Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Trends Parasitol ; 26(3): 125-9, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20056487

RESUMO

The emergence of artemisinin resistance could adversely impact the current strategy for malaria treatment; thus, new drugs are urgently needed. A possible approach to developing new antimalarials is to find new uses for old drugs. Some anticancer agents such as methotrexate and trimetrexate are active against malaria. However, they are commonly perceived to be toxic and thus not suitable for malaria treatment. In this opinion article, we examine how the toxicity of anticancer agents is just a matter of dose or 'only dose makes the poison', as coined in Paracelsus' law. Thus, the opportunity exists to discover new antimalarials using the anticancer pharmacopoeia.


Assuntos
Antineoplásicos/uso terapêutico , Malária/tratamento farmacológico , Antimaláricos/uso terapêutico , Antineoplásicos/toxicidade , Humanos , Metotrexato/uso terapêutico , Metotrexato/toxicidade , Trimetrexato/uso terapêutico , Trimetrexato/toxicidade
2.
Biochem Pharmacol ; 57(5): 567-77, 1999 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-9952321

RESUMO

Folic acid (PteGlu)-enhanced intense synergy has been observed between nonpolyglutamylatable dihydrofolate reductase (DHFR) inhibitors and polyglutamylatable inhibitors of other folate-requiring enzymes, such as glycinamide ribonucleotide formyltransferase (GARFT) and thymidylate synthase. Since this phenomenon is potentially therapeutically useful, we explored its universality by examining the combined action of a DHFR inhibitor, trimetrexate (TMQ), with a GARFT inhibitor, 4-[2-(2-amino-4-oxo-4,6,7,8-tetrahydro-3H-pyrimidino[5,4,6][1,4]++ +thiazin-6-yl)-(S)-ethyl]-2,5-thienoylamino-L-glutamic acid (AG2034), in eight human cultured cell lines. Using a 96-well plate cell growth inhibition assay, four ileocecal adenocarcinoma cell lines [HCT-8, HCT-8/DW2 (Tomudex-resistant), HCT-8/DF2 (Tomudex-/FdUrd-resistant), and HCT-8/50 (adapted to 50 nM PteGlu)], three head and neck carcinoma cell lines [A253, FaDu, and Hep-2/500 (FdUrd-resistant)], and a non-small cell lung carcinoma cell line [H460] were treated for 96 hr with TMQ + AG2034 in the presence of 23 or 40 microM PteGlu. Cell growth was measured with the sulforhodamine B assay at the end of this period. Drug interactions were assessed by fitting a 7-parameter model including a synergism parameter, alpha, to data with weighted nonlinear regression. Isobologram analysis was also applied. At 23 microM PteGlu, cells exhibited similar intensities of Loewe synergy for the combination of TMQ + AG2034. Loewe synergy was abolished in HCT-8/50 cells cultured and studied in 50 nM PteGlu. At 40 microM PteGlu, the intensity of the combined action in all cell lines was increased However, the most intense Loewe synergy was seen with HCT-8, HCT-8/DF2, H460, FaDu, A253, and Hep-2/500 cells, whereas the HCT-8/50 subculture showed less of the phenomenon, and PteGlu enhancement was the least with HCT-8/DW2, a subline deficient in folylpolyglutamate synthetase (FPGS). The universality of the PteGlu-enhanced intense synergy phenomenon is suggested. Impaired FPGS activity and low-folate adaptation prior to treatment significantly lessen the degree of PteGlu enhancement.


Assuntos
Antineoplásicos/toxicidade , Antagonistas do Ácido Fólico/toxicidade , Ácido Fólico/farmacologia , Glutamatos/toxicidade , Hidroximetil e Formil Transferases/antagonistas & inibidores , Pirimidinas/toxicidade , Trimetrexato/toxicidade , Adenocarcinoma , Carcinoma de Células Escamosas , Divisão Celular/efeitos dos fármacos , Neoplasias do Colo , Resistência a Múltiplos Medicamentos , Sinergismo Farmacológico , Neoplasias de Cabeça e Pescoço , Humanos , Cinética , Neoplasias Pulmonares , Fosforribosilglicinamido Formiltransferase , Células Tumorais Cultivadas
3.
Blood ; 90(9): 3546-54, 1997 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9345038

RESUMO

Antifolates such as methotrexate (MTX) and trimetrexate (TMTX) are widely used in the treatment of cancer and nonmalignant disorders. Transient, yet sometimes severe myelosuppression is an important limitation to the use of these drugs. It has previously been shown that clonogenic myeloid progenitors and colony-forming units-spleen are resistant to antifolates, suggesting that myelotoxicity occurs late in hematopoietic development. The goal of this study was to define the mechanisms by which primitive hematopoietic cells resist the toxic effects of antifolate drugs. To test the hypothesis that myeloid progenitors may salvage extracellular nucleotide precursors to resist TMTX toxicity, a defined liquid culture system was developed to measure TMTX toxicity in expanding progenitor populations. These in vitro experiments showed that both human and murine progenitors can resist TMTX toxicity by importing thymidine and hypoxanthine from the serum. As predicted from these findings, several drugs that block thymidine transport sensitized progenitors to TMTX in vitro, although to differing degrees. These nucleoside transport inhibitors were used to test whether progenitors and hematopoietic stem cells (HSCs) could be sensitized to TMTX in vivo. Treatment of mice with TMTX and nitrobenzylmercaptopurineriboside phosphate (NBMPR-P), a potent transport inhibitor, caused significant depletions of clonogenic progenitors within the bone marrow (20-fold) and spleen (6-fold). Furthermore, NBMPR-P administration dramatically sensitized HSCs to TMTX, with dual-treated mice showing a greater than 90% reduction in bone marrow repopulating activity. These studies demonstrate that both myeloid progenitor cells and HSCs resist TMTX by using nucleotide salvage mechanisms and that these pathways can be pharmacologically blocked in vivo using nucleoside transport inhibitors. These results have important implications regarding the use of transport inhibitors for cancer therapy and for using variants of dihydrofolate reductase for in vivo selection of genetically modified HSCs.


Assuntos
Antimetabólitos Antineoplásicos/toxicidade , Resistencia a Medicamentos Antineoplásicos/fisiologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/fisiologia , Tioinosina/análogos & derivados , Tionucleotídeos/farmacologia , Timidina/metabolismo , Trimetrexato/toxicidade , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Morte Celular/efeitos dos fármacos , Feminino , Células-Tronco Hematopoéticas/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Tioinosina/farmacologia
4.
Jpn J Cancer Res ; 88(9): 900-6, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9369939

RESUMO

Cytotoxicity of trimetrexate (TMQ), a lipophilic dihydrofolate reductase inhibitor, was examined in antifolate-resistant human T-cell leukemia cell lines developed in oxidized or reduced folate. An approximately 60-fold methotrexate (MTX)-resistant subline was developed in oxidized folate (pteroylglutamic acid: PGA) (CCRF-CEM/MTX60-PGA) from human T-cell leukemia cell line CCRF-CEM; this line exhibited impaired membrane transport of the drug. Further enhancement of MTX resistance resulted in selection of an approximately 5000-fold MTX-resistant subline (CCRF-CEM/ MTX5000-PGA), which showed increased dihydrofolate reductase activity due to gene amplification in addition to further impairment of MTX transport. An approximately 140-fold MTX-resistant subline, and then a 1500-fold MTX-resistant subline were developed in reduced folate (10 nM leucovorin) (CCRF-CEM/MTX140-LV and CCRF-CEM/MTX1500-LV); they exhibited increased dihydrofolate reductase due to gene amplification accompanied by increased intracellular drug accumulation of MTX. While CCRF-CEM/MTX140-LV and CCRF-CEM/MTX1500-LV cells showed cross-resistance to TMQ, CCRF-CEM/MTX60-PGA and CCRF-CEM/MTX5000-PGA cells were at least as sensitive to TMQ as the parent cells. TMQ was more potent against approximately 200-fold N10-propargyl-5,8-dideazafolic-acid (CB3717)-resistant human T-cell leukemia MOLT-3 sublines developed in PGA (MOLT-3/CB3717(200)-PGA) or leucovorin (MOLT-3/CB3717(200)-LV), as compared to the parent cells; MOLT-3/CB3717(200)-PGA and MOLT-3/CB3717(200)-LV cells were resistant to CB3717 by virtue of impaired transport, only the former possessing gene amplification of thymidylate synthase. The cytotoxicity of TMQ in both MOLT-3/CB3717(200)-PGA and MOLT-3/CB3717(200)-LV cells was reduced by addition of leucovorin in a dose-dependent manner, suggesting intracellular folate deficiency as a cause of TMQ sensitivity. These results demonstrate that TMQ overcomes transport-impaired antifolate resistance, irrespective of gene amplification of dihydrofolate reductase or thymidylate synthase. Types of folate used during the development of antifolate resistance seem to be important in relation to the mechanism of TMQ responsiveness as well as that of antifolate resistance.


Assuntos
Antagonistas do Ácido Fólico/toxicidade , Ácido Fólico/metabolismo , Metotrexato/toxicidade , Trimetrexato/toxicidade , Antimetabólitos Antineoplásicos/farmacocinética , Antimetabólitos Antineoplásicos/toxicidade , Transporte Biológico , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Antagonistas do Ácido Fólico/farmacocinética , Humanos , Leucemia de Células T , Oxirredução , Trimetrexato/farmacocinética , Células Tumorais Cultivadas
5.
Anticancer Res ; 17(6D): 4519-24, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9494561

RESUMO

The synergistic effect of two different inhibitors of thymidylate synthase-FdUrd and sulphonamide derivatives on murine leukemia cells-5178Y (parental subline) and 5178Y/F (its fluorodeoxyuridine resistant subline) in culture was examined. Upon the exposure of cultures from both lines to a slightly inhibitory concentration of FdUrd (1 nM) in combination with 2-desamino-2-methyl-10-propargyl-5,8-dideaza-pteroylsulphogluta mine or -glycine a synergistic effect of antimetabolites on cell growth was observed. This was accompanied by a marked reduction in intracellular concentration in both cell lines of 5,10CH2H4PteGlu; the intracellular concentration of 5,10CH2H4PteGlu(n) in the resistant subline was 3 times higher than in parental line. The inhibitory effect of combined drugs on the cellular pool of 5178Y of the two antimetabolites also depends on the sequence of their addition; however in the FdUrd resistant cell-line the dependence on the sequence of the addition was not observed. The results obtained strongly suggest that under certain conditions inhibition of thymidylate synthesis by antifolates is intensified by proprior use of FdUrd.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Floxuridina/toxicidade , Antagonistas do Ácido Fólico/toxicidade , Leucemia L5178/patologia , Timidilato Sintase/antagonistas & inibidores , Animais , Bovinos , Divisão Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Floxuridina/farmacocinética , Ácido Fólico/análogos & derivados , Ácido Fólico/toxicidade , Antagonistas do Ácido Fólico/farmacocinética , Fígado/enzimologia , Metotrexato/toxicidade , Camundongos , Tetra-Hidrofolato Desidrogenase/metabolismo , Trimetrexato/toxicidade , Células Tumorais Cultivadas
6.
Cancer Lett ; 88(1): 93-9, 1995 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-7850779

RESUMO

Various alterations of the dihydrofolate reductase (DHFR) gene are involved in resistance. In order to understand the mechanism that induce such gene alterations in human leukemia cells, we studied the expression products of DHFR gene in trimetrexate (TMQ)- and/or methotrexate (MTX)-resistant sublines derived from a MOLT-3 human leukemia cell line. A 200-fold TMQ-resistant subline (MOLT-3/TMQ200) expressed the mutated DHFR mRNA, with a base change (T-->C) at the second position of codon 31, as well as the wild type gene. A MTX-resistant subline derived from MOLT-3/TMQ200 (MOLT-3/TMQ200-MTX500) showed a further increase in the expression of the mutated DHFR mRNA, compared to MOLT-3/TMQ200, with a marked decrease of expression of the wild type DHFR mRNA, which is confirmation of amplification of the mutated DHFR gene. By contrast, a 10,000-fold MTX-resistant subline (MOLT-3/MTX10,000) over-expressed the wild type DHFR mRNA, which is confirmation of amplification of the wild type gene. Increased levels of the DHFR enzyme in these sublines were proportional to expression levels of the DHFR mRNA. The DHFR enzyme expressed in MOLT-3/TMQ200-MTX500 cells showed a 40-fold increase in the Ki values for both MTX and TMQ, compared with values for the wild type DHFR expressed in both MOLT-3/MTX10,000 and its parent cell line. These findings suggest that the altered DHFR gene, which was introduced in MOLT-3 cells by exposure to TMQ, gave rise to a variant enzyme with reduced affinity to antifolates, and that complex DHFR alterations confer drug-resistant phenotypes in antifolate-resistance. Structural difference between the antifolates could be important in the introduction of the differential DHFR gene alterations in the antifolate resistance.


Assuntos
Antagonistas do Ácido Fólico/metabolismo , Tetra-Hidrofolato Desidrogenase/metabolismo , Trimetrexato/metabolismo , Sequência de Bases , Primers do DNA/química , Resistência a Medicamentos , Humanos , Técnicas In Vitro , Cinética , Leucemia de Células T , Metotrexato/farmacologia , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Neoplásico/genética , Trimetrexato/toxicidade , Células Tumorais Cultivadas
7.
Cancer Chemother Pharmacol ; 36(2): 165-71, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-7767954

RESUMO

Antifolates have been shown to increase the DNA strand breaks produced by the topoisomerase inhibitor etoposide. PT523 is a potent new antifolate that cannot be polyglutamated. Human SCC-25 squamous carcinoma cells were exposed to methotrexate, trimetrexate or PT523 at a concentration of 5 microM for 24 h along with various concentrations of etoposide or novobiocin during the final 2 h. Isobologram analysis of the treatment combinations indicated that exposure of the cells to PT523/etoposide, methotrexate/etoposide, PT523/novobiocin, methotrexate/novobiocin and trimetrexate/novobiocin resulted in greater than additive cytotoxicity. DNA alkaline elution studies with the same drug combinations indicated that there were three- to four-fold increases in the radiation equivalent (rad equivalent) strand breaks in the cellular DNA with etoposide or novobiocin along with the antifolate compared with the topoisomerase II inhibitors alone. Tumor growth delay studies were carried out in the murine SCC VII squamous carcinoma. PT523 (0.5 mg/kg) and methotrexate (2 mg/kg) were administered by 7-day continuous infusion while trimetrexate (3.75 mg/kg) was administered intraperitoneally daily on days 7-9. Etoposide (10 mg/kg) and novobiocin (100 mg/kg) were administered intraperitoneally on alternate days (7, 9, 11). The combinations of PT523 with etoposide or novobiocin were significantly more effective than methotrexate and etoposide or novobiocin, producing tumor growth delays of 8.4 days and 6.9 days, respectively. Overall, the antifolate/topoisomerase II inhibitor treatment combinations produced tumor growth delays that were apparently additive to greater than additive.


Assuntos
Antineoplásicos/toxicidade , Carcinoma de Células Escamosas/tratamento farmacológico , Dano ao DNA , Antagonistas do Ácido Fólico/toxicidade , Ornitina/análogos & derivados , Pterinas/toxicidade , Pterinas/uso terapêutico , Inibidores da Topoisomerase II , Animais , Antineoplásicos/uso terapêutico , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , DNA de Neoplasias/efeitos dos fármacos , Sinergismo Farmacológico , Etoposídeo/uso terapêutico , Etoposídeo/toxicidade , Antagonistas do Ácido Fólico/uso terapêutico , Neoplasias de Cabeça e Pescoço , Humanos , Masculino , Metotrexato/uso terapêutico , Metotrexato/toxicidade , Camundongos , Camundongos Endogâmicos C3H , Novobiocina/uso terapêutico , Novobiocina/toxicidade , Ornitina/uso terapêutico , Ornitina/toxicidade , Trimetrexato/uso terapêutico , Trimetrexato/toxicidade , Células Tumorais Cultivadas
8.
Br J Cancer ; 70(6): 1075-84, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7981057

RESUMO

We examined the cytotoxicity and biochemical effects of the lipophilic antifol trimetrexate (TMQ) in two human colon carcinoma cell lines, SNU-C4 and NCI-H630, with different inherent sensitivity to TMQ. While a 24 h exposure to 0.1 microM TMQ inhibited cell growth by 50-60% in both cell lines, it did not reduce clonogenic survival. A 24 h exposure to 1 and 10 microM TMQ produced 42% and 50% lethality in C4 cells, but did not affect H630 cells. Dihydrofolate reductase (DHFR) and thymidylate synthase were quantitatively and qualitatively similar in both lines. During drug exposure, DHFR catalytic activity was inhibited by > or = 85% in both cell lines; in addition, the reduction in apparent free DHFR binding capacity (< or = 20% of control), depletion of dTTP, ATP and GTP pools and inhibition of [6-3H]deoxyuridine incorporation into DNA were similar in C4 and H630 cells. TMQ produced a more striking alteration of the pH step alkaline elution profile of newly synthesised DNA in C4 cells compared with 630 cells, however, indicating greater interference with DNA chain elongation or more extensive DNA damage. When TMQ was removed after a 24 h exposure to 0.1 microM, recovery of DHFR catalytic activity and apparent free DHFR binding sites was evident over the next 24-48 h in both cell lines. With 1 and 10 microM, however, persistent inhibition of DHFR was evident in C4 cells, whereas DHFR recovered in H630 cells. These data suggest that, although DHFR inhibition during TMQ exposure produced growth inhibition, DHFR catalytic activity 48 h after drug removal was a more accurate predictor of lethality in these two cell lines. Several factors appeared to influence the duration of DHFR inhibition after drug removal, including initial TMQ concentration, declining cytosolic TMQ levels after drug removal, the ability to acutely increase total DHFR content and the extent of TMQ-mediated DNA damage. The greater sensitivity of C4 cells to TMQ-associated lethality may be attributed to the greater extent of TMQ-mediated DNA damage and more prolonged duration of DHFR inhibition after drug exposure.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Trimetrexato/toxicidade , Transporte Biológico , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , DNA de Neoplasias/biossíntese , Humanos , Técnicas In Vitro , Tetra-Hidrofolato Desidrogenase/metabolismo , Timidilato Sintase/metabolismo , Nucleotídeos de Timina/metabolismo , Fatores de Tempo , Trimetrexato/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos
9.
Mutat Res ; 304(2): 243-60, 1994 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-7506368

RESUMO

We have studied cell lines of rodent and human origin for their propensity to become resistant to antifolates (methotrexate, trimetrexate), phosphonacetyl-L-aspartate (PALA), and colcemid, resistances associated with amplification of the DHFR, CAD, and MDR1 genes, respectively. We have employed two different methods: (1) a shallow step-wise selection protocol, where time to attain specified resistance is the quantitative measure, (2) the frequency of resistant colonies at specified drug concentrations. Although there are advantages and disadvantages to both methods, the two methods gave the same relative ranking of cell lines. Striking differences in the propensity for gene amplification (resistance) were found: human cell lines were less prone to amplify genes than Chinese hamster ovary (CHO) cells. This ranking was similar with all of the agents employed. Additionally, we observed that whereas PALA resistance in CHO cells is associated with amplification of the CAD gene, PALA resistance in the two human cell lines studied (HeLaS3 and VA13) was not associated with amplification and/or overexpression of the CAD gene, and thus this resistance to PALA occurs by an unknown mechanism.


Assuntos
Antimetabólitos Antineoplásicos/toxicidade , Aspartato Carbamoiltransferase/genética , Ácido Aspártico/análogos & derivados , Carbamoil Fosfato Sintase (Glutamina-Hidrolizante)/genética , Di-Hidro-Orotase/genética , Resistência a Medicamentos/genética , Amplificação de Genes , Complexos Multienzimáticos/genética , Ácido Fosfonoacéticos/análogos & derivados , Animais , Ácido Aspártico/toxicidade , Células CHO , Linhagem Celular Transformada , Cricetinae , Demecolcina/toxicidade , Relação Dose-Resposta a Droga , Fibroblastos/efeitos dos fármacos , Antagonistas do Ácido Fólico/toxicidade , Células HeLa , Humanos , Dose Letal Mediana , Metotrexato/toxicidade , Ácido Fosfonoacéticos/toxicidade , RNA Mensageiro/análise , Projetos de Pesquisa , Tetra-Hidrofolato Desidrogenase/genética , Trimetrexato/toxicidade
10.
Fundam Appl Toxicol ; 21(2): 244-52, 1993 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8405788

RESUMO

Repeated high doses of trimetrexate (TMX), a potent non-classical antifolate, have been administered as an experimental treatment for life-threatening Pneumocystis carinii infections in man. This therapy includes the coadministration of leucovorin, a reduced folate cofactor, to prevent antifolate toxicity in the host. The purpose of this investigation was to assess possible toxicologic sequelae of this combination regimen in an animal model. TMX at daily oral doses of 25, 35, and 45 mg/kg produced dose-related myelosuppression, thymic lymphoid depletion, seminiferous tubular atrophy, and degenerative lesions of the gastrointestinal tract. Mortality observed with TMX alone occurred earlier at higher doses and was specifically associated with severe degenerative enteropathy of the cecum. Oral leucovorin doses of 1, 5, 20, or 50 mg/kg administered twice daily, at the time of TMX administration and 6 hr later, protected against TMX lethality and target organ toxicity in a dose-related manner. Leucovorin was only partially protective against TMX-induced macrocytic anemia and the degree of protection was not dose-related. Leucovorin protection against cecal enteropathy was associated with increased DNA synthetic rates and higher mitotic activity of cecal epithelium than those in rats administered TMX alone. Importantly, the combination of daily administration of high dose TMX for 4 weeks with protective coadministration of leucovorin did not result in target organ toxicities that differed from TMX alone.


Assuntos
Leucovorina/farmacologia , Trimetrexato/antagonistas & inibidores , Administração Oral , Animais , Contagem de Células Sanguíneas , Medula Óssea/efeitos dos fármacos , Sistema Digestório/efeitos dos fármacos , Leucovorina/administração & dosagem , Tecido Linfoide/efeitos dos fármacos , Masculino , Ratos , Ratos Wistar , Testículo/efeitos dos fármacos , Trimetrexato/toxicidade
11.
Fundam Appl Toxicol ; 19(1): 6-14, 1992 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-1397803

RESUMO

Trimetrexate was administered to rats in an interrupted treatment regimen comparable to proposed human clinical treatment. Forty-five rats of each sex were dosed intravenously with trimetrexate at 0, 1, 10, or 30 mg/kg (0, 6, 60, or 180 mg/m2), once daily for 5 consecutive days, followed by a 23-day recovery period. This cycle of dosing and recovery was repeated for a total of six cycles. Hematology, urinalysis, clinical chemistry, and gross and microscopic pathology examinations were conducted for animals euthanatized 3 and 21 days after dosing cycles 1, 3, and 6. Additional rats in each group were maintained without dosing for an additional 56 days (77 days after the last trimetrexate dose) to assess the long-term reversibility of pathologic changes. Target organs were typical for an antifolate and included gastrointestinal tract, lymphoid tissues, and the hematopoietic and male reproductive systems. No toxicity was observed at 1 mg/kg. Treatment-related changes in hematology parameters following 10 and 30 mg/kg were fully reversible within 3 weeks of each dosing cycle. Except for testis and cecum, histopathological changes were also reversible within 21 days of dosing. Trimetrexate-induced testicular changes persisting during the course of multiple cycles of dosing were not reversible within 21 days, but required an additional 56 days for essentially complete recovery.


Assuntos
Trimetrexato/toxicidade , Animais , Peso Corporal/efeitos dos fármacos , Medula Óssea/efeitos dos fármacos , Células da Medula Óssea , Ceco/efeitos dos fármacos , Ceco/patologia , Relação Dose-Resposta a Droga , Esquema de Medicação , Ingestão de Alimentos/efeitos dos fármacos , Contagem de Eritrócitos/efeitos dos fármacos , Feminino , Contagem de Leucócitos/efeitos dos fármacos , Masculino , Necrose , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Wistar , Albumina Sérica/efeitos dos fármacos , Testículo/efeitos dos fármacos , Testículo/patologia , Fatores de Tempo
12.
Fundam Appl Toxicol ; 18(1): 115-25, 1992 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1534775

RESUMO

Trimetrexate is a nonclassical folate antagonist that is active against a number of experimental murine and human tumor cell lines. To assess its toxicity, rats were administered single or repeated (daily x5) doses by either the oral or the intravenous route. Oral doses were 0, 90, 180, 295, and 375 mg/kg (single dose) and 0, 32, 65, and 80 mg/kg (daily x5). Intravenous doses were 0, 6, 20, and 60 mg/kg (single dose) and 0, 10, 20, and 30 mg/kg (daily x5). In the oral studies, signs of toxicity first appeared 2 to 3 days after initiation of dosing. Clinical signs included hypoactivity, diarrhea, urine scald, rhinorrhea, emaciation, and death. Significant pathologic findings were degenerative enteropathy in small and large intestines, bone marrow hypocellularity, decreased WBCs (neutrophils, lymphocytes), generalized lymphoid depletion, and testicular tubular degeneration. Except for the testicular changes, these effects were most severe in animals dosed at 65 and 80 mg/kg in the oral x5 study (65-70% mortality). Repeated oral doses at 32 mg/kg and single oral doses through 375 mg/kg caused only mild to moderate effects and less than 5% mortality. In contrast, single intravenous doses at 60 mg/kg resulted in immediate death (20% mortality) due to apparent CNS toxicity. Intravenous doses below 60 mg/kg were essentially asymptomatic. Toxicity in the intravenous studies was limited to decreased WBCs, splenic and thymic lymphoid depletion (repeated dosing), and testicular tubular degeneration and/or atrophy. Except for the testicular lesions, most of the effects in the oral and intravenous studies were reversible within 4 weeks. The results show that the acute toxicity of trimetrexate in rats is somewhat dependent on its route of administration, although the spectrum of effects is qualitatively similar to that observed in other species and with other folate antagonists. The dose-limiting toxicity of trimetrexate in rats common to both routes of administration is myelosuppression.


Assuntos
Trimetrexato/toxicidade , Administração Oral , Animais , Contagem de Células Sanguíneas , Peso Corporal/efeitos dos fármacos , Medula Óssea/patologia , Doenças da Medula Óssea/induzido quimicamente , Doenças da Medula Óssea/patologia , Ingestão de Alimentos/efeitos dos fármacos , Feminino , Injeções Intravenosas , Masculino , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Endogâmicos , Doenças Testiculares/induzido quimicamente , Doenças Testiculares/patologia , Trimetrexato/administração & dosagem
13.
Toxicol Pathol ; 19(2): 184-8, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-1837612

RESUMO

Proliferative endosteal lesions were observed in metaphysis and diaphysis of femur and sternebra of Wistar (CRL:[WI]BR) rats administered 3 chemically-distinct anticancer compounds with dissimilar mechanisms of action: trimetrexate glucuronate, an antifolate; pentostatin, an adenosine deaminase inhibitor; and CI-980, a mitotic inhibitor. Islands of woven bone, often circumscribed by conspicuous myelostromal proliferation, were seen on Days 8-28 in rats given trimetrexate glucuronate daily by gavage, and on Day 4 but not Day 29 in rats given a single intravenous dose of pentostatin. Intravenous administration of CI-980 for 1 or 5 days resulted in marrow necrosis, marked centripetal new bone formation, and myelostromal proliferation on Days 4 and 8, respectively. These lesions were not present at the termination of these latter studies (Days 29 and 35, respectively). In conclusion, anticancer compounds induced local bone marrow injury and the release of local inflammatory mediators which may have provided the stimulus for bone formation and myelostromal proliferation.


Assuntos
Antineoplásicos/toxicidade , Doenças Ósseas/induzido quimicamente , Osso e Ossos/patologia , Animais , Antineoplásicos/administração & dosagem , Desenvolvimento Ósseo/efeitos dos fármacos , Doenças Ósseas/patologia , Medula Óssea/efeitos dos fármacos , Medula Óssea/patologia , Osso e Ossos/efeitos dos fármacos , Carbamatos/administração & dosagem , Carbamatos/toxicidade , Divisão Celular/efeitos dos fármacos , Combinação de Medicamentos , Feminino , Glucuronatos/administração & dosagem , Glucuronatos/toxicidade , Injeções Intravenosas , Masculino , Pentostatina/administração & dosagem , Pentostatina/toxicidade , Pirazinas/administração & dosagem , Pirazinas/toxicidade , Piridinas/administração & dosagem , Piridinas/toxicidade , Ratos , Ratos Endogâmicos , Trimetrexato/administração & dosagem , Trimetrexato/análogos & derivados , Trimetrexato/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...