1
|
D’Amato A, Mariconda A, Iacopetta D, Ceramella J, Catalano A, Sinicropi MS, Longo P. Complexes of Ruthenium(II) as Promising Dual-Active Agents against Cancer and Viral Infections. Pharmaceuticals (Basel) 2023; 16:1729. [PMID: 38139855 PMCID: PMC10747139 DOI: 10.3390/ph16121729] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Poor responses to medical care and the failure of pharmacological treatment for many high-frequency diseases, such as cancer and viral infections, have been widely documented. In this context, numerous metal-based substances, including cisplatin, auranofin, various gold metallodrugs, and ruthenium complexes, are under study as possible anticancer and antiviral agents. The two Ru(III) and Ru(II) complexes, namely, BOLD-100 and RAPTA-C, are presently being studied in a clinical trial and preclinical studies evaluation, respectively, as anticancer agents. Interestingly, BOLD-100 has also recently demonstrated antiviral activity against SARS-CoV-2, which is the virus responsible for the COVID-19 pandemic. Over the last years, much effort has been dedicated to discovering new dual anticancer-antiviral agents. Ru-based complexes could be very suitable in this respect. Thus, this review focuses on the most recent studies regarding newly synthesized Ru(II) complexes for use as anticancer and/or antiviral agents.
Collapse
Affiliation(s)
- Assunta D’Amato
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (A.D.); (P.L.)
| | | | - Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (D.I.); (J.C.); (M.S.S.)
| | - Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (D.I.); (J.C.); (M.S.S.)
| | - Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70126 Bari, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (D.I.); (J.C.); (M.S.S.)
| | - Pasquale Longo
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (A.D.); (P.L.)
| |
Collapse
|
2
|
Kavukcu S, Ensarioğlu HK, Karabıyık H, Vatansever HS, Türkmen H. Cell Death Mechanism of Organometallic Ruthenium(II) and Iridium(III) Arene Complexes on HepG2 and Vero Cells. ACS OMEGA 2023; 8:37549-37563. [PMID: 37841164 PMCID: PMC10569012 DOI: 10.1021/acsomega.3c05898] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023]
Abstract
Due to side effects and toxicity associated with platinum-derived metal-based drugs, extensive research has been conducted on ruthenium (Ru) complexes. We aim to synthesize a highly oil soluble Ru(II)-p-cymene complex (Ru1) with an aliphatic chain group, a bimetallic Ru(II)-p-cymene complex (Ru2) with N,S,S triple-coordination and a bimetallic Ir(III)-pentamethylcyclopentadienyl complex (Ir1) with S,S double-coordination. Subsequently, we investigate the effects of these complexes on Vero and HepG2 cell lines, focusing on cell death mechanisms. Characterization of the complexes is performed through nuclear magnetic resonance spectroscopy (1H and 13C NMR) and Fourier-transform infrared spectroscopy. The effective doses are determined using the (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide) (MTT) assay, applying different doses of the complexes to the two cell lines for 24 and 48 h, respectively. Immunoreactivities of Bax, Bcl2, caspase-3, RIP3, and RIPK1 are analyzed using the indirect immunoperoxidase technique. Notably, all the complexes (Ru1, Ru2, and Ir1) exhibit distinct cell death mechanisms, showing greater effectiveness than cisplatin. This study reveals the diverse mechanisms of action of Ru and Ir complexes based on different ligands. To the best of our knowledge, this study represents the first investigation of a novel RAED-type complex (Ru1) and unexpected bimetallic complexes (Ru2 and Ir1).
Collapse
Affiliation(s)
| | - Hilal Kabadayı Ensarioğlu
- Manisa
Celal Bayar University, Faculty of Medicine,
Department of Histology and Embryology, Manisa 45030, Turkey
| | - Hande Karabıyık
- Dokuz
Eylül University, Faculty of Science,
Department of Physics, Izmir 35390, Turkey
| | - Hafize Seda Vatansever
- Manisa
Celal Bayar University, Faculty of Medicine,
Department of Histology and Embryology, Manisa 45030, Turkey
- Near
East University, DESAM Institute, Mersin 10, Turkey 99138
| | - Hayati Türkmen
- Ege
University, Faculty of Science,
Department of Chemistry, Izmir 35100, Turkey
| |
Collapse
|
3
|
Bresciani G, Vančo J, Funaioli T, Zacchini S, Malina T, Pampaloni G, Dvořák Z, Trávníček Z, Marchetti F. Anticancer Potential of Diruthenium Complexes with Bridging Hydrocarbyl Ligands from Bioactive Alkynols. Inorg Chem 2023; 62:15875-15890. [PMID: 37713240 PMCID: PMC10548421 DOI: 10.1021/acs.inorgchem.3c01731] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Indexed: 09/16/2023]
Abstract
Diruthenacyclopentenone complexes of the general composition [Ru2Cp2(CO)2{μ-η1:η3-CH═C(C(OH)(R))C(═O)}] (2a-c; Cp = η5-C5H5) were synthesized in 94-96% yields from the reactions of [Ru2Cp2(CO)2{μ-η1:η3-C(Ph)═C(Ph)C(═O)}] (1) with 1-ethynylcyclopentanol, 17α-ethynylestradiol, and 17-ethynyltestosterone, respectively, in toluene at reflux. Protonation of 2a-c by HBF4 afforded the corresponding allenyl derivatives [Ru2Cp2(CO)3{μ-η1:η2-CH═C═R}]BF4 (3a-c) in 85-93% yields. All products were thoroughly characterized by elemental analysis, mass spectrometry, and IR, UV-vis, and nuclear magnetic resonance spectroscopy. Additionally, 2a and 3a were investigated by cyclic voltammetry, and the single-crystal diffraction method was employed to establish the X-ray structures of 2b and 3a. The cytotoxicity in vitro of 2b and 3a-c was evaluated against nine human cancer cell lines (A2780, A2780R, MCF-7, HOS, A549, PANC-1, Caco-2, PC-3, and HeLa), while the selectivity was assessed on normal human lung fibroblast (MRC-5). Overall, complexes exert stronger cytotoxicity than cisplatin, and 3b (comprising 17α-estradiol derived ligand) emerged as the best-performing complex. Inductively coupled plasma mass spectrometry cellular uptake studies in A2780 cells revealed a higher level of internalization for 3b and 3c compared to 2b, 3a, and the reference compound RAPTA-C. Experiments conducted on A2780 cells demonstrated a noteworthy impact of 3a and 3b on the cell cycle, leading to the majority of the cells being arrested in the G0/G1 phase. Moreover, 3a moderately induced apoptosis and oxidative stress, while 3b triggered autophagy and mitochondrial membrane potential depletion.
Collapse
Affiliation(s)
- Giulio Bresciani
- University
of Pisa, Dipartimento di Chimica e Chimica
Industriale, Via G. Moruzzi
13, I-56124 Pisa, Italy
| | - Ján Vančo
- Regional
Centre of Advanced Technologies and Materials, Czech Advanced Technology
and Research Institute, Palacký University, Šlechtitelů 27, CZ-779 00 Olomouc, Czech Republic
| | - Tiziana Funaioli
- University
of Pisa, Dipartimento di Chimica e Chimica
Industriale, Via G. Moruzzi
13, I-56124 Pisa, Italy
| | - Stefano Zacchini
- University
of Bologna, Dipartimento di Chimica Industriale
“Toso Montanari”, Viale del Risorgimento 4, I-40136 Bologna, Italy
| | - Tomáš Malina
- Regional
Centre of Advanced Technologies and Materials, Czech Advanced Technology
and Research Institute, Palacký University, Šlechtitelů 27, CZ-779 00 Olomouc, Czech Republic
| | - Guido Pampaloni
- University
of Pisa, Dipartimento di Chimica e Chimica
Industriale, Via G. Moruzzi
13, I-56124 Pisa, Italy
| | - Zdeněk Dvořák
- Department
of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, CZ-779
00 Olomouc, Czech
Republic
| | - Zdeněk Trávníček
- Regional
Centre of Advanced Technologies and Materials, Czech Advanced Technology
and Research Institute, Palacký University, Šlechtitelů 27, CZ-779 00 Olomouc, Czech Republic
| | - Fabio Marchetti
- University
of Pisa, Dipartimento di Chimica e Chimica
Industriale, Via G. Moruzzi
13, I-56124 Pisa, Italy
| |
Collapse
|
4
|
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
|
5
|
Bresciani G, Boni S, Funaioli T, Zacchini S, Pampaloni G, Busto N, Biver T, Marchetti F. Adding Diversity to a Diruthenium Biscyclopentadienyl Scaffold via Alkyne Incorporation: Synthesis and Biological Studies. Inorg Chem 2023; 62:12453-12467. [PMID: 37478132 PMCID: PMC10410612 DOI: 10.1021/acs.inorgchem.3c01644] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Indexed: 07/23/2023]
Abstract
We report the synthesis and the assessment of the anticancer potential of two series of diruthenium biscyclopentadienyl carbonyl complexes. Novel dimetallacyclopentenone compounds (2-4) were obtained (45-92% yields) from the thermal reaction (PhCCPh exchange) of [Ru2Cp2(CO)(μ-CO){μ-η1:η3-C(Ph)═C(Ph)C(═O)}], 1, with alkynes HCCR [R = C5H4FeCp (Fc), 3-C6H4(Asp), 2-naphthyl; Cp = η5-C5H5, Asp = OC(O)-2-C6H4C(O)Me]. Protonation of 1-3 by HBF4 afforded the corresponding μ-alkenyl derivatives 5-7, in 40-86% yields. All products were characterized by IR and NMR spectroscopy; moreover, cyclic voltammetry (1, 2, 5, 7) and single-crystal X-ray diffraction (5, 7) analyses were performed on representative compounds. Complexes 5-7 revealed a cytotoxic activity comparable to that of cisplatin in A549 (lung adenocarcinoma), SW480 (colon adenocarcinoma), and ovarian (A2780) cancer cell lines, and 2, 5, 6, and 7 overcame cisplatin resistance in A2780cis cells. Complexes 2, 5, and 7 (but not the aspirin derivative 6) induced an increase in intracellular ROS levels. Otherwise, 6 strongly stabilizes and elongates natural DNA (from calf thymus, CT-DNA), suggesting a possible intercalation binding mode, whereas 5 is less effective in binding CT-DNA, and 7 is ineffective. This trend is reversed concerning RNA, and in particular, 7 is able to bind poly(rA)poly(rU) showing selectivity for this nucleic acid. Complexes 5-7 can interact with the albumin protein with a thermodynamic signature dominated by hydrophobic interactions. Overall, we show that organometallic species based on the Ru2Cp2(CO)x scaffold (x = 2, 3) are active against cancer cells, with different incorporated fragments influencing the interactions with nucleic acids and the production of ROS.
