Wang 2018

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Journal of Functional Foods 40 (2018) 292–298

Contents lists available at ScienceDirect

Journal of Functional Foods


journal homepage: www.elsevier.com/locate/jff

Food–drug interactions involving multiple mechanisms: A case study with MARK


effect of Capsaicin on the pharmacokinetics of Irinotecan and its main
metabolites in rat
⁎ ⁎
Nanxi Wang, Chaoran Zhu, Xinlin Zhang, Xuejia Zhai , Yongning Lu
Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China

A R T I C L E I N F O A B S T R A C T

Keywords: Capsaicin (CAP), the main ingredient of chili peppers, displays potent anti-tumor activity in several human
Capsaicin cancers. Irinotecan, an anticancer agent with narrow therapeutic index, is subjected to disposition by several
Irinotecan enzymes or transporters modulated by CAP. This study systematically investigated the CAP–drug interactions in
Metabolite vitro, in situ and in animals. Results indicated that after 7 days of 3 mg/kg CAP treatment, the AUC0-∞ of SN-38
Mechanism
was significantly increased to 1.48-fold compared to the vehicle treated rats. Meanwhile, it caused a significant
Food–drug interactions
plasma protein binding displacement of SN-38, lowered the liver to plasma concentration ratio and slight re-
duction of biliary excretion after CAP treatment. These results demonstrated that ingestion of CAP would in-
crease the systemic exposure and toxicity of SN-38 to a significant extent in rats by affecting multiple enzymes
and transporters at various stages.

1. Introduction that CAP could significantly decrease the bioavailability of digoxin and
simvastatin, which were mainly metabolized by CYP3A (Kato et al.,
Hot pepper fruits, as traditional food or additives, are among the 2012; Zhai, Chen, Liu, Shi, & Lu, 2013). However, some researchers
most heavily consumed spices throughout the world (Hernandez-Ortega found CAP could increase the systemic exposure of the substrate of the
et al., 2012). Capsaicin (CAP, trans-8-methyl-N-vanillyl-6-nonenamide) CYP3A enzyme and P-glycoprotein (P–gp) transporter, and showed a
derived from the natural food, is the most abundant pungent compo- dual inhibition of CYP3A and P–gp (Zhai, Shi, Chen, & Lu, 2013).
nent in chili pepper (Bode & Dong, 2011). It is a valuable pharmaco- Meanwhile, other reports indicated that CAP exhibited inhibition on
logical agent that has been used for chronic pain treatment, gastro OATP1B1 and BCRP transporter (Chen, Zhai, Zhu, & Lu, 2015; Duan
protection and pruritus in various clinical conditions (Gooding, Canter, et al., 2013), which also mediated the pharmacokinetic behaviors of
Coelho, Boddy, & Ernst, 2010). Moreover, emerging studies indicated abovementioned drugs. Hence, these paradoxical results indicated that
that CAP displayed potent anti-tumor activity in various types of cancer other intrinsic underlying mechanisms such as OATP1B1 and BCRP
and could act as a cancer preventive agent, which has motivated re- transporter inhibition may play an important role in these complex
generated attraction for human health and disease (Chapa-Oliver & DDIs. These significant CAP–drug interactions have been recognized for
Mejia-Teniente, 2016; Reyes-Escogido, Gonzalez-Mondragon, & many years, but the biochemical mechanisms have not been fully ex-
Vazquez-Tzompantzi, 2011). plained.
In daily life, the food or spice supplements such as CAP are generally Irinotecan (CPT-11) plays an important role in several cancer regi-
considered safe. However, several studies demonstrated that CAP might mens mainly in advanced colorectal cancer (Vanhoefer et al., 2001). Its
widely influence the drug metabolizing enzymes (DMEs) and drug pharmacokinetics and pharmacodynamics were extremely complex,
transporters (DTs), which may cause complex drug-drug interactions involving multiple DMEs and DTs (Newton, Newman, & Hill, 2012). In
(DDIs) (Babbar, Chanda, & Bley, 2010; Takanohashi, Isaka, Ubukata, short, it was subjected to extensive metabolism by various enzymes
Mihara, & Bernard, 2010). Our previous investigations demonstrated including CES2 to form its active metabolite SN-38, then UGT1A1 to

Abbreviations: APC, 7-ethyl-10-[4-N-(5-aminopentanoic acid)-1-piperidino] carbonyloxy-camptothecin; AUC, Area under the plasma concentration–time curve; CAP, Capsaicin; CES,
Carboxylesterase; CPT, Camptothecin; CPT-11, Irinotecan; CYP, cytochrome P450; DMEs, Drug-metabolizing enzymes; DTs, Drug transporters; LC–MS/MS, Liquid chromatography-
tandem mass spectrometry; RLM, Rat liver microsomes; SD rats, Sprague-Dawley rats; SN-38, 7-ethyl-10-hydroxycamptothecin; SN-38G, SN-38 glucuronide; UGT, Uridine diphosphate
glucuronosyl transferase esterase

Corresponding authors at: No. 1277, Jiefang Avenue, Wuhan, Hubei Province, China.
E-mail addresses: zhaixuejia@163.com (X. Zhai), luyn_union@163.com (Y. Lu).

https://doi.org/10.1016/j.jff.2017.11.014
Received 23 July 2017; Received in revised form 8 November 2017; Accepted 11 November 2017
1756-4646/ © 2017 Elsevier Ltd. All rights reserved.
N. Wang et al. Journal of Functional Foods 40 (2018) 292–298

form SN-38G and CYP3A4 to form APC. Meanwhile, several membrane in biological samples. Briefly, an aliquot of 15 μL of CPT (IS, 1 μg/mL)
transporters, notably BCRP and P–gp, play important roles in their bile was added to 150 μL of rat plasma/tissue homogenate in an Eppendorf
elimination, which greatly affected their pharmacokinetics in vivo tube. After vortex-mixing for 30 s, the mixture was added with 300 μL
(Smith, Figg, & Sparreboom, 2006). For SN-38, the hepatic uptake extraction solvent (methanol containing 0.1% formic acid) to pre-
process was mainly dependent on OATP1B1 transporter (Iusuf et al., cipitate protein, and then the sample was vortex mixed for 5 min and
2014). Based on the aforementioned modulations on DMEs and DTs, centrifuged at 10,000g for 10 min at 4 °C. The clear supernatant fluid
CAP was highly suspected to cause the complex interaction with CPT-11 was injected into the LC–MS/MS system for concentration analysis.
and its metabolites. In addition, SN-38 and CAP both were strongly The chromatographic separation work was carried on an Agilent
bound to albumin in plasma (Fujita et al., 2016), then whether CAP 1200 HPLC system with Agilent ZORBAX Eclipse XDB–C18 column
would increase the free fraction of SN-38 at the target site to cause (2.1 × 50 mm, 3.5 μm). The mobile phase was composed of acetonitrile
adverse effects that was still unknown. – water containing 0.1% formic acid (27:73, v/v) at flow rate of 0.3 mL/
This study performed the in vivo pharmacokinetics study of CPT-11 min, under 35 °C. The mass spectrometer was operated using an AB
and its main metabolites with or without CAP pretreatment, and me- Sciex Qtrap® 4000 tandem mass spectrometer equipped with an elec-
chanistically investigated the behavioral change in the distribution, trospray (ESI) source in positive mode. The optimized condition was
metabolism and excretion phases to elucidate the underlying mechan- consisted of a collision-activated dissociation (CAD) gas of 8 psi, a
isms. It would provide some suggestions for the rational use of CPT-11 curtain gas of 25 psi, a nebulizer gas (GS1) of 40 psi, a Turbo Ion–Spray
and foster the hope of innovative applications of CAP in human disease. gas (GS2) of 40 psi, an ion spray voltage of 5500 V, a source tem-
perature of 400 °C. The monitored ion transitions and collision energy
2. Materials and methods (CE) were as follows: m/z 587.0 → 124.2, 54 V for CPT-11, m/z
393.2 → 349.1, 36 V for SN-38, m/z 569.4 → 393.2, 39 V for SN-38G,
2.1. Chemicals and reagents m/z 619.3 → 393.2, 42 V for APC and m/z 349.1 → 305.1, 34 V for CPT.
Calibration curves were linear over the concentration range of
CPT-11, SN-38, CPT (internal standard, IS) and CAP were purchased 16.9–6760 ng/mL for CPT-11, 0.58–920 ng/mL for SN-38,
from Dalian Meilun Biology Technology Co., Ltd. (Dalian, China). SN- 2.5–1000 ng/mL for SN-38G and 1.25–250 ng/mL for APC. The intra–
38G and APC were purchased from TRC (Toronto, Canada). and inter– batch precision and accuracy of the validated method were
HPLC–grade methanol and acetonitrile were obtained from Merck within the acceptable limits of less than 15%. The assay was sufficiently
(Darmstadt, Germany). Formic acid was purchased from Sigma (St. robust; there was no significant matrix effect from different bio-ma-
Louis, MO, USA). The water used for HPLC was purified by use of a trices or tissues. Stability of the samples under the assay conditions was
Milli–Q system (Millipore, Milford, MA, USA). All other chemicals were secured. Data acquisition and integration were carried out by the
analytical grade and were commercially available. Analyst 1.6.1 software (AB SCIEX).

2.2. Pharmacokinetic studies in rat 2.4. Estimation of plasma protein binding (PPB) by ultrafiltration

Male Sprague–Dawley rats (200 ± 20 g) were obtained from the The plasma protein binding rates of SN-38 in the presence or ab-
Laboratory Animal Center of the Tongji Medical College of Huazhong sence of CAP (0.5 μg/mL) were determined by ultrafiltration method.
University of Science and Technology (Hubei, China quality certifica- Briefly, the fresh rat plasma was prepared from untreated rats and then
tion number: SCXK (E) 2016-0009). The rats were allowed to standard SN-38 was added to the blank plasma in vitro to prepare SN-38 plasma
rat chow and water and maintained under controlled temperature and samples at three concentration levels (0.2, 0.5, 1 μg/mL). The series
humidity (22 ± 2 °C and 45 ± 5%). All animal experimentation pro- concentrations of SN-38 were enough to cover the effective con-
cedures were carried out according to the ‘‘Guiding Principles in the centration of clinical dosing range (Marangon, Posocco, Mazzega, &
Use of Animals in Toxicology’’ adopted by the Society of Toxicology Toffoli, 2015) and CAP were incubated at plasma concentration ob-
(USA) in July 1989 and revised in March 1999 and were approved by tained from published literature (Suresh & Srinivasan, 2010). The
the Ethics Committee of Union Hospital of Huazhong University of prepared samples were placed in a water bath incubator set at 37 °C. At
Science and Technology (Permit Number: 2016-S216, Wuhan, China). the end of the incubation time, 400 μL plasma was put in the ultra-
The rats were randomly divided into three groups (n = 6, each): filtration apparatus (Molecular weight cutoff: 30 KDa, Millipore), cen-
control group (pretreated with 0.5% CMC-Na); positive control group trifuged to obtain ultrafiltrate. The concentrations in the plasma and
(pretreated with repeated doses 10 mg/kg of CsA in 0.5% CMC-Na); filtrate measured at each point were used to calculate the unbound
CAP pretreated group (pretreated with repeated doses 3 mg/kg of CAP fraction through dividing the unbound concentration by the total
in 0.5% CMC-Na). All the rats were given vehicle or drug daily by oral plasma concentration. For sample analysis, 100 μL of filtrate was mixed
gavage for 7 days. In oral gavage operation, wiped the gavage needle with 10 μL of the internal standard solution, then added with 100 μL of
clean after drawing CAP solution, then injected the CAP directly into methanol, vortex-mixed for 30 s and centrifuged at 10,000g for 5 min at
stomach to avoid leaving CAP on the mouth and skin, thereby stimu- 4 °C, and take the supernatant fluid for concentration analysis. For re-
lating animals showed spicy and hot reaction. Twelve hours before covery test of ultrafiltration, SN-38 PBS buffer samples at high, medium
intravenous administration of CPT-11, access to diet was removed and and low (375, 40, 5 ng/mL) concentrations (Cpre) were respectively
only water was provided. On the 7th day, 30 min after pretreatment placed in blank ultrafiltration tube, and centrifuged to obtain ultra-
with CAP, all rats were intravenously injected with CPT-11(HENGRUI filtrate (Cpost) at the same ultrafiltration condition. The recovery rates
MEDICINE, Jiangsu, China) at the dose of 20 mg/kg. Blood samples of the ultrafiltration apparatus were calculated as: Recovery
were collected after injection of CPT-11 at the times of 10, 20, 30 min rate = Cpost/Cpre × 100%. Followed by initial experiment to de-
and 1, 1.5, 2, 3, 4, 6, 8, 10, 24 h. Immediately after sampling, sample termine the incubation time and ultrafiltration condition of SN-38, each
tubes were briefly immersed into ice-water, then immediately cen- experiment was conducted in 5 replicates.
trifuged to obtain plasma, which were stored at −80 °C until analysis.
2.5. Distribution of CPT-11 and its active metabolite in rat liver
2.3. Analytical methods
In this liver distribution study, rats were randomized and orally
In present paper, a rapid, robust and sensitive LC–MS/MS method administered by 0.5% CMC-Na or 3 mg/kg CAP in 0.5% CMC-Na for
was developed and validated to detect CPT-11 and its main metabolites 7 days. On the 7th day, 30 min after pretreatment with CAP, all rats

