Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters










Publication year range
1.
Structure ; 5(10): 1373-83, 1997 Oct 15.
Article in English | MEDLINE | ID: mdl-9351810

ABSTRACT

BACKGROUND: Purine nucleoside phosphorylase (PNP) from Escherichia coli is a hexameric enzyme that catalyzes the reversible phosphorolysis of 6-amino and 6-oxopurine (2'-deoxy)ribonucleosides to the free base and (2'-deoxy)ribose-1-phosphate. In contrast, human and bovine PNPs are trimeric and accept only 6-oxopurine nucleosides as substrates. The difference in the specificities of these two enzymes has been utilized in gene therapy treatments in which certain prodrugs are cleaved by E. coli PNP but not the human enzyme. The trimeric and hexameric PNPs show no similarity in amino acid sequence, even though they catalyze the same basic chemical reaction. Structural comparison of the active sites of mammalian and E. coli PNPs would provide an improved basis for the design of potential prodrugs that are specific for E. coli PNP. RESULTS: The crystal structure of E. coli PNP at 2.0 A resolution shows that the overall subunit topology and active-site location within the subunit are similar to those of the subunits from human PNP and E. coli uridine phosphorylase. Nevertheless, even though the overall geometry of the E. coli PNP active site is similar to human PNP, the active-site residues and subunit interactions are strikingly different. In E. coli PNP, the purine- and ribose-binding sites are generally hydrophobic, although a histidine residue from an adjacent subunit probably forms a hydrogen bond with a hydroxyl group of the sugar. The phosphate-binding site probably consists of two main-chain nitrogen atoms and three arginine residues. In addition, the active site in hexameric PNP is much more accessible than in trimeric PNP. CONCLUSIONS: The structures of human and E. coli PNP define two possible classes of nucleoside phosphorylase, and help to explain the differences in specificity and efficiency between trimeric and hexameric PNPs. This structural data may be useful in designing prodrugs that can be activated by E. coli PNP but not the human enzyme.


Subject(s)
Escherichia coli/enzymology , Protein Conformation , Purine-Nucleoside Phosphorylase/chemistry , Binding Sites , Catalysis , Crystallography, X-Ray , Humans , Models, Molecular , Protein Folding , Protein Structure, Secondary , Purine-Nucleoside Phosphorylase/metabolism , Sequence Alignment , Substrate Specificity
2.
Antimicrob Agents Chemother ; 41(5): 1082-93, 1997 May.
Article in English | MEDLINE | ID: mdl-9145874

ABSTRACT

1592U89, (-)-(1S,4R)-4-[2-amino-6-(cyclopropylamino)-9H-purin-9-yl]-2-cyclo pentene-1-methanol, is a carbocyclic nucleoside with a unique biological profile giving potent, selective anti-human immunodeficiency virus (HIV) activity. 1592U89 was selected after evaluation of a wide variety of analogs containing a cyclopentene substitution for the 2'-deoxyriboside of natural deoxynucleosides, optimizing in vitro anti-HIV potency, oral bioavailability, and central nervous system (CNS) penetration. 1592U89 was equivalent in potency to 3'-azido-3'-deoxythymidine (AZT) in human peripheral blood lymphocyte (PBL) cultures against clinical isolates of HIV type 1 (HIV-1) from antiretroviral drug-naive patients (average 50% inhibitory concentration [IC50], 0.26 microM for 1592U89 and 0.23 microM for AZT). 1592U89 showed minimal cross-resistance (approximately twofold) with AZT and other approved HIV reverse transcriptase (RT) inhibitors. 1592U89 was synergistic in combination with AZT, the nonnucleoside RT inhibitor nevirapine, and the protease inhibitor 141W94 in MT4 cells against HIV-1 (IIIB). 1592U89 was anabolized intracellularly to its 5'-monophosphate in CD4+ CEM cells and in PBLs, but the di- and triphosphates of 1592U89 were not detected. The only triphosphate found in cells incubated with 1592U89 was that of the guanine analog (-)-carbovir (CBV). However, the in vivo pharmacokinetic, distribution, and toxicological profiles of 1592U89 were distinct from and improved over those of CBV, probably because CBV itself was not appreciably formed from 1592U89 in cells or animals (<2%). The 5'-triphosphate of CBV was a potent, selective inhibitor of HIV-1 RT, with Ki values for DNA polymerases (alpha, beta, gamma, and epsilon which were 90-, 2,900-, 1,200-, and 1,900-fold greater, respectively, than for RT (Ki, 21 nM). 1592U89 was relatively nontoxic to human bone marrow progenitors erythroid burst-forming unit and granulocyte-macrophage CFU (IC50s, 110 microM) and human leukemic and liver tumor cell lines. 1592U89 had excellent oral bioavailability (105% in the rat) and penetrated the CNS (rat brain and monkey cerebrospinal fluid) as well as AZT. Having demonstrated an excellent preclinical profile, 1592U89 has progressed to clinical evaluation in HIV-infected patients.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , Anti-HIV Agents/pharmacokinetics , Dideoxynucleosides/pharmacokinetics , Acquired Immunodeficiency Syndrome/metabolism , Adenosine Deaminase/metabolism , Administration, Oral , Animals , Anti-HIV Agents/blood , Anti-HIV Agents/chemistry , Anti-HIV Agents/urine , Antiviral Agents/blood , Antiviral Agents/pharmacokinetics , Area Under Curve , Biotransformation , Cells, Cultured , Dideoxynucleosides/blood , Dideoxynucleosides/chemistry , Dideoxynucleosides/urine , Drug Resistance, Microbial , Female , HIV-1/drug effects , Half-Life , Humans , Injections, Intravenous , Macaca fascicularis , Male , Rats , Structure-Activity Relationship
3.
Drug Metab Dispos ; 23(11): 1242-7, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8591725

ABSTRACT

Ganciclovir (GCV), which is used in the treatment of human cytomegalovirus infections, is poorly absorbed orally. A double prodrug of GCV, the dipivalate ester of 6-deoxy-GCV (6-dGCV) (called 6-dGCV-DPiv), was given orally to rats (25 mg/kg) and resulted in a nearly 7-fold enhancement of GCV bioavailability compared with administration of GCV alone and a 2-fold increase compared with administration of 6-dGCV. The prodrug was rapidly hydrolyzed and extensively oxidized by first-pass metabolism in such a way that only GCV, 6-dGCV, and a small amount of the monopivalate ester of 6-dGCV were observed in rat plasma. In cynomolgus monkey was given the prodrug orally (22.5 mg/kg), two additional metabolites were observed--the 8-hydroxy analogs of GCV and dGCV. The double prodrug approach demonstrated the potential for enhanced oral delivery of GCV in humans.


Subject(s)
Antiviral Agents/pharmacokinetics , Ganciclovir/analogs & derivatives , Ganciclovir/pharmacokinetics , Prodrugs/pharmacokinetics , Administration, Oral , Animals , Antiviral Agents/chemical synthesis , Chromatography, High Pressure Liquid , Feces/chemistry , Female , Ganciclovir/chemical synthesis , Half-Life , In Vitro Techniques , Injections, Intravenous , Macaca fascicularis , Male , Oxidation-Reduction , Prodrugs/chemical synthesis , Rats , Species Specificity
4.
Cancer Res ; 55(15): 3352-6, 1995 Aug 01.
Article in English | MEDLINE | ID: mdl-7614470

ABSTRACT

Earlier studies have shown guanine arabinoside (ara-G) is an effective agent against growth of T-cell lines and freshly isolated human T-leukemic cells. However, poor water solubility of ara-G limits clinical use. 2-Amino-6-methoxypurine arabinoside (506U) is a water-soluble prodrug converted to ara-G by adenosine deaminase. 506U is not a substrate for deoxycytidine kinase, adenosine kinase, or purine nucleoside phosphorylase and is phosphorylated by mitochondrial deoxyguanosine kinase at a rate 4% that of ara-G phosphorylation. Mitochondrial DNA polymerase was the least sensitive to ara-GTP inhibition of the five human DNA polymerases tested. [3H]506U was anabolized to ara-G 5'-phosphates in CEM cells but not to phosphorylated metabolites of 506U. 506U was selective for transformed T over B cells and also inhibited growth in two of three monocytic lines tested. 506U given i.v. to cynomolgus monkeys was rapidly converted to ara-G; the ara-G had a half-life of approximately 2 h. 506U had in vivo dose-dependent efficacy against human T-cell tumors in immunodeficient mice. A Phase 1 trial of 506U against refractory hematological malignancies is now in progress at two study sites.


Subject(s)
Antineoplastic Agents/therapeutic use , Arabinonucleosides/therapeutic use , Leukemia, T-Cell/drug therapy , Prodrugs/therapeutic use , Animals , Antineoplastic Agents/metabolism , Arabinonucleosides/metabolism , Arabinonucleotides/metabolism , Drug Screening Assays, Antitumor , Female , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/metabolism , Humans , Leukemia, B-Cell/drug therapy , Leukemia, T-Cell/metabolism , Macaca fascicularis/metabolism , Mice , Mice, Nude , Nucleic Acid Synthesis Inhibitors , Prodrugs/metabolism , Tumor Cells, Cultured
5.
Biochem Pharmacol ; 46(12): 2209-18, 1993 Dec 14.
Article in English | MEDLINE | ID: mdl-8274154

ABSTRACT

The varicella-zoster virus (VZV) thymidine kinase (TK) EC 2.7.2.21) catalyzes the phosphorylation of many anti-VZV nucleosides. Purified, bacterially expressed VZV TK was characterized with regard to N-terminal amino acid sequence, pI value, pH optimum, metal ion requirement, phosphate donor and acceptor specificity, and inhibition by dTTP. Initial velocities of thymidine phosphorylation with variable MgATP concentrations fit a two-site model with apparent Km values for MgATP of 0.10 and 900 microM. dTTP was a noncompetitive inhibitor of thymidine phosphorylation but was competitive with MgATP. Phosphate donor and acceptor specificities of the bacterially expressed enzyme were indistinguishable from those of VZV TK purified from infected cells. Detailed studies of the nucleoside specificity with the bacterially expressed enzyme showed that, for a given sugar moiety, thymine nucleosides were the most efficient substrates followed by nucleosides of cytosine, uracil, adenine, and with some exceptions, guanine. For a given pyrimidine or purine (except guanine), 2'-deoxyribonucleosides were the most efficient substrates, followed by arabinosides, ribonucleosides, 2',3'-dideoxyribonucleosides, and the acyclic moiety of acyclovir.


Subject(s)
Herpesvirus 3, Human/enzymology , Thymidine Kinase/metabolism , Adenosine Triphosphate/pharmacology , Amino Acid Sequence , Carbohydrate Metabolism , Carbohydrates/chemistry , Escherichia coli/genetics , Hydrogen-Ion Concentration , Isoelectric Point , Metals , Organophosphorus Compounds/metabolism , Phosphorylation , Pyrimidine Nucleosides/chemistry , Pyrimidine Nucleosides/metabolism , Substrate Specificity , Thymidine Kinase/antagonists & inhibitors , Thymidine Kinase/genetics
6.
J Med Chem ; 36(3): 378-84, 1993 Feb 05.
Article in English | MEDLINE | ID: mdl-8426365

ABSTRACT

Twenty-one 6-alkoxypurine 2',3'-dideoxynucleosides were enzymatically synthesized with nucleoside phosphorylases purified from E. coli. Eighteen analogs exhibited anti-HIV-1 activity in MT4 cells. Two analogs, 6-(hexyloxy)-(17) and 6-(heptyloxy)-(18) purine 2',3'-dideoxynucleoside, were as potent as 2',3'-dideoxyinosine (ddI, didanosine, Videx). Although the antiviral activities of 17 and 18 were equivalent, 18 was more cytotoxic. Analogs containing less than four carbons in the 6-alkoxypurine substituent exhibited weak anti-HIV-1 activity. Analogs containing more than seven carbons in the 6-alkoxypurine substituent were too cytotoxic to be effectively evaluated for antiviral activity. Several 6-alkoxypurine 2',3'-dideoxynucleosides were evaluated for substrate activity with calf intestinal adenosine deaminase (ADA). Increasing the carbon chain length of the 6-alkoxypurine substituent decreased the rate of dealkoxylation. The best substrate in this series was 6-methoxypurine 2',3'-dideoxynucleaside (1); however, the rate of dealkoxylation of 100 microM 1 was 0.17% of the rate of deamination of 100 microM 2',3'-dideoxyadenosine. Compound 17, the most potent anti-HIV-1 analog, was not a substrate for ADA. EHNA (erthro-9-(2-hydroxy-3-nonyl)adenine), a potent inhibitor of ADA, had little effect on the antiviral activities of 17 and ddI. In contrast, coformycin, a potent inhibitor of both ADA and AMP deaminase, dramatically decreased the antiviral activity of 17, but not the antiviral activity of ddI. Thus, AMP deaminase appeared to be involved in the anabolism of 17. The pharmacokinetic profile of 17, the most promising analog in this series, was determined in the rat. At least seventeen metabolites of 17, including ddI, were detected in plasma samples. This analog also had poor oral bioavailability.


Subject(s)
Dideoxynucleosides/pharmacology , HIV-1/drug effects , Purine Nucleosides/pharmacology , Adenosine Deaminase/metabolism , Animals , Chemical Phenomena , Chemistry, Physical , Cytopathogenic Effect, Viral/drug effects , Dideoxynucleosides/chemistry , Dideoxynucleosides/metabolism , Humans , In Vitro Techniques , Male , Mice , Purine Nucleosides/chemistry , Purine Nucleosides/metabolism , Purine-Nucleoside Phosphorylase/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Thymidine Phosphorylase/metabolism
7.
Antiviral Res ; 20(1): 13-20, 1993 Jan.
Article in English | MEDLINE | ID: mdl-8457145

ABSTRACT

6-Dimethylamino-9-(beta-D-arabinofuranosyl)-9H-purine (ara-DMAP) effectively prevented the development of rash and appreciably reduced viremia in simian varicella virus-infected monkeys. Doses of 100 and 50 mg/kg/day, administered orally, were highly effective. The lowest dose of 20 mg/kg/day was much less effective in preventing moderate viremia. However, the 20 mg/kg/day did prevent the development of rash in two of three monkeys. All three doses of ara-DMAP reduced liver infection as reflected by lower aspartate aminotransferase values in the sera of the African green monkeys. Orally administered ara-DMAP was rapidly absorbed. However, significant variation among individual monkeys in the AUC values, peak plasma levels, and plasma half-lives were observed.


Subject(s)
Antiviral Agents/pharmacokinetics , Chickenpox/drug therapy , Vidarabine/analogs & derivatives , Administration, Oral , Animals , Animals, Wild , Antiviral Agents/blood , Antiviral Agents/therapeutic use , Aspartate Aminotransferases/blood , Chlorocebus aethiops , Drug Evaluation , Half-Life , Relative Biological Effectiveness , Skin/pathology , Treatment Outcome , Vidarabine/administration & dosage , Vidarabine/pharmacokinetics , Vidarabine/therapeutic use , Viremia/drug therapy
8.
J Med Virol ; Suppl 1: 139-45, 1993.
Article in English | MEDLINE | ID: mdl-8245881

ABSTRACT

Research leading to the new anti-herpesvirus compounds discussed here has come from three approaches. The first approach was directed towards improving the bioavailability of acyclovir by examining the potential of a variety of prodrugs, leading to the new compound valaciclovir hydrochloride. The second approach was to examine a large number of 5-substituted pyrimidines for activity against those viruses which were not as potently inhibited by acyclovir as are herpes simplex viruses, i.e., varicella zoster virus (VZV) and human cytomegalovirus (HCMV). This research led to the new chemical entity 882C for VZV. A third approach has been to examine drug combinations with acyclovir. This research led to the compound 348U, an inhibitor of herpes simplex virus ribonucleotide reductase which acts synergistically in combination with acyclovir. This manuscript will focus on the first two approaches leading to new compounds valaciclovir hydrochloride and 882C since Dr. Safrin details such background for 348U/acyclovir. Attempts to improve the bioavailability of acyclovir began a decade ago. Early prodrugs were compounds with alterations in the 6-substituent of the purine ring of acyclovir. The 6-amino congener required the cellular enzyme adenosine deaminase for conversion to acyclovir and the 6-deoxycongener was dependent on cellular xanthine oxidase for conversion. Neither of these prodrugs had a chronic toxicity profile in laboratory animals as good as acyclovir. Efforts were directed towards simpler esters and 18 amino acid esters were made. The pharmacokinetic profile of each prodrug was determined in rats by measuring the recovery of acyclovir in urine after oral dosing.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Acyclovir/analogs & derivatives , Antiviral Agents/chemistry , Arabinofuranosyluracil/analogs & derivatives , Drug Design , Herpesvirus 3, Human/drug effects , Valine/analogs & derivatives , Acyclovir/chemistry , Acyclovir/pharmacology , Antiviral Agents/pharmacology , Arabinofuranosyluracil/chemistry , Arabinofuranosyluracil/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacology , Valacyclovir , Valine/chemistry , Valine/pharmacology
9.
J Med Chem ; 36(1): 119-25, 1993 Jan 08.
Article in English | MEDLINE | ID: mdl-8421277

ABSTRACT

Twenty purine 2'-deoxy-2'-fluororibosides were synthesized by enzymic pentosyl transfer from 2'-deoxy-2'-fluorouridine. Each nucleoside analogue was assayed for cytotoxicity in uninfected Madin-Darby canine kidney cells and for their ability to suppress influenza A virus infections in these cells. The most potent antivirial activity was observed with analogues having an amino group in the 2-position of the purine moiety. All 2-unsubstituted analogues were less potent than their 2-amino counterparts. Furthermore, 2-methyl,2-methoxy, or 2-fluoro substitution obliterated antivirial activity. The most cytotoxic member of the series was the 2-fluoro-6-amino analogue (IC50 = 120 microM). 2'-Deoxy-2'-fluoroguanosine and those congeners readily converted to it by adenosine deaminase showed the most potent antivirial activity (IC50 = 15-23 microM). Little cytotoxicity was observed with this subgroup of analogues which renders them worthy of further investigation as potential antiinfluenza agents.


Subject(s)
Antiviral Agents/chemical synthesis , Orthomyxoviridae/drug effects , Purines/chemical synthesis , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Dogs , Hydrocarbons, Fluorinated/chemistry , Kidney/drug effects , Purines/pharmacology , Structure-Activity Relationship
10.
Arch Biochem Biophys ; 296(1): 161-9, 1992 Jul.
Article in English | MEDLINE | ID: mdl-1605627

ABSTRACT

A novel nucleoside phosphotransferase, referred to as adenosine phosphotransferase (Ado Ptase), was partially purified 1230-fold from human placenta. This enzyme differed from other known nucleoside phosphotransferases in its substrate specificity. Using AMP as the phosphate donor, it readily phosphorylated Ado. Changes in the sugar moiety were tolerated. dAdo and ddAdo were phosphate acceptors and dAMP was a donor. No other nucleotide or nucleoside common in nature displayed appreciable activity as donor or acceptor substrate, respectively. In the absence of nucleoside, the enzyme catalyzed the hydrolysis of AMP, typical of other nucleoside phosphotransferases. However, in the presence of Ado, little, if any, hydrolysis occurred. Ado Ptase had an absolute requirement for a metal cation, with Mg2+ and, to a lesser extent, Mn2+ fulfilling this requisite. The apparent Km for Ado was 0.2 mM. However, the donor AMP displayed cooperativity in both transfer and hydrolytic reactions. This cooperativity was eliminated by nucleotides, 2,3-diphosphoglycerate, and inorganic phosphate. ADP and 2,3-diphosphoglycerate were especially potent. In the presence of these effectors, the apparent Km for AMP was 3.0 mM in the transfer reaction and 4.0 mM in the hydrolytic reaction. Kinetic data suggest that there are two nucleotide binding sites on Ado Ptase, one for the donor, the other for an effector. AMP appeared to bind to both sites. Although this novel enzyme might play a role in the anabolism of nucleoside analogues, the normal physiological role of this nucleoside phosphotransferase is not understood.


Subject(s)
Adenosine Kinase/metabolism , Placenta/enzymology , Adenosine Kinase/isolation & purification , Chromatography, Affinity , Chromatography, Gel , Chromatography, Ion Exchange , Female , Humans , Kinetics , Nucleosides/metabolism , Nucleotides/pharmacology , Pregnancy , Substrate Specificity
11.
J Med Chem ; 35(1): 56-63, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1732533

ABSTRACT

6-Methoxypurine arabinoside (9-beta-D-arabinofuranosyl-6-methoxy-9H-purine, 1) has potent and selective activity against varicella-zoster virus in vitro. An unfavourable metabolic profile observed with oral dosing in the rat led to the preparation of a variety of 2',3',5'-triesters (2a-n) and several 2',3'-, 2',5'-, and 3',5'-diesters of this arabinoside (3a-n, 4a-f, and 5a-j, respectively). The compounds were evaluated as prodrugs by measuring the urinary levels of 1 in rat urine after oral dosing. With the exception of triacetate 2a, the triesters failed to significantly enhance bioavailability. Administration of compound 2a resulted in a 3-fold increase in systemic availability of 1, possibly because of its increased water solubility (1.6 times more soluble than 1) and only slightly increased relative log P value (1.93 vs 0.50 for 1). The longer chain aliphatic triesters and aromatic triesters had lower water solubilities and increased lipophilic partitioning. These factors might account for the lower systemic bioavailability of these compounds. In contrast, the diesters, especially the aliphatic diesters, showed significantly improved systemic availability. This might be a consequence of the higher aqueous solubilities and enhanced partition coefficients seen with these compounds. 2',3'-Diacetate 3a showed the best combination of high systemic availability and water solubility of all the prodrugs of 1.


Subject(s)
Antiviral Agents/chemical synthesis , Arabinonucleosides/chemistry , Arabinonucleosides/chemical synthesis , Prodrugs/chemical synthesis , Administration, Oral , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Arabinonucleosides/pharmacokinetics , Biological Availability , Esters/chemical synthesis , Esters/chemistry , Esters/pharmacology , Male , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Rats , Structure-Activity Relationship
12.
Antimicrob Agents Chemother ; 35(10): 2116-20, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1722079

ABSTRACT

6-Methoxypurine arabinoside (ara-M) is a highly selective inhibitor of varicella-zoster virus (VZV). It belongs to a class of purine arabinosides whose anti-VZV activity in vitro correlates with substrate utilization by the VZV-encoded thymidine kinase (TK) (D. R. Averett, G. W. Koszalka, J. A. Fyfe, G. B. Roberts, D. J. M. Purifoy, and T. A. Krenitsky, Antimicrob Agents Chemother. 35:851-857, 1991). In this study, the mechanism of action of ara-M was explored. VZV-infected human fibroblasts selectively accumulated ara-M and its phosphorylated metabolites, whereas in uninfected fibroblasts or in those infected with a TK-deficient strain of VZV, there was virtually no cellular uptake of ara-M. The major intracellular metabolite of ara-M in VZV-infected cells was identified as the triphosphate of adenine arabinoside (ara-ATP). Appreciable levels of ara-ADP, ara-AMP, and ara-MMP were also detected. However, di- or triphosphorylated forms of ara-M were not detected. Moreover, in VZV-infected cells, the concentrations of ara-ATP which accumulated in the presence of ara-M were up to eightfold higher than those generated with ara-A itself. In contrast, in uninfected cells, the levels of ara-ATP which accumulated in the presence of ara-M were barely detectable. Clearly, Ara-M activation was dependent on the activity of the virus-encoded TK, while ara-A anabolism resulted primarily from the activity of host cell enzymes. Therefore, ara-M selectively generates the DNA polymerase inhibitor ara-ATP in the VZV-infected cell.


Subject(s)
Arabinonucleosides/metabolism , Herpesvirus 3, Human , Cells, Cultured , Chromatography, High Pressure Liquid , Fibroblasts/metabolism , Half-Life , Humans , Vidarabine/metabolism , Vidarabine Phosphate/analogs & derivatives , Vidarabine Phosphate/metabolism
13.
Antimicrob Agents Chemother ; 35(10): 2121-4, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1662024

ABSTRACT

6-Methoxypurine arabinoside (ara-M) exhibits potent activity against varicella-zoster virus (VZV) as a result of ara-M's anabolism to the triphosphate of adenine arabinoside (ara-ATP) in VZV-infected cells. The adenosine deaminase inhibitor erythro-9-(2-hydroxy-3-nonyl)adenine (EHNA) enhanced the formation of ara-ATP by inhibiting ara-M demethoxylation. In contrast, deoxycoformycin and coformycin, inhibitors of both adenosine deaminase and AMP deaminase, blocked the formation of ara-ATP and reversed the anti-VZV activity of ara-M. These results indicate that after the initial phosphorylation of ara-M by the VZV-coded thymidine kinase, the monophosphate is demethoxylated by AMP deaminase to form ara-IMP, which is converted to ara-ATP by the sequential actions of the cellular adenylosuccinate synthetase, adenylosuccinate lyase, and nucleotide kinases.


Subject(s)
Arabinonucleosides/metabolism , Herpesvirus 3, Human , 5'-Nucleotidase/metabolism , AMP Deaminase/metabolism , Adenine/analogs & derivatives , Adenine/metabolism , Cells, Cultured , Fibroblasts/metabolism , Humans
14.
Biochem J ; 278 ( Pt 2): 487-91, 1991 Sep 01.
Article in English | MEDLINE | ID: mdl-1910332

ABSTRACT

1. [2'-2H]Inosine was made from inosine by tetraisopropyldisiloxanyl protection of the 3'- and 5'-positions, oxidation with dimethyl sulphoxide and acetic anhydride, immediate NaB2H4 reduction of the oxo sugar product and inversion at C-2' of the resultant protected [2'-2H]arabino-inosine by trifluoromethanesulphonylation and reaction with caesium propionate, followed by deprotection. 2. The equilibrium-perturbation technique was used to measure beta 2H(V/K) for phosphorolysis of this compound by the purine nucleoside phosphorylase of Escherichia coli as a function of pH. 3. The pH variation indicates an intrinsic effect of 1.068 masked by isotopically silent steps near the pH optimum. 4. The similar pH variation of these beta-deuterium effects and the alpha-deuterium effects measured previously [Stein & Cordes (1981) J. Biol. Chem. 256, 767-772; Lehikoinen, Sinnott & Krenitsky (1989) Biochem. J. 257, 355-359] for this reaction provides the first experimental reassurance for the common assumption that pH changes merely mask and unmask the chemical steps in an enzyme-catalysed reaction, and do not detectably alter transition-state structure. 5. The dihedral angle between the C-H-2' bond and the electron-deficient p-orbital at the transition state is in the range 32-48 degrees, in accord with an essentially planar furanose ring.


Subject(s)
Deuterium/chemistry , Purine-Nucleoside Phosphorylase/chemistry , Escherichia coli/enzymology , Hydrogen-Ion Concentration , Inosine/chemistry , Isotopes , Kinetics , Spectrophotometry, Ultraviolet
15.
Proc Natl Acad Sci U S A ; 88(18): 8039-43, 1991 Sep 15.
Article in English | MEDLINE | ID: mdl-1654555

ABSTRACT

An approach involving retroviral-mediated gene therapy for the treatment of neoplastic disease is described. This therapeutic approach is called "virus-directed enzyme/prodrug therapy" (VDEPT). The VDEPT approach exploits the transcriptional differences between normal and neoplastic cells to achieve selective killing of neoplastic cells. We now describe development of the VDEPT approach for the treatment of hepatocellular carcinoma. Replication-defective, amphotrophic retroviruses were constructed containing a chimeric varicella-zoster virus thymidine kinase (VZV TK) gene that is transcriptionally regulated by either the hepatoma-associated alpha-fetoprotein or liver-associated albumin transcriptional regulatory sequences. Subsequent to retroviral infection, expression of VZV TK was limited to either alpha-fetoprotein- or albumin-positive cells, respectively. VZV TK metabolically activated the nontoxic prodrug 6-methoxypurine arabinonucleoside (araM), ultimately leading to the formation of the cytotoxic anabolite adenine arabinonucleoside triphosphate (araATP). Cells that selectively expressed VZV TK became selectively sensitive to araM due to the VZV TK-dependent anabolism of araM to araATP. Hence, these retroviral-delivered chimeric genes generated tissue-specific expression of VZV TK, tissue-specific anabolism of araM to araATP, and tissue-specific cytotoxicity due to araM exposure. By utilizing such retroviral vectors, araM was anabolized to araATP in hepatoma cells, producing a selective cytotoxic effect.


Subject(s)
Arabinonucleosides/administration & dosage , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Prodrugs/administration & dosage , Animals , Base Sequence , Cloning, Molecular , Gene Expression , Genetic Therapy , Genetic Vectors , Growth Inhibitors/administration & dosage , Herpesvirus 3, Human/genetics , Humans , In Vitro Techniques , Molecular Sequence Data , Oligonucleotides/chemistry , Rats , Thymidine Kinase/administration & dosage , Thymidine Kinase/genetics , Tumor Cells, Cultured
16.
Antimicrob Agents Chemother ; 35(7): 1437-43, 1991 Jul.
Article in English | MEDLINE | ID: mdl-1656862

ABSTRACT

A series of 6-alkylaminopurine arabinosides were synthesized and found to inhibit varicella-zoster virus (VZV). The antiviral activities of these nucleosides were limited to VZV. None of the other viruses tested in the herpesvirus family were affected. The in vitro antiviral potencies of the 18 arabinosides correlated with their efficiencies as substrates of the VZV-encoded thymidine kinase in all but one case. The arabinosides of 6-methylaminopurine and 6-dimethylaminopurine were the most potent analogs, with 50% inhibitory concentrations against VZV of 3 and 1 microM, respectively. They were not cytotoxic to uninfected MRC-5 cells, human Detroit 98 cells, or mouse L cells (50% inhibitory concentration, greater than 100 microM). Neither 6-methylaminopurine arabinoside nor 6-dimethylaminopurine arabinoside was detectably phosphorylated by either adenosine kinase or 2'-deoxycytidine kinase. These two alkylaminopurine arabinosides were also resistant to deamination catalyzed by adenosine deaminase. The VZV-dependent phosphorylation of these nucleosides offers the possibility of a potent and highly selective therapy for VZV infection.


Subject(s)
Antiviral Agents/chemical synthesis , Herpesvirus 3, Human/drug effects , Vidarabine/analogs & derivatives , Adenosine Deaminase Inhibitors , Animals , Antiviral Agents/pharmacology , Chemical Phenomena , Chemistry, Physical , L Cells/drug effects , Magnetic Resonance Spectroscopy , Mice , Phosphotransferases/antagonists & inhibitors , Vidarabine/chemical synthesis , Vidarabine/pharmacology
17.
Antimicrob Agents Chemother ; 35(6): 1165-73, 1991 Jun.
Article in English | MEDLINE | ID: mdl-1929259

ABSTRACT

The metabolism and pharmacokinetics of 6-methoxypurine arabinoside (ara-M), a potent and selective inhibitor of varicella-zoster virus, were investigated in rats and monkeys. In Long Evans rats, orally administered [8-14C]ara-M (10 mg/kg) was well absorbed but extensively metabolized to hypoxanthine arabinoside (ara-H), hypoxanthine, xanthine, uric acid, and allantoin. Only 4% of an oral dose was recovered in the urine as unchanged drug, compared with 40% of an intravenous dose, indicating significant presystemic metabolism. Pretreatment of rats with 1-aminobenzotriazole, an inhibitor of cytochrome P-450, did not alter this metabolism. Pretreatment with deoxycoformycin or erythro-9-(2-hydroxy-3-nonyl)adenine hydrochloride, inhibitors of adenosine deaminase, resulted in a marked decrease in ara-M metabolism, indicating that adenosine deaminase plays a major role in the biotransformation of ara-M. In cynomolgus monkeys, [8-14C]ara-M (10 mg/kg) administered intravenously or orally was extensively metabolized to ara-H. Several minor urinary metabolites were detected in both rats and monkeys. However, adenine arabinoside was not found in urine or plasma from either rats or monkeys after administration of ara-M, except for a very low level detected in the urine of rats pretreated with deoxycoformycin. The elimination half-lives of intravenously administered ara-M in rats and monkeys were 29 and 45 min, respectively. The corresponding half-lives of the primary metabolite, ara-H, were 44 min and 2.3 h. Plasma profiles of orally administered ara-M in both rats and monkeys demonstrated the poor oral bioavailability of this arabinoside. The results of these studies indicate that ara-M is not well suited for oral administration because of extensive presystemic metabolism.


Subject(s)
Arabinonucleosides/pharmacokinetics , Administration, Oral , Animals , Arabinonucleosides/metabolism , Biotransformation , Chromatography, High Pressure Liquid , Drug Evaluation, Preclinical , In Vitro Techniques , Injections, Intravenous , Macaca fascicularis , Microsomes, Liver , Protein Binding , Rats , Species Specificity
18.
Antimicrob Agents Chemother ; 35(5): 851-7, 1991 May.
Article in English | MEDLINE | ID: mdl-1649571

ABSTRACT

Seven 6-alkoxypurine arabinosides were synthesized and evaluated for in vitro activity against varicella-zoster virus (VZV). The simplest of the series, 6-methoxypurine arabinoside (ara-M), was the most potent, with 50% inhibitory concentrations ranging from 0.5 to 3 microM against eight strains of VZV. This activity was selective. The ability of ara-M to inhibit the growth of a variety of human cell lines was at least 30-fold less (50% effective concentration, greater than 100 microM) than its ability to inhibit the virus. Enzyme studies suggested the molecular basis for these results. Of the seven 6-alkoxypurine arabinosides, ara-M was the most efficient substrate for VZV-encoded thymidine kinase as well as the most potent antiviral agent. In contrast, it was not detectably phosphorylated by any of the three major mammalian nucleoside kinases. Upon direct comparison, ara-M was appreciably more potent against VZV than either acyclovir or adenine arabinoside (ara-A). However, in the presence of an adenosine deaminase inhibitor, the arabinosides of adenine and 6-methoxypurine were equipotent but not equally selective; the adenine congener had a much less favorable in vitro chemotherapeutic index. Again, this result correlated with data from enzyme studies in that ara-A, unlike ara-M, was a substrate for two mammalian nucleoside kinases. Unlike acyclovir and ara-A, ara-M had no appreciable activity against other viruses of the herpes group. The potency and selectivity of ara-M as an anti-VZV agent in vitro justify its further study.


Subject(s)
Antiviral Agents/pharmacology , Arabinonucleosides/pharmacology , Herpesvirus 3, Human/drug effects , Acyclovir/pharmacology , Arabinonucleosides/chemistry , Cell Survival/drug effects , Cells, Cultured , Herpesvirus 3, Human/enzymology , Humans , Kinetics , Thymidine Kinase/metabolism , Vidarabine/pharmacology
19.
J Biol Chem ; 265(23): 14016-22, 1990 Aug 15.
Article in English | MEDLINE | ID: mdl-2199449

ABSTRACT

The three-dimensional structure of thymidine phosphorylase from Escherichia coli has been determined at 2.8 A resolution using multiple-isomorphous-replacement techniques. The amino acid sequence deduced from the deoA DNA sequence is also reported. Thymidine phosphorylase exists in the crystal as an S-shaped dimer in which the subunits are related by a crystallographic 2-fold axis. Each subunit is composed of a small alpha-helical domain of six helices and a large alpha/beta domain. The alpha/beta domain includes a six-stranded mixed beta-sheet and a four-stranded antiparallel beta-sheet. The active site has been identified by difference Fourier analyses of the binding of thymine and thymidine and lies in a cavity between the small and large domains. The central beta-sheet is splayed open to accommodate a putative phosphate-binding site which is probably occupied by a sulfate ion in the crystal.


Subject(s)
Escherichia coli/enzymology , Pentosyltransferases , Thymidine Phosphorylase , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Crystallization , DNA, Bacterial/genetics , Ligands , Models, Molecular , Molecular Sequence Data , Pentosyltransferases/genetics , Pentosyltransferases/isolation & purification , Protein Conformation , Thymidine Phosphorylase/genetics , Thymidine Phosphorylase/isolation & purification , X-Ray Diffraction
20.
J Biol Chem ; 265(6): 3066-9, 1990 Feb 25.
Article in English | MEDLINE | ID: mdl-2105941

ABSTRACT

The diphosphate of the antiherpetic agent acyclovir [9-[(2-hydroxyethoxy)methyl]guanine] has been shown to inhibit purine nucleoside phosphorylase with unique potency (Tuttle, J. V., and Krenitsky, T. A. (1984) J. Biol. Chem. 259, 4065-4069). A major factor contributing to the superior inhibition by this diphosphate over the corresponding mono- and triphosphates is revealed here. Homologues of acyclovir mono- and diphosphate that extend the ethoxy moiety by one to four methylene groups were synthesized. These homologues were evaluated for their ability to inhibit human purine nucleoside phosphorylase. Within the diphosphate series, the Ki values increased progressively with increasing chain length. With the monophosphates, the Ki values reached a minimum with the homologue containing a pentoxy moiety. A plot of chain length versus Ki values for both mono- and diphosphates showed that both series had similar optimal distances between the aminal carbon and the terminal oxygen anion. Monophosphates with optimal positioning were somewhat less potent than diphosphates with similar positioning. Nevertheless, it was clear that a major factor in determining potency of inhibition was the distance of the terminal phosphate from the guanine moiety.


Subject(s)
Acyclovir/analogs & derivatives , Acyclovir/pharmacology , Antiviral Agents , Nucleotides/pharmacology , Pentosyltransferases/antagonists & inhibitors , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Erythrocytes/enzymology , Humans , Kinetics , Nucleotides/chemical synthesis , Purine-Nucleoside Phosphorylase/blood , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...