Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 204
Filtrar
1.
Blood Cancer J ; 6(9): e465, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27588518

RESUMEN

Approximately 10-20% of chronic lymphocytic leukemia (CLL) patients exhibit del(11q22-23) before treatment, this cohort increases to over 40% upon progression following chemoimmunotherapy. The coding sequence of the DNA damage response gene, ataxia-telangiectasia-mutated (ATM), is contained in this deletion. The residual ATM allele is frequently mutated, suggesting a relationship between gene function and clinical response. To investigate this possibility, we sought to develop and validate an assay for the function of ATM protein in these patients. SMC1 (structural maintenance of chromosomes 1) and KAP1 (KRAB-associated protein 1) were found to be unique substrates of ATM kinase by immunoblot detection following ionizing radiation. Using a pool of eight fluorescence in situ hybridization-negative CLL samples as a standard, the phosphorylation of SMC1 and KAP1 from 46 del (11q22-23) samples was analyzed using normal mixture model-based clustering. This identified 13 samples (28%) that were deficient in ATM function. Targeted sequencing of the ATM gene of these samples, with reference to genomic DNA, revealed 12 somatic mutations and 15 germline mutations in these samples. No strong correlation was observed between ATM mutation and function. Therefore, mutation status may not be taken as an indicator of ATM function. Rather, a direct assay of the kinase activity should be used in the development of therapies.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Deleción Cromosómica , Cromosomas Humanos Par 11 , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/metabolismo , Mutación , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/metabolismo , Metilación de ADN , Eliminación de Gen , Mutación de Línea Germinal , Humanos , Fosforilación , Regiones Promotoras Genéticas , Proteína 28 que Contiene Motivos Tripartito/metabolismo
2.
Leukemia ; 26(3): 443-50, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21904380

RESUMEN

Triptolide, isolated from the herb Tripterygium wilfordii, has been shown to potently induce apoptosis in various malignant cells by inhibiting RNA synthesis and nuclear factor-κB activity. Previously, we showed that triptolide promotes apoptosis in acute myeloid leukemia (AML) cells via the mitochondria-mediated pathway, in part, by decreasing levels of the anti-apoptotic proteins XIAP and Mcl-1. MRx102 is a triptolide derivative, currently in preclinical development. Here we show that MRx102 potently promoted apoptosis in AML cell lines, with EC(50) values of 14.5±0.6 nM and 37.0±0.9 nM at 48 h for OCI-AML3 and MV4-11 cells, respectively. MRx102, at low nanomolar concentrations, also induced apoptosis in bulk, CD34(+) progenitor, and more importantly, CD34(+)CD38(-) stem/progenitor cells from AML patients, even when they were protected by coculture with bone marrow derived mesenchymal stromal cells. MRx102 decreased XIAP and Mcl-1 protein levels and inhibited RNA synthesis in OCI-AML3 cells. In vivo, MRx102 greatly decreased leukemia burden and increased survival time in non-obese diabetic/severe combined immunodeficiency mice harboring Ba/F3-ITD cells. Collectively, we demonstrated that MRx102 has potent antileukemic activity both in vitro and in vivo, has the potential to eliminate AML stem/progenitor cells and overcome microenvironmental protection of leukemic cells, and warrants clinical investigation.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Fenantrenos/farmacología , Fenantrenos/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Médula Ósea/inmunología , Médula Ósea/metabolismo , Médula Ósea/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Leucemia Mieloide Aguda/mortalidad , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transcripción Genética/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Phys Chem Chem Phys ; 12(26): 6984-9, 2010 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-20485727

RESUMEN

Rhombohedral shaped, single crystal hematite particles with narrow size distribution (D(TEM) = 93 +/- 2 nm) were prepared by hydrolysis of iron chloride and polymerisation in water. The results of field dependent magnetization measurements at different warming-cooling rates and ac susceptibility measurements at varying frequencies are reported and discussed. Thermal hysteresis (DeltaT(M)) associated with the Morin transition and field dependence of the Morin temperature (T(M)) are observed in warming-cooling cycles (DeltaT(M) = 25 and 13 K for H = 0.1 and 3 T, respectively) due to the first order phase transition. A frequency dependence of ac susceptibility is observed above T(M), as a result of the relaxation of the magnetic moment of hematite particles in the weak-ferromagnetic phase.

