Volume 14 Issue 4
Apr.  2024
Turn off MathJax
Article Contents
Lan Chen, Yuan Zhang, Yi-Xin Zhang, Wei-Lai Wang, De-Mei Sun, Peng-Yun Li, Xue-Song Feng, Yue Tan. Pretreatment and analysis techniques development of TKIs in biological samples for pharmacokinetic studies and therapeutic drug monitoring[J]. Journal of Pharmaceutical Analysis, 2024, 14(4): 100899. doi: 10.1016/j.jpha.2023.11.006
Citation: Lan Chen, Yuan Zhang, Yi-Xin Zhang, Wei-Lai Wang, De-Mei Sun, Peng-Yun Li, Xue-Song Feng, Yue Tan. Pretreatment and analysis techniques development of TKIs in biological samples for pharmacokinetic studies and therapeutic drug monitoring[J]. Journal of Pharmaceutical Analysis, 2024, 14(4): 100899. doi: 10.1016/j.jpha.2023.11.006

Pretreatment and analysis techniques development of TKIs in biological samples for pharmacokinetic studies and therapeutic drug monitoring

doi: 10.1016/j.jpha.2023.11.006
Funds:

This work was supported by the Natural Science Foundation of Liaoning Province, China (Grant No.: 2023-MS-172).

  • Received Date: Jun. 10, 2023
  • Accepted Date: Nov. 15, 2023
  • Rev Recd Date: Oct. 26, 2023
  • Publish Date: Nov. 28, 2023
  • Tyrosine kinase inhibitors (TKIs) have emerged as the first-line small molecule drugs in many cancer therapies, exerting their effects by impeding aberrant cell growth and proliferation through the modulation of tyrosine kinase-mediated signaling pathways. However, there exists a substantial inter-individual variability in the concentrations of certain TKIs and their metabolites, which may render patients with compromised immune function susceptible to diverse infections despite receiving theoretically efficacious anticancer treatments, alongside other potential side effects or adverse reactions. Therefore, an urgent need exists for an up-to-date review concerning the biological matrices relevant to bioanalysis and the sampling methods, clinical pharmacokinetics, and therapeutic drug monitoring of different TKIs. This paper provides a comprehensive overview of the advancements in pretreatment methods, such as protein precipitation (PPT), liquid-liquid extraction (LLE), solid-phase extraction (SPE), micro-SPE (μ-SPE), magnetic SPE (MSPE), and vortex-assisted dispersive SPE (VA-DSPE) achieved since 2017. It also highlights the latest analysis techniques such as newly developed high performance liquid chromatography (HPLC) and high-resolution mass spectrometry (HRMS) methods, capillary electrophoresis (CE), gas chromatography (GC), supercritical fluid chromatography (SFC) procedures, surface plasmon resonance (SPR) assays as well as novel nanoprobes-based biosensing techniques. In addition, a comparison is made between the advantages and disadvantages of different approaches while presenting critical challenges and prospects in pharmacokinetic studies and therapeutic drug monitoring.
  • loading
  • [1]
    E. Cardoso, M. Guidi, B. Blanchet, et al., Therapeutic drug monitoring of targeted anticancer protein kinase inhibitors in routine clinical use: A critical review, Ther. Drug Monit. 42 (2020) 33-44.
    [2]
    N. Verougstraete, V. Stove, A.G. Verstraete, et al., Quantification of eight hematological tyrosine kinase inhibitors in both plasma and whole blood by a validated LC-MS/MS method, Talanta 226 (2021), 122140.
    [3]
    H.H. Huynh, C. Pressiat, H. Sauvageon, et al., Development and validation of a simultaneous quantification method of 14 tyrosine kinase inhibitors in human plasma using LC-MS/MS, Ther. Drug Monit. 39 (2017) 43-54.
    [4]
    S. Farag, R.B. Verheijen, J. Martijn Kerst, et al., Imatinib pharmacokinetics in a large observational cohort of gastrointestinal stromal tumour patients, Clin. Pharmacokinet. 56 (2017) 287-292.
    [5]
    R. Xu, Q. Lin, X. Qiu, et al., UPLC-MS/MS method for the simultaneous determination of imatinib, voriconazole and their metabolites concentrations in rat plasma, J. Pharm. Biomed. Anal. 166 (2019) 6-12.
    [6]
    Y. Xia, S. Chen, M. Luo, et al., Correlations between imatinib plasma trough concentration and adverse reactions in Chinese patients with gastrointestinal stromal tumors, Cancer 126 (2020) 2054-2061.
    [7]
    J.C. Yang, N. Reguart, J. Barinoff, et al., Diarrhea associated with afatinib: An oral ErbB family blocker, Expert Rev. Anticancer Ther. 13 (2013) 729-736.
    [8]
    X. Lu, S. Liu, X. Yang, et al., Determination of tyrosine kinase inhibitor afatinib in rat plasma using LC-MS/MS and its application to in vivo pharmacokinetic studies of afatinib liposomes, J. Pharm. Biomed. Anal. 164 (2019) 181-186.
    [9]
    D.N. Suresha, T. Pramila, T. Tamizh Mani, Method development and validation of Imatinib Mesylate-Review, Int. J. Pharm. Pharm. Anal. 1 (2016) 1-11.
    [10]
    N. Li, T. Zhang, G. Chen, et al., Recent advances in sample preparation techniques for quantitative detection of pharmaceuticals in biological samples, Trends Analyt. Chem. 142 (2021), 116318.
    [11]
    H. Li, D. Zhang, X. Cheng, et al., A validated 2D-LC-UV method for simultaneous determination of imatinib and N-desmethylimatinib in plasma and its clinical application for therapeutic drug monitoring with GIST patients, Curr. Pharm. Anal. 18 (2022) 122-131.
    [12]
    M. Zhang, X. Liu, Z. Chen, et al., Method development and validation for simultaneous determination of six tyrosine kinase inhibitors and two active metabolites in human plasma/serum using UPLC-MS/MS for therapeutic drug monitoring, J. Pharm. Biomed. Anal. 211 (2022), 114562.
    [13]
    L. Adlnasab, M. Ezoddin, R.A. Shojaei, et al., Ultrasonic-assisted dispersive micro solid-phase extraction based on melamine-phytate supermolecular aggregate as a novel bio-inspired magnetic sorbent for preconcentration of anticancer drugs in biological samples prior to HPLC-UV analysis, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 1095 (2018) 226-234.
    [14]
    P. Khodayari, N. Jalilian, H. Ebrahimzadeh, et al., Trace-level monitoring of anti-cancer drug residues in wastewater and biological samples by thin-film solid-phase micro-extraction using electrospun polyfam/Co-MOF-74 composite nanofibers prior to liquid chromatography analysis, J. Chromatogr. A 1655 (2021), 462484.
    [15]
    N. Rahimi Kakavandi, T. Asadi, B. Jannat, et al., Method development for determination of imatinib and its major metabolite, N-desmethyl imatinib, in biological and environmental samples by SA-SHS-LPME and HPLC, Biomed. Chromatogr. 35 (2021), e5088.
    [16]
    A. Kazemi, H. Ahmad Panahi, R. Safaeijavan, Thermosensitive molecularly imprinted poly(1-vinyl-2-pyrrolidone/methyl methacrylate/N-vinylcaprolactam) for selective extraction of imatinib mesylate in human biological fluid, J. Sep. Sci. 43 (2020) 614-621.
    [17]
    S. Hooshmand, Z. Es’haghi, Hydrophilic modified magnetic multi-walled carbon nanotube for dispersive solid/liquid phase microextraction of sunitinib in human samples, Anal. Biochem. 542 (2018) 76-83.
    [18]
    A.T.M. da Silva, C.D.P.B. Bessa, W. de S. Borges, et al., Bioanalytical methods for determining ecstasy components in biological matrices: A review, Trends Analyt. Chem. 108 (2018) 323-346.
    [19]
    C.C.L.M. Boons, L. Timmers, J.J.W.M. Janssen, et al., Feasibility of and patients’ perspective on nilotinib dried blood spot self-sampling, Eur. J. Clin. Pharmacol. 75 (2019) 825-829.
    [20]
    J. Lee, S.Y. Jung, M.Y. Choi, et al., Development of a dried blood spot sampling method towards therapeutic monitoring of radotinib in the treatment of chronic myeloid leukaemia, J. Clin. Pharm. Ther. 45 (2020) 1006-1013.
    [21]
    T. Tuzimski, A. Petruczynik, Review of chromatographic methods coupled with modern detection techniques applied in the therapeutic drugs monitoring (TDM), Molecules 25 (2020), 4026.
    [22]
    Y. Mukai, T. Yoshida, T. Kondo, et al., Development and validation of a simple method for simultaneously measuring the concentrations of BCR-ABL and bruton tyrosine kinase inhibitors in dried blood spot (DBS): A pilot study to obtain candidate conversion equations for predicting plasma concentration based on DBS concentration, Ther. Drug Monit. 44 (2022) 762-770.
    [23]
    R. Longuespee, D. Theile, M. Fresnais, et al., Approaching sites of action of drugs in clinical pharmacology: New analytical options and their challenges, Br. J. Clin. Pharmacol. 87 (2021) 858-874.
    [24]
    J. Portnow, B. Badie, S. Markel, et al., A neuropharmacokinetic assessment of bafetinib, a second generation dual BCR-Abl/Lyn tyrosine kinase inhibitor, in patients with recurrent high-grade gliomas, Eur. J. Cancer 49 (2013) 1634-1640.
    [25]
    M. Tamminga, S. de Wit, E. Schuuring, et al., Circulating tumor cells in lung cancer are prognostic and predictive for worse tumor response in both targeted-and chemotherapy, Transl. Lung Cancer Res. 8 (2019) 854-861.
    [26]
    Z. Ye, L. Wu, X. Zhang, et al., Quantification of sorafenib, lenvatinib, and apatinib in human plasma for therapeutic drug monitoring by UPLC-MS/MS, J. Pharm. Biomed. Anal. 202 (2021), 114161.
    [27]
    M. Allard, N. Khoudour, B. Rousseau, et al., Simultaneous analysis of regorafenib and sorafenib and three of their metabolites in human plasma using LC-MS/MS, J. Pharm. Biomed. Anal. 142 (2017) 42-48.
    [28]
    A. Chokshi, A. Gajjar, P. Bhanushali, et al., Quantification of antileukemic drug Dasatinib in human plasma: Application of a sensitive liquid chromatographic method, J. Chem. Metrol. 15 (2021) 152-162.
    [29]
    K. Kleigrewe, A.C. Söhnel, H.U. Humpf, A new high-performance liquid chromatography-tandem mass spectrometry method based on dispersive solid phase extraction for the determination of the mycotoxin fusarin C in corn ears and processed corn samples, J. Agric. Food Chem. 59 (2011) 10470-10476.
    [30]
    M. Sakhi, A. Khan, I. Khan, et al., A new sensitive HPLC/UV method for simultaneous determination of paclitaxel, sorafenib and omeprazole in standard solutions and spiked plasma: Application to in-vitro and in-vivo evaluation of paclitaxel polymeric nanoformulations, Trop. J. Pharm. Res. 20 (2021) 1949-1959.
    [31]
    Z. Wang, L. Lian, Y. Dong, et al., Determination of anlotinib, a tyrosine kinase inhibitor, in rat plasma by UHPLC-MS/MS and its application to a pharmacokinetic study, J. Anal. Methods Chem. 2019 (2019), 5016757.
    [32]
    R.R. Yaragal, D. Kumar, S. Mutnuri, Development of UPLC-MS/MS method for analyzing phorate: Application to wastewater treatment, J. Iran. Chem. Soc. 17 (2020) 2923-2931.
    [33]
    W. Zhuang, H.-B. Qiu, X.-M. Chen, et al., Simultaneous quantification of imatinib and its main metabolite N-demethyl-imatinib in human plasma by liquid chromatography-tandem mass spectrometry and its application to therapeutic drug monitoring in patients with gastrointestinal stromal tumor, Biomed. Chromatogr. 31 (2017), e4022.
    [34]
    J. Zeng, H.L. Cai, Z.P. Jiang, et al., A validated UPLC-MS/MS method for simultaneous determination of imatinib, dasatinib and nilotinib in human plasma, J. Pharm. Anal. 7 (2017) 374-380.
    [35]
    M. Gurjar, P. Mehta, J. Sharma, et al., An HPLC method for simultaneous quantification of sunitinib and its active metabolite, SU12662, using hydrophilic interaction chromatography principle, Bioanalysis 12 (2020) 75-85.
    [36]
    T. Zhao, L. Wang, D.D.Y. Chen, Quantification of imatinib and related compounds using capillary electrophoresis-tandem mass spectrometry with field-amplified sample stacking, Electrophoresis 41 (2020) 1843-1850.
    [37]
    E. Ezzeldin, M. Iqbal, R.N. Herqash, et al., Simultaneous quantitative determination of seven novel tyrosine kinase inhibitors in plasma by a validated UPLC-MS/MS method and its application to human microsomal metabolic stability study, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 1136 (2020), 121851.
    [38]
    J. Jolibois, A. Schmitt, B. Royer, A simple and fast LC-MS/MS method for the routine measurement of cabozantinib, olaparib, palbociclib, pazopanib, sorafenib, sunitinib and its main active metabolite in human plasma, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 1132 (2019), 121844.
    [39]
    M. Ni, J. Zhou, H. Li, et al., Simultaneous determination of six tyrosine kinase inhibitors in human plasma using HPLC-Q-Orbitrap mass spectrometry, Bioanalysis 9 (2017) 925-935.
    [40]
    Z. Xu, C. Zhang, X. Yu, et al., Microwave-assisted solid-phase synthesis of nitrogen-doping carbon dot with good solvent compatibility and its sensing of sunitinib, Anal. Bioanal. Chem. 413 (2021) 6435-6447.
    [41]
    L. Zhou, S. Wang, M. Chen, et al., Simultaneous and rapid determination of 12 tyrosine kinase inhibitors by LC-MS/MS in human plasma: Application to therapeutic drug monitoring in patients with non-small cell lung cancer, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 1175 (2021), 122752.
    [42]
    O.S. Ahmed, Y. Ladner, J. Xia, et al., A fully automated on-line salting-out assisted liquid-liquid extraction capillary electrophoresis methodology: Application to tyrosine kinase inhibitors in human plasma, Talanta 208 (2020), 120391.
    [43]
    O.S. Ahmed, Y. Ladner, J. Montels, et al., Coupling of salting-out assisted liquid-liquid extraction with on-line stacking for the analysis of tyrosine kinase inhibitors in human plasma by capillary zone electrophoresis, J. Chromatogr. A 1579 (2018) 121-128.
    [44]
    A.Z. Moghaddam, A.E. Bameri, M.R. Ganjali, et al., A low-voltage electro-membrane extraction for quantification of imatinib and sunitinib in biological fluids, Bioanalysis 13 (2021) 1401-1413.
    [45]
    H. Liu, M. Zhang, X. Wang, et al., Extraction and determination of polybrominated diphenyl ethers in water and urine samples using solidified floating organic drop microextraction along with high performance liquid chromatography, Microchim. Acta 176 (2012) 303-309.
    [46]
    M. Cruz-Vera, R. Lucena, S. Cardenas, et al., One-step in-syringe ionic liquid-based dispersive liquid-liquid microextraction, J. Chromatogr. A 1216 (2009) 6459-6465.
    [47]
    M. Ghazaghi, H.Z. Mousavi, H. Shirkhanloo, et al., Stirring-controlled solidified floating solid-liquid drop microextraction as a new solid phase-enhanced liquid-phase microextraction method by exploiting magnetic carbon nanotube-nickel hybrid, Anal. Chim. Acta 951 (2017) 78-88.
    [48]
    C. Merienne, M. Rousset, D. Ducint, et al., High throughput routine determination of 17 tyrosine kinase inhibitors by LC-MS/MS, J. Pharm. Biomed. Anal. 150 (2018) 112-120.
    [49]
    N.Z. Alzoman, H.M. Maher, S.M. Shehata, et al., UPLC-MS/MS study of the effect of dandelion root extract on the plasma levels of the selected irreversible tyrosine kinase inhibitors dasatinib, imatinib and nilotinib in rats: Potential risk of pharmacokinetic interactions, Biomed. Chromatogr. 33 (2019), e4674.
    [50]
    A. Wojnicz, B. Colom-Fernandez, J.L. Steegmann, et al., Simultaneous determination of imatinib, dasatinib, and nilotinib by liquid chromatography-tandem mass spectrometry and its application to therapeutic drug monitoring, Ther. Drug Monit. 39 (2017) 252-262.
    [51]
    I. Vrobel, H. Janeckova, E. Faber, et al., Ultrafast online SPE-MS/MS method for quantification of 3 tyrosine kinase inhibitors in human plasma, Ther. Drug Monit. 38 (2016) 516-524.
    [52]
    H.M. Maher, N.Z. Alzoman, S.M. Shehata, et al., Comparative pharmacokinetic profiles of selected irreversible tyrosine kinase inhibitors, neratinib and pelitinib, with apigenin in rat plasma by UPLC-MS/MS, J. Pharm. Biomed. Anal. 137 (2017) 258-267.
    [53]
    J. Li, L. Zhao, C. Wei, et al., Preparation of restricted access media molecularly imprinted polymers for efficient separation and enrichment ofloxacin in bovine serum samples, J. Sep. Sci. 42 (2019) 2491-2499.
    [54]
    T. Khezeli, A. Daneshfar, Development of dispersive micro-solid phase extraction based on micro and nano sorbents, Trends Analyt. Chem. 89 (2017) 99-118.
    [55]
    D. Koller, V. Vaitsekhovich, C. Mba, et al., Effective quantification of 11 tyrosine kinase inhibitors and caffeine in human plasma by validated LC-MS/MS method with potent phospholipids clean-up procedure. Application to therapeutic drug monitoring, Talanta 208 (2020), 120450.
    [56]
    N. Li, Q. Zhu, Y. Yang, et al., A novel dispersive solid-phase extraction method using metal-organic framework MIL-101 as the adsorbent for the analysis of benzophenones in toner, Talanta 132 (2015) 713-718.
    [57]
    C. Qi, Q. Cai, P. Zhao, et al., The metal-organic framework MIL-101(Cr) as efficient adsorbent in a vortex-assisted dispersive solid-phase extraction of imatinib mesylate in rat plasma coupled with ultra-performance liquid chromatography/mass spectrometry: Application to a pharmacokinetic study, J. Chromatogr. A 1449 (2016) 30-38.
    [58]
    L. Chen, M. Zhang, F. Fu, et al., Facile synthesis of magnetic covalent organic framework nanobeads and application to magnetic solid-phase extraction of trace estrogens from human urine, J. Chromatogr. A 1567 (2018) 136-146.
    [59]
    L. Chen, Y. He, Z. Lei, et al., Preparation of core-shell structured magnetic covalent organic framework nanocomposites for magnetic solid-phase extraction of bisphenols from human serum sample, Talanta 181 (2018) 296-304.
    [60]
    H. Jiang, N. Li, L. Cui, et al., Recent application of magnetic solid phase extraction for food safety analysis, Trends Analyt. Chem. 120 (2019), 115632.
    [61]
    G. Li, M. Zhao, L. Zhao, Development and validation of an UPLC-MS/MS method for simultaneous determination of fifteen targeted anti-cancer drugs in human plasma and its application in therapeutic drug monitoring, J. Pharm. Biomed. Anal. 212 (2022), 114517.
    [62]
    M. Arvand, A.N. Masouleh, Magnetic solid-phase extraction of imatinib and doxorubicin as cytostatic drugs by Fe3O4/graphene oxide nanocomposite, J. Iran. Chem. Soc. 14 (2017) 1673-1682.
    [63]
    H. Sahebi, S.M. Pourmortazavi, H. Zandavar, et al., Chitosan grafted onto Fe3O4@poly(N-vinylcaprolactam) as a new sorbent for detecting Imatinib mesylate in biosamples using UPLC-MS/MS, Analyst 144 (2019) 7336-7350.
    [64]
    H. Sahebi, E. Konoz, A. Ezabadi, et al., Sensitive determination of imatinib mesylate in human plasma using DABCO-based ionic liquid-modified magnetic nanoparticles, Chromatographia 83 (2020) 1009-1019.
    [65]
    M. Pirdadeh-Beiranvand, A. Afkhami, T. Madrakian, Magnetic molecularly imprinted electrospun nanofibers for selective extraction of nilotinib from human serum, Anal. Bioanal. Chem. 412 (2020) 1629-1637.
    [66]
    D. Liu, J. Peng, L. Chen, et al., Solid phase extraction-based magnetic carbon nitride/metal organic framework composite with high performance liquid chromatography for the determination of tyrosine kinase inhibitors in urine samples, Anal. Methods 12 (2020) 4798-4805.
    [67]
    E.A. Souza Silva, S. Risticevic, J. Pawliszyn, Recent trends in SPME concerning sorbent materials, configurations and in vivo applications, Trends Analyt. Chem. 43 (2013) 24-36.
    [68]
    E.A. Souza-Silva, N. Reyes-Garces, G.A. Gomez-Rios, et al., A critical review of the state of the art of solid-phase microextraction of complex matrices III. Bioanalytical and clinical applications, Trends Analyt. Chem. 71 (2015) 249-264.
    [69]
    B. Bojko, E. Cudjoe, G.A. Gomez-Rios, et al., SPME: Quo vadis? Anal. Chim. Acta 750 (2012) 132-151.
    [70]
    G.A. Gomez-Rios, N. Reyes-Garces, B. Bojko, et al., Biocompatible solid-phase microextraction nanoelectrospray ionization: An unexploited tool in bioanalysis, Anal. Chem. 88 (2016) 1259-1265.
    [71]
    A. Miodek, N. Mejri-Omrani, R. Khoder, et al., Electrochemical functionalization of polypyrrole through amine oxidation of poly(amidoamine) dendrimers: Application to DNA biosensor, Talanta 154 (2016) 446-454.
    [72]
    M. Zhou, Y. Wang, Y. Zhai, et al., Controlled synthesis of large-area and patterned electrochemically reduced graphene oxide films, Chemistry 15 (2009) 6116-6120.
    [73]
    B. Hatamluyi, Z. Es’haghi, A layer-by-layer sensing architecture based on dendrimer and ionic liquid supported reduced graphene oxide for simultaneous hollow-fiber solid phase microextraction and electrochemical determination of anti-cancer drug imatinib in biological samples, J. Electroanal. Chem. 801 (2017) 439-449.
    [74]
    A. Jerath, Q.J. Yang, K.S. Pang, et al., Tranexamic acid dosing for cardiac surgical patients with chronic renal dysfunction: A new dosing regimen, Anesth. Analg. 127 (2018) 1323-1332.
    [75]
    Y. Cui, S. Liu, K. Wei, et al., Magnetic solid-phase extraction of trace-level mercury(II) ions using magnetic core-shell nanoparticles modified with thiourea-derived chelating agents, Microchim. Acta 182 (2015) 1337-1344.
    [76]
    M. Ghazaghi, H.Z. Mousavi, H. Shirkhanloo, et al., Ultrasound assisted dispersive micro solid-phase extraction of four tyrosine kinase inhibitors from serum and cerebrospinal fluid by using magnetic nanoparticles coated with nickel-doped silica as an adsorbent, Microchim. Acta 183 (2016) 2779-2789.
    [77]
    W. Xiong, J.H. Kang, Y. Jung, Preparation of nitrogen-doped porous carbon from melamine-formaldehyde resins crosslinked by phytic acid, Int. J. Electrochem. Sci. 13 (2018) 852-862.
    [78]
    M. Ghorbani, M. Aghamohammadhassan, M. Chamsaz, et al., Dispersive solid phase microextraction, Trends Analyt. Chem. 118 (2019) 793-809.
    [79]
    S. Jo, H. Jeong, S.R. Bae, et al., Modified platinum electrode with phytic acid and single-walled carbon nanotube: Application to the selective determination of dopamine in the presence of ascorbic and uric acids, Microchem. J. 88 (2008) 1-6.
    [80]
    Y. Liu, H. Liu, Z. Xia, et al., Simultaneous and rapid determination of six tyrosine kinase inhibitors in patients with non-small cell lung cancer using HPLC-MS/MS, Int. J. Anal. Chem. 2021 (2021), 5524361.
    [81]
    Q. Zhang, Z. Li, K. Xu, et al., Intracellular concentration and transporters in imatinib resistance of gastrointestinal stromal tumor, Scand. J. Gastroenterol. 54 (2019) 220-226.
    [82]
    T. Hirasawa, M. Kikuchi, K. Shigeta, et al., High-throughput liquid chromatography/electrospray ionization-tandem mass spectrometry method using in-source collision-induced dissociation for simultaneous quantification of imatinib, dasatinib, bosutinib, nilotinib, and ibrutinib in human plasma, Biomed. Chromatogr. 35 (2021), e5124.
    [83]
    H.M. Maher, N.Z. Alzoman, S.M. Shehata, Ultra-performance LC-MS/MS study of the pharmacokinetic interaction of imatinib with selected vitamin preparations in rats, Bioanalysis 10 (2018) 1099-1113.
    [84]
    J. Wen, S. Bao, Y. Cai, et al., A reliable and stable method for determination of brigatinib in rat plasma by UPLC-MS/MS: Application to a pharmacokinetic study, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 1068-1069 (2017) 84-89.
    [85]
    H. Son, W. Kang, Quantitative determination of bilobetin in rat plasma by HPLC-MS/MS and its application to a pharmacokinetic study, Biomed. Chromatogr. 34 (2020), e4784.
    [86]
    B. Pasquini, S. Orlandini, S. Furlanetto, et al., Quality by Design as a risk-based strategy in pharmaceutical analysis: Development of a liquid chromatography-tandem mass spectrometry method for the determination of nintedanib and its impurities, J. Chromatogr. A 1611 (2020), 460615.
    [87]
    P.J. Rudzki, E. Gniazdowska, K. Bus-Kwasnik, Quantitative evaluation of the matrix effect in bioanalytical methods based on LC-MS: A comparison of two approaches, J. Pharm. Biomed. Anal. 155 (2018) 314-319.
    [88]
    G.D.M. Veerman, P. de Bruijn, A.C. Dingemans, et al., To quantify the small-molecule kinase inhibitors ceritinib, dacomitinib, lorlatinib, and nintedanib in human plasma by liquid chromatography/triple-quadrupole mass spectrometry, J. Pharm. Biomed. Anal. 193 (2021), 113733.
    [89]
    V. Iacuzzi, M. Zanchetta, S. Gagno, et al., A LC-MS/MS method for therapeutic drug monitoring of sorafenib, regorafenib and their active metabolites in patients with hepatocellular carcinoma, J. Pharm. Biomed. Anal. 187 (2020), 113358.
    [90]
    H.M. Maher, N.Z. Alzoman, S.M. Shehata, et al., Validated UPLC-MS/MS method for the quantification of dasatinib in plasma: Application to pharmacokinetic interaction studies with nutraceuticals in Wistar rats, PLoS One 13 (2018), e0199208.
    [91]
    D. Dutta, S. Das, J.A. Seijas, et al., Validated stability-indicating HPTLC method for nintedanib & characterization of degradants by LC-MSn, The 23rd International Electronic Conference on Synthetic Organic Chemistry, November 15-December 15, 2019, https://doi.org/10.3390/ecsoc-23-06502.
    [92]
    Y. He, L. Zhou, S. Gao, et al., Development and validation of a sensitive LC-MS/MS method for simultaneous determination of eight tyrosine kinase inhibitors and its application in mice pharmacokinetic studies, J. Pharm. Biomed. Anal. 148 (2018) 65-72.
    [93]
    Y. Mukai, T. Yoshida, T. Kondo, et al., Novel high-performance liquid chromatography-tandem mass spectrometry method for simultaneous quantification of BCR-ABL and Bruton’s tyrosine kinase inhibitors and their three active metabolites in human plasma, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 1137 (2020), 121928.
    [94]
    A.S. Abdelhameed, M.W. Attwa, N.S. Al-Shaklia, et al., A highly sensitive LC-MS/MS method to determine novel Bruton’s tyrosine kinase inhibitor spebrutinib: Application to metabolic stability evaluation, R. Soc. Open Sci. 6 (2019), 190434.
    [95]
    P. Johnsirani, A.A. Wani, P.V. Bharatam, et al., LC-ESI-QTOF-MS analysis utilizing gas-phase fragmentation reactions subjected to ESI-IS-CID and ESI-CID-MS/MS conditions to study the degradation behaviour of sorafenib tosylate: NMR and in vitro cytotoxicity and apoptosis detection studies of hydrolytic degradation products, J. Pharm. Biomed. Anal. 177 (2020), 112881.
    [96]
    J. Roosendaal, S.L. Groenland, H. Rosing, et al., Determination of the absolute bioavailability of oral imatinib using a stable isotopically labeled intravenous imatinib-d8 microdose, Eur. J. Clin. Pharmacol. 76 (2020) 1075-1082.
    [97]
    E. Pirro, S. De Francia, F. De Martino, et al., A new HPLC-UV validated method for therapeutic drug monitoring of tyrosine kinase inhibitors in leukemic patients, J. Chromatogr. Sci. 49 (2011) 753-757.
    [98]
    G.P. Kocan, M. Huang, F. Li, et al., A sensitive LC-MS-MS assay for the determination of lapatinib in human plasma in subjects with end-stage renal disease receiving hemodialysis, J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 1097-1098 (2018) 74-82.
    [99]
    F. Hasin, M.T. Islam, M.F. Ahmad, et al., Validation of assay method for the estimation of imatinib mesylate in tablet dosage form by HPLC, Eur. J. Biomed. Pharm. Sci. 4 (2017) 74-81.
    [100]
    O. Coban, Z. Degim, Development and validation of highly selective method for the determination of imatinib mesylate and dexketoprofen trometamol combination in three different media, Braz. J. Pharm. Sci. 56 (2020), e18583.
    [101]
    P. Shah, N. Shah, R. Shah, Method development and validation of a stability indicating RP-HPLC method for assay determination of imatinib in imatinib mesylate tablets dosage form, Int. J. Curr. Pharm. Rev. Res. 6 (2015) 4453-4468.
    [102]
    H.A. Alhazmi, D.A. Moraya, E. Alahdal, et al., Ultrafast monolithic HPLC method for simultaneous quantification of the anticancer agents, imatinib and sorafenib: Application to tablet dosage forms, Trop. J. Pharm Res 17 (2018) 1127-1134.
    [103]
    A.C. Alaejos, S.J. Cabrera, B.C. Rodriguez, et al., Validation and comparison of two analytical methods for imatinib therapeutic drug monitoring, Chromatographia 84 (2021) 589-596.
    [104]
    A.K. Kuna, G. Seru, G.V. Radha, Analytical method development and validation for the estimation of Imatinib mesylate and its dimer impurity in pharmaceutical formulation by reverse-phase high-performance liquid chromatography, Asian J. Pharm. Clin. Res. 11 (2018) 136-139.
    [105]
    Y. Zhou, J. Lai, F. Qiu, et al., Development and validation of an HPLC-UV method for routine trough plasma concentration monitoring of imatinib in Chinese patients with gastrointestinal stromal tumor, J. Chin. Pharm. Sci. 29 (2020) 637-648.
    [106]
    N.S. Lakka, C. Kuppan, K.S. Srinivas, et al., Separation and characterization of new forced degradation products of dasatinib in tablet dosage formulation using LC-MS and stability-indicating HPLC methods, Chromatographia 83 (2020) 947-962.
    [107]
    B. Mohan, D. Ravi Kumar, R.S.K. Sharma, et al., Validated RP-HPLC method for estimation of related impurities in dasatinib, Eurasian J. Analyt. Chem. 15 (2020) 51-58.
    [108]
    D.M. Patel, D. Patel, A. Patel, et al., Method development and validation for simultaneous estimation of benidipine hydrochloride and metoprolol succinate in tablet, J. Drug Delivery Ther. 9 (2019) 28-33.
    [109]
    P.R. Sankar, S. Anusha, Development and validation of RP-HPLC method for the determination of dasatinib in the tablet dosage form, Int. J. Pharm. Sci. Res. 10 (2019) 4531-4537.
    [110]
    C. Sojitra, A. Tehare, C. Dholakia, et al., Development of chromatographic method for determination of impurities in solid dispersion of dasatinib, Braz. J. Anal. Chem. 5 (2018) 19-29.
    [111]
    P. Ravi Sankar, A. Viswanath, M.M. Eswarudu, et al., Development and validation of RP-HPLC method for the determination of sorafenib in pharmaceutical dosage form, Int. J. Pharm. Sci. Rev. Res. 69 (2021) 15-23.
    [112]
    S.K. Shukla, H. Kadry, J.A. Bhatt, et al., Statistical optimization and validation of a novel ultra-performance liquid chromatography method for estimation of nintedanib in rat and human plasma, Bioanalysis 12 (2020) 159-174.
    [113]
    R. Kumar, V.K. Munipalli, R.M. Singh, et al., Validated RP-HPLC method for determination and quantification of nintedanib in pharmaceutical formulation, J. Adv. Pharmacol. 1 (2020) 38-47.
    [114]
    R. Lalitha, K. Gandla, P.V.T. Shruthi, et al., Method development and validation of sunitinib in bulk and pharmaceutical dosage form by RP-HPLC method, Int. J. Pharmacogn. Chem. 1 (2020) 70-73.
    [115]
    B. Jayagopal, S. Murugesh, QbD-mediated RP-UPLC method development invoking an FMEA-based risk assessment to estimate nintedanib degradation products and their pathways, Arab. J. Chem. 13 (2020) 7087-7103.
    [116]
    T. Sharma, R.K. Khurana, A. Jain, et al., Development of a validated liquid chromatographic method for quantification of sorafenib tosylate in the presence of stress-induced degradation products and in biological matrix employing analytical quality by design approach, Biomed. Chromatogr. 32 (2018), e4169.
    [117]
    S.A. Waghmare, M. Sumithra, QbD based development and validation of RP-HPLC method for nintedanib esylate: Application to bioanalytical and stability study in plasma, Anal. Chem. Lett. 11 (2021) 392-408.
    [118]
    Z. Wu, J. Liu, M. Liang, et al., Detection of imatinib based on electrochemical sensor constructed using biosynthesized graphene-silver nanocomposite, Front. Chem. 9 (2021), 670074.
    [119]
    F. Tahernejad-Javazmi, M. Shabani-Nooshabadi, H. Karimi-Maleh, Gold nanoparticles and reduced graphene oxide-amplified label-free DNA biosensor for dasatinib detection, New J. Chem. 42 (2018) 16378-16383.
    [120]
    B. Vercelli, S. Crotti, M. Agostini, Voltammetric responses at modified electrodes and aggregation effects of two anticancer molecules: Irinotecan and sunitinib, New J. Chem. 44 (2020) 18233-18241.
    [121]
    H. Chen, K. Luo, K. Li, A facile electrochemical sensor based on NiO-ZnO/MWCNT-COOH modified GCE for simultaneous quantification of imatinib and itraconazole, J. Electrochem. Soc. 166 (2019) B697-B707.
    [122]
    B. Hassan Pour, N. Haghnazari, F. Keshavarzi, et al., High sensitive electrochemical sensor for imatinib based on metal-organic frameworks and multiwall carbon nanotubes nanocomposite, Microchem. J. 165 (2021), 106147.
    [123]
    N. Rezvani Jalal, T. Madrakian, A. Afkhami, et al., In situ growth of metal-organic framework HKUST-1 on graphene oxide nanoribbons with high electrochemical sensing performance in imatinib determination, ACS Appl. Mater. Interfaces 12 (2020) 4859-4869.
    [124]
    S. Mirsadeghi, H. Zandavar, M. Rahimi, et al., Photocatalytic reduction of imatinib mesylate and imipenem on electrochemical synthesized Al2W3O12 nanoparticle: Optimization, investigation of electrocatalytic and antimicrobial activity, Colloids Surf. A Physicochem. Eng. Asp. 586 (2020), 124254.
    [125]
    S.A.R. Alavi-Tabari, M.A. Khalilzadeh, H. Karimi-Maleh, Simultaneous determination of doxorubicin and dasatinib as two breast anticancer drugs uses an amplified sensor with ionic liquid and ZnO nanoparticle, J. Electroanal. Chem. 811 (2018) 84-88.
    [126]
    M. Ghapanvari, T. Madrakian, A. Afkhami, Sensitive determination of imatinib in human biological samples by differential pulse voltammetry based on carbon paste electrode modified by MMWCNTs/PAN NFs, (2018).
    [127]
    A. Yarahmadi, T. Madrakian, A. Afkhami, et al., Electrochemical determination of sunitinib in biological samples using polyacrylonitrile nanofibers/nickel-zinc-ferrite nanocomposite/carbon paste electrode, J. Electrochem. Soc. 166 (2019) B1268-B1275.
    [128]
    M. Ghapanvari, T. Madrakian, A. Afkhami, et al., A modified carbon paste electrode based on Fe3O4@multi-walled carbon nanotubes@polyacrylonitrile nanofibers for determination of imatinib anticancer drug, J. Appl. Electrochem. 50 (2020) 281-294.
    [129]
    J. Rodriguez, G. Castaneda, I. Lizcano, Electrochemical sensor for leukemia drug imatinib determination in urine by adsorptive striping square wave voltammetry using modified screen-printed electrodes, Electrochim. Acta 269 (2018) 668-675.
    [130]
    P.K. Kalambate, Y. Li, Y. Shen, et al., Mesoporous Pd@Pt core-shell nanoparticles supported on multi-walled carbon nanotubes as a sensing platform: Application in simultaneous electrochemical detection of anticancer drugs doxorubicin and dasatinib, Anal. Methods 11 (2019) 443-453.
    [131]
    A. Moghaddam, H.A. Zamani, H. Karimi-Maleh, A new electrochemical platform for dasatinib anticancer drug sensing using Fe3O4-SWCNTs/ionic liquid paste sensor, Micromachines 12 (2021), 437.
    [132]
    R.V. Rele, P. Tiwatane, A non-aqueous potentiometric titration method for validation of imatinib mesylate from pharmaceutical dosages, Asia. J. Rese. Chem. 12 (2019) 307-310.
    [133]
    W. Yang, M. Zhou, N. Oturan, et al., Electrocatalytic destruction of pharmaceutical imatinib by electro-Fenton process with graphene-based cathode, Electrochim. Acta 305 (2019) 285-294.
    [134]
    S. Tartaggia, A. Meneghello, O. Bellotto, et al., An SPR investigation into the therapeutic drug monitoring of the anticancer drug imatinib with selective aptamers operating in human plasma, Analyst 146 (2021) 1714-1724.
    [135]
    S. Fornasaro, A. Bonifacio, E. Marangon, et al., Label-free quantification of anticancer drug imatinib in human plasma with surface enhanced Raman spectroscopy, Anal. Chem. 90 (2018) 12670-12677.
    [136]
    P. Ravisankar, S. Anusha, P. Srinivasa Babu, Development and validation of UV-spectrophotometric method for determination of dasatinib in bulk and pharmaceutical dosage form and its degradation behaviour under various stress conditions, Int. J. Pharm. Sci. Rev. Res. 53 (2020) 45-50.
    [137]
    P. Ravisankar, P.S. Babu, S.M. Taslim, et al., Development and validation of UV-spectrophotometric method for determination of sorafenib in pharmaceutical dosage form and its degradation behaviour under various stress conditions, Int. J. Pharm. Sci. Rev. Res. 56 (2019) 12-17.
    [138]
    E. Souri, E. Amoon, N. Shabani Ravari, et al., Spectrophotometric methods for determination of sunitinib in pharmaceutical dosage forms based on ion-pair complex formation, Iran. J. Pharm. Res. 19 (2020) 103-109.
    [139]
    I.A. Darwish, N.Y. Khalil, H.W. Darwish, et al., Spectrophotometric and computational investigations of charge transfer complexes of chloranilic acid with tyrosine kinase inhibitors and application to development of novel universal 96-microwell assay for their determination in pharmaceutical formulations, Spectrochim. Acta A Mol. Biomol. Spectrosc. 252 (2021), 119482.
    [140]
    I.A. Darwish, H.W. Darwish, N.Y. Khalil, et al., Experimental and computational evaluation of chloranilic acid as an universal chromogenic reagent for the development of a novel 96-microwell spectrophotometric assay for tyrosine kinase inhibitors, Molecules 26 (2021), 744.
    [141]
    A.S. Abdelhameed, M.W. Attwa, M.I. Attia, et al., Development of novel univariate and multivariate validated chemometric methods for the analysis of dasatinib, sorafenib, and vandetanib in pure form, dosage forms and biological fluids, Spectrochim. Acta A Mol. Biomol. Spectrosc. 264 (2022), 120336.
    [142]
    A.S. Abdelhameed, E.S. Hassan, M.W. Attwa, et al., Simple and efficient spectroscopic-based univariate sequential methods for simultaneous quantitative analysis of vandetanib, dasatinib, and sorafenib in pharmaceutical preparations and biological fluids, Spectrochim. Acta A Mol. Biomol. Spectrosc. 260 (2021), 119987.
    [143]
    F. Belal, F. Ibrahim, Z.A. Sheribah, et al., New spectrophotometric/chemometric assisted methods for the simultaneous determination of imatinib, gemifloxacin, nalbuphine and naproxen in pharmaceutical formulations and human urine, Spectrochim. Acta A Mol. Biomol. Spectrosc. 198 (2018) 51-60.
    [144]
    Z. Zhang, J. Chen, Q. Yang, et al., Eco-friendly intracellular microalgae synthesis of fluorescent CdSe QDs as a sensitive nanoprobe for determination of imatinib, Sens. Actuat. B Chem. 263 (2018) 625-633.
    [145]
    D.W. Zidan, W.S. Hassan, M.S. Elmasry, et al., A novel spectrofluorimetric method for determination of imatinib in pure, pharmaceutical preparation, human plasma, and human urine, Luminescence 33 (2018) 232-242.
    [146]
    H.W. Darwish, A.H. Bakheit, N.S. Al-Shakliah, et al., Development of innovative artificial neural networks for simultaneous determination of lapatinib and foretinib in human urine by micellar enhanced synchronous spectrofluorimetry, Spectrochim. Acta A Mol. Biomol. Spectrosc. 238 (2020), 118438.
    [147]
    H.W. Darwish, A.H. Bakheit, N.S. Al-Shakliah, et al., Experimental and computational evaluation of kolliphor RH 40 as a new fluorescence enhancer in development of a micellar-based spectrofluorimetric method for determination of lapatinib in tablets and urine, PLoS One 15 (2020), e0239918.
    [148]
    H. Salem, F.A. Abo Elsoud, D. Heshmat, et al., Resonance Rayleigh scattering technique-using erythrosine B, as novel spectrofluorimetric method for determination of anticancer agent nilotinib: Application for capsules and human plasma, Spectrochim. Acta A Mol. Biomol. Spectrosc. 251 (2021), 119428.
    [149]
    M. Pirdadeh-Beiranvand, A. Afkhami, T. Madrakian, Ni0.5Zn0.5Fe2O4 nanoparticles-decorated poly (vinyl alcohol) nanofiber as resonance light scattering probe for determination of sunitinib in serum samples, Talanta 218 (2020), 121190.
    [150]
    H.M. Kashani, T. Madrakian, A. Afkhami, Highly fluorescent nitrogen-doped graphene quantum dots as a green, economical and facile sensor for the determination of sunitinib in real samples, New J. Chem. 41 (2017) 6875-6882.
    [151]
    M. Forough, K. Farhadi, A. Eyshi, et al., Rapid ionic liquid-supported nano-hybrid composite reinforced hollow-fiber electromembrane extraction followed by field-amplified sample injection-capillary electrophoresis: An effective approach for extraction and quantification of Imatinib mesylate in human plasma, J. Chromatogr. A 1516 (2017) 21-34.
    [152]
    A.G. Gonzalez, L. Taraba, J. Hranicek, et al., Determination of dasatinib in the tablet dosage form by ultra high performance liquid chromatography, capillary zone electrophoresis, and sequential injection analysis, J. Sep. Sci. 40 (2017) 400-406.
    [153]
    G. Sodeifian, F. Razmimanesh, S. Ali Sajadian, Solubility measurement of a chemotherapeutic agent (Imatinib mesylate) in supercritical carbon dioxide: Assessment of new empirical model, J. Supercrit. Fluids 146 (2019) 89-99.
    [154]
    G. Sodeifian, F. Razmimanesh, S. Ali Sajadian, Prediction of solubility of sunitinib malate (an anti-cancer drug) in supercritical carbon dioxide (SC-CO2): Experimental correlations and thermodynamic modeling, J. Mol. Liq. 297 (2020), 111740.
    [155]
    S. Zhang, W. Jin, Y. Yang, Simultaneous identification and determination of eleven tyrosine kinase inhibitors by supercritical fluid chromatography-mass spectrometry, Anal. Methods 11 (2019) 2211-2222.
    [156]
    H.A. Alhazmi, A. Ali Bokar Nasib, Y. Ali Musleh, et al., Application of drug-metal ion interaction principle in conductometric determination of imatinib, sorafenib, gefitinib and bosutinib, Open Chem. 18 (2020) 798-807.
    [157]
    L. Smy, A.J. Sadler, G.A. McMillin, Evaluation of imatinib concentrations in samples submitted for BCR-ABL1 or imatinib testing-evidence to support therapeutic drug monitoring for dose optimization? Ther. Drug Monit. 42 (2020) 559-564.
    [158]
    T. Saita, Y. Yamamoto, K. Hosoya, et al., An ultra-specific and sensitive sandwich ELISA for imatinib using two anti-imatinib antibodies, Anal. Chim. Acta 969 (2017) 72-78.
    [159]
    D. Dutta, S. Das, M. Ghosh, Validated HPTLC method for the determination of nintedanib in bulk drug, 22nd International Electronic Conference on Synthetic Organic Chemistry, November 15-December 15, 2018.
    [160]
    R. Bhole, T. Zombade, C.G. Bonde, et al., Identification and characterization of degradation products by using Ms-Ms studies for developed and validated stability indicating HPTLC method for estimation of Nintedanib Esylate in pharmaceutical dosage form, Eurasian J. Anal. Chem. 14 (2019) 60-70.
    [161]
    T. Sharma, R. Kaur Khurana, B. Borges, et al., An HPTLC densitometric method for simultaneous quantification of sorafenib tosylate and chrysin: Analytical method development, validation and applications, Microchem. J. 162 (2021), 105821.
    [162]
    D. Westover, J. Zugazagoitia, B.C. Cho, et al., Mechanisms of acquired resistance to first-and second-generation EGFR tyrosine kinase inhibitors, Ann. Oncol. 29 (2018) i10-i19.
    [163]
    C. Pottier, M. Fresnais, M. Gilon, et al., Tyrosine kinase inhibitors in cancer: Breakthrough and challenges of targeted therapy, Cancers 12 (2020), 731.
    [164]
    Q. Jiao, L. Bi, Y. Ren, et al., Advances in studies of tyrosine kinase inhibitors and their acquired resistance, Mol. Cancer 17 (2018), 36.
    [165]
    S. Wind, D. Schnell, T. Ebner, et al., Clinical pharmacokinetics and pharmacodynamics of afatinib, Clin. Pharmacokinet. 56 (2017) 235-250.
    [166]
    B.J. Smith, Y. Pithavala, H. Bu, et al., Pharmacokinetics, metabolism, and excretion of [14C] axitinib, a vascular endothelial growth factor receptor tyrosine kinase inhibitor, in humans, Drug Metab. Dispos. 42 (2014) 918-931.
    [167]
    R. Abbas, P.-H. Hsyu, Clinical pharmacokinetics and pharmacodynamics of bosutinib, Clin. Pharmacokinet. 55 (2016) 1191-1204.
    [168]
    T.R. Johnson, W. Tan, L. Goulet, et al., Metabolism, excretion and pharmacokinetics of [14C] crizotinib following oral administration to healthy subjects, Xenobiotica 45 (2015) 45-59.
    [169]
    N.P. van Erp, H. Gelderblom, H.J. Guchelaar, Clinical pharmacokinetics of tyrosine kinase inhibitors, Cancer Treat. Rev. 35 (2009) 692-706.
    [170]
    A. Puszkiel, G. Noe, A. Bellesoeur, et al., Clinical pharmacokinetics and pharmacodynamics of dabrafenib, Clin. Pharmacokinet. 58 (2019) 451-467.
    [171]
    D. Leveque, G. Becker, K. Bilger, et al., Clinical pharmacokinetics and pharmacodynamics of dasatinib, Clin. Pharmacokinet. 59 (2020) 849-856.
    [172]
    R.B. Verheijen, J.H. Beijnen, J.H.M. Schellens, et al., Clinical pharmacokinetics and pharmacodynamics of pazopanib: Towards optimized dosing, Clin. Pharmacokinet. 56 (2017) 987-997.
    [173]
    N.I. Narasimhan, D.J. Dorer, K. Niland, et al., Effects of food on the pharmacokinetics of ponatinib in healthy subjects, J. Clin. Pharm. Ther. 38 (2013) 440-444.
    [174]
    A. Hulin, J. Stocco, M. Bouattour, Clinical pharmacokinetics and pharmacodynamics of transarterial chemoembolization and targeted therapies in hepatocellular carcinoma, Clin. Pharmacokinet. 58 (2019) 983-1014.
    [175]
    Y. Ogama, T. Mineyama, A. Yamamoto, et al., A randomized dose-escalation study to assess the safety, tolerability, and pharmacokinetics of ruxolitinib (INC424) in healthy Japanese volunteers, Int. J. Hematol. 97 (2013) 351-359.
    [176]
    S. Johansson, J. Read, S. Oliver, et al., Pharmacokinetic evaluations of the co-administrations of vandetanib and metformin, digoxin, midazolam, omeprazole or ranitidine, Clin. Pharmacokinet. 53 (2014) 837-847.
    [177]
    H.I. Hurwitz, A. Dowlati, S. Saini, et al., Phase I trial of pazopanib in patients with advanced cancer, Clin. Cancer Res. 15 (2009) 4220-4227.
    [178]
    B. Peng, P. Lloyd, H. Schran, Clinical pharmacokinetics of imatinib, Clin. Pharmacokinet. 44 (2005) 879-894.
    [179]
    A.J. Montero, D. Kwon, A. Flores, et al., A phase I clinical, pharmacokinetic, and pharmacodynamic study of weekly or every three week ixabepilone and daily sunitinib in patients with advanced solid tumors, Clin. Cancer Res. 22 (2016) 3209-3217.
    [180]
    C. Lathia, J. Lettieri, F. Cihon, et al., Lack of effect of ketoconazole-mediated CYP3A inhibition on sorafenib clinical pharmacokinetics, Cancer Chemother. Pharmacol. 57 (2006) 685-692.
    [181]
    X. Tian, H. Zhang, T. Heimbach, et al., Clinical pharmacokinetic and pharmacodynamic overview of nilotinib, a selective tyrosine kinase inhibitor, J. Clin. Pharmacol. 58 (2018) 1533-1540.
    [182]
    M. Fukudo, Y. Ikemi, Y. Togashi, et al., Population pharmacokinetics/pharmacodynamics of erlotinib and pharmacogenomic analysis of plasma and cerebrospinal fluid drug concentrations in Japanese patients with non-small cell lung cancer, Clin. Pharmacokinet. 52 (2013) 593-609.
    [183]
    B. Thiessen, C. Stewart, M. Tsao, et al., A phase I/II trial of GW572016 (lapatinib) in recurrent glioblastoma multiforme: Clinical outcomes, pharmacokinetics and molecular correlation, Cancer Chemother. Pharmacol. 65 (2010) 353-361.
    [184]
    H.C. Swaisland, M. Ranson, R.P. Smith, et al., Pharmacokinetic drug interactions of gefitinib with rifampicin, itraconazole and metoprolol, Clin. Pharmacokinet. 44 (2005) 1067-1081.
    [185]
    A. Arora, E.M. Scholar, Role of tyrosine kinase inhibitors in cancer therapy, J. Pharmacol. Exp. Ther. 315 (2005) 971-979.
    [186]
    D.H. Josephs, D.S. Fisher, J. Spicer, et al., Clinical pharmacokinetics of tyrosine kinase inhibitors: Implications for therapeutic drug monitoring, Ther. Drug Monit. 35 (2013) 562-587.
    [187]
    P. Herviou, E. Thivat, D. Richard, et al., Therapeutic drug monitoring and tyrosine kinase inhibitors, Oncol. Lett. 12 (2016) 1223-1232.
    [188]
    R. Lewis, R. Bruggemann, C. Padoin, et al., Triazole antifungal therapeutic drug monitoring, Sixth European Conference on Infections in Leukaemia Meeting, Sophia Antipolis, France, September 11-12, 2015.
    [189]
    R.A. Ghiculescu, Therapeutic drug monitoring: Which drugs, why, when and how to do it, Aust. Prescr. 31 (2008) 42-44.
    [190]
    M. Baccarani, M.W. Deininger, G. Rosti, et al., European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013, Blood 122 (2013) 872-884.
    [191]
    M. Garcia-Ferrer, A. Wojnicz, G. Mejia, et al., Utility of therapeutic drug monitoring of imatinib, nilotinib, and dasatinib in chronic myeloid leukemia: A systematic review and meta-analysis, Clin. Ther. 41 (2019) 2558-2570.e7.
    [192]
    M. Miura, Therapeutic drug monitoring of imatinib, nilotinib, and dasatinib for patients with chronic myeloid leukemia, Biol. Pharm. Bull. 38 (2015) 645-654.
  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Figures(1)

    Article Metrics

    Article views (119) PDF downloads(16) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return