You are on page 1of 9

Toxicology and Applied Pharmacology 424 (2021) 115595

Contents lists available at ScienceDirect

Toxicology and Applied Pharmacology


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

Comparison of the drug-drug interactions potential of ibrutinib and


acalabrutinib via inhibition of UDP-glucuronosyltransferase
Xiaoyu Wang a, Zhe Wang a, Xiaoyu Fan a, Mingrui Yan a, Lili Jiang a, Yangliu Xia a, Jun Cao b, **,
Yong Liu a, *
a
School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
b
Department of Occupational and Environmental Health, Dalian Medical University, Dalian 116044, China

A R T I C L E I N F O A B S T R A C T

Keywords: Ibrutinib and acalabrutinib are two Bruton's tyrosine kinase (BTK) inhibitors which have gained Food and Drug
Ibrutinib Administration (FDA) approval for the treatment of various B cell malignancies. Herein, we investigated the
Acalabrutinib effects of the two drugs on UDP-glucuronosyltransferase (UGT) activities to evaluate their potential risk for drug-
UDP-glucuronosyltransferase
drug interactions (DDIs) via UGT inhibition. Our data indicated that ibrutinib exerted broad inhibition on most of
Drug-drug interaction
UGTs, including a potent competitive inhibition against UGT1A1 with a Ki value of 0.90 ± 0.03 μM, a
noncompetitive inhibition against UGT1A3 and UGT1A7 with Ki values of 0.88 ± 0.03 μM and 2.52 ± 0.23 μM,
respectively, while acalabrutinib only exhibited weak UGT inhibition towards all tested UGT isoforms. DDI risk
prediction suggested that the inhibition against UGT1A1 and UGT1A3 by ibrutinib might bring a potential DDIs
risk, while acalabrutinib was unlikely to trigger clinically significant UGT-mediated DDIs due to its weak effects.
Our study raises an alarm bell about potential DDI risk associated with ibrutinib, however, the extrapolation
from in vitro data to in vivo drug interactions should be taken with caution, and additional systemic study is
needed.

1. Introduction 29- and 24-fold, respectively, so that lead to potential adverse events at
therapeutic dose (de Zwart et al., 2016). Therefore, DDIs based on drug
Ibrutinib and acalabrutinib are two oral small-molecule Bruton's metabolizing enzyme remain a problem of considerable clinical
tyrosine kinase (BTK) inhibitors approved for the treatment of adult significance.
patients with chronic lymphocytic leukemia (CLL) and mantle cell In addition to giving focus on CYP enzyme driving DDIs, indeed,
lymphoma (MCL) by blocking B-cell receptor signaling pathway to UDP-glycosyltransferase (UGT) inhibition could also be implicated in
induce tumor cell death (Advani et al., 2013; Byrd et al., 2016). Phase III hepatotoxicity and associated with clinically DDIs (Meech et al., 2019).
trials of both two drugs are currently underway worldwide to evaluate UGT is an important class of phase II metabolizing enzyme superfamily
their treatment effects and give more combinations of drugs, meaning which plays a critical role in the elimination of numerous exogenous
that drug-drug interactions (DDIs) may first be encountered during chemicals and endogenous substances and in the pathological proced­
clinical development (Cameron and Sanford, 2014; Markham and ures of some diseases (Zhou et al., 2013). Particularly, UGT1A1, for
Dhillon, 2018). Previous study has suggested that co-administration which participates in bilirubin glucuronidation and has a high level of
acalabrutinib with itraconazole, a cytochrome P450 (CYP) 3A inhibi­ polymorphism, is a major concern. Several tyrosine kinase inhibitors
tor, will be expected to increase its plasma concentrations which may (TKIs) have been observed as potent UGT inhibitors implying a risk for
result in toxicity (Zhou et al., 2019). Co-treatment with ketoconazole (a adverse DDIs in clinic. For example, concomitant administration of
known CYP3A inhibitor) will increase the Cmax and AUC of ibrutinib by gefitinib (700 mg/day) and irinotecan may increase the SN-38 AUC by

* Correspondence to: Y. Liu, School of Life and Pharmaceutical Sciences, Dalian University of Technology, 2 Dagong Road, Liaodongwan New District, Panjin
124221, China.
** Correspondence to: J. Cao, Department of Occupational and Environmental Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044,
China.
E-mail addresses: caojunly@163.com (J. Cao), yliu@dlut.edu.cn (Y. Liu).

https://doi.org/10.1016/j.taap.2021.115595
Received 31 January 2021; Received in revised form 18 May 2021; Accepted 21 May 2021
Available online 24 May 2021
0041-008X/© 2021 Elsevier Inc. All rights reserved.
X. Wang et al. Toxicology and Applied Pharmacology 424 (2021) 115595

was consisted of solvent A (0.1% formic acid in water) and solvent B


(0.1% formic acid in acetonitrile) with a flow rate of 0.3 mL/min. The
gradient was used as follows: 5% (v/v) B at 0–2 min, 5%–50% (v/v) B at
2–2.4 min, 50% (v/v) B at 2.4–3 min, 50%–5% (v/v) B at 3–4 min, 5%
(v/v) B at 4–5 min. The MRM m/z transitions monitored were 353.3/
177.2 for 4-MUG and 163.1/107.1 for internal standard in positive ion
mode which were identified to be optimal for quantitative analysis.
Calibration standard of 4-MUG was prepared at concentrations of
0.125–15 μM and the accuracy was within 10%.
In addition, trifluoperazine (TFP) was used as the selective substrate
in the UGT1A4 inhibition studies (Uchaipichat et al., 2006) and the
supernatant of reactions were separated by HPLC system (Waters, Mil­
ford, MA) with a Tnature C18 column (250 × 4.6 mm, 5 μm). Mobile
phase was containing 0.5% formic acid in water (A) and acetonitrile (B).
A gradient program was as follows: 29–48% (v/v) B at 0–7 min, 48%–
29% (v/v) B at 7.0–7.5 min, 29% (v/v) B at 7.5–9 min. The flow rate was
maintained at 1 mL/min and analytes were quantified with UV detection
at 254 nm. Umbelliferone (18 μM) was used as the internal standard.
Fig. 1. Chemical structures of ibrutinib and acalabrutinib. Calibration standard of TFP was prepared at concentrations of 0.025–10
μM since no TFP-G standards were available (Seo et al., 2014).
49% via UGT1A1 inhibition (Li et al., 2015). Regorafenib and sorafenib
can potently inhibit UGT1A1 activity, which contributed significantly to 2.3. Determination of IC50 and Ki
the hyperbilirubinemia observed in patients (Miners et al., 2017).
However, the issue of prediction of DDIs risk by acalabrutinib and The same incubation condition as for the preliminary inhibition
ibrutinib via UGT inhibition is far from exhaustive. study was used for each UGT isoform. The relative formation rate of 4-
Here, we aimed to evaluate the inhibitory effects of two BTK in­ MUG or TFP-G in the presence vs. absence of inhibitors was estimated.
hibitors ibrutinib and acalabrutinib towards the activity of 12 human For UGT isoforms whose activity is inhibited by >50% by individual
recombinant UGT isoforms and identify the potential DDIs in vivo based BTK inhibitors, half maximal inhibitory concentration (IC50) values for
on in vitro-in vivo extrapolation (IVIVE). These results provide a new TKIs were determined. Subsequent inhibition kinetic experiments were
sight for explanation of the toxicity of BTK-TKIs and might be helpful to performed for the lower IC50 values groups. The most appropriate in­
develop new clinical combination therapies. hibition type and Ki value were ascertained from comparison of
goodness-of-fit parameters using equations for competitive inhibition
2. Materials and methods (eq. 1), noncompetitive inhibition (eq. 2), uncompetitive (eq. 3), or
mixed inhibition (eq. 4).
2.1. Chemical reagents
v = (Vmax S)/(Km (1 + I/Ki ) + S ) (1)
Ibrutinib, acalabrutinib, and trifluoperazine (TFP) were obtained v = (Vmax S)/(Km + S)(1 + I/Ki ) (2)
from Selleck Chemicals (Houston, TX, USA), umbelliferone, 4-methyl­
umbelliferone (4-MU), 4-methylumbelliferone-β-D-glucuronide (4- v = (Vmax S)/(Km + (1 + I/Ki )S ) (3)
MUG), and uridine 5′ -diphosphoglucuronic acid trisodium salt (UDPGA)
were obtained from Sigma-Aldrich (St. Louis, MO, USA). Tris-HCl, v = (Vmax S)/[Km (1 + I/Ki ) + S(1 + I/αK i ) ] (4)
MgCl2 were purchased from Aladdin (Shanghai, China). All other
chemicals used were of HPLC grade commercially available. where v is the velocity of the reaction, Vmax is the maximum velocity, S
Recombinant UGT supersomes (UGT1A1, UGT1A3, UGT1A4, and I are the substrate and inhibitor concentrations, respectively. Km is
UGT1A6, UGT1A7, UGT1A8, UGT1A9, UGT1A10, UGT2B4, UGT2B7, Michaelis constant (the substrate concentration at half of the Vmax of the
UGT2B15, and UGT2B17) were purchased from Corning Gentest reaction), Ki is inhibition constant. α reflects the effect of inhibitor on the
(Tewksbury, MA, USA). affinity of the enzyme for its substrate.

2.4. Predicted concentrations of TKIs at UGT active site


2.2. Inhibition of recombinant human UGT enzymes by BTK inhibitors
For IVIVE, the inhibitor concentrations ([I]) after oral administration
The incubation systems were conducted in a final volume of 200 μL
can be calculated using the following equation: average systemic plasma
in 50 mM Tris-HCl buffer (pH 7.4) with 10 mM MgCl2, 5 mM UDPGA,
concentration ([I]av) (eq.5), maximum systemic plasma concentration
UGTs and 4-MU. Ibrutinib and acalabrutinib (structures are shown in
([I]max) (eq.6), and maximum hepatic input concentration ([I]in) (eq.7)
Fig. 1) were dissolved in DMSO. The linear initial velocity enzymatic
to evaluate the highest risk (Ito et al., 2004).
conditions were used as described previously (Wang et al., 2020). Re­
/
actions were pre-incubated for 5 min at 37 ◦ C and initiated by addition [I]av = (D/τ) (CL/F) (5)
of 20 μL UDPGA. Enzymatic reactions were terminated by adding equal
( )/
volume of ice-cold acetonitrile containing 1 μM umbelliferone as an [I]max = [I]av κτ (1 − exp( − κτ) ) (6)
internal standard. Samples were centrifuged at 20,050g for 20 min at
/
4 ◦ C to remove the protein. [I]in = [I]av + ka Fa D Qh (7)
Detection of 4-MUG was performed by ultra-performance liquid
chromatography-tandem mass spectrometry (UPLC-MS/MS) system where D and τ are oral dose and dosing interval, respectively, CL/F is
consisting of ACQUITY UPLC module (Waters, Milford, MA) and Qtrap apparent (oral) drug clearance, k is the elimination rate constant which
6500 (SCIEX, San Jose, CA, USA). Aliquot 2 μL of each sample was is expressed as k = 0.693/t1/2 (t1/2, half time), ka is the absorption rate
injected on a BEH C18 column (50 × 2.1 mm, 1.7 μm). The mobile phase constant, Fa is the fraction absorbed from gut into the portal vein, and Qh

2
X. Wang et al. Toxicology and Applied Pharmacology 424 (2021) 115595

Fig. 2. The inhibition of ibrutinib and acalabrutinib towards human recombinant UGT isoforms. TFP was used as substrate for the inhibition studies with UGT1A4, 4-
MU was used as the substrate for all other UGT isoforms. Each bar represents the mean ± standard error of duplicate experiments.

is the hepatic blood flow rate, assumed to be 1450 mL/min (Davies and inhibited enzyme which ranged of 0.1–1. Here, [I]in was used to predict
Morris, 1993). The parameters of CL/F, t1/2, ka and Fa obtained from the highest potential risk from the perspective of pharmacokinetics
previous publications for ibrutinib (Pharmacyclics Inc, 2013; de Jong theory (Oda et al., 2015). As for UGT1A7, only expressed in extrahepatic
et al., 2015; Marostica et al., 2015) and acalabrutinib (Markham and tissues (Strassburg et al., 1997), [I]max was used to represent the in­
Dhillon, 2018; Zhou et al., 2019) were shown in Table 2. hibitor concentrations at UGT enzyme active site.
In the present analysis, the DDIs potential for ibrutinib and acalab­
rutinib were evaluated by calculating the AUC ratios. The FDA draft
2.5. Prediction of in vivo drug–drug interactions magnitude
guidance on drug interaction studies proposes that a 25% increase in the
AUC of victim drugs, which is normally considered to be bioequivalent,
The potential DDIs risk arising from inhibition of a drug metabolizing
should not merit full consideration for clinically relevant DDIs (Yu and
enzyme can be determined from the Ki value generated in vitro using the
Tweedie, 2013). As such, the AUC ratio cut-off value of 1.25 was used
following equations (eq.8) for drugs with negligible renal clearance
according to the FDA Guidance (USFDA, 2020) and interactions with
(Miners et al., 2010).
AUC ratio changes more than 5-fold, 2-to 5-fold, or 1.25-to 2-fold were
AUCi /AUC = 1/(fm /(1 + [I]/Ki ) + (1 − fm ) (8) considered strong, moderate, or weak inhibition drug interactions,
respectively (Yu et al., 2019).
where the victim drug is metabolized by a single enzyme or has a high
hepatic clearance (fm assumed as 1), eq. (8) simplifies to 3. Results
AUCi /AUC = 1 + [I]/Ki (9)
3.1. Inhibition of UGT activities by ibrutinib and acalabrutinib
where AUCi/AUC is the ratio of areas under the plasma concentration
against time of the victim drug in the presence and absence of the in­ The inhibitory effects of ibrutinib and acalabrutinib on UGT activ­
hibitor and fm is the fraction of UGT substrates metabolized by the ities were shown in Fig. 2. Ibrutinib (100 μM) inhibited almost all the

3
X. Wang et al. Toxicology and Applied Pharmacology 424 (2021) 115595

Fig. 3. Concentration-dependent inhibition of ibrutinib in recombinant human UGTs.

Fig. 4. Concentration-dependent inhibition of acalabrutinib in recombinant human UGT1A1, UGT1A3, UGT1A7, and UGT2B15.

UGTs in reducing the substrate glucuronidation more than 70%. The


Table 1
inhibition of acalabrutinib was observed against UGT1A1, 1A3, 1A7,
The IC50 of ibrutinib, acalabrutinib for the inhibition against UGT activities.
and 2B15 with corresponding residual activities were 48.68%, 28.57%,
UGTs Ibrutinib (μM) Acalabrutinib (μM) 28.13%, and 11.8%, respectively. To further investigate the inhibitory
UGT1A1 0.70 ± 0.05 17.89 ± 0.72 potentials of both two BTK-inhibitors, IC50 values were further deter­
UGT1A3 0.79 ± 0.05 8.54 ± 0.75 mined and corresponding inhibition curves of ibrutinib and acalabruti­
UGT1A4 20.26 ± 0.41 > 100 nib were shown in Figs. 3 and 4, respectively. As shown in Table 1,
UGT1A6 24.92 ± 0.93 > 100
UGT1A7 1.05 ± 0.01 27.62 ± 2.08
ibrutinib potently inhibited the enzyme activities of UGT1A1 (IC50 0.70
UGT1A8 16.19 ± 0.71 > 100 ± 0.05 μM), UGT1A3 (IC50 0.79 ± 0.05 μM), and UGT1A7 (IC50 1.05 ±
UGT1A9 11.69 ± 0.48 > 100 0.01 μM) (Fig. 3), and moderately inhibited of UGT1A9 (IC50 11.69 ±
UGT1A10 13.65 ± 0.11 > 100 0.48 μM), and UGT2B7 (6.94 ± 0.08 μM), weakly inhibited of other UGT
UGT2B4 80.84 ± 4.07 > 100
isoforms which IC50 values ranged from 13.65 to 80.84 μM. Meanwhile,
UGT2B7 6.94 ± 0.08 > 100
UGT2B15 21.84 ± 0.53 19.84 ± 0.13 acalabrutinib demonstrated moderate or slight inhibition of UGT1A1,
UGT2B17 26.08 ± 4.56 > 100 1A3, 1A7 and UGT2B15 activities with IC50 values ranged from 8.54 to
27.62 μM, since these values are more than an order of magnitude higher
Data represent the mean ± standard error of duplicates experiments.
than the reported Cmax (0.69 μM) (Markham and Dhillon, 2018), and
hence Ki was not determined but estimated based on the equation Ki =
1/2 × IC50, assuming competitive inhibition (Haupt et al., 2015).

4
X. Wang et al. Toxicology and Applied Pharmacology 424 (2021) 115595

Fig. 5. Representative Lineweaver-Burk plots and Dixon plots for the inhibition of.
4-MU glucuronidation by ibrutinib in human recombinant UGT1A1(A and B), UGT1A3(C and D), UGT1A7(E and F). All data points shown represent the mean ±
standard error of duplicate measurements.

5
X. Wang et al. Toxicology and Applied Pharmacology 424 (2021) 115595

Table 2 with Ki values of 0.88 ± 0.03 and 2.52 ± 0.23 μM, respectively.
Calculation of possible concentrations of ibrutinib and acalabrutinib.
Drugs Ibrutinib Acalabrutinib
3.3. Quantitative prediction of DDIs potential risk
Dose (mg) 560 200
Dosing Interval (hr) 24 12
Cmax (μM) 0.29 0.69 The calculated concentrations of [I]in and [I]max of ibrutinib and
Absorption Rate Constant (ka, h− 1) 0.46 1.65 acalabrutinib were shown in Table 2. Then the prediction results for
Plasma Unbound Fraction (fu,p) 2.70% 2.50% each TKI were calculated by eq. (8) and reported as isolines plot in
t1/2 (h) 4 1.57
Figs. 6 and 7, respectively. Values in the line represented the AUC ratio
CL/F (L/h) 1060 159
Oral bioavailability (F) 67% 25%
(AUCi/AUC) and an isoline was joining equal values of AUC ratio
Calculated concentrations Iav (μM) 0.05 0.23 generated from the corresponding oral dose and fm.
Imax (μM) 0.21 1.20 As shown in Fig. 6A, when the oral dose of ibrutinib is 560 mg/day,
Iin (μM) 2.05 1.17 co-administered substrate is mainly metabolized by UGT1A1 (fm > 0.8),
the predicted AUC ratio is approximately 2.40, which means that an
3.2. Inhibition kinetics in recombinant human UGTs by ibrutinib increase in the AUC of substrate of more than 140% might be observed
when substrates are co-administrated with ibrutinib. Also, when the
Kinetic experiments were performed to further characterize the in­ dose is more than 560 mg/day and fm is 1, the predicted AUC ratio is
hibition of UGT1A1, UGT1A3 and UGT1A7 activities by ibrutinib 3.05 (Fig. 6B), which means that the AUC of substrate totally metabo­
(Fig. 5). Nonlinear regression analysis indicated that 4-MU glucur­ lized by UGT1A3 will increase more than 2-fold in vivo. Specifically,
onidation in UGT1A1 was greatly inhibited by ibrutinib in a competitive when the dose of ibrutinib was 280 mg/day and fm was more than 0.4,
manner with a Ki value of 0.90 ± 0.03 μM, whereas inhibition of the AUC ratio of victim drug cleared by UGT1A1 or UGT1A3 were
UGT1A3 and UGT1A7 followed noncompetitive inhibition mechanism increased by at least 1.34, which indicated a clinically relevant DDI
potential for both UGT isoforms substrate. However, even when

Fig. 6. Isolines plots for relationship of AUC ratio against oral dose of ibrutinib and fm by UGT1A1 (A), UGT1A3 (B), and UGT1A7 (C) for DDI study. An isoline was
joining equal points, values in each line represent the AUC ratio (AUCi/AUC) calculated by the eq. (8) according to the corresponding oral dose of ibrutinib and fm.

6
X. Wang et al. Toxicology and Applied Pharmacology 424 (2021) 115595

Fig. 7. Isolines plots for relationship of AUC ratio against oral dose of acalabrutinib and fm by UGT1A1 (A), UGT1A3 (B), and UGT1A7 (C) for DDI study. An isoline
was joining equal points, values in each line represent the AUC ratio (AUCi/AUC) calculated by the eq. (8) according to the corresponding oral dose of acalabrutinib
and fm.

administered at the highest dose (700 mg/day) and an fm is 1, the AUC there is an urgent need for carefully evaluation of the clinical DDIs po­
ratio is 1.10 (less than the cut-off value 1.25) for the substrates metab­ tential based on methodology and modeling methods in vitro to recog­
olized by UGT1A7 (Fig. 6C), indicating minor clinical impacts on the nize whether adjust dose regiments (Tornio et al., 2019). It has been
pharmacokinetics of UGT1A7 substrates. Consequently, ibrutinib is reported that strong inhibitors or inducers of CYP3A may have a rele­
likely to result into the potential for clinically significant DDIs when co- vant effect on the PK of both two drugs and lead to a series of adverse
administered with UGT1A1, or UGT1A3 substrates through inhibition of events, the effects on human UGTs still need further discussed. Previous
glucuronidation. study revealed the inhibitory effects of ibrutinib on UGT enzymes and
In this study, we assumed the Ki values of UGT1A1,1A3, and 1A7 by gave the prediction of DDIs risk of UGT1A1, 1A4 and 1A9 (Korpra­
acalabrutinib were 4.27, 8.56, and 13.81 μM, respectively. As shown in sertthaworn et al., 2019). In the current study, our data offered evidence
Fig. 7, when administered acalabrutinib at the recommended dose of that potent inhibition against UGT1A1 and UGT1A3 by ibrutinib might
200 mg/day and fm is 1, the AUC ratio of victim drugs cleared by increase the risk of clinically significant DDIs when UGT1A1, or
UGT1A1,1A3 and 1A7 are increased by 1.28, 1.14 and 1.09-fold, UGT1A3 substrates were co-administered. Acalabrutinib was unlikely to
respectively, which indicated that acalabrutinib could not bring the trigger clinically significant DDIs through UGT inhibition due to its weak
evident changes in the AUC of the victim drugs. effects.
UGT1A1 is the only enzyme that metabolized bilirubin and facilitates
4. Discussion its elimination from the body (Bosma et al., 1994). Inhibition of UGT1A1
by ibrutinib may cause hyperbilirubinemia in vivo. This result might
The advent of BTK inhibitors ibrutinib and acalabrutinib represents a provide a better understanding about ibrutinib-induced acute liver
major breakthrough in the treatment of CLL and other B cell malig­ injury which initially characterized by marked elevations of bilirubin
nancies (Sibaud et al., 2020). However, the coadministration with a levels (Nandikolla et al., 2017; Tafesh et al., 2019). Indeed, several TKIs
range of concomitant treatments is expected to under the high risks of such as erlotinib, nilotinib, pazopanib, vemurafenib with potent inhib­
DDIs. As these kinds of events are by nature essentially unprevented, itory effect on UGT1A1 have been reported to have a high incidence of

7
X. Wang et al. Toxicology and Applied Pharmacology 424 (2021) 115595

hyperbilirubinemia (Qosa et al., 2018). Particularly, pazopanib inhibi­ smoking) and diseases, will need consideration to progress the predic­
ted UGT1A1-mediated SN-38 (active metabolite of irinotecan) glucur­ tion towards a quantitative basis (Rostami-Hodjegan and Tucker, 2007).
onidation and increased SN-38 AUC in recombinant human UGT1A1 Thus, assessing DDIs involving UGT inhibition remains challenging.
Supersomes by a factor of 2.2 (Iwase et al., 2019). These results were However, this research was a preliminary study. Further mechanistic
comparable to the ~90% increase in the AUC of SN-38 clinically and clinical studies still need to be performed to gain more accurate
observed in patients who received 800 mg pazopanib once daily (Ben­ evaluation of DDIs potential associated with ibrutinib and acalabrutinib.
nouna et al., 2015). The prediction from in vitro data demonstrated that In conclusion, results from this research provide essential informa­
atazanavir (a potent UGT1A1 inhibitor) increased in molidustat AUC of tion about the inhibition of ibrutinib and acalabrutinib towards UGTs
2.9-fold. In the clinical DDI study, a 2-fold increase of AUC for moli­ isoforms and evaluate the possible DDIs risk. The present findings raise
dustat was also observed when pre- and co-treatment with atazanavir an alarm bell about potential DDI risk associated with ibrutinib, how­
which helps to confirm the assumptions made from the in vitro-in vivo ever, the extrapolation from in vitro data to in vivo drug interactions
correlation(van der Mey et al., 2021). In addition, common genetic should be taken with caution, and additional systemic study is needed.
polymorphisms of UGT1A1*28 will lead to lower drug clearance and
have an increased risk of DDIs. Therefore, co-administered of antineo­ Declaration of Competing Interest
plastic agents with a narrow therapeutic index such as etoposide and
irinotecan could result in excessive cytotoxic drugs exposure even a The authors declare that there is no conflict of interests regarding the
slight increase of AUC(Wen et al., 2007) (Ichikawa et al., 2008). Taken publication of this article.
together, more attention should be paid when ibrutinib is co-
administered with UGT1A1 substrates which may cause a buildup of Acknowledgements
endogenous substances or toxic drug levels.
UGT1A3 is mainly responsible for metabolizing nonsteroidal anti- This study was financially supported by the National Key Research
inflammatory drugs (NSAIDs) and some important endogenous sub­ and Development Program of China (2017YFC1702006), the Funda­
stances, such as bile acids, estrogens (Lepine et al., 2004; Kuehl et al., mental Research Funds for the Central Universities (DUT21LK11).
2005). Inhibition of UGT1A3 might affect the conjugation of cheno­
deoxycholic acid and thus to cholestasis (Trottier et al., 2006). Prior
References
studies have noted that cholestatic features were shown in patients with
severe acute liver injury after ibrutinib treatment (Tafesh et al., 2019). Advani, R.H., Buggy, J.J., Sharman, J.P., Smith, S.M., Boyd, T.E., Grant, B., Kolibaba, K.
Additionally, UGT1A3 activity is subject to about 60-fold inter- S., Furman, R.R., Rodriguez, S., Chang, B.Y., Sukbuntherng, J., Izumi, R., Hamdy, A.,
individual variability, implying increased clinical risks such as signifi­ Hedrick, E., Fowler, N.H., 2013. Bruton tyrosine kinase inhibitor ibrutinib (PCI-
32765) has significant activity in patients with relapsed/refractory B-cell
cant variations of plasma concentrations and subsequent toxicity (Izu­ malignancies. J. Clin. Oncol. 31, 88–94.
kawa et al., 2009). Evidence suggested that the observed increase in Bennouna, J., Deslandres, M., Senellart, H., de Labareyre, C., Ruiz-Soto, R., Wixon, C.,
atorvastatin lactonization in carriers of one or two UGT1A3*2 alleles, Botbyl, J., Suttle, A.B., Delord, J.P., 2015. A phase I open-label study of the safety,
tolerability, and pharmacokinetics of pazopanib in combination with irinotecan and
was the result of the strong increase in UGT1A3 expression. Also, data cetuximab for relapsed or refractory metastatic colorectal cancer. Investig. New
demonstrated that UGT1A3*2 genotype has a very substantial increase Drugs 33, 138–147.
in mRNA (5-fold) and protein (7.3-fold) in the human liver (Riedmaier Bosma, P.J., Seppen, J., Goldhoorn, B., Bakker, C., Oude Elferink, R.P., Chowdhury, J.R.,
Chowdhury, N.R., Jansen, P.L., 1994. Bilirubin UDP-glucuronosyltransferase 1 is the
et al., 2010).Therefore, it is conceivable that individuals with only relevant bilirubin glucuronidating isoform in man. J. Biol. Chem. 269,
UGT1A3*2/*2 may endure the toxic side effects caused by atorvastatin 17960–17964.
lactone. In addition, UGT1A3 has been reported to catalyze the glu­ Byrd, J.C., Harrington, B., O’Brien, S., Jones, J.A., Schuh, A., Devereux, S., Chaves, J.,
Wierda, W.G., Awan, F.T., Brown, J.R., Hillmen, P., Stephens, D.M., Ghia, P.,
curonidation of ibuprofen and ketoprofen (Sakaguchi et al., 2004), so it
Barrientos, J.C., Pagel, J.M., Woyach, J., Johnson, D., Huang, J., Wang, X.,
represents bleeding risk whilst taking ibrutinib with anti-inflammatory Kaptein, A., Lannutti, B.J., Covey, T., Fardis, M., McGreivy, J., Hamdy, A.,
drugs. Although the level of UGT1A3 expression in human liver is Rothbaum, W., Izumi, R., Diacovo, T.G., Johnson, A.J., Furman, R.R., 2016.
Acalabrutinib (ACP-196) in relapsed chronic lymphocytic leukemia. N. Engl. J. Med.
much lower than that of other UGT1A isoforms(Green et al., 1998), the
374, 323–332.
co-administrated ibrutinib with xenobiotic/drug mainly cleared by Cameron, F., Sanford, M., 2014. Ibrutinib: first global approval. Drugs 74, 263–271.
UGT1A3 needs to be taken additional caution. Carlini, L.E., Meropol, N.J., Bever, J., Andria, M.L., Hill, T., Gold, P., Rogatko, A.,
Since UGT1A7 is an important extra-hepatic UGT isoform that in­ Wang, H., Blanchard, R.L., 2005. UGT1A7 and UGT1A9 polymorphisms predict
response and toxicity in colorectal cancer patients treated with capecitabine/
fluences the disposition of drugs within the gut (Court et al., 2012) and irinotecan. Clin. Cancer Res. 11, 1226–1236.
plays a prominent role in the detoxification of tobacco carcinogens Court, M.H., Zhang, X., Ding, X., Yee, K.K., Hesse, L.M., Finel, M., 2012. Quantitative
benzo(α)pyrene (Fang et al., 2002). Previous study has described a distribution of mRNAs encoding the 19 human UDP-glucuronosyltransferase
enzymes in 26 adult and 3 fetal tissues. Xenobiotica 42, 266–277.
striking correlation between the UGT1A7 genotypes and the occurrence Davies, B., Morris, T., 1993. Physiological parameters in laboratory animals and humans.
of irinotecan-induced toxicity that inhibition of UGT1A7-medicated Pharm. Res. 10, 1093–1095.
glucuronidation pathway in the gut may predispose for irinotecan- de Jong, J., Sukbuntherng, J., Skee, D., Murphy, J., O’Brien, S., Byrd, J.C., James, D.,
Hellemans, P., Loury, D.J., Jiao, J., Chauhan, V., Mannaert, E., 2015. The effect of
induced diarrhea (Carlini et al., 2005). Additionally, it was reported food on the pharmacokinetics of oral ibrutinib in healthy participants and patients
that variations in the UGT1A7 gene that reduce UGT1A7 activity may with chronic lymphocytic leukemia. Cancer Chemother. Pharmacol. 75, 907–916.
increase the risk of smoking-related orolaryngeal cancer (Zheng et al., de Zwart, L., Snoeys, J., De Jong, J., Sukbuntherng, J., Mannaert, E., Monshouwer, M.,
2016. Ibrutinib dosing strategies based on interaction potential of CYP3A4
2001). A meta-analysis also demonstrated that individuals with
perpetrators using physiologically based pharmacokinetic modeling. Clin.
UGT1A7*3 allele (intermediate, and low activity UGT1A7 genotypes) Pharmacol. Ther. 100, 548–557.
were involved in a high risk of cancer (Lu et al., 2011). It can be Fang, J.L., Beland, F.A., Doerge, D.R., Wiener, D., Guillemette, C., Marques, M.M.,
Lazarus, P., 2002. Characterization of benzo(a)pyrene-trans-7,8-dihydrodiol
anticipated that inhibition of UGT1A7 and UGT1A7*3 genotype in vivo
glucuronidation by human tissue microsomes and overexpressed UDP-
deserved more attention. Although the degree of DDIs risk via UGT1A7 glucuronosyltransferase enzymes. Cancer Res. 62, 1978–1986.
inhibition is low in our prediction, the potent inhibition of ibrutinib on Green, M.D., King, C.D., Mojarrabi, B., Mackenzie, P.I., Tephly, T.R., 1998.
UGT1A7 in the gut is still noteworthy. Glucuronidation of amines and other xenobiotics catalyzed by expressed human
UDP-glucuronosyltransferase 1A3. Drug Metab. Dispos. 26, 507–512.
There are several limitations to this study. First, DDIs risk based-on Haupt, L.J., Kazmi, F., Ogilvie, B.W., Buckley, D.B., Smith, B.D., Leatherman, S., Paris, B.,
IVIVE approach would be regarded as an initial discriminating screen, Parkinson, O., Parkinson, A., 2015. The reliability of estimating Ki values for direct,
therefore, prediction from in vitro data to in vivo drug interactions must reversible inhibition of cytochrome P450 enzymes from corresponding IC50 values:
a retrospective analysis of 343 experiments. Drug Metab. Dispos. 43, 1744–1750.
be taken with caution (Ito et al., 2004). Moreover, variable factors such Ichikawa, W., Araki, K., Fujita, K., Yamamoto, W., Endo, H., Nagashima, F., Tanaka, R.,
as transporters, various metabolic enzymes, the extrinsic factors (diet, Miya, T., Kodama, K., Sunakawa, Y., Narabayashi, M., Ando, Y., Akiyama, Y.,

8
X. Wang et al. Toxicology and Applied Pharmacology 424 (2021) 115595

Kawara, K., Sasaki, Y., 2008. Re: UGT1A1*28 genotype and irinotecan-induced affect atorvastatin lactonization in vitro and in vivo. Clin. Pharmacol. Ther. 87,
neutropenia: dose matters. J. Natl. Cancer Inst. 100, 224–225 (author reply 225). 65–73.
Ito, K., Brown, H.S., Houston, J.B., 2004. Database analyses for the prediction of in vivo Rostami-Hodjegan, A., Tucker, G.T., 2007. Simulation and prediction of in vivo drug
drug-drug interactions from in vitro data. Br. J. Clin. Pharmacol. 57, 473–486. metabolism in human populations from in vitro data. Nat. Rev. Drug Discov. 6,
Iwase, M., Fujita, K.I., Nishimura, Y., Seba, N., Masuo, Y., Ishida, H., Kato, Y., Kiuchi, Y., 140–148.
2019. Pazopanib interacts with irinotecan by inhibiting UGT1A1-mediated Sakaguchi, K., Green, M., Stock, N., Reger, T.S., Zunic, J., King, C., 2004.
glucuronidation, but not OATP1B1-mediated hepatic uptake, of an active metabolite Glucuronidation of carboxylic acid containing compounds by UDP-
SN-38. Cancer Chemother. Pharmacol. 83, 993–998. glucuronosyltransferase isoforms. Arch. Biochem. Biophys. 424, 219–225.
Izukawa, T., Nakajima, M., Fujiwara, R., Yamanaka, H., Fukami, T., Takamiya, M., Seo, K.A., Kim, H.J., Jeong, E.S., Abdalla, N., Choi, C.S., Kim, D.H., Shin, J.G., 2014. In
Aoki, Y., Ikushiro, S., Sakaki, T., Yokoi, T., 2009. Quantitative analysis of UDP- vitro assay of six UDP-glucuronosyltransferase isoforms in human liver microsomes,
glucuronosyltransferase (UGT) 1A and UGT2B expression levels in human livers. using cocktails of probe substrates and liquid chromatography-tandem mass
Drug Metab. Dispos. 37, 1759–1768. spectrometry. Drug Metab. Dispos. 42, 1803–1810.
Korprasertthaworn, P., Chau, N., Nair, P.C., Rowland, A., Miners, J.O., 2019. Inhibition Sibaud, V., Beylot-Barry, M., Protin, C., Vigarios, E., Recher, C., Ysebaert, L., 2020.
of human UDP-glucuronosyltransferase (UGT) enzymes by kinase inhibitors: effects Dermatological toxicities of Bruton’s tyrosine kinase inhibitors. Am. J. Clin.
of dabrafenib, ibrutinib, nintedanib, trametinib and BIBF 1202. Biochem. Dermatol. 21, 799–812.
Pharmacol. 169, 113616. Strassburg, C.P., Oldhafer, K., Manns, M.P., Tukey, R.H., 1997. Differential expression of
Kuehl, G.E., Lampe, J.W., Potter, J.D., Bigler, J., 2005. Glucuronidation of nonsteroidal the UGT1A locus in human liver, biliary, and gastric tissue: identification of UGT1A7
anti-inflammatory drugs: identifying the enzymes responsible in human liver and UGT1A10 transcripts in extrahepatic tissue. Mol. Pharmacol. 52, 212–220.
microsomes. Drug Metab. Dispos. 33, 1027–1035. Tafesh, Z.H., Coleman, M., Fulmer, C., Nagler, J., 2019. Severe hepatotoxicity due to
Lepine, J., Bernard, O., Plante, M., Tetu, B., Pelletier, G., Labrie, F., Belanger, A., ibrutinib with a review of published cases. Case Rep. Gastroenterol. 13, 357–363.
Guillemette, C., 2004. Specificity and regioselectivity of the conjugation of estradiol, Tornio, A., Filppula, A.M., Niemi, M., Backman, J.T., 2019. Clinical studies on drug-drug
estrone, and their catecholestrogen and methoxyestrogen metabolites by human interactions involving metabolism and transport: methodology, pitfalls, and
uridine diphospho-glucuronosyltransferases expressed in endometrium. J. Clin. interpretation. Clin. Pharmacol. Ther. 105, 1345–1361.
Endocrinol. Metab. 89, 5222–5232. Trottier, J., Verreault, M., Grepper, S., Monte, D., Belanger, J., Kaeding, J., Caron, P.,
Li, W., Xing, Y., Liu, Y., 2015. Inhibition of SN-38 glucuronidation by gefitinib and its Inaba, T.T., Barbier, O., 2006. Human UDP-glucuronosyltransferase (UGT)1A3
metabolite. Cancer Chemother. Pharmacol. 75, 1253–1260. enzyme conjugates chenodeoxycholic acid in the liver. Hepatology 44, 1158–1170.
Lu, P.H., Chen, M.B., Wu, X.Y., Gu, J.H., Liu, Y., Gu, R.M., 2011. Genetic polymorphisms Uchaipichat, V., Mackenzie, P.I., Elliot, D.J., Miners, J.O., 2006. Selectivity of substrate
of UGT1A7 and cancer risk: evidence from 21 case-control studies. Cancer Investig. (trifluoperazine) and inhibitor (amitriptyline, androsterone, canrenoic acid,
29, 645–654. hecogenin, phenylbutazone, quinidine, quinine, and sulfinpyrazone) “probes” for
Markham, A., Dhillon, S., 2018. Acalabrutinib: first global approval. Drugs 78, 139–145. human udp-glucuronosyltransferases. Drug Metab. Dispos. 34, 449–456.
Marostica, E., Sukbuntherng, J., Loury, D., de Jong, J., de Trixhe, X.W., Vermeulen, A., USFDA, 2020. Guidance for Industry: In Vitro Drug Interaction Studies—Cytochrome
De Nicolao, G., O’Brien, S., Byrd, J.C., Advani, R., McGreivy, J., Poggesi, I., 2015. P450 Enzyme-and Transporter-Mediated Drug Interactions (Draft Guidance). Center
Population pharmacokinetic model of ibrutinib, a Bruton tyrosine kinase inhibitor, for Drug Evaluation and Research. FDA, Silver Spring, MD.
in patients with B cell malignancies. Cancer Chemother. Pharmacol. 75, 111–121. van der Mey, D., Gerisch, M., Jungmann, N.A., Kaiser, A., Yoshikawa, K., Schulz, S.,
Meech, R., Hu, D.G., McKinnon, R.A., Mubarokah, S.N., Haines, A.Z., Nair, P.C., Radtke, M., Lentini, S., 2021. Drug-drug interaction of atazanavir on UGT1A1-
Rowland, A., Mackenzie, P.I., 2019. The UDP-glycosyltransferase (UGT) superfamily: mediated glucuronidation of molidustat in human. Basic Clin. Pharmacol. Toxicol.
new members, new functions, and novel paradigms. Physiol. Rev. 99, 1153–1222. 128, 511–524.
Miners, J.O., Mackenzie, P.I., Knights, K.M., 2010. The prediction of drug- Wang, Z., Wang, X., Jia, Y., Yin, H., Feng, Y., Jiang, L., Cao, J., Liu, Y., 2020. Inhibition of
glucuronidation parameters in humans: UDP-glucuronosyltransferase enzyme- human UDP-glucuronosyltransferase enzymes by midostaurin and ruxolitinib:
selective substrate and inhibitor probes for reaction phenotyping and in vitro-in vivo implications for drug-drug interactions. Biopharm. Drug Dispos. 41, 231–238.
extrapolation of drug clearance and drug-drug interaction potential. Drug Metab. Wen, Z., Tallman, M.N., Ali, S.Y., Smith, P.C., 2007. UDP-glucuronosyltransferase 1A1 is
Rev. 42, 196–208. the principal enzyme responsible for etoposide glucuronidation in human liver and
Miners, J.O., Chau, N., Rowland, A., Burns, K., McKinnon, R.A., Mackenzie, P.I., intestinal microsomes: structural characterization of phenolic and alcoholic
Tucker, G.T., Knights, K.M., Kichenadasse, G., 2017. Inhibition of human UDP- glucuronides of etoposide and estimation of enzyme kinetics. Drug Metab. Dispos.
glucuronosyltransferase enzymes by lapatinib, pazopanib, regorafenib and sorafenib: 35, 371–380.
implications for hyperbilirubinemia. Biochem. Pharmacol. 129, 85–95. Yu, H., Tweedie, D., 2013. A perspective on the contribution of metabolites to drug-drug
Nandikolla, A.G., Derman, O., Nautsch, D., Liu, Q., Massoumi, H., Venugopal, S., interaction potential: the need to consider both circulating levels and inhibition
Braunschweig, I., Janakiram, M., 2017. Ibrutinib-induced severe liver injury. Clin. potency. Drug Metab. Dispos. 41, 536–540.
Case Rep. 5, 735–738. Yu, J., Petrie, I.D., Levy, R.H., Ragueneau-Majlessi, I., 2019. Mechanisms and clinical
Oda, S., Fujiwara, R., Kutsuno, Y., Fukami, T., Itoh, T., Yokoi, T., Nakajima, M., 2015. significance of pharmacokinetic-based drug-drug interactions with drugs approved
Targeted screen for human UDP-glucuronosyltransferases inhibitors and the by the U.S. Food and Drug Administration in 2017. Drug Metab. Dispos. 47,
evaluation of potential drug-drug interactions with zafirlukast. Drug Metab. Dispos. 135–144.
43, 812–818. Zheng, Z., Park, J.Y., Guillemette, C., Schantz, S.P., Lazarus, P., 2001. Tobacco
Pharmacyclics Inc, 2013. Imbruvica™ (ibrutinib) Capsules for Oral Use; US Prescribing carcinogen-detoxifying enzyme UGT1A7 and its association with orolaryngeal
Information. http://www.accessdata.fda.gov/drugsatfda_docs/label/2013/20 cancer risk. J. Natl. Cancer Inst. 93, 1411–1418.
5552s000lbl.pdf. (Accessed 10 December 2013). Zhou, X., Xie, Y., Qi, Q., Cheng, X., Liu, F., Liao, K., Wang, G., Hao, H., 2013. Disturbance
Qosa, H., Avaritt, B.R., Hartman, N.R., Volpe, D.A., 2018. In vitro UGT1A1 inhibition by of hepatic and intestinal UDP-glucuronosyltransferase in rats with trinitrobenzene
tyrosine kinase inhibitors and association with drug-induced hyperbilirubinemia. sulfonic acid-induced colitis. Drug Metab. Pharmacokinet. 28, 305–313.
Cancer Chemother. Pharmacol. 82, 795–802. Zhou, D., Podoll, T., Xu, Y., Moorthy, G., Vishwanathan, K., Ware, J., Slatter, J.G., Al-
Riedmaier, S., Klein, K., Hofmann, U., Keskitalo, J.E., Neuvonen, P.J., Schwab, M., Huniti, N., 2019. Evaluation of the drug-drug interaction potential of acalabrutinib
Niemi, M., Zanger, U.M., 2010. UDP-glucuronosyltransferase (UGT) polymorphisms and its active metabolite, ACP-5862, using a physiologically-based pharmacokinetic
modeling approach. CPT Pharmacometrics Syst. Pharmacol. 8, 489–499.

You might also like