Logo Passei Direto
Buscar
Material
páginas com resultados encontrados.
páginas com resultados encontrados.

Prévia do material em texto

Toxicokinetics of Drugs of Abuse: Current Knowledge
of the Isoenzymes Involved in the Human Metabolism
of Tetrahydrocannabinol, Cocaine, Heroin, Morphine,
and Codeine
Hans H. Maurer, Christoph Sauer, and Denis S. Theobald
Abstract: This review summarizes the major metabolic pathways
of the drugs of abuse, tetrahydrocannabinol, cocaine, heroin,
morphine, and codeine, in humans including the involvement of
isoenzymes. This knowledge may be important for predicting
their possible interactions with other xenobiotics, understanding
pharmaco-/toxicokinetic and pharmacogenetic variations, tox-
icological risk assessment, developing suitable toxicological
analysis procedures, and finally for understanding certain
pitfalls in drug testing. The detection times of these drugs and/
or their metabolites in biological samples are summarized and
the implications of the presented data on the possible inter-
actions of drugs of abuse with other xenobiotics, ie, inhibition or
induction of individual polymorphic and nonpolymorphic
isoenzymes, discussed.
Key Words: drugs of abuse, metabolism, toxicokinetics, cocaine,
codeine, heroin, morphine, tetrahydrocannabinol
(Ther Drug Monit 2006;28:447–453)
Individual variations in the pharmacological or toxico-logical responses to the same drug dose may be caused
by a variety of factors such as body mass, age, sex, kidney
and liver function, drug–drug (food–drug) interactions,
and genetic variability.1 Detailed knowledge of the
metabolism of drugs (of abuse) allows for the prediction
of possible interactions with other xenobiotics because of,
for example, inhibition or induction of individual meta-
bolic isoenzymes by poisons, drugs (of abuse), alcohol,
tobacco smoke or food ingredients. It is thus a
prerequisite for understanding pharmaco-/toxicokinetics,
pharmacogenetic variations, evidence-based case inter-
pretation, toxicological risk assessment, developing
toxicological analysis procedures, and understanding
pitfalls in drug testing.
Drug metabolism is largely determined by the
relevant genetic variants of metabolic enzymes or trans-
port proteins. The most important variants are the
following polymorphically expressed proteins: the cyto-
chrome P450 (CYP) isoenzymes CYP2C9, CYP2C19, and
CYP2D6, the alcohol dehydrogenase ADH2 and the
aldehyde dehydrogenase ALDH2, the phase II enzymes
uridine diphosphate glucuronyltransferases (UGT)
UGT1A1, thiopurin methyltransferases, arylamine N-
acetyl transferases NAT2 and glutathione S-transferases
GSTM1 and GSTT1, and finally transporters like P-
glycoprotein.1 Genetic variants of CYP have been
discovered because of the atypical clinical responses of
certain individuals to certain drugs, which could later be
attributed to a reduced ability of these individuals to
metabolize the respective drugs. At least 30 drugs have
subsequently been shown to be oxidized by CYP2D6,
including dextromethorphan, tricyclic antidepressants,
some neuroleptics, beta-blockers, and antiarrhythmic
agents such as perhexiline and flecainide, and drugs
of abuse including eg, codeine, dihydrocodeine, and
oxycodone.2 Also designer drugs like para-methoxyam-
phetamine,3 para-methoxymethamphetamine,4 or the
piperazines 1-(3-chlorophenyl)piperazine (mCPP),5 1-(4-
methoxyphenyl)piperazine,6 and 1-(3-trifluoromethylphe-
nyl)piperazine7 were shown to be predominantly or
exclusively metabolized by this enzyme. CYP2D6-
mediated metabolism of such drugs is known to be a
major source of pharmacokinetic variations or variations
in drug effects.
In the last few years, many studies have been
published regarding the impact of pharmacogenetic
variability, for example, on drug transport and meta-
bolism resulting in different pharmacokinetic behavior or
toxic risks.1,2,8–15 In addition, variations resulting from
interactions with other drugs of abuse, therapeutically
administered drugs, food ingredients and/or smoking and
the impact of supra-pharmacological doses on drug
disposition must be taken into consideration.16–21 For
assessment or prediction of all these variations, it is
important to know whether an individual has a reduced
or an increased turnover in these steps due to geneticCopyright r 2006 by Lippincott Williams & Wilkins
Received for publication January 23, 2006; accepted February 27, 2006.
From the Department of Experimental and Clinical Toxicology,
Institute of Experimental and Clinical Pharmacology and Toxico-
logy, University of Saarland, D–66421 Homburg (Saar), Germany.
Dedicated to Prof Dr Irving Sunshine at the occasion of his 90th
birthday.
Reprints: Prof Hans H. Maurer, PhD, Department of Experimental and
Clinical Toxicology, University of Saarland, D-66421 Homburg
(Saar), Germany (e-mail: hans.maurer@uniklinikum-saarland.de).
REVIEW ARTICLE
Ther Drug Monit � Volume 28, Number 3, June 2006 447
variations or drug/food interactions via enzyme inhibition
or induction. Furthermore, knowledge about the iso-
enzymes involved in the particular metabolic steps of all
used drugs is of great importance. Today, pharmaceutical
companies are required to provide such data for all new
drug entities introduced into the market. They are usually
published in review articles2,22 or drug information
handbooks or databases (eg, Refs. 23, 24). In contrast,
such data on illicit drugs of abuse are found in various
publications. Therefore, in the following pages, the main
metabolic steps and the isoenzymes involved therein will
be reviewed for delta9-tetrahydrocannabinol, cocaine,
heroin, morphine, and codeine. Data for amphetamines
and designer drugs of the ecstasy, the piperazine-, and the
pyrrolidinophenone-type are not included here, because
they have recently been reviewed elsewhere.25–29 Know-
ledge of the toxicokinetics of drugs of abuse is important
to find out which drug can be detected in which body
sample (urine, blood, saliva, sweat, hair, nails), via which
target analyte (parent drug or metabolite) and for how
long.30–32 In addition, this knowledge is needed to find
out whether a detected compound is formed by a licit
(eg, codeine) or an illicit drug (eg, heroin). Finally, for
pharmacological and/or toxicological interpretation of
the analytical results, pharmacokinetic calculations are
often used as a basis.33 However, kinetic data coming
either from controlled studies or from single case reports
may not reflect kinetic data in drug abuse situations and
must therefore be used very carefully.
METABOLISM OF TETRAHYDROCANNABINOL
Delta9-tetrahydrocannabinol (THC, also named
delta1-tetrahydrocannabinol according to the monoter-
penoid nomenclature), the primary hallucinogenic con-
stituent of Cannabis sativa, is subject to hepatic
metabolism primarily by hydroxylation (Fig. 1). The
primary metabolite 11-hydroxy-THC (HO-THC), which
is pharmacologically active, is formed by CYP2C9,34–37
and further oxidized, probably by alcohol dehydrogenase
or microsomal alcohol oxygenase, to the intermediate
aldehyde 11-oxo-THC followed by oxidation to 11-nor-9-
carboxy-THC (THC-COOH) catalyzed by a microsomal
aldehyde oxygenase, a member of the CYP2C subfam-
ily.35,38,39 After glucuronidation of the carboxy group,40
THC-COOH is excreted in urine and can be detected up
to 4 days after smoking 1 marijuana cigarette and up to 4
weeks after frequent use.30,31 In blood, THC levels increase
rapidly after smoking, peak before the end of smoking, and
quickly dissipate. Mean peak HO-THC levels are sub-
stantially lower than THC levels and occur immediately
after the end of smoking. THC-COOH levels increase
slowly and plateau for an extended period. The mean peak
time for THC-COOH is about 2 hours after smoking and a
correspondingly longer time course of detection can be
observed.41 THC-COOH glucuronide is present in blood at
considerably higher concentrations than the unconjugated
acid and is unstable to hydrolysis.42 This may cause
misinterpretation when using models for the prediction
of time of cannabis exposure.43
Further minor metabolic steps yieldseveral hydroxy
metabolites at C1’ to C5’ followed by beta-oxidation
to the corresponding carboxylic acids ((CH2)nCOOH),
37
2 diastereomeric 8-hydroxy metabolites followed by
dehydration,36,37 and an epoxide at C9-10 followed
by hydrolysis or glutathione conjugation.36 Formation
of 8-hydroxy-THC and the epoxide are catalyzed by
CYP3A4.36 Finally, several combinations of the meta-
bolic steps have been described.37
Given the high affinity of CYP2C9 for the hydro-
xylation of THC, the potential for an interaction was
evaluated between THC, HO-THC, or THC-COOH and
the CYP2C9 substrate phenytoin.34 Surprisingly, THC
increased the rate of phenytoin hydroxylation in human
liver microsomes and expressed CYP2C9 enzyme (Super-
somes). Similar increases in the rate of phenytoin meta-
bolism were observed with coincubation of HO-THC and
THC-COOH. These in vitro data suggest the potential for
FIGURE 1. Major metabolic pathways
of delta9-tetrahydrocannabinol in hu-
mans with the isoenzymes involved.
Maurer et al Ther Drug Monit � Volume 28, Number 3, June 2006
448 r 2006 Lippincott Williams & Wilkins
an interaction from the concomitant administration of
THC and phenytoin that could result in decreased
phenytoin concentrations in vivo and thus an increased
risk of epileptic seizures.34
In the context of the use of cannabis-based medicine
extracts for therapeutic purposes, a study was performed
in a double-blind and placebo-controlled cross-over
design in which each of 24 volunteers received an oral
dose of THC, of a cannabis extract containing THC and
cannabidiol (CBD) or placebo in weekly intervals.38
Despite the large variation of the kinetic data, evidence
emerged from the total of the results that CBD partially
inhibits the CYP2C9-catalyzed hydroxylation of THC to
HO-THC. The probability for this inhibition is particu-
larly high for oral intake because THC and CBD attain
relatively high concentrations in the liver and because of
the high first-pass metabolism of THC. However, the
effect of CBD seems to be small in comparison to the
variability caused by other factors. Therefore, a pharma-
cokinetic reason for the differences determined between
pure THC and cannabis extract is improbable at the doses
chosen in this study.
METABOLISM OF COCAINE
Cocaine, the main alkaloid of Erythroxylum coca,
is a powerful stimulant that is primarily metabolized
by the 3 esterases pseudocholinesterase, human carbo-
xylesterase-1 (hCE-1), and human carboxylesterase-2
(hCE-2).44–48 Figure 2 shows that cocaine is hydrolyzed
mainly by hCE-1 to benzoylecgonine, the primary
metabolite excreted in the urine, or by pseudocholinester-
ase and hCE-2 to ecgonine methyl ester. In the presence
of ethanol, hCE-1 catalyzes transesterification of cocaine
to cocaethylene, a toxic metabolite, that can be further
hydrolyzed by hCE-1 to benzoylecgonine or by hCE-2 to
ecgonine ethyl ester.44,45,47
The main metabolite benzoylecgonine could be
detected in urine for 2 to 3 days after an intranasal dose
of 100mg of cocaine, for 1.5 days after a single
intravenous dose of 20mg, and for 10 days after chronic
intravenous doses of 8 g/d.31 The detection time of
cocaine in blood is 4 to 6 hours after 20mg and 12 hours
after 100mg. In the serum of chronic users, benzoylecgo-
nine was detectable for 5 days on average.31 At this point,
it must be mentioned that cocaine may be enzymatically
degraded in blood samples during transport and storage,
which can be avoided by the use of blood sampling
devices containing the cholinesterase inhibitor sodium
fluoride.49
The first step in the oxidative metabolism of cocaine
is N-demethylation to pharmacologically active norco-
caine catalyzed by 2 alternate pathways, one involving
only CYP3A450 and the other requiring both CYP and
flavin-containing monooxygenase . In the first pathway,
cocaine was directly N-demethylated to norcocaine by
CYP and the second route was found to be a 2-step
reaction involving cocaine N-oxide as an intermediate. In
this pathway, cocaine is first oxidized to cocaine N-oxide
by flavin-containing monooxygenase, followed by a CYP-
catalyzed N-demethylation to norcocaine.51 Norcocaine is
further metabolized by N–hydroxylation at least partly
catalyzed by several CYP subfamilies including 1A, 2A,
3A and possibly 2B.52 In addition, aromatic m- and p-
hydroxylation of cocaine partly followed by hydrolysis to
the corresponding hydroxy benzoylecgonine isomers
(minor pathways not shown in Fig. 2).53,54 Unfortunately,
the involved isoenzymes are not yet identified. Again, in
the presence of ethanol, hCE-1 catalyzes transesterifica-
tion of norcocaine to norcocaethylene that can be further
hydrolyzed by hCE-1 to benzoylnorecgonine.44,45,47
Crack is cocaine that has been processed from
cocaine hydrochloride to a free base for smoking. When
cocaine is smoked, a pyrolytic product, methylecgonidine
(anhydroecgonine methylester), is also consumed with the
cocaine. The amount of methylecgonidine formed
depends on the pyrolytic conditions and composition of
the illicit cocaine. Besides cocaine and its metabolites,
FIGURE 2. Major metabolic pathways
of cocaine in humans with the iso-
enzymes involved.
Ther Drug Monit � Volume 28, Number 3, June 2006 Toxicokinetics of Drugs of Abuse
r 2006 Lippincott Williams & Wilkins 449
3 metabolites of methylecgonidine could be identified:
ecgonidine (anhydroecgonine), ethyl ecgonidine (anhy-
droecgonine ethyl ester) and nor-ecgonidine (nor-
anhydroecgonine).54,55 Unfortunately, the isoenzymes
involved in the metabolism of methylecgonidine are not
yet identified.
More recently, the activity of several human
cholinesterase variants with cocaine has been examined.56
One atypical cholinesterase had 10-fold lower binding
efficiency for cocaine and 10-fold lower catalytic effi-
ciency. Although this evidence suggests the possibility
that individual responses to cocaine may be mediated in
part by greater or lesser metabolic capacity, genetically
determined variations in cholinesterase activity, or in
activity of the carboxylesterase, have not been investi-
gated in persons with addictive diseases.48
METABOLISM OF MORPHINE AND HEROIN
Morphine is the main alkaloid of Papaver somnifer-
um and is used therapeutically as a potent analgesic
although its 3,6-diacetyl derivative heroin is one of the
most dangerous drugs of abuse. The oral bioavailability
of heroin is poor only due to complete first-pass
metabolism by hepatic and extrahepatic factors.48
It exerts its effects only after metabolism to 6-monoace-
tylmorphine and morphine catalyzed in the liver by hCE-1
and hCE-2, in the serum by pseudocholinesterase, and also
nonenzymatically in the serum (Fig. 3). The catalytic
efficiency for both hydrolysis steps is substantially greater
for hCE-2 than for the other esterases.45,46,48,57
Morphine administered as a drug or formed
metabolically from heroin is mainly metabolized by
UGT to the inactive metabolite morphine-3-glucuronide
(M3G) and, to a lesser extent, to morphine-6-glucuronide
(M6G),48 that has equivalent analgesic effects to mor-
phine and an improved side effect profile with a reduced
tendency to cause nausea, vomiting, sedation and
respiratory depression.58 UGT2B7 is the major enzyme
involved in morphine 3- and 6-glucuronidation, but 1A1,
1A3, 1A6, 1A8, 1A9, 1A10 also more or less contribute to
M3G formation.59 Finally, morphine is N-demethylated
to normorphine by hepatic CYP3A4 and to a lesser extent
by CYP2C8.60 Today, all major heroin and morphine
metabolites can be monitored in urine and blood.61,62
The detection time of morphine in blood is 20 hours after
intravenous administration of 12 or 20mg heroin in a
subject.63 After being smoked, the detection time for
10.5mg of heroin varied between 22 minutes and
2 hours.63 In the blood of chronic users, total morphine
was detectable for 29.2 hours on average and free
morphine for 14.4 hours.64 After administration of 3 to
7 or 10.5 to 13.9mg heroin intravenously, 6-acetylmor-
phine was detectablein urine up to 5.1 hours (median 2.3)
and 2.3 to 11.2 hours (median 4.5), respectively, by
GC-MS with a cutoff of 10 ng/mL, whereas total
morphine was detectable in urine for 7.4 to 31.9 hours
(median 15.7) and 10.7 to 53.5 hours (median 34.4),
respectively, by GC-MS with a cutoff of 300 ng/mL.65
A recent study of 5 subjects with Gilbert syndrome,
characterized by impaired glucuronidation due to a
polymorphism in the gene encoding UGT1A1, did not
show altered morphine clearance or difference in the
plasma concentration versus time curves for M6G or
M3G compared with controls.66 The discussion on the
effect of UGT2B7 polymorphisms on M6G and M3G
formation and clearance is controversial.11,48
METABOLISM OF CODEINE
Codeine (3-methylmorphine) is a minor alkaloid of
Papaver somniferum. Synthesized from morphine, it is
used therapeutically primarily as a potent antitussive, but
also misused, for example, by opiate addicts because it is
metabolized to morphine which is further metabolized by
N-demethylation and glucuronidation as described earlier
FIGURE 3. Major metabolic pathways
of heroin and morphine in humans with
the isoenzymes involved.
Maurer et al Ther Drug Monit � Volume 28, Number 3, June 2006
450 r 2006 Lippincott Williams & Wilkins
(Fig. 4). The O-dealkylation to morphine is catalyzed by
polymorphically expressed CYP2D6 as is also the case for
its congeners dihydrocodeine, ethylmorphine, hydroco-
done, or oxocodone.11,12,48 Some variants of the CYP2D6
gene increase metabolism of these drugs to their more
potent metabolites, whereas others decrease metabolism,
which may influence the analgesic potency and abuse
liability.67–69 However, their clinical relevance, though
controvertial, is being discussed.48
In addition, codeine is N-demethylated by
CYP3A470,71 and conjugated equally by UGT2B4 and
UGT2B7 to codeine-6-glucuronide so that UGT2B7
polymorphism plays no significant role in this step.72
CONCLUSIONS
The knowledge of major metabolic pathways of
drugs of abuse in humans and the isoenzymes involved
therein may be important to predict possible interactions
with other xenobiotics, to understand pharmaco-/toxico-
kinetics and pharmacogenetic variations, to understand
toxic risks, to develop suitable toxicological analysis
procedures, and finally to understand pitfalls in drug
testing.
ACKNOWLEDGMENTS
The authors would like to thank Andreas H. Ewald
and Frank T. Peters for their help.
REFERENCES
1. Evans WE, McLeod HL. Pharmacogenomics-drug disposition, drug
targets, and side effects. N Engl J Med. 2003;348:538–549.
2. Rendic S. Summary of information on human CYP enzymes:
human P450 metabolism data. Drug Metab Rev. 2002;34:83–448.
3. Kitchen I, Tremblay J, Andre J, et al. Interindividual and
interspecies variation in the metabolism of the hallucinogen 4-
methoxyamphetamine. Xenobiotica. 1979;9:397–404.
4. Staack RF, Theobald DS, Paul LD, et al. Identification of human
cytochrome p450 2D6 as major enzyme involved in the o-
demethylation of the designer drug p-methoxymethamphetamine.
Drug Metab Dispos. 2004;32:379–381.
5. Rotzinger S, Fang J, Coutts RT, et al. Human CYP2D6 and
metabolism of m-chlorophenylpiperazine. Biol Psychiatry. 1998;44:
1185–1191.
6. Staack RF, Theobald DS, Paul LD, et al. In vivo metabolism of the
new designer drug 1-(4-methoxyphenyl)piperazine (MeOPP) in rat
and identification of the human cytochrome P450 enzymes
responsible for the major metabolic step. Xenobiotica. 2004;34:
179–192.
7. Staack RF, Paul LD, Springer D, et al. Cytochrome P450 dependent
metabolism of the new designer drug 1-(3-trifluoromethylphenyl)pi-
perazine (TFMPP). In vivo studies in wistar and dark agouti rats as
well as in vitro studies in human liver microsomes. Biochem
Pharmacol. 2004;67:235–244.
8. Zhang ZY, Wong YN. Enzyme kinetics for clinically relevant CYP
inhibition. Curr Drug Metab. 2005;6:241–257.
9. Eichelbaum M, Ingelman-Sundberg M, Evans WE. Pharmaco-
genomics and individualized drug therapy. Annu Rev Med. 2006;
57:119–137.
10. Tang C, Lin JH, Lu AY. Metabolism-based drug-drug interactions:
what determines individual variability in cytochrome P450 induc-
tion? Drug Metab Dispos. 2005;33:603–613.
11. Lotsch J, Skarke C, Liefhold J, et al. Genetic predictors of the
clinical response to opioid analgesics: clinical utility and future
perspectives. Clin Pharmacokinet. 2004;43:983–1013.
12. Zanger UM, Raimundo S, Eichelbaum M. Cytochrome P450 2D6:
overview and update on pharmacology, genetics, biochemistry.
Naunyn Schmiedebergs Arch Pharmacol. 2004;369:23–37.
13. Pritzker D, Kanungo A, Kilicarslan T, et al. Designer drugs that are
potent inhibitors of CYP2D6. J Clin Psychopharmacol. 2002;
22:330–332.
14. Antoniou T, Tseng AL. Interactions between recreational drugs and
antiretroviral agents. Ann Pharmacother. 2002;36:1598–1613.
15. Howard LA, Sellers EM, Tyndale RF. The role of pharmacogen-
etically-variable cytochrome P450 enzymes in drug abuse and
dependence. Pharmacogenomics. 2002;3:185–199.
16. Maurer HH. The relevance of pharmacogenetics and of
metabolic interactions for clinical and forensic toxicology. In:
Pragst F, Aderjan R, eds. Proceedings of the XIIIth GTFCh
FIGURE 4. Major metabolic pathways
of codeine in humans with the isoen-
zymes involved.
Ther Drug Monit � Volume 28, Number 3, June 2006 Toxicokinetics of Drugs of Abuse
r 2006 Lippincott Williams & Wilkins 451
Symposium in Mosbach. Heppenheim, Germany: Helm-Verlag;
2003:197–203.
17. Wojnowski L. Genetics of the variable expression of CYP3A in
humans. Ther Drug Monit. 2004;26:192–199.
18. Thummel KE, Wilkinson GR. In vitro and in vivo drug interactions
involving human CYP3A. Annu Rev Pharmacol Toxicol. 1998;
38:389–430.
19. Evans AM. Influence of dietary components on the gastrointestinal
metabolism and transport of drugs. Ther Drug Monit. 2000;
22:131–136.
20. Ozdemir V, Kalowa W, Tang BK, et al. Evaluation of the genetic
component of variability in CYP3A4 activity: a repeated drug
administration method. Pharmacogenetics. 2000;10:373–388.
21. Gibson GG, Plant NJ, Swales KE, et al. Receptor-dependent
transcriptional activation of cytochrome P4503A genes: induction
mechanisms, species differences and interindividual variation in
man. Xenobiotica. 2002;32:165–206.
22. Rendic S, Di Carlo FJ. Human cytochrome P450 enzymes: a status
report summarizing their reactions, substrates, inducers, and
inhibitors. Drug Metab Rev. 1997;29:413–580.
23. Bundesverband der Pharmazeutischen Industrie (BPI). Fachinfo CD
version 2004. Aulendorf, Germany: Editio Cantor Verlag; 2004.
24. Indiana University Department of Medicine -Division of Clinical
Pharmacology - Indianapolis. Drug Interactions -Cytochrome P450
System (http://medicine.iupui.edu/flockhart/), 2005.
25. Kraemer T, Maurer HH. Toxicokinetics of amphetamines: Meta-
bolism and toxicokinetic data of designer drugs, of amphetamine,
methamphetamine and their N-alkyl derivatives. Ther Drug Monit.
2002;24:277–289.
26. de la Torre R, Farre M, Roset PN, et al. Human pharmacology of
MDMA: pharmacokinetics, metabolism, and disposition. Ther Drug
Monit. 2004;26:137–144.
27. de la Torre R, Farre M. Neurotoxicity of MDMA (ecstasy): the
limitations of scaling from animals to humans. Trends Pharmacol
Sci. 2004;25:505–508.
28. Maurer HH, Kraemer T, Springer D, et al. Chemistry, pharmaco-
logy, toxicology, and hepatic metabolism of designer drugs of the
amphetamine (ecstasy), piperazine, and pyrrolidinophenone types, a
synopsis. Ther Drug Monit. 2004;26:127–131.
29. Staack RF, Maurer HH. Metabolism of designer drugs of abuse.
Curr Drug Metab. 2005;6:259–274.
30. Vandevenne M, Vandenbussche H, Verstraete A. Detection time of
drugs of abuse in urine. Acta Clin Belg. 2000;55:323–333.
31. Verstraete AG. Detection times of drugs of abuse in blood, urine,
and oral fluid. Ther Drug Monit. 2004;26:200–205.
32. Maurer HH. Position of chromatographic techniques in screening
for detection of drugs or poisons in clinical and forensic toxico-
logy and/or dopingcontrol. Clin Chem Lab Med. 2004;42:
1310–1324.
33. Toennes SWH, Maurer HH. Microsoft Excel in pharmacokinetic-
s–an easy way to solve kinetic problems in clinical toxicology, legal
medicine or doping control. In: Sachs H, Bernhard W, Jaeger A, eds.
Proceedings of the 34th International TIAFT Meeting, Interlaken.
Leipzig, Germany: Molina-Press; 1997;201–204.
34. Bland TM, Haining RL, Tracy TS, et al. CYP2C-catalyzed delta9-
tetrahydrocannabinol metabolism: kinetics, pharmacogenetics and
interaction with phenytoin. Biochem Pharmacol. 2005;70:1096–1103.
35. Watanabe K, Matsunaga T, Yamamoto I, et al. Involvement of
CYP2C in the metabolism of cannabinoids by human hepatic
microsomes from an old woman. Biol Pharm Bull.
1995;18:1138–1141.
36. Bornheim LM, Lasker JM, Raucy JL. Human hepatic microsomal
metabolism of delta 1-tetrahydrocannabinol. Drug Metab Dispos.
1992; 20:241–246.
37. Halldin MM, Widman M, Bahr V, et al. Identification of in vitro
metabolites of delta 1-tetrahydrocannabinol formed by human
livers. Drug Metab Dispos. 1982;10:297–301.
38. Nadulski T, Pragst F, Weinberg G, et al. Randomized, double-blind,
placebo-controlled study about the effects of cannabidiol (CBD) on
the pharmacokinetics of delta9-tetrahydrocannabinol (THC) after
oral application of THC verses standardized cannabis extract. Ther
Drug Monit. 2005;27:799–810.
39. Wall ME, Sadler BM, Brine D, et al. Metabolism, disposition, and
kinetics of delta-9-tetrahydrocannabinol in men and women. Clin
Pharmacol Ther. 1983;34:352–363.
40. Williams PL, Moffat AC. Identification in human urine of delta 9-
tetrahydrocannabinol-11-oic acid glucuronide: a tetrahydrocanna-
binol metabolite. J Pharm Pharmacol. 1980;32:445–448.
41. Huestis MA, Henningfield JE, Cone EJ. Blood cannabinoids. I.
Absorption of THC and formation of 11-OH-THC and THCCOOH
during and after smoking marijuana. J Anal Toxicol. 1992;
16:276–282.
42. Maralikova B, Weinmann W. Simultaneous determination of
Delta9-tetrahydrocannabinol, 11-hydroxy-Delta9-tetrahydrocanna-
binol and 11-nor-9-carboxy- Delta9-tetrahydrocannabinol in human
plasma by high-performance liquid chromatography/tandem mass
spectrometry. J Mass Spectrom. 2004;39:526–531.
43. Huestis MA, Henningfield JE, Cone EJ. Blood cannabinoids. II.
Models for the prediction of time of marijuana exposure from
plasma concentrations of delta 9-tetrahydrocannabinol (THC) and
11-nor-9-carboxy-delta 9-tetrahydrocannabinol (THCCOOH). J
Anal Toxicol. 1992;16:283–290.
44. Dean RA, Christian CD, Sample RH, et al. Human liver cocaine
esterases: ethanol-mediated formation of ethylcocaine. FASEB J.
1991;5:2735–2739.
45. Brzezinski MR, Spink BJ, Dean RA, et al. Human liver
carboxylesterase hCE-1: binding specificity for cocaine, heroin,
and their metabolites and analogs. Drug Metab Dispos. 1997;
25:1089–1096.
46. Bencharit S, Morton CL, Xue Y, et al. Structural basis of heroin and
cocaine metabolism by a promiscuous human drug-processing
enzyme. Nat Struct Biol. 2003;10:349–356.
47. Laizure SC, Mandrell T, Gades NM, et al. Cocaethylene metabolism
and interaction with cocaine and ethanol: role of carboxylesterases.
Drug Metab Dispos. 2003;31:16–20.
48. Kreek MJ, Bart G, Lilly C, et al. Pharmacogenetics and human
molecular genetics of opiate and cocaine addictions and their
treatments. Pharmacol Rev. 2005;57:1–26.
49. Toennes SW, Kauert GF. Importance of vacutainer selection in
forensic toxicological analysis of drugs of abuse. J Anal Toxicol.
2001;25:339–343.
50. LeDuc BW, Sinclair PR, Shuster L, et al. Norcocaine and N-
hydroxynorcocaine formation in human liver microsomes: role of
cytochrome P-450 3A4. Pharmacology. 1993;46:294–300.
51. Kloss MW, Rosen GM, Rauckman EJ. N-demethylation of cocaine
to norcocaine. Evidence for participation by cytochrome P-450 and
FAD-containing monooxygenase.Mol Pharmacol. 1983;23:482–485.
52. Pellinen P, Kulmala L, Konttila J, et al. Kinetic characteristics of
norcocaine N-hydroxylation in mouse and human liver microsomes:
involvement of CYP enzymes. Arch Toxicol. 2000;74:511–520.
53. Zhang JY, Foltz RL. Cocaine metabolism in man: identification of
four previously unreported cocaine metabolites in human urine.
J Anal Toxicol. 1990;14:201–205.
54. Cone EJ, Tsadik A, Oyler J, et al. Cocaine metabolism and urinary
excretion after different routes of administration. Ther Drug Monit.
1998;20:556–560.
55. Paul BD, Lalani S, Bosy T, et al. Concentration profiles of cocaine,
pyrolytic methyl ecgonidine and thirteen metabolites in human
blood and urine: determination by gas chromatography-mass
spectrometry. Biomed Chromatogr. 2005;19:677–688.
56. Xie W, Altamirano CV, Bartels CF, et al. An improved cocaine
hydrolase: the A328Y mutant of human butyrylcholinesterase is
4-fold more efficient. Mol Pharmacol. 1999;55:83–91.
57. Kamendulis LM, Brzezinski MR, Pindel EV, et al. Metabolism of
cocaine and heroin is catalyzed by the same human liver
carboxylesterases. J Pharmacol Exp Ther. 1996;279:713–717.
58. Kilpatrick GJ, Smith TW. Morphine-6-glucuronide: actions and
mechanisms. Med Res Rev. 2005;25:521–544.
59. Stone AN, Mackenzie PI, Galetin A, et al. Isoform selectivity and
kinetics of morphine 3- and 6-glucuronidation by human udp-
glucuronosyltransferases: evidence for atypical glucuronidation
Maurer et al Ther Drug Monit � Volume 28, Number 3, June 2006
452 r 2006 Lippincott Williams & Wilkins
kinetics by UGT2B7. Drug Metab Dispos. 2003;31:
1086–1089.
60. Projean D, Morin PE, Tu TM, et al. Identification of CYP3A4 and
CYP2C8 as the major cytochrome P450 s responsible for morphine
N-demethylation in human liver microsomes. Xenobiotica. 2003;
33:841–854.
61. von Euler M, Villen T, Svensson JO, et al. Interpretation of the
presence of 6-monoacetylmorphine in the absence of morphine-3-
glucuronide in urine samples: evidence of heroin abuse. Ther Drug
Monit. 2003;25:645–648.
62. Rook EJ, Hillebrand MJ, Rosing H, et al. The quantitative analysis
of heroin, methadone and their metabolites and the simultaneous
detection of cocaine, acetylcodeine and their metabolites in human
plasma by high-performance liquid chromatography coupled with
tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed
Life Sci. 2005;824:213–221.
63. Jenkins AJ, Keenan RM, Henningfield JE, et al. Pharmacokinetics
and pharmacodynamics of smoked heroin. J Anal Toxicol. 1994;
18:317–330.
64. Reiter A, Hake J, Meissner C, et al. Time of drug elimination in
chronic drug abusers. Case study of 52 patients in a ‘‘low-step’’
detoxification ward. Forensic Sci Int. 2001;119:248–253.
65. Smith ML, Shimomura ET, Summers J, et al. Urinary excretion
profiles for total morphine, free morphine, and 6-acetylmorphine
following smoked and intravenous heroin. J Anal Toxicol. 2001;
25:504–514.
66. Skarke C, Schmidt H, Geisslinger G, et al. Pharmacokinetics of
morphine are not altered in subjects with Gilbert’s syndrome. Br J
Clin Pharmacol. 2003;56:228–231.
67. Sindrup SH, Brosen K, Bjerring P, et al. Codeine increases pain
thresholds to copper vapor laser stimuli in extensive but not poor
metabolizers of sparteine. Clin Pharmacol Ther. 1990;48:686–693.
68. Yue QY, Alm C, Svensson JO, et al. Quantification of the O- and
N-demethylated and the glucuronidated metabolites of codeine
relative to the debrisoquine metabolic ratio in urine in ultrarapid,
rapid, and poor debrisoquine hydroxylators. Ther Drug Monit.
1997;19:539–542.
69. Kathiramalainathan K, Kaplan HL, Romach MK, et al. Inhibition
of cytochrome P450 2D6 modifies codeine abuse liability. J Clin
Psychopharmacol. 2000;20:435–444.
70. Yue QY, Sawe J. Different effects of inhibitors on the O- and N-
demethylation of codeine in human liver microsomes. Eur J Clin
Pharmacol. 1997;52:41–47.
71. Caraco Y, Tateishi T, Guengerich FP, et al. Microsomal codeine N-
demethylation: cosegregation with cytochrome P4503A4 activity.
Drug Metab Dispos. 1996;24:761–764.
72. Court MH, Krishnaswamy S, Hao Q, et al. Evaluation of 30-azido-
30-deoxythymidine, morphine, and codeineas probe substrates for
UDP-glucuronosyltransferase 2B7 (UGT2B7) in human liver
microsomes: specificity and influence of the UGT2B7*2 polymorph-
ism. Drug Metab Dispos. 2003;31:1125–1133.
Ther Drug Monit � Volume 28, Number 3, June 2006 Toxicokinetics of Drugs of Abuse
r 2006 Lippincott Williams & Wilkins 453

Mais conteúdos dessa disciplina