You are on page 1of 24

Coordination Chemistry Reviews 427 (2021) 213600

Contents lists available at ScienceDirect

Coordination Chemistry Reviews


journal homepage: www.elsevier.com/locate/ccr

Molecular probes for human cytochrome P450 enzymes: Recent


progress and future perspectives
Jingjing Wu a,⇑, Xiaoqing Guan b, Ziru Dai c, Rongjing He b, Xinxin Ding d, Ling Yang b,⇑, Guangbo Ge b,⇑
a
Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian 116044, China
b
Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
c
Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy
of Medical Sciences & Peking Union Medical College, Beijing 100193, China
d
Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA

a r t i c l e i n f o a b s t r a c t

Article history: The cytochrome P450 enzymes (P450s or CYPs) are a class of heme-containing monooxygenases respon-
Received 22 July 2020 sible for the oxidative metabolism of a wide range of endogenous substances and foreign chemicals.
Accepted 7 September 2020 Deciphering the physiological functions of CYPs and their relevance to human diseases requires reliable
tools for sensing target enzyme in complex biological systems. Over the past few decades, great efforts
have been made to develop isoform-specific probe substrates for human CYP(s) and use them in both
Keywords: basic researches and translational applications. The growing database of the ligands and 3D-structures
Cytochrome P450 enzymes (CYPs)
of human CYPs strongly facilitates the design and development of CYP probe substrates with improved
Probe substrates
Design principles
specificity. Herein, we summarize the recent progress in the development and applications of molecular
Inhibitor screening probes for human CYPs. The structural and catalytic features of human CYPs, as well as the design prin-
Biomedical applications ciples and strategies used in the development of CYP probe substrates, have been well summarized. Two
comprehensive lists of molecular probes for various human CYP enzymes, including non-optical and opti-
cal probe substrates, along with their structural information and kinetic parameters, are presented.
Finally, the current challenges and future perspectives in this field are proposed and highlighted. The
information and knowledge presented here provides practical tools for sensing CYP activities in complex
biological systems, exploring the relevance of CYPs to human diseases, as well as screening and charac-
terizing of CYP modulators.
Ó 2020 Elsevier B.V. All rights reserved.

Abbreviations: ADR, adverse drug reactions; AHMC, 3-[2-(diethylamino)-ethyl]-7-hydroxy-4-methylcoumarin; AMMC, 3-[2-(N,N-diethyl-N-methylammonium)ethyl]-7-


methoxy-4-methylcoumarin; BFBFC, 2,5-bis(trifluoromethyl)-7-benzyloxy-4-trifluoromethyl-coumarin; BFC, 7-benzyloxy-4-(trifluoromethyl)-coumarin; BnXPI, (E)-2-(2-(6-
p-methoxybenzy 2,3-dihydro-1H-xanthen-4-yl)vinyl)-3,3-dimethyl-1-propyl-3H-indol-1-ium iodide; HXPI, (E)-2-(2-(6-hydroxy-2, 3-dihydro-1H-xanthen-4-yl)vinyl)-3,
3-dimethyl-1-propyl-3H-indol-1-ium iodide; BOMCC, 7-benzyloxy-methyloxy 3-cyano-coumarin; BOMF, benzyloxy-methyl-fluorescein; BOMFC, 7-benzyloxy-methyloxy-
4-(trifluoromethyl)-coumarin; BOMR, benzyloxy-methyl-resorufin; BQ, 7-benzyloxy-quinoline; CEC, 3-cyano-7-ethoxycoumarin; CHPO, chloroethyl derivative (7-(2-chlor
oethoxy)–3H-phenoxazin-3-one; CYP, cytochrome P450; rCYPs, recombinant cytochrome P450 enzymes; DBF, dibenzyl-fluorescein; DBOMF, di-(benzyl-O-methyl)-
fluorescein; DDI, drug-drug interactions; EMA, European Medicines Agency; ER, 7-ethoxy-resorufin; EOMCC, 7-ethoxy-methyloxy-3-cyano-coumarin; EFC, 7-ethoxy-4-
(trifluoromethyl)-coumarin; FDA, Food and Drug Administration; HAMC, 7-hydroxy-4-(aminomethyl)-coumarin; HDI, herb-drug interactions; HN, 4-hydroxy-1,
8-naphthalimide; HTS, high-throughput screening; Km, the substrate concentration that leads to half maximal velocity; MAMC, 7-methoxy-4-(aminomethyl)-coumarin;
MFC, 7-methoxy-4-(trifluoromethyl) -coumarin; MOBFC, 7-p-methoxy-benzyloxy-4-trifluoro-coumarin; NBCeN, N-(4-butyl)-4-chloroethoxy-1,8-naphthalimide; NBHN,
N-(4-butyl)-4-hydroxy-1,8-naphthalimide; iPrBN, 4-isopropoxy-7H-benzo[de]benzo[4,5]imidazo[2,1-a]isoquinolin-7-one; HBN, 4-hydroxy-7H-benzo[de]benzo, [4,5]imi
dazo[2,1-a]isoquinolin-7-one; NCMN, N-(3-carboxy propyl)-4-methoxy-1, 8-naphthalimide; NCHN, N-(3-carboxy propyl)-4-hydroxy-1,8-naphthalimide; NEMN, N-((2-
hydroxyl ethoxy) ethyl)-4-methoxy-1,8-naphthalimide; NEHN, N-((2-hydroxyethoxy)ethyl)-4-hydroxy-1,8-naphthalimide; NEiPN, N-((2-hydroxyl ethoxy)ethyl)-4-isopro
poxy-1,8-naphthalimide; DPCI, 2-(3-(4-(2-chloroethoxy)styryl)-5,5-dimethylcyclohex-2-enylidene)malononitrile; DPOH, 2-(3-(4-hydroxystyryl)-5,5-dimethylcyclohex-2-e
nylidene) malononitrile; NEHN, N-(3-ethyl)-4-hydroxy-1,8-naphthalimide; NEN, N-(3-ethyl)-1,8-naphthalimide; NTI, narrow therapeutic index; OMF, 3-O-
methylfluorescein; PDB, Protein Data Bank; Vivid Cyan, 7-hydroxy-4-trifluoromethyl-coumarin; Vivid Blue, 3-cyano-7-hydroxy-coumarin; Vivid Red, resorufin; Vivid
Green, fluorescein; Vmax, the enzyme’s maximum velocity.
⇑ Corresponding authors.
E-mail addresses: wjj@dmu.edu.cn (J. Wu), yling@dicp.ac.cn (L. Yang), geguangbo@shutcm.edu.cn (G. Ge).

https://doi.org/10.1016/j.ccr.2020.213600
0010-8545/Ó 2020 Elsevier B.V. All rights reserved.
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Structural and catalytic features of human CYPs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.1. Structural features of human CYPs. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.1.1. General structural features of human CYPs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.1.2. Structural variability of human CYP enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.2. CYP-mediated reactions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3. In silico prediction of CYP mediated metabolism. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.1. Prediction of ligand binding to CYP enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.2. Prediction of the site of metabolism (SOM) by CYP enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
4. Approaches for the development of CYP probes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
4.1. Phenotype-based screening . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
4.2. Phenotype screening combined with computer-aided design . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
5. Non-optical probes for human CYPs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
6. Optical probes for human CYPs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
6.1. Fluorogenic probes for human CYPs. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
6.2. Bioluminogenic probes for human CYPs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
7. Biomedical applications of the probes for human CYPs. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
7.1. Sensing enzymatic activity in biological samples . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
7.2. Screening of CYP modulators . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
7.3. CYP–ligand interactions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
8. Challenges and future perspectives. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
Declaration of Competing Interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20

1. Introduction variety of fatty acids, such as lauric acid, myristic acid, arachidonic
acid and eicosadienoic acid [7]. Generally, the xenobiotic-
The cytochrome P450 (P450s or CYPs) superfamily of metabolizing CYPs are mainly related to the metabolism and the
heme-containing monooxygenase enzymes are responsible for treatment outcomes (such as efficacy and toxicity) of the drugs
the oxidative metabolism of a wide range of endogenous sub- or toxins, while endobiotic-metabolizing CYPs may be associated
stances and foreign chemicals in almost all living organisms. Mam- with the etiology of some diseases [2].
malian CYPs have a broad substrate specificity, and they play The expression and function of mammalian CYPs, which can be
crucial roles in the biotransformation of numerous physiologically, affected by genetic, physiological, pathophysiological and environ-
pharmacologically, or toxicologically important molecules, includ- mental factors, have been recognized as critical determinants of
ing steroids, chemical carcinogens, environmental pollutants, and inter-individual variability in drug disposition and clinical out-
therapeutic drugs [1]. As one of the largest enzyme superfamily, come [8–10]. Significant changes in both expression and function
CYPs have been extensively investigated by many researchers in of human CYPs may bring various undesirable effects, including
various fields, such as biochemistry, pharmacology, toxicology, therapeutic failure, idiosyncratic toxicity [11], multiple-organ
genetics, environmental biology, chemical biology and molecular injury and drug-drug interactions (DDI) [12] (Fig. 1). By far, inhibi-
biology. In humans, 57 genes that encode CYPs have been identi- tion of CYP activities is the most common mechanism leading to
fied (Table 1), which are classified into 18 families and 44 subfam- DDI [13]. The incidence of serious adverse drug reactions (ADRs)
ilies based on sequence similarity [2]. Of the identified 18 human was found to be extremely high among hospitalized patients, mak-
CYP families, three (i.e., CYP1, CYP2, and CYP3) are primarily ing ADR as one of the leading causes of death [14]. Since DDI is an
responsible for the metabolism of xenobiotics [3]. By contrast, important cause of ADRs, various regulatory agencies including the
the majority of the other CYP families are involved in the metabo- United States Food and Drug Administration (USFDA) and the
lism of endobiotic substances. For instances, it has been reported European Medicines Agency (EMA), have recommended to assay
that CYP2S1 is involved in polyunsaturated fatty acid x-1 hydrox- the DDI potential of an investigational drug or drug candidate
ylation [4], while CYP2W1 selective catalyzes oxidation of toward the key hepatic CYPs involved in drug metabolism (such
lysophospholipids [5]. Meanwhile, CYP4F22 has been reported to as CYP1A2, CYP2C8, CYP2C9, CYP2C19 and CYP3A4) before
play an essential role in the transformation of ceramide to acylce- approval [15]. Therefore, the identification or development of pre-
ramide [6], and CYP2Z1 catalyzes the oxidative metabolism of a ferred substrates for individual CYPs, to be used as ‘‘probe sub-
strates” to accurately assess the activities of a target CYP enzyme
Table 1 in vivo, ex vivo, or in other biological systems such as microsomal
Classification of human CYPs based on their substrate spectra. preparations, and evaluate the potentials of DDI, have received
Substrate class Human CYP enzymes great attention from both basic science researchers and the phar-
maceutical industry over the past two decades.
Xenobiotics 1A1, 1A2, 2A6, 2A13, 2B6, 2C8, 2C9, 2C18, 2C19, 2D6,
2E1, 2F1, 3A4, 3A5, 3A7, 3A43
Steroids 1B1, 7A1, 7B1, 8B1, 11A1, 11B1, 11B2, 17A1, 19A1, 21A2, 2. Structural and catalytic features of human CYPs
27A1, 39A1, 46A1, 51A1
Fatty Acids 2J2, 2S1, 2U1, 4A11, 4B1, 4F11, 4F12, 4V2, 4Z1
Lysophospholipids 2W1
2.1. Structural features of human CYPs
Eicosanoids 4F2, 4F3, 4F8, 5A1, 8A1
Vitamins 2R1, 24A1, 26A1, 26B1, 26C1, 27B1, 27C1 2.1.1. General structural features of human CYPs
Ceramides 4F22 The structural features of CYPs are invaluable for understanding
Unknown 2A7, 4A22, 4X1, 20A1
the structural basis of the selectivity of CYP ligands and gaining
2
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

Fig. 1. Interindividual variability in CYPs and its clinical implications.

metabolizing CYPs have been revealed and found in the Protein


Data Bank (PDB). The representative protein crystals for major
human CYPs are summarized in Table 2. The three-dimensional
structures of CYP enzymes provide crucial insight into the active
site architectures, the key domains and amino acids responsible
for ligand binding [19]. With the help of the structures of CYP
enzymes, further investigations on the molecular basis of the
structure–activity relationships, and the altered enzyme activity
arising from genetic polymorphism, can be conducted more effi-
ciently. More importantly, the crystal structures of CYPs strongly
facilitate the rational design of CYP ligands (such as specific inhibi-
tors or probe substrates) that may confer potential therapeutic
benefits or serve as diagnostic tools [46].

2.1.2. Structural variability of human CYP enzymes


The variability in the amino acid sequence and structural fea-
tures of the active site among various CYP enzymes is substantial,
Fig. 2. The general structure of human CYPs. The heme iron and the cysteine despite the overall conservation of three-dimensional structures in
residue located on L helix are colored in red and blue, respectively. all mammalian CYPs. The most structurally variable regions are
located in the B–C and F–G helices, and the L helix [22]. Some
examples of the structural diversity in these regions are high-
deeper insight into the structure-function relationships. One of the lighted below in the context of functional consequences. 1) CYP1A2
most conserved elements of CYPs is the cysteine residue located on favors small planar aromatic or heterocyclic amine ligands, which
a loop region in front of the L helix, which provides the sulfur atom can be attributed to several polar residues (Thr118, Ser122, and
to the heme iron; another one is the heme moiety surrounded by Thr124) on the B0 –C region that face toward the active site, as well
helices I and L (Fig. 2) [16,17]. The catalytic domain is connected as Thr223 on F helix and Asp320 on I helix that line the roof of the
to the NH2-terminal transmembrane helix of CYPs by a proline- active cavity [22]. 2) CYP2A6 preferentially binds small aromatic
rich region, and the substrate-binding area is located on the distal ligands [47], due to the fact that this enzyme has a correspondingly
side of the heme [17,18]. Overall, the secondary structures of mam- small active site pocket (260 Å3), while Asn297 is considered as a
malian CYPs are conserved, and all human CYPs contain 12 a- key residue for substrate orientation [23]. 3) CYP2C9 favors weakly
helices (A-L) and 4 b-sheets (1–4) [19]. The B0 helix and regions acidic ligands [27], owing to that its structure (PDB 1R9O) shows a
between the F and G helices show the greatest degree of variation disordered conformation of the F–G loop and an extra turn at the
and undergo remarkable conformational changes to allow the NH2-terminus of helix A, which is quite different from that of other
ligands to enter [16]. Notably, several additional helices are pre- members from CYP2C subfamily [28]. The active site cavity of
sent, such as the F’ and G’ helices found in many human CYPs. CYP2C9 is smaller than that of CYP2C8 but larger than that of
The F and G helices and the F–G loop, together with the F’ and G’ CYP2A6 [48]. 4) CYP2E1 possesses one of the smallest active-site
helices play a key role in substrate access to the active site, by pockets (190 Å3), and this enzyme favors neutral compounds with
forming a ‘‘roof” for the active site opposite to the heme ‘‘floor” low molecular weight and fatty acids [31]. CYP2E1 has a conserved
[1,20]. The conserved I helix, the longest helix in human CYPs, is Asp295 in the active site cavity, and this amino acid plays a pivotal
adjacent to heme and runs through the catalytic domain [17]. role in substrate recognition and ligand binding. 5) CYP3A4 is a
Currently, the crystal structures of more than one hundred highly flexible enzyme that possesses a large binding cavity
human drug-metabolizing CYPs and 43 human steroid- (1173 Å3 ~ 1332 Å3), leading to the binding of a wide range of
3
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

Table 2
Representative crystal structures of the major human CYP enzymes.

Enzyme PDB ID Resolution (Å) Ligand Refs.


CYP1A1 a
4I8V 2.60 Å a-naphthoflavone [21]
CYP1A2 a
2HI4 1.95 Å a-naphthoflavone [22]
a
CYP2A6 1Z10 1.90 Å Coumarin [23]
1Z11 2.05 Å Methoxsalen [23]
a
CYP2A13 4EJG 2.50 Å Nicotine [24]
CYP2B6 a 4I91 2.00 Å (+)-a-Pinene [25]
CYP2C8 a 2VN0 2.70 Å Troglitazone [26]
2NNJ 2.28 Å Felodipine [26]
2NNI 2.80 Å Montelukast [26]
a
CYP2C9 1OG5 2.55 Å S-wafarin [27]
1R9O 2.00 Å Flurbiprofen [28]
CYP2C19 a 4GQS 2.87 Å 0XV b [29]
CYP2D6 a 4WNU 2.26 Å Quinidine [30]
CYP2E1 a 3E6I 2.20 Å Indazole [31]
CYP3A4 a 5TE8 2.70 Å Midazolam [32]
3TJS 2.25 Å DTMCR c [33]
3NXU 2.00 Å Ritonavir [34]
CYP3A5 a 5VEU 2.91 Å Ritonavir [35]
CYP1B1 a 3PM0 2.70 Å a-naphthoflavone [36]
CYP7A1 3V8D 1.90 Å 7-ketocholesterol [37]
3SN5 2.75 Å Cholest-4-en-3-one [37]
CYP11A1 3N9Y 2.60 Å Cholesterol [38]
a
CYP17A1 3RUK 2.60 Å Abiraterone [39]
CYP19A1 3EQM 2.90 Å Androstenedione [40]
CYP21A2 4Y8W 2.64 Å Progesterone [41]
a
CYP46A1 2Q9F 1.90 Å Cholesterol-3-sulphate [42]
4ENH 2.50 Å Fluvoxamine [43]
3MDV 2.40 Å Clotrimazole [44]
CYP51A1 3LD6 2.80 Å Ketoconazole [45]
a
The recombinant CYPs are commercially available.
b
An inhibitor of CYP2C19.
c
Desthiazolylmethyloxycarbonyl ritonavir.

structurally diverse ligands [49]. CYP3A4 has several distinguish- via NADPH-CYP reductase, and generally involves a distal water
ing structural features, such as short F and G helices and several displacement from the heme iron [55]. Once the molecular oxygen
large hydrophobic phenylalanine residues that form a cluster in is bound to the heme, an oxy-iron-CYP complex is formed after the
the ligand-free structure at the roof of the active site. The pheny- transfer of a second electron and oxygen activation. The oxy-iron-
lalanine cluster is distorted when the enzyme binds to ligands CYP complex is reduced, generating a peroxo-ferric-CYP complex,
[33,50,51]. 6) the structures of CYP3A5 and CYP3A4 are very simi- which is further converted to a hydroperoxo-iron-P450 complex
lar due to their high homology (~83% identity) in amino acid via protonation of the terminal oxygen. Then, the latter complex
sequence, but the architectures of their active cavities are different. undergoes proton-assisted oxygen–oxygen bond scission, forming
Especially as the Phe210 located on the extended portion of helix F a reactive, and high-valent oxo-iron-CYP complex. Finally, sub-
in CYP3A5 is longer than Leu210 of CYP3A4, which expands the strate oxidation by this reactive complex produces the oxidized
size of the upper region of the substrate binding cavity in CYP3A5 metabolite, with CYP regenerated to its initial ferric state [56,57].
by increasing the space between the helix F-F’ and helix G’-G con- The schematic catalytic cycle of CYP enzymes is shown in Fig. 4.
nectors, and this space can be further enlarged by replacing Phe108 CYP-catalyzed reactions are very diverse, including hydrocar-
in CYP3A4 using a shorter Leu108 in CYP3A5 [35]. bon hydroxylation; epoxidation; O-, S-, and N-dealkylation; dehy-
A visual comparison of the substrate binding cavities of the drogenation; dehalogenation, oxidative deamination,
above mentioned CYPs is shown in Fig. 3. It is apparent that the decarboxylation; reductive dehalogenation; N-oxide, and epoxide
structural variability, primarily in the regions of F–G helices and reduction; isomerization and ring formation. Among all known
B0 helix among various CYPs, contributes to the distinct shapes CYP-catalyzed reactions, monooxygenation is the most common
and sizes of the active cavities. Recent advances in deciphering reaction [55,58]. A clear understanding of the enzymology of vari-
structural and mechanistic properties of human CYPs have signifi- ous types of CYP-catalyzed reactions is important for the rational
cantly facilitated the development of highly specific ligands for design and development of ligands for a CYP enzyme.
various CYP enzymes.

2.2. CYP-mediated reactions 3. In silico prediction of CYP mediated metabolism

As their name implies, the P450 hemoproteins have a character- 3.1. Prediction of ligand binding to CYP enzymes
istic absorption band at 450 nm when the ferrous heme iron binds
to carbon monoxide [52]. During the catalytic process, CYPs bind The metabolism occurs when the compound has affinity for CYP
the molecular oxygen, split it into its component atoms and trans- subtypes and is able to react with the activated heme group. The
fer one oxygen atom to a substrate in the CYP active site [53]. The existing approaches for predicting the specificity of CYP isoforms
binding of ligands to CYPs occur mainly when the heme iron is in can be divided into two major categories: ligand-based methods
the ferric state, with water bound [54]. At most cases, the catalytic and structure-based methods [59]. The ligand-based approach is
cycle of CYPs is initiated by binding of a substrate to the ferric to establish mathematical models based on the biological activities
enzyme, with the first electron provided to the CYP from NADPH and descriptors of ligands, such as physicochemical and structural
4
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

Fig. 3. Comparisons of the substrate binding cavity of six major human hepatic CYPs: (a) CYP1A2 (2HI4)- a-naphthoflavone, (b) CYP2A6 (1Z10)-coumarin, (c) CYP2C9 (1OG5)-
flurbiprofen, (d) CYP2E1 (3E6I)- indazole, (e) CYP3A4 (3NXU)-ritonavir, (f) CYP3A5 (5VEU)-ritonavir. The greatest degree of variation includes the region between the F and G
helices (green) and the B0 helix (pink).

Fig. 4. The schematic catalytic cycle of CYPs.

5
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

properties. Machine learning methods including neural networks, based screening systems and very limited access to purified human
support vector machines and random forests have been developed CYP enzymes. During the 1980 s, in addition to the phenotype-
as primary methods for predicting enzyme specificity on account based assays using purified human CYPs, other approaches, includ-
of the complex nonlinear relationships in enzyme-ligands interac- ing correlation of enzyme activities toward individual substrates
tion data [60]. More detailed ligand-based methods and tools have and the application of selective chemical inhibitors, were devel-
recently been reviewed [61]. In structure-based methods, protein oped and used in drug metabolism related fields [16]. After that,
structure information is essential for understanding protein- application of recombinant DNA technology yielded a panel of
ligand interaction details. Structural characteristics of main CYP cDNAs for CYPs, and then heterologous expression of mammalian
subtypes have been described above. Both the key information of CYPs was achieved in mammalian and insect cells, yeast and bac-
substrate recognition and catalysis in target enzyme (ligand- terial systems, in the early 1990 s [16,71].
based methods) as well as protein structure information The currently used specific probe substrates for human CYPs are
(structure-based methods) would contribute to the computer- mainly derived from endogenous substance or therapeutic drugs.
aided design and development of the molecular probes for a target For example, testosterone and cortisol can be selectively catalyzed
CYP. Molecular docking and molecular dynamics (MD) simulations, by CYP3A to form 6b-hydroxylated products [72,73]. Many drugs,
as well as quantum mechanics/molecular mechanics (QM/MM) such as ethoxyresorufin (CYP1A), paclitaxel (CYP2C8), and nifedip-
methods are the most commonly used structure-based methods ine (CYP3A), have been found with high specificity towards a par-
to predict binding affinity and binding mode [62]. Based on the ticular CYP, which qualifies them as CYP probe substrates [74].
predicted binding affinity by scoring functions or more accurate Over the past two decades, equipped with a deeper understanding
free energy values in MD simulations, binders and non-binders of the structure and catalytic behaviors of various CYPs, research-
can be classified. Notably, multiple binding poses are observed in ers have determined the relationships between substrate structure
CYP2C9 and 2D6, which needs to be taken into account to interpret and CYP selectivity, which are very helpful for discovery or rational
the experimental data [63,64]. For CYP3A4, binding cooperativity design of the probe substrate(s) for a target CYP enzyme.
effect of more than one ligand is proposed [65]. Especially, due In recent years, several groups have systematically investigated
to the active sites of CYP are deeply buried inside the enzyme, the structure-metabolism relationships for a series of natural com-
the flexibility and adaptability of accessible channels have been pounds, one of which are the dibenzocyclooctadiene lignans iso-
simulated and proved to be crucial for ligand passage and selectiv- lated from Fructus Schisandrae. These lignans exhibited obvious
ity [66]. metabolic preferences for CYP3A. For example, the dibenzocyclooc-
tadiene lignans with six methoxy groups on the biphenyl rings are
co-substrates of both CYP3A4 and CYP3A5, such as deoxyschizan-
3.2. Prediction of the site of metabolism (SOM) by CYP enzymes
drin [75]. By contrast, the C-7 hydroxylated dibenzocyclooctadiene
lignans are selectively metabolized by CYP3A4, but not CYP3A5,
Because of the variety of CYP mediated reactions, it may exist a
such as gomisin A and schizandrin [76,77]. Conversely, schisan-
few potential reactive sites of a substrate . Identification of SOMs is
therin E, a natural dibenzocyclooctadiene lignan containing a ben-
critical for design of compounds with good ADMET properties. So
zoyl group at C-6 and a hydroxyl group at C-12, can be selectively
far, hybrid QM/MM calculation is considered as the most accurate
metabolized to a 2-O-demethylated product by CYP3A5 [78]. The
method which describes the active sites by accurate QM method,
above results suggest that the substituents on the octatomic and
while capturing the effects of protein/solvent environments by
biphenyl rings are crucial to the metabolic selectivity for CYP
MM method [54,67]. It is notable that the complete catalytic cycle
enzymes. The structure-CYP metabolism selectivity relationships
of CYP can now be studied by QM/MM methods. However, these
of dibenzocyclooctadiene lignans are summarized in Fig. 5.
calculations are so time-consuming that it is not yet possible to
Another notable case is the naturally occurring bufadienolides,
predict a large number of ligands [54,67]. Therefore, reactivity pre-
the major constitutes in traditional Chinese medicine Chansu.
dictions of small ligands are always based on simplified models of
These compounds also showed evident metabolic preferences to
the heme interactions which ignore the protein environment
CYP3A enzymes. Several groups have reported that some bufa-
[59,61]. Studies have shown that for most CYP isoenzymes, such
dienolides bearing an acetyl substituent at the C-16 site (such as
as CYP3A4, 1A2, 2A6, 2B6, and 2E1, the prediction sites with low
cinobufagin and bufotalin), can be metabolized by both CYP3A4
activation energies are the major determinants of SOMs [50,68].
and CYP3A5 [79]. By contrast, resibufogenin and bufalin, two nat-
While in the case of CYP2D6 and 2C9, it is insufficient to predict
ural bufadienolides without an acetyl substituent at the C-16 site,
SOMs by assaying reactivity alone rather than combining with
could be selectively metabolized by CYP3A4 to form the corre-
binding poses [69,70]. The common SOM prediction tools have
sponding 5b-hydroxylated product [80]. Further investigations
been summarized by some previous literatures [61,62]. In addition
revealed that the hydroxyl group at the C-3 site and the epoxy
to the ligand reactivity prediction, the accessibility of ligands to the
group at the C-14/15 site of these bufadienolides are crucial for
vicinity of active site can be simulated by docking and MD
CYP3A4-catalyzed oxidation, while the acetyl substituent at the
approaches. A simple metabolic criterion is based on the distance
C-16 site may contribute to metabolic increase of CYP3A5. The
(less than 6 Å) between the SOM of a substrate and the heme iron
structure-metabolic selectivity relationships of bufadienolide com-
ion, and more elaborate criteria including orientation are also pro-
pounds are summarized in Fig. 6.
posed for improvement [67].

4.2. Phenotype screening combined with computer-aided design


4. Approaches for the development of CYP probes
In view of the fact that identification of probe substrates for a
4.1. Phenotype-based screening target CYP isoenzyme from endogenous substances or approved
drugs is always time-consuming, the researchers have begun to
The discovery and development of the first-generation CYP rationally design CYP probes with the help of the structural and
probes was made primarily through phenotype-based screening. catalytic features of target enzyme(s). Computer-aided design of
Before the 1980 s, it was difficult to determine whether a com- specific probes for a target enzyme is mainly based on the 3D
pound could act as a highly specific substrate (or ‘‘probe”) for a tar- structure of the target enzyme and the key information of sub-
get human CYP, owing to the incomplete nature of the phenotype- strate recognition and catalysis in target enzyme(s). Over the past
6
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

Fig. 5. The substrate structure-CYP selectivity relationships of dibenzocyclooctadiene lignans.

Fig. 6. The substrate structure-CYP selectivity relationships of bufadienolides.

decade, in order to design a specific substrate for a particular designed on the basis of structural features (especially the amino
human CYP enzyme in a more efficient way, molecular docking acids in the active site cavity) of CYP1A that play key roles in
and molecular dynamic simulations are often utilized to explore ligand-enzyme interactions, while the substrate preferences of
the key interactions between CYPs and their ligands CYP1A are also considered. In these studies, 4-hydroxy-1,8-
[46,78,81,82]. Commonly, the key interactions contributing to sub- naphthalimide (HN) was selected as the basic fluorophore due to
strate recognition and catalysis can guide the structural modifica- its excellent optical properties, including high fluorescence quan-
tion of the ligands (such as substrate) to make their interactions tum yield, large stokes shifts (>120 nm), good photostability and
with target enzymes more specific and stronger [78,81]. With the significant two-photon absorbability characteristics [84-86]. On
help of computer-aided design and virtual screening technology, the baisi of the subtle differences in substrate preference and 3D
the biochemists can rationally design novel, man-made probe sub- structure of CYP1A1 and CYP1A2, an isoform-specific probe
strates with improved specificity for target CYPs, as well as develop (NBCeN) for CYP1A1 was successfully developed by Dai et al, via
enzyme-specific probe substrates via modification of a common chemical modifications of the local structure at the metabolic site
substrate for multiple CYPs [82,83]. on a known co-substrate for CYP1A isoenzymes [83]. To obtain a
In 2015, Dai et al. have developed several two-photon ratiomet- highly sensitive and specific probe for the target enzyme, a panel
ric fluorescent probes (i.e., NCMN and NBCeN) for CYP1A (including of O-alkylated HN derivatives were synthesized and used to
CYP1A1 and CYP1A2) and CYP1A1 by virtual screening combined explore the structure-selectivity relationship with the help of
with CYP phenotyping assays [81,83]. NCMN and NBCeN display molecular docking and phenotyping-based screening (Fig. 7). The
the best combination of selectivity, sensitivity, ideal kinetic behav- results demonstrate that the introduction of a chloroethyl group
iors and ratiometric fluorescence response following CYP1A or to HN can yield the best specificity towards CYP1A1 over other
CYP1A1-catalyzed O-dealkylation [81,83]. These probes are CYPs including CYP1A2 [83]. This newly developed CYP1A1 probe

7
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

Fig. 7. A practical strategy proposed for the design and development of a specific fluorogenic probe for a given CYP isozyme.

has been successfully used to real-time monitor the activities of (turnover) (Vmax or kcat), as well as commercial availability and
CYP1A1 in living systems with very high sensitivity and specificity. good chemical stability for both substrate and metabolite(s)
These findings suggest that fluorescent probes hold great promise [93,94]. It is well-known that enzyme kinetic behavior is a key con-
for sensing the biological function of target CYPs in living systems; cern for the quantitative applications of probe substrates, thus the
their application will be very helpful for exploring the relevance of probe substrates that follow classic Michaelis-Menten kinetics are
CYPs to human diseases. Furthermore, the strategy used for the highly desirable. The preferred and acceptable probe substrates for
development of NBCeN blazes a new trail for the development of the major hepatic human CYPs have been well documented and
enzyme-specific probes for other CYPs. recommended by FDA [95]. All these probes and their metabolites
Following the development of NBCeN as an isoform-specific flu- are detected by LC combined with ultraviolet or tandem mass
orescent probe substrate for CYP1A1, Zhang et al. further investi- spectrometry. However, most of these non-fluorogenic CYP probes
gated a series of 1,8-naphthalimide-based two-photon are derived from drugs, which commonly display low detection
fluorescent chromophores and evaluated their potentials as sensitivity and inapplicable for high throughput screening of CYP
CYP1A1 substrate using computer-aided calculations [87]. The inhibitors [74,92]. Despite the aforementioned issues, non-
docking results showed that NCMN-3, containing an electron- fluorogenic probes are still used by pharmaceutical researchers,
withdrawing benzothiadiazole group, and NCMN-5, with an in consideration of their ‘‘drug-like” properties. The commonly
electron-donating carbazole group, fit the active site cavity of used non-optical substrates for human CYPs, along with the LC-
CYP1A1 well, with the carbon-iron distances of 3.31 Å and tandem mass spectrometry methods used for their analysis, are
4.18 Å, respectively, suggesting that NCMN-3 and NCMN-5 might listed in Table 3.
be specific substrates for CYP1A [87]. Nevertheless, it should be Besides of the CYPs in the CYP1-3 families, steroid-metabolizing
noted that all virtual screening approaches require further experi- CYPs have also received increased attentions, as they are closely
mental validation by reaction phenotyping assays and enzymatic related to certain pathological conditions. Multiple human CYP
kinetic analyses [88]. Furthermore, to ensure the isoform speci- enzymes are known to contribute to the production of steroid hor-
ficity, results from the biochemical assays for target enzyme activ- mones [117–120]. Among these enzymes, CYP17A1 is the sole
ity using fluorogenic probes should be verified with other methods, enzyme responsible for converting progestogens to androgens in
such as LC-MS/MS-based proteomics or western-blotting, or with humans, and the latter drives prostate cancer proliferation. Thus,
assay using known non-fluorogenic specific probe substrates for CYP17A1 is recognized as an attractive therapeutic target for
the target enzyme [81,89,90]. blocking androgen biosynthesis [121], implying that inhibition of
More recently, Ning et al developed a near-infrared fluorescent this enzyme can be therapeutically useful [122]. One CYP17A1
probe for CYP2J2 (BnXPI) by virtual screening combined with CYP inhibitor, abiraterone, progresses rapidly through clinical trials,
phenotyping assays. Based on the active cavity structure character- and has received FDA approval in 2012 for administration [123].
istics, Ning et al. designed a series of self-immolative linker into CYP46A1, which is a cholesterol 24-hydroxylase, is specifically
the HXPI derivatives with a long aromatic skeleton, in order to expressed in the brain. The metabolite 24S-hydroxycholesterol
adapt to the narrow substrate channel of CYP2J2. These modifica- shows elevated levels in the cerebrospinal fluid of Alzheimer
tions aim to shorten the spatial distance between the O-alkyl group patients and could induce cell death, suggesting that CYP46A1 is
(metabolic moiety) and the catalytic center of CYP2J2. Further- involved in the pathology of Alzheimer’s disease [124]. Therefore,
more, based on the CYP phenotyping assays, BnXPI possesses a CYP46A1 inhibition therapy may be therapeutically beneficial for
benzyl as a self-immolative linker, which was found to exhibit the patients with Alzheimer’s disease [125]. Overall, either for
the optimal combination of flexibility and specificity towards the human drug-metabolizing CYPs or for the steroid-
CYP2J2 amongst all tested O-alkylated HXPI derivatives [91]. metabolizing CYPs, the highly specific probe substrates are crucial
for activity sensing and screening of selective modulators of the
target CYPs.
5. Non-optical probes for human CYPs

Probe substrates are defined as the compounds that are exclu- 6. Optical probes for human CYPs
sively metabolized in vitro by an individual CYP enzyme in a given
biological system [92]. Generally, an excellent probe targeting a As mentioned above, quantification of the metabolite of non-
single enzyme is recommended to meet the following require- optical CYP substrates requires liquid chromatography combined
ments: high selectivity, good sensitivity, and high conversion rate with UV or mass spectrometry detector, such assays are
8
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

Table 3
Commonly used non-optical CYP probes that are analyzed using LC/MS-based methods.
a
Enzyme Tissue Probe substrates Chemical structure Catalytic Applications Km Vmax MRM Refs.
sites monitor or Kcat or kmax
CYP1A2 Liver Phenacetin O-deethylation In vitro (rCYPs, 47.0 688b 151.9/ [96]
microsomes, living 110.0
cells)

CYP2A6 Liver, Coumarin 7-hydroxylation In vitro (rCYPs, 0.84 940b 161.0/ [96]
Lung microsomes, living 133.0 g
cells)

CYP2A13 Lung Phenacetin O-deethylation In vitro (rCYPs, 10.7 3.82f 151.9/ [97]
microsomes, living 110.0
cells)

CYP2B6 Liver, Bupropion Hydroxylation In vitro (rCYPs, 81.7 413b 256.2/ [96]
Lung microsomes, living 139.0
cells)

CYP2C8 Liver Paclitaxel 6a- In vitro (rCYPs, 18.3 250b 870.6/ [98]
hydroxylation microsomes, living 286.1
cells)In vivo (rat)

Amodiaquine N-dealkylation In vitro (rCYPs, 1.89 1480b 328.0/ [96]


microsomes, living 283.0
cells)

CYP2C9 Liver, Diclofenac 4-hydroxylation In vitro (rCYPs, 27.6 967b 312.0/ [99,100]
Intestine microsomes, living 231.1
cells)In vivo (rat)

d
(S)-Warfarin 7-hydroxylation In vitro (rCYPs, 4.60 2.33 323.0/ [101]
microsomes, living 177.0 g
cells)In vivo (human
and rat)

Tolbutamide 4-hydroxylation In vitro (rCYPs, 147 276b 287.0/ [96]


microsomes, living 171.0
cells)In vivo (human
and rat)
d
CYP2C19 Liver (R)-Omeprazole 5- hydroxylation In vitro (rCYPs, 3.99 12.9 362.2/ [102]
microsomes, living 214.1
cells)In vivo (human
and rat)

(S)-Mephenytoin 40 -hydroxylation In vitro (rCYPs, 57.2 58.3b 235.0/ [96,100]


microsomes, living 150.1
cells)

CYP2D6 Liver Bufuralol 10 -hydroxylation In vitro (rCYPs, 7.28 118.9b 278.1/ [100,103]
microsomes, living 186.1
cells)In vivo (human
and rat)

Dextromethorphan O-demethylation In vitro (rCYPs, 4.64 202b 258.2/ [96,100]


microsomes, living 157.0
cells)In vivo (human
and rat)

CYP2E1 Liver, Chlorzoxazone 6-hydroxylation In vitro (rCYPs, 73.9 2360b 184.2/ [96]
Lung microsomes, living 120.0 g
cells)In vivo (human
and rat)

(continued on next page)

9
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

Table 3 (continued)
a
Enzyme Tissue Probe substrates Chemical structure Catalytic Applications Km Vmax MRM Refs.
sites monitor or Kcat or kmax
CYP2J2 Heart, Astemizole O-demethylation In vitro (rCYPs, 1.80 152c 445.0/ [104]
Liver microsomes, living 204.0
cells)In vivo (human
and rat)

d
Terfenadine Hydroxylation In vitro (rCYPs, 1.60 29.4 488.3/ [105]
microsomes, living 452.2
cells)In vivo (human
and rat)

CYP3A4 Liver, Midazolam 10 -hydroxylation In vitro (rCYPs, 2.27 1220b 342.1/ [96]
Intestine microsomes, living 203.1
cells)In vivo (human
and rat)

Testosterone 6b- In vitro (rCYPs, 46.4 5260b 305.0/ [96]


hydroxylation microsomes, living 269.0
cells)

Nifedipine Dehydrogenation In vitro (rCYPs, 15.3 4461b 345.0/ [75,106]


microsomes, living 122.0
cells)

Bufalin 5b- In vitro (rCYPs, 8.00 2620b 403.3/ [94]


hydroxylation microsomes, living 349.3
cells)

Deoxyschizandrin 7-hydroxylation In vitro (rCYPs, 1.60 623b 250 nm [75]


microsomes, living
cells)

Gomisin A 8-hydroxylation In vitro (rCYPs, 6.30 47.5b 245 nm [76]


microsomes, living
cells)

d
CYP3A5 Liver, Schisantherin E 2-O- In vitro (rCYPs, 5.90 63.4 230 nm [78]
Lung, demethylation microsomes, living
Kidney cells)

10
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

Table 3 (continued)
a
Enzyme Tissue Probe substrates Chemical structure Catalytic Applications Km Vmax MRM Refs.
sites monitor or Kcat or kmax
d
T-5 N-oxidation In vitro (rCYPs, 2.60 13.9 584.2/ [107]
microsomes, living 476.2
cells)

d
Maraviroc Hydroxylation In vitro (rCYPs, 48.9 0.93 530.0/ [108]
microsomes, living 296.0
cells)

CYP4F2 Liver Leukotriene x-hydroxylation In vitro (rCYPs, 60.0 2.70 d


335.0/ [109,110]
microsomes, living 194.9 g
cells)

CYP7A1 Liver Cholesterol 7a- In vitro (rCYPs, 16.4 397b 385.1/ [111,112]
hydroxylation microsomes, living 159.1
cells)

CYP17A1 Adrenal Progesterone 17a- In vitro (rCYPs, 11.4 1.31f 331.2/ [39,113]
hydroxylation microsomes, living 97.00
cells)

e
CYP21A2 Adrenal Progesterone 21- In vitro (rCYPs, 1.05 3.73 331.2/ [114]
hydroxylation microsomes, living 97.00
cells)

CYP46A1 Brain 24(S)- 24,25- and In vitro (rCYPs, 3.90 0.92f 210 nm [42,115]
hydroxycholesterol 24,27- microsomes, living
dihydroxylation cells)

Cholesterol 24(S)- In vitro (rCYPs, 5.40 0.11f 420.3/ [42,116]


hydroxylation microsomes, living 385.5
cells)

a
Km values were lM; b Vmax values were in pmol/min/mg human liver microsome; c Vmax values were in pmol/min/mg human small intestinal microsome; d Vmax or kcat
values were in nmol/min/nmol P450 for CYP enzymes; e Vmax values were in nmol/min/mg transfected cells; f kcat values were in min1; g: negative mode; MRM: multiple
reaction monitoring; kmax: wavelength of maximum UV absorption. Red arrows indicate the major site of metabolism by target CYP(s).

11
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

time-consuming in both sample preparation and analysis, and of NBCeN, Ning et al have reported a new two-photon fluorescent
require skilled operators and expensive equipment. By contrast, probe for CYP1A1 (termed iPrBN) using 4-hydroxy-7H-benzo[de]
the fluorogenic or luminescent substrates based optical assays benzo[4,5]imidazo[2,1-a] isoquinolin-7-one (HBN) as a basic skele-
are time- and- cost-saving, easy to made and can be adapted for ton. This fluorogenic probe is designed on the basis of the substrate
96- and 384-well microplate formats, representing a highly effi- preferences of CYP1A1 and the principle of intramolecular charge
cient approach for high-throughput screening of inhibitors or stim- transfer (ICT). Further investigations demonstrate iPrBN displays
ulators of a target enzyme. Hence, the optical probe substrates for excellent specificity and ultrahigh sensitivity towards CYP1A1,
human CYPs are more and more favored by the researchers. with the detection limit as low as 0.036 nM [82]. More recently,
Jin et al have designed and developed a resorufin-based isoform-
6.1. Fluorogenic probes for human CYPs specific fluorescent probe (CHPO) for highly selective and sensitive
sensing CYP1A1 activities in complex biological systems. CHPO is a
Compared to non-optical probes, fluorogenic probes have many cell membrane permeable agent, which has been successfully used
inherent advantages [81,117], including highly sensitive (the for imaging of endogenous CYP1A1 in living cells and tissue slice
detection limit of fluorogenic probes can be up to picomole level), [130]. The reported fluorogenic probes for CYP1A1 have provided
easy to manage, time- and- cost-saving (capable for high- promising tools for high-throughput screening of CYP1A1 inhibi-
throughput detection) and can be adapted for 96- and 384-well tors and sensing CYP1A1 activities in living systems, which
microplate formats, as well as in-situ imaging with high resolution strongly facilitate the investigations on the relevance of CYP1A1
(commonly up to sub-nanometer spatial resolution and sub- to human diseases and the regulatory effects of CYP1A1 by xenobi-
millisecond time resolution). These advantages allow most fluoro- otics. Besides of the development of a variety of CYP1A1 fluoro-
genic probes be used to real-time sensing CYP activities in living genic probes, another major breakthrough in this field is the
cells or in vivo. Generally, CYP optical probes or sensors can be development of an isoform-specific fluorogenic probe for CYP3A4
divided into several categories, including substrate-based sensors (termed NEN), which has been reported recently by Ning et al.
and electrochemical biosensors [118,119]. Among these, [137]. NEN was developed by a novel two-dimensional strategy.
substrate-based sensors are widely used for screening of CYP mod- In the first dimension, docking based virtual screening was used
ulators, and in other drug metabolism and pharmacokinetics to select a suitable two-proton fluorophore as the substrate
(DMPK) related studies [119]. Substrate-based activity sensors candidate for CYP3A4. In the second dimensional design, chemical
can readily record the alterations in CYP activities in the presence modifications on the selected fluorophore (N-substituted
of drug candidate(s), which strongly facilitate the development of 1,8-naphthalimide) were performed to optimize the catalytic
the new chemical entities, and decrease the rates of failure later activity and isoform-selectivity. Notably, although great break-
in the drug development process [120]. through has been made by researchers in this field, the highly
The reported fluorogenic probes for human CYPs have been specific fluorogenic probes for target human CYP isoforms are
summarized in Table 4. It is should be noted that most of reported rarely reported. Particularly, the highly specific fluorogenic probes
fluorogenic probes for human CYPs displayed poor selectivity for target endobiotic-metabolizing CYPs have not been reported
[129–138], except for NBCeN (CYP1A1), iPrBN (CYP1A1), coumarin yet. Hence, much more efforts should be made to develop more
(CYP2A6), BQ (CYP3A4), NEN (CYP3A4) and BFBFC (CYP3A4). The practical and highly specific fluorogenic probes for target human
non-specific fluorogenic probes for CYPs can only be used for CYPs.
screening CYP inhibitors in vitro using individual recombinant
CYP enzyme as enzyme source. For example, 3-O- 6.2. Bioluminogenic probes for human CYPs
methylfluorescein (OMF), a so-called fluorogenic probes for
CYP2C19, could also be O-demethylated by CYP2C9 and CYP1A1 Unlike fluorescent based assays, bioluminescent based CYP
[126]. Another example is 7-benzyloxy-4-(trifluoromethyl)-cou activity assays always involves two reaction steps. First, the
marin (BFC), which can be metabolized by both CYP1A2 and luminogenic precusor produces D-luciferin, the natural substrate
CYP3A4. Kinetic analyses demonstrated a lower Km with CYP1A2, for firefly luciferase. Second, the luciferase is added and catalyzes
but a higher Vmax with CYP3A4 [127]. By contrast, 2,5-bis(trifluoro luciferin to yield oxyluciferin that bring photons in the presence
methyl)-7-benzyloxy-4-trifluoromethyl-coumarin (BFBFC), which of ATP, magnesium ion and oxygen (Fig. 8). Bioluminescence
has two additional trifluoromethyl groups compared to BFC, was occurs during the oxidation process, which can be detected as light
selectively metabolized by CYP3A4 [128]. Notably, in most cases, output in a luminometer [142]. The bioluminescent based assays
the isoform-specificity of reported fluorogenic substrates for possess the following merits [142]: 1) obviates the need for pro-
CYP3A4 over CYP3A5 has not been well-studied, except for duct extractions prior to detection by the method of chromatogra-
dibenzyl-fluorescein (DBF). phy or mass spectrometry, 2) eliminates safety concerns associated
A milestone fluorogenic probe to note is NBCeN, which is the with radiometric assays, 3) obviates the interferences associated
first reported isoform-specific fluorogenic probe for CYP1A1. From with fluorescent analytes in fluorescent assays. Notably, biolumi-
the views of 3D structures of two CYP1A isoforms in humans nescent based assays also have some demerits. The major demerit
(CYP1A1 and CYP1A2), the volume of the catalytic cavity of CYP1A1 of bioluminescent assay is the limited tissue penetration ability of
is larger than that of CYP1A2 (524 Å3 VS 375 Å3). Meanwhile, pre- emitted light resulting in quenching of the bioluminescence by tis-
vious studies have found that CYP1A2 preferred to catalyze O- sue components [143,144]. In recent years, many efforts has been
demethylation of a set of substrates with O-methyl group(s), while made to synthesize red-shifted D-luciferin analogues to improve
CYP1A1 preferred to catalyze O-dealkylation of relatively large the tissue penetration [145]. Furthermore, oxyluciferin, the
ethers, such as O-ethyl or O-chloroethyl [3,48]. These observations luminescence product, could also regenerate into Luciferin. The
inspired the researchers to develop a panel of isoform-specific flu- luciferin recycling process also follows two-step reaction: 1) trans-
orogenic probes for CYP1A1 via adjusting the volume of the leaving formation of oxyluciferin to 2-cyano-6-hydroxybenzothiazole, and
ether group(s), such as CHPO and iPrBN [82,130]. Furthermore, the 2) condensation of 2-cyano-6-hydroxybenzothiazole with
strategies used in the design and development of NBCeN have D -cysteine to yield luciferin [146]. In addition, it has been reported
greatly promoted the development of fluorescent probes for other that some endogenous compounds can strongly inhibit the lucifer-
target enzymes, such as human carboxylesterases and human ase, which in turn, affect the readouts of the bioluminescent based
UDP-glucuronosyltransferases [84]. Inspired by the development assays [147,148].
12
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

Table 4
Reported fluorogenic probes for human CYPs.

Enzyme Substrate Substrate structure Metabolite Applications Km Vmax or kex kem Ref.
name (lM) Kcat (nm) (nm)
a
CYP1A1 NBCeN NBHN In vitro (rCYPs, microsomes, living 0.84 18.99 450 562 [83]
cells)

a
iPrBN HBN In vitro (rCYPs, microsomes, living 0.29 5.54 470 525 [82]
cells)

a
NCMN NCHN In vitro (rCYPs, microsomes, living 11.99 17.44 450 564 [81]
cells)

NEMN NEHN In vitro (rCYPs, microsomes, living N.A. N.A. 458 552 [129]
cells)

NEiPN NEHN In vitro (rCYPs, microsomes, living N.A. N.A. 446 550 [130]
cells)

DPCI DPOH In vitro (rCYPs, microsomes, living N.A. N.A. 555 673 [131]
cells)

a
ER Vivid Red In vitro (rCYPs, microsomes, living 2.80 18.2 530 590 [132]
cells)

a
CHPO Vivid Red In vitro (rCYPs, microsomes) 0.278 9.17 540 590 [133]

a
CEC Vivid Blue In vitro (rCYPs, microsomes) 1.2 24.2 409 460 [134]

a
CYP1A2 NCMN NCHN In vitro (rCYPs, microsomes, living 9.8 32.06 450 564 [81]
cells)

NEMN NEHN In vitro (rCYPs, microsomes, living N.A. N.A. 458 552 [129]
cells)

a
ER Vivid Red In vitro (rCYPs, microsomes, living 3.0 0.23 530 590 [132]
cells)

a
CEC Vivid Blue In vitro (rCYPs, microsomes) 1.14 7.13 409 460 [134]

EOMCC Vivid Blue In vitro (rCYPs, microsomes) 11 21.0c 415 460 [135]

a
BFC Vivid Cyan In vitro (rCYPs, microsomes) 4.1 11.1 410 510 [127]

a
CYP1B1 ER Vivid Red In vitro (rCYPs, microsomes, living 1.0 1.33 530 590 [132]
cells)

(continued on next page)

13
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

Table 4 (continued)

Enzyme Substrate Substrate structure Metabolite Applications Km Vmax or kex kem Ref.
name (lM) Kcat (nm) (nm)
CYP2A6 Coumarin 7-OH- In vitro (rCYPs, microsomes) 2.1 0.79b 390 460 [136]
coumarin

a
CYP2B6 EFC Vivid Cyan In vitro (rCYPs, microsomes) 8.5 14.0 409 530 [137]

a
MFC Vivid Cyan In vitro (rCYPs, microsomes) 6.79 6.48 409 530 [134]

BOMCC Vivid Blue In vitro (rCYPs, microsomes) 52 66.0c 415 460 [135]

BOMR Vivid Red In vitro (rCYPs, microsomes) 0.73 0.80c 550 590 [135]

CYP2C8 DBOMF Vivid In vitro (rCYPs, microsomes) 3.6 2.10c 490 520 [135]
Green

CYP2C9 BOMCC Vivid Blue In vitro (rCYPs, microsomes) 43 2.10c 425 460 [135]

BOMF Vivid In vitro (rCYPs, microsomes) 4.5 4.50c 490 520 [135]
Green

a
MFC Vivid Cyan In vitro (rCYPs, microsomes) 54.7 0.56 409 530 [134]

a
CYP2C19 CEC Vivid Blue In vitro (rCYPs, microsomes) 9.11 0.84 409 460 [134]

EOMCC Vivid Blue In vitro (rCYPs, microsomes) 16.6 5.70c 415 460 [135]

OMF Vivid In vitro (rCYPs, microsomes) 1.14 0.01b 485 530 [126]
Green

CYP2D6 MAMC HAMC In vitro (rCYPs, microsomes) 11.5 2.81c 390 460 [138]

a
AMMC AHMC In vitro (rCYPs, microsomes) 4.81 6.19 390 460 [134]

MOBFC Vivid Cyan In vitro (rCYPs, microsomes) 5.0 0.60c 405 490 [135]

14
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

Table 4 (continued)

Enzyme Substrate Substrate structure Metabolite Applications Km Vmax or kex kem Ref.
name (lM) Kcat (nm) (nm)
a
CYP2E1 EOMCC Vivid Blue In vitro (rCYPs, microsomes) 22 2.10 415 460 [139]

d
CYP2J2 BnXPI HXPI In vitro (rCYPs, microsomes, living 4.2 0.44 656 718 [91]
cells)

a
CYP3A4 NEN NEHN In vitro (rCYPs, microsomes, living 59.8 2.18 450 558 [140]
cells and tissues)

BQ Quinolinol In vitro (rCYPs, microsomes) 70 3.39b 420 530 [128]

a
DBF Vivid In vitro (rCYPs, microsomes) 1.03 1.55 485 538 [134]
Green

DBOMF Vivid In vitro (rCYPs, microsomes) 7.8 16.0c 490 520 [135]
Green

BOMR Vivid Red In vitro (rCYPs, microsomes) 1.4 1.10c 550 590 [135]

a
BFC Vivid Cyan In vitro (rCYPs, microsomes) 26 61.2 410 510 [127]

BFBFC Vivid Cyan In vitro (rCYPs, microsomes) 4.6 0.02b 410 510 [128]

BOMCC Vivid Blue In vitro (rCYPs, microsomes) 89 10.0c 415 460 [135]

BOMFC Vivid Cyan In vitro (rCYPs, microsomes) 22 4.00c 405 490 [135]

CYP3A5 BOMCC Vivid Blue In vitro (rCYPs, microsomes) N.A. N.A. 415 460 [141]

DBOMF Vivid In vitro (rCYPs, microsomes) N.A. N.A. 490 520 [141]
Green

(continued on next page)

15
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

Table 4 (continued)

Enzyme Substrate Substrate structure Metabolite Applications Km Vmax or kex kem Ref.
name (lM) Kcat (nm) (nm)
a
DBF Vivid In vitro (rCYPs, microsomes) 2.17 1.28 490 520 [134]
Green

a
Vmax values were in nmol/min/nmol P450 for CYP enzymes; b Vmax values were in nmol/min/mg human liver microsome; c kcat or Vmax values were in min1; d Vmax values
were in nmol/min/mg CYP2J2; kex: excitation wavelength; kem: emission wavelength. NA.: Not available or not assayed. Red arrows indicate the major site of metabolism by
CYP.

Fig. 8. Two reaction steps involved in bioluminescent CYP assays and luciferin recycling process.

Over the past few decades, multiple bioluminogenic CYP sub- 7. Biomedical applications of the probes for human CYPs
strates have been developed under the trade name P450-GloTM
from Promega Corp. [149]. Amongst these bioluminogenic sub- 7.1. Sensing enzymatic activity in biological samples
strates, several have been identified as specific substrates for a tar-
get CYP isoenzyme, others have a broader enzyme profile. As listed As the most important xenobiotic-metabolizing enzymes in
in Table 5, the relatively selective luminogenic substrates for mammals, CYPs play a crucial role in detoxification and metabolic
CYP1A1, 1A2, 2B6, 2D6, 2C9, 2C19, 2J2, 3A4, 3A7, 4A and CYP4F clearance of a wide range of xenobiotics. Dysfunction or inhibition
have been reported. However, except Luciferin-H and Luciferin- of CYPs may have great impact on drug safety, efficacy and dose
IPA have been validated as highly specific probe substrates for requirement. Increasing evidence has indicated that the large vari-
CYP2A9, and CYP3A4, respectively, majority of luminogenic CYP ability in the function of some key CYPs may result in significant
probes display poor selectivity, these luminogenic substrates are interindividual variations in drug disposition and treatment out-
limited to inhibitor screening using recombinant CYP enzymes as comes, especially those drugs with narrow therapeutic index
enzyme sources. As far as we know, the specificity of a given (NTI), such as warfarin, digoxin and paclitaxel [157,158]. The
luminogenic substrate towards a target CYP was mainly affected highly specific molecular probes for target human CYPs have pro-
by the chemical structure of the luciferin derivatives and the most vided powerful tools for reliable sensing the real activities of target
labile sites of metabolism of CYPs. Two common CYP reactions, O- CYPs in various biological systems. In fact, some molecular probes
dealkylation and aromatic hydroxylation, are usually involved in for human CYPs have been utilized to investigate the interindivid-
the design and development of luminogenic probes [56,150]. In ual variability in CYP activities. For example, Court’s group has
addition, the modifications of these luminogenic substrates are investigated the interindividual variations in the activities of sev-
based on the structural features of target CYP (including catalytic eral key drug metabolizing enzymes in human liver microsomes
cavity and the key residues surrounding the catalytic site) and (n = 55) from individual donors, using several specific non-
the structures of their preferred ligands. For example, a previous fluorogenic probes [10]. The results reveal that CYP3A4 (midazo-
study haa found that the molecular weights and critical volumes lam 10 -hydroxylation) shows the highest interindividual variability
of Luciferin-H (a CYP2C9 luminogenic substrate) are similar to that (111% CV), while other hepatic CYPs, including CYP2E1 (chlorzox-
of flurbiprofen (a conventional CYP2C9 probe substrate), and azone 6-hydroxylation), display moderate interindividual variabil-
Luciferin-H was modeled in the CYP2C9 active site starting with ity (35% CV) [10]. Furthermore, Qiao’s group has determined the
the carboxylic acid anchored by a hydrogen bond and salt bridge activities of nine CYP enzymes in 78 individual human liver micro-
to the same residues as observed with flurbiprofen [142]. In future, somes using non-fluorogenic CYP probes [159]. The CYP activities
more detailed in silico molecular modelling and docking simula- show large individual variations, with the largest interindividual
tions should be carry out to find out the clarification of these highly variations occurred in the microsomal activities (VM) of CYP2C19
specific luminogenic substrates for CYPs. and CYP2A6, reaching 232 and 109-fold, respectively, followed by

16
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

Table 5
The bioluminogenic substrates for human CYPs.
c
Enzyme Substrate Substrate structure Metabolite Applications Km k (nm) Refs.
name (lM)
CYP1A1 Luciferin- D- In vitro (rCYPs, 3.0 560 [142,151]
a
1A1 Luciferin microsomes)

CYP1A2 Luciferin- D- In vitro (rCYPs, 6.0 560 [142,149]


a
1A2 Luciferin microsomes)

CYP2B6 Luciferin- D- In vitro (rCYPs, 3.0 560 [149]


a
2B6 Luciferin microsomes)

CYP2C9 Luciferin-H D- In vitro (rCYPs, 100 560 [142,149]


Luciferin microsomes)

CYP2C19 Luciferin-H D- In vitro (rCYPs, 10 560 [149]


EGE Luciferin microsomes)

CYP2D6 Luciferin- D- In vitro (rCYPs, 30 560 [149]


ME EGE Luciferin microsomes)

CYP2J2 Luciferin- D- In vitro (rCYPs, 1.0 560 [142,152]


2 J2/4F12 * Luciferin microsomes)

CYP3A4 Luciferin- D- In vitro (rCYPs, 3.0 560 [142,149]


IPA Luciferin microsomes, living
cells)

CYP3A7 Luciferin- D- In vitro (rCYPs, 33b 560 [142,153]


3A7 Luciferin microsomes)

CYP4A11 Luciferin- D- In vitro (rCYPs, 80 560 [142,154]


4A* Luciferin microsomes)

CYP4F3B Luciferin- D- In vitro (rCYPs, ~10b 560 [142,155]


4F2/3* Luciferin microsomes)

(continued on next page)

17
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

Table 5 (continued)
c
Enzyme Substrate Substrate structure Metabolite Applications Km k (nm) Refs.
name (lM)
CYP4F12 Luciferin- D- In vitro (rCYPs, 10 560 [142,156]
4F12* Luciferin microsomes)

a
The product of the Luciferin-1A1, Luciferin-1A2 and Luciferin-2B6 is a luciferin precursor, which can further generate D-luciferin by addition of luciferin detection reagent
supplemented with d-Cysteine (Fig. 8). b The reactions showed atypical kinetics. c k: the bioluminescence wavelength. Red arrows indicate the major site of metabolism by
CYP. * Currently, Luciferin-2J2/4F12, Luciferin-4A, Luciferin-4F2/3, and Luciferin-4F12, are not commercially available.

that of CYP3A4/5, CYP2D6, CYP2C8, CYP2B6, CYP1A2 and CYP2E1 sible for non-NTI drugs, such as statins. Regulators, including FDA,
(Fig. 9). Notably, interindividual variations of CYP activities based have therefore issued guidance for in vitro and in vivo drug interac-
on liver tissues (VL) were even more pronounced, except for tion studies that are recommended to be conducted during devel-
CYP2C19 [159]. More recently, Vos’s group has measured multiple opment [11]. Predicting the DDI potential for a new drug usually
drug metabolizing enzyme activities in a set of 20 individual liver relies on the evaluation of the effect on the rate of a probe reaction
homogenates, among which, the strongest inter-individual vari- that represents the activity of a specific CYP enzyme [74]. Over the
ability in enzyme-specific activities were observed for CYP2C19 past few decades, various non-fluorogenic CYP probes have been
and CYP2B6 (CV ~ 78%) by employing mephenytoin and bupropion utilized for screening and characterization of CYP modulators,
as the probe substrates, respectively [8]. and subsequently to assess their potential risks to trigger CYP-
mediated DDI in humans [11]. Meanwhile, fluorogenic probes have
gotten more and more attention in the assessment of DDI, due to
7.2. Screening of CYP modulators their advantage of throughput capabilities [163,164]. For example,
7-ethoxy-resorufin (ER) has been used as a fluorogenic probe for
Numerous clinical surveys have highlighted continuing issues CYP1A, which allows high-throughput screening (HTS). The inhibi-
related to drug/herb-drug interactions (DDI/HDI) and the resulting tory potentials of rutaecarpine, evodiamine, and dehydroevodi-
serious adverse events in clinical settings [160,161]. Assessing the amine, the alkaloids isolated from a traditional Chinese medicine
potential risks of DDI/HDI is an important task in the development towards CYP1A1 and CYP1A2, were evaluated using ER, and the
of new drugs. Inhibition of human CYPs responsible for metabolic results indicated that rutaecarpine is a potent inhibitor of CYP1A2
clearance of many therapeutic drugs has been considered as one of in human liver microsomes [165]. Furthermore, the fluorogenic
the key causes of DDI/HDI. Therefore, assessing the inhibition probe of CYP is highly favoured in evaluating the induction poten-
potency of human CYPs by a new chemical entity has become a tial of CYP inducers in living cells, due to its high sensitivity. For
routine practice during the drug discovery stage [162]. The large example, NBCeN, a CYP1A1 probe, has been successfully used to
changes in exposure triggered by modulation (inhibition, induction evaluate the activity of CYP1A1 in hepG2 cells pre-treated with
or stimulation) of CYP activities can alter the efficacy and safety 2,3,7,8-tetrachlorodibenzo-p-dioxin, a well-known CYP1A inducer
profile of a drug. This is most obvious for NTI drugs, but is also pos- [81], and the results showed that CYP1A1 activity agreed well with
its protein levels (Fig. 10). Taken together, chemical probes have
become promising tools for screening and characterization of CYP
modulators, and more practical probe substrates with high speci-
ficity and high sensitivity are highly desirable.

Fig. 9. The individual fold-change of CYP activity in human livers (n = 78). The fold-
change of CYP activity is expressed as the ratio between the maximal (Max) and
minimal (Min) values of CYP metabolic rate. Phenacetin (CYP1A2), coumarin
(CYP2A6), bupropion (CYP2B6), paclitaxel (CYP2C8), tolbutamide (CYP2C9),
omeprazole (CYP2C19), dextromethorphan (CYP2D6), chlorzoxazone (CYP2E1),
and midazolam (CYP3A4/5) were used as the isoform-specific probes, respectively.
VM: metabolic rate of each CYP isozymes per mg microsomal protein; VL: metabolic Fig. 10. Comparison of the CYP1A1 activity and protein levels in HepG2 cells pre-
rate of each CYP isozymes per gram liver tissue, which derived from the VM values treated with CYP1A inducer by using NBCeN as a CYP1A1 probe and western blot
multiplied by the individual microsomal protein per gram of liver. See ref. [159]. analysis, respectively. See ref. [83].

18
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

7.3. CYP–ligand interactions CYPs in living cells or tissues. Although a wide range of specific
probes for human CYPs have been reported, most of known CYP
Understanding the key structural elements and the key moiety probe substrates are hardly used in living systems, due to low sen-
of ligands that dictate their orientation in the active site of the tar- sitivity, poor chemical stability, high cytotoxicity or other safety
get CYP is very helpful for deep understanding of CYP–ligand inter- concerns. Thus, more practical molecular probes for highly specific
actions, which may aid the design and development of highly and highly sensitive sensing of target CYP enzymes in living sys-
specific ligands (such as highly specific inhibitors and substrates) tems are always desirable.
[49]. For some key human CYPs (such as CYP3A4), there are multi- The major challenge to develop practical CYP probes is the lim-
ple ligand-binding sites in the catalytic cavity. In these cases, the ited ability to enhance their specificity and sensitivity. With the
researches should know whether the optical substrates for the tar- help of structural biology techniques that make the crystal struc-
get enzyme could replace its physiological substrates or its tures of more CYP enzymes available, the structure-based probe
substrate-drugs. [75,84,166]. An inappropriate selection of a probe development using computer-aided design and virtual screening
substrate may lead to false positive or negative results to predict will likely improve the isoform-specificity of CYP probes to a large
the DDI risks [74]. For example, testosterone, midazolam, and extent. To improve the sensitivity of candidate probe substrates,
nifedipine are frequently used as three substrate subgroups for several feasible strategies, such as increasing turnover rate,
CYP3A4, and multiple molecular probes are recommended to be enhancing the fluorescence quantum yield of fluorogenic sub-
used for CYP3A4-mediated DDI assays [166]. Wu et al. have strates, or dwindling the reaction group size to reduce the energy
reported that deoxyschizandrin is a good substrate to replace consumption of CYP reactions, could be used for future design
midazolam and testosterone, but not nifedipine (Fig. 11) [75]. This and development of chemical probes for CYPs.
was concluded from mutual inhibition assays. Testosterone and In light of the fact that fluorescence-based assays have multiple
midazolam were shown to compete with deoxyschizandrin for inherent advantages, such as highly sensitive, easy to manage, and
the mutual binding site. However, deoxyschizandrin showed only applicable to HTS, there is a great deal of interest in the design and
slight inhibitory effects on nifedipine oxidation even though development of highly specific and practical fluorescent probes for
nifedipine was able to inhibit deoxyschizandrin hydroxylation via target CYPs. The design strategies and experiences in the develop-
a noncompetitive manner [75]. Taken together, site-specific probes ment of isoform-specific fluorogenic substrates for CYP1A1 and
for CYPs are in high demand for the studies on interactions other CYP enzymes may assist others to develop more practical flu-
between the small-molecule ligands and CYPs, especially the CYPs orescent substrates for highly specific and sensitive sensing of tar-
with multiple ligand-binding sites. get CYP enzyme(s) in complex biological systems. These optical
probes can be used to develop a set of fluorescence-based CYP
detection microarrays, which in turn, at least partially, take place
8. Challenges and future perspectives
of the physiological substrates or those CYP substrates with rela-
tively poor enzyme-specificity and low conversion rate [171]. Of
Molecular probes are practical tools for deciphering the physio-
particular note that the optical probes has been tried ~15 years
logical functions of target enzyme, as well as for exploring the
ago in the pharmaceutical industry, but there are a number of
interactions between target enzyme and its ligands [167–170].
issues & many companies still use the physiological substrates
Over the past few decades, many strategies and approaches have
(such as testosterone) or the drug substrates (such as diclofenac)
been used to develop specific molecular probes for sensing the
for CYP inhibition assays, owing to the man-made substrates
activities of CYPs in complex biological systems, living cells or
may bind on the target enzyme (such as CYP3A4) at a distinct
in vivo [81–83]. At present, CYP probes have been widely used in
ligand-binding site [166]. Therefore, it is highly recommended that
biological and biomedical fields, including sensing CYP activity in
the difference in ligand-binding site and the binding mode of the
various biological samples, high-throughput screening of CYP inhi-
man-made optical substrates and the physiological or drug sub-
bitors or inducers, as well as biological imaging of intracellular
strates on a target enzyme should be tested, prior to inhibition
assays [84].
Although significant breakthroughs have been achieved in the
design and development of optical probes for human CYPs, the
emission wavelengths of most reported molecular probes for
human CYPs and their corresponding metabolites are less than
560 nm, which may be interfered by the background fluorescence
signals in living systems. To get more practical CYP probes, more
fluorogens or luminogens with diverse chemical structures (such
as green fluorescent protein (GFP)-like and red fluorescent protein
(GFP)-like fluorophores, as well as aggregation-induced emission
luminogens) should be used to construct the fluorogenic probe
substrates for a target human CYP [172–174]. Meanwhile, exten-
sive investigations on the design and development of CYP probes
with high specificity, excellent optical properties (e.g., near-
infrared wavelength probes), improved safety profiles and good
biological compatibility are highly desirable, which will strongly
facilitate real-time monitoring and in situ imaging of the activities
of target CYPs in vivo, and exploring the relevance of CYP enzymes
to human diseases. Furthermore, most previous studies are focused
on the development of molecular probes for xenobiotic-endobio
tic-metabolizing CYPs. Until now, no optical probes have been
Fig. 11. The proposed binding site distribution of MDZ, TST, NIF and DS within
developed for in vivo or in situ sensing or imaging of endobiotic-
CYP3A4 enzyme. DS: deoxyschizandrin, MDZ: midazolam, TST: testosterone, NIF: metabolizing CYPs, owing to the lack of 3D structural information
nifedipine. of the target endobiotic-metabolizing CYPs and the limited
19
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

knowledge about their substrate preferences. In future, more conduct of in vitro and in vivo drug-drug interaction studies: a
Pharmaceutical Research and Manufacturers of America (PhRMA)
investigations still need to be done using isoform-specific sub-
perspective, Drug Metab Dispos, 31 (2003) 815-832. https://doi.org/
strates for target human CYP(s), to gain better understanding of 10.1124/dmd.31.7.815.
the biological functions, substrate preferences and the mechanisms [12] A. Guglielmi, A. Ruzzenente, S. Conci, A. Valdegamberi, C. Iacono, How much
of catalysis of these key oxidative enzymes, as well as to explain remnant is enough in liver resection?, Dig Surg 29 (2012) 6–17, https://
doiorg/10.1159/000335713.
how regulation of these enzymes is beneficial for improving or [13] A.S. Kalgutkar, R.S. Obach, T.S. Maurer, Mechanism-based inactivation of
maintaining of human health. cytochrome P450 enzymes: chemical mechanisms, structure-activity
relationships and relationship to clinical drug-drug interactions and
idiosyncratic adverse drug reactions, Curr. Drug Metab. 8 (2007) 407–447,
Declaration of Competing Interest https://doi.org/10.2174/138920007780866807.
[14] J. Lazarou, B.H. Pomeranz, P.N. Corey, Incidence of adverse drug reactions in
The authors declare that they have no known competing finan- hospitalized patients: a meta-analysis of prospective studies, JAMA 279
(1998) 1200–1205, https://doi.org/10.1001/jama.279.15.1200.
cial interests or personal relationships that could have appeared [15] R. Leone, L. Magro, U. Moretti, P. Cutroneo, M. Moschini, D. Motola, M.
to influence the work reported in this paper. Tuccori, A. Conforti, Identifying adverse drug reactions associated with drug-
drug interactions, Drug Saf. 33 (2010) 667–675, https://doi.org/10.2165/
11534400-000000000-00000.
Acknowledgements [16] T.L. Poulos, E.F. Johnson, Structures of Cytochrome P450 Enzymes, in: P.R.
Ortiz de Montellano (Ed.), Cytochrome P450: Structure, Mechanism, and
This work was supported by the National Natural Science Foun- Biochemistry, Springer, Cham, New York, 2015, pp. 3–32.
[17] M. Otyepka, J. Skopalik, E. Anzenbacherova, P. Anzenbacher, What common
dation of China (81922070, 81973286, 81773687), the National
structural features and variations of mammalian P450s are known to date?,
Key Research and Development Program of China Bba-Gen Subjects 1770 (2007) 376–389, https://doi.org/10.1016/j.
(2020YFC0845400, 2017YFC1700200, 2017YFC1702000), the bbagen.2006.09.013.
[18] E.F. Johnson, C.D. Stout, Structural diversity of human xenobiotic-
Three-year Action Plan of Shanghai TCM Development (ZY-(2018-
metabolizing cytochrome P450 monooxygenases, Biochem. Biophys. Res.
2020)-CCCX-5001), Liaoning Revitalization Talents Program Commun. 338 (2005) 331–336, https://doi.org/10.1016/j.bbrc.2005.08.190.
(XLYC1907103), Program of Shanghai Academic/Technology [19] E.F. Johnson, C.D. Stout, Structural diversity of eukaryotic membrane
Research Leader (18XD1403600), the 111 Project (Grant B16047), cytochrome P450s, J. Biol. Chem. 288 (2013) 17082–17090, https://doi.org/
10.1074/jbc.R113.452805.
and Shuguang Program (No. 18SG40) supported by Shanghai Edu- [20] P.A. Williams, J. Cosme, D.M. Vinkovic, A. Ward, H.C. Angove, P.J. Day, C.
cation Development Foundation and Shanghai Municipal Educa- Vonrhein, I.J. Tickle, H. Jhoti, Crystal structures of human cytochrome P450
tion Commission. 3A4 bound to metyrapone and progesterone, Science 305 (2004) 683–686,
https://doi.org/10.1126/science.1099736.
[21] A.A. Walsh, G.D. Szklarz, E.E. Scott, Human Cytochrome P450 1A1 Structure
References and Utility in Understanding Drug and Xenobiotic Metabolism, J. Biol. Chem.
288 (2013) 12932–12943, https://doi.org/10.1074/jbc.M113.452953.
[1] A.W. Munro, H.M. Girvan, A.E. Mason, A.J. Dunford, K.J. McLean, What makes a [22] S. Sansen, J.K. Yano, R.L. Reynald, G.A. Schoch, K.J. Griffin, C.D. Stout, E.F.
P450 tick?, Trends Biochem. Sci. 38 (2013) 140–150, https://doi.org/10.1016/ Johnson, Adaptations for the oxidation of polycyclic aromatic hydrocarbons
j.tibs.2012.11.006. exhibited by the structure of human P450 1A2, J. Biol. Chem. 282 (2007)
[2] F.P. Guengerich, Human cytochrome P450 enzymes, in: P.R. Ortiz de 14348–14355, https://doi.org/10.1074/jbc.M611692200.
Montellano (Ed.), Cytochrome P450, Springer, Cham, New York, 2015, pp. [23] J.K. Yano, M.H. Hsu, K.J. Griffin, C.D. Stout, E.F. Johnson, Structures of human
523–785. microsomal cytochrome P450 2A6 complexed with coumarin and
[3] U.M. Zanger, M. Schwab, Cytochrome P450 enzymes in drug metabolism: methoxsalen, Nat. Struct. Mol. Biol. 12 (2005) 822–823, https://doi.org/
regulation of gene expression, enzyme activities, and impact of genetic 10.1038/nsmb971.
variation, Pharmacol. Ther. 138 (2013) 103–141, https://doi.org/10.1016/j. [24] N.M. DeVore, E.E. Scott, Nicotine and 4-(methylnitrosamino)-1-(3-pyridyl)-1-
pharmthera.2012.12.007. butanone binding and access channel in human cytochrome P450 2A6 and
[4] M.I. Fekry, Y. Xiao, J.Z. Berg, F.P. Guengerich, A role for the orphan human 2A13 enzymes, J. Biol. Chem. 287 (2012) 26576–26585, https://doi.org/
cytochrome P450 2S1 in polyunsaturated fatty acid x-1 hydroxylation using 10.1074/jbc.M112.372813.
an untargeted metabolomic approach, Drug Metab. Dispos. 47 (2019) 1325– [25] P.R. Wilderman, M.B. Shah, H.H. Jang, C.D. Stout, J.R. Halpert, Structural and
1332, https://doi.org/10.1124/dmd.119.089086. thermodynamic basis of (+)-alpha-pinene binding to human cytochrome
[5] Y. Xiao, F.P. Guengerich, Metabolomic analysis and identification of a role for P450 2B6, J. Am. Chem. Soc. 135 (2013) 10433–10440, https://doi.org/
the orphan human cytochrome P450 2W1 in selective oxidation of 10.1021/ja403042k.
lysophospholipids, J. Lipid Res. 53 (2012) 1610–1617, https://doi.org/ [26] G.A. Schoch, J.K. Yano, S. Sansen, P.M. Dansette, C.D. Stout, E.F. Johnson,
10.1194/jlr.M027185. Determinants of cytochrome P450 2C8 substrate binding: structures of
[6] Y. Ohno, S. Nakamichi, A. Ohkuni, N. Kamiyama, A. Naoe, H. Tsujimura, U. complexes with montelukast, troglitazone, felodipine, and 9-cis-retinoic acid,
Yokose, K. Sugiura, J. Ishikawa, M. Akiyama, Essential role of the cytochrome J. Biol. Chem. 283 (2008) 17227–17237, https://doi.org/10.1074/jbc.
P450 CYP4F22 in the production of acylceramide, the key lipid for skin M802180200.
permeability barrier formation, Proc. Natl. Acad. Sci. 112 (2015) 7707–7712, [27] P.A. Williams, J. Cosme, A. Ward, H.C. Angove, D. Matak, Vinkovic H. Jhoti,
https://doi.org/10.1073/pnas.1503491112. Crystal structure of human cytochrome P450 2C9 with bound warfarin,
[7] Q. Yan, D. Machalz, A. Zöllner, E.J. Sorensen, G. Wolber, M. Bureik, Efficient Nature 424 (2003) 464–468, https://doi.org/10.1038/nature01862.
substrate screening and inhibitor testing of human CYP4Z1 using [28] M.R. Wester, J.K. Yano, G.A. Schoch, C. Yang, K.J. Griffin, C.D. Stout, E.F.
permeabilized recombinant fission yeast, Biochem. Pharmacol. 146 (2017) Johnson, The structure of human cytochrome P450 2C9 complexed with
174–187, https://doi.org/10.1016/j.bcp.2017.09.011. flurbiprofen at 2.0-A resolution, J. Biol. Chem. 279 (2004) 35630–35637,
[8] S.P. Den Braversewradj, M.W. Den Braver, M. Van Dijk, Y. Zhang, S.J. Dekker, L. https://doi.org/10.1074/jbc.M405427200.
Wijaya, N.P.E. Vermeulen, L. Richert, J.N.M. Commandeur, J.C. Vos, Inter- [29] R.L. Reynald, S. Sansen, C.D. Stout, E.F. Johnson, Structural characterization of
individual variability in activity of the major drug metabolizing enzymes in human cytochrome P450 2C19: active site differences between P450s 2C8,
liver homogenates of 20 Individuals, Curr. Drug Metab. 19 (2018) 370–381, 2C9, and 2C19, J. Biol. Chem. 287 (2012) 44581–44591, https://doi.org/
https://doi.org/10.2174/1389200219666180108160046. 10.1074/jbc.M112.424895.
[9] B. Achour, M.R. Russell, J. Barber, A. Rostami-Hodjegan, Simultaneous [30] A. Wang, C.D. Stout, Q.H. Zhang, E.F. Johnson, Contributions of ionic
quantification of the abundance of several cytochrome P450 and uridine 5 interactions and protein dynamics to cytochrome P450 2D6 (CYP2D6)
’-diphospho-glucuronosyltransferase enzymes in human liver microsomes substrate and inhibitor binding, J. Biol. Chem. 290 (2015) 5092–5104,
using multiplexed targeted proteomics, Drug Metab. Dispos. 42 (2014) 500– https://doi.org/10.1074/jbc.M114.627661.
510, https://doi.org/10.1124/dmd.113.055632. [31] P.R. Porubsky, K.M. Meneely, E.E. Scott, Structures of human cytochrome P-
[10] M.H. Court, Interindividual variability in hepatic drug glucuronidation: 450 2E1. Insights into the binding of inhibitors and both small molecular
studies into the role of age, sex, enzyme inducers, and genetic weight and fatty acid substrates, J Biol Chem 283 (2008) 33698–33707,
polymorphism using the human liver bank as a model system, Drug Metab. https://doi.org/10.1074/jbc.M805999200.
Rev. 42 (2010) 209–224, https://doi.org/10.3109/03602530903209288. [32] I.F. Sevrioukova, T.L. Poulos, Structural basis for regiospecific midazolam
[11] T. D. Bjornsson, J. T. Callaghan, H. J. Einolf, V. Fischer, L. Gan, S. Grimm, J. Kao, oxidation by human cytochrome P450 3A4, Proc. Natl. Acad. Sci. U.S.A. 114
S. P. King, G. Miwa, L. Ni, G. Kumar, J. McLeod, R. S. Obach, S. Roberts, A. Roe, A. (2017) 486–491, https://doi.org/10.1073/pnas.1616198114.
Shah, F. Snikeris, J. T. Sullivan, D. Tweedie, J. M. Vega, J. Walsh, S. A. Wrighton, [33] I.F. Sevrioukova, T.L. Poulos, Interaction of human cytochrome P4503A4 with
R. Pharmaceutical, G. Manufacturers of America Drug Metabolism/Clinical ritonavir analogs, Arch. Biochem. Biophys. 520 (2012) 108–116, https://doi.
Pharmacology Technical Working, F. D. A. C. f. D. Evaluation,Research, The org/10.1016/j.abb.2012.02.018.

20
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

[34] I.F. Sevrioukova, T.L. Poulos, Structure and mechanism of the complex [58] R. Bernhardt, Cytochromes P450 as versatile biocatalysts, J. Biotechnol. 124
between cytochrome P4503A4 and ritonavir, Proc. Natl. Acad. Sci. U.S.A. 107 (2006) 128–145, https://doi.org/10.1016/j.jbiotec.2006.01.026.
(2010) 18422–18427, https://doi.org/10.1073/pnas.1010693107. [59] L. Olsen, C. Oostenbrink, F.S. Jorgensen, Prediction of cytochrome P450
[35] M.H. Hsu, U. Savas, E.F. Johnson, The X-ray crystal structure of the human mediated metabolism, Adv. Drug Deliv. Rev. 86 (2015) 61–71, https://doi.org/
mono-oxygenase cytochrome P450 3A5-ritonavir complex reveals active site 10.1016/j.addr.2015.04.020.
differences between P450s 3A4 and 3A5, Mol. Pharmacol. 93 (2018) 14–24, [60] Y. Xiong, Y. Qiao, D. Kihara, H. Zhang, X. Zhu, D. Wei, Survey of machine
https://doi.org/10.1124/mol.117.109744. learning techniques for prediction of the isoform specificity of cytochrome
[36] A. Wang, U. Savas, C.D. Stout, E.F. Johnson, Structural characterization of the P450 substrates, Curr. Drug Metab. 20 (2019) 229–235, https://doi.org/
complex between alpha-naphthoflavone and human cytochrome P450 1B1, J. 10.2174/1389200219666181019094526.
Biol. Chem. 286 (2011) 5736–5743, https://doi.org/10.1074/jbc. [61] J.D. Tyzack, J. Kirchmair, Computational methods and tools to predict
M110.204420. cytochrome P450 metabolism for drug discovery, Chem. Biol. Drug Des. 93
[37] W. Tempel, I. Grabovec, F. MacKenzie, Y.V. Dichenko, S.A. Usanov, A.A. Gilep, (2019) 377–386, https://doi.org/10.1111/cbdd.13445.
H.W. Park, N. Strushkevich, Structural characterization of human cholesterol [62] J. Kirchmair, M.J. Williamson, J.D. Tyzack, L. Tan, P.J. Bond, A. Bender, R.C.
7alpha-hydroxylase, J. Lipid Res. 55 (2014) 1925–1932, https://doi.org/ Glen, Computational prediction of metabolism: sites, products, SAR, P450
10.1194/jlr.M050765. enzyme dynamics, and mechanisms, J. Chem. Inf. Model. 52 (2012) 617–648,
[38] N. Strushkevich, F. MacKenzie, T. Cherkesova, I. Grabovec, S. Usanov, H.W. https://doi.org/10.1021/ci200542m.
Park, Structural basis for pregnenolone biosynthesis by the mitochondrial [63] E. Stjernschantz, C. Oostenbrink, Improved ligand-protein binding affinity
monooxygenase system, Proc. Natl. Acad. Sci. U.S.A. 108 (2011) 10139– predictions using multiple binding modes, Biophys. J. 98 (2010) 2682–2691,
10143, https://doi.org/10.1073/pnas.1019441108. https://doi.org/10.1016/j.bpj.2010.02.034.
[39] N.M. DeVore, E.E. Scott, Structures of cytochrome P450 17A1 with prostate [64] L. Perichassler, E. Stjernschantz, C. Oostenbrink, D.P. Geerke, CYP 2D6 binding
cancer drugs abiraterone and TOK-001, Nature 482 (2012) 116–119, https:// affinity predictions using multiple ligand and protein conformations, Int. J.
doi.org/10.1038/nature10743. Mol. Sci. 14 (2013) 24514–24530, https://doi.org/10.3390/ijms141224514.
[40] D. Ghosh, J. Griswold, M. Erman, W. Pangborn, Structural basis for androgen [65] U. Bren, C. Oostenbrink, Cytochrome P450 3A4 inhibition by ketoconazole:
specificity and oestrogen synthesis in human aromatase, Nature 457 (2009) tackling the problem of ligand cooperativity using molecular dynamics
219–223, https://doi.org/10.1038/nature07614. simulations and free-energy calculations, J. Chem. Inf. Model. 52 (2012)
[41] P. S. Pallan, C. Wang, L. Lei, F. K. Yoshimoto, R. J. Auchus, M. R. Waterman, F. P. 1573–1582, https://doi.org/10.1021/ci300118x.
Guengerich,M. Egli, Human Cytochrome P450 21A2, the Major Steroid 21- [66] M. Paloncýova, V. Navratilova, K. Berka, A. Laio, M. Otyepka, Role of enzyme
Hydroxylase: STRUCTURE OF THE ENZYME.PROGESTERONE SUBSTRATE flexibility in ligand access and egress to active site: bias-exchange
COMPLEX AND RATE-LIMITING C-H BOND CLEAVAGE, J Biol Chem, 290 metadynamics study of 1,3,7-trimethyluric acid in cytochrome P450 3A4, J.
(2015) 13128-13143. https://doi.org/10.1074/jbc.M115.646307. Chem. Theory Comput. 12 (2016) 2101–2109, https://doi.org/10.1021/acs.
[42] N. Mast, M.A. White, I. Bjorkhem, E.F. Johnson, C.D. Stout, I.A. Pikuleva, Crystal jctc.6b00075.
structures of substrate-bound and substrate-free cytochrome P450 46A1, the [67] M.A. Olah, J.N. Harvey, Understanding the determinants of selectivity in drug
principal cholesterol hydroxylase in the brain, Proc. Natl. Acad. Sci. U.S.A. 105 metabolism through modeling of dextromethorphan oxidation by
(2008) 9546–9551, https://doi.org/10.1073/pnas.0803717105. cytochrome P450, Proc. Natl. Acad. Sci. U.S.A. 108 (2011) (2011) 6050–
[43] N. Mast, M. Linger, M. Clark, J. Wiseman, C.D. Stout, I.A. Pikuleva, In silico and 6055, https://doi.org/10.1073/pnas.1010194108.
intuitive predictions of CYP46A1 inhibition by marketed drugs with [68] P. Rydberg, M. Rostkowski, D.E. Gloriam, L. Olsen, The contribution of atom
subsequent enzyme crystallization in complex with fluvoxamine, Mol. accessibility to site of metabolism models for cytochromes P450, Mol. Pharm.
Pharmacol. 82 (2012) 824–834, https://doi.org/10.1124/mol.112.080424. 10 (2013) 1216–1223, https://doi.org/10.1021/mp3005116.
[44] N. Mast, C. Charvet, I.A. Pikuleva, C.D. Stout, Structural basis of drug binding [69] P. Rydberg, L. Olsen, Predicting drug metabolism by cytochrome P450 2C9:
to CYP46A1, an enzyme that controls cholesterol turnover in the brain, J. Biol. comparison with the 2D6 and 3A4 isoforms, Chem. Med. Chem. 7 (2012)
Chem. 285 (2010) 31783–31795, https://doi.org/10.1074/jbc.M110.143313. 1202–1209, https://doi.org/10.1002/cmdc.201200160.
[45] N. Strushkevich, S.A. Usanov, H.W. Park, Structural basis of human CYP51 [70] R. Liu, J. Liu, G. Tawa, A. Wallqvist, 2D SMARTCyp reactivity-based site of
inhibition by antifungal azoles, J. Mol. Biol. 397 (2010) 1067–1078, https:// metabolism prediction for major drug-metabolizing cytochrome P450
doi.org/10.1016/j.jmb.2010.01.075. enzymes, J. Chem. Inf. Model. 52 (2012) 1698–1712, https://doi.org/
[46] P.C. Nair, R.A. Mckinnon, J.O. Miners, Cytochrome P450 structure–function: 10.1021/ci3001524.
insights from molecular dynamics simulations, Drug Metab. Rev. 48 (2016) [71] H.J. Barnes, M.P. Arlotto, M.R. Waterman, Expression and enzymatic-activity
434–452, https://doi.org/10.1080/03602532.2016.1178771. of recombinant cytochrome-P450 17-alpha-hydroxylase in Escherichia-Coli,
[47] K.M. Huttunen, N. Mahonen, H. Raunio, J. Rautio, Cytochrome P450-activated Proc. Natl. Acad. Sci. U.S.A. 88 (1991) 5597–5601, https://doi.org/10.1073/
prodrugs: targeted drug delivery, Curr. Med. Chem. 15 (2008) 2346–2365, pnas.88.13.5597.
https://doi.org/10.2174/092986708785909120. [72] M.M. Galteau, F. Shamsa, Urinary 6b-hydroxycortisol: a validated test for
[48] D. Dong, B. Wu, D. Chow, M. Hu, Substrate selectivity of drug-metabolizing evaluating drug induction or drug inhibition mediated through CYP3A in
cytochrome P450s predicted from crystal structures and in silico modeling, humans and in animals, Eur. J. Clin. Pharmacol. 59 (2003) 713–733, https://
Drug Metab. Rev. 44 (2012) 192–208, https://doi.org/10.3109/ doi.org/10.1007/s00228-003-0690-3.
03602532.2011.645580. [73] T. Miura, M. Komori, M. Iwasaki, K. Kurozumi, K. Ohta, S. Ohmori, M. Kitada, T.
[49] S.C. Gay, A.G. Roberts, J.R. Halpert, Structural features of cytochromes P450 Kamataki, Sex-related difference in oxidative metabolism of testosterone and
and ligands that affect drug metabolism as revealed by X-ray crystallography erythromycin by hamster liver microsomes, FEBS Lett. 231 (1988) 183–186,
and NMR, Future Med. Chem. 2 (2010) 1451–1468, https://doi.org/10.4155/ https://doi.org/10.1016/0014-5793(88)80727-0.
fmc.10.229. [74] R. Yuan, S. Madani, X.X. Wei, K. Reynolds, Evaluation of cytochrome P450
[50] M. Ekroos, T. Sjogren, Structural basis for ligand promiscuity in cytochrome probe substrates commonly used by the pharmaceutical industry to study
P450 3A4, Proc. Natl. Acad. Sci. U.S.A. 103 (2006) 13682–13687, https://doi. in vitro drug interactions, Drug Metab. Dispos. 30 (2002) 1311–1319, https://
org/10.1073/pnas.0603236103. doi.org/10.1124/dmd.30.12.1311.
[51] J.K. Yano, M.R. Wester, G.A. Schoch, K.J. Griffin, C.D. Stout, E.F. Johnson, The [75] J.J. Wu, Y.F. Cao, Y.Y. Zhang, Y. Liu, J.Y. Hong, L.L. Zhu, G.B. Ge, L. Yang,
structure of human microsomal cytochrome P450 3A4 determined by X-ray Deoxyschizandrin, a naturally occurring lignan, is a specific probe substrate
crystallography to 2.05-A resolution, J. Biol. Chem. 279 (2004) 38091–38094, of human cytochrome P450 3A, Drug Metab. Dispos. 42 (2014) 94–104,
https://doi.org/10.1074/jbc.C400293200. https://doi.org/10.1124/dmd.113.053884.
[52] T. Omura, R. Sato, The Carbon Monoxide-Binding Pigment of Liver [76] J.J. Wu, G.B. Ge, Y.Q. He, P. Wang, Z.R. Dai, J. Ning, L.H. Hu, L. Yang, Gomisin A
Microsomes. I. Evidence for Its Hemoprotein Nature, J. Biol. Chem. 239 is a novel isoform-specific probe for the selective sensing of human
(1964) 2370–2378. cytochrome P450 3A4 in liver microsomes and living cells, AAPS J. 18
[53] A.W. Munro, H.M. Girvan, K.J. McLean, Variations on a (t)heme–novel (2016) 134–145, https://doi.org/10.1208/s12248-015-9827-4.
mechanisms, redox partners and catalytic functions in the cytochrome [77] Y.F. Cao, Y.Y. Zhang, J. Li, G.B. Ge, D. Hu, H.X. Liu, T. Huang, Y.C. Wang, Z.Z.
P450 superfamily, Nat. Prod. Rep. 24 (2007) 585–609, https://doi.org/ Fang, D.X. Sun, H. Huo, J. Yin, L. Yang, CYP3A catalyses schizandrin
10.1039/b604190f. biotransformation in human, minipig and rat liver microsomes, Xenobiotica
[54] S. Shaik, S. Cohen, Y. Wang, H. Chen, D. Kumar, W. Thiel, P450 enzymes: their 40 (2010) 38–47, https://doi.org/10.3109/00498250903366052.
structure, reactivity, and selectivitys modeled by QM/MM calculations, Chem. [78] J.J. Wu, Y.F. Cao, L. Feng, Y.Q. He, J.Y. Hong, T.Y. Dou, P. Wang, D.C. Hao, G.B.
Rev. 110 (2010) 949–1017, https://doi.org/10.1021/cr030722j. Ge, L. Yang, A naturally occurring isoform-specific probe for highly selective
[55] E.M. Isin, F.P. Guengerich, Complex reactions catalyzed by cytochrome P450 and sensitive detection of human cytochrome P450 3A5, J. Med. Chem. 60
enzymes, Bba-Gen Subjects 1770 (2007) 314–329, https://doi.org/10.1016/j. (2017) 3804–3813, https://doi.org/10.1021/acs.jmedchem.7b00001.
bbagen.2006.07.003. [79] Z.R. Dai, J. Ning, G.B. Sun, P. Wang, F. Zhang, H.Y. Ma, L.W. Zou, J. Hou, J.J. Wu, G.B.
[56] F.P. Guengerich, Common and uncommon cytochrome P450 reactions related Ge, X.B. Sun, L. Yang, Cytochrome P450 3A enzymes are key contributors for
to metabolism and chemical toxicity, Chem. Res. Toxicol. 14 (2001) 611–650, hepatic metabolism of bufotalin, a natural constitute in chinese medicine chansu,
https://doi.org/10.1021/tx0002583. Front. Pharmacol. 10 (2019) 52, https://doi.org/10.3389/fphar.2019.00052.
[57] I. Schlichting, J. Berendzen, K. Chu, A.M. Stock, S.A. Maves, D.E. Benson, B.M. [80] J. Ning, Z. Yu, L. Hu, C. Wang, X. Huo, S. Deng, J. Hou, J. Wu, G. Ge, X. Ma,
Sweet, D. Ringe, G.A. Petsko, S.G. Sligar, The catalytic pathway of cytochrome Characterization of phase I metabolism of resibufogenin and evaluation of the
P450cam at atomic resolution, Science 287 (2000) 1615–1622, https://doi. metabolic effects on its antitumor activity and toxicity, Drug Metab. Dispos.
org/10.1126/science.287.5458.1615. 43 (2015) 299–308, https://doi.org/10.1124/dmd.114.060996.

21
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

[81] Z.R. Dai, G.B. Ge, L. Feng, J. Ning, L.H. Hu, Q. Jin, D.D. Wang, X. Lv, T.Y. Dou, J.N. [102] A. Abelo, T.B. Andersson, M. Antonsson, A.K. Naudot, I. Skanberg, L. Weidolf,
Cui, L. Yang, A highly selective ratiometric two-photon fluorescent probe for Stereoselective metabolism of omeprazole by human cytochrome P450
human cytochrome P450 1A, J. Am. Chem. Soc. 137 (2015) 14488–14495, enzymes, Drug Metab. Dispos. 28 (2000) 966–972.
https://doi.org/10.1021/jacs.5b09854. [103] R.S. Foti, M.B. Fisher, Impact of incubation conditions on bufuralol human
[82] J. Ning, Z.H. Tian, B. Wang, G.B. Ge, Y. An, J. Hou, C. Wang, X.Y. Zhao, Y.N. Li, X. clearance predictions: enzyme lability and nonspecific binding, Drug Metab.
G. Tian, Z.L. Yu, X.K. Huo, C.P. Sun, L. Feng, J.N. Cui, X.C. Ma, A highly sensitive Dispos. 32 (2004) 295–304, https://doi.org/10.1124/dmd.32.3.295.
and selective two-photon fluorescent probe for real-time sensing of [104] S. Matsumoto, T. Hirama, T. Matsubara, K. Nagata, Y. Yamazoe, Involvement
cytochrome P450 1A1 in living systems, Mater. Chem. Front. 2 (2018) of CYP2J2 on the intestinal first-pass metabolism of antihistamine drug,
2013–2020, https://doi.org/10.1039/c8qm00372f. astemizole, Drug Metab. Dispos. 30 (2002) 1240–1245, https://doi.org/
[83] Z.R. Dai, L. Feng, Q. Jin, H. Cheng, Y. Li, J. Ning, Y. Yu, G.B. Ge, J.N. Cui, L. Yang, A 10.1124/dmd.30.11.1240.
practical strategy to design and develop an isoform-specific fluorescent probe [105] E.A. Evangelista, R. Kaspera, N.A. Mokadam, J.P. Jones 3rd, R.A. Totah, Activity,
for a target enzyme: CYP1A1 as a case study, Chem. Sci. 8 (2017) 2795–2803, inhibition, and induction of cytochrome P450 2J2 in adult human primary
https://doi.org/10.1039/c6sc03970g. cardiomyocytes, Drug Metab. Dispos. 41 (2013) 2087–2094, https://doi.org/
[84] X. Lv, L. Feng, C.Z. Ai, J. Hou, P. Wang, L.W. Zou, J. Cheng, G.B. Ge, J.N. Cui, L. 10.1124/dmd.113.053389.
Yang, A Practical and High-Affinity fluorescent probe for uridine diphosphate [106] J. Zhang, Y. Chen, Y. Sun, R. Wang, J. Zhang, Z. Jia, Plateau hypoxia attenuates
glucuronosyltransferase 1A1: a good surrogate for bilirubin, J. Med. Chem. 60 the metabolic activity of intestinal flora to enhance the bioavailability of
(2017) 9664–9675, https://doi.org/10.1021/acs.jmedchem.7b01097. nifedipine, Drug Deliv. 25 (2018) 1175–1181, https://doi.org/10.1080/
[85] H. Yu, Y. Xiao, L. Jin, A lysosome-targetable and two-photon fluorescent probe 10717544.2018.1469687.
for monitoring endogenous and exogenous nitric oxide in living cells, J. Am. [107] X. Li, V. Jeso, S. Heyward, G.S. Walker, R. Sharma, G.C. Micalizio, M.D.
Chem. Soc. 134 (2012) 17486–17489, https://doi.org/10.1021/ja308967u. Cameron, Characterization of T-5 N-oxide formation as the first highly
[86] X.L. Liu, X.J. Du, C.G. Dai, Q.H. Song, Ratiometric two-photon fluorescent selective measure of CYP3A5 activity, Drug Metab. Dispos. 42 (2014) 334–
probes for mitochondrial hydrogen sulfide in living cells, J. Org. Chem. 79 342, https://doi.org/10.1124/dmd.113.054726.
(2014) 9481–9489, https://doi.org/10.1021/jo5014838. [108] Y.H. Lu, C.W. Hendrix, N.N. Bumpus, Cytochrome P450 3A5 Plays a Prominent
[87] C. Zhang, A.M. Ren, J.F. Guo, D. Wang, L.Y. Yu, Theoretical design and Role in the Oxidative Metabolism of the Anti-Human Immunodeficiency
investigation of 1,8-naphthalimide-based two-photon fluorescent probes for Virus Drug Maraviroc, Drug Metab. Dispos. 40 (2012) 2221–2230, https://doi.
detecting cytochrome P450 1A with separated fluorescence signal, PCCP 20 org/10.1124/dmd.112.048298.
(2018) 13290–13305, https://doi.org/10.1039/c8cp01754a. [109] Y. Kikuta, E. Kusunose, M. Kusunose, Characterization of human liver
[88] P.H. Keizers, C. de Graaf, F.J. de Kanter, C. Oostenbrink, K.A. Feenstra, J.N. leukotriene B(4) omega-hydroxylase P450 (CYP4F2), J. Biochem. 127 (2000)
Commandeur, N.P. Vermeulen, Metabolic regio- and stereoselectivity of 1047–1052, https://doi.org/10.1093/oxfordjournals.jbchem.a022696.
cytochrome P450 2D6 towards 3,4-methylenedioxy-N-alkylamphetamines: [110] W. Lin, M.Q. Huang, X. Xue, K. Bertelsen, G. Chen, H. Zhao, Z.J. Lin, A. Fourie, J.
in silico predictions and experimental validation, J. Med. Chem. 48 (2005) de Jong, N. Weng, A highly sensitive and selective method for the
6117–6127, https://doi.org/10.1021/jm050338+. determination of leukotriene B4 (LTB4) in ex vivo stimulated human
[89] X. Liu, L. Hu, G. Ge, B. Yang, J. Ning, S. Sun, L. Yang, K. Pors, J. Gu, Quantitative plasma by ultra fast liquid chromatography-tandem mass spectrometry, J
analysis of cytochrome P450 isoforms in human liver microsomes by the Chromatogr B Analyt Technol Biomed Life Sci 925 (2013) 54–62, https://doi.
combination of proteomics and chemical probe-based assay, Proteomics 14 org/10.1016/j.jchromb.2013.02.038.
(2014) 1943–1951, https://doi.org/10.1002/pmic.201400025. [111] K.O. Martin, K. Budai, N.B. Javitt, Cholesterol and 27-hydroxycholesterol 7
[90] F. Weis, H. Hammer, K. Klein, H. Planatscher, U.M. Zanger, A. Noren, C. Wegler, alpha-hydroxylation: evidence for two different enzymes, J. Lipid Res. 34
P. Artursson, T.O. Joos, O. Poetz, Direct quantification of cytochromes P450 (1993) 581–588.
and drug transporters—a rapid, targeted mass spectrometry-based [112] C.H. Yun, T.J. Yin, K. Shatzer, D.G. Burrin, L.W. Cui, Y.F. Tu, M. Hu,
immunoassay panel for tissues and cell culture lysates, Drug Metab. Dispos. Determination of 7 alpha-OH cholesterol by LC-MS/MS: application in
46 (2018) 387–396, https://doi.org/10.1124/dmd.117.078626. assessing the activity of CYP7A1 in cholestatic minipigs, J. Chromatogr. B
[91] J. Ning, T. Liu, P. Dong, W. Wang, G.B. Ge, B. Wang, Z.L. Yu, L. Shi, X. Tian, X.K. 1025 (2016) 76–82, https://doi.org/10.1016/j.jchromb.2016.05.005.
Huo, L. Feng, C. Wang, C.P. Sun, J.N. Cui, T.D. James, X. Ma, Molecular design [113] S. Zhang, S.R. Mada, S. Sharma, M. Torch, D. Mattison, S. Caritis, R.
strategy to construct the near-infrared fluorescent probe for selectively Venkataramanan, Simultaneous quantitation of 17alpha-
sensing human cytochrome P450 2J2, J. Am. Chem. Soc. 141 (2019) 1126– hydroxyprogesterone caproate, 17alpha-hydroxyprogesterone and
1134, https://doi.org/10.1021/jacs.8b12136. progesterone in human plasma using high-performance liquid
[92] D.S. Streetman, J.S. Bertino Jr., A.N. Nafziger, Phenotyping of drug- chromatography-mass spectrometry (HPLC-MS/MS), J. Pharmaceut. Biomed.
metabolizing enzymes in adults: a review of in-vivo cytochrome P450 48 (2008) 1174–1180, https://doi.org/10.1016/j.jpba.2008.08.024.
phenotyping probes, Pharmacogenetics 10 (2000) 187–216, https://doi.org/ [114] P. Concolino, E. Mello, M.C. Patrosso, S. Penco, C. Zuppi, E. Capoluongop.,
10.1097/00008571-200004000-00001. H282N and p.Y191H: 2 novel CYP21A2 mutations in Italian congenital
[93] R.S. Foti, D.A. Rock, L.C. Wienkers, J.L. Wahlstrom, Selection of alternative adrenal hyperplasia patients, Metabolism 61 (2012) 519–524, https://doi.
CYP3A4 probe substrates for clinical drug interaction studies using in vitro org/10.1016/j.metabol.2011.08.008.
data and in vivo simulation, Drug Metab. Dispos. 38 (2010) 981–987, https:// [115] Z. Zhang, D.S. Li, D.E. Blanchard, S.R. Lear, S.K. Erickson, T.A. Spencer, Key
doi.org/10.1124/dmd.110.032094. regulatory oxysterols in liver: analysis as Delta(4)-3-ketone derivatives by
[94] G.B. Ge, J. Ning, L.H. Hu, Z.R. Dai, J. Hou, Y.F. Cao, Z.W. Yu, C.Z. Ai, J.K. Gu, X.C. HPLC and response to physiological perturbations, J. Lipid Res. 42 (2001)
Ma, L. Yang, A highly selective probe for human cytochrome P450 3A4: 649–658.
isoform selectivity, kinetic characterization and its applications, Chem. [116] V.V. Bandaru, N.J. Haughey, Quantitative detection of free 24S-
Commun. (Camb.) 49 (2013) 9779–9781, https://doi.org/10.1039/ hydroxycholesterol, and 27-hydroxycholesterol from human serum, BMC
c3cc45250f. Neurosci. 15 (2014) 137, https://doi.org/10.1186/s12868-014-0137-z.
[95] FDA, Clinical Drug Interaction Studies — Cytochrome P450 Enzyme- and [117] V. Miller, D. Stresser, A. Blanchard, S. Turner, C. Crespi, Fluorometric high-
Transporter-Mediated Drug Interactions Final Guidance. https://www. throughput screening for inhibitors of cytochrome P450, Ann. N. Y. Acad. Sci.
fda.gov/drugs/guidances-drugs/clinical-drug-interaction-studies- 919 (2000) 26–32, https://doi.org/10.1111/j.1749-6632.2000.tb06864.x.
cytochrome-p450-enzyme-and-transporter-mediated-drug-interactions, [118] E. Schneider, D.S. Clark, Cytochrome P450 (CYP) enzymes and the
2020 (accessed 02/28 2020). development of CYP biosensors, Biosens. Bioelectron. 39 (2013) 1–13,
[96] R. Walsky, R. Obach, Validated assays for human cytochrome P450 activities, https://doi.org/10.1016/j.bios.2012.05.043.
Drug Metab. Dispos. 32 (2004) 647–660, https://doi.org/10.1124/ [119] A.T. Wright, J.D. Song, B.F. Cravatt, A suite of activity-based probes for human
dmd.32.6.647. cytochrome P450 enzymes, J. Am. Chem. Soc. 131 (2009) 10692–10700,
[97] N.M. DeVore, B.D. Smith, M.J. Urban, E.E. Scott, Key residues controlling https://doi.org/10.1021/ja9037609.
phenacetin metabolism by human cytochrome P450 2A enzymes, Drug [120] A.T. Wright, B.F. Cravatt, Chemical proteomic probes for profiling cytochrome
Metab. Dispos. 36 (2008) 2582–2590, https://doi.org/10.1124/ p450 activities and drug interactions in vivo, Chem. Biol. 14 (2007) 1043–
dmd.108.023770. 1051, https://doi.org/10.1016/j.chembiol.2007.08.008.
[98] Y.Y. Zhang, Y. Liu, J.W. Zhang, G.B. Ge, L.M. Wang, J. Sun, L. Yang, [121] C. Fehl, C.D. Vogt, R. Yadav, K.L. Li, E.E. Scott, J. Aube, Structure-based design
Characterization of human cytochrome P450 isoforms involved in the of inhibitors with improved selectivity for steroidogenic cytochrome P450
metabolism of 7-epi-paclitaxel, Xenobiotica 39 (2009) 283–292, https://doi. 17A1 over cytochrome P450 21A2, J. Med. Chem. 61 (2018) 4946–4960,
org/10.1080/00498250802714907. https://doi.org/10.1021/acs.jmedchem.8b00419.
[99] U. Yasar, E. Eliasson, C. Forslund-Bergengren, G. Tybring, M. Gadd, F. Sjöqvist, [122] G.A. Potter, S.E. Barrie, M. Jarman, M.G. Rowlands, Novel steroidal inhibitors
M. Dahl, The role of CYP2C9 genotype in the metabolism of diclofenac in vivo of human cytochrome P45017 alpha (17 alpha-hydroxylase-C17,20-lyase):
and in vitro, Eur. J. Clin. Pharmacol. 57 (2001) 729–735, https://doi.org/ potential agents for the treatment of prostatic cancer, J. Med. Chem. 38
10.1007/s00228-001-0376-7. (1995) 2463–2471, https://doi.org/10.1021/jm00013a022.
[100] M.G. Shou, R. Norcross, G. Sandig, P. Lu, Y.H. Li, Y. Lin, Q. Mei, A.D. Rodrigues, [123] K. Fizazi, H.I. Scher, A. Molina, C.J. Logothetis, K.N. Chi, R.J. Jones, J.N. Staffurth,
T.H. Rushmore, Substrate specificity and kinetic properties of seven S. North, N.J. Vogelzang, F. Saad, P. Mainwaring, S. Harland, O.B. Goodman, C.
heterologously expressed dog cytochromes P450, Drug Metab. Dispos. 31 N. Sternberg, J.H. Li, T. Kheoh, C.M. Haqq, J.S. de Bono, Abiraterone acetate for
(2003) 1161–1169, https://doi.org/10.1124/dmd.31.9.1161. treatment of metastatic castration-resistant prostate cancer: final overall
[101] V. Kumar, D.A. Rock, C.J. Warren, T.S. Tracy, J.L. Wahlstrom, Enzyme source survival analysis of the COU-AA-301 randomised, double-blind, placebo-
effects on CYP2C9 kinetics and inhibition, Drug Metab. Dispos. 34 (2006) controlled phase 3 study, Lancet Oncol. 13 (2012) 983–992, https://doi.org/
1903–1908, https://doi.org/10.1124/dmd.106.010249. 10.1016/s1470-2045(12)70379-0.

22
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

[124] P. Schonknecht, D. Lutjohann, J. Pantel, H. Bardenheuer, T. Hartmann, K. von analysis, biology and medicine, Chem. Soc. Rev. 45 (2016) 6048–6077,
Bergmann, K. Beyreuther, J. Schroder, Cerebrospinal fluid 24S- https://doi.org/10.1039/C6CS00296J.
hydroxycholesterol is increased in patients with Alzheimer’s disease [146] K. Gomi, N. Kajiyama, Oxyluciferin, a luminescence product of firefly
compared to healthy controls, Neurosci. Lett. 324 (2002) 83–85, https://doi. luciferase, is enzymatically regenerated into luciferin, J. Biol. Chem. 276
org/10.1016/s0304-3940(02)00164-7. (2001) 36508–36513, https://doi.org/10.1074/jbc.M105528200.
[125] F. Djelti, J. Braudeau, E. Hudry, M. Dhenain, J. Varin, I. Bieche, C. Marquer, F. [147] E.V. Vetrova, N.S. Kudryasheva, K.H. Cheng, Effect of quinone on the
Chali, S. Ayciriex, N. Auzeil, S. Alves, D. Langui, M.C. Potier, O. Laprevote, M. fluorescence decay dynamics of endogenous flavin bound to bacterial
Vidaud, C. Duyckaerts, R. Miles, P. Aubourg, N. Cartier, CYP46A1 inhibition, luciferase, Biophys. Chem. 141 (2009) 59–65, https://doi.org/10.1016/j.
brain cholesterol accumulation and neurodegeneration pave the way for bpc.2008.12.012.
Alzheimer’s disease, Brain 138 (2015) 2383–2398, https://doi.org/10.1093/ [148] E. Vetrova, N. Kudryasheva, V. Kratasyuk, Redox compounds influence on the
brain/awv166. NAD (P) H: FMN–oxidoreductase–luciferase bioluminescent system,
[126] S. Sudsakorn, J. Skell, D.A. Williams, T.J. O’Shea, H.L. Liu, Evaluation of 3-O- Photochem. Photobiol. Sci. 6 (2007) 35–40, https://doi.org/10.1039/
methylfluorescein as a selective fluorometric substrate for CYP2C19 in B608152E.
human liver microsomes, Drug Metab. Dispos. 35 (2007) 841–847, https:// [149] Promega, P450-Glo assay technical bulletin. . http://www.promega.com/
doi.org/10.1124/dmd.106014472. ~/media/Files/Resources/Protocols/Technical%20Bulletins/101/P450%20Glo%
[127] A.B. Renwick, D. Surry, R.J. Price, B.G. Lake, D.C. Evans, Metabolism of 7- 20Assays%20Protocol.pdf, 2012 (accessed 19 Junly 2020).
benzyloxy-4-trifluoromethyl-coumarin by human hepatic cytochrome P450 [150] J.J. Cali, D. Ma, M. Sobol, D.J. Simpson, S. Frackman, T.D. Good, W.J. Daily, D.
isoforms, Xenobiotica 30 (2000) 955–969, https://doi.org/10.1080/ Liu, Luminogenic cytochrome P450 assays, Expert Opin. Drug Metab. Toxicol.
00498250050200113. 2 (2006) 629–645, https://doi.org/10.1517/17425255.2.4.629.
[128] A.B. Renwick, D.F.V. Lewis, S. Fulford, D. Surry, B. Williams, P.D. Worboys, X. [151] J. J. Cali,D. Ma, A Rapid Cell Based Bioluminescent CYP1A1 Aryl Hydrocarbon
Cai, R.W. Wang, R.J. Price, B.G. Lake, D.C. Evans, Metabolism of 2,5-bis Receptor Activation Assay. https://www.promega.com.cn/
(trifluoromethyl)-7-benzyloxy-4-trifluoromethylcoumarin by human hepatic resources/scientific_posters/posters/a-rapid-cell-based-bioluminscent-
CYP isoforms: evidence for selectivity towards CYP3A4, Xenobiotica 31 cyp1a1-aryl-hydrocarbon-receptor-assay-poster/, 2012 (accessed 19 Junly
(2001) 187–204, https://doi.org/10.1080/00498250110043526. 2020).
[129] X. Zhang, Y. Zhou, X. Gu, Y. Cheng, M. Hong, L. Yan, F. Ma, Z. Qi, Synthesis of a [152] D. Ma, M. Sobol, C. C. Woodroofe,J. J. Cali, Cytochrome P450 2J2 enzyme assay
selective ratiometric fluorescent probe based on Naphthalimide and its using a novel bioluminescent probe substrate. . https://www.promega.com.
application in human cytochrome P450 1A, Talanta 186 (2018) 413–420, cn/resources/pubhub/enotes/cytochrome-p450-2j2-enzyme-assay-using-a-
https://doi.org/10.1016/j.talanta.2018.04.079. novel-bioluminescent-probe-substrate/, 2008 (accessed 19 Junly 2020).
[130] H. Ji, X. Zhang, Y. Dai, T. Xue, S. Misal, Z. Qi, A highly selective ratiometric [153] M. Sobol, D. Ma,J. J. Cali, Selective cytochrome p450 3A7 enzyme assay using
fluorescent probe based on naphthalimide for detection and imaging of a novel bioluminescent probe substrate. https://www.promega.com.cn/
CYP1A1 in living cells and zebrafish, Analyst 144 (2019) 7390–7397, https:// resources/pubhub/enotes/selective-cytochrome-p450-3a7-enzyme-assay-
doi.org/10.1039/c9an01767d. using-a-novel-bioluminescent-probe-substrate/, 2007 (accessed 19 Junly
[131] T. Xue, Y. Dai, X. Zhang, Y. Cheng, X. Gu, H. Ji, S. Misal, Z. Qi, Ultrasensitive 2020).
near-infrared fluorescent probe with large stokes shift for real-time tracing of [154] M. Sobol, D. Ma,J. J. Cali, Selective Cytochrome P450 4A11 Enzyme Assay
CYP1A1 in living cells and zebrafish model, Sens. Actuat. B-Chem. 293 (2019) Using a Novel Bioluminescent Probe Substrate. http://www.promega.com/
265–272, https://doi.org/10.1016/j.snb.2019.04.147. resources/articles/pubhub/enotes/selective-cytochrome-p450-4a11-
[132] T. Shimada, E.M. Gillam, T.R. Sutter, P.T. Strickland, F.P. Guengerich, H. enzyme-assay-using-a-novel-bioluminescent-probe-substrate/, 2012
Yamazaki, Oxidation of xenobiotics by recombinant human cytochrome P450 (accessed 16 January 2012).
1B1, Drug Metab. Dispos. 25 (1997) 617–622. [155] M. Sobol, D. Ma, C. C. Woodroofe,J. J. Cali, Cytochrome P450 4F2 and 4F3B
[133] Q. Jin, Y. Ma, L. Feng, P. Wang, R. He, L. Yang, G.B. Ge, Sensing cytochrome Enzyme Assays Using a Novel Bioluminescent Probe Substrate. https://www.
P450 1A1 activity by a resorufin-based isoform-specific fluorescent probe, promega.com.cn/Resources/PubHub/eNotes/Cytochrome%20P450%204F2%
Chin. Chem. Lett. (2020), https://doi.org/10.1016/j.cclet.2020.05.038. 20and%204F3B%20Enzyme%20Assays%20Using%20a%20Novel%
[134] A. Ghosal, N. Hapangama, Y. Yuan, X. Lu, D. Horne, J.E. Patrick, S. Zbaida, Rapid 20Bioluminescent%20Probe%20Substrate/, 2008 (accessed 19 Junly 2020).
determination of enzyme activities of recombinant human cytochromes [156] D. Ma, M. Sobol, C. C. Woodroofe,J. J. Cali, Cytochrome P450 4F12 Enzyme
P450, human liver microsomes and hepatocytes, Biopharm. Drug Dispos. 24 Assay Using a Novel Bioluminescent Probe Substrate. https://www.
(2003) 375–384, https://doi.org/10.1002/bdd.374. promega.com.cn/Resources/PubHub/eNotes/Cytochrome%20P450%204F12%
[135] L. Makings, G. Zlokarnik, Optical molecular sensors for cytochrome P450 20Enzyme%20Assay%20Using%20a%20Novel%20Bioluminescent%20Probe%
activity. https://patents.google.com/patent/US6514687B1/en, 2003 (accessed 20Substrate/, 2007 (accessed 27 August 2020).
19 Junly 2020). [157] H.S. Blix, K.K. Viktil, T.A. Moger, A. Reikvam, Drugs with narrow therapeutic
[136] Y. Li, N.Y. Li, E.M. Sellers, Comparison of CYP2A6 catalytic activity on index as indicators in the risk management of hospitalised patients, Pharm.
coumarin 7-hydroxylation in human and monkey liver microsomes, Eur. J. Pract. 8 (2010) 50–55, https://doi.org/10.4321/s1886-36552010000100006.
Drug Metab. 22 (1997) 295–304, https://doi.org/10.1007/BF03190960. [158] M.E. de Jonge, A.D. Huitema, J.H. Schellens, S. Rodenhuis, J.H. Beijnen,
[137] M. Spatzenegger, H. Liu, Q. Wang, A. Debarber, D. Koop, J. Halpert, Analysis of Population pharmacokinetics of orally administered paclitaxel formulated in
differential substrate selectivities of CYP2B6 and CYP2E1 by site-directed Cremophor EL, Br. J. Clin. Pharmacol. 59 (2005) 325–334, https://doi.org/
mutagenesis and molecular modeling, J. Pharmacol. Exp. Ther. 304 (2002) 10.1111/j.1365-2125.2004.02325.x.
477–487, https://doi.org/10.1124/jpet.102.043323. [159] H. Zhang, N. Gao, X. Tian, T. Liu, Y. Fang, J. Zhou, Q. Wen, B. Xu, B. Qi, J. Gao, H.
[138] J. Venhorst, A. ter Laak, J. Commandeur, Y. Funae, T. Hiroi, N. Vermeulen, Li, L. Jia, H. Qiao, Content and activity of human liver microsomal protein and
Homology modeling of rat and human cytochrome P450 2D (CYP2D) prediction of individual hepatic clearance in vivo, Sci. Rep. 5 (2015) 17671,
isoforms and computational rationalization of experimental ligand-binding https://doi.org/10.1038/srep17671.
specificities, J. Med. Chem. 46 (2003) 74–86, https://doi.org/10.1021/ [160] D.N. Juurlink, Drug-drug interactions among elderly patients hospitalized for
jm0209578. drug toxicity, JAMA 289 (2003) 1652–1658, https://doi.org/
[139] B.D. Marks, R.W. Smith, H.A. Braun, T.A. Goossens, M. Christenson, M.S. Ozers, 10.1001/jama.289.13.1652.
C.S. Lebakken, O.V. Trubetskoy, A high throughput screening assay to screen [161] C. Zhan, R. Correa-de-Araujo, A.S. Bierman, J. Sangl, M.R. Miller, S.W. Wickizer,
for CYP2E1 metabolism and inhibition using a fluorogenic vivid p450 D. Stryer, Suboptimal prescribing in elderly outpatients: potentially harmful
substrate, Assay Drug Dev. Technol. 1 (2002) 73–81, https://doi.org/ drug-drug and drug-disease combinations, J. Am. Geriatr. Soc. 53 (2005) 262–
10.1089/154065802761001329. 267, https://doi.org/10.1111/j.1532-5415.2005.53112.x.
[140] N. Ning, W. Wang, G.B. Ge, P. Chu, F. Long, Y. Yang, Y. Peng, L. Feng, X. Ma, T.D. [162] S. Fowler, H. Zhang, In vitro evaluation of reversible and irreversible
James, Targeted enzyme activated two-photon fluorescent probes: a case cytochrome P450 inhibition: current status on methodologies and their
study of CYP3A4 using a two-dimensional design strategy, Angew. Chem. Int. utility for predicting drug-drug interactions, AAPS J. 10 (2008) 410–424,
Ed. Engl. 58 (2019) 9959–9963, https://doi.org/10.1002/anie.201903683. https://doi.org/10.1208/s12248-008-9042-7.
[141] R. Foti, L. Wienkers, J. Wahlstrom, Application of cytochrome P450 drug [163] L.H. Cohen, M.J. Remley, D. Raunig, A.D. Vaz, In vitro drug interactions of
interaction screening in drug discovery, Comb. Chem. High T Scr. 13 (2010) cytochrome p450: an evaluation of fluorogenic to conventional substrates,
145–158, https://doi.org/10.2174/138620710790596718. Drug Metab. Dispos. 31 (2003) 1005–1015, https://doi.org/10.1124/
[142] J.J. Cali, D. Ma, M.G. Wood, P.L. Meisenheimer, D.H. Klaubert, Bioluminescent dmd.31.8.1005.
assays for ADME evaluation: dialing in CYP selectivity with luminogenic [164] L. Bell, S. Bickford, P.H. Nguyen, J. Wang, T. He, B. Zhang, Y. Friche, A. Zimmerlin,
substrates, Expert Opin. Drug Metab. Toxicol. 8 (2012) 1115–1130, https:// L. Urban, D. Bojanic, Evaluation of fluorescence- and mass spectrometry-based
doi.org/10.1517/17425255.2012.695345. CYP inhibition assays for use in drug discovery, J. Biomol. Screen. 13 (2008)
[143] J. Li, L. Chen, L. Du, M. Li, Cage the firefly luciferin!–a strategy for developing 343–353, https://doi.org/10.1177/1087057108317480.
bioluminescent probes, Chem. Soc. Rev. 42 (2013) 662–676, https://doi.org/ [165] Y.F. Ueng, W.C. Jan, L.C. Lin, T.L. Chen, F.P. Guengerich, C.F. Chen, The alkaloid
10.1039/C2CS35249D. rutaecarpine is a selective inhibitor of cytochrome P450 1A in mouse and
[144] T.A. Su, K.J. Bruemmer, C.J. Chang, Caged luciferins for bioluminescent human liver microsomes, Drug Metab. Dispos. 30 (2002) 349–353, https://
activity-based sensing, Curr. Opin. Biotechnol. 60 (2019) 198–204, https:// doi.org/10.1124/dmd.30.3.349.
doi.org/10.1016/j.copbio.2019.05.002. [166] A. Galetin, K. Ito, D. Hallifax, J.B. Houston, CYP3A4 substrate selection and
[145] Z.M. Kaskova, A.S. Tsarkova, I.V. Yampolsky, 1001 lights: luciferins, substitution in the prediction of potential drug-drug interactions, J.
luciferases, their mechanisms of action and applications in chemical Pharmacol. Exp. Ther. 314 (2005) 180–190, https://doi.org/10.1124/
jpet.104.082826.

23
J. Wu et al. Coordination Chemistry Reviews 427 (2021) 213600

[167] L. Yu, Y.M. Qiao, L.X. Miao, Y.Q. He, Y. Zhou, Recent progress in fluorescent [171] S.M. Sukumaran, B. Potsaid, M.Y. Lee, D.S. Clark, J.S. Dordick, Development of
and colorimetric sensors for the detection of ions and biomolecules, Chin. a fluorescence-based, ultra high-throughput screening platform for
Chem. Lett. 29 (2018) 1545–1559, https://doi.org/10.1016/j. nanoliter-scale cytochrome p450 microarrays, J. Biomol. Screen. 14 (2009)
cclet.2018.09.005. 668–678, https://doi.org/10.1177/1087057109336592.
[168] J. Zhang, X. Chai, X.P. He, H.J. Kim, J. Yoon, H. Tian, Fluorogenic probes for [172] J. Hao, J. Long, B. Li, X. Li, S. Zhang, F. Yang, X. Zeng, Z. Yang, W.K. Pang, Z. Guo,
disease-relevant enzymes, Chem. Soc. Rev. 48 (2019) 683–722, https://doi. Toward high-performance hybrid zn-based batteries via deeply
org/10.1039/c7cs00907k. understanding their mechanism and using electrolyte additive, Adv. Funct.
[169] H.W. Liu, L. Chen, C. Xu, Z. Li, H. Zhang, X.B. Zhang, W. Tan, Recent progresses Mater. 29 (2019) 1903605, https://doi.org/10.1002/adfm.201903605.
in small-molecule enzymatic fluorescent probes for cancer imaging, Chem. [173] M. Gao, B.Z. Tang, AIE-based cancer theranostics, Coord. Chem. Rev. 402
Soc. Rev. 47 (2018) 7140–7180, https://doi.org/10.1039/c7cs00862g. (2020), https://doi.org/10.1016/j.ccr.2019.213076 213076.
[170] W. Chyan, R.T. Raines, Enzyme-activated fluorogenic probes for live-cell and [174] A.D. Halleran, A. Swaminathan, R.M. Murray, Single day construction of
in vivo imaging, ACS Chem. Biol. 13 (2018) 1810–1823, https://doi.org/ multigene circuits with 3G assembly, ACS Synth. Biol. 7 (2018) 1477–1480,
10.1021/acschembio.8b00371. https://doi.org/10.1021/acssynbio.8b00060.

24

You might also like