Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Biol Inorg Chem ; 28(7): 669-678, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37624480

RESUMEN

(1R,2R-diaminocyclohexane)(dihydropyrophosphato) platinum(II), also abbreviated as RRD2, belongs to a class of potent antitumor platinum cytostatics called phosphaplatins. Curiously, several published studies have suggested significant mechanistic differences between phosphaplatins and conventional platinum antitumor drugs. Controversial findings have been published regarding the role of RRD2 binding to DNA in the mechanism of its antiproliferative activity in cancer cells. This prompted us to perform detailed studies to confirm or rule out the role of RRD2 binding to DNA in its antiproliferative effect in cancer cells. Here, we show that RRD2 exhibits excellent antiproliferative activity in various cancer cell lines, with IC50 values in the low micromolar or submicromolar range. Moreover, the results of this study demonstrate that DNA lesions caused by RRD2 contribute to killing cancer cells treated with this phosphaplatin derivative. Additionally, our data indicate that RRD2 accumulates in cancer cells but to a lesser extent than cisplatin. On the other hand, the efficiency of cisplatin and RRD2, after they accumulate in cancer cells, in binding to nuclear DNA is similar. Our results also show that RRD2 in the medium, in which the cells were cultured before RRD2 accumulated inside the cells, remained intact. This result is consistent with the view that RRD2 is activated by releasing free pyrophosphate only in the environment of cancer cells, thereby allowing RRD2 to bind to nuclear DNA.


Asunto(s)
Antineoplásicos , Neoplasias , Oxaliplatino/farmacología , Cisplatino/farmacología , Platino (Metal)/farmacología , Difosfatos/farmacología , Compuestos Organoplatinos/farmacología , Compuestos Organoplatinos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/metabolismo , ADN/metabolismo
2.
Inorg Chem ; 62(16): 6474-6487, 2023 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-37040203

RESUMEN

We present the synthesis and characterization of six new heteroleptic osmium(II) complexes of the type [Os(C^N)(N^N)2]OTf (N^N = 2,2'-bipyridine and dipyrido[3,2-d:2',3'-f]quinoxaline; C^N = deprotonated methyl 1-butyl-2aryl-benzimidazolecarboxylate) with varying substituents in the R3 position of the phenyl ring of the cyclometalating C^N ligand. The new compounds are highly kinetically inert and absorb a full-wavelength range of visible light. An investigation of the antiproliferative activity of the new compounds has been performed using a panel of human cancer and noncancerous 2D cell monolayer cultures under dark conditions and green light irradiation. The results demonstrate that the new Os(II) complexes are markedly more potent than conventional cisplatin. The promising antiproliferative activity of selected Os(II) complexes was also confirmed using 3D multicellular tumor spheroids, which have the characteristics of solid tumors and can mimic the tumor tissue microenvironment. The mechanism of antiproliferative action of complexes has also been investigated and revealed that the investigated Os(II) complexes activate the endoplasmic reticulum stress pathway in cancer cells and disrupt calcium homeostasis.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Neoplasias , Humanos , Relación Estructura-Actividad , Osmio/farmacología , Calcio , Línea Celular Tumoral , Bencimidazoles/farmacología , Homeostasis , Antineoplásicos/farmacología , Complejos de Coordinación/farmacología
3.
J Biol Inorg Chem ; 25(6): 913-924, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32851480

RESUMEN

The search for more effective platinum anticancer drugs has led to the design, synthesis, and preclinical testing of hundreds of new platinum complexes. This search resulted in the recognition and subsequent FDA approval of the third-generation Pt(II) anticancer drug, [Pt(1,2-diaminocyclohexane)(oxalate)], oxaliplatin, as an effective agent in treating colorectal and gastrointestinal cancers. Another promising example of the class of anticancer platinum(II) complexes incorporating the Pt(1,n-diaminocycloalkane) moiety is kiteplatin ([Pt(cis-1,4-DACH)Cl2], DACH = diaminocyclohexane). We report here our progress in evaluating the role of the cycloalkyl moiety in these complexes focusing on the synthesis, characterization, evaluation of the antiproliferative activity in tumor cells and studies of the mechanism of action of new [Pt(cis-1,3-diaminocycloalkane)Cl2] complexes wherein the cis-1,3-diaminocycloalkane group contains the cyclobutyl, cyclopentyl, and cyclohexyl moieties. We demonstrate that [Pt(cis-1,3-DACH)Cl2] destroys cancer cells with greater efficacy than the other two investigated 1,3-diamminocycloalkane derivatives, or cisplatin. Moreover, the investigated [Pt(cis-1,3-diaminocycloalkane)Cl2] complexes show selectivity toward tumor cells relative to non-tumorigenic normal cells. We also performed several mechanistic studies in cell-free media focused on understanding some early steps in the mechanism of antitumor activity of bifunctional platinum(II) complexes. Our data indicate that reactivities of the investigated [Pt(cis-1,3-diaminocycloalkane)Cl2] complexes and cisplatin with glutathione and DNA binding do not correlate with antiproliferative activity of these platinum(II) complexes in cancer cells. In contrast, we show that the higher antiproliferative activity in cancer cells of [Pt(cis-1,3-DACH)Cl2] originates from its highest hydrophobicity and most efficient cellular uptake.


