Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Acta Chim Slov ; 68(4): 970-982, 2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34918770

RESUMO

Effective treatment of glioma still stands as a challenge in medical science. The work aims for the fabrication and evaluation of lipid based nanostructures for improved delivery of lomustine to brain tumor cells. Experimental formulations (LNLs) were developed by modified lipid layer hydration technique and evaluated for different in vitro characteristics like particle size analysis, surface charge, surface morphology, internal structure, in vitro drug loading, drug release profile etc. Anticancer potential of selected LNLs was tested in vitro on C6 glioma cell line. Electron microscopic study depicted a size of less than 50 nm for the selected LNLs along 8.8% drug loading with a sustained drug release tendency over 48 h study period. Confocal microscopy revealed extensive internalization of the selected LNL in C6 cells. LNLs were found more cytotoxic than free drug and blank nanocarriers as depicted from MTT assay. The selected LNL showed improved pharmacokinetic profile both in blood and brain in the experimental mice models along with negligible hemolysis in mice blood cells. Further studies are warranted for the future translation of LNLs at clinics.


Assuntos
Antineoplásicos/química , Lipídeos/química , Lomustina/química , Nanoestruturas/química , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Liberação Controlada de Fármacos , Feminino , Glioma/metabolismo , Glioma/patologia , Meia-Vida , Hemólise/efeitos dos fármacos , Cinética , Masculino , Camundongos , Nanoestruturas/toxicidade
2.
Artigo em Inglês | MEDLINE | ID: mdl-32013840

RESUMO

AIMS: The present research work discussed the preparation of lomustine loaded with chitosan nanoparticles (LNCp) by ionic gelation method with homogenization using the design on experiments by Box-Behnken design. METHODS: The nanoparticles are evaluated by particle size, zeta potential, surface morphology, drug content, entrapment efficiency and in-vitro drug release. RESULTS: The FT-IR results support that drug have no interaction with excipients, which are used in the preparation of nanoparticle. The particle size, drug content and encapsulation efficiency of the developed nanoparticles ranged from 190 to 255 nm, 80.88% to 94.02%, and 77.12 to 88.74%, respectively. The drug release rate is diffusion-controlled over 8 hours. The F-value for all of the responses shows that the models are significant. The p-value, less than 0.05 for all the responses reveals the significance of the models. Graphical optimisation is done by desirability plot and overlay plot, which contains optimal values of independent variables with the desirability of 1. CONCLUSION: In conclusion, the results suggested that the optimised lomustine loaded chitosan nanoparticles are useful for brain targeting hence hold the potential for further research and clinical application.


Assuntos
Antineoplásicos Alquilantes/química , Quitosana/química , Lomustina/química , Nanopartículas/química , Encéfalo , Desenho de Fármacos , Liberação Controlada de Fármacos , Microscopia Eletrônica de Transmissão , Nanopartículas/ultraestrutura , Tamanho da Partícula
3.
J Pharm Biomed Anal ; 176: 112786, 2019 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-31398506

RESUMO

Electrochemical techniques were used to investigate the behavior of lomustine (CCNU) and its degradation in aqueous solution at a glassy carbon electrode (GCE). The in situ interaction of CCNU and chemically degraded CCNU (cdCCNU) with dsDNA was then investigated in dsDNA incubated solutions, using dsDNA electrochemical biosensors and comet assays. CCNU undergoes electrochemical reduction in two irreversible, diffusion-controlled, and pH-dependent redox processes, each with transfer of two electrons and one proton. At pH ≥ 10.1, the peak potential for the two processes was essentially pH-independent and involved only one electron. A mechanism was proposed for the reduction of CCNU in a neutral medium. In addition, it was found that CCNU underwent spontaneous degradation during incubation in aqueous solution, without the formation of electroactive degradation products. The degradation process was faster in basic media. Moreover, this pro-drug interacted with the DNA. Its metabolite(s) initially caused condensation of the double helix chains, followed by the unwinding of these chains. In addition, free guanine (Gua) was released from the dsDNA and oxidative damage to the DNA by the CCNU metabolite(s) was evidenced from the detection of 8-oxoGua and 2,8-oxoAde. These results were confirmed by the poly(dA)- and poly(dG)-polyhomonucleotide biosensors, which revealed the oxidative damage caused to both bases (guanine and adenine) of the dsDNA by the CCNU metabolite(s). The comet assay indicated breaks in the single strand DNA, complementing the results of the studies using differential pulse voltammetry. Conformational changes of dsDNA caused by CCNU and cdCCNU were confirmed using comet assays.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Quebras de DNA de Cadeia Simples/efeitos dos fármacos , DNA/efeitos dos fármacos , Lomustina/farmacologia , Antineoplásicos Alquilantes/química , Técnicas Biossensoriais , DNA/química , Difusão , Estabilidade de Medicamentos , Técnicas Eletroquímicas , Eletrodos , Lomustina/química , Conformação de Ácido Nucleico/efeitos dos fármacos , Água
4.
ACS Nano ; 13(2): 1365-1384, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30721028

RESUMO

Glioblastoma multiforme (GBM) is an aggressive primary brain tumor, for which there is no cure. Treatment effectiveness for GBM has been limited due to tumor heterogeneity, an immunosuppressive tumor microenvironment (TME), and the presence of the blood-brain barrier, which hampers the transport of chemotherapeutic compounds to the central nervous system (CNS). High-density lipoprotein (HDL)-mimicking nanodiscs hold considerable promise to achieve delivery of bioactive compounds into tumors. Herein, we tested the ability of synthetic HDL nanodiscs to deliver chemotherapeutic agents to the GBM microenvironment and elicit tumor regression. To this end, we developed chemo-immunotherapy delivery vehicles based on sHDL nanodiscs loaded with CpG, a Toll-like receptor 9 (TLR9) agonist, together with docetaxel (DTX), a chemotherapeutic agent, for targeting GBM. Our data show that delivery of DTX-sHDL-CpG nanodiscs into the tumor mass elicited tumor regression and antitumor CD8+ T cell responses in the brain TME. We did not observe any overt off-target side effects. Furthermore, the combination of DTX-sHDL-CpG treatment with radiation (IR), which is the standard of care for GBM, resulted in tumor regression and long-term survival in 80% of GBM-bearing animals. Mice remained tumor-free upon tumor cell rechallenge in the contralateral hemisphere, indicating the development of anti-GBM immunological memory. Collectively, these data indicate that sHDL nanodiscs constitute an effective drug delivery platform for the treatment of GBM, resulting in tumor regression, long-term survival, and immunological memory when used in combination with IR. The proposed delivery platform has significant potential for clinical translation.


Assuntos
Glioblastoma/tratamento farmacológico , Glioblastoma/terapia , Imunoterapia/métodos , Lipoproteínas HDL/química , Lipoproteínas HDL/uso terapêutico , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Docetaxel/química , Docetaxel/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Lomustina/química , Lomustina/uso terapêutico , Camundongos , Modelos Biológicos , Paclitaxel/química , Paclitaxel/uso terapêutico , Ratos , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo
5.
J Am Vet Med Assoc ; 250(3): 322-326, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-28117638

RESUMO

OBJECTIVE To determine the lomustine content (potency) in compounded and FDA-approved lomustine capsules. DESIGN Evaluation study. SAMPLE 2 formulations of lomustine capsules (low dose [7 to 11 mg] and high dose [40 to 48 mg]; 5 capsules/dose/source) from 3 compounders and from 1 manufacturer of FDA-approved capsules. PROCEDURES Lomustine content was measured by use of a validated high-pressure liquid chromatography method. An a priori acceptable range of 90% to 110% of the stated lomustine content was selected on the basis of US Pharmacopeia guidelines. RESULTS The measured amount of lomustine in all compounded capsules was less than the stated content (range, 59% to 95%) and was frequently outside the acceptable range (failure rate, 2/5 to 5/5). Coefficients of variation for lomustine content ranged from 4.1% to 16.7% for compounded low-dose capsules and from 1.1% to 10.8% for compounded high-dose capsules. The measured amount of lomustine in all FDA-approved capsules was slightly above the stated content (range, 104% to 110%) and consistently within the acceptable range. Coefficients of variation for lomustine content were 0.5% for low-dose and 2.3% for high-dose FDA-approved capsules. CONCLUSIONS AND CLINICAL RELEVANCE Compounded lomustine frequently did not contain the stated content of active drug and had a wider range of lomustine content variability than did the FDA-approved product. The sample size was small, and larger studies are needed to confirm these findings; however, we recommend that compounded veterinary formulations of lomustine not be used when appropriate doses can be achieved with FDA-approved capsules or combinations of FDA-approved capsules.


Assuntos
Antineoplásicos Alquilantes/química , Composição de Medicamentos , Lomustina/química , United States Food and Drug Administration , Antineoplásicos Alquilantes/normas , Cápsulas/química , Cápsulas/normas , Lomustina/normas , Estados Unidos
6.
J Vet Intern Med ; 30(1): 242-6, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26682700

RESUMO

BACKGROUND: Compounded lomustine is used commonly in veterinary patients. However, the potential variability in these formulations is unknown and concern exists that compounded formulations of drugs may differ in potency from Food and Drug Administration (FDA)-approved products. HYPOTHESIS/OBJECTIVES: The initial objective of this study was to evaluate the frequency and severity of neutropenia in dogs treated with compounded or FDA-approved formulations of lomustine. Subsequent analyses aimed to determine the potency of lomustine obtained from several compounding pharmacies. ANIMALS: Thirty-seven dogs treated with FDA-approved or compounded lomustine. METHODS: Dogs that received compounded or FDA-approved lomustine and had pretreatment and nadir CBCs performed were eligible for inclusion. Variables assessed included lomustine dose, neutrophil counts, and severity of neutropenia. Lomustine 5 mg capsules from 5 compounding sources were tested for potency using high-pressure liquid chromatography (HPLC) with ultraviolet (UV) detection. RESULTS: Twenty-one dogs received FDA-approved lomustine and 16 dogs were treated with lomustine prescribed from a single compounding pharmacy. All dogs treated with FDA-approved lomustine were neutropenic after treatment; 15 dogs (71%) developed grade 3 or higher neutropenia. Four dogs (25%) given compounded lomustine became neutropenic, with 2 dogs (12.5%) developing grade 3 neutropenia. The potency of lomustine from 5 compounding pharmacies ranged from 50 to 115% of the labeled concentration, with 1 sample within ±10% of the labeled concentration. CONCLUSIONS AND CLINICAL IMPORTANCE: These data support broader investigation into the potency and consistency of compounded chemotherapy drugs and highlight the potential need for greater oversight of these products.


Assuntos
Antineoplásicos Alquilantes/efeitos adversos , Doenças do Cão/induzido quimicamente , Composição de Medicamentos , Lomustina/efeitos adversos , Neoplasias/veterinária , Neutropenia/veterinária , Animais , Doenças do Cão/tratamento farmacológico , Cães , Feminino , Lomustina/química , Lomustina/uso terapêutico , Masculino , Neoplasias/tratamento farmacológico , Neutropenia/induzido quimicamente , Farmácia/normas
7.
J Biomol Struct Dyn ; 33(8): 1653-68, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25350567

RESUMO

Chloroethyl nitrosoureas constitute an important family of cancer chemotherapeutic agents, used in the treatment of various types of cancer. They exert antitumor activity by inducing DNA interstrand cross-links. Semustine, a chloroethyl nitrosourea, is a 4-methyl derivative of lomustine. There exist some interesting reports dealing with DNA-binding properties of chloroethyl nitrosoureas; however, underlying mechanism of cytotoxicity caused by semustine has not been precisely and completely delineated. The present work focuses on understanding semustine-DNA interaction to comprehend its anti-proliferative action at molecular level using various spectroscopic techniques. Attenuated total reflection-Fourier transform infrared (ATR-FTIR) spectroscopy is used to determine the binding site of semustine on DNA. Conformational transition in DNA after semustine complexation is investigated using circular dichroism (CD) spectroscopy. Stability of semustine-DNA complexes is determined using absorption spectroscopy. Results of the present study demonstrate that semustine performs major-groove-directed DNA alkylation at guanine residues in an incubation-time-drug-concentration-dependent manner. CD spectral outcomes suggest partial transition of DNA from native B-conformation to C-form. Calculated binding constants (Ka) for semustine and lomustine interactions with DNA are 1.53 × 10(3) M(-1) and 8.12 × 10(3) M(-1), respectively. Moreover, molecular modeling simulation is performed to predict preferential binding orientation of semustine with DNA that corroborates well with spectral outcomes. Results based on comparative study of DNA-binding properties of semustine and lomustine, presented here, may establish a correlation between molecular structure and cytotoxicity of chloroethyl nitrosoureas that may be instrumental in the designing and synthesis of new nitrosourea therapeutics possessing better efficacy and fewer side effects.


Assuntos
Antineoplásicos Alquilantes/química , Adutos de DNA/química , DNA/química , Lomustina/química , Modelos Moleculares , Semustina/química , Algoritmos , Antineoplásicos Alquilantes/metabolismo , Dicroísmo Circular , DNA/metabolismo , Lomustina/metabolismo , Conformação Molecular , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Espectroscopia de Infravermelho com Transformada de Fourier
8.
J Photochem Photobiol B ; 130: 281-6, 2014 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-24368412

RESUMO

Investigation of drug-DNA interaction is important for understanding the drug action at molecular level and for designing specific DNA targeted drug. Lomustine (CCNU=1-[2-chloroethyl]-3-cyclohexyl-1-nitroso-urea) is an alkylating antineoplastic nitrosourea derivative, used to treat different types of cancer. In the present study, conformational and structural effects of lomustine on DNA are investigated using different spectroscopic approaches. Different drug/DNA molar ratios are analyzed to determine the binding sites and binding mode of lomustine with DNA. Fourier transform infrared spectroscopic (FTIR) results suggest binding of lomustine with nitrogenous bases guanine and cytosine along with weak interaction to the sugar-phosphate backbone of DNA. Circular dichroism (CD) spectroscopic results show perturbation in the local conformation of DNA upon binding of lomustine with DNA helix. These local conformational changes may act as recognition site for alkylating enzymes that further causes alkylation of DNA. Spectroscopic results confirm the formation of an intermediate stage of DNA that occurs during the transition of B-conformation into A-conformation.


Assuntos
Antineoplásicos Alquilantes/química , DNA/química , Lomustina/química , Dicroísmo Circular , Espectroscopia de Infravermelho com Transformada de Fourier
10.
Chem Pharm Bull (Tokyo) ; 59(3): 315-20, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21372411

RESUMO

The aim of this work was to prepare chitosan nanoparticles loaded with antineoplastic drug Lomustine (LCNPs), by ionic-gelation method with homogenization. The nanoparticles were characterized for particle size, polydispersity index (PDI), surface morphology, encapsulation efficiency, in-vitro drug release and cytotoxicity on human lung cancer cell line L132 by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The particle size, zeta potential and encapsulation efficiency of prepared nanoparticles ranged from 75 ± 1.1 to 637 ± 1.6 nm (PDI from 0.05 ± 0.001 to 0.18 ± 0.007), 37.2 ± 0.21 to 53.8 ± 0.18 mV and 66.74 ± 1.4 to 98.0 ± 1.8% respectively. The particles were spherical in shape with smooth surface in scanning electron microscopy (SEM) and transmission electron microscopy (TEM) images. Mechanical shearing by homogenization treatment significantly changed the nanoparticle size. The drug release rate was biphasic and diffusion controlled over the 8 h. LCNPs greatly inhibited the growth of the L132 cancer cell line used in this study in comparison to the native Lomustine (LMT).


Assuntos
Antineoplásicos/química , Quitosana/química , Lomustina/química , Neoplasias Pulmonares/tratamento farmacológico , Nanopartículas/química , Antineoplásicos/uso terapêutico , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Humanos , Lomustina/uso terapêutico , Lomustina/toxicidade , Nanopartículas/ultraestrutura , Tamanho da Partícula , Espectroscopia de Infravermelho com Transformada de Fourier
11.
J Pharm Biomed Anal ; 54(1): 213-6, 2011 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-20719460

RESUMO

A simple, inexpensive and rapid liquid chromatography (LC) method has been developed for the quantitative determination of Lomustine, an chemotherapy drug. Degradation studies were performed on the bulk drug by heating to 60 °C, exposure to UV light at an energy of 200 Wh/m(2)and to visible light at an illumination of not less than 1.2 million lux hours, acid (0.1N hydrochloric acid), base (0.1N sodium hydroxide) aqueous hydrolysis and oxidation with 6.0% (v/v) hydrogen peroxide. Good resolution between the peaks corresponding to impurities produced during synthesis, degradation products and the analyte was achieved on a Symmetry C 8 LC column using a mobile phase consisting of a mixture of aqueous potassium dihydrogen phosphate and acetonitrile. The degradation samples were assayed against the reference standard of Lomustine and the mass balance in each case was close to 99.9%. Validation of the method was carried out as per International Conference on Harmonization (ICH) requirements.


Assuntos
Química Farmacêutica/métodos , Cromatografia Líquida/métodos , Lomustina/análise , Acetonitrilas/química , Técnicas de Química Analítica , Cromatografia/métodos , Estabilidade de Medicamentos , Peróxido de Hidrogênio/química , Hidrólise , Lomustina/química , Modelos Químicos , Reprodutibilidade dos Testes , Temperatura , Raios Ultravioleta
12.
Mol Pharm ; 6(2): 504-12, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19718801

RESUMO

The present study describes a novel nonradioactive methodology for in vivo noninvasive, real-time imaging of blood-brain barrier (BBB) permeability for conventional drugs, using nitroxyl radicals as spin-labels and magnetic resonance imaging (MRI). Two TEMPO-labeled analogues (SLENU and SLCNUgly) of the anticancer drug lomustine [1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea] were synthesized, using a substitution of the cyclohexyl part with nitroxyl radical. Nonmodified nitroxyl radical TEMPOL was used for comparison. The nitroxyl derivatives were injected intravenously in healthy mice via the tail vein, and MR imaging of the brain was performed on a 7.0 T MRI. The MRI signal dynamic of SLENU and SLCNUgly followed the same kinetics as nonmodified TEMPO radical. SLENU and SLCNUgly were rapidly transported and randomly distributed in the brain tissue, which indicated that the exchange of cyclohexyl part of lomustine with TEMPO radical did not suppress the permeability of the anticancer drug for BBB. The selected nitroxyl derivatives possessed different hydrophobicity, cell permeabilization ability, and blood clearance. Based on these differences, we investigated the relationship betweenthe structure of nitroxyl derivatives, their half-life in the circulation, and their MRI signal dynamic in the brain. This information was important for estimation of the merits and demerits of the described methodology and finding pathways for overcoming the restrictions.


Assuntos
Antineoplásicos/química , Barreira Hematoencefálica/anatomia & histologia , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Óxidos N-Cíclicos/química , Lomustina/química , Óxidos de Nitrogênio/química , Animais , Antineoplásicos/farmacocinética , Velocidade do Fluxo Sanguíneo , Espectroscopia de Ressonância de Spin Eletrônica , Lomustina/análogos & derivados , Lomustina/farmacocinética , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Marcadores de Spin , Distribuição Tecidual
14.
Int J Pharm ; 222(2): 237-42, 2001 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-11427354

RESUMO

Physicochemical properties such as alkylating and carbamoylating activity and in vivo antimelanomic effects against B16 melanoma of the spin labeled (nitroxyl free radical containing) glycine nitrosourea (SLCNUgly) and its nonlabeled analogue (ChCNUgly), synthesized in our laboratory are studied and compared to those of antitumour drug 3-cyclohexyl-1-(2-chloroethyl)-1-nitrosourea (CCNU). We have demonstrated that introducing of glycine moiety in the nitrosourea structure in practice does not affect either alkylating or carbamoylating activity. On the other hand replacement of cyclohexyl moiety in ChCNUgly structure with nitroxyl free radical leads to a decrease in carbamoylating activity and an increase in alkylating activity. Compound ChCNUgly showed in vivo a higher antimelanomic activity against B16 melanoma in comparison with CCNU and SLCNUgly. It completely inhibited B16 melanoma growth (TGI=100%) at a dose 64.0 mg/kg. Moreover, we established that joint i.p. application in normal mice of SLCNUgly plus a new immunostimulator (C3bgp) formerly isolated in our laboratory led to a 75% restoration in immune function with respect to antibody production measured by Jerne hemolytic plaque assay. In contrast, no immunostimulation was found after joint application of C3bgp plus ChCNUgly or CCNU at the same experimental conditions. Based on these preliminary results, a possibility for developing of new combination immunochemotherapy schemes for treatment of human cancers is discussed.


Assuntos
Adjuvantes Imunológicos/química , Antineoplásicos/química , Glicina/química , Lomustina/química , Melanoma Experimental/tratamento farmacológico , Compostos de Nitrosoureia/química , Adjuvantes Imunológicos/uso terapêutico , Animais , Antineoplásicos/uso terapêutico , Lomustina/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Compostos de Nitrosoureia/uso terapêutico , Marcadores de Spin
15.
Cancer Biochem Biophys ; 17(1-2): 99-108, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10738906

RESUMO

The modulating effect of newly synthesized alkylating spin labeled triazene and spin labeled nitrosourea derivatives on the DOPA-oxidase activity of mushroom tyrosinase has been investigated by Bumett's spectrophotometric method (Burnett et al., 1967). All spin labeled triazenes have exhibited activating effect on DOPA-oxidase activity of tyrosinase, whereas clinically used triazene (DTIC), which does not contain nitroxide moiety, have showed inhibiting effect. At the same experimental conditions the spin labeled aminoacid nitrosoureas have showed dual effect - activating, in the beginning of the enzyme reaction and inhibiting later on. It is deduced that the activating effect of the spin labeled compounds is due to the nitroxide moiety and the inhibiting effect of all compounds depends on their half-life time. This study might contribute to make more clear the mechanism of action of the new compounds and on the other hand would come in quite useful as a preliminary prognosis for their antimelanomic activity.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Inibidores Enzimáticos/farmacologia , Melanoma Experimental/enzimologia , Monofenol Mono-Oxigenase/antagonistas & inibidores , Compostos de Nitrosoureia/farmacologia , Marcadores de Spin , Triazenos/farmacologia , Animais , Catálise , Óxidos N-Cíclicos/química , Óxidos N-Cíclicos/farmacologia , Dacarbazina/química , Dacarbazina/farmacologia , Di-Hidroxifenilalanina/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores Enzimáticos/química , Lomustina/química , Lomustina/farmacologia , Melanoma Experimental/tratamento farmacológico , Camundongos , Estrutura Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Oxirredução , Piperidinas/química , Piperidinas/farmacologia , Proteínas de Plantas/antagonistas & inibidores , Espectrofotometria , Relação Estrutura-Atividade , Triazenos/química , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/enzimologia
16.
Free Radic Biol Med ; 9(5): 423-6, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-1963416

RESUMO

We studied the formation of a free radical induced by UV irradiation of 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (CCNU) in benzene. It was determined a stable nitroxide radical by ESR spectroscopy. We confirmed that sterically hindered cyclic amine 2,2,6,6-tetramethyl-4-piperidone (TMPone) was transformed into a corresponding stable free nitroxyl radical during UV irradiation. When CCNU was present, the rate of free radical formation from TMPone increased considerably.


Assuntos
Lomustina/efeitos da radiação , Benzeno , Espectroscopia de Ressonância de Spin Eletrônica , Radicais Livres , Lomustina/química , Triacetonamina-N-Oxil , Raios Ultravioleta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA