Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Biomed Res Int ; 2020: 9514357, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32775454

RESUMO

OBJECTIVE: A DNA intercalating agent reversibly stacks between the adjacent base pairs of DNA and thus is expected to exhibit preferential localization in the tumorous lesions as tumors are associated with enhanced DNA replication. Therefore, radiolabeled DNA intercalators are supposed to have potential to be used in targeted tumor therapy. Working in this direction, an attempt was made to radiolabel 9-aminoacridine, a DNA intercalator, with 177Lu, one of the most useful therapeutic radionuclides, and study the potential of 177Lu-acridine in targeted tumor therapy. Experiments. 9-Aminoacridine was coupled with p-NCS-benzyl-DOTA to facilitate radiolabeling, and the conjugate was radiolabeled with 177Lu. Different reaction parameters were optimized in order to obtain 177Lu-acridine complex with maximum radiochemical purity. In vitro stability of the radiolabeled complex was studied in normal saline and human blood serum. Biological behavior of the radiolabeled agent was studied both in vitro and in vivo using the Raji cell line and fibrosarcoma tumor-bearing Swiss mice, respectively. RESULTS: 177Lu-acridine complex was obtained with ~100% radiochemical purity under the optimized reaction conditions involving incubation of 1.5 mg/mL of ligand with 177Lu (1 mCi, 37 MBq) at 100°C at pH ~5 for 45 minutes. The complex maintained a radiochemical purity of >85% in saline at 6 d and >70% in human serum at 2 d postpreparation. In vitro cellular study showed uptake of the radiotracer (5.3 ± 0.13%) in the Raji cells along with significant cytotoxicity (78.06 ± 2.31% after 6 d). Biodistribution study revealed considerable accumulation of the radiotracer in tumor 9.98 ± 0.13 %ID/g within 1 h postadministration and retention therein till 6 d postadministration 4.00 ± 0.16 %ID/g with encouraging tumor to nontarget organ uptake ratios. CONCLUSIONS: The present study, although preliminary, indicates the potential of 177Lu-acridine and thus radiolabeled DNA intercalators in targeted tumor therapy. However, further detailed evaluation is required to explore the actual potential of such agents in targeted tumor therapy.


Assuntos
Acridinas , Substâncias Intercalantes , Lutécio , Neoplasias , Radioisótopos , Compostos Radiofarmacêuticos , Acridinas/química , Acridinas/farmacocinética , Acridinas/farmacologia , Animais , Linhagem Celular Tumoral , Humanos , Substâncias Intercalantes/química , Substâncias Intercalantes/farmacocinética , Substâncias Intercalantes/farmacologia , Lutécio/química , Lutécio/farmacocinética , Lutécio/farmacologia , Camundongos , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/radioterapia , Radioisótopos/química , Radioisótopos/farmacocinética , Radioisótopos/farmacologia , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacocinética , Compostos Radiofarmacêuticos/farmacologia , Distribuição Tecidual
2.
Anticancer Agents Med Chem ; 20(13): 1538-1547, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32357824

RESUMO

INTRODUCTION: Acridine is a well-known DNA intercalator and thereby gets easily inserted within DNA. As uncontrolled rapid cell division is one of the primary characteristics of the tumors, it is expected that acridine or its suitable derivatives will have preferential accumulation in the tumorous lesions. Therefore, an attempt was made to radiolabel an acridine derivative with 68Ga and study the potential of the 68Ga-acridine complex as a PET agent for tumor imaging. METHODS: 9-aminoacridine was coupled with p-NCS-benzyl-DOTA to render it suitable for labeling with 68Ga. The purified acridine-DOTA conjugate was radiolabeled with 68Ga, eluted from a 68Ge/68Ga radionuclide generator. Various radiolabeling parameters were optimized and the stability of the radiolabeled preparation was studied. The biological behavior of the 68Ga-acridine complex was studied both in vitro and in vivo using Raji cell line and fibrosarcoma tumor bearing Swiss mice, respectively. RESULTS: 68Ga-acridine complex was obtained with ~100% radiochemical purity under the optimized reaction conditions involving incubation of 2mg/mL of ligand at 100°C for 30 minutes. The complex maintained a radiochemical purity of >95% in normal saline and >65% in human blood serum at 3h post-incubation. In vitro cellular study showed (3.2±0.1)% uptake of the radiotracer in the Raji cells. Biodistribution study revealed significant tumor accumulation [(11.41±0.41)% injected activity in per gram] of the radiotracer within 1h postadministration along with uptake in other non-target organs such as, blood, liver, GIT kidney etc. Conclusion: The present study indicates the potential of 68Ga-acridine as a PET agent for imaging of tumorous lesions. However, further detailed evaluation of the agent is warranted to explore its actual potential.


Assuntos
Acridinas/química , Linfoma de Burkitt/diagnóstico por imagem , Fibrossarcoma/diagnóstico por imagem , Substâncias Intercalantes/química , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/química , Acridinas/síntese química , Acridinas/farmacocinética , Animais , Linhagem Celular Tumoral , Radioisótopos de Gálio , Humanos , Substâncias Intercalantes/síntese química , Substâncias Intercalantes/farmacocinética , Camundongos , Estrutura Molecular , Neoplasias Experimentais/diagnóstico por imagem , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/farmacocinética , Distribuição Tecidual
3.
Chemistry ; 24(65): 17239-17254, 2018 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-30222214

RESUMO

After uptake by U87 MG and A375 cancer cells, cobaltabisdicarbollide [COSAN]- distributes between membrane and nucleus and presents no relevant cytotoxicity against both cell lines even for long incubation times. The cytotoxicity of Na[COSAN] was also tested towards one normal cell line, the V79 fibroblasts, in order to ascertain the noncytotoxic profile of the compound. As the cell's nucleus contains DNA, the interaction between [COSAN]- and double-stranded calf thymus DNA (CT-dsDNA) has been investigated. There is a strong interaction between both molecules forming a nanohybrid CT-dsDNA-[COSAN] biomaterial, which was fully characterized. Moreover, Na[COSAN] shows characteristic redox peaks ascribed to the oxidation/reduction of Co3+/2+ at a formal potential of -1.444 V and it can be accumulated at a surface-immobilized DNA layer of glassy carbon electrodes. The equilibrium surface-binding constants (Kox /Kred ), which confirm that [COSAN]- interacts with DNA by an intercalative or electrostatic mode, depending on the ionic strength of the solution, were estimated. In addition, high binding affinity of Na[COSAN] to proteins was observed by 11 B{1 H} NMR and confirmed in vivo. Finally, biodistribution studies of [COSAN]- in normal mice were run. After administration, Na[COSAN] was distributed into many organs but mainly accumulated in the reticuloendothelial system (RES), including liver and spleen. After 1 h, the formation of aggregates by plasma protein interaction plays a role in the biodistribution profile; the aggregates accumulate mostly in the lungs. Na[COSAN], which displays low toxicity and high uptake by relevant cancer cells accumulating boron within the nucleus, could act as a suitable compound for further developments as boron neutron capture therapy (BNCT) agents.


Assuntos
Antineoplásicos/farmacologia , Boranos/farmacologia , DNA/metabolismo , Compostos Organometálicos/farmacologia , Animais , Antineoplásicos/química , Transporte Biológico , Boranos/química , Boranos/farmacocinética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Técnicas Eletroquímicas , Eletrodos , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Humanos , Substâncias Intercalantes/química , Substâncias Intercalantes/farmacocinética , Substâncias Intercalantes/farmacologia , Camundongos Endogâmicos BALB C , Estrutura Molecular , Compostos Organometálicos/química , Compostos Organometálicos/farmacocinética , Oxirredução , Eletricidade Estática , Propriedades de Superfície , Termodinâmica , Distribuição Tecidual
4.
Angew Chem Int Ed Engl ; 56(41): 12628-12633, 2017 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-28834038

RESUMO

Using a new mononuclear "building block," for the first time, a dinuclear RuII (dppn) complex and a heteroleptic system containing both RuII (dppz) and RuII (dppn) moieties are reported. The complexes, including the mixed dppz/dppn system, are 1 O2 sensitizers. However, unlike the homoleptic dppn systems, the mixed dppz/dppn complex also displays a luminescence "switch on" DNA light-switch effect. In both cisplatin sensitive and resistant human ovarian carcinoma lines the dinuclear complexes show enhanced uptake compared to their mononuclear analogue. Thanks to a favorable combination of singlet oxygen generation and cellular uptake properties all three of the new complexes are phototoxic and display potent activity against chemotherapeutically resistant cells.


Assuntos
Complexos de Coordenação/farmacologia , Substâncias Intercalantes/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Fármacos Fotossensibilizantes/farmacologia , Rutênio/farmacologia , Linhagem Celular Tumoral , Complexos de Coordenação/química , Complexos de Coordenação/farmacocinética , DNA/metabolismo , Feminino , Humanos , Substâncias Intercalantes/química , Substâncias Intercalantes/farmacocinética , Neoplasias Ovarianas/metabolismo , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacocinética , Rutênio/química , Rutênio/farmacocinética , Oxigênio Singlete/metabolismo
5.
J Inorg Biochem ; 160: 94-105, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26795497

RESUMO

New rhenium(I) tricarbonyl complexes with the quinolone antimicrobial agents oxolinic acid (Hoxo) and enrofloxacin (Herx) and containing methanol, triphenylphosphine (PPh3) or imidazole (im) as unidentate co-ligands, were synthesized and characterized. The crystal structure of complex [Re(CO)3(oxo)(PPh3)]∙0.5MeOH was determined by X-ray crystallography. The deprotonated quinolone ligands are bound bidentately to rhenium(I) ion through the pyridone oxygen and a carboxylate oxygen. The binding of the rhenium complexes to calf-thymus DNA (CT DNA) was monitored by UV spectroscopy, viscosity measurements and competitive studies with ethidium bromide; intercalation was suggested as the most possible mode and the DNA-binding constants of the complexes were calculated. The rhenium complex [Re(CO)3(erx)(im)] was assayed for its topoisomerase IIα inhibition activity and was found to be active at 100µM concentration. The interaction of the rhenium complexes with human or bovine serum albumin was investigated by fluorescence emission spectroscopy (through the tryptophan quenching) and the corresponding binding constants were determined. The tracer complex [(99m)Tc(CO)3(erx)(im)] was synthesized and identified by comparative HPLC analysis with the rhenium analog. The (99m)Tc complex was found to be stable in solution. Upon injection in healthy mice, fast tissue clearance of the (99m)Tc complex was observed, while both renal and hepatobiliary excretion took place. Preliminary studies in human K-562 erythroleukemia cells showed cellular uptake of the (99m)Tc tracer with distribution primarily in the cytoplasm and the mitochondria and less in the nucleus. These preliminary results indicate that the quinolone (99m)Tc/Re complexes show promise to be further evaluated as imaging or therapeutic agents.


Assuntos
Meios de Contraste/síntese química , Complexos de Coordenação/síntese química , Substâncias Intercalantes/síntese química , Quinolonas/química , Rênio/química , Tecnécio/química , Animais , Antígenos de Neoplasias/química , Sítios de Ligação , Bovinos , Meios de Contraste/farmacocinética , Complexos de Coordenação/farmacocinética , Cristalografia por Raios X , DNA/química , DNA Topoisomerases Tipo II/química , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/química , Enrofloxacina , Etídio/química , Fluoroquinolonas/química , Humanos , Imidazóis/química , Substâncias Intercalantes/farmacocinética , Células K562 , Cinética , Metanol/química , Camundongos , Compostos Organofosforados/química , Ácido Oxolínico/química , Ligação Proteica , Albumina Sérica/química
6.
Spectrochim Acta A Mol Biomol Spectrosc ; 136 Pt C: 1666-70, 2015 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-25459730

RESUMO

The interaction between apigenin and calf thymus deoxyribonucleic acid (ctDNA) in a pH 7.4 Tris-HCl buffer solution was investigated by UV-Vis spectroscopy, fluorescence spectroscopy, DNA melting techniques, and viscosity measurements. It was found that apigenin molecules could intercalate into the base pairs of DNA, forming a apigenin-DNA complex with a binding constant of K310K=6.4×10(4)Lmol(-1). The thermodynamic parameters enthalpy change (ΔH), entropy change (ΔS) and Gibbs free energy (ΔG) were calculated to be 7.36×10(4)Jmol(-1), 329JK(-1)mol(-1) and -2.84×10(4)Jmol(-1) at 310K, respectively. Hydrophobic interaction was the predominant intermolecular force in stabilizing the apigenin-DNA complex. Thermal denaturation study suggested that the stabilization of the ctDNA helix was increased when the apigenin binding to ctDNA as indicated by the increase in thermal denaturation temperature of ctDNA at around 5.0°C in the presence of apigenin. Spectroscopic techniques together with melting techniques and viscosity determination provided evidences of intercalation mode of binding for the interaction between apigenin and ctDNA.


Assuntos
Apigenina/farmacocinética , DNA/metabolismo , Apigenina/química , Sítios de Ligação , Dicroísmo Circular , DNA/química , DNA/efeitos dos fármacos , Entropia , Substâncias Intercalantes/química , Substâncias Intercalantes/farmacocinética , Desnaturação de Ácido Nucleico , Espectrometria de Fluorescência , Espectrofotometria Ultravioleta , Temperatura , Termodinâmica , Viscosidade
7.
J Drug Target ; 19(9): 846-55, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21692679

RESUMO

BACKGROUND: Increasing attention is currently focussed on the issue of finding strategies for the delivery of Auger-electron-emitting radionuclides into tumor cell nuclei. PURPOSE: In this study, we investigated tumor-cell uptake and cell-killing ability in vitro as well as in vivo biodistribution of an (125)I-labelled anthracycline derivative administered by means of HER2-targeted liposomes. METHODS: Anthracycline derivative Comp1 was radiolabelled with Auger-emitting (125)I and encapsulated in liposomes (DSPC:Chol:DSPE-PEG) using pH-gradient loading. Single-chain fragment F5 was anchored to the liposomes as targeting device for HER2. Uptake and specificity of (125)I-Comp1 delivered via targeting and non-targeting liposomes were analysed in cultured HER2-overexpressing cells. Cell-killing efficacy was evaluated in SKOV3 cells and biodistribution for up to 48 h was studied after intraperitoneal injection in tumor-bearing female BALB/c nu/nu mice. RESULTS: (125)I-Comp1 was specifically taken up by the cultured cells when administered by means of HER2-targeted liposomes and a clear dose-effect correlation in survival of cells was seen with increasing specific activity. The biodistribution studies revealed that (125)I-Comp1 accumulated in tumors when distributed using HER2-targeted liposomes and that this effect was absent when using non-targeting liposomes. CONCLUSION: The HER2-targeted liposomes possess the properties needed to bring about tumor-specific delivery and therapeutic effect of (125)I-Comp1.


Assuntos
Daunorrubicina/análogos & derivados , Sistemas de Liberação de Medicamentos , Substâncias Intercalantes/administração & dosagem , Neoplasias Ovarianas/tratamento farmacológico , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Daunorrubicina/administração & dosagem , Daunorrubicina/farmacocinética , Relação Dose-Resposta a Droga , Feminino , Humanos , Injeções Intraperitoneais , Substâncias Intercalantes/farmacocinética , Radioisótopos do Iodo , Lipossomos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/patologia , Receptor ErbB-2/metabolismo , Distribuição Tecidual
8.
Invest New Drugs ; 29(6): 1253-63, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20567996

RESUMO

The increasing incidence of melanoma and the lack of effective therapy on the disseminated form have led to an urgent need for new specific therapies. Several iodobenzamides or analogs are known to possess specific affinity for melanoma tissue. New heteroaromatic derivatives have been designed with a cytotoxic moiety and termed DNA intercalating agents. These compounds could be applied in targeted radionuclide therapy using (125)I, which emits Auger electrons and gives high-energy, localized irradiation. Two iodinated acridine derivatives have been reported to present an in vivo kinetic profile conducive to application in targeted radionuclide therapy. The aim of the present study was to perform a preclinical evaluation of these compounds. The DNA intercalating property was confirmed for both compounds. After radiolabeling with (125)I, the two compounds induced in vitro a significant radiotoxicity to B16F0 melanoma cells. Nevertheless, the acridine compound appeared more radiotoxic than the acridone compound. While cellular uptake was similar for both compounds, SIMS analysis and in vitro protocol showed a stronger affinity for melanin with acridone derivative, which was able to induce a predominant scavenging process in the melanosome and restrict access to the nucleus. In conclusion, the acridine derivative with a higher nuclear localization appeared a better candidate for application in targeted radionuclide therapy using (125)I.


Assuntos
Acridinas/farmacologia , Substâncias Intercalantes/farmacologia , Radioisótopos do Iodo/administração & dosagem , Melanoma Experimental/radioterapia , Acridinas/química , Acridinas/farmacocinética , Animais , Núcleo Celular/metabolismo , Elétrons , Substâncias Intercalantes/química , Substâncias Intercalantes/farmacocinética , Melaninas/metabolismo , Melanossomas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
9.
Med Chem ; 5(6): 549-57, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20041834

RESUMO

Cancer is one of the life threatening diseases and the development of novel anticancer molecules is limited by many reasons. In the present investigation, some novel benzo[a]phenazine-5-sulfonic acid derivatives as DNA intercalator was designed with optimized pharmacokinetic features for cancer treatment. The compounds with desired pharmacokinetic profile were synthesized and structurally characterized. Cytotoxic activity study against HL-60 tumor cell lines shows that 10-dimethyl carboxamido derivative of benzo[a]phenazine-5-sulfonic acid is found to be the most active in the series with cytotoxic activity (IC(50) = 19 microM) comparable to cisplatin (IC(50) = 7 microM). The study concluded that the novel benzo[a]phenazine-5-sulfonic acid derivatives were found to have enhanced DNA binding affinity and exhibited significant activity in vitro against HL-60 cell lines. This work will also guide for further development of effective DNA intercalators for cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/farmacocinética , Biologia Computacional , Ácidos Sulfônicos/farmacocinética , Ácidos Sulfônicos/toxicidade , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Bovinos , DNA/química , DNA/metabolismo , Células HL-60 , Humanos , Substâncias Intercalantes/síntese química , Substâncias Intercalantes/química , Substâncias Intercalantes/farmacocinética , Substâncias Intercalantes/toxicidade , Desnaturação de Ácido Nucleico/efeitos dos fármacos , Relação Estrutura-Atividade , Ácidos Sulfônicos/síntese química , Ácidos Sulfônicos/química
10.
Artigo em Inglês | MEDLINE | ID: mdl-18722155

RESUMO

A systematic comparative study of the binding of antitumor Morin and its complexes with DNA has been investigated in the Britton-Robison (BR) buffer solutions using voltammetric and spectroscopic methods. The results show that Morin molecule, acting as an intercalator, is inserted into the cavity of the beta-cyclodextrin (beta-CD) as well as into the base stacking domain of the DNA double helix. The interaction of Morin-Cu complex or the inclusion complex of Morin-beta-CD with ds-DNA causes hypochromism in the absorption spectra, along with pronounced changes in the electrochemical behavior of the Morin complexes. An isobestic point and a new spectrum band appeared indicating the formation of the new system of Morin-Cu-DNA at lambda(m)=391 nm and Morin-beta-CD-DNA at lambda(m)=375 nm. The intercalation of Morin-Cu and Morin-beta-CD complexes with DNA produces an electrochemically inactive supramolecular complex. The binding constants were calculated from the increase of the solubility, the strong hypochromism, and the decrease in peak current of Morin and its complexes upon the addition of the host molecules. Calculation of the thermodynamic parameters of the interaction of the inclusion complex of Morin-beta-CD with DNA, including Gibbs free energy change, Helmholz free energy and entropy change shows that the complexation is a spontaneous process of association.


Assuntos
Cobre/farmacocinética , DNA/metabolismo , Flavonoides/farmacocinética , beta-Ciclodextrinas/farmacocinética , Antineoplásicos/química , Antineoplásicos/farmacocinética , Cobre/química , DNA/efeitos dos fármacos , Eletroquímica/métodos , Flavonoides/química , Substâncias Intercalantes/química , Substâncias Intercalantes/farmacocinética , Modelos Biológicos , Espectrometria de Fluorescência , beta-Ciclodextrinas/química
11.
J Am Chem Soc ; 130(23): 7182-3, 2008 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-18489094

RESUMO

Here we report on a phenanthridine derivative which has a covalently linked fluorescein molecule in order to increase the light absorption and hence fluorescence signal intensity when bound to duplex RNA. Steady-state fluorescence shows that the energy transfer efficiency from the fluorescein to the phenanthridine fluorophore is approximately 77%, which results in the probe being over 5x brighter than other phenanthridine derivatives when bound to RNA. Due to the relatively long lifetime (approximately 20 ns) of the probe, time-resolved fluorescence is used to increase the signal to background ratio in cell growth medium from 7 (steady-state value) to over 40. Moreover, fluorescence images of cells containing the probe show that the fluorescein signal is readily apparent along with that of the intercalated fluorophore, allowing this probe to be used as a dual color probe which simultaneously reports the probes' location and that of RNA.


Assuntos
Fluoresceínas/química , Corantes Fluorescentes/química , Substâncias Intercalantes/química , Fenantridinas/química , RNA/análise , Animais , Neoplasias da Mama/química , Neoplasias da Mama/metabolismo , Fluoresceínas/farmacocinética , Transferência Ressonante de Energia de Fluorescência/métodos , Corantes Fluorescentes/farmacocinética , Humanos , Substâncias Intercalantes/farmacocinética , Neoplasias Mamárias Experimentais/química , Neoplasias Mamárias Experimentais/metabolismo , Microscopia de Fluorescência , Modelos Moleculares , Fenantridinas/farmacocinética , RNA/química , RNA Fúngico/análise , Espectrometria de Fluorescência/métodos , Leveduras/química , Leveduras/genética
12.
DNA Cell Biol ; 27(2): 81-90, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17924822

RESUMO

There is compelling evidence that cellular DNA is the target of many anticancer agents. Consequently, elucidation of the molecular nature governing the interaction of small molecules to DNA is paramount to the progression of rational drug design strategies. In this study, we have compared the binding and thermodynamic aspects of two known DNA-binding agents, quinacrine (QNA) and methylene blue (MB), with calf thymus (CT) DNA. The study revealed noncooperative binding phenomena for both the drugs to DNA with an affinity one order higher for QNA compared to MB as observed from diverse techniques, but both bindings obeyed neighbor exclusion principle. The data of the salt dependence of QNA and MB from the plot of log K versus log [Na+] revealed a slope of 1.06 and 0.93 consistent with the values predicted by theories for the binding of monovalent cations, and have been analyzed for contributions from polyelectrolytic and nonpolyelectrolytic forces. The binding of both drugs was further characterized by strong stabilization of DNA against thermal strand separation in both optical melting and differential scanning calorimetry studies. The binding data analyzed from the thermal denaturation and from isothermal titration calorimetry (ITC) were in close proximity to those obtained from spectral titration data. ITC results revealed the binding to be exothermic and favored by both negative enthalpy and positive entropy changes. The heat capacity changes obtained from temperature dependence of enthalpy indicated -146 and -78 cal/(mol.K), respectively, for the binding of QNA and MB to CT DNA. Circular dichroism study further characterized the structural changes on DNA upon intercalation of these molecules. Molecular aspects of interaction of these molecules to DNA are discussed.


Assuntos
DNA/metabolismo , Substâncias Intercalantes/farmacocinética , Azul de Metileno/farmacocinética , Quinacrina/farmacocinética , Ligação Competitiva , Dicroísmo Circular , DNA/efeitos dos fármacos , Temperatura Alta , Modelos Biológicos , Desnaturação de Ácido Nucleico/efeitos dos fármacos , Termodinâmica , Titulometria
13.
DNA Cell Biol ; 27(2): 101-7, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17970617

RESUMO

Platinum(II)-based anticancer drugs are effective for the management and treatment of several types of cancer. Cis-diamminedichloroplatinum(II) (cisplatin) exerts its antitumor activity by binding to DNA via intrastrand cross-links to d(GpG) (dG = deoxyguanosine) and to d(ApG) (dA = deoxyadenosine), causing DNA bending and interfering with DNA replication and transcription. However, the exact binding modes of other platinum(II)-based antitumor drugs to DNA duplex and their mechanism of action have not been clearly investigated. The aim of this study was to examine the binding of a novel anticancer estradiol-platinum(II) hybrid molecule (CD-37) with calf-thymus DNA in vitro and to compare the results with those obtained with cisplatin drug. Solutions containing various CD-37 or cisplatin concentrations were reacted with DNA at physiological pH. Then, using Fourier transform infrared, ultraviolet-visible, and circular dichroism spectroscopic methods, it was possible to characterize the drug binding mode, the binding constant, and structural variations of DNA in aqueous solution. Spectroscopic evidence showed that cisplatin binds to guanine N7 site with minor perturbations of the backbone phosphate group with an overall binding constant of K(cisPt) = 5.73 (+/- 0.45) x 10(4) M(-1). CD-37 binds to DNA duplex via H-bonding network at low drug concentrations with minor perturbations of guanine N7 site at high drug content and with a binding constant of K(CD-37) = 1.0 (+/- 0.15) x 10(4) M(-1). DNA aggregation occurs at high drug concentration, while DNA remains in the B-family structure.


Assuntos
Cisplatino/farmacocinética , DNA/metabolismo , Estradiol/análogos & derivados , Estradiol/farmacocinética , Compostos Organoplatínicos/farmacocinética , Antineoplásicos/química , Antineoplásicos/farmacocinética , Dicroísmo Circular , DNA/química , DNA/efeitos dos fármacos , Estradiol/química , Substâncias Intercalantes/química , Substâncias Intercalantes/farmacocinética , Modelos Biológicos , Conformação de Ácido Nucleico/efeitos dos fármacos , Compostos Organoplatínicos/química , Espectroscopia de Infravermelho com Transformada de Fourier
14.
Acta Biochim Pol ; 54(2): 297-306, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17446935

RESUMO

Triazoloacridinones (TA) are a new group of potent antitumor compounds, from which the most active derivative, C-1305, has been selected for extended preclinical trials. This study investigated the mechanism of TA binding to DNA. Initially, for selected six TA derivatives differing in chemical structures as well as cytotoxicity and antitumor activity, the capability of noncovalent DNA binding was analyzed. We showed that all triazoloacridinones studied stabilized the DNA duplex at a low-concentration buffer but not at a salt concentration corresponding to that in cells. DNA viscometric studies suggested that intercalation to DNA did not play a major role in the mechanism of the cytotoxic action of TA. Studies involving cultured cells revealed that triazoloacridinone C-1305 after previous metabolic activation induced the formation of interstrand crosslinks in DNA of some tumor and fibroblast cells in a dose dependent manner. However, the detection of crosslink formation was possible only when the activity of topoisomerase II in cells was lowered. Furthermore, it was impossible to validate the relevance of the ability to crosslink DNA to biological activity of TA derivatives.


Assuntos
Acridonas/farmacologia , Antineoplásicos/farmacologia , DNA/efeitos dos fármacos , Acridinas/metabolismo , Acridinas/farmacocinética , Acridinas/farmacologia , Acridonas/metabolismo , Acridonas/farmacocinética , Animais , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Biotransformação , Reagentes de Ligações Cruzadas/metabolismo , Reagentes de Ligações Cruzadas/farmacocinética , Reagentes de Ligações Cruzadas/farmacologia , DNA/química , DNA/metabolismo , DNA de Neoplasias/química , DNA de Neoplasias/efeitos dos fármacos , DNA de Neoplasias/metabolismo , Células HL-60 , Células HeLa , Humanos , Substâncias Intercalantes/metabolismo , Substâncias Intercalantes/farmacocinética , Substâncias Intercalantes/farmacologia , Camundongos , Células NIH 3T3 , Conformação de Ácido Nucleico/efeitos dos fármacos , Triazóis/metabolismo , Triazóis/farmacocinética , Triazóis/farmacologia
15.
J Struct Biol ; 155(1): 38-44, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16630726

RESUMO

SRIXE mapping has been used to gain insight into the fate of platinum(II) and platinum(IV) complexes in cells and tumours treated with anticancer active complexes to facilitate the development of improved drugs. SRIXE maps were collected of thin sections of human ovarian (A2780) cancer cells treated with bromine containing platinum complexes, cis-[PtCl(2)(3-Brpyr)(NH(3))] (3-Brpyr=3-bromopyridine) and cis,trans,cis-[PtCl(2)(OAcBr)(2)(NH(3))(2)] (OAcBr=bromoacetate), or a platinum complex with an intercalator attached cis-[PtCl(2)(2-[(3-aminopropyl)amino]-9,10-anthracenedione)(NH(3))]. After 24h the complexes appear to be localised in the cell nucleus with a lower concentration in the surrounding cytoplasm. In cells treated with cis-[PtCl(2)(3-Brpyr)(NH(3))] the concentration of bromine was substantially higher than in control cells and the bromine was co-localised with the platinum consistent with the 3-bromopyridine ligand remaining bound to the platinum. The cells treated with cis,trans,cis-[PtCl(2)(OAcBr)(2)(NH(3))(2)] also showed an increased level of bromine, but to a much lesser extent than for those treated with cis-[PtCl(2)(3-Brpyr)(NH(3))] suggestive of substantial reduction of the platinum(IV) complex. Maps were also collected from thin sections of a 4T1.2 neo 1 mammary tumour xenograft removed from a mouse 3h after treatment with cis,trans,cis-[PtCl(2)(OH)(2)(NH(3))(2)] and revealed selective uptake of platinum by one cell.


Assuntos
Antineoplásicos/farmacocinética , Microanálise por Sonda Eletrônica/métodos , Neoplasias/metabolismo , Compostos Organoplatínicos/farmacocinética , Animais , Compostos de Bromo/análise , Feminino , Humanos , Substâncias Intercalantes/farmacocinética , Neoplasias Mamárias Animais/diagnóstico por imagem , Neoplasias Mamárias Animais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/diagnóstico por imagem , Neoplasias Ovarianas/diagnóstico por imagem , Neoplasias Ovarianas/metabolismo , Radiografia , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Bioconjug Chem ; 16(3): 582-7, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15898725

RESUMO

A trifunctional bioconjugate consisting of the SV40 nuclear localization signal (NLS) peptide, an aliphatic triamine ligand, and the DNA intercalating pyrene has been synthesized and quantitatively labeled with [(99m)Tc(OH(2))(3)(CO)(3)](+). The radiotoxicity of the resulting nucleus-targeting radiopharmaceutical on B16F1 mouse melanoma cells has been investigated to evaluate the activity of Auger and Coster-Kronig electrons on the viability of cells. We found a dose-dependent significant radiotoxicity of the nucleus-targeting radiopharmaceutical clearly related to the low energy decay of (99m)Tc. These principal results imply a possible therapeutic strategy based on the use of the low-energy Auger electron-emitting (99m)Tc radionuclide attached to nucleus-targeting molecules and comprising an intercalator. Highly efficient DNA targeting vectors could complement the usual role of (99m)Tc in diagnostic applications. The Auger electrons emitted by the (99m)Tc nuclide induce DNA damage leading ultimately, through a mitotic catastrophe pathway, to necrotic cell death. Non-DNA-targeting (99m)Tc complexes display much lower radiotoxicity.


Assuntos
Substâncias Intercalantes/farmacocinética , Substâncias Intercalantes/toxicidade , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/farmacocinética , Compostos de Organotecnécio/toxicidade , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Relação Dose-Resposta à Radiação , Substâncias Intercalantes/química , Camundongos , Estrutura Molecular , Compostos de Organotecnécio/química , Compostos de Organotecnécio/farmacocinética
17.
J Phys Chem B ; 109(44): 21140-4, 2005 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-16853738

RESUMO

The semirigid binuclear ruthenium complex Delta,Delta-[mu-(11,11'-bidppz)(phen)(4)Ru(2)](4+) has been shown to rearrange slowly from an initial groove-bound nonluminescent state to a final intercalated emissive state by threading one of its bulky Ru(phen)(2) moieties through the DNA base stack. When this complex binds to poly[d(A-T)(2)], a further increase in emission from the complex is observed after completion of the intercalation, assigned to reorganization of the intercalated complex. We here report a study of the threading process in poly[d(A-T)(2)], in which the minor groove binding dye DAPI is used as an energy transfer probe molecule to assess the distribution of ruthenium complex during and also after the actual threading phase. The emission from DAPI is found to change with the same rate as the emission from the ruthenium complex, and furthermore, DAPI does not disturb the binding kinetics of the latter, justifying it as a good probe of both the threading and the reorganization processes. We conclude from the change in the emission from both DAPI and the ruthenium complex with time that DAPI-ruthenium interactions are most pronounced during the process of threading of the complex, suggesting that the complexes are initially threaded slightly anticooperatively and thereafter redistribute along the DNA to reach their thermodynamically most favorable distribution. The final distribution is characterized by a small but significant binding cooperativity, probably as a result of hydrophobic interactions between the complex ions despite their tetravalent positive charges. The mechanism of "shuffling" the complex along the DNA chain is discussed, i.e., whether the ruthenium complex remains threaded (requiring sequential base-pair openings) or if unthreading followed by lateral diffusion within the ionic atmosphere of the DNA and rethreading occurs.


Assuntos
DNA/metabolismo , Substâncias Intercalantes/química , Rutênio/química , Difusão , Transferência de Energia , Interações Hidrofóbicas e Hidrofílicas , Indóis , Substâncias Intercalantes/farmacocinética , Cinética , Compostos Organometálicos , Rutênio/farmacocinética
18.
J Pharmacol Exp Ther ; 297(3): 1088-98, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11356933

RESUMO

There is considerable evidence that DNA intercalating drugs fail to penetrate tumor tissue efficiently. This study used the multicellular layer (MCL) experimental model, in conjunction with computational modeling, to test the hypothesis that a DNA intercalator in phase II clinical trial, N-[2-(dimethylamino)-ethyl]acridine-4-carboxamide (DACA), has favorable extravascular transport properties. Single cell uptake and metabolism of DACA and the related but more basic aminoacridine 9-[3-(dimethylamino)propylamino]acridine (DAPA), and penetration through V79 and EMT6 MCL, were investigated by high-performance liquid chromatography. DACA was accumulated by cells to a lesser extent than DAPA and was metabolized to the previously unreported acridan by V79 but not EMT6 cells. Despite this metabolism, flux of DACA through MCL was much faster than that of DAPA. Modeling MCL transport as diffusion with reaction (metabolism and reversible binding) showed that the faster flux of DACA was due to a 3-fold higher free drug diffusion coefficient and 10-fold lower binding site density. The MCL transport parameters were used to develop a spatially resolved pharmacokinetic model for the extravascular compartment in tumors, which provided a reasonable prediction of measured average tumor concentrations from plasma pharmacokinetics in mice. Area under the curve was essentially independent of distance from blood vessels, although the combined pharmacokinetic/pharmacodynamic model predicted a modest decrease in cytotoxicity (from 1.8 to 1.1 logs of cell kill) across a 125-microm region. In conclusion, this study demonstrates that it is possible to design DNA intercalators that diffuse efficiently in tumor tissue, and that there is little impediment to DACA transport over distances required for its antitumor action.


Assuntos
Acridinas/farmacocinética , Antineoplásicos/farmacocinética , Substâncias Intercalantes/farmacocinética , Neoplasias Mamárias Experimentais/metabolismo , Modelos Biológicos , Acridinas/química , Acridinas/metabolismo , Animais , Área Sob a Curva , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Compartimentos de Líquidos Corporais , Radioisótopos de Carbono , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Simulação por Computador , Cricetinae , Difusão , Cultura em Câmaras de Difusão , Desenho de Fármacos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Camundongos , Nitracrina/análogos & derivados , Nitracrina/farmacocinética , Inibidores da Topoisomerase II , Ureia/metabolismo
19.
Cancer Res ; 61(7): 2935-44, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11306471

RESUMO

The tricyclic carboxamide N-[2-(dimethylamino)ethyl]acridine-4-carboxamide (DACA) is a DNA-intercalating agent capable of inhibiting both topoisomerases I and II and is currently in Phase II clinical trial. Many related analogues have been developed, but despite their potent in vitro cytotoxicities, they exhibit poor extravascular distribution. As part of an ongoing drug development program to obtain related "minimal intercalators" with lower DNA association constants, we have compared the biodistribution and metabolite profiles of the prototype compound, DACA, with three analogues to aid rational drug selection. All of these compounds share a common structural feature, N-dimethyl side chain, which was radiolabeled with the positron-emitting radioisotope, carbon-11. This strategy was selected because it allows promising candidates emerging from preclinical studies in animals to be evaluated rapidly in humans using positron emission tomography (PET). The acridine DACA, the phenazine SN 23490, the pyridoquinoline SN 23719, and the dibenzodioxin SN 23935 were found to be cytotoxic in in vitro assays with an IC50 of 1.4-1.8 microM, 0.4-0.6 microM, 1.3-1.6 microM, and 24-36 microM, respectively, in HT29, U87MG, and A375M cell lines. Ex vivo biodistribution studies with carbon-11 radiolabeled compounds in mice bearing human tumor xenografts showed rapid clearance of 11C-radioactivity (parent drug and metabolites) from blood and the major organs. Rapid hepatobiliary clearance and renal excretion were also observed. There was low [<5% of injected dose/gram (%ID/g)] and variable uptake of 11C-radioactivity in three tumor types for all of the compounds. Tumor (U87MG) to blood 11C-radioactivity for [11C]DACA, [11C](9-methoxyphenazine-1-carboxamide (SN 23490), [11C]2-(4-pyridyl)quinoline-8-carboxamide (SN 23719), and [11C]dibenzo[1,4]dioxin-1-carboxamide (SN 23935) at 30 min were 2.9 +/- 1.1, 2.3 +/- 0.6, 2.6 +/- 0.6, and 0.7 +/- 0.2, respectively. For SN 23719, the distribution of 11C-radioactivity in normal tissues and tumors determined ex vivo was in broad agreement with that determined in vivo by whole body PET scanning. [11C]DACA was rapidly and extensively metabolized to several plasma metabolites and a major tumor metabolite. In contrast, [11C]SN 23935, [11C]SN 23490, and [11C]SN 23719 showed less extensive metabolism. In the tumor samples, the parent [11C]DACA and [11C]SN 23935 represented between 0.3 and 1.5%ID/g, whereas [11C]SN 23490 and [11C]SN 23719 represented between 1.5 and 2.8%ID/g. In conclusion, by using a strategy with 11C-labeling, we have determined the tissue distribution and metabolic stability of novel tricyclic carboxamides with the view of selecting analogues with potentially better in vivo activity against solid tumors. SN 23490 and SN 23719 had more favorable distribution and metabolic stability compared with DACA and SN 23935 and may warrant further development. The radiolabeling strategy used allows ex vivo and in vivo evaluation of promising anticancer agents in animals and offers the potential of rapid translation to studies in humans using PET.


Assuntos
Acridinas/farmacocinética , Antineoplásicos/farmacocinética , Substâncias Intercalantes/farmacocinética , Acridinas/química , Acridinas/metabolismo , Acridinas/toxicidade , Animais , Antineoplásicos/metabolismo , Antineoplásicos/toxicidade , Radioisótopos de Carbono , Feminino , Glioma/tratamento farmacológico , Glioma/metabolismo , Células HT29 , Humanos , Concentração Inibidora 50 , Substâncias Intercalantes/metabolismo , Substâncias Intercalantes/toxicidade , Marcação por Isótopo , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Camundongos Nus , Distribuição Tecidual , Tomografia Computadorizada de Emissão , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
20.
J Clin Oncol ; 19(3): 857-69, 2001 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11157040

RESUMO

PURPOSE: To determine the maximum-tolerated dose and characterize the pharmacokinetic behavior of LU79553, a novel bisnaphthalimide antineoplastic agent, when administered as a daily intravenous infusion for 5 days every 3 weeks. PATIENTS AND METHODS: Patients with advanced solid malignancies received escalating doses of LU79553. Plasma sampling and urine collections were performed on both days 1 and 5 of the first course. RESULTS: Thirty patients received 105 courses of LU79553 at doses ranging from 2 to 24 mg/m(2)/d. Proximal myopathy, erectile dysfunction, and myelosuppression precluded the administration of multiple courses at doses above 18 mg/m(2)/d. These toxicities were intolerable in two of six patients after receiving three courses at the 24-mg/m(2)/d dose level. At the 18-mg/m(2)/d dose, one of six patients developed febrile neutropenia and grade 2 proximal myopathy after three courses of LU79553. The results of electrophysiologic, histopathologic, and ultrastructural studies supported a drug-induced primary myopathic process. A patient with a platinum- and taxane-resistant papillary serous carcinoma of the peritoneum experienced a partial response lasting 22 months. Pharmacokinetics were dose-independent, optimally described by a three-compartment model, and there was modest drug accumulation over the 5 days of treatment. CONCLUSION: Although no dose-limiting events were noted in the first two courses of LU79553, cumulative muscular toxicity precluded repetitive treatment with LU79553 at doses above 18 mg/m(2)/d, which is the recommended dose for subsequent disease-directed evaluations. The preliminary antitumor activity noted is encouraging, but the qualitative and cumulative nature of the principal toxicities, as well as the relatively small number of patients treated repetitively, mandate that rigorous and long-term toxicologic monitoring be performed in subsequent evaluations of this unique agent.


Assuntos
Amidas/efeitos adversos , Amidas/farmacocinética , Substâncias Intercalantes/efeitos adversos , Substâncias Intercalantes/farmacocinética , Isoquinolinas/efeitos adversos , Isoquinolinas/farmacocinética , Neoplasias/metabolismo , Adulto , Idoso , Amidas/uso terapêutico , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Humanos , Infusões Intravenosas , Substâncias Intercalantes/uso terapêutico , Isoquinolinas/uso terapêutico , Masculino , Pessoa de Meia-Idade , Doenças Musculares/induzido quimicamente , Neoplasias/tratamento farmacológico , Neutropenia/induzido quimicamente , Trombocitopenia/induzido quimicamente
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA