You are on page 1of 14

Clinical and Translational Oncology

https://doi.org/10.1007/s12094-020-02325-7

REVIEW ARTICLE

Drug‑metabolizing enzymes: role in drug resistance in cancer


G. Kaur1   · S. K. Gupta1 · P. Singh1 · V. Ali1 · V. Kumar1 · M. Verma1

Received: 1 November 2019 / Accepted: 18 February 2020


© Federación de Sociedades Españolas de Oncología (FESEO) 2020

Abstract
Although continuous researches are going on for the discovery of new chemotherapeutic agents, resistance to these antican-
cer agents has made it really difficult to reach the fruitful results. There are many causes for this resistance that are being
studied by the researchers across the world, but still, success is far because there are several factors that are going along
unattended or have been studied less. Drug-metabolizing enzymes (DMEs) are one of these factors, on which less study has
been conducted. DMEs include Phase I and Phase II enzymes. Cytochrome P450s (CYPs) are major Phase I enzymes while
glutathione-S-transferases (GSTs), UDP-glucuronosyltransferases (UGTs), dihydropyrimidine dehydrogenases are the major
enzymes belonging to the Phase II enzymes. These enzymes play an important role in detoxification of the xenobiotics as well
as the metabolism of drugs, depending upon the tissue in which they are expressed. When present in tumorous tissues, they
cause resistance by metabolizing the drugs and rendering them inactive. In this review, the role of these various enzymes in
anticancer drug metabolism and the possibilities for overcoming the resistance have been discussed.

Keywords  Drug-metabolizing enzymes (DMEs) · Drug metabolism · Drug resistance · DME inhibitors · Prodrugs

Introduction to treat cancer. Drug resistance is well known to us since


very old times. It is a phenomenon when a disease becomes
At present, cancer is one of those diseases on which exten- resistant to pharmaceutical treatments. The concept was first
sive research is going on. It has become the topic of con- noticed in bacteria against some antibiotics after that other
cern because of its fatal nature and the increasing number diseases were also found to show this phenomenon. Cancer
of cancer cases and deaths all over the world. Although a is one among them. Cancerous cells can acquire resistance
huge number of resources are invested for the development against cytotoxic drugs in many ways [3]. Some of the major
of preventive, diagnostic and therapeutic strategies for the mechanisms responsible for the resistance against anticancer
control of cancer, it is difficult to tackle it with the current chemotherapeutic agents are acquired mutations (primary
knowledge. The causes of cancer are very vast and so is and secondary), drug metabolism and inactivation, cross-
its prevention and cure [1]. In the 1990s, the research was talk between kinases signaling, alteration in regulation of
mainly focused on the development of chemotherapeu- cell cycle and checkpoints, blocked apoptosis, cancer stem
tic agents; those were at that time used as the first line of cells, immune system evasion, DNA damage and repair, epi-
therapy, until later on many heterocyclic compounds were genetic modifications like DNA methylation, histone modi-
approved by FDA for treating cancer. After that, the research fication, etc. [4].
diverted towards the development of various agents that Drug resistance is a major pitfall to cancer therapy as
could overcome various resistance mechanisms (Fig. 1) most of the chemotherapy will lead to resistance after long
against the already existing anticancer drugs [2]. The devel- use [4]. In the last few decades, studies have shown that
opment of various resistances has made it more difficult cancer cells differ in their metabolism from the normal
cells. These metabolic changes are one of the main reasons
for normal cells transforming into cancerous cells and also
* M. Verma a cause for the resistance to various types of chemothera-
malkhey.verma@cup.edu.in; malkhey@yahoo.com peutic drugs. The changed expression and activity of drug-
1
Department of Biochemistry, School of Basic and Applied
metabolizing enzymes play a considerable role in the drug
Sciences, Central University of Punjab, Bathinda 151001, resistance due to which metabolic and signaling pathways
India

13
Vol.:(0123456789)
Clinical and Translational Oncology

Fig. 1  Various causes of anti-


cancer drug resistance

become altered in cancerous cells [5]. These drug-metab- types of cancers cause rapid turnover and the elimination of
olizing enzymes have been broadly divided into two cat- the drug before reaching to the target [7]. Some of the Phase
egories—Phase I and Phase II drug-metabolizing enzymes. II drug-metabolizing enzymes that will be discussed in this
Phase I enzymes include the oxidases, dehydrogenases, review are GSTs, UGTs and DPDs.
deaminases and hydrolases. Phase II enzymes comprise
UDP-glucuronosyl transferases (UGTs), sulfotransferases Cytochrome P450s
(SULTs), glutathione transferases (GSTs), N-acetyl trans-
ferases (NSTs), etc., that generate water-soluble compounds Cytochrome P450s (CYPs) are a large group of enzymes
by the conjugation of products of Phase I reactions or parent present in the membrane of the endoplasmic reticulum.
compounds with suitable functional groups (Fig. 2). These They are terminal oxidases and belong to the heme-thiolate
compounds can be easily eliminated. The Phase I enzymes multigene family. CYPs need molecular oxygen and cou-
that play a major role in drug metabolism are the oxidases- pling to NADPH reductase. They take part in oxidative
Cytochrome P450 [6]. As they take part in the metabolism of reactions involved in the metabolism of many of the mar-
various anticancer drugs, their elevated expression in various keted drugs [8]. Cytochrome P450s have been evolved to

Fig. 2  Phase II enzymatic reac-


tions in drug metabolism

13
Clinical and Translational Oncology

protect the organisms against toxic compounds [9]. How- reductase; Class II P450s use NADPH-P450 reductase hav-
ever, cytochrome P450-mediated biotransformation may ing FAD/FMN and Class III does not need a separate pro-
result in metabolic stimulation of environmental chemical tein for reduction [15]. The typical structure of cytochrome
compounds leads to the production of carcinogens which is P450 consists of two domains—one having a majorly
called ‘lethal synthesis’ [10]. CYPs play an important role in α-helix structure and is about 70% of the protein and the
detoxification of xenobiotics as well as various endogenous other formed mainly of β-sheet. There are three conserved
compounds such as bile acids, steroids, prostaglandins, leu- residues among the P450 superfamily: (i) cysteine, present
kotrienes and unsaturated fats (Table 1) [11]. in heme binding region, coordinates the iron present in the
CYP enzymes are broadly divided into two classes: Class heme group; (ii) glutamine and (iii) arginine, both present
I and Class II. The Class I is composed of well-conserved in the α-helix [15].
members CYP1A1, CYP1A2, CYP2E1, and CYP3A4; not CYPs catalyze many oxidative reactions like CYP2C8
having important functional polymorphisms and are actively is involved in particular hydroxylation, N-demethylations
involved in the metabolism of drugs and procarcinogens. and N-de-ethylations. CYP2C8 can catalyze substrates of
Class II is composed of highly polymorphic CYP2B6, different sizes and structures, because of its structure. It is
CYP2C9, CYP2C19, and CYP2D6 which actively partici- involved in the metabolism of anti-microtubule agent pacli-
pate in drug metabolism but not in procarcinogen metabo- taxel, which is converted to 6α-hydroxy paclitaxel. CYP3A4
lism [4]. CYPs are divided into families and subfamilies, also metabolizes paclitaxel to 39-phenyl-hydroxypaclitaxel.
drug-metabolizing enzymes being confined to subfamilies 1, These metabolites are further metabolized to 6α, 3′-p-dihy-
2, 3 and 4 [8]. There are about 57 genes and 58 pseudogenes droxy paclitaxel [16]. CYP3A4 is involved in the metabo-
in human differing in their substrate specificity and tissue lism of many of the known drugs including anticancer drugs
expression profile (Table 1). There are about 18 distinct like taxol, tamoxifen, ifosfamide, VP-16 and Vinca alkaloids
P450s in the liver, out of which only 10 are from families 1, [17]. The anticancer drugs flutamide, mitoxantrone, pacli-
2, and 3; metabolize the most of the marketed drugs. These taxel and docetaxel have also been proposed to be the sub-
are CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, strates of the isozyme CYP1B1 as they inhibited the activity
CYP2C19, CYP2D6, CYP2E1, CYP3A4 and CYP3A5. of this enzyme in a competitive manner [18].
Some of these enzymes are also expressed in extrahepatic
tissues like kidneys, lungs, small intestine, brain, skin and
placenta. CYP3 group is the most abundant enzyme belong- Catalytic cycle of CYP enzymes
ing to the Phase I drug-metabolizing enzymes [8].
The enzyme CYP2C8 has been extensively studied for its CYP enzymes have a wide range of substrate specificity and
crystal structure and was resolved [13]. The enzyme exists catalyze a number of redox reactions such as hydroxylation,
as a dimer and the approximate molecular weight is 54 kDa. heteroatom oxygenation, dealkylation, epoxidation, desatu-
The structure contains a heme moiety as a prosthetic group ration, and several others [19, 20]. CYP mediated monoox-
that can bind carbon monoxide very tightly. The name P450 ygenation reactions incorporate one oxygen atom into the
has been given because the microsomes with carbon mon- substrate, while the other oxygen atom is reduced to water
oxide exposure show a strong absorption band at 450 nm, [19]. A characteristic CYP enzyme catalyzed reaction is:
and P stands for pigment. The substrates of P450s range RH + O2 + 2e− + 2H+ → ROH + H2 O
from smaller (ethylene, MW = 28) to larger (cyclosporine,
MW = 1201) molecules [14]. P450s can be classified into The amount of energy required to break the bond between
three classes according to the redox partner they need, Class the oxygen molecules is provided by the transfer of elec-
I P450s need two redox cofactors, ferredoxin (an iron–sul- trons from NADPH through FAD and FMN mainly by
fur protein) and a FAD-containing NAD(P)H-ferredoxin enzyme NADPH cytochrome P450 reductase or in certain

Table 1  The classification of human CYP enzymes on the basis of major substrate classes [12]
Class of substrates Human CYP enzymes

Sterols 1B1, 7A1, 7B1, 8B1, 11A1, 11B1, 11B2, 17A1, 19A1, 21A2, 27A1, 39A1, 46A1, 51A1
Xenobiotics 1A1, 1A2, 2A6, 2A13, 2B6, 2C8, 2C9, 2C18, 2C19, 2D6, 2E1, 2F1, 3A4, 3A5, 3A7
Fatty acids 2J2, 4A11, 4B1, 4F12
Eicosanoids 4F2, 4F3, 4F8, 5A1, 8A1
Vitamins 2R1, 24A1, 26A1, 26B1, 26C1, 27B1
Unknown 2A7, 2S1, 2U1, 2W1, 3A43, 4A22, 4F11, 4F22, 4V2, 4X1, 4Z1, 20A1, 27C1

13
Clinical and Translational Oncology

cases, cytochrome b5 in the smooth endoplasmic reticu- the role of CYP1 enzymes in the bioactivation of these com-
lum. Cytochrome b5 is the smaller partner of eukaryotic pounds was provided by the experiments carried out using
cytochrome P450 enzymes which act as an allosteric the inhibitors of CYP1, i.e, acacetin and α-naphthoflavone
modulator for numerous CYPs involved in the regulation [28].
of mammalian steroidogenesis. In reality, cytochrome b5 Cyclophosphamide, ifosfamide and trofosfamide belong
transfers the second electron to particular CYPs faster than to the oxazaphosphorine class of prodrugs. They are bio-
CYP reductase [19]. Finally, the electrons are transferred activated by CYP enzymes to alkylating anticancer agents.
from NADPH to CYP inside the mitochondria [9]. There Cyclophosphamide when introduced initially in 1960 as
are several donors of oxygen atom for CYP enzymes such an anticancer drug, it was supposed to be activated by the
as hydroperoxides, peracids, perborate, percarbonate, perio- enzyme phosphoramidase to bis-(chloroethyl) amine. But
date, chlorite, iodosobenzene and N-oxides. However, after later it was found to be oxidized by hepatic CYP enzymes
passing through a series of reactions CYP iron–oxygen com- to 4-hydroxycyclophosphamide. So, cyclophosphamide may
plex produce intermediates such as ferric-peroxo anion spe- be considered one of the pioneering prodrugs designed for
cies ­(FeIIIOO−), ferric-hydroperoxo species (Fe(III)–OOH) improving the therapeutic index of highly toxic therapeutic
and Fe(III)–(H2O2) complex in CYP mediated monooxy- agents [29]. 1,4-Bis-([2-(dimethylamino-N-oxide) ethyl]
genation reactions [20]. amino) 5,8-dihydroxy anthracene-9,10-dione (AQ4N), a
Cytochrome P450 enzymes contribute a major part to the bioreductive prodrug also require activation by CYP2S1 and
metabolizing enzymes in humans, catalyzing the biotrans- CYP2W1 in tumor tissues and is converted to topoisomer-
formation of various exogenous compounds, foods, drugs ase II inhibitor AQ4 [1,4-bis-5,8-dihydroxy-anthracene-
and also the endogenous compounds [21]. CYP1, 2 and 3 9,10-dione]. Other prodrugs activated by CYPs include
families of the CYP isozymes have also been found to cata- procarbazine, tegafur, and thiotepa. They are activated by
lyze the biotransformation of various anticancer drugs [22]. the enzymes in the liver [30].
CYPs mainly expressed in the liver (accounts for 90% of
the total body) [23] have also been shown to be conserved Glutathione S‑transferases
in cancer cells and also been reviewed in tumors and can-
cer cell lines [24]. So, it indicates that CYPs may have a Glutathione S-transferases (GSTs) belong to the family of
role in detoxification and biotransformation of anticancer Phase II detoxification enzymes, which protects cellular
drugs. Interestingly, many reports underline the coinciding macromolecules from the attack of reactive electrophiles.
substrate specificity of CYP3A and ABCB1 (ATP-binding Precisely, GSTs catalyze the reaction in which glutathione
cassette B1) transporters [25]. This combination of mecha- (GSH) is conjugated with the wide variety of exogenous
nisms that have possibly co-evolved over billions of years and endogenous xenobiotics. This conjugation process is
may possibly lead to drug resistance by reducing the con- the first step of the mercapturic acid pathway, and it helps
centration of active drugs in both, systemic circulation and in the elimination of toxic compounds from the cell. On the
the target cells [26]. basis of location, GSTs are divided into two super-family
Various CYPs have been found to be inhibited by vari- members: the cytosolic and the membrane-bound micro-
ous chemicals like CYP3A4 have been found to be inhib- somal family members. Again, the cytosolic GSTs due to
ited by ritonavir. As CYP3A is known to metabolize anti- significant genetic polymorphisms in human populations are
cancer drugs like paclitaxel and docetaxel, reducing their divided into six classes, which share nearly 30% sequence
oral availability, co-administration of ritonavir with these identity, and are labeled by Greek letters α, μ, ω, π, θ and ζ.
drugs has been shown to enhance their bioavailability [17]. Though having few genetic similarities, the affinities of these
α-Naphthoflavone has also been shown to inhibit CYP1A GSTs for the substrates differ due to genetic mutation and
isoforms, in vitro. Furafylline, which is a methylxanthine have different substrate specificity (Table 2).
analogue also known to inhibit CYP1A2-mediated reactions, The GST is a dimeric enzyme and each subunit of the
where it is a potent inhibitor while the effect is not prominent enzyme has an active site with two distinctive functional
in case of CYP1A1 [27]. regions: hydrophilic G-Site (GSH binding site) and hydro-
Like other anticancer drug-metabolizing enzymes, the phobic H-site; the binding site of structurally different elec-
overexpression of CYPs in tumor cells can also be utilized trophilic substrates. The G-site is the conservative site, while
by designing prodrugs that are activated by CYPs (Table 3). H-site has affinity for variable substrates that regulate the
To this approach a series of chalcone derivatives have been specificity of GST isozymes [32]. The link between two
synthesized that showed anti-proliferative activities in subunits of GST constructs the intrasubunit binding site for
human breast cancer cell lines expressing CYP1A1 and ligands, which facilitates the GSH conjugate formed by one
CYP1B1 and also showed lesser toxicity towards the normal subunit to be insulated from the adjacent subunit and hence
breast cell line having no CYP expression. The evidence for limit the product inhibition [33]. In the reactions catalyzed

13
Clinical and Translational Oncology

Table 2  Different classes of Compounds Xenobiotics GSTs


GSTs with their substrates [31]
Exogenous Polycyclic aromatic hydrocarbons π GSTs
α,β unsaturated aldehydes π GSTs
Molecule with epoxide group θ GSTs
Chemotherapeutic agents α GSTs, π GSTs
Endogenous O-Quinones of catecholamines and dopamine μ GSTs
Prostaglandins α GSTs, μ, microsomal GSTs
Lipid peroxidation products generated by reactive μ GSTs, μ, microsomal
oxygen species

by GSTs, GSH does the nucleophilic attack on electrophilic association with the Kelch-like ECH-associated-protein 1
substrates after bringing the substrate into nearby vicinity (Keap1) and cullin3, where Nrf2 is degraded by ubiquitina-
with GSH at the active site of the enzyme. The distribution tion. But, oxidative stress interrupts the cysteine residue in
of GSTs among organ is also diverse and depends on age, the Keap1 and so disruption of (Keap1) and cullin3 ubiq-
sex and species. GSTA1, GSTA2 and GSTM1 are highly uitination system occur. This further leads to the translo-
expressed during entire life, while GSTP1 are expressed in cation of Nrf2 from the cytoplasm to nucleus, and in the
embryonic and fetal organs that are diminished at the end of nucleus Nrf2 carry out the transcription of antioxidative
prenatal period [34]. The allocation of GSTs also depends on genes. The nucleophilic attack by GSTs on the sulfur atom
the organ, like GSTA1 is expressed in the liver, kidney and of GSH on the electrophilic group of several xenobiotics
testis; GSTP1 in lung and brain; GSTM2 is highly expressed could reduce the activity of the drugs and favor their elimi-
in the brain while GSTM1 in expressed at the highest level nation by making them water soluble. Hence, the catalytic
in the liver [35]. It is found that the expression of GSTs activity of GSTs could play a key role in the detoxification
also varies from tissue to tissue within the same organ, e.g., of anticancer drugs such as cyclophosphamide, chlorambu-
GSTA1 and GSTA2 is found in the duodenum and small cil, carmustine, melphalan, cisplatin, ifosfamide, busulfan,
intestine, while GSTP1 is found in the stomach [36]. The thiotepa and mitoxantrone [38].
distribution of GSTs also depends on the cell division stage, Besides its role as the detoxifying agent, GSTs also regu-
like the erythroleukemia cells (K562) have GSTP1 in its late apoptosis by acting as the inhibitor of c-Jun N-termi-
proliferating stage and during differentiation, this GST is nal kinase 1 (JNK1), a kinase involved in stress response,
replaced by GSTA1 [37]. The expression of GSTs in the apoptosis, and cellular proliferation. Firstly, the GST was
human body is also regulated by chemicals and xenobiotics. predicted to be ligand because of its binding capability with
The expression of GSTs in the cancer cell lines makes them the substrates, which are not linked to the enzymatic mecha-
resistant to drugs, had led the idea to use GSTs as the marker nisms and hence entitled as ‘ligandin’ [31]. Recently, the
of tumor progression. regulatory role of the π and μ classes of GSTs have been
Among all GSTs, GSTP1 expression is highly coupled identified in the mitogen-activated protein (MAP) kinase
with the development of drug resistance in tumor cell lines. pathway, one of the signaling pathways in the cell. In nor-
The GST families include many isozymes with wide sub- mal cellular condition, low JNK activity was observed due
strate specificities, but the polymorphisms within these to the formation of GSTπ–JNK complex, but under oxida-
isozymes not only contribute to inter-individual differ- tive stresses, the complex is degraded, and further GSTπ is
ences in response to the xenobiotics but also to anti-cancer oligomerized and induction of apoptosis occurs [39]. The
drugs [31]. In the tumor cells drug resistance develops after JNK is also activated either by immunodepletion or inhibi-
induction and activation of the efflux transporter proteins, tion of GSTπ. So, for the treatment of cancer, the researchers
the mutation in the topoisomerase II, enhancement in DNA are focusing on the GSTs inhibitors, which might break the
repair, alteration in genes which are responsible for apopto- GST–JNK complexes and then JNK will be free to carry out
sis and at last by expressing the detoxifying enzymes. The the apoptosis in the cancer cells.
alkylating agents which act as the anticancer agents are the The efflux pumps were found to be over-expressed in sev-
substrates of the GST. The evidence showed that the over- eral cancers and they mediate the extrusion of drugs from
expression of GSTs and GSH in tumors is associated with the cells. Similar associations have been found between
the development of drug resistance. GSTs and efflux pumps which lead to a reduction in drug
The level of GST increases by the action of the nuclear accumulation and further triggers drug resistance. The com-
factor-erythroid 2 p45-related factor 2 (Nrf2). Under normal monly found efflux transporters are the ABC transporters
conditions, the Nrf2 is seized in the cytoplasm due to its Pgp, ABCG2 and mitoxantrone-resistance proteins (MRPs)

13
Clinical and Translational Oncology

[40]. Out of these transporters, the MRPs are involved in resistance. GST-activated prodrugs use the catalytic power
the transport of GSH, glucuronate or sulphate conjugates of these enzymes, particularly GSTP1-1, to activate them-
of xenobiotics that result from after the detoxification reac- selves into cytotoxic agents. Some of these molecules yield
tions carried out by Phase II conjugating enzymes such as promising results and are currently under clinical trials for
GSTs, sulfotransferases and UDP-glucuronosyltransferases the treatment of specific forms of advanced cancers. While
(UGT). GSTs coexpress with MRP result in drug resistance great strides have been made in overcoming drug resistance,
in the melanoma cells against vincristine [41], and also further investigation of both existing and novel strategies
against other anticancer drugs, i.e., chlorambucil, vincris- may be the approach necessary to surmount this phenom-
tine, ethacrynic acid, and etoposide [42]. The cis-acting enon [38, 53]. In recent years there has been particular
regulatory elements ARE, the promoter site for the antioxi- interest in the link between the GSTP1-1 isoenzyme and
dant enzymes, also play a central role in the coexpression of the MAPK pathway that mediates both stress and apoptotic
GSTs and MRPs. Nrf2 also regulates the expression of the cellular responses. In fact, several findings indicate that a
efflux pumps, MRP1 and MRP2 efflux pumps [40]. promising new strategy for tumor treatment may be the acti-
To cope with the drug resistance, the researchers devel- vation of this pathway through molecules that selectively
oped inhibitors of GSTs and the prodrugs (Table 3). One target GSTP1-1. After first attempts with the analogues of
of the most specific GSTs inhibitors is TER 199/TLK199 GSH, the search for new proapoptotic agents that do not
(γ-Glutamyl-S-(benzyl)-cysteinyl-R(-)-phenylglycine interact with the multidrug transporters is yielding encour-
diethyl ester). TLK199 has a phenylglycine moiety which aging results and are currently underway at the preclinical
interacts precisely with a hydrophobic pocket close to the level [38].
GSH-binding site of GST and enhances the effect of anti- In the past few years, along with the GSH-conjugating
tumor drugs, for example, melphalan and chlorambucil catalytic activity, GSTs are additionally found to have the
[43]. Another GST inhibitor, the 7-nitro-2,1,3-benzoxadi- non-enzymatic activity. It regulates several signaling path-
azole derivative NBDHEX binds to the cytosolic GST and ways related to cell division, differentiation and cell death
induce the dissociation of JNK–GST complex that leads to [54]. Among different GST isozymes, GSTP1-1 found
the activation of the JNK-mediated pathway [44]. Prodrugs to be highly expressed in cancer cells where it provides
are the class of cytotoxic agents that comprise molecules drug resistance to those cells [55]. So, the GST inhibitors,
which exploit the catalytic capability of GSTs, and these especially for GSTP1-1, may provide the chemotherapy
drugs have different affinities for the various isoenzymes for cancer and other diseases linked with abnormal cell
of GST. Findlay and his colleagues synthesized a prodrug proliferation.
called ­[O2-{2,4-dinitro-5-[4-(N-methylamino) benzoyloxy]
phenyl}1-(N,N-dimethylamino)diazen-1-ium-1,2-diolate]
(PABA/NO) which had the capability to release cytotoxic Uridine diphospho‑glucuronosyltransferases
levels of nitric oxide [45]. PABA/NO is an anticancer pro-
drug which transfers its aryl groups to nucleophiles (i.e., Like GSTs, uridine diphospho-glucuronosyltransferases
GSH) along with cogeneration of ions which instinctively (UGTs) also belong to Phase II enzyme classification and it
release NO more efficiently at an appropriate pH after catalyzes the reactions related to glucuronidation. In glucu-
metabolization by GSTs [46]. Later, in vitro transport studies ronidation, UGTs transfer glucuronosyl group from uridine
revealed that PABA/NO are effluxed by MRP1, and hence 5′-diphospho-glucuronic acid (UDPGA) to the compounds
the case of drug resistance was observed [45]. Additionally, having oxygen, nitrogen, sulfur, or carboxyl functional
PABA/NO are found to be not very much soluble and are group. UGTs are the undoubtedly one of the important con-
suddenly hydrolyzed. After this, Cui and colleagues syn- jugating enzymes of the liver, which ease the elimination of
thesized pH-controlled NO donors which are 4-Aryl-1,3,2- several endobiotics (thyroid and steroid hormones, bilirubin,
oxathiazolylium-5-olate (OZO) derivatives [47]. The OZO bile acids and retinoids), as well as xenobiotics (environmen-
derivatives react with GSH only in the presence of GST tal chemicals, pollutants and drugs). Due to glucuronidation,
and further the reactions affect the catalytic properties of these compounds become highly soluble in blood and are
GSTs and activation of JNK to carry out the apoptosis in easily excreted from the body via kidney. The process of
the cancer cells. Few other prodrugs include GST-activated glucuronidation is carried out by UGTs in the liver, intesti-
alkylating generators, for example, TER286 [48], ethacra- nal mucosa and kidney; and the resultant glucuronides are
platin [49], 2-crotonyloxymethyl-2-cyclohexenone (COMC) secreted in bile and urine. These are highly expressed in the
[50], and brostallicin [51, 52]. liver but are also widespread in the gastrointestinal tract,
The catalytic activity and binding properties of some kidney, lungs, brain, and reproductive tissues [56]. Similar
members of the GST superfamily have been exploited to to GSTs, human UGT superfamily is divided into three sub-
generate anticancer agents efficient at overcoming drug families: UGT1A, 2A, 2B, which after alternative splicing

13
Clinical and Translational Oncology

and translation further categorized into different functional of UGT1A1 gene, i.e., UGT1A1*28 results in reduced excre-
proteins. The UGT1A is located on chromosome 2q37 and tion of irinotecan metabolites, and due to the accumulation
codes for nine functional proteins (UGT1A1, UGT1A3, of these metabolites in blood, the development of neutrope-
UGT1A4, UGT1A5, UGT1A6, UGT1A7, UGT1A8, nia occurs [65].
UGT1A9, and UGT1A10), while the UGT2 family is located The role of UGTs in breast cancer, the most common can-
on chromosome 4q13 and encodes for eight functional pro- cer, is also detected. For the growth of breast, the estrogen is
teins (UGT2A1, UGT2B4, UGT2B7, UGT2B10, UGT2B11, required, but excessive estrogen level in cell signaling cause
UGT2B15, UGT2B17, and UGT2B28). This mechanism of breast cancer. The relation between the activity of UGT1A1
alternative splicing is conserved in human, mouse, and rat alleles and breast cancer is mixed type because of results
UGTs [57]. from different studies. Four studies reported link between
UGTs are type-I transmembrane glycoproteins mainly low activity UGT1A1 alleles (i.e., *28, *34) and increased
located within the smooth endoplasmic reticulum (ER), breast cancer threat [66], while this increased risk was not
although some isoforms can be found in the nuclear enve- identified by few other studies [67]. A case study of Nigerian
lope [58]. The detoxification enzymes cytochromes P450, women confirmed a negative association between low activ-
CYP (Phase I enzyme) and UGTs (Phase II enzyme) are ity UGT1A1*28 allele and breast cancer [68].
found to be related, as CYP add the functional groups to A case study showed the potential involvement of the
the molecules that have to be metabolized by UGTs [59]. UGT1A1 promoter TATA box polymorphism and endome-
Apart from the process of detoxification, few times the glu- trial cancer risk [69]. The results from this study revealed
curonidation can activate metabolites, like in case of mor- that women having homozygous (*28/*28) or heterozy-
phine which is converted into morphine 3- and 6-glucuron- gous (*1/*28) UGT1A1*28 alleles had a reduced risk of
ides and proved to be more potent analgesic than morphine endometrial cancer in comparison to women homozy-
[60]. Rarely, it happens that glucuronidation metabolizes gous for UGT1A1*1 allele (*1/*1). Normally, UGT1A*1
substrates into their toxic forms, such as steroid D-ring allele of UGT1A1 in the uterus show high activity toward
glucuronides, which could be the mediator for intrahepatic 2-hydroestradiol, an antiproliferative metabolite of estradiol,
cholestasis of pregnancy [61]. while UGT1A1*28 allele diminishes the capability of the
The UGT1A1 gene codes for an enzyme which conjugates body to glucuronidate 2-hydroestradiol in the endometrium
bilirubin to facilitate its removal from the human body. The and hence reduces the risk of developing endometrial cancer
UGT1A1 gene comprises a TA repeat promoter polymor- [69].
phism, located 41 nucleotides upstream of the translational Since last 15 years, numerous case–control studies have
start site adjacent to a presumed TATA box, is responsible evaluated that the polymorphism in UGT genes (primarily
for the decreased enzymatic activity of UGT1A1. In the UGT1A and UGT2B genes), increase the risk of cancers
normal state, the serum bilirubin has the antioxidant and (bladder, breast, colorectal, endometrial, esophageal, head
cytoprotective properties, but in the individuals with seven and neck, liver, lungs, prostate, and thyroid). Few reports
thymine-adenine (TA) repeats the condition of hyperbiliru- proved that polymorphism in a single UGT allele might give
binemia occurs. This elevation in bilirubin level may further rise to multiple cancers. In the coming future, the assured
cause brain damage in neonates and could also modulate association of several cancers with UGTs polymorphism
susceptibility to oxidative damage and cancer. The polymor- requires detailed studies with a large number of sample size
phism in UGT1A1 gene causes the production of UGT1A1 in different situations to end the conflicts between the results
variants, and these variants may cause jaundice. Presently, of different studies.
around 135 genetic variants of UGT1A1 have been reported
in which the most common variant allele is UGT1A1*28 Dihydropyrimidine dehydrogenases (DPDs)
[62]. This variant has seven TA repeats within the TATA
box promoter region in comparison to 6 TA repeats of the DPD is also known as dihydrouracil dehydrogenase, dihy-
wild-type allele [63]. drothymine dehydrogenase and uracil reductase. DPD plays
UGT1A1*28 is also associated with drug toxicity, an important role in catabolizing various nitrogen-contain-
which leads to neutropenia following intravenous and iri- ing molecules like pyrimidines: uracil and thymine. Along
notecan therapy. Irinotecan (brand name Camptosar), is with these endogenous nitrogenous bases, DPD also play
an anti-cancer drug for colorectal cancer and the role of an important role in 5-FU (5-fluorouracil, an anticancer
UGT1A1 enzyme in its metabolization is highly studied. drug) metabolism (Table 3). As it is the major enzyme in
This drug is a topoisomerase I inhibitor and is converted 5-FU catabolism, metabolizing about 85% of the adminis-
into its active form (SN-38) in the human body. SN-38 is tered amount, into a compound 5-fluoro-5,6-dihydrouracil
further inactivated and detoxified by enzyme encoded by the (5-FUH2). This inactivation of 5-FU reduces its anabolism
UGT1A1 gene [64]. But the presence of modified structure and thus its overall metabolism. So, the increased level of

13
Clinical and Translational Oncology

DPD in tumors results in the nitrogen heterocycle based anti- case report was about a female patient with rigorous fluoro-
cancer drug resistance [2]. The enzyme in its cytosolic form pyrimidine-induced neurotoxicity, a familial DPD deficiency
is present in a many of the tissues with major activity being [74]. Various case reports identified the relation between
found in liver, catalyzes the reduction of the pyrimidines, DPD deficiency and fluoropyrimidine toxicities [75]. For
uracil and thymine in an NADPH dependent manner, leading the identification of the molecular background of this heredi-
to the 5-FUH2 formation (Fig. 3) [70]. Researchers are able tary defect, cloning of DPYD stage was set [76]. The most
to measure 5-FU metabolites with the innovation of new common familial DPD deficiency was recognized which is
techniques like high-pressure liquid chromatography [71]. related to a fault in the processing of the DPD precursor
On the basis of these findings it is confirmed that DPD is the mRNA, specifically an exon-skipping abnormality that cause
first rate-limiting enzyme involved in the catabolic process the decline of 165 nucleotides from the fully spliced mRNA
of 5-FU, but it did not show the clinical importance of these [77]. One year later, two different researchers published the
results [71]. Shortly after that in clinical literature, they iden- first DNA sequence level recognition of this DPYD varia-
tified the key role of DPD activity in catabolism of fluoropy- tion; they represented similar results within two different
rimidine and the consequences of DPD deficit for fluoropy- families. Both identified a single nucleotide polymorphism
rimidine related severe adverse events (AEs). The first case (SNP) inside DPYD that depicts a new splice site, which
related to this catabolic enzyme reported that patient had results into exon 14 skipping. The resulting DPD protein has
oral ulceration, neurotoxicity and severe myelosuppression, loss of its function completely [78]. Now this DPYD variant
and further analysis of case confirmed that patient had famil- is generally called as DPYD*2A (or called as rs3918290 or
ial pyrimidinemia and pyrimidinuria as a result of uracil c.1905 + 1G > A). Around 2% of Caucasians of European
and thymine in blood and urine [72]. The removal of 5-FU descent possess this DPYD*2A allele. 50% reduction of
in cancer patients takes place via DPD dependent metabo- DPD activity is exhibited in heterozygous carriers allele,
lism. Based on this study it was found that the removal of besides that is very rare (~ 1:1000), homozygous DPYD*2A
more than 80% of systemic 5-FU, along with 95% of the patients determine complete DPD deficiency [79].
ultimate metabolite via urine is solely responsible for the Knowing the DPD activity is essential for understanding
catabolic pathway (Fig. 3) [73]. Diasio and his colleagues the link between DPD deficiency and severe fluoropyrimi-
issued a case report after the confirmation of DPD as the dine-related AEs. DPD activity follows a circadian rhythm
vital metabolic enzyme causative for 5-FU elimination, the and its activity was recorded at peak in the midnight with the

Table 3  Drug-metabolizing enzymes overexpressed in various tumors and their substrates and prodrugs

Sr. no. Enzymes Tumor overexpression Resistance against drugs Prodrugs

1. CYPs Breast cancer, colon cancer, stomach Paclitaxel, taxol, tamoxifen, VP-16, Chalcone derivatives, cyclophosphamide,
cancer flutamide, mitoxantrone docetaxel ifosfamide, trofosfamide
2. GSTs Colorectal tumor, prostate cancer, ovar- Cis-platin, chlorambucil adriamicin, Acrylic acid, sulphonamide derivative of
ian cancer, mantle cell lymphomas, melphalan doxorubicin, ANS-etoposide, ANS-
breast cancer cells DOX
3. UGTs Human colorectal tumor, human Irinotecan sorafenib, PR-104A, –
esophageal tumor, lung cancer cells GS-1101 methotrexate
breast cancer cells, chronic lympho-
cytic leukemia
4. DPDs Lung cancer, colorectal cancer, breast 5-Fluorouracil 1-Ethoxymethyl 5-fluorouracil, tegafur
cancer

Fig. 3  Inactivation and elimination of 5-FU by the action of an onate (FUPA) and the third enzyme beta-ureidopropionase (UPB1)
enzyme dihydropyrimidine dehydrogenase (DPD) that converted into activity finally terminates in urinary elimination of fluoro-beta-ala-
5-dihydrofluorouracil (5-DHFU). The second enzyme dihydropy- nine (FBAL)
rimidinase (DPYS) causes the formation of fluoro-beta-ureidopropi-

13
Clinical and Translational Oncology

trough in the early afternoon [80]. An interesting discovery differ among healthy and malignant tissues of the identical
was related to the difference in the systemic 5-FU disclosure organ [86].
through persisting uninterrupted infusion, with a change of DPD enzyme has been isolated and purified from various
2.3-fold from peak to trough in the level of systemic 5-FU sources like pig, rat, bovine and human. It is homodimer
during the 5-day course [81]. Despite that there is little of each monomer being 111 kDa. It has a large number of
agreement on the rhythm’s physiological consequence [82]. redox cofactors. These are one FAD, one FMN, 16 acid-
Generally, it was comprehended that hepatic DPD activity labile sulfur and 16 non-heme-iron atoms, present on each
is causative for the bulk of 5-FU clearance [74], but at the (1025 amino acid residue long) subunit. According to the
population level it follows an ordinary division [83]. The amino acid sequence analysis, the enzyme most likely has
DPD activity from the frozen liver samples was measured four [4Fe–4S] cluster in each subunit, two being in an N-ter-
by radiolabeled biochemical assay. They showed a power- minal Cys-rich stretch, and the other two in a 50 amino acid
ful connection among level of DPD protein expression and long stretch at the C-terminus. According to two-site ping-
enzyme activity [83]. Alternate work has been done to esti- pong kinetic mechanism, there are two separate binding sites
mate the DPD activity required to correlate mRNA expres- for NADPH/NADP+ and pyrimidine/5,6-dihydropyrimidine,
sion with DPD activity. But, later the conclusion of the study respectively (Fig. 4) [70].
was demolished as it was understood that elevated expres- DPD is an important regulatory enzyme in the metabo-
sion of mRNA might not recompense for inactive enzyme. lism of the anticancer drug 5-FU [2]. 5-FU is counted in the
Thus, DPD mRNA levels might not provide satisfactory con- most widely used drugs and has been in use for a long time.
sideration of 5-FU elimination. Throughout the examination In 1994, for the first time, DPD activity was shown to be
of peripheral blood mononuclear cells (PBMCs), researchers significantly related to the sensitivity of 5-FU. An experi-
have desired to understand population difference in DPD ment was performed to check the co-relation between DPD
activity, however significant correlation among PBMC DPD activity and 5-FU sensitivity, in which 19 human tumor cell
activity and hepatocyte DPD activity R2 < 0.6 [84], that cat- lines, from digestive tract, breast, head and neck cancer cells
egorize the significance of PBMC DPD activity studies fur- were taken into the investigation. Marked differences were
ther difficult. Besides that, the relationship among PBMC shown between the cell lines. DPD activity was shown in
DPD activity and systemic 5-FU clearance established weak all cell lines except four. The cell lines showed the inverse
associations than PBMC DPD activity and hepatic DPD relation between DPD activity and 5-FU sensitivity [87].
activity [84, 85]. Finally, PBMC DPD activity established Further several studies show that DPD can be a major fac-
larger difference than established in examination of hepatic tor for variation in effectiveness of anti-tumor agent 5-FU
DPD activity, wherein PBMC DPD confirmed difference of among various tumors as well as people. Few studies on
activity among 8- to 21-fold based on the study [85]. Thus, DPD have shown that there is variable expression of DPD
the gain of PBMC DPD activity in illustrating the popula- in various tumors. These varying levels of DPD in tumors
tion difference of endogenous DPD activity remains tough are in accordance with the variance in the responses of the
to understand. Beside that DPD activity is acknowledged to tumors towards 5-FU [2]. In another study, head and neck

Fig. 4  Deactivation of 5-FU by
DPD

13
Clinical and Translational Oncology

squamous cell carcinoma (HNSCC) were used to check the There are several other anticancer drugs involved in the
relation between DPD expression and cytotoxic activity of cure of cancer through different mechanisms. Angiostatin
5-FU. DPD cDNA was constitutively expressed and 5-FU K13 is a potent inhibitor of endothelial cell growth and
cytotoxicity was examined. The cytotoxicity was shown to angiogenesis and acts as an anticancer agent that targets
be decreased with the overexpression of DPD [88]. VEGF and prevents angiogenesis [93]. Staurosporine is a
Knowing the role of DPD in the varying sensitivity to powerful inhibitor that stops angiogenesis by suppressing
5-FU and also their negative correlation, it became a point up-regulated VEGF expression in tumor cells and prevents
of interest for researchers to consider inhibiting DPD in their angiogenesis [94]. A well-documented inhibitor of
order to eliminate the varying responses to 5-FU. Few years microsomal UGT1 A4 is 1-naphthol UGTI-3. The mode of
ago, attempts have been made to synthesize the inhibitors action of 1-naphthol includes its higher glucuronidation via
of DPD. New fluoropyrimidine drugs have been introduced UGT1 A6, which results in the decreased level of UDP-glu-
using the DPD inhibition agents called as dihydropyrimidine curonic acid and a higher level of UDP and plays an impor-
dehydrogenase inhibitory fluoropyrimidines (DIFs). tant role in competitive inhibition of UDP-glucuronic acid
There are four DIF drugs: UFT, ethynyluracil, S-1, and [95]. Recently, kinase inhibitors against UGTs such as lapat-
BOF-A2. These drugs differ in both type and degree of DPD inib UGTI-8, pazopanib UGTI-9, regorafenib UGTI-10, and
inhibition produced. UFT was the first DIF drug to be syn- sorafenib UGTI-11 are reported to inhibit the UGTs enzyme
thesized. It is a combination of uracil, the naturally occur- [96]. Pilocarpine and sulfaphenazole both inhibit CYP2C9,
ring pyrimidine, and the 5-FU prodrug-tegafur. Actually, it the ketoconazole inhibits CYP3A4, and quinidine inhib-
is competitive inhibition of DPD. In the presence of excess its CYP2D6 vigorously [97]. Cimetidine is a well-known
uracil, there is a competition at DPD level, so the 5-FU will inhibitor of P450-linked reaction. It has been shown that
not be degraded rapidly and its presence will be prolonged. cimetidine interacts with human hepatic CYP1A, 2C, 2D,
This effect is reversible, so it is better than the earlier inhibi- 2E, and 3A forms and inhibits P-450-linked reactions [98].
tors [89]. Ethynyluracil (eniluracil) is a pyrimidine structur- Gemcitabine (2′,2′-difluoro-2′-deoxycytidine, dFdC) is intra-
ally similar to uracil and 5-FU. It is an irreversible inhibitor cellularly metabolized to 5′diphosphate (dFdCDP). It inhib-
of DPD. In its presence, the oral bioavailability of 5-FU its ribonucleotide reductase and an encouraging anticancer
is increased to 100%, which was earlier hampered due to drug [99]. HDAC inhibitors such as vorinostat and romidep-
the variable activity of DPD enzyme. Phase I and Phase II sin have been approved by the Food and Drug Administra-
trials have been done, which shows the increased half-life tion, USA, as anticancer drugs [100]. The metal-binding
and decreased clearance of 5-FU. So, eniluracil is a promis- compounds such as clioquinol (a zinc ionophore) is another
ing approach that allows reliable and safer oral administra- anticancer drug that targets cyclin D1 gene at the levels both
tion of 5-FU and overcomes the resistance caused by the transcriptional and post-transcriptional regulation in cancer
overexpression of DPD [90]. S-1 is a combination of three cells. It encourages mRNA degeneration of the cyclin D1
drugs—tegafur (prodrug); 5-chloro-2,4-dihydroxypyridine gene controlled by miR-302C. It means the metal-binding
[CDHP] (a DPD inhibitor); and potassium oxalate in a molar compounds have an impact on gene expression at different
ratio of 1:0.4:1. This combination along with the previous regulatory levels. In this case, the post-transcriptional gene
inhibitor provides the relief from GI-tract toxicity. Potassium regulation can be a probable target for chemotherapy [101].
oxalate selectively inhibits 5-FU phosphorylation caused by
the enzyme, orotate phosphoribosyltransferase, in the gas-
trointestinal tract and not in tumor cells [89, 91]. BOF-A2 Conclusion
is still another attempt towards improved fluoropyrimidine.
In this drug combination, 1-ethoxymethyl 5-fluorouracil Drug-metabolizing enzymes are one of the important fac-
(EM-FU)-the prodrug is combined with the DPD inhibitor tors causing resistance against various anticancer drugs. In
3-cyano-2,6-dehydropyrimidine (CNDP) in a molar ratio of the last two decades, people have worked on DMEs, but
1:1. EM-FU is resistant to degradation and is converted to yet their role in the anticancer drug resistance has not been
5-FU by liver microsomes [89]. TAS-114 also inhibits DPD fully understood. In this review, significant data have been
enzyme, it targets the metabolism of 5-FU to enhance its collected to show the role DMEs play in drug resistance.
anticancer activity and also modulates the catabolic path- So, while designing new drugs, the role of these enzymes
way to enhance the systemic availability of 5-FU. It affects should be taken into consideration. Various inhibitors of
the DPD enzyme moderately and in irreversible manner. It these enzymes have been discussed that can be used as a
enhanced the bioavailability of 5-FU on co-administration combination with the anticancer drugs to overcome the
with capecitabine in mice and improved the therapeutic resistance. The overexpression of these enzymes in cancers
index of capecitabine [92]. can be turned into an advantage by the use of prodrugs that
are being activated by these enzymes. In that way, they will

13
Clinical and Translational Oncology

help in making the anticancer drugs more specific to the of xenobiotic and natural chemicals. Chem Res Toxicol.
cancer cells. As these enzymes are overexpressed in cancer 2014;28(1):38–42.
12. Nelson DR, Zeldin DC, Hoffman SM, Maltais LJ, Wain HM,
cells, the prodrug will remain inactive in the normal cells Nebert DW. Comparison of cytochrome P450 (CYP) genes from
decreasing the cytotoxicity of the drugs on normal cells. the mouse and human genomes, including nomenclature recom-
Hence, there is still a need for research to be done in this mendations for genes, pseudogenes and alternative-splice vari-
area of anticancer drug resistance and should be taken as a ants. Pharmacogenetics Genomics. 2004;14(1):1–8.
13. Schoch GA, Yano JK, Wester MR, Griffin KJ, Stout CD, John-
potential factor of consideration while designing anticancer son EF. Structure of human microsomal cytochrome P450 2C8
drugs. Evidence for a peripheral fatty acid binding site. J Biol Chem.
2004;279(10):9497–503.
Acknowledgements  The authors acknowledge the support received 14. Testa B, Krämer SD. The biochemistry of drug metabolism–an
from the Central University of Punjab, Bhatinda, India, in writing this introduction: part 2. Redox reactions and their enzymes. Chem
manuscript. Biodiversity. 2007;4(3):257–405.
15. Graham-Lorence S, Peterson JA. P450s: structural similarities
Authors’ contribution  MV conceived and designed the research work. and functional differences. FASEB J. 1996;10(2):206–14.
GK and SKG both have first authorship. PS and VA further discussed 16. Backman JT, Filppula AM, Niemi M, Neuvonen PJ. Role of
and improved the review. All authors read and approved the final cytochrome P450 2C8 in drug metabolism and interactions.
manuscript. Pharmacol Rev. 2016;68(1):168–241.
17. Hendrikx JJ, Lagas JS, Rosing H, Schellens JH, Beijnen JH,
Schinkel AH. P-glycoprotein and cytochrome P450 3A act
Compliance with ethical standard  together in restricting the oral bioavailability of paclitaxel. Int J
Cancer. 2013;132(10):2439–47.
Conflict of interest  The authors declare no conflicting interests. 18. Rochat B, Morsman JM, Murray GI, Figg WD, McLeod HL.
Human CYP1B1 and anticancer agent metabolism: mechanism
Ethical approval  This article does not contain any studies with human for tumor-specific drug inactivation? J Pharmacol Exp Ther.
participants or animals performed by any of the authors. 2001;296(2):537–41.
19. Isin EM, Guengerich FP. Complex reactions catalyzed by
Informed consent  For this type of study, formal consent is not required. cytochrome P450 enzymes. Biochimica et Biophysica Acta
(BBA)-General Subjects. 2007;1770(3):314–29.
20. Hrycay EG, Bandiera SM. Monooxygenase, peroxidase and per-
oxygenase properties and reaction mechanisms of cytochrome
P450 enzymes. Monooxygenase, peroxidase and peroxyge-
References nase properties and mechanisms of cytochrome P450. Cham:
Springer; 2015. p. 1–61.
1. Kumar S, Ahmad MK, Waseem M, Pandey AK. Drug targets for 21. Ingelman-Sundberg M, Daly AK, Oscarson M, Nebert DW.
cancer treatment: an overview. Med Chem. 2015;5:115–23. Human cytochrome P450 (CYP) genes: recommendations
2. Pathania S, Bhatia R, Baldi A, Singh R, Rawal RK. Drug metabo- for the nomenclature of alleles. Pharmacogenet Genomics.
lizing enzymes and their inhibitors’ role in cancer resistance. 2000;10(1):91–3.
Biomed Pharmacother. 2018;105:53–65. 22. Iyer L, Ratain MJ. Pharmacogenetics and cancer chemotherapy.
3. Housman G, Byler S, Heerboth S, Lapinska K, Longacre M, Sny- Eur J Cancer. 1998;34(10):1493–9.
der N, Sarkar S. Drug resistance in cancer: an overview. Cancers. 23. Krishna DR, Klotz U. Extrahepatic metabolism of drugs in
2014;6(3):1769–92. humans. Clin Pharmacokinet. 1994;26(2):144–60.
4. Leary M, Heerboth S, Lapinska K, Sarkar S. Sensitization of 24. Doherty MM, Michael M. Tumoral drug metabolism: per-
drug resistant cancer cells: a matter of combination therapy. Can- spectives and therapeutic implications. Curr Drug Metab.
cers. 2018;10(12):483. 2003;4(2):131–49.
5. Rochat B. Importance of influx and efflux systems and xenobiotic 25. Huang L, Wring SA, Woolley JL, Brouwer KR, Serabjit-Singh
metabolizing enzymes in intratumoral disposition of anticancer C, Polli JW. Induction of P-glycoprotein and cytochrome
agents. Curr Cancer Drug Targets. 2009;9(5):652–74. P450 3A by HIV protease inhibitors. Drug Metab Disposition.
6. Jain AK, Jain S, Rana AC. Metabolic enzyme considerations in 2001;29(5):754–60.
cancer therapy. Malays J Med Sci. 2007;14(1):10. 26. Cummings J, Zelcer N, Allen JD, Yao D, Boyd G, Maliepaard
7. Doehmer J, Goeptar AR, Vermeulen NP. Cytochromes P450 and M, Friedberg TH, Smyth JF, Jodrell DI. Glucuronidation as a
drug resistance. Cytotechnology. 1993;12(1–3):357–66. mechanism of intrinsic drug resistance in colon cancer cells:
8. Penner N, Woodward C, Prakash C. Drug metabolizing enzymes contribution of drug transport proteins. Biochem Pharmacol.
and biotransformation reactions. In: Zhang D, Surapaneni S, edi- 2004;67(1):31–9.
tors. ADME enabling technologies in drug design and develop- 27. Pelkonen O, Mäeenpäeä J, Taavitsainen P, Rautio A, Raunio
ment. Hoboken: Wiley; 2012. p. 545–65. H. Inhibition and induction of human cytochrome P450 (CYP)
9. Guengerich FP, Waterman MR, Egli M. Recent structural enzymes. Xenobiotica. 1998;28(12):1203–53.
insights into cytochrome P450 function. Trends Pharmacol Sci. 28. Ruparelia KC, Zeka K, Ijaz T, Ankrett DN, Wilsher NE, Butler
2016;37(8):625–40. PC, Tan HL, Lodhi S, Bhambra AS, Potter GA, Arroo RR. The
10. Wahlang B, Falkner KC, Cave MC, Prough RA. Role of synthesis of chalcones as anticancer prodrugs and their bioac-
cytochrome P450 monooxygenase in carcinogen and chemo- tivation in CYP1 expressing breast cancer cells. Med Chem.
therapeutic drug metabolism. Advances in Pharmacology. Cam- 2018;14(4):322–32.
bridge: Academic Press; 2015. p. 1–33. 29. Stella V, Borchardt R, Hageman M, Oliyai R, Maag H, Tilley J,
11. Rendic S, Guengerich FP. Survey of human oxidoreductases editors. Prodrugs: challenges and rewards. Springer Science &
and cytochrome P450 enzymes involved in the metabolism Business Media; 2007.

13
Clinical and Translational Oncology

30. Nishida CR, Lee M, De Montellano PR. Efficient hypoxic activa- 48. Vergote I, Finkler N, Del Campo J, Lohr A, Hunter J, Matei
tion of the anticancer agent AQ4N by CYP2S1 and CYP2W1. D, Kavanagh J, Vermorken JB, Meng L, Jones M, Brown G.
Mol Pharmacol. 2010;78(3):497–502. Phase 3 randomised study of canfosfamide (­ Telcyta®, TLK286)
31. Townsend DM, Tew KD. The role of glutathione-S-transferase versus pegylated liposomal doxorubicin or topotecan as third-
in anti-cancer drug resistance. Oncogene. 2003;22(47):7369. line therapy in patients with platinum-refractory or-resistant
32. Deponte M. Glutathione catalysis and the reaction mechanisms of ovarian cancer. Eur J Cancer. 2009;45(13):2324–32.
glutathione-dependent enzymes. Biochimica et Biophysica Acta 49. Ang WH, Khalaila I, Allardyce CS, Juillerat-Jeanneret L,
(BBA) Gen Subj. 2013;1830(5):3217–66. Dyson PJ. Rational design of platinum (IV) compounds to
33. Wu B, Dong D. Human cytosolic glutathione transferases: overcome glutathione-S-transferase mediated drug resistance.
structure, function, and drug discovery. Trends Pharmacol Sci. J Am Chem Soc. 2005;127(5):1382–3.
2012;33(12):656–68. 50. Hamilton DS, Zhang X, Ding Z, Hubatsch I, Mannervik B,
34. Raijmakers MT, Steegers EA, Peters WH. Glutathione S-trans- Houk KN, Ganem B, Creighton DJ. Mechanism of the glu-
ferases and thiol concentrations in embryonic and early fetal tis- tathione transferase-catalyzed conversion of antitumor 2-crot-
sues. Hum Reprod. 2001;16(11):2445–50. onyloxymethyl-2-cycloalkenones to GSH adducts. J Am Chem
35. Sherratt PJ, Hayes JD. Glutathione S-transferases. Enzyme Sys- Soc. 2003;125(49):15049–58.
tems Metab Drugs Other Xenobiotics. 2001;2002:319–52. 51. Lorusso D, Mainenti S, Pietragalla A, Ferrandina G, Foco G,
36. Coles BF, Chen G, Kadlubar FF, Radominska-Pandya A. Inter- Masciullo V, Scambia G. Brostallicin (PNU 166196), a new
individual variation and organ-specific patterns of glutathione minor groove DNA binder: preclinical and clinical activity.
S-transferase alpha, mu, and pi expression in gastrointestinal Expert Opin Investig Drugs. 2009;18(12):1939–46.
tract mucosa of normal individuals. Arch Biochem Biophys. 52. Pezzola S, Antonini G, Geroni C, Beria I, Colombo M, Brog-
2002;403(2):270–6. gini M, Mongelli N, Leboffe L, MacArthur R, Mozzi AF, Fed-
37. Schnekenburger M, Morceau F, Henry E, Blasius R, Dicato M, erici G. Role of glutathione transferases in the mechanism of
Trentesaux C, Diederich M. Transcriptional and post-transcrip- brostallicin activation. Biochemistry. 2009;49(1):226–35.
tional regulation of glutathione S-transferase P1 expression dur- 53. Ruzza P, Calderan A. Glutathione transferase (GST)-activated
ing butyric acid-induced differentiation of K562 cells. Leukemia prodrugs. Pharmaceutics. 2013;5(2):220–31.
Res. 2006;30(5):561–8. 54. Cho SG, Lee YH, Park HS, Ryoo K, Kang KW, Park J,
38. Sau A, Tregno FP, Valentino F, Federici G, Caccuri AM. Eom SJ, Kim MJ, Chang TS, Choi SY, Shim J. Glutathione
Glutathione transferases and development of new princi- S-transferase mu modulates the stress-activated signals by sup-
ples to overcome drug resistance. Arch Biochem Biophys. pressing apoptosis signal-regulating kinase 1. J Biol Chem.
2010;500(2):116–22. 2001;276(16):12749–55.
39. Adler V, Yin Z, Fuchs SY, Benezra M, Rosario L, Tew KD, Pin- 55. Tew KD, Monks A, Barone L, Rosser D, Akerman G, Montali
cus MR, Sardana M, Henderson CJ, Wolf CR, Davis RJ. Regula- JA, Wheatley JB, Schmidt DE. Glutathione-associated enzymes
tion of JNK signaling by GSTp. EMBO J. 1999;18(5):1321–34. in the human cell lines of the National Cancer Institute Drug
40. Meijerman I, Beijnen JH, Schellens JH. Combined action and Screening Program. Mol Pharmacol. 1996;50(1):149–59.
regulation of phase II enzymes and multidrug resistance pro- 56. Riches Z, Collier AC. Posttranscriptional regulation of uridine
teins in multidrug resistance in cancer. Cancer Treat Rev. diphosphate glucuronosyltransferases. Exp Opin Drug Metabol
2008;34(6):505–20. Toxicol. 2015;11(6):949–65.
41. Depeille P, Cuq P, Mary S, Passagne I, Evrard A, Cupissol D, 57. Caspersen CS, Reznik B, Weldy PL, Abildskov KM, Stark RI,
Vian L. Glutathione S-transferase M1 and multidrug resistance Garland M. Molecular cloning of the baboon UDP-glucurono-
protein 1 act in synergy to protect melanoma cells from vincris- syltransferase 1A gene family: evolution of the primate UGT1
tine effects. Mol Pharmacol. 2004;65(4):897–905. locus and relevance for models of human drug metabolism.
42. Depeille P, Cuq P, Passagne I, Evrard A, Vian L. Combined Pharmacogenetics Genomics. 2007;17(1):11–24.
effects of GSTP1 and MRP1 in melanoma drug resistance. Br J 58. Fry DJ, Wishart GJ. Apparent induction by phenobarbital of
Cancer. 2005;93(2):216. uridine diphosphate glucuronyltransferase activity in nuclear
43. Morgan AS, Ciaccio PJ, Tew KD, Kauvar LM, Ciaccio FJ. envelopes of embryonic-chick liver. Biochem Soc Trans.
Isozyme-specific glutathione S-transferase inhibitors potenti- 1976;4:265–6.
ate drug sensitivity in cultured human tumor cell lines. Cancer 59. Fremont JJ, Wang RW, King CD. Coimmunoprecipitation of
Chemother Pharmacol. 1996;37(4):363–70. UDP-glucuronosyltransferase isoforms and cytochrome P450
44. Federici L, Sterzo CL, Pezzola S, Di Matteo A, Scaloni F, Fed- 3A4. Mol Pharmacol. 2005;67(1):260–2.
erici G, Caccuri AM. Structural basis for the binding of the 60. Abbott FV, Palmour RM. Morphine-6-glucuronide: anal-
anticancer compound 6-(7-nitro-2, 1, 3-benzoxadiazol-4-ylthio) gesic effects and receptor binding profile in rats. Life Sci.
hexanol to human glutathione S-transferases. Cancer Res. 1988;43(21):1685–95.
2009;69(20):8025–34. 61. Vore M, Slikker W. Steroid D-ring glucuronides: a new class
45. Findlay VJ, Townsend DM, Saavedra JE, Buzard GS, Citro ML, of cholestatic agents. Trends Pharmacol Sci. 1985;1(6):256–9.
Keefer LK, Ji X, Tew KD. Tumor cell responses to a novel glu- 62. Strassburg CP. Pharmacogenetics of Gilbert’s syndrome. Phar-
tathione S-transferase–activated nitric oxide-releasing prodrug. macogenomics. 2008;9(6):703–15.
Mol Pharmacol. 2004;65(5):1070–9. 63. Hall D, Ybazeta G, Destro-Bisol G, Petzl-Erler ML, Di AR.
46. Saavedra JE, Srinivasan A, Bonifant CL, Chu J, Shanklin AP, Variability at the uridine diphosphate glucuronosyltransferase
Flippen-Anderson JL, Rice WG, Turpin JA, Davies KM, Keefer 1A1 promoter in human populations and primates. Pharmaco-
LK. The secondary amine/nitric oxide complex ion R2N [N (O) genetics. 1999;9(5):591–9.
NO]-as nucleophile and leaving group in SNAr reactions. J Org 64. Dean L. Irinotecan therapy and UGT1A1 genotype. Medical
Chem. 2001;66(9):3090–8. Genetics Summaries 2018;. National Center for Biotechnology
47. Cui H, Shen J, Lu D, Zhang T, Zhang W, Sun D, Wang PG. Information (US).
4-Aryl-1, 3, 2-oxathiazolylium-5-olate: a novel GST inhibitor 65. Liu X, Cheng D, Kuang Q, Liu G, Xu W. Association of
to release JNK and activate c-Jun for cancer therapy. Cancer UGT1A1* 28 polymorphisms with irinotecan induced
Chemother Pharmacol. 2008;62(3):509–15.

13
Clinical and Translational Oncology

toxicities in colorectal cancer: a meta-analysis in Caucasians. 5-fluorouracil levels with evidence for circadian variation of
Pharmacogenomics J. 2014;14(2):120. enzyme activity and plasma drug levels in cancer patients
66. Justenhoven C, Winter S, Dünnebier T, Hamann U, Baisch C, receiving 5-fluorouracil by protracted continuous infusion.
Rabstein S, Spickenheuer A, Harth V, Pesch B, Brüning T, Ko Cancer Res. 1990;50(1):197–201.
YD. Combined UGT1A1 and UGT1A6 genotypes together with 81. Petit E, Milano G, Lévi F, Thyss A, Bailleul F, Schneider M.
a stressful life event increase breast cancer risk. Breast Cancer Circadian rhythm-varying plasma concentration of 5-fluoroura-
Res Treatment. 2010;124(1):289–92. cil during a five-day continuous venous infusion at a constant
67. Clendenen T, Zeleniuch-Jacquotte A, Wirgin I, Koenig KL, Afa- rate in cancer patients. Cancer Res. 1988;48(6):1676–9.
nasyeva Y, Lundin E, Arslan AA, Axelsson T, Försti A, Hallmans 82. Jacobs BA, Deenen MJ, Pluim D, van Hasselt JC, Krähenbühl
G, Hemminki K. Genetic variants in hormone-related genes and MD, van Geel RM, de Vries N, Rosing H, Meulendijks D,
risk of breast cancer. PLoS ONE. 2013;8(7):e69367. Burylo AM, Cats A. Pronounced between-subject and circa-
68. Huo D, Kim HJ, Adebamowo CA, Ogundiran TO, Akang EE, dian variability in thymidylate synthase and dihydropyrimidine
Campbell O, Adenipekun A, Niu Q, Sveen L, Fackenthal JD, dehydrogenase enzyme activity in human volunteers. Br J Clin
Fackenthal DL. Genetic polymorphisms in uridine diphospho- Pharmacol. 2016;82(3):706–16.
glucuronosyltransferase 1A1 and breast cancer risk in Africans. 83. Lu Z, Zhang R, Diasio RB. Population characteristics of
Breast Cancer Res Treatment. 2008;110(2):367–76. hepatic dihydropyrimidine dehydrogenase activity, a key meta-
69. Duguay Y, McGrath M, Lépine J, Gagné JF, Hankinson bolic enzyme in 5-fluorouracil chemotherapy. Clin Pharmacol
SE, Colditz GA, Hunter DJ, Plante M, Têtu B, Bélanger Ther. 1995;58(5):512–22.
A, Guillemette C. The functional UGT1A1 promoter poly- 84. Fleming RA, Milano G, Thyss A, Etienne MC, Renée N, Sch-
morphism decreases endometrial cancer risk. Cancer Res. neider M, Demard F. Correlation between dihydropyrimidine
2004;64(3):1202–7. dehydrogenase activity in peripheral mononuclear cells and
70. Dobritzsch D, Schneider G, Schnackerz KD, Lindqvist Y. Crystal systemic clearance of fluorouracil in cancer patients. Cancer
structure of dihydropyrimidine dehydrogenase, a major determi- Res. 1992;52(10):2899–902.
nant of the pharmacokinetics of the anti-cancer drug 5-fluoroura- 85. Etienne MC, Lagrange JL, Dassonville O, Fleming R, Thyss
cil. EMBO J. 2001;20(4):650–60. A, Renee N, Schneider M, Demard F, Milano G. Population
71. Sommadossi JP, Gewirtz DA, Diasio RB, Aubert C, Cano JP, study of dihydropyrimidine dehydrogenase in cancer patients.
Goldman ID. Rapid catabolism of 5-fluorouracil in freshly iso- J Clin Oncol. 1994;12(11):2248–53.
lated rat hepatocytes as analyzed by high performance liquid 86. Guimbaud R, Guichard S, Dusseau C, Bertrand V, Aparicio
chromatography. J Biol Chem. 1982;257(14):8171–6. T, Lochon I, Chatelut E, Couturier D, Bugat R, Chaussade S,
72. Tuchman M, Stoeckeler JS, Kiang DT, O’Dea RF, Ramnaraine Canal P. Dihydropyrimidine dehydrogenase activity in normal,
ML, Mirkin BL. Familial pyrimidinemia and pyrimidinuria inflammatory and tumour tissues of colon and liver in humans.
associated with severe fluorouracil toxicity. N Engl J Med. Cancer Chemother Pharmacol. 2000;45(6):477–82.
1985;313(4):245–9. 87. Beck A, Etienne MC, Cheradame S, Fischel JL, Formento P,
73. Heggie GD, Sommadossi JP, Cross DS, Huster WJ, Diasio RB. Renee N, Milano G. A role for dihydropyrimidine dehydroge-
Clinical pharmacokinetics of 5-fluorouracil and its metabolites nase and thymidylate synthase in tumour sensitivity to fluoro-
in plasma, urine, and bile. Cancer Res. 1987;47(8):2203–6. uracil. Eur J Cancer. 1994;30(10):1517–22.
74. Diasio RB, Harris BE. Clinical pharmacology of 5-fluorouracil. 88. Yasumatsu R, Nakashima T, Uryu H, Masuda M, Hirakawa N,
Clin Pharmacokinet. 1989;16(4):215–37. Shiratsuchi H, Tomita K, Fukushima M, Komune S. The role
75. Beuzeboc P, Pierga JY, Stoppa-Lyonnet D, Etienne MC, Milano of dihydropyrimidine dehydrogenase expression in resistance
G, Fouillait P. Severe 5-fluorouracil toxicity possibly second- to 5-fluorouracil in head and neck squamous cell carcinoma
ary to dihydropyrimidine dehydrogenase deficiency in a breast cells. Oral Oncol. 2009;45(2):141–7.
cancer patient with osteogenesis imperfecta. Eur J Cancer. 89. Robert B, Diasio M. Clinical implications of dihydropyrimi-
1996;32(2):369–70. dine dehydrogenase inhibition. Phys Pract. 1997;7(3):13–21.
76. Yokota H, Fernandez-Salguero P, Furuya H, Lin K, McBride OW, 90. Schilsky RL, Kindler HL. Eniluracil: an irreversible inhibitor
Podschun B, Schnackerz KD, Gonzalez FJ. cDNA cloning and of dihydropyrimidine dehydrogenase. Expert Opin Investig
chromosome mapping of human dihydropyrimidine dehydroge- Drugs. 2000;9(7):1635–49.
nase, an enzyme associated with 5-fluorouracil toxicity and con- 91. Shigeishi H, Biddle A, Gammon L, Rodini CO, Yamasaki M,
genital thymine uraciluria. J Biol Chem. 1994;269(37):23192–6. Seino S, Sugiyama M, Takechi M, Mackenzie IC. Elevation
77. Meinsma R, Fernandez-Salguero P, Van Kuilenburg AB, Van in 5-FU-induced apoptosis in Head and Neck Cancer Stem
Gennip AH, Gonzalez FJ. Human polymorphism in drug metab- Cells by a combination of CDHP and GSK 3β inhibitors. J Oral
olism: mutation in the dihydropyrimidine dehydrogenase gene Pathol Med. 2015;44(3):201–7.
results in exon skipping and thymine uracilurea. DNA Cell Biol. 92. Yano W, Yokogawa T, Wakasa T, Yamamura K, Fujioka A,
1995;14(1):1–6. Yoshisue K, Matsushima E, Miyahara S, Miyakoshi H, Tagu-
78. Vreken PA, Van Kuilenburg AB, Meinsma R, Smit GP, Bakker chi J, Chong KT. TAS-114, a first-in-class dual dUTPase/DPD
HD, De Abreu RA, Van Gennip AH. A point mutation in an inhibitor, demonstrates potential to improve therapeutic effi-
invariant splice donor site leads to exon skipping in two unre- cacy of fluoropyrimidine-based chemotherapy. Mol Cancer
lated Dutch patients with dihydropyrimidine dehydrogenase Ther. 2018;17(8):1683–93.
deficiency. J Inherit Metab Dis. 1996;19(5):645–54. 93. Wahl ML, Kenan DJ, Gonzalez-Gronow M, Pizzo SV. Angio-
79. Amstutz U, Henricks LM, Offer SM, Barbarino J, Schellens JH, statin’s molecular mechanism: aspects of specificity and regu-
Swen JJ, Klein TE, McLeod HL, Caudle KE, Diasio RB, Schwab lation elucidated. J Cell Biochem. 2005;96(2):242–61.
M. Clinical Pharmacogenetics Implementation Consortium 94. Stepczynska A, Lauber K, Engels IH, Janssen O, Kabelitz
(CPIC) guideline for dihydropyrimidine dehydrogenase geno- D, Wesselborg S, Schulze-Osthoff K. Staurosporine and con-
type and fluoropyrimidine dosing: 2017 update. Clin Pharmacol ventional anticancer drugs induce overlapping, yet distinct
Ther. 2018;103(2):210–6. pathways of apoptosis and caspase activation. Oncogene.
80. Harris BE, Song R, Soong SJ, Diasio RB. Relationship 2001;20(10):1193.
between dihydropyrimidine dehydrogenase activity and plasma

13
Clinical and Translational Oncology

95. Landmann H, Proia DA, He S, Ogawa LS, Kramer F, Beißbarth 99. Pereira S, Fernandes PA, Ramos MJ. Mechanism for ribonucleo-
T, Grade M, Gaedcke J, Ghadimi M, Moll U, Dobbelstein M. tide reductase inactivation by the anticancer drug gemcitabine. J
UDP glucuronosyltransferase 1A expression levels determine the Comput Chem. 2004;25(10):1286–94.
response of colorectal cancer cells to the heat shock protein 90 100. Campas-Moya C. Romidepsin for the treatment of cutane-
inhibitor ganetespib. Cell death & disease. 2014;5(9):e1411. ous T-cell lymphoma. Drugs Today (Barcelona, Spain: 1998).
96. Miners JO, Chau N, Rowland A, Burns K, McKinnon RA, Mac- 2009;45(11):787–95.
kenzie PI, Tucker GT, Knights KM, Kichenadasse G. Inhibition 101. Zheng J, Benbrook DM, Yu H, Ding WQ. Clioquinol suppresses
of human UDP-glucuronosyltransferase enzymes by lapatinib, cyclin D1 gene expression through transcriptional and post-tran-
pazopanib, regorafenib and sorafenib: implications for hyper- scriptional mechanisms. Anticancer Res. 2011;31:2739–47.
bilirubinemia. Biochem Pharmacol. 2017;1(129):85–95.
97. Bourrié M, Meunier V, Berger Y, Fabre G. Cytochrome P450 Publisher’s Note Springer Nature remains neutral with regard to
isoform inhibitors as a tool for the investigation of metabolic jurisdictional claims in published maps and institutional affiliations.
reactions catalyzed by human liver microsomes. J Pharmacol
Exp Ther. 1996;277(1):321–32.
98. Knodell RG, Browne DG, Gwozdz GP, Brian WR, Guengerich
FP. Differential inhibition of individual human liver cytochromes
P-450 by cimetidine. Gastroenterology. 1991;101(6):1680–91.

13

You might also like