4-Dehydrodebrisoquine , a Novel Debrisoquine Metabolite Formed from 4-Hydroxydebrisoquine That Affects the CYP 2 D 6 Metabolic Ratio

Considerable unexplained intersubject variability in the debrisoquine metabolic ratio (urinary debrisoquine/4-hydroxydebrisoquine) exists within individual CYP2D6 genotypes. We speculated that debrisoquine was converted to as yet undisclosed metabolites. Thirteen healthy young volunteers, nine CYP2D6*1 homozygotes [extensive metabolizers (EMs)] and four CYP2D6*4 homozygotes [poor metabolizers (PMs)] took 12.8 mg of debrisoquine hemisulfate by mouth and collected 0to 8and 8to 24-h urines, which were analyzed by gas chromatography-mass spectrometry (GCMS) before and after treatment with -glucuronidase. Authentic 3,4-dehydrodebrisoquine was synthesized and characterized by GCMS, liquid chromatography-tandem mass spectrometry, and H NMR. 3,4-Dehydrodebrisoquine is a novel metabolite of debrisoquine excreted variably in 0to 24-h urine, both in EMs (3.1–27.6% of dose) and PMs (0–2.1% of dose). This metabolite is produced from 4-hydroxydebrisoquine in vitro by human and rat liver microsomes. A previously unstudied CYP2D6*1 homozygote was administered 10.2 mg of 4-hydroxydebrisoquine orally and also excreted 3,4-dehydrodebrisoquine. EMs excreted 6-hydroxydebrisoquine (0–4.8%) and 8-hydroxydebrisoquine (0–1.3%), but these phenolic metabolites were not detected in PM urine. Debrisoquine and 4-hydroxydebrisoquine glucuronides were excreted in a highly genotype-dependent manner. A microsomal activity that probably does not involve cytochrome P450 participates in the further metabolism of 4-hydroxydebrisoquine, which we speculate may also lead to the formation of 1and 3-hydroxy-debrisoquine and their ringopened products. In conclusion, this study suggests that the traditional metabolic ratio is not a true measure of the debrisoquine 4-hydroxylation capacity of an individual and thus may, in part, explain the wide intragenotype variation in metabolic ratio. Debrisoquine (3,4-dihydro-2(1H)-isoquinoline carboxamidine) sulfate was patented in the United States by Hoffmann-La Roche in 1964 (Wenner, 1964) and immediately went into clinical trials as an antihypertensive agent (Talbot, 1965; Rosendorff et al., 1968; Somers et al., 1968; Blechman et al., 1969). Surprisingly, at the time of its launch, little, if anything was known about the metabolic disposition of debrisoquine. Workers at Hoffmann-La Roche UK (Allen et al., 1975) reported on a study in which C-labeled debrisoquine was administered to rats (50 mg/kg) and a single hypertensive patient (2.6 mg, on top of 15 mg q.d.s. therapeutic dose). Debrisoquine was excreted unchanged in urine of both human and rat, together with 4-hydroxydebrisoquine as the major metabolite, and traces of the phenolic metabolites 5-, 6-, 7-, and 8-hydroxydebrisoquine. In addition, both rats and humans excrete 10 to 15% of the dose as two ring-opened acidic metabolites, presumed to arise from hydroxylation of debrisoquine in positions 1 and 3 (Fig. 1A). The nature of these metabolites was subsequently confirmed (Allen et al., 1976; Eiermann et al., 1998). For the rat, 70% of the administered radioactivity could be accounted for as debrisoquine and these seven aforementioned metabolites. In the single patient who was studied, 66% of the radioactivity was similarly accounted for (Allen et al., 1975). It was noted that there was considerable variation (7-fold) in the extent of urinary excretion of unchanged debrisoquine in 10 volunteers given 40 mg of debrisoquine sulfate by mouth and that urinary debrisoquine excretion correlated positively with hypotensive response to debrisoquine (Angelo et al., 1975). This same group performed a second C-debrisoquine study in four human volunteers, for which 71.3 to 76.7% of the radioactivity was recovered in the urine within 24 h (Angelo et al., 1976). These authors identified debrisoquine and 4-hydroxydebrisoquine in human urine, and tentatively Supported by the National Cancer Institute Intramural Research Program (F.J.G.). J.R.I. is grateful to U.S. Smokeless Tobacco Company for a grant for collaborative research. Article, publication date, and citation information can be found at http://dmd.aspetjournals.org. doi:10.1124/dmd.105.008920. ABBREVIATIONS: MR, metabolic rate; EM, extensive metabolizer; IM, intermediate metabolizer; P450, cytochrome P450; PM, poor metabolizer; UM, ultrarapid metabolizer; GCMS, gas chromatography-mass spectrometry; LC-MS/MS, liquid chromatography-tandem mass spectrometry; UPLC-TOFMS, ultra-performance liquid chromatography-coupled time-of-flight mass spectrometry. 0090-9556/06/3409-1563–1574 DRUG METABOLISM AND DISPOSITION Vol. 34, No. 9 U.S. Government work not protected by U.S. copyright 8920/3134741 DMD 34:1563–1574, 2006 Printed in U.S.A. 1563 at A PE T Jornals on Sptem er 0, 2017 dm d.aspurnals.org D ow nladed from

[1]  I. Wilson,et al.  High resolution "ultra performance" liquid chromatography coupled to oa-TOF mass spectrometry as a tool for differential metabolic pathway profiling in functional genomic studies. , 2005, Journal of proteome research.

[2]  Y. Funae,et al.  Effect of genetic polymorphism on the metabolism of endogenous neuroactive substances, progesterone and p-tyramine, catalyzed by CYP2D6. , 2004, Brain research. Molecular brain research.

[3]  J. Idle,et al.  Polymorphic Cytochrome P450 2D6: Humanized Mouse Model and Endogenous Substrates , 2004, Drug metabolism reviews.

[4]  P. Philip,et al.  Ifosfamide plasma clearance in relation to polymorphic debrisoquine oxidation , 2004, Cancer Chemotherapy and Pharmacology.

[5]  G. Shenfield,et al.  Lack of relationship between tolbutamide metabolism and debrisoquine oxidation phenotype , 2004, European Journal of Clinical Pharmacology.

[6]  M. Eichelbaum,et al.  CYP2D6 genotyping strategy based on gene copy number determination by TaqMan real‐rime PCR , 2003, Human mutation.

[7]  W. Steimer,et al.  Optimized strategy for rapid cytochrome P450 2D6 genotyping by real-time long PCR. , 2003, Clinical chemistry.

[8]  J. Idle,et al.  Screening for endogenous substrates reveals that CYP2D6 is a 5-methoxyindolethylamine O-demethylase. , 2003, Pharmacogenetics.

[9]  W. Koch,et al.  Comparison of two CYP2D6 genotyping methods and assessment of genotype-phenotype relationships. , 2003, Clinical chemistry.

[10]  J. Idle,et al.  The Relative Contribution of Monoamine Oxidase and Cytochrome P450 Isozymes to the Metabolic Deamination of the Trace Amine Tryptamine , 2003, Journal of Pharmacology and Experimental Therapeutics.

[11]  Ulrich M. Zanger,et al.  Cytochrome P450 2D6: overview and update on pharmacology, genetics, biochemistry , 2003, Naunyn-Schmiedeberg's Archives of Pharmacology.

[12]  D. Greenblatt,et al.  Effect of zolpidem on human Cytochrome P450 activity, and on transport mediated by P‐glycoprotein , 2002, Biopharmaceutics & drug disposition.

[13]  J. Idle,et al.  4-Hydroxylation of debrisoquine by human CYP1A1 and its inhibition by quinidine and quinine. , 2002, The Journal of pharmacology and experimental therapeutics.

[14]  M. Galteau,et al.  Short communication. The effects of drinking and smoking on the CYP2D6 metabolic capacity. , 2000, Drug metabolism and disposition: the biological fate of chemicals.

[15]  S D Hall,et al.  Regioselective and stereoselective metabolism of ibuprofen by human cytochrome P450 2C. , 1997, Biochemical pharmacology.

[16]  J. Brockmöller,et al.  Cytochrome P450 2D6 variants in a Caucasian population: allele frequencies and phenotypic consequences. , 1997, American journal of human genetics.

[17]  C. Bonfils,et al.  Oxidative metabolism of zolpidem by human liver cytochrome P450S. , 1995, Drug metabolism and disposition: the biological fate of chemicals.

[18]  J. Idle,et al.  Genetic and metabolic criteria for the assignment of debrisoquine 4-hydroxylation (cytochrome P4502D6) phenotypes. , 1991, Pharmacogenetics.

[19]  D. Nebert,et al.  Characterization of the common genetic defect in humans deficient in debrisoquine metabolism , 1988, Nature.

[20]  L. Bertilsson,et al.  Amitriptyline metabolism: Association with debrisoquin hydroxylation in nonsmokers , 1986, Clinical pharmacology and therapeutics.

[21]  J. Poirier,et al.  ECOGENETICS OF PARKINSON'S DISEASE: 4-HYDROXYLATION OF DEBRISOQUINE , 1985, The Lancet.

[22]  M. Hetzel,et al.  Metabolic oxidation phenotypes as markers for susceptibility to lung cancer , 1984, Nature.

[23]  L. Bertilsson,et al.  Nortriptyline and debrisoquine hydroxylations in Ghanaian and Swedish subjects , 1984, Clinical pharmacology and therapeutics.

[24]  W. Kalow,et al.  Comparative pharmacogenetics of sparteine and debrisoquine , 1983, Clinical pharmacology and therapeutics.

[25]  J. Idle,et al.  Genetic polymorphism of phenformin 4‐hydroxylation , 1982, Clinical pharmacology and therapeutics.

[26]  L. Bertilsson,et al.  E‐ and Z‐10‐hydroxylation of nortriptyline: Relationship to polymorphic debrisoquine hydroxylation , 1981, Clinical pharmacology and therapeutics.

[27]  J. Idle,et al.  A study of the debrisoquine hydroxylation polymorphism in a Nigerian population. , 1980, Xenobiotica; the fate of foreign compounds in biological systems.

[28]  J. Idle,et al.  Debrisoquin hydroxylation polymorphism among Ghanaians and Caucasians , 1979, Clinical pharmacology and therapeutics.

[29]  J. Idle,et al.  The metabolism of [14C]-debrisoquine in man. , 1979, British journal of clinical pharmacology.

[30]  J. Idle,et al.  POLYMORPHIC HYDROXYLATION OF DEBRISOQUINE IN MAN , 1977, The Lancet.

[31]  L. Dring,et al.  The metabolism of debrisoquine in rat and man. , 1976, Biochemical Society transactions.

[32]  Cockcroft Dw,et al.  Prediction of Creatinine Clearance from Serum Creatinine , 1976 .

[33]  A. Brown,et al.  Metabolism of debrisoquine sulphate in rat, dog and man. , 1976, Xenobiotica; the fate of foreign compounds in biological systems.

[34]  A. Latham,et al.  Proceedings: A correlation between the response to debrisoquine and the amount of unchanged drug excreted in the urine. , 1975, British journal of pharmacology.

[35]  J. Haigh,et al.  Metabolism of debrisoquine sulfate. Identification of some urinary metabolites in rat and man. , 1975, Drug metabolism and disposition: the biological fate of chemicals.

[36]  T. Goehl,et al.  Gas-liquid chromatographic electron capture determination of some monosubstituted guanido-containing drugs. , 1975, Analytical chemistry.

[37]  M. Moser,et al.  Debrisoquin sulfate: a clinical evaluation of a new antihypertensive agent. , 1969, Current therapeutic research, clinical and experimental.

[38]  W. I. Cranston,et al.  Clinical evaluation of debrisoquin in the treatment of hypertension. , 1968, Archives of internal medicine.

[39]  K. Somers,et al.  A Clinical Evaluation of Debrisoquine Sulphate , 1968, The British journal of clinical practice.

[40]  F. Talbot Clinical evaluation of the antihypertensive effects of debrisoquin, a new postganglionic vasodepressor agent. , 1965, The Medical annals of the District of Columbia.

[41]  E. Boyland Detoxication Mechanisms , 1948, Nature.