Ruijuan Liuab,
Yao Wuab,
Minlu Chengab,
Pan Guab,
Lu Zhengc,
Yujie Liuc,
Pengcheng Ma*d and
Li Ding*ab
aDepartment of Pharmaceutical Analysis, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China. E-mail: dinglidl@hotmail.com; Fax: +86 025 83271485; Tel: +86 025 83271485
bNanjing Clinical Tech Laboratories Inc., Nanjing 211000, China
cYangtze River Pharmaceutical Group, Taizhou 225321, China
dInstitute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, 12 Jiangwangmiao Street, Nanjing 210042, China. E-mail: mpc815@163.com; Fax: +86 25 85471862; Tel: +86 25 85478929
First published on 13th November 2015
Shuanghua Baihe tablets (SBT) are a traditional Chinese medicine formula which is used for treatment of oral mucositis (OM) for more than 30 years in China. So far, there has been no report of the quantification of the major components in SBT and their pharmacokinetics in humans. In this study, high performance liquid chromatography coupled to triple-quadrupole mass spectrometry was used to determine the content of the major compounds in SBT and to evaluate the pharmacokinetics of nine major constituents (berberine, epiberberine, coptisine, palmatine, jatrorrhizine, magnoflorine, berberrubine, corynoline and acetylcorynoline) following single and multiple oral administrations of SBT in healthy subjects. This study was an open-label and 2-period clinical trial. It was conducted in 12 Chinese healthy subjects. Each subject received a single dose in period 1 and multiple doses in period 2. Blood samples were collected and determined over 96 h. Subjects underwent safety assessments and were monitored for adverse events. There was no serious adverse event, death or withdrawal. Gender had a significant effect on the pharmacokinetics of the active alkaloids except for magnoflorine, berberrubine, corynoline and acetylcorynoline. Compared with the single dosing, the exposure of the alkaloids increased significantly except for berberrubine after multiple dosing. Single and multiple oral doses of SBT were safe in healthy Chinese subjects. The accumulation of the alkaloids to different extents was observed with repeated dosing except for berberrubine. This work provides useful information for clinical application in the treatment of OM, and a scientific basis for the study on the material foundation of the medicinal effectiveness of SBT and its mechanism of action.
Traditional Chinese medicines (TCMs) have been used for thousands of years in Asia and attracted increasing research and application in Western countries.7,8 Most herbal medicines are prescribed in combination based on the theory of TCM to obtain synergistic effects or diminish the possible adverse reactions. These multiple herbs are called “TCM formula”.9 Shuanghua Baihe tablets (SBT) is a TCM formula that has been clinically employed for the prevention and treatment of OM for more than 30 years in China.10,11 Because of the long-term clinical practices and good therapeutic outcomes, it had been approved by the China Food and Drug Administration (CFDA) in 2012 (approval number Z20123033) for treating OM. SBT is composed of ten crude herbs, consisting of Coptidis Rhizoma, Corydalis Bungeanae Herba, Isatidis Radix, Arnebiae Radix, Lonicerae Japonicae Flos, Lophatheri Herba, Rehmanniae Radix, Lilii bulbus, Asari radix et rhizoma and Snake Bile.12 In this formula, Coptidis Rhizoma is the monarch drug. Corydalis Bungeanae Herba, Isatidis Radix and Arnebiae Radix are the minister drugs, and others are adjuvant and courier herbs. Coptidis Rhizoma and Corydalis Bungeanae Herba are well-known TCMs widely used in many prescriptions. They have pharmacological effects such as heat-clearing, damp-drying and detoxification.13,14 SBT have the functions of invigorating blood circulation, antibiotic, anti-inflammatory, anti-ulcer and regulation of immune function.15
Currently, the studies on the clinical efficacy of SBT for treating OM have been reported in China.10,15 Generally, the therapeutic and pharmacological effects of TCM are usually attributed to synergism among multiple herbs and constituents, termed ‘‘TCM formula compatibility’’.16 In order to indicate the active ingredients in SBT and its mechanism of action and promote the internationalization and modernization of SBT, well studies are needed to be done. In our previous study,12 the chemical compounds in SBT and constituents in rat plasma following oral administration of SBT were initially identified using liquid chromatography-high resolution mass spectrometry techniques. Besides, pharmacokinetic studies are useful to explain and predict a variety of events related to the efficacy and toxicity of drugs, thus it is valuable to perform pharmacokinetic studies for evaluating the rationality and compatibility of herbs or prescriptions.17–20 Until now, there has been no report about the pharmacokinetic study of SBT. As multiple compounds exist in herbal medicines, we should choose representative active compounds and be able to explain pharmacokinetic behaviors and drug–drug interactions in herbal medicines.21 According to our previous study,12 nine major bioactive alkaloids (Fig. 1; berberine, epiberberine, coptisine, palmatine, jatrorrhizine, magnoflorine and berberrubine derived from Coptidis Rhizoma; corynoline and acetylcorynoline derived from Corydalis Bungeanae Herba) were selected as indicative compounds for the pharmacokinetic study.
![]() | ||
Fig. 1 Chemical structures of the nine active alkaloids in the traditional Chinese medicine formula Shuanghua Baihe tablets. |
Additionally, the prevalence of use of TCMs is high and continues to increase in the world.22 Moreover, global interests in the safety of herbal medicines have grown.23 Since producers may omit safety information on product labels at their own discretion, labels themselves may lack clinically pertinent information.24 Hence, it is very important to be aware of the safety issues associated with the administration of SBT. Therefore, the objective of our study was to evaluate the pharmacokinetics and safety of the nine major bioactive ingredients following single and multiple oral administrations of SBT in healthy subjects.
The LC system was coupled to an Agilent 6410B triple quadrupole mass spectrometer (USA) equipped with an ESI source.† The mass spectrometer was operated in the positive ESI† mode with the drying gas temperature of 300 °C with N2 gas flow at 10 L min−1, nebulizer pressure of 40 psi, and capillary voltage of 4000 V. The specific other mass parameters for each analyte are displayed in Table 1. The signal acquisition and peak integration were performed using the MassHunter Qualitative Analysis Software (B.03.01) supplied by Agilent Technologies.
Analytes | Precursor ion (m/z) | Product ion (m/z) | Fragmentor voltage (V) | Collosion energy (eV) |
---|---|---|---|---|
Coptisine | 319.8 | 291.9 | 160 | 25 |
Berberrubine | 322.1 | 307.1 | 130 | 25 |
Berberine | 336.0 | 292.0 | 140 | 30 |
Epiberberine | 336.0 | 292.0 | 140 | 30 |
Jatrorrhizine | 338.0 | 323.0 | 145 | 20 |
Magnoflorine | 341.9 | 296.9 | 130 | 15 |
Palmatine | 352.0 | 336.0 | 140 | 25 |
Corynoline | 368.0 | 288.9 | 140 | 25 |
Acetylcorynoline | 410.0 | 288.9 | 130 | 25 |
Donepezil (IS) | 380.2 | 91.2 | 105 | 46 |
SBT (ten tablets) were pulverized in a mortar. Then, 0.1 g of SBT powder was accurately weighed into a volumetric flask and subjected to ultrasonic treatment at room temperature with 10 mL methanol/water (70:
30, v/v) for 30 min. The methanol extraction was centrifuged at 16
000 rpm for 10 min. The supernatant was collected and filtered through a 0.22 μm membrane. Then the solutions of SBT were diluted 200 times with methanol/water (30
:
70, v/v). An aliquot of 10 μL supernatant was then used for the LC-MS/MS analysis.
Subjects were screened 2 weeks before the initialization of this study. Eligibility criteria for male and female subjects included age 18–45 years, weight not less than 50 kg and a body mass index (BMI) in the normal range (19–24 kg m−2 inclusive). Subjects were in good health as evidenced by their medical history, physical examination, vital signs, 12-lead electrocardiogram (ECG) and laboratory profile. The following exclusion criteria were applied for subjects in this clinical trial: a history of clinically significant cardiovascular, renal, urinary tract, hepatic, pulmonary, gastrointestinal diseases; a history of known allergy or intolerance to any drugs; a history of tobacco, alcohol or drug abuse; those with abnormalities in clinical laboratory parameters; those who had received an investigational drug, or donation of blood in the preceding 3 months, or had received any drug within 4 weeks before the study start date, or was considered by the investigator, for any reason, to be an unsuitable candidate for receiving SBT. Females who were lactating or who had a positive pregnancy test were also ineligible. A total of 12 subjects, 6 male and 6 female, participated in this study.
In the first period, all the eligible subjects received a single dose (4 tablets) of SBT with 250 mL water at 7:00 am on day 1 after an overnight fast at least of 10 h. Blood samples were collected from each subject at pre-dose, 0.25, 0.5, 0.75, 1, 1.33, 1.67, 2, 2.5, 3, 4, 5, 6, 8, 12, 24, 48, 72 and 96 h after dosing. Water intake was allowed 2 h after administration of the drug, and standard meals were provided 4 h after administration of the drug.
After an 8 day wash out period, the same subjects participated in the multiple dose design. In this second period, all subjects received multiple doses (4 tablets at once, 3 times a day) of SBT at 7:00 am, 1:00 pm and 7:00 pm from day 9 to day 13 about half an hour before the meals and a single dose (4 tablets) at morning of day 14 under fasted condition. Blood samples used to confirm the steady-state were drawn before administration (pre-dose) on day 12 to day 14. Besides, blood sample collection on day 14 was the same as that on day 1. All doses were administered with 250 mL of water. Morning doses were administered after at least a 10 h overnight fasting.
In both periods of the study, approximately 5 mL of blood were drawn from a suitable forearm vein using an indwelling catheter into heparin containing tubes. These samples were centrifuged at 1700g for 10 min at 4 °C. The separated plasma was transferred to labelled tubes and stored at −80 °C until analysis.
Constituents | Calibration curve | Regression coefficient (r2) | Content (mg per tablet) |
---|---|---|---|
Coptisine | y = 0.001464x + 0.1946 | 0.9976 | 5.108 |
Berberrubine | y = 0.02828x + 1.255 | 0.9990 | 1.041 |
Berberine | y = 0.01127x + 6.624 | 0.9993 | 18.01 |
Epiberberine | y = 0.003179x + 0.1311 | 0.9990 | 5.378 |
Jatrorrhizine | y = 0.005598x + 0.6717 | 0.9978 | 2.423 |
Magnoflorine | y = 0.0005506x + 0.1376 | 0.9961 | 1.903 |
Palmatine | y = 0.005624x + 0.1811 | 0.9994 | 4.922 |
Corynoline | y = 0.005519x + 0.006205 | 0.9991 | 0.4646 |
Acetylcorynoline | y = 0.002698x − 0.001770 | 0.9991 | 0.06112 |
Subject | Gender | Age (years) | Height (cm) | Body weight (kg) | BMI (kg m−2) |
---|---|---|---|---|---|
a M = male; F = female; BMI = Body Mass Index; data are presented as mean with standard deviation (SD). | |||||
A | M | 20 | 186 | 82 | 23.70 |
B | M | 24 | 175 | 65 | 21.22 |
C | M | 21 | 170 | 60 | 20.76 |
D | M | 19 | 176 | 60 | 19.37 |
E | M | 20 | 160 | 60 | 23.44 |
F | M | 20 | 165 | 52 | 19.10 |
G | F | 21 | 163 | 55 | 20.70 |
H | F | 22 | 165 | 55 | 20.20 |
I | F | 22 | 163 | 54 | 20.32 |
J | F | 24 | 157 | 53 | 21.50 |
K | F | 25 | 160 | 52 | 20.31 |
L | F | 23 | 155 | 50 | 20.81 |
Mean | — | 22 | 166 | 58 | 20.95 |
SD | — | 2 | 9 | 9 | 1.39 |
Parameters/ingredients | Cmax (pg mL−1) | Tmax (h) | t1/2 (h) | MRT0–96 (h) | CL/F (×103 L h−1) | V/F (×103 L) | AUC0–96 (pg h mL−1) |
---|---|---|---|---|---|---|---|
a Data are presented as mean ± SD (standard deviation). The dose represents the content of each alkaloid in four tablets of Shanghua Baihe tablets. Abbreviations are as follows: Cmax, maximum plasma concentration; Tmax, time to reach Cmax; t1/2, terminal elimination half-life; MRT0–96, mean residence time from 0 to 96 h; CL/F, apparent clearance; V/F, apparent volume of distribution; AUC0–96, area under the plasma concentration–time curve from time 0 to 96 hours after administration. | |||||||
Berberine | 281.1 ± 88.6 | 1.6 ± 0.9 | 48.5 ± 35.7 | 22.3 ± 13.2 | 28.51 ± 26.88 | 1105 ± 687 | 3133 ± 1838 |
Coptisine | 296.2 ± 143.5 | 2.0 ± 0.6 | 46.3 ± 28.3 | 23.3 ± 11.5 | 7.554 ± 9.708 | 261.3 ± 128.9 | 4333 ± 3214 |
Epiberberine | 135.6 ± 37.2 | 1.5 ± 0.7 | 33.5 ± 22.0 | 12.3 ± 7.2 | 15.34 ± 17.42 | 444.3 ± 212.6 | 1266 ± 887 |
Jatrorrhizine | 25.74 ± 12.96 | 2.1 ± 1.1 | 31.6 ± 17.8 | 20.2 ± 8.5 | 26.64 ± 14.04 | 971.1 ± 421.3 | 355.2 ± 186.4 |
Magnoflorine | 2106 ± 654 | 2.7 ± 1.3 | 18.7 ± 13.4 | 14.1 ± 5.6 | 0.389 ± 0.156 | 9.425 ± 6.141 | 21![]() |
Berberrubine | 2961 ± 1171 | 1.1 ± 0.5 | 19.8 ± 13.1 | 8.7 ± 2.8 | 0.336 ± 0.111 | 9.190 ± 6.579 | 13![]() |
Palmatine | 53.37 ± 22.55 | 1.8 ± 0.9 | 37.5 ± 21.1 | 24.2 ± 7.8 | 33.01 ± 19.47 | 1428 ± 481 | 616 ± 253 |
Corynoline | 473.9 ± 442.1 | 1.5 ± 0.5 | 3.0 ± 2.3 | 3.2 ± 1.3 | 2.958 ± 2.570 | 7.680 ± 4.483 | 1625 ± 1887 |
Acetylcorynoline | 369.9 ± 146.6 | 1.4 ± 0.5 | 6.8 ± 5.5 | 3.8 ± 1.1 | 0.217 ± 0.072 | 1.814 ± 1.007 | 1086 ± 1887 |
Parameters/ingredients | Gender | Cmax (pg mL−1) | Tmax (h) | t1/2 (h) | MRT0–96 (h) | CL/F (×103 L h−1) | V/F (×103 L) | AUC0–96 (pg h mL−1) |
---|---|---|---|---|---|---|---|---|
a Data are presented as mean ± SD (standard deviation). Abbreviations are as follows: M, male; F, female; Cmax, maximum plasma concentration; Tmax, time to reach Cmax; t1/2, terminal elimination half-life; MRT0–96, mean residence time from 0 to 96 h; CL/F, apparent clearance; V/F, apparent volume of distribution; AUC0–96, area under the plasma concentration–time curve from time 0 to 96 hours after administration; asterisks (*) indicate significant difference (P < 0.05) between male and female subjects. | ||||||||
Berberine | M | 230.0 ± 94.4 | 1.2 ± 0.4 | 33.3 ± 37.8 | 14.6 ± 14.5* | 45.39 ± 29.91* | 1218 ± 928 | 1762 ± 1254* |
F | 332.3 ± 45.6 | 2.0 ± 1.1 | 63.6 ± 28.9 | 30.0 ± 5.4* | 11.63 ± 3.36* | 991.8 ± 381.8 | 4503 ± 1163* | |
Coptisine | M | 304.3 ± 185.8 | 1.7 ± 0.6 | 38.9 ± 32.6 | 15.3 ± 8.8* | 11.55 ± 12.70 | 295.3 ± 153.5 | 3027 ± 3232 |
F | 288.0 ± 103.0 | 2.2 ± 0.6 | 53.9 ± 23.6 | 31.4 ± 7.6* | 3.559 ± 2.764 | 227.3 ± 100.9 | 5639 ± 2860 | |
Epiberberine | M | 126.4 ± 49.1 | 1.4 ± 0.6 | 32.6 ± 30.4 | 7.2 ± 2.8* | 21.60 ± 23.50 | 483.6 ± 288.3 | 693.9 ± 282.0* |
F | 144.7 ± 20.9 | 1.7 ± 0.9 | 34.4 ± 11.7 | 17.3 ± 6.8* | 9.086 ± 4.565 | 404.9 ± 112.4 | 1839 ± 930* | |
Jatrorrhizine | M | 15.53 ± 4.84* | 1.9 ± 1.1 | 23.9 ± 14.1 | 16.7 ± 6.5 | 36.88 ± 12.49* | 1108 ± 494 | 219.3 ± 52.5* |
F | 35.94 ± 9.82* | 2.2 ± 1.2 | 39.4 ± 18.8 | 23.7 ± 9.3 | 16.41 ± 5.13* | 834.1 ± 319.2 | 491.1 ± 171.4* | |
Magnoflorine | M | 2232 ± 359 | 2.6 ± 1.1 | 15.3 ± 6.1 | 13.7 ± 5.7 | 0.368 ± 0.165 | 7.201 ± 2.567 | 22![]() |
F | 1981 ± 881 | 2.8 ± 1.6 | 22.1 ± 18.2 | 14.5 ± 6.0 | 0.410 ± 0.159 | 11.65 ± 8.032 | 19![]() |
|
Berberrubine | M | 2670 ± 851 | 0.8 ± 0.5 | 21.3 ± 18.1 | 9.4 ± 3.9 | 0.393 ± 0.124 | 10.93 ± 8.83 | 11![]() |
F | 3252 ± 1445 | 1.3 ± 0.4 | 18.3 ± 6.9 | 8.0 ± 0.9 | 0.279 ± 0.061 | 7.449 ± 3.166 | 15![]() |
|
Palmatine | M | 40.87 ± 21.88 | 1.4 ± 0.4 | 27.3 ± 10.7 | 18.7 ± 6.3* | 46.35 ± 19.20* | 1680 ± 507 | 406.7 ± 144.2* |
F | 65.88 ± 16.27 | 2.2 ± 1.2 | 47.7 ± 24.8 | 29.7 ± 4.8* | 19.67 ± 6.20* | 1176 ± 317 | 825.0 ± 124.5* | |
Corynoline | M | 373.7 ± 388.3 | 1.3 ± 0.5 | 1.8 ± 0.8 | 2.7 ± 0.7 | 3.766 ± 2.863 | 7.825 ± 5.545 | 1071 ± 1144 |
F | 574.1 ± 505.1 | 1.6 ± 0.4 | 4.2 ± 2.8 | 3.8 ± 1.5 | 2.150 ± 2.184 | 7.534 ± 3.662 | 2178 ± 2406 | |
Acetylcorynoline | M | 374.4 ± 186.1 | 1.4 ± 0.7 | 5.2 ± 2.9 | 3.4 ± 1.0 | 0.235 ± 0.089 | 1.541 ± 0.423 | 1052 ± 427 |
F | 365.4 ± 112.2 | 1.4 ± 0.2 | 8.4 ± 7.2 | 4.1 ± 1.2 | 0.198 ± 0.052 | 2.087 ± 1.368 | 1120 ± 250 |
Following single oral administration of SBT to human volunteers, most of the alkaloids were rapidly absorbed and exhibited good oral exposure. Peak plasma concentrations were observed within 1.1–2.7 h post-dose, and t1/2 ranged from 3.0 h (corynoline) to 48.5 h (berberine). The differences of pharmacokinetic parameters between male and female subjects in the first period of the study were estimated. After single dose administration, no significant differences (P > 0.05) were observed in Tmax, t1/2, MRT0–96, CL/F, V/F, lnAUC*0–96 (AUC had been body weight-normalized) and ln
C*max (Cmax had been body weight-normalized) of the four alkaloids (magnoflorine, berberrubine, corynoline and acetylcorynoline) for male and female subjects. However, the ln
AUC*0–96, MRT0–96 and CL/F of berberine for female subjects were 1–2 times, 2–3 times and 3–4 times higher than those of the male subjects, respectively; the MRT0–96 of coptisine for female subjects was about 2 times that of male subjects; the ln
AUC*0–96 and MRT0–96 of epiberberine for female subjects were both 2–3 times higher than those of male subjects, respectively; the ln
C*max and ln
AUC*0–96 of jatrorrhizine for female subjects were both 1–2 times more than those of the male subjects, respectively; the ln
AUC*0–96 and MRT0–96 of palmatine for female subjects were both 1–2 times more than those of male subjects, respectively.
Ingredients/parameters | Berberine | Coptisine | Epiberberine | Jatrorrhizine | Magnoflorine | Berberrubine | Palmatine | Corynoline | Acetylcorynoline |
---|---|---|---|---|---|---|---|---|---|
a Data are presented as mean ± SD (standard deviation). Abbreviations are as follows: Cmax, maximum plasma concentration; Cmin,ss, minimum plasma concentration at steady state; Cav, average value of the steady-state plasma concentration; Tmax, time to reach Cmax; t1/2, terminal elimination half-life; MRT0–96, mean residence time from 0 to 96 h; CL/F, apparent clearance; V/F, apparent volume of distribution; AUCss, AUC at steady state from time 0 to τ (τ was the dosing interval 6 h); AUC0–96, area under the plasma concentration–time curve from time 0 to 96 hours after administration; DF, the degree of fluctuation; RAUC, accumulation ratio calculated based on AUC; RCmax, accumulation ratio calculated based on Cmax; asterisks (*) indicate significant difference (P < 0.05) between multiple doses and single dose. | |||||||||
Cmax (pg mL−1) | 389.5 ± 125.1* | 483.1 ± 171.3* | 279.3 ± 109.3* | 56.23 ± 19.63* | 2902 ± 872* | 2620 ± 1129 | 110.3 ± 48.0* | 1818 ± 1146* | 659.9 ± 296.8* |
Css,min (pg mL−1) | 193.7 ± 71.7 | 201.7 ± 125.2 | 102.5 ± 39.4 | 27.77 ± 9.91 | 741.2 ± 347.3 | 330.7 ± 112.8 | 57.42 ± 20.29 | 423.0 ± 338.4 | 141.7 ± 39.0 |
Cav (pg mL−1) | 261.1 ± 77.7 | 280.4 ± 102.1 | 171.0 ± 56.4 | 38.09 ± 12.07 | 1978 ± 504 | 1251 ± 404 | 78.62 ± 24.85 | 944.9 ± 610.8 | 345.7 ± 128.2 |
Tmax (h) | 1.9 ± 0.8 | 1.6 ± 0.6 | 1.5 ± 0.5 | 2.0 ± 1.1 | 2.1 ± 1.1 | 1.0 ± 0.5 | 1.8 ± 0.9 | 1.7 ± 0.9 | 1.4 ± 0.6 |
t1/2 (h) | 56.3 ± 18.0 | 50.5 ± 22.9 | 49.0 ± 21.3 | 45.2 ± 16.3* | 15.6 ± 12.0 | 35.5 ± 26.9 | 53.3 ± 9.5 | 31.4 ± 9.7* | 56.7 ± 20.5* |
MRT0–96 (h) | 36.5 ± 3.4* | 35.0 ± 3.2* | 31.2 ± 7.3* | 33.6 ± 2.7* | 14.4 ± 5.1 | 14.0 ± 1.8* | 35.8 ± 3.0* | 22.3 ± 5.7* | 32.1 ± 4.0* |
CL/F (×103 L h−1) | 50.92 ± 18.74* | 14.08 ± 6.16* | 23.49 ± 8.87 | 47.20 ± 17.86* | 0.688 ± 0.203* | 0.614 ± 0.223* | 45.78 ± 15.30* | 0.480 ± 0.306* | 0.134 ± 0.048* |
V/F (×103 L) | 367.2 ± 176.8* | 102.5 ± 66.0* | 197.9 ± 92.2* | 327.7 ± 136.4* | 5.908 ± 6.238 | 12.64 ± 7.99 | 345.1 ± 137.9* | 6.718 ± 5.277 | 1.730 ± 0.486 |
AUCss (pg h mL−1) | 1567 ± 466 | 1683 ± 613 | 1026 ± 338.3 | 228.5 ± 72.4 | 11![]() |
7508 ± 2421 | 471.7 ± 149.1 | 5669 ± 3665 | 2074 ± 769 |
AUC0–96 (pg h mL−1) | 12![]() |
13![]() |
6458 ± 2808* | 1675 ± 680* | 32![]() |
15![]() |
3635 ± 1283* | 19![]() ![]() |
9191 ± 2950* |
DF | 0.750 ± 0.261 | 1.046 ± 0.568 | 1.017 ± 0.246 | 0.737 ± 0.243 | 1.089 ± 0.264 | 1.771 ± 0.393 | 0.666 ± 0.280 | 1.581 ± 0.679 | 1.442 ± 0.369 |
RAUC | 1.92 ± 0.35 | 1.72 ± 0.39 | 2.51 ± 0.79 | 2.87 ± 0.55 | 1.50 ± 0.37 | 0.89 ± 0.11 | 3.10 ± 0.81 | 6.26 ± 3.01 | 2.38 ± 0.48 |
RCmax | 1.45 ± 0.43 | 1.86 ± 1.06 | 2.10 ± 0.72 | 2.45 ± 0.83 | 1.45 ± 0.48 | 0.88 ± 0.20 | 2.38 ± 1.15 | 5.38 ± 3.16 | 1.81 ± 0.45 |
To evaluate whether steady-state had been reached, a statistical analysis of the Cmin values obtained at days 12, 13 and 14 was performed. The results showed that there were no significant differences in the Cmin values of the active alkaloids expect epiberberine, jatrorrhizine and palmatine at the administered doses (P > 0.05). Therefore, it could be proposed that steady-state had been reached for berberine, coptisine, magnoflorine, berberrubine, corynoline and acetylcorynoline after 5 days of administration of SBT (4 tablets at once, three times a day). Small fluctuations between maximum and minimum concentrations for all the nine ingredients were observed following repeat dosing of SBT with the DF ranging from 0.75 to 1.77. No significant differences (P > 0.05) were observed in Tmax of the nine ingredients after single- and multiple-dose administration of SBT. Median Tmax did not change after multiple oral administration of SBT for 5 days. Compared with the single dosing, the Cmax and AUC of the alkaloids increased significantly except berberrubine after 5 days of multiple dosing. The accumulation index calculated based on AUC (RAUC) and Cmax(RCmax) indicated that the exposure of eight ingredients (except berberrubine) increased in different extent with 1.5–6.3 times after repeated doses. Compared with the single dosing, the Cmax and AUCss of epiberberine, jatrorrhizine, palmatine, corynoline and acetylcorynoline were significantly increased by more than 2-fold after multiple dosing with SBT, especially corynoline, which increased by 5–6 times, but they could not be detected from human body essentially 96 h after the last administration.
The statistical results of other pharmacokinetic parameters (t1/2, MRT0–96, CL/F, V/F) of the nine alkaloids after single- and multiple-dose administration of SBT were as follows: except for jatrorrhizine, corynoline and acetylcorynoline, no significant differences (P > 0.05) were observed in t1/2 of nine alkaloids; there were significant differences (P < 0.05) in MRT0–96 of the nine alkaloids except magnoflorine; significant differences (P < 0.05) were observed in CL/F of nine alkaloids except epiberberine; no significant differences (P > 0.05) were observed in V/F of nine alkaloids except magnoflorine, berberrubine, corynoline and acetylcorynoline.
Pharmacokinetics is one of the key properties for the design of a safe and effective drug. Data and information obtained from the pharmacokinetic study could help clarify the complex interactions between clinically used medicines. The studies described in this report represent the initial first in man studies to examine the pharmacokinetic and safety profiles after single and multiple oral doses of SBT in healthy volunteers.
In the present study, the most significant differences of the main pharmacokinetic parameters (Cmax, AUCss and t1/2) of corynoline were observed between the single- and multiple-dose studies. The exposure of corynoline was increased significantly, and t1/2 was obviously prolonged without affecting Tmax noticeably by comparison to those of single dose. These results could be attributed to compatible effects of the ten herbs composed of SBT and auto-induction of metabolism. Distinct from chemical drugs, herbs medicines are multi-components and may exert a holistic treatment to diseases and show potential clinical benefits.26 The co-existence of multiple compounds may lead to metabolic and pharmacokinetic interactions.27,28 Those are the current hot topics of herb–drug and herb–herb interactions. Cytochrome P450 (CYP450) inhibition or induction is probably the most common mechanism for the pharmacokinetic interactions of herbs and drugs. Similarly, herbal constituents can be substrates, inhibitors, or inducers of CYP450 and have an impact on the pharmacokinetics of any co-administered drugs.27 One study demonstrated that corynoline could be metabolized by CYP2C9 and CYP3A4, and corynoline was also demonstrated to inhibit CYP2C9 (competitively) and CYP3A4 (noncompetitively).29 Besides, we identified that CYP2D6 also participated in the metabolism of corynoline (data not shown). Rhizoma Coptidis is the monarch (main) drug in SBT, and berberine is the highest and most effective constituent. Another study showed that repeated administration of berberine decreased CYP2D6, CYP2C9, and CYP3A4 activities, and herb–drug interactions should be considered when berberine is co-administered.30 Consequently, the Cmax and AUC of corynoline were increased, as less parent drug was metabolized by CYP3A4, CYP2D6 and CYP2C9. Half-life of corynoline was prolonged and the eliminated rate of corynoline was reduced because of the same reason mentioned above, which was consistent with the results of the study on corynoline and its potential interaction in SBT in rats.31
In addition, compared with the pharmacokinetic parameters of berberine reported in the literature32 after oral administration of single berberine in healthy volunteers, the Tmax of berberine was obviously shortened from 9.8 to 1.6 h after oral administration of the formula SBT, the Cmax and AUC (dose-normalized) of berberine were significantly increased by 2–3 times, and t1/2 was significantly prolonged from 28.6 to 48.5 h. The results indicated that other ingredients in the formula SBT could enhance the adsorption rate and bioavailability of berberine. The metabolism of berberine is mainly mediated by CYP1A2, 3A4, 2D6 enzymes.33 It implied that corynoline or other ingredients in the formula SBT might possess an inhibitory effect on the metabolism of berberine, which would be studied in our later research. Further studies on the herb–drug or herb–herb interactions are needed and more research on the clinical pharmacology mechanism of action should be conducted.
Several AEs occurred in two female subjects during the multiple doses study. One subject experienced two events with the symptoms of hiccups and heartburn, 20 minutes after taking the drug on the evening of day 13, which was considered unlikely to be related to the study drug by investigators according to the judgment rules of AEs. The other subject experienced two events with the symptoms of repeated abdominal pain and diarrhea after taking SBT on the evening of day 13 and morning of day 14, which was considered to be related to the study medication. An abnormality was found in the ECG of the latter subject when comparing baseline and end of study evaluations, which was considered to be possibly related to study drug. All the observed AEs were generally classified as mild events, and resolved without dose interruption, treatment or sequelae. No other individual clinically significant changes from baseline in laboratory evaluations, physical examinations, or vital signs were observed during the study.
It was observed that the incidence of AEs was higher in subjects receiving multiple doses than the single dose, and it was also higher in female subjects than male subjects. It was interesting that the phenomenon seems to be related to the results of the pharmacokinetic parameters of SBT, in which the exposure (AUC and Cmax) of some alkaloids increased significantly after multiple dosing and they were higher in female subjects. However, further research with a larger number of subjects is needed to see whether the exposure of the alkaloids has any link to the incidence of AEs. Besides, future investigations to evaluate the effect of long-term doses of SBT on the OM, abdominal pain, diarrhea and ECG are warned.
Footnote |
† Electronic supplementary information (ESI) available. See DOI: 10.1039/c5ra18665j |
This journal is © The Royal Society of Chemistry 2015 |