Collapse
Affiliation(s)
- Giulio Bresciani
- University
of Pisa, Dipartimento di
Chimica e Chimica Industriale, Via G. Moruzzi 13, I-56124 Pisa, Italy
| | - Serena Boni
- University
of Pisa, Dipartimento di
Chimica e Chimica Industriale, Via G. Moruzzi 13, I-56124 Pisa, Italy
| | - Tiziana Funaioli
- University
of Pisa, Dipartimento di
Chimica e Chimica Industriale, Via G. Moruzzi 13, I-56124 Pisa, Italy
| | - Stefano Zacchini
- University
of Bologna, Dipartimento di
Chimica Industriale “Toso Montanari”, Viale del Risorgimento 4, I-40136 Bologna, Italy
| | - Guido Pampaloni
- University
of Pisa, Dipartimento di
Chimica e Chimica Industriale, Via G. Moruzzi 13, I-56124 Pisa, Italy
| | - Natalia Busto
- University
of Burgos, Departamento de
Química, Plaza
Misael Bañuelos s/n, 09001 Burgos, Spain
| | - Tarita Biver
- University
of Pisa, Dipartimento di
Chimica e Chimica Industriale, Via G. Moruzzi 13, I-56124 Pisa, Italy
| | - Fabio Marchetti
- University
of Pisa, Dipartimento di
Chimica e Chimica Industriale, Via G. Moruzzi 13, I-56124 Pisa, Italy
| |
Collapse
|
6
|
Lenis Rojas OA, Cordeiro S, Baptista PV, Fernandes AR. Half-sandwich Ru(II) N-heterocyclic carbene complexes in anticancer drug design. J Inorg Biochem 2023; 245:112255. [PMID: 37196411 DOI: 10.1016/j.jinorgbio.2023.112255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/10/2023] [Accepted: 05/10/2023] [Indexed: 05/19/2023]
Abstract
The ruthenium arene fragment is a rich source for the design of anticancer drugs; in this design, the co-ligand is a critical factor for obtaining effective anticancer complexes. In comparison with other types of ligands, N-heterocyclic carbenes (NHCs) have been less explored, despite the versatility in structural modifications and the marked stabilization of metal ions, being these characteristics important for the design of metal drugs. However, notable advances have been made in the development of NHC Ruthenium arene as anticancer agents. These advances include high antitumor activities, proven both in in vitro and in in vivo models and, in some cases, with marked selectivity against tumorigenic cells. The versatility of the structure has played a fundamental role, since they have allowed a selective interaction with their molecular targets through, for example, bio-conjugation with known anticancer molecules. For this reason, the structure-activity relationship of the imidazole, benzimidazole, and abnormal NHC ruthenium (II) η6-arene complexes have been studied. Taking into account this study, several synthetic aspects are provided to contribute to the next generations of this kind of complexes. Moreover, in recent years nanotechnology has provided innovative nanomedicines, where half-sandwich Ruthenium(II) complexes are paving their way. In this review, the recent developments in nanomaterials functionalized with Ruthenium complexes for targeted drug delivery to tumors will also be highlighted.
Collapse
Affiliation(s)
- Oscar A Lenis Rojas
- Instituto de Tecnologia Química e Biológica António Xavier, ITQB, Av. da República, EAN, 2780-157 Oeiras, Portugal.
| | - Sandra Cordeiro
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Pedro V Baptista
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Alexandra R Fernandes
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal.
| |
Collapse
|
7
|
Swaminathan S, Karvembu R. Dichloro Ru(II)- p-cymene-1,3,5-triaza-7-phosphaadamantane (RAPTA-C): A Case Study. ACS Pharmacol Transl Sci 2023; 6:982-996. [PMID: 37470017 PMCID: PMC10353064 DOI: 10.1021/acsptsci.3c00085] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Indexed: 07/21/2023]
Abstract
The use of organometallic compounds to treat various phenotypes of cancer has attracted increased interest in recent decades. Organometallic compounds, which are transitional between conventional inorganic and organic materials, have outstanding and one-of-a-kind features that offer fresh insight into the development of inorganic medicinal chemistry. The therapeutic potential of ruthenium(II)-arene RAPTA-type compounds is being thoroughly investigated, specifically owing to the excellent antimetastatic property of the initial candidate RAPTA-C. This review gives a thorough analysis of this complex and its evolution as a potential anticancer drug candidate. The numerous mechanistic investigations of RAPTA-C are discussed, and they are connected to the macroscopic biological characteristics that have been found. The "multitargeted" complex described here target enzymes, peptides, and intracellular proteins in addition to DNA that allow it to specifically target cancer cells. Understanding these may allow researchers to find specific targets and tune a new-generation organometallic complex accordingly.
Collapse
Affiliation(s)
- Srividya Swaminathan
- Department
of Chemistry, National Institute of Technology, Tiruchirappalli 620015, Tamil Nadu, India
- Center
for Computational Modeling, Chennai Institute
of Technology (CIT), Chennai 600069, India
| | - Ramasamy Karvembu
- Department
of Chemistry, National Institute of Technology, Tiruchirappalli 620015, Tamil Nadu, India
| |
Collapse
|
8
|
Kološa K, Žegura B, Štampar M, Filipič M, Novak M. Adverse Toxic Effects of Tyrosine Kinase Inhibitors on Non-Target Zebrafish Liver (ZFL) Cells. Int J Mol Sci 2023; 24:ijms24043894. [PMID: 36835302 PMCID: PMC9965539 DOI: 10.3390/ijms24043894] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023] Open
Abstract
Over the past 20 years, numerous tyrosine kinase inhibitors (TKIs) have been introduced for targeted therapy of various types of malignancies. Due to frequent and increasing use, leading to eventual excretion with body fluids, their residues have been found in hospital and household wastewaters as well as surface water. However, the effects of TKI residues in the environment on aquatic organisms are poorly described. In the present study, we investigated the cytotoxic and genotoxic effects of five selected TKIs, namely erlotinib (ERL), dasatinib (DAS), nilotinib (NIL), regorafenib (REG), and sorafenib (SOR), using the in vitro zebrafish liver cell (ZFL) model. Cytotoxicity was determined using the MTS assay and propidium iodide (PI) live/dead staining by flow cytometry. DAS, SOR, and REG decreased ZFL cell viability dose- and time-dependently, with DAS being the most cytotoxic TKI studied. ERL and NIL did not affect viability at concentrations up to their maximum solubility; however, NIL was the only TKI that significantly decreased the proportion of PI negative cells as determined by the flow cytometry. Cell cycle progression analyses showed that DAS, ERL, REG, and SOR caused the cell cycle arrest of ZFL cells in the G0/G1 phase, with a concomitant decrease of cells in the S-phase fraction. No data could be obtained for NIL due to severe DNA fragmentation. The genotoxic activity of the investigated TKIs was evaluated using comet and cytokinesis block micronucleus (CBMN) assays. The dose-dependent induction of DNA single strand breaks was induced by NIL (≥2 μM), DAS (≥0.006 μM), and REG (≥0.8 μM), with DAS being the most potent. None of the TKIs studied induced micronuclei formation. These results suggest that normal non-target fish liver cells are sensitive to the TKIs studied in a concentration range similar to those previously reported for human cancer cell lines. Although the TKI concentrations that induced adverse effects in exposed ZFL cells are several orders of magnitude higher than those currently expected in the aquatic environment, the observed DNA damage and cell cycle effects suggest that residues of TKIs in the environment may pose a hazard to non-intentionally exposed organisms living in environments contaminated with TKIs.
Collapse
Affiliation(s)
- Katja Kološa
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia
| | - Bojana Žegura
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia
- Jozef Stefan International Postgraduate School, 1000 Ljubljana, Slovenia
- Correspondence:
| | - Martina Štampar
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia
| | - Metka Filipič
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia
- Jozef Stefan International Postgraduate School, 1000 Ljubljana, Slovenia
| | - Matjaž Novak
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia
| |
Collapse
|
9
|
Dual drug delivery system of RAPTA-C and paclitaxel based on fructose coated nanoparticles for metastatic cancer treatment. Biochem Biophys Res Commun 2023; 640:134-141. [PMID: 36508926 DOI: 10.1016/j.bbrc.2022.12.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/22/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022]
Abstract
Ruthenium complexes have been widely studied as potential alternatives to platinum-type anticancer drugs due to their unique medical properties such as high selectivity, strong ability to inhibit solid tumour metastasis. However, non-specific biodistribution, and weak lethality of ruthenium to cancer cells limit its use in medical application. Drug delivery systems offer the ability to integrate multiple drugs in one system, which is particularly important to enhance the chemotherapeutic efficacy and to potentially achieve a synergistic effect of both drugs. Here, we report a dual drug nanocarrier that is based on a self-assembled biodegradable block copolymer, where the ruthenium complex (RAPTA-C) is chemically attached to the polymer chain, while another drug, paclitaxel (PTX), is entrapped in the core of the micelle. The dual drug delivery system was studied via in vitro tests using MDA-MB-231 breast cancer cells and it was observed that RAPTA-C in combination with PTX significantly enhanced anti-tumour and anti-metastasis activity.
Collapse
|
10
|
Mansouri F, Ortiz D, Dyson PJ. Competitive binding studies of the nucleosomal histone targeting drug, [Ru(η 6-p-cymene)Cl 2(pta)] (RAPTA-C), with oligonucleotide-peptide mixtures. J Inorg Biochem 2023; 238:112043. [PMID: 36370502 DOI: 10.1016/j.jinorgbio.2022.112043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/11/2022] [Accepted: 10/19/2022] [Indexed: 11/07/2022]
Abstract
Protein crystallography and biochemical assays reveal that the organometallic drug, [Ru(η6-p-cymene)Cl2(pta)] (RAPTA-C), preferentially binds to nucleosomal histone proteins in chromatin. To better understand the binding mechanism we report here a mass spectrometric-based competitive binding study between a model peptide from the acidic patch region of the H2A histone protein (the region where RAPTA-C is known to bind) and an oligonucleotide. In contrast to the protein crystallography and biochemical assays, RAPTA-C preferentially binds to the oligonucleotide, confirming that steric factors, rather than electronic effects, primarily dictate binding of RAPTA-C to histone proteins within the nucleosome.
Collapse
Affiliation(s)
- Farangis Mansouri
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne CH-1015, Switzerland; Department of Chemistry Institute for Advanced Studies in Basic Sciences (IASBS), 444 Prof. Sobouti Blvd., Gava Zang, Zanjan 45137-66731, Iran
| | - Daniel Ortiz
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne CH-1015, Switzerland
| | - Paul J Dyson
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne CH-1015, Switzerland.
| |
Collapse
|
11
|
Lenis-Rojas OA, Roma-Rodrigues C, Carvalho B, Cabezas-Sainz P, Fernández Vila S, Sánchez L, Baptista PV, Fernandes AR, Royo B. In Vitro and In Vivo Biological Activity of Ruthenium 1,10-Phenanthroline-5,6-dione Arene Complexes. Int J Mol Sci 2022; 23:13594. [PMID: 36362381 PMCID: PMC9656482 DOI: 10.3390/ijms232113594] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/18/2022] [Accepted: 11/02/2022] [Indexed: 12/28/2023] Open
Abstract
Ruthenium(II) arene complexes exhibit promising chemotherapeutic properties. In this study, the effect of the counter anion in Ru(II) complexes was evaluated by analyzing the biological effect of two Ru(II) p-cymene derivatives with the 1,10-phenanthroline-5,6-dione ligand of general-formula [(η6-arene)Ru(L)Cl][X] X = CF3SO3 (JHOR10) and PF6 (JHOR11). The biological activity of JHOR10 and JHOR11 was examined in the ovarian carcinoma cell line A2780, colorectal carcinoma cell line HCT116, doxorubicin-resistant HCT116 (HCT116-Dox) and in normal human dermal fibroblasts. Both complexes JHOR10 and JHOR11 displayed an antiproliferative effect on A2780 and HCT116 cell lines, and low cytotoxicity in fibroblasts. Interestingly, JHOR11 also showed antiproliferative activity in the HCT116-Dox cancer cell line, while JHOR10 was inactive. Studies in A2780 cells showed that JHOR11 induced the production of reactive oxygen species (ROS) that trigger autophagy and cellular senescence, but no apoptosis induction. Further analysis showed that JHOR11 presented no tumorigenicity, with no effect in the cellular mobility, as evaluated by thye wound scratch assay, and no anti- or pro-angiogenic effect, as evaluated by the ex-ovo chorioallantoic membrane (CAM) assay. Importantly, JHOR11 presented no toxicity in chicken and zebrafish embryos and reduced in vivo the proliferation of HCT116 injected into zebrafish embryos. These results show that these are suitable complexes for clinical applications with improved tumor cell cytotoxicity and low toxicity, and that counter-anion alteration might be a viable clinical strategy for improving chemotherapy outcomes in multidrug-resistant (MDR) tumors.
Collapse
Affiliation(s)
- Oscar A. Lenis-Rojas
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Catarina Roma-Rodrigues
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Beatriz Carvalho
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Pablo Cabezas-Sainz
- Departamento de Zoología Genética y Antropología Física, Facultad de Veterinaria, Universidad de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain
| | - Sabela Fernández Vila
- Departamento de Zoología Genética y Antropología Física, Facultad de Veterinaria, Universidad de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain
| | - Laura Sánchez
- Departamento de Zoología Genética y Antropología Física, Facultad de Veterinaria, Universidad de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain
- Preclinical Animal Models Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Pedro V. Baptista
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Alexandra R. Fernandes
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Beatriz Royo
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| |
Collapse
|
12
|
Highly Charged Ru(II) Polypyridyl Complexes as Photosensitizer Agents in Photodynamic Therapy of Epithelial Ovarian Cancer Cells. Int J Mol Sci 2022; 23:ijms232113302. [DOI: 10.3390/ijms232113302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/22/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Ovarian cancer recurrence is frequent and associated with chemoresistance, leading to extremely poor prognosis. Herein, we explored the potential anti-cancer effect of a series of highly charged Ru(II)-polypyridyl complexes as photosensitizers in photodynamic therapy (PDT), which were able to efficiently sensitize the formation of singlet oxygen upon irradiation (Ru12+ and Ru22+) and to produce reactive oxygen species (ROS) in their corresponding dinuclear metal complexes with the Fenton active Cu(II) ion/s ([CuRu1]4+ and [Cu2Ru2]6+). Their cytotoxic and anti-tumor effects were evaluated on human ovarian cancer A2780 cells both in the absence or presence of photoirradiation, respectively. All the compounds tested were well tolerated under dark conditions, whereas they switched to exert anti-tumor activity following photoirradiation. The specific effect was mediated by the onset of programed cell death, but only in the case of Ru12+ and Ru22+ was preceded by the loss of mitochondrial membrane potential soon after photoactivation and ROS production, thus supporting the occurrence of apoptosis via type II photochemical reactions. Thus, Ru(II)-polypyridyl-based photosensitizers represent challenging tools to be further investigated in the identification of new therapeutic approaches to overcome the innate chemoresistance to platinum derivatives of some ovarian epithelial cancers and to find innovative drugs for recurrent ovarian cancer.
Collapse
|
13
|
Beirne DF, Dalla Via M, Velasco-Torrijos T, Montagner D. Metal-Tyrosine Kinase Inhibitors: Targeted metal-drug conjugates. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
14
|
Iacopini D, Vančo J, Di Pietro S, Bordoni V, Zacchini S, Marchetti F, Dvořák Z, Malina T, Biancalana L, Trávníček Z, Di Bussolo V. New glycoconjugation strategies for Ruthenium(II) arene complexes via phosphane ligands and assessment of their antiproliferative activity. Bioorg Chem 2022; 126:105901. [DOI: 10.1016/j.bioorg.2022.105901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 11/29/2022]
|
15
|
Pete S, Roy N, Kar B, Paira P. Construction of homo and heteronuclear Ru(II), Ir(III) and Re(I) complexes for target specific cancer therapy. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214462] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
16
|
P K A, Roy N, Das U, Varddhan S, Sahoo SK, Paira P. [Ru(η 6- p-cymene)(N^O 8-hydroxyquinoline)(PTA)] complexes as rising stars in medicinal chemistry: synthesis, properties, biomolecular interactions, in vitro anti-tumor activity toward human brain carcinomas, and in vivo biodistribution and toxicity in a zebrafish model. Dalton Trans 2022; 51:8497-8509. [PMID: 35606053 DOI: 10.1039/d2dt00666a] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Herein, we have introduced a class of half-sandwich [Ru(η6-p-cymene)(N^O 8-hydroxyquinoline)(PTA)] complexes for brain cancer therapy. Among all the complexes, [RuL3PTA] and [RuL4PTA] exhibited excellent cytotoxicity profiles against T98G, LN229, and U87MG cancer cells. Notably, the antiproliferative activities of the relevant complexes were also supported by neurosphere, DNA intercalation, agarose gel electrophoresis, and time-dependent ROS detection assay studies. Detailed molecular assays were obtained via real-time reverse transcription (RT)-polymerase chain reaction (PCR) experiments. Moreover, the in vivo biodistribution of the [RuL4PTA] complex in different organs and the morphological patterns of zebrafish embryos due to toxic effects have been evaluated.
Collapse
Affiliation(s)
- Anuja P K
- Department of Chemistry, School of advanced sciences, Vellore Institute of Technology, Vellore-632014, Tamilnadu, India.
| | - Nilmadhab Roy
- Department of Chemistry, School of advanced sciences, Vellore Institute of Technology, Vellore-632014, Tamilnadu, India.
| | - Utpal Das
- Department of Chemistry, School of advanced sciences, Vellore Institute of Technology, Vellore-632014, Tamilnadu, India.
| | - Seshu Varddhan
- Department of Chemistry, School of advanced sciences, Vellore Institute of Technology, Vellore-632014, Tamilnadu, India.
| | - Suban K Sahoo
- Department of Chemistry, School of advanced sciences, Vellore Institute of Technology, Vellore-632014, Tamilnadu, India.
| | - Priyankar Paira
- Department of Chemistry, School of advanced sciences, Vellore Institute of Technology, Vellore-632014, Tamilnadu, India.
| |
Collapse
|
17
|
Swaminathan S, Haribabu J, Balakrishnan N, Vasanthakumar P, Karvembu R. Piano stool Ru(II)-arene complexes having three monodentate legs: A comprehensive review on their development as anticancer therapeutics over the past decade. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214403] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
18
|
Hildebrandt J, Häfner N, Kritsch D, Görls H, Dürst M, Runnebaum IB, Weigand W. Highly Cytotoxic Osmium(II) Compounds and Their Ruthenium(II) Analogues Targeting Ovarian Carcinoma Cell Lines and Evading Cisplatin Resistance Mechanisms. Int J Mol Sci 2022; 23:ijms23094976. [PMID: 35563367 PMCID: PMC9102668 DOI: 10.3390/ijms23094976] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 12/14/2022] Open
Abstract
(1) Background: Ruthenium and osmium complexes attract increasing interest as next generation anticancer drugs. Focusing on structure-activity-relationships of this class of compounds, we report on 17 different ruthenium(II) complexes and four promising osmium(II) analogues with cinnamic acid derivatives as O,S bidentate ligands. The aim of this study was to determine the anticancer activity and the ability to evade platin resistance mechanisms for these compounds. (2) Methods: Structural characterizations and stability determinations have been carried out with standard techniques, including NMR spectroscopy and X-ray crystallography. All complexes and single ligands have been tested for cytotoxic activity on two ovarian cancer cell lines (A2780, SKOV3) and their cisplatin-resistant isogenic cell cultures, a lung carcinoma cell line (A549) as well as selected compounds on three non-cancerous cell cultures in vitro. FACS analyses and histone γH2AX staining were carried out for cell cycle distribution and cell death or DNA damage analyses, respectively. (3) Results: IC50 values show promising results, specifically a high cancer selective cytotoxicity and evasion of resistance mechanisms for Ru(II) and Os(II) compounds. Histone γH2AX foci and FACS experiments validated the high cytotoxicity but revealed diminished DNA damage-inducing activity and an absence of cell cycle disturbance thus pointing to another mode of action. (4) Conclusion: Ru(II) and Os(II) compounds with O,S-bidentate ligands show high cytotoxicity without strong effects on DNA damage and cell cycle, and this seems to be the basis to circumvent resistance mechanisms and for the high cancer cell specificity.
Collapse
Affiliation(s)
- Jana Hildebrandt
- Institut für Anorganische und Analytische Chemie Friedrich-Schiller Universität Jena, Humboldtstraße 8, 07743 Jena, Germany; (J.H.); (H.G.)
- Department of Gynecology, Jena University Hospital—Friedrich-Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (N.H.); (D.K.); (M.D.)
| | - Norman Häfner
- Department of Gynecology, Jena University Hospital—Friedrich-Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (N.H.); (D.K.); (M.D.)
| | - Daniel Kritsch
- Department of Gynecology, Jena University Hospital—Friedrich-Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (N.H.); (D.K.); (M.D.)
| | - Helmar Görls
- Institut für Anorganische und Analytische Chemie Friedrich-Schiller Universität Jena, Humboldtstraße 8, 07743 Jena, Germany; (J.H.); (H.G.)
| | - Matthias Dürst
- Department of Gynecology, Jena University Hospital—Friedrich-Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (N.H.); (D.K.); (M.D.)
| | - Ingo B. Runnebaum
- Department of Gynecology, Jena University Hospital—Friedrich-Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (N.H.); (D.K.); (M.D.)
- Correspondence: (I.B.R.); (W.W.); Tel.: +49-3641-9329101 (I.B.R.); +49-3641-948160 (W.W.)
| | - Wolfgang Weigand
- Institut für Anorganische und Analytische Chemie Friedrich-Schiller Universität Jena, Humboldtstraße 8, 07743 Jena, Germany; (J.H.); (H.G.)
- Correspondence: (I.B.R.); (W.W.); Tel.: +49-3641-9329101 (I.B.R.); +49-3641-948160 (W.W.)
| |
Collapse
|
19
|
Fakhri S, Zachariah Moradi S, DeLiberto LK, Bishayee A. Cellular senescence signaling in cancer: A novel therapeutic target to combat human malignancies. Biochem Pharmacol 2022; 199:114989. [DOI: 10.1016/j.bcp.2022.114989] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 12/26/2022]
|
20
|
Sonkar C, Sarkar S, Mukhopadhyay S. Ruthenium(ii)-arene complexes as anti-metastatic agents, and related techniques. RSC Med Chem 2022; 13:22-38. [PMID: 35224494 PMCID: PMC8792825 DOI: 10.1039/d1md00220a] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/15/2021] [Indexed: 09/18/2023] Open
Abstract
With the discovery of cisplatin, a vast area of applications of metallodrugs in cancer treatment was opened but due to the side effects caused by the cisplatin complexes, researchers began to look for alternatives with similar anticancer properties but fewer side effects. Ruthenium was found to be a promising candidate, considering its significant anticancer properties and low side effects. Several ruthenium complexes, viz. NAMI-A, KP1019, KP1339, and TLD1433, have entered clinical trials. Some other arene ruthenium complexes such as RM175 and RAPTA-C have also entered clinical trials but very few of them have shown anti-metastatic properties. Herein, we provide information and probable mechanistic pathways for ruthenium(ii)-arene complexes that have been studied, so far, for their anti-metastatic activities. Also, we discuss the techniques and their significance for determining the anti-metastatic effects of the complexes.
Collapse
Affiliation(s)
- Chanchal Sonkar
- Department of Biosciences and Biomedical Engineering, School of Engineering, Indian Institute of Technology Indore Khandwa Road, Simrol Indore 453552 MP India
| | - Sayantan Sarkar
- Department of Chemistry, School of Basic Sciences, Indian Institute of Technology Indore Khandwa Road, Simrol Indore 453552 MP India
| | - Suman Mukhopadhyay
- Department of Biosciences and Biomedical Engineering, School of Engineering, Indian Institute of Technology Indore Khandwa Road, Simrol Indore 453552 MP India
- Department of Chemistry, School of Basic Sciences, Indian Institute of Technology Indore Khandwa Road, Simrol Indore 453552 MP India
| |
Collapse
|
21
|
Chakraborty A, Roy S, Chakraborty MP, Roy SS, Purkait K, Koley TS, Das R, Acharya M, Mukherjee A. Cytotoxic Ruthenium(II) Complexes of Pyrazolylbenzimidazole Ligands That Inhibit VEGFR2 Phosphorylation. Inorg Chem 2021; 60:18379-18394. [PMID: 34780170 DOI: 10.1021/acs.inorgchem.1c02979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Eight new ruthenium(II) complexes of N,N-chelating pyrazolylbenzimidazole ligands of the general formula [RuII(p-cym)(L)X]+ [where the ligand L is 2-(1H-pyrazol-1-yl)-1H-benzo[d]imidazole (L1) substituted at the 4 position of the pyrazole ring by Cl (L2), Br (L3), or I (L4) and X = Cl- and I-] were synthesized and characterized using various analytical techniques. Complexes 1 and 3 were also characterized by single-crystal X-ray crystallography, and they crystallized as a monoclinic crystal system in space groups P21/n and P21/c, respectively. The complexes display good solution stability at physiological pH 7.4. The iodido-coordinated pyrazolylbenzimidazole ruthenium(II) p-cymene complexes (2, 4, 6, and 8) are more resistant toward hydrolysis and have less tendency to form monoaquated complexes in comparison to their chlorido analogues (1, 3, 5, and 7). The halido-substituted 2-(1H-pyrazol-1-yl)-1H-benzo[d]imidazole ligands, designed as organic-directing molecules, inhibit vascular endothelial growth factor receptor 2 (VEGFR2) phosphorylation. In addition, the ruthenium(II) complexes display a potential to bind to DNA bases. The cytotoxicity profile of the complexes (IC50 ca. 9-12 μM for 4-8) against the triple-negative breast cancer cells (MDA-MB-231) show that most of the complexes are efficient. The lipophilicity and cellular accumulation data of the complexes show a good correlation with the cytotoxicity profile of 1-8. The representative complexes 3 and 7 demonstrate the capability of arresting the cell cycle in the G2/M phase and induce apoptosis. The inhibition of VEGFR2 phosphorylation with the representative ligands L2 and L4 and the corresponding metal complexes 3 and 7 in vitro shows that the organic-directing ligands and their complexes inhibit VEGFR2 phosphorylation. Besides, L2, L4, 3, and 7 inhibit the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) and proto-oncogene tyrosine-protein kinase (Src), capable of acting downstream of VEGFR2 as well as independently. Compounds L2, L4, 3, and 7 have a lesser effect on ERK1/2 and more prominently affect Src phosphorylation. We extended the study for L2 and 3 in the Tg(fli1:gfp) zebrafish model and found that L2 is more effective in vivo compared to 3 in inhibiting angiogenesis.
Collapse
|
22
|
Anti-Inflammatory Mechanisms of Novel Synthetic Ruthenium Compounds. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app112110092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Inflammation is the primary biological reaction to induce severe infection or injury in the immune system. Control of different inflammatory cytokines, such as nitric oxide (NO), interleukins (IL), tumor necrosis factor alpha-(TNF-α), noncytokine mediator, prostaglandin E2 (PGE2), mitogen activated protein kinases (MAPKs) and nuclear factor kappa B (NF-κB), facilitates anti-inflammatory effect of different substances. Coordination metal complexes have been applied as metallo-drugs. Several metal complexes have found to possess potent biological activities, especially anticancer, cardioprotective, chondroprotective and anti-parasitosis activities. Among the metallo drugs, ruthenium-based (Ru) complexes have paid much attention in clinical applications. Despite the kinetic nature of Ru complexes is similar to platinum in terms of cell division events, their toxic effect is lower than that of cisplatin. This paper reviews the anti-inflammatory effect of novel synthetic Ru complexes with potential molecular mechanisms that are actively involved.
Collapse
|
23
|
Rubio AR, González R, Busto N, Vaquero M, Iglesias AL, Jalón FA, Espino G, Rodríguez AM, García B, Manzano BR. Anticancer Activity of Half-Sandwich Ru, Rh and Ir Complexes with Chrysin Derived Ligands: Strong Effect of the Side Chain in the Ligand and Influence of the Metal. Pharmaceutics 2021; 13:1540. [PMID: 34683834 PMCID: PMC8537477 DOI: 10.3390/pharmaceutics13101540] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/09/2021] [Accepted: 09/17/2021] [Indexed: 11/16/2022] Open
Abstract
An important challenge in the field of anticancer chemotherapy is the search for new species to overcome the resistance of standard drugs. An interesting approach is to link bioactive ligands to metal fragments. In this work, we have synthesized a set of p-cymene-Ru or cyclopentadienyl-M (M = Rh, Ir) complexes with four chrysin-derived pro-ligands with different -OR substituents at position 7 of ring A. The introduction of a piperidine ring on chrysin led to the highly cytotoxic pro-ligand HL4 and its metal complexes L4-M (SW480 and A549 cell lines, cytotoxic order: L4-Ir > L4-Ru ≈ L4-Rh). HL4 and its complexes induce apoptosis and can overcome cis-platinum resistance. However, HL4 turns out to be more cytotoxic in healthy than in tumor cells in contrast to its metal complexes which displayed higher selectivity than cisplatin towards cancer cells. All L4-M complexes interact with double stranded DNA. Nonetheless, the influence of the metal is clear because only complex L4-Ir causes DNA cleavage, through the generation of highly reactive oxygen species (1O2). This result supports the hypothesis of a potential dual mechanism consisting of two different chemical pathways: DNA binding and ROS generation. This behavior provides this complex with a great effectivity in terms of cytotoxicity.
Collapse
Affiliation(s)
- Ana R. Rubio
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, Plaza Misael Bañuelos s/n, 09001 Burgos, Spain; (A.R.R.); (M.V.); (G.E.); (B.G.)
| | - Rocío González
- Facultad de Ciencias y Tecnologías Químicas-IRICA, Universidad de Castilla-La Mancha, Avda. C. J. Cela 10, 13071 Ciudad Real, Spain; (R.G.); (A.L.I.); (F.A.J.)
| | - Natalia Busto
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, Plaza Misael Bañuelos s/n, 09001 Burgos, Spain; (A.R.R.); (M.V.); (G.E.); (B.G.)
| | - Mónica Vaquero
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, Plaza Misael Bañuelos s/n, 09001 Burgos, Spain; (A.R.R.); (M.V.); (G.E.); (B.G.)
| | - Ana L. Iglesias
- Facultad de Ciencias y Tecnologías Químicas-IRICA, Universidad de Castilla-La Mancha, Avda. C. J. Cela 10, 13071 Ciudad Real, Spain; (R.G.); (A.L.I.); (F.A.J.)
- Facultad de Ciencias de la Ingeniería y Tecnología (FCITEC), Universidad Autónoma de Baja California, Blvd. Universitario # 1000, Unidad Valle de las Palmas, Baja California, Tijuana 21500, Mexico
| | - Félix A. Jalón
- Facultad de Ciencias y Tecnologías Químicas-IRICA, Universidad de Castilla-La Mancha, Avda. C. J. Cela 10, 13071 Ciudad Real, Spain; (R.G.); (A.L.I.); (F.A.J.)
| | - Gustavo Espino
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, Plaza Misael Bañuelos s/n, 09001 Burgos, Spain; (A.R.R.); (M.V.); (G.E.); (B.G.)
| | - Ana M. Rodríguez
- Departamento de Química Inorgánica, Orgánica y Bioquímica, Escuela Técnica Superior de Ingenieros Industriales, Universidad de Castilla-La Mancha, Avda. C. J. Cela 2, 13071 Ciudad Real, Spain;
| | - Begoña García
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, Plaza Misael Bañuelos s/n, 09001 Burgos, Spain; (A.R.R.); (M.V.); (G.E.); (B.G.)
| | - Blanca R. Manzano
- Facultad de Ciencias y Tecnologías Químicas-IRICA, Universidad de Castilla-La Mancha, Avda. C. J. Cela 10, 13071 Ciudad Real, Spain; (R.G.); (A.L.I.); (F.A.J.)
| |
Collapse
|
24
|
Hongthong K, Nhukeaw T, Temboot P, Dyson PJ, Ratanaphan A. Anticancer activity of RAPTA-EA1 in triple-negative BRCA1 proficient breast cancer cells: single and combined treatment with the PARP inhibitor olaparib. Heliyon 2021; 7:e07749. [PMID: 34430738 PMCID: PMC8371217 DOI: 10.1016/j.heliyon.2021.e07749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 06/08/2021] [Accepted: 08/06/2021] [Indexed: 11/26/2022] Open
Abstract
RAPTA-EA1 is a promising glutathione transferase (GSTP-1) inhibitor that has previously been shown to inhibit the growth of various breast cancer cells. We studied the anticancer activity of RAPTA-EA1 on triple-negative BRCA1 competent breast cancer MDA-MB-231 cells. MDA-MB-231 cells are significantly more sensitive to RAPTA-EA1 than MCF-7 cells. Treatment reveals a higher degree of cytotoxicity than cisplatin against both cell lines. Ruthenium accumulation in MDA-MB-231 cells is mainly in the nuclear fraction (43%), followed by the cytoplasm (30%), and the mitochondria (27%). RAPTA-EA1 blocks cell growth at the G2/M phase, leading to nuclear condensation and cell death. The compound slightly inhibits DNA replication of the 3,426-bp fragment of the BRCA1 exon 11 of the cells, with approximately 0.6 lesion per the BRCA1 fragment. The expression of BRCA1 mRNA and its protein in the Ru-treated cells is curtailed by 50–80% compared to the untreated controls. Growth inhibition of the triple-negative BRCA1 wild-type MDA-MB-231 and the sporadic BRCA1 wild-type MCF-7 cells by olaparib (a poly [ADP-ribose] polymerase (PARP) inhibitor) is dose-dependent, with MDA-MB-231 cells being two-fold less susceptible to the drug than MCF-7 cells. Combining olaparib with RAPTA-EA1 results in a combination index (CI) of 0.78 (almost additive) in MDA-MB-231 cells and 0.24 (potent synergy) in the MCF-7 cells. The PARP inhibitor alone differently regulates the expression of BRCA1 mRNA in both cell lines, whereas the olaparib-RAPTA-EA1 combination induces overexpression of BRCA1 mRNA in these cells. However, the expression level of the BRCA1 protein is dramatically reduced after treatment with the combined inhibitors, compared with the untreated controls. This observation highlights the cellular responses of triple-negative BRCA1 proficient breast cancer MDA-MB-231 cells to RAPTA-EA1 through BRCA1 inhibition and provides insights into alternative treatments for breast cancer.
Collapse
Affiliation(s)
- Khwanjira Hongthong
- Laboratory of Pharmaceutical Biotechnology, Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai, Songkhla 90112, Thailand
| | - Tidarat Nhukeaw
- Laboratory of Pharmaceutical Biotechnology, Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai, Songkhla 90112, Thailand
| | - Pornvichai Temboot
- Laboratory of Pharmaceutical Biotechnology, Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai, Songkhla 90112, Thailand
| | - Paul J Dyson
- Institute of Chemical Sciences, and Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Adisorn Ratanaphan
- Laboratory of Pharmaceutical Biotechnology, Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai, Songkhla 90112, Thailand
| |
Collapse
|
25
|
Oliveira KM, Peterson EJ, Carroccia MC, Cominetti MR, Deflon VM, Farrell NP, Batista AA, Correa RS. Ru(II)-Naphthoquinone complexes with high selectivity for triple-negative breast cancer. Dalton Trans 2021; 49:16193-16203. [PMID: 32329497 DOI: 10.1039/d0dt01091j] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Six new ruthenium(ii) complexes with lapachol (Lap) and lawsone (Law) with the general formula [Ru(L)(P-P)(bipy)]PF6, where L = Lap or Law, P-P = 1,2'-bis(diphenylphosphino)ethane (dppe), 1,4'-bis(diphenylphosphino)butane (dppb), 1,1'-bis(diphenylphosphino)ferrocene (dppf) and bipy = 2,2'-bipyridine, were synthesized, fully characterized by elemental analysis, molar conductivity, NMR, cyclic voltammetry, UV-vis, IR spectroscopies and three of them by X-ray crystallography. All six complexes were active against breast (MCF-7 and MDA-MB-231) and prostate (DU-145) cancer cell lines with lower IC50 values than cisplatin. Complex [Ru(Lap)(dppe)(bipy)]PF6 (1a) showed significant selectivity for MDA-MB-231, a model of triple-negative breast cancer (TNBC), as compared to the "normal-like" human breast epithelial cell line, MCF-10A. Complex (1a) inhibited TNBC colony formation and induced loss of cellular adhesion. Furthermore, the complex (1a) induced mitochondrial dysfunction and generation of ROS, as is involved in the apoptotic cell death pathway. Preferential cellular uptake of complex (1a) was observed in MDA-MB-231 cells compared to MCF-10A cells, consistent with the observed selectivity for tumorigenic vs. non-tumorigenic cells. Taken together, these results indicate that ruthenium complexes containing lapachol and lawsone as ligands are promising candidates as chemotherapeutic agents.
Collapse
Affiliation(s)
- Katia M Oliveira
- Departamento de Química, Universidade Federal de São Carlos - UFSCar, Rodovia Washington Luiz, KM 235 CP 676, CEP 13561-901, São Carlos, SP, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Drug Repurposing to Identify a Synergistic High-Order Drug Combination to Treat Sunitinib-Resistant Renal Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13163978. [PMID: 34439134 PMCID: PMC8391235 DOI: 10.3390/cancers13163978] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary In this study, drug combination screening was used to design a multidrug combination consisting of repurposed drugs to treat sunitinib-resistant clear cell renal cell carcinoma. In the frame of this project, the multidrug combination has been optimized and validated and an insight into the mechanism of action is given. The multidrug combinations significantly altered the transcription of genes related to apoptosis and metabolic pathways. Further analysis of the metabolism revealed strong upregulation of the presence of sphingolipids after multidrug combination treatment. Final evaluation for translation of the multidrug combination in ex vivo organoid-like cultures demonstrated significant anti-cancer efficacy. Abstract Repurposed drugs have been evaluated for the management of clear cell renal cell carcinoma (ccRCC), but only a few have influenced the overall survival of patients with advanced disease. To combine repurposed non-oncology with oncological drugs, we applied our validated phenotypic method, which consisted of a reduced experimental part and data modeling. A synergistic optimized multidrug combination (ODC) was identified to significantly reduce the energy levels in cancer remaining inactive in non-cancerous cells. The ODC consisted of Rapta-C, erlotinib, metformin and parthenolide and low doses. Molecular and functional analysis of ODC revealed a loss of adhesiveness and induction of apoptosis. Gene-expression network analysis displayed significant alterations in the cellular metabolism, confirmed by LC-MS based metabolomic analysis, highlighting significant changes in the lipid classes. We used heterotypic in vitro 3D co-cultures and ex vivo organoids to validate the activity of the ODC, maintaining an efficacy of over 70%. Our results show that repurposed drugs can be combined to target cancer cells selectively with prominent activity. The strong impact on cell adherence and metabolism indicates a favorable mechanism of action of the ODC to treat ccRCC.
Collapse
|
27
|
Synthesis and Antiparasitic Activity of New Conjugates—Organic Drugs Tethered to Trithiolato-Bridged Dinuclear Ruthenium(II)–Arene Complexes. INORGANICS 2021. [DOI: 10.3390/inorganics9080059] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Tethering known drugs to a metalorganic moiety is an efficient approach for modulating the anticancer, antibacterial, and antiparasitic activity of organometallic complexes. This study focused on the synthesis and evaluation of new dinuclear ruthenium(II)–arene compounds linked to several antimicrobial compounds such as dapsone, sulfamethoxazole, sulfadiazine, sulfadoxine, triclosan, metronidazole, ciprofloxacin, as well as menadione (a 1,4-naphtoquinone derivative). In a primary screen, 30 compounds (17 hybrid molecules, diruthenium intermediates, and antimicrobials) were assessed for in vitro activity against transgenic T. gondii tachyzoites constitutively expressing β-galactosidase (T. gondii β-gal) at 0.1 and 1 µM. In parallel, the cytotoxicity in noninfected host cells (human foreskin fibroblasts, HFF) was determined by an alamarBlue assay. When assessed at 1 µM, five compounds strongly impaired parasite proliferation by >90%, and HFF viability was retained at 50% or more, and they were further subjected to T. gondii β-gal dose-response studies. Two compounds, notably 11 and 13, amide and ester conjugates with sulfadoxine and metronidazole, exhibited low IC50 (half-maximal inhibitory concentration) values 0.063 and 0.152 µM, and low or intermediate impairment of HFF viability at 2.5 µM (83 and 64%). The nature of the anchored drug as well as that of the linking unit impacted the biological activity.
Collapse
|
28
|
Liang W, Shi J, Xia H, Wei X. A Novel Ruthenium-Fluvastatin Complex Downregulates SNCG Expression to Modulate Breast Carcinoma Cell Proliferation and Apoptosis via Activating the PI3K/Akt/mTOR/VEGF/MMP9 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5537737. [PMID: 34221232 PMCID: PMC8221895 DOI: 10.1155/2021/5537737] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/16/2021] [Accepted: 05/07/2021] [Indexed: 12/19/2022]
Abstract
Breast cancer is the most common cause of malignancy and cancer-related morbidity and death worldwide that requests effective and safe chemotherapy. Evaluation of metallodrug-based anticancer agents and statins as chemotherapeutics with fewer side effects is a largely unexplored research field. Synthesis and characterization of the ruthenium-fluvastatin complex were achieved using multiple spectroscopic techniques and thus further examined to evaluate its chemotherapeutic prospects in both MDA-MB-231 and MCF-7 cancer lines and eventually in vivo models of DMBA-induced mammary carcinogenesis in rodents. Our studies indicate that the metal and ligand chelation was materialized by the ligand's functional groups of carbonyl (=O) oxygen and hydroxyl (-OH), and the complex has been observed to be crystalline and able to chelate with CT-DNA. The complex was able to reduce cell proliferation and activate apoptotic events in breast carcinoma cell lines MCF-7 and MDA-MB-231. In addition, the complex was able to modify p53 expressions to interfere with apoptosis in the carcinoma of the breast, stimulated by the intrinsic apoptotic path assisted by Bcl2 and Bax in vivo, yet at the same point, controlling the PI3K/Akt/mTOR/VEGF pathway, as obtained from western blotting, correlates with the MMP9-regulated tumor mechanisms. Our research reveals that ruthenium-fluvastatin chemotherapy may disrupt, rescind, or interrupt breast carcinoma progression by modifying intrinsic apoptosis as well as the antiangiogenic cascade, thereby taking the role of a potential candidate in cancer therapy for the immediate future.
Collapse
Affiliation(s)
- Wei Liang
- Department of Oncology, Nanjing First Hospital Nanjing Medical University, Nanjing 210006, China
| | - Junfeng Shi
- Department of Oncology, Nanjing First Hospital Nanjing Medical University, Nanjing 210006, China
| | - Haiyan Xia
- Department of Oncology, Nanjing First Hospital Nanjing Medical University, Nanjing 210006, China
| | - Xiaowei Wei
- Department of Oncology, Nanjing First Hospital Nanjing Medical University, Nanjing 210006, China
| |
Collapse
|
29
|
Wu S, Wu Z, Ge Q, Zheng X, Yang Z. Antitumor activity of tridentate pincer and related metal complexes. Org Biomol Chem 2021; 19:5254-5273. [PMID: 34059868 DOI: 10.1039/d1ob00577d] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Pincer complexes featuring tunable tridentate ligand frameworks are one of the most actively studied classes of metal-based complexes. Currently, growing attention is devoted to the cytotoxicity of pincer and related metal complexes. The antiproliferative activity of numerous pincer complexes has been reported. Pincer tridentate ligand scaffolds show different coordination modes and offer multiple options for directed structural modifications. This review summarizes the significant progress in the research studies of the antitumor activity of pincer and related platinum(ii), gold(iii), palladium(ii), copper(ii), iron(iii), ruthenium(ii), nickel(ii) and some other metal complexes, in order to provide a reference for designing novel metal coordination drug candidates with promising antitumor activity.
Collapse
Affiliation(s)
- Shulei Wu
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, Affiliated Nanhua Hospital, University of South China, 28 Western Changsheng Road, Hengyang 421001, Hunan, PR China.
| | - Zaoduan Wu
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, Affiliated Nanhua Hospital, University of South China, 28 Western Changsheng Road, Hengyang 421001, Hunan, PR China.
| | - Qianyi Ge
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, Affiliated Nanhua Hospital, University of South China, 28 Western Changsheng Road, Hengyang 421001, Hunan, PR China.
| | - Xing Zheng
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, Affiliated Nanhua Hospital, University of South China, 28 Western Changsheng Road, Hengyang 421001, Hunan, PR China.
| | - Zehua Yang
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, Affiliated Nanhua Hospital, University of South China, 28 Western Changsheng Road, Hengyang 421001, Hunan, PR China.
| |
Collapse
|
30
|
Sathiya Kamatchi T, Mohamed Subarkhan MK, Ramesh R, Wang H, Małecki JG. Investigation into antiproliferative activity and apoptosis mechanism of new arene Ru(ii) carbazole-based hydrazone complexes. Dalton Trans 2021; 49:11385-11395. [PMID: 32776042 DOI: 10.1039/d0dt01476a] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ruthenium complexes with bioactive ligands are becoming promising substitutes for platinum complexes due to their precise action against various cancers. In the present study, the synthesis of three new arene Ru(ii) complexes containing new carbazole-based hydrazone ligands of general formula [(η6-benzene)Ru(L)Cl] (1-3; L = carbazolone benzhydrazone ligands), and their anticancer properties are described. The structural characterization of the ligands and their ruthenium complexes has been realized with the aid of elemental analysis, IR, UV-vis, NMR and HR-MS techniques. The molecular structures of all three complexes have been elucidated by single crystal X-ray crystallography and reveal the existence of pseudo-octahedral geometry around the ruthenium. The in vitro cancer cell growth inhibition property of the complexes against A549 (lung carcinoma), A2780 (ovarian adenocarcinoma) and non-cancerous 16HBE (human lung bronchial epithelium) cells were examined by MTT assay. All the complexes display good cytotoxicity towards both of these types of cancer cell compared to the standard drug cisplatin, with low IC50 values. Remarkably, complex 3, which contains an electron-donating substituent, induces a significant reduction of viability in A2780 cells. The inhibition capacity of the complexes towards A2780 cells proliferation was further confirmed using 5-ethynyl-2-deoxyuridine (EdU) assay via minimal DNA synthesis. The result of the acridine orange-ethidium bromide (AO-EB) fluorescent staining assay establishes that the cytotoxicity of the complexes was mediated by apoptosis in cancer cells. Furthermore, flow cytometry using Annexin V-FITC/propidium iodide (PI) double staining determines the quantitative discrimination of early apoptosis by the externalization of phosphatidylserine. In addition, cell cycle distribution indicates that the complexes block the cell cycle progression in the S-phase. The outcome of our investigation shows the promising scope and potency of tailored arene ruthenium complexes for precise cancer chemotherapy beyond platinum drugs.
Collapse
Affiliation(s)
- Thangavel Sathiya Kamatchi
- Centre for Organometallic Chemistry, School of Chemistry, Bharathidasan University, Tiruchirappalli-620 024, Tamil Nadu, India.
| | - Mohamed Kasim Mohamed Subarkhan
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, PR China
| | - Rengan Ramesh
- Centre for Organometallic Chemistry, School of Chemistry, Bharathidasan University, Tiruchirappalli-620 024, Tamil Nadu, India.
| | - Hangxiang Wang
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, PR China
| | - Jan Grzegorz Małecki
- Department of Crystallography, Institute of Chemistry, University of Silesia, 40-006, Katowice, Poland
| |
Collapse
|
31
|
Fibronectin 1: A Potential Biomarker for Ovarian Cancer. DISEASE MARKERS 2021; 2021:5561651. [PMID: 34093898 PMCID: PMC8164534 DOI: 10.1155/2021/5561651] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/26/2021] [Accepted: 05/11/2021] [Indexed: 11/17/2022]
Abstract
Methods OVCAR3 and A2780 are the two common cell lines that are used for ovarian cancer studies. The different invasion and migration abilities were observed by scratch tests and transwell experiments in our preliminary study. Gene chip was used to screen the expression gene in these two different cell lines, and then, the differentially expressed genes (at least 2-fold difference, P value < 0.05) were analyzed using KEGG. Result Fibronectin 1 (FN1) was found to be the most strongly correlated with the invasion and migration abilities of the OVCAR3 cells. Real-time PCR and FN1 knockout cell line was conducted and confirmed this finding. Based on the Oncomine database analysis, comparing with normal people, ovarian cancer patients exhibited high levels of FN1 expression. Additionally, higher FN1 expression was found in patients with higher FIGO stages of cancer. Conclusion FN1 could be a new biomarker for ovarian cancer detection and progress indicator.
Collapse
|
32
|
Balahbib A, El Omari N, Hachlafi NE, Lakhdar F, El Menyiy N, Salhi N, Mrabti HN, Bakrim S, Zengin G, Bouyahya A. Health beneficial and pharmacological properties of p-cymene. Food Chem Toxicol 2021; 153:112259. [PMID: 33984423 DOI: 10.1016/j.fct.2021.112259] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/28/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023]
Abstract
p-cymene also known as p-cymol or p-isopropyltoluene is an alkyl-substituted aromatic compound naturally occurring in essential oils (EOs) of various aromatic plants, including the genus of Artemisia, Protium, Origanum, and Thymus. It is related to the family of terpenes, especially monocyclic monoterpenes. p-cymene is also present in several food-based plants such as carrots, orange juice, grapefruit, tangerine, raspberries and several spices. Numerous studies have demonstrated the pharmacological properties of the monoterpenes p-cymene, including antioxidant, anti-inflammatory, antiparasitic, antidiabetic, antiviral, antitumor, antibacterial, and antifungal activities. The p-cymene has also been reported to act as an analgesic, antinociceptive, immunomodulatory, vasorelaxant and neuroprotective agent. Its anticancer effects are related to some mechanisms such as the inhibition of apoptosis and cell cycle arrest. In this review, we critically highlighted the in vitro and in vivo pharmacological properties of the p-cymene molecule, providing insight into its mechanisms of action and potential applications in drug discovery. In light of this finding, in-depth in vivo studies are strongly required to validate the safety and beneficial effects of the p-cymene molecule in human healthcare and industrial applications as a potential source of drug discovery.
Collapse
Affiliation(s)
- Abdelaali Balahbib
- Laboratory of Biodiversity, Ecology, and Genome, Faculty of Sciences, Mohammed V University in Rabat, Morocco
| | - Nasreddine El Omari
- Laboratory of Histology, Embryology, And Cytogenetic, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Naoufal El Hachlafi
- Microbial Biotechnology and Bioactive Molecules Laboratory, Sciences and Technologies Faculty, Sidi Mohmed Ben Abdellah University, P.O.Box-2002, Imouzzer Road Fez, Morocco
| | - Fatima Lakhdar
- Department of Biology, Laboratory of Marine Biotechnology and Environment, Faculty of Sciences, ChouaibDoukkali University, BP 20, El Jadida, 24000, Morocco
| | - Naoual El Menyiy
- Laboratory of Natural Substances, Pharmacology, Environment, Modeling, Health and of Life (SNAMOPEQ). Faculty of Sciences Dhar El Mahraz. University Sidi Mohamed Ben Abdellah, Fez, Morocco
| | - Najoua Salhi
- Laboratory of Pharmacology and Toxicology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Hanae Naceiri Mrabti
- Laboratory of Pharmacology and Toxicology, Bio Pharmaceutical and Toxicological Analysis Research Team, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, BP 6203, Rabat, Morocco
| | - Saad Bakrim
- Laboratory of Molecular Engineering, Valorization and Environment, Department of Sciences and Techniques, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Taroudant, Morocco
| | - Gokhan Zengin
- Physiology and Biochemistry Research Laboratory, Department of Biology, Science Faculty, Selcuk University, Konya, Turkey.
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, And Genomic Center of Human Pathologies, Mohammed V University in Rabat, Morocco.
| |
Collapse
|
33
|
Lenis-Rojas OA, Cabral R, Carvalho B, Friães S, Roma-Rodrigues C, Fernández JAA, Vila SF, Sanchez L, Gomes CSB, Fernandes AR, Royo B. Triazole-Based Half-Sandwich Ruthenium(II) Compounds: From In Vitro Antiproliferative Potential to In Vivo Toxicity Evaluation. Inorg Chem 2021; 60:8011-8026. [PMID: 33973771 DOI: 10.1021/acs.inorgchem.1c00527] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A new series of half-sandwich ruthenium(II) compounds [(η6-arene)Ru(L)Cl][CF3SO3] bearing 1,2,3-triazole ligands (arene = p-cymene, L = L1 (1); arene = p-cymene, L = L2 (2); arene = benzene, L = L1 (3); arene = benzene, L2 (4); L1 = 2-[1-(p-tolyl)-1H-1,2,3-triazol-4-yl]pyridine and L2 = 1,1'-di-p-tolyl-1H,1'H-4,4'-bi(1,2,3-triazole) have been synthesized and fully characterized by 1H and 13C NMR and IR spectroscopy, mass spectrometry, and elemental analysis. The molecular structures of 1, 2, and 4 have been determined by single-crystal X-ray diffraction. The cytotoxic activity of 1-4 was evaluated using the MTS assay against human tumor cells, namely ovarian carcinoma (A2780), colorectal carcinoma (HCT116), and colorectal carcinoma resistant to doxorubicin (HCT116dox), and against normal primary fibroblasts. Whereas compounds 2 and 4 showed no cytotoxic activity toward tumor cell lines, compounds 1 and 3 were active in A2780, while showing no antiproliferative effect in human normal dermal fibroblasts at the IC50 concentrations of the A2780 cell line. Exposure of ovarian carcinoma cells to IC50 concentrations of compound 1 or 3 led to an accumulation of reactive oxygen species and an increase of apoptotic and autophagic cells. While compound 3 displayed low levels of angiogenesis induction, compound 1 showed an ability to induce cell cycle delay and to interfere with cell migration. When the in vivo toxicity studies using zebrafish and chicken embryos are considered, compounds 1 and 3, which were not lethal, are promising candidates as anticancer agents against ovarian cancer due to their good cytotoxic activity in tumor cells and their low toxicity both in vitro and in vivo.
Collapse
Affiliation(s)
- Oscar A Lenis-Rojas
- Instituto de Tecnologia Química e Biológica António Xavier, ITQB NOVA, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Rui Cabral
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Beatriz Carvalho
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Sofia Friães
- Instituto de Tecnologia Química e Biológica António Xavier, ITQB NOVA, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Catarina Roma-Rodrigues
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Jhonathan A A Fernández
- Laboratory of Zebrafish, Department of Medical Genetics and Genomic Medicine- School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-887, Sao Paulo, Brazil.,Departamento de Zoología Genética y Antropología Física, Facultad de Veterinaria, Universidad de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain
| | - Sabela F Vila
- Departamento de Zoología Genética y Antropología Física, Facultad de Veterinaria, Universidad de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain
| | - Laura Sanchez
- Departamento de Zoología Genética y Antropología Física, Facultad de Veterinaria, Universidad de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain.,Preclinical Animal Models Group. Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, A Coruña, Spain
| | - Clara S B Gomes
- LAQV-REQUIMTE UCIBIO, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - Alexandra R Fernandes
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Beatriz Royo
- Instituto de Tecnologia Química e Biológica António Xavier, ITQB NOVA, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| |
Collapse
|
34
|
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]
|
35
|
|
36
|
Gopalakrishnan D, Saravanan S, Merckx R, Madan Kumar A, Khamrang T, Velusamy M, Vasanth K, Sunitha S, Hoogenboom R, Maji S, Ganeshpandian M. N, N-Ru(II)- p-cymene-poly( N-vinylpyrrolidone) surface functionalized gold nanoparticles: from organoruthenium complex to nanomaterial for antiproliferative activity. Dalton Trans 2021; 50:8232-8242. [PMID: 34037018 DOI: 10.1039/d1dt00694k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Organometallic Ru-arene complexes are promising as anticancer agents, but the lack of tumor uptake and poor solubility in the physiological medium impede their development. In order to deal with these challenges, we developed gold nanoparticles coated with Ru(arene)-functionalized PNVP-Py, where PNVP-Py is pyridine end-functionalized poly(N-vinylpyrrolidone). It is demonstrated that these particles exhibit higher anti-proliferative activity than the individual organometallic ruthenium(ii) complex of the type [Ru(η6-p-cymene)(NN)Cl]PF6, where NN is bis(4-methoxyphenylimino)acenaphthene, against colorectal adenocarcinoma cell lines. More specifically, a RuII(η6-p-cymene) complex containing a NN bidentate ligand has been prepared and characterized by spectral studies and X-ray crystallography. To tether the isolated complex onto the surface of the AuNPs, PNVP-Py, which contains a pyridine group at one end to coordinate to the Ru-complex and a suitable functional group at the other end to bind on the surface of the AuNPs, has been prepared and utilized to obtain the macromolecular complex [Ru(η6-p-cymene)(NN)(PNVP-Py)]Cl2. Next, stable Ru(p-cym)(NN)(PNVP-Py)@AuNPs were obtained via a ligand exchange reaction of citrate-stabilized AuNPs with a macromolecular complex by a direct 'grafting to' approach and characterized well. Despite the lower DNA cleavage activity, the nanoconjugate exhibits better cytotoxicity than the individual complex against HT-29 colorectal adenocarcinoma cells on account of its enhanced permeability across the cell membrane. The AO/EB staining assay revealed that the nanoconjugate is able to induce an apoptotic mode of cell death, which was further quantitatively evaluated by Annexin V-FITC/PI double assay. An immunofluorescence assay indicated the higher potency of the nanoconjugate to inhibit cyclin D1 gene expression that is required for cancer cell growth. To the best of our knowledge, this is the first report of the modification of an organometallic Ru(arene) complex into a Ru(arene)metallopolymer-gold nanoconjugate for the development of ruthenium-based nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Durairaj Gopalakrishnan
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu, Chennai, TN, India.
| | - S Saravanan
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu, Chennai, TN, India.
| | - Ronald Merckx
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281 S4, Ghent, Belgium
| | - Arumugam Madan Kumar
- Department of Chemistry, Sathyabama Institute of Science and Technology, Chennai - 600119, India
| | - Themmila Khamrang
- Department of Chemistry, C. I. College, Bishnupur 795126, Manipur, India
| | - Marappan Velusamy
- Department of Chemistry, North Eastern Hill University, Shillong 793022, India
| | - K Vasanth
- Division of Molecular Biology, Interdisciplinary Institute of Indian System of Medicine (IIISM), SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu, Chennai, TN, India
| | - S Sunitha
- Department of Chemistry, Sathyabama Institute of Science and Technology, Chennai - 600119, India
| | - Richard Hoogenboom
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281 S4, Ghent, Belgium
| | - Samarendra Maji
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu, Chennai, TN, India.
| | - Mani Ganeshpandian
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu, Chennai, TN, India.
| |
Collapse
|
37
|
Studer V, Anghel N, Desiatkina O, Felder T, Boubaker G, Amdouni Y, Ramseier J, Hungerbühler M, Kempf C, Heverhagen JT, Hemphill A, Ruprecht N, Furrer J, Păunescu E. Conjugates Containing Two and Three Trithiolato-Bridged Dinuclear Ruthenium(II)-Arene Units as In Vitro Antiparasitic and Anticancer Agents. Pharmaceuticals (Basel) 2020; 13:E471. [PMID: 33339451 PMCID: PMC7767221 DOI: 10.3390/ph13120471] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022] Open
Abstract
The synthesis, characterization, and in vitro antiparasitic and anticancer activity evaluation of new conjugates containing two and three dinuclear trithiolato-bridged ruthenium(II)-arene units are presented. Antiparasitic activity was evaluated using transgenic Toxoplasmagondii tachyzoites constitutively expressing β-galactosidase grown in human foreskin fibroblasts (HFF). The compounds inhibited T.gondii proliferation with IC50 values ranging from 90 to 539 nM, and seven derivatives displayed IC50 values lower than the reference compound pyrimethamine, which is currently used for treatment of toxoplasmosis. Overall, compound flexibility and size impacted on the anti-Toxoplasma activity. The anticancer activity of 14 compounds was assessed against cancer cell lines A2780, A2780cisR (human ovarian cisplatin sensitive and resistant), A24, (D-)A24cisPt8.0 (human lung adenocarcinoma cells wild type and cisPt resistant subline). The compounds displayed IC50 values ranging from 23 to 650 nM. In A2780cisR, A24 and (D-)A24cisPt8.0 cells, all compounds were considerably more cytotoxic than cisplatin, with IC50 values lower by two orders of magnitude. Irrespective of the nature of the connectors (alkyl/aryl) or the numbers of the di-ruthenium units (two/three), ester conjugates 6-10 and 20 exhibited similar antiproliferative profiles, and were more cytotoxic than amide analogues 11-14, 23, and 24. Polynuclear conjugates with multiple trithiolato-bridged di-ruthenium(II)-arene moieties deserve further investigation.
Collapse
Affiliation(s)
- Valentin Studer
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland; (V.S.); (O.D.); (T.F.)
| | - Nicoleta Anghel
- Vetsuisse Faculty, Institute of Parasitology, University of Bern, Länggassstrasse 122, CH-3012 Bern, Switzerland; (N.A.); (G.B.); (Y.A.); (J.R.)
| | - Oksana Desiatkina
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland; (V.S.); (O.D.); (T.F.)
| | - Timo Felder
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland; (V.S.); (O.D.); (T.F.)
| | - Ghalia Boubaker
- Vetsuisse Faculty, Institute of Parasitology, University of Bern, Länggassstrasse 122, CH-3012 Bern, Switzerland; (N.A.); (G.B.); (Y.A.); (J.R.)
| | - Yosra Amdouni
- Vetsuisse Faculty, Institute of Parasitology, University of Bern, Länggassstrasse 122, CH-3012 Bern, Switzerland; (N.A.); (G.B.); (Y.A.); (J.R.)
- Laboratoire de Parasitologie, Institution de la Recherche et de l’Enseignement Supérieur Agricoles, École Nationale de Médecine Vétérinaire de Sidi Thabet, University of Manouba, Sidi Thabet 2020, Tunisia
| | - Jessica Ramseier
- Vetsuisse Faculty, Institute of Parasitology, University of Bern, Länggassstrasse 122, CH-3012 Bern, Switzerland; (N.A.); (G.B.); (Y.A.); (J.R.)
| | - Martin Hungerbühler
- Department of BioMedical Research, Experimental Radiology, University of Bern, CH-3008 Bern, Switzerland; (M.H.); (C.K.); (J.T.H.)
- Department of Diagnostic, Interventional and Pediatric Radiology, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland
| | - Christoph Kempf
- Department of BioMedical Research, Experimental Radiology, University of Bern, CH-3008 Bern, Switzerland; (M.H.); (C.K.); (J.T.H.)
- Department of Diagnostic, Interventional and Pediatric Radiology, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland
| | - Johannes Thomas Heverhagen
- Department of BioMedical Research, Experimental Radiology, University of Bern, CH-3008 Bern, Switzerland; (M.H.); (C.K.); (J.T.H.)
- Department of Diagnostic, Interventional and Pediatric Radiology, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland
| | - Andrew Hemphill
- Vetsuisse Faculty, Institute of Parasitology, University of Bern, Länggassstrasse 122, CH-3012 Bern, Switzerland; (N.A.); (G.B.); (Y.A.); (J.R.)
| | - Nico Ruprecht
- Department of BioMedical Research, Experimental Radiology, University of Bern, CH-3008 Bern, Switzerland; (M.H.); (C.K.); (J.T.H.)
- Department of Diagnostic, Interventional and Pediatric Radiology, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland
| | - Julien Furrer
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland; (V.S.); (O.D.); (T.F.)
| | - Emilia Păunescu
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland; (V.S.); (O.D.); (T.F.)
| |
Collapse
|
38
|
Lee SY, Kim CY, Nam TG. Ruthenium Complexes as Anticancer Agents: A Brief History and Perspectives. Drug Des Devel Ther 2020; 14:5375-5392. [PMID: 33299303 PMCID: PMC7721113 DOI: 10.2147/dddt.s275007] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/03/2020] [Indexed: 12/28/2022] Open
Abstract
Platinum (Pt)-based anticancer drugs such as cisplatin have been used to treat various cancers. However, they have some limitations including poor selectivity and toxicity towards normal cells and increasing chemoresistance. Therefore, there is a need for novel metallo-anticancers, which has not been met for decades. Since the initial introduction of ruthenium (Ru) polypyridyl complex, a number of attempts at structural evolution have been conducted to improve efficacy. Among them, half-sandwich Ru-arene complexes have been the most prominent as an anticancer platform. Such complexes have clearly shown superior anticancer profiles such as increased selectivity toward cancer cells and ameliorating toxicity against normal cells compared to existing Pt-based anticancers. Currently, several Ru complexes are under human clinical trials. For improvement in selectivity and toxicity associated with chemotherapy, Ru complexes as photodynamic therapy (PDT), and photoactivated chemotherapy (PACT), which can selectively activate prodrug moieties in a specific region, have also been investigated. With all these studies on these interesting entities, new metallo-anticancer drugs to at least partially replace existing Pt-based anticancers are anticipated. This review covers a brief description of Ru-based anticancer complexes and perspectives.
Collapse
Affiliation(s)
- Sang Yeul Lee
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do15588, Republic of Korea
| | - Chul Young Kim
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do15588, Republic of Korea
| | - Tae-Gyu Nam
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do15588, Republic of Korea
| |
Collapse
|
39
|
Sudhindra P, Ajay Sharma S, Roy N, Moharana P, Paira P. Recent advances in cytotoxicity, cellular uptake and mechanism of action of ruthenium metallodrugs: A review. Polyhedron 2020. [DOI: 10.1016/j.poly.2020.114827] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
40
|
Roy N, Sen U, Madaan Y, Muthukumar V, Varddhan S, Sahoo SK, Panda D, Bose B, Paira P. Mitochondria-Targeting Click-Derived Pyridinyltriazolylmethylquinoxaline-Based Y-Shaped Binuclear Luminescent Ruthenium(II) and Iridium(III) Complexes as Cancer Theranostic Agents. Inorg Chem 2020; 59:17689-17711. [PMID: 33210921 DOI: 10.1021/acs.inorgchem.0c02928] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Due to several negative issues, market available drugs have been gradually losing their importance in the treatment of cancer. With a view to discover suitable drugs capable of diagnosing as well as inhibiting the growth of cancer cells, we have aspired to develop a group of theranostic metal complexes which will be (i) target specific, (ii) cytoselective, thus rendering the normal cell unaffected, (iii) water-soluble, (iv) cancer cell permeable, and (v) luminescent, being beneficial for healing the cancer eternally. Therefore, to reach our goal, we have prepared novel Ru(II)- and Ir(III)-based bimetallic and hetero bimetallic scaffolds using click-derived pyridinyltriazolylmethylquinoxaline ligands followed by metal coordination. Most of the compounds have displayed significant cytoselectivity against colorectal adenocarcinoma (Caco-2) and epithiloid cervical carcinoma (HeLa) cells with respect to normal human embryonic kidney cells (HEK-293) compared to cisplatin [cis-diamminedichloroplatinum(II)] along with excellent binding efficacy with DNA as well as serum albumin. Complex [(η6-p-cymene)(η5-Cp*)RuIIIrIIICl2(K2-N,N-L)](PF6)2 [RuIrL] exhibited the best cytoselectivity against all the human cancer cells and was identified as the most significant cancer theranostic agent in terms of potency, selectivity, and fluorescence quantum yield. Investigation of the localization of complex [Ir2L] and [RuIrL] in the more aggressive colorectal adenocarcinoma cell HT-29 indicates that mitochondria are the key cellular target for destroying cancer cells. Mitochondrial dysfunction and G2/M phase cell cycle arrest in HT-29 cell were found to be involved in the apoptotic cell death pathway induced by the test complexes [Ir2L] and [RuIrL]. These results validate the concept that these types of complexes will be reasonably able to exert great potential for tumor diagnosis as well as therapy in the near future.
Collapse
Affiliation(s)
- Nilmadhab Roy
- Department of Chemistry, School of advanced sciences, Vellore Institute of Technology Vellore-632014, Tamilnadu, India
| | - Utsav Sen
- Department Stem Cells and Regenerative Medicine Centre, Institution Yenepoya Research Centre, Yenepoya University, University Road, Derlakatte, Mangalore 575018, Karnataka, India
| | - Yukti Madaan
- Department of Chemistry, School of advanced sciences, Vellore Institute of Technology Vellore-632014, Tamilnadu, India
| | - Venkatesan Muthukumar
- Department of Chemistry, School of advanced sciences, Vellore Institute of Technology Vellore-632014, Tamilnadu, India
| | - Seshu Varddhan
- Department of Applied Chemistry, S. V. National Institute of Technology (SVNIT) Ichchanath, Surat, Gujrat-395007, India
| | - Suban K Sahoo
- Department of Applied Chemistry, S. V. National Institute of Technology (SVNIT) Ichchanath, Surat, Gujrat-395007, India
| | - Debashis Panda
- Department of Basic Sciences and Humanities, Rajiv Gandhi Institute of Petroleum Technology, An Institution of National Importance, Jais, Amethi-229304, Uttar Pradesh, India
| | - Bipasha Bose
- Department Stem Cells and Regenerative Medicine Centre, Institution Yenepoya Research Centre, Yenepoya University, University Road, Derlakatte, Mangalore 575018, Karnataka, India
| | - Priyankar Paira
- Department of Chemistry, School of advanced sciences, Vellore Institute of Technology Vellore-632014, Tamilnadu, India
| |
Collapse
|
41
|
Yusoh NA, Ahmad H, Gill MR. Combining PARP Inhibition with Platinum, Ruthenium or Gold Complexes for Cancer Therapy. ChemMedChem 2020; 15:2121-2135. [PMID: 32812709 PMCID: PMC7754470 DOI: 10.1002/cmdc.202000391] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Indexed: 12/24/2022]
Abstract
Platinum drugs are heavily used first-line chemotherapeutic agents for many solid tumours and have stimulated substantial interest in the biological activity of DNA-binding metal complexes. These complexes generate DNA lesions which trigger the activation of DNA damage response (DDR) pathways that are essential to maintain genomic integrity. Cancer cells exploit this intrinsic DNA repair network to counteract many types of chemotherapies. Now, advances in the molecular biology of cancer has paved the way for the combination of DDR inhibitors such as poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) and agents that induce high levels of DNA replication stress or single-strand break damage for synergistic cancer cell killing. In this review, we summarise early-stage, preclinical and clinical findings exploring platinum and emerging ruthenium anti-cancer complexes alongside PARPi in combination therapy for cancer and also describe emerging work on the ability of ruthenium and gold complexes to directly inhibit PARP activity.
Collapse
Affiliation(s)
- Nur Aininie Yusoh
- Department of ChemistryFaculty of ScienceUniversiti Putra Malaysia43400 UPMSerdang, SelangorMalaysia
| | - Haslina Ahmad
- Department of ChemistryFaculty of ScienceUniversiti Putra Malaysia43400 UPMSerdang, SelangorMalaysia
- Integrated Chemical BiophysicsFaculty of ScienceUniversiti Putra Malaysia43400 UPMSerdang, SelangorMalaysia
| | - Martin R. Gill
- Department of ChemistrySwansea UniversitySwanseaWales (UK
| |
Collapse
|
42
|
Kar B, Roy N, Pete S, Moharana P, Paira P. Ruthenium and iridium based mononuclear and multinuclear complexes: A Breakthrough of Next-Generation anticancer metallopharmaceuticals. Inorganica Chim Acta 2020. [DOI: 10.1016/j.ica.2020.119858] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
43
|
Oncometabolites lactate and succinate drive pro-angiogenic macrophage response in tumors. Biochim Biophys Acta Rev Cancer 2020; 1874:188427. [PMID: 32961257 DOI: 10.1016/j.bbcan.2020.188427] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/21/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023]
Abstract
Macrophages are innate phagocytic leukocytes that are highly present in solid tumors, where they are referred to as tumor-associated macrophages (TAMs). In solid tumors, the microenvironment is often immunosuppressive and hypoxic regions are prevalent. These hypoxic conditions impose tumor cells to reprogram their metabolism, shifting from oxidative phosphorylation to anaerobic glycolysis. This so-called glycolytic switch enables hypoxic tumor cells to survive, proliferate, and eventually to outcompete untransformed cells. The hypoxia-induced change in tumor cell metabolism leads to the production of oncometabolites, among which are the glycolytic end-metabolite lactate and the tricarboxylic acid cycle intermediate succinate. TAMs can react to these oncometabolites, resulting in an altered maturation and the adoption of pro-angiogenic features. These angiogenesis-promoting TAMs have been reported to cooperate with tumor cells in the formation of new vessels, and even have been considered an important cause of resistance against anti-angiogenic therapies. For a long time, the mechanisms by which lactate and succinate activated pro-angiogenic TAMs were not understood. Researchers now start to unravel and understand some of the underlying mechanisms. Here, the importance of microenvironmental cues in inducing different macrophage activation states is discussed, as well as the role of hypoxia in the recruitment and activation of pro-angiogenic macrophages. In addition, the latest findings on the oncometabolites lactate and succinate in the activation of angiogenesis supporting macrophages are reviewed. Finally, various oncometabolite-targeting therapeutic strategies are proposed that could improve the response to anti-angiogenic therapies. SIGNIFICANCE STATEMENT: Tumor-associated macrophages (TAMs) are known promotors of tumor neovascularization, and significantly contribute to the emergence of resistance to anti-angiogenic therapies. Recent evidence suggests that the angiogenesis promoting phenotype of TAMs can be activated by hypoxic tumor cell-derived oncometabolites, including lactate and succinate. Here, the latest findings into the lactate- and succinate-mediated mechanistic activation of pro-angiogenic TAMs are reviewed, and therapeutic strategies that interfere with this mechanism and may delay or even prevent acquired resistance to anti-angiogenic agents are discussed.
Collapse
|
44
|
A System Biology-Based Approach for Designing Combination Therapy in Cancer Precision Medicine. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5072697. [PMID: 32908895 PMCID: PMC7471815 DOI: 10.1155/2020/5072697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/07/2020] [Accepted: 07/22/2020] [Indexed: 12/02/2022]
Abstract
In this paper, we have used an agent-based stochastic tumor growth model and presented a mathematical and theoretical perspective to cancer therapy. This perspective can be used to theoretical study of precision medicine and combination therapy in individuals. We have conducted a series of in silico combination therapy experiments. Based on cancer drugs and new findings of cancer biology, we hypothesize relationships between model parameters which in some cases represent individual genome characteristics and cancer drugs, i.e., in our approach, therapy players are delegated by biologically reasonable parameters. In silico experiments showed that combined therapies are more effective when players affect tumor via different mechanisms and have different physical dimensions. This research presents for the first time an algorithm as a theoretical viewpoint for the prediction of effectiveness and classification of therapy sets.
Collapse
|
45
|
Karges J, Chao H, Gasser G. Synthesis, Characterization, and Biological Evaluation of the Polymeric Encapsulation of a Ruthenium(II) Polypyridine Complex with Pluronic F‐127/Poloxamer‐407 for Photodynamic Therapy Applications. Eur J Inorg Chem 2020. [DOI: 10.1002/ejic.202000545] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Johannes Karges
- Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology Chimie ParisTech, PSL University, CNRS 75005 Paris France
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry Sun Yat‐Sen University 510275 Guangzhou People's Republic of China
| | - Gilles Gasser
- Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology Chimie ParisTech, PSL University, CNRS 75005 Paris France
| |
Collapse
|
46
|
Gonchar MR, Matnurov EM, Burdina TA, Zava O, Ridel T, Milaeva ER, Dyson PJ, Nazarov AA. Ruthenium(II)–arene and triruthenium-carbonyl cluster complexes with new water-soluble phopsphites based on glucose: Synthesis, characterization and antiproliferative activity. J Organomet Chem 2020. [DOI: 10.1016/j.jorganchem.2020.121312] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
47
|
Identification of low-dose multidrug combinations for sunitinib-naive and pre-treated renal cell carcinoma. Br J Cancer 2020; 123:556-567. [PMID: 32439932 PMCID: PMC7435198 DOI: 10.1038/s41416-020-0890-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 04/04/2020] [Accepted: 04/23/2020] [Indexed: 12/11/2022] Open
Abstract
Background Combinations of drugs can improve the efficacy of cancer treatment, enable the reduction of side effects and the occurrence of acquired drug resistance. Methods We approached this challenge mathematically by using the validated technology called the Therapeutically Guided Multidrug Optimization (TGMO) method. In a set of genetically distinct human renal cell carcinoma (RCC) cell lines, either treated chronically with sunitinib (−ST) or sunitinib-naive, we identified cell line-specific low-dose-optimised drug combinations (ODC). Results Six cell-type-specific low-dose drug combinations for three sunitinib-naive as well as three sunitinib pre-treated cells were established. These ODCs effectively inhibited the RCC cell metabolic activity while being ineffective in non-cancerous cells. Based on a single screening test and three searches, starting with ten drugs, we identified highly efficacious drug mixtures containing four drugs. All ODCs contained AZD4547 (FGFR signalling pathway inhibitor) and pictilisib (pan-phosphatidylinositol 3-kinase inhibitor), but varied in the third and fourth drug. ODC treatment significantly decreased cell metabolic activity (up to 70%) and induced apoptosis, independent of the pretreatment with sunitinib. The ODCs outperformed sunitinib, the standard care for RCC. Moreover, short-term starvation potentiated the ODC activity. The translation of the 2D-based results to 3D heterotypic co-culture models revealed significant inhibition of the spheroid growth (up to 95%). Conclusion We demonstrate a promising low-dose drug combination development to obtain drug combinations effective in naive as well as resistant tumours. Nevertheless, we emphasise the need for further mechanistic investigation and preclinical development.
Collapse
|
48
|
Balaji S, Mohamed Subarkhan MK, Ramesh R, Wang H, Semeril D. Synthesis and Structure of Arene Ru(II) N∧O-Chelating Complexes: In Vitro Cytotoxicity and Cancer Cell Death Mechanism. Organometallics 2020. [DOI: 10.1021/acs.organomet.0c00092] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Sundarraman Balaji
- Centre for Organometallic Chemistry, School of Chemistry, Bharathidasan University, Tiruchirappalli 620 024, India
| | - Mohamed Kasim Mohamed Subarkhan
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Rengan Ramesh
- Centre for Organometallic Chemistry, School of Chemistry, Bharathidasan University, Tiruchirappalli 620 024, India
| | - Hangxiang Wang
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - David Semeril
- Laboratoire de Chimie Inorganique et Catalyse, Institut de Chimie, UMR 7177, CNRS, Universite de Strasbourg, Strasbourg 67008, France
| |
Collapse
|
49
|
Shu L, Ren L, Wang Y, Fang T, Ye Z, Han W, Chen C, Wang H. Niacin-ligated platinum(iv)-ruthenium(ii) chimeric complexes synergistically suppress tumor metastasis and growth with potentially reduced toxicity in vivo. Chem Commun (Camb) 2020; 56:3069-3072. [PMID: 32049075 DOI: 10.1039/c9cc09016a] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Niacin-ligated platinum(iv)-ruthenium(ii) chimeric complexes (PtRu 1-4) have been synthesized and evaluated for their antitumor performance. Using the optimal complex, PtRu-1, we show that this water-soluble chimeric prodrug not only potently inhibits the metastasis and proliferation of tumor cells but also has an unexpectedly higher safety margin in animals compared with the traditionally-used, clinically approved drug cisplatin.
Collapse
Affiliation(s)
- Liwei Shu
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China. and Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, P. R. China
| | - Lulu Ren
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, P. R. China
| | - Yuchen Wang
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China. and Department of Chemical Engineering, Zhejiang, University, Hangzhou, P. R. China
| | - Tao Fang
- Jinhua People's Hospital, Jinhua, Zhejiang Province 321000, P. R. China
| | - Zhijian Ye
- Jinhua People's Hospital, Jinhua, Zhejiang Province 321000, P. R. China
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, P. R. China
| | - Chao Chen
- College of Life Sciences, Huzhou University, Huzhou, 313000, P. R. China.
| | - Hangxiang Wang
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, P. R. China.
| |
Collapse
|
50
|
Seiler KM, Bajinting A, Alvarado DM, Traore MA, Binkley MM, Goo WH, Lanik WE, Ou J, Ismail U, Iticovici M, King CR, VanDussen KL, Swietlicki EA, Gazit V, Guo J, Luke CJ, Stappenbeck T, Ciorba MA, George SC, Meacham JM, Rubin DC, Good M, Warner BW. Patient-derived small intestinal myofibroblasts direct perfused, physiologically responsive capillary development in a microfluidic Gut-on-a-Chip Model. Sci Rep 2020; 10:3842. [PMID: 32123209 PMCID: PMC7051952 DOI: 10.1038/s41598-020-60672-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 02/13/2020] [Indexed: 02/07/2023] Open
Abstract
The development and physiologic role of small intestine (SI) vasculature is poorly studied. This is partly due to a lack of targetable, organ-specific markers for in vivo studies of two critical tissue components: endothelium and stroma. This challenge is exacerbated by limitations of traditional cell culture techniques, which fail to recapitulate mechanobiologic stimuli known to affect vessel development. Here, we construct and characterize a 3D in vitro microfluidic model that supports the growth of patient-derived intestinal subepithelial myofibroblasts (ISEMFs) and endothelial cells (ECs) into perfused capillary networks. We report how ISEMF and EC-derived vasculature responds to physiologic parameters such as oxygen tension, cell density, growth factors, and pharmacotherapy with an antineoplastic agent (Erlotinib). Finally, we demonstrate effects of ISEMF and EC co-culture on patient-derived human intestinal epithelial cells (HIECs), and incorporate perfused vasculature into a gut-on-a-chip (GOC) model that includes HIECs. Overall, we demonstrate that ISEMFs possess angiogenic properties as evidenced by their ability to reliably, reproducibly, and quantifiably facilitate development of perfused vasculature in a microfluidic system. We furthermore demonstrate the feasibility of including perfused vasculature, including ISEMFs, as critical components of a novel, patient-derived, GOC system with translational relevance as a platform for precision and personalized medicine research.
Collapse
Grants
- R01 HD105301 NICHD NIH HHS
- R01 DK106382 NIDDK NIH HHS
- T32 DK007130 NIDDK NIH HHS
- R01 DK104698 NIDDK NIH HHS
- R01 DK114047 NIDDK NIH HHS
- R03 DK111473 NIDDK NIH HHS
- R01 DK109384 NIDDK NIH HHS
- R01 DK118568 NIDDK NIH HHS
- R01 DK112378 NIDDK NIH HHS
- K08 DK101608 NIDDK NIH HHS
- P30 DK052574 NIDDK NIH HHS
- T32 HD043010 NICHD NIH HHS
- K01 DK109081 NIDDK NIH HHS
- Association for Academic Surgery Foundation (AASF)
- Children’s Discovery Institute of Washington University in St. Louis and St. Louis Children’s Hospital MI-F-2017-629; National Institutes of Health 4T32HD043010-14
- National Institutes of Health 3T32DK007130-45S1
- Givin’ it all for Guts Foundation (https://givinitallforguts.org/), Lawrence C. Pakula MD IBD Research, Innovation, and Education Fund, National Institutes of Health R01DK109384
- National Institutes of Health R03DK111473, R01DK118568, and K08DK101608, Children’s Discovery Institute of Washington University in St. Louis and St. Louis Children’s Hospital MI-FR-2017-596, March of Dimes Foundation Grant No. 5-FY17-79, Department of Pediatrics at Washington University School of Medicine, St. Louis
Collapse
Affiliation(s)
- Kristen M Seiler
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Adam Bajinting
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Saint Louis University School of Medicine, St. Louis, Missouri, United States
| | - David M Alvarado
- Division of Gastroenterology and the Inflammatory Bowel Diseases Center, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Mahama A Traore
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| | - Michael M Binkley
- Department of Mechanical Engineering & Materials Science, Washington University McKelvey School of Engineering, St. Louis, MO, United States
| | - William H Goo
- Washington University, St. Louis, Missouri, United States
| | - Wyatt E Lanik
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Jocelyn Ou
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Usama Ismail
- Department of Mechanical Engineering & Materials Science, Washington University McKelvey School of Engineering, St. Louis, MO, United States
| | - Micah Iticovici
- Division of Gastroenterology and the Inflammatory Bowel Diseases Center, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Cristi R King
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Kelli L VanDussen
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Elzbieta A Swietlicki
- Division of Gastroenterology and the Inflammatory Bowel Diseases Center, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Vered Gazit
- Division of Gastroenterology and the Inflammatory Bowel Diseases Center, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Jun Guo
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Cliff J Luke
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Thaddeus Stappenbeck
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Matthew A Ciorba
- Division of Gastroenterology and the Inflammatory Bowel Diseases Center, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, California, United States
| | - J Mark Meacham
- Department of Mechanical Engineering & Materials Science, Washington University McKelvey School of Engineering, St. Louis, MO, United States
| | - Deborah C Rubin
- Division of Gastroenterology and the Inflammatory Bowel Diseases Center, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Misty Good
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Brad W Warner
- Division of Pediatric Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, United States.
| |
Collapse
|