Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
J Antimicrob Chemother ; 79(6): 1423-1431, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38708557

ABSTRACT

BACKGROUND: Bemnifosbuvir (AT-527) is a novel oral guanosine nucleotide antiviral drug for the treatment of persons with COVID-19. Direct assessment of drug disposition in the lungs, via bronchoalveolar lavage, is necessary to ensure antiviral drug levels at the primary site of SARS-CoV-2 infection are achieved. OBJECTIVES: This Phase 1 study in healthy subjects aimed to assess the bronchopulmonary pharmacokinetics, safety and tolerability of repeated doses of bemnifosbuvir. METHODS: A total of 24 subjects were assigned to receive bemnifosbuvir twice daily at doses of 275, 550 or 825 mg for up to 3.5 days. RESULTS: AT-511, the free base of bemnifosbuvir, was largely eliminated from the plasma within 6 h post dose in all dosing groups. Antiviral drug levels of bemnifosbuvir were consistently achieved in the lungs with bemnifosbuvir 550 mg twice daily. The mean level of the guanosine nucleoside metabolite AT-273, the surrogate of the active triphosphate metabolite of the drug, measured in the epithelial lining fluid of the lungs was 0.62 µM at 4-5 h post dose. This exceeded the target in vitro 90% effective concentration (EC90) of 0.5 µM for antiviral drug exposure against SARS-CoV-2 replication in human airway epithelial cells. Bemnifosbuvir was well tolerated across all doses tested, and most treatment-emergent adverse events reported were mild in severity and resolved. CONCLUSIONS: The favourable pharmacokinetics and safety profile of bemnifosbuvir demonstrates its potential as an oral antiviral treatment for COVID-19, with 550 mg bemnifosbuvir twice daily currently under further clinical evaluation in persons with COVID-19.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , Prodrugs , SARS-CoV-2 , Humans , Antiviral Agents/pharmacokinetics , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Male , Adult , Prodrugs/pharmacokinetics , Prodrugs/administration & dosage , Female , SARS-CoV-2/drug effects , Middle Aged , Administration, Oral , COVID-19 , Young Adult , Lung/drug effects , Lung/metabolism , Lung/virology , Healthy Volunteers , Guanosine/analogs & derivatives , Guanosine/pharmacokinetics , Guanosine/administration & dosage
2.
J Inorg Biochem ; 226: 111660, 2022 01.
Article in English | MEDLINE | ID: mdl-34801970

ABSTRACT

Nucleos(t)ide analogues (NA) belong to a family of compounds widely used in anticancer/antiviral treatments. They generally exhibit a cell toxicity limited by cellular uptake levels and the resulting nucleos(t)ides metabolism modifications, interfering with the cell machinery for nucleic acids synthesis. We previously synthesized purine nucleos(t)ide analogues N7-coordinated to a platinum centre with unaltered sugar moieties of the type: [Pt(dien)(N7-dGuo)]2+ (1; dien = diethylenetriamine; dGuo = 2'-deoxy-guanosine), [Pt(dien)(N7-dGMP)] (2; dGMP = 5'-(2'-deoxy)-guanosine monophosphate), and [Pt(dien)(N7-dGTP)]2- (3; dGTP = 5'-(2'-deoxy)-guanosine triphosphate), where the indicated electric charge is calculated at physiological pH (7.4). In this work, we specifically investigated the uptake of these complexes (1-3) at the plasma membrane level. Specific experiments on HeLa cervical cancer cells indicated a relevant cellular uptake of the model platinated deoxynucleos(t)ide 1 and 3 while complex 2 appeared unable to cross the cell plasma membrane. Obtained data buttress an uptake mechanism involving Na+-dependent concentrative transporters localized at the plasma membrane level. Consistently, 1 and 3 showed higher cytotoxicity with respect to complex 2 also suggesting selective possible applications as antiviral/antitumor drugs among the used model compounds.


Subject(s)
Cell Membrane/metabolism , Cytotoxins , Guanosine , Organoplatinum Compounds , Biological Transport , Cytotoxins/chemical synthesis , Cytotoxins/chemistry , Cytotoxins/pharmacokinetics , Cytotoxins/pharmacology , Guanosine/analogs & derivatives , Guanosine/chemistry , Guanosine/pharmacokinetics , Guanosine/pharmacology , HeLa Cells , Humans , Organoplatinum Compounds/chemical synthesis , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/pharmacokinetics , Organoplatinum Compounds/pharmacology
3.
PLoS One ; 15(1): e0227104, 2020.
Article in English | MEDLINE | ID: mdl-31914458

ABSTRACT

Despite the availability of highly effective direct-acting antiviral (DAA) regimens for the treatment of hepatitis C virus (HCV) infections, sustained viral response (SVR) rates remain suboptimal for difficult-to-treat patient populations such as those with HCV genotype 3, cirrhosis or prior treatment experience, warranting development of more potent HCV replication antivirals. AT-527 is the hemi-sulfate salt of AT-511, a novel phosphoramidate prodrug of 2'-fluoro-2'-C-methylguanosine-5'-monophosphate that has potent in vitro activity against HCV. The EC50 of AT-511, determined using HCV laboratory strains and clinical isolates with genotypes 1-5, ranged from 5-28 nM. The active 5'-triphosphate metabolite, AT-9010, specifically inhibited the HCV RNA-dependent RNA polymerase. AT-511 did not inhibit the replication of other selected RNA or DNA viruses in vitro. AT-511 was approximately 10-fold more active than sofosbuvir (SOF) against a panel of laboratory strains and clinical isolates of HCV genotypes 1-5 and remained fully active against S282T resistance-associated variants, with up to 58-fold more potency than SOF. In vitro, AT-511 did not inhibit human DNA polymerases or elicit cytotoxicity or mitochondrial toxicity at concentrations up to 100 µM. Unlike the other potent guanosine analogs PSI-938 and PSI-661, no mutagenic O6-alkylguanine bases were formed when incubated with cytochrome P450 (CYP) 3A4, and AT-511 had IC50 values ≥25 µM against a panel of CYP enzymes. In hepatocytes from multiple species, the active triphosphate was the predominant metabolite produced from the prodrug, with a half-life of 10 h in human hepatocytes. When given orally to rats and monkeys, AT-527 preferentially delivered high levels of AT-9010 in the liver in vivo. These favorable preclinical attributes support the ongoing clinical development of AT-527 and suggest that, when used in combination with an HCV DAA from a different class, AT-527 may increase SVR rates, especially for difficult-to-treat patient populations, and could potentially shorten treatment duration for all patients.


Subject(s)
Antiviral Agents/pharmacology , Guanosine/pharmacology , Hepacivirus/drug effects , Hepatitis C/drug therapy , Prodrugs/pharmacology , Animals , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Antiviral Agents/pharmacokinetics , Cell Line , Drug Discovery , Drug Evaluation, Preclinical , Female , Guanosine/analogs & derivatives , Guanosine/metabolism , Guanosine/pharmacokinetics , Haplorhini , Hepacivirus/genetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Male , Mice , Prodrugs/chemistry , Prodrugs/metabolism , Prodrugs/pharmacokinetics , Rats
4.
Purinergic Signal ; 12(1): 149-59, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26695181

ABSTRACT

In addition to its intracellular roles, the nucleoside guanosine (GUO) also has extracellular effects that identify it as a putative neuromodulator signaling molecule in the central nervous system. Indeed, GUO can modulate glutamatergic neurotransmission, and it can promote neuroprotective effects in animal models involving glutamate neurotoxicity, which is the case in brain ischemia. In the present study, we aimed to investigate a new in vivo GUO administration route (intranasal, IN) to determine putative improvement of GUO neuroprotective effects against an experimental model of permanent focal cerebral ischemia. Initially, we demonstrated that IN [(3)H] GUO administration reached the brain in a dose-dependent and saturable pattern in as few as 5 min, presenting a higher cerebrospinal GUO level compared with systemic administration. IN GUO treatment started immediately or even 3 h after ischemia onset prevented behavior impairment. The behavior recovery was not correlated to decreased brain infarct volume, but it was correlated to reduced mitochondrial dysfunction in the penumbra area. Therefore, we showed that the IN route is an efficient way to promptly deliver GUO to the CNS and that IN GUO treatment prevented behavioral and brain impairment caused by ischemia in a therapeutically wide time window.


Subject(s)
Brain Ischemia/drug therapy , Guanosine/administration & dosage , Guanosine/therapeutic use , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Stroke/drug therapy , Administration, Intranasal , Animals , Behavior, Animal , Brain Ischemia/psychology , Cerebral Infarction/pathology , Cerebral Infarction/prevention & control , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Guanosine/cerebrospinal fluid , Guanosine/pharmacokinetics , Male , Mitochondria/drug effects , Neuroprotective Agents/cerebrospinal fluid , Neuroprotective Agents/pharmacokinetics , Rats , Rats, Wistar , Stroke/psychology
5.
Eur J Drug Metab Pharmacokinet ; 41(5): 567-74, 2016 Oct.
Article in English | MEDLINE | ID: mdl-25898809

ABSTRACT

IDX184 is a phosphoramidate prodrug of 2'-methylguanosine-5'-monophosphate, developed to treat patients infected with hepatitis C virus. A mass balance study of radiolabeled IDX184 and pharmacokinetic studies of IDX184 in portal vein-cannulated monkeys revealed relatively low IDX184 absorption but higher exposure of IDX184 in the portal vein than in the systemic circulation, indicating >90 % of the absorbed dose was subject to hepatic extraction. Systemic exposures to the main metabolite, 2'-methylguanosine (2'-MeG), were used as a surrogate for liver levels of the pharmacologically active entity 2'-MeG triphosphate, and accordingly, systemic levels of 2'-MeG in the monkey were used to optimize formulations for further clinical development of IDX184. Capsule formulations of IDX184 delivered acceptable levels of 2'-MeG in humans; however, the encapsulation process introduced low levels of the genotoxic impurity ethylene sulfide (ES), which necessitated formulation optimization. Animal pharmacokinetic data guided the development of a tablet with trace levels of ES and pharmacokinetic performance equal to that of the clinical capsule in the monkey. Under fed conditions in humans, the new tablet formulation showed similar exposure to the capsule used in prior clinical trials.


