Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Available online at www.sciencedirect.

com

Cancer Letters 259 (2008) 1–15


www.elsevier.com/locate/canlet

Mini-review

Cytochrome P450s in the development


of target-based anticancer drugs
a,* b,1 b
Kedar Purnapatre , Sunil K. Khattar , Kulvinder Singh Saini
a
Infectious Diseases, New Drug Discovery Research, Ranbaxy Research Laboratories, Plot No. 20, Sector 18,
Udyog Vihar Industrial Area, Gurgaon 122001, Haryana, India
b
Biotechnology, New Drug Discovery Research, Ranbaxy Research Laboratories, Gurgaon 122001, Haryana, India

Received 27 August 2007; received in revised form 16 October 2007; accepted 17 October 2007

Abstract

Enzymes of the cytochrome P450 (CYP) superfamily are the major determinants of half-life and execute pharmacolog-
ical effects of many therapeutic drugs. In new drug discovery research, recombinant (human) CYPs are also used for iden-
tifying active or inactive metabolites that could lead to increased potency or toxicity of a molecule. In addition, CYP
inhibition by anticancer drugs might lead to adverse drug reactions, multiple-drug resistance, and drug–drug interactions.
During the discovery and pre-clinical evaluation of a New Chemical Entity (NCE), large amounts of purified recombinant
CYPs are required for studying metabolism and pharmacokinetic parameters. Therefore, present research efforts are
focused to over-express these human CYPs in bacteria, yeast, insect and mammalian cells, followed by their purification
on an industrial scale to facilitate identification of novel anticancer drugs. This review summarizes the merits and limita-
tions of these expression systems for an optimized production of individual CYP isoforms, and their usefulness in the dis-
covery and development of target-based, safe and efficacious NCEs for the treatment of cancer.
 2007 Elsevier Ireland Ltd. All rights reserved.

Keywords: Recombinant P450; CYP; Expression; Pharmacokinetics; Drug discovery

1. Introduction window between the drug safety parameters and its


therapeutic potential. Cytochrome P450 (CYP)
During the drug discovery research and develop- superfamily is the major determinant of half-life of
ment process, studies on the Metabolism and Phar- many therapeutic molecules and exerts their physio-
macokinetics (PK) of New Chemical Entity (NCE) logical effects through modulation of its enzyme
play a vital role in the determination of an optimum activity and by guiding some of these molecules
on the proteasomal degradation pathway. There
are 57 functional genes (and 58 pseudo genes) which
*
Corresponding author. Tel.: +91 124 4194400X5312; fax: +91 code for active CYP enzymes present in humans.
124 2343544. The enzymes of the CYP1, 2, and 3 families are
E-mail addresses: kedar.purnapatre@ranbaxy.com (K. Pur-
napatre), skhattar@umd.edu (S.K. Khattar).
involved in the biotransformation of xenobiotics
1
Present address: University of Maryland, Department of and chemicals, while the other CYP families
Veterinary Medicine, College Park, MD 20742, USA. (CYP4, 11, 17, 19, and 21) are involved in the

0304-3835/$ - see front matter  2007 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.canlet.2007.10.024
2 K. Purnapatre et al. / Cancer Letters 259 (2008) 1–15

‘‘housekeeping’’ metabolism of endogenous mole- tration of the inhibitor (3 lM) in a 96-well plate
cules [1,2]. The precise biochemical role of 58 format offers an economical and rapid screening
pseudo genes, in the overall metabolism of CYPs procedure for predicting drug–drug interactions
needs to be further evaluated, particularly if any in early phase of drug discovery [5]. Large
of these pseudo gene(s) have RNA interference amounts of CYP enzymes are required to perform
(RNAi) like regulatory properties. these HTS assays. Currently, there is considerable
Specific CYPs (primarily 1A2, 3A4, 2D6, 2C9, & effort by the pharmaceutical industry to over-
2C19) catalyze oxidative metabolism of over 90% of express human CYPs in bacteria, yeast, insect
the human drugs and are the major determinants of and mammalian cells and each of these systems
elimination or systemic clearance and bioavailabil- has been further exploited for the commercial pro-
ity of therapeutic compounds. These enzymes share duction of recombinant CYPs. However, these
considerable homology at the nucleotide level [3] expression systems need to be supplemented with
and contribution of the major isoforms to overall NADPH P450 oxidoreductase (OR) that provides
drug metabolism is as follows: 1A2 (4%), 2A6 reducing equivalents from NADPH for the activa-
(2%), 2C9 (10%), 2C19 (2%), 2E1 (2%), 2D6 tion of CYP. In addition, ancillary protein like
(30%), and 3A4 (50%). Interaction of CYP with cytochrome b5 may also enhance the activity of
drugs inside human body could lead to various out- P450s.
comes, which are listed in Fig. 1. Therefore, early Cancer patients, at times, undergo multiple treat-
determination of CYP inhibition potential and iden- ment regimes, which increases the risk of drug–drug
tification of CYP-mediated metabolism of NCEs is interactions followed by adverse drug reactions. In
absolutely necessary for the discovery of safe and addition, genetic polymorphism in CYP genes could
efficacious drugs. further complicate such therapy among various eth-
The combinatorial synthetic approach used in nic populations. Altered isoforms could either
the new drug discovery demands screening of a decrease or increase metabolism of certain antican-
large number of compounds at early drug discov- cer drugs leading to cytotoxicity in some patients.
ery phase. Such evaluation of the in vitro pharma- Over and above, CYP isoform(s) present in the
cokinetics is performed in a high throughput tumor cells can play an important role in tumor
screening (HTS) mode [4]. Optimization of HTS development [6]. Inside cancer cells, CYPs could
has been exploited well in the pharmaceutical either inactivate the anticancer drugs, like 2-meth-
industry. A comparative study evaluating 10-, 3- oxy estradiol, or activate the tumor-promoting com-
and single point at 3 lM screening assays was per- pound like 4-hydroxylestradiol [7,8]. Therefore,
formed for 569 data points [5]. Statistical analysis determination of CYP mediated metabolism
of these data demonstrated that variation and ana- appears to be mandatory for the discovery of novel
lytical precision for the IC50 generated by 10-point and safer anticancer therapies.
and 3-point methods were comparable to single This review describes methods of production of
point assay. Thus, utilization of a single concen- recombinant CYPs and their role in the metabolism

Fig. 1. Several outcomes of CYP–drug interaction: CYP metabolites can either have an increased potency (bioactivation) or are
hydrophilic intermediates that are eliminated from the body (elimination) or generate toxic intermediates. A drug can either inhibit or
induce CYP leading to drug–drug interaction during multi-drug therapy.
K. Purnapatre et al. / Cancer Letters 259 (2008) 1–15 3

of existing anticancer molecules, drug–drug interac- has been observed that the expression of human
tion and activation of a pro-drug into an active ther- 2C9 and 2C19 can be enhanced in bacteria by intro-
apeutic agent. In addition, the exploitation of ducing eight silent mutations in the N-terminal
single-nucleotide-polymorphism (SNP) of CYPs codons which reduced the formation of stable sec-
towards the discovery and development of target- ondary structures [35]. In addition, when the codon
based, safe, efficacious and personalized therapies specifying alanine (GCT) at the second position of
will be discussed. This review also briefly outlines transcripts was substituted for tryptophan (TGG),
the ‘‘targeted regulation’’ of CYP genes by RNAi, the resultant enzymes show catalytic properties sim-
like nucleotides. ilar to the native enzymes isolated from various ani-
mal sources [36].
2. Expression and production of recombinant CYPs
2.1.2. Fusion with leader peptide
Large number of pharmaceutical R&D compa-
Presence of hydrophobic membrane anchoring
nies are generating in-house recombinant human
sequences at the N-terminal of human CYPs is
CYPs for the determination of in vitro PK proper-
known to be deleterious for their expression in bac-
ties of their NCEs [9,10]. Recombinant CYPs with
teria [15]. This problem was overcome by construct-
the catalytic properties comparable to those of the
ing an in frame fusion of the CYP cDNA with a 21
human liver microsomes (HLM) have now been
amino acid long leader sequence of E. coli outer
expressed in bacteria, such as, Escherichia coli and
membrane protein, ompA, in frame with the CYP
Salmonella typhimurium; baker yeast Saccharomyces
initiation codon. In addition, a spacer sequence
cerevisiae. Insect cells with baculoviral vectors, or
for Ala-Pro was added at the ompA–CYP junction
mammalian cells such as lymphoblastoid cells,
which, leads to the proteolytic degradation of leader
COS cells, V79 CHO cells, HepG2, etc., with
sequence and thereby restores the production of the
vaccinia viral vectors have been extensively used
native CYP protein. Similarly, CYP reductase fused
[11–14]. Table 1 summarizes the strengths and chal-
to a pelB leader sequence was found to be optimal
lenges of various recombinant systems used for the
for the expression of OR on bacterial membranes.
production of CYP enzymes. CYPs expressed using
However, such N-terminal modifications are not
all these recombinant systems are available from
reported for the expression of CYPs in yeast, bacul-
different commercial sources and these enzymes
oviral or mammalian expression systems.
have shown comparable properties vis-a-vis HLM.

2.1. Strategies used to enhance expression of 2.1.3. Co-expression of CYP and OR


recombinant CYPs Functional monooxygenase system required for
CYP activity can be reconstituted by the co-expres-
CYPs are associated with microsomal mem- sion of CYP isozyme and OR. To achieve this objec-
branes and require NADPH reductase for their tive, the following protocols have been employed
functional activation [33]. Emerging strategies extensively to optimize the expression of individual
employed for the expression of a eukaryotic protein CYPs:
in a bacterial host have been appropriately In E. coli, co-expression of OR is achieved by
addressed and reviewed in a recent article from construction of CYP–OR fusion proteins [37], or
our laboratory [34]. Review of the recent literature CYP and OR are expressed separately using two
strongly suggests utilization of various modifica- different compatible plasmids like CYP gene in
tions in the recombinant CYPs as well as existing pCWOri+ vector and the OR gene in pACYC-184
protocols leading to the optimal expression of vector [38].
recombinant CYPs in various expression systems. In yeast cells, low levels of endogenous OR
These important observations are discussed in the (120–140 nmol/min/mg vs. 220 nmol/min/mg in
following section. mammalian tissue) were found to be insufficient
for the activation of CYP [23]. Catalytic properties
2.1.1. Modification of N-terminal of CYP expressed in yeast were improved by the
Deletion of strong secondary sequences around co-expression of human OR using high copy num-
the initiation codon of human CYPs are required ber plasmid-based expression system. Recently, a
for the expression of CYPs in E. coli [27,28]. It yeast strain co-expressing hOR and hCYP3A4 has
4
Table 1
Comparative analysis of various expression systems for P450 production
Expression Timea Typical Strengths Challenges
system yield
E. coli 3 months 150–500 • Availability of multiple cloning vectors & related recombi- • Often requires modifications of N-terminal sequences [26,27]
(bacteria) nmol/L nant technologies [18,19] • Catalytic activity of P450s requires addition of heme precursor like
[15] • Relatively inexpensive compared to other expression sys- delta amino levulinic acid to the growth media [28]
tems [20] • Temperature optimization is necessary to ensure adequate heme
• Requires short culture time incorporation and proper folding of recombinant P450s.
• Low maintenance & ease of purification [21] • Catalytic activity of bacterially expressed P450s requires reconstitu-
• Amenability to large scale production [22] tion with phospholipid(s) and accessory electron transfer proteins,
such as P450 oxidoreductase (OR) and cytochrome b5 [29]
S. cerevisiae 3 months 1–3 nmol/ • Ease of growth and purification • Yeast system possess low level of endogenous yeast reductase, which

K. Purnapatre et al. / Cancer Letters 259 (2008) 1–15


(yeast) L [15] • Similar to the mammalian cells in protein synthesis, process- results in low specific activity of recombinant human P450 [30]
ing, and membrane compartmentalization [23] • Endogenous cytochrome b5 in yeast is unable to enhance the activity
• Does not require any modification in the cDNA sequence of human CYPs, and is usually supplemented in the microsomal
coding for the P450 protein preparations [23]
• Endogenous membrane anchoring sequences target it to • Provision of heme is often limiting and endogenous CYPs compli-
endoplasmic reticulum and integrate into the ER cate interpretation of spectra and catalytic data in some strains [30]
• Yeast microsomes prepared from such recombinant strains
offer a rich source of P450s for in vitro studies [23]
Insect cells 2–3 103 ± 29 • Expression levels are high as compared to other systems [16] • Requires co-expression of OR, as insect cells lack endogenous OR,
with months nmol/L • Allows proper targeting of the recombinant protein into the and culture media must be supplemented with hemin [31,24]
baculoviral [16] host’s sub-cellular compartment [13] • Microsomes prepared from the insect cells, even in the presence of
vectors hemin contain significant amount of heme lacking apoprotein. This
apoprotein fraction is catalytically inactive and not spectroscopically
detected by CO difference spectra but could easily be detected by
western blotting [32]
• Cumbersome scale-up issues
Mammalian 6 months 20– • Mammalian cell lines endogenously express small quantities • Lymphoblastoid cells used for stable expression of CYPs contain
cells 100 pmol/ of OR and b5, which seems sufficient to activate expressed negligible amount of constitutive CYPs [24]
mg protein recombinant CYPs [15,24] • The expression level of recombinant P450s in mammalian cells is rel-
[17] • They possess the machinery for sub-cellular localization of atively low compared to other expression systems
P450 & electron transport chains. • Expensive growth culture media
• Capable of carrying out second phase metabolism, after the • Industrial production of CYPs is commercially a poor ‘‘return-on-
P450 reaction(s). investment’’ [17]
• Transient expression systems based on viral expression vec-
tors; e.g., SV40 based vectors in COS cells and vaccinia
virus system in HepG2 are available [25]
• The episomal mammalian stable expression system using
Epstein bar virus in human B lymphoid cells is commer-
cially used for the production of CYPs [15]
a
Timelines are only indicative, and denotes authors’ personal experience. It includes cloning, stable expression, and/or purification of a target P450.
K. Purnapatre et al. / Cancer Letters 259 (2008) 1–15 5

been shown to have 30-fold enhanced activity of the for catalytic activity. However, the conditions for
CYP3A4, compared to its parental strain [23]. reconstitution vary for each CYP, e.g., CYP3A4
In insect cells, OR required for the catalytically requires cytochrome b5 for its complete catalytic
active CYP isozymes is absent. This deficiency is activity. The optimal conditions for reconstitution
overcome by providing OR to the insect cell line of CYP1A2, 2C9, 2E1, and 3A4 can be found in a
using the following approaches: Both CYP and recent report by Yamazaki and Shimada [46].
OR can be produced individually and mixed
together to reconstitute active CYP system 2.2. Purification of recombinant CYPs
[31,39,40], or the desired CYP and reductase partner
were co-expressed using a single virus [41]. In Microsomes prepared from yeast, insect or mam-
another approach, insect cells were co-infected with malian cell lines with CYP coding plasmids provide
independent viruses containing individual CYP and rich source of CYP enzymes for in vitro PK studies.
a reductase partner [42]. For structural studies, CYP isoform 1A2, 2C9, 2D6,
In mammalian lymphoblast cell lines, certain and 3A4 have been purified from yeast microsomes
CYP isoforms require additional expression of [47]. CYP proteins were solubilized from yeast
active OR. Here, the activities of 1A2, 2B6, and microsomes in the presence of 0.6% sodium cholate
2C19 were supported by the endogenously and subjected to octylamino-sepharose 4B column
expressed OR, whereas 2A6, 2C8, 2C9, 2D6, and chromatography. CYPs eluted with 0.2–0.5% Emul-
3A4 required cotransfection with the OR cDNA gen were subjected to DEAE column chromatogra-
to bring a significant improvement in the enzymatic phy. The flowthrough fractions containing CYPs
activity of individual CYPs [32]. were subjected to hydroxylapatite column chroma-
tography. CYPs were further eluted by applying
2.1.4. Addition of delta-amino levulinic acid (d-ALA) 0.01–0.35 M Phosphate gradient. Typical yields
d-ALA is the precursor for the heme-moiety in obtained were 17 pmol/mg for CYP2C19 and 9–
CYP and needs to be supplemented in the growth 14 nmol/mg for CYP1A2, 2C9, 2D6, and 3A4 [47].
media for optimum production of CYP. In For the purification of CYP3A4 from insect cells,
E. coli, the yield of CYP1B1 could be enhanced microsomes were solubilized with 0.6% cholate and
by 20-fold with the inclusion of 2.5 mM d-ALA subjected to DEAE column chromatography. CYP
in the growth medium [28]. In yeast, excessive glu- from flow through and wash fractions was loaded
cose (30%), heat shock, and the addition of the d- on to Hydroxylapatite column to get rid of the
ALA favorably improved the activity of CYP3A4. detergent. CYP3A4 was recovered by washing the
Additional glucose helps to increase the growth column with 300 mM phosphate buffer. Typical
rate and protein expression levels of yeast cells, yield obtained was 12.7 nmol CYP/mg protein
and d-ALA, helps to improve both the stability [16]. From bacterial cells, CYPs were purified from
and activity of CYP enzymes [23]. In Insect cells, its membranes fractions. C-terminal His tag CYP
Hemin chloride or iron-citrate and d-ALA were 3A4 was solubilized from bacterial membranes by
added to the culture media as a source of heme non-ionic detergents and purified to homogeneity
to compensate low levels of endogenous heme. A using sequential DEAE and metal affinity chroma-
sharp increase in the synthesis of functional CYP tography [21].
2E1 was seen in the Tni cells supplemented with For crystallization studies, CYP3A4 with C-ter-
of 0.2 lg/ml heme-albumin [43]. minal His tag was solubilized with CHAPS and
purified by metal ion affinity chromatography fol-
2.1.5. Addition of cytochrome b5 lowed by CM-Sepharose [48].
Cytochrome b5 is an alternate electron carrier for
the second electron transfer involved in the catalytic 2.3. Validation of recombinant CYPs
activity of CYP. Co-expression of cytochrome b5
along with CYP3A4 and OR has been shown to CYP content from various recombinant expres-
increase the oxidation of nifedipine and testosterone sion systems is quantified by calculating the differ-
by CYP 3A4 to 167% in bacterial membrane prepa- ence between Fe2+ vs Fe2+-CO absorption
rations [44,45]. spectrum at 450 nm [49]. CYP reductase is measured
Recombinant CYPs are required to be supple- by cytochrome c reductase assay performed at
mented with OR, and membrane phospholipids 550 nm [50]. Recombinant CYPs obtained from het-
6 K. Purnapatre et al. / Cancer Letters 259 (2008) 1–15

erologous expression systems usually retain catalytic determination of Km and Ki are essential for the
properties, and substrate specificities like their prediction of in vitro–in vivo extrapolations in new
native human liver microsomes (HLM) counter- drug discovery.
parts. Recombinant CYPs are normally validated
by comparing the specificity, affinity and capacity 3. Role of CYPs in anticancer drug discovery
of the enzymes with those from the HLM [51].
The validation of specificity requires screening of Recombinant CYPs are used routinely in new
the metabolic activity for each isoform towards a drug discovery to predict in vitro PK properties that
panel of standard substrates. Affinity can be vali- can be extrapolated to in vivo PK of the NCE. Var-
dated by studying kinetics of biotransformation of ious functional assays are used to screen NCEs in
standard substrate. If Km values for standard sub- new drug discovery and some of key strategies for
strates are similar then affinity of both enzymes is the use of CYPs in the discovery and development
considered identical. Kcat or turn over number is of new drug candidates are described below:
another measure of affinity of enzyme towards the
substrate. The concentration of accessory electron 3.1. Role of CYPs in metabolism of existing
transfer proteins and the lipid composition of the anticancer drugs
membrane could alter the kinetic properties of
recombinant CYPs. Optimal molar ratio of OR to Hepatic CYP-mediated oxidation and conjuga-
recombinant CYPs is important for the maximum tion reactions lead to the elimination of compounds
activity of individual enzyme. For bacterially from human body [55]. Thus, the residence time of a
expressed 2C19, the OR/2C19 a ratio of 20:1 was compound in systemic circulation is directly depen-
found to be optimal for diazepam-N methylation dent on the resistance to this first pass metabolism.
activity [52]. In addition, membrane proteins and Intrinsic clearance (CLintr) is determined to extrap-
lipids from recombinant system might exhibit non- olate the in vitro data for prediction of in vivo
specific binding to compounds leading to decreased metabolism. It is a direct measure of enzyme activity
amount of ‘‘metabolically free’’ drug. Apparently towards a drug and is not influenced by other deter-
only unbound drug interacts freely with these minants like hepatic blood flow or plasma protein
metabolizing enzymes and such non-specific interac- binding.
tions could further lead to altered kinetic properties More than 30 CYP isoforms are known to
of recombinant enzymes [53]. metabolize xenobiotics and are used extensively to
Appropriate purification steps are required for study the metabolism of NCEs. Several CYPs, most
the recombinant proteins to obtain results compa- notably, CYP 1A2, 1B1, 2A6, 2C9, 2C19, 2D6, 2E1,
rable with the CYPs expressed from HLM. IC50s and 3A4/5 are responsible for the metabolism of
should be determined against a series of standard known anticancer drugs [56]. Specific CYP isoforms
inhibitors for both recombinant and HLM and their targeted anticancer drugs are listed in
enzymes. Due to high purity and specific activity Table 2.
of recombinant proteins total protein concentra- Endogenous CYP isoforms expressed in tumor
tions tends to be lower in incubation with recombi- cells may lead to the metabolism of active drug into
nant enzymes. In a study of recombinant CYP2D6 inactive or less potent form, thereby, altering the
inhibition, a 10-fold increase in total protein con- half-life and kinetics of the administered therapeutic
centration with constant CYP2D6 content led to molecule. One such example is the hydroxylation of
two to ninefold higher IC50 values for inhibition Paclitaxel into 6-hydroxy metabolite that decreases
of bufuralol 1 hydroxylation by fluoxetine, ezlopit- the potency of the drug by 30-fold [75].
ant and imipramine [53]. The use of RAF (relative In a rat oncology model, regulation of CYPs was
activity factor) is recommended to normalize values reported to occur primarily at post-transcriptional
to CYP content. RAF is the ratio of Vmax values level. Animals were treated with a single dose of Tri-
for the metabolism of marker substrates in incuba- acid (a metabolite of dehydrotarplatin), which led to
tion with HLM and recombinant enzymes [54]. the down-regulation of androgen-dependent male-
Also, addition of control protein can be optimized specific CYP2C11 mRNA and its apoprotein [76].
for IC50 determination using recombinant enzymes. Recently, heavy metal ions; arsenate, cadmium,
Taken together, these proposed mechanisms clearly and chromium, were shown to regulate CYP1A1
suggest that the optimization of conditions for the mRNA at post-transcriptional level in Hepa
K. Purnapatre et al. / Cancer Letters 259 (2008) 1–15 7

Table 2
Current anticancer drugs that are substrate for CYP 450 enzymes
Cancer Drugs CYP isoforms involved in References
Metabolism Activation
Breast Docetaxel CYP3A, CYP1B1 [57,58]
Ellipticine CYP3A4, CYP1A1, CYP1A2, [59]
CYP1B1, CYP2C9
Exemestane, Fulvestrant, CYP3A [57,60,61]
Vinblastine, Vinorelbine
Letrozole CYP3A, CYP2A6 [60]
Mitoxantrone CYP3A, CYP1B1 [58,62]
Paclitaxel CYP3A, CYP2C8 [58,63]
Tamoxifen CYP3A, CYP2D6, CYP2C9, [58,62–64,67,65]
CYP1B1, CYP2C19
Toremifene CYP3A, CYP1A2 [60]
Tegafur CYP2A6, CYP2C8, [66–68]
CYP1A2
Thiotepa CYP3A, CYP2B6 [69]
NSCLC Vinorelbine, Erlotinib, CYP3A4 [57,60,61]
Vindesine
Docetaxel CYP3A, CYP1B1 [57,58]
Gefitinib CYP3A, CYP2D6 [70]
Paclitaxel CYP3A, CYP2C8 [58,63,71]
SCLC Etoposide CYP3A4, CYP2E1, CYP1A2 [57,72,73]
Topotecan CYP3A [74]
Leukemias Idarubicin CYP2D6, CYP2C9 [60]
Imatinib, Teniposide, CYP3A [73]
Vindesine
Mitoxantrone CYP3A, CYP1B1 [58,62]
Cyclophosphamide CYP2B6, CYP2C19, [57,66]
CYP3A4
Sarcoma Paclitaxel CYP3A, CYP2C8 [63,71]
Vinblastine, Vincristine CYP3A [60,57,61]
Ifosfamide CYP3A, CYP2B6 [57,66]
Ovarian Paclitaxel CYP3A, CYP2C8 [58,63]
Topotecan CYP3A [74]
Thiotepa CYP3A, CYP2B6 [60]
Prostate Docetaxel CYP3A, CYP1B1 [60]
Flutamide CYP1A2 [60]
Colon Irinotecan CYP3A [66]
Tegafur CYP2A6, CYP2C8, [66–68]
CYP1A2
Lymphomas Vinblastine, Vincristine CYP3A [60,57,61]
Cyclophosphamide CYP2B6, CYP2C19, [57,66]
CYP3A4

1C1C7 cells, where chromium increased the half-life metabolic pathways involved will allow us to predict
of CYP1A1 [77]. Relatively little is known with with certainty, the efficacy and safety of novel anti-
respect to the stability of various CYP mRNAs cancer molecules under development [78]. Antican-
and their proteins after treatment with different che- cer agents are incubated with HLM or
motherapeutic agents. recombinant cytochrome CYPs and their metabo-
lites are identified by Mass Spectrometry. The data
3.2. CYP metabolism and the discovery of new generated from such studies will help us in:
anticancer molecules
(1) Designing new chemotherapeutic agent which
A significant mechanistic insight into the role of retain anticancer properties but show reduced
CYPs in the metabolism of anticancer drugs has CYP inhibitory activity or CYP induction
now been elucidated. A clear understanding of these potential. Such drugs will also help in reduc-
8 K. Purnapatre et al. / Cancer Letters 259 (2008) 1–15

