1
|
Antonets AA, Spitsyna EV, Tyurin VY, Mazur DM, Yakovlev DS, Babkov DA, Pshenichnikova MS, Spasov AA, Milaeva ER, Nazarov AA. Ruthenium complexes with abiraterone acetate as antiproliferative agents. J Inorg Biochem 2025; 262:112754. [PMID: 39383670 DOI: 10.1016/j.jinorgbio.2024.112754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
This study is dedicated to the development of multimodal anticancer agents. We have obtained ruthenium complexes conjugated with the steroid-type antitumor drug abiraterone acetate in order to take advantage of the dual antitumor properties of both ruthenium and abiraterone. The compounds exhibit good antiproliferative activity against cancer cells, with selectivity over primary fibroblasts. Real-time cell analysis revealed that compound dichlorido(η6-p-cymene)(abiraterone acetate)ruthenium(II) had pronounced antiproliferation activity compared to abiraterone acetate. Flow cytometric studies on the mechanism of cell death have revealed that the most active compound induces apoptosis more effectively than abiraterone acetate. Our findings demonstrate the potential of this novel dual-action compound as promising candidates for further development as anticancer agents.
Collapse
Affiliation(s)
- Anastasia A Antonets
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Ekaterina V Spitsyna
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Vladimir Yu Tyurin
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia; Department of Materials Science, MSU-BIT University, Shenzhen 517182, China
| | - Dmitrii M Mazur
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Dmitry S Yakovlev
- Volgograd State Medical University, Pavshikh Bortsov Sq. 1, 400131 Volgograd, Russia
| | - Denis A Babkov
- Volgograd State Medical University, Pavshikh Bortsov Sq. 1, 400131 Volgograd, Russia
| | | | - Alexander A Spasov
- Volgograd State Medical University, Pavshikh Bortsov Sq. 1, 400131 Volgograd, Russia
| | - Elena R Milaeva
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Alexey A Nazarov
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia.
| |
Collapse
|
2
|
Li A, Pan W, Zhang Z, Yang F, Gou Y, Zhang Y, Ma L. Hydrazone copper(II) complexes suppressed lung adenocarcinoma by activating multiple anticancer pathway. Bioorg Chem 2024; 154:107994. [PMID: 39603071 DOI: 10.1016/j.bioorg.2024.107994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/06/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
Activating multiple anti-cancer pathways has great potential for tumor treatment. Herein, we synthesized two binuclear Cu(II) hydrazone complexes ([Cu2(HL1)2Cl2] 1 and [Cu2(HL1)2Br2] 2) and two mononuclear hydrazone-Cu(II) complexes ([Cu(HL2)Cl]·CH3OH 3 and [Cu(HL2)(H2O)Br]·2H2O 4), to evaluate their anti-lung cancer activities. MTT assays revealed that the Cu(II) complexes demonstrate superior anticancer activity compared to cisplatin. Among them, complex 3 exhibited selective toxicity towards A549 cancer cells in comparison to normal cells and demonstrated hemolytic activity comparable to cisplatin. The low toxicity and effective antitumor capabilities of complex 3 have been confirmed in xenograft experiments using A549 tumor-bearing mice. Interestingly, complex 3 eradicates lung tumor cells both in vivo and in vitro by initiating multiple anticancer pathways, including cuproptosis. Our research extends the study of hydrazone copper complexes and provides strategies for the treatment of lung cancer.
Collapse
Affiliation(s)
- Aili Li
- The Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China; Key Laboratory of Basic Research on Respiratory Diseases, Guangxi Health Commission, Guilin, Guangxi, China; Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, Guangxi, China
| | - Weiping Pan
- The Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - ZhenLei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, China
| | - Yi Gou
- The Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China; Key Laboratory of Basic Research on Respiratory Diseases, Guangxi Health Commission, Guilin, Guangxi, China.
| | - Ye Zhang
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, Guangxi, China.
| | - Libing Ma
- The Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China; Key Laboratory of Basic Research on Respiratory Diseases, Guangxi Health Commission, Guilin, Guangxi, China.
| |
Collapse
|
3
|
Falcone E, Vigna V, Schueffl H, Stellato F, Vileno B, Bouraguba M, Mazzone G, Proux O, Morante S, Heffeter P, Sicilia E, Faller P. When Metal Complexes Evolve, and a Minor Species is the Most Active: the Case of Bis(Phenanthroline)Copper in the Catalysis of Glutathione Oxidation and Hydroxyl Radical Generation. Angew Chem Int Ed Engl 2024:e202414652. [PMID: 39363702 DOI: 10.1002/anie.202414652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024]
Abstract
Several copper-ligands, including 1,10-phenanthroline (Phen), have been investigated for anticancer purposes based on their capacity to bind excess copper (Cu) in cancer tissues and form redox active complexes able to catalyse the formation of reactive oxygen species (ROS), ultimately leading to oxidative stress and cell death. Glutathione (GSH) is a critical compound as it is highly concentrated intracellularly and can reduce and dissociate copper(II) from the ligand forming poorly redox-active copper(I)-thiolate clusters. Here we report that Cu-Phen2 speciation evolves in physiologically relevant GSH concentrations. Experimental and computational experiments suggest that at pH 7.4 mostly copper(I)-GSH clusters are formed, but a minor species of copper(I) bound to one Phen and forming ternary complexes with GSH (GS-Cu-Phen) is the redox active species, oxidizing quite efficiently GSH to GSSG and forming HO⋅ radicals. This minor active species becomes more populated at lower pH, such as typical lysosomal pH 5, resulting in faster GSH oxidation and HO⋅ production. Consistently, cell culture studies showed lower toxicity of Cu-Phen2 upon inhibition of lysosomal acidification. Overall, this study underscores that sub-cellular localisation can considerably influence the speciation of Cu-based drugs and that minor species can be the most redox- and biologically-active.
Collapse
Affiliation(s)
- Enrico Falcone
- Institut de Chimie (UMR 7177), University of Strasbourg, CNRS, 4 Rue Blaise Pascal, 67081, Strasbourg, France
- current address: Laboratoire de Chimie de Coordination (UPR 8142), CNRS, 31077, Toulouse, France
| | - Vincenzo Vigna
- Department of Chemistry and Chemical Technologies, Università della Calabria, 87036, Arcavacata di Rende, CS, Italy
| | - Hemma Schueffl
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Francesco Stellato
- Department of Physics, Università di Roma Tor Vergata, Via della Ricerca Scientifica 1, 00133, Roma, Italy
- INFN, Università di Roma Tor Vergata, Via della Ricerca Scientifica 1, 00133, Roma, Italy
| | - Bertrand Vileno
- Institut de Chimie (UMR 7177), University of Strasbourg, CNRS, 4 Rue Blaise Pascal, 67081, Strasbourg, France
| | - Merwan Bouraguba
- Institut de Chimie (UMR 7177), University of Strasbourg, CNRS, 4 Rue Blaise Pascal, 67081, Strasbourg, France
| | - Gloria Mazzone
- Department of Chemistry and Chemical Technologies, Università della Calabria, 87036, Arcavacata di Rende, CS, Italy
| | - Olivier Proux
- Observatoire des Sciences de l'Univers de Grenoble, UAR 832, CNRS-Université Grenoble Alpes, 38041, Grenoble, France
| | - Silvia Morante
- Department of Physics, Università di Roma Tor Vergata, Via della Ricerca Scientifica 1, 00133, Roma, Italy
- INFN, Università di Roma Tor Vergata, Via della Ricerca Scientifica 1, 00133, Roma, Italy
| | - Petra Heffeter
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Emilia Sicilia
- Department of Chemistry and Chemical Technologies, Università della Calabria, 87036, Arcavacata di Rende, CS, Italy
| | - Peter Faller
- Institut de Chimie (UMR 7177), University of Strasbourg, CNRS, 4 Rue Blaise Pascal, 67081, Strasbourg, France
- Institut Universitaire de France (IUF), 1 rue Descartes, 75231, Paris, France
| |
Collapse
|
4
|
Mamindla A, Murugan D, Varadhan M, Ajaykamal T, Rangasamy L, Palaniandavar M, Rajendiran V. Mixed-ligand copper(ii)-diimine complexes of 3-formylchromone- N 4-phenyl thiosemicarbazone: 5,6-dmp co-ligand confers enhanced cytotoxicity. RSC Adv 2024; 14:31704-31722. [PMID: 39376525 PMCID: PMC11457010 DOI: 10.1039/d4ra04997g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/22/2024] [Indexed: 10/09/2024] Open
Abstract
The promising biological applications of thiosemicarbazone derivatives have inspired the design, synthesis, and study of their Cu(ii) complexes for anticancer therapeutic applications. Herein, we have evaluated the DNA/protein binding, DNA cleaving, and cytotoxic properties of four mixed-ligand Cu(ii) complexes of the type [Cu(L)(diimine)](NO3) 1-4, where HL is 4-oxo-4H-chromene-3-carbaldehyde-4(N)-phenylthiosemicarbazone and diimine is 2,2'-bipyridine (bpy, 1) 1,10-phenanthroline (phen, 2), 5,6-dimethyl-1,10-phenanthroline (5,6-dmp, 3), or dipyrido-[3,2-f:2',3'-h]-quinoxaline (dpq, 4). Interestingly, complex 3 with higher lipophilicity shows stronger DNA binding and oxidative DNA cleavage, higher ROS production, and more reversible redox behaviour, resulting in its remarkable cytotoxicity (IC50, 1.26 μM) against HeLa cervical cancer cells, and rendering it 5 times more potent than the widely used drug cisplatin. The same complex induces enhanced apoptotic cell death on HeLa cells but lower toxicity towards the non-cancerous PBMC cells. Molecular docking studies suggest that all the complexes bind in the minor groove of DNA and subdomain II of HSA, which is in close agreement with the experimental results. Also, 3 shows cytotoxicity higher than the analogous mixed ligand Cu(ii) complexes, reported already, emphasizing the importance of co-ligand in tuning the anticancer activity.
Collapse
Affiliation(s)
- Anjaneyulu Mamindla
- Department of Chemistry, School of Basic and Applied Sciences, Central University of Tamil Nadu Thiruvarur 610005 India
| | - Dhanashree Murugan
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT) Vellore 632014 Tamilnadu India
| | - Manikandan Varadhan
- Department of Chemistry, School of Basic and Applied Sciences, Central University of Tamil Nadu Thiruvarur 610005 India
| | | | - Loganathan Rangasamy
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT) Vellore 632014 Tamilnadu India
| | | | - Venugopal Rajendiran
- Department of Chemistry, School of Basic and Applied Sciences, Central University of Tamil Nadu Thiruvarur 610005 India
| |
Collapse
|
5
|
Hu H, Chen J, Zhang F, Sheng Z, Yang Y, Xie Y, Zhou L, Liu Y. Evaluation of Efficiency of Liposome-Entrapped Iridium(III) Complexes Inhibiting Tumor Growth In Vitro and In Vivo. J Med Chem 2024; 67:16195-16208. [PMID: 39264254 DOI: 10.1021/acs.jmedchem.4c01026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
In this paper, three new iridium(III) complexes: [Ir(piq)2(DFIPP)]PF6 (piq = deprotonated 1-phenylisoquinoline, DFIPP = 3,4-difluoro-2-(1H-imidazo[4,5-f][1,10]phenenthrolin-2-yl)phenol, 3a), [Ir(bzq)2(DFIPP)]PF6 (bzq = deprotonated benzo[h]quinoline, 3b), and [Ir(ppy)2(DFIPP)]PF6 (ppy = deprotonated 1-phenylpyridine, 3c), were synthesized and characterized. The complexes were found to be nontoxic to tumor cells via 3-(4,5-dimethylthiazole-2-yl)-diphenyltetrazolium bromide (MTT) assay. Surprisingly, its liposome-entrapped complexes 3alip, 3blip, and 3clip on B16 cells showed strong cytotoxicity (IC50 = 13.6 ± 2.8, 9.6 ± 1.1, and 18.9 ± 2.1 μM). Entry of 3alip, 3blip, and 3clip into B16 cells decreases mitochondrial membrane potential, regulates Bcl-2 family proteins, releases cytochrome c, triggers caspase family cascade reaction, and induces apoptosis. In addition, we also found that 3alip, 3blip, and 3clip triggered ferroptosis and autophagy. In vivo studies demonstrated that 3blip inhibited melanoma growth in C57 mice with a high inhibitory rate of 83.95%, and no organic damage was found in C57 mice.
Collapse
Affiliation(s)
- Huiyan Hu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Jing Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Fan Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Zhujun Sheng
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yan Yang
- Department of Pharmacy, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, PR China
| | - Yufeng Xie
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Lin Zhou
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| |
Collapse
|
6
|
Goodman DM, Ritter CU, Chen E, Tong KKH, Riisom M, Söhnel T, Jamieson SMF, Anderson RF, Brothers PJ, Ware DC, Hartinger CG. Masking the Bioactivity of Hydroxamic Acids by Coordination to Cobalt: Towards Bioreductive Anticancer Agents. Chemistry 2024; 30:e202401724. [PMID: 38853639 DOI: 10.1002/chem.202401724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/11/2024]
Abstract
The clinical use of many potent anticancer agents is limited by their non-selective toxicity to healthy tissue. One of these examples is vorinostat (SAHA), a pan histone deacetylase inhibitor, which shows high cytotoxicity with limited discrimination for cancerous over healthy cells. In an attempt to improve tumor selectivity, we exploited the properties of cobalt(III) as a redox-active metal center through stabilization with cyclen and cyclam tetraazamacrocycles, masking the anticancer activity of SAHA and other hydroxamic acid derivatives to allow for the complex to reach the hypoxic microenvironment of the tumor. Biological assays demonstrated the desired low in vitro anticancer activity of the complexes, suggesting effective masking of the activity of SAHA. Once in the tumor, the bioactive moiety may be released through the reduction of the CoIII center. Investigations revealed long-term stability of the complexes, with cyclic voltammetry and chemical reduction experiments supporting the design hypothesis of SAHA release through the reduction of the CoIII prodrug. The results highlight the potential for further developing this complex class as novel anticancer agents by masking the high cytotoxicity of a given drug, however, the cellular uptake needs to be improved.
Collapse
Affiliation(s)
- David M Goodman
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Cornelia U Ritter
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Erin Chen
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Kelvin K H Tong
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Mie Riisom
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
- MacDiarmid Institute for Advanced Materials and Nanotechnology, Victoria University of Wellington, PO Box 600, Wellington, 6140, New Zealand
| | - Tilo Söhnel
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
- MacDiarmid Institute for Advanced Materials and Nanotechnology, Victoria University of Wellington, PO Box 600, Wellington, 6140, New Zealand
| | - Stephen M F Jamieson
- Auckland Cancer Society Research Centre, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Robert F Anderson
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
- Auckland Cancer Society Research Centre, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
- Maurice Wilkins Centre, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Penelope J Brothers
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - David C Ware
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Christian G Hartinger
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| |
Collapse
|
7
|
Coverdale JPC, Bedford RA, Carter OWL, Cao S, Wills M, Sadler PJ. In-cell Catalysis by Tethered Organo-Osmium Complexes Generates Selectivity for Breast Cancer Cells. Chembiochem 2024; 25:e202400374. [PMID: 38785030 DOI: 10.1002/cbic.202400374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 05/25/2024]
Abstract
Anticancer agents that exhibit catalytic mechanisms of action offer a unique multi-targeting strategy to overcome drug resistance. Nonetheless, many in-cell catalysts in development are hindered by deactivation by endogenous nucleophiles. We have synthesised a highly potent, stable Os-based 16-electron half-sandwich ('piano stool') catalyst by introducing a permanent covalent tether between the arene and chelated diamine ligand. This catalyst exhibits antiproliferative activity comparable to the clinical drug cisplatin towards triple-negative breast cancer cells and can overcome tamoxifen resistance. Speciation experiments revealed Os to be almost exclusively albumin-bound in the extracellular medium, while cellular accumulation studies identified an energy-dependent, protein-mediated Os accumulation pathway, consistent with albumin-mediated uptake. Importantly, the tethered Os complex was active for in-cell transfer hydrogenation catalysis, initiated by co-administration of a non-toxic dose of sodium formate as a source of hydride, indicating that the Os catalyst is delivered to the cytosol of cancer cells intact. The mechanism of action involves the generation of reactive oxygen species (ROS), thus exploiting the inherent redox vulnerability of cancer cells, accompanied by selectivity for cancerous cells over non-tumorigenic cells.
Collapse
Affiliation(s)
- J P C Coverdale
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, B15 2TT, UK
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| | - R A Bedford
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, B15 2TT, UK
| | - O W L Carter
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| | - S Cao
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, B15 2TT, UK
| | - M Wills
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| | - P J Sadler
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| |
Collapse
|
8
|
Hata M, Kadoya Y, Ueno J, Taki M, Kodera M. Dicopper Complexes of p-Cresol-2,6-bis(amide-tether-dpa 4-X) (X = MeO and Cl): Selective ROS Generation and Cytotoxicity Enhancement Controlled by Electronic and Hydrophobic Effects of the MeO and Cl Groups. Inorg Chem 2024; 63:13893-13902. [PMID: 39011904 DOI: 10.1021/acs.inorgchem.4c01072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Two new p-cresol-2,6-bis(amide-tether-dpa4-X) ligands (HL4-X, X = MeO and Cl) and their dicopper complexes [Cu2(μ-1,1-OAc)(μ-1,3-OAc)(L4-MeO)]Y (Y = PF6 1a, OAc 1b) and [Cu2(μ-1,3-OAc)2(L4-Cl)]Y (Y = ClO4 2a, OAc 2b) were synthesized. The electronic and hydrophobic effects of the MeO and Cl groups were examined compared with nonsubstituted complex [Cu2(μ-1,1-OAc)(μ-1,3-OAc)(L)]+ (3). The electronic effects were found in crystal structures, spectroscopic characterization, and redox potentials of these complexes. 1b and 2b were reduced to Cu(I)Cu(I) with sodium ascorbate and reductively activated O2 to produce H2O2 and HO•. The H2O2 release and HO• generation are promoted by the electronic effects. The hydrophobic effects increased the lipophilicity of 1b and 2b. Cellular ROS generation of 1b, 2b, and 3 was visualized by DCFH-DA. To examine the intracellular behavior, boron dipyrromethene (Bodipy)-modified complexes 4B and 5B corresponding to 1b and 2b were synthesized. These support that 1b and 2b are localized at the ER and Golgi apparatus. The cytotoxicity of 1b and 2b against various cell lines was examined by MTT assay. 1b and 2b were 7- and 41-fold more cytotoxic than 3. 1b generated ROS selectively in cancer cell but 2b nonselectively in cancer and normal cells, causing cancer- and normal-cell-selective cytotoxicity, respectively.
Collapse
Affiliation(s)
- Machi Hata
- Department of Molecular Chemistry and Biochemistry, Doshisha University, Kyotanabe Kyoto 610-0321, Japan
| | - Yuki Kadoya
- Department of Molecular Chemistry and Biochemistry, Doshisha University, Kyotanabe Kyoto 610-0321, Japan
| | - Jin Ueno
- Department of Molecular Chemistry and Biochemistry, Doshisha University, Kyotanabe Kyoto 610-0321, Japan
| | - Masayasu Taki
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo, Chikusa, Nagoya 464-8601, Japan
| | - Masahito Kodera
- Department of Molecular Chemistry and Biochemistry, Doshisha University, Kyotanabe Kyoto 610-0321, Japan
| |
Collapse
|
9
|
Besleaga I, Raptová R, Stoica AC, Milunovic MNM, Zalibera M, Bai R, Igaz N, Reynisson J, Kiricsi M, Enyedy ÉA, Rapta P, Hamel E, Arion VB. Are the metal identity and stoichiometry of metal complexes important for colchicine site binding and inhibition of tubulin polymerization? Dalton Trans 2024; 53:12349-12369. [PMID: 38989784 PMCID: PMC11264232 DOI: 10.1039/d4dt01469c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 06/29/2024] [Indexed: 07/12/2024]
Abstract
Quite recently we discovered that copper(II) complexes with isomeric morpholine-thiosemicarbazone hybrid ligands show good cytotoxicity in cancer cells and that the molecular target responsible for this activity might be tubulin. In order to obtain better lead drug candidates, we opted to exploit the power of coordination chemistry to (i) assemble structures with globular shape to better fit the colchicine pocket and (ii) vary the metal ion. We report the synthesis and full characterization of bis-ligand cobalt(III) and iron(III) complexes with 6-morpholinomethyl-2-formylpyridine 4N-(4-hydroxy-3,5-dimethylphenyl)-3-thiosemicarbazone (HL1), 6-morpholinomethyl-2-acetylpyridine 4N-(4-hydroxy-3,5-dimethylphenyl)-3-thiosemicarbazone (HL2), and 6-morpholinomethyl-2-formylpyridine 4N-phenyl-3-thiosemicarbazone (HL3), and mono-ligand nickel(II), zinc(II) and palladium(II) complexes with HL1, namely [CoIII(HL1)(L1)](NO3)2 (1), [CoIII(HL2)(L2)](NO3)2 (2), [CoIII(HL3)(L3)](NO3)2 (3), [FeIII(L2)2]NO3 (4), [FeIII(HL3)(L3)](NO3)2 (5), [NiII(L1)]Cl (6), [Zn(L1)Cl] (7) and [PdII(HL1)Cl]Cl (8). We discuss the effect of the metal identity and metal complex stoichiometry on in vitro cytotoxicity and antitubulin activity. The high antiproliferative activity of complex 4 correlated well with inhibition of tubulin polymerization. Insights into the mechanism of antiproliferative activity were supported by experimental results and molecular docking calculations.
Collapse
Affiliation(s)
- Iuliana Besleaga
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Straße 42, A-1090 Vienna, Austria.
| | - Renáta Raptová
- Institute of Physical Chemistry and Chemical Physics, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, SK-81237 Bratislava, Slovakia
- Institute of Physical and Theoretical Chemistry, Graz University of Technology, Stremayrgasse 9/II, A-8010 Graz, Austria
| | - Alexandru-Constantin Stoica
- Inorganic Polymers Department, "Petru Poni" Institute of Macromolecular Chemistry, Aleea Gr. Ghica Voda 41 A, Iasi 700487, Romania
| | - Miljan N M Milunovic
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Straße 42, A-1090 Vienna, Austria.
| | - Michal Zalibera
- Institute of Physical Chemistry and Chemical Physics, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, SK-81237 Bratislava, Slovakia
| | - Ruoli Bai
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Diagnosis and Treatment, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Maryland 21702, USA
| | - Nóra Igaz
- Department of Biochemistry and Molecular Biology, University of Szeged, Közép fasor 52, H-6726 Szeged, Hungary
| | - Jóhannes Reynisson
- School of Pharmacy and Bioengineering, Keele University, Newcastle-under-Lyme, Staffordshire ST5 5BG, UK
| | - Mónika Kiricsi
- School of Pharmacy and Bioengineering, Keele University, Newcastle-under-Lyme, Staffordshire ST5 5BG, UK
| | - Éva A Enyedy
- Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8, H-6720 Szeged, Hungary.
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720 Szeged, Hungary
| | - Peter Rapta
- Institute of Physical Chemistry and Chemical Physics, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, SK-81237 Bratislava, Slovakia
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Diagnosis and Treatment, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Maryland 21702, USA
| | - Vladimir B Arion
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Straße 42, A-1090 Vienna, Austria.
- Inorganic Polymers Department, "Petru Poni" Institute of Macromolecular Chemistry, Aleea Gr. Ghica Voda 41 A, Iasi 700487, Romania
| |
Collapse
|
10
|
Hagar FF, Abbas SH, Atef E, Abdelhamid D, Abdel-Aziz M. Benzimidazole scaffold as a potent anticancer agent with different mechanisms of action (2016-2023). Mol Divers 2024:10.1007/s11030-024-10907-8. [PMID: 39031290 DOI: 10.1007/s11030-024-10907-8] [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: 03/24/2024] [Accepted: 05/30/2024] [Indexed: 07/22/2024]
Abstract
Benzimidazole scaffolds have potent anticancer activity due to their structure similarity to nucleoside. In addition, benzimidazoles could function as hydrogen donors or acceptors and bind to different drug targets that participate in cancer progression. The literature had many anticancer agents containing benzimidazole cores that gained much interest. Provoked by our endless interest in benzimidazoles as anticancer agents, we summarized the successful trials of the benzimidazole scaffolds in this concern. Moreover, we discuss the substantial opportunities in cancer treatment using benzimidazole-based drugs that may direct medicinal chemists for a compelling future design of more active chemotherapeutic agents with potential clinical applications. The uniqueness of this work lies in the highlighted benzimidazole scaffold hybridization with different molecules and benzimidazole-metal complexes, detailed mechanisms of action, and the IC50 of the developed compounds determined by different laboratories after 2015.
Collapse
Affiliation(s)
- Fatma Fouad Hagar
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Samar H Abbas
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt.
| | - Eman Atef
- College of Pharmacy, West Coast University, Los Angeles, CA, USA
| | - Dalia Abdelhamid
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt.
- Raabe College of Pharmacy, Ohio Northern University, Ohio, USA.
| | - Mohamed Abdel-Aziz
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| |
Collapse
|
11
|
Lončarević A, Clara-Trujillo S, Martínez-Férriz A, Blanco-Gómez M, Gallego-Ferrer G, Rogina A. Chitosan-copper microparticles as doxorubicin microcarriers for bone tumor therapy. Int J Pharm 2024; 659:124245. [PMID: 38772497 DOI: 10.1016/j.ijpharm.2024.124245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/30/2024] [Accepted: 05/17/2024] [Indexed: 05/23/2024]
Abstract
Doxorubicin (DOX) is a chemotherapeutic drug used in osteosarcoma treatments, usually administrated in very high dosages. This study proposes novel DOX microcarriers based on chitosan (CHT) physically crosslinked with copper(II) ions that will act synergically to inhibit tumor growth at lower drug dosage without affecting the healthy cells. Spherical CHT-Cu microparticles with a smooth surface and an average size of 30.1 ± 9.1 µm were obtained by emulsion. The release of Cu2+ ions from the CHT-Cu microparticles showed that 99.4 % of added cupric ions were released in 72 h of incubation in a complete cell culture medium (CCM). DOX entrapment in microparticles was conducted in a phosphate buffer solution (pH 6), utilizing the pH sensitivity of the polymer. The successful drug-loading process was confirmed by DOX emitting red fluorescence from drug-loaded microcarriers (DOX@CHT-Cu). The drug release in CCM showed an initial burst release, followed by sustained release. Biological assays indicated mild toxicity of CHT-Cu microparticles on the MG-63 osteosarcoma cell line, without affecting the viability of human mesenchymal stem cells (hMSCs). The DOX@CHT-Cu microparticles at concentration of 0.5 mg mL‒1 showed selective toxicity toward MG-63 cells.
Collapse
Affiliation(s)
- Andrea Lončarević
- University of Zagreb, Faculty of Chemical Engineering and Technology, Trg Marka Marulića 19, HR-10000 Zagreb, Croatia.
| | - Sandra Clara-Trujillo
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain.
| | - Arantxa Martínez-Férriz
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain.
| | - Mireia Blanco-Gómez
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain.
| | - Gloria Gallego-Ferrer
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain.
| | - Anamarija Rogina
- University of Zagreb, Faculty of Chemical Engineering and Technology, Trg Marka Marulića 19, HR-10000 Zagreb, Croatia.
| |
Collapse
|
12
|
Milunovic MM, Ohui K, Besleaga I, Petrasheuskaya TV, Dömötör O, Enyedy ÉA, Darvasiova D, Rapta P, Barbieriková Z, Vegh D, Tóth S, Tóth J, Kucsma N, Szakács G, Popović-Bijelić A, Zafar A, Reynisson J, Shutalev AD, Bai R, Hamel E, Arion VB. Copper(II) Complexes with Isomeric Morpholine-Substituted 2-Formylpyridine Thiosemicarbazone Hybrids as Potential Anticancer Drugs Inhibiting Both Ribonucleotide Reductase and Tubulin Polymerization: The Morpholine Position Matters. J Med Chem 2024; 67:9069-9090. [PMID: 38771959 PMCID: PMC11181322 DOI: 10.1021/acs.jmedchem.4c00259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/19/2024] [Accepted: 05/09/2024] [Indexed: 05/23/2024]
Abstract
The development of copper(II) thiosemicarbazone complexes as potential anticancer agents, possessing dual functionality as inhibitors of R2 ribonucleotide reductase (RNR) and tubulin polymerization by binding at the colchicine site, presents a promising avenue for enhancing therapeutic effectiveness. Herein, we describe the syntheses and physicochemical characterization of four isomeric proligands H2L3-H2L6, with the methylmorpholine substituent at pertinent positions of the pyridine ring, along with their corresponding Cu(II) complexes 3-6. Evidently, the position of the morpholine moiety and the copper(II) complex formation have marked effects on the in vitro antiproliferative activity in human uterine sarcoma MES-SA cells and the multidrug-resistant derivative MES-SA/Dx5 cells. Activity correlated strongly with quenching of the tyrosyl radical (Y•) of mouse R2 RNR protein, inhibition of RNR activity in the cancer cells, and inhibition of tubulin polymerization. Insights into the mechanism of antiproliferative activity, supported by experimental results and molecular modeling calculations, are presented.
Collapse
Affiliation(s)
| | - Katerina Ohui
- Institute
of Inorganic Chemistry, University of Vienna, Vienna A-1090, Austria
| | - Iuliana Besleaga
- Institute
of Inorganic Chemistry, University of Vienna, Vienna A-1090, Austria
| | - Tatsiana V. Petrasheuskaya
- Department
of Molecular and Analytical Chemistry, Interdisciplinary Excellence
Centre, University of Szeged, Dóm tér 7-8, Szeged H-6720, Hungary
- MTA-SZTE
Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary
| | - Orsolya Dömötör
- Department
of Molecular and Analytical Chemistry, Interdisciplinary Excellence
Centre, University of Szeged, Dóm tér 7-8, Szeged H-6720, Hungary
- MTA-SZTE
Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary
| | - Éva A. Enyedy
- Department
of Molecular and Analytical Chemistry, Interdisciplinary Excellence
Centre, University of Szeged, Dóm tér 7-8, Szeged H-6720, Hungary
- MTA-SZTE
Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary
| | - Denisa Darvasiova
- Institute
of Physical Chemistry and Chemical Physics, Faculty of Chemical and
Food Technology, Slovak University of Technology
in Bratislava, Bratislava SK-81237, Slovakia
| | - Peter Rapta
- Institute
of Physical Chemistry and Chemical Physics, Faculty of Chemical and
Food Technology, Slovak University of Technology
in Bratislava, Bratislava SK-81237, Slovakia
| | - Zuzana Barbieriková
- Institute
of Physical Chemistry and Chemical Physics, Faculty of Chemical and
Food Technology, Slovak University of Technology
in Bratislava, Bratislava SK-81237, Slovakia
| | - Daniel Vegh
- Institute
of Organic Chemistry, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Bratislava SK-81237, Slovakia
| | - Szilárd Tóth
- Institute
of Molecular Life Sciences, HUN-REN Research
Centre for Natural Sciences, Hungarian Research Network, Magyar Tudósok körútja
2, Budapest H-1117, Hungary
| | - Judit Tóth
- Institute
of Molecular Life Sciences, HUN-REN Research
Centre for Natural Sciences, Hungarian Research Network, Magyar Tudósok körútja
2, Budapest H-1117, Hungary
| | - Nóra Kucsma
- Institute
of Molecular Life Sciences, HUN-REN Research
Centre for Natural Sciences, Hungarian Research Network, Magyar Tudósok körútja
2, Budapest H-1117, Hungary
| | - Gergely Szakács
- Institute
of Molecular Life Sciences, HUN-REN Research
Centre for Natural Sciences, Hungarian Research Network, Magyar Tudósok körútja
2, Budapest H-1117, Hungary
- Center
for Cancer Research, Medical University
of Vienna, Vienna A-1090, Austria
| | - Ana Popović-Bijelić
- Faculty
of Physical Chemistry, University of Belgrade, Belgrade 11158, Serbia
| | - Ayesha Zafar
- School
of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Jóhannes Reynisson
- School
of Pharmacy and Bioengineering, Keele University, Newcastle-under-Lyme, Staffordshire ST5 5BG, United
Kingdom
| | - Anatoly D. Shutalev
- N.
D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow 119991, Russian Federation
| | - Ruoli Bai
- Molecular
Pharmacology Branch, Developmental Therapeutics Program, Division
of Cancer Diagnosis and Treatment, National Cancer Institute, Frederick
National Laboratory for Cancer Research, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Ernest Hamel
- Molecular
Pharmacology Branch, Developmental Therapeutics Program, Division
of Cancer Diagnosis and Treatment, National Cancer Institute, Frederick
National Laboratory for Cancer Research, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Vladimir B. Arion
- Institute
of Inorganic Chemistry, University of Vienna, Vienna A-1090, Austria
- Inorganic
Polymers Department, “Petru Poni”
Institute of Macromolecular Chemistry, Aleea Gr. Ghica Voda 41 A, Iasi 700487, Romania
| |
Collapse
|
13
|
Jin Z, Jiang L, He Q. Critical learning from industrial catalysis for nanocatalytic medicine. Nat Commun 2024; 15:3857. [PMID: 38719843 PMCID: PMC11079063 DOI: 10.1038/s41467-024-48319-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/26/2024] [Indexed: 05/12/2024] Open
Abstract
Systematical and critical learning from industrial catalysis will bring inspiration for emerging nanocatalytic medicine, but the relevant knowledge is quite limited so far. In this review, we briefly summarize representative catalytic reactions and corresponding catalysts in industry, and then distinguish the similarities and differences in catalytic reactions between industrial and medical applications in support of critical learning, deep understanding, and rational designing of appropriate catalysts and catalytic reactions for various medical applications. Finally, we summarize/outlook the present and potential translation from industrial catalysis to nanocatalytic medicine. This review is expected to display a clear picture of nanocatalytic medicine evolution.
Collapse
Affiliation(s)
- Zhaokui Jin
- Medical Center on Aging, Ruijin Hospital; Shanghai Key Laboratory of Hydrogen Science & Center of Hydrogen Science, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 510182, China
| | - Lingdong Jiang
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Qianjun He
- Medical Center on Aging, Ruijin Hospital; Shanghai Key Laboratory of Hydrogen Science & Center of Hydrogen Science, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
14
|
Wilke N, Frias C, Berkessel A, Prokop A. (2,6-Dimethylphenyl)arsonic Acid Induces Apoptosis through the Mitochondrial Pathway, Downregulates XIAP, and Overcomes Multidrug Resistance to Cytostatic Drugs in Leukemia and Lymphoma Cells In Vitro. Int J Mol Sci 2024; 25:4713. [PMID: 38731935 PMCID: PMC11083614 DOI: 10.3390/ijms25094713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/13/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Cancer treatment is greatly challenged by drug resistance, highlighting the need for novel drug discoveries. Here, we investigated novel organoarsenic compounds regarding their resistance-breaking and apoptosis-inducing properties in leukemia and lymphoma. Notably, the compound (2,6-dimethylphenyl)arsonic acid (As2) demonstrated significant inhibition of cell proliferation and induction of apoptosis in leukemia and lymphoma cells while sparing healthy leukocytes. As2 reached half of its maximum activity (AC50) against leukemia cells at around 6.3 µM. Further experiments showed that As2 overcomes multidrug resistance and sensitizes drug-resistant leukemia and lymphoma cell lines to treatments with the common cytostatic drugs vincristine, daunorubicin, and cytarabine at low micromolar concentrations. Mechanistic investigations of As2-mediated apoptosis involving FADD (FAS-associated death domain)-deficient or Smac (second mitochondria-derived activator of caspases)/DIABLO (direct IAP binding protein with low pI)-overexpressing cell lines, western blot analysis of caspase-9 cleavage, and measurements of mitochondrial membrane integrity identified the mitochondrial apoptosis pathway as the main mode of action. Downregulation of XIAP (x-linked inhibitor of apoptosis protein) and apoptosis induction independent of Bcl-2 (B-cell lymphoma 2) and caspase-3 expression levels suggest the activation of additional apoptosis-promoting mechanisms. Due to the selective apoptosis induction, the synergistic effects with common anti-cancer drugs, and the ability to overcome multidrug resistance in vitro, As2 represents a promising candidate for further preclinical investigations with respect to refractory malignancies.
Collapse
Affiliation(s)
- Nathalie Wilke
- Department of Pediatric Hematology/Oncology, Children’s Hospital Cologne, Amsterdamer Straße 59, 50735 Cologne, Germany
| | - Corazon Frias
- Department of Pediatric Hematology/Oncology, Children’s Hospital Cologne, Amsterdamer Straße 59, 50735 Cologne, Germany
- Department of Pediatric Oncology/Hematology, Helios Clinics Schwerin, Wismarsche Straße 393–397, 19049 Schwerin, Germany
- Medical School Hamburg (MSH), University of Applied Sciences and Medical University, Am Kaiserkai 1, 20457 Hamburg, Germany
| | - Albrecht Berkessel
- Department of Chemistry, Organic Chemistry, University of Cologne, Greinstraße 4, 50939 Cologne, Germany
| | - Aram Prokop
- Department of Pediatric Hematology/Oncology, Children’s Hospital Cologne, Amsterdamer Straße 59, 50735 Cologne, Germany
- Department of Pediatric Oncology/Hematology, Helios Clinics Schwerin, Wismarsche Straße 393–397, 19049 Schwerin, Germany
- Medical School Hamburg (MSH), University of Applied Sciences and Medical University, Am Kaiserkai 1, 20457 Hamburg, Germany
| |
Collapse
|
15
|
Bonsignore R, Trippodo E, Di Gesù R, Carreca AP, Rubino S, Spinello A, Terenzi A, Barone G. Novel half Salphen cobalt(III) complexes: synthesis, DNA binding and anticancer studies. Dalton Trans 2024; 53:6311-6322. [PMID: 38487871 DOI: 10.1039/d4dt00092g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
While platinum(II)-based drugs continue to be employed in cancer treatments, the escalating occurrence of severe side effects has spurred researchers to explore novel sources for potential therapeutic agents. Notably, cobalt(III) has emerged as a subject of considerable interest due to its ubiquitous role in human physiology. Several studies investigating the anticancer effects of Salphen complexes derived from cobalt(III) have unveiled intriguing antiproliferative properties. In a bid to enhance our understanding of this class of compounds, we synthesized and characterized two novel half Salphen cobalt(III) complexes. Both compounds exhibited notable stability, even in the presence of physiologically relevant concentrations of glutathione. The application of spectroscopic and computational methodologies unravelled their interactions with duplex and G4-DNAs, suggesting an external binding affinity for these structures, with preliminary indications of selectivity trends. Importantly, antiproliferative assays conducted on 3D cultured SW-1353 cancer cells unveiled a compelling anticancer activity at low micromolar concentrations, underscoring the potential therapeutic efficacy of this novel class of cobalt(III) complexes.
Collapse
Affiliation(s)
- Riccardo Bonsignore
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, Palermo 90128, Italy.
| | - Elisa Trippodo
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, Palermo 90128, Italy.
| | | | | | - Simona Rubino
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, Palermo 90128, Italy.
| | - Angelo Spinello
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, Palermo 90128, Italy.
| | - Alessio Terenzi
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, Palermo 90128, Italy.
| | - Giampaolo Barone
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, Palermo 90128, Italy.
| |
Collapse
|
16
|
Chen L, Zhao Z, Diarimalala RO, Chen Z, Wang Y, Zhan T, Zhao Y, Ma C, Wang X, Zhao C, Xiao Z, Hu K, Wu P. Tris-Functionalized Polyoxotungstovanadate-Mediated Antitumor Efficacy Involves Multiple Cell Death Pathways. Chem Biodivers 2024; 21:e202301898. [PMID: 38369765 DOI: 10.1002/cbdv.202301898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/27/2023] [Accepted: 02/17/2024] [Indexed: 02/20/2024]
Abstract
Polyoxometalates (POMs) are promising inorganic drug candidates for cancer chemotherapy. They are becoming attractive because of their easy accessibility and low cost. Herein, we report the synthesis and antitumor activity studies of four Lindqvist-type POMs with mixed-addenda atoms Na2[V4W2O16{(OCH2)3CR}] (R=-CH2OH, -CH3, -CH2CH3) and (Bu4N)2[V3W3{(OCH2)3CH2OOCCH2CH3}]. Compared with the current clinical applied antitumor drug 5-fluorouracil (5-FU) or Gemcitabine, analysis of MTT/CCK-8 assay, colony formation and wound healing assay revealed that the {V4W2} POMs had acceptable cytotoxicity in normal cells (293T) and significant inhibitory effects on cell proliferation and migration in three human tumor cell lines: human lung carcinoma cells (A549), human cervical carcinoma cells (HeLa), and human breast cancer cells (MCF-7). Interestingly, among the POMs analyzed, the therapeutic index (TI) of the {V4W2} POM with R= -CH2OH was relatively the most satisfactory. Thus, it was subsequently used for further studies. Flow cytometry analysis showed it prompted cellular apoptosis rate. qRT-PCR and Western blotting analysis indicated that multiple cell death pathways were activated including apoptosis, autophagy, necroptosis and pyroptosis during the POM-mediated antitumor process. In conclusion, our study shows that the polyoxotungstovanadate has great potential to be developed into a broad-spectrum antitumor chemotherapeutic drug.
Collapse
Affiliation(s)
- Lihong Chen
- Institute of POM-based Materials, New Materials and Green Manufacturing Talent Introduction and Innovation Demonstration Base, School of Materials and Chemical Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, PR China
| | - Zijia Zhao
- Sino-German Biomedical Center, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan, 430068, Hubei, PR China
| | - Rominah Onintsoa Diarimalala
- Sino-German Biomedical Center, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan, 430068, Hubei, PR China
| | - Zhongwei Chen
- Sino-German Biomedical Center, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan, 430068, Hubei, PR China
| | - Yu Wang
- Institute of POM-based Materials, New Materials and Green Manufacturing Talent Introduction and Innovation Demonstration Base, School of Materials and Chemical Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, PR China
| | - Taozhu Zhan
- Institute of POM-based Materials, New Materials and Green Manufacturing Talent Introduction and Innovation Demonstration Base, School of Materials and Chemical Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, PR China
| | - Yanchao Zhao
- Institute of POM-based Materials, New Materials and Green Manufacturing Talent Introduction and Innovation Demonstration Base, School of Materials and Chemical Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, PR China
| | - Chunhui Ma
- Institute of POM-based Materials, New Materials and Green Manufacturing Talent Introduction and Innovation Demonstration Base, School of Materials and Chemical Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, PR China
| | - Xingyue Wang
- Institute of POM-based Materials, New Materials and Green Manufacturing Talent Introduction and Innovation Demonstration Base, School of Materials and Chemical Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, PR China
| | - Chenqi Zhao
- Institute of POM-based Materials, New Materials and Green Manufacturing Talent Introduction and Innovation Demonstration Base, School of Materials and Chemical Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, PR China
| | - Zicheng Xiao
- Institute of POM-based Materials, New Materials and Green Manufacturing Talent Introduction and Innovation Demonstration Base, School of Materials and Chemical Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, PR China
| | - Kanghong Hu
- Sino-German Biomedical Center, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan, 430068, Hubei, PR China
| | - Pingfan Wu
- Institute of POM-based Materials, New Materials and Green Manufacturing Talent Introduction and Innovation Demonstration Base, School of Materials and Chemical Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, PR China
| |
Collapse
|
17
|
García-García A, Cristobal-Cueto P, Hidalgo T, Vitórica-Yrezábal IJ, Rodríguez-Diéguez A, Horcajada P, Rojas S. Potential antiprostatic performance of novel lanthanide-complexes based on 5-nitropicolinic acid. J Biol Inorg Chem 2024; 29:331-338. [PMID: 38717473 PMCID: PMC11111526 DOI: 10.1007/s00775-024-02054-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/20/2024] [Indexed: 05/24/2024]
Abstract
Two new lanthanide-complexes based on the 5-nitropicolinate ligand (5-npic) were obtained and fully characterized. Single-crystal X-ray diffraction revealed that these compounds are isostructural to a Dy-complex, previously published by us, based on dinuclear monomers link together with an extended hydrogen bond network, providing a final chemical formula of [Ln2(5-npic)6(H2O)4]·(H2O)2, where Ln = Dy (1), Gd (2), and Tb (3). Preliminary photoluminescent studies exhibited a ligand-centered emission for all complexes. The potential antitumoral activity of these materials was assayed in a prostatic cancer cell line (PC-3; the 2nd most common male cancerous disease), showing a significant anticancer activity (50-60% at 500 μg·mL-1). In turn, a high biocompatibility by both, the complexes and their precursors in human immunological HL-60 cells, was evidenced. In view of the strongest toxic effect in the tumoral cell line provided by the free 5-npic ligand (~ 40-50%), the overall anticancer complex performance seems to be triggered by the presence of this molecule.
Collapse
Affiliation(s)
- Amalia García-García
- Department of Inorganic Chemistry, Faculty of Science, University of Granada, Av. Fuente Nueva S/N, 18071, Granada, Spain
| | - Pablo Cristobal-Cueto
- Advanced Porous Material Unit, IMDEA Energy Institute, Av. Ramón de La Sagra 3, 28935, Móstoles, Madrid, Spain
| | - Tania Hidalgo
- Advanced Porous Material Unit, IMDEA Energy Institute, Av. Ramón de La Sagra 3, 28935, Móstoles, Madrid, Spain
| | - Iñigo J Vitórica-Yrezábal
- Department of Inorganic Chemistry, Faculty of Science, University of Granada, Av. Fuente Nueva S/N, 18071, Granada, Spain
| | - Antonio Rodríguez-Diéguez
- Department of Inorganic Chemistry, Faculty of Science, University of Granada, Av. Fuente Nueva S/N, 18071, Granada, Spain
| | - Patricia Horcajada
- Advanced Porous Material Unit, IMDEA Energy Institute, Av. Ramón de La Sagra 3, 28935, Móstoles, Madrid, Spain.
| | - Sara Rojas
- Department of Inorganic Chemistry, Faculty of Science, University of Granada, Av. Fuente Nueva S/N, 18071, Granada, Spain.
| |
Collapse
|
18
|
Grabher P, Kapitza P, Hörmann N, Scherfler A, Hermann M, Zwerger M, Varbanov HP, Kircher B, Baecker D, Gust R. Development of Cytotoxic GW7604-Zeise's Salt Conjugates as Multitarget Compounds with Selectivity for Estrogen Receptor- Positive Tumor Cells. J Med Chem 2024; 67:4870-4888. [PMID: 38478882 PMCID: PMC10983001 DOI: 10.1021/acs.jmedchem.3c02454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/07/2024] [Accepted: 02/28/2024] [Indexed: 04/04/2024]
Abstract
(E/Z)-3-(4-((E)-1-(4-Hydroxyphenyl)-2-phenylbut-1-enyl)phenyl)acrylic acid (GW7604) as a carrier was esterified with alkenols of various lengths and coordinated through the ethylene moiety to PtCl3, similar to Zeise's salt (K[PtCl3(C2H4)]). The resulting GW7604-Alk-PtCl3 complexes (Alk = Prop, But, Pent, Hex) degraded in aqueous solution only by exchange of the chlorido ligands. For example, GW7604-Pent-PtCl3 coordinated the amino acid alanine in the cell culture medium, bound the isolated nucleotide 5'-GMP, and interacted with the DNA (empty plasmid pSport1). It accumulated in estrogen receptor (ER)-positive MCF-7 cells primarily via cytosolic vesicles, while it was only marginally taken up in ER-negative SKBr3 cells. Accordingly, GW7604-Pent-PtCl3 and related complexes were inactive in SKBr3 cells. GW7604-Pent-PtCl3 showed high affinity to ERα and ERβ without mediating agonistic or ER downregulating properties. GW7604-Alk ligands also increased the cyclooxygenase (COX)-2 inhibitory potency of the complexes. In contrast to Zeise's salt, the GW7604-Alk-PtCl3 complexes inhibited COX-1 and COX-2 to the same extent.
Collapse
Affiliation(s)
- Patricia Grabher
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy, Center for Molecular
Biosciences Innsbruck, University of Innsbruck, Innrain 80/82, Innsbruck A-6020, Austria
| | - Paul Kapitza
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy, Center for Molecular
Biosciences Innsbruck, University of Innsbruck, Innrain 80/82, Innsbruck A-6020, Austria
| | - Nikolas Hörmann
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy, Center for Molecular
Biosciences Innsbruck, University of Innsbruck, Innrain 80/82, Innsbruck A-6020, Austria
| | - Amelie Scherfler
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy, Center for Molecular
Biosciences Innsbruck, University of Innsbruck, Innrain 80/82, Innsbruck A-6020, Austria
| | - Martin Hermann
- Department
of Anesthesiology & Critical Care Medicine, Medical University Innsbruck, Anichstraße 35, Innsbruck A-6020, Austria
| | - Michael Zwerger
- Department
of Pharmacognosy, Institute of Pharmacy, Center for Molecular Biosciences
Innsbruck, University of Innsbruck, Innrain 80/82, Innsbruck A-6020, Austria
| | - Hristo P. Varbanov
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy, Center for Molecular
Biosciences Innsbruck, University of Innsbruck, Innrain 80/82, Innsbruck A-6020, Austria
| | - Brigitte Kircher
- Department
of Internal Medicine V, Haematology & Oncology, Immunobiology
and Stem Cell Laboratory, Medical University
Innsbruck, Anichstraße
35, Innsbruck A-6020, Austria
- Tyrolean
Cancer Research Institute, Innrain 66, Innsbruck A-6020, Austria
| | - Daniel Baecker
- Department
of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße
2 + 4, Berlin D-14195, Germany
| | - Ronald Gust
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy, Center for Molecular
Biosciences Innsbruck, University of Innsbruck, Innrain 80/82, Innsbruck A-6020, Austria
| |
Collapse
|
19
|
Alajroush DR, Smith CB, Anderson BF, Oyeyemi IT, Beebe SJ, Holder AA. A Comparison of In Vitro Studies between Cobalt(III) and Copper(II) Complexes with Thiosemicarbazone Ligands to Treat Triple Negative Breast Cancer. Inorganica Chim Acta 2024; 562:121898. [PMID: 38282819 PMCID: PMC10810091 DOI: 10.1016/j.ica.2023.121898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Metal complexes have gained significant attention as potential anti-cancer agents. The anti-cancer activity of [Co(phen)2(MeATSC)](NO3)3•1.5H2O•C2H5OH 1 (where phen = 1,10-phenanthroline and MeATSC = 9-anthraldehyde-N(4)-methylthiosemicarbazone) and [Cu(acetylethTSC)Cl]Cl•0.25C2H5OH 2 (where acetylethTSC = (E)-N-ethyl-2-[1-(thiazol-2-yl)ethylidene]hydrazinecarbothioamide) was investigated by analyzing DNA cleavage activity. The cytotoxic effect was analyzed using CCK-8 viability assay. The activities of caspase 3/7, 9, and 1, reactive oxygen species (ROS) production, cell cycle arrest, and mitochondrial function were further analyzed to study the cell death mechanisms. Complex 2 induced a significant increase in nicked DNA. The IC50 values of complex 1 were 17.59 μM and 61.26 μM in cancer and non-cancer cells, respectively. The IC50 values of complex 2 were 5.63 and 12.19 μM for cancer and non-cancer cells, respectively. Complex 1 induced an increase in ROS levels, mitochondrial dysfunction, and activated caspases 3/7, 9, and 1, which indicated the induction of intrinsic apoptotic pathway and pyroptosis. Complex 2 induced cell cycle arrest in the S phase, ROS generation, and caspase 3/7 activation. Thus, complex 1 induced cell death in the breast cancer cell line via activation of oxidative stress which induced apoptosis and pyroptosis while complex 2 induced cell cycle arrest through the induction of DNA cleavage.
Collapse
Affiliation(s)
- Duaa R. Alajroush
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, U.S.A
| | - Chloe B. Smith
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, U.S.A
| | - Brittney F. Anderson
- Department of Biological Sciences, University of the Virgin Islands, 2 John Brewers Bay, St. Thomas, VI 00802, U.S.A
| | - Ifeoluwa T. Oyeyemi
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, U.S.A
- Department of Biological Sciences, University of Medical Sciences, Ondo City, Nigeria
| | - Stephen J. Beebe
- Frank Reidy Research center for Bioelectrics, Old Dominion University, 4211 Monarch Way, Suite 300, Norfolk, VA, 23508, U.S.A
| | - Alvin A. Holder
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, U.S.A
| |
Collapse
|
20
|
Marotta C, Cirri D, Kanavos I, Ronga L, Lobinski R, Funaioli T, Giacomelli C, Barresi E, Trincavelli ML, Marzo T, Pratesi A. Oxaliplatin(IV) Prodrugs Functionalized with Gemcitabine and Capecitabine Induce Blockage of Colorectal Cancer Cell Growth-An Investigation of the Activation Mechanism and Their Nanoformulation. Pharmaceutics 2024; 16:278. [PMID: 38399332 PMCID: PMC10892879 DOI: 10.3390/pharmaceutics16020278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
The use of platinum-based anticancer drugs, such as cisplatin, oxaliplatin, and carboplatin, is a common frontline option in cancer management, but they have debilitating side effects and can lead to drug resistance. Combination therapy with other chemotherapeutic agents, such as capecitabine and gemcitabine, has been explored. One approach to overcome these limitations is the modification of traditional Pt(II) drugs to obtain new molecules with an improved pharmacological profile, such as Pt(IV) prodrugs. The design, synthesis, and characterization of two novel Pt(IV) prodrugs based on oxaliplatin bearing the anticancer drugs gemcitabine or capecitabine in the axial positions have been reported. These complexes were able to dissociate into their constituents to promote cell death and induce apoptosis and cell cycle blockade in a representative colorectal cancer cell model. Specifically, the complex bearing gemcitabine resulted in being the most active on the HCT116 colorectal cancer cell line with an IC50 value of 0.49 ± 0.04. A pilot study on the encapsulation of these complexes in biocompatible PLGA-PEG nanoparticles is also included to confirm the retention of the pharmacological properties and cellular drug uptake, opening up to the possible delivery of the studied complexes through their nanoformulation.
Collapse
Affiliation(s)
- Carlo Marotta
- Department of Chemistry and Industrial Chemistry, University of Pisa, 56124 Pisa, Italy; (C.M.); (T.F.)
| | - Damiano Cirri
- Department of Chemistry and Industrial Chemistry, University of Pisa, 56124 Pisa, Italy; (C.M.); (T.F.)
| | - Ioannis Kanavos
- Institute of Analytical and Physical Chemistry for the Environment and Materials (IPREM-UMR 5254), Pau University, E2S UPPA, CNRS, 64053 Pau, France; (I.K.); (L.R.); (R.L.)
| | - Luisa Ronga
- Institute of Analytical and Physical Chemistry for the Environment and Materials (IPREM-UMR 5254), Pau University, E2S UPPA, CNRS, 64053 Pau, France; (I.K.); (L.R.); (R.L.)
| | - Ryszard Lobinski
- Institute of Analytical and Physical Chemistry for the Environment and Materials (IPREM-UMR 5254), Pau University, E2S UPPA, CNRS, 64053 Pau, France; (I.K.); (L.R.); (R.L.)
| | - Tiziana Funaioli
- Department of Chemistry and Industrial Chemistry, University of Pisa, 56124 Pisa, Italy; (C.M.); (T.F.)
| | - Chiara Giacomelli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.G.); (E.B.); (M.L.T.); (T.M.)
| | - Elisabetta Barresi
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.G.); (E.B.); (M.L.T.); (T.M.)
| | | | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.G.); (E.B.); (M.L.T.); (T.M.)
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry, University of Pisa, 56124 Pisa, Italy; (C.M.); (T.F.)
| |
Collapse
|
21
|
Jiménez-Pérez A, Fernández-Fariña S, Pedrido R, García-Tojal J. Desulfurization of thiosemicarbazones: the role of metal ions and biological implications. J Biol Inorg Chem 2024; 29:3-31. [PMID: 38148423 DOI: 10.1007/s00775-023-02037-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/26/2023] [Indexed: 12/28/2023]
Abstract
Thiosemicarbazones are biologically active substances whose structural formula is formed by an azomethine, an hydrazine, and a thioamide fragments, to generate a R2C=N-NR-C(=S)-NR2 backbone. These compounds often act as ligands to generate highly stable metal-organic complexes. In certain experimental conditions, however, thiosemicarbazones undergo reactions leading to the cleavage of the chain. Sometimes, the breakage involves desulfurization processes. The present work summarizes the different chemical factors that influence the desulfurization reactions of thiosemicarbazones, such as pH, the presence of oxidant reactants or the establishment of redox processes as those electrochemically induced, the effects of the solvent, the temperature, and the electromagnetic radiation. Many of these reactions require coordination of thiosemicarbazones to metal ions, even those present in the intracellular environment. The nature of the products generated in these reactions, their detection in vivo and in vitro, together with the relevance for the biological activity of these compounds, mainly as antineoplastic agents, is discussed.
Collapse
Affiliation(s)
- Alondra Jiménez-Pérez
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001, Burgos, Spain
| | - Sandra Fernández-Fariña
- Departamento de Química Inorgánica, Facultade de Química, Campus Vida, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Rosa Pedrido
- Departamento de Química Inorgánica, Facultade de Química, Campus Vida, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain.
| | - Javier García-Tojal
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001, Burgos, Spain.
| |
Collapse
|
22
|
Al-Salmi IK, Shongwe MS. Ternary Phenolate-Based Thiosemicarbazone Complexes of Copper(II): Magnetostructural Properties, Spectroscopic Features and Marked Selective Antiproliferative Activity against Cancer Cells. Molecules 2024; 29:431. [PMID: 38257344 PMCID: PMC10819714 DOI: 10.3390/molecules29020431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/06/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
The new diprotic ligand 3,5-di-tert-butylsalicylaldehyde 4-ethyl-3-thiosemicarbazone, abbreviated H2(3,5-t-Bu2)-sal4eT, exists as the thio-keto tautomer and adopts the E-configuration with respect to the imine double bond, as evidenced by single-crystal X-ray analysis and corroborated by spectroscopic characterisation. Upon treatment with Cu(OAc)2·H2O in the presence of either 2,9-dimethyl-1,10-phenanthroline (2,9-Me2-phen) or 1,10-phenanthroline (phen) as a co-ligand in MeOH, this thiosemicarbazone undergoes conformational transformation (relative donor-atom orientations: syn,anti → syn,syn) concomitantly with tautomerisation and double deprotonation to afford the ternary copper(II) complexes [Cu{(3,5-t-Bu2)-sal4eT}(2,9-Me2-phen)] (1) and [Cu2{3,5-t-Bu2)-sal4eT}2(phen)] (2). Crystallographic elucidation has revealed that complex 1 is a centrosymmetric dimer of mononuclear copper(II) complex molecules brought about by intermolecular H-bonding. The coordination geometry at the copper(II) centre is best described as distorted square pyramidal in accordance with the trigonality index (τ = 0.14). The co-ligand adopts an axial-equatorial coordination mode; hence, there is a disparity between its two Cu-N coordinate bonds arising from weakening of the apical one as a consequence of the tetragonal distortion. The axial X-band ESR spectrum of complex 1 is consistent with retention of this structure in solution. Complex 2 is a centrosymmetric dimer of dinuclear copper(II) complex molecules exhibiting intermolecular H-bonding and π-π-stacking interactions. The two copper(II) centres, which are 4.8067(18) Å apart and bridged by the thio-enolate nitrogen of the quadridentate thiosemicarbazonate ligand, display two different coordination geometries, one distorted square planar (τ4 = 0.082) and the other distorted square pyramidal (τ5 = 0.33). Such dinuclear copper(II) thiosemicarbazone complexes, which are crystallographically characterised, are extremely rare. In vitro, complexes 1 and 2 outperform cisplatin as antiproliferative agents in terms of potency and selectivity towards HeLa and MCF-7 cancer cell lines.
Collapse
Affiliation(s)
| | - Musa S. Shongwe
- Department of Chemistry, College of Science, Sultan Qaboos University, P.O. Box 36, Al-Khod 123, Muscat, Oman
| |
Collapse
|
23
|
Ma FJ, Huang X, Li XY, Tang SL, Li DJ, Cheng YZ, Azam M, Zhang LP, Sun D. Synthesis, structure and biological activity of silver(I) complexes containing triphenylphosphine and non-steroidal anti-inflammatory drug ligands. J Inorg Biochem 2023; 250:112404. [PMID: 39492372 DOI: 10.1016/j.jinorgbio.2023.112404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 11/05/2024]
Abstract
Two Ag(I) complexes containing triphenylphosphine and non-steroidal anti-inflammatory drug ligands were synthesized and investigated using various spectroscopic studies and single crystal X-ray crystallography. The binding properties of tolfenamic acid, ibuprofen and the two complexes with DNA and BSA were investigated using UV or fluorescence spectroscopy. The results showed that two Ag(I) complexes bound to DNA by the intercalation mode and interacted with BSA using a static quenching procedure. Furthermore, the results of fluorescence titration suggested that the complexes had good affinity for BSA and one binding site close to BSA. The in vitro cytotoxicity of tolfenamic acid, ibuprofen, and the two complexes against four human carcinoma cell lines (MCF-7, HepG-2, A549, and HeLa cells) was tested using an MTT assay. Complex 1 had higher cytotoxicity against HeLa cells. The intracellular reactive oxygen species (ROS) assay showed complex 1 induced the ROS generation in HeLa cells in a concentration dependent manner. Flow cytometry analysis showed complex 1 could suppress the HeLa cells growth during the G0/G1 phase and induce apoptosis in dose-depended manner.
Collapse
Affiliation(s)
- Feng-Jie Ma
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China
| | - Xiang Huang
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China
| | - Xue-Ying Li
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China
| | - Shi-Li Tang
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China
| | - De-Jun Li
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China
| | - Yuan-Zheng Cheng
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China.
| | - Mohammad Azam
- Department of Chemistry, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Li-Ping Zhang
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China.
| | - Di Sun
- School of Chemistry and Chemical Engineering, State Key Laboratory of Crystal Materials, Shandong University, Ji'nan 250100, PR China.
| |
Collapse
|
24
|
Tam JP, Huang J, Loo S, Li Y, Kam A. Ginsentide-like Coffeetides Isolated from Coffee Waste Are Cell-Penetrating and Metal-Binding Microproteins. Molecules 2023; 28:6556. [PMID: 37764332 PMCID: PMC10538209 DOI: 10.3390/molecules28186556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Coffee processing generates a huge amount of waste that contains many natural products. Here, we report the discovery of a panel of novel cell-penetrating and metal ion-binding microproteins designated coffeetide cC1a-c and cL1-6 from the husk of two popular coffee plants, Coffea canephora and Coffea liberica, respectively. Combining sequence determination and a database search, we show that the prototypic coffeetide cC1a is a 37-residue, eight-cysteine microprotein with a hevein-like cysteine motif, but without a chitin-binding domain. NMR determination of cC1a reveals a compact structure that confers its resistance to heat and proteolytic degradation. Disulfide mapping together with chemical synthesis reveals that cC1a has a ginsentide-like, and not a hevein-like, disulfide connectivity. In addition, transcriptomic analysis showed that the 98-residue micrcoproten-like coffeetide precursor contains a three-domain arrangement, like ginsentide precursors. Molecular modeling, together with experimental validation, revealed a Mg2+ and Fe3+ binding pocket at the N-terminus formed by three glutamic acids. Importantly, cC1a is amphipathic with a continuous stretch of 19 apolar amino acids, which enables its cell penetration to target intracellular proteins, despite being highly negatively charged. Our findings suggest that coffee by-products could provide a source of ginsentide-like bioactive peptides that have the potential to target intracellular proteins.
Collapse
Affiliation(s)
- James P. Tam
- Synthetic Enzymes and Natural Products Center, School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore; (J.H.); (S.L.); (Y.L.); (A.K.)
| | - Jiayi Huang
- Synthetic Enzymes and Natural Products Center, School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore; (J.H.); (S.L.); (Y.L.); (A.K.)
| | - Shining Loo
- Synthetic Enzymes and Natural Products Center, School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore; (J.H.); (S.L.); (Y.L.); (A.K.)
- Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Yimeng Li
- Synthetic Enzymes and Natural Products Center, School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore; (J.H.); (S.L.); (Y.L.); (A.K.)
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Antony Kam
- Synthetic Enzymes and Natural Products Center, School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore; (J.H.); (S.L.); (Y.L.); (A.K.)
- Department of Biological Sciences, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China
| |
Collapse
|
25
|
Favaron C, Gabano E, Zanellato I, Gaiaschi L, Casali C, Bottone MG, Ravera M. Effects of Ferrocene and Ferrocenium on MCF-7 Breast Cancer Cells and Interconnection with Regulated Cell Death Pathways. Molecules 2023; 28:6469. [PMID: 37764244 PMCID: PMC10537025 DOI: 10.3390/molecules28186469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
The effects of ferrocene (Fc) and ferrocenium (Fc+) induced in triple negative human breast cancer MCF-7 cells were explored by immunofluorescence, flow cytometry, and transmission electron microscopy analysis. The different abilities of Fc and Fc+ to produce reactive oxygen species and induce oxidative stress were clearly observed by activating apoptosis and morphological changes after treatment, but also after tests performed on the model organism D. discoideum, particularly in the case of Fc+. The induction of ferroptosis, an iron-dependent form of regulated cell death driven by an overload of lipid peroxides in cellular membranes, occurred after 2 h of treatment with Fc+ but not Fc. However, the more stable Fc showed its effects by activating necroptosis after a longer-lasting treatment. The differences observed in terms of cell death mechanisms and timing may be due to rapid interconversion between the two oxidative forms of internalized iron species (from Fe2+ to Fe3+ and vice versa). Potential limitations include the fact that iron metabolism and mitophagy have not been investigated. However, the ability of both Fc and Fc+ to trigger different and interregulated types of cell death makes them suitable to potentially overcome the shortcomings of traditional apoptosis-mediated anticancer therapies.
Collapse
Affiliation(s)
- Cristina Favaron
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy; (C.F.); (L.G.); (C.C.)
| | - Elisabetta Gabano
- Department of Sustainable Development and Ecological Transition, University of Piemonte Orientale, Piazza S. Eusebio 5, 13100 Vercelli, Italy;
| | - Ilaria Zanellato
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, Viale Teresa Michel 11, 15121 Alessandria, Italy
| | - Ludovica Gaiaschi
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy; (C.F.); (L.G.); (C.C.)
| | - Claudio Casali
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy; (C.F.); (L.G.); (C.C.)
| | - Maria Grazia Bottone
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy; (C.F.); (L.G.); (C.C.)
| | - Mauro Ravera
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, Viale Teresa Michel 11, 15121 Alessandria, Italy
| |
Collapse
|
26
|
Zhou L, Zhang Y, Xu Y, Jiang T, Tang L. Identification of a novel prognostic signature composed of 3 cuproptosis-related transcription factors in colon adenocarcinoma. Genes Genomics 2023; 45:1047-1061. [PMID: 37318704 DOI: 10.1007/s13258-023-01406-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/17/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Since the mechanism of cuproptosis was recently revealed, many molecules related to this pathway have been widely concerned and exploited to have prognostic potential. However, it is still unknown whether the transcription factors related to cuproptosis could be competent as tumor biomarkers of colon adenocarcinoma (COAD). OBJECTIVE To analyze the prognostic potential of cuproptosis-related transcription factors in COAD, and validate the representative molecule. METHODS Transcriptome data and patients' clinical parameters were obtained from the TCGA and GEO database. 19 cuproptosis genes were identified through literature consulting. Cuproptosis-related transcription factors were screened by COX regression analyses. Multivariate Cox regression was applied to construct the signature. Prognostic effects were evaluated by Kaplan Meier survival analyses and ROC analyses. KEGG, GO, and ssGSEA analyses were performed for function prediction. 48 COAD tissues were collected for immunohistochemistry stain to observe the expression level and prognostic value of E2F3. qRT-PCR was performed to detect mRNA expression levels, while cell viability assay was applied to detect the response of COAD cells to elesclomol treatment. RESULTS A novel signature based on 3 prognostic transcription factors related to cuproptosis was successfully established and verified. Patients in the low-risk group tended to have better overall survival and lower immune phenotype scores than those in the high-risk group. Meanwhile, we also constructed a nomogram based on this signature and predict 10 candidate compounds targeting this signature. As an essential member of this signature, E2F3 was confirmed to be overexpressed in COAD tissues and was associated with poor prognosis of COAD patients. Importantly, CuCl2 and cuproptosis inducer elesclomol treatment could increase the expression of E2F3 in COAD cell while the overexpression of E2F3 significantly enhanced the resistance of COAD cells to elesclomol treatment. CONCLUSION Our research has identified a new prognostic biomarker and provides some innovative insights into the diagnosis and therapy of patients with COAD.
Collapse
Affiliation(s)
- Lei Zhou
- The Graduate School, Dalian Medical University, Dalian, Liaoning, China
- Department of General Surgery, Yancheng Third People's Hospital, The Clinical Teaching Hospital of Jiangsu Vocational College of Medicine, Yancheng, Jiangsu, China
| | - Yuwan Zhang
- School of Management, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yixin Xu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Tao Jiang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China.
| | - Liming Tang
- The Graduate School, Dalian Medical University, Dalian, Liaoning, China.
- Department of General Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, No. 68 Gehu Middle Road, Changzhou, 213164, Jiangsu, China.
| |
Collapse
|
27
|
Engrácia DM, Pinto CIG, Mendes F. Cancer 3D Models for Metallodrug Preclinical Testing. Int J Mol Sci 2023; 24:11915. [PMID: 37569291 PMCID: PMC10418685 DOI: 10.3390/ijms241511915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 08/13/2023] Open
Abstract
Despite being standard tools in research, the application of cellular and animal models in drug development is hindered by several limitations, such as limited translational significance, animal ethics, and inter-species physiological differences. In this regard, 3D cellular models can be presented as a step forward in biomedical research, allowing for mimicking tissue complexity more accurately than traditional 2D models, while also contributing to reducing the use of animal models. In cancer research, 3D models have the potential to replicate the tumor microenvironment, which is a key modulator of cancer cell behavior and drug response. These features make cancer 3D models prime tools for the preclinical study of anti-tumoral drugs, especially considering that there is still a need to develop effective anti-cancer drugs with high selectivity, minimal toxicity, and reduced side effects. Metallodrugs, especially transition-metal-based complexes, have been extensively studied for their therapeutic potential in cancer therapy due to their distinctive properties; however, despite the benefits of 3D models, their application in metallodrug testing is currently limited. Thus, this article reviews some of the most common types of 3D models in cancer research, as well as the application of 3D models in metallodrug preclinical studies.
Collapse
Affiliation(s)
- Diogo M. Engrácia
- Center for Nuclear Sciences and Technologies, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal; (D.M.E.); (C.I.G.P.)
| | - Catarina I. G. Pinto
- Center for Nuclear Sciences and Technologies, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal; (D.M.E.); (C.I.G.P.)
| | - Filipa Mendes
- Center for Nuclear Sciences and Technologies, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal; (D.M.E.); (C.I.G.P.)
- Department of Nuclear Sciences and Engineering, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal
| |
Collapse
|
28
|
Zimmeter K, Vileno B, Platas-Iglesias C, Vinjamuri B, Sour A, Faller P. Derivatization of the Peptidic Xxx-Zzz-His Motif toward a Ligand with Attomolar Cu II Affinity under Maintaining High Selectivity and Fast Redox Silencing. Inorg Chem 2023. [PMID: 37269299 DOI: 10.1021/acs.inorgchem.3c00480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Cu chelation in biological systems is of interest as a tool to study the metabolism of this essential metal or for applications in the case of diseases with a systemic or local Cu overload, such as Wilson's or Alzheimer's disease. The choice of the chelating agent must meet several criteria. Among others, affinities and kinetics of metal binding and related metal selectivity are important parameters of the chelators to consider. Here, we report on the synthesis and characterization of Cu-binding properties of two ligands, L1 and L2, derivatives of the well-known peptidic CuII-binding motif Xxx-Zzz-His (also called ATCUN), where CuII is bound to the N-terminal amine, two amidates, and the imidazole. In either L, the N-terminal amine was replaced with a pyridine, and for L2, one amide was replaced with an amine compared to Xxx-Zzz-His. In particular, L2 showed several interesting features, including a CuII-binding affinity with a log KDapp = -16.0 similar to that of EDTA and stronger than all reported ATCUN peptides. L2 showed high selectivity for CuII over ZnII and other essential metal ions, even under the challenging conditions of the presence of human serum albumin. Further, L2 showed fast and efficient CuII redox silencing qualities and CuII-L2 was stable in the presence of mM GSH concentrations. Benefitting the fact that L2 can be easily elongated on its peptide part by standard SPPS to add other functions, L2 has attractive properties as a CuII chelator for application in biological systems.
Collapse
Affiliation(s)
- Katharina Zimmeter
- Institut de Chimie (UMR 7177), Université de Strasbourg, CNRS, 4 Rue Blaise Pascal, 67000 Strasbourg, France
| | - Bertrand Vileno
- Institut de Chimie (UMR 7177), Université de Strasbourg, CNRS, 4 Rue Blaise Pascal, 67000 Strasbourg, France
| | - Carlos Platas-Iglesias
- Departamento de Química Fundamental, Universidade da Coruña, Campus da Zapateira, Rúa da Fraga 10, 15008 A Coruña, Spain
| | - Bharath Vinjamuri
- Institut de Chimie (UMR 7177), Université de Strasbourg, CNRS, 4 Rue Blaise Pascal, 67000 Strasbourg, France
| | - Angélique Sour
- Institut de Chimie (UMR 7177), Université de Strasbourg, CNRS, 4 Rue Blaise Pascal, 67000 Strasbourg, France
| | - Peter Faller
- Institut de Chimie (UMR 7177), Université de Strasbourg, CNRS, 4 Rue Blaise Pascal, 67000 Strasbourg, France
- Institut Universitaire de France (IUF), 1 rue Descartes, 75231 Paris, France
| |
Collapse
|
29
|
Alfadul SM, Matnurov EM, Varakutin AE, Babak MV. Metal-Based Anticancer Complexes and p53: How Much Do We Know? Cancers (Basel) 2023; 15:2834. [PMID: 37345171 DOI: 10.3390/cancers15102834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 06/23/2023] Open
Abstract
P53 plays a key role in protecting the human genome from DNA-related mutations; however, it is one of the most frequently mutated genes in cancer. The P53 family members p63 and p73 were also shown to play important roles in cancer development and progression. Currently, there are various organic molecules from different structural classes of compounds that could reactivate the function of wild-type p53, degrade or inhibit mutant p53, etc. It was shown that: (1) the function of the wild-type p53 protein was dependent on the presence of Zn atoms, and (2) Zn supplementation restored the altered conformation of the mutant p53 protein. This prompted us to question whether the dependence of p53 on Zn and other metals might be used as a cancer vulnerability. This review article focuses on the role of different metals in the structure and function of p53, as well as discusses the effects of metal complexes based on Zn, Cu, Fe, Ru, Au, Ag, Pd, Pt, Ir, V, Mo, Bi and Sn on the p53 protein and p53-associated signaling.
Collapse
Affiliation(s)
- Samah Mutasim Alfadul
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, China
| | - Egor M Matnurov
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, China
| | - Alexander E Varakutin
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, China
| | - Maria V Babak
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, China
| |
Collapse
|
30
|
Abdolmaleki S, Panjehpour A, Aliabadi A, Khaksar S, Motieiyan E, Marabello D, Faraji MH, Beihaghi M. Cytotoxicity and mechanism of action of metal complexes: An overview. Toxicology 2023; 492:153516. [PMID: 37087063 DOI: 10.1016/j.tox.2023.153516] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 04/24/2023]
Abstract
After the discovery of cisplatin, many metal compounds were investigated for the therapy of diseases, especially cancer. The high therapeutic potential of metal-based compounds is related to the special properties of these compounds, such as their redox activity and ability to target vital biological sites. The overproduction of ROS and the consequent destruction of the membrane potential of mitochondria and/or the DNA helix is one of the known pathways leading to the induction of apoptosis by metal complexes. The apoptosis process can occur via the death receptor pathway and/or the mitochondrial pathway. The expression of Bcl2 proteins and the caspase family play critical roles in these pathways. In addition to apoptosis, autophagy is another process that regulates the suppression or promotion of various cancers through a dual action. On the other hand, the ability to interact with DNA is an important property found in several metal complexes with potent antiproliferative effects against cancer cells. These interactions were classified into two important categories: covalent/coordinated or subtle, and non-coordinated interactions. The anticancer activity of metal complexes is sometimes achieved by the simultaneous combination of several mechanisms. In this review, the anticancer effect of metal complexes is mechanistically discussed by different pathways, and some effective agents on their antiproliferative properties are explained.
Collapse
Affiliation(s)
- Sara Abdolmaleki
- School of Science and Technology, The University of Georgia, Tbilisi, Georgia
| | - Akram Panjehpour
- Department of Chemistry, Faculty of Sciences, Tarbiat Modares University, P.O. Box 14115-175, Tehran, Iran
| | - Alireza Aliabadi
- Pharmaceutical Sciences Research Center, Health Institute, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Samad Khaksar
- School of Science and Technology, The University of Georgia, Tbilisi, Georgia
| | - Elham Motieiyan
- Department of Chemistry, Payame Noor University, P. O. BOX 19395-4697, Tehran, Iran
| | - Domenica Marabello
- Dipartimento di Chimica, University of Torino Via P. Giuria 7, 10125 Torino, Italy; Interdepartmental Centre for Crystallography, University of Torino, Italy
| | - Mohammad Hossein Faraji
- Physiology Division, Department of Basic Science, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Maria Beihaghi
- School of Science and Technology, The University of Georgia, Tbilisi, Georgia; Department of Biology, Kavian Institute of Higher Education, Mashhad, Iran
| |
Collapse
|
31
|
Lončarević A, Ostojić K, Urlić I, Rogina A. Preparation and Properties of Bimetallic Chitosan Spherical Microgels. Polymers (Basel) 2023; 15:polym15061480. [PMID: 36987262 PMCID: PMC10057022 DOI: 10.3390/polym15061480] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023] Open
Abstract
The aim of this work was to prepare bimetallic chitosan microgels with high sphericity and investigate the influences of metal-ion type and content on the size, morphology, swelling, degradation and biological properties of microgels. Amino and hydroxyl groups of chitosan (deacetylation degree, DD, of 83.2% and 96.9%) served as ligands in the Cu2+–Zn2+/chitosan complexes with various contents of cupric and zinc ions. The electrohydrodynamic atomization process was used to produce highly spherical microgels with a narrow size distribution and with surface morphology changing from wrinkled to smooth by increasing Cu2+ ions’ quantity in bimetallic systems for both used chitosans. The size of the bimetallic chitosan particles was estimated to be between 60 and 110 µm for both used chitosans, and FTIR spectroscopy indicated the formation of complexes through physical interactions between the chitosans’ functional groups and metal ions. The swelling capacity of bimetallic chitosan particles decreases as the DD and copper (II) ion content increase as a result of stronger complexation with respect to zinc (II) ions. Bimetallic chitosan microgels showed good stability during four weeks of enzymatic degradation, and bimetallic systems with smaller amounts of Cu2+ ions showed good cytocompatibility for both used chitosans.
Collapse
Affiliation(s)
- Andrea Lončarević
- Faculty of Chemical Engineering and Technology, University of Zagreb, Trg Marka Marulića 19, HR-10000 Zagreb, Croatia
- Correspondence: (A.L.); (A.R.)
| | - Karla Ostojić
- Faculty of Science, University of Zagreb, Horvatovac 102a, HR-10000 Zagreb, Croatia
| | - Inga Urlić
- Faculty of Science, University of Zagreb, Horvatovac 102a, HR-10000 Zagreb, Croatia
| | - Anamarija Rogina
- Faculty of Chemical Engineering and Technology, University of Zagreb, Trg Marka Marulića 19, HR-10000 Zagreb, Croatia
- Correspondence: (A.L.); (A.R.)
| |
Collapse
|
32
|
Salerno S, Barresi E, Baglini E, Poggetti V, Da Settimo F, Taliani S. Target-Based Anticancer Indole Derivatives for the Development of Anti-Glioblastoma Agents. Molecules 2023; 28:molecules28062587. [PMID: 36985576 PMCID: PMC10056347 DOI: 10.3390/molecules28062587] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/28/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive and frequent primary brain tumor, with a poor prognosis and the highest mortality rate. Currently, GBM therapy consists of surgical resection of the tumor, radiotherapy, and adjuvant chemotherapy with temozolomide. Consistently, there are poor treatment options and only modest anticancer efficacy is achieved; therefore, there is still a need for the development of new effective therapies for GBM. Indole is considered one of the most privileged scaffolds in heterocyclic chemistry, so it may serve as an effective probe for the development of new drug candidates against challenging diseases, including GBM. This review analyzes the therapeutic benefit and clinical development of novel indole-based derivatives investigated as promising anti-GBM agents. The existing indole-based compounds which are in the pre-clinical and clinical stages of development against GBM are reported, with particular reference to the most recent advances between 2013 and 2022. The main mechanisms of action underlying their anti-GBM efficacy, such as protein kinase, tubulin and p53 pathway inhibition, are also discussed. The final goal is to pave the way for medicinal chemists in the future design and development of novel effective indole-based anti-GBM agents.
Collapse
|
33
|
Romani D, Marchetti F, Di Nicola C, Cuccioloni M, Gong C, Eleuteri AM, Galindo A, Fadaei-Tirani F, Nabissi M, Pettinari R. Multitarget-Directed Gallium(III) Tris(acyl-pyrazolonate) Complexes Induce Ferroptosis in Cancer Cells via Dysregulation of Cell Redox Homeostasis and Inhibition of the Mevalonate Pathway. J Med Chem 2023; 66:3212-3225. [PMID: 36802330 PMCID: PMC10009752 DOI: 10.1021/acs.jmedchem.2c01374] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
A series of Ga(Qn)3 coordination compounds have been synthesized, where HQn is 1-phenyl-3-methyl-4-RC(═O)-pyrazolo-5-one. The complexes have been characterized through analytical data, NMR and IR spectroscopy, ESI mass spectrometry, elemental analysis, X-ray crystallography, and density functional theory (DFT) studies. Cytotoxic activity against a panel of human cancer cell lines was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, with interesting results in terms of both cell line selectivity and toxicity values compared with cisplatin. The mechanism of action was explored by spectrophotometric, fluorometric, chromatographic, immunometric, and cytofluorimetric assays, SPR biosensor binding studies, and cell-based experiments. Cell treatment with gallium(III) complexes promoted several cell death triggering signals (accumulation of p27, PCNA, PARP fragments, activation of the caspase cascade, and inhibition of the mevalonate pathway) and induced changes in cell redox homeostasis (decreased levels of GSH/GPX4 and NADP(H), increased reactive oxygen species (ROS) and 4-hydroxynonenal (HNE), mitochondrial damage, and increased activity of CPR and CcO), identifying ferroptosis as the mechanism responsible for cancer cell death.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Agustín Galindo
- Departamento de Química Inorgánica, Facultad de Química, Universidad de Sevilla, Aptdo 1203, 41071 Sevilla, Spain
| | - Farzaneh Fadaei-Tirani
- Institut of Chemical Sciences and Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne CH-1015, Switzerland
| | | | | |
Collapse
|
34
|
Sunitha N, Raj CIS, Kumari BS. SYNTHESIS, SPECTRAL STUDIES, BIOLOGICAL EVALUATION AND MOLECULAR DOCKING STUDIES OF METAL COMPLEXES FROM COUMARIN DERIVATIVE. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
|
35
|
Nongpiur CGL, Verma AK, Ghate MM, Poluri KM, Kaminsky W, Kollipara MR. Synthesis, cytotoxicity and antibacterial activities of ruthenium, rhodium and iridium metal complexes containing diazafluorene functionalized ligands. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
36
|
Kazimir A, Schwarze B, Lönnecke P, Jelača S, Mijatović S, Maksimović-Ivanić D, Hey-Hawkins E. Metallodrugs against Breast Cancer: Combining the Tamoxifen Vector with Platinum(II) and Palladium(II) Complexes. Pharmaceutics 2023; 15:pharmaceutics15020682. [PMID: 36840003 PMCID: PMC9959148 DOI: 10.3390/pharmaceutics15020682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/30/2023] [Accepted: 02/09/2023] [Indexed: 02/19/2023] Open
Abstract
The luminal A-subtype of breast cancer, where the oestrogen receptor α (ERα) is overexpressed, is the most frequent one. The prodrug tamoxifen (1) is the clinically used agent, inhibiting the ERα activity via the formation of several active metabolites, such as 4-hydroxytamoxifen (2) or 4,4'-dihydroxytamoxifen (3). In this study, we present the tamoxifen derivative 4-[1,1-bis(4-methoxyphenyl)but-1-en-2-yl]-2,2'-bipyridine (4), which was combined with platinum or palladium dichloride, the former a well-known scaffold in anticancer treatment, to give [PtCl2(4-κ2N,N')] (5) or [PdCl2(4-κ2N,N'] (6). To prevent fast exchange of weakly coordinating chlorido ligands in aqueous solution, a bulky, highly stable and hydrophobic nido-carborate(-2) ([C2B9H11]2-) was incorporated. The resulting complexes [3-(4-κ2N,N')-3,1,2-PtC2B9H11] (7) and [3-(4-κ2N,N')-3,1,2-PdC2B9H11] (8) exhibit a dramatic change in electronic and biological properties compared to 5 and 6. Thus, 8 is highly selective for triple-negative MDA-MB-231 cells (IC50 = 3.7 μM, MTT test), while 7 is completely inactive against this cell line. The observed cytotoxicity of compounds 4-6 and 8 against this triple-negative cell line suggests off-target mechanisms rather than only ERα inhibition, for which these compounds were originally designed. Spectroscopic properties and electronic structures of the metal complexes were investigated for possible explanations of the biological activities.
Collapse
Affiliation(s)
- Aleksandr Kazimir
- Institute of Inorganic Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University, 04103 Leipzig, Germany
| | - Benedikt Schwarze
- Institute for Medical Physics and Biophysics, Medical Faculty, Leipzig University, 04107 Leipzig, Germany
| | - Peter Lönnecke
- Institute of Inorganic Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University, 04103 Leipzig, Germany
| | - Sanja Jelača
- Department of Immunology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 11060 Belgrade, Serbia
| | - Sanja Mijatović
- Department of Immunology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 11060 Belgrade, Serbia
| | - Danijela Maksimović-Ivanić
- Department of Immunology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 11060 Belgrade, Serbia
| | - Evamarie Hey-Hawkins
- Institute of Inorganic Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University, 04103 Leipzig, Germany
- Correspondence:
| |
Collapse
|
37
|
Influence of the Fatty Acid Metabolism on the Mode of Action of a Cisplatin(IV) Complex with Phenylbutyrate as Axial Ligands. Pharmaceutics 2023; 15:pharmaceutics15020677. [PMID: 36839999 PMCID: PMC9967619 DOI: 10.3390/pharmaceutics15020677] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/08/2023] [Accepted: 02/11/2023] [Indexed: 02/19/2023] Open
Abstract
For a variety of cancer types, platinum compounds are still among the best treatment options. However, their application is limited by side effects and drug resistance. Consequently, multi-targeted platinum(IV) prodrugs that target specific traits of the malignant tissue are interesting new candidates. Recently, cisPt(PhB)2 was synthesized which, upon reduction in the malignant tissue, releases phenylbutyrate (PhB), a metabolically active fatty acid analog, in addition to cisplatin. In this study, we in-depth investigated the anticancer properties of this new complex in cell culture and in mouse allograft experiments. CisPt(PhB)2 showed a distinctly improved anticancer activity compared to cisplatin as well as to PhB alone and was able to overcome various frequently occurring drug resistance mechanisms. Furthermore, we observed that differences in the cellular fatty acid metabolism and mitochondrial activity distinctly impacted the drug's mode of action. Subsequent analyses revealed that "Warburg-like" cells, which are characterized by deficient mitochondrial function and fatty acid catabolism, are less capable of coping with cisPt(PhB)2 leading to rapid induction of a non-apoptotic form of cell death. Summarizing, cisPt(PhB)2 is a new orally applicable platinum(IV) prodrug with promising activity especially against cisplatin-resistant cancer cells with "Warburg-like" properties.
Collapse
|
38
|
Bourouai MA, Si Larbi K, Bouchoucha A, Terrachet-Bouaziz S, Djebbar S. New Ni(II) and Pd(II) complexes bearing derived sulfa drug ligands: synthesis, characterization, DFT calculations, and in silico and in vitro biological activity studies. Biometals 2023; 36:153-188. [PMID: 36427181 DOI: 10.1007/s10534-022-00469-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/16/2022] [Indexed: 11/26/2022]
Abstract
In the present study, the synthesis of six new Ni(II) and Pd(II) complexes with three derived sulfamethoxazole drug ligands is reported. The coordination mode, geometry, and chemical formula of all the synthesized compounds have been determined by elemental analysis, mass spectrometry, emission atomic spectroscopy, conductivity measurements, magnetic susceptibility, FTIR, TGA, 1H-NMR, electronic absorption spectroscopy, SEM-EDX along with DFT calculations. The Schiff Base ligands were found to be bidentate and coordinated to the metal ions through sulfonamidic nitrogen and oxazolic nitrogen atoms leading to a square planar geometry for palladium (II) while a distorted octahedral geometry around Nickel (II) ion was suggested. Biological applications of the new complexes including in vitro antimicrobial, antioxidant and anticancer properties were investigated. The results showed that the new metal (II) compounds exhibit remarkable antibacterial inhibition activity against both Gram-positive and Gram-negative bacteria, in addition to noticeable DPPH free radical scavenging activity. The in vitro cytotoxicity assay of the complexes against cell lines of chronic myelogenous leukaemia (K562) showed promising potential for the application of the coordination compounds in antitumor therapy. Subsequently, to evaluate the pharmaceutical potential of the metal-containing compounds, pharmacokinetics and toxicity were studied by ADMET simulations while interactions between the complexes and bacterial proteins were evaluated by molecular docking.
Collapse
Affiliation(s)
- Mohamed Amine Bourouai
- Hydrometallurgy and Molecular Inorganic Chemistry Laboratory, Faculty of Chemistry, University of Science and Technology Houari Boumediene, BP 32 El Alia, 16111, Algiers, Algeria
| | - Karima Si Larbi
- Hydrometallurgy and Molecular Inorganic Chemistry Laboratory, Faculty of Chemistry, University of Science and Technology Houari Boumediene, BP 32 El Alia, 16111, Algiers, Algeria
| | - Afaf Bouchoucha
- Hydrometallurgy and Molecular Inorganic Chemistry Laboratory, Faculty of Chemistry, University of Science and Technology Houari Boumediene, BP 32 El Alia, 16111, Algiers, Algeria.
| | | | - Safia Djebbar
- Hydrometallurgy and Molecular Inorganic Chemistry Laboratory, Faculty of Chemistry, University of Science and Technology Houari Boumediene, BP 32 El Alia, 16111, Algiers, Algeria
| |
Collapse
|
39
|
Silva HVR, da Silva GÁF, Zavan B, Machado RP, de Araujo-Neto JH, Ellena JA, Ionta M, Barbosa MIF, Doriguetto AC. The nicotinamide ruthenium(II) complex induces the production of reactive oxygen species (ROS), cell cycle arrest, and apoptosis in melanoma cells. Polyhedron 2023. [DOI: 10.1016/j.poly.2022.116267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
40
|
Bonsignore G, Martinotti S, Ranzato E. Endoplasmic Reticulum Stress and Cancer: Could Unfolded Protein Response Be a Druggable Target for Cancer Therapy? Int J Mol Sci 2023; 24:ijms24021566. [PMID: 36675080 PMCID: PMC9865308 DOI: 10.3390/ijms24021566] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Unfolded protein response (UPR) is an adaptive response which is used for re-establishing protein homeostasis, and it is triggered by endoplasmic reticulum (ER) stress. Specific ER proteins mediate UPR activation, after dissociation from chaperone Glucose-Regulated Protein 78 (GRP78). UPR can decrease ER stress, producing an ER adaptive response, block UPR if ER homeostasis is restored, or regulate apoptosis. Some tumour types are linked to ER protein folding machinery disturbance, highlighting how UPR plays a pivotal role in cancer cells to keep malignancy and drug resistance. In this review, we focus on some molecules that have been revealed to target ER stress demonstrating as UPR could be a new target in cancer treatment.
Collapse
|
41
|
Latonduine-1-Amino-Hydantoin Hybrid, Triazole-Fused Latonduine Schiff Bases and Their Metal Complexes: Synthesis, X-ray and Electron Diffraction, Molecular Docking Studies and Antiproliferative Activity. INORGANICS 2023. [DOI: 10.3390/inorganics11010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
A series of latonduine derivatives, namely 11-nitro-indolo[2,3-d]benzazepine-7-(1-amino-hydantoin) (B), triazole-fused indolo[2,3-d]benzazepine-based Schiff bases HL1 and HL2 and metal complexes [M(p-cymene)(HL1)Cl]Cl, where M = Ru (1), Os (2), and [Cu(HL2)Cl2] (3) were synthesized and characterized by spectroscopic techniques (UV–vis, 1H, 13C, 15N–1H HSQC NMR) and ESI mass spectrometry. The molecular structures of B and HL1 were confirmed by single-crystal X-ray diffraction, while that of 3 by electron diffraction of nanometer size crystalline sample. Molecular docking calculations of species B in the binding pocket of PIM-1 enzyme revealed that the 1-amino-hydantoin moiety is not involved in any hydrogen-bonding interactions, even though a good accommodation of the host molecule in the ATP binding pocket of the enzyme was found. The antiproliferative activity of organic compounds B, HL1 and HL2, as well as complexes 1–3 was investigated in lung adenocarcinoma A549, colon adenocarcinoma LS-174 and triple-negative breast adenocarcinoma MDA-MB-231 cells and normal human lung fibroblast cells MRC-5 by MTT assays; then, the results are discussed.
Collapse
|
42
|
Targeting emerging cancer hallmarks by transition metal complexes: Epigenetic reprogramming and epitherapies. Part II. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
43
|
Marotta C, Giorgi E, Binacchi F, Cirri D, Gabbiani C, Pratesi A. An overview of recent advancements in anticancer Pt(IV) prodrugs: New smart drug combinations, activation and delivery strategies. Inorganica Chim Acta 2023. [DOI: 10.1016/j.ica.2023.121388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
44
|
Liu Y, Zhou Z, Li Y, Qin J, Wang X, Lu C, Wu W. Efficient Removal of Dibenzothiophene (DBT) over WO3–Mo–Al Catalysts in the Extractive Catalytic Oxidative Desulfurization (ECODS) System. RUSSIAN JOURNAL OF PHYSICAL CHEMISTRY A 2022. [DOI: 10.1134/s0036024422120329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
45
|
Ertürk AG, Sekeroglu V, Yildirim E, Dindaroglu G, Sekeroglu ZA. Antipyrine derived-Schiff base copper complex: Synthesis, characterization, and in vitro evaluation. Inorganica Chim Acta 2022. [DOI: 10.1016/j.ica.2022.121146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
46
|
Mone N, Harihar S, Salunke-Gawali S, Satpute S, Patil A, Mokashi V, Jadhav M, Butcher RJ. Metal complexes of ‘Heena’ (2-hydroxy-1,4-naphthoquinone): Synthesis Characterization and Anticancer activity. Inorganica Chim Acta 2022. [DOI: 10.1016/j.ica.2022.121290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
47
|
Murillo MI, Gaiddon C, Le Lagadec R. Targeting of the intracellular redox balance by metal complexes towards anticancer therapy. Front Chem 2022; 10:967337. [PMID: 36034648 PMCID: PMC9405673 DOI: 10.3389/fchem.2022.967337] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
The development of cancers is often linked to the alteration of essential redox processes, and therefore, oxidoreductases involved in such mechanisms can be considered as attractive molecular targets for the development of new therapeutic strategies. On the other hand, for more than two decades, transition metals derivatives have been leading the research on drugs as alternatives to platinum-based treatments. The success of such compounds is particularly due to their attractive redox kinetics properties, favorable oxidation states, as well as routes of action different to interactions with DNA, in which redox interactions are crucial. For instance, the activity of oxidoreductases such as PHD2 (prolyl hydroxylase domain-containing protein) which can regulate angiogenesis in tumors, LDH (lactate dehydrogenase) related to glycolysis, and enzymes, such as catalases, SOD (superoxide dismutase), TRX (thioredoxin) or GSH (glutathione) involved in controlling oxidative stress, can be altered by metal effectors. In this review, we wish to discuss recent results on how transition metal complexes have been rationally designed to impact on redox processes, in search for effective and more specific cancer treatments.
Collapse
Affiliation(s)
- María Isabel Murillo
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, Mexico
| | - Christian Gaiddon
- Strasbourg Université, Inserm UMR_S U1113, IRFAC, Strasbourg, France
| | - Ronan Le Lagadec
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, Mexico
- *Correspondence: Ronan Le Lagadec,
| |
Collapse
|
48
|
Falcone E, Ritacca AG, Hager S, Schueffl H, Vileno B, El Khoury Y, Hellwig P, Kowol CR, Heffeter P, Sicilia E, Faller P. Copper-Catalyzed Glutathione Oxidation is Accelerated by the Anticancer Thiosemicarbazone Dp44mT and Further Boosted at Lower pH. J Am Chem Soc 2022; 144:14758-14768. [PMID: 35929814 PMCID: PMC9389589 DOI: 10.1021/jacs.2c05355] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
![]()
Glutathione (GSH) is the most abundant thiol in mammalian
cells
and plays a crucial role in maintaining redox cellular homeostasis.
The thiols of two GSH molecules can be oxidized to the disulfide GSSG.
The cytosolic GSH/GSSG ratio is very high (>100), and its reduction
can lead to apoptosis or necrosis, which are of interest in cancer
research. CuII ions are very efficient oxidants of thiols,
but with an excess of GSH, CuIn(GS)m clusters are formed, in which CuI is very slowly reoxidized by O2 at pH 7.4 and
even more slowly at lower pH. Here, the aerobic oxidation of GSH by
CuII was investigated at different pH values in the presence
of the anticancer thiosemicarbazone Dp44mT, which accumulates in lysosomes
and induces lysosomal membrane permeabilization in a Cu-dependent
manner. The results showed that CuII-Dp44mT catalyzes GSH
oxidation faster than CuII alone at pH 7.4 and hence accelerates
the production of very reactive hydroxyl radicals. Moreover, GSH oxidation
and hydroxyl radical production by CuII-Dp44mT were accelerated
at the acidic pH found in lysosomes. To decipher this unusually faster
thiol oxidation at lower pH, density functional theory (DFT) calculations,
electrochemical and spectroscopic studies were performed. The results
suggest that the acceleration is due to the protonation of CuII-Dp44mT on the hydrazinic nitrogen, which favors the rate-limiting
reduction step without subsequent dissociation of the CuI intermediate. Furthermore, preliminary biological studies in cell
culture using the proton pump inhibitor bafilomycin A1 indicated that
the lysosomal pH plays a role in the activity of CuII-Dp44mT.
Collapse
Affiliation(s)
- Enrico Falcone
- Institut de Chimie (UMR 7177), University of Strasbourg - CNRS, 4 Rue Blaise Pascal, 67081 Strasbourg, France
| | - Alessandra G Ritacca
- Department of Chemistry and Chemical Technologies, Università della Calabria, Ponte P. Bucci, 87036 Arcavacata di Rende, (CS), Italy
| | - Sonja Hager
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Hemma Schueffl
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Bertrand Vileno
- Institut de Chimie (UMR 7177), University of Strasbourg - CNRS, 4 Rue Blaise Pascal, 67081 Strasbourg, France
| | - Youssef El Khoury
- Laboratoire de bioélectrochimie et spectroscopie, UMR 7140, CNRS, Université de Strasbourg, 4 Rue Blaise Pascal, 67081 Strasbourg, France
| | - Petra Hellwig
- Laboratoire de bioélectrochimie et spectroscopie, UMR 7140, CNRS, Université de Strasbourg, 4 Rue Blaise Pascal, 67081 Strasbourg, France
| | - Christian R Kowol
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Straße 42, 1090 Vienna, Austria
| | - Petra Heffeter
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Emilia Sicilia
- Department of Chemistry and Chemical Technologies, Università della Calabria, Ponte P. Bucci, 87036 Arcavacata di Rende, (CS), Italy
| | - Peter Faller
- Institut de Chimie (UMR 7177), University of Strasbourg - CNRS, 4 Rue Blaise Pascal, 67081 Strasbourg, France.,Institut Universitaire de France (IUF), 1 rue Descartes, 75231 Paris, France
| |
Collapse
|
49
|
Ashfaq M, Ali A, Tahir MN, Kuznetsov A, Munawar KS, Muhammad S. Synthesis, single-crystal exploration, hirshfeld surface analysis, and DFT investigation of the thiosemicarbazones. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
50
|
Abstract
The first appearance of SARS-CoV-2 is dated back to 2019. This new member of the coronavirus family has caused more than 5 million deaths worldwide up until the end of January 2022. At the moment, and after intensive vaccination programmes throughout the world, the pandemic is still active, whilst new mutations constantly appear. Researchers are working intensively to discover antiviral drugs to combat the severe cases in intensive care units, giving the overloaded hospital units a breather. Alongside various research projects focusing on developing small pharmaceutical molecules, a significant proportion of the research community has shifted towards paying attention to metal drugs. In this small review, we make brief reference to the use of metal drugs in therapeutics and provide some examples of metal drugs that are of extreme interest in the current pandemic. At the same time, we will also examine some of their promising mechanisms of action and possible effectiveness against COVID-19.
Collapse
Affiliation(s)
- Kyriacos Ioannou
- Department of Life and Health Sciences, University of Nicosia, 2417, Nicosia, Cyprus
| | - Manos C Vlasiou
- Department of Life and Health Sciences, University of Nicosia, 2417, Nicosia, Cyprus.
| |
Collapse
|