1
|
Ferraro G, Terán A, Galardo F, Lucignano R, Picone D, Massai L, Fasulo F, Muñoz-García AB, Messori L, Herrero S, Merlino A. Deciphering the role of neutral diruthenium complexes in protein binding. Int J Biol Macromol 2024; 283:137691. [PMID: 39551297 DOI: 10.1016/j.ijbiomac.2024.137691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/19/2024]
Abstract
The charge of paddlewheel diruthenium complexes has a major role in defining their interaction with proteins: negatively charged complexes bind proteins non-covalently, while cationic complexes form adducts where the Ru2 core binds to Asp side chains at the equatorial sites, or to the main chain carbonyl groups or the side chains of His, Arg or Lys residues at the axial sites. Here we study the interactions of the neutral compound [Ru2(D-p-FPhF)(O2CCH3)2(O2CO)]·3H2O (D-p-FPhF- = N,N'-bis(4-fluorophenyl)formamidinate), a very rare example of a paddlewheel diruthenium compound with three different equatorial ligands, with the model protein bovine pancreatic ribonuclease (RNase A) by means of UV-visible absorption spectroscopy, circular dichroism (CD), electrospray ionization mass spectrometry (ESI-MS) and X-ray crystallography. It is the first attempt to investigate the binding of a neutral diruthenium compound to a protein. ESI-MS data indicate that, in solution, under the investigated experimental conditions, the diruthenium compound binds the protein upon the loss of an acetate ligand. The crystallographic results indicate the replacement of an acetate by two water molecules and the coordination of the [Ru2(D-p-FPhF)(O2CCH3)2(O2CO)(OH2)2]+ ion, that is expected to be a highly reactive species in the absence of the protein, to the imidazole ring of His105 at the axial site. The side chains of Glu9 and His119 are also identified as possible diruthenium binding sites. The binding significantly affects the protein ability to form dimers and higher-order oligomers, without significantly altering its secondary structure content and thermal stability. These data show that: i) Glu side chain has to be considered as a possible alternative binding site for diruthenium compounds, ii) diruthenium containing fragments that would be unstable in solution can be formed upon reaction of diruthenium compounds with a protein, iii) diruthenium compounds could be used as modulators of protein aggregation.
Collapse
Affiliation(s)
- Giarita Ferraro
- Department of Chemical Sciences, University of Naples Federico II, Complesso Univ. di Monte Sant'Angelo, via Cinthia, 26, 80126 Naples, Italy
| | - Aarón Terán
- Department of Physics "Ettore Pancini", University of Naples Federico II, Complesso Univ. di Monte Sant'Angelo, via Cinthia, 26, 80126 Naples, Italy
| | - Francesco Galardo
- Department of Chemical Sciences, University of Naples Federico II, Complesso Univ. di Monte Sant'Angelo, via Cinthia, 26, 80126 Naples, Italy
| | - Rosanna Lucignano
- Department of Chemical Sciences, University of Naples Federico II, Complesso Univ. di Monte Sant'Angelo, via Cinthia, 26, 80126 Naples, Italy
| | - Delia Picone
- Department of Chemical Sciences, University of Naples Federico II, Complesso Univ. di Monte Sant'Angelo, via Cinthia, 26, 80126 Naples, Italy
| | - Lara Massai
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 3-13, 50019, Sesto Fiorentino, Florence, Italy
| | - Francesca Fasulo
- Department of Physics "Ettore Pancini", University of Naples Federico II, Complesso Univ. di Monte Sant'Angelo, via Cinthia, 26, 80126 Naples, Italy
| | - Ana B Muñoz-García
- Department of Physics "Ettore Pancini", University of Naples Federico II, Complesso Univ. di Monte Sant'Angelo, via Cinthia, 26, 80126 Naples, Italy
| | - Luigi Messori
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 3-13, 50019, Sesto Fiorentino, Florence, Italy
| | - Santiago Herrero
- MatMoPol Research Group, Department of Inorganic Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, Avda. Complutense s/n, 28040, Madrid, Spain; Knowledge Technology Institute, Complutense University of Madrid, Campus de Somosaguas, 28223, Pozuelo de Alarcón, Madrid, Spain
| | - Antonello Merlino
- Department of Chemical Sciences, University of Naples Federico II, Complesso Univ. di Monte Sant'Angelo, via Cinthia, 26, 80126 Naples, Italy.
| |
Collapse
|
2
|
Cirri D, Di Leo R, Chiaverini L, Tolbatov I, Marrone A, Messori L, Pratesi A, La Mendola D, Marzo T. Non-Medical Applications of Inorganic Medicines. A Switch Based on Mechanistic Knowledge. Chemistry 2024; 30:e202402647. [PMID: 39158114 DOI: 10.1002/chem.202402647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 08/20/2024]
Abstract
Metals have been used in medicine for centuries. However, it was not until much later that the effects of inorganic drugs could be rationalized from a mechanistic point of view. Today, thanks to the technologies available, this approach has been functionally developed and implemented. It has been found that there is probably no single biological target for the pharmacological effects of most inorganic drugs. Herein, we present an overview of some integrated and multi-technique approaches to elucidate the molecular interactions underlying the biological effects of metallodrugs. On this premise, selected examples are used to illustrate how the information obtained on metal-based drugs and their respective mechanisms can become relevant for applications in fields other than medicine. For example, some well-known metallodrugs, which have been shown to bind specific amino acid residues of proteins, can be used to solve problems related to protein structure elucidation in crystallographic studies. Diruthenium tetraacetate can be used to catalyze the conversion of hydroxylamines to nitrones with a high selectivity when bound to lysozyme. Finally, a case study is presented in which an unprecedented palladium/arsenic-mediated catalytic cycle for nitrile hydration was discovered thanks to previous studies on the solution chemistry of the anticancer compound arsenoplatin-1 (AP-1).
Collapse
Affiliation(s)
- Damiano Cirri
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi, 13, 56124, Pisa, Italy
| | - Riccardo Di Leo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Lorenzo Chiaverini
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Iogann Tolbatov
- Department of Physics and Astronomy, University of Padova, Via F. Marzolo 8, 35131, Padova, Italy
| | - Alessandro Marrone
- Department of Pharmacy, University "G. D'Annunzio" Chieti-Pescara, Via dei Vestini, 31, 66100, Chieti, Italy
| | - Luigi Messori
- Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto Fiorentino, Italy
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi, 13, 56124, Pisa, Italy
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| |
Collapse
|
3
|
Cirri D, Marzo T, Mastrorilli P, Petrelli V, Todisco S, De Giglio E, Gellini C, Ricci M, Pratesi A, Messori L. Description of a Non-Canonical AsPt Blue Species Originating from the Aerobic Oxidation of AP-1 in Aqueous Solution. Int J Mol Sci 2024; 25:7408. [PMID: 39000514 PMCID: PMC11242394 DOI: 10.3390/ijms25137408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
The peculiar behavior of arsenoplatin-1, ([Pt(µ-NHC(CH3)O)2ClAs(OH)2], AP-1), in aqueous solution and the progressive appearance of a characteristic and intense blue color led us to carry out a more extensive investigation to determine the nature of this elusive chemical species, which we named "AsPt blue". A multi-technique approach was therefore implemented to describe the processes involved in the formation of AsPt blue, and some characteristic features of this intriguing species were revealed.
Collapse
Affiliation(s)
- Damiano Cirri
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy;
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy;
| | - Piero Mastrorilli
- Department of Civil, Environmental, Land, Building and Chemical Engineering (DICATECh), Polytechnic University of Bari, Via Orabona 4, 70125 Bari, Italy; (P.M.); (V.P.); (S.T.)
| | - Valentina Petrelli
- Department of Civil, Environmental, Land, Building and Chemical Engineering (DICATECh), Polytechnic University of Bari, Via Orabona 4, 70125 Bari, Italy; (P.M.); (V.P.); (S.T.)
| | - Stefano Todisco
- Department of Civil, Environmental, Land, Building and Chemical Engineering (DICATECh), Polytechnic University of Bari, Via Orabona 4, 70125 Bari, Italy; (P.M.); (V.P.); (S.T.)
| | - Elvira De Giglio
- Department of Chemistry, University of Bari “Aldo Moro”, Via Orabona 4, 70125 Bari, Italy;
| | - Cristina Gellini
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy; (C.G.); (M.R.)
| | - Marilena Ricci
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy; (C.G.); (M.R.)
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy;
| | - Luigi Messori
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy; (C.G.); (M.R.)
| |
Collapse
|
4
|
Nayek U, Shenoy TN, Abdul Salam AA. Data mining of arsenic-based small molecules geometrics present in Cambridge structural database. CHEMOSPHERE 2024; 360:142349. [PMID: 38763400 DOI: 10.1016/j.chemosphere.2024.142349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 04/27/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024]
Abstract
Arsenic, ubiquitous in various industrial processes and consumer products, presents both essential functions and considerable toxicity risks, driving extensive research into safer applications. Our investigation, drawing from 7182 arsenic-containing molecules in the Cambridge Structural Database (CSD), outlines their diverse bonding patterns. Notably, 51% of these molecules exhibit cyclic connections, while 49% display acyclic ones. Arsenic forms eight distinct bonding types with other elements, with significant interactions observed, particularly with phenyl rings, O3 and F6 moieties. Top interactions involve carbon, nitrogen, oxygen, fluorine, sulfur, and arsenic itself. We meticulously evaluated average bond lengths under three conditions: without an R-factor cut-off, with R-factor ≤0.075, and with R-factor ≤0.05, supporting the credibility of our results. Comparative analysis with existing literature data enriches our understanding of arsenic's bonding behaviour. Our findings illuminate the structural attributes, molecular coordination, geometry, and bond lengths of arsenic with 68 diverse atoms, enriching our comprehension of arsenic chemistry. These revelations not only offer a pathway for crafting innovative and safer arsenic-based compounds but also foster the evolution of arsenic detoxification mechanisms, tackling pivotal health and environmental challenges linked to arsenic exposure across different contexts.
Collapse
Affiliation(s)
- Upendra Nayek
- Department of Atomic and Molecular Physics, Manipal Academy of Higher Education, Manipal, 576 104, Karnataka, India
| | - Thripthi Nagesh Shenoy
- Department of Atomic and Molecular Physics, Manipal Academy of Higher Education, Manipal, 576 104, Karnataka, India
| | - Abdul Ajees Abdul Salam
- Department of Atomic and Molecular Physics, Manipal Academy of Higher Education, Manipal, 576 104, Karnataka, India.
| |
Collapse
|
5
|
Zhao J, Li X, Ma T, Chang B, Zhang B, Fang J. Glutathione-triggered prodrugs: Design strategies, potential applications, and perspectives. Med Res Rev 2024; 44:1013-1054. [PMID: 38140851 DOI: 10.1002/med.22007] [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: 10/17/2023] [Revised: 11/20/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023]
Abstract
The burgeoning prodrug strategy offers a promising avenue toward improving the efficacy and specificity of cytotoxic drugs. Elevated intracellular levels of glutathione (GSH) have been regarded as a hallmark of tumor cells and characteristic feature of the tumor microenvironment. Considering the pivotal involvement of elevated GSH in the tumorigenic process, a diverse repertoire of GSH-triggered prodrugs has been developed for cancer therapy, facilitating the attenuation of deleterious side effects associated with conventional chemotherapeutic agents and/or the attainment of more efficacious therapeutic outcomes. These prodrug formulations encompass a spectrum of architectures, spanning from small molecules to polymer-based and organic-inorganic nanomaterial constructs. Although the GSH-triggered prodrugs have been gaining increasing interests, a comprehensive review of the advancements made in the field is still lacking. To fill the existing lacuna, this review undertakes a retrospective analysis of noteworthy research endeavors, based on a categorization of these molecules by their diverse recognition units (i.e., disulfides, diselenides, Michael acceptors, and sulfonamides/sulfonates). This review also focuses on explaining the distinct benefits of employing various chemical architecture strategies in the design of these prodrug agents. Furthermore, we highlight the potential for synergistic functionality by incorporating multiple-targeting conjugates, theranostic entities, and combinational treatment modalities, all of which rely on the GSH-triggering. Overall, an extensive overview of the emerging field is presented in this review, highlighting the obstacles and opportunities that lie ahead. Our overarching goal is to furnish methodological guidance for the development of more efficacious GSH-triggered prodrugs in the future. By assessing the pros and cons of current GSH-triggered prodrugs, we expect that this review will be a handful reference for prodrug design, and would provide a guidance for improving the properties of prodrugs and discovering novel trigger scaffolds for constructing GSH-triggered prodrugs.
Collapse
Affiliation(s)
- Jintao Zhao
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Xinming Li
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
| | - Tao Ma
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Bingbing Chang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Baoxin Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Jianguo Fang
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
6
|
Troisi R, Tito G, Ferraro G, Sica F, Massai L, Geri A, Cirri D, Messori L, Merlino A. On the mechanism of action of arsenoplatins: arsenoplatin-1 binding to a B-DNA dodecamer. Dalton Trans 2024; 53:3476-3483. [PMID: 38270175 DOI: 10.1039/d3dt04302a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
The reaction of Pt-based anticancer agents with arsenic trioxide affords robust complexes known as arsenoplatins. The prototype of this family of anticancer compounds is arsenoplatin-1 (AP-1) that contains an As(OH)2 fragment linked to a Pt(II) moiety derived from cisplatin. Crystallographic and spectrometric studies of AP-1 binding to a B-DNA double helix dodecamer are presented here, in comparison with cisplatin and transplatin. Results reveal that AP-1, cisplatin and transplatin react differently with the DNA model system. Notably, in the AP-1/DNA systems, the Pt-As bond can break down with time and As-containing fragments can be released. These results have implications for the understanding of the mechanism of action of arsenoplatins.
Collapse
Affiliation(s)
- Romualdo Troisi
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, 80126, Naples, Italy.
- Institute of Biostructures and Bioimaging, CNR, via Pietro Castellino 111, 80131 Naples, Italy
| | - Gabriella Tito
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, 80126, Naples, Italy.
| | - Giarita Ferraro
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, 80126, Naples, Italy.
| | - Filomena Sica
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, 80126, Naples, Italy.
| | - Lara Massai
- Department of Chemistry "U. Schiff", University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Andrea Geri
- Department of Chemistry "U. Schiff", University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Damiano Cirri
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy
| | - Luigi Messori
- Department of Chemistry "U. Schiff", University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Antonello Merlino
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, 80126, Naples, Italy.
| |
Collapse
|
7
|
Bowden G, Scott PJH, Boros E. Radiochemistry: A Hot Field with Opportunities for Cool Chemistry. ACS CENTRAL SCIENCE 2023; 9:2183-2195. [PMID: 38161375 PMCID: PMC10755734 DOI: 10.1021/acscentsci.3c01050] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 01/03/2024]
Abstract
Recent Food and Drug Administration (FDA) approval of diagnostic and therapeutic radiopharmaceuticals and concurrent miniaturization of particle accelerators leading to improved access has fueled interest in the development of chemical transformations suitable for short-lived radioactive isotopes on the tracer scale. This recent renaissance of radiochemistry is paired with new opportunities to study fundamental chemical behavior and reactivity of elements to improve their production, separation, and incorporation into bioactive molecules to generate new radiopharmaceuticals. This outlook outlines pertinent challenges in the field of radiochemistry and indicates areas of opportunity for chemical discovery and development, including those of clinically established (C-11, F-18) and experimental radionuclides in preclinical development across the periodic table.
Collapse
Affiliation(s)
- Gregory
D. Bowden
- Department
of Radiology, University of Michigan, 1301 Catherine, Ann Arbor, Michigan 48109, United States
- Werner
Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, 72074 Tuebingen, Germany
- Cluster
of Excellence iFIT (EXC 2180) “Image Guided and Functionally
Instructed Tumor Therapies”, Eberhard
Karls University of Tuebingen, 72074 Tuebingen, Germany
| | - Peter J. H. Scott
- Department
of Radiology, University of Michigan, 1301 Catherine, Ann Arbor, Michigan 48109, United States
| | - Eszter Boros
- Department
of Chemistry, University of Wisconsin, 1101 University Avenue, Madison, Wisconsin 53706, United States
| |
Collapse
|
8
|
Zhang J, Bhattacharya S, Nisar T, Wagner V, Kortz U. Discrete Platinum(II/IV)-Arsenito Clusters with Pt-As and Pt-O Bonding: [Pt IV(As 3O 6) 2] 2-, [Pt 4II(H 2AsO 3) 6(HAsO 3) 2] 2-, and [Pt 2IIAs 6W 4O 28] 10. Inorg Chem 2023; 62:19603-19611. [PMID: 37971601 DOI: 10.1021/acs.inorgchem.3c02967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
The first two discrete, fully inorganic platinum(II/IV)-arsenito clusters, [fac-PtIV(As3O6)2]2- (PtAs6) and [Pt4II(H2AsO3)6(HAsO3)2]2- (Pt4As8), as well as the platinum(II)-arsenito heteropolytungstate [Pt2IIAs6W4O28]10- (Pt2As6W4), have been synthesized in aqueous media using simple one-pot reaction conditions. In PtAs6, a PtIV ion is coordinated to two cyclic, tridentate As3O6 units via oxo-donation (PtIV-O ∼ 2.02 Å). In Pt4As8, each PtII ion is coordinated to four AsO3 ligands via two oxygens and two AsIII atoms in a square-planar fashion (PtII-AsIII 2.31 Å, PtII-O 2.07 Å), resulting in an open cage-like structure, which forms a strong tetrameric assembly in the solid state mediated by two K+ counterions. In Pt2As6W4, each PtII ion is coordinated by the As atoms of three AsO3 ligands (PtII-AsIII 2.38 Å) and an oxo group (PtII-O 2.07 Å) in addition to bridging two tungsten ions, and this polyanion was characterized in solution by 195Pt NMR.
Collapse
Affiliation(s)
- Jiayao Zhang
- School of Science, Constructor University, Campus Ring 1, 28759 Bremen, Germany
| | - Saurav Bhattacharya
- School of Science, Constructor University, Campus Ring 1, 28759 Bremen, Germany
- Department of Chemistry, BITS Pilani K. K. Birla Goa Campus, Zuarinagar 403726, Goa, India
| | - Talha Nisar
- School of Science, Constructor University, Campus Ring 1, 28759 Bremen, Germany
| | - Veit Wagner
- School of Science, Constructor University, Campus Ring 1, 28759 Bremen, Germany
| | - Ulrich Kortz
- School of Science, Constructor University, Campus Ring 1, 28759 Bremen, Germany
| |
Collapse
|
9
|
Paul NP, Galván AE, Yoshinaga-Sakurai K, Rosen BP, Yoshinaga M. Arsenic in medicine: past, present and future. Biometals 2023; 36:283-301. [PMID: 35190937 PMCID: PMC8860286 DOI: 10.1007/s10534-022-00371-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 02/05/2022] [Indexed: 12/17/2022]
Abstract
Arsenicals are one of the oldest treatments for a variety of human disorders. Although infamous for its toxicity, arsenic is paradoxically a therapeutic agent that has been used since ancient times for the treatment of multiple diseases. The use of most arsenic-based drugs was abandoned with the discovery of antibiotics in the 1940s, but a few remained in use such as those for the treatment of trypanosomiasis. In the 1970s, arsenic trioxide, the active ingredient in a traditional Chinese medicine, was shown to produce dramatic remission of acute promyelocytic leukemia similar to the effect of all-trans retinoic acid. Since then, there has been a renewed interest in the clinical use of arsenicals. Here the ancient and modern medicinal uses of inorganic and organic arsenicals are reviewed. Included are antimicrobial, antiviral, antiparasitic and anticancer applications. In the face of increasing antibiotic resistance and the emergence of deadly pathogens such as the severe acute respiratory syndrome coronavirus 2, we propose revisiting arsenicals with proven efficacy to combat emerging pathogens. Current advances in science and technology can be employed to design newer arsenical drugs with high therapeutic index. These novel arsenicals can be used in combination with existing drugs or serve as valuable alternatives in the fight against cancer and emerging pathogens. The discovery of the pentavalent arsenic-containing antibiotic arsinothricin, which is effective against multidrug-resistant pathogens, illustrates the future potential of this new class of organoarsenical antibiotics.
Collapse
Affiliation(s)
- Ngozi P Paul
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Adriana E Galván
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Kunie Yoshinaga-Sakurai
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Barry P Rosen
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA.
| | - Masafumi Yoshinaga
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| |
Collapse
|
10
|
Merlino A. Metallodrug binding to serum albumin: Lessons from biophysical and structural studies. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
11
|
Marzo T, Messori L. Protein targets for anticancer metal based drugs. COMPREHENSIVE INORGANIC CHEMISTRY III 2023:794-807. [DOI: 10.1016/b978-0-12-823144-9.00078-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
12
|
Chiaverini L, Marzo T, La Mendola D. AS101: An overview on a leading tellurium-based prodrug. Inorganica Chim Acta 2022. [DOI: 10.1016/j.ica.2022.121048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
13
|
Ferraro G, Merlino A. Metallodrugs: Mechanisms of Action, Molecular Targets and Biological Activity. Int J Mol Sci 2022; 23:ijms23073504. [PMID: 35408863 PMCID: PMC8998277 DOI: 10.3390/ijms23073504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/21/2022] [Indexed: 01/27/2023] Open
Affiliation(s)
- Giarita Ferraro
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cinthia 21, I-80126 Naples, Italy
| | - Antonello Merlino
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cinthia 21, I-80126 Naples, Italy
| |
Collapse
|
14
|
Tolbatov I, Cirri D, Tarchi M, Marzo T, Coletti C, Marrone A, Messori L, Re N, Massai L. Reactions of Arsenoplatin-1 with Protein Targets: A Combined Experimental and Theoretical Study. Inorg Chem 2022; 61:3240-3248. [PMID: 35137586 PMCID: PMC8864615 DOI: 10.1021/acs.inorgchem.1c03732] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Indexed: 11/30/2022]
Abstract
Arsenoplatin-1 (AP-1) is a dual-action anticancer metallodrug with a promising pharmacological profile that features the simultaneous presence of a cisplatin-like center and an arsenite center. We investigated its interactions with proteins through a joint experimental and theoretical approach. The reactivity of AP-1 with a variety of proteins, including carbonic anhydrase (CA), superoxide dismutase (SOD), myoglobin (Mb), glyceraldehyde 3-phosphate dehydrogenase (GAPDH), and human serum albumin (HSA), was analyzed by means of electrospray ionization mass spectrometry (ESI MS) measurements. In accordance with previous observations, ESI MS experiments revealed that the obtained metallodrug-protein adducts originated from the binding of the [(AP-1)-Cl]+ fragment to accessible protein residues. Remarkably, in two cases, i.e., Mb and GAPDH, the formation of a bound metallic fragment that lacked the arsenic center was highlighted. The reactions of AP-1 with various nucleophiles side chains of neutral histidine, methionine, cysteine, and selenocysteine, in neutral form as well as cysteine and selenocysteine in anionic form, were subsequently analyzed through a computational approach. We found that the aquation of AP-1 is energetically disfavored, with a reaction free energy of +19.2 kcal/mol demonstrating that AP-1 presumably attacks its biological targets through the exchange of the chloride ligand. The theoretical analysis of thermodynamics and kinetics for the ligand-exchange processes of AP-1 with His, Met, Cys, Sec, Cys-, and Sec- side chain models unveils that only neutral histidine and deprotonated cysteine and selenocysteine are able to effectively replace the chloride ligand in AP-1.
Collapse
Affiliation(s)
- Iogann Tolbatov
- Institut
de Chimie Moleculaire de l’Université de Bourgogne (ICMUB),
Université de Bourgogne Franche-Comté (UBFC), Avenue Alain Savary 9, 21078 Dijon, France
| | - Damiano Cirri
- Department
of Chemistry and Industrial Chemistry, University
of Pisa, Via G. Moruzzi
13, 56124 Pisa, Italy
| | - Matteo Tarchi
- Department
of Chemistry, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| | - Tiziano Marzo
- Department
of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy
- CISUP
- Centre for Instrumentation Sharing (Centro per l’Integrazione
della Strumentazione Scientifica), University
of Pisa, 56126 Pisa, Italy
- University
Consortium for Research in the Chemistry of Metal ions in Biological
Systems (CIRCMSB), Via
Celso Ulpiani 27, 70126 Bari, Italy
| | - Cecilia Coletti
- Dipartimento
di Farmacia, Università “G
d’Annunzio” di Chieti-Pescara, Via dei Vestini 31, 66013, Chieti, Italy
| | - Alessandro Marrone
- Dipartimento
di Farmacia, Università “G
d’Annunzio” di Chieti-Pescara, Via dei Vestini 31, 66013, Chieti, Italy
| | - Luigi Messori
- Department
of Chemistry, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| | - Nazzareno Re
- Dipartimento
di Farmacia, Università “G
d’Annunzio” di Chieti-Pescara, Via dei Vestini 31, 66013, Chieti, Italy
| | - Lara Massai
- Department
of Chemistry, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
15
|
Shahabadi N, Mahdavi M, Zendehcheshm S. Can polyoxometalates (POMs) prevent of coronavirus 2019-nCoV cell entry? Interaction of POMs with TMPRSS2 and spike receptor domain complexed with ACE2 (ACE2-RBD): Virtual screening approaches. INFORMATICS IN MEDICINE UNLOCKED 2022; 29:100902. [PMID: 35284620 PMCID: PMC8896857 DOI: 10.1016/j.imu.2022.100902] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/14/2022] [Accepted: 03/03/2022] [Indexed: 12/22/2022] Open
Abstract
The unexpected appearance and global spread of COVID-19 create significant difficulties for healthcare systems and present an unusual challenge for the fast discovery of medicines to combat this fatal disease. Screening metallodrugs libraries from the medicinal inorganic chemistry society may expand the studied ‘chemical space’ and improve the probability of discovering effective anti-COVID drugs, including polyoxometalates. POMs are an oxygen-rich family of inorganic cluster systems that have previously been tested for antiviral action against different types of viruses. Human angiotensin-converting enzyme 2 (ACE2), human transmembrane protease serine 2 (TMPRSS2), and the SARS-CoV-2 spike glycoprotein are required for host cell-mediated viral entrance. Targeting these proteins demonstrates potential possibilities for preventing infections and transmissions in the initial stage. As a result, POMs with known antiviral effects were investigated for this purpose using molecular docking and dynamic simulations. This research shows that POMs can prevent SARS CoV-2 from entering cells by blocking TMPRSS2, which SARS-CoV-2 uses for spike glycoprotein priming. They may also engage with ACE2 and the spike glycoprotein and disrupt their binding by blocking the active sites. We think that a thorough investigation of POMs as possible anti-COVID-19 drugs will provide significant opportunities.
Collapse
|
16
|
Ferraro G, Demitri N, Vitale L, Sciortino G, Sanna D, Ugone V, Garribba E, Merlino A. Spectroscopic/Computational Characterization and the X-ray Structure of the Adduct of the V IVO-Picolinato Complex with RNase A. Inorg Chem 2021; 60:19098-19109. [PMID: 34847328 PMCID: PMC8693189 DOI: 10.1021/acs.inorgchem.1c02912] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Indexed: 12/12/2022]
Abstract
The structure, stability, and enzymatic activity of the adduct formed upon the reaction of the V-picolinato (pic) complex [VIVO(pic)2(H2O)], with an octahedral geometry and the water ligand in cis to the V═O group, with the bovine pancreatic ribonuclease (RNase A) were studied. While electrospray ionization-mass spectrometry, circular dichroism, and ultraviolet-visible absorption spectroscopy substantiate the interaction between the metal moiety and RNase A, electron paramagnetic resonance (EPR) allows us to determine that a carboxylate group, stemming from Asp or Glu residues, and imidazole nitrogen from His residues are involved in the V binding at acidic and physiological pH, respectively. Crystallographic data demonstrate that the VIVO(pic)2 moiety coordinates the side chain of Glu111 of RNase A, by substituting the equatorial water molecule at acidic pH. Computational methods confirm that Glu111 is the most affine residue and interacts favorably with the OC-6-23-Δ enantiomer establishing an extended network of hydrogen bonds and van der Waals stabilizations. By increasing the pH around neutrality, with the deprotonation of histidine side chains, the binding of the V complex to His105 and His119 could occur, with that to His105 which should be preferred when compared to that to the catalytically important His119. The binding of the V compound affects the enzymatic activity of RNase A, but it does not alter its overall structure and stability.
Collapse
Affiliation(s)
- Giarita Ferraro
- Department
of Chemical Sciences, University of Naples
Federico II, I-80126 Napoli, Italy
| | - Nicola Demitri
- Elettra−Sincrotrone
Trieste, S.S. 14 km 163.5
in Area Science Park, 34149 Trieste, Italy
| | - Luigi Vitale
- Department
of Chemical Sciences, University of Naples
Federico II, I-80126 Napoli, Italy
| | - Giuseppe Sciortino
- Institute
of Chemical Research of Catalonia (ICIQ), The Barcelona Institute
of Science and Technology, 43007 Tarragona, Spain
| | - Daniele Sanna
- Istituto
di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Trav. La Crucca 3, I-07100 Sassari, Italy
| | - Valeria Ugone
- Istituto
di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Trav. La Crucca 3, I-07100 Sassari, Italy
| | - Eugenio Garribba
- Dipartimento
di Scienze Mediche, Chirurgiche e Sperimentali, Università di Sassari, Viale San Pietro, I-07100 Sassari, Italy
| | - Antonello Merlino
- Department
of Chemical Sciences, University of Naples
Federico II, I-80126 Napoli, Italy
| |
Collapse
|
17
|
In Vitro Anti-SARS-CoV-2 Activity of Selected Metal Compounds and Potential Molecular Basis for Their Actions Based on Computational Study. Biomolecules 2021; 11:biom11121858. [PMID: 34944502 PMCID: PMC8699537 DOI: 10.3390/biom11121858] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/24/2022] Open
Abstract
Metal-based drugs represent a rich source of chemical substances of potential interest for the treatment of COVID-19. To this end, we have developed a small but representative panel of nine metal compounds, including both synthesized and commercially available complexes, suitable for medical application and tested them in vitro against the SARS-CoV-2 virus. The screening revealed that three compounds from the panel, i.e., the organogold(III) compound Aubipyc, the ruthenium(III) complex KP1019, and antimony trichloride (SbCl3), are endowed with notable antiviral properties and an acceptable cytotoxicity profile. These initial findings prompted us to perform a computational study to unveil the likely molecular basis of their antiviral actions. Calculations evidenced that the metalation of nucleophile sites in SARS-CoV-2 proteins or nucleobase strands, induced by Aubipyc, SbCl3, and KP1019, is likely to occur. Remarkably, we found that only the deprotonated forms of Cys and Sec residues can react favorably with these metallodrugs. The mechanistic implications of these findings are discussed.
Collapse
|
18
|
Zhao S, Yang Z, Jiang G, Huang S, Bian M, Lu Y, Liu W. An overview of anticancer platinum N-heterocyclic carbene complexes. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214217] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
19
|
Song ZL, Zhang J, Xu Q, Shi D, Yao X, Fang J. Structural Modification of Aminophenylarsenoxides Generates Candidates for Leukemia Treatment via Thioredoxin Reductase Inhibition. J Med Chem 2021; 64:16132-16146. [PMID: 34704769 DOI: 10.1021/acs.jmedchem.1c01441] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Upregulation of the selenoprotein thioredoxin reductase (TrxR) is of pathological significance in maintaining tumor phenotypes. Thus, TrxR inhibitors are promising cancer therapeutic agents. We prepared different amino-substituted phenylarsine oxides and evaluated their cytotoxicity and inhibition of TrxR. Compared with our reported p-substituted molecule (8), the o-substituted molecule (10) shows improved efficacy (nearly a fourfold increase) to kill leukemia HL-60 cells. Although the compounds 8 and 10 display similar potency to inhibit the purified TrxR, the o-substitution 10 exhibits higher potency than the p-substitution 8 to inhibit the cellular TrxR activity. Molecular docking results demonstrate the favorable weak interactions of the o-amino group with the TrxR C-terminal active site. Efficient inhibition of TrxR consequently induces the oxidative stress-mediated apoptosis of cancer cells. Silence of the TrxR expression sensitizes the cells to the arsenic compound treatment, further supporting the critical involvement of TrxR in the cellular actions of compound 10.
Collapse
Affiliation(s)
- Zi-Long Song
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.,Botanical Agrochemicals Research & Development Center, Lanzhou Jiaotong University, Lanzhou 730070, China
| | - Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Qianhe Xu
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Danfeng Shi
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Xiaojun Yao
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
20
|
Miodragović Ð, Qiang W, Sattar Waxali Z, Vitnik Ž, Vitnik V, Yang Y, Farrell A, Martin M, Ren J, O’Halloran TV. Iodide Analogs of Arsenoplatins-Potential Drug Candidates for Triple Negative Breast Cancers. Molecules 2021; 26:molecules26175421. [PMID: 34500854 PMCID: PMC8434261 DOI: 10.3390/molecules26175421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 01/18/2023] Open
Abstract
Patients with triple negative breast cancers (TNBCs)—highly aggressive tumors that do not express estrogen, progesterone, and human epidermal growth factor 2 receptors—have limited treatment options. Fewer than 30% of women with metastatic TNBC survive five years after their diagnosis, with a mortality rate within three months after a recurrence of 75%. Although TNBCs show a higher response to platinum therapy compared to other breast cancers, drug resistance remains a major obstacle; thus, platinum drugs with novel mechanisms are urgently needed. Arsenoplatins (APs) represent a novel class of anticancer agents designed to contain the pharmacophores of the two FDA approved drugs cisplatin and arsenic trioxide (As2O3) as one molecular entity. Here, we present the syntheses, crystal structures, DFT calculations, and antiproliferative activity of iodide analogs of AP-1 and AP-2, i.e., AP-5 and AP-4, respectively. Antiproliferative studies in TNBC cell lines reveal that all AP family members are more potent than cisplatin and As2O3 alone. DFT calculations demonstrate there is a low energy barrier for hydrolysis of the platinum-halide bonds in arsenoplatins, possibly contributing to their higher cytotoxicities compared to cisplatin.
Collapse
Affiliation(s)
- Ðenana Miodragović
- Department of Chemistry, Northeastern Illinois University, 5500 St. Louis Ave, Chicago, IL 60625, USA; (Ð.M.); (M.M.)
- Chemistry of Life Processes Institute, Northwestern University, 2170 Campus Drive, Evanston, IL 60208, USA; (W.Q.); (Z.S.W.); (Y.Y.); (J.R.)
| | - Wenan Qiang
- Chemistry of Life Processes Institute, Northwestern University, 2170 Campus Drive, Evanston, IL 60208, USA; (W.Q.); (Z.S.W.); (Y.Y.); (J.R.)
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 East Superior Street, Chicago, IL 60611, USA
| | - Zohra Sattar Waxali
- Chemistry of Life Processes Institute, Northwestern University, 2170 Campus Drive, Evanston, IL 60208, USA; (W.Q.); (Z.S.W.); (Y.Y.); (J.R.)
| | - Željko Vitnik
- Institute of Chemistry, Technology and Metallurgy, University of Belgrade, Njegoševa 12, 11000 Belgrade, Serbia; (Ž.V.); (V.V.)
| | - Vesna Vitnik
- Institute of Chemistry, Technology and Metallurgy, University of Belgrade, Njegoševa 12, 11000 Belgrade, Serbia; (Ž.V.); (V.V.)
| | - Yi Yang
- Chemistry of Life Processes Institute, Northwestern University, 2170 Campus Drive, Evanston, IL 60208, USA; (W.Q.); (Z.S.W.); (Y.Y.); (J.R.)
| | - Annie Farrell
- Department of Chemistry, University of Illinois at Urbana Champaign, 102 N. Neil St., Champaign, IL 61820, USA;
| | - Matthew Martin
- Department of Chemistry, Northeastern Illinois University, 5500 St. Louis Ave, Chicago, IL 60625, USA; (Ð.M.); (M.M.)
| | - Justin Ren
- Chemistry of Life Processes Institute, Northwestern University, 2170 Campus Drive, Evanston, IL 60208, USA; (W.Q.); (Z.S.W.); (Y.Y.); (J.R.)
| | - Thomas V. O’Halloran
- Chemistry of Life Processes Institute, Northwestern University, 2170 Campus Drive, Evanston, IL 60208, USA; (W.Q.); (Z.S.W.); (Y.Y.); (J.R.)
- Department of Chemistry and Department of Microbiology & Molecular Genetics, Michigan State University, 567 Wilson Rd., East Lansing, MI 48824, USA
- Correspondence: or ; Tel.: +1-847-491-5060; Fax: +1-847-467-1566
| |
Collapse
|
21
|
Sarpong-Kumankomah S, Gailer J. Application of a Novel Metallomics Tool to Probe the Fate of Metal-Based Anticancer Drugs in Blood Plasma: Potential, Challenges and Prospects. Curr Top Med Chem 2021; 21:48-58. [PMID: 32600232 DOI: 10.2174/1568026620666200628023540] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/20/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022]
Abstract
Although metallodrugs are used to treat a variety of human disorders and exhibit a remarkable diversity of therapeutic properties, they constitute only a tiny minority of all medicinal drugs that are currently on the market. This undesirable situation must be partially attributed to our general lack of understanding the fate of metallodrugs in the extremely ligand-rich environment of the bloodstream. The challenge of gaining insight into these bioinorganic processes can be overcome by the application of 'metallomics tools', which involve the analysis of biological fluids (e.g., blood plasma) with a separation method in conjunction with multi-element specific detectors. To this end, we have developed a metallomics tool that is based on size-exclusion chromatography (SEC) hyphenated to an inductively coupled plasma atomic emission spectrometer (ICP-AES). After the successful application of SEC-ICPAES to analyze plasma for endogenous copper, iron and zinc-metalloproteins, it was subsequently applied to probe the metabolism of a variety of metal-based anticancer drugs in plasma. The versatility of this metallomics tool is exemplified by the fact that it has provided insight into the metabolism of individual Pt-based drugs, the modulation of the metabolism of cisplatin by sulfur-containing compounds, the metabolism of two metal-based drugs that contain different metals as well as a bimetallic anticancer drug, which contained two different metals. After adding pharmacologically relevant doses of metallodrugs to plasma, the temporal analysis of aliquots by SEC-ICP-AES allows to observe metal-protein adducts, metallodrug-derived degradation products and the parent metallodrug(s). This unique capability allows to obtain comprehensive insight into the fate of metal-based drugs in plasma and can be extended to in vivo studies. Thus, the application of this metallomics tool to probe the fate of novel metalcomplexes that exert the desired biological activity in plasma has the potential to advance more of these to animal/preclinical studies to fully explore the potential that metallodrugs inherently offer.
Collapse
Affiliation(s)
| | - Jürgen Gailer
- Department of Chemistry, Faculty of Science, University of Calgary, Calgary, Canada
| |
Collapse
|
22
|
Marzo T, Mendola DL. Looking Beyond DNA as the Target of Inorganic Chemotherapy Agents: The Key Role of Protein Metalation and Metal Homeostasis. Curr Top Med Chem 2021; 21:3-5. [PMID: 33443001 DOI: 10.2174/156802662101201211094545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| |
Collapse
|
23
|
Marzo T, La Mendola D. The Effects on Angiogenesis of Relevant Inorganic Chemotherapeutics. Curr Top Med Chem 2021; 21:73-86. [PMID: 33243124 DOI: 10.2174/1568026620666201126163436] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Angiogenesis is a key process allowing the formation of blood vessels. It is crucial for all the tissues and organs, ensuring their function and growth. Angiogenesis is finely controlled by several mechanisms involving complex interactions between pro- or antiangiogenic factors, and an imbalance in this control chain may result in pathological conditions. Metals as copper, zinc and iron cover an essential role in regulating angiogenesis, thus therapies having physiological metals as target have been proposed. In addition, some complexes of heavier metal ions (e.g., Pt, Au, Ru) are currently used as established or experimental anticancer agents targeting genomic or non-genomic targets. These molecules may affect the angiogenic mechanisms determining different effects that have been only poorly and non-systematically investigated so far. Accordingly, in this review article, we aim to recapitulate the impact on the angiogenic process of some reference anticancer drugs, and how it is connected to the overall pharmacological effects. In addition, we highlight how the activity of these drugs can be related to the role of biological essential metal ions. Overall, this may allow a deeper description and understanding of the antineoplastic activity of both approved or experimental metal complexes, providing important insights for the synthesis of new inorganic drugs able to overcome resistance and recurrence phenomena.
Collapse
Affiliation(s)
- Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| |
Collapse
|
24
|
Ferraro G, Loreto D, Merlino A. Interaction of Platinum-based Drugs with Proteins: An Overview of Representative Crystallographic Studies. Curr Top Med Chem 2021; 21:6-27. [PMID: 32579504 DOI: 10.2174/1568026620666200624162213] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 12/27/2022]
Abstract
Pt-based drugs are widely used in clinics for the treatment of cancer. The mechanism of action of these molecules relies on their interaction with DNA. However, the recognition of these metal compounds by proteins plays an important role in defining pharmacokinetics, side effects and their overall pharmacological profiles. Single crystal X-ray diffraction studies provided important information on the molecular mechanisms at the basis of this process. Here, the molecular structures of representative adducts obtained upon reaction with proteins of selected Pt-based drugs, including cisplatin, carboplatin and oxaliplatin, are briefly described and comparatively examined. Data indicate that metal ligands play a significant role in driving the reaction of Pt compounds with proteins; non-covalent interactions that occur in the early steps of Pt compound/protein recognition process play a crucial role in defining the structure of the final Pt-protein adduct. In the metallated protein structures, Pt centers coordinate few protein side chains, such as His, Met, Cys, Asp, Glu and Lys residues upon releasing labile ligands.
Collapse
Affiliation(s)
- Giarita Ferraro
- Department of Chemistry Ugo Schiff, University of Florence, Sesto Fiorentino, Firenze, Italy
| | - Domenico Loreto
- Department of Chemical Sciences, University of Naples Federico II, Napoli, Italy
| | - Antonello Merlino
- Department of Chemical Sciences, University of Naples Federico II, Napoli, Italy
| |
Collapse
|
25
|
Wahiduzzaman M, Ota A, Hosokawa Y. Novel Mechanistic Insights into the Anti-cancer Mode of Arsenic Trioxide. Curr Cancer Drug Targets 2021; 20:115-129. [PMID: 31736446 DOI: 10.2174/1568009619666191021122006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/23/2019] [Accepted: 09/19/2019] [Indexed: 12/19/2022]
Abstract
Arsenic, a naturally-occurring toxic element, and a traditionally-used drug, has received a great deal of attention worldwide due to its curative anti-cancer properties in patients with acute promyelocytic leukemia. Among the arsenicals, arsenic trioxide has been most widely used as an anti-cancer drug. Recent advances in cancer therapeutics have led to a paradigm shift away from traditional cytotoxic drugs towards the targeting of proteins closely associated with driving the cancer phenotype. Due to the diverse anti-cancer effects of ATO on different types of malignancies, numerous studies have made efforts to uncover the mechanisms of ATO-induced tumor suppression. From in vitro cellular models to studies in clinical settings, ATO has been extensively studied. The outcomes of these studies have opened doors to establishing improved molecular-targeted therapies for cancer treatment. The efficacy of ATO has been augmented by combination with other drugs. In this review, we discuss recent arsenic-based cancer therapies and summarize the novel underlying molecular mechanisms of the anti-cancer effects of ATO.
Collapse
Affiliation(s)
- Md Wahiduzzaman
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| | - Akinobu Ota
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| | - Yoshitaka Hosokawa
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| |
Collapse
|
26
|
Cirri D, Bartoli F, Pratesi A, Baglini E, Barresi E, Marzo T. Strategies for the Improvement of Metal-Based Chemotherapeutic Treatments. Biomedicines 2021; 9:504. [PMID: 34064364 PMCID: PMC8147839 DOI: 10.3390/biomedicines9050504] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/12/2022] Open
Abstract
This article provides an overview of the various research approaches we have explored in recent years to improve metal-based agents for cancer or infection treatments. Although cisplatin, carboplatin, and oxaliplatin remain the cornerstones in tumor chemotherapy, the discovery and approval of novel inorganic anticancer drugs is a very slow process. Analogously, although a few promising inorganic drugs have found clinical application against parasitic or bacterial infections, their use remains relatively limited. Moreover, the discovery process is often affected by small therapeutic enhancements that are not attractive for the pharmaceutical industry. However, the availability of increasing mechanistic information for the modes of action of established inorganic drugs is fueling the exploration of various approaches for developing effective inorganic chemotherapy agents. Through a series of examples, some from our own research experience, we focus our attention on a number of promising strategies, including (1) drug repurposing, (2) the simple modification of the chemical structures of approved metal-based drugs, (3) testing novel drug combinations, and (4) newly synthesized complexes coupling different anticancer drugs. Accordingly, we aim to suggest and summarize a series of reliable approaches that are exploitable for the development of improved and innovative treatments.
Collapse
Affiliation(s)
- Damiano Cirri
- Department of Chemistry and Industrial Chemistry (DCCI), Univerisity of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy;
| | - Francesco Bartoli
- Department of Translational Research and of New Surgical and Medical Technologies, Univerisity of Pisa, Via Risorgimento, 36, 56126 Pisa, Italy;
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry (DCCI), Univerisity of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy;
| | - Emma Baglini
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy; (E.B.); (E.B.)
| | - Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy; (E.B.); (E.B.)
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy; (E.B.); (E.B.)
| |
Collapse
|
27
|
Chu B, Mo X, Chen Z, Zhang M, Liang Y, Hu H, Liu D, Liang F. Synthesis and anticancer activity of mixed ligand 3d metal complexes. Metallomics 2021; 13:6184049. [PMID: 33755727 DOI: 10.1093/mtomcs/mfab011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/13/2021] [Accepted: 03/17/2021] [Indexed: 11/13/2022]
Abstract
Our previously reported copper-based complexes of tropolone show nice antitumor effects, but with high cytotoxicity to normal cells, which is presumably caused by copper ions. Here, we managed to achieve this challenge by using other 3D metals to replace copper ions. We thus prepared four mononuclear 3D metal complexes [M(phen)L2] (M = Mn, Co, Ni, and Zn for 1-4, respectively). Complexes 1 and 4 show selectivity on different cancer cell lines with much lower cytotoxicity to normal cells than cisplatin. The anticancer effects for complexes 2 and 3 on the tested cancer cell lines are very poor. It revealed a tuning effect of different metal ions on the anticancer activities with those for Mn(II) and Zn(II) being much higher than those for Co(II) and Ni(II) in this system. Among them, complex 1 presents a best anticancer effect on HeLa cells comparable to cisplatin. It overcame the afore-mentioned shortage of high cytotoxicity to normal cells for the reported Cu(II) complexes. It revealed from the mechanistic studies that complex 1 mainly induces apoptosis through the mitochondrial pathway by increasing intracellular reactive oxygen species, releasing Ca2+, and activating Caspase 9 and proapoptotic gene Bax.
Collapse
Affiliation(s)
- Bo Chu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China
| | - Xiyu Mo
- Department of Food and Chemical Engineering, Liuzhou Institute of Technology, Liuzhou 545616, P. R. China
| | - Zilu Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China
| | - Mingling Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China
| | - Yuning Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China
| | - Huancheng Hu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China
| | - Dongcheng Liu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China
| | - Fupei Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, P. R. China.,Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin 541004, P. R. China
| |
Collapse
|
28
|
Arsenoplatin-Ferritin Nanocage: Structure and Cytotoxicity. Int J Mol Sci 2021; 22:ijms22041874. [PMID: 33668605 PMCID: PMC7918638 DOI: 10.3390/ijms22041874] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/28/2021] [Accepted: 02/04/2021] [Indexed: 01/07/2023] Open
Abstract
Arsenoplatin-1 (AP-1), the prototype of a novel class of metallodrugs containing a PtAs(OH)2 core, was encapsulated within the apoferritin (AFt) nanocage. UV-Vis absorption spectroscopy and inductively coupled plasma-atomic emission spectroscopy measurements confirmed metallodrug encapsulation and allowed us to determine the average amount of AP-1 trapped inside the cage. The X-ray structure of AP-1-encapsulated AFt was solved at 1.50 Å. Diffraction data revealed that an AP-1 fragment coordinates the side chain of a His residue. The biological activity of AP-1-loaded AFt was comparatively tested on a few representative cancer and non-cancer cell lines. Even though the presence of the cage reduces the overall cytotoxicity of AP-1, it improves its selectivity towards cancer cells.
Collapse
|
29
|
Parise A, Russo N, Marino T. The platination mechanism of RNase A by arsenoplatin: insight from the theoretical study. Inorg Chem Front 2021. [DOI: 10.1039/d0qi01165g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A detailed metalation process of the bovine pancreatic ribonuclease (RNase A) by a novel multitarget anti-cancer agent arsenoplatin-1, ([Pt(μ-NHC(CH3)O)2ClAs(OH)2]), performed at DFT level and using different models size is provided.
Collapse
Affiliation(s)
- A. Parise
- Dipartimento di Chimica e Tecnologie Chimiche
- Università della Calabria
- 87036 Arcavacata di Rende
- Italy
- Université Paris-Saclay
| | - N. Russo
- Dipartimento di Chimica e Tecnologie Chimiche
- Università della Calabria
- 87036 Arcavacata di Rende
- Italy
| | - T. Marino
- Dipartimento di Chimica e Tecnologie Chimiche
- Università della Calabria
- 87036 Arcavacata di Rende
- Italy
| |
Collapse
|
30
|
Abstract
Recent advances in structural studies unveiling the basis of the metal compounds/protein recognition process are discussed.
Collapse
Affiliation(s)
- Antonello Merlino
- Department of Chemical Sciences
- University of Naples Federico II
- Complesso Universitario di Monte Sant’Angelo
- Napoli
- Italy
| |
Collapse
|
31
|
Ferraro G, Cirri D, Marzo T, Pratesi A, Messori L, Merlino A. The first step of arsenoplatin-1 aggregation in solution unveiled by solving the crystal structure of its protein adduct. Dalton Trans 2020; 50:68-71. [PMID: 33320144 DOI: 10.1039/d0dt04068a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Arsenoplatin-1 (AP-1) is an innovative dual-action anticancer agent that contains a platinum(ii) center coordinated to an arsenous acid moiety. We found that AP-1 spontaneously aggregates in aqueous solutions generating oligomeric species of increasing length. Afterward, we succeeded in solving the crystal structure of the adduct formed between the model protein lysozyme and an early AP-1 oligomer that turned out to be a trimer. Remarkably, this crystal structure traps an early stage of AP-1 aggregation offering detailed insight into the molecular process of the oligomer's growth.
Collapse
Affiliation(s)
- Giarita Ferraro
- Department of Chemistry, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, FI, Italy.
| | | | | | | | | | | |
Collapse
|
32
|
Kaspi-Kaneti AW, Bhandari S, Schubert A, Huang SD, Dunietz BD. Cyanide Bridged Platinum-Iron Complexes as Cisplatin Prodrug Systems: Design and Computational Study. Chemphyschem 2020; 22:106-111. [PMID: 33098742 DOI: 10.1002/cphc.202000748] [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: 08/30/2020] [Revised: 10/13/2020] [Indexed: 11/09/2022]
Abstract
The potential role of cyanide-bridged platinum-iron complexes as an anti-cancer Pt(IV) prodrug is studied. We present design principles of a dual-function prodrug that can upon reduction dissociate and release concurrently six cisplatin units and a ferricyanide anion per prodrug unit. The prodrug molecule is a unique complex of hepta metal centers consisting of a ferricyanide core with six Pt(IV) centers each bonded to the Fe(III) core through a cyano ligand. The functionality of the prodrug is addressed through density functional theory (DFT) calculations.
Collapse
Affiliation(s)
- Ariela W Kaspi-Kaneti
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH, 44242, USA
| | - Srijana Bhandari
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH, 44242, USA
| | - Alexander Schubert
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH, 44242, USA.,Present address: Institute of Physical Chemistry, Friedrich Schiller University Jena, 07743, Jena, Germany
| | - Songping D Huang
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH, 44242, USA
| | - Barry D Dunietz
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH, 44242, USA
| |
Collapse
|
33
|
Tolbatov I, Coletti C, Marrone A, Re N. Reactivity of arsenoplatin complex versus water and thiocyanate: a DFT benchmark study. Theor Chem Acc 2020. [DOI: 10.1007/s00214-020-02694-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AbstractSeven different density functionals, including GGAs, meta-GGAs, hybrids and range-separated hybrids, and considering Grimme’s empirical dispersion correction (M06-L, M06-2X, PBE0, B3LYP, B3LYP-D3, CAM-B3LYP, ωB97X) have been tested for their performance in the prediction of molecular structures, energies and energy barriers for a class of newly developed antitumor platinum complexes involving main group heavy elements such as arsenic. The calculated structural parameters, energies and energy barriers have been compared to the available experimental data. The results show that range-separated hybrid functionals CAM-B3LYP and ωB97X give good results in predicting both geometrical parameters and isomerization energies and barrier heights and are promising new tools for the theoretical study of novel platinum(II) arsenic compounds.
Collapse
|
34
|
Alberto ME, Mazzone G, Regina C, Russo N, Sicilia E. Theoretical exploration of the photophysical properties of two-component Ru II-porphyrin dyes as promising assemblies for a combined antitumor effect. Dalton Trans 2020; 49:12653-12661. [PMID: 32870211 DOI: 10.1039/d0dt02197k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Due to the extraordinary success of porphyrins in photodynamic therapy (PDT) and Ru compounds as chemotherapeutics, a series of RuII-porphyrin complexes have recently been synthesized and proposed as promising dual-action therapeutic agents. The results of a careful DFT and TDDFT investigation on four mononuclear pyridyl triphenylporphyrin RuII-arene complexes are herein reported and compared with those obtained for the metal-free derivatives. The investigation aims at shedding light on the modulation of the photophysical properties of the light absorber upon metalation and exploring the hydrolysis process of the RuII-moiety in the presence of the bulky porphyrin unit. Type I and Type II photoreactions were analyzed computing absorption spectra, singlet-triplet energy gaps, spin orbit coupling constants and vertical electron affinity (VEA) along with ionization potentials (VIP) for all the investigated compounds, while the chloride/water exchange reaction kinetics were determined by exploring the first and second aquation reactions of the Ru-moiety. Despite the highly similar photophysical properties displayed by the members of this class of compounds, an analysis of the hydrolysis processes in the dark allows to point out an interesting difference related to the type of pyridylporphyrin isomer and could be a preliminary explanation of the greater phototoxicity experimentally found for 3'-pyridyl substituted compounds.
Collapse
Affiliation(s)
- Marta Erminia Alberto
- Department of Chemistry and Chemical Technologies, Università della Calabria, 87036, Arcavacata di Rende, CS, Italy.
| | - Gloria Mazzone
- Department of Chemistry and Chemical Technologies, Università della Calabria, 87036, Arcavacata di Rende, CS, Italy.
| | - Claudia Regina
- Department of Chemistry and Chemical Technologies, Università della Calabria, 87036, Arcavacata di Rende, CS, Italy.
| | - Nino Russo
- Department of Chemistry and Chemical Technologies, Università della Calabria, 87036, Arcavacata di Rende, CS, Italy.
| | - Emilia Sicilia
- Department of Chemistry and Chemical Technologies, Università della Calabria, 87036, Arcavacata di Rende, CS, Italy.
| |
Collapse
|
35
|
Cirri D, Pratesi A, Marzo T, Messori L. Metallo therapeutics for COVID-19. Exploiting metal-based compounds for the discovery of new antiviral drugs. Expert Opin Drug Discov 2020; 16:39-46. [PMID: 32915656 DOI: 10.1080/17460441.2020.1819236] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The COVID-19 pandemic poses an unprecedented challenge for the rapid discovery of drugs against this life-threatening disease. Owing to the peculiar features of the metal centers that are currently used in medicinal chemistry, metallodrugs might offer an excellent opportunity to achieve this goal. AREAS COVERED Two main strategies for developing metal-based drugs against the SARS-CoV-2 are herein illustrated. Firstly, a few clinically approved metallodrugs could be evaluated in patients according to a 'drug repurposing' approach. To this respect, the gold drug auranofin seems a promising candidate, but some other clinically established metal compounds are worthy of a careful evaluation as well. On the other hand, libraries of inorganic compounds, featuring a large chemical diversity, should be screened to identify the most effective molecules. This second strategy might be assisted by a pathway-driven discovery approach arising from a preliminary knowledge of the mode of action, exploitable to inhibit the functional activities of the key viral proteins. Also, attention must be paid to selectivity and toxicity issues. EXPERT OPINION The medicinal inorganic chemistry community may offer a valuable contribution against COVID-19. The screening of metallodrugs' libraries can expand the explored 'chemical space' and increase the chance of finding effective anti-COVID agents.
Collapse
Affiliation(s)
- Damiano Cirri
- Department of Chemistry and Industrial Chemistry, University of Pisa , Pisa, Italy.,Laboratory of Metals in Medicine (Metmed), Department of Chemistry "U. Schiff", University of Florence , Sesto Fiorentino, Italy
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry, University of Pisa , Pisa, Italy
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa , Pisa, Italy
| | - Luigi Messori
- Laboratory of Metals in Medicine (Metmed), Department of Chemistry "U. Schiff", University of Florence , Sesto Fiorentino, Italy
| |
Collapse
|
36
|
Zhang C, Gao L, Yuan Q, Zhao L, Niu W, Cai P, Li J, Han X, He Z, Gao F, Wang Y, Jiang H, Chai Z, Gao X. Is GSH Chelated Pt Molecule Inactive in Anti-Cancer Treatment? A Case Study of Pt 6 GS 4. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2002044. [PMID: 32500659 DOI: 10.1002/smll.202002044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/24/2020] [Indexed: 06/11/2023]
Abstract
Platinum (Pt) drugs are widely used in anti-cancer treatment although many reports advocated that tumor cells could inactivate Pt drugs via glutathione-Pt (GSH-Pt) adducts formation. To date, GSH chelated Pt molecules have not been assessed in cancer treatment because GSH-Pt adducts are not capable of killing cancer cells, which is widely accepted and well followed. In this report, endogenous biothiol is utilized to precisely synthesize a GSH chelated Pt molecule (Pt6 GS4 ). This Pt6 GS4 molecule can be well taken up by aggressive triple negative breast cancer (TNBC) cells. Subsequently, its metabolites could enter nuclei to interact with DNA, finally the DNA-Pt complex triggers TNBC cell apoptosis via the p53 pathway. Impressively, high efficacy for anti-cancer treatment is achieved by Pt6 GS4 both in vitro and in vivo when compared with traditional first-line carboplatin in the same dosage. Compared with carboplatin, Pt6 GS4 keeps tumor bearing mice alive for a longer time and is non-toxic for the liver and kidneys. This work opens a route to explore polynuclear Pt compound with accurate architecture for enhancing therapeutic effects and reducing systemic toxicity.
Collapse
Affiliation(s)
- Chunyu Zhang
- Department of Chemistry and Chemical Engineering, Beijing University of Technology, Beijing, 100124, P. R. China
| | - Liang Gao
- Department of Chemistry and Chemical Engineering, Beijing University of Technology, Beijing, 100124, P. R. China
| | - Qing Yuan
- Department of Chemistry and Chemical Engineering, Beijing University of Technology, Beijing, 100124, P. R. China
| | - Lina Zhao
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Wenchao Niu
- Department of Chemistry and Chemical Engineering, Beijing University of Technology, Beijing, 100124, P. R. China
| | - Pengju Cai
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jiaojiao Li
- Department of Chemistry and Chemical Engineering, Beijing University of Technology, Beijing, 100124, P. R. China
| | - Xu Han
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zhesheng He
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Fuping Gao
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yaling Wang
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Huaidong Jiang
- School of Physical Science and Technology, Shanghai Tech University, Shanghai, 201210, P. R. China
| | - Zhifang Chai
- CAS Key Laboratory for the Biological Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xueyun Gao
- Department of Chemistry and Chemical Engineering, Beijing University of Technology, Beijing, 100124, P. R. China
| |
Collapse
|
37
|
An anticancer gold(III)-activated porphyrin scaffold that covalently modifies protein cysteine thiols. Proc Natl Acad Sci U S A 2020; 117:1321-1329. [PMID: 31896586 DOI: 10.1073/pnas.1915202117] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Cysteine thiols of many cancer-associated proteins are attractive targets of anticancer agents. Herein, we unequivocally demonstrate a distinct thiol-targeting property of gold(III) mesoporphyrin IX dimethyl ester (AuMesoIX) and its anticancer activities. While the binding of cysteine thiols with metal complexes usually occurs via M-S bond formation, AuMesoIX is unique in that the meso-carbon atom of the porphyrin ring is activated by the gold(III) ion to undergo nucleophilic aromatic substitution with thiols. AuMesoIX was shown to modify reactive cysteine residues and inhibit the activities of anticancer protein targets including thioredoxin, peroxiredoxin, and deubiquitinases. Treatment of cancer cells with AuMesoIX resulted in the formation of gold-bound sulfur-rich protein aggregates, oxidative stress-mediated cytotoxicity, and accumulation of ubiquitinated proteins. Importantly, AuMesoIX exhibited effective antitumor activity in mice. Our study has uncovered a gold(III)-induced ligand scaffold reactivity for thiol targeting that can be exploited for anticancer applications.
Collapse
|
38
|
Newman DJ. Modern traditional Chinese medicine: Identifying, defining and usage of TCM components. PHARMACOLOGICAL ADVANCES IN NATURAL PRODUCT DRUG DISCOVERY 2020; 87:113-158. [DOI: 10.1016/bs.apha.2019.07.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
39
|
Tracking the cellular targets of platinum anticancer drugs: Current tools and emergent methods. Inorganica Chim Acta 2019. [DOI: 10.1016/j.ica.2019.118984] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
40
|
Miodragović Ð, Swindell EP, Waxali ZS, Bogachkov A, O'Halloran TV. Beyond Cisplatin: Combination Therapy with Arsenic Trioxide. Inorganica Chim Acta 2019; 496:119030. [PMID: 32863421 PMCID: PMC7453736 DOI: 10.1016/j.ica.2019.119030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Platinum drugs (cisplatin, oxaliplatin, and carboplatin) and arsenic trioxide are the only commercial inorganic non-radioactive anticancer drugs approved by the US Food and Drug Administration. Numerous efforts are underway to take advantage of the synergy between the anticancer activity of cisplatin and arsenic trioxide - two drugs with strikingly different mechanisms of action. These include co-encapsulation of the two drugs in novel nanoscale delivery systems as well as the development of small molecule agents that combine the activity of these two inorganic materials. Several of these new molecular entities containing Pt-As bonds have broad anticancer activity, are robust in physiological buffer solutions, and form stable complexes with biopolymers. This review summarizes results from a number of preclinical studies involving the combination of cisplatin and As2O3, co-encapsulation and nanoformulation efforts, and the chemistry and cytotoxicity of the first member of platinum anticancer agents with an arsenous acid moiety bound to the platinum(II) center: arsenoplatins.
Collapse
Affiliation(s)
- Ðenana Miodragović
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- Northeastern Illinois University, 5500 North St Louis Avenue, Chicago, Illinois 60625, United States
| | - Elden P Swindell
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Zohra Sattar Waxali
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Abraham Bogachkov
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Thomas V O'Halloran
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
41
|
Wang FY, Liu R, Huang KB, Feng HW, Liu YN, Liang H. New platinum(II)-based DNA intercalator: Synthesis, characterization and anticancer activity. INORG CHEM COMMUN 2019. [DOI: 10.1016/j.inoche.2019.04.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|