Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Curr Med Chem ; 22(34): 3991-4028, 2015.
Article in English | MEDLINE | ID: mdl-26295463

ABSTRACT

About 500 NAD (P)-dependent enzymes in the cell use NAD (P) as a cofactor or a substrate. This family of broadly diversified enzymes is crucial for maintaining homeostasis of all living organisms. The NAD binding domain of these enzymes is conserved and it was believed that NAD mimics would not be of therapeutic value due to lack of selectivity. Consequently, only mycophenolic acid which selectively binds at the cofactor pocket of NAD-dependent IMP-dehydrogenase (IMPDH) has been approved as an immunosuppressant. Recently, it became clear that the NAD (P)-binding domain was structurally much more diversified than anticipated and numerous highly potent and selective inhibitors of NAD (P) dependent enzymes have been reported. It is likely, that as in the case of protein kinases inhibitors, inhibitors of NAD (P)-dependent enzymes would find soon their way to the clinic. In this review, recent developments of selective inhibitors of NAD-dependent human IMPDH, as well as inhibitors of IMPDHs from parasites, and from bacterial sources are reported. Therapies against Cryptosporidium parvum and the development of new antibiotics that are on the horizon will be discussed. New inhibitors of bacterial NAD-ligases, NAD-kinases, NMN-adenylyl transferases, as well as phosphoribosyl transferases are also described. Although none of these compounds has yet to be approved, the progress in revealing and understanding crucial factors that might allow for designing more potent and efficient drug candidates is enormous and highly encouraging.


Subject(s)
Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemistry , NAD/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bacteria/drug effects , Binding Sites , Cell Survival/drug effects , DNA Ligases/antagonists & inhibitors , DNA Ligases/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , IMP Dehydrogenase/antagonists & inhibitors , IMP Dehydrogenase/metabolism , Molecular Dynamics Simulation , NAD/pharmacology , NAD/therapeutic use , Neoplasms/drug therapy , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism
2.
Curr Med Chem ; 18(13): 1891-908, 2011.
Article in English | MEDLINE | ID: mdl-21517781

ABSTRACT

A large number of enzymes that use nicotinamide adenine dinucleotide NAD or its phosphorylated form NADP as a cofactor or substrate were found to play an important role in the growth and reproduction of living organisms. NAD(P)-dependent and NAD(P)-utilizing enzymes [NAD(P)-addicted?] have been extensively investigated and implicated in a wide variety of diseases. NAD, generally considered a key component involved in redox reactions, has been found to participate in a broad spectrum of cellular processes, including signal transduction, DNA repair, and post-translational protein modifications. The reduced form of NADP, i.e. NADPH, guards the cell against oxidative stress and it has been suggested that suppression of NADPH oxidase activity could result in anti-angiogenesis and anticancer effects. Consequently, small molecule NAD(P)-based inhibitors that selectively bind at the NAD(P)-binding domain of the targeted enzyme have been designed for novel treatment of medical disorders. The NAD(P)-binding domain is modular in nature; it can be divided into three sub-sites, the nicotinamide monophosphate (NMN) binding sub-site (N sub-site), the adenosine monophosphate (AMP) binding sub-site (A sub-site), and the pyrophosphate binding sub-site (P sub-site or P-groove). Each sub-site plays an important role in securing proper and tight binding; however, each has its own requirements. In this review we discuss a number of conformational and structural factors that might affect (improve) the affinity of various inhibitors to these sub-sites, as well as to the whole binding domain. We have focused on potential selectivity of NAD(P)-like molecules toward targeted enzymes and their potential application in biology and medicine.


Subject(s)
Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Enzymes/metabolism , NADP/metabolism , NAD/metabolism , Animals , Enzyme Inhibitors/chemistry , Enzymes/chemistry , Humans , NAD/chemistry , NAD/pharmacology , NADP/chemistry , NADP/pharmacology , Protein Structure, Tertiary , Substrate Specificity
3.
Curr Med Chem ; 15(7): 650-70, 2008.
Article in English | MEDLINE | ID: mdl-18336280

ABSTRACT

Nicotinamide adenine dinucleotide (NAD), generally considered a key component involved in redox reactions, has been found to participate in an increasingly diverse range of cellular processes, including signal transduction, DNA repair, and post-translational protein modifications. In recent years, medicinal chemists have become interested in the therapeutic potential of molecules affecting interactions of NAD with NAD-dependent enzymes. Also, enzymes involved in de novo biosynthesis, salvage pathways, and down-stream utilization of NAD have been extensively investigated and implicated in a wide variety of diseases. These studies have bolstered NAD-based therapeutics as a new avenue for the discovery and development of novel treatments for medical conditions ranging from cancer to aging. Industrial and academic groups have produced structurally diverse molecules which target NAD metabolic pathways, with some candidates advancing into clinical trials. However, further intensive structural, biological, and medical studies are needed to facilitate the design and evaluation of new generations of NAD-based therapeutics. At this time, the field of NAD-therapeutics is most likely at a stage similar to that of the early successful development of protein kinase inhibitors, where analogs of ATP (a more widely utilized metabolite than NAD) began to show selectivity against target enzymes. This review focuses on key representative opportunities for research in this area, which extends beyond the scope of this article.


Subject(s)
Histone Deacetylase Inhibitors , IMP Dehydrogenase/antagonists & inhibitors , NAD/pharmacology , Nicotinamide-Nucleotide Adenylyltransferase/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors , Protein Kinase Inhibitors/pharmacology , Drug Design , Humans , Molecular Structure , NAD/chemistry , NAD/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Stereoisomerism
4.
Biochemistry ; 39(43): 13241-50, 2000 Oct 31.
Article in English | MEDLINE | ID: mdl-11052677

ABSTRACT

Using off-resonance Raman spectroscopy, we have examined each complex along the catalytic pathway of the DNA repair enzyme uracil DNA glycosylase (UDG). The binding of undamaged DNA to UDG results in decreased intensity of the DNA Raman bands, which can be attributed to an increased level of base stacking, with little perturbation in the vibrational modes of the DNA backbone. A specific complex between UDG and duplex DNA containing 2'-beta-fluorodeoxyuridine shows similar increases in the level of DNA base stacking, but also a substrate-directed conformational change in UDG that is not observed with undamaged DNA, consistent with an induced-fit mechanism for damage site recognition. The similar increases in the level of DNA base stacking for the nonspecific and specific complexes suggest a common enzyme-induced distortion in the DNA, potentially DNA bending. The difference spectrum of the extrahelical uracil base in the substrate-analogue complexes reveals only a small electron density reorganization in the uracil ring for the ground state complex, but large 34 cm(-)(1) downshifts in the carbonyl normal modes. Thus, UDG activates the uracil ring in the ground state mainly through H bonds to its C=O groups, without destroying its quasi-aromaticity. This result is at variance with the conclusion from a recent crystal structure, in which the UDG active site significantly distorts the flipped-out pseudouridine analogue such that a change in hybridization at C1 occurs [Parikh, S. S., et al. (2000) Proc. Natl. Acad. Sci. USA 97, 5083]. The Raman vibrational signature of the bound uracil product differs significantly from that of free uracil at neutral pH, and indicates that the uracil is anionic. This is consistent with recent NMR results, which established that the enzyme stabilizes the uracil anion leaving group by 3.4 pK(a) units compared to aqueous solution, contributing significantly to catalysis. These observations are generally not apparent from the high-resolution crystal structures of UDG and its complexes with DNA; thus, Raman spectroscopy can provide unique and valuable insights into the nature of enzyme-DNA interactions.


Subject(s)
DNA Damage , DNA Glycosylases , DNA, Bacterial/chemistry , N-Glycosyl Hydrolases/chemistry , Base Composition , Catalysis , DNA Repair , DNA-Binding Proteins/chemistry , Escherichia coli/chemistry , Escherichia coli/genetics , Floxuridine/chemistry , Furans , Glycosides/chemistry , Hydrogen Bonding , Hydrogen-Ion Concentration , Macromolecular Substances , Nucleic Acid Conformation , Solvents , Spectrum Analysis, Raman , Uracil/chemistry , Uracil-DNA Glycosidase
5.
Carbohydr Res ; 327(1-2): 87-105, 2000 Jul 10.
Article in English | MEDLINE | ID: mdl-10968677

ABSTRACT

The synthesis and biological activity of deoxyfluoro nucleosides are reviewed.


Subject(s)
Fluorine/chemistry , Nucleosides/chemistry , Antineoplastic Agents/chemistry , Antiviral Agents/chemistry
6.
Biochemistry ; 38(37): 11876-86, 1999 Sep 14.
Article in English | MEDLINE | ID: mdl-10508390

ABSTRACT

The nature of the putative general acid His187 in the reaction catalyzed by Escherichia coli uracil DNA glycosylase (UDG) was investigated using X-ray crystallography and NMR spectroscopy. The crystal structures of H187Q UDG, and its complex with uracil, have been solved at 1.40 and 1.60 A resolution, respectively. The structures are essentially identical to those of the wild-type enzyme, except that the side chain of Gln187 is turned away from the uracil base and cannot interact with uracil O2. This result provides a structural basis for the similar kinetic properties of the H187Q and H187A enzymes. The ionization state of His187 was directly addressed with (1)H-(15)N NMR experiments optimized for histidine ring spin systems, which established that His187 is neutral in the catalytically active state of the enzyme (pK(a) <5.5). These NMR experiments also show that His187 is held in the N(epsilon)()2-H tautomeric form, consistent with the crystallographic observation of a 2.9 A hydrogen bond from the backbone nitrogen of Ser189 to the ring N(delta)()1 of His187. The energetic cost of breaking this hydrogen bond may contribute significantly to the low pK(a) of His187. Thus, the traditional view that a cationic His187 donates a proton to uracil O2 is incorrect. Rather, we propose a concerted mechanism involving general base catalysis by Asp64 and electrophilic stabilization of the developing enolate on uracil O2 by a neutral His187.


Subject(s)
DNA Glycosylases , Escherichia coli/enzymology , Histidine/chemistry , N-Glycosyl Hydrolases/chemistry , Uracil/chemistry , Binding Sites/genetics , Carbon Isotopes , Catalysis , Crystallography, X-Ray , Enzyme Stability , Glutamine/genetics , Histidine/genetics , Histidine/metabolism , Hydrogen Bonding , Hydrogen-Ion Concentration , Mutagenesis, Site-Directed , N-Glycosyl Hydrolases/genetics , N-Glycosyl Hydrolases/metabolism , Nitrogen Isotopes , Nuclear Magnetic Resonance, Biomolecular , Protons , Substrate Specificity , Uracil/metabolism , Uracil-DNA Glycosidase
7.
Nucleosides Nucleotides ; 18(4-5): 927-32, 1999.
Article in English | MEDLINE | ID: mdl-10432711

ABSTRACT

An effective treatment of myeloid leukemias would rely on inducing myeloid cells to undergo differentiation. It has been demonstrated that inhibition of IMPDH with mycophenolic acid or tiazofurin resulted in inhibition of cell growth as well as induction of differentiation. We synthesized a number of bis(phosphonate) analogues of tiazofurin-, benzamide-, and mycophenolic-adenine dinucleotide which were found to be cytotoxic as well as effective inducers of cell differentiation.


Subject(s)
Adenine Nucleotides/pharmacology , Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Adenine Nucleotides/chemistry , Adenine Nucleotides/therapeutic use , Antineoplastic Agents/therapeutic use , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , IMP Dehydrogenase/antagonists & inhibitors , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/pathology , Mycophenolic Acid/chemistry , Organophosphorus Compounds/chemistry , Tumor Cells, Cultured
8.
Curr Med Chem ; 6(7): 629-34, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10390605

ABSTRACT

Mycophenolic acid (MPA) is the most potent and specific inhibitor of inosine monophosphate dehydrogenase (IMPDH). This compound was reported to bind the NAD site of IMPDH and mimic the binding of nicotinamide moiety of nicotinamide adenine dicnucleotide. We linked MPA derivatives with the adenine moiety of NAD through a methylenebis(phonphonate) birdge to form novel mycophenolic adenine dinucleotides (MADs) which resemble well the intact natural cofactor. The MAD analogues differ by the length of the side chain (linker) between the aromatic ring of mycophenolic derivative and the beta-phosphorus atom of the adenosine bis(phosphonate) moiety. Regardless of the linker size, MADs were found to be potent inhibitors of human IMPDH type I and type II with Ki's = 0.25-0.52 microM, an order of magnitude less potent than MPA itself (Ki = 0.01-0.04 microM). The growth of K562 cells was inhibited by MPA (IC50 = 0.03 microM) and the MAD analogues (IC50 = 0.01-1.15 microM) with a similar potency. Accordingly, a suppression of alloantigen- induced proliferation of human lymphocytes by the MAD analogues at concentration of 10-20 microM was equally effective as that observed for MPA. In contrast to MPA, MAD analogues were found to be resistant to glucuronidation in vitro. Since therapeutic potential of MPA is limited by its undesirable glucuronidation, the glucuronidation- resistant MAD analogues may be superior immunosuppressants if they are not glucuronidated in vivo.


Subject(s)
Immunosuppressive Agents/chemical synthesis , Immunosuppressive Agents/pharmacology , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/pharmacology , Dose-Response Relationship, Drug , HT29 Cells , Humans , IMP Dehydrogenase/antagonists & inhibitors , K562 Cells , Kinetics , Mycophenolic Acid/chemical synthesis , NAD/analogs & derivatives
9.
Nucleosides Nucleotides ; 18(3): 363-76, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10358941

ABSTRACT

A synthesis of the C-nucleoside, 2-amino-7-(2-deoxy-beta-D-erythro- pentofuranosyl)-3H,5H-pyrrolo[3,2-d]pyrimidin-4-one (9-deaza-2'-deoxyguanosine) was achieved starting from 2-amino-6-methyl-3H-pyrimidin-4-one (5) and methyl 2-deoxy-3,5-di-O-(p-nitrobenzoyl)-D-erythro-pento-furanoside (11). The anomeric configuration of the C-nucleoside was established by 1H NMR, NOEDS and ROESY. This C-nucleoside did not inhibit the growth of T-cell lymphoma cells.


Subject(s)
Antimetabolites, Antineoplastic/chemical synthesis , Deoxyguanosine/analogs & derivatives , Animals , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/toxicity , Cell Survival/drug effects , Deoxyguanosine/chemical synthesis , Deoxyguanosine/chemistry , Deoxyguanosine/toxicity , Drug Screening Assays, Antitumor , Indicators and Reagents , Leukemia L1210 , Lymphoma, T-Cell , Magnetic Resonance Spectroscopy , Mice , Molecular Conformation , Molecular Structure , Tumor Cells, Cultured
10.
Biochemistry ; 38(3): 952-63, 1999 Jan 19.
Article in English | MEDLINE | ID: mdl-9893991

ABSTRACT

The DNA repair enzyme uracil DNA glycosylase (UDG) catalyzes hydrolytic cleavage of the N-glycosidic bond of premutagenic uracil residues in DNA by flipping the uracil base from the DNA helix. The mechanism of base flipping and the role this step plays in site-specific DNA binding and catalysis by enzymes are largely unknown. The thermodynamics and kinetics of DNA binding and uracil flipping by UDG have been studied in the absence of glycosidic bond cleavage using substrate analogues containing the 2'-alpha and 2'-beta fluorine isomers of 2'-fluoro-2'-deoxyuridine (Ubeta, Ualpha) positioned adjacent to a fluorescent nucleotide reporter group 2-aminopurine (2-AP). Activity measurements show that DNA containing a Ubeta or Ualpha nucleotide is a 10(7)-fold slower substrate for UDG (t1/2 approximately 20 h), which allows measurements of DNA binding and base flipping in the absence of glycosidic bond cleavage. When UDG binds these analogues, but not other DNA molecules, a 4-8-fold 2-AP fluorescence enhancement is observed, as expected for a decrease in 2-AP base stacking resulting from enzymatic flipping of the adjacent uracil. Thermodynamic measurements show that UDG forms weak nonspecific complexes with dsDNA (KDns = 1.5 microM) and binds approximately 25-fold more tightly to Ubeta containing dsDNA (KDapp approximately 50 nM). Thus, base flipping contributes less than approximately 2 kcal/mol to the free energy of binding and is not a major component of the >10(6)-fold catalytic specificity of UDG. Kinetic studies at 25 degrees C show that site-specific binding occurs by a two-step mechanism. The first step (E + S left and right arrow ES) involves the diffusion-controlled binding of UDG to form a weak nonspecific complex with the DNA (KD approximately 1.5-3 microM). The second step (ES left and right arrow E'F) involves a rapid step leading to reversible uracil flipping (kmax approximately 1200 s-1). This step is followed closely by a conformational change in UDG that was monitored by the quenching of tryptophan fluorescence. The results provide evidence for an enzyme-assisted mechanism for uracil flipping and exclude a passive mechanism in which the enzyme traps a transient extrahelical base in the free substrate. The data suggest that the duplex structure of the DNA is locally destabilized before the base-flipping step, thereby facilitating extrusion of the uracil. Thus, base flipping contributes little to the free energy of DNA binding but contributes greatly to specificity through an induced-fit mechanism.


Subject(s)
DNA Damage , DNA Glycosylases , DNA Repair , Escherichia coli/enzymology , N-Glycosyl Hydrolases/chemistry , Uracil/chemistry , Binding Sites , Escherichia coli/genetics , Floxuridine/analogs & derivatives , Floxuridine/chemistry , Kinetics , Macromolecular Substances , Spectrometry, Fluorescence , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substrate Specificity , Thermodynamics , Uracil-DNA Glycosidase
11.
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
12.
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
13.
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
14.
Pharmacol Ther ; 76(1-3): 89-100, 1997.
Article in English | MEDLINE | ID: mdl-9535171

ABSTRACT

Synthetic nicotinamide adenine dinucleotide (NAD) analogues containing 5-beta-D-ribofuranosylnicotinamide (C-NAD), 6-beta-D-ribofuranosylpicolinamide (C-PAD), 3-beta-D-ribofuranosylbenzamide (BAD), and 2-beta-D-ribofuranosylthiazole-4-carboxamide (TAD) in place of the nicotinamide riboside moiety are described and evaluated as potential inhibitors of inosine monophosphate dehydrogenase (IMPDH). TAD and BAD showed potent inhibitory activity against the enzyme in the form of pyrophosphates, as well as metabolically stable methylene- and difluoromethylenebis(phosphonate)s. Fluorination at the C2' (ribo and arabino configuration) and C3' (ribo) of the adenosine moiety of TAD afforded analogues highly potent against IMPDH, but weakly active against alcohol dehydrogenase. With the exception of the methylenebis(phosphonate) analogue of TAD compounds containing a methylene bridge were poor inhibitors of growth of K562 cells. On the other hand, NAD analogues containing difluoromethylene linkage were highly effective in inhibition of K562 cell growth, as well as potent inducers of K562 cell differentiation. Such compounds, therefore, may be of potential therapeutic interest.


Subject(s)
Antineoplastic Agents/therapeutic use , NAD/analogs & derivatives , NAD/therapeutic use , Animals , Antineoplastic Agents/chemistry , Enzyme Inhibitors/classification , Enzyme Inhibitors/pharmacology , Humans , IMP Dehydrogenase/antagonists & inhibitors , NAD/chemical synthesis , Neoplasms/drug therapy , Structure-Activity Relationship
15.
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
16.
Acta Biochim Pol ; 43(1): 183-93, 1996.
Article in English | MEDLINE | ID: mdl-8790723

ABSTRACT

Cofactor type inhibitors (NAD-analogues) of IMP-dehydrogenase (IMPDH) were synthesized and their application as potential anticancer agents are discussed. C-nucleoside isosteres of NAD, C-NAD and C-PAD, showed an effective competitive inhibition of IMPDH, C-NAD but not C-PAD caused extremely potent inhibition of alcohol dehydrogenase. We also synthesized compounds in which nicotinamide riboside was replaced with tiazofurin (TAD-analogues) and the 2' and 3'-positions of adenosine part were fluorinated. The ribose ring of 2'-deoxy-2'-fluoroadenosine is in the C3'-endo conformation whereas 3'-deoxy-3'-fluoroadenosine favors the C2'-endo sugar pucker. These derivatives are good inhibitors of IMPDH type II, the isoenzyme dominant in neoplastic cells. In contrast, all these analogues showed rather week inhibitory activity against alcohol dehydrogenase. Nicotinamide riboside derivatives in which the base and the sugar are linked through an oxygen or a methylene bridge were synthesized. NAD-analogues containing such conformationally restricted nicotinamide nucleoside moiety (syn or anti) are expected to be selective inhibitors of B-specific (IMPDH) or A-specific dehydrogenases, respectively.


Subject(s)
Antimetabolites, Antineoplastic/chemical synthesis , IMP Dehydrogenase/antagonists & inhibitors , NAD/analogs & derivatives , NAD/chemical synthesis , Animals , Antimetabolites, Antineoplastic/chemistry , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/therapeutic use , Humans , Molecular Conformation , Molecular Structure , NAD/chemistry , NAD/therapeutic use , Stereoisomerism
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...