Journal of Clinical Trials

Journal of Clinical Trials
Open Access

ISSN: 2167-0870

Editorial - (2016) Volume 6, Issue 1

Altered Expression/Activity of Cytochrome P450 (CYP) 3A4 Enzyme: Implications in Drug Safety and Efficacy

Romi Ghose*
Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 1441 Mousund Street (TMC), Room 517, Houston, TX 77030, USA
*Corresponding Author: Romi Ghose, Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 1441 Mousund Street (TMC), Room 517, Houston, TX 77030, USA, Tel: 713-795-8343, Fax: 832-842-8305 Email:

Editorial

Cytochrome P450 (CYP) 3A4 enzyme alterations during diseases. Transcriptional or post-transcriptional down-regulation of hepatic CYP enzymes is a characteristic feature of many diseases including infections; cancer, cardiovascular diseases and liver disorders [1-5]. This disrupts CYP-mediated drug metabolism and clearance in these patients. The major drug metabolizing enzyme (DME) in human liver is CYP3A4, which metabolizes ~60% of drugs [6,7]. Recent studies in pediatric patients show that critical illness is the primary determinant of the clearance of the CYP3A4 substrate, midazolam [8]. Furthermore, diabetic kidney transplant patients had low metabolism of the immunosuppressant, cyclosporine A, likely due to reduced CYP3A4 expression [9]. These clinical observations warrant further studies to understand the molecular mechanism by which human CYP enzymes are down-regulated during diseases.

Role of CYP3A4 enzyme in herb-drug interactions. Furthermore, changes in CYP3A4 expression/activity has been shown to be associated with herb-drug interactions. According to recent epidemiological reports, ~40% of Americans use CAM “natural products” including herbal medicines, botanicals, and other dietary supplements during their lifetime [10-14]. These supplements are often self-administered primarily to manage side effects of drugs and/or to improve overall physical and mental health along with therapeutic drugs. Moreover, patients diagnosed with HIV or cancer exhibit a higher CAM use; concomitant use of CAM and prescription medications have been reported in 70%-90% HIV and cancer patients [15-17]. Natural products/herbs are complex mixtures of many molecular entities. Both the putative active ingredient(s) and other constituents present in that mixture have the potential to interact with various classes of drugs, which may lead to dangerous clinical consequences.

A majority of these interactions has been attributed to CYP3A4 enzyme. For example, the herbal compound St. John’s wort induced CYP3A4, which decreased the oral bioavailability of a number of therapeutic drugs, including anti-depressants, anti-HIV agents, anticancer drugs, leading to failure of therapy [18-20]. Studies in animal models, humans or cell culture have shown that herbal compounds such as licorice, ginkgo, echinacea, quercetin, etc. significantly induced CYP3A expression/activity, leading to altered therapeutic potency of co-administered medications [16-20].

Regulation of CYP3A4 gene expression. Changes in CYP3A4 enzymes cause accumulation of the parent compound or the metabolite, which increases the risk of drug-drug interactions and adverse drug reactions (ADRs) in individual patients. A meta-analysis of 39 prospective studies revealed that ~2 million cases of hospitalization and ≥ 100,000 deaths/year can be attributed to ADRs in U.S.21 ADRs cause ~3%-6% of all hospital admissions and ~15% of hospitalized patients experience a serious adverse reaction to drugs [21-23]. Liver injury is one of the major adverse effects associated with drugs and accounts for ~50% of the cases of acute liver failure in this country [24].

Altered drug metabolism in diseases is associated with the induction of inflammatory responses. For example, CYP-mediated drug metabolism is disrupted and inflammatory markers are induced in patients undergoing treatment for several diseases including rheumatoid arthritis, cancer, organ transplantation, liver disorders [1-5]. Thus, treatment of rodents or liver cells with inflammatory mediators such as cytokines or lipopolysaccharide (LPS) has been widely used to investigate down-regulation of CYP enzymes in the liver [25-27].

CYP3A4 gene expression is regulated by basal transcription factors, and can be induced by diverse chemicals which bind to and activate nuclear receptors (NRs) [28,29]. The mouse xenobiotic NR, pregnane X receptor (PXR) is activated by the ligand, pregnenolone-16α- carbonitrile, PCN. Activated PXR binds to conserved sequences in the proximal promoter and the distal xenobiotic-responsive enhancer module (XREM) in the CYP3A4 gene [28,29]. The XREM is located -7.2 kb and -7.8 kb upstream of the transcription initiation site of CYP3A4. It contains PXR-responsive elements (PXREs) and works cooperatively with a PXRE located within the proximal promoter [28,29]. Basal and inducible expression of CYP3A4 gene is also regulated by the orphan NR, hepatocyte nuclear factor (HNF4)α [30]. NRs activate their target genes by recruiting or exchanging coactivators with the corepressor complex to the chromatins [31-32]. Chromatin remodeling enzymes catalyze histone modification to allow the binding of transcription factors.

CYP3A4 regulation is also modulated by a broad variety of kinases which phosphorylate the regulatory NRs or their associated proteins. For example, casein kinase (CK) 2-dependent phosphorylation of the heat shock protein 90β (Hsp90β; which binds to and retains PXR in the cytoplasm) was required for induction of a PXR target gene, multidrug resistance (MDR)1 by the prototypical human PXR (hPXR) ligand, Rifampicin (RIF) [33-35]. Furthermore, the mitogen-activated protein kinase (MAPK), c-JUN N-terminal kinase (JNK) was required for CYP3A4 induction by the vitamin D receptor (VDR) ligand, 1,25- Dihydroxyvitamin D(3) (1,25(OH)(2)D(3)) [36].

Interestingly, phosphorylation was shown to interfere with PXR’s subcellular localization, dimerization, DNA binding, and co-regulator interactions, leading to inhibition of its transcriptional activity [33-35]. Kinases such as cyclic-AMP dependent protein kinase A (PKA), PKC, cyclin dependent kinases, p70 S6K (70-kDa form of S6 kinase), Ca2+/ calmodulin-dependent protein kinase II (CaMKII), serine/threonine kinase DYRK2 can phosphorylate PXR, leading to CYP3A4 inhibition [33-35]. PXR also undergoes additional post-translational modification (PTM)s including ubiquitination, acetylation and SUMOylation, which can impact gene induction by PXR [33-35]. Crosstalk between these PTMs has been reported to affect function of the transcriptional regulators, NF-κB and p53, and it was shown that phosphorylation at Ser 350 affected PXR acetylation [33-35]. Thus, protein phosphorylation may impact multiple PTMs in one or more proteins, and this complex regulatory mechanism may be responsible for CYP3A4 induction by PXR.

Regulation of CYP3A4 enzyme by microRNAs (miRNAs). Posttranscriptional or transcriptional changes in CYP3A4 may be mediated by miRNAs directly targeting the 3’-untranslated region (3’UTR) of CYP3A4 mRNA and indirectly targeting the 3’UTR of NR mRNAs, respectively [37-39]. The involvement of miRNAs in the regulation of CYP enzymes and NRs indicates potential role of miRNAs in the integrated response of cells to drugs and toxins. miRNAs are 19 to 25- nucleotide RNAs that bind to complementary sequences in the 3′-UTR of mRNAs. This recruits a RNA-induced silencing complex to mRNAs to repress protein translation, cleave targeted messages, and degrade mRNAs [37-39]. The broadly conserved miR-27b has been shown to target the 3′UTR of CYP3A4 in vitro [38].

Thus, induction or down-regulation of CYP3A4 enzyme can alter the metabolism and clearance of one or more medications in individual patients. These patients will be at a higher risk of undesirable effects of medications. Thus, alteration of CYP3A4- mediated drug metabolism should be a major consideration in developing/implementing treatment regimens for individual patients.

References

  1. Chang KC, Bell TD, Lauer BA, Chai H (1978) Altered theophylline pharmacokinetics during acute respiratory viral illness. Lancet 1:1132-1133.
  2. Haas CE, Kaufman DC, Jones CE, Burstein AH, Reiss W (2003) Cytochrome P450 3A4 activity after surgical stress. Crit Care Med 31: 1338-1346.
  3. Frye RF, Schneider VM, Frye CS, Feldman AM (2002) Plasma levels of TNF-alpha and IL-6 are inversely related to cytochrome P450-dependent drug metabolism in patients with congestive heart failure. J Card Fail 8: 315-319.
  4. Rivory LP, Slaviero KA, Clarke SJ (2002) Hepatic cytochrome P450 3A drug metabolism is reduced in cancer patients who have an acute-phase response. Br J Cancer 87: 277-280.
  5. George J, Liddle C, Murray M, Byth K, Farrell GC (1995) Pre-translational regulation of cytochrome P450 genes is responsible for disease-specific changes of individual P450 enzymes among patients with cirrhosis. BiochemPharmacol 49: 873-881.
  6. Nelson DR, Koymans L, Kamataki T, Stegeman JJ, Feyereisen R, et al. (1996) P450 superfamily: update on new sequences, gene mapping, accession numbers and nomenclature. Pharmacogenetics 6: 1-42.
  7. Guengerich FP (2008) Cytochrome p450 and chemical toxicology. Chem Res Toxicol 21: 70-83.
  8. Ince I, de Wildt SN, Peeters MY, Murry DJ, Tibboel D, et al. (2012) Critical illness is a major determinant of midazolam clearance in children aged 1 month to 17 years. Ther Drug Monit 34: 381-389.
  9. Akhlaghi F, Dostalek M, Falck P, Mendonza AE, Amundsen R, et al. (2012) The concentration of cyclosporine metabolites is significantly lower in kidney transplant recipients with diabetes mellitus. Ther Drug Monit 34: 38-45.
  10. Tindle HA, Davis RB, Phillips RS, Eisenberg DM (2005) Trends in use of complementary and alternative medicine by US adults: 1997-2002. AlternTher Health Med 11: 42-49.
  11. Lee LS, Andrade AS, Flexner C (2006) Interactions between natural health products and antiretroviral drugs: pharmacokinetic and pharmacodynamic effects. Clin Infect Dis 43: 1052-1059.
  12. Windrum P, Hull DR, Morris TC (2000) Herb-drug interactions. Lancet 355: 1019-1020.
  13. Hu Z, Yang X, Ho PC, Chan SY, Heng PW, et al. (2005) Herb-drug interactions: a literature review. Drugs 65: 1239-1282.
  14. Risa KJ, Nepon L, Justis JC, Panwalker A, Berman SM, et al. (2002) Alternative therapy use in HIV-infected patients receiving highly active antiretroviral therapy. Int J STD AIDS 13: 706-713.
  15. Meijerman I, Beijnen JH, Schellens JH (2006) Herb-drug interactions in oncology: focus on mechanisms of induction. Oncologist 11: 742-752.
  16. Goey AK, Mooiman KD, Beijnen JH, Schellens JH, Meijerman I (2013) Relevance of in vitro and clinical data for predicting CYP3A4-mediated herb-drug interactions in cancer patients. Cancer Treat Rev 39: 773-783.
  17. Lau WC, Welch TD, Shields T, Rubenfire M, Tantry US, et al. (2011) The effect of St John's Wort on the pharmacodynamic response of clopidogrel in hyporesponsive volunteers and patients: increased platelet inhibition by enhancement of CYP3A4 metabolic activity. J CardiovascPharmacol 57: 86-93.
  18. Roby CA, Anderson GD, Kantor E, Dryer DA, Burstein AH (2000) St John's Wort: effect on CYP3A4 activity. ClinPharmacolTher 67: 451-457.
  19. Markowitz JS, Donovan JL, DeVane CL, Taylor RM, Ruan Y, et al. (2003) Effect of St John's wort on drug metabolism by induction of cytochrome P450 3A4 enzyme. JAMA 290: 1500-1504.
  20. Lazarou J, Pomeranz BH, Corey PN (1998) Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA 279: 1200-1205.
  21. Brvar M, Fokter N, Bunc M, Mozina M (2009) The frequency of adverse drug reaction related admissions according to method of detection, admission urgency and medical department specialty. BMC clinical pharmacology 9: 8.
  22. Davies EC, Green CF, Taylor S, Williamson PR, Mottram DR, et al. (2009) Adverse drug reactions in hospital in-patients: a prospective analysis of 3695 patient-episodes. PLoS One 4: e4439.
  23. Aitken AE, Morgan ET (2007) Gene-specific effects of inflammatory cytokines on cytochrome P450 2C, 2B6 and 3A4 mRNA levels in human hepatocytes. Drug MetabDispos 35: 1687-1693.
  24. Aitken AE, Richardson TA, Morgan ET (2006) Regulation of drug-metabolizing enzymes and transporters in inflammation. Annu Rev PharmacolToxicol 46: 123-149.
  25. Ghose R, White D, Guo T, Vallejo J, Karpen SJ (2008) Regulation of hepatic drug-metabolizing enzyme genes by Toll-like receptor 4 signaling is independent of Toll-interleukin 1 receptor domain-containing adaptor protein. Drug MetabDispos 36: 95-101.
  26. Xie W (2009) Nuclear Receptors in Drug Metabolism. A John Wiley & Sons, USA.
  27. Bombail V, Taylor K, Gibson GG, Plant N (2004) Role of Sp, C/EBP alpha, HNF, and PXR in the basal- and xenobiotic-mediated regulation of the CYP3A4 gene. Drug MetabDispos 32: 525-535.
  28. Tirona RG, Lee W, Leake BF, Lan LB, Cline CB, et al. (2003) The orphan nuclear receptor HNF4alpha determines PXR- and CAR-mediated xenobiotic induction of CYP3A4. Nat Med 9: 220-224.
  29. Urnov FD, Wolffe AP (2001) A necessary good: nuclear hormone receptors and their chromatin templates. MolEndocrinol 15: 1-16.
  30. McKenna NJ, Lanz RB, O'Malley BW (1999) Nuclear receptor coregulators: cellular and molecular biology. Endocr Rev 20: 321-344.
  31. Elias A, High AA, Mishra A, Ong SS, Wu J, et al. (2014) Identification and characterization of phosphorylation sites within the pregnane X receptor protein. BiochemPharmacol 87: 360-370.
  32. Wang YM, Chai SC, Lin W, Chai X, Elias A, et al. (2015) Serine 350 of human pregnane X receptor is crucial for its heterodimerization with retinoid X receptor alpha and transactivation of target genes in vitro and in vivo. BiochemPharmacol 96: 357-368.
  33. Staudinger JL, Xu C, Biswas A, Mani S (2011) Post-translational modification of pregnane x receptor. Pharmacol Res 64: 4-10.
  34. Yasunami Y, Hara H, Iwamura T, Kataoka T, Adachi T (2004) C-jun N-terminal kinase modulates 1,25-dihydroxyvitamin D3-induced cytochrome P450 3A4 gene expression. Drug MetabDispos 32: 685-688.
  35. Gomez A, Ingelman-Sundberg M (2009) Epigenetic and microRNA-dependent control of cytochrome P450 expression: a gap between DNA and protein. Pharmacogenomics 10: 1067-1076.
  36. Pan YZ, Gao W, Yu AM (2009) MicroRNAs regulate CYP3A4 expression via direct and indirect targeting. Drug MetabDispos 37: 2112-2117.
  37. Ramamoorthy A, Skaar TC (2011) In silico identification of microRNAs predicted to regulate the drug metabolizing cytochrome P450 genes. Drug MetabLett 5: 126-131.
Citation: Ghose R (2016) Altered Expression/Activity of Cytochrome P450 (CYP) 3A4 Enzyme: Implications in Drug Safety and Efficacy. J Clin Trials 6:e123.

Copyright: © 2016 Ghose R. 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.
Top