Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Viruses ; 13(10)2021 10 02.
Article in English | MEDLINE | ID: mdl-34696413

ABSTRACT

The causative agent of COVID-19, SARS-CoV-2, gains access to cells through interactions of the receptor-binding domain (RBD) on the viral S protein with angiotensin-converting enzyme 2 (ACE2) on the surface of human host cells. Systematic evolution of ligands by exponential enrichment (SELEX) was used to generate aptamers (nucleic acids selected for high binding affinity to a target) to the RBD made from 2'-fluoro-arabinonucleic acid (FANA). The best selected ~79 nucleotide aptamers bound the RBD (Arg319-Phe541) and the larger S1 domain (Val16-Arg685) of the 1272 amino acid S protein with equilibrium dissociation constants (KD,app) of ~10-20 nM, and binding half-life for the RBD, S1 domain, and full trimeric S protein of 53 ± 18, 76 ± 5, and 127 ± 7 min, respectively. Aptamers inhibited the binding of the RBD to ACE2 in an ELISA assay. Inhibition, on a per weight basis, was similar to neutralizing antibodies that were specific for RBD. Aptamers demonstrated high specificity, binding with about 10-fold lower affinity to the related S1 domain from the original SARS virus, which also binds to ACE2. Overall, FANA aptamers show affinities comparable to previous DNA aptamers to RBD and S1 protein and directly block receptor interactions while using an alternative Xeno-nucleic acid (XNA) platform.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Antiviral Agents/pharmacology , Aptamers, Nucleotide/pharmacology , Arabinonucleotides/pharmacology , SARS-CoV-2/drug effects , Virus Attachment/drug effects , Humans , Protein Binding/drug effects , Protein Domains/drug effects , Protein Domains/genetics , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/metabolism , COVID-19 Drug Treatment
2.
ChemMedChem ; 9(9): 2138-49, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25125220

ABSTRACT

A group of acidic nucleosides were synthesized to develop a new class of ribonuclease A (RNase A) inhibitors. Our recent study on carboxymethylsulfonyl-modified nucleosides revealed some interesting results in RNase A inhibition. This positive outcome triggered an investigation of the role played by secondary sugar hydroxy groups in inhibiting RNase A activity. Uridines and cytidines modified with SO2 CH2 COOH groups at the 2'- and 3'-positions show good inhibitory properties with low inhibition constant (Ki ) values in the range of 109-17 µM. The present work resulted in a set of inhibitors that undergo more effective interactions with the RNase A active site, as visualized by docking studies.


Subject(s)
Arabinonucleotides/chemical synthesis , Arabinonucleotides/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Ribonuclease, Pancreatic/antagonists & inhibitors , Catalytic Domain/drug effects , Computational Biology , Cytidine/chemistry , Kinetics , Models, Molecular , Uridine/chemistry
3.
Respir Res ; 10: 39, 2009 May 20.
Article in English | MEDLINE | ID: mdl-19457265

ABSTRACT

BACKGROUND: Recent development in the field of COPD has focused on strategies aimed at reducing the underlying inflammation through selective inhibition of the phosphodiesterase type IV (PDE4) isoform. Although the anti-inflammatory and bronchodilator activity of selective PDE4 inhibitors has been well documented, their low therapeutic ratio and dose-dependent systemic side effects have limited their clinical utility. This study examined the effect of 2'-deoxy-2'-Fluoro-beta-D-Arabinonucleic Acid (FANA)-containing antisense oligonucleotides (AON) targeting the mRNA for the PDE4B/4D and 7A subtypes on lung inflammatory markers, both in vitro and in vivo. METHODS: Normal human bronchial epithelial (NHBE) cells were transfected with FANA AON against PDE4B/4D and 7A alone or in combination. mRNA levels for target PDE subtypes, as well as secretion of pro-inflammatory chemokines were then measured following cell stimulation. Mice were treated with combined PDE4B/4D and 7A AON via endo-tracheal delivery, or with roflumilast via oral delivery, and exposed to cigarette smoke for one week. Target mRNA inhibition, as well as influx of inflammatory cells and mediators were measured in lung lavages. A two-week smoke exposure protocol was also used to test the longer term potency of PDE4B/4D and 7A AONs. RESULTS: In NHBE cells, PDE4B/4D and 7A AONs dose-dependently and specifically inhibited expression of their respective target mRNA. When used in combination, PDE4B/4D and 7A AONs significantly abrogated the cytokine-induced secretion of IL-8 and MCP-1 to near baseline levels. In mice treated with combined PDE4B/4D and 7A AONs and exposed to cigarette smoke, significant protection against the smoke-induced recruitment of neutrophils and production of KC and pro-MMP-9 was obtained, which was correlated with inhibition of target mRNA in cells from lung lavages. In this model, PDE AONs exerted more potent and broader anti-inflammatory effects against smoke-induced lung inflammation than roflumilast. Moreover, the protective effect of PDE4B/4D and 7A AON was maintained when a once-weekly treatment schedule was used. CONCLUSION: These results indicate that inhaled AON against PDE4B/4D and 7A have unique effects on biomarkers that are believed to be important in the pathophysiology of COPD, which supports further development as a potential therapy in this disease.


Subject(s)
Arabinonucleotides/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Cyclic Nucleotide Phosphodiesterases, Type 7/genetics , Inflammation/physiopathology , Lung/physiopathology , Smoke/adverse effects , Smoking/adverse effects , Animals , Bronchi/drug effects , Bronchi/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 7/drug effects , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Humans , Inflammation/enzymology , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Lung/enzymology , Mice , Oligonucleotides, Antisense/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/genetics , Transfection , Tumor Necrosis Factor-alpha/pharmacology
4.
J Med Chem ; 49(26): 7711-20, 2006 Dec 28.
Article in English | MEDLINE | ID: mdl-17181153

ABSTRACT

Cytotoxic nucleosides have proven to be ineffective for the treatment of hepatocellular carcinoma (HCC) due, in part, to their inadequate conversion to their active nucleoside triphosphates (NTP) in the liver tumor and high conversion in other tissues. These characteristics lead to poor efficacy, high toxicity, and a drug class associated with an unacceptable therapeutic index. Cyclic 1-aryl-1,3-propanyl phosphate prodrugs selectively release the monophosphate of a nucleoside (NMP) into CYP3A4-expressing cells, such as hepatocytes, while leaving the prodrug intact in plasma and extrahepatic tissues. This prodrug strategy was applied to the monophosphate of the well-known cytotoxic nucleoside cytosine-1-beta-D-arabinofuranoside (cytarabine, araC). Compound 19S (MB07133), in mice, achieves good liver targeting compared to araC, generating >19-fold higher cytarabine triphosphate (araCTP) levels in the liver than levels of araC in the plasma and >12-fold higher araCTP levels in the liver than in the bone marrow, representing a >120-fold and >28-fold improvement, respectively, over araC administration.


Subject(s)
Antineoplastic Agents/pharmacology , Arabinonucleotides/chemical synthesis , Carcinoma, Hepatocellular/drug therapy , Cytidine Monophosphate/analogs & derivatives , Liver/drug effects , Prodrugs/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Arabinofuranosylcytosine Triphosphate/blood , Arabinonucleotides/pharmacokinetics , Arabinonucleotides/pharmacology , Bone Marrow/drug effects , Bone Marrow/metabolism , Chromatography, High Pressure Liquid , Cytidine Monophosphate/chemical synthesis , Cytidine Monophosphate/pharmacokinetics , Cytidine Monophosphate/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver Neoplasms/drug therapy , Male , Mice , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Rats , Tissue Distribution
5.
Ann N Y Acad Sci ; 1082: 91-102, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17145930

ABSTRACT

Antisense oligonucleotides (AON) are being developed for a wide array of therapeutic applications. Significant improvements in their serum stability, target affinity, and safety profile have been achieved with the development of chemically modified oligonucleotides. Here, we compared 2'-deoxy-2'-fluoro-beta-D-arabinonucleic acid (FANA)-containing AONs with phosphorothioate oligodeoxynucleotides (PS-DNA), 2'-O-methyl-RNA/DNA chimeras and short interfering RNAs (siRNA) with respect to their target knockdown efficacy, duration of action and resistance to nuclease degradation. Results show that two different configurations of FANA/DNA chimeras (altimers and gapmers) were found to have potent antisense activity. Specific target inhibition was observed with both FANA configurations with an estimated EC50 value comparable to that of an siRNA but 20-to 100-fold lower than the other commonly used AONs. Moreover, the FANA/DNA chimeras showed increased serum stability that was correlated with sustained antisense activity for up to 4 days. Taken together, these results indicate that chimeric FANA/DNA AONs are promising new tools for therapeutic gene silencing when increased potency and duration of action are required.


Subject(s)
Arabinonucleotides/pharmacology , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacology , Drug Stability , Gene Silencing , HeLa Cells , Humans , Inhibitory Concentration 50 , Luciferases/antagonists & inhibitors , Luciferases/genetics , RNA, Small Interfering/pharmacology , Structure-Activity Relationship , Thionucleotides/pharmacology , Time Factors
6.
Biochem Pharmacol ; 68(9): 1757-66, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15450941

ABSTRACT

Previous studies including ours have demonstrated that DNA repair is one of the important targets of fludarabine. The aim of this study is to clarify a mechanistic interaction of carboplatin and F-ara-A, from the perspective of F-ara-A-mediated inhibition of DNA repair initiated by carboplatin. Using human quiescent lymphocytes, we focused on DNA repair, since these cells provide a model of dormant cells. To evaluate the carboplatin-induced DNA incision and its repair, we used the alkaline comet assay. When lymphocytes were incubated with carboplatin, a dose-dependent increase in the tail-moment was observed. Then, tail-moment decreased in proportion to the incubation period in fresh media and recovered to the control level at 4 h. DNA rejoining was completely inhibited by F-ara-A at 10 microM through 0 to 6 h after washing out of these drugs and this F-ara-A-induced inhibition was concentration-dependent. Cellular damage after drug exposure was evaluated with the induction of apoptosis as well as cytotoxic effect. Exposure to carboplatin alone did not induce any apparent cellular damage in quiescent lymphocytes. In contrast, a more than additive induction of apoptosis as well as an enhancement of cytotoxic action was observed in cells treated with a combination of carboplatin and F-ara-A. In the CEM cell line, there was no enhancement of the cytotoxic action of these drugs, despite the clear demonstration of an inhibitory effect on DNA repair. These results indicate that chemotherapy with carboplatin opened a new target for F-ara-A by initiating DNA repair in quiescent cells.


Subject(s)
Antineoplastic Agents/pharmacology , Carboplatin/pharmacology , DNA Damage/drug effects , DNA Repair/drug effects , Lymphocytes/drug effects , Vidarabine/analogs & derivatives , Vidarabine/pharmacology , Apoptosis , Arabinonucleotides/pharmacology , Cell Proliferation/drug effects , Drug Interactions , Humans , Lymphocytes/pathology , Tumor Cells, Cultured
7.
Article in English | MEDLINE | ID: mdl-15043161

ABSTRACT

Nucleoside analogs were investigated for their potential to inhibit cowpox virus (a surrogate for variola and monkeypox viruses) in cell culture and in lethal respiratory infections in mice. Cell culture antiviral activity was determined by plaque reduction assays, with cytotoxicity determined by cell proliferation assays. Selectivity indices (SI's, 50% cytotoxic concentration divided by 50% virus-inhibitory concentration) were determined for 15 compounds. Three arabinofuranosyl (Ara) nucleosides showed activity in mouse mammary tumor (C127I) cells: guanine (Ara-G), thymine (Ara-T), and adenine (Ara-A) with SI's of 113, 61, and 95, respectively. The 2'-fluoro-Ara nucleosides of 5-F-cytosine (FIAC), 5-methyluracil (FMAU), and 5-iodouracil (FIAU) exhibited SI's of 148, 77, and 29, respectively. Other potent compounds included cidofovir (a positive control) and 3'-O-methyladenosine, with SI values of 164 and 56, respectively. In general, assays performed in African green monkey kidney (Vero) cells produced lower SI's than in C127I cells, except for 5-iodo-2'-deoxyuridine (IDU) which had an SI of > 71 in Vero cells and 3.1 in C127I cells. Intranasal infection of mice with cowpox virus was followed a day later by twice daily intraperitoneal treatment with compounds for 5 days. Ara-A was active at 300 mg/kg/day (40% survival), FMAU at 100 mg/kg/day (70% survival), and cidofovir (given for 1 day only) at 100 mg/kg (80-100% survival). None of the other compounds, including IDU, prevented death nor delayed the time to death. Cidofovir had the best potential for treating orthopoxvirus infections of those tested.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/pharmacology , Antiviral Agents/pharmacology , Arabinonucleotides/pharmacology , Cowpox virus/drug effects , Cowpox/drug therapy , Animals , Chlorocebus aethiops , Cowpox/veterinary , Female , Mice , Vero Cells
8.
Clin Cancer Res ; 7(11): 3580-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11705880

ABSTRACT

PURPOSE: Chronic lymphocytic leukemia (CLL) lymphocytes respond to DNA alkylation by excision repair, with the extent of repair increasing as the cells acquire resistance to alkylating agents. Because incorporation of nucleotide analogues into the repair patches elicits death signals in quiescent cells, the increased capacity for excision repair in alkylator-resistant cells could facilitate incorporation of nucleotide analogues. We hypothesized that the mechanism-based interaction of nucleoside analogues with alkylating agents could elicit greater than additive killing of CLL cells. EXPERIMENTAL DESIGN: Lymphocytes from 50 patients with CLL that were not refractory to alkylators were treated in vitro with 4-hydroperoxycyclophosphamide (4-HC) with or without prior incubation with fludarabine nucleoside (F-ara-A) or with clofarabine (Cl-F-ara-A). DNA damage repair kinetics were determined by the single-cell gel electrophoresis (comet) assay. Cytotoxicity was assessed by staining with annexin V. RESULTS: CLL lymphocytes promptly initiated and completed excision repair in response to 4-HC. A 2-h preincubation with 10 microM F-ara-A or 10 microM Cl-F-ara-A inhibited the repair initiated by 4-HC, with inhibition peaking at the intracellular concentrations of 50 microM F-ara-ATP or 5 microM Cl-F-ara-ATP. Combining 4-HC with either F-ara-A or Cl-F-ara-A produced more than additive apoptotic cell death than the sum of each alone. The increase in cytotoxicity was proportional to the initial magnitude of the DNA incision and to the extent of repair inhibition by the nucleoside analogues, suggesting close correlation between the repair inhibition and induction of cell death. CONCLUSIONS: DNA repair, which is active in CLL lymphocytes, may be a biological target for facilitating the incorporation of nucleoside analogues and increasing their cytotoxicity. Thus, the increased repair capacity associated with resistant disease may be manipulated to therapeutic advantage.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclophosphamide/analogs & derivatives , DNA Repair/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Lymphocytes/drug effects , Vidarabine/analogs & derivatives , Adenine Nucleotides , Apoptosis/drug effects , Arabinonucleosides/pharmacology , Arabinonucleotides/pharmacology , Clofarabine , Cyclophosphamide/pharmacology , DNA/biosynthesis , DNA/drug effects , DNA/genetics , Dose-Response Relationship, Drug , Humans , Lymphocytes/metabolism , Nucleosides/chemistry , Nucleosides/pharmacology , Time Factors , Vidarabine/chemistry , Vidarabine/pharmacology
9.
J Biol Chem ; 275(33): 25814-9, 2000 Aug 18.
Article in English | MEDLINE | ID: mdl-10833512

ABSTRACT

A 3'-5' exonuclease that excises the nucleotide analogs 1-beta-d-arabinofuranosylcytosine monophosphate and 9-beta-d-arabinofuranosyl-2-fluoroadenine 5'-monophosphate incorporated at 3' ends of DNA was purified from the nuclei of: 1) primary human chronic lymphocytic leukemia cells, 2) primary and established human acute myeloblastic leukemia cells, and 3) lymphocytes obtained from healthy individuals. The activity of this nuclear exonuclease (exoN) is elevated approximately 6-fold in 1-beta-d-arabinofuranosylcytosine-resistant leukemia cells as compared with drug-sensitive cells, and it differs between two healthy individuals and among three leukemia patients. exoN is a 46-kDa monomer, requires 50 mm KCl and 1 mm magnesium for optimal activity, and shows a preference for single-stranded over duplex DNA. Its physical and enzymatic properties indicate that exoN is a previously uncharacterized enzyme whose activity may confer resistance to clinical nucleoside analogs in leukemia cells.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Arabinonucleotides/pharmacology , Cytidine Monophosphate/analogs & derivatives , Drug Resistance, Neoplasm , Exodeoxyribonucleases/metabolism , Exonucleases/biosynthesis , Exonucleases/chemistry , Leukemia/enzymology , Neoplasm Proteins , Vidarabine Phosphate/analogs & derivatives , Cell Nucleus/enzymology , Cells, Cultured , Cytidine Monophosphate/pharmacology , DNA-Directed DNA Polymerase/metabolism , Exodeoxyribonuclease V , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/isolation & purification , Exonucleases/metabolism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/enzymology , Lymphocytes/enzymology , Magnesium/metabolism , Potassium Chloride/metabolism , Protein Biosynthesis , Time Factors , Transcription, Genetic , Tumor Cells, Cultured , Up-Regulation , Vidarabine Phosphate/pharmacology
10.
Nucleic Acids Symp Ser ; (44): 175-6, 2000.
Article in English | MEDLINE | ID: mdl-12903325

ABSTRACT

Telomerase, which synthesizes telomeric DNA in eukaryotic cells, is classified as a reverse transcriptase. To clarify the susceptibility of telomerase to nucleoside 5'-triphosphates bearing a hydrophobic group on the base moiety, we studied the inhibitory effects of 2',3'-dideoxy-5-styryluridine 5'-triphosphate analogues and 9-(beta-D-arabinofuranosyl)-2-(p-n-butylanilino)purine 5'-triphosphate analogues on telomerase activity using a quantitative 'stretch PCR' assay. 2',3'-Dideoxy-5-styryluridine 5'-triphosphate (StddUTP) showed more potent inhibition than 2',3'-dideoxythymidine 5'-triphosphate (ddTTP). On the other hand, 9-(beta-D-arabinofuranosyl)-2-(p-n-butylphenyl)guanine 5'-triphosphate (BuParaGTP) showed no inhibition, even though 9-(beta-D-arabinofuranosyl)guanine 5'-triphosphate (araGTP) is a potent inhibitor of telomerase. The influence on telomerase of hydrophobic substituents on the base moieties of nucleotides is described.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Guanosine Triphosphate/analogs & derivatives , Nucleosides/chemistry , Nucleosides/pharmacology , Telomerase/antagonists & inhibitors , Arabinonucleotides/chemistry , Arabinonucleotides/pharmacology , Dideoxynucleotides , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/pharmacology , Humans , In Vitro Techniques , Kinetics , Structure-Activity Relationship , Thymine Nucleotides/chemistry , Thymine Nucleotides/pharmacology , Uridine Triphosphate/analogs & derivatives , Uridine Triphosphate/chemistry , Uridine Triphosphate/pharmacology
11.
J Biol Chem ; 270(44): 26664-9, 1995 Nov 03.
Article in English | MEDLINE | ID: mdl-7592892

ABSTRACT

Oligonucleotides containing a specific initiation site for polymerase alpha-primase (pol alpha-primase) were used to measure the effects of cytosine arabinoside triphosphate and cytosine arabinoside monophosphate (araCMP) in DNA on RNA-primed DNA synthesis. Primase inserts araCMP at the 3' terminus of a full-length RNA primer with a 400-fold preference over CMP. The araCMP is elongated efficiently by pol alpha in the primase-coupled reaction. Extension from RNA 3'-araCMP is 50-fold less efficient than from CMP, and extension from DNA 3'-araCMP is 1600-fold less efficient than from dCMP. Using araCMP-containing templates, primer synthesis is reduced 2-3-fold, and RNA-primed DNA synthesis is reduced 2-8-fold. The efficiency of polymerization past a template araCMP by pol alpha is reduced 180-fold during insertion of dGMP opposite araCMP and 35-fold during extension from the araCMP:dGMP 3' terminus. These results show that the pol alpha-primase efficiently incorporates araCMP as the border nucleotide between RNA and DNA and suggest that the inhibitory effects of araC most likely result from slowed elongation of pol alpha and less so from inhibition of primer synthesis by primase.


Subject(s)
Arabinonucleotides/pharmacology , Cytarabine/pharmacology , Cytidine Monophosphate/analogs & derivatives , DNA Replication/drug effects , RNA Nucleotidyltransferases/metabolism , Base Sequence , Cytidine Monophosphate/pharmacology , DNA Primase , DNA Primers , Kinetics , Molecular Sequence Data , RNA Nucleotidyltransferases/drug effects , Templates, Genetic
12.
Nucleic Acids Res ; 22(6): 893-900, 1994 Mar 25.
Article in English | MEDLINE | ID: mdl-8152919

ABSTRACT

The ribonucleoprotein enzyme telomerase is a specialized type of cellular reverse transcriptase which synthesizes one strand of telomeric DNA, using as the template a sequence in the RNA moiety of telomerase. We analyzed the effects of various nucleoside analogs, known to be chain-terminating inhibitors of retroviral reverse transcriptases, on Tetrahymena thermophila telomerase activity in vitro. We also analyzed the effects of such analogs on telomere length and maintenance in vivo, and on vegetative growth and mating of Tetrahymena cells. Arabinofuranyl-guanosine triphosphate (Ara-GTP) and ddGTP both efficiently inhibited telomerase activity in vitro, while azidothymidine triphosphate (AZT-TP), dideoxyinosine triphosphate (ddITP) or ddTTP were less efficient inhibitors. All of these nucleoside triphosphate analogs, however, produced analog-specific alterations of the normal banding patterns seen upon gel electrophoresis of the synthesis products of telomerase, suggesting that their chain terminating and/or competitive actions differ at different positions along the RNA template. The analogs AZT, 3'-deoxy-2',3'-didehydrothymidine (d4T) and Ara-G in nucleoside form caused consistent and rapid telomere shortening in vegetatively growing Tetrahymena. In contrast, ddG or ddI had no effect on telomere length or cell growth rates. AZT caused growth rates and viability to decrease in a fraction of cells, while Ara-G had no such effects even after several weeks in culture. Neither AZT, Ara-G, acycloguanosine (Acyclo-G), ddG nor ddI had any detectable effect on cell mating, as assayed by quantitation of the efficiency of formation of progeny from mated cells. However, AZT decreased the efficiency of programmed de novo telomere addition during macronuclear development in mating cells.


Subject(s)
DNA Nucleotidylexotransferase/antagonists & inhibitors , Nucleotides/pharmacology , Telomere/drug effects , Tetrahymena thermophila/enzymology , Tetrahymena thermophila/genetics , Animals , Arabinonucleotides/pharmacology , Base Sequence , Cell Survival/drug effects , DNA Nucleotidylexotransferase/metabolism , Deoxyguanine Nucleotides/pharmacology , Dideoxynucleotides , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/pharmacology , Molecular Sequence Data , Telomere/physiology , Telomere/ultrastructure , Tetrahymena thermophila/physiology , Thymine Nucleotides/pharmacology , Zidovudine/analogs & derivatives , Zidovudine/pharmacology
13.
Cancer Res ; 54(1): 109-13, 1994 Jan 01.
Article in English | MEDLINE | ID: mdl-8261429

ABSTRACT

1-beta-D-Arabinofuranosylcytosine-5'-stearylphosphate (cytarabine ocfosfate, stearyl-ara-CMP) is a newly synthesized 5'-alkylphosphate derivative of 1-beta-D-arabinofuranosylcytosine (ara-C), which is lipophilic, resistant to inactivation by deamination, and orally active. Pharmacology of this drug was studied in patients with hematological malignancies. The concentrations of stearyl-ara-CMP, ara-C (its active metabolite), and 1-beta-D-arabinofuranosyluracil (ara-U, its inactive metabolite) were determined by radioimmunoassay. When six patients received a single p.o. dose of the drug (500 mg/m2), stearyl-ara-CMP, ara-C, and ara-U could be detected in the plasma for at least 72 h afterwards. The plasma disappearance curve of stearyl-ara-CMP corresponded to a one-compartment open model with first-order absorption kinetics. The peak plasma level (Cmax) was 322 +/- 218 nM, and the predicted time to reach Cmax (Tmax) was 6.5 +/- 4.5 h, while the elimination half-life (t1/2) was very long (32.0 +/- 8.4 h). The plasma ara-C level increased slowly to a Cmax of 26.3 +/- 12.7 nM (Tmax, 13.3 +/- 4.7 h) after stearyl-ara-CMP administration. This level was quite low compared with that achieved by low-dose s.c. ara-C therapy, but ara-C persisted longer in the plasma in the former case, and the area under the curve was similar for both regimens. For ara-U, the Cmax, Tmax, and t1/2 were 483 +/- 315 nM, 23.6 +/- 4.0 h, and 19.6 +/- 5.3 h, respectively. No stearyl-ara-CMP was detected in the urine, and only 8.0% of the administered dose was excreted as ara-C and ara-U within 72 h. The stearyl-ara-CMP concentration in the cerebrospinal fluid was below the limit of detection in three patients without meningeal involvement at 6 h. During clinical use of stearyl-ara-CMP, macrocytic anemia was observed, and some patients also developed megaloblastic change of their erythroblasts, suggesting a mild and persistent cytostatic effect. In conclusion, p.o. therapy with stearyl-ara-CMP achieved prolonged maintenance of the plasma drug level. Thus, the drug released a very low dose of ara-C over a long period in plasma and tissues and had a prolonged mild antineoplastic effect in patients with hematological malignancies.


Subject(s)
Anemia, Refractory, with Excess of Blasts/drug therapy , Antineoplastic Agents/pharmacology , Arabinonucleotides/pharmacology , Cytidine Monophosphate/analogs & derivatives , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myeloid/drug therapy , Acute Disease , Adult , Aged , Anemia, Refractory, with Excess of Blasts/metabolism , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Arabinonucleotides/administration & dosage , Arabinonucleotides/pharmacokinetics , Cytidine Monophosphate/administration & dosage , Cytidine Monophosphate/pharmacokinetics , Cytidine Monophosphate/pharmacology , Female , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myeloid/metabolism , Male , Middle Aged , Polycythemia Vera/drug therapy , Polycythemia Vera/metabolism
14.
Biochemistry ; 31(19): 4720-8, 1992 May 19.
Article in English | MEDLINE | ID: mdl-1581321

ABSTRACT

Inhibition of DNA primase and polymerase alpha from calf thymus was examined. DNA primase requires a 3'-hydroxyl on the incoming NTP in order to polymerize it, while the 2'-hydroxyl is advantageous, but not essential. Amazingly, primase prefers to polymerize araATP rather than ATP by 4-fold (kcat/KM). However, after incorporation of an araNMP into the growing primer, further synthesis is abolished. The 2'- and 3'-hydroxyls of the incoming nucleotide appear relatively unimportant for nucleotide binding to primase. Polymerization of nucleoside triphosphates by DNA polymerase alpha onto a DNA primer was similarly analyzed. Removing the 3'-hydroxyl of the incoming triphosphate decreases the polymerization rate greater than 1000-fold (kcat/KM), while a 2'-hydroxyl in the ribo configuration abolishes polymerization. If the 2'-hydroxyl is in the ara configuration, there is almost no effect on polymerization. An araCMP or ddCMP at the 3'-terminus of a DNA primer slightly decreased DNA binding as well as binding of the next correct 2'-dNTP. Changing the primer from DNA to RNA dramatically and unpredictably altered the interactions of pol alpha with araNTPs and ddNTPs. Compared to the identical DNA primer, pol alpha discriminated 4-fold better against araCTP polymerization when the primer was RNA, but 85-fold worse against ddCTP polymerization. Additionally, pol alpha elongated RNA primers containing 3'-terminal araNMPs more efficiently than the identical DNA substrate.


Subject(s)
Arabinonucleotides/pharmacology , DNA Polymerase II/antagonists & inhibitors , RNA Nucleotidyltransferases/antagonists & inhibitors , Animals , Arabinofuranosylcytosine Triphosphate/pharmacology , Base Sequence/drug effects , Catalysis , Cattle , DNA Primase , DNA Replication , Molecular Sequence Data , Structure-Activity Relationship
15.
J Biol Chem ; 267(4): 2345-9, 1992 Feb 05.
Article in English | MEDLINE | ID: mdl-1310314

ABSTRACT

9-beta-D-Arabinofuranosyl-2-fluoroadenine (F-ara-A) is an analogue of adenosine and deoxyadenosine with potent anti-tumor activity. The mechanism of action for this compound has been elucidated as the inhibition of DNA and RNA synthesis, induction of DNA fragmentation, and genetic damage. This study demonstrated that DNA ligase I, an enzyme involved in DNA replication, is a target for the drug action. F-ara-adenine triphosphate (F-ara-ATP) at 80 microM inhibited the activity of DNA ligase I by more than 90%. In contrast, eight other related nucleoside analogues showed no effect on the enzyme activity at 200 microM. F-ara-ATP inhibited DNA ligation in two distinct ways. First, F-ara-ATP directly interacted with DNA ligase I and inhibited the formation of the ligase-AMP complex. This inhibition could not be reversed when free F-ara-ATP was eliminated from the treated enzyme; however, the addition of pyrophosphate, followed by gel filtration chromatography, restored enzyme activity, indicating that F-ara-ATP bound to the enzyme and altered the AMP-binding site. Secondly, the activity of DNA ligase I was inhibited when F-ara-ATP was incorporated into the 3' terminus of the DNA substrate. The dual mode of inhibition of DNA ligase I by F-ara-ATP indicates that its effect on DNA ligation may be important in the inhibition of DNA synthesis and the cytotoxicity of F-ara-A.


Subject(s)
Arabinonucleotides/pharmacology , DNA Ligases/antagonists & inhibitors , Adenosine Monophosphate/metabolism , Base Sequence , Cell Line , Chromatography, Gel , DNA/drug effects , DNA Ligase ATP , DNA Ligases/metabolism , Diphosphates/pharmacology , Electrophoresis, Polyacrylamide Gel , Humans , Molecular Sequence Data , Plasmids
16.
Cancer Chemother Pharmacol ; 31(1): 11-7, 1992.
Article in English | MEDLINE | ID: mdl-1458554

ABSTRACT

Exponentially growing K562 cells incubated with 1-beta-D-arabinofuranosylcytosine (ara-C) accumulate ara-C triphosphate (ara-CTP) at a higher rate and to a greater concentration after pretreatment with 9-beta-D-arabinofuranosyl-2-fluoroadenine (F-ara-A) than do cells treated with ara-C alone. Potentiation of ara-C metabolism is due in part to an indirect effect of F-ara-A triphosphate (F-ara-ATP)-mediated reduction in deoxynucleotide pools and consequent activation of deoxycytidine kinase. Because the levels of deoxynucleotide pools and the activity of deoxycytidine kinase are cell cycle-specific, we investigated the effect of cell cycle phases on the accumulation of ara-CTP and the influence of F-ara-A pretreatment on such accumulation. Exponentially growing K562 cells were fractionated into G1, S, and G2+M phase-enriched subpopulations (each enriched by > 60%) by centrifugal elutriation. The rate of ara-CTP accumulation was 22, 25, and 14 microM/h and the rate of F-ara-ATP accumulation was 38, 47, and 33 microM/h in the G1, S, and G2+M subpopulations, respectively. The rate of elimination of arabinosyl triphosphates was similar among the different phases of the cell cycle. After pretreatment with F-ara-A, the rate of ara-CTP accumulation in the G1, S, and G2+M phase-enriched subpopulations was 43, 37, and 26 microM/h, indicating a 1.7-, 1.5-, and 1.9-fold increase, respectively. These results suggest that a combination of F-ara-A and ara-C may effectively potentiate ara-CTP accumulation in all phases of the cell cycle. This observation is consistent with the results of studies on the modulation of ara-C metabolism by F-ara-A in lymphocytes and leukemia blasts obtained from patients with chronic lymphocytic leukemia and acute myelogenous leukemia, respectively.


Subject(s)
Antineoplastic Agents/metabolism , Arabinonucleotides/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Arabinofuranosylcytosine Triphosphate/metabolism , Arabinofuranosylcytosine Triphosphate/pharmacokinetics , Arabinonucleotides/pharmacokinetics , Arabinonucleotides/pharmacology , Cell Cycle/physiology , Cytarabine/metabolism , Deoxycytidine Kinase/metabolism , Drug Synergism , G1 Phase/physiology , G2 Phase/physiology , Humans , Intracellular Fluid/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Metaphase/physiology , Nucleotides/metabolism , S Phase/physiology , Sensitivity and Specificity , Tumor Cells, Cultured , Vidarabine/analogs & derivatives , Vidarabine/pharmacology
17.
J Biol Chem ; 265(27): 16617-25, 1990 Sep 25.
Article in English | MEDLINE | ID: mdl-1697861

ABSTRACT

The action of 9-beta-D-arabinofuranosyl-2-fluoroadenine (F-ara-A) on DNA synthesis was evaluated both in whole cells and in vitro. 9-beta-D-Arabinofuranosyl-2-fluoroadenine was converted to its 5'-triphosphate 9-beta-D-arabinofuranosyl-2-fluoroadenine 5'-triphosphate (F-ara-ATP) in cells and then incorporated into DNA in a self-limiting manner. More than 94% of the analogue was incorporated into DNA at the 3' termini, indicating a chain termination action. In vitro DNA primer extension experiments further revealed that F-ara-ATP compared with dATP for incorporation into the A site of the extending DNA strand. The incorporation of F-ara-AMP into DNA resulted in termination of DNA strand elongation. Human DNA polymerase alpha incorporated more F-ara-AMP into DNA than polymerase epsilon (proliferating cell nuclear antigen-independent DNA polymerase delta) and was more sensitive to inhibition by F-ara-ATP. On the other hand, DNA polymerase epsilon was able to excise the incorporated F-ara-AMP from DNA in vitro. The incorporation of F-ara-AMP into DNA was linearly correlated both with inhibition of DNA synthesis and with loss of clonogenicity; thus it may be the mechanism of cytotoxicity.


Subject(s)
Cell Survival/drug effects , DNA Replication/drug effects , DNA-Directed DNA Polymerase/metabolism , Vidarabine/analogs & derivatives , Aphidicolin , Arabinonucleotides/metabolism , Arabinonucleotides/pharmacology , Cell Line , Clone Cells , Diterpenes/pharmacology , Guanosine Monophosphate/pharmacology , Humans , Kinetics , Nucleic Acid Synthesis Inhibitors , Vidarabine/pharmacology , Vidarabine Phosphate/analogs & derivatives , Vidarabine Phosphate/metabolism
18.
Gan To Kagaku Ryoho ; 17(8 Pt 1): 1437-45, 1990 Aug.
Article in Japanese | MEDLINE | ID: mdl-2389942

ABSTRACT

4-Amino-1-beta-D-arabinofuranosyl-2(1H)-pyrimidinone 5'-(sodium octadecyl phosphate) monohydrate (YNK01) was an orally active depot form of 1-beta-D-arabinofuranosylcytosine (Ara-C). In the present study, antitumor activity of YNK01 was compared with it of Ara-C in vitro and in vivo. The activity of a main metabolite of YNK01, 5'-carboxypropylphosphate of Ara-C (C-C3PCA), was also studied. Growth inhibitory activity of YNK01 against various cultured tumor cells was 1/32-1/1,100 of that of Ara-C. YNK01 exhibited antitumor activity against L1210 leukemia in mice after i.v., i.p. or p.o. administration. The activity did not depend on the administration routes. Compared with Ara-C, the activity was comparable in both i.v. and i.p. administrations, but greater in p.o. administration. Oral administration of YNK01 showed similar antitumor spectrum to i.p. administration of Ara-C. Oral activity of YNK01 against L1210 leukemia did not depend on the administration schedules but depended on a total administration dose. In contrast, activity of Ara-C greatly depended on the schedules, and the frequent i.p, administration showed greatest activity. Growth inhibitory activity of C-C3PCA against cultured tumor cells was 1/2-1/7 of Ara-C. The metabolite exhibited activity against L1210 leukemia in mice after i.p. administration. These results suggest that YNK01 is a clinically useful drug with p.o. administration for cancers as well as Ara-C.


Subject(s)
Antineoplastic Agents , Arabinonucleotides/pharmacology , Cytidine Monophosphate/pharmacology , Cytosine Nucleotides/pharmacology , Neoplasms, Experimental/drug therapy , Administration, Oral , Animals , Arabinonucleotides/administration & dosage , Arabinonucleotides/therapeutic use , Cytarabine/pharmacology , Cytidine Monophosphate/administration & dosage , Cytidine Monophosphate/analogs & derivatives , Cytidine Monophosphate/therapeutic use , Drug Administration Schedule , Drug Screening Assays, Antitumor , Female , Injections, Intraperitoneal , Injections, Intravenous , Leukemia L1210/drug therapy , Male , Mice , Neoplasms, Experimental/pathology , Tumor Cells, Cultured/drug effects
19.
Eur J Biochem ; 190(3): 463-7, 1990 Jul 05.
Article in English | MEDLINE | ID: mdl-2164928

ABSTRACT

(E)-5-(2-Bromovinyl)-2'-deoxyuridine 5'-triphosphate (BrVdUTP) and (E)-5-(2-bromovinyl)-1-beta-D-arabinofuranosyluracil 5'-triphosphate (BrVarafUTP), which are known as specific inhibitors of herpes simplex viral (type 1 and 2) DNA polymerase, were found to be strong inhibitors of DNA polymerase gamma from human KB and murine myeloma cells. In fact BrVdUTP and BrVarafUTP were found to be stronger inhibitors of DNA polymerase gamma than of other DNA polymerases having viral (herpes simplex virus or retrovirus) origin or cellular (eukaryotic alpha and beta, or prokaryotic) origin. The mode of inhibition of DNA polymerase gamma by BrVdUTP and BrVarafUTP was competitive with respect to dTTP, the normal substrate. Whereas BrVdUTP was an efficient substrate for DNA polymerase gamma and other DNA polymerases that were examined, BrVarafUTP failed to serve as a substrate for DNA synthesis. Ki values for BrVdUTP (40 nM) and BrVarafUTP (7 nM) with DNA polymerase gamma, as determined with (rA)n.(dT) as the template.primer, were much smaller than the Km values for dTTP (0.16 microM and 0.71 microM for murine and human DNA polymerase gamma, respectively). Thus, the affinity of BrVdUTP or BrVarafUTP for DNA polymerase gamma was much stronger than that of dTTP.


Subject(s)
Antiviral Agents/pharmacology , Arabinonucleotides/pharmacology , Bromodeoxyuridine/analogs & derivatives , DNA Polymerase III/antagonists & inhibitors , DNA Replication/drug effects , Nucleic Acid Synthesis Inhibitors , Animals , Bromodeoxyuridine/pharmacology , Cell Line , DNA Polymerase I/antagonists & inhibitors , DNA Polymerase I/isolation & purification , DNA Polymerase II/antagonists & inhibitors , DNA Polymerase II/isolation & purification , DNA Polymerase III/isolation & purification , Escherichia coli/enzymology , Humans , KB Cells , Kinetics , Simplexvirus/enzymology
20.
Cancer Res ; 50(12): 3675-80, 1990 Jun 15.
Article in English | MEDLINE | ID: mdl-2340517

ABSTRACT

This investigation analyzed the metabolism of 2',2'-difluorodeoxycytidine (dFdC) in K562 human leukemia cells and evaluated it as a biochemical modulator for the phosphorylation of several arabinosyl nucleosides. The rate of accumulation of dFdC triphosphate was linear up to 3 h and maximal during incubation with 10 microM dFdC (92 microM/h). Deoxynucleotides analyzed at this time showed a decrease in dCTP, dATP, and dGTP levels, indicating an inhibitory role of dFdC nucleotides in ribonucleotide reduction. We evaluated the hypothesis that dFdC-mediated deoxyribonucleoside triphosphate perturbation enhances the phosphorylation of substrates that use deoxycytidine kinase or deoxyguanosine kinase, because these enzymes are inhibited by dCTP or dGTP, respectively. When the activity of these nucleoside kinases was rate limiting to triphosphate formation, the accumulation of triphosphates of deoxycytidine, 1-beta-D-arabinofuranosylcytosine, and 1-beta-D-arabinofuranosylguanine was potentiated in cells pretreated with dFdC. In contrast, the phosphorylation of 9-beta-D-arabinofuranosyladenine was not affected, since it is mainly phosphorylated by adenosine kinase, which is not influenced by deoxyribonucleoside triphosphates. Treatment of cells with dFdC followed by 1-beta-D-arabinofuranosylcytosine resulted in greater cytotoxicity than sum effects of each drug alone. The data indicate that an enhanced cytotoxicity could be obtained by administering dFdC as a modulator followed by 1-beta-D-arabinofuranosylcytosine or 1-beta-D-arabinofuranosylguanine in optimal sequence, suggesting that these results should be considered in the design of combination clinical protocols.


Subject(s)
Arabinonucleosides/metabolism , Deoxycytidine/analogs & derivatives , Leukemia/metabolism , Arabinonucleosides/pharmacology , Arabinonucleotides/metabolism , Arabinonucleotides/pharmacology , Cytarabine/metabolism , Cytarabine/pharmacology , Deoxycytidine/metabolism , Humans , Phosphorylation , Tumor Cells, Cultured/metabolism , Vidarabine/metabolism , Vidarabine/pharmacology , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...