1
|
Elias MG, Fatima S, Mann TJ, Karan S, Mikhael M, de Souza P, Gordon CP, Scott KF, Aldrich-Wright JR. Anticancer Effect of Pt IIPHEN SS, Pt II5ME SS, Pt II56ME SS and Their Platinum(IV)-Dihydroxy Derivatives against Triple-Negative Breast Cancer and Cisplatin-Resistant Colorectal Cancer. Cancers (Basel) 2024; 16:2544. [PMID: 39061185 PMCID: PMC11274883 DOI: 10.3390/cancers16142544] [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: 05/31/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Development of resistance to cisplatin, oxaliplatin and carboplatin remains a challenge for their use as chemotherapies, particularly in breast and colorectal cancer. Here, we compare the anticancer effect of novel complexes [Pt(1,10-phenanthroline)(1S,2S-diaminocyclohexane)](NO3)2 (PtIIPHENSS), [Pt(5-methyl-1,10-phenanthroline)(1S,2S-diaminocyclohexane)](NO3)2 (PtII5MESS) and [Pt(5,6-dimethyl-1,10-phenanthroline)(1S,2S-diaminocyclohexane)](NO3)2 (PtII56MESS) and their platinum(IV)-dihydroxy derivatives with cisplatin. Complexes are greater than 11-fold more potent than cisplatin in both 2D and 3D cell line cultures with increased selectivity for cancer cells over genetically stable cells. ICP-MS studies showed cellular uptake occurred through an active transport mechanism with considerably altered platinum concentrations found in the cytoskeleton across all complexes after 24 h. Significant reactive oxygen species generation was observed, with reduced mitochondrial membrane potential at 72 h of treatment. Late apoptosis/necrosis was shown by Annexin V-FITC/PI flow cytometry assay, accompanied by increased sub-G0/G1 cells compared with untreated cells. An increase in S and G2+M cells was seen with all complexes. Treatment resulted in significant changes in actin and tubulin staining. Intrinsic and extrinsic apoptosis markers, MAPK/ERK and PI3K/AKT activation markers, together with autophagy markers showed significant activation of these pathways by Western blot. The proteomic profile investigated post-72 h of treatment identified 1597 MDA-MB-231 and 1859 HT29 proteins quantified by mass spectroscopy, with several differentially expressed proteins relative to no treatment. GO enrichment analysis revealed a statistically significant enrichment of RNA/DNA-associated proteins in both the cell lines and specific additional processes for individual drugs. This study shows that these novel agents function as multi-mechanistic chemotherapeutics, offering promising anticancer potential, and thereby supporting further research into their application as cancer therapeutics.
Collapse
Affiliation(s)
- Maria George Elias
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (S.K.); (M.M.); (C.P.G.)
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.)
| | - Shadma Fatima
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Timothy J. Mann
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Shawan Karan
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (S.K.); (M.M.); (C.P.G.)
| | - Meena Mikhael
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (S.K.); (M.M.); (C.P.G.)
| | - Paul de Souza
- Nepean Clinical School, Faculty of Medicine and Health, University of Sydney, Kingswood, NSW 2747, Australia;
| | - Christopher P. Gordon
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (S.K.); (M.M.); (C.P.G.)
| | - Kieran F. Scott
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Janice R. Aldrich-Wright
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (S.K.); (M.M.); (C.P.G.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| |
Collapse
|
2
|
McAdam AD, Batchelor LK, Romano-deGea J, Vasilyev D, Dyson PJ. Thermoresponsive carboplatin-releasing prodrugs. J Inorg Biochem 2024; 254:112505. [PMID: 38377623 DOI: 10.1016/j.jinorgbio.2024.112505] [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: 11/28/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 02/22/2024]
Abstract
Platinum-based anticancer drugs, while potent, are associated with numerous and severe side effects. Hyperthermia therapy is an effective adjuvant in anticancer treatment, however, clinically used platinum drugs have not been optimised for combination with hyperthermia. The derivatisation of existing anticancer drugs with appropriately chosen thermoresponsive moieties results in drugs being activated only at the heated site. Perfluorinated chains of varying lengths were installed on carboplatin, a clinically approved drug, leading to the successful synthesis of a series of mono- and di- substituted platinum(IV) carboplatin prodrugs. Some of these complexes display relevant thermosensitivity on ovarian cancer cell lines, i.e., being inactive at 37 °C while having comparable activity to carboplatin under mild hyperthermia (42 °C). Nuclear magnetic resonance spectroscopy and mass spectrometry indicated that carboplatin is likely the active platinum(II) anticancer agent upon reduction and cyclic voltammetry revealed that the length of the fluorinated alkyl chain has a strong influence on the rate of carboplatin formation, regulating the subsequent cytotoxicity.
Collapse
Affiliation(s)
- Aemilia D McAdam
- Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Lucinda K Batchelor
- Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Jan Romano-deGea
- Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Dmitry Vasilyev
- Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Paul J Dyson
- Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| |
Collapse
|
3
|
Mao L, Qin Y, Fan J, Yang W, Li B, Cao L, Yuan L, Wang M, Liu B, Wang W. Rapid discovery of a novel "green" and natural GST inhibitor for sensitizing hepatocellular carcinoma to Cisplatin by visual screening strategy. J Pharm Anal 2024; 14:100923. [PMID: 38799232 PMCID: PMC11127223 DOI: 10.1016/j.jpha.2023.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/24/2023] [Accepted: 12/16/2023] [Indexed: 05/29/2024] Open
Abstract
Over-expression of glutathione S-transferase (GST) can promote Cisplatin resistance in hepatocellular carcinoma (HCC) treatment. Hence, inhibiting GST is an attractive strategy to improve Cisplatin sensitivity in HCC therapy. Although several synthesized GST inhibitors have been developed, the side effects and narrow spectrum for anticancer seriously limit their clinical application. Considering the abundance of natural compounds with anticancer activity, this study developed a rapid fluorescence technique to screen "green" natural GST inhibitors with high specificity. The fluorescence assay demonstrated that schisanlactone B (hereafter abbreviated as C1) isolated from Xue tong significantly down-regulated GST levels in Cisplatin-resistant HCC cells in vitro and in vivo. Importantly, C1 can selectively kill HCC cells from normal liver cells, effectively improving the therapeutic effect of Cisplatin on HCC mice by down-regulating GST expression. Considering the high GST levels in HCC patients, this compound demonstrated the high potential for sensitizing HCC therapy in clinical practice by down-regulating GST levels.
Collapse
Affiliation(s)
- Linxi Mao
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yan Qin
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
- College of Biology, Hunan University, Changsha, 410082, China
| | - Jialong Fan
- College of Biology, Hunan University, Changsha, 410082, China
| | - Wei Yang
- College of Foreign Languages, Hunan Women's University, Changsha, 410004, China
| | - Bin Li
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Liang Cao
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Liqin Yuan
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Mengyun Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Bin Liu
- College of Biology, Hunan University, Changsha, 410082, China
| | - Wei Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| |
Collapse
|
4
|
Ahmedova A, Mihaylova R, Stoykova S, Mihaylova V, Burdzhiev N, Elincheva V, Momekov G, Momekova D. Pyrenebutyrate Pt(IV) Complexes with Nanomolar Anticancer Activity. Pharmaceutics 2023; 15:2310. [PMID: 37765279 PMCID: PMC10537052 DOI: 10.3390/pharmaceutics15092310] [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: 08/22/2023] [Revised: 09/06/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023] Open
Abstract
Research on platinum-based anticancer drugs continuously strives to develop new non-classical platinum complexes. Pt(IV) prodrugs are the most promising, and their activation-by-reduction mechanism of action is being explored as a prospect for higher selectivity and efficiency. Herein, we present the anticancer potency and chemical reactivity of Pt(IV) complexes formed by linking pyrene butyric acid with cisplatin. The results from cytotoxicity screening on 10 types of cancer cell lines and non-malignant cells (HEK-293) indicated IC50 values as low as 50-70 nM for the monosubstituted Pt(IV) complex against leukemia cell lines (HL-60 and SKW3) and a cisplatin-resistant derivative (HL-60/CDDP). Interestingly, the bis-substituted complex is virtually non-toxic to both healthy and cancerous cells of adherent types. Nevertheless, it shows high cytotoxicity against multidrug-resistant derivatives HL-60/CDDP and HL-60/Dox. The reactivity of the complexes with biological reductants was monitored by the NMR method. Furthermore, the platinum uptake by the treated cells was examined on two types of cellular cultures: adherent and suspension growing, and proteome profiling was conducted to track expression changes of key apoptosis-related proteins in HL-60 cells. The general conclusion points to a possible cytoskeletal entrapment of the bulkier bis-pyrene complex that could be limiting its cytotoxicity to adherent cells, both cancerous and healthy ones.
Collapse
Affiliation(s)
- Anife Ahmedova
- Faculty of Chemistry and Pharmacy, Sofia University, 1, J. Bourchier Blvd., 1164 Sofia, Bulgaria; (S.S.); (V.M.); (N.B.)
| | - Rositsa Mihaylova
- Faculty of Pharmacy, Medical University-Sofia, 2 Dunav Street, 1000 Sofia, Bulgaria; (R.M.); (G.M.); (D.M.)
| | - Silviya Stoykova
- Faculty of Chemistry and Pharmacy, Sofia University, 1, J. Bourchier Blvd., 1164 Sofia, Bulgaria; (S.S.); (V.M.); (N.B.)
| | - Veronika Mihaylova
- Faculty of Chemistry and Pharmacy, Sofia University, 1, J. Bourchier Blvd., 1164 Sofia, Bulgaria; (S.S.); (V.M.); (N.B.)
| | - Nikola Burdzhiev
- Faculty of Chemistry and Pharmacy, Sofia University, 1, J. Bourchier Blvd., 1164 Sofia, Bulgaria; (S.S.); (V.M.); (N.B.)
| | - Viktoria Elincheva
- Faculty of Pharmacy, Medical University-Sofia, 2 Dunav Street, 1000 Sofia, Bulgaria; (R.M.); (G.M.); (D.M.)
| | - Georgi Momekov
- Faculty of Pharmacy, Medical University-Sofia, 2 Dunav Street, 1000 Sofia, Bulgaria; (R.M.); (G.M.); (D.M.)
| | - Denitsa Momekova
- Faculty of Pharmacy, Medical University-Sofia, 2 Dunav Street, 1000 Sofia, Bulgaria; (R.M.); (G.M.); (D.M.)
| |
Collapse
|
5
|
Mazari AMA, Zhang L, Ye ZW, Zhang J, Tew KD, Townsend DM. The Multifaceted Role of Glutathione S-Transferases in Health and Disease. Biomolecules 2023; 13:688. [PMID: 37189435 PMCID: PMC10136111 DOI: 10.3390/biom13040688] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
In humans, the cytosolic glutathione S-transferase (GST) family of proteins is encoded by 16 genes presented in seven different classes. GSTs exhibit remarkable structural similarity with some overlapping functionalities. As a primary function, GSTs play a putative role in Phase II metabolism by protecting living cells against a wide variety of toxic molecules by conjugating them with the tripeptide glutathione. This conjugation reaction is extended to forming redox sensitive post-translational modifications on proteins: S-glutathionylation. Apart from these catalytic functions, specific GSTs are involved in the regulation of stress-induced signaling pathways that govern cell proliferation and apoptosis. Recently, studies on the effects of GST genetic polymorphisms on COVID-19 disease development revealed that the individuals with higher numbers of risk-associated genotypes showed higher risk of COVID-19 prevalence and severity. Furthermore, overexpression of GSTs in many tumors is frequently associated with drug resistance phenotypes. These functional properties make these proteins promising targets for therapeutics, and a number of GST inhibitors have progressed in clinical trials for the treatment of cancer and other diseases.
Collapse
Affiliation(s)
- Aslam M. A. Mazari
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 70 President Street, DDB410, Charleston, SC 29425, USA
| | - Leilei Zhang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 70 President Street, DDB410, Charleston, SC 29425, USA
| | - Zhi-Wei Ye
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 70 President Street, DDB410, Charleston, SC 29425, USA
| | - Jie Zhang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 70 President Street, DDB410, Charleston, SC 29425, USA
| | - Kenneth D. Tew
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 70 President Street, DDB410, Charleston, SC 29425, USA
| | - Danyelle M. Townsend
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, 274 Calhoun Street, MSC141, Charleston, SC 29425, USA
| |
Collapse
|
6
|
Yu L, Lee H, Rho SB, Park MK, Lee CH. Ethacrynic Acid: A Promising Candidate for Drug Repurposing as an Anticancer Agent. Int J Mol Sci 2023; 24:ijms24076712. [PMID: 37047688 PMCID: PMC10094867 DOI: 10.3390/ijms24076712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023] Open
Abstract
Ethacrynic acid (ECA) is a diuretic that inhibits Na-K-2Cl cotransporter (NKCC2) present in the thick ascending loop of Henle and muculo dens and is clinically used for the treatment of edema caused by excessive body fluid. However, its clinical use is limited due to its low bioavailability and side effects, such as liver damage and hearing loss at high doses. Despite this, ECA has recently emerged as a potential anticancer agent through the approach of drug repositioning, with a novel mechanism of action. ECA has been shown to regulate cancer hallmark processes such as proliferation, apoptosis, migration and invasion, angiogenesis, inflammation, energy metabolism, and the increase of inhibitory growth factors through various mechanisms. Additionally, ECA has been used as a scaffold for synthesizing a new material, and various derivatives have been synthesized. This review explores the potential of ECA and its derivatives as anticancer agents, both alone and in combination with adjuvants, by examining their effects on ten hallmarks of cancer and neuronal contribution to cancer. Furthermore, we investigated the trend of synthesis research of a series of ECA derivatives to improve the bioavailability of ECA. This review highlights the importance of ECA research and its potential to provide a cost-effective alternative to new drug discovery and development for cancer treatment.
Collapse
Affiliation(s)
- Lu Yu
- College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Ho Lee
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy National Cancer Center, Goyang 10408, Republic of Korea
| | - Seung Bae Rho
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy National Cancer Center, Goyang 10408, Republic of Korea
| | - Mi Kyung Park
- College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy National Cancer Center, Goyang 10408, Republic of Korea
| | - Chang Hoon Lee
- College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| |
Collapse
|
7
|
Zhao J, Gao Y, He W, Wang W, Hu W, Sun Y. Synthesis, characterization and biological evaluation of two cyclometalated iridium(III) complexes containing a glutathione S-transferase inhibitor. J Inorg Biochem 2023; 238:112050. [PMID: 36332411 DOI: 10.1016/j.jinorgbio.2022.112050] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/12/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
The cyclometalated iridium(III) compounds have been intensively studied for health-related applications due to their outstanding luminescent properties and multiple anticancer modes of action. Herein, two iridium(III) compounds Ir-1 and Ir-3 containing glutathione S-transferase inhibitor (GSTi) were developed and studied together with two unfunctionalized compounds Ir-2 and Ir-4 as a comparison. Biological study indicated that GSTi-bearing complexes Ir-1 and Ir-3 exert a synergistic effect on the inhibition of cancer cells. The photophysical properties of Ir-1 ∼ Ir-4 were investigated by UV/vis absorption and fluorescence spectroscopy and rationalized with TD-DFT calculations. As expected, GSTi-bearing complexes Ir-1 and Ir-3 exhibited considerable cytotoxicity against both A549 and cisplatin-resistant A549/cis cancer cells, much higher than the unfunctionalized iridium compounds Ir-2 and Ir-4. Further study indicated that Ir-1 and Ir-3 mainly localize in the mitochondria of tumor cells, and exert their cytotoxicity via generating ROS and inhibiting GST activity. The flow cytometry investigations demonstrated that Ir-1 and Ir-3 can arrest the cell cycle in S phase and induce the cell death through apoptosis process. Overall, the complexation of GST inhibitors with cyclometalated iridium(III) agents provides an effective way for potentiating the cytotoxicity of iridium(III) anticancer agents and resensitizing the efficacy against cisplatin resistant cancer cells.
Collapse
Affiliation(s)
- Jian Zhao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China; Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huaian 223003, China.
| | - Ya Gao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Weiyu He
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Wei Wang
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Weiwei Hu
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huaian 223003, China.
| | - Yanyan Sun
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, China.
| |
Collapse
|
8
|
Kasparkova J, Kostrhunova H, Novohradsky V, Ma L, Zhu G, Milaeva ER, Shtill AA, Vinck R, Gasser G, Brabec V, Nazarov AA. Is antitumor Pt(IV) complex containing two axial lonidamine ligands a true dual- or multi-action prodrug? METALLOMICS : INTEGRATED BIOMETAL SCIENCE 2022; 14:6618656. [PMID: 35759404 DOI: 10.1093/mtomcs/mfac048] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/09/2022] [Indexed: 11/14/2022]
Abstract
This work studied the mechanism of action of a Pt(IV) complex 2 bearing two axial lonidamine ligands, which are selective inhibitors of aerobic glycolysis. The presence of two lonidamine ligands in 2 compared to the parent Pt(II) complex increased its antiproliferative activity, cellular accumulation, and changed its cell cycle profile and mechanism of cell death. In 3D cell culture, 2 showed exceptional antiproliferative activity with IC50 values as low as 1.6 μM in MCF7 cells. The study on the influence of the lonidamine ligands in the Pt complex on glycolysis showed only low potency of ligands to affect metabolic processes in cancer cells, making the investigated complex, not a dual- or multi-action prodrug. However, the Pt(IV) prodrug effectively delivers the cytotoxic Pt(II) complex into cancer cells.
Collapse
Affiliation(s)
- Jana Kasparkova
- Czech Academy of Sciences, Institute of Biophysics, Brno CZ-61265, Czech Republic
| | - Hana Kostrhunova
- Czech Academy of Sciences, Institute of Biophysics, Brno CZ-61265, Czech Republic
| | - Vojtech Novohradsky
- Czech Academy of Sciences, Institute of Biophysics, Brno CZ-61265, Czech Republic
| | - Lili Ma
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, P. R. China
| | - Guangyu Zhu
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, P. R. China
| | - Elena R Milaeva
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russian Federation
| | - Alexender A Shtill
- Blokhin Cancer Center, Russian Academy of Medical Sciences, 115478 Moscow, Russian Federation
| | - Robin Vinck
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, 75005 Paris, France
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, 75005 Paris, France
| | - Viktor Brabec
- Czech Academy of Sciences, Institute of Biophysics, Brno CZ-61265, Czech Republic
| | - Alexey A Nazarov
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russian Federation
| |
Collapse
|
9
|
Fronik P, Gutmann M, Vician P, Stojanovic M, Kastner A, Heffeter P, Pirker C, Keppler BK, Berger W, Kowol CR. A platinum(IV) prodrug strategy to overcome glutathione-based oxaliplatin resistance. Commun Chem 2022; 5:46. [PMID: 36697790 PMCID: PMC9814792 DOI: 10.1038/s42004-022-00661-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 03/07/2022] [Indexed: 02/01/2023] Open
Abstract
Clinical efficacy of oxaliplatin is frequently limited by severe adverse effects and therapy resistance. Acquired insensitivity to oxaliplatin is, at least in part, associated with elevated levels of glutathione (GSH). In this study we report on an oxaliplatin-based platinum(IV) prodrug, which releases L-buthionine-S,R-sulfoximine (BSO), an inhibitor of glutamate-cysteine ligase, the rate-limiting enzyme in GSH biosynthesis. Two complexes bearing either acetate (BSO-OxOAc) or an albumin-binding maleimide (BSO-OxMal) as second axial ligand were synthesized and characterized. The in vitro anticancer activity of BSO-OxOAc was massively reduced in comparison to oxaliplatin, proving its prodrug nature. Nevertheless, the markedly lower intracellular oxaliplatin uptake in resistant HCT116/OxR cells was widely overcome by BSO-OxOAc resulting in distinctly reduced resistance levels. Platinum accumulation in organs of a colorectal cancer mouse model revealed higher tumor selectivity of BSO-OxMal as compared to oxaliplatin. This corresponded with increased antitumor activity, resulting in significantly enhanced overall survival. BSO-OxMal-treated tumors exhibited reduced GSH levels, proliferative activity and enhanced DNA damage (pH2AX) compared to oxaliplatin. Conversely, pH2AX staining especially in kidney cells was distinctly increased by oxaliplatin but not by BSO-OxMal. Taken together, our data provide compelling evidence for enhanced tumor specificity of the oxaliplatin(IV)/BSO prodrug.
Collapse
Affiliation(s)
- Philipp Fronik
- grid.10420.370000 0001 2286 1424University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry, Waehringer Strasse 42, 1090 Vienna, Austria
| | - Michael Gutmann
- grid.22937.3d0000 0000 9259 8492Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria ,Research Cluster “Translational Cancer Therapy Research”, 1090 Vienna, Austria
| | - Petra Vician
- grid.22937.3d0000 0000 9259 8492Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Mirjana Stojanovic
- grid.22937.3d0000 0000 9259 8492Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Alexander Kastner
- grid.10420.370000 0001 2286 1424University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry, Waehringer Strasse 42, 1090 Vienna, Austria
| | - Petra Heffeter
- grid.22937.3d0000 0000 9259 8492Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria ,Research Cluster “Translational Cancer Therapy Research”, 1090 Vienna, Austria
| | - Christine Pirker
- grid.22937.3d0000 0000 9259 8492Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria ,Research Cluster “Translational Cancer Therapy Research”, 1090 Vienna, Austria
| | - Bernhard K. Keppler
- grid.10420.370000 0001 2286 1424University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry, Waehringer Strasse 42, 1090 Vienna, Austria ,Research Cluster “Translational Cancer Therapy Research”, 1090 Vienna, Austria
| | - Walter Berger
- grid.22937.3d0000 0000 9259 8492Center of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria ,Research Cluster “Translational Cancer Therapy Research”, 1090 Vienna, Austria
| | - Christian R. Kowol
- grid.10420.370000 0001 2286 1424University of Vienna, Faculty of Chemistry, Institute of Inorganic Chemistry, Waehringer Strasse 42, 1090 Vienna, Austria ,Research Cluster “Translational Cancer Therapy Research”, 1090 Vienna, Austria
| |
Collapse
|
10
|
Network Biology and Artificial Intelligence Drive the Understanding of the Multidrug Resistance Phenotype in Cancer. Drug Resist Updat 2022; 60:100811. [DOI: 10.1016/j.drup.2022.100811] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/07/2023]
|
11
|
Moynihan E, Bassi G, Ruffini A, Panseri S, Montesi M, Velasco-Torrijos T, Montagner D. Click Pt(IV)-Carbohydrates Pro-Drugs for Treatment of Osteosarcoma. Front Chem 2021; 9:795997. [PMID: 34950638 PMCID: PMC8688915 DOI: 10.3389/fchem.2021.795997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/08/2021] [Indexed: 11/29/2022] Open
Abstract
The selectivity vs. cancer cells has always been a major challenge for chemotherapeutic agents and in particular for cisplatin, one of the most important anticancer drugs for the treatment of several types of tumors. One strategy to overtake this challenge is to modify the coordination sphere of the metallic center with specific vectors whose receptors are overexpressed in the tumoral cell membrane, such as monosaccharides. In this paper, we report the synthesis of four novel glyco-modified Pt(IV) pro-drugs, based on cisplatin scaffold, and their biological activity against osteosarcoma (OS), a malignant tumor affecting in particular adolescents and young adults. The sugar moiety and the Pt scaffold are linked exploiting the Copper Azide Alkyne Cycloaddition (CUAAC) reaction, which has become the flagship of click chemistry due to its versatility and mild conditions. Cytotoxicity and drug uptake on three different OS cell lines as well as CSCs (Cancer Stem Cell) are described.
Collapse
Affiliation(s)
- Eoin Moynihan
- Department of Chemistry, Maynooth University, Maynooth, Ireland
| | - Giada Bassi
- Institute of Science and Technology for Ceramics-National Research Council, Faenza, Italy
| | - Andrea Ruffini
- Institute of Science and Technology for Ceramics-National Research Council, Faenza, Italy
| | - Silvia Panseri
- Institute of Science and Technology for Ceramics-National Research Council, Faenza, Italy
| | - Monica Montesi
- Institute of Science and Technology for Ceramics-National Research Council, Faenza, Italy
| | - Trinidad Velasco-Torrijos
- Department of Chemistry, Maynooth University, Maynooth, Ireland.,Kathleen Londsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Diego Montagner
- Department of Chemistry, Maynooth University, Maynooth, Ireland.,Kathleen Londsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| |
Collapse
|
12
|
Predarska I, Saoud M, Morgan I, Eichhorn T, Kaluđerović GN, Hey-Hawkins E. Cisplatin-cyclooxygenase inhibitor conjugates, free and immobilised in mesoporous silica SBA-15, prove highly potent against triple-negative MDA-MB-468 breast cancer cell line. Dalton Trans 2021; 51:857-869. [PMID: 34877948 DOI: 10.1039/d1dt03265h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
For the development of anticancer drugs with higher activity and reduced toxicity, two approaches were combined: preparation of platinum(IV) complexes exhibiting higher stability compared to their platinum(II) counterparts and loading them into mesoporous silica SBA-15 with the aim to utilise the passive enhanced permeability and retention (EPR) effect of nanoparticles for accumulation in tumour tissues. Three conjugates based on a cisplatin scaffold bearing the anti-inflammatory drugs naproxen, ibuprofen or flurbiprofen in the axial positions (1, 2 and 3, respectively) were synthesised and loaded into SBA-15 to afford the mesoporous silica nanoparticles (MSNs) SBA-15|1, SBA-15|2 and SBA-15|3. Superior antiproliferative activity of both free and immobilised conjugates in a panel of four breast cancer cell lines (MDA-MB-468, HCC1937, MCF-7 and BT-474) with markedly increased cytotoxicity with respect to cisplatin was demonstrated. All compounds exhibit highest activity against the triple-negative cell line MDA-MB-468, with conjugate 1 being the most potent. However, against MCF-7 and BT-474 cell lines, the most notable improvement was found, with IC50 values up to 240-fold lower than cisplatin. Flow cytometry assays clearly show that all compounds induce apoptotic cell death elevating the levels of both early and late apoptotic cells. Furthermore, autophagy as well as formation of reactive oxygen species (ROS) and nitric oxide (NO) were elevated to a similar or greater extent than with cisplatin.
Collapse
Affiliation(s)
- Ivana Predarska
- Universität Leipzig, Faculty of Chemistry and Mineralogy, Institute of Inorganic Chemistry, Johannisallee 29, 04103 Leipzig, Germany. .,Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Str. 2, 06217 Merseburg, Germany
| | - Mohamad Saoud
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D 06120 Halle (Saale), Germany.
| | - Ibrahim Morgan
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D 06120 Halle (Saale), Germany.
| | - Thomas Eichhorn
- Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Str. 2, 06217 Merseburg, Germany
| | - Goran N Kaluđerović
- Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Str. 2, 06217 Merseburg, Germany.,Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D 06120 Halle (Saale), Germany.
| | - Evamarie Hey-Hawkins
- Universität Leipzig, Faculty of Chemistry and Mineralogy, Institute of Inorganic Chemistry, Johannisallee 29, 04103 Leipzig, Germany.
| |
Collapse
|
13
|
Peng K, Liang BB, Liu W, Mao ZW. What blocks more anticancer platinum complexes from experiment to clinic: Major problems and potential strategies from drug design perspectives. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214210] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
14
|
El Abbouchi A, El Brahmi N, Hiebel MA, Bignon J, Guillaumet G, Suzenet F, El Kazzouli S. Synthesis and evaluation of a novel class of ethacrynic acid derivatives containing triazoles as potent anticancer agents. Bioorg Chem 2021; 115:105293. [PMID: 34426162 DOI: 10.1016/j.bioorg.2021.105293] [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: 03/18/2021] [Revised: 07/03/2021] [Accepted: 08/17/2021] [Indexed: 12/30/2022]
Abstract
For unmet clinical needs, a novel class of ethacrynic acid (EA) derivatives containing triazole moieties (3a-i and 8) were designed, synthesized and evaluated as new anticancer agents. The in vitro anti-proliferative activities were assessed first on HL60 cell line and in a second stage, the two selected compounds 3a and 3c were tested on a panel of human cancer cell lines (A549, MCF7, PC3, U87-MG, SKOV3 and HCT116) and on a normal cell line (MCR5). Compound3c exhibited very good antitumor activities with IC50 values of 20.2, 56.5 and 76.8 nM against A549, PC3 and U87-MG cell lines respectively, which is 2.8- and 1.3-fold more active than doxorubicin on A549 and U87-MG cancer cells, respectively. In addition, compound 3c displays a very good safety index (SI) of 82 fold for A549. Compound 3a showed also good IC50 values of 50 nM on both A549 and PC3 cells and lower selectivity compared to 3c for A549 and PC3 vs. MCR5 with SI of 33 and 18 fold, respectively. The measurement of mitochondrial membrane potential on HCT116 cells after treatments by either 3a or 3c showed that both compounds induced mitochondrial dysfunctions causing thus caspase-induced apoptosis.
Collapse
Affiliation(s)
- Abdelmoula El Abbouchi
- Euromed Research Center, Engineering School of Biomedical and Biotechnology, Euromed University of Fes (UEMF)-Route de Meknès, 30000 Fez, Morocco; Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, BP 6759, Orléans cedex 2 54067, France
| | - Nabil El Brahmi
- Euromed Research Center, Engineering School of Biomedical and Biotechnology, Euromed University of Fes (UEMF)-Route de Meknès, 30000 Fez, Morocco
| | - Marie-Aude Hiebel
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, BP 6759, Orléans cedex 2 54067, France
| | - Jérôme Bignon
- Institut de Chimie des Substances Naturelles, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Gérald Guillaumet
- Euromed Research Center, Engineering School of Biomedical and Biotechnology, Euromed University of Fes (UEMF)-Route de Meknès, 30000 Fez, Morocco; Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, BP 6759, Orléans cedex 2 54067, France.
| | - Franck Suzenet
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, BP 6759, Orléans cedex 2 54067, France.
| | - Saïd El Kazzouli
- Euromed Research Center, Engineering School of Biomedical and Biotechnology, Euromed University of Fes (UEMF)-Route de Meknès, 30000 Fez, Morocco.
| |
Collapse
|
15
|
Han T, Wu Y, Han W, Yan K, Zhao J, Sun Y. Antitumor Effect of Organometallic Half-Sandwich Ru(II)-Arene Complexes Bearing a Glutathione S-Transferase Inhibitor. Inorg Chem 2021; 60:13051-13061. [PMID: 34369147 DOI: 10.1021/acs.inorgchem.1c01482] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The facile modification of the ligands in organometallic Ru(II)-arene complexes offers more opportunities to optimize their pharmacological profiles. Herein, three Ru(II)-arene complexes containing a glutathione S-transferase (GST) inhibitor (NBDHEX) in chelate ligand have been designed and synthesized in this study. In vitro results indicated that the ligation with NBDHEX significantly increased the activities and selectivities of the organometallic Ru(II)-arene complexes against tumor cells, especially complex 3, which was the most active compound among the tested compounds. DFT calculations and hydrolysis results demonstrated that complex 3 with more alkyl groups in the arene ligand has increased electron density at the Ru(II) center as compared with complexes 1 and 2, thus resulting in the improved hydrolysis rate, which may be responsible for its higher anticancer activity. Further studies showed that complexes 1-3 can cause the loss of the mitochondrial membrane potential and upregulate the expression of Bcl-2 and Bax in A549 cells, suggesting that complexes 1-3-induced cell death may be mediated via the mitochondrial apoptotic pathway. Thus, these findings suggested that simultaneous modification of the chelate ligands and arene rings in the organometallic Ru(II)-arene complexes is an effective way to improve their pharmacological properties.
Collapse
Affiliation(s)
- Tianyu Han
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Yuying Wu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Weinan Han
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Kaiwen Yan
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Jian Zhao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Yanyan Sun
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, China
| |
Collapse
|
16
|
Valente A, Podolski-Renić A, Poetsch I, Filipović N, López Ó, Turel I, Heffeter P. Metal- and metalloid-based compounds to target and reverse cancer multidrug resistance. Drug Resist Updat 2021; 58:100778. [PMID: 34403910 DOI: 10.1016/j.drup.2021.100778] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/18/2021] [Accepted: 08/03/2021] [Indexed: 12/19/2022]
Abstract
Drug resistance remains the major cause of cancer treatment failure especially at the late stage of the disease. However, based on their versatile chemistry, metal and metalloid compounds offer the possibility to design fine-tuned drugs to circumvent and even specifically target drug-resistant cancer cells. Based on the paramount importance of platinum drugs in the clinics, two main areas of drug resistance reversal strategies exist: overcoming resistance to platinum drugs as well as multidrug resistance based on ABC efflux pumps. The current review provides an overview of both aspects of drug design and discusses the open questions in the field. The areas of drug resistance covered in this article involve: 1) Altered expression of proteins involved in metal uptake, efflux or intracellular distribution, 2) Enhanced drug efflux via ABC transporters, 3) Altered metabolism in drug-resistant cancer cells, 4) Altered thiol or redox homeostasis, 5) Altered DNA damage recognition and enhanced DNA damage repair, 6) Impaired induction of apoptosis and 7) Altered interaction with the immune system. This review represents the first collection of metal (including platinum, ruthenium, iridium, gold, and copper) and metalloid drugs (e.g. arsenic and selenium) which demonstrated drug resistance reversal activity. A special focus is on compounds characterized by collateral sensitivity of ABC transporter-overexpressing cancer cells. Through this approach, we wish to draw the attention to open research questions in the field. Future investigations are warranted to obtain more insights into the mechanisms of action of the most potent compounds which target specific modalities of drug resistance.
Collapse
Affiliation(s)
- Andreia Valente
- Centro de Química Estrutural and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisboa, Portugal
| | - Ana Podolski-Renić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Serbia
| | - Isabella Poetsch
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Nenad Filipović
- Department of Chemistry and Biochemistry, Faculty of Agriculture, University of Belgrade, Belgrade, Serbia
| | - Óscar López
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Sevilla, Spain
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
17
|
Harringer S, Hejl M, Enyedy ÉA, Jakupec MA, Galanski MS, Keppler BK, Dyson PJ, Varbanov HP. Multifunctional Pt(iv) prodrug candidates featuring the carboplatin core and deferoxamine. Dalton Trans 2021; 50:8167-8178. [PMID: 34031671 DOI: 10.1039/d1dt00214g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The synergistic combination of the anticancer drug carboplatin and the iron chelator deferoxamine (DFO) served as a foundation for the development of novel multifunctional prodrugs. Hence, five platinum(iv) complexes, featuring the equatorial coordination sphere of carboplatin, and one or two DFO units incorporated at axial positions, were synthesized and characterized using ESI-HRMS, multinuclear (1H, 13C, 15N, 195Pt) NMR spectroscopy and elemental analysis. Analytical studies demonstrated that the chelating properties of the DFO moiety were not compromised after coupling to the platinum(iv) core. The cytotoxic activity of the compounds was evaluated in monolayer (2D) and spheroid (3D) cancer cell models, derived from ovarian teratocarcinoma (CH1/PA-1), colon carcinoma (SW480) and non-small cell lung cancer (A549). The platinum(iv)-DFO prodrugs demonstrated moderate in vitro cytotoxicity (a consequence of their slow activation kinetics) but with less pronounced differences between intrinsically chemoresistant and chemosensitive cell lines as well as between 2D and 3D models than the clinically used platinum(ii) drug carboplatin.
Collapse
Affiliation(s)
- Sophia Harringer
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, 1090 Vienna, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Dang Y, Ruan L, Tian Y, Xu Z, Zhang W. Nitric Oxide Prodrug Delivery and Release Monitoring Based on a Galactose-Modified Multifunctional Nanoprobe. Anal Chem 2021; 93:7625-7634. [PMID: 34010568 DOI: 10.1021/acs.analchem.1c00287] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Nitric oxide (NO)-based cancer therapy has attracted much attention in recent years owing to its broad effects on cancer. Low concentrations of NO stimulate cancer cell progression, while its higher levels induce cell apoptosis, and thus, it has motivated the development of probes for in situ NO release monitoring. In this work, a galactose-modified benzothiadiazole-based fluorescent probe (GalNONP/C) was synthesized as both a NO-responsive nanoprobe and NO prodrug carrier. The probe exhibited far-red emission in the range from 550 to 800 nm, and the response showed acidity preference. The galactose on the probe enabled selective targeting of hepatocellular carcinoma (HCC) cells by binding to the asialoglycoprotein receptor (ASGPR) on the cell surface. The probe also delivered low-molecular weight NO prodrug JS-K into cells and monitored the real-time release of the generated NO. Furthermore, in vivo NO imaging with tumor targeting was demonstrated in HCC orthotopic transplantation nude mice and liver sections. Compared with the control experiment using a probe without NO prodrug loading, higher fluorescence response of NO was detected in the cell (3.0 times) and liver slices of the HCC tumor model (2.7 times). This strategy may pave the way to develop nanoprobes for in situ NO monitoring and therapy evaluation in NO-related cancer therapy.
Collapse
Affiliation(s)
- Yijing Dang
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Liting Ruan
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Yang Tian
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Zhiai Xu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Wen Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, East China Normal University, Shanghai 200062, China
| |
Collapse
|
19
|
Glutathione S-Transferases in Cancer. Antioxidants (Basel) 2021; 10:antiox10050701. [PMID: 33946704 PMCID: PMC8146591 DOI: 10.3390/antiox10050701] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 02/06/2023] Open
Abstract
In humans, the glutathione S-transferases (GST) protein family is composed of seven members that present remarkable structural similarity and some degree of overlapping functionalities. GST proteins are crucial antioxidant enzymes that regulate stress-induced signaling pathways. Interestingly, overactive GST proteins are a frequent feature of many human cancers. Recent evidence has revealed that the biology of most GST proteins is complex and multifaceted and that these proteins actively participate in tumorigenic processes such as cell survival, cell proliferation, and drug resistance. Structural and pharmacological studies have identified various GST inhibitors, and these molecules have progressed to clinical trials for the treatment of cancer and other diseases. In this review, we discuss recent findings in GST protein biology and their roles in cancer development, their contribution in chemoresistance, and the development of GST inhibitors for cancer treatment.
Collapse
|
20
|
Gibson D. Platinum(IV) anticancer agents; are we en route to the holy grail or to a dead end? J Inorg Biochem 2021; 217:111353. [PMID: 33477089 DOI: 10.1016/j.jinorgbio.2020.111353] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/02/2020] [Accepted: 12/13/2020] [Indexed: 01/23/2023]
Abstract
Pt(IV) complexes are designed as prodrugs that are intended to overcome resistance. Pt(IV) prodrugs are activated inside cancer cells releasing cytotoxic Pt(II) drugs as well as two axial ligands that can be used to confer favorable pharmacological properties to the prodrug. The ligands can be innocent spectators, cancer targeting agents or bioactive moieties. The choice of axial ligands determines the chemical and pharmacological properties of the prodrugs. Over the years, several approaches were employed in attempts to increase the selectivity of the prodrugs to cancer cells and to utilize multi-action prodrugs to overcome resistance. In this review, we critically examine several of these approaches in order to evaluate the validity of some of the working hypotheses that are driving the current research.
Collapse
Affiliation(s)
- Dan Gibson
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel.
| |
Collapse
|
21
|
Chen H, Chen F, Wang X, Gou S. Multifunctional Pt(iv) complexes containing a glutathione S-transferase inhibitor lead to enhancing anticancer activity and preventing metastasis of osteosarcoma cells. Metallomics 2020; 11:317-326. [PMID: 30560252 DOI: 10.1039/c8mt00296g] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cisplatin has been clinically applied in the treatment of osteosarcoma (OS), but its efficacy is severely limited due to drug resistance and metastasis. One of the chief culprits is the overexpression of glutathione S-transferases (GSTs) in cancer cells, which can accelerate the interaction of glutathione (GSH) with cisplatin, reducing its biological effects. In this study, three Pt(iv) complexes derived from cisplatin conjugated with a GST inhibitor (NBDHEX) were designed and synthesized. The stabilities and releasing capabilities of these complexes, as well as their abilities to inhibit GSTs, were investigated together with their in vitro anticancer activities toward osteosarcoma cells. Among them, complex 2, bearing one NBDHEX derivative and a hydroxyl group at the axial positions, could markedly kill human OS cells due to its suitable stability and prominent ability to inhibit GSTs. Meanwhile, it can prevent the metastasis of OS via down-regulating Akt. Thus, complex 2 has the potential for further research for the treatment of OS.
Collapse
Affiliation(s)
- Hong Chen
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, Southeast University, Nanjing 211189, China.
| | | | | | | |
Collapse
|
22
|
|
23
|
Yempala T, Babu T, Karmakar S, Nemirovski A, Ishan M, Gandin V, Gibson D. Expanding the Arsenal of Pt IV Anticancer Agents: Multi-action Pt IV Anticancer Agents with Bioactive Ligands Possessing a Hydroxy Functional Group. Angew Chem Int Ed Engl 2019; 58:18218-18223. [PMID: 31599054 DOI: 10.1002/anie.201910014] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Indexed: 12/16/2022]
Abstract
Most multi-action PtIV prodrugs have bioactive ligands containing carboxylates. This is probably due to the ease of carboxylating the OH axial ligands and because following reduction, the active drug is released. A major challenge is to expand the arsenal of bioactive ligands to include those without carboxylates. We describe a general approach for synthesis of PtIV prodrugs that release drugs with OH groups. We linked the OH groups of gemcitabine (Gem), paclitaxel (Tax), and estramustine (EM) to the PtIV derivative of cisplatin by a carbonate bridge. Following reduction, the axial ligands lost CO2 , rapidly generating the active drugs. In contrast, succinate-linked drugs did not readily release the free drugs. The carbonate-bridged ctc-[Pt(NH3 )2 (PhB)(Gem-Carb)Cl2 ] was significantly more cytotoxic than the succinate-bridged ctc-[Pt(NH3 )2 (PhB)(Gem-Suc)Cl2 ], and more potent and less toxic than gemcitabine, cisplatin, and co-administration of cisplatin and gemcitabine.
Collapse
Affiliation(s)
- Thirumal Yempala
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Tomer Babu
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Subhendu Karmakar
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Alina Nemirovski
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Maisaloon Ishan
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| | - Valentina Gandin
- Dipartimento di Scienze del Farmaco, Università di Padova, Via Marzolo 5, 35131, Padova, Italy
| | - Dan Gibson
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, 9112102, Jerusalem, Israel
| |
Collapse
|
24
|
Yempala T, Babu T, Karmakar S, Nemirovski A, Ishan M, Gandin V, Gibson D. Expanding the Arsenal of Pt
IV
Anticancer Agents: Multi‐action Pt
IV
Anticancer Agents with Bioactive Ligands Possessing a Hydroxy Functional Group. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201910014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Thirumal Yempala
- Institute for Drug ResearchSchool of PharmacyThe Hebrew University of Jerusalem 9112102 Jerusalem Israel
| | - Tomer Babu
- Institute for Drug ResearchSchool of PharmacyThe Hebrew University of Jerusalem 9112102 Jerusalem Israel
| | - Subhendu Karmakar
- Institute for Drug ResearchSchool of PharmacyThe Hebrew University of Jerusalem 9112102 Jerusalem Israel
| | - Alina Nemirovski
- Institute for Drug ResearchSchool of PharmacyThe Hebrew University of Jerusalem 9112102 Jerusalem Israel
| | - Maisaloon Ishan
- Institute for Drug ResearchSchool of PharmacyThe Hebrew University of Jerusalem 9112102 Jerusalem Israel
| | - Valentina Gandin
- Dipartimento di Scienze del FarmacoUniversità di Padova Via Marzolo 5 35131 Padova Italy
| | - Dan Gibson
- Institute for Drug ResearchSchool of PharmacyThe Hebrew University of Jerusalem 9112102 Jerusalem Israel
| |
Collapse
|
25
|
Shaili E, Salassa L, Woods JA, Clarkson G, Sadler PJ, Farrer NJ. Platinum(iv) dihydroxido diazido N-(heterocyclic)imine complexes are potently photocytotoxic when irradiated with visible light. Chem Sci 2019; 10:8610-8617. [PMID: 31803436 PMCID: PMC6844273 DOI: 10.1039/c9sc02644d] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 07/28/2019] [Indexed: 01/05/2023] Open
Abstract
A series of trans-di-(N-heterocyclic)imine dihydroxido diazido PtIV complexes of the form trans,trans,trans-[Pt(N3)2(OH)2(L1)(L2)] where L = pyridine, 2-picoline, 3-picoline, 4-picoline, thiazole and 1-methylimidazole have been synthesised and characterised, and their photochemical and photobiological activity evaluated. Notably, complexes 19 (L1 = py, L2 = 3-pic) and 26 (L1 = L2 = 4-pic) were potently phototoxic following irradiation with visible light (420 nm), with IC50 values of 4.0 μM and 2.1 μM respectively (A2780 cancer cell line), demonstrating greater potency than the previously reported complex 1 (L1 = L2 = py; 6.7 μM); whilst also being minimally toxic in the absence of irradiation. Complexes with mixed N-(heterocyclic)imine ligands 19 and 20 (L1 = py, L2 = 4-pic) were particularly photocytotoxic towards cisplatin resistant (A2780cis) cell lines. Complex 18 (L1 = py, L2 = 2-pic) was comparatively less photocytotoxic (IC50 value 14.5 μM) than the other complexes, despite demonstrating the greatest absorbance at the irradiation wavelength and the fastest half-life for loss of the N3 → Pt LMCT transition upon irradiation (λ irr = 463 nm) in aqueous solution. Complex 29 (X1 = X2 = thiazole) although potently phototoxic (2.4 μM), was also toxic towards cells in the absence of irradiation.
Collapse
Affiliation(s)
- Evyenia Shaili
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK
| | - Luca Salassa
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK
| | - Julie A Woods
- Photobiology Unit , Department of Dermatology and Photobiology , Ninewells Hospital , Dundee , DD1 9SY , UK
| | - Guy Clarkson
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK
| | - Peter J Sadler
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK
| | - Nicola J Farrer
- Chemistry Research Laboratory , University of Oxford , 12 Mansfield Road , Oxford , OX1 3TA , UK . ; ; Tel: +44 (0)1865 285131
| |
Collapse
|
26
|
Yao K, Bertran A, Howarth A, Goicoechea JM, Hare SM, Rees NH, Foroozandeh M, Bowen AM, Farrer NJ. A visible-light photoactivatable di-nuclear Pt IV triazolato azido complex. Chem Commun (Camb) 2019; 55:11287-11290. [PMID: 31475995 PMCID: PMC6984334 DOI: 10.1039/c9cc05310g] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 07/30/2019] [Indexed: 12/14/2022]
Abstract
A novel PtIV triazolato azido complex [3]-[N1,N3] has been synthesised via a strain-promoted double-click reaction (SPDC) between a PtIV azido complex (1) and the Sondheimer diyne (2). Photoactivation of [3]-[N1,N3] with visible light (452 nm) in the presence of 5'-guanosine monophosphate (5'-GMP) produced both PtIV and PtII 5'-GMP species; EPR spectroscopy confirmed the production of both azidyl and hydroxyl radicals. Spin-trapping of photogenerated radicals - particularly hydroxyl radicals - was significantly reduced in the presence of 5'-GMP.
Collapse
Affiliation(s)
- Kezi Yao
- Chemistry Research Laboratory
, University of Oxford
,
12 Mansfield Road
, Oxford
, OX1 3TA
, UK
.
; Tel: +44 (0)1865 285131
| | - Arnau Bertran
- Chemistry Research Laboratory
, University of Oxford
,
12 Mansfield Road
, Oxford
, OX1 3TA
, UK
.
; Tel: +44 (0)1865 285131
| | - Alison Howarth
- Chemistry Research Laboratory
, University of Oxford
,
12 Mansfield Road
, Oxford
, OX1 3TA
, UK
.
; Tel: +44 (0)1865 285131
| | - Jose M. Goicoechea
- Chemistry Research Laboratory
, University of Oxford
,
12 Mansfield Road
, Oxford
, OX1 3TA
, UK
.
; Tel: +44 (0)1865 285131
| | - Samuel M. Hare
- Chemistry Research Laboratory
, University of Oxford
,
12 Mansfield Road
, Oxford
, OX1 3TA
, UK
.
; Tel: +44 (0)1865 285131
| | - Nicholas H. Rees
- Chemistry Research Laboratory
, University of Oxford
,
12 Mansfield Road
, Oxford
, OX1 3TA
, UK
.
; Tel: +44 (0)1865 285131
| | - Mohammadali Foroozandeh
- Chemistry Research Laboratory
, University of Oxford
,
12 Mansfield Road
, Oxford
, OX1 3TA
, UK
.
; Tel: +44 (0)1865 285131
| | - Alice M. Bowen
- Chemistry Research Laboratory
, University of Oxford
,
12 Mansfield Road
, Oxford
, OX1 3TA
, UK
.
; Tel: +44 (0)1865 285131
| | - Nicola J. Farrer
- Chemistry Research Laboratory
, University of Oxford
,
12 Mansfield Road
, Oxford
, OX1 3TA
, UK
.
; Tel: +44 (0)1865 285131
| |
Collapse
|
27
|
Verma H, Singh Bahia M, Choudhary S, Kumar Singh P, Silakari O. Drug metabolizing enzymes-associated chemo resistance and strategies to overcome it. Drug Metab Rev 2019; 51:196-223. [DOI: 10.1080/03602532.2019.1632886] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Himanshu Verma
- MolecularModelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | | | - Shalki Choudhary
- MolecularModelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Pankaj Kumar Singh
- MolecularModelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Om Silakari
- MolecularModelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| |
Collapse
|
28
|
Ravera M, Gabano E, McGlinchey MJ, Osella D. A view on multi-action Pt(IV) antitumor prodrugs. Inorganica Chim Acta 2019. [DOI: 10.1016/j.ica.2019.04.025] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
29
|
Gabano E, Ravera M, Perin E, Zanellato I, Rangone B, McGlinchey MJ, Osella D. Synthesis and characterization of cyclohexane-1R,2R-diamine-based Pt(iv) dicarboxylato anticancer prodrugs: their selective activity against human colon cancer cell lines. Dalton Trans 2019; 48:435-445. [PMID: 30539948 DOI: 10.1039/c8dt03950j] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Three pairs of asymmetric dicarboxylato derivatives based on the cisplatin and oxaliplatin-like skeletons have been synthesized de novo or re-synthesized. The axial ligands consist of one medium-chain fatty acid (MCFA), namely clofibrate (i.e. 2-(p-chlorophenoxy)-2-methylpropionic acid, CA), heptanoate (HA) or octanoate (OA), respectively, and an inactive acetato ligand that imparts acceptable water solubility to such conjugates. Stability tests provided evidence for the partial formation of two hydrolyzed products, corresponding to two monoaqua diastereomers derived from the substitution of an equatorial chlorido ligand with a water molecule. The complexes have been tested on three different colon cancer cell lines having different histological history, and also on the cisplatin-sensitive A2780 ovarian cancer cell line for comparison. This allowed the evaluation not only of the increase in activity on passing from Pt(ii) to Pt(iv) derivatives, but also the selectivity towards colon cancer cells brought about by the cyclohexane-1R,2R-diamine carrier ligand.
Collapse
Affiliation(s)
- E Gabano
- Dipartimento di Scienze e Innovazione Tecnologica, Università del Piemonte Orientale, Viale Michel 11, 15121 Alessandria, Italy.
| | | | | | | | | | | | | |
Collapse
|
30
|
Chen H, Wang X, Gou S. A cisplatin-based platinum(IV) prodrug containing a glutathione s-transferase inhibitor to reverse cisplatin-resistance in non-small cell lung cancer. J Inorg Biochem 2019; 193:133-142. [PMID: 30731264 DOI: 10.1016/j.jinorgbio.2019.01.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/25/2019] [Accepted: 01/27/2019] [Indexed: 12/16/2022]
Abstract
A Pt(IV) prodrug of cisplatin containing a glutathione s-transferase (GSTs) inhibitor 6-(7-nitro-2,1,3-benzoxadiazol-4-ylthio)hexanol (NBDHEX), complex 1, was designed and studied aiming to overcome cisplatin-resistance and reduce its toxicity by inhibiting GSTs overexpressed in cancer cells. The complex could be reduced to release its active Pt(II) species and axial ligand in the presence of ascorbic acid. In cytotoxicity study, complex 1 showed more potent anticancer activity than cisplatin and NBDHEX against all the tested cancer cells, especially toward cisplatin resistant A549/DDP cells with a resistance factor value of 0.37. By effectively inhibiting GSTs, complex 1 was found to be able to promote higher platinum uptake and cause more severe DNA damage in both A549 cells and A549/DDP cells as compared with cisplatin. Further mechanism study indicated that it could trigger cell death via an apoptotic pathway. In vivo tests on A549 xenograft tumor mice model showed that complex 1 presented higher tumor inhibiting rate and lower toxicity than cisplatin as well. In all, the Pt(IV) prodrug has potential to be developed as an anticancer agent.
Collapse
Affiliation(s)
- Hong Chen
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, Southeast University, Nanjing 211189, China
| | - Xinyi Wang
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, Southeast University, Nanjing 211189, China
| | - Shaohua Gou
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, Southeast University, Nanjing 211189, China.
| |
Collapse
|
31
|
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: 409] [Impact Index Per Article: 81.8] [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
|
32
|
Montagner D, Tolan D, Andriollo E, Gandin V, Marzano C. A Pt(IV) Prodrug Combining Chlorambucil and Cisplatin: a Dual-Acting Weapon for Targeting DNA in Cancer Cells. Int J Mol Sci 2018; 19:ijms19123775. [PMID: 30486477 PMCID: PMC6321036 DOI: 10.3390/ijms19123775] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/24/2018] [Accepted: 11/26/2018] [Indexed: 12/25/2022] Open
Abstract
In this study, two DNA-targeting agents, cisplatin and chlorambucil, were combined in a Pt(IV) prodrug, 1, which was thoroughly characterized by means of spectroscopic and spectrometric techniques. Tested towards a panel of various human tumor cell lines, this compound showed superior in vitro antitumor potential than the reference drug cisplatin. In addition, an antitumor potential of 1 was found, which is comparable to that of oxaliplatin in 3D spheroid models of colon cancer cells. Mechanistic studies performed in colon cancer cells confirmed that the conjugation of chlorambucil to Pt(IV) cisplatin-based scaffold tunes the lipophilicity of the prodrug, consequently improving the ability of the compound to accumulate into cancer cells and to target DNA, ultimately leading to apoptotic cancer cell death.
Collapse
Affiliation(s)
- Diego Montagner
- Department of Chemistry, Maynooth University, Maynooth zip code, Ireland.
| | - Dina Tolan
- School of Chemistry, National University of Ireland Galway, Galway zip code, Ireland.
- Department of Chemistry, Faculty of Science, El-Menoufia University, Shebin El-Kom 32511, Egypt.
| | - Emma Andriollo
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Padova 35131, Italy.
| | - Valentina Gandin
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Padova 35131, Italy.
| | - Cristina Marzano
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Padova 35131, Italy.
| |
Collapse
|
33
|
Gibson D. Multi-action Pt(IV) anticancer agents; do we understand how they work? J Inorg Biochem 2018; 191:77-84. [PMID: 30471522 DOI: 10.1016/j.jinorgbio.2018.11.008] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 12/12/2022]
Abstract
Pt(IV) complexes act as prodrugs that are activated inside cancer cells releasing cytotoxic Pt(II) drugs such as cisplatin as well as two axial ligands. These ligands can be used to confer favorable pharmacological properties to the prodrug. They can be innocent spectators, targeting agents or bioactive moieties. When the ligands are bioactive moieties such as enzyme inhibitors or antiproliferative agents, the prodrug attacks several cellular targets at the same time acting as a multi-action prodrug. These compounds are very potent and often overcome resistance to cisplatin. Despite solid rationalization and careful design, often there is no correlation between the ability of the bioactive ligand to inhibit the target enzyme and the cytotoxicity. This might be because most bioactive ligands affect several cellular functions and not only the ones they were designed to inhibit. Thus, even "dual action" prodrugs might in reality be multi-action prodrugs. This class of multi-action Pt(IV) prodrugs seems to have great potential in the attempts to overcome resistance.
Collapse
Affiliation(s)
- Dan Gibson
- Institute for Drug Research, School of Pharmacy, The Hebrew University, Jerusalem, 91120, Israel.
| |
Collapse
|
34
|
Li X, Liu Y, Tian H. Current Developments in Pt(IV) Prodrugs Conjugated with Bioactive Ligands. Bioinorg Chem Appl 2018; 2018:8276139. [PMID: 30402082 PMCID: PMC6191961 DOI: 10.1155/2018/8276139] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/19/2018] [Accepted: 09/06/2018] [Indexed: 12/21/2022] Open
Abstract
To overcome the side effects of and resistance to cisplatin, a variety of Pt(IV) prodrugs were designed and synthesized via different modifications including combination with lipid chains to increase hydrophobicity, conjugation with short peptide chains or nanoparticles to improve drug delivery, or addition of bioactive ligands to the axial positions of Pt(IV) complexes to exert dual-function effects. This review summarizes the recent progress in the development of Pt(IV) prodrugs conjugated with bioactive-targeting ligands, including histone deacetylase inhibitors, p53 agonists, alkylating agents, and nonsteroidal anti-inflammatory agents. Although Pt(IV) complexes that conjugated with bioactive ligands show satisfactory anticancer effects, none has been approved for clinical use. Therefore, we hope that this review will contribute to further study and development of Pt(IV) complexes conjugated with bioactive and other ligands.
Collapse
Affiliation(s)
- Xuejiao Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yahong Liu
- Tianjin Binjiang Pharma, Inc., Tianjin 300192, China
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
35
|
Lo Re D, Montagner D, Tolan D, Di Sanza C, Iglesias M, Calon A, Giralt E. Increased immune cell infiltration in patient-derived tumor explants treated with Traniplatin: an original Pt(iv) pro-drug based on Cisplatin and Tranilast. Chem Commun (Camb) 2018; 54:8324-8327. [PMID: 29796549 DOI: 10.1039/c8cc02071j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Elevated intra-tumoral immune infiltrate is associated with an improved prognosis in cancer of distinct origins. Traniplatin (TPT) is a novel platinum(iv) pro-drug based on Cisplatin (CDDP) and the marketed drug Tranilast. When compared in vitro to Cisplatin, TPT showed increased cytotoxic activity against colon and lung cancer cells but decreased activity against immune cells. In addition, TPT efficiency was evaluated in tumor explants derived from colorectal cancer samples from patients subjected to intended curative surgery. TPT induced strong intra-tumoral cytotoxic activity yet was associated with an elevated presence of immune cell infiltrate, suggesting a reduced cytotoxic activity against immune cells in colorectal cancer.
Collapse
Affiliation(s)
- Daniele Lo Re
- Institute for Research in Biomedicine (IRB Barcelona), C/Baldiri Reixac 10, Barcelona, E-08028, Spain.
| | | | | | | | | | | | | |
Collapse
|