Foretinib

JOURNAL OF CHROMATOGRAPHY B 

Simultaneous quantitative determination of seven novel tyrosine kinase inhib￾itors in plasma by a validated UPLC-MS/MS method and its application to
human microsomal metabolic stability study
ABSTRACT
A rapid, sensitive and reproducible ultra-high performance liquid chromatography mass
spectrometry method was developed and validated for simultaneous determination of seven
tyrosine kinase inhibitors (dasatinib, foretinib, osimertinib, gefitinib, ibrutinib, linifanib and
motesanib) in human plasma samples using quizartinib as internal standard (IS). The sample
preparation was performed by liquid-liquid extraction method, using a mixture of ethyl acetate
and tert butyl methyl ether (50:50, v/v) as extracting solvents. Chromatographic separation was
achieved using acquity UPLC BEH C18, 1.7 µm 2.1 X 100 mm column and a mobile phase
consisting of a mixture of acetonitrile (0.1% formic acid) and 20 mM ammonium acetate (95:5)
at a flow rate of 0.25 ml/min. All the analytes and IS were eluted within 2 min, with total run
time of 3 min only. The electrospray ionization in positive mode was used and all analytes were
monitored using multiple reaction monitoring (MRM) mode. The method was linear and
reproducible for all the compounds in the range 5 -1000 ng/mL. Between- and within-run
accuracy ranged from 86.7% – 92.5%, and the precision was less than 11.9 % for all seven
compounds. The developed method was successfully applied to in-vitro human microsomal
metabolic stability study. This method could be useful in clinical application for therapeutic drug
monitoring (TDM) of these analytes and for individualization of therapeutic regimens.
Keywords: UPLC-MS/MS, Tyrosine kinase inhibitors, Validation, Plasma, metabolic stability
1. INTRODUCTION
Cancer is a leading cause of death worldwide and is responsible for 8.8 million deaths per
year, mostly in middle- and low-income countries [1]. Tyrosine kinase inhibitors (TKIs) are
cancer growth blockers that inhibit growth factors signaling, thereby preventing cancer cell
proliferation. TKIs have made profound progress in cancer treatment as a type of targeted
therapy. TKIs can block one or multiple types of tyrosine kinases and thus attack specific types
of malignancies with limited toxicity to normal cells. Various preclinical and clinical studies
showed that therapeutic treatment by using TKIs in combinations have a promising curative
potential [2-3]. However, these drugs have narrow therapeutic index and large inter individual
variability in their exposure [4-5]. The large inter-individual variability is mainly attributed to
genetic differences between individuals, tumor sensitivity to treatment, patient compliance and
drug-drug interaction. Therefore, therapeutic drug monitoring (TDM) is necessary to avoid
treatment resistance (sub-therapeutic exposure) or toxicity (overexposure) [6] during TKIs
therapy.
Herein, seven of the most commonly used TKIs for treatment of different types of
malignancies were selected for this study, including osimertinib (OSIM), foretinib (FOT),
dasatinib (DAS), gefitinib (GIF), ibrutinib (IBR), linifanib (LNF), and motesanib (MOT)
(Figure1). OSIM is the first-line treatment for advanced non-small cell lung cancer (NSCLC) [8-
9]. FOT has been used for treatment of breast cancer [7]. DAS has been reported for treatment of
certain hematological malignancies and is available in two formulations (immediate release
tablet and a pediatric powder for oral suspension) [10-11]. GIF is classified as a signal
transduction inhibitor [12] and its combination with metformin is used for treatment of bladder
cancer via intravesical administration [13]. IBR is commonly used for treatment of B-cell
malignancies and for targeted therapy in patients with chronic lymphocytic leukemia [14]. LNF
in combination with other chemotherapies has a remarkable cytotoxic effect on human gastric
cancer [15]. It has been reported that MOT has an antiproliferative activity against colorectal
tumors [16].
Ultra-high performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS)
is a powerful tool for the detection and quantitation of drug and its metabolites in biological
fluids [17-19]. A highly sensitive and reproducible method for the determination of TKIs in
plasma is required to guide therapeutic dose individualization that optimizes therapeutic
effectiveness and minimizes the side effects. In previous studies, we have reported the
quantitative determination of DAS by HPLC-florescence detector [20], MOT, IBR and LNF by
UPLC-MS/MS method [18, 21, 22] in plasma samples. In addition, determination of OSIM [23-
25], FOT [26], DAS [27], IBR [28, 29], GIF [30-32] and LNF [33] has been also reported as
parent drug or with its metabolites in biological fluids. Moreover, determination of DAS, IBR
and GIF with other TKIs in plasma samples has been also reported in literatures [34, 35], but
during the recent literature searched, simultaneous determination of above mentioned seven TKIs
has been not identified. Therefore, in the present study, an UPLC-MS/MS method was developed
and validated for simultaneous determination of new seven tyrosine kinase inhibitors in human
plasma using quizartinib (QUIZ) as internal standard (IS). In vitro metabolic studies are a high￾throughput screening tool to predict various aspects of drugs metabolism and pharmacokinetics
[36].Hence, In vitro metabolic stability study was also carried out to evaluate the metabolic
characteristics of these compounds in human microsomes.
2. Materials and Methods
2.1. Materials
DAS was purchased from TRC Chemicals (Ontario, Canada). LNF, GIF, IBR, and OSIM
were obtained from Beijing Mesochem Technology (Beijing, China), whereas MOT, QUIZ and
FOT were purchased from Enzo Life Sciences (UK) Ltd (Exeter, UK). Ammonium acetate was
obtained from Qualikems Fine Chem Pvt. Ltd (Delhi, India) tert-butyl methyl ether and ethyl
acetate was from Scharalb SL (Barcelona, Spain). Formic acid and acetonitrile (HPLC grade)
were purchased from VWR International Ltd (Poole, England). Ultrapure demineralized water
was generated from the Milli-Q system (Millipore, Molsheim Cedex, France).
2.2. Chromatographic and mass spectroscopy conditions
Analysis was performed by Acquity UPLC-MS/MS system (Waters, Milford, USA). The
triple quadrupole mass spectrometer was operated in MRM mode and the detection was achieved
using electrospray ionization in positive mode. Mass spectrometry parameters including parent to
daughter ion transition, collision energy, cone voltage, and dwell time were optimized for the
determination of each analyte (Table1). Source temperature was adjusted at 150 °C and the
capillary voltage was maintained at 4kV. Collision gas (argon) flow rate was kept at 0.1 mL/min
and desolvatiog gas (nitrogen) flow rate was optimized to 600 L/h. Chromatographic separations
was achieved using an Acquity UPLC BEH C18 1.7 µm 2.1 x 100 mm column (Waters, Milford,
USA) and the mobile phase consists of 0.1% formic acid in acetonitrile and 20 mM ammonium
acetate (95:5) at a flow rate of 0.25 ml/min.
2.3. Standards & quality control samples preparation
The stock standard solutions (10 mg/mL) of all analytes were prepared by dissolving their
reference standards in 1 mL dimethyl sulphoxide (DMSO) and then further diluted to 10 mL
methanol. The working TKIs standard mixture was prepared by dilution of stock solutions in
methanol to reach a final concentration of 10 µg/mL for all the analytes. The IS stock solution (1
mg/mL) was prepared in 1 mL DMSO and then being diluted to 10 mL methanol. A further
dilution was made in acetonitrile to give IS working solution (20 µg/mL). Standard calibration
samples were prepared in human plasma at eight concentrations of 5.0, 10.0, 20.0, 50.0, 100.0,
200.0, 500.0 and 1000.0 ng/mL and quality control (QC) samples were prepared at
concentrations of 5.0, 15.0, 100.0 and 800.0 ng/mL. All standard calibration and QC samples
were stored at -80 °C until analysis.
2.4. Sample preparation
The 10 µL of IS working solution (20 µg/mL) was added to 100 µL of calibrators and QCs
samples and then mixed for 5 sec. 50 µL of acetonitrile was added to the tubes and vortex mixed
for 20 sec. 1 mL of a mixture of ethyl acetate and tert butyl methyl ether (1:1) was added to the
each tubes. The tubes, then were vortex mixed for 30 sec and centrifuged at 10000 x g for 10
minutes. The upper organic layer was then transferred to a clean tube and evaporated to dryness
at 40 °C. The dried extracts were reconstituted with 100 µL mobile phase and 5µLwas injected
to UPLC-MS/MS instrument.
2.5. Method validation
The method validation and run acceptance criteria was carried out in accordance with US
FDA and European Medical Agency (EMA) Guidance for bioanalytical method validation [37,
38].
2.5.1. Selectivity and specificity
Method selectivity was assessed by analyzing blank human plasma samples from 6
different sources to investigate the potential interfering peaks at the retention times of the
analytes and the IS. The blank plasma and plasma spiked with all analytes at lower limit of
quantification (LLOQ) were processed and analyzed. The method confirmed as selective and
specific if the response of blank plasma samples were less than 20 % and 5 % for IS.
2.5.2. Linearity and calibration curves
Calibration standard curves were prepared in human plasma at eight concentration levels
ranging from 5.0 to 1000.0 ng/mL. Five calibration curves were used to evaluate the linearity of
the assay using the least square regression method and a correlation coefficient (r2) of 0.99 or
better
2.5.3. Sensitivity and LLOQ
Sensitivity of the method is defined by the lower limit of quantitation (LLOQ). The
analytes signal to noise ratio in LLOQ should be at least 5:1
LLOQ is the lowest calibration curve concentration where the analyte response should be at least
5 times the blank response. LLOQ was determined for each analyte based on blank plasma
response and accuracy and precision acceptance criteria should be ± 20%.
2.5.4. Recovery and matrix effects
The absolute recovery of the assay was determined by comparing the average of peak
area measurements obtained from blank plasma spiked with quality control (low, middle and
high) samples prior to extraction to those obtained from plasma spiked after extraction. Matrix
effects evaluation was carried out by comparing the peak area ratios of post extracted plasma
spiked with same of quality control samples to the peak area ratios obtained from the spiking
solutions.
2.5.5. Precision and accuracy
Precision and accuracy of the method were determined by the analysis of four quality
control (QC) samples representing the entire range of the calibration curve (LLOQ, low, medium
and high QC samples). Within-batch and between-batch accuracy and precision were measured
using 6 replicates of LLOQ and three QC sample in three successive days. The within-batch and
between-batch mean accuracy should be within ± 15% of the nominal values for QC samples and
20% for LLOQ. The within-batch and between-batch precision should not exceed 15% RSD for
QC samples and 20% for LLOQ.
2.5.6. Stability
Stability of TKIs in human plasma was evaluated by the analysis of two quality control
samples (MQC and HQC) in 6 replicates under various stress conditions including short term
stability at room temperature (~22 °C) for 6 hours, long term stability at approximately -80 °C
for 4 weeks, auto-sampler stability at ambient temperature for 24 hours and freeze-thaw stability.
2.6. In-vitro metabolic stability study
The metabolic stability of the mixture of TKYs was studied using human microsomes (50
Donors) [39]. 10 µL of freshly prepared NADPH (20 mM) was added to a 1.5 mL in Eppendorf
tube containing 2 µL TKIs mixture (800 ng/mL) and 200 mL of pre-warmed 0.1 M phosphate
buffer (37°C). The reaction was initiated by adding 5 µL microsomes (0.5 mg/mL) into all the
tubes. After mixing and incubation at 37 °C in a shaking water bath for 1 h, 50 µL cold
acetonitrile containing IS (100 ng/ mL) was added to terminate the reaction at different time
intervals (0.0, 5.0, 10.0, 20.0, 30.0 and 60.0 min). After mixing and centrifugation at 10,500 g for
5 min, the supernatant was transferred to Eppendorf tube for evaporation. The residues were
reconstituted in 50 µL of acetonitrile and 5 µl was injected to UPLC-MS/MS.
3. Results and discussion
3.1. Mass spectrometry
To optimize mass spectrometer conditions, a solution containing 500 ng/mL for each
analyte was directly infused to mass spectrometer at a flow rate of 10 µl/min. The most efficient
ionization with higher sensitivity was achieved in positive ionization mode. The most
predominate precursor ions [M + H]+
for FOT, OSIM, DAS, GIF, IBR, LNF, MOT and QUIZ
were found at m/z 633.21, m/z 500.72, m/z 488.17, m/z 447.1, m/z 441.16, m/z 374.16, m/z
374.16, and m/z 561.31, respectively. After fragmentation, the most stable product ions were
obtained at m/z 128.08, m/z 72.01, m/z 401.17, m/z 128.08, m/z 84.0, m/z 251.1, m/z 212.1 and
m/z 114.01, respectively (Figure 2). The compound dependent and source dependent parameters
were optimized to achieve the highest response for IS and all the analytes (Table 1).
3.2. Chromatography
Chromatographic conditions were investigated and optimized to achieve short run time,
symmetrical peaks and good resolution of the analytes and IS. Different types of column were
evaluated during method development and the best chromatographic resolution of the analyts
was achieved on Acquity UPLC BEH C18, 1.7 µm 2.1 x 100 mm column (Waters, Milford MD,
USA). The mobile phase compositions of different acetonitrile ratios and various strengths of
ammonium acetate were examined. The best chromatographic separation with symmetric peak
shapes and minimum matrix interference were obtained using 0.1% formic acid in ACN: 20 mM
ammonium acetate (95:5) at a flow rate of 0.25 mL/min. The addition of 0.1% formic acid to
acetonitrile increased the sensitivity and improved peak shape.
3.3. Method validation
Method validation was performed in accordance with FDA and EMA [37, 38] guidance
of bioanalysis. The parameters determined for method validation were sensitivity, selectivity,
low limit of quantitation, linearity, extraction recovery, matrix effect, carryover assessment,
accuracy and precision, and stability.
3.3.1 Selectivity
To evaluate the selectivity, human plasma from six different sources were analyzed. All
test compounds were eluted within 2.5 min and no interfering peaks from endogenous
compounds were found to interfere with the retention time of analytes and IS. This results
confirmed that the developed method is selective and specific for these selected analytes. Typical
chromatograms of the analytes and IS in blank plasma are shown in Figure 3.
3.3.2. Linearity, calibration curves and sensitivity
The calibration curves of seven TKIs were established by 1/x2 weighted linear regression
model. Linearity range was 5.0 – 1000 ng/mL for all tested compounds with correlation
coefficients (r2) ≥ 0.99. The linear regression equations and the mean correlation coefficients of
calibration curves are listed in Table 2. The LLOQ was 5.0 ng/mL for all the analytes in human
plasma. Typical chromatograms of the analytes and IS spiked at LLOQ levels are shown in
Figure 4.
3.3.3. Precision and accuracy
Between-day and within-day precision and accuracy of the method were assessed by
analyzing QC samples at four different concentration levels (LLOQ, low, medium and high) with
6 replicates at each level in three successive days. The precision was expressed as the relative
standard deviation (RSD %). Between-day and within-day precision and accuracy results are
summarized in Table 3. The values were within the acceptable range for all TKIs at all the
concentration levels, suggesting that the method was accurate and precise for determination of
the tested TKIs.
3.3.4. Recovery and matrix effects
The recovery was evaluated by comparing the response of extracted samples at low,
medium and high QC concentrations with the response of blank plasma extracts spiked with the
analytes post extraction at corresponding concentrations. The recovery results are presented in
Table 4. The extraction recoveries were ≥ 69% for all the analytes at the three QC concentration
levels, showing that the investigated TKIs were well recovered during the extraction process.
The average recovery for IS was > 85%. This results confirmed that the recovery of investigated
TKIs are consistent and concentration independent.
Matrix effect was assessed at low, medium and high QC concentrations using 6 different
plasma sources. Peak area measurements obtained from plasma spiked after extraction with the
seven TKIs and IS were compared to those obtained from the spiking solutions (Table 4). The
matrix effect ranged from 83.8 to 94.7%. Therefore, no significant matrix effect was observed in
this method. %. So a slight ions suppression effects has been observed for all TKIs which was
insignificant in this method.
3.3.5. Carryover
Auto-sampler carryover was evaluated by monitoring analytes residues in blank samples
after injection of the upper limit of quantification for 3 cycles. Carryover percentages were less
than 20% of the LLOQ for all analytes, and 5% of the IS.
3.3.6. Stability
The stability of the seven TKIs in human plasma was studied under different conditions
of processing and storage. The results are shown in Table 5. The measured concentrations of all
storage samples were within the acceptance criteria of 85-115%, indicating that all analytes were
stable under the conditions investigated in this study.
3.4. In-vitro metabolic stability study
Calibration curves were constructed in the buffer between the area ratio of each drug of
the mixture and IS versus nominal concentrations of each drug. The metabolic reaction of the
investigated TKIs was terminated at specific time points. The concentrations at different time
intervals were calculated using the corresponding calibration curve. Figure 5 shows the plot of
the relationship between the ln of the % remaining concentration of the studied TKIs versus
incubation time. The residual concentrations were different among the tested drugs as human
microsomal enzymes degraded them at different rates. The results demonstrated that all drugs
were rapidly metabolized during the first 30 min.
4. Conclusion
A reliable, sensitive and high-throughput LC–MS-MS method was developed and
validated for simultaneous quantification of FOT, OSIM, DAS, GIF, IBR, LN and MOT in
human plasma. The method allows high throughput bioanalysis of FOT, OSIM, DAS, GIF, IBR,
LN and MOT in routine clinical analysis including therapeutic drug monitoring and
pharmacokinetics application. The method can provide an appropriate tool for dose
individualization to improve the efficacy and safety of the seven anticancer therapy. The method
was successfully applied in in vitro metabolic studies for the seven TKIs in human microsomes.
The in vitro metabolic studies revealed that the seven TKIs are rapidly metabolizedinhuman
hepatic microsomes.
Acknowledgement
The authors extend their appreciation to the Deanship of Scientific Research at King
Saud University for funding this work through the research group project no. RGP–1435-072.
REFERENCES
1. WHO, https://www.who.int/news-room/fact-sheets/detail/cancer
2. J.T. Hartmann, M, Haap, H.G. Kopp, HP, Lipp, Curr. Drug Metab. Tyrosine kinase
inhibitors – a review on pharmacology, metabolism and side effects, Curr Drug Metab.
10(5) (2009) 470- 481.
3. C.A London, Tyrosine kinase inhibitors in veterinary medicine, Top Companion Anim
Med. 24(3) (2009) 106-112.
4. P. Herviou , E. Thivat , D. Richard, L. Roche, J. Dohou, M. Pouget , A Eschalier, X.
Durando, N. Authier, Therapeutic drug monitoring and tyrosine kinase inhibitors, Oncol
Lett. 12(2) (2016) 1223-1232.
5. M. Herbrink B. Nuijen, J.H. Schellens , J.H. Beijnen, Variability in bioavailability of
small molecular tyrosine kinase inhibitors, Cancer Treat Rev. 41(5) (2015) 412-422.
6. N. Widmer , C. Bardin , E. Chatelut , A. Paci , J. Beijnen , D. Levêque, G. Veal , A.
Astier, Review of therapeutic drug monitoring of anticancer drugs part two–targeted
therapies, Eur. J. Cancer. 50(12) (2014) 2020- 2036.
7. S.K. Chia , S.L. Ellard , M. Mates , S. Welch , C. Mihalcioiu , Miller WH Jr, K. Gelmon C. Lohrisch , V. Kumar , S. Taylor , Hagerman L., Goodwin R., T. Wang, S. Sakashita ,
M.S. Tsao , E. Eisenhauer , P. Bradbury, A phase-I study of lapatinib in combination with
foretinib, a c-MET, AXL and vascular endothelial growth factor receptor inhibitor, in
human epidermal growth factor receptor 2 (HER-2)-positive metastatic breast cancer,
Breast Cancer Res. 19(1) (2017) 54. https://doi: 10.1186/s13058-017-0836-3
8. K. Namba, K. Shien, Y. Takahash , H. Torigoe, H. Sato, T. Yoshioka, T. Takeda, E.
Kurihara , Y. Ogoshi , Y. H. amamoto, J .Soh, S. Tomida , S Toyooka , Activation of
AXL as a preclinical acquired resistance mechanism against osimertinib treatment in
EGFR-mutant non-small cell lung cancer cells, Mol Cancer Res. (2018) https://doi:
10.1158/1541-7786.MCR-18-0628
9. P. Zhou, G. Chen, M. Gao , J. Wu, Design, synthesis and evaluation of the osimertinib
analogue (C-005) as potent EGFR inhibitor against NSCLC, Bioorg Med Chem. 26 (23-
24) (2018) 6135 – 6145.
10. S. Vaidhyanathan , X. Wang , J. Crison, S. Varia, J.Z. Gao, A. Saxena, D. Good,
Bioequivalence comparison of pediatric Dasatinib formulations and elucidation of
absorptionmechanisms through integrated PBPK modeling. J Pharm Sci. 108(1) (2019)
741-749.
11. E.H. McCafferty, S. Dhillon, E.D. Deeks. Dasatinib: A Review in Pediatric Chronic
Myeloid Leukemia, Paediatr Drugs. 20(6) (2018) 593-600.
12. M. Arakawa, K. Nakamura, Y. Yamada, C.J. Rosser, M. Tobiume, H Saito, T. Hasegawa,
N. Honda, Association between gefitinib and hemorrhagic cystitis and severely
contracted bladder, a case report. BMC Urol. (2010) 10:6. doi: 10.1186/1471-2490-10-6.
13. J. Deng, M. Peng, Z. Wang, S. Zhou, D. Xiao, J. Deng, X. Yang , J. Peng, X. Yang,
Novel Application of Metformin Combined with Targeted Drugs on Anticancer
Treatment, Cancer Sci. 110 (1) (2019) 23-30.
14. H.Y. Nam, J.H. Nam, G. Yoon, J.Y. Lee, Y. Nam, H.J. Kang, H.J. Cho, J. Kim, H.S.
Hoe, Ibrutinib suppresses LPS-induced neuroinflammatory responses in BV2 microglial
cells and wild-type mice. J Neuroinflammation. 15(1) 2018 271. https:// doi:
10.1186/s12974-018-1308-0.
15. J. Chen, J. Guo, Z. Chen, J. Wang, M. Liu, X. Pang, Linifanib (ABT- 869) Potentiates
the Efficacy of Chemotherapeutic Agents through the Suppression of Receptor Tyrosine
Kinase-Mediated AKT/m TOR Signaling Pathways in Gastric Cancer, Sci Rep. 6:29382
(2016) https:// doi: 10.1038/srep29382. 29382.
16. T.T. Kaya, A. Altun, N.H. Turgut, H. Ataseven, G. Koyluoglu, Effects of a Multikinase
Inhibitor Motesanib (AMG 706) Alone and Combined with the Selective DuP-697 COX-
2 Inhibitor on Colorectal Cancer Cells, Asian Pac J Cancer Prev. 17(3) (2016) 1103-
1110.
17. E. Ezzeldin , G.A. Mostafa , M. Iqbal , K.A. Al-Rashood , Y. Asiri , T. El-Nahhas .
UPLC/MS-MS assay development for estimation of mozavaptan in plasma and its
pharmacokinetic study in rats. Bioanalysis. 14 (2018):1077-1086.
18. E. Ezzeldin , M. Iqbal , R. Al-Salahi , T. El-Nahhas. Development and validation of a
UPLC-MS/MS method for determination of motesanib in plasma: Application to
metabolic stability and pharmacokinetic studies in rats. J Pharm Biomed Anal. ; 166
(2019)244-251.
19. M. Iqbal , E. Ezzeldin , N.Y. Khalil , P. Alam , K.A. Al-Rashood . UPLC-MS/MS
determination of suvorexant in urine by a simplified dispersive liquid-liquid micro￾extraction followed by ultrasound assisted back extraction from solidified floating
organic droplets. J Pharm Biomed Anal. 5 (2019)164:1-8.
20. Kassem MG1, Ezzeldin E, Korashy HM, Mostafa GA. High-performance liquid
chromatographic method for the determination of dasatinib in rabbit plasma using
fluorescence detection and its application to a pharmacokinetic study. J Chromatogr B
Analyt Technol Biomed Life Sci. 2013 Nov 15; 939:73-9.
21. Iqbal M, Ezzeldin E, Wani TA, Khalil NY. Simple, sensitive and rapid determination of linifanib (ABT-869), a novel tyrosine kinase inhibitor in rat plasma by UHPLC-MS/MS.
Chem Cent J. 2014 Feb 17; 8(1):13.
22. Iqbal M, Shakeel F, Anwer T. Simple and Sensitive UPLC-MS/MS Method for High￾Throughput Analysis of Ibrutinib in Rat Plasma: Optimization by Box-Behnken
Experimental Design. J AOAC Internatioanl. 2016; 99 (3):618-25.
23. Rood JJM, van Bussel MTJ, Schellens JHM, Beijnen JH, Sparidans RW. Liquid
chromatography-tandem mass spectrometric assay for the T790M mutant EGFR inhibitor
osimertinib (AZD9291) in human plasma. J Chromatogr B Analyt Technol Biomed Life
Sci. 2016 Sep 15; 1031:80-85.
24. Zheng X, Wang W, Zhang Y, Ma Y, Zhao H, Hu P, Jiang J. Development and validation
of a UPLC-MS/MS method for quantification of osimertinib (AZD9291) and its
metabolite AZ5104 in human plasma. Biomed Chromatogr. 2018 Dec; 32(12):e4365.
25. Mitchell R, Bailey C, Ewles M, Swan G, Turpin P. Determination of osimertinib in
human plasma, urine and cerebrospinal fluid. Bioanalysis. 2019 May;11(10):987-1001.
26. Attwa MW, Kadi AA, Darwish HW, Amer SM, Alrabiah H. A reliable and stable method
for the determination of foretinib in human plasma by LC-MS/MS: Application to
metabolic stability investigation and excretion rate. Eur J Mass Spectrom (Chichester).
2018 Aug;24(4):344-351.
27. Maher HM, Alzoman NZ, Shehata SM, Abanmy NO. Validated UPLC-MS/MS method
for the quantification of dasatinib in plasma: Application to pharmacokinetic interaction
studies with nutraceuticals in Wistar rats. PLoS One. 2018 Jun 14;13(6):e0199208.
28. Rood JJM, van Hoppe S, Schinkel AH, Schellens JHM, Beijnen JH, Sparidans RW.
Liquid chromatography-tandem mass spectrometric assay for the simultaneous
determination of the irreversible BTK inhibitor ibrutinib and its dihydrodiol-metabolite in
plasma and its application in mouse pharmacokinetic studies. J Pharm Biomed Anal.
2016 Jan 25;118:123-131.
29. Rood JJM, Dormans PJA, van Haren MJ, Schellens JHM, Beijnen JH, Sparidans RW.
Bioanalysis of ibrutinib, and its dihydrodiol- and glutathione cycle metabolites by liquid
chromatography-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed
Life Sci. 2018 Jul 15; 1090:14-21.
30. Zhao M, Hartke C, Jimeno A, Li J, He P, Zabelina Y, Hidalgo M, Baker SD. Specific
method for determination of gefitinib in human plasma, mouse plasma and tissues using
high performance liquid chromatography coupled to tandem mass spectrometry. J
Chromatogr B Analyt Technol Biomed Life Sci. 2005 May 5;819(1):73-80.
31. Zheng N, Zhao C, He XR, Jiang ST, Han SY, Xu GB, Li PP. Simultaneous determination
of gefitinib and its major metabolites in mouse plasma by HPLC-MS/MS and its
application to a pharmacokinetics study. J Chromatogr B Analyt Technol Biomed Life
Sci. 2016 Feb 1; 1011:215-22.
32. Guan S, Chen X, Wang F, Xin S, Feng W, Zhu X, Liu S, Zhuang W, Zhou S, Huang M,Wang X, Zhang L. Development and validation of a sensitive LC-MS/MS method for
determination of gefitinib and its major metabolites in human plasma and its application
in non-small cell lung cancer patients. J Pharm Biomed Anal. 2019 Aug 5; 172:364-371.
33. Rodila RC, Kim JC, Ji QC, El-Shourbagy TA. A high-throughput, fully automated
liquid/liquid extraction liquid chromatography/mass spectrometry method for the
quantitation of a new investigational drug ABT-869 and its metabolite A-849529 in
human plasma samples. Rapid Commun Mass Spectrom. 2006;20(20):3067-75.
34. Merienne C, Rousset M, Ducint D, Castaing N, Titier K, Molimard M, Bouchet S. High
throughput routine determination of 17 tyrosine kinase inhibitors by LC-MS/MS. J Pharm
Biomed Anal. 2018 Feb 20;150:112-120.
35. Huynh HH, Pressiat C, Sauvageon H, Madelaine I, Maslanka P, Lebbé C, Thieblemont C,
Goldwirt L, Mourah S. Development and Validation of a Simultaneous Quantification
Method of 14 Tyrosine Kinase Inhibitors in Human Plasma Using LC-MS/MS.Ther Drug
Monit. 2017 Feb;39(1):43-54.
36. Clarke, S.E. and P. Jeffrey, Utility of metabolic stability screening: comparison of in vitro and in vivo clearance. Xenobiotica, 2001. 31(8-9): p. 591-8.
37. Guidance for Industry on Bioanalytical Method Validation (2001) (Center for Drug
Evaluation and Research, US Food and Drug Administration, Rockville, MD.
https://www.fda.gov/downloads/Drugs/Guidance/ucm070107.pdf (Accessed 01.03.2018)
38. European Medicines Agency, Guideline on Bioanalytical Method Validation, 2012.

https://www.ema.europa.eu/documents/scientific-guideline/guideline-bioanalytical￾method-validation_en.pdf.

39. Hill JR. In vitro drug metabolism using liver microsomes, Curr. Protoc. Pharmacol.
7(2003) 7.7.1-7.8.11.Legend
Figure 1: Chemical structure of Foretinib (A), Osemeritinib (B), Motesanib (C), Gifitinib
(D). Ibrutinib (E), Linifanib (F), Dasatinib (G) and Quizartinib (H).
Figure2: Product ion spectra of the Foretinib seven TKIs drugs and IS in positive ionization mode.
Figure 3. Chromatographic separation. MRM chromatograms of blank plasma of
Foretinib (A), Osemeritinib (B), Motesanib (C), Gifitinib (D). Ibrutinib (E), Linifanib (F),
Dasatinib (G) and Quizartinib (H).
Figure 4. Chromatographic separation. MRM chromatograms of human blank plasma
spiked with low limit of detection of each of Foretinib (A), Osemeritinib (B), Motesanib
(C), Gifitinib (D). Ibrutinib (E), Linifanib (F), Dasatinib (G) and Quizartinib (H).
Figure 5: Percentage drug remaining versus incubation time profile of all TKIs.