Home | Register | Login | Inquiries | Alerts | Sitemap |  


Advanced Search
JKM > Volume 40(4); 2019 > Article
Hu, Jang, Kang, Ku, Choi, and Lee: Effects of mixed formulation of tamoxifen and blue honeysuckle on the pharmacokinetics profiles of tamoxifen after single oral administration

Abstract

Objectives

Here, we investigated the effects of concentrated and lyophilized powders Blue honeysuckle (BH) on the PK of tamoxifen, to establish the pharmacokinetics (PK) profiles as one of essential process in new drug development.

Methods

After single oral treatment of 0.4 mg/ml of tamoxifen or tamoxifen 0.4 with BH 40, 20 and 10 mg/ml, the plasma were collected at 0.5 hr before administration, 0.5, 1, 2, 3, 4, 6, 8 and 24 hr after end of single or mixed formula treatment. Plasma concentrations of tamoxifen were analyzed using LC-MS/MS methods. Tmax, Cmax, AUC, t1/2 and MRTinf were analyzed using noncompartmental PK data analyzer programs.

Results

Tamoxifen and BH 40 mg/ml did not induce any significant change on the plasma tamoxifen concentrations, while significant decreases were observed in tamoxifen and BH 10 mg/ml from 2 to 8 hr as compared with tamoxifen only, respectively. Furthermore, significant increases of Tmax in tamoxifen and BH 40 mg/ml, significant decreases of Cmax in tamoxifen and BH 20 mg/ml, significant decreases of AUC0-t, AUC0-inf and MRTinf in tamoxifen and BH 10 mg/ml were demonstrated as compared with tamoxifen only.

Conclusion

Taken together, tamoxifen and BH 10 mg/ml induced significant decrease of the oral bioavailability of tamoxifen, while tamoxifen and BH 40 or 20 mg/ml did not critically influenced, suggesting formulated BH concentration-independencies. It, therefore, seems to be needed that pharmacokinetic study after repeated administration should be tested to conclude the effects of BH on the pharmacokinetics of tamoxifen.

Introduction

Pharmacokintics (PK) is a branch of pharmacology dedicated to the determination of the fate of substances administered externally to a living organism. PK includes the study of the mechanisms of absorption and distribution of an administered drug, the rate at which a drug action begins and the duration of the effect, the chemical changes of the substance in the body1,2). PK of drugs in animals, especially rodents have been provided a valuable data for absorption, distribution, metabolism and elimination of drugs in vivo, directly related with their pharmacodynamics. Therefore, the PK profiles have been observed as one of essential process in new drug development including natural products14). In addition, they also provided constantly reliable data on the possible potential drug-drug interactions, when drugs were co-administered24).
Tamoxifen is a nonsteroidal estrogen agonist -antagonist antineoplastic agent has been used for breast cancer5). It is the usual endocrine (anti-estrogen) therapy for hormone receptor-positive breast cancer in pre-menopausal women, and is also a standard in post-menopausal women although aromatase inhibitors are also frequently used in that setting6,7). Tamoxifen causes cells to remain in the G0 and G1 phases of the cell cycle. Because it prevents (pre)cancerous cells from dividing but does not cause cell death, tamoxifen is cytostatic rather than cytocidal8,9). However, various side effects related to tamoxifen treatment also have been arise as bone loss in premenopausal women who continue to menstruate after adjuvant chemotherapy10), endometrial changes, including cancer, are among tamoxifen’s side effects11), increased risk of thromboembolism12), cause of fatty liver13), reduced cognition14), semantic memory scores15) and libido16,17), and premature growth plate fusion18). Tamoxifen also depress the immune response19,20), and it also known that hypersensitivity to tamoxifen or any ingredient in the formulation21,22).
Tamoxifen is contraindicated, when used in women with ductal carcinoma in situ and women at high risk for breast cancer, concurrent anticoagulant therapy with a warfarin derivative23), and be used with caution in patients with leukopenia or thrombocytopenia24) and pregnant25,26). Hot flashes, vaginal discharge, menstrual irregularities and weight loss are common side effects related with tamoxifen treatment25,27).
As results of combination therapies with other drugs to improve the side effects of tamoxifen or to achieve synergic effects, various drug-drug interactions of tamoxifen have been evaluated; Because tamoxifen was metabolized by a substrate of CYP3A, 2C9, 2D628), it interacted with various drugs, namely, combinations containing any of the following medications, depending on the amount present, may also interact with aminoglutethimide - decreased plasma tamoxifen and N-desmethyltamoxifen concentrations29), anticoagulants - enhanced warfarin effects30,31), bromocriptine - increased plasma tamoxifen and N-desmethyltamoxifen concentrations32), letrozole - decreased plasma letrozole concentrations33), medroxyprogesterone - decreased plasma N-desmethyltamoxifen concentrations but did not reduce plasma tamoxifen concentrations34), phenobarbital – decreased plasma tamoxifen concentrations35), rifampin - decreased plasma tamoxifen and N-desmethyltamoxifen concentrations36), and cyclosporine, erythromycin, diltiazem, erythromycin and nifedipine - competitively inhibited formation of N-desmethyltamoxifen in vitro3739), respectively. However, interactions with herbal products have not been established except for some restricted natural compounds30,31).
Until now, we have observed the possible interactions with Korean traditional polyherbal formulas; oral co-administration of Jaeumkanghwa-tang, a traditional yin-tonifying herbal medicine used for various oriental obstetrical and gynecological fields within 5 min did not critically influenced on the pharmacokinetics profiles of tamoxifen after single40) and repeated41) co-administration at dosage levels of 50 mg/kg in tamoxifen and 100 mg/kg in Jaeumkanghwa-tang, respectively. It was also demonstrated that single co-administration of Gamiondam-tang within 5 min and with 2.5 hr-intervals critically influenced on the oral bioavailability of tamoxifen through variable influences on the absorption and excretion of tamoxifen at dosage levels of GMODT 100 mg/kg and tamoxifen 50 mg/kg, can be influenced on the toxicity or pharmacodynamic of tamoxifen in our previous studies42,43).
Blue honeysuckle (Berries of Lonicera caerulea L., Caprifoliaceae) is a traditional shrub used in folk medicine in northern Russia, China, and Japan, but its fruits are little known as edible berries in North America and Europe, and also in Korea44). The berries are a rich source of ascorbic acid and phenolic components, particularly anthocyanins, flavonoids and low molecular weight phenolic acids44). These compounds have been reported to have multiple biological activities including strong antioxidant activity44). Recently, orally administered blue honeysuckle was reported to protect mice against ionizing radiation45), ameliorates abnormal lipid and glucose metabolism in rats46), hepatoprotective effects47), anti-inflammatory effects48). Especially blue honeysuckle extracts have been showed the strongest antioxidant potent among 12 types of colored berries49), and phenolic rich extract of BH has been shown to possess anti-inflammatory and wound-healing effects in vitro and in vivo48) and skin protective effects to ultraviolet-induced damages50) with less toxicity51).
In the present study, the effects of BH on the pharmacokinetics of tamoxifen were examined in rats after mixed formulation as tamoxifen 0.4 and BH 40, 20 and 10 mg/ml concentrations. After single oral treatment of 4 mg/kg of tamoxifen single formula or tamoxifen 0.4 with BH 40, 20 and 10 mg/ml concentration mixed formulas, the plasma were collected at 30 min before administration, 30 min, 1, 2, 3, 4, 6, 8 and 24 hr after end of single or mixed formula treatment. Plasma concentrations of tamoxifen were analyzed using LC-MS/MS methods. PK parameters of tamoxifen (Tmax, Cmax, AUC, t1/2 and MRTinf) were analysis as compared with tamoxifen single formula administered rats.

Materials & methods

1. Animals and husbandry

A total of fifty-three male SPF/VAF Outbred Crl:CD [SD] rats (OrientBio, Seungnam, Korea) were used after acclimatization for 15 days. Animals were allocated five per polycarbonate cage in a temperature (20–25°C) and humidity (40–45%) controlled room. Light: dark cycle was 12hr: 12hr and feed (Samyang, Korea) and water were supplied free to access. After 15 days of acclimatization, four groups of 5 rats each were selected based on the body weights, and used further experiments, in the present study. All animals were marked by picric acid, and overnight fasted (about 18 hr; water was not restricted) before treatment, and further fasted during 3 hr after end of treatment. Animal experiments were conducted according to the national regulations of the usage and welfare of laboratory animals, and approved by the Institutional Animal Care and Use Committee in Daegu Haany University (Gyeongsan, Gyeongbuk, Korea) [Approval Approval No DHU2014-081].

2. Test articles

Deep purple colored solution of concentrated blue honeysuckle, about 63 brix, were supplied by H&K Bioscience Co., Ltd. (Seoul, Korea) and used as same as our previous single oral dose toxicity test51) in this experiment. The brief process for making BH was as follows. 200 g of 63 brix concentrated blue honeysuckle solutions supplied by H&K Bioscience were diluted into 25 brix using distilled water, and then completely lyophilized by programmable freeze dryer (Operon FDB-5503, Kimpo, Korea). Total 124.40 g (yield = 62.2%) of BH were acquired. At proximate analysis of BH by Association of Official Analytical Chemists (AOAC)52) methods, BHcL contains energy 380 kcal/100 ml, carbohydrate 93 g/100 ml, sugar 41 g/100 ml, protein 2 g/100 ml, sodium 20 mg/100 ml, but it did not contains total lipids (0 mg/100 ml), saturated lipids (0 mg/100 ml), trans-fat (0 mg/100 ml), cholesterol (0 mg/100 ml), respectively. In addition, phytochemical analysis of BH reveals that it contains 4.54 ± 0.09% of betaine by high performance liquid chromatography (HPLC), 210.63 ± 23.65 mg gallic acid equivalents (GAE)/g of total phenols by Folin-Ciocalteu colorimetric method53), 159.30 ± 12.51 mg catechin equivalents (CE)/g of total flavonoids by a modified colorimetric method54) and 133.57 ± 4.06 mg malvidin-3-O-glucoside equivalents (M3GE)/g of total antocyanins by a modified pH differential method55), respectively. In addition, white crystalline powders of tamoxifen (Kunshan SanYou Pharmaceutical Material Co., Ltd., Suzhou, China) was used as control drug as listed follows. Tamoxifen and BH were stored in a refrigerator at 4°C and −20 °C to protect from light and degeneration until use. Both drugs are well dissolved (up to 40 mg/ml solutions in BH and upto 0.4 mg/ml solutions in tamoxifen) in distilled water as vehicle, respectively.

3. Groupings, test article formulation and administration

Four groups of 5 rats each were used in this study as follows. The dosages of tamoxifen 4 mg/kg were selected, considering the clinical dosage in human and body surface of rats. Single formula of tamoxifen was prepared as dissolved in distilled water (0.4 mg/ml concentration), and each mixed formula consisted of tamoxifen and BH was prepared by dissolved of approximate amounts of tamoxifen (0.4 mg/ml) and BH (40, 20 or 10 mg/ml) in distilled water. Tamoxifen single formula and all three types of mixed formula consisted of tamoxifen and BH were once orally administered, in a volume of 10 ml/kg, respectively.

4. Plasma collections

All rats were anesthetized with 2 to 3% isoflurane (Hana Pharm. Co., Hwasung, Korea) in the mixture of 70% N2O and 28.5% O2, and blood samples (0.5 ml) were collected into 50 IU heparinized 0.7 ml Eppendorf tubes through the orbital plexus at 30 min before treatment (as a control), 30 min, 1, 2, 3, 4, 6, 8 and 24 hr after end of single oral administration of tamoxifen single or mixed formula with three different dosages of BH, respectively. Blood samples were immediately centrifuged for 10 min at 13,000 rpm and about 0.3 ml aliquots of plasma were stored in a −150 °C deep freezer until analysis of tamoxifen.

5. Sample preparation and calibrations

Primary stock solution, 1.0 mg/ml of tamoxifen in 50% acetonitrile (Sigma-Aldrich, St Louise, MO, USA) mixtures with distilled water and internal standard working solution, carbamazepine (Sigma-Aldrich, St Louise, MO, USA) 500 ng/ml in acetonitrile were prepared. Working standard solutions were prepared by dilution with acetonitrile. All standard solutions were stored at −20°C in the dark when not in use, and calibrated the standard samples as 100 μl of blank plasma; working standard solutions and internal standard working solution were mixed with 100 μl of acetonitrile. In addition, 100 μl of sample plasma and internal standard working solution were mixed with 200 μl of acetonitrile. The mixtures were mixed by vortex-mixing and centrifuged at 12,000 rpm for 10 min at 4°C. Clear supernatants (150 μ l) were directly transferred to injection vials and the aliquot (5.0 μl) was injected into the LC-MS/MS system.

6. LC-MS/MS conditions

Concentrations of tamoxifen in the rat plasma samples were determined LC-MS/MS method. Chromatographic analysis was performed using an Agilent 1100 Series HPLC (Agilent Technologies, Santa Clara, CA, USA) equipped with on-line degasser, binary pump, autosampler and column compartment. Separation of the analyte from potentially interfering material was achieved at ambient temperature using Waters SymmetryTM C18 (2.1×50mm, 3.5μm) (Waters Corp., Milford, MA, USA) at column oven 30°C. The mobile phase used for the chromatographic separation was composed of 50% distilled water (0.1% formic acid) / 50% acetonitrile and was delivered isocratically at a flow rate of 0.35 ml/min. The column effluent was monitored using an API 2000 triple-quadruple mass-spectometric detector (Applied Biosystems, Foster City, CA, USA). The instrument was equipped with an electrospray interface in positive ion mode, and controlled by the Analyst version 1.4.1 software (Applied Biosystems, Foster City, CA, USA). Samples were introduced to the interface through a Turbo Ion Spray with the temperature set at 500°C. A high positive voltage of 4.0 kV was applied to the ion spray. Nitrogen was used as the nebulizer gas, curtain gas, and collision gas with the settings of 70, 20, and 7, respectively. The multiple reaction monitoring (MRM) detection method was employed for the detection of tamoxifen; the transitions monitored were carbamazepine (IS): m/z 237>194 (Retention time: 0.74 min), tamoxifen: 372>178 (Retention time: 0.55 min). Calibration curves of tamoxifen were linear over the ranges studied with r2>0.994. The lower limit of quantification of the tamoxifen in the rat plasma was 0.5 ng/ml.

7. Pharmacokinetic analysis

The plasma concentration data were analyzed using a noncompartmental method on commercial pharmacokinetics data analyzer programs (PK solutions2.0; Summit, Montrose, CO, USA)56). The elimination rate constant (Kel) was calculated by the log-linear regression of tamoxifen concentration data during the elimination phase, and the terminal half-life (t1/2) was calculated by 0.693/Kel. The peak concentration (Cmax) and time to reach the peak concentration (Tmax) of tamoxifen in the plasma were obtained by visual inspection of the data in the concentration-time curve. The area under the plasma concentration-time curve (AUC0-t) from time zero to the time of the last measured concentration (Clast) was calculated using the linear trapezoidal rule57). The AUC zero to infinity (AUC0-inf) was obtained by adding AUC0-t and the extrapolated area was determined by Clast/Kel. The mean residence time infinity (MRTinf) was calculated by dividing the first moment of AUC (AUMC0-inf) by AUC0-inf.

8. Statistical analyses

All the means are presented with their standard deviation (SD) of five rats (Mean±SD of five rat plasma tamoxifen concentrations). The pharmacokinetic parameters were compared using a non-parametric comparison test, Mann-Whitney U (MW) test, on the SPSS for Windows (Release 14.0K, SPSS Inc., Chicago, IL, USA). A p-value <0.05 was considered statistically significant. In addition, the percent-point changes between tamoxifen single formula treated rats and tamoxifen with BH 40, 20 or 10 mg/ml mixed formula administered rats were calculated to help the understanding of the effects after mix formulations, according to our previous study58): Percentage-point changes as compared with tamoxifen 0.4 mg/ml single formula treated mice (%) = [((Data of mixed formula administrated rats – data of tamoxifen single formula treated rats)/Data of tamoxifen single formula treated rats) × 100].

Results

1. Changes on the plasma concentrations of tamoxifen

Tamoxifen was detected from 30 min to 24 hr after end of administration in tamoxifen single formula and all three different concentrations of mixed formulas with BH administered rats, respectively. Although single oral administration of mixed formulas consisted of tamoxifen and BH 40 mg/ml did not induced any significant changed on the plasma tamoxifen concentrations, but significant (p<0.01 or p<0.05) decreases of plasma tamoxifen concentrations were demonstrated in tamoxifen and BH 20 mg/ml mixed formula treated rats at 1, 2 and 6 hr after administration, and also in BH 10 mg/ml mixed formula treated rats from 2 to 8 hr after end of treatment as compared with tamoxifen 0.4 mg/ml single formula treated rats, in the current result (Fig 1).

2. Changes on the Tmax of tamoxifen

The Tmax of tamoxifen were significantly (p<0.05) increased as 172.73% points in tamoxifen and BH 40 mg/ml mixed formula treated rats (3.00±1.00 hr) as compared with tamoxifen single formula treated rats (1.10±0.55 hr). The Tmax of tamoxifen were non-significantly changed as 27.27 and 9.09% points in tamoxifen and BH 20 or 10 mg/ml mixed formula treated rats as compared with tamoxifen single formula treated rats; they were detected as 1.40±0.89 and 1.20±0.45 hr in tamoxifen and BH 20 or 10 mg/ml mixed formula treated rats, respectively (Table 1).

3.3. Changes on the Cmax of tamoxifen

The Cmax of tamoxifen were significantly (p<0.05) decreased as −32.90% points in tamoxifen and BH 20 mg/ml mixed formula treated rats (21.46±4.01 ng/ml) as compared with tamoxifen single formula treated rats (31.98±8.98 ng/ml). The Cmax of tamoxifen were non-significantly changed as −15.13 and −33.77% points in tamoxifen and BH 40 or 10 mg/ml mixed formula treated rats as compared with tamoxifen single formula treated rats; they were detected as 27.14±7.24 and 21.18±7.61 ng/ml in tamoxifen and BH 20 or 10 mg/ml mixed formula treated rats, respectively (Table 1).

4. Changes on the AUC of tamoxifen

The AUC0-t of tamoxifen were non-significantly changed as 6.41 and −33.69% points in tamoxifen and BH 40 or 20 mg/ml mixed formula treated rats as compared with tamoxifen single formula treated rats, respectively. They were detected as 260.11±46.59 and 162.10±65.97 hr · ng/ml in tamoxifen and BH 40 or 20 mg/ml mixed formula treated rats, and as 244.44±85.90 hr · ng/ml in tamoxifen single formula treated rats, respectively. However, the AUC0-t of tamoxifen was significantly (p<0.05) decreased as -56.63% points in tamoxifen and BH 10 mg/ml mixed formula treated rats (106.01±37.79 hr · ng/ml) as compared with tamoxifen single formula treated rats, in this experiment. In addition, the AUC0-inf of tamoxifen were non-significantly changed as 5.12 and −34.68% points in tamoxifen and BH 40 or 20 mg/ml mixed formula treated rats as compared with tamoxifen single formula treated rats, respectively. They were detected as 271.99±56.50 and 169.01±66.94 hr · ng/ml in tamoxifen and BH 40 or 20 mg/ml mixed formula treated rats, and as 258.74±81.02 hr · ng/ml in tamoxifen single formula treated rats, respectively. However, the AUC0-inf of tamoxifen was significantly (p<0.01) decreased as −53.83% points in tamoxifen and BH 10 mg/ml mixed formula treated rats (119.45±33.45 hr · ng/ml) as compared with tamoxifen single formula treated rats, in the current result (Table 1).

5. Changes on the t1/2 of tamoxifen

The t1/2 of tamoxifen were non-significantly changed as 12.31, 1.32 and −29.50% points in tamoxifen and all three different concentrations of BH mixed formula treated rats as compared with tamoxifen single formula treated rats, respectively. They were detected as 4.98±1.20, 4.50±1.28 and 3.13±1.23 hr in tamoxifen and BH 40, 20 or 10 mg/ml mixed formula treated rats, and as 4.44±0.98 hr in tamoxifen single formula treated rats, in our experiment (Table 1).

6. Changes on the MRTinf of tamoxifen

The MRTinf of tamoxifen were significantly (p<0.05) decreased as −21.43% points in tamoxifen and BH 10 mg/ml mixed formula treated rats (4.83±0.91 hr) as compared with tamoxifen single formula treated rats (6.15±0.63 hr). The MRTinf of tamoxifen were non-significantly changed as 15.40 and −5.00% points in tamoxifen and BH 40 or 20 mg/ml mixed formula treated rats as compared with tamoxifen single formula treated rats; they were detected as 7.10±1.32 and 5.84±1.16 hr in tamoxifen and BH 40 or 10 mg/ml mixed formula treated rats, in this study (Table 1).

Discussion

Although single oral administration of mixed formulas consisted of tamoxifen and BH 40 mg/ml did not induced any significant changed on the plasma tamoxifen concentrations, but significant decreases of plasma tamoxifen concentrations were demonstrated in tamoxifen and BH 20 mg/ml mixed formula treated rats at 1, 2 and 6 hr after administration, and also in BH 10 mg/ml mixed formula treated rats from 2 to 8 hr after end of treatment as compared with tamoxifen 0.4 mg/ml single formula treated rats, respectively. In addition, significant increases of Tmax in tamoxifen and BH 40 mg/ml mixed formula treated rats, significant decreases of Cmax in tamoxifen and BH 20 mg/ml mixed formula treated rats, and significant decreases of AUC0-t, AUC0-inf and MRTinf in tamoxifen and BH 10 mg/ml mixed formula treated rats were demonstrated as compared with tamoxifen 0.4 mg/ml single formula treated rats, respectively. These findings are considered as direct evidences that single oral administration of mixed formulation with BH 10 mg/ml concentration significantly decreased the oral bioavailability of tamoxifen through decrease of the absorptions, but not in tamoxifen and BH 40 and 20 mg/ml mixed formulas, suggesting formulated BH concentration-independencies in the current study. It, therefore, seems to be need that pharmacokinetic study after repeated administration of mixed formulas for considerable periods should be tested to conclude the effects of mixed formulation with BH on the pharmacokinetics of tamoxifen, and can be used as potent comprehensive and integrative medicine for the mammary cancer.
Tamoxifen was absorbed slowly following oral administration and Tmax of tamoxifen occur about 3–6 hr after a single dose6,61) but it rapidly and extensively metabolized in the liver, through a substrate of CYP3A, 2C9, 2D626 including an active major metabolite, N-desmethyltamoxifen has biologic activity similar to that of the parent drug60,61). Steady-state concentrations of tamoxifen are attained after 3–4 weeks and those of N-desmethyltamoxifen, an active metabolite, are attained after 3–8 weeks62). Tamoxifen excreted principally in feces as polar conjugates63) with about 5–7 days of t1/2 in tamoxifen and 9–14 days in N-desmethyltamoxifen59). Clearance of tamoxifen is higher in female children 2–10 years of age than in women64,65). In the present study, Tmax of tamoxifen in tamoxifen 0.4 mg/ml single formula oral treated rats was detected as 1.10±0.55 hr, and Cmax, AUC0-t, AUC0-inf, t1/2 and MRTinf were detected as 31.98±8.98 ng/ml, 244.44±85.90 hr · ng/ml, 258.74±81.02 hr · ng/ml, 4.44±0.98 hr and 6.15±0.63 hr, respectively. In tamoxifen and BH 40 mg/ml mixed formula administrated rats, Tmax, Cmax, AUC0-t, AUC0-inf, t1/2 and MRTinf of tamoxifen were detected as 3.00±1.00 hr, 27.14±7.24 ng/ml, 260.11±46.59 hr · ng/ml, 271.99±56.50 hr · ng/ml, 4.98±1.20 hr and 7.10±1.32 hr as changed as 172.73, −15.13, 6.41, 5.12, 12.31 and 15.40% points as compared with tamoxifen 0.4 mg/ml single formula treated rats, in the present study. They showed significant (p<0.05) decreases of Tmax, but not in AUC0-t and AUC0-inf as compared with tamoxifen 0.4 mg/ml single formula treated rats, respectively. In addition, tamoxifen and BH 20 mg/ml mixed formula administrated rats also showed significant (0<0.01) reduced of Cmax, but not in AUC0-t and AUC0-inf as compared with tamoxifen 0.4 mg/ml single formula treated rats, respectively. They showed 27.27, −32.90, −33.69, −34.68, 1.32 and −5.00% points of Tmax, Cmax, AUC0-t, AUC0-inf, t1/2 and MRTinf as compared with tamoxifen 0.4 mg/ml single formula treated rats, and detetced as 1.40±0.89 hr, 21.46±4.01 ng/ml, 162.10±65.97 hr · ng/ml, 169.01±66.94 hr · ng/ml, 4.50±1.28 hr and 5.84±1.16 hr hr, in our result. However, Tmax, Cmax, AUC0-t, AUC0-inf, t1/2 and MRTinf of tamoxifen in tamoxifen and BH 10 mg/ml mixed formula administrated rats were detected as 1.20±0.45 hr, 21.18±7.61 ng/ml, 106.01± 37.79 hr · ng/ml, 119.45±33.45 hr · ng/ml, 3.13±1.23 hr and 4.83±0.91 hr as changed as 9.09, −33.77, −56.63, −53.83, −29.50 and −21.43% points as compared with tamoxifen 0.4 mg/ml single formula treated rats; They showed significant (p<0.01 or p<0.05) decreases of AUC0-t, AUC0-inf and MRTinf as compared with tamoxifen 0.4 mg/ml single formula treated rats, in the current study.
Tamoxifen rapidly and extensively metabolized in the liver, through a substrate of CYP3A, 2C9, 2D626 to active major metabolite, N-desmethyltamoxifen6,61) and, therefore, tamoxifen can be interacted with various drugs2936). In addition the possibilities that tamoxifen competitively interacted with cyclosporine, erythromycin, diltiazem, erythromycin and nifedipine were also suggested in vitro experiments3739). The severities of various side effects arise from tamoxifen treatment, especially bone loss10), endometrial cancer11), thromboembolism12), fatty liver13), reduced cognition14), semantic memory scores15) and libido16,17), premature growth plate fusion18), immune suppression19,20) and hypersensitivity21,22) are considered as directly co-related with absorption and excretion of tamoxifen or pharmacodynamics. In the present study, it is demonstrated that single oral administration of mixed formulation with BH 10 mg/ml concentration significantly decreased the oral bioavailability of tamoxifen through decrease of the absorptions, but not in tamoxifen and BH 40 and 20 mg/ml mixed formulas, suggesting formulated BH concentration-independencies and repeated administration of mixed formulas for considerable periods seems to be need to conclude the effects of mixed formulation with BH on the pharmacokinetics of tamoxifen and whether can be adjusted to mammary cancer patient as potent comprehensive and integrative medicine, at least in a condition of this experiment.

Conclusions

Taken together, single oral administration of tamoxifen 0.4 mg/ml and BH 10 mg/ml mixed formulation induced significant decrease of the oral bioavailability of tamoxifen through inhibition of absorptions. However, tamoxifen 0.4 mg/ml and BH 40 or 20 mg/ml mixed formulation did not critically influenced on the oral bioavailability (AUCs) of tamoxifen, suggesting formulated BH concentration-independencies. It, therefore, seems to be need that pharmacokinetic study after repeated administration of mixed formulas for considerable periods should be tested to conclude the effects of mixed formulation with BH on the pharmacokinetics of tamoxifen. It can be used as potent comprehensive and integrative medicine for the mammary cancer.

Acknowledgments

This work was supported by the National Research Foundation of Korea(NRF) grant funded by the Korea government(MSIT) (No.2018R1A5A 2025272), and by grant of Korea of Health & Welfare, Republic of Korea (Project No: 20-11-0-090-091-3000-3033-320).

Fig. 1
Plasma concentrations of tamoxifen after single oral administration of tamoxifen single or mixed formulas with BH 40, 20 or 10 mg/ml in male rats. Values are expressed as mean ± S.D. of five rats (ng/ml). BH = Blue honeysuckle (fruit parts of Lonicera caerulea L., Caprifoliaceae) concentrated and lyophilized powder.
jkm-40-4-1f1.gif
Table 1
Pharmacokinetic parameters of tamoxifen after single oral administration of tamoxifen single or mixed formulas with BH 40, 20 or 10 mg/ml in male rats.
Treatment PK Parameters Tamoxifen single formula Tamoxifen mixed formulas with BH

0.4 mg/ml 40 mg/ml 20 mg/ml 10 mg/ml
Tmax (hr) 1.10±0.55 3.00±1.00 b 1.40±0.89 1.20±0.45
Cmax (ng/ml) 31.98±8.98 27.14±7.24 21.46±4.01 b 21.18±7.61
AUC0-t (hr · ng/ml) 244.44±85.90 260.11±46.59 162.10±65.97 106.01±37.79 b
AUC0-inf (hr · ng/ml) 258.74±81.02 271.99±56.50 169.01±66.94 119.45±33.45 b
t1/2 (hr) 4.44±0.98 4.98±1.20 4.50±1.28 3.13±1.23
MRTinf (hr) 6.15±0.63 7.10±1.32 5.84±1.16 4.83±0.91 b

Values are expressed as mean ± S.D. of five rats. BH = Blue honeysuckle (fruit parts of Lonicera caerulea L., Caprifoliaceae) concentrated and lyophilized powder. Cmax: The peak plasma concentration; Tmax: Time to reach Cmax; AUC0-t: The total area under the plasma concentration-time curve from time zero to time measured; AUC0-inf: The total area under the plasma concentration-time curve from time zero to time infinity; t1/2: Half life; MRTinf: Mean residence to time infinity.

a p<0.01 and

b p<0.05 as compared with tamoxifen single formula administered rats.

References

1. Ruiz-Garcia A, Bermejo M, Moss A, Casabo VG. Pharmacokinetics in drug discovery. Journal of pharmaceutical sciences. 2008; 97:2. 654–90.
crossref pmid

2. Kang SB, Shon HS, Park SJ, Song CH, Ku SK. Effects of Chungsinoryungsan, a polyherbal complex, on the pharmacokinetic profiles of perindopril in rats. Biomedical reports. 2014; 2:6. 855–60.
crossref

3. Baek KM, Kwon OD, Kim HS, Park SJ, Song CH, Ku SK. Pharmacokinetic Profiles of Donepezil in Combination with Gwibi-Chongmyungtang in Rats. Int J Pharmacol. 2015; 11:4. 343–50.
crossref

4. Kim DJ, Ryu HM, Park Si, Park SJ, Song CH, Ku SK. Pharmacokinetic properties of ondansetron in combination with ijintang-gamibang, polyherbal complex in rats. Int J Pharmacol. 2015; 11:351–8.
crossref

5. Mouridsen H, Giobbie-Hurder A, Goldhirsch A, Thurlimann B, Paridaens R; , et alBIG 1–98 Collaborative Group. Letrozole therapy alone or in sequence with tamoxifen in women with breast cancer. The New England journal of medicine. 2009; 361:8. 766–76.
crossref pmid pmc

6. Jordan VC. Fourteenth Gaddum Memorial Lecture. A current view of tamoxifen for the treatment and prevention of breast cancer. British journal of pharmacology. 1993; 110:2. 507–17.
crossref pmid pmc

7. Jordan VC. Tamoxifen (ICI46,474) as a targeted therapy to treat and prevent breast cancer. British journal of pharmacology. 2006; 147:Suppl 1. S269–76.
crossref pmid pmc

8. Massarweh S, Osborne CK, Creighton CJ, Qin L, Tsimelzon A, Huang S, et al. Tamoxifen resistance in breast tumors is driven by growth factor receptor signaling with repression of classic estrogen receptor genomic function. Cancer research. 2008; 68:3. 826–33.
crossref pmid

9. Hurtado A, Holmes KA, Geistlinger TR, Hutcheson IR, Nicholson RI, Brown M, et al. Regulation of ERBB2 by oestrogen receptor-PAX2 determines response to tamoxifen. Nature. 2008; 456:7222. 663–6.
crossref pmid pmc

10. Vehmanen L, Elomaa I, Blomqvist C, Saarto T. Tamoxifen treatment after adjuvant chemotherapy has opposite effects on bone mineral density in premenopausal patients depending on menstrual status. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2006; 24:4. 675–80.
crossref

11. Grilli S. Tamoxifen (TAM): the dispute goes on. Annali dell’Istituto superiore di sanita. 2006; 42:2. 170–3.
pmid

12. Decensi A, Maisonneuve P, Rotmensz N, Bettega D, Costa A, Sacchini V, et al. Effect of tamoxifen on venous thromboembolic events in a breast cancer prevention trial. Circulation. 2005; 111:5. 650–6.
crossref pmid

13. Osman KA, Osman MM, Ahmed MH. Tamoxifen-induced non-alcoholic steatohepatitis: where are we now and where are we going? Expert opinion on drug safety. 2007; 6:1. 1–4.
crossref pmid

14. Paganini-Hill A, Clark LJ. Preliminary assessment of cognitive function in breast cancer patients treated with tamoxifen. Breast cancer research and treatment. 2000; 64:2. 165–76.
crossref pmid

15. Eberling JL, Wu C, Tong-Turnbeaugh R, Jagust WJ. Estrogen- and tamoxifen-associated effects on brain structure and function. NeuroImage. 2004; 21:1. 364–71.
crossref pmid

16. Mortimer JE, Boucher L, Baty J, Knapp DL, Ryan E, Rowland JH. Effect of tamoxifen on sexual functioning in patients with breast cancer. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 1999; 17:5. 1488–92.
crossref

17. Cella D, Fallowfield L, Barker P, Cuzick J, Locker G, Howell A, et al. Quality of life of postmenopausal women in the ATAC (“Arimidex”, tamoxifen, alone or in combination) trial after completion of 5 years’ adjuvant treatment for early breast cancer. Breast cancer research and treatment. 2006; 100:3. 273–84.
crossref pmid

18. Karimian E, Chagin AS, Gjerde J, Heino T, Lien EA, Ohlsson C, et al. Tamoxifen impairs both longitudinal and cortical bone growth in young male rats. Journal of bone and mineral research: the official journal of the American Society for Bone and Mineral Research. 2008; 23:8. 1267–77.
crossref

19. Wilson SC, Knight PG, Cunningham FJ. Evidence for the involvement of central conversion of testosterone to oestradiol-17 beta in the regulation of luteinizing hormone secretion in the cockerel. The Journal of endocrinology. 1983; 99:2. 301–10.
crossref pmid

20. Nalbandian G, Paharkova-Vatchkova V, Mao A, Nale S, Kovats S. The selective estrogen receptor modulators, tamoxifen and raloxifene, impair dendritic cell differentiation and activation. Journal of immunology (Baltimore, Md: 1950). 2005; 175:4. 2666–75.
crossref

21. Berstein LM, Wang JP, Zheng H, Yue W, Conaway M, Santen RJ. Long-term exposure to tamoxifen induces hypersensitivity to estradiol. Clinical cancer research: an official journal of the American Association for Cancer Research. 2004; 10:4. 1530–4.
crossref

22. Rousset-Jablonski C, Thalabard JC, Gompel A. Tamoxifen contraindicated in women with hereditary angioedema? Annals of oncology: official journal of the European Society for Medical Oncology. 2009; 20:7. 1281–2.
crossref

23. Fisher B, Costantino JP, Wickerham DL, Redmond CK, Kavanah M, Cronin WM, et al. Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study. Journal of the National Cancer Institute. 1998; 90:18. 1371–88.
crossref pmid

24. Pienta KJ, Redman BG, Esper PS, Flaherty LE. A phase II evaluation of oral tamoxifen and intermittent intravenous vinblastine in hormone-refractory adenocarcinoma of the prostate. American journal of clinical oncology. 1996; 19:5. 500–3.
crossref pmid

25. Zeneca Pharmaceuticals. Nolvadex (tamoxifen citrate) prescribing information. 1998.


26. Braems G, Denys H, De Wever O, Cocquyt V, Van den Broecke R. Use of tamoxifen before and during pregnancy. The oncologist. 2011; 16:11. 1547–51.
crossref pmid pmc

27. Karn A, Jha AK, Shrestha S, Acharya B, Poudel S, Bhandari RB. Tamoxifen for breast cancer. JNMA; journal of the Nepal Medical Association. 2010; 49:177. 62–7.
crossref pmid

28. Notley LM, Crewe KH, Taylor PJ, Lennard MS, Gillam EM. Characterization of the human cytochrome P450 forms involved in metabolism of tamoxifen to its alpha-hydroxy and alpha,4-dihydroxy derivatives. Chemical research in toxicology. 2005; 18:10. 1611–8.
crossref pmid

29. Lien EA, Anker G, Lonning PE, Solheim E, Ueland PM. Decreased serum concentrations of tamoxifen and its metabolites induced by aminoglutethimide. Cancer research. 1990; 50:18. 5851–7.
pmid

30. Ritchie LD, Grant SM. Tamoxifen-warfarin interaction: the Aberdeen hospitals drug file. BMJ. 1989; 298:6682. 1253
crossref pmid pmc

31. Tenni P, Lalich DL, Byrne MJ. Life threatening interaction between tamoxifen and warfarin. BMJ. 1989; 298:6666. 93
crossref

32. Lamberts SW, Verleun T, Hofland L, Oosterom R. Differences in the interaction between dopamine and estradiol on prolactin release by cultured normal and tumorous human pituitary cells. The Journal of clinical endocrinology and metabolism. 1986; 63:6. 1342–7.
crossref

33. Dowsett M, Pfister C, Johnston SR, Miles DW, Houston SJ, Verbeek JA, et al. Impact of tamoxifen on the pharmacokinetics and endocrine effects of the aromatase inhibitor letrozole in postmenopausal women with breast cancer. Clinical cancer research: an official journal of the American Association for Cancer Research. 1999; 5:9. 2338–43.


34. Reid AD, Horobin JM, Newman EL, Preece PE. Tamoxifen metabolism is altered by simultaneous administration of medroxyproge-sterone acetate in breast cancer patients. Breast cancer research and treatment. 1992; 22:2. 153–6.
crossref

35. Dehal SS, Brodie AM, Kupfer D. The aromatase inactivator 4-hydroxyandrostenedione (4-OH-A) inhibits tamoxifen metabolism by rat hepatic cytochrome P-450 3A: potential for drug-drug interaction of tamoxifen and 4-OH-A in combined anti-breast cancer therapy. Drug metabolism and disposition: the biological fate of chemicals. 1999; 27:3. 389–94.


36. West CM, Reeves SJ, Brough W. Additive interaction between tamoxifen and rifampicin in human biliary tract carcinoma cells. Cancer letters. 1990; 55:2. 159–63.
crossref

37. Mani C, Gelboin HV, Park SS, Pearce R, Parkinson A, Kupfer D. Metabolism of the antimammary cancer antiestrogenic agent tamoxifen. I. Cytochrome P-450-catalyzed N-demethylation and 4-hydroxylation. Drug metabolism and disposition: the biological fate of chemicals. 1993; 21:4. 645–56.


38. Mani C, Pearce R, Parkinson A, Kupfer D. Involvement of cytochrome P4503A in catalysis of tamoxifen activation and covalent binding to rat and human liver microsomes. Carcinogenesis. 1994; 15:12. 2715–20.
crossref

39. Williams JA, Ring BJ, Cantrell VE, Jones DR, Eckstein J, Ruterbories K, et al. Comparative metabolic capabilities of CYP3A4, CYP3A5, and CYP3A7. Drug metabolism and disposition: the biological fate of chemicals. 2002; 30:8. 883–91.
crossref

40. Kwak MA, Park SJ, Park SH, Lee YJ, Ku SK. Effect of Jaeumkanghwatang (JEKHT), a Polyherbal Formula on the Pharmacokinetics Profiles of Tamoxifen in Male SD Rats (1)-Single Oral Combination Treatment of Tamoxifen 50 mg/kg with JEKHT 100 mg/kg within 5 min -. J Korean Med. 2016; 37:2. 1–11.
crossref

41. Park SJ, Kwak MA, Park SH, Lee YJ, Ku SK. Effect of Jaeumkanghwatang (JEKHT), a Polyherbal Formula on the Pharmacokinetics Profiles of Tamoxifen in Male SD Rats (2) - Oral Combination Treatment of Tamoxifen 50 mg/kg with JEKHT 100 mg/kg on JEKHT 6-day Repeated Pretreated Rats with 8-day Repeated Co-administration -. J Soc Preventive Korean Med. 2016; 20:2. 97–109.


42. Ryu EA, Kang SJ, Song CH, Lee BH, Choi SH, Han CH, et al. Effect of Gamiondam-tang (GMODT), a Polyherbal Formula on the Pharmacokinetics Profiles of Tamoxifen in Male SD Rats. J Korean Med. 2017; 38:2. 61–72.
crossref

43. Ryu EA, Kang SJ, Song CH, Lee BH, Choi SH, Han CH, et al. Effect of Gamiondam-tang (GMODT), a Polyherbal Formula on the Pharmacokinetics Profiles of Tamoxifen in Male SD Rats (2) - Single Oral Combination Treatment of Tamoxifen 50 mg/kg with GMODT 100 mg/kg with 2.5 hr-intervals -. Journal of Society of Preventive Korean Medicine. 2017; 21:2. 127–37.
crossref

44. Svarcova I, Heinrich J, Valentova K. Berry fruits as a source of biologically active compounds: the case of Lonicera caerulea . Biomedical papers of the Medical Faculty of the University Palacky, Olomouc, Czechoslovakia. 2007; 151:2. 163–74.
crossref

45. Zhao H, Wang Z, Ma F, Yang X, Cheng C, Yao L. Protective effect of anthocyanin from Lonicera Caerulea var. Edulis on radiation-induced damage in mice. International journal of molecular sciences. 2012; 13:9. 11773–82.
crossref

46. Jurgoński A, Juśkiewicz J, Zduńczyk Z. An anthocyanin-rich extract from Kamchatka honeysuckle increases enzymatic activity within the gut and ameliorates abnormal lipid and glucose metabolism in rats. Nutrition. 2013; 29:6. 898–902.
crossref

47. Palíková I, Valentová K, Oborná I, Ulrichová J. Protectivity of blue honeysuckle extract against oxidative human endothelial cells and rat hepatocyte damage. Journal of agricultural and food chemistry. 2009; 57:15. 6584–9.
crossref

48. Jin XH, Ohgami K, Shiratori K, Suzuki Y, Koyama Y, Yoshida K, et al. Effects of blue honeysuckle (Lonicera caerulea L.) extract on lipopolysaccharide-induced inflammation in vitro and in vivo. Experimental eye research. 2006; 82:5. 860–7.
crossref

49. Chen L, Xin X, Yuan Q, Su D, Liu W. Phytochemical properties and antioxidant capacities of various colored berries. Journal of the science of food and agriculture. 2014; 94:2. 180–8.
crossref

50. Vostálová J, Galandáková A, Paliková I, Ulrichová J, Doležal D, Lichnovská R, et al. Lonicera caerulea fruits reduce UVA-induced damage in hairless mice. Journal of photochemistry and photobiology B, Biology. 2013; 128:1–11.
crossref

51. Kim HS, Park SI, Choi SH, Song CH, Park SJ, Shin YK, et al. Single oral dose toxicity test of blue honeysuckle concentrate in mice. Toxicological research. 2015; 31:1. 61–8.
crossref

52. AOAC (Association of Official Analytical Chemists). Official Methods of Analysis International. 17th ed. AOAC;2000.


53. Singleton VL, Timberlake CF, Lea AGH. The phenolic cinnamates of white grapes and wine. Journal of the science of food and agriculture. 1978; 29:4. 403–10.
crossref

54. Yang J, Meyers KJ, van der Heide J, Liu RH. Varietal differences in phenolic content and antioxidant and antiproliferative activities of onions. Journal of agricultural and food chemistry. 2004; 52:22. 6787–93.
crossref

55. Boyles MJ, Wrolstad RE. Anthocyanin Composition of Red Raspberry Juice: Influences of Cultivar, Processing, and Environmental Factors. J Food Sci. 1993; 58:5. 1135–41.
crossref

56. Bailer AJ. Testing for the equality of area under the curves when using destructive measurement techniques. Journal of pharmacokinetics and biopharmaceutics. 1988; 16:3. 303–9.
crossref

57. Chiou WL. Critical evaluation of the potential error in pharmacokinetic studies of using the linear trapezoidal rule method for the calculation of the area under the plasma level-time curve. Journal of pharmacokinetics and biopharmaceutics. 1978; 6:6. 539–46.
crossref

58. Kang SJ, Lee JE, Lee EK, Jung DH, Song CH, Park SJ, et al. Fermentation with Aquilariae Lignum enhances the anti-diabetic activity of green tea in type II diabetic db/db mouse. Nutrients. 2014; 6:9. 3536–71.
crossref

59. Adam HK, Patterson JS, Kemp JV. Studies on the metabolism and pharmacokinetics of tamoxifen in normal volunteers. Cancer treatment reports. 1980; 64:6–7. 761–4.


60. Fabian C, Sternson L, Barnett M. Clinical pharmacology of tamoxifen in patients with breast cancer: comparison of traditional and loading dose schedules. Cancer treatment reports. 1980; 64:6–7. 765–73.


61. Murphy C, Fotsis T, Pantzar P, Adlercreutz H, Martin F. Analysis of tamoxifen and its metabolites in human plasma by gas chromatography-mass spectrometry (GC-MS) using selected ion monitoring (SIM). Journal of steroid biochemistry. 1987; 26:5. 547–55.
crossref

62. Jordan VC. Metabolites of tamoxifen in animals and man: identification, pharmacology, and significance. Breast cancer research and treatment. 1982; 2:2. 123–38.
crossref

63. Sun D, Sharma AK, Dellinger RW, Blevins-Primeau AS, Balliet RM, Chen G, et al. Glucuronidation of active tamoxifen metabolites by the human UDP glucuronosyltransferases. Drug metabolism and disposition: the biological fate of chemicals. 2007; 35:11. 2006–14.
crossref

64. AstraZeneca Pharmaceuticals. Nolvadex (tamoxifen citrate) prescribing information. 2003.


65. Hall WA, Doolittle ND, Daman M, Bruns PK, Muldoon L, Fortin D, et al. Osmotic blood-brain barrier disruption chemotherapy for diffuse pontine gliomas. Journal of neuro-oncology. 2006; 77:3. 279–84.
crossref

Editorial office contact information
3F, #26-27 Gayang-dong, Gangseo-gu Seoul, 157-200 Seoul, Korea
The Society of Korean Medicine
Tel : +82-2-2658-3627   Fax : +82-2-2658-3631   E-mail : skom1953.journal@gmail.com
About |  Browse Articles |  Current Issue |  For Authors and Reviewers
Developed in M2PI