Subject(s)
Guanosine Monophosphate/analogs & derivatives , Guanosine/analogs & derivatives , Liver/drug effects , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Animals , Capsules/administration & dosage , Capsules/pharmacokinetics , Chemistry, Pharmaceutical/methods , Guanosine/administration & dosage , Guanosine/pharmacokinetics , Guanosine Monophosphate/administration & dosage , Guanosine Monophosphate/pharmacokinetics , Haplorhini , Humans , Male , Tablets/administration & dosage , Tablets/pharmacokinetics
6.
Biol Pharm Bull ; 38(3): 380-8, 2015.
Article in English | MEDLINE | ID: mdl-25757919

ABSTRACT

A conventional, rapid and high throughput method for tissue extraction and accurate and selective LC-MS/MS quantification of 2'-C-methylguanosine triphosphate (2'-MeGTP) in mouse liver was developed and qualified. Trichloroacetic acid (TCA) was used as the tissue homogenization reagent that overcomes instability challenges of liver tissue nucleotide triphosphates due to instant ischemic degradation to mono- and diphosphate nucleotides. Degradation of 2'-MeGTP was also minimized by harvesting livers using in situ clamp-freezing or snap-freezing techniques. The assay also included a sample clean-up procedure using weak anion exchange solid phase extraction followed by ion exchange chromatography and tandem mass spectrometry detection. The linear assay range was from 50 to 10000 pmol/mL concentration in liver homogenate (250-50000 pmol/g in liver tissue). The method was qualified over three intraday batches for accuracy, precision, selectivity and specificity. The assay was successfully applied to pharmacokinetic studies of 2'-MeGTP in liver tissue samples after single oral doses of IDX184, a nucleotide prodrug inhibitor of the viral polymerase for the treatment of hepatitis C, to mice. The study results suggested that the clamp-freezing liver collection method was marginally more effective in preventing 2'-MeGTP degradation during liver tissue collection compared to the snap-freezing method.


Subject(s)
Guanosine Monophosphate/analogs & derivatives , Guanosine Triphosphate/metabolism , Guanosine/analogs & derivatives , Liver/metabolism , Nucleotides/metabolism , Prodrugs/metabolism , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacokinetics , Chromatography, Ion Exchange/methods , Chromatography, Liquid/methods , Freezing , Guanosine/metabolism , Guanosine/pharmacokinetics , Guanosine Monophosphate/metabolism , Guanosine Monophosphate/pharmacokinetics , Guanosine Triphosphate/analogs & derivatives , Hepatitis C/drug therapy , Male , Mice , Prodrugs/pharmacokinetics , Solid Phase Extraction/methods , Tandem Mass Spectrometry/methods , Trichloroacetic Acid/chemistry
7.
J Biol Regul Homeost Agents ; 26(1): 51-65, 2012.
Article in English | MEDLINE | ID: mdl-22475097

ABSTRACT

Guanosine has long been known as an endogenous purine nucleoside deeply involved in the modulation of several intracellular processes, especially G-protein activity. More recently, it has been reported to act as an extracellular signaling molecule released from neurons and, more markedly, from astrocytes either in basal conditions or after different kinds of stimulation including hypoxia. Moreover, in vivo studies have shown that guanosine plays an important role as both a neuroprotective and neurotrophic agent in the central nervous system. Specific high-affinity binding sites for this nucleoside have been found on membrane preparations from rat brain. The present study was undertaken to investigate the distribution and metabolic profiles of guanosine after administering the nucleoside to gain a better understanding of the biological effects of this potential drug candidate. Rats were given an intraperitonal (i.p.) injection of 2, 4, 8 or 16 mg/kg of guanosine combined with 0.05% of [3H]guanosine. Plasma samples were collected 7.5, 15, 30, 60 and 90 min after the guanosine-mixture administration and analyzed by either a liquid scintillation counter or by HPLC connected to a UV and to an on-line radiochemical detector to measure the levels of guanosine and its metabolic products guanine, xanthine and uric acid. The levels of guanosine, guanine and xanthine were also measured in brain, lung, heart, kidney and liver tissue homogenates at the defined time points after the injection of 8 mg/kg of the guanosine-mixture. We found that the levels of radioactivity in plasma increased linearly in a dose- and time-dependent manner. Guanosine was widely distributed in all tissues examined in the present study, at almost twice its usual levels. In addition, guanine levels dramatically increased in all the organs. Interestingly, enzymatic analysis of the plasma samples showed the presence of a soluble purine nucleoside phosphorylase, a key enzyme in the purine salvage pathway and nucleoside catabolism. Since guanosine has been shown to be neuroprotective and astrocytes have been reported to play critical roles in mediating neuronal survival and functions in different neurodegenerative disorders, we also performed uptake and release.


Subject(s)
Guanosine/pharmacokinetics , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Brain/cytology , Brain/metabolism , Cells, Cultured , Guanine/metabolism , Guanosine/administration & dosage , Guanosine/blood , Injections, Intraperitoneal , Kidney/metabolism , Liver/metabolism , Lung/metabolism , Male , Myocardium/metabolism , Purine-Nucleoside Phosphorylase/blood , Purines/metabolism , Rats , Rats, Sprague-Dawley , Xanthine/metabolism
8.
J Med Chem ; 54(24): 8632-45, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22039920

ABSTRACT

We herein report phosphorodiamidates as a significant new phosphate prodrug motif. Sixty-seven phosphorodiamidates are reported of two 6-O-alkyl 2'-C-methyl guanosines, with significant variation in the diamidate structure. Both symmetrical and asymmetric phosphorodiamidates are reported, derived from various esterified amino acids, both d and l, and also from various simple amines. All of the compounds were evaluated versus hepatitis C virus in replicon assay, and nanomolar activity levels were observed. Many compounds were noncytotoxic at 100 µM, leading to high antiviral selectivities. The agents are stable in acidic, neutral, and moderately basic media and in selected biological media but show efficient processing by carboxypeptidases and efficiently yield the free nucleoside monophosphate in cells. On the basis of in vitro data, eight leads were selected for additional in vivo evaluation, with the intent of selecting one candidate for progression toward clinical studies. This phosphorodiamidate prodrug method may have broad application outside of HCV and antivirals as it offers many of the advantages of phosphoramidate ProTides but without the chirality issues present in most cases.


