1
|
Tabrizi L, M. Jones A, Romero-Canelon I, Erxleben A. Multiaction Pt(IV) Complexes: Cytotoxicity in Ovarian Cancer Cell Lines and Mechanistic Studies. Inorg Chem 2024; 63:14958-14968. [PMID: 39083592 PMCID: PMC11323244 DOI: 10.1021/acs.inorgchem.4c01586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 08/02/2024]
Abstract
Ovarian cancer has the worst case-to-fatality ratio of all gynecologic malignancies. The main reasons for the high mortality rate are relapse and the development of chemoresistance. In this paper, the cytotoxic activity of two new multiaction platinum(IV) derivatives of cisplatin and oxaliplatin in a panel of ovarian cancer cells is reported. Cis,cis,trans-[Pt(NH3)2Cl2(IPA)(DCA)] (1) and trans-[Pt(DACH)(OX)(IPA)(DCA)] (2) (IPA = indole-3-propionic acid, DCA = dichloroacetate, DACH = 1R,2R-1,2-diaminocyclohexane, OX = oxalate) were synthesized and characterized by elemental analysis, ESI-MS, FT-IR, and 1H, 13C, and195Pt NMR spectroscopy. The biological activity was evaluated in A2780, PEA1, PEA2, SKOV3, SW626, and OVCAR3 cells. Both complexes are potent cytotoxins. Remarkably, complex 2 is 14 times more active in OVCAR3 cells than cisplatin and is able to overcome cisplatin resistance in PEA2 and A2780cis cells, which are models of post-treatment patient-developed and laboratory-induced resistance. This complex also shows activity in 3D cancer models of the A2780 cells. Mechanistic studies revealed that the complexes induce apoptosis via DNA damage and ROS generation.
Collapse
Affiliation(s)
- Leila Tabrizi
- School
of Biological and Chemical Sciences, University
of Galway, Galway H91 TK33, Ireland
- School
of Chemical Sciences, Dublin City University, Dublin D09W6Y4, Ireland
| | - Alan M. Jones
- School
of Pharmacy, University of Birmingham, Birmingham B15 2TT, U.K.
| | - Isolda Romero-Canelon
- School
of Pharmacy, University of Birmingham, Birmingham B15 2TT, U.K.
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
| | - Andrea Erxleben
- School
of Biological and Chemical Sciences, University
of Galway, Galway H91 TK33, Ireland
- Synthesis
and Solid State Pharmaceutical Centre (SSPC), Limerick V94 T9PX, Ireland
| |
Collapse
|
2
|
Yang B, Lin Y, Huang Y, Shen YQ, Chen Q. Thioredoxin (Trx): A redox target and modulator of cellular senescence and aging-related diseases. Redox Biol 2024; 70:103032. [PMID: 38232457 PMCID: PMC10827563 DOI: 10.1016/j.redox.2024.103032] [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: 10/14/2023] [Revised: 12/03/2023] [Accepted: 01/04/2024] [Indexed: 01/19/2024] Open
Abstract
Thioredoxin (Trx) is a compact redox-regulatory protein that modulates cellular redox state by reducing oxidized proteins. Trx exhibits dual functionality as an antioxidant and a cofactor for diverse enzymes and transcription factors, thereby exerting influence over their activity and function. Trx has emerged as a pivotal biomarker for various diseases, particularly those associated with oxidative stress, inflammation, and aging. Recent clinical investigations have underscored the significance of Trx in disease diagnosis, treatment, and mechanistic elucidation. Despite its paramount importance, the intricate interplay between Trx and cellular senescence-a condition characterized by irreversible growth arrest induced by multiple aging stimuli-remains inadequately understood. In this review, our objective is to present a comprehensive and up-to-date overview of the structure and function of Trx, its involvement in redox signaling pathways and cellular senescence, its association with aging and age-related diseases, as well as its potential as a therapeutic target. Our review aims to elucidate the novel and extensive role of Trx in senescence while highlighting its implications for aging and age-related diseases.
Collapse
Affiliation(s)
- Bowen Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Yumeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Yibo Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Ying-Qiang Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
3
|
Pivarcsik T, Kiss MA, Rapuš U, Kljun J, Spengler G, Frank É, Turel I, Enyedy ÉA. Organometallic Ru(II), Rh(III) and Re(I) complexes of sterane-based bidentate ligands: synthesis, solution speciation, interaction with biomolecules and anticancer activity. Dalton Trans 2024; 53:4984-5000. [PMID: 38406993 DOI: 10.1039/d3dt04138g] [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: 02/27/2024]
Abstract
In this study, we present the synthesis, characterization and in vitro cytotoxicity of six organometallic [Ru(II)(η6-p-cymene)(N,N)Cl]Cl, [Rh(III)(η5-C5Me5)(N,N)Cl]Cl and [Re(I)(CO)3(N,N)Cl] complexes, in which the (N,N) ligands are sterane-based 2,2'-bipyridine derivatives (4-Me-bpy-St-OH, 4-Ph-bpy-St-OH). The solution chemical behavior of the ligands and the complexes was explored by UV-visible spectrophotometry and 1H NMR spectroscopy. The ligands and their Re(I) complexes are neutral at pH = 7.40; this contributes to their highly lipophilic character (log D7.40 > +3). The Ru(II) and Rh(III) half-sandwich complexes are much more hydrophilic, and this property is greatly affected by the actual chloride ion content of the medium. The half-sandwich Ru and Rh complexes are highly stable in 30% (v/v) DMSO/water (<5% dissociation at pH = 7.40); this is further increased in water. The Rh(III)(η5-C5Me5) complexes were characterized by higher water/chloride exchange and pKa constants compared to their Ru(II)(η6-p-cymene) counterparts. The Re(I)(CO)3 complexes are also stable in solution over a wide pH range (2-12) without the release of the bidentate ligand; only the chlorido co-ligand can be replaced with OH- at higher pH values. A comprehensive discussion of the binding affinity of the half-sandwich Ru(II) and Rh(III) complexes toward human serum albumin and calf-thymus DNA is also provided. The Ru(II)(η6-p-cymene) complexes interact with human serum albumin via intermolecular forces, while for the Rh(III)(η5-C5Me5) complexes the coordinative binding mode is suggested as well. They are also able to interact with calf-thymus DNA, most likely via the coordination of the guanine nitrogen. The Ru(II)(η6-p-cymene) complexes were found to be the most promising among the tested compounds as they exhibited moderate-to-strong cytotoxic activity (IC50 = 3-11 μM) in LNCaP as well as in PC3 prostate cells in an androgen receptor-independent manner. They were also significantly cytotoxic in breast and colon adenocarcinoma cancer cell lines and showed good selectivity for cancer cells.
Collapse
Affiliation(s)
- Tamás Pivarcsik
- MTA-SZTE Lendület Functional Metal Complexes Research Group, Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8., H-6720 Szeged, Hungary.
- Department of Molecular and Analytical Chemistry, University of Szeged, Dóm tér 7-8., H-6720 Szeged, Hungary
| | - Márton A Kiss
- Department of Molecular and Analytical Chemistry, University of Szeged, Dóm tér 7-8., H-6720 Szeged, Hungary
| | - Uroš Rapuš
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Jakob Kljun
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Gabriella Spengler
- MTA-SZTE Lendület Functional Metal Complexes Research Group, Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8., H-6720 Szeged, Hungary.
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Éva Frank
- Department of Molecular and Analytical Chemistry, University of Szeged, Dóm tér 7-8., H-6720 Szeged, Hungary
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Éva A Enyedy
- MTA-SZTE Lendület Functional Metal Complexes Research Group, Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8., H-6720 Szeged, Hungary.
- Department of Molecular and Analytical Chemistry, University of Szeged, Dóm tér 7-8., H-6720 Szeged, Hungary
| |
Collapse
|
4
|
Sumithaa C, Gajda-Morszewski P, Ishaniya W, Khamrang T, Velusamy M, Bhuvanesh N, Brindell M, Mazuryk O, Ganeshpandian M. Design of an anticancer organoruthenium complex as the guest and polydiacetylene-coated fluorogenic nanocarrier as the host: engineering nanocarrier using ene-yne conjugation for sustained guest release, enhanced anticancer activity and reduced in vivo toxicity. Dalton Trans 2024; 53:966-985. [PMID: 38054338 DOI: 10.1039/d3dt03358a] [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: 12/07/2023]
Abstract
Despite the enormous efforts made over the past two decades to develop metallodrugs and nanocarriers for metallodrug delivery, there are still few precise strategies that aim to optimize the design of both metallodrugs and metallodrug carriers jointly in a concerted effort. In this work, three half-sandwich ruthenium(II) complexes with pyridylimidazo[1,5-a]pyridine ligand functionalized with polycyclic aromatic moiety (Ru(nap), Ru(ant), Ru(pyr)) are evaluated as possible anticancer candidates and polydiacetylene (PDA)-coated amino-functionalized mesoporous silica nanoparticles (AMSNs) are designed as a functional nanocarrier for drug delivery. Ru(pyr) exhibits higher cytotoxicity in HT-29 colorectal cancer cells compared to other complexes and cis-platin, but it does not exhibit better cellular uptake. Ru(pyr) is found to be preferentially accumulated in plasma, mitochondria, and ER-Golgi membrane. The complex induces cell cycle arrest in the G0/G1 phase, while higher concentrations cause programmed cell death via apoptosis. Ru(pyr) influences cancer cell adhesion property and acts as an antioxidant in HT-29 cells. In order to modulate the anticancer potency of Ru(pyr), AMSNs are used to encapsulate the complex, and then diacetylene self-assembly is allowed to deposit on the surface of the nanoparticles. Subsequently, the nanoparticles undergo topopolymerization, which results in π-conjugated PDA-Ru(pyr)@AMSNs. Owing to the ene-yne polymeric skeleton in the backbone, the non-fluorescent AMSNs turn into red-emissive particles, which are exploited for cell imaging applications. The release profile analysis reveals that such a π-conjugated polymer prevents the premature release of the complex from porous silica nanoparticles with the accelerated release of the complex in an acidic medium compared to physiological conditions. The PDA gatekeepers have also been proven to enhance the cellular internalization of Ru(pyr) with slow continuous release from the nanoformulation. Zebrafish embryo toxicity analysis suggests that the PDA-coated nanocarriers could be suitable candidates for in vivo investigations.
Collapse
Affiliation(s)
- Chezhiyan Sumithaa
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| | - Przemyslaw Gajda-Morszewski
- Faculty of Chemistry, Department of Inorganic Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Cracow, Poland.
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Łojasiewicza 11, 30-348 Cracow, Poland
| | - Wickneswaran Ishaniya
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| | - Themmila Khamrang
- Department of Chemistry, Dhanamanjuri University, Manipur 795001, India
| | - Marappan Velusamy
- Department of Chemistry, North Eastern Hill University, Shillong 793022, India
| | - Nattamai Bhuvanesh
- X-ray Diffraction Lab, Department of Chemistry, Texas A&M University, College Station, TX 77842, USA
| | - Malgorzata Brindell
- Faculty of Chemistry, Department of Inorganic Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Cracow, Poland.
| | - Olga Mazuryk
- Faculty of Chemistry, Department of Inorganic Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Cracow, Poland.
| | - Mani Ganeshpandian
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
5
|
Balsa LM, Solernó LM, Rodriguez MR, Parajón-Costa BS, Gonzalez-Baró AC, Alonso DF, Garona J, León IE. Cu(II)-acylhydrazone complex, a potent and selective antitumor agent against human osteosarcoma: Mechanism of action studies over in vitro and in vivo models. Chem Biol Interact 2023; 384:110685. [PMID: 37666443 DOI: 10.1016/j.cbi.2023.110685] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/05/2023] [Accepted: 08/26/2023] [Indexed: 09/06/2023]
Abstract
Osteosarcoma (OS) is a frequent bone cancer, affecting largely children and young adults. Cisplatin (CDDP) has been efficacious in the treatment of different cancer such us OS but the development of chemoresistance and important side effects leading to therapeutic failure. Novel therapies including copper compounds have shown to be potentially effective as anticancer drugs and one alternative to usually employed platinum compounds. The goal of this work is the evaluation of the in vitro and in vivo antitumoral activity and dilucidate the molecular target of a Cu(II) cationic complex containing a tridentate hydrazone ligand, CuHL for short, H2L=N'-'-(2-hydroxy-3-methoxybenzylidene)thiophene-2-carbohydrazide, against human OS MG-63 cells. Anticancer activity on MG-63 cell line was evaluated in OS monolayer and spheroids. CuHL significantly impaired cell viability in both models (IC50 2D: 2.1 ± 0.3 μM; 3D: 9.1 ± 1.0 μM) (p < 0.001). Additional cell studies demonstrated the copper compound inhibits cell proliferation and conveys cells to apoptosis, determined by flow cytometry. CuHL showed a great genotoxicity, evaluated by comet assay. Proteomic analysis by Orbitrap Mass Spectometry identified 27 differentially expressed proteins: 17 proteins were found overexpressed and 10 underexpressed in MG-63 cells after the CuHL treatment. The response to unfolded protein was the most affected biological process. In addition, in vivo antitumor effects of the compound were evaluated on human OS tumors xenografted in nude mice. CuHL treatment, at a dose of 2 mg/kg i.p., given three times/week for one month, significantly inhibited the progression of OS xenografts and was associated to a reduction in mitotic index and to an increment of tumor necrosis (p < 0.01). Administration of standard-of-care cytotoxic agent CDDP, following the same treatment schedule as CuHL, failed to impair OS growth and progression.
Collapse
Affiliation(s)
- Lucia 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
| | - Luisina M Solernó
- Centro de Oncología Molecular y Traslacional (COMTra), Universidad Nacional de Quilmes, Argentina; Centro de Medicina Traslacional (Unidad 6), Hospital de Alta Complejidad en Red El Cruce "Dr. Néstor Carlos Kirchner" S.A.M.I.C, Argentina
| | - Maria R Rodriguez
- 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
| | - Beatriz S Parajón-Costa
- 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
| | - Ana C Gonzalez-Baró
- 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
| | - Daniel F Alonso
- Centro de Oncología Molecular y Traslacional (COMTra), Universidad Nacional de Quilmes, Argentina
| | - Juan Garona
- Centro de Oncología Molecular y Traslacional (COMTra), Universidad Nacional de Quilmes, Argentina; Centro de Medicina Traslacional (Unidad 6), Hospital de Alta Complejidad en Red El Cruce "Dr. Néstor Carlos Kirchner" S.A.M.I.C, 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
|
6
|
Salmain M, Gaschard M, Baroud M, Lepeltier E, Jaouen G, Passirani C, Vessières A. Thioredoxin Reductase and Organometallic Complexes: A Pivotal System to Tackle Multidrug Resistant Tumors? Cancers (Basel) 2023; 15:4448. [PMID: 37760418 PMCID: PMC10526406 DOI: 10.3390/cancers15184448] [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: 07/26/2023] [Revised: 09/01/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Cancers classified as multidrug-resistant (MDR) are a family of diseases with poor prognosis despite access to increasingly sophisticated treatments. Several mechanisms explain these resistances involving both tumor cells and their microenvironment. It is now recognized that a multi-targeting approach offers a promising strategy to treat these MDR tumors. Inhibition of thioredoxin reductase (TrxR), a key enzyme in maintaining redox balance in cells, is a well-identified target for this approach. Auranofin was the first inorganic gold complex to be described as a powerful inhibitor of TrxR. In this review, we will first recall the main results obtained with this metallodrug. Then, we will focus on organometallic complexes reported as TrxR inhibitors. These include gold(I), gold(III) complexes and metallocifens, i.e., organometallic complexes of Fe and Os derived from tamoxifen. In these families of complexes, similarities and differences in the molecular mechanisms of TrxR inhibition will be highlighted. Finally, the possible relationship between TrxR inhibition and cytotoxicity will be discussed and put into perspective with their mode of action.
Collapse
Affiliation(s)
- Michèle Salmain
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire (IPCM), 4 Place Jussieu, F-75005 Paris, France; (M.S.); (M.G.); (G.J.); (A.V.)
| | - Marie Gaschard
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire (IPCM), 4 Place Jussieu, F-75005 Paris, France; (M.S.); (M.G.); (G.J.); (A.V.)
| | - Milad Baroud
- Micro & Nanomedecines Translationnelles (MINT), University of Angers, Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, F-49000 Angers, France; (M.B.); (E.L.)
| | - Elise Lepeltier
- Micro & Nanomedecines Translationnelles (MINT), University of Angers, Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, F-49000 Angers, France; (M.B.); (E.L.)
| | - Gérard Jaouen
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire (IPCM), 4 Place Jussieu, F-75005 Paris, France; (M.S.); (M.G.); (G.J.); (A.V.)
| | - Catherine Passirani
- Micro & Nanomedecines Translationnelles (MINT), University of Angers, Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, F-49000 Angers, France; (M.B.); (E.L.)
| | - Anne Vessières
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire (IPCM), 4 Place Jussieu, F-75005 Paris, France; (M.S.); (M.G.); (G.J.); (A.V.)
| |
Collapse
|
7
|
Kljun J, Rebernik M, Balsa LM, Kladnik J, Rapuš U, Trobec T, Sepčić K, Frangež R, León IE, Turel I. Exploring pta Alternatives in the Development of Ruthenium-Arene Anticancer Compounds. Molecules 2023; 28:molecules28062499. [PMID: 36985471 PMCID: PMC10058425 DOI: 10.3390/molecules28062499] [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: 01/31/2023] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/30/2023] Open
Abstract
Organoruthenium pyrithione (1-hydroxypyridine-2-thione) complexes have been shown in our recent studies to be a promising family of compounds for development of new anticancer drugs. The complex [(η6-p-cymene)Ru(pyrithionato)(pta)]PF6 contains phosphine ligand pta (1,3,5-triaza-7-phosphaadamantane) as a functionality that improves the stability of the complex and its aqueous solubility. Here, we report our efforts to find pta alternatives and discover new structural elements to improve the biological properties of ruthenium anticancer drugs. The pta ligand was replaced by a selection of phosphine, phosphite, and arsine ligands to identify new functionalities, leading to improvement in inhibitory potency towards enzyme glutathione S-transferase. In addition, cytotoxicity in breast, bone, and colon cancers was investigated.
Collapse
Affiliation(s)
- Jakob Kljun
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Mihaela Rebernik
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - 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
| | - Jerneja Kladnik
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Uroš Rapuš
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Tomaž Trobec
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, SI-1000 Ljubljana, Slovenia
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty, University of LjubljanaJamnikarjeva 101, SI-1000 Ljubljana, Slovenia
| | - Robert Frangež
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, SI-1000 Ljubljana, Slovenia
| | - 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
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
8
|
Sumithaa C, Ganeshpandian M. Half-Sandwich Ruthenium Arene Complexes Bearing Clinically Approved Drugs as Ligands: The Importance of Metal-Drug Synergism in Metallodrug Design. Mol Pharm 2023; 20:1453-1479. [PMID: 36802711 DOI: 10.1021/acs.molpharmaceut.2c01027] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
A novel strategy in metallodrug discovery today is incorporating clinically approved drugs into metal complexes as coordinating ligands. Using this strategy, various drugs have been repurposed to prepare organometallic complexes to overcome the resistance of drugs and to design promising alternatives to currently available metal-based drugs. Notably, the combination of organoruthenium moiety and clinical drug in a single molecule has been shown, in some instances, to enhance pharmacological activity and reduce toxicity in comparison to the parent drug. Thus, for the past two decades, there has been increasing interest in exploiting metal-drug synergism to develop multifunctional organoruthenium drug candidates. Herein, we summarized the recent reports of rationally designed half-sandwich Ru(arene) complexes containing different FDA-approved drugs. This review also focuses on the mode of coordination of drugs, ligand-exchange kinetics, mechanism of action, and structure-activity relationship of organoruthenated complexes containing drugs. We hope this discussion may serve to shed light on future developments in ruthenium-based metallopharmaceuticals.
Collapse
Affiliation(s)
- Chezhiyan Sumithaa
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603 203, India
| | - Mani Ganeshpandian
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603 203, India
| |
Collapse
|
9
|
Catalano A, Mariconda A, Sinicropi MS, Ceramella J, Iacopetta D, Saturnino C, Longo P. Biological Activities of Ruthenium NHC Complexes: An Update. Antibiotics (Basel) 2023; 12:365. [PMID: 36830276 PMCID: PMC9952499 DOI: 10.3390/antibiotics12020365] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Ruthenium N-heterocyclic carbene (NHC) complexes have unique physico-chemical properties as catalysts and a huge potential in medicinal chemistry and pharmacology, exhibiting a variety of notable biological activities. In this review, the most recent studies on ruthenium NHC complexes are summarized, focusing specifically on antimicrobial and antiproliferative activities. Ruthenium NHC complexes are generally active against Gram-positive bacteria, such as Bacillus subtilis, Staphylococcus aureus, Micrococcus luteus, Listeria monocytogenes and are seldom active against Gram-negative bacteria, including Salmonella typhimurium, Pseudomonas aeruginosa and Escherichia coli and fungal strains of Candida albicans. The antiproliferative activity was tested against cancer cell lines of human colon, breast, cervix, epidermis, liver and rat glioblastoma cell lines. Ruthenium NHC complexes generally demonstrated cytotoxicity higher than standard anticancer drugs. Further studies are needed to explore the mechanism of action of these interesting compounds.
Collapse
Affiliation(s)
- Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70126 Bari, Italy
| | | | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Carmela Saturnino
- Department of Science, University of Basilicata, 85100 Potenza, Italy
| | - Pasquale Longo
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| |
Collapse
|
10
|
Albanell-Fernández M, Oltra SS, Orts-Arroyo M, Ibarrola-Villava M, Carrasco F, Jiménez-Martí E, Cervantes A, Castro I, Martínez-Lillo J, Ribas G. RUNAT-BI: A Ruthenium(III) Complex as a Selective Anti-Tumor Drug Candidate against Highly Aggressive Cancer Cell Lines. Cancers (Basel) 2022; 15:cancers15010069. [PMID: 36612065 PMCID: PMC9817854 DOI: 10.3390/cancers15010069] [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: 11/08/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Ruthenium compounds have demonstrated promising activity in different cancer types, overcoming several limitations of platinum-based drugs, yet their global structure-activity is still under debate. We analyzed the activity of Runat-BI, a racemic Ru(III) compound, and of one of its isomers in eight tumor cell lines of breast, colon and gastric cancer as well as in a non-tumoral control. Runat-BI was prepared with 2,2'-biimidazole and dissolved in polyethylene glycol. We performed assays of time- and dose-dependent viability, migration, proliferation, and expression of pro- and antiapoptotic genes. Moreover, we studied the growth rate and cell doubling time to correlate it with the apoptotic effect of Runat-BI. As a racemic mixture, Runat-BI caused a significant reduction in the viability and migration of three cancer cell lines from colon, gastric and breast cancer, all of which displayed fast proliferation rates. This compound also demonstrated selectivity between tumor and non-tumor lines and increased proapoptotic gene expression. However, the isolated isomer did not show any effect. Racemic Runat-BI is a potential drug candidate for treatment of highly aggressive tumors. Further studies should be addressed at evaluating the role of the other isomer, for a more precise understanding of its antitumoral potential and mechanism of action.
Collapse
Affiliation(s)
- Marta Albanell-Fernández
- INCLIVA Biomedical Research Institute, Hospital Clínico Universitario Valencia, University of Valencia, 46010 Valencia, Spain
| | - Sara S. Oltra
- INCLIVA Biomedical Research Institute, Hospital Clínico Universitario Valencia, University of Valencia, 46010 Valencia, Spain
| | - Marta Orts-Arroyo
- Instituto de Ciencia Molecular (ICMol)/Departament de Química Inorgànica, University of Valencia, 46980 Valencia, Spain
| | - Maider Ibarrola-Villava
- INCLIVA Biomedical Research Institute, Hospital Clínico Universitario Valencia, University of Valencia, 46010 Valencia, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), 28029 Madrid, Spain
| | - Fany Carrasco
- INCLIVA Biomedical Research Institute, Hospital Clínico Universitario Valencia, University of Valencia, 46010 Valencia, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), 28029 Madrid, Spain
| | - Elena Jiménez-Martí
- INCLIVA Biomedical Research Institute, Hospital Clínico Universitario Valencia, University of Valencia, 46010 Valencia, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), 28029 Madrid, Spain
- Departament de Bioquímica i Biología Molecular, Facultat de Medicina, University of Valencia, 46010 Valencia, Spain
| | - Andrés Cervantes
- INCLIVA Biomedical Research Institute, Hospital Clínico Universitario Valencia, University of Valencia, 46010 Valencia, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), 28029 Madrid, Spain
| | - Isabel Castro
- Instituto de Ciencia Molecular (ICMol)/Departament de Química Inorgànica, University of Valencia, 46980 Valencia, Spain
| | - José Martínez-Lillo
- Instituto de Ciencia Molecular (ICMol)/Departament de Química Inorgànica, University of Valencia, 46980 Valencia, Spain
- Correspondence: (J.M.-L.); (G.R.); Tel.: +34-9635-44460 (J.M.-L.)
| | - Gloria Ribas
- INCLIVA Biomedical Research Institute, Hospital Clínico Universitario Valencia, University of Valencia, 46010 Valencia, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), 28029 Madrid, Spain
- Correspondence: (J.M.-L.); (G.R.); Tel.: +34-9635-44460 (J.M.-L.)
| |
Collapse
|
11
|
Kladnik J, Dolinar A, Kljun J, Perea D, Grau-Expósito J, Genescà M, Novinec M, Buzon MJ, Turel I. Zinc pyrithione is a potent inhibitor of PL Pro and cathepsin L enzymes with ex vivo inhibition of SARS-CoV-2 entry and replication. J Enzyme Inhib Med Chem 2022; 37:2158-2168. [PMID: 35943189 PMCID: PMC9367663 DOI: 10.1080/14756366.2022.2108417] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Zinc pyrithione (1a), together with its analogues 1b–h and ruthenium pyrithione complex 2a, were synthesised and evaluated for the stability in biologically relevant media and anti-SARS-CoV-2 activity. Zinc pyrithione revealed potent in vitro inhibition of cathepsin L (IC50=1.88 ± 0.49 µM) and PLPro (IC50=0.50 ± 0.07 µM), enzymes involved in SARS-CoV-2 entry and replication, respectively, as well as antiviral entry and replication properties in an ex vivo system derived from primary human lung tissue. Zinc complexes 1b–h expressed comparable in vitro inhibition. On the contrary, ruthenium complex 2a and the ligand pyrithione a itself expressed poor inhibition in mentioned assays, indicating the importance of the selection of metal core and structure of metal complex for antiviral activity. Safe, effective, and preferably oral at-home therapeutics for COVID-19 are needed and as such zinc pyrithione, which is also commercially available, could be considered as a potential therapeutic agent against SARS-CoV-2.
Collapse
Affiliation(s)
- Jerneja Kladnik
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Ana Dolinar
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Jakob Kljun
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - David Perea
- Infectious Diseases Department, Vall d'Hebron Research Institute (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Judith Grau-Expósito
- Infectious Diseases Department, Vall d'Hebron Research Institute (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Meritxell Genescà
- Infectious Diseases Department, Vall d'Hebron Research Institute (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Marko Novinec
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Maria J Buzon
- Infectious Diseases Department, Vall d'Hebron Research Institute (VHIR), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
12
|
Special Issue: "New Diagnostic and Therapeutic Tools against Multidrug-Resistant Tumors (STRATAGEM Special Issue, EU-COST CA17104)". Cancers (Basel) 2022; 14:cancers14225491. [PMID: 36428584 PMCID: PMC9688366 DOI: 10.3390/cancers14225491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 10/31/2022] [Indexed: 11/11/2022] Open
Abstract
Cancer drug resistance, either intrinsic or acquired, often causes treatment failure and increased mortality [...].
Collapse
|
13
|
Kljun J, Pavlič R, Hafner E, Lipec T, Moreno-Da Silva S, Tič P, Turel I, Büdefeld T, Stojan J, Rižner TL. Ruthenium complexes show potent inhibition of AKR1C1, AKR1C2, and AKR1C3 enzymes and anti-proliferative action against chemoresistant ovarian cancer cell line. Front Pharmacol 2022; 13:920379. [PMID: 36034868 PMCID: PMC9403717 DOI: 10.3389/fphar.2022.920379] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/28/2022] [Indexed: 12/04/2022] Open
Abstract
In this study, we present the synthesis, kinetic studies of inhibitory activity toward aldo-keto reductase 1C (AKR1C) enzymes, and anticancer potential toward chemoresistant ovarian cancer of 10 organoruthenium compounds bearing diketonate (1–6) and hydroxyquinolinate (7–10) chelating ligands with the general formula [(η6-p-cymene)Ru(chel)(X)]n+ where chel represents the chelating ligand and X the chlorido or pta ligand. Our studies show that these compounds are potent inhibitors of the AKR enzymes with an uncommon inhibitory mechanism, where two inhibitor molecules bind to the enzyme in a first fast and reversible step and a second slower and irreversible step. The binding potency of each step is dependent on the chemical structure of the monodentate ligands in the metalloinhibitors with the chlorido complexes generally acting as reversible inhibitors and pta complexes as irreversible inhibitors. Our study also shows that compounds 1–9 have a moderate yet better anti-proliferative and anti-migration action on the chemoresistant ovarian cancer cell line COV362 compared to carboplatin and similar effects to cisplatin.
Collapse
Affiliation(s)
- Jakob Kljun
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Renata Pavlič
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Eva Hafner
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tanja Lipec
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Sara Moreno-Da Silva
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
- Faculty of Chemical Sciences, Universidad Complutense de Madrid, Madrid, Spain
| | - Primož Tič
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Iztok Turel
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
- *Correspondence: Iztok Turel, ; Tea Lanišnik Rižner,
| | - Tomaž Büdefeld
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jure Stojan
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tea Lanišnik Rižner
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- *Correspondence: Iztok Turel, ; Tea Lanišnik Rižner,
| |
Collapse
|
14
|
Guo W, Ji T, Deng Y, Liu J, Gou Y, Dong W. Facile synthesis of a glutathione-depleting Cu(II)-half-salamo-based coordination polymer for enhanced chemodynamic therapy. Dalton Trans 2022; 51:11884-11891. [PMID: 35876194 DOI: 10.1039/d2dt01786e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chemodynamic therapy (CDT), utilizing Fenton catalysts to convert intracellular H2O2 into toxic hydroxyl radicals (˙OH) to kill cancer cells, has a wide application prospect in tumor treatment because of its high selectivity. Its anticancer effect, however, is unsatisfactory due to the overexpressed glutathione (GSH). Herein, a GSH-depleting Cu(II)-half-salamo-based coordination polymer (CuCP) was prepared and validated by single crystal X-ray crystallography, Hirshfeld surface analyses and DFT calculations. The Cu(II) ions in the coordination polymer are five-coordinated bearing slightly twisted square pyramidal coordination environments and are bridged by phenoxy and alkoxy groups. After internalization by tumor cells, the CuCP could be biodegraded and reduced by GSH to generate a large amount of Cu(I), simultaneously depleting GSH. Subsequently, the Cu(I) ions interact with H2O2 to generate toxic ˙OH through a Fenton-like reaction to enhance their anticancer efficacy. Our study provides useful insights into designing smarter metal-based anticancer agents to improve the CDT efficiency in cancer therapy.
Collapse
Affiliation(s)
- Wenting Guo
- School of Chemistry and Chemical Engineering, Lanzhou Jiaotong University, Lanzhou, Gansu 730070, China.
| | - Tongxi Ji
- School of Chemistry and Chemical Engineering, Lanzhou Jiaotong University, Lanzhou, Gansu 730070, China.
| | - Yunhu Deng
- School of Chemistry and Chemical Engineering, Lanzhou Jiaotong University, Lanzhou, Gansu 730070, China.
| | - Jia Liu
- School of Chemistry and Chemical Engineering, Lanzhou Jiaotong University, Lanzhou, Gansu 730070, China.
| | - Yantong Gou
- School of Chemistry and Chemical Engineering, Lanzhou Jiaotong University, Lanzhou, Gansu 730070, China.
| | - Wenkui Dong
- School of Chemistry and Chemical Engineering, Lanzhou Jiaotong University, Lanzhou, Gansu 730070, China.
| |
Collapse
|
15
|
Hildebrandt J, Häfner N, Kritsch D, Görls H, Dürst M, Runnebaum IB, Weigand W. Highly Cytotoxic Osmium(II) Compounds and Their Ruthenium(II) Analogues Targeting Ovarian Carcinoma Cell Lines and Evading Cisplatin Resistance Mechanisms. Int J Mol Sci 2022; 23:ijms23094976. [PMID: 35563367 PMCID: PMC9102668 DOI: 10.3390/ijms23094976] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 12/14/2022] Open
Abstract
(1) Background: Ruthenium and osmium complexes attract increasing interest as next generation anticancer drugs. Focusing on structure-activity-relationships of this class of compounds, we report on 17 different ruthenium(II) complexes and four promising osmium(II) analogues with cinnamic acid derivatives as O,S bidentate ligands. The aim of this study was to determine the anticancer activity and the ability to evade platin resistance mechanisms for these compounds. (2) Methods: Structural characterizations and stability determinations have been carried out with standard techniques, including NMR spectroscopy and X-ray crystallography. All complexes and single ligands have been tested for cytotoxic activity on two ovarian cancer cell lines (A2780, SKOV3) and their cisplatin-resistant isogenic cell cultures, a lung carcinoma cell line (A549) as well as selected compounds on three non-cancerous cell cultures in vitro. FACS analyses and histone γH2AX staining were carried out for cell cycle distribution and cell death or DNA damage analyses, respectively. (3) Results: IC50 values show promising results, specifically a high cancer selective cytotoxicity and evasion of resistance mechanisms for Ru(II) and Os(II) compounds. Histone γH2AX foci and FACS experiments validated the high cytotoxicity but revealed diminished DNA damage-inducing activity and an absence of cell cycle disturbance thus pointing to another mode of action. (4) Conclusion: Ru(II) and Os(II) compounds with O,S-bidentate ligands show high cytotoxicity without strong effects on DNA damage and cell cycle, and this seems to be the basis to circumvent resistance mechanisms and for the high cancer cell specificity.
Collapse
Affiliation(s)
- Jana Hildebrandt
- Institut für Anorganische und Analytische Chemie Friedrich-Schiller Universität Jena, Humboldtstraße 8, 07743 Jena, Germany; (J.H.); (H.G.)
- Department of Gynecology, Jena University Hospital—Friedrich-Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (N.H.); (D.K.); (M.D.)
| | - Norman Häfner
- Department of Gynecology, Jena University Hospital—Friedrich-Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (N.H.); (D.K.); (M.D.)
| | - Daniel Kritsch
- Department of Gynecology, Jena University Hospital—Friedrich-Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (N.H.); (D.K.); (M.D.)
| | - Helmar Görls
- Institut für Anorganische und Analytische Chemie Friedrich-Schiller Universität Jena, Humboldtstraße 8, 07743 Jena, Germany; (J.H.); (H.G.)
| | - Matthias Dürst
- Department of Gynecology, Jena University Hospital—Friedrich-Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (N.H.); (D.K.); (M.D.)
| | - Ingo B. Runnebaum
- Department of Gynecology, Jena University Hospital—Friedrich-Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (N.H.); (D.K.); (M.D.)
- Correspondence: (I.B.R.); (W.W.); Tel.: +49-3641-9329101 (I.B.R.); +49-3641-948160 (W.W.)
| | - Wolfgang Weigand
- Institut für Anorganische und Analytische Chemie Friedrich-Schiller Universität Jena, Humboldtstraße 8, 07743 Jena, Germany; (J.H.); (H.G.)
- Correspondence: (I.B.R.); (W.W.); Tel.: +49-3641-9329101 (I.B.R.); +49-3641-948160 (W.W.)
| |
Collapse
|
16
|
Peña Q, Wang A, Zaremba O, Shi Y, Scheeren HW, Metselaar JM, Kiessling F, Pallares RM, Wuttke S, Lammers T. Metallodrugs in cancer nanomedicine. Chem Soc Rev 2022; 51:2544-2582. [PMID: 35262108 DOI: 10.1039/d1cs00468a] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Metal complexes are extensively used for cancer therapy. The multiple variables available for tuning (metal, ligand, and metal-ligand interaction) offer unique opportunities for drug design, and have led to a vast portfolio of metallodrugs that can display a higher diversity of functions and mechanisms of action with respect to pure organic structures. Clinically approved metallodrugs, such as cisplatin, carboplatin and oxaliplatin, are used to treat many types of cancer and play prominent roles in combination regimens, including with immunotherapy. However, metallodrugs generally suffer from poor pharmacokinetics, low levels of target site accumulation, metal-mediated off-target reactivity and development of drug resistance, which can all limit their efficacy and clinical translation. Nanomedicine has arisen as a powerful tool to help overcome these shortcomings. Several nanoformulations have already significantly improved the efficacy and reduced the toxicity of (chemo-)therapeutic drugs, including some promising metallodrug-containing nanomedicines currently in clinical trials. In this critical review, we analyse the opportunities and clinical challenges of metallodrugs, and we assess the advantages and limitations of metallodrug delivery, both from a nanocarrier and from a metal-nano interaction perspective. We describe the latest and most relevant nanomedicine formulations developed for metal complexes, and we discuss how the rational combination of coordination chemistry with nanomedicine technology can assist in promoting the clinical translation of metallodrugs.
Collapse
Affiliation(s)
- Quim Peña
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany.
| | - Alec Wang
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany.
| | - Orysia Zaremba
- BCMaterials, Bld. Martina Casiano, 3rd. Floor, UPV/EHU Science Park, 48940, Leioa, Spain
| | - Yang Shi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany.
| | - Hans W Scheeren
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany.
| | - Josbert M Metselaar
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany.
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany
| | - Roger M Pallares
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany.
| | - Stefan Wuttke
- BCMaterials, Bld. Martina Casiano, 3rd. Floor, UPV/EHU Science Park, 48940, Leioa, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany.
| |
Collapse
|
17
|
Therapeutic strategies to overcome cisplatin resistance in ovarian cancer. Eur J Med Chem 2022; 232:114205. [DOI: 10.1016/j.ejmech.2022.114205] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 12/15/2022]
|
18
|
Pyrithione metal (Cu, Ni, Ru) complexes as photo-catalysts for styrene oxide production. Sci Rep 2021; 11:23810. [PMID: 34893654 PMCID: PMC8664809 DOI: 10.1038/s41598-021-03085-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 11/17/2021] [Indexed: 11/08/2022] Open
Abstract
Selective photochemical oxidation of styrene was performed in an active acetonitrile medium, using H2O2 with or without ultraviolet (UV) light radiation. Pyrithione metal complexes (M-Pth: M = Cu(II), Ni(II), Ru(II); Pth = 2-mercaptopyridine-N-oxide) were used as catalysts. Catalytic testing measurements were done by varying the time, chemical reaction temperature and H2O2 concentration with or without UV energy. Epoxide styrene oxide (SO), benzaldehyde and acetophenone were the major synthesized products. A high batch rate, conversion and selectivity towards SO was shown in the presence of UV. A minor constant formation of CO2 was observed in the stream. Coordinated Ru-based compounds demonstrated the highest process productivity of SO at 60 °C. The effect of the functional alkyl substituent on the ligand Pth, attached to the specific ruthenium(II) centre, decreased the activity of the substance. Ni-Pth selectively yielded benzaldehyde. The stability of the catalysts was examined by applying nuclear magnetic resonance (NMR) spectroscopy and thermogravimetric analysis coupled with mass spectrometry. Tested metal complexes with pyrithione (M-Pth) exhibited excellent reuse recyclability up to 3 cycles.
Collapse
|
19
|
Trobec T, Sepčić K, Žužek MC, Kladnik J, Podjed N, Cardoso Páscoa C, Turel I, Frangež R. Fine Tuning of Cholinesterase and Glutathione-S-Transferase Activities by Organoruthenium(II) Complexes. Biomedicines 2021; 9:biomedicines9091243. [PMID: 34572429 PMCID: PMC8467340 DOI: 10.3390/biomedicines9091243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/26/2021] [Accepted: 09/13/2021] [Indexed: 11/27/2022] Open
Abstract
Cholinesterases (ChEs) show increased activities in patients with Alzheimer’s disease, and remain one of the main therapeutic targets for treatment of this neurodegenerative disorder. A library of organoruthenium(II) complexes was prepared to investigate the influence of their structural elements on inhibition of ChEs, and on another pharmacologically important group of enzymes, glutathione S-transferases (GSTs). Two groups of organoruthenium(II) compounds were considered: (i) organoruthenium(II) complexes with p-cymene as an arene ligand, and (ii) organoruthenium(II) carbonyl complexes as CO-releasing molecules. Eight organoruthenium complexes were screened for inhibitory activities against ChEs and GSTs of human and animal origins. Some compounds inhibited all of these enzymes at low micromolar concentrations, while others selectively inhibited either ChEs or GSTs. This study demonstrates the importance of the different structural elements of organoruthenium complexes for their inhibitory activities against ChEs and GSTs, and also proposes some interesting compounds for further preclinical testing as ChE or GST inhibitory drugs.
Collapse
Affiliation(s)
- Tomaž Trobec
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.T.); (M.C.Ž.)
| | - Kristina Sepčić
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia
- Correspondence: (K.S.); (I.T.); (R.F.); Tel.: +386-1-3203419 (K.S.); +386-1-4798525 (I.T.); +386-1-4779131 (R.F.)
| | - Monika Cecilija Žužek
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.T.); (M.C.Ž.)
| | - Jerneja Kladnik
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.K.); (N.P.); (C.C.P.)
| | - Nina Podjed
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.K.); (N.P.); (C.C.P.)
| | - Catarina Cardoso Páscoa
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.K.); (N.P.); (C.C.P.)
- NOVA School of Science and Technology, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Iztok Turel
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.K.); (N.P.); (C.C.P.)
- Correspondence: (K.S.); (I.T.); (R.F.); Tel.: +386-1-3203419 (K.S.); +386-1-4798525 (I.T.); +386-1-4779131 (R.F.)
| | - Robert Frangež
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.T.); (M.C.Ž.)
- Correspondence: (K.S.); (I.T.); (R.F.); Tel.: +386-1-3203419 (K.S.); +386-1-4798525 (I.T.); +386-1-4779131 (R.F.)
| |
Collapse
|
20
|
Valente A, Podolski-Renić A, Poetsch I, Filipović N, López Ó, Turel I, Heffeter P. Metal- and metalloid-based compounds to target and reverse cancer multidrug resistance. Drug Resist Updat 2021; 58:100778. [PMID: 34403910 DOI: 10.1016/j.drup.2021.100778] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/18/2021] [Accepted: 08/03/2021] [Indexed: 12/19/2022]
Abstract
Drug resistance remains the major cause of cancer treatment failure especially at the late stage of the disease. However, based on their versatile chemistry, metal and metalloid compounds offer the possibility to design fine-tuned drugs to circumvent and even specifically target drug-resistant cancer cells. Based on the paramount importance of platinum drugs in the clinics, two main areas of drug resistance reversal strategies exist: overcoming resistance to platinum drugs as well as multidrug resistance based on ABC efflux pumps. The current review provides an overview of both aspects of drug design and discusses the open questions in the field. The areas of drug resistance covered in this article involve: 1) Altered expression of proteins involved in metal uptake, efflux or intracellular distribution, 2) Enhanced drug efflux via ABC transporters, 3) Altered metabolism in drug-resistant cancer cells, 4) Altered thiol or redox homeostasis, 5) Altered DNA damage recognition and enhanced DNA damage repair, 6) Impaired induction of apoptosis and 7) Altered interaction with the immune system. This review represents the first collection of metal (including platinum, ruthenium, iridium, gold, and copper) and metalloid drugs (e.g. arsenic and selenium) which demonstrated drug resistance reversal activity. A special focus is on compounds characterized by collateral sensitivity of ABC transporter-overexpressing cancer cells. Through this approach, we wish to draw the attention to open research questions in the field. Future investigations are warranted to obtain more insights into the mechanisms of action of the most potent compounds which target specific modalities of drug resistance.
Collapse
Affiliation(s)
- Andreia Valente
- Centro de Química Estrutural and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisboa, Portugal
| | - Ana Podolski-Renić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Serbia
| | - Isabella Poetsch
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Nenad Filipović
- Department of Chemistry and Biochemistry, Faculty of Agriculture, University of Belgrade, Belgrade, Serbia
| | - Óscar López
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Sevilla, Spain
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
21
|
Pivarcsik T, Tóth G, Szemerédi N, Bogdanov A, Spengler G, Kljun J, Kladnik J, Turel I, Enyedy ÉA. Comparison of Solution Chemical Properties and Biological Activity of Ruthenium Complexes of Selected β-Diketone, 8-Hydroxyquinoline and Pyrithione Ligands. Pharmaceuticals (Basel) 2021; 14:518. [PMID: 34072270 PMCID: PMC8226722 DOI: 10.3390/ph14060518] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 12/18/2022] Open
Abstract
In this work, the various biological activities of eight organoruthenium(II) complexes were evaluated to reveal correlations with their stability and reactivity in aqueous media. Complexes with general formula [Ru(η6-p-cymene)(X,Y)(Z)] were prepared, where (X,Y) represents either an O,O-ligand (β-diketone), N,O-ligand (8-hydroxyquinoline) or O,S-pyrithione-type ligands (pyrithione = 1-hydroxypyridine-2(1H)-thione) with Cl- or 1,3,5-triaza-7-phosphaadamantane (PTA) as a co-ligand (Z). The tested complexes inhibit the chlamydial growth on HeLa cells, and one of the complexes inhibits the growth of the human herpes simplex virus-2. The chlorido complexes with N,O- and O,S-ligands displayed strong antibacterial activity on Gram-positive strains including the resistant S. aureus (MRSA) and were cytotoxic in adenocarcinoma cell lines. Effect of the structural variation on the biological properties and solution stability was clearly revealed. The decreased bioactivity of the β-diketone complexes can be related to their lower stability in solution. In contrast, the O,S-pyrithione-type complexes are highly stable in solution and the complexation prevents the oxidation of the O,S-ligands. Comparing the binding of PTA and the chlorido co-ligands, it can be concluded that PTA is generally more strongly coordinated to ruthenium, which at the same time decreased the reactivity of complexes with human serum albumin or 1-methylimidazole as well as diminished their bioactivity.
Collapse
Affiliation(s)
- Tamás Pivarcsik
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary; (T.P.); (G.T.)
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary
| | - Gábor Tóth
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary; (T.P.); (G.T.)
| | - Nikoletta Szemerédi
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (N.S.); (A.B.)
| | - Anita Bogdanov
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (N.S.); (A.B.)
| | - Gabriella Spengler
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary; (T.P.); (G.T.)
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center, Faculty of Medicine, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (N.S.); (A.B.)
| | - Jakob Kljun
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.K.); (J.K.)
| | - Jerneja Kladnik
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.K.); (J.K.)
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.K.); (J.K.)
| | - Éva A. Enyedy
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary; (T.P.); (G.T.)
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm Tér 7, H-6720 Szeged, Hungary
| |
Collapse
|