tion of drug–drug interactions in patients its polymorphs [60]. Class I enzymes are well con-
undergoing multiple therapies. A SAR of served and are active in the metabolism of drugs
ADS102550, thienyl based hydroxamic acids and pre-carcinogens & includes CYP1A1, 1A2,
series of HDAC inhibitors has lead to identifi- 2E1, and 3A4, whereas Class II category of enzymes
cation of a compound with reduced CYP inhi- have important functional polymorphs and are
bition potential, retaining the HDAC involved in the metabolism of drugs. This latter
inhibitory activity [79]. class includes CYP 2B6, 2C9, 2A6, and 2D6.
(2) Further identification of active metabolites Identification of CYP SNPs is done using poly-
can be useful in understanding mechanism of merase chain reaction-restriction fragment polymor-
action of new chemotherapeutics. Novel anti- phism genotyping. Genotyping of 500 Caucasian
neoplastic agent, ellipticine, was shown to patients from Europe have identified the
act by intercalation into DNA and inhibition CYP3A5*11 allele. Functional characterization of
of DNA topoisomerase II. Recently, a novel recombinant CYP3A5*11 purified from E. coli dem-
mode of action mediated by CYP3A was iden- onstrated lower 3A5 activity [84]. Another SNP
tified. 9-hydroxy and 7-hydroxyellipticine CYP 2A6*7 represents gene deletion mostly in Asian
metabolites of ellipticine by CYP3A and per- population, at a frequency of 7–22% as compared to
oxidases lead to formation of DNA adducts 0.5–1% in Caucasians. In a recent study, patients
increasing the susceptibility to ellipticine [80]. with poor metabolizing activity of tegafur were
(3) Inhibitors or substrates of CYP responsible found to be heterozygous for 2A6*4 and 2A6*11
for the metabolism of anticancer agents can [85]. These observations clearly make a point that
be co-administered to increase the half-life of before including patients in clinical trials, they
drug and thus reduce dosing in patients. Co- should be screened for appropriate CYP poly-
administration of cyclosporine (substrate for morph(s) to avoid adverse drug reactions. Important
CYP3A4) increased oral bioavailability of CYP polymorphs metabolizing existing anticancer
docetaxol in cancer patients [81]. drugs are listed in Table 3. A Database of ethnic fre-
(4) CYP metabolism of inactive pro-drug into quency of clinically relevant CYP polymorphs and
cytotoxic drug can be explored in designing adverse drug reaction of commonly used medicines
safer potent anticancer agents. Several new has been used in creating a pharmocogenetic drug
agents now entering clinical trials (e.g., Phor- monitoring (PDM) system in Korea [86]. This
tress and AQ4N) are specifically designed to PDM demonstrated that CYP 2C9, 19, and 2D6
exploit tumor cytochrome CYP, resulting in are major CYP enzymes of clinical importance for
local bioactivation of the cytotoxicity within pharmacological effects and safety in the Korean
the tumor cells [82]. population [86]. Therefore, it appears highly likely
that recombinant polymorphic CYPs will be useful
in predicting certain pharmacological effects of exist-
3.3. Polymorphism in CYPs and anticancer therapy ing chemotherapeutics in a particular population.

Reaction phenotyping is employed to identify the 3.4. Role of CYPs in activation of anticancer pro-
isoform(s) that catalyze the metabolism of a particu- drugs
lar drug. Several polymorphs of human CYPs are
reported in the literature and have been extensively Idiosyncratic effects of a drug can occur by pro-
categorized. These reactions are used for predicting longed exposure in humans. It has been suggested
the consequences of drug administration to a partic- that bioactivation caused by CYPs could lead to
ular patient population, which are carrier of a partic- formation of reactive metabolites that are electro-
ular allele. Enzyme deficiency due to CYP2C19 philic in nature and are capable of causing covalent
alleles, 2C19*2 and 2C19*3, are predominant modification of protein or nucleic acids [87]. Some
amongst Asian populations, and should be included of these reactions are performed by pre-incubating
in reaction phenotyping, while conducting clinical tri- high concentrations of compounds with an excess
als [83]. Several recombinant human isoforms are of enzyme in the presence of glutathione or cyanide.
available commercially for reaction phenotyping. Resulting metabolites are purified by chromatogra-
Recently CYPs have been classified into two phy and further detected by mass spectroscopy [88].
groups based on the functional characterization of Recombinant enzymes are preferred in these reac-
K. Purnapatre et al. / Cancer Letters 259 (2008) 1–15 9

Table 3
Polymorphic P450 enzymes metabolizing anticancer drugs*
CYP isozymes No. of variants Important isoform Functional alteration Altered amino acid
1A1 15
1A2 36
2A6 52 2A6*4 Gene deletion (inactive)
2B6 53
2C8 12 2C8*3 Altered substrate specificity R139K,
K399R
2C8*4 I264M
2C9 37 2C9*2 Decreased enzyme activity Cys144 (R144C)
2C9*3 Decreased enzyme activity Leu359 (I359L)
2C19 25 2C19*2 Premature stop codon (inactive) splicing defect; I331V
2C19*3 W212X, I331V
2C19*17 Mutation in promoter. Increased transcription I331V
2D6 106 2D6*1
2D6*10 P34S, S486T
2E1 13 2E*3 Active V389I
2E*2 30% active R76H
3A4 40 3A4*1B Gene alteration in promoter
3A 4*2 Decreased enzyme activity S222P
1B1 23 1B1*7 Decreased Estradiol metabolism R48G, A119S, L432V, A443G
*Compiled from CYP allele homepage (http://www.imm.ki.se/cypalleles), [56,60].

tions as they exhibit higher specific activity com- patients undergoing multiple therapies [93]. Com-
pared to HLM. mercially available recombinant CYP inhibition kits
Various CYP isoforms are quite often over- offer HTS mode of predicting inhibition potential of
expressed during the development phase of a cancer a large number of compounds under investigation.
cell from a benign form to a highly metastatic malig- Two different approaches are normally followed to
nant phenotype. Two recent examples include the streamline the screening of the compounds. (1) If
over-expression of 1A1 & 1B1 isotypes in various a particular class of drugs has liability with respect
cancers [89,90]. This ability of using these over- to a single CYP, screening can be done initially
expressed CYP isoforms for the bioactivation of for that isoform to narrow down the number of
inactive pro-drugs into an active cytotoxic molecule compounds to be screened with other isoforms.
in tumor tissues is being exploited for designing che- While working with macrolides, we screen initially
motherapeutic agents. CYP1B1 has been shown to for liability against CYP3A4. Compounds showing
convert inactive pro-drug, reseveratrol, into active reduced liabilities can be taken further to determine
anticancer agent piceatannol in cancer cells [91] liability for other major hepatic CYP isoforms. (2)
and was also reported to convert DMU 135 to In another parallel approach, liabilities of major
DMU 117. DMU 117 is a non-selective tyrosine CYPs can be checked at a single concentration of
kinase (and COX) inhibitor that is being explored compounds, usually at 25 lM. Exact IC50s could
as a therapeutic molecule against gastrointestinal be determined further for selected compounds that
cancers [92]. In solid tumors, such bio-activation show greater inhibition at this single concentration.
of a pro-drug by CYPs, will aid in decreasing the IC50s of compounds for all five major CYPs (1A2,
cytotoxicity to the adjacent normal cells, and will 2D6, 3A4, 2C9, & 2C19) play important role in
also go a long way in targeting tumor-specific acti- deciding the molecules to be taken forward for
vation of chemotherapeutic agents. developmental studies.
Anticancer agents, like other therapeutics, are
3.5. CYP inhibition potential of anticancer drugs and known to have its own pharmacological profile and
its implications for drug–drug interactions PK properties, which may influence the PK parame-
ters of another drug administered simultaneously
Inhibition of CYP could affect accumulation or [93]. This will require careful monitoring, as patients
elimination of drugs administered simultaneously, with advanced cancer quite often require other med-
and might cause irreversible tissue damage to ications as well. Such drug–drug interaction could
10 K. Purnapatre et al. / Cancer Letters 259 (2008) 1–15

alter the efficacy of both the drugs and might lead to and colon cancers. CYP2J2 was found to be over-
adverse reactions in some patients. E7070 was dis- expressed in esophageal, adenocarcinoma, pulmon-
covered as novel sulfonamide anticancer agent that ary squamous carcinoma, and breast carcinoma
arrests cancer cells at the G1/S boundary of the cell tissues [98]. Thus careful determination of expres-
cycle. In vitro studies showed that E7070 has the sion levels of these CYPs could also serve as a good
potential to inhibit several CYP enzymes, including bio-marker for early detection of cancer in tumor
CYP2C9, CYP2C19, CYP2D6, CYP2E1, and biopsies. Indeed, CYP1B1 is already being used as
CYP3A4. The oral anticoagulant, Acenocoumaro, a prognostic marker for cancer [Patent No:
is metabolized in humans by CYP2C9. Upon co- WO0135810]. Depending upon the secretion &
administration, E7070 decreased metabolism of Ace- detection of some of these CYPs into blood, urine,
nocoumarol even at a very low concentration sputum & stool, non-invasive diagnostic and/or
(2.1 lM), thereby reducing the systemic clearance prognostic test could also be developed for various
of Acenocoumarol [94]. In the absence of careful cancers.
analysis of individual patients, this drug–drug inter-
action may result in hypoprothrombinemia and a 3.7. CYPs inhibitors as anticancer drugs
hemorrhagic tendency [94].
Changes in PK properties of co-administered Are tumor cells capable of efflux of chemothera-
drugs are also regulated by drug efflux pump; P-gly- peutic drugs out of their cellular compartments with
coprotein (Pgp, product of MDR1) [93]. Most com- help from the CYP enzymes? Inhibition of these
pounds, which are substrates for Pgp are also endogenous CYPs in cancerous cells might thus offer
substrates for CYP3A4 and can act synergistically a novel target for designing new anticancer agents,
in vivo. Gleevac (imatinib mesylate) is administered whereas simultaneous activation of drug-specific
orally for the treatment of chronic myeloid leuke- CYPs inside the normal cells (adjacent to the tumors)
mia (CML). It is a substrate for 3A4 but simulta- might help in reducing adverse reactions. Extra-
neously inhibits CYP3A4, 2C9, 2D6. Therefore, hepatic CYPs, that are involved in the metabolism
agents like Erythromycin or ketokonazole (3A4 of hormones, and are not responsible for xenobiotic
inhibitors) and dexamethhazone or carbamazepine metabolism, are also being targeted using this
(3A4 inducers) will change concentrations and bio- approach. For example, inhibitors of CYPs responsi-
availability of Gleevac in a multi-drug administra- ble for the metabolism of hormones or vitamins, are
tion scenario, and might lead to adverse events in being used as anticancer drugs for hormone depen-
patients [93,95]. dent cancers. CYP 19, an aromatase, catalyses a
Determination of the in vitro CYP inhibition rate-limiting step in the conversion of androgen to
potential of novel anticancer drugs is a tool, which estrogen. Aromatase inhibitors, like exemestane, or
could help us in predicting possible drug–drug inter- non-steroidal trazoles, letrazole(femera), and anas-
actions. It may also be useful in designing a treat- trozole are used successfully to maintain low levels
ment regime to be followed in patients undergoing of estrogen in post-menopausal women patients with
chemotherapy, where some of these CYPs could hormone sensitive breast cancer [99].
be monitored as prognostic bio-markers. Vitamin D3 metabolite, 1a,25(OH)2D3 (calci-
trol), inhibits proliferation, promotes differentiation
3.6. CYPs as prognostics/diagnostic markers for the and induces apoptosis in colon and prostrate cancer
detection of tumors cells [100]. However, deactivation of 1a,25(OH)2D3
occurs due to hydroxylation at C4 via CYP 24
Certain CYP isoforms like 1B1 and 1A2 are expressed in kidney cells. QW-1642, a synthetic ana-
induced in response to environmental mutagens like logue of 1a,25(OH)2D3 was identified, which inhib-
UV [96]. In addition, altered gene regulation in its the expression of CYP24, and further inhibited
tumor cells could lead to the activation of endoge- cell proliferation. This synthetic analogue has
nously expressed CYPs. Over-expression of shown good efficacy in pre-clinical studies and could
CYP1B1, CYP2J2, and CYP2W1 has been reported be developed as a chemotherapeutic agent for the
in various cancer tissues [90,97,98]. Furthermore, prostate cancer [101].
several carcinogens that are metabolized by CYP Various strategies to inhibit or exploit CYP
enzymes binds to AhR, thereby inducing the expres- enzymes for the treatment of cancer have been
sion of CYP1B1 in prostate, kidney, ovarian, breast, recently reviewed by Bruno and Njar [99].
K. Purnapatre et al. / Cancer Letters 259 (2008) 1–15 11

3.8. SiRNA and CYP metabolism used commercially as they possess most of the ancil-
lary proteins required for the activation of CYPs.
RNAi offers a powerful tool to achieve target Recombinant CYPs play a major role in the dis-
selective knockdown of specific genes. Recently covery of novel anticancer compounds. Individual
hPXR-siRNA was used for down-regulating the CYP enzymes are used to study the metabolism
expression of CYP genes controlled by the PXR ele- and inhibition of a potential new molecule. Inside
ment in primary human hepatocytes [102]. These the tumor cell, metabolism by the endogenous
siRNAs not only suppressed 3A4 transcript levels CYPs is being exploited for the activation of a
to about 18% in primary hepatocytes but it also sup- non-toxic pro-drug into a cytotoxic drug. Simulta-
pressed the induction of 3A4 by inducers like Phe- neously, such studies could give insights into possi-
nobarbital (1 mM) and Clotrimazole (10 lM). ble drug–drug interactions and they may also help
Besides CYP3A4, levels of 2A6, 2C8, and 3A5 were in determining the safety and efficacy of these
also suppressed by these siRNAs. In another study, NCEs.
3A4III siRNAs (specific for suppression of Polymorphism in CYPs could lead to the identi-
CYP3A4) significantly diminished cytotoxicity of fication of the genetic parameters which may cause
two CYP3A4 substrate drugs, cyclophosphamide varied therapeutic response in different ethnic
and ifosamide in CHL-3A4 cells [103]. It is clear groups. Recombinant allelic CYPs are being used
that the siRNA technology could be explored fur- to predict the efficacy of new molecules in geneti-
ther to suppress organ specific toxicity caused by cally diverse populations, and some of these individ-
induction or suppression of different CYP isoforms ual CYP SNPs data has been compiled onto
by anticancer drugs [103]. Furthermore, RNAi tech- microarray slides, which are now available from
nology may be able to target multiple genes in met- commercial vendors. Inhibitors of endogenously
abolically diverse pathways, and eventually help us expressed CYPs, like 1B1, are being explored by
override drug-resistance issues normally encoun- many pharmaceutical companies as possible thera-
tered with single target-based drugs. Co-administra- peutic targets. Over-expressed CYPs, with an
tion of siRNAs along with a therapeutic drug will altered substrate/inhibitor specificity, offer great
not only reduce drug–drug interaction but can also advantage for the designing of safe anticancer mol-
offer altered metabolism leading ultimately to an ecules, as levels of these CYPs are either undetect-
increased specificity and reduced toxicity. Most able or miniscule in normal cells. Recombinant
importantly, unlike the small molecules that inhibit CYPs will enable us to screen compounds that mod-
various CYP polymorphs differentially, siRNAs will ulate CYP levels inside the tumor cells. Co-adminis-
not discriminate amongst these isoforms and can tration of anticancer drugs with CYP activity
regulate all the polymorphs uniformly. modulators can activate/inactivate pro-drugs, or
A detailed analysis of this very powerful technol- could reduce the formation of cytotoxic metabo-
ogy in cancer is beyond the scope of this review, lite(s), thereby leading to the discovery and develop-
although two recent reviews outlining RNAi-medi- ment of safe and efficacious NCEs. It remains to be
ated anti-tumorigenesis for drug-target validation seen that at what stage of cancer development such
have appeared in the literature [104,105]. Effective a combination strategy will be most effective and
applications of such siRNAs further warrant the will be close to the tolerated doses. Further, with
development of more efficient in vivo siRNA deliv- RNAi technology, we may be able to dissect not
ery systems and then analyzing their therapeutic only the mechanism of action of NCEs under devel-
potential in animal models of cancer. opment, but also design compounds which will tar-
get multiple ‘‘culprit genes’’ in the signal
transduction pathway of cancer.
4. Conclusion and future directions
Acknowledgements
In this review, we have summarized methods of
production of recombinant CYPs in various expres- This work was funded by Ranbaxy Research
sion systems. E. coli and yeast offer simple & eco- Laboratories, India. We thankfully acknowledge
nomical modes of production, while insect cells our recombinant P450 group: Dr. Deepika Singh,
with baculovirus expression system have shown Ms. Alka Khanna, Ms. Pratibha Sharma and Mr.
highest yields. Human lymphoblast cells are also Hemant Kumar Chavan for their assistance during
12 K. Purnapatre et al. / Cancer Letters 259 (2008) 1–15

the compilation of this review and for generating the intralaboratory comparison, Drug Metabol. Rev. 31 (1999)
unpublished data. 523–544.
[16] J.T. Buters, K.R. Korzekwa, K.L. Kunze, Y. Omata, J.P.
Hardwick, F.J. Gonzalez, cDNA-directed expression of
References human cytochrome P450 CYP3A4 using baculovirus, Drug
Metab. Dispos. 22 (1994) 688–692.
[1] F.J. Gonzalez, The molecular biology of cytochrome P450s, [17] F.J. Gonzale, K.R. Korzekawa, Cytochrome P450 expres-
Pharmacol. Rev. 40 (1988) 243–288. sion systems, Annu. Rev. Pharmacol. Toxicol. 35 (1995)
[2] B.S. Kalra, Cytochrome P450 enzyme isoforms and their 369–390.
therapeutic implications: an update, Indian J. Med. Sci. 61 [18] F.P. Guengerich, A. Parikh, E.F. Johnson, T.H. Richard-
(2007) 102–116. son, von C. Wachenfeldt, J. Cosme, F. Jung, C.P. Strass-
[3] A. Juneja, K.S. Saini, R. Soni, Discerning relationships burg, M.P. Manns, R.H. Tukey, M. Pritchard, S. Fournel-
among human cytochrome P450s by computational anal- Gigleux, B. Burchell, Heterologous expression of human
yses, Bioinf. Trends 1 (2006) 1–12. drug-metabolizing enzymes, Drug Metab. Dispos. 25 (1997)
[4] G. Zlokarnik, P.D. Grootenhuis, J.B. Watson, High 1234–1241.
throughput P450 inhibition screens in early drug discovery, [19] T. Uno, A. Nakao, S. Masuda, Y. Taniguchi, K. Kana-
Drug Discov. Today 10 (2005) 1443–1450. maru, H. Yamagata, M. Nakamura, H. Imaishi, K. Oono,
[5] F. Gao, D.L. Johnson, S. Ekins, Optimizing higher Modification of small molecules by using cytochrome P450
throughput methods to assess drug–drug interactions for expressed in Escherichia coli, J. Ind. Microbiol. Biotechnol.
CYP1A2, CYP2C9, CYP2C19, CYP2D6, rCYP2D6, and 33 (2006) 1043–1050.
CYP3A4 in vitro using a single point IC(5), J. Biomol. [20] M.P. Pritchard, L. McLaughlin, T. Friedberg, Establish-
Screen. 7 (2002) 373–382. ment of functional human cytochrome P450 monooxygen-
[6] M.C. McFadyen, W.T. Melvin, G.I. Murray, Cytochrome ase systems in Escherichia coli, Methods Mol. Biol. 320
P450 enzymes: novel options for cancer therapeutics, Mol. (2006) 19–29.
Cancer Ther. 3 (2004) 363–371. [21] F.P. Guengerich, M.V. Martin, Purification of cytochromes
[7] N.J. Lakhani, M.A. Sarkar, J. Venitz, W.D. Figg, 2- P450: products of bacterial recombinant expression sys-
Methoxyestradiol, a promising anticancer agent, Pharma- tems, Methods Mol. Biol. 320 (2006) 31–37.
cotherapy 23 (2003) 165–172. [22] Y. Kanamori, K. Fujita, K. Nakayana, H. Kawai, T.
[8] S. Jennewein, C.D. Rithner, R.M. Williams, R.B. Croteau, Kamataki, Large-scale production of genetically engineered
Taxol biosynthesis: taxane 13 alpha-hydroxylase is a CYP3A4 in E. coli: application of a Jarfermenter, Drug
cytochrome P450-dependent monooxygenase, Proc. Natl. Metab. Pharmacokin. 18 (2003) 42–47.
Acad. Sci. USA 98 (2001) 13595–13600. [23] J. Cheng, D.F. Wan, J.R. Gu, Y. Gong, S.L. Yang,
[9] C.M. Masimirembwa, C. Otter, M. Berg, M. Jönsson, B. D.C. Hao, L. Yang, Establishment of a yeast system
Leidvik, E. Jonsson, T. Johansson, A. Bäckman, A. that stably expresses human cytochrome P450 reductase:
Edlund, T.B. Andersson, Heterologous expression and application for the study of drug metabolism of
kinetic characterization of human cytochromes P-450: cytochrome P450s in vitro, Protein Expr. Purif. 47
validation of a pharmaceutical tool for drug metabolism (2006) 467–476.
research, Drug Metab. Dispos. 27 (1999) 1117–1122. [24] C.L. Crespi, B.W. Penman, Use of cDNA-expressed human
[10] R.B. Vail, M.J. Homann, I. Hanna, A. Zaks, Preparative cytochrome P450 enzymes to study potential drug–drug
synthesis of drug metabolites using human cytochrome interactions, Adv. Pharmacol. 43 (1997) 171–188.
P450s 3A4, 2C9 and 1A2 with NADPH-P450 reductase [25] T. Aoyama, S. Yamano, P.S. Guzelian, H.V. Gelboin, F.J.
expressed in Escherichia coli, J. Ind. Microbiol. Biotechnol. Gonzalez, Five of 12 forms of vaccinia virus-expressed
32 (2005) 67–74. human hepatic cytochrome P450 metabolically activate
[11] F.P. Guengerich, A. Parikh, Expression of drug-metabo- aflatoxin B1, Proc. Natl. Acad. Sci. USA 87 (1990) 4790–
lizing enzymes, Curr. Opin. Biotechnol. 8 (1997) 623–628. 4793.
[12] P. Urban, C. Mignotte, M. Kazmaier, F. Delorme, D. [26] M.P. Pritchard, R. Ossetian, D.N. Li, C.J. Henderson, B.
Pompon, Cloning, yeast expression, and characterization of Burchell, C.R. Wolf, T. Friedberg, A general strategy for
the coupling of two distantly related Arabidopsis thaliana the expression of recombinant human cytochrome P450s in
NADPH-cytochrome P450 reductases with P450 Escherichia coli using bacterial signal peptides: expression
CYP73A5, J. Biol. Chem. 272 (1997) 19176–19186. of CYP3A4, CYP2A6, and CYP2E1, Arch. Biochem.
[13] C.A. Lee, T.A. Kost, C.J. Serabjit-Singh, Recombinant Biophys. 345 (1997) 342–354.
baculovirus strategy for coexpression of functional human [27] T.D. Porter, S. Chang, Strategies to enhance the coexpres-
cytochrome P450 and P450 reductase, Methods Enzymol. sion of cytochrome P450 2E1 and reductase in bacteria,
272 (1996) 86–95. Drug Metab. Rev. 31 (1999) 159–174.
[14] C.L. Crespi, V.P. Miller, The use of heterologously [28] I. Jansson, I. Stoilov, M. Sarfarazi, J.B. Schenkman,
expressed drug metabolizing enzymes-state of the art and Enhanced expression of CYP1B1 in Escherichia coli,
prospects for the future, Pharmacol. Ther. 84 (1999) 121– Toxicology 144 (2000) 211–219.
131. [29] P.M. Shaw, N.A. Hosea, D.V. Thompson, J.M. Lenius,
[15] T. Friedberg, M.P. Pritchard, M. Bbandera, S.P. Hanlon, F.P. Guengerich, Reconstitution premixes for assays using
D. Yao, L.A. McLaughlin, S. Ding, B. Burchell, C.R. Wolf, purified recombinant human cytochrome P450, NADPH-
Merits and limitations of recombinant models for the study cytochrome P450 reductase, and cytochrome b5, Arch.
of human P450-mediated drug metabolism and toxicity: an Biochem. Biophys. 348 (1997) 107–115.
K. Purnapatre et al. / Cancer Letters 259 (2008) 1–15 13

[30] P. Urban, C. Cullin, D. Pompon, Maximizing the expres- activities by cytochrome b (5) in bacterial membranes, Drug
sion of mammalian cytochrome P-450 monooxygenase Metab. Dispos. 27 (1999) 999–1004.
activities in yeast cells, Biochimie 72 (1990) 463–472. [45] M.W. Voice, Y. Zhang, C.R. Wolf, B. Burchell, T.
[31] S. Tamura, K.R. Korzekwa, S. Kimura, H.V. Gelboin, F.J. Friedberg, Effects of human cytochrome b5 on CYP3A4
Gonzalez, Baculovirus-mediated expression and functional activity and stability in vivo, Arch. Biochem. Biophys. 366
characterization of human NADPH-P450 oxidoreductase, (1999) 116–124.
Arch. Biochem. Biophys. 293 (1992) 219–223. [46] H. Yamazaki, T. Shimada, Cytochrome P450 reconstitu-
[32] K. Venkatakrishnan, L.L. VonMoltke, D.J. Greenblatt, tion systems, Methods Mol. Biol. 320 (2006) 61–71.
Human drug metabolism and the cytochromes P450: [47] S. Imaoka, T. Yamada, T. Hiroi, K. Hayashi, T. Sakaki, Y.
application and relevance of in vitro models, J. Clin. Yabusaki, Y. Funae, Multiple forms of human P450
Pharmacol. 41 (2001) 1149–1179. expressed in Saccharomyces cerevisiae. Systematic charac-
[33] I.F. Sevrioukova, J.A. Peterson, NADPH-P-450 reductase: terization and comparison with those of the rat, Biochem.
structural and functional comparisons of the eukaryotic Pharmacol. 51 (1996) 1041–1050.
and prokaryotic isoforms, Biochimie 77 (1995) 562–572. [48] J.K. Yano, M.R. Wester, G.A. Schoch, K.J. Griffin, C.D.
[34] S. Sahdev, S.K. Khattar, K.S. Saini, Production of active Stout, E.F. Johnson, The structure of human microsomal
eukaryotic proteins through bacterial expression systems: a cytochrome P450 3A4 determined by X-ray crystallography
review of the existing biotechnology strategies, Mol. Cell. to 2.05-A resolution, J. Biol. Chem. 279 (2004) 38091–
Biochem., in press. 38094.
[35] T.H. Richardson, F. Jung, K.J. Griffin, M. Wester, J.L. [49] T. Omura, R. Sato, The carbon monoxide-binding pigment
Raucy, B. Kemper, L.M. Bornheim, C. Hassett, C.J. of liver microsomes. I. Evidence for its hemoprotein nature,
Omiecinski, E.F. Johnson, A universal approach to the J. Biol. Chem. 239 (1964) 2370–2378.
expression of human and rabbit cytochrome P450s of the [50] I.R. Phillips, E.A. Shephard, Cytochrome P450 protocols,
2C subfamily in Escherichia coli, Arch. Biochem. Biophys. Methods Mol. Biol. 320 (2006) 1–10.
323 (1995) 87–96. [51] K. Venkatakrishnan, L.L. von Moltke, D.J. Greenblatt,
[36] K. Fujita, T. Kamataki, Genetically engineered bacterial Application of the relative activity factor approach in
cells co-expressing human cytochrome P450 with NADPH- scaling from heterologously expressed cytochromes P450 to
cytochrome P450 reductase: prediction of metabolism and human liver microsomes: studies on amitriptyline as a
toxicity of drugs in humans, Drug Metab. Pharmacokinet. model substrate, J. Pharmacol. Exp. Ther. 297 (2001) 326–
17 (2002) 1–22. 337.
[37] A. Parikh, E.M. Gillam, F.P. Guengerich, Drug metabo- [52] D.F. McGinnity, R.J. Riley, Predicting drug pharmacoki-
lism by Escherichia coli expressing human cytochromes netics in humans from in vitro metabolism studies,
P450, Nat. Biotechnol. 15 (1997) 784–788. Biochem. Soc. Trans. 29 (2001) 135–139.
[38] F.P. Guengerich, Cytochrome P450 proteins and potential [53] J.M. Margolis, R.S. Obach, Impact of nonspecific binding
utilization in biodegradation, Environ. Health Perspect. 103 to microsomes and phospholipid on the inhibition of
(1995) 25–28. cytochrome P4502D6: implications for relating in vitro
[39] J.T.M. Buters, M. Shou, J.P. Hardwick, K.R. Korzekwa, inhibition data to in vivo drug interactions, Drug Metab.
F.J. Gonzalez, cDNA-directed expression of human cyto- Dispos. 31 (2003) 606–611.
chrome P450 CYP1A1 using Baculovirus, Drug Metab. [54] W. Tang, R.W. Wang, A.Y. Lu, Utility of recombinant
Dispos. 23 (1995) 696–701. cytochrome P450 enzymes: a drug metabolism perspective,
[40] C.A. Lee, S.H. Kandwell, T.A. Kost, C.J. Serabjit-Singh, Curr. Drug Metab. 6 (2005) 503–517.
CYP3A4 expressed by insect cells infected with a recombi- [55] S.A. Wrighton, J.C. Stevens, The human hepatic cyto-
nant baculovirus containing both CYP3A4 and human chromes P450 involved in drug metabolism, Crit. Rev.
NADPH-cytochrome P450 reductase is catalytically similar Toxicol. 22 (1992) 1–21.
to human liver microsomal CYP3A4, Arch. Biochem. [56] I. Sekine, N. Saijo, Polymorphisms of metabolizing
Biophys. 319 (1995) 157–167. enzymes and transporter proteins involved in the clearance
[41] A. Yu, H. Dong, D. Lang, R.L. Haining, Characterization of anticancer agents, Ann. Oncol. 12 (2001) 1515–1525.
of dextromethorphan O- and N-demethylation catalyzed by [57] K.T. Kivisto, H.K. Kroemer, M. Eichelbaum, The role of
highly purified recombinant human CYP2D6, Drug Metab. human cytochrome P450 enzymes in the metabolism of
Dispos. 29 (2001) 1362–1365. anti-cancer agents: implications for drug interactions, Br. J.
[42] L. Chen, J.T. Buters, J.P. Hardwick, S. Tamura, B.W. Clin. Pharmacol. 40 (1995) 523–530.
Penman, F.J. Gonzalez, C.L. Crespi, Coexpression of [58] K.M.L. Crommentuyn, J.H.M. Schellens, J.D. vander
cytochrome P4502A6 and human NADPH-P450 oxidore- Berg, J.H. Beijnen, In vitro metabolism of anti-cancer
ductase in the baculovirus system, Drug Metab. Dispos. 25 drugs, methods and applications: paclitaxel, docetaxel,
(1997) 399–405. tamoxifen and ifosfamide, Cancer Treat. Rev. 24 (1998)
[43] J. Grogan, M. Shou, E.A. Andrusiak, S. Tamuro, J.T.M. 345–366.
Buters, F.J. Gonzalez, K.R. Korzekwa, Cytochrome P450 [59] M. Stiborova, C.A. Bieler, M. Wiessler, E. Frei, The
2A1, 2E1, and 2C9 cDNA-expression by insect cells and anticancer agent ellipticine on activation by cytochrome
partial purification using hydrophobic chromatography, P450 forms covalent DNA adducts, Biochem. Pharmacol.
Biochem. Pharmacol. 50 (1995) 1509–1515. 62 (2001) 1675–1684.
[44] H. Yamazaki, M. Nakajima, M. Nakamura, S. Asahi, N. [60] C. Rodriguez-Antona, M. Ingelman-Sundberg, Cyto-
Shimada, E.M. Gillam, F.P. Guengerich, T. Shimada, T. chrome P450 pharmacogenetics and cancer, Oncogene 25
Yokoi, Enhancement of cytochrome P-450 3A4 catalytic (2006) 1679–1691.
14 K. Purnapatre et al. / Cancer Letters 259 (2008) 1–15

[61] D. Yao, S. Ding, B. Burchell, C.R. Wolf, T. Friedberg, metabolism by CYP2C8 and CYP3A4, J. Pharmacol. Sci.
Detoxication of Vinca alkaloids by human P450 CYP3A4- 97 (2005) 83–90.
mediated metabolism: implications for the development of [76] A. Nannelli, A. Messina, S. Marini, S. Trasciatti, V. Longo,
drug resistance, J. Pharmacol. Exp. Ther. 294 (2000) 387– P.G. Gervasi, Effects of the anticancer dehydrotarplatin on
395. cytochrome P450 and antioxidant enzymes in male rat
[62] B. Rochat, J.M. Morsman, G.I. Murray, W.D. Figg, H.L. tissues, Arch. Toxicol. 81 (2007) 479–487.
McLeod, Human CYP1B1 and anticancer agent metabo- [77] R.H. Elbekia, A.O. El-Kadi, Transcriptional activation and
lism: mechanism for tumor-specific drug inactivation?, J post-transcriptional modification of CYPlal by arsenite,
Pharmacol. Exp. Ther. 296 (2001) 537–541. cadmium, and chromium, Toxicol. Lett. 172 (2007) 106–
[63] S.S. Dehal, D. Kupfer, CYP2D6 catalyzes tamoxifen 4- 119.
hydroxylation in human liver, Cancer Res. 57 (1997) 3402– [78] C.D. Scripture, A. Sparreboom, W.D. Figg, Modulation of
3406. cytochrome P450 activity: implications for cancer therapy,
[64] H.K. Crewe, L.M. Notley, R.M. Wunsch, M.S. Lennard, Lancet Oncol. 6 (2005) 780–789.
E.M. Gillam, Metabolism of tamoxifen by recombinant [79] S. Price, W. Bordogna, R. Braganza, R.J. Bull, H.J. Dyke,
human cytochrome P450 enzymes: formation of the 4- S. Gardan, M. Gill, N.V. Harris, R.A. Heald, M. van den
hydroxy, 4 0 -hydroxy and N-desmethyl metabolites and Heuvel, P.M. Lockey, J. Lloyd, A.G. Molina, A.G. Roach,
isomerization of trans-4-hydroxytamoxifen, Drug Metab. F. Roussel, J.M. Sutton, A.B. White, Identification and
Dispos. 30 (2002) 869–874. optimisation of a series of substituted 5-pyridin-2-yl-thio-
[65] H.K. Crewe, S.W. Ellis, M.S. Lennard, G.T. Tucker, phene-2-hydroxamic acids as potent histone deacetylase
Variable contribution of cytochromes P450 2D6, 2C9 (HDAC) inhibitors, Bioorgan. Med. Chem. Lett. 17 (2007)
and 3A4 to the 4-hydroxylation of tamoxifen by human 363–369.
liver microsomes, Biochem. Pharmacol. 53 (1997) 171– [80] M. Stiborova, M. Rupertova, H.H. Schmeiser, E. Frei,
178. Molecular mechanisms of antineoplastic action of an
[66] L.H. Patterson, G.I. Murray, Tumor cytochrome P450 and anticancer drug elipticine, Biomed. Pap. Med. Fac. Univ.
drug activation, Curr. Pharm. Des. 8 (2002) 1335–1447. Palacky Olomouc Czech Repub. 150 (2006) 13–23.
[67] J. Kajita, E. Fuse, T. Kuwabara, H. Kobayashi, The [81] M.M. Malingre, D.J. Richel, J.H. Beijnen, H. Rosing, F.J.
contribution of cytochrome P450 to the metabolism of Koopman, W.W. Tem Bokkel Huinink, M.E. Schot, J.H.
tegafur in human liver, Drug Metab. Pharmacokinet. 18 Schellens, Coadministration of cyclosporine strongly
(2003) 303–309. enhances the oral bioavailability of docetaxel, J. Clin.
[68] K. Ikeda, K. Yoshisue, E. Matsushima, S. Nagayama, K. Oncol. 19 (2001) 1160–1166.
Kobayashi, C.A. Tyson, K. Chiba, Y. Kawaguchi, Bioac- [82] P.H. Rooney, C. Telfer, M.C. McFadyen, W.T. Melvin,
tivation of tegafur to 5-fluorouracil is catalyzed by cyto- G.I. Murray, The role of cytochrome P450 in cytotoxic
chrome P-450 2A6 in human liver microsomes in vitro, bioactivation: future therapeutic directions, Curr. Cancer
Clin. Cancer Res. 6 (2000) 4409–4415. Drug Targets 4 (2004) 257–265.
[69] P.A. Jacobson, K. Green, A. Birnbaum, R.P. Remmel, [83] Z. Desta, X. Zhao, J.G. Shin, D.A. Flockhart, Clinical
Cytochrome P450 isozymes 3A4 and 2B6 are involved in significance of the cytochrome P450 2C19 genetic polymor-
the in vitro human metabolism of thiotepa to TEPA, phism, Clin. Pharmacokinet. 41 (2002) 913–958.
Cancer Chemother. Pharmacol. 49 (2002) 461–467. [84] S. Lee, I.P. van der Heiden, J.A. Goldstein, R.H.N. van
[70] D. McKillop, A.D. McCormick, A. Millar, G.S. Miles, P.J. Schaik, A new CYP3A5 variant, CYP3A5*11, is shown to
Phillips, M. Hutchison, Cytochrome P450-dependent be defective in Nifedipine metabolism in a recombinant
metabolism of gefitinib, Xenobiotica 35 (2005) 39–50. cDNA expression system, Drug Metab. Dispos. 35 (2007)
[71] T. Cresteil, B. Monsarrat, J. Dubois, M. Sonnier, P. 67–71.
Alvinerie, F. Gueritte, Regioselective metabolism of taxoids [85] S. Daigo, Y. Takahashi, M. Fujieda, N. Ariyoshi, H.
by human CYP3A4 and 2C8: structure–activity relation- Yamazaki, W. Koizumi, S. Tanabe, K. Saigenji, S. Nagay-
ship, Drug Metab. Dispos. 30 (2002) 438–445. ama, K. Ikeda, Y. Nishioka, T. Kamataki, A novel mutant
[72] T. Kawashiro, K. Yamashita, X.J. Zhao, E. Koyama, M. allele of the CYP2A6 gene (CYP2A6*11) found in a cancer
Tani, K. Chiba, T. Ishizaki, A study on the metabolism of patient who showed poor metabolic phenotype towards
etoposide and possible interactions with antitumor or tegafur, Pharmacogenetics 12 (2002) 299–306.
supporting agents by human liver microsomes, J. Pharma- [86] Y.M. Kim, S.H. Yoo, R.Y. Kang, M.J. Kim, Y.Y. Bae,
col. Exp. Ther. 286 (1998) 1294–1300. Y.K. Lee, S.J. Jeon, K.J. Chon, S.M. Shin, S.G. Kim, K.H.
[73] M.V. Relling, J. Nemec, E.G. Schuetz, J.D. Schuetz, F.J. Park, I.J. Son, Identifying drugs needing pharmacogenetic
Gonzalez, K.R. Korzekwa, O-demethylation of epipodo- monitoring in a Korean hospital, Am. J. Health Syst.
phyllotoxins is catalyzed by human cytochrome P450 3A4, Pharm. 64 (2007) 166–175.
Mol. Pharmacol. 45 (1994) 352–358. [87] B.K. Park, M. Pirmohamed, N.R. Kitteringham, The role
[74] G. Vassal, C. Pondarre, I. Boland, C. Cappelli, A. Santos, of cytochrome P450 enzymes in hepatic and extrahepatic
C. Thomas, E. Lucchi, K. Imadalou, F. Pein, J. Morizet, A. human drug toxicity, Pharmacol. Ther. 68 (1995) 385–424.
Gouyette, Preclinical development of camptothecin deriv- [88] W. Tang, Miller, in: Z. Yan Caldwell (Ed.), Methods in
atives and clinical trials in pediatric oncology, Biochimie 80 Pharmacology and Toxicology: Optimization in Drug
(1998) 271–280. Discovery: In vitro Methods, Humana Press, Totowa, NJ,
[75] R. Taniguchi, T. Kumai, N. Matsumoto, M. Watanabe, K. 2004, pp. 369–383.
Kamio, S. Suzuki, S. Kobayashi, Utilization of human liver [89] S. Wenzlaff, M.L. Cote, C.H. Bock, S.J. Land, S.K. Santer,
microsomes to explain individual differences in paclitaxel D.R. Schwartz, A.G. Schwartz, CYP1A1 and CYP1B1
K. Purnapatre et al. / Cancer Letters 259 (2008) 1–15 15

polymorphisms and risk of lung cancer among never [97] M. Karlgren, M. Ingelman-Sundberg, Tumour-specific
smokers: a population-based study, Carcinogenesis 26 expression of CYP2W1: its potential as a drug target in
(2005) 2207–2212. cancer therapy, Expert Opin Ther. Targets 11 (2007) 61–67.
[90] T.M. Sissung, D.K. Price, A. Sparreboom, W.D. Figg, [98] J.G. Jiang, C.L. Chen, J.W. Card, S. Yang, J.X. Chen, X.N.
Pharmacogenetics and regulation of human cytochrome Fu, Y.G. Ning, X. Xiao, D.C. Zeldin, D.W. Wang,
P450 1B1: implications in hormone-mediated tumor metab- Cytochrome P450 2J2 promotes the neoplastic phenotype
olism and a novel target for therapeutic intervention, Mol. of carcinoma cells and is up-regulated in human tumors,
Cancer Res. 4 (2006) 135–150. Cancer Res. 65 (2005) 4707–4715.
[91] G.A. Potter, L.H. Patterson, E. Wanogho, P.J. Perry, P.C. [99] R.D. Bruno, V.C. Njar, Targeting cytochrome P450
Butler, T. Ijaz, K.C. Ruparelia, J.H. Lamb, P.B. Farmer, enzymes: a new approach in anti-cancer drug development,
L.A. Stanley, M.D. Burke, The cancer preventative agent Bioorg. Med. Chem. 15 (2007) 5047–5060.
resveratrol is converted to the anticancer agent piceatannol [100] K.K. Deeb, D.L. Trump, C.S. Johnson, Vitamin D
by the cytochrome P450 enzyme CYP1B1, Br. J. Cancer 86 signaling pathways in cancer: potential for anticancer
(2002) 774–778. therapeutics, Nat. Rev. Cancer 7 (2007) 684–700.
[92] S. Sale, R.G. Tunstall, K.C. Ruparelia, P.C. Butler, G.A. [101] S. Sundaram, M.J. Beckman, A. Bajwa, J. Wei, K.M.
Potter, W.P. Steward, A.J. Gescher, Effects of the potential Smith, G.H. Posner, D.A. Gewirtz, QW-1624F2-2, a
chemopreventive agent DMU-135 on adenoma develop- synthetic analogue of 1,25-dihydroxyvitamin D3, enhances
ment in the ApcMin+ mouse, Invest. New Drugs. 24 (2006) the response to other deltanoids and suppresses the
459–464. invasiveness of human metastatic breast tumor cells, Mol.
[93] D. Pal, A.K. Mitra, CYP3A4 and MDR mediated interac- Cancer Ther. 5 (2006) 2806–2814.
tions in drug therapy, Clin. Res. Regul. Affairs 23 (2006) [102] K. Kojima, K. Nagata, T. Matsubara, Y. Yamazoe, Broad
125–163. but distinct role of pregnane X receptor on the expression
[94] H.J.G.D. van den Bongard, R.W. Sparidans, D.J.P. of individual cytochrome P450s in human hepatocytes,
Critchley, J.H. Beijnen, J.H.M. Schellens, Pharmacokinetic Drug Metab. Pharmacokinet. 22 (2007) 276–286.
drug–drug interaction of the novel anticancer agent E7070 [103] J. Chen, X.X. Yang, M. Huang, Z.P. Hu, M. He, W. Duan,
and acenocoumarol, Invest. New Drugs 22 (2004) 151–158. E. Chan, F.S. Sheu, X. Chen, S.F. Zhou, Small interfering
[95] Y. Sai, R. Dai, T.J. Yang, K.W. Krausz, F.J. Gonzalez, RNA-mediated silencing of cytochrome P450 3A4 gene,
H.V. Gelboin, M. Shou, Assessment of specificity of eight Drug Metab. Dispos. 34 (2006) 1650–1657.
chemical inhibitors using cDNA-expressed cytochromes [104] M. Izquierdo, Short interfering RNAs as a tool for cancer
P450, Xenobiotica 30 (2000) 327–343. gene therapy, Cancer Gene. Ther. 12 (2005) 217–227.
[96] N. Ahmad, H. Mukhtar, Cytochrome p450: a target for [105] S.l. Pai, Y.Y. Lin, B. Macaes, A. Meneshian, C.F. Hung,
drug development for skin diseases, J. Invest. Dermatol. T.C. Wu, Prospects of RNA interference therapy for
123 (2004) 417–425. cancer, Gene Ther. 13 (2006) 464–477.

You might also like