1
|
Balgera F, Tijani MK, Wennerberg J, Persson KEM, Nordlander E, Ferreira RJ. Evaluation of Au(III) complexes as Plasmodium falciparum aquaglyceroporin (PfAQP) inhibitors by in silico and in vitro methods. J Biol Inorg Chem 2024; 29:821-836. [PMID: 39579246 DOI: 10.1007/s00775-024-02081-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: 06/24/2024] [Accepted: 10/20/2024] [Indexed: 11/25/2024]
Abstract
The onset of resistance to artemisinin for malaria treatment has stimulated the quest for novel antimalarial drugs. Herein, the gold(III) coordination complexes Aubipy [Au(bipy)Cl]+ (bipy = 2,2'-bipyridine), Auphen [Au(phen)Cl]+ (phen = phenanthroline), Auterpy [Au(terpy)Cl]2+ (terpy = 2,2';6',2″-terpyridine), and corresponding hydrolyzed species, have been investigated as inhibitors of the Plasmodium falciparum aquaglyceroporin (PfAQP) protein by computational methods. Through an in-silico approach using an Umbrella Sampling protocol to sample how Aubipy, Auphen, and Auterpy permeate through the PfAQP, their permeability coefficients were estimated using the Inhomogeneous Solubility Diffusion (ISD) model with promising results. The efficacy of the gold complexes was then probed by an in vitro assay testing the growth inhibition in chloroquine sensitive and resistant P. falciparum strains. In accordance with the computational data, Auterpy achieved the highest efficiency with an IC50 in the nanomolar range (590 nM) on resistant strain cultures, additionally revealing a good selectivity as compared to its activity against the human aquaglyceroporin 3.
Collapse
Affiliation(s)
- Federico Balgera
- Red Glead Discovery AB, Medicon Village, Scheelevägen 10, 223 63, Lund, Sweden
- Chemical Physics, Department of Chemistry, Lund University, Box 124, 221 00, Lund, Sweden
| | | | - Johan Wennerberg
- Red Glead Discovery AB, Medicon Village, Scheelevägen 10, 223 63, Lund, Sweden
- Organic Chemistry, Faculty of Engineering, Lund University, Box 124, 221 00, Lund, Sweden
| | - Kristina E M Persson
- Department of Laboratory Medicine, Lund University, Lund, Sweden
- Clinical Chemistry and Pharmacology, Laboratory Medicine, Office for Medical Services, Region Skåne, Lund, Sweden
| | - Ebbe Nordlander
- Chemical Physics, Department of Chemistry, Lund University, Box 124, 221 00, Lund, Sweden
| | - Ricardo J Ferreira
- Red Glead Discovery AB, Medicon Village, Scheelevägen 10, 223 63, Lund, Sweden.
| |
Collapse
|
2
|
Andrés CMC, Pérez de la Lastra JM, Bustamante Munguira E, Andrés Juan C, Pérez-Lebeña E. Anticancer Activity of Metallodrugs and Metallizing Host Defense Peptides-Current Developments in Structure-Activity Relationship. Int J Mol Sci 2024; 25:7314. [PMID: 39000421 PMCID: PMC11242492 DOI: 10.3390/ijms25137314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
This article provides an overview of the development, structure and activity of various metal complexes with anti-cancer activity. Chemical researchers continue to work on the development and synthesis of new molecules that could act as anti-tumor drugs to achieve more favorable therapies. It is therefore important to have information about the various chemotherapeutic substances and their mode of action. This review focuses on metallodrugs that contain a metal as a key structural fragment, with cisplatin paving the way for their chemotherapeutic application. The text also looks at ruthenium complexes, including the therapeutic applications of phosphorescent ruthenium(II) complexes, emphasizing their dual role in therapy and diagnostics. In addition, the antitumor activities of titanium and gold derivatives, their side effects, and ongoing research to improve their efficacy and reduce adverse effects are discussed. Metallization of host defense peptides (HDPs) with various metal ions is also highlighted as a strategy that significantly enhances their anticancer activity by broadening their mechanisms of action.
Collapse
Affiliation(s)
| | - José Manuel Pérez de la Lastra
- Institute of Natural Products and Agrobiology, CSIC-Spanish Research Council, Avda. Astrofísico Fco. Sánchez, 3, 38206 La Laguna, Spain
| | | | - Celia Andrés Juan
- Cinquima Institute and Department of Organic Chemistry, Faculty of Sciences, Valladolid University, Paseo de Belén, 7, 47011 Valladolid, Spain
| | | |
Collapse
|
3
|
Mattioli EJ, Cipriani B, Zerbetto F, Marforio TD, Calvaresi M. Interaction of Au(III) with amino acids: a vade mecum for medicinal chemistry and nanotechnology. J Mater Chem B 2024; 12:5162-5170. [PMID: 38687242 DOI: 10.1039/d4tb00204k] [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: 05/02/2024]
Abstract
Au(III) is highly reactive. At odds with its reduced counterpart, Au(I), it is hardly present in structural databases. And yet, it is the starting reactant to form gold nanoclusters (AuNCs) and the constitutive component of a new class of drugs. Its reactivity is a world apart from that of the iso-electronic Pt(II) species. Rather than DNA, it targets proteins. Its interaction with amino acid residues is manifold. It can strongly interact with the residue backbones, amino acid side chains and protein ends, it can form appropriate complexes whose stabilization energy reaches up to more than 40 kcal mol-1, it can affect the pKa of amino acid residues, and it can promote charge transfer from the residues to the amount that it is reduced. Here, quantum chemical calculations provide quantitative information on all the processes where Au(III) can be involved. A myriad of structural arrangements are examined in order to determine the strongest interactions and quantify the amount of charge transfer between protonated and deprotonated residues and Au(III). The calculated interaction energies of the amino acid side chains with Au(III) quantitatively reproduce the experimental tendency of Au(III) to interact with selenocysteine, cysteine and histidine and negatively charged amino acids such as Glu and Asp. Also, aromatic residues such as tyrosine and tryptophan strongly interact with Au(III). In proteins, basic pH plays a role in the deprotonation of cysteine, lysine and tyrosine and strongly increases the binding affinity of Au(III) toward these amino acids. The amino acid residues in the protein can also trigger the reduction of Au(III) ions. Sulfur-containing amino acids (cysteine and methionine) and selenocysteine provide almost one electron to Au(III) upon binding. Tyrosine also shows a considerable tendency to act as a reductant. Other amino acids, commonly identified in Au-protein adducts, such as Ser, Trp, Thr, Gln, Glu, Asn, Asp, Lys, Arg and His, possess a notable reducing power toward Au(III). These results and their discussion form a vade mecum that can find application in medicinal chemistry and nanotech applications of Au(III).
Collapse
Affiliation(s)
- Edoardo Jun Mattioli
- Dipartimento di Chimica ''G. Ciamician'', Alma Mater Studiorum - Universita di Bologna, via F. Selmi 2, 40126 Bologna, Italy.
| | - Beatrice Cipriani
- Dipartimento di Chimica ''G. Ciamician'', Alma Mater Studiorum - Universita di Bologna, via F. Selmi 2, 40126 Bologna, Italy.
| | - Francesco Zerbetto
- Dipartimento di Chimica ''G. Ciamician'', Alma Mater Studiorum - Universita di Bologna, via F. Selmi 2, 40126 Bologna, Italy.
| | - Tainah Dorina Marforio
- Dipartimento di Chimica ''G. Ciamician'', Alma Mater Studiorum - Universita di Bologna, via F. Selmi 2, 40126 Bologna, Italy.
| | - Matteo Calvaresi
- Dipartimento di Chimica ''G. Ciamician'', Alma Mater Studiorum - Universita di Bologna, via F. Selmi 2, 40126 Bologna, Italy.
| |
Collapse
|
4
|
Borges AP, Obata MMS, Libardi SH, Trevisan RO, Deflon VM, Abram U, Ferreira FB, Costa LAS, Patrocínio AOT, da Silva MV, Borges JC, Maia PIS. Gold(I) and Silver(I) Complexes Containing Hybrid Sulfonamide/Thiourea Ligands as Potential Leishmanicidal Agents. Pharmaceutics 2024; 16:452. [PMID: 38675113 PMCID: PMC11053681 DOI: 10.3390/pharmaceutics16040452] [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: 02/29/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Leishmaniasis is a group of parasitic diseases with the potential to infect more than 1 billion people; however, its treatment is still old and inadequate. In order to contribute to changing this view, this work consisted of the development of complexes derived from MI metal ions with thioureas, aiming to obtain potential leishmanicidal agents. The thiourea ligands (HLR) were obtained by reactions of p-toluenesulfohydrazide with R-isothiocyanates and were used in complexation reactions with AgI and AuI, leading to the formation of complexes of composition [M(HLR)2]X (M = Ag or Au; X = NO3- or Cl-). All compounds were characterized by FTIR, 1H NMR, UV-vis, emission spectroscopy and elemental analysis. Some representatives were additionally studied by ESI-MS and single-crystal XRD. Their properties were further analyzed by DFT calculations. Their cytotoxicity on Vero cells and the extracellular leishmanicidal activity on Leishmania infantum and Leishmania braziliensis cells were evaluated. Additionally, the interaction of the complexes with the Old Yellow enzyme of the L. braziliensis (LbOYE) was examined. The biological tests showed that some compounds present remarkable leishmanicidal activity, even higher than that of the standard drug Glucantime, with different selectivity for the two species of Leishmania. Finally, the interaction studies with LbOYE revealed that this enzyme could be one of their biological targets.
Collapse
Affiliation(s)
- Alice P. Borges
- Bioactive Compounds Development Research Group, Federal University of Triângulo Mineiro, Av. Dr. Randolfo Borges 1400, Uberaba 38025-440, MG, Brazil;
| | - Malu M. S. Obata
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Av. Getúlio Guarita, Uberaba 38025-440, MG, Brazil; (M.M.S.O.); (R.O.T.); (M.V.d.S.)
| | - Silvia H. Libardi
- São Carlos Institute of Chemistry, University of São Paulo, Av. Trabalhador São Carlense, 400, São Carlos 13566-590, SP, Brazil; (S.H.L.); (V.M.D.); (J.C.B.)
| | - Rafael O. Trevisan
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Av. Getúlio Guarita, Uberaba 38025-440, MG, Brazil; (M.M.S.O.); (R.O.T.); (M.V.d.S.)
| | - Victor M. Deflon
- São Carlos Institute of Chemistry, University of São Paulo, Av. Trabalhador São Carlense, 400, São Carlos 13566-590, SP, Brazil; (S.H.L.); (V.M.D.); (J.C.B.)
| | - Ulrich Abram
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 34/36, 14195 Berlin, Germany;
| | - Francis B. Ferreira
- Associated Faculties of Uberaba, Av. do Tutuna 720, Uberaba 38061-500, MG, Brazil;
| | - Luiz Antônio S. Costa
- Computational Chemistry Research Group, Institute of Exact Sciences, Federal University of Juiz de Fora, Campus Universitário s/n Martelos, Juiz de Fora 36036-900, MG, Brazil;
| | - Antonio O. T. Patrocínio
- Laboratory of Photochemistry and Material Science, Institute of Chemistry, Federal University of Uberlândia, Av. João Naves de Ávila 2121, Uberlândia 38400-902, MG, Brazil;
| | - Marcos V. da Silva
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Av. Getúlio Guarita, Uberaba 38025-440, MG, Brazil; (M.M.S.O.); (R.O.T.); (M.V.d.S.)
| | - Júlio C. Borges
- São Carlos Institute of Chemistry, University of São Paulo, Av. Trabalhador São Carlense, 400, São Carlos 13566-590, SP, Brazil; (S.H.L.); (V.M.D.); (J.C.B.)
| | - Pedro I. S. Maia
- Bioactive Compounds Development Research Group, Federal University of Triângulo Mineiro, Av. Dr. Randolfo Borges 1400, Uberaba 38025-440, MG, Brazil;
| |
Collapse
|
5
|
Baitullina A, Claude G, Sucena SF, Nisli E, Scholz C, Bhardwaj P, Amthauer H, Brenner W, Geppert C, Gorges C, Abram U, da Silva Maia PI, Spreckelmeyer S. Metallacages with 2,6-dipicolinoylbis(N,N-dialkylthioureas) as novel platforms in nuclear medicine for 68Ga, 177Lu and 198Au. EJNMMI Radiopharm Chem 2023; 8:40. [PMID: 37982944 PMCID: PMC10661681 DOI: 10.1186/s41181-023-00225-z] [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: 09/05/2023] [Accepted: 11/06/2023] [Indexed: 11/21/2023] Open
Abstract
BACKGROUND Heterometallic gold metallacages are of great interest for the incorporation of several cations. Especially in nuclear medicine, those metallacages can serve as a platform for radionuclides relevant for imaging or therapy (e.g. 68Ga or 177Lu). Moreover, the radionuclide 198Au is an attractive beta emitter, for potential application in nuclear medicine. Here, we aim to synthesize a new set of gold metallacages and to study their ability to coordinate to 68Ga, 177Lu and 198Au. RESULTS New heterometallic gold metallacages of composition [M{Au(Lmorph-κS)}3] (M = La3+, Tb3+, Lu3+ or Y3+) and [Ga{Au(Lmorph-κS)}2]NO3 have been synthesized from 2,6-dipicolinoylbis(N,N-morpholinylthiourea) (H2Lmorph) with [AuCl(THT)] and the target M3+ metal ions in yields ranging from 33 (Lu) to 62% (Tb). The characterization of the compounds bases on ESI-MS, 1H NMR, IR, EA and single-crystal X-ray diffraction techniques (all except the Ga derivative). Selected gold cages derived from H2Lmorph were compared to previously reported gold cages that were derived from 2,6-dipicolinoylbis(N,N-diethylthiourea) (H2Ldiethyl). The tested metallacages show similar IC50 values close to that of auranofin in four different cancer cell lines (MCF-7, PC-3, U383, U343), e.g. 4.5 ± 0.7 µM for [Ga{Au(Ldiethyl)}2]NO3 on PC-3. The radiolabeling experiments thereof show high radiochemical purities with 68Ga and 198Au and low radiochemical purity with 177Lu. CONCLUSIONS The results indicate that these gold metallacages could serve as a novel platform for inclusion of different (radio)nuclides with potential theranostic applications in nuclear medicine.
Collapse
Affiliation(s)
- Anna Baitullina
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 34-36, 14195, Berlin, Germany
| | - Guilhem Claude
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 34-36, 14195, Berlin, Germany
| | - Suelen F Sucena
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 34-36, 14195, Berlin, Germany
| | - Eda Nisli
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Nuclear Medicine, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Cedric Scholz
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 34-36, 14195, Berlin, Germany
| | - Punita Bhardwaj
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Nuclear Medicine, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Holger Amthauer
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Nuclear Medicine, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Winfried Brenner
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Nuclear Medicine, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Christopher Geppert
- Forschungsreaktor TRIGA Mainz, Johannes Gutenberg-Universität Mainz, Fritz-Strassmann-Weg 2, 55128, Mainz, Germany
| | - Christian Gorges
- Forschungsreaktor TRIGA Mainz, Johannes Gutenberg-Universität Mainz, Fritz-Strassmann-Weg 2, 55128, Mainz, Germany
| | - Ulrich Abram
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 34-36, 14195, Berlin, Germany
| | - Pedro Ivo da Silva Maia
- Núcleo de Desenvolvimento de Compostos Bioativos (NDCBio), Universidade Federal do Triângulo Mineiro, Uberaba, MG, 38025-440, Brazil.
| | - Sarah Spreckelmeyer
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Nuclear Medicine, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
6
|
Sucena SF, Demirer TI, Baitullina A, Hagenbach A, Grewe J, Spreckelmeyer S, März J, Barkleit A, Maia PIDS, Nguyen HH, Abram U. Gold-Based Coronands as Hosts for M 3+ Metal Ions: Ring Size Matters. Molecules 2023; 28:5421. [PMID: 37513293 PMCID: PMC10385047 DOI: 10.3390/molecules28145421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The controlled, self-assembled synthesis of multinuclear coordination compounds can be performed via different approaches. Frequently, steric, geometric and/or electronic factors located at the ligand systems predefine the way in which metal ions can assemble them to large aggregates. For the compounds in the present paper, also the Pearson's acidities and preferred coordination geometries of the metal ions were used as organization principles. The ligand under study, 2,6-dipicolinoylbis(N,N-diethylthiourea), H2L1ethyl, possesses 'soft' sulfur and 'hard' nitrogen and oxygen donors. One-pot reactions of this compound with [AuCl(tht)] (tht = tetrahydrothiophene) and M3+ salts (M = Sc, Y, La, Ln, Ga, In) give products with gold-based {Au3(L1ethyl)3}3+ or {Au2(L1ethyl)2}2+ coronands, which host central M3+ ions. The formation of such units is templated by the M3+ ions and the individual size of the coronand rings is dependent on the ionic radii of the central ions in a way that small ions such as Ga3+ form a [Ga⊂{Au2(L1ethyl)2}]+ assembly, while larger ions (starting from Sc3+/In3+) establish neutral [M⊂{Au3(L1ethyl)3}] units with nine-coordinate central ions.
Collapse
Affiliation(s)
- Suelen Ferreira Sucena
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 34/36, 14195 Berlin, Germany
| | - Türkan Ilgin Demirer
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 34/36, 14195 Berlin, Germany
| | - Anna Baitullina
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 34/36, 14195 Berlin, Germany
| | - Adelheid Hagenbach
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 34/36, 14195 Berlin, Germany
| | - Jacqueline Grewe
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 34/36, 14195 Berlin, Germany
| | - Sarah Spreckelmeyer
- Department of Nuclear Medicine and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Juliane März
- Institute of Resource Ecology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstr. 400, 01328 Dresden, Germany
| | - Astrid Barkleit
- Institute of Resource Ecology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstr. 400, 01328 Dresden, Germany
| | - Pedro Ivo da Silva Maia
- Núcleo de Desenvolvimento de Compostos Bioativos (NDCBio), Universidade Federal do Triângulo Mineiro, Av. Dr. Randolfo Borges 1400, Uberaba 38025-440, MG, Brazil
| | - Hung Huy Nguyen
- Department of Inorganic Chemistry, VNU University of Science, 19 Le Thanh Tong, Hanoi 100000, Vietnam
| | - Ulrich Abram
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 34/36, 14195 Berlin, Germany
| |
Collapse
|
7
|
Ratia C, Ballén V, Gabasa Y, Soengas RG, Velasco-de Andrés M, Iglesias MJ, Cheng Q, Lozano F, Arnér ESJ, López-Ortiz F, Soto SM. Novel gold(III)-dithiocarbamate complex targeting bacterial thioredoxin reductase: antimicrobial activity, synergy, toxicity, and mechanistic insights. Front Microbiol 2023; 14:1198473. [PMID: 37333656 PMCID: PMC10272563 DOI: 10.3389/fmicb.2023.1198473] [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: 04/01/2023] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction Antimicrobial resistance is a pressing global concern that has led to the search for new antibacterial agents with novel targets or non-traditional approaches. Recently, organogold compounds have emerged as a promising class of antibacterial agents. In this study, we present and characterize a (C^S)-cyclometallated Au(III) dithiocarbamate complex as a potential drug candidate. Methods and results The Au(III) complex was found to be stable in the presence of effective biological reductants, and showed potent antibacterial and antibiofilm activity against a wide range of multidrug-resistant strains, particularly gram-positive strains, and gram-negative strains when used in combination with a permeabilizing antibiotic. No resistant mutants were detected after exposing bacterial cultures to strong selective pressure, indicating that the complex may have a low propensity for resistance development. Mechanistic studies indicate that the Au(III) complex exerts its antibacterial activity through a multimodal mechanism of action. Ultrastructural membrane damage and rapid bacterial uptake suggest direct interactions with the bacterial membrane, while transcriptomic analysis identified altered pathways related to energy metabolism and membrane stability including enzymes of the TCA cycle and fatty acid biosynthesis. Enzymatic studies further revealed a strong reversible inhibition of the bacterial thioredoxin reductase. Importantly, the Au(III) complex demonstrated low cytotoxicity at therapeutic concentrations in mammalian cell lines, and showed no acute in vivo toxicity in mice at the doses tested, with no signs of organ toxicity. Discussion Overall, these findings highlight the potential of the Au(III)-dithiocarbamate scaffold as a basis for developing novel antimicrobial agents, given its potent antibacterial activity, synergy, redox stability, inability to produce resistant mutants, low toxicity to mammalian cells both in vitro and in vivo, and non-conventional mechanism of action.
Collapse
Affiliation(s)
- Carlos Ratia
- Barcelona Institute for Global Health (ISGlobal), Universitat de Barcelona, Barcelona, Spain
| | - Victoria Ballén
- Barcelona Institute for Global Health (ISGlobal), Universitat de Barcelona, Barcelona, Spain
| | - Yaiza Gabasa
- Barcelona Institute for Global Health (ISGlobal), Universitat de Barcelona, Barcelona, Spain
| | - Raquel G. Soengas
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, Almería, Spain
| | | | - María José Iglesias
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, Almería, Spain
| | - Qing Cheng
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Francisco Lozano
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Servei d’Immunologia, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain
- Department de Biomedicina, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Elias S. J. Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Department of Selenoprotein Research and the National Tumor Biology Laboratory, Budapest, Hungary
| | - Fernando López-Ortiz
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, Almería, Spain
| | - Sara M. Soto
- Barcelona Institute for Global Health (ISGlobal), Universitat de Barcelona, Barcelona, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
8
|
Mertens RT, Gukathasan S, Arojojoye AS, Olelewe C, Awuah SG. Next Generation Gold Drugs and Probes: Chemistry and Biomedical Applications. Chem Rev 2023; 123:6612-6667. [PMID: 37071737 PMCID: PMC10317554 DOI: 10.1021/acs.chemrev.2c00649] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
The gold drugs, gold sodium thiomalate (Myocrisin), aurothioglucose (Solganal), and the orally administered auranofin (Ridaura), are utilized in modern medicine for the treatment of inflammatory arthritis including rheumatoid and juvenile arthritis; however, new gold agents have been slow to enter the clinic. Repurposing of auranofin in different disease indications such as cancer, parasitic, and microbial infections in the clinic has provided impetus for the development of new gold complexes for biomedical applications based on unique mechanistic insights differentiated from auranofin. Various chemical methods for the preparation of physiologically stable gold complexes and associated mechanisms have been explored in biomedicine such as therapeutics or chemical probes. In this Review, we discuss the chemistry of next generation gold drugs, which encompasses oxidation states, geometry, ligands, coordination, and organometallic compounds for infectious diseases, cancer, inflammation, and as tools for chemical biology via gold-protein interactions. We will focus on the development of gold agents in biomedicine within the past decade. The Review provides readers with an accessible overview of the utility, development, and mechanism of action of gold-based small molecules to establish context and basis for the thriving resurgence of gold in medicine.
Collapse
Affiliation(s)
- R Tyler Mertens
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Sailajah Gukathasan
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Adedamola S Arojojoye
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Chibuzor Olelewe
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
- University of Kentucky Markey Cancer Center, Lexington, Kentucky 40536, United States
| |
Collapse
|
9
|
Moreno-Alcántar G, Picchetti P, Casini A. Gold Complexes in Anticancer Therapy: From New Design Principles to Particle-Based Delivery Systems. Angew Chem Int Ed Engl 2023; 62:e202218000. [PMID: 36847211 DOI: 10.1002/anie.202218000] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 02/28/2023]
Abstract
The discovery of the medicinal properties of gold complexes has fuelled the design and synthesis of new anticancer metallodrugs, which have received special attention due to their unique modes of action. Current research in the development of gold compounds with therapeutic properties is predominantly focused on the molecular design of drug leads with superior pharmacological activities, e.g., by introducing targeting features. Moreover, intensive research aims at improving the physicochemical properties of gold compounds, such as chemical stability and solubility in the physiological environment. In this regard, the encapsulation of gold compounds in nanocarriers or their chemical grafting onto targeted delivery vectors could lead to new nanomedicines that eventually reach clinical applications. Herein, we provide an overview of the state-of-the-art progress of gold anticancer compounds, andmore importantly we thoroughly revise the development of nanoparticle-based delivery systems for gold chemotherapeutics.
Collapse
Affiliation(s)
- Guillermo Moreno-Alcántar
- Chair of Medicinal and Bioinorganic Chemistry, School of Natural Sciences, Department of Chemistry, Technical University of Munich (TUM), Lichtenbergstr. 4, 85748, Garching b. München, Germany
| | - Pierre Picchetti
- Karlsruhe Institute of Technology (KIT), Institute of Nanotechnology, Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Angela Casini
- Chair of Medicinal and Bioinorganic Chemistry, School of Natural Sciences, Department of Chemistry, Technical University of Munich (TUM), Lichtenbergstr. 4, 85748, Garching b. München, Germany
| |
Collapse
|
10
|
Ratia C, Sueiro S, Soengas RG, Iglesias MJ, López-Ortiz F, Soto SM. Gold(III) Complexes Activity against Multidrug-Resistant Bacteria of Veterinary Significance. Antibiotics (Basel) 2022; 11:antibiotics11121728. [PMID: 36551386 PMCID: PMC9774617 DOI: 10.3390/antibiotics11121728] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
The emergence and spread of multidrug-resistant bacteria are a global concern. The lack of new antibiotics in the pipeline points to the need for developing new strategies. In this sense, gold(III) complexes (G3Cs) could be a promising alternative due to their recently described antibacterial activity. The aim of this study was to evaluate the antimicrobial activity of G3Cs alone and in combination with colistin against pathogenic bacteria from veterinary sources. Minimal inhibitory concentration (MIC) values were determined by broth microdilution and compared with clinically relevant antibiotics. Antibiofilm activity was determined by crystal violet staining. Combinations of selected G3Cs with colistin and cytotoxicity in commercial human cell lines were evaluated. Four and seven G3Cs showed antibacterial effect against Gram-negative and Gram-positive strains, respectively, with this activity being higher among Gram-positive strains. The G3Cs showed antibiofilm activity against Gram-negative species at concentrations similar or one to four folds higher than the corresponding MICs. Combination of G3Cs with colistin showed a potential synergistic antibacterial effect reducing concentrations and toxicity of both agents. The antimicrobial and antibiofilm activity, the synergistic effect when combined with colistin and the in vitro toxicity suggest that G3Cs would provide a new therapeutic alternative against multidrug-resistant bacteria from veterinary origin.
Collapse
Affiliation(s)
- Carlos Ratia
- ISGlobal, Hospital Clínic—Universitat de Barcelona, 08036 Barcelona, Spain
| | - Sara Sueiro
- ISGlobal, Hospital Clínic—Universitat de Barcelona, 08036 Barcelona, Spain
| | - Raquel G. Soengas
- Departamento de Química Orgánica e Inorgánica, Universidad de Oviedo, Julián Clavería 7, 33006 Oviedo, Spain
| | - María José Iglesias
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, 04120 Almería, Spain
| | - Fernando López-Ortiz
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, 04120 Almería, Spain
- Correspondence: (F.L.-O.); (S.M.S.)
| | - Sara María Soto
- ISGlobal, Hospital Clínic—Universitat de Barcelona, 08036 Barcelona, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (F.L.-O.); (S.M.S.)
| |
Collapse
|
11
|
Banerjee S, Banerjee S. Metal-Based Complexes as Potential Anti-cancer Agents. Anticancer Agents Med Chem 2022; 22:2684-2707. [PMID: 35362388 DOI: 10.2174/1871520622666220331085144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/16/2021] [Accepted: 02/03/2022] [Indexed: 11/22/2022]
Abstract
Metal based therapy is no new in biomedical research. In early days the biggest limitation was the inequality among therapeutical and toxicological dosages. Ever since, Barnett Rosenberg discovered cisplatin, a new era has begun to treat cancer with metal complexes. Platinum complexes such as oxaliplatin, cisplatin, and carboplatin, seem to be the foundation of metal/s-based components to challenge malignancies. With an advancement in the biomolemoecular mechanism, researchers have started developing non-classical platinum-based complexes, where a different mechanistic approach of the complexes is observed towards the biomolecular target. Till date, larger number of metal/s-based complexes was synthesized by overhauling the present structures chemically by substituting the ligand or preparing the whole novel component with improved cytotoxic and safety profiles. Howsoever, due to elevated accentuation upon the therapeutic importance of metal/s-based components, a couple of those agents are at present on clinical trials and several other are in anticipating regulatory endorsement to enter the trial. This literature highlights the detailed heterometallic multinuclear components, primarily focusing on platinum, ruthenium, gold and remarks on possible stability, synergism, mechanistic studies and structure activity relationships.
Collapse
Affiliation(s)
- Sabyasachi Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Ashram More, G.T. Road, Asansol-713301, West Bengal, India
| | - Subhasis Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Ashram More, G.T. Road, Asansol-713301, West Bengal, India
| |
Collapse
|
12
|
Ratia C, Cepas V, Soengas R, Navarro Y, Velasco-de Andrés M, Iglesias MJ, Lozano F, López-Ortiz F, Soto SM. A C ∧S-Cyclometallated Gold(III) Complex as a Novel Antibacterial Candidate Against Drug-Resistant Bacteria. Front Microbiol 2022; 13:815622. [PMID: 35308343 PMCID: PMC8928146 DOI: 10.3389/fmicb.2022.815622] [Citation(s) in RCA: 7] [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: 11/15/2021] [Accepted: 01/13/2022] [Indexed: 12/14/2022] Open
Abstract
The worldwide emergence and spread of infections caused by multidrug-resistant bacteria endangers the efficacy of current antibiotics in the clinical setting. The lack of new antibiotics in the pipeline points to the need of developing new strategies. Recently, gold-based drugs are being repurposed for antibacterial applications. Among them, gold(III) complexes have received increasing attention as metal-based anticancer agents. However, reports on their antibacterial activity are scarce due to stability issues. The present work demonstrates the antibacterial activity of the gold(III) complex 2 stabilized as C∧S-cycloaurated containing a diphenylphosphinothioic amide moiety, showing minimum inhibitory concentration (MIC) values that ranged from 4 to 8 and from 16 to 32 mg/L among Gram-positive and Gram-negative multidrug-resistant (MDR) pathogens, respectively. Complex 2 has a biofilm inhibitory activity of only two to four times than its MIC. We also describe for the first time a potent antibacterial synergistic effect of a gold(III) complex combined with colistin, showing a bactericidal effect in less than 2 h; confirming the role of the outer membrane as a permeability barrier. Complex 2 shows a low rate of internalization in Staphylococcus aureus and Acinetobacter baumannii; it does not interact with replication enzymes or efflux pumps, causes ultrastructural damages in both membrane and cytoplasmic levels, and permeabilizes the bacterial membrane. Unlike control antibiotics, complex 2 did not generate resistant mutants in 30-day sequential cultures. We detected lower cytotoxicity in a non-tumoral THLE-2 cell line (IC50 = 25.5 μM) and no acute toxicity signs in vivo after an i.v. 1-mg/kg dose. The characterization presented here reassures the potential of complex 2 as a new chemical class of antimicrobial agents.
Collapse
Affiliation(s)
- Carlos Ratia
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Virginio Cepas
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Raquel Soengas
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, Almería, Spain
| | - Yolanda Navarro
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, Almería, Spain
| | - María Velasco-de Andrés
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - María José Iglesias
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, Almería, Spain
| | - Francisco Lozano
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Servei d’Immunologia, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain
- Departament de Biomedicina, Universitat de Barcelona, Barcelona, Spain
| | - Fernando López-Ortiz
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, Almería, Spain
| | - Sara M. Soto
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
13
|
Gold(III) heteroleptic complexes with SNS-thiosemicarbazonate ligands as cytotoxic agents: experimental and computational insights into the mechanism of action. Polyhedron 2022. [DOI: 10.1016/j.poly.2022.115767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
14
|
Vedenyapina MD, Kulaishin SA, Kuznetsov VV, Makhova NN, Kazakova MM. Kinetics and mechanism of gold anode corrosion in a weakly basic aqueous solution of hexamethylenetetramine (urotropine). Russ Chem Bull 2022. [DOI: 10.1007/s11172-022-3375-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
15
|
Kyhoiesh HAK, Al-Adilee KJ. Synthesis, spectral characterization and biological activities of Ag(I), Pt(IV) and Au(III) complexes with novel azo dye ligand (N, N, O) derived from 2-amino-6-methoxy benzothiazole. CHEMICAL PAPERS 2022. [DOI: 10.1007/s11696-022-02072-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
16
|
Dandash F, Leger DY, Diab-Assaf M, Sol V, Liagre B. Porphyrin/Chlorin Derivatives as Promising Molecules for Therapy of Colorectal Cancer. Molecules 2021; 26:7268. [PMID: 34885849 PMCID: PMC8659284 DOI: 10.3390/molecules26237268] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 12/27/2022] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related death. The demand for new therapeutic approaches has increased attention paid toward therapies with high targeting efficiency, improved selectivity and few side effects. Porphyrins are powerful molecules with exceptional properties and multifunctional uses, and their special affinity to cancer cells makes them the ligands par excellence for anticancer drugs. Porphyrin derivatives are used as the most important photosensitizers (PSs) for photodynamic therapy (PDT), which is a promising approach for anticancer treatment. Nevertheless, the lack of solubility and selectivity of the large majority of these macrocycles led to the development of different photosensitizer complexes. In addition, targeting agents or nanoparticles were used to increase the efficiency of these macrocycles for PDT applications. On the other hand, gold tetrapyrrolic macrocycles alone showed very interesting chemotherapeutic activity without PDT. In this review, we discuss the most important porphyrin derivatives, alone or associated with other drugs, which have been found effective against CRC, as we describe their modifications and developments through substitutions and delivery systems.
Collapse
Affiliation(s)
- Fatima Dandash
- Doctoral School of Sciences and Technology, Lebanese University, Hadath, Beirut 21219, Lebanon; (F.D.); (M.D.-A.)
| | - David Y. Leger
- Laboratoire PEIRENE EA 7500, Faculté de Pharmacie et Faculté des Sciences et Techniques, Université de Limoges, 2 Rue du Dr Marcland, CEDEX, 87025 Limoges, France; (D.Y.L.); (V.S.)
| | - Mona Diab-Assaf
- Doctoral School of Sciences and Technology, Lebanese University, Hadath, Beirut 21219, Lebanon; (F.D.); (M.D.-A.)
| | - Vincent Sol
- Laboratoire PEIRENE EA 7500, Faculté de Pharmacie et Faculté des Sciences et Techniques, Université de Limoges, 2 Rue du Dr Marcland, CEDEX, 87025 Limoges, France; (D.Y.L.); (V.S.)
| | - Bertrand Liagre
- Laboratoire PEIRENE EA 7500, Faculté de Pharmacie et Faculté des Sciences et Techniques, Université de Limoges, 2 Rue du Dr Marcland, CEDEX, 87025 Limoges, France; (D.Y.L.); (V.S.)
| |
Collapse
|
17
|
Pradhan AK, Shyam A, Mondal P. Quantum Chemical Investigations on the Hydrolysis of Gold(III)-Based Anticancer Drugs and Their Interaction with Amino Acid Residues. ACS OMEGA 2021; 6:28084-28097. [PMID: 34723008 PMCID: PMC8552358 DOI: 10.1021/acsomega.1c04168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/06/2021] [Indexed: 06/13/2023]
Abstract
A comprehensive hydrolysis mechanism of the promising class of Au(III) anticancer drugs [Au(DMDT)Cl2] (DMDT = N,N-dimethyldithiocarbamate) (R) and [Au(damp)Cl2] (damp = 2-[(dimethylamino)methyl]phenyl) (R') was done by means of density functional theory (DFT) in combination with the CPCM solvation model to explore the solution behavior and stability under physiological conditions. The activation free energies (ΔG) for the second hydrolysis, R (13.7 kcal/mol) and R' (10.0 kcal/mol) are found to be relatively lower in comparison to the first hydrolysis, and their rate constant values are computed to be 5.62 × 102 and 2.90 × 105 s-1, respectively. Besides these, the interaction mechanisms of aquated R and R' with the potential protein-binding sites cysteine (Cys) and selenocysteine (Sec) were also investigated in detail. The kinetic study and activation Gibbs free energy profiles reveal that the aquated complexes of R and R' bind more effectively to the Se site of Sec than to the S site of Cys. Intra- and intermolecular hydrogen bonding play a pivotal role in stabilizing the intermediates and transition states involved in the ligand substitution reactions of R and R'. Natural population analysis (NPA) was done to determine the charge distributions on important atoms during the hydrolysis and ligand substitution reactions.
Collapse
|
18
|
Mazzei L, Massai L, Cianci M, Messori L, Ciurli S. Medicinal Au(I) compounds targeting urease as prospective antimicrobial agents: unveiling the structural basis for enzyme inhibition. Dalton Trans 2021; 50:14444-14452. [PMID: 34585201 DOI: 10.1039/d1dt02488d] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A few gold compounds were recently found to show antimicrobial properties in vitro, holding great promise for the discovery of new drugs to overcome antibiotic resistance. Here, the inhibition of the bacterial virulence factor urease by four Au(I)-compounds, namely Au(PEt3)Cl, Au(PEt3)Br, Au(PEt3)I and [Au(PEt3)2]Cl, obtained from the antiarthritic Au(I)-drug Auranofin and earlier reported to act as antimicrobials, is investigated. The three monophosphino Au(I) complexes showed IC50 values in the 30-100 nM range, while the diphosphino Au(I) complex, though being less active, still showed a IC50 value of 7 μM. The structural basis for this inhibition was provided by solving the crystal structures of urease co-crystallized with Au(PEt3)I and [Au(PEt3)2]Cl: at least two Au(I) ions bind the enzyme in a flap domain involved in the catalysis, thus obliterating enzyme activity. Peculiar changes observed in the two structures reveal implications for the mechanism of soft metal binding and enzyme inactivation.
Collapse
Affiliation(s)
- Luca Mazzei
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Giuseppe Fanin 40, I-40127 Bologna, Italy.
| | - Lara Massai
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, I-50019 Sesto Fiorentino, Italy
| | - Michele Cianci
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, I-60131 Ancona, Italy
| | - Luigi Messori
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, I-50019 Sesto Fiorentino, Italy
| | - Stefano Ciurli
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Giuseppe Fanin 40, I-40127 Bologna, Italy.
| |
Collapse
|
19
|
Ostrovskaya LA, Korman DB, Nekrasova EI, Bluhterova NV, Fomina MM, Rikova VA, Hochenkova UA, Abzaeva KA. The Antitumor and Cytotoxic Effects of Polyacrylates of Noble Metals. Biophysics (Nagoya-shi) 2021. [DOI: 10.1134/s000635092105016x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
20
|
Fontinha D, Sousa SA, Morais TS, Prudêncio M, Leitão JH, Le Gal Y, Lorcy D, Silva RAL, Velho MFG, Belo D, Almeida M, Guerreiro JF, Pinheiro T, Marques F. Gold(iii) bis(dithiolene) complexes: from molecular conductors to prospective anticancer, antimicrobial and antiplasmodial agents. Metallomics 2021; 12:974-987. [PMID: 32391537 DOI: 10.1039/d0mt00064g] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The anticancer, antimicrobial and antiplasmodial activities of six gold(iii) bis(dithiolene) complexes were studied. Complexes 1-6 showed relevant anticancer properties against A2780/A2780cisR ovarian cancer cells (IC50 values of 0.08-2 μM), also being able to overcome cisplatin resistance in A2780cisR cells. Complex 1 also exhibited significant antimicrobial activity against Staphylococcus aureus (minimum inhibitory concentration (MIC) values of 12.1 ± 3.9 μg mL-1) and both Candida glabrata and Candida albicans (MICs of 9.7 ± 2.7 and 19.9 ± 2.4 μg mL-1, respectively). In addition, all complexes displayed antiplasmodial activity against the Plasmodium berghei parasite liver stages, even exhibiting better results than the ones obtained using primaquine, an anti-malarial drug. Mechanistic studies support the idea that thioredoxin reductase, but not DNA, is a possible target of these complexes. Complex 1 is stable under biological conditions, which would be important if this compound is ever to be considered as a drug. Overall, the results obtained evidenced the promising biological activity of complex 1, which might have potential as a novel anticancer, antimicrobial and antiplasmodial agent to be used as an alternative to current therapeutics.
Collapse
Affiliation(s)
- Diana Fontinha
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Sílvia A Sousa
- iBB-Institute for Bioengineering and Biosciences, Departmento de Bioengenharia, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Tânia S Morais
- Centro de Química Estrutural, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Miguel Prudêncio
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Jorge H Leitão
- iBB-Institute for Bioengineering and Biosciences, Departmento de Bioengenharia, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Yann Le Gal
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000 Rennes, France
| | - Dominique Lorcy
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000 Rennes, France
| | - Rafaela A L Silva
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal.
| | - Mariana F G Velho
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal. and Instituto de Telecomunicações, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001, Lisboa, Portugal
| | - Dulce Belo
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal.
| | - M Almeida
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal.
| | - Joana F Guerreiro
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal.
| | - Teresa Pinheiro
- iBB-Institute for Bioengineering and Biosciences, Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal
| | - Fernanda Marques
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal.
| |
Collapse
|
21
|
Abbasi M, Yaqoob M, Haque RA, Iqbal MA. Potential of Gold Candidates against Human Colon Cancer. Mini Rev Med Chem 2021; 21:69-78. [PMID: 32767935 DOI: 10.2174/1389557520666200807130721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 11/22/2022]
Abstract
Development of novel metallodrugs with pharmacological profile plays a significant role in modern medicinal chemistry and drug design. Metal complexes have shown remarkable clinical results in current cancer therapy. Gold complexes have attained attention due to their high antiproliferative potential. Gold-based drugs are used for the treatment of rheumatoid arthritis. Gold-containing compounds with selective and specific targets are capable to assuage the symptoms of a range of human diseases. Gold (I) species with labile ligands (such as Cl in TEPAuCl) interact with isolated DNA; therefore, this biomolecule has been considered as a target for gold drugs. Gold (I) has a high affinity towards sulfur and selenium. Due to this, gold (I) drugs readily interact with cysteine or selenocysteine residue of the enzyme to form protein-gold(I) thiolate or protein-gold (I) selenolate complexes that lead to inhibition of the enzyme activity. Au(III) compounds due to their square-planner geometriesthe same as found in cisplatin, represent a good source for the development of anti-tumor agents. This article aims to review the most important applications of gold products in the treatment of human colon cancer and to analyze the complex interplay between gold and the human body.
Collapse
Affiliation(s)
- Mahvish Abbasi
- Department of Chemistry, University of Agriculture Faisalabad-38040, Pakistan
| | - Munazzah Yaqoob
- Department of Chemistry, University of Agriculture Faisalabad-38040, Pakistan
| | - Rosenani A Haque
- School of Chemical Sciences, Universiti Sains Malaysia, 11800-USM, Penang, Malaysia
| | | |
Collapse
|
22
|
Possato B, Dalmolin LF, Pereira LM, Alves JQ, Silva RTC, Gelamo RV, Yatsuda AP, Lopez RFV, de Albuquerque S, Leite NB, Maia PIDS. Gold(III) complexes with thiosemicarbazonate ligands as potential anticancer agents: Cytotoxicity and interactions with biomolecular targets. Eur J Pharm Sci 2021; 162:105834. [PMID: 33826936 DOI: 10.1016/j.ejps.2021.105834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/17/2021] [Accepted: 03/31/2021] [Indexed: 01/23/2023]
Abstract
Gold(III) complexes have been studied for the past years due to their anticancer properties and great affinity to biotargets, such as enzymes and proteins, which support their pharmacological applications. Within this scope, in this work the antiproliferative activities of two Au(III)-thiosemicarbazonate complexes, [AuClL1] (1, L1: (E,Z)-N-ethyl-N'-(3-nitroso-kN)butan-2-ylidene)carbamohydrazonothioato-k2N2,S) and [Au(Hdamp)L2]Cl (2, L2: N-(N'',N''-diethylaminothiocarbonyl)-N'(N''', N'''-dimethylcarbothioamide)benzamidineto-kN,k2S and Hdamp: 2-(N,N-dimethylaminomethyl)-phenyl-C1), and their affinities to possible biological targets were investigated. Three different tumor cell lines were used to perform the cytotoxicity assays, including one cisplatin-resistant model, and the results showed lower EC50 for 1 over 2 in every case: B16F10 (4.1 μM and 15.6 μM), A431 (4.0 μM and >50 μM) and OVCAR3 (4.2 μM and 24.5 μM). However, a lower toxicity to fibroblast 3T3 cell line was observed for 2 (30.58 μM) when compared to 1 (7.17 μM), resulting in comparable therapeutic indexes. Both complexes presented strong affinity to HSA: they distorted the secondary structure of the protein, as verified by circular dichroism, but 1 additionally presented the apparent fluorescence quenching constant (Kapp) ten times greater than 2, which was probably due to the fact of 1 being able to denature HSA. The ethidium bromide displacement assay showed that neither 1 nor 2 are strong DNA intercalators, which is in agreement with what was observed through the UV-vis titration. In both cases, the 260 nm band presented hyperchromism, which can indicate ionic interactions or DNA damage. In fact, 1 was able to damage the pGEM plasmid, similarly to cisplatin, as verified by agarose gel electrophoresis and Atomic Force Microscopy. Biophysical studies in cancer cells model membranes were also performed in order to investigate the interaction of the gold complexes to lipid bilayers and revealed that the compounds interact with the membranes by exhibiting partition coefficients of 103 order of magnitude. Overall, both complexes were found to be promising candidates for the development of a future anticancer drug against low sensitive or cisplatin resistant tumors.
Collapse
Affiliation(s)
- Bruna Possato
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Luciana Falcco Dalmolin
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Luiz Miguel Pereira
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | | | | | - Rogerio Valentim Gelamo
- Instituto de Ciências Tecnológicas e Exatas, Universidade Federal do Triângulo Mineiro, Uberaba, Brazil
| | - Ana Patrícia Yatsuda
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | | | - Sérgio de Albuquerque
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Natália Bueno Leite
- Núcleo de Desenvolvimento de Compostos Bioativos (NDCBio), Universidade Federal do Triângulo Mineiro, Uberaba, Brazil.
| | - Pedro Ivo da Silva Maia
- Núcleo de Desenvolvimento de Compostos Bioativos (NDCBio), Universidade Federal do Triângulo Mineiro, Uberaba, Brazil.
| |
Collapse
|
23
|
Silva MJSA, Gois PMP, Gasser G. Unveiling the Potential of Transition Metal Complexes for Medicine: Translational in Situ Activation of Metal-Based Drugs from Bench to in Vivo Applications. Chembiochem 2021; 22:1740-1742. [PMID: 33507625 DOI: 10.1002/cbic.202100015] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/28/2021] [Indexed: 02/06/2023]
Abstract
The development of metal-based anticancer drugs has been hampered, among other reasons, by their lack of selectivity for cancer cells. In a recent article, Zou and co-workers presented the successful intracellular activation of organogold(I) complexes for potential cancer treatment through Pd(II)-mediated transmetallation, overcoming some off-target activity of novel gold-based drugs. This unique strategy builds the perfect bridge between metallodrug usage and bioorthogonal intracellular catalysis for more advanced and selective therapies. Such an approach will hopefully pave the way for forthcoming studies in medicinal inorganic chemistry.
Collapse
Affiliation(s)
- Maria J S A Silva
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences Laboratory for Inorganic Chemical Biology, FR-75005, Paris, France.,Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Pedro M P Gois
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences Laboratory for Inorganic Chemical Biology, FR-75005, Paris, France
| |
Collapse
|
24
|
Galassi R, Luciani L, Gambini V, Vincenzetti S, Lupidi G, Amici A, Marchini C, Wang J, Pucciarelli S. Multi-Targeted Anticancer Activity of Imidazolate Phosphane Gold(I) Compounds by Inhibition of DHFR and TrxR in Breast Cancer Cells. Front Chem 2021; 8:602845. [PMID: 33490036 PMCID: PMC7821381 DOI: 10.3389/fchem.2020.602845] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/09/2020] [Indexed: 12/09/2022] Open
Abstract
A class of phosphane gold(I) compounds, made of azoles and phosphane ligands, was evaluated for a screening on the regards of Breast Cancer cell panels (BC). The compounds possess N-Au-P or Cl-Au-P bonds around the central metal, and they differ for the presence of aprotic or protic polar groups in the azoles and/or the phosphane moieties to tune their hydrophilicity. Among the six candidates, only the compounds having the P-Au-N environment and not displaying neither the hydroxyl nor carboxyl groups in the ligands were found active. The compounds were screened by MTT tests in SKBR3, A17, and MDA-MB231 cancer cells, and two compounds (namely the 4,5-dicyano-imidazolate-1yl-gold(I)-(triphenylphosphane, 5, and 4,5-dichloro-imidazolate-1yl-gold(I)-triphenylphosphane, 6) were found very cytotoxic, with the most active with an IC50 value of 3.46 μM in MDA-MB231 cells. By performing enzymatic assays in the treated cells lysates, the residual enzymatic activity of dihydrofolate reductase (DHFR) has been measured after cell treatment for 4 or 12 h in comparison with control cells. Upon 12 h of treatment, the activity of DHFR was significantly reduced in both SKBR3 and A17 cells by compounds 5 and 6, but not in human MDA-MB231 cells; interestingly, it was found remarkably high after 4 h of treatment, revealing a time dependence for the DHFR enzymatic assays. The DHFR inhibition data have been compared to those for the thioredoxin reductase (TrxR), the most recognized molecular target for gold compounds. For this latter, similar residual activities (i.e., 37 and 49% for the match of SKBR3 cells and compound 5 or 6, respectively) were found. Binding studies on the regards of ct-DNA (calf-thymus-DNA) and of plasma transporters proteins, such as BSA (bovine serum albumin) and ATF (apo transferrin), were performed. As expected for gold compounds, the data support strong binding to proteins (Ksv values range: 1.51 ÷ 2.46 × 104 M−1) and a weaker interaction with ct-DNA's minor groove (Ksv values range: 1.55 ÷ 6.12 × 103 M−1).
Collapse
Affiliation(s)
- Rossana Galassi
- School of Science and Technology, University of Camerino, Camerino, Italy
| | - Lorenzo Luciani
- School of Science and Technology, University of Camerino, Camerino, Italy
| | - Valentina Gambini
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Silvia Vincenzetti
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Giulio Lupidi
- School of Drugs and Health Products Sciences, University of Camerino, Camerino, Italy
| | - Augusto Amici
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Cristina Marchini
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Junbiao Wang
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Stefania Pucciarelli
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| |
Collapse
|
25
|
|
26
|
Espinosa AV, Costa DDS, Tunes LG, Monte‐Neto RLD, Grazul RM, Almeida MV, Silva H. Anticancer and antileishmanial in vitro activity of gold(I) complexes with 1,3,4‐oxadiazole‐2(
3H
)‐thione ligands derived from δ‐D‐gluconolactone. Chem Biol Drug Des 2020; 97:41-50. [DOI: 10.1111/cbdd.13757] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 06/15/2020] [Accepted: 06/28/2020] [Indexed: 12/18/2022]
Affiliation(s)
| | - Danilo de Souza Costa
- Departamento de Química ICEUniversidade Federal de Juiz de Fora Juiz de Fora MG Brazil
| | | | | | | | - Mauro Vieira Almeida
- Departamento de Química ICEUniversidade Federal de Juiz de Fora Juiz de Fora MG Brazil
| | - Heveline Silva
- Departamento de Química ICExUniversidade Federal de Minas Gerais Belo Horizonte MG Brazil
| |
Collapse
|
27
|
Khan E, Khan S, Gul Z, Muhammad M. Medicinal Importance, Coordination Chemistry with Selected Metals (Cu, Ag, Au) and Chemosensing of Thiourea Derivatives. A Review. Crit Rev Anal Chem 2020; 51:812-834. [DOI: 10.1080/10408347.2020.1777523] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Ezzat Khan
- Department of Chemistry, University of Malakand, Chakdara, Khyber Pakhtunkhwa, Pakistan
| | - Sikandar Khan
- Department of Chemistry, University of Malakand, Chakdara, Khyber Pakhtunkhwa, Pakistan
| | - Zarif Gul
- Department of Chemistry, University of Malakand, Chakdara, Khyber Pakhtunkhwa, Pakistan
| | - Mian Muhammad
- Department of Chemistry, University of Malakand, Chakdara, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
28
|
Tunes LG, Morato RE, Garcia A, Schmitz V, Steindel M, Corrêa-Junior JD, Dos Santos HF, Frézard F, de Almeida MV, Silva H, Moretti NS, de Barros ALB, do Monte-Neto RL. Preclinical Gold Complexes as Oral Drug Candidates to Treat Leishmaniasis Are Potent Trypanothione Reductase Inhibitors. ACS Infect Dis 2020; 6:1121-1139. [PMID: 32283915 DOI: 10.1021/acsinfecdis.9b00505] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The drugs currently used to treat leishmaniases have limitations concerning cost, efficacy, and safety, making the search for new therapeutic approaches urgent. We found that the gold(I)-derived complexes were active against L. infantum and L. braziliensis intracellular amastigotes with IC50 values ranging from 0.5 to 5.5 μM. All gold(I) complexes were potent inhibitors of trypanothione reductase (TR), with enzyme IC50 values ranging from 1 to 7.8 μM. Triethylphosphine-derived complexes enhanced reactive oxygen species (ROS) production and decreased mitochondrial respiration after 2 h of exposure, indicating that gold(I) complexes cause oxidative stress by direct ROS production, by causing mitochondrial damage or by impairing TR activity and thus accumulating ROS. There was no cross-resistance to antimony; in fact, SbR (antimony-resistant mutants) strains were hypersensitive to some of the complexes. BALB/c mice infected with luciferase-expressing L. braziliensis or L. amazonensis and treated orally with 12.5 mg/kg/day of AdT Et (3) or AdO Et (4) presented reduced lesion size and parasite burden, as revealed by bioimaging. The combination of (3) and miltefosine allowed for a 50% reduction in miltefosine treatment time. Complexes 3 and 4 presented favorable pharmacokinetic and toxicity profiles that encourage further drug development studies. Gold(I) complexes are promising antileishmanial agents, with a potential for therapeutic use, including in leishmaniasis caused by antimony-resistant parasites.
Collapse
Affiliation(s)
- Luiza G. Tunes
- Instituto René Rachou/Fiocruz Minas−Fundação Oswaldo Cruz, Belo Horizonte 30190-009, Brasil
| | - Roberta E. Morato
- Instituto René Rachou/Fiocruz Minas−Fundação Oswaldo Cruz, Belo Horizonte 30190-009, Brasil
| | - Adriana Garcia
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora 36036-900, Brasil
| | - Vinicius Schmitz
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora 36036-900, Brasil
| | - Mario Steindel
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina, Florianópolis 88040-900, Brasil
| | - José D. Corrêa-Junior
- Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brasil
| | - Hélio F. Dos Santos
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora 36036-900, Brasil
| | - Frédéric Frézard
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brasil
| | - Mauro V. de Almeida
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora 36036-900, Brasil
| | - Heveline Silva
- Departamento de Química, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brasil
| | - Nilmar S. Moretti
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo 04023-062, Brasil
| | - André L. B. de Barros
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brasil
| | | |
Collapse
|
29
|
Adokoh CK. Therapeutic potential of dithiocarbamate supported gold compounds. RSC Adv 2020; 10:2975-2988. [PMID: 35496096 PMCID: PMC9048446 DOI: 10.1039/c9ra09682e] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/08/2020] [Indexed: 12/24/2022] Open
Abstract
Chrysotherapy or aurotherapy, the use of gold as medicine, is two thousand years old. Hitherto, numerous diverse gold stabilizing ligands for instance vitamins, pyridine, phosphines, naphthylamine and xanthanes have been developed and their 'chelating effect' in addition to their anti-proliferative properties have been extensively studied. Recent advances in the field of bioinorganic chemistry have led to the design of biologically relevant metal complexes with appropriate fine-tuned ligands such as metallic conjugates of dithiocarbamates (DTCs). DTC compounds have been recognised to possess diverse applications and have demonstrated interesting biological properties. For instance, the chemoprotective and antitumour properties of gold metal ions and DTC compounds respectively, presents an innovative and effective approach to cancer management. This review presents therefore the therapeutic potential of DTC ligand systems as a support for gold compounds. The importance of dithiocarbamate supported gold compounds as potential therapeutic agents is highlighted with emphasis on the therapeutic potential of gold(iii) and gold(i) dithiocarbamate derivatives.
Collapse
Affiliation(s)
- Christian K Adokoh
- Department of Forensic Sciences, School of Biological Sciences, College of Agriculture and Natural Sciences, University of Cape Coast Cape Coast Ghana
| |
Collapse
|
30
|
Ghosh S, Khan MA, Bhattacharyya A, Alam MA, Zangrando E, Guchhait N. Cu(ii)-induced twisting of the biphenyl core: exploring the effect of structure and coordination environment of biphenyl-based chiral copper(ii) complexes on interaction with calf-thymus DNA. NEW J CHEM 2020. [DOI: 10.1039/c9nj06184c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Biphenyl core-based clip-like receptors get twisted after complexation with Cu2+. The extent of interaction of the optically active complexes with ct-DNA varies depending on the structure and coordination environment.
Collapse
Affiliation(s)
- Soumen Ghosh
- Department of Chemistry
- University of Calcutta
- Kolkata 700 009
- India
| | - Mehebub Ali Khan
- Department of Chemistry
- Aliah University
- IIA/27
- Action Area II
- Newtown
| | | | | | - Ennio Zangrando
- Department of Chemical and Pharmaceutical Sciences, University of Trieste
- 34127 Trieste
- Italy
| | - Nikhil Guchhait
- Department of Chemistry
- University of Calcutta
- Kolkata 700 009
- India
| |
Collapse
|
31
|
Group 10 metal complexes with a tetradentate thiosemicarbazonate ligand: Synthesis, crystal structures and computational insights into the catalysis for C–C coupling via Mizoroki-Heck reaction. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2019.126997] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
32
|
Ceramella J, Mariconda A, Iacopetta D, Saturnino C, Barbarossa A, Caruso A, Rosano C, Sinicropi MS, Longo P. From coins to cancer therapy: Gold, silver and copper complexes targeting human topoisomerases. Bioorg Med Chem Lett 2019; 30:126905. [PMID: 31874823 DOI: 10.1016/j.bmcl.2019.126905] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/07/2019] [Accepted: 12/09/2019] [Indexed: 02/07/2023]
Abstract
Cancer is a complex issue and, even though the prevention basics and therapy have been implemented, it is still the second leading death cause worldwide. With the hope to discover new powerful and safer molecules to fight cancer, many researchers focused their attention on metal-based compounds, starting from the most famous and successfully employed anticancer drug, i.e. cisplatin. The current article aims to report the most recent discoveries about the use of gold, silver and copper complexes as antitumor agents, highlighting their influences on important enzymes, namely human topoisomerases. The latter are fundamental for the cell life and, if overexpressed, strongly implicated in cancer onset and progression. The identification of lead complexes targeting human topoisomerases and gifted with the appropriate chemical and pharmacological properties represents a fecund starting point to obtain new and more effective anticancer molecules.
Collapse
Affiliation(s)
- Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | | | - Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy.
| | - Carmela Saturnino
- Department of Science, University of Basilicata, 85100 Potenza, Italy
| | - Alexia Barbarossa
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Anna Caruso
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Camillo Rosano
- Biopolymers and Proteomics IRCCS, Ospedale Policlinico San Martino - IST, 16132 Genova, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Pasquale Longo
- Department of Biology and Chemistry, University of Salerno, 84084 Fisciano, Italy
| |
Collapse
|
33
|
Abstract
Abstract
The earliest therapy use of gold compounds dated in 2500 AD. In 1960s gold compounds were used in the treatment of rheumatoid arthritis, discoid lupus erythematosus, etc. Although the compounds of Au(I) are well-known and used in the modern medicine, new complexes of Au(III) and ligands with different donor atoms are synthetized and examined for their biological (antitumor, antimicrobial) activity. So far conducted studies show that complexes with Au(III) as central metal ions have greater activity compared with free ligands. Future syntheses should be based on the increase of biological activity with complexes that have necessary stability under physiological conditions.
Collapse
|
34
|
Lopes CD, Possato B, Gaspari APS, Oliveira RJ, Abram U, Almeida JPA, Rocho FDR, Leitão A, Montanari CA, Maia PIS, da Silva JS, de Albuquerque S, Carneiro ZA. Organometallic Gold(III) Complex [Au(Hdamp)(L1 4)] + (L1 = SNS-Donating Thiosemicarbazone) as a Candidate to New Formulations against Chagas Disease. ACS Infect Dis 2019; 5:1698-1707. [PMID: 31419384 DOI: 10.1021/acsinfecdis.8b00284] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chagas disease remains a serious public health concern with unsatisfactory treatment outcomes due to strain-specific drug resistance and various side effects. To identify new therapeutic drugs against Trypanosoma cruzi, we evaluated both the in vitro and in vivo activity of the organometallic gold(III) complex [Au(III)(Hdamp)(L14)]Cl (L1 = SNS-donating thiosemicarbazone), henceforth denoted 4-Cl. Our results demonstrated that 4-Cl was more effective than benznidazole (Bz) in eliminating both the extracellular trypomastigote and intracellular amastigote forms of the parasite without cytotoxic effects on mammalian cells. In in vivo assays, 4-Cl in PBS solution loses the protonation and becomes the 4-neutral. 4-Neutral reduced parasitaemia and tissue parasitism in addition to protecting the liver and heart from tissue damage at 2.8 mg/kg/day. All these changes resulted in the survival of 100% of the mice treated with the gold complex during the acute phase. Analyzing the surviving animals of the acute infection, the parasite load after 150 days of infection was equivalent to those treated with the standard dose of Bz without demonstrating the hepatotoxicity of the latter. In addition, we identified a modulation of interferon gamma (IFN-γ) levels that may be targeting the disease's positive outcome. To the best of our knowledge, this is the first gold organometallic study that shows promise in an in vivo experimental model against Chagas disease.
Collapse
Affiliation(s)
- Carla Duque Lopes
- Department of Clinical Toxicological and Bromatological Analysis School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo 14040-903, Brazil
- Departament of Biochemistry and Immunology, School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Bruna Possato
- Department of Clinical Toxicological and Bromatological Analysis School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo 14040-903, Brazil
| | - Ana Paula S. Gaspari
- Department of Clinical Toxicological and Bromatological Analysis School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo 14040-903, Brazil
| | - Ronaldo J. Oliveira
- Núcleo de Desenvolvimento de Compostos Bioativos (NDCBio), Universidade Federal do Triângulo Mineiro, Uberaba, Minas Gerais 38025-470, Brazil
| | - Ulrich Abram
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin D-14195, Germany
| | - José P. A. Almeida
- Department of Clinical Toxicological and Bromatological Analysis School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo 14040-903, Brazil
| | - Fernanda dos Reis Rocho
- Grupo de Estudos em Química Medicinal de Produtos Naturais−NEQUIMED-PN, Instituto de Química de São Carlos, Universidade de São Paulo, Av. Trabalhador Sancarlense 400, P.O. Box 780, São Carlos, São Paulo 13560-960, Brazil
| | - Andrei Leitão
- Grupo de Estudos em Química Medicinal de Produtos Naturais−NEQUIMED-PN, Instituto de Química de São Carlos, Universidade de São Paulo, Av. Trabalhador Sancarlense 400, P.O. Box 780, São Carlos, São Paulo 13560-960, Brazil
| | - Carlos A. Montanari
- Grupo de Estudos em Química Medicinal de Produtos Naturais−NEQUIMED-PN, Instituto de Química de São Carlos, Universidade de São Paulo, Av. Trabalhador Sancarlense 400, P.O. Box 780, São Carlos, São Paulo 13560-960, Brazil
| | - Pedro I. S. Maia
- Núcleo de Desenvolvimento de Compostos Bioativos (NDCBio), Universidade Federal do Triângulo Mineiro, Uberaba, Minas Gerais 38025-470, Brazil
| | - João S. da Silva
- Departament of Biochemistry and Immunology, School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Sérgio de Albuquerque
- Department of Clinical Toxicological and Bromatological Analysis School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo 14040-903, Brazil
| | - Zumira A. Carneiro
- Department of Clinical Toxicological and Bromatological Analysis School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo 14040-903, Brazil
| |
Collapse
|
35
|
Salsi F, Bulhões Portapilla G, Schutjajew K, Roca Jungfer M, Goulart A, Hagenbach A, de Albuquerque S, Abram U. Organometallic Gold(III) Complexes with Tridentate Halogen‐Substituted Thiosemicarbazones: Effects of Halogenation on Cytotoxicity and Anti‐Parasitic Activity. Eur J Inorg Chem 2019. [DOI: 10.1002/ejic.201900904] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Federico Salsi
- Institute of Chemistry and Biochemistry Freie Universität Berlin Fabeckstr. 34–36 D‐14195 Berlin Germany
| | - Gisele Bulhões Portapilla
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto Universidade de São Paulo Av. do Café – Vila Monte Alegre 14040‐903 Ribeirão Preto Brazil
| | - Konstantin Schutjajew
- Institute of Chemistry and Biochemistry Freie Universität Berlin Fabeckstr. 34–36 D‐14195 Berlin Germany
| | - Maximilian Roca Jungfer
- Institute of Chemistry and Biochemistry Freie Universität Berlin Fabeckstr. 34–36 D‐14195 Berlin Germany
| | - Amanda Goulart
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto Universidade de São Paulo Av. do Café – Vila Monte Alegre 14040‐903 Ribeirão Preto Brazil
| | - Adelheid Hagenbach
- Institute of Chemistry and Biochemistry Freie Universität Berlin Fabeckstr. 34–36 D‐14195 Berlin Germany
| | - Sérgio de Albuquerque
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto Universidade de São Paulo Av. do Café – Vila Monte Alegre 14040‐903 Ribeirão Preto Brazil
| | - Ulrich Abram
- Institute of Chemistry and Biochemistry Freie Universität Berlin Fabeckstr. 34–36 D‐14195 Berlin Germany
| |
Collapse
|
36
|
|
37
|
Salsi F, Bulhões Portapilla G, Simon S, Roca Jungfer M, Hagenbach A, de Albuquerque S, Abram U. Effect of Fluorination on the Structure and Anti-Trypanosoma cruzy Activity of Oxorhenium(V) Complexes with S,N,S-Tridentate Thiosemicarbazones and Benzoylthioureas. Synthesis and Structures of Technetium(V) Analogues. Inorg Chem 2019; 58:10129-10138. [DOI: 10.1021/acs.inorgchem.9b01260] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Federico Salsi
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstrasse 34-36, D-14195 Berlin, Germany
| | - Gisele Bulhões Portapilla
- Faculdade de Ciencias Farmaceuticas de Ribeirão Preto, Universidade de São Paulo, Av. do Cafe - Vila Monte Alegre, Ribeirão Preto, São Paulo 14040-903, Brazil
| | - Saskia Simon
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstrasse 34-36, D-14195 Berlin, Germany
| | - Maximilian Roca Jungfer
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstrasse 34-36, D-14195 Berlin, Germany
| | - Adelheid Hagenbach
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstrasse 34-36, D-14195 Berlin, Germany
| | - Sérgio de Albuquerque
- Faculdade de Ciencias Farmaceuticas de Ribeirão Preto, Universidade de São Paulo, Av. do Cafe - Vila Monte Alegre, Ribeirão Preto, São Paulo 14040-903, Brazil
| | - Ulrich Abram
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstrasse 34-36, D-14195 Berlin, Germany
| |
Collapse
|
38
|
Cu(I) complexes with thiosemicarbazides derived from p-toluenesulfohydrazide: Structural, luminescence and biological studies. Polyhedron 2018. [DOI: 10.1016/j.poly.2018.08.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
39
|
Aliaga-Lavrijsen M, Herrera RP, Villacampa MD, Gimeno MC. Efficient Gold(I) Acyclic Diaminocarbenes for the Synthesis of Propargylamines and Indolizines. ACS OMEGA 2018; 3:9805-9813. [PMID: 31459109 PMCID: PMC6645035 DOI: 10.1021/acsomega.8b01352] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/09/2018] [Indexed: 06/10/2023]
Abstract
Mononuclear gold(I) acyclic diaminocarbenes (ADCs) were prepared by the reaction of 1,2-cyclohexanediamine with the corresponding isocyanide complexes [AuCl(CNR)] (R = Cy, t Bu). The three-component coupling of aldehydes, amines, and alkynes was investigated by using these gold(I) ADC complexes. The new gold(I) metal complexes are highly efficient catalysts for the synthesis of propargylamines and indolizines in the absence of solvent and in mild conditions. This method affords the corresponding final products with excellent yields in short reaction times. Additionally, chiral gold(I) complexes with ADCs have been prepared and tried in the enantioselective synthesis of propargylamines.
Collapse
Affiliation(s)
- Mélanie Aliaga-Lavrijsen
- Departamento
de Química Inorgánica, Instituto de Síntesis
Química y Catálisis Homogénea (ISQCH), and Departamento
de Química Orgánica, Laboratorio de Organocatálisis
Asimétrica, Instituto de Síntesis Química y Catálisis
Homogénea (ISQCH), CSIC-Universidad
de Zaragoza, C/ Pedro Cerbuna, No. 12, E-50009 Zaragoza, Spain
| | - Raquel P. Herrera
- Departamento
de Química Inorgánica, Instituto de Síntesis
Química y Catálisis Homogénea (ISQCH), and Departamento
de Química Orgánica, Laboratorio de Organocatálisis
Asimétrica, Instituto de Síntesis Química y Catálisis
Homogénea (ISQCH), CSIC-Universidad
de Zaragoza, C/ Pedro Cerbuna, No. 12, E-50009 Zaragoza, Spain
| | - M. Dolores Villacampa
- Departamento
de Química Inorgánica, Instituto de Síntesis
Química y Catálisis Homogénea (ISQCH), and Departamento
de Química Orgánica, Laboratorio de Organocatálisis
Asimétrica, Instituto de Síntesis Química y Catálisis
Homogénea (ISQCH), CSIC-Universidad
de Zaragoza, C/ Pedro Cerbuna, No. 12, E-50009 Zaragoza, Spain
| | - M. Concepción Gimeno
- Departamento
de Química Inorgánica, Instituto de Síntesis
Química y Catálisis Homogénea (ISQCH), and Departamento
de Química Orgánica, Laboratorio de Organocatálisis
Asimétrica, Instituto de Síntesis Química y Catálisis
Homogénea (ISQCH), CSIC-Universidad
de Zaragoza, C/ Pedro Cerbuna, No. 12, E-50009 Zaragoza, Spain
| |
Collapse
|
40
|
Maia PIDS, Carneiro ZA, Lopes CD, Oliveira CG, Silva JS, de Albuquerque S, Hagenbach A, Gust R, Deflon VM, Abram U. Organometallic gold(iii) complexes with hybrid SNS-donating thiosemicarbazone ligands: cytotoxicity and anti-Trypanosoma cruzi activity. Dalton Trans 2018; 46:2559-2571. [PMID: 28154849 DOI: 10.1039/c6dt04307k] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Stable organogold(iii) compounds of the composition [AuIII(Hdamp)(L1)]Cl are formed from reactions of [AuCl2(damp)] with H2L1 (damp- = dimethylaminomethylphenyl; H2L1 = N'-(diethylcarbamothioyl)benzimidothiosemicarbazides). The cationic complexes can be neutralized by reactions with weak bases under the formation of [AuIII(damp)(L1)] compounds. The structures of the products show interesting features like relatively short AuH contacts between the methylene protons of the Hdamp ligand and the gold(iii) ions. Preliminary biological studies on the uncoordinated compounds H2L1 and their gold complexes indicate considerable cytotoxicity for the [AuIII(Hdamp)(L1)]Cl complexes against MCF-7 cells. The in vitro trypanocidal activity was evaluated against the intracellular form of Trypanosoma cruzi. The organometallic complexes display a remarkable activity, which is dependent on the alkyl substituents of the thiosemicarbazone building blocks of the ligands. One representative of the cationic [AuIII(Hdamp)(L1)]Cl complexes, where H2L1 contains a dimethylthiosemicarbazide building block, shows a trypanocidal activity against the intracellular amastigote form in the same order of magnitude as that of the standard drug benznidazole. Furthermore, no appreciable toxicity to mice spleen cells is observed for this compound resulting in a therapeutic index of about 30, which strongly recommends it as a promising candidate for the development of a future antiparasitic drug.
Collapse
Affiliation(s)
- P I da S Maia
- Departamento de Química, Universidade Federal do Triângulo Mineiro, 38025-440, Uberaba, MG, Brazil.
| | - Z A Carneiro
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP-USP, Universidade de São Paulo, Avenida do Café s/n, 14040-903, Ribeirão Preto, SP, Brazil and Department of Biochemistry and Immunology, School of Medicine, University of São Paulo, Av. Bandeirantes 3900, 14040-900, Ribeirão Preto, SP, Brazil
| | - C D Lopes
- Department of Biochemistry and Immunology, School of Medicine, University of São Paulo, Av. Bandeirantes 3900, 14040-900, Ribeirão Preto, SP, Brazil
| | - C G Oliveira
- Instituto de Química de São Carlos, Universidade de São Paulo, Av. Trabalhador São Carlense, 400, 13566-590, São Carlos, SP, Brazil
| | - J S Silva
- Department of Biochemistry and Immunology, School of Medicine, University of São Paulo, Av. Bandeirantes 3900, 14040-900, Ribeirão Preto, SP, Brazil
| | - S de Albuquerque
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP-USP, Universidade de São Paulo, Avenida do Café s/n, 14040-903, Ribeirão Preto, SP, Brazil
| | - A Hagenbach
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Fabeckstr. 34-36, D-14195 Berlin, Germany.
| | - R Gust
- Freie Universität Berlin, Institute of Pharmacy, Königin-Luise-Str. 2+4, 14195 Berlin, Germany
| | - V M Deflon
- Instituto de Química de São Carlos, Universidade de São Paulo, Av. Trabalhador São Carlense, 400, 13566-590, São Carlos, SP, Brazil
| | - U Abram
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Fabeckstr. 34-36, D-14195 Berlin, Germany.
| |
Collapse
|
41
|
Yeo CI, Ooi KK, Tiekink ERT. Gold-Based Medicine: A Paradigm Shift in Anti-Cancer Therapy? Molecules 2018; 23:molecules23061410. [PMID: 29891764 PMCID: PMC6100309 DOI: 10.3390/molecules23061410] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/23/2018] [Accepted: 05/28/2018] [Indexed: 11/16/2022] Open
Abstract
A new era of metal-based drugs started in the 1960s, heralded by the discovery of potent platinum-based complexes, commencing with cisplatin [(H₃N)₂PtCl₂], which are effective anti-cancer chemotherapeutic drugs. While clinical applications of gold-based drugs largely relate to the treatment of rheumatoid arthritis, attention has turned to the investigation of the efficacy of gold(I) and gold(III) compounds for anti-cancer applications. This review article provides an account of the latest research conducted during the last decade or so on the development of gold compounds and their potential activities against several cancers as well as a summary of possible mechanisms of action/biological targets. The promising activities and increasing knowledge of gold-based drug metabolism ensures that continued efforts will be made to develop gold-based anti-cancer agents.
Collapse
Affiliation(s)
- Chien Ing Yeo
- Research Centre for Crystalline Materials, School of Science and Technology, Sunway University. No. 5, Jalan Universiti, Bandar Sunway 47500, Malaysia.
| | - Kah Kooi Ooi
- Research Centre for Crystalline Materials, School of Science and Technology, Sunway University. No. 5, Jalan Universiti, Bandar Sunway 47500, Malaysia.
| | - Edward R T Tiekink
- Research Centre for Crystalline Materials, School of Science and Technology, Sunway University. No. 5, Jalan Universiti, Bandar Sunway 47500, Malaysia.
| |
Collapse
|
42
|
Patanjali P, Kumar R, Sourabh, Kumar A, Chaudhary P, Singh R. Reviewing Gold(III) complexes as effective biological operators. MAIN GROUP CHEMISTRY 2018. [DOI: 10.3233/mgc-180247] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Pooja Patanjali
- Department of Chemistry, Material/Organometallics Laboratory, Atma Ram Sanatan Dharma College, University of Delhi, Dhaula Kuan, New Delhi, India
| | - Ramesh Kumar
- Department of Chemistry, Material/Organometallics Laboratory, Atma Ram Sanatan Dharma College, University of Delhi, Dhaula Kuan, New Delhi, India
| | - Sourabh
- Department of Chemistry, Material/Organometallics Laboratory, Atma Ram Sanatan Dharma College, University of Delhi, Dhaula Kuan, New Delhi, India
| | - Amit Kumar
- Department of Chemistry, Material/Organometallics Laboratory, Atma Ram Sanatan Dharma College, University of Delhi, Dhaula Kuan, New Delhi, India
| | - Pratibha Chaudhary
- Maitreyi College, University of Delhi, Bapudham Complex, Chanakyapuri, New Delhi, India
| | - Rajeev Singh
- Department of Chemistry, Material/Organometallics Laboratory, Atma Ram Sanatan Dharma College, University of Delhi, Dhaula Kuan, New Delhi, India
| |
Collapse
|
43
|
Ostrovskaya LA, Korman DB, Grehova AK, Osipov AN, Bluhterova NV, Fomina MM, Rikova VA, Abzaeva KA. Experimental study of the antitumor effect of aurumacryl. Russ Chem Bull 2018. [DOI: 10.1007/s11172-017-2025-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
44
|
Altaf M, Monim-Ul-Mehboob M, Kawde AN, Corona G, Larcher R, Ogasawara M, Casagrande N, Celegato M, Borghese C, Siddik ZH, Aldinucci D, Isab AA. New bipyridine gold(III) dithiocarbamate-containing complexes exerted a potent anticancer activity against cisplatin-resistant cancer cells independent of p53 status. Oncotarget 2018; 8:490-505. [PMID: 27888799 PMCID: PMC5341752 DOI: 10.18632/oncotarget.13448] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 11/12/2016] [Indexed: 11/25/2022] Open
Abstract
We synthesized, characterized and tested in a panel of cancer cell lines, nine new bipyridine gold(III) dithiocarbamate-containing complexes. In vitro studies demonstrated that compounds 1, 2, 4, 5, 7 and 8 were the most cytotoxic in prostate, breast, ovarian cancer cell lines and in Hodgkin lymphoma cells with IC50 values lower than the reference drug cisplatin. The most active compound 1 was more active than cisplatin in ovarian (A2780cis and 2780CP-16) and breast cancer cisplatin-resistant cells. Compound 1 determined an alteration of the cellular redox homeostasis leading to increased ROS levels, a decrease in the mitochondrial membrane potential, cytochrome-c release from the mitochondria and activation of caspases 9 and 3. The ROS scavenger NAC suppressed ROS generation and rescued cells from damage. Compound 1 resulted more active in tumor cells than in normal human Mesenchymal stromal cells. Gold compounds were active independent of p53 status: exerted cytotoxic effects on a panel of non-small cell lung cancer cell lines with different p53 status and in the ovarian A2780 model where the p53 was knocked out. In conclusion, these promising results strongly indicate the need for further preclinical evaluation to test the clinical potential of these new gold(III) complexes.
Collapse
Affiliation(s)
- Muhammad Altaf
- Center of Excellence in Nanotechnology (CENT), King Fahd University of Petroleum and Minerals, Dhahran, Saudi Arabia
| | | | - Abdel-Nasser Kawde
- Department of Chemistry, King Fahd University of Petroleum and Minerals, Dhahran, Saudi Arabia
| | - Giuseppe Corona
- Department of Translational Research, CRO Aviano National Cancer Institute, Aviano, PN, Italy
| | - Roberto Larcher
- Center for Technological Transfer, Edmund Mach Foundation, Trento, Italy
| | - Marcia Ogasawara
- The University of Texas MD Anderson Cancer Center, Department of Experimental Therapeutics, Houston, Texas, USA
| | - Naike Casagrande
- Department of Experimental Oncology 2, CRO Aviano National Cancer Institute, Aviano, PN, Italy
| | - Marta Celegato
- Department of Experimental Oncology 2, CRO Aviano National Cancer Institute, Aviano, PN, Italy
| | - Cinzia Borghese
- Department of Experimental Oncology 2, CRO Aviano National Cancer Institute, Aviano, PN, Italy
| | - Zahid H Siddik
- The University of Texas MD Anderson Cancer Center, Department of Experimental Therapeutics, Houston, Texas, USA
| | - Donatella Aldinucci
- Department of Experimental Oncology 2, CRO Aviano National Cancer Institute, Aviano, PN, Italy
| | - Anvarhusein A Isab
- Department of Chemistry, King Fahd University of Petroleum and Minerals, Dhahran, Saudi Arabia
| |
Collapse
|
45
|
Borges AP, Gaspari APS, Oliveira CG, de Sousa SF, da Silva RS, Deflon VM, Machado AEH, Patrocínio AOT, Maia PIS. Photophysical and DFT Studies of Cationic Ag(I) Complexes with Thiosemicarbazides Derived from p
-Toluenesulfohydrazide. ChemistrySelect 2018. [DOI: 10.1002/slct.201800079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Alice P. Borges
- Department of Chemistry; Universidade Federal do Triângulo Mineiro, Av. Dr. Randolfo Borges 1400; 38025-440 Uberaba - MG Brazil
| | - Ana P. S. Gaspari
- Department of Chemistry; Faculdade de Filosofia; Ciências e Letras de Ribeirão Preto - FFCLRP-USP; Universidade de São Paulo, Avenida do Café s/n; 14040-903, Ribeirão Preto, SP Brazil
| | - Carolina G. Oliveira
- Department of Molecular Chemistry and Physics; Instituto de Química de São Carlos; Universidade de São Paulo, Av. Trabalhador Sãocarlense, 400; 13566-590, São Carlos - SP Brazil
| | - Sinval F. de Sousa
- Instituto de Química; Laboratório de Fotoquímica e Ciência de Materiais; Universidade Federal de Uberlândia, Av. João Naves de Ávila 2121; 38400-902, Uberlândia - MG Brazil
| | - Roberto S. da Silva
- Department of Chemistry; Faculdade de Filosofia; Ciências e Letras de Ribeirão Preto - FFCLRP-USP; Universidade de São Paulo, Avenida do Café s/n; 14040-903, Ribeirão Preto, SP Brazil
| | - Victor M. Deflon
- Department of Molecular Chemistry and Physics; Instituto de Química de São Carlos; Universidade de São Paulo, Av. Trabalhador Sãocarlense, 400; 13566-590, São Carlos - SP Brazil
| | - Antonio E. H. Machado
- Instituto de Química; Laboratório de Fotoquímica e Ciência de Materiais; Universidade Federal de Uberlândia, Av. João Naves de Ávila 2121; 38400-902, Uberlândia - MG Brazil
| | - Antonio O. T. Patrocínio
- Instituto de Química; Laboratório de Fotoquímica e Ciência de Materiais; Universidade Federal de Uberlândia, Av. João Naves de Ávila 2121; 38400-902, Uberlândia - MG Brazil
| | - Pedro I. S. Maia
- Department of Chemistry; Universidade Federal do Triângulo Mineiro, Av. Dr. Randolfo Borges 1400; 38025-440 Uberaba - MG Brazil
| |
Collapse
|
46
|
Porchia M, Pellei M, Marinelli M, Tisato F, Del Bello F, Santini C. New insights in Au-NHCs complexes as anticancer agents. Eur J Med Chem 2018; 146:709-746. [PMID: 29407992 DOI: 10.1016/j.ejmech.2018.01.065] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 01/18/2018] [Accepted: 01/19/2018] [Indexed: 01/03/2023]
Abstract
Within the research field of antitumor metal-based agents alternative to platinum drugs, gold(I/III) coordination complexes have always been in the forefront due mainly to the familiarity of medicinal chemists with gold compounds, whose application in medicine goes back in the ancient times, and to the rich chemistry shown by this metal. In the last decade, N-heterocyclic carbene ligands (NHC), a class of ligands that largely resembles the chemical properties of phosphines, became of interest for gold(I) medicinal applications, and since then, the research on NHC-gold(I/III) coordination complexes as potential antiproliferative agents boosted dramatically. Different classes of gold(I/III)-NHC complexes often showed an outstanding in vitro antiproliferative activity, however up to now very few in vivo data have been reported to corroborate the in vitro results. This review summarizes all achievements in the field of gold (I/III) complexes comprising NHC ligands proposed as potential antiproliferative agents in the period 2004-2016, and critically analyses biological data (mainly IC50 values) in relation to the chemical structures of Au compounds. The state of art of the in vivo studies so far described is also reported.
Collapse
Affiliation(s)
| | - Maura Pellei
- School of Science and Technology, Chemistry Division, University of Camerino, via S. Agostino 1, 62032 Camerino, Macerata, Italy.
| | - Marika Marinelli
- School of Science and Technology, Chemistry Division, University of Camerino, via S. Agostino 1, 62032 Camerino, Macerata, Italy
| | | | - Fabio Del Bello
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Macerata, Italy
| | - Carlo Santini
- School of Science and Technology, Chemistry Division, University of Camerino, via S. Agostino 1, 62032 Camerino, Macerata, Italy
| |
Collapse
|
47
|
Gonçalves AC, Carneiro ZA, Oliveira CG, Danuello A, Guerra W, Oliveira RJ, Ferreira FB, Veloso-Silva LL, Batista FA, Borges JC, de Albuquerque S, Deflon VM, Maia PI. Pt II , Pd II and Au III complexes with a thiosemicarbazone derived from diacethylmonooxime: Structural analysis, trypanocidal activity, cytotoxicity and first insight into the antiparasitic mechanism of action. Eur J Med Chem 2017; 141:615-631. [DOI: 10.1016/j.ejmech.2017.10.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/19/2017] [Accepted: 10/07/2017] [Indexed: 11/28/2022]
|
48
|
Casado-Sánchez A, Martín-Santos C, Padrón JM, Mas-Ballesté R, Navarro-Ranninger C, Alemán J, Cabrera S. Effect of electronic and steric properties of 8-substituted quinolines in gold(III) complexes: Synthesis, electrochemistry, stability, interactions and antiproliferative studies. J Inorg Biochem 2017; 174:111-118. [DOI: 10.1016/j.jinorgbio.2017.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/22/2017] [Accepted: 06/13/2017] [Indexed: 11/30/2022]
|
49
|
Ostrovskaya LA, Grehova AK, Korman DB, Osipov AN, Bluhterova NV, Fomina MM, Rikova VA, Abzaeva KA. Cellular effects of the antitumor drug aurumacryl. Biophysics (Nagoya-shi) 2017. [DOI: 10.1134/s0006350917030150] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
50
|
Milaeva E, Shpakovsky D, Dyadchenko V, Gryzlov A, Gracheva Y, Antonenko T, Parulava M, Albov D, Aslanov L, Dubova L, Shevtsov P, Neganova M, Shevtsova E. Synthesis and biological activity of novel Au(I) complexes with a protective antioxidant 2,6-di-tert-butylphenol group. Polyhedron 2017. [DOI: 10.1016/j.poly.2016.08.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|