Short Communication
Impact of CYP2D6 Ultra Rapid Metabolism on Therapeutic Drug Efficacy in Type 2 Diabetic Patients
A Pineda, N Williams, N Chaudhry, M Pietruszka, K Daldalyan, Y Valles-Ayoub, M Robinson*
Corresponding Author: Michael F Robinson, MD, Firmalab, CPI Director, Marina Diabetes Center, Marina del Rey, California
Received: April 30, 2020; Revised: August 19, 2020; Accepted: May 19, 2020
Citation: Pineda A, Williams N, Chaudhry N, Pietruszka M, Daldalyan K, et al. (2020) Impact of CYP2D6 Ultra Rapid Metabolism on Therapeutic Drug Efficacy in Type 2 Diabetic Patients. J Genet Cell Biol, 3(2): 174-179.
Copyrights: ©2020 Robinson M, Pineda A, Williams N, Chaudhry N, Pietruszka M, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Share :
  • 950

    Views & Citations
  • 10

    Likes & Shares

Duplication of the CYP2D6 allele results in excess enzyme activity and can alter the effects of administered drugs. This ultra-rapid metabolizer phenotype and its impact on conventional treatment of Type 2 Diabetes Mellitus is the focus of the present communication. Genomic DNA was extracted from buccal swab samples taken from 215 patients and analyzed for CYP2D6 activity. Results indicated that 22 patients (10.2%) were positive for CYP2D6 duplication. Of these 22 ultra-rapid metabolizers, 12 patients (54.5%) were poor responders to conventional Type 2 Diabetes treatment. These results may reflect the small population size used in the study. A larger sample with a focus on the mechanism of drug metabolism in the future may elucidate a more causal relationship between CYP2D6 duplication and optimized therapeutic outcome of conventional Type 2 Diabetes treatment regimens.

 

Keywords: Cytochrome P450, CYP2D6, Ultra rapid metabolizers, Diabetes, Pharmacogenetics

 

Abbreviations: CYP450: Cytochrome P450; CYP2D6: Debrisoquine 4-hydroxylase; EM: Extensive Normal; UM: Ultra-rapid metabolizer; PM: Poor metabolizer; T2D: Type 2 Diabetes

INTRODUCTION

Cytochrome P450 (CYP450) enzymes are essential for the metabolism of drugs. They account for around 75 percent of enzymes involved in drug metabolism [1]. An important cytochrome P450 (CYP) enzyme, CYP2D6 (debrisoquine 4-hydroxylase), represents 1-5% of the CYP liver content and is responsible for the hydroxylation or de-alkylation of up to 25% of commonly prescribed drug classes drugs such as analgesics, anticonvulsants, antidepressants, antipsychotics, opioids, antiarrhythmics and tamoxifen, many of which have a narrow therapeutic window [2-5].

CYP2D6 is highly polymorphic and allele variations alter the enzyme’s rate of drug metabolism [6]. These differing rates of metabolism are categorized into the following phenotypes: extensive normal (EM), ultra-rapid metabolizers (UM), intermediate metabolizers (IM) and poor metabolizers (PM) [7]. Poor metabolizers have a complete or near-complete loss of CYP2D6 function while ultra-rapid metabolizers have multiple copies of the gene and therefore metabolize their substrates ultra-rapidly [8]. These changes in metabolism may cause alterations to the therapeutic effect of the drugs being metabolized and the impact can be clinically significant [9-11]. For example, a poor metabolizer is unable to convert codeine into its biologically active form, morphine, thereby eliminating its therapeutic effect [12]. On the other side of the spectrum, giving a normal dose of an opioid to an ultra-rapid metabolizer could result in excessive drug effect and increased activity of the CYP2D6 variants thus resulting in toxic levels of potent metabolites of the parent drug [13] and accumulation of drug in breast milk [14,15].

A compilation of data from across the world concerning CYP2D6 predicted PM prevalence to be between 0.4-5.4%, IM between 0.4 and 11%, EM between 67-90% and UM between 1 to 21% [16]. In addition, there were significant variations of allele prevalence between ethnic groups [17,18]. Prevalence variability across race groups specifically of UMs has been well studied. Some notable UM prevalence’s include Ethiopians (29%) [19], Saudi Arabians (21%) [20-22], Spaniards (10%) [23], Turkish (5.6%) [24], African Americans (4.9%) [25-28] Caucasion Americans (4.3%) [25], Croatians (4%) [29], Colombians (1.9%) [30], European white populations (0.8-1.0%) [31-33], and Chinese (0.9%) [34,35].

CYP2D6 duplication in relation to Type 2 Diabetes Mellitus (T2D) is not well understood. T2D is a major public health concern with prevalence reaching 8.5% in adults in the United States [36]. The International Diabetes Federation estimates that, worldwide, 425 million adults were living with diabetes mellitus in 2017, with about 90% of those cases being Type 2 [37]. Projections report that this number may reach 629 million by 2045 [37]. It has been demonstrated that T2D has the ability to alter CYP450 enzyme activity, which in turn impacts the patient’s response to treatment [38,39]. Understanding the role of CYP2D6 duplication in patients with T2D may be useful for personalized treatment and improvement of therapeutic outcomes [40,41].

MATERIALS AND METHODS

Nucleic acid isolation

Buccal epithelial cells were obtained with the Hydra Flock 6" Sterile Elongated Flock Swab w/Plastic Handle & Dry Transport Tube (Puritan Medical Products, Glendora, United States, Cat. #25-3606-H BT.) Cellular DNA was isolated using the Quick-DNA Miniprep Plus Kit (Zymo Research, Irvine, United States, Cat. # D4068) following manufacturer’s instructions. DNA optical density was measured using the ND-1000 Spectrophotometer (Thermo Fisher Scientific, Waltham, United States) for concentration determination. After DNA quantitation, DNA was diluted using RNAse free H2O to a concentration of 20 ng/μl. Isolated DNA was stored at -20°C as necessary.

PCR

To identify individuals with a duplicated CYP2D6 gene, nucleic acid is amplified using primers designed to amplify a 3200 bp segment (CYP-17f: 5’-TCCCCCACTGACCCAACTCT-3’) CYP-32r: 5’-CACGTGCAGGGCACCTAGAT-3’). Patient samples are amplified alongside a positive control obtained from Coriell in a Techne TC-412 Thermal Cycler. Each 12.5 μl reaction was comprised of 7.02 μl RNAse free H2O, 1.25 μl 10X Long Range PCR buffer, 0.63 μl 10 mM dNTP mix, 1 μl 10 μmol of primer mix (CYP207F/32R), 0.1 μl Qiagen Long Range enzyme, and 2.5 μl (50ng total) DNA. The thermocycler program consists of a 3 minute enzyme activation step at 93°C, followed by 35 PCR cycles, each of which included three steps: denaturation of DNA template and primers for 15 seconds at 93°C, annealing of primers to single-stranded DNA template for 30 s at 62°C, and extension of amplicon strand (complementary to DNA template strand) for 5 min at 68°C. The PCR included a final holding temperature at 4°C.

Gel electrophoresis and imaging

Each gel was electrophoresed with 3 μl Kapa Express DNA Ladder kit (Kapa Biosystems, Boston, United States) as a molecular weight marker. Kapa loading dye (6X) was added to each PCR tube containing a total reaction volume of 12.5 μl. 10 μl of each amplicon was loaded into a 1% Lonza Reliant Minigel TAE (Lonza Group AG, Basel, Switzerland, Cat. #54801) and electrophoresed at 90V for 30 minutes. The gels were stained in a 25 μl Sybr®-Safe DNA gel stain (Thermo Fisher, Carlsbad, United States, Cat. #S33102) and 250 μl 1X TBE buffer bath. The gels were then viewed and imaged in a UVP Epi Chemi II Darkroom (UVP, United States) imaging system and analyzed for amplification of the duplicated CYP2D6 region. A positive signal for CYP2D6 duplication yields a 3200 bp band (Figure 1).


Sequencing analysis

The duplicated patients’ CYP2D6 exons 1, 2, 3, 4, 5, 6, 7, and 9 genotypes were analyzed for other variations via target exon sequencing. The target exons were Sanger sequenced using ABI 3130xl Genetic Analyzer (Thermo Fisher Scientific, Waltham, MA, USA) and cross-referenced to NCBI Reference Sequence NC_000022.10 [42], using Sequencher software (Gene Codes Corporation, Ann Arbor, MI, USA) for analysis [43].

RESULTS

Images obtained of the gel electrophoresis using UVP Epi Chemi Darkroom and Lab works Image Acquisition and Analysis software, identified 22 patients out of 215, positive for CYP2D6 duplication. A positive signal was denoted by a 3200 bp band found on the gel as denoted in Figure 1. This yielded a 10.2% rate of ultra-rapid metabolizers in a sample of 215 patients. Using further analysis, it was discovered that 54.5% of the 22 patients were also poor responders to conventional type 2 diabetes treatment regimens. Sequencing analysis revealed that 45% of the CYP2D6 duplicated patients also had at least one additional variation in conjunction with CYP2D6 duplication. These variations included CYP2D6*2, CYP2D6*3, CYP2D6*4, CYP2D6*10, CYP2D6*17, and CYP2D6*41. Five of the twenty-two (22.7%) CYP2D6 duplicated patients specifically had the CYP2D6*10 polymorphism (Figure 2).

 DISCUSSION

Our findings suggest that CYP2D6 genotyping of diabetic patients may have clinical significance in prescription planning with respect to therapeutic efficacy. To the extent that phenotypic expression of CYP activity corresponds to CYP genotype, it may be possible to design optimized therapeutic regimens for selective CYP substrates based on knowledge of a patient's CYP genotype.

CYP2D6 is fixed in the phospholipid bilayer of the endoplasmic reticulum. Switching between the opened and closed conformations allows for substrate access to the active site [45,], [46]. The active site of CYP2D6 is bordered by iron-protoporphyrin IX and lined by amino acid residues, which can bind numerous substrates with distinct features [46,47]. This accounts for the ability of CYP2D6 to metabolize a broad range of substrates. The substrates are typically lipophilic bases with a planar hydrophobic aromatic ring and a nitrogen atom [46]. Polymorphisms affecting polypeptide sequences that compose active sites change the enzyme’s ability to bind substrates and thereby affect observed metabolic phenotype. It is possible that a polymorphism that changes the binding pocket of the enzyme [45] combined with duplication of CYP2D6 can alter one’s ability to metabolize medications used in conventional diabetes treatments. Future studies should seek to understand which medications could be most impacted within diabetes treatment regimens by a duplication of CYP2D6. Additionally, a more in depth look at patient demographics such as age, sex, ethnicity and disease duration should be undertaken and presented in future studies. 

CONCLUSION

Testing for CYP2D6 for a more customized prescription planning is being used increasingly in clinical practice. This research has revealed a possible link between CYP2D6 duplication and treatment response in patients with Type 2 Diabetes Mellitus. Our findings suggest that CYP2D6 variations may alter metabolic responses to diabetes treatment. CYP2D6 duplication in conjunction with other CYP2D6 variations produce various phenotypes with ranging degrees of functionality. Following genetic analysis, it is important to determine the phenotype by referencing CPIC guidelines in order to adjust treatment accordingly [48]. It is necessary to confirm a causative link using larger multi-center prospective clinical studies. A larger clinical study would allow for analysis of CYP2D6 allelic variations and would improve the causational link between the ultra-rapid metabolizers and Type 2 Diabetes Mellitus treatment. Subsequently, the results of such studies can justify the use of actionable CYP2D6 testing to help better predict diabetic drug response. Furthermore, such clinical studies can be applied to numerous drugs with a narrow therapeutic range to identify patients at risk for drug metabolism inefficiency.


1.          Guengerich FP (2008) Cytochrome p450 and chemical toxicology. Chem Res Toxicol 21: 70-83.

2.          Zhou SF, Di YM, Chan E, Du YM, Chow VD, et al. (2008) Clinical pharmacogenetics and potential application in personalized medicine. Curr Drug Metab 9: 738-784.

3.          McConnachie L, Bodor M, Kowdley K, Levy A, Tung B, et al. (2004) Human Liver Cytochrome P450 2D6 Genotype, Full-length Messenger Ribonucleic Acid, and Activity Assessed with a Novel Cytochrome P450 2D6 Substrate. Clin Pharmacol Ther 75: 282-297.

4.          Zanger UM, Schwab M (2013) Cytochrome P450 enzymes in drug metabolism: Regulation of gene expression, enzyme activities and impact of genetic variation. Pharmacol Ther 138: 103-141.

5.          Teh LK, Bertilsson L (2012) Pharmacogenomics of CYP2D6: Molecular genetics, interethnic differences and clinical importance. Drug Metab Pharmacokinet 27: 55-67.

6.          Yang Y, Botton MR, Scott ER, Scott SA (2017) Sequencing the CYP2D6 gene: From variant allele discovery to clinical pharmacogenetic testing. Pharmacogenomics 18: 673-685.

7.          Ingelman-Sundberg M, Sim SC, Gomez A, Rodriguez-Antona C (2007) Influence of cytochrome P450 polymorphisms on drug therapies: Pharmacogenetic, pharmacoepigenetic and clinical aspects. Pharmacol Ther 116: 496-526.

8.          St Sauver JL, Olson JE, Roger VL, Nicholson WT, Black JL, et al. (2017) CYP2D6 phenotypes are associated with adverse outcomes related to opioid medications. Pharmgenomics Pers Med 10: 217-227.

9.          Martinez-Matilla M, Blanco-Verea A, Santori M, Ansede-Bermejo J, Ramos-Luis E, et al. (2019) Genetic susceptibility in pharmacodynamic and pharmacokinetic pathways underlying drug-induced arrhythmia and sudden unexplained deaths. Forensic Sci Int Genet 42: 203-212.

10.      MacLeod AK, McLaughlin LA, Henderson CJ, Wolf CR (2017) Application of mice humanized for CYP2D6 to the study of tamoxifen metabolism and drug-drug interaction with antidepressants. Drug Metab Dispos 45: 17-22.

11.      Bahar MA, Setiawan D, Hak E, Wilffert B (2017) Pharmacogenetics of drug-drug interaction and drug-drug-gene interaction: A systematic review on CYP2C9, CYP2C19 and CYP2D6. Pharmacogenomics 18: 701-739.

12.      Smith HS (2009) Opioid Metabolism. Mayo Clin Proc 84: 613-624.

13.      Cavallari LH, Johnson JA (2019) A case for genotype-guided pain management. Pharmacogenomics 20: 705-708.

14.      Madadi P, Koren G, Cairns J, Chitayat D, Gaedigk A, et al. (2007) Safety of codeine during breastfeeding: Fatal morphine poisoning in the breastfed neonate of a mother prescribed codeine. Can Fam Physician 53: 33-35.

15.      Ito S (2018) Opioids in breast milk: Pharmacokinetic principles and clinical implications. J Clin Pharmacol 58: S151-S163.

16.      Gaedigk A, Sangkuhl K, Whirl-Carrillo M, Klein T, Leeder JS, et al. (2017) Prediction of CYP2D6 phenotype from genotype across world populations. Genet Med 19: 69-76.

17.      Bernard S, Neville KA, Nguyen AT, Flockhart DA (2006) Interethnic differences in genetic polymorphisms of CYP2D6 in the U.S. population: Clinical implications. Oncologist 11: 126-135.

18.      Naranjo MG, Rodrigues-Soares F, Peñas-Lledó EM, Tarazona-Santos E, Fariñas H, et al. (2018) Interethnic variability in CYP2D6, CYP2C9, and CYP2C19 genes and predicted drug metabolism phenotypes among 6060 Ibero- and Native Americans: RIBEF-CEIBA consortium report on population pharmacogenomics. OMICS 22: 575-578.

19.      Aklillu E, Persson I, Bertilsson L, Johansson I, Rodrigues F, et al. (1996) Frequent distribution of ultrarapid metabolizers of debrisoquine in an ethiopian population carrying duplicated and multiduplicated functional CYP2D6 alleles. J Pharmacol Exp Ther 278: 441-446.

20.      Ingelman-Sundberg M, Oscarson M, McLellan RA (1999) Polymorphic human cytochrome P450 enzymes: An opportunity for individualized drug treatment. Trends Pharmacol Sci 20: 342-349.

21.      McLellan RA, Oscarson M, Seidegård J, Evans DA, Ingelman-Sundberg M, et al. (1997) Frequent occurrence of CYP2D6 gene duplication in Saudi Arabians. Pharmacogenetics 7: 187-191.

22.      Islam SI, Idle JR, Smith RL (1980) The polymorphic 4-hydroxylation of debrisoquine in a Saudi arab population. Xenobiotica 10: 819-825.

23.      Bernal ML, Sinues B, Johansson I, McLellan RA, Wennerholm A, et al. (1999) Ten percent of North Spanish individuals carry duplicated or triplicated CYP2D6 genes associated with ultrarapid metabolism of debrisoquine. Pharmacogenetics 9: 657-660.

24.      Petrović J, Pešić V, Lauschke VM (2019) Frequencies of clinically important CYP2C19 and CYP2D6 alleles are graded across Europe. Eur J Hum Genet 28: 88-94.

25.      London SJ, Daly AK, Leathart JB, Navidi WC, Carpenter CC, et al. (1997) Genetic polymorphism of CYP2D6 and lung cancer risk in African-Americans and Caucasians in Los Angeles County. Carcinogenesis 18: 1203-1214

26.      He N, Daniel HI, Hajiloo L, Shockley D (1999) Dextromethorphan O-demethylation polymorphism in an African-American population. E J Clin Pharmacol 55: 457-459.

27.      Marinac JS, Foxworth JW, Willsie SK (1995) Dextromethorphan polymorphic hepatic oxidation (CYP2D6) in healthy black American adult subjects. Ther Drug Monit 17: 120-124.

28.      Gaedigk A, Bradford LD, Marcucci KA, Leeder JS (2002) Unique CYP2D6 activity distribution and genotype-phenotype discordance in black Americans. Clin Pharmacol Ther 72: 76-89.

29.      Bozina N, Granić P, Lalić Z, Tramisak I, Lovrić M, et al. (2003) Genetic polymorphisms of cytochromes P450: CYP2C9, CYP2C19, and CYP2D6 in Croatian population. Croat Med J 44: 425-428.

30.      Isaza CA, Henao J, López AM, Cacabelos R (2000) Isolation, sequence and genotyping of the drug metabolizer CYP2D6 gene in the Colombian population. Methods Find Exp Clin Pharmacol 22: 695-705.

31.      Bathum L, Johansson I, Ingelman-Sundberg M, Hørder M, Brøsen K, et al. (1998) Ultrarapid metabolism of sparteine: frequency of alleles with duplicated CYP2D6 genes in a Danish population as determined by restriction fragment length polymorphism and long polymerase chain reaction. Pharmacogenetics 8: 119-123.

32.      Griese EU, Zanger UM, Brudermanns U, Gaedigk A, Mikus G, et al. (1998) Assessment of the predictive power of genotypes for the in-vivo catalytic function of CYP2D6 in a German population. Pharmacogenetics 8: 15-26.

33.      Dahl ML, Johansson I, Bertilsson L, Ingelman-Sundberg M, Sjöqvist F (1995) Ultrarapid hydroxylation of debrisoquine in a Swedish population. Analysis of the molecular genetic basis. J Pharmacol Exp Ther 274: 516-520.

34.      Johansson I, Oscarson M, Yue QY, Bertilsson L, Sjöqvist F, et al. (1994) Genetic analysis of the Chinese cytochrome P4502D locus: Characterization of variant CYP2D6 genes present in subjects with diminished capacity for debrisoquine hydroxylation. Mol Pharmacol 46: 452-459.

35.      Lou YC, Ying L, Bertilsson L, Sjöqvist F (1987) Low frequency of slow debrisoquine hydroxylation in a native Chinese population. Lancet 2: 852-853.

36.      Xu G, Liu B, Sun Y, Du Y, Snetselaar LG, et al. (2018) Prevalence of diagnosed type 1 and type 2 diabetes among US adults in 2016 and 2017: Population based study. BMJ 362: k1497.

37.      International Diabetes Federation (2017) IDF Diabetes Atlas.

38.      Cheng PY, Morgan ET (2001) Hepatic cytochrome P450 regulation in disease states. Curr Drug Metab 2: 165-183.

39.      Gravel S, Chiasson JL, Dallaire S, Turgeon J, Michaud V (2018) Evaluating the impact of type 2 diabetes mellitus on CYP450 metabolic activities: Protocol for a case-control. pharmacokinetic study BMJ Open 8.

40.      He ZX, Chen XW, Zhou ZW, Zhou SF (2015) Impact of physiological, pathological and environmental factors on the expression and activity of human cytochrome P450 2D6 and implications in precision medicine. Drug Metab Rev 47: 470-519.

41.      Tomalik-Scharte D (2008) Application of pharmacogenetics in dose individualization in diabetes, psychiatry, cancer and cardiology. EJIFCC 19: 54-61.

42.      p13 Primary Assembly (2013) Homo sapiens chromosome 22, GRCh37. Accessed on April 22, 2020.

43.      Valles-Ayoub Y, Esfandiarifard S, No D,Sinai P, Khokher Z, et al. (2011) Wolman Disease (LIPA p.G87V) Genotype Frequency in People of Iranian-Jewish Ancestry. Genet Test Mol Bioma 15: 395-398.

44.      NCBI (2020) National Center for Biotechnology Information. ClinVar [VCV000016893.3] Accessed on: April 23, 2020.

45.      Poulos TL, Finzel BC, Howard AJ (1987) High-resolution crystal structure of cytochrome P450cam. J Mol Biol 195: 687-700.

46.      ScienceDirect (2020) CYP2D6 pharmacogenomics. Accessed on April 28, 2020.

47.      Zhou SF, Liu JP, Lai XS (2009) Substrate specificity, inhibitors and regulation of human cytochrome P450 2D6 and implications in drug development. Curr Med Chem 16: 2661-2805.

48.      Clinical Pharmacokinetics Implementation Consortium (2020) Genes-Drugs.