Abstract
We aimed to use physiologically based pharmacokinetic (PBPK) modeling and simulation to predict imatinib steady‐state plasma exposure in patients with chronic myeloid leukemia (CML) to investigate variability in outcomes. A validated imatinib PBPK model (Simcyp Simulator) was used to predict imatinib AUCss, Css,min and Css,max for patients with CML (n = 68) from a real‐world retrospective observational study. Differences in imatinib exposure were evaluated based on clinical outcomes, (a) Early Molecular Response (EMR) achievement and (b) occurrence of grade ≥3 adverse drug reactions (ADRs), using the Kruskal‐Wallis rank sum test. Sensitivity analyses explored the influence of patient characteristics and drug interactions on imatinib exposure. Simulated imatinib exposure was significantly higher in patients who achieved EMR compared to patients who did not (geometric mean AUC0‐24,ss 51.2 vs. 42.7 μg h mL−1, p < 0.05; Css,min 1.1 vs. 0.9 μg mL−1, p < 0.05; Css,max 3.4 vs. 2.8 μg mL−1, p < 0.05). Patients who experienced grade ≥3 ADRs had a significantly higher simulated imatinib exposure compared to patients who did not (AUC0‐24,ss 56.1 vs. 45.9 μg h mL−1, p < 0.05; Css,min 1.2 vs. 1.0 μg mL−1, p < 0.05; Css,max 3.7 vs. 3.0 μg mL−1, p < 0.05). Simulations identified a range of patient (sex, age, weight, abundance of hepatic CYP2C8 and CYP3A4, α1‐acid glycoprotein concentrations, liver and kidney function) and medication‐related factors (dose, concomitant CYP2C8 modulators) contributing to the inter‐individual variability in imatinib exposure. Relationships between imatinib plasma exposure, EMR achievement and ADRs support the rationale for therapeutic drug monitoring to guide imatinib dosing to achieve optimal outcomes in CML.
Keywords: adverse drug reactions, drug interactions, early molecular response, exposure‐response, imatinib, physiologically based pharmacokinetic (PBPK), simulation
Combining real word clinical data and pharmacokinetic modeling, this study investigated the exposure‐response relationship for imatinib in people with chronic myeloid leukemia. Patients with higher imatinib exposure were more likely to achieve clinical response, but also the risk of imatinib‐related harms.

Abbreviations
- AAG
α1‐acid glycoprotein
- ADR
adverse‐drug reaction
- ADRs
adverse drug reactions
- AUCss
area under the concentration‐time curve at steady state
- BSA
body surface area
- CI
confidence interval
- CLR
renal clearance
- CML
chronic myeloid leukemia
- DMR
deep molecular response
- EMR
Early Molecular Response
- fup
unbound fraction in plasma
- HLM
human liver microsomes
- MMR
major molecular response
- OR
Odds ratio
- PBPK
physiologically based pharmacokinetic
- PK
pharmacokinetics
- SD
standard deviation
- sDMR
sustained deep molecular response
- TDM
therapeutic drug monitoring
- TKI
tyrosine kinase inhibitor
1. INTRODUCTION
Imatinib, a BCR‐ ABL tyrosine kinase inhibitor (TKI), has significantly changed the treatment landscape of Philadelphia chromosome‐positive chronic myeloid leukemia (CML). Despite showing significantly superior survival outcomes and improved tolerability compared to interferon‐α plus low‐dose cytarabine, 1 many patients need to discontinue imatinib treatment and switch to another TKI. 2 We recently conducted a retrospective observational study of real‐world patients with CML in Australia and observed that 57% (95% confidence interval [CI], 46–66%) of patients had experienced an imatinib‐related adverse‐drug reaction (ADR) necessitating imatinib dose changes or discontinuation, and 53% (95% CI, 42–63%) a grade ≥3 ADR, by 18 months of treatment. 2 These patients required increased monitoring and utilization of healthcare services, with many needing to commence new long‐term medicines for the management of imatinib‐related ADRs. 2 A large proportion of patients never achieved sustained deep molecular response (sDMR; BCR‐ABL1 IS ≤0.01% maintained for at least 2 consecutive years) with imatinib treatment (5‐year cumulative incidence of 41%; 95% CI, 27–55%) 2 ; a minimum requirement for attempting TKI treatment discontinuation. 3 , 4 Furthermore, one in four patients failed to achieve Early Molecular Response (EMR; BCR‐ABL1 IS ≤ 10%) by 3 months of imatinib treatment 2 ; an endpoint predictive for major molecular response (MMR), deep molecular response (DMR) and sDMR achievement. 2 , 5 , 6
Inter‐individual variability in imatinib pharmacokinetics (PK) is a possible determinant of variability in imatinib response. 7 Large inter‐individual variability has been observed in imatinib trough plasma concentrations at steady‐state (Css,min) in patients administered the same dose, with reported coefficient of variations (CV%) of 40 to 60%. 8 , 9 , 10 , 11 , 12 As a drug with a low hepatic extraction ratio, steady‐state total plasma imatinib concentrations are determined by variability in intrinsic hepatic clearance (metabolism and transport) and plasma protein binding. 13 Both imatinib and its major metabolite N‐desmethyl imatinib undergo mostly hepatic elimination, with biotransformation of imatinib to N‐desmethyl imatinib by cytochrome P450 (CYP)3A4 and CYP2C8 a clinically important inactivating process. 7 , 13 Imatinib is also a substrate of P‐glycoprotein (P‐gp) and breast cancer resistant protein (BCRP) efflux transporters, which contribute to the net intestinal absorption of imatinib (via expression of transporters on apical/luminal membrane of enterocytes), biotransformation and excretion of imatinib (via expression of transporters on the bile caniculi of hepatocytes), 7 , 13 and retention of imatinib in target cancer cells. 14 , 15 , 16 , 17 The expression and activities of drug‐metabolizing enzymes and transporters, which can be affected by genetic or environmental factors (e.g., concomitant medicine use and diet), 18 are hypothesized to influence imatinib plasma and intracellular concentrations, and thus the efficacy and tolerability of imatinib. Patients receiving imatinib treatment are likely to be receiving multiple concomitant medicines for management of their comorbidities (and treatment related ADRs), due to the older median age at CML diagnosis and expected longer survival of patients treated with TKIs. 19 , 20 , 21 As a basic compound, imatinib exhibits concentration‐dependent binding with high affinity to α1‐acid glycoprotein (AAG), with an unbound fraction in plasma (fup) of 0.05. 22 As an acute phase protein, AAG demonstrates highly variable concentrations, increasing 2 to 6‐fold in response to injury, infection, inflammation and cancer. 22 , 23 Furthermore, there are different genetic polymorphisms of AAG, with different binding properties of variants reported. 24
Mechanistic modeling and simulation approaches can be used for predicting and understanding the PK of a drug. Physiologically based pharmacokinetic (PBPK) modeling integrates drug‐specific parameters (e.g., plasma protein binding and clearance) and drug independent system‐specific parameters (e.g., population variables such as organ blood flow, organ volumes and tissue composition) to simulate the time course of drug concentrations in different clinical populations. 25 , 26 Using a PBPK model, the drug concentration‐time profile from a specific dose regimen, in a patient from a population for which clinical data was not available, can be predicted. The concentration‐time profile is predicted by considering key determinants of PK in an individual patient, such as geographic ancestry, weight, age, kidney and hepatic function, and genetic polymorphisms in drug‐metabolizing enzymes or transporters. PBPK modeling can also be used to understand possible drug–drug interactions, 25 , 26 , 27 , 28 , 29 , 30 and to predict characteristics that influence inter‐individual variability in drug exposure. 31 , 32 Identifying factors that contribute to achievement of EMR and occurrence of imatinib‐related ADRs can help inform initial imatinib dose selection, identify patients that may benefit from an alternative TKI, and inform the use of therapeutic drug monitoring (TDM) to guide dose adjustments.
The aim of this study is to investigate predictions of imatinib steady‐state area under the concentration‐time curve (AUCss), maximum concentration (Css,max), Css,min, and apparent clearance (CL/F) using a PBPK model in virtual patients with the demographic and clinical characteristics identical to those patients in the real‐world observational cohort study, 2 and to investigate the association between predicted plasma exposure and clinical outcomes (severe ADRs, failure to achieve EMR). The PBPK model will also be used to provide mechanistic insight into factors contributing to variability in imatinib PK, and to explore the PK implications of concomitant medicines identified in the real‐world patient cohort study.
2. METHODS
A schematic representation of the study workflow is presented in Figure 1. All PBPK modeling and simulations were conducted using the Simcyp Population‐based Simulator (version 18, Certara UK Limited, Simcyp Division). 33
FIGURE 1.

Schematic representation of the workflow of this study. ADR, adverse drug reaction; AUC, area under the plasma concentration‐time curve; Cmax, maximum (peak) plasma concentration; Cmin, minimum (trough) plasma concentration; CYP, cytochrome P450; EMR, early molecular response; PBPK, physiologically based pharmacokinetic.
2.1. Prediction of imatinib exposure in patients based on clinical outcomes
A previously developed and validated PBPK model of imatinib in adults 34 was used to predict imatinib steady‐state systemic exposure in patients with CML from the real‐world retrospective observational study described in Adattini et al. 2 The imatinib PBPK model has been verified extensively using clinical pharmacokinetic data from healthy and patient populations (gastrointestinal stromal tumor and CML) across different geographic ancestries and dosing regimens. 34 , 35 The imatinib PBPK model has also demonstrated excellent performance in predicting the drug interaction between imatinib (chronic use) and carbamazepine, a CYP3A4 and CYP2C8 inducer, with predicted pharmacokinetic parameters within 1.25‐fold of the clinically observed values. 34
Details of the drug‐related input parameters from the imatinib PBPK model are listed in Supplementary Table 1. The fup of 0.05 for AAG binding was assigned to the PBPK model of imatinib based on the reported value in healthy Europeans. 36 , 37 Although higher plasma AAG concentrations have been reported in patients with solid tumors (1650 ± 160 mg L−1 in lung cancer; 1600 mg L−1 in breast cancer), 38 plasma AAG concentrations reported in patients with chronic phase chronic myeloid leukemia (CML; 1200 ± 120 mg L−1 at diagnosis, and 1100 ± 60 mg L−1 at follow‐up) 39 , 40 , 41 are closer to those reported in healthy individuals (mean 910 ± 40 mg L−1), 38 corresponding to a similar unbound fraction in plasma (fup) of imatinib in healthy individuals and patients with chronic phase CML. 41 The corresponding dissociation constant was fixed and assumed to be independent of plasma AAG concentrations, with the variability in fup reflecting interindividual variability in AAG concentrations. The intrinsic clearances of imatinib to N‐desmethyl imatinib and other metabolites were estimated from in vitro kinetic data using recombinant CYP3A4 and human liver microsomes (HLM, in the presence of azamulin, a selective irreversible inhibitor of CYP3A4), with HLM (supplemented with azamulin) representing the CYP2C8 metabolic pathway given the assumed minor contribution (3%) of other CYP enzymes to imatinib metabolism. 42 The mechanism‐based inhibition of CYP3A4 following multiple doses of imatinib was modeled by an increase in degradation of the active enzyme and thus, a decrease in enzyme activity over time, based on a turnover model detailed elsewhere. 43 P‐gp and BCRP‐mediated biliary clearance of imatinib were modeled using data from previous in vitro studies. 44 , 45 The renal clearance (CLR) value for imatinib was taken from a study in patients with CML and Philadelphia chromosome‐positive acute lymphatic leukemia. 46 Model assumptions are further detailed in Adiwidjaja et al. 34 As the PBPK model of imatinib by Adiwidjaja et al 34 used the Simcyp Simulator version 17 for model development and verification, the imatinib PBPK model was reverified using the Simcyp Simulator version 18 (Supplementary Methods, Supplementary Table 2).
Patient characteristics from the real‐world retrospective observational study 2 were grouped according to achievement of the following clinical outcome measures:
Achievement of EMR (BCR‐ABL1 IS ≤ 10%) at 3 months of imatinib treatment (as defined by the National Comprehensive Cancer Network 47 )
Occurrence of a grade ≥3 imatinib‐related ADR within the follow‐up period (defined using the Common Terminology Criteria for Adverse Events version 5 grading scale 48 )
In each scenario, individual patient parameters were added into the PBPK model for simulation of individual patient imatinib plasma concentration‐time profiles and estimation of AUCss (μg h mL−1), Css,max (μg mL−1), Css,min (μg mL−1) and CL/F (L h−1). Parameters used for virtual patients matched the real‐world patient cohort 2 with respect to age (years), sex, actual body weight (ABW in kg; if available), height (cm; if available), serum creatinine concentration (μmol L−1; if available), hepatic function (defined by presence and severity of cirrhosis), kidney function (defined using glomerular filtration rate [GFR], estimated via the Chronic Kidney Disease – Epidemiology Collaboration equation 49 ), geographic ancestry, imatinib dose (mg/day) and concomitant medicine use (if a validated model was available). The “Sim‐NEurCaucasian” population available in the Simcyp library was used for patients of European ancestry, and the “Sim‐Chinese” 50 and “Sim‐Japanese” 51 populations were used for patients of Chinese and Japanese ancestry, respectively. Details of the virtual populations used in the PBPK simulations, including for patients with hepatic or kidney dysfunction, are provided in the Supplementary Methods. PBPK model input variables of all patients from the real‐world observational study 2 that were included in the PBPK simulations are presented in Supplementary Tables 3 and 4.
A total of 10 virtual trials for each simulation were carried out. A multiple‐dosing regimen of imatinib given for 14 days was used to achieve steady‐state. Potential differences in imatinib AUCss, Css,max, Css,min and CL/F within different clinical scenario outcomes (i.e., between those who achieved EMR and those who did not) were evaluated by the independent two sample t‐test or the Kruskal‐Wallis rank sum test (if residuals were not normally distributed). A Css,min of 1002 ng mL−1 was the exposure target for clinical response used to interpret the simulated exposures, with previous studies correlating this threshold with a significantly higher probability of MMR achievement. 9 , 52 All reported p values are two‐sided, with a significance level of 0.05. All statistical analyses were conducted using R (version 3.3.3). 53
2.2. Sensitivity analyses
Sensitivity analyses were conducted to evaluate the influence of variation in input parameters on the simulated imatinib plasma concentration at steady‐state. 33 , 54 , 55 These analyses were conducted using either (a) the automated sensitivity analysis tool available in the Simcyp Simulator, or (b) repeated simulations using a trial design with virtual age and sex‐matched subjects, whilst adjusting values of the input parameter of interest (a total of 10 virtual trials for each subgroup within the input parameter, with 10 subjects per trial). Parameters selected for sensitivity analyses included sex, age, ABW, kidney or hepatic dysfunction (eGFR and cirrhosis, respectively), hepatic and intestinal CYP3A4 enzyme abundance, hepatic CYP2C8 enzyme abundance, hepatic P‐gp transporter abundance, hepatic BCRP transporter abundance, and AAG abundance.
To investigate the influence of variation in kidney function on imatinib exposure, default “Sim‐RenalGFR_less30” and “Sim‐RenalGFR_30to60” populations were used. Sensitivity analyses were also conducted using the modified kidney dysfunction populations (adjusted CYP2C8 and CYP3A4 abundances) described in Supplementary Methods. To investigate the influence of variation in hepatic function on imatinib exposure, default “Sim‐Cirrhosis‐CP” populations were used. 56 The values for AAG abundance, drug‐metabolizing enzyme abundances, and transporter abundances used in the sensitivity analyses reflect physiologically plausible ranges. A meta‐analysis on the abundance of human hepatic CYP enzymes in Caucasian adult livers reported a CYP3A4 abundance of 93.0 pmol mg−1 protein (range, 0–600.0), and CYP2C8 abundance of 22.4 pmol mg−1 protein (range, 0–85.0), with a positive correlation between expression levels of CYP3A4 and CYP2C8 (Rs 0.68; p < 0.001). 57 In healthy Caucasian adults, AAG concentration ranges from 0.36 to 1.46 g L−1. 58 However, as an acute phase protein, AAG can increase approximately 2 to 6‐fold in many disease states, with concentrations up to 3.15 g L−1 reported. 22 A meta‐analysis on the abundance of hepatic transporters in liver tissue of healthy Caucasian adults reported a weighted mean P‐gp abundance of 0.201 pmol per 106 hepatocytes (CV 46%), and weighted mean BCRP abundance of 0.044 pmol per 106 hepatocytes (CV 40%). 59 Different types of liver damage have been shown to affect hepatocellular transport abundance to various extents. 59 , 60 Compared to healthy livers, hepatic P‐gp abundance was reported to increase up to 350% in cholestatic liver disease, and up to 410% in autoimmune hepatitis, with no significant differences observed in fatty liver disease, alcoholic liver disease, or hepatic virus C‐induced liver damage. 59 , 60 Conversely, hepatic BCRP abundance appears preserved in the these types of liver damage. 59 , 60 If examining hepatic transporter abundance according to the functional state of the liver, a Child‐Pugh class C score has been reported to result in a 260% increase in protein abundance of P‐gp, with no change in BCRP abundance. 60
To determine whether model input parameters had statistically significant effects on imatinib PK parameters (AUCss, Css,max, Css,min and CL/F), statistical analyses were conducted as described in the previous subsection. The parametric tests for comparing two (the independent two sample t‐test) or more groups (one‐way analysis of variance) were used. If the assumptions of normality and homogeneity of variances were not met, the corresponding non‐parametric tests (Wilcoxon‐Mann–Whitney and Kruskal‐Wallis rank sum test, respectively) were used.
2.3. Drug–drug interaction simulations
Imatinib is a substrate of CYP3A4 and CYP2C8, 13 and has also been shown to inhibit its own CYP3A‐mediated metabolism after multiple‐dose administration. 42 As such, PBPK simulations were performed to predict the extent of interaction between imatinib and CYP3A4/CYP2C8 modulators frequently used by patients in the real‐world retrospective observational study 2 ; verapamil (a moderate CYP3A4 inhibitor), diltiazem (a moderate CYP3A4 inhibitor), trimethoprim (a weak CYP2C8 inhibitor), clopidogrel (moderate CYP2C8 inhibitor) and hyperforin (a CYP3A4 inducer in intestinal enzymes). The default PBPK model library files for verapamil and its metabolite norverapamil, diltiazem and its metabolite N‐desmethyldiltiazem, and trimethoprim were used as included in the Simcyp Simulator. The previously verified PBPK models of clopidogrel and its clopidogrel glucuronide metabolite 61 were used to evaluate the potential CYP2C8 mediated drug–drug interaction with clopidogrel and imatinib. A verified PBPK model of hyperforin 62 was used to predict the potential drug–drug interaction between St John's Wort and imatinib.
The simulations were performed using the default “Sim‐NEurCaucasian” population with a multiple‐dosing regimen of imatinib 400 mg daily for 14 days, with the trial design representative of the demographic characteristics of patients in the real‐world observational study (10 subjects, 4 females, age range 20–91 years). 2 The CYP modulators were simulated at clinically relevant dose regimens for 14 days, except for trimethoprim which was dosed for 7 days to reflect clinical practice guidelines when treating urinary tract infections. A total of 10 virtual trials for each simulation were carried out to capture population variability. The interaction between imatinib and CYP modulators at steady‐state are reported as the geometric mean ratio of simulated PK parameters of imatinib (AUCss, Css,max, Css,min and CL/F) with and without the perpetrator drugs (CYP modulators), and their associated 90% confidence intervals. If the predicted ratio of PK parameters fell within 1.25‐fold (0.80–1.25), the interaction was not considered clinically significant. 63
Key protein targets and ligands in this article are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY, 64 and are permanently archived in the Concise Guide to PHARMACOLOGY 2019/20. 65
3. RESULTS
3.1. Prediction of imatinib exposure in patients based on clinical outcomes
Individual patient characteristics, imatinib dose regimens and simulated PK parameters are presented in Supplementary Tables 3 and 4. The simulated geometric mean plasma concentration‐time profiles of imatinib at steady‐state for different clinical scenarios are summarized in Table 1 and Figures 2 and 3. Notably, the simulated geometric mean AUC during 24 h after the dose (AUC0‐24,ss), Css,max and Css,min of imatinib were significantly higher in patients who achieved EMR at 3 months, compared to those who did not (p < 0.05). Unlike patients who did not achieve EMR, the geometric mean Css,min of patients who achieved EMR fell above the efficacy target of 1002 ng mL−1. 9 , 52 Moreover, imatinib AUC0‐24,ss, Css,max, and Css,min were significantly higher in patients who experienced a grade ≥3 ADR on imatinib treatment compared to those who did not (p < 0.05).
TABLE 1.
Summary of PBPK model predicted PK parameters of imatinib following multiple‐dose regimens, in patients with CML followed in the real‐world retrospective observational study, 2 grouped by clinical scenario outcome.
| PBPK parameter prediction | EMR achieved (n = 35) | EMR not achieved (n = 10) | Significance of predicted EMR difference | Grade ≥3 ADR experienced (n = 47) | Grade ≥3 ADR not experienced (n = 21) | Significance of predicted ADR difference |
|---|---|---|---|---|---|---|
| AUC0‐24,ss (μg h mL−1) | 51.17 (48.43—54.07) | 42.68 (38.01—47.91) | p < 0.001* | 56.09 (53.56—58.73) | 45.91 (42.54—49.54) | p < 0.001* |
| Css,min (μg mL−1) | 1.06 (0.97—1.16) | 0.91 (0.77—1.07) | p < 0.05* | 1.23 (1.14—1.32) | 1.00 (0.90—1.12) | p < 0.05* |
| Css,max (μg mL−1) | 3.41 (3.27—3.55) | 2.81 (2.55—3.09) | p < 0.001* | 3.73 (3.60—3.86) | 2.96 (2.78—3.15) | p < 0.001* |
| CL/F (L h−1) | 9.81 (9.28—10.38) | 10.22 (9.14—11.43) | p = 0.32 | 8.96 (8.58—9.36) | 10.53 (9.82—11.30) | p < 0.001* |
Notes. A total of 10 trials for each simulation were carried out. All pharmacokinetic parameters are reported as geometric mean values (95% confidence intervals) of PBPK model predictions.
Abbreviations: ADR, adverse drug reaction; AUC0‐24,ss, area under the plasma concentration‐time curve from time zero to 24 h after a dose at steady‐state; Css,max, maximum (peak) plasma concentration at steady‐state; Css,min, minimum (trough) plasma concentration at steady‐state; CL/F, apparent clearance; CML, chronic myeloid leukemia; EMR, early molecular response at 3 months; PBPK, physiologically based pharmacokinetic; PK, pharmacokinetic.
Statistical significance evaluated using Kruskal‐Wallis Rank Sum Test.
FIGURE 2.

PBPK model simulated plasma concentration‐time profiles of imatinib in patients treated in the real‐world retrospective observational study, 2 grouped by clinical outcome; (A) achievement of EMR at 3 months (N = 45), (B) occurrence of a grade ≥3 imatinib‐related ADR (N = 68). Geometric mean simulated plasma concentration time curves of imatinib at steady‐state in patients who experienced (blue dotted line) and did not experience (orange line) the clinical outcome of interest in the retrospective observational study are presented. The Css,min target of 1.002 μg mL−1 is shown by dashed black lines. ADR, adverse drug reaction; Css,min, minimum (trough) plasma concentration at steady‐state; EMR, early molecular response at 3 months; PBPK, physiologically based pharmacokinetic.
FIGURE 3.

PBPK model simulated PK parameters of imatinib in patients treated in the real‐world retrospective observational study, 2 grouped by achievement of EMR or occurrence of a grade ≥3 imatinib‐related ADR. Simulation results are depicted as geometric mean predicted Css,max, Css,min, AUC0‐24,ss and CL/F of imatinib (and 95% confidence intervals) in patients who experienced the clinical outcome of interest (blue line) and in patients who did not experience the clinical outcome of interest (orange line) in the retrospective observational study. 35 patients achieved EMR, whilst 10 did not achieve EMR. 47 patients experienced a grade ≥3 ADR, whilst 21 did not. *Statistically significant difference (p < 0.05; Kruskal‐Wallis rank sum test). ADR, adverse drug reaction; AUC0‐24,ss, area under the plasma concentration‐time curve from time zero to 24 h after a dose at steady‐state; Css,max, maximum (peak) plasma concentration at steady‐state; Css,min, minimum (trough) plasma concentration at steady‐state; CL/F, apparent clearance; EMR, early molecular response at 3 months; PBPK, physiologically based pharmacokinetic; PK, pharmacokinetic.
3.2. Sensitivity analyses
The impact of various parameters on simulated imatinib systemic exposure is shown in Supplementary Figure 1. The simulated plasma concentration‐time profile of imatinib was sensitive to variation in subject age, in which a change in age from 20 to 55 years increased imatinib AUC0‐24,ss by 30% (p < 0.05) and increased Css,min by 46% (p < 0.05). The effect was notably larger in older age, with a change in age from 55 to 90 years increasing imatinib AUC0‐24,ss and Css,min 2‐fold and 2.6‐fold, respectively (p < 0.05). Increased ABW (45–125 kg) was associated with significantly decreased imatinib AUC 0‐24,ss (44%) and Css,min (40%, p < 0.05). Although Css,max was modestly higher in females (18%; p < 0.05), other PK parameters were not significantly altered by sex.
Variation in hepatic CYP2C8 and CYP3A4 enzyme abundances, and in AAG concentrations, significantly influenced imatinib systemic exposure. A hepatic CYP2C8 abundance range of 0–85 pmol mg−1 protein decreased imatinib AUC0‐24,ss and Css,min by 70% and 85%, respectively (p < 0.05). A hepatic CYP3A4 enzyme abundance range of 0 to 600 pmol mg−1 protein decreased imatinib AUC0‐24,ss and Css,min by 50% and 68%, respectively (p < 0.05). As AAG concentration increased from 0.3 to 3.2 mg/mL, total plasma imatinib AUC0‐24,ss increased by 8.4‐fold (p < 0.05) and Css,min increased by 26‐fold (p < 0.05). Intestinal CYP3A4 enzyme abundance, and hepatobiliary P‐gp and BCRP transporter abundances, had no significant impact on simulated imatinib exposure.
Simulated imatinib plasma concentration‐time profiles were sensitive to varying degrees of hepatic impairment (measured as liver cirrhosis) and kidney dysfunction (measured as eGFR). Child‐Pugh B cirrhosis was associated with a 36% increase in imatinib AUC0‐24,ss and 56% increase in Css,min compared to healthy (“Sim‐NEurCaucasian”) adults (p < 0.05), whilst Child‐Pugh C cirrhosis was associated with a 66% increase in AUC0‐24,ss and 100% increase in Css,min relative to healthy adults (p < 0.05). Child‐Pugh A cirrhosis did not significantly alter simulated imatinib exposure. In the default kidney dysfunction simulations, a GFR of 30–60 mL/min/1.73 m2 was associated with a 53% higher AUC0‐24,ss and 85% higher Css,min relative to healthy adults (p < 0.05), whilst a GFR < 30 mL/min/1.73 m2 was associated with a 2.6‐fold increase in AUC0‐24,ss and 3.4‐fold increase in Css,min relative to healthy adults (p < 0.05). Simulated imatinib systemic exposure remained sensitive to kidney dysfunction in the modified populations (CYP enzyme abundance changed to default values in general populations), however, to a lesser extent (AUC0‐24,ss and Css,min increased by 1.75 and 2.3‐fold, respectively, for GFR < 30 mL/min/1.73 m2).
3.3. Drug–drug interaction simulations
The results of the simulations to predict the interactions between selected CYP modulators and imatinib are detailed in Figure 4 and Supplementary Table 5. Notably, the AUC0‐24,ss and Css,min of imatinib were predicted to increase by 23% and 34%, respectively, in the presence of trimethoprim. In the presence of clopidogrel, the AUC0‐24,ss of imatinib was simulated to increase by 32% and Css,min predicted to increase by 54%. Meanwhile, the interactions between imatinib and diltiazem (AUC0‐24,ss ratio = 1.04), and between imatinib and verapamil (AUC0‐24,ss ratio = 1.06) were predicted to be non‐clinically significant at steady‐state. At the recommended dosing regimen of St John's Wort (hyperforin 45 mg daily), there was minimal interaction with imatinib at steady‐state (AUC0‐24,ss ratio = 0.95; Css,max ratio = 0.96; Css,min ratio = 0.93; CL/F ratio = 1.05). This mirrors the geometric mean ratios reported in the PBPK drug–drug interaction simulations of imatinib with hyperforin by Adiwidjaja et al 62 (AUC0‐24,ss ratio = 0.94; Css,max ratio = 0.95).
FIGURE 4.

PBPK model prediction of the interactions between imatinib and a set of CYP modulators at steady‐state (A) diltiazem 45 mg QID for 14 days, (B) verapamil 60 mg TDS for 14 days, (C) hyperforin (St John's Wort) 45 mg daily for 14 days, (D) clopidogrel 75 mg daily for 14 days, (E) trimethoprim 300 mg daily for 7 days (day 8–14). Imatinib was administered at 400 mg daily for 14 days in a North European Caucasian population, with 10 subjects (4 females) and an age range of 20–91 years. Simulation results are depicted as geometric mean predicted plasma concentrations of imatinib in the presence and absence of coadministration with the CYP modulator. The extent of interactions was evaluated based on predicted pharmacokinetic parameter ratios (ratio in the presence and absence of CYP modulators) and their 90% confidence intervals (CI). A ratio of 1 indicates absence of drug interactions with imatinib. AUC0‐24,ss, area under the plasma concentration‐time curve from time zero to 24 h after a dose at steady‐state; Css,min, minimum (trough) plasma concentration at steady‐state; CYP, cytochrome P450; PBPK, physiologically based pharmacokinetic; QID, four times a day; TDS, three times a day.
4. DISCUSSION
This study utilized a PBPK approach to predict the imatinib plasma concentration‐time profile in patients with CML included in a real‐world study to investigate whether imatinib systemic exposure differed for patients who achieved EMR or experienced severe ADRs. The PBPK model predicted significantly higher imatinib AUCss, Css,max and Css,min in patients who achieved EMR at 3 months, compared to patients who did not. This relationship between predicted imatinib systemic exposure and EMR achievement is a key finding of this analysis and has not been previously reported in the literature. Previous studies have described the relationship between imatinib systemic exposure and other clinical response measures. A population pharmacokinetic analysis of imatinib in a European cohort (n = 20) demonstrated a significant correlation between lower imatinib unbound clearance and achievement of hematological response (Odds ratio [OR], 0.8 ± standard error 0.9; p < 0.05). 66 Many studies have correlated imatinib Css,min with complete cytogenetic response (CCyR) and MMR achievement. 8 , 9 , 11 , 52 , 67 , 68 , 69 , 70 , 71 , 72 , 73 In a European cohort study of patients treated with imatinib for at least 12 months (n = 68), mean (± standard deviation [SD]) Css,min was significantly higher in those who had achieved CCyR than in those who had not (1123 ± 617 ng mL−1 vs. 694 ± 556 ng mL−1, respectively; p < 0.05), and again higher in the group that had achieved MMR than in the group who had not (1452 ± 649 ng mL−1 vs. 869 ± 427 ng mL−1, respectively; p < 0.001). 9 A pharmacokinetic analysis of the TOPS study (n = 240) showed significantly faster times to MMR, and higher rates of both MMR and CCyR at 6 and 12 months, when Css,min ≥ 1165 ng mL−1. 67 Imatinib Css,min > 1000 ng mL−1 predicted improved MMR, event‐free and failure‐free survival in a cohort of Chinese patients with CML (n = 110), with those who achieved MMR showing higher mean imatinib Css,min compared with those not achieving MMR (1474 ± SD 419 ng mL−1 vs. 986 ± 213 ng mL−1, respectively; p < 0.001). 73 A study in Japanese patients with CML treated with imatinib (n = 254) observed a significantly lower probability of achieving MMR in patients with Css,min < 1002 ng mL−1. 52
Another major finding from these PBPK simulations was the significantly higher mean simulated imatinib systemic exposure in CML patients who experienced a grade ≥3 imatinib‐related ADR, compared to patients who did not. Previous studies have correlated imatinib plasma concentration with toxicity8, 66, 67, 74 however, a consistent threshold concentration has not been observed. In a sub‐analysis of the IRIS study (n = 351), fluid retention, rash, myalgia, and anemia were more common in patients who demonstrated higher plasma imatinib concentrations (Css,min > 1170 ng mL−1) compared to those with lower concentrations (Css,min ≤ 1170 ng mL−1). 8 A population pharmacokinetic model (n = 20) demonstrated that unbound imatinib AUC was significantly correlated with the occurrence and number of side effects (OR, 6.1 ± standard error 4.5; p < 0.05). 66 A cohort study in South Asian patients (n = 111) demonstrated a higher Css,min in patients who experienced grade ≥2 thrombocytopaenia compared to those who did not experience thrombocytopaenia (Css,min 1582 ± SD 355 ng mL−1 vs. 1337 ± 366 ng mL−1, respectively; p < 0.05). 74 Imatinib Css,min > 1000 ng mL−1 was correlated with leukopenia or neutropenia, diarrhea, periorbital and limb oedema, and rash (p < 0.05) in a Chinese cohort (n = 110), with an AUCss threshold of 1685 ng mL−1 identified for diarrhea and 1575 ng mL−1 for periorbital and limb oedema. 73 The TOPS study (n = 240) noted a significantly higher incidence of all‐grade neutropenia, anemia, leukopaenia, rash, diarrhea, arthralgia/myalgia and all‐cause oedema in patients with a Css,min > 3180 ng mL−1. 67 Further research is required to define the concentration‐threshold for imatinib‐related ADRs, which may help facilitate TDM‐guided dose adjustments and improve tolerability outcomes.
The predicted exposures for the various treatment outcome groups were a consequence of the differences in patient and medication‐related factors, and relationships between these covariates, that could be accommodated within the PBPK modeling and simulation software. Sensitivity analyses provided mechanistic insight into the factors contributing to inter‐individual variability in imatinib PK. The predicted imatinib plasma concentration–time profile was sensitive to changes in ABW, in line with previous studies where higher ABW was correlated to increased imatinib apparent volume of distribution (V/F) and CL/F, 75 , 76 , 77 , 78 and lower Css,min. 8 , 67 Another body size metric, body surface area (BSA), has been demonstrated as an important covariate of imatinib PK variability. 8 , 67 , 79 Accordingly, in both univariable and multivariable analyses in the retrospective observational study, 2 lower ABW was associated with higher rates of occurrence of grade ≥3 ADR during imatinib treatment.
Sensitivity analysis identified age as another influential factor contributing to variability in imatinib PK. Older age was associated with reduced imatinib CL/F and increased Css,min, Css,max and AUCss, confirming results from other PK studies. 8 , 80 The effect of age on imatinib PK supports the findings of the retrospective observational study, 2 whereby older age was associated with superior sDMR rates in univariable regression, and a higher Charlson Comorbidity Index score (of which age is included as a predictor) was associated with increased grade ≥3 ADRs in both univariable and multivariable regression. 2
Interestingly, sex was identified as an influential factor in the variability of imatinib Css,max, with no significant influence on other PK parameters. Previous studies in patients with CML have observed significantly higher imatinib Css,min in females compared to males, with authors highlighting differences in body weight and BSA as potential contributors. 67 , 81 , 82 Regression analysis from the observational study presented in Adattini et al 2 identified female sex as independently predictive of recurrent imatinib‐related grade ≥3 ADRs, which could be attributed to lower ABW (median 64.5 vs. 82 kg in males, p < 0.001). Although not accounted for in these PBPK simulations, sex differences in expression levels of hepatic P‐gp transporters may also play a role in altered imatinib PK, 59 however, the evidence is conflicting. 83
In line with previous studies, the simulated imatinib plasma concentration was sensitive to changes in AAG abundance. A reduction in AAG concentration leads to an increase in the unbound fraction of imatinib and a reduction in total plasma imatinib concentrations, due to the rapid distribution of unbound imatinib into extravascular space and increased total clearance. 77 , 84 , 85 , 86 , 87 , 88 , 89 , 90 Elevated AAG concentrations in patients have also been correlated with delayed response or resistance to imatinib treatment in CML, with studies suggesting that higher AAG concentrations are associated with reduced distribution into leukemic cells. 23 , 86 , 88 As discussed previously, AAG demonstrates highly variable concentrations, increasing 2‐ to 6‐fold in response to injury, infection, inflammation, and cancer. 22 , 23 Males have significantly higher mean AAG concentrations than females. 91 There are also inter‐ethnic differences in AAG concentrations, with individuals of Asian ancestry reported to have lower AAG concentrations than Europeans. 91 , 92 , 93 Additionally, drug–drug interactions can influence protein binding and subsequently the unbound concentration of a drug, as observed in interaction between clindamycin and imatinib. 85
Liver dysfunction was identified in these PBPK simulations as influential on imatinib PK, with increasing severities of liver dysfunction associated with increased imatinib systemic exposure. Similarly, Lankheet et al 10 observed a tendency toward higher imatinib plasma concentrations with decreasing liver function. A higher rate of imatinib‐related haematologic toxicities and more frequent dose reductions have also been observed in patients with pre‐existing liver dysfunction. 94 The decline in activity of CYP enzymes in enterocytes and hepatocytes is more pronounced as the severity of cirrhosis increases, 95 explaining the different magnitude of effects simulated on imatinib PK in patients with Child‐Pugh B/C cirrhosis and Child‐Pugh A cirrhosis. In advanced cirrhosis, kidney dysfunction is often also present, thereby reducing renal elimination. 96 Aside from PK changes, patients with cirrhosis can exhibit different pharmacological and toxicological responses to imatinib due to other pathophysiological changes. 96
The observed influence of kidney dysfunction on the PK variability of imatinib is supported in previous studies. A study by the National Cancer Institute Organ Dysfunction Working Group reported significantly higher imatinib exposure (dose‐normalized AUC0‐24,ss and Css,max), lower V/F and CL/F, and higher incidences of serious ADRs in patients with a creatinine clearance (CrCL) < 60 mL/min compared to normal kidney function. 88 Analogous to these findings, Tong et al 94 reported a higher rate of imatinib‐related hematological toxicities and more frequent dose reductions in patients with pre‐existing kidney dysfunction (CrCL <60 mL/min) compared to normal kidney function. Only a relatively small fraction of imatinib is excreted unchanged in the urine, 97 and hence decreased imatinib clearance in patients with kidney dysfunction is likely to be associated with altered tissue binding and plasma protein binding, the latter of which was accounted for in the virtual population with renal impairment during PBPK simulations by higher concentrations of plasma AAG. 98 , 99
Geographic ancestry is another factor that can contribute to inter‐individual variability in imatinib PK and outcomes, due to ethnic variations in the expression or activity of genes encoding for drug‐metabolizing enzymes, plasma protein binding and drug transporters, ethnic variation in complementary and herbal medicine use and dietary habits, and ethnic variation in body composition. 18 A Japanese study (n = 31) observed a mean Css,min of 1400 ± 570 ng mL−1 with imatinib 400 mg/day, and demonstrated that a lower imatinib dose of 300 mg/day was successful in maintaining a Css,min above the threshold of 1002 ng mL−1 (mean Css,min of 1150 ± SD 440 ng mL−1). 100 Similarly, a lower imatinib dose of 300 mg/day in a Chinese cohort (n = 16) was successful in maintaining concentrations required for MMR achievement, with a mean imatinib Css,min of 1034 ng mL−1 ± 576 ng mL−1. 73 European cohorts have reported lower systemic exposures with imatinib 400 mg/day, with an overall mean Css,min of 979 ± 530 ng mL−1 in the IRIS trial 8 and 1058 ± 557 ng mL−1 in a French study. 9 Interestingly, our retrospective study observed a lower hazard of recurrent imatinib‐related grade ≥3 ADRs in patients of European ancestry compared to East Asian ancestry. 2
In this study, a PBPK modeling approach was utilized to evaluate drug–drug interactions with imatinib and CYP modulators at steady‐state. The predicted drug–drug interaction effects between imatinib and verapamil, diltiazem and hyperforin (St John's Wort) at steady‐state were not clinically significant. Verapamil and diltiazem are inhibitors of CYP3A4 (via a mechanism‐based inhibition) and P‐gp, whilst hyperforin is an inducer of CYP3A4, CYP2C9, CYP2C19 and P‐gp. Repeat‐dose administration of imatinib results in autoinhibition of CYP3A4‐mediated metabolism, assigning a key role to other biotransformation pathways such as CYP2C8, and diminishing the effect of CYP3A4 modulators on imatinib at steady‐state. 42 , 101 In agreement with this, sensitivity analysis in this study demonstrated a negligible change in imatinib systemic exposure at steady‐state when CYP3A4 enzyme abundance in hepatocytes dropped from 93 pmol mg−1 protein (mean abundance in healthy Caucasian livers 57 ) to 0 pmol mg−1 protein (analogous to mechanism‐based inhibition of hepatic CYP3A4). Conversely, at steady‐state, imatinib is predicted to be more susceptible to drug interactions with inducers of hepatic CYP3A4. Significant decreases of imatinib Css,min have been observed in patients receiving CYP3A4 enzyme‐inducing anti‐epileptic drugs, such as carbamazepine and phenytoin. 10 , 102 However, since imatinib undergoes little to no metabolism in enterocytes, 62 it is unlikely that CYP3A inducers confined to intestinal enzymes (e.g., hyperforin) will affect the steady‐state CL/F of imatinib. This is reflected in the sensitivity analysis of CYP3A4 in this study, whereby a change in CYP3A4 enzyme abundance in enterocytes resulted in negligible changes to imatinib systemic exposure.
A PBPK‐based analysis provided a prediction of imatinib drug–drug interactions with CYP2C8 inhibitors, with simulations of repeat dosing of trimethoprim and clopidogrel (and its glucuronide metabolite) increasing the AUCss of imatinib by 23% and 32%, respectively. Clinical drug–drug interaction data with CYP2C8 inhibitors and imatinib at steady‐state is lacking, however, a candidate‐gene study of CYP2C8 supports the clinically significant effect of reduced CYP2C8 function on imatinib Css,min. 12 Whilst the dosing regimen and potency of the inhibitor determine the extent to which it affects systemic exposure of the victim drug, recent studies have demonstrated that genomic data may be used to estimate the magnitude of potential drug–drug interactions (and vice versa). 103 , 104 CYP2C8 inducers may also influence imatinib disposition, however clinical PK data are limited. Accounting for CYP2C8 induction in a PBPK model of carbamazepine (and its active metabolite carbamazepine‐10,11‐epoxide) improved the prediction of its interaction with imatinib at steady‐state, 34 highlighting the importance of accounting for CYP2C8‐based interactions with imatinib. Together with in vitro and in silico studies, 34 , 42 , 62 , 101 , 103 these simulations suggest that pharmacogenetic polymorphisms and drug–drug interactions affecting CYP2C8 activity may be another confounding factor on the total plasma imatinib concentration‐response relationship. CYP2C8 inhibition could be the mechanism behind the superior molecular response rates observed in patients treated with imatinib on concomitant potentially interacting medicines in the retrospective study. 2
TDM has been advocated as an integral part for dose optimisation of imatinib, given the high inter‐patient PK variability and established relationship between exposure and drug response (pharmacological effect and treatment‐related toxicities). 73 , 105 , 106 The PBPK modeling approach may help guide dose selection of imatinib for individual patients based on patient characteristics and has an advantage over the population pharmacokinetic counterpart in predicting the effect of comedications, particularly for previously unstudied/unidentified clinical interactions. Implementing PBPK model‐informed precision medicine in clinical practice involves the concept of matching the attributes of the real patient to those of his or her ‘virtual twin’, informed by covariates known to influence the drugs pharmacokinetics. 107 , 108 This approach may decrease variability in drug exposure via individualized dose adjustment. 108 Although the PBPK model of imatinib 34 may be useful to guide dose selection and optimisation of imatinib in real‐world clinical practice, further prospective PBPK model‐informed TDM studies are desirable to evaluate the robustness of the model.
A limitation of this research is that the PBPK model of imatinib used in these simulations did not include transporter mediated process in the apical/luminal membrane of enterocytes. 34 Therefore, there is a possibility that the drug–drug interaction simulations could have underestimated imatinib plasma‐concentration in the presence of P‐gp inhibitors. Another limitation is the inability to predict intracellular concentrations of imatinib. Imatinib has an intracellular site of action, therefore variability in imatinib disposition into target CML cells may have important implications on imatinib response. 7 In vitro studies have demonstrated reduced intracellular concentrations of imatinib, and reduced phosphorylation of the BCR‐ABL1 downstream effector phospho‐CRKL, in BCR‐ABL1 positive and P‐gp positive cells—a possible mechanism of imatinib resistance. 14 , 15 , 16 , 17 Further research is required to investigate the CML cell intracellular concentration‐response relationship. Another limitation of this study is that potential non‐adherence to imatinib treatment could not be accounted for. An assumption was made that patients took the prescribed dose 100% of the time leading up to the outcome of interest. Due to the prolonged (potentially life‐long) course of CML treatment, adherence is critical to achieving and maintaining adequate response. There is a clear correlation between adherence to TKI treatment and achieving optimal efficacy outcomes. 109 , 110 , 111 , 112 Studies in patients with CML reported as few as 14% to 33% of patients were highly adherent to their TKI treatment. 109 , 113 , 114 , 115 Therefore, poor adherence could be an additional factor contributing to the lack of achievement of molecular response outcomes in the retrospective observational study. 2 Furthermore, due to the lack of clinical drug–drug interaction studies of imatinib with trimethoprim, clopidogrel, verapamil and diltiazem, the PBPK simulated drug–drug interactions could not be verified with clinically observed concentrations. Further clinical studies are required to confirm the drug–drug interactions simulated in this study.
This study demonstrates the utility of PBPK modeling in providing a mechanistic framework that can extend our understanding of the inter‐individual variability in treatment outcomes of patients in routine daily clinical practice. We present new insights on the relationship between imatinib systemic exposure and imatinib treatment outcomes, and identify patient and medication‐related factors that could influence imatinib systemic exposure in real‐world populations. The outcomes of this study support the rationale for TDM to guide imatinib dosing to achieve optimal outcomes in patients with CML.
AUTHOR CONTRIBUTIONS
All authors conceived the study and contributed to the interpretation. J.A.A performed the simulation, analyzed the data and drafted the manuscript. J.A, A.S.G and A.J.M critically revised the manuscript. All authors approved the final manuscript.
FUNDING INFORMATION
This work was supported by the Peter Coates Postgraduate Scholarship in Ethnopharmacology provided by GlaxoSmithKline.
CONFLICT OF INTEREST STATEMENT
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
ETHICS STATEMENT
The real world clinical data included in this study was collected with the approval of the Sydney Local Health District Human Research Ethics Committee (reference number: LNR/17/CRGH/248).
Supporting information
Data S1: Supporting Information
ACKNOWLEDGMENTS
This work was supported by the Peter Coates Postgraduate Scholarship in Ethnopharmacology provided by GlaxoSmithKline. Certara UK Limited (Simcyp Division) is gratefully acknowledged for providing access to the Simcyp Simulator. The authors would like to thank Dr Manoranjenni Chetty for the training provided on using the Simcyp Simulator.
Adattini JA, Adiwidjaja J, Gross AS, McLachlan AJ. Application of physiologically based pharmacokinetic modeling to understand real‐world outcomes in patients receiving imatinib for chronic myeloid leukemia. Pharmacol Res Perspect. 2023;11:e01082. doi: 10.1002/prp2.1082
DATA AVAILABILITY STATEMENT
The data that support the findings of this study are available on request from the corresponding author. The data are not publicly available because of privacy or ethical restrictions.
REFERENCES
- 1. O'Brien SG, Guilhot F, Larson RA, et al. Imatinib compared with interferon and low‐dose cytarabine for newly diagnosed chronic‐phase chronic myeloid leukemia. N Engl J Med. 2003;348(11):994‐1004. doi: 10.1056/NEJMoa022457 [DOI] [PubMed] [Google Scholar]
- 2. Adattini JA, Gross AS, Wong Doo N, McLachlan AJ. Real‐world efficacy and safety outcomes of imatinib treatment in patients with chronic myeloid leukaemia: an Australian experience. Pharmacol Res Perspect. 2022;10(5):e01005. doi: 10.1002/prp2.1005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Hochhaus A, Larson RA, Guilhot F, et al. Long‐term outcomes of imatinib treatment for chronic myeloid leukemia. N Engl J Med. 2017;376(10):917‐927. doi: 10.1056/NEJMoa1609324 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Etienne G, Guilhot J, Rea D, et al. Long‐term follow‐up of the french stop imatinib (STIM1) study in patients with chronic myeloid leukemia. J Clin Oncol. 2017;35(3):298‐305. doi: 10.1200/jco.2016.68.2914 [DOI] [PubMed] [Google Scholar]
- 5. Marin D, Ibrahim AR, Lucas C, et al. Assessment of BCR‐ABL1 transcript levels at 3 months is the only requirement for predicting outcome for patients with chronic myeloid leukemia treated with tyrosine kinase inhibitors. J Clin Oncol. 2012;30(3):232‐238. doi: 10.1200/jco.2011.38.6565 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Kim DD, Lee H, Kamel‐Reid S, Lipton JH. BCR‐ABL1 transcript at 3 months predicts long‐term outcomes following second generation tyrosine kinase inhibitor therapy in the patients with chronic myeloid leukaemia in chronic phase who failed imatinib. Br J Haematol. 2013;160(5):630‐639. doi: 10.1111/bjh.12187 [DOI] [PubMed] [Google Scholar]
- 7. Barratt D, Somogyi A. Role of pharmacogenetics in personalised imatinib dosing. Transl Cancer Res. 2017;6(Suppl 10):S1541‐S1557. [Google Scholar]
- 8. Larson RA, Druker BJ, Guilhot F, et al. Imatinib pharmacokinetics and its correlation with response and safety in chronic‐phase chronic myeloid leukemia: a subanalysis of the IRIS study. Blood. 2008;111(8):4022‐4028. doi: 10.1182/blood-2007-10-116475 [DOI] [PubMed] [Google Scholar]
- 9. Picard S, Titier K, Etienne G, et al. Trough imatinib plasma levels are associated with both cytogenetic and molecular responses to standard‐dose imatinib in chronic myeloid leukemia. Blood. 2007;109(8):3496‐3499. doi: 10.1182/blood-2006-07-036012 [DOI] [PubMed] [Google Scholar]
- 10. Lankheet NA, Knapen LM, Schellens JH, Beijnen JH, Steeghs N, Huitema AD. Plasma concentrations of tyrosine kinase inhibitors imatinib, erlotinib, and sunitinib in routine clinical outpatient cancer care. Ther Drug Monit. 2014;36(3):326‐334. [DOI] [PubMed] [Google Scholar]
- 11. Bouchet S, Titier K, Moore N, et al. Therapeutic drug monitoring of imatinib in chronic myeloid leukemia: experience from 1216 patients at a centralized laboratory. Fundam Clin Pharmacol. 2013;27(6):690‐697. doi: 10.1111/fcp.12007 [DOI] [PubMed] [Google Scholar]
- 12. Barratt DT, Cox HK, Menelaou A, et al. CYP2C8 genotype significantly alters imatinib metabolism in chronic myeloid leukaemia patients. Clin Pharmacokinet. 2017;56(8):977‐985. doi: 10.1007/s40262-016-0494-0 [DOI] [PubMed] [Google Scholar]
- 13. Whirl‐Carrillo M, McDonagh EM, Hebert JM, et al. Pharmacogenomics knowledge for personalized medicine. Clin Pharmacol Ther. 2012;92(4):414‐417. doi: 10.1038/clpt.2012.96 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Illmer T, Schaich M, Platzbecker U, et al. P‐glycoprotein‐mediated drug efflux is a resistance mechanism of chronic myelogenous leukemia cells to treatment with imatinib mesylate. Leukemia. 2004;18(3):401‐408. doi: 10.1038/sj.leu.2403257 [DOI] [PubMed] [Google Scholar]
- 15. Mahon FX, Deininger MW, Schultheis B, et al. Selection and characterization of BCR‐ABL positive cell lines with differential sensitivity to the tyrosine kinase inhibitor STI571: diverse mechanisms of resistance. Blood. 2000;96(3):1070‐1079. [PubMed] [Google Scholar]
- 16. Rumpold H, Wolf AM, Gruenewald K, Gastl G, Gunsilius E, Wolf D. RNAi‐mediated knockdown of P‐glycoprotein using a transposon‐based vector system durably restores imatinib sensitivity in imatinib‐resistant CML cell lines. Exp Hematol. 2005;33(7):767‐775. doi: 10.1016/j.exphem.2005.03.014 [DOI] [PubMed] [Google Scholar]
- 17. Widmer N, Rumpold H, Untergasser G, Fayet A, Buclin T, Decosterd LA. Resistance reversal by RNAi silencing of MDR1 in CML cells associated with increase in imatinib intracellular levels. Leukemia. 2007;21(7):1561‐1562. doi: 10.1038/sj.leu.2404671 [DOI] [PubMed] [Google Scholar]
- 18. Touma JA, McLachlan AJ, Gross AS. The role of ethnicity in personalized dosing of small molecule tyrosine kinase inhibitors used in oncology. Transl Cancer Res. 2017;6(Suppl 10):S1558‐S1591. doi: 10.21037/tcr.2017.09.09 [DOI] [Google Scholar]
- 19. Gora‐Tybor J, Sacha T, Waclaw J, et al. Comorbidity burden and use of concomitant medications at CML diagnosis: a retrospective analysis of 470 patients from the polish adult leukemia group (PALG) registry. Blood. 2016;128(22):1909. doi: 10.1182/blood.V128.22.1909.1909 [DOI] [PubMed] [Google Scholar]
- 20. Saussele S, Krauss MP, Hehlmann R, et al. Impact of comorbidities on overall survival in patients with chronic myeloid leukemia: results of the randomized CML study IV. Blood. 2015;126(1):42‐49. doi: 10.1182/blood-2015-01-617993 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Iurlo A, Nobili A, Latagliata R, et al. Imatinib and polypharmacy in very old patients with chronic myeloid leukemia: effects on response rate, toxicity and outcome. Oncotarget. 2016;7(48):80083‐80090. doi: 10.18632/oncotarget.11657 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Smith SA, Waters NJ. Pharmacokinetic and pharmacodynamic considerations for drugs binding to alpha‐1‐acid glycoprotein. Pharm Res. 2018;36:30. doi: 10.1007/s11095-018-2551-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. le Coutre P, Kreuzer KA, Na IK, et al. Determination of alpha‐1 acid glycoprotein in patients with Ph+ chronic myeloid leukemia during the first 13 weeks of therapy with STI571. Blood Cells Mol Dis. 2002;28(1):75‐85. doi: 10.1006/bcmd.2002.0493 [DOI] [PubMed] [Google Scholar]
- 24. Fitos I, Visy J, Zsila F, Mády G, Simonyi M. Selective binding of imatinib to the genetic variants of human alpha1‐acid glycoprotein. Biochim Biophys Acta. 2006;1760(11):1704‐1712. doi: 10.1016/j.bbagen.2006.08.015 [DOI] [PubMed] [Google Scholar]
- 25. Zhao P, Zhang L, Grillo JA, et al. Applications of physiologically based pharmacokinetic (PBPK) modeling and simulation during regulatory review. Clin Pharmacol Ther. 2011;89(2):259‐267. doi: 10.1038/clpt.2010.298 [DOI] [PubMed] [Google Scholar]
- 26. Sinha V, Zhao P, Huang SM, Zineh I. Physiologically based pharmacokinetic modeling: from regulatory science to regulatory policy. Clin Pharmacol Ther. 2014;95(5):478‐480. doi: 10.1038/clpt.2014.46 [DOI] [PubMed] [Google Scholar]
- 27. Wagner C, Pan Y, Hsu V, et al. Predicting the effect of cytochrome P450 inhibitors on substrate drugs: analysis of physiologically based pharmacokinetic modeling submissions to the US Food and Drug Administration. Clin Pharmacokinet. 2015;54(1):117‐127. doi: 10.1007/s40262-014-0188-4 [DOI] [PubMed] [Google Scholar]
- 28. Wagner C, Pan Y, Hsu V, Sinha V, Zhao P. Predicting the effect of CYP3A inducers on the pharmacokinetics of substrate drugs using physiologically based pharmacokinetic (PBPK) modeling: an analysis of PBPK submissions to the US FDA. Clin Pharmacokinet. 2016;55(4):475‐483. doi: 10.1007/s40262-015-0330-y [DOI] [PubMed] [Google Scholar]
- 29. Wagner C, Zhao P, Pan Y, et al. Application of physiologically based pharmacokinetic (PBPK) modeling to support dose selection: report of an FDA public workshop on PBPK. CPT Pharmacometrics Syst Pharmacol. 2015;4(4):226‐230. doi: 10.1002/psp4.33 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Samant TS, Huth F, Umehara K, et al. Ribociclib drug‐drug interactions: clinical evaluations and physiologically‐based pharmacokinetic modeling to guide drug labeling. Clin Pharmacol Ther. 2020;108(3):575‐585. doi: 10.1002/cpt.1950 [DOI] [PubMed] [Google Scholar]
- 31. Rowland A, van Dyk M, Hopkins AM, et al. Physiologically based pharmacokinetic modeling to identify physiological and molecular characteristics driving variability in drug exposure. Clin Pharmacol Ther. 2018;104(6):1219‐1228. doi: 10.1002/cpt.1076 [DOI] [PubMed] [Google Scholar]
- 32. Emoto C, Johnson TN, Hahn D, et al. A theoretical physiologically‐based pharmacokinetic approach to ascertain covariates explaining the large interpatient variability in tacrolimus disposition. CPT Pharmacometrics Syst Pharmacol. 2019;8(5):273‐284. doi: 10.1002/psp4.12392 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Jamei M, Marciniak S, Edwards D, et al. The simcyp population based simulator: architecture, implementation, and quality assurance. In Silico Pharmacol. 2013;1:9. doi: 10.1186/2193-9616-1-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Adiwidjaja J, Boddy AV, McLachlan AJ. Implementation of a physiologically based pharmacokinetic modeling approach to guide optimal dosing regimens for imatinib and potential drug interactions in paediatrics. Front Pharmacol. 2020;10:1672. doi: 10.3389/fphar.2019.01672 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Adiwidjaja J, Gross AS, Boddy AV, McLachlan AJ. Physiologically‐based pharmacokinetic model predictions of inter‐ethnic differences in imatinib pharmacokinetics and dosing regimens. Br J Clin Pharmacol. 2022;88(4):1735‐1750. [DOI] [PubMed] [Google Scholar]
- 36. Smith P, Bullock JM, Booker BM, Haas CE, Berenson CS, Jusko WJ. The influence of St. John's wort on the pharmacokinetics and protein binding of imatinib mesylate. Pharmacotherapy. 2004;24(11):1508‐1514. [DOI] [PubMed] [Google Scholar]
- 37. Filppula AM, Tornio A, Niemi M, Neuvonen PJ, Backman JT. Gemfibrozil impairs imatinib absorption and inhibits the CYP2C8‐mediated formation of its main metabolite. Clin Pharmacol Ther. 2013;94(3):383‐393. [DOI] [PubMed] [Google Scholar]
- 38. Fraeyman NF, Dello CD, Belpaire FM. Alpha 1‐acid glycoprotein concentration and molecular heterogeneity: relationship to oxprenolol binding in serum from healthy volunteers and patients with lung carcinoma or cirrhosis. Br J Clin Pharmacol. 1988;25(6):733‐740. doi: 10.1111/j.1365-2125.1988.tb05260.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Al‐Dewik NI, Jewell AP, Yassin MA, Morsi HM. Studying the impact of presence of alpha acid glycoprotein and protein glycoprotein in chronic myeloid leukemia patients treated with imatinib mesylate in the State of Qatar. Biomark Cancer. 2015;7:63‐67. doi: 10.4137/bic.s31427 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Jorgensen HG, Elliott MA, Allan EK, Carr CE, Holyoake TL, Smith KD. Alpha1‐acid glycoprotein expressed in the plasma of chronic myeloid leukemia patients does not mediate significant in vitro resistance to STI571. Blood. 2002;99(2):713‐715. [DOI] [PubMed] [Google Scholar]
- 41. Gandia P, Arellano C, Lafont T, Huguet F, Malard L, Chatelut E. Should therapeutic drug monitoring of the unbound fraction of imatinib and its main active metabolite N‐desmethyl‐imatinib be developed? Cancer Chemother Pharmacol. 2013;71(2):531‐536. doi: 10.1007/s00280-012-2035-3 [DOI] [PubMed] [Google Scholar]
- 42. Filppula AM, Neuvonen M, Laitila J, Neuvonen PJ, Backman JT. Autoinhibition of CYP3A4 leads to important role of CYP2C8 in imatinib metabolism: variability in CYP2C8 activity may alter plasma concentrations and response. Drug Metab Dispos. 2013;41(1):50‐59. doi: 10.1124/dmd.112.048017 [DOI] [PubMed] [Google Scholar]
- 43. Rowland‐Yeo K, Jamei M, Yang J, Tucker GT, Rostami‐Hodjegan A. Physiologically based mechanistic modelling to predict complex drug‐drug interactions involving simultaneous competitive and time‐dependent enzyme inhibition by parent compound and its metabolite in both liver and gut ‐ the effect of diltiazem on the time‐course of exposure to triazolam. Eur J Pharm Sci. 2010;39(5):298‐309. [DOI] [PubMed] [Google Scholar]
- 44. Dai H, Marbach P, Lemaire M, Hayes M, Elmquist WF. Distribution of STI‐571 to the brain is limited by P‐glycoprotein‐mediated efflux. J Pharmacol Exp Ther. 2003;304(3):1085‐1092. doi: 10.1124/jpet.102.045260 [DOI] [PubMed] [Google Scholar]
- 45. Breedveld P, Pluim D, Cipriani G, et al. The effect of Bcrp1 (Abcg2) on the in vivo pharmacokinetics and brain penetration of imatinib mesylate (Gleevec): implications for the use of breast cancer resistance protein and Pglycoprotein inhibitors to enable the brain penetration of imatinib in patients. Cancer Res. 2005;65(7):2577‐2582. doi: 10.1158/0008-5472.CAN-04-2416 [DOI] [PubMed] [Google Scholar]
- 46. Bornhauser M, Pursche S, Bonin M, et al. Elimination of imatinib mesylate and its metabolite N‐desmethylimatinib. J Clin Oncol. 2005;23(16):3855‐3856. author reply 3857‐3858. doi: 10.1200/JCO.2005.05.246 [DOI] [PubMed] [Google Scholar]
- 47. Deininger MW, Shah NP, Altman JK, et al. Chronic myeloid leukemia, version 2.2021, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2020;18(10):1385‐1415. doi: 10.6004/jnccn.2020.0047 [DOI] [PubMed] [Google Scholar]
- 48. National Cancer Institute (U.S.) . Common Terminology Criteria for Adverse Events (CTCAE) v5.0. U.S. Department of Health and Human Services; 2017. https://ctep.cancer.gov/protocoldevelopment/electronic_applications/ctc.htm [Google Scholar]
- 49. Levey AS, Stevens LA, Schmid CH, et al. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150(9):604‐612. doi: 10.7326/0003-4819-150-9-200905050-00006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Wang HY, Chen X, Jiang J, Shi J, Hu P. Evaluating a physiologically based pharmacokinetic model for predicting the pharmacokinetics of midazolam in Chinese after oral administration. Acta Pharmacol Sin. 2016;37(2):276‐284. doi: 10.1038/aps.2015.122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Matsumoto Y, Cabalu T, Sandhu P, et al. Application of physiologically based pharmacokinetic modeling to predict pharmacokinetics in healthy Japanese subjects. Clin Pharmacol Ther. 2019;105(4):1018‐1030. doi: 10.1002/cpt.1240 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Takahashi N, Wakita H, Miura M, et al. Correlation between imatinib pharmacokinetics and clinical response in Japanese patients with chronic‐phase chronic myeloid leukemia. Clin Pharmacol Ther. 2010;88(6):809‐813. doi: 10.1038/clpt.2010.186 [DOI] [PubMed] [Google Scholar]
- 53. R: A language and environment for statistical computing . http://www.R‐project.org/
- 54. McNally K, Cotton R, Loizou GD. A workflow for global sensitivity analysis of PBPK models. Front Pharmacol. 2011;2:31. doi: 10.3389/fphar.2011.00031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Zhang XY, Trame MN, Lesko LJ, Schmidt S. Sobol sensitivity analysis: a tool to guide the development and evaluation of systems pharmacology models. CPT Pharmacometrics Syst Pharmacol. 2015;4(2):69‐79. doi: 10.1002/psp4.6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Johnson TN, Boussery K, Rowland‐Yeo K, Tucker GT, Rostami‐Hodjegan A. A semi‐mechanistic model to predict the effects of liver cirrhosis on drug clearance. Clin Pharmacokinet. 2010;49(3):189‐206. doi: 10.2165/11318160-000000000-00000 [DOI] [PubMed] [Google Scholar]
- 57. Achour B, Barber J, Rostami‐Hodjegan A. Expression of hepatic drug‐metabolizing cytochrome P450 enzymes and their intercorrelations: a meta‐analysis. Drug Metab Dispos. 2014;42(8):1349‐1356. doi: 10.1124/dmd.114.058834 [DOI] [PubMed] [Google Scholar]
- 58. Blain PG, Mucklow JC, Rawlins MD, Roberts DF, Routledge PA, Shand DG. Determinants of plasma alpha 1‐acid glycoprotein (AAG) concentrations in health. Br J Clin Pharmacol. 1985;20(5):500‐502. doi: 10.1111/j.1365-2125.1985.tb05107.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Burt HJ, Riedmaier AE, Harwood MD, Crewe HK, Gill KL, Neuhoff S. Abundance of hepatic transporters in Caucasians: a meta‐analysis. Drug Metab Dispos. 2016;44(10):1550‐1561. doi: 10.1124/dmd.116.071183 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Drozdzik M, Szelag‐Pieniek S, Post M, et al. Protein abundance of hepatic drug transporters in patients with different forms of liver damage. Clin Pharmacol Ther. 2020;107(5):1138‐1148. doi: 10.1002/cpt.1717 [DOI] [PubMed] [Google Scholar]
- 61. Shebley M, Fu W, Badri P, Bow D, Fischer V. Physiologically based pharmacokinetic modeling suggests limited drug‐drug interaction between clopidogrel and dasabuvir. Clin Pharmacol Ther. 2017;102(4):679‐687. doi: 10.1002/cpt.689 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Adiwidjaja J, Boddy AV, McLachlan AJ. Physiologically based pharmacokinetic modelling of hyperforin to predict drug interactions with St John's wort. Clin Pharmacokinet. 2019;58(7):911‐926. doi: 10.1007/s40262-019-00736-6 [DOI] [PubMed] [Google Scholar]
- 63. Center for Drug Evaluation and Research . Clinical drug interaction studies —cytochrome P450 enzyme and transporter‐mediated drug interactions guidance for industry. Food and Drug Administration. Updated January 01, 2020. Accessed November 20, 2020. https://www.fda.gov/regulatory‐information/search‐fda‐guidance‐documents/clinical‐drug‐interaction‐studies‐cytochrome‐p450‐enzyme‐and‐transporter‐mediated‐drug‐interactions [Google Scholar]
- 64. Harding SD, Sharman JL, Faccenda E, et al. The IUPHAR/BPS guide to PHARMACOLOGY in 2019: updates and expansion to encompass the new guide to IMMUNOPHARMACOLOGY. Nucleic Acids Res. 2018;46:D1091‐D1106. doi: 10.1093/nar/gkx1121 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Alexander SPH, Kelly E, Mathie A, et al. THE CONCISE GUIDE TO PHARMACOLOGY 2021/22: introduction and other protein targets. Br J Pharmacol. 2021;178:S1‐S26. doi: 10.1111/bph.15537 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Widmer N, Decosterd LA, Leyvraz S, et al. Relationship of imatinib‐free plasma levels and target genotype with efficacy and tolerability. Br J Cancer. 2008;98(10):1633‐1640. doi: 10.1038/sj.bjc.6604355 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Guilhot F, Hughes TP, Cortes J, et al. Plasma exposure of imatinib and its correlation with clinical response in the tyrosine kinase inhibitor optimization and selectivity trial. Haematologica. 2012;97(5):731‐738. doi: 10.3324/haematol.2011.045666 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Malhotra H, Sharma P, Bhargava S, Rathore OS, Malhotra B, Kumar M. Correlation of plasma trough levels of imatinib with molecular response in patients with chronic myeloid leukemia. Leuk Lymphoma. 2014;55(11):2614‐2619. doi: 10.3109/10428194.2014.885515 [DOI] [PubMed] [Google Scholar]
- 69. Singh N, Kumar L, Meena R, Velpandian T. Drug monitoring of imatinib levels in patients undergoing therapy for chronic myeloid leukaemia: comparing plasma levels of responders and non‐responders. Eur J Clin Pharmacol. 2009;65(6):545‐549. doi: 10.1007/s00228-009-0621-z [DOI] [PubMed] [Google Scholar]
- 70. Awidi A, Ayed AO, Bsoul N, et al. Relationship of serum imatinib trough level and response in CML patients: long term follow‐up. Leuk Res. 2010;34(12):1573‐1575. doi: 10.1016/j.leukres.2010.07.014 [DOI] [PubMed] [Google Scholar]
- 71. Ishikawa Y, Kiyoi H, Watanabe K, et al. Trough plasma concentration of imatinib reflects BCR‐ABL kinase inhibitory activity and clinical response in chronic‐phase chronic myeloid leukemia: a report from the BINGO study. Cancer Sci. 2010;101(10):2186‐2192. doi: 10.1111/j.1349-7006.2010.01643.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Golabchifar AA, Rezaee S, Ghavamzadeh A, Alimoghaddam K, Dinan NM, Rouini MR. Population pharmacokinetics of imatinib in Iranian patients with chronic‐phase chronic myeloid leukemia. Cancer Chemother Pharmacol. 2014;74(1):85‐93. doi: 10.1007/s00280-014-2473-1 [DOI] [PubMed] [Google Scholar]
- 73. Cheng F, Zeng F, Li Q, et al. Imatinib dose optimization based on therapeutic drug monitoring in Chinese patients with chronic‐phase chronic myeloid leukemia. Cancer. 2022. Nov 15;128(22):3951‐3958. doi: 10.1002/cncr.34478 [DOI] [PubMed] [Google Scholar]
- 74. Francis J, Dubashi B, Sundaram R, Pradhan SC, Chandrasekaran A. A study to explore the correlation of ABCB1, ABCG2, OCT1 genetic polymorphisms and trough level concentration with imatinib mesylate‐induced thrombocytopenia in chronic myeloid leukemia patients. Cancer Chemother Pharmacol. 2015;76(6):1185‐1189. doi: 10.1007/s00280-015-2905-6 [DOI] [PubMed] [Google Scholar]
- 75. Judson I, Ma P, Peng B, et al. Imatinib pharmacokinetics in patients with gastrointestinal stromal tumour: a retrospective population pharmacokinetic study over time. EORTC soft tissue and bone sarcoma group. Cancer Chemother Pharmacol. 2005;55(4):379‐386. doi: 10.1007/s00280-004-0876-0 [DOI] [PubMed] [Google Scholar]
- 76. Schmidli H, Peng B, Riviere GJ, et al. Population pharmacokinetics of imatinib mesylate in patients with chronic‐phase chronic myeloid leukaemia: results of a phase III study. Br J Clin Pharmacol. 2005;60(1):35‐44. doi: 10.1111/j.1365-2125.2005.02372.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Widmer N, Decosterd LA, Csajka C, et al. Population pharmacokinetics of imatinib and the role of alpha‐acid glycoprotein. Br J Clin Pharmacol. 2006;62(1):97‐112. doi: 10.1111/j.1365-2125.2006.02719.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Menon‐Andersen D, Mondick JT, Jayaraman B, et al. Population pharmacokinetics of imatinib mesylate and its metabolite in children and young adults. Cancer Chemother Pharmacol. 2009;63(2):229‐238. doi: 10.1007/s00280-008-0730-x [DOI] [PubMed] [Google Scholar]
- 79. Sakai M, Miyazaki Y, Matsuo E, et al. Long‐term efficacy of imatinib in a practical setting is correlated with imatinib trough concentration that is influenced by body size: a report by the Nagasaki CML study group. Int J Hematol. 2009;89(3):319‐325. doi: 10.1007/s12185-009-0263-z [DOI] [PubMed] [Google Scholar]
- 80. Ohnishi K, Nakaseko C, Takeuchi J, et al. Long‐term outcome following imatinib therapy for chronic myelogenous leukemia, with assessment of dosage and blood levels: the JALSG CML202 study. Cancer Sci. 2012;103(6):1071‐1078. doi: 10.1111/j.1349-7006.2012.02253.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Belsey SL, Ireland R, Lang K, et al. Women administered standard dose imatinib for chronic myeloid leukemia have higher dose‐adjusted plasma imatinib and norimatinib concentrations than men. Ther Drug Monit. 2017;39(5):499‐504. doi: 10.1097/ftd.0000000000000440 [DOI] [PubMed] [Google Scholar]
- 82. Li QB, Chen C, Chen ZC, et al. Imatinib plasma trough concentration and its correlation with characteristics and response in Chinese CML patients. Acta Pharmacol Sin. 2010;31(8):999‐1004. doi: 10.1038/aps.2010.79 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Prasad B, Evers R, Gupta A, et al. Interindividual variability in hepatic organic anion‐transporting polypeptides and P‐glycoprotein (ABCB1) protein expression: quantification by liquid chromatography tandem mass spectroscopy and influence of genotype, age, and sex. Drug Metab Dispos. 2014;42(1):78‐88. doi: 10.1124/dmd.113.053819 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Bins S, Eechoute K, Kloth JS, et al. Prospective analysis in GIST patients on the role of alpha‐1 acid glycoprotein in imatinib exposure. Clin Pharmacokinet. 2017;56(3):305‐310. doi: 10.1007/s40262-016-0441-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Gambacorti‐Passerini C, Zucchetti M, Russo D, et al. Alpha1 acid glycoprotein binds to imatinib (STI571) and substantially alters its pharmacokinetics in chronic myeloid leukemia patients. Clin Cancer Res. 2003;9(2):625‐632. [PubMed] [Google Scholar]
- 86. Zhong JS, Meng FY, Xu D, Dai M, Wei YQ, Zhou HS. Serum alpha1‐acid glycoprotein, imatinib concentration and efficacy in chronic myeloid leukemia patients. Zhonghua Yi Xue Za Zhi. 2011;91(30):2120‐2123. [PubMed] [Google Scholar]
- 87. Delbaldo C, Chatelut E, Ré M, et al. Pharmacokinetic‐pharmacodynamic relationships of imatinib and its main metabolite in patients with advanced gastrointestinal stromal tumors. Clin Cancer Res. 2006;12:6073‐6078. doi: 10.1158/1078-0432.CCR-05-2596 [DOI] [PubMed] [Google Scholar]
- 88. Kretz O, Weiss HM, Schumacher MM, Gross G. In vitro blood distribution and plasma protein binding of the tyrosine kinase inhibitor imatinib and its active metabolite, CGP74588, in rat, mouse, dog, monkey, healthy humans and patients with acute lymphatic leukaemia. Br J Clin Pharmacol. 2004;58(2):212‐216. doi: 10.1111/j.1365-2125.2004.02117.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Gibbons J, Egorin MJ, Ramanathan RK, et al. Phase I and pharmacokinetic study of imatinib mesylate in patients with advanced malignancies and varying degrees of renal dysfunction: a study by the National Cancer Institute organ dysfunction working group. J Clin Oncol. 2008;26(4):570‐576. doi: 10.1200/jco.2007.13.3819 [DOI] [PubMed] [Google Scholar]
- 90. Gotta V, Buclin T, Csajka C, Widmer N. Systematic review of population pharmacokinetic analyses of imatinib and relationships with treatment outcomes. Ther Drug Monit. 2013;35(2):150‐167. doi: 10.1097/FTD.0b013e318284ef11 [DOI] [PubMed] [Google Scholar]
- 91. Colombo S, Buclin T, Décosterd LA, et al. Orosomucoid (alpha1‐acid glycoprotein) plasma concentration and genetic variants: effects on human immunodeficiency virus protease inhibitor clearance and cellular accumulation. Clin Pharmacol Ther. 2006;80(4):307‐318. doi: 10.1016/j.clpt.2006.06.006 [DOI] [PubMed] [Google Scholar]
- 92. Zhou HH, Adedoyin A, Wilkinson GR. Differences in plasma binding of drugs between Caucasians and Chinese subjects. Clin Pharmacol Ther. 1990;48(1):10‐17. doi: 10.1038/clpt.1990.111 [DOI] [PubMed] [Google Scholar]
- 93. Feely J, Grimm T. A comparison of drug protein binding and alpha 1‐acid glycoprotein concentration in Chinese and Caucasians. Br J Clin Pharmacol. 1991;31(5):551‐552. doi: 10.1111/j.1365-2125.1991.tb05579.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Tong WG, Kantarjian H, O'Brien S, et al. Imatinib front‐line therapy is safe and effective in patients with chronic myelogenous leukemia with pre‐existing liver and/or renal dysfunction. Cancer. 2010;116(13):3152‐3159. doi: 10.1002/cncr.25071 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Verbeeck RK. Pharmacokinetics and dosage adjustment in patients with hepatic dysfunction. Eur J Clin Pharmacol. 2008;64(12):1147‐1161. doi: 10.1007/s00228-008-0553-z [DOI] [PubMed] [Google Scholar]
- 96. Weersink RA, Burger DM, Hayward KL, Taxis K, Drenth JP, Borgsteede SD. Safe use of medication in patients with cirrhosis: pharmacokinetic and pharmacodynamic considerations. Expert Opin Drug Metab Toxicol. 2020;16(1):45‐57. doi: 10.1080/17425255.2020.1702022 [DOI] [PubMed] [Google Scholar]
- 97. Peng B, Lloyd P, Schran H. Clinical pharmacokinetics of imatinib. Clin Pharmacokinet. 2005;44(9):879‐894. doi: 10.2165/00003088-200544090-00001 [DOI] [PubMed] [Google Scholar]
- 98. Pichette V, Leblond FA. Drug metabolism in chronic renal failure. Curr Drug Metab. 2003;4(2):91‐103. doi: 10.2174/1389200033489532 [DOI] [PubMed] [Google Scholar]
- 99. Rowland‐Yeo K, Aarabi M, Jamei M, Rostami‐Hodjegan A. Modeling and predicting drug pharmacokinetics in patients with renal impairment. Expert Rev Clin Pharmacol. 2011;4(2):261‐274. doi: 10.1586/ecp.10.143 [DOI] [PubMed] [Google Scholar]
- 100. Kawaguchi T, Hamada A, Hirayama C, et al. Relationship between an effective dose of imatinib, body surface area, and trough drug levels in patients with chronic myeloid leukemia. Int J Hematol. 2009;89(5):642‐648. doi: 10.1007/s12185-009-0315-4 [DOI] [PubMed] [Google Scholar]
- 101. van Erp NP, Gelderblom H, Karlsson MO, et al. Influence of CYP3A4 inhibition on the steady‐state pharmacokinetics of imatinib. Clin Cancer Res. 2007;13(24):7394‐7400. doi: 10.1158/1078-0432.Ccr-07-0346 [DOI] [PubMed] [Google Scholar]
- 102. Pursche S, Schleyer E, von Bonin M, et al. Influence of enzyme‐inducing antiepileptic drugs on trough level of imatinib in glioblastoma patients. Curr Clin Pharmacol. 2008;3(3):198‐203. doi: 10.2174/157488408785747656 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Lagishetty CV, Deng J, Lesko LJ, Rogers H, Pacanowski M, Schmidt S. How informative are drug‐drug interactions of gene‐drug interactions? J Clin Pharmacol. 2016;56(10):1221‐1231. doi: 10.1002/jcph.743 [DOI] [PubMed] [Google Scholar]
- 104. Filppula AM, Laitila J, Neuvonen PJ, Backman JT. Potent mechanism‐based inhibition of CYP3A4 by imatinib explains its liability to interact with CYP3A4 substrates. Br J Pharmacol. 2012;165(8):2787‐2798. doi: 10.1111/j.1476-5381.2011.01732.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. IJzerman NS, Groenland SL, Koenen AM, et al. Therapeutic drug monitoring of imatinib in patients with gastrointestinal stromal tumours ‐ results from daily clinical practice. Eur J Cancer. 2020;136:140‐148. doi: 10.1016/j.ejca.2020.05.025. Epub 2020 Jul 17 PMID: 32688207. [DOI] [PubMed] [Google Scholar]
- 106. Westerdijk K, Desar IME, Steeghs N, van der Graaf WTA, van Erp NP, Dutch Pharmacology and Oncology Group (DPOG) . Imatinib, sunitinib and pazopanib: from flat‐fixed dosing towards a pharmacokinetically guided personalized dose. Br J Clin Pharmacol. 2020;86(2):258‐273. doi: 10.1111/bcp.14185 Epub 2020 Jan 21. PMID: 31782166; PMCID: PMC7015742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Polasek TM, Rostami‐Hodjegan A. Virtual twins: understanding the data required for model‐informed precision dosing. Clin Pharmacol Ther. 2020. Apr;107(4):742‐745. doi: 10.1002/cpt.1778 [DOI] [PubMed] [Google Scholar]
- 108. Polasek TM, Tucker GT, Sorich MJ, et al. Prediction of olanzapine exposure in individual patients using physiologically based pharmacokinetic modelling and simulation. Br J Clin Pharmacol. 2018. Mar;84(3):462‐476. doi: 10.1111/bcp.13480 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Marin D, Bazeos A, Mahon FX, et al. Adherence is the critical factor for achieving molecular responses in patients with chronic myeloid leukemia who achieve complete cytogenetic responses on imatinib. J Clin Oncol. 2010;28(14):2381‐2388. doi: 10.1200/jco.2009.26.3087 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Ganesan P, Sagar TG, Dubashi B, et al. Nonadherence to imatinib adversely affects event free survival in chronic phase chronic myeloid leukemia. Am J Hematol. 2011;86(6):471‐474. doi: 10.1002/ajh.22019 [DOI] [PubMed] [Google Scholar]
- 111. Ibrahim AR, Eliasson L, Apperley JF, et al. Poor adherence is the main reason for loss of CCyR and imatinib failure for chronic myeloid leukemia patients on long‐term therapy. Blood. 2011;117(14):3733‐3736. doi: 10.1182/blood-2010-10-309807 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Yoshida C, Komeno T, Hori M, et al. Adherence to the standard dose of imatinib, rather than dose adjustment based on its plasma concentration, is critical to achieve a deep molecular response in patients with chronic myeloid leukemia. Int J Hematol. 2011;93(5):618‐623. doi: 10.1007/s12185-011-0838-3 [DOI] [PubMed] [Google Scholar]
- 113. Geissler J, Sharf G, Bombaci F, et al. Factors influencing adherence in CML and ways to improvement: results of a patient‐driven survey of 2546 patients in 63 countries. J Cancer Res Clin Oncol. 2017;143(7):1167‐1176. doi: 10.1007/s00432-017-2372-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Noens L, van Lierde MA, De Bock R, et al. Prevalence, determinants, and outcomes of nonadherence to imatinib therapy in patients with chronic myeloid leukemia: the ADAGIO study. Blood. 2009;113(22):5401‐5411. doi: 10.1182/blood-2008-12-196543 [DOI] [PubMed] [Google Scholar]
- 115. Kapoor J, Agrawal N, Ahmed R, Sharma SK, Gupta A, Bhurani D. Factors influencing adherence to imatinib in Indian chronic myeloid leukemia patients: a cross‐sectional study. Mediterr J Hematol Infect Dis. 2015;7(1):e2015013. doi: 10.4084/MJHID.2015.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data S1: Supporting Information
Data Availability Statement
The data that support the findings of this study are available on request from the corresponding author. The data are not publicly available because of privacy or ethical restrictions.
