1
|
Pelliccia S, Russomanno P, Barone S, Mateu B, Alfano AI, Miranda M, Coretti L, Lembo F, Piccolo M, Irace C, Friggeri L, Hargrove TY, Curtis A, Lepesheva GI, Kavanagh K, Buommino E, Brindisi M. A First-in-Class Pyrazole-isoxazole Enhanced Antifungal Activity of Voriconazole: Synergy Studies in an Azole-Resistant Candida albicans Strain, Computational Investigation and in Vivo Validation in a Galleria mellonella Fungal Infection Model. J Med Chem 2024; 67:14256-14276. [PMID: 39115219 PMCID: PMC11482282 DOI: 10.1021/acs.jmedchem.4c01109] [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] [Indexed: 08/23/2024]
Abstract
The widespread and irrational use of azole antifungal agents has led to an increase of azole-resistant Candida albicans strains with an urgent need for combination drug therapy, enhancing the treatment efficacy. Here, we report the discovery of a first-in-class pyrazole-isoxazole, namely, 5b, that showed remarkable growth inhibition against the C. albicans ATCC 10231 strain in combination with voriconazole, acting as a downregulator of ERG 11 (Cyp51) gene expression with a significant reduction of the yeast-to-hypha morphological transition. Furthermore, C. albicans CYP51 enzyme assay and in-depth molecular docking studies unveiled the unique ability of the combination of 5b and voriconazole to completely fill the CYP51 binding sites. In vivo studies using a Galleria mellonella model confirmed the previously in vitro observed synergistic effect of 5b with voriconazole. Also considering its biocompatibility in a cellular model of human keratinocytes, these results indicate that 5b represents a promising compound for a further optimization campaign.
Collapse
Affiliation(s)
- Sveva Pelliccia
- Department of Pharmacy (DoE 2023-2027), University of Naples Federico II, 80131 Naples, Italy
| | - Pasquale Russomanno
- Magnetic Resonance Centre (CERM), Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (CIRMMP) and Department of Chemistry “Ugo Schiff”, University of Florence, Sesto Fiorentino 50019, Italy
| | - Simona Barone
- Department of Pharmacy (DoE 2023-2027), University of Naples Federico II, 80131 Naples, Italy
| | - Baptiste Mateu
- Department of Pharmacy (DoE 2023-2027), University of Naples Federico II, 80131 Naples, Italy
| | - Antonella Ilenia Alfano
- Department of Pharmacy (DoE 2023-2027), University of Naples Federico II, 80131 Naples, Italy
| | - Martina Miranda
- Department of Pharmaceutical and Cell Biological Chemistry, Pharmaceutical Institute, University of Bonn, 53121 Bonn, Germany
| | - Lorena Coretti
- Department of Pharmacy (DoE 2023-2027), University of Naples Federico II, 80131 Naples, Italy
| | - Francesca Lembo
- Department of Pharmacy (DoE 2023-2027), University of Naples Federico II, 80131 Naples, Italy
| | - Marialuisa Piccolo
- BioChemLab, Department of Pharmacy,School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Carlo Irace
- BioChemLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Laura Friggeri
- Department of Cell and Development Biology, U4225 Medical Research Building III, Nashville, Tennessee 37232, United States
| | - Tatiana Y. Hargrove
- Department of Biochemistry,Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Aaron Curtis
- Department of Biology, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| | - Galina I. Lepesheva
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Kevin Kavanagh
- Department of Biology, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| | - Elisabetta Buommino
- Department of Pharmacy (DoE 2023-2027), University of Naples Federico II, 80131 Naples, Italy
| | - Margherita Brindisi
- Department of Pharmacy (DoE 2023-2027), University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
2
|
Capuano A, D’Urso G, Gazzillo E, Lauro G, Chini MG, D’Auria MV, Ferraro MG, Iazzetti F, Irace C, Bifulco G, Casapullo A. Fatty Acid Synthase as Interacting Anticancer Target of the Terpenoid Myrianthic Acid Disclosed by MS-Based Proteomics Approaches. Int J Mol Sci 2024; 25:5918. [PMID: 38892106 PMCID: PMC11172900 DOI: 10.3390/ijms25115918] [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: 04/24/2024] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
This research focuses on the target deconvolution of the natural compound myrianthic acid, a triterpenoid characterized by an ursane skeleton isolated from the roots of Myrianthus arboreus and from Oenothera maritima Nutt. (Onagraceae), using MS-based chemical proteomic techniques. Application of drug affinity responsive target stability (DARTS) and targeted-limited proteolysis coupled to mass spectrometry (t-LiP-MS) led to the identification of the enzyme fatty acid synthase (FAS) as an interesting macromolecular counterpart of myrianthic acid. This result, confirmed by comparison with the natural ursolic acid, was thoroughly investigated and validated in silico by molecular docking, which gave a precise picture of the interactions in the MA/FAS complex. Moreover, biological assays showcased the inhibitory activity of myrianthic acid against the FAS enzyme, most likely related to its antiproliferative activity towards tumor cells. Given the significance of FAS in specific pathologies, especially cancer, the myrianthic acid structural moieties could serve as a promising reference point to start the potential development of innovative approaches in therapy.
Collapse
Affiliation(s)
- Alessandra Capuano
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
- PhD Program in Drug Discovery and Development, Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Gilda D’Urso
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
| | - Erica Gazzillo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
- PhD Program in Drug Discovery and Development, Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Gianluigi Lauro
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
| | - Maria Giovanna Chini
- Department of Biosciences and Territory, University of Molise, C.da Fonte Lappone, 86090 Pesche, Italy
| | - Maria Valeria D’Auria
- Department of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano 49, 80131 Naples, Italy;
| | - Maria Grazia Ferraro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy;
| | - Federica Iazzetti
- Biochem Lab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via Domenico Montesano 49, 80131 Naples, Italy; (F.I.); (C.I.)
| | - Carlo Irace
- Biochem Lab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via Domenico Montesano 49, 80131 Naples, Italy; (F.I.); (C.I.)
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
| | - Agostino Casapullo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
| |
Collapse
|
3
|
Mansour N, Mehanna S, Bodman-Smith K, Daher CF, Khnayzer RS. A Ru(II)-Strained Complex with 2,9-Diphenyl-1,10-phenanthroline Ligand Induces Selective Photoactivatable Chemotherapeutic Activity on Human Alveolar Carcinoma Cells via Apoptosis. Pharmaceuticals (Basel) 2023; 17:50. [PMID: 38256884 PMCID: PMC10819265 DOI: 10.3390/ph17010050] [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: 10/25/2023] [Revised: 11/20/2023] [Accepted: 11/27/2023] [Indexed: 01/24/2024] Open
Abstract
[Ru(bipy)2(dpphen)]Cl2 (where bipy = 2,2'-bipyridine and dpphen = 2,9-diphenyl-1,10-phenanthroline) (complex 1) is a sterically strained compound that exhibits promising in vitro photocytotoxicity on an array of cell lines. Since lung adenocarcinoma cancer remains the most common lung cancer and the leading cause of cancer deaths, the current study aims to evaluate the plausible effect and uptake of complex 1 on human alveolar carcinoma cells (A549) and mesenchymal stem cells (MSC), and assess its cytotoxicity in vitro while considering its effect on cell morphology, membrane integrity and DNA damage. MSC and A549 cells showed similar rates of complex 1 uptake with a plateau at 12 h. Upon photoactivation, complex 1 exhibited selective, potent anticancer activity against A549 cells with phototoxicity index (PI) values of 16, 25 and 39 at 24, 48 and 72 h, respectively. This effect was accompanied by a significant increase in A549-cell rounding and detachment, loss of membrane integrity and DNA damage. Flow cytometry experiments confirmed that A549 cells undergo apoptosis when treated with complex 1 followed by photoactivation. In conclusion, this present study suggests that complex 1 might be a promising candidate for photochemotherapy with photoproducts that possess selective anticancer effects in vitro. These results are encouraging to probe the potential activity of this complex in vivo.
Collapse
Affiliation(s)
- Najwa Mansour
- Department of Natural Sciences, Lebanese American University, Chouran, Beirut 1102-2801, Lebanon; (N.M.); (S.M.); (C.F.D.)
| | - Stephanie Mehanna
- Department of Natural Sciences, Lebanese American University, Chouran, Beirut 1102-2801, Lebanon; (N.M.); (S.M.); (C.F.D.)
| | - Kikki Bodman-Smith
- Department of Microbial and Cellular Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK;
| | - Costantine F. Daher
- Department of Natural Sciences, Lebanese American University, Chouran, Beirut 1102-2801, Lebanon; (N.M.); (S.M.); (C.F.D.)
| | - Rony S. Khnayzer
- Department of Natural Sciences, Lebanese American University, Chouran, Beirut 1102-2801, Lebanon; (N.M.); (S.M.); (C.F.D.)
| |
Collapse
|
4
|
Gazzillo E, Pierri M, Colarusso E, Chini MG, Ferraro MG, Piccolo M, Irace C, Bruno I, Bifulco G, Terracciano S, Lauro G. Exploring the chemical space of functionalized [1,2,4]triazolo[4,3-a]quinoxaline-based compounds targeting the bromodomain of BRD9. Bioorg Chem 2023; 139:106677. [PMID: 37352721 DOI: 10.1016/j.bioorg.2023.106677] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 06/25/2023]
Abstract
Here we report a detailed structure-activity relationship (SAR) study related to [1,2,4]triazolo[4,3-a]quinoxaline-based compounds targeting the reader module of bromodomain containing-protein 9 (BRD9). 3D structure-based pharmacophore models, previously introduced by us, were here employed to evaluate a second generation of compounds, exploring different substitution patterns on the heterocyclic core. Starting from the promising data obtained from our previously identified [1,2,4]triazolo[4,3-a]quinoxaline-based compounds 1-4, the combination of in silico studies, chemical synthesis, biophysical and in vitro assays led to the identification of a new set of derivatives, selected for thoroughly exploring the chemical space of the bromodomain binding site. In more details, the investigation of different linkers at C-4 position highlighted the amine spacer as mandatory for the binding with the protein counterpart and the crucial role of the alkyl substituents at C-1 for increasing the selectivity toward BRD9. Additionally, the importance of a hydrogen bond donor group, critical to anchor the ZA region and required for the interaction with Ile53 residue, was inferred from the analysis of our collected results. Herein we also propose an optimization and an update of our previously reported "pharm-druglike2" 3D structure-based pharmacophore model, introducing it as "pharm-druglike2.1". Compounds 24-26, 32, 34 and 36 were identified as new valuable BRD9 binders featuring IC50 values in the low micromolar range. Among them, 24 and 36 displayed an excellent selectivity towards BRD9 and a good antiproliferative effect on a panel of leukemia models, especially toward CCRF-CEM cell line, with no cytotoxicity on healthy cells. Notably, the interaction of 24 and 36 with the bromodomain and PHD finger-containing protein 1 (BRPF1) also emerged, disclosing them as new and unexplored dual inhibitors for these two proteins highly involved in leukemia. These findings highlight the potential for the identification of new attractive dual epidrugs as well as a promising starting point for the development of chemical degraders endowed with anticancer activities.
Collapse
Affiliation(s)
- Erica Gazzillo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano 84084, Italy; PhD Program in Drug Discovery and Development, University of Salerno, Via Giovanni Paolo II 132, Fisciano 84084, Italy
| | - Martina Pierri
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano 84084, Italy; PhD Program in Drug Discovery and Development, University of Salerno, Via Giovanni Paolo II 132, Fisciano 84084, Italy
| | - Ester Colarusso
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano 84084, Italy
| | - Maria Giovanna Chini
- Department of Biosciences and Territory, University of Molise, C.da Fonte Lappone, Pesche 86090, Italy
| | - Maria Grazia Ferraro
- BioChem Lab, Department of Pharmacy, School of Medicine and Surgery, University of Naples, Via Domenico Montesano 49, Naples 80131, Italy
| | - Marialuisa Piccolo
- BioChem Lab, Department of Pharmacy, School of Medicine and Surgery, University of Naples, Via Domenico Montesano 49, Naples 80131, Italy
| | - Carlo Irace
- BioChem Lab, Department of Pharmacy, School of Medicine and Surgery, University of Naples, Via Domenico Montesano 49, Naples 80131, Italy
| | - Ines Bruno
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano 84084, Italy
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano 84084, Italy
| | - Stefania Terracciano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano 84084, Italy.
| | - Gianluigi Lauro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano 84084, Italy.
| |
Collapse
|
5
|
Pereira SAP, Romano-deGea J, Barbosa AI, Costa Lima SA, Dyson PJ, Saraiva MLMFS. Fine-tuning the cytotoxicity of ruthenium(II) arene compounds to enhance selectivity against breast cancers. Dalton Trans 2023; 52:11679-11690. [PMID: 37552495 PMCID: PMC10442743 DOI: 10.1039/d3dt02037a] [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: 06/29/2023] [Accepted: 07/28/2023] [Indexed: 08/09/2023]
Abstract
Ruthenium-based complexes have been suggested as promising anticancer drugs exhibiting reduced general toxicity compared to platinum-based drugs. In particular, Ru(η6-arene)(PTA)Cl2 (PTA = 1,3,5-triaza-7-phosphaadamantane), or RAPTA, complexes have demonstrated efficacy against breast cancer by suppressing metastasis, tumorigenicity, and inhibiting the replication of the human tumor suppressor gene BRCA1. However, RAPTA compounds have limited cytotoxicity, and therefore comparatively high doses are required. This study explores the activity of a series of RAPTA-like ruthenium(II) arene compounds against MCF-7 and MDA-MB-231 breast cancer cell lines and [Ru(η6-toluene)(PPh3)2Cl]+ was identified as a promising candidate. Notably, [Ru(η6-toluene)(PPh3)2Cl]Cl was found to be remarkably stable and highly cytotoxic, and selective to breast cancer cells. The minor groove of DNA was identified as a relevant target.
Collapse
Affiliation(s)
- Sarah A P Pereira
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, no 228, 4050-313 Porto, Portugal.
- Institut des Sciences et Ingénierie Chimiques, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| | - Jan Romano-deGea
- Institut des Sciences et Ingénierie Chimiques, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| | - Ana Isabel Barbosa
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, no 228, 4050-313 Porto, Portugal.
| | - Sofia A Costa Lima
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, no 228, 4050-313 Porto, Portugal.
| | - Paul J Dyson
- Institut des Sciences et Ingénierie Chimiques, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| | - M Lúcia M F S Saraiva
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira, no 228, 4050-313 Porto, Portugal.
| |
Collapse
|
6
|
Gonçalves YG, Becceneri AB, Graminha AE, Miranda VM, Rios RR, Rinaldi-Neto F, Costa MS, Gonçalves ACR, Deflon VM, Yoneyama KAG, Maia PIS, Franca EF, Cominetti MR, Silva RS, Von Poelhsitz G. New ruthenium(II) complexes with cyclic thio- and semicarbazone: evaluation of cytotoxicity and effects on cell migration and apoptosis of lung cancer cells. Dalton Trans 2023. [PMID: 37377063 DOI: 10.1039/d3dt00750b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
We describe the synthesis, physicochemical characterization, and in vitro antitumor assays of four novel analogous ruthenium(II) complexes with general formula cis-[RuII(N-L)(P-P)2]PF6, where P-P = bis(diphenylphosphine)methane (dppm, in complexes 1 and 2) or bis(diphenylphosphine)ethane (dppe, in complexes 3 and 4) and N-L = 5,6-diphenyl-4,5-dihydro-2H-[1,2,4]triazine-3-thione (Btsc, in complexes 1 and 3) or 5,6-diphenyltriazine-3-one (Bsc, in complexes 2 and 4). The data were consistent with cis arrangement of the biphosphine ligands. For the Btsc and Bsc ligands, the data pointed to monoanionic bidentate coordination to ruthenium(II) through N,S and N,O, respectively. Single-crystal X-ray diffraction showed that complex 1 crystallized in the monoclinic system, space group P21/c. Determination of the cytotoxicity profiles of complexes 1-4 gave SI values ranging from 1.19 to 3.50 against the human lung adenocarcinoma cell line A549 and the non-tumor lung cell line MRC-5. Although the molecular docking studies suggested that the interaction between DNA and complex 4 was energetically favorable, the experimental results showed that they interacted weakly. Overall, our results demonstrated that these novel ruthenium(II) complexes have interesting in vitro antitumor potential and this study may contribute to further studies in medicinal inorganic chemistry.
Collapse
Affiliation(s)
- Yasmim G Gonçalves
- Chemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil.
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Amanda B Becceneri
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Angelica E Graminha
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
- Gerontology Department, Federal University of São Carlos, São Carlos, SP, Brazil
| | - Victor M Miranda
- Grupo de Química Inorgânica Estrutural e Biológica, Chemistry Institute of São Carlos, Universidade de São Paulo, USP - São Carlos, São Carlos, SP, Brazil
| | - Rafaella R Rios
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Francisco Rinaldi-Neto
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Mônica S Costa
- Genetic and Biochemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Ana C R Gonçalves
- Exacts, Natural Sciences, and Education Institute, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Victor M Deflon
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Kelly A G Yoneyama
- Genetic and Biochemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Pedro I S Maia
- Exacts, Natural Sciences, and Education Institute, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Eduardo F Franca
- Laboratório de Cristalografia e Química Computacional, Chemistry Institute, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Márcia R Cominetti
- Gerontology Department, Federal University of São Carlos, São Carlos, SP, Brazil
| | - Roberto S Silva
- Laboratório de Fotoquímica e Química Bioinorgânica, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | |
Collapse
|
7
|
Persico M, Galatello P, Ferraro MG, Irace C, Piccolo M, Abduvakhidov A, Tkachuk O, d'Aulisio Garigliota ML, Campiglia P, Iannece P, Varra M, Ramunno A, Fattorusso C. Tetrasubstituted Pyrrole Derivative Mimetics of Protein-Protein Interaction Hot-Spot Residues: A Promising Class of Anticancer Agents Targeting Melanoma Cells. Molecules 2023; 28:molecules28104161. [PMID: 37241902 DOI: 10.3390/molecules28104161] [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: 03/29/2023] [Revised: 05/08/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
A new series of tetrasubstituted pyrrole derivatives (TSPs) was synthesized based on a previously developed hypothesis on their ability to mimic hydrophobic protein motifs. The resulting new TSPs were endowed with a significant toxicity against human epithelial melanoma A375 cells, showing IC50 values ranging from 10 to 27 μM, consistent with the IC50 value of the reference compound nutlin-3a (IC50 = 15 μM). In particular, compound 10a (IC50 = 10 μM) resulted as both the most soluble and active among the previous and present TSPs. The biological investigation evidenced that the anticancer activity is related to the activation of apoptotic cell-death pathways, supporting our rational design based on the ability of TSPs to interfere with PPI involved in the cell cycle regulation of cancer cells and, in particular, the p53 pathway. A reinvestigation of the TSP pharmacophore by using DFT calculations showed that the three aromatic substituents on the pyrrole core are able to mimic the hydrophobic side chains of the hot-spot residues of parallel and antiparallel coiled coil structures suggesting a possible molecular mechanism of action. A structure-activity relationship (SAR) analysis which includes solubility studies allows us to rationalize the role of the different substituents on the pyrrole core.
Collapse
Affiliation(s)
- Marco Persico
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Napoli, NA, Italy
| | - Paola Galatello
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84100 Fisciano, SA, Italy
| | - Maria Grazia Ferraro
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Napoli, NA, Italy
| | - Carlo Irace
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Napoli, NA, Italy
| | - Marialuisa Piccolo
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Napoli, NA, Italy
| | - Avazbek Abduvakhidov
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Napoli, NA, Italy
| | - Oleh Tkachuk
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Napoli, NA, Italy
| | | | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84100 Fisciano, SA, Italy
| | - Patrizia Iannece
- Department of Chemistry and Biology, University of Salerno, Via G. Paolo II 132, 84100 Fisciano, SA, Italy
| | - Michela Varra
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Napoli, NA, Italy
| | - Anna Ramunno
- Department of Pharmacy, University of Salerno, Via G. Paolo II 132, 84100 Fisciano, SA, Italy
| | - Caterina Fattorusso
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Napoli, NA, Italy
| |
Collapse
|
8
|
Ferraro MG, Bocchetti M, Riccardi C, Trifuoggi M, Paduano L, Montesarchio D, Misso G, Santamaria R, Piccolo M, Irace C. Triple Negative Breast Cancer Preclinical Therapeutic Management by a Cationic Ruthenium-Based Nucleolipid Nanosystem. Int J Mol Sci 2023; 24:ijms24076473. [PMID: 37047448 PMCID: PMC10094725 DOI: 10.3390/ijms24076473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023] Open
Abstract
Based on compelling preclinical evidence concerning the progress of our novel ruthenium-based metallotherapeutics, we are focusing research efforts on challenging indications for the treatment of invasive neoplasms such as the triple-negative breast cancer (TNBC). This malignancy mainly afflicts younger women, who are black, or who have a BRCA1 mutation. Because of faster growing and spreading, TNBC differs from other invasive breast cancers having fewer treatment options and worse prognosis, where existing therapies are mostly ineffective, resulting in a large unmet biomedical need. In this context, we benefited from an experimental model of TNBC both in vitro and in vivo to explore the effects of a biocompatible cationic liposomal nanoformulation, named HoThyRu/DOTAP, able to effectively deliver the antiproliferative ruthenium(III) complex AziRu, thus resulting in a prospective candidate drug. As part of the multitargeting mechanisms featuring metal-based therapeutics other than platinum-containing agents, we herein validate the potential of HoThyRu/DOTAP liposomes to act as a multimodal anticancer agent through inhibition of TNBC cell growth and proliferation, as well as migration and invasion. The here-obtained preclinical findings suggest a potential targeting of the complex pathways network controlling invasive and migratory cancer phenotypes. Overall, in the field of alternative chemotherapy to platinum-based drugs, these outcomes suggest prospective brand-new settings for the nanostructured AziRu complex to get promising goals for the treatment of metastatic TNBC.
Collapse
|
9
|
Riccardi C, Campanella A, Montesarchio D, Del Vecchio P, Oliva R, Paduano L. Investigating the Interaction of an Anticancer Nucleolipidic Ru(III) Complex with Human Serum Proteins: A Spectroscopic Study. Molecules 2023; 28:molecules28062800. [PMID: 36985771 PMCID: PMC10055563 DOI: 10.3390/molecules28062800] [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: 02/16/2023] [Revised: 03/09/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Ruthenium(III) complexes are very promising candidates as metal-based anticancer drugs, and several studies have supported the likely role of human serum proteins in the transport and selective delivery of Ru(III)-based compounds to tumor cells. Herein, the anticancer nanosystem composed of an amphiphilic nucleolipid incorporating a Ru(III) complex, which we named DoHuRu, embedded into the biocompatible cationic lipid DOTAP, was investigated as to its interaction with two human serum proteins thought to be involved in the mechanism of action of Ru(III)-based anticancer drugs, i.e., human serum albumin (HSA) and human transferrin (hTf). This nanosystem was studied in comparison with the simple Ru(III) complex named AziRu, a low molecular weight metal complex previously designed as an analogue of NAMI-A, decorated with the same ruthenium ligands as DoHuRu but devoid of the nucleolipid scaffold and not inserted in liposomal formulations. For this study, different spectroscopic techniques, i.e., Fluorescence Spectroscopy and Circular Dichroism (CD), were exploited, showing that DoHuRu/DOTAP liposomes can interact with both serum proteins without affecting their secondary structures.
Collapse
Affiliation(s)
- Claudia Riccardi
- Department of Chemical Sciences, University Federico II of Napoli, Via Cintia 21, 80126 Napoli, Italy
| | - Antonella Campanella
- Department of Chemical Sciences, University Federico II of Napoli, Via Cintia 21, 80126 Napoli, Italy
| | - Daniela Montesarchio
- Department of Chemical Sciences, University Federico II of Napoli, Via Cintia 21, 80126 Napoli, Italy
- CINMPIS-Consorzio Interuniversitario Nazionale di Ricerca in Metodologie e Processi Innovativi di Sintesi, Via E. Orabona 4, 70125 Bari, Italy
| | - Pompea Del Vecchio
- Department of Chemical Sciences, University Federico II of Napoli, Via Cintia 21, 80126 Napoli, Italy
| | - Rosario Oliva
- Department of Chemical Sciences, University Federico II of Napoli, Via Cintia 21, 80126 Napoli, Italy
| | - Luigi Paduano
- Department of Chemical Sciences, University Federico II of Napoli, Via Cintia 21, 80126 Napoli, Italy
- CSGI-Consorzio Interuniversitario per Lo Sviluppo dei Sistemi a Grande Interfase, Via della Lastruccia 3, 50019 Florence, Italy
| |
Collapse
|
10
|
Naskar N, Liu W, Qi H, Stumper A, Fischer S, Diemant T, Behm RJ, Kaiser U, Rau S, Weil T, Chakrabortty S. A Carbon Nanodot Based Near-Infrared Photosensitizer with a Protein-Ruthenium Shell for Low-Power Photodynamic Applications. ACS APPLIED MATERIALS & INTERFACES 2022; 14:48327-48340. [PMID: 36269223 DOI: 10.1021/acsami.2c08585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Near-infrared (NIR) light-activated photosensitization represents an encouraging therapeutic method in photodynamic therapy, especially for deep tissue penetration. In this context, two-photon activation, i.e., utilization of photons with relatively low energy but high photon flux for populating a virtual intermediate state leading to an excited state, is attractive. This concept would be highly advantageous in photodynamic therapy due to its minimal side effects. Herein, we propose that the combination of plasma protein serum albumin (HSA) containing several Ru complexes and NIR two-photon excitable carbon nanodots (Cdots), termed HSA-Ru-Cdots, provides several attractive features for enhancing singlet oxygen formation within the mitochondria of cancer cells stimulated by two-photon excitation in the NIR region. HSA-Ru-Cdot features biocompatibility, water solubility, and photostability as well as uptake into cancer cells with an endosomal release, which is an essential feature for subcellular targeting of mitochondria. The NIR two-photon excitation induced visible emission of the Cdots allows fluorescence resonance energy transfer (FRET) to excite the metal-to-ligand charge transfer of the Ru moiety, and fluorescence-lifetime imaging microscopy (FLIM) has been applied to demonstrate FRET within the cells. The NIR two-photon excitation is indirectly transferred to the Ru complexes, which leads to the production of singlet oxygen within the mitochondria of cancer cells. Consequently, we observe the destruction of filamentous mitochondrial structures into spheroid aggregates within various cancer cell lines. Cell death is induced by the long-wavelength NIR light irradiation at 810 nm with a low power density (7 mW/cm2), which could be attractive for phototherapy applications where deeper tissue penetration is crucial.
Collapse
Affiliation(s)
- Nilanjon Naskar
- Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, D-89081Ulm, Germany
| | - Weina Liu
- Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, D-89081Ulm, Germany
| | - Haoyuan Qi
- Central Facility for Materials Science Electron Microscopy, Ulm University, Albert-Einstein-Allee 11, D-89081Ulm, Germany
- Center for Advancing Electronics Dresden (cfaed) & Faculty of Chemistry and Food Chemistry, Dresden University of Technology, Mommsenstrasse 4, D-01062Dresden, Germany
| | - Anne Stumper
- Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, D-89081Ulm, Germany
| | - Stephan Fischer
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Albert-Einstein-Allee 11, D-89081Ulm, Germany
| | - Thomas Diemant
- Institute of Surface Chemistry and Catalysis, Ulm University, Albert-Einstein-Allee 47, D-89081Ulm, Germany
| | - R Jürgen Behm
- Institute of Surface Chemistry and Catalysis, Ulm University, Albert-Einstein-Allee 47, D-89081Ulm, Germany
| | - Ute Kaiser
- Central Facility for Materials Science Electron Microscopy, Ulm University, Albert-Einstein-Allee 11, D-89081Ulm, Germany
| | - Sven Rau
- Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, D-89081Ulm, Germany
| | - Tanja Weil
- Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, D-89081Ulm, Germany
| | - Sabyasachi Chakrabortty
- Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, D-89081Ulm, Germany
- Department of Chemistry, SRM University AP Andhra Pradesh, Andhra Pradesh522502, India
| |
Collapse
|
11
|
Riccardi C, Piccolo M, Ferraro MG, Graziano R, Musumeci D, Trifuoggi M, Irace C, Montesarchio D. Bioengineered lipophilic Ru(III) complexes as potential anticancer agents. BIOMATERIALS ADVANCES 2022; 139:213016. [PMID: 35882162 DOI: 10.1016/j.bioadv.2022.213016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/21/2022] [Accepted: 07/02/2022] [Indexed: 06/15/2023]
Abstract
Lipid-conjugated Ru(III) complexes - designed to obtain lipophilic analogues of the low molecular weight derivative AziRu, which is a NAMI-A-like anticancer agent - have been synthesized and fully characterized. A detailed biophysical investigation, including multiple, integrated techniques, allowed determining their molecular and self-assembling properties in aqueous solutions mimicking the extracellular environment, showing that our design produced a protective effect from hydrolysis of the Ru(III) complexes. In vitro biological experiments, carried out in comparison with AziRu, demonstrated that, among the novel lipophilic Ru(III) complexes synthesized, the compounds derivatized with palmitic and stearic acid, that we named PalmiPyRu and StePyRu respectively, showed attractive features and a promising antiproliferative activity, selective on specific breast cancer phenotypes. To get a deeper insight into their interactions with potential biomacromolecular targets, their ability to bind both bovine serum albumin (BSA), an abundant serum carrier protein, and some DNA model systems, including duplex and G-quadruplex structures, has been investigated by spectroscopic techniques. Inductively coupled plasma-mass spectrometry (ICP-MS) analysis of the ruthenium amount incorporated in human MCF-7 and MDA-MB-231 breast cancer cells, after incubation in parallel experiments with PalmiPyRu and AziRu, showed a markedly higher cell uptake of the lipophilic Ru(III) complex with respect to AziRu. These data confirmed that the proper lipidic tail decorating the metal complex not only favoured the formation of aggregates in the extracellular media but also improved their cell membrane penetration, thus leading to higher antiproliferative activity selective on breast cancer cells.
Collapse
Affiliation(s)
- Claudia Riccardi
- Department of Chemical Sciences, University of Naples "Federico II", Via Cintia 21, 80126 Naples, Italy
| | - Marialuisa Piccolo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy
| | - Maria Grazia Ferraro
- Department of Pharmacy, School of Medicine and Surgery, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy
| | - Raffaele Graziano
- Department of Chemical Sciences, University of Naples "Federico II", Via Cintia 21, 80126 Naples, Italy; Department of Pharmacy, School of Medicine and Surgery, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy
| | - Domenica Musumeci
- Department of Chemical Sciences, University of Naples "Federico II", Via Cintia 21, 80126 Naples, Italy; Institute of Biostructures and Bioimages, CNR, Naples, Italy
| | - Marco Trifuoggi
- Department of Chemical Sciences, University of Naples "Federico II", Via Cintia 21, 80126 Naples, Italy
| | - Carlo Irace
- Department of Pharmacy, School of Medicine and Surgery, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples "Federico II", Via Cintia 21, 80126 Naples, Italy.
| |
Collapse
|
12
|
Ferraro MG, Piccolo M, Misso G, Santamaria R, Irace C. Bioactivity and Development of Small Non-Platinum Metal-Based Chemotherapeutics. Pharmaceutics 2022; 14:pharmaceutics14050954. [PMID: 35631543 PMCID: PMC9147010 DOI: 10.3390/pharmaceutics14050954] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023] Open
Abstract
Countless expectations converge in the multidisciplinary endeavour for the search and development of effective and safe drugs in fighting cancer. Although they still embody a minority of the pharmacological agents currently in clinical use, metal-based complexes have great yet unexplored potential, which probably hides forthcoming anticancer drugs. Following the historical success of cisplatin and congeners, but also taking advantage of conventional chemotherapy limitations that emerged with applications in the clinic, the design and development of non-platinum metal-based chemotherapeutics, either as drugs or prodrugs, represents a rapidly evolving field wherein candidate compounds can be fine-tuned to access interactions with druggable biological targets. Moving in this direction, over the last few decades platinum family metals, e.g., ruthenium and palladium, have been largely proposed. Indeed, transition metals and molecular platforms where they originate are endowed with unique chemical and biological features based on, but not limited to, redox activity and coordination geometries, as well as ligand selection (including their inherent reactivity and bioactivity). Herein, current applications and progress in metal-based chemoth are reviewed. Converging on the recent literature, new attractive chemotherapeutics based on transition metals other than platinum—and their bioactivity and mechanisms of action—are examined and discussed. A special focus is committed to anticancer agents based on ruthenium, palladium, rhodium, and iridium, but also to gold derivatives, for which more experimental data are nowadays available. Next to platinum-based agents, ruthenium-based candidate drugs were the first to reach the stage of clinical evaluation in humans, opening new scenarios for the development of alternative chemotherapeutic options to treat cancer.
Collapse
Affiliation(s)
- Maria Grazia Ferraro
- BioChemLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (R.S.)
| | - Marialuisa Piccolo
- BioChemLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (R.S.)
| | - Gabriella Misso
- Department of Precision Medicine, School of Medicine and Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Correspondence: (G.M.); (C.I.)
| | - Rita Santamaria
- BioChemLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (R.S.)
| | - Carlo Irace
- BioChemLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (M.G.F.); (M.P.); (R.S.)
- Correspondence: (G.M.); (C.I.)
| |
Collapse
|
13
|
Khan AA, Ahmad R, Alanazi AM, Alsaif N, Abdullah M, Wani TA, Bhat MA. Determination of anticancer potential of a novel pharmacologically active thiosemicarbazone derivative in colorectal cancer cell lines. Saudi Pharm J 2022; 30:815-824. [PMID: 35812146 PMCID: PMC9257852 DOI: 10.1016/j.jsps.2022.03.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/18/2022] [Indexed: 12/16/2022] Open
Abstract
Thiosemicarbazones have received noteworthy attention due to their numerous pharmacological activities. Various thiosemicarbazone derivatives have been reported to play a key role as potential chemotherapeutic agents for the management of cancer. Herein, we aimed to establish the anticancer efficacy of novel thiosemicarbazone derivative C4 against colon cancer in vitro. The MTT viability assay identified C4 as a promising anticancer compound in a panel of cancer cell lines with the most potent activity against colon cancer cells. Further, anticancer potential of C4 was evaluated against HT-29 and SW620 colon cancer cell lines considering the factors like cell adhesion and migration, oxidative stress, cell cycle arrest, and apoptosis. Our results showed that C4 significantly inhibited the migration and adhesion of colon cancer cells. C4 significantly increased the intracellular reactive oxygen species (ROS) and induced apoptotic cell death. Cell cycle analysis revealed that C4 interfered in the cell cycle distribution and arrested the cells at the G2/M phase of the cell cycle. Consistent with these results C4 also down-regulated the Bcl-XL and Bcl-2 and up-regulated the caspase-3 expression. These findings introduced C4 as the potential anticancer agent against colon cancer.
Collapse
Affiliation(s)
- Azmat Ali Khan
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, P.O. Box 2457, Saudi Arabia
- Corresponding authors.
| | - Rehan Ahmad
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh 11451, P.O. Box 2457, Saudi Arabia
- Corresponding authors.
| | - Amer M. Alanazi
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, P.O. Box 2457, Saudi Arabia
| | - Nawaf Alsaif
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, P.O. Box 2457, Saudi Arabia
| | - Maha Abdullah
- Colorectal Research Chair, Department of Surgery, College of Medicine, King Saud University, Riyadh 11451, P.O. Box 2457, Saudi Arabia
| | - Tanveer A. Wani
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, P.O. Box 2457, Saudi Arabia
| | - Mashooq A. Bhat
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, P.O. Box 2457, Saudi Arabia
| |
Collapse
|
14
|
Nel J, Siniscalco D, Hognon C, Bouché M, Touche N, Brunner É, Gros PC, Monari A, Grandemange S, Francius G. Structural and morphological changes of breast cancer cells induced by iron(II) complexes. NANOSCALE 2022; 14:2735-2749. [PMID: 35112689 DOI: 10.1039/d1nr08301e] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Metal-based complexes are well-established cancer chemotherapeutic drug candidates. Although our knowledge regarding their exact activity vs. toxicity profile is incomplete, changes in cell membrane biophysical properties and cytoskeletal structures have been implicated as part of the mechanism of action. Thus, in this work, we characterised the effects of iron(II)-based complexes on the structural and morphological properties of epithelial non-tumorigenic (MCF 10A) and tumorigenic (MDA-MB-231) breast cell lines using atomic force microscopy (AFM), flow cytometry and immunofluorescence microscopy. At 24 h of exposure, both the MCF 10A and MDA-MB-231 cells experienced a cell softening, and an increase in size followed by a re-stiffening at 96 h. In addition, the triple negative breast cancer cell line, MDA-MB-231, sustained a notable cytoskeletal and mitochondrial reorganization with increased actin stress fibers and cell-to-cell communication structures. An extensive all-atom molecular dynamic simulation suggests a possible direct and unassisted internalization of the metallodrug candidate, and confirmed that the cellular effects could not be ascribed to the simple physical interaction of the iron-based complexes with the biological membrane. These observations provide an insight into a link between the mechanisms of action of such iron-based complexes as anti-cancer treatment and cytoskeletal architecture.
Collapse
Affiliation(s)
- Janske Nel
- Université de Lorraine, LIBio, F-54000, Nancy, France
| | - David Siniscalco
- Université de Lorraine and CNRS, LPCME UMR 7564, F-54000 Nancy, France.
| | - Cécilia Hognon
- Université de Lorraine and CNRS, LPCT UMR 7019, F-54000 Nancy, France.
| | - Mathilde Bouché
- Université de Lorraine and CNRS, L2CM UMR 7053, F-54000, Nancy, France
| | - Nadége Touche
- Université de Lorraine and CNRS, CRAN UMR 7039, F-54000 Nancy, France.
| | - Émilie Brunner
- Université de Lorraine and CNRS, CRAN UMR 7039, F-54000 Nancy, France.
| | - Philippe C Gros
- Université de Lorraine and CNRS, L2CM UMR 7053, F-54000, Nancy, France
| | - Antonio Monari
- Université de Lorraine and CNRS, LPCT UMR 7019, F-54000 Nancy, France.
- Université de Paris, ITODYS, CNRS, F-75006, Paris, France
| | | | - Grégory Francius
- Université de Lorraine and CNRS, LPCME UMR 7564, F-54000 Nancy, France.
| |
Collapse
|
15
|
Riccardi C, Napolitano F, Montesarchio D, Sampaolo S, Melone MAB. Nanoparticle-Guided Brain Drug Delivery: Expanding the Therapeutic Approach to Neurodegenerative Diseases. Pharmaceutics 2021; 13:1897. [PMID: 34834311 PMCID: PMC8623286 DOI: 10.3390/pharmaceutics13111897] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 10/31/2021] [Accepted: 11/04/2021] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases (NDs) represent a heterogeneous group of aging-related disorders featured by progressive impairment of motor and/or cognitive functions, often accompanied by psychiatric disorders. NDs are denoted as 'protein misfolding' diseases or proteinopathies, and are classified according to their known genetic mechanisms and/or the main protein involved in disease onset and progression. Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD) are included under this nosographic umbrella, sharing histopathologically salient features, including deposition of insoluble proteins, activation of glial cells, loss of neuronal cells and synaptic connectivity. To date, there are no effective cures or disease-modifying therapies for these NDs. Several compounds have not shown efficacy in clinical trials, since they generally fail to cross the blood-brain barrier (BBB), a tightly packed layer of endothelial cells that greatly limits the brain internalization of endogenous substances. By engineering materials of a size usually within 1-100 nm, nanotechnology offers an alternative approach for promising and innovative therapeutic solutions in NDs. Nanoparticles can cross the BBB and release active molecules at target sites in the brain, minimizing side effects. This review focuses on the state-of-the-art of nanoengineered delivery systems for brain targeting in the treatment of AD, PD and HD.
Collapse
Affiliation(s)
- Claudia Riccardi
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Naples, Italy; (C.R.); (D.M.)
| | - Filomena Napolitano
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, Via Sergio Pansini, 5, I-80131 Naples, Italy; (F.N.); (S.S.)
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Naples, Italy; (C.R.); (D.M.)
| | - Simone Sampaolo
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, Via Sergio Pansini, 5, I-80131 Naples, Italy; (F.N.); (S.S.)
| | - Mariarosa Anna Beatrice Melone
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, Via Sergio Pansini, 5, I-80131 Naples, Italy; (F.N.); (S.S.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA 19122-6078, USA
| |
Collapse
|
16
|
Synthesis and Characterization of Multifunctional Nanovesicles Composed of POPC Lipid Molecules for Nuclear Imaging. Molecules 2021; 26:molecules26216591. [PMID: 34770999 PMCID: PMC8587727 DOI: 10.3390/molecules26216591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/02/2022] Open
Abstract
The integration of nuclear imaging analysis with nanomedicine has tremendously grown and represents a valid and powerful tool for the development and clinical translation of drug delivery systems. Among the various types of nanostructures used as drug carriers, nanovesicles represent intriguing platforms due to their capability to entrap both lipophilic and hydrophilic agents, and their well-known biocompatibility and biodegradability. In this respect, here we present the development of a labelling procedure of POPC (1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine)-based liposomes incorporating an ad hoc designed lipophilic NOTA (1,4,7-triazacyclononane-1,4,7-triacetic acid) analogue, derivatized with an oleic acid residue, able to bind the positron emitter gallium-68(III). Based on POPC features, the optimal conditions for liposome labelling were studied with the aim of optimizing the Ga(III) incorporation and obtaining a significant radiochemical yield. The data presented in this work demonstrate the feasibility of the labelling procedure on POPC liposomes co-formulated with the ad hoc designed NOTA analogue. We thus provided a critical insight into the practical aspects of the development of vesicles for theranostic approaches, which in principle can be extended to other nanosystems exploiting a variety of bioconjugation protocols.
Collapse
|
17
|
Buommino E, De Marino S, Sciarretta M, Piccolo M, Festa C, D’Auria MV. Synergism of a Novel 1,2,4-oxadiazole-containing Derivative with Oxacillin against Methicillin-Resistant Staphylococcus aureus. Antibiotics (Basel) 2021; 10:antibiotics10101258. [PMID: 34680838 PMCID: PMC8532612 DOI: 10.3390/antibiotics10101258] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 11/25/2022] Open
Abstract
Staphylococcusaureus is an important opportunistic pathogen that causes many infections in humans and animals. The inappropriate use of antibiotics has favored the diffusion of methicillin-resistant S. aureus (MRSA), nullifying the efforts undertaken in the discovery of antimicrobial agents. Oxadiazole heterocycles represent privileged scaffolds for the development of new drugs because of their unique bioisosteric properties, easy synthesis, and therapeutic potential. A vast number of oxadiazole-containing derivatives have been discovered as potent antibacterial agents against multidrug-resistant MRSA strains. Here, we investigate the ability of a new library of oxadiazoles to contrast the growth of Gram-positive and Gram-negative strains. The strongest antimicrobial activity was obtained with compounds 3 (4 µM) and 12 (2 µM). Compound 12, selected for further evaluation, was found to be noncytotoxic on the HaCaT cell line up to 25 µM, bactericidal, and was able to improve the activity of oxacillin against the MRSA. The highest synergistic interaction was obtained with the combination values of 0.78 μM for compound 12, and 0.06 μg/mL for oxacillin. The FIC index value of 0.396 confirms the synergistic effect of compound 12 and oxacillin. MRSA treatment with compound 12 reduced the expression of genes included in the mec operon. In conclusion, 12 inhibited the growth of the MRSA and restored the activity of oxacillin, thus resulting in a promising compound in the treatment of MRSA infection.
Collapse
Affiliation(s)
| | | | | | | | - Carmen Festa
- Correspondence: ; Tel.: +39-081678987; Fax: +39-081678552
| | | |
Collapse
|
18
|
Safety and Efficacy Evaluation In Vivo of a Cationic Nucleolipid Nanosystem for the Nanodelivery of a Ruthenium(III) Complex with Superior Anticancer Bioactivity. Cancers (Basel) 2021; 13:cancers13205164. [PMID: 34680314 PMCID: PMC8534243 DOI: 10.3390/cancers13205164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022] Open
Abstract
Simple Summary The availability of selective, effective, and safe anticancer agents is a major challenge in the field of cancer research. As part of a multidisciplinary research project, in recent years our group has proposed an original class of nanomaterials for the delivery of new anticancer drugs based on ruthenium(III) complexes. In cellular models, these nanosystems have been shown to be effective in counteracting growth and proliferation of human breast cancer cells. Compared to conventional metallochemotherapeutics such as platinum-based agents whose clinical practice is associated with serious undesirable effects, ruthenium complexes share improved biochemical profiles making them more selective towards cancer cells and less cytotoxic to healthy cells. Their combination with biocompatible nanocarriers further enhances these promising features, as here showcased by our research carried out in an animal model which underscores the efficacy and safety in vivo of one of our most promising ruthenium-based nanosystems. Abstract Selectivity and efficacy towards target cancer cells, as well as biocompatibility, are current challenges of advanced chemotherapy powering the discovery of unconventional metal-based drugs and the search for novel therapeutic approaches. Among second-generation metal-based chemotherapeutics, ruthenium complexes have demonstrated promising anticancer activity coupled to minimal toxicity profiles and peculiar biochemical features. In this context, our research group has recently focused on a bioactive Ru(III) complex—named AziRu—incorporated into a suite of ad hoc designed nucleolipid nanosystems to ensure its chemical stability and delivery. Indeed, we proved that the structure and properties of decorated nucleolipids can have a major impact on the anticancer activity of the ruthenium core. Moving in this direction, here we describe a preclinical study performed by a mouse xenograft model of human breast cancer to establish safety and efficacy in vivo of a cationic Ru(III)-based nucleolipid formulation, named HoThyRu/DOTAP, endowed with superior antiproliferative activity. The results show a remarkable reduction in tumour with no evidence of animal suffering. Blood diagnostics, as well as biochemical analysis in both acute and chronic treated animal groups, demonstrate a good tolerability profile at the therapeutic regimen, with 100% of mice survival and no indication of toxicity. In addition, ruthenium plasma concentration analysis and tissue bioaccumulation were determined via appropriate sampling and ICP-MS analysis. Overall, this study supports both the efficacy of our Ru-containing nanosystem versus a human breast cancer model and its safety in vivo through well-tolerated animal biological responses, envisaging a possible forthcoming use in clinical trials.
Collapse
|
19
|
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: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
20
|
Ruthenium Complexes as Promising Candidates against Lung Cancer. Molecules 2021; 26:molecules26154389. [PMID: 34361543 PMCID: PMC8348655 DOI: 10.3390/molecules26154389] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 11/17/2022] Open
Abstract
Lung cancer is one of the most common malignancies with the highest mortality rate and the second-highest incidence rate after breast cancer, posing a serious threat to human health. The accidental discovery of the antitumor properties of cisplatin in the early 1960s aroused a growing interest in metal-based compounds for cancer treatment. However, the clinical application of cisplatin is limited by serious side effects and drug resistance. Therefore, other transition metal complexes have been developed for the treatment of different malignant cancers. Among them, Ru(II/III)-based complexes have emerged as promising anticancer drug candidates due to their potential anticancer properties and selective cytotoxic activity. In this review, we summarized the latest developments of Ru(II/III) complexes against lung cancer, focusing mainly on the mechanisms of their biological activities, including induction of apoptosis, necroptosis, autophagy, cell cycle arrest, inhibition of cell proliferation, and invasion and metastasis of lung cancer cells.
Collapse
|
21
|
Nayeem N, Contel M. Exploring the Potential of Metallodrugs as Chemotherapeutics for Triple Negative Breast Cancer. Chemistry 2021; 27:8891-8917. [PMID: 33857345 DOI: 10.1002/chem.202100438] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Indexed: 12/11/2022]
Abstract
This review focuses on studies of coordination and organometallic compounds as potential chemotherapeutics against triple negative breast cancer (TNBC) which has one of the poorest prognoses and worst survival rates from all breast cancer types. At present, chemotherapy is still the standard of care for TNBC since only one type of targeted therapy has been recently developed. References for metal-based compounds studied in TNBC cell lines will be listed, and those of metal-specific reviews, but a detailed overview will also be provided on compounds studied in vivo (mostly in mice models) and those compounds for which some preliminary mechanistic data was obtained (in TNBC cell lines and tumors) and/or for which bioactive ligands have been used. The main goal of this review is to highlight the most promising metal-based compounds with potential as chemotherapeutic agents in TNBC.
Collapse
Affiliation(s)
- Nazia Nayeem
- Brooklyn College Cancer Center BCCC-CURE, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York, 11210, USA.,Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York, 11210, USA.,Biology PhD Program, The Graduate Center, The City University of New York, 365 5th Avenue, New York, New York, 11006, USA
| | - Maria Contel
- Brooklyn College Cancer Center BCCC-CURE, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York, 11210, USA.,Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York, 11210, USA.,Biology PhD Program, The Graduate Center, The City University of New York, 365 5th Avenue, New York, New York, 11006, USA.,Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, 365 5th Avenue, New York, New York, 11006, USA.,University of Hawaii Cancer Center, 701 Ilalo St, Honolulu, Hawaii, 96813, USA
| |
Collapse
|
22
|
Hairat S, Zaki M. Half sandwiched RutheniumII complexes: En Route towards the targeted delivery by Human Serum Albumin (HSA). J Organomet Chem 2021. [DOI: 10.1016/j.jorganchem.2021.121732] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
23
|
Ferraro MG, Piccolo M, Pezzella A, Guerra F, Maione F, Tenore GC, Santamaria R, Irace C, Novellino E. Promelanogenic Effects by an Annurca Apple-Based Natural Formulation in Human Primary Melanocytes. Clin Cosmet Investig Dermatol 2021; 14:291-301. [PMID: 33790611 PMCID: PMC8008161 DOI: 10.2147/ccid.s299569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 02/25/2021] [Indexed: 01/21/2023]
Abstract
Introduction Melanocytes are engaged in synthesis, transport, and release of pigments at the epidermal-melanin units in response to the finely regulated melanogenic pathway. A multifaceted combination of both intrinsic and extrinsic factors – from endocrine and paracrine dynamics to exogenous stimuli such as sunlight and xenobiotics – modulates expression and activity of proteins involved in pigmentation, including the rate-limiting enzyme tyrosinase. As well as playing critical physiological functions comprising skin photoprotection, melanins define hair and skin pigmentation which in turn have impacted considerably to human social communication since time immemorial. Additionally, numerous skin diseases based on pigmentation alterations can have serious public influence. While several melanogenesis inhibitors are already available, the number of melanin activators and tyrosinase stimulators as drug-like agents is still limited. Methods To explore the biological effects of an Annurca Apple-based nutraceutical preparation (AMS) on melanin production, experiments in cellular models of human skin were performed. Both primary cultures and co-cultures of epidermal melanocytes (HEMa) and follicular keratinocytes (HHFK) were used. Results We show that AMS, by now branded for its cutaneous beneficial effects, induces in total biocompatibility a significant promelanogenic effect in human primary melanocytes. In line, we found melanin cytosolic accumulation consistent with tyrosinase up-regulation. Conclusion Disposal of skin pigmenting agents would be attractive for the treatment of hypopigmentation disorders, to postpone skin photoaging or simply for fashion, so that discovery and development of melanogenesis stimulators, especially from natural sources, is nowadays a dynamic area of research.
Collapse
Affiliation(s)
- Maria Grazia Ferraro
- Department of Pharmacy, School of Medicine and Surgery, University of Naples "Federico II", Naples, 80131, Italy
| | - Marialuisa Piccolo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples "Federico II", Naples, 80131, Italy
| | - Alessandro Pezzella
- Department of Chemical Sciences, University of Naples "Federico II", Naples, 80126, Italy
| | - Fabrizia Guerra
- Department of Pharmacy, School of Medicine and Surgery, University of Naples "Federico II", Naples, 80131, Italy
| | - Francesco Maione
- Department of Pharmacy, School of Medicine and Surgery, University of Naples "Federico II", Naples, 80131, Italy
| | - Gian Carlo Tenore
- Department of Pharmacy, School of Medicine and Surgery, University of Naples "Federico II", Naples, 80131, Italy
| | - Rita Santamaria
- Department of Pharmacy, School of Medicine and Surgery, University of Naples "Federico II", Naples, 80131, Italy
| | - Carlo Irace
- Department of Pharmacy, School of Medicine and Surgery, University of Naples "Federico II", Naples, 80131, Italy
| | - Ettore Novellino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples "Federico II", Naples, 80131, Italy
| |
Collapse
|
24
|
Hu Z, Fang W, Liu Y, Liang H, Chen W, Wang H. Acute glucose fluctuation promotes RAGE expression via reactive oxygen species‑mediated NF‑κB activation in rat podocytes. Mol Med Rep 2021; 23:330. [PMID: 33760170 PMCID: PMC7974412 DOI: 10.3892/mmr.2021.11969] [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: 10/26/2020] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetic nephropathy (DN) is a common chronic complication of diabetes, for which acute glucose fluctuation (AGF) is a potential risk factor. Fluctuating hyperglycemia has been confirmed to induce more serious kidney damage than hyperglycemia in diabetic rats; however, the mechanism remains unknown. The purpose of this study was to explore the potential role of AGF in the progression of DN. Viability of rat podocytes following 72-h AGF treatment was detected using Cell Counting-Kit-8. The rates of apoptosis and the level of reactive oxygen species (ROS) in rat podocytes were assessed by flow cytometry. Western blotting and reverse transcription-quantitative PCR were performed to measure relative protein and mRNA expression levels, respectively. Transfection with an mRFP-GFP-LC3 adenoviral vector was used to track autophagic flux under confocal microscopy. The results indicated that AGF could inhibit cell proliferation, promote TNF-α, interleukin-1β (IL-1β), and reactive oxygen species (ROS) generation, and increase autophagy in rat podocytes. Moreover, AGF upregulated receptor for advanced glycation end products (RAGE) expression via activation of NF-κB/p65 and IκBα. Pretreatment with 5 mM N-Acetyl-L-cysteine or 10 µM pyrrolidine dithiocarbamate effectively reduced cellular damage and inhibited activation of the NF-κB/RAGE signaling pathway. Thus, AGF induces rat podocyte injury by aggravating oxidative stress, promoting the inflammatory response, and regulating ROS-mediated NF-κB/RAGE activation.
Collapse
Affiliation(s)
- Zhangjie Hu
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Wenming Fang
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Yi Liu
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Haowei Liang
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Wei Chen
- Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Hui Wang
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
25
|
Silvestri S, Cirilli I, Marcheggiani F, Dludla P, Lupidi G, Pettinari R, Marchetti F, Di Nicola C, Falcioni G, Marchini C, Orlando P, Tiano L, Amici A. Evaluation of anticancer role of a novel ruthenium(II)-based compound compared with NAMI-A and cisplatin in impairing mitochondrial functionality and promoting oxidative stress in triple negative breast cancer models. Mitochondrion 2020; 56:25-34. [PMID: 33220497 DOI: 10.1016/j.mito.2020.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/22/2020] [Accepted: 11/02/2020] [Indexed: 01/18/2023]
Abstract
Platinum-based compounds are the most widely used anticancer drugs but, their elevated toxicity and chemoresistance has stimulated the study of others, such as ruthenium-based compounds. NAMI-A and UNICAM-1 were tested in vitro, comparing the mechanisms of toxicity, in terms of mitochondrial functionality and cellular oxidative stress. UNICAM-1, showed a clear mitochondrial target and a cytotoxic dose-dependent response thanks to its ability to promote an imbalance of cellular redox status. It impaired directly mitochondrial respiratory chain, promoting mitochondrial superoxide anion production, leading to mitochondrial membrane depolarization. All these aspects, could make UNICAM-1 a valid alternative for chemotherapy treatment of breast cancer.
Collapse
Affiliation(s)
- Sonia Silvestri
- Department of Life and Environmental Sciences, DISVA-Biochemistry, Polytechnic University of Marche, 60131 Ancona, Italy; Biomedfood srl, Ex-Spinoff of Polytechnic University of Marche, 60125 Ancona, Italy
| | - Ilenia Cirilli
- Department of Life and Environmental Sciences, DISVA-Biochemistry, Polytechnic University of Marche, 60131 Ancona, Italy; School of Pharmacy, University of Camerino, Camerino, MC, Italy
| | - Fabio Marcheggiani
- Department of Life and Environmental Sciences, DISVA-Biochemistry, Polytechnic University of Marche, 60131 Ancona, Italy
| | - Phiwayinkosi Dludla
- Department of Life and Environmental Sciences, DISVA-Biochemistry, Polytechnic University of Marche, 60131 Ancona, Italy; Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa
| | - Giulio Lupidi
- School of Pharmacy, University of Camerino, Camerino, MC, Italy
| | | | - Fabio Marchetti
- School of Sciences and Technology, University of Camerino, Camerino, MC, Italy
| | - Corrado Di Nicola
- School of Sciences and Technology, University of Camerino, Camerino, MC, Italy
| | | | - Cristina Marchini
- University of Camerino, via Gentile III da Varano, 62032 Camerino, Italy
| | - Patrick Orlando
- Department of Life and Environmental Sciences, DISVA-Biochemistry, Polytechnic University of Marche, 60131 Ancona, Italy.
| | - Luca Tiano
- Department of Life and Environmental Sciences, DISVA-Biochemistry, Polytechnic University of Marche, 60131 Ancona, Italy
| | - Augusto Amici
- University of Camerino, via Gentile III da Varano, 62032 Camerino, Italy
| |
Collapse
|
26
|
A photoactivatable Ru (II) complex bearing 2,9-diphenyl-1,10-phenanthroline: A potent chemotherapeutic drug inducing apoptosis in triple negative human breast adenocarcinoma cells. Chem Biol Interact 2020; 336:109317. [PMID: 33197429 DOI: 10.1016/j.cbi.2020.109317] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/23/2020] [Accepted: 11/06/2020] [Indexed: 12/26/2022]
Abstract
The photoactivatable Ru (II) complex 1 [Ru(bipy)2(dpphen)]Cl2 (where bipy = 2,2'-bipyridine and dpphen = 2,9-diphenyl-1,10-phenanthroline) has been shown to possess promising anticancer activity against triple negative adenocarcinoma MDA-MB-231 cells. The present study aims to elucidate the plausible mechanism of action of the photoactivatable complex 1 against MDA-MB-231 cells. Upon photoactivation, complex 1 exhibited time-dependent cytotoxic activity with a phototoxicity index (P Index) of >100 after 72 h. A significant increase in cell rounding and detachment, loss of membrane integrity, ROS accumulation and DNA damage was observed. Flow cytometry and a fluorescent apoptosis/necrosis assay showed an induction of cell apoptosis. Western blot analysis revealed the induction of intrinsic and extrinsic pathways and inhibition of the MAPK and PI3K pathways. The photoproduct of complex 1 showed similar effects on key apoptotic protein expression confirming that it is behind the observed cell death. In conclusion, the present study revealed that complex 1 is a potent multi-mechanistic photoactivatable chemotherapeutic drug that may serve as a potential lead molecule for targeted cancer chemotherapy.
Collapse
|
27
|
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: 4.5] [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
|
28
|
Gupta PK, Mishra L. Ecofriendly ruthenium-containing nanomaterials: synthesis, characterization, electrochemistry, bioactivity and catalysis. NANOSCALE ADVANCES 2020; 2:1774-1791. [PMID: 36132502 PMCID: PMC9418862 DOI: 10.1039/d0na00051e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/27/2020] [Indexed: 05/07/2023]
Abstract
Among transition metals, ruthenium being an in-demand element along with its complexes with multidimensional applications in biology, catalysis (especially photocatalysis), and several other aspects of industrial materials, is lacking regards for the potential aspect of its nanoparticles. In the modern synthetic scenario, green synthesis of novel ruthenium nanoparticles for the development of novel materials with potential applications has become a focus. Ru-containing nanomaterials (Ru-cNMs) combined with metals like platinum and palladium or with non-metals like phosphorus and oxygen have shown applications as an anticancer, antimicrobial, and antioxidant agents along with wide-ranging catalytic applications. Reduction of Ru salts using biomaterials including plants etc. has emerged enabling the synthesis of Ru-cNMs. In this context, authors realize that poor availability of literature in this area of research seems to be one of the major handicaps that perhaps could be limiting its attractiveness to researchers. Therefore, it was thought worthwhile to present a review article to encourage, guide, and facilitate scientific researches in green ruthenium nanochemistry embodying synthesis, characterization and biological as well as catalytic applications.
Collapse
Affiliation(s)
- Pranshu K Gupta
- Department of Chemistry, Institute of Science, Banaras Hindu University Varanasi-221005 India
| | - Lallan Mishra
- Department of Chemistry, Institute of Science, Banaras Hindu University Varanasi-221005 India
| |
Collapse
|
29
|
Rationally Designed Ruthenium Complexes for Breast Cancer Therapy. Molecules 2020; 25:molecules25020265. [PMID: 31936496 PMCID: PMC7024301 DOI: 10.3390/molecules25020265] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/31/2019] [Accepted: 01/01/2020] [Indexed: 12/11/2022] Open
Abstract
Since the discovery of the anticancer potential of ruthenium-based complexes, several species were reported as promising candidates for the treatment of breast cancer, which accounts for the greatest number of new cases in women every year worldwide. Among these ruthenium complexes, species containing bioactive ligand(s) have attracted increasing attention due to their potential multitargeting properties, leading to anticancer drug candidates with a broader range of cellular targets/modes of action. This review of the literature aims at providing an overview of the rationally designed ruthenium-based complexes that have been reported to date for which ligands were carefully selected for the treatment of hormone receptor positive breast cancers (estrogen receptor (ER+) or progesterone receptor (PR+)). In addition, this brief survey highlights some of the most successful examples of ruthenium complexes reported for the treatment of triple negative breast cancer (TNBC), a highly aggressive type of cancer, regardless of if their ligands are known to have the ability to achieve a specific biological function.
Collapse
|
30
|
Subasi E, Atalay EB, Erdogan D, Sen B, Pakyapan B, Kayali HA. Synthesis and characterization of thiosemicarbazone-functionalized organoruthenium (II)-arene complexes: Investigation of antitumor characteristics in colorectal cancer cell lines. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 106:110152. [DOI: 10.1016/j.msec.2019.110152] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 08/27/2019] [Accepted: 08/30/2019] [Indexed: 01/21/2023]
|
31
|
Bellavita R, Raucci F, Merlino F, Piccolo M, Ferraro MG, Irace C, Santamaria R, Iqbal AJ, Novellino E, Grieco P, Mascolo N, Maione F. Temporin L-derived peptide as a regulator of the acute inflammatory response in zymosan-induced peritonitis. Biomed Pharmacother 2019; 123:109788. [PMID: 31865142 DOI: 10.1016/j.biopha.2019.109788] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/08/2019] [Accepted: 12/09/2019] [Indexed: 01/29/2023] Open
Abstract
Antimicrobial peptides (AMPs) are an ancient group of defense molecules distributed in nature being found in mammals, birds, amphibians, insects, plants, and microorganisms. They display antimicrobial as well as immunomodulatory properties. The aim of this study was to investigate, for the first time, the anti-inflammatory activities of two synthetic temporin-L analogues (here named peptide 1 and 2) by an in vivo model of inflammation caused by intraperitoneal sub-lethal dose of zymosan. Our results show that peptide 1 and 2 exert anti-inflammatory activity in vivo in response to zymosan-induce peritonitis. Simultaneous administration of 10 mg/kg of both temporins, with a sub-lethal dose of zymosan (500 mg/kg), significantly rescued mice from the classical hallmarks of inflammation, including leukocyte infiltration and synthesis of inflammatory mediators including IL-6, TNF-α and MCP-1. More importantly, flow cytometry analysis highlighted a selective modulation of infiltrating inflammatory monocytes (defined as B220-/GR1hi-F480hi/CD115+) after peptide 2 treatment. Our results and presented models offer the possibility to test, in a preclinical setting, the potential of temporin analogues as anti-inflammatory agents.
Collapse
Affiliation(s)
- Rosa Bellavita
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Federica Raucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Francesco Merlino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Marialuisa Piccolo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Maria Grazia Ferraro
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Carlo Irace
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Rita Santamaria
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Asif J Iqbal
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Ettore Novellino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Paolo Grieco
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy.
| | - Nicola Mascolo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Francesco Maione
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy.
| |
Collapse
|
32
|
Fernandes AC. Synthesis, Biological Activity and Medicinal Applications of Ruthenium Complexes Containing Carbohydrate Ligands. Curr Med Chem 2019; 26:6412-6437. [DOI: 10.2174/0929867326666190124124350] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 10/15/2018] [Accepted: 12/11/2018] [Indexed: 12/24/2022]
Abstract
The search for new metal-efficient drugs has attracted considerable attention of the
scientific community. Among them, ruthenium complexes have emerged as an excellent alternative
of platinum complexes. This review presents a thorough and timely coverage of the synthesis,
biological activity and medicinal applications of ruthenium complexes bearing carbohydrate ligands,
allowing a large community of readers, in particularly the community that works in organic,
inorganic, bioorganometallic and medicinal chemistry, ready access to the most relevant examples.
Collapse
Affiliation(s)
- Ana Cristina Fernandes
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049- 001 Lisboa, Portugal
| |
Collapse
|
33
|
Induction of Hair Keratins Expression by an Annurca Apple-Based Nutraceutical Formulation in Human Follicular Cells. Nutrients 2019; 11:nu11123041. [PMID: 31847069 PMCID: PMC6950555 DOI: 10.3390/nu11123041] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/03/2019] [Accepted: 12/11/2019] [Indexed: 11/16/2022] Open
Abstract
Hair disorders may considerably impact the social and psychological well-being of an individual. Recent advances in the understanding the biology of hair have encouraged the research and development of novel and safer natural hair growth agents. In this context, we have previously demonstrated—at both preclinical and clinical level—that an Annurca apple-based dietary supplement (AMS), acting as a nutraceutical, is endowed with an intense hair-inductive activity (trichogenicity), at once increasing hair tropism and keratin content. Herein, in the framework of preclinical investigations, new experiments in primary human models of follicular keratinocytes and dermal papilla cells have been performed to give an insight around AMS biological effects on specific hair keratins expression. As well as confirming the biocompatibility and the antioxidant proprieties of our nutraceutical formulation, we have proven an engagement of trichokeratins production underlying its biological effects on human follicular cells. Annurca apples are particularly rich in oligomeric procyanidins, natural polyphenols belonging to the broader class of bioflavonoids believed to exert many beneficial health effects. To our knowledge, none of the current available remedies for hair loss has hitherto shown to stimulate the production of hair keratins so clearly.
Collapse
|
34
|
Riccardi C, Musumeci D, Trifuoggi M, Irace C, Paduano L, Montesarchio D. Anticancer Ruthenium(III) Complexes and Ru(III)-Containing Nanoformulations: An Update on the Mechanism of Action and Biological Activity. Pharmaceuticals (Basel) 2019; 12:E146. [PMID: 31561546 PMCID: PMC6958509 DOI: 10.3390/ph12040146] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 09/22/2019] [Accepted: 09/23/2019] [Indexed: 12/15/2022] Open
Abstract
The great advances in the studies on metal complexes for the treatment of different cancer forms, starting from the pioneering works on platinum derivatives, have fostered an increasingly growing interest in their properties and biomedical applications. Among the various metal-containing drugs investigated thus far, ruthenium(III) complexes have emerged for their selective cytotoxic activity in vitro and promising anticancer properties in vivo, also leading to a few candidates in advanced clinical trials. Aiming at addressing the solubility, stability and cellular uptake issues of low molecular weight Ru(III)-based compounds, some research groups have proposed the development of suitable drug delivery systems (e.g., taking advantage of nanoparticles, liposomes, etc.) able to enhance their activity compared to the naked drugs. This review highlights the unique role of Ru(III) complexes in the current panorama of anticancer agents, with particular emphasis on Ru-containing nanoformulations based on the incorporation of the Ru(III) complexes into suitable nanocarriers in order to enhance their bioavailability and pharmacokinetic properties. Preclinical evaluation of these nanoaggregates is discussed with a special focus on the investigation of their mechanism of action at a molecular level, highlighting their pharmacological potential in tumour disease models and value for biomedical applications.
Collapse
Affiliation(s)
- Claudia Riccardi
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Naples, Italy.
| | - Domenica Musumeci
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Naples, Italy.
| | - Marco Trifuoggi
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Naples, Italy.
| | - Carlo Irace
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, I-80131 Naples, Italy.
| | - Luigi Paduano
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Naples, Italy.
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Naples, Italy.
| |
Collapse
|
35
|
Halevas E, Mavroidi B, Swanson CH, Smith GC, Moschona A, Hadjispyrou S, Salifoglou A, Pantazaki AA, Pelecanou M, Litsardakis G. Magnetic cationic liposomal nanocarriers for the efficient drug delivery of a curcumin-based vanadium complex with anticancer potential. J Inorg Biochem 2019; 199:110778. [PMID: 31442839 DOI: 10.1016/j.jinorgbio.2019.110778] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/12/2019] [Accepted: 07/14/2019] [Indexed: 01/10/2023]
Abstract
In this work novel magnetic cationic liposomal nanoformulations were synthesized for the encapsulation of a crystallographically defined ternary V(IV)-curcumin-bipyridine (VCur) complex with proven bioactivity, as potential anticancer agents. The liposomal vesicles were produced via the thin film hydration method employing N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium (DOTAP) and egg phosphatidylcholine lipids and were magnetized through the addition of citric acid surface-modified monodispersed magnetite colloidal magnetic nanoparticles. The obtained nanoformulations were evaluated for their structural and textural properties and shown to have exceptional stability and enhanced solubility in physiological media, demonstrated by the entrapment efficiency and loading capacity results and the in vitro release studies of their cargo. Furthermore, the generated liposomal formulations preserved the superparamagnetic behavior of the employed magnetic core maintaining the physicochemical and morphological requirements for targeted drug delivery applications. The novel nanomaterials were further biologically evaluated for their DNA interaction potential and were found to act as intercalators. The findings suggest that the positively charged magnetic liposomal nanoformulations can generate increased concentration of their cargo at the DNA site, offering a further dimension in the importance of cationic liposomes as nanocarriers of hydrophobic anticancer metal ion complexes for the development of new multifunctional pharmaceutical nanomaterials with enhanced bioavailability and targeted antitumor activity.
Collapse
Affiliation(s)
- Eleftherios Halevas
- Laboratory of Materials for Electrotechnics, Department of Electrical and Computer Engineering, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece; Institute of Biosciences & Applications, National Centre for Scientific Research "Demokritos", 15310 Athens, Greece.
| | - Barbara Mavroidi
- Institute of Biosciences & Applications, National Centre for Scientific Research "Demokritos", 15310 Athens, Greece
| | - Claudia H Swanson
- Department of Natural Sciences, University of Chester, Thornton Science Park, Chester CH2 4NU, UK
| | - Graham C Smith
- Department of Natural Sciences, University of Chester, Thornton Science Park, Chester CH2 4NU, UK
| | - Alexandra Moschona
- Laboratory of Organic Chemistry, Department of Chemical Engineering, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Spyros Hadjispyrou
- Laboratory of Inorganic Chemistry, Department of Chemical Engineering, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Athanasios Salifoglou
- Laboratory of Inorganic Chemistry, Department of Chemical Engineering, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Anastasia A Pantazaki
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Maria Pelecanou
- Institute of Biosciences & Applications, National Centre for Scientific Research "Demokritos", 15310 Athens, Greece
| | - George Litsardakis
- Laboratory of Materials for Electrotechnics, Department of Electrical and Computer Engineering, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| |
Collapse
|
36
|
Mitochondria as playmakers of apoptosis, autophagy and senescence. Semin Cell Dev Biol 2019; 98:139-153. [PMID: 31154010 DOI: 10.1016/j.semcdb.2019.05.022] [Citation(s) in RCA: 300] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 05/21/2019] [Accepted: 05/22/2019] [Indexed: 12/16/2022]
Abstract
Mitochondria are the key energy-producing organelles and cellular source of reactive species. They are responsible for managing cell life and death by a balanced homeostasis passing through a network of structures, regulated principally via fission and fusion. Herein we discuss about the most advanced findings considering mitochondria as dynamic biophysical systems playing compelling roles in the regulation of energy metabolism in both physiologic and pathologic processes controlling cell death and survival. Precisely, we focus on the mitochondrial commitment to the onset, maintenance and counteraction of apoptosis, autophagy and senescence in the bioenergetic reprogramming of cancer cells. In this context, looking for a pharmacological manipulation of cell death processes as a successful route for future targeted therapies, there is major biotechnological challenge in underlining the location, function and molecular mechanism of mitochondrial proteins. Based on the critical role of mitochondrial functions for cellular health, a better knowledge of the main molecular players in mitochondria disfunction could be decisive for the therapeutical control of degenerative diseases, including cancer.
Collapse
|
37
|
Pelliccia S, Amato J, Capasso D, Di Gaetano S, Massarotti A, Piccolo M, Irace C, Tron GC, Pagano B, Randazzo A, Novellino E, Giustiniano M. Bio-Inspired Dual-Selective BCL-2/c-MYC G-Quadruplex Binders: Design, Synthesis, and Anticancer Activity of Drug-like Imidazo[2,1-i]purine Derivatives. J Med Chem 2019; 63:2035-2050. [DOI: 10.1021/acs.jmedchem.9b00262] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Sveva Pelliccia
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Jussara Amato
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Domenica Capasso
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Sonia Di Gaetano
- Institute of Biostructures and Bioimaging, CNR, Via Mezzocannone 16, 80134 Naples, Italy
| | - Alberto Massarotti
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Marialuisa Piccolo
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Carlo Irace
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Gian Cesare Tron
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Bruno Pagano
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Antonio Randazzo
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Ettore Novellino
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Mariateresa Giustiniano
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| |
Collapse
|
38
|
Luchini A, Vitiello G. Understanding the Nano-bio Interfaces: Lipid-Coatings for Inorganic Nanoparticles as Promising Strategy for Biomedical Applications. Front Chem 2019; 7:343. [PMID: 31165058 PMCID: PMC6534186 DOI: 10.3389/fchem.2019.00343] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/25/2019] [Indexed: 12/26/2022] Open
Abstract
Inorganic nanoparticles (NPs) exhibit relevant physical properties for application in biomedicine and specifically for both the diagnosis and therapy (i.e. theranostic) of severe pathologies, such as cancer. The inorganic NP core is often not stable in aqueous suspension and can induce cytotoxic effects. For this reason, over the years, several coating strategies were suggested to improve the NP stability in aqueous solutions as well as the NP biocompatibility. Among the various components which can be used for NP coatings, lipids, and in particular phospholipids emerged as versatile molecular building blocks for the production of NP coatings suitable for biomedical application. The recent synthetic efforts in NP lipid coatings allows today to introduce on the NP surface a large variety of lipid molecules eventually in mixture with amphiphilic or hydrophobic drugs or active molecules for cell targeting. In this review, the most relevant examples of NP lipid-coatings are presented and grouped in two main categories: supported lipid bilayers (SLB) and hybrid lipid bilayers (HLB). The discussed scientific cases take into account the most commonly used inorganic NP for biomedical applications in cancer therapy and diagnosis.
Collapse
Affiliation(s)
| | - Giuseppe Vitiello
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples, Italy
- CSGI, Center for Colloids and Surface Science, Sesto Fiorentino, Italy
| |
Collapse
|
39
|
Piccolo M, Misso G, Ferraro MG, Riccardi C, Capuozzo A, Zarone MR, Maione F, Trifuoggi M, Stiuso P, D'Errico G, Caraglia M, Paduano L, Montesarchio D, Irace C, Santamaria R. Exploring cellular uptake, accumulation and mechanism of action of a cationic Ru-based nanosystem in human preclinical models of breast cancer. Sci Rep 2019; 9:7006. [PMID: 31065032 PMCID: PMC6505035 DOI: 10.1038/s41598-019-43411-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 01/15/2019] [Indexed: 01/07/2023] Open
Abstract
According to WHO, breast cancer incidence is increasing so that the search for novel chemotherapeutic options is nowadays an essential requirement to fight neoplasm subtypes. By exploring new effective metal-based chemotherapeutic strategies, many ruthenium complexes have been recently proposed as antitumour drugs, showing ability to impact on diverse cellular targets. In the framework of different molecular pathways leading to cell death in human models of breast cancer, here we demonstrate autophagy involvement behind the antiproliferative action of a ruthenium(III)-complex incorporated into a cationic nanosystem (HoThyRu/DOTAP), proved to be hitherto one of the most effective within the suite of nucleolipidic formulations we have developed for the in vivo transport of anticancer ruthenium(III)-based drugs. Indeed, evidences are implicating autophagy in both cancer development and therapy, and anticancer interventions endowed with the ability to trigger this biological response are currently considered attractive oncotherapeutic approaches. Moreover, crosstalk between apoptosis and autophagy, regulated by finely tuned metallo-chemotherapeutics, may provide novel opportunities for future improvement of cancer treatment. Following this line, our in vitro and in vivo preclinical investigations suggest that an original strategy based on suitable formulations of ruthenium(III)-complexes, inducing sustained cell death, could open new opportunities for breast cancer treatment, including the highly aggressive triple-negative subtype.
Collapse
Affiliation(s)
- Marialuisa Piccolo
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131, Naples, Italy
| | - Gabriella Misso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138, Naples, Italy
| | - Maria Grazia Ferraro
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131, Naples, Italy
| | - Claudia Riccardi
- Department of Chemical Sciences, University of Naples "Federico II", Via Cintia 21, 80126, Naples, Italy
| | - Antonella Capuozzo
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131, Naples, Italy
| | - Mayra Rachele Zarone
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138, Naples, Italy
| | - Francesco Maione
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131, Naples, Italy
| | - Marco Trifuoggi
- Department of Chemical Sciences, University of Naples "Federico II", Via Cintia 21, 80126, Naples, Italy
| | - Paola Stiuso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138, Naples, Italy
| | - Gerardino D'Errico
- Department of Chemical Sciences, University of Naples "Federico II", Via Cintia 21, 80126, Naples, Italy
- CSGI - Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Florence, Via della Lastruccia 3, 50019, Sesto Fiorentino (FI), Italy
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138, Naples, Italy
| | - Luigi Paduano
- Department of Chemical Sciences, University of Naples "Federico II", Via Cintia 21, 80126, Naples, Italy.
- CSGI - Consorzio Sistemi a Grande Interfase, Department of Chemistry, University of Florence, Via della Lastruccia 3, 50019, Sesto Fiorentino (FI), Italy.
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples "Federico II", Via Cintia 21, 80126, Naples, Italy.
| | - Carlo Irace
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131, Naples, Italy.
| | - Rita Santamaria
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131, Naples, Italy
| |
Collapse
|
40
|
Naves MA, Graminha AE, Vegas LC, Luna-Dulcey L, Honorato J, Menezes ACS, Batista AA, Cominetti MR. Transport of the Ruthenium Complex [Ru(GA)(dppe)2]PF6 into Triple-Negative Breast Cancer Cells Is Facilitated by Transferrin Receptors. Mol Pharm 2019; 16:1167-1183. [DOI: 10.1021/acs.molpharmaceut.8b01154] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | | | | | | | | | - Antônio C. S. Menezes
- Campus of Exact Sciences and Technology (CCET), State University of Goiás, CEP 75132-903 Anápolis, GO, Brazil
| | | | | |
Collapse
|
41
|
Non-Platinum Metal Complexes as Potential Anti-Triple Negative Breast Cancer Agents. CRYSTALS 2018. [DOI: 10.3390/cryst8100369] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Breast cancer (BC) is the most common cancer in women worldwide, with a mortality rate that has been forecasted to rise in the next decade. This is especially worrying for people with triple-negative BC (TNBC), because of its unresponsiveness to current therapies. Different drugs to treat TNBC have been assessed, and, although platinum chemotherapy drugs seem to offer some hope, their drawbacks have motivated extensive investigations into alternative metal-based BC therapies. This paper aims to: (i) describe the preliminary in vitro and in vivo anticancer properties of non-platinum metal-based complexes (NPMBC) against TNBC; and (ii) analyze the likely molecular targets involved in their anticancer activity.
Collapse
|
42
|
Di Cesare Mannelli L, Piccolo M, Maione F, Ferraro MG, Irace C, De Feo V, Ghelardini C, Mascolo N. Tanshinones from Salvia miltiorrhiza Bunge revert chemotherapy-induced neuropathic pain and reduce glioblastoma cells malignancy. Biomed Pharmacother 2018; 105:1042-1049. [DOI: 10.1016/j.biopha.2018.06.047] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/12/2018] [Accepted: 06/12/2018] [Indexed: 12/28/2022] Open
|
43
|
Riccardi C, Fàbrega C, Grijalvo S, Vitiello G, D'Errico G, Eritja R, Montesarchio D. AS1411-decorated niosomes as effective nanocarriers for Ru(iii)-based drugs in anticancer strategies. J Mater Chem B 2018; 6:5368-5384. [PMID: 32254501 DOI: 10.1039/c8tb01563e] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Niosomes are self-assembled vesicles made up of single chain non-ionic surfactants combined with appropriate amounts of cholesterol or other lipids, exploited as carriers for hydrophilic or lipophilic drugs. Compared to liposomes, niosomes are typically more stable, less expensive and, being generally obtained from synthetic surfactants, more easily derivatizable, providing vesicular structures with a higher versatility and chemical diversity. Herein, we investigated the physico-chemical and biological properties of niosomes loaded with two active ingredients, i.e. the nucleolipidic Ru(iii)-complex HoThyRu, selected as an anticancer agent, and the nucleolin-targeting AS1411 aptamer, allowing selective recognition of cancer cells. The morphology, average size, zeta potential, electrophoretic mobility, and stability over time of the functionalized niosomes were analyzed using different biophysical techniques. These formulations, tested on both cancer and normal cells, showed promising antiproliferative activity on HeLa cells, with a higher efficacy associated with the nanosystems containing both AS1411 and HoThyRu with respect to the controls. In all the tested cell lines, AS1411 proved to markedly enhance the bioactivity of the Ru(iii)-containing niosomes.
Collapse
Affiliation(s)
- Claudia Riccardi
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126, Napoli, Italy.
| | | | | | | | | | | | | |
Collapse
|
44
|
Gouveia M, Figueira J, Jardim MG, Castro R, Tomás H, Rissanen K, Rodrigues J. Poly(alkylidenimine) Dendrimers Functionalized with the Organometallic Moiety [Ru(η⁵-C₅H₅)(PPh₃)₂]⁺ as Promising Drugs Against Cisplatin-Resistant Cancer Cells and Human Mesenchymal Stem Cells. Molecules 2018; 23:E1471. [PMID: 29914219 PMCID: PMC6100097 DOI: 10.3390/molecules23061471] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 06/11/2018] [Accepted: 06/12/2018] [Indexed: 02/05/2023] Open
Abstract
Here and for the first time, we show that the organometallic compound [Ru(η⁵-C₅H₅)(PPh₃)₂Cl] (RuCp) has potential to be used as a metallodrug in anticancer therapy, and further present a new approach for the cellular delivery of the [Ru(η⁵-C₅H₅)(PPh₃)₂]⁺ fragment via coordination on the periphery of low-generation poly(alkylidenimine) dendrimers through nitrile terminal groups. Importantly, both the RuCp and the dendrimers functionalized with [Ru(η⁵-C₅H₅)(PPh₃)₂]⁺ fragments present remarkable toxicity towards a wide set of cancer cells (Caco-2, MCF-7, CAL-72, and A2780 cells), including cisplatin-resistant human ovarian carcinoma cell lines (A2780cisR cells). Also, RuCp and the prepared metallodendrimers are active against human mesenchymal stem cells (hMSCs), which are often found in the tumor microenvironment where they seem to play a role in tumor progression and drug resistance.
Collapse
Affiliation(s)
- Marisol Gouveia
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390 Funchal, Portugal.
| | - João Figueira
- Department of Chemistry, ScilifeLab, Umeå University, KBC-Building, Linnaeus väg 6, 90736 Umeå, Sweden.
| | - Manuel G Jardim
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390 Funchal, Portugal.
| | - Rita Castro
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390 Funchal, Portugal.
| | - Helena Tomás
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390 Funchal, Portugal.
| | - Kari Rissanen
- Department of Chemistry, University of Jyvaskyla, P.O. Box. 35, FI-40014 Jyväskylä, Finland.
| | - João Rodrigues
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390 Funchal, Portugal.
- School of Materials Science and Engineering/Center for Nano Energy Materials, Northwestern Polytechnical University, Xi'an 710072, China.
| |
Collapse
|
45
|
Gouveia M, Figueira J, Jardim M, Castro R, Tomás H, Rissanen K, Rodrigues J. Poly(alkylidenimine) Dendrimers Functionalized with the Organometallic Moiety [Ru(η5-C5H5)(PPh3)2]+ as Promising Drugs Against Cisplatin-Resistant Cancer Cells and Human Mesenchymal Stem Cells. Molecules 2018. [DOI: https://doi.org/10.3390/molecules23061471] [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] Open
|
46
|
Russo G, Capuozzo A, Barbato F, Irace C, Santamaria R, Grumetto L. Cytotoxicity of seven bisphenol analogues compared to bisphenol A and relationships with membrane affinity data. CHEMOSPHERE 2018; 201:432-440. [PMID: 29529570 DOI: 10.1016/j.chemosphere.2018.03.014] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 02/15/2018] [Accepted: 03/03/2018] [Indexed: 05/25/2023]
Abstract
Bisphenol A (BPA) is a chemical used in numerous industrial applications. Due to its well ascertained toxicity as endocrine disruptor, industries have started to replace it with other bisphenols whose alleged greater safety is scarcely supported by literature studies. In this study, the toxicity of seven BPA analogues was evaluated using both in silico and in vitro techniques, as compared to BPA toxicity. Furthermore, their affinity indexes for phospholipids (i.e. phospholipophilicity) were determined by immobilized artificial membrane liquid chromatography (IAM-LC) and possible relationships with in vitro toxic activity were also investigated. The results on four different cell cultures yielded similar ranking of toxicity for the bisphenols considered, with IC50 values confirming their poor acute toxicity. As compared to BPA, bisphenol AF, bisphenol B, bisphenol M, and bisphenol A diglycidyl ether resulted more toxic, while bisphenol S, bisphenol F and bisphenol E were found as the less toxic congeners. These results are partly consistent with the scale of phospholipid affinity showing that toxicity increases at increasing membrane affinity. Therefore, phospholipophilicity determination can be assumed as a useful preliminary tool to select less toxic congeners to surrogate BPA in industrial applications.
Collapse
Affiliation(s)
- Giacomo Russo
- Pharm-Analysis & Bio-Pharm Laboratory - Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via D. Montesano, 49, I-80131 Naples, Italy
| | - Antonella Capuozzo
- BioChem Laboratory - Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via D. Montesano, 49, I-80131 Naples, Italy
| | - Francesco Barbato
- Pharm-Analysis & Bio-Pharm Laboratory - Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via D. Montesano, 49, I-80131 Naples, Italy
| | - Carlo Irace
- BioChem Laboratory - Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via D. Montesano, 49, I-80131 Naples, Italy
| | - Rita Santamaria
- BioChem Laboratory - Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via D. Montesano, 49, I-80131 Naples, Italy.
| | - Lucia Grumetto
- Pharm-Analysis & Bio-Pharm Laboratory - Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via D. Montesano, 49, I-80131 Naples, Italy; Consorzio Interuniversitario INBB, Viale Medaglie d'Oro, 305, I-00136 Rome, Italy.
| |
Collapse
|
47
|
Thota S, Rodrigues DA, Crans DC, Barreiro EJ. Ru(II) Compounds: Next-Generation Anticancer Metallotherapeutics? J Med Chem 2018; 61:5805-5821. [PMID: 29446940 DOI: 10.1021/acs.jmedchem.7b01689] [Citation(s) in RCA: 318] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Metal based therapeutics are a precious class of drugs in oncology research that include examples of theranostic drugs, which are active in both diagnostic, specifically imaging, and therapeutics applications. Ruthenium compounds have shown selective bioactivity and the ability to overcome the resistance that platinum-based therapeutics face, making them effective oncotherapeutic competitors in rational drug invention approaches. The development of antineoplastic ruthenium therapeutics is of particular interest because ruthenium containing complexes NAMI-A, KP1019, and KP1339 entered clinical trials and DW1/2 is in preclinical levels. The very robust, conformationally rigid organometallic Ru(II) compound DW1/2 is a protein kinase inhibitor and presents new Ru(II) compound designs as anticancer agents. Over the recent years, numerous strategies have been used to encapsulate Ru(II) derived compounds in a nanomaterial system, improving their targeting and delivery into neoplastic cells. A new photodynamic therapy based Ru(II) therapeutic, TLD-1433, has also entered clinical trials. Ru(II)-based compounds can also be photosensitizers for photodynamic therapy, which has proven to be an effective new, alternative, and noninvasive oncotherapy modality.
Collapse
Affiliation(s)
- Sreekanth Thota
- National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Fundação Oswaldo Cruz, Ministério da Saúde, Av. Brazil 4036, Prédio da Expansão, 8° Andar, Sala 814, Manguinhos , 21040-361 Rio de Janeiro , RJ , Brazil.,Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Institute of Biomedical Sciences , Federal University of Rio de Janeiro (UFRJ) , P.O. Box 68023, 21941-902 Rio de Janeiro , RJ , Brazil
| | - Daniel A Rodrigues
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Institute of Biomedical Sciences , Federal University of Rio de Janeiro (UFRJ) , P.O. Box 68023, 21941-902 Rio de Janeiro , RJ , Brazil
| | - Debbie C Crans
- Department of Chemistry , Colorado State University , Fort Collins , Colorado 80523 , United States
| | - Eliezer J Barreiro
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Institute of Biomedical Sciences , Federal University of Rio de Janeiro (UFRJ) , P.O. Box 68023, 21941-902 Rio de Janeiro , RJ , Brazil
| |
Collapse
|
48
|
Riccardi C, Musumeci D, Capuozzo A, Irace C, King S, Russo Krauss I, Paduano L, Montesarchio D. "Dressing up" an Old Drug: An Aminoacyl Lipid for the Functionalization of Ru(III)-Based Anticancer Agents. ACS Biomater Sci Eng 2017; 4:163-174. [PMID: 33418686 DOI: 10.1021/acsbiomaterials.7b00547] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In the search for more efficient anticancer treatments, Ru(III) complexes have attracted much interest among metal-based candidate drugs, showing marked antitumor and antimetastatic activity associated with lower systemic toxicity. Remarkable examples are the Ru(III) complexes NAMI-A and KP1019, which have reached advanced clinical evaluation. In order to improve the in vivo stability of Ru(III)-based drugs, as well as their cellular uptake and effectiveness, a new approach has been proposed by our research group, based on the incorporation of the active, NAMI-A-like Ru(III) complex into highly functionalized nucleolipidic structures, i.e., hybrid molecules containing a nucleoside or nucleotide central core derivatized with a lipid chain, ensuring both efficient protection against extracellular degradation and high cellular internalization of the metal. Aiming at expanding the chemical diversity of available amphiphilic Ru(III) complexes, we here selected a trifunctional α-amino acid to replace the nucleosidic core of previously prepared nucleolipid-based Ru(III) complexes. The amino acidic scaffold, linked to the Ru(III) complex, is decorated with both hydrophilic and lipophilic moieties, conferring high propensity to form stable aggregates in water, which is required to obtain a suitable nanocarrier for the drug delivery. Following this approach, a novel compound, indicated here as compound I, was successfully prepared and characterized, then studied in coformulation with the biocompatible cationic lipid 1,2-dioleyl-3-trimethylammoniumpropane chloride (DOTAP) by dynamic light scattering (DLS), small angle neutron scattering (SANS), and UV-vis analysis. Evaluated in vitro on a panel of human and nonhuman cell lines, it showed good antiproliferative activity on cancer cells, with IC50 values in the μM range, and no relevant cytotoxicity on the healthy cells used as control.
Collapse
Affiliation(s)
- Claudia Riccardi
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Via Cintia 21, 80126 Napoli, Italy
| | - Domenica Musumeci
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Via Cintia 21, 80126 Napoli, Italy.,CNR, Istituto di Biostrutture e Bioimmagini, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Antonella Capuozzo
- Dipartimento di Farmacia, Università di Napoli Federico II, Via D. Montesano 49, 80131 Napoli, Italy
| | - Carlo Irace
- Dipartimento di Farmacia, Università di Napoli Federico II, Via D. Montesano 49, 80131 Napoli, Italy
| | - Stephen King
- ISIS Facility, Science and Technology Facilities Council, Rutherford Appleton Laboratory, Harwell Campus, Didcot OX11 0QX, United Kingdom
| | - Irene Russo Krauss
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Via Cintia 21, 80126 Napoli, Italy.,CSGI, Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase, Via della Lastruccia 3, 50019 Sesto Fiorentino (Florence), Italy
| | - Luigi Paduano
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Via Cintia 21, 80126 Napoli, Italy.,CSGI, Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase, Via della Lastruccia 3, 50019 Sesto Fiorentino (Florence), Italy
| | - Daniela Montesarchio
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Via Cintia 21, 80126 Napoli, Italy.,Istituto per l'Endocrinologia e l'Oncologia "Gaetano Salvatore", Consiglio Nazionale delle Ricerche (CNR), Via Pansini 5, 80131 Napoli, Italy
| |
Collapse
|