We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Simultaneous determination of six tyrosine kinase inhibitors in human plasma using HPLC-Q-Orbitrap mass spectrometry

    Mao-Wei Ni

    Zhejiang Cancer Hospital, Hangzhou, Zhejiang, PR China

    Key Laboratory Diagnosis & Treatment Technology on Thoracic Oncology, Zhejiang Province, Hangzhou 310022, PR China

    ,
    Jie Zhou

    Zhejiang Academy of Traditional Chinese Medicine, Development & Research Center of Official Silkworm Resources, Hangzhou, Zhejiang, PR China

    ,
    Hui Li

    Zhejiang Cancer Hospital, Hangzhou, Zhejiang, PR China

    ,
    Wei Chen

    Zhejiang Cancer Hospital, Hangzhou, Zhejiang, PR China

    ,
    Han-Zhou Mou

    Zhejiang Cancer Hospital, Hangzhou, Zhejiang, PR China

    &
    Zhi-Guo Zheng

    *Author for correspondence:

    E-mail Address: zhengzg@zjcc.org.cn

    Zhejiang Cancer Hospital, Hangzhou, Zhejiang, PR China

    Published Online:https://doi.org/10.4155/bio-2017-0031

    Aim: Gefitinib, erlotinib, icotinib, crizotinib, lapatinib and apatinib are targeted cancer therapy agents acting through inhibition of tyrosine kinase. Method for quantifying these six drugs in human plasma of patients was required. Materials & methods: An HPLC-Q-Orbitrap method (based on HPLC–MS/MS) was developed and validated for the simultaneous detection and quantitation of six tyrosine kinase inhibitors in human plasma. Sample was extracted by liquid–liquid extraction (ethyl acetate: tert-Butyl methyl ether, 1:1 v/v). The method shows a high level of accuracy and reproducibility. The lower limit of quantification was 0.02 ng/ml for apatinib, 0.1 ng/ml for crizotinib, 2.0 ng/ml for lapatinib and 0.05 ng/ml for erlotinib, gefitinib and icotinib. This method was successfully used for apatinib monitoring in plasma of patients with NSCLC. Conclusion: This simple and reproducible method has potential for monitoring of tyrosine kinase inhibitors in patients’ plasma.

    References

    • 1 Moyer JD, Barbacci EG, Iwata KK et al. Induction of apoptosis and cell cycle arrest by CP-358,774, an inhibitor of epidermal growth factor receptor tyrosine kinase. Cancer Res. 57(21), 4838–4848 (1997).
    • 2 Glade-Bender J, Kandel JJ, Yamashiro DJ. VEGF blocking therapy in the treatment of cancer. Expert Opin. Biol. Ther. 3(2), 263–276 (2003).
    • 3 Kwak EL, Bang YJ, Camidge DR et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N. Engl. J. Med. 363(18), 1693–1703 (2010).
    • 4 Hoelder S, Clarke PA, Workman P. Discovery of small molecule cancer drugs: successes, challenges and opportunities. Mol. Oncol. 6(2), 155–176 (2012).
    • 5 Kris MG, Natale RB, Herbst RS et al. Efficacy of gefitinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase, in symptomatic patients with non-small cell lung cancer: a randomized trial. JAMA 290(16), 2149–2158 (2003).
    • 6 Ciardiello F, Tortora G. A novel approach in the treatment of cancer: targeting the epidermal growth factor receptor. Clin. Cancer Res. 7(10), 2958–2970 (2001).
    • 7 Hidalgo M, Siu LL, Nemunaitis J et al. Phase I and pharmacologic study of OSI-774, an epidermal growth factor receptor tyrosine kinase inhibitor, in patients with advanced solid malignancies. J. Clin. Oncol. 19(13), 3267–3279 (2001).
    • 8 Huo ZJ, Wang SJ, Wang ZQ et al. Novel nanosystem to enhance the antitumor activity of lapatinib in breast cancer treatment: therapeutic efficacy evaluation. Cancer Sci. 106(10), 1429–1437 (2015).
    • 9 Ding L, Li QJ, You KY, Jiang ZM, Yao HR. The use of apatinib in treating nonsmall-cell lung cancer case report and review of literature. Medicine 95(20), e3598 (2016).
    • 10 Sandborn WJ, Hanauer SB, Pierre-Louis B, Lichtenstein GR. certolizumab pegol plasma concentration and clinical remission in Crohn's disease. Gastroenterology 142(Suppl. 1), S563 (2012).
    • 11 Corsonello A, Pedone C, Incalz RA. Age-related pharmacokinetic and pharmacodynamic changes and related risk of adverse drug reactions. Curr. Med. Chem. 17, 14 (2010).
    • 12 Cimino GD, Pan CX, Henderson PT. Personalized medicine for targeted and platinum-based chemotherapy of lung and bladder cancer. Bioanalysis 5(3), 369–391 (2013).
    • 13 Zheng N, Zhao C, He XR et al. 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. 1011, 215–222 (2016).
    • 14 Lankheet NA, Schaake EE, Rosing H et al. Quantitative determination of erlotinib and O-desmethyl erlotinib in human EDTA plasma and lung tumor tissue. Bioanalysis 4(21), 2563–2577 (2012).
    • 15 Liu D, Jiang J, Hu P, Tan F, Wang Y. Quantitative determination of icotinib in human plasma and urine using liquid chromatography coupled to tandem mass spectrometry. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 877(30), 3781–3786 (2009).
    • 16 Roberts MS, Turner DC, Broniscer A, Stewart CF. Determination of crizotinib in human and mouse plasma by liquid chromatography electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS). J. Chromatogr. B 960, 151–157 (2014).
    • 17 Musijowski J, Filist M, Rudzki PJ. Sensitive single quadrupole LC/MS method for determination of lapatinib in human plasma. Acta Pol. Pharm. 71(6), 1029–1036 (2014).
    • 18 Ding J, Chen X, Dai X, Zhong D. Simultaneous determination of apatinib and its four major metabolites in human plasma using liquid chromatography–tandem mass spectrometry and its application to a pharmacokinetic study. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 895–896, 108–115 (2012).
    • 19 Venugopal N, Reddy AVB, Madhavi G. Development and validation of a systematic UPLC–MS/MS method for simultaneous determination of three phenol impurities in ritonavir. J. Pharmaceut. Biomed. 90, 127–133 (2014).
    • 20 ThermoFisher. www.thermofisher.com/content/dam/tfs/ATG/CMD/CMD%20Documents/HUPO-2011-Hao-P1123-Final.pdf.
    • 21 Sturm RM, Jones BR, Mulvana DE, Lowes S. HRMS using a Q-Exactive series mass spectrometer for regulated quantitative bioanalysis: how, when, and why to implement. Bioanalysis 8(16), 1709–1721 (2016).
    • 22 Guidance for Industry Bioanalytical Method Validation. US Department of Health and Human Services, US FDA (2001). www.fda.gov/downloads/Drugs/Guidance/ucm070107.pdf.
    • 23 Dolan JW. When should an internal standard be used? LCGC N. Am. 30(6), 474 (2012).
    • 24 Lynch KL. CLSI C62-A: a new standard for clinical mass spectrometry. Clin. Chem. 62(1), 24–29 (2016).
    • 25 Yang ZZ, Wang L, Xu MC, Gu JK, Yu LS, Zeng S. Simultaneous analysis of gemfibrozil, morphine, and its two active metabolites in different mouse brain structures using solid-phase extraction with ultra-high performance liquid chromatography and tandem mass spectrometry with a deuterated internal standard. J. Sep. Sci. 39(11), 2087–2096 (2016).
    • 26 Guo C, Shi F, Jiang S et al. Simultaneous identification, confirmation and quantitation of illegal adulterated antidiabetics in herbal medicines and dietary supplements using high-resolution benchtop quadrupole-Orbitrap mass spectrometry. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 967, 174–182 (2014).