首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
The binding of transition metal compounds to nucleic acids is discussed in the perspectives of kinetics and their anticancer activity. Kinetics of ligand exchange is primarily determined by the intrinsic properties of the metal ions, and to a lesser degree by the ligands coordinated already to the metal ion. Metal compounds having ligand-exchange rates of the same order of magnitude as cell-division processes, e.g. many Pt(iIIi), Ru(II) and Ru(III) compounds, are in use as chemotherapeutic drugs. Detailed knowledge of ligand exchange in such compounds is important for design of derivative and entirely new compounds. Metal coordination compounds of metal ions with much faster ligand-exchange reactions interact with DNA in a quite different way, namely primarily by compensation of negative charge of the polyanionic chain and are usually not active as anticancer agents. Examples of our recent work are presented in relation with experiments performed by others on new generations of platinum anti-cancer drugs.  相似文献   

2.
The clinical application of Pt-based anticancer drugs has inspired the development of novel chemotherapeutic metallodrugs with improved efficacies. Pt(IV) prodrugs are one of the most promising successors of Pt(II) drugs and have displayed great anticancer performance. In particular, judicious modification of axial ligands endows Pt(IV) complexes with unique properties that enable them to overcome the limitations of conventional Pt(II) drugs. Herein, we summarize recent developments in Pt(IV) anticancer complexes, with a focus on their axial functionalization with other anticancer agents, immunotherapeutic agents, photosensitive ligands, peptides, and theranostic agents. We hope that this concise view of recently reported Pt(IV) coordination complexes will help researchers to design next-generation multi-functional anticancer agents based on a comprehensive Pt(IV) platform.  相似文献   

3.
An unusual coordination mode of salicylaldehyde-N-phenylthiosemicarbazone (H2-Sal-Ptsc) ligand was observed in unusual ruthenium(III) carbonyl complex for the first time when it was reacted with [RuHCl(CO)(PPh3)3]. The EPR and electrochemical analysis conformed the formation of Ru(III) species.  相似文献   

4.
A novel class of ruthenium (III) complexes of formulas K[Ru(sar)2Cl2].12H2O and K2[Ru(dmgly)Cl4].2H2O, containing bidentate chelates N-methylglycine (sarcosine, sar) or N,N-dimethylglycine (dmgly) and additional chloro ligands were synthesized. The complexes have been obtained by direct reaction of ruthenium(III) chloride with corresponding bidentate ligand followed by addition of base (KOH). These new complexes were characterized by elemental analysis, IR and electronic absorption spectroscopy. As astrocytomas, the most common of all brain tumors, are still very difficult to treat, we examined the influence of newly synthesized ruthenium-based complexes, as well as the earlier synthesized analogue platinum(IV) complexes [Pt(dmgly)2Cl2], [Pt(sar)2Br2] and [Pt(dmgly)2Br2], on rat astrocytoma C6 cells in vitro. Among these complexes only K2[Ru(dmgly)Cl4].2H2O and [Pt(dmgly)2Br2] markedly inhibited the viability of non-confluent C6 cells. Furthermore, only complex K2[Ru(dmgly)Cl4].2H2O was able to reduce viability in confluent C6 cultures. Importantly, this complex was not toxic to primary rat astrocytes or macrophages. Having in mind that appropriate chemotherapy should be effective against tumor cells without harming normal tissues, complex K2[Ru(dmgly)Cl4].2H2O could be a promising agent for developing therapeutics against astrocytomas.  相似文献   

5.
The rise of supramolecular chemistry offers new tools to design therapeutics and delivery platforms for biomedical applications. This review aims to highlight the recent developments that harness host-guest interactions and self-assembly to design novel supramolecular Pt complexes as anticancer agents and drug delivery systems. These complexes range from small host-guest structures to large metallosupramolecules and nanoparticles. These supramolecular complexes integrate the biological properties of Pt compounds and novel supramolecular structures, which inspires new designs of anticancer approaches that overcome problems in conventional Pt drugs. Based on the differences in Pt cores and supramolecular structures, this review focuses on five different types of supramolecular Pt complexes, and they include host-guest complexes of the FDA-approved Pt(II) drugs, supramolecular complexes of nonclassical Pt(II) metallodrugs, supramolecular complexes of fatty acid-like Pt(IV) prodrugs, self-assembled nanotherapeutics of Pt(IV) prodrugs, and self-assembled Pt-based metallosupramolecules.  相似文献   

6.
Interest in Ru anticancer drugs has been growing rapidly since NAMI-A ((ImH(+))[Ru(III)Cl(4)(Im)(S-dmso)], where Im = imidazole and S-dmso = S-bound dimethylsulfoxide) or KP1019 ((IndH(+))[Ru(III)Cl(4)(Ind)(2)], where Ind = indazole) have successfully completed phase I clinical trials and an array of other Ru complexes have shown promise for future development. Herein, the recent literature is reviewed critically to ascertain likely mechanisms of action of Ru-based anticancer drugs, with the emphasis on their reactions with biological media. The most likely interactions of Ru complexes are with: (i) albumin and transferrin in blood plasma, the former serving as a Ru depot, and the latter possibly providing active transport of Ru into cells; (ii) collagens of the extracellular matrix and actins on the cell surface, which are likely to be involved in the specific anti-metastatic action of Ru complexes; (iii) regulatory enzymes within the cell membrane and/or in the cytoplasm; and (iv) DNA in the cell nucleus. Some types of Ru complexes can also promote the intracellular formation of free radical species, either through irradiation (photodynamic therapy), or through reactions with cellular reductants. The metabolic pathways involve competition among reduction, aquation, and hydrolysis in the extracellular medium; binding to transport proteins, the extracellular matrix, and cell-surface biomolecules; and diffusion into cells; with the extent to which individual drugs participate in various steps along these pathways being crucial factors in determining whether they are mainly anti-metastatic or cytotoxic. This diversity of modes of action of Ru anticancer drugs is also likely to enhance their anticancer activities and to reduce the potential for them to develop tumour resistance. New approaches to metabolic studies, such as X-ray absorption spectroscopy and X-ray fluorescence microscopy, are required to provide further mechanistic insights, which could lead to the rational design of improved Ru anticancer drugs.  相似文献   

7.
The reactions of [Ru(PPh3)3Cl2], N-(benzoyl)-N′-(5-R-salicylidene)hydrazines (H2bhsR, R = H, OCH3, Cl, Br and NO2) and triethylamine (1:1:2 mole ratio) in methanol afford mononuclear ruthenium(III) complexes having the general formula trans-[Ru(bhsR)(PPh3)2Cl]. In the case of R = H, a dinuclear ruthenium(III) complex of formula [Ru2(μ-OCH3)2(bhsH)2(PPh3)2] has been isolated as a minor product. The complexes are characterized by elemental analysis, magnetic, spectroscopic and electrochemical measurements. The crystal structures of the dinuclear complex and two mononuclear complexes have been determined. In the dinuclear complex, each metal centre is in distorted octahedral NO4P coordination sphere constituted by the two bridging methoxide groups, one PPh3 molecule and the meridionally spanning phenolate-O, imine-N and amide-O donor bhsH2−. The terminal PPh3 ligands are trans to each other. In the mononuclear complexes, bhsR2− and the chlorine atom form an NO2Cl square-plane around the metal centre and the P-atoms of the two PPh3 molecules occupy the remaining two axial sites to complete a distorted octahedral NO2ClP2 coordination sphere. All the complexes display ligand-to-metal charge transfer bands in the visible region of the electronic spectra. The cryomagnetic measurements reveal the antiferromagnetic character of the diruthenium(III) complex. The low-spin mononuclear ruthenium(III) complexes as well as the diruthenium(III) complex display rhombic EPR spectra in frozen solutions. All the complexes are redox active in CH2Cl2 solutions. Two successive metal centred oxidations at 0.69 and 1.20 V (versus Ag/AgCl) are observed for the dinuclear complex. The mononuclear complexes display a metal centred reduction in the potential range −0.53 to −0.27 V. The trend in these potential values reflects the polar effect of the substituents on the salicylidene moiety of the tridentate ligand.  相似文献   

8.
A novel Ru(III) complex, mer-[RuCl(3)(CH(3)CN)(dpq)] (1), has been synthesized and characterized by X-ray diffraction, where dpq=dipyrido[3,2-d:2',3'-f]quinoxaline. Its chemical and biological properties have been intensively compared with those of mer-[RuCl(3)(DMSO)(dpq)] (DMSO=dimethyl sulfoxide) (2). It has been found that the stability in buffered solutions and the reduction potential for the Ru(III)/Ru(II) couple can be modulated by changing the small molecule bonded to the Ru(III) center. Interactions of 1 with DNA have been investigated by DNA melting experiments, DNA competitive binding with EB (ethidium bromide), plasmid DNA cleavage experiments and viscosity measurements. The interaction of 1 and 2 with BSA (bovine serum albumin) has also been studied using fluorescent quenching method. The experimental results show that 1 exerts higher affinity towards DNA and BSA than 2 does. The cytotoxicity of 1 has been evaluated by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) method, and 2 shows slightly higher anticancer potency than 1 does against all the cell lines screened. Attempts are made to clarify the possible antitumor mechanisms of these two complexes by analyzing the experimental results presented.  相似文献   

9.
Ascorbic acid has been previously discussed to have antitumor potential through its interaction with transition metal ions such as iron and copper. Furthermore, ascorbic acid may act as a reducing agent for Ru(III) compounds such as indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (KP1019), an investigational anticancer drug which is supposed to be activated by reduction, prior to binding to cellular target proteins. Therefore, we investigated the influence of ascorbic acid on the activity of this antitumor metal complex in cell culture studies. We show that co-incubation of equicytotoxic, constant amounts of KP1019 with high concentrations of ascorbic acid (50–700 μM) increases cytotoxicity of the ruthenium anticancer drug in the human colon carcinoma cell line SW480, human cervical carcinoma KB-3-1 cells, and the multidrug-resistant subline KBC-1, whereas addition of low concentrations (2.7–50 μM) has a strong chemoprotective effect in the human colon carcinoma cell line SW480, but not in multidrug-resistant KBC-1 cells. Although cellular uptake of KP1019 is not altered, ascorbic acid induce stronger interaction of the ruthenium compound with DNA both in SW480 cells and under cell-free conditions with plasmid DNA. Even if DNA interactions probably play a subordinate role in vivo given the extensive protein binding of the compound, our data exemplify that ascorbic acid enhances the reactivity of KP1019 with biomolecules. Moreover, we demonstrate that the levels of KP1019-generated reactive oxygen species are markedly decreased by co-incubation with ascorbic acid. Conclusively, our results indicate that application of high doses of ascorbic acid might increase the anticancer effects of KP1019.  相似文献   

10.
Development of new chemotherapeutic agents to treat microbial infections and recurrent cancers is of pivotal importance. Metal based drugs particularly ruthenium complexes have the uniqueness and desired properties that make them suitable candidates for the search of potential chemotherapeutic agents. In this study, two mixed ligand Ru(III) complexes [Ru(Cl)2(SB)(Phen] (RC-1) and [Ru(Cl)2(SB)(Bipy)] (RC-2) were synthesised and characterized by elemental analysis, IR, UV–Vis, 1H, 13C NMR spectroscopic techniques and their molecular structure was confirmed by X-ray crystallography. Antibacterial activity evaluation against two Gram-positive (S. pneumonia and E. faecalis) and four Gram-negative strains (P. aurogenosa, K. pneumoniae, S. enterica, and E. coli) revealed their moderate antibacterial activity with MIC value of ≥250 μg/mL. Anticancer activity evaluation against a non-small lung cancer cell line (H1299) revealed the tremendous anticancer activity of these complexes which was further validated by DNA binding and docking results. DNA binding profile of the complexes studied by UV–Visible and fluorescence spectroscopy showed an intercalative binding mode with CT-DNA and an intrinsic binding constant in the range of 3.481–1.015× 105 M−1. Both the complexes were also found to exert weak toxicity to human erythrocytes by haemolytic assay compared to cisplatin. Potential of these complexes as anticancer agents will be further delineated by in vivo studies.  相似文献   

11.
The reduction of Cl(NH3)5Ru(III) and subsequent binding of heterocyclic ligands by the resultant (H2O)(NH3)5Ru(II) ion is shown to be catalyzed by components of rat-liver cells. The presence of air significantly decreases the rate of heterocyclic ligand binding. In the case of microsome and soluble component catalysis, this is probably due to oxidation of the Ru(II) ion prior to complexation. Various inhibitors of electron-transfer proteins were employed in an effort to determine the preferred reducing species. These results lend support to the hypothesis that the antitumor activity of acido ruthenium(III) ammine complexes involves activation by reduction in vivo prior to metal coordination to nucleic acids. Anticancer drugs functioning by this mechanism may be preferentially toxic to or may localize in hypoxic areas of tumors.  相似文献   

12.
DNA is believed to be the primary target for many metal-based drugs. For example, platinum-based anticancer drugs can form specific lesions on DNA that induce apoptosis. New platinum drugs can be designed that have novel modes of interaction with DNA, such as the trinuclear platinum complex BBR3464. Also it is possible to design inert platinum(IV) pro-drugs which are non-toxic in the dark, but lethal when irradiated with certain wavelengths of light. This gives rise to novel DNA lesions which are not as readily repaired as those induced by cisplatin, and provides the basis for a new type of photoactivated chemotherapy. Finally, newly emerging ruthenium(II) organometallic complexes not only bind to DNA coordinatively, but also by H-bonding and hydrophobic interactions triggered by the introduction of extended arene rings into their versatile structures. Intriguingly osmium (the heavier congener of ruthenium) reacts differently with DNA but can also give rise to highly cytotoxic organometallic complexes.  相似文献   

13.
Metal-based anticancer agents occupy a distinct chemical space due to their particular coordination geometry and reactivity. Despite the initial DNA-targeting paradigm for this class of compounds, it is now clear that they can also be tuned to target proteins in cells, depending on the metal and ligand scaffold. Since metallodrug discovery is dominated by phenotypic screenings, tailored proteomics strategies were crucial to identify and validate protein targets of several investigative and clinically advanced metal-based drugs. Here, such experimental approaches are discussed, which showed that metallodrugs based on ruthenium, gold, rhenium and even platinum, can selectively and specifically target proteins with clear-cut down-stream effects. Target identification strategies are expected to support significantly the mechanism-driven clinical translation of metal-based drugs.  相似文献   

14.
15.
Titanium(II) solutions, prepared by dissolving titanium wire in triflic acid + HF, contain equimolar quantities of Ti(IV). Treatment of such solutions with excess Fe(III) or Ru(III) complexes yield Ti(IV), but reactions with Ti(II) in excess give Ti(III). Oxidations by (NH3)5Ru(III) complexes, but not by Fe(III) species, are catalyzed by titanium(IV) and by fluoride. Stoichiometry is unchanged. The observed rate law for the Ru(III)-Ti(II)-Ti(IV) reactions in fluoride media points to competing reaction paths differing by a single F, with both routes involving a Ti(II)-Ti(IV) complex which is activated by deprotonation. It is suggested that coordination of Ti(IV) to TiII(aq) minimizes the mismatch of Jahn-Teller distortions which would be expected to lower the Ti(II,III) self-exchange rate.  相似文献   

16.
Two ruthenium(II) complexes, Λ-[Ru(phen)2(p-HPIP)]2+ and Δ-[Ru(phen)2(p-HPIP)]2+, were synthesized and characterized via proton nuclear magnetic resonance spectroscopy, electrospray ionization-mass spectrometry, and circular dichroism spectroscopy. This study aims to clarify the anticancer effect of metal complexes as novel and potent telomerase inhibitors and cellular nucleus target drug. First, the chiral selectivity of the compounds and their ability to stabilize quadruplex DNA were studied via absorption and emission analyses, circular dichroism spectroscopy, fluorescence-resonance energy transfer melting assay, electrophoretic mobility shift assay, and polymerase chain reaction stop assay. The two chiral compounds selectively induced and stabilized the G-quadruplex of telomeric DNA with or without metal cations. These results provide new insights into the development of chiral anticancer agents for G-quadruplex DNA targeting. Telomerase repeat amplification protocol reveals the higher inhibitory activity of Λ-[Ru(phen)2(p-HPIP)]2+ against telomerase, suggesting that Λ-[Ru(phen)2(p-HPIP)]2+ may be a potential telomerase inhibitor for cancer chemotherapy. MTT assay results show that these chiral complexes have significant antitumor activities in HepG2 cells. More interestingly, cellular uptake and laser-scanning confocal microscopic studies reveal the efficient uptake of Λ-[Ru(phen)2(p-HPIP)]2+ by HepG2 cells. This complex then enters the cytoplasm and tends to accumulate in the nucleus. This nuclear penetration of the ruthenium complexes and their subsequent accumulation are associated with the chirality of the isomers as well as with the subtle environment of the ruthenium complexes. Therefore, the nucleus can be the cellular target of chiral ruthenium complexes for anticancer therapy.  相似文献   

17.
Over the past decade, interest on multitarget anticancer drugs -including heterometallic compounds-has increased considerably. Heterometallic species display improved efficacy and physicochemical properties compared to the individual metallic fragments for a variety of metal pair combinations. By 2018, several compounds had emerged as promising candidates against cisplatin resistant cancers. Here, we summarize research contributions to this topic over the past four years (July 2018–July 2022). In particular, we highlight five articles reporting on the in vivo activity and preliminary mechanisms of action for five groups of compounds. From this selection, we further feature two families of compounds based on Pt(IV)-Gd(III) and Ti(IV)–Au(I) metal combinations, given their potential for clinical translation.  相似文献   

18.
A new series of glucose-conjugated Pt(IV) complexes that target tumor-specific glucose transporters (GLUTs) was designed, synthesized, and evaluated for their anticancer activities. All six compounds, namely, A1-A6, exhibited increased cytotoxicity that were almost six fold higher than that of oxaliplatin to MCF-7 cells. These Pt(IV) complexes can be reduced to release Pt(II) complexes and cause the death of tumor cells. Simultaneously, the glycosylated Pt(IV) complexes (30.21–91.33?μM) showed lower cytotoxicity that normal LO2 cells compared with cisplatin (5.25?μM) and oxaliplatin (8.34?μM). The intervention of phlorizin as a GLUTs inhibitor increased the IC50 value of the glycosylated Pt(IV) complexes, thereby indicating the potential GLUT transportability. The introduction of glucose moiety to Pt(IV) complexes can effectively enhance the Pt cellular uptake and DNA platination. Results suggested glucose-conjugated Pt(IV) complexes had potential for further study as new anticancer agents.  相似文献   

19.
Using bis(3,5-dimethylpyrazol-1-yl)methane as the bidentate N donor ligand L, the yellow compound trans-[RuIIIL2(OMe)2]ClO4 · CH2Cl2 is synthesized. It is a rare example of a mononuclear dialkoxo complex of Ru(III). It shows a quasireversible Ru(II/III) couple at −0.65 V versus NHE in acetonitrile at a Pt electrode. Its magnetic moment at room temperature corresponds to one unpaired electron. It displays a rhombic EPR spectrum in acetone at 77 K with g = 2.219, 2.062 and 1.855.  相似文献   

20.
The oxidation of an amino acid, dl-ornithine monohydrochloride (OMH) by diperiodatoargentate(III) (DPA) was carried out both in the absence and presence of ruthenium(III) catalyst in alkaline medium at 25 °C and a constant ionic strength of 0.10 mol dm−3 spectrophotometrically. The reaction was of first order in both catalyzed and uncatalyzed cases, with respect to [DPA] and was less than unit order in [OMH] and negative fraction in [alkali]. The order with respect to [OMH] changes from first order to zero order as the [OMH] increases. The order with respect to Ru(III) was unity. The uncatalyzed reaction in alkaline medium has been shown to proceed via a DPA-OMH complex, which decomposes in a rate determining step to give the products. Where as in catalyzed reaction, it has been shown to proceed via a Ru(III)-OMH complex, which further reacts with two molecules of DPA in a rate determining step to give the products. The reaction constants involved in the different steps of the mechanisms were calculated for both the reactions. The catalytic constant (KCat.const.) was also calculated for catalyzed reaction at different temperatures. The activation parameters with respect to slow step of the mechanism and also the thermodynamic quantities were determined.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号