1
|
Maximiano I, Henriques C, Teixeira RG, Marques F, Valente A, Antunes AMM. Lead to hit ruthenium-cyclopentadienyl anticancer compounds: Cytotoxicity against breast cancer cells, metabolic stability and metabolite profiling. J Inorg Biochem 2024; 251:112436. [PMID: 38016328 DOI: 10.1016/j.jinorgbio.2023.112436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 11/30/2023]
Abstract
The successful choice of hit compounds during drug development programs involves the integration of structure-activity relationship (SAR) studies with pharmacokinetic determinations, including metabolic stability assays and metabolite profiling. A panel of nine ruthenium-cyclopentadienyl (RuCp) compounds with the general formula [Ru(η5-C5H4R)(PPh3)(bipyR')]+ (with R = H, CHO, CH2OH; R' = H, CH3, CH2OH, CH2Biotin) has been tested against hormone-dependent MCF-7 and triple negative MDA-MB-231 breast cancer cells. In general, all compounds showed important cytotoxicity against both cancer cell lines and were able to inhibit the formation of MDA-MB-231 colonies in a dose-dependent manner, while showing selectivity for cancer cells over normal fibroblasts. Among them, four compounds stood out as lead structures to be further studied. Cell distribution assays revealed their preference for the accumulation at cell membrane (Ru quantification by ICP-MS) and the mechanism of cell death seemed to be mediated by apoptosis. Potential structural liabilities of lead compounds were subsequently flagged upon in vitro metabolic stability assays and metabolite profiling. The implementation of this integrated strategy led to the selection of RT151 as a promising hit compound.
Collapse
Affiliation(s)
- Inês Maximiano
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisboa 1749-016, Portugal; Centro de Química Estrutural (CQE), Institute of Molecular Sciences, Departamento de Engenharia Química, Instituto Superior Técnico (IST), Universidade de Lisboa, Lisboa 1049-001, Portugal
| | - Catarina Henriques
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisboa 1749-016, Portugal; Centro de Química Estrutural (CQE), Institute of Molecular Sciences, Departamento de Engenharia Química, Instituto Superior Técnico (IST), Universidade de Lisboa, Lisboa 1049-001, Portugal
| | - Ricardo G Teixeira
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisboa 1749-016, Portugal
| | - Fernanda Marques
- Centro de Ciências e Tecnologias Nucleares (C(2)TN) and Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, Bobadela LRS 2695-066, Portugal
| | - Andreia Valente
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisboa 1749-016, Portugal.
| | - Alexandra M M Antunes
- Centro de Química Estrutural (CQE), Institute of Molecular Sciences, Departamento de Engenharia Química, Instituto Superior Técnico (IST), Universidade de Lisboa, Lisboa 1049-001, Portugal.
| |
Collapse
|
2
|
Borutzki Y, Skos L, Gerner C, Meier‐Menches SM. Exploring the Potential of Metal-Based Candidate Drugs as Modulators of the Cytoskeleton. Chembiochem 2023; 24:e202300178. [PMID: 37345897 PMCID: PMC10946712 DOI: 10.1002/cbic.202300178] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 06/23/2023]
Abstract
During recent years, accumulating evidence suggested that metal-based candidate drugs are promising modulators of cytoskeletal and cytoskeleton-associated proteins. This was substantiated by the identification and validation of actin, vimentin and plectin as targets of distinct ruthenium(II)- and platinum(II)-based modulators. Despite this, structural information about molecular interaction is scarcely available. Here, we compile the scattered reports about metal-based candidate molecules that influence the cytoskeleton, its associated proteins and explore their potential to interfere in cancer-related processes, including proliferation, invasion and the epithelial-to-mesenchymal transition. Advances in this field depend crucially on determining binding sites and on gaining comprehensive insight into molecular drug-target interactions. These are key steps towards establishing yet elusive structure-activity relationships.
Collapse
Affiliation(s)
- Yasmin Borutzki
- Institute of Inorganic ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Doctoral School of ChemistryUniversity of Vienna1090ViennaAustria
| | - Lukas Skos
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Doctoral School of ChemistryUniversity of Vienna1090ViennaAustria
| | - Christopher Gerner
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Joint Metabolome FacilityUniversity of Vienna and Medical University Vienna1090ViennaAustria
| | - Samuel M. Meier‐Menches
- Institute of Inorganic ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
- Joint Metabolome FacilityUniversity of Vienna and Medical University Vienna1090ViennaAustria
| |
Collapse
|
3
|
Brás AR, Fernandes P, Moreira T, Morales-Sanfrutos J, Sabidó E, Antunes AMM, Valente A, Preto A. New Ruthenium-Cyclopentadienyl Complexes Affect Colorectal Cancer Hallmarks Showing High Therapeutic Potential. Pharmaceutics 2023; 15:1731. [PMID: 37376178 DOI: 10.3390/pharmaceutics15061731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Colorectal cancer (CRC) is among the most deadly cancers worldwide. Current therapeutic strategies have low success rates and several side effects. This relevant clinical problem requires the discovery of new and more effective therapeutic alternatives. Ruthenium drugs have arisen as one of the most promising metallodrugs, due to their high selectivity to cancer cells. In this work we studied, for the first time, the anticancer properties and mechanisms of action of four lead Ru-cyclopentadienyl compounds, namely PMC79, PMC78, LCR134 and LCR220, in two CRC-derived cell lines (SW480 and RKO). Biological assays were performed on these CRC cell lines to evaluate cellular distribution, colony formation, cell cycle, proliferation, apoptosis, and motility, as well as cytoskeleton and mitochondrial alterations. Our results show that all the compounds displayed high bioactivity and selectivity, as shown by low half-maximal inhibitory concentrations (IC50) against CRC cells. We observed that all the Ru compounds have different intracellular distributions. In addition, they inhibit to a high extent the proliferation of CRC cells by decreasing clonogenic ability and inducing cell cycle arrest. PMC79, LCR134, and LCR220 also induce apoptosis, increase the levels of reactive oxygen species, lead to mitochondrial dysfunction, induce actin cytoskeleton alterations, and inhibit cellular motility. A proteomic study revealed that these compounds cause modifications in several cellular proteins associated with the phenotypic alterations observed. Overall, we demonstrate that Ru compounds, especially PMC79 and LCR220, display promising anticancer activity in CRC cells with a high potential to be used as new metallodrugs for CRC therapy.
Collapse
Affiliation(s)
- Ana Rita Brás
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- Institute of Science and Innovation for Bio-Sustainability (IB-S), University of Minho, 4710-057 Braga, Portugal
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Pedro Fernandes
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- Institute of Science and Innovation for Bio-Sustainability (IB-S), University of Minho, 4710-057 Braga, Portugal
| | - Tiago Moreira
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- Institute of Science and Innovation for Bio-Sustainability (IB-S), University of Minho, 4710-057 Braga, Portugal
| | - Julia Morales-Sanfrutos
- Proteomics Unit, Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Catalonia, 08003 Barcelona, Spain
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Eduard Sabidó
- Proteomics Unit, Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Catalonia, 08003 Barcelona, Spain
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Alexandra M M Antunes
- Centro de Química Estrutural (CQE), Institute of Molecular Sciences, Departamento de Engenharia Química, Instituto Superior Técnico (IST), Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Andreia Valente
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Ana Preto
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- Institute of Science and Innovation for Bio-Sustainability (IB-S), University of Minho, 4710-057 Braga, Portugal
| |
Collapse
|
4
|
Hou Z, Vanecek AS, Tepe JJ, Odom AL. Synthesis, structure, properties, and cytotoxicity of a (quinoline)RuCp + complex. Dalton Trans 2023; 52:721-730. [PMID: 36562335 DOI: 10.1039/d2dt03484k] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A rare example of a structurally characterized metal quinoline complex was prepared using a non-covalent quinoline-based proteasome inhibitor (Quin1), and a related complex bearing an inactive quinoline ligand (Quin2) was also synthesized. The quinolines are prepared by a one-pot procedure involving titanium-catalyzed alkyne iminoamination and are bound to ruthenium by reaction with CpRu(NCMe)3+ PF6- in CH2Cl2. The arene of the quinoline is η6-bonded to the ruthenium metal center. The kinetics of quinoline displacement were investigated, and reactivity with deuterated solvents follows the order acetonitrile > DMSO > water. Quinolines with more methyl groups on the arene are more kinetically stable, and RuCp(Quin1)+ PF6- (1), which has two methyl groups on the arene, is stable for days in DMSO. In contrast, a very similar complex (2) made with Quin2 having no methyl groups on the arene was readily displaced by DMSO. Both 1 and 2 are stable in 9 : 1 water/DMSO for days with no measurable displacement of the quinoline. The cytotoxicity of the quinolines, their CpRu+-complexes, and CpRu(DMSO)3+ PF6- was investigated towards two multiple myeloma cell lines: MC/CAR and RPMI 8226. To determine whether the activity of the complexes was related to the nature of the quinoline ligands, two structurally similar quinoline ligands with vastly different biological properties were investigated. Quin1 is a cytotoxic proteasome inhibitor, whereas Quin2 is not a proteasome inhibitor and showed no discernable cytotoxicity. The ruthenium complexes showed poor cellular proteasome inhibition. However, both 1 and 2 showed good cytotoxicity towards RPMI 8226 and MC/CAR, with 1 being slightly more cytotoxic. For example, 1 has a CC50 = 2 μM in RPMI 8226, and 2 has a CC50 = 5 μM for the same cell line. In contrast, CpRu(DMSO)3+ PF6- was quite active towards MC/CAR with CC50 = 2.8 μM but showed no discernible cytotoxicity toward RPMI 8226. The mechanism of action responsible for the observed cytotoxicity is not known, but the new Ru(Cp)(Quin)+ PF6- complexes do not cross-link DNA as found for platinum-based drugs. It is concluded that the Ru(Cp)(Quin)+ PF6- complexes remain intact in the cellular assays and constitute a new class of cytotoxic metal complexes.
Collapse
Affiliation(s)
- Zhilin Hou
- Michigan State University, Department of Chemistry, 578 S. Shaw Ln, East Lansing, MI 48824, USA.
| | - Allison S Vanecek
- Michigan State University, Department of Chemistry, 578 S. Shaw Ln, East Lansing, MI 48824, USA.
| | - Jetze J Tepe
- Michigan State University, Department of Chemistry, 578 S. Shaw Ln, East Lansing, MI 48824, USA.
| | - Aaron L Odom
- Michigan State University, Department of Chemistry, 578 S. Shaw Ln, East Lansing, MI 48824, USA.
| |
Collapse
|
5
|
Polymeric Nanosystems Applied for Metal-Based Drugs and Photosensitizers Delivery: The State of the Art and Recent Advancements. Pharmaceutics 2022; 14:pharmaceutics14071506. [PMID: 35890401 PMCID: PMC9320085 DOI: 10.3390/pharmaceutics14071506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/03/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022] Open
Abstract
Nanotechnology-based approaches for targeting the delivery and controlled release of metal-based therapeutic agents have revealed significant potential as tools for enhancing the therapeutic effect of metal-based agents and minimizing their systemic toxicities. In this context, a series of polymer-based nanosized systems designed to physically load or covalently conjugate metal-based therapeutic agents have been remarkably improving their bioavailability and anticancer efficacy. Initially, the polymeric nanocarriers were applied for platinum-based chemotherapeutic agents resulting in some nanoformulations currently in clinical tests and even in medical applications. At present, these nanoassemblies have been slowly expanding for nonplatinum-containing metal-based chemotherapeutic agents. Interestingly, for metal-based photosensitizers (PS) applied in photodynamic therapy (PDT), especially for cancer treatment, strategies employing polymeric nanocarriers have been investigated for almost 30 years. In this review, we address the polymeric nanocarrier-assisted metal-based therapeutics agent delivery systems with a specific focus on non-platinum systems; we explore some biological and physicochemical aspects of the polymer–metallodrug assembly. Finally, we summarize some recent advances in polymeric nanosystems coupled with metal-based compounds that present potential for successful clinical applications as chemotherapeutic or photosensitizing agents. We hope this review can provide a fertile ground for the innovative design of polymeric nanosystems for targeting the delivery and controlled release of metal-containing therapeutic agents.
Collapse
|
6
|
Côrte-Real L, Brás AR, Pilon A, Mendes N, Ribeiro AS, Martins TD, Farinha JPS, Oliveira MC, Gärtner F, Garcia MH, Preto A, Valente A. Biotinylated Polymer-Ruthenium Conjugates: In Vitro and In Vivo Studies in a Triple-Negative Breast Cancer Model. Pharmaceutics 2022; 14:1388. [PMID: 35890283 PMCID: PMC9315599 DOI: 10.3390/pharmaceutics14071388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/15/2022] [Accepted: 06/28/2022] [Indexed: 12/10/2022] Open
Abstract
The need for new therapeutic approaches for triple-negative breast cancer is a clinically relevant problem that needs to be solved. Using a multi-targeting approach to enhance cancer cell uptake, we synthesized a new family of ruthenium(II) organometallic complexes envisaging simultaneous active and passive targeting, using biotin and polylactide (PLA), respectively. All compounds with the general formula, [Ru(η5-CpR)(P)(2,2'-bipy-4,4'-PLA-biotin)][CF3SO3], where R is -H or -CH3 and P is P(C6H5)3, P(C6H4F)3 or P(C6H4OCH3)3, were tested against triple-negative breast cancer cells MDA-MB-231 showing IC50 values between 2.3-14.6 µM, much better than cisplatin, a classical chemotherapeutic drug, in the same experimental conditions. We selected compound 1 (where R is H and P is P(C6H5)3), for further studies as it was the one showing the best biological effect. In a competitive assay with biotin, we showed that cell uptake via SMVT receptors seems to be the main transport route into the cells for this compound, validating the strategy of including biotin in the design of the compound. The effects of the compound on the hallmarks of cancer show that the compound leads to apoptosis, interferes with proliferation by affecting the formation of cell colonies in a dose-dependent manner and disrupts the cell cytoskeleton. Preliminary in vivo assays in N: NIH(S)II-nu/nu mice show that the concentrations of compound 1 used in this experiment (maximum 4 mg/kg) are safe to use in vivo, although some signs of liver toxicity are already found. In addition, the new compound shows a tendency to control tumor growth, although not significantly. In sum, we showed that compound 1 shows promising anti-cancer effects, bringing a new avenue for triple-negative breast cancer therapy.
Collapse
Affiliation(s)
- Leonor Côrte-Real
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (L.C.-R.); (A.R.B.); (A.P.); (M.H.G.)
| | - Ana Rita Brás
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (L.C.-R.); (A.R.B.); (A.P.); (M.H.G.)
- Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal;
- Institute of Science and Innovation for Bio-Sustainability, University of Minho, Campus de Gualtar, Edifício 18, 4710-057 Braga, Portugal
| | - Adhan Pilon
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (L.C.-R.); (A.R.B.); (A.P.); (M.H.G.)
| | - Nuno Mendes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (N.M.); (A.S.R.); (F.G.)
| | - Ana Sofia Ribeiro
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (N.M.); (A.S.R.); (F.G.)
| | - Tiago D. Martins
- Centro de Química Estrutural, Institute of Molecular Sciences and Department of Chemical Engineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (T.D.M.); (J.P.S.F.); (M.C.O.)
| | - José Paulo S. Farinha
- Centro de Química Estrutural, Institute of Molecular Sciences and Department of Chemical Engineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (T.D.M.); (J.P.S.F.); (M.C.O.)
| | - M. Conceição Oliveira
- Centro de Química Estrutural, Institute of Molecular Sciences and Department of Chemical Engineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (T.D.M.); (J.P.S.F.); (M.C.O.)
| | - Fátima Gärtner
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (N.M.); (A.S.R.); (F.G.)
| | - M. Helena Garcia
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (L.C.-R.); (A.R.B.); (A.P.); (M.H.G.)
| | - Ana Preto
- Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal;
- Institute of Science and Innovation for Bio-Sustainability, University of Minho, Campus de Gualtar, Edifício 18, 4710-057 Braga, Portugal
| | - Andreia Valente
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (L.C.-R.); (A.R.B.); (A.P.); (M.H.G.)
| |
Collapse
|
7
|
Ruthenium(II)-Cyclopentadienyl-Derived Complexes as New Emerging Anti-Colorectal Cancer Drugs. Pharmaceutics 2022; 14:pharmaceutics14061293. [PMID: 35745864 PMCID: PMC9228117 DOI: 10.3390/pharmaceutics14061293] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 12/15/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies and one of the leading causes of cancer-related death worldwide, urging the need for new and more efficient therapeutic approaches. Ruthenium complexes have emerged as attractive alternatives to traditional platinum-based compounds in the treatment of CRC. This work aims to evaluate anti-CRC properties, as well as to identify the mechanisms of action of ruthenium complexes with the general formula [Ru(η5-C5H4R)(PPh3)(4,4′-R′-2,2′-bipyridine)][CF3SO3], where R = CH3, CHO or CH2OH and R′ = H, CH3, CH2OH, or dibiotin ester. The complexes (Ru 1–7) displayed high bioactivity, as shown by low IC50 concentrations against CRC cells, namely, RKO and SW480. Four of the most promising ruthenium complexes (Ru 2, 5–7) were phenotypically characterized and were shown to inhibit cell viability by decreasing cell proliferation, inducing cell cycle arrest, and increasing apoptosis. These findings were in accordance with the inhibition of MEK/ERK and PI3K/AKT signaling pathways. Ruthenium complexes also led to a decrease in cellular clonogenic ability and cell migration, which was associated with the disruption of F-actin cytoskeleton integrity. Here, we demonstrated that ruthenium complexes, especially Ru7, have a high anticancer effect against CRC cells and are promising drugs to be used as a new therapeutical strategy for CRC treatment.
Collapse
|
8
|
Karas BF, Hotz JM, Gural BM, Terez KR, DiBona VL, Côrte-Real L, Valente A, Buckley BT, Cooper KR. Anticancer Activity and In Vitro to In Vivo Mechanistic Recapitulation of Novel Ruthenium-Based Metallodrugs in the Zebrafish Model. Toxicol Sci 2021; 182:29-43. [PMID: 33822233 DOI: 10.1093/toxsci/kfab041] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Ruthenium is popular as a metal core for chemotherapeutics, due to versatile molecular coordination. Because new metallodrugs are synthesized at high rates, our studies included assays in zebrafish to expedite the initial evaluation as anticancer agents. Here we evaluated novel metallodrugs (PMC79 and LCR134), and cisplatin, a widely used platinum-based chemotherapeutic. We hypothesized that this model could characterize anticancer properties and recapitulate previous in vitro results in vivo. Our findings suggest anticancer properties of PMC79 and LCR134 were similar with less toxicity than cisplatin. Exposures from 24 to 72 h at or below the LOAELs of PMC79 and LCR134 (3.9 µM and 13.5 µm, respectively), impaired blood vessel development and tailfin regeneration. Blood vessel examination through live imaging of larvae revealed distinct regional antiangiogenic impacts. The significant decrease in gene expression of the VEGF-HIF pathway and beta-actin could explain the morphological effects observed in the whole organism following exposure. Tailfin amputation in larvae exposed to PMC79 or LCR134 inhibited tissue regrowth and cell division, but did not impact normal cell proliferation unlike cisplatin. This suggests Ru drugs may be more selective in targeting cancerous cells than cisplatin. Additionally, in vitro mechanisms were confirmed. PMC79 disrupted cytoskeleton formation in larvae and P-glycoprotein transporters in vivo was inhibited at low doses which could limit off-target effects of chemotherapeutics. Our results demonstrate the value for using the zebrafish in metallodrug research to evaluate mechanisms and off-target effects. In light of the findings reported in this article, future investigation of PMC79 and LCR134 are warranted in higher vertebrate models.
Collapse
Affiliation(s)
- Brittany F Karas
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey 08854, USA
| | - Jordan M Hotz
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey 08854, USA.,Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Brian M Gural
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey 08854, USA
| | - Kristin R Terez
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey 08854, USA
| | - Victoria L DiBona
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey 08854, USA
| | - Leonor Côrte-Real
- Centro de Química Estrutural and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa 1749-016, Portugal
| | - Andreia Valente
- Centro de Química Estrutural and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa 1749-016, Portugal
| | - Brian T Buckley
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Keith R Cooper
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey 08854, USA
| |
Collapse
|
9
|
Valente A, Podolski-Renić A, Poetsch I, Filipović N, López Ó, Turel I, Heffeter P. Metal- and metalloid-based compounds to target and reverse cancer multidrug resistance. Drug Resist Updat 2021; 58:100778. [PMID: 34403910 DOI: 10.1016/j.drup.2021.100778] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/18/2021] [Accepted: 08/03/2021] [Indexed: 12/19/2022]
Abstract
Drug resistance remains the major cause of cancer treatment failure especially at the late stage of the disease. However, based on their versatile chemistry, metal and metalloid compounds offer the possibility to design fine-tuned drugs to circumvent and even specifically target drug-resistant cancer cells. Based on the paramount importance of platinum drugs in the clinics, two main areas of drug resistance reversal strategies exist: overcoming resistance to platinum drugs as well as multidrug resistance based on ABC efflux pumps. The current review provides an overview of both aspects of drug design and discusses the open questions in the field. The areas of drug resistance covered in this article involve: 1) Altered expression of proteins involved in metal uptake, efflux or intracellular distribution, 2) Enhanced drug efflux via ABC transporters, 3) Altered metabolism in drug-resistant cancer cells, 4) Altered thiol or redox homeostasis, 5) Altered DNA damage recognition and enhanced DNA damage repair, 6) Impaired induction of apoptosis and 7) Altered interaction with the immune system. This review represents the first collection of metal (including platinum, ruthenium, iridium, gold, and copper) and metalloid drugs (e.g. arsenic and selenium) which demonstrated drug resistance reversal activity. A special focus is on compounds characterized by collateral sensitivity of ABC transporter-overexpressing cancer cells. Through this approach, we wish to draw the attention to open research questions in the field. Future investigations are warranted to obtain more insights into the mechanisms of action of the most potent compounds which target specific modalities of drug resistance.
Collapse
Affiliation(s)
- Andreia Valente
- Centro de Química Estrutural and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisboa, Portugal
| | - Ana Podolski-Renić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Serbia
| | - Isabella Poetsch
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Nenad Filipović
- Department of Chemistry and Biochemistry, Faculty of Agriculture, University of Belgrade, Belgrade, Serbia
| | - Óscar López
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Sevilla, Spain
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
10
|
|
11
|
Gou Y, Huang G, Li J, Yang F, Liang H. Versatile delivery systems for non-platinum metal-based anticancer therapeutic agents. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213975] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
12
|
Han Q, Huang L, Wang Y, Sun S, Huang H, Li F, Wang F, Chen L, Zhang H, Wang Y. Platinum (II)-coordinated Portulaca oleracea polysaccharides as metal-drug based polymers for anticancer study. Colloids Surf B Biointerfaces 2021; 201:111628. [PMID: 33639509 DOI: 10.1016/j.colsurfb.2021.111628] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 01/15/2021] [Accepted: 02/13/2021] [Indexed: 12/12/2022]
Abstract
Novel polysaccharide-platinum conjugated polymers bearing alendronate on Portulaca oleracea polysaccharides (PPS) were designed and synthesized. Their chemical structures and properties were characterized by Fourier transform infrared spectroscopy (FT-IR), 1H NMR and 31P NMR spectroscopy, Thermogravimetric analysis (TGA), X-ray powder diffraction (XRD), UV-vis spectrophotometer (UV-vis) and other analysis methods. The results demonstrated that alendronate can be used as the linker of Portulaca oleracea polysaccharides and platinum compounds. Portulaca oleracea polysaccharides-alendronate (PPS-ALN) conjugates exhibited stronger antioxidant ability than PPS. The cytotoxicity assay to cancer cells was tested in vitro, and the Portulaca oleracea polysaccharides-alendronate-platinum (PPS-ALN-Pt) conjugates strongly inhibited the proliferation of cancer cells than PPS and PPS-ALN. The evaluation of complexes affinity toward supercoiled plasmid DNA, displayed a high DNA interaction. Interestingly, the platinum conjugates displayed immunological competence in HeLa cells by cellular immunofluorescence assay. Besides, the cellular platinum accumulation of PPS-ALN-Pt conjugates was higher than that of cisplatin in HeLa cells, implying that the polysaccharide-platinum conjugated polymers might have a synergistically therapeutic application in metal anticancer drug delivery.
Collapse
Affiliation(s)
- Qianqian Han
- Institute of Environmental Toxicology and Environmental Ecology, Yancheng Teachers University, Yancheng City, Jiangsu Province, 224051, People's Republic of China; Chemistry and Chemical Engineering, Nanjing University of Technology, Nanjing City, Jiangsu Province, 210009, People's Republic of China
| | - Lirong Huang
- Cardio-Thoracic Surgery, Yancheng First People's Hospital, Yancheng, 224006, People's Republic of China
| | - Ying Wang
- Institute of Environmental Toxicology and Environmental Ecology, Yancheng Teachers University, Yancheng City, Jiangsu Province, 224051, People's Republic of China; Chemistry and Chemical Engineering, Nanjing University of Technology, Nanjing City, Jiangsu Province, 210009, People's Republic of China
| | - Shixin Sun
- Institute of Environmental Toxicology and Environmental Ecology, Yancheng Teachers University, Yancheng City, Jiangsu Province, 224051, People's Republic of China.
| | - Hao Huang
- Institute of Environmental Toxicology and Environmental Ecology, Yancheng Teachers University, Yancheng City, Jiangsu Province, 224051, People's Republic of China
| | - Fei Li
- Institute of Environmental Toxicology and Environmental Ecology, Yancheng Teachers University, Yancheng City, Jiangsu Province, 224051, People's Republic of China
| | - Fangtian Wang
- Institute of Environmental Toxicology and Environmental Ecology, Yancheng Teachers University, Yancheng City, Jiangsu Province, 224051, People's Republic of China
| | - Ligen Chen
- Department of Bioengineering School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, 224054, People's Republic of China
| | - Hongmei Zhang
- Institute of Environmental Toxicology and Environmental Ecology, Yancheng Teachers University, Yancheng City, Jiangsu Province, 224051, People's Republic of China.
| | - Yanqing Wang
- Institute of Environmental Toxicology and Environmental Ecology, Yancheng Teachers University, Yancheng City, Jiangsu Province, 224051, People's Republic of China.
| |
Collapse
|
13
|
Lenis-Rojas OA, Robalo MP, Tomaz AI, Fernandes AR, Roma-Rodrigues C, Teixeira RG, Marques F, Folgueira M, Yáñez J, Gonzalez AA, Salamini-Montemurri M, Pech-Puch D, Vázquez-García D, Torres ML, Fernández A, Fernández JJ. Half-Sandwich Ru( p-cymene) Compounds with Diphosphanes: In Vitro and In Vivo Evaluation As Potential Anticancer Metallodrugs. Inorg Chem 2021; 60:2914-2930. [PMID: 33570919 DOI: 10.1021/acs.inorgchem.0c02768] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Ruthenium(II) complexes are currently considered attractive alternatives to the widely used platinum-based drugs. We present herein the synthesis and characterization of half-sandwich ruthenium compounds formulated as [Ru(p-cymene)(L)Cl][CF3SO3] (L = 1,1-bis(methylenediphenylphosphano)ethylene, 1; L = 1,1-bis(diphenylphosphano)ethylene, 2), which were characterized by elemental analysis, mass spectrometry, 1H and 31P{1H} NMR, UV-vis and IR spectroscopy, conductivity measurements and cyclic voltammetry. The molecular structures for both complexes were determined by single-crystal X-ray diffraction. Their cytotoxic activity was evaluated using the MTT assay against human tumor cells, namely ovarian (A2780) and breast (MCF7 and MDA-MB-231). Both complexes were active against breast adenocarcinoma cells, with complex 1 exhibiting a quite remarkable cytotoxicity in the submicromolar range. Interestingly, at concentrations equivalent to the IC50 values in the MCF7 cancer cells, complexes 1 and 2 presented lower cytotoxicity in normal human primary fibroblasts. The antiproliferative effects of 1 and 2 in MCF7 cells might be associated with the induction of reactive oxygen species (ROS), leading to a combined cell death mechanism via apoptosis and autophagy. Despite the fact that in vitro a partial intercalation between complexes and DNA was observed, no MCF7 cell cycle delay or arrest was observed, indicating that DNA might not be a direct target. Complexes 1 and 2 both exhibited a moderate to strong interaction with human serum albumin, suggesting that protein targets may be involved in their mode of action. Their acute toxicity was evaluated in the zebrafish model. Complex 1 (the most toxic of the two) exhibited a lethal toxicity LC50 value about 1 order of magnitude higher than any IC50 concentrations found for the cancer cell models used, highlighting its therapeutic relevance as a drug candidate in cancer chemotherapy.
Collapse
Affiliation(s)
- Oscar A Lenis-Rojas
- Instituto de Tecnologia Química e Biológica António Xavier, ITQB, Av. da República, EAN, 2780-157 Oeiras, Portugal
| | - M Paula Robalo
- Área Departamental de Engenharia Química, ISEL-Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, Rua Conselheiro Emídio Navarro, 1, 1959-007 Lisboa, Portugal.,Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Ana Isabel Tomaz
- Centro de Química Estrutural and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1049-016 Lisboa, Portugal
| | - Alexandra R Fernandes
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Catarina Roma-Rodrigues
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Ricardo G Teixeira
- Centro de Química Estrutural and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1049-016 Lisboa, Portugal
| | - Fernanda Marques
- Centro de Ciências e Tecnologías Nucleares (C2TN), Instituto Superior Técnico, Universidade de Lisboa, E.N. 10 (km 139.7), 2695-066 Bobadela LRS, Portugal
| | - Mónica Folgueira
- Neurover Group, Centro de Investigacións Científicas Avanzadas (CICA) and Department of Biology, Universidade da Coruña, 15008 A Coruña, Spain.,Department of Cell and Developmental Biology, University College London, Gower Street, London WC1 6BT, U.K
| | - Julián Yáñez
- Neurover Group, Centro de Investigacións Científicas Avanzadas (CICA) and Department of Biology, Universidade da Coruña, 15008 A Coruña, Spain
| | - Anabel Alba Gonzalez
- Neurover Group, Centro de Investigacións Científicas Avanzadas (CICA) and Department of Biology, Universidade da Coruña, 15008 A Coruña, Spain
| | - Martín Salamini-Montemurri
- Neurover Group, Centro de Investigacións Científicas Avanzadas (CICA) and Department of Biology, Universidade da Coruña, 15008 A Coruña, Spain
| | - Dawrin Pech-Puch
- Departamento de Química & Centro de Investigaciones Científicas Avanzadas (CICA), Universidade da Coruña, 15008 A Coruña, Spain.,Departamento de Biología Marina, Universidad Autónoma de Yucatán, Km. 15.5, carretera Mérida-Xmatkuil, A.P. 4-116 Itzimná, C.P. 97100, Mérida, Yucatán, Mexico
| | - Digna Vázquez-García
- Departamento de Química & Centro de Investigaciones Científicas Avanzadas (CICA), Universidade da Coruña, 15008 A Coruña, Spain
| | - Margarita López Torres
- Departamento de Química & Centro de Investigaciones Científicas Avanzadas (CICA), Universidade da Coruña, 15008 A Coruña, Spain
| | - Alberto Fernández
- Departamento de Química & Centro de Investigaciones Científicas Avanzadas (CICA), Universidade da Coruña, 15008 A Coruña, Spain
| | - Jesús J Fernández
- Departamento de Química & Centro de Investigaciones Científicas Avanzadas (CICA), Universidade da Coruña, 15008 A Coruña, Spain
| |
Collapse
|
14
|
Kar B, Roy N, Pete S, Moharana P, Paira P. Ruthenium and iridium based mononuclear and multinuclear complexes: A Breakthrough of Next-Generation anticancer metallopharmaceuticals. Inorganica Chim Acta 2020. [DOI: 10.1016/j.ica.2020.119858] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Sun YM, Jiang X, Liu ZY, Liu LG, Liao YH, Zeng L, Ye Y, Liu HY. Hydroxy-corrole and its gallium(III) complex as new photosensitizer for photodynamic therapy against breast carcinoma. Eur J Med Chem 2020; 208:112794. [PMID: 32916313 DOI: 10.1016/j.ejmech.2020.112794] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/27/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022]
Abstract
Three mono-hydroxy corroles 1-3 and their gallium(III) complexes Ga1-3 were synthesized, and their photodynamic antitumour activities towards breast cancer cells were investigated. All corroles showed excellent cytotoxicity against the MDA-MB-231 and 4T1 cell lines upon light irradiation at 625 nm. Ga3 exhibited excellent phototoxicity and selectivity against MDA-MB-231 cells, with an IC50 of 0.06 ± 0.03 μM and a selective index value of 1338.83 (relative to human normal Huvec cells). The performance of Ga3 was even better than that of the clinical photodynamic therapy drug m-THPC. A preliminary mechanistic investigation revealed that corrole 3 and Ga3 were mainly located in the cytoplasm. Upon irradiation, they could generate intracellular reactive oxygen to destroy the mitochondrial membrane potential and arrest the cell cycle at the sub-G1 phase. Flow cytometry revealed that corrole 3 and Ga3 induced cancer cell apoptosis after photodynamic treatment. Corrole 3 and Ga3 displayed negligible cytotoxicity in the dark. These results suggest that corrole 3 and Ga3 are promising candidates for use in the photodynamic therapy of breast cancer.
Collapse
Affiliation(s)
- Yan-Mei Sun
- School of Chemistry and Chemical Engineering, Key Laboratory of Functional Molecular Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510641, China
| | - Xiao Jiang
- School of Chemistry and Chemical Engineering, Key Laboratory of Functional Molecular Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510641, China
| | - Ze-Yu Liu
- School of Chemistry and Chemical Engineering, Key Laboratory of Functional Molecular Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510641, China
| | - Ling-Gui Liu
- School of Chemistry and Chemical Engineering, Key Laboratory of Functional Molecular Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510641, China
| | - Yu-Hui Liao
- Molecular Diagnosis & Treatment of Central Infectious Diseases, Dermatology Hospital, Southern Medical University, Guangzhou, 510091, China
| | - Lei Zeng
- Foresea Life Insurance Guangzhou General Hospital, Guangzhou, 511300, China
| | - Yong Ye
- School of Chemistry and Chemical Engineering, Key Laboratory of Functional Molecular Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510641, China
| | - Hai-Yang Liu
- School of Chemistry and Chemical Engineering, Key Laboratory of Functional Molecular Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510641, China.
| |
Collapse
|
16
|
Pilon A, Brás AR, Côrte-Real L, Avecilla F, Costa PJ, Preto A, Garcia MH, Valente A. A New Family of Iron(II)-Cyclopentadienyl Compounds Shows Strong Activity Against Colorectal and Triple Negative Breast Cancer Cells. Molecules 2020; 25:E1592. [PMID: 32235674 PMCID: PMC7180908 DOI: 10.3390/molecules25071592] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/22/2020] [Accepted: 03/25/2020] [Indexed: 01/29/2023] Open
Abstract
A family of compounds with the general formula [Fe(η5-C5H5)(CO)(PPh3)(NCR)]+ has been synthesized (NCR = benzonitrile (1); 4-hydroxybenzonitrile (2); 4-hydroxymethylbenzonitrile (3); 4-aminobenzonitrile (4); 4-bromobenzonitrile (5); and, 4-chlorocinnamonitrile (6)). All of the compounds were obtained in good yields and were completely characterized by standard spectroscopic and analytical techniques. Compounds 1, 4, and 5 crystallize in the monoclinc P21/c space group and packing is determined by short contacts between the phosphane phenyl rings and cyclopentadienyl (compounds 1 and 4) or π-π lateral interactions between the benzonitrile molecules (complex 5). DFT and TD-DFT calculations were performed to help in the interpretation of the experimental UV-Vis. data and assign the electronic transitions. Cytotoxicity studies in MDA-MB-231 breast and SW480 colorectal cancer-derived cell lines showed IC50 values at a low micromolar range for all of the compounds in both cell lines. The determination of the selectivity index for colorectal cells (SW480 vs. NCM460, a normal colon-derived cell line) indicates that the compounds have some inherent selectivity. Further studies on the SW480 cell line demonstrated that the compounds induce cell death by apoptosis, inhibit proliferation by inhibiting the formation of colonies, and affect the actin-cytoskeleton of the cells. These results are not observed for the hydroxylated compounds 2 and 3, where an alternative mode of action might be present. Overall, the results indicate that the substituent at the nitrile-based ligand is associated to the biological activity of the compounds.
Collapse
Affiliation(s)
- Adhan Pilon
- Centro de Química Estrutural, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (A.P.); (A.R.B.); (L.C.-R.)
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Ana Rita Brás
- Centro de Química Estrutural, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (A.P.); (A.R.B.); (L.C.-R.)
- Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal;
- Institute of Science and Innovation for Bio-Sustainability, University of Minho, Campus de Gualtar, Edifício 18, 4710-057 Braga, Portugal
| | - Leonor Côrte-Real
- Centro de Química Estrutural, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (A.P.); (A.R.B.); (L.C.-R.)
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Fernando Avecilla
- Grupo Xenomar, Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Química, Facultade de Ciencias, Universidade da Coruña, Campus de A Coruña, 15071 A Coruña, Spain;
| | - Paulo J. Costa
- Faculty of Sciences, University of Lisboa, BioISI—Biosystems & Integrative Sciences Institute, Campo Grande, C8 bdg, 1749-016 Lisboa, Portugal;
| | - Ana Preto
- Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal;
- Institute of Science and Innovation for Bio-Sustainability, University of Minho, Campus de Gualtar, Edifício 18, 4710-057 Braga, Portugal
| | - M. Helena Garcia
- Centro de Química Estrutural, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (A.P.); (A.R.B.); (L.C.-R.)
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Andreia Valente
- Centro de Química Estrutural, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (A.P.); (A.R.B.); (L.C.-R.)
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|
17
|
Golbaghi G, Pitard I, Lucas M, Haghdoost MM, de Los Santos YL, Doucet N, Patten SA, Sanderson JT, Castonguay A. Synthesis and biological assessment of a ruthenium(II) cyclopentadienyl complex in breast cancer cells and on the development of zebrafish embryos. Eur J Med Chem 2020; 188:112030. [PMID: 31945643 PMCID: PMC7221417 DOI: 10.1016/j.ejmech.2019.112030] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/17/2019] [Accepted: 12/30/2019] [Indexed: 12/31/2022]
Abstract
Ruthenium-based complexes currently attract great attention as they hold promise to replace platinum-based drugs as a first line cancer treatment. Whereas ruthenium arene complexes are some of the most studied species for their potential anticancer properties, other types of ruthenium complexes have been overlooked for this purpose. Here, we report the synthesis and characterization of Ru(II) cyclopentadienyl (Cp), Ru(II) cyclooctadienyl (COD) and Ru(III) complexes bearing anastrozole or letrozole ligands, third-generation aromatase inhibitors currently used for the treatment of estrogen receptor positive (ER +) breast cancer. Among these complexes, Ru(II)Cp 2 was the only one that displayed a high stability in DMSO and in cell culture media and consequently, the only complex for which the in vitro and in vivo biological activities were investigated. Unlike anastrozole alone, complex 2 was considerably cytotoxic in vitro (IC50 values < 1 μM) in human ER + breast cancer (T47D and MCF7), triple negative breast cancer (TNBC) (MBA-MB-231), and in adrenocortical carcinoma (H295R) cells. Theoretical (docking simulation) and experimental (aromatase catalytic activity) studies suggested that an interaction between 2 and the aromatase enzyme was not likely to occur and that the bulkiness of the PPh3 ligands could be an important factor preventing the complex to reach the active site of the enzyme. Exposure of zebrafish embryos to complex 2 at concentrations around its in vitro cytotoxicity IC50 value (0.1-1 μM) did not lead to noticeable signs of toxicity over 96 h, making it a suitable candidate for further in vivo investigations. This study confirms the potential of Ru(II)Cp complexes for breast cancer therapy, more specifically against TNBCs that are usually not responsive to currently used chemotherapeutic agents.
Collapse
Affiliation(s)
- Golara Golbaghi
- Organometallic Chemistry Laboratory for the Design of Catalysts and Therapeutics, and Endocrine Toxicology Laboratory, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, Canada
| | - Irène Pitard
- Organometallic Chemistry Laboratory for the Design of Catalysts and Therapeutics, and Endocrine Toxicology Laboratory, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, Canada
| | - Matthieu Lucas
- Organometallic Chemistry Laboratory for the Design of Catalysts and Therapeutics, and Endocrine Toxicology Laboratory, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, Canada
| | - Mohammad Mehdi Haghdoost
- Organometallic Chemistry Laboratory for the Design of Catalysts and Therapeutics, and Endocrine Toxicology Laboratory, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, Canada
| | - Yossef López de Los Santos
- Organometallic Chemistry Laboratory for the Design of Catalysts and Therapeutics, and Endocrine Toxicology Laboratory, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, Canada
| | - Nicolas Doucet
- Organometallic Chemistry Laboratory for the Design of Catalysts and Therapeutics, and Endocrine Toxicology Laboratory, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, Canada
| | - Shunmoogum A Patten
- Organometallic Chemistry Laboratory for the Design of Catalysts and Therapeutics, and Endocrine Toxicology Laboratory, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, Canada
| | - J Thomas Sanderson
- Organometallic Chemistry Laboratory for the Design of Catalysts and Therapeutics, and Endocrine Toxicology Laboratory, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, Canada
| | - Annie Castonguay
- Organometallic Chemistry Laboratory for the Design of Catalysts and Therapeutics, and Endocrine Toxicology Laboratory, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, Canada.
| |
Collapse
|
18
|
Karas BF, Côrte-Real L, Doherty C, Valente A, Cooper KR, Buckley BT. A novel screening method for transition metal-based anticancer compounds using zebrafish embryo-larval assay and inductively coupled plasma-mass spectrometry analysis. J Appl Toxicol 2019; 39:1173-1180. [PMID: 30963621 PMCID: PMC6625851 DOI: 10.1002/jat.3802] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/19/2019] [Accepted: 02/28/2019] [Indexed: 12/20/2022]
Abstract
As novel metallodrugs continue to emerge, they are evaluated using models, including zebrafish, that offer unique sublethal endpoints. Testing metal-based anticancer compounds with high-throughput zebrafish toxicological assays requires analytical methods with the sensitivity to detect these sublethal tissue doses in very small sample masses (e.g., egg mass 100 μg). A robust bioanalytical model, zebrafish embryos coupled with inductively coupled plasma-mass spectrometry (ICPMS) for measurement of delivered dose, creates a very effective means for screening metal-based chemotherapeutic agents. In this study, we used ICPMS quantitation with the zebrafish embryo assays to detect metal equivalents at multiple response endpoints for two compounds, the chemotherapeutic agent cisplatin and ruthenium (Ru)-based prospective metallodrug, PMC79. We hypothesized that cisplatin and PMC79 have different mechanisms for inducing apoptosis and result in similar lesions but different potencies following water-borne exposure. An ICPMS method was developed to detect the metal in waterborne solution and tissue (detection limit: 5 parts per trillion for Ru or platinum [Pt]). The Ru-based compound was more potent (LC50 : 7.8 μm) than cisplatin (LC50 : 158 μm) and induced disparate lesions. Lethality from cisplatin exposure exhibited a threshold (values >15 mg/L) while no threshold was observed for delayed hatching (lowest observed adverse effect level 3.75 mg/L cisplatin; 8.7 Pt (ng)/organism). The Ru organometallic did not have a threshold for lethality. Cisplatin-induced delayed hatching was investigated further by larval-Pt distribution and preferentially distributed to the chorion. We propose that zebrafish embryo-larval assays coupled with ICPMS serve as a powerful platform to evaluate relative potency and toxic effects of metallodrug candidates.
Collapse
Affiliation(s)
- Brittany F. Karas
- Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Road, Piscataway NJ, 08854, U.S.A
- Department of Biochemistry and Microbiology, Rutgers University, 76 Lipman Drive, New Brunswick NJ, 08854, U.S.A
| | - Leonor Côrte-Real
- Centro de Química Estrutural, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Cathleen Doherty
- Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Road, Piscataway NJ, 08854, U.S.A
| | - Andreia Valente
- Centro de Química Estrutural, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Keith R. Cooper
- Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Road, Piscataway NJ, 08854, U.S.A
- Department of Biochemistry and Microbiology, Rutgers University, 76 Lipman Drive, New Brunswick NJ, 08854, U.S.A
| | - Brian T. Buckley
- Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Road, Piscataway NJ, 08854, U.S.A
| |
Collapse
|
19
|
Sojka M, Fojtu M, Fialova J, Masarik M, Necas M, Marek R. Locked and Loaded: Ruthenium(II)-Capped Cucurbit[ n]uril-Based Rotaxanes with Antimetastatic Properties. Inorg Chem 2019; 58:10861-10870. [PMID: 31355636 DOI: 10.1021/acs.inorgchem.9b01203] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
We report here the first coupling of Ru(II) units with cucurbit[6/7]uril-based pseudorotaxane ligands meant for biological application. The resulting ruthenium-capped rotaxanes were fully characterized, and a structure of one supramolecular system was determined by X-ray diffraction. Because the biological properties of Ru-based metallodrugs are tightly linked to the ligand-exchange processes, the effect of salt concentration on the hydrolysis of chlorides from the Ru(II) center was monitored by using 1H NMR spectroscopy. The biological activity of Ru(II)-based rotaxanes was evaluated for three selected mammalian breast cell lines, HBL-100, MCF-7, and MDA-MB-231. The antimetastatic activity of the assembled cationic Ru(II)-rotaxane systems, evaluated in migration assays against MCF-7 and MDA-MB-231 cell lines, is notably enhanced compared to that of RAPTA-C, a reference that was used. The indicated synergistic effect of combining Ru(II) with a pseudorotaxane unit opens a new direction in searching for anticancer supramolecular metallodrugs.
Collapse
Affiliation(s)
- Martin Sojka
- Department of Chemistry, Faculty of Science , Masaryk University , Kamenice 5 , CZ-62500 Brno , Czechia.,CEITEC-Central European Institute of Technology , Masaryk University , Kamenice 5 , CZ-62500 Brno , Czechia
| | - Michaela Fojtu
- CEITEC-Central European Institute of Technology , Masaryk University , Kamenice 5 , CZ-62500 Brno , Czechia.,Department of Pathological Physiology, Faculty of Medicine , Masaryk University , Kamenice 5 , CZ-62500 Brno , Czechia.,Department of Physiology, Faculty of Medicine , Masaryk University , Kamenice 5 , CZ-62500 Brno , Czechia
| | - Jindriska Fialova
- Department of Physiology, Faculty of Medicine , Masaryk University , Kamenice 5 , CZ-62500 Brno , Czechia
| | - Michal Masarik
- CEITEC-Central European Institute of Technology , Masaryk University , Kamenice 5 , CZ-62500 Brno , Czechia.,Department of Pathological Physiology, Faculty of Medicine , Masaryk University , Kamenice 5 , CZ-62500 Brno , Czechia.,Department of Physiology, Faculty of Medicine , Masaryk University , Kamenice 5 , CZ-62500 Brno , Czechia
| | - Marek Necas
- Department of Chemistry, Faculty of Science , Masaryk University , Kamenice 5 , CZ-62500 Brno , Czechia.,CEITEC-Central European Institute of Technology , Masaryk University , Kamenice 5 , CZ-62500 Brno , Czechia
| | - Radek Marek
- Department of Chemistry, Faculty of Science , Masaryk University , Kamenice 5 , CZ-62500 Brno , Czechia.,CEITEC-Central European Institute of Technology , Masaryk University , Kamenice 5 , CZ-62500 Brno , Czechia
| |
Collapse
|
20
|
Côrte-Real L, Karas B, Brás AR, Pilon A, Avecilla F, Marques F, Preto A, Buckley BT, Cooper KR, Doherty C, Garcia MH, Valente A. Ruthenium-Cyclopentadienyl Bipyridine-Biotin Based Compounds: Synthesis and Biological Effect. Inorg Chem 2019; 58:9135-9149. [PMID: 31241925 DOI: 10.1021/acs.inorgchem.9b00735] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Prospective anticancer metallodrugs should consider target-specific components in their design in order to overcome the limitations of the current chemotherapeutics. The inclusion of vitamins, which receptors are overexpressed in many cancer cell lines, has proven to be a valid strategy. Therefore, in this paper we report the synthesis and characterization of a set of new compounds [Ru(η5-C5H5)(P(C6H4R)3)(4,4'-R'-2,2'-bpy)]+ (R = F and R' = H, 3; R = F and R' = biotin, 4; R = OCH3 and R' = H, 5; R = OCH3 and R' = biotin, 6), inspired by the exceptional good results recently obtained for the analogue bearing a triphenylphosphane ligand. The precursors for these syntheses were also described following modified literature procedures, [Ru(η5-C5H5)(P(C6H4R)3)2Cl], where R is -F (1) or -OCH3 (2). The structure of all compounds is fully supported by spectroscopic and analytical techniques and by X-ray diffraction studies for compounds 2, 3, and 5. All cationic compounds are cytotoxic in the two breast cancer cell lines tested, MCF7 and MDA-MB-231, and much better than cisplatin under the same experimental conditions. The cytotoxicity of the biotinylated compounds seems to be related with the Ru uptake by the cells expressing biotin receptors, indicating a potential mediated uptake. Indeed, a biotin-avidin study confirmed that the attachment of biotin to the organometallic fragment still allows biotin recognition by the protein. Therefore, the biotinylated compounds might be potent anticancer drugs as they show cytotoxic effect in breast cancer cells at low dose dependent on the compounds' uptake, induce cell death by apoptosis and inhibit the colony formation of cancer cells causing also less severe side effects in zebrafish.
Collapse
Affiliation(s)
- Leonor Côrte-Real
- Centro de Química Estrutural , Faculdade de Ciências da Universidade de Lisboa , Campo Grande , 1749-016 Lisboa , Portugal
| | - Brittany Karas
- Environmental and Occupational Health Sciences Institute , Rutgers University , 170 Frelinghuysen Road , Piscataway New Jersey 08854 , United States.,Department of Biochemistry and Microbiology , Rutgers University , 76 Lipman Drive , New Brunswick New Jersey 08854 , United States
| | - Ana Rita Brás
- Centro de Química Estrutural , Faculdade de Ciências da Universidade de Lisboa , Campo Grande , 1749-016 Lisboa , Portugal.,Centre of Molecular and Environmental Biology (CBMA), Department of Biology , University of Minho , Portugal. Campus de Gualtar , Braga 4710-057 , Portugal
| | - Adhan Pilon
- Centro de Química Estrutural , Faculdade de Ciências da Universidade de Lisboa , Campo Grande , 1749-016 Lisboa , Portugal
| | - Fernando Avecilla
- Grupo Xenomar, Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Química, Facultade de Ciencias , Universidade da Coruña , Campus de A Coruña , 15071 A Coruña , Spain
| | - Fernanda Marques
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Te'cnico (C2TN/IST) , Universidade de Lisboa , Estrada Nacional 10 (km 139.7) , 2695-066 Bobadela LRS , Portugal
| | - Ana Preto
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology , University of Minho , Portugal. Campus de Gualtar , Braga 4710-057 , Portugal
| | - Brian T Buckley
- Environmental and Occupational Health Sciences Institute , Rutgers University , 170 Frelinghuysen Road , Piscataway New Jersey 08854 , United States
| | - Keith R Cooper
- Department of Biochemistry and Microbiology , Rutgers University , 76 Lipman Drive , New Brunswick New Jersey 08854 , United States
| | - Cathleen Doherty
- Environmental and Occupational Health Sciences Institute , Rutgers University , 170 Frelinghuysen Road , Piscataway New Jersey 08854 , United States
| | - M Helena Garcia
- Centro de Química Estrutural , Faculdade de Ciências da Universidade de Lisboa , Campo Grande , 1749-016 Lisboa , Portugal
| | - Andreia Valente
- Centro de Química Estrutural , Faculdade de Ciências da Universidade de Lisboa , Campo Grande , 1749-016 Lisboa , Portugal
| |
Collapse
|