Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Biochemistry ; 40(32): 9532-8, 2001 Aug 14.
Article in English | MEDLINE | ID: mdl-11583152

ABSTRACT

This study examines the ability of P450cam to catalyze the formation of 2-ethylhexanoic acid from 2-ethylhexanol relative to its activity on the natural substrate camphor. As is the case for camphor, the P450cam exhibits stereoselectivity for binding (R)- and (S)-2-ethylhexanol. Kinetic studies indicate (R)-2-ethylhexanoic acid is produced 3.5 times as fast as the (S)-enantiomer. In a racemic mixture of 2-ethylhexanol, P450cam produces 50% more (R)-2-ethylhexanoic acid than (S)-2-ethylhexanoic acid. The reason for stereoselective 2-ethylhexanoic acid production is seen in regioselectivity assays, where (R)-2-ethylhexanoic acid comprises 50% of total products while (S)-2-ethylhexanoic acid comprises only 13%. (R)- and (S)-2-ethylhexanol exhibit similar characteristics with respect to the amount of oxygen and reducing equivalents consumed, however, with (S)-2-ethylhexanol turnover producing more water than the (R)-enantiomer. Crystallographic studies of P450cam with (R)- or (S)-2-ethylhexanoic acid suggest that the (R)-enantiomer binds in a more ordered state. These results indicate that wild-type P450cam displays stereoselectivity toward 2-ethylhexanoic acid synthesis, providing a platform for rational active site design.


Subject(s)
Camphor 5-Monooxygenase/metabolism , Caproates/metabolism , Computer Simulation , Protein Structure, Tertiary , Camphor/metabolism , Camphor 5-Monooxygenase/isolation & purification , Crystallography, X-Ray , Kinetics , Models, Molecular , Molecular Structure , Pseudomonas putida/enzymology
4.
J Med Chem ; 43(7): 1264-70, 2000 Apr 06.
Article in English | MEDLINE | ID: mdl-10753464

ABSTRACT

The syntheses of furanthiofurin [5beta-D-(4'-thioribofuranosyl)furan-3-carboxamide, 1] and thiophenthiofurin [5beta-D-(4'-thioribofuranosyl)thiophene-3-carboxamide, 2], two C-thioribonucleoside analogues of tiazofurin, are described. Direct trifluoroacetic acid-catalyzed C-glycosylation of ethyl furan-3-carboxylate with 1-O-acetyl-2,3,5-tri-O-benzyl-4-thio-D-ribofuranose gave 2- and 5-glycosylated regioisomers, as a mixture of alpha and beta anomers. Ethyl 5-(2,3,5-tri-O-benzyl)-beta-D-(4'-thioribofuranosyl)furan-3-carboxylate (6beta) was debenzylated and then converted into the corresponding amide (furanthiofurin) by reaction with ammonium hydroxide. A similar C-glycosylation of ethyl thiophene-3-carboxylate with 1,2,3,5-tetra-O-acetyl-4-thio-D-ribofuranose catalyzed by stannic chloride afforded an anomeric mixture of 2- and 5-glycosylated regioisomers. Deacetylation of ethyl 5-(2,3,5-tri-O-acetyl)-beta-D-(4'-thioribofuranosyl)thiophene-3-carboxylate (13beta) with methanolic ammonia and treatment of the ethyl ester with ammonium hydroxide gave thiophenthiofurin. The glycosylation site and anomeric configuration were established by (1)H NMR spectroscopy. Thiophenthiofurin was found to be cytotoxic in vitro toward human myelogenous leukemia K562, albeit 39-fold less than thiophenfurin, while furanthiofurin proved to be inactive. K562 cells incubated with thiophenthiofurin resulted in inhibition of inosine 5'-monophosphate dehydrogenase (IMPDH) and an increase in IMP pools with a concurrent decrease in GTP levels. From computational studies it was deduced that, among the C-nucleoside analogues of tiazofurin, activity requires an electrophilic sulfur adjacent to the C-glycosidic bond and an energetically favorable conformer around chi = 0 degrees. Among these, the more constrained (least flexible) compounds (tiazofurin and thiophenfurin) are more active than the less constrained thiophenthiofurin. Those compounds which contain a nucleophilic oxygen in place of the thiazole or thiophene (oxazofurin, furanfurin, and furanthiofurin) show the least activity.


Subject(s)
Antineoplastic Agents/chemical synthesis , Furans/chemical synthesis , Ribavirin/chemical synthesis , Ribose/chemical synthesis , Thiophenes/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Furans/chemistry , Furans/pharmacology , Glycosylation , Humans , IMP Dehydrogenase/antagonists & inhibitors , Magnetic Resonance Spectroscopy , Molecular Conformation , Ribavirin/analogs & derivatives , Ribavirin/chemistry , Ribavirin/pharmacology , Ribonucleosides/chemistry , Ribose/analogs & derivatives , Ribose/chemistry , Ribose/pharmacology , Structure-Activity Relationship , Thermodynamics , Thiophenes/chemistry , Thiophenes/pharmacology , Tumor Cells, Cultured
5.
Curr Med Chem ; 6(7): 519-36, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10390598

ABSTRACT

Inosine monophosphate dehydrogenase (IMPDH, E.C. 1.1.1.205) is recognized as an important target for both antileukemic and immunosuppressive therapy. IMPDH catalyzes the NAD-dependent oxidation of inosine 5 monophosphate (IMP) to xanthosine 5 monophosphate. Several classes of IMPDH inhibitors are now in use or under development. These include agents that bind at either the substrate site (e.g. ribavirin and mizoribine) or at the NAD site (mycophenolic acid and thiazole-4-carboxamide adenine dinucleotide). All suffer from some degree of toxicity and/or susceptibility to metabolic inactivation. The finding that IMPDH exists as two isoforms, one of which (type II) is induced in tumor cells, has led to the search for potentially more effective isoform-specific agents. Recently, a number of crystal structures of IMPDH have become available. These include structures of the human type II, hamster, Tritrichomonas foetus, Streptococcus pyogenes and Borrelia burgdorferi enzymes. Each structure crystallizes as a tetramer of a/b barrels, with the active site located partly at the monomer-monomer interface. The substrate and cofactor bind in a continuous cleft on the C-terminal face of each barrel. The IMP base is well positioned to stack against the NAD nicotinamide ring to facilitate hydride transfer. The active site cleft is further bounded by a highly flexible flap and loop. These structures reveal enzyme-ligand interactions which suggest strategies for the design of improved inhibitors.


Subject(s)
IMP Dehydrogenase/antagonists & inhibitors , IMP Dehydrogenase/chemistry , Amino Acids/chemistry , Animals , Binding Sites , Borrelia burgdorferi Group/enzymology , Cricetinae , Crystallography, X-Ray , Drug Design , Humans , Models, Biological , Models, Molecular , Protein Binding , Protein Conformation , Streptococcus pyogenes/enzymology , Tritrichomonas foetus/enzymology
6.
Proc Natl Acad Sci U S A ; 96(7): 3531-6, 1999 Mar 30.
Article in English | MEDLINE | ID: mdl-10097070

ABSTRACT

Inosine monophosphate dehydrogenase (IMPDH) controls a key metabolic step in the regulation of cell growth and differentiation. This step is the NAD-dependent oxidation of inosine 5' monophosphate (IMP) to xanthosine 5' monophosphate, the rate-limiting step in the synthesis of the guanine nucleotides. Two isoforms of IMPDH have been identified, one of which (type II) is significantly up- regulated in neoplastic and differentiating cells. As such, it has been identified as a major target in antitumor and immunosuppressive drug design. We present here the 2.9-A structure of a ternary complex of the human type II isoform of IMPDH. The complex contains the substrate analogue 6-chloropurine riboside 5'-monophosphate (6-Cl-IMP) and the NAD analogue selenazole-4-carboxamide adenine dinucleotide, the selenium derivative of the active metabolite of the antitumor drug tiazofurin. The enzyme forms a homotetramer, with the dinucleotide binding at the monomer-monomer interface. The 6 chloro-substituted purine base is dehalogenated, forming a covalent adduct at C6 with Cys-331. The dinucleotide selenazole base is stacked against the 6-Cl-IMP purine ring in an orientation consistent with the B-side stereochemistry of hydride transfer seen with NAD. The adenosine end of the ligand interacts with residues not conserved between the type I and type II isoforms, suggesting strategies for the design of isoform-specific agents.


Subject(s)
IMP Dehydrogenase/chemistry , IMP Dehydrogenase/metabolism , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/chemistry , Adenosine Diphosphate/metabolism , Amino Acid Sequence , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Binding Sites , Crystallography, X-Ray , Drug Design , Humans , IMP Dehydrogenase/antagonists & inhibitors , Immunosuppressive Agents/chemical synthesis , Immunosuppressive Agents/chemistry , Inosine Monophosphate/analogs & derivatives , Inosine Monophosphate/chemistry , Inosine Monophosphate/metabolism , Ligands , Macromolecular Substances , Models, Molecular , Molecular Sequence Data , Organoselenium Compounds/chemistry , Organoselenium Compounds/metabolism , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Structure, Secondary , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
7.
Biochemistry ; 37(26): 9295-304, 1998 Jun 30.
Article in English | MEDLINE | ID: mdl-9649310

ABSTRACT

The oxidation of alcohol to aldehyde by horse liver alcohol dehydrogenase (LADH) requires the transfer of a hydride ion from the alcohol substrate to the cofactor nicotinamide adenine dinucleotide (NAD). A quantum mechanical tunneling contribution to this hydride transfer step has been demonstrated in a number of LADH mutants designed to enhance or diminish this effect [Bahnson, B. J., et al. (1997) Proc. Natl. Acad. Sci. U.S.A. 94, 12797-12802]. The active site double mutant Phe93 --> Trp/Val203 --> Ala shows a 75-fold reduction in catalytic efficiency relative to that of the native enzyme, and reduced tunneling relative to that of either single mutant. We present here two crystal structures of the double mutant: a 2.0 A complex with NAD and the substrate analogue trifluoroethanol and a 2.6 A complex with the isosteric NAD analogue CPAD and ethanol. Changes at the active site observed in both complexes are consistent with reduced activity and tunneling. The NAD-trifluoroethanol complex crystallizes in the closed conformation characteristic of the active enzyme. However, the NAD nicotinamide ring rotates away from the substrate, toward the space vacated by replacement of Val203 with the smaller alanine. Replacement of Phe93 with the larger tryptophan also produces unfavorable steric contacts with the nicotinamide carboxamide group, potentially destabilizing hydrogen bonds required to maintain the closed conformation. These contacts are relieved in the second complex by rotation of the CPAD pyridine ring into an unusual syn orientation. The resulting loss of the carboxamide hydrogen bonds produces an open conformation characteristic of the apoenzyme.


