Accepted Manuscript Title: Plasma pharmacokinetics, tissue distribution and excretion studies of corydaline in rats using an ultra performance liquid chromatography–tandem mass spectrometry (UPLC–MS/MS) Authors: Jianfeng Wang Lishuang Liang Xingang Li Zhijian Fu PII: DOI: Reference:

S1570-0232(13)00556-4 http://dx.doi.org/doi:10.1016/j.jchromb.2013.10.018 CHROMB 18585

To appear in:

Journal of Chromatography B

Received date: Revised date: Accepted date:

10-7-2013 2-10-2013 11-10-2013

Please cite this article as: http://dx.doi.org/10.1016/j.jchromb.2013.10.018 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Plasma pharmacokinetics, tissue distribution and excretion studies of corydaline in rats using an ultra performance liquid chromatography–tandem mass spectrometry (UPLC–MS/MS)

Department of Pain managent, Qilu Hospital of Shandong University, 107 Wenhua West

b

us

Road, Jinan, 250012, China

cr

a

ip t

Jianfeng Wang a, 1, Lishuang Liang a, 1, Xingang Li b, *, Zhijian Fu c, **

Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua West Road,

c

an

Jinan, 250012, China

Department of Pain Management, Provincial Hospital Affiliated to Shandong University,

1

Authors contributed equally to this work

d

Corresponding author. Tel: +86 531 82166317; fax: +86 531 86927544; Email:

te

*

M

Jinan, Shandong 250021, China

[email protected]

Corresponding author. Tel: +86 531-82169114; fax: +86 531-82169022; Email:

Ac ce p

**

[email protected]

1  

Page 1 of 33

Abstract A rapid resolution ultra performance liquid chromatography (UPLC) coupled with electrospray ionization (ESI) mass spectrometry method was developed and validated for the

ip t

quantitative analysis of corydaline in rats' plasma and various tissues for pharmacokinetic,

cr

tissue distribution and excretion studies of corydaline. The analytes were separated on an

us

Acquity UPLC BEH C18 column (2.1 mm × 100 mm, 1.7 μm) and detected with a triple quadrupole mass spectrometer using positive ion ESI in the multiple reaction monitoring

an

(MRM) mode. The MS/MS ion transitions monitored were m/z 370.0→192.0 for corydaline and 354.1→188.0 for IS, respectively. Calibration curves (1/x2 weighted) offered satisfactory

M

linearity (r2 > 0.9984) within 1-1000 ng/mL. The accuracy and precision ranged from -7.4 to 8.5% and 3.4 to 12.8%, respectively. The absolute matrix effect (94.2–119.2%), relative

te

d

matrix effect (1.7–9.6%) and recoveries (81.4–93.7%) were satisfactory in all the biological matrices examined. The assay was successfully applied to the plasma pharmacokinetics,

Ac ce p

tissue distribution and excretion studies of corydaline in rats. The pharmacokinetic parameters such as half-life (t1/2), mean residence time (MRT) and maximum concentration (Cmax) were determined. These preclinical data of corydaline would be useful for the clinical reference.

Keywords: Corydaline; Pharmacokinetic; Tissue distribution; Excretion; Analysis; Determination

2  

Page 2 of 33

1. Introduction Corydaline, 2,3,9,10-tetramethoxy-13-methyl-5,8,13,13a-tetrahydro-6H-isoquino [3,2-a] isoquinoline, is an alkdoid which was discovered in 1826 by Wachroder during his investion

ip t

of Corydalis tuberosa, a herb which is native to Central Europe [1]. Corydaline has since

cr

been isolated from many species of Corydalis yields ranging from 0.01 to 2.0%. After

us

intensive studies by many workers, a correct structural formula was proposed and confirmed by total synthesis or biosynthesis of (±)-corydaline [1-4]. Corydaline was a good candidate

an

drug, showing various pharmacological activities such as acetylcholinesterase inhibitory [5-7], butyrylcholinesterase inhibitory [7], antiallergic [8], antinociceptive [9], multiple cytochrome

M

P450 and UDP-glucuronosyltransferase enzymes inhibition effect [10]. In addition, corydaline could promote gastric emptying and small intestinal transit and facilitates gastric

te

d

accommodation [11]. The absorption and transportation of corydaline in the human Caco-2 cell monolayer was mainly via passive diffusion mechanism [12]. In vivo, O-demethylation

Ac ce p

and hydroxylation were the major metabolic pathways for the metabolism of corydaline in human liver [13].

So far, the in vivo plasma pharmacokinetics, tissue distribution and excretion of

corydaline were still unknown. In the present study, an LC–ESI–MS/MS method was established for the determination of corydaline in rat biological samples and was successfully applied to the pharmacokinetic, tissue distribution and excretion studies of corydaline. To the best of our knowledge, it is the first time to report the pharmacokinetic, tissue distribution and excretion profiles of corydaline. The chemical structures of corydaline and the internal standard (IS) protopine are shown in Fig. 1. 3  

Page 3 of 33

ip t

Fig. 1

2. Material and methods

cr

2.1. Chemicals and reagents

us

Corydaline (purity > 98%) and protopine (IS, purity > 98%) was purchased from the National Institute for the Control of Pharmaceutical and Biological Products (Beijing, China).

an

HPLC grade methanol and formic acid were purchased from TEDIA Inc. (USA). Ultra pure ammonium acetate was purchased from Sangon Biotech Co., Ltd. (Shanghai, China).

M

Analytical grade ethyl acetate was purchased from Sinopharm Chemical Reagent Co., Ltd. (Shanghai, China). Solutol® HS 15 was purchased from Sigma-Aldrich (USA). Ultrapure

Ac ce p

2.2. Instrumentation

te

d

water (18.2 MΩ) was obtained from a Milli-Q Reference system (Millipore, USA).

A Waters AcquityTM TQD with an ultra performance liquid chromatography (Waters,

Milford, MA, USA) was used. Chromatographic separation was achieved on an Acquity UPLC BEH C18 column (2.1 mm × 100 mm, 1.7 μm; Waters, USA). Data acquisition and processing were performed using Masslynx 4.1 software and Quanlynx V4.1 (Micromass, Manchester, UK). All centrifugation were performed on an Eppendorf 5415R Refrigerated Microcentrifuge (Eppendorf, Germany).

2.3. Animals 4  

Page 4 of 33

Male Sprague–Dawley (SD) rats (200–220 g) were obtained from the Animal Center of Shandong Academy of Medical Sciences (Jinan, China). Animals were bred in a breeding room with temperature at 25 °C, humidity of 50 ± 10%, and a 12 h dark–light cycle. They

ip t

had free access to water and rodent chow all the time. All the experiment animals were

cr

housed under the above conditions for one week for acclimation, and were fasting overnight

us

before starting any treatment. The study was approved by the Animal Ethics Committee of

an

Shandong University.

2.4. Standard and sample preparation

M

2.4.1. Preparation of stock and working solutions

The stock solutions of corydaline (1 mg/mL) and the IS (1 mg/mL) were prepared with

te

d

methanol, respectively. The stock solution of corydaline was diluted with water to make a series of working solutions of 20 ng/mL, 100 ng/mL, 200 ng/mL, 1 μg/mL, 2 μg/mL, 10

Ac ce p

μg/mL, 20 μg/mL. The stock solution of IS was diluted with water to make a 1 μg/mL working solution. All stock and working solutions were kept at 4 °C away from light and brought to room temperature before use.

2.4.2. Preparation of quality control (QC) samples Series of standard working solutions (1, 5, 10, 50, 100, 500 and 1000 ng/mL) were prepared by spiking 95 μL blank biological matrix with 5 μL corydaline working solutions of different concentrations as mentioned above. No IS quality-control (QC) samples at three levels (low: 1.2 ng/mL; medium: 50 ng/mL; high: 800 ng/mL) were also independently 5  

Page 5 of 33

prepared in the same way. The calibration working solutions and QC samples were freshly

ip t

prepared before use.

2.5. Sample pretreatment

cr

In the present study, a conventional liquid–liquid extraction (LLE) method was applied to

us

extract corydaline and IS from biological samples (plasma, tissue homogenates, urine, feces or bile). Biological samples were taken out from -80 °C storage and thawed at room

an

temperature. Then 95 μL biological samples were extracted with 400 μL ethyl acetate after the addition of 5 μL water and 5 μL IS working solution (1 μg/mL). After vortex for 1 min

M

and centrifugation at 13,000 rpm for 10 min, 300 μL supernatant was removed and evaporated to dryness under vacuum at room temperature. The residue was reconstituted in

te

d

100 μL methanol-H2O (50:50, v/v), followed by vortexing for 1 min and centrifuged for 10

Ac ce p

min at 13,000 rpm. Finally, the supernatant was injected for LC–MS/MS analysis.

2.6. Chromatographic conditions

The mobile phase was delivered at a flow rate of 0.3 mL/min using a gradient elution

profile consisting of 5 mmol/L ammonium acetate with 0.2% formic acid (A) and methanol with 0.2% formic acid (B). The initial mobile phase composition was A-B (90:10, v/v) with a 1.0 min hold and followed by a linear increase to A-B (5:95, v/v) over a 1.5 min period, then held for 1 min, and finally returned to A-B (90:10, v/v) during 0.5 min and re–equilibrated for 1 min.

6  

Page 6 of 33

2.7. Mass spectrometer conditions Analytes were detected by MS/MS with an electrospray ionization (ESI) interface in positive multiple reaction monitoring (MRM) mode. Mass transitions of corydaline (m/z

ip t

370.0→192.0) and IS (m/z 354.1→188.0) were optimized. The ESI–MS/MS operating

cr

parameters used in this study were as follows: nitrogen was used as the desolvation and cone

us

gas with a flow rate of 500 and 50 L/h, respectively. The source and desolvation gas temperatures were set at 120 and 350 °C, respectively. The capillary voltage was set as 2.9

an

kV, and the cone voltage, extractor and radio frequency lens voltages were 30, 3 and 0.1 V, respectively. The collision energy was set as 30 and 35 eV for corydaline and IS,

M

respectively.

te

d

2.8. Method validation

A thorough and complete method validation for assaying corydaline and PTP in

Ac ce p

biological matrices was done following the US Food and Drug Administration (FDA) guidelines [14]. The validation parameters included selectivity, sensitivity, linearity, accuracy and precision, recovery, matrix effect and stability. 2.8.1. Selectivity

The selectivity of the method was determined by measuring the level of interfering

components in six individual sources of blank biological matrix. 2.8.2. Linearity and sensitivity The calibration curves were constructed by linear regression of the peak area ratio of corydaline to IS (Y–axis) and the nominal standard concentration (1, 5, 10, 50, 100, 500 and 7  

Page 7 of 33

1000 ng/mL) (X–axis) with a 1/x2 weighting factor, described as y = a + bx. Concentrations of QCs and samples were calculated using the regression equation of the calibration curve. The LLOQ was determined as the lowest concentration of the calibration curve (S/N >

ip t

10).

cr

2.8.3. Accuracy and precision

us

Accuracy and precision were determined from quintuplicate measurements on five different validation days of QC samples at three different concentrations (high, medium, low)

an

and from back calculated values of the calibration curve. The accuracy was expressed by the relative error (RE) and the precision by relative standard deviation (RSD). The acceptance

M

values used for validation of RSD and RE were within 15%, except LLOQ (within 20%). 2.8.4. Recovery

te

d

The extraction recovery of corydaline and IS from biological matrices was determined by comparing the response of corydaline and IS in biological matrices after extraction to the

Ac ce p

response of the same concentration of analyte spiked into the solution extracted from blank biological matrices. The recovery of corydaline was determined at three levels (high, medium, low), while IS at a single concentration of 50 ng/mL. 2.8.5. Matrix effect

The absolute matrix effect was evaluated by comparing the peak areas of samples spiked

post-extraction with corresponding peak areas of samples dissolved in mobile phase at high, medium and low level. The assessment of a relative matrix effect was made based on direct comparison of the responses (peak areas) of the analyte spiked into extracts originating from five different lots 8  

Page 8 of 33

(sources) of biofluids. The variability in these responses, determined as RSD (%), was considered as a measure of the relative matrix effect for a given analyte [15-16]. 2.8.6. Stability

ip t

The stability of analyte in rat biological samples was investigated by quintuplicate

cr

determinations of QC samples (high, medium, low levels) for each concentration. The

us

stability of corydaline was tested under the following conditions: (1) post–preparative stability during storage in the auto–sampler at 4 °C for 24 h; (2) short–term stability at room

an

temperature for 8 h; (3) freeze–thaw stability through three freeze–thaw cycles; (4) long–term

M

stability stored at −80 °C for 40 days.

2.9. Pharmacokinetics

te

d

Twelve rats were divided equally into two groups (six rats each group) at random for oral administration (i.g.) and tail intravenous injection (i.v.), respectively. Corydaline solution for

Ac ce p

administration was prepared in 0.9% sterile saline containing 3% ethanol (v/v) and 3% Solutol® HS 15 (v/v). A single dose (10 mg/kg for i.g. and 1.0 mg/kg for i.v.) of corydaline was orally administrated to rats after an overnight fasting. Plasma samples (about 500 μL per blood sample) were collected from the retro–orbital sinus prior to dosing and at 2 min, 5 min, 15 min, 30 min, 1 h, 2 h, 4 h, 6 h, 8 h, 10 h and 12 h. The blood samples were centrifuged for 15 min at 4,000 rpm at 4 °C. The supernatant was transferred into tubes.

2.10. Tissue distribution studies Three groups of male SD rats (n = 6 per group) were orally administrated at a single dose 9  

Page 9 of 33

of 10 mg/kg. After perfusion, tissues including heart, liver, brain, lung, kidney and spleen were removed at 10 min, 30 min and 3 h after dosing, followed by washing with normal

cr

normal saline solution (250 mg tissue / mL normal saline solution).

ip t

saline and drying with filter paper. Then the tissues were weighed and homogenized in

us

2.11. Excretion studies

Rats were randomly divided into two groups (n = 6). One group of six rats was orally

an

administrated at a single dose of 10 mg/kg corydaline. Afterward, the rats were individually placed in stainless-steel cages, which allowed the separate collection of urine and feces. The

M

urine and feces samples were collected at 0–2, 2–4, 4–6, 6–8, 8–10, 10–12 and 12–24 h post dosing. For the other group, rats were given a same oral dosage after the bile duct cannulation.

te

d

The bile samples were collected at 0–2, 2–4, 4–6, 6–8, 8–10, 10–12 and 12–24 h intervals. The blank bile was collected 2 h before drug administration. The volume of urine and bile

Ac ce p

samples and the weight of feces samples were recorded. The feces samples were homogenized with normal saline solution (250 mg feces /mL normal saline solution). All prepared plasma, tissue, urine, feces and bile samples were stored at −80 °C until

analysis.

2.12. Calculation All calculations were performed using Microsoft Excel 2007 (Microsoft Co., USA) or OriginPro 8.0 (OriginLab Co., USA) software. In order to acquire the pharmacokinetic parameters of corydaline and PTP, the concentration – time data were analyzed by DAS 2.0 10  

Page 10 of 33

software (SFDA). Data were expressed as mean ± SD.

ip t

3. Results and Discussion 3.1. Method validation

cr

3.1.1. Selectivity

us

The selectivity of the method was determined by extracting and analyzing pooled drug-free rat plasma. Representative ion chromatograms of drug-free and spiked rat plasma

an

containing 1 ng/mL of corydine and 50 ng/mL IS are shown in Fig. 2A and 2B. Representative ion chromatograms of plasma sample obtained 1.0 h after an i.g.

M

administration of 10 mg/kg corydaline are illustrated as Fig. 2C. The absence of peaks at the measured mass transitions and retention times of corydaline (m/z 370.0→192.0) and internal

te

d

standard (m/z 354.1→188.0) in the drug-free trace provide evidence of no endogenous

Ac ce p

interferences in drug-free plasma.

Fig. 2

3.1.2. Linearity and sensitivity

Linearity of the method was observed between corydaline concentrations of 1 and 1,000

ng/mL as detected from a 100 μL sample size of plasma. The resulting regression coefficients of the composite calibration curves were greater than 0.9984 on each day of validation (Table 1).

11  

Page 11 of 33

ip t

Table 1

3.1.3. Accuracy and precision

cr

Precision is represented as the relative standard deviation (%RSD) and accuracy was

us

calculated as the relative error (%RE) from the respective nominal concentration. The maximum acceptable limit for precision and accuracy was set at 15%. The intra-day accuracy

an

of the method ranged from -4.5% to 6.2%, while the range of intra-day precision was 3.4– 8.5% of the validation for rat plasma. The inter-day accuracy ranged from -7.4% to 8.5% and

M

inter-day precision ranged from 6.7% to 12.8% (Table 2).

te

d

Table 2

Ac ce p

3.1.4. Recovery and matrix effect

The average recoveries of corydaline from plasma, tissues, urine, feces and bile at the

three concentrations assayed was determined and shown in Table 3. The results demonstrated the efficiency of the sample preparation with little variation. The absolute matrix effects of plasma, tissues, urine, feces and bile on corydaline ranged

from 94.2% to 118.5%, while the relative matrix effects were over the range of 1.7-9.6% (Table 3), indicating that the extraction matrix had little coeluting endogenous substances that could influence the ionization of the analytes and IS.

12  

Page 12 of 33

Table 3

3.1.5. Stability

ip t

The results of stability investigation indicated that corydaline was stable in autosampler

cr

(24 h) at 4 °C, on bench–top (8 h) at room temperature, after repeated three freeze/thaw

an

routine pharmacokinetic and tissue distribution studies.

us

cycles and under the frozen condition at -80 °C for 40 days (Table 4), which could satisfy

M

Table 4

3.2. Plasma pharmacokinetics

te

d

The plasma concentration–time course of corydaline in rats given a single dose (10 mg/kg for i.g. and 1.0 mg/kg for i.v.) was illustrated in Fig. 3. The non-compartmental

Ac ce p

pharmacokinetic parameters, as showed in Table 5, were calculated from the plasma concentration versus time data using DAS 2.0 software. For i.g., during 0.5 h after dosing, plasma concentrations of corydaline increased very

quickly to Cmax (133.4 ± 19.84 ng/mL). Plasma concentrations decreased rapidly in the following 1.5 hours to 64.76 ± 11.43 ng/mL, and followed by the gradually decrease to LLOQ. The half-life (t1/2) was 3.009 ± 0.038 h. For i.v., the plasma concentration of corydaline first decreased sharply, and then equilibrium was reached at 2 h. The t1/2 was 1.327 ± 0.76 h, much shorter than that for i.g. The MRT0−t were 3.840 ± 0.382 h and 1.667 ± 0.182 h for i.g. and i.v., respectively, revealing that the residence time of corydine in vivo was 13  

Page 13 of 33

very short, both for i.g. and i.v. The absolute bioavailability (F) of corydaline was estimated as 10.0 ± 1.47% (F % = [AUC0–∞(i.g.) × Dose(i.v.)] / [AUC0–∞(i.v.) × Dose(i.g.)] × 100) [17-20], based on the calculation of AUC0–∞ obtained from i.g. and i.v. administration.

ip t

Pharmacokinetic studies have shown that corydaline could rapidly reach its Cmax and

cr

eliminate fast from the body, which implied that corydaline can show quick but short

us

pharmacological effects in vivo.

Tetrahydropulmatine was a very similar structural analogue of corydaline. The only

an

difference was the lack of methyl in the B ring of tetrahydropulmatine. The same with corydaline, tetrahydropulmatine was also a main active ingredient of the famous Traditional

M

Chinese medicine (TCM) Rhizoma Corydalis (yanhusuo). The pharmacokinetic studies of tetrahydropulmatine (administered in the form of pure compund or an ingredient in the TCM)

te

d

have been performed in rats [21-24], dogs [21, 25], rabbits [26] or human [27]. In rats, the range of Tmax, t1/2 and MRT0−t were 0.5-1.5 h, 4.2-6.68 h and 7.21-9.04 h, acquired by

Ac ce p

different groups. Compared with tetrahydropulmatine, the absorption of corydaline was similar (Tmax 0.5 h vs. 0.5-1.5 h), the elimination was slower (t1/2 3.009 h vs. 4.2-6.68 h), and the residence time was much longer (MRT0−∞ 3.840 h vs. 7.21-9.04 h) in rats. These results indicated that the methyl in the B ring play an important role in the pharmacokinetics of corydaline and tetrahydropulmatine. In addition, as the dosage was virous, the Cmax and AUC0–∞ were hardly to compare.

Fig. 3 Table 5 14  

Page 14 of 33

3.3. Tissue distribution Tissue distribution of corydaline was investigated in rats following a single i.g. dose of

ip t

corydaline (10 mg/kg). The results (Fig. 4) demonstrated that the corydaline underwent a

cr

rapid and wide distribution to tissues within the time course examined. The level of

us

corydaline in liver was significantly higher than other tissues, indicating its accumulation in liver. Furthermore, this phenomenon implied the extensive metabolism of corydaline in liver.

an

From Fig. 4, it can be observed that the level of corydaline in brain was close to that in heart, lung, kidney and spleen, showing that corydaline could effectively cross the blood–brain

M

barrier (BBB). This finding implied that corydaline may have central pharmacological

Ac ce p

Fig. 4

te

d

activity, though related reports had not been seen hitherto.

3.4. Excretion

The cumulative excretion of corydaline in urine, feces and bile after a single oral

administration (10 mg/kg) was investigated. Excretion data of corydaline in urine, feces and bile indicated that merely 18.5% (10.1%, 8.38% and 0.063%, respectively) of the dose administered was excreted as unconverted form, suggesting that corydaline was mainly excreted as the metabolites form. This finding was corresponding to the deduction of the tissue distribution results in section 3.3.

15  

Page 15 of 33

4. Conclusions For the first time, a method of analysis of corydaline in rat biological matrices using LC–MS/MS was developed and validated with respect to selectivity, accuracy, precision,

ip t

recovery, and successfully applied to evaluate the pharmacokinetics, tissue distribution of

cr

corydaline in rats. The Tmax 0.5 h of corydaline in plasma showed corydaline was rapidly

us

absorbed, while the t1/2 3.009 h of corydaline in plasma exhibited that corydaline could be fast eliminated from the body. Tissue distribution studies demonstrated that corydaline could

an

be rapidly and widely distributed into tissues and it can effectively cross the BBB in rats. The low absolute bioavailability (10.0 ± 1.47%) and accumulation in liver suggested an extensive

M

metabolism of corydaline in liver. Only 18.5% of the administered corydaline was excreted

Ac ce p

te

the metabolites form.

d

as unconverted form in urine, feces and bile, implying that corydaline was mainly excreted as

16  

Page 16 of 33

cr

Fig. 1. Chemical structures of corydaline and protopine (IS).

ip t

Figure captions:

us

Fig. 2. Representative MRM chromatograms of: (A) blank rat plasma; (B) rat plasma spiked with 1 ng/mL corydaline (LLOQ) and 50 ng/mL IS; (C) plasma sample from a rat obtained

an

1.0 h after an i.g. administration of 10 mg/kg corydaline.

M

Fig. 3. The plasma concentration – time course in rats given a single i.g. dose of 10 mg/kg (A)

te

d

or i.v. dose of 1.0 mg/kg (B) of corydaline (n = 6).

Fig. 4. Tissue distribution in rats after a single i.g. administration of 10 mg/kg of corydaline

Ac ce p

(n = 6).

17  

Page 17 of 33

References

M. Cushman, F.W. Dekow, Tetrahedron 34 (1978) 1435.

[2]

B. Ribar, D. Lazar, P. Radivojevic, P. Engel, O. Gasic, I. Kanyo, Acta Crystallogr.,

cr us

Sect. C: Cryst. Struct. Commun 48 (1992) 1864. [3]

ip t

[1]

H.L. Holland, M. Castillo, D.B. Maclean, I.D. Spenser, Can. J. Chem. 52 (1974)

an

2818.

C. Saa, E. Guitian, L. Castedo, R. Suau, J.M. Saa, J. Org. Chem. 51 (1986) 2781.

[5]

S. Berkov, J. Bastida, M. Nikolova, F. Viladomat, C. Codina, Phytochem. Anal. 19

M

[4]

(2008) 411.

H.T. Xiao, J. Peng, Y. Liang, J. Yang, X. Bai, X.Y. Hao, F.M. Yang, Q.Y. Sun, Nat.

te

d

[6]

Prod. Res. 25 (2011) 1418.

[8]

A. Adsersen, A. Kjolbye, O. Dall, A.K. Jager, J. Ethnopharmacol. 113 (2007) 179.

Ac ce p

[7]

S. Saito, M. Tanaka, K. Matsunaga, Y.S. Li, Y. Ohizumi, Biol. Pharm. Bull. 27 (2004) 1270.

[9]

[10]

C. Wang, S.W. Wang, G.R. Fan, H.F. Zou, Anal. Bioanal. Chem. 396 (2010) 1731.

H.Y. Ji, K.H. Liu, H. Lee, S.R. Im, H.J. Shim, M. Son, H.S. Lee, Molecules 16 (2011)

6591. [11]

T.H. Lee, M. Son, S.Y. Kim, Biol. Pharm. Bull. 33 (2010) 958.

[12]

Y. Liu, X. Yang, X. Yang, C. Yao, L. Ran, S. Wu, W. Xu, J. Liu, Zhongguo Zhong Yao Za Zhi 38 (2013) 112. 18

 

Page 18 of 33

[13]

H.Y. Ji, H. Lee, J.H. Kim, K.H. Kim, K.R. Lee, H.J. Shim, M. Son, H.S. Lee, J. Sep. Sci. 35 (2012) 1102.

[14]

USFDA. Guidance for Industry: Bioanalytical Method Validation 2001; Available

ip t

from:

cr

www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidanc

[15]

us

es/ucm070107.pdf.

A. Cappiello, G. Famiglini, P. Palma, E. Pierini, V. Termopoli, H. Trufelli, Anal.

[16]

an

Chem. 80 (2008) 9343.

B.K. Matuszewski, M.L. Constanzer, C.M. Chavez-Eng, Anal. Chem. 75 (2003)

[17]

M

3019.

R. Bhamidipati, S. Mujeeb, P.V. Dravid, A.A. Khan, S.K. Singh, Y.K. Rao, R.

[18]

te

d

Mullangi, N.R. Srinivas, Xenobiotica 35 (2005) 253. C. Guo, C. Yu, L. Li, Y. Wang, S. Wang, W. Wang, H. Hu, S. Xu, L. Yu, H. Jiang, S.

Ac ce p

Zeng, Xenobiotica 42 (2012) 466.

[19]

A. Treiber, O. Morand, M. Clozel, Xenobiotica 37 (2007) 298.

[20]

Y.T. Zheng, J.L. Xu, G. Ma, J.L. Zhang, Q. Zhu, H.R. Liu, P. Zhang, Y.Z. Zhu, W.M.

Cai, Xenobiotica 42 (2012) 304.

[21]

Z.Y. Hong, G.R. Fan, Y.F. Chai, X.P. Yin, Y.T. Wu, Chirality 17 (2005) 293.

[22]

Z.Y. Hong, G.R. Fan, J. Le, Y.F. Chai, X.P. Yin, Y.T. Wu, Biopharm. Drug Dispos. 27 (2006) 111.

[23]

H.D. Ma, Y.J. Wang, T. Guo, Z.G. He, X.Y. Chang, X.H. Pu, J. Pharm. Biomed. Anal. 49 (2009) 440. 19

 

Page 19 of 33

[24]

Z.Y. Hong, G.J. Cai, W.T. Ma, J. Wen, Y.F. Chai, G.R. Fan, Biomed. Chromatogr. 26 (2012) 749. Z.Y. Hong, G.R. Fan, Y.F. Chai, X.P. Yin, J. Wen, Y.T. Wu, J. Chromatogr. B 826 (2005) 108.

ip t

[25]

Z.C. Fan, C.J. Xie, Z.Q. Zhang, Chromatographia 64 (2006) 577.

[27]

C. Li, S. Zhang, H. Gao, X. Zhang, Eur. J. Drug Metabol. Pharmacokinet. 36 (2011)

us

cr

[26]

Ac ce p

te

d

M

an

257.

20  

Page 20 of 33

Table 1 Calibration curves, correlation coefficients and linear ranges of corydaline in different  matrices. 

Matrix 

Calibration curve 

Correlation coefficient (R2) 

Linear range (ng/mL) 

Plasma 

Y = 0.01718 × C − 0.008678 

0.9995 

Heart 

Y = 0.02246 × C + 0.001935 

0.9990 

Liver 

Y = 0.03447 × C – 0.01159 

0.9998 

ip t

 

Brain 

Y = 0.02486 × C – 0.005521 

0.9987 

Lung 

Y = 0.02075 × C + 0.003682 

0.9986 

1‐1000 

Kidney 

Y = 0.03608 × C – 0.01854 

0.9991 

1‐1000 

Spleen 

Y = 0.01823 × C + 0.004305 

0.9988 

1‐1000 

Urine 

Y = 0.02573 × C – 0.006279 

0.9984 

1‐1000 

Feces 

Y = 0.01654 × C – 0.008310 

0.9995 

1‐1000 

1‐1000 

1‐1000 

cr

us

an M

1‐1000 

1‐1000 

 

Ac ce p

te

d

                           

  21  

Page 21 of 33

Table 2 Intra– and inter–day accuracy and precision for the determination of corydaline in  rat plasma, liver, urine and feces (n = 5).   

Sample matrix 

Intra–day 

Inter–day 

concentration  RSD (%) 

RE (%) 

RSD (%) 

RE (%) 

8.4 

6.3 

1.2 

6.6 

3.5 

Plasma 

50 

3.4 

4.8 

 

800 

5.2 

‐2.7 

 

 

 

 

 

1.2 

4.6 

5.7 

Liver 

50 

7.8 

 

800 

6.5 

 

 

 

 

1.2 

5.6 

Urine 

50 

 

800 

 

 

8.5  3.4 

 

 

9.8 

‐1.8 

‐3.6 

12.6 

‐7.4 

5.1 

10.9 

8.2 

 

 

 

4.2 

8.4 

‐3.7 

‐4.5 

9.6 

2.4 

6.4 

‐3.9 

10.7 

5.6 

 

 

 

 

d

M

an

7.6 

te

6.1 

Ac ce p

6.7 

us

 

cr

(ng/mL) 

 

ip t

QC 

 

1.2 

8.5 

‐3.0 

12.8 

‐5.6 

Feces 

50 

7.8 

5.3 

12.0 

‐1.2 

 

800 

8.4 

6.2 

11.3 

6.8 

   

22  

Page 22 of 33

Table 3 Matrix effect and extraction recovery for the assay of corydaline and IS in rat plasma,  liver, urine and feces (n = 5).    QC 

Absolute 

Relative 

Extraction 

concentration 

matrix effect 

matrix effect 

recovery 

(ng/mL) 

(Mean ± SD, %) 

(RSD, %) 

Analyte  matrix  Corydaline 

1.2 

118.5 ± 9.7 

6.3 

 

 

50 

97.6 ± 11.5 

4.0 

 

 

800 

103.2 ± 8.3 

 

IS 

50 

104.1 ± 6.7 

 

 

 

 

Liver 

Corydaline 

1.2 

 

 

50 

 

 

800 

 

IS 

50 

 

 

 

Urine 

Corydaline 

 

 

(Mean ± SD, %)  88.3 ± 8.5 

us

cr

Plasma 

ip t

Sample 

92.6 ± 6.7 

84.5 ± 2.1 

1.7 

83.7 ± 11.0 

 

 

114.1 ± 12.6 

5.6 

83.4 ± 7.2 

106.4 ± 11.4 

3.5 

85.8 ± 4.4 

95.7 ± 8.6 

2.9 

90.5 ± 5.9 

94.6 ± 3.5 

4.0 

85.7 ± 9.2 

 

 

 

1.2 

119.2 ± 9.5 

5.4 

93.7 ± 6.4 

50 

113.0 ± 12.3 

6.6 

91.6 ± 7.8 

Ac ce p

te

d

M

an

4.5 

 

 

800 

102.4 ± 7.1 

4.1 

86.8 ± 7.0 

 

IS 

50 

98.6 ± 4.4 

5.2 

87.5 ± 8.2 

 

 

 

 

 

 

Feces 

Corydaline 

1.2 

94.2 ± 11.0 

7.8 

86.0 ± 9.3 

 

 

50 

117.4 ± 13.4 

7.4 

81.4 ± 8.5 

 

 

800 

104.0 ± 9.1 

5.2 

84.5 ± 5.7 

 

IS 

50 

104.6 ± 5.5 

9.6 

82.6 ± 13.0 

   

23  

Page 23 of 33

Table 4 Stability of corydaline in rat plasma, liver, urine, feces and bile (n = 5).   

QC 

Freeze and thaw 

Short‐term 

Long‐term 

Post‐preparative 

concentration 

stability 

stability 

stability 

stability 

(ng/mL) 

(Mean ± SD, %) 

(Mean ± SD, %) 

(Mean ± SD, %) 

(Mean ± SD, %) 

Plasma 

1.2 

98.2 ± 2.8 

94.2 ± 4.5 

97.7 ± 8.3 

93.1 ± 6.0 

 

50 

95.9 ± 5.3 

96.8 ± 2.3 

95.8 ± 7.4 

 

800 

108.3 ± 3.0 

96.1 ± 2.8 

94.5 ± 6.0 

 

 

 

 

Liver 

1.2 

94.5 ± 6.4 

105.6 ± 4.6 

 

50 

107.9 ± 2.1 

101.4 ± 2.6 

 

800 

105.4 ± 4.6 

 

 

 

Urine 

1.2 

97.1 ± 3.2 

 

50 

95.5 ± 4.4 

 

800 

106.2 ± 6.7 

 

 

Feces 

1.2 

Sample 

us

cr

96.5 ± 1.8 

94.9 ± 4.7   

94.6 ± 5.5 

93.7 ± 7.6 

92.4 ± 8.3 

97.2 ± 8.8 

96.8 ± 5.2 

104.0 ± 7.1 

94.3 ± 6.7 

 

 

 

101.3 ± 3.7 

87.4 ± 6.7 

106.7 ± 3.5 

104.8 ± 7.6 

93.4 ± 4.7 

92.6 ± 2.7 

96.5 ± 4.4 

91.5 ± 8.6 

105.4 ± 4.8 

 

 

 

 

94.2 ± 4.8 

96.2 ± 5.9 

88.3 ± 6.2 

97.4 ± 5.2 

te

d

M

an

 

Ac ce p

 

ip t

matrix 

 

50 

98.5 ± 5.1 

102.4 ± 4.3 

92.6 ± 3.6 

102.5 ± 6.4 

 

800 

103.7 ± 7.2 

95.5 ± 5.6 

90.2 ± 5.4 

95.8 ± 3.3 

   

24  

Page 24 of 33

  Table 5 Pharmacokinetic parameters of corydaline in rats following i.g. (10 mg/kg) or i.v. (1.0  mg/kg) administration (n = 6). 

i.v. 

Parameters 

Mean ± SD 

Parameters 

Cmax (μg/L) 

133.4 ± 19.84 

Cmax (μg/L) 

Tmax (h) 

0.5 ± 0 

Tmax (h) 

t1/2z (h) 

3.009 ± 0.038 

t1/2z (h) 

1.327 ± 0.76 

AUC0–t (μg/L×h) 

550.9 ± 78.30 

AUC0–t (μg/L×h) 

559.7 ± 59.95 

AUC0–∞ (μg/L×h) 

592.9 ± 86.97 

AUC0–∞ (μg/L×h) 

590.7 ± 72.15 

CLz/F (L/h/kg) 

17.11 ± 2.473 

CLz (L/h/kg) 

1.711 ± 0.217 

Vz/F (L/Kg) 

74.30 ± 11.02 

Vz (L/Kg) 

7.150 ± 1.713 

MRT0−t (h) 

3.840 ± 0.382 

MRT0−t (h) 

1.667 ± 0.182 

MRT0−∞ (h) 

4.718 ± 0.433 

MRT0−∞ (h) 

2.334 ± 0.288 

Zeta 

0.23 ± 0.003 

Zeta 

0.254 ± 0.1 

 

te

i.g. 

ip t

 

 

 

 

10.0 ± 1.47 

 

 

1123 ± 130.4 

cr

us

an

M

d

Ac ce p

Bioavailability (%) 

Mean ± SD 

0.033 ± 0 

                 

25  

Page 25 of 33

Highlights 

Ac ce p

te

d

M

an

us

cr

ip t

The PK, tissue distribution and excretion of corydaline was investigated.  Corydaline was quickly absorbed and eliminated.  Rapid and wide distribution into tissues and BBB penetration ability of corydaline  Few of corydaline were excreted unchangingly.   

26  

Page 26 of 33

Ac

ce

pt

ed

M

an

us

cr

i

Figure 1

Page 27 of 33

Ac

ce

pt

ed

M

an

us

cr

i

Figure 2A

Page 28 of 33

Ac

ce

pt

ed

M

an

us

cr

i

Figure 2B

Page 29 of 33

Ac

ce

pt

ed

M

an

us

cr

i

Figure 2C

Page 30 of 33

Ac

ce

pt

ed

M

an

us

cr

i

Figure 3A

Page 31 of 33

Ac

ce

pt

ed

M

an

us

cr

i

Figure 3B

Page 32 of 33

Ac

ce

pt

ed

M

an

us

cr

i

Figure 4

Page 33 of 33

Preclinical pharmacokinetics, tissue distribution and excretion studies of a potential analgesics - corydaline using an ultra performance liquid chromatography-tandem mass spectrometry.

A rapid resolution ultra performance liquid chromatography (UPLC) coupled with electrospray ionization (ESI) mass spectrometry method was developed an...
467KB Sizes 0 Downloads 0 Views