Subject(s)
Antiviral Agents/chemical synthesis , Guanosine/analogs & derivatives , Guanosine/chemical synthesis , Hepacivirus/drug effects , Organophosphorus Compounds/chemical synthesis , Prodrugs/chemical synthesis , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Cathepsin A/metabolism , Cell Line , Drug Stability , Guanosine/pharmacokinetics , Guanosine/pharmacology , Hepacivirus/genetics , Humans , Liver/metabolism , Male , Models, Molecular , Organophosphorus Compounds/pharmacokinetics , Organophosphorus Compounds/pharmacology , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Rats , Rats, Sprague-Dawley , Serum , Structure-Activity Relationship
9.
Antimicrob Agents Chemother ; 55(5): 1843-51, 2011 May.
Article in English | MEDLINE | ID: mdl-21357300

ABSTRACT

INX-08189 is an aryl-phosphoramidate of 6-O-methyl-2'-C-methyl guanosine. INX-08189 was highly potent in replicon assays, with a 50% effective concentration of 10±6 nM against hepatitis C genotype 1b at 72 h. The inhibitory effect on viral replication was rapid, with a 50% effective concentration (EC50) of 35±8 nM at 24 h. An intracellular 2'-C-methyl guanosine triphosphate (2'-C-MeGTP) concentration of 2.43±0.42 pmol/10(6) cells was sufficient to achieve 90% inhibition of viral replication. In vitro resistance studies confirmed that the S282T mutation in the NS5b gene conferred an approximately 10-fold reduction in sensitivity to INX-08189. However, the complete inhibition of S282T mutant replicons still could be achieved with an EC90 of 344±170 nM. Drug combination studies of INX-08189 and ribavirin indicated significant synergy in antiviral potency both in wild-type and S282T-expressing replicons. Genotype 1b replicons could be cleared after 14 days of culture when exposed to as little as 20 nM INX-08189. No evidence of mitochondrial toxicity was observed after 14 days of INX-08189 exposure in both HepG2 and CEM human cell lines. In vivo studies of rats and cynomolgus monkeys demonstrated that 2'-C-MeGTP concentrations in liver equivalent to the EC90 could be attained after a single oral dose of INX-08189. Rat liver 2'-C-MeGTP concentrations were proportional to dose, sustained for greater than 24 h, and correlated with plasma concentrations of the nucleoside metabolite 2'-C-methyl guanosine. The characteristics displayed by INX-08189 support its continued development as a clinical candidate for the treatment of chronic HCV infection.


Subject(s)
Amides/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/pharmacokinetics , Guanosine/pharmacology , Guanosine/pharmacokinetics , Hepacivirus/drug effects , Phosphoric Acids/chemistry , Prodrugs/pharmacology , Prodrugs/pharmacokinetics , Animals , Cell Line , Cell Line, Tumor , Guanosine/analogs & derivatives , Guanosine/chemistry , Humans , Macaca fascicularis , Male , Prodrugs/chemistry , Rats , Rats, Sprague-Dawley , Virus Replication/drug effects
10.
Antimicrob Agents Chemother ; 53(1): 150-6, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19001108

ABSTRACT

Accumulation of antiviral nucleotides in renal proximal tubules is controlled by their basolateral uptake via the human renal organic anion transporters type 1 (hOAT1) and 3 (hOAT3) and apical efflux via the multidrug resistance protein 4 (MRP4). GS-9148 is a novel ribose-modified nucleotide human immunodeficiency virus (HIV) reverse transcriptase inhibitor, and its oral prodrug GS-9131 is currently being evaluated in the clinic as an anti-HIV agent. To assess the potential of GS-9148 for nephrotoxicity, its mechanism of renal transport, cytotoxicity, and renal accumulation were explored in vitro and in vivo. In comparison with the acyclic nucleotides cidofovir, adefovir, and tenofovir, GS-9148 showed 60- to 100-fold lower efficiency of transport (V(max)/K(m)) by hOAT1 and was 20- to 300-fold less cytotoxic in cells overexpressing hOAT1, indicating its lower hOAT1-mediated intracellular accumulation and reduced intrinsic cytotoxicity. GS-9148 was also relatively inefficiently transported by hOAT3. Similar to acyclic nucleotides, GS-9148 was a substrate for MRP4 as evidenced by its reduced intracellular retention in cells overexpressing the efflux pump. Consistent with these molecular observations, GS-9148 was inefficiently taken up by fresh human renal cortex tissue in vitro and showed a limited accumulation in kidneys in vivo following oral administration of [(14)C]GS-9131 to dogs. Compared to acyclic nucleotide analogs, GS-9148 was also found to have lower net active tubular secretion in dogs. Collectively, these results suggest that GS-9148 exhibits a low potential for renal accumulation and nephrotoxicity.


Subject(s)
Guanosine/analogs & derivatives , HIV Reverse Transcriptase/antagonists & inhibitors , Kidney/drug effects , Kidney/metabolism , Reverse Transcriptase Inhibitors/pharmacology , Reverse Transcriptase Inhibitors/pharmacokinetics , Adenine/analogs & derivatives , Adenine/chemistry , Adenine/pharmacokinetics , Adenine/pharmacology , Animals , CHO Cells , Cell Line , Cell Survival/drug effects , Cidofovir , Cricetinae , Cricetulus , Cytosine/analogs & derivatives , Cytosine/chemistry , Cytosine/pharmacokinetics , Cytosine/pharmacology , Guanosine/chemistry , Guanosine/pharmacokinetics , Guanosine/pharmacology , Humans , In Vitro Techniques , Kidney/cytology , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Organophosphonates/chemistry , Organophosphonates/pharmacokinetics , Organophosphonates/pharmacology , Reverse Transcriptase Inhibitors/chemistry , Tenofovir
11.
Arch Pharm Res ; 31(10): 1347-53, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18958427

ABSTRACT

A 1:1 mixture of acriflavine (ACF; CAS 8063-24-9) and guanosine is under evaluation in preclinical studies as a possible antitumor agent. Guanosine is known to potentiate the anti-cancer activity of ACF. We therefore investigated the pharmacokinetics of guanosine following administration of the ACF/guanosine mixture in rats. Rats were given guanosine (1 or 5 mg/kg) or ACF/guanosine (2 or 10 mg/kg) by i.v. bolus; or guanosine (3 or 15 mg/kg) or ACF/guanosine (6 or 30 mg/kg) by i.m. injection. We found that guanosine was rapidly cleared from the blood and transferred to tissues after i.m. administration of ACF/guanosine. The mean plasma half-lives (t(1/2)) at the alpha and beta phases were 0.091 and 6.86 h, or 0.09 and 7.51 h at a dose of 1 or 5 mg/kg guanosine, respectively. ACF had no effect on the plasma disappearance of guanosine following either i.v. bolus or i.m. administration of the combination mixture. Moreover, the ACF combination with guanosine did not significantly alter the values of MRT, V(dss), and CL(t) of guanosine. Guanosine exhibited linear pharmacokinetics over the dose range from 1 to 5 mg/kg for i.v. doses and 3 to 15 mg/kg for i.m. doses. The bioavailability of guanosine after i.m. administration was 84% for 3 mg/kg dose and 88% for 15 mg/kg dose. ACF had no effects on biliary and urinary excretion of guanosine after i.m. administration. The cumulative amount of guanosine in urine after i.m. administration was about 5-fold larger than that in bile, indicating that guanosine is mostly excreted into the urine. Guanosine was widely distributed in all tissues examined in this study, but was most highly concentrated in the kidney after i.m. administration, followed by slow excretion to bile or urine. ACF had no effect on the tissue distribution of guanosine following i.m. administration. These characterizations of the pharmacokinetics of guanosine after administration of the ACF/guanosine combination will be useful in providing preclinical and clinical bases for the potential application of this combination to the treatment of cancer.


Subject(s)
Acriflavine , Antineoplastic Agents , Guanosine/pharmacokinetics , Animals , Area Under Curve , Drug Combinations , Guanosine/administration & dosage , Indicators and Reagents , Injections, Intramuscular , Injections, Intravenous , Male , Rats , Rats, Sprague-Dawley , Tissue Distribution
12.
Nucleosides Nucleotides Nucleic Acids ; 27(6): 673-80, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18600525

ABSTRACT

Intraperitoneal administration of guanosine to rats with chronic spinal cord injury stimulates remyelination and functional recovery. If guanosine produced its effects in the nervous system, it should enter it and elevate endogenous concentrations. [(3)H]-guanosine (8 mg/kg) was administered intraperitoneally to rats and its distribution and concentration in different sites determined. Guanosine rapidly entered all tissues; its concentration peaked at about 15 minutes except in adipose tissue and CNS where it continued to rise for 30 minutes. Its chief metabolic product in all sites was guanine with over twice as much guanine as guanosine present in CNS after 30 minutes.


Subject(s)
Guanosine/metabolism , Guanosine/pharmacokinetics , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Animals , Cell Membrane/metabolism , Female , Guanine/blood , Guanine/metabolism , Guanine/pharmacokinetics , Guanosine/blood , Guanosine/therapeutic use , Injections, Intraperitoneal , Rats , Tissue Distribution , Xanthine/blood , Xanthine/metabolism , Xanthine/pharmacokinetics
13.
Am J Physiol Endocrinol Metab ; 290(6): E1124-30, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16390860

ABSTRACT

Glucose transport is regarded as the principal rate control step governing insulin-stimulated glucose utilization by skeletal muscle. To assess this step in human skeletal muscle, quantitative PET imaging of skeletal muscle was performed using 3-O-methyl-[11C]glucose (3-[11C]OMG) in healthy volunteers during a two-step insulin infusion [n = 8; 30 and 120 mU.min(-1).m(-2), low (LO) and high (HI)] and during basal conditions (n = 8). Positron emission tomography images were coregistered with MRI to assess 3-[11C]OMG activity in regions of interest placed on oxidative (soleus) compared with glycolytic (tibialis anterior) muscle. Insulin dose-responsive increases of 3-[11C]OMG activity in muscle were observed (P < 0.01). Tissue activity was greater in soleus than in tibialis anterior (P < 0.05). Spectral analysis identified that two mathematical components interacted to shape tissue activity curves. These two components were interpreted physiologically as likely representing the kinetics of 3-[11C]OMG delivery from plasma to tissue and the kinetics of bidirectional glucose transport. During low compared with basal, there was a sixfold increase in k3, the rate constant attributed to inward glucose transport, and another threefold increase during HI (0.012 +/- 0.003, 0.070 +/- 0.014, 0.272 +/- 0.059 min(-1), P < 0.001). Values for k3 were similar in soleus and tibialis anterior, suggesting similar kinetics for transport, but compartmental modeling indicated a higher value in soleus for k1, denoting higher rates of 3-[11C]OMG delivery to soleus than to tibialis anterior. In summary, in healthy volunteers there is robust dose-responsive insulin stimulation of glucose transport in skeletal muscle.


Subject(s)
Blood Glucose/metabolism , Guanosine/analogs & derivatives , Insulin/administration & dosage , Insulin/blood , Muscle, Skeletal/metabolism , Biological Transport , Dose-Response Relationship, Drug , Fatty Acids, Nonesterified/blood , Glucose Clamp Technique , Guanosine/pharmacokinetics , Humans , Magnetic Resonance Imaging , Muscle, Skeletal/physiology , Positron-Emission Tomography , Time Factors
14.
Xenobiotica ; 35(7): 755-73, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16323364

ABSTRACT

Acriflavine (ACF; CAS 8063-24-9), a mixture of trypaflavine (TRF) and proflavine (PRF) at a ratio of 2:1 is being investigated in rodents as an anticancer agent. However, its pharmacokinetics have not been investigated in mammals. Guanosine is known to potentiate the anticancer activity of some compounds. The pharmacokinetics of AG60, a 1:1 mixture of ACF and guanosine, were therefore investigated in rats. Rats were given 2 or 10 mg kg(-1) AG60 by intravenous bolus or 6 or 30 mg kg (-1) intramuscularly. An HPLC-based method was developed to analyse the levels of TRF, PRF, and their metabolites in plasma, bile, urine and tissue homogenates. The plasma concentrations of TRF and PRF decreased rapidly after intravenous administration and more slowly after intramuscular administration. Both TRF and PRF were distributed widely, most notably in the kidney, and were eliminated slowly. Three glucuronosyl conjugate metabolite peaks were tentatively identified in the bile. The intramuscular route leads to a prolongation of TRF or PRF plasma levels, and the systemic exposures for both TRF and PRF were both relatively high. These observations indicate that the intramuscular route may be the best way to administer AG60 for various clinical applications.


Subject(s)
Acriflavine/pharmacokinetics , Anti-Infective Agents, Local/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Guanosine/pharmacokinetics , Acriflavine/administration & dosage , Acriflavine/metabolism , Animals , Anti-Infective Agents, Local/administration & dosage , Anti-Infective Agents, Local/metabolism , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , Guanosine/administration & dosage , Guanosine/metabolism , Injections, Intramuscular/methods , Injections, Intravenous/methods , Male , Rats , Rats, Sprague-Dawley
15.
J Clin Endocrinol Metab ; 90(3): 1752-9, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15613423

ABSTRACT

Insulin-stimulated glucose transport in skeletal muscle is regarded as a key determinant of insulin sensitivity, yet isolation of this step for quantification in human studies is a methodological challenge. One notable approach is physiological modeling of dynamic positron emission tomography (PET) imaging using 2-[18-fluoro]2-deoxyglucose ([(18)F]FDG); however, this has a potential limitation in that deoxyglucose undergoes phosphorylation subsequent to transport, complicating separate estimations of these steps. In the current study we explored the use of dynamic PET imaging of [(11)C]3-O-methylglucose ([(11)C]3-OMG), a glucose analog that is limited to bidirectional glucose transport. Seventeen lean healthy volunteers with normal insulin sensitivity participated; eight had imaging during basal conditions, and nine had imaging during euglycemic insulin infusion at 30 mU/min.m(2). Dynamic PET imaging of calf muscles was conducted for 90 min after the injection of [(11)C]3-OMG. Spectral analysis of tissue activity indicated that a model configuration of two reversible compartments gave the strongest statistical fit to the kinetic pattern. Accordingly, and consistent with the structure of a model previously used for [(18)F]FDG, a two-compartment model was applied. Consistent with prior [(18)F]FDG findings, insulin was found to have minimal effect on the rate constant for movement of [(11)C]3-OMG from plasma to tissue interstitium. However, during insulin infusion, a robust and highly significant increase was observed in the kinetics of inward glucose transport; this and the estimated tissue distribution volume for [(11)C]3-OMG increased 6-fold compared with basal conditions. We conclude that dynamic PET imaging of [(11)C]3-OMG offers a novel quantitative approach that is both chemically specific and tissue specific for in vivo assessment of glucose transport in human skeletal muscle.


Subject(s)
Glucose/metabolism , Guanosine/analogs & derivatives , Guanosine/pharmacokinetics , Monosaccharide Transport Proteins/metabolism , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/metabolism , Positron-Emission Tomography/methods , Adult , Carbon Radioisotopes , Female , Glucose Clamp Technique , Humans , Male , Models, Biological
16.
Pharm Res ; 21(6): 982-8, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15212163

ABSTRACT

PURPOSE: To investigate the effects of potential inhibitors of membrane transport on the tubular secretion of AM188, an antiviral guanosine analog, in the isolated perfused rat kidney (IPK). METHODS: AM188 was administered to the IPK perfusate as a bolus/infusion regimen. In inhibitor groups, probenecid, p-aminohippuric acid (PAH), cimetidine, or nitrobenzylthioinosine was added to the perfusing medium. RESULTS: In control IPKs, the ratio of renal clearance of AM188 (CLR) to GFR was 7.7 +/- 0.51 (mean +/- SD). The CL(R)/GFR ratio for AM188 was 6.20 +/- 0.41*, 2.85 +/- 0.20*, 1.45 +/- 0.07*, and 0.80 +/- 0.01* when the concentration of probenecid in perfusate was 10, 50, 100, and 1000 microM, respectively (*p < 0.05 compared to control group); the ratio was 7.71 +/- 0.38, 6.02 +/- 0.42*, 1.71 +/- 0.15*, and 0.91 +/- 0.07* for the PAH group and 6.42 +/- 1.70*, 5.33 +/- 1.53*, 3.16 +/- 0.81*, and 1.21 +/- 0.20* for the cimetidine group when the concentrations were 10, 100, 1000 and 10,000 microM, respectively; and the ratio was 5.33 +/- 0.21* when the concentration of nitrobenzylthioinosine was 5 microM. CONCLUSIONS: These results suggest that renal tubular secretion of AM188 involves organic anion and cation transport systems.


Subject(s)
Cimetidine/pharmacology , Guanosine/analogs & derivatives , Guanosine/antagonists & inhibitors , Guanosine/metabolism , Nephrons/drug effects , Nephrons/metabolism , Probenecid/pharmacology , Thioinosine/analogs & derivatives , p-Aminohippuric Acid/pharmacology , Animals , Antiviral Agents/antagonists & inhibitors , Antiviral Agents/metabolism , Antiviral Agents/pharmacokinetics , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/physiology , Guanosine/pharmacokinetics , Male , Metabolic Clearance Rate/drug effects , Metabolic Clearance Rate/physiology , Perfusion/methods , Rats , Thioinosine/pharmacology , Tissue Distribution/drug effects
17.
Am J Physiol Cell Physiol ; 286(4): C792-7, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14644775

ABSTRACT

Initial rates of glucose entry into isolated bovine mammary epithelial cells display moderate degrees of asymmetry and cooperative interactions between export and import sites. The present study examined the hypothesis that these kinetic features are due to compartmentalization of intracellular glucose. Net uptake of 3-O-methyl-d-[1-(3)H]glucose (3-OMG) by isolated bovine mammary epithelial cells was measured at 37 degrees C. The time course of 3-OMG net uptake was better fitted by a double-exponential equation than by a single- or triple-exponential equation. Compartmental analysis of the time course curve suggested that translocated 3-OMG is distributed into two compartments with fractional volumes of 32.6 +/- 5.7% and 67.4 +/- 5.7%, respectively. The results support the view that glucose transport in bovine mammary epithelial cells is a multistep process consisting of two serial steps: fast, carrier-mediated, symmetric translocation of sugar across the cell plasma membrane into a small compartment and subsequent slow exchange of posttranslocated sugar between two intracellular compartments. A three-compartment model of this system successfully simulated the observed time course of 3-OMG net uptake and the observed dependence of unidirectional entry rates on intra- and extracellular 3-OMG concentrations. Simulations indicated that backflux of radiolabeled sugar from the small compartment to extracellular space during 15 s of incubation gives rise to the apparent asymmetry, trans-stimulation, and cooperativity of mammary glucose transport kinetics. The fixed-site carrier model overestimated the rate of glucose accumulation in cells, and its features can be accounted for by the compartmentalization of intracellular sugar.


Subject(s)
Cell Compartmentation/physiology , Epithelial Cells/metabolism , Glucose/metabolism , Guanosine/analogs & derivatives , Mammary Glands, Animal/cytology , Models, Biological , Animals , Cattle , Epithelial Cells/cytology , Female , Guanosine/pharmacokinetics
18.
Clin Exp Pharmacol Physiol ; 30(12): 943-50, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14678234

ABSTRACT

1. We tested the hypothesis that the cGMP-dependent protein kinase has major negative functional effects in cardiac myocytes and that the importance of this pathway is reduced in thyroxine (T4; 0.5 mg/kg per day for 16 days) hypertrophic myocytes. 2. Using isolated ventricular myocytes from control (n = 7) and T4-treated (n = 9) rabbit hypertrophic hearts, myocyte shortening was studied with a video edge detector. Oxygen consumption was measured using O2 electrodes. Protein phosphorylation was measured autoradiographically. 3. Data were collected following treatment with: (i) 8-(4-chlorophenylthio)guanosine-3',5'-monophosphate (PCPT; 10-7 or 10-5 mol/L); (ii) 8-bromo-cAMP (10-5 mol/L) followed by PCPT; (iii) beta-phenyl-1,N2-etheno-8-bromoguanosine-3',5'-monophosphorothioate, SP-isomer (SP; 10-7 or 10-5 mol/L); or (iv) 8-bromo-cAMP (10-5 mol/L) followed by SP. 4. There were no significant differences between groups in baseline percentage shortening (Pcs; 4.9 +/- 0.2 vs 5.6 +/- 0.4% for control and T4 groups, respectively) and maximal rate of shortening (Rs; 64.8 +/- 5.9 vs 79.9 +/- 7.1 micro m/ s for control and T4 groups, respectively). Both SP and PCPT decreased Pcs (-43 vs-21% for control and T4 groups, respectively) and Rs (-36 vs-22% for control and T4 groups, respectively), but the effect was significantly reduced in T4 myocytes. 8-Bromo-cAMP similarly increased Pcs (28 vs 23% for control and T4 groups, respectively) and Rs (20 vs 19% for control and T4 groups, respectively). After 8-bromo-cAMP, SP and PCPT decreased Pcs (-34%) and Rs (-29%) less in the control group. However, the effects of these drugs were not altered in T4 myocytes (Pcs -24%; Rs -22%). Both PCPT and cAMP phosphorylated the same five protein bands. In T4 myocytes, these five bands were enhanced less. 5. We conclude that, in control ventricular myocytes, the cGMP-dependent protein kinase exerted major negative functional effects but, in T4-induced hypertrophic myocytes, the importance of this pathway was reduced and the interaction between cAMP and the cGMP protein kinase was diminished.


Subject(s)
Cyclic AMP/analogs & derivatives , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/metabolism , Guanosine/analogs & derivatives , Hypertrophy/chemically induced , Thyroxine/adverse effects , gamma-Aminobutyric Acid/analogs & derivatives , 8-Bromo Cyclic Adenosine Monophosphate , Animals , Body Weight/drug effects , Cyclic AMP/adverse effects , Cyclic AMP/pharmacokinetics , Cyclic GMP/metabolism , Drug Administration Schedule , Drug Synergism , Guanosine/adverse effects , Guanosine/pharmacokinetics , Heart Ventricles/cytology , Heart Ventricles/drug effects , Heart Ventricles/pathology , Hypertrophy/enzymology , Injections, Intramuscular , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Myocytes, Cardiac/chemistry , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Organ Size/drug effects , Oxygen Consumption/drug effects , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Phosphorylation/drug effects , Rabbits , Stimulation, Chemical , Thyroxine/administration & dosage , Time Factors
19.
J Appl Physiol (1985) ; 94(4): 1373-9, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12496137

ABSTRACT

Physical activity is known to increase insulin action in skeletal muscle, and data have indicated that 5'-AMP-activated protein kinase (AMPK) is involved in the molecular mechanisms behind this beneficial effect. 5-Aminoimidazole-4-carboxamide-1-beta-d-ribofuranoside (AICAR) can be used as a pharmacological tool to repetitively activate AMPK, and the objective of this study was to explore whether the increase in insulin-stimulated glucose uptake after either long-term exercise or chronic AICAR administration was followed by fiber-type-specific changes in insulin signaling and/or changes in GLUT-4 expression. Wistar rats were allocated into three groups: an exercise group trained on treadmill for 5 days, an AICAR group exposed to daily subcutaneous injections of AICAR, and a sedentary control group. AMPK activity, insulin-stimulated glucose transport, insulin signaling, and GLUT-4 expression were determined in muscles characterized by different fiber type compositions. Both exercised and AICAR-injected animals displayed a fiber-type-specific increase in glucose transport with the most marked increase in muscles with a high content of type IIb fibers. This increase was accompanied by a concomitant increase in GLUT-4 expression. Insulin signaling as assessed by phosphatidylinositol 3-kinase and PKB/Akt activity was enhanced only after AICAR administration and in a non-fiber-type-specific manner. In conclusion, chronic AICAR administration and long-term exercise both improve insulin-stimulated glucose transport in skeletal muscle in a fiber-type-specific way, and this is associated with an increase in GLUT-4 content.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Glucose/pharmacokinetics , Guanosine/analogs & derivatives , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Monosaccharide Transport Proteins/metabolism , Motor Activity/physiology , Muscle Proteins , Muscle, Skeletal/metabolism , Ribonucleotides/pharmacology , Signal Transduction/drug effects , AMP-Activated Protein Kinases , Animals , Biological Transport , Glucose Transporter Type 4 , Guanosine/pharmacokinetics , Immunoblotting , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Male , Multienzyme Complexes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Wistar
20.
J Dairy Sci ; 85(9): 2308-14, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12362464

ABSTRACT

In previous work, a comparatively high capacity for Na(+)-dependent transport of nucleosides across the intestinal brush border membrane (BBM) was observed in dairy cows, which might be related to digestion of the large amount of nucleic acids present in ruminal microorganisms in the ruminant small intestine. If this were the case, the capacity for Na(+)-dependent intestinal nucleoside transport should be much lower in veal calves, in which only small amounts of nucleic acids, nucleotides, and nucleosides reach the small intestine via the milk replacer. To test this hypothesis, we investigated Na(+)-dependent transport of 3H-labeled thymidine and guanosine across the BBM using BBM vesicles (BBMV) isolated from the small intestine of veal calves. In the presence of a transmembrane Na+ gradient both substrates were transported against a concentration gradient. Inhibitory studies showed that thymidine and guanosine are transported by two different transporters with overlapping substrate specificity, one accepting predominantly pyrimidine nucleosides (N2) and one accepting particularly purine nucleosides (N1). Nucleoside transport was inhibited by glucose along the whole small intestine. Maximal transport rates similar to those in dairy cows were obtained for the proximal, mid-, and distal small intestine. These findings suggest that the high absorptive capacity for nucleosides is a genetically fixed property in the bovine small intestine, which is already present in the preruminant state of veal calves. It may contribute to the high digestibility of nucleic acids observed by others in veal calves receiving milk replacer supplemented with RNA. Its main function may be the efficient absorption of nucleosides resulting from the digestion of nucleic acids associated with desquamated enterocytes. Due to the limited de novo synthesis of nucleotides in enterocytes intracellular uptake of nucleosides across the BBM may contribute to nucleic acid synthesis in enterocytes and thus may have a trophic effect on the intestinal epithelium.


Subject(s)
Cattle/metabolism , Intestinal Absorption , Intestine, Small/ultrastructure , Microvilli/metabolism , Nucleosides/pharmacokinetics , Sodium/pharmacology , Animals , Biological Transport/drug effects , Glucose/pharmacology , Guanosine/pharmacokinetics , Kinetics , Thymidine/pharmacokinetics , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL
...