Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
1.
Adv Mater ; 36(14): e2310298, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38145801

RESUMEN

Photodynamic therapy (PDT), as a new type of light-mediated reactive oxygen species (ROS) cancer therapy, has the advantages of high therapeutic efficiency, non-resistance, and less trauma than traditional cancer therapy such as surgery, radiotherapy, and chemotherapy. However, oxygen-dependent PDT further exacerbates tumor metastasis. To this end, a strategy that circumvents tumor metastasis to improve the therapeutic efficacy of PDT is proposed. Herein, a near-infrared light-activated photosensitive polymer is synthesized and branched the anti-metastatic ruthenium complex NAMI-A on the side, which is further assembled to form nanoparticles (NP2) for breast cancer therapy. NP2 can kill tumor cells by generating ROS under 808 nm radiation (NP2 + L), reduce the expression of matrix metalloproteinases (MMP2/9) in cancer cells, decrease the invasive and migration capacity of cancer cells, and eliminate cancer cells. Further animal experiments show that NP2 + L can inhibit tumor growth and reduce liver and lung metastases. In addition, NP2 + L can activate the immune system in mice to avoid tumor recurrence. In conclusion, a PDT capable of both preventing tumor metastasis and precisely hitting the primary tumor to achieve effective treatment of highly metastatic cancers is developed.


Asunto(s)
Dimetilsulfóxido/análogos & derivados , Nanopartículas , Compuestos Organometálicos , Fotoquimioterapia , Compuestos de Rutenio , Animales , Ratones , Especies Reactivas de Oxígeno/metabolismo , Recurrencia Local de Neoplasia/tratamiento farmacológico , Nanopartículas/uso terapéutico , Polímeros , Línea Celular Tumoral , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico
2.
Int J Mol Sci ; 22(17)2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-34502079

RESUMEN

The formation of amyloid-like structures by metabolites is associated with several inborn errors of metabolism (IEMs). These structures display most of the biological, chemical and physical properties of protein amyloids. However, the molecular interactions underlying the assembly remain elusive, and so far, no modulating therapeutic agents are available for clinical use. Chemical chaperones are known to inhibit protein and peptide amyloid formation and stabilize misfolded enzymes. Here, we provide an in-depth characterization of the inhibitory effect of osmolytes and hydrophobic chemical chaperones on metabolite assemblies, thus extending their functional repertoire. We applied a combined in vivo-in vitro-in silico approach and show their ability to inhibit metabolite amyloid-induced toxicity and reduce cellular amyloid content in yeast. We further used various biophysical techniques demonstrating direct inhibition of adenine self-assembly and alteration of fibril morphology by chemical chaperones. Using a scaffold-based approach, we analyzed the physiochemical properties of various dimethyl sulfoxide derivatives and their role in inhibiting metabolite self-assembly. Lastly, we employed whole-atom molecular dynamics simulations to elucidate the role of hydrogen bonds in osmolyte inhibition. Our results imply a dual mode of action of chemical chaperones as IEMs therapeutics, that could be implemented in the rational design of novel lead-like molecules.


Asunto(s)
Amiloide/efectos de los fármacos , Dimetilsulfóxido/farmacología , Adenina/química , Adenina/metabolismo , Amiloide/química , Amiloide/metabolismo , Dimetilsulfóxido/análogos & derivados , Simulación de Dinámica Molecular , Polimerizacion/efectos de los fármacos , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo
3.
Crit Rev Oncog ; 26(2): 73-78, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34347974

RESUMEN

The expansion of metal-based complexes in the last 20 years has been very intense and many metals have been involved. Among the many compounds studied, the ruthenium-based complex NAMI-A embodies the unique paradigm of the ability to selectively inhibiting and preventing the development and the growth of distant metastases originating from solid tumors in all the tumor models on which it has been tested. An activity that can be detected only in vivo since the compound is virtually free of measurable direct cell cytotoxicity in vitro. Recently, a published paper reported on a significant in vitro cytotoxicity against some leukemic cells. The present study was undertaken to reproduce those experiments to further support this novel antileukemic activity that would have put NAMI-A on a new trajectory for development. Our results do not confirm the efficacy of NAMI-A in vitro against the human HL-60 promyelocytic leukemia cell line either using test cultures identical to those reported in the study of reference or in even more stressed conditions, supporting the lack of in vitro direct cell cytotoxicity of NAMI-A. The present study also helps to elucidate that many factors can influence the outcome of in vitro tests of cytotoxicity and suggests caution to speculate on possible therapeutic properties based on the results of simple and reductive in vitro tests of cytotoxicity.


Asunto(s)
Antineoplásicos , Leucemia , Compuestos Organometálicos , Compuestos de Rutenio , Antineoplásicos/farmacología , Línea Celular Tumoral , Dimetilsulfóxido/análogos & derivados , Humanos , Leucemia/tratamiento farmacológico , Compuestos Organometálicos/farmacología , Compuestos de Rutenio/farmacología
4.
J Inorg Biochem ; 223: 111545, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34303108

RESUMEN

Three Ru(II)-DMSO complexes (1-3) containing 2-(3-pyrazolyl)pyridine (PzPy), 2-pyrazol-3-ylfuran (PzO), or 2-pyrazol-3-ylthiophene (PzS) ligand, were synthesized and characterized. The monodentate coordination of the heterocyclic pyrazolyl ligand (PzPy) with Ru(II) ion via N atom was confirmed by single crystal X-ray diffraction. Complex 1 could be converted to the known η2-bidentate PzPy complex cis(Cl), cis(S)-[RuCl2(PzPy)(DMSO)2] (4) under reflux conditions. The mechanism underlying binding mode transformation was studied by 1H NMR spectroscopy and density functional theory (DFT) calculations. The binding abilities of the complexes (1-4) with calf-thymus (CT) DNA and bovine serum albumin (BSA) were investigated using spectroscopic and molecular docking techniques. Among the four Ru(II) complexes, complexes 1 and 3 inhibited the long-term proliferation of human breast cancer cells, whereas complexes 2 and 4 did not inhibit their proliferation to a considerable extent. Interestingly, complexes 1 and 3 did not induce significant cell death but rather attenuated the clonogenicity of breast cancer cells by upregulating reactive oxygen species (ROS), endoplasmic reticulum (ER) and autophagic stress.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Dimetilsulfóxido/análogos & derivados , Dimetilsulfóxido/farmacología , Pirazoles/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Bovinos , Línea Celular Tumoral , Complejos de Coordinación/síntesis química , Complejos de Coordinación/metabolismo , ADN/metabolismo , Dimetilsulfóxido/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Unión Proteica , Pirazoles/síntesis química , Pirazoles/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Rutenio/química , Albúmina Sérica Bovina/metabolismo
5.
J Thromb Thrombolysis ; 51(3): 577-583, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33389608

RESUMEN

In recent years a variety of metals (cadmium, chromium, copper, iron) have been demonstrated to modulate coagulation in vitro and in vivo. One group of metals, the platinoids, have not been assessed, and such investigation is justified given the thromboembolic phenomena associated with platinum-based chemotherapy. Thus, the goal of the present investigation was to assess the effects of carboplatin, cisplatin (platinum compounds), NAMI-A, and ruthenium chloride (ruthenium compounds) on human plasmatic coagulation. Human plasma was exposed to clinically relevant, equimolar concentrations of the aforementioned platinum and ruthenium compounds, with changes in plasmatic coagulation assessed via thrombelastography. The first series of experiments demonstrated no significant modulation of coagulation by the platinum compounds, while NAMI-A demonstrated mild hypercoagulability and ruthenium chloride exerted marked hypercoagulability. A second series of experiments utilizing a variety of specialized modifications of thrombelastography focused on ruthenium chloride revealed that this compound enhances prothrombin activation. While the hypercoagulability associated with platinum compounds in vivo do not appear to have a basis in plasmatic biochemistry, it appears that ruthenium compounds can exert procoagulant properties by enhancing the common pathway of human plasmatic coagulation. Future investigation of Ru based chemotherapeutic agents in development to assess procoagulant activity as part of evaluating their potential clinical safety is warranted.


Asunto(s)
Coagulación Sanguínea , Carboplatino/farmacología , Cisplatino/farmacología , Dimetilsulfóxido/análogos & derivados , Compuestos Organometálicos/farmacología , Protrombina/metabolismo , Compuestos de Rutenio/farmacología , Tromboelastografía/métodos , Antineoplásicos/farmacología , Coagulación Sanguínea/efectos de los fármacos , Coagulación Sanguínea/fisiología , Pruebas de Coagulación Sanguínea/métodos , Dimetilsulfóxido/farmacología , Humanos , Compuestos de Platino/farmacología , Trombofilia/sangre , Trombofilia/inducido químicamente
6.
Mitochondrion ; 56: 25-34, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33220497

RESUMEN

Platinum-based compounds are the most widely used anticancer drugs but, their elevated toxicity and chemoresistance has stimulated the study of others, such as ruthenium-based compounds. NAMI-A and UNICAM-1 were tested in vitro, comparing the mechanisms of toxicity, in terms of mitochondrial functionality and cellular oxidative stress. UNICAM-1, showed a clear mitochondrial target and a cytotoxic dose-dependent response thanks to its ability to promote an imbalance of cellular redox status. It impaired directly mitochondrial respiratory chain, promoting mitochondrial superoxide anion production, leading to mitochondrial membrane depolarization. All these aspects, could make UNICAM-1 a valid alternative for chemotherapy treatment of breast cancer.


Asunto(s)
Cisplatino/farmacología , Dimetilsulfóxido/análogos & derivados , Mitocondrias/efectos de los fármacos , Compuestos Organometálicos/farmacología , Compuestos de Rutenio/farmacología , Superóxidos/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Antineoplásicos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cisplatino/química , Dimetilsulfóxido/química , Dimetilsulfóxido/farmacología , Femenino , Humanos , Mitocondrias/metabolismo , Compuestos Organometálicos/química , Estrés Oxidativo , Compuestos de Rutenio/química , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico
7.
Chem Commun (Camb) ; 56(9): 1397-1400, 2020 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-31912815

RESUMEN

NAMI-A is highly reactive to Sp1, a tumor metastasis related protein, resulting in the perturbation of the protein structure and disruption of the DNA recognition of Sp1. Interestingly, Sp1 is more susceptible than other zinc finger proteins to NAMI-A, suggesting that Sp1 could be the anti-metastasis target of NAMI-A.


Asunto(s)
Antineoplásicos/química , Dimetilsulfóxido/análogos & derivados , Compuestos Organometálicos/química , Compuestos de Rutenio/química , Factor de Transcripción Sp1/química , ADN/metabolismo , Dimetilsulfóxido/química , Glutatión/química , Unión Proteica , Multimerización de Proteína/efectos de los fármacos , Estructura Secundaria de Proteína/efectos de los fármacos , Desplegamiento Proteico/efectos de los fármacos , Factor de Transcripción Sp1/metabolismo
8.
Vox Sang ; 114(3): 283-289, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30734294

RESUMEN

BACKGROUND AND OBJECTIVES: Umbilical cord blood is considered an alternative source of hematopoietic stem cells. Standard banking procedures use 50/55% DMSO in dextran 40 for cryopreservation and dextran-based solutions for thawing, however, due to the potential risk of crystallization of dextran, dextran 40 approved for clinical use has become limited or unavailable. This affects cryopreservation and thawing procedures. Carbohydrates, in particular sucrose, trehalose and glucose, have been shown to be effective in reducing cell damage during dehydration and have cryoprotective potential. We aim to study a 50/55% DMSO in 5% dextrose cryopreservation solution as an alternative to DMSO dextran. MATERIALS AND METHODS: Eighteen samples were divided into two aliquots and cryopreserved, one using standard solution and the other with DMSO dextrose experimental solution. Both aliquots were thawed and diluted with PBS or saline. Total nucleated cells counts, 7-AAD viability of CD45+ cells and recovery of CD34+ viable cells were assessed on thawed samples and compared between pair of aliquots. RESULTS: No differences were observed in the total nucleated cells recovery between cryopreservation solutions, however, higher viability and CD34+ viable cells recoveries were observed using the experimental solution. CONCLUSION: Results showed that DMSO dextrose cryopreservation solution had better results than the standard solution when thawed in an isotonic solution. This indicates that DMSO dextrose is probably a better alternative for direct infusion or when dextran thawing solutions are unavailable. Viability of CD45+ cells and recovery of CD34+ viable cells have positive correlation with engraftment, highlighting the relevance of the optimization of the cryopreservation and thawing process.


Asunto(s)
Conservación de la Sangre/métodos , Criopreservación/métodos , Crioprotectores/efectos adversos , Dimetilsulfóxido/análogos & derivados , Sangre Fetal/efectos de los fármacos , Supervivencia Celular , Crioprotectores/farmacología , Dextranos/efectos adversos , Dextranos/farmacología , Glucosa/efectos adversos , Glucosa/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos
9.
J Phys Chem A ; 122(42): 8397-8411, 2018 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-30114366

RESUMEN

Density functional theory method in combination with a continuum solvation model is used to understand the role of hydrogen bonding in the interactions of tertiary nitrogen centers of guanine and adenine with monoaqua and diaqua NAMI-A. In the case of adenine, the interaction of N3 with monoaqua NAMI-A is preferred over that of N7 and N1 whereas, N7 site is the most preferred site over N3 and N1 in the diaqua ruthenium-adenine interaction. In the monoaqua and diaqua NAMI-A-guanine interactions, the N7 site is the most preferred site over the N3 site. Here, the strength and number of H-bonds play important roles in stabilizing intermediates and transition states involved in the interaction of NAMI-A and purine bases. Atoms in molecules and Becke surface analysis confirm that the interactions between monoaqua and diaqua NAMI-A with the base pairs of GC and AT dinucleotides leads to the structural deformation in the geometry of the base pairs of dinucleotides. The diaqua NAMI-A adducts induce more disruption in the base pairs as compared to monoaqua NAMI-A adducts. which suggests that diaqua NAMI-A could be a better anticancer agent than monoaqua NAMI-A. This study can be extended to envisage the potential applications of computational studies in the development of new drugs and targeted drug delivery systems.


Asunto(s)
ADN/química , Dimetilsulfóxido/análogos & derivados , Nucleótidos/química , Compuestos Organometálicos/química , Teoría Cuántica , Dimetilsulfóxido/química , Enlace de Hidrógeno , Conformación Molecular , Compuestos de Rutenio
10.
Dalton Trans ; 47(33): 11429-11437, 2018 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-30063237

RESUMEN

The reaction of the antimetastatic ruthenium(iii) drug NAMI A with human H-chain ferritin (HuHf) was investigated through a variety of biophysical methods. We observed that the addition of HuHf to NAMI A solutions significantly increases the rate of spontaneous NAMI A hydrolysis suggesting the occurrence of a direct metallodrug-protein interaction. The resulting hydrolyzed Ru species binds the protein mostly forming a relatively tight 1 : 1 ruthenium/ferritin (subunit) adduct that was then separated and characterized. Notably, this adduct shows a characteristic CD spectrum in the visible region, which is diagnostic of the existence of at least one protein bound ruthenium center. The crystal structure of this NAMI A/HuHf adduct was subsequently solved at 1.58 Å resolution; clear evidence is given for the selective binding of a single Ru ion to His105 of each subunit with concomitant release of all other original Ru ligands in agreement with previous observations. We also noted that NAMI A produces a partial inhibition of HuHf ferroxidase activity. The implications of the above results are discussed.


Asunto(s)
Dimetilsulfóxido/análogos & derivados , Ferritinas/metabolismo , Compuestos Organometálicos/metabolismo , Dimetilsulfóxido/metabolismo , Ferritinas/química , Humanos , Enlace de Hidrógeno , Modelos Moleculares , Unión Proteica , Conformación Proteica , Compuestos de Rutenio
11.
Chem Commun (Camb) ; 54(65): 8987-8990, 2018 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-29951655

RESUMEN

IT127 is a dinuclear transition metal complex that contains a Pt(ii) and a Ru(iii) metal center. We have shown that IT127 is significantly more effective in binding the 29-base sarcin ricin loop (SRL) RNA in comparison to Cisplatin, a hallmark anticancer agent. Binding site analysis shows that IT127 prefers purine bases and the GAGA tetraloop region of SRL RNA. Our results with a dihydrofolate reductase (DHFR) model system reveal that IT127 binding to mRNA reduces translation of DHFR enzyme and that the Ru(iii) and Pt(ii) centers in IT127 appear to work in a synergistic manner.


Asunto(s)
Complejos de Coordinación/química , Platino (Metal)/química , ARN Mensajero/química , ARN Ribosómico 28S/química , Rutenio/química , Antineoplásicos/química , Sitios de Unión , Cisplatino/química , Dimetilsulfóxido/análogos & derivados , Dimetilsulfóxido/química , NADP/metabolismo , Conformación de Ácido Nucleico , Compuestos Organometálicos/química , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Compuestos de Rutenio , Tetrahidrofolato Deshidrogenasa/química
12.
J Inorg Biochem ; 182: 177-183, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29501978

RESUMEN

The ruthenium-based anticancer agent NAMI-A (ImH[trans-RuCl4(dmso)(Im)], where Im = imidazole) has been shown to interact with RNA in vivo and in vitro. We hypothesized that the similarly structured drug KP1019 (IndH[trans-RuCl4(Ind)2], where Ind = indazole) binds to RNA as well. Fluorescence spectroscopy was employed to assay the interactions between either NAMI-A or KP1019 and tRNAPhe through an intrinsic fluorophore wybutosine (Y) base and by extrinsic displacement of the intercalating agent ethidium bromide. In both the intrinsic Y-base and extrinsic ethidium bromide studies, KP1019 exhibited tighter binding to phenylalanine-specific tRNA (tRNAPhe) than NAMI-A. In the ethidium bromide study, reducing both drugs from RuIII to RuII resulted in a significant decrease in binding. Our findings suggest that the relatively large heteroaromatic indazole ligands of KP1019 intercalate in the π-stacks of tRNAPhe within structurally complex binding pockets. In addition, NAMI-A appears to be sensitive to destabilizing electrostatic interactions with the negative phosphate backbone of tRNAPhe. Interactions with additional tRNA molecules and other types of RNA require further evaluation to determine the role of RNA in the mechanisms of action for KP1019 and to better understand how Ru drugs fundamentally interact with biomolecules that are more structurally sophisticated than short DNA oligonucleotides. To the best of our knowledge, this is the first study to report KP1019 binding interactions with RNA.


Asunto(s)
Antineoplásicos/química , Dimetilsulfóxido/análogos & derivados , Indazoles/química , Compuestos Organometálicos/química , ARN de Transferencia de Fenilalanina/química , Rutenio/química , Dimetilsulfóxido/química , ARN/química , Compuestos de Rutenio
13.
Met Ions Life Sci ; 182018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29394024

RESUMEN

The general interest in anticancer metal-based drugs and some encouraging pharmacological results obtained at the beginning of the investigations on innovative Ru-based drugs triggered a lot of attention on NAMI-A and KP1019, the two Ru(III) coordination compounds that are the subject of this review. This great attention led to a considerable amount of scientific results and, more importantly, to their eventual admission into clinical trials. Both complexes share a relatively low systemic toxicity that allows reaching rather high dosages, comparable to those of carboplatin. Soon it became evident that NAMI-A and KP1019, in spite of their structural similarity, manifest very distinct chemical and biological properties. The pharmacological performances qualified KP1019 mainly as a cytotoxic drug for the treatment of platinum-resistant colorectal cancers, whereas NAMI-A gained the reputation of a potential anticancer drug with negligible effects on the primary tumor but a pronounced ability to affect metastases. We believe that a strictly comparative exam of NAMI-A and KP1019, based on the substantial body of studies accomplished since their discovery almost 30 years ago, might be an useful exercise, both for assessing the state of the art in terms of biological and clinical profiles, and of the inherent mechanisms, and for envisaging possible future developments in the light of past achievements.


Asunto(s)
Antineoplásicos/farmacología , Dimetilsulfóxido/análogos & derivados , Descubrimiento de Drogas/métodos , Indazoles/farmacología , Neoplasias/tratamiento farmacológico , Compuestos Organometálicos/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/historia , Antineoplásicos/metabolismo , Muerte Celular/efectos de los fármacos , Complejos de Coordinación , Dimetilsulfóxido/química , Dimetilsulfóxido/historia , Dimetilsulfóxido/metabolismo , Dimetilsulfóxido/farmacología , Descubrimiento de Drogas/historia , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Indazoles/química , Indazoles/historia , Indazoles/metabolismo , Modelos Moleculares , Estructura Molecular , Neoplasias/historia , Neoplasias/metabolismo , Neoplasias/patología , Compuestos Organometálicos/química , Compuestos Organometálicos/historia , Compuestos Organometálicos/metabolismo , Unión Proteica , Compuestos de Rutenio , Relación Estructura-Actividad , Distribución Tisular
14.
Drug Deliv ; 25(1): 321-329, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29350051

RESUMEN

Multi-drug delivery systems, which may be promising solution to overcome obstacles, have limited the clinical success of multi-drug combination therapies to treat cancer. To this end, we used three different anticancer agents, Cu(BpT)Br, NAMI-A, and doxorubicin (DOX), to build human serum albumin (HSA)-based multi-drug delivery systems in a breast cancer model to investigate the therapeutic efficacy of overcoming single drug (DOX) resistance to cancer cells in vivo, and to regulate the drugs' release from HSA. The HSA complex structure revealed that NAMI-A and Cu(BpT)Br bind to the IB and IIA sub-domain of HSA by N-donor residue replacing a leaving group and coordinating to their metal centers, respectively. The MALDI-TOF mass spectra demonstrated that one DOX molecule is conjugated with lysine of HSA by a pH-sensitive linker. Furthermore, the release behavior of three agents form HSA can be regulated at different pH levels. Importantly, in vivo results revealed that the HSA-NAMI-A-Cu(BpT)Br-DOX complex not only increases the targeting ability compared with a combination of the three agents (the NAMI-A/Cu(BpT)Br/DOX mixture), but it also overcomes DOX resistance to drug-resistant breast cancer cell lines.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/química , Neoplasias de la Mama/tratamiento farmacológico , Liberación de Fármacos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Albúmina Sérica Humana/química , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Dimetilsulfóxido/administración & dosificación , Dimetilsulfóxido/análogos & derivados , Dimetilsulfóxido/química , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Sistemas de Liberación de Medicamentos/métodos , Quimioterapia Combinada/métodos , Femenino , Humanos , Células MCF-7 , Ratones , Compuestos Organometálicos/administración & dosificación , Compuestos Organometálicos/química , Compuestos de Rutenio
15.
J Inorg Biochem ; 168: 90-97, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28064102

RESUMEN

Solid tumours are constituted of tumour cells, healthy cells recruited from the host tissues and soluble factors released by both these cell types. The present investigation examines the capacity of co-cultures between the HCEC colon epithelial cells and the HCT-116 colorectal cancer cells (mimicking the primary site of tumour growth) and between IHH hepatocytes and the HCT-116 colorectal cancer cells (metastatic site) to influence the effects of NAMI-A (imidazolium trans-imidazoledimethylsulphoxidetetrachloro ruthenate) on the tumour cells themselves. The growth of HCT-116 cells is significantly influenced when the cancer cells are sown on a monolayer of HCEC. The release of soluble factors by the healthy cells promotes, in HCT-116 colorectal cancer cells, the transcription of genes involved in growth, invasion and migration. NAMI-A is not cytotoxic to HCT-116 cells grown on plastics or co-cultured with HCEC or IHH cells, and maintains its ability to control the cell pseudo-metastatic ability, mimicked by the migration in the scratch test. The effects of NAMI-A on HCT-116 migration are supported by its inhibition of the transcription of the ABL-2, ATF-3 and RND-1 genes. In conclusion the study highlights the need of test systems more complex than a single cancer cell culture to study an anticancer drug in vitro and reinforces the hypothesis that NAMI-A targets the ability of the cancer cell to interact with the tumour microenvironment and with the signals that support its metastatic ability.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Dimetilsulfóxido/análogos & derivados , Compuestos Organometálicos/farmacología , Microambiente Tumoral/fisiología , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Dimetilsulfóxido/química , Dimetilsulfóxido/farmacología , Células Epiteliales/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Hepatocitos/efectos de los fármacos , Humanos , Compuestos Organometálicos/química , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Compuestos de Rutenio
16.
Acta Chim Slov ; 64(4): 763-770, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29318318

RESUMEN

Novel tetrachloridoruthenium(III) complex Na[trans-RuCl4(DMSO)(PyrDiaz)] (3) with pyridine-tethered diazenedicarboxamide PyrDiaz ligand (PyrDiaz = N1-(4-isopropylphenyl)-N2-(pyridin-2-ylmethyl)diazene-1,2-dicarboxamide) was synthesized by direct coupling of PyrDiaz with sodium trans-bis(dimethyl sulfoxide)tetrachloridoruthenate(III) (Na-[trans-Ru(DMSO)2Cl4]). Compound 3 is the analogue of the antimetastatic Ru(III) complex NAMI-A and NAMI-Pyr. Single crystal X.


Asunto(s)
Antineoplásicos/síntesis química , Dimetilsulfóxido/análogos & derivados , Compuestos Organometálicos/química , Rutenio/química , Antineoplásicos/química , Cristalografía por Rayos X , Dimetilsulfóxido/química , Imidas/síntesis química , Imidas/química
17.
Biometals ; 29(6): 1035-1046, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27812766

RESUMEN

The antimetastatic ruthenium(III) complex (H2Im)[trans-RuCl4(HIm)(DMSO)] (NAMI-A) as well as its two analogues (H2Ind)[trans-RuCl4(HInd)(DMSO)] (Ru-Ind) and (HIsq)[trans-RuCl4(Isq)(DMSO)] (Ru-Isq) (HIm-imidazole, HInd-indazole, Isq-isoquinoline, DMSO-dimethyl sulfoxide) were tested for their effect on endothelial cell functions in vitro on human skin microvascular endothelial cells (HSkMEC) and human endothelial progenitor cells (HPEC-CB.2) under normoxic (21 % O2) and hypoxic (1 % O2) conditions. All studied complexes showed very low cytotoxicity profiles towards both mature microvascular and precursor endothelial cells (ECs), independently of oxygen concentration. Among tested compounds Ru-Ind exhibited the highest cytotoxicity. The antiangiogenic activity of ruthenium complexes was evaluated for their influence on pseudo-vessels formation by microvascular endothelial cells (HSkMEC) because of their involvement in melanoma progression. Our studies indicated that Ru-Ind and Ru-Isq exhibited hypoxia- and dose-dependent-inhibition of angiogenesis on Matrigel™. Significant hypoxia-selective downregulation of pseudo-vessels formation by Ru-Isq correlates with efficient inhibition of cell motility. Interestingly, in the applied concentration doses migration of endothelial cells was also inhibited by NAMI-A, but the pseudo-vessels formation on Matrigel™ was unaffected. Angiogenesis-related genes expression profile for both mature and precursor ECs indicated that inhibition of angiogenesis, mainly due to Ru-Isq, as compared to NAMI-A and Ru-Ind correlated with downregulation of CD31 and CD144 expression and upregulation of NOTCH4 expression in mature ECs, which is essential for endothelial cell motility and stalk cells organization control. The hypoxia-selective antiangiogenic activity of Ru-Ind and Ru-Isq, NAMI-A analogues makes them potent antimetastatic therapeutics for their selective action in hypoxia which controls tumor pathologic angiogenesis.


Asunto(s)
Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Dimetilsulfóxido/análogos & derivados , Compuestos Organometálicos/química , Rutenio/química , Antineoplásicos/química , Hipoxia de la Célula/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Dimetilsulfóxido/química , Evaluación Preclínica de Medicamentos/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Neovascularización Patológica/genética , Compuestos de Rutenio , Hipoxia Tumoral/efectos de los fármacos
18.
Metallomics ; 8(8): 762-73, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27460862

RESUMEN

NAMI-A and KP1019 are Ru(III)-based anti-metastatic and cytotoxic anti-cancer drugs, respectively, and have been proposed to be activated by reduction to Ru(II). The potential reduction of NAMI-A and KP1019 in the hypoxic environment of a tumour model of neuroblastoma was examined. Normoxic, hypoxic and necrotic tumour tissues were modelled by multicellular spheroids of SH-SY5Y human neuroblastoma cells of various diameters (50-800 µm). The variation in spheroid environment was confirmed with pimonidazole staining. Laser-ablation inductively-coupled plasma mass spectrometry showed KP1019 and NAMI-A penetration into the spheroid hypoxic region. XANES showed that the speciation of NAMI-A biotransformation products did not change significantly as hypoxia levels increased. KP1019 metabolites showed a correlation between the degree of spheroid hypoxia and the Ru K-edge energy consistent with either partial reduction of Ru(III) to Ru(II) in tumour microenvironments, increased S/Cl coordination or a reduced fraction of polynuclear Ru species. EXAFS spectroscopy was undertaken in an attempt to distinguish between these scenarios but was inconclusive.


Asunto(s)
Antineoplásicos/farmacología , Dimetilsulfóxido/análogos & derivados , Hipoxia/fisiopatología , Indazoles/farmacología , Neuroblastoma/patología , Compuestos Organometálicos/farmacología , Rutenio/farmacología , Esferoides Celulares/patología , Antineoplásicos/química , Dimetilsulfóxido/química , Dimetilsulfóxido/farmacología , Humanos , Indazoles/química , Neuroblastoma/tratamiento farmacológico , Compuestos Organometálicos/química , Rutenio/química , Compuestos de Rutenio , Esferoides Celulares/efectos de los fármacos , Células Tumorales Cultivadas , Microambiente Tumoral/efectos de los fármacos , Espectroscopía de Absorción de Rayos X
19.
J BUON ; 21(2): 482-90, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27273962

RESUMEN

PURPOSE: Recently, we reported the synthesis and characterization of two complexes of general formula cis-[Ru(S-DMSO)3(R-CO-CH=CH-R')Cl] (R = 2-hydroxyphenyl for both, R' = thiophene (1), 3-methyl thiophene (2)) that showed remarkable topoisomerase II inhibition and strong binding with DNA. The aim of this study was the investigation of cytotoxic properties of these complexes against a panel of human tumor cell lines, with elucidation of their anticancer mechanisms in HeLa cells. METHODS: Characterization of anticancer activity of the investigated ruthenium complexes 1 and 2 included analysis of cytotoxicity by MTT assay. Cell cycle phase disruption of HeLa cells treated with complexes 1 and 2 was analyzed by flow cytometry after propidium iodide (PI) staining. Annexin V-FITC/PI double staining and further flow cytometry analysis and acridine orange (AO)/ethidium bromide (EB) double staining and fluorescent microscopy were used to determine the apoptotic potential of the investigated ruthenium complexes. The inhibitory effect on gelatinases (MMP-2 and MMP-9) as an indication of possible antimetastatic potential was also analyzed using gelatine zymography. RESULTS: The 50% cell growth inhibition (IC50) values of the investigated complexes ranged between 22.9 and 76.8 µM, with complex 2 being more cytotoxic. Both complexes induced G2 phase cell cycle arrest and apoptosis in HeLa cells. Inhibitory effect of complex 2 on MMP-2 activity was detected. CONCLUSIONS: This work revealed the potential of the investigated Ru(II)-DMSO-chalcone complexes as anticancer agents with cytotoxic and pro-apoptotic activity and indicated complex 2 as leading compound for further chemical modifications and anticancer research.


Asunto(s)
Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Chalconas/farmacología , Dimetilsulfóxido/farmacología , Neoplasias/tratamiento farmacológico , Compuestos de Rutenio/farmacología , Inhibidores de Topoisomerasa II/farmacología , Proliferación Celular/efectos de los fármacos , Chalconas/síntesis química , Dimetilsulfóxido/análogos & derivados , Dimetilsulfóxido/síntesis química , Relación Dosis-Respuesta a Droga , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Células HeLa , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Estructura Molecular , Invasividad Neoplásica , Neoplasias/patología , Compuestos de Rutenio/síntesis química , Relación Estructura-Actividad , Factores de Tiempo , Inhibidores de Topoisomerasa II/síntesis química
20.
Future Med Chem ; 8(5): 527-44, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-27096164

RESUMEN

Research on the field of metal complexes for the treatment of cancer diseases has attracted increasing interest due to the urgency in finding more efficient and selective treatments. Owing to their wide structural diversity, organometallic complexes appear as potential alternatives to the design of new anticancer candidates. Herein, we review recent progress in our work toward the development of new drugs based on Ru(II)- and Fe(II)-cyclopentadienyl scaffolds. Their design and chemical properties are reviewed and correlated with their biological effects, in particular the key role that coligands play in the overall behavior of the complex.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Compuestos Ferrosos/farmacología , Neoplasias/tratamiento farmacológico , Rutenio/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Complejos de Coordinación/química , Complejos de Coordinación/uso terapéutico , Ciclopentanos/química , Dimetilsulfóxido/análogos & derivados , Dimetilsulfóxido/química , Dimetilsulfóxido/farmacología , Dimetilsulfóxido/uso terapéutico , Descubrimiento de Drogas , Compuestos Ferrosos/química , Compuestos Ferrosos/uso terapéutico , Humanos , Indazoles/química , Indazoles/farmacología , Indazoles/uso terapéutico , Terapia Molecular Dirigida , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacología , Compuestos Organometálicos/uso terapéutico , Unión Proteica , Rutenio/química , Rutenio/uso terapéutico , Compuestos de Rutenio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...