Asunto(s)
Antineoplásicos/síntesis química , Hidrocarburos Cíclicos/síntesis química , Compuestos Organometálicos/síntesis química , Platino (Metal)/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Cisplatino/normas , ADN/química , Ensayos de Selección de Medicamentos Antitumorales , Glutatión/química , Humanos , Compuestos Organometálicos/farmacología
4.
J Biol Inorg Chem ; 25(2): 339-350, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32112290

RESUMEN

The adverse side effects and acquired resistance associated with the clinical application of traditional platinum-based anticancer drugs have forced investigation of alternative transition metal-based compounds and their cytostatic properties. Over the last years, the anticancer potential of cobalt complexes has been extensively studied, and in-depth analyses of their mode of action have been conducted. In this work, we present antiproliferative activity against human cancer cells of the dinuclear Co(III) complexes bearing the quinizarin ligand and tris(2-aminoethyl)amine (tren, compound 1) or tris(2-pyridylmethyl)amine (tpa, compound 2) co-ligands. To contribute the understanding mechanisms of biological action of these compounds, their association with DNA in the cells, DNA binding in cell-free media, and DNA cleavage capability were investigated in detail. The results demonstrate that both complexes interact with DNA in tumor cells. However, their mechanism of antiproliferative action is different, and this difference is mirrored by distinct antiproliferative activity. The antiproliferative effect of 1 is connected with its ability to intercalate into DNA and subsequently to inhibit activities of DNA processing enzymes. In contrast, the total antiproliferative efficiency of 2, thanks to its redox properties, appears to be connected with its ability to form radicals and, consequently, with the ability of 2 to cleave DNA. Hence, the findings presented in this study may significantly contribute to understanding the antitumor potential of cobalt complexes. Dinuclear Co(III) complexes containing the bioactive quinizarin ligand exhibit antiproliferative activity based on distinct mechanism.


Asunto(s)
Antraquinonas/farmacología , Antineoplásicos/farmacología , Cobalto/farmacología , Complejos de Coordinación/farmacología , ADN/química , Antraquinonas/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Sitios de Unión/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cobalto/química , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , División del ADN , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ligandos , Conformación Molecular , Células Tumorales Cultivadas
5.
Inorg Chem ; 59(5): 3304-3311, 2020 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-32064865

RESUMEN

The water-compatible optically pure metallohelices made by self-assembly of simple nonpeptidic organic components around Fe(II) ions are now recognized as a distinct subclass of helicates that exhibit similar architecture to some natural cationic antimicrobial peptides. Notably, a new series of metallohelices was recently shown to exhibit biological activity, displaying high, structure-dependent activity against bacteria. It is also important that, thanks to their properties, such metallohelices can exhibit specific interactions with biomacromolecules. Here, following our prior report on the metallohelices that have high, structure-dependent activity against bacteria, we investigated the interactions of the series of iron(II) metallohelices with DNA, which is a potential pharmacological target of this class of coordination compounds. The results obtained with the aid of biophysical and molecular biology methods show that the investigated metallohelices accumulate in eukaryotic cells and that a significant fraction of the metallohelices accumulates in the cell nucleus, allowing them to interact also with nuclear DNA. Additionally, we have demonstrated that some metallohelices have a high affinity to DNA and are able to condense/aggregate DNA molecules more efficiently than conventional DNA-condensing agents, such as polyamines. Moreover, this capability of the metallohelices correlates with their efficiency to inhibit DNA-related enzymatic activities, such as those connected with DNA transcription, catalysis of DNA relaxation by DNA topoisomerase I, and cleavage by restriction enzymes.


Asunto(s)
Núcleo Celular/química , ADN-Topoisomerasas de Tipo I/metabolismo , ADN/antagonistas & inhibidores , Compuestos Ferrosos/farmacología , Inhibidores de Topoisomerasa I/farmacología , Núcleo Celular/metabolismo , ADN/genética , ADN/metabolismo , Compuestos Ferrosos/síntesis química , Compuestos Ferrosos/química , Células HCT116 , Humanos , Estructura Molecular , Fenómenos Ópticos , Inhibidores de Topoisomerasa I/síntesis química , Inhibidores de Topoisomerasa I/química
6.
Angew Chem Int Ed Engl ; 59(34): 14677-14685, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32489012

RESUMEN

Monosaccharides are added to the hydrophilic face of a self-assembled asymmetric FeII metallohelix, using CuAAC chemistry. The sixteen resulting architectures are water-stable and optically pure, and exhibit improved antiproliferative selectivity against colon cancer cells (HCT116 p53+/+ ) with respect to the non-cancerous ARPE-19 cell line. While the most selective compound is a glucose-appended enantiomer, its cellular entry is not mainly glucose transporter-mediated. Glucose conjugation nevertheless increases nuclear delivery ca 2.5-fold, and a non-destructive interaction with DNA is indicated. Addition of the glucose units affects the binding orientation of the metallohelix to naked DNA, but does not substantially alter the overall affinity. In a mouse model, the glucose conjugated compound was far better tolerated, and tumour growth delays for the parent compound (2.6 d) were improved to 4.3 d; performance as good as cisplatin but with the advantage of no weight loss in the subjects.


Asunto(s)
Glicoconjugados/química , Metales/química , Neoplasias/patología , Células HCT116 , Humanos , Espectroscopía de Protones por Resonancia Magnética , Espectrometría de Masa por Ionización de Electrospray
7.
Chemistry ; 24(18): 4607-4619, 2018 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-29369444

RESUMEN

A series of five kinetically inert bis-cyclometalated IrIII complexes of general formula [Ir(C^N)2 (N^N)][PF6 ] [C^N=2-phenyl-1-[4-(trifluoromethyl)benzyl]-1H-benzo[d]imidazol-κN,C; N^N=1,10-phenanthroline (phen, 1), dipyrido[3,2-d:2',3'-f]quinoxaline (dpq, 2), dipyrido[3,2-a:2',3'-c]phenazine (dppz, 3), benzo[i]dipyrido[3,2-a:2',3'-c]phenazine (dppn, 4), and dipyrido[3,2-a:2',3'-c]phenazine-10,11-imidazolone (dppz-izdo, 5)] were designed and synthesized to explore the effect of the degree of π conjugation of the polypyridyl ligand on their toxicity in cancer cells. We show that less-lipophilic complexes 1 and 2 exhibit the highest toxicity [sub-micromolar inhibitory concentration (IC50 ) values] in A2780, HeLa, and MCF-7 cancer cells, and they are markedly more efficient than clinically used platinum drugs. It is noteworthy that the investigated Ir agents display the capability to overcome acquired and inherent resistance to conventional cisplatin (in A2780cisR and MCF-7 cells, respectively). We demonstrate that the Ir complexes, unlike clinically used platinum antitumor drugs, do not kill cells through DNA-damage response. Rather, they kill cells by inhibiting protein translation by targeting preferentially the endoplasmic reticulum. Our findings also reveal that the toxic effect of the Ir complexes can be significantly potentiated by irradiation with visible light (by more than two orders of magnitude). The photopotentiation of the investigated Ir complexes can be attributed to a marked increase (≈10-30-fold) in intracellular reactive oxygen species. Collectively, these data highlight the functional diversity of antitumor metal-based drugs and the usefulness of a mechanism-based rationale for selecting candidate agents that are effective against chemoresistant tumors for further preclinical testing.


Asunto(s)
Antineoplásicos/farmacología , Polímeros/farmacología , Piridinas/farmacología , Animales , Antineoplásicos/química , Ligandos , Polímeros/química , Piridinas/química
8.
J Biol Inorg Chem ; 18(5): 579-89, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23674329

RESUMEN

The cisplatin analogues cis-[PtCl2(3ClHaza)2] (1) and cis-[PtCl2(3IHaza)2] (2) (3ClHaza and 3IHaza are 3-chloro-7-azaindole and 3-iodo-7-azaindole, respectively) are quite toxic to ovarian tumor cells, with moderately better IC50 values than for cisplatin in the cisplatin-sensitive cell line A2780. We investigated potential factors which might be involved in the mechanism underlying the cytotoxic effects of 1 and 2 and compared these factors with those involved in the mechanism underlying the effects of conventional cisplatin. Our data indicate that the higher cytotoxicity of 1 and 2 originates mainly from their efficient cellular accumulation, different effects at the level of cell cycle regulation, and reduced propensity for DNA adduct repair. Studies of their reactivity toward cellular components reveal efficient binding to DNA, which is typically required for an active platinum drug. Further results suggest that 1 and 2 are capable of circumventing resistance to cisplatin induced by alterations in cellular accumulation and DNA repair. Hence, the latter two factors appear to be responsible for differences in the toxicity of 1 or 2, and cisplatin in tumor cells. The results of this work reinforce the idea that direct analogues of conventional cisplatin-containing halogeno-substituted 7-azaindoles offer much promise for the design of novel therapeutic agents.


Asunto(s)
Compuestos Organoplatinos/toxicidad , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Reparación del ADN , Glutatión/química , Humanos , Compuestos Organoplatinos/química
9.
Chemistry ; 19(29): 9578-91, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23733242

RESUMEN

Platinum diam(m)ine complexes, such as cisplatin, are successful anticancer drugs, but suffer from problems of resistance and side-effects. Photoactivatable Pt(IV) prodrugs offer the potential of targeted drug release and new mechanisms of action. We report the synthesis, X-ray crystallographic and spectroscopic properties of photoactivatable diazido complexes trans,trans,trans-[Pt(N3)2(OH)2(MA)(Py)] (1; MA=methylamine, Py=pyridine) and trans,trans,trans-[Pt(N3)2(OH)2(MA)(Tz)] (2; Tz=thiazole), and interpret their photophysical properties by TD-DFT modelling. The orientation of the azido groups is highly dependent on H bonding and crystal packing, as shown by polymorphs 1p and 1q. Complexes 1 and 2 are stable in the dark towards hydrolysis and glutathione reduction, but undergo rapid photoreduction with UVA or blue light with minimal amine photodissociation. They are over an order of magnitude more potent towards HaCaT keratinocytes, A2780 ovarian, and OE19 oesophageal carcinoma cells than cisplatin and show particular potency towards cisplatin-resistant human ovarian cancer cells (A2780cis). Analysis of binding to calf-thymus (CT), plasmids, oligonucleotide DNA and individual nucleotides reveals that photoactivated 1 and 2 form both mono- and bifunctional DNA lesions, with preference for G and C, similar to transplatin, but with significantly larger unwinding angles and a higher percentage of interstrand cross-links, with evidence for DNA strand cross-linking further supported by a comet assay. DNA lesions of 1 and 2 on a 50 bp duplex were not recognised by HMGB1 protein, in contrast to cisplatin-type lesions. The photo-induced platination reactions of DNA by 1 and 2 show similarities with the products of the dark reactions of the Pt(II) compounds trans-[PtCl2(MA)(Py)] (5) and trans-[PtCl2(MA)(Tz)] (6). Following photoactivation, complex 2 reacted most rapidly with CT DNA, followed by 1, whereas the dark reactions of 5 and 6 with DNA were comparatively slow. Complexes 1 and 2 can therefore give rapid potent photocytotoxicity and novel DNA lesions in cancer cells, with no activity in the absence of irradiation.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Azidas/química , Azidas/farmacología , Cisplatino/química , Aductos de ADN/química , ADN/química , Compuestos Organoplatinos/química , Compuestos Organoplatinos/farmacología , Platino (Metal)/química , Profármacos/química , Piridinas/química , Química Farmacéutica , Cisplatino/farmacología , Cristalografía por Rayos X , Humanos , Luz
10.
Chem Res Toxicol ; 25(5): 1099-111, 2012 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-22420335

RESUMEN

The Pt(IV) diazido complex trans,trans,trans-[Pt(N(3))(2)(OH)(2)(pyridine)(2)] (1) is unreactive in the dark but is cytotoxic when photoactivated by UVA and visible light. We have shown that 1 when photoactivated accumulates in tumor cells and binds strongly to nuclear DNA under conditions in which it is toxic to tumor cells. The nature of the DNA adducts, including conformational alterations, induced by photoactivated 1 are distinctly different from those produced in DNA by conventional cisplatin or transplatin. In addition, the observation that major DNA adducts of photoactivated 1 are able to efficiently stall RNA polymerase II more efficiently than cisplatin suggests that transcription inhibition may contribute to the cytotoxicity levels observed for photoactivated 1. Hence, DNA adducts of 1 could trigger a number of downstream cellular effects different from those triggered in cancer cells by DNA adducts of cisplatin. This might lead to the therapeutic effects that could radically improve chemotherapy by platinum complexes. The findings of the present work help to explain the different cytotoxic effects of photoactivated 1 and conventional cisplatin and thereby provide new insights into mechanisms associated with the antitumor effects of platinum complexes photoactivated by UVA and visible light.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Aductos de ADN/metabolismo , Compuestos Organoplatinos/química , Compuestos Organoplatinos/farmacología , Piridinas/química , Piridinas/farmacología , Animales , Azidas/química , Azidas/farmacología , Bovinos , Línea Celular Tumoral , ADN/metabolismo , Aductos de ADN/química , Femenino , Humanos , Luz , Neoplasias Ováricas/tratamiento farmacológico , Tiourea/metabolismo , Rayos Ultravioleta
11.
Inorg Chem Front ; 7: 4150-4159, 2020 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-34540235

RESUMEN

The platinum(IV) prodrug trans,trans,trans-[Pt(N3)2(OH)2(py)2] (1) is stable and non-toxic in the dark, but potently cytotoxic to cancer cells when irradiated by visible light, including cisplatin-resistant cells. On irradiation with visible light, it generates reactive Pt(II) species which can attack DNA, and produces reactive oxygen species (ROS) and reactive nitrogen species (RNS) which exert unusual effects on biochemical pathways. We now show that its novel mechanism of action includes induction of immunogenic cell death (ICD). Treatment of cancer cells with 1 followed by photoirradiation with visible light induces calreticulin (CRT) expression at the surface of dying cancer cells. This is accompanied by release of high mobility group protein-1B (HMGB1) and the secretion of ATP. Autophagy appears to play a key role in this chemotherapeutically-stimulated ICD. The observed uneven distribution of ecto-CRT promotes phagocytosis, confirmed by the observation of engulfment of photoirradiated CT26 colorectal cancer cells treated with 1 by J774.A1 macrophages. The photoactivatable prodrug 1 has a unique mechanism of action which distinguishes it from other platinum drugs due to its immunomodulating properties, which may enhance its anticancer efficacy.

12.
Dalton Trans ; 47(35): 12197-12208, 2018 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-30112527

RESUMEN

There is an urgent need to discover new, selective compounds to add to the limited arsenal of chemotherapeutics displaying selective toxicity for aggressive triple-negative breast cancer (TNBC) cells. The effect of two, recently developed metal-based half-sandwich complexes [Os(η6-pcym)(bphen)(dca)]PF6 (Os-dca) and [Ru(η6-pcym)(bphen)(dca)]PF6 (Ru-dca) [pcym = 1-methyl-4-(propan-2-yl)benzene (p-cymene); bphen = 4,7-diphenyl-1,10-phenanthroline (bathophenanthroline); dca = dichloroacetate] on triple-negative breast cancer cells MDA-MB-231 is reported. The complexes display selective toxicity in several tumor cells (at submicromolar concentrations), and a prominent effect is observed against highly progressive triple negative breast cancer MDA-MB-231 cells for Os-dca. The lower potency of Ru-dca in comparison with Os-dca is apparently connected with a relatively quick release of the dca ligand due to the hydrolysis of Ru-dca before this complex enters the cells. Remarkably, both Os-dca and Ru-dca reduce successfully metastasis-related properties of the triple-negative breast cancer cells such as migration, invasion, and re-adhesion. The anti-metastatic effects of Os-dca and Ru-dca are associated with their ability to suppress matrix metalloproteinase activity and/or production and reduce the expression of aquaporins. Further detailed mechanistic studies reveal that Os-dca reverses Warburg's effect and oncosis seems to be a prominent mode of cell death that predominates over apoptosis. As such, Os-dca can efficiently overcome the resistance of cancer cells to clinically-used apoptotic inducers cisplatin and carboplatin. The cytostatic and anti-metastatic properties of Os-dca in MDA-MB-231 provide a strong impetus for the development of new metal-based compounds to target hardly treatable human TNBC cells and displaying different modes of action compared to the antitumor metallodrugs in clinical use.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Osmio/farmacología , Fenantrolinas/farmacología , Rutenio/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Estructura Molecular , Osmio/química , Fenantrolinas/química , Rutenio/química
13.
Dalton Trans ; 46(41): 14139-14148, 2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-28972623

RESUMEN

One of the promising new antitumor platinum complexes is a large-ring chelate complex [PtCl2(cis-1,4-DACH)] (DACH = diaminocyclohexane) (kiteplatin). Recently, new platinum(ii) derivatives of kiteplatin with pyrophosphate as a carrier ligand have been synthesized and tested on a panel of human cancer cell lines. These derivatives of kiteplatin were found to be more effective than clinically used anticancer platinum drugs. The design of kiteplatin pyrophosphate derivatives was based on the concept of pyrophosphate coordinated platinum complexes, phosphaplatins. Phosphaplatins have been shown to function without binding to DNA and hence DNA has been excluded as the target of phosphaplatins in contrast to conventional antitumor platinum drugs. Cytotoxicity, major cellular targets and DNA interactions of the new anticancer platinum drug were characterized by standard biochemical methods and methods of molecular and cellular biology. We demonstrate that, in contrast to what has been reported on closely related phosphaplatins, the derivatives of kiteplatin with the pyrophosphate carrier ligand are activated in the cellular environment. This activation, which yields species capable of platination of DNA, very likely comprises the hydrolytic release of the pyrophosphate ligand that could be enzymatically catalyzed. Collectively, these data provide convincing evidence that unexpectedly DNA is an important target for the biological activity of the kiteplatin pyrophosphate derivatives, although the overall mechanism of action might be different from those of conventional platinum drugs.


Asunto(s)
Antineoplásicos/química , Complejos de Coordinación/metabolismo , ADN/metabolismo , Difosfatos/química , Compuestos Organoplatinos/química , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Células CHO , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Cricetinae , Cricetulus , ADN/química , Reparación del ADN/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Escherichia coli/fisiología , Humanos , Pruebas de Mutagenicidad , Plásmidos/genética , Plásmidos/metabolismo , Análisis de la Célula Individual
14.
Sci Rep ; 7(1): 3751, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28623355

RESUMEN

We present the anticancer properties of cis, cis, trans-[Pt(IV)(NH3)2Cl2(OA)2] [Pt(IV)diOA] (OA = octanoato), Pt(IV) derivative of cisplatin containing two OA units appended to the axial positions of a six-coordinate Pt(IV) center. Our results demonstrate that Pt(IV)diOA is a potent cytotoxic agent against many cancer cell lines (the IC50 values are approximately two orders of magnitude lower than those of clinically used cisplatin or Pt(IV) derivatives with biologically inactive axial ligands). Importantly, Pt(IV)diOA overcomes resistance to cisplatin, is significantly more potent than its branched Pt(IV) valproato isomer and exhibits promising in vivo antitumor activity. The potency of Pt(IV)diOA is a consequence of several factors including enhanced cellular accumulation correlating with enhanced DNA platination and cytotoxicity. Pt(IV)diOA induces DNA hypermethylation and reduces mitochondrial membrane potential in cancer cells at levels markedly lower than the IC50 value of free OA suggesting the synergistic action of platinum and OA moieties. Collectively, the remarkable antitumor effects of Pt(IV)diOA are a consequence of the enhanced cellular uptake which makes it possible to simultaneously accumulate high levels of both cisplatin and OA in cells. The simultaneous dual action of cisplatin and OA by different mechanisms in tumor cells may result in a markedly enhanced and unique antitumor effects of Pt(IV) prodrugs.


Asunto(s)
Antineoplásicos , Cisplatino , Metilación de ADN/efectos de los fármacos , ADN de Neoplasias/metabolismo , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Ováricas , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Línea Celular Tumoral , Cisplatino/análogos & derivados , Cisplatino/farmacocinética , Cisplatino/farmacología , Femenino , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología
15.
J Inorg Biochem ; 160: 149-55, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26778426

RESUMEN

We report DNA cleavage by ruthenium(II) arene anticancer complex [(η(6)-p-terp)Ru(II)(en)Cl](+) (p-terp=para-terphenyl, en=1,2-diaminoethane, complex 1) after its photoactivation by UVA and visible light, and the toxic effects of photoactivated 1 in cancer cells. It was shown in our previous work (T. Bugarcic et al., J. Med. Chem. 51 (2008) 5310-5319) that this complex exhibits promising toxic effects in several human tumor cell lines and concomitantly its DNA binding mode involves combined intercalative and monofunctional (coordination) binding modes. We demonstrate in the present work that when photoactivated by UVA or visible light, 1 efficiently photocleaves DNA, also in hypoxic media. Studies of the mechanism underlying DNA cleavage by photoactivated 1 reveal that the photocleavage reaction does not involve generation of reactive oxygen species (ROS), although contribution of singlet oxygen ((1)O2) to the DNA photocleavage process cannot be entirely excluded. Notably, the mechanism of DNA photocleavage by 1 appears to involve a direct modification of mainly those guanine residues to which 1 is coordinatively bound. As some tumors are oxygen-deficient and cytotoxic effects of photoactivated ruthenium compounds containing {Ru(η(6)-arene)}(2+) do not require the presence of oxygen, this class of ruthenium complexes may be considered potential candidate agents for improved photodynamic anticancer chemotherapy.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , División del ADN/efectos de los fármacos , Etilenodiaminas/química , Compuestos Organometálicos/farmacología , Rutenio/química , Compuestos de Terfenilo/química , Antineoplásicos/síntesis química , Secuencia de Bases , Cationes Bivalentes , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Complejos de Coordinación/síntesis química , ADN/química , División del ADN/efectos de la radiación , Humanos , Luz , Compuestos Organometálicos/síntesis química , Fotoquimioterapia , Plásmidos/química , Oxígeno Singlete/química
16.
Dalton Trans ; 45(33): 13179-86, 2016 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-27396365

RESUMEN

The limitations associated with the clinical utility of conventional platinum anticancer drugs have stimulated research leading to the design of new metallodrugs with improved pharmacological properties, particularly with increased selectivity for cancer cells. Very recent research has demonstrated that photoactivation or photopotentiation of platinum drugs can be one of the promising approaches to tackle this challenge. This is so because the application of irradiation can be targeted exclusively to the tumor tissue so that the resulting effects could be much more selective and targeted to the tumor. We show in this work that the presence of 1-methyl-7-azaindole in trans-[PtCl2(NH3)(L)] (L = 1-methyl-7-azaindole, compound 1) markedly potentiated the DNA binding ability of 1 when irradiated by UVA light in a cell-free medium. Concomitantly, the formation of cytotoxic bifunctional cross-links was markedly enhanced. In addition, 1, when irradiated with UVA, was able to effectively cleave the DNA backbone also in living cells. The incorporation of 1-methyl-7-azaindole moiety had also a profound effect on the photophysical properties of 1, which can generate singlet oxygen responsible for the DNA cleavage reaction. Finally, we found that 1, upon irradiation with UVA light, exhibited a pronounced dose-dependent decrease in viability of A2780 cells whereas it was markedly less cytotoxic if the cells were treated in the absence of light. Hence, it is possible to conclude that 1 is amenable to photodynamic therapy.


Asunto(s)
Antineoplásicos , Cisplatino , Indoles , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/efectos de la radiación , Línea Celular Tumoral , Cisplatino/química , Cisplatino/farmacología , Cisplatino/efectos de la radiación , ADN/efectos de los fármacos , ADN/metabolismo , División del ADN , Fragmentación del ADN , Humanos , Indoles/química , Indoles/farmacología , Indoles/efectos de la radiación , Ligandos , Especies Reactivas de Oxígeno/metabolismo , Oxígeno Singlete/química , Rayos Ultravioleta
17.
Dalton Trans ; 44(8): 3573-82, 2015 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-25406573

RESUMEN

The toxicity of the new derivative of transplatin, namely trans-[PtCl2(DEA)(NH3)] (DEA = diethylamine), in which only one NH3 group was replaced by a small, non-bulky DEA ligand, in the cisplatin sensitive and resistant tumor cell lines was examined. The results indicate that this very small modification of the transplatin molecule results in a considerable enhancement of toxicity in the cancer cells. Thus, this finding is consistent with the thesis that the trans geometry in Pt(II)-dichlorido compounds can also be effectively activated by the replacement of only one NH3 ligand in transplatin by the non-bulky ligand, such as the short aliphatic amine, (C2H5)2NH. We also demonstrate that trans-[PtCl2(DEA)(NH3)], in contrast to transplatin, can be grouped with the coordination compounds exhibiting antitumor activity and capable of inducing lysis in lysogenic bacteria. Thus, these results afford, for the first time, experimental support for the view that DNA is the potential cellular target also for antitumor derivatives of transplatin. DNA binding mode of trans-[PtCl2(DEA)(NH3)] in cell-free media was examined as well. The results show that the small aliphatic DEA ligand has significant consequences for the DNA conformational changes. Unlike 'classical' transplatin, modification of DNA with trans-[PtCl2(DEA)(NH3)] leads to mainly bifunctional cross-links. The extent and destabilization of the double-helical structure of DNA by this new trans-platinum complex are similar to those induced by cisplatin. As a consequence the lesions effectively inhibit transcription of template DNA similarly to the lesions of cisplatin, but markedly more than the lesions of transplatin. The stronger inhibition of DNA transcription by the adducts of trans-[PtCl2(DEA)(NH3)] in comparison with the adducts of transplatin adds a new dimension to the impact of the activated trans geometry in platinum compounds on biological processes, possibly including DNA transcription.


Asunto(s)
Antineoplásicos/química , Cisplatino/química , ADN/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Dicroismo Circular , Cisplatino/farmacología , ADN/metabolismo , Aductos de ADN/química , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Células MCF-7 , Transcripción Genética/efectos de los fármacos
18.
PLoS One ; 10(4): e0123595, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25875850

RESUMEN

The moderate-to-high in vitro cytotoxicity against ovarian A2780 (IC50 = 4.7-14.4 µM), prostate LNCaP (IC50 = 18.7-30.8 µM) and prostate PC-3 (IC50 = 17.6-42.3 µM) human cancer cell lines of the platinum(II) cyclobutane-1,1'-dicarboxylato complexes [Pt(cbdc)(naza)2] (1-6; cbdc = cyclobutane-1,1'-dicarboxylate(2-); naza = halogeno-substituted 7-azaindoles), derived from the anticancer metallodrug carboplatin, are reported. The complexes containing the chloro- and bromo-substituted 7-azaindoles (1, 2, and 4-6) showed a significantly higher (p < 0.05) cytotoxicity against A2780 cell line as compared to cisplatin used as a reference drug. Addition of the non-toxic concentration (5.0 µM) of L-buthionine sulfoximine (L-BSO, an effective inhibitor of γ-glutamylcysteine synthase) markedly increases the in vitro cytotoxicity of the selected complex 3 against A2780 cancer cell line by a factor of about 4.4. The cytotoxicity against A2780 and LNCaP cells, as well as the DNA platination, were effectively enhanced by UVA light irradiation (λmax = 365 nm) of the complexes, with the highest phototoxicity determined for compound 3, resulting in a 4-fold decline in the A2780 cells viability from 25.1% to 6.1%. The 1H NMR and ESI-MS experiments suggested that the complexes did not interact with glutathione as well as their ability to interact with guanosine monophosphate. The studies also confirmed UVA light induced the formation of the cis [Pt(H2O)2(cbdc`)(naza)] intermediate, where cbdc` represents monodentate-coordinated cbdc ligand, which is thought to be responsible for the enhanced cytotoxicity. This is further supported by the results of transcription mapping experiments showing that the studied complexes preferentially form the bifunctional adducts with DNA under UVA irradiation, in contrast to the formation of the less effective monofunctional adducts in dark.


Asunto(s)
Antineoplásicos/farmacología , Carboplatino/farmacología , Indoles/farmacología , Rayos Ultravioleta , Antineoplásicos/química , Butionina Sulfoximina/farmacología , Carboplatino/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Aductos de ADN/química , Aductos de ADN/genética , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Sinergismo Farmacológico , Humanos , Indoles/química , Estructura Molecular , Neoplasias/genética , Neoplasias/patología , Conformación de Ácido Nucleico , Espectroscopía de Protones por Resonancia Magnética , Espectrometría de Masa por Ionización de Electrospray
19.
J Med Chem ; 58(2): 847-59, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25496325

RESUMEN

The current work investigates the effect of new bifunctional and mononuclear Pt(II) compounds, the cis- and trans-isomers of [PtCl2(NH3)(L)] (L = 1-methyl-7-azaindole, compounds 1 and 2, respectively), on growth and viability of human carcinoma cells as well as their putative mechanism(s) of cytotoxicity. The results show that substitution of 1-methyl-7-azaindole for ammine in cisplatin or transplatin results in an increase of the toxic efficiency, selectivity for tumor cells in cisplatin-resistant cancer cells, and activation of the trans geometry. The differences in the cytotoxic activities of 1 and 2 were suggested to be due to their different DNA binding mode, different capability to induce cell cycle perturbations, and fundamentally different role of transcription factor p53 in their mechanism of action. Interestingly, both isomers make it possible to detect their cellular uptake and distribution in living cells by confocal microscopy without their modification with an optically active tag.


Asunto(s)
Antineoplásicos/síntesis química , Cisplatino/análogos & derivados , Compuestos Organoplatinos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , ADN/metabolismo , Humanos , Indoles/química , Compuestos Organoplatinos/química , Compuestos Organoplatinos/farmacología , Unión Proteica , Proteína p53 Supresora de Tumor/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA