Development of an LC–MS/MS method for quantifying two main metabolites of abivertinib
in human plasma
Xin Zhenga
, Weicong Wangb
, Yanbao Zhanga
, Yuxiang Mac
, Hongyun Zhaoc
,Huitao Gaoa
,Pei
Hua
, Ji Jianga*
aClinical Pharmacology Research Center, Peking Union Medical College Hospital, Peking
Union Medical College and Chinese Academy of Medical Sciences, China
bDepartment of Clinical Trial Center, China National Clinical Research Center for
Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, China
cDepartment of Medical Oncology, Sun Yatsen University Cancer Center, State Key
Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer
Medicine, Guangzhou, China
Abstract
Abivertinib represents a highly selective irreversible epidermal growth factor receptor
tyrosine kinase inhibitor. Two major metabolites of abivertinib, M7 and MII-6, were detected
in human plasma, which are recommended to be monitored for safety reasons in clinical trial.
A high throughput quantification method utilizing liquid chromatography-tandem mass
spectrometry was designed and verified to quantify abivertinib’s primary metabolites in
human plasma. Solid phase extraction (SPE) was used to process plasma, and then the
analytes underwent a gradient elution separation in an Aquity UPLC BEH C18 column (1.7
µm, 2.1×50 mm) with mobile phase A (10mM ammonium acetate comprised of 0.1% formic
acid) and mobile phase B (methanol : acetonitrile (2:8, v/v) with 0.1% formic acid). Ion
transitions of M7 (m/z 490.2→405.1) and MII-6(m/z 476.2→391.1) were monitored under
the mode of multiple reaction monitoring (MRM) and electrospray ionization in positive ion
mode. This simultaneous determination method was found to have acceptable precision,
accuracy and linearity in 0.5-500 ng/mL range for M7 as well as the 0.5-500 ng/mL range for
MII-6, accompanied with a mild matrix effect but high recovery. Further stability
This article is protected by copyright. All rights reserved.
assessments indicated that both analytes remained stable throughout the entire experimental
process beginning from harvesting whole blood to plasma extracting and analyzing.
Keywords: Abivertinib; metabolites; UPLC-MS/MS; Human plasma; Pharmacokinetics
1. Introduction
Lung cancer is a primary cause of death associated with cancer in US and China. One
extremely frequently occurring subtype of lung cancer, non-small cell lung cancer (NSCLC),
is responsible for an estimated 85% of all incidences of pulmonary malignancies(Jemal A et
al.,2011;Haghgoo SM et al.,2015;Reck M et al.,2013). Epidermal growth factor receptor
(EGFR) mutations, mostly seen on exon 21 or exon 19, has been identified as an oncogenic
driver that culminates in continuous cell signaling activation as well as enhanced cell
proliferation and metastasis.(D.S. Salomon et al.,1995) Consequently, EGFR tyrosine kinase
inhibitors (EGFR-TKIs), such as afatinib, erlotinib and gefitinib, have brought an inspiring
revolution in the treatment of NSCLC patients who possess mutations sensitive to EGFR.
EGFR-TKIs treatment in these subsets of the cancer cohort invoke better response and
superior overall survival rates as well as progression-free survival rates in contrast to
chemotherapy involving platinum compounds.
Despite great benefits gained from the first and second generation of EGFR TKIs, acquired
resistance and toxicity have been found in most cases, with disease progression observed
mostly within 9-12 months after treatment (Mok TS et al.,2009;Rosell R et al.,2012;Mok TS
et al.,2017). A secondary mutation in gatekeeper T790M, which occurs in 49% to 60% of
patients, has been identified to be of critical importance in the development of resistance
towards EGFR-TKI therapy (Sequist LV et al.,2011). For this reason, third-generation EGFR
TKIs were formulated to overcome this hurdle. Abivertinib represents a novel
third-generation, irreversible and highly selective inhibitor of EGFR for both T790M
resistance mutations and exons EGFR19 and 21 sensitive mutations (Xu X et al., 2016).
Metabolite profiling was conducted in mouse, rat, dog, monkey, and human liver microsomes.
Six metabolites were found in preclinical investigations, which were M2、M4、M1、M7、
This article is protected by copyright. All rights reserved.
MII-6 and MII-2.(Xu X et al., 2016). In the further study, the exposure of other metabolites
was relatively low. Given the favorable safety and efficacy data demonstrated in Phase I and
Phase II clinical trials, marketing application of NMPA for abivertinib was filed (National
Medical Products Administration, NMPA) in 2018.
In human pharmacokinetic studies, the exposure (AUC) in plasma of the two major
metabolites of abivertinib, M7 and MII-6, were found both around 10% of parent drug. For
safety reasons, these two metabolites are recommended to be measured in clinical trials.
The current study elucidates the development of an uncomplicated, quick and accurate
high-performing liquid chromatographic tandem mass spectrometric (UPLC–MS/MS)
method to quantitate abivertinib’s major metabolites, M7 and MII-6, in human plasma for the
first time. We then successfully applied our method in the analysis of M7 and MII-6
concentrations in plasma samples of advanced NSCLC patients. This method provides a
simple way for detecting M7 and MII-6 which has a broad linear range and a short analytical
run-time.
2. Experimental methods
2.1. Standards, reagents & materials
Standards of M7 (purity 96.3%), MII-6 (purity 98.6%) and Internal standard(purity 97.2%)
were supplied by Hangzhou ACEA Pharmaceutical Research Co., Ltd (Hangzhou, China).,
who also sponsored abivertinib pharmacokinetic research in cancer volunteers. HPLC-grade
methanol and acetonitrile were purchased from Fisher, USA. Ammonium acetate was bought
from Sinopharm Chemical Reagent Co., Ltd. (Beijing, China). Sigma-Aldrich Corp. (St.
Louis, MO, USA) supplied the formic acid. A water purification Milli-Q
system (Millipore,
MA, USA) was used to purify all water used in this study. Healthy volunteers supplied
plasma which was used as controls and was stored in heparin sodium tubes and kept in a
-30 °C freezer until further use.
2.2. Liquid chromatographic and mass spectrometric conditions
The ACQUITY UPLC system (Waters Corp., USA) was used to perform the LC separation
and comprised of an autosampler, a binary solvent delivery manager and a 5500 Qtrap mass
This article is protected by copyright. All rights reserved.
spectrometer (AB Sciex, USA) with an electrospray ionization (ESI) interface were used to
perform the mass spectrometric detection. LC Separation took place via a gradient elusion
with an Aquity UPLC BEH C18 column (1.7 μm, 2.1×50 mm) consisting of 10mM
ammonium acetate with 0.1% formic acid (A) and methanol:acetonitrile (2:8, v/v) with 0.1%
formic acid (B), at the flow rate of 0.4 mL/min.
The gradient of the mobile phase initialed at 30% mobile phase B for 0.8 min, and then the
gradient elevated to 35% within 2.5 min, subsequently, mobile phase B proportion remaining
in the mobile phase was programmed to run over 0.01 minutes at 90%, while maintaining this
state for the next 10 seconds. The mobile phase was finally reprogrammed to 30% mobile
phase B in the last 0.5 min. Each sample ran for a total of 3.5 min. Auto-sampler and column
temperatures were maintained consistently at 10 °C and 35 °C, respectively.
Optimal source/gas parameters were set as follows: gas1, 60; gas2, 60; collision gas, 7;
curtain gas, nitrogen, 45; temperature, 550 °C. Each transition had a dwell time of 100 ms on
multiple reaction monitoring mode. Table 1 depicts several other optimized ionization
conditions. Figure 1 depicts the product ion spectra of the major metabolites M7, MII-6 and
the internal standard. The Analyst Data Acquisition and Processing software (Version 1.5.1,
AB Sciex, USA) was used to collect data.
2.3. Stock solutions, calibration standards (CS) and quality control (QC) samples
1 mg/mL stock solutions of M7 and MII-6 for CS and QC were prepared separately in
dimethyl sulfoxide (DMSO) and were kept at -80 °C. Methanol-acetonitrile-water (7:28:65,
v/v/v) were used to further dilute the stock solutions to prepare working solutions. M7 and
MII-6 concentrations of working solutions were 20, 40, 200, 800, 2000, 4000, 10000, 20000
ng/mL, and the concentrations of QC samples were 60, 1000 and 16000 ng/mL. QC and CS
samples in human plasma were prepared by diluting 25 μL working solution of M7 and MII-6
with 975 μL drug-free human plasma. Finally, M7 and MII-6 concentration levels of CS
samples in plasma were 0.5, 1, 5, 20, 50, 100, 250, 500 ng/mL, and the concentrations of QC
samples were 1.5, 25 and 400 ng/mL. Internal standard (IS) working solution (ISw, 1.5
This article is protected by copyright. All rights reserved.
μg/mL) was diluted with methanol-acetonitrile-water (7:28:65, v/v/v) from 1 mg/mL IS stock
solution.
2.4. Sample preparation
Solid-phase extraction was used to prepare all plasma samples. Add 1 mL methanol to the
Oasis® HLB 96-well plate 30 m (10 mg), drip dry and then add 1 mL water. Briefly, 50 μL
plasma aliquots was first spiked into an Eppendorf micro tube, and mixed with a vortex
machine for 15 seconds with 50 μL IS and 250 μL pure water. After 15 seconds, Solid-phase
extraction plate was conditioned with 350 μL aliquot plasma sample and washed with
methanol-water (1:9, v/v) for twice. Then, 800 μL methanol was used to elute the analytes
and evaporated with Nitrogen at 40 °C. Finally, analytes were dissolved with 500 L solution
comprised of methanol-acetonitrile-water (7:28:65, v/v/v). Detection of the plasma samples
was performed by injecting 10 µL of the solution into the LC–MS/MS system.
2.5. Data analysis
For acquiring chromatography–mass spectrometric data, Analyst® software (version 1.5.1,
AB SCIEX) was used. The peak area ratio of analyte to the corresponding IS being the
explained variable (using a weighting factor of 1/x2
) and concentrations being the explanatory
variable were the parameters for linear regression. Acceptable biases were no more than 15%
for calibration standards against the nominal concentrations in each calibration curve, while
20% was taken to be acceptable for LLOQ. Each calibration curve should have a regression
coefficient of more than 0.98 while at least more than 75% of the calibration standards were
required to surpass the stipulated criterion in each analytical run.
2.6. Method validation
Validation of this developed method was conducted according to the guidelines of the US
Food and Drug Administration (FDA) (2013), European Medicines Agency (EMA) (2011)
and NMPA guidelines for the validation of bioanalytical methods (2005), Pharmacopoeia of
the People’s Republic of China (2015), the LC–MS/MS procedure allowed validation in terms
This article is protected by copyright. All rights reserved.
of linearity, sensitivity, specificity, selectivity, accuracy, precision, recovery, dilution
integrity, stability, matrix effect, and carry-over.
The linearity of each calibration curve was determined by plotting the peak area ratio (y) of
the analyte/IS versus the nominal concentration (x) of the analyte with weighed (1/x2
) least
square linear regression. The selectivity was assessed by analyzing six individual human
plasma lots to determine the presence of endogenous compound interference.
Accuracy and precision of the intra- and inter-day assay were determined with six duplicates
of LLOQ and QC samples in plasma (low, medium, and high concentration level) over three
successive batch runs. Relative error (RE%) and relative standard deviation (RSD%) were
used to evaluate the intra- and inter-day accuracy and precision, respectively. RSD% and
RE% should be within 15% for all other calibration levels in the intra-batch and inter-batch
assays but within 20% for LLOQ.
The extraction recovery was calculated by contrasting the average peak area of analyte spiked
post-extraction to those that spiked prior to extraction at three different levels of QC
concentrations, with six replicates each.
The matrix effect was determined through calculation of the ratio of the peak area of
matrix-free solutions to the peak area of analyte spiked post-extraction at three different QC
concentrations using 6 individual donor blank matrix lots. The matrix effect RSD% was kept
at no more than 15%.
The stabilities of M7 and MII-6 in reconstituted solution and in biological matrix at various
storage conditions were investigated using samples maintained under the following
conditions: Long-term stability (-80 °C for 6 months), short-term stability (room temperature
for 8 hours), freeze-thaw stability (six freeze-thaw cycles) and auto-sampler stability (10 °C
for 72 hours). QC samples of low, medium and high concentration levels were assessed
through six replicates each. Stability was assumed if the RE% and RSD% was no more than
15%.
The dilution integrity was evaluated at the concentration levels of 1000 ng/mL for M7 and
1000 ng/mL for MII-6 utilizing a dilution factor of 10, respectively. Six replicate samples
were prepared and their concentrations were calculated, by applying the dilution factor of 10
This article is protected by copyright. All rights reserved.
against the freshly prepared calibration curve for M7 and MII-6. The RE% and RSD% for six
replicates post-dilution was maintained at ≤15%.
Samples that did not contain internal standards or analytes (double blank plasma samples)
were inserted after the upper limit of quantification (ULOQ) in each validation batch in order
to assess the carry-over of this method. The threshold of carry-over should not exceed 5% of
the IS and 20% of the LLOQ in the blank samples.
2.7. Method application
The validated LC–MS/MS method was then utilized for investigations of the
pharmacokinetic characteristics of M7 and MII-6 in a phase I clinical trial which was
conducted between October 2014 and December 2016 (data cutoff). This is a randomized,
open, single-center dose escalation clinical trial, which was conducted in accordance with the
principles of the Declaration of Helsinki and the Good Clinical Practice (GCP) and approved
by the Ethics Committee of Sun Yat-sen University Cancer Center (NCT02274337).
During the study, the improved Fibonacci method was adopted in the dose escalation phase.
A plurality of patient cohorts received abivertinib capsules of 7 oral dose levels, 50 mg, 100
mg, 200 mg, 350 mg, 500 mg, 550 mg and 600 mg, respectively. Each patient was initially
administered a single dose of abivertinib, and received the PK assessment during the
following 7 days; multiple dose assessment was applied thereafter. Heparin sodium tubes
were used to harvest blood samples at the following time points: Day 1 administration: within
5 minutes before the first dose, 1, 2, 3, 4, 5, 6, 8, 12, 24 and 48 hours after the first dose. For
QD multiple dose ,within 5 minutes before the first dose on day 8,15 and 22; Drug
administration on day 28: pre-dose, 1, 2, 3, 4, 5, 6, 8, 12, 24, and 48 hours post-dose. A total
of 25 blood samples were collected from each subject. Harvested blood samples were then
centrifuged for 10 minutes at speeds of 13300 g with a temperature of 4 °C to obtain plasma.
An ultra-deep freezer was used to store all plasma samples at −80 °C until required for
further analysis.
This article is protected by copyright. All rights reserved.
3. Results and discussion
3.1. Development of method
3.1.1. Optimization of mass spectrometric parameters
Sensitive and selective quantification of abivertinib’s main metabolites were achieved by the
MRM mode. Method parameter settings were optimized for all target analytes to obtain the
maximum abundance of product ions using both negative and positive ionization modes. The
product and precursor ions were determined by injecting 10 ng/mL of each analyte mixed
with methanol-water (50:50, v/v) into the mass spectrometer through a syringe pump
functioning at a continuous flow rate of 10 μL/min. The precursor ions for M7, MII-6 and
IS-0010 were observed at m/z 490.2, 476.2 and 470.2. respectively. The major product ions
of M7, MII-6 and IS-0010 at m/z 405.1, 391.1 and 385.1, respectively, were selected to
quantify.
MS parameters including the ion source temperature (◦C), collision energy (eV), (V) were
optimized separately in order to obtain the highest ion abundance and most stable compound.
Table 1 depicts the optimized ionization conditions, while Figure 1 illustrates the product ion
spectra of M7, MII-6, and IS.
3.1.2. Chromatographic condition optimization
To improve the peaks resolution and the peak-to-noise ratio, different volume ratios of
acetonitrile and methanol were tested as the organic mobile phase, and finally regarding its
best separating effect and low background noise the organic mobile phase composited by
acetonitrile: methanol (8:2, v/v) was selected.
Upon optimizing the LC separation gradient program, chromatographic retention times for
M7, MII-6 and IS-0010 were at 2.00, 1.40 and 1.64 min, all of which maintained acceptable
peak symmetry and shape. With these results, our method is proven to be high-throughput
and suitable for use to analyze plasma in a pharmacokinetic study.
This article is protected by copyright. All rights reserved.
3.2. Method validation
3.2.1. Specificity and selectivity
Typical chromatograms obtained from LLOQ sample, clinical sample extracted 4 hours
post-administration of 50 mg abivertinib capsule as well as blank sample are shown in Fig. 2.
In processed blank samples, no endogenous interference was observed at the analyte retention
times (2.00 min for M7, 1.40 for MII-6 and 1.64 min for IS), which suggested that the assay
demonstrated the enough specificity. Plasma samples which were separately spiked with M7
and MII-6 at ULOQ concentrations, and with IS only are used to investigate the selectivity.
No cross-analytes interference was observed in all test samples.
3.2.2. Linearity
Linearity for M7 and MII6 both was obtained the concentration ranged from 0.5 to 500
ng/mL. A least-squares linear regression with a weighting factor of 1/x2 was applied to assess
the calibration curves and Correlation coefficient (r2
) of all standard curves was equal to or
greater than 0.99. Eight non-zero calibration standards were used for each calibration curve
and at least 75% of the calibration standards were within 15% of the nominal concentrations,
or within 20% for the LLOQ, in three separate runs.
3.2.3. LLOQ, Accuracy and precision
For both M7 and MII-6, the signals at LLOQ level were both at least 5 times as high as the
signal in the blank sample. The signal-to-noise ratios were 17.9 and 25.1 for M7 and MII-6,
respectively. Representative chromatogram of LLOQ sample is showed in Figure 2.
The inter- and intra-batch accuracy and precision data of M7 and MII-6 are presented in
Table 2. Accuracy values (expressed as the bias) and precision values (expressed as the
coefficient of variation (CV)) were kept within 20% for the LLOQ and within 15% for high,
medium, and low QC’s. This indicates that the current method met the criteria of accuracy
and precision.
This article is protected by copyright. All rights reserved.
3.2.4. Matrix effect and extraction recovery
The matrix factor (MF) was calculated by dividing the peak area in matrix present sample to
the peak area in neat solution (matrix absent). The IS-normalized MF of M7 was ranged from
101% to 103%, and 102% for MII-6 (Table 3). The CVs of the IS-normalized MF for the six
batches of M7 and MII-6 was lower than 5.0 % and 2.7 %, respectively. Based on these
results, it was concluded that the matrix has no effect on the precision of the methods for all
three matrices.
The average extraction recoveries derived from three separate QC plasma sample
concentrations were within the range of 88.0–91.8% for M7, 88.2–90.8% for MII-6, 96.7%
for IS, respectively.
This highlights that solid-phase extraction appears to be an easily carried out and fast
procedure for human plasma analytes. Overall, the small extraction losses and the negligible
relevant matrix effects were invaluable in validating this method.
3.2.5. Stability
The storage and processing conditions of stability tests were set to cover different storage and
processing conditions expected during clinical samples routine analysis.
M7 and MII-6 remained stable after being stored at room temperature (25 ℃) for 8 hours or
being stored at -80 ℃ for an extended period of 12 months.
Plasma samples subjected to six freeze-thaw cycles still remained stable following from
-80℃ to 25℃.
M7 and MII-6 also showed good stability at 10℃ for 72 hours after sample preparation
respectively.
Compounds that were stable ensured the veracity of quantitation results obtained by this
method. The results of the stability are presented in Table 4.
3.2.6. Dilution integrity
Six replicates of human plasma samples spiked with concentrations of M7 and MII-6 around
2 times the ULOQ were diluted 10-fold with pooled blank human plasma. The bias was
This article is protected by copyright. All rights reserved.
within 15% and the CVs were ≤15% for both analytes. Therefore, adequate dilution of the
plasma samples with the concentration higher than ULOQ was able to be achieved by
utilizing the dilution factors.
3.2.7. Carryover
Two double blank samples (without M7/MII-6 and IS) were analyzed directly follow an
upper limit of quantification sample to determine carry-over effects. No peaks of all analytes
were present at the retention time in the condition of the present method, indicating that the
carryovers of this method may be neglected.
3.3. Method Application
After a successful validation procedure, this developed LC–MS/MS method was used to
support a clinical study in which the plasma pharmacokinetic profiles of M7 and MII6 in
NSCLC patients were investigated. 52 subjects were enrolled in the study and the center
participated in the testing of samples from 19 patients. The robustness of the method was
observed with 859 plasma samples that were successfully analyzed with a lack of significant
issue or problems. Typical plasma concentration-time curves of M7 and MII-6 in cancer
volunteers post oral administration of 500 mg and 600 mg of M7 and MII-6 were depicted in
Figure 3.
Table 5 and Table 6 depict the corresponding non-compartmental analysis (NCA)
pharmacokinetic profiles of 500 mg, 600 mg M7 and MII-6. M7 in the dose of 500 mg and
600 mg achieved a maximum concentration (Cmax) of 169.3±22.7 ng/mL and 168.6±80.2
ng/mL, respectively. For MII-6, Cmax was 192.7±21.0 and 98.9±67.5, respectively.
4. Conclusion
In this study, a highly specific and sensitive LC-MS/MS method for simultaneous
quantification of two main metabolites of the novel anticancer agent (abivertinib) was
developed. This method which involves efficient preparation provided broad linearity and
possessed a low LLOQ (0.5 ng/mL for both M7 and MII-6). The assay was validated in a
This article is protected by copyright. All rights reserved.
range of 0.5-100 ng/mL in plasma. Samples with concentrations above the ULOQ can
reliably be diluted 10 times to quantify the analytes in the validated concentration ranges. The
presented method in this study assay was used to support a clinical trial of abivertinib
(NCT02274337).
5. Future perspective
EGFR TKI inhibitors have shown good therapeutic effects and higher overall survival in the
treatment of various cancers such as lung cancer. We have benefited greatly from the
discovery of first – and second-generation EGFR TKIs. Abivertinib, a third-generation,
high-choice EGFR inhibitor designed to overcome resistance, is currently being evaluated in
the second phase of drug development. To support clinical studies, a simple, quick and
accurate UPLC-MS /MS method for the detection of abivertinib’s active metabolites M7 and
MII-6 in human plasma was described. This method is suitable for future studies on
pharmacokinetics of abivertinib metabolites in patients.
Competing financial interests
The authors declare no conflicts of interest.
Funding information
13th Five-Year” National Major New Drug Projects (No: 2018ZX09734006-001)
This article is protected by copyright. All rights reserved.
Reference
Jemal, A., Bray, F., Center, MM., Ferlay,J., Ward,E., Forman,D. (2011). Global cancer
statistic. CA Cancer J Clin, 61, 69-90. https://doi.org/10.3322/caac.20107
Haghgoo, SM., Allameh, A., Mortaz, E., Garssen, J., Folkerts, G., Barnes,PJ., Adcock, IM.
(2015). Pharmacogenomics and targeted therapy of cancer: Focusing on non-small cell lung
cancer. Eur J Pharmacol, 754, 82-91. https://doi.org/10.1016/j.ejphar.2015.02.029
Reck, M., Heigener, DF., Mok,T., Soria, JC., Rabe, KF. (2013). Management of
non-small-cell lung cancer: recent developments. Lancet, 382, 709-719.
https://doi.org/10.1016/S0140-6736(13)61502-0
Salomon, DS., Brandt, R., Ciardiello, F., Normanno, N. (1995). Epidermal growth factor
related peptides and their receptors in human malignancies. Crit Rev Oncol Hematol, 19,
183–232. https://doi.org/10.1016/1040-8428(94)00144-I
Mok, TS., Wu, YL., Thongprasert, S., Yang, CH., Chu, DT., Saijo, N., Fukuoka, M. (2009).
Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med, 361(10),
947-57. https://doi.org/10.1056/NEJMoa0810699
Rosell, R., Carcereny, E., Gervais, R., Vergnenegre, A., Massuti, B., Felip, E., Spanish Lung
Cancer Group in collaboration with Groupe Français de Pneumo-Cancérologie and
Associazione Italiana Oncologia Toracica.(2012). Erlotinib versus standard chemotherapy as
first-line treatment for European patients with advanced EGFR mutation-positive
non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial.
Lancet Oncol, 13(3), 239-46. https://doi.org/10.1016/S1470-2045(11)70393-X
Mok, TS., Wu, Y-L., Ahn, M-J., Garassino, MC., Kim, HR., Ramalingam, SS., AURA3
Investigators.(2017). Osimertinib or Platinum-Pemetrexed in EGFR T790M-Positive Lung
Cancer. N Engl J Med, 376(7), 629-640. https://doi.org/10.1056/NEJMoa1612674
Sequist, LV., Waltman, BA., Dias-Santagata, D., Digumarthy, S., Turke, AB., Fidias, P.,
Engelman, JA. (2011). Genotypic and histological evolution of lung cancers acquiring
resistance to EGFR inhibitors . Sci Transl Med, 3(75), 75ra26 .
https://doi.org/10.1126/scitranslmed.3002003
Xu, X., Mao, L., Xu, W., Tang, W., Zhang, X., Xi, B., Zhang, L.(2016). AC0010, an
Irreversible EGFR Inhibitor Selectively Targeting Mutated EGFR and Overcoming
This article is protected by copyright. All rights reserved.
T790M-Induced Resistance in Animal Models and Lung Cancer Patients. Mol Cancer Ther,
15 (11) , 2586-2597. https://doi.org/10.1158/1535-7163.MCT-16-0281
Food and Drug Administration (FDA), Draft Guidance for Industry inBioanalytical Method
Validation. 2013. Available at:
https://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/
ucm368107.pdf.
European Agency for the Evaluation of Medicinal Products (EMEA),
Guideline on Bioanalytical Method
Validation. 2011. Available at: http://www.ema.europa.eu/docs/en GB/document
library/Scientific guideline/2011/08/WC500109686.pdf.
China food and drug administration, 2005. Available at:
http://www.sda.gov.cn/gsz05106/08.pdf.
This article is protected by copyright. All rights reserved.
Table 1 Optimized parameters on mass to charge (m/z) transition and DP, EP, CE and
CXP of M7, MII-6 and IS
Analytes m/z DP (V) EP (V) CE (V) CXP (V)
M7 490.2/405.1 135 11 46 27
MII-6 476.2/391.1 210 10 43 26
IS-0010 470.2/385.1 134 12 44 28
Table 3 Matrix effect and extraction recovery of M7 and MII-6 in human plasma (n=6)
Analytes Concentration Matrix effect Extraction recovery
(ng/mL) Mean±SD (%) RSD% Mean RSD%
M7 1.5 102±2.73 2.7 88.0 4.9
25 101±0.602 0.6 91.8 3.9
400 103±1.07 1.0 88.9 2.0
MII-6 1.5 102±2.73 2.7 88.2 2.3
25 102±0.973 1.0 90.8 3.4
400 102±0.867 0.8 88.5 1.9
IS-0010 96.7 3.3
This article is protected by copyright. All rights reserved.
Table 4 Stability Assessments for M7 and MII-6 in human plasma (n = 6)
Analytes Stability types
Nominal
concentration
(ng/ml)
Mean RSD% RE%
M7 Short-term
(room
temperature
for 8h)
1.5 1.49 2.5 -0.6
25 25.2 0.9 0.9
400 403 2.2 0.7
Auto-sampler
(10℃ for 72h)
1.5 1.28 4.8 -14.9
25 22.1 3.3 -11.7
400 344 1.7 -14.1
Freeze-thaw
(6 cycles)
1.5 1.53 4.3 2.1
25 25.6 1.0 2.2
400 404 1.2 1.0
Long term
(-80℃,
6 months)
1.5 1.52 4.4 1.4
25 25.8 3.0 3.1
400 397 1.6 -0.8
MII6 Short-term
(room
temperature
for 8h)
1.6 1.60 4.1 6.7
25 26.4 2.3 5.7
400 419 1.9 4.8
Auto-sampler
(10℃ for 72h)
1.5 1.34 5.7 -10.4
25 25.3 3.2 1.1
400 394 3.5 -1.4
Freeze-thaw
(6 cycles)
1.5 1.64 6.6 9.0
25 26.4 0.9 5.7
400 417 0.7 4.2
Long term
(-80℃,
6 months)
1.5 1.57 2.4 4.9
25 26.1 2.4 4.5
400 411 2.0 2.6
This article is protected by copyright. All rights reserved.
Table 5 Pharmacokinetic parameters M7 in patients with NSCLC after a single oral
dose of 500, 600 mg of Abivertinib (Mean±SD, n=18)
Dose
t1/2 Tmax Cmax AUClast
(h) (h) (ug/L) (h*ug/L)
500 mg 28.57±16.44 3.67±0.58 169.3±22.7 4331.0±1090.3
600 mg 14.07±3.95 4.33±2.38 168.6±80.2 2881.9±2119.2
Table 6 Pharmacokinetic parameters MII-6 in patients with NSCLC after a single oral
dose of 500, 600 mg of Abivertinib (Mean±SD, n=18)
Dose
t1/2 Tmax Cmax AUClast
(h) (h) (ug/L) (h*ug/L)
500 mg 30.24±11.83 6.33±1.53 192. 7±21.0 5781.2±1394.8
600 mg 24.29±14.58 6.27±4.98 98.9±67.5 2321.5±1580.2
This article is protected by copyright. All rights reserved.
Fig. 1. Product ion spectra: (A) IS-0010 (B) M7; (C) MII-6;
This article is protected by copyright. All rights reserved.
Fig. 2. Typical MRM chromatograms of plasma samples: (A) blank plasma; (B) plasma
spiked with the analytes at LLOQ level and IS; (C) plasma sample obtained 2 h after 50 mg Avitinib
oral administration of Abivertinib.
This article is protected by copyright. All rights reserved.
Fig. 3 Concentration-time profiles in plasma of patients with advanced NSCLC after oral
dose of 500 mg and 600 mg of Abivertinib. (A)M7 (mean±SD); (B) MII-6