Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Cancer Res ; 42(7): 2587-91, 1982 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7083151

RESUMO

The biochemical basis for the differential therapeutic activity of equally toxic doses of 9-beta-D-arabinofuranosyl-2-fluoroadenine (F-ara-A) administered on two schedules to tumor-bearing mice has been studied. A single dose (234 mg/kg) of F-ara-A in solution decreased the number of P388 leukemia cells by greater than 10(3), whereas a multiple-dose regimen (41 mg/kg every day for 5 days) of equal toxicity to the host was ineffective at reducing the tumor burden. No antitumor activity was seen when an equal dose of the relatively insoluble F-ara-A was injected as a suspension. The 5'-triphosphate of F-ara-A accumulated in P388 cells in levels proportional to the dose of the nucleoside and disappeared from these cells at an exponential rate with a half-life of 2.9 hr, which was independent of the cellular concentration of the nucleotide. The extent and duration of the inhibition of DNA synthesis of P388 cells was dependent on the dose of F-ara-A, but the rates of recovery were similar and in proportion to the cellular concentration of the analog triphosphate. The extent of the inhibition of DNA synthesis in host bone marrow and intestinal mucosa was also related to the dose of F-ara-A, but the recovery of these tissues proceeded to similar, incomplete levels (less than 60% of initial) 24 hr after F-ara-A injection of either 41 or 234 mg/kg. These results suggest that the equal toxicity of the two regimens of F-ara-A may be attributed to the similar extent of inhibition of host-tissue DNA synthesis evoked by each. In contrast, the greater extent and longer duration of inhibition of P388 cell DNA synthesis caused by the single dose of F-ara-A was responsible for its superior therapeutic activity. Measurements of F-ara-A triphosphate concentrations and the DNA-synthetic capacity of tumor and host tissues are determinants of the action of F-ara-A and may be used to predict optimal therapeutic dose schedules.


Assuntos
Leucemia P388/tratamento farmacológico , Leucemia Experimental/tratamento farmacológico , Vidarabina/análogos & derivados , Animais , Medula Óssea/metabolismo , Feminino , Mucosa Intestinal/metabolismo , Cinética , Leucemia P388/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Vidarabina/metabolismo , Vidarabina/uso terapêutico , Vidarabina/toxicidade
2.
Cancer Res ; 47(14): 3672-8, 1987 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-2439195

RESUMO

The human lymphoid cell lines CCRF/CEM/0 and the deoxycytidine kinase (dCk)-deficient CCRF/CEM/dCk- were treated with various 5-azacytidine (5-aza-C) nucleosides and the effect on DNA methylation and dCk activity were examined. 5-Azacytidine (5-aza-C), 5,6-dihydro-5-azacytidine (DHAC), 5-aza-2'-deoxycytidine (5-aza-Cdr), and 1-beta-D-arabinofuranosyl-5-azacytidine (ara-AC) reduced the DNA 5-methylcytosine level in the CEM/0 cells, down to approximately 10% of the level in untreated cells. The dCk activity was increased after treatment with the 5-aza-C nucleosides approximately 10% compared to untreated cells. In CEM/dCk- cells DNA hypomethylation between 50 and 25% of control was seen only after treatment with DHAC and 5-aza-C. No decrease in the methylation level was seen after treatment with 5-aza-Cdr or ara-AC. The dCk activity was increased up to 37% after treatment with DHAC or 5-aza-C but no increase was observed after treatment with 5-aza-Cdr or ara-AC. CEM/dCk- cells treated with DHAC showed a revertant frequency to cells expressing dCk activity of between 0.1 and 0.6%. Cloned revertant CEM/dCk- cells isolated from soft agar had dCk activity between 31 and 113% compared to the activity in untreated CEM/0 cells. This in vitro study indicates that DHAC and 5-aza-C induced dCk re-expression in the CEM/dCk- cells whereas 5-aza-Cdr and ara-AC did not.


Assuntos
Azacitidina/farmacologia , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA/metabolismo , Desoxicitidina Quinase/metabolismo , Tecido Linfoide/enzimologia , Fosfotransferases/metabolismo , Azacitidina/análogos & derivados , Linhagem Celular , Citarabina/farmacologia , Decitabina , Resistência a Medicamentos , Humanos , Tecido Linfoide/metabolismo , Metilação
3.
Cancer Res ; 47(24 Pt 1): 6786-92, 1987 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-3479250

RESUMO

The pharmacodynamic parameters of 1-beta-D-arabinofuranosylcytosine (ara-C) in patient plasma and its active anabolite 1-beta-D-arabinofuranosylcytosine-5-triphosphate (ara-CTP) in circulating and bone marrow blast cells were studied in 20 pediatric patients with acute leukemia. ara-C (3 g/m2) was administered as a short-term infusion over 3 h every 12 h for a total of eight doses. The peak plasma concentration of ara-C ranged from 0.02 to 5.6 microM after the first dose of ara-C. The area under the concentration-time curve (AUC) of ara-C in plasma ranged from 302 to 20,298 microMh after the first dose of ara-C. The half-life of elimination (t1/2,el) of ara-C from plasma was 2.4 +/- 1.5 h in three patients with acute nonlymphoblastic leukemia (ANLL) and 4.78 +/- 4.1 h in 9 patients with acute lymphoblastic leukemia (ALL). The intracellular peak concentration of ara-CTP in circulating blast cells averaged 432.2 +/- 14.5 microM and 544.3 +/- 330 microM in patients with ANLL and ALL, respectively. The elimination kinetics of ara-CTP was monoexponential with t1/2,el of 3.30 +/- 0.8 h and 6.9 +/- 2.8 h in patients with ANLL and ALL. DNA synthetic capacity (DSC) of the blast cells was inhibited to between 24 and 64% of control after the first dose of ara-C and it declined further to between 1 and 32% after four doses of ara-C. The AUC of ara-CTP in leukemic cells ranged from 1,073 to 14,751 microMh and it was not related to the AUC of ara-C in plasma. The pharmacodynamic parameters of ara-CTP in circulating blast cells were more homogeneous in patients with ANLL than in patients with ALL. Four of six patients (67%) with ANLL and six of 14 patients (43%) with ALL achieved either complete remission or partial remission with high dose ara-C. We conclude that treatment of pediatric patients with leukemia in relapse with high dose ara-C is tolerable and moderately successful. Inhibition of DSC is positively correlated with the probability of having zero nadir peripheral blast cells. In turn there is a trend for a zero nadir peripheral blast cell count to be related to achievement of a response to therapy. This latter result is consistent with the results of larger studies in adults with leukemia.


Assuntos
Citarabina/farmacocinética , Leucemia/tratamento farmacológico , Arabinofuranosilcitosina Trifosfato/sangue , Criança , Citarabina/administração & dosagem , Citarabina/efeitos adversos , Meia-Vida , Humanos , Leucemia/sangue , Leucemia Linfoide/sangue , Leucemia Linfoide/tratamento farmacológico , Neutropenia/induzido quimicamente
4.
Cancer Res ; 49(1): 241-7, 1989 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-2908850

RESUMO

In an attempt to maximize the therapeutic index and to overcome the large variations in 1-beta-D-arabinofuranosylcytosine (ara-C) plasma levels and host toxicities that have been documented with standard HDara-C regimens (3 g/m2 over 3 h every 12 h x 8 or x12 doses), pediatric patients with acute lymphocytic leukemia or lymphoma in relapse were treated with a regimen of loading bolus followed immediately by continuous infusion of ara-C. In addition, patients received a single dose of etoposide (VP-16, 1 g/m2) prior to the ara-C administration. In four patients, total body irradiation was administered as part of a bone marrow transplantation preparative regimen after the ara-C administration. The regimen was designed to attain and maintain plasma steady-state concentrations (Css) of ara-C three to four times the Km2 value of ara-C, which was determined with purified deoxycytidine kinase from the patients' tumor cells prior to treatment. Eight patients age 0.75 to 16 years with relapsed acute lymphocytic leukemia (three patients) or lymphoma (five patients, one with bone marrow involvement), received a test dose of 3 g/m2 ara-C injected over 1 h, and the plasma kinetics were determined. The peak plasma ara-C concentration of ara-C ranged from 57 to 199 microM with an average concentration of 103 +/- 49 microM; the half-lives of distribution (t1/2, alpha) and elimination (t1/2, beta) averaged 17 +/- 7 min and 4.04 +/- 3.1 h, respectively. The mean area under the plasma concentration time curve from 0 to 12 h (AUC0----12 h) of ara-C averaged 386.8 +/- 328.0 microMh (mean, +/- SD, n = 8). The peak concentration of uracil arabinoside averaged 501 +/- 123 microM, and it was eliminated with a t1/2, el of 2.3 +/- 0.6 h. The patients then received an individualized loading bolus (mean = 0.5 g/m2) followed by a continuous infusion regimen of ara-C (mean = 130 mg/m2/h), to achieve a Css in the range of 20 to 35 microM. The obtained plasma Css were similar to the desired ones, averaging in variation 10.7% +/- 8.2%. The percentage of variation of correlation of the AUC following the loading bolus plus the continuous infusion from 12 to 72 h was only 12.4% (mean = 2158 microMh, n = 8), whereas the percentage of variation of correlation of the AUC after the test dose of ara-C in the same patients was 84.8%.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Citarabina/farmacocinética , Leucemia/tratamento farmacológico , Linfoma/tratamento farmacológico , Adolescente , Criança , Pré-Escolar , Citarabina/administração & dosagem , Citarabina/efeitos adversos , Desoxicitidina Quinase/análise , Humanos , Lactente
5.
Cancer Res ; 50(22): 7226-31, 1990 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-1699658

RESUMO

Fludarabine phosphate is a nucleotide analogue of adenine arabinoside with antitumor activity in murine and human lymphoid malignancies; it has occasional, unpredictable neurotoxicity after high dose bolus injections in adults. To avoid this toxicity, we studied a loading dose plus 5-day continuous infusion in 47 evaluable pediatric patients. Dose limiting myelosuppression was seen in children with solid tumors after a loading dose of 8 mg/m2 followed by 23.5 mg/m2/day for 5 days. In children with leukemia, no dose limiting toxicity was seen at dose level 6, consisting of a loading dose of 10 mg/m2 and an infusion of 30.5 mg/m2/day for 5 days. One complete and 3 partial remissions were seen in 26 evaluable children with acute lymphoblastic leukemia. 9-beta-D-arabinofuranosyl-2-fluoroadenine plasma concentrations and the area under the moment curve increased linearly with dose. The terminal half-life was similar, while the total body clearance was shorter than that reported for adults receiving bolus or continuous doses. Lymphoblasts isolated from 2 patients during fludarabine phosphate (9-beta-D-arabinofuranosyl-2-fluoroadenine) treatment increased their ability to convert 1-beta-D-arabinofuranosylcytosine to 1-beta-D-arabinofuranosylcytosine 5'-triphosphate by more than 10-fold. The antileukemic activity of 9-beta-D-arabinofuranosyl-2-fluoroadenine 5'-phosphate and its ability to alter the metabolism of 1-beta-D-arabinofuranosylcytosine indicate that timed combinations of these 2 agents should be tested.


Assuntos
Fosfato de Vidarabina/análogos & derivados , Antimetabólitos Antineoplásicos , Arabinofuranosilcitosina Trifosfato/metabolismo , Pré-Escolar , DNA de Neoplasias/biossíntese , Relação Dose-Resposta a Droga , Esquema de Medicação , Avaliação de Medicamentos , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Fosfato de Vidarabina/administração & dosagem , Fosfato de Vidarabina/efeitos adversos , Fosfato de Vidarabina/farmacocinética
6.
Oncogene ; 20(43): 6111-22, 2001 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-11593419

RESUMO

Protein phosphatase 1 (PP1) plays important roles in many different aspects of cellular activities including cell cycle control. One important function of PP1 is to activate the retinoblastoma protein pRB. Here we show that pRB is one of PP1's downstream targets during apoptosis. When HL-60 cells synchronized at the G1/S boundary were treated with pro-apoptotic cytosine arabinoside (araC), PP1alpha protein increased twofold and PP1 activity about 30% within 1 h. This was followed by pRB dephosphorylation, pRB cleavage by caspases, DNA fragmentation, the appearance of cells with <2n DNA content and finally, dying and dead cells. In vitro, pRB was protected from caspase-3 digestion by prior Cdk-mediated phosphorylation, whereas PP1alpha converted phospho-pRB into an efficient substrate for caspase-3. Introduction of active PP1alpha into HL-60 cells by electroporation was sufficient to induce characteristics of apoptosis. Similarly, araC-resistant cells, normally unable to die in response to araC, initiated apoptosis when electroporated with active PP1alpha. This was also accompanied by pRB cleavage. In contrast, introduction of inhibitor-2 delayed the onset of araC-induced apoptosis, whereas concomitant introduction of PP1alpha and inhibitor-2 completely prevented PP1alpha-induced apoptosis. These results suggest that dephosphorylation of key proteins by PP1alpha may be crucial for the initiation of apoptosis and further support the concept of PP1 serving as a potential target for anti-cancer therapy.


Assuntos
Apoptose/efeitos dos fármacos , Fosfoproteínas Fosfatases/metabolismo , Proteína do Retinoblastoma/metabolismo , Antimetabólitos Antineoplásicos/farmacologia , Western Blotting , Caspase 3 , Caspases/metabolismo , Citarabina/farmacologia , Fragmentação do DNA , Citometria de Fluxo , Vetores Genéticos , Células HL-60 , Humanos , Fosforilação , Testes de Precipitina , Ligação Proteica , Proteína Fosfatase 1 , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Fatores de Tempo
7.
J Clin Oncol ; 13(4): 894-901, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7707116

RESUMO

PURPOSE: Treatment of neuroblastoma cell lines with 13-cis-retinoic acid (cis-RA) can cause sustained inhibition of proliferation. Since cis-RA has demonstrated clinical responses in neuroblastoma patients, it may be effective in preventing relapse after cytotoxic therapy. This phase I trial was designed to determine the maximal-tolerated dosage (MTD), toxicities, and pharmacokinetics of cis-RA administered on an intermittent schedule in children with neuroblastoma following bone marrow transplantation (BMT). PATIENTS AND METHODS: Fifty-one assessable patients, 2 to 12 years of age, were treated with oral cis-RA administered in two equally divided doses daily for 2 weeks, followed by a 2-week rest period, for up to 12 courses. The dose was escalated from 100 to 200 mg/m2/d until dose-limiting toxicity (DLT) was observed. A single intrapatient dose escalation was permitted. RESULTS: The MTD of cis-RA was 160 mg/m2/d. Dose-limiting toxicities in six of nine patients at 200 mg/m2/d included hypercalcemia (n = 3), rash (n = 2), and anemia/thrombocytopenia/emesis/rash (n = 1). All toxicities resolved after cis-RA was discontinued. Three complete responses were observed in marrow metastases. Serum levels of 7.4 +/- 3.0 mumol/L (peak) and 4.0 +/- 2.8 mumol/L (trough) at the MTD were maintained during 14 days of therapy. The DLT correlated with serum levels > or = 10 mumol/L. CONCLUSION: The MTD of cis-RA given on this intermittent schedule was 160 mg/m2/d. Serum levels known to be effective against neuroblastoma in vitro were achieved at this dose. The DLT included hypercalcemia, and may be predicted by serum cis-RA levels. Monitoring of serum calcium and cis-RA levels is indicated in future trials.


Assuntos
Transplante de Medula Óssea , Isotretinoína/uso terapêutico , Neuroblastoma/tratamento farmacológico , Criança , Pré-Escolar , Terapia Combinada , Feminino , Humanos , Hipercalcemia/induzido quimicamente , Isotretinoína/administração & dosagem , Isotretinoína/efeitos adversos , Isotretinoína/farmacocinética , Masculino , Neuroblastoma/sangue , Neuroblastoma/terapia , Indução de Remissão
8.
J Clin Oncol ; 15(8): 2780-5, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9256119

RESUMO

PURPOSE: The Children's Cancer Group (CCG) undertook a phase I study (CCG-0922) to determine a tolerable dose of idarubicin given with fludarabine and cytarabine in children with relapsed or refractory leukemia. The phase I study was extended to a limited phase II study to assess the activity of this combination in children with acute myelogenous leukemia (AML). PATIENTS AND METHODS: This was a multiinstitutional study within the CCG. Eleven patients were entered onto the phase I study: seven with AML, three with acute lymphoblastic leukemia (ALL), and one with chronic myelogenous leukemia (CML). The maximal-tolerated dose (MTD) of fludarabine and cytarabine determined in a previous study was a fludarabine loading dose (LD) of 10.5 mg/m2 followed by a continuous infusion (CI) of 30.5 mg/m2/24 hours for 48 hours, followed by cytarabine LD 390 mg/m2, then CI 101 mg/m2/h for 72 hours. Idarubicin was given at three dose levels: 6, 9, and 12 mg/m2 intravenously (I.V.) on days 0, 1, and 2. The phase II portion of the trial included 10 additional patients with relapsed or refractory AML. RESULTS: A dose of idarubicin 12 mg/m2/d for 3 days given in combination with fludarabine and cytarabine was tolerated. The major toxicity encountered was hematologic. Nonhematologic toxicities included transaminase elevations, hyperbilirubinemia, and infections. Eight of 10 patients with AML in the phase II portion (12 mg/m2 idarubicin) achieved a complete remission (CR). CONCLUSION: This combination is active in patients with relapsed or refractory AML. The major toxicity encountered is hematologic. This regimen may be useful therapy for AML and should be compared with standard induction therapy in children with newly diagnosed AML.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Leucemia/tratamento farmacológico , Doença Aguda , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Criança , Pré-Escolar , Citarabina/administração & dosagem , Citarabina/efeitos adversos , Feminino , Humanos , Idarubicina/administração & dosagem , Idarubicina/efeitos adversos , Lactente , Infusões Intravenosas , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mieloide Aguda/tratamento farmacológico , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Vidarabina/administração & dosagem , Vidarabina/efeitos adversos , Vidarabina/análogos & derivados
9.
Clin Cancer Res ; 4(1): 45-52, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9516951

RESUMO

The sequential administration of fludarabine followed by cytosine arabinoside (ara-C) has demonstrated significant synergistic effects against the CEM human leukemic cell line. This in vitro synergism was investigated in a Phase I trial in pediatric patients with relapsed acute leukemia. The optimum concentrations of 9-beta-D-arabinofuranosyl 2-fluoroadenine and ara-C necessary to achieve significant drug synergism from in vitro studies were between 10 and 20 microM. Fludarabine was infused at a dose to attain a target plasma concentration of 10 microM for 48 h, followed by a continuous infusion of escalated ara-C doses to maintain plasma ara-C concentrations of 10, 12.5, 15, or 17.5 microM for 72 h. Thirteen patients with acute lymphocytic leukemia and 18 with acute myelocytic leukemia were entered into the study, 30 of whom were clinically evaluable for toxicity. Pharmacokinetic and pharmacodynamic studies were performed on specimens from 20 patients. The optimal 9-beta-D-arabinofuranosyl 2-fluoroadenine and ara-C concentrations in plasma were easily achieved after continuous infusion regimens of both drugs. Cellular ara-CTP is augmented 5-8-fold in leukemic cells from patients receiving fludarabine phosphate treatment followed by ara-C. The maximum tolerated plasma concentrations for this combination regimen was 10 microM fludarabine for 48 h followed by 72 h of 15 microM ara-C, which were achieved at dose level 3. A significant number of responses were also seen. Nine of 18 evaluable patients (50%) with acute myelocytic leukemia achieved complete or partial responses, and 3 of 9 evaluable patients with acute lymphocytic leukemia achieved complete or partial responses. Fludarabine and ara-C successfully eradicated bone marrow disease in 16 of 27 patients (59%), 23 patients of which had been treated previously with high-dose ara-C. These results verified the synergistic effect fludarabine exhibited in augmenting ara-CTP concentrations in patients' leukemic blasts, thus improving the clinical response in relapsed pediatric leukemias.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Citarabina/administração & dosagem , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Vidarabina/análogos & derivados , Adolescente , Adulto , Arabinofuranosilcitosina Trifosfato/farmacocinética , Criança , Pré-Escolar , Citarabina/efeitos adversos , Citarabina/farmacocinética , Humanos , Lactente , Vidarabina/administração & dosagem , Vidarabina/efeitos adversos , Vidarabina/farmacocinética
10.
Clin Cancer Res ; 2(9): 1533-42, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9816330

RESUMO

Combined immunochemotherapy regimens using the investigational biotherapeutic agent B43(anti-CD19)-poke-weed antiviral protein (PAP) immunotoxin may offer an effective treatment for refractory B-cell precursor leukemias. The purpose of the present study was to explore and identify effective combinations of B43-PAP with standard chemotherapeutic drugs, including the anthracyclin doxorubicin, the epipodophyllotoxin etoposide, the nitrosurea carmustine, and the antimetabolite cytosine arabinoside. Here, we report that the B43-PAP plus cytosine arabinoside combination has potent antileukemic activity against human B-cell precursor leukemia in SCID mice and leads to 100% long-term event-free survival from an otherwise invariably fatal leukemia. Surprisingly, none of the other treatment protocols tested, including combinations of B43-PAP with carmustine, doxorubicin, or etoposide, proved more effective than B43-PAP alone.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Citarabina/uso terapêutico , N-Glicosil Hidrolases , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamento farmacológico , Animais , Anticorpos Monoclonais/uso terapêutico , Carmustina/uso terapêutico , Doxorrubicina/uso terapêutico , Etoposídeo/uso terapêutico , Feminino , Humanos , Imunotoxinas/uso terapêutico , Masculino , Camundongos , Camundongos SCID , Transplante de Neoplasias , Proteínas de Plantas/uso terapêutico , Proteínas Inativadoras de Ribossomos Tipo 1 , Organismos Livres de Patógenos Específicos , Resultado do Tratamento
11.
AIDS ; 3(7): 417-22, 1989 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-2504244

RESUMO

HIV is the causative agent of AIDS. The purpose of this study was to examine the biochemical pharmacology of the anti-viral agent zidovudine (AZT) in the T-cell origin line (CEM). We have shown that zidovudine is activated by thymidine kinase (TK) in CEM cells to the triphosphate anabolite, which is incorporated into DNA. One microM zidovudine is sufficient for saturation of activation by TK and also of zidovudine monophosphate, by thymidylate kinase, to the diphosphate. Zidovudine triphosphate peaked 4 h after initiation of drug administration in CEM cells and then declined biexponentially. Nucleoside triphosphate (NTP) cellular concentrations declined rapidly in the cells after exposure to zidovudine. Concomitantly phosphorylation of zidovudine to zidovudine monophosphate and zidovudine monophosphate to zidovudine diphosphate declined in a similar manner in the CEM cells. The amount of zidovudine anabolite incorporated into purified DNA peaked 1 h after zidovudine treatment and declined thereafter with first order elimination kinetics. These studies elucidate the cellular activation of zidovudine in a T-cell line, CEM, and enhance our understanding of this important anti-HIV drug.


Assuntos
Linfócitos T/efeitos dos fármacos , Zidovudina/metabolismo , Divisão Celular/efeitos dos fármacos , Linhagem Celular , DNA/metabolismo , Humanos , Linfócitos T/metabolismo , Zidovudina/farmacologia
12.
J Acquir Immune Defic Syndr (1988) ; 6(12): 1287-96, 1993 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8254464

RESUMO

Clinical reports indicate that the development of drug resistance to AZT after chronic administration is common. In order to study this phenomenon, the T-cell line Jurkat E6-1 was treated continuously in vitro with low, gradually increased, concentrations of azidothymidine (AZT). Initially, 1 microM AZT significantly retarded the cell line from reaching confluence. However, after 10 weeks the T-cell line was able to grow in 10 microM AZT without any evidence of growth inhibition. Subsequently, cell isolates could grow continuously in the presence of 20, 50, and 100 microM AZT without growth inhibition. These T-cell lines (Jurkat E6-1/AZT-10, Jurkat E6-1/AZT-20, Jurkat E6-1/AZT-50, and Jurkat E6-1/AZT-100) were tested for AZT anabolism using purified [3H]AZT, and the results were compared to the wild-type untreated Jurkat E6-1 cell line. Similar intracellular AZT anabolites concentrations were determined in all cell lines. However, a four- to sixfold lower cellular concentration of mono-, di-, and triphosphate anabolites of AZT was determined in the Jurkat E6-1/AZT-10 cell line after 1 microM AZT incubation and 6.5-fold lower after 10 microM AZT treatment. In general, a five- to sixfold reduction in the phosphorylation rates were estimated in the AZT resistant T-cell line. Pharmacology studies of [3H]AZT in the Jurkat E6-1/AZT-100 cell line showed a much lower level of activation of the pro-drug (28-fold), due to lack of thymidine kinase (TK) activity when compared to the Jurkat E6-1/AZT-10 T-cell line. A similar level of resistance was obtained at the thymidylate (dTMP) kinase level. Concurrently an additional mode of resistance (407-fold) was seen on the incorporation of the AZT triphosphate anabolite (AZTTP) into cellular DNA. The formation of this cell line in a period of < or = 4 months coincides with the evidence of the clinical development of "resistance" to AZT in patients who receive the drug continuously. In addition, these T-cell lines have been infected with HIV, and studies on the development of collaterally sensitive regimens are under way.


Assuntos
Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Leucemia de Células T/patologia , Linfócitos T/efeitos dos fármacos , Zidovudina/farmacologia , Adolescente , Adulto , Antivirais/metabolismo , Divisão Celular/efeitos dos fármacos , Criança , Pré-Escolar , Células Clonais , DNA de Neoplasias/metabolismo , Didesoxinucleotídeos , Resistência a Medicamentos , Resistência Microbiana a Medicamentos , Humanos , Lactente , Linfócitos T/enzimologia , Timidina Quinase/metabolismo , Nucleotídeos de Timina/metabolismo , Células Tumorais Cultivadas , Zidovudina/análogos & derivados , Zidovudina/metabolismo , Zidovudina/uso terapêutico
13.
Cancer Chemother Pharmacol ; 21(2): 117-21, 1988.
Artigo em Inglês | MEDLINE | ID: mdl-2450689

RESUMO

Dihydro-5-azacytidine (DHAC) is a hydrolytically stable congener of 5-azacytidine, which retains antileukemic activity against experimental leukemias. The biochemical pharmacology of DHAC was studied in tumor-bearing mice in order to elucidate the mode of action of this drug. We found that after an LD10 dose of DHAC, the plasma peak concentration achieved was 317 microM and was eliminated biexponentially, with a t1/2 alpha of 1.03 h and a t1/2 beta of 5 h. By 4 h, an unidentified metabolite of [3H]DHAC peaked and was eliminated biexponentially, with a t1/2 alpha of 1.06 h and a t1/2 beta of 10.6 h. [3H]DHACTP was the major anabolite in the L1210/0 cells, and was also eliminated biexponentially, with a t1/2 alpha of 4.3 h and a t1/2 beta of 12.2 h. An unknown anabolite of [3H]DHAC that eluted 5 min after [3H]DHACTP, between UTP and ATP, peaked at 3 h and could possibly be the deoxy-derivative [3H]DHAdCTP. A tissue distribution study revealed that the liver, L1210/0, and lung accumulate the most radioactivity per gram of wet tissue. Methylation studies showed that an LD10 dose of [3H]DHAC resulted in a 25.06% hypomethylation of DNA in L1210/0 cells and a 46.32% hypomethylation in a deoxycytidine kinase mutant cell line L1210/dCK(-), compared with their respective controls.


Assuntos
Azacitidina/análogos & derivados , DNA de Neoplasias/metabolismo , Leucemia L1210/tratamento farmacológico , Animais , Azacitidina/farmacocinética , Azacitidina/farmacologia , Azacitidina/uso terapêutico , Medula Óssea/metabolismo , Encéfalo/metabolismo , Cromatografia Líquida de Alta Pressão , Eritrócitos/metabolismo , Mucosa Intestinal/metabolismo , Rim/metabolismo , Leucemia L1210/metabolismo , Fígado/metabolismo , Pulmão/metabolismo , Masculino , Metilação , Camundongos , Músculos/metabolismo , Miocárdio/metabolismo , Baço/metabolismo , Testículo/metabolismo , Distribuição Tecidual , Células Tumorais Cultivadas
14.
Cancer Chemother Pharmacol ; 39(1-2): 42-50, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8995498

RESUMO

We examined the population pharmacokinetics (PPK) of cytosine arabinoside (ara-C) after high-dose ara-C (HDara-C) (3 g/m2 every 12 h) and after a loading bolus (LB) plus continuous infusion (C1) of ara-C for 72 h in 52 pediatric patients with leukemias, enrolled in four clinical trials. The PPK analyses of the drug were performed using the NONMEM program. The patients' ages ranged from 2 months to 19 years. The ara-C data were analyzed using a two-compartment open model. Interindividual variability was described by the constant coefficient of variation (CCV) model, while the intraindividual variability was described by a combined additive and CCV error model. The covariates age (AGE) and surface area (SA) were tested to examine their influence on the estimation of the ara-C PPK parameters. In the absence of model covariates, the data fit was characterized by considerable bias, as indicated by the plot of measured vs predicted ara-C concentrations. The fit of the data was greatly improved when the parameters total body clearance (CL), intercompartmental clearance (Q), and volumes of distribution of central (Vd1) and peripheral (Vd2) compartments were expressed as linear functions of the covariate product, AGE x SA. The final parameter estimates were: CL = 2.59 x AGE x SA 1/h, Q = 2.01 x AGE x SA 1/h, Vd1 = 0.48 x AGE x SA1, and Vd2 = 38.1 x AGE x SA1. The coefficients of variation of CL, Q, Vd1 and Vd2 were 83.79%, 12.08%, 40.0%, and 52.54%, respectively, indicating substantial interindividual variability. In separate NONMEM analyses, the PK of ara-C and its metabolite uracil arabinoside (ara-U) were modeled simultaneously in order to investigate whether the dependence of ara-C on patient age was due to increased deamination of ara-C to ara-U. The PK of ara-C were described by the two-compartment open model while the PK of ara-U were simultaneously described by the one-compartment open model. The conversion of ara-C to ara-U was modeled as a first-order kinetic process due to the relatively low concentrations of ara-C in plasma. These PPK analyses indicated that elimination of ara-C from the central compartment occurs primarily by its metabolic conversion to ara-U and that the rate of conversion of ara-C to ara-U increases with increasing patient age, which explains the higher ratios of ara-U to ara-C and, hence, the increased ara-C clearance observed in older children as compared to infants. We conclude that the NONMEM PPK methodology allowed the simultaneous analyses of data from different doses and dose regimens and explained phenomena that prior standard two-stage analyses could not.


Assuntos
Citarabina/farmacocinética , Leucemia/tratamento farmacológico , Leucemia/metabolismo , Modelos Biológicos , Adolescente , Adulto , Arabinofuranosiluracila/sangue , Arabinofuranosiluracila/farmacocinética , Criança , Pré-Escolar , Cromatografia Líquida de Alta Pressão , Citarabina/administração & dosagem , Citarabina/sangue , Meia-Vida , Humanos , Lactente , Infusões Intravenosas , Taxa de Depuração Metabólica
15.
Cancer Chemother Pharmacol ; 35(3): 191-9, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-7805176

RESUMO

Selective combinations of purine and pyrimidine analogs increase remission rates in pediatric patients with relapsed leukemias. The combination of 6-mercaptopurine (6-MP) and cytosine arabinoside (ara-C) may exhibit synergism similar to that observed for fludarabine and ara-C and may diminish the potential for development of resistance since the two drugs are activated by separate enzymatic pathways. To determine the efficacy of the combination against human leukemia cells, we investigated the time-concentration relationships of the drugs given alone or in combination to the resultant cytotoxicity. To determine whether the combination leads to enhanced activity of deoxycytidine kinase (dCk), the rate-limiting enzyme in ara-C activation, we characterized the cellular dCk in CCRF/CEM/0, CCRF/CEM/ara-C/7A, and CCRF/CEM/ara-C/3A monoclonal cells before and after treatment with 6-MP. CCRF/CEM/0 (wild type), CCRF/CEM/ara-C/7A (approximately 50% ara-C-resistant as determined by ara-C sensitivity assay and dCk characterization), and CCRF/CEM/ara-C/3A (approximately 90% resistant to ara-C) human leukemia cells were incubated with various concentrations of 6-MP and ara-C given alone or in combination. Cell survival, inhibition of DNA synthetic capacity (DSC), ara-CTP anabolism, and dCk enzymatic characteristics were studied. Incubation of CEM/0 cells with 6-MP for 24 h, followed by ara-C for 48 h, increased cell-growth inhibition by approximately 0.5-1 log10, corresponding to 5- to 10-fold synergism, as compared with ara-C alone after identical drug incubation in all cell lines. Simultaneous administration showed no synergism, whereas reversal of the sequence produced an antagonistic effect. The ara-CTP levels were 2- to 3.5-fold and 3- to 5-fold higher in CEM/0 and CEM/ara-C/7A cells, respectively, in cells exposed to 6-MP followed by ara-C than in those exposed to ara-C alone at the same concentrations. Furthermore, a progressive increase in ara-CTP levels was noted in CEM/0 cells exposed to increasing concentrations of 6-MP followed by 10 or 20 microM ara-C. A significant decrease in DSC was observed upon treatment of wild-type and ara-C-resistant cells with 6-MP and ara-C. The combination of 6-MP and ara-C exhibits significant sequence-specific synergism in both wild-type and partially ara-C-resistant leukemia cell lines. The combination also exerts collateral sensitivity in the ara-C-resistant cell lines. 6-MP pretreatment may play a role in enhancing ara-C activation, thus producing drug synergism in sensitive and resistant leukemia cell lines.


Assuntos
Citarabina/farmacologia , Leucemia/tratamento farmacológico , Mercaptopurina/farmacologia , Arabinofuranosilcitosina Trifosfato/metabolismo , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citarabina/uso terapêutico , DNA de Neoplasias/biossíntese , DNA de Neoplasias/efeitos dos fármacos , Desoxicitidina Quinase/metabolismo , Relação Dose-Resposta a Droga , Resistência a Medicamentos , Sinergismo Farmacológico , Humanos , Cinética , Mercaptopurina/uso terapêutico , Células Tumorais Cultivadas
16.
Cancer Chemother Pharmacol ; 25(1): 19-24, 1989.
Artigo em Inglês | MEDLINE | ID: mdl-2480187

RESUMO

1-beta-D-arabinosyl-5-azacytosine (ara-AC) is a relatively new antitumor agent under clinical investigation, which has the 2'-beta arabinosyl configuration found in the tumoricidal drug ara-C and the nitrogen substitution in the 5-position of the pyrimidine ring found in 5-azacytidine (5-aza-C). The present study examined the cellular metabolism and the effect on DNA methylation of ara-AC in human CCRF/CEM cells sensitive and resistant to ara-C. The triphosphate anabolite of the drug, ara-ACTP, was the major anabolite in the CEM cellular extracts, peaking at 50.6 +/- 23 microM 4 h after incubation with IC50 concentrations (0.25 microM) of [3H]ara-AC. The mono- and diphosphate anabolites accumulated 10-fold lower cellular concentrations than ara-ACTP. The nucleoside triphosphate (NTP) pools and, especially, cellular ATP declined significantly by 9 h after the initiation of drug treatment and remained depleted for the 24-h treatment. The drug anabolite was gradually incorporated into both RNA and DNA, peaking in CEM/0 at 3.44 and 0.14 nmol/10(7) cells, respectively. The DNA methylation levels in these cells declined rapidly after treatment with ara-AC, attaining a nadir plateau at 29% of control methylation value. The deoxycytidine kinase (dCK) mutant CEM cell line [CEM/dCk(-)] neither activated ara-AC at appreciable levels nor induced DNA hypomethylation at low concentrations (0.25-1 microM). However, the drug was activated at 0.2-1 microM extracellular concentrations of ara-AC, probably by an as yet unknown nucleoside kinase at approximately 10% of the amount in CEM/0 cells. Ara-AC appears to mediate its cytotoxic action through the accumulation of its triphosphate anabolite, ara-ACTP, and the subsequent incorporation into nucleic acids. DNA methylation may also contribute to its cytotoxicity.


Assuntos
Antineoplásicos/farmacocinética , Azacitidina/farmacocinética , DNA de Neoplasias/metabolismo , Leucemia/metabolismo , RNA Neoplásico/metabolismo , Antineoplásicos/farmacologia , Azacitidina/farmacologia , Linhagem Celular/efeitos dos fármacos , Linhagem Celular/metabolismo , DNA de Neoplasias/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Metilação , RNA Neoplásico/efeitos dos fármacos , Estereoisomerismo , Fatores de Tempo , Trítio , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
17.
Cancer Chemother Pharmacol ; 24(3): 155-60, 1989.
Artigo em Inglês | MEDLINE | ID: mdl-2472227

RESUMO

5,6-Dihydro-5-azacytidine (DHAC) is a hydrolytically stable analog of 5-azacytidine (5-aza-C) that has antileukemic activity against experimental leukemias and, like 5-aza-C, causes DNA hypomethylation. We report the cellular metabolism of DHAC and its incorporation into nucleic acids in the CCRF/CEM/O and deoxycytidine kinase mutant CCRF/CEM/dCk(-) human lymphoid cell lines. The cells were incubated with their respective IC50 concentrations for 24 h, then aliquot samples were removed at predetermined intervals and extracted for nucleotides. The acid-soluble extracts of the cells were assayed on HPLC for nucleotides of DHAC. The major anabolite of [3H]DHAC, [3H]DHACTP, peaked at 110.3 +/- 30.7 microM in CEM/O and at 96.3 +/- 41.9 microM in CEM/dCk(-) cells at 9 and 12 h, respectively. The intracellular concentrations of the deoxyribonucleoside triphosphate, [3H]DHAdCTP, peaked at 13.5 +/- 7.7 microM at 4 h in CEM/O and at 80.8 +/- 13.8 microM at 12 h, a 6-fold greater cellular concentration, in the dCk mutant cell line. The amount of DHAC anabolites incorporated into CEM/O nucleic acids reached a plateau in RNA at 552.6 +/- 7.8 pmol/10(7) cells and in DNA at 64.55 +/- 10.0 pmol/10(7) cells. In CEM/dCk(-) cells, DHAC anabolites reached a plateau in RNA and DNA at 4,256.3 +/- 631.0 and 395.5 +/- 145.4 pmol/10(7) cells, respectively. Thus, with equitoxic treatments of DHAC, the incorporation of its analog anabolites into RNA and DNA was 8- and 6-fold greater in CEM/dCk(-) cells. DNA methylation levels were depressed equally despite a 6-fold greater incorporation of the analog in DNA in the CEM/dCk(-) cells indicating that hypomethylation may be saturated after DHAC treatment. The DNA methylation levels reached a nadir of 0.19% and 0.20% methyl-C (percentage of methylation) in the two cell lines at 6 and 12 h after the beginning of drug treatment and remained relatively constant for the duration of the 24-h treatment. A curve-linear relationship was obtained between the DNA methylation levels in both cell lines and the amounts of DHAC anabolite incorporated into DNA.


Assuntos
Antimetabólitos Antineoplásicos/farmacocinética , Azacitidina/análogos & derivados , DNA de Neoplasias/metabolismo , Leucemia/metabolismo , Linfócitos/metabolismo , RNA Neoplásico/metabolismo , Azacitidina/farmacocinética , Linhagem Celular , Humanos , Metilação , Mutação , Fatores de Tempo , Trítio , Células Tumorais Cultivadas
18.
Cancer Chemother Pharmacol ; 33(3): 197-202, 1993.
Artigo em Inglês | MEDLINE | ID: mdl-8269600

RESUMO

O-6-methylguanine (O6-mG), a guanine analog recently shown to be a potent inhibitor of alkylguanine-DNA alkyltransferase, has been found to potentiate the antitumor activity of nitrosoureas, in particular, carmustine (BCNU), in resistant cell lines (HT-29 mer+) and is targeted for development as a modulating agent with chloroethyl nitrosoureas. A high-performance liquid chromatography (HPLC) assay of O6-mG in plasma has been developed using a microC18 reverse-phase column. O6-mG and the internal standard deoxyguanosine (dGuo) were eluted with a linear gradient of from 15% to 35% methanol in 0.5 M ammonium acetate (pH 6.5) at a flow rate of 1 ml/min. The assay was linear over a 4-log concentration range with a detection limit of 0.1 microgram/ml. The within-run and between-run coefficients of variation (CV) were found to be 8.1% and 9.3%, respectively. The pharmacokinetics (PK) of O6-mG were investigated in healthy CDF1 mice following separate i.v. and i.p. administrations. At 20 mg/kg i.v., plasma O6-mG gave a biexponential profile with a terminal half-life (t1/2) of 24 min and a total clearance (CLT) of 23.7 ml min-1 kg-1. Higher doses (40-80 mg/kg) revealed a fluctuating third phase, probably due to enterohepatic cycling. Dose-dependent kinetics as measured by CLT and area under the plasma-concentration curve (AUC) values were also seen. Following i.p. dosing, O6-mG was completely absorbed and available to the circulation. No acute toxicity was observed in the animals, except for mild sedation, a possible side effect of the 10% ethanol used in the formulation. Studies on the cellular metabolism of highly purified [3H]-O6-mG have shown that the compound is not anabolized by a human lymphoblastoid cell line (CEM). Biochemistry studies have shown that the parent molecule is inactivating the alkylguanine-DNA alkyltransferase (AGT), thus exerting its pharmacological effect.


Assuntos
Guanina/análogos & derivados , Animais , Proteínas Sanguíneas/metabolismo , Cromatografia Líquida de Alta Pressão , Avaliação Pré-Clínica de Medicamentos , Guanina/administração & dosagem , Guanina/metabolismo , Guanina/farmacocinética , Meia-Vida , Humanos , Injeções Intraperitoneais , Injeções Intravenosas , Leucemia Linfoide/metabolismo , Leucemia Linfoide/patologia , Camundongos , Ligação Proteica , Ratos , Células Tumorais Cultivadas
19.
Cancer Chemother Pharmacol ; 24(4): 203-10, 1989.
Artigo em Inglês | MEDLINE | ID: mdl-2473850

RESUMO

The primary development of clinical resistance to 1-beta-D-arabinofuranosyl cytosine (ara-C) in leukemic blast cells is expressed as decreased cellular concentrations of its active anabolite. Correlations exist between the cellular concentrations of 1-beta-D-arabinofuranosyl cytosine 5'-triphosphate (ara-CTP) in leukemic blast cells and inhibition of DNA synthetic capacity with the clinical response to high-dose cytosine arabinoside (HDara-C). 5-Azacytidine (5-Aza-C) and its congeners are potent DNA hypomethylating agents, an action closely associated with the reexpression of certain genes such as that for deoxycytidine kinase (dCk) in ara-C-resistant mouse and human leukemic cells. Reexpression of dCk could increase the cellular ara-CTP concentrations and the sensitivity to ara-C. A total of 17 pediatric patients with refractory acute lymphocytic leukemia (ALL) received a continuous infusion of 5-Aza-C at 150 mg/m2 daily for 5 days after not responding to (13/17) or relapsing from (4/17) an HDara-C regimen (3 g/m2 over 3 h, every 12 h, x 8 doses). Approximately 3 days after the end of the 5-Aza-C infusion, the HDara-C regimen was given again with the idea that the induced DNA hypomethylation in the leukemic cells may have increased the dCk activity and that a reversal of the tumor drug resistance to ara-C could have occurred. Deoxycytidine kinase (expressed as cellular ara-CTP concentrations) in untreated blasts, DNA synthetic capacity (DSC), and the percentage of DNA methylcytidine levels were determined before and after 5-Aza-C administration. Cellular ara-CTP was enhanced to varying degrees in 15 of 16 patients after 5-Aza-C treatment. The average cellular concentration of ara-CTP determined in vitro by the sensitivity test was 314 +/- 390 microM, 2.3-fold higher than the average value before 5-Aza-C treatment. In 12 patients in whom the DNA methylation studies were completed before and after 5-Aza-C treatment, the average DNA hypomethylation level was 55.6% + 15.8% of pretreatment values (n = 13; mean +/- SD). DSC showed a profound decline in 2/9 evaluable patients who achieved a complete response (CR) after this regimen. The data suggest that treatment with a cytostatic but DNA-modulatory regimen of 5-Aza-C causes DNA hypomethylation in vivo, which is associated with dCk reexpression in the patients' leukemic blasts. The partial reversal of drug resistance to ara-C by 5-Aza-C yielded two CRs in this poor-prognosis, multiply relapsed patient population with refractory ALL.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Citarabina/farmacologia , DNA de Neoplasias/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Adolescente , Adulto , Arabinofuranosilcitosina Trifosfato/metabolismo , Azacitidina/administração & dosagem , Criança , Pré-Escolar , Citarabina/administração & dosagem , DNA de Neoplasias/biossíntese , DNA de Neoplasias/efeitos dos fármacos , Desoxicitidina Quinase/genética , Resistência a Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Metilação , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Indução de Remissão , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
20.
Cancer Chemother Pharmacol ; 39(1-2): 34-41, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8995497

RESUMO

A phase I clinical trial of 13-cis-retinoic acid (cis-RA) was undertaken to determine the maximally tolerated dose (MTD) and pharmacokinetics (PK) of cis-RA following bone marrow transplantation (BMT) in children with high-risk neuroblastoma. Mean peak serum levels of cis-RA in 31 pediatric patients ranged from 4.9 to 8.9 microM following doses of 100-200 mg/m2 per day, divided into two doses every 12 h administered orally. The PK of cis-RA obeyed a single-compartment model following first-order absorption in the majority of patients. A linear increase in the mean peak serum levels and area under the time-concentration curve (AUC) with increasing dose was observed. The average half-lives of absorption and elimination were 1.0 and 5.8 h, respectively. At the MTD of 160 mg/m2 per day, the mean cis-RA peak serum concentration was 7.2 +/- 5.3 microM. AUC values were not altered significantly during a 2-week course of treatment or over a long period of multiple courses. Levels of trans-retinoic acid, a metabolite of cis-RA, remained low but were similar on days 1 and 14, whereas the 4-oxo-13-cis-RA metabolite had increased in 64% of patients by day 14. Peak serum cis-RA concentrations correlated with clinical toxicity as grade 3 to 4 toxicity was seen in 44% of patient-courses (8/18) with peak serum levels > 10 microM, but only 13% (12/96) with peak serum levels < 10 microM. These results show that cis-RA given at 160 mg/m2 to children achieved serum concentrations known to be effective against neuroblastoma in vitro, and the PK for cis-RA differs from that reported for trans-retinoic acid in children.


Assuntos
Isotretinoína/farmacocinética , Ceratolíticos/farmacocinética , Neuroblastoma/metabolismo , Administração Oral , Área Sob a Curva , Transplante de Medula Óssea , Criança , Pré-Escolar , Cromatografia Líquida de Alta Pressão , Terapia Combinada , Esquema de Medicação , Feminino , Humanos , Isotretinoína/administração & dosagem , Isotretinoína/sangue , Ceratolíticos/administração & dosagem , Ceratolíticos/sangue , Masculino , Neuroblastoma/terapia , Fatores Sexuais , Estereoisomerismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA