Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET

User menu

  • My alerts
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Drug Metabolism & Disposition
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET
  • My alerts
  • Log in
  • Log out
  • My Cart
Drug Metabolism & Disposition

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Visit dmd on Facebook
  • Follow dmd on Twitter
  • Follow ASPET on LinkedIn
Research ArticleArticle

Expression of Hepatic Drug-Metabolizing Cytochrome P450 Enzymes and Their Intercorrelations: A Meta-Analysis

Brahim Achour, Jill Barber and Amin Rostami-Hodjegan
Drug Metabolism and Disposition August 2014, 42 (8) 1349-1356; DOI: https://doi.org/10.1124/dmd.114.058834
Brahim Achour
Manchester Pharmacy School, University of Manchester, Manchester, United Kingdom (B.A., J.B., A.R-H.), and Simcyp Limited, a Certara Company, Sheffield, United Kingdom (A.R-H.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jill Barber
Manchester Pharmacy School, University of Manchester, Manchester, United Kingdom (B.A., J.B., A.R-H.), and Simcyp Limited, a Certara Company, Sheffield, United Kingdom (A.R-H.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Amin Rostami-Hodjegan
Manchester Pharmacy School, University of Manchester, Manchester, United Kingdom (B.A., J.B., A.R-H.), and Simcyp Limited, a Certara Company, Sheffield, United Kingdom (A.R-H.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF + SI
  • PDF
Loading

Abstract

Cytochrome P450 is a family of enzymes that catalyze reactions involved in the metabolism of drugs and other xenobiotics. These enzymes are therefore important in pharmacologic and toxicologic studies, and information on their abundances is of value in the process of scaling in vitro data to in vivo metabolic parameters. A meta-analysis was applied to data on the abundance of human hepatic cytochrome P450 enzymes in Caucasian adult livers (50 studies). Despite variations in the methods used to measure the abundance of enzymes, agreement between the studies in 26 different laboratories was generally good. Nonetheless, some heterogeneity was detected (Higgins and Thompson heterogeneity test). More importantly, large interindividual variability was observed in the collated data. Positive correlations between the expression levels of some cytochrome P450 enzymes were found in the abundance data, including the following pairs: CYP3A4/CYP3A5*1/*3 (Rs = 0.70, P < 0.0001, n = 52), CYP3A4/CYP2C8 (Rs = 0.68, P < 0.0001, n = 134), CYP3A4/CYP2C9 (Rs = 0.55, P < 0.0001, n = 71), and CYP2C8/CYP2C9 (Rs = 0.55, P < 0.0001, n = 99). These correlations can be used to demonstrate common genetic transcriptional mechanisms.

Introduction

Cytochrome P450 enzymes constitute the main family of enzymes responsible for phase I metabolism reactions (Al Omari and Murry, 2007). The survey by Wienkers and Heath (2005) indicated the involvement of these enzymes in the metabolic clearance of nearly 75% of the 200 most prescribed therapeutic drugs in the United States in 2002. A report by Zanger et al. (2014) described the contribution of each drug-metabolizing cytochrome P450 enzyme to drug metabolism pathways, as well as the various genetic and nongenetic factors that affect variability in enzymatic activity and, therefore, the clinical outcome of therapy.

Many studies have attempted to quantify the abundance of cytochrome P450 enzymes and to describe the genetic and epigenetic factors that affect the expression of these enzymes. As indicated by Proctor et al. (2004), abundance data of cytochrome P450 can be used for extrapolation from in vitro data to in vivo pharmacokinetic parameters, particularly when constructing the population variability is of interest. Hence, these data can be used for simulation experiments involving virtual populations for the pharmacologic and toxicologic assessment of drugs (Rostami-Hodjegan and Tucker, 2007). Correlations of expression between enzymes have been reported in the literature (Jover et al., 2009), and although in the current framework of Monte-Carlo simulations for creating virtual populations (Rostami-Hodjegan and Tucker, 2007) these correlations are not considered, such correlations, if established, can assist in producing more reliable scenarios.

Many efforts have been made to quantify cytochrome P450 enzymes, and some of these studies attempted to assess correlations of expression. These quantitative data were obtained using a variety of methods [Western blotting, enzyme-linked immunosorbent assay (ELISA), or liquid chromatography in conjunction with mass spectrometry (LC-MS)] and using different numbers of subjects from different age groups and ethnic backgrounds.

Meta-analysis is a process of combining different studies to obtain an overall figure for a measured variable, detect the degree of heterogeneity between the individual studies, and describe inconsistency and its possible sources (Sánchez-Meca and Martín-Martínez, 1997). With the growing number of studies that quantify pharmacokinetically relevant enzymes, there is a pressing need for a meta-analysis of these data to identify the weighted mean abundance levels of the enzymes, the degree of variability in these abundance data, and the degree of heterogeneity between these studies. The most recent meta-analysis of abundance data of drug-metabolizing cytochrome P450 enzymes, published in 2004, analyzed data from 19 studies and was a comprehensive and influential analysis, even though it did not distinguish between CYP3A isoforms (Rowland-Yeo et al., 2004) (see Supplemental Fig. 1). Since 2004, an additional 31 important studies of cytochrome P450 abundance have been published; for this reason, we now present a meta-analysis of the literature on cytochrome P450 abundance published up to the beginning of 2014 and provide correlations of expression where data are available. This meta-analysis will assist different groups who work with physiologically based pharmacokinetic models to incorporate more representative values for the system parameters related to the expression levels of cytochrome P450 enzymes.

Materials and Methods

Collection of Data.

Two electronic databases, Medline (http://www.nlm.nih.gov/bsd/pmresources.html) and Web of Knowledge (http://wok.mimas.ac.uk/) (between the years 1980 and 2014), were searched for relevant literature on the abundance of enzymes by using appropriate key words (hepatic/liver cytochrome P450 abundance, hepatic/liver CYP abundance, correlation of expression). Other key words were also used (e.g., quantity, concentration, content, quantification, measurement) instead of abundance to widen the search scope. Resources cited by the collected articles were also inspected to locate additional literature that could be used. A set of predefined criteria were used for inclusion of identified studies. The selected studies were those conducted on individual liver microsomal samples (rather than pooled samples or cell lines), where absolute protein abundance was measured in Caucasian adults. In addition, studies quoting abundance data in arbitrary, relative, or nonstandard units (other than pmol mg−1) were excluded. Finally, studies that quantified mRNA levels or enzyme activities only were also excluded. The sources of data were identified to ensure that none of the data used were duplicated in the analysis. Where data were not quoted and graphs contained data points, GetData Graph Digitizer version 2.25 (available at: http://www.getdata-graph-digitizer.com/) was used to obtain abundance values.

Calculation of Weighted Means and Coefficients of Variation.

For each enzyme, abundance values were tested for heterogeneity and normality of distribution. The Higgins and Thompson heterogeneity test (Cochran, 1954; Higgins and Thompson, 2002; Higgins et al., 2003) determines whether the 50 studies are consistent with one another, whereas the normality test determines whether the combined data are normally distributed. The normality test was performed according to the method of Kolmogorov and Smirnov.

The data from the individual studies were combined. The weighted means (Embedded Image) and the weighted coefficients of variation (WCVs) of the abundance of the different enzymes from the collected studies were calculated using eqs. 1 and 2 (Armitage et al., 2001), respectively:Embedded Image(1)Embedded Image(2)where (Embedded Image) and WCV represent the weighted mean and weighted coefficient of variation, respectively. Subscript j indicates the study, nj the number of samples in study j, and Embedded Image the mean abundance of a particular enzyme in study j.

Assessment of Heterogeneity between Studies.

To assess the homogeneity between the means and coefficients of variation of individual studies and the overall mean and variability of the collated data, eqs. 3, 4, and 5 were used:Embedded Image(3)Embedded Image(4)Embedded Image(5)where wj is the weight of study j expressed as the variance and calculated using eq. 4 (the inverse of standard deviation, sdj, squared), (Embedded Image) is the variance in the weighted mean of the data from all the studies. Q is the coefficient of heterogeneity (eq. 5) of the collated data [Cochran’s Q test (Cochran, 1954)] expressed as the collective weighted squared differences between the mean of each study and the variance in the weighted mean. A higher value of Q indicates greater heterogeneity.

The degree of heterogeneity can be assessed using the I2 index (eq. 6) proposed by Higgins and Thompson (2002). This index provides a percentage of overall heterogeneity that can be interpreted as proposed by Higgins et al. (2003) as follows: around 0%, no heterogeneity, around 25%, low heterogeneity, around 50%, moderate heterogeneity, and around 75%, high heterogeneity:Embedded Image(6)where I2 is the index of heterogeneity, Q is Cochran’s heterogeneity coefficient, and (k – 1) is the number of degrees of freedom defined as the number of studies, k, minus one. Where I2 is negative, it is set to zero. The probability, P, of the test can be quoted by comparing the Q value with a χ2 distribution with the same number of degrees of freedom (Higgins et al., 2003).

Assessing Correlations between the Abundances of Drug-Metabolizing Enzymes.

The Spearman rank test (Armitage et al., 2001) was used to assess correlations of the expression levels of the enzymes. Before correlation calculations, collated data were normalized using mean values of reference studies (eq. 7).Embedded Image(7)where Embedded Image is the normalized abundance value of the abundance measurement x in study j, Embedded Image is the mean abundance value of study j, and Embedded Image is the mean abundance of the same enzyme in the reference study used for the normalization process. The reference abundance values used for normalization were those obtained in this meta-analysis (Table 1).

View this table:
  • View inline
  • View popup
TABLE 1

The weighted means, coefficients of variation (CV), ranges, and heterogeneity analysis of the analyzed hepatic cytochrome P450 enzyme abundance data from 50 studies

Calculations of the Spearman correlation coefficients, Rs, and probabilities, P, were carried using a t-Student distribution. A Bonferroni correction was used to define the P value for statistical significance for the number of tests applied on each enzyme (a maximum of 14 tests for each enzyme).

Assessment of Gender- and Age-Related Differences in Expression.

Differences between expression levels in Caucasian adult liver cytochrome P450 enzymes in male and female subjects and in different age groups were investigated where data were available. Gender-related differences were investigated using the Mann-Whitney rank order U test, and correlation of expression with age was examined using the Spearman rank correlation test.

Results

Studies Used for the Meta-analysis.

The number of studies used in the meta-analysis was 50 of 94 studies collected using the search strategy. Several criteria were used to select the studies to be used for the meta-analysis. Studies were excluded where the samples were not individual liver microsomal samples (either recombinantly expressed systems, cell lines, or pooled samples; 13 studies), where enzymatic activity or mRNA levels of expression were the only quantification data (19 studies) and where all the study subjects were from a non-Caucasian ethnic group (six studies) or from a pediatric age group (four studies). Additionally, studies quoting abundance data in arbitrary, relative, or nonstandard units (other than pmol mg−1) were also excluded (seven studies). Some studies were excluded for more than one of these reasons.

The data in the selected studies for the meta-analysis were obtained in 26 different laboratories worldwide using two different types of experimental method for abundance measurement: immunoquantification (Western blotting or ELISA) and LC-MS. Most of the studies included in the meta-analysis used Western blotting (44 studies), whereas ELISA was used in three studies (Stresser and Kupfer, 1999; Snawder and Lipscomb, 2000; Barter et al., 2010) and LC-MS was used in five studies (Wang et al., 2008; Langenfeld et al., 2009; Seibert et al., 2009; Ohtsuki et al., 2012; Achour et al., 2014a). Wang et al. (2008); Langenfeld et al. (2009) used both immunoquantification and LC-MS. The range of enzymes quantified and the number of livers used in the studies were different for each enzyme (Table 1).

For correlation analysis, additional studies conducted on samples from non-Caucasian (Kawakami et al., 2011) and pediatric populations (Koukouritaki et al., 2004; Hines, 2007) were also included. Studies expressing abundance in relative units (Wrighton et al., 1987; Wrighton et al., 1993) were also used.

Meta-analysis of Cytochrome P450 Enzyme Abundance.

The data from available literature were collated and tested for normality and heterogeneity. Data did not show normal distribution. The data from different studies did not show extensive heterogeneity except for the CYP3A43 data set, which came from only two independent studies. Table 1 shows the results of the heterogeneity tests and the WXs and WCVs. Figure 1 shows the results of the meta-analysis for the 15 drug-metabolizing cytochrome P450 enzymes and a pie chart of the distribution of expression of these enzymes in hepatic tissue. The bar graph in Fig. 1 was divided into panels A and B to make it possible to visualize clearly the low abundance enzymes (CYP2C18, CYP2J2, and CYP3A43) using two different scales. Cytochrome P450 protein abundance pie charts from this meta-analysis and previous studies (Shimada et al., 1994; Rowland-Yeo et al., 2004) are shown in Supplemental Fig. 1.

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Bar graph (A and B) and pie chart (C) of weighted mean abundances of cytochrome P450 enzymes in livers from adult Caucasians. Error bars represent weighted standard deviation values. n, the number of livers.

Correlation Analysis of the Abundance of Cytochrome P450 Enzymes.

Where more than one enzyme was quantified in the same samples, the data were used for correlation analysis. This analysis included data from other ethnic backgrounds and from pediatric samples. Since the data were not normally distributed, the Spearman rank correlation test was used after normalizing the collated data using mean values obtained in this meta-analysis (Table 1). Table 2 shows the correlation matrix obtained using the correlation analysis, and Fig. 2 shows examples of statistically significant weak and strong correlations.

View this table:
  • View inline
  • View popup
TABLE 2

Correlations matrix of the abundance data of 15 cytochrome P450 enzymes from 32 studies

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Plots of examples of significant correlations of hepatic cytochrome P450 expression levels. Correlations between CYP2B6 and CYP3A4 (A) and between CYP2C9 and CYP2C19 (E) are weak, whereas correlations between CYP2C8 and CYP2C9 (B), CYP2C8 and CYP3A4 (C), and CYP2C9 and CYP3A4 (D) are strong. The strongest correlation was found between CYP3A4 and CYP3A5*1/*3 (F). Plots show normalized collated data.

Gender- and Age-Related Differences of Expression.

No statistically significant differences in cytochrome P450 abundance levels were found between male and female Caucasian subjects (Mann-Whitney U test, P > 0.05) for all data sets except CYP3A4, which was higher in livers from female subjects (n = 105) than in those from male subjects (n = 114) (Mann-Whitney U test, P < 0.0001) (Fig. 3). In addition, no correlation between hepatic levels of enzyme expression and age was found in Caucasian adults (Supplemental Fig. 2) except in the case of CYP2C9 (Spearman correlation test, P < 0.05, n = 60).

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Expression of cytochrome P450 in male and female Caucasian subjects. No sex differences were detected (U-test, P > 0.05) in any cases, except between levels of CYP3A4 in male and female livers (U-test, P < 0.0001). F, female; M, male; n, number of subjects in each set.

Discussion

Abundance levels and activities of drug-metabolizing enzymes play major roles in determining the fate of drugs and other xenobiotics in the body. In addition, pharmacokinetic evaluation of a new therapeutic entity, in the process of drug development, has become increasingly reliant on the use of in vitro systems that are dependent on scaling factors, which include the abundance data of enzymes involved in metabolism pathways relevant to the particular drug (Barter et al., 2007; Rostami-Hodjegan and Tucker, 2007).

This study aimed to analyze the published literature on the abundance of cytochrome P450 enzymes in adult Caucasian livers. Using the defined criteria, 50 individual studies from 26 different laboratories were selected for analysis. The number of livers used to quantify each enzyme ranged from 23 to 713 livers, with the CYP3A4 data set having the highest number of livers (n = 713) with the largest number of studies (k = 31). Some heterogeneity was found between data from different studies, especially in the case of CYP3A43; however, in the absence of information on interlaboratory consistency of assays, it is difficult to assign these differences to heterogeneity between samples. Nevertheless, it was clear that when Western blotting was the method of quantification, significant differences were found between abundance levels when different standards were used. Heterogeneity also tended to become less noticeable between data sets generated using the same quantitative method (immunoquantification or LC-MS). Data sets collected from a small number of studies tended to show higher levels of heterogeneity (especially in the case of CYP3A43), which should be interpreted with caution as the decreased numbers of degrees of freedom in these cases significantly diminish the power of the heterogeneity test (Higgins and Thompson, 2002; Higgins et al., 2003). Other sources of heterogeneity may include different protocols of the same method (especially in the case of Western blotting) and low numbers of samples in some studies, which can cause the effect of outliers to be magnified.

The data obtained in this study can be used as scaling factors for in vitro-in vivo extrapolation of measurements from in vitro recombinant enzyme systems and in simulations of drug trials and pharmacokinetic experiments (Table 1) (Barter et al., 2007; Rostami-Hodjegan and Tucker, 2007). The meta-analysis data revealed large interindividual variation across cytochrome P450 enzymes (5- to 1300-fold difference), highlighting the importance of taking variability into account in the process of in vitro-in vivo extrapolation (Rostami-Hodjegan and Tucker, 2007). Another aspect that can be incorporated in the process of pharmacokinetic simulation and the building of virtual populations is the correlation of expression between pharmacokinetically relevant proteins (Table 2). The strongest correlation was seen between levels of CYP3A4 and CYP3A5*1/*3 genetic variant (Rs = 0.70, P < 0.0001, n = 52, nine studies). This correlation was not strong when the CYP3A5 genotype was ignored (Rs = 0.06, P = 0.37, n = 218), a consistent observation in the literature (Lin et al., 2002; Barter et al., 2010; Achour et al., 2014a). Strong and statistically significant correlations between cytochrome P450 enzymes identified also include the following pairs: CYP3A4/CYP2C8 (Rs = 0.68, P < 0.0001, n = 134), CYP3A4/CYP2C9 (Rs = 0.55, P < 0.0001, n = 71), CYP2C8/CYP2C9 (Rs = 0.55, P < 0.0001, n = 99), and CYP1A2/CYP2C9 (Rs = 0.56, P < 0.0001, n = 53). Although a relatively strong correlation between CYP2B6 and CYP3A4 expression levels was reported in individual studies in the literature (Rs > 0.5) (Mimura et al., 1993; Totah et al., 2008; Achour et al., 2014a), this correlation was weaker on analysis of all the collected data (Rs = 0.46, P < 0.0001, n = 124, five studies). Correlations of expression have also been reported at the mRNA level (Wortham et al., 2007), which further supports reports of common genetic regulation of the expression of cytochrome P450 enzymes (Jover et al., 2009).

Although some of the correlations demonstrated by this meta-analysis are strong, it is necessary to exercise caution in interpreting these relationships. All the correlations observed were positive, and although this would be expected [correlation arising from common regulatory receptors and pathways (Wortham et al., 2007; Jover et al., 2009)], there appears to be a background positive correlation between all the cytochrome P450 enzymes under study. The background correlation is a necessary artifact of measuring abundance levels in units of pmol mg−1. A Bonferroni correction of the P value made the test stricter and provided more confidence in the strong correlations observed. Recently, it has been proposed that abundance levels would be more usefully measured in units of micrograms per gram of tissue in the case of uridine 5′-glucuronosyltransferase (UGT) enzymes (Milne et al., 2011), and the findings reported here may lend support to this suggestion.

Aging in adults (18 years onward) seemed to correlate with an overall apparent decline in expression of cytochrome P450 enzymes; however, correlation between age and levels of cytochrome P450 was not statistically significant (Spearman rank test, P > 0.05; Supplemental Fig. 2) for all the data sets except one (CYP2C9). In adult subjects, age was previously reported to have minimal effect on the abundance and activity of cytochrome P450 enzymes per mass unit of liver (King et al., 2003; Wolbold et al., 2003; Galetin et al., 2004). However, analysis of reported metabolic ratios (which were constant with age) indicated a decrease in the liver metabolic activity mediated by cytochrome P450 enzymes that is paralleled by the decline in renal function with age (Rostami-Hodjegan et al., 1999). Two contributing factors to these seemingly contradictory observations (i.e., no change in abundance per unit mass with age and reduced total metabolic activity) can be liver shrinkage that occurs when body size decreases with age (Johnson et al., 2005) and the decrease in the amount of total microsomal protein per gram liver (Barter et al., 2008). Therefore, although the abundance levels of expressed cytochrome P450 enzymes seem not to be affected by aging to a significant extent, the literature suggests that overall activity may be affected. The results of a recent study by Polasek et al. (2013) are also supportive of this view, although the area requires further investigation.

Gender differences in expression of cytochrome P450 were not statistically significant except in the case of CYP3A4, in line with literature on CYP3A4 abundance (Wolbold et al., 2003). This finding is supported by reports of more efficient clearance of CYP3A substrates, such as nifedipine (Krecic-Shepard et al., 2000), verapamil (Wolbold et al., 2003), and cyclosporine (Kahan et al., 1986), in female subjects. CYP3A4 is highly inducible, and only one study (Wolbold et al., 2003) has distinguished between inducer-exposed and control subjects. In the controls, CYP3A4 levels were on average 2.6 times higher in female than in male subjects (n = 39), whereas in inducer-exposed patients, the levels were much closer (1.7-fold difference in the mean, n = 55). The headline numbers would suggest that gender might influence clinical practice in prescribing drugs metabolized by CYP3A4; the reality, however, is that there is overlap between males and females in CYP3A4 expression and that exposure to inducers is much more significant than gender.

It is interesting to compare the present work with the single study (Shimada et al., 1994) published in 1994 and with the first meta-analysis (Rowland-Yeo et al., 2004) published in 2004 (see Supplemental Fig. 1). When restricted to the enzymes detectable in 1994 (CYP1A2, 2A6, 2B6, 2C, 2D6, 2E1, 3A), the pie charts are very similar, with the most striking difference being the growth of the relative abundance of CYP2B6 at the expense of CYP3A. We attribute this difference to the cross-reactivity of antibodies (early antibodies to CYP3A4 were rather nonspecific). The major difference between this work and earlier work, however, is the sheer number of individual enzymes detected; 15 individual enzymes have now been quantified, allowing a much more detailed human liver “pie” to be created and complementing our recent UGT liver pie (Achour et al., 2014b).

We look forward to extending this work to describe the expression profiles of cytochrome P450 in different ethnic groups and in pediatric samples in addition to investigating correlations of expression between different proteins (enzymes and transporters) that govern the processes of metabolism, disposition, and elimination of drugs in different organs, including the liver, the intestine, and the kidney.

Acknowledgments

The authors thank Eleanor Savill for assistance in preparing the manuscript.

Authorship Contributions

Participated in research design: Achour, Barber, Rostami-Hodjegan.

Performed data analysis: Achour.

Wrote or contributed to the writing of the manuscript: Achour, Barber, Rostami-Hodjegan.

Footnotes

    • Received April 30, 2014.
    • Accepted May 30, 2014.
  • dx.doi.org/10.1124/dmd.114.058834.

  • ↵Embedded ImageThis article has supplemental material available at dmd.aspetjournals.org.

Abbreviations

ELISA
enzyme-linked immunosorbent assay
LC-MS
liquid chromatography in conjunction with mass spectrometry
P450
cytochrome P450
WCV
weighted coefficient of variation
WX
weighted mean
  • Copyright © 2014 by The American Society for Pharmacology and Experimental Therapeutics

References

  1. ↵
    1. Achour B,
    2. Russell MR,
    3. Barber J, and
    4. Rostami-Hodjegan A
    (2014a) Simultaneous quantification of the abundance of several cytochrome P450 and uridine 5′-diphospho-glucuronosyltransferase enzymes in human liver microsomes using multiplexed targeted proteomics. Drug Metab Dispos 42:500–510.
    OpenUrlAbstract/FREE Full Text
  2. ↵
    1. Achour B,
    2. Rostami-Hodjegan A, and
    3. Barber J
    (2014b) Protein expression of various hepatic uridine 5′-glucuronosyltransferase (UGT) enzymes and their inter-correlations: a meta-analysis. Biopharm Drug Dispos (In press).
  3. ↵
    1. Al Omari A and
    2. Murry DJ
    (2007) Pharmacogenetics of the cytochrome P450 enzyme system: review of current knowledge and clinical significance. J Pharm Pract 20:206–218.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    Armitage P, Berry G, and Matthews JNS(2001) Statistical Methods in Medical Research, 4th ed. Wiley-Blackwell, Oxford.
  5. ↵
    1. Barter ZE,
    2. Bayliss MK,
    3. Beaune PH,
    4. Boobis AR,
    5. Carlile DJ,
    6. Edwards RJ,
    7. Houston JB,
    8. Lake BG,
    9. Lipscomb JC,
    10. Pelkonen OR,
    11. et al.
    (2007) Scaling factors for the extrapolation of in vivo metabolic drug clearance from in vitro data: reaching a consensus on values of human microsomal protein and hepatocellularity per gram of liver. Curr Drug Metab 8:33–45.
    OpenUrlCrossRefPubMed
  6. ↵
    1. Barter ZE,
    2. Chowdry JE,
    3. Harlow JR,
    4. Snawder JE,
    5. Lipscomb JC, and
    6. Rostami-Hodjegan A
    (2008) Covariation of human microsomal protein per gram of liver with age: absence of influence of operator and sample storage may justify interlaboratory data pooling. Drug Metab Dispos 36:2405–2409.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    1. Barter ZE,
    2. Perrett HF,
    3. Yeo KR,
    4. Allorge D,
    5. Lennard MS, and
    6. Rostami-Hodjegan A
    (2010) Determination of a quantitative relationship between hepatic CYP3A5*1/*3 and CYP3A4 expression for use in the prediction of metabolic clearance in virtual populations. Biopharm Drug Dispos 31:516–532.
    OpenUrlCrossRefPubMed
  8. ↵
    1. Cochran WG
    (1954) The combination of estimates from different experiments. Biometrics 10:101–129.
    OpenUrl
    1. Code EL,
    2. Crespi CL,
    3. Penman BW,
    4. Gonzalez FJ,
    5. Chang TK, and
    6. Waxman DJ
    (1997) Human cytochrome P4502B6: interindividual hepatic expression, substrate specificity, and role in procarcinogen activation. Drug Metab Dispos 25:985–993.
    OpenUrlAbstract/FREE Full Text
    1. Dennison JB,
    2. Jones DR,
    3. Renbarger JL, and
    4. Hall SD
    (2007) Effect of CYP3A5 expression on vincristine metabolism with human liver microsomes. J Pharmacol Exp Ther 321:553–563.
    OpenUrlAbstract/FREE Full Text
    1. Ekins S,
    2. Vandenbranden M,
    3. Ring BJ,
    4. Gillespie JS,
    5. Yang TJ,
    6. Gelboin HV, and
    7. Wrighton SA
    (1998) Further characterization of the expression in liver and catalytic activity of CYP2B6. J Pharmacol Exp Ther 286:1253–1259.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    1. Galetin A,
    2. Brown C,
    3. Hallifax D,
    4. Ito K, and
    5. Houston JB
    (2004) Utility of recombinant enzyme kinetics in prediction of human clearance: impact of variability, CYP3A5, and CYP2C19 on CYP3A4 probe substrates. Drug Metab Dispos 32:1411–1420.
    OpenUrlAbstract/FREE Full Text
    1. Guengerich FP and
    2. Turvy CG
    (1991) Comparison of levels of several human microsomal cytochrome P-450 enzymes and epoxide hydrolase in normal and disease states using immunochemical analysis of surgical liver samples. J Pharmacol Exp Ther 256:1189–1194.
    OpenUrlAbstract/FREE Full Text
    1. Haberl M,
    2. Anwald B,
    3. Klein K,
    4. Weil R,
    5. Fuss C,
    6. Gepdiremen A,
    7. Zanger UM,
    8. Meyer UA, and
    9. Wojnowski L
    (2005) Three haplotypes associated with CYP2A6 phenotypes in Caucasians. Pharmacogenet Genomics 15:609–624.
    OpenUrlCrossRefPubMed
    1. Hanna IH,
    2. Reed JR,
    3. Guengerich FP, and
    4. Hollenberg PF
    (2000) Expression of human cytochrome P450 2B6 in Escherichia coli: characterization of catalytic activity and expression levels in human liver. Arch Biochem Biophys 376:206–216.
    OpenUrlCrossRefPubMed
    1. Hesse LM,
    2. He P,
    3. Krishnaswamy S,
    4. Hao Q,
    5. Hogan K,
    6. von Moltke LL,
    7. Greenblatt DJ, and
    8. Court MH
    (2004) Pharmacogenetic determinants of interindividual variability in bupropion hydroxylation by cytochrome P450 2B6 in human liver microsomes. Pharmacogenetics 14:225–238.
    OpenUrlCrossRefPubMed
  10. ↵
    1. Higgins JPT and
    2. Thompson SG
    (2002) Quantifying heterogeneity in a meta-analysis. Stat Med 21:1539–1558.
    OpenUrlCrossRefPubMed
  11. ↵
    1. Higgins JPT,
    2. Thompson SG,
    3. Deeks JJ, and
    4. Altman DG
    (2003) Measuring inconsistency in meta-analyses. BMJ 327:557–560.
    OpenUrlFREE Full Text
  12. ↵
    1. Hines RN
    (2007) Ontogeny of human hepatic cytochromes P450. J Biochem Mol Toxicol 21:169–175.
    OpenUrlCrossRefPubMed
    1. Imaoka S,
    2. Yamada T,
    3. Hiroi T,
    4. Hayashi K,
    5. Sakaki T,
    6. Yabusaki Y, and
    7. Funae Y
    (1996) Multiple forms of human P450 expressed in Saccharomyces cerevisiae: systematic characterization and comparison with those of the rat. Biochem Pharmacol 51:1041–1050.
    OpenUrlCrossRefPubMed
  13. ↵
    1. Johnson TN,
    2. Tucker GT,
    3. Tanner MS, and
    4. Rostami-Hodjegan A
    (2005) Changes in liver volume from birth to adulthood: a meta-analysis. Liver Transpl 11:1481–1493.
    OpenUrlCrossRefPubMed
  14. ↵
    1. Jover R,
    2. Moya M, and
    3. Gómez-Lechón MJ
    (2009) Transcriptional regulation of cytochrome p450 genes by the nuclear receptor hepatocyte nuclear factor 4-alpha. Curr Drug Metab 10:508–519.
    OpenUrlCrossRefPubMed
    1. Jung F,
    2. Richardson TH,
    3. Raucy JL, and
    4. Johnson EF
    (1997) Diazepam metabolism by cDNA-expressed human 2C P450s: identification of P4502C18 and P4502C19 as low K(M) diazepam N-demethylases. Drug Metab Dispos 25:133–139.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    1. Kahan BD,
    2. Kramer WG,
    3. Wideman C,
    4. Flechner SM,
    5. Lorber MI, and
    6. Van Buren CT
    (1986) Demographic factors affecting the pharmacokinetics of cyclosporine estimated by radioimmunoassay. Transplantation 41:459–464.
    OpenUrlCrossRefPubMed
    1. Kamdem LK,
    2. Meineke I,
    3. Koch I,
    4. Zanger UM,
    5. Brockmöller J, and
    6. Wojnowski L
    (2004) Limited contribution of CYP3A5 to the hepatic 6β-hydroxylation of testosterone. Naunyn Schmiedebergs Arch Pharmacol 370:71–77.
    OpenUrlPubMed
  16. ↵
    1. Kawakami H,
    2. Ohtsuki S,
    3. Kamiie J,
    4. Suzuki T,
    5. Abe T, and
    6. Terasaki T
    (2011) Simultaneous absolute quantification of 11 cytochrome P450 isoforms in human liver microsomes by liquid chromatography tandem mass spectrometry with in silico target peptide selection. J Pharm Sci 100:341–352.
    OpenUrlCrossRefPubMed
    1. Kharasch ED,
    2. Hoffer C,
    3. Whittington D, and
    4. Sheffels P
    (2004) Role of hepatic and intestinal cytochrome P450 3A and 2B6 in the metabolism, disposition, and miotic effects of methadone. Clin Pharmacol Ther 76:250–269.
    OpenUrlCrossRefPubMed
  17. ↵
    1. King BP,
    2. Leathart JB,
    3. Mutch E,
    4. Williams FM, and
    5. Daly AK
    (2003) CYP3A5 phenotype-genotype correlations in a British population. Br J Clin Pharmacol 55:625–629.
    OpenUrlCrossRefPubMed
  18. ↵
    1. Koukouritaki SB,
    2. Manro JR,
    3. Marsh SA,
    4. Stevens JC,
    5. Rettie AE,
    6. McCarver DG, and
    7. Hines RN
    (2004) Developmental expression of human hepatic CYP2C9 and CYP2C19. J Pharmacol Exp Ther 308:965–974.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    1. Krecic-Shepard ME,
    2. Park K,
    3. Barnas C,
    4. Slimko J,
    5. Kerwin DR, and
    6. Schwartz JB
    (2000) Race and sex influence clearance of nifedipine: results of a population study. Clin Pharmacol Ther 68:130–142.
    OpenUrlCrossRefPubMed
    1. Kuehl P,
    2. Zhang J,
    3. Lin Y,
    4. Lamba J,
    5. Assem M,
    6. Schuetz J,
    7. Watkins PB,
    8. Daly A,
    9. Wrighton SA,
    10. Hall SD,
    11. et al.
    (2001) Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nat Genet 27:383–391.
    OpenUrlCrossRefPubMed
    1. Lamba V,
    2. Lamba J,
    3. Yasuda K,
    4. Strom S,
    5. Davila J,
    6. Hancock ML,
    7. Fackenthal JD,
    8. Rogan PK,
    9. Ring B,
    10. Wrighton SA,
    11. et al.
    (2003) Hepatic CYP2B6 expression: gender and ethnic differences and relationship to CYP2B6 genotype and CAR (constitutive androstane receptor) expression. J Pharmacol Exp Ther 307:906–922.
    OpenUrlAbstract/FREE Full Text
    1. Lang T,
    2. Klein K,
    3. Fischer J,
    4. Nüssler AK,
    5. Neuhaus P,
    6. Hofmann U,
    7. Eichelbaum M,
    8. Schwab M, and
    9. Zanger UM
    (2001) Extensive genetic polymorphism in the human CYP2B6 gene with impact on expression and function in human liver. Pharmacogenetics 11:399–415.
    OpenUrlCrossRefPubMed
  20. ↵
    1. Langenfeld E,
    2. Zanger UM,
    3. Jung K,
    4. Meyer HE, and
    5. Marcus K
    (2009) Mass spectrometry-based absolute quantification of microsomal cytochrome P450 2D6 in human liver. Proteomics 9:2313–2323.
    OpenUrlCrossRefPubMed
    1. Läpple F,
    2. von Richter O,
    3. Fromm MF,
    4. Richter T,
    5. Thon KP,
    6. Wisser H,
    7. Griese E-U,
    8. Eichelbaum M, and
    9. Kivistö KT
    (2003) Differential expression and function of CYP2C isoforms in human intestine and liver. Pharmacogenetics 13:565–575.
    OpenUrlCrossRefPubMed
    1. Lasker JM,
    2. Wester MR,
    3. Aramsombatdee E, and
    4. Raucy JL
    (1998) Characterization of CYP2C19 and CYP2C9 from human liver: respective roles in microsomal tolbutamide, S-mephenytoin, and omeprazole hydroxylations. Arch Biochem Biophys 353:16–28.
    OpenUrlCrossRefPubMed
  21. ↵
    1. Lin YS,
    2. Dowling AL,
    3. Quigley SD,
    4. Farin FM,
    5. Zhang J,
    6. Lamba J,
    7. Schuetz EG, and
    8. Thummel KE
    (2002) Co-regulation of CYP3A4 and CYP3A5 and contribution to hepatic and intestinal midazolam metabolism. Mol Pharmacol 62:162–172.
    OpenUrlAbstract/FREE Full Text
    1. McSorley LC and
    2. Daly AK
    (2000) Identification of human cytochrome P450 isoforms that contribute to all-trans-retinoic acid 4-hydroxylation. Biochem Pharmacol 60:517–526.
    OpenUrlCrossRefPubMed
  22. ↵
    1. Milne AM,
    2. Burchell B, and
    3. Coughtrie MWH
    (2011) A novel method for the immunoquantification of UDP-glucuronosyltransferases in human tissue. Drug Metab Dispos 39:2258–2263.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    1. Mimura M,
    2. Baba T,
    3. Yamazaki H,
    4. Ohmori S,
    5. Inui Y,
    6. Gonzalez FJ,
    7. Guengerich FP, and
    8. Shimada T
    (1993) Characterization of cytochrome P-450 2B6 in human liver microsomes. Drug Metab Dispos 21:1048–1056.
    OpenUrlAbstract
    1. Nakajima M,
    2. Nakamura S,
    3. Tokudome S,
    4. Shimada N,
    5. Yamazaki H, and
    6. Yokoi T
    (1999) Azelastine N-demethylation by cytochrome P-450 (CYP)3A4, CYP2D6, and CYP1A2 in human liver microsomes: evaluation of approach to predict the contribution of multiple CYPs. Drug Metab Dispos 27:1381–1391.
    OpenUrlAbstract/FREE Full Text
    1. Naraharisetti SB,
    2. Lin YS,
    3. Rieder MJ,
    4. Marciante KD,
    5. Psaty BM,
    6. Thummel KE, and
    7. Totah RA
    (2010) Human liver expression of CYP2C8: gender, age, and genotype effects. Drug Metab Dispos 38:889–893.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    1. Ohtsuki S,
    2. Schaefer O,
    3. Kawakami H,
    4. Inoue T,
    5. Liehner S,
    6. Saito A,
    7. Ishiguro N,
    8. Kishimoto W,
    9. Ludwig-Schwellinger E,
    10. Ebner T,
    11. et al.
    (2012) Simultaneous absolute protein quantification of transporters, cytochromes P450, and UDP-glucuronosyltransferases as a novel approach for the characterization of individual human liver: comparison with mRNA levels and activities. Drug Metab Dispos 40:83–92.
    OpenUrlAbstract/FREE Full Text
    1. Olesen OV and
    2. Linnet K
    (2000) Identification of the human cytochrome P450 isoforms mediating in vitro N-dealkylation of perphenazine. Br J Clin Pharmacol 50:563–571.
    OpenUrlCrossRefPubMed
    1. Olesen OV and
    2. Linnet K
    (2001) Contributions of five human cytochrome P450 isoforms to the N-demethylation of clozapine in vitro at low and high concentrations. J Clin Pharmacol 41:823–832.
    OpenUrlCrossRefPubMed
    1. Paine MF,
    2. Khalighi M,
    3. Fisher JM,
    4. Shen DD,
    5. Kunze KL,
    6. Marsh CL,
    7. Perkins JD, and
    8. Thummel KE
    (1997) Characterization of interintestinal and intraintestinal variations in human CYP3A-dependent metabolism. J Pharmacol Exp Ther 283:1552–1562.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    1. Polasek TM,
    2. Patel F,
    3. Jensen BP,
    4. Sorich MJ,
    5. Wiese MD, and
    6. Doogue MP
    (2013) Predicted metabolic drug clearance with increasing adult age. Br J Clin Pharmacol 75:1019–1028.
    OpenUrlCrossRefPubMed
  26. ↵
    1. Proctor NJ,
    2. Tucker GT, and
    3. Rostami-Hodjegan A
    (2004) Predicting drug clearance from recombinantly expressed CYPs: intersystem extrapolation factors. Xenobiotica 34:151–178.
    OpenUrlCrossRefPubMed
  27. ↵
    1. Rostami-Hodjegan A,
    2. Kroemer HK, and
    3. Tucker GT
    (1999) In-vivo indices of enzyme activity: the effect of renal impairment on the assessment of CYP2D6 activity. Pharmacogenetics 9:277–286.
    OpenUrlCrossRefPubMed
  28. ↵
    1. Rostami-Hodjegan A and
    2. Tucker GT
    (2007) Simulation and prediction of in vivo drug metabolism in human populations from in vitro data. Nat Rev Drug Discov 6:140–148.
    OpenUrlCrossRefPubMed
  29. ↵
    1. Rowland-Yeo K,
    2. Rostami-Hodjegan A, and
    3. Tucker GT
    (2004) Abundance of cytochrome P450 in human liver: a meta-analysis. Br J Clin Pharmacol 57:687–688.
    OpenUrl
  30. ↵
    1. Sánchez-Meca J and
    2. Martín-Martínez F
    (1997) Homogeneity tests in meta-analysis: a Monte Carlo comparison of statistical power and type I error. Qual Quant 31:385–399.
    OpenUrlCrossRef
  31. ↵
    1. Seibert C,
    2. Davidson BR,
    3. Fuller BJ,
    4. Patterson LH,
    5. Griffiths WJ, and
    6. Wang Y
    (2009) Multiple-approaches to the identification and quantification of cytochromes P450 in human liver tissue by mass spectrometry. J Proteome Res 8:1672–1681.
    OpenUrlCrossRefPubMed
  32. ↵
    1. Shimada T,
    2. Yamazaki H,
    3. Mimura M,
    4. Inui Y, and
    5. Guengerich FP
    (1994) Interindividual variations in human liver cytochrome P-450 enzymes involved in the oxidation of drugs, carcinogens and toxic chemicals: studies with liver microsomes of 30 Japanese and 30 Caucasians. J Pharmacol Exp Ther 270:414–423.
    OpenUrlAbstract/FREE Full Text
    1. Sim SC,
    2. Edwards RJ,
    3. Boobis AR, and
    4. Ingelman-Sundberg M
    (2005) CYP3A7 protein expression is high in a fraction of adult human livers and partially associated with the CYP3A7*1C allele. Pharmacogenet Genomics 15:625–631.
    OpenUrlCrossRefPubMed
  33. ↵
    1. Snawder JE and
    2. Lipscomb JC
    (2000) Interindividual variance of cytochrome P450 forms in human hepatic microsomes: correlation of individual forms with xenobiotic metabolism and implications in risk assessment. Regul Toxicol Pharmacol 32:200–209.
    OpenUrlCrossRefPubMed
  34. ↵
    1. Stresser DM and
    2. Kupfer D
    (1999) Monospecific antipeptide antibody to cytochrome P-450 2B6. Drug Metab Dispos 27:517–525.
    OpenUrlAbstract/FREE Full Text
    1. Tateishi T,
    2. Watanabe M,
    3. Moriya H,
    4. Yamaguchi S,
    5. Sato T, and
    6. Kobayashi S
    (1999) No ethnic difference between Caucasian and Japanese hepatic samples in the expression frequency of CYP3A5 and CYP3A7 proteins. Biochem Pharmacol 57:935–939.
    OpenUrlCrossRefPubMed
  35. ↵
    1. Totah RA,
    2. Sheffels P,
    3. Roberts T,
    4. Whittington D,
    5. Thummel K, and
    6. Kharasch ED
    (2008) Role of CYP2B6 in stereoselective human methadone metabolism. Anesthesiology 108:363–374.
    OpenUrlCrossRefPubMed
    1. Venkatakrishnan K,
    2. von Moltke LL,
    3. Court MH,
    4. Harmatz JS,
    5. Crespi CL, and
    6. Greenblatt DJ
    (2000) Comparison between cytochrome P450 (CYP) content and relative activity approaches to scaling from cDNA-expressed CYPs to human liver microsomes: ratios of accessory proteins as sources of discrepancies between the approaches. Drug Metab Dispos 28:1493–1504.
    OpenUrlPubMed
    1. von Richter O,
    2. Burk O,
    3. Fromm MF,
    4. Thon KP,
    5. Eichelbaum M, and
    6. Kivistö KT
    (2004) Cytochrome P450 3A4 and P-glycoprotein expression in human small intestinal enterocytes and hepatocytes: a comparative analysis in paired tissue specimens. Clin Pharmacol Ther 75:172–183.
    OpenUrlCrossRefPubMed
    1. Wandel C,
    2. Böcker RH,
    3. Böhrer H,
    4. deVries JX,
    5. Hofmann W,
    6. Walter K,
    7. Kleingeist B,
    8. Neff S,
    9. Ding R,
    10. Walter-Sack I,
    11. et al.
    (1998) Relationship between hepatic cytochrome P450 3A content and activity and the disposition of midazolam administered orally. Drug Metab Dispos 26:110–114.
    OpenUrlAbstract/FREE Full Text
    1. Wang YH,
    2. Jones DR, and
    3. Hall SD
    (2005) Differential mechanism-based inhibition of CYP3A4 and CYP3A5 by verapamil. Drug Metab Dispos 33:664–671.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    1. Wang MZ,
    2. Wu JQ,
    3. Dennison JB,
    4. Bridges AS,
    5. Hall SD,
    6. Kornbluth S,
    7. Tidwell RR,
    8. Smith PC,
    9. Voyksner RD,
    10. Paine MF,
    11. et al.
    (2008) A gel-free MS-based quantitative proteomic approach accurately measures cytochrome P450 protein concentrations in human liver microsomes. Proteomics 8:4186–4196.
    OpenUrlCrossRefPubMed
    1. Wester MR,
    2. Lasker JM,
    3. Johnson EF, and
    4. Raucy JL
    (2000) CYP2C19 participates in tolbutamide hydroxylation by human liver microsomes. Drug Metab Dispos 28:354–359.
    OpenUrlAbstract/FREE Full Text
    1. Westlind-Johnsson A,
    2. Malmebo S,
    3. Johansson A,
    4. Otter C,
    5. Andersson TB,
    6. Johansson I,
    7. Edwards RJ,
    8. Boobis AR, and
    9. Ingelman-Sundberg M
    (2003) Comparative analysis of CYP3A expression in human liver suggests only a minor role for CYP3A5 in drug metabolism. Drug Metab Dispos 31:755–761.
    OpenUrlAbstract/FREE Full Text
  37. ↵
    1. Wienkers LC and
    2. Heath TG
    (2005) Predicting in vivo drug interactions from in vitro drug discovery data. Nat Rev Drug Discov 4:825–833.
    OpenUrlCrossRefPubMed
  38. ↵
    1. Wolbold R,
    2. Klein K,
    3. Burk O,
    4. Nüssler AK,
    5. Neuhaus P,
    6. Eichelbaum M,
    7. Schwab M, and
    8. Zanger UM
    (2003) Sex is a major determinant of CYP3A4 expression in human liver. Hepatology 38:978–988.
    OpenUrlCrossRefPubMed
  39. ↵
    1. Wortham M,
    2. Czerwinski M,
    3. He L,
    4. Parkinson A, and
    5. Wan Y-JY
    (2007) Expression of constitutive androstane receptor, hepatic nuclear factor 4 α, and P450 oxidoreductase genes determines interindividual variability in basal expression and activity of a broad scope of xenobiotic metabolism genes in the human liver. Drug Metab Dispos 35:1700–1710.
    OpenUrlAbstract/FREE Full Text
  40. ↵
    1. Wrighton SA,
    2. Stevens JC,
    3. Becker GW, and
    4. VandenBranden M
    (1993) Isolation and characterization of human liver cytochrome P450 2C19: correlation between 2C19 and S-mephenytoin 4′-hydroxylation. Arch Biochem Biophys 306:240–245.
    OpenUrlCrossRefPubMed
  41. ↵
    1. Wrighton SA,
    2. Thomas PE,
    3. Willis P,
    4. Maines SL,
    5. Watkins PB,
    6. Levin W, and
    7. Guzelian PS
    (1987) Purification of a human liver cytochrome P-450 immunochemically related to several cytochromes P-450 purified from untreated rats. J Clin Invest 80:1017–1022.
    OpenUrlCrossRefPubMed
    1. Yamazaki H,
    2. Inoue K,
    3. Chiba K,
    4. Ozawa N,
    5. Kawai T,
    6. Suzuki Y,
    7. Goldstein JA,
    8. Guengerich FP, and
    9. Shimada T
    (1998) Comparative studies on the catalytic roles of cytochrome P450 2C9 and its Cys- and Leu-variants in the oxidation of warfarin, flurbiprofen, and diclofenac by human liver microsomes. Biochem Pharmacol 56:243–251.
    OpenUrlCrossRefPubMed
    1. Yamazaki H,
    2. Inoue K,
    3. Shaw PM,
    4. Checovich WJ,
    5. Guengerich FP, and
    6. Shimada T
    (1997) Different contributions of cytochrome P450 2C19 and 3A4 in the oxidation of omeprazole by human liver microsomes: effects of contents of these two forms in individual human samples. J Pharmacol Exp Ther 283:434–442.
    OpenUrlAbstract/FREE Full Text
    1. Yang TJ,
    2. Krausz KW,
    3. Shou M,
    4. Yang SK,
    5. Buters JT,
    6. Gonzalez FJ, and
    7. Gelboin HV
    (1998) Inhibitory monoclonal antibody to human cytochrome P450 2B6. Biochem Pharmacol 55:1633–1640.
    OpenUrlCrossRefPubMed
    1. Yasumori T,
    2. Nagata K,
    3. Yang SK,
    4. Chen LS,
    5. Murayama N,
    6. Yamazoe Y, and
    7. Kato R
    (1993) Cytochrome P450 mediated metabolism of diazepam in human and rat: involvement of human CYP2C in N-demethylation in the substrate concentration-dependent manner. Pharmacogenetics 3:291–301.
    OpenUrlCrossRefPubMed
    1. Zanger UM,
    2. Fischer J,
    3. Raimundo S,
    4. Stüven T,
    5. Evert BO,
    6. Schwab M, and
    7. Eichelbaum M
    (2001) Comprehensive analysis of the genetic factors determining expression and function of hepatic CYP2D6. Pharmacogenetics 11:573–585.
    OpenUrlCrossRefPubMed
  42. ↵
    1. Zanger UM,
    2. Klein K,
    3. Thomas M,
    4. Rieger JK,
    5. Tremmel R,
    6. Kandel BA,
    7. Klein M, and
    8. Magdy T
    (2014) Genetics, epigenetics, and regulation of drug-metabolizing cytochrome p450 enzymes. Clin Pharmacol Ther 95:258–261.
    OpenUrlCrossRef
PreviousNext
Back to top

In this issue

Drug Metabolism and Disposition: 42 (8)
Drug Metabolism and Disposition
Vol. 42, Issue 8
1 Aug 2014
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Editorial Board (PDF)
  • Front Matter (PDF)
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Drug Metabolism & Disposition article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Expression of Hepatic Drug-Metabolizing Cytochrome P450 Enzymes and Their Intercorrelations: A Meta-Analysis
(Your Name) has forwarded a page to you from Drug Metabolism & Disposition
(Your Name) thought you would be interested in this article in Drug Metabolism & Disposition.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Research ArticleArticle

Meta-analysis of P450 Abundance and Correlations

Brahim Achour, Jill Barber and Amin Rostami-Hodjegan
Drug Metabolism and Disposition August 1, 2014, 42 (8) 1349-1356; DOI: https://doi.org/10.1124/dmd.114.058834

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Research ArticleArticle

Meta-analysis of P450 Abundance and Correlations

Brahim Achour, Jill Barber and Amin Rostami-Hodjegan
Drug Metabolism and Disposition August 1, 2014, 42 (8) 1349-1356; DOI: https://doi.org/10.1124/dmd.114.058834
Reddit logo Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Authorship Contributions
    • Footnotes
    • Abbreviations
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF + SI
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • A PBPK model for CBD in adults and children
  • rs2242480 Regulates the Expression of CYP3A4 and CYP3A5
  • Absorption, Metabolism, and Excretion of Taselisib
Show more Articles

Similar Articles

Advertisement
  • Home
  • Alerts
Facebook   Twitter   LinkedIn   RSS

Navigate

  • Current Issue
  • Fast Forward by date
  • Fast Forward by section
  • Latest Articles
  • Archive
  • Search for Articles
  • Feedback
  • ASPET

More Information

  • About DMD
  • Editorial Board
  • Instructions to Authors
  • Submit a Manuscript
  • Customized Alerts
  • RSS Feeds
  • Subscriptions
  • Permissions
  • Terms & Conditions of Use

ASPET's Other Journals

  • Journal of Pharmacology and Experimental Therapeutics
  • Molecular Pharmacology
  • Pharmacological Reviews
  • Pharmacology Research & Perspectives
ISSN 1521-009X (Online)

Copyright © 2023 by the American Society for Pharmacology and Experimental Therapeutics