4.
Oncogene ; 27(50): 6522-37, 2008 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-18955977

RESUMEN

Nucleoside analogs are structurally similar antimetabolites that have a broad range of action and are clinically active in both solid tumors and hematological malignancies. Many of these agents are incorporated into DNA by polymerases during normal DNA synthesis, an action that blocks further extension of the nascent strand and causes stalling of replication forks. The molecular mechanisms that sense stalled replication forks activate cell cycle checkpoints and DNA repair processes, which may contribute to drug resistance. When replication forks are not stabilized by these molecules or when subsequent DNA repair processes are overwhelmed, apoptosis is initiated either by these same DNA damage sensors or by alternative mechanisms. Recently, strategies aimed at targeting DNA damage checkpoints or DNA repair processes have demonstrated effectiveness in sensitizing cells to nucleoside analogs, thus offering a means to elude drug resistance. In addition to their DNA synthesis-directed actions many nucleoside analogs trigger apoptosis by unique mechanisms, such as causing epigenetic modifications or by direct activation of the apoptosome. A review of the cellular and molecular responses to clinically relevant agents provides an understanding of the mechanisms that cause apoptosis and may provide rationale for the development of novel therapeutic strategies.


Asunto(s)
Apoptosis/efectos de los fármacos , Nucleósidos/farmacología , Transducción de Señal , Animales , Daño del ADN , Metilación de ADN , Reparación del ADN , Replicación del ADN , Humanos , Nucleósidos/química , Nucleósidos/metabolismo
5.
Leukemia ; 20(4): 610-9, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16482209

RESUMEN

17-Allylamino-17-demethoxygeldanamycin (17-AAG) is a new anticancer agent currently in clinical trials. The ability of 17-AAG to abrogate the function of heat-shock protein Hsp90 and modulate cellular sensitivity to anticancer agents has prompted recent research to use this compound in drug combination therapy. Here we report that 17-AAG has striking opposite effects on the activity of arsenic trioxide (ATO) and ara-C. Combination of 17-AAG with ATO exhibited a synergistic effect in leukemia cells, whereas coincubation of 17-AAG and ara-C showed antagonistic activity. Mechanistic studies revealed that ATO exerted cytotoxic action by reactive oxygen species generation, and activated Akt survival pathway. 17-AAG abrogated Akt activation and enhanced the activity of ATO. In contrast, treatment of leukemia cells with 17-AAG caused a G1 arrest, a decrease in DNA synthesis and reduced ara-C incorporation into DNA, leading to antagonism. The ability of 17-AAG to enhance the antileukemia activity of ATO was further demonstrated in primary leukemia cells isolated from patients with acute myeloid leukemia and chronic lymphocytic leukemia, including cells from refractory patients. Our data suggest that combination of 17-AAG and ATO may be an effective therapeutic regimen. Caution should be exercised in using 17-AAG together with ara-C, as their combination effects are schedule dependent.


Asunto(s)
Antineoplásicos/farmacología , Arsenicales/farmacología , Benzoquinonas/farmacología , Citarabina/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Lactamas Macrocíclicas/farmacología , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Mieloide/metabolismo , Óxidos/farmacología , Enfermedad Aguda , Apoptosis/efectos de los fármacos , Trióxido de Arsénico , Arsenicales/antagonistas & inhibidores , Citarabina/antagonistas & inhibidores , ADN/biosíntesis , ADN/efectos de los fármacos , ADN/metabolismo , Esquema de Medicación , Antagonismo de Drogas , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Quimioterapia Combinada , Fase G1/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/fisiología , Células HeLa , Humanos , Células Jurkat , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Mieloide/tratamiento farmacológico , Óxidos/antagonistas & inhibidores , Sensibilidad y Especificidad , Relación Estructura-Actividad , Células Tumorales Cultivadas
6.
Leuk Lymphoma ; 43(9): 1755-62, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12685828

RESUMEN

Resistance to purine analogs is emerging as a major problem in the management of patients with chronic lymphocytic leukemia (CLL). Most of these patients have already been exposed to and have become refractory to alkylating agents. To define the natural history of fludarabine (Fludara) refractory patients with CLL, we reviewed the response to first salvage therapy of 147 patients who were refractory to Fludara or had a remission less than six months in duration after a Fludara-containing regimen. Thirty-three (22%) patients responded to their first salvage attempt. However, the median survival was only 10 months. Responders survived significantly longer than non-responders. The most effective salvage regimens were combinations of purine analogs and cyclophosphamide. Patients still possibly sensitive to alkylating agents had a superior response than alkylating agent resistant or naive patients. Subsequent salvage therapy was administered to 61 patients. The most promising results noted in the group were transplantation and the use of Campath-1H antibody. The major morbidity and cause of death were associated with infections. The probability of infection was most strongly associated with the response to salvage therapy. Gram-positive organisms were most commonly associated with infection. However, gram-negative bacilli or opportunistic infection such as fungi, Pneumocystis carinii, acid-fast bacilli and legionella were prominent causes of infection. Fludara-refractory patients are a poor prognosis group and need more effective therapeutic regimens and well-designed infection prophylactic regimens.


Asunto(s)
Resistencia a Antineoplásicos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Terapia Recuperativa , Vidarabina/análogos & derivados , Vidarabina/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/uso terapéutico , Antineoplásicos Alquilantes/uso terapéutico , Ciclofosfamida/uso terapéutico , Femenino , Humanos , Leucemia Linfocítica Crónica de Células B/mortalidad , Masculino , Persona de Mediana Edad , Análisis Multivariante , Factores de Tiempo
7.
Cancer Res ; 61(24): 8723-9, 2001 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11751391

RESUMEN

Therapeutic nucleoside analogues such as ara-C, gemcitabine, and fludarabine exert their cytotoxic activity against cancer cells mainly by incorporation into DNA and disruption of further DNA synthesis, resulting in the triggering of apoptosis. However, the molecules that recognize the incorporated analogues in DNA and subsequently initiate the downstream cellular responses remain to be identified. Here, we report that the DNA-dependent protein kinase (DNA-PK) and p53 are able to form a protein complex that interacts with the gemcitabine-containing DNA and plays a role in signaling to apoptotic pathways. DNA-PK/Ku and p53 were copurified in a protein fraction that binds to gemcitabine-containing DNA in preference to normal DNA. Immunoprecipitation experiments revealed that the two proteins physically associate in a complex. Treatment with gemcitabine resulted in an increase of DNA-PK and p53 protein and an increase in the phosphorylation of p53 at Ser15. Furthermore, confocal microscopy demonstrated a colocalization of DNA-PK and p53 to the nucleus in cells treated with gemcitabine. The nuclear localization of the DNA-PK/p53 complex was coincident with the induction of apoptosis in these cells. Although the wild-type p53 present in the protein complex exhibited 3'-5' exonuclease activity, it was incapable of excising the incorporated gemcitabine from DNA. The binding of the p53/DNA-PK complex to DNA substantially blocked further DNA synthesis by DNA polymerases alpha and epsilon in vitro, indicating a stalling of this complex at the site of drug incorporation. These data suggest that DNA-PK and p53 may form a sensor complex that detects the disruption of DNA replication caused by nucleoside analogue incorporation and may subsequently signal for apoptosis.


Asunto(s)
Antígenos Nucleares , Antimetabolitos Antineoplásicos/farmacología , Daño del ADN , ADN Helicasas , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Antimetabolitos Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Unión Competitiva , Reparación del ADN , Replicación del ADN/efectos de los fármacos , ADN de Neoplasias/efectos de los fármacos , ADN de Neoplasias/metabolismo , Proteína Quinasa Activada por ADN , Proteínas de Unión al ADN/metabolismo , Desoxicitidina/metabolismo , Humanos , Autoantígeno Ku , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Proteínas Nucleares/metabolismo , Fosforilación , Células Tumorales Cultivadas , Gemcitabina
8.
Clin Cancer Res ; 7(11): 3580-9, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11705880

RESUMEN

PURPOSE: Chronic lymphocytic leukemia (CLL) lymphocytes respond to DNA alkylation by excision repair, with the extent of repair increasing as the cells acquire resistance to alkylating agents. Because incorporation of nucleotide analogues into the repair patches elicits death signals in quiescent cells, the increased capacity for excision repair in alkylator-resistant cells could facilitate incorporation of nucleotide analogues. We hypothesized that the mechanism-based interaction of nucleoside analogues with alkylating agents could elicit greater than additive killing of CLL cells. EXPERIMENTAL DESIGN: Lymphocytes from 50 patients with CLL that were not refractory to alkylators were treated in vitro with 4-hydroperoxycyclophosphamide (4-HC) with or without prior incubation with fludarabine nucleoside (F-ara-A) or with clofarabine (Cl-F-ara-A). DNA damage repair kinetics were determined by the single-cell gel electrophoresis (comet) assay. Cytotoxicity was assessed by staining with annexin V. RESULTS: CLL lymphocytes promptly initiated and completed excision repair in response to 4-HC. A 2-h preincubation with 10 microM F-ara-A or 10 microM Cl-F-ara-A inhibited the repair initiated by 4-HC, with inhibition peaking at the intracellular concentrations of 50 microM F-ara-ATP or 5 microM Cl-F-ara-ATP. Combining 4-HC with either F-ara-A or Cl-F-ara-A produced more than additive apoptotic cell death than the sum of each alone. The increase in cytotoxicity was proportional to the initial magnitude of the DNA incision and to the extent of repair inhibition by the nucleoside analogues, suggesting close correlation between the repair inhibition and induction of cell death. CONCLUSIONS: DNA repair, which is active in CLL lymphocytes, may be a biological target for facilitating the incorporation of nucleoside analogues and increasing their cytotoxicity. Thus, the increased repair capacity associated with resistant disease may be manipulated to therapeutic advantage.


Asunto(s)
Antineoplásicos/farmacología , Ciclofosfamida/análogos & derivados , Reparación del ADN/efectos de los fármacos , Leucemia Linfocítica Crónica de Células B/sangre , Linfocitos/efectos de los fármacos , Vidarabina/análogos & derivados , Nucleótidos de Adenina , Apoptosis/efectos de los fármacos , Arabinonucleósidos/farmacología , Arabinonucleotidos/farmacología , Clofarabina , Ciclofosfamida/farmacología , ADN/biosíntesis , ADN/efectos de los fármacos , ADN/genética , Relación Dosis-Respuesta a Droga , Humanos , Linfocitos/metabolismo , Nucleósidos/química , Nucleósidos/farmacología , Factores de Tiempo , Vidarabina/química , Vidarabina/farmacología
10.
Curr Opin Oncol ; 13(6): 484-90, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11673689

RESUMEN

The mammalian cell cycle is exquisitely controlled by the cyclin-dependent kinases, which regulate cell cycle progression. Cell cycle transitions are, in turn, controlled by checkpoints that monitor the integrity and replication status of the genetic material before cells commit to either replicate or segregate their DNA. On activation, checkpoints interface with cyclin-Cdk complexes to block the cell cycle. Pharmacologic compounds that exploit our current knowledge of cell cycle and checkpoint pathway regulation offer insights into the development of novel therapeutic strategies.


Asunto(s)
Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Quinasas Ciclina-Dependientes/metabolismo , Replicación del ADN , Diseño de Fármacos , Animales , Daño del ADN , Humanos , Mamíferos , Neoplasias/tratamiento farmacológico
11.
J Clin Oncol ; 19(15): 3483-9, 2001 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-11481354

RESUMEN

PURPOSE: To evaluate the efficacy, toxicity, and optimal dose rate of gemcitabine in adult patients with advanced soft tissue sarcomas (STS) by comparing levels of gemcitabine triphosphate (GTP) in peripheral-blood mononuclear cells (PBMCs) of patients receiving two different dose rates. PATIENTS AND METHODS: Fifty-six assessable patients with STS (17 gastrointestinal [GI] leiomyosarcomas and 39 other histologies) were treated on a two-arm phase II study. Gemcitabine was given at 1 g/m2 as a 30-minute infusion weekly for up to 7 weeks followed by 1 week of rest and reassessment of tumor. Subsequent cycles were given at 1 g/m2 weekly for 3 weeks followed by 1 week of rest. Nine patients underwent cellular pharmacologic studies at two different dose rates (1 g/m2 over a standard 30-minute infusion on week 1 and over pharmacologically based infusion of 150 minutes on week 2) to evaluate GTP levels in PBMCs. RESULTS: Seven partial responses were noted among 39 patients, for an overall response rate of 18% (95% confidence interval, 7% to 29%). Median duration of response was 3.5 months (range, 2 to 13 months). Four of 10 patients with non-GI leiomyosarcomas achieved a partial response. No objective responses were noted in 17 patients with GI leiomyosarcomas. One patient had a mixed response. Median time to progression for all patients (both arms) was 3 months; median survival was 13.9 months. Treatment was generally well tolerated. Comparison of cellular pharmacology demonstrated a significant 1.4-fold increase in the concentration of GTP with the 150-minute infusion. CONCLUSION: Given the limited therapeutic armamentarium for STS, the activity of gemcitabine is encouraging. Its potential for combination therapy in the salvage setting should be studied with pharmacologically guided fixed dose-rate infusion.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Desoxicitidina/administración & dosificación , Neoplasias Gastrointestinales/tratamiento farmacológico , Leiomiosarcoma/tratamiento farmacológico , Adulto , Anciano , Antimetabolitos Antineoplásicos/efectos adversos , Antimetabolitos Antineoplásicos/farmacocinética , Desoxicitidina/efectos adversos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacocinética , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Neoplasias Gastrointestinales/sangre , Humanos , Infusiones Intravenosas , Leiomiosarcoma/sangre , Leucocitos Mononucleares/metabolismo , Masculino , Persona de Mediana Edad , Fosfatos/sangre , Sarcoma/sangre , Sarcoma/tratamiento farmacológico , Neoplasias de los Tejidos Blandos/sangre , Neoplasias de los Tejidos Blandos/tratamiento farmacológico , Gemcitabina
12.
Cancer ; 92(1): 194-9, 2001 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-11443627

RESUMEN

BACKGROUND: The authors investigated the maximum tolerated dose (MTD) and dose limiting toxicity (DLT) associated with the addition of a biomodulating dose of gemcitabine to an established regimen of ifosfamide and doxorubicin as part of a program to explore the potential of low-dose gemcitabine to modulate the activity of alkylating agents. METHODS: A Phase I trial was carried out in a population of patients with bladder or pelvic carcinoma for whom no standard therapy was available. Doses of ifosfamide and doxorubicin were held fixed at 2 g/m(2) for 4 days and 20 mg/m(2) for 3 days, respectively. Gemcitabine was given on Day 2 and Day 4 at doses of 90 mg/m(2), 150 mg/m(2), and 200 mg/m(2) per dose. RESULTS: A total of 18 patients received 53 courses of therapy. Myelosuppression was dose limiting. Nonhematologic toxicity also was significant, with 10 of 18 patients experiencing toxicity of Grade 3 or greater. For previously untreated patients with an intact performance status, the MTD for gemcitabine in this context was at least 150 mg/m(2) per dose. According to an intent-to-treat analysis, 11 of 18 patients demonstrated a clinically significant response to this regimen. CONCLUSIONS: The regimen of ifosfamide and doxorubicin with the addition of gemcitabine was significantly toxic but has promising activity. Based on the observed activity and the generally reversible nature of the toxicity, the authors have initiated a Phase II trial of this regimen in patients with untreated, metastatic urothelial carcinoma.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Desoxicitidina/administración & dosificación , Neoplasias Pélvicas/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Adulto , Anciano , Alquilantes/administración & dosificación , Antimetabolitos Antineoplásicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Desoxicitidina/efectos adversos , Desoxicitidina/análogos & derivados , Doxorrubicina/administración & dosificación , Femenino , Humanos , Ifosfamida/administración & dosificación , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Gemcitabina
13.
Biochem Pharmacol ; 61(12): 1497-507, 2001 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-11377379

RESUMEN

The pharmacokinetics and pharmacodynamics of the novel clinical candidate 2'-C-cyano-2'-deoxy-1-beta-D-arabino-pentofuranosylcytosine (CNDAC) were investigated in human lymphoblastoid CCRF-CEM cells and human myeloblastic leukemia ML-1 cells. Formation of CNDAC 5'-mono-, di-, and triphosphate (CNDACTP) was concentration-dependent; nucleotide accumulation was greater in the lymphoid cells than in the myeloid cells. The nucleotides were eliminated with linear kinetics from both lines, but were retained more effectively by the ML-1 cells. DNA synthesis was selectively inhibited by a 4-hr treatment with CNDAC in CCRF-CEM and ML-1 cells; the IC(50) values were 1 and 0.8 microM, respectively. Evaluation of the polymerization reaction of a primer on an M13mp19(+) template by human DNA polymerase alpha indicated that CNDACTP was incorporated effectively (K(m) = 0.22 microM) opposite a complementary dGMP in the template strand. CNDACTP competed with the normal substrate, dCTP, for incorporation, and the two nucleotides showed similar substrate efficiencies (V(max)/K(m): dCTP = 0.91; CNDACTP = 0.77). Primer extension was potently inhibited by CNDAC triphosphate (K(i) = 23 nM); once the analog had been incorporated, further extension was not observed in vitro, suggesting that primers containing a 3'-terminal nucleotide analog were high K(m) substrates for polymerase alpha. Thus, the ability of human leukemia cells to effectively accumulate and retain CNDACTP, coupled with the favorable kinetics of competition for incorporation into DNA, and the relatively strong ability of the analog to terminate further extension, are likely to contribute to the cytotoxic action of CNDAC.


Asunto(s)
Antineoplásicos/farmacología , Citarabina/farmacología , ADN/efectos de los fármacos , Antineoplásicos/metabolismo , División Celular/efectos de los fármacos , Citarabina/análogos & derivados , Citarabina/metabolismo , ADN/biosíntesis , Nucleótidos de Desoxicitosina/metabolismo , Desoxirribonucleótidos/metabolismo , Humanos , ARN/biosíntesis , ARN/efectos de los fármacos , Células Tumorales Cultivadas
14.
J Clin Oncol ; 19(8): 2142-52, 2001 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-11304766

RESUMEN

PURPOSE: A pilot protocol was designed to evaluate the efficacy of fludarabine with nelarabine (the prodrug of arabinosylguanine [ara-G]) in patients with hematologic malignancies. The cellular pharmacokinetics was investigated to seek a relationship between response and accumulation of ara-G triphosphate (ara-GTP) in circulating leukemia cells and to evaluate biochemical modulation of cellular ara-GTP metabolism by fludarabine triphosphate. PATIENTS AND METHODS: Nine of the 13 total patients had indolent leukemias, including six whose disease failed prior fludarabine therapy. Two patients had T-acute lymphoblastic leukemia, one had chronic myelogenous leukemia, and one had mycosis fungoides. Nelarabine (1.2 g/m(2)) was infused on days 1, 3, and 5. On days 3 and 5, fludarabine (30 mg/m(2)) was administered 4 hours before the nelarabine infusion. Plasma and cellular pharmacokinetic measurements were conducted during the first 5 days. RESULTS: Seven patients had a partial or complete response, six of whom had indolent leukemias. The disease in four responders had failed prior fludarabine therapy. The median peak intracellular concentrations of ara-GTP were significantly different (P =.001) in responders (890 micromol/L, n = 6) and nonresponders (30 micromol/L, n = 6). Also, there was a direct relationship between the peak fludarabine triphosphate and ara-GTP in each patient (r = 0.85). The cellular elimination of ara-GTP was slow (median, 35 hours; range, 18 to > 48 hours). The ratio of ara-GTP to its normal counterpart, deoxyguanosine triphosphate, was higher in each patient (median, 42; range, 14 to 1,092) than that of fludarabine triphosphate to its normal counterpart, deoxyadenosine triphosphate (median, 2.2; range, 0.2 to 27). CONCLUSION: Fludarabine plus nelarabine is an effective, well-tolerated regimen against leukemias. Clinical responses suggest the need for further exploration of nelarabine against fludarabine-refractory diseases. Determination of ara-GTP levels in the target tumor population may provide a prognostic test for the activity of nelarabine.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Prolinfocítica/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Vidarabina/análogos & derivados , Adulto , Anciano , Anciano de 80 o más Años , Arabinonucleósidos/administración & dosificación , Arabinonucleósidos/farmacocinética , Arabinonucleósidos/farmacología , Arabinonucleotidos/análisis , Arabinonucleotidos/metabolismo , Biomarcadores/análisis , Femenino , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/análisis , Guanosina Trifosfato/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Resultado del Tratamiento , Vidarabina/administración & dosificación , Vidarabina/farmacocinética , Vidarabina/farmacología
15.
Mol Pharmacol ; 59(4): 725-31, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11259616

RESUMEN

The mechanism of 2'-C-cyano-2'-deoxy-1-beta-D-arabino-pentofuranosylcytosine (CNDAC) action was investigated in human lymphoblastoid CEM cells and myeloblastic leukemia ML-1 cells. CNDAC was metabolized to its 5'-triphosphate and incorporated into DNA, which was associated with inhibition of DNA synthesis. After incubation of cells with [(3)H]CNDAC, metabolites were detected in 3'-->5' phosphodiester linkage and at the 3' terminus of cellular DNA. Specific enzymatic hydrolysis of DNA demonstrated that the parent nucleoside and its 2'-epimer 2'-C-cyano-2'-deoxy-2-ribo-pentofuranosylcytosine accounted for approximately 65% of the total analogs incorporated into DNA and essentially all of the drug in the 3'-->5' phosphodiester linkage. In contrast, all detectable radioactivity at 3' termini was associated with 2'-C-cyano-2',3'-didehydro-2',3'-dideoxycytidine. This de facto DNA chain-terminating nucleotide arises from an electronic characteristic and cleavage of the 3'-phosphodiester bond subsequent to the addition of a nucleotide to the incorporated CNDAC moiety by beta-elimination, a process that generates a single strand break in DNA. Investigation of the biological consequences of these actions indicated that, after incubation with cytostatic concentrations of CNDAC, cell cycle progression was delayed during S phase, but that cells arrested predominantly in the G(2) phase. This differed from the S phase-arresting actions of ara-C and gemcitabine, other deoxycytidine analogs that inhibit DNA replication but do not cause strand breaks. Thus, once incorporated into DNA, the CNDAC molecule appears to act by a dual mechanism that 1) delays the progress of further DNA replication, but 2) upon addition of a deoxynucleotide results in the conversion of the incorporated analog to a de facto DNA chain terminator at the 3' terminus of a single strand break. It is likely that DNA strand breaks trigger cell cycle arrest in G(2).


Asunto(s)
Antineoplásicos/metabolismo , Citarabina/metabolismo , ADN de Neoplasias/efectos de los fármacos , Fase G2/efectos de los fármacos , Leucemia Mieloide Aguda/metabolismo , Linfocitos/metabolismo , Antineoplásicos/farmacología , Proteína Quinasa CDC2/metabolismo , Rotura Cromosómica/genética , Ciclina B/metabolismo , Ciclina B1 , Citarabina/análogos & derivados , Citarabina/farmacología , ADN de Neoplasias/metabolismo , Desoxicitidina/análogos & derivados , Desoxicitidina/metabolismo , Desoxicitidina/farmacología , Humanos , Leucemia Mieloide Aguda/genética , Linfocitos/citología , Linfocitos/efectos de los fármacos , Fosforilación/efectos de los fármacos , ARN/efectos de los fármacos , ARN/metabolismo , Células Tumorales Cultivadas , Gemcitabina
16.
Cancer Res ; 61(3): 1065-72, 2001 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11221834

RESUMEN

The mechanisms of resistance to nucleoside analogues established in preclinical models are rarely found in primary tumors resistant to therapy with these agents. We tested the hypothesis that cells sense sublethal incorporation of analogues into DNA during replication and react by arresting further DNA synthesis and cell cycle progression. After removal of drug, cells may be able to repair damaged DNA and continue proliferation, thus escaping nucleoside analogue toxicity. As a corollary, we evaluated whether dysregulation of this mechanism causes cell death. Using gemcitabine as a model of S-phase-specific nucleoside analogues in human acute myelogenous leukemia ML-1 cells, we found that DNA synthesis decreased, cells arrested in S-phase transit, and 60-70% of the population accumulated in S-phase in response to cytostatic conditions. Proliferation continued after washing the cells into drug-free medium. S-phase-arrested cells were then treated with otherwise nontoxic concentrations of UCN-01, which caused rapid onset of apoptosis without cell cycle progression specifically in cells with an S-phase DNA content. Thus, S-phase arrest by nucleoside analogues sensitizes cells to UCN-01, which appears to activate signaling for death mechanisms and/or inhibit survival pathways. These results differ from those in cells arrested at the G2 checkpoint, in which UCN-01 abrogates cell cycle arrest, permitting cells to progress in the cell cycle before apoptosis.


Asunto(s)
Alcaloides/farmacología , Antimetabolitos Antineoplásicos/farmacología , Antineoplásicos/farmacología , Desoxicitidina/farmacología , Fase S/efectos de los fármacos , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/patología , Estaurosporina/análogos & derivados , Células Tumorales Cultivadas , Gemcitabina
17.
Cancer Chemother Pharmacol ; 46(5): 382-6, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11127942

RESUMEN

PURPOSE: To determine the efficacy and pharmacokinetics of intraventricular cytosine arabinoside (Ara-C) as front-line treatment for leptomeningeal metastases from breast cancer. METHODS: Ten patients newly diagnosed with leptomeningeal metastases (LMM) from breast cancer were treated with 100 mg intraventricular cytosine arabinoside (IVT Ara-C) via an Ommaya reservoir. Treatment was administered three times a week for 2 weeks, then once a week for 4 weeks, and then once every 6 weeks for four cycles to responding patients. Nine patients were evaluable clinically, and seven patients underwent testing to determine the pharmacokinetic profile of Ara-C in the cerebrospinal fluid (CSF). RESULTS: Two patients had partial responses lasting 9 and 40 weeks, respectively. Two other patients had stable disease. The median survival duration was 30 weeks (range: 5-58 weeks). Seven patients died from LMM. Acute toxic effects associated with IVT Ara-C included meningismus, nausea, vomiting, and myelosuppression. The median peak Ara-C level in CSF was 16.69+/-6.30 mM (SD). The half life for elimination was 1.45+/-0.61 h (SD) There was no drug accumulation between courses. Neuropsychological evaluations were completed in eight patients, six (75%) of whom had preexisting cognitive deficits. Their condition generally improved over the course of treatment until the LMM progressed. No neurotoxic side effects of IVT Ara-C were observed in the two patients who had normal baseline cognitive assessments. CONCLUSIONS: IVT Ara-C at this dose and schedule has minimal activity as initial treatment for LMM from breast cancer despite achievement of high peak levels of the drug in the cerebrospinal fluid. A liposomal Ara-C formulation is currently under investigation.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Neoplasias de la Mama/patología , Citarabina/uso terapéutico , Neoplasias Meníngeas/tratamiento farmacológico , Neoplasias Meníngeas/secundario , Adulto , Anciano , Antimetabolitos Antineoplásicos/efectos adversos , Antimetabolitos Antineoplásicos/farmacocinética , Cognición/fisiología , Citarabina/efectos adversos , Citarabina/farmacocinética , Femenino , Humanos , Neoplasias Meníngeas/metabolismo , Persona de Mediana Edad , Análisis de Supervivencia , Resultado del Tratamiento
18.
Cancer Res ; 60(22): 6408-15, 2000 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-11103806

RESUMEN

Activation of the c-Jun NH2-terminal kinase type 1 (JNK1) signaling pathway is often associated with apoptosis. In this report, we elucidated the role of this kinase in the programmed cell death induced by the nucleoside analogue 9-beta-D-arabinosyl-2-fluoroadenine (F-ara-A). Treatment of ML-1 cells with 3 or 10 microM F-ara-A specifically killed cells in the S-phase of the population. Incorporation of F-ara-ATP, the nucleoside triphosphate of F-ara-A, into DNA resulted in the activation of JNK1 in a time- and dose-dependent fashion. Activation of JNK1 temporally preceded DNA fragmentation. When incorporation of F-ara-A into DNA was blocked by pretreatment of the cells with aphidicolin to inhibit DNA synthesis, neither JNK1 signaling nor apoptosis was evident. Furthermore, inhibition of JNK1 by treatment of the cells with forskolin or by pretreatment with an antisense oligonucleotide directed against JNK1 mRNA resulted in a decrease in F-ara-A-induced apoptosis. Finally, the JNK1 signaling pathway appeared to be upstream to that of the effector caspases in nucleoside analogue-induced apoptosis. Thus, our data strongly suggest that JNK1 is involved in transduction of F-ara-A-induced distress signals into an apoptotic response.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/fisiología , Daño del ADN/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Fosfato de Vidarabina/análogos & derivados , Vidarabina/análogos & derivados , Vidarabina/farmacología , Apoptosis/efectos de los fármacos , Arabinonucleotidos/metabolismo , Caspasas/metabolismo , ADN/efectos de los fármacos , ADN/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Leucemia Mieloide , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 8 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fase S/fisiología , Células Tumorales Cultivadas , Células U937 , Vidarabina/metabolismo , Fosfato de Vidarabina/metabolismo
19.
Nature ; 407(6802): 390-5, 2000 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-11014196

RESUMEN

Superoxide dismutases (SOD) are essential enzymes that eliminate superoxide radical (O2-) and thus protect cells from damage induced by free radicals. The active O2- production and low SOD activity in cancer cells may render the malignant cells highly dependent on SOD for survival and sensitive to inhibition of SOD. Here we report that certain oestrogen derivatives selectively kill human leukaemia cells but not normal lymphocytes. Using complementary DNA microarray and biochemical approaches, we identify SOD as a target of this drug action and show that chemical modifications at the 2-carbon (2-OH, 2-OCH3) of the derivatives are essential for SOD inhibition and for apoptosis induction. Inhibition of SOD causes accumulation of cellular O2- and leads to free-radical-mediated damage to mitochondrial membranes, the release of cytochrome c from mitochondria and apoptosis of the cancer cells. Our results indicate that targeting SOD may be a promising approach to the selective killing of cancer cells, and that mechanism-based combinations of SOD inhibitors with free-radical-producing agents may have clinical applications.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Estradiol/análogos & derivados , Estradiol/farmacología , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia/tratamiento farmacológico , Superóxido Dismutasa/antagonistas & inhibidores , 2-Metoxiestradiol , Apoptosis , Ciclo Celular , Ensayos de Selección de Medicamentos Antitumorales , Regulación Enzimológica de la Expresión Génica , Células HL-60 , Humanos , Linfocitos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Relación Estructura-Actividad , Superóxido Dismutasa/genética , Células Tumorales Cultivadas
20.
J Chromatogr B Biomed Sci Appl ; 745(2): 421-30, 2000 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-11043760

RESUMEN

A gradient anion-exchange high-performance liquid chromatographic assay was developed for the simultaneous determination and quantitation of the cytotoxic triphosphates of arabinosylguanine (ara-GTP) and fludarabine (F-ara-ATP). The method was validated with respect to selectivity, recovery, linearity, precision, and accuracy using authentic standards. To test this assay in a more complex biological matrix, perchloric acid extracts of circulating human leukemia cells spiked with known concentrations of ara-GTP and F-ara-ATP were examined. Finally, to assess the clinical utility of our method, perchloric acid extracts of circulating human leukemia cells isolated from patients treated with fludarabine and nelarabine were analyzed. The range of quantitation was 0.0125-10 nmol for the ara- and native NTPs in cellular extracts. This assay should be helpful in establishing the mechanistic rationales for drug scheduling and combinations of nelarabine and fludarabine, and for correlating the therapeutic efficacy and levels of the cytotoxic triphosphates in target cells.


Asunto(s)
Arabinonucleotidos/análisis , Cromatografía Líquida de Alta Presión/métodos , Guanosina Trifosfato/análogos & derivados , Guanosina Trifosfato/análisis , Vidarabina/análogos & derivados , Vidarabina/análisis , Calibración , Humanos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Espectrofotometría Ultravioleta , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...