1
|
Garrosa-Miró Y, Muñoz-Moreno L, D'Errico G, Tancredi M, Carmena MJ, Ottaviani MF, Ortega P, de la Mata J. Ruthenium(II) and copper(II) polyamine complexes as promising antitumor agents: synthesis, characterization, and biological evaluation. Dalton Trans 2025; 54:7506-7521. [PMID: 40232207 DOI: 10.1039/d4dt03377a] [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: 04/16/2025]
Abstract
Ruthenium or copper complexes have emerged as some of the most promising alternatives for the treatment of many types of cancer. They have enhanced activity, greater selectivity and reduced side effects compared to their predecessors, cisplatin and its analogues. On the other hand, polyamine metabolism is often deregulated in cancer, leading to increased intracellular concentrations of polyamines that promote cell proliferation, differentiation, and tumorigenesis. In the present work, we report the synthesis and characterization of a family of mono- and binuclear Ru(II) and Cu(II) complexes functionalized with polyamine ligands derived from norspermine. The computer-aided analysis of the electron paramagnetic resonance (EPR) spectra provided magnetic and dynamic parameters, which helped to identify prevalent Cu-N2 coordination in a partially distorted square planar geometry of the Cu(II) complexes and the flexibility of the complexes in solution, slowed down by both the complex size and the hydrophobic interactions between chains. In vitro studies focused on advanced prostate cancer have demonstrated that these new metal complexes present a high level of cytotoxicity against PC3 cells. Furthermore, these metallic compounds exhibit the ability to inhibit cell adhesion and migration while reducing intracellular reactive oxygen species levels, which are key factors of metastasis.
Collapse
Affiliation(s)
- Yoel Garrosa-Miró
- Universidad de Alcalá, Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. del Río" (IQAR), Madrid, Spain.
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
- Institute "Ramón y Cajal" for Health Research (IRYCIS), Spain
| | - Laura Muñoz-Moreno
- Universidad de Alcalá, Department of Biology of Systems, Biochemistry and Molecular Biology Unit, Madrid, Spain
| | - Gerardino D'Errico
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 4, Complesso Universitario di Monte Sant'Angelo, I-80126 Naples, Italy
| | - Matilde Tancredi
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 4, Complesso Universitario di Monte Sant'Angelo, I-80126 Naples, Italy
| | - M Jose Carmena
- Universidad de Alcalá, Department of Biology of Systems, Biochemistry and Molecular Biology Unit, Madrid, Spain
| | - M Francesca Ottaviani
- Department of Pure and Applied Sciences, University of Urbino "Carlo Bo", Via Saffi 2, I-61029 Urbino, Italy
| | - Paula Ortega
- Universidad de Alcalá, Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. del Río" (IQAR), Madrid, Spain.
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
- Institute "Ramón y Cajal" for Health Research (IRYCIS), Spain
| | - Javier de la Mata
- Universidad de Alcalá, Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. del Río" (IQAR), Madrid, Spain.
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
- Institute "Ramón y Cajal" for Health Research (IRYCIS), Spain
| |
Collapse
|
2
|
Ganci D, D'Anna L, Abruscato G, Le Chevalier M, Quideau O, Cataldo S, Pettignano A, Rubino S, Chiarelli R, Barone G, Luparello C, Bonsignore R. Harnessing redox reactions for anticancer effects: A copper(II) Schiff base complex induces apoptosis in HepG2 liver cancer cells via ROS generation. J Inorg Biochem 2025; 270:112938. [PMID: 40344694 DOI: 10.1016/j.jinorgbio.2025.112938] [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: 12/02/2024] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/11/2025]
Abstract
This study uncovers the potential of a copper(II) Schiff base complex, CuL2+, to access the Cu(I) oxidation state and generate reactive oxygen species (ROS), highlighting its significance in eventual therapeutic applications. UV-vis absorption spectroscopy was used to follow the redox stability of the metal complex, also in the presence of reducing agents, such as ascorbic acid and glutathione, and of the copper(I) chelator, bathocuproine disulfonate. Utilizing human tumor cell lines HepG2 (hepatocarcinoma cells), we assessed its efficacy in reducing cell viability, increasing the sub-G0/G1 cell fraction, and initiating apoptotic pathways. Cell viability assays demonstrated a dose-dependent cytotoxicity with pronounced effects at sub-micromolar concentrations. Flow cytometry revealed significant ROS production, followed by mitochondrial membrane potential dissipation, and caspase activation, underscoring CuL2+'s mechanism of action. These findings position CuL2+ as a promising candidate for cancer therapy, providing insights into copper complexes' therapeutic application through oxidative stress and apoptosis modulation.
Collapse
Affiliation(s)
- Daniela Ganci
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Luisa D'Anna
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Giulia Abruscato
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy
| | | | - Océane Quideau
- Institut National des Sciences Appliquées de Rouen, 76800 Saint-Étienne-du-Rouvray, France
| | - Salvatore Cataldo
- Dipartimento di Fisica e Chimica - Emilio Segrè, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Alberto Pettignano
- Dipartimento di Fisica e Chimica - Emilio Segrè, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Simona Rubino
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Roberto Chiarelli
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Giampaolo Barone
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Claudio Luparello
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Riccardo Bonsignore
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy.
| |
Collapse
|
3
|
Deng Y, Mo X, Lai SKM, Haw SC, Au-Yeung HY, Tse ECM. Mechanical and Covalent Tailoring of Copper Catenanes for Selective Aqueous Nitrate-to-Ammonia Electrocatalysis. J Am Chem Soc 2025; 147:14316-14325. [PMID: 40260598 PMCID: PMC12046556 DOI: 10.1021/jacs.4c18547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 04/23/2025]
Abstract
Electrocatalytic nitrate reduction reaction (NO3RR) for the selective generation of ammonia (NH3) enables the removal of deleterious nitrate pollutants while simultaneously upcycling them into a value-added fertilizer. The development of nonprecious metal-derived catalysts such as those featuring copper (Cu) as earth-abundant alternatives for the state-of-the-art precious metal catalysts is of urgent need yet suffering from the activity-selectivity-durability trilemma. Rational design of molecular Cu complexes with well-defined coordination structures permitting systematic structure-activity relationship (SAR) investigations is key to addressing the challenge. Here, a series of molecular Cu(I) complexes with [2]catenane ligands are developed as NO3RR electrocatalysts for the first time. By engineering multiple cationic ammoniums on the catenane backbone, acceptance of the anionic nitrate substrate as well as the release of the cationic ammonium product are promoted, thereby facilitating a higher Faradaic efficiency and product selectivity toward ammonia via an 8e- pathway. Of note, the mutual Coulombic repulsion between the multiply charged ligands is overcome by the mechanical interlocking such that the catalyst integrity can be maintained under practical conditions. This report highlights the promise of employing mechanically interlocked ligands as a platform for customizing metal complexes as catalysts for redox processes involving multiple proton-coupled electron transfer steps.
Collapse
Affiliation(s)
- Yulin Deng
- HKU-CAS
Joint Laboratory on New Materials & Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, P. R. China
| | - Xiaoyong Mo
- HKU-CAS
Joint Laboratory on New Materials & Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, P. R. China
| | - Samuel Kin-Man Lai
- HKU-CAS
Joint Laboratory on New Materials & Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, P. R. China
| | - Shu-Chih Haw
- National
Synchrotron Radiation Research Center, 101 Hsin-Ann Road, Hsinchu 30076, Taiwan
| | - Ho Yu Au-Yeung
- HKU-CAS
Joint Laboratory on New Materials & Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, P. R. China
- State
Key Laboratory of Synthetic Chemistry, The
University of Hong Kong, Pokfulam Road, Hong Kong SAR, P. R. China
| | - Edmund C. M. Tse
- HKU-CAS
Joint Laboratory on New Materials & Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, P. R. China
| |
Collapse
|
4
|
Riediger T, Stammler A, Oldengott J, Walleck S, Glaser T. Support for the Anticipated Binding Mode of a Cytotoxic Dinuclear Copper Complex to Two Neighboring Phosphate Esters of the DNA Backbone. Inorg Chem 2025. [PMID: 40298861 DOI: 10.1021/acs.inorgchem.5c00505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The cytotoxic dinuclear complex [(Htom6-Me){CuII(OAc)}2](OAc) (H2tom6-Me = 2,7-bis(di(6-methylpyridine-2-yl-methyl)aminomethyl)-1,8-naphthalenediol) binds to DNA, interferes with DNA synthesis in PCR at lower concentrations than cisplatin, and kills human cancer cells more efficiently than human stem cells of the same proliferation rate. It is supposed that the OAc- ligands dissociate in aqueous buffer, providing [(Htom6-Me){CuII(OH2)}2]3+, while the fragment [(Htom6-Me)CuII2]3+ binds to two neighboring phosphate diesters of the DNA backbone. Here, we report the hydrolysis of [(Htom6-Me){CuII(OAc)}2](OAc) in phosphate buffer providing the anticipated [(Htom6-Me){CuII(OH2)}2]3+. Moreover, the same reaction in H2O/CH3OH yields the complex [(Htom6-Me){CuII(H2PO4)}2]+. The molecular structures of [(Htom6-Me){CuII(OH2)}2]3+ and [(Htom6-Me){CuII(H2PO4)}2]+ show different coordination modes around the CuII ions demonstrating a significant flexibility of the fragment [(Htom6-Me)CuII2]3+ for binding to exogenous ligands despite its rigid naphthalene backbone. The coupling is weakly antiferromagnetic in [(Htom6-Me){CuII(H2PO4)}2]+, while that in tricationic [(Htom6-Me){CuII(OH2)}2]3+ is weakly ferromagnetic and can be attributed to the orthogonal orientation of the basal planes. UV-vis-NIR spectra of [(Htom6-Me){CuII(H2PO4)}2]+ but also of [(Htom6-Me){CuII(OAc)}2](OAc) in aqueous solution resemble that of [(Htom6-Me){CuII(OH2)}2]3+ demonstrating our earlier assumption that [(Htom6-Me){CuII(OH2)}2]3+ is the active species in buffer solutions for interaction with DNA. Moreover, the exchange of the OAc- ligands by H2PO4- ligands models the anticipated binding of [(Htom6-Me){CuII(OH2)}2]3+ to the phosphates of DNA.
Collapse
Affiliation(s)
- Thomas Riediger
- Lehrstuhl für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, Universitätsstr. 25, Bielefeld D-33615, Germany
| | - Anja Stammler
- Lehrstuhl für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, Universitätsstr. 25, Bielefeld D-33615, Germany
| | - Jan Oldengott
- Lehrstuhl für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, Universitätsstr. 25, Bielefeld D-33615, Germany
| | - Stephan Walleck
- Lehrstuhl für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, Universitätsstr. 25, Bielefeld D-33615, Germany
| | - Thorsten Glaser
- Lehrstuhl für Anorganische Chemie I, Fakultät für Chemie, Universität Bielefeld, Universitätsstr. 25, Bielefeld D-33615, Germany
| |
Collapse
|
5
|
Pisanu F, Sciortino G, Maseras F, Ugone V, Sanna D, Garribba E. Binding of potential antitumor Casiopeínas® to small proteins. Dalton Trans 2025. [PMID: 40260577 DOI: 10.1039/d5dt00228a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
Casiopeínas® are a family of patented CuII anticancer compounds. Cas II-gly and Cas VII-gly are formed by 4,7-dimethyl-1,10-phenanthroline (Me2phen) and 1,10-phenanthroline (phen), respectively, and the bidentate glycinato ligand (Gly), along with a nitrate anion acting as a counterion. In biological fluids, they can maintain their identity or form mixed species and adducts with several bioligands, particularly proteins. In this study, the binding of Cas II-gly, and, for comparison, Cas VII-gly, to small proteins such as myoglobin (Mb), ubiquitin (Ub), and lysozyme (Lyz) was evaluated through a combination of instrumental (ESI-MS and EPR) and computational (dockings) methods. Simulations of the peak signals in the ESI-MS spectra confirmed the formation of the adducts. The results indicated that in all systems, adducts with the formula protein-[CuII(Me2phen)]n (with n = 1-3) were formed after the replacement of glycinato in the equatorial positions by side-chain donors. Docking studies showed that the three proteins used different donor sets to bind the CuII(Me2phen)2+ fragment: (NHis, NHis) or (NHis, COO-Asp/Glu) for Mb; NHis68 or (COO-Glu/Asp, COO-Glu/Asp) for Ub; and (COO-Glu/Asp, CO) or only a monodentate O donor for Lyz. Computational exploration of the protein structure revealed that more than one metal fragment could bind to the macromolecule. At present, it is not clear whether the formation of the adducts improves or worsens the activity of Casiopeínas®. However, the results suggested that, at the low copper concentrations found in the organism, the species protein-[CuII(Me2phen)]n coexist with [CuII(Me2phen)(Gly)]+ and the fragment CuII(Me2phen)2+, which - in turn - could partially dissociate into Cu2+ ions and free Me2phen ligands. Therefore, a mixture of species could be responsible for the biological activity of Casiopeínas®.
Collapse
Affiliation(s)
- Federico Pisanu
- Dipartimento di Medicina, Chirurgia e Farmacia, Università di Sassari, Viale San Pietro, I-07100 Sassari, Italy.
| | - Giuseppe Sciortino
- Departament de Química, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallés, Barcelona, Spain
| | - Feliu Maseras
- Institute of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology (BIST), 43007 Tarragona, Spain
| | - Valeria Ugone
- Istituto CNR di Chimica Biomolecolare, Trav. La Crucca 3, I-07100 Sassari, Italy.
| | - Daniele Sanna
- Istituto CNR di Chimica Biomolecolare, Trav. La Crucca 3, I-07100 Sassari, Italy.
| | - Eugenio Garribba
- Dipartimento di Medicina, Chirurgia e Farmacia, Università di Sassari, Viale San Pietro, I-07100 Sassari, Italy.
| |
Collapse
|
6
|
Li Y, Han L, Hu H. Research progress on cuproptosis and copper related anti-tumor therapy. Discov Oncol 2025; 16:584. [PMID: 40257639 PMCID: PMC12011693 DOI: 10.1007/s12672-025-02335-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 04/08/2025] [Indexed: 04/22/2025] Open
Abstract
Copper is a trace element which is essential for biological organisms, and its homeostatic balance is important for living organisms to maintain the normal function. When the copper homeostasis is disordered, the cellular function and structure will be disrupted. Excess copper cause oxidative stress and DNA damage in cells, thereby inducing regulated cell death such as apoptosis and necroptosis. Excess copper in mitochondria can bind to lipoylated proteins in the tricarboxylic acid (TCA) cycle and cause them to aggregate, resulting in proteotoxic stress and eliciting a novel cell death modality: cuproptosis. Cancer cells have a greater demand for copper compared to normal tissue, and high levels of copper ions are closely associated with tumour proliferation and metastasis. The anti-tumor mechanisms of copper include the production of oxidative stress, inhibition of the ubiquitin-proteasome system, suppression of angiogenesis, and induction of copper-dependent cell death. Targeting copper is one of the current directions in oncology research, including the use of copper ion carriers to increase intracellular copper levels to induce oxidative stress and cuproptosis, as well as the use of copper ion chelators to reduce copper bioavailability. However, copper complexes have certain toxicity, so their biosafety needs to be improved. Emerging nanotechnology is expected to solve this problem by utilizing copper-based nanomaterials (Cu-based NMs) to deliver copper ions and a variety of drugs with different functions, thereby improving the anti-tumor efficacy and reducing the side effects. Therefore, a thorough understanding of copper metabolic processes and the mechanism of cuproptosis will greatly benefit anti-tumor therapy. This review summarizes the processes of copper metabolism and the mechanism of cuproptosis. In addition, we discuss the current anti-tumor paradigms related to copper, we also discuss current nanotherapeutic approaches to copper mortality and provide prospective insights into the future copper-mediated cancer therapy.
Collapse
Affiliation(s)
- Yichen Li
- School of Medicine, Southeast University, No. 87, Dingjiaqiao, Hunan Road, Gulou District, Nanjing, 210009, China
| | - Lifei Han
- Breast Disease Diagnosis and Treatment Center, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, China
| | - Haolin Hu
- Breast Disease Diagnosis and Treatment Center, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, China.
| |
Collapse
|
7
|
Roy I, Mathur S, Deb S, Rathnam SSV, Tuti N, Shaji UP, L K, Roy A, Maji S. Benzimidazole-based mononuclear polypyridyl Cu(II) complexes: DNA binding, cleavage, and in vitro antiproliferative studies. Dalton Trans 2025; 54:6386-6401. [PMID: 40062962 DOI: 10.1039/d4dt03379e] [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: 04/16/2025]
Abstract
This paper addresses the synthesis, characterization, DNA binding, cleavage, and in vitro antiproliferative activity studies of a series of heteroleptic mononuclear copper(II) complexes [Cu(L)(bpy)](ClO4)2, {1}; [Cu(L)(phen)](ClO4)2, {2}; and [Cu(L)(Mephen)](ClO4)2, {3} derived from different polypyridyl ligands, where in the complex architecture, one 2,6-bis(1-methyl-1H-benzo[d]imidazol-2-yl)pyridine(Mebzimpy) (L) moiety is connected to the central Cu metal in a tridentate fashion and the bidentate co-ligands are 2,2'-bipyridine (bpy), 1,10-phenanthroline (phen) and 2,9-dimethyl-1,10-phenanthroline (Mephen). All the synthesized complexes were characterized using various spectroscopic and analytical methods, along with the single-crystal X-ray diffraction (SCXRD) technique. The complexes crystallize in a penta-coordinated distorted square pyramidal geometry. The redox properties of the complexes were also studied by using cyclic voltammetry (CV) and differential pulse voltammetry (DPV). The DNA binding nature of the complexes was investigated utilizing absorbance spectral measurement and fluorescence quenching experiments with ethidium bromide (EB) as a DNA intercalator, employing double-stranded salmon sperm DNA (ss-DNA). Both the binding constant (Kb) and the Stern-Volmer constant (KSV) were found to be in the order of 104. In silico molecular docking analysis confirmed that all the complexes could interact with the minor groove of duplex DNA. The DNA cleaving ability of the complexes was studied by gel electrophoresis using supercoiled plasmid DNA; however, no DNA cleavage was found. DNA-binding polypyridyl complexes are well known to disrupt DNA metabolic pathways and cause cytotoxicity to rapidly growing cancer cells. Hence, cell viability analysis was also carried out with complexes 1-3. It was observed that complexes 2 and 3 prevented the proliferation of the human osteosarcoma cell line U2OS and the triple-negative breast cancer cell line MDA-MB-231. Overall, these findings could be beneficial in the design and development of future antitumor agents.
Collapse
Affiliation(s)
- Indrajit Roy
- Department of Chemistry, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India.
| | - Shobhit Mathur
- Department of Chemistry, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India.
| | - Sourav Deb
- Department of Chemistry, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India.
| | | | - Nikhil Tuti
- Department of Biotechnology, Indian Institute of Technology, Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| | | | - Karthikeyan L
- Department of Chemistry, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India.
| | - Anindya Roy
- Department of Biotechnology, Indian Institute of Technology, Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| | - Somnath Maji
- Department of Chemistry, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India.
| |
Collapse
|
8
|
Gupta S, Shukla A, Pandey SK, Jha S, Zewde B, Acharya A, Butcher RJ, Bharty MK. Piperazine-Based Co(III), Ni(II), Cu(II), and Zn(II) Carbodithioate Complexes as Potential Anticancer Agent. ACS OMEGA 2025; 10:13829-13838. [PMID: 40256499 PMCID: PMC12004137 DOI: 10.1021/acsomega.4c06972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 03/01/2025] [Accepted: 03/11/2025] [Indexed: 04/22/2025]
Abstract
The development of facile and cost-effective anticancer metallodrugs possessing minimal side effects is urgently needed. Piperazine-containing anticancer drugs are already available on the market. A piperazine-based potassium 4-(ethoxycarbonyl)piperazine-1-carbodithioate [pecpcdt] (L) ligand and its metal complexes [Co(ecpcdt)3] (1), [Ni(ecpcdt)2] (2), [Cu(ecpcdt)2] (3), and [Zn(ecpcdt)2] (4) were synthesized. These compounds were characterized by different spectroscopic methods and single-crystal X-ray crystallography data. Ni(II) and Cu(II) complexes have distorted square planar geometry, whereas the Co(III) complex has distorted octahedral geometry around the metal ions. Complexes are weakly fluorescent in the solution compared to the free ligand. The complexes were further examined for their in vitro anticancer activities against the primary Dalton's lymphoma (DL) cells along with standard drug cisplatin. The anticancer studies of metal complexes have been performed through various biochemical assays, and the findings thus obtained suggest that they demonstrate an effective anticancer activity. [Co(ecpcdt)3] (1) shows superior cytotoxicity against DL cells than complexes [Cu(ecpcdt)2] (3), [Zn(ecpcdt)2] (4), and cisplatin. The superiority preferences of these complexes follows [Co(ecpcdt)3] (1) > [pecpcdt] > [Cu(ecpcdt)2] (3) > [Ni(ecpcdt)2] (2) > [Zn(ecpcdt)2] (4). Further assays were performed on a cobalt(III) complex having the highest efficacy to gain insights into the mechanism of cell death and showed that reduced mitochondrial membrane potential and increased mitochondrial ROS production, highlighting mitochondrial-dependent apoptosis as the major mechanism for tumor cell death. On the other hand, the viability of normal splenocytes was minimally affected by the [Co(ecpcdt)3] (1) treatment.
Collapse
Affiliation(s)
- Seema Gupta
- Department
of Chemistry, Banaras Hindu University, Varanasi 221005, India
| | - Alok Shukla
- Department
of Zoology, Banaras Hindu University, Varanasi 221005, India
| | | | - Shalini Jha
- Department
of Chemistry, Banaras Hindu University, Varanasi 221005, India
| | - Berhanu Zewde
- Department
of Chemistry, Howard University, 525 College Street NW, Washington, District of Columbia 20059, United States
| | - Arbind Acharya
- Department
of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Raymond John Butcher
- Department
of Chemistry, Howard University, 525 College Street NW, Washington, District of Columbia 20059, United States
| | - M. K. Bharty
- Department
of Chemistry, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
9
|
Mihajlović E, Biancalana L, Mojić M, Jelača S, Chiaverini L, Zacchini S, Mijatović S, Maksimović-Ivanić D, Marchetti F. Anticancer activity promoted by ligand diversity in diiron thiocarbyne complexes. Eur J Med Chem 2025; 287:117364. [PMID: 39923532 DOI: 10.1016/j.ejmech.2025.117364] [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: 12/03/2024] [Revised: 01/31/2025] [Accepted: 02/02/2025] [Indexed: 02/11/2025]
Abstract
Mononuclear iron (II) complexes have been intensively investigated with the aim of developing efficacious anticancer drugs that can overcome the serious limitations associated with the platinum complexes currently employed in chemotherapy. Combining a promising antitumor potential with appropriate physicochemical properties, such as aqueous stability and a balanced hydrophilic/lipophilic character, is essential for clinical progression. We prepared six highly functionalized diiron(I) complexes from the μ-thiocarbyne precursor [Fe2Cp2(CO)2(μ-CO)(μ-CSMe)]CF3SO3, 1 (Cp = η5-C5H5), through the substitution of one carbonyl ligand with isocyanides (2-4) and the subsequent substitution of a second CO with N- or P-ligands (5-7). All products 2-7 were structurally characterized using IR and multinuclear NMR spectroscopy. One compound from series (7) was also characterized by single crystal X-ray diffraction. Complexes 2-7 exhibit outstanding stability in physiological-like solutions, with 92-97 % of the compounds unchanged after storing in DMEM at 37 °C for 24 h, and substantial amphiphilicity, with most of Log Pow values falling in the range -1 to +1. Complexes 3, 4, 5 and 7 exhibited cytotoxic activity against human (HCT 116, MCF-7, A2780) and murine (CT26, 4T1, B16-F1, B16-F10) cancer cell lines with IC50 values up to the nanomolar range, along with moderate selectivity toward the malignant phenotype. The induction of cell differentiation, senescence, and apoptotic cell death with cell-specific redox response were in the background of cytotoxic activity. However, limited tumor volume reduction and observed systemic toxicity in vivo indicated the need for additional structure-activity relationship studies to optimize the compounds anticancer profile.
Collapse
Affiliation(s)
- Ekatarina Mihajlović
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia
| | - Lorenzo Biancalana
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, I-56124 Pisa, Italy.
| | - Marija Mojić
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia
| | - Sanja Jelača
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia
| | - Lorenzo Chiaverini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, I-56124 Pisa, Italy
| | - Stefano Zacchini
- Department of Industrial Chemistry "Toso Montanari", University of Bologna, Via Piero Gobetti 85, I-40129 Bologna, Italy
| | - Sanja Mijatović
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia
| | - Danijela Maksimović-Ivanić
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11108 Belgrade, Serbia.
| | - Fabio Marchetti
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, I-56124 Pisa, Italy.
| |
Collapse
|
10
|
Carrasco CJ, Pastor A, Conejo MDM, Álvarez E, Calderón-Montaño JM, López-Lázaro M, Galindo A. Synthesis, Characterization, and Preliminary In Vitro Anticancer Activity of Zinc Complexes Containing Amino Acid-Derived Imidazolium-Based Dicarboxylate Ligands. Int J Mol Sci 2025; 26:3202. [PMID: 40244013 PMCID: PMC11989707 DOI: 10.3390/ijms26073202] [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: 03/04/2025] [Revised: 03/27/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
Coordination polymers containing zinc and imidazolium-based dicarboxylate ligands, [LR]-, were synthesized by reacting zinc acetate with HLR compounds, 1. The resulting complexes were characterized and structurally identified using single-crystal X-ray diffraction, revealing polymeric structures for the complexes [Zn(LR)2]n (R = Gly, 2a; βAla, 2b) and [Zn(LLeu)2(H2O)2]n (2c). In these structures, the [LR]- ligands adopt a bridging monodentate μ-κ1-O1,κ1-O3 coordination mode, resulting in distorted tetrahedral (2a, 2b) or octahedral (2c) geometries around the zinc center. When the synthesis was carried out in the presence of amino acids, mixed ligand complexes [Zn(LR)(aa)(H2O)]n (R = aa = Val, 2d, and R = aa = Ile, 2e) were formed. Complexes 2d-2e were also structurally characterized using single-crystal X-ray crystallography, revealing that the ligand [LR]- maintained the same coordination mode, while the zinc center adopted a five-coordinated geometry. The cytotoxic activity of complexes 2a-2e was evaluated against three cancer cell lines and one non-cancerous cell line. Remarkably, these complexes exhibited higher toxicity against cancer cells than against the non-cancerous cell line, and they showed greater selectivity than carboplatin, a commonly used chemotherapy drug. Although, in general, these complexes did not surpass the selectivity of gemcitabine, complex 2c stood out for exhibiting a selectivity index value similar to that of gemcitabine against melanoma cells. Among the series, compounds 2a-2c demonstrated the highest activity, with 2a being the only complex with some selective activity against lung cancer. Complex 2b was the most active, though with low selectivity, while complex 2c exhibited the highest selectivity for melanoma and bladder cancer (selectivity index of 3.0).
Collapse
Affiliation(s)
- Carlos J. Carrasco
- Departamento de Química Inorgánica, Facultad de Química, Universidad de Sevilla, 41071 Sevilla, Spain; (C.J.C.); (A.P.); (M.d.M.C.)
| | - Antonio Pastor
- Departamento de Química Inorgánica, Facultad de Química, Universidad de Sevilla, 41071 Sevilla, Spain; (C.J.C.); (A.P.); (M.d.M.C.)
| | - María del Mar Conejo
- Departamento de Química Inorgánica, Facultad de Química, Universidad de Sevilla, 41071 Sevilla, Spain; (C.J.C.); (A.P.); (M.d.M.C.)
| | - Eleuterio Álvarez
- Instituto de Investigaciones Químicas, CSIC-Universidad de Sevilla, Avda. Américo Vespucio 49, 41092 Sevilla, Spain;
| | - José Manuel Calderón-Montaño
- Departamento de Farmacología, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; (J.M.C.-M.); (M.L.-L.)
| | - Miguel López-Lázaro
- Departamento de Farmacología, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; (J.M.C.-M.); (M.L.-L.)
| | - Agustín Galindo
- Departamento de Química Inorgánica, Facultad de Química, Universidad de Sevilla, 41071 Sevilla, Spain; (C.J.C.); (A.P.); (M.d.M.C.)
| |
Collapse
|
11
|
Boga S, Bouzada D, Lopez-Blanco R, Sarmiento A, Salvadó I, Alvar Gil D, Brea J, Loza MI, Barreiro-Piñeiro N, Martínez-Costas J, Mena S, Guirado G, Santoro A, Faller P, Vázquez ME, Vázquez López M. Copper(II) Cyclopeptides with High ROS-Mediated Cytotoxicity. Bioconjug Chem 2025; 36:500-509. [PMID: 40059798 DOI: 10.1021/acs.bioconjchem.4c00561] [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: 03/20/2025]
Abstract
Cu(II) coordination complexes are emerging as promising anticancer agents due to their ability to induce oxidative stress through reactive oxygen species (ROS) generation. In this study, we synthesized and characterized two novel Cu(II) metallopeptide systems, 1/Cu(II) and 2/Cu(II), derived from the oligocationic bipyridyl cyclopeptides 1 and 2, and designed to enhance the transport of Cu(II) into cells and increase ROS levels. Spectroscopic and electrochemical analyses confirmed the formation of stable metallopeptide species in aqueous media. Inductively coupled plasma mass spectrometry (ICP-MS) studies demonstrated that both metallopeptides significantly increase intracellular Cu(II) accumulation in NCI/ADR-RES cancer cells, highlighting their role as efficient Cu(II) transporters. Additionally, ROS generation assays revealed that 1/Cu(II) induces a substantial increase in intracellular ROS levels, supporting the hypothesis of oxidative stress-induced cytotoxicity. Cell-viability assays further confirmed that both 1/Cu(II) and 2/Cu(II) exhibit strong anticancer activity in a number of cancer cell lines, with IC50 values significantly lower than those of their free cyclopeptide counterparts or Cu(II) alone, showing an order of activity higher than that of cisplatin. Finally, molecular modeling studies provided further insights into the structural stability and coordination environment of Cu(II) within the metallopeptide complexes. These findings suggest that these Cu(II) cyclometallopeptide systems hold potential as novel metal-based therapeutic agents, leveraging Cu(II) transport and ROS increase as key strategies for cancer treatment.
Collapse
Affiliation(s)
- Sonia Boga
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - David Bouzada
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Roi Lopez-Blanco
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Axel Sarmiento
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Inorgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Iria Salvadó
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Inorgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - David Alvar Gil
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Inorgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - José Brea
- Innopharma Drug Screening and Pharmacogenomics Platform. Center for Research in Molecular Medicine and Chronic Diseases (CiMUS). Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - María Isabel Loza
- Innopharma Drug Screening and Pharmacogenomics Platform. Center for Research in Molecular Medicine and Chronic Diseases (CiMUS). Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Natalia Barreiro-Piñeiro
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Bioquímica e Bioloxía Molecular, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - José Martínez-Costas
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Bioquímica e Bioloxía Molecular, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Silvia Mena
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Gonzalo Guirado
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Alice Santoro
- Institut de Chimie (UMR 7177), University of Strasbourg─CNRS, 67081 Strasbourg, France
| | - Peter Faller
- Institut de Chimie (UMR 7177), University of Strasbourg─CNRS, 67081 Strasbourg, France
- Institut Universitaire de France (IUF), 75231 Paris, France
| | - M Eugenio Vázquez
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Miguel Vázquez López
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Inorgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
12
|
O'Sullivan E, O'Shea D, Devereux M, Howe O. Apoptotic cell death induced by copper (II), manganese (II) and silver (I) complexes containing bridging dicarboxylate and 1,10-phenanthroline ligands: one of the multi-modes of anticancer activity? Biometals 2025:10.1007/s10534-025-00676-8. [PMID: 40095185 DOI: 10.1007/s10534-025-00676-8] [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: 11/18/2024] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
Cu(II), Mn(II) and Ag(I) complexes incorporating bridging dicarboxylate and 1,10-phenanthroline ligands have exhibited anti-cancer potential with significant in vitro and in vivo cytotoxic efficacies. Our study focuses on regulated cell death process of apoptosis as a mode of action of the anti-cancer activity by the complexes. Cytotoxicity screening of the complexes demonstrated all the metal-dicarboxylate-phenanthroline complexes exhibit superior activity compared to their non-phenanthroline containing precursors, in addition to cisplatin. The Cu(II) and Mn(II) complexes were shown to induce reactive oxygen species (ROS) but this was not observed for the Ag(I) analogues. Furthermore, apoptosis was found to be induced by all the metal-phenanthroline complexes to varying degrees contingent on the type of metal centre in the complex. Apoptotic gene expression analysis established the predominant activation of the intrinsic apoptotic pathway, with co-stimulation of the extrinsic pathway observed in some cases. The mechanistic data provided within this study highlights the multi-modal activity of the metal-phenanthroline complexes contingent on the type of metal present, warranting continued investigation of their biological modes of action beyond apoptosis induction.
Collapse
Affiliation(s)
- Ella O'Sullivan
- School of Biological, Health and Sports Sciences, Technological University Dublin, Dublin, Ireland.
- Sustainability and Health Research Hub, Technological University Dublin, Dublin, Ireland.
| | - Denis O'Shea
- School of Food Science and Environmental Health, Technological University Dublin, Dublin, Ireland
| | - Michael Devereux
- The Physical to Life Sciences Research Hub, Technological University Dublin, Dublin, Ireland
| | - Orla Howe
- School of Biological, Health and Sports Sciences, Technological University Dublin, Dublin, Ireland
- The Physical to Life Sciences Research Hub, Technological University Dublin, Dublin, Ireland
| |
Collapse
|
13
|
Stocchetti S, Vančo J, Bresciani G, Biancalana L, Belza J, Zacchini S, Dvořák Z, Benetti S, Biver T, Bortoluzzi M, Trávníček Z, Marchetti F. Anticancer diiron aminocarbyne complexes with labile N-donor ligands. Eur J Med Chem 2025; 286:117304. [PMID: 39862748 DOI: 10.1016/j.ejmech.2025.117304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/14/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
The novel diiron amine complexes [Fe2Cp2(CO)(NH2R')(μ-CO){μ-CN(Me)(Cy)}]CF3SO3 [R' = H, 3; Cy, 4; CH2CH2NH2, 5; CH2CH2NMe2, 6; CH2CH2(4-C6H4OMe), 7; CH2CH2(4-C6H4OH), 8; Cp = η5-C5H5, Cy = C6H11 = cyclohexyl] were synthesized in 49-92 % yields from [Fe2Cp2(CO)2(μ-CO){μ-CN(Me)(Cy)}]CF3SO3, 1a, using a straightforward two-step procedure. They were characterized by IR and multinuclear NMR spectroscopy, and the structure of 7 was confirmed through X-ray diffraction analysis. Complexes 3-8 and the acetonitrile adducts [Fe2Cp2(CO)(NCMe)(μ-CO){μ-CN(Me)(R)}]CF3SO3 (R = Cy, 2a; Me, 2b; Xyl = 2,6-C6H3Me2, 2c) were assessed for their water solubility, octanol-water partition coefficient and stability in physiological-like solutions. The in vitro antiproliferative activity of 2a-c and 3-8 was tested on seven human cancer cell lines (A2780, A2780R, PC3, A549, MCF7, HOS and HT-29), while the selectivity was evaluated using normal MRC-5 cells. Overall, the complexes exhibited variable cytotoxicity, with IC50 values reaching the low micromolar range for 3, 7 and 8 in A2780 and A2780R cells, along with significant selectivity. Targeted experiments covered cell cycle modification, induction of cell death, mitochondrial membrane potential, ROS production and interaction with DNA and bovine serum albumin (BSA) as a model protein. The interaction of 3 with BSA was further investigated through computational studies. Results showed a negligible increase in intracellular ROS levels (except for 2b) and insignificant changes in mitochondrial membrane potential.
Collapse
Affiliation(s)
- Sara Stocchetti
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124, Pisa, Italy
| | - Ján Vančo
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University, Šlechtitelů 27, CZ-779 00, Olomouc, Czech Republic
| | - Giulio Bresciani
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124, Pisa, Italy
| | - Lorenzo Biancalana
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124, Pisa, Italy
| | - Jan Belza
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University, Šlechtitelů 27, CZ-779 00, Olomouc, Czech Republic
| | - Stefano Zacchini
- University of Bologna, Department of Industrial Chemistry "Toso Montanari", I-40136, Bologna, Italy
| | - Zdeněk Dvořák
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, CZ-779 00, Olomouc, Czech Republic
| | - Sara Benetti
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124, Pisa, Italy
| | - Tarita Biver
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124, Pisa, Italy
| | - Marco Bortoluzzi
- University of Venezia "Ca' Foscari", Department of Molecular Science and Nanosystems, Via Torino 155, I-30170, Mestre (VE), Italy
| | - Zdeněk Trávníček
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University, Šlechtitelů 27, CZ-779 00, Olomouc, Czech Republic.
| | - Fabio Marchetti
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124, Pisa, Italy.
| |
Collapse
|
14
|
Balewski Ł, Inkielewicz-Stępniak I, Gdaniec M, Turecka K, Hering A, Ordyszewska A, Kornicka A. Synthesis, Structure, and Stability of Copper(II) Complexes Containing Imidazoline-Phthalazine Ligands with Potential Anticancer Activity. Pharmaceuticals (Basel) 2025; 18:375. [PMID: 40143151 PMCID: PMC11946467 DOI: 10.3390/ph18030375] [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: 01/30/2025] [Revised: 02/27/2025] [Accepted: 03/02/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Recently, there has been great interest in metallopharmaceuticals as potential anticancer agents. In this context, presented studies aim to synthesize and evaluate of two copper(II) complexes derived from phthalazine- and imidazoline-based ligands against on three human cancer cell lines: cervix epithelial cell line (HeLa), breast epithelial-like adenocarcinoma (MCF-7), and triple-negative breast epithelial cancer cell line (MDA-MB-231), as well as non-tumorigenic cell line (HDFa). Moreover their antimicrobial, and antioxidant properties were assessed. Methods: The synthetized compounds-both free ligands L1, L2, L3 and copper(II) complexes C1 and C2-were characterized by elemental analysis, infrared spectroscopy. Additionally, a single-crystal X-ray diffraction studies we performed for free ligand L3 and its copper(II) complex C2. The stability of Cu(II)-complexes C1 and C2 was evaluated by UV-Vis spectroscopy. The cytotoxic potency of free ligands and their copper(II) complexes was estimated on HeLa, MCF-7, MDA-MB-231, as well as non-cancerous HDFa by use of an MTT assay after 48 h of incubation. Moreover, the antimicrobial activity of ligands L1 and L3 and their copper(II) complexes C1 and C2 was evaluated using reference strains of the following bacteria and yeasts: Staphylococcus aureus, Escherichia coli, and Candida albicans. The free radical scavenging properties of free ligands L1, L3 and the corresponding copper(II) complexes C1, C2 was tested with two colorimetric methods-ABTS, DPPH, and reduction ability assay (FRAP). Additionally, the ADME webtool was used to assess the drug-likeness of the synthesized compounds, as well as their physicochemical and pharmacokinetic properties. Results: Copper(II) complex C2 exhibited antitumor properties towards MDA-MB-231 compared with Cisplatin (cancer cell viability rate of 23.6% vs. 22.5%). At a concentration of 200 μg/mL, complexes C1 and C2 were less cytotoxic than the reference Cisplatin against a normal, non-cancerous skin fibroblast cell line (HDFa). According to in vitro tests, C2 reduced the viability of HeLa, MCF-7, and MDA-MB-231 cells by about 57.5-81.2%. It was evident that all compounds were devoid of antibacterial or antifungal activity. In vitro assays revealed that a moderate antiradical effect was observed for free ligand L1 containing phthalazin-1(2H)-imine in the ABTS radical scavenging assay (IC50 = 23.63 µg/mL). Conclusions: The anticancer studies revealed that the most potent compound was copper(II) complex C2 bearing a phthalazin-1(2H)-one scaffold. None of the tested compounds showed antimicrobial or antifungal activity. This feature seems to be beneficial in terms of their potential uses as anticancer agents in the future. In vitro antiradical assays revealed that a moderate antioxidant effect was observed only for free ligand L1 containing phthalazin-1(2H)-imine.
Collapse
Affiliation(s)
- Łukasz Balewski
- Department of Chemical Technology of Drugs, Faculty of Pharmacy, Medical University of Gdansk, Gen. J. Hallera 107, 80-416 Gdańsk, Poland
| | - Iwona Inkielewicz-Stępniak
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, Gen. Dębinki 7, 80-211 Gdańsk, Poland
| | - Maria Gdaniec
- Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland
| | - Katarzyna Turecka
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Medical University of Gdansk, Gen. J. Hallera 107, 80-416 Gdańsk, Poland
| | - Anna Hering
- Department of Biology and Pharmaceutical Botany, Faculty of Pharmacy, Medical University of Gdansk, Gen. J. Hallera 107, 80-416 Gdańsk, Poland
| | - Anna Ordyszewska
- Department of Inorganic Chemistry, Faculty of Chemistry and Advanced Materials Centers, Gdańsk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
| | - Anita Kornicka
- Department of Chemical Technology of Drugs, Faculty of Pharmacy, Medical University of Gdansk, Gen. J. Hallera 107, 80-416 Gdańsk, Poland
| |
Collapse
|
15
|
Kazemi Z, Moini N, Rudbari HA, Micale N. A comprehensive review on the development of chiral Cu, Ni, and Zn complexes as pharmaceutical agents over the past decades: Synthesis, molecular structure and biological activity. Med Res Rev 2025; 45:654-754. [PMID: 39297288 DOI: 10.1002/med.22083] [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: 08/26/2022] [Revised: 07/09/2024] [Accepted: 08/25/2024] [Indexed: 02/06/2025]
Abstract
Chirality is a fundamental and widespread geometric structural property in living organisms that most biomacromolecules including nucleic acids, proteins and enzymes, possess. Consequently, the development of chiral drugs capable of binding specific targets have gradually gained wide attention in recent decades due to their selective effects on a broad spectrum of biological events ranging from cell metabolism to cell fate. In this context, the synthesis of chiral compounds as promising therapeutic candidates has assumed a major role in drug discovery. Among them, chiral metal complexes have attracted considerable interest due to their unique and intriguing structural features that could enable overcoming side effects and drug-resistance phenomena of metal-based drugs currently in the market such as cisplatin. In the current scenario, an in-depth overview of non-platinum chiral complexes needs to be presented and carried forward. Therefore, in this perspective article, an update of the scientific development of bioactive chiral copper, zinc and nickel complexes have been reported since they have not been thoroughly reviewed so far. Specifically, we focused the article mainly on metal complexes containing chiral ligands (type 2 chirality) as in literature they are more numerous than those with chirality at the metal center (type 1 chirality). Herein, not only their biological activity but also their mechanism of action is summarized. Furthermore, in the final section of the article we have highlighted copper-based complexes as those with a superior biological activity profile and greater prospects for development as a drug.
Collapse
Affiliation(s)
- Zahra Kazemi
- Department of Chemistry, University of Isfahan, Isfahan, Iran
| | - Nakisa Moini
- Department of Inorganic Chemistry, Faculty of Chemistry, Alzahra University, Tehran, Iran
| | | | - Nicola Micale
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
16
|
Jhetam Z, Martins-Furness C, Slabber C, Munro OQ, Nel M, Harmse L. Copper complexes induce haem oxygenase-1 (HMOX1) and cause apoptotic cell death in pancreatic cancer cells. J Inorg Biochem 2025; 264:112815. [PMID: 39740375 DOI: 10.1016/j.jinorgbio.2024.112815] [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: 07/27/2024] [Revised: 12/14/2024] [Accepted: 12/15/2024] [Indexed: 01/02/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common pancreatic malignancy, has a dismal 5-year survival rate, making palliative chemotherapy the only treatment option. Targeted therapy has limited efficacy in PDAC, underscoring the need for novel therapeutic approaches. The inducible stress-response protein, haem oxygenase-1 (HMOX1), has been implicated in treatment failure in PDAC. Copper coordination complexes have shown promise as anticancer agents against various cancers, and are associated with apoptotic cell death. The different ligands to which copper is complexed, determine the specificity and efficacy of each complex. Three different classes of copper complexes were evaluated for anti-cancer activity against AsPC-1 and MIA PaCa-2 pancreatic cancer cell lines. A copper-phenanthroline-theophylline complex (CuPhTh2), a copper-8-aminoquinoline-naphthyl complex (Cu8AqN), and two copper-aromatic-isoindoline complexes (CuAIsI) were effective inhibitors of cell proliferation with clinically relevant IC50 values below 5 μM. The copper complexes caused reactive oxygen species (ROS) formation, promoted annexin-V binding, disrupted the mitochondrial membrane potential (MMP) and activated caspase-9 and caspase-3/7, confirming apoptotic cell death. Expression of nuclear HMOX1 was increased in both cell lines, with the CuPhTh2 complex being the most active. Inhibition of HMOX1 activity significantly decreased the IC50 values of these copper complexes suggesting that HMOX1 inhibition may alter treatment outcomes in PDAC.
Collapse
Affiliation(s)
- Zakeeya Jhetam
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, South Africa
| | - Carla Martins-Furness
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, South Africa
| | - Cathy Slabber
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smut Ave, Braamfontein, Johannesburg 2017, South Africa
| | - Orde Q Munro
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smut Ave, Braamfontein, Johannesburg 2017, South Africa; School of Chemistry, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, UK
| | - Marietha Nel
- Dept of Surgery, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, South Africa
| | - Leonie Harmse
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, South Africa.
| |
Collapse
|
17
|
Zhao P, Zhang X, Dong J, Li L, Meng X, Gao L. In vitro study of the pro-apoptotic mechanism of amino acid Schiff base copper complexes on anaplastic thyroid cancer. Eur J Pharm Sci 2025; 206:107005. [PMID: 39799912 DOI: 10.1016/j.ejps.2025.107005] [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: 09/26/2024] [Revised: 12/19/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
In the endocrine system, anaplastic thyroid cancer (ATC) is extremely aggressive since it inhibits the majority of medications and treatments. Therefore, there is an immediate demand to identify new treatment approaches or drugs to deal with ATC. Recently, amino acid Schiff base copper complexes have received great attention due to their excellent anti-tumor activity. In this research, three copper(II) complexes, [Cu(o-van-D-Trp)(phen)](1), [Cu(o-van-D-Trp)(bipy)](2), [Cu(naph-D-Trp)(bipy)](3), [D-Trp = D-tryptophan; o-van = o-vanillin; naph = 2‑hydroxy-1- naphthaldehyde; phen = 1,10-phenanthroline; bipy = 2,2-biprydine], have been synthesized and investigated as potential anticancer agents. The crystal structure data of the complexes demonstrate that the central copper (II) atom forms a twisted polyhedral environment with nitrogen and oxygen atoms. The MTT results demonstrated that three complexes exhibited superior cytotoxicity against five cell lines of thyroid cancer (Cal-62 cells, ARO cells, KHM-5 m cells, BHP10-3 cells and K1 cells), especially complex 1 with the IC50 values of 0.59±0.05 μM, 2.36±0.47 μM, 1.10±0.87 μM, 0.75±0.09 μM, 1.72±0.06 μM, when cisplatin was used as a control. Research on antitumor mechanisms has demonstrated that complex 1 can significantly reduce the mitochondrial membrane potential, raise autophagy, and produce reactive oxygen species (ROS) in ARO cells in a dose-dependent manner. RNA sequencing study reveals that complex 1 may cause apoptosis in ARO cells and exhibit anticancer efficacy in vitro through ROS-mediated downregulation of Akt and p38 MAPK activation.
Collapse
Affiliation(s)
- Peiran Zhao
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng 252000, PR China
| | - Xinyan Zhang
- Department of Pathology, Liaocheng People's Hospital, Liaocheng 252000, PR China
| | - Jianfang Dong
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng 252059, PR China; Department of Information Engineering, Shandong Polytechnic Technician College, Liaocheng 252000, PR China
| | - Lianzhi Li
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng 252059, PR China
| | - Xiao Meng
- Department of Pathology, Liaocheng People's Hospital, Liaocheng 252000, PR China.
| | - Lei Gao
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng 252000, PR China.
| |
Collapse
|
18
|
Khan S, Tauqeer M, Arjmand F, Tabassum S. Synthesis and structure elucidation of tailored metal-based intercalative agents derived from anthraldehyde & L-valine that show selective inhibition against triple-negative resistant breast cancer cells. Polyhedron 2025; 269:117398. [DOI: 10.1016/j.poly.2025.117398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
|
19
|
Espindola-Moreno O, da Silva Moura F, Santa Maria de la Parra L, Stellet C, Serna JDP, Diniz R, Rey NA, León IE. Antiproliferative activity of a series of copper(II) complexes derived from a furan-containing N-acylhydrazone: monomers, dimers, charge status, and cell mechanistic studies on triple negative breast cancer cells. Dalton Trans 2025; 54:3872-3886. [PMID: 39883470 DOI: 10.1039/d4dt03445g] [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: 01/31/2025]
Abstract
In this work, we evaluated the anticancer activity of compounds 1 (mononuclear) and 2 (dinuclear) copper(II) coordination compounds derived from the ligand 5-methylsalicylaldehyde 2-furoyl hydrazone (H2L) over MDA-MB-231 Triple-negative breast cancer (TNBC) cells, and compared their activities with that of a newly synthesized, protonated, dinuclear analogue of 2 (complex 3). Here, we report the synthesis of compound 3 and it has been characterized in the solid state (X-ray diffraction, FTIR) and in solution (EPR, UV-Vis, ESI) as well as its electrochemical profile. Complexes 1-3 impaired cell viability from 0.5 to 2.5 μM, with IC50 values around 1.25 μM for complexes 2 and 3 and a slightly higher value of 2.0 μM for the monomer 1. It is important to highlight that the three compounds are more active than cisplatin (CDDP), by a factor of 100 in the case of 2 and 3. Our results indicate that the protonation status of the amide group in H2L plays an important role in the stability of the dimer, being compound 2 (amide-deprotonated) fairly stable in solution so we decided to continue the study of mechanism of action using this compound. Complex 2 increases the ROS production and induces cell programmed death on TNBC cells at very low micromolar concentrations (0.5-1.5 μM). Moreover, the compound decreased the amount of breast CSCs on MDA-MB-231 cells reducing the percentage of CD44+/CD24-/low cells at 1 and 1.5 μM.
Collapse
Affiliation(s)
- Olivia Espindola-Moreno
- CEQUINOR (UNLP, CCT-CONICET La Plata, asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 No. 1465, La Plata (1900), Argentina.
| | - Fagner da Silva Moura
- LABSO-Bio (CTC, PUC-Rio), Departamento de Química, Pontifícia Universade Católica do Rio de Janeiro, Rua Marquês de São Vicente, 225 - Gávea, Rio de Janeiro (22453-900), RJ, Brasil.
| | - Lucía Santa Maria de la Parra
- CEQUINOR (UNLP, CCT-CONICET La Plata, asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 No. 1465, La Plata (1900), Argentina.
| | - Carolina Stellet
- LABSO-Bio (CTC, PUC-Rio), Departamento de Química, Pontifícia Universade Católica do Rio de Janeiro, Rua Marquês de São Vicente, 225 - Gávea, Rio de Janeiro (22453-900), RJ, Brasil.
| | - Jilder D P Serna
- Centro Brasileiro de Pesquisas Físicas, Rua Dr. Xavier Sigaud, 150 - Botafogo, Rio de Janeiro (22290-180), RJ, Brasil
| | - Renata Diniz
- Grupo de Cristalografia Química (ICEx, UFMG), Departamento de Química, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627 - Pampulha, Belo Horizonte (31270-901), MG, Brasil
| | - Nicolás A Rey
- LABSO-Bio (CTC, PUC-Rio), Departamento de Química, Pontifícia Universade Católica do Rio de Janeiro, Rua Marquês de São Vicente, 225 - Gávea, Rio de Janeiro (22453-900), RJ, Brasil.
| | - Ignacio E León
- CEQUINOR (UNLP, CCT-CONICET La Plata, asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 No. 1465, La Plata (1900), Argentina.
- Cátedra de Fisiopatología, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, La Plata 1900, Argentina
| |
Collapse
|
20
|
Pellei M, Santini C, Caviglia M, Del Gobbo J, Battocchio C, Meneghini C, Amatori S, Donati C, Zampieri E, Gandin V, Marzano C. Anticancer potential of copper(i) complexes based on isopropyl ester derivatives of bis(pyrazol-1-yl)acetate ligands. RSC Med Chem 2025; 16:849-861. [PMID: 39618961 PMCID: PMC11605304 DOI: 10.1039/d4md00610k] [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: 08/07/2024] [Accepted: 11/07/2024] [Indexed: 02/21/2025] Open
Abstract
In this paper, the isopropyl ester derivatives LOiPr and L2OiPr of bis(pyrazol-1-yl)acetic acid and bis(3,5-dimethyl-pyrazol-1-yl)acetic acid were used as chelators for the preparation of new Cu(i) phosphane complexes 1-4. They were synthesized by the reaction of [Cu(CH3CN)4]PF6 and triphenylphosphine or 1,3,5-triaza-7-phosphaadamantane with LOiPr and L2OiPr ligands, in acetonitrile or acetonitrile/methanol solution. The authenticity of the compounds was confirmed by CHN analysis, 1H-, 13C- and 31P-NMR, FT-IR spectroscopy, and electrospray ionization mass spectrometry (ESI-MS). Furthermore, the electronic and molecular structures of the selected Cu(i) coordination compound 3 were investigated by synchrotron radiation-induced X-ray photoelectron spectroscopy (SR-XPS), and the local structure around the copper ion site was studied combining X-ray absorption fine structure (XAFS) spectroscopy techniques and DFT modelling. Triphenylphosphine as a coligand confers to [Cu(LOiPr)(PPh3)]PF6 (1) and [Cu(L2OiPr)(PPh3)]PF6 (3) a significant antitumor activity in 3D spheroidal models of human colon cancer cells. Investigations focused on the mechanism of action evidenced protein disulfide-isomerase (PDI) as an innovative molecular target for this class of phosphane copper(i) complexes. By hampering PDI activity, copper(i) complexes were able to cause an imbalance in cancer cell redox homeostasis thus leading to cancer cell death - a non-apoptotic programmed cell death.
Collapse
Affiliation(s)
- Maura Pellei
- School of Science and Technology, Chemistry Division, University of Camerino Via Madonna delle Carceri (ChIP) 62032 Camerino Italy
| | - Carlo Santini
- School of Science and Technology, Chemistry Division, University of Camerino Via Madonna delle Carceri (ChIP) 62032 Camerino Italy
| | - Miriam Caviglia
- School of Science and Technology, Chemistry Division, University of Camerino Via Madonna delle Carceri (ChIP) 62032 Camerino Italy
| | - Jo' Del Gobbo
- School of Science and Technology, Chemistry Division, University of Camerino Via Madonna delle Carceri (ChIP) 62032 Camerino Italy
| | - Chiara Battocchio
- Department of Science, Roma Tre University Via della Vasca Navale 79 00146 Roma Italy
| | - Carlo Meneghini
- Department of Science, Roma Tre University Via della Vasca Navale 79 00146 Roma Italy
| | - Simone Amatori
- Department of Science, Roma Tre University Via della Vasca Navale 79 00146 Roma Italy
| | - Chiara Donati
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova Via Marzolo 5 35131 Padova Italy
| | - Eleonora Zampieri
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova Via Marzolo 5 35131 Padova Italy
| | - Valentina Gandin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova Via Marzolo 5 35131 Padova Italy
| | - Cristina Marzano
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova Via Marzolo 5 35131 Padova Italy
| |
Collapse
|
21
|
Argirova M, Cherneva E, Mihaylova R, Momekov G, Yancheva D. New metal complexes of 1H-benzimidazole-2-yl hydrazones: Cytostatic, proapoptotic and modulatory activity on kinase signaling pathways. Arch Biochem Biophys 2025; 764:110245. [PMID: 39617120 DOI: 10.1016/j.abb.2024.110245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
The copper complexes of two 1H-benzimidazole-2-yl hydrazones were obtained by complexation with copper chloride. The molecular structure of the complexes was studied by microchemical analysis, SEM-EDX, IR and micro-Raman spectroscopy and DFT calculations. It was found that both ligands form 1:1 complexes with the copper, where the Cu ions are coordinated by N-atom from the benzimidazole ring, N-atom of the azomethine bond, O-atom from the ortho-OH group of the aromatic ring and one chlorine atom. The coordination process significantly affected their cytotoxicity profile. The conversion of 2-(2-hydroxybenzylidene)-1-(1H-benzimidazol-2-yl)hydrazine 1.1. into a Cu complex 2.1. led to a 2.4-fold increase in its antileukemic activity against AR-230 cells and an 8-fold increase in the cytostatic activity against MCF-7 breast cancer cell line. The growth-inhibitory effect of the Cu complex of 2-(2-hydroxy-4-methoxybenzylidene)-1-(1H-benzimidazol-2-yl)hydrazine 2.2. on the MCF-7 cells was comparable to that of the respective ligand, however lacked towards the leukemic AR-230 cell population. Regarding their cytotoxic potential towards CCL-1 cells, both Cu complexes exhibited a weaker selectivity pattern as compared to their ligands. The proapoptotic and modulatory activity of 1.1 and 2.1. on key kinase signaling pathways was further studied in the ER + breast cancer (MCF-7) and bcr-abl + leukemic (AR-230) in vitro tumor models in a comparative manner to the reference drugs tamoxifen and imatinib, respectively. Inhibition of the JAK/STAT signaling pathway was outlined as a prominent mechanism in the antileukemic activity against the Ph + AR-230 in vitro model, whereas recruitment and activation of the extrinsic apoptotic pathway was established in the MCF-7 cells.
Collapse
Affiliation(s)
- Maria Argirova
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., build. 9, 1113, Sofia, Bulgaria
| | - Emiliya Cherneva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., build. 9, 1113, Sofia, Bulgaria; Faculty of Pharmacy, Medical University of Sofia, 2 Dunav str., 1000, Sofia, Bulgaria.
| | - Rositsa Mihaylova
- Faculty of Pharmacy, Medical University of Sofia, 2 Dunav str., 1000, Sofia, Bulgaria
| | - Georgi Momekov
- Faculty of Pharmacy, Medical University of Sofia, 2 Dunav str., 1000, Sofia, Bulgaria
| | - Denitsa Yancheva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., build. 9, 1113, Sofia, Bulgaria; University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756, Sofia, Bulgaria.
| |
Collapse
|
22
|
Rossi A, Biancalana L, Vančo J, Malina T, Zacchini S, Dvořák Z, Trávníček Z, Marchetti F. The effect of a varying pyridine ligand on the anticancer activity of Diiron(I) bis-cyclopentadienyl complexes. Chem Biol Interact 2025; 406:111318. [PMID: 39603517 DOI: 10.1016/j.cbi.2024.111318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/16/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024]
Abstract
The new diiron complexes [Fe2Cp2(CO)(L)(μ-CO){μ-CN(Me)(Cy)}]CF3SO3 (L = pyridine, 3a; 4-aminopyridine, 3b; 4-dimethylaminopyridine, 3c; 4-trifluoromethylpyridine, 3d; nicotinic acid, 4; Cp = η5-C5H5, Cy = C6H11 = cyclohexyl) were synthesized in moderate to high yields using two distinct synthetic routes from the precursors 1 (L = CO, for 4) and 2 (L = NCMe, for 3a-d), respectively. All products were characterized by IR and multinuclear NMR spectroscopy, and the structures of 3b and 3d were ascertained by X-ray diffraction studies. The behavior of the complexes in aqueous solutions (solubility, Log Pow, stability) was assessed using NMR and UV-Vis methods. The in vitro antiproliferative activity of 3a-c and 4 was evaluated against seven human cancer cell lines (A2780, A2780R, A549, MCF-7, PC3, HOS and HT-29) and one normal cell line (MRC-5), following 24 h of incubation (MTT test). Overall, 3-4 demonstrated stronger cytotoxicity than cisplatin, with 3c emerging as the most potent compound. The activity seems primarily linked to the inhibition of metabolic processes in the cancer cells, including depletion of reactive oxygen species (ROS) levels. However, subtle differences have been observed between the complexes, with 4 exerting its cytotoxicity through a distinct multimodal mechanism.
Collapse
Affiliation(s)
- Annachiara Rossi
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via Giuseppe Moruzzi 13, I-56124, Pisa, Italy
| | - Lorenzo Biancalana
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via Giuseppe Moruzzi 13, I-56124, Pisa, Italy
| | - Ján Vančo
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University, Šlechtitelů 27, CZ-779 00, Olomouc, Czech Republic
| | - Tomáš Malina
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University, Šlechtitelů 27, CZ-779 00, Olomouc, Czech Republic; Nanotechnology Centre, Centre for Energy and Environmental Technologies, VŠB-Technical University of Ostrava, 17. listopadu 2172/15, CZ-708 00, Ostrava, Poruba, Czech Republic
| | - Stefano Zacchini
- University of Bologna, Dipartimento di Chimica Industriale "Toso Montanari", Via Piero Gobetti 85, I-40129, Bologna, Italy
| | - Zdeněk Dvořák
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, CZ-779 00, Olomouc, Czech Republic
| | - Zdeněk Trávníček
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University, Šlechtitelů 27, CZ-779 00, Olomouc, Czech Republic.
| | - Fabio Marchetti
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via Giuseppe Moruzzi 13, I-56124, Pisa, Italy.
| |
Collapse
|
23
|
Pastuch-Gawołek G, Szreder J. Effect of Glycoconjugation on Cytotoxicity and Selectivity of 8-Aminoquinoline Derivatives Compared to 8-Hydroxyquinoline. Molecules 2025; 30:427. [PMID: 39860296 PMCID: PMC11767929 DOI: 10.3390/molecules30020427] [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: 12/21/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Numerous emerging chemotherapeutic agents incorporate N-heterocyclic fragments in their structures, with the quinoline skeleton being particularly significant. Our recent works have focused on glycoconjugates of 8-hydroxyquinoline (8-HQ), which demonstrated enhanced bioavailability and solubility compared to their parent compounds, although they fell short in selectivity. In this study, our objective was to improve the selectivity of glycoconjugates by replacing the oxygen atom with nitrogen by substituting the 8-HQ moiety with 8-aminoquinoline (8-AQ). The 8-AQ derivatives were functionalized through the amino group and linked to sugar derivatives (D-glucose or D-galactose) that were modified with an azide, alkylazide, or propargyl group at the anomeric position by copper(I)-catalyzed 1,3-dipolar azido-alkyne cycloaddition (CuAAC). The resulting glycoconjugates, as well as their potential metabolites, were evaluated for their ability to inhibit the proliferation of cancer cell lines (including HCT 116 and MCF-7) and a healthy cell line (NHDF-Neo). Two of the synthesized glycoconjugates (17 and 18) demonstrated higher cytotoxicity than their oxygen-containing counterparts and showed improved selectivity for cancer cells, thus enhancing their anticancer potential. Furthermore, it was found that glycoconjugates exhibited greater cytotoxicity in comparison to their potential metabolites.
Collapse
Affiliation(s)
- Gabriela Pastuch-Gawołek
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland;
- Biotechnology Center, Silesian University of Technology, B. Krzywoustego 8, 44-100 Gliwice, Poland
| | - Julia Szreder
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland;
| |
Collapse
|
24
|
Fu Y, Zeng S, Wang Z, Huang H, Zhao X, Li M. Mechanisms of Copper-Induced Autophagy and Links with Human Diseases. Pharmaceuticals (Basel) 2025; 18:99. [PMID: 39861161 PMCID: PMC11768742 DOI: 10.3390/ph18010099] [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: 12/18/2024] [Revised: 01/04/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
As a structural and catalytic cofactor, copper is involved in many biological pathways and is required for the biochemistry of all living organisms. However, excess intracellular copper can induce cell death due to its potential to catalyze the generation of reactive oxygen species, thus copper homeostasis is strictly regulated. And the deficiency or accumulation of intracellular copper is connected with various pathological conditions. Since the success of platinum-based compounds in the clinical treatment of various types of neoplasias, metal-based drugs have shown encouraging perspectives for drug development. Compared to platinum, copper is an essential intracellular trace element that may have better prospects for drug development than platinum. Recently, the potential therapeutic role of copper-induced autophagy in chronic diseases such as Parkinson's, Wilson's, and cardiovascular disease has already been demonstrated. In brief, copper ions, numerous copper complexes, and copper-based nano-preparations could induce autophagy, a lysosome-dependent process that plays an important role in various human diseases. In this review, we not only focus on the current advances in elucidating the mechanisms of copper or copper-based compounds/preparations on the regulation of autophagy but also outline the association between copper-induced autophagy and human diseases.
Collapse
Affiliation(s)
- Yuanyuan Fu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Shuyan Zeng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhenlin Wang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Huiting Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xin Zhao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Min Li
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
25
|
Singh K, Northcote-Smith J, Feng X, Singh K, Suntharalingam K. The Anti-Cancer Stem Cell Properties of Copper(II)-Terpyridine Complexes with Attached Salicylaldehyde Moieties. Chembiochem 2025; 26:e202400703. [PMID: 39401178 DOI: 10.1002/cbic.202400703] [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: 08/27/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 11/13/2024]
Abstract
We report the synthesis, characterisation, and anti-breast cancer stem cell (CSC) properties of two copper(II)-terpyridine complexes with bidentate salicylaldehyde moieties (2-hydroxybenzaldehyde for 1 and 2-hydroxy-1-naphthaldehyde for 2). The copper(II)-terpyridine complexes 1 and 2 are stable in biologically relevant aqueous solutions and display micromolar potency towards breast CSCs. The most effective complex 1 is 5-fold and 6.6-fold more potent towards breast CSCs than salinomycin and cisplatin, respectively. The copper(II)-terpyridine complexes 1 and 2 also decrease the formation and viability of three-dimensionally cultured mammospheres within the micromolar range. Notably complex 1 is up to 7-fold more potent towards mammospheres than salinomycin or cisplatin. Mechanistic studies suggest that the copper(II)-terpyridine complexes 1 and 2 are able to readily enter breast CSCs, elevate intracellular reactive oxygen species levels, induce DNA damage (presumably by oxidative DNA cleavage), and evoke apoptosis that is independent of caspases. This study shows that the copper(II)-terpyridine motif is a useful building block for the design of anti-breast CSC agents and reinforces the therapeutic potential of copper coordination complexes.
Collapse
Affiliation(s)
- Karampal Singh
- School of Chemistry, University, of Leicester, Leicester, UK
| | | | - Xiao Feng
- School of Chemistry, University, of Leicester, Leicester, UK
| | - Kuldip Singh
- School of Chemistry, University, of Leicester, Leicester, UK
| | | |
Collapse
|
26
|
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 2025; 64:e202414652. [PMID: 39363702 PMCID: PMC11720388 DOI: 10.1002/anie.202414652] [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: 08/02/2024] [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, CNRS4 Rue Blaise Pascal67081StrasbourgFrance
- current address: Laboratoire de Chimie de Coordination (UPR 8142)CNRS31077ToulouseFrance
| | - Vincenzo Vigna
- Department of Chemistry and Chemical TechnologiesUniversità della Calabria87036Arcavacata di RendeCSItaly
| | - Hemma Schueffl
- Center for Cancer Research and Comprehensive Cancer CenterMedical University of Vienna1090ViennaAustria
| | - Francesco Stellato
- Department of PhysicsUniversità di Roma Tor VergataVia della Ricerca Scientifica 100133RomaItaly
- INFNUniversità di Roma Tor VergataVia della Ricerca Scientifica 100133RomaItaly
| | - Bertrand Vileno
- Institut de Chimie (UMR 7177)University of Strasbourg, CNRS4 Rue Blaise Pascal67081StrasbourgFrance
| | - Merwan Bouraguba
- Institut de Chimie (UMR 7177)University of Strasbourg, CNRS4 Rue Blaise Pascal67081StrasbourgFrance
| | - Gloria Mazzone
- Department of Chemistry and Chemical TechnologiesUniversità della Calabria87036Arcavacata di RendeCSItaly
| | - Olivier Proux
- Observatoire des Sciences de l'Univers de Grenoble, UAR 832CNRS-Université Grenoble Alpes38041GrenobleFrance
| | - Silvia Morante
- Department of PhysicsUniversità di Roma Tor VergataVia della Ricerca Scientifica 100133RomaItaly
- INFNUniversità di Roma Tor VergataVia della Ricerca Scientifica 100133RomaItaly
| | - Petra Heffeter
- Center for Cancer Research and Comprehensive Cancer CenterMedical University of Vienna1090ViennaAustria
| | - Emilia Sicilia
- Department of Chemistry and Chemical TechnologiesUniversità della Calabria87036Arcavacata di RendeCSItaly
| | - Peter Faller
- Institut de Chimie (UMR 7177)University of Strasbourg, CNRS4 Rue Blaise Pascal67081StrasbourgFrance
- Institut Universitaire de France (IUF)1 rue Descartes75231ParisFrance
| |
Collapse
|
27
|
Scalise M, Scanga R, Console L, Galluccio M, Pochini L, Indiveri C. Lysine 204 is crucial for the antiport function of the human LAT1 transporter. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2025; 1866:149520. [PMID: 39428051 DOI: 10.1016/j.bbabio.2024.149520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/11/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
LAT1 (SLC7A5) catalyzes an antiport reaction of amino acids with specificity towards the essential ones. It is mainly expressed at the Blood Brain Barrier and placenta barriers, but it becomes over-expressed in virtually all human cancers even if originating from tissues with lower expression levels. The antiport reaction of LAT1 is crucial at the BBB since its inherited loss causes Autism Spectrum Disorder. We have investigated the possible molecular determinant of the antiport by site-directed mutagenesis, in vitro transport assay and computational analysis. Previous data indicated that mutation of K204 impairs, but does not knock-out LAT1 functionality. We have investigated the activity changes in the K204Q mutant by following the transport of [3H]-histidine, one of the major substrates, in proteoliposomes harbouring the WT or K204Q. In the mutant, the [3H]-histidine uptake and efflux are not more stimulated by the counter-substrate as they occur in the WT. Moreover, the mutation strongly decreases the substrate affinity and alters the pH dependence of K204Q. Molecular Dynamics analysis correlates well with the experimental data since it shows that substrate prematurely escapes the binding site. In addition, the K204Q shows a strongly increased mobility in those regions, transmembrane domains and random coils, involved in the transport cycle. The identified Lys residue could be responsible of the same phenomenon in those members of the SLC7 family, described as antiporters, in which it is conserved.
Collapse
Affiliation(s)
- Mariafrancesca Scalise
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy
| | - Raffaella Scanga
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy
| | - Lara Console
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy
| | - Michele Galluccio
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy
| | - Lorena Pochini
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy; CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, via Amendola 165/A, 70126 Bari, Italy.
| | - Cesare Indiveri
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via P. Bucci 4C, 87036 Arcavacata di Rende, Italy; CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, via Amendola 165/A, 70126 Bari, Italy.
| |
Collapse
|
28
|
Karim A, Ullah N, Iqbal M, Malekshah RE, Ali S, Hsu SCN. In silico drug encapsulation using 2-hydroxypropyl-β-CD, tyrosine kinase and tyrosinase inhibition of dinuclear Cu(II) carboxylate complexes. J Mol Graph Model 2025; 134:108903. [PMID: 39541649 DOI: 10.1016/j.jmgm.2024.108903] [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: 09/11/2024] [Revised: 10/29/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
In recent years, copper carboxylate complexes have garnered significant interest for biological applications. This study focuses on 20 Cu(II) carboxylate complexes selected from our previous research. Due to the hydrophobic nature of these complexes, the 2-hydroxypropyl-β-cyclodextrin (2HPβCD) was employed as a carrier to reduce toxicity and increase solubility for controlling drug delivery. Monte Carlo calculations were performed to confirm the interaction between the optimized structures of Cu(II) complexes and 2HPβCD, forming a host-guest system. All the structures were simulated and optimized using DFT-D calculations in Material Studio 2017. The results indicated that a neutral medium is more favorable for the adsorption of these complexes into 2HPβCD. More negative binding energy values suggested strong and energetically favorable adsorption on 2HPβCD. Complexes 4, 5, and 7 exhibited the highest interaction, making them excellent candidates for drug delivery systems. DFT-D calculations were also used to investigate the release of complexes, revealing that complexes 5, 14, and 19 were difficult to release due to their lowest energy. In contrast, complexes 8, 9, and 16 were found to be most efficient to release due to weak non-covalent interactions with 2HPβCD as we can predict from binding energy obtained by DFT-D. No specific trend was observed in the interaction of the complexes with 2HPβCD. Additionally, the effects of these complexes on c-kit tyrosine kinase and Mushroom tyrosinase were studied by molecular docking. The results demonstrated that all the complexes interacted with the active site of respective receptors through hydrophobic interactions. Complexes containing 1,10-phenanthroline and 2,2-bipyrdine were identified as having a strong, spontaneous binding ability with receptors.
Collapse
Affiliation(s)
- Amir Karim
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Chemistry Bacha Khan University Charsadda 24420, KPK, Pakistan
| | - Najeeb Ullah
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Chemistry Bacha Khan University Charsadda 24420, KPK, Pakistan
| | - Muhammad Iqbal
- Department of Chemistry Bacha Khan University Charsadda 24420, KPK, Pakistan
| | - Rahime Eshaghi Malekshah
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Chemistry, Semnan University, Semnan, Iran.
| | - Saqib Ali
- Department of Chemistry Quaid-i-Azam University Islamabad 45320, Pakistan
| | - Sodio C N Hsu
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| |
Collapse
|
29
|
Elwell CE, Stein E, Lewis A, Hamaway S, Alexis KA, Tanski JM, Barnum TJ, Connelly CM, Tyler LA. Synthesis, characterization and comparative biological activity of a novel set of Cu(II) complexes containing azole-based ligand frames. J Inorg Biochem 2025; 262:112736. [PMID: 39332382 DOI: 10.1016/j.jinorgbio.2024.112736] [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: 06/26/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 09/29/2024]
Abstract
The synthesis and spectroscopic characterization of three complexes containing a substituted 2-(2-pyridyl)benzothiazole (PyBTh) group in the ligand frame are reported along with the comparative biological activity. The ligands have been substituted at the 6-position with either a methoxy (Py(OMe)BTh) or a methyl group (Py(Me)BTh). Reaction of Py(OMe)BTh with either CuCl2 or Cu(NO3)2·2.5 H2O yielded the monomeric [Cu(Py(OMe)BTh))2(NO3)]NO3·1.5 MeOH, (1·1.5 MeOH) complex or the dimeric [Cu(Py(OMe)BTh)Cl2]2 (2), respectively, with the nuclearity of the complex dependent on the starting Cu(II) salt. Reaction between the methyl substituted ligand and Cu(NO3)2·2.5 H2O resulted in the isolation of Cu(Py(Me)BTh)(NO3)2·0.5 THF (3·0.5 THF). Complexes 1-3 were fully characterized. Cyclic voltammetry measurements were performed on all three complexes as well as on [Cu(PyBTh)2(H2O)](BF4)2 (4), a compound previously reported by us which contains the unsubstituted 2-(2-pyridyl)benzothiazole ligand. The biological activity was studied and included concentration dependent DNA binding and cleavage, antibacterial activity, and cancer cell toxicity. All complexes exhibited DNA cleavage activity, however 2 and 4 were found to be the most potent. Mechanistic studies revealed that the nuclease activity is dependent on an oxidative mechanism reliant principally on O2-. Antibacterial studies revealed complex 4 was more potent compared to 1-3. Cancer cell toxicity studies were carried out on HeLa, PC-3, and MCF7 cells with 1-4, Cu(QBTh)(NO3)2(H2O) and Cu(PyBIm)3(BF4)2. The differences in the observed toxicities suggests the importance of the ligand and its substituents in modulating cell death.
Collapse
Affiliation(s)
- Courtney E Elwell
- Department of Chemistry and Biochemistry, Union College, Schenectady, NY 12308, United States
| | - Emily Stein
- Department of Chemistry and Biochemistry, Union College, Schenectady, NY 12308, United States
| | - Adam Lewis
- Department of Chemistry and Biochemistry, Union College, Schenectady, NY 12308, United States
| | - Stefan Hamaway
- Department of Chemistry and Biochemistry, Union College, Schenectady, NY 12308, United States
| | - Kennedy A Alexis
- Department of Chemistry and Biochemistry, Union College, Schenectady, NY 12308, United States
| | - Joseph M Tanski
- Department of Chemistry, Vassar College, Poughkeepsie, NY 12604, United States
| | - Timothy J Barnum
- Department of Chemistry and Biochemistry, Union College, Schenectady, NY 12308, United States
| | - Colleen M Connelly
- Department of Chemistry and Biochemistry, Union College, Schenectady, NY 12308, United States.
| | - Laurie A Tyler
- Department of Chemistry and Biochemistry, Union College, Schenectady, NY 12308, United States.
| |
Collapse
|
30
|
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 2025; 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] [MESH Headings] [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
|
31
|
Man X, Li W, Zhu M, Li S, Xu G, Zhang Z, Liang H, Yang F. Anticancer Tetranuclear Cu(I) Complex Catalyzes a Click Reaction to Synthesize a Chemotherapeutic Agent in situ to Achieve Targeted Dual-Agent Combination Therapy for Cancer. Angew Chem Int Ed Engl 2024; 63:e202411846. [PMID: 39295439 DOI: 10.1002/anie.202411846] [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: 06/24/2024] [Revised: 09/02/2024] [Accepted: 09/18/2024] [Indexed: 09/21/2024]
Abstract
To develop next-generation metal-based drugs and dual-drug combination therapy for cancer, we proposed to develop a copper (Cu) complex that exerts anticancer function by integrating chemotherapy, immunotherapy and catalyzes a click reaction for the in situ synthesis of a chemotherapeutic agent, thereby achieving targeted dual-agent combination therapy. We designed and synthesized a tetranuclear Cu(I) complex (Cu4) with remarkable cytotoxicity and notable catalytic ability for the in situ synthesis of a chemotherapeutic agent via Cu(I)-catalyzed azide-alkyne 1,3-cycloaddition (CuAAC). We also constructed an apoferritin (AFt)-Cu4 nanoparticles (NPs) delivery system. Aft-Cu4 NPs not only showed an enhanced performance of tumor growth inhibition, but also improved the targeting ability and reduced the systemic toxicity of Cu4 in vivo. Importantly, the anticancer effect was enhanced by combining the Aft-Cu4 NPs with the resveratrol analogue obtained from the CuAAC reaction in situ. Finally, we revealed the anticancer mechanism of the Cu4/Aft-Cu4 NPs, which involves both cuproptosis and cuproptosis-induced systemic immune response.
Collapse
Affiliation(s)
- Xueyu Man
- 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), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, 541004, China
| | - Wenjuan Li
- 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), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, 541004, China
| | - Minghui Zhu
- 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), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, 541004, China
| | - Shanhe Li
- 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), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, 541004, China
| | - Gang Xu
- 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), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, 541004, 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), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, 541004, China
| | - Hong Liang
- 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), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, 541004, 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), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi, 541004, China
- School of Pharmaceutical Sciences, Jiangxi Normal University, Nanchang, Jiangxi, 330022, China
| |
Collapse
|
32
|
Fabra D, Melones-Herrero J, Velazquez-Gutierrez J, Matesanz AI, Aliseda PD, Figueiras S, Aguilar-Rico F, Calés C, Sánchez-Pérez I, Quiroga AG. A select thiosemicarbazone copper(II) complex induces apoptosis in gastric cancer and targets cancer stem cells reducing pluripotency markers. Eur J Med Chem 2024; 280:116994. [PMID: 39489985 DOI: 10.1016/j.ejmech.2024.116994] [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: 09/06/2024] [Revised: 10/11/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Copper(II)-based complexes are promising candidates as anti-cancer agents due to their ability to target cancer cells. Here we describe the synthesis and characterization of two copper(II) thiosemicarbazone complexes with the ligands 4-(dimethylamino)benzaldehyde N4-methylthiosemicarbazone (HL1) and 4-(dimethylamino)benzaldehyde N4-(4-(dimethylamino)phenylthiosemicarbazone (HL2) and general formula [Cu(L)2]. The complexes show stability in aqueous solution with 1 % of DMSO that allows to stablish its solution profile in biological buffers. Compound [Cu(L1)₂] lipophilicity was lower than [Cu(L2)₂], however, its solubility in biological buffer was not only better but also its DLS and ζ-potential data. In vitro studies demonstrate a higher cytotoxic effect of [Cu(L1)₂] on gastric cancer cells. The proposed mechanism of action consists in the generation of free radicals that induce DNA lesions, oxidative stress and ultimately autophagy deregulation and apoptosis. Additionally, [Cu(L1)₂] is equally active on gastric cancer stem cells and tumor cells resistant to cisplatin. More importantly, stem cells treated with [Cu(L1)₂] show a downregulation of pluripotency markers such as TWIST, NANOG and OCT4. Overall, our results with [Cu(L1)₂] prompt a significant advancement in the development of rational-designed pharmaceuticals for combating cancer.
Collapse
Affiliation(s)
- David Fabra
- Department of Inorganic Chemistry, School of Sciences, Universidad Autónoma de Madrid (UAM), Madrid, 28049, Spain
| | - Jorge Melones-Herrero
- Department of Biochemistry. School of Medicine, UAM, Madrid, 28029, Spain; Instituto de Investigaciones Biomédicas "Sols-Morreale" (IIBM), CSIC-UAM, Madrid, 28029, Spain; Biomarkers and Personalized Approach to Cancer (BioPAC) Group. Area 3 Cancer -Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, 28034, Spain
| | - Javier Velazquez-Gutierrez
- Department of Biochemistry. School of Medicine, UAM, Madrid, 28029, Spain; Instituto de Investigaciones Biomédicas "Sols-Morreale" (IIBM), CSIC-UAM, Madrid, 28029, Spain
| | - Ana I Matesanz
- Department of Inorganic Chemistry, School of Sciences, Universidad Autónoma de Madrid (UAM), Madrid, 28049, Spain
| | - Patricia D Aliseda
- Department of Biochemistry. School of Medicine, UAM, Madrid, 28029, Spain; Instituto de Investigaciones Biomédicas "Sols-Morreale" (IIBM), CSIC-UAM, Madrid, 28029, Spain; Biomarkers and Personalized Approach to Cancer (BioPAC) Group. Area 3 Cancer -Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, 28034, Spain
| | - Sofia Figueiras
- Department of Biochemistry. School of Medicine, UAM, Madrid, 28029, Spain; Instituto de Investigaciones Biomédicas "Sols-Morreale" (IIBM), CSIC-UAM, Madrid, 28029, Spain; Biomarkers and Personalized Approach to Cancer (BioPAC) Group. Area 3 Cancer -Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, 28034, Spain
| | - Francisco Aguilar-Rico
- Department of Inorganic Chemistry, School of Sciences, Universidad Autónoma de Madrid (UAM), Madrid, 28049, Spain
| | - Carmela Calés
- Department of Biochemistry. School of Medicine, UAM, Madrid, 28029, Spain; Instituto de Investigaciones Biomédicas "Sols-Morreale" (IIBM), CSIC-UAM, Madrid, 28029, Spain; Biomarkers and Personalized Approach to Cancer (BioPAC) Group. Area 3 Cancer -Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, 28034, Spain
| | - Isabel Sánchez-Pérez
- Department of Biochemistry. School of Medicine, UAM, Madrid, 28029, Spain; Instituto de Investigaciones Biomédicas "Sols-Morreale" (IIBM), CSIC-UAM, Madrid, 28029, Spain; Biomarkers and Personalized Approach to Cancer (BioPAC) Group. Area 3 Cancer -Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, 28034, Spain; Unidad Asociada de Biomedicina UCLM-CSIC, Madrid, 28029, Spain; Centro de Investigación Biomédica en Red, Área Rare Diseases, CIBERER, ISCIII, Madrid, 28029, Spain.
| | - Adoracion G Quiroga
- Department of Inorganic Chemistry, School of Sciences, Universidad Autónoma de Madrid (UAM), Madrid, 28049, Spain; Institute for Advance Research in Chemistry UAM, Madrid, 28049, Spain.
| |
Collapse
|
33
|
Kuila A, Maity R, Acharya P, Sarkar T, Bhakat A, Brandao P, Pattanayak S, Maity T, Dalai S, Sarkar K, Samanta BC. Exploring the Potential Fungicidal Applications of a Cu(II) Complex with Schiff Base and Carboxylates against Fusarium equisetum. ACS OMEGA 2024; 9:48273-48284. [PMID: 39676998 PMCID: PMC11635468 DOI: 10.1021/acsomega.4c05824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/19/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024]
Abstract
Given the critical need to preserve agricultural sustainability, there is an urgent call to address fungal infections. Our study presents a promising approach by focusing on SIX (Secreted in Xylem) proteins as a pivotal target for the development of innovative fungicidal strategies. Within the sphere of this study, we meticulously scrutinize the antifungal efficacy of our synthesized Cu(II) complex formulated as [Cu(L1)2(L2)]+(ClO4)-, where L1 represents (E)-cyclohexyl-N(pyridine-2-xlmethylene) methanamine and L2H denotes cinnamic acid, compared against a commercially available fungicide comprising 4% hexaconazole and 68% zineb. Employing in silico methodologies, we undertake a comparative analysis targeting SIX proteins to discern the potency of our compound. The X-ray diffraction, 1H NMR, and FTIR spectroscopic techniques were utilized to elucidate the structure of the complex methodically. The lipophilicity test of the complex signifies its potential lipophilic nature and prompted further investigation into the complex's interaction with DNA (DNA) and bovine serum albumin (BSA). The binding constant values suggested a notable interaction between the complex and both DNA and BSA. The antifungal test reveal that our complex emerges as a potent contender in the battle against Fusarium equisetum (F.E.), exhibiting a commendable efficacy that positions it as a viable substitute for the incumbent commercial fungicide. This discovery predicts well the prospect of bolstering agricultural resilience and safeguarding global food security in the face of pervasive fungal threats.
Collapse
Affiliation(s)
- Arun Kuila
- Department
of Chemistry, Mugberia Gangadhar Mahavidyalaya, Bhupatinagar, Purba Medinipur, 721425 Contai, West Bengal, India
- Department
of Chemistry and Chemical Technology, Vidyasagar
University, Midnapore 721102, West Bengal, India
| | - Ribhu Maity
- Department
of Chemistry, Mugberia Gangadhar Mahavidyalaya, Bhupatinagar, Purba Medinipur, 721425 Contai, West Bengal, India
| | - Prasun Acharya
- Department
of Chemistry, Mugberia Gangadhar Mahavidyalaya, Bhupatinagar, Purba Medinipur, 721425 Contai, West Bengal, India
- Department
of Chemistry and Chemical Technology, Vidyasagar
University, Midnapore 721102, West Bengal, India
| | - Tuhin Sarkar
- Department
of Microbiology, University of Kalyani, West Bengal, Kalyani 741235, India
| | - Ankika Bhakat
- Department
of Microbiology, University of Kalyani, West Bengal, Kalyani 741235, India
| | - Paula Brandao
- Departamento
de Química, CICECO, Universidade
de Aveiro, 3810-193 Aveiro, Portugal
| | - Satyajit Pattanayak
- Department
of Chemistry, Mugberia Gangadhar Mahavidyalaya, Bhupatinagar, Purba Medinipur, 721425 Contai, West Bengal, India
| | - Tithi Maity
- Department
of Chemistry, Prabhat Kumar College, Purba Medinipur, 721401 Contai, West Bengal, India
| | - Sudipta Dalai
- Department
of Chemistry and Chemical Technology, Vidyasagar
University, Midnapore 721102, West Bengal, India
| | - Keka Sarkar
- Department
of Microbiology, University of Kalyani, West Bengal, Kalyani 741235, India
| | - Bidhan Chandra Samanta
- Department
of Chemistry, Mugberia Gangadhar Mahavidyalaya, Bhupatinagar, Purba Medinipur, 721425 Contai, West Bengal, India
| |
Collapse
|
34
|
Wang K, Wang L, Shang Z, Yang X, Li H, Wang X, Zhu M, Meng Q. A series of DNA targeted Cu (II) complexes containing 1,8-naphthalimide ligands: Synthesis, characterization and in vitro anticancer activity. J Inorg Biochem 2024; 261:112721. [PMID: 39236444 DOI: 10.1016/j.jinorgbio.2024.112721] [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: 06/21/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/07/2024]
Abstract
Copper(II) complexes are very promising candidates for platinum-based anticancer agents. Herein, three Cu (II) complexes (1-3) containing 1,8-naphthalimide ligands were synthesized and characterized by FT-IR, elemental analysis, ESI-MS and single crystal X-ray diffraction (complex 3). In addition, a control compound (complex 4) without 1,8-naphthalimide ligand was synthesized and characterized. The in vitro anticancer activity of the synthesized complexes against five cancer cell lines and one normal cell line was evaluated by MTS assay. The results displayed the antitumor activity of complexes 1-3 was controlled by the aliphatic chain length of ligands, their cytotoxicity was in the order 3 > 2 > 1, giving the IC50 values ranging from 2.874 ± 0.155 μM to 31.47 ± 0.29 μM against five cancer cell lines. Complex 4 showed less activity in comparison with complex 1-3. Notably, complexes 1-3 displayed much higher selectivity (SI = 2.65 to 10.16) compared to complex 4 (SI = 1.0), indicated that the introduction of 1,8-naphthalimide group not only increased the activity of this series of compounds but also enhanced their specific selectivity to cancer cells. Compound 3 induced apoptosis in cancer cells and blocked the S-phase and G2/M of cancer cells. The interaction with DNA of complexes 3 and 4 was studied by UV/Vis spectroscopic titrations, competitive DNA-binding experiment, viscometry and CD spectra. The results showed that complex 3 interacted with DNA in an intercalating mode, but the interaction mode of compound 4 with DNA was electrostatic interaction.
Collapse
Affiliation(s)
- Kehua Wang
- School of Chemical Engineering, University of Science and Technology Liaoning, Anshan, Liaoning 114051, PR China; Liaoning Key Laboratory of Development and Utilization for Natural Products Active Molecules, Anshan Normal University, Anshan, Liaoning 114007, PR China
| | - Ling Wang
- The Key Laboratory of the Inorganic Molecule-Based Chemistry of Liaoning Province, Department of Coordination Chemistry, Shenyang University of Chemical Technology, Shenyang 110142, PR China
| | - Zhuye Shang
- School of Chemical Engineering, University of Science and Technology Liaoning, Anshan, Liaoning 114051, PR China
| | - Xingzhi Yang
- Center for Natural Drug Activity Screening, Kunming Institute of Botany, Chinese Academy of Sciences, PR China
| | - Hongmei Li
- Center for Natural Drug Activity Screening, Kunming Institute of Botany, Chinese Academy of Sciences, PR China
| | - Xiaochun Wang
- Liaoning Key Laboratory of Development and Utilization for Natural Products Active Molecules, Anshan Normal University, Anshan, Liaoning 114007, PR China
| | - Mingchang Zhu
- The Key Laboratory of the Inorganic Molecule-Based Chemistry of Liaoning Province, Department of Coordination Chemistry, Shenyang University of Chemical Technology, Shenyang 110142, PR China
| | - Qingtao Meng
- School of Chemical Engineering, University of Science and Technology Liaoning, Anshan, Liaoning 114051, PR China.
| |
Collapse
|
35
|
El Yaagoubi OM, Oularbi L, Salhi O, Samaki H, El Rhazi M, Aboudkhil S. Novel copper complex inhibits the proteasome in skin squamous cell carcinoma induced by DMBA in mice. J Trace Elem Med Biol 2024; 86:127533. [PMID: 39321648 DOI: 10.1016/j.jtemb.2024.127533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/11/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024]
Abstract
The proteasomal system is becoming a target for the treatment of several diseases, especially in cancer therapy. The present study aims to develop a novel copper complex that inhibits the proteasome in skin squamous cell carcinoma. New molecules based on the copper complex were synthesized for the first time to assess their potential as proteasome inhibitors, specifically targeting squamous cell carcinoma induced by 7,12-dimethylbenz(a)anthracene (DMBA) in mouse models. Fourier Transform Infrared (FTIR), X-ray diffraction (XRD), scanning electron microscopy (SEM), nuclear magnetic resonance (NMR), and energy dispersive X-ray analysis (EDX) were carried out to characterize this new copper complex. Notably, the presence of a papilloma (skin tumor) was confirmed by histopathological analysis. Subsequent investigation included the quantification of proteasome levels using a sandwich ELISA test, and the catalytic activity of the 20S proteasome was determined by measuring the fluorescence emitted after the cleavage of 7-amino-4-methylcoumarin (AMC). Hence, X-ray crystallography indicates that all Cu atoms are five-coordinated in a square-pyramidal configuration and biological activity of copper Schiff base complex, which exhibits high proteasome inhibitory activities with particular selectivity of β5 subunit. The pharmacokinetic properties (ADMET) of the copper complex named Cu(L1) showed encouraging results with very low toxicity, distribution, and absorption. Structure-activity relationship (SAR) information obtained from Cu(L1) demonstrated its selectivity and potent inhibition for β5 subunit. In this regard, this copper complex has emerged as a novel therapy for skin cancer.
Collapse
Affiliation(s)
- Ouadie Mohamed El Yaagoubi
- Laboratory of Biochemistry, Environment and Agri-Food (URAC 36), Faculty of Sciences and Techniques - Mohammedia, Hassan II University, Casablanca, Morocco.
| | - Larbi Oularbi
- Laboratory of Materials Membranes and Environment, P.B 146, Faculty of Sciences and Techniques - Mohammedia, Hassan II University, Casablanca, Morocco; Supramolecular Nanomaterials Group (SNG), Mohammed VI Polytechnic University, Benguerir, Morocco.
| | - Ouissal Salhi
- Laboratory of Materials Membranes and Environment, P.B 146, Faculty of Sciences and Techniques - Mohammedia, Hassan II University, Casablanca, Morocco.
| | - Hamid Samaki
- National Institute of Social Action (INAS), Tangier, Morocco.
| | - Mama El Rhazi
- Laboratory of Materials Membranes and Environment, P.B 146, Faculty of Sciences and Techniques - Mohammedia, Hassan II University, Casablanca, Morocco.
| | - Souad Aboudkhil
- Laboratory of Biochemistry, Environment and Agri-Food (URAC 36), Faculty of Sciences and Techniques - Mohammedia, Hassan II University, Casablanca, Morocco.
| |
Collapse
|
36
|
Vušak D, Šimunović Letić M, Tašner M, Matković-Čalogović D, Jurec J, Žilić D, Prugovečki B. Solvatomorphic Diversity in Coordination Compounds of Copper(II) with l-Homoserine and 1,10-Phenanthroline: Syntheses, Crystal Structures and ESR Study. Molecules 2024; 29:5621. [PMID: 39683780 DOI: 10.3390/molecules29235621] [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: 10/24/2024] [Revised: 11/13/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
In this study, we report the syntheses, crystal structures and magnetic properties of ternary copper(II) coordination compounds with l-homoserine (l-Hhser) and 1,10-phenanthroline (phen). Six new coordination compounds were obtained: [Cu(l-hser)(H2O)(phen)]2SO4·5H2O (1·5H2O), [Cu(μ-l-hser)(H2O)(phen)][Cu(l-hser)(H2O)(phen)]3(SO4)2∙12H2O (2·12H2O), {[Cu(μ-l-hser)(H2O)(phen)][Cu(μ-l-hser)(phen)]SO4·6H2O}n (3·6H2O), {[Cu(μ-l-hser)(H2O)(phen)]2SO4·3H2O}n (4·3H2O), [Cu(l-hser)(H2O)(phen)][Cu(l-hser)(CH3OH)(phen)]SO4·4H2O (5·4H2O) and {[Cu(l-hser)(CH3OH)(phen)][Cu(μ-l-hser)(phen)]SO4·5CH3OH}n (6·5CH3OH). It was shown that slight differences in water content in the synthetic mixtures highly influence the final product, so in some cases, two or three different products were obtained. The compounds were characterized by single-crystal X-ray diffraction and ESR spectroscopy. Crystal packings are based on intensive networks of hydrogen bonds and π interactions. Most water solvent molecules in these microporous compounds are found in discrete pockets (1∙5H2O, 2∙12H2O, 3∙6H2O, 4∙3H2O). In 5∙4H2O, water molecules are packed in pockets and 1D channels and in 6∙5CH3OH methanol solvent molecules form 1D channels. ESR spectroscopy measured from room down to liquid nitrogen temperature was used for local magnetic characterization of copper centers. The spin Hamiltonian parameters obtained from the spectral simulation revealed copper coordination geometry that is in agreement with the structural results. Furthermore, ESR spectra revealed no significant exchange coupling between copper ions. 3·6H2O showed pronounced antiproliferative activity toward human colon cancer cell lines (HCT116), human breast cancer cell line (MCF-7) and human lung cancer cell lines (H460).
Collapse
Affiliation(s)
- Darko Vušak
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102A, HR-10000 Zagreb, Croatia
| | - Marta Šimunović Letić
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102A, HR-10000 Zagreb, Croatia
| | - Marina Tašner
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102A, HR-10000 Zagreb, Croatia
| | - Dubravka Matković-Čalogović
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102A, HR-10000 Zagreb, Croatia
| | - Jurica Jurec
- Laboratory for Magnetic Resonances, Division of Physical Chemistry, Ruđer Bošković Institute, Bijenička Cesta 54, HR-10000 Zagreb, Croatia
| | - Dijana Žilić
- Laboratory for Magnetic Resonances, Division of Physical Chemistry, Ruđer Bošković Institute, Bijenička Cesta 54, HR-10000 Zagreb, Croatia
| | - Biserka Prugovečki
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102A, HR-10000 Zagreb, Croatia
| |
Collapse
|
37
|
Roy A, Khatun S, Dewale PD, Rengan AK, Chinta JP. Copper-assisted anticancer activity of hydroxycinnamic acid terpyridine conjugates on triple-negative breast cancer. Dalton Trans 2024; 53:18640-18652. [PMID: 39479915 DOI: 10.1039/d4dt02516d] [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: 11/02/2024]
Abstract
The development of active therapeutic agents to treat highly metastatic cancer while minimizing damage to healthy cells is of utmost importance. Due to potential antioxidant properties, hydroxycinnamic acid derivatives (caffeic acid and p-coumaric acids) were found to inhibit highly metastatic breast cancer cell growth both in vitro and in vivo without much effect on normal cells. Especially due to the structure-activity relationships, ester and amide derivatives of hydroxycinnamic acids are reported to gain much higher radical scavenging ability than their naked hydroxycinnamic acid analogs like caffeic acid and p-coumaric acid. These results prompted us to design a set of ligands by incorporating an amide moiety on caffeic acid and p-coumaric acid to achieve the least toxicity towards healthy cell lines. Further, the Cu(II) complexes of amide-coupled caffeic acid and p-coumaric acid ligands have been explored for their therapeutic activity on triple-negative breast cancer and other cancer cells like colon, and prostate cancer. The Cu(II) complexes (4 & 5) were characterized by UV-Vis spectroscopy, FTIR, and X-band EPR spectroscopy. The trigonal bipyramidal geometry of complexes was confirmed by the X-band EPR spectra recorded in solution state at liquid N2 temperature. The purity of the complexes was determined by elemental analysis and HPLC traces. Initially, Calf thymus DNA (ct-DNA) binding studies with the complexes were explored. The results suggested the complexes (4 & 5) bind majorly through an intercalative mode of binding with ct-DNA, whereas no significant binding was observed for the bare organic ligands (2 & 3). The intercalation binding modes of 4 and 5 were further supported by UV-visible spectroscopy, ct-DNA melting point analysis, and CD spectroscopy. Moreover, these complexes showed better activity towards cisplatin-resistant TNBC cell lines (4T1, a TNBC cell line derived from the mammary gland tissue of a mouse). The combination of antioxidants and Cu(II) as the metal center made the complexes more cytotoxic toward cancer cell lines (4T1) (IC50 ∼ 3.5 ± 2.5 μM) and the least toxic toward healthy cells (L929) (IC50 ∼ 15 ± 5 μM). Finally, the mechanism of cell death was studied using JC-1 staining and a cell colony formation assay. These studies might help in designing safer anticancer drugs for treating more aggressive types of cancer.
Collapse
Affiliation(s)
- Anindya Roy
- Department of Chemistry, National Institute of Technology Warangal, Warangal, Telangana 506004, India.
| | - Sajmina Khatun
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Hyderabad 5022854, India
| | | | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Hyderabad 5022854, India
| | - Jugun Prakash Chinta
- Department of Chemistry, National Institute of Technology Warangal, Warangal, Telangana 506004, India.
| |
Collapse
|
38
|
Balsa LM, Santa Maria de la Parra L, Ferretti V, León IE. Deciphering the Effect of a Cu(II)-hydrazone Complex on Intracellular Cell Signalling Pathways in a Human Osteosarcoma 2D and 3D Models. Chembiochem 2024; 25:e202400373. [PMID: 39121373 DOI: 10.1002/cbic.202400373] [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: 04/22/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/11/2024]
Abstract
New therapeutic strategies for osteosarcoma (OS) have demonstrated the potential efficacy of copper compounds as anticancer drugs and as a substitute for the often used platinum compounds. OS is a type of bone cancer, primarily affecting young adults and children.The main objective of this work is to discover the molecular targets and cellular pathways related to the antitumor properties of a Cu(II)-hydrazone toward human OS 2D and 3D systems. Cell viability study using MG-63 cells was evaluated in OS monolayer and spheroids. CuHL significantly reduced cell viability in OS models (IC50 2D: 2.6±0.3 μM; IC50 3D: 9.9±1.4 μM) (p<0.001). Also, CuHL inhibits cell proliferation and it induces cells to apoptosis. The main mechanism of action found for CuHL are the interaction with DNA, genotoxicity, the ROS generation and the proteasome activity inhibition. Besides, 67 differentially expressed proteins were found using proteomic approaches. Of those 67 proteins, 40 were found overexpressed and 27 underexpressed. The response to stress and to unfolded protein, as well as ATP synthesis were the most affected biological process among upregulated proteins, whilst proteins related to DNA replication and redox homeostasis were downregulated.
Collapse
Affiliation(s)
- Lucía M Balsa
- CEQUINOR (UNLP, CCT-CONICET La Plata, asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 N° 1465, La Plata, (1900), Argentina
| | - Lucía Santa Maria de la Parra
- CEQUINOR (UNLP, CCT-CONICET La Plata, asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 N° 1465, La Plata, (1900), Argentina
| | - Valeria Ferretti
- CEQUINOR (UNLP, CCT-CONICET La Plata, asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 N° 1465, La Plata, (1900), Argentina
| | - Ignacio E León
- CEQUINOR (UNLP, CCT-CONICET La Plata, asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Blvd. 120 N° 1465, La Plata, (1900), Argentina
- Cátedra de Fisiopatología, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 47 y 115, La Plata, 1900), Argentina
| |
Collapse
|
39
|
Rogalewicz B, Sierański T, Szczesio M, Olczak A, Gobis K, Orlewska C, Korona-Głowniak I, Korga-Plewko A, Iwan M, Michalczuk M, Kubik J, Adamczuk G, Korga M, Rutkowska N, Boruta T, Gas K, Sawicki M, Poleszak E, Maniukiewicz W, Świątkowski M, Czylkowska A. Physicochemical properties and mechanism of action of a new copper(ii) pyrazine-based complex with high anticancer activity and selectivity towards cancer cells. RSC Adv 2024; 14:36295-36307. [PMID: 39534047 PMCID: PMC11556459 DOI: 10.1039/d4ra06874b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Two compounds, benzyl-2-(amino(pyrazin-2-yl)methylene)-1-methylhydrazine-1-carbodithioate (L) and its copper(ii) complex Cu(L) were synthesized and studied in terms of their physicochemical properties, including single crystal, spectroscopic and magnetic properties; in silico simulations, including DFT calculations and pharmacokinetic profile analysis; and in vitro biological activity. The Cu(L) compound was found to exhibit good anticancer activity against A375, PANC-1, MKN-74, T-47D, HeLa, and NCI-H1563 cells, with the IC50 value against the HeLa cell line reaching 17.50 μM, significantly surpassing the activity of the organic ligand. Moreover, at the same time, the Cu(L) complex did not exhibit significant toxicity towards healthy cells. Mechanism of action studies revealed that its activity is connected with the oxidative stress and redox imbalance caused by the upregulation of genes encoding superoxide dismutase (SOD2) and catalase (CAT) antioxidant enzymes. The reported results further underscore the anticancer potential of pyrazine-based copper(ii) complexes.
Collapse
Affiliation(s)
- B Rogalewicz
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology Żeromskiego 116 Lodz 90-924 Poland
| | - T Sierański
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology Żeromskiego 116 Lodz 90-924 Poland
| | - M Szczesio
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology Żeromskiego 116 Lodz 90-924 Poland
| | - A Olczak
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology Żeromskiego 116 Lodz 90-924 Poland
| | - K Gobis
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Gdansk Gen. Hallera 107 Gdańsk 80-416 Poland
| | - C Orlewska
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Gdansk Gen. Hallera 107 Gdańsk 80-416 Poland
| | - I Korona-Głowniak
- Department of Pharmaceutical Microbiology, Medical University of Lublin Lublin 20-093 Poland
| | - A Korga-Plewko
- Independent Medical Biology Unit, Medical University of Lublin Jaczewskiego 8b Lublin 20-093 Poland
| | - M Iwan
- Department of Toxicology, Medical University of Lublin Chodźki 8b Lublin 20-093 Poland
| | - M Michalczuk
- Independent Medical Biology Unit, Medical University of Lublin Jaczewskiego 8b Lublin 20-093 Poland
| | - J Kubik
- Independent Medical Biology Unit, Medical University of Lublin Jaczewskiego 8b Lublin 20-093 Poland
| | - G Adamczuk
- Independent Medical Biology Unit, Medical University of Lublin Jaczewskiego 8b Lublin 20-093 Poland
| | - M Korga
- Independent Medical Biology Unit, Medical University of Lublin Jaczewskiego 8b Lublin 20-093 Poland
| | - N Rutkowska
- Institute of Molecular and Industrial Biotechnology, Lodz University of Technology Stefanowskiego 2/22 Lodz 90-537 Poland
| | - T Boruta
- Faculty of Process and Environmental Engineering, Department of Bioprocess Engineering, Lodz University of Technology ul. Wolczanska 213 Lodz 93-005 Poland
| | - K Gas
- Institute of Physics, Polish Academy of Sciences Aleja Lotnikow 32/46 Warsaw PL-02668 Poland
| | - M Sawicki
- Institute of Physics, Polish Academy of Sciences Aleja Lotnikow 32/46 Warsaw PL-02668 Poland
| | - E Poleszak
- Laboratory of Preclinical Testing, Chair and Department of Applied and Social Pharmacy, Medical University of Lublin Chodzki 1 Lublin 20-093 Poland
| | - W Maniukiewicz
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology Żeromskiego 116 Lodz 90-924 Poland
| | - M Świątkowski
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology Żeromskiego 116 Lodz 90-924 Poland
| | - A Czylkowska
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology Żeromskiego 116 Lodz 90-924 Poland
| |
Collapse
|
40
|
Pinheiro T, Alves LC, Matos AP, Correia I, Costa Pessoa J, Marques F. Cellular targets of cytotoxic copper phenanthroline complexes: a multimodal imaging quantitative approach in single PC3 cells. Metallomics 2024; 16:mfae051. [PMID: 39510960 DOI: 10.1093/mtomcs/mfae051] [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: 06/13/2024] [Accepted: 11/06/2024] [Indexed: 11/15/2024]
Abstract
Metal complexes are emerging as promising alternatives to traditional platinum-based cancer treatments, offering reduced side effects. However, understanding their cellular uptake and distribution and quantifying their presence at the single cell level remains challenging. Advanced imaging techniques, including transmission electron microscopy, synchrotron radiation X-ray fluorescence, and energetic ion beam-based nuclear microscopy (scanning transmission ion microscopy, particle-induced X-ray emission, elastic backscattering spectrometry), allow detailed high-resolution visualization of structure and morphology, high sensitivity for elemental detection with quantification within single cells, and the construction of 3D models of metal distribution, positioning them as powerful tools for assessing the cellular uptake and compartmentalization of complexes. Three Cu(II) complexes [Cu(phen)2(H2O)](NO3)2 (1), [Cu(Me2phen)2(NO3)]NO3 (2) and [Cu(amphen)2(H2O)](NO3)2 (3), (phen = 1,10-phenanthroline, Me2phen = 4,7-dimethyl-1,10-phen, amphen = 5-amino-phen) were investigated for Cu uptake and distribution in PC3 prostate cancer cells. All complexes show significant Cu uptake regardless of media concentration. Cu concentrations in the cytoplasm and nucleus are similar between treatments. Complexes 1 and 3 concentrate Cu in the nuclear region and show a vesicle-like pattern around the nucleus, while 2 shows a dispersed cytoplasmic pattern with large vesicles. The 3D models confirm that Cu is not retained at the plasma membrane, with complex 1 targeting the nucleus and 2 remaining in the cytoplasm. These results highlight the importance of quantifying metal distribution and correlating it with structural changes to understand the relevance of the ligand in the mechanisms of cellular uptake and targeting, crucial for the development of effective metal-based cancer therapies.
Collapse
Affiliation(s)
- Teresa Pinheiro
- Instituto de Bioengenharia e Biociências, Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal
| | - Luís C Alves
- Centro de Ciências e Tecnologias Nucleares, Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| | - António P Matos
- Egas Moniz Center for Interdisciplinary Research, Egas Moniz School of Health and Science, Campus Universitário, Quinta da Granja, Monte de Caparica, 2829-511 Caparica, Portugal
| | - Isabel Correia
- Centro de Química Estrutural, Institute of Molecular Sciences, and Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - João Costa Pessoa
- Centro de Química Estrutural, Institute of Molecular Sciences, and Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Fernanda Marques
- Centro de Ciências e Tecnologias Nucleares, Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| |
Collapse
|
41
|
Varna D, Geromichalos GD, Dalezis P, Hatzidimitriou AG, Psomas G, Zachariadis G, Psatha K, Aivaliotis M, Papi R, Trafalis D, Angaridis PA. Amine-substituted heterocyclic thioamide Cu(I) and Ag(I) complexes as effective anticancer and antibacterial agents targeting the periplasm of E. coli bacteria. Eur J Med Chem 2024; 277:116746. [PMID: 39146831 DOI: 10.1016/j.ejmech.2024.116746] [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: 05/15/2024] [Revised: 07/31/2024] [Accepted: 08/03/2024] [Indexed: 08/17/2024]
Abstract
Metal complexes showing dual activity against cancer and bacterial infections are currently the focus of significant interest for their potential in treating life-threatening diseases. Aiming to investigate the impact of ligand substituents on these bioactivity properties of Group 11 d10 metal complexes, we herein present a series of mononuclear Cu(I) and Ag(I) complexes featuring the bis-NH2-substituted heterocyclic thioamide dap2SH (=4,6-diaminopyrimidine-2-thione), namely [AgCl(dap2SH)(PPh3)2] (1), [CuBr(dap2SH)(PPh3)2] (2), [CuBr(dap2SH)(xantphos)] (3), [Ag(dap2S)(xantphos)] (4), and [Cu(dap2S)(xantphos)] (5) (xantphos = 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene). Complexes were characterized by means of different physicochemical methods (i.e., single crystal X-ray diffraction as well as FTIR, NMR, UV-Vis and fluorescence spectroscopy), and studied in-vitro for their antibacterial and anticancer activity against a variety of bacterial strains and cancer cell lines. Complexes 1-3 effectively inhibited both Gram (+) and Gram (-) bacterial growth, while cellular uptake studies for the most potent complex 1 against E. coli bacteria revealed the accumulation of Ag(I) ions in the periplasm of the bacteria. A high anti-proliferative effect was observed for 1 and 5 against A549, MCF7 and PC3 cancer cell lines, with 1 being capable of inducing apoptosis in A549 cells, as suggested by flow cytometry analysis. DNA interaction studies revealed the capacity of 1 to intercalate between base-pairs of CT DNA. All complexes had a moderate-to-high capacity to scavenge free radicals preventing oxidative stress. Molecular docking calculations, in combination with the experimentally obtained data, provided insights for potential mechanisms of the bioactivity of the complexes.
Collapse
Affiliation(s)
- Despoina Varna
- Laboratory of Inorganic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - George D Geromichalos
- Laboratory of Inorganic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Panagiotis Dalezis
- Laboratory of Pharmacology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527, Athens, Greece
| | - Antonios G Hatzidimitriou
- Laboratory of Inorganic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - George Psomas
- Laboratory of Inorganic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Georgios Zachariadis
- Laboratory of Analytical Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Konstantina Psatha
- Laboratory of Medical Biology - Medical Genetics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece; Laboratory of Biological Chemistry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece; Functional Proteomics and Systems Biology (FunPATh) - Center for Interdisciplinary Research and Innovation (CIRI-AUTH), 57001, Thessaloniki, Greece
| | - Michalis Aivaliotis
- Laboratory of Biological Chemistry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece; Functional Proteomics and Systems Biology (FunPATh) - Center for Interdisciplinary Research and Innovation (CIRI-AUTH), 57001, Thessaloniki, Greece; Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Rigini Papi
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Dimitrios Trafalis
- Laboratory of Pharmacology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527, Athens, Greece.
| | - Panagiotis A Angaridis
- Laboratory of Inorganic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece.
| |
Collapse
|
42
|
Paliwal K, Swain A, Mishra DP, Sudhadevi Antharjanam PK, Kumar M. A novel copper(II) complex with a salicylidene carbohydrazide ligand that promotes oxidative stress and apoptosis in triple negative breast cancer cells. Dalton Trans 2024; 53:17702-17720. [PMID: 39420621 DOI: 10.1039/d4dt01914h] [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: 10/19/2024]
Abstract
We report the synthesis, characterization, anti-cancer activity and mechanism of action of a novel water-soluble Cu(II) complex with salicylidene carbohydrazide as the ligand and o-phenanthroline as the co-ligand. The synthesized complex (1) was characterized by FT-IR, EPR, and electronic spectroscopy, as well as single crystal X-ray diffraction. This compound was found to be paramagnetic from EPR spectra and X-ray crystallography revealed that the molecule crystallized in an orthorhombic crystal system. The crystal lattice was asymmetric containing two distinct binuclear copper complexes containing the Schiff base as the major ligand, o-phenanthroline as a co-ligand, two nitrate anions, and two water molecules. The Cu(II) in the first site coordinated with the enolised ligand comprising enolate O-, phenolate O-, and the imine N and N,N from o-phen. The major part of this complex exists as Cu(II) coordinated with two H2O molecules at the second site with nitrate acting as the counter anion. However, a smaller portion of the complex exists where Cu(II) is coordinated with NO3- and H2O, and the remaining water molecule acts as lattice water. It was tested for DNA binding and cleavage properties which revealed that it binds in an intercalative mode to CT-DNA with Kb value of 1.25 × 104 M-1. Furthermore, cleavage studies reveal that the complex has potential for efficient DNA cleavage under both oxidative and hydrolytic conditions. It was able to enhance the rate of cleavage by 2.8 × 108 times. The complex shows good cytotoxicity to breast cancer monolayer (2D) as well as spheroid (3D) systems. The IC50 values for MDA-MB-231 and MCF-7 monolayer culture was calculated as 1.86 ± 0.17 μM and 2.22 ± 0.08 μM, respectively, and in (3D) spheroids of MDA-MB-231 cells, the IC50 value was calculated to be 1.51 ± 0.29 μM. It was observed that the complex outperformed cisplatin in both breast cancer cell lines. The cells treated with complex 1 underwent severe DNA damage, increased oxidative stress and cell cycle arrest which finally led to programmed cell death or apoptosis in triple negative breast cancer cells through an intrinsic pathway.
Collapse
Affiliation(s)
- Kumudini Paliwal
- Department of Chemical Engineering, Birla Institute of Technology and Science-Pilani, K.K. Birla Goa Campus, Zuarinagar, Goa 403726, India.
| | - Abinash Swain
- Cell Death Research Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Lucknow, Uttar Pradesh 226031, India
| | - Durga Prasad Mishra
- Cell Death Research Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Lucknow, Uttar Pradesh 226031, India
| | - P K Sudhadevi Antharjanam
- Sophisticated Analytical Instrument Facility, Indian Institute of Technology-Madras, Chennai 600 036, India
| | - Manjuri Kumar
- Department of Chemical Engineering, Birla Institute of Technology and Science-Pilani, K.K. Birla Goa Campus, Zuarinagar, Goa 403726, India.
| |
Collapse
|
43
|
Fernández CY, Alvarez N, Rocha A, Mendes LFS, Costa-Filho AJ, Ellena J, Batista AA, Facchin G. Phenanthroline and phenyl carboxylate mixed ligand copper complexes in developing drugs to treat cancer. J Inorg Biochem 2024; 260:112700. [PMID: 39163715 DOI: 10.1016/j.jinorgbio.2024.112700] [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: 04/03/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/22/2024]
Abstract
The success of a classic inorganic coordination compound, Cisplatin, cis-[Pt(NH3)2Cl2], as the first anticancer metallodrug started a field of research dedicated to discovering coordination compounds with antitumor activity, encompassing various metals. Among these, copper complexes have emerged as interesting candidates to develop drugs to treat cancer. In this work, mixed ligand complexes of Cu(II) with diimines (phenanthroline or 4-methylphenanthroline) and 3-(4-hydroxyphenyl)propanoate, phenylcarboxylate or phenylacetate were synthesized. They were characterized in the solid state, including a new crystal structure of [Cu2(3-(4-hydroxyphenyl)propanoate)3(phenanthroline)2]Cl·H2O. The obtained complexes presented a variety of stoichiometries. In solution, complexes were partially dissociated in the corresponding Cu-diimine complex. The complexes bound to the DNA by partial intercalation and groove binding, as assessed by Circular Dichroism, relative viscosity change and UV-Vis titration. The cytotoxicity of the complexes was determined in vitro on MDA-MB-231, MCF-7 (human metastatic breast adenocarcinomas, the first triple negative), MCF-10A (breast nontumoral), A549 (human lung epithelial carcinoma), and MRC-5 (human nontumoral lung epithelial cells), finding an activity higher than that of Cisplatin, although with less selectivity.
Collapse
Affiliation(s)
- Carlos Y Fernández
- Facultad de Química, Universidad de la República, Av. General Flores 2124, Montevideo, Uruguay; Programa de Posgrados de la Facultad de Química, Facultad de Química, Universidad de la República, Gral. Flores 2124, Montevideo 11800, Uruguay
| | - Natalia Alvarez
- Facultad de Química, Universidad de la República, Av. General Flores 2124, Montevideo, Uruguay
| | - Analu Rocha
- Departamento de Química, Universidade Federal de São Carlos, CP 676, 13565-905, São Carlos (SP), Brazil; Instituto de Física de São Carlos, Universidade de São Paulo, Av. do Trabalhador São-carlense 400, 143566-590, São Carlos (SP), Brazil
| | - Luis Felipe S Mendes
- Instituto de Física de São Carlos, Universidade de São Paulo, Av. do Trabalhador São-carlense 400, 143566-590, São Carlos (SP), Brazil
| | - Antonio J Costa-Filho
- Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes, 14040-901, Ribeirão Preto (SP), Brazil
| | - Javier Ellena
- Instituto de Física de São Carlos, Universidade de São Paulo, Av. do Trabalhador São-carlense 400, 143566-590, São Carlos (SP), Brazil
| | - Alzir A Batista
- Departamento de Química, Universidade Federal de São Carlos, CP 676, 13565-905, São Carlos (SP), Brazil
| | - Gianella Facchin
- Facultad de Química, Universidad de la República, Av. General Flores 2124, Montevideo, Uruguay.
| |
Collapse
|
44
|
Małyszko M, Przybyłkowski A. Copper and Colorectal Cancer. Cancers (Basel) 2024; 16:3691. [PMID: 39518128 PMCID: PMC11544869 DOI: 10.3390/cancers16213691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Minerals constitute only 5% of the typical human diet but are vital for health and functionality. Copper, a trace element, is absorbed by the human gut at 30-40% from diets typical of industrialized countries. The liver produces metallothioneins, which store copper. Copper is crucial for mitochondrial respiration, pigmentation, iron transport, antioxidant defense, hormone production, and extracellular matrix biosynthesis. Copper deficiency, often caused by mutations in the ATP7A gene, results in Menkes disease, an X-linked recessive disorder. On the contrary, Wilson disease is characterized by toxic copper accumulation. Cuproptosis, a unique form of cell death regulated by copper, is a subtype of necrosis induced by enhanced mitochondrial metabolism and intracellular copper accumulation. This process can reduce the malignant potential of tumor cells by inhibiting glucose metabolism. Therapeutically, copper and its complexes have shown efficacy in malignancy treatments. The disruption of copper homeostasis and excessive cuproplasia are significant in colorectal cancer development and metastasis. Therefore, manipulating copper status presents a potential therapeutic target for colorectal cancer, using copper chelators to inhibit copper formation or copper ion carriers to promote cuproptosis. This review highlights the role of copper in human physiology and pathology, emphasizing its impact on colorectal cancer and potential therapeutic strategies. Future AI-based approaches are anticipated to accelerate the development of new compounds targeting cuproptosis and copper disruption in colorectal cancer.
Collapse
Affiliation(s)
| | - Adam Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland;
| |
Collapse
|
45
|
Yao Y, Chen Y, Zhou C, Zhang Q, He X, Dong K, Yang C, Chu B, Qian Z. Bioorthogonal chemistry-based prodrug strategies for enhanced biosafety in tumor treatments: current progress and challenges. J Mater Chem B 2024; 12:10818-10834. [PMID: 39352785 DOI: 10.1039/d4tb01413h] [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: 10/04/2024]
Abstract
Cancer is a significant global health challenge, and while chemotherapy remains a widely used treatment, its non-specific toxicity and broad distribution can lead to systemic side effects and limit its effectiveness against tumors. Therefore, the development of safer chemotherapy alternatives is crucial. Prodrugs hold great promise, as they remain inactive until they reach the cancer site, where they are selectively activated by enzymes or specific factors, thereby reducing side effects and improving targeting. However, subtle differences in the microenvironments between tumors and normal tissue may still result in unintended cytotoxicity. Bioorthogonal reactions, known for their selectivity and precision without interfering with natural biochemical processes, are gaining attention. When combined with prodrug strategies, these reactions offer the potential to create highly effective chemotherapy drugs. This review examines the safety and efficacy of prodrug strategies utilizing various bioorthogonal reactions in cancer treatment.
Collapse
Affiliation(s)
- Yongchao Yao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
- Precision Medicine Translational Research Center (PMTRC), West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ying Chen
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China
| | - Chang Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Quanzhi Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Xun He
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Kai Dong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Chengli Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Bingyang Chu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Zhiyong Qian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
46
|
Abreu K, Viana JR, Oliveira Neto JG, Dias TG, Reis AS, Lage MR, da Silva LM, de Sousa FF, dos Santos AO. Exploring Thermal Stability, Vibrational Properties, and Biological Assessments of Dichloro(l-histidine)copper(II): A Combined Theoretical and Experimental Study. ACS OMEGA 2024; 9:43488-43502. [PMID: 39493995 PMCID: PMC11525524 DOI: 10.1021/acsomega.4c05029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 11/05/2024]
Abstract
Dichloro(l-histidine)copper(II) crystal ([Cu(l-His)Cl2] complex) was obtained by the slow evaporation method and characterized concerning its thermal stability, phase transformations, and electronic and vibrational properties. X-ray diffraction (XRPD) confirmed that this complex crystallizes with an orthorhombic structure (P212121 space group). Thermal analyses (TG and DTA) demonstrate stability from ambient temperature up to 460 K, followed by a phase transition from the orthorhombic structure to the amorphous form around 465 K, as confirmed by temperature-dependent XRPD studies. The active modes in Fourier transform infrared (FT-IR) and Raman spectroscopy spectra were suitably assigned via density functional theory (DFT) calculations. Additionally, Hirshfeld surface analysis uncovered the prominence of Cl···H, O···H, and H···H interactions as the primary intermolecular forces within the crystal structure. The antimicrobial activity of the [Cu(l-His)Cl2] complex was investigated, demonstrating significant efficacy against Gram-positive bacteria (Staphylococcus aureus), Gram-negative bacteria (Pseudomonas aeruginosa), and fungi (Candida albicans). The minimum inhibitory concentration and cell viability tests showed that the complex inhibits the growth of S. aureus bacteria at a concentration of 1.5 μM without causing damage to the human cell line. The pharmacokinetic parameters corroborate the other tested parameters and highlight the [Cu(l-His)Cl2] complex as a promising alternative for future clinical trials and medicinal applications. The alignment of the pharmacokinetic parameters with other tested criteria highlights the potential of the [Cu(l-His)Cl2] complex as a promising candidate for future clinical studies.
Collapse
Affiliation(s)
- Kamila
R. Abreu
- Center
for Sciences of Imperatriz, Federal University
of Maranhao (UFMA), 65900-410 Imperatriz, MA, Brazil
| | - Jailton R. Viana
- Center
for Sciences of Imperatriz, Federal University
of Maranhao (UFMA), 65900-410 Imperatriz, MA, Brazil
| | - João G. Oliveira Neto
- Center
for Sciences of Imperatriz, Federal University
of Maranhao (UFMA), 65900-410 Imperatriz, MA, Brazil
| | - Tatielle G. Dias
- Center
for Sciences of Imperatriz, Federal University
of Maranhao (UFMA), 65900-410 Imperatriz, MA, Brazil
| | - Aramys S. Reis
- Center
for Sciences of Imperatriz, Federal University
of Maranhao (UFMA), 65900-410 Imperatriz, MA, Brazil
| | - Mateus R. Lage
- Center
for Sciences of Imperatriz, Federal University
of Maranhao (UFMA), 65900-410 Imperatriz, MA, Brazil
| | - Luzeli M. da Silva
- Center
for Sciences of Imperatriz, Federal University
of Maranhao (UFMA), 65900-410 Imperatriz, MA, Brazil
| | - Francisco F. de Sousa
- Institute
of Exact and Natural Sciences, Federal University
of Para (UFPA), 66075-110 Belem, PA, Brazil
| | - Adenilson O. dos Santos
- Center
for Sciences of Imperatriz, Federal University
of Maranhao (UFMA), 65900-410 Imperatriz, MA, Brazil
| |
Collapse
|
47
|
Zhu C, Li J, Sun W, Li D, Wang Y, Shen XC. Signaling Mechanism of Cuproptosis Activating cGAS-STING Immune Pathway. JACS AU 2024; 4:3988-3999. [PMID: 39483232 PMCID: PMC11522904 DOI: 10.1021/jacsau.4c00712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/12/2024] [Accepted: 09/12/2024] [Indexed: 11/03/2024]
Abstract
Copper-mediated programmed cell death, which influences the regulation of tumor progression, is an effective approach for antitumor molecular therapy. Unlike apoptosis, copper complex-induced cuproptosis by lipid-acylated protein aggregation triggers the mitochondrial proteotoxic stress response, which could be associated with immunomodulation. However, it remains a great challenge to understand the distinctive molecular mechanisms that presumably activate immunity by cuproptosis. Here, the new nonlabeling fluorescent molecular tools of Cu-DPPZ-Py+ and Cu-DPPZ-Ph are synthesized and used to investigate the differential immune signaling mechanisms induced by copper-mediated cuproptosis or apoptosis. With Cu-DPPZ-Py+ and Cu-Elesclomol, there is strong evidence that the triggering cuproptosis significantly drives mitochondrial DNA (mtDNA) release to activate innate immunity via cyclic GMP-AMP synthase-stimulation of interferon genes (cGAS-STING), which can improve T cell antitumor immunity in vivo. By contrast, it is observed that Cu-DPPZ-Ph treated tumor cells could release intracellular caspase-3, resulting in apoptosis-associated immunosuppression. This study supports insights into how cuproptosis bridges cGAS-STING immune pathways, contributing to the development of cuproptosis-based antitumor immunotherapy.
Collapse
Affiliation(s)
- Chengyuan Zhu
- 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, Guilin 541004, China
| | - Jialiang Li
- 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, Guilin 541004, China
| | - Wanying Sun
- 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, Guilin 541004, China
| | - Desheng Li
- 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, Guilin 541004, China
| | - Yiliang Wang
- 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, Guilin 541004, China
| | - Xing-Can Shen
- 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, Guilin 541004, China
| |
Collapse
|
48
|
Lin Y, Lin W, Lai Y, Chen Y. Trends of Copper Homeostasis in Neoplasms: A Systemic Bibliometric from 2013 to 2023. Biol Trace Elem Res 2024:10.1007/s12011-024-04413-z. [PMID: 39379668 DOI: 10.1007/s12011-024-04413-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 10/05/2024] [Indexed: 10/10/2024]
Abstract
Copper dysregulation is pivotal in tumorigenesis. This study aimed to elucidate the knowledge framework of copper homeostasis within neoplasms utilizing bibliometric analysis. Publications about copper homeostasis in neoplasms from 2013 to 2023 were retrieved from the Web of Science Core Collection and PubMed databases. The bibliometric analysis was conducted using the RStudio, the CiteSpace software, the VOSviewer software, and a bibliometric analysis platform. A total of 1701 articles were indexed from 73 countries and regions. China has emerged as the leading country with a publication proportion of 32.45%, followed by the USA (10.35%) and India (8.41%). Significant collaborations have been conducted among Chinese academic institutions. The Journal of Inorganic Biochemistry emerged as the most prominent journal. Among the 4841 keywords extracted from 671 journals, concepts of cell death, positron emission tomography, and tumor microenvironment emerged as the most significant hotspots. This bibliometric study reviewed significant academic development and synthesized research trends on copper homeostasis in neoplasms, providing a valuable reference for scholars.
Collapse
Affiliation(s)
- Yuhai Lin
- Department of General Surgery, Shantou Central Hospital, Shantou, China
| | - Wenhao Lin
- Department of Part-Time Master of Business Administration, Antai College of Economics and Management, Shanghai Jiao Tong University, Shanghai, China
| | - Yucheng Lai
- Department of Anesthesiology, Guangxi Hospital Division of the First Affiliated Hospital, Sun Yat-Sen University, Nanning, China
| | - Yuqiang Chen
- Department of General Surgery, Gaozhou People's Hospital, Gaozhou, China.
| |
Collapse
|
49
|
Miglioli F, De Franco M, Bartoli J, Scaccaglia M, Pelosi G, Marzano C, Rogolino D, Gandin V, Carcelli M. Anticancer activity of new water-soluble sulfonated thiosemicarbazone copper(II) complexes targeting disulfide isomerase. Eur J Med Chem 2024; 276:116697. [PMID: 39047610 DOI: 10.1016/j.ejmech.2024.116697] [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: 04/30/2024] [Revised: 06/26/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Copper complexes have shown promising anticancer properties, but they are often poorly soluble in aqueous solutions, thus limiting their possible medical developments and applications. We have recently isolated some copper(II) complexes with salicylaldehyde thiosemicarbazone ligands exhibiting remarkable nanomolar cytotoxic activity, but in vivo tests evidenced several difficulties related to their poor solubility. To overcome these limitations and increase solubility in aqueous solution, herein we report the synthetic strategy that led to the introduction of the sulfonic group on the ligands, then separated as salts (NaH2L1 - NaH2L5), as well as the synthesis and characterization of the related copper(II) complexes. The characterization of the complexes is completed by the analysis of the structures obtained by X-rays diffraction on single crystals on the species [Cu(HL5)(H2O)]2.2H2O, [Cu(HL2)(H2O)2].2H2O, and [Cu(HL1)(H2O]2.2H2O. While the uncoordinated ligands do not affect cancer cell viability, copper(II) complexes exhibit low to sub-micromolar cytotoxic activity, which is maintained in 3D (HCT-15 and 2008) spheroidal models of cancer cell. Notably, copper(II) complexes were selective towards cancer cells, showing high selectivity indexes. Investigations focused on elucidating the mechanism of action evidenced the protein disulfide-isomerase as an innovative molecular target for this class of water-soluble copper(II) complexes. Finally, preliminary in vivo experiments performed with the most representative derivative in the murine Lewis Lung Carcinoma, highlight its significant antitumor efficacy and better tolerability profile with respect to the reference metallodrug, suggesting for this sulfonated Cu(II) complex a potential clinical relevance.
Collapse
Affiliation(s)
- Francesca Miglioli
- Department of Chemistry, Life Sciences, Environmental Sustainability, University of Parma, Parco Area Delle Scienze 11/A, 43124, Parma, Italy
| | - Michele De Franco
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131, Padova, Italy
| | - Jennifer Bartoli
- Department of Chemistry, Life Sciences, Environmental Sustainability, University of Parma, Parco Area Delle Scienze 11/A, 43124, Parma, Italy
| | - Mirco Scaccaglia
- Department of Chemistry, Life Sciences, Environmental Sustainability, University of Parma, Parco Area Delle Scienze 11/A, 43124, Parma, Italy
| | - Giorgio Pelosi
- Department of Chemistry, Life Sciences, Environmental Sustainability, University of Parma, Parco Area Delle Scienze 11/A, 43124, Parma, Italy
| | - Cristina Marzano
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131, Padova, Italy
| | - Dominga Rogolino
- Department of Chemistry, Life Sciences, Environmental Sustainability, University of Parma, Parco Area Delle Scienze 11/A, 43124, Parma, Italy
| | - Valentina Gandin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131, Padova, Italy.
| | - Mauro Carcelli
- Department of Chemistry, Life Sciences, Environmental Sustainability, University of Parma, Parco Area Delle Scienze 11/A, 43124, Parma, Italy.
| |
Collapse
|
50
|
Ren H, Qi F, Zhao Y, Labidi A, Miao Z. Synthesis, Crystal Structure and Antifungal Activity of ( E)-1-(4-Methylbenzylidene)-4-(3-Isopropylphenyl) Thiosemicarbazone: Quantum Chemical and Experimental Studies. Molecules 2024; 29:4702. [PMID: 39407629 PMCID: PMC11477955 DOI: 10.3390/molecules29194702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
A novel (E)-1-(4-methylbenzylidene)-4-(3-isopropylphenyl) thiosemicarbazone was synthesized in a one-pot four-step synthetic route. Fourier transform infrared spectroscopy (FTIR), 1H and 13C nuclear magnetic resonances (NMR), single-crystal X-ray diffraction, and UV-visible absorption spectroscopy were utilized to confirm the successful preparation of the title compound. Single-crystal data indicated that the intramolecular hydrogen bond N(3)-H(3)···N(1) and intermolecular hydrogen bond N(2)-H(2)···S(1) (1 - x, 1 - y, 1 - z) existed in the crystal structure and packing of the title compound. Besides the covalent interaction, the non-covalent weak intramolecular hydrogen bond N(3)-H(3)···N(1) discussed by atoms in molecules (AIM) theory also functioned in maintaining the title compound's crystal structure. The strong intermolecular hydrogen bond N(2)-H(2)···S(1) (1 - x, 1 - y, 1 - z) discussed by Hirshfeld surface analysis played a major role in maintaining the title compound's crystal packing. The local maximum and minimum electrostatic potential of the title compound was predicted by electrostatic potential (ESP) analysis. The UV-visible spectra and HOMO-LUMO analysis revealed that the title compound has a low ΔEHOMO-LUMO energy gap (3.86 eV), which implied its high chemical reactivity due to the easy occurrence of charge transfer interactions within the molecule. Molecular docking and in vitro antifungal assays evidenced that its antifungal activity is comparable to the reported pyrimethanil, indicating its usage as a potential candidate for future antifungal drugs.
Collapse
Affiliation(s)
- Haitao Ren
- Technological Institute of Materials & Energy Science (TIMES), Xijing University, Xi’an 710123, China
| | - Fan Qi
- State Key Laboratory of Medicinal Chemical, College of Pharmacy, Nankai University, Tianjin 300071, China
| | - Yuzhen Zhao
- Technological Institute of Materials & Energy Science (TIMES), Xijing University, Xi’an 710123, China
| | - Abdelkader Labidi
- School of Environmental Science and Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China
| | - Zongcheng Miao
- Technological Institute of Materials & Energy Science (TIMES), Xijing University, Xi’an 710123, China
- School of Artificial Intelligence, Optics and Electronics (iOPEN), Northwestern Polytechnical University, Xi’an 710072, China
| |
Collapse
|