Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Ann Oncol ; 26(7): 1481-7, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25899782

RESUMEN

BACKGROUND: The use of Escherichia coli purine nucleoside phosphorylase (PNP) to activate fludarabine has demonstrated safety and antitumor activity during preclinical analysis and has been approved for clinical investigation. PATIENTS AND METHODS: A first-in-human phase I clinical trial (NCT 01310179; IND 14271) was initiated to evaluate safety and efficacy of an intratumoral injection of adenoviral vector expressing E. coli PNP in combination with intravenous fludarabine for the treatment of solid tumors. The study was designed with escalating doses of fludarabine in the first three cohorts (15, 45, and 75 mg/m(2)) and escalating virus in the fourth (10(11)-10(12) viral particles, VP). RESULTS: All 12 study subjects completed therapy without dose-limiting toxicity. Tumor size change from baseline to final measurement demonstrated a dose-dependent response, with 5 of 6 patients in cohorts 3 and 4 achieving significant tumor regression compared with 0 responsive subjects in cohorts 1 and 2. The overall adverse event rate was not dose-dependent. Most common adverse events included pain at the viral injection site (92%), drainage/itching/burning (50%), fatigue (50%), and fever/chills/influenza-like symptoms (42%). Analysis of serum confirmed the lack of systemic exposure to fluoroadenine. Antibody response to adenovirus was detected in two patients, suggesting that neutralizing immune response is not a barrier to efficacy. CONCLUSIONS: This first-in-human clinical trial found that localized generation of fluoroadenine within tumor tissues using E. coli PNP and fludarabine is safe and effective. The pronounced effect on tumor volume after a single treatment cycle suggests that phase II studies are warranted. CLINICALTRIALSGOV IDENTIFIER: NCT01310179.


Asunto(s)
Escherichia coli/enzimología , Terapia Genética , Vectores Genéticos/uso terapéutico , Neoplasias/genética , Neoplasias/terapia , Purina-Nucleósido Fosforilasa/administración & dosificación , Vidarabina/análogos & derivados , Adenoviridae/genética , Anciano , Anciano de 80 o más Años , Terapia Combinada , Relación Dosis-Respuesta a Droga , Femenino , Estudios de Seguimiento , Humanos , Inyecciones Intralesiones , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias/patología , Pronóstico , Purina-Nucleósido Fosforilasa/genética , Células Tumorales Cultivadas , Vidarabina/uso terapéutico
2.
Cancer Res ; 36(11 Pt 1): 3917-23, 1976 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-975040

RESUMEN

8-Azainosine (8-aza-HR) is of interest because of its activity against experimental tumors. Metabolic studies in cell cultures were performed with 8-aza-HR and with the structurally related nucleoside, 8-azaadenosine (9-beta-D-ribofuranosyl-8-azaadenine) (8-aza-AR), which has a lower degree of antitumor activity than does 8-aza-HR. In H. Ep. 2 cells and in Ca755 cells, both 14C-labeled nucleosides were metabolized to nucleotides of 8-azaadenine (8-aza-A) and 8-azaguanine (8-aza-G) and incorporated into polynucleotides as 8-aza-A and 8-aza-G. 8-Aza HR was incorporated primarily as 8-aza-G, whereas 8-aza-AR was incorporated about equally as 8-aza-A and 8-aza-G. In H. Ep. 2 cells, the extent of incorporation of 8-aza-HR as 8-aza-G was about one-half that found when [14C]-8-aza-G was the precursor. In the H. Ep. 2/FA/FAR cell line, 8-aza-AR and 8-aza-HR were metabolized similarly, in that both were incorporated into polynucleotides principally as 8-aza-G; apparently, in this cell line which is deficient in adenosine kinase and adenine phosphoribosyltransferase, 8-aza-AR is metabolized by conversion to 8-aza-HR. A cell line (H. Ep 2/8-aza HR), which was resistant to 8-aza-HR but sensitive to 8-aza-AR and which retained hypoxanthine (guanine)-phosphoribosyltransferase activity, metabolized 8-aza-HR to only a small extent. However, in this cell-line, 8-aza-AR was more extensively metabolized and was incorporated primarily as 8-aza-A. The failure of these cells to convert 8-aza-AR or 8-aza-HR to 8-aza-G indicates that the basis for resistance may be a change in the substrate specificities of the enzymes of guanosine monophosphate synthesis such that these cells no longer effectively convert 8-azainosine monophosphate to 8-azaguanosine monophosphate. 8-Aza-AR was a potent inhibitor of purine synthesis de novo, but 8-aza-HR, at concentrations much higher than the inhibitory concentration of 8-aza-AR, did not inhibit this process. In H. Ep. 2 cells, 8-aza-HR blocked the conversion of orotic acid to uridine nucleotides and caused an accumulation of orotidine. This inhibition of pyrimidine biosynthesis apparently does not contribute significantly to the cytotoxicity of 8-aza-HR because uridine provided no degree of reversal of its inhibition of the growth of cell cultures.


Asunto(s)
Adenosina/análogos & derivados , Inosina/análogos & derivados , Adenosina/metabolismo , Adenosina/farmacología , Animales , Azaguanina/metabolismo , Línea Celular , Cromatografía en Capa Delgada , Humanos , Técnicas In Vitro , Inosina/farmacología , Ratones , Ácido Orótico/metabolismo , Polinucleótidos/biosíntesis , Purinas/biosíntesis , Nucleótidos de Pirimidina/biosíntesis , Timidina/metabolismo , Uridina/metabolismo
3.
Cancer Res ; 54(7): 1742-5, 1994 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-8137289

RESUMEN

6-Thio-2'-deoxyguanosine (T-dGuo) has been reported to be both phosphorylated by deoxycytidine kinase and converted to 6-thioguanine by purine nucleoside phosphorylase (PNP). Combination of T-dGuo with an inhibitor of PNP would be expected to generate the 5'-triphosphate of T-dGuo and limit or prevent the formation of 6-thioguanosine triphosphate. Because the incorporation of 6-thioguanine into DNA is believed to be primarily responsible for the antitumor activity of the thiopurines, this treatment might result in enhanced activity against certain tumors, particularly those of T-cell origin. We have evaluated the metabolic basis of this strategy by examining the effects of 9-benzyl-9-deazaguanine (BDG), a potent inhibitor of PNP, on the metabolism of T-dGuo in CEM cells. The concentration of T-dGuo required to inhibit cell growth by 50% was approximately 50-fold greater in the presence of 8.0 microM BDG than in its absence. As expected, the addition of BDG to cells treated with T-dGuo prevented the metabolism of T-dGuo to 6-thio-guanine-containing ribo-nucleotides, but, unexpectedly, no 6-thio-2'-deoxyguanosine 5'-triphosphate was detected. In cells treated with T-dGuo plus BDG, the major phosphorylated metabolite was T-dGMP. These results indicated that even in the absence of PNP activity, T-dGuo cannot be phosphorylated directly to 6-thio-2'-deoxyguanosine 5'-triphosphate due to the inability of guanylate kinase to utilize T-dGMP as a substrate.


Asunto(s)
Compuestos de Bencilo/farmacología , Desoxiguanosina/análogos & derivados , Guanina/análogos & derivados , Purina-Nucleósido Fosforilasa/antagonistas & inhibidores , Tionucleósidos/toxicidad , Biotransformación , División Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Desoxiguanosina/metabolismo , Desoxiguanosina/toxicidad , Relación Dosis-Respuesta a Droga , Guanina/farmacología , Humanos , Tionucleósidos/metabolismo
4.
Artículo en Inglés | MEDLINE | ID: mdl-16248054

RESUMEN

A novel series of 6-methylpurine nucleoside derivatives with substitutions at 5-position have been synthesised These compounds bear a 5'-heterocycle such as triazole or a imidazole with a two carbon chain, and an ether, thio ether or amine. To extend the SAR study of 2-fluoroadenine and 6-methyl purine nucleosides, their corresponding alpha-linker nucleosides with L-xylose and L-lyxose were also synthesized. All of these compounds have been evaluated for their substrate activity with E. coli PNP.


Asunto(s)
Adenina/análogos & derivados , Terapia Genética/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Nucleósidos/síntesis química , Profármacos/farmacología , Purinas/química , Adenina/farmacología , Antineoplásicos/farmacología , Carbono/química , Escherichia coli/enzimología , Humanos , Modelos Químicos , Mutación , Nucleósidos/química , Profármacos/química , Nucleósidos de Purina/química , Purina-Nucleósido Fosforilasa/química , Especificidad por Sustrato , Xilosa/química
5.
Hum Gene Ther ; 8(14): 1637-44, 1997 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-9322865

RESUMEN

We have developed a new strategy for the gene therapy of cancer based on the activation of purine nucleoside analogs by transduced E. coli purine nucleoside phosphorylase (PNP, E.C. 2.4.2.1). The approach is designed to generate antimetabolites intracellularly that would be too toxic for systemic administration. To determine whether this strategy could be used to kill tumor cells without host toxicity, nude mice bearing human malignant D54MG glioma tumors expressing E. coli PNP (D54-PNP) were treated with either 6-methylpurine-2'-deoxyriboside (MeP-dR) or arabinofuranosyl-2-fluoroadenine monophosphate (F-araAMP, fludarabine, a precursor of F-araA). Both prodrugs exhibited significant antitumor activity against established D54-PNP tumors at doses that produced no discernible systemic toxicity. Significantly, MeP-dR was curative against this slow growing solid tumor after only 3 doses. The antitumor effects showed a dose dependence on both the amount of prodrug given and the level of E. coli PNP expression within tumor xenografts. These results indicated that a strategy using E. coli PNP to create highly toxic, membrane permeant compounds that kill both replicating and nonreplicating cells is feasible in vivo, further supporting development of this cancer gene therapy approach.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Terapia Genética/métodos , Glioma/tratamiento farmacológico , Profármacos/farmacología , Purina-Nucleósido Fosforilasa/fisiología , Animales , Antimetabolitos Antineoplásicos/toxicidad , Escherichia coli/enzimología , Escherichia coli/genética , Vectores Genéticos/genética , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Nucleósidos de Purina/uso terapéutico , Nucleósidos de Purina/toxicidad , Purina-Nucleósido Fosforilasa/genética , Retroviridae/genética , Fosfato de Vidarabina/análogos & derivados , Fosfato de Vidarabina/uso terapéutico , Fosfato de Vidarabina/toxicidad
6.
J Med Chem ; 29(10): 2069-74, 1986 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-3489838

RESUMEN

The synthesis and characterization of 8-amino-6-fluoro-9-beta-D-ribofuranosyl-9H-purine (3a) are presented. This compound is a substrate for adenosine deaminase and adenosine kinase. In L1210 cells 3a is converted to 8-aminoinosine monophosphate (4b), apparently by the action of AMP deaminase on the monophosphate of 3a, as well as to the triphosphate derivative of 3a. Pentostatin was used to inhibit adenosine deaminase, and coformycin was used to inhibit AMP deaminase in experiments designed to delineate the metabolic fate of 3a. Pentostatin was without influence on the cytotoxicity of 3a, but coformycin potentiated the cytotoxicity. The potentiation was associated with an increased cellular concentration of phosphates of 3a and a decreased concentration of 4b.


Asunto(s)
Antineoplásicos/síntesis química , Nucleósidos de Purina/síntesis química , AMP Desaminasa/antagonistas & inhibidores , Inhibidores de la Adenosina Desaminasa , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Coformicina/análogos & derivados , Coformicina/farmacología , Inosina Monofosfato/metabolismo , Cinética , Leucemia L1210/metabolismo , Pentostatina , Nucleósidos de Purina/farmacología
7.
Biochem Pharmacol ; 37(7): 1233-44, 1988 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-3355597

RESUMEN

3-Deazaadenine, 3-deazaadenosine, and the carbocyclic analog of 3-deazaadenosine produced similar effects on nucleotide pools of L1210 cells in culture: each caused an increase in IMP and a decrease in adenine nucleotides and had no effect on nucleotides of uracil and cytosine. Concentrations of 50-100 microM were required to produce these effects. Although 3-deazaadenosine and carbocyclic 3-deazaadenosine are known to be potent inhibitors of adenosylhomocysteine hydrolase, the effects on nucleotide pools apparently are not mediated via this inhibition because they are also produced by the base, 3-deazaadenine, and because the concentrations required are higher than those required to inhibit the hydrolase. Cells grown in the presence of 3-deazaadenine or 3-deazaadenosine contained phosphates of 3-deazaadenosine (the mono- and triphosphates were isolated); from cells grown in the presence of the carbocyclic analog of 3-deazaadenosine, the monophosphate was isolated, but evidence for the presence of the triphosphate was not obtained. A cell-free supernatant fraction from L1210 cells supplemented with ATP catalyzed the formation of monophosphates from 3-deazaadenosine or carbocyclic 3-deazaadenosine, and a cell-free supernatant fraction supplemented with 5-phosphoribosyl 1-pyrophosphate (PRPP) catalyzed the formation of 3-deaza-AMP from 3-deazaadenine. Adenosine kinase apparently was not solely responsible for the phosphorylation of the nucleosides because a cell line that lacked this enzyme converted 3-deazaadenosine to phosphates. No evidence was obtained that the effects on nucleotide pools resulted from a block of the IMP-AMP conversion, but the results could be rationalized as a consequence of increased AMP deaminase activity. This explanation is supported by two observations: (a) coformycin, an inhibitor of AMP deaminase, prevented the effects on nucleotide pools, and (b) 3-deazaadenine decreased the conversion of carbocyclic adenosine to carbocyclic ATP and increased its conversion to carbocyclic GTP. The latter conversion requires the action of AMP deaminase and the observed effects can be rationalized by a nucleoside analog-mediated increase in AMP deaminase activity. Because these effects on nucleotide pools are produced only by concentrations higher than those required to inhibit adenosylhomocysteine hydrolase, they may not contribute significantly to the biological effects of 3-deazaadenosine or carbocyclic 3-deazaadenosine.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
AMP Desaminasa/fisiología , Adenina/análogos & derivados , Antibacterianos/farmacología , Nucleótido Desaminasas/fisiología , Nucleótidos/análisis , Tubercidina/análogos & derivados , Tubercidina/farmacología , Adenina/metabolismo , Adenina/farmacología , Adenosina Quinasa/fisiología , Alanina/análogos & derivados , Alanina/farmacología , Aminoglicósidos , Animales , Supervivencia Celular/efectos de los fármacos , Coformicina/farmacología , Hipoxantina , Hipoxantinas/metabolismo , Ratones , Factores de Tiempo , Tubercidina/metabolismo
8.
Biochem Pharmacol ; 55(10): 1673-81, 1998 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-9634004

RESUMEN

Activation of purine nucleoside analogs by Escherichia coli purine nucleoside phosphorylase (PNP) is being evaluated as a suicide gene therapy strategy for the treatment of cancer. Because the mechanisms of action of two toxic purine bases, 6-methylpurine (MeP) and 2-fluoroadenine (F-Ade), that are generated by this approach are poorly understood, mechanistic studies were initiated to learn how these compounds differ from agents that are being used currently. The concentration of F-Ade, MeP, or 5-fluorouracil required to inhibit CEM cell growth by 50% after a 4-hr incubation was 0.15, 9, or 120 microM, respectively. F-Ade and MeP were also toxic to quiescent MRC-5, CEM, and Balb 3T3 cells. Treatment of CEM, MRC-5, or Balb 3T3 cells with either F-Ade or MeP resulted in the inhibition of protein, RNA, and DNA syntheses. CEM cells converted F-Ade and MeP to F-ATP and MeP-ribonucleoside triphosphate (MeP-R-TP), respectively. The half-life for disappearance of HeP-ribonucleoside triphosphate from CEM cells was approximately 48 hr, whereas the half-lives of F-ATP and ATP were approximately 5 hr. Both MeP and F-Ade were incorporated into the RNA and DNA of CEM cells. These studies indicated that the mechanisms of action of F-Ade and MeP were quite different from those of other anticancer agents, and suggested that the generation of these agents in tumor cells by E. coli PNP could result in significant advantages over those generated by either herpes simplex virus thymidine kinase or E. coli cytosine deaminase. These advantages include a novel mechanism of action resulting in toxicity to nonproliferating and proliferating tumor cells and the high potency of these agents during short-term treatment.


Asunto(s)
Adenina/análogos & derivados , Purinas/metabolismo , Células 3T3 , Adenina/metabolismo , Animales , División Celular/efectos de los fármacos , Línea Celular , Cicloheximida/farmacología , ADN/efectos de los fármacos , ADN/metabolismo , Fluorouracilo/farmacología , Humanos , Ratones , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Inhibidores de la Síntesis de la Proteína/farmacología , ARN/efectos de los fármacos , ARN/metabolismo
9.
Biochem Pharmacol ; 40(7): 1515-22, 1990 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-2171531

RESUMEN

The carbocyclic analog of 2'-deoxyguanosine [(+-)-2-amino-1,9-dihydro-9-[(1 alpha,3 beta,4 alpha)-3-hydroxy-4-(hydroxymethyl)cyclopentyl]-6H-purine-6-one] (2'-CDG) is highly active in cell culture against strains S148 and E377 of herpes simplex virus type 1 (HSV-1), both of which code for thymidine kinase, and much less active against strain BW10168 which is deficient in this enzyme activity. Antiviral activity is associated primarily with the D-enantiomer; the L-enantiomer has much lower but significant activity. The metabolism of racemic 2'-CDG and its D- and L-enantiomers was studied in uninfected HEp-2 cells and in HEp-2 cells infected with the S148 or BW10168 strains of HSV-1. Nucleotides were separated by HPLC, and their elution was monitored by spectrophotometry. The chromatograms of extracts of cells infected with the S148 strain and treated with (+/-)-2'-CDG or D-2'-CDG included a new peak which appeared in the triphosphate region. This peak, the area of which exceeded that of the GTP peak, was shown to be due to the triphosphate of 2'-CDG. The new peak was not observed by HPLC of extracts of uninfected cells treated with (+/-)-2'-CDG or either of its enantiomers, cells infected with the S148 strain and treated with L-2'-CDG, or cells infected with the BW10168 strain and treated with (+/-)-2'-CDG or either of its enantiomers. The results were similar when these studies were performed with uninfected Vero cells and with Vero cells infected with strain S148 of HSV-1. In experiments with D-[8-3H]-2'-CDG, small amounts of phosphates of 2'-CDG could also be detected in uninfected HEp-2 cells and in cells infected with the BW10168 strain of HSV-1. Thus, 2'-CDG apparently is a good substrate for the virus-coded kinase and a very poor substrate for cellular phosphorylating enzymes. The selective phosphorylation of 2'-CDG by the virus-specific kinase presumably is critical for its antiviral activity as it is for that of acyclovir and other acyclic derivatives of guanine.


Asunto(s)
Antivirales/metabolismo , Desoxiguanosina/análogos & derivados , Simplexvirus/enzimología , Timidina Quinasa/metabolismo , Animales , Línea Celular , Cromatografía Líquida de Alta Presión , Desoxiguanosina/metabolismo , Herpes Simple/metabolismo , Mutación , Nucleótidos/aislamiento & purificación , Fosforilación , Simplexvirus/genética , Timidina Quinasa/genética
14.
Cancer Gene Ther ; 18(6): 390-8, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21394111

RESUMEN

The use of E. coli purine nucleoside phosphorylase (PNP) to activate prodrugs has demonstrated excellent activity in the treatment of various human tumor xenografts in mice. E. coli PNP cleaves purine nucleoside analogs to generate toxic adenine analogs, which are activated by adenine phosphoribosyl transferase (APRT) to metabolites that inhibit RNA and protein synthesis. We created tumor cell lines that encode both E. coli PNP and excess levels of human APRT, and have used these new cell models to test the hypothesis that treatment of otherwise refractory human tumors could be enhanced by overexpression of APRT. In vivo studies with 6-methylpurine-2'-deoxyriboside (MeP-dR), 2-F-2'-deoxyadenosine (F-dAdo) or 9-ß-D-arabinofuranosyl-2-fluoroadenine 5'-monophosphate (F-araAMP) indicated that increased APRT in human tumor cells coexpressing E. coli PNP did not enhance either the activation or the anti-tumor activity of any of the three prodrugs. Interestingly, expression of excess APRT in bystander cells improved the activity of MeP-dR, but diminished the activity of F-araAMP. In vitro studies indicated that increasing the expression of APRT in the cells did not significantly increase the activation of MeP. These results provide insight into the mechanism of bystander killing of the E. coli PNP strategy, and suggest ways to enhance the approach that are independent of APRT.


Asunto(s)
Adenina Fosforribosiltransferasa/metabolismo , Escherichia coli/enzimología , Profármacos/farmacología , Purina-Nucleósido Fosforilasa/metabolismo , Animales , Línea Celular Tumoral , Escherichia coli/metabolismo , Terapia Genética , Vectores Genéticos/genética , Humanos , Ratones , Profármacos/uso terapéutico , Nucleósidos de Purina/metabolismo , Trasplante Heterólogo , Fosfato de Vidarabina/análogos & derivados , Fosfato de Vidarabina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA