Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci Methods ; 222: 131-7, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-24269253

RESUMO

BACKGROUND: Convection-enhanced delivery (CED) has been developed as a potentially effective drug-delivery strategy into the central nervous system. In contrast to systemic intravenous administration, local delivery achieves high concentration and prolonged retention in the local tissue, with increased chance of local toxicity, especially with toxic agents such as chemotherapeutic agents. Therefore, the factors that affect local toxicity should be extensively studied. NEW METHOD: With the assumption that concentration-oriented evaluation of toxicity is important for local CED, we evaluated the appearance of local toxicity among different agents after delivery with CED and studied if it is dose dependent or concentration dependent. RESULTS: Local toxicity profile of chemotherapeutic agents delivered via CED indicates BCNU was dose-dependent, whereas that of ACNU was concentration-dependent. On the other hand, local toxicity for doxorubicin, which is not distributed effectively by CED, was dose-dependent. Local toxicity for PLD, which is extensively distributed by CED, was concentration-dependent. COMPARISON WITH EXISTING METHOD: Traditional evaluation of drug induced toxicity was dose-oriented. This is true for systemic intravascular delivery. However, with local CED, toxicity of several drugs exacerbated in concentration-dependent manner. From our study, local toxicity of drugs that are likely to distribute effectively tended to be concentration-dependent. CONCLUSION: Concentration rather than dose may be more important for the toxicity of agents that are effectively distributed by CED. Concentration-oriented evaluation of toxicity is more important for CED.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/toxicidade , Encéfalo/efeitos dos fármacos , Sistemas de Liberação de Medicamentos/métodos , Animais , Antineoplásicos/farmacocinética , Encéfalo/patologia , Carmustina/administração & dosagem , Carmustina/farmacocinética , Carmustina/toxicidade , Convecção , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/patologia , Difusão , Relação Dose-Resposta a Droga , Doxorrubicina/administração & dosagem , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacocinética , Doxorrubicina/toxicidade , Imuno-Histoquímica , Masculino , Nimustina/administração & dosagem , Nimustina/farmacocinética , Nimustina/toxicidade , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/farmacocinética , Polietilenoglicóis/toxicidade , Ratos Endogâmicos F344
2.
Cell Cycle ; 11(14): 2606-19, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22751442

RESUMO

Chloroethylnitrosureas (CNUs) are powerful DNA-reactive alkylating agents used in cancer therapy. Here, we analyzed cyto- and genotoxicity of nimustine (ACNU), a representative of CNUs, in synchronized cells and in cells deficient in repair proteins involved in homologous recombination (HR) or nonhomologous end-joining (NHEJ). We show that HR mutants are extremely sensitive to ACNU, as measured by colony formation, induction of apoptosis and chromosomal aberrations. The NHEJ mutants differed in their sensitivity, with Ku80 mutants being moderately sensitive and DNA-PKcs mutated cells being resistant. HR mutated cells displayed a sustained high level of γH2AX foci, which co-stained with Rad51 and 53BP1, indicating DNA double-strand breaks (DSB) to be formed. Using synchronized cells, we analyzed whether DSB formation after ACNU treatment was replication-dependent. We show that γH2AX foci were not induced in G(1) but increased significantly in S phase and remained at a high level in G(2), where a fraction of cells became arrested and underwent, with a delay of > 12 h, cell death by apoptosis and necrosis. Rad51, ATM, MDC-1 and RPA-2 foci were also formed and shown to co-localize with γH2AX foci induced in S phase, indicating that the DNA damage response was activated. All effects observed were abrogated by MGMT, which repairs O(6)-chloroethylguanine that is converted into DNA cross-links. We deduce that the major genotoxic and killing lesion induced by CNUs are O(6)-chloroethylguanine-triggered cross-links, which give rise to DSBs in the treatment cell cycle, and that HR, but not NHEJ, is the major route of protection against this group of anticancer drugs. Base excision repair had no significant impact on ACNU-induced cytotoxicity.


Assuntos
Antineoplásicos/toxicidade , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Reparo do DNA por Junção de Extremidades/efeitos dos fármacos , Recombinação Homóloga/efeitos dos fármacos , Nimustina/toxicidade , Animais , Antígenos Nucleares/metabolismo , Apoptose , Células CHO , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Cricetinae , Cricetulus , Proteínas de Ligação a DNA/metabolismo , Fase G1 , Fase G2 , Histonas/metabolismo , Autoantígeno Ku , Necrose , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Rad51 Recombinase/metabolismo , Fase S
3.
Anticancer Res ; 22(2A): 697-701, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12014639

RESUMO

BCNU was reported to have about a 6- to 8- fold lower cytotoxic potency than ACNU in cell lines naturally deficient in O6-AGT. In seven tumor cell lines with an O6-AGT activity ranging from 40 to 360 fmol/mg the cytotoxic potency of BCNU, ACNU and HeCNU, without and after O6-AGT depletion by O6-BG, was determined. Without O6-AGT depletion, BCNU was superior to both other drugs in tumor cells with high O6-AGT activity. After O6-AGT depletion, the cytotoxic potency (comparison of IC50 values) of ACNU was higher than that of BCNU (p=0.016) or that of HeCNU (p=0.016) in all tumor cell lines. We conclude that (without O6-AGT depletion) BCNU is the drug of choice especially in tumor cells with high transferase activity. The higher cytotoxic potency of ACNU after O6-AGT depletion as compared to BCNU after O6-AGT depletion is countered by the higher toxicity of ACNU in patients necessitating a clinical dose reduction as compared to BCNU. Thus, we would not expect superiority of ACNU + O6-BG over BCNU+ O6-BG after systemic administration.


Assuntos
Antineoplásicos Alquilantes/toxicidade , Guanina/análogos & derivados , Compostos de Nitrosoureia/toxicidade , O(6)-Metilguanina-DNA Metiltransferase/deficiência , Carmustina/toxicidade , Sinergismo Farmacológico , Guanina/metabolismo , Guanina/farmacologia , Humanos , Nimustina/toxicidade , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Células Tumorais Cultivadas
4.
Eur J Haematol ; 67(1): 2-13, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11553261

RESUMO

Overexpression of O(6)-methylguanine DNA methyltransferase (MGMT) can protect hematopoietic cells from O(6)-alkylation damage. To identify possible clinical applications of this technology we compared the effect of MGMT gene transfer on the hematotoxicity induced by different O(6)-alkylating agents in clinical use: the chloroethylnitrosoureas ACNU, BCNU, CCNU and the tetrazine derivative temozolomide. In addition, various retroviral vectors expressing the MGMT-cDNA were investigated to identify optimal viral backbones for hematoprotection by MGMT expression. Protection from ACNU, BCNU, CCNU or temozolomide toxicity was evaluated utilizing a Moloney murine leukemia virus-based retroviral vector (N2/Zip-PGK-MGMT) to transduce primary murine bone marrow cells. Increased resistance in murine colony-forming units (CFU) was demonstrated for all four drugs. In comparison to mock-transduced controls, after transduction with N2/Zip-PGK-MGMT the IC50 for CFU increased on average 4.7-fold for ACNU, 2.5-fold for BCNU, 6.3-fold for CCNU and 1.5-fold for temozolomide. To study the effect of the retroviral backbone on hematoprotection various vectors expressing the human MGMT-cDNA from a murine embryonic sarcoma virus LTR (MSCV-MGMT) or a hybrid spleen focus-forming/murine embryonic sarcoma virus LTR (SF1-MGMT) were compared with the N2/Zip-PGK-MGMT vector. While all vectors increased resistance of transduced human CFU to ACNU, the SF1-MGMT construct was most efficient especially at high ACNU concentrations (8-12 microg/ml). Similar results were obtained for protection of murine high-proliferative-potential colony-forming cells. These data may help to optimize treatment design and retroviral constructs in future clinical studies aiming at hematoprotection by MGMT gene transfer.


Assuntos
Alquilantes/toxicidade , Dano ao DNA , Resistência a Medicamentos/genética , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/efeitos dos fármacos , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Retroviridae/genética , Alquilação , Animais , Carmustina/toxicidade , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , DNA Complementar/genética , Dacarbazina/análogos & derivados , Dacarbazina/toxicidade , Células-Tronco Hematopoéticas/enzimologia , Humanos , Lomustina/toxicidade , Camundongos , Vírus da Leucemia Murina de Moloney/genética , Nimustina/toxicidade , O(6)-Metilguanina-DNA Metiltransferase/genética , Proteínas Recombinantes de Fusão/fisiologia , Vírus do Sarcoma Murino/genética , Vírus Formadores de Foco no Baço/genética , Temozolomida , Sequências Repetidas Terminais , Transfecção
5.
J Neurooncol ; 48(1): 63-73, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-11026699

RESUMO

PURPOSE: To evaluate activity and toxicity of simultaneous ACNU and Ara-C with concurrent accelerated hyperfractionated radiotherapy in the treatment of high-grade glioma. PATIENTS AND METHODS: Thirty patients aged 23-71 years (median 47.5), 16 patients with glioblastoma multiforme (GBM) and 14 patients with grade-III glioma, received 93 courses of ACNU/Ara-C (median 4 courses) at following dose levels (ACNU/Ara-C in mg/m2/day): 70/90 (11 courses), 75/100 (36 courses) and 90/120 (46 courses). ACNU was administered IV on day 1 of each cycle, Ara-C as a 2 h-intravenous infusion on days 1-3. Patients received concomitant radiation therapy with 2 daily fractions of 1.75 Gy up to 57 Gy (median). RESULTS: Median survival of all patients was 13 months, 11 months for GBM and > 28 months for grade-III glioma; 31% (9 patients) survived longer than 24 months. The percentage of grade IV hematological toxicity was dose-dependent: 33% at the 70/90 dose level, 40% at 75/100 and 58% at 90/120. Six patients required platelet transfusion, 1 patient red blood cells; no febrile neutropenia occurred. Among 18 patients evaluable for response, 3 (17%) showed PR, 8 (44%) NC and 7 (39%) PD at completion of chemoradiation. No acute or late neurological toxicity occurred in this study. Younger age (p = 0.0001) and grade-III histology (p = 0.0009) were important prognostic factors for prolonged survival. CONCLUSION: This chemoradiation regimen is active in malignant gliomas and can be safely recommended at a dose level using 70 mg/m2 ACNU together with 90 mg/m2 Ara-C.


Assuntos
Antimetabólitos Antineoplásicos/administração & dosagem , Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/radioterapia , Citarabina/administração & dosagem , Glioblastoma/tratamento farmacológico , Glioblastoma/radioterapia , Nimustina/administração & dosagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Antimetabólitos Antineoplásicos/toxicidade , Antineoplásicos/toxicidade , Neoplasias Encefálicas/mortalidade , Terapia Combinada , Citarabina/toxicidade , Feminino , Glioblastoma/mortalidade , Humanos , Masculino , Pessoa de Meia-Idade , Nimustina/toxicidade , Radioterapia/métodos , Taxa de Sobrevida
6.
Carcinogenesis ; 21(10): 1879-83, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11023546

RESUMO

O(6)-methylguanine-DNA methyltransferase plays vital roles in preventing induction of mutations and cancer as well as cell death related to alkylating agents. Mice defective in the MGMT: gene, encoding the methyltransferase, were used to evaluate cell death-inducing and tumorigenic activities of therapeutic agents which have alkylation potential. MGMT(-/-) mice were considerably more sensitive to dacarbazine, a monofunctional triazene, than were wild-type mice, in terms of survival. When dacarbazine was administered i.p. to 6-week-old mice and survival at 30 days was enumerated, LD(50) values of MGMT(-/-) and MGMT(+/+) mice were 20 and 450 mg/kg body wt, respectively. Increased sensitivity of MGMT(-/-) mice to 1-(4-amino-2-methyl-5-pyrimidinyl)methyl-3-(2-chloroethyl)-3-nitrosou rea (ACNU), a bifunctional nitrosourea, was also noted. On the other hand, there was no difference in survival of MGMT(+/+) and MGMT(-/-) mice exposed to cyclophosphamide, a bifunctional nitrogen mustard. It appears that dacarbazine and ACNU produce O(6)-alkylguanine as a major toxic lesion, while cyclophosphamide yields other types of modifications in DNA which are not subjected to the action of the methyltransferase. MGMT(-/-) mice seem to be less refractory to the tumor-inducing effect of dacarbazine than are MGMT(+/+) mice. Thus, the level of O(6)-methylguanine-DNA methyltransferase activity is an important factor when determining susceptibility to drugs with the potential for alkylation.


Assuntos
Antineoplásicos Alquilantes/toxicidade , O(6)-Metilguanina-DNA Metiltransferase/deficiência , Adenoma/induzido quimicamente , Animais , Morte Celular/efeitos dos fármacos , Ciclofosfamida/toxicidade , Dacarbazina/toxicidade , Feminino , Dose Letal Mediana , Neoplasias Pulmonares/induzido quimicamente , Linfoma/induzido quimicamente , Masculino , Camundongos , Camundongos Knockout , Nimustina/toxicidade , O(6)-Metilguanina-DNA Metiltransferase/genética , Testículo/efeitos dos fármacos , Neoplasias do Timo/induzido quimicamente
7.
J Interferon Cytokine Res ; 20(6): 539-45, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10888110

RESUMO

The effect of recombinant human interleukin-11 (rHuIL-11) on myelosuppressive nimustine (ACNU)-induced thrombocytopenia was assessed in nonhuman primates. A single intravenous (i.v.) injection of ACNU (15 mg/kg) was administered to cynomolgus monkeys on day 0. rHuIL-11 (100 microg/kg/day) or the vehicle was given subcutaneously (s.c.) from day 1 to day 21. In monkeys receiving ACNU, the circulating platelet count decreased to a low of 42 +/- 6 x 10(9)/L by day 21 but returned to pretreatment levels (375 +/- 48 x 10(9)/L) on day 30. Administration of rHuIL-11 prevented severe thrombocytopenia; the platelet count fell only to 138 +/- 23 x 10(9)/L on day 18, and platelet recovery was faster (458 +/- 91 x 10(9)/L by day 27) compared with that of the control animals. The size of bone marrow megakaryocytes from rHuIL-11-treated animals was larger than that of the controls, indicating that rHuIL-11 stimulated megakaryopoiesis in a myelosuppressive condition. Treatment with ACNU also caused leukopenia and moderate anemia. rHuIL-11 transiently and slightly decreased the white blood cell (WBC) and red blood cell (RBC) counts. Conversely, rHuIL-11 accelerated recovery of RBC count in the late administration period. These results support the assertion that rHuIL-11 may be an important therapeutic agent for reducing the severity and duration of thrombocytopenia following cancer chemotherapy.


Assuntos
Antineoplásicos/toxicidade , Hematopoese/efeitos dos fármacos , Interleucina-11/farmacologia , Nimustina/toxicidade , Trombocitopenia/induzido quimicamente , Trombocitopenia/tratamento farmacológico , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/patologia , Contagem de Eritrócitos , Humanos , Contagem de Leucócitos , Macaca fascicularis , Masculino , Contagem de Plaquetas , Proteínas Recombinantes/farmacologia , Trombocitopenia/sangue
8.
Blood ; 95(12): 3771-80, 2000 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10845909

RESUMO

We examined the chemoprotective effects of KF41399, a novel derivative of carbazole compounds, on severe thrombocytopenia induced by nimustine (ACNU, 45 mg/kg administered for 2 consecutive days intravenously) in mice. Administration schedule studies revealed that pretreatment of mice with KF41399 was necessary to improve thrombocytopenia. Oral administration of KF41399 ameliorated thrombocytopenia induced by ACNU and accelerated the rate of platelet recovery in a dose-dependent fashion. In addition, KF41399 pretreatment improved the decrease in body weight and spleen weight and in the colony-forming activity of bone marrow mononuclear cells (MNC). Oral administration of KF41399 to normal mice induced G(0)/G(1)-phase accumulation of MNC as well as hematopoietic progenitor cells (lineage negative cells [Lin(-)]) and reduced the colony-forming activity of MNC. In Lin(-) cells derived from KF41399-treated mice, up-regulation of Bcl-2 and down-regulation of cyclin E and cyclin A proteins were observed. In the same cells, a decrease in the phosphorylated form of Rb protein and an increase in the p130 protein were observed without changes in the protein level of cell cycle-dependent kinase 2 (Cdk2), Cdk4, and Cdk6. More important, KF41399 did not affect the antitumor activity of ACNU against mouse Sarcoma180 and human lung cancer LC-6. However, 25-mg/kg KF41399 treatment reduced the antitumor activity of ACNU against human lung cancer Lu-65, and 5 mg/kg KF41399 caused a slight reduction of the antitumor activity of ACNU without inducing thrombocytopenia. These results suggest that KF41399 might be useful as a chemoprotective agent to improve chemotherapy-induced thrombocytopenia and types of other toxicity. (Blood. 2000;95:3771-3780)


Assuntos
Células da Medula Óssea/citologia , Carbazóis/farmacologia , Células-Tronco Hematopoéticas/citologia , Nimustina/toxicidade , Sarcoma 180/tratamento farmacológico , Trombocitopenia/prevenção & controle , Adenocarcinoma/tratamento farmacológico , Administração Oral , Animais , Células da Medula Óssea/efeitos dos fármacos , Carbazóis/administração & dosagem , Carbazóis/uso terapêutico , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Células K562 , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nimustina/uso terapêutico , Contagem de Plaquetas/efeitos dos fármacos , Baço/efeitos dos fármacos , Trombocitopenia/induzido quimicamente , Transplante Heterólogo
9.
Mutat Res ; 401(1-2): 133-41, 1998 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-9639692

RESUMO

Retrovirus-mediated transfer of O6-methylguanine-DNA methyltransferase (MGMT; E.C. 2.1.1.63) and a human multidrug-resistance gene (MDR1) confers resistance to nitrosoureas and natural product antitumor agents, respectively. In a previous study, we constructed two bicistronic retroviral vectors, Ha-MDR-IRES-MGMT and Ha-MGMT-IRES-MDR, that allow co-expression of the MGMT gene and the MDR1 gene to protect cells from the toxicity of combination chemotherapy. Each cell transduced with Ha-MDR-IRES-MGMT or Ha-MGMT-IRES-MDR showed high-level resistance to vincristine and 1-(4-amino-2-methyl-5-pyrimidinyl)methyl-3-(2-chloroethyl)-3-nitrosou rea (ACNU), indicating that the two drug-resistance genes can be functionally co-expressed from these vectors. In the present study, we examined whether the expression of MGMT from these MDR1-MGMT bicistronic retroviral vectors could protect cells from the genotoxicity of nitrosoureas. Three independent Ha-MDR-IRES-MGMT-transduced clones and three independent Ha-MGMT-IRES-MDR-transduced clones of HeLa MR cells showed 12-23-fold and 27-30-fold higher MGMT activity than the parental cells. These clones are more resistant to ACNU mutagenicity measured by the frequency of the emergence of 6-thioguanine-resistant colonies after ACNU treatment over the frequency seen in the parental cells. The ACNU-induced sister chromatid exchange (SCE) was markedly suppressed in these clones. Murine bone marrow cells were transduced with either Ha-MDR-IRES-MGMT or Ha-MGMT-IRES-MDR. Non-selected populations of the transduced cells showed only marginal increases in drug resistance and MGMT activity. Remarkable increase in drug resistance and MGMT activity were observed after a short exposure of the transduced cells to vincristine. The Ha-MDR-IRES-MGMT-transduced, vincristine-selected bone marrow cells showed 27-fold resistance to vincristine, 7-fold resistance to ACNU, and 10-fold higher MGMT activity than the non-transduced, non-selected cells. The Ha-MGMT-IRES-MDR-transduced, vincristine-selected cells showed 8-fold resistance to vincristine, 16-fold resistance to ACNU and 19-fold higher MGMT activity than the non-transduced, non-selected cells. The rates of ACNU-induced SCE in the vincristine-selected cells were as follows: non-transduced cells (non-selected) and HaMDR-transduced cells>Ha-MDR-IRES-MGMT-transduced cells>Ha-MGMT-IRES-MDR-transduced cells. Again, the only marginal levels of increases in the rates of ACNU-induced SCE were observed in non-selected population of the transduced cells. These results indicate that the MDR1-MGMT bicistronic retrovirus vectors would be useful to protect normal hematopoietic cells from nitrosourea-induced mutagenesis, and drug-selectable bicistronic constructs would have great advantage over non-selectable vectors.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Resistência a Múltiplos Medicamentos/genética , Genes , Nimustina/toxicidade , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Troca de Cromátide Irmã/efeitos dos fármacos , Animais , Células da Medula Óssea/citologia , Vetores Genéticos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Testes de Mutagenicidade , O(6)-Metilguanina-DNA Metiltransferase/biossíntese , O(6)-Metilguanina-DNA Metiltransferase/genética , Proibitinas , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Retroviridae , Transfecção , Vincristina/toxicidade
10.
Clin Cancer Res ; 3(6): 947-54, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9815770

RESUMO

Drug resistance genes can protect normal hematopoietic cells from the toxicity of anticancer agents. Because chemotherapeutic agents are often used in combination in current clinical protocols, coexpression of two different drug resistance genes should be useful in protecting normal bone marrow cells from the hematotoxicities caused by combination chemotherapy. In this study, we have combined the human multidrug resistance gene (MDR1) and human O6-methylguanine DNA methyltransferase (MGMT) gene as drug resistance genes. For the coexpression of two drug resistance genes, we have constructed two bicistronic retrovirus vectors. One vector is Ha-MDR-IRES-MGMT, in which translation of the MDR1 cDNA is cap-dependent and MGMT translation is dependent on an internal ribosome entry site (IRES). The other is Ha-MGMT-IRES-MDR, which has cap-dependent MGMT translation and IRES-dependent MDR1 translation. MGMT-negative HeLa derivative (MR) cells transduced with these retroviruses showed resistance to vincristine (from MDR1) and 1-(4-amino-2-methyl-5-pyrimidinyl)methyl-3-(2-chloroethyl)-3-nitrosou rea (ACN; from MGMT). Cells transduced with Ha-MDR-IRES-MGMT showed higher resistance to vincristine and lower resistance to ACNU than those transduced with Ha-MGMT-IRES-MDR. In any case, the resistance levels of cells transduced with either vector were high enough to select transduced cells with vincristine or ACNU. The expression levels of P-glycoprotein or MGMT in the transduced cells determined by FACS and Western blot analysis correlated well with the extent of resistance to vincristine and ACNU, respectively. All of the MGMT-transduced cells expressed higher amounts of MGMT than the MGMT-expressing parental cell line HeLa S3. Murine bone marrow cells transduced with Ha-MDR-IRES-MGMT and selected with vincristine also showed simultaneous resistance to vincristine and ACNU. These results suggest that bicistronic retroviral vectors allow the functional coexpression of two different types of drug resistance genes. This strategy could be applicable to any combination of drug resistance genes.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Antineoplásicos/toxicidade , Resistência a Múltiplos Medicamentos/genética , O(6)-Metilguanina-DNA Metiltransferase/genética , Retroviridae/genética , Células 3T3 , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Animais , Divisão Celular/efeitos dos fármacos , Doxorrubicina/toxicidade , Etoposídeo/toxicidade , Regulação da Expressão Gênica , Vetores Genéticos , Células HeLa , Humanos , Metilnitronitrosoguanidina/toxicidade , Camundongos , Nimustina/toxicidade , O(6)-Metilguanina-DNA Metiltransferase/biossíntese , Paclitaxel/toxicidade , Proibitinas , Proteínas Recombinantes/biossíntese , Transfecção/métodos , Vincristina/toxicidade
11.
Anticancer Res ; 17(3C): 1933-8, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9216647

RESUMO

We examined the genomic status of the p16 gene in 5 human glioma cell lines by Southern blot analysis. The p16 gene was located in the 9p21 chromosomal region and homozygous deletion was detected in 4 of 5 (80%) human glioma cell lines and 5 of 15 (33%) clinical samples. We transfected the full-length human p16 gene into p16-null human glioma cell line, U251MG cells, using the plasmid vector pRc/CMV-p16 and evaluated the effect of p16 gene transfer on the growth suppression of malignant glioma cells. The transfection of p16 cDNA caused growth suppression through G1 cell cycle arrest in U251MG cells. We also examined the effect of p16 gene transfer on the chemosensitivity to cis-diamminedichloroplatinum II (CDDP), 1-(4-amino-2-methyl-5-pyrimidinyl)methyl-3-(2-chloroethyl) -3-nitrosourea hydrochloride (ACNU), and 5'-azacytidine (AZC). We did not detect any change in them after p16 gene transfer. These results might suggest that deletion of p16 genes promoted unrestrained growth in human glioma but has no relationship to the chemosensitivity to CDDP, ACNU and AZC.


Assuntos
Antineoplásicos/toxicidade , Neoplasias Encefálicas/genética , Proteínas de Transporte/biossíntese , Genes Supressores de Tumor , Glioma/genética , Azacitidina/toxicidade , Neoplasias Encefálicas/patologia , Proteínas de Transporte/genética , Divisão Celular , Sobrevivência Celular/efeitos dos fármacos , Mapeamento Cromossômico , Cromossomos Humanos Par 9 , Cisplatino/toxicidade , Inibidor p16 de Quinase Dependente de Ciclina , DNA Complementar , Fase G1 , Deleção de Genes , Glioma/patologia , Humanos , Nimustina/toxicidade , Proteínas Recombinantes/biossíntese , Transfecção , Células Tumorais Cultivadas
12.
Anticancer Res ; 17(6D): 4611-4, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9494577

RESUMO

As part of the study of the appropriate usage of anticancer drugs, and the effects of the administration method (single dose, or divided doses) on the death rate from toxicity, five anticancer drugs (nimustine hydrochloride = ACNU, cyclophosphamide = CY, carboplatine = CBDCA, mitomycin C = MMC, vindesine = VDS) were studied in F-344 rats. A reduced death rate from toxicity was achieved by divided-dose administration of ACNU and CBDCA (group 1), but it was higher for CY and VDS (group 2), and there was no significant difference with MMC (group 3). In conclusion, the drugs were divided into three groups according to the administration method-related toxicity patterns. Improved knowledge of the properties of anticancer drugs is important for the success of chemotherapy.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/toxicidade , Transplante de Medula Óssea/fisiologia , Animais , Carboplatina/administração & dosagem , Carboplatina/toxicidade , Ciclofosfamida/administração & dosagem , Ciclofosfamida/toxicidade , Esquema de Medicação , Injeções Intravenosas , Masculino , Mitomicina/administração & dosagem , Mitomicina/toxicidade , Nimustina/administração & dosagem , Nimustina/toxicidade , Ratos , Ratos Endogâmicos F344 , Transplante Isogênico , Vindesina/administração & dosagem , Vindesina/toxicidade
13.
Biochem Biophys Res Commun ; 227(2): 558-63, 1996 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-8878552

RESUMO

We investigated the expression of DNA polymerase beta (beta-pol) and O6-methylguanine-DNA methyltransferase (MGMT) in human glioma cells with acquired resistance to 1-(4-amino-2-methyl-5-pyrimidinyl)-methyl-3-(2-chloroethyl)-3-nitrosoure a (ACNU) and in the parent cells. ACNU-resistant T430 (T430R) and A172 (A172R) glioma cell lines were established following repeated exposure to ACNU. The level of MGMT mRNA expression was elevated in T430R, but not in A172R. In contrast, the level of beta-pol mRNA expression and the level of beta-pol protein were elevated in A172R, compared with the parent cells. While the mechanism of MGMT repair has been considered to be important in the drug resistance of human brain tumors to ACNU, our present results demonstrate that beta-pol may also play an important role in the acquisition of tumor cell resistance to ACNU in human gliomas.


Assuntos
Antineoplásicos/toxicidade , DNA Polimerase I/biossíntese , Resistencia a Medicamentos Antineoplásicos , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Nimustina/toxicidade , Northern Blotting , Southern Blotting , Linhagem Celular , DNA Polimerase I/genética , Glioblastoma , Glioma , Humanos , RNA Mensageiro/biossíntese , Transcrição Gênica/efeitos dos fármacos , Células Tumorais Cultivadas
14.
Anticancer Res ; 16(1): 135-40, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8615598

RESUMO

In vitro assays for neurotoxicity with the aid of cultured mouse fetal neurons and glial cells were applied to investigate neurotoxicity of recombinant murine interferon-beta (rMuIFN-beta). These data were compared with those for MTX, ADR, and ACNU. The range of concentrations of the drugs used in these experiments spanned their clinically achievable concentrations in patient serum (IFN-beta: 1 x 10(4) IU/ml, MTX: 100 micrograms/ml, ADR: 20 micrograms/ml, ACNU: 20 micrograms/ml). rMuIFN- beta damaged both neurons and glial cells at concentrations of more than 1 x 10(5) IU/ml but did not damage them at 1 x 10(4) IU/ml or less. Microtubule-associated protein 1A (MAP1A) staining was decreased in rMuIFN-beta-treated (more than 1 x 10(5) IU/ml) neutrons. In conclusion, since IFN-beta may have some neurotoxic effects at concentrations higher than 1 x 10(5) IU/ml, it should be administered carefully, as should other antitumor agents, into the tumor cavity in the CNS following surgery.


Assuntos
Antineoplásicos/toxicidade , Encefalopatias/induzido quimicamente , Animais , Doxorrubicina/toxicidade , Feto , Glioma/tratamento farmacológico , Injeções Intraventriculares , Injeções Espinhais , Interferon beta/toxicidade , Metotrexato/toxicidade , Camundongos , Camundongos Endogâmicos C3H , Neuroglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Nimustina/toxicidade , Proteínas Recombinantes/toxicidade , Células Tumorais Cultivadas
15.
Int J Cancer ; 63(1): 148-51, 1995 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-7558444

RESUMO

Activity of O6-methylguanine-DNA methyltransferase (MGMT) is well related with drug resistance of tumor cells to chloroethylnitrosoureas (CENUs), because MGMT removes CENU-induced O6-alkylguanines in DNA by accepting the alkyl group at a cysteine moiety. Inactivation of MGMT is a feasible way to overcome MGMT-related resistance of tumor cells to CENUs. O6-Benzylguanine is known to be a strong depleter of MGMT. We previously reported the potentiation effect of O6-arylmethylguanine derivatives on cytotoxicity of 1-(4-amino-2-methyl-5-pyrimidinyl)methyl-3-(2-chloroethyl)-3-nitrosourea hydrochloride (ACNU), a CENU, for HeLa S3 cells. In this study, we tested the activity of these O6-arylmethylguanine derivatives as depleters of MGMT using HeLa S3 cell-free extract. The O6-arylmethylguanine derivatives tested were O6-benzylguanine (1), O6-(4-, 3-, and 2-fluorobenzyl)guanines (2-4), O6-(4, 3-, and 2-trifluoromethylbenzyl)guanines (5-7), O6-(4-, 3-, and 2-pyridylmethyl)guanines (8-10), and O6-(2- and 1-naphtylmethyl)guanines (11,12). Among these, compounds 1-3, 5, 8, 9 and 11 showed a strong MGMT depletion activity, whereas compounds 4, 6, 7, 10 and 12 were inactive. These inactive compounds, except for 6, have a substituent at the alpha position of the benzyl group (4, 7, 12) or are an alpha nitrogen analogue of 1 (10). There was a good relation (r = -0.856, p < 0.001) between the MGMT depletion activity of O6-arylmethylguanine derivatives and their potentiation activity of ACNU cytotoxicity. These results suggest that the alpha position of the benzyl group plays an important role in the interaction of O6-arylmethylguanine derivatives with MGMT to result in the inactivation of MGMT. Potent MGMT inactivators (1-3, 5, 8, 9, 11) sensitize tumor cells to CENU chemotherapy.


Assuntos
Inibidores Enzimáticos/farmacologia , Guanina/análogos & derivados , Guanina/farmacologia , Metiltransferases/antagonistas & inibidores , Antineoplásicos/farmacologia , Sinergismo Farmacológico , Guanina/química , Células HeLa , Humanos , Técnicas In Vitro , Nimustina/toxicidade , O(6)-Metilguanina-DNA Metiltransferase , Relação Estrutura-Atividade
16.
Biol Pharm Bull ; 18(3): 424-30, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7550096

RESUMO

It was reported recently that monomeric O6-benzylguanine (1) acts as an alternative substrate for a DNA repair enzyme, O6-alkylguanine-DNA alkyltransferase (AGT), and that therefore pretreatment of cells with 1 induces depletion of AGT resulting in an enhanced cytotoxic response to alkylating antitumor agents. In order to study the interaction of O6-benzylguanine derivatives with AGT and to obtain greater AGT depletion, we synthesized the following O6-arylmethylguanine derivatives and related compounds: O6-(4-, 3- and 2-fluorobenzyl)guanines (2, 3, 4), O6-(4-, 3- and 2-trifluoromethylbenzyl)guanines (5, 6, 7), O6-(4-, 3- and 2-pyridylmethyl)guanines (8, 9, 10), O6-(2- and 1-naphthylmethyl)guanines (11, 12), O6-biphenylmethylguanine (13), S and Se analogues of O6-benzylguanine (14, 15) and O6-phenylguanine (16). Ten of these are new compounds. All these compounds were tested for their potentiation of N'-[(4-amino-2-methyl-5-pyrimidinyl)methyl]-N-(2-chloroethyl)-N-nitrosou rea (ACNU) cytotoxicity using HeLa S3 and C6-1 cells. Compounds 2, 3, 5, 8, 9, 11 and 13 were active, as was 1. Compounds 7 and 12, with a substituent at the alpha position of the benzyl group, and compound 10, the alpha-nitrogen analogue of 1, were almost completely devoid of potentiating activity. These results suggest that the alpha-position of the O6-benzyl group plays an important role in the interaction of O6-benzylguanines with AGT. Of the other compounds, 4 and 6 exhibited very weak activity and 14, 15 and 16 were inactive. Possible reasons for these differences in activity are discussed in relation to the biomimetic dealkylation rates of O6-benzylguanine derivatives and the chemical characteristics of their substituents.


Assuntos
Antineoplásicos/toxicidade , Guanina/análogos & derivados , Nimustina/toxicidade , Alquilação , Animais , Fenômenos Químicos , Físico-Química , Sinergismo Farmacológico , Glioma/tratamento farmacológico , Guanina/síntese química , Guanina/farmacocinética , Guanina/farmacologia , Células HeLa , Humanos , Metiltransferases/metabolismo , O(6)-Metilguanina-DNA Metiltransferase , Fenóis/metabolismo , Proibitinas , Ratos , Células Tumorais Cultivadas
17.
Neurol Res ; 16(4): 234-40, 1994 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7984253

RESUMO

A simple and convenient technique for in situ quantification of DNA damage induced by 1-(4-amino-2-methyl-5-pyrimidinyl) methyl-3-(2-chloro-ethyl)-3-nitrosourea hydrochloride (ACNU) an alkylating agent, or irradiation was demonstrated in C6 glioma cells using a single cell gel electrophoresis. Treatment with ACNU or irradiation caused a dose dependent DNA damage which was detected by measuring the length of migration of fragmentary DNA in individual cells. Wild type C6 cells treated with ACNU (0, 10, 30, 60 micrograms ml-1) for one hour showed longer distance of migration of DNA than the ACNU-resistant subtype cells (C6R), indicating that ACNU-sensitive C6 cells were more vulnerable to ACNU than C6R cells. The results of DNA migration in C6 and C6R cells treated with ACNU were consistent with that from MTT assay which had been regarded as a standard method for chemosensitivity test. Furthermore, a time course study for DNA repair activity of C6 and C6R cells was also performed by measuring the length of DNA migration after incubation (0, 15, 30, 60, 120 min) of cells treated with 60 micrograms ml-1 ACNU. C6R cells repaired DNA damage more rapidly than C6 cells. In addition, the technique was also used to measure the DNA damage in C6 cells exposed to 0, 2, 6, 8, 10 Gy of x-ray irradiation, and a dose-dependent DNA migration after radiation injury was observed. This technique appears to be simple and useful for assessing chemosensitivity or radiosensitivity in individual glioma cells.


Assuntos
Alquilantes/toxicidade , Dano ao DNA , Reparo do DNA , Nimustina/toxicidade , Animais , Linhagem Celular , Sobrevivência Celular , DNA de Neoplasias/efeitos dos fármacos , DNA de Neoplasias/efeitos da radiação , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Glioma , Cinética , Ratos , Células Tumorais Cultivadas , Raios X
18.
J Neurooncol ; 22(1): 23-31, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-7714548

RESUMO

Several anticancer drugs have recently been shown to induce cell death in a manner similar to programmed cell death or apoptosis. The purpose of this study is to explore the mode of cell death caused by ACNU, a water-soluble nitrosourea. Exposure of rat glioma cell line KEG-1 to ACNU for 2 hours resulted in oligonucleosomal DNA fragmentation, creating a 'ladder' on agarose gel electrophoresis. DNA fragmentation began 18 hours after ACNU treatment, and preceded loss of membrane integrity as evaluated by the trypan blue exclusion test. The extent of DNA fragmentation increased in a dose-dependent manner, and the cell survival rate decreased reciprocally. A translational inhibitor, cycloheximide, suppressed this DNA fragmentation and enhanced cell survival rate with partial inhibition of protein synthesis. However, a transcriptional inhibitor, actinomycin D, failed to inhibit DNA fragmentation or enhance cell survival. Cycloheximide-inhibitable DNA fragmentation was also found in the KEG-1 implanted in vivo rat model following the administration of ACNU. These findings suggest that ACNU induces cell death associates with DNA fragmentation and partially with protein synthesis.


Assuntos
Cicloeximida/farmacologia , Nimustina/toxicidade , Animais , Morte Celular , Dano ao DNA , Dactinomicina/farmacologia , Masculino , Ratos , Ratos Wistar
19.
Neurol Med Chir (Tokyo) ; 33(2): 65-70, 1993 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7682667

RESUMO

The effects of ACNU and cranial irradiation on the immune system were studied in three groups of 90 mice: Group A, intraperitoneal injection of ACNU (30 mg/kg); Group B, single exposure of 10 Gy to the head; and Group C, intraperitoneal injection of ACNU (30 mg/kg) and single exposure of 10 Gy to the head. Peripheral white blood cell counts, spleen cell subsets, natural killer (NK) cell activity, lymphocyte blastogenesis, and production of interferon (IFN)-gamma were analyzed once a week for 6 weeks after treatment. In Group A, NK cell activity decreased between weeks 4-5, concanavalin A blastogenesis decreased during weeks 1-5, and the levels of L3T4 (CD4) and Lyt2 cells (CD8) and IFN-gamma production decreased during weeks 2-5. However, all tested parameters returned to the normal range at 6 weeks. In Group B, all parameters except for the L3T4 cell level and the IFN-gamma production decreased during week 1, and returned to the normal range thereafter. The concentration of L3T4 cells decreased during week 2 and between weeks 5-6. The IFN-gamma production increased during week 1, decreased during week 2, and returned to the normal range thereafter. In Group C, the suppressive effects were severe and continued for a longer period than in either Group A or B. Concanavalin A blastogenesis, L3T4 cell concentration, and IFN-gamma production were still suppressed after 6 weeks. Therefore, intensive radiochemotherapy for brain tumor may suppress the immunological function.


Assuntos
Irradiação Craniana/efeitos adversos , Síndromes de Imunodeficiência/etiologia , Nimustina/toxicidade , Animais , Contagem de Células/efeitos dos fármacos , Contagem de Células/efeitos da radiação , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/efeitos da radiação , Síndromes de Imunodeficiência/induzido quimicamente , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/efeitos da radiação , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/efeitos da radiação , Subpopulações de Linfócitos/efeitos dos fármacos , Subpopulações de Linfócitos/efeitos da radiação , Macrófagos/efeitos dos fármacos , Macrófagos/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos C57BL/imunologia , Nimustina/farmacologia , Organismos Livres de Patógenos Específicos , Fatores de Tempo
20.
J Neurooncol ; 14(2): 159-68, 1992 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1331350

RESUMO

The effect of recombinant human interleukin 1 beta (rHuIL-1 beta) on myelosuppression induced by 3-[(4-amino-2-methyl-5-pyrimidynyl)methyl]-1-(2-chloroethyl)-1-nit rosourea hydrochloride (ACNU) was studied. In in vivo study using BALB/c mice, pretreatment with 1 microgram/mouse of rHuIL-1 beta as a single intraperitoneal (i.p.) injection had a significant preventive effect on thrombocytopenia as well as granulocytopenia induced by ACNU at an intravenous dose of 60 mg/kg. Facilitated recovery by rHuIL-1 beta administered seven days after injection of high-dose ACNU was also observed. Experimental combination immunochemotherapy with high-dose ACNU and rHuIL-1 beta was performed in nude mice inoculated with human glioblastoma subcutaneously. The elongation of the survival time of the tumor bearing nude mice was also observed in combined use of high dose ACNU with rHuIL-1 beta. Seven patients with malignant brain tumors received intravenous 2.5-3 mg/kg ACNU. All patients were subcutaneously injected with 2 x 10(4)-U or more rHuIL-1 beta twice a week or daily. The mean nadir of leukocyte, granulocyte, and thrombocyte counts of the 7 patients received 2.5-3 mg/kg ACNU were significantly higher than in matched historical controls. In combination with rHuIL-1 beta, it may be possible to use chemotherapeutic agents at a relatively high dose.


Assuntos
Agranulocitose/prevenção & controle , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Fatores Imunológicos/uso terapêutico , Interleucina-1/uso terapêutico , Nimustina/efeitos adversos , Trombocitopenia/prevenção & controle , Idoso , Agranulocitose/induzido quimicamente , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/cirurgia , Terapia Combinada , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Glioblastoma/tratamento farmacológico , Glioblastoma/radioterapia , Glioblastoma/cirurgia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Nimustina/uso terapêutico , Nimustina/toxicidade , Proteínas Recombinantes/uso terapêutico , Indução de Remissão , Trombocitopenia/induzido quimicamente , Transplante Heterólogo , Vincristina/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...