Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
RSC Chem Biol ; 2(4): 1263-1273, 2021 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-34458840

RESUMEN

Fungal infections represent a global problem, notably for immunocompromised patients in hospital, COVID-19 patient wards and care home settings, and the ever-increasing emergence of multidrug resistant fungal strains is a sword of Damocles hanging over many healthcare systems. Azoles represent the mainstay of antifungal drugs, and their mode of action involves the binding mode of these molecules to the fungal lanosterol 14α-demethylase target enzyme. In this study, we have prepared and characterized four novel organometallic derivatives of the frontline antifungal drug fluconazole (1a-4a). Very importantly, enzyme inhibition and chemogenomic profiling demonstrated that lanosterol 14α-demethylase, as for fluconazole, was the main target of the most active compound of the series, (N-(ferrocenylmethyl)-2-(2,4-difluorophenyl)-2-hydroxy-N-methyl-3-(1H-1,2,4-triazol-1-yl)propan-1-aminium chloride, 2a). Transmission electron microscopy (TEM) studies suggested that 2a induced a loss in cell wall integrity as well as intracellular features ascribable to late apoptosis or necrosis. The impressive activity of 2a was further confirmed on clinical isolates, where antimycotic potency up to 400 times higher than fluconazole was observed. Also, 2a showed activity towards azole-resistant strains. This finding is very interesting since the primary target of 2a is the same as that of fluconazole, emphasizing the role played by the organometallic moiety. In vivo experiments in a mice model of Candida infections revealed that 2a reduced the fungal growth and dissemination but also ameliorated immunopathology, a finding suggesting that 2a is active in vivo with added activity on the host innate immune response.

2.
Chem Commun (Camb) ; 56(92): 14373-14376, 2020 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-33140750

RESUMEN

Novel tetraplatinated metalloporphyrin-based photosensitizers (PSs) are reported, which show excellent phototoxic indexes (PIs) up to 5800 against HeLa cells, which is, to the best of our knowledge, the highest value reported for any porphyrin so far. Furthermore, 67Zn isotope labelling allowed the determination of the ratio of zinc to platinum inside the cells using ICP-MS.


Asunto(s)
Metaloporfirinas/química , Fármacos Fotosensibilizantes/química , Platino (Metal)/química , Isótopos de Zinc/química , Cobre/química , Células HeLa , Humanos , Marcaje Isotópico , Ligandos , Espectrometría de Masas , Metaloporfirinas/efectos adversos , Imagen Óptica , Fotoquimioterapia , Fármacos Fotosensibilizantes/efectos adversos , Platino (Metal)/efectos adversos , Relación Estructura-Actividad , Distribución Tisular
3.
Chembiochem ; 21(20): 2966-2973, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32473056

RESUMEN

In the fight against cancer, photodynamic therapy is generating great interest thanks to its ability to selectively kill cancer cells without harming healthy tissues. In this field, ruthenium(II) polypyridyl complexes, and more specifically, complexes with dipyrido[3,2-a:2',3'-c]phenazine (dppz) as a ligand are of particular interest due to their DNA-binding and photocleaving properties. However, ruthenium(II) polypyridyl complexes can sometimes suffer from low lipophilicity, which hampers cellular internalisation through passive diffusion. In this study, four new [Ru(dppz-X2 )3 ]2+ complexes (X=H, F, Cl, Br, I) were synthesized and their lipophilicity (logP), cytotoxicity and phototoxicity on cancerous and noncancerous cell lines were assessed. This study shows that, counterintuitively, the phototoxicity of these complexes decreases as their lipophilicity increases; this could be due solely to the atomic radius of the halogen substituents.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Hidrocarburos Halogenados/farmacología , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Halogenación , Humanos , Hidrocarburos Halogenados/síntesis química , Hidrocarburos Halogenados/química , Interacciones Hidrofóbicas e Hidrofílicas , Ligandos , Estructura Molecular , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/química , Piridinas/química , Piridinas/farmacología , Rutenio/química , Rutenio/farmacología , Oxígeno Singlete/metabolismo
4.
J Med Chem ; 63(10): 5568-5584, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32319768

RESUMEN

Chemotherapy remains one of the dominant treatments to cure cancer. However, due to the many inherent drawbacks, there is a search for new chemotherapeutic drugs. Many classes of compounds have been investigated over the years to discover new targets and synergistic mechanisms of action including multicellular targets. In this work, we designed a new chemotherapeutic drug candidate against cancer, namely, [Ru(DIP)2(sq)](PF6) (Ru-sq) (DIP = 4,7-diphenyl-1,10-phenanthroline; sq = semiquinonate ligand). The aim was to combine the great potential expressed by Ru(II) polypyridyl complexes and the singular redox and biological properties associated with the catecholate moiety. Experimental evidence (e.g., X-ray crystallography, electron paramagnetic resonance, electrochemistry) demonstrates that the semiquinonate is the preferred oxidation state of the dioxo ligand in this complex. The biological activity of Ru-sq was then scrutinized in vitro and in vivo, and the results highlight the promising potential of this complex as a chemotherapeutic agent against cancer.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/metabolismo , Quinonas/química , Quinonas/metabolismo , Rutenio/química , Rutenio/metabolismo , Animales , Antineoplásicos/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Femenino , Células HeLa , Humanos , Ligandos , Ratones , Ratones Desnudos , Oxidación-Reducción/efectos de los fármacos , Quinonas/farmacología , Rutenio/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
5.
Chemistry ; 26(22): 4997-5009, 2020 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-32065454

RESUMEN

Cancer is one of the main causes of death worldwide. Chemotherapy, despite its severe side effects, is to date one of the leading strategies against cancer. Metal-based drugs present several potential advantages when compared to organic compounds and they have gained trust from the scientific community after the approval on the market of the drug cisplatin. Recently, we reported the ruthenium complex ([Ru(DIP)2 (sq)](PF6 ) (where DIP is 4,7-diphenyl-1,10-phenantroline and sq is semiquinonate) with a remarkable potential as chemotherapeutic agent against cancer, both in vitro and in vivo. In this work, we analyse a structurally similar compound, namely [Ru(DIP)2 (mal)](PF6 ), carrying the flavour-enhancing agent approved by the FDA, maltol (mal). To possess an FDA approved ligand is crucial for a complex, whose mechanism of action might include ligand exchange. Herein, we describe the synthesis and characterisation of [Ru(DIP)2 (mal)](PF6 ), its stability in solutions and under conditions that resemble the physiological ones, and its in-depth biological investigation. Cytotoxicity tests on different cell lines in 2D model and on HeLa MultiCellular Tumour Spheroids (MCTS) demonstrated that our compound has higher activity than cisplatin, inspiring further tests. [Ru(DIP)2 (mal)](PF6 ) was efficiently internalised by HeLa cells through a passive transport mechanism and severely affected the mitochondrial metabolism.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Complejos de Coordinación/farmacología , Pironas/farmacología , Rutenio/química , Antineoplásicos/química , Cisplatino/química , Complejos de Coordinación/química , Células HeLa , Humanos , Ligandos , Estructura Molecular , Pironas/química , Rutenio/farmacología
6.
Metallomics ; 11(2): 462-474, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30620038

RESUMEN

A library of 18 dinuclear-thiolato bridged arene ruthenium complexes, some of which with demonstrated activity against cancer cells, was screened for activity against a transgenic Neospora caninum strain that constitutively expresses beta-galactosidase. Initial assessments were done at concentrations of 2500, 250, 25 and 2.5 nM, and 5 compounds were further evaluated with regard to their half maximal proliferation-inhibiting concentration (IC50). Among those, [(η6-p-MeC6H4Pri)2Ru2(µ2-SC6H4-p-CH3)3]Cl (1), [(η6-p-MeC6H4Pri)2Ru2(µ2-SC6H4-p-But)3]Cl (2) and [(η6-p-MeC6H4Pri)2Ru2(µ2-SCH2C6H4-p-But)2(µ2-SC6H4-p-OH)]BF4 (9) inhibited N. caninum proliferation with low C50 values of 15, 5 and 1 nM, respectively, while [(η6-p-MeC6H4Pri)2Ru2(µ2-SC6H4-p-OH)3]Cl (3) and [(η6-p-MeC6H4Pri)2Ru2(µ2-SC6H4-p-mco)3]Cl (5, mco = 4-methylcoumarinyl) were less active (IC50 = 280 and 108 nM, respectively). These compounds did not affect human foreskin fibroblast (HFF) host cells at dosages of 5 µM and above, but impaired proliferation of the human ovarian carcinoma cell line A2780 (IC50 values of 130 nM (1), 30 nM (2), 530 nM (3), 7730 nM (5), 130 nM (9)). A2780 cancer cells were treated with complexes 1, 2, and 5, and biodistribution analysis using inductively coupled plasma mass spectrometry (ICP-MS) showed that most of the drugs accumulated in the mitochondrial fractions. Transmission electron microscopy showed that the parasite mitochondrion is the primary target also in N. caninum tachyzoites, but these compounds, when applied at 200 nM for 15 days in vitro, did not act parasiticidal. Complexes 1, 2 and 9 applied orally at 2 and 10 mg kg-1 day-1 during 5 days in a neosporosis mouse model did not reduce parasite load and did not limit parasite dissemination to the central nervous system. In accordance with these results, ICP-MS carried out on different organs of mice orally administrated with complexes 1 and 9, demonstrated that the drugs were readily absorbed, and after 3 and 48 h, were mainly detected in liver and kidney, but were largely absent from the brain. Thus, dinuclear thiolato-bridged arene ruthenium complexes exhibit interesting activities against N. caninum in vitro, but further modifications of these promising molecules are required to improve their bioavailability and pharmacokinetic properties in order to exert a pronounced and selective effect against N. caninum in vivo.


Asunto(s)
Antiparasitarios/química , Antiparasitarios/farmacología , Mitocondrias/metabolismo , Rutenio/química , Rutenio/farmacología , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Coccidios/efectos de los fármacos , Coccidios/patogenicidad , Humanos , Ratones , Microscopía Electrónica de Transmisión , Neospora/metabolismo , Células Vero , beta-Galactosidasa/metabolismo
7.
Artículo en Inglés | MEDLINE | ID: mdl-28652238

RESUMEN

The in vitro effects of 18 dinuclear thiolato-bridged arene ruthenium complexes (1 monohiolato compound, 4 dithiolato compounds, and 13 trithiolato compounds), originally designed as anticancer agents, on the apicomplexan parasite Toxoplasma gondii grown in human foreskin fibroblast (HFF) host cells were studied. Some trithiolato compounds exhibited antiparasitic efficacy at concentrations of 250 nM and below. Among those, complex 1 and complex 2 inhibited T. gondii proliferation with 50% inhibitory concentrations (IC50s) of 34 and 62 nM, respectively, and they did not affect HFFs at dosages of 200 µM or above, resulting in selectivity indices of >23,000. The IC50s of complex 9 were 1.2 nM for T. gondii and above 5 µM for HFFs. Transmission electron microscopy detected ultrastructural alterations in the matrix of the parasite mitochondria at the early stages of treatment, followed by a more pronounced destruction of tachyzoites. However, none of the three compounds applied at 250 nM for 15 days was parasiticidal. By affinity chromatography using complex 9 coupled to epoxy-activated Sepharose followed by mass spectrometry, T. gondii translation elongation factor 1α and two ribosomal proteins, RPS18 and RPL27, were identified to be potential binding proteins. In conclusion, organometallic ruthenium complexes exhibit promising activities against Toxoplasma, and the potential mechanisms of action of these compounds as well as their prospective applications for the treatment of toxoplasmosis are discussed.


Asunto(s)
Antiparasitarios/farmacología , Compuestos Organometálicos/farmacología , Compuestos de Rutenio/farmacología , Toxoplasma/efectos de los fármacos , Toxoplasmosis/tratamiento farmacológico , Animales , Antiparasitarios/química , Línea Celular , Chlorocebus aethiops , Humanos , Microscopía Electrónica de Transmisión , Mitocondrias/efectos de los fármacos , Compuestos Organometálicos/química , Pruebas de Sensibilidad Parasitaria , Factor 1 de Elongación Peptídica/metabolismo , Proteínas Ribosómicas/metabolismo , Toxoplasma/crecimiento & desarrollo , Células Vero
8.
Dalton Trans ; 46(42): 14523-14536, 2017 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-28164201

RESUMEN

In this article, we report on the development of new metal-based anticancer agents with imaging, chemotherapeutic and photosensitizing properties. Hence, a new heterobimetallic complex (Pt-LQ-Re) was prepared by connecting a non-conventional trans-chlorido Pt(ii) complex to a photoactive Re tricarbonyl unit (LQ-Re), which can be replaced by 99mTc to allow for in vivo imaging. We describe the photophysical and biological properties of the new complexes, in the dark and upon light irradiation (DNA interaction, cellular localization and uptake, and cytotoxicity). Furthermore, planar scintigraphic images of mice injected with Pt-LQ-Tc clearly showed that the radioactive compound is taken up by the excretory system organs, namely liver and kidneys, without significant retention in other tissues. All in all, the strategy of conjugating a chemotherapeutic compound with a PDT photosensitizer endows the resulting complexes with an intrinsic cytotoxic activity in the dark, driven by the non-classical platinum core, and a selective activity upon light irradiation. Most importantly, the possibility of integrating a SPECT imaging radiometal (99mTc) in the structure of these new heterobimetallic complexes might allow for in vivo non-invasive visualization of their tumoral accumulation, a crucial issue to predict therapeutic outcomes.


Asunto(s)
Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Platino (Metal)/química , Cintigrafía/métodos , Renio/química , Tecnecio/química , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Transporte Biológico , Línea Celular Tumoral , Complejos de Coordinación/metabolismo , Complejos de Coordinación/farmacocinética , Humanos , Ratones , Oxígeno Singlete/metabolismo , Distribución Tisular
9.
Chem Sci ; 7(9): 6115-6124, 2016 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-27708751

RESUMEN

Photodynamic therapy (PDT) is an attractive, complementary medical technique to chemotherapy. Among the different photosensitizers (PSs) employed, Ru(ii) polypyridyl complexes were found to be valid substitutes to porphyrin-based or phthalocyanine-based PSs. Here, we confirm that one such complex, namely [Ru(bipy)2-dppz-7-methoxy][PF6]2 (Ru65), which localizes in the nucleus of various cancer and normal cells, displays cytotoxicity only upon UV-A irradiation. Importantly, we disclose the molecular mechanism of the UV-A mediated cytotoxic action of Ru65. We demonstrate that Ru65 intercalates in DNA and, upon light irradiation, promotes guanine oxidation, resulting in nicks in the double helix. We confirm this mechanism of action in living cells, showing that the UV-A irradiation of cells loaded with Ru65 results in a transient DNA damage response and cell death. Strikingly, the photo-irradiation of Ru65 triggered distinct mechanisms of cell death in interphase or mitotic cells. The former underwent cell cycle arrest at the G2/M phase and massive cytoplasmic vacuolation, which was paralleled by an unfolded-protein stress response, resulting in a reduction of viability and cell death through a paraptosis-like mechanism. On the other hand, the UV-A irradiation of Ru65 in cells synchronized by G2/M block-release with a selective CDK1 inhibitor led to blocking mitotic entry and rapid cell death through classic apoptotic pathways. Importantly, targeting mitotic cells with Ru65 allowed increasing its photo-toxicity by a factor of 3.6. Overall, our findings show that the use of a combination of a cell cycle inhibitor and a PS targeting the nucleus could open up new avenues in PDT.

10.
Bioconjug Chem ; 27(8): 1942-8, 2016 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-27459208

RESUMEN

The current interest for platinum N-heterocyclic carbene complexes in cancer research stems from their impressive toxicity reported against a range of different human cancer cells. To date, the demonstration of their in vivo efficacy relative to that of established platinum-based drugs has not been specifically addressed. Here, we introduce an innovative approach to increase the NHC-Pt complex potency whereby multiple NHC-Pt(II) complexes are coordinated along a polyethylenimine polymer (PEI) chain. We show that such NHC-Pt(II)-PEI conjugates induce human cancer cell death in vitro and in vivo in a xenograft mouse model with no observable side effects in contrast to oxaliplatin. Additional studies indicate nucleus and mitochondria targeting and suggest various mechanisms of action compared to classical platinum-based anticancer drugs.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Compuestos Organoplatinos/química , Compuestos Organoplatinos/farmacología , Polietileneimina/química , Animales , Antineoplásicos/metabolismo , Transporte Biológico , Línea Celular Tumoral , Humanos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Compuestos Organoplatinos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Chembiochem ; 17(11): 1004-7, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-26991635

RESUMEN

An organometallic derivative of praziquantel was studied directly in worms by using inductively coupled plasma-mass spectrometry (ICP-MS) for quantification and synchrotron-based imaging. X-ray fluorescence (XRF) and IR absorption spectromicroscopy were used for the first time in combination to directly locate this organometallic drug candidate in schistosomes. The detection of both CO (IR) and Cr (XRF) signatures proved that the Cr(CO)3 core remained intact in the worms. Images showed a preferential accumulation at the worm's tegument, consistent with a possible targeting of the calcium channel but not excluding other biological targets inside the worm.


Asunto(s)
Praziquantel/química , Schistosoma mansoni/química , Animales , Cromo/química , Espectrometría de Masas , Microscopía , Imagen Óptica , Praziquantel/síntesis química , Praziquantel/farmacología , Schistosoma mansoni/efectos de los fármacos , Schistosoma mansoni/metabolismo , Espectrofotometría Infrarroja , Estereoisomerismo , Espectroscopía de Absorción de Rayos X
12.
Molecules ; 21(2)2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26861271

RESUMEN

A new bifunctional macrocyclic chelator featuring a conjugatable alkynyl-naphthalimide fluorophore pendant group has been prepared and its Gd(III) complex coupled to a cell-penetrating lipidated azido-Tat peptide derivative using Cu(I)-catalysed "click" chemistry. The resulting fluorescent conjugate is able to enter CAL-33 tongue squamous carcinoma cells, as revealed by confocal microscopy, producing a very modest anti-proliferative effect (IC50 = 93 µM). Due to the photo-reactivity of the naphthalimide moiety, however, the conjugate's cytotoxicity is significantly enhanced (IC50 = 16 µM) upon brief low-power UV-A irradiation.


Asunto(s)
Antineoplásicos/metabolismo , Complejos de Coordinación/metabolismo , Naftalimidas/metabolismo , Fármacos Fotosensibilizantes/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quelantes/metabolismo , Quelantes/farmacología , Química Clic , Complejos de Coordinación/farmacología , Gadolinio/química , Compuestos Heterocíclicos con 1 Anillo/química , Humanos , Naftalimidas/farmacología , Fármacos Fotosensibilizantes/farmacología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química
13.
Inorg Chem ; 55(4): 1674-82, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26821062

RESUMEN

Two new bifunctional macrocyclic chelate ligands that form luminescent terbium(III) complexes featuring an alkyne group for conjugation to (bio)molecules via the Cu(I)-catalyzed "click" reaction were synthesized. Upon ligation, the complexes exhibit a significant luminescent enhancement when excited at the λ(max) of the "clicked" products. To demonstrate the utility of the complexes for luminescent labeling, they were conjugated in vitro to E. coli aspartate/glutamate-binding protein incorporating a genetically encoded p-azido-L-phenylalanine or p-(azidomethyl)-L-phenylalanine residue. The complexes may prove useful for time-gated assay applications.


Asunto(s)
Alquinos/química , Proteínas/química , Terbio/química , Luminiscencia
14.
Angew Chem Int Ed Engl ; 55(8): 2792-5, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26799241

RESUMEN

Doxorubicin, a well-established chemotherapeutic agent, is known to accumulate in the cell nucleus. By using ICP-MS, we show that the conjugation of two small organometallic rhenium complexes to this structural motif results in a significant redirection of the conjugates from the nucleus to the mitochondria. Despite this relocation, the two bioconjugates display excellent toxicity toward HeLa cells. In addition, we carried out a preliminarily investigation of aspects of cytotoxicity and present evidence that the conjugates disrupt the mitochondrial membrane potential, are strong inhibitors of human Topoisomerase II, and induce apoptosis. Such derivatives may enhance the therapeutic index of the aggressive parent drug and overcome drug resistance by influencing nuclear and mitochondrial homeostasis.


Asunto(s)
Antibióticos Antineoplásicos/farmacocinética , Doxorrubicina/farmacocinética , Mitocondrias/efectos de los fármacos , Compuestos Organometálicos/farmacología , Renio/farmacología , Células HeLa , Humanos , Microscopía Fluorescente , Renio/química
15.
ChemMedChem ; 10(11): 1901-14, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26332425

RESUMEN

In the search for alternative photosensitizers for use in photodynamic therapy (PDT), herein we describe two new water-soluble porphyrins, a neutral fourfold-symmetric compound and a +3-charged tris-methylpyridinium derivative, in which either four or one [1,4,7]-triazacyclononane (TACN) units are connected to the porphyrin macrocycle through a hydrophilic linker; we also report their corresponding tetracationic Re(I) conjugates. The in vitro (photo)toxic effects of the compounds toward the human cell lines HeLa (cervical cancer), H460M2 (non-small-cell lung carcinoma), and HBL-100 (non-tumorigenic epithelial cells) are reported. Three of the compounds are not cytotoxic in the dark up to 100 µm, and the fourfold-symmetric couple revealed very good phototoxic indexes (PIs). The intracellular localization of all derivatives was studied in HeLa cells by confocal fluorescence microscopy. Although low nuclear localization was observed for some of them, it still prompted us to investigate their capacity to bind both quadruplex and duplex DNA; we observed significant selectivity in the tris-methylpyridinium derivatives for G-quadruplex interactions.


Asunto(s)
ADN/química , ADN/efectos de los fármacos , Compuestos Organometálicos/toxicidad , Fármacos Fotosensibilizantes/toxicidad , Porfirinas/química , Renio/química , Agua/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , División del ADN , Relación Dosis-Respuesta a Droga , G-Cuádruplex/efectos de los fármacos , Células HeLa , Humanos , Luz , Estructura Molecular , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/química , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/química , Solubilidad , Relación Estructura-Actividad
16.
Chemistry ; 21(3): 1179-83, 2015 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-25421757

RESUMEN

Novel photoactive (metallo)porphyrins were synthesised and characterised. When irradiated with light at a wavelength greater than 600 nm, these porphyrins act as photosensitisers and show high cytotoxicity towards two different human cancer cell lines with IC50 values down to 0.4 µM. A paramagnetic copper(II) porphyrin is the first photosensitiser to display excellent phototoxicity, explained by the electron paramagnetic resonance (EPR) spin trapping of hydroxy radicals and experimentally confirmed by the discovery of elevated levels of reactive oxygen species (ROS) inside A2780 cells after irradiation with red light. This finding indicates that paramagnetic compounds should be considered for photodynamic therapy (PDT). Furthermore, an additive effect of cisplatin and a zinc porphyrin, both at subtherapeutic concentrations of 0.22 µM, was observed.


Asunto(s)
Antineoplásicos/química , Fármacos Fotosensibilizantes/química , Porfirinas/química , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisplatino/química , Cisplatino/uso terapéutico , Cisplatino/toxicidad , Complejos de Coordinación/química , Complejos de Coordinación/uso terapéutico , Complejos de Coordinación/toxicidad , Cobre/química , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Luz , Neoplasias/tratamiento farmacológico , Fotoquimioterapia , Fármacos Fotosensibilizantes/uso terapéutico , Fármacos Fotosensibilizantes/toxicidad , Porfirinas/toxicidad , Especies Reactivas de Oxígeno/metabolismo
17.
ChemMedChem ; 9(12): 2781-90, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25377650

RESUMEN

Six phosphorescent (2-phenyl)pyridine (ppy) gold(III) 2,4,6-tris(trifluoromethyl)phenyl (FMes) complexes were synthesized and investigated for their anticancer potential. The compounds demonstrated strong antiproliferative activity, with EC50 values in the low micromolar range, along with significant accumulation in HeLa cancer cells after treatment for only 6 h (up to 119 ng gold per milligram of protein as measured by high-resolution continuum source atomic spectroscopy). Enzyme inhibition studies showed interaction of the gold(III) complexes with thioredoxin reductase (TrxR), a key homeostasis-regulation flavoprotein. TrxR was inhibited with IC50 values in the micromolar range. Furthermore, five of the complexes displayed selectivity toward TrxR against glutathione reductase (GR, a disulfide reductase structurally related to TrxR) by up to >49-fold. Because no major differences in bioactivity were observed across the series, [(ppy)Au(FMes)(PPh3 )OTf] (complex 4) was chosen for further in-depth biological characterization. Complex 4 was also found to interact with guanosine monophosphate in (1) H NMR studies under long incubation times. Interestingly, 4 induced a significant increase in intracellular levels of reactive oxygen species, which led to late apoptotic events and cytocidal effects.


Asunto(s)
Antineoplásicos/química , Complejos de Coordinación/química , Oro/química , Piridinas/química , Antineoplásicos/metabolismo , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Complejos de Coordinación/metabolismo , Complejos de Coordinación/toxicidad , Cristalografía por Rayos X , Estabilidad de Medicamentos , Glutatión Reductasa/antagonistas & inhibidores , Glutatión Reductasa/metabolismo , Células HeLa , Humanos , Conformación Molecular , Unión Proteica , Especies Reactivas de Oxígeno/metabolismo , Reductasa de Tiorredoxina-Disulfuro/antagonistas & inhibidores , Reductasa de Tiorredoxina-Disulfuro/metabolismo
18.
Chemistry ; 20(44): 14421-36, 2014 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-25213439

RESUMEN

Six substitutionally inert [Ru(II) (bipy)2 dppz](2+) derivatives (bipy=2,2'-bipyridine, dppz=dipyrido[3,2-a:2',3'-c]phenazine) bearing different functional groups on the dppz ligand [NH2 (1), OMe (2), OAc (3), OH (4), CH2 OH (5), CH2 Cl (6)] were synthesized and studied as potential photosensitizers (PSs) in photodynamic therapy (PDT). As also confirmed by DFT calculations, all complexes showed promising (1) O2 production quantum yields, well comparable with PSs available on the market. They can also efficiently intercalate into the DNA double helix, which is of high interest in view of DNA targeting. The cellular localization and uptake quantification of 1-6 were assessed by confocal microscopy and high-resolution continuum source atomic absorption spectrometry. Compound 1, and especially 2, showed very good uptake in cervical cancer cells (HeLa) with preferential nuclear accumulation. None of the compounds studied was found to be cytotoxic in the dark on both HeLa cells and, interestingly, on noncancerous MRC-5 cells (IC50 >100 µM). However, 1 and 2 showed very promising behavior with an increment of about 150 and 42 times, respectively, in their cytotoxicities upon light illumination at 420 nm in addition to a very good human plasma stability. As anticipated, the preferential nuclear accumulation of 1 and 2 and their very high DNA binding affinity resulted in very efficient DNA photocleavage, suggesting a DNA-based mode of phototoxic action.


Asunto(s)
Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Sustancias Intercalantes/química , Sustancias Intercalantes/farmacología , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Rutenio/química , 2,2'-Dipiridil/química , Complejos de Coordinación/síntesis química , Cristalografía por Rayos X , ADN/efectos de los fármacos , Células HeLa , Humanos , Sustancias Intercalantes/síntesis química , Procesos Fotoquímicos , Fotoquimioterapia , Fármacos Fotosensibilizantes/síntesis química
19.
Mol Cancer ; 13: 221, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25253202

RESUMEN

BACKGROUND: Cancer cells in the advanced stage show aberrant antioxidant capacity to detoxify excessive ROS resulting in the compensation for intrinsic oxidative stress and therapeutic resistance. PDAC is one of the most lethal cancers and often associated with a high accumulation of ROS. Recent studies identified gold(I) NHC complexes as potent TrxR inhibitors suppressing cell growth in a wide spectrum of human malignant cell lines at the low micromolar concentration. However, the mechanism of action is not completely elucidated yet. METHODS: To understand the biological function of gold(I) NHC complexes in PDAC, we used a recently published gold(I) NHC complex, MC3, and evaluated its anti-proliferative effect in four PDAC cell lines, determined by MTT and SRB assays. In further detailed analysis, we analyzed cellular ROS levels using the ROS indicator DHE and mitochondrial membrane potential indicated by the dye JC-1 in Panc1. We also analyzed cell cycle arrest and apoptosis by FACS. To elucidate the role of specific cell signaling pathways in MC3-induced cell death, co-incubation with ROS scavengers, a p38-MAPK inhibitor and siRNA mediated depletion of ASK1 were performed, and results were analyzed by immunoblotting, ELISA-microarrays, qRT-PCR and immunoprecipitation. RESULTS: Our data demonstrate that MC3 efficiently suppressed cell growth, and induced cell cycle arrest and apoptosis in pancreatic cancer cells, in particular in the gemcitabine-resistant cancer cells Panc1 and ASPC1. Treatment with MC3 resulted in a substantial alteration of the cellular redox homeostasis leading to increased ROS levels and a decrease in the mitochondrial membrane potential. ROS scavengers suppressed ROS formation and rescued cells from damage. On the molecular level, MC3 blocked the interaction of Trx with ASK1 and subsequently activated p38-associated signaling. Furthermore, inhibition of this pathway by using ASK1 siRNA or a p38 inhibitor clearly attenuated the effect of MC3 on cell proliferation in Panc1 and ASPC1. CONCLUSIONS: Our results confirm that MC3 is a TrxR inhibitor and show MC3 induced apoptosis in gemcitabine-resistant PDACs. MC3 mediated cell death could be blocked by using anti-oxidants, ASK1 siRNA or p38 inhibitor suggesting that the Trx-ASK1-p38 signal cascade played an important role in gold(I) NHC complexes-mediated cellular damage.


Asunto(s)
Apoptosis/efectos de los fármacos , MAP Quinasa Quinasa Quinasa 5/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Compuestos Orgánicos de Oro/farmacología , Neoplasias Pancreáticas/patología , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Densitometría , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Depuradores de Radicales Libres/farmacología , Homeostasis/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Oxidación-Reducción/efectos de los fármacos , Neoplasias Pancreáticas/enzimología , Unión Proteica/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Tiorredoxinas/metabolismo , Gemcitabina
20.
Parasit Vectors ; 7: 424, 2014 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-25190030

RESUMEN

BACKGROUND: Schistosomiasis is a neglected tropical disease and drug-repurposing is a useful strategy to fill its exhausted drug development pipeline. The ferrocenyl analogue of chloroquine, ferroquine, is an antimalarial in late-stage drug development. The aim of the present work was to study the antischistosomal activity of ferroquine against Schistosoma mansoni adult worms and newly transformed schistosomula (NTS) in vitro and in vivo. Hydroxyl-ferroquine and ruthenoquine were included to study the potential role of reactive oxygen species in the antischistosomal activity. Chloroquine and mefloquine, the later described for its antischistosomal properties, served as comparators. FINDINGS: All metal complexes were shown to be moderately cytotoxic on human cervix HeLa cancer cells and human fetal lung fibroblasts MRC-5. 72 hours post-incubation NTS exposed to 33.3 µM ruthenoquine had died, while ferroquine and hydroxyl-ferroquine treated worms were strongly affected. No activity was observed treating NTS with chloroquine at 33.3 µM. Incubation of adult S. mansoni with 33.3 µM of the organometallic derivatives were highly affected in viability but were still alive 72 hours post-incubation. Mefloquine showed the highest activity against NTS and adult S. mansoni. Low total worm burden reductions of 0-36% were observed following oral administration of 200-800 mg/kg of the ferroquine derivatives to S. mansoni-infected mice. CONCLUSIONS: The organometallic compounds evaluated in this study revealed moderate in vitro activity against both larval and adult stages of S. mansoni but low in vivo activity. No correlation can be drawn between the antimalarial and antischistosomal activity of chloroquine analogues and oxidative shock does not seem to play a role in the activity of these compounds against S. mansoni.


Asunto(s)
Aminoquinolinas/farmacología , Compuestos Ferrosos/farmacología , Schistosoma mansoni/efectos de los fármacos , Esquistosomiasis mansoni/tratamiento farmacológico , Esquistosomicidas/farmacología , Aminoquinolinas/química , Animales , Femenino , Compuestos Ferrosos/química , Células HeLa , Humanos , Metalocenos , Ratones , Estructura Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...