Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Res ; 61(10): 4017-23, 2001 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11358820

RESUMO

This study describes a novel approach to the treatment of brain tumors with the combination of recombinant L-methionine-alpha-deamino-gamma-lyase and chemotherapeutic regimens that are currently used against such tumors. The growth of Daoy, SWB77, and D-54 xenografts in athymic mice was arrested after the depletion of mouse plasma methionine (MET) with a combination of a MET- and choline-free diet and recombinant L-methionine-alpha-deamino-gamma-lyase. The treated tumor-bearing mice were rescued from the toxic effects of MET withdrawal with daily i.p. homocystine. This regimen suppressed plasma MET to levels below 5 microM for several days, with no treatment-related deaths. MET depletion for 10-12 days induced mitotic and cell cycle arrest, apoptotic death, and widespread necrosis in tumors but did not prevent tumor regrowth after cessation of the regimen. However, when a single dose of 35 mg/m(2) of N,N'-bis(2-chloroethyl)-N-nitrosourea (BCNU), which was otherwise ineffective as a single therapy in any of the tumors tested, was given at the end of the MET depletion regimen, a more than 80-day growth delay was observed for Daoy and D-54, whereas the growth of SWB77 was delayed by 20 days. MET-depleting regimens also trebled the efficacy of temozolomide (TMZ) against SWB77 when TMZ was given to animals as a single dose of 180 mg/m(2) at the end of a 10-day period of MET depletion. The enhanced responses of both Daoy and SWB77 to DNA alkylating agents such as BCNU and TMZ could be attributed to the down-regulation of O(6)-methylguanine-DNA methyltransferase activity. However, the synergy of MET depletion and BCNU observed with D-54 tumors, which do not express measurable O(6)-methylguanine-DNA methyltransferase protein, is probably mediated by a different mechanism. MET depletion specifically sensitizes tumors to alkylating agents and does not significantly lower the toxicity of either BCNU or TMZ for the host. In this regard, the combination approach of MET depletion and genotoxic chemotherapy demonstrates significant promise for clinical evaluation.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Antineoplásicos Alquilantes/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Liases de Carbono-Enxofre/farmacologia , Dacarbazina/análogos & derivados , Glioblastoma/tratamento farmacológico , Metionina/deficiência , Animais , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/metabolismo , Carmustina/farmacologia , Colina/metabolismo , Dacarbazina/farmacologia , Dieta , Regulação para Baixo , Sinergismo Farmacológico , Glioblastoma/enzimologia , Glioblastoma/metabolismo , Humanos , Metionina/sangue , Metionina/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , O(6)-Metilguanina-DNA Metiltransferase/biossíntese , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Proteínas Recombinantes/farmacologia , Temozolomida , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Neuro Oncol ; 3(2): 99-112, 2001 04.
Artigo em Inglês | MEDLINE | ID: mdl-11296486

RESUMO

Glial tumors may originate from the malignant transformation of multipotent glial progenitor cells, but tools to study malignant transformation leading to gliomas are limited by the lack of biological systems that represent early stages of this disease in adult animals. In order to characterize the initiated cells that give rise to gliomas, we have employed the N-methylnitrosourea (MNU) model for induction of brain tumors in adult rats (Rushing et al., 1998). Specifically, we have isolated and cultured transformed (premalignant) cells from normal-appearing brains of rats exposed to MNU for 10 weeks and from histologically abnormal brains of rats exposed to MNU for 15 weeks. We compared them with cells cultured from control animals under identical conditions. Cultured cells were classified according to their morphology, immunophenotype, karyotype, proliferation capacity, and tumorigenicity in athymic mice. Cultures from untreated normal rat brains grew as monolayers and had normal karyotypes (42 X,Y), epithelioid morphology, and slow proliferative capacity (doubling time > 120 h). In contrast, cultured cells from brains of MNU-exposed animals had karyotypes that ranged from normal to highly aneuploid. Aneuploid lines grew rapidly in multilayers (doubling time < 24 h), had differentiated astrocytic or oligodendroglial morphology and immunohistochemical staining profile, and yielded tumors in athymic mice. Initiated cells with minor chromosomal aberrations assumed mixed bipolar or tripolar morphologies in high density cultures, proliferated rapidly, but showed contact inhibition and failed to induce tumors when injected s.c. in athymic mice. In general, lines showing no evidence of chromosomal aberrations had the most epithelioid morphology, proliferated slowly (doubling time > 72 h), and retained strict contact growth inhibition. The presumed undifferentiated glial progenitor cells in culture from either control or MNU-treated rats variably expressed markers such as vimentin, nestin, and NG2 proteoglycan, and they weakly expressed the mature astrocytic or oligodendroglial markers glial fibrillary acidic protein or galactocerbroside, respectively. These cultures differentiated to bipolar-tripolar morphology with concomitant maturation to a GFAP+ or GalC+ phenotype upon exposure to secondary messengers such as dibutyryl-cyclic-AMP and/or growth factors such as basic fibrillary growth factor. Continuous stimulation with these messengers resulted in terminal differentiation and consequent death upon withdrawal of the stimulus. These results provide information that could lead to detailed characterization of initiated, premalignant cells in the adult brain and to a better understanding of glial carcinogenesis.


Assuntos
Neoplasias do Sistema Nervoso Central/patologia , Sistema Nervoso Central/citologia , Animais , Técnicas de Cultura de Células , Transformação Celular Neoplásica/induzido quimicamente , Sistema Nervoso Central/patologia , Neoplasias do Sistema Nervoso Central/induzido quimicamente , Imuno-Histoquímica , Cariotipagem , Masculino , Metilnitrosoureia , Camundongos , Modelos Animais , Ratos , Ratos Sprague-Dawley , Células Tumorais Cultivadas
3.
Clin Cancer Res ; 7(2): 421-8, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11234899

RESUMO

Bis-2-chloroethylnitrosourea (BCNU) or temozolomide (TMZ) were tested alone or in combination with the AGT inhibitors O6-benzyl-2'-deoxyguanosine (dBG) or O6-benzylguanine (BG) against human glial tumor xenografts growing s.c. in athymic mice. Four glioblastoma (SWB77, SWB40, SWB39, and D-54) and one anaplastic oligodendroglioma (SWB61) xenografts having O6-alkylguanine-DNA alkyltransferase (AGT) activities of 75, 45, 10, < 10, and 16 fmol/mg protein, respectively, were used. BCNU at 35 mg/m2 was ineffective against these tumors, although 70 mg/m2 (LD10, 75 mg/m2) produced a marked tumor growth delay (T-C) in D54 but had no effect against SWB40 or SWB77. Coadministration of BG or dBG and BCNU necessitated reduction of the BCNU dose to a maximum of 30 and 35 mg/m2, respectively, because of increased toxicity. Optimized treatment with dBG (250 mg/m2) and BCNU (35 mg/m2) resulted in T-Cs of 30, 29, 11, 16, and 14 days for SWB77, SWB40, SWB39, D-54 and SWB61, respectively. These delays were more pronounced than those induced with optimized, isotoxic treatments with BG (180 mg/m2) and BCNU (30 mg/m2). In comparison to BCNU, TMZ was less toxic, with an LD10 of 400 mg/m2. TMZ (300 mg/m2) was more effective than BCNU against SWB77, SWB40, and SWB61, inducing T-Cs of 23, 53, and 56 days, respectively. BG and dBG enhanced the toxicity of TMZ in athymic mice by decreasing the LD10 from 400 to 200 mg/m2. TMZ (180 mg/m2) with either BG (180 mg/m2) or dBG (250 mg/m2) resulted in T-Cs of 31 and 49 days in SWB77, respectively, as compared with 16 days for TMZ (180 mg/m2) alone. In SWB40, the combination of TMZ with dBG, but not with BG, was significantly more effective than the maximum tolerated dose of TMZ (300 mg/m2) alone. The combination of TMZ with AGT inactivators had no benefit, as compared with TMZ alone, against xenografts with marginal AGT activity. In conclusion, at equimolar doses dBG was less toxic than BG in athymic mice when combined with either BCNU or TMZ. In this regard, BCNU or TMZ can be used at higher doses in combination with dBG than with BG. This study further demonstrates that there is a significant benefit of depleting AGT with nonspecific AGT inhibitors prior to treatment with either BCNU or TMZ in tumors having AGT activity >45 fmol/mg protein.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Carmustina/uso terapêutico , Dacarbazina/análogos & derivados , Dacarbazina/uso terapêutico , Desoxiguanosina/análogos & derivados , Glioma/tratamento farmacológico , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Animais , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptose , Desoxiguanosina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/uso terapêutico , Glioma/enzimologia , Glioma/patologia , Guanina/análogos & derivados , Guanina/uso terapêutico , Humanos , Camundongos , Camundongos Nus , O(6)-Metilguanina-DNA Metiltransferase/antagonistas & inibidores , Temozolomida , Transplante Heterólogo , Células Tumorais Cultivadas
4.
Cancer Chemother Pharmacol ; 45(1): 69-77, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10647505

RESUMO

PURPOSE: O6-Benzylguanine (BG), an O6-methylguanine-DNA methyltransferase (MGMT) inactivator, potentiates the efficacy of 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) and of other DNA chloroethylating and methylating anticancer drugs and is currently undergoing clinical trials. O6-Benzyl-2'-deoxyguanosine (dBG), a less effective MGMT inactivator than BG in vitro, is at least as effective as BG in combination with BCNU against tumor xenografts in athymic mice. In order to identify the mechanism of dBG activation in in vivo systems we tested the metabolism, ability to inactivate MGMT, and efficacy to potentiate BCNU in vivo of two additional 9-substituted derivatives of BG, namely O6-benzyl-9-cyanomethylguanine (CMBG) and O6-benzylguanosine (BGS). METHODS: Metabolism and disposition of these drugs was examined in athymic mice and Sprague-Dawley rats. MGMT suppression was determined in human medulloblastoma (Daoy) tumor xenografts in athymic mice following treatment with BGS, dBG, and CMBG and was compared with the loss of resistance to BCNU as determined by tumor growth delays. RESULTS: Growth delays at 25 mg/m2 BCNU and 133 mg/m2 BG or equimolar doses of CMBG, BGS or dBG were 23.0, 2.5, 21.3 days. and 30.4 days, respectively. The above differences did not correlate with the ED50S of 0.2, 13, 11 microM, and 2 microM determined for the above compounds, respectively, in cell free extracts. Differences in the efficacies of the 9-substituted compounds did correlate, however, with the extent of their metabolic conversion to BG. The maximum concentrations of BG in blood achieved after the administration of equimolar (250 micromol/kg) doses of CMBG, BGS and dBG were 10, 30 microM, and 55 microM, respectively. Although such levels were lower than those achieved in circulation by administration of an equimolar amount of BG, BG levels persisted longer following treatments with BGS or dBG than after treatment with BG itself. Formation of BG was required for continuous and prolonged (> 16 h) suppression of MGMT activity to non-detectable levels (< 5 fmol/mg protein). CONCLUSION: Metabolism of BGS and dBG to BG explains the unexpected high efficacy of these compounds in potentiating the antitumor activity of BCNU in the athymic mouse model. The faster and more effective suppression of tumor MGMT by dBG and its greater efficacy, as compared with BGS, also correlates with a more rapid accumulation of BG in blood after dBG than after BGS administration, which results in faster and complete suppression of MGMT in Daov xenografts. Thus, metabolism of dBG and BGS to BG appears to be the determining factor for continuous and prolonged suppression of MGMT activity, and that near complete suppression of such activity during and following BCNU administration is required for the higher efficacy of treatments. Similarly, the failure of CMBG to suppress tumor MGMT to the same extent as BGS, in spite of their similar ED50 values, could be attributed to the metabolism of this compound mainly by pathways other than conversion to BG.


Assuntos
Antineoplásicos/farmacologia , Carmustina/farmacologia , Guanina/análogos & derivados , Animais , Encéfalo/metabolismo , Sinergismo Farmacológico , Guanina/metabolismo , Guanina/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , O(6)-Metilguanina-DNA Metiltransferase/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley
5.
Clin Cancer Res ; 5(11): 3676-81, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10589786

RESUMO

O6-Benzyl-2'-deoxyguanosine (dBG), a water-soluble inhibitor of O6-methylguanine-DNA methyltransferase (MGMT), potentiates the efficacy of 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) against MGMT-positive, BCNU-resistant Daoy human medulloblastoma tumor xenografts in athymic mice (S. C. Schold et al., Cancer Res., 56: 2076-2081, 1996). Such potentiation was comparable to that observed for O6-benzylguanine, the prototype MGMT inhibitor that is currently undergoing clinical trials. In this study, we optimized the therapeutic effect of the dBG and BCNU combination against brain tumor xenografts without inducing substantial toxicity in the host by adjusting the doses of both compounds. dBG was escalated from 133 mg/m2 to 200 and 300 mg/m2, whereas corresponding doses of BCNU were reduced from 25 mg/m2 to 17 and 11 mg/m2, respectively. The growth delays of 30.2, 38.4, and 22.3 days, respectively, observed for the above regimens suggest that the optimal drug combination is not achieved with maximum doses of dBG. In fact, the highest doses of dBG (300 mg/m2) contributed to more frequent BCNU-related toxicities, despite the reduced BCNU dosage, and a reduction of the therapeutic effect. Toxicity was related to the depletion of MGMT activity in the gut of host mice and was manifested by edema, inflammation, and hemorrhage in the bowel wall by subsequent BCNU administration. With additional dosage adjustments, we found that tumor suppression of >90 days without toxicity was observed at 200 mg/m2 dBG and 23 mg/m2 BCNU. At these doses, tumors were eradicated (regressed to an undetectable size for >90 days) in 8 of 12 animals. Thus, dBG is the first of the MGMT inhibitors to show a curative effect in combination with BCNU against a human central nervous system tumor xenograft in athymic mice.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/toxicidade , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Cerebelares/tratamento farmacológico , Desoxiguanosina/análogos & derivados , Meduloblastoma/tratamento farmacológico , O(6)-Metilguanina-DNA Metiltransferase/antagonistas & inibidores , Animais , Apoptose , Carmustina/administração & dosagem , Carmustina/toxicidade , Neoplasias Cerebelares/patologia , Desoxiguanosina/administração & dosagem , Desoxiguanosina/uso terapêutico , Desoxiguanosina/toxicidade , Humanos , Inflamação , Mucosa Intestinal/enzimologia , Meduloblastoma/patologia , Camundongos , Camundongos Nus , Mitose , Transplante Heterólogo , Células Tumorais Cultivadas , Redução de Peso/efeitos dos fármacos
6.
Clin Cancer Res ; 5(1): 209-13, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9918221

RESUMO

O6-benzylguanine (BG) is a potent, specific inactivator of the DNA repair protein O6-alkylguanine-DNA alkyltransferase (AGT), which enhances sensitivity to nitrosoureas in cells and tumor-bearing animals. BG is presently undergoing clinical trials for development as an agent to enhance the therapeutic index of alkylating agent chemotherapy. It has been reported that a polymorphism exists in the human agt gene, with about 15% of the Japanese population having arginine at codon 160 instead of glycine on the polypeptide (Y. Imai et al., Carcinogenesis, 16: 2441-2445, 1995). The resultant mutant AGT protein is equally effective against both methylated DNA as compared with wild type protein. However, this mutant AGT protein was less sensitive to inactivation by BG with a 20-fold increase in the ED50 value. This observation raised the possibility that a subpopulation of patients may be resistant to BG due to a single base change. We have demonstrated that this alteration also reduces the sensitivity to O6-benzyl-8-oxoguanine, an equally potent, yet much longer-lived human metabolite of BG. To test the possibility that this germ-line mutation of the agt gene might explain resistance to BG and O6-benzyl-8-oxoguanine of patients on our Phase I clinical trials, we evaluated the DNA from lymphocytes of 18 patients. The G160R mutation was not found in any of the 18 patients. To determine the frequency of this mutation in the United States population, DNA from 181 healthy individuals were investigated and, again, the mutation was not observed in this cohort. Therefore, if the mutation exists, it is in statistically <1.6% of the United States noncancerous population. To investigate the possibility that this mutation might be somatic, we evaluated genomic DNA samples from 94 human primary cancers of four different histological subtypes (brain, colon, esophageal, and head and neck). Again, none were found to have the G160R mutation.


Assuntos
Códon , Neoplasias/enzimologia , O(6)-Metilguanina-DNA Metiltransferase/genética , Polimorfismo Genético , Animais , Antineoplásicos/farmacologia , Bovinos , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Guanina/análogos & derivados , Guanina/farmacologia , Humanos , Mutação , Neoplasias/tratamento farmacológico , Neoplasias/genética , O(6)-Metilguanina-DNA Metiltransferase/antagonistas & inibidores , Reação em Cadeia da Polimerase
7.
J Clin Oncol ; 16(11): 3570-5, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9817277

RESUMO

PURPOSE: The major mechanism of resistance to alkylnitrosourea therapy is the DNA repair protein O6-alkylguanine-DNA alkyltransferase (AGT), which removes chlorethylation or methylation damage from the O6-position of guanine. O6-benzylguanine (O6-BG) is an AGT substrate that inhibits AGT by suicide inactivation. We conducted a phase I trial to define the presurgical dose required for depletion of tumor AGT activity in patients with malignant glioma. MATERIALS AND METHODS: Patients were to be treated 18 hours before craniotomy with intravenous doses that ranged between 40 and 100 mg/m2 given over 1 hour. Resected tumor was snap-frozen in liquid nitrogen and AGT activity analyzed by high-pressure liquid chromatography (HPLC). Up to 13 patients were treated at a specific dose of O6-BG, with a target end point of > or = 11 of 13 patients with undetectable tumor AGT levels (< 10 fmol/mg protein). RESULTS: Thirty patients with malignant gliomas were enrolled, with 11 of 11 patients treated at 100 mg/m2 O6-BG demonstrating tumor AGT levels less than 10 fmol/mg protein. No toxicity was noted in any patient treated. CONCLUSION: These results indicate that 100 mg/m2 of O6-BG can maintain tumor AGT levels less than 10 fmol/mg protein for at least 18 hours after treatment, a time interval in which bis(2-chloroethyl)nitrosourea (BCNU)-induced chloroethyl adducts are fully converted into interstrand cross-links. A 100-mg/m2 dose of O6-BG will be used in combination with BCNU in another phase I trial designed to determine the maximal-tolerated dose of BCNU.


Assuntos
Neoplasias Encefálicas/cirurgia , Inibidores Enzimáticos/administração & dosagem , Glioblastoma/cirurgia , Guanina/análogos & derivados , Adulto , Idoso , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/enzimologia , Glioblastoma/tratamento farmacológico , Glioblastoma/enzimologia , Guanina/administração & dosagem , Humanos , Pessoa de Meia-Idade , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Cuidados Pré-Operatórios
8.
J Neuropathol Exp Neurol ; 57(11): 1053-60, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9825942

RESUMO

Gliomas were induced in adult male Sprague-Dawley rats by continuous exposure to 100 ppm of N-nitrosmethylurea (MNU) in drinking water. Latency periods for such tumors were 20 and 50 weeks following completion of exposure intervals of 20, 15, and 10 weeks, respectively. Based on histomorphology and the pattern of GFAP immunoreactivity, a large percentage of MNU-induced tumors (>40%) were anaplastic mixed gliomas, having both neoplastic astrocytic and oligodendroglial components. Typical oligodendrogliomas and astrocytomas also occurred less frequently. Unlike the majority of tumors induced by ethylnitrosourea (ENU), MNU yielded glial tumors that did not express synaptophysin. Anaplastic mixed gliomas and glioblastoma multiforme (GBMs) had no missense p53 mutations in the commonly mutated exons 4 through 8 and did not overexpress wild-type p53, suggesting that MNU-induced oncogenesis in rat brain tumors may not require inactivation/alteration of the p53 tumor suppressor gene. The K-ras gene was also analyzed and found to have no activating mutations in brain tumors. This model is suitable for studying genetic events leading to the majority of gliomas that apparently express functional p53.


Assuntos
Carcinógenos , Glioma/induzido quimicamente , Glioma/genética , Metilnitrosoureia , Mutação de Sentido Incorreto/genética , Proteína Supressora de Tumor p53/genética , Animais , Astrócitos/patologia , Éxons/genética , Genes ras/genética , Glioma/patologia , Masculino , Oligodendroglia/patologia , Ratos , Ratos Sprague-Dawley
9.
Cancer Res ; 57(23): 5360-8, 1997 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-9393761

RESUMO

Pancreatic adenocarcinomas rarely respond to radiation or chemotherapy, indicating that a large percentage of these tumors possess complex mechanisms of resistance. The failure of alkylating agents, such as carmustine [1,3-bis(2-chloroethyl)-1-nitrosourea; BCNU], lomustine [1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea; CCNU], and streptozotocin, to yield consistent therapeutic results further suggests that one of these mechanisms may be the high expression of O6-methylguanine-DNA methyltransferase (MGMT). All 12 human pancreatic ductal adenocarcinomas assayed for MGMT activity showed unusually high levels, implying that these malignancies are efficient in repairing genotoxic O6-alkylguanine lesions induced by methylating (streptozotocin) and 2-chloroethylating (BCNU and CCNU) chemotherapeutic genotoxic agents. Immunohistochemical analysis of an additional 15 pancreatic tumors showed that high levels of MGMT protein reside in the nucleus and the cytoplasm of malignant cells. Both nuclear and cytoplasmic staining were absent in hyperplastic duct epithelium, but staining was invariably present in moderate to highly dysplastic foci and especially strong in invasive components of the tumor. With the exception of lymphocytes that were MGMT positive, acinar, ductal, and islet cells did not stain for MGMT in histologically normal pancreata. These data indicate that MGMT activity is up-regulated in dysplastic epithelium, and its expression increases during tumor progression, reaching the highest levels in the invasive components of the tumor. Resistance of pancreatic tumor cells to alkylating agents was verified with four pancreatic tumor cell lines. CAPAN-2, CFPAC-1, PANC-1, and MIAPaCa-2, having MGMT levels of 1800, 987, 700, and 880 fmol/mg protein, respectively, were resistant to BCNU, but their resistance declined sharply following pretreatment with the MGMT inhibitor O6-benzylguanine (O6-BG). On the other hand, PANC-1 and MIAPaCa-2 could not be eradicated with N-methylnitrosourea (MNU) at concentrations as high as 2 mM, even when pretreated with O6-BG. These two lines were shown to be modified genetically in microsatellite sequences by MNU and are believed to have a defective mismatch repair system, which may explain their resistance to methylating agents. Failure of pancreatic tumors to respond to nitrosoureas is related to high levels of MGMT expression and in some cases to genomic instability. However, these tumors can be sensitized to chloroethylating drugs and eradicated following the elimination of MGMT activity by O6-BG or homologous MGMT inhibitors.


Assuntos
Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/enzimologia , Antineoplásicos Alquilantes/toxicidade , Resistencia a Medicamentos Antineoplásicos , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/enzimologia , Adenocarcinoma/patologia , Adenocarcinoma/cirurgia , Adulto , Idoso , Antineoplásicos Alquilantes/uso terapêutico , Apoptose/efeitos dos fármacos , Carmustina/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Lomustina/toxicidade , Masculino , Pessoa de Meia-Idade , Mitose/efeitos dos fármacos , Estadiamento de Neoplasias , O(6)-Metilguanina-DNA Metiltransferase/biossíntese , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/cirurgia , Estreptozocina/toxicidade , Taxa de Sobrevida , Células Tumorais Cultivadas
10.
Nutr Cancer ; 29(3): 195-204, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9457739

RESUMO

Depletion of plasma methionine is expected to inhibit or reverse growth of methionine-dependent tumors; however, modulation of methionine and other sulfur amino acids is not a trivial task in experimental animals. L-Methioninase from Pseudomonas putida at 1,000 U/kg causes acute reduction of plasma methionine by 80% in mice, but recovery occurs within 14 hours. Restriction of dietary choline and replacement of dietary methionine with homocystine results in 50% chronic reduction of plasma methionine. A > 70% reduction can be accomplished with a diet deficient in methionine, homocystine, and choline, but ultimately this diet is lethal. Plasma methionine can be lowered to a steady state of < 5 microM in mice with a combination of dietary restriction of methionine, homocysteine, and choline and synchronous treatments with intraperitoneal injections of 1,000 U/kg L-methioninase and 25-50 mg/kg homocystine, each administered at 12-hour intervals. Modulation of plasma methionine by this means causes no weight loss or pathologies in liver or pancreas, and it does not markedly alter levels of cysteine, homocysteine, or glutathione in plasma or in hepatic tissue. When this procedure is applied to athymic mice bearing human medulloblastoma (Daoy) tumors subcutaneously, tumor growth is inhibited. Methionine deprivation arrests mitosis by blocking the cell cycle in G2 and induces apoptosis. Tumor stasis was achieved in 100% of treated animals within 4 days of treatment, and regression was seen in one-third of animals after a 10-day period. These data strongly support the use of methionine-depleting regimens for tumor treatments.


Assuntos
Neoplasias Encefálicas/patologia , Dieta , Meduloblastoma/patologia , Metionina/sangue , Animais , Antimetabólitos Antineoplásicos/metabolismo , Antimetabólitos Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/sangue , Neoplasias Encefálicas/terapia , Liases de Carbono-Enxofre/metabolismo , Liases de Carbono-Enxofre/uso terapêutico , Cisteína/análise , Cisteína/sangue , Glutationa/análise , Glutationa/sangue , Homocisteína/análise , Homocisteína/sangue , Humanos , Fígado/química , Meduloblastoma/sangue , Meduloblastoma/terapia , Metionina/deficiência , Metionina/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Fatores de Tempo , Transplante Heterólogo
11.
Br J Cancer ; 75(6): 779-88, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9062396

RESUMO

Methionine (MET)-dependent cell lines require MET to proliferate, and homocysteine (HCY) does not act as a substitute for this requirement. From six O6-methylguanine-DNA methyltransferase (MGMT)-efficient (mer+) cell lines tested, two medulloblastomas (Daoy and D-341) and a lung non-small-cell adenocarcinoma with metastatic potential (H-1623) were most sensitive to MET deprivation, while two glioblastomas (U-138, D-263) and a small-cell lung carcinoma H-1944 were moderately to weakly dependent. Regardless of the degree of MET dependence, all of these lines down-regulated their MGMT activity within 48-72 h of transfer from MET+HCY- to MET-HCY+ media, long before the eradication of the culture. Reduction of MGMT activity was due to a decline of both MGMT mRNA and protein levels. However, the reduction was not related to the methylation status of the MGMT promoter at the SmaI site or the HpaII sites in the body of the gene; such sites have been shown to be associated in MGMT regulation and in defining the mer phenotype. MET-dependent, mer+ tumour cells cultured in MET-HCY+ were more sensitive to BCNU (IC50 = 5-10 microM) than those cultured in MET+HCY-(IC50 = 45-90 microM), while MET-independent or mer- cell lines were unaffected. This indicates that reduction of MGMT, imposed by the absence of MET, renders mer+ tumour cells more susceptible to alkylating agents. The relatively selective suppression of MGMT activity in mer+ MET-dependent tumour cells, in combination with the inability of such cells to proliferate in the absence of MET, may lead to the development of more effective treatment strategies for mer+ MET-dependent tumours.


Assuntos
Metionina/deficiência , Metiltransferases/metabolismo , Células 3T3/metabolismo , Células 3T3/patologia , Animais , Antineoplásicos Alquilantes , Carmustina/toxicidade , Contagem de Células , Divisão Celular , Regulação para Baixo , Sinergismo Farmacológico , Fase G1 , Fase G2 , Humanos , Metionina/metabolismo , Camundongos , O(6)-Metilguanina-DNA Metiltransferase , Fase S , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/patologia
12.
In Vivo ; 10(3): 297-306, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8797031

RESUMO

O6-Benzyl-2'-deoxyguanosine (O6-BzldGuo) was found to be a potent inhibitor of O6-methylguanine-DNA methyltransferase (MGMT) activity in vivo, despite its demonstrated lower activity compared to O6-benzylguanine (O6-BzlGua) for inactivation of MGMT in cell cultures or cell free extracts. This difference is probably due to metabolism and possibly to enhanced systemic availability of O6-BzldGuo compared to O6-BzlGua in animals. At doses above 50 mg/kg, O6-BzldGuo completely suppresses the MGMT activity in liver, lung and brain for 12 to 18 hrs following treatment. However, it fails to suppress the activity in jejunum and possibly in other tissues with low blood perfusion rates. O6-BzldGuo is metabolized mainly in the liver, and possibly in kidney and lung, to yield the more potent MGMT inhibitors O6-BzlGua and O6-Benzyl-7,8-dihydro-8-oxoguanine (O6-Bzl(8-OX)Gua), that reach maximum levels following the decline of the parent compound both in tissues and the circulation. Although O6-BzldGuo may cause the initial drop of MGMT activity, continued prolonged suppression is likely due to the persistence of its MGMT-inhibiting metabolites. Severe suppression of MGMT activity and the presence of O6-BzldGuo and its metabolites in the brain indicates that this inhibitor crosses the blood-brain barrier. Prolonged suppression of MGMT in brain following a complete disappearance of the parent compound from the tissue also demonstrates the importance of metabolites in MGMT suppression. Such metabolites, especially O6-Bzl(8-OX)Gua, appear to accumulate in brain, possibly due to their hydrophobic nature. Accumulation of the parent compound in the pancreatic ducts may explain the unusual high concentrations of this compound in pancreas as compared to the spleen. Similarly, high concentrations of O6-BzldGuo in kidneys may be related to accumulation of the compound in proximal tubules and its poor reabsorption into the circulation. Overall, O6-BzldGuo shows tissue specificity which is modulated by dose. Such specificity seems to be related to accumulation of the parent compound in certain tissues, but more importantly, to site-specific metabolism and the persistence of MGMT inhibitory metabolites in these tissues. The combination of enhanced systemic availability and site specific metabolic activation and/or inactivation of O6-BzldGuo may be related to the enhanced antitumor therapeutic index against human tumor xenografts for this compound which equals or even exceeds that of O6-BzlGua.


Assuntos
Desoxiguanosina/análogos & derivados , Inibidores Enzimáticos/farmacologia , Metiltransferases/antagonistas & inibidores , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Desoxiguanosina/farmacologia , Intestinos/efeitos dos fármacos , Intestinos/enzimologia , Rim/efeitos dos fármacos , Rim/enzimologia , Cinética , Fígado/efeitos dos fármacos , Fígado/enzimologia , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Masculino , Metiltransferases/metabolismo , O(6)-Metilguanina-DNA Metiltransferase , Especificidade de Órgãos , Pâncreas/efeitos dos fármacos , Pâncreas/enzimologia , Ratos , Ratos Sprague-Dawley , Baço/efeitos dos fármacos , Baço/enzimologia
13.
Cancer Res ; 56(9): 2076-81, 1996 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-8616853

RESUMO

O6-Methylguanine-DNA methyltransferase (MGMT), a constitutively expressed DNA repair protein, removes alkyl groups from the O6-position of guanine in DNA. Tumor cells with high MGMT activity are resistant to nitrosoureas and other agents that form toxic O6-alkyl adducts. O6-Benzylguanine (BG) inactivates the MGMT protein and thereby enhances the sensitivity of tumor cells to alkylating drugs. However, the therapeutic potential of BG is limited by its poor solubility and its nonspecific inactivation of MGMT in normal tissues as well as in tumor tissues. Consequently, BG analogues are being developed to identify agents that have more favorable pharmacological characteristics. We evaluated O6-benzyl-2'-deoxyguanosine (dBG), the 2'-deoxyribonucleoside analogue of BG, for its ability to inhibit MGMT and to potentiate 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) in a MGMT-positive human brain tumor xenograft, Daoy. When given i.p. 1 h before BCNU (25 mg/m2) to animals bearing s.c. tumors, dBG (134 mg/m2) produced a growth delay of 24.7 days, compared to 21.6 days after treatment with an equimolar dose of BG (90 mg/m2) plus BCNU and -0.6 days after treatment with BCNU alone. The combination of dBG + BCNU also increased the survival of animals bearing intracranial tumors by 65%. By increasing the dose of dBG to 300 mg/m2 (the maximum dose that could be delivered i.p. in a standard treatment volume), the growth delay of s.c. tumors increased from -0.1 days with BCNU alone to 39.3 days. dBG suppressed both tumor and liver MGMT activity to less than 1.5% of baseline, and dBG + BCNU induced extensive perivascular apoptosis. Because dBG is a 10-fold less potent MGMT inhibitor than BG in HT-29 cell extracts, these results illustrate the capacity of BG analogues to potentiate BCNU toxicity, despite less in vitro activity than the parent compound, and emphasize the importance of in vivo evaluation of BG analogues.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidade , Neoplasias Encefálicas/patologia , Carmustina/administração & dosagem , Carmustina/toxicidade , Morte Celular/efeitos dos fármacos , Desoxiguanosina/administração & dosagem , Desoxiguanosina/análogos & derivados , Desoxiguanosina/toxicidade , Humanos , Metiltransferases/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , O(6)-Metilguanina-DNA Metiltransferase , Transplante Heterólogo
14.
Carcinogenesis ; 17(2): 303-9, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8625455

RESUMO

Ductal adenocarcinoma, the most common form of pancreatic cancer in humans, is associated with activation of the K-ras oncogene in approximately 90% of cases. In contrast, K-ras mutations are found in < 50% of the relatively rare intraductal papillary mucinous tumor (IPMT), which arises in the main pancreatic ducts. Since both adenocarcinomas and IPMTs are believed to arise from ductal cells and progress through similar sequences of morphological changes (i.e. flat hyperplasia, papillary hyperplasia, atypia and carcinoma in situ), it is clear that such progression may not always necessitate activation of the ras oncogene. Experimentally ductal adenocarcinomas of the pancreas can be induced in the hamster model by a brief treatment with N-nitroso(2-hydroxypropyl)(2-oxopropyl)amine (HPOP), while IPMTs can be induced by a combined treatment with HPOP and orotic acid (OA) in an initiation/promotion schedule. Since animals are exposed to the carcinogen only once, initiated normal epithelium is expected to give rise to a wide spectrum of neoplastic and preneoplastic lesions, progression of which will depend on the extent of mutagenesis induced at initiation in the targeted cells. In order to investigate the role of K-ras in progression of IPMTs as compared with adenocarcinomas we have examined the presence of K-ras mutations in the above two types of experimentally induced pancreatic cancers, as well as in associated and preneoplastic lesions. K-ras mutations at codons 12, 13 and 61 were determined by a designed restriction fragment length polymorphism method using mismatched nested primers in 77 neoplastic and preneoplastic foci microdissected from 20 pancreases. Mutations were found in all foci of atypical hyperplasia, in carcinomas in situ and invasive cancer, whether such lesions originated in lobular tissue or in the main pancreatic duct. Mutations were also found in papillary hyperplasia and flat hyperplasia in small ducts and also in the main duct at high frequency. With one exception, all ras mutations were G-->A transitions at the second base of codon 12. Mutations were occasionally accompanied by excessive presence of the mutant ras allele or loss of the wild-type ras allele, events that were more frequent in atypical hyperplasia (5/17), carcinomas in situ (5/14), IPMTs (2/5) and invasive adenocarcinomas (2/5) than in flat hyperplasia (0/6) or papillary hyperplasia (2/18). Our results demonstrate that: (i) K-ras mutations, predominantly G-->A transitions, are present in all experimentally induced hamster tumors; (ii) the incidence of K-ras mutations in IPMTs is lower in humans than in the hamster model; (iii) advanced lesions in both adenocarcinomas and IPMTs were frequently associated with an excess of the mutant over the wild-type K-ras allele. It is likely that both adenocarcinomas and IPMTs induced chemically in the hamster model arise by mechanisms which involve early activation of K-ras. Such a mechanism seems to be applicable only in a fraction of human IPMTs.


Assuntos
Adenocarcinoma Mucinoso/genética , Carcinoma Ductal de Mama/genética , Genes ras/efeitos dos fármacos , Mutação , Neoplasias Pancreáticas/genética , Adenocarcinoma Mucinoso/induzido quimicamente , Animais , Sequência de Bases , Carcinoma Ductal de Mama/induzido quimicamente , Cricetinae , Feminino , Genes ras/genética , Neoplasias Hepáticas Experimentais/induzido quimicamente , Mesocricetus , Dados de Sequência Molecular , Neoplasias Pancreáticas/induzido quimicamente , Polimorfismo de Fragmento de Restrição
15.
Chem Res Toxicol ; 7(6): 762-9, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-7696530

RESUMO

O6-Benzyl-2'-deoxyguanosine is a potential antitumor drug modulator that is intended to reduce or eliminate O6-alkylguanine-DNA alkyltransferase activity in tumors prior to treatment with genotoxic chemotherapeutic alkylating agents. The rationale for using this compound instead of the more active O6-benzylguanine and its substituted benzyl derivatives at the benzyl ring is its greater solubility in aqueous media and potential pharmacologic advantage. Metabolism and disposition of O6-benzyl-2'-deoxyguanosine was determined in adult male Sprague-Dawley rats following an ip injection of 100 mg/kg. Under these conditions, the compound was partially metabolized to yield a glucuronic acid conjugate, which was secreted exclusively in the bile. Removal of the 2'-deoxyribose or the benzyl group to yield O6-benzylguanine and 2'-deoxyguanosine, respectively, occurred to a lesser extent. Metabolism accounted for the clearance of at least 58% of the total dose and took place primarily in the liver. Direct excretion of unchanged drug, mainly in urine, accounted for the remainder of the dose. Analysis of venous blood showed the presence of O6-benzyl-2'-deoxyguanosine and O6-benzylguanine at concentrations which are considered to be effective in depleting alkyltransferase activity. Levels of the nucleoside reached a maximum of 45 microM at 2 h, while those of O6-benzylguanine peaked to 20 microM at 4 h and remained at that level for at least 4 more hours. Transport of O6-benzyl-2'-deoxyguanosine in C6 glioma cells increased linearly with the extracellular concentration of the drug up to 600 microM. Intracellular levels of the drug reached 1.2 pmol per microM of extracellular compound per 10(6) cells as soon as 30 s after exposure and remained as high for at least 1 h. Such levels indicate that entrapment of the nucleoside inside cells by either phosphorylation or other means is probably not an important feature for this drug.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Desoxiguanosina/análogos & derivados , Animais , Bile/metabolismo , Cromatografia Líquida de Alta Pressão , Desoxiguanosina/metabolismo , Injeções Intraperitoneais , Masculino , Ratos , Ratos Sprague-Dawley
16.
Cancer Res ; 54(20): 5324-32, 1994 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-7923160

RESUMO

The effect of dietary orotic acid (OA) in liver-pancreas carcinogenesis induced in female Syrian hamsters by N-Nitroso(2-hydroxypropyl) (2-oxopropyl)amine (HPOP) was evaluated. All animals infused with the carcinogen received the same doses. Results of the control group which received no OA or carcinogen were compared with the results of: (a) hamsters treated with HPOP and fed a regular 20% protein synthetic diet (group 1); (b) hamsters fed the OA diet for a brief time period during initiation with the carcinogen (group 2); and (c) hamsters in which OA was administered after carcinogen infusion for life (group 3). All animals of the control group were normal at autopsy, while those in group 1 (HPOP alone) revealed the spectrum of lesions accepted as classical in the multistep hyperplasia-dysplasia-carcinoma in situ (CIS) sequence of carcinogenesis. Results of group 2, in light of group 1, revealed an increased incidence of the following lesions in the common pancreatic duct: dilatation, 2.5 times; flat and papillary hyperplasia, 2 times; and dysplasia (atypical hyperplasia), 12 times. No significant increase of CIS and invasive cancer in the body and tail of the pancreas was observed; in addition, the incidence, nature, and location of pancreatic adenocarcinomas were not affected. Yet, the effect of OA administered after carcinogen infusion (group 3) when compared to group 1 seemed to enhance a further increase in the incidence of practically all lesions throughout the pancreas. An obvious overall step-up incidence along the multistep hyperplasia-dysplasia-CIS-invasive cancer process in the pancreas was observed. The increase in incidence of flat, papillary, and atypia of the epithelium of the common pancreatic duct in group 3 was mild compared to that found in the same duct of group 2, but the increase in incidence of these same three lesions when found in the main ducts was marked: flat hyperplasia, 3-fold; papillary hyperplasia, 2.5-fold; atypical hyperplasia, 3-fold. The increase in incidence of CIS in this group was 5-fold and papillary adenocarcinomas, 3-fold, when compared to 5% found in groups 1 and 2. Hepatic malignancies (cholangiocarcinomas) occurred in 6% of the cases in group 3 compared to none in group 2; the incidence of malignancy in the gallbladder was the same in groups 2 and 3 but three times greater than that in group 1.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Carcinógenos , Colangiocarcinoma/induzido quimicamente , Neoplasias da Vesícula Biliar/induzido quimicamente , Neoplasias Hepáticas Experimentais/induzido quimicamente , Nitrosaminas , Ácido Orótico , Neoplasias Pancreáticas/induzido quimicamente , Lesões Pré-Cancerosas/induzido quimicamente , Animais , Colangiocarcinoma/patologia , Cricetinae , Sinergismo Farmacológico , Feminino , Neoplasias da Vesícula Biliar/patologia , Hiperplasia/induzido quimicamente , Neoplasias Hepáticas Experimentais/patologia , Mesocricetus , Pâncreas/efeitos dos fármacos , Pâncreas/patologia , Neoplasias Pancreáticas/patologia , Lesões Pré-Cancerosas/patologia
17.
Carcinogenesis ; 14(8): 1705-9, 1993 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8353856

RESUMO

Metabolism of 14C labeled N-nitrosobis(2-oxopropyl)amine (BOP), N-nitroso(2-hydroxypropyl)(2-oxopropyl)amine (HPOP) and N-nitrosobis(2-hydroxypropyl)amine (BHP) by pancreatic duct cells in culture involves the following two pathways: reduction or oxidation reactions at the beta-carbon which result in the inter-conversion of these nitrosamines and activation reactions which result in the decomposition of the nitrosamine, the evolution of 14CO2 and the labeling of macromolecules. Reduction of BOP to HPOP seems to contribute significantly to the metabolism of the former nitrosamine by pancreatic duct cells, however, redox reactions at the beta-carbon of HPOP or BHP are not extensive. In terms of DNA damage, all three nitrosamines yield methyl and hydroxypropyl adducts. As expected, HPOP and BHP yield higher levels of O6-hydroxypropylguanine than BOP, while the latter yields higher levels of O6-methylguanine. There is no correlation between the ability of these nitrosamines to alkylate duct cell DNA in vitro and their carcinogenic potency in vivo. Concentrations of DNA adducts induced by pancreas specific nitrosamines (PSNs) in cultured duct cells at concentrations comparable to those found in the pancreatic juice of animals treated with BOP, are almost an order of magnitude lower than those induced in the pancreas of such animals. Discrepancies between in vitro and in vivo formation of active metabolites and DNA adducts may be attributed to the decline of the cells' ability to activate PSNs during culturing. In the same vein, the ductal cell may not be the main source of active metabolites targeting its DNA in the animal model.


Assuntos
Carcinógenos/farmacocinética , Nitrosaminas/farmacocinética , Pâncreas/metabolismo , Animais , Biotransformação , Radioisótopos de Carbono , Carcinógenos/metabolismo , Transformação Celular Neoplásica/efeitos dos fármacos , Células Cultivadas , Cricetinae , DNA/efeitos dos fármacos , Dano ao DNA , Masculino , Mesocricetus , Modelos Biológicos , Mutação , Nitrosaminas/metabolismo , Nitrosaminas/toxicidade , Pâncreas/citologia
18.
Cancer Res ; 53(12): 2790-5, 1993 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-8504421

RESUMO

N-Nitroso(2-hydroxypropyl)(2-oxopropyl)amine (HPOP) is a complete pancreatic carcinogen in female hamsters at a dose of 210 mg/kg given via an Alzet 2001 pump implanted s.c. Ultimate carcinogenic metabolites of HPOP target DNA to yield 7-and O6-methylguanines (7-mGua, O6-mGua) and 7- and O6-hydroxypropylguanines (7-HpGua, O6-HpGua). During continuous administration of HPOP, levels of DNA alkylation increase linearly with time of exposure and reach a maximum at the end of treatment. Such levels are markedly lower in pancreas or in its component duct or acinar cells than in liver or in other extrahepatic organs examined, indicating that the organotropy of HPOP does not directly correlate to its extent of activation by various tissues. After continuous treatment with HPOP, all major DNA adducts with the exception of 7-mGua in liver are repaired at rates slower than those measured after a single injection of the carcinogen. Half-lives for the repair of O6-mGua are 240 h in liver and considerably longer in extrahepatic tissues. Half-lives for the removal of 7-mGua are 46, 55, 72, and 96 h in liver, kidney, lung, and pancreas; while respective values for 7-HpGua are 216, 216, 132, and 140 h. No significant repair is observed for O6-HpGua for at least 8 days. The above differences in half-lives result in the gradual increase of 7-HpGua and O6-HpGua relative to their methyl counterparts. DNA synthesis progressively increases during HPOP infusion in all the tissues examined, and reaches maximum levels 3 to 4 days after termination of treatment. In pancreas, such levels are up to 5 times greater in HPOP treated animals than in controls. The increase in DNA synthesis during carcinogen treatment is due to the greater number of cells entering S phase rather than to an increase in the rate of proliferation of a certain population of cells. The mitogenic effect of HPOP in the pancreas and the persistence of highly promutagenic hydroxypropyl adducts are postulated to contribute to the initiation of pancreatic cancer in the hamster model.


Assuntos
Carcinógenos/metabolismo , Dano ao DNA , Reparo do DNA , DNA/biossíntese , Nitrosaminas/metabolismo , Neoplasias Pancreáticas/induzido quimicamente , Alquilação , Animais , Divisão Celular/efeitos dos fármacos , Cricetinae , DNA/efeitos dos fármacos , Feminino , Guanina/análogos & derivados , Guanina/metabolismo , Mesocricetus
19.
Carcinogenesis ; 13(5): 759-65, 1992 May.
Artigo em Inglês | MEDLINE | ID: mdl-1586988

RESUMO

Levels of methyl and hydroxypropyl adducts induced by single s.c. injections of various doses of tritium-labeled N-nitrosobis(2-hydroxypropyl)amine ([1-3H]BHP) were determined in the liver, pancreas, kidney and lung of hamsters and rats. At doses of BHP used in carcinogenesis studies (100-500 mg/kg), methylation of DNA was more extensive than its hydroxypropylation; however, it did not increase proportionally with the dose and gradually became secondary to hydroxypropylation at higher doses of the carcinogen. Ratios of hydroxypropyl versus methyl adducts also varied significantly depending on the tissue and species. In both species ratios of N7-hydroxypropylguanine (N7-HpG) versus N7-methylguanine (N7-MeG) were greater in kidney and pancreas than in liver or lung. Due to apparent differences in the repair of O6-methylguanine (O6-MeG) and O6-hydroxypropylguanine (O6-HpG), and the propensity of 2-hydroxypropylating as compared to methylating agents to yield a greater percentage of oxygen adducts, ratios of O6-HpG versus O6-MeG were markedly greater than those of N7-HpG versus N7-MeG. Levels of O6-HpG were greater than those of O6-MeG in rat liver, pancreas and kidney and also in hamster kidney, while such levels were similar in rat lung and also in hamster liver, pancreas and lung. Like N-nitrosobis(2-oxopropyl)amine (BOP) and N-nitroso(2-hydroxypropyl) (2-oxopropyl)amine (HPOP), BHP was activated primarily in the liver and induced substantially greater DNA damage in this than in any other tissue examined. However, unlike BOP and HPOP, which induced similar levels of hepatic DNA damage in the above two species, BHP methylated and hydroxypropylated hamster liver DNA more extensively than that of the rat. Differences between BOP and BHP were also observed regarding levels and distribution of DNA adducts in extrahepatic tissues. In rats, BHP induced greater levels of methylation and hydroxypropylation in lung than in kidney, while the reverse was observed with BOP. Apparently reduction of the beta-carbon of pancreas-specific nitrosamine carcinogens results in a shift of alkylation from kidney to the lung. Excretion of HPOP in the urine of BHP-treated animals and the observed saturation of DNA methylation at high doses of BHP, supported the hypothesis that the BHP-induced methylation of DNA proceeded via the intermediate formation of HPOP. This was further supported by the observation that both excretion of HPOP and levels of methyl adducts were greater in hamsters than in rats.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Carcinógenos/metabolismo , DNA/metabolismo , Nitrosaminas/metabolismo , Alquilação , Animais , Cricetinae , Relação Dose-Resposta a Droga , Guanina/análogos & derivados , Guanina/metabolismo , Rim/metabolismo , Fígado/metabolismo , Pulmão/metabolismo , Mesocricetus , Metilação , Nitrosaminas/urina , Pâncreas/metabolismo , Ratos , Ratos Endogâmicos , Especificidade da Espécie
20.
Carcinogenesis ; 12(2): 181-6, 1991 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-1995183

RESUMO

The effect of orotic acid (OA) on the carcinogenicity of N-nitroso(2-hydroxypropyl)(2-oxopropyl)amine (HPOP) in rats was evaluated. A group of 5 week old Sprague-Dawley male rats were placed on a synthetic 20% protein diet containing 1% OA. A second group was placed on a regular, OA-free diet of similar composition. Approximately 2 weeks later, animals from both groups grown to 100 g were treated with 400 mg/kg HPOP delivered continuously for 14 days via 2002 Alzet osmotic pumps implanted s.c. Rats fed the OA diet were kept under this diet for 13 weeks following initiation of HPOP treatment and subsequently were placed on the regular diet for another 12 weeks, at which time they were killed. In the absence of OA, HPOP-treated rats developed adenomas in the kidney and lungs at incidences of 5 and 33% respectively, while pancreas and liver were unaffected. On the other hand, rats fed the OA diet and treated with HPOP developed renal mesenchymal tumors and pulmonary adenomas at incidences of 70 and 65% respectively. In addition, HPOP induced cystic lesions in the pancreas of animals fed the OA diet. The enhancement of the tumorigenic effectiveness of HPOP was at least partly ascribed to the effect of OA treatment on the rate by which carcinogen-induced alkylation of DNA was repaired in various tissues. Accumulation of N7-methylguanine in kidney, lung and pancreas of rats fed the OA diet was 1.6, 1.9 and 2.4 times higher than in respective organs of animals fed the regular diet. Similarly, concentrations of the premutagenic O6-methylguanine (O6-MeG) were 3.0, 3.1 and 2 times greater in the kidney, lung and pancreas of rats fed the OA diet than in the respective organs of those fed the regular diet. Feeding an OA diet to HPOP-treated rats did not have an effect on either the resistance of the liver to this carcinogen or on the level of O6-MeG accumulation in the DNA of this tissue.


Assuntos
Carcinógenos/toxicidade , Nitrosaminas/toxicidade , Ácido Orótico/farmacologia , Animais , DNA/metabolismo , Sinergismo Farmacológico , Rim/efeitos dos fármacos , Neoplasias Renais/induzido quimicamente , Fígado/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Neoplasias Pulmonares/induzido quimicamente , Neoplasias Pulmonares/patologia , Masculino , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Neoplasias Pancreáticas/induzido quimicamente , Ratos , Ratos Endogâmicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...