Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
1.
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
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.
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
4.
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
5.
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
6.
Inorg Chem ; 55(1): 177-90, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26652771

RESUMEN

A series of novel ferrocene (Fc) functionalized Ru(III) complexes was synthesized and characterized. These compounds are derivatives of the anti-metastatic Ru(III) complex imidazolium [trans-RuCl4(1H-imidazole) (DMSO-S)] (NAMI-A) and are derived from its pyridine analogue (NAMI-Pyr), with direct coupling of Fc to pyridine at the 4 or 3 positions, or at the 4 position via a two-carbon linker, which is either unsaturated (vinyl) or saturated (ethyl). Electron paramagnetic resonance (EPR) and UV-vis spectroscopic studies of the ligand exchange processes of the compounds in phosphate buffered saline (PBS) report similar solution behavior to NAMI-Pyr. However, the complex with Fc substitution at the 3 position of the coordinated pyridine shows greater solution stability, through resistance to the formation of oligomeric species. Further EPR studies of the complexes with human serum albumin (hsA) indicate that the Fc groups enhance noncoordinate interactions with the protein and help to inhibit the formation of protein-coordinated species, suggesting the potential for enhanced bioavailability. Cyclic voltammetry measurements demonstrate that the Fc groups modestly reduce the reduction potential of the Ru(III) center as compared to NAMI-Pyr, while the reduction potentials of the Fc moieties of the four compounds vary by 217 mV, with the longer linkers giving significantly lower values of E1/2. EPR spectra of the compounds with 2-carbon linkers show the formation of a high-spin Fe(III) species (S = 5/2) in PBS with a distinctive signal at g = 4.3, demonstrating oxidation of the Fe(II) ferrocene center and likely reflecting degradation products. Density functional theory calculations and paramagnetic (1)H NMR describe delocalization of spin density onto the ligands and indicate that the vinyl linker could be a potential pathway for electron transfer between the Ru and Fe centers. In the case of the ethyl linker, electron transfer is suggested to occur via an indirect mechanism enabled by the greater flexibility of the ligand. In vitro assays with the SW480 cell line reveal cytotoxicity induced by the ruthenium ferrocenylpyridine complexes that is at least an order of magnitude higher than the unfunctionalized complex, NAMI-Pyr. Furthermore, migration studies with LNCaP cells reveal that Fc functionalization does not reduce the ability of the compounds to inhibit cell motility. Overall, these studies demonstrate that NAMI-A-type compounds can be functionalized with redox-active ligands to produce both cytotoxic and anti-metastatic activity.


Asunto(s)
Dimetilsulfóxido/análogos & derivados , Compuestos Ferrosos/química , Metástasis de la Neoplasia/prevención & control , Compuestos Organometálicos/química , Piridinas/química , Cristalografía por Rayos X , Dimetilsulfóxido/química , Dimetilsulfóxido/farmacología , Espectroscopía de Resonancia por Spin del Electrón , Metalocenos , Estructura Molecular , Compuestos Organometálicos/farmacología , Compuestos de Rutenio
7.
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
8.
Adv Exp Med Biol ; 893: 211-224, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26667346

RESUMEN

Lung cancer is the second most common cancer in both men and women and thus a leading cause of cancer-related deaths worldwide. New efficient treatments especially for its advanced stages and metastases are desperately needed, particularly with regard to overcoming the resistance which thwarts the efficacy of most clinically established drugs such as the platinum complexes. Glimpses of hope are new metal-based drugs that have emerged over the past decade which displayed efficacy in patients with platinum-resistant tumors and metastases. This chapter provides an overview of the latest developments of such metal-based drugs against lung cancer.


Asunto(s)
Neoplasias Pulmonares/tratamiento farmacológico , Cisplatino/uso terapéutico , Dimetilsulfóxido/análogos & derivados , Dimetilsulfóxido/uso terapéutico , Compuestos Ferrosos/uso terapéutico , Humanos , Neoplasias Pulmonares/patología , Metalocenos , Metaloporfirinas/uso terapéutico , Compuestos Organometálicos/uso terapéutico , Compuestos de Rutenio
9.
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
10.
Invest New Drugs ; 33(1): 53-63, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25338748

RESUMEN

AIM OF THE STUDY: The tumor metastases targeting ruthenium complex NAMI-A synergistically improves the activity of gemcitabine in combination therapies. High-throughput screening was used to identify other potential drug combinations from a library of FDA approved drugs. Doxorubicin was identified as a hit compound and was therefore evaluated in combination with NAMI-A in vitro and in a preclinical in vivo model. RESULTS: High-throughput screening identified eight structurally diverse compounds that synergize with NAMI-A including doxorubicin. The combination index on MCF-7 cells showed synergism as the concentration of NAMI-A increases independent of the doxorubicin concentration. In MCa mammary carcinoma of CBA mice, NAMI-A (35 mg/kg/day i.p. on days 7-12) followed by doxorubicin (10 mg/kg i.p. on day 16), significantly increased the effects of the individual drugs on metastases with 70 % animals resulting free of macroscopically detectable tumor nodules in the lungs at sacrifice. NAMI-A, unlike doxorubicin, cured 60 % of the treated mice but the combination therapy was toxic to the animals. CONCLUSIONS: The combined therapy of NAMI-A with doxorubicin synergizes on lung metastasis in a preclinical mouse model. The combination therapy at the maximum tolerated doses of the two drugs is toxic. Hence, this combination is not suitable for clinical studies using maximum tolerated doses.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Mamarias Animales/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Supervivencia Celular/efectos de los fármacos , Dimetilsulfóxido/administración & dosificación , Dimetilsulfóxido/análogos & derivados , Dimetilsulfóxido/farmacocinética , Doxorrubicina/administración & dosificación , Humanos , Riñón/metabolismo , Hígado/metabolismo , Pulmón/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Células MCF-7 , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones Endogámicos CBA , Compuestos Organometálicos/administración & dosificación , Compuestos Organometálicos/farmacocinética , Rutenio/metabolismo , Compuestos de Rutenio
11.
Invest New Drugs ; 33(1): 201-14, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25344453

RESUMEN

BACKGROUND: This phase I/II study determined the maximal tolerable dose, dose limiting toxicities, antitumor activity, the pharmacokinetics and pharmacodynamics of ruthenium compound NAMI-A in combination with gemcitabine in Non-Small Cell Lung Cancer patients after first line treatment. METHODS: Initial dose escalation of NAMI-A was performed in a 28 day cycle: NAMI-A as a 3 h infusion through a port-a-cath at a starting dose of 300 mg/m(2) at day 1, 8 and 15, in combination with gemcitabine 1,000 mg/m(2) at days 2, 9 and 16. Subsequently, dose escalation of NAMI-A in a 21 day schedule was explored. At the maximal tolerable dose level of this schedule an expansion group was enrolled of which 15 patients were evaluable for response. RESULTS: Due to frequent neutropenic dose interruptions in the third week, the 28 day schedule was amended into a 21 day schedule. The maximal tolerable dose was 300 and 450 mg/m(2) of NAMI-A (21 day schedule). Main adverse events consisted of neutropenia, anemia, elevated liver enzymes, transient creatinine elevation, nausea, vomiting, constipation, diarrhea, fatigue, and renal toxicity. CONCLUSION: NAMI-A administered in combination with gemcitabine is only moderately tolerated and less active in NSCLC patients after first line treatment than gemcitabine alone.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Desoxicitidina/administración & dosificación , Desoxicitidina/efectos adversos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacocinética , Dimetilsulfóxido/administración & dosificación , Dimetilsulfóxido/efectos adversos , Dimetilsulfóxido/análogos & derivados , Dimetilsulfóxido/farmacocinética , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Compuestos Organometálicos/administración & dosificación , Compuestos Organometálicos/efectos adversos , Compuestos Organometálicos/farmacocinética , Rutenio/administración & dosificación , Rutenio/efectos adversos , Rutenio/sangre , Rutenio/farmacocinética , Compuestos de Rutenio , Resultado del Tratamiento , Gemcitabina
12.
J Biol Inorg Chem ; 20(4): 695-703, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25792379

RESUMEN

Imidazolium trans-tetrachloridodimethylsulfoxideimidazolruthenate(III), NAMI-A, a novel antimetastatic ruthenium complex was investigated towards affinity to transferrin (Tf), whether Tf-Ru adducts might be formed after its intravenous injection. Studies were focused on the holotransferrin due to its preferential binding to transferrin receptor. Here, we showed that holotransferrin is able to bind NAMI-A as readily as apotransferrin. The simulation of biological conditions of human serum performed by application of simplified serum models allowed to analyse ruthenium distribution between transferrin and albumin. The presence of physiological concentration of albumin (ca. 18-fold excess over Tf) resulted in a twofold decrease of ruthenium binding to Tf. Interestingly, the introducing of low-molecular-mass components of serum dramatically increased the ruthenation of Tf. Intermolecular competition binding studies between transferrin and albumin showed that both proteins bound similar amount of ruthenium species. Investigation of NAMI-A binding to Tf in human serum showed that this protein was not the major binding partner for Ru complex. However, in spite of many competing proteins still the ruthenation of Tf was observed. The lack of free Ru species (protein unbounded) after incubation with human serum allowed to make an assumption of high affinity of NAMI-A towards serum proteins.


Asunto(s)
Dimetilsulfóxido/análogos & derivados , Compuestos Organometálicos/química , Transferrina/química , Sitios de Unión , Dimetilsulfóxido/sangre , Dimetilsulfóxido/química , Humanos , Modelos Moleculares , Conformación Molecular , Peso Molecular , Compuestos Organometálicos/sangre , Compuestos de Rutenio
13.
J Biol Inorg Chem ; 20(7): 1163-73, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26369538

RESUMEN

The ruthenium-based drug NAMI-A, characterised by its selectivity against solid tumour metastases, promotes TGF-ß1-dependent fibrosis and the reduction of the release of MMPs in the primary tumour. The aim of the study was to examine the interaction of NAMI-A with TGF-ß1 in the process of metastasis formation. NAMI-A (1) affects the secretion of TGF-ß1 in metastatic MDA-MB-231 cells rather than in non-tumorigenic HBL-100 cells, (2) prevails over TGF-ß1 with regard to the invasive capacity of the treated cells, and (3) contrasts integrin-dependent migration stimulated by TGF-ß1. It, thus, appears that the effects of NAMI-A on cell invasion and migration are best summarised as an interference with TGF-ß1 and a reduction of its activity in these events. At a molecular level, the similar activity of NAMI-A and TGF-ß1 on RhoA GTPase supports its interaction with cell surface integrins while TGF-ß1 can activate it by interaction with its TGFßR receptor. The inhibition of TGF-ß1-induced migration of MDA-MB-231 cells by NAMI-A cannot simply be attributed to a modulation of the Smad2 and p38MAPK pathways. In conclusion, the effects of NAMI-A on the biological role of TGF-ß1 in cancer metastasis are insufficient to attribute the responsibility for the anti-metastatic activity of the ruthenium-based drug to this target alone.


Asunto(s)
Dimetilsulfóxido/análogos & derivados , Compuestos Organometálicos/farmacología , Rutenio/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacología , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dimetilsulfóxido/química , Dimetilsulfóxido/farmacocinética , Dimetilsulfóxido/farmacología , Humanos , Estructura Molecular , Metástasis de la Neoplasia/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Neoplasias/fisiopatología , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacocinética , Rutenio/química , Compuestos de Rutenio
14.
J Biol Inorg Chem ; 20(5): 831-40, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25982099

RESUMEN

The ruthenium-based drug imidazolium trans-imidazoledimethylsulphoxidetetrachlorido ruthenate (NAMI-A) is a novel antitumour drug under clinical evaluation. In this study, NAMI-A is tested on aortic rings in vitro and on the systolic blood pressure in vivo with the aim of evaluating its effects on smooth muscle cells and, more in general, on the vascular system. Pre-incubation of aortic rings with 10 µM NAMI-A for 10 min potentiates the contraction induced by phenylephrine (PE). The reduction of the B max value of [(3)H]-prazosin bound to NAMI-A-treated aortic rings and the ability of NAMI-A to displace [(3)H]-prazosin and [(3)H]-IP3 binding by 25 and 42%, respectively, suggest the involvement of α1-adrenoceptor in mediating the effects on smooth muscle cells. NAMI-A also decreases the number of maximal sites of [(3)H]-prazosin bound to kidney membrane preparation from 34 to 24 fmol/mg proteins. A single i.p. dose (105 mg/kg) or a repeated treatment for 6 consecutive days (17 mg/kg/day) in Wistar rats increases the systolic blood pressure, respectively, 1 h and 3 days after treatment, and the responsiveness of rat aortic rings to PE. Atomic absorption spectroscopy confirms the presence of ruthenium in the aortic rings excised from the treated rats. These findings suggest monitoring the cardiovascular parameters when the drug is used in humans for treating cancer patients, particularly if the drug is associated with chemicals that are potentially active at the cardiovascular level.


Asunto(s)
Antineoplásicos/farmacología , Aorta/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Dimetilsulfóxido/análogos & derivados , Contracción Muscular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Compuestos Organometálicos/farmacología , Fenilefrina/farmacología , Animales , Antineoplásicos/química , Aorta/citología , Dimetilsulfóxido/química , Dimetilsulfóxido/farmacología , Relación Dosis-Respuesta a Droga , Masculino , Miocitos del Músculo Liso/citología , Compuestos Organometálicos/química , Fenilefrina/química , Ratas , Ratas Wistar , Compuestos de Rutenio , Relación Estructura-Actividad
15.
J Biol Inorg Chem ; 19(6): 1049-53, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24458238

RESUMEN

Single drug-based cancer therapies are frequently associated with the development of drug resistance. To overcome this problem, combination therapy with two or more anticancer drugs is a promising strategy, but clinical studies are logistically challenging and costly. Intermediary in vitro studies, however, can provide critical insight to decide whether one should proceed to in vivo studies. To this end, cisplatin and the Ru-based anticancer drug NAMI-A were added to human plasma and the size distribution of Pt-containing and Ru-containing entities was determined over a 2 h period. The results revealed a dramatically different rate of plasma protein binding for each drug and/or their hydrolysis products. Both drugs bound to the same apparent plasma proteins, but crucially they did not adversely affect each other's metabolism. Therefore, combination therapy of patients with these metallodrugs should be further assessed in clinical studies in order to systematically develop an effective combination therapy protocol to prevent the resurgence of cancer.


Asunto(s)
Cisplatino/sangre , Cisplatino/metabolismo , Dimetilsulfóxido/análogos & derivados , Compuestos Organometálicos/sangre , Compuestos Organometálicos/metabolismo , Cisplatino/química , Dimetilsulfóxido/sangre , Dimetilsulfóxido/química , Dimetilsulfóxido/metabolismo , Humanos , Compuestos Organometálicos/química , Compuestos de Rutenio , Espectrofotometría Atómica
16.
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
18.
Inorg Chem ; 51(2): 954-66, 2012 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-22224431

RESUMEN

A series of pyridine-based derivatives of the antimetastatic Ru(III) complex imidazolium [trans-RuCl(4)(1H-imidazole)(DMSO-S)] (NAMI-A) have been synthesized along with their sodium-ion compensated analogues. These compounds have been characterized by X-ray crystallography, electron paramagnetic resonance (EPR), NMR, and electrochemistry, with the goal of probing their noncovalent interactions with human serum albumin (hsA). EPR studies show that the choice of imidazolium ligands and compensating ions does not strongly influence the rates of ligand exchange processes in aqueous buffer solutions. By contrast, the rate of formation and persistence of interactions of the complexes with hsA is found to be strongly dependent on the properties of the axial ligands. The stability of noncovalent binding is shown to correlate with the anticipated ability of the various pyridine ligands to interact with the hydrophobic binding domains of hsA. These interactions prevent the oligomerization of the complexes in solution and limit the rate of covalent binding to albumin amino acid side chains. Electrochemical studies demonstrate relatively high reduction potentials for these complexes, leading to the formation of Ru(II) species in aqueous solutions containing biological reducing agents, such as ascorbate. However, EPR measurements indicate that while noncovalent interactions with hsA do not prevent reduction, covalent binding produces persistent mononuclear Ru(III) species under these conditions.


Asunto(s)
Antineoplásicos/química , Dimetilsulfóxido/análogos & derivados , Compuestos Organometálicos/química , Piridinas/química , Albúmina Sérica/química , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Sitios de Unión , Cristalografía por Rayos X , Dimetilsulfóxido/química , Electroquímica , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Espectroscopía de Resonancia Magnética , Estructura Molecular , Oxidación-Reducción , Compuestos de Rutenio , Albúmina Sérica/metabolismo , Soluciones/química , Relación Estructura-Actividad
19.
J Biol Inorg Chem ; 16(8): 1177-85, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21739255

RESUMEN

[ImH][trans-Ru(III)Cl(4)(DMSO)(Im)] (where DMSO is dimethyl sulfoxide and Im is imidazole) (NAMI-A) is an antimetastatic prodrug currently in phase II clinical trials. The mechanisms of action of this and related Ru-based anticancer agents are not well understood, but several cellular targets have been suggested. Although Ru has been observed to bind to DNA following in vitro NAMI-A exposure, little is known about Ru-DNA interactions in vivo and even less is known about how this or related metallodrugs might influence cellular RNA. In this study, Ru accumulation in cellular RNA was measured following treatment of Saccharomyces cerevisiae with NAMI-A. Drug-dependent growth and cell viability indicate relatively high tolerance, with approximately 40% cell death occurring at 6 h for 450 µM NAMI-A. Significant dose-dependent accumulation of Ru in cellular RNA was observed by inductively coupled plasma mass spectrometry measurements on RNA extracted from yeast treated with NAMI-A. In vitro, binding of Ru species to drug-treated model DNA and RNA oligonucleotides at pH 6.0 and 7.4 was characterized by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry in the presence and absence of the reductant ascorbate. The extent of Ru-nucleotide interactions increases slightly with lower pH and significantly in the presence of ascorbate, with differences in observed species distribution. Taken together, these studies demonstrate the accumulation of aquated and reduced derivatives of NAMI-A on RNA in vitro and in cellulo, and enhanced binding with nucleic acid targets in a tumorlike acidic, reducing environment. To our knowledge, this is also the first study to characterize NAMI-A treatment of S. cerevisiae, a genetically tractable model organism.


Asunto(s)
ADN/química , Dimetilsulfóxido/análogos & derivados , Compuestos Organometálicos/química , ARN/química , Rutenio/química , Rutenio/metabolismo , Saccharomyces cerevisiae/metabolismo , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Ácido Ascórbico/química , Supervivencia Celular/efectos de los fármacos , Dimetilsulfóxido/síntesis química , Dimetilsulfóxido/química , Dimetilsulfóxido/farmacología , Concentración de Iones de Hidrógeno , Hidrólisis , Estructura Molecular , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/farmacología , Profármacos/síntesis química , Profármacos/química , Profármacos/farmacología , Rutenio/farmacología , Compuestos de Rutenio
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA