Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Xenobiotica ; 46(4): 379-84, 2016.
Article in English | MEDLINE | ID: mdl-26338263

ABSTRACT

1. The extent of sulfoxidation of the drug, S-carboxymethyl-L-cysteine, has been shown to vary between individuals, with this phenomenon being mooted as a biomarker for certain disease states and susceptibilities. Studies in vitro have indicated that the enzyme responsible for this reaction was phenylalanine monooxygenase but to date no in vivo evidence exists to support this assumption. Using the mouse models of mild hyperphenylalaninamia (enu1 PAH variant) and classical phenylketonuria (enu2 PAH variant), the sulfur oxygenation of S-carboxymethyl-L-cysteine has been investigated. 2. Compared to the wild type (wt/wt) mice, both the heterozygous dominant (wt/enu1 and wt/enu2) mice and the homozygous recessive (enu1/enu1 and enu2/enu2) mice were shown to have significantly increased Cmax, AUC(0-180 min) and AUC(0-∞ min) values (15 - to 20-fold higher). These results were primarily attributable to the significantly reduced clearance of S-carboxymethyl-L-cysteine (13 - to 22-fold lower). 3. Only the wild type mice produced measurable quantities of the parent S-oxide metabolites. Those mice possessing one or more allelic variant showed no evidence of blood SCMC (R/S) S-oxides. These observations support the proposition that differences in phenylalanine hydroxylase activity underlie the variation in S-carboxymethyl-L-cysteine sulfoxidation and that no other enzyme is able to undertake this reaction.


Subject(s)
Carbocysteine/metabolism , Oxygen/metabolism , Phenylalanine Hydroxylase/metabolism , Sulfur/metabolism , Animals , Carbocysteine/blood , Carbocysteine/pharmacokinetics , Female , Male , Mice , Time Factors
2.
Drug Metab Rev ; 44(2): 129-47, 2012 May.
Article in English | MEDLINE | ID: mdl-22497630

ABSTRACT

S-carboxymethyl-L-cysteine, the side-chain carboxymethyl derivative of the sulfur-containing amino acid, cysteine, has been known and available for almost 80 years. During this time, it has been put to a variety of uses, but it is within the field of respiratory medicine that, presently, it has found a clinical niche. Early studies indicated that this compound underwent a rather simplistic, predictable pattern of metabolism, whereas later investigations alluded to more subtle interactions with the pathways of intermediary metabolism, as may be expected for an amino acid derivative. In addition, suggestions of polymorphic influences and circadian rhythms within metabolic profiles have emerged. These latter factors may underlie the conflicting reports regarding the therapeutic efficacy of this compound: that it appears to work well in some patients, but has no measurable effects in others. The relevant literature pertaining to the fate of this compound within living systems has been reviewed and a comprehensive précis advanced. Hopefully, this article will serve as a vade mecum for those interested in S-carboxymethyl-L-cysteine and as a catalyst for future research.


Subject(s)
Carbocysteine/pharmacokinetics , Acetylation , Animals , Carbocysteine/chemical synthesis , Carbocysteine/pharmacology , Carbocysteine/therapeutic use , Circadian Rhythm , Glucuronides/metabolism , Humans , Respiratory Tract Diseases/drug therapy , Respiratory Tract Diseases/metabolism , Sulfoxides/metabolism
3.
Eur J Pharm Sci ; 39(4): 219-23, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20035864

ABSTRACT

The aim of this study was to investigate the feasibility of employing S-carboxymethyl-L-cysteine as a treatment of chronic obstructive pulmonary disease in dogs. To this end the pharmacokinetic parameters of orally administered S-carboxymethyl-L-cysteine were determined in the dog, cow and sheep. Six healthy beagle dogs, six endogenous Greek sheep and four Holstein Fresian calves were orally dosed with 10 mg/kg body weight of S-carboxymethyl-L-cysteine. No significant differences in T(max) and T(1/2) were reported between the species. However, significantly higher AUC((0-last)), 21.56+/-6.67 microg h ml(-1) and AUC((0-infinity)), 21.63+/-6.68 microg h ml(-1) were seen in the dogs compared to the sheep and calves. The calculated V(D) was significantly higher in the sheep (10.4+/-2.7 L kg(-1)) and the calves (3.8+/-0.7 L kg(-1)) compared to the dogs (1.0+/-0.6 L kg(-1)). The rank order of increasing C(L) was sheep (3.4+/-2.7 L h(-1)kg(-1))>calves (2.7+/-0.4 L h(-1) kg(-1))>dogs (0.5+/-0.2 L h(-1)kg(-1)). The result for the dogs was significantly lower that the calculated C(L) for the sheep and calves. All these results indicate that the oral administration of S-carboxymethyl-L-cysteine may be useful during the therapeutic management of chronic obstructive pulmonary disease in dogs.


Subject(s)
Carbocysteine/administration & dosage , Carbocysteine/pharmacokinetics , Administration, Oral , Animals , Carbocysteine/blood , Cattle , Dogs , Male , Sheep , Species Specificity , Tissue Distribution/drug effects , Tissue Distribution/physiology
4.
Expert Opin Pharmacother ; 10(4): 693-703, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19239402

ABSTRACT

BACKGROUND: Carbocysteine is a muco-active drug with free radical scavenging and anti-inflammatory properties. It is actually approved for clinical use as adjunctive therapy of respiratory tract disorders characterized by excessive, viscous mucus, including chronic obstructive airways disease (COPD). OBJECTIVE: The intriguing antioxidant and anti-inflammatory properties of carbocysteine, beyond its known mucolytic activity, are described to explain its therapeutic efficacy and suggest new clinical uses. METHODS: After reviewing physiology and preclinical studies, human studies on the use of carbocysteine in chronic inflammatory diseases, i.e., COPD and cancer cachexia, are reviewed. RESULTS/CONCLUSIONS: Carbocysteine has been recently recognized as an effective and safe treatment for the long-term management of COPD, able to reduce the incidence of exacerbations and improve patient quality of life. Moreover, carbocysteine was effective in counteracting some symptoms associated with cancer cachexia. Preclinical and clinical studies have demonstrated that the antioxidant and anti-inflammatory properties of carbocysteine are more important than mucolysis itself for its therapeutic efficacy. Therefore, carbocysteine may be able to reverse the oxidative stress associated with several chronic inflammatory diseases, such as cardiovascular diseases and neurodegenerative disorders. Controlled, randomized studies in humans are warranted.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Cachexia/drug therapy , Carbocysteine/therapeutic use , Inflammation/drug therapy , Neoplasms , Pulmonary Disease, Chronic Obstructive/drug therapy , Anti-Inflammatory Agents/pharmacokinetics , Cachexia/etiology , Cachexia/metabolism , Carbocysteine/pharmacokinetics , Humans , Inflammation/metabolism , Neoplasms/complications , Oxidative Stress/drug effects , Pulmonary Disease, Chronic Obstructive/metabolism , Treatment Outcome
5.
Drug Metabol Drug Interact ; 23(3-4): 261-82, 2008.
Article in English | MEDLINE | ID: mdl-19326770

ABSTRACT

The purpose of this investigation was to reaction phenotype the identity of the cytosolic enzyme responsible for the S-oxidation of S-carboxymethyl-L-cysteine (SCMC) in female human hepatic cytosolic fractions. The identity of this enzyme in the female Wistar rat hepatic cytosolic fraction was found to be phenylalanine 4-monooxygenase (PAH). In pooled female human hepatic cytosolic fractions the calculated K(m) and V(max) for substrate (SCMC) activated PAH was 16.22 +/- 11.31 mM and 0.87 +/- 0.41 nmoles x min(-1) mg(-1). The experimental data modelled to the Michaelis-Menten equation with noncompetitive substrate inhibition. When the cytosolic fractions were activated with lysophophatidylcholine the V(max) increased to 52.31 +/- 11.72 nmoles x min(-1) mg(-1) but the K(m) remained unchanged at 16.53 +/- 2.32 mM. A linear correlation was seen in the production of Tyr and SCMC R/S S-oxide in 20 individual female hepatic cytosolic fractions for both substrate and lysophosphatidylcholine activated PAH (r(s) > 0.96). Inhibitor studies found that the specific chemical and antibody inhibitors of PAH reduced the production of Tyr and SCMC R/S S-oxide in these in vitro PAH assays. An investigation of the mechanism of interaction of SCMC with PAH indicated that the drug was a competitive inhibitor of the aromatic C-oxidation of Phe with a calculated K(i) of 17.23 +/- 4.15 mM. The requirement of BH4 as cofactor and the lack of effect of the specific tyrosine hydroxylase, tryptophan hydroxylase and nitric oxide synthase inhibitors on the S-oxidation of SCMC all indicate that PAH was the enzyme responsible for this biotransformation reaction in human hepatic cytosolic fractions.


Subject(s)
Carbocysteine/pharmacokinetics , Cytosol/metabolism , Hepatocytes/metabolism , Phenylalanine Hydroxylase/physiology , Biotransformation , Carbocysteine/pharmacology , Coenzymes/metabolism , Cytosol/enzymology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Free Radical Scavengers/pharmacokinetics , Free Radical Scavengers/pharmacology , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Oxidation-Reduction , Phenylalanine/pharmacology , Phenylalanine Hydroxylase/antagonists & inhibitors
7.
Pharmazie ; 61(5): 446-9, 2006 May.
Article in English | MEDLINE | ID: mdl-16724543

ABSTRACT

The bioequivalence of two carbocysteine capsulae preparations was assessed in 18 healthy volunteers who received a single 750 mg dose of each carbocysteine formulation, and a new sensitive method for the quantification of carbocysteine in human plasma was developed. The study was conducted using an open, randomized, two-sequence, two-period crossover design with a week washout period between the succesive treatments. Plasma samples were obtained over a 12-hour period and analyzed by high performance liquid chromatography coupled to electrospray ionization-mass spectrometry. Either a multiplicative statistic model for concentration-dependent parameters or an additive approach for time-related parameters were used for the comparison of pharmacokinetic parameters describing both the early and total exposure to carbocysteine. The respective 90% confidence limits [CL] of the individual ratios of geometric means were 0.898 to 1.112 [point estimate 0.999] for Cmax and 0.923 to 1.210 [point estimate 1.057] for AUC(0-infinity), while the difference between times elapsed to reach Cmax was insignificant [p = 0.4497]. Since both 90% CL for the log-transformed AUC(0-infinity) and Cmax geometric mean ratios were included in the proposed 0.80-1.25 interval, test drug (Bronchobos capsules) was considered bioequivalent to the reference one (Mucopront capsules).


Subject(s)
Antitussive Agents/administration & dosage , Antitussive Agents/pharmacokinetics , Carbocysteine/administration & dosage , Carbocysteine/pharmacokinetics , Area Under Curve , Biological Availability , Capsules , Chemistry, Pharmaceutical , Chromatography, High Pressure Liquid , Cross-Over Studies , Drug Compounding , Drugs, Generic , Gas Chromatography-Mass Spectrometry , Half-Life , Humans , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization , Therapeutic Equivalency
9.
Toxicol Lett ; 161(3): 188-94, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16229978

ABSTRACT

The clinical use of the alkylating oxazaphosphorine ifosfamide is hampered by a potentially severe encephalopathy. S-carboxymethylcysteine (SCMC), a metabolite of ifosfamide (IF), activates the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)/kainate receptor, causes neuronal acidification, and could thus be responsible for the encephalopathy. Since the presence of SCMC in brain has not been documented following administration of IF, SCMC was measured in the brain of mice following both the individual i.p. administration of IF and SCMC. SCMC was found in a concentration of 108.2 +/- 29.7 nmol/g following IF, but was detectable at much lower levels following the administration of SCMC (21.1 +/- 21.2 nmol/g). Together with the observation that the concentration of SCMC was 10-fold higher in liver than in brain 1h after administration of SCMC, these findings suggest that the SCMC found after IF was formed in the brain in situ. The concentration of glutamic acid was similar in IF and SCMC treated animals. Methylene blue, which is used clinically to treat and to prevent IF encephalopathy, did not decrease the formation of SCMC in brain. By inhibiting monoamine oxidase activity it did, however, markedly increase the concentration of serotonin in brain which could modulate the effects of SCMC on AMPA/kainate receptors. Thus, SCMC is present in brain following the administration of IF and could contribute to the IF-associated encephalopathy by activation of AMPA/kainate receptors.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacokinetics , Brain Diseases/metabolism , Brain/metabolism , Carbocysteine/pharmacokinetics , Ifosfamide/pharmacokinetics , Animals , Antineoplastic Agents, Alkylating/toxicity , Brain/drug effects , Brain Diseases/chemically induced , Carbocysteine/toxicity , Drug Interactions , Drug Therapy, Combination , Female , Glutamic Acid/metabolism , Ifosfamide/toxicity , Methylene Blue/pharmacology , Mice , Monoamine Oxidase Inhibitors/pharmacology , Serotonin/metabolism
10.
Rapid Commun Mass Spectrom ; 17(3): 192-6, 2003.
Article in English | MEDLINE | ID: mdl-12539182

ABSTRACT

A sensitive liquid chromatography/tandem mass spectrometry (LC/MS/MS) method was developed to determine carbocysteine in human plasma using 2-pyridylacetic acid as the internal standard (IS). The method employed derivatization with 10 M hydrochloric acid/methanol, which significantly improved the ionization efficiency of carbocysteine. After methanol-induced protein precipitation of plasma samples, carbocysteine and the IS were derivatized and subjected to LC/MS/MS analysis using atmospheric pressure chemical ionization. The method has a lower limit of quantitation of 20 ng/mL for a 0.2-mL plasma aliquot. The intra- and inter-day precision (RSD), calculated from quality control (QC) samples, was less than 7%. The accuracy, determined using QC samples, was within +/- 1%. The method offered increased sensitivity, selectivity and speed of analysis over existing methods. The method was utilized to support clinical pharmacokinetic studies of carbocysteine in volunteers following oral administration.


Subject(s)
Carbocysteine/blood , Chromatography, Liquid/methods , Mass Spectrometry/methods , Calibration , Carbocysteine/pharmacokinetics , Humans , Molecular Structure , Reproducibility of Results , Sensitivity and Specificity
11.
Drug Metabol Drug Interact ; 19(4): 241-55, 2003.
Article in English | MEDLINE | ID: mdl-14768973

ABSTRACT

A nearly complete recovery of radioactivity was achieved over 14 days following the oral administration of [35S]-S-methyl-L-cysteine sulphoxide and [35S]-S-carboxymethyl-L-cysteine sulphoxide to four healthy male volunteers. The urine was the major pathway of excretion of radioactivity (c. 96% in 0-14 days; c. 59% in 0-24 hours), with the faecal route being relatively unimportant (c. 1.7% in 0-3 days). Inorganic sulphate was an important degradation product, incorporating a substantial proportion of radioactive sulphur derived from these molecules (c. 40% in 0-14 days; c. 20% in 0-24 hours). Subtle differences were noted in the pattern of radioactive sulphate excretion following administration of the two cysteine-sulphoxide compounds, suggesting that their sulphur-containing moieties may enter different catabolic routes.


Subject(s)
Carbocysteine/pharmacokinetics , Cysteine/analogs & derivatives , Cysteine/pharmacokinetics , Sulfates/urine , Administration, Oral , Adult , Biodegradation, Environmental , Carbocysteine/urine , Cysteine/urine , Feces/chemistry , Humans , Male , Sulfates/metabolism , Sulfur Radioisotopes
12.
Drug Metab Dispos ; 27(9): 1092-7, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10460812

ABSTRACT

The routes of metabolism of S-carboxymethyl-L-cysteine in humans are dependent on the time of dosing. Administration of 750 mg of S-carboxymethyl-L-cysteine (Day 1) during the day at 8:00 AM followed by a 8:00 AM to 4:00 PM urine collection revealed that S-carboxymethyl-L-cysteine S-oxide was the major urinary metabolite produced. The 4:00 PM to midnight urine collection resulted in S-(carboxymethylthio)-L-cysteine being identified as the major urinary metabolite. However, the administration of 750 mg of S-carboxymethyl-L-cysteine (day 15) during the night at midnight and analysis of the midnight to 8:00 AM urine collection found that thiodiglycolic acid was the major urinary metabolite, whereas thiodiglycolic S-oxide was identified as the major urinary metabolite in the 8:00 AM to 4:00 PM urine collection. A diurnal variation in the metabolism of S-carboxymethyl-L-cysteine was seen and, in particular, the timing of S-carboxymethyl-L-cysteine administration had a profound effect on the identity of urinary S-oxide metabolites produced. After administration at 8:00 AM the urinary S-oxides produced were S-carboxymethyl-L-cysteine S-oxide and S-methyl-L-cysteine S-oxide but at midnight the major urinary S-oxide metabolite produced was thiodiglycolic acid S-oxide.


Subject(s)
Anti-Infective Agents, Local/pharmacokinetics , Carbocysteine/pharmacokinetics , Circadian Rhythm/physiology , Adult , Anti-Infective Agents, Local/administration & dosage , Anti-Infective Agents, Local/urine , Biotransformation , Carbocysteine/administration & dosage , Carbocysteine/urine , Chromatography, High Pressure Liquid , Humans , Male , Oxidation-Reduction
13.
Toxicology ; 111(1-3): 43-65, 1996 Jul 17.
Article in English | MEDLINE | ID: mdl-8711748

ABSTRACT

Proper bodily response to environmental toxicants presumably requires proper function of the xenobiotic (foreign chemical) detoxification pathways. Links between phenotypic variations in xenobiotic metabolism and adverse environmental response have long been sought. Metabolism of the drug S-carboxymethyl-L-cysteine (SCMC) is polymorphous in the population, having a bimodal distribution of metabolites, 2.5% of the general population are thought to be nonmetabolizers. The researchers developing this data feel this implies a polymorphism in sulfoxidation of the amino acid cysteine to sulfate. While this interpretation is somewhat controversial, these metabolic differences reflected may have significant effects. Additionally, a significant number of individuals with environmental intolerance or chronic disease have impaired sulfation of phenolic xenobiotics. This impairment is demonstrated with the probe drug acetaminophen and is presumably due to starvation of the sulfotransferases for sulfate substrate. Reduced metabolism of SCMC has been found with increased frequency in individuals with several degenerative neurological and immunological conditions and drug intolerances, including Alzheimer's disease, Parkinson's disease, motor neuron disease, rheumatoid arthritis, and delayed food sensitivity. Impaired sulfation has been found in many of these conditions, and preliminary data suggests that it may be important in multiple chemical sensitivities and diet responsive autism. In addition, impaired sulfation may be relevant to intolerance of phenol, tyramine, and phenylic food constituents, and it may be a factor in the success of the Feingold diet. These studies indicate the need for the development of genetic and functional tests of xenobiotic metabolism as tools for further research in epidemiology and risk assessment.


Subject(s)
Carbocysteine/analogs & derivatives , Environmental Pollutants/pharmacokinetics , Sulfur/metabolism , Xenobiotics/pharmacokinetics , Carbocysteine/pharmacokinetics , Environmental Pollutants/adverse effects , Genetic Variation , Humans , Inactivation, Metabolic , Multiple Chemical Sensitivity/etiology , Multiple Chemical Sensitivity/genetics , Multiple Chemical Sensitivity/metabolism , Phenotype , Xenobiotics/adverse effects
14.
Pharmacogenetics ; 3(5): 270-4, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8287066

ABSTRACT

It has been claimed that the amino acid derivative carbocisteine is predominantly metabolized by sulfoxidation and that this pathway exhibits a genetic polymorphism. Moreover, those subjects with a 'poor metabolizer' phenotype have been thought to have a genetic predisposition to developing certain diseases. We have confirmed the observations of others that this marker drug does not undergo significant S-oxidation. Furthermore, a novel urinary metabolite, S-(carboxymethylthio)-L-cysteine (CMTC) has recently been identified. To determine if a genetic polymorphism for this biotransformation pathway exists, metabolic ratios (% urinary excretion carbocisteine/% urinary excretion CMTC) for 120 healthy volunteers were assessed using high-performance thin-layer chromatography. Urinary excretion of the parent drug ranged from 6% of the dose administered to 56% (mean +/- SD, 23.4 +/- 0.8%). No cysteinyl sulfoxide metabolites were identified in the urine samples. The amount excreted as CMTC exhibited a 12-fold variation but only accounted for mean of 4.4% (1-12%) of the dose given. Two individuals initially had high metabolic ratios (> 30), however, on rechallenge both their MRs were less than 5. Therefore, carbocisteine is not an appropriate probe drug for sulfoxidation. The formation of the novel metabolite CMTC appears to exhibit polymorphism, although the considerable intra-subject variation for its formation does not allow assignment of a phenotype.


Subject(s)
Carbocysteine/metabolism , Adult , Aged , Aged, 80 and over , Biotransformation , Carbocysteine/analogs & derivatives , Carbocysteine/pharmacokinetics , Carbocysteine/urine , Female , Humans , Male , Middle Aged , Phenotype , Polymorphism, Genetic , United Kingdom , White People/genetics
16.
Eur J Clin Pharmacol ; 40(4): 387-92, 1991.
Article in English | MEDLINE | ID: mdl-2050173

ABSTRACT

A quantitative study has been made of the metabolism of S-carboxymethyl-L-cysteine (CMC) and its sulphoxides in volunteers by HPLC. Precolumn derivatization was applied prior to gradient reversed phase HPLC separation and fluorescence detection. For CMC and its metabolites containing a primary amino group the reagent 9-fluorenylmethylchloroformate was used. The other metabolites of CMC were derivatized at their carboxylic group with 1-pyrenyldiazomethane to give stable fluorescent products. Urine samples were collected for 8 h after oral administration of 1.125 g CMC to 33 healthy volunteers. Elimination of CMC in urine as sulphoxides did not account for more than 1% of the dose in any of the volunteers. Thus, CMC-sulphoxide metabolites are not quantitatively important. Recovery of the original substance in 8-hour urines ranged from 10 to 30% and a further 2 to 20% was recovered as the metabolite thiodiglycolic acid. Oral doses of 0.19, 1.125, and 2.25 g CMC in a second group of 12 healthy volunteers did not reveal dose dependence of the urinary excretion of the sulphoxides or of thiodiglycolic acid. Serum concentration-time-curves of CMC, (S)- and (R)-CMC sulphoxide were measured in a group of 9 healthy volunteers. The CMC sulphoxides in serum reached 1.5% of the parent substance after 4 hours. The ratio of CMC to its sulphoxide metabolites was similar in serum and urine. Pharmacogenetic polymorphism of sulphoxidation was not confirmed by the specific HPLC methods used.


Subject(s)
Carbocysteine/metabolism , Sulfoxides/urine , Administration, Oral , Adult , Carbocysteine/analogs & derivatives , Carbocysteine/pharmacokinetics , Chromatography, High Pressure Liquid/methods , Female , Humans , Male , Middle Aged
17.
Respiration ; 57(6): 353-8, 1990.
Article in English | MEDLINE | ID: mdl-2099568

ABSTRACT

The rheological behavior and clearance of bronchial mucus samples collected by protected expectoration from 24 out-patients with simple chronic bronchitis were investigated before, at the end of a short period of treatment (4 days) with a single oral dose of 2.7 g (sachet) of carbocysteine-lysine (evening meal), and on the 4th and 8th days after the end of treatment versus placebo. In the group treated with carbocysteine-lysine, there were significant reductions in viscosity (-67, -48, -62%) and increases in mucociliary transport (+41, +31, +34%) at the three times mentioned. The most striking finding was that the improvements were still present 8 days after cessation of treatment. The elasticity parameter was not affected in any statistically significant way (-10, -24, +65%). These findings suggest the presence of some type of 'post-mucoactive' effect.


Subject(s)
Bronchitis/metabolism , Carbocysteine/pharmacokinetics , Mucus/metabolism , Adult , Aged , Chronic Disease , Female , Humans , Male , Middle Aged , Mucociliary Clearance , Random Allocation , Rheology , Viscosity
18.
J Inorg Biochem ; 37(4): 309-23, 1989 Dec.
Article in English | MEDLINE | ID: mdl-2628548

ABSTRACT

S-carboxymethyl-L-cysteine (SCC) is a mucolytic agent extensively used in the treatment of respiratory tract disorders. Some of the undesirable side effects observed during SCC therapy being reminiscent of symptoms characteristic of copper and zinc imbalances, the objective of this paper was to test the possible interference of SCC with the metabolism of these two metals. Copper(II)- and zinc(II)-SCC complex equilibria have thus been investigated under physiological conditions by means of classical potentiometry combined with computer-assisted calculation techniques. Formation constants derived from these studies have then been used to simulate 1) the potential influence of SCC on the distribution of the above metals in blood plasma and 2) the extent to which gastrointestinal interactions between the drug and each metal ion in turn are likely to affect the bioavailability of each other. The results of these simulations show that 1) plasma therapeutic levels of SCC are not likely to induce dramatic changes in the distributions of copper(II) and zinc(II) low molecular weight fractions, 2) the gastrointestinal distribution of the drug is not affected by standard dietary doses of these metals, and 3) in contrast, therapeutic concentrations of SCC are capable of mobilizing significant fractions of both metals into tissue-diffusible electrically neutral complexes. In conclusion significant depletions of neither copper nor zinc are to be expected from oral administration of SCC. While the drug may to some extent facilitate the excretion of Cu2+ and Zn2+ ions from blood plasma, its gastrointestinal influence is, on the contrary, favorable to a better absorption of these two metals.


Subject(s)
Carbocysteine/metabolism , Computer Simulation , Copper/metabolism , Cysteine/analogs & derivatives , Zinc/metabolism , Absorption , Biological Availability , Carbocysteine/pharmacokinetics , Carbocysteine/pharmacology , Copper/blood , Digestive System/drug effects , Digestive System/metabolism , Humans , Hydrogen-Ion Concentration , Potentiometry , Zinc/blood
19.
J Hepatol ; 7(1): 72-8, 1988 Aug.
Article in English | MEDLINE | ID: mdl-3183354

ABSTRACT

On the basis of previous experimental studies we postulated that individuals who were phenotypically good hydroxylators but poor sulphoxidisers would be susceptible to chlorpromazine jaundice. Sulphoxidation capacity was assessed in 12 subjects with a history of chlorpromazine jaundice, using S-carboxymethyl-L-cysteine as an in vivo probe. Following an oral dose of 750 mg, unchanged compound and sulphoxide metabolites were measured in urine. All 12 subjects (100%) were shown to be poor sulphoxidisers compared to 22% of normal controls (P less than 0.001) and 23.8% of liver disease controls (P less than 0.001). No subjects with a history of chlorpromazine jaundice had an impaired hydroxylation capacity as assessed by recovery of 4-hydroxydebrisoquine in urine following oral debrisoquine. The results support the hypothesis and demonstrate an inherent metabolic basis of susceptibility to chlorpromazine jaundice.


Subject(s)
Chlorpromazine/adverse effects , Cholestasis/chemically induced , Mixed Function Oxygenases/metabolism , Sulfoxides/metabolism , Adult , Aged , Aged, 80 and over , Carbocysteine/pharmacokinetics , Cholestasis/metabolism , Female , Humans , Hydroxylation , Male , Middle Aged
20.
J Chromatogr ; 428(2): 301-10, 1988 Jul 15.
Article in English | MEDLINE | ID: mdl-3215933

ABSTRACT

A method is described for the determination of S-(carboxymethyl)-L-cysteine in serum. After addition of S-(carboxyethyl)-L-cysteine as internal standard, both compounds are extracted into methanol, converted into fluorescent derivatives with o-phthalaldehyde and quantitatively determined by reversed-phase high-performance liquid chromatography. Chromatography of unwanted amino acid derivatives is avoided by column switching, thereby shortening analysis time and increasing column lifetime. The technique was applied in a study of the bioavailability of S-(carboxymethyl)-L-cysteine after oral administration to humans. The concentration-response curve was linear from 2 to 16 micrograms/ml; mean serum concentrations are reported.


Subject(s)
Carbocysteine/blood , Cysteine/analogs & derivatives , Carbocysteine/pharmacokinetics , Chromatography, Liquid , Humans , Indicators and Reagents , o-Phthalaldehyde
SELECTION OF CITATIONS
SEARCH DETAIL
...