Subject(s)
Alcohol Dehydrogenase/chemistry , Alcohol Dehydrogenase/genetics , Liver/enzymology , Alanine/genetics , Alcohol Dehydrogenase/metabolism , Alcohol Oxidoreductases/chemistry , Animals , Binding Sites/genetics , Coenzymes/chemistry , Crystallography, X-Ray , Horses , Ligands , Models, Molecular , NAD/analogs & derivatives , NAD/chemistry , Niacinamide/chemistry , Phenylalanine/genetics , Structure-Activity Relationship , Substrate Specificity , Tryptophan/genetics , Valine/genetics
8.
J Med Chem ; 41(4): 618-22, 1998 Feb 12.
Article in English | MEDLINE | ID: mdl-9484510

ABSTRACT

Mycophenolic alcohol (MPAlc), obtained by reduction of the carboxylic group of mycophenolic acid (MPA), was coupled with 2',3'-O-isopropylideneadenosine 5'-methylenebis(phosphonate) (4) in the presence of diisopropylcarbodiimide (DIC) to give P1-(2',3'-O-isopropylideneadenosin-5'-yl)-P2-(mycophenolic alcohol-6'-yl)methylenebis(phosphonate) (8) in 32% yield. Deisopropy-lidenation of 8 with CF3COOH/H2O afforded the methylenebis(phosphonate) analogue 3 of mycophenolic adenine dinucleotide (MAD). Compound 3, beta-methylene-MAD, was found to be a potent inhibitor of inosine monophosphate dehydrogenase (IMPDH) type II (Ki = 0.3 microM) as well as an inhibitor of growth of K562 cells (IC50 = 1.5 microM). In contrast to MPA and mycophenolic alcohol, beta-methylene-MAD was not converted into the glucuronide when incubated with uridine 5'-diphosphoglucuronyltransferase.


Subject(s)
Antineoplastic Agents/chemical synthesis , Glucuronosyltransferase/metabolism , IMP Dehydrogenase/antagonists & inhibitors , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/chemical synthesis , NAD/analogs & derivatives , Adenine Nucleotides , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Biotransformation , Cell Division/drug effects , Glucuronates , Humans , Indicators and Reagents , Molecular Structure , Mycophenolic Acid/chemistry , Mycophenolic Acid/pharmacology , Mycophenolic Acid/toxicity , Structure-Activity Relationship , Tumor Cells, Cultured
9.
Proc Natl Acad Sci U S A ; 94(24): 12797-802, 1997 Nov 25.
Article in English | MEDLINE | ID: mdl-9371755

ABSTRACT

We present evidence that the size of an active site side chain may modulate the degree of hydrogen tunneling in an enzyme-catalyzed reaction. Primary and secondary kH/kT and kD/kT kinetic isotope effects have been measured for the oxidation of benzyl alcohol catalyzed by horse liver alcohol dehydrogenase at 25 degrees C. As reported in earlier studies, the relationship between secondary kH/kT and kD/kT isotope effects provides a sensitive probe for deviations from classical behavior. In the present work, catalytic efficiency and the extent of hydrogen tunneling have been correlated for the alcohol dehydrogenase-catalyzed hydride transfer among a group of site-directed mutants at position 203. Val-203 interacts with the opposite face of the cofactor NAD+ from the alcohol substrate. The reduction in size of this residue is correlated with diminished tunneling and a two orders of magnitude decrease in catalytic efficiency. Comparison of the x-ray crystal structures of a ternary complex of a high-tunneling (Phe-93 --> Trp) and a low-tunneling (Val-203 --> Ala) mutant provides a structural basis for the observed effects, demonstrating an increase in the hydrogen transfer distance for the low-tunneling mutant. The Val-203 --> Ala ternary complex crystal structure also shows a hyperclosed interdomain geometry relative to the wild-type and the Phe-93 --> Trp mutant ternary complex structures. This demonstrates a flexibility in interdomain movement that could potentially narrow the distance between the donor and acceptor carbons in the native enzyme and may enhance the role of tunneling in the hydride transfer reaction.


Subject(s)
L-Lactate Dehydrogenase/metabolism , Amino Acids/chemistry , Amino Acids/genetics , Catalysis , Crystallography, X-Ray , Hydrogen , Isotopes , L-Lactate Dehydrogenase/chemistry , L-Lactate Dehydrogenase/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation
10.
J Med Chem ; 40(16): 2533-8, 1997 Aug 01.
Article in English | MEDLINE | ID: mdl-9258359

ABSTRACT

Thiazole-4-carboxamide adenine dinucleotide (TAD) analogue 7 containing a fluorine atom at the C2' arabino configuration of the adenine nucleoside moiety was found to be a potent inducer of differentiation of K562 erythroid leukemia cells. This finding prompted us to synthesize its hydrolysis-resistant methylenebis(phosphonate) and difluoromethylenebis(phosphonate) analogues 8 and 9, respectively. Since both TAD and benzamide adenine dinucleotide (BAD) are potent inhibitors of inosine monophosphate dehydrogenase (IMPDH), the corresponding fluorine-substituted methylenebis(phosphonate) analogue 12 of BAD was also synthesized. Thus, 9-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl)adenine (13) was converted in five steps into the corresponding methylenebis(phosphonate) analogue 18. Dehydration of 18 with DCC led to the formation of the bicyclic trisanhydride intermediate 19a, which upon reaction with 2',3'-O-isopropylidenetiazofurin (20) or -benzamide riboside (21) followed by hydrolysis and deprotection afforded the desired methylene-bridged dinucleotides 8 and 12, respectively. The similar displacement of the 5'-mesyl function of 2',3'-O-isopropylidene-5'-O-mesyltiazofurin (24) with the difluoromethylenebis(phosphonic acid) derivative gave the phosphonate 25 which was coupled with 13 to afford 26. The desired difluoromethylenebis(phosphonate) analogue 9 was obtained by deprotection with Dowex 50/H+. This compound as well as beta-CF2-TAD (4) showed improved differentiation-inducing activity over beta-CH2-TAD (3), whereas analogues containing the -CH2-linkage (8 and 12) were inactive.


Subject(s)
Adenine Nucleotides/chemical synthesis , Antimetabolites, Antineoplastic/chemical synthesis , Benzamides/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Fluorine , IMP Dehydrogenase/antagonists & inhibitors , Adenine Nucleotides/pharmacology , Animals , Antimetabolites, Antineoplastic/pharmacology , Benzamides/pharmacology , Cell Differentiation/drug effects , Chromatography, High Pressure Liquid , Enzyme Inhibitors/pharmacology , Humans , Tumor Cells, Cultured
11.
J Med Chem ; 40(11): 1731-7, 1997 May 23.
Article in English | MEDLINE | ID: mdl-9171883

ABSTRACT

The synthesis and biological activity of selenophenfurin (5-beta-D-ribofuranosylselenophene-3-carboxamide, 1), the selenophene analogue of selenazofurin, are described. Glycosylation of ethyl selenophene-3-carboxylate (6) under stannic chloride-catalyzed conditions gave 2- and 5-glycosylated regioisomers, as a mixture of alpha- and beta-anomers, and the beta-2,5-diglycosylated derivative. Deprotected ethyl 5-beta-D-ribofuranosylselenophene-3-carboxylate (12 beta) was converted into selenophenfurin by ammonolysis. The structure of 12 beta was determined by 1H- and 13C-NMR, crystallographic, and computational studies. Selenophenfurin proved to be antiproliferative against a number of leukemia, lymphoma, and solid tumor cell lines at concentrations similar to those of selenazofurin but was more potent than the thiophene and thiazole analogues thiophenfurin and tiazofurin. Incubation of K562 cells with selenophenfurin resulted in inhibition of IMP dehydrogenase (IMPDH) (76%) and an increase in IMP pools (14.5-fold) with a concurrent decrease in GTP levels (58%). The results obtained confirm the hypothesis that the presence of heteroatoms such as S or Se in the heterocycle in position 2 with respect to the glycosidic bond is essential for both cytotoxicity and IMP dehydrogenase inhibitory activity in this type of C-nucleosides.


Subject(s)
Antineoplastic Agents , Enzyme Inhibitors/chemical synthesis , IMP Dehydrogenase/antagonists & inhibitors , Organoselenium Compounds/chemistry , Organoselenium Compounds/chemical synthesis , Organoselenium Compounds/pharmacology , Ribonucleosides/chemistry , Ribonucleosides/chemical synthesis , Ribonucleosides/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Computer Simulation , Crystallography, X-Ray , Guanosine Triphosphate/metabolism , Humans , Inosine Monophosphate/metabolism , Leukemia/pathology , Lymphoma/pathology , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Molecular Structure , Neoplasms/pathology , Ribavirin/analogs & derivatives , Tumor Cells, Cultured
12.
J Med Chem ; 40(8): 1287-91, 1997 Apr 11.
Article in English | MEDLINE | ID: mdl-9111303

ABSTRACT

beta-Methylene-BAD (8), a nonhydrolyzable analogue of benzamide adenine dinucleotide (BAD), was synthesized as potential inhibitor of human inosine monophosphate dehydrogenase (IMPDH). Treatment of 2',3'-O-isopropylideneadenosine 5'-methylenebisphosphonate (15) with DCC afforded P1,P4-bis(2',3'-O-isopropylideneadenosine) 5'-P1,P2:P3,P4-dimethylenetetrakisphosphonate (17). This compound was further converted with DCC to an active intermediate 18 which upon reaction with 3-(2',3'-O-isopropylidene-beta-D-ribofuranosyl)benzamide (19) gave, after hydrolysis and deisopropylidenation, the desired beta-methylene-BAD (8) in 95% yield. In a similar manner, treatment of 18 with 2',3'-O-isopropylidenetiazofurin (21) followed by hydrolysis and deprotection afforded beta-methylene-TAD (5) in 91% yield. Compound 8 (IC50 = 0.665 microM) was found to be a 6-8 times less potent inhibitor of IMPDH than 5 (IC50 = 0.107 microM) and was almost equally potent against IMPDH type I and type II. Although TAD and beta-methylene-TAD were bound by LADH with the same affinity, the binding affinity of 8 toward LADH (Ki = 333 microM) was found to be 50-fold lower than that of the parent pyrophosphate 7 (Ki = 6.3 microM).


Subject(s)
Adenine Nucleotides/chemical synthesis , Antimetabolites, Antineoplastic/chemical synthesis , Benzamides/chemical synthesis , Enzyme Inhibitors/chemical synthesis , IMP Dehydrogenase/antagonists & inhibitors , Adenine Nucleotides/chemistry , Adenine Nucleotides/pharmacology , Alcohol Dehydrogenase/antagonists & inhibitors , Animals , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacology , Benzamides/chemistry , Benzamides/pharmacology , Chromatography, High Pressure Liquid , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Horses , Humans , Isoenzymes/antagonists & inhibitors , Kinetics , Liver/enzymology , Tumor Cells, Cultured
13.
J Med Chem ; 39(12): 2422-6, 1996 Jun 07.
Article in English | MEDLINE | ID: mdl-8691436

ABSTRACT

Treatment of 3-(2,3-O-isopropylidene-beta-D-ribofuranosyl)benzamide (6) with POCl3 in (EtO)3-PO afforded only little phosphorylation product (8, 5%), but the major product was 5'-chlorobenzamide riboside (7, 85%). Reaction of 6 with 2-cyanoethyl N,N-diisopropylchlorophosphoramidite followed by 2-cyanoethanol/tetrazole treatment and oxidation with tert-butyl peroxide gave a 1:1 mixture of the desired 5'-O-bis(2-cyanoethyl) phosphate 9 and the chloro derivative 7. This mixture was treated with methanolic ammonia and partitioned between CHCl3 and water. The 2',3'-O-isopropylidenebenzamide mononucleotide (8) was obtained in 21.2% overall yield from the aqueous layer. Compound 8 was then converted into the corresponding imidazolide 11b which, upon coupling with 2',3'-O-acetonide of AMP, afforded the acetonide of benzamide adenine dinucleotide (15) in 94% yield together with small amounts of symmetrical pyrophosphates P1,P2-bis(2',3'-O-isopropylideneadenosin-5'-yl)pyrophosphate (13, 3%) and P1,P2-bis(2',3'-O-isopropylidene-3-(carbamoylphenyl)-5'-ribosyl)py rophosphate (14, 2%). Deprotection of 15 with Dowex 50/H+ in water afforded the desired benzamide adenine dinucleotide (BAD) in 93% yield. BAD inhibits inosine monophosphate dehydrogenase type I (IC50 = 0.78 microM) and type II (IC50 = 0.88 microM) with same degree of potency.


Subject(s)
Adenine Nucleotides/chemical synthesis , Antimetabolites, Antineoplastic/chemical synthesis , Benzamides/chemical synthesis , Enzyme Inhibitors/chemical synthesis , IMP Dehydrogenase/antagonists & inhibitors , Isoenzymes/antagonists & inhibitors , Adenine Nucleotides/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Benzamides/pharmacology , Chromatography, High Pressure Liquid , Enzyme Inhibitors/pharmacology , Humans , Molecular Structure , Neoplasm Proteins/antagonists & inhibitors , Structure-Activity Relationship
14.
J Med Chem ; 38(19): 3829-37, 1995 Sep 15.
Article in English | MEDLINE | ID: mdl-7562914

ABSTRACT

The syntheses of furan and thiophene analogues of tiazofurin (furanfurin and thiophenfurin, respectively) are described. Direct stannic chloride-catalyzed C-glycosylation of ethyl 3-furan-carboxylate (6) or ethyl 3-thiophencarboxylate (18) with 1,2,3,5-tetra-O-acetyl-D-ribofuranose gave 2- and 5-glycosylated regioisomers, as a mixture of alpha- and beta-anomers, and the beta-2,5-diglycosylated derivatives. Deprotection of ethyl 5-(2,3,5-tri-O-acetyl-beta-D-ribofuranosyl)furan-3-carboxylate (9 beta) and ethyl 5-(2,3,5-tri-O-acetyl-beta-D-ribofuranosyl)thiophene-3-carboxylate (20 beta) with sodium ethoxide afforded ethyl 5-beta-D-ribofuranosylfuran-3-carboxylate (12 beta) and ethyl 5-beta-D-ribofuranosylthiophene-3-carboxylate (23 beta) which were converted into 5-beta-D-ribofuranosylfuran-3-carboxamide (furanfurin, 4) and 5-beta-D-ribofuranosylthiophene-3-carboxamide (thiophenfurin, 5) by reaction with ammonium hydroxide. The anomeric configuration and the site of glycosylation were established by 1H-NMR and proton-proton nuclear Overhauser effect difference spectroscopy. The structure of compound 23 beta was confirmed by X-ray crystallography. Thiophenfurin was found to be cytotoxic in vitro toward murine lymphocytic leukemia P388 and L1210, human myelogenous leukemia K562, human promyelocytic leukemia HL-60, human colon adenocarcinoma LoVo, and B16 melanoma at concentrations similar to that of tiazofurin. In the same test furanfurin proved to be inactive. Thiophenfurin was found active in vivo in BD2F1 mice inoculated with L1210 cells with a % T/C of 168 at 25 mg/kg. K562 cells incubation with thiophenfurin resulted in inhibition of inosine monophosphate (IMP) dehydrogenase (63%) and an increase in IMP pools (6-fold) with a concurrent decrease in GTP levels (42%). Incubation of adenosine-labeled K562 cells with tiazofurin, thiophenfurin, and furanfurin resulted in a 2-fold higher NAD analogue formulation by thiophenfurin than by tiazofurin. Furanfurin was converted to the NAD analogue with only 10% efficiency. The results obtained support the hypothesis that the presence of S in the heterocycle in position 2 with respect to the glycosidic bond is essential for the cytotoxicity and IMP dehydrogenase activity of tiazofurin, while the N atom is not.


Subject(s)
Antineoplastic Agents/chemical synthesis , IMP Dehydrogenase/antagonists & inhibitors , Ribonucleosides/chemical synthesis , Ribonucleosides/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Crystallography, X-Ray , Humans , Inosine Monophosphate/metabolism , Magnetic Resonance Spectroscopy , Mice , NAD/analogs & derivatives , Neoplasms/drug therapy , Ribavirin/analogs & derivatives , Ribavirin/chemistry , Ribavirin/pharmacology , Ribonucleosides/chemistry , Ribonucleotides/metabolism , Tumor Cells, Cultured
15.
J Med Chem ; 38(7): 1098-105, 1995 Mar 31.
Article in English | MEDLINE | ID: mdl-7707313

ABSTRACT

Three analogues of thiazole-4-carboxamide adenine dinucleotide (TAD) (1-3) containing a fluorine atom at the C2' of the adenine nucleoside (in the ribo and arabino configuration) and at the C3' (in the ribo configuration) were synthesized in high yield from the corresponding 5'-monophosphates of 2'-deoxy-2'-fluoroadenosine (9), 9-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl)-adenine (17), and 3'-deoxy-3'-fluoroadenosine (14), respectively. Pure 2',3'-O-isopropylidene-tiazofurin 5'-phosphorimidazolide (8) was obtained by phosphorylation of the protected tiazofurin followed by treatment with carbonyldiimidazole and HPLC purification. Reaction of 8 with 9 in DMF-d7 (monitored by 1H and 31P NMR) afforded the desired dinucleotide 12, which after deisopropylidenation gave 1 in 82% yield. Small amounts of symmetrical dinucleotides AppA (10, 7.2%) and TRppTR (11, 8.0%) were also isolated during HPLC purification of the major product 12. In a similar manner, compounds 2 and 3 were obtained by coupling of 8 with 14 and 17 in 80% and 76% yield, respectively. All newly prepared fluoro-substituted compounds as well as beta-CF2-TAD, earlier synthesized by us, showed good inhibitory activity against inosine monophosphate dehydrogenase type II, the isozyme which is predominant in neoplastic cells. Binding of 1 (Kis = 0.5 microM), 2 (Kis = 0.7 microM), and 3 (Kis = 2.9 microM) was comparable to that of TAD (Ki = 0.2 microM). The difluoromethylene bisphosphonate analogue, beta-CF2-TAD (Ki = 0.17 microM), was found to be equally effective as the best cofactor-type inhibitor, beta-CH2-TAD (Ki = 0.11 microM). Interestingly, the level of inhibition of horse liver alcohol dehydrogenase by these compounds was found to be much lower (0.1 mM for 1 and 2 and no inhibition up to 10 mM for 3). These findings show that inhibition of tumor-induced inosine monophosphate dehydrogenase type II is selective and may be of therapeutic interest.


Subject(s)
Adenine Nucleotides/chemical synthesis , Adenosine Diphosphate/analogs & derivatives , IMP Dehydrogenase/antagonists & inhibitors , NAD/analogs & derivatives , Thiazoles/chemistry , Adenosine Diphosphate/chemistry , Animals , Fluorine , Horses , Humans , NAD/chemistry , Recombinant Proteins
16.
Biochemistry ; 33(39): 11734-44, 1994 Oct 04.
Article in English | MEDLINE | ID: mdl-7918390

ABSTRACT

CNAD (5-beta-D-ribofuranosylnicotinamide adenine dinucleotide) is an isosteric C-glycosidic analogue of NAD(H) containing a neutral pyridine ring. CPAD (5-beta-D-ribofuranosylpicolinamide adenine dinucleotide) is a closely related pyridine-containing analogue with the pyridine nitrogen on the opposite side of the ring. CNAD is a potent and specific inhibitor of horse liver alcohol dehydrogenase (LADH), binding with a dissociation constant in the nanomolar range. CPAD binds LADH with an affinity comparable to that of NAD. Crystal structures of CNAD and CPAD bound to LADH are presented at 2.4 and 2.7 A, respectively. The two complexes are isomorphous, crystallizing in the triclinic system with cell dimensions different from those seen in previous ternary LADH complexes. Structures were solved using the molecular replacement method and refined to crystallographic R values of 18% (CNAD) and 17% (CPAD). Both inhibitors bind to the "closed" form of LADH in the normal cofactor-binding cleft. The conformation of LADH-bound CPAD closely mimics that of LADH-bound NAD(H). The data suggest that alcohol substrate binds directly to the catalytic zinc atom. In the CNAD complex, the pyridine nitrogen replaces alcohol as the fourth coordination ligand to the active site zinc atom, while all other polar interactions remain the same as those of bound NAD(H). The zinc-nitrogen ligand explains the high affinity of CNAD for LADH.


Subject(s)
Alcohol Dehydrogenase/chemistry , NAD/analogs & derivatives , Alcohol Dehydrogenase/metabolism , Animals , Binding Sites , Crystallography, X-Ray , Ethanol/chemistry , Ethanol/metabolism , Horses , Liver/enzymology , Models, Molecular , Molecular Conformation , Molecular Mimicry , NAD/chemistry , NAD/metabolism , Zinc/chemistry
17.
J Med Chem ; 37(11): 1684-8, 1994 May 27.
Article in English | MEDLINE | ID: mdl-8201602

ABSTRACT

Oxazofurin is the inactive oxazole analogue of the C-glycosyl thiazole antitumor agent tiazofurin. Replacement of the thiazole sulfur in tiazofurin with the oxazole oxygen in oxazofurin produces conformational effects that are examined using crystallographic and computational methods. The crystal structure of oxazofurin contains six molecules in the asymmetric unit and has been refined to a standard R value of 6.8% for all data. The six oxazofurin conformers show an average C-glycosidic torsion angle of 70(9) degrees. This value is significantly higher than the average absolute C-glycosidic torsion angle of 24(10) degrees obtained from previous thiazole nucleoside structures. Previous studies suggest that, in tiazofurin, an electrostatic interaction between a positively charged thiazole sulfur and negatively charged furanose oxygen constrains the C-glycosidic torsion angle to a relatively small value. Ab initio molecular orbital studies presented here suggest that the higher C-glycosidic angles observed in the oxazofurin structures result from a repulsive interaction between negatively charged oxazole and furanose oxygens. Thus, it is likely that differences in activity between oxazo- and tiazofurin are either (1) due directly to differences in electronic properties between the thiazole and oxazole rings or (2) due to the variation in C-glycosidic bond conformation resulting from the alteration in the charge distribution of the heterocycle.


Subject(s)
Antineoplastic Agents/chemistry , Oxazoles/chemistry , Ribavirin/analogs & derivatives , Ribose/analogs & derivatives , Chemical Phenomena , Chemistry, Physical , Crystallization , Crystallography, X-Ray , Electrochemistry , Glycosides/chemistry , Glycosylation , Models, Molecular , Molecular Conformation , Ribavirin/chemistry , Ribose/chemistry , Structure-Activity Relationship , Thermodynamics
18.
J Med Chem ; 37(3): 392-9, 1994 Feb 04.
Article in English | MEDLINE | ID: mdl-8308865

ABSTRACT

CNAD (5-beta-D-ribofuranosylnicotinamide adenine dinucleotide) is an isosteric and isomeric analogue of NAD, in which the nicotinamide ring is linked to the sugar via a C-glycosyl (C5-C1') bond. CNAD acts as a general dehydrogenase inhibitor but shows unusual specificity and affinity for liver alcohol dehydrogenase (ADH, EC 1.1.1.1). The pattern of inhibition is congruent to 4 nM, with NAD as the variable substrate. These values are 3-5 orders of magnitude smaller than those obtained for CNAD in other dehydrogenases and are comparable to values observed for the tightest binding ADH inhibitors known. The specificity and affinity of CNAD for ADH are likely due to coordination of the zinc cation at the ADH catalytic site by the CNAD pyridine nitrogen. This is supported by kinetic and computational studies of ADH-CNAD complexes. These results are compared with those for a related analogue, CPAD. In this analogue, displacement of the pyridine nitrogen to the opposite side of the ring removes the specificity for ADH.


Subject(s)
Alcohol Dehydrogenase/antagonists & inhibitors , Liver/enzymology , NAD/pharmacology , Alcohol Dehydrogenase/chemistry , Alcohol Dehydrogenase/metabolism , Animals , Binding Sites , Binding, Competitive , Cattle , Computer Simulation , Horses , Kinetics , Models, Molecular , Molecular Structure , NAD/analogs & derivatives , NAD/chemistry , NAD/metabolism , Thermodynamics
19.
Biochemistry ; 33(1): 23-32, 1994 Jan 11.
Article in English | MEDLINE | ID: mdl-8286346

ABSTRACT

Thiazole-4-carboxamide adenine dinucleotide (TAD) is the active anabolite of the antitumor drug tiazofurin. Beta-methylene TAD (beta-TAD) is a phosphodiesterase-resistant analogue of TAD, active in tiazofurin-resistant cells. Beta-methylene SAD (beta-SAD) is the active selenium derivative of beta-TAD. Both agents are analogues of the cofactor NAD and are capable of acting as general dehydrogenase inhibitors. Crystal structures of beta-TAD and beta-SAD bound to horse liver alcohol dehydrogenase (LADH) are presented at 2.9 and 2.7 A, respectively. Both complexes crystallize in the orthorhombic space group C222(1) and are isomorphous to apo-LADH. Complexes containing beta-TAD and beta-SAD were refined to crystallographic R values of 15% and 16%, respectively, for reflections between 8 A and the minimum d spacing. Conformations of both inhibitors are similar. beta-TAD and beta-SAD bind to the "open" form of LADH in the normal cofactor-binding cleft between the coenzyme and catalytic domains of each monomer. Binding at the adenosine end of each inhibitor resembles that of NAD. However, the positions of the thiazole and selenazole heterocycles are displaced away from the catalytic Zn cation by approximately 4 A. Close intramolecular S-O and Se-O contacts observed in the parent nucleoside analogues are maintained in both LADH-bound beta-TAD and beta-SAD, respectively. These conformational constraints may influence the binding specificity of the inhibitors.


Subject(s)
Adenosine Diphosphate/analogs & derivatives , Alcohol Dehydrogenase/chemistry , Organoselenium Compounds/chemistry , Protein Conformation , Ribavirin/analogs & derivatives , Thiazoles/chemistry , Adenosine Diphosphate/chemistry , Adenosine Diphosphate/metabolism , Alcohol Dehydrogenase/metabolism , Amino Acid Sequence , Animals , Antineoplastic Agents , Binding Sites , Crystallography, X-Ray/methods , Horses , Ligands , Liver/enzymology , Models, Molecular , Molecular Structure , Organoselenium Compounds/metabolism , Thiazoles/metabolism
20.
Carbohydr Res ; 249(1): 95-108, 1993 Oct 18.
Article in English | MEDLINE | ID: mdl-8252557

ABSTRACT

Synthesis of an analogue 3 of thiazole-4-carboxamide adenine-dinucleotide (TAD) in which the beta-oxygen atom of the pyrophosphate bridge is replaced by a difluoromethylene group has been achieved. Likewise, 2'-deoxy-2'-fluoroadenosine containing analogues of TAD (4) and its difluoromethylenediphosphonate congener (5) have been synthesized. Adenosine 5'-difluoromethylenediphosphonate (8) was prepared from 5'-O-tosyladenosine (6) and tris(tetra-n-butylammonium)difluoromethylenediphosphonate (7) by a modified procedure of Poulter's. Compound 8 was converted into the 2',3'-cyclic carbonate 9 by treatment with triethyl orthoformate. Treatment of 9 with 2',3'-O-isopropylidenetiazofurin (10) in pyridine in the presence of DCC gave a mixture of dinucleotide 11 and the isopropylidene-protected diadenosine tetraphosphonate 12. After deprotection of 11, the desired beta-difluoromethylene TAD (3) was separated by HPLC as the minor product. The diadenosine tetraphosphonate 12, an analogue of Ap4A, was obtained as the major component. Alternatively, 2',3'-O-isopropylidenetiazofurin (10) was tosylated, and the product 13 was further converted into the corresponding difluoromethylenediphosphonate 14 by coupling with 7. DCC-catalyzed coupling of 14 with 2'-deoxy-2'-fluoroadenosine (15) followed by deisopropylidenation afforded the analogue 5. Again the corresponding tetraphosphonate analogue of tiazofurin 17 was the predominant product. Dinucleotide 4 was obtained by coupling of the carbonyldiimidazole-activated tiazofurin 5'-monophosphate with 2'-deoxy-2'-fluoroadenosine 5'-monophosphate. 2'-Deoxy-2'-fluoroadenosine (15) was prepared efficiently from the known N6-benzoyl-3'-O-tetrahydropyranyladenosine (18), which was converted into 3'-O-tetrahydropyranyl-2'-O-triflyl-5'-O-trityladenosine (20) by tritylation and triflation. Treatment of 20 with sodium acetate in hexamethylphosphoric triamide, followed by deacetylation afforded 9-(3-O-tetrahydropyranyl-5-O-trityl-beta-D- arabinofuranosyl)-N6-benzoyladenine (22), which was then treated with DAST. After deprotection of the product, 15 was obtained in good yield.


Subject(s)
Adenine Nucleotides/chemical synthesis , Adenosine/analogs & derivatives , Thiazoles/chemical synthesis , Adenine Nucleotides/chemistry , Adenine Nucleotides/toxicity , Fluorine , Indicators and Reagents , Magnetic Resonance Spectroscopy , Molecular Structure , Spectrometry, Mass, Fast Atom Bombardment , Thiazoles/chemistry , Thiazoles/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL