Journal of Pharmaceutical and Biomedical Analysis 90 (2014) 134–138

Contents lists available at ScienceDirect

Journal of Pharmaceutical and Biomedical Analysis journal homepage: www.elsevier.com/locate/jpba

Short communication

Determination of CUDC-101 in rat plasma by liquid chromatography mass spectrometry and its application to a pharmacokinetic study Qingwei Zhang a , Congcong Wen b , Zheng Xiang c , Jianshe Ma b , Xianqin Wang c,∗ a b c

Shanghai Institute of Pharmaceutical Industry, Shanghai 200437, China Function Experiment Teaching Center, Wenzhou Medical University, Wenzhou 325035, China Analytical and Testing Center of Wenzhou Medical University, Wenzhou 325035, China

a r t i c l e

i n f o

Article history: Received 5 October 2013 Received in revised form 25 November 2013 Accepted 29 November 2013 Available online 4 December 2013 Keywords: CUDC-101 LC–MS Pharmacokinetics Rat plasma HDAC

a b s t r a c t CUDC-101 is a multi-targeted, small-molecule inhibitor of histone deacetylase (HDAC), epidermal growth factor receptor tyrosine kinase (EGFR/ErbB1), and human epidermal growth factor receptor 2 tyrosine kinase (HER2/neu or ErbB2) with potential antineoplastic activity. A sensitive and selective liquid chromatography mass spectrometry method for determination of CUDC-101 in rat plasma was developed. After addition of carbamazepine as internal standard (IS), protein precipitation by acetonitrile–methanol (9:1, v/v) was used as sample preparation. Chromatographic separation was achieved on a Zorbax SB-C18 (2.1 mm × 150 mm, 5 ␮m) column with acetonitrile–0.1% formic acid in water as mobile phase with gradient elution. An electrospray ionization source was applied and operated in positive ion mode; selective ion monitoring (SIM) mode was used for quantification using target fragment ions m/z 435 for CUDC-101 and m/z 237 for the IS. Calibration plots were linear over the range of 5–2000 ng/mL for CUDC-101 in rat plasma. Mean recoveries of CUDC-101 in rat plasma were in the range of 84.0–90.5%. RSD of intra-day and inter-day precision were both 98%) was a gift from Shanghai Institute of Pharmaceutical Industry (Shanghai, China). Carbamazepine (IS, purity >98%) was purchased from the National Institute for Control of Pharmaceutical and Biological Products (Beijing, China). LC-grade acetonitrile and methanol were purchased from Merck Company (Darmstadt, Germany). Ultra-pure water was prepared by Millipore Milli-Q purification system (Bedford, MA, USA). The rat blank plasma samples were supplied from drug-free rats. 2.2. Instrumentation and conditions All analysis was performed with a 1200 Series liquid chromatograph (Agilent Technologies, Waldbronn, Germany) and a Bruker Esquire HCT ion-trap mass spectrometer (Bruker Technologies, Bremen, Germany) equipped with an electrospray ion source and controlled by ChemStation software (Version B.01.03 [204], Agilent Technologies, Waldbronn, Germany). Chromatographic separation was achieved on an Agilent Zorbax SB-C18 (2.1 mm × 150 mm, 5 ␮m) column at 40 ◦ C, with acetonitrile–0.1% formic acid as mobile phase. The flow rate was 0.4 mL/min. A gradient elution program was conducted for chromatographic separation with mobile phase A (0.1% formic acid), and mobile phase B (acetonitrile) as follows: 0–4.0 min (10–80% B), 4.0–7.0 min (80–80% B), 7.0–8.0 min (80–10% B), 8.0–12.0 min (10–10% B). Drying gas flow and nebulizer pressure was set at 7 L/min and 25 psi. Dry gas temperature and capillary voltage of the system were adjusted at 350 ◦ C and 3500 V. LC–MS was performed with SIM mode using target ions at m/z 435 for CUDC-101 (Fig. 1a) and m/z 237 for carbamazepine (IS, Fig. 1b), in positive ion electrospray ionization interface, respectively. 2.3. Calibration standards and quality control samples The stock solutions of CUDC-101 (100 ␮g/mL) was prepared in methanol–water (50:50) and carbamazepine (IS) (100 ␮g/mL) was prepared in methanol–water (50:50). Working solutions for calibration and controls were prepared from the stock solution by dilution using methanol. The 2.0 ␮g/mL working standard solution of IS was prepared from the IS stock solution by dilution using methanol. All of the solutions were stored at 4 ◦ C and were brought to room temperature before use. CUDC-101 calibration standards were prepared by spiking blank rat plasma with appropriate amounts of the working solutions. Calibration plots were constructed in the range of 5–2000 ng/mL for CUDC-101 in rat plasma (5, 10, 20, 50, 100, 200, 500, 1000 and 2000 ng/mL). Quality-control (QC) samples were prepared by the same way as the calibration standards, three different plasma concentrations (12, 800 and 1600 ng/mL). The analytical standards and QC samples were stored at −20 ◦ C. 2.4. Sample preparation Before analysis, the plasma sample was thawed to room temperature. In a 1.5 mL centrifuge tube, an aliquot of 10 ␮L of the IS working solution (2.0 ␮g/mL) was added to 100 ␮L of collected plasma sample followed by the addition of 200 ␮L acetonitrile–methanol (9:1, v/v). The tubes were vortex mixed for

Fig. 1. Mass spectrum of CUDC-101 (a) and carbamazepine (IS, b).

1.0 min. After centrifugation at 14 900 g for 10 min, the supernatant (2 ␮L) was injected into the LC–ESI–MS system for analysis. 2.5. Method validation The method was validated for selectivity, linearity, accuracy, precision, recovery and stability according to the guidelines set by the United States Food and Drug Administration (FDA) and European Medicines Agency (EMA) for validation of bioanalytical method [17,18]. Validation runs were conducted on three consecutive days. Each validation run consisted of one set of calibration standards and six replicates of QC plasma samples. The selectivity of the method was evaluated by analyzing blank rat plasma, blank plasma spiked CUDC-101 and IS and a rat plasma sample. Calibration curves were constructed by analyzing spiked calibration samples on three separate days. Peak area ratios of CUDC-101 to IS were plotted against analyte concentrations, and standard curves were well fitted to the equations by linear regression with a weighting factor of the reciprocal of the concentration (1/x) in the concentration range of 5–2000 ng/mL. The LLOQ was defined as the lowest concentration on the calibration curves. To evaluate the matrix effect, blank rat plasma were extracted and then spiked with the analyte at 12, 800 and 1600 ng/mL. The corresponding peak areas were then compared to those of neat standard solutions at equivalent concentrations, and this peak area ratio is defined as the matrix effect (ME). The matrix effect of IS was evaluated at the concentration (200 ng/mL) in the same manner. Accuracy and precision were assessed by the determination of QC samples at three concentration levels in six replicates (12, 800 and 1600 ng/mL) in three validation days. The precision was expressed by RSD.

136

Q. Zhang et al. / Journal of Pharmaceutical and Biomedical Analysis 90 (2014) 134–138

The recovery of CUDC-101 was evaluated by comparing peak area of extracted QC samples with those of reference QC solutions reconstituted in blank plasma extracts (n = 6). The recovery of the IS was determined in a similar way. Carry-over was assessed following injection of a blank plasma sample immediately after 3 repeats of the upper limit of quantification (ULOQ) and the response was checked [19]. The stabilities of CUDC-101 in rat plasma were evaluated by analyzing three replicates of plasma samples at the concentrations of 12 and 1600 ng/mL, which were exposed to different conditions. These results were compared with those freshly prepared plasma samples. The short-term stability was determined after the exposure of the spiked samples at room temperature for 2 h, and the ready-to-inject samples (after protein precipitation) in the HPLC autosampler at room temperature for 24 h. The freeze/thaw stability was evaluated after three complete freeze/thaw cycles (−20 to 25 ◦ C) on consecutive days. The long-term stability was assessed after storage of the standard spiked plasma samples at −20 ◦ C for 10 days. The stability of the IS (200 ng/mL) was evaluated in a similar way. 2.6. Pharmacokinetic study Male Sprague-Dawley rats (200–220 g) were obtained from Laboratory Animal Center of Wenzhou Medical University (Wenzhou, China) used to study the pharmacokinetics of CUDC-101. All six rats were housed at Laboratory Animal Center of Wenzhou Medical University. All experimental procedures and protocols were reviewed and approved by the Animal Care and Use Committee of Wenzhou Medical University and were in accordance with the Guide for the Care and Use of Laboratory Animals. Diet was prohibited for 12 h before the experiment but water was freely available. Blood samples (0.3 mL) were collected from the tail vein into heparinized 1.5 mL polythene tubes at 0.0833, 0.25, 0.5, 0.75, 1, 1.5, 2, 3, 4, 6, 8, 12 h after intravenous administration of CUDC-101 (5 mg/kg, 30% captisol solution). The samples were immediately centrifuged at 3000 g for 10 min. The plasma obtained (100 ␮L) was stored at −20 ◦ C until analysis. Plasma CUDC-101 concentration versus time data for each rat was analyzed by DAS (Drug and statistics) software (Version 2.0, Wenzhou Medical University, China). 3. Results and discussion 3.1. Method development Electrospray ionization (ESI+) source exhibited more sensitivity and better reproducibility for CUDC-101 compared with an atmospheric pressure chemical ionization (APCI+) interface, and ESI+ was used in this work. It was found that the sensitivity of SIM mode was much higher than the multiple reaction monitoring (MRM). Therefore, we chose the SIM mode for quantitative determination in this study. The liquid chromatographic conditions were developed to separate as many interfering compounds as possible from the analyte and IS. Different columns, such as Zorbax SB-C18 (50 mm × 2.1 mm, 3.5 ␮m), Zorbax SB-C18 (100 mm × 2.1 mm, 3.5 ␮m) and Zorbax SB-C18 (150 mm × 2.1 mm, 5 ␮m) were compared for chromatographic separation. The Zorbax SB-C18 (150 mm × 2.1 mm, 5 ␮m) column demonstrated proper retention time and better peak shape for CUDC-101 and IS than other columns. The mobile phase played a critical role in achieving good chromatographic behavior and appropriate ionization. Various combinations of methanol, acetonitrile, water and 0.1% formic acid in with water changed content of each component were investigated and compared to identify the optimal mobile phase. Acetonitrile was chosen as the organic phase

Fig. 2. Representative LC–MS chromatograms of CUDC-101 (1) and carbamazepine (IS, 2), (a) blank plasma; (b) blank plasma spiked with CUDC-101 (5 ng/mL) and IS (200 ng/mL); (c) a rat plasma sample 2 h after intravenous administration of single dosage 5 mg/kg CUDC-101 (68 ng/mL).

because it could provide sharper peak shape and lower pump pressure compared with methanol. Formic acid (0.1%) added into the water could improve the sensitivity, therefore consist of acetonitrile and 0.1% formic acid in water was chosen as mobile phase. An efficient clean-up for bio-samples to remove protein and potential interferences prior to LC–MS analysis was an important point in the method development. The effective and simple protein precipitation was employed in our work. Acetonitrile–methanol (9:1, v/v) was chosen as the protein precipitation solvent because it exhibited better effect than acetonitrile, methanol, perchloric acid (6%) and trichloroacetic acid (10%). 3.2. Selectivity and matrix effect Fig. 2 shows the typical chromatograms of a blank plasma sample, a blank plasma sample spiked with CUDC-101 and IS, and a

Q. Zhang et al. / Journal of Pharmaceutical and Biomedical Analysis 90 (2014) 134–138

137

Table 1 Precision, accuracy and recovery for CUDC-101 of QC sample in rat plasma (n = 6). Concentration (ng/mL)

RSD (%)

5 12 800 1600

Accuracy (%)

Recovery (%)

Intra-day

Inter-day

Intra-day

Inter-day

10.3 7.4 4.2 2.3

13.6 6.7 4.1 4.4

94.1 96.2 106.8 98.7

91.2 103.6 95.8 96.4

84.0 87.5 90.5 89.3

± ± ± ±

5.5 6.5 3.8 3.2

plasma sample. No interfering endogenous substance was observed at the retention time of the analyte and IS. The ME for CUDC-101 at concentrations of 12, 800 and 1600 ng/mL were measured to be 97.6 ± 8.6%, 105.6 ± 6.3% and 102.3 ± 4.8% (n = 6), respectively. The ME for IS (200 ng/mL) was 104.5 ± 5.4% (n = 6). As a result, ME from plasma was negligible in this method. 3.3. Calibration curve and sensitivity The linear regressions of the peak area ratios versus concentrations were fitted over the concentration range 5–2000 ng/mL for CUDC-101 in rat plasma. Typical equation of the calibration curve was: y = (0.00315 ± 0.00027)x − (0.00897 ± 0.03510), r = 0.9991, where y represents the ratios of CUDC-101 peak area to that of IS and x represents the plasma concentration. The LLOQ for the determination of CUDC-101 in plasma was 5 ng/mL. The precision and accuracy at LLOQ were 10.3% and 94.1%, respectively. The LOD, defined as a signal/noise ratio of 3, was 2 ng/mL for CUDC-101 in plasma. 3.4. Precision, accuracy and recovery The precision of the method was determined by calculating RSD for QCs at three concentration levels over three validation days. Intra-day precision was 11% or less and the inter-day precision was 14% or less at each QC level. The accuracy of the method ranged from 91.2% to 106.8% at each QC level. Mean recoveries of CUDC101 were better than 84.0%. The recovery of the IS (200 ng/mL) was 90.3 ± 3.2%. Assay performance data were presented in Table 1. 3.5. Carry-over None of the analytes showed any significant peak (≥20% of the LLOQ and 5% of the IS) in blank samples injected after the ULOQ samples. Adding 4 extra minutes to the end of the gradient elution effectively washed the system between samples thereby eliminating carry-over [19]. Table 2 Summary of stability of CUDC-101 and IS under various storage conditions (n = 3). Condition

Concentration (ng/mL)

RSD (%)

Fig. 3. Mean plasma concentration time profile after intravenous administration of single dosage 5 mg/kg CUDC-101 in six rats. Table 3 The main pharmacokinetic parameters after intravenous administration of single dosage 5 mg/kg CUDC-101 in six rats. Parameters

Unit

Mean (±SD)

AUC(0–t) AUC(0–∞) MRT(0–t) MRT(0–∞) t1/2 CL V Cmax

ng/mL h ng/mL h h h h L/h/kg L/kg ng/mL

581 585 1.4 1.5 2.0 9.2 25.9 1085

± ± ± ± ± ± ± ±

166 168 0.1 0.2 0.5 2.8 8.2 171

3.6. Stability The auto-sampler, room temperature, freeze–thaw and longterm (10 days) stability results indicated that the analyte was stable under the storage conditions described above since the bias in concentrations were within ±15% of their nominal values, and the established method was suitable for the pharmacokinetic study (Table 2).

Accuracy (%)

3.7. Application

Added

Found

Ambient, 2 h

12 1600 (IS) 200

12.4 ± 0.8 1636.8 ± 37.6 192.6 ± 8.3

6.3 2.3 4.3

103.1 102.3 96.3

−20 ◦ C, 10 days

12 1600 (IS) 200

11.8 ± 1.1 1524.8 ± 115.9 203.8 ± 12.8

9.5 7.6 6.3

98.6 95.3 101.9

3 freeze thaw

12 1600 (IS) 200

12.5 ± 1.1 1561.6 ± 114.0 206.6 ± 14.3

8.6 7.3 6.9

104.4 97.6 103.3

4. Conclusion

12 1600 (IS) 200

11.6 ± 0.5 1619.2 ± 69.6 195.2 ± 10.3

4.5 4.3 5.3

96.7 101.2 97.6

The LC–MS method described here allowed accurate and reproducible method for determination of CUDC-101 in rat plasma utilizing 100 ␮L of plasma with an LLOQ of 5.0 ng/mL. A time-saving

Autosampler ambient 24 h

The method was applied to a pharmacokinetic study in rats. The mean plasma concentration–time curve after intravenous administration of a single 5 mg/kg dose of CUDC-101 was shown in Fig. 3. The main pharmacokinetic parameters from non-compartment model analysis were summarized in Table 3.

138

Q. Zhang et al. / Journal of Pharmaceutical and Biomedical Analysis 90 (2014) 134–138

protein precipitation procedure made the method readily applicable in a further clinical study. The LC–MS method successfully applied to a pharmacokinetic study of CUDC-101 after intravenous administration of single dosage 5 mg/kg in rats. Acknowledgment This work was supported by fund of the National Natural Science Foundation of China, No. 81102297. References [1] A. Gschwind, O.M. Fischer, A. Ullrich, The discovery of receptor tyrosine kinases: targets for cancer therapy, Nat. Rev. Cancer 4 (2004) 361–370. [2] A.M. Xu, P.H. Huang, Receptor tyrosine kinase coactivation networks in cancer, Cancer Res. 70 (2010) 3857–3860. [3] B.A. Baldo, N.H. Pham, Drugs used for chemotherapy, in: Drug Allergy, Springer, 2013, pp. 399–418. [4] P. Ulivi, W. Zoli, L. Capelli, E. Chiadini, D. Calistri, D. Amadori, Target therapy in NSCLC patients: relevant clinical agents and tumour molecular characterisation, Mol. Clin. Oncol. 1 (2013) 575–581 (Review). [5] Q. Xie, G.F. Vande Woude, M.E. Berens, RTK inhibition: looking for the right pathways toward a miracle, Future Oncol. 8 (2012) 1397–1400. [6] A. Tartarone, C. Lazzari, R. Lerose, V. Conteduca, G. Improta, A. Zupa, A. Bulotta, M. Aieta, V. Gregorc, Mechanisms of resistance to EGFR tyrosine kinase inhibitors gefitinib/erlotinib and to ALK inhibitor crizotinib, Lung Cancer 81 (2013) 328–336. [7] C.-M. Choi, J.C. Lee, Mechanisms of acquired resistance to epidermal growth factor receptor inhibitors and overcoming strategies in lung cancer, J. Lung Cancer 11 (2012) 59–65. [8] M. Garofalo, G. Romano, G. Di Leva, G. Nuovo, Y.-J. Jeon, A. Ngankeu, J. Sun, F. Lovat, H. Alder, G. Condorelli, EGFR and MET receptor tyrosine kinase-altered microRNA expression induces tumorigenesis and gefitinib resistance in lung cancers, Nat. Med. 18 (2011) 74–82.

[9] K. Tanaka, I. Babic, D. Nathanson, D. Akhavan, D. Guo, B. Gini, J. Dang, S. Zhu, H. Yang, J. De Jesus, Oncogenic EGFR signaling activates an mTORC2–NF-␬B pathway that promotes chemotherapy resistance, Cancer Discov. 1 (2011) 524–538. [10] T. Nakagawa, S. Takeuchi, T. Yamada, H. Ebi, T. Sano, S. Nanjo, D. Ishikawa, M. Sato, Y. Hasegawa, Y. Sekido, EGFR-TKI resistance due to BIM polymorphism can be circumvented in combination with HDAC inhibition, Cancer Res. 73 (2013) 2428–2434. [11] L. Bianchi, F. Bruzzese, A. Leone, A. Gagliardi, M. Puglia, E. Di Gennaro, M. Rocco, A. Gimigliano, B. Pucci, A. Armini, Proteomic analysis identifies differentially expressed proteins after HDAC vorinostat and EGFR inhibitor gefitinib treatments in Hep-2 cancer cells, Proteomics 11 (2011) 3725–3742. [12] H. Song, C.-W. Li, A.M. Labaff, S.-O. Lim, L.-Y. Li, S.-F. Kan, Y. Chen, K. Zhang, J. Lang, X. Xie, Acetylation of EGF receptor contributes to tumor cell resistance to histone deacetylase inhibitors, Biochem. Biophys. Res. Commun. 404 (2011) 68–73. [13] X. Cai, C. Qian, Discovery of HDAC-inhibiting multi-target inhibitors Designing Multi-target Drugs, vol. 21, 2012, pp. 221. [14] X. Cai, H.-X. Zhai, J. Wang, J. Forrester, H. Qu, L. Yin, C.-J. Lai, R. Bao, C. Qian, Discovery of 7-(4-(3-ethynylphenylamino)-7-methoxyquinazolin-6yloxy)-N-hydroxyheptanamide (CUDC-101) as a potent multi-acting HDAC, EGFR, and HER2 inhibitor for the treatment of cancer, J. Med. Chem. 53 (2010) 2000–2009. [15] J. Wang, N.W. Pursell, M.E. Samson, R. Atoyan, A.W. Ma, A. Selmi, W. Xu, X. Cai, M. Voi, P. Savagner, C.J. Lai, Potential advantages of CUDC-101, a multitargeted HDAC, EGFR, and HER2 inhibitor, in treating drug resistance and preventing cancer cell migration and invasion, Mol. Cancer Ther. 12 (2013) 925–936. [16] C.-J. Lai, R. Bao, X. Tao, J. Wang, R. Atoyan, H. Qu, D.-G. Wang, L. Yin, M. Samson, J. Forrester, CUDC-101, a multitargeted inhibitor of histone deacetylase, epidermal growth factor receptor, and human epidermal growth factor receptor 2, exerts potent anticancer activity, Cancer Res. 70 (2010) 3647–3656. [17] US Food and Drug Administration, Guidance for Industry: Bioanalytical Method Validation, 2001. [18] European Medicines Agency (EMA), Guideline on Bioanalytical Method Validation, 2011. [19] J.S. Williams, S.H. Donahue, H. Gao, C.L. Brummel, Universal LC–MS method for minimized carryover in a discovery bioanalytical setting, Bioanalysis 4 (2012) 1025–1037.

Determination of CUDC-101 in rat plasma by liquid chromatography mass spectrometry and its application to a pharmacokinetic study.

CUDC-101 is a multi-targeted, small-molecule inhibitor of histone deacetylase (HDAC), epidermal growth factor receptor tyrosine kinase (EGFR/ErbB1), a...
750KB Sizes 0 Downloads 0 Views

Recommend Documents