1
|
Mohanty M, Das S, Pattanayak PD, Lima S, Kaminsky W, Dinda R. Ru III-Morpholine-Derived Thiosemicarbazone-Based Metallodrugs: Lysosome-Targeted Anticancer Agents. ACS APPLIED BIO MATERIALS 2025; 8:1210-1226. [PMID: 39806879 DOI: 10.1021/acsabm.4c01536] [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] [Indexed: 01/16/2025]
Abstract
The idea of coordinating biologically active ligand systems to metal centers to exploit their synergistic effects has gained momentum. Therefore, in this report, three RuIII complexes 1-3 of morpholine-derived thiosemicarbazone ligands have been prepared and characterized by spectroscopy and HRMS along with the structure of 2 through a single-crystal X-ray diffraction study. The solution stability of 1-3 was tested using conventional techniques such as UV-vis and HRMS. Further, the anticancer activity of 1-3 was tested in HT-29 and HeLa cancer cell lines. To gain insight into their mechanism of action, the cytotoxicity, hydrophobicity, and the interaction of 1-3 with DNA and HSA were evaluated by different conventional methods such as absorption, fluorescence, and circular dichroism studies. Along with favorable biomolecule interaction, 1-3 revealed potent selectivity toward cancer cells, which is a prerequisite for the generation of an anticancer drug. According to cell viability results, 1 has the highest cytotoxicity among all in the group, against both cells, respectively. Additionally, the fluorescence-active ruthenium complexes selectively target lysosomes, which is evaluated by live-cell imaging. 1-3 disrupt the lysosome membrane potential by generating an excessive amount of reactive oxygen species, which results in an apoptotic mode of cell death.
Collapse
Affiliation(s)
- Monalisa Mohanty
- Department of Chemistry, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Sanchita Das
- Department of Chemistry, National Institute of Technology, Rourkela, Odisha 769008, India
| | | | - Sudhir Lima
- Department of Chemistry, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Werner Kaminsky
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Rupam Dinda
- Department of Chemistry, National Institute of Technology, Rourkela, Odisha 769008, India
| |
Collapse
|
2
|
Teixeira R, Stefanelli A, Pilon A, Warmers R, Fontrodona X, Romero I, Costa PJ, Villa de Brito MJ, Hudec X, Pirker C, Türck S, Antunes AMM, Kowol CR, Ott I, Brozovic A, Sombke A, Eckhard M, Tomaz AI, Heffeter P, Valente A. Paraptotic Cell Death as an Unprecedented Mode of Action Observed for New Bipyridine-Silver(I) Compounds Bearing Phosphane Coligands. J Med Chem 2024; 67:6081-6098. [PMID: 38401050 PMCID: PMC11056982 DOI: 10.1021/acs.jmedchem.3c01036] [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: 06/26/2023] [Revised: 01/20/2024] [Accepted: 02/12/2024] [Indexed: 02/26/2024]
Abstract
In this work, we investigated the anticancer activity of several novel silver(I) 2,2'-bipyridine complexes containing either triphenylphosphane (PPh3) or 1,2-bis(diphenylphosphino)ethane (dppe) ligands. All compounds were characterized by diverse analytical methods including ESI-MS spectrometry; NMR, UV-vis, and FTIR spectroscopies; and elemental analysis. Moreover, several compounds were also studied by X-ray single-crystal diffraction. Subsequently, the compounds were investigated for their anticancer activity against drug-resistant and -sensitive cancer cells. Noteworthily, neither carboplatin and oxaliplatin resistance nor p53 deletion impacted on their anticancer efficacy. MES-OV cells displayed exceptional hypersensitivity to the dppe-containing drugs. This effect was not based on thioredoxin reductase inhibition, enhanced drug uptake, or apoptosis induction. In contrast, dppe silver drugs induced paraptosis, a novel recently described form of programmed cell death. Together with the good tumor specificity of this compound's class, this work suggests that dppe-containing silver complexes could be interesting drug candidates for the treatment of resistant ovarian cancer.
Collapse
Affiliation(s)
- Ricardo
G. Teixeira
- Centro
de Química Estrutural, Institute of Molecular Sciences, Departamento
de Química e Bioquímica, Faculdade
de Ciências, Universidade de Lisboa, Campo Grande, Lisboa 1749-016, Portugal
| | - Alessia Stefanelli
- Center
for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna 1090, Austria
| | - Adhan Pilon
- Centro
de Química Estrutural, Institute of Molecular Sciences, Departamento
de Química e Bioquímica, Faculdade
de Ciências, Universidade de Lisboa, Campo Grande, Lisboa 1749-016, Portugal
| | - Rebecca Warmers
- Center
for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna 1090, Austria
| | - Xavier Fontrodona
- Departament
de Química and Serveis Tècnics de Recerca, Universitat de Girona, Campus de Montilivi, Girona 17071, Spain
| | - Isabel Romero
- Departament
de Química and Serveis Tècnics de Recerca, Universitat de Girona, Campus de Montilivi, Girona 17071, Spain
| | - Paulo J. Costa
- BioISI
- Instituto de Biosistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, Lisboa 1749-016, Portugal
| | - Maria J. Villa de Brito
- Centro
de Química Estrutural, Institute of Molecular Sciences, Departamento
de Química e Bioquímica, Faculdade
de Ciências, Universidade de Lisboa, Campo Grande, Lisboa 1749-016, Portugal
| | - Xenia Hudec
- Center
for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna 1090, Austria
| | - Christine Pirker
- Center
for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna 1090, Austria
| | - Sebastian Türck
- Institute
of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstr. 55, Braunschweig 38106, Germany
| | - Alexandra M. M. Antunes
- Centro de
Química Estrutural (CQE), Institute of Molecular Sciences,
Departamento de Engenharia Química, Instituto Superior Técnico
(IST), Universidade de Lisboa, Av Rovisco Pais 1, Lisboa 1049-001, Portugal
| | - Christian R. Kowol
- Institute
of Inorganic Chemistry, Faculty of Chemistry,
University of Vienna, Waehringerstrasse 42, Vienna 1090, Austria
| | - Ingo Ott
- Institute
of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstr. 55, Braunschweig 38106, Germany
| | - Anamaria Brozovic
- Division
of Molecular Biology, Ruđer Bošković
Institute, Bijenička
cesta 54,Zagreb 10000, Croatia
| | - Andy Sombke
- Center
for Anatomy and Cell Biology, Cell and Developmental Biology, Medical University of Vienna, Schwarzspanierstraße 17, Vienna 1090, Austria
| | - Margret Eckhard
- Center
for Anatomy and Cell Biology, Cell and Developmental Biology, Medical University of Vienna, Schwarzspanierstraße 17, Vienna 1090, Austria
| | - Ana Isabel Tomaz
- Centro
de Química Estrutural, Institute of Molecular Sciences, Departamento
de Química e Bioquímica, Faculdade
de Ciências, Universidade de Lisboa, Campo Grande, Lisboa 1749-016, Portugal
| | - Petra Heffeter
- Center
for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna 1090, Austria
| | - Andreia Valente
- Centro
de Química Estrutural, Institute of Molecular Sciences, Departamento
de Química e Bioquímica, Faculdade
de Ciências, Universidade de Lisboa, Campo Grande, Lisboa 1749-016, Portugal
| |
Collapse
|
3
|
Sumithaa C, Gajda-Morszewski P, Ishaniya W, Khamrang T, Velusamy M, Bhuvanesh N, Brindell M, Mazuryk O, Ganeshpandian M. Design of an anticancer organoruthenium complex as the guest and polydiacetylene-coated fluorogenic nanocarrier as the host: engineering nanocarrier using ene-yne conjugation for sustained guest release, enhanced anticancer activity and reduced in vivo toxicity. Dalton Trans 2024; 53:966-985. [PMID: 38054338 DOI: 10.1039/d3dt03358a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Despite the enormous efforts made over the past two decades to develop metallodrugs and nanocarriers for metallodrug delivery, there are still few precise strategies that aim to optimize the design of both metallodrugs and metallodrug carriers jointly in a concerted effort. In this work, three half-sandwich ruthenium(II) complexes with pyridylimidazo[1,5-a]pyridine ligand functionalized with polycyclic aromatic moiety (Ru(nap), Ru(ant), Ru(pyr)) are evaluated as possible anticancer candidates and polydiacetylene (PDA)-coated amino-functionalized mesoporous silica nanoparticles (AMSNs) are designed as a functional nanocarrier for drug delivery. Ru(pyr) exhibits higher cytotoxicity in HT-29 colorectal cancer cells compared to other complexes and cis-platin, but it does not exhibit better cellular uptake. Ru(pyr) is found to be preferentially accumulated in plasma, mitochondria, and ER-Golgi membrane. The complex induces cell cycle arrest in the G0/G1 phase, while higher concentrations cause programmed cell death via apoptosis. Ru(pyr) influences cancer cell adhesion property and acts as an antioxidant in HT-29 cells. In order to modulate the anticancer potency of Ru(pyr), AMSNs are used to encapsulate the complex, and then diacetylene self-assembly is allowed to deposit on the surface of the nanoparticles. Subsequently, the nanoparticles undergo topopolymerization, which results in π-conjugated PDA-Ru(pyr)@AMSNs. Owing to the ene-yne polymeric skeleton in the backbone, the non-fluorescent AMSNs turn into red-emissive particles, which are exploited for cell imaging applications. The release profile analysis reveals that such a π-conjugated polymer prevents the premature release of the complex from porous silica nanoparticles with the accelerated release of the complex in an acidic medium compared to physiological conditions. The PDA gatekeepers have also been proven to enhance the cellular internalization of Ru(pyr) with slow continuous release from the nanoformulation. Zebrafish embryo toxicity analysis suggests that the PDA-coated nanocarriers could be suitable candidates for in vivo investigations.
Collapse
Affiliation(s)
- Chezhiyan Sumithaa
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| | - Przemyslaw Gajda-Morszewski
- Faculty of Chemistry, Department of Inorganic Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Cracow, Poland.
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Łojasiewicza 11, 30-348 Cracow, Poland
| | - Wickneswaran Ishaniya
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| | - Themmila Khamrang
- Department of Chemistry, Dhanamanjuri University, Manipur 795001, India
| | - Marappan Velusamy
- Department of Chemistry, North Eastern Hill University, Shillong 793022, India
| | - Nattamai Bhuvanesh
- X-ray Diffraction Lab, Department of Chemistry, Texas A&M University, College Station, TX 77842, USA
| | - Malgorzata Brindell
- Faculty of Chemistry, Department of Inorganic Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Cracow, Poland.
| | - Olga Mazuryk
- Faculty of Chemistry, Department of Inorganic Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Cracow, Poland.
| | - Mani Ganeshpandian
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
4
|
The Anticancer Ruthenium Compound BOLD-100 Targets Glycolysis and Generates a Metabolic Vulnerability towards Glucose Deprivation. Pharmaceutics 2022; 14:pharmaceutics14020238. [PMID: 35213972 PMCID: PMC8875291 DOI: 10.3390/pharmaceutics14020238] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 12/13/2022] Open
Abstract
Cellular energy metabolism is reprogrammed in cancer to fuel proliferation. In oncological therapy, treatment resistance remains an obstacle and is frequently linked to metabolic perturbations. Identifying metabolic changes as vulnerabilities opens up novel approaches for the prevention or targeting of acquired therapy resistance. Insights into metabolic alterations underlying ruthenium-based chemotherapy resistance remain widely elusive. In this study, colon cancer HCT116 and pancreatic cancer Capan-1 cells were selected for resistance against the clinically evaluated ruthenium complex sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (BOLD-100). Gene expression profiling identified transcriptional deregulation of carbohydrate metabolism as a response to BOLD-100 and in resistance against the drug. Mechanistically, acquired BOLD-100 resistance is linked to elevated glucose uptake and an increased lysosomal compartment, based on a defect in downstream autophagy execution. Congruently, metabolomics suggested stronger glycolytic activity, in agreement with the distinct hypersensitivity of BOLD-100-resistant cells to 2-deoxy-d-glucose (2-DG). In resistant cells, 2-DG induced stronger metabolic perturbations associated with ER stress induction and cytoplasmic lysosome deregulation. The combination with 2-DG enhanced BOLD-100 activity against HCT116 and Capan-1 cells and reverted acquired BOLD-100 resistance by synergistic cell death induction and autophagy disturbance. This newly identified enhanced glycolytic activity as a metabolic vulnerability in BOLD-100 resistance suggests the targeting of glycolysis as a promising strategy to support BOLD-100 anticancer activity.
Collapse
|
5
|
Valente A, Podolski-Renić A, Poetsch I, Filipović N, López Ó, Turel I, Heffeter P. Metal- and metalloid-based compounds to target and reverse cancer multidrug resistance. Drug Resist Updat 2021; 58:100778. [PMID: 34403910 DOI: 10.1016/j.drup.2021.100778] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/18/2021] [Accepted: 08/03/2021] [Indexed: 12/19/2022]
Abstract
Drug resistance remains the major cause of cancer treatment failure especially at the late stage of the disease. However, based on their versatile chemistry, metal and metalloid compounds offer the possibility to design fine-tuned drugs to circumvent and even specifically target drug-resistant cancer cells. Based on the paramount importance of platinum drugs in the clinics, two main areas of drug resistance reversal strategies exist: overcoming resistance to platinum drugs as well as multidrug resistance based on ABC efflux pumps. The current review provides an overview of both aspects of drug design and discusses the open questions in the field. The areas of drug resistance covered in this article involve: 1) Altered expression of proteins involved in metal uptake, efflux or intracellular distribution, 2) Enhanced drug efflux via ABC transporters, 3) Altered metabolism in drug-resistant cancer cells, 4) Altered thiol or redox homeostasis, 5) Altered DNA damage recognition and enhanced DNA damage repair, 6) Impaired induction of apoptosis and 7) Altered interaction with the immune system. This review represents the first collection of metal (including platinum, ruthenium, iridium, gold, and copper) and metalloid drugs (e.g. arsenic and selenium) which demonstrated drug resistance reversal activity. A special focus is on compounds characterized by collateral sensitivity of ABC transporter-overexpressing cancer cells. Through this approach, we wish to draw the attention to open research questions in the field. Future investigations are warranted to obtain more insights into the mechanisms of action of the most potent compounds which target specific modalities of drug resistance.
Collapse
Affiliation(s)
- Andreia Valente
- Centro de Química Estrutural and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisboa, Portugal
| | - Ana Podolski-Renić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Serbia
| | - Isabella Poetsch
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Nenad Filipović
- Department of Chemistry and Biochemistry, Faculty of Agriculture, University of Belgrade, Belgrade, Serbia
| | - Óscar López
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Sevilla, Spain
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
6
|
Davis J, Cetto A, Campbell M, Scoggins S, Stultz L, Hanson P. DMSO reduces the cytotoxicity of anticancer ruthenium complex KP1019 in yeast. MICROPUBLICATION BIOLOGY 2021; 2021:10.17912/micropub.biology.000436. [PMID: 34377963 PMCID: PMC8339913 DOI: 10.17912/micropub.biology.000436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 11/25/2022]
Abstract
Low solubility in aqueous solutions is a significant limitation of the otherwise promising anticancer ruthenium complex KP1019. In laboratory studies, this challenge is often overcome by using DMSO to help drive the drug into solution. Since DMSO was previously shown to alter the bioactivity of platinum-based chemotherapeutics, here we examine DMSO's effects on KP1019. Using Saccharomyces cerevisiae as a model organism, we apply multiple measures of growth inhibition to demonstrate that DMSO reduces the drug's toxicity. This reduction in bioactivity correlates with spectrophotometric changes consistent with DMSO-dependent increases in the stability of the KP1019 pro-drug. The impact of DMSO on the biology and chemistry of KP1019 suggests this solvent should not be used in studies of this and similar anticancer ruthenium complexes.
Collapse
Affiliation(s)
- Jonathan Davis
- Department of Biology, Furman University, Greenville, SC 29613, USA
| | - Anne Cetto
- Department of Chemistry, Birmingham-Southern College, Birmingham, AL 35254, USA
| | - Mary Campbell
- Department of Chemistry, Birmingham-Southern College, Birmingham, AL 35254, USA
| | - Seth Scoggins
- Department of Chemistry, Birmingham-Southern College, Birmingham, AL 35254, USA
| | - Laura Stultz
- Department of Chemistry, Birmingham-Southern College, Birmingham, AL 35254, USA
| | - Pamela Hanson
- Department of Biology, Furman University, Greenville, SC 29613, USA,
Correspondence to: Pamela Hanson ()
| |
Collapse
|
7
|
Lazić D, Scheurer A, Ćoćić D, Milovanović J, Arsenijević A, Stojanović B, Arsenijević N, Milovanović M, Rilak Simović A. A new bis-pyrazolylpyridine ruthenium(III) complex as a potential anticancer drug: in vitro and in vivo activity in murine colon cancer. Dalton Trans 2021; 50:7686-7704. [PMID: 33982702 DOI: 10.1039/d1dt00185j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We synthesized and characterized the ruthenium(iii) pincer-type complex [RuCl3(H2Lt-Bu] (H2Lt-Bu = 2,6-bis(5-tert-butyl-1H-pyrazol-3-yl)pyridine, 1) by elemental analysis, IR and UV-Vis spectroscopy, and the mass spectrometry (MS) method ESI Q-TOF. For comparison reasons, we also studied ruthenium(iii) terpyridine complexes of the general formula [Ru(N-N-N)Cl3], where N-N-N = 4'-chloro-terpyridine (Cl-tpy; 2) or 4'-chlorophenyl-terpyridine (Cl-Ph-tpy; 3). A kinetic study of the substitution reactions of 1-3 with biomolecules showed that the rate constants depend on the properties of the spectator ligand and the nature of the entering nucleophile. The DNA/HSA binding study showed that in comparison to complex 1 (bis-pyrazolylpyridine), the other two (2 and 3) terpyridine complexes had a slightly better binding affinity to calf thymus DNA (CT DNA), while in the case of human serum albumin (HSA), complex 1 exhibited the strongest quenching ability. We demonstrated that 1 possesses significant in vitro cytotoxic activity against mouse colon carcinoma CT26 cells and in vivo antitumor activity in murine heterotopic colon carcinoma. Complex 1 induced G0/G1 cell cycle arrest and apoptotic death in CT26 cells. Additionally, 1 showed antiproliferative activity, as evaluated by the detection of the expression levels of the Ki67 protein. Furthermore, the in vivo results showed that 1 reduced primary tumour growth and the number and growth of lung and liver metastases, significantly prolonging the treated mice's survival rate. This study highlighted that 1 does not show hepato- and nephrotoxicity. Our data demonstrated the considerable antitumor activity of the ruthenium(iii) pincer complex against CT26 tumour cells and implicated further investigations of its role as a potential chemotherapeutic agent for colon carcinoma.
Collapse
Affiliation(s)
- Dejan Lazić
- Department of Surgery, Faculty of Medical Sciences, University of Kraujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Andreas Scheurer
- Inorganic Chemistry, Department of Chemistry and Pharmacy, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Dušan Ćoćić
- University of Kragujevac, Faculty of Science, Radoja Domanovića 12, P. O. Box 60, 34000 Kragujevac, Serbia
| | - Jelena Milovanović
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia. and Department of Histology, Faculty of Medical Sciences, University of Kragujevac, Serbia
| | - Aleksandar Arsenijević
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.
| | - Bojana Stojanović
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia. and Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Serbia
| | - Nebojša Arsenijević
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.
| | - Marija Milovanović
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.
| | - Ana Rilak Simović
- University of Kragujevac, Institute for Information Technologies Kragujevac, Department of Natural Sciences, Jovana Cvijića bb, 34000 Kragujevac, Serbia.
| |
Collapse
|
8
|
Shutkov IA, Antonets AA, Tyurin VY, Milaeva ER, Nazarov AA. Ruthenium(III) Complexes of NAMI-A Type with Ligands Based on Lonidamine and Bexarotene as Antiproliferative Agents. RUSS J INORG CHEM+ 2021. [DOI: 10.1134/s0036023621030177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
9
|
Hager S, Pape VFS, Pósa V, Montsch B, Uhlik L, Szakács G, Tóth S, Jabronka N, Keppler BK, Kowol CR, Enyedy ÉA, Heffeter P. High Copper Complex Stability and Slow Reduction Kinetics as Key Parameters for Improved Activity, Paraptosis Induction, and Impact on Drug-Resistant Cells of Anticancer Thiosemicarbazones. Antioxid Redox Signal 2020; 33:395-414. [PMID: 32336116 DOI: 10.1089/ars.2019.7854] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Aims: Due to their significant biological activity, thiosemicarbazones (TSCs) are promising candidates for anticancer therapy. In part, the efficacy of TSCs is linked to their ability to chelate essential metal ions such as copper and iron. Triapine, the best-studied anticancer TSC, has been tested clinically with promising results in hematological diseases. During the past few years, a novel subclass of TSCs with improved anticancer activity was found to induce paraptosis, a recently characterized form of cell death. The aim of this study was to identify structural and chemical properties associated with anticancer activity and paraptosis induction of TSCs. Results: When testing a panel of structurally related TSCs, compounds with nanomolar anticancer activity and paraptosis-inducing properties showed higher copper(II) complex solution stability and a slower reduction rate, which resulted in reduced redox activity. In contrast, TSCs with lower anticancer activity induced higher levels of superoxide that rapidly stimulated superoxide dismutase expression in treated cells, effectively protecting the cells from drug-induced redox stress. Innovation: Consequently, we hypothesize that in the case of close Triapine derivatives, intracellular reduction leads to rapid dissociation of intracellularly formed copper complexes. In contrast, TSCs characterized by highly stable, slowly reducible copper(II) complexes are able to reach new intracellular targets such as the endoplasmic reticulum-resident protein disulfide isomerase. Conclusion: The additional modes of actions observed with highly active TSC derivatives are based on intracellular formation of stable copper complexes, offering a new approach to combat (drug-resistant) cancer cells.
Collapse
Affiliation(s)
- Sonja Hager
- Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
- Research Cluster 'Translational Cancer Therapy Research,' Vienna, Austria
| | - Veronika F S Pape
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Vivien Pósa
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Szeged, Hungary
| | - Bianca Montsch
- Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
- Research Cluster 'Translational Cancer Therapy Research,' Vienna, Austria
| | - Lukas Uhlik
- Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
- Research Cluster 'Translational Cancer Therapy Research,' Vienna, Austria
| | - Gergely Szakács
- Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Szilárd Tóth
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Nikolett Jabronka
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Bernhard K Keppler
- Research Cluster 'Translational Cancer Therapy Research,' Vienna, Austria
- Faculty of Chemistry, Institute of Inorganic Chemistry, University of Vienna, Vienna, Austria
| | - Christian R Kowol
- Research Cluster 'Translational Cancer Therapy Research,' Vienna, Austria
- Faculty of Chemistry, Institute of Inorganic Chemistry, University of Vienna, Vienna, Austria
| | - Éva A Enyedy
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Szeged, Hungary
| | - Petra Heffeter
- Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
- Research Cluster 'Translational Cancer Therapy Research,' Vienna, Austria
| |
Collapse
|
10
|
Marker SC, King AP, Swanda RV, Vaughn B, Boros E, Qian SB, Wilson JJ. Exploring Ovarian Cancer Cell Resistance to Rhenium Anticancer Complexes. Angew Chem Int Ed Engl 2020; 59:13391-13400. [PMID: 32396709 PMCID: PMC7482417 DOI: 10.1002/anie.202004883] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/11/2020] [Indexed: 11/09/2022]
Abstract
Rhenium tricarbonyl complexes have been recently investigated as novel anticancer agents. However, little is understood about their mechanisms of action, as well as the means by which cancer cells respond to chronic exposure to these compounds. To gain a deeper mechanistic insight into these rhenium anticancer agents, we developed and characterized an ovarian cancer cell line that is resistant to a previously studied compound [Re(CO)3 (dmphen)(ptolICN)]+ , where dmphen=2,9-dimethyl-1,10-phenanthroline and ptolICN=para-tolyl isonitrile, called TRIP. This TRIP-resistant ovarian cancer cell line, A2780TR, was found to be 9 times less sensitive to TRIP compared to the wild-type A2780 ovarian cancer cell line. Furthermore, the cytotoxicities of established drugs and other rhenium anticancer agents in the TRIP-resistant cell line were determined. Notably, the drug taxol was found to exhibit a 184-fold decrease in activity in the A2780TR cell line, suggesting that mechanisms of resistance towards TRIP and this drug are similar. Accordingly, expression levels of the ATP-binding cassette transporter P-glycoprotein, an efflux transporter known to detoxify taxol, were found to be elevated in the A2780TR cell line. Additionally, a gene expression analysis using the National Cancer Institute 60 cell line panel identified the MT1E gene to be overexpressed in cells that are less sensitive to TRIP. Because this gene encodes for metallothioneins, this result suggests that detoxification by this class of proteins is another mechanism for resistance to TRIP. The importance of this gene in the A2780TR cell line was assessed, confirming that its expression is elevated in this cell line as well. As the first study to investigate and identify the cancer cell resistance pathways in response to a rhenium complex, this report highlights important similarities and differences in the resistance responses of ovarian cancer cells to TRIP and conventional drugs.
Collapse
Affiliation(s)
- Sierra C. Marker
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - A. Paden King
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Robert V. Swanda
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, 14853, United States
| | - Brett Vaughn
- Department of Chemistry, Stony Brook University, Stony Brook, New York, 11794, United States
| | - Eszter Boros
- Department of Chemistry, Stony Brook University, Stony Brook, New York, 11794, United States
| | - Shu-Bing Qian
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, 14853, United States
| | - Justin J. Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
11
|
Marker SC, King AP, Swanda RV, Vaughn B, Boros E, Qian SB, Wilson JJ. Exploring ovarian cancer cell resistance to rhenium anticancer complexes. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 132:13493-13502. [PMID: 34366495 PMCID: PMC8340908 DOI: 10.1002/ange.202004883] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Indexed: 12/29/2022]
Abstract
Rhenium tricarbonyl complexes have been recently investigated as novel anticancer agents. However, little is understood about their mechanisms of action, as well as the means by which cancer cells respond to chronic exposure to these compounds. To gain a deeper mechanistic insight into these rhenium anticancer agents, we developed and characterized an ovarian cancer cell line that is resistant to a previously studied compound [Re(CO)3(dmphen)(ptolICN)]+, where dmphen = 2,9-dimethyl-1,10-phenanthroline and ptolICN = para-tolyl isonitrile, called TRIP. This TRIP-resistant ovarian cancer cell line, A2780TR, was found to be 9 times less sensitive to TRIP compared to the wild-type A2780 ovarian cancer cell line. Furthermore, the cytotoxicities of established drugs and other rhenium anticancer agents in the TRIP-resistant cell line were determined. Notably, the drug taxol was found to exhibit a 184-fold decrease in activity in the A2780TR cell line, suggesting that mechanisms of resistance towards TRIP and this drug are similar. Accordingly, expression levels of the ATP-binding cassette transporter P-glycoprotein, an efflux transporter known to detoxify taxol, were found to be elevated in the A2780TR cell line. Additionally, a gene expression analysis using the National Cancer Institute 60 cell line panel identified the MT1E gene to be overexpressed in cells that are less sensitive to TRIP. Because this gene encodes for metallothioneins, this result suggests that detoxification by this class of proteins is another mechanism for resistance to TRIP. The importance of this gene in the A2780TR cell line was assessed, confirming that its expression is elevated in this cell line as well. As the first study to investigate and identify the cancer cell resistance pathways in response to a rhenium complex, this report high-lights important similarities and differences in the resistance responses of ovarian cancer cells to TRIP and conventional drugs.
Collapse
Affiliation(s)
- Sierra C. Marker
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - A. Paden King
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Robert V. Swanda
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, 14853, United States
| | - Brett Vaughn
- Department of Chemistry, Stony Brook University, Stony Brook, New York, 11794, United States
| | - Eszter Boros
- Department of Chemistry, Stony Brook University, Stony Brook, New York, 11794, United States
| | - Shu-Bing Qian
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, 14853, United States
| | - Justin J. Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
12
|
Stultz LK, Hunsucker A, Middleton S, Grovenstein E, O'Leary J, Blatt E, Miller M, Mobley J, Hanson PK. Proteomic analysis of the S. cerevisiae response to the anticancer ruthenium complex KP1019. Metallomics 2020; 12:876-890. [PMID: 32329475 PMCID: PMC7362344 DOI: 10.1039/d0mt00008f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Like platinum-based chemotherapeutics, the anticancer ruthenium complex indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(iii)], or KP1019, damages DNA, induces apoptosis, and causes tumor regression in animal models. Unlike platinum-based drugs, KP1019 showed no dose-limiting toxicity in a phase I clinical trial. Despite these advances, the mechanism(s) and target(s) of KP1019 remain unclear. For example, the drug may damage DNA directly or by causing oxidative stress. Likewise, KP1019 binds cytosolic proteins, suggesting DNA is not the sole target. Here we use the budding yeast Saccharomyces cerevisiae as a model in a proteomic study of the cellular response to KP1019. Mapping protein level changes onto metabolic pathways revealed patterns consistent with elevated synthesis and/or cycling of the antioxidant glutathione, suggesting KP1019 induces oxidative stress. This result was supported by increased fluorescence of the redox-sensitive dye DCFH-DA and increased KP1019 sensitivity of yeast lacking Yap1, a master regulator of the oxidative stress response. In addition to oxidative and DNA stress, bioinformatic analysis revealed drug-dependent increases in proteins involved ribosome biogenesis, translation, and protein (re)folding. Consistent with proteotoxic effects, KP1019 increased expression of a heat-shock element (HSE) lacZ reporter. KP1019 pre-treatment also sensitized yeast to oxaliplatin, paralleling prior research showing that cancer cell lines with elevated levels of translation machinery are hypersensitive to oxaliplatin. Combined, these data suggest that one of KP1019's many targets may be protein metabolism, which opens up intriguing possibilities for combination therapy.
Collapse
Affiliation(s)
- Laura K Stultz
- Department of Chemistry, Birmingham-Southern College, Birmingham, AL 35254, USA
| | - Alexandra Hunsucker
- Department of Biology, Birmingham-Southern College, Birmingham, AL 35254, USA
| | - Sydney Middleton
- Department of Chemistry, Birmingham-Southern College, Birmingham, AL 35254, USA
| | - Evan Grovenstein
- Department of Biology, Birmingham-Southern College, Birmingham, AL 35254, USA
| | - Jacob O'Leary
- Department of Chemistry, Birmingham-Southern College, Birmingham, AL 35254, USA
| | - Eliot Blatt
- Department of Biology, Rhodes College, Memphis, TN 38112, USA
| | - Mary Miller
- Department of Biology, Rhodes College, Memphis, TN 38112, USA
| | - James Mobley
- Department of Surgery, University of Alabama at Birmingham, School of Medicine, Birmingham, AL 35294, USA
| | - Pamela K Hanson
- Department of Biology, Furman University, Greenville, SC 29613, USA.
| |
Collapse
|
13
|
Development and biological investigations of hypoxia-sensitive prodrugs of the tyrosine kinase inhibitor crizotinib. Bioorg Chem 2020; 99:103778. [PMID: 32229347 DOI: 10.1016/j.bioorg.2020.103778] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/21/2020] [Accepted: 03/18/2020] [Indexed: 12/24/2022]
Abstract
Despite the huge success of tyrosine kinase inhibitors as anticancer agents, severe side effects are a major problem. In order to overcome this drawback, the first hypoxia-activatable 2-nitroimidazole-based prodrugs of the clinically approved ALK and c-MET inhibitor crizotinib were developed. The 2-aminopyridine functionality of crizotinib (essential for target kinase binding) was considered as ideal position for prodrug derivatization. Consequently, two different prodrugs were synthesized with the nitroimidazole unit attached to crizotinib either via carbamoylation (A) or alkylation (B) of the 2-aminopyridine moiety. The successful prodrug design could be proven by docking studies and a dramatically reduced ALK and c-MET kinase-inhibitory potential. Furthermore, the prodrugs showed high stability in serum and release of crizotinib in an enzymatic nitroreductase-based cleavage assay was observed for prodrug A. The in vitro activity of both prodrugs was investigated against ALK- and c-MET-dependent or -overexpressing cells, revealing a distinct hypoxia-dependent activation for prodrug A. Finally, inhibition of c-MET phosphorylation and cell proliferation could also be proven in vivo. In summary of the theoretical, chemical and biological studies, prodrug derivatization of the 2-aminopyridine position can be considered as a promising strategy to reduce the side effects and improve the anticancer activity of crizotinib.
Collapse
|
14
|
Ravi C, Vuradi RK, Avudoddi S, Ramchander M, Satyanarayana S. Induction of Apoptosis in SKOV3 and DNA Binding by Cobalt(III) Polypyridyl Complexes. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2019. [DOI: 10.1134/s1068162019040095] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
15
|
NAMI-A and KP1019/1339, Two Iconic Ruthenium Anticancer Drug Candidates Face-to-Face: A Case Story in Medicinal Inorganic Chemistry. Molecules 2019; 24:molecules24101995. [PMID: 31137659 PMCID: PMC6571951 DOI: 10.3390/molecules24101995] [Citation(s) in RCA: 240] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 05/22/2019] [Indexed: 01/23/2023] Open
Abstract
NAMI-A ((ImH)[trans-RuCl4(dmso-S)(Im)], Im = imidazole) and KP1019/1339 (KP1019 = (IndH)[trans-RuCl4(Ind)2], Ind = indazole; KP1339 = Na[trans-RuCl4(Ind)2]) are two structurally related ruthenium(III) coordination compounds that have attracted a lot of attention in the medicinal inorganic chemistry scientific community as promising anticancer drug candidates. This has led to a considerable amount of studies on their respective chemico-biological features and to the eventual admission of both to clinical trials. The encouraging pharmacological performances qualified KP1019 mainly as a cytotoxic agent for the treatment of platinum-resistant colorectal cancers, whereas the non-cytotoxic NAMI-A has gained the reputation of being a very effective antimetastatic drug. A critical and strictly comparative analysis of the studies conducted so far on NAMI-A and KP1019 allows us to define the state of the art of these experimental ruthenium drugs in terms of the respective pharmacological profiles and potential clinical applications, and to gain some insight into the inherent molecular mechanisms. Despite their evident structural relatedness, deeply distinct biological and pharmacological profiles do emerge. Overall, these two iconic ruthenium complexes form an exemplary and unique case in the field of medicinal inorganic chemistry.
Collapse
|
16
|
Pötsch I, Baier D, Keppler BK, Berger W. Challenges and Chances in the Preclinical to Clinical Translation of Anticancer Metallodrugs. METAL-BASED ANTICANCER AGENTS 2019. [DOI: 10.1039/9781788016452-00308] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Despite being “sentenced to death” for quite some time, anticancer platinum compounds are still the most frequently prescribed cancer therapies in the oncological routine and recent exciting news from late-stage clinical studies on combinations of metallodrugs with immunotherapies suggest that this situation will not change soon. It is perhaps surprising that relatively simple molecules like cisplatin, discovered over 50 years ago, are still widely used clinically, while none of the highly sophisticated metal compounds developed over the last decade, including complexes with targeting ligands and multifunctional (nano)formulations, have managed to obtain clinical approval. In this book chapter, we summarize the current status of ongoing clinical trials for anticancer metal compounds and discuss the reasons for previous failures, as well as new opportunities for the clinical translation of metal complexes.
Collapse
Affiliation(s)
- Isabella Pötsch
- University of Vienna, Department of Inorganic Chemistry Währingerstrasse Vienna 1090 Austria
- Medical University of Vienna, Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I Borschkegasse 8a 1090 Vienna Austria
| | - Dina Baier
- University of Vienna, Department of Inorganic Chemistry Währingerstrasse Vienna 1090 Austria
- Medical University of Vienna, Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I Borschkegasse 8a 1090 Vienna Austria
| | - Bernhard K. Keppler
- University of Vienna, Department of Inorganic Chemistry Währingerstrasse Vienna 1090 Austria
| | - Walter Berger
- Medical University of Vienna, Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I Borschkegasse 8a 1090 Vienna Austria
| |
Collapse
|
17
|
|
18
|
Hager S, Korbula K, Bielec B, Grusch M, Pirker C, Schosserer M, Liendl L, Lang M, Grillari J, Nowikovsky K, Pape VFS, Mohr T, Szakács G, Keppler BK, Berger W, Kowol CR, Heffeter P. The thiosemicarbazone Me 2NNMe 2 induces paraptosis by disrupting the ER thiol redox homeostasis based on protein disulfide isomerase inhibition. Cell Death Dis 2018; 9:1052. [PMID: 30323190 PMCID: PMC6189190 DOI: 10.1038/s41419-018-1102-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 09/04/2018] [Accepted: 09/21/2018] [Indexed: 12/16/2022]
Abstract
Due to their high biological activity, thiosemicarbazones have been developed for treatment of diverse diseases, including cancer, resulting in multiple clinical trials especially of the lead compound Triapine. During the last years, a novel subclass of anticancer thiosemicarbazones has attracted substantial interest based on their enhanced cytotoxic activity. Increasing evidence suggests that the double-dimethylated Triapine derivative Me2NNMe2 differs from Triapine not only in its efficacy but also in its mode of action. Here we show that Me2NNMe2- (but not Triapine)-treated cancer cells exhibit all hallmarks of paraptotic cell death including, besides the appearance of endoplasmic reticulum (ER)-derived vesicles, also mitochondrial swelling and caspase-independent cell death via the MAPK signaling pathway. Subsequently, we uncover that the copper complex of Me2NNMe2 (a supposed intracellular metabolite) inhibits the ER-resident protein disulfide isomerase, resulting in a specific form of ER stress based on disruption of the Ca2+ and ER thiol redox homeostasis. Our findings indicate that compounds like Me2NNMe2 are of interest especially for the treatment of apoptosis-resistant cancer and provide new insights into mechanisms underlying drug-induced paraptosis.
Collapse
Affiliation(s)
- Sonja Hager
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
- Research Cluster "Translational Cancer Therapy Research", Vienna, Austria
| | - Katharina Korbula
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
- Research Cluster "Translational Cancer Therapy Research", Vienna, Austria
| | - Björn Bielec
- Research Cluster "Translational Cancer Therapy Research", Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Str. 42, A-1090, Vienna, Austria
| | - Michael Grusch
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Christine Pirker
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Markus Schosserer
- Department of Biotechnology, BOKU-University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, A-1190, Vienna, Austria
| | - Lisa Liendl
- Department of Biotechnology, BOKU-University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, A-1190, Vienna, Austria
| | - Magdalena Lang
- Department of Biotechnology, BOKU-University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, A-1190, Vienna, Austria
| | - Johannes Grillari
- Department of Biotechnology, BOKU-University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, A-1190, Vienna, Austria
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Muthgasse 18, A-1190, Vienna, Austria
- Evercyte GmbH, Muthgasse 18, A-1190, Vienna, Austria
| | - Karin Nowikovsky
- Department of Internal Medicine I and Comprehensive Cancer Center, Medical University of Vienna, Lazarettgasse 14, A-1090, Vienna, Austria
| | - Veronika F S Pape
- Department of Physiology, Faculty of Medicine, Semmelweis University, Tűzoltó utca 37-47, H-1094, Budapest, Hungary
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, H-1117, Budapest, Hungary
| | - Thomas Mohr
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
- Science Consult DI Thomas Mohr KG, Enzianweg 10a, A-2353, Guntramsdorf, Austria
| | - Gergely Szakács
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, H-1117, Budapest, Hungary
| | - Bernhard K Keppler
- Research Cluster "Translational Cancer Therapy Research", Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Str. 42, A-1090, Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
- Research Cluster "Translational Cancer Therapy Research", Vienna, Austria
| | - Christian R Kowol
- Research Cluster "Translational Cancer Therapy Research", Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Str. 42, A-1090, Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria.
- Research Cluster "Translational Cancer Therapy Research", Vienna, Austria.
| |
Collapse
|
19
|
Orlowska E, Babak MV, Dömötör O, Enyedy EA, Rapta P, Zalibera M, Bučinský L, Malček M, Govind C, Karunakaran V, Farid YCS, McDonnell TE, Luneau D, Schaniel D, Ang WH, Arion VB. NO Releasing and Anticancer Properties of Octahedral Ruthenium–Nitrosyl Complexes with Equatorial 1H-Indazole Ligands. Inorg Chem 2018; 57:10702-10717. [DOI: 10.1021/acs.inorgchem.8b01341] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Ewelina Orlowska
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Strasse 42, A-1090 Vienna, Austria
| | - Maria V. Babak
- Department of Chemistry, National University of Singapore, 3 Science Drive 2, 117543 Singapore
| | - Orsolya Dömötör
- Department of Inorganic and Analytical Chemistry, University of Szeged, Dom ter 7, H-6720 Szeged, Hungary
| | - Eva A. Enyedy
- Department of Inorganic and Analytical Chemistry, University of Szeged, Dom ter 7, H-6720 Szeged, Hungary
| | - Peter Rapta
- Slovak University of Technology, Institute of Physical Chemistry and Chemical Physics, Radlinského 9, SK-81237 Bratislava, Slovakia
| | - Michal Zalibera
- Slovak University of Technology, Institute of Physical Chemistry and Chemical Physics, Radlinského 9, SK-81237 Bratislava, Slovakia
| | - Lukáš Bučinský
- Slovak University of Technology, Institute of Physical Chemistry and Chemical Physics, Radlinského 9, SK-81237 Bratislava, Slovakia
| | - Michal Malček
- Slovak University of Technology, Institute of Physical Chemistry and Chemical Physics, Radlinského 9, SK-81237 Bratislava, Slovakia
- LAQV@REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Chinju Govind
- Photosciences and Photonics Section, Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram 695019 Kerala India
| | - Venugopal Karunakaran
- Photosciences and Photonics Section, Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram 695019 Kerala India
| | | | - Tara E. McDonnell
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Kensington, Sydney, New South Wales 2052, Australia
| | - Dominique Luneau
- Laboratoire des Multimatériaux et Interfaces (UMR5615), Université Claude Bernard Lyon 1, Campus de la Doua, 69622 Villeurbanne Cedex, France
| | | | - Wee Han Ang
- Department of Chemistry, National University of Singapore, 3 Science Drive 2, 117543 Singapore
| | - Vladimir B. Arion
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Strasse 42, A-1090 Vienna, Austria
| |
Collapse
|
20
|
Pal M, Nandi U, Mukherjee D. Detailed account on activation mechanisms of ruthenium coordination complexes and their role as antineoplastic agents. Eur J Med Chem 2018; 150:419-445. [DOI: 10.1016/j.ejmech.2018.03.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 03/02/2018] [Accepted: 03/03/2018] [Indexed: 10/17/2022]
|
21
|
Gichumbi JM, Friedrich HB, Omondi B, Lazarus GG, Singh M, Chenia HY. Synthesis, characterization, anticancer and antimicrobial study of arene ruthenium(II) complexes with 1,2,4-triazole ligands containing an α-diimine moiety. ACTA ACUST UNITED AC 2018. [DOI: 10.1515/znb-2017-0145] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
The reaction of the ruthenium arene dimers [(η
6-arene)Ru(μ-Cl)Cl]2 (where arene=benzene or p-cymene) with the ligands 4-benzylidene-3,5-di(2′-pyridyl)-4-amino-1,2,4-triazole (L1
), 2-methoxybenzylidene-3,5-di(2′-pyridyl)-4-amino-1,2,4-triazole (L2
), 4-methylbenzylidene-3,5-di(2′-pyridyl)-4-amino-1,2,4-triazole (L3
) and indole-3-carbaldehyde-3,5-di(2′-pyridyl)-4-amino-1,2,4-triazole (L4
) in a 1:2 ratio gives the new complexes [(η
6-arene)RuCl(L)]+ [arene=C6H6 (with L=L1(1), L2(3), L4(7), with PF6
− as a counter ion, and L4 (6), with Cl− as a counter ion) or p-cymene with L=L1(2), L2(4), L3(5), L4(8) with PF6
− as a counter ion]. All complexes were fully characterized using 1H and 13C NMR, elemental analyses, UV/Vis and IR spectroscopy. The single crystal X-ray structures of ligand L2
and complex 1 have been determined. The structure of 1 has the Ru atom coordinated with the arene group and to the N,N′-bidentate ligand and to the Cl atom. The arene group occupies the apex, while the ligand and the Cl atom are at the base of a pseudo-octahedral three-legged piano stool. The cytotoxicity of these mononuclear complexes was established in the human epithelial colorectal adenocarcinoma cell line (Caco-2) and for selectivity in the non-cancerous human embryonic kidney cell line (HEK293), using 5-fluorouracil (5-FU) as the reference anticancer drug. Compounds 1 and 7 were relatively inactive toward the Caco-2 tumor cells (IC50>200), while complexes 2–5 showed moderate anti-proliferative properties (IC50>100–200). Compound 6, however, displayed better anti-proliferative properties with an IC50 value lower than that of the reference drug, 5-FU, and was therefore further investigated for its antimicrobial activity against six Gram-positive and four Gram-negative bacteria.
Collapse
Affiliation(s)
- Joel M. Gichumbi
- School of Chemistry and Physics , University of KwaZulu-Natal , Private Bag X54001 , Durban 4000 , South Africa
| | - Holger B. Friedrich
- School of Chemistry and Physics , University of KwaZulu-Natal , Private Bag X54001 , Durban 4000 , South Africa
| | - Bernard Omondi
- School of Chemistry and Physics , University of KwaZulu-Natal , Private Bag X54001 , Durban 4000 , South Africa
| | - Geraldine G. Lazarus
- School of Life Sciences , University of KwaZulu-Natal , Private Bag X54001 , Durban 4000 , South Africa
| | - Moganavelli Singh
- School of Life Sciences , University of KwaZulu-Natal , Private Bag X54001 , Durban 4000 , South Africa
| | - Hafizah Y. Chenia
- School of Life Sciences , University of KwaZulu-Natal , Private Bag X54001 , Durban 4000 , South Africa
| |
Collapse
|
22
|
Nišavić M, Janjić GV, Hozić A, Petković M, Milčić MK, Vujčić Z, Cindrić M. Positive and negative nano-electrospray mass spectrometry of ruthenated serum albumin supported by docking studies: an integrated approach towards defining metallodrug binding sites on proteins. Metallomics 2018; 10:587-594. [DOI: 10.1039/c7mt00330g] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Negative mode nanoLC/nano ESI MS was used for determing Ru(ii) binding sites on protein.
Collapse
Affiliation(s)
- Marija Nišavić
- Department of Physical Chemistry
- “Vinča” Institute of Nuclear Sciences
- University of Belgrade
- 11000 Belgrade
- Serbia
| | - Goran V. Janjić
- Institute of Chemistry, Technology and Metallurgy
- University of Belgrade
- 11000 Belgrade
- Serbia
| | - Amela Hozić
- Centre for Proteomics and Mass Spectrometry
- Division of Molecular Medicine
- Ruđer Bošković Institute
- 10000 Zagreb
- Croatia
| | - Marijana Petković
- Department of Physical Chemistry
- “Vinča” Institute of Nuclear Sciences
- University of Belgrade
- 11000 Belgrade
- Serbia
| | - Miloš K. Milčić
- Faculty of Chemistry
- University of Belgrade
- 11000 Belgrade
- Serbia
| | - Zoran Vujčić
- Faculty of Chemistry
- University of Belgrade
- 11000 Belgrade
- Serbia
| | - Mario Cindrić
- Centre for Proteomics and Mass Spectrometry
- Division of Molecular Medicine
- Ruđer Bošković Institute
- 10000 Zagreb
- Croatia
| |
Collapse
|
23
|
Golla U, Swagatika S, Chauhan S, Tomar RS. A systematic assessment of chemical, genetic, and epigenetic factors influencing the activity of anticancer drug KP1019 (FFC14A). Oncotarget 2017; 8:98426-98454. [PMID: 29228701 PMCID: PMC5716741 DOI: 10.18632/oncotarget.21416] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/28/2017] [Indexed: 12/11/2022] Open
Abstract
KP1019 ([trans-RuCl4(1H-indazole)2]; FFC14A) is one of the promising ruthenium-based anticancer drugs undergoing clinical trials. Despite the pre-clinical and clinical success of KP1019, the mode of action and various factors capable of modulating its effects are largely unknown. Here, we used transcriptomics and genetic screening approaches in budding yeast model and deciphered various genetic targets and plethora of cellular pathways including cellular signaling, metal homeostasis, vacuolar transport, and lipid homeostasis that are primarily targeted by KP1019. We also demonstrated that KP1019 modulates the effects of TOR (target of rapamycin) signaling pathway and induces accumulation of neutral lipids (lipid droplets) in both yeast and HeLa cells. Interestingly, KP1019-mediated effects were found augmented with metal ions (Al3+/Ca2+/Cd2+/Cu2+/Mn2+/Na+/Zn2+), and neutralized by Fe2+, antioxidants, osmotic stabilizer, and ethanolamine. Additionally, our comprehensive screening of yeast histone H3/H4 mutant library revealed several histone residues that could significantly modulate the KP1019-induced toxicity. Altogether, our findings in both the yeast and HeLa cells provide molecular insights into mechanisms of action of KP1019 and various factors (chemical/genetic/epigenetic) that can alter the therapeutic efficiency of this clinically important anticancer drug.
Collapse
Affiliation(s)
- Upendarrao Golla
- Laboratory of Chromatin Biology, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Bhopal 462066, India
| | - Swati Swagatika
- Laboratory of Chromatin Biology, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Bhopal 462066, India
| | - Sakshi Chauhan
- Laboratory of Chromatin Biology, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Bhopal 462066, India
| | - Raghuvir Singh Tomar
- Laboratory of Chromatin Biology, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Bhopal 462066, India
| |
Collapse
|
24
|
Fuereder T, Berger W. Metal drugs become targeted. ESMO Open 2017; 2:e000239. [PMID: 29225925 PMCID: PMC5604710 DOI: 10.1136/esmoopen-2017-000239] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 01/10/2017] [Accepted: 06/29/2017] [Indexed: 11/28/2022] Open
Affiliation(s)
- Thorsten Fuereder
- Department of Medicine I and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Walter Berger
- Department of Medicine I, Institute of Cancer Research, Medical University Vienna, Vienna, Austria
| |
Collapse
|
25
|
Schoenhacker-Alte B, Mohr T, Pirker C, Kryeziu K, Kuhn PS, Buck A, Hofmann T, Gerner C, Hermann G, Koellensperger G, Keppler BK, Berger W, Heffeter P. Sensitivity towards the GRP78 inhibitor KP1339/IT-139 is characterized by apoptosis induction via caspase 8 upon disruption of ER homeostasis. Cancer Lett 2017; 404:79-88. [PMID: 28716523 DOI: 10.1016/j.canlet.2017.07.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/05/2017] [Accepted: 07/07/2017] [Indexed: 11/30/2022]
Abstract
The ruthenium drug and GRP78 inhibitor KP1339/IT-139 has already demonstrated promising anticancer activity in a phase I clinical trial. This study aimed to identify mechanisms underlying increased sensitivity to KP1339 treatment. Based on a screen utilizing 23 cell lines, a small panel was selected to compare KP1339-sensitive and low-responsive models. KP1339 sensitivity was neither based on differences in ruthenium accumulation, nor sensitivity to oxidative stress or constituents of KP1339 (ruthenium chloride and indazole). Subsequently, the biochemical response to KP1339 was analyzed using whole genome expression arrays indicating that, while sensitive cell lines were characterized by "response to chemical stimuli" and "regulation of cell death", low-responsive cells preferentially activated pathways controlling cell cycle, DNA repair, and metabolism. Cell culture experiments confirmed that, while low-responsive cells executed cell cycle arrest in G2 phase, pronounced apoptosis induction via activation of caspase 8 was found in sensitive cells. Cell death induction is based on a unique disruption of the ER homeostasis by depletion of key cellular chaperones including GRP78 in combination with enhanced KP1339-mediated protein damage.
Collapse
Affiliation(s)
- Beatrix Schoenhacker-Alte
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090, Vienna, Austria; Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Thomas Mohr
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Christine Pirker
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Kushtrim Kryeziu
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Paul-Steffen Kuhn
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090, Vienna, Austria
| | - Alicia Buck
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Thilo Hofmann
- Department of Environmental Geosciences, University of Vienna, Althanstraße 14, A-1090, Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, A-1090, Vienna, Austria
| | - Gerrit Hermann
- Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, A-1090, Vienna, Austria
| | - Gunda Koellensperger
- Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, A-1090, Vienna, Austria
| | - Bernhard K Keppler
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090, Vienna, Austria; Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Walter Berger
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria; Research Cluster "Translational Cancer Therapy Research", Vienna, Austria
| | - Petra Heffeter
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria; Research Cluster "Translational Cancer Therapy Research", Vienna, Austria.
| |
Collapse
|
26
|
Panchuk RR, Skorokhyd NR, Kozak YS, Lehka LV, Moiseenok AG, Stoika RS. Tissue-protective activity of selenomethionine and D-panthetine in B16 melanoma-bearing mice under doxorubicin treatment is not connected with their ROS scavenging potential. Croat Med J 2017; 58:171-184. [PMID: 28409500 PMCID: PMC5410729 DOI: 10.3325/cmj.2017.58.171] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Aim To evaluate molecular mechanisms of tissue-protective effects of antioxidants selenomethionine (SeMet) and D-pantethine (D-Pt) applied in combination with doxorubicin (Dx) in B16 melanoma-bearing-mice. Methods Impact of the chemotherapy scheme on a survival of tumor-bearing animals, general nephro- and hepatotoxicity, blood cell profile in vivo, and ROS content in B16 melanoma cells in vitro was compared with the action of Dx applied alone. Nephrotoxicity of the drugs was evaluated by measuring creatinine indicator assay, hepatotoxicity was studied by measuring the activity of ALT/AST enzymes, and myelotoxicity was assessed by light microscopic analysis of blood smears. Changes in ROS content in B16 melanoma cells under Dx, SeMet, and D-Pt action in vitro were measured by incubation with fluorescent dyes dihydrodichlorofluoresceindiacetate (DCFDA, H2O2-specific) and dihydroethidium (DHE, O2--specific), and further analysis at FL1 (DCFDA) or FL2 channels (DHE) of FACScan flow cytometer. The impact of aforementioned compounds on functional status of mitochondria was measured by Rhodamine 123 assay and further analysis at FL1 channel of FACScan flow cytometer. Results Selenomethionine (1200 µg/kg) and D-pantethine (500 mg/kg) in combination with Dx (10 mg/kg) significantly reduced tumor-induced neutrophilia, lymphocytopenia, and leukocytosis in comparison to Dx treatment alone. Moreover, SeMet and D-Pt decreased several side effects of Dx, namely an elevated creatinine level in blood and monocytosis, thus normalizing health conditions of B16 melanoma-bearing animals. Conclusions Our results showed that antioxidants selenomethionine and D-pantethine possess significant nephroprotective and myeloprotective activity toward Dx action on murine B16 melanoma in vivo, but fail to boost a survival of B16 melanoma-bearing animals. The observed cytoprotective effects of studied antioxidants are not directly connected with their ROS scavenging.
Collapse
Affiliation(s)
- Rostyslav R Panchuk
- Rostyslav R. Panchuk, Institute of Cell Biology, National Academy of Sciences of Ukraine, Drahomanov Street 14/16, 79005, Lviv, Ukraine,
| | | | | | | | | | | |
Collapse
|
27
|
Panchuk RR, Lehka LV, Terenzi A, Matselyukh BP, Rohr J, Jha AK, Downey T, Kril IJ, Herbacek I, van Schoonhoven S, Heffeter P, Stoika RS, Berger W. Rapid generation of hydrogen peroxide contributes to the complex cell death induction by the angucycline antibiotic landomycin E. Free Radic Biol Med 2017; 106:134-147. [PMID: 28189848 PMCID: PMC5552372 DOI: 10.1016/j.freeradbiomed.2017.02.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 02/06/2017] [Accepted: 02/08/2017] [Indexed: 01/14/2023]
Abstract
Landomycin E (LE) is an angucycline antibiotic produced by Streptomyces globisporus. Previously, we have shown a broad anticancer activity of LE which is, in contrast to the structurally related and clinically used anthracycline doxorubicin (Dx), only mildly affected by multidrug resistance-mediated drug efflux. In the present study, cellular and molecular mechanisms underlying the anticancer activity of landomycin E towards Jurkat T-cell leukemia cells were dissected focusing on the involvement of radical oxygen species (ROS). LE-induced apoptosis distinctly differed in several aspects from the one induced by Dx. Rapid generation of both extracellular and cell-derived hydrogen peroxide already at one hour drug exposure was observed in case of LE but not found before 24h for Dx. In contrast, Dx but not LE induced production of superoxide radicals. Mitochondrial damage, as revealed by JC-1 staining, was weakly enhanced already at 3h LE treatment and increased significantly with time. Accordingly, activation of the intrinsic apoptosis pathway initiator caspase-9 was not detectable before 12h exposure. In contrast, cleavage of the down-stream caspase substrate PARP-1 was clearly induced already at the three hour time point. Out of all caspases tested, only activation of effector caspase-7 was induced at this early time points paralleling the LE-induced oxidative burst. Accordingly, this massive cleavage of caspase-7 at early time points was inhibitable by the radical scavenger N-acetylcysteine (NAC). Additionally, only simultaneous inhibition of multiple caspases reduced LE-induced apoptosis. Specific scavengers of both H2O2 and OH• effectively decreased LE-induced ROS production, but only partially inhibited LE-induced apoptosis. In contrast, NAC efficiently blocked both parameters. Summarizing, rapid H2O2 generation and a complex caspase activation pattern contribute to the antileukemic effects of LE. As superoxide generation is considered as the main cardiotoxic mechanism of Dx, LE might represent a better tolerable drug candidate for further (pre)clinical development.
Collapse
Affiliation(s)
| | - Lilya V Lehka
- Institute of Cell Biology NAS of Ukraine, Lviv, Ukraine
| | - Alessio Terenzi
- Institute of Inorganic Chemistry, University of Vienna, Vienna, Austria
| | - Bohdan P Matselyukh
- D.K. Zabolotny Institute of Microbiology and Virology NAS of Ukraine, Lviv, Ukaine
| | - Jürgen Rohr
- University of Kentucky, College of Pharmacy, Lexington, USA
| | - Amit K Jha
- University of Kentucky, College of Pharmacy, Lexington, USA
| | - Theresa Downey
- University of Kentucky, College of Pharmacy, Lexington, USA
| | - Iryna J Kril
- Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Irene Herbacek
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Sushilla van Schoonhoven
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | | | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
28
|
Electronic State of Sodium trans-[Tetrachloridobis(1H-indazole)ruthenate(III)] (NKP-1339) in Tumor, Liver and Kidney Tissue of a SW480-bearing Mouse. Sci Rep 2017; 7:40966. [PMID: 28112202 PMCID: PMC5256101 DOI: 10.1038/srep40966] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/13/2016] [Indexed: 12/19/2022] Open
Abstract
Ruthenium complexes are promising candidates for anticancer agents, especially NKP-1339 (sodium trans-[tetrachloridobis(1H-indazole)ruthenate(III)]), which is on the edge to clinical applications. The anticancer mechanism seems to be tightly linked to the redox chemistry but despite progress in human clinical trials the in vivo Ru oxidation state and the coordination of Ru remains unclear. The Ru-based anticancer drug NKP-1339 was studied applying XANES (Cl K- and Ru L2,3-edges) in tumor, kidney and liver tissue of a SW480 bearing mouse. Based on coordination charge and 3D XANES plots containing a series of model compounds as well as pre-edge analysis of the ligand Cl K-edge it is suggested that NKP-1339 remains in its +III oxidation state after 24 hours and at least one of the four chlorido ligands remain covalently bound to the Ru ion showing a biotransformation from RuIIIN2Cl4 to RuIIIClx(N/O)6−x (X = 1 or 2).
Collapse
|
29
|
Inorganic mesoporous silicas as vehicles of two novel anthracene-based ruthenium metalloarenes. J Inorg Biochem 2016; 166:87-93. [PMID: 27838582 DOI: 10.1016/j.jinorgbio.2016.11.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/14/2016] [Accepted: 11/03/2016] [Indexed: 12/19/2022]
Abstract
Two novel anthracene-based half-sandwich organometallic Ru(II) compounds, namely, [Ru(p-cymene)(L1)Cl2] (1) and [Ru(p-cymene)(L2)Cl2] (2) (L1=1-(anthracen-9-yl)-N-(pyridin-3-ylmethyl)methanamine; L2=1-(anthracen-9-yl)-N-(pyridin-4-ylmethyl)methanamine) have been synthesized and characterized. We demonstrate that the fluorescence properties of these complexes are highly affected by the linking position of the anthracene unit, as only 2 shows fluorescence emission in the blue region. Regarding their biological activity, both ruthenium metallodrugs show interaction with different biological targets such as S-donor amino acids (cysteine) and proteases (cysteine cathepsin B). Moreover, 1 and 2 show in vitro cytotoxicity against HL-60 cancer cell line (IC50=84.5 and 87.0μM for 1 and 2, respectively), with cell death occurring via apoptosis. Further studies have shown that diffusion into cells is the main mechanism of metallodrug uptake. Finally, as a proof of concept, these ruthenium complexes have been successfully encapsulated into MCM-41 and SBA-15 mesoporous silicas using two different incorporation strategies (impregnation and grinding).
Collapse
|
30
|
Bijelic A, Theiner S, Keppler BK, Rompel A. X-ray Structure Analysis of Indazolium trans-[Tetrachlorobis(1H-indazole)ruthenate(III)] (KP1019) Bound to Human Serum Albumin Reveals Two Ruthenium Binding Sites and Provides Insights into the Drug Binding Mechanism. J Med Chem 2016; 59:5894-903. [PMID: 27196130 PMCID: PMC4921950 DOI: 10.1021/acs.jmedchem.6b00600] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
![]()
Ruthenium(III) complexes are promising
candidates for anticancer
drugs, especially the clinically studied indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (KP1019)
and its analogue sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (NKP-1339). Several studies have
emphasized the likely role of human serum proteins in the transportation
and accumulation of ruthenium(III) complexes in tumors. Therefore,
the interaction between KP1019 and human serum albumin was investigated
by means of X-ray crystallography and inductively coupled plasma mass
spectrometry (ICP-MS). The structural data unambiguously reveal the
binding of two ruthenium atoms to histidine residues 146 and 242,
which are both located within well-known hydrophobic binding pockets
of albumin. The ruthenium centers are octahedrally coordinated by
solvent molecules revealing the dissociation of both indazole ligands
from the ruthenium-based drug. However, a binding mechanism is proposed
indicating the importance of the indazole ligands for binding site
recognition and thus their indispensable role for the binding of KP1019.
Collapse
Affiliation(s)
- Aleksandar Bijelic
- Fakultät für Chemie, Institut für Biophysikalische Chemie, Universität Wien , Althanstraße 14, 1090 Wien, Austria
| | | | | | - Annette Rompel
- Fakultät für Chemie, Institut für Biophysikalische Chemie, Universität Wien , Althanstraße 14, 1090 Wien, Austria
| |
Collapse
|
31
|
Molecular mode of action of NKP-1339 - a clinically investigated ruthenium-based drug - involves ER- and ROS-related effects in colon carcinoma cell lines. Invest New Drugs 2016; 34:261-8. [PMID: 26988975 PMCID: PMC4859864 DOI: 10.1007/s10637-016-0337-8] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 03/01/2016] [Indexed: 12/15/2022]
Abstract
Sodium trans-[tetrachloridobis(1H-indazole)ruthenate(III)] (NKP-1339) is a clinically investigated ruthenium-based metal complex, which shows promising results in solid tumors, such as non-small cell lung cancer, colorectal carcinoma, and most distinctively in gastrointestinal neuroendocrine tumors. In previous studies, fast binding to albumin as well as transferrin could be shown. The enhanced permeability and retention (EPR) effect, which is diversely being exploited for tumor targeting, could therefore be applicable for NKP-1339. Here we studied the serum dependence of its biological activity in various methods, influencing its cellular accumulation, cytotoxicity as well as the generation of reactive oxygen species (ROS). ROS lead to Nrf2 activation, which is known to activate antioxidant response gene transcription. GRP78 down-regulation on the protein level suggests ER associated protein degradation (ERAD) as a mode of action, as RNA levels are only mildly affected. Another important part for the mode of action is endoplasmic reticulum (ER) stress, as different factors are highly upregulated on the protein level. For example PERK, a transmembrane receptor which is released by GRP78 when the ER is disturbed, is upregulated and phosphorylated. EIF2α is phosphorylated, which leads to an inhibition of CAP-dependent translation and other stress responses. The transcription factor CHOP (DDIT3), which promotes ER stress dependent apoptosis, is time and concentration dependently upregulated. Finally cytotoxicity tests could prove that inhibition of ER stress and ER stress-mediated apoptosis leads to decreased cytotoxic effects of NKP-1339, which highlights the involvement of this mechanism in the mode of action.
Collapse
|
32
|
DNA Damage Response Checkpoint Activation Drives KP1019 Dependent Pre-Anaphase Cell Cycle Delay in S. cerevisiae. PLoS One 2015; 10:e0138085. [PMID: 26375390 PMCID: PMC4572706 DOI: 10.1371/journal.pone.0138085] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 08/25/2015] [Indexed: 12/19/2022] Open
Abstract
Careful regulation of the cell cycle is required for proper replication, cell division, and DNA repair. DNA damage–including that induced by many anticancer drugs–results in cell cycle delay or arrest, which can allow time for repair of DNA lesions. Although its molecular mechanism of action remains a matter of debate, the anticancer ruthenium complex KP1019 has been shown to bind DNA in biophysical assays and to damage DNA of colorectal and ovarian cancer cells in vitro. KP1019 has also been shown to induce mutations and induce cell cycle arrest in Saccharomyces cerevisiae, suggesting that budding yeast can serve as an appropriate model for characterizing the cellular response to the drug. Here we use a transcriptomic approach to verify that KP1019 induces the DNA damage response (DDR) and find that KP1019 dependent expression of HUG1 requires the Dun1 checkpoint; both consistent with KP1019 DDR in budding yeast. We observe a robust KP1019 dependent delay in cell cycle progression as measured by increase in large budded cells, 2C DNA content, and accumulation of Pds1 which functions to inhibit anaphase. Importantly, we also find that deletion of RAD9, a gene required for the DDR, blocks drug-dependent changes in cell cycle progression, thereby establishing a causal link between the DDR and phenotypes induced by KP1019. Interestingly, yeast treated with KP1019 not only delay in G2/M, but also exhibit abnormal nuclear position, wherein the nucleus spans the bud neck. This morphology correlates with short, misaligned spindles and is dependent on the dynein heavy chain gene DYN1. We find that KP1019 creates an environment where cells respond to DNA damage through nuclear (transcriptional changes) and cytoplasmic (motor protein activity) events.
Collapse
|
33
|
Martins A, Sipos P, Dér K, Csábi J, Miklos W, Berger W, Zalatnai A, Amaral L, Molnár J, Szabó-Révész P, Hunyadi A. Ecdysteroids sensitize MDR and non-MDR cancer cell lines to doxorubicin, paclitaxel, and vincristine but tend to protect them from cisplatin. BIOMED RESEARCH INTERNATIONAL 2015; 2015:895360. [PMID: 26075272 PMCID: PMC4449901 DOI: 10.1155/2015/895360] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 02/25/2015] [Accepted: 03/10/2015] [Indexed: 01/14/2023]
Abstract
Ecdysteroids, analogs of the insect molting hormone, are known for their various mild, nonhormonal bioactivities in mammals. Previously, we reported that less-polar ecdysteroids can modulate the doxorubicin resistance of a multidrug resistant (MDR) mouse lymphoma cell line expressing the human ABCB1 transporter. Here, we describe the ability of 20-hydroxyecdysone (1) and its mono- (2) and diacetonide (3) derivatives to sensitize various MDR and non-MDR cancer cell lines towards doxorubicin, paclitaxel, vincristine, or cisplatin. Drug IC50 values with or without ecdysteroid were determined by MTT assay. Compound 3 significantly sensitized all cell lines to each chemotherapeutic except for cisplatin, whose activity was decreased. In order to overcome solubility and stability issues for the future in vivo administration of compound 3, liposomal formulations were developed. By means of their combination index values obtained via checkerboard microplate method, a formulation showed superior activity to that of compound 3 alone. Because ecdysteroids act also on non-ABCB1 expressing (sensitive) cell lines, our results demonstrate that they do not or not exclusively exert their adjuvant anticancer activity as ABCB1 inhibitors, but other mechanisms must be involved, and they opened the way towards their in vivo bioactivity testing against various cancer xenografts.
Collapse
Affiliation(s)
- Ana Martins
- Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, Dóm tér 10, Szeged 6720, Hungary
- Unidade de Parasitologia e Microbiologia Médica, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira 100, 1349-008 Lisbon, Portugal
| | - Péter Sipos
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Szeged, Eötvös Ucta 6, Szeged 6720, Hungary
| | - Katalin Dér
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Szeged, Eötvös Ucta 6, Szeged 6720, Hungary
| | - József Csábi
- Institute of Pharmacognosy, Faculty of Pharmacy, University of Szeged, Eötvös Ucta 6, Szeged 6720, Hungary
| | - Walter Miklos
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8A, 1090 Vienna, Austria
| | - Walter Berger
- Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8A, 1090 Vienna, Austria
- Institute of Inorganic Chemistry and Research Platform “Translational Cancer Therapy Research”, University of Vienna, Waehringer Strasse 42, 1090 Vienna, Austria
| | - Attila Zalatnai
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Leonard Amaral
- Centro de Malária e Outras Doenças Tropicais (CMDT), Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira 100, 1349-008 Lisbon, Portugal
| | - Joseph Molnár
- Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, Dóm tér 10, Szeged 6720, Hungary
| | - Piroska Szabó-Révész
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Szeged, Eötvös Ucta 6, Szeged 6720, Hungary
| | - Attila Hunyadi
- Institute of Pharmacognosy, Faculty of Pharmacy, University of Szeged, Eötvös Ucta 6, Szeged 6720, Hungary
| |
Collapse
|
34
|
Seršen S, Kljun J, Kryeziu K, Panchuk R, Alte B, Körner W, Heffeter P, Berger W, Turel I. Structure-Related Mode-of-Action Differences of Anticancer Organoruthenium Complexes with β-Diketonates. J Med Chem 2015; 58:3984-96. [PMID: 25856666 DOI: 10.1021/acs.jmedchem.5b00288] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
A series of organoruthenium(II) chlorido complexes with fluorinated O,O-ligands [(η(6)-p-cymene)Ru(F3C-acac-Ar)Cl] (1a-6a) and their respective 1,3,5-triaza-7-phosphaadamantane (pta) derivatives [(η(6)-p-cymene)Ru(F3C-acac-Ar)pta]PF6 (1b-6b) were synthesized and fully characterized in both solution and solid state. All complexes were inactive against nonmalignant keratinocytes but displayed variable activity against cancer cell models (ovarian, osteosarcoma). Compounds with a ligand containing the 4-chlorophenyl substituent (6a and 6b) exhibited the strongest anticancer effects. Despite a marginally lower cellular Ru accumulation compared to the chlorido complexes, pta analogues showed higher activity especially in the osteosarcoma model. Reduction of glutathione levels by buthionine sulfoximine (BSO) significantly enhanced the activity of all compounds with the most pronounced effects being observed for the pta series resulting in IC50 values down to the nanomolar range. While all chlorido complexes potently induce reactive oxygen species, DNA damage, and apoptosis, the respective pta compounds widely lacked ROS production but blocked cell cycle progression in G0/G1 phase.
Collapse
Affiliation(s)
- Sara Seršen
- †Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Jakob Kljun
- †Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Kushtrim Kryeziu
- ‡Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Rostyslav Panchuk
- §Institute of Cell Biology, National Academy of Sciences of Ukraine, Lviv, 79005, Ukraine
| | - Beatrix Alte
- ‡Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Wilfried Körner
- ∥Department of Environmental Geosciences, University of Vienna, 1090, Vienna, Austria
| | - Petra Heffeter
- ‡Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Walter Berger
- ‡Department of Medicine I, Institute of Cancer Research and Comprehensive Cancer Center, Medical University Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Iztok Turel
- †Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
35
|
Matczuk M, Kupiec M, Legat J, Pawlak K, Timerbaev AR, Jarosz M. A shotgun metalloproteomic approach enables identification of proteins involved in the speciation of a ruthenium anticancer drug in the cytosol of cancer cells. Analyst 2015; 140:3492-9. [DOI: 10.1039/c5an00426h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Development of a versatile analytical methodology for characterization of the cancer cytosol species formed between ruthenium originating from a Ru(iii) drug and cellular proteins.
Collapse
Affiliation(s)
- Magdalena Matczuk
- Chair of Analytical Chemistry
- Faculty of Chemistry
- Warsaw University of Technology
- 00-664 Warsaw
- Poland
| | - Monika Kupiec
- Chair of Analytical Chemistry
- Faculty of Chemistry
- Warsaw University of Technology
- 00-664 Warsaw
- Poland
| | - Joanna Legat
- Chair of Analytical Chemistry
- Faculty of Chemistry
- Warsaw University of Technology
- 00-664 Warsaw
- Poland
| | - Katarzyna Pawlak
- Chair of Analytical Chemistry
- Faculty of Chemistry
- Warsaw University of Technology
- 00-664 Warsaw
- Poland
| | - Andrei R. Timerbaev
- Vernadsky Institute of Geochemistry and Analytical Chemistry
- 119991 Moscow
- Russian Federation
| | - Maciej Jarosz
- Chair of Analytical Chemistry
- Faculty of Chemistry
- Warsaw University of Technology
- 00-664 Warsaw
- Poland
| |
Collapse
|
36
|
Jones MR, Mu C, Wang MCP, Webb MI, Walsby CJ, Storr T. Modulation of the Aβ peptide aggregation pathway by KP1019 limits Aβ-associated neurotoxicity. Metallomics 2014; 7:129-35. [PMID: 25387614 DOI: 10.1039/c4mt00252k] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is increasing worldwide due to increased life expectancy. AD is characterized by two pathological hallmarks in the brain: amyloid-β (Aβ) plaque deposits and neurofibrillary tangles. A focus of AD research has concentrated on either inhibiting Aβ peptide aggregation that leads to plaque formation or breaking down pre-formed Aβ peptide aggregates. An alternative approach is to modulate the Aβ aggregation profile by facilitating the formation of Aβ species that are off-pathway and non-toxic. Herein, we report the re-purposing of the widely studied Ru(iii) anti-cancer complex KP1019, towards regulating the aggregation profile of the Aβ peptide. Using electron paramagnetic resonance (EPR) spectroscopy, we conclude that KP1019 binds to histidine residues, located at the N-terminus of the peptide, in a rapid and robust fashion. Native gels and transmission electron microscopy (TEM) analyses have provided insight into the species and structures that are generated by KP1019-Aβ interactions. Finally, incubation in an in vitro human neuronal cell model has demonstrated that the formation of KP1019-Aβ species rescues cell viability from Aβ-associated neurotoxicity. Modulation of the Aβ aggregation pathway via covalent interactions with small molecules is thus a promising AD therapeutic strategy.
Collapse
Affiliation(s)
- Michael R Jones
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A1S6, Canada.
| | | | | | | | | | | |
Collapse
|
37
|
Spreckelmeyer S, Orvig C, Casini A. Cellular transport mechanisms of cytotoxic metallodrugs: an overview beyond cisplatin. Molecules 2014; 19:15584-610. [PMID: 25268716 PMCID: PMC6271550 DOI: 10.3390/molecules191015584] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 09/17/2014] [Accepted: 09/22/2014] [Indexed: 12/21/2022] Open
Abstract
The field of medicinal inorganic chemistry has grown consistently during the past 50 years; however, metal-containing coordination compounds represent only a minor proportion of drugs currently on the market, indicating that research in this area has not yet been thoroughly realized. Although platinum-based drugs as cancer chemotherapeutic agents have been widely studied, exact knowledge of the mechanisms governing their accumulation in cells is still lacking. However, evidence suggests active uptake and efflux mechanisms are involved; this may be involved also in other experimental metal coordination and organometallic compounds with promising antitumor activities in vitro and in vivo, such as ruthenium and gold compounds. Such knowledge would be necessary to elucidate the balance between activity and toxicity profiles of metal compounds. In this review, we present an overview of the information available on the cellular accumulation of Pt compounds from in vitro, in vivo and clinical studies, as well as a summary of reports on the possible accumulation mechanisms for different families of experimental anticancer metal complexes (e.g., Ru Au and Ir). Finally, we discuss the need for rationalization of the investigational approaches available to study metallodrug cellular transport.
Collapse
Affiliation(s)
- Sarah Spreckelmeyer
- Dept. Pharmacokinetics, Toxicology and Targeting, Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, The Netherlands
| | - Chris Orvig
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T1Z1, Canada
| | - Angela Casini
- Dept. Pharmacokinetics, Toxicology and Targeting, Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, The Netherlands.
| |
Collapse
|
38
|
Anti-cancer drug KP1019 induces Hog1 phosphorylation and protein ubiquitylation in Saccharomyces cerevisiae. Eur J Pharmacol 2014; 736:77-85. [DOI: 10.1016/j.ejphar.2014.04.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/22/2014] [Accepted: 04/23/2014] [Indexed: 01/29/2023]
|
39
|
Jungwirth U, Gojo J, Tuder T, Walko G, Holcmann M, Schöfl T, Nowikovsky K, Wilfinger N, Schoonhoven S, Kowol CR, Lemmens-Gruber R, Heffeter P, Keppler BK, Berger W. Calpain-Mediated Integrin Deregulation as a Novel Mode of Action for the Anticancer Gallium Compound KP46. Mol Cancer Ther 2014; 13:2436-49. [DOI: 10.1158/1535-7163.mct-14-0087] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
40
|
Trondl R, Heffeter P, Kowol CR, Jakupec MA, Berger W, Keppler BK. NKP-1339, the first ruthenium-based anticancer drug on the edge to clinical application. Chem Sci 2014. [DOI: 10.1039/c3sc53243g] [Citation(s) in RCA: 489] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
41
|
Anti-cancer drug KP1019 modulates epigenetics and induces DNA damage response inSaccharomyces cerevisiae. FEBS Lett 2014; 588:1044-52. [DOI: 10.1016/j.febslet.2014.02.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 01/28/2014] [Accepted: 02/05/2014] [Indexed: 12/11/2022]
|
42
|
Hummer AA, Rompel A. The use of X-ray absorption and synchrotron based micro-X-ray fluorescence spectroscopy to investigate anti-cancer metal compounds in vivo and in vitro. Metallomics 2013; 5:597-614. [PMID: 23558305 DOI: 10.1039/c3mt20261e] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
X-ray absorption spectroscopy (XAS) and micro-synchrotron based X-ray fluorescence (micro-SXRF) are element specific spectroscopic techniques and have been proven to be valuable tools for the investigation of changes in the chemical environment of metal centres. XAS allows the determination of the oxidation state, the coordination motif of the probed element, the identity and the number of adjacent atoms and the absorber-ligand distances. It is further applicable to nearly all types of samples independent of their actual physical state (solid, liquid, gaseous) down to μM concentrations. Micro-SXRF can provide information on the distribution and concentration of multiple elements within a sample simultaneously, allowing for the chemical state of several elements within subcellular compartments to be probed. Modern third generation synchrotrons offer the possibility to investigate the majority of the biologically relevant elements. The biological mode of action of metal-based compounds often involves interactions with target and/or transport molecules. The presence of reducing agents may also give rise to changes in the coordination sphere and/or the oxidation state. XAS and micro-SXRF are ideal techniques for investigating these issues. This tutorial review introduces the use of XAS and micro-SXRF techniques into the field of inorganic medicinal chemistry. The results obtained for platinum, ruthenium, gallium, gold and cobalt compounds within the last few years are presented.
Collapse
Affiliation(s)
- Alfred A Hummer
- Institut für Biophysikalische Chemie, Universität Wien, Althanstr. 14, 1090 Wien, Austria
| | | |
Collapse
|
43
|
Webb MI, Wu B, Jang T, Chard RA, Wong EWY, Wong MQ, Yapp DTT, Walsby CJ. Increasing the Bioavailability of RuIIIAnticancer Complexes through Hydrophobic Albumin Interactions. Chemistry 2013; 19:17031-42. [DOI: 10.1002/chem.201302671] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Indexed: 11/08/2022]
|
44
|
Bury M, Novo-Uzal E, Andolfi A, Cimini S, Wauthoz N, Heffeter P, Lallemand B, Avolio F, Delporte C, Cimmino A, Dubois J, Van Antwerpen P, Zonno MC, Vurro M, Poumay Y, Berger W, Evidente A, De Gara L, Kiss R, Locato V. Ophiobolin A, a sesterterpenoid fungal phytotoxin, displays higher in vitro growth-inhibitory effects in mammalian than in plant cells and displays in vivo antitumor activity. Int J Oncol 2013; 43:575-85. [PMID: 23754298 DOI: 10.3892/ijo.2013.1979] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 03/21/2013] [Indexed: 11/06/2022] Open
Abstract
Ophiobolin A, a sesterterpenoid produced by plant pathogenic fungi, was purified from the culture extract of Drechslera gigantea and tested for its growth-inhibitory activity in both plant and mammalian cells. Ophiobolin A induced cell death in Nicotiana tabacum L. cv. Bright Yellow 2 (TBY-2) cells at concentrations ≥10 µM, with the TBY-2 cells showing typical features of apoptosis-like cell death. At a concentration of 5 µM, ophiobolin A did not affect plant cell viability but prevented cell proliferation. When tested on eight cancer cell lines, concentrations <1 µM of ophiobolin A inhibited growth by 50% after 3 days of culture irrespective of their multidrug resistance (MDR) phenotypes and their resistance levels to pro-apoptotic stimuli. It is, thus, unlikely that ophiobolin A exerts these in vitro growth-inhibitory effects in cancer cells by activating pro-apoptotic processes. Highly proliferative human keratinocytes appeared more sensitive to the growth-inhibitory effects of ophiobolin A than slowly proliferating ones. Ophiobolin A also displayed significant antitumor activity at the level of mouse survival when assayed at 10 mg/kg in the B16F10 mouse melanoma model with lung pseudometastases. Ophiobolin A could, thus, represent a novel scaffold to combat cancer types that display various levels of resistance to pro-apoptotic stimuli and/or various MDR phenotypes.
Collapse
Affiliation(s)
- Marina Bury
- Laboratoire de Toxicologie, Faculté de Pharmacie, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Heffeter P, Atil B, Kryeziu K, Groza D, Koellensperger G, Körner W, Jungwirth U, Mohr T, Keppler BK, Berger W. The ruthenium compound KP1339 potentiates the anticancer activity of sorafenib in vitro and in vivo. Eur J Cancer 2013; 49:3366-75. [PMID: 23790465 PMCID: PMC3807657 DOI: 10.1016/j.ejca.2013.05.018] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 05/20/2013] [Indexed: 10/30/2022]
Abstract
KP1339 is a promising ruthenium-based anticancer compound in early clinical development. This study aimed to test the effects of KP1339 on the in vitro and in vivo activity of the multi-kinase inhibitor sorafenib, the current standard first-line therapy for advanced hepatoma. Anticancer activity of the parental compounds as compared to the drug combination was tested against a panel of cancer cell lines with a focus on hepatoma. Combination of KP1339 with sorafenib induced in the majority of all cases distinctly synergistic effects, comprising both sorafenib-resistant as well as sorafenib-responsive cell models. Several mechanisms were found to underlie these multifaceted synergistic activities. Firstly, co-exposure induced significantly enhanced accumulation levels of both drugs resulting in enhanced apoptosis induction. Secondly, sorafenib blocked KP1339-mediated activation of P38 signalling representing a protective response against the ruthenium drug. In addition, sorafenib treatment also abrogated KP1339-induced G2/M arrest but resulted in check point-independent DNA-synthesis block and a complete loss of the mitotic cell populations. The activity of the KP1339/sorafenib combination was evaluated in the Hep3B hepatoma xenograft. KP1339 monotherapy led to a 2.4-fold increase in life span and, thus, was superior to sorafenib, which induced a 1.9-fold prolonged survival. The combined therapy further enhanced the mean survival by 3.9-fold. Synergistic activity was also observed in the VM-1 melanoma xenograft harbouring an activating braf mutation. Together, our data indicate that the combination of KP1339 with sorafenib displays promising activity in vitro and in vivo especially against human hepatoma models.
Collapse
Affiliation(s)
- Petra Heffeter
- Institute of Cancer Research, Department of Medicine I, Medical University Vienna, Austria; Comprehensive Cancer Center of the Medical University Vienna, Austria; Research and Platform "Translational Cancer Therapy Research" Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Hummer AA, Heffeter P, Berger W, Filipits M, Batchelor D, Büchel GE, Jakupec MA, Keppler BK, Rompel A. X-ray absorption near edge structure spectroscopy to resolve the in vivo chemistry of the redox-active indazolium trans-[Tetrachlorobis(1H-indazole)ruthenate(III)] (KP1019). J Med Chem 2013; 56:1182-96. [PMID: 23282017 PMCID: PMC3579476 DOI: 10.1021/jm301648f] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
![]()
Indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (1, KP1019) and
its analogue
sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)]
(2, KP1339) are promising redox-active anticancer drug
candidates that were investigated with X-ray absorption near edge
structure spectroscopy. The analysis was based on the concept of the
coordination charge and ruthenium model compounds representing possible
coordinations and oxidation states in vivo. 1 was investigated
in citrate saline buffer (pH 3.5) and in carbonate buffer (pH 7.4)
at 37 °C for different time intervals. Interaction studies on 1 with glutathione in saline buffer and apo-transferrin in
carbonate buffer were undertaken, and the coordination of 1 and 2 in tumor tissues was studied too. The most likely
coordinations and oxidation states of the compound under the above
mentioned conditions were assigned. Microprobe X-ray fluorescence
of tumor thin sections showed the strong penetration of ruthenium
into the tumor tissue, with the highest concentrations near blood
vessels and in the edge regions of the tissue samples.
Collapse
Affiliation(s)
- Alfred A Hummer
- Institut für Biophysikalische Chemie, Universität Wien, Althanstrasse 14, 1090 Wien, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Stevens SK, Strehle AP, Miller RL, Gammons SH, Hoffman KJ, McCarty JT, Miller ME, Stultz LK, Hanson PK. The anticancer ruthenium complex KP1019 induces DNA damage, leading to cell cycle delay and cell death in Saccharomyces cerevisiae. Mol Pharmacol 2013; 83:225-34. [PMID: 23090979 DOI: 10.1124/mol.112.079657] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The anticancer ruthenium complex trans-[tetrachlorobis(1H-indazole)ruthenate(III)], otherwise known as KP1019, has previously been shown to inhibit proliferation of ovarian tumor cells, induce DNA damage and apoptosis in colon carcinoma cells, and reduce tumor size in animal models. Notably, no dose-limiting toxicity was observed in a Phase I clinical trial. Despite these successes, KP1019's precise mechanism of action remains poorly understood. To determine whether Saccharomyces cerevisiae might serve as an effective model for characterizing the cellular response to KP1019, we first confirmed that this drug is internalized by yeast and induces mutations, cell cycle delay, and cell death. We next examined KP1019 sensitivity of strains defective in DNA repair, ultimately showing that rad1Δ, rev3Δ, and rad52Δ yeast are hypersensitive to KP1019, suggesting that nucleotide excision repair (NER), translesion synthesis (TLS), and recombination each play a role in drug tolerance. These data are consistent with published work showing that KP1019 causes interstrand cross-links and bulky DNA adducts in mammalian cell lines. Published research also showed that mammalian cell lines resistant to other chemotherapeutic agents exhibit only modest resistance, and sometimes hypersensitivity, to KP1019. Here we report similar findings for S. cerevisiae. Whereas gain-of-function mutations in the transcription activator-encoding gene PDR1 are known to increase expression of drug pumps, causing resistance to structurally diverse toxins, we now demonstrate that KP1019 retains its potency against yeast carrying the hypermorphic alleles PDR1-11 or PDR1-3. Combined, these data suggest that S. cerevisiae could serve as an effective model system for identifying evolutionarily conserved modulators of KP1019 sensitivity.
Collapse
Affiliation(s)
- Shannon K Stevens
- Department of Biology, Birmingham-Southern College, 900 Arkadelphia Rd. Birmingham, AL 35254, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Heffeter P, Pirker C, Kowol CR, Herrman G, Dornetshuber R, Miklos W, Jungwirth U, Koellensperger G, Keppler BK, Berger W. Impact of terminal dimethylation on the resistance profile of α-N-heterocyclic thiosemicarbazones. Biochem Pharmacol 2012; 83:1623-33. [PMID: 22426010 PMCID: PMC3342514 DOI: 10.1016/j.bcp.2012.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 03/05/2012] [Accepted: 03/06/2012] [Indexed: 12/24/2022]
Abstract
Triapine is an α-N-heterocyclic thiosemicarbazone with promising anticancer activity against hematologic malignancies but widely ineffective against solid tumor types in clinical trials. The anticancer activity of thiosemicarbazones can be dramatically increased by terminal dimethylation. KP1089 is a gallium compound containing two terminal dimethylated thiosemicarbazone ligands. To gain insights on the vulnerability of this highly active terminal dimethylated thiosemicarbazone to drug resistance mechanisms, a new cell model with acquired resistance against the lead compound KP1089 was established. Subsequent genomic analyses (arrayCGH and FISH) revealed amplification of the ABCC1 gene on double minute chromosomal DNA in KP1089-resistant cells as well as overexpression of ABCC1 and ABCG2 on the protein level. KP1089 was further confirmed as a substrate of ABCC1 and ABCG2 but not of ABCB1 using a panel of ABC transporter-overexpressing cell models as well as ABC transporter inhibitors. Moreover, glutathione depletion strongly enhanced KP1089 activity, although no glutathione conjugate formation by glutathione-S-transferase was observed. Thus, a co-transport of KP1089 together with glutathione is suggested. Finally, a panel of thiosemicarbazone derivatives was tested on the new KP1089-resistant cell line. Notably, KP1089-resistant cells were not cross-resistant against thiosemicarbazones lacking terminal dimethylation (e.g. Triapine) which are less active than KP1089. This suggests that terminal dimethylation of thiosemicarbazones – linked with distinctly enhanced anticancer activity – leads to altered resistance profiles compared to classical thiosemicarbazones making this compound class of interest for further (pre)clinical evaluation.
Collapse
Affiliation(s)
- Petra Heffeter
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
The binding and transport of alternative metals by transferrin. Biochim Biophys Acta Gen Subj 2012; 1820:362-78. [DOI: 10.1016/j.bbagen.2011.07.003] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Revised: 06/28/2011] [Accepted: 07/06/2011] [Indexed: 11/24/2022]
|
50
|
Bartel C, Egger AE, Jakupec MA, Heffeter P, Galanski M, Berger W, Keppler BK. Influence of ascorbic acid on the activity of the investigational anticancer drug KP1019. J Biol Inorg Chem 2011; 16:1205-15. [DOI: 10.1007/s00775-011-0809-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 06/11/2011] [Indexed: 11/28/2022]
|