1
|
Ceccherini V, Giorgi E, Mannelli M, Cirri D, Gamberi T, Gabbiani C, Pratesi A. Synthesis, Chemical Characterization, and Biological Evaluation of Hydrophilic Gold(I) and Silver(I) N-Heterocyclic Carbenes as Potential Anticancer Agents. Inorg Chem 2024; 63:16949-16963. [PMID: 39226133 DOI: 10.1021/acs.inorgchem.4c02581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
A series of new gold(I) and silver(I) N-heterocyclic carbenes bearing a 1-thio-β-d-glucose tetraacetate moiety was synthesized and chemically characterized. The compounds' stability and solubility in physiological conditions were investigated employing a multitechnique approach. Interaction studies with biologically relevant proteins, such as superoxide dismutase (SOD) and human serum albumin (HSA), were conducted via UV-vis absorption spectroscopy and high-resolution ESI mass spectrometry. The biological activity of the compounds was evaluated in the A2780 and A2780R (cisplatin-resistant) ovarian cancer cell lines and the HSkMC (human skeletal muscle) healthy cell line. Inhibition studies of the selenoenzyme thioredoxin reductase (TrxR) were also carried out. The results highlighted that the gold complexes are more stable in aqueous environment and capable of interaction with SOD and HSA. Moreover, these carbenes strongly inhibited the TrxR activity. In contrast, the silver ones underwent structural alterations in the aqueous medium and showed greater antiproliferative activity.
Collapse
Affiliation(s)
- Valentina Ceccherini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Ester Giorgi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Michele Mannelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Damiano Cirri
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Chiara Gabbiani
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| |
Collapse
|
2
|
Coscione F, Zineddu S, Vitali V, Fondi M, Messori L, Perrin E. The Many Lives of Auranofin: How an Old Anti-Rheumatic Agent May Become a Promising Antimicrobial Drug. Antibiotics (Basel) 2024; 13:652. [PMID: 39061334 PMCID: PMC11274207 DOI: 10.3390/antibiotics13070652] [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: 06/21/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Auranofin (AF) is a gold-based compound with a well-known pharmacological and toxicological profile, currently used in the treatment of some severe forms of rheumatoid arthritis. Over the last twenty years, AF has also been repurposed as antiviral, antitumor, and antibacterial drug. In this review we focused on the antibacterial properties of AF, specifically researching the minimal inhibitory concentrations (MIC) of AF in both mono- and diderm bacteria reported so far in literature. AF proves to be highly effective against monoderm bacteria, while diderm are far less susceptible, probably due to the outer membrane barrier. We also reported the current mechanistic hypotheses concerning the antimicrobial properties of AF, although a conclusive description of its antibacterial mode of action is not yet available. Even if its mechanism of action has not been fully elucidated yet and further studies are required to optimize its delivery strategy, AF deserves additional investigation because of its unique mode of action and high efficacy against a wide range of pathogens, which could lead to potential applications in fighting antimicrobial resistance and improving therapeutic outcomes in infectious diseases.
Collapse
Affiliation(s)
- Francesca Coscione
- Department of Biology, University of Florence, Via Madonna del Piano 6, I-50019 Sesto Fiorentino, Italy; (F.C.); (M.F.)
| | - Stefano Zineddu
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3-13, I-50019 Sesto Fiorentino, Italy; (S.Z.); (V.V.)
| | - Valentina Vitali
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3-13, I-50019 Sesto Fiorentino, Italy; (S.Z.); (V.V.)
| | - Marco Fondi
- Department of Biology, University of Florence, Via Madonna del Piano 6, I-50019 Sesto Fiorentino, Italy; (F.C.); (M.F.)
| | - Luigi Messori
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3-13, I-50019 Sesto Fiorentino, Italy; (S.Z.); (V.V.)
| | - Elena Perrin
- Department of Biology, University of Florence, Via Madonna del Piano 6, I-50019 Sesto Fiorentino, Italy; (F.C.); (M.F.)
| |
Collapse
|
3
|
Vitali V, Massai L, Messori L. Strategies for the design of analogs of auranofin endowed with anticancer potential. Expert Opin Drug Discov 2024; 19:855-867. [PMID: 38803122 DOI: 10.1080/17460441.2024.2355329] [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: 03/06/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024]
Abstract
INTRODUCTION Auranofin (AF) is a well-established, FDA-approved, antiarthritic gold drug that is currently being reevaluated for a variety of therapeutic indications through drug repurposing. AF has shown great promise as a potential anticancer agent and has been approved for a few clinical trials in cancer. The renewed interest in AF has led to extensive research into the design, preparation and biological evaluation of auranofin analogs, which may have an even better pharmacological profile than the parent drug. AREAS COVERED This article reviews the strategies for chemical modification of the AF scaffold. Several auranofin analogs have been prepared and characterized for medical application in the field of cancer treatment over the last 20 years. Some emerging structure-function relationships are proposed and discussed. EXPERT OPINION The chemical modification of the AF scaffold has been the subject of intense activity in recent years and this strategy has led to the preparation and evaluation of several AF analogs. The case of iodauranofin is a particularly promising example. The availability of homogeneous biological data for a group of AF derivatives allows some initial structure-function relationships to be proposed, which may inspire the design and synthesis of new and better AF analogs for cancer treatment.
Collapse
Affiliation(s)
- Valentina Vitali
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Lara Massai
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Luigi Messori
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| |
Collapse
|
4
|
Giorgi E, Mannelli M, Gamberi T, Durante M, Gabbiani C, Cirri D, Pratesi A. Cytotoxic auranofin analogues bearing phosphine, arsine and stibine ligands: A study on the possible role of the ligand on the biological activity. J Inorg Biochem 2024; 251:112452. [PMID: 38070433 DOI: 10.1016/j.jinorgbio.2023.112452] [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: 09/25/2023] [Revised: 11/24/2023] [Accepted: 12/01/2023] [Indexed: 12/25/2023]
Abstract
Three gold(I) linear compounds, sharing the general formula [AuI(LPh3)], have been synthesized and characterized. The nature of the ligand has been modified by moving down among some of the elements of group 15, i.e. phosphorus, arsenic and antimony. The structures of derived compounds have been solved through XRD and the reactivity behaviour towards selected biomolecules has been investigated through a multi-technique approach involving NMR, high-resolution mass spectrometry and IR. Moreover, the biological activity of the investigated compounds has been comparatively analyzed through classical methodologies and the disclosed differences are discussed in detail.
Collapse
Affiliation(s)
- Ester Giorgi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Michele Mannelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Maria Durante
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Chiara Gabbiani
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Damiano Cirri
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy.
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy.
| |
Collapse
|
5
|
Geri A, Massai L, Novinec M, Turel I, Messori L. Reactions of Medicinal Gold Compounds with Cathepsin B Explored through Electrospray Mass Spectrometry Measurements. Chempluschem 2024; 89:e202300321. [PMID: 37930642 DOI: 10.1002/cplu.202300321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Abstract
Medicinal gold compounds, a novel class of potential anticancer drugs, are believed to produce their pharmacological effects mainly through direct gold binding to protein targets at the level of solvent exposed cysteine (or selenocysteine) residues. We have explored therein the reactions of a panel of seven representative gold compounds with the cysteine protease cathepsin B according to an established ESI MS approach. Detailed information on the mode of protein binding of these gold compounds is gained; notably, quite distinct patterns of cathepsin B metalation have emerged from these studies. It is shown that panel gold compounds interact preferentially, often exclusively, with the free cysteine located in the active site of the enzyme.
Collapse
Affiliation(s)
- Andrea Geri
- Department of Chemistry "Ugo Schiff", Università di Firenze, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Lara Massai
- Department of Chemistry "Ugo Schiff", Università di Firenze, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Marko Novinec
- Faculty of Chemistry and Chemical Technology, Department of Chemistry and Biochemistry, University of Ljubljana, Večna pot 113, 1000, Ljubljana, Slovenia
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, Department of Chemistry and Biochemistry, University of Ljubljana, Večna pot 113, 1000, Ljubljana, Slovenia
| | - Luigi Messori
- Department of Chemistry "Ugo Schiff", Università di Firenze, Via della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| |
Collapse
|
6
|
Casagrande N, Borghese C, Corona G, Aldinucci D, Altaf M, Sulaiman AAA, Isab AA, Ahmad S, Peedikakkal AMP. Dinuclear gold(I) complexes based on carbene and diphosphane ligands: bis[2-(dicyclohexylphosphano)ethyl]amine complex inhibits the proteasome activity, decreases stem cell markers and spheroid viability in lung cancer cells. J Biol Inorg Chem 2023; 28:751-766. [PMID: 37955736 DOI: 10.1007/s00775-023-02025-x] [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: 05/02/2023] [Accepted: 10/02/2023] [Indexed: 11/14/2023]
Abstract
Three new dinuclear gold(I) complexes (1-3) containing a carbene (1,3-Bis(2,6-di-isopropylphenyl)imidazol-2-ylidene (IPr)) and diphosphane ligands [bis(1,2-diphenylphosphano)ethane (Dppe), bis(1,3-diphenylphosphano)propane (Dppp) and bis[2-(dicyclohexylphosphano)ethyl]amine (DCyPA)], were synthesized and characterized by elemental analysis and, ESI-MS, mid FT-IR and NMR spectroscopic methods. The structures of complexes 2 and 3 were determined by X-ray crystallography, which revealed that the complexes are dinuclear having gold(I) ions linearly coordinated. The anticancer activities of the complexes (1-3) were evaluated in lung (A549), breast (MC-F7), prostate (PC-3), osteosarcoma (MG-63) and ovarian (A2780 and A2780cis) cancer models. Growth inhibition by the new complexes was higher than cisplatin in all cell lines tested. The mechanism of action of complex 3 was investigated in A549 cells using 2-dimensional (2D) models and 3D-multicellular tumor spheroids. Treatment of A549 cells with complex 3 caused: the induction of apoptosis and the generation of reactive oxygen species; the cell cycle arrest in the G0/G1 phase; the inhibition of both the proteasome and the NF-kB activity; the down-regulation of lung cancer stem cell markers (NOTCH1, CD133, ALDH1 and CD44). Complex 3 was more active than cisplatin also in 3D models of A549 lung cancer cells.
Collapse
Affiliation(s)
- Naike Casagrande
- Molecular Oncology, Centro Di Riferimento Oncologico Di Aviano (CRO) IRCCS, 33081, Aviano, Italy
| | - Cinzia Borghese
- Molecular Oncology, Centro Di Riferimento Oncologico Di Aviano (CRO) IRCCS, 33081, Aviano, Italy
| | - Giuseppe Corona
- Immunopathology and Cancer Biomarkers Unit, Centro Di Riferimento Oncologico Di Aviano (CRO), IRCCS, 33081, Aviano, Italy
| | - Donatella Aldinucci
- Molecular Oncology, Centro Di Riferimento Oncologico Di Aviano (CRO) IRCCS, 33081, Aviano, Italy.
| | - Muhammad Altaf
- Department of Chemistry, Government College University Lahore, Lahore, 54000, Pakistan
| | - Adam A A Sulaiman
- Core Research Facilities (CRF), King Fahd University of Petroleum and Minerals, 31261, Dhahran, Saudi Arabia.
- Department of Chemistry, King Fahd University of Petroleum and Minerals, 31261, Dhahran, Saudi Arabia.
| | - Anvarhusein A Isab
- Department of Chemistry, King Fahd University of Petroleum and Minerals, 31261, Dhahran, Saudi Arabia.
- Interdisciplinary Research Center for Advanced Materials, King Fahd University of Petroleum and Minerals, 31261, Dhahran, Saudi Arabia.
| | - Saeed Ahmad
- Department of Chemistry, College of Sciences and Humanities, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| | - Abdul Malik P Peedikakkal
- Department of Chemistry, King Fahd University of Petroleum and Minerals, 31261, Dhahran, Saudi Arabia
- Interdisciplinary Research Center for Hydrogen and Energy Storage, King Fahd University of Petroleum and Minerals, 31261, Dhahran, Saudi Arabia
| |
Collapse
|
7
|
Chiaverini L, Baglini E, Mannelli M, Poggetti V, Da Settimo F, Taliani S, Gamberi T, Barresi E, La Mendola D, Marzo T. A complex bearing TSPO PIGA ligand coordinated to the [Au(PEt 3)] + pharmacophore is highly cytotoxic against ovarian cancer cells. Biometals 2023; 36:961-968. [PMID: 36869967 PMCID: PMC10545567 DOI: 10.1007/s10534-023-00496-8] [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: 12/19/2022] [Accepted: 02/09/2023] [Indexed: 03/05/2023]
Abstract
Auranofin ([1-(thio-κS)-β-D-glucopyranose-2,3,4,6-tetraacetato](triethylphosphine)-gold) is a leading gold-based drug clinically used to treat arthritis. In the last years, it entered various drug reprofiling programs, and it has been found promising against various forms of tumor, including ovarian cancer. Evidence showed as its antiproliferative profile mainly depends on the inhibition of thioredoxin reductase (TrxR), being this mitochondrial system its main target. In this context, we report here the synthesis and biological evaluation of a novel complex designed as auranofin analogue obtained through the conjugation of a phenylindolylglyoxylamide ligand (which belongs to the so-called PIGA TSPO ligand family) with the auranofin-derived cationic fragment [Au(PEt3)]+. This complex is characterized by two parts. The phenylindolylglyoxylamide moiety, owing to its high affinity for TSPO (in the low nM range) should drive the compound to target mitochondria, whereas the [Au(PEt3)]+ cation is the actual anticancer-active molecular fragment. Overall, we wanted to offer the proof-of-concept that by coupling PIGA ligands to anticancer gold active moieties, it is possible to preserve and even improve anticancer effects, opening the avenue to a reliable approach for targeted therapy.
Collapse
Affiliation(s)
- Lorenzo Chiaverini
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Emma Baglini
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Michele Mannelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Valeria Poggetti
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Federico Da Settimo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy.
| | - Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy.
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
- University Consortium for Research in the Chemistry of Metal Ions in Biological Systems (CIRCMSB), Via Celso Ulpiani 27, 70126, Bari, Italy
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
- University Consortium for Research in the Chemistry of Metal Ions in Biological Systems (CIRCMSB), Via Celso Ulpiani 27, 70126, Bari, Italy
| |
Collapse
|
8
|
Zarewa SA, Binobaid L, Sulaiman AAA, Sobeai HMA, Alotaibi M, Alhoshani A, Isab AA. Synthesis, Characterization, and Anticancer Activity of Phosphanegold(i) Complexes of 3-Thiosemicarbano-butan-2-one Oxime. Biomedicines 2023; 11:2512. [PMID: 37760953 PMCID: PMC10525815 DOI: 10.3390/biomedicines11092512] [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: 07/22/2023] [Revised: 08/23/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Four novel phosphanegold(I) complexes of the type [Au(PR3)(DMT)].PF6 (1-4) were synthesized from 3-Thiosemicarbano-butan-2-one oxime ligand (TBO) and precursors [Au(PR3)Cl], (where R = methyl (1), ethyl (2), tert-butyl (3), and phenyl (4)). The resulting complexes were characterized by elemental analyses and melting point as well as various spectroscopic techniques, including FTIR and (1H, 13C, and 31P) NMR spectroscopy. The spectroscopic data confirmed the coordination of TBO ligands to phosphanegold(I) moiety. The solution chemistry of complexes 1-4 indicated their stability in both dimethyl sulfoxide (DMSO) and a mixture of EtOH:H2O (1:1). In vitro cytotoxicity of the complexes was evaluated relative to cisplatin using an MTT assay against three different cancer cell lines: HCT116 (human colon cancer), MDA-MB-231 (human breast cancer), and B16 (murine skin cancer). Complexes 2, 3, and 4 exhibited significant cytotoxic effects against all tested cancer cell lines and showed significantly higher activity than cisplatin. To elucidate the mechanism underlying the cytotoxic effects of the phosphanegold(I) TBO complexes, various assays were employed, including mitochondrial membrane potential, ROS production, and gene expression analyses. The data obtained suggest that complex 2 exerts potent anticancer activity against breast cancer cells (MDA-MB-231) through the induction of oxidative stress, mitochondrial dysfunction, and apoptosis. Gene expression analyses showed an increase in the activity of the proapoptotic gene caspase-3 and a reduction in the activity of the antiapoptotic gene BCL-xL, which supported the findings that apoptosis had occurred.
Collapse
Affiliation(s)
- Sani A. Zarewa
- Department of Chemistry, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia;
| | - Lama Binobaid
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.B.); (H.M.A.S.); (M.A.); (A.A.)
| | - Adam A. A. Sulaiman
- Department of Chemistry, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia;
- Core Research Facilities (CRF), King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
| | - Homood M. As Sobeai
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.B.); (H.M.A.S.); (M.A.); (A.A.)
| | - Moureq Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.B.); (H.M.A.S.); (M.A.); (A.A.)
| | - Ali Alhoshani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (L.B.); (H.M.A.S.); (M.A.); (A.A.)
| | - Anvarhusein A. Isab
- Department of Chemistry, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia;
- Interdisciplinary Research Center for Advanced Materials, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
| |
Collapse
|
9
|
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
|
10
|
Barresi E, Robello M, Baglini E, Poggetti V, Viviano M, Salerno S, Da Settimo F, Taliani S. Indol-3-ylglyoxylamide as Privileged Scaffold in Medicinal Chemistry. Pharmaceuticals (Basel) 2023; 16:997. [PMID: 37513909 PMCID: PMC10386336 DOI: 10.3390/ph16070997] [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: 06/12/2023] [Revised: 07/05/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
In recent years, indolylglyoxylamide-based derivatives have received much attention due to their application in drug design and discovery, leading to the development of a wide array of compounds that have shown a variety of pharmacological activities. Combining the indole nucleus, already validated as a "privileged structure," with the glyoxylamide function allowed for an excellent template to be obtained that is suitable to a great number of structural modifications aimed at permitting interaction with specific molecular targets and producing desirable therapeutic effects. The present review provides insight into how medicinal chemists have elegantly exploited the indolylglyoxylamide moiety to obtain potentially useful drugs, with a particular focus on compounds exhibiting activity in in vivo models or reaching clinical trials. All in all, this information provides exciting new perspectives on existing data that can be useful in further design of indolylglyoxylamide-based molecules with interesting pharmacological profiles. The aim of this report is to present an update of collection data dealing with the employment of this moiety in the rational design of compounds that are able to interact with a specific target, referring to the last 20 years.
Collapse
Affiliation(s)
- Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Marco Robello
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Emma Baglini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Valeria Poggetti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Monica Viviano
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Silvia Salerno
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Federico Da Settimo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| |
Collapse
|
11
|
Ronga L, Tolbatov I, Giorgi E, Pisarek P, Enjalbal C, Marrone A, Tesauro D, Lobinski R, Marzo T, Cirri D, Pratesi A. Mechanistic Evaluations of the Effects of Auranofin Triethylphosphine Replacement with a Trimethylphosphite Moiety. Inorg Chem 2023; 62:10389-10396. [PMID: 37342994 PMCID: PMC10324304 DOI: 10.1021/acs.inorgchem.3c01280] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Indexed: 06/23/2023]
Abstract
Auranofin, a gold(I)-based complex, is under clinical trials for application as an anticancer agent for the treatment of nonsmall-cell lung cancer and ovarian cancer. In the past years, different derivatives have been developed, modifying gold linear ligands in the search for new gold complexes endowed with a better pharmacological profile. Recently, a panel of four gold(I) complexes, inspired by the clinically established compound auranofin, was reported by our research group. As described, all compounds possess an [Au{P(OMe)3}]+ cationic moiety, in which the triethylphosphine of the parent compound auranofin was replaced with an oxygen-rich trimethylphosphite ligand. The gold(I) linear coordination geometry was complemented by Cl-, Br-, I-, and the auranofin-like thioglucose tetraacetate ligand. As previously reported, despite their close similarity to auranofin, the panel compounds exhibited some peculiar and distinctive features, such as lower log P values which can induce relevant differences in the overall pharmacokinetic profiles. To get better insight into the P-Au strength and stability, an extensive study was carried out for relevant biological models, including three different vasopressin peptide analogues and cysteine, using 31P NMR and LC-ESI-MS. A DFT computational study was also carried out for a better understanding of the theoretical fundamentals of the disclosed differences with regard to triethylphosphine parent compounds.
Collapse
Affiliation(s)
- Luisa Ronga
- Université
de Pau et des Pays de l’Adour, E2S UPPA, CNRS, IPREM, 64053 Pau, France
| | - Iogann Tolbatov
- Institute
of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology, Av. Paisos Catalans 16, 43007 Tarragona, Spain
| | - Ester Giorgi
- Department
of Chemistry and Industrial Chemistry, University
of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Paulina Pisarek
- Université
de Pau et des Pays de l’Adour, E2S UPPA, CNRS, IPREM, 64053 Pau, France
| | - Christine Enjalbal
- IBMM,
Université de Montpellier, CNRS, ENSCM, UMR 5247, 34293 Montpellier, France
| | - Alessandro Marrone
- Department
of Pharmacy, University “G. D’Annunzio”
Chieti-Pescara, Via dei
Vestini, 31, 66100 Chieti, Italy
| | - Diego Tesauro
- Department
of Pharmacy and CIRPeB, Università
degli Studi di Napoli Federico II, 80131 Naples, Italy
| | - Ryszard Lobinski
- Université
de Pau et des Pays de l’Adour, E2S UPPA, CNRS, IPREM, 64053 Pau, France
- Chair
of Analytical Chemistry, Department of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Tiziano Marzo
- Department
of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, 56126 Pisa, Italy
| | - Damiano Cirri
- Department
of Chemistry and Industrial Chemistry, University
of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Alessandro Pratesi
- Department
of Chemistry and Industrial Chemistry, University
of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| |
Collapse
|
12
|
König P, Zhulenko R, Suparman E, Hoffmeister H, Bückreiß N, Ott I, Bendas G. A biscarbene gold(I)-NHC-complex overcomes cisplatin-resistance in A2780 and W1 ovarian cancer cells highlighting pERK as regulator of apoptosis. Cancer Chemother Pharmacol 2023; 92:57-69. [PMID: 37272932 PMCID: PMC10261188 DOI: 10.1007/s00280-023-04548-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/25/2023] [Indexed: 06/06/2023]
Abstract
PURPOSE Cisplatin resistance is the major obstacle in the clinical treatment of ovarian cancer patients. Molecular mechanisms of cisplatin resistance are multifaceted. Gold(I)-compounds, i.e. N-heterocyclic carbene-gold(I)-complexes (NHC-Au(I)) has been regarded as promising cytotoxic drug candidates. However, their potential to overcome cisplatin resistance has hardly been addressed yet. Here we investigated the activity of the gold(I) drug auranofin and the NHC-Au(I)-compound MC3 in W1CR and A2780cis cisplatin-resistant ovarian cancer cells. METHODS Cytotoxicity of auranofin and MC3 was detected by MTT assay, correlated with intracellular gold(I) content, analyzed by AAS, and with flow cytometric detection of the cell cycle. Insight into cellular redox balance was provided by fluorimetric ROS-formation assay and western blotting thioredoxin (Trx) and Nrf2. The role of ERK was elucidated by using the inhibitor SCH772984 and its impact on cytotoxicity upon co-treatment with cisplatin and Au(I)-compounds, respectively. RESULTS MC3 overcomes cisplatin resistance in A2780cis and W1CR, and auranofin in W1CR cells completely, which is neither reflected by intracellular gold levels nor cell cycle changes. Upregulated redox balance appears as a basis for resistance. W1CR cells possess higher Trx levels, whereas A2780cis cells display strong Nrf2 expression as anti-oxidative protection. Nevertheless, overcoming redox balance appears not primary mode of activity comparing cisplatin and gold(I)-compounds. pERK emerges as a critical component and thus a promising target for overcoming resistance, regulating apoptosis differently in response to either gold(I) or cisplatin in A2780 cells. CONCLUSION These data reflect the complexity of cisplatin resistance in cell models and emphasize NHC-Au(I)-complexes as prospective cytotoxic agents for further investigations in that respect.
Collapse
Affiliation(s)
- Philipp König
- Department of Pharmacy, University Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Roman Zhulenko
- Department of Pharmacy, University Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Eloy Suparman
- Department of Pharmacy, University Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Henrik Hoffmeister
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, 38106, Brunswick, Germany
| | - Nico Bückreiß
- Department of Pharmacy, University Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Ingo Ott
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, 38106, Brunswick, Germany
| | - Gerd Bendas
- Department of Pharmacy, University Bonn, An der Immenburg 4, 53121, Bonn, Germany.
| |
Collapse
|
13
|
Schleser SW, Ghosh H, Hörner G, Seib J, Bhattacharyya S, Weber B, Schobert R, Dandawate P, Biersack B. New 4,5-Diarylimidazol-2-ylidene-iodidogold(I) Complexes with High Activity against Esophageal Adenocarcinoma Cells. Int J Mol Sci 2023; 24:5738. [PMID: 36982817 PMCID: PMC10052191 DOI: 10.3390/ijms24065738] [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: 02/10/2023] [Revised: 03/11/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
Inspired by the vascular-disrupting agent combretastatin A-4 and recently published anticancer active N-heterocyclic carbene (NHC) complexes of Au(I), a series of new iodidogold(I)-NHC complexes was synthesized and characterized. The iodidogold(I) complexes were synthesized by a route involving van Leusen imidazole formation and N-alkylation, followed by complexation with Ag2O, transmetalation with chloro(dimethylsulfide)gold(I) [Au(DMS)Cl], and anion exchange with KI. The target complexes were characterized by IR spectroscopy, 1H and 13C NMR spectroscopy, and mass spectrometry. The structure of 6c was validated via single-crystal X-ray diffraction. A preliminary anticancer screening of the complexes using two esophageal adenocarcinoma cell lines showed promising nanomolar activities for certain iodidogold(I) complexes accompanied with apoptosis induction, as well as c-Myc and cyclin D1 suppression in esophageal adenocarcinoma cells treated with the most promising derivative 6b.
Collapse
Affiliation(s)
- Sebastian W. Schleser
- Organic Chemistry 1, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| | - Hindole Ghosh
- Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Gerald Hörner
- Inorganic Chemistry IV, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| | - Jonathan Seib
- Organic Chemistry 1, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| | - Sangita Bhattacharyya
- Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Birgit Weber
- Inorganic Chemistry IV, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| | - Rainer Schobert
- Organic Chemistry 1, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| | - Prasad Dandawate
- Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Bernhard Biersack
- Organic Chemistry 1, University of Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany
| |
Collapse
|
14
|
Aragoni MC, Podda E, Caria V, Carta SA, Cherchi MF, Lippolis V, Murgia S, Orrù G, Pippia G, Scano A, Slawin AMZ, Woollins JD, Pintus A, Arca M. [Au III(N^N)Br 2](PF 6): A Class of Antibacterial and Antibiofilm Complexes (N^N = 2,2'-Bipyridine and 1,10-Phenanthroline Derivatives). Inorg Chem 2023; 62:2924-2933. [PMID: 36728360 PMCID: PMC9930124 DOI: 10.1021/acs.inorgchem.2c04410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A series of new complexes of general formula [AuIII(N^N)Br2](PF6) (N^N = 2,2'-bipyridine and 1,10-phenanthroline derivatives) were prepared and characterized by spectroscopic, electrochemical, and diffractometric techniques and tested against Gram-positive and Gram-negative bacterial strains (Staphylococcus aureus, Streptococcus intermedius, Pseudomonas aeruginosa, and Escherichia coli), showing promising antibacterial and antibiofilm properties.
Collapse
Affiliation(s)
- M. Carla Aragoni
- Dipartimento
di Scienze Chimiche e Geologiche, Università
degli Studi di Cagliari, S. S. 554 bivio per Sestu, Monserrato Cagliari09042, Italy
| | - Enrico Podda
- Dipartimento
di Scienze Chimiche e Geologiche, Università
degli Studi di Cagliari, S. S. 554 bivio per Sestu, Monserrato Cagliari09042, Italy,Centro
Servizi di Ateneo per la Ricerca (CeSAR), Università degli Studi di Cagliari, S. S. 554 bivio per Sestu, Monserrato Cagliari09042, Italy
| | - Veronica Caria
- Dipartimento
di Scienze Chimiche e Geologiche, Università
degli Studi di Cagliari, S. S. 554 bivio per Sestu, Monserrato Cagliari09042, Italy
| | - Silvia A. Carta
- Dipartimento
di Scienze Chimiche e Geologiche, Università
degli Studi di Cagliari, S. S. 554 bivio per Sestu, Monserrato Cagliari09042, Italy
| | - M. Francesca Cherchi
- Dipartimento
di Scienze Chimiche e Geologiche, Università
degli Studi di Cagliari, S. S. 554 bivio per Sestu, Monserrato Cagliari09042, Italy
| | - Vito Lippolis
- Dipartimento
di Scienze Chimiche e Geologiche, Università
degli Studi di Cagliari, S. S. 554 bivio per Sestu, Monserrato Cagliari09042, Italy
| | - Simone Murgia
- Dipartimento
di Scienze Chimiche e Geologiche, Università
degli Studi di Cagliari, S. S. 554 bivio per Sestu, Monserrato Cagliari09042, Italy
| | - Germano Orrù
- Dipartimento
di Scienze Chirurgiche, University of Cagliari, Cagliari09042, Italy
| | - Gabriele Pippia
- Dipartimento
di Scienze Chimiche e Geologiche, Università
degli Studi di Cagliari, S. S. 554 bivio per Sestu, Monserrato Cagliari09042, Italy
| | - Alessandra Scano
- Dipartimento
di Scienze Chirurgiche, University of Cagliari, Cagliari09042, Italy
| | - Alexandra M. Z. Slawin
- EaStCHEM
School of Chemistry, University of St. Andrews, North Haugh, Fife, St. AndrewsKY16 9ST, U.K.
| | - J. Derek Woollins
- EaStCHEM
School of Chemistry, University of St. Andrews, North Haugh, Fife, St. AndrewsKY16 9ST, U.K.,Department
of Chemistry, Khalifa University, Abu Dhabi127788, United Arab Emirates
| | - Anna Pintus
- Dipartimento
di Scienze Chimiche e Geologiche, Università
degli Studi di Cagliari, S. S. 554 bivio per Sestu, Monserrato Cagliari09042, Italy,
| | - Massimiliano Arca
- Dipartimento
di Scienze Chimiche e Geologiche, Università
degli Studi di Cagliari, S. S. 554 bivio per Sestu, Monserrato Cagliari09042, Italy,
| |
Collapse
|
15
|
Chemical Modification of Auranofin Yields a New Family of Anticancer Drug Candidates: The Gold(I) Phosphite Analogues. Molecules 2023; 28:molecules28031050. [PMID: 36770719 PMCID: PMC9920260 DOI: 10.3390/molecules28031050] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
A panel of four novel gold(I) complexes, inspired by the clinically established gold drug auranofin (1-Thio-β-D-glucopyranosatotriethylphosphine gold-2,3,4,6-tetraacetate), was prepared and characterized. All these compounds feature the replacement of the triethylphosphine ligand of the parent compound auranofin with a trimethylphosphite ligand. The linear coordination around the gold(I) center is completed by Cl-, Br-, I- or by the thioglucose tetraacetate ligand (SAtg). The in-solution behavior of these gold compounds as well as their interactions with some representative model proteins were comparatively analyzed through 31PNMR and ESI-MS measurements. Notably, all panel compounds turned out to be stable in aqueous media, but significant differences with respect to auranofin were disclosed in their interactions with a few leading proteins. In addition, the cytotoxic effects produced by the panel compounds toward A2780, A2780R and SKOV-3 ovarian cancer cells were quantitated and found to be in the low micromolar range, since the IC50 of all compounds was found to be between 1 μM and 10 μM. Notably, these novel gold complexes showed large and similar inhibition capabilities towards the key enzyme thioredoxin reductase, again comparable to those of auranofin. The implications of these results for the discovery of new and effective gold-based anticancer agents are discussed.
Collapse
|
16
|
Sulaiman AAA, Casagrande N, Borghese C, Corona G, Isab AA, Ahmad S, Aldinucci D, Altaf M. Design, Synthesis, and Preclinical Activity in Ovarian Cancer Models of New Phosphanegold(I)-N-heterocyclic Carbene Complexes. J Med Chem 2022; 65:14424-14440. [DOI: 10.1021/acs.jmedchem.2c00737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Adam A. A. Sulaiman
- Department of Chemistry, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
| | - Naike Casagrande
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Cinzia Borghese
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Giuseppe Corona
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Anvarhusein A. Isab
- Department of Chemistry, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
- Interdisciplinary Research Center for Advanced Materials, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
| | - Saeed Ahmad
- Department of Chemistry, College of Sciences and Humanities, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Donatella Aldinucci
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Muhammad Altaf
- Department of Chemistry, Government College University Lahore, Lahore 54000, Pakistan
| |
Collapse
|
17
|
Cirri D, Massai L, Giacomelli C, Trincavelli ML, Guerri A, Gabbiani C, Messori L, Pratesi A. Synthesis, chemical characterization, and biological evaluation of a novel auranofin derivative as an anticancer agent. Dalton Trans 2022; 51:13527-13539. [PMID: 36000524 DOI: 10.1039/d2dt00836j] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A novel gold(I) complex inspired by the known medicinal inorganic compounds auranofin and thimerosal, namely ethylthiosalicylate(triethylphosphine)gold(I) (AFETT hereafter), was synthesized and characterised and its structure was resolved through X-ray diffraction. The solution behavior of AFETT and its interactions with two biologically relevant proteins (i.e. human serum albumin and haemoglobin) and with a synthetic dodecapeptide reproducing the C-terminal portion of thioredoxin reductase were comparatively analyzed through 31P NMR and ESI-MS. Remarkable binding properties toward these biomolecules were disclosed. Moreover, the cytotoxic effects produced by AFETT on two ovarian cancer cell lines (A2780 and A2780 R) and one colorectal cancer cell line (HCT116) were analyzed and found to be strong and nearly superimposable to those of auranofin. Interestingly, for both compounds, the ability to induce downregulation of vimentin expression in A2780 R cells was evidenced. Despite its close similarity to auranofin, AFETT is reported to exhibit some peculiar and distinctive features such as a lower lipophilicity, an increased water solubility and a faster reactivity towards the selected target biomolecules. These differences might confer to AFETT significant pharmaceutical and therapeutic advantages over auranofin itself.
Collapse
Affiliation(s)
- Damiano Cirri
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy.
| | - Lara Massai
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, FI, Italy.
| | - Chiara Giacomelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | | | - Annalisa Guerri
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, FI, Italy.
| | - Chiara Gabbiani
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy.
| | - Luigi Messori
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, FI, Italy.
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy.
| |
Collapse
|
18
|
Aires RL, Santos IA, Fontes JV, Bergamini FRG, Jardim ACG, Abbehausen C. Triphenylphosphine gold(I) derivatives promote antiviral effects against the Chikungunya virus. METALLOMICS : INTEGRATED BIOMETAL SCIENCE 2022; 14:6650674. [PMID: 35894863 DOI: 10.1093/mtomcs/mfac056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/11/2022] [Indexed: 11/14/2022]
Abstract
Herein a systematic series of four [AuLL']n+ n = 0, +1 complexes, where L = 1,3-bis(mesityl)imidazole-2-ylidene (IMes), or triphenylphosphine (PPh3), and L' = chloride, or 4-dimethylaminopyridine (DMAP), had their in vitro antiviral activity assessed against Chikungunya virus (CHIKV). The PPh3 derivatives inhibited viral replication by 99%, whereas the IMes derivatives about 50%. The lipophilicity of the PPh3 derivatives is higher than the IMes-bearing compounds, which can be related to their more prominent antiviral activities. The dissociation of DMAP is faster than chloride in solution for both IMes and PPh3 derivatives; however, it does not significantly affect their in vitro activities, showing a higher dependence on the nature of L rather than L' towards their antiviral effects. All complexes bind to N-acetyl-L-cysteine, with the Ph3P-bearing complexes coordinating at a faster rate to this amino acid. The binding constants to bovine serum albumin (BSA) are in the order of 104, slightly higher for the DMAP complexes in both PPh3 and IMes derivatives. Mechanistic investigations of the PPh3 complexes showed a ubiquitous protective effect of the compounds in the pre-treatment, early stages, and post-entry assays. The most significant inhibition was observed in post-entry activity, in which the complexes blocked viral replication in 99%, followed by up to 95% inhibition of the early stages of infection. Pre-treatment assays showed a 92% and 80% replication decrease for the chloride and DMAP derivatives, respectively. dsRNA binding assays showed a significant interaction of the compounds with dsRNA, an essential biomolecule to viral replication.
Collapse
Affiliation(s)
- Rochanna L Aires
- Institute of Chemistry, University of Campinas-UNICAMP, Campinas-SP, 13083-871, Brazil
| | - Igor A Santos
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia-MG 38405-302, Brazil
| | - Josielle V Fontes
- Institute of Chemistry, University of Campinas-UNICAMP, Campinas-SP, 13083-871, Brazil
| | - Fernando R G Bergamini
- Laboratory of Synthesis of Bioinspired Molecules, Institute of Chemistry, Federal University of Uberlândia, MG 38408-100, Brazil.,Max-Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Ana Carolina G Jardim
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia-MG 38405-302, Brazil.,Institute of Biosciences, Humanities and Exact Sciences (Ibilce), São Paulo State University (Unesp), Campus São José do Rio Preto, São José do Rio Preto, SP, Brazil
| | - Camilla Abbehausen
- Institute of Chemistry, University of Campinas-UNICAMP, Campinas-SP, 13083-871, Brazil
| |
Collapse
|
19
|
Chiappetta G, Gamberi T, Faienza F, Limaj X, Rizza S, Messori L, Filomeni G, Modesti A, Vinh J. Redox proteome analysis of auranofin exposed ovarian cancer cells (A2780). Redox Biol 2022; 52:102294. [PMID: 35358852 PMCID: PMC8966199 DOI: 10.1016/j.redox.2022.102294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/16/2022] [Indexed: 01/03/2023] Open
Abstract
The effects of Auranofin (AF) on protein expression and protein oxidation in A2780 cancer cells were investigated through a strategy based on simultaneous expression proteomics and redox proteomics determinations. Bioinformatics analysis of the proteomics data supports the view that the most critical cellular changes elicited by AF treatment consist of thioredoxin reductase inhibition, alteration of the cell redox state, impairment of the mitochondrial functions, metabolic changes associated with conversion to a glycolytic phenotype, induction of ER stress. The occurrence of the above cellular changes was extensively validated by performing direct biochemical assays. Our data are consistent with the concept that AF produces its effects through a multitarget mechanism that mainly affects the redox metabolism and the mitochondrial functions and results into severe ER stress. Results are discussed in the context of the current mechanistic knowledge existing on AF. Redox proteomics allows to underline cell adaptation mechanisms in response to Auranofin treatment in ovarian cancer cells. BRCA1 is one of the major candidates of the ovarian cancer cell adaptation to Auranofin treatment. Auranofin alters the oxidative phosphorylation and mitochondrial protein import machinery. TRAP1 C501 modulates Auranofin toxicity. Auranofin induces severe stress of the endoplasmic reticulum.
Collapse
Affiliation(s)
- Giovanni Chiappetta
- Biological Mass Spectrometry and Proteomics Group, SMBP, PDC CNRS UMR, 8249, ESPCI Paris, Université PSL, 10 rue Vauquelin, 75005, Paris, France.
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134, Florence, Italy.
| | - Fiorella Faienza
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Xhesika Limaj
- Biological Mass Spectrometry and Proteomics Group, SMBP, PDC CNRS UMR, 8249, ESPCI Paris, Université PSL, 10 rue Vauquelin, 75005, Paris, France
| | - Salvatore Rizza
- Redox Signaling and Oxidative Stress Group, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Luigi Messori
- Metmed Lab, Department of Chemistry, University of Florence, via della lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Giuseppe Filomeni
- Department of Biology, University of Rome Tor Vergata, Rome, Italy; Redox Signaling and Oxidative Stress Group, Danish Cancer Society Research Center, Copenhagen, Denmark; Center for Healthy Aging, University of Copenhagen, Denmark
| | - Alessandra Modesti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Joelle Vinh
- Biological Mass Spectrometry and Proteomics Group, SMBP, PDC CNRS UMR, 8249, ESPCI Paris, Université PSL, 10 rue Vauquelin, 75005, Paris, France
| |
Collapse
|
20
|
Ferraro MG, Piccolo M, Misso G, Santamaria R, Irace C. Bioactivity and Development of Small Non-Platinum Metal-Based Chemotherapeutics. Pharmaceutics 2022; 14:pharmaceutics14050954. [PMID: 35631543 PMCID: PMC9147010 DOI: 10.3390/pharmaceutics14050954] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023] Open
Abstract
Countless expectations converge in the multidisciplinary endeavour for the search and development of effective and safe drugs in fighting cancer. Although they still embody a minority of the pharmacological agents currently in clinical use, metal-based complexes have great yet unexplored potential, which probably hides forthcoming anticancer drugs. Following the historical success of cisplatin and congeners, but also taking advantage of conventional chemotherapy limitations that emerged with applications in the clinic, the design and development of non-platinum metal-based chemotherapeutics, either as drugs or prodrugs, represents a rapidly evolving field wherein candidate compounds can be fine-tuned to access interactions with druggable biological targets. Moving in this direction, over the last few decades platinum family metals, e.g., ruthenium and palladium, have been largely proposed. Indeed, transition metals and molecular platforms where they originate are endowed with unique chemical and biological features based on, but not limited to, redox activity and coordination geometries, as well as ligand selection (including their inherent reactivity and bioactivity). Herein, current applications and progress in metal-based chemoth are reviewed. Converging on the recent literature, new attractive chemotherapeutics based on transition metals other than platinum—and their bioactivity and mechanisms of action—are examined and discussed. A special focus is committed to anticancer agents based on ruthenium, palladium, rhodium, and iridium, but also to gold derivatives, for which more experimental data are nowadays available. Next to platinum-based agents, ruthenium-based candidate drugs were the first to reach the stage of clinical evaluation in humans, opening new scenarios for the development of alternative chemotherapeutic options to treat cancer.
Collapse
Affiliation(s)
- Maria Grazia Ferraro
- BioChemLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (R.S.)
| | - Marialuisa Piccolo
- BioChemLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (R.S.)
| | - Gabriella Misso
- Department of Precision Medicine, School of Medicine and Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Correspondence: (G.M.); (C.I.)
| | - Rita Santamaria
- BioChemLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (R.S.)
| | - Carlo Irace
- BioChemLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (R.S.)
- Correspondence: (G.M.); (C.I.)
| |
Collapse
|
21
|
Drug combination study of novel oxorhenium(V) complexes. J Inorg Biochem 2022; 231:111807. [DOI: 10.1016/j.jinorgbio.2022.111807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/11/2022] [Accepted: 03/21/2022] [Indexed: 11/23/2022]
|
22
|
Tolbatov I, Marrone A. Selenocysteine of thioredoxin reductase as the primary target for the antitumor metallodrugs: A computational point of view. J Organomet Chem 2022. [DOI: 10.1016/j.jorganchem.2022.122330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
23
|
Luo M, Ma X, Jiang W, Zhang J, Liu W, Wei S, Liu H. Novel phosphanegold(I) thiolate complexes suppress de novo lipid synthesis in human lung cancer. Eur J Med Chem 2022; 232:114168. [DOI: 10.1016/j.ejmech.2022.114168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/17/2022] [Accepted: 01/30/2022] [Indexed: 12/13/2022]
|
24
|
Sulaiman AAA, Ahmad S, Mujahid Hashimi S, Alqosaibi AI, Peedikakkal AMP, Alhoshani A, Alsaleh NB, Isab AA. Novel dinuclear gold( i) complexes containing bis(diphenylphosphano)alkanes and (biphenyl-2-yl)(di- tert-butyl)phosphane: synthesis, structural characterization and anticancer activity. NEW J CHEM 2022. [DOI: 10.1039/d2nj01680j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Four novel dinuclear phosphanegold(I) complexes containing bis(diphenylphosphano)alkanes and related phosphano alkanes were synthesized and characterized by elemental analysis, FTIR, NMR spectroscopy, and X-ray crystallography.
Collapse
Affiliation(s)
- Adam A. A. Sulaiman
- Core Research Facilities (CRF), King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
- Department of Chemistry, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
| | - Saeed Ahmad
- Department of Chemistry, College of Sciences and Humanities, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Saeed Mujahid Hashimi
- School of Medical Science, and Menzies Health Institute Queensland, Griffith University, Parklands, QLD, Australia
| | - Amany I. Alqosaibi
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441, Dammam, Saudi Arabia
| | | | - Ali Alhoshani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nasser B. Alsaleh
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Anvarhusein A. Isab
- Department of Chemistry, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
- Interdisciplinary Research Center for Advanced Materials, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
| |
Collapse
|
25
|
Tolbatov I, Marrone A, Coletti C, Re N. Computational Studies of Au(I) and Au(III) Anticancer MetalLodrugs: A Survey. Molecules 2021; 26:7600. [PMID: 34946684 PMCID: PMC8707411 DOI: 10.3390/molecules26247600] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/29/2021] [Accepted: 12/12/2021] [Indexed: 11/16/2022] Open
Abstract
Owing to the growing hardware capabilities and the enhancing efficacy of computational methodologies, computational chemistry approaches have constantly become more important in the development of novel anticancer metallodrugs. Besides traditional Pt-based drugs, inorganic and organometallic complexes of other transition metals are showing increasing potential in the treatment of cancer. Among them, Au(I)- and Au(III)-based compounds are promising candidates due to the strong affinity of Au(I) cations to cysteine and selenocysteine side chains of the protein residues and to Au(III) complexes being more labile and prone to the reduction to either Au(I) or Au(0) in the physiological milieu. A correct prediction of metal complexes' properties and of their bonding interactions with potential ligands requires QM computations, usually at the ab initio or DFT level. However, MM, MD, and docking approaches can also give useful information on their binding site on large biomolecular targets, such as proteins or DNA, provided a careful parametrization of the metal force field is employed. In this review, we provide an overview of the recent computational studies of Au(I) and Au(III) antitumor compounds and of their interactions with biomolecular targets, such as sulfur- and selenium-containing enzymes, like glutathione reductases, glutathione peroxidase, glutathione-S-transferase, cysteine protease, thioredoxin reductase and poly (ADP-ribose) polymerase 1.
Collapse
Affiliation(s)
- Iogann Tolbatov
- Institut de Chimie Moleculaire de l’Université de Bourgogne (ICMUB), Université de Bourgogne Franche-Comté (UBFC), Avenue Alain Savary 9, 21078 Dijon, France;
| | - Alessandro Marrone
- Dipartimento di Farmacia, Università degli Studi “G. D’Annunzio” Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (A.M.); (C.C.)
| | - Cecilia Coletti
- Dipartimento di Farmacia, Università degli Studi “G. D’Annunzio” Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (A.M.); (C.C.)
| | - Nazzareno Re
- Dipartimento di Farmacia, Università degli Studi “G. D’Annunzio” Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (A.M.); (C.C.)
| |
Collapse
|
26
|
Abdalbari FH, Telleria CM. The gold complex auranofin: new perspectives for cancer therapy. Discov Oncol 2021; 12:42. [PMID: 35201489 PMCID: PMC8777575 DOI: 10.1007/s12672-021-00439-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022] Open
Abstract
Advanced stages of cancer are highly associated with short overall survival in patients due to the lack of long-term treatment options following the standard form of care. New options for cancer therapy are needed to improve the survival of cancer patients without disease recurrence. Auranofin is a clinically approved agent against rheumatoid arthritis that is currently enrolled in clinical trials for potential repurposing against cancer. Auranofin mainly targets the anti-oxidative system catalyzed by thioredoxin reductase (TrxR), which protects the cell from oxidative stress and death in the cytoplasm and the mitochondria. TrxR is over-expressed in many cancers as an adaptive mechanism for cancer cell proliferation, rendering it an attractive target for cancer therapy, and auranofin as a potential therapeutic agent for cancer. Inhibiting TrxR dysregulates the intracellular redox state causing increased intracellular reactive oxygen species levels, and stimulates cellular demise. An alternate mechanism of action of auranofin is to mimic proteasomal inhibition by blocking the ubiquitin-proteasome system (UPS), which is critically important in cancer cells to prevent cell death when compared to non-cancer cells, because of its role on cell cycle regulation, protein degradation, gene expression, and DNA repair. This article provides new perspectives on the potential mechanisms used by auranofin alone, in combination with diverse other compounds, or in combination with platinating agents and/or immune checkpoint inhibitors to combat cancer cells, while assessing the feasibility for its repurposing in the clinical setting.
Collapse
Affiliation(s)
- Farah H Abdalbari
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Carlos M Telleria
- Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.
- Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
27
|
Cirri D, Landini I, Massai L, Mini E, Maestrelli F, Messori L. Cyclodextrin Inclusion Complexes of Auranofin and Its Iodido Analog: A Chemical and Biological Study. Pharmaceutics 2021; 13:pharmaceutics13050727. [PMID: 34063389 PMCID: PMC8155929 DOI: 10.3390/pharmaceutics13050727] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 01/25/2023] Open
Abstract
Auranofin (AF) and its iodido analog, i.e., Au(PEt3) I (AFI), were reported to exhibit very promising anticancer properties both in vitro and in vivo. However, both these gold compounds have a scarce aqueous solubility that hampers their pharmaceutical use. Here, we explore whether encapsulation of these metallodrugs inside hydroxypropyl-beta–cyclodextrin (HPβ–CD) may lead to an improved biopharmaceutical profile for the resulting adducts. Phase solubility studies, performed at 25 °C in an aqueous buffer, revealed, in both cases, the formation of a 1:1 drug to cyclodextrin complex; a far greater apparent stability constant (K1:1) was measured for AFI compared to AF (331 M−1 versus ca. 30 M−1). NMR studies conducted on the AFI/HPβ–CD system confirmed the formation of a stable 1:1 adduct. Then, binary systems of AF and AFI with HPβ–CD were prepared by colyophilization and characterized by DSC and PXRD. The results revealed the occurrence of drug complexation and/or amorphization for the AFI/HPβ–CD binary system. Afterwards, the antiproliferative properties of the two cyclodextrin adducts and of the corresponding free drugs were comparatively evaluated in vitro in three representative ovarian cancer cell lines, i.e., A2780, SKOV3, and IGROV-1. The results, in all cases, point out that CD complexation of the two gold drugs does not substantially affect their biological activity. The implications of these findings are discussed in the frame of the current knowledge of AF and its analogs.
Collapse
Affiliation(s)
- Damiano Cirri
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Moruzzi 13, 56124 Pisa, Italy;
| | - Ida Landini
- Department of Health Sciences, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy;
| | - Lara Massai
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy;
| | - Enrico Mini
- Department of Health Sciences, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy;
- Correspondence: (E.M.); (F.M.); (L.M.)
| | - Francesca Maestrelli
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy;
- Correspondence: (E.M.); (F.M.); (L.M.)
| | - Luigi Messori
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy;
- Correspondence: (E.M.); (F.M.); (L.M.)
| |
Collapse
|
28
|
Chupakhin E, Krasavin M. Thioredoxin reductase inhibitors: updated patent review (2017-present). Expert Opin Ther Pat 2021; 31:745-758. [PMID: 33666133 DOI: 10.1080/13543776.2021.1899160] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Thioredoxin reductase (TrxR) is a selenocysteine-containing enzyme which is responsible - as a part of the thioredoxin system - for maintaining redox homeostasis in cells. It is upregulated in cancerous state as a defense against oxidative stress. TrxR has been mostly considered an anticancer drug target although it has applications in other therapeutic areas such as neurodegeneration, inflammation, microbial infections, and neonatal hyperoxic lung injury.Areas covered: The present review covers the patent literature that appeared in the period 2017-2020, i.e. since the publication of the previous expert opinion patent review on TrxR inhibitors. The recent additions to the following traditional classes of inhibitors are discussed: metal complexes, Michael acceptors as well as arsenic and selenium compounds. At the same time, a novel group of nitro (hetero)aromatic compounds have emerged which likely acts via covalent inhibition mechanism. Several miscellaneous chemotypes are grouped under Miscellaneous subsection.Expert opinion: While specificity over glutathione reductase is achieved easily, TrxR is still moving toward the later stages of development at a very slow rate. Michael acceptors, particularly based on TRXR substrate-mimicking scaffolds, are gaining impetus and so are dual and hybrid compounds. The development prospects of the emerging nitro (hetero)aromatic chemotypes remain uncertain.
Collapse
Affiliation(s)
- Evgeny Chupakhin
- Institute of Chemistry, Saint Petersburg State University, Saint Petersburg Russian Federation.,Institute for Living Systems, Immanuel Kant Baltic Federal University, Kaliningrad Russian Federation
| | - Mikhail Krasavin
- Institute of Chemistry, Saint Petersburg State University, Saint Petersburg Russian Federation.,Institute for Living Systems, Immanuel Kant Baltic Federal University, Kaliningrad Russian Federation
| |
Collapse
|
29
|
Silva DES, Becceneri AB, Santiago JVB, Gomes Neto JA, Ellena J, Cominetti MR, Pereira JCM, Hannon MJ, Netto AVG. Silver(I) complexes of 3-methoxy-4-hydroxybenzaldehyde thiosemicarbazones and triphenylphosphine: structural, cytotoxicity, and apoptotic studies. Dalton Trans 2020; 49:16474-16487. [PMID: 32914824 DOI: 10.1039/d0dt01134g] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Novel silver(i) complexes of the type [AgCl(PPh3)2(L)] {PPh3 = triphenylphosphine; L = VTSC = 3-methoxy-4-hydroxybenzaldehyde thiosemicarbazone (1); VMTSC = 3-methoxy-4-[2-(morpholine-1-yl)ethoxy]benzaldehyde thiosemicarbazone (2); VPTSC = 3-methoxy-4-[2-(piperidine-1-yl)ethoxy]benzaldehyde thiosemicarbazone (3)} were synthesized and fully characterized by spectroscopic techniques. The molecular structures of complexes 2 and 3 were determined by single crystal X-ray diffraction. Compounds 1-3 exhibited appreciable cytotoxic activity against human tumor cells (lung A549, breast MDA-MB-231 and MCF-7) with IC50 values in 48 h of incubation ranging from 5.6 to 18 μM. Cellular uptake studies showed that complexes 1-3 were efficiently internalized after 3 hours of treatment in MDA-MB-231 cells. The effects of complex 1 on the cell morphology, cell cycle, induction of apoptosis, mitochondrial membrane potential (Δψm), and reactive oxygen species (ROS) production have been evaluated in triple negative breast cancer (TNBC) cells MDA-MB-231. Our results showed that complex 1 induced typical morphological alterations of cell death, an increase in cells at the sub-G1 phase, apoptosis, and mitochondrial membrane depolarization. Furthermore, DNA binding studies evidenced that 1 can bind to ct-DNA and does so without modifying the B-structure of the DNA, but that the binding is weak compared to that of Hoechst 33258.
Collapse
Affiliation(s)
- Débora E S Silva
- Department of General and Inorganic Chemistry, Department of Analytical Chemistry, UNESP - São Paulo State University, Institute of Chemistry, CEP 14800-060 Araraquara, SP, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Antiproliferative Properties of a Few Auranofin-Related Gold(I) and Silver(I) Complexes in Leukemia Cells and their Interferences with the Ubiquitin Proteasome System. Molecules 2020; 25:molecules25194454. [PMID: 32998355 PMCID: PMC7582876 DOI: 10.3390/molecules25194454] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 01/01/2023] Open
Abstract
A group of triethylphosphine gold(I) and silver(I) complexes, structurally related to auranofin, were prepared and investigated as potential anticancer drug candidates. The antiproliferative properties of these metal compounds were assessed against two leukemia cell lines, i.e., CCRF-CEM and its multidrug-resistant counterpart, CEM/ADR5000. Interestingly, potent cytotoxic effects were disclosed for both series of compounds against leukemia cells, with IC50 values generally falling in the low-micromolar range, the gold derivatives being on the whole more effective than the silver analogues. Some initial structure-function relationships were drawn. Subsequently, the ability of the study compounds to inhibit the three main catalytic activities of the proteasome was investigated. Different patterns of enzyme inhibition emerged for the various metal complexes. Notably, gold compounds were able to inhibit effectively both the trypsin-like and chymotrypsin-like proteasome activities, being less effective toward the caspase-like catalytic activity. In most cases, a significant selectivity of the study compounds toward the proteasome proteolytic activities was detected when compared to other proteases. The implications of the obtained results are discussed.
Collapse
|