1
|
Steel TR, Stjärnhage J, Lin Z, Bloomfield HO, Herbert CD, Astin JW, Krawczyk K, Rychlik B, Plażuk D, Jamieson SMF, Hartinger CG. Biotin functionalization of 8-hydroxyquinoline anticancer organometallics: low in vivo toxicity but potent in vitro activity. Dalton Trans 2024. [PMID: 39659246 DOI: 10.1039/d4dt02296c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
[M(arene)(HQ)Cl] complexes (M = RuII/OsII/RhIII/IrIII; HQ = 8-hydroxyquinoline) have shown promise as anticancer agents. To assess the effect of conjugating biotin (vitamin B7) to such compounds and improve their tumor-targeting ability through interaction with the sodium-dependent multivitamin transporter (SMVT), the chlorido co-ligand was exchanged with biotinylated 6-aminoindazole. The complexes were characterized by NMR spectroscopy and mass spectrometry, and purity was determined by elemental analysis. The compounds were shown to be stable in aqueous solution but reacted in particular with biologically relevant nitrogen-donor ligands. The biotinylated organometallics were shown to be able to interact with the high-affinity biotin-binding protein streptavidin using molecular modelling. High antiproliferative activity of the biotinylated Rh complex (IC50 = 1.1-10 μM) and its chlorido precursor (IC50 = 2.1-7.0 μM) was demonstrated in human HCT116, NCI-H460, COLO 205, SW620, A2780 and A2780cis cancer cells, which feature differing levels of SMVT expression. While there was no clear relationship between the anticancer activity in cells and SMVT expression, the complexes showed similar activity in cisplatin-sensitive and -resistant cells. The most potent was the biotinylated Rh derivative which displayed low toxicity toward zebrafish embryos with >75% survival up to day 4 and after treatment with up to 32 μM complex.
Collapse
Affiliation(s)
- Tasha R Steel
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Julia Stjärnhage
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Zexiong Lin
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Hugh O Bloomfield
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Caitlin D Herbert
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Jonathan W Astin
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Krzysztof Krawczyk
- Centre for Digital Biology and Biomedical Science - Biobank® Lodz, Faculty of Biology and Environmental Protection, University of Lodz, ul. Pomorska 141/143, 90-236 Łódź, Poland
| | - Błażej Rychlik
- Centre for Digital Biology and Biomedical Science - Biobank® Lodz, Faculty of Biology and Environmental Protection, University of Lodz, ul. Pomorska 141/143, 90-236 Łódź, Poland
| | - Damian Plażuk
- Laboratory of Molecular Spectroscopy, Department of Organic Chemistry, Faculty of Chemistry, University of Lodz, Tamka 12, 91-403 Łódź, Poland
| | - Stephen M F Jamieson
- Auckland Cancer Society Research Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Christian G Hartinger
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|
2
|
Swaminathan S, Haribabu J, Karvembu R. From Concept to Cure: The Road Ahead for Ruthenium-Based Anticancer Drugs. ChemMedChem 2024; 19:e202400435. [PMID: 39374112 DOI: 10.1002/cmdc.202400435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/19/2024] [Indexed: 10/09/2024]
Abstract
The evolution of chemotherapy, especially the dawn of metal-based drugs, represents a transformative era in cancer treatment. From the serendipitous discovery of mustard gas's cytotoxic effects to the sophisticated development of targeted therapies, chemotherapy has significantly refined. Central to this progression is the incorporation of metal-based compounds, such as platinum (Pt), ruthenium (Ru), and gold (Au), which offer unique mechanisms of action, distinguishing them from organic therapeutics. Among these, Ru complexes, exemplified by BOLD-100 and TLD1433, have shown exceptional promise due to their selective activity, lower propensity for resistance, and the ability to target spescific cellular pathways. This paper explores the journey of such Ru candidates, focusing on the mechanisms, efficacy, and clinical potential of these Ru-based drugs, which stand at the forefront of current research, aiming to provide more targeted, less toxic, and highly effective cancer treatments.
Collapse
Affiliation(s)
- Srividya Swaminathan
- Center for Computational Modelling, Chennai Institute of Technology, Chennai, Tamil Nadu, 600069, India
- Inorganic and Physical Chemistry Laboratory, CSIR-CLRI, Chennai, Tamil Nadu, 600020, India
| | - Jebiti Haribabu
- Faculty of Medicine, University of Atacama, Los Carreras 1579, Copiapo, 1532502, Chile
| | - Ramasamy Karvembu
- Department of Chemistry, National Institute of Technology, Tiruchirappalli, Tamil Nadu, 620015, India
| |
Collapse
|
3
|
Sumithaa C, Sugantharam K, Karanath-Anilkumar A, Munuswamy-Ramanujam G, Ganeshpandian M. RAPTA-coordinated polydiacetylene self-assembly: A chameleon-like prodrug with a dual-lock strategy for real-time release monitoring of metallodrug. Chem Commun (Camb) 2024; 60:9566-9569. [PMID: 39139058 DOI: 10.1039/d4cc03368j] [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: 08/15/2024]
Abstract
Herein, we report the first-ever design strategy of modifying RAPTA-C into a self-reporting prodrug candidate based on Ru-coordinated polydiacetylene self-assembly. This nanosystem exhibits a dual lock strategy that responds to visible light and pH-stimuli sequentially one by one with a concomitant color change for controlled RAPTA-C release and real-time release monitoring in human gastric cancer cells.
Collapse
Affiliation(s)
- Chezhiyan Sumithaa
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| | - Karnan Sugantharam
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| | - Aswathy Karanath-Anilkumar
- Molecular Biology and Immunobiology Division, Interdisciplinary Institute of Indian System of Medicine, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
- Department of Biotechnology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Ganesh Munuswamy-Ramanujam
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
- Molecular Biology and Immunobiology Division, Interdisciplinary Institute of Indian System of Medicine, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Mani Ganeshpandian
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
4
|
Roth E, Listyarini RV, Hofer TS, Cziferszky M. Host-Guest Interactions of Ruthenium(II) Arene Complexes with Cucurbit[7/8]uril. Inorg Chem 2024; 63:14021-14031. [PMID: 39016439 PMCID: PMC11289748 DOI: 10.1021/acs.inorgchem.4c01755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/18/2024]
Abstract
Cucurbit[n]urils (CB[n]s) have been recognized for their chemical and thermal stability, and their ability to bind many neutral and cationic guest molecules makes them excellent hosts in a range of supramolecular applications. In drug delivery, CB[n]s can enhance drug solubility, improve chemical and physical drug stability, and allow for triggered and controlled release. This study aimed to investigate the ability of CB[7] and CB[8] as molecular hosts to bind ruthenium(II) arene complexes that are current anticancer lead structures in the area of metallodrugs. Both, experimental and computational methods, led to insights into the binding preferences and geometries of [RuII(cym)Cl2]2 (1; cym = η6-p-cymene), [RuII(cym)(dmb)Cl2]) (2; cym = η6-p-cymene; dmb = 1,3-dimethylbenzimidazol-2-ylidene), and [RuII(cym)(pta)Cl2] (3, RAPTA-C; cym = η6-p-cymene; pta = 1,3,5-triaza-7-phospha-adamantane) with CB[7] and CB[8]. Competition experiments by mass spectrometry revealed clear preferences of 2 for CB[8] and 3 for CB[7]. Based on a comparison of the associated interaction energies from quantum chemical calculations as well as experimental data, 3@CB[7] clearly prefers a binding mode, where the pta ligand is located inside the cavity of the host, and the metal ion interacts with two of the carbonyl groups on the rim of CB[7]. In contrast, complex 2 binds in two different orientations with interaction energies similar to those of both CB[n]s, with the cym ligand being either inside or outside of the cavity. These findings suggest that ruthenium(II) arene complexes are able to form stable host-guest interactions with CB[n]s, which can be exploited as drug delivery vehicles in further metallodrug development to improve their chemical stability.
Collapse
Affiliation(s)
- Elisa Roth
- Institute
for Pharmacy, Pharmaceutical Chemistry, Department of Chemistry and
Pharmacy, University of Innsbruck, Innrain 80/82, A-6020 Innsbruck, Austria
| | - Risnita Vicky Listyarini
- Institute
of General, Inorganic and Theoretical Chemistry, Center for Chemistry
and Biomedicine, University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
- Chemistry
Education Study Program, Sanata Dharma University, Yogyakarta 55282, Indonesia
| | - Thomas S. Hofer
- Institute
of General, Inorganic and Theoretical Chemistry, Center for Chemistry
and Biomedicine, University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Monika Cziferszky
- Institute
for Pharmacy, Pharmaceutical Chemistry, Department of Chemistry and
Pharmacy, University of Innsbruck, Innrain 80/82, A-6020 Innsbruck, Austria
| |
Collapse
|
5
|
Dai G, Sun Y. Knockdown of GNL3 inhibits LUAD cell growth by regulating Wnt-β-catenin pathway. Allergol Immunopathol (Madr) 2024; 52:46-52. [PMID: 38970264 DOI: 10.15586/aei.v52i4.1117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 05/26/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a leading cause of tumor-associated mortality, and it is needed to find new target to combat this disease. Guanine nucleotide-binding -protein-like 3 (GNL3) mediates cell proliferation and apoptosis in several cancers, but its role in LUAD remains unclear. OBJECTIVE To explore the expression and function of Guanine nucleotide-binding protein-like 3 (GNL3) in lung adenocarcinoma (LUAD) and its potential mechanism in inhibiting the growth of LUAD cells. METHODS We evaluated the expression of GNL3 in LUAD tissues and its association with patient prognosis using databases and immunohistochemistry. Cell proliferation was assessed by CCK-8 assay as well as colony formation, while apoptosis was evaluated by FCM. The effect of GNL3 knockdown on the Wnt/β-catenin axis was investigated by Immunoblot analysis. RESULTS GNL3 is overexpressed in LUAD tissues and is correlated with poor prognosis. Knockdown of GNL3 significantly inhibited the growth as well as induced apoptosis in A549 as well as H1299 cells. Furthermore, we found that the inhibitory effect of GNL3 knockdown on LUAD cell growth is associated with the downregulation of the Wnt/β-catenin axis. CONCLUSION GNL3 is key in the progression of LUAD by metiating Wnt/β-catenin axis. Targeting GNL3 may represent a novel therapeutic method for LUAD treatment.
Collapse
Affiliation(s)
- Guihong Dai
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu Province China
| | - Yuejun Sun
- Department of Pathology, Affiliated Jiangyin Clinical College of Xuzhou Medical University, Jiangyin, Jiangsu Province, China;
| |
Collapse
|
6
|
Chen Y, Wang C, Qi M, Wei Y, Jiang H, Du Z. Molecular targets of cisplatin in HeLa cells explored through competitive activity-based protein profiling strategy. J Inorg Biochem 2024; 254:112518. [PMID: 38460483 DOI: 10.1016/j.jinorgbio.2024.112518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/23/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
Cisplatin is widely used as anticancer drugs, and DNA is considered as the main target. Considering its high affinity towards cysteines and the important role of cystine containing proteins, we applied a competitive activity-based protein profiling strategy to identify protein cysteines that bind with cisplatin in HeLa cells. Living cells were treated with cisplatin at cytotoxic concentrations, then the protein was extracted. After labeling with desthiobiotin iodoacetamide (DBIA) probe, protein was precipitated, digested and isotopically labeled, subsequently the peptides were combined, and the biotinylated cysteine-containing peptides were enriched and quantified by LC-MS/MS. A total of 3571 peptides which originated from 1871 proteins were identified using the DBIA probe. Among them, 46 proteins were screened as targets, including proteins that have been identified as binding proteins by previous study. A novel cisplatin target, calpain-1 (CAPN1), was identified and validated as binding with cisplatin in vitro.
Collapse
Affiliation(s)
- Yi Chen
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chenxi Wang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Meiling Qi
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yinyu Wei
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongliang Jiang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhifeng Du
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
7
|
Ramos R, Karaiskou A, Botuha C, Amhaz S, Trichet M, Dingli F, Forté J, Lam F, Canette A, Chaumeton C, Salome M, Chenuel T, Bergonzi C, Meyer P, Bohic S, Loew D, Salmain M, Sobczak-Thépot J. Identification of Cellular Protein Targets of a Half-Sandwich Iridium(III) Complex Reveals Its Dual Mechanism of Action via Both Electrophilic and Oxidative Stresses. J Med Chem 2024; 67:6189-6206. [PMID: 38577779 DOI: 10.1021/acs.jmedchem.3c02000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Identification of intracellular targets of anticancer drug candidates provides key information on their mechanism of action. Exploiting the ability of the anticancer (C∧N)-chelated half-sandwich iridium(III) complexes to covalently bind proteins, click chemistry with a bioorthogonal azido probe was used to localize a phenyloxazoline-chelated iridium complex within cells and profile its interactome at the proteome-wide scale. Proteins involved in protein folding and actin cytoskeleton regulation were identified as high-affinity targets. Upon iridium complex treatment, the folding activity of Heat Shock Protein HSP90 was inhibited in vitro and major cytoskeleton disorganization was observed. A wide array of imaging and biochemical methods validated selected targets and provided a multiscale overview of the effects of this complex on live human cells. We demonstrate that it behaves as a dual agent, inducing both electrophilic and oxidative stresses in cells that account for its cytotoxicity. The proposed methodological workflow can open innovative avenues in metallodrug discovery.
Collapse
Affiliation(s)
- Robin Ramos
- Sorbonne Université, INSERM, Centre de Recherche Saint Antoine, 184 rue du Faubourg Saint Antoine, F-75012 Paris, France
| | - Anthi Karaiskou
- Sorbonne Université, INSERM, Centre de Recherche Saint Antoine, 184 rue du Faubourg Saint Antoine, F-75012 Paris, France
| | - Candice Botuha
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, 4 place Jussieu, F-75005 Paris, France
| | - Sadek Amhaz
- Sorbonne Université, INSERM, Centre de Recherche Saint Antoine, 184 rue du Faubourg Saint Antoine, F-75012 Paris, France
| | - Michaël Trichet
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Service d'imagerie cellulaire, F-75005 Paris, France
| | - Florent Dingli
- Institut Curie, PSL Research University, CurieCoreTech Mass Spectrometry Proteomics, F-75248 Paris, France
| | - Jérémy Forté
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, 4 place Jussieu, F-75005 Paris, France
| | - France Lam
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Service d'imagerie cellulaire, F-75005 Paris, France
| | - Alexis Canette
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Service d'imagerie cellulaire, F-75005 Paris, France
| | - Chloé Chaumeton
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Service d'imagerie cellulaire, F-75005 Paris, France
| | - Murielle Salome
- ESRF, The European Synchrotron Research Facility, F-38043 Grenoble cedex 9, France
| | - Thomas Chenuel
- Sorbonne Université, PSL, CNRS, UMR8226, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, F-75005 Paris, France
| | - Céline Bergonzi
- Sorbonne Université, PSL, CNRS, UMR8226, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, F-75005 Paris, France
| | - Philippe Meyer
- Sorbonne Université, PSL, CNRS, UMR8226, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, F-75005 Paris, France
| | - Sylvain Bohic
- Université Grenoble Alpes, INSERM, UA7 STROBE, Synchrotron Radiation for Biomedicine, F-38400 Saint Martin d'Hères, France
| | - Damarys Loew
- Institut Curie, PSL Research University, CurieCoreTech Mass Spectrometry Proteomics, F-75248 Paris, France
| | - Michèle Salmain
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, 4 place Jussieu, F-75005 Paris, France
| | - Joëlle Sobczak-Thépot
- Sorbonne Université, INSERM, Centre de Recherche Saint Antoine, 184 rue du Faubourg Saint Antoine, F-75012 Paris, France
| |
Collapse
|
8
|
Coverdale JPC, Polepalli S, Arruda MAZ, da Silva ABS, Stewart AJ, Blindauer CA. Recent Advances in Metalloproteomics. Biomolecules 2024; 14:104. [PMID: 38254704 PMCID: PMC10813065 DOI: 10.3390/biom14010104] [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: 10/20/2023] [Revised: 11/17/2023] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Interactions between proteins and metal ions and their complexes are important in many areas of the life sciences, including physiology, medicine, and toxicology. Despite the involvement of essential elements in all major processes necessary for sustaining life, metalloproteomes remain ill-defined. This is not only owing to the complexity of metalloproteomes, but also to the non-covalent character of the complexes that most essential metals form, which complicates analysis. Similar issues may also be encountered for some toxic metals. The review discusses recently developed approaches and current challenges for the study of interactions involving entire (sub-)proteomes with such labile metal ions. In the second part, transition metals from the fourth and fifth periods are examined, most of which are xenobiotic and also tend to form more stable and/or inert complexes. A large research area in this respect concerns metallodrug-protein interactions. Particular attention is paid to separation approaches, as these need to be adapted to the reactivity of the metal under consideration.
Collapse
Affiliation(s)
- James P. C. Coverdale
- School of Pharmacy, Institute of Clinical Sciences, University of Birmingham, Edgbaston B15 2TT, UK;
| | | | - Marco A. Z. Arruda
- Institute of Chemistry, Department of Analytical Chemistry, Universidade Estadual de Campinas, Campinas 13083-970, Brazil; (M.A.Z.A.); (A.B.S.d.S.)
| | - Ana B. Santos da Silva
- Institute of Chemistry, Department of Analytical Chemistry, Universidade Estadual de Campinas, Campinas 13083-970, Brazil; (M.A.Z.A.); (A.B.S.d.S.)
| | - Alan J. Stewart
- School of Medicine, University of St. Andrews, St Andrews KY16 9TF, UK
| | | |
Collapse
|
9
|
Riisom M, Jamieson SMF, Hartinger CG. Critical evaluation of cell lysis methods for metallodrug studies in cancer cells. Metallomics 2023; 15:mfad048. [PMID: 37596065 DOI: 10.1093/mtomcs/mfad048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/01/2023] [Indexed: 08/20/2023]
Abstract
Intracellular accumulation studies are a key step in metallodrug development but often variable results are obtained. Therefore, we aimed here to investigate different protocols for efficient and reproducible lysis of cancer cells in terms of protein content in lysates and in cell uptake studies of the Ru anticancer complex [chlorido(8-oxyquinolinato)(η6-p-cymene)ruthenium(II)] ([Ru(cym)(HQ)Cl]). The physical lysis methods osmosis and sonication were chosen for comparison with chemical lysis with the radioimmunoprecipitation assay (RIPA) buffer. Based on the protein content and the total Ru accumulated in the lysates, the latter determined using inductively coupled plasma-mass spectrometry, RIPA buffer was the most efficient lysis method. Measurements of plastic adsorption blanks revealed that the higher Ru content determined in the RIPA buffer lysis samples may be due a higher amount of Ru extracted from the plastic incubation plates compared with osmosis and sonication. Overall, we found that the choice of lysis method needs to be matched to the information sought and we suggest the least disruptive osmosis method might be the best choice for labile drug-biomolecule adducts. Minimal differences were found for experiments aimed at measuring the overall cell uptake of the Ru complex.
Collapse
Affiliation(s)
- Mie Riisom
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Auckland Cancer Society Research Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Stephen M F Jamieson
- Auckland Cancer Society Research Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Christian G Hartinger
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
10
|
Kumar S, Riisom M, Jamieson SMF, Kavianinia I, Harris PWR, Metzler-Nolte N, Brimble MA, Hartinger CG. On-Resin Conjugation of the Ruthenium Anticancer Agent Plecstatin-1 to Peptide Vectors. Inorg Chem 2023; 62:14310-14317. [PMID: 37611203 DOI: 10.1021/acs.inorgchem.3c01718] [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: 08/25/2023]
Abstract
Ruthenium piano-stool complexes have been explored for their anticancer activity and some promising compounds have been reported. Herein, we conjugated a derivative of plecstatin-1 to peptides in order to increase their cancer cell targeting ability. For this purpose, plecstatin-1 was modified at the arene ligand to introduce a functional amine handle (3), which resulted in a compound that showed similar activity in an in vitro anticancer activity assay. The cell-penetrating peptide TAT48-60, tumor-targeting neurotensin8-13, and plectin-targeting peptide were functionalized with succinyl or β-Ala-succinyl linkers under standard solid-phase peptide synthesis (SPPS) conditions to spatially separate the peptide backbones from the bioactive metal complexes. These modifications allowed for conjugating precursor 3 to the peptides on resin yielding the desired metal-peptide conjugates (MPCs), as confirmed by high-performance liquid chromatography (HPLC), NMR spectroscopy, and mass spectrometry (MS). The MPCs were studied for their behavior in aqueous solution and under acidic conditions and resembled that of the parent compound plecstatin-1. In in vitro anticancer activity studies in a small panel of cancer cell lines, the TAT-based MPCs showed the highest activity, while the other MPCs were virtually inactive. However, the MPCs were significantly less active than the small molecules plecstatin-1 and 3, which can be explained by the reduced cell uptake as determined by inductively coupled plasma MS (ICP-MS). Although the MPCs did not display potent anticancer activities, the developed conjugation strategy can be extended toward other metal complexes, which may be able to utilize the targeting properties of peptides.
Collapse
Affiliation(s)
- Saawan Kumar
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Mie Riisom
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Auckland Cancer Society Research Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Stephen M F Jamieson
- Auckland Cancer Society Research Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Iman Kavianinia
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Paul W R Harris
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Nils Metzler-Nolte
- Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Bochum 44801, Germany
| | - Margaret A Brimble
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Christian G Hartinger
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
11
|
Iacobucci I, La Manna S, Cipollone I, Monaco V, Canè L, Cozzolino F. From the Discovery of Targets to Delivery Systems: How to Decipher and Improve the Metallodrugs' Actions at a Molecular Level. Pharmaceutics 2023; 15:1997. [PMID: 37514183 PMCID: PMC10385150 DOI: 10.3390/pharmaceutics15071997] [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: 06/05/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Metals are indispensable for the life of all organisms, and their dysregulation leads to various disorders due to the disruption of their homeostasis. Nowadays, various transition metals are used in pharmaceutical products as diagnostic and therapeutic agents because their electronic structure allows them to adjust the properties of molecules differently from organic molecules. Therefore, interest in the study of metal-drug complexes from different aspects has been aroused, and numerous approaches have been developed to characterize, activate, deliver, and clarify molecular mechanisms. The integration of these different approaches, ranging from chemoproteomics to nanoparticle systems and various activation strategies, enables the understanding of the cellular responses to metal drugs, which may form the basis for the development of new drugs and/or the modification of currently used drugs. The purpose of this review is to briefly summarize the recent advances in this field by describing the technological platforms and their potential applications for identifying protein targets for discovering the mechanisms of action of metallodrugs and improving their efficiency during delivery.
Collapse
Affiliation(s)
- Ilaria Iacobucci
- UMR7042 CNRS-Unistra-UHA, Laboratoire d'Innovation Moléculaire et Applications (LIMA), European School of Chemistry, Polymers and Materials (ECPM), 67087 Strasbourg, France
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Sara La Manna
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Irene Cipollone
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
- CEINGE Biotecnologie Avanzate "Franco Salvatore" S.c.a r.l., 80131 Naples, Italy
| | - Vittoria Monaco
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
- CEINGE Biotecnologie Avanzate "Franco Salvatore" S.c.a r.l., 80131 Naples, Italy
| | - Luisa Canè
- CEINGE Biotecnologie Avanzate "Franco Salvatore" S.c.a r.l., 80131 Naples, Italy
- Department of Translational Medical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Flora Cozzolino
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
- CEINGE Biotecnologie Avanzate "Franco Salvatore" S.c.a r.l., 80131 Naples, Italy
| |
Collapse
|
12
|
de Araujo-Neto JH, Guedes APM, Leite CM, Moraes CAF, Santos AL, Brito RDS, Rocha TL, Mello-Andrade F, Ellena J, Batista AA. "Half-Sandwich" Ruthenium Complexes with Alizarin as Anticancer Agents: In Vitro and In Vivo Studies. Inorg Chem 2023; 62:6955-6969. [PMID: 37099760 DOI: 10.1021/acs.inorgchem.3c00183] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Upon exploration of the chemistry of the combination of ruthenium/arene with anthraquinone alizarin (L), three new complexes with the general formulas [Ru(L)Cl(η6-p-cymene)] (C1), [Ru(L)(η6-p-cymene)(PPh3)]PF6 (C2), and [Ru(L)(η6-p-cymene)(PEt3)]PF6 (C3) were synthesized and characterized using spectroscopic techniques (mass, IR, and 1D and 2D NMR), molar conductivity, elemental analysis, and X-ray diffraction. Complex C1 exhibited fluorescence, such as free alizarin, while in C2 and C3, the emission was probably quenched by monophosphines and the crystallographic data showed that hydrophobic interactions are predominant in intermolecular contacts. The cytotoxicity of the complexes was evaluated in the MDA-MB-231 (triple-negative breast cancer), MCF-7 (breast cancer), and A549 (lung) tumor cell lines and MCF-10A (breast) and MRC-5 (lung) nontumor cell lines. Complexes C1 and C2 were more selective to the breast tumor cell lines, and C2 was the most cytotoxic (IC50 = 6.5 μM for MDA-MB-231). In addition, compound C1 performs a covalent interaction with DNA, while C2 and C3 present only weak interactions; however, internalization studies by flow cytometry and confocal microscopy showed that complex C1 does not accumulate in viable MDA-MB-231 cells and is detected in the cytoplasm only after cell permeabilization. Investigations of the mechanism of action of the complexes indicate that C2 promotes cell cycle arrest in the Sub-G1 phase in MDA-MB-231, inhibits its colony formation, and has a possible antimetastatic action, impeding cell migration in the wound-healing experiment (13% of wound healing in 24 h). The in vivo toxicological experiments with zebrafish indicate that C1 and C3 exhibit the most zebrafish embryo developmental toxicity (inhibition of spontaneous movements and heartbeats), while C2, the most promising anticancer drug in the in vitro preclinical tests, revealed the lowest toxicity in in vivo preclinical screening.
Collapse
Affiliation(s)
- João Honorato de Araujo-Neto
- Instituto de Física de São Carlos, Universidade de São Paulo (USP), São Carlos, São Paulo 13566-590, Brazil
- Departamento de Química, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | - Adriana P M Guedes
- Departamento de Química, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | - Celisnolia M Leite
- Instituto de Física de São Carlos, Universidade de São Paulo (USP), São Carlos, São Paulo 13566-590, Brazil
| | - Carlos André F Moraes
- Departamento de Química, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | - Andressa L Santos
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás 74605-050, Brazil
| | - Rafaella da S Brito
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás 74605-050, Brazil
| | - Thiago L Rocha
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás 74605-050, Brazil
| | - Francyelli Mello-Andrade
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás 74605-050, Brazil
- Instituto Federal de Educação Ciência e Tecnologia (IFG), Goiânia, Goiás 74055-110, Brazil
| | - Javier Ellena
- Instituto de Física de São Carlos, Universidade de São Paulo (USP), São Carlos, São Paulo 13566-590, Brazil
| | - Alzir A Batista
- Departamento de Química, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| |
Collapse
|
13
|
Skos L, Borutzki Y, Gerner C, Meier-Menches SM. Methods to identify protein targets of metal-based drugs. Curr Opin Chem Biol 2023; 73:102257. [PMID: 36599256 DOI: 10.1016/j.cbpa.2022.102257] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/02/2022] [Accepted: 12/04/2022] [Indexed: 01/03/2023]
Abstract
Metal-based anticancer agents occupy a distinct chemical space due to their particular coordination geometry and reactivity. Despite the initial DNA-targeting paradigm for this class of compounds, it is now clear that they can also be tuned to target proteins in cells, depending on the metal and ligand scaffold. Since metallodrug discovery is dominated by phenotypic screenings, tailored proteomics strategies were crucial to identify and validate protein targets of several investigative and clinically advanced metal-based drugs. Here, such experimental approaches are discussed, which showed that metallodrugs based on ruthenium, gold, rhenium and even platinum, can selectively and specifically target proteins with clear-cut down-stream effects. Target identification strategies are expected to support significantly the mechanism-driven clinical translation of metal-based drugs.
Collapse
Affiliation(s)
- Lukas Skos
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; Doctoral School of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Yasmin Borutzki
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; Doctoral School of Chemistry, University of Vienna, 1090 Vienna, Austria; Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; Joint Metabolome Facility, University of Vienna and Medical University Vienna, 1090 Vienna, Austria
| | - Samuel M Meier-Menches
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; Joint Metabolome Facility, University of Vienna and Medical University Vienna, 1090 Vienna, Austria.
| |
Collapse
|
14
|
Chrzanowska M, Katafias A, van Eldik R. Reactivity of non-organometallic ruthenium(II) polypyridyl complexes and their application as catalysts for hydride transfer reactions. Front Chem 2023; 11:1150164. [PMID: 37007058 PMCID: PMC10050333 DOI: 10.3389/fchem.2023.1150164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/21/2023] [Indexed: 03/17/2023] Open
Abstract
Recently, we investigated the substitution behavior of a series of ruthenium(II) complexes of the general formula [RuII(terpy)(N∧N)Cl]Cl, where terpy = 2,2′:6′,2″-terpyridine, N∧N = bidentate ligand, in aqueous solutions. We have shown that the most and least reactive complexes of the series are [RuII(terpy)(en)Cl]Cl (en = ethylenediamine) and [RuII(terpy)(phen)Cl]Cl (phen = 1, 10-phenantroline), respectively, as a result of different electronic effects provided by the bidentate spectator chelates. Polypyridyl amine Ru(II) complex, viz. [Ru(terpy)(en)Cl]Cl and [Ru(terpy)(ampy)Cl]Cl (where ampy = 2-(aminomethyl)pyridine), in which the terpy chelate labilizes the metal center, are able to catalyze the conversion of NAD+ to 1,4-NADH using sodium formate as a source of hydride. We showed that this complex can control the [NAD+]/[NADH] ratio and potentially induce reductive stress in living cells, which is accepted as an effective method to kill cancer cells. Polypyridyl Ru(II) complexes, characterized in terms of the behavior in aqueous solutions, can be used as model systems to monitor heterogeneous multiphase ligand substitution reactions at the solid-liquid interface. Colloidal coordination compounds in the submicron range were synthesized from Ru(II)-aqua derivatives of starting chlorido complexes via the anti-solvent procedure and stabilized by a surfactant shell layer.
Collapse
Affiliation(s)
- Marta Chrzanowska
- Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Anna Katafias
- Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Rudi van Eldik
- Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Toruń, Poland
- Department of Chemistry and Pharmacy, University of Erlangen-Nuremberg, Erlangen, Germany
- *Correspondence: Rudi van Eldik,
| |
Collapse
|
15
|
Mir MA, Qayoom H, Sofi S, Jan N. Proteomics: A groundbreaking development in cancer biology. Proteomics 2023. [DOI: 10.1016/b978-0-323-95072-5.00004-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
16
|
Yan X, Zhou Y, Li H, Jiang G, Sun H. Metallomics and metalloproteomics. COMPREHENSIVE INORGANIC CHEMISTRY III 2023:53-76. [DOI: 10.1016/b978-0-12-823144-9.00060-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
17
|
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]
|
18
|
Mansouri F, Ortiz D, Dyson PJ. Competitive binding studies of the nucleosomal histone targeting drug, [Ru(η 6-p-cymene)Cl 2(pta)] (RAPTA-C), with oligonucleotide-peptide mixtures. J Inorg Biochem 2023; 238:112043. [PMID: 36370502 DOI: 10.1016/j.jinorgbio.2022.112043] [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: 08/13/2022] [Revised: 10/11/2022] [Accepted: 10/19/2022] [Indexed: 11/07/2022]
Abstract
Protein crystallography and biochemical assays reveal that the organometallic drug, [Ru(η6-p-cymene)Cl2(pta)] (RAPTA-C), preferentially binds to nucleosomal histone proteins in chromatin. To better understand the binding mechanism we report here a mass spectrometric-based competitive binding study between a model peptide from the acidic patch region of the H2A histone protein (the region where RAPTA-C is known to bind) and an oligonucleotide. In contrast to the protein crystallography and biochemical assays, RAPTA-C preferentially binds to the oligonucleotide, confirming that steric factors, rather than electronic effects, primarily dictate binding of RAPTA-C to histone proteins within the nucleosome.
Collapse
Affiliation(s)
- Farangis Mansouri
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne CH-1015, Switzerland; Department of Chemistry Institute for Advanced Studies in Basic Sciences (IASBS), 444 Prof. Sobouti Blvd., Gava Zang, Zanjan 45137-66731, Iran
| | - Daniel Ortiz
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne CH-1015, Switzerland
| | - Paul J Dyson
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne CH-1015, Switzerland.
| |
Collapse
|
19
|
Kljun J, Pavlič R, Hafner E, Lipec T, Moreno-Da Silva S, Tič P, Turel I, Büdefeld T, Stojan J, Rižner TL. Ruthenium complexes show potent inhibition of AKR1C1, AKR1C2, and AKR1C3 enzymes and anti-proliferative action against chemoresistant ovarian cancer cell line. Front Pharmacol 2022; 13:920379. [PMID: 36034868 PMCID: PMC9403717 DOI: 10.3389/fphar.2022.920379] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/28/2022] [Indexed: 12/04/2022] Open
Abstract
In this study, we present the synthesis, kinetic studies of inhibitory activity toward aldo-keto reductase 1C (AKR1C) enzymes, and anticancer potential toward chemoresistant ovarian cancer of 10 organoruthenium compounds bearing diketonate (1–6) and hydroxyquinolinate (7–10) chelating ligands with the general formula [(η6-p-cymene)Ru(chel)(X)]n+ where chel represents the chelating ligand and X the chlorido or pta ligand. Our studies show that these compounds are potent inhibitors of the AKR enzymes with an uncommon inhibitory mechanism, where two inhibitor molecules bind to the enzyme in a first fast and reversible step and a second slower and irreversible step. The binding potency of each step is dependent on the chemical structure of the monodentate ligands in the metalloinhibitors with the chlorido complexes generally acting as reversible inhibitors and pta complexes as irreversible inhibitors. Our study also shows that compounds 1–9 have a moderate yet better anti-proliferative and anti-migration action on the chemoresistant ovarian cancer cell line COV362 compared to carboplatin and similar effects to cisplatin.
Collapse
Affiliation(s)
- Jakob Kljun
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Renata Pavlič
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Eva Hafner
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tanja Lipec
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Sara Moreno-Da Silva
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
- Faculty of Chemical Sciences, Universidad Complutense de Madrid, Madrid, Spain
| | - Primož Tič
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Iztok Turel
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
- *Correspondence: Iztok Turel, ; Tea Lanišnik Rižner,
| | - Tomaž Büdefeld
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jure Stojan
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tea Lanišnik Rižner
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- *Correspondence: Iztok Turel, ; Tea Lanišnik Rižner,
| |
Collapse
|
20
|
Milutinović MM, Caković AZ, Ćoćić D, Rais E, Schoch R, Marković BS, Arsenijević N, Volarević V, Jovanović-Stević S, Bogojeski JV, Wilhelm R. Unique enantiopure camphor-based neutral arene–ruthenium(II) complexes: DNA/BSA binding, kinetic and cytotoxic studies. J COORD CHEM 2022. [DOI: 10.1080/00958972.2022.2106562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
| | | | - Dušan Ćoćić
- Faculty of Science, University of Kragujevac, Kragujevac, Serbia
| | - Eduard Rais
- Department of Chemistry, University of Paderborn, Paderborn, Germany
| | - Roland Schoch
- Department of Chemistry, University of Paderborn, Paderborn, Germany
| | - Bojana Simović Marković
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nebojša Arsenijević
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Vladislav Volarević
- Department of Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | | | - René Wilhelm
- Institute of Organic Chemistry, Clausthal University of Technology, Clausthal-Zellerfeld, Germany
| |
Collapse
|
21
|
Riisom M, Eade L, Tremlett WDJ, Hartinger CG. The aqueous stability and interactions of organoruthenium compounds with serum proteins, cell culture medium, and human serum. Metallomics 2022; 14:mfac043. [PMID: 35751650 PMCID: PMC9314723 DOI: 10.1093/mtomcs/mfac043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/26/2022] [Indexed: 11/12/2022]
Abstract
Metal complexes bind to a wide variety of biomolecules and the control of the reactivity is essential when designing anticancer metallodrugs with a specific mode of action in mind. In this study, we used the highly cytotoxic compound [RuII(cym)(8-HQ)Cl] (cym = η6-p-cymene, 8-HQ = 8-hydroxyquinoline), the more inert derivative [RuII(cym)(8-HQ)(PTA)](SO3CF3) (PTA = 1,3,5-triaza-7-phosphaadamantane), and [RuII(cym)(PCA)Cl]Cl (PCA = pyridinecarbothioamide) as a complex with a different coordination environment about the Ru center and investigated their stability, interactions with proteins, and behavior in medium (αMEM) and human serum by capillary zone electrophoresis. The developed method was found to be robust and provides a quick and low-cost technique to monitor the interactions of such complexes with biomolecules. Each complex was found to behave very differently, emphasizing the importance of the choice of ligands and demonstrating the applicability of the developed method. Additionally, the human serum albumin binding site preference of [RuII(cym)(8-HQ)Cl] was investigated through displacement studies, revealing that the compound was able to bind to both sites I and site II, and the type of adducts formed with transferrin was determined by mass spectrometry.
Collapse
Affiliation(s)
- Mie Riisom
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| | - Liam Eade
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| | - William D J Tremlett
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| | - Christian G Hartinger
- School of Chemical Sciences, University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| |
Collapse
|
22
|
Chiaverini L, Cirri D, Tolbatov I, Corsi F, Piano I, Marrone A, Pratesi A, Marzo T, La Mendola D. Medicinal Hypervalent Tellurium Prodrugs Bearing Different Ligands: A Comparative Study of the Chemical Profiles of AS101 and Its Halido Replaced Analogues. Int J Mol Sci 2022; 23:ijms23147505. [PMID: 35886853 PMCID: PMC9317073 DOI: 10.3390/ijms23147505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 02/01/2023] Open
Abstract
Ammonium trichloro (dioxoethylene-O,O′) tellurate (AS101) is a potent immunomodulator prodrug that, in recent years, entered various clinical trials and was tested for a variety of potential therapeutic applications. It has been demonstrated that AS101 quickly activates in aqueous milieu, producing TeOCl3−, which likely represents the pharmacologically active species. Here we report on the study of the activation process of AS101 and of two its analogues. After the synthesis and characterization of AS101 and its derivatives, we have carried out a comparative study through a combined experimental and computational analysis. Based on the obtained results, we describe here, for the first time, the detailed reaction that AS101 and its bromido- and iodido-replaced analogues undergo in presence of water, allowing the conversion of the original molecule to the likely true pharmacophore. Interestingly, moving down in the halogens’ group we observed a higher tendency to react, attributable to the ligands’ effect. The chemical and mechanistic implications of these meaningful differences are discussed.
Collapse
Affiliation(s)
- Lorenzo Chiaverini
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, 56126 Pisa, Italy; (L.C.); (F.C.); (I.P.); (D.L.M.)
| | - Damiano Cirri
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi, 13, 56124 Pisa, Italy; (D.C.); (A.P.)
| | - Iogann Tolbatov
- Institute of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology, 43007 Tarragona, Spain
- Correspondence: (I.T.); (T.M.)
| | - Francesca Corsi
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, 56126 Pisa, Italy; (L.C.); (F.C.); (I.P.); (D.L.M.)
| | - Ilaria Piano
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, 56126 Pisa, Italy; (L.C.); (F.C.); (I.P.); (D.L.M.)
| | - Alessandro Marrone
- Dipartimento di Farmacia, Università degli Studi “G. D’Annunzio” Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy;
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi, 13, 56124 Pisa, Italy; (D.C.); (A.P.)
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, 56126 Pisa, Italy; (L.C.); (F.C.); (I.P.); (D.L.M.)
- Correspondence: (I.T.); (T.M.)
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, 56126 Pisa, Italy; (L.C.); (F.C.); (I.P.); (D.L.M.)
| |
Collapse
|
23
|
Swaminathan S, Haribabu J, Balakrishnan N, Vasanthakumar P, Karvembu R. Piano stool Ru(II)-arene complexes having three monodentate legs: A comprehensive review on their development as anticancer therapeutics over the past decade. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214403] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
24
|
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
|
25
|
Abstract
Metals are essential components in life processes and participate in many important biological processes. Dysregulation of metal homeostasis is correlated with many diseases. Metals are also frequently incorporated into diagnosis and therapeutics. Understanding of metal homeostasis under (patho)physiological conditions and the molecular mechanisms of action of metallodrugs in biological systems has positive impacts on human health. As an emerging interdisciplinary area of research, metalloproteomics involves investigating metal-protein interactions in biological systems at a proteome-wide scale, has received growing attention, and has been implemented into metal-related research. In this review, we summarize the recent advances in metalloproteomics methodologies and applications. We also highlight emerging single-cell metalloproteomics, including time-resolved inductively coupled plasma mass spectrometry, mass cytometry, and secondary ion mass spectrometry. Finally, we discuss future perspectives in metalloproteomics, aiming to attract more original research to develop more advanced methodologies, which could be utilized rapidly by biochemists or biologists to expand our knowledge of how metal functions in biology and medicine. Expected final online publication date for the Annual Review of Biochemistry, Volume 91 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, University of Hong Kong, Hong Kong SAR, China; ,
| | - Hongyan Li
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, University of Hong Kong, Hong Kong SAR, China; ,
| | - Hongzhe Sun
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, University of Hong Kong, Hong Kong SAR, China; ,
| |
Collapse
|
26
|
|
27
|
Mihajlović-Lalić LE, Poljarević J, Grgurić-Šipka S. Metal complexes with α-picolinic acid frameworks and their antitumor activity. Inorganica Chim Acta 2021. [DOI: 10.1016/j.ica.2021.120582] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
28
|
Current and emerging mass spectrometry methods for the preclinical development of metal-based drugs: a critical appraisal. Anal Bioanal Chem 2021; 414:95-102. [PMID: 34642780 DOI: 10.1007/s00216-021-03718-5] [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: 08/18/2021] [Revised: 09/23/2021] [Accepted: 10/04/2021] [Indexed: 10/20/2022]
Abstract
This Trends article highlights the multiple ways in which the state-of-the-art molecular mass spectrometry can support the preclinical development of novel metal-based anticancer drugs. Examples from the recent literature-beyond routine characterization applications-are presented to illustrate what analytical and experimental design challenges are to be addressed to facilitate the translation of promising drug candidates to clinical practice.
Collapse
|
29
|
Design concepts of half-sandwich organoruthenium anticancer agents based on bidentate bioactive ligands. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213950] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
30
|
Trivedi M, Sharma P, Pandey IK, Kumar A, Kumar S, Rath NP. Acid-assisted hydrogenation of CO 2 to methanol using Ru(II) and Rh(III) RAPTA-type catalysts under mild conditions. Chem Commun (Camb) 2021; 57:8941-8944. [PMID: 34397067 DOI: 10.1039/d1cc03049c] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A highly efficient homogeneous catalyst system for production of CH3OH from CO2 using single molecular defined ruthenium and rhodium RAPTA-type catalysts [Ru(η6-p-cymene)X2(PTA)] (X = I(1), Cl(2); PTA = 1,3,5-triaza-7-phosphaadamantane) and rhodium catalysts [Rh(η5-C5Me5)X2(PTA/PTA-BH3)] (X = Cl(3), H(4) and PTA-BH3, H(5)) developed in acidic media under mild conditions. A TON of 4752 is achieved using a [Ru(η6-p-cymene)I2(PTA)] catalyst which represents the first example of CO2 hydrogenation to CH3OH using single molecular defined Ru and Rh RAPTA-type catalysts.
Collapse
Affiliation(s)
- Manoj Trivedi
- Department of Chemistry, University of Delhi, Delhi-110007, India. and Department of Chemistry, Sri Venkateswara College, University of Delhi, New Delhi-110021, India
| | - Pooja Sharma
- Department of Chemistry, Dhirendra Mahila PG College, Varanasi-221005, India
| | | | - Abhinav Kumar
- Department of Chemistry, University of Lucknow, Lucknow-226007, India
| | - Sanjay Kumar
- Department of Chemistry, Sri Venkateswara College, University of Delhi, New Delhi-110021, India
| | - Nigam P Rath
- Department of Chemistry & Biochemistry and Centre for Nanoscience, University of Missouri-St. Louis, One University Boulevard, St. Louis, MO 63121-4499, USA.
| |
Collapse
|
31
|
Drug Repurposing to Identify a Synergistic High-Order Drug Combination to Treat Sunitinib-Resistant Renal Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13163978. [PMID: 34439134 PMCID: PMC8391235 DOI: 10.3390/cancers13163978] [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: 07/05/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary In this study, drug combination screening was used to design a multidrug combination consisting of repurposed drugs to treat sunitinib-resistant clear cell renal cell carcinoma. In the frame of this project, the multidrug combination has been optimized and validated and an insight into the mechanism of action is given. The multidrug combinations significantly altered the transcription of genes related to apoptosis and metabolic pathways. Further analysis of the metabolism revealed strong upregulation of the presence of sphingolipids after multidrug combination treatment. Final evaluation for translation of the multidrug combination in ex vivo organoid-like cultures demonstrated significant anti-cancer efficacy. Abstract Repurposed drugs have been evaluated for the management of clear cell renal cell carcinoma (ccRCC), but only a few have influenced the overall survival of patients with advanced disease. To combine repurposed non-oncology with oncological drugs, we applied our validated phenotypic method, which consisted of a reduced experimental part and data modeling. A synergistic optimized multidrug combination (ODC) was identified to significantly reduce the energy levels in cancer remaining inactive in non-cancerous cells. The ODC consisted of Rapta-C, erlotinib, metformin and parthenolide and low doses. Molecular and functional analysis of ODC revealed a loss of adhesiveness and induction of apoptosis. Gene-expression network analysis displayed significant alterations in the cellular metabolism, confirmed by LC-MS based metabolomic analysis, highlighting significant changes in the lipid classes. We used heterotypic in vitro 3D co-cultures and ex vivo organoids to validate the activity of the ODC, maintaining an efficacy of over 70%. Our results show that repurposed drugs can be combined to target cancer cells selectively with prominent activity. The strong impact on cell adherence and metabolism indicates a favorable mechanism of action of the ODC to treat ccRCC.
Collapse
|
32
|
Klaimanee E, Nhukeaw T, Saithong S, Ratanaphan A, Phongpaichit S, Tantirungrotechai Y, Leesakul N. Half-sandwich ruthenium (II) p-cymene complexes based on organophosphorus ligands: Structure determination, computational investigation, in vitro antiproliferative effect in breast cancer cells and antimicrobial activity. Polyhedron 2021. [DOI: 10.1016/j.poly.2021.115244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
33
|
Steel TR, Walsh F, Wieczorek-Błauż A, Hanif M, Hartinger CG. Monodentately-coordinated bioactive moieties in multimodal half-sandwich organoruthenium anticancer agents. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213890] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
34
|
Rafols L, Josa D, Aguilà D, Barrios LA, Roubeau O, Cirera J, Soto-Cerrato V, Pérez-Tomás R, Martínez M, Grabulosa A, Gamez P. Piano-Stool Ruthenium(II) Complexes with Delayed Cytotoxic Activity: Origin of the Lag Time. Inorg Chem 2021; 60:7974-7990. [PMID: 33979132 PMCID: PMC8659375 DOI: 10.1021/acs.inorgchem.1c00507] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
![]()
We have recently reported a series
of piano-stool ruthenium(II)
complexes of the general formula [RuCl2(η6-arene)(P(1-pyrenyl)R2R3)] showing excellent
cytotoxic activities (particularly when R2 = R3 = methyl). In the present study, new members of this family of compounds
have been prepared with the objective to investigate the effect of
the steric hindrance of a bulky phosphane ligand, namely diisopropyl(1-pyrenyl)phosphane
(L), on exchange reactions involving the coordinated
halides (X = Cl, I). Two η6-arene rings were used,
i.e. η6-methyl benzoate (mba) and η6-p-cymene (p-cym), and four complexes
were synthesized, namely [RuCl2(mba)(L)] (1Cl2iPr), [RuI2(mba)(L)] (1I2iPr), [RuCl2(p-cym)(L)] (2Cl2iPr), and [RuI2(p-cym)(L)]
(2I2iPr). Unexpectedly, all of
the complexes exhibited poor cytotoxic activities after 24 h of incubation
with cells, in contrast to the related compounds previously reported.
However, it was observed that aged DMSO solutions of 2I2iPr (from 2 to 7 days) exhibited better activities
in comparison to freshly prepared solutions and that the activity
improved over “aging” time. Thorough studies were therefore
performed to uncover the origin of this lag time in the cytotoxicity
efficiency. The data achieved clearly demonstrated that compounds 2I2iPr and 2Cl2iPr were undergoing a series of transformation reactions in DMSO (with
higher rates for the iodido complex 2I2iPr), ultimately generating cyclometalated species through a mechanism
involving DMSO as a coordinated proton abstractor. The cyclometalated
complexes detected in solution were subsequently prepared; hence,
pure [RuCl(p-cym)(κ2C-diisopropyl(1-pyrenyl)phosphane)] (3CliPr), [RuI(p-cym)(κ2C-diisopropyl(1-pyrenyl)phosphane)]
(3IiPr), and [Ru(p-cym)(κS-dmso)(κ2C-diisopropyl(1-pyrenyl)phosphane)]PF6 (3dmsoiPr) were synthesized and fully
characterized. Remarkably, 3CliPr, 3IiPr, and 3dmsoiPr are all very efficient cytotoxic agents,
exhibiting slightly better activities in comparison to the chlorido
noncyclometalated complexes [RuCl2(η6-arene)(P(1-pyrenyl)R2R3)] described in an earlier report. For comparison
purposes, the iodido compounds [RuI2(mba)(dimethyl(1-pyrenyl)phosphane)]
(1I2Me) and [RuI2(p-cym)(dimethyl(1-pyrenyl)phosphane)] (2I2Me), bearing the less hindered dimethyl(1-pyrenyl)phosphane ligand,
have also been prepared. The cytotoxic and chemical behaviors of 1I2Me and 1I2Me were comparable to those of their chlorido counterparts reported
previously. DMSO gradually converts half-sandwich,
1-pyrenyl-containing
ruthenium(II) complexes into cyclometalated species showing notable
cytotoxic properties.
Collapse
Affiliation(s)
- Laia Rafols
- Departament de Química Inorgànica i Orgànica, Facultat de Química, Secció de Química Inorgànica, Universitat de Barcelona, Martí i Franquès, 1-11, 08028 Barcelona, Spain
| | - Dana Josa
- Departament de Química Inorgànica i Orgànica, Facultat de Química, Secció de Química Inorgànica, Universitat de Barcelona, Martí i Franquès, 1-11, 08028 Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - David Aguilà
- Departament de Química Inorgànica i Orgànica, Facultat de Química, Secció de Química Inorgànica, Universitat de Barcelona, Martí i Franquès, 1-11, 08028 Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Leoní A Barrios
- Departament de Química Inorgànica i Orgànica, Facultat de Química, Secció de Química Inorgànica, Universitat de Barcelona, Martí i Franquès, 1-11, 08028 Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Olivier Roubeau
- Instituto de Ciencia de Materiales de Aragón (ICMA), CSIC and Universidad de Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Jordi Cirera
- Departament de Química Inorgànica i Orgànica, Facultat de Química, Secció de Química Inorgànica, Universitat de Barcelona, Martí i Franquès, 1-11, 08028 Barcelona, Spain.,Institut de Recerca de Química Teórica i Computacional, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Vanessa Soto-Cerrato
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, University of Barcelona, Campus Bellvitge, Feixa Llarga s/n, 08907 L'Hospitalet de Llobregat (Barcelona), Spain.,Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ricardo Pérez-Tomás
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, University of Barcelona, Campus Bellvitge, Feixa Llarga s/n, 08907 L'Hospitalet de Llobregat (Barcelona), Spain.,Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), 08907 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Manuel Martínez
- Departament de Química Inorgànica i Orgànica, Facultat de Química, Secció de Química Inorgànica, Universitat de Barcelona, Martí i Franquès, 1-11, 08028 Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Arnald Grabulosa
- Departament de Química Inorgànica i Orgànica, Facultat de Química, Secció de Química Inorgànica, Universitat de Barcelona, Martí i Franquès, 1-11, 08028 Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Patrick Gamez
- Departament de Química Inorgànica i Orgànica, Facultat de Química, Secció de Química Inorgànica, Universitat de Barcelona, Martí i Franquès, 1-11, 08028 Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, 08028 Barcelona, Spain.,Catalan Institution for Research and Advanced Studies, Passeig Lluís Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
35
|
Neuditschko B, Legin AA, Baier D, Schintlmeister A, Reipert S, Wagner M, Keppler BK, Berger W, Meier‐Menches SM, Gerner C. Die Wechselwirkung mit ribosomalen Proteinen begleitet die Stressinduktion des Wirkstoffkandidaten BOLD-100/KP1339 im endoplasmatischen Retikulum. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 133:5121-5126. [PMID: 38505777 PMCID: PMC10947255 DOI: 10.1002/ange.202015962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Indexed: 11/09/2022]
Abstract
AbstractDer metallhaltige Wirkstoff BOLD‐100/KP1339 zeigte bereits vielversprechende Resultate in verschiedenen In vitro‐ und In vivo‐Tumormodellen sowie in klinischen Studien. Der detaillierte Wirkmechanismus wurde jedoch noch nicht komplett aufgeklärt. Als entscheidende Wirkstoffeffekte kristallisierten sich kürzlich die Stressinduktion im endoplasmatischen Retikulum (ER) und die damit einhergehende Modulierung von HSPA5 (GRP78) heraus. Das spontane und stabile Addukt zwischen BOLD‐100 und menschlichem Serumalbumin wurde als Immobilisierungsstrategie ausgewählt, um einen chemoproteomischen Ansatz auszuführen, der die ribosomalen Proteine RPL10, RPL24 und den Transkriptionsfaktor GTF2I als potentielle Interaktoren dieser Ru(III)‐Verbindung identifizierten. Dieses Ergebnis wurde mit proteomischen und transkriptomischen Profiling‐Experimenten kombiniert, was die Interpretation einer ribosomalen Beeinträchtigung sowie der Induktion von ER‐Stress unterstützte. Die Bildung von Polyribosomen und begleitende ER‐Schwellungen in behandelten Krebszellen wurden zudem durch TEM‐Messungen bestätigt. Somit scheint eine direkte Wechselwirkung von BOLD‐100 mit ribosomalen Proteinen die ER‐Stressinduktion und die Modulierung von GRP78 in Krebszellen zu begleiten.
Collapse
Affiliation(s)
- Benjamin Neuditschko
- Institut für Anorganische ChemieFakultät für ChemieUniversität WienWähringer Str. 421090WienÖsterreich
- Institut für Analytische ChemieFakultät für ChemieUniversität WienWähringer Str. 381090WienÖsterreich
| | - Anton A. Legin
- Institut für Anorganische ChemieFakultät für ChemieUniversität WienWähringer Str. 421090WienÖsterreich
- Forschungsnetzwerk “Chemistry, Microbiology and Environmental Systems Science”Universität WienWähringer Str. 421090WienÖsterreich
| | - Dina Baier
- Institut für Anorganische ChemieFakultät für ChemieUniversität WienWähringer Str. 421090WienÖsterreich
- Institut für Krebsforschung und Comprehensive Cancer CenterUniversitätsklinik für Innere Medizin IMedizinische Universität WienBorschkegasse 8a1090WienÖsterreich
- Forschungscluster “Translational Cancer Therapy Research”Universität WienWähringer Str. 421090WienÖsterreich
| | - Arno Schintlmeister
- Forschungsnetzwerk “Chemistry, Microbiology and Environmental Systems Science”Universität WienWähringer Str. 421090WienÖsterreich
- Großgeräteeinrichtung für Umwelt- und Isotopen-MassenspektrometrieZentrum für Mikrobiologie und UmweltsystemwissenschaftUniversität WienAlthanstr. 141090WienÖsterreich
| | - Siegfried Reipert
- Core Facility für Cell Imaging und UltrastrukturforschungAlthanstr. 141090WienÖsterreich
| | - Michael Wagner
- Forschungsnetzwerk “Chemistry, Microbiology and Environmental Systems Science”Universität WienWähringer Str. 421090WienÖsterreich
- Großgeräteeinrichtung für Umwelt- und Isotopen-MassenspektrometrieZentrum für Mikrobiologie und UmweltsystemwissenschaftUniversität WienAlthanstr. 141090WienÖsterreich
| | - Bernhard K. Keppler
- Institut für Anorganische ChemieFakultät für ChemieUniversität WienWähringer Str. 421090WienÖsterreich
- Forschungsnetzwerk “Chemistry, Microbiology and Environmental Systems Science”Universität WienWähringer Str. 421090WienÖsterreich
- Forschungscluster “Translational Cancer Therapy Research”Universität WienWähringer Str. 421090WienÖsterreich
| | - Walter Berger
- Institut für Krebsforschung und Comprehensive Cancer CenterUniversitätsklinik für Innere Medizin IMedizinische Universität WienBorschkegasse 8a1090WienÖsterreich
- Forschungscluster “Translational Cancer Therapy Research”Universität WienWähringer Str. 421090WienÖsterreich
| | - Samuel M. Meier‐Menches
- Institut für Analytische ChemieFakultät für ChemieUniversität WienWähringer Str. 381090WienÖsterreich
- Forschungscluster “Translational Cancer Therapy Research”Universität WienWähringer Str. 421090WienÖsterreich
| | - Christopher Gerner
- Institut für Analytische ChemieFakultät für ChemieUniversität WienWähringer Str. 381090WienÖsterreich
- Joint Metabolome FacilityUniversität Wien und Medizinische Universität WienWähringer Str. 381090WienÖsterreich
| |
Collapse
|
36
|
Neuditschko B, Legin AA, Baier D, Schintlmeister A, Reipert S, Wagner M, Keppler BK, Berger W, Meier‐Menches SM, Gerner C. Interaction with Ribosomal Proteins Accompanies Stress Induction of the Anticancer Metallodrug BOLD-100/KP1339 in the Endoplasmic Reticulum. Angew Chem Int Ed Engl 2021; 60:5063-5068. [PMID: 33369073 PMCID: PMC7986094 DOI: 10.1002/anie.202015962] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Indexed: 02/06/2023]
Abstract
The ruthenium-based anticancer agent BOLD-100/KP1339 has shown promising results in several in vitro and in vivo tumour models as well as in early clinical trials. However, its mode of action remains to be fully elucidated. Recent evidence identified stress induction in the endoplasmic reticulum (ER) and concomitant down-modulation of HSPA5 (GRP78) as key drug effects. By exploiting the naturally formed adduct between BOLD-100 and human serum albumin as an immobilization strategy, we were able to perform target-profiling experiments that revealed the ribosomal proteins RPL10, RPL24, and the transcription factor GTF2I as potential interactors of this ruthenium(III) anticancer agent. Integrating these findings with proteomic profiling and transcriptomic experiments supported ribosomal disturbance and concomitant induction of ER stress. The formation of polyribosomes and ER swelling of treated cancer cells revealed by TEM validated this finding. Thus, the direct interaction of BOLD-100 with ribosomal proteins seems to accompany ER stress-induction and modulation of GRP78 in cancer cells.
Collapse
Affiliation(s)
- Benjamin Neuditschko
- Institute of Inorganic ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 421090ViennaAustria
- Department of Analytical ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 381090ViennaAustria
| | - Anton A. Legin
- Institute of Inorganic ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 421090ViennaAustria
- Research Network “Chemistry, Microbiology and Environmental Systems Science”University of ViennaWähringer Str. 421090ViennaAustria
| | - Dina Baier
- Institute of Inorganic ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 421090ViennaAustria
- Institute of Cancer Research and Comprehensive Cancer CenterDepartment of Medicine IMedical University of ViennaBorschkegasse 8a1090ViennaAustria
- Research Cluster “Translational Cancer Therapy Research”University of ViennaWaehringer Str. 421090ViennaAustria
| | - Arno Schintlmeister
- Research Network “Chemistry, Microbiology and Environmental Systems Science”University of ViennaWähringer Str. 421090ViennaAustria
- Large-Instrument Facility for Environmental and Isotope Mass SpectrometryCentre for Microbiology and Environmental Systems ScienceUniversity of ViennaAlthanstr. 141090ViennaAustria
| | - Siegfried Reipert
- Core Facility Cell Imaging and Ultrastructure ResearchAlthanstr. 141090ViennaAustria
| | - Michael Wagner
- Research Network “Chemistry, Microbiology and Environmental Systems Science”University of ViennaWähringer Str. 421090ViennaAustria
- Large-Instrument Facility for Environmental and Isotope Mass SpectrometryCentre for Microbiology and Environmental Systems ScienceUniversity of ViennaAlthanstr. 141090ViennaAustria
| | - Bernhard K. Keppler
- Institute of Inorganic ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 421090ViennaAustria
- Research Network “Chemistry, Microbiology and Environmental Systems Science”University of ViennaWähringer Str. 421090ViennaAustria
- Research Cluster “Translational Cancer Therapy Research”University of ViennaWaehringer Str. 421090ViennaAustria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer CenterDepartment of Medicine IMedical University of ViennaBorschkegasse 8a1090ViennaAustria
- Research Cluster “Translational Cancer Therapy Research”University of ViennaWaehringer Str. 421090ViennaAustria
| | - Samuel M. Meier‐Menches
- Department of Analytical ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 381090ViennaAustria
- Research Cluster “Translational Cancer Therapy Research”University of ViennaWaehringer Str. 421090ViennaAustria
| | - Christopher Gerner
- Department of Analytical ChemistryFaculty of ChemistryUniversity of ViennaWaehringer Str. 381090ViennaAustria
- Joint Metabolome FacilityUniversity of Vienna and Medical University of ViennaWaehringer Str. 381090ViennaAustria
| |
Collapse
|
37
|
Vidal A, Battistin F, Milani B, Balducci G, Alessio E. Stereoisomeric Control in [RuCl
2
(PTA)
2
(2L)] Complexes (2L=2py or bpy): From Theoretical Calculations to a 2+2 Metallacycle of Pyridylporphyrins. Eur J Inorg Chem 2021. [DOI: 10.1002/ejic.202000996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Alessio Vidal
- Department of Chemical and Pharmaceutical Sciences University of Trieste Via L. Giorgieri 1 34127 Trieste Italy
| | - Federica Battistin
- Department of Chemical and Pharmaceutical Sciences University of Trieste Via L. Giorgieri 1 34127 Trieste Italy
- Current address: IMDEA Nanociencia Ciudad Universitaria de Cantoblanco Faraday 9 28049 Madrid Spain
| | - Barbara Milani
- Department of Chemical and Pharmaceutical Sciences University of Trieste Via L. Giorgieri 1 34127 Trieste Italy
| | - Gabriele Balducci
- Department of Chemical and Pharmaceutical Sciences University of Trieste Via L. Giorgieri 1 34127 Trieste Italy
| | - Enzo Alessio
- Department of Chemical and Pharmaceutical Sciences University of Trieste Via L. Giorgieri 1 34127 Trieste Italy
| |
Collapse
|
38
|
Kaim V, Kaur-Ghumaan S. Mononuclear Mn complexes featuring N,S-/N,N-donor and 1,3,5-triaza-7-phosphaadamantane ligands: synthesis and electrocatalytic properties. NEW J CHEM 2021. [DOI: 10.1039/d1nj02104d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mononuclear Mn(i) carbonyl complexes incorporating 2-mercaptobenzothiazole or 2-mercaptobenzimidazole and phosphaadamantane ligands were evaluated as electrocatalysts for the HER both in acetonitrile and acetonitrile/water.
Collapse
Affiliation(s)
- Vishakha Kaim
- Department of Chemistry, University of Delhi, Delhi 110007, India
| | | |
Collapse
|
39
|
Anthony EJ, Bolitho EM, Bridgewater HE, Carter OWL, Donnelly JM, Imberti C, Lant EC, Lermyte F, Needham RJ, Palau M, Sadler PJ, Shi H, Wang FX, Zhang WY, Zhang Z. Metallodrugs are unique: opportunities and challenges of discovery and development. Chem Sci 2020; 11:12888-12917. [PMID: 34123239 PMCID: PMC8163330 DOI: 10.1039/d0sc04082g] [Citation(s) in RCA: 351] [Impact Index Per Article: 70.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/13/2020] [Indexed: 12/15/2022] Open
Abstract
Metals play vital roles in nutrients and medicines and provide chemical functionalities that are not accessible to purely organic compounds. At least 10 metals are essential for human life and about 46 other non-essential metals (including radionuclides) are also used in drug therapies and diagnostic agents. These include platinum drugs (in 50% of cancer chemotherapies), lithium (bipolar disorders), silver (antimicrobials), and bismuth (broad-spectrum antibiotics). While the quest for novel and better drugs is now as urgent as ever, drug discovery and development pipelines established for organic drugs and based on target identification and high-throughput screening of compound libraries are less effective when applied to metallodrugs. Metallodrugs are often prodrugs which undergo activation by ligand substitution or redox reactions, and are multi-targeting, all of which need to be considered when establishing structure-activity relationships. We focus on early-stage in vitro drug discovery, highlighting the challenges of evaluating anticancer, antimicrobial and antiviral metallo-pharmacophores in cultured cells, and identifying their targets. We highlight advances in the application of metal-specific techniques that can assist the preclinical development, including synchrotron X-ray spectro(micro)scopy, luminescence, and mass spectrometry-based methods, combined with proteomic and genomic (metallomic) approaches. A deeper understanding of the behavior of metals and metallodrugs in biological systems is not only key to the design of novel agents with unique mechanisms of action, but also to new understanding of clinically-established drugs.
Collapse
Affiliation(s)
- Elizabeth J Anthony
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Elizabeth M Bolitho
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Hannah E Bridgewater
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Oliver W L Carter
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Jane M Donnelly
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Cinzia Imberti
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Edward C Lant
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Frederik Lermyte
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
- Department of Chemistry, Technical University of Darmstadt Alarich-Weiss-Strasse 4 64287 Darmstadt Germany
| | - Russell J Needham
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Marta Palau
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Peter J Sadler
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Huayun Shi
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Fang-Xin Wang
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Wen-Ying Zhang
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Zijin Zhang
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| |
Collapse
|
40
|
Steel TR, Hartinger CG. Metalloproteomics for molecular target identification of protein-binding anticancer metallodrugs. Metallomics 2020; 12:1627-1636. [PMID: 33063808 DOI: 10.1039/d0mt00196a] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Proteomics has played an important role in elucidating the fundamental processes occuring in living cells. Translating these methods to metallodrug research ('metalloproteomics') has provided a means for molecular target identification of metal-based anticancer agents which should signifcantly advance the research field. In combination with biological assays, these techniques have enabled the mechanisms of action of metallodrugs to be linked to their interactions with molecular targets and aid understanding of their biological properties. Such investigations have profoundly increased our knowledge of the complex and dynamic nature of metallodrug-biomolecule interactions and have provided, at least for some compound types, a more detailed picture on their specific protein-binding patterns. This perspective highlights the progression of metallodrug proteomics research for the identification of non-DNA targets from standard analytical techniques to powerful metallodrug pull-down methods.
Collapse
Affiliation(s)
- Tasha R Steel
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | | |
Collapse
|
41
|
Ribeiro GH, Guedes APM, de Oliveira TD, de Correia CRSTB, Colina-Vegas L, Lima MA, Nóbrega JA, Cominetti MR, Rocha FV, Ferreira AG, Castellano EE, Teixeira FR, Batista AA. Ruthenium(II) Phosphine/Mercapto Complexes: Their in Vitro Cytotoxicity Evaluation and Actions as Inhibitors of Topoisomerase and Proteasome Acting as Possible Triggers of Cell Death Induction. Inorg Chem 2020; 59:15004-15018. [PMID: 32997499 DOI: 10.1021/acs.inorgchem.0c01835] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In this paper, a series of new ruthenium complexes of the general formula [Ru(NS)(dpphpy)(dppb)]PF6 (Ru1-Ru3), where dpphpy = diphenyl-2-pyridylphosphine, NS ligands = 2-thiazoline-2-thiol (tzdt, Ru1), 2-mercaptopyrimidine (pySm, Ru2), and 4,6-diamino-2-mercaptopyrimidine (damp, Ru3), and dppb = 1,4-bis(diphenylphosphino)butane, were synthesized and characterized by elemental analysis, spectroscopic techniques (IR, UV/visible, and 1D and 2D NMR), and X-ray diffraction. In the characterization, the correlation between the phosphorus atoms and their respective aromatic hydrogen atoms of the compounds in the assignment stands outs, by 1H-31P HMBC experiments. The compounds show anticancer activities against A549 (lung) and MDA-MB-231 (breast) cancer cell lines, higher than the clinical drug cisplatin. All of the complexes are more cytotoxic against the cancer cell lines than against the MRC-5 (lung) and MCF-10A (breast) nontumorigenic human cell lines. For A549 tumor cells, cell cycle analysis upon treatment with Ru2 showed that it inhibits the mitotic phase because arrest was observed in the Sub-G1 phase. Additionally, the compound induces cell death by an apoptotic pathway in a dose-dependent manner, according to annexin V-PE assay. The multitargeted character of the compounds was investigated, and the biomolecules were DNA, topoisomerase IB, and proteasome, as well as the fundamental biomolecule in the pharmacokinetics of drugs, human serum albumin. The experimental results indicate that the complexes do not target DNA in the cells. At low concentrations, the compounds showed the ability to partially inhibit the catalytic activity of topoisomerase IB in the process of relaxation of the DNA plasmid. Among the complexes assayed in cultured cells, complex Ru3 was able to diminish the proteasomal chymotrypsin-like activity to a greater extent.
Collapse
Affiliation(s)
- Gabriel H Ribeiro
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Adriana P M Guedes
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Tamires D de Oliveira
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Camila R S T B de Correia
- Departamento de Genética e Evolução, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Legna Colina-Vegas
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil.,Instituto de Química, Universidade Federal do Rio Grande do Sul, CP 15003, 91501-970 Porto Alegre, Rio Grande do Sul, Brazil
| | - Mauro A Lima
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Joaquim A Nóbrega
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Márcia R Cominetti
- Departamento de Gerontologia, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo Brazil
| | - Fillipe V Rocha
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Antônio G Ferreira
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Eduardo E Castellano
- Instituto de Física de São Carlos, Universidade de São Paulo, CEP 13560-970 São Carlos, São Paulo, Brazil
| | - Felipe R Teixeira
- Departamento de Genética e Evolução, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Alzir A Batista
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| |
Collapse
|
42
|
Rafols L, Torrente S, Aguilà D, Soto-Cerrato V, Pérez-Tomás R, Gamez P, Grabulosa A. Expanding the Range of Pyrenylphosphines and Their Derived Ru(II)-Arene Complexes. Organometallics 2020. [DOI: 10.1021/acs.organomet.0c00302] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Laia Rafols
- Departament de Quı́mica Inorgànica i Orgànica, Secció de Quı́mica Inorgànica, Universitat de Barcelona, Martı́ i Franquès, 1-11, E-08028 Barcelona, Spain
| | - Sara Torrente
- Departament de Quı́mica Inorgànica i Orgànica, Secció de Quı́mica Inorgànica, Universitat de Barcelona, Martı́ i Franquès, 1-11, E-08028 Barcelona, Spain
| | - David Aguilà
- Departament de Quı́mica Inorgànica i Orgànica, Secció de Quı́mica Inorgànica, Universitat de Barcelona, Martı́ i Franquès, 1-11, E-08028 Barcelona, Spain
- Institut de Nanociència i Nanotecnologia (IN2UB), Universitat de Barcelona, E-08028 Barcelona, Spain
| | - Vanessa Soto-Cerrato
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, University of Barcelona, Campus Bellvitge, Feixa Llarga s/n, 08907 L’Hospitalet de Llobregat, Spain
- Oncobell Program, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, E-08908 Barcelona, Spain
| | - Ricardo Pérez-Tomás
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, University of Barcelona, Campus Bellvitge, Feixa Llarga s/n, 08907 L’Hospitalet de Llobregat, Spain
- Oncobell Program, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, E-08908 Barcelona, Spain
| | - Patrick Gamez
- Institut de Nanociència i Nanotecnologia (IN2UB), Universitat de Barcelona, E-08028 Barcelona, Spain
- Institució Catalana de Rercerca i Estudis Avançats (ICREA), Passeig Lluı́s Companys 23, E-08010, Barcelona, Spain
- NanoBIC, Departament de Quı́mica Inorgànica i Orgànica, Secció de Quı́mica Inorgànica, Universitat de Barcelona, Martı́ i Franquès, 1-11, E-08028 Barcelona, Spain
| | - Arnald Grabulosa
- Departament de Quı́mica Inorgànica i Orgànica, Secció de Quı́mica Inorgànica, Universitat de Barcelona, Martı́ i Franquès, 1-11, E-08028 Barcelona, Spain
- Institut de Nanociència i Nanotecnologia (IN2UB), Universitat de Barcelona, E-08028 Barcelona, Spain
| |
Collapse
|
43
|
Battistin F, Vidal A, Balducci G, Alessio E. Investigating the reactivity of neutral water-soluble Ru(ii)-PTA carbonyls towards the model imine ligands pyridine and 2,2'-bipyridine. RSC Adv 2020; 10:26717-26727. [PMID: 35515784 PMCID: PMC9055427 DOI: 10.1039/d0ra05898j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/07/2020] [Indexed: 01/09/2023] Open
Abstract
As a continuation of our strategy for preparing new Ru(ii) precursors to be exploited as building blocks in the construction of metal-mediated supramolecular assemblies with improved solubility in water, here we describe the reactivity of selected neutral Ru(ii)-PTA carbonyls (PTA = 1,3,5-triaza-7-phosphaadamantane) towards the model imine ligands pyridine (py) and 2,2'-bipyridine (bpy) and the preparation and characterization of several neutral and cationic water-soluble derivatives: trans,trans,trans-[RuCl2(CO)(py)(PTA)2] (7), cis,cis,trans-[RuCl2(CO)2(py)(PTA)] (9), cis,trans-[Ru(bpy)Cl(CO)(PTA)2]Cl (10), mer-[Ru(bpy)(CO)(PTA)3](Cl)2 (12), cis,trans-[Ru(bpy)(CO)2Cl(PTA)]Cl (13), cis,trans-[Ru(bpy)(CO)2(PTA)2](NO3)2 (14NO3). In addition, we found that light-induced isomerization in some bpy compounds could be induced. The following species, either side-products isolated in low yield or compounds obtained exclusively in solution, were also unambiguously identified: cis,cis,trans-[RuCl2(CO)(py)(PTA)2] (8), trans-[RuCl2(bpy)(CO)(PTA)] (11), cis,cis-[Ru(bpy)Cl(CO)(PTA)2]Cl (15) and cis,cis-[Ru(bpy)(CO)2Cl(PTA)]Cl (16). The X-ray structures of 7, 11·H2O, and 12·7H2O are also reported. All compounds are new and - with few exceptions - show a good solubility in water.
Collapse
Affiliation(s)
- Federica Battistin
- Department of Chemical and Pharmaceutical Sciences, University of Trieste Via L. Giorgieri 1 34127 Trieste Italy
| | - Alessio Vidal
- Department of Chemical and Pharmaceutical Sciences, University of Trieste Via L. Giorgieri 1 34127 Trieste Italy
| | - Gabriele Balducci
- Department of Chemical and Pharmaceutical Sciences, University of Trieste Via L. Giorgieri 1 34127 Trieste Italy
| | - Enzo Alessio
- Department of Chemical and Pharmaceutical Sciences, University of Trieste Via L. Giorgieri 1 34127 Trieste Italy
| |
Collapse
|
44
|
Ferraro MG, Piccolo M, Misso G, Maione F, Montesarchio D, Caraglia M, Paduano L, Santamaria R, Irace C. Breast Cancer Chemotherapeutic Options: A General Overview on the Preclinical Validation of a Multi-Target Ruthenium(III) Complex Lodged in Nucleolipid Nanosystems. Cells 2020; 9:E1412. [PMID: 32517101 PMCID: PMC7349411 DOI: 10.3390/cells9061412] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022] Open
Abstract
In this review we have showcased the preclinical development of original amphiphilic nanomaterials designed for ruthenium-based anticancer treatments, to be placed within the current metallodrugs approach leading over the past decade to advanced multitarget agents endowed with limited toxicity and resistance. This strategy could allow for new options for breast cancer (BC) interventions, including the triple-negative subtype (TNBC) with poor therapeutic alternatives. BC is currently the second most widespread cancer and the primary cause of cancer death in women. Hence, the availability of novel chemotherapeutic weapons is a basic requirement to fight BC subtypes. Anticancer drugs based on ruthenium are among the most explored and advanced next-generation metallotherapeutics, with NAMI-A and KP1019 as two iconic ruthenium complexes having undergone clinical trials. In addition, many nanomaterial Ru complexes have been recently conceived and developed into anticancer drugs demonstrating attractive properties. In this field, we focused on the evaluation of a Ru(III) complex-named AziRu-incorporated into a suite of both zwitterionic and cationic nucleolipid nanosystems, which proved to be very effective for the in vivo targeting of breast cancer cells (BBC). Mechanisms of action have been widely explored in the context of preclinical evaluations in vitro, highlighting a multitarget action on cell death pathways which are typically deregulated in neoplasms onset and progression. Moreover, being AziRu inspired by the well-known NAMI-A complex, information on non-nanostructured Ru-based anticancer agents have been included in a precise manner.
Collapse
Affiliation(s)
- Maria Grazia Ferraro
- Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (F.M.)
| | - Marialuisa Piccolo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (F.M.)
| | - Gabriella Misso
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (G.M.); (M.C.)
| | - Francesco Maione
- Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (F.M.)
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cintia 421, 80126 Naples, Italy; (D.M.); (L.P.)
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (G.M.); (M.C.)
| | - Luigi Paduano
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cintia 421, 80126 Naples, Italy; (D.M.); (L.P.)
| | - Rita Santamaria
- Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (F.M.)
| | - Carlo Irace
- Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (F.M.)
| |
Collapse
|
45
|
Meier-Menches SM, Zappe K, Bileck A, Kreutz D, Tahir A, Cichna-Markl M, Gerner C. Time-dependent shotgun proteomics revealed distinct effects of an organoruthenium prodrug and its activation product on colon carcinoma cells. Metallomics 2020; 11:118-127. [PMID: 30106070 DOI: 10.1039/c8mt00152a] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Activation kinetics of metallo-prodrugs control the types of possible interactions with biomolecules. The intact metallo-prodrug is able to engage with potential targets by purely non-covalent bonding, while the activated metallodrug can form additional coordination bonds. It is hypothesized that the additional coordinative bonding might be favourable with respect to the target selectivity of activated metallodrugs. Thus, a time-dependent shotgun proteomics study was conducted in HCT116 colon carcinoma cells with plecstatins, which are organoruthenium anticancer drug candidates. First, the target selectivity was evaluated in a time-dependent fashion, which accounted for their hydrolysis kinetics. The binding selectivity increased from 50- to 160-fold and the average specificity from 0.72 to 0.86, respectively, from the 2 h to the 4 h target profiling experiment. Target profiling after 19 h did not reveal significant enrichments, possibly due to deactivation of the probe via arene cleavage. Up to 450 interactors were identified in the target profiling experiments. A plecstatin analogue that substituted a hydrogen bond acceptor with a hydrogen bond donor abrogated the target selectivity for plectin in HCT116 whole cell lysates, underlining the necessity of this hydrogen bond acceptor for a strong interaction between plecstatin and plectin. Second, time-dependent response profiling experiments provided evidence that plecstatin-2 induced an integrated stress response (ISR) in HCT116 cell culture. The phosphorylation of eIF2α, a key mediator of the ISR, after 3 h treatment indicated that this perturbation was initiated by the intact plecstatin-2 prodrug, while the effects of plectin-targeting are mediated by activated plecstatin-2.
Collapse
|
46
|
Mészáros JP, Geisler H, Poljarević JM, Roller A, Legina MS, Hejl M, Jakupec MA, Keppler BK, Kandioller W, Enyedy ÉA. Naphthoquinones of natural origin: Aqueous chemistry and coordination to half-sandwich organometallic cations. J Organomet Chem 2020. [DOI: 10.1016/j.jorganchem.2019.121070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
47
|
Gopalakrishnan D, Sumithaa C, Kumar AM, Bhuvanesh NSP, Ghorai S, Das P, Ganeshpandian M. Encapsulation of a Ru(η6-p-cymene) complex of the antibacterial drug trimethoprim into a polydiacetylene-phospholipid assembly to enhance its in vitro anticancer and antibacterial activities. NEW J CHEM 2020. [DOI: 10.1039/d0nj03664a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The first report of a Ru(arene)–liposome nanoaggregate to enhance the in vitro anticancer activity of a Ru–arene complex in liver cancer cells.
Collapse
Affiliation(s)
| | | | - Arumugam Madan Kumar
- Cancer Biology Lab
- Molecular and Nanomedicine Research Unit
- Sathyabama Institute of Science and Technology
- Chennai
- India
| | | | - Suvankar Ghorai
- Department of Biotechnology
- SRM Institute of Science and Technology
- Kattankulathur 603 203
- India
| | - Priyadip Das
- Department of Chemistry
- SRM Institute of Science and Technology
- India
| | | |
Collapse
|
48
|
Hanif M, Hartinger CG. From the hypothesis-driven development of organometallic anticancer drugs to new methods in mode of action studies. Med Chem 2020. [DOI: 10.1016/bs.adioch.2019.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
49
|
Malviya N, Sonkar C, Ganguly R, Bhattacherjee D, Bhabak KP, Mukhopadhyay S. Novel Approach to Generate a Self-Deliverable Ru(II)-Based Anticancer Agent in the Self-Reacting Confined Gel Space. ACS APPLIED MATERIALS & INTERFACES 2019; 11:47606-47618. [PMID: 31755256 DOI: 10.1021/acsami.9b17075] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Finding the most effective method for cancer treatment is one of the thought-provoking tasks. Drug delivery by collapsing of metallogel to the cancer cell is an appealing way out. Cancer cells have an acidic environment due to excessive accumulation of lactic acid. In this work, the novel G5 gelator with a strategically free carboxylic acid arm has been designed and fabricated and characterized by several spectroscopic and microscopic techniques. These experiments suggest the formation of an ordered supramolecular gel with clover-leaf-like morphology. Mechanical properties from rheological measurements suggest the viscoelastic nature of the gel. Furthermore, we have obtained crystals of G5 from the pure dimethyl sulfoxide solution, whereas gelation gets induced by addition of water. This G5 gelator loses its gelation capability once the carboxylate is esterified by layering with methanol, which furnished the crystals of Me-G5' (G5' = G5-H). Further, the G5 gelator is used for the formation of ruthenium metallogel. Interestingly, we obtained the monomeric species [Ru(G5')(η6-p-cymene)Cl] [Ru(II)G5] only in confined gel space upon addition of a [Ru2(η6-p-cymene)2Cl4] dimer to G5. The Ru(II)G5 metallogel has an inherent anticancer property with an IC50 value of 10.53 μM for the A549 cancer cell line. Treatment of the Ru(II)G5 metallogel by lactic acid for mimicking the acidic environment of the malignant cell results in collapsing of the gel by releasing the ruthenium metal ion. This released ruthenium ion binds with the lactic acid derivative making the gelator G5 free and producing a new compound Ru(II)L, which has also shown the anticancer property. The molecular docking study revealed that the released G5 could interact with a monocarboxylate transporter to disrupt the lactate transport chain, which might induce apoptosis.
Collapse
Affiliation(s)
| | | | - Rakesh Ganguly
- Division of Chemistry and Biological Chemistry , Nanyang Technological University , 639798 Singapore
| | - Debojit Bhattacherjee
- Department of Chemistry , Indian Institute of Technology Guwahati , Guwahati 781039 , India
| | - Krishna Pada Bhabak
- Department of Chemistry , Indian Institute of Technology Guwahati , Guwahati 781039 , India
| | | |
Collapse
|
50
|
Koch V, Meschkov A, Feuerstein W, Pfeifer J, Fuhr O, Nieger M, Schepers U, Bräse S. Synthesis, Characterization, and Biological Properties of Steroidal Ruthenium(II) and Iridium(III) Complexes Based on the Androst-16-en-3-ol Framework. Inorg Chem 2019; 58:15917-15926. [DOI: 10.1021/acs.inorgchem.9b02402] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Vanessa Koch
- Institute of Organic Chemistry (IOC), Karlsruhe Institute of Technology (KIT), Fritz-Haber-Weg 6, 76131 Karlsruhe, Germany
| | - Anna Meschkov
- Institute of Organic Chemistry (IOC), Karlsruhe Institute of Technology (KIT), Fritz-Haber-Weg 6, 76131 Karlsruhe, Germany
- Institute of Functional Interfaces (IFG), Karlsruhe Institute of Technology (KIT), Hermann von Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Wolfram Feuerstein
- Institute of Inorganic Chemistry, Division Molecular Chemistry, Karlsruhe Institute of Technology (KIT), Engesserstr. 15, 76131 Karlsruhe, Germany
| | - Juliana Pfeifer
- Institute of Functional Interfaces (IFG), Karlsruhe Institute of Technology (KIT), Hermann von Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Olaf Fuhr
- Institute for Nanotechnology (INT) and Karlsruhe Nano Micro Facility (KNMF), Karlsruhe Institute of Technology (KIT), Hermann von Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Martin Nieger
- Department of Chemistry, University of Helsinki, P.O. Box 55, 00014 Helsinki, Finland
| | - Ute Schepers
- Institute of Organic Chemistry (IOC), Karlsruhe Institute of Technology (KIT), Fritz-Haber-Weg 6, 76131 Karlsruhe, Germany
- Institute of Functional Interfaces (IFG), Karlsruhe Institute of Technology (KIT), Hermann von Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Stefan Bräse
- Institute of Organic Chemistry (IOC), Karlsruhe Institute of Technology (KIT), Fritz-Haber-Weg 6, 76131 Karlsruhe, Germany
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, D-76344 Eggenstein-Leopoldshafen, Germany
| |
Collapse
|