Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Chemother Pharmacol ; 85(3): 573-583, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31915968

RESUMO

Treatment with fludarabine phosphate (9-ß-D-arabinofuranosyl-2-F-adenine 5'-phosphate, F-araAMP) leads to regressions and cures of human tumor xenografts that express Escherichia coli purine nucleoside phosphorylase (EcPNP). This occurs despite the fact that fludarabine (F-araA) is a relatively poor substrate for EcPNP, and is cleaved to liberate 2-fluoroadenine at a rate only 0.3% that of the natural E. coli PNP substrate, adenosine. In this study, we investigated a panel of naturally occurring PNPs to identify more efficient enzymes that may be suitable for metabolizing F-araA as part of experimental cancer therapy. We show that Trichomonas vaginalis PNP (TvPNP) cleaves F-araA with a catalytic efficiency 25-fold greater than the prototypic E. coli enzyme. Cellular extracts from human glioma cells (D54) transduced with lentivirus stably expressing TvPNP (D54/TvPNP) were found to cleave F-araA at a rate similar to extracts from D54 cells expressing EcPNP, although much less enzyme was expressed per cell in the TvPNP transduced condition. As a test of safety and efficacy using TvPNP, human head and neck squamous cell carcinoma (FaDu) xenografts expressing TvPNP were studied in nude mice and shown to exhibit robust tumor regressions, albeit with partial weight loss that resolved post-therapy. F-araAMP was also a very effective treatment for mice bearing D54/TvPNP xenografts in which approximately 10% of tumor cells expressed the enzyme, indicating pronounced ability to kill non-transduced tumor cells (high bystander activity). Moreover, F-araAMP demonstrated activity against D54 tumors injected with an E1, E3 deleted adenoviral vector encoding TvPNP. In that setting, despite higher F-araA cleavage activity using TvPNP, tumor responses were similar to those obtained with EcPNP, indicating factors other than F-Ade production may limit regressions of the D54 murine xenograft model. Our results establish that TvPNP is a favorable enzyme for activating F-araA, and support further studies in combination with F-araAMP for difficult-to-treat human cancers.


Assuntos
Glioma/tratamento farmacológico , Purina-Núcleosídeo Fosforilase/genética , Trichomonas vaginalis/enzimologia , Vidarabina/análogos & derivados , Animais , Linhagem Celular Tumoral , Escherichia coli/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Glioma/genética , Humanos , Lentivirus/genética , Camundongos , Camundongos Nus , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Vidarabina/farmacologia
2.
Eur J Med Chem ; 108: 616-622, 2016 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-26724729

RESUMO

Impressive antitumor activity has been observed with fludarabine phosphate against tumors that express Escherichia coli purine nucleoside phosphorylase (PNP) due to the liberation of 2-fluoroadenine in the tumor tissue. 6-Methylpurine (MeP) is another cytotoxic adenine analog that does not exhibit selectivity when administered systemically, and could be very useful in a gene therapy approach to cancer treatment involving E. coli PNP. The prototype MeP releasing prodrug 9-(2-deoxy-ß-d-ribofuranosyl)-6-methylpurine (1) [MeP-dR] has demonstrated good activity against tumors expressing E. coli PNP, but its antitumor activity is limited due to toxicity resulting from the generation of MeP from gut bacteria. Therefore, we have embarked on a medicinal chemistry program to identify a combination of non-toxic MeP prodrugs and non-human adenosine glycosidic bond cleaving enzymes. The two best MeP-based substrates with M64V-E coli PNP, a mutant which was engineered to tolerate modification at the 5'-position of adenosine and its analogs, were 9-(6-deoxy-α-l-talofuranosyl)-6-methylpurine (3) [methyl(talo)-MeP-R] and 9-(α-l-lyxofuranosyl)6-methylpurine (4) [lyxo-MeP-R]. The detailed synthesis methyl(talo)-MeP-R and lyxo-MeP-R, and the evaluation of their substrate activity with 4 enzymes not normally associated with cancer patients is described. In addition, we have determined the intraperitoneal pharmacokinetic (ip-PK) properties of methyl(talo)-MeP-R and have determined its in vivo bystander activity in mice bearing D54 tumors that express M64V PNP. The observed good in vivo bystander activity of [methyl(talo)-MeP-R/M64V-E coli PNP combination suggests that these agents could be useful for the treatment of cancer.


Assuntos
Antineoplásicos/farmacologia , Carboidratos/farmacologia , Inibidores Enzimáticos/farmacologia , Escherichia coli/enzimologia , Nucleosídeos/farmacologia , Purina-Núcleosídeo Fosforilase/antagonistas & inibidores , Purinas/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Carboidratos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Camundongos , Estrutura Molecular , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/patologia , Nucleosídeos/química , Purina-Núcleosídeo Fosforilase/metabolismo , Purinas/síntese química , Purinas/química , Relação Estrutura-Atividade
3.
Bioorg Chem ; 65: 9-16, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26745284

RESUMO

6-Methylpurine (MeP) is cytotoxic adenine analog that does not exhibit selectivity when administered systemically, and could be very useful in a gene therapy approach to cancer treatment involving Escherichia coli PNP. The prototype MeP releasing prodrug, 9-(ß-d-ribofuranosyl)-6-methylpurine, MeP-dR has demonstrated good activity against tumors expressing E. coli PNP, but its antitumor activity is limited due to toxicity resulting from the generation of MeP from gut bacteria. Therefore, we have embarked on a medicinal chemistry program to identify non-toxic MeP prodrugs that could be used in conjunction with E. coli PNP. In this work, we report on the synthesis of 9-(6-deoxy-ß-d-allofuranosyl)-6-methylpurine (3) and 9-(6-deoxy-5-C-methyl-ß-d-ribo-hexofuranosyl)-6-methylpurine (4), and the evaluation of their substrate activity with several phosphorylases. The glycosyl donors; 1,2-di-O-acetyl-3,5-di-O-benzyl-α-d-allofuranose (10) and 1-O-acetyl-3-O-benzyl-2,5-di-O-benzoyl-6-deoxy-5-C-methyl-ß-d-ribohexofuran-ose (15) were prepared from 1,2:5,6-di-O-isopropylidine-α-d-glucofuranose in 9 and 11 steps, respectively. Coupling of 10 and 15 with silylated 6-methylpurine under Vorbrüggen glycosylation conditions followed conventional deprotection of the hydroxyl groups furnished 5'-C-methylated-6-methylpurine nucleosides 3 and 4, respectively. Unlike 9-(6-deoxy-α-l-talo-furanosyl)-6-methylpurine, which showed good substrate activity with E. coli PNP mutant (M64V), the ß-d-allo-furanosyl derivative 3 and the 5'-di-C-methyl derivative 4 were poor substrates for all tested glycosidic bond cleavage enzymes.


Assuntos
Carboidratos/química , Nucleosídeos/síntese química , Nucleosídeos/farmacologia , Purina-Núcleosídeo Fosforilase/metabolismo , Purinas/química , Humanos , Conformação Molecular , Nucleosídeos/química , Purina-Núcleosídeo Fosforilase/química , Especificidade por Substrato
4.
Biochem Biophys Res Commun ; 461(1): 65-9, 2015 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-25849889

RESUMO

In Vibrio cholerae, the genes required for biofilm development are repressed by quorum sensing at high cell density due to the accumulation in the medium of two signaling molecules, cholera autoinducer 1 (CAI-1) and autoinducer 2 (AI-2). A significant fraction of toxigenic V. cholerae isolates, however, exhibit dysfunctional quorum sensing pathways. It was reported that transition state analogs of the enzyme methylthioadenosine/S-adenosylhomocysteine nucleosidase (MtnN) required to make AI-2 inhibited biofilm formation in the prototype quorum sensing-deficient strain N16961. This finding prompted us to examine the role of both autoinducers and MtnN in biofilm development and virulence gene expression in a quorum sensing-deficient genetic background. Here we show that deletion of mtnN encoding methylthioadenosine/S-adenosylhomocysteine nucleosidase, cqsA (CAI-1), and/or luxS (AI-2) do not prevent biofilm development. However, two independent mtnN mutants exhibited diminished growth rate and motility in swarm agar plates suggesting that, under certain conditions, MtnN could influence biofilm formation indirectly. Nevertheless, MtnN is not required for the development of a mature biofilm.


Assuntos
Proteínas de Bactérias/metabolismo , Biofilmes/crescimento & desenvolvimento , Liases de Carbono-Enxofre/metabolismo , Cetonas/metabolismo , N-Glicosil Hidrolases/metabolismo , Purina-Núcleosídeo Fosforilase/metabolismo , Percepção de Quorum/fisiologia , Vibrio cholerae/fisiologia , Movimento Celular/fisiologia
5.
Cancer Chemother Pharmacol ; 74(2): 291-302, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24908436

RESUMO

PURPOSE: Currently approved DNA hypomethylating nucleosides elicit their effects in part by depleting DNA methyltransferase I (DNMT1). However, their low response rates and adverse effects continue to drive the discovery of newer DNMT1 depleting agents. Herein, we identified two novel 2'-deoxycytidine (dCyd) analogs, 4'-thio-2'-deoxycytidine (T-dCyd) and 5-aza-4'-thio-2'-deoxycytidine (aza-T-dCyd) that potently deplete DNMT1 in both in vitro and in vivo models of cancer and concomitantly inhibit tumor growth. METHODS: DNMT1 protein levels in in vitro and in vivo cancer models were determined by Western blotting and antitumor efficacy was evaluated using xenografts. Effects on CpG methylation were evaluated using methylation-specific PCR. T-dCyd metabolism was evaluated using radiolabeled substrate. RESULTS: T-dCyd markedly depleted DNMT1 in CCRF-CEM and KG1a leukemia and NCI-H23 lung carcinoma cell lines, while it was ineffective in the HCT-116 colon or IGROV-1 ovarian tumor lines. On the other hand, aza-T-dCyd potently depleted DNMT1 in all of these lines indicating that dCyd analogs with minor structural dissimilarities induce different DNMT1 turnover mechanisms. Although T-dCyd was deaminated to 4'-thio-2'-deoxyuridine, very little was converted to 4'-thio-thymidine nucleotides, suggesting that inhibition of thymidylate synthase would be minimal with 4'-thio dCyd analogs. Both T-dCyd and aza-T-dCyd also depleted DNMT1 in human tumor xenografts and markedly reduced in vivo tumor growth. Interestingly, the selectivity index of aza-T-dCyd was at least tenfold greater than that of decitabine. CONCLUSIONS: Collectively, these data show that 4'-thio modified dCyd analogs, such as T-dCyd or aza-T-dCyd, could be a new source of clinically effective DNMT1 depleting anticancer compounds with less toxicity.


Assuntos
Azacitidina/análogos & derivados , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/efeitos dos fármacos , Desoxicitidina/análogos & derivados , Neoplasias Experimentais/metabolismo , Tionucleosídeos/farmacologia , Animais , Azacitidina/farmacologia , Western Blotting , Proliferação de Células/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferase 1 , DNA de Neoplasias/genética , Desoxicitidina/farmacologia , Feminino , Humanos , Camundongos , Camundongos Nus , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/patologia , Reação em Cadeia da Polimerase em Tempo Real , Células Tumorais Cultivadas
6.
Cancer Chemother Pharmacol ; 70(2): 321-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22760227

RESUMO

PURPOSE: Systemically administered fludarabine phosphate (F-araAMP) slows growth of human tumor xenografts that express Escherichia coli purine nucleoside phosphorylase (PNP). However, this treatment has been limited by the amount of F-araAMP that can be administered in vivo. The current study was designed to (1) determine whether efficacy of this overall strategy could be improved by intratumoral administration of F-araAMP, (2) test enhancement of the approach with external beam radiation, and (3) optimize recombinant adenovirus as a means to augment PNP delivery and bystander killing in vivo. METHODS: The effects of systemic or intratumoral F-araAMP in mice were investigated with human tumor xenografts (300 mg), in which 10 % of the cells expressed E. coli PNP from a lentiviral promoter. Tumors injected with an adenoviral vector expressing E. coli PNP (Ad/PNP; 2 × 10(11) viral particles, 2 times per day × 3 days) and the impact of radiotherapy on tumors treated by this approach were also studied. Radiolabeled F-araAMP was used to monitor prodrug activation in vivo. RESULTS: Intratumoral administration of F-araAMP in human tumor xenografts expressing E. coli PNP resulted in complete regressions and/or prolonged tumor inhibition. External beam radiation significantly augmented this effect. Injection of large human tumor xenografts (human glioma, nonsmall cell lung cancer, or malignant prostate tumors) with Ad/PNP followed by intratumoral F-araAMP resulted in excellent antitumor activity superior to that observed following systemic administration of prodrug. CONCLUSION: Activation of F-araAMP by E. coli PNP results in destruction of large tumor xenografts in vivo, augments radiotherapy, and promotes robust bystander killing. Our results indicate that intratumoral injection of F-araAMP leads to ablation of tumors in vivo with minimal toxicity.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Terapia Genética , Pró-Fármacos/uso terapêutico , Purina-Núcleosídeo Fosforilase/genética , Fosfato de Vidarabina/análogos & derivados , Adenoviridae/genética , Animais , Antimetabólitos Antineoplásicos/administração & dosagem , Antimetabólitos Antineoplásicos/farmacocinética , Efeito Espectador/efeitos dos fármacos , Efeito Espectador/genética , Efeito Espectador/efeitos da radiação , Linhagem Celular Tumoral , Terapia Combinada , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/genética , Resistencia a Medicamentos Antineoplásicos/efeitos da radiação , Escherichia coli/genética , Vetores Genéticos , Glioma/tratamento farmacológico , Glioma/genética , Glioma/radioterapia , Humanos , Injeções Intralesionais , Camundongos , Camundongos Nus , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacocinética , Purina-Núcleosídeo Fosforilase/metabolismo , Transfecção , Transplante Heterólogo , Fosfato de Vidarabina/administração & dosagem , Fosfato de Vidarabina/farmacocinética , Fosfato de Vidarabina/uso terapêutico
7.
Eur J Med Chem ; 47(1): 167-74, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22112758

RESUMO

A series of C-6 alkyl, cycloalkyl, and aryl-9-(ß-d-ribofuranosyl)purines were synthesized and their substrate activities with Escherichia coli purine nucleoside phosphorylase (E. coli PNP) were evaluated. (Ph(3)P)(4)Pd-mediated cross-coupling reactions of 6-chloro-9-(2,3,5-tri-O-acetyl-ß-d-ribofuranosyl)-purine (6) with primary alkyl (Me, Et, n-Pr, n-Bu, isoBu) zinc halides followed by treatment with NH(3)/MeOH gave the corresponding 6-alkyl-9-(ß-d-ribofuranosyl)purine derivatives 7-11, respectively, in good yields. Reactions of 6 with cycloalkyl(propyl, butyl, pentyl)zinc halides and aryl (phenyl, 2-thienyl)zinc halides gave under similar conditions the corresponding 6-cyclopropyl, cyclobutyl, cyclopentyl, phenyl, and thienyl -9-(ß-d-ribofuranosyl)purine derivatives 12-16, respectively in high yields. E. coli PNP showed a high tolerance to the steric and hydrophobic environment at the 6-position of the synthesized purine ribonucleosides. Significant cytotoxic activity was observed for 8, 12, 15, and 16. Evaluation of 12 and 16 against human tumor xenografts in mice did not demonstrate any selective antitumor activity. In addition, 6-methyl-9-(ß-d-arabinofuranosyl)purine (18) was prepared and evaluated.


Assuntos
Escherichia coli/enzimologia , Halogenação , Paládio/química , Nucleosídeos de Purina/química , Nucleosídeos de Purina/metabolismo , Purina-Núcleosídeo Fosforilase/metabolismo , Ribonucleosídeos/química , Ribonucleosídeos/metabolismo , Zinco/química , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Catálise , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Camundongos , Nucleosídeos de Purina/síntese química , Nucleosídeos de Purina/farmacologia , Ribonucleosídeos/síntese química , Ribonucleosídeos/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Biochemistry ; 50(30): 6549-58, 2011 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-21707079

RESUMO

Uridine phosphorylase (UP), a key enzyme in the pyrimidine salvage pathway, catalyzes the reversible phosphorolysis of uridine or 2'-deoxyuridine to uracil and ribose 1-phosphate or 2'-deoxyribose 1-phosphate. This enzyme belongs to the nucleoside phosphorylase I superfamily whose members show diverse specificity for nucleoside substrates. Phylogenetic analysis shows Streptococcus pyogenes uridine phosphorylase (SpUP) is found in a distinct branch of the pyrimidine subfamily of nucleoside phosphorylases. To further characterize SpUP, we determined the crystal structure in complex with the products, ribose 1-phosphate and uracil, at 1.8 Å resolution. Like Escherichia coli UP (EcUP), the biological unit of SpUP is a hexamer with an α/ß monomeric fold. A novel feature of the active site is the presence of His169, which structurally aligns with Arg168 of the EcUP structure. A second active site residue, Lys162, is not present in previously determined UP structures and interacts with O2 of uracil. Biochemical studies of wild-type SpUP showed that its substrate specificity is similar to that of EcUP, while EcUP is ∼7-fold more efficient than SpUP. Biochemical studies of SpUP mutants showed that mutations of His169 reduced activity, while mutation of Lys162 abolished all activity, suggesting that the negative charge in the transition state resides mostly on uracil O2. This is in contrast to EcUP for which transition state stabilization occurs mostly at O4.


Assuntos
Proteínas de Bactérias/química , Família Multigênica , Streptococcus pyogenes/enzimologia , Uridina Fosforilase/química , Proteínas de Bactérias/genética , Catálise , Domínio Catalítico/genética , Cristalografia por Raios X , Mutagênese Sítio-Dirigida , Dobramento de Proteína , Ribosemonofosfatos/química , Eletricidade Estática , Especificidade por Substrato/genética , Uracila/química , Uridina Fosforilase/genética
9.
Acta Crystallogr D Biol Crystallogr ; 66(Pt 2): 155-62, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20124695

RESUMO

Purine nucleoside phosphorylase (PNP) catalyzes the reversible phosphorolysis of purine ribonucleosides to the corresponding free bases and ribose 1-phosphate. The crystal structure of grouper iridovirus PNP (givPNP), corresponding to the first PNP gene to be found in a virus, was determined at 2.4 A resolution. The crystals belonged to space group R3, with unit-cell parameters a = 193.0, c = 105.6 A, and contained four protomers per asymmetric unit. The overall structure of givPNP shows high similarity to mammalian PNPs, having an alpha/beta structure with a nine-stranded mixed beta-barrel flanked by a total of nine alpha-helices. The predicted phosphate-binding and ribose-binding sites are occupied by a phosphate ion and a Tris molecule, respectively. The geometrical arrangement and hydrogen-bonding patterns of the phosphate-binding site are similar to those found in the human and bovine PNP structures. The enzymatic activity assay of givPNP on various substrates revealed that givPNP can only accept 6-oxopurine nucleosides as substrates, which is also suggested by its amino-acid composition and active-site architecture. All these results suggest that givPNP is a homologue of mammalian PNPs in terms of amino-acid sequence, molecular mass, substrate specificity and overall structure, as well as in the composition of the active site.


Assuntos
Purina-Núcleosídeo Fosforilase/química , Ranavirus/enzimologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Sequência Conservada , Cristalografia por Raios X , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Fosfatos/química , Fosfatos/metabolismo , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Purina-Núcleosídeo Fosforilase/genética , Purina-Núcleosídeo Fosforilase/metabolismo , Ranavirus/genética , Alinhamento de Sequência , Especificidade por Substrato
10.
Nucleosides Nucleotides Nucleic Acids ; 28(5): 642-56, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-20183607

RESUMO

Metalation of 6-methyl-9-(tetrahydro-2H-pyran-2-yl)purine (10) with lithiating agents of varying basicities such as n-BuLi and LiHMDS in THF at -78 degrees C resulted in metalation at both of the 6-CH(3) moiety and the 8-CH position, irrespective of the molar equivalence of the base. On the other hand, a regioselective metalation at the 6-CH(3) moiety of 10 was observed with NaHMDS or KHMDS, under similar conditions. Treatment of the potassium salts of 10 and of the protected riboside derivative 6-methyl-9-(beta-D-2,3,5-tri-O-tert-butyldimethylsilylribofuranosyl)purine (22) with N-fluorobenzenesulfonamide (NFSI) at -78 degrees C gave the corresponding 6-fluoromethylpurine derivatives 11 and 23, respectively, in good yields. Deprotection of 11 and 23 under standard conditions gave 6-fluoromethylpurine (6-FMeP, 3) and 6-fluoromethyl-9-(beta-D-ribofuranosyl)purine (6-FMePR, 4), respectively, in high yield. Both 3 and 4 demonstrated cytotoxic activity against CCRF-CEM cells in culture. 6-FMePR is a good substrate for E. coli purine nucleoside phosphorylase (E. coli PNP) with a comparable substrate activity to that of the parent nucleoside, 6-methyl-9-(beta-D-ribofuranosyl)purine (6-MePR, 21). The cytotoxic activity of 6-FMeP along with the substrate activity of 6-FMePR with E. coli PNP meet the fundamental requirements for using 6-FMeP as a potential toxin in PNP/prodrug based cancer gene therapy.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Halogenação , Neoplasias/tratamento farmacológico , Purinas/química , Purinas/farmacologia , Antineoplásicos/síntese química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Escherichia coli/enzimologia , Terapia Genética , Humanos , Purina-Núcleosídeo Fosforilase/metabolismo , Purinas/síntese química
11.
J Antimicrob Chemother ; 59(1): 118-21, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17085766

RESUMO

OBJECTIVES: Analyse a series of halogenated 3-deaza-adenosine analogues for efficacy against Mycobacterium tuberculosis H37Ra and determine if adenosine (Ado) kinase plays a role in the mechanism of action of these compounds. METHODS: The MIC as determined by microdilution broth assay provided a measure of antitubercular efficacy. MIC values were measured in M. tuberculosis strains H37Ra, SRICK1 (an Ado kinase-deficient strain of M. tuberculosis derived from H37Ra) and SRICK1 complemented with adoK, the gene which codes for Ado kinase in M. tuberculosis, in order to determine if Ado kinase played a role in the mechanism of action of these compounds. Furthermore, each compound was analysed as both a substrate and inhibitor for purified Ado kinases from M. tuberculosis and human sources. RESULTS: 2-Fluoro-3-deaza-adenosine, 3-fluoro-3-deaza-adenosine and 2,3-difluoro-3-deaza-adenosine exhibited antitubercular activity that was Ado kinase-dependent. Furthermore, these compounds were at least 10-fold better substrates for M. tuberculosis Ado kinase than the human homologue. CONCLUSIONS: The Ado kinase-dependent antitubercular activity exhibited by several of the halogenated 3-deaza-adenosine analogues investigated in this study warrants further investigation of these compounds as antitubercular agents. Furthermore, substrate and inhibition studies provided insight into the Ado-binding domain of Ado kinase, indicating that steric hindrance may limit the size of exocyclic modifications at the 3-position of Ado.


Assuntos
Adenosina Quinase/fisiologia , Antituberculosos/farmacologia , Mycobacterium tuberculosis/efeitos dos fármacos , Tubercidina/farmacologia , Humanos , Ligantes , Relação Estrutura-Atividade , Especificidade por Substrato
12.
Artigo em Inglês | MEDLINE | ID: mdl-16247956

RESUMO

Our studies have led to the identification of an E. coli PNP mutant (M64V) that is able to cleave numerous 5'-modified nucleoside analogs with much greater efficiency than the wild-type enzyme. The biological activity of the three best substrates of this mutant (9-[6-deoxy-alpha-L-talofuranosyl]-6-methylpurine (methyl(talo)-MeP-R), 9-[6-deoxy-alpha-L-talofuranosyl]-2-F-adenine, and 9-[alpha-L-lyxofuranosyl]-2-F-adenine) were evaluated so that we can optimally utilize these compounds. Our results indicated that the mechanism of toxicity of methyl(talo)-MeP-R to mice was due to its cleavage to MeP by a bacterial enzyme, and that the toxicity of the two F-Ade analogs was due to their cleavage to F-Ade by mammalian methylthioadenosine phosphorylase.


Assuntos
Escherichia coli/enzimologia , Mutação , Nucleosídeos/síntese química , Nucleosídeos/farmacologia , Pró-Fármacos/farmacologia , Purina-Núcleosídeo Fosforilase/genética , Purina-Núcleosídeo Fosforilase/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Química Farmacêutica/métodos , Desenho de Fármacos , Escherichia coli/genética , Humanos , Concentração Inibidora 50 , Camundongos , Modelos Químicos , Nucleosídeos/química , Pró-Fármacos/síntese química , Especificidade por Substrato , Tionucleosídeos/química
13.
Hum Gene Ther ; 16(3): 339-47, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15812229

RESUMO

The E. coli PNP suicide gene sensitizes solid tumors to nucleoside prodrugs, such as 6-methylpurine-2'-deoxyriboside (MeP-dR). In this study using lentiviral, MuLv, and HSV-based gene transfer, we quantified thresholds for inhibition of tumor growth and bystander killing by E. coli PNP and tested the role of intestinal flora in this process. Regressions of human glioma tumors following retroviral transduction exhibited dose dependence on both the level of PNP expression and the dose of MeP-dR administered, including strong tumor inhibition when 90-99% bystander cells comprised the tumor mass. A replication competent, non-neurovirulent herpes simplex virus (HSV) deficient in both copies of the gamma-1 34.5 gene was next engineered to express E. coli PNP under the egr-1 promoter (HSV-PNP). HSV-PNP injected intratumorally (17 million pfu/0.05 ml) in nude mice bearing 300 mg human glioma flank tumors produced a delay in tumor growth (approximately 24 days delay to one doubling). MeP-dR treatment after antibiotic therapy (to eliminate enteric flora encoding PNP enzymes) resulted in antitumor enhancement, with arrest of tumor growth (delay to doubling >50 days). Bystander killing of the magnitude described here has been difficult to accomplish with other suicide genes, such as HSV-tk or cytosine deaminase. The results establish a model for applying E. coli PNP to HSV treatment of glioma.


Assuntos
Genes Transgênicos Suicidas/genética , Terapia Genética/métodos , Glioma/terapia , Nucleosídeos de Purina/uso terapêutico , Purina-Núcleosídeo Fosforilase/metabolismo , Purina-Núcleosídeo Fosforilase/uso terapêutico , Animais , Antibacterianos/uso terapêutico , Linhagem Celular Tumoral , Escherichia coli/genética , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Vetores Genéticos/uso terapêutico , Vida Livre de Germes , Glioma/genética , Lentivirus/genética , Camundongos , Camundongos Nus , Pró-Fármacos/metabolismo , Pró-Fármacos/uso terapêutico , Nucleosídeos de Purina/metabolismo , Purina-Núcleosídeo Fosforilase/genética , Simplexvirus/genética , Simplexvirus/metabolismo , Fatores de Tempo
14.
Cancer Res ; 64(18): 6610-5, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15374975

RESUMO

Escherichia coli purine nucleoside phosphorylase (PNP) expressed in tumors converts relatively nontoxic prodrugs into membrane-permeant cytotoxic compounds with high bystander activity. In the present study, we examined tumor regressions resulting from treatment with E. coli PNP and fludarabine phosphate (F-araAMP), a clinically approved compound used in the treatment of hematologic malignancies. We tested bystander killing with an adenoviral construct expressing E. coli PNP and then more formally examined thresholds for the bystander effect, using both MuLv and lentiviral vectoring. Because of the importance of understanding the mechanism of bystander action and the limits to this anticancer strategy, we also evaluated in vivo variables related to the expression of E. coli PNP (level of E. coli PNP activity in tumors, ectopic expression in liver, percentage of tumor cells transduced in situ, and accumulation of active metabolites in tumors). Our results indicate that F-araAMP confers excellent in vivo dose-dependent inhibition of bystander tumor cells, including strong responses in subcutaneous human glioma xenografts when 95 to 97.5% of the tumor mass is composed of bystander cells. These findings define levels of E. coli PNP expression necessary for antitumor activity with F-araAMP and demonstrate new potential for a clinically approved compound in solid tumor therapy.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Escherichia coli/genética , Terapia Genética/métodos , Purina-Núcleosídeo Fosforilase/genética , Fosfato de Vidarabina/análogos & derivados , Fosfato de Vidarabina/farmacologia , Adenoviridae/genética , Animais , Antimetabólitos Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Terapia Combinada , Relação Dose-Resposta a Droga , Escherichia coli/enzimologia , Vetores Genéticos/genética , Glioma/tratamento farmacológico , Glioma/enzimologia , Glioma/genética , Humanos , Lentivirus/genética , Camundongos , Camundongos Nus , Vírus da Leucemia Murina de Moloney/genética , Purina-Núcleosídeo Fosforilase/biossíntese , Purina-Núcleosídeo Fosforilase/metabolismo , Transfecção/métodos , Fosfato de Vidarabina/farmacocinética , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Tuberculosis (Edinb) ; 84(5): 327-36, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15207808

RESUMO

2-Methyladenosine (methyl-Ado) has selective activity against Mycobacterium tuberculosis (M. tuberculosis). In an effort to better understand its mechanism of action, we have characterized its metabolism in M. tuberculosis cells. The primary intracellular metabolite of methyl-Ado was 2-methyl-adenylate (methyl-AMP). Very little of the methyl-AMP was metabolized further. A M. tuberculosis strain that was resistant to methyl-Ado did not express adenosine kinase and did not convert methyl-Ado to methyl-AMP in intact cells. In contrast to these results, the primary intracellular metabolite of adenosine in M. tuberculosis cells was ATP, which was readily incorporated into RNA. The rate of metabolism of methyl-Ado to methyl-AMP was similar to the rate of metabolism of adenosine to ATP. Treatment of M. tuberculosis with methyl-Ado did not affect intracellular ATP levels. Methyl-Ado and Ado were also cleaved to 2-methyladenine and adenine, respectively, which accumulated in the medium outside the cells. These studies suggested that methyl-AMP was the active metabolite responsible for the cytotoxicity of this agent. Furthermore, because methyl-Ado was poorly metabolized in human cells, these studies indicated that the selective activity of methyl-Ado was due to its selective activation by M. tuberculosis. These studies have identified two enzyme reactions (Ado kinase and Ado cleavage) in M. tuberculosis that could be exploited for the rational design of new and selective anti-M. tuberculosis agents.


Assuntos
Adenosina/análogos & derivados , Adenosina/metabolismo , Mycobacterium tuberculosis/metabolismo , Trifosfato de Adenosina/metabolismo , Técnicas Bacteriológicas
16.
Artigo em Inglês | MEDLINE | ID: mdl-14565269

RESUMO

A selective metalation at the 6-CH3 over C-8 of 6-methylpurine derivative 6 was observed with softer counter cation (Na+ or K+) of the base, while the harder Li+ showed no selectivity. In the presence of N-fluorobenzenesulfonamide (NFSI), this property was utilized for the synthesis of 6-fluoromethylpurine derivatives 4 and 5 as potential toxins for suicide gene therapy.


Assuntos
Metais , Nucleosídeos de Purina/química , Nucleosídeos de Purina/síntese química , Purinas/química , Terapia Genética/métodos , Indicadores e Reagentes , Metilação , Purinas/síntese química
17.
J Biol Chem ; 278(47): 47110-8, 2003 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-12937174

RESUMO

Purine nucleoside phosphorylase catalyzes reversible phosphorolysis of purine nucleosides and 2'-deoxypurine nucleosides to the free base and ribose (or 2'-deoxyribose) 1-phosphate. Whereas the human enzyme is specific for 6-oxopurine ribonucleosides, the Escherichia coli enzyme accepts additional substrates including 6-oxopurine ribonucleosides, 6-aminopurine ribonucleosides, and to a lesser extent purine arabinosides. These differences have been exploited in a potential suicide gene therapy treatment for solid tumors. In an effort to optimize this suicide gene therapy approach, we have determined the three-dimensional structure of the E. coli enzyme in complex with 10 nucleoside analogs and correlated the structures with kinetic measurements and computer modeling. These studies explain the preference of the enzyme for ribose sugars, show increased flexibility for active site residues Asp204 and Arg24, and suggest that interactions involving the 1- and 6-positions of the purine and the 4'- and 5'-positions of the ribose provide the best opportunities to increase prodrug specificity and enzyme efficiency.


Assuntos
Proteínas de Escherichia coli/química , Purina-Núcleosídeo Fosforilase/química , Proteínas de Escherichia coli/metabolismo , Cinética , Ligantes , Modelos Moleculares , Estrutura Molecular , Nucleosídeos de Purina/química , Nucleosídeos de Purina/metabolismo , Purina-Núcleosídeo Fosforilase/metabolismo , Ribose , Especificidade por Substrato
18.
J Pharmacol Exp Ther ; 304(3): 1280-4, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12604707

RESUMO

We have demonstrated antitumor activity against refractory human glioma and pancreatic tumors with 6-methylpurine (MeP) using either a suicide gene therapy strategy to selectively release 6-methylpurine in tumor cells or direct intratumoral injection of 6-methylpurine itself. A single i.p. injection in mice of the prodrug 9-beta-D-[2-deoxyribofuranosyl]-6-methylpurine (MeP-dR; 134 mg/kg) caused sustained regression lasting over 70 days of D54 (human glioma) tumors transduced with the Escherichia coli purine nucleoside phosphorylase (PNP), and a single intratumoral injection of 6-methylpurine (5-10 mg/kg) elicited prolonged delays of the growth of D54 tumors and CFPAC human pancreatic carcinoma. Because the D54 tumor doubling time is >15 days, the experiments indicate that prodrug activation by E. coli PNP engenders destruction of both dividing and nondividing tumor compartments in vivo and, therefore, address a fundamental barrier that has limited the development of suicide gene strategies in the past. A prolonged retention time of 6-methylpurine metabolites in tumors was noted in vivo (T(1/2) >24 h compared with a serum half-life of <1 h). By high-pressure liquid chromatography, metabolites of [(3)H]MeP-dR were 5- to 6-fold higher in tumors expressing E. coli PNP. These experiments point to new endpoints for monitoring E. coli PNP suicide gene therapy, including intratumoral enzymatic activity, in situ (intratumoral) prodrug conversion, and tumor regressions after direct injection of a suicide gene toxin. The findings also help explain the strong in vivo bystander killing mechanism ascribed by several laboratories to E. coli PNP in the past.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Experimentais/tratamento farmacológico , Purinas/uso terapêutico , Animais , Modelos Animais de Doenças , Glioma/patologia , Humanos , Camundongos , Camundongos SCID , Transplante de Neoplasias , Resultado do Tratamento , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Chem Biol ; 10(12): 1173-81, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14700625

RESUMO

Activation of prodrugs by Escherichia coli purine nucleoside phosphorylase (PNP) provides a method for selectively killing tumor cells expressing a transfected PNP gene. This gene therapy approach requires matching a prodrug and a known enzymatic activity present only in tumor cells. The specificity of the method relies on avoiding prodrug cleavage by enzymes already present in the host cells or the intestinal flora. Using crystallographic and computer modeling methods as guides, we have redesigned E. coli PNP to cleave new prodrug substrates more efficiently than does the wild-type enzyme. In particular, the M64V PNP mutant cleaves 9-(6-deoxy-alpha-L-talofuranosyl)-6-methylpurine with a kcat/Km over 100 times greater than for native E. coli PNP. In a xenograft tumor experiment, this compound caused regression of tumors expressing the M64V PNP gene.


Assuntos
Escherichia coli/enzimologia , Terapia Genética/métodos , Pró-Fármacos/metabolismo , Pró-Fármacos/uso terapêutico , Engenharia de Proteínas , Purina-Núcleosídeo Fosforilase/química , Purina-Núcleosídeo Fosforilase/metabolismo , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Simulação por Computador , Desenho Assistido por Computador , Cristalografia por Raios X , Escherichia coli/genética , Feminino , Humanos , Cinética , Camundongos , Modelos Moleculares , Estrutura Molecular , Mutação/genética , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Especificidade de Órgãos , Pró-Fármacos/efeitos adversos , Conformação Proteica , Purina-Núcleosídeo Fosforilase/genética , Especificidade por Substrato
20.
Cancer Gene Ther ; 10(1): 23-9, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12489025

RESUMO

The selective expression of Escherichia coli purine nucleoside phosphorylase (PNP) in solid tumors has been successfully used to activate two purine nucleoside analogs [9-(2-deoxy-beta-D-ribofuranosyl)-6-methylpurine (MeP-dR) and 9-beta-D-arabinofuranosyl-2-fluoroadenine (F-araA)] resulting in lasting tumor regressions and cures. E. coli PNP also cleaves 2-fluoro-2'-deoxyadenosine (F-dAdo) to 2-F-adenine, which is the toxic purine analog liberated from F-araA that has high bystander activity and is active against nonproliferating tumor cells. As F-dAdo is 3000 times better than F-araA as a substrate for E. coli PNP, we have evaluated its antitumor activity against D54 gliomas that express E. coli PNP and have characterized its in vivo metabolism in order to better understand its mechanism of action with respect to the other two agents. Like MeP-dR and F-araA-5'-monophosphate (F-araAMP, a prodrug of F-araA), treatment of mice bearing D54 tumors that express E. coli PNP with F-dAdo resulted in excellent antitumor activity. Although F-dAdo was as active as MeP-dR and better than F-araAMP, it was not dramatically better than either compound because of its short plasma half-life and the limited activation of F-adenine to toxic metabolites. Regardless, these results indicated that F-dAdo was also an excellent prodrug for use with gene vectors that deliver E. coli PNP to tumor cells.


Assuntos
Antineoplásicos/uso terapêutico , Desoxiadenosinas/farmacologia , Escherichia coli/enzimologia , Glioma/tratamento farmacológico , Glioma/enzimologia , Purina-Núcleosídeo Fosforilase/metabolismo , Animais , Regulação Bacteriana da Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/genética , Camundongos , Camundongos Nus , Transplante de Neoplasias , Pró-Fármacos/farmacologia , Purina-Núcleosídeo Fosforilase/genética , Retroviridae/genética , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...