1
|
Kumarasamy K, Devendhiran T, Marthandam Asokan S, Ramaswamy M, Lin MC, Chien WJ, Kumar Ramasamy S, Huang CY. Synthesis and structural characterization of C,N-benzimidazole based ruthenium(II) complex with in vitro anticancer activity. INORG CHEM COMMUN 2023. [DOI: 10.1016/j.inoche.2023.110662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
|
2
|
Khater M, Brazier JA, Greco F, Osborn HMI. Anticancer evaluation of new organometallic ruthenium(ii) flavone complexes. RSC Med Chem 2023; 14:253-267. [PMID: 36846373 PMCID: PMC9945865 DOI: 10.1039/d2md00304j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/22/2022] [Indexed: 12/23/2022] Open
Abstract
Targeting multiple malignancy features such as angiogenesis, proliferation and metastasis with one molecule is an effective strategy in developing potent anticancer agents. Ruthenium metal complexation to bioactive scaffolds is reported to enhance their biological activities. Herein, we evaluate the impact of Ru chelation on the pharmacological activities of two bioactive flavones (1 and 2) as anticancer candidates. The novel Ru complexes (1Ru and 2Ru) caused a loss of their parent molecules' antiangiogenic activities in an endothelial cell tube formation assay. 1Ru enhanced the antiproliferative and antimigratory activities of its 4-oxoflavone 1 on MCF-7 breast cancer cells (IC50 = 66.15 ± 5 μM and 50% migration inhibition, p < 0.01 at 1 μM). 2Ru diminished 4-thioflavone's (2) cytotoxic activity on MCF-7 and MDA-MB-231 yet significantly enhanced 2's migration inhibition (p < 0.05) particularly on the MDA-MB-231 cell line. The test derivatives also showed non-intercalative interaction with VEGF and c-myc i-motif DNA sequences.
Collapse
Affiliation(s)
- Mai Khater
- School of Pharmacy, University of Reading Whiteknights Reading RG6 6AD UK .,Therapeutic Chemistry Department, Pharmaceutical & Drug Industries Research Division, National Research Centre Cairo Egypt
| | - John A. Brazier
- School of Pharmacy, University of ReadingWhiteknightsReadingRG6 6ADUK
| | - Francesca Greco
- School of Pharmacy, University of Reading Whiteknights Reading RG6 6AD UK
| | | |
Collapse
|
3
|
He SF, Liao JX, Huang MY, Zhang YQ, Zou YM, Wu CL, Lin WY, Chen JX, Sun J. Rhenium-guanidine complex as photosensitizer: trigger HeLa cell apoptosis through death receptor-mediated, mitochondria-mediated and cell cycle arrest pathways. Metallomics 2022; 14:6527583. [PMID: 35150263 DOI: 10.1093/mtomcs/mfac008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/27/2022] [Indexed: 11/12/2022]
Abstract
During the last decades, growing evidence indicates that the photodynamic antitumor activity of transition metal complexes, and Re(I) compounds are potential candidates for photodynamic therapy (PDT). This study reports the synthesis, characterization, and anti-tumor activity of three new Re(I)-guadinium complexes. Cytotoxicity tests reveal that complex Re1 increased cytotoxicity by 145-fold from IC50 > 180 μM in the dark to 1.3 ± 0.7 μM following 10 min of light irradiation (425 nm) in HeLa cells. Further, the mechanism by which Re1 induces apoptosis in the presence or absence of light irradiation was investigated, and results indicate that cell death was caused through different pathways. Upon irradiation, Re1 first accumulates on the cell membrane and interacts with death receptors to activate the extrinsic death receptor-mediated signaling pathway, then is transported into the cell cytoplasm. Most of the intracellular Re1 locates within mitochondria, improving the ROS level, and decreasing MMP and ATP levels, and inducing the activation of caspase-9 and, thus, apoptosis. Subsequently, the residual Re1 can translocate into the cell nucleus, and activates the p53 pathway, causing cell-cycle arrest and eventually cell death.
Collapse
Affiliation(s)
- Shu-Fen He
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.,Department of Pharmacy, Dongguan Peaple's Hospital, Dongguan, 523059, China
| | - Jia-Xin Liao
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Min-Ying Huang
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Yu-Qing Zhang
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Yi-Min Zou
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Ci-Ling Wu
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Wen-Yuan Lin
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Jia-Xi Chen
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Jing Sun
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| |
Collapse
|
4
|
Adibi H, Beyhaghi E, Hayati S, Hosseinzadeh L, Amin N. In Vitro Cytotoxicity and Apoptosis Inducing Evaluation of Novel Halogenated Isatin Derivatives. Anticancer Agents Med Chem 2022; 22:2439-2447. [PMID: 35043767 DOI: 10.2174/1871520622666220119091642] [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: 10/14/2021] [Revised: 11/04/2021] [Accepted: 12/07/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Isatin (1H-indole-2,3-dione) and its derivatives have been shown their responsibility in a wide range of biological activities. Among the range of beneficial properties, anticancer compounds were the most extensively highlighted and explored. OBJECTIVE Herein, we report the targeting effect of halogenated isatin derivatives on cancer cell mitochondria and their antiproliferative mechanism. METHOD A series of novel 5-halo-Isatin derivatives consisting 5-Amino-1,3,4-thiadiazole-2-thiol scaffold were synthesized and easily conducted in good yields through a condensation reaction between keto groups of Isatin and primary amine under alcoholic conditions, followed by S-benzylation. The compounds were fully characterized using spectroscopic methods such as 1H-NMR, FTIR, mass spectroscopy and then tested in vitro towards three cancer cell lines HT-29 (colon cancer), MCF-7 (breast cancer) and SKNMC (neuroblastoma). Apoptosis induction was investigated through assessment of caspase 3 and mitochondrial membrane potential. RESULTS The most potent compounds of 5b, 5r (IC50 = 18,13 µM) and 5n (IC50 = 20,17 µM) were found to show strong anticancer activity, especially for MCF7 cells. Further anticancer mechanism studies indicated that 5b and 5r induced apoptosis through the intrinsic mitochondrial pathway. CONCLUSION This research demonstrated that 5b and 5r have an anticancer property via the modulation of oxidative stress-mediated mitochondrial apoptosis and immune response, which deserves further studies on their clinical applications.
Collapse
Affiliation(s)
- Hadi Adibi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ehsan Beyhaghi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sonya Hayati
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Leila Hosseinzadeh
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Niloufar Amin
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
5
|
Chakraborty A, Roy S, Chakraborty MP, Roy SS, Purkait K, Koley TS, Das R, Acharya M, Mukherjee A. Cytotoxic Ruthenium(II) Complexes of Pyrazolylbenzimidazole Ligands That Inhibit VEGFR2 Phosphorylation. Inorg Chem 2021; 60:18379-18394. [PMID: 34780170 DOI: 10.1021/acs.inorgchem.1c02979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Eight new ruthenium(II) complexes of N,N-chelating pyrazolylbenzimidazole ligands of the general formula [RuII(p-cym)(L)X]+ [where the ligand L is 2-(1H-pyrazol-1-yl)-1H-benzo[d]imidazole (L1) substituted at the 4 position of the pyrazole ring by Cl (L2), Br (L3), or I (L4) and X = Cl- and I-] were synthesized and characterized using various analytical techniques. Complexes 1 and 3 were also characterized by single-crystal X-ray crystallography, and they crystallized as a monoclinic crystal system in space groups P21/n and P21/c, respectively. The complexes display good solution stability at physiological pH 7.4. The iodido-coordinated pyrazolylbenzimidazole ruthenium(II) p-cymene complexes (2, 4, 6, and 8) are more resistant toward hydrolysis and have less tendency to form monoaquated complexes in comparison to their chlorido analogues (1, 3, 5, and 7). The halido-substituted 2-(1H-pyrazol-1-yl)-1H-benzo[d]imidazole ligands, designed as organic-directing molecules, inhibit vascular endothelial growth factor receptor 2 (VEGFR2) phosphorylation. In addition, the ruthenium(II) complexes display a potential to bind to DNA bases. The cytotoxicity profile of the complexes (IC50 ca. 9-12 μM for 4-8) against the triple-negative breast cancer cells (MDA-MB-231) show that most of the complexes are efficient. The lipophilicity and cellular accumulation data of the complexes show a good correlation with the cytotoxicity profile of 1-8. The representative complexes 3 and 7 demonstrate the capability of arresting the cell cycle in the G2/M phase and induce apoptosis. The inhibition of VEGFR2 phosphorylation with the representative ligands L2 and L4 and the corresponding metal complexes 3 and 7 in vitro shows that the organic-directing ligands and their complexes inhibit VEGFR2 phosphorylation. Besides, L2, L4, 3, and 7 inhibit the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) and proto-oncogene tyrosine-protein kinase (Src), capable of acting downstream of VEGFR2 as well as independently. Compounds L2, L4, 3, and 7 have a lesser effect on ERK1/2 and more prominently affect Src phosphorylation. We extended the study for L2 and 3 in the Tg(fli1:gfp) zebrafish model and found that L2 is more effective in vivo compared to 3 in inhibiting angiogenesis.
Collapse
|
6
|
Chen L, Wang J, Cai X, Chen S, Zhang J, Li B, Chen W, Guo X, Luo H, Chen J. Cyclometalated Ru(II)-isoquinoline complexes overcome cisplatin resistance of A549/DDP cells by downregulation of Nrf2 via Akt/GSK-3β/Fyn pathway. Bioorg Chem 2021; 119:105516. [PMID: 34856444 DOI: 10.1016/j.bioorg.2021.105516] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 11/18/2021] [Accepted: 11/21/2021] [Indexed: 01/03/2023]
Abstract
Both ruthenium (Ru) and isoquinoline (IQ) compounds are regarded as potential anticancer drug candidates. Here, we report the synthesis and characterization of three novel cyclometalated Ru(II)-isoquinoline complexes: RuIQ-3, RuIQ-4, and RuIQ-5, and evaluation of their in vitro cytotoxicities against a panel of cell lines including A549/DDP, a cisplatin-resistant human lung cancer cell line. A549/DDP 3D multicellular tumor spheroids (MCTSs) were also used to detect the drug resistance reversal effect of Ru(II)-IQ complexes. Our results indicated that the cytotoxic activities against cancer cells of Ru(II)-IQ complexes, especially RuIQ-5, were superior compared with cisplatin. In addition, RuIQ-5 exhibited low toxicity towards both normal HBE cells in vitro and zebrafish embryos in vivo. Further investigation on cellular mechanism of action indicated that after absorption by A549/DDP cells, RuIQ-5 was mainly distributed in the nucleus, which is different from cisplatin. Besides, RuIQ-5 could induce apoptosis through mitochondrial dysfunction, reactive oxygen species (ROS) accumulation, ROS-mediated DNA damage, and cycle arrest at both S and G2/M phases. Moreover, RuIQ-5 could inhibit the overexpression of Nrf2 through regulation of Akt/GSK-3β/Fyn signaling pathway and hindering the nuclear translocation of Nrf2. Based on these findings, we firmly believe that the studied Ru(II)-IQ complexes hold great promise as anticancer therapeutics with high effectiveness and low toxicity.
Collapse
Affiliation(s)
- Lanmei Chen
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, Guangdong 524023, China
| | - Jie Wang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China
| | - Xianhong Cai
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China
| | - Suxiang Chen
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Western Australia 6150, Australia
| | - Jingjing Zhang
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang 524001, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, China
| | - Baojun Li
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China
| | - Weigang Chen
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China
| | - Xinhua Guo
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China
| | - Hui Luo
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, Guangdong 524023, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, China.
| | - Jincan Chen
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, Guangdong 524023, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|
7
|
Mello-Andrade F, Guedes APM, Pires WC, Velozo-Sá VS, Delmond KA, Mendes D, Molina MS, Matuda L, de Sousa MAM, Melo-Reis P, Gomes CC, Castro CH, Almeida MAP, Menck CFM, Batista AA, Burikhanov R, Rangnekar VM, Silveira-Lacerda E. Ru(II)/amino acid complexes inhibit the progression of breast cancer cells through multiple mechanism-induced apoptosis. J Inorg Biochem 2021; 226:111625. [PMID: 34655962 DOI: 10.1016/j.jinorgbio.2021.111625] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/20/2022]
Abstract
For some cancer subtypes, such as triple-negative breast cancer, there are no specific therapies, which leads to a poor prognosis associated with invasion and metastases. Ruthenium complexes have been developed to act in all steps of tumor growth and its progression. In this study, we investigated the effects of Ruthenium (II) complexes coupled to the amino acids methionine (RuMet) and tryptophan (RuTrp) on the induction of cell death, clonogenic survival ability, inhibition of angiogenesis, and migration of MDA-MB-231 cells (human triple-negative breast cancer). The study also demonstrated that the RuMet and RuTrp complexes induce cell cycle blockage and apoptosis of MDA-MB-231 cells, as evidenced by an increase in the number of Annexin V-positive cells, p53 phosphorylation, caspase 3 activation, and poly(ADP-ribose) polymerase cleavage. Moreover, morphological changes and loss of mitochondrial membrane potential were detected. The RuMet and RuTrp complexes induced DNA damage probably due to reactive oxygen species production related to mitochondrial membrane depolarization. Therefore, the RuMet and RuTrp complexes acted directly on breast tumor cells, leading to cell death and inhibiting their metastatic potential; this reveals the potential therapeutic action of these drugs.
Collapse
Affiliation(s)
- Francyelli Mello-Andrade
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás 74690-900, Brazil; Department of Chemistry, Federal Institute of Education, Science and Technology of Goiás, Goiânia, Goiás 74055-110, Brazil.
| | - Adriana P M Guedes
- Department of Chemistry, Federal University of São Carlos, São Carlos, SP 13565-905, Brazil
| | - Wanessa C Pires
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás 74690-900, Brazil
| | - Vivianne S Velozo-Sá
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás 74690-900, Brazil
| | - Kezia A Delmond
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás 74690-900, Brazil
| | - Davi Mendes
- Department of Microbiology, Laboratory of DNA Repair, Institute of Biomedical Sciences, University of São Paulo - USP, São Paulo, SP 05508-900, Brazil
| | - Matheus S Molina
- Department of Microbiology, Laboratory of DNA Repair, Institute of Biomedical Sciences, University of São Paulo - USP, São Paulo, SP 05508-900, Brazil
| | - Larissa Matuda
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás - UFG, Goiânia, GO 74690-900, Brazil
| | | | - Paulo Melo-Reis
- Departament of Biomedicine, Pontifical Catholic University of Goiás, Goiânia, GO, Brazil
| | - Clever C Gomes
- Department of Morphology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO 74690-900, Brazil
| | - Carlos Henrique Castro
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás - UFG, Goiânia, GO 74690-900, Brazil
| | - Márcio Aurélio P Almeida
- Coordination of Science and Technology, Federal University of Maranhão, São Luís, MA 65080-805, Brazil
| | - Carlos F M Menck
- Department of Microbiology, Laboratory of DNA Repair, Institute of Biomedical Sciences, University of São Paulo - USP, São Paulo, SP 05508-900, Brazil
| | - Alzir A Batista
- Department of Chemistry, Federal University of São Carlos, São Carlos, SP 13565-905, Brazil
| | - Ravshan Burikhanov
- Department of Radiation Medicine, University of Kentucky, Lexington, KY 40536, United States of America
| | - Vivek M Rangnekar
- Department of Radiation Medicine, University of Kentucky, Lexington, KY 40536, United States of America; L. P. Markey Cancer Center, University of Kentucky, Lexington, KY 40536, United States of America
| | - Elisângela Silveira-Lacerda
- Department of Genetics, Laboratory of Molecular Genetics and Cytogenetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Goiás 74690-900, Brazil.
| |
Collapse
|
8
|
Zhou L, Liu H, Liu K, Wei S. Gold Compounds and the Anticancer Immune Response. Front Pharmacol 2021; 12:739481. [PMID: 34588987 PMCID: PMC8473785 DOI: 10.3389/fphar.2021.739481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/31/2021] [Indexed: 12/20/2022] Open
Abstract
Gold compounds are not only well-explored for cytotoxic effects on tumors, but are also known to interact with the cancer immune system. The immune system deploys innate and adaptive mechanisms to protect against pathogens and prevent malignant transformation. The combined action of gold compounds with the activated immune system has shown promising results in cancer therapy through in vivo and in vitro experiments. Gold compounds are known to induce innate immune responses; however, these responses may contribute to adaptive immune responses. Gold compounds play the role of a major hapten that acts synergistically in innate immunity. Gold compounds support cancer cell antigenicity and promote anti-tumor immune response by inducing the release of CRT, ATP, HMGB1, HSP, and NKG2D to enhance immunogenicity. Gold compounds affect various immune cells (including suppressor regulatory T cells), inhibit myeloid derived suppressor cells, and enhance the function and number of dendritic cells. Gold nanoparticles (AuNPs) have potential for improving the effect of immunotherapy and reducing the toxicity and side effects of the treatment process. Thus, AuNPs provide an ideal opportunity for exploring the combination of anticancer gold compounds and immunotherapeutic interventions.
Collapse
Affiliation(s)
- Ling Zhou
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Kui Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Cyclometalated Ru(II) β-carboline complexes induce cell cycle arrest and apoptosis in human HeLa cervical cancer cells via suppressing ERK and Akt signaling. J Biol Inorg Chem 2021; 26:793-808. [PMID: 34459988 DOI: 10.1007/s00775-021-01894-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/17/2021] [Indexed: 10/20/2022]
Abstract
Two new cyclometalated Ru(II)-β-carboline complexes, [Ru(dmb)2(Cl-Ph-βC)](PF6) (dmb = 4,4'-dimethyl-2,2'-bipyridine; Cl-Ph-βC = Cl-phenyl-9H-pyrido[3,4-b]indole; RuβC-3) and [Ru(bpy)2(Cl-Ph-βC)](PF6) (bpy = 2,2'-bipyridine; RuβC-4) were synthesized and characterized. The Ru(II) complexes display high cytotoxicity against HeLa cells, the stabilized human cervical cancer cell, with IC50 values of 3.2 ± 0.4 μM (RuβC-3) and 4.1 ± 0.6 μM (RuβC-4), which were considerably lower than that of non-cyclometalated Ru(II)-β-carboline complex [Ru(bpy)2(1-Py-βC)] (PF6)2 (61.2 ± 3.9 μM) by 19- and 15-folds, respectively. The mechanism studies indicated that both Ru(II) complexes could significantly inhibit HeLa cell migration and invasion, and effectively induce G0/G1 cell cycle arrest. The new Ru(II) complexes could also trigger apoptosis through activating caspase-3 and poly (ADP-ribose) polymerase (PARP), increasing the Bax/Bcl-2 ratio, enhancing reactive oxygen species (ROS) generation, decreasing mitochondrial membrane potential (MMP), and inducing cytochrome c release from mitochondria. Further research revealed that RuβC-3 could deactivate the ERK/Akt signaling pathway thus inhibiting HeLa cell invasion and migration, and inducing apoptosis. In addition, RuβC-3-induced apoptosis in HeLa cells was closely associated with the increase of intracellular ROS levels, which may act as upstream factors to regulate ERK and Akt pathways. More importantly, RuβC-3 exhibited low toxicity on both normal BEAS-2B cells in vitro and zebrafish embryos in vivo. Consequently, the developed Ru(II) complexes have great potential on developing novel low-toxic anticancer drugs.
Collapse
|
10
|
Dorairaj DP, Lin YF, Haribabu J, Murugan T, Narwane M, Karvembu R, Neelakantan MA, Kao CL, Chiu CC, Hsu SCN. Binding mode transformation and biological activity on the Ru(II)-DMSO complexes bearing heterocyclic pyrazolyl ligands. J Inorg Biochem 2021; 223:111545. [PMID: 34303108 DOI: 10.1016/j.jinorgbio.2021.111545] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/28/2021] [Accepted: 07/12/2021] [Indexed: 12/19/2022]
Abstract
Three Ru(II)-DMSO complexes (1-3) containing 2-(3-pyrazolyl)pyridine (PzPy), 2-pyrazol-3-ylfuran (PzO), or 2-pyrazol-3-ylthiophene (PzS) ligand, were synthesized and characterized. The monodentate coordination of the heterocyclic pyrazolyl ligand (PzPy) with Ru(II) ion via N atom was confirmed by single crystal X-ray diffraction. Complex 1 could be converted to the known η2-bidentate PzPy complex cis(Cl), cis(S)-[RuCl2(PzPy)(DMSO)2] (4) under reflux conditions. The mechanism underlying binding mode transformation was studied by 1H NMR spectroscopy and density functional theory (DFT) calculations. The binding abilities of the complexes (1-4) with calf-thymus (CT) DNA and bovine serum albumin (BSA) were investigated using spectroscopic and molecular docking techniques. Among the four Ru(II) complexes, complexes 1 and 3 inhibited the long-term proliferation of human breast cancer cells, whereas complexes 2 and 4 did not inhibit their proliferation to a considerable extent. Interestingly, complexes 1 and 3 did not induce significant cell death but rather attenuated the clonogenicity of breast cancer cells by upregulating reactive oxygen species (ROS), endoplasmic reticulum (ER) and autophagic stress.
Collapse
Affiliation(s)
- Dorothy Priyanka Dorairaj
- Department of Medicinal and Applied Chemistry, Drug Development and Value Creation Research Centre, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Chemistry, National Institute of Technology, Tiruchirappalli 620 015, India
| | - Ya-Fan Lin
- Department of Medicinal and Applied Chemistry, Drug Development and Value Creation Research Centre, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Fragrance and Cosmetic Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jebiti Haribabu
- Department of Medicinal and Applied Chemistry, Drug Development and Value Creation Research Centre, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Chemistry, National Institute of Technology, Tiruchirappalli 620 015, India
| | - Theetharappan Murugan
- Chemistry Research Centre, National Engineering College, K.R. Nagar, Kovilpatti, 628503 Thoothukudi District, Tamilnadu, India
| | - Manmath Narwane
- Department of Medicinal and Applied Chemistry, Drug Development and Value Creation Research Centre, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ramasamy Karvembu
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620 015, India
| | | | - Chai-Lin Kao
- Department of Medicinal and Applied Chemistry, Drug Development and Value Creation Research Centre, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chien-Chih Chiu
- Department of Medical Research, Kaohsiung Medical University Hospital, Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Sodio C N Hsu
- Department of Medicinal and Applied Chemistry, Drug Development and Value Creation Research Centre, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
11
|
Liu C, Lai H, Chen T. Boosting Natural Killer Cell-Based Cancer Immunotherapy with Selenocystine/Transforming Growth Factor-Beta Inhibitor-Encapsulated Nanoemulsion. ACS NANO 2020; 14:11067-11082. [PMID: 32806028 DOI: 10.1021/acsnano.9b10103] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Natural killer (NK) cell-based immunotherapy represents a promising strategy to overcome the bottlenecks of cancer treatment. However, the therapeutic efficacy is greatly limited by downregulation of recognition ligands on the tumor cell surface, and the immunosuppressive effects can be thwarted by the tumor microenvironment such as secretion of transforming growth factor-beta (TGF-β), which could stunt the NK cell-mediated immune response. To overcome these limitations, herein we developed a nanoemulsion system (SSB NMs) to co-deliver TGF-β inhibitor and selenocysteine (SeC) to achieve amplified anticancer efficacy. SSB NMs significantly enhanced the lytic potency of NK92 cells by 2.1-fold. Moreover, a subtoxic dose of SSB NMs effectively sensitized MDA-MB-231 triple-negative breast cancer (TNBC) cells to NK cells derived from seven clinical patients, resulting in an up to 13.8-fold increase in cancer lysis. Mechanistic studies reveal that the sensitizing effects relied on natural killer group 2, member D (NKG2D)/NKG2D ligands (NKG2DLs) signaling with the involvement of DNA damage response. SSB NMs also effectively restrained TGF-β/TGF-β RI/Smad2/3 signaling, which thus enhanced NKG2DL expression on tumor cells and stimulated NKG2D surface expression on NK92 cells, ultimately contributing to the enhanced immune response. Furthermore, SSB NMs sustained release of SeC and TGF-β inhibitor and synergized with NK92 cells to induce significant anticancer effects in vivo. Together, this study not only demonstrates a simple strategy for the design of a nanoemulsion to co-deliver synergistic drugs but also sheds light on the application and action mechanisms in NK cell adaptive therapy against breast cancer, especially TNBCs.
Collapse
Affiliation(s)
- Chang Liu
- The First Affiliated Hospital and Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Haoqiang Lai
- The First Affiliated Hospital and Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Tianfeng Chen
- The First Affiliated Hospital and Department of Chemistry, Jinan University, Guangzhou 510632, China
| |
Collapse
|
12
|
Chen J, Wang J, Deng Y, Li B, Li C, Lin Y, Yang D, Zhang H, Chen L, Wang T. Novel cyclometalated Ru(II) complexes containing isoquinoline ligands: Synthesis, characterization, cellular uptake and in vitro cytotoxicity. Eur J Med Chem 2020; 203:112562. [PMID: 32698112 DOI: 10.1016/j.ejmech.2020.112562] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/29/2020] [Accepted: 06/10/2020] [Indexed: 01/25/2023]
Abstract
Two novel cyclometalated Ru(II) complexes containing isoquinoline ligand, [Ru(bpy)2(1-Ph-IQ)](PF6), (bpy = 2,2'-bipyridine; 1-Ph-IQ = 1-phenylisoquinoline; RuIQ-1) and [Ru(phen)2(1-Ph-IQ)](PF6) (phen = 1,10-phenanthroline; RuIQ-2) were found to show high cytotoxic activity against NCI-H460, A549, HeLa and MCF-7 cell lines. Notably, both of them exhibited IC50 values that were an order of magnitude lower than those of clinical cisplatin and two structurally similar Ru(II)-isoquinoline complexes [Ru(bpy)2(1-Py-IQ)](PF6)2 (Ru3) and [Ru(phen)2(1-Py-IQ)](PF6)2 (Ru4) (1-Py-IQ = 1-pyridine-2-yl). The cellular uptake and intracellular localization displayed that the two cyclometalated Ru(II) complexes entered NCI-H460 cancer cells dominantly via endocytosis pathway, and preferentially distributed in the nucleus. Further investigations on the apoptosis-inducing mechanisms of RuIQ-1 and RuIQ-2 revealed that the two complexes could cause S, G2/M double-cycle arrest by regulating cell cycle related proteins. The two complexes also could reduce the mitochondrial membrane potential (MMP), promote the generation of intracellular ROS and trigger DNA damage, and then lead to apoptosis-mediated cell death. More importantly, RuIQ-2 exhibits low toxicity both towards normal HBE cells in vitro and zebrafish embryos in vivo. Accordingly, the developed complexes hold great potential to be developed as novel therapeutics for effective and low-toxic cancer treatment.
Collapse
Affiliation(s)
- Jincan Chen
- Guangdong Key Laboratory for Research and Development of Nature Drugs, Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, China
| | - Jie Wang
- Guangdong Key Laboratory for Research and Development of Nature Drugs, Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, China
| | - Yuanyuan Deng
- Guangdong Key Laboratory for Research and Development of Nature Drugs, Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, China
| | - Baojun Li
- Guangdong Key Laboratory for Research and Development of Nature Drugs, Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, China
| | - Chengpeng Li
- The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, China
| | - Yuxue Lin
- Guangdong Key Laboratory for Research and Development of Nature Drugs, Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, China
| | - Dongbin Yang
- The Affiliated People's Hospital of Hebi of Henan University, Hebi, 456030, China
| | - Huanyun Zhang
- The Affiliated People's Hospital of Hebi of Henan University, Hebi, 456030, China
| | - Lanmei Chen
- Guangdong Key Laboratory for Research and Development of Nature Drugs, Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, China.
| | - Tao Wang
- The College of Nursing and Health, Zhengzhou University, Zhengzhou, 450001, China; Centre for Comparative Genomics, Murdoch University, Perth, WA, 6150, Australia.
| |
Collapse
|
13
|
Shu L, Ren L, Wang Y, Fang T, Ye Z, Han W, Chen C, Wang H. Niacin-ligated platinum(iv)-ruthenium(ii) chimeric complexes synergistically suppress tumor metastasis and growth with potentially reduced toxicity in vivo. Chem Commun (Camb) 2020; 56:3069-3072. [PMID: 32049075 DOI: 10.1039/c9cc09016a] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Niacin-ligated platinum(iv)-ruthenium(ii) chimeric complexes (PtRu 1-4) have been synthesized and evaluated for their antitumor performance. Using the optimal complex, PtRu-1, we show that this water-soluble chimeric prodrug not only potently inhibits the metastasis and proliferation of tumor cells but also has an unexpectedly higher safety margin in animals compared with the traditionally-used, clinically approved drug cisplatin.
Collapse
Affiliation(s)
- Liwei Shu
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China. and Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, P. R. China
| | - Lulu Ren
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, P. R. China
| | - Yuchen Wang
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China. and Department of Chemical Engineering, Zhejiang, University, Hangzhou, P. R. China
| | - Tao Fang
- Jinhua People's Hospital, Jinhua, Zhejiang Province 321000, P. R. China
| | - Zhijian Ye
- Jinhua People's Hospital, Jinhua, Zhejiang Province 321000, P. R. China
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, P. R. China
| | - Chao Chen
- College of Life Sciences, Huzhou University, Huzhou, 313000, P. R. China.
| | - Hangxiang Wang
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China.
| |
Collapse
|
14
|
Synthesis, crystal structures and antioxidant studies of Pd(II) and Ru(II) complexes of 2-(4-methoxyphenyltelluro) ethanol. J Organomet Chem 2019. [DOI: 10.1016/j.jorganchem.2019.120967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
15
|
Lai H, Zeng D, Liu C, Zhang Q, Wang X, Chen T. Selenium-containing ruthenium complex synergizes with natural killer cells to enhance immunotherapy against prostate cancer via activating TRAIL/FasL signaling. Biomaterials 2019; 219:119377. [DOI: 10.1016/j.biomaterials.2019.119377] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/20/2019] [Accepted: 07/22/2019] [Indexed: 12/14/2022]
|
16
|
Zhu H, Dai C, He L, Xu A, Chen T. Iron (II) Polypyridyl Complexes as Antiglioblastoma Agents to Overcome the Blood-Brain Barrier and Inhibit Cell Proliferation by Regulating p53 and 4E-BP1 Pathways. Front Pharmacol 2019; 10:946. [PMID: 31551768 PMCID: PMC6733960 DOI: 10.3389/fphar.2019.00946] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 07/24/2019] [Indexed: 12/28/2022] Open
Abstract
Background and Purpose: It is urgently required to develop promising candidates to permeate across blood-brain barrier (BBB) efficiently with simultaneous disrupting vasculogenic mimicry capability of gliomas. Previously, a series of iron (II) complexes were synthesized through a modified method. Hence, the aim of this study was to evaluate anticancer activity of Fe(PIP)3SO4 against glioma cancer cells. Methods: Cytotoxic effects were determined via MTT assay, and IC50 values were utilized to evaluate the cytotoxicity. Cellular uptake of Fe(PIP)3SO4 between U87 and HEB cells was conducted by subtracting content of the complex remaining in the cell culture supernatants. Propidium Iodide (PI)-flow cytometric analysis was used to analyze cell cycle proportion of U87 cells treated with Fe(PIP)3SO4. The reactive oxygen species levels induced by Fe(PIP)3SO4 were measured by 2'-deoxycoformycin (DCF) probe; ABTS assay was utilized to examine the radical scavenge capacity of Fe(PIP)3SO4. To study the bind efficiency to thioredoxin reductase (TrxR), Fe(PIP)3SO4 was introduced into solution containing TrxR. To verify if Fe(PIP)3SO4 could penetrate BBB, HBMEC/U87 coculture as BBB model was established, and penetrating capability of Fe(PIP)3SO4 was tested. In vitro U87 tumor spheroids were formed to test the permeating ability of Fe(PIP)3SO4. Acute toxicity and biodistribution of Fe(PIP)3SO4 were tested on mice for 72 h. Protein profiles associated with U87 cells treated with Fe(PIP)3SO4 were determined by Western blotting analysis. Results: Results showed that Fe(PIP)3SO4 could suppress cell proliferation by inducing G2/M phase cycle retardation and apoptotic pathways, which was related with expression of p53 and initiation factor 4E binding protein 1. In addition, Fe complex could suppress cell proliferation by downregulating reactive oxygen species levels via scavenging free radicals and interaction with TrxR. Furthermore, Fe(PIP)3SO4 could permeate across BBB and simultaneously inhibited the vasculogenic mimicry-channel of U87 cells, suggesting favorable antiglioblastoma efficacy. Acute toxicity manifested lower degree of the complex compared with cisplatin and temozolomide. Conclusion: Fe(PIP)3SO4 exhibited favorable anticancer activity against glioma cells associated with p53 and 4E binding protein 1, accompanied with negligible toxic effects on normal tissues. Herein, Fe(PIP)3SO4 could be developed as a promising metal-based chemotherapeutic agent to overcome BBB and antagonize glioblastomas.
Collapse
Affiliation(s)
- Huili Zhu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Chengli Dai
- The First Affiliated Hospital and the Department of Chemistry, Jinan University, Guangzhou, China
| | - Lizhen He
- The First Affiliated Hospital and the Department of Chemistry, Jinan University, Guangzhou, China
| | - Anding Xu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Tianfeng Chen
- The First Affiliated Hospital and the Department of Chemistry, Jinan University, Guangzhou, China
| |
Collapse
|
17
|
Rausch M, Dyson PJ, Nowak‐Sliwinska P. Recent Considerations in the Application of RAPTA‐C for Cancer Treatment and Perspectives for Its Combination with Immunotherapies. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900042] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Magdalena Rausch
- Molecular Pharmacology GroupSchool of Pharmaceutical Sciences, Faculty of SciencesUniversity of Lausanne and University of Geneva Rue Michel‐Servet 1, 1211 Geneva 4 Switzerland
| | - Paul J. Dyson
- Institute of Chemical Sciences and EngineeringEcole Polytechnique Fédérale de Lausanne (EPFL) 1015 Lausanne Switzerland
| | - Patrycja Nowak‐Sliwinska
- Molecular Pharmacology GroupSchool of Pharmaceutical Sciences, Faculty of SciencesUniversity of Lausanne and University of Geneva Rue Michel‐Servet 1, 1211 Geneva 4 Switzerland
- Translational Research Centre in Oncohaematology Geneva, Switzerland, 1211 Geneva 4 Switzerland
| |
Collapse
|
18
|
Ruthenium(II) salicylate complexes inducing ROS-mediated apoptosis by targeting thioredoxin reductase. J Inorg Biochem 2019; 193:112-123. [DOI: 10.1016/j.jinorgbio.2019.01.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 01/19/2019] [Accepted: 01/20/2019] [Indexed: 12/16/2022]
|
19
|
Abstract
After nearly 20 years of research on the use of ruthenium in the fight against cancer, only two Ru(III) coordination complexes have advanced to clinical trials. During this time, the field has produced excellent candidate drugs with outstanding in vivo and in vitro activity; however, we have yet to find a ruthenium complex that would be a viable alternative to platinum drugs currently used in the clinic. We aimed to explore what we have learned from the most prominent complexes in the area, and to challenge new concepts in chemical design. Particularly relevant are studies involving NKP1339, NAMI-A, RM175, and RAPTA-C, which have paved the way for current research. We explored the development of the ruthenium anticancer field considering that the mechanism of action of complexes no longer focuses solely on DNA interactions, but explores a diverse range of cellular targets involving multiple chemical strategies.
Collapse
|
20
|
Qin QP, Wang SL, Tan MX, Luo DM, Wang ZF, Wei QM, Wu XY, Zou BQ, Liu YC. 3-(1H-benzoimidazol-2-yl)-chromen-2-ylideneamine platinum(II) and ruthenium(II) complexes exert their high in vitro antitumor activity by inducing S-phase arrest and disrupting mitochondrial functions in SK-OV-3/DDP tumor cells. Polyhedron 2019. [DOI: 10.1016/j.poly.2018.10.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
21
|
Englinger B, Pirker C, Heffeter P, Terenzi A, Kowol CR, Keppler BK, Berger W. Metal Drugs and the Anticancer Immune Response. Chem Rev 2018; 119:1519-1624. [DOI: 10.1021/acs.chemrev.8b00396] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Bernhard Englinger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Christine Pirker
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Alessio Terenzi
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Christian R. Kowol
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Bernhard K. Keppler
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
| |
Collapse
|
22
|
Lai H, Fu X, Sang C, Hou L, Feng P, Li X, Chen T. Selenadiazole Derivatives Inhibit Angiogenesis-Mediated Human Breast Tumor Growth by Suppressing the VEGFR2-Mediated ERK and AKT Signaling Pathways. Chem Asian J 2018; 13:1447-1457. [DOI: 10.1002/asia.201800110] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 03/08/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Haoqiang Lai
- Department of Chemistry; Jinan University; Guangzhou 510632 China
| | - Xiaoyan Fu
- Department of Chemistry; Jinan University; Guangzhou 510632 China
| | - Chengcheng Sang
- Department of Chemistry; Jinan University; Guangzhou 510632 China
| | - Liyuan Hou
- Department of Chemistry; Jinan University; Guangzhou 510632 China
| | - Pengju Feng
- Department of Chemistry; Jinan University; Guangzhou 510632 China
| | - Xiaoling Li
- Institute of Food Safety and Nutrition; Jinan University; Guangzhou 510632 China
| | - Tianfeng Chen
- Department of Chemistry; Jinan University; Guangzhou 510632 China
| |
Collapse
|
23
|
Fang X, Li C, Zheng L, Yang F, Chen T. Dual-Targeted Selenium Nanoparticles for Synergistic Photothermal Therapy and Chemotherapy of Tumors. Chem Asian J 2018; 13:996-1004. [DOI: 10.1002/asia.201800048] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/01/2018] [Indexed: 11/11/2022]
Affiliation(s)
- Xueyang Fang
- Department of Chemistry; Jinan University; Guangzhou 510632 P.R. China
| | - Chang'e Li
- Department of Chemistry; Jinan University; Guangzhou 510632 P.R. China
| | - Lan Zheng
- Department of Chemistry; Jinan University; Guangzhou 510632 P.R. China
| | - Fang Yang
- Department of Chemistry; Jinan University; Guangzhou 510632 P.R. China
| | - Tianfeng Chen
- Department of Chemistry; Jinan University; Guangzhou 510632 P.R. China
| |
Collapse
|
24
|
Mohan N, Mohamed Subarkhan MK, Ramesh R. Synthesis, antiproliferative activity and apoptosis-promoting effects of arene ruthenium(II) complexes with N, O chelating ligands. J Organomet Chem 2018. [DOI: 10.1016/j.jorganchem.2018.01.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
25
|
Anticancer Efficacy of Targeted Shikonin Liposomes Modified with RGD in Breast Cancer Cells. Molecules 2018; 23:molecules23020268. [PMID: 29382149 PMCID: PMC6017468 DOI: 10.3390/molecules23020268] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 01/22/2018] [Accepted: 01/26/2018] [Indexed: 12/04/2022] Open
Abstract
Shikonin (SHK) has been proven to have a good anti-tumor effect. However, poor water solubility and low bioavailability limit its wide application in clinical practice. In this study, to overcome these drawbacks, RGD-modified shikonin-loaded liposomes (RGD-SSLs-SHK) were successfully prepared. It exhibited excellent physicochemical characteristics including particle size, zeta potential, encapsulation efficiency, and delayed release time. Meanwhile, the targeting activity of the RGD-modified liposomes was demonstrated by flow cytometry and confocal microscopy in the αvβ3-positive MDA-MB-231 cells. Besides exhibiting greater cytotoxicity in vitro, compared with non-targeted shikonin-loaded liposomes (SSLs-SHK), RGD-SSLs-SHK could also evidently induce apoptosis by decreasing the expression of Bcl-2 and increasing the expression of Bax. It could also inhibit cell proliferation, migration, invasion, and adhesion by reducing the expression of MMP-9 and the level of NF-κB p65, but did not affect the expression of MMP-2 in the MDA-MB-231 cells. Therefore, these findings indicated that the strategy to use RGD-modified liposomes as carriers for targeted delivery of shikonin is a very promising approach to achieve breast cancer targeted therapy.
Collapse
|
26
|
Chen J, Zhang Y, Li G, Peng F, Jie X, She J, Dongye G, Zou Z, Rong S, Chen L. Cytotoxicity in vitro, cellular uptake, localization and apoptotic mechanism studies induced by ruthenium(II) complex. J Biol Inorg Chem 2017; 23:261-275. [DOI: 10.1007/s00775-017-1528-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/13/2017] [Indexed: 12/22/2022]
|
27
|
Synthesis, characterization, cellular uptake and apoptosis-inducing properties of two highly cytotoxic cyclometalated ruthenium(II) β-carboline complexes. Eur J Med Chem 2017; 140:104-117. [PMID: 28923379 DOI: 10.1016/j.ejmech.2017.09.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/26/2017] [Accepted: 09/04/2017] [Indexed: 01/08/2023]
Abstract
Two new cyclometalated Ru(II) complexes of the general formula [Ru(N-N)2(1-Ph-βC)](PF6), where N-N = 4,4'-dimethyl-2,2'-bipyridine (dmb, Ru1), 2,2'-bipyridine (bpy, Ru2), and 1-Ph-βC (1-phenyl-9H-pyrido[3,4-b]indole) is a β-carboline alkaloids derivatives, have been synthesized and characterized. The in vitro cytotoxicities, cellular uptake and localization, cell cycle arrest and apoptosis-inducing mechanisms of these complexes have been extensively explored by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, inductively coupled plasma mass spectrometry (ICP-MS), flow cytometry, comet assay, inverted fluorescence microscope as well as western blotting experimental techniques. Notably, Ru1 and Ru2 exhibit potent antiproliferative activities against selected human cancer cell lines with IC50 values lower than those of cisplatin and other non-cyclometalated Ru(II) β-carboline complexes. The cellular uptake and localization exhibit that these complexes can accumulate in the cell nuclei. Further antitumor mechanism studies show that Ru1 and Ru2 can cause cell cycle arrest in the G0/G1 phase by regulating cell cycle relative proteins and induce apoptosis through mitochondrial dysfunction, reactive oxygen species (ROS) accumulation and ROS-mediated DNA damage.
Collapse
|
28
|
A novel ruthenium (II)-derived organometallic compound, TQ-6, potently inhibits platelet aggregation: Ex vivo and in vivo studies. Sci Rep 2017; 7:9556. [PMID: 28842683 PMCID: PMC5573325 DOI: 10.1038/s41598-017-09695-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/26/2017] [Indexed: 01/18/2023] Open
Abstract
Arterial thrombosis plays a key role in cardiovascular diseases. Hence, developing more effective antithrombotic agents is necessary. We designed a ruthenium (II)-derived complex, [Ru(η6-cymene)2-(1H-benzoimidazol-2-yl)-quinoline Cl]BF4 (TQ-6), as a new antiplatelet drug. TQ-6 (0.3 µM) exhibited extremely strong inhibitory activity against platelet aggregation, Src, and Syk phosphorylation stimulated by agonists in human platelets. In collagen-activated platelets, TQ-6 also inhibited ATP-release, [Ca+2]i, P-selectin expression, FITC-PAC-1 binding, and hydroxyl radical formation, as well as the phosphorylation of phospholipase Cγ2, protein kinase C, mitogen-activated protein kinases, and Akt. Neither FITC-JAQ1 nor FITC-triflavin binding or integrin β3 phosphorylation stimulated by immobilized fibrinogen were diminished by TQ-6. Furthermore, TQ-6 had no effects in cyclic nucleotide formation. Moreover, TQ-6 substantially prolonged the closure time in whole blood, increased the occlusion time of thrombotic platelet plug formation and bleeding time in mice. In conclusion, TQ-6 has a novel role in inhibiting platelet activation through the inhibition of the agonist receptors-mediated inside-out signaling such as Src-Syk-PLCγ2 cascade and subsequent suppression of granule secretion, leading to disturb integrin αIIbβ3-mediated outside-in signaling, and ultimately inhibiting platelet aggregation. Therefore, TQ-6 has potential to develop as a therapeutic agent for preventing or treating thromboembolic disorders.
Collapse
|
29
|
Lai H, Zhang X, Feng P, Xie L, Chen J, Chen T. Enhancement of Antiangiogenic Efficacy of Iron(II) Complex by Selenium Substitution. Chem Asian J 2017; 12:982-987. [DOI: 10.1002/asia.201700272] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Haoqiang Lai
- Department of Chemistry; Jinan University; Guangzhou 510632 China
| | - Xiang Zhang
- Department of Chemistry; Jinan University; Guangzhou 510632 China
| | - Pengju Feng
- Department of Chemistry; Jinan University; Guangzhou 510632 China
| | - Lina Xie
- Department of Chemistry; Jinan University; Guangzhou 510632 China
| | - Jinjin Chen
- Department of Chemistry; Jinan University; Guangzhou 510632 China
| | - Tianfeng Chen
- Department of Chemistry; Jinan University; Guangzhou 510632 China
| |
Collapse
|
30
|
Deng Z, Gao P, Yu L, Ma B, You Y, Chan L, Mei C, Chen T. Ruthenium complexes with phenylterpyridine derivatives target cell membrane and trigger death receptors-mediated apoptosis in cancer cells. Biomaterials 2017; 129:111-126. [PMID: 28340357 DOI: 10.1016/j.biomaterials.2017.03.017] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 03/10/2017] [Accepted: 03/11/2017] [Indexed: 01/04/2023]
Abstract
Elucidation of the communication between metal complexes and cell membrane may provide useful information for rational design of metal-based anticancer drugs. Herein we synthesized a novel class of ruthenium (Ru) complexes containing phtpy derivatives (phtpy = phenylterpyridine), analyzed their structure-activity relationship and revealed their action mechanisms. The result showed that, the increase in the planarity of hydrophobic Ru complexes significantly enhanced their lipophilicity and cellular uptake. Meanwhile, the introduction of nitro group effectively improved their anticancer efficacy. Further mechanism studies revealed that, complex (2c), firstly accumulated on cell membrane and interacted with death receptors to activate extrinsic apoptosis signaling pathway. The complex was then transported into cell cytoplasm through transferrin receptor-mediated endocytosis. Most of the intracellular 2c accumulated in cell plasma, decreasing the level of cellular ROS, inducing the activation of caspase-9 and thus intensifying the apoptosis. At the same time, the residual 2c can translocate into cell nucleus to interact with DNA, induce DNA damage, activate p53 pathway and enhance apoptosis. Comparing with cisplatin, 2c possesses prolonged circulation time in blood, comparable antitumor ability and importantly, much lower toxicity in vivo. Taken together, this study uncovers the role of membrane receptors in the anticancer actions of Ru complexes, and provides fundamental information for rational design of membrane receptor targeting anticancer drugs.
Collapse
Affiliation(s)
- Zhiqin Deng
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Pan Gao
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Lianling Yu
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Bin Ma
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Yuanyuan You
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Leung Chan
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Chaoming Mei
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Tianfeng Chen
- Department of Chemistry, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
31
|
Xie L, Luo Z, Zhao Z, Chen T. Anticancer and Antiangiogenic Iron(II) Complexes That Target Thioredoxin Reductase to Trigger Cancer Cell Apoptosis. J Med Chem 2016; 60:202-214. [DOI: 10.1021/acs.jmedchem.6b00917] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Lina Xie
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Zuandi Luo
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Zhennan Zhao
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Tianfeng Chen
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| |
Collapse
|
32
|
Liu D, Sun W, Ren R, Wang Y, Shen Z. Poly[6-(2,6-bis(1′-methylbenzimidazolyl)pyridin-4-yloxy)hexyl acrylate] (PBIP) and its terbium (III) complex (PBIP-Tb 3+): Homopolymerization, optical, and magnetic performance. J Appl Polym Sci 2016. [DOI: 10.1002/app.44249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Dizheng Liu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization; Department of Polymer Science and Engineering, Zhejiang University; Hangzhou 310027 People's Republic of China
| | - Weilin Sun
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization; Department of Polymer Science and Engineering, Zhejiang University; Hangzhou 310027 People's Republic of China
| | - Rong Ren
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization; Department of Polymer Science and Engineering, Zhejiang University; Hangzhou 310027 People's Republic of China
| | - Yanhua Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization; Department of Polymer Science and Engineering, Zhejiang University; Hangzhou 310027 People's Republic of China
| | - Zhiquan Shen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization; Department of Polymer Science and Engineering, Zhejiang University; Hangzhou 310027 People's Republic of China
| |
Collapse
|
33
|
Kwong WL, Lam KY, Lok CN, Lai YT, Lee PY, Che CM. A Macrocyclic Ruthenium(III) Complex Inhibits Angiogenesis with Down-Regulation of Vascular Endothelial Growth Factor Receptor-2 and Suppresses Tumor Growth In Vivo. Angew Chem Int Ed Engl 2016; 55:13524-13528. [DOI: 10.1002/anie.201608094] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 09/07/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Wai-Lun Kwong
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Kar-Yee Lam
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Chun-Nam Lok
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Yau-Tsz Lai
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Pui-Yan Lee
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Chi-Ming Che
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
- HKU Shenzhen Institute of Research and Innovation; Shenzen 518053 P.R. China
| |
Collapse
|
34
|
Kwong WL, Lam KY, Lok CN, Lai YT, Lee PY, Che CM. A Macrocyclic Ruthenium(III) Complex Inhibits Angiogenesis with Down-Regulation of Vascular Endothelial Growth Factor Receptor-2 and Suppresses Tumor Growth In Vivo. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201608094] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Wai-Lun Kwong
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Kar-Yee Lam
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Chun-Nam Lok
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Yau-Tsz Lai
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Pui-Yan Lee
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
| | - Chi-Ming Che
- State Key Laboratory of Synthetic Chemistry; Chemical Biology Centre and Department of Chemistry; The University of Hong Kong; Pokfulam Road Hong Kong P.R. China
- HKU Shenzhen Institute of Research and Innovation; Shenzen 518053 P.R. China
| |
Collapse
|
35
|
Huang W, Wu H, Li X, Chen T. Facile One-Pot Synthesis of Tellurium Nanorods as Antioxidant and Anticancer Agents. Chem Asian J 2016; 11:2301-11. [PMID: 27325381 DOI: 10.1002/asia.201600757] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Indexed: 11/09/2022]
Abstract
Nanorods have been utilized in targeted therapy, controlled release, molecular diagnosis, and molecule imaging owing to their large surface area and optical, magnetic, electronic, and structural properties. However, low stability and complex synthetic methods have substantially limited the application of tellurium nanorods for use as antioxidant and anticancer agents. Herein, a facile one-pot synthesis of functionalized tellurium nanorods (PTNRs) by using a hydrothermal synthetic system with a polysaccharide-protein complex (PTR), which was extracted from Pleurotus tuber-regium, as a capping agent is described. PTNRs remained stable in water and in phosphate-buffered saline and exhibited high hemocompatibility. Interestingly, these nanorods possessed strong antioxidant activity for scavenging 2,2'-azinobis-(3-ethylbenzothiazoline-6-sulfonic acid radical cation (ABTS(.+) ) and 2,2-diphenyl-1-picrylhydrazylhydrate (DPPH) free radicals and demonstrated novel anticancer activities. However, these nanorods exhibited low cytotoxicity toward normal human cells. In addition, the PTNRs effectively induced a decrease in the mitochondrial membrane potential in a dose-dependent manner, which indicated that mitochondrial dysfunction might play an important role in PTNR-induced apoptosis. Therefore, this study provides a one-pot strategy for the facile synthesis of tellurium nanorods with novel antioxidant and anticancer application potentials.
Collapse
Affiliation(s)
- Wei Huang
- Department of Chemistry, Jinan University, Guangzhou, 510631, P.R China
| | - Hualian Wu
- Department of Chemistry, Jinan University, Guangzhou, 510631, P.R China
| | - Xiaoling Li
- Department of Chemistry, Jinan University, Guangzhou, 510631, P.R China.
| | - Tianfeng Chen
- Department of Chemistry, Jinan University, Guangzhou, 510631, P.R China.
| |
Collapse
|
36
|
Gou Y, Wang J, Chen S, Zhang Z, Zhang Y, Zhang W, Yang F. α-N-heterocyclic thiosemicarbazone Fe(III) complex: Characterization of its antitumor activity and identification of anticancer mechanism. Eur J Med Chem 2016; 123:354-364. [PMID: 27487566 DOI: 10.1016/j.ejmech.2016.07.041] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 07/12/2016] [Accepted: 07/19/2016] [Indexed: 12/19/2022]
Abstract
We synthesized an α-N-heterocyclic thiosemicarbazone ligand (L) and its Fe complex (C1) and assessed their chemical and biological properties in order to understand their marked activity. Electrochemical studies and ascorbate oxidation studies demonstrated that C1 shows considerable redox activity, and Fe(III/II) redox potentials was within the range accessible to cellular oxidants and reductants. Absorption spectral, emission spectral and viscosity analysis reveal that L and C1 interacted with DNA through intercalation and C1 exhibited a higher DNA binding ability. Agarose gel electrophoresis experiments indicated that C1 exhibited the highest pBR322 DNA cleaving ability. In vitro, C1 showed significantly more anticancer activity than the ligand alone. Moreover, C1 induces production of reactive oxygen species (ROS) and DNA damage, resulting in activation of the p53 pathway, cell cycle arrest at the S phase, and mitochondria-mediated apoptosis by regulating the expression of Bcl-2 family proteins.
Collapse
Affiliation(s)
- Yi Gou
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Jun Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Shifang Chen
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Zhan Zhang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yao Zhang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Wei Zhang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Feng Yang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
37
|
Liu DZ, Sun WL, Ren R, Wang YH, Shen ZQ. Synthesis of poly[6-(2,6-bis(1′-methylbenzimidazolyl)pyridin-4-yloxy)hexyl acrylate] (PBIP) and magnetic property of its neodymium complex (PBIP-Nd3+). CHINESE JOURNAL OF POLYMER SCIENCE 2016. [DOI: 10.1007/s10118-016-1804-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
38
|
Kljun J, Anko M, Traven K, Sinreih M, Pavlič R, Peršič Š, Ude Ž, Codina EE, Stojan J, Lanišnik Rižner T, Turel I. Pyrithione-based ruthenium complexes as inhibitors of aldo-keto reductase 1C enzymes and anticancer agents. Dalton Trans 2016; 45:11791-800. [PMID: 27357845 DOI: 10.1039/c6dt00668j] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Four ruthenium complexes of clinically used zinc ionophore pyrithione and its oxygen analog 2-hydroxypyridine N-oxide were prepared and evaluated as inhibitors of enzymes of the aldo-keto reductase subfamily 1C (AKR1C). A kinetic study assisted with docking simulations showed a mixed type of inhibition consisting of a fast reversible and a slow irreversible step in the case of both organometallic compounds 1A and 1B. Both compounds also showed a remarkable selectivity towards AKR1C1 and AKR1C3 which are targets for breast cancer drug design. The organoruthenium complex of ligand pyrithione as well as pyrithione itself also displayed toxicity on the hormone-dependent MCF-7 breast cancer cell line with EC50 values in the low micromolar range.
Collapse
Affiliation(s)
- Jakob Kljun
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Chow MJ, Babak MV, Wong DYQ, Pastorin G, Gaiddon C, Ang WH. Structural Determinants of p53-Independence in Anticancer Ruthenium-Arene Schiff-Base Complexes. Mol Pharm 2016; 13:2543-54. [PMID: 27174050 DOI: 10.1021/acs.molpharmaceut.6b00348] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
p53 is a key tumor suppressor gene involved in key cellular processes and implicated in cancer therapy. However, it is inactivated in more than 50% of all cancers due to mutation or overexpression of its negative regulators. This leads to drug resistance and poor chemotherapeutic outcome as most clinical drugs act via a p53-dependent mechanism of action. An attractive strategy to circumvent this resistance would be to identify new anticancer drugs that act via p53-independent mode of action. In the present study, we identified 9 Ru (II)-Arene Schiff-base (RAS) complexes able to induce p53-independent cytotoxicity and discuss structural features that are required for their p53-independent activity. Increasing hydrophobicity led to an increase in cellular accumulation in cells with a corresponding increase in efficacy. We further showed that all nine complexes demonstrated p53-independent activity. This was despite significant differences in their physicochemical properties, suggesting that the iminoquinoline ligand, a common structural feature for all the complexes, is required for the p53-independent activity.
Collapse
Affiliation(s)
- Mun Juinn Chow
- Department of Chemistry, National University of Singapore , 3 Science Drive 3, 117543 Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , 28 Medical Drive, 117456 Singapore
| | - Maria V Babak
- Department of Chemistry, National University of Singapore , 3 Science Drive 3, 117543 Singapore
| | - Daniel Yuan Qiang Wong
- Department of Chemistry, National University of Singapore , 3 Science Drive 3, 117543 Singapore
| | - Giorgia Pastorin
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , 28 Medical Drive, 117456 Singapore.,Department of Pharmacy, National University of Singapore , 18 Science Drive 4, 117543 Singapore
| | - Christian Gaiddon
- U1113 INSERM, 3 Avenue Molière, Strasbourg 67200, France.,Oncology Section, FMTS, Université de Strasbourg , F-67081 Strasbourg, France
| | - Wee Han Ang
- Department of Chemistry, National University of Singapore , 3 Science Drive 3, 117543 Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , 28 Medical Drive, 117456 Singapore
| |
Collapse
|
40
|
Musumeci D, Rozza L, Merlino A, Paduano L, Marzo T, Massai L, Messori L, Montesarchio D. Interaction of anticancer Ru(III) complexes with single stranded and duplex DNA model systems. Dalton Trans 2016; 44:13914-25. [PMID: 26154188 DOI: 10.1039/c5dt01105a] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The interaction of the anticancer Ru(iii) complex AziRu - in comparison with its analogue NAMI-A, currently in advanced clinical trials as an antimetastatic agent - with DNA model systems, both single stranded and duplex oligonucleotides, was investigated using a combined approach, including absorption UV-vis spectroscopy, circular dichroism (CD) and electrospray mass spectrometry (ESI-MS) techniques. UV-vis absorption spectra of the Ru complexes were recorded at different times in a pseudo-physiological solution, to monitor the ligand exchange processes in the absence and in the presence of the examined oligonucleotides. CD experiments provided information on the overall conformational changes of the DNA model systems induced by these metal complexes. UV- and CD-monitored thermal denaturation studies were performed to analyse the effects of AziRu and NAMI-A on the stability of the duplex structures. ESI-MS experiments, carried out on the oligonucleotide/metal complex mixtures under investigation, allowed us to detect the formation of stable adducts between the guanine-containing oligomers and the ruthenium complexes. These data unambiguously demonstrate that both AziRu and NAMI-A can interact with the DNA model systems. Although very similar in their structures, the two metal compounds manifest a markedly different reactivity with the examined sequences, respectively, with either a naked Ru(3+) ion or a Ru(Im)(3+) (Im = imidazole) fragment being incorporated into the oligonucleotide structure via stable linkages.
Collapse
Affiliation(s)
- Domenica Musumeci
- Department of Chemical Sciences, University of Napoli Federico II, Via Cintia 21, I-80126 Napoli, Italy
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Fu X, Yang Y, Li X, Lai H, Huang Y, He L, Zheng W, Chen T. RGD peptide-conjugated selenium nanoparticles: antiangiogenesis by suppressing VEGF-VEGFR2-ERK/AKT pathway. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1627-39. [PMID: 26961468 DOI: 10.1016/j.nano.2016.01.012] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 10/25/2015] [Accepted: 01/21/2016] [Indexed: 12/31/2022]
Abstract
Angiogenesis is essential for tumorigenesis, progression and metastasis. Herein we described the synthesis of RGD peptide-decorated and doxorubicin-loaded selenium nanoparticles (RGD-NPs) targeting tumor vasculature to enhance the cellular uptake and antiangiogenic activities in vitro and in vivo. After internalization by receptor-mediated endocytosis, this nanosystem disassembled under acidic condition with the presence of lysozymes and cell lysate, leading to bioresponsive triggered drug release. Mechanistic investigation revealed that RGD-NPs inhibited angiogenesis through induction of apoptosis and cell cycle arrest in human umbilical vein endothelial cells (HUVECs) via suppression of VEGF-VEGFR2-ERK/AKT signaling axis by triggering ROS-mediated DNA damage. Additionally, RGD-NPs can inhibit MCF-7 tumor growth and angiogenesis in nude mice via down-regulation of VEGF-VEGFR2, effectively reduce the toxicity and prolong the blood circulation in vivo. Our results suggest that the strategy to use RGD-peptide functionalized SeNPs as carriers of anticancer drugs is an efficient way to achieve cancer-targeted antiangiogenesis synergism.
Collapse
Affiliation(s)
- Xiaoyan Fu
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Yahui Yang
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Xiaoling Li
- Department of Chemistry, Jinan University, Guangzhou, China.
| | - Haoqiang Lai
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Yanyu Huang
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Lizhen He
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Wenjie Zheng
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Tianfeng Chen
- Department of Chemistry, Jinan University, Guangzhou, China.
| |
Collapse
|
42
|
Zhou B, Huang Y, Yang F, Zheng W, Chen T. Dual-Functional Nanographene Oxide as Cancer-Targeted Drug-Delivery System to Selectively Induce Cancer-Cell Apoptosis. Chem Asian J 2016; 11:1008-19. [DOI: 10.1002/asia.201501277] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 12/28/2015] [Indexed: 01/19/2023]
Affiliation(s)
- Binwei Zhou
- Department of Chemistry; Jinan University; Guangzhou 510631 P.R. China
| | - Yanyu Huang
- Department of Chemistry; Jinan University; Guangzhou 510631 P.R. China
| | - Fang Yang
- Department of Chemistry; Jinan University; Guangzhou 510631 P.R. China
| | - Wenjie Zheng
- Department of Chemistry; Jinan University; Guangzhou 510631 P.R. China
| | - Tianfeng Chen
- Department of Chemistry; Jinan University; Guangzhou 510631 P.R. China
| |
Collapse
|
43
|
Garcia CV, Parrilha GL, Rodrigues BL, Teixeira SF, de Azevedo RA, Ferreira AK, Beraldo H. Tricarbonylrhenium(i) complexes with 2-acetylpyridine-derived hydrazones are cytotoxic to NCI-H460 human large cell lung cancer. NEW J CHEM 2016. [DOI: 10.1039/c6nj00050a] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Tricarbonylrhenium(i)-hydrazone complexes showed antiproliferative activity against NCI-H460 cells. The mode of action involves apoptosis and ROS scavenge.
Collapse
Affiliation(s)
- Camila Vargas Garcia
- Departamento de Química
- Instituto de Ciências Exatas
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | - Gabrieli Lessa Parrilha
- Departamento de Química
- Instituto de Ciências Exatas
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | - Bernardo Lages Rodrigues
- Departamento de Química
- Instituto de Ciências Exatas
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | - Sarah Fernandes Teixeira
- Departamento de Imunologia
- Instituto de Ciências Biomédicas
- Universidade de São Paulo
- São Paulo
- Brazil
| | | | - Adilson Kleber Ferreira
- Departamento de Imunologia
- Instituto de Ciências Biomédicas
- Universidade de São Paulo
- São Paulo
- Brazil
| | - Heloisa Beraldo
- Departamento de Química
- Instituto de Ciências Exatas
- Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| |
Collapse
|
44
|
Liu M, Chang Y, Yang J, You Y, He R, Chen T, Zhou C. Functionalized halloysite nanotube by chitosan grafting for drug delivery of curcumin to achieve enhanced anticancer efficacy. J Mater Chem B 2016; 4:2253-2263. [DOI: 10.1039/c5tb02725j] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A new HNTs-based drug delivery system to improve the bioavailability of curcumin for cancer therapy is proposed.
Collapse
Affiliation(s)
- Mingxian Liu
- Department of Materials Science and Engineering
- Jinan University
- Guangzhou 510632
- China
| | - Yanzhou Chang
- Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
| | - Jing Yang
- Department of Materials Science and Engineering
- Jinan University
- Guangzhou 510632
- China
| | - Yuanyuan You
- Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
| | - Rui He
- Department of Materials Science and Engineering
- Jinan University
- Guangzhou 510632
- China
| | - Tianfeng Chen
- Department of Chemistry
- Jinan University
- Guangzhou 510632
- China
| | - Changren Zhou
- Department of Materials Science and Engineering
- Jinan University
- Guangzhou 510632
- China
| |
Collapse
|
45
|
Yang Y, Deng S, Zeng Q, Hu W, Chen T. Highly stable selenadiazole derivatives induce bladder cancer cell apoptosis and inhibit cell migration and invasion through the activation of ROS-mediated signaling pathways. Dalton Trans 2016; 45:18465-18475. [PMID: 27711726 DOI: 10.1039/c6dt02045c] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Herein highly stable selenadiazole derivatives were synthesized and found to be able to induce bladder cancer cell apoptosis and inhibit cell migration and invasion through the activation of ROS-mediated pathways.
Collapse
Affiliation(s)
- Yahui Yang
- Department of Chemistry
- Jinan University Guangzhou 510632
- China
| | - Shulin Deng
- Department of Chemistry
- Jinan University Guangzhou 510632
- China
| | - Qinsong Zeng
- Department of Urology
- General Hospital of Guangzhou Military Command of PLA
- Guangzhou 510010
- China
| | - Weilie Hu
- Department of Urology
- General Hospital of Guangzhou Military Command of PLA
- Guangzhou 510010
- China
| | - Tianfeng Chen
- Department of Chemistry
- Jinan University Guangzhou 510632
- China
| |
Collapse
|
46
|
Chan L, Huang Y, Chen T. Cancer-targeted tri-block copolymer nanoparticles as payloads of metal complexes to achieve enhanced cancer theranosis. J Mater Chem B 2016; 4:4517-4525. [DOI: 10.1039/c6tb00514d] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
47
|
Editorial of Special Issue Ruthenium Complex: The Expanding Chemistry of the Ruthenium Complexes. Molecules 2015; 20:17244-74. [PMID: 26393560 PMCID: PMC6332046 DOI: 10.3390/molecules200917244] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/09/2015] [Accepted: 09/11/2015] [Indexed: 12/18/2022] Open
Abstract
Recent trends in Ru complex chemistry are surveyed with emphasis on the development of anticancer drugs and applications in catalysis, polymers, materials science and nanotechnology.
Collapse
|
48
|
Yellol J, Pérez SA, Buceta A, Yellol G, Donaire A, Szumlas P, Bednarski PJ, Makhloufi G, Janiak C, Espinosa A, Ruiz J. Novel C,N-Cyclometalated Benzimidazole Ruthenium(II) and Iridium(III) Complexes as Antitumor and Antiangiogenic Agents: A Structure-Activity Relationship Study. J Med Chem 2015; 58:7310-27. [PMID: 26313136 DOI: 10.1021/acs.jmedchem.5b01194] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A series of novel C,N-cyclometalated benzimidazole ruthenium(II) and iridium(III) complexes of the types [(η(6)-p-cymene)RuCl(κ(2)-N,C-L)] and [(η(5)-C5Me5)IrCl(κ(2)-N,C-L)] (HL = methyl 1-butyl-2-arylbenzimidazolecarboxylate) with varying substituents (H, Me, F, CF3, MeO, NO2, and Ph) in the R4 position of the phenyl ring of 2-phenylbenzimidazole chelating ligand of the ruthenium (3a-g) and iridium complexes (4a-g) have been prepared. The cytotoxic activity of the new ruthenium(II) and iridium(III) compounds has been evaluated in a panel of cell lines (A2780, A2780cisR, A427, 5637, LCLC, SISO, and HT29) in order to investigate structure-activity relationships. Phenyl substitution at the R4 position shows increased potency in both Ru and Ir complexes (3g and 4g, respectively) as compared to their parent compounds (3a and 4a) in all cell lines. In general, ruthenium complexes are more active than the corresponding iridium complexes. The new ruthenium and iridium compounds increased caspase-3 activity in A2780 cells, as shown for 3a,d and 4a,d. Compound 4g is able to increase the production of ROS in A2780 cells. Furthermore, all the new compounds are able to overcome the cisplatin resistance in A2780cisR cells. In addition, some of the metal complexes effectively inhibit angiogenesis in the human umbilical vein endothelial cell line EA.hy926 at 0.5 μM, the ruthenium derivatives 3g (Ph) and 3d (CF3) being the best performers. QC calculations performed on some ruthenium model complexes showed only moderate or slight electron depletion at the phenyl ring of the C,N-cyclometalated ligand and the chlorine atom on increasing the electron withdrawing effect of the R substituent.
Collapse
Affiliation(s)
- Jyoti Yellol
- Departamento de Química Inorgánica and Regional Campus of International Excellence "Campus Mare Nostrum", Universidad de Murcia and Institute for Bio-Health Research of Murcia IMIB-Arrixaca , E-30071 Murcia, Spain
| | - Sergio A Pérez
- Departamento de Química Inorgánica and Regional Campus of International Excellence "Campus Mare Nostrum", Universidad de Murcia and Institute for Bio-Health Research of Murcia IMIB-Arrixaca , E-30071 Murcia, Spain
| | - Alicia Buceta
- Departamento de Química Inorgánica and Regional Campus of International Excellence "Campus Mare Nostrum", Universidad de Murcia and Institute for Bio-Health Research of Murcia IMIB-Arrixaca , E-30071 Murcia, Spain
| | - Gorakh Yellol
- Departamento de Química Inorgánica and Regional Campus of International Excellence "Campus Mare Nostrum", Universidad de Murcia and Institute for Bio-Health Research of Murcia IMIB-Arrixaca , E-30071 Murcia, Spain
| | - Antonio Donaire
- Departamento de Química Inorgánica and Regional Campus of International Excellence "Campus Mare Nostrum", Universidad de Murcia and Institute for Bio-Health Research of Murcia IMIB-Arrixaca , E-30071 Murcia, Spain
| | - Piotr Szumlas
- Pharmaceutical and Medicinal Chemistry, Institut für Pharmazie, EMA-University of Greifswald , D-17487 Greifswald, Germany
| | - Patrick J Bednarski
- Pharmaceutical and Medicinal Chemistry, Institut für Pharmazie, EMA-University of Greifswald , D-17487 Greifswald, Germany
| | - Gamall Makhloufi
- Institut für Anorganische Chemie und Strukturchemie, Heinrich-Heine-Universität Düsseldorf , Universitätsstrasse 1, D-40225 Düsseldorf, Germany
| | - Christoph Janiak
- Institut für Anorganische Chemie und Strukturchemie, Heinrich-Heine-Universität Düsseldorf , Universitätsstrasse 1, D-40225 Düsseldorf, Germany
| | - Arturo Espinosa
- Departamento de Química Orgánica, Universidad de Murcia , E-30071 Murcia, Spain
| | - José Ruiz
- Departamento de Química Inorgánica and Regional Campus of International Excellence "Campus Mare Nostrum", Universidad de Murcia and Institute for Bio-Health Research of Murcia IMIB-Arrixaca , E-30071 Murcia, Spain
| |
Collapse
|
49
|
Păunescu E, Nowak-Sliwinska P, Clavel CM, Scopelliti R, Griffioen AW, Dyson PJ. Anticancer Organometallic Osmium(II)-p-cymene Complexes. ChemMedChem 2015; 10:1539-1547. [PMID: 26190176 DOI: 10.1002/cmdc.201500221] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Indexed: 01/04/2025]
Abstract
Osmium compounds are attracting increasing attention as potential anticancer drugs. In this context, a series of bifunctional organometallic osmium(II)-p-cymene complexes functionalized with alkyl or perfluoroalkyl groups were prepared and screened for their antiproliferative activity. Three compounds from the series display selectivity toward cancer cells, with moderate cytotoxicity observed against human ovarian carcinoma (A2780) cells, whereas no cytotoxicity was observed on non-cancerous human embryonic kidney (HEK-293) cells and human endothelial (ECRF24) cells. Two of these three cancer-cell-selective compounds induce cell death largely via apoptosis and were also found to disrupt vascularization in the chicken embryo chorioallantoic membrane (CAM) model. Based on these promising properties, these compounds have potential clinical applications.
Collapse
Affiliation(s)
- Emilia Păunescu
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne (Switzerland)
| | - Patrycja Nowak-Sliwinska
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne (Switzerland)
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, 1081 HV Amsterdam (The Netherlands)
| | - Catherine M Clavel
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne (Switzerland)
| | - Rosario Scopelliti
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne (Switzerland)
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, 1081 HV Amsterdam (The Netherlands)
| | - Paul J Dyson
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne (Switzerland).
| |
Collapse
|
50
|
Bergamo A, Sava G. Linking the future of anticancer metal-complexes to the therapy of tumour metastases. Chem Soc Rev 2015; 44:8818-35. [DOI: 10.1039/c5cs00134j] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cancer chemotherapy is almost always applied to patients with one or more diagnosed metastases and is expected to impact these lesions, thus providing significant benefits to the patient.
Collapse
Affiliation(s)
| | - Gianni Sava
- Callerio Foundation Onlus
- 34127 Trieste
- Italy
- Department of Life Sciences
- University of Trieste
| |
Collapse
|