293
N. Wang et al. Journal of Functional Foods 40 (2018) 292–298

Fig. 1. Plasma concentrations of CPT-11 and its main metabolites after intravenous administration of CPT-11 (20 mg/kg) to rats given oral 0.5% CMC-Na (control group), CsA (10 mg/kg,
positive control group) and CAP (3 mg/kg) for seven consecutive days (n = 6). Data are expressed as mean ± S.D.

were intravenously given by CPT-11 at the dose of 20 mg/kg. Then, the shaking water bath. The Ces2 enzyme incubation mixture with a total
blood and liver samples were immediately collected at 1, 2, 4 h (n = 5, volume of 100 μL contained 40 μL phosphate buffer (pH = 7.4), 10 μL
each). The liver tissue was washed with cold physiological saline, CPT-11 solution (10 μM at final concentration) and 50 μL of microsome
blotted on filter paper and weighed, then cut into pieces. Each gram of protein (0.3 mg/mL at final concentration). The cofactors and substrate
liver tissue was ground into homogenate with 3 mL ice-cold physiolo- of Ugt1a1 enzyme incubation system were performed separately in a
gical saline. The samples were processed by above described method for 37 °C water bath for 5 min. Then, the reaction was started by adding
analysis. 10 μL UDPGA–generating system, 12.5 μL MgCl2, 5 μL SN-38 (5 μM at
final concentration) and 22.5 μL phosphate buffer to 50 μL of micro-
2.6. Metabolism studies some protein (0.3 mg/mL protein). All above system were terminated
by 200 μL of cold methanol contained 0.1% formic and 1000 ng/mL of
To examine the inhibitory potential of CAP against the key enzymes IS, and placed on ice for 3 min. Then, the samples were vortexed for
mediated CPT-11 metabolism, we investigated the CAP effect on the 30 s, centrifuged at 10,000g for 10 min at 4 °C, and the supernatant
activity of Ces2 and Ugt1a1 enzymes in short- and long-term treatment. injected for further analysis.
For short-term treatment, normal rat liver microsomes (RLM) were
incubated CPT-11 or SN-38 with increasing concentrations of CAP
(1 μM, 5 μM, 10 μM, 20 μM, 40 μM, 60 μM, 80 μM and 100 μM) to de- 2.7. Drug interaction in biliary excretion
termine IC50. The rats were given oral 0.5% CMC-Na (control group)
and CAP (3 mg/kg) for seven consecutive days and sacrificed on the In the biliary excretion experiments, male SD rats were randomly
seventh day to get the long-term treated RLM. Then, we also detected divided into control group and CAP pretreated group as described
the enzyme activity by only incubation with CPT-11 or SN-38. above, to give vehicle or CAP daily by oral gavage for 7 days. On the
According to the above process, incubated metabolites with CPT-11 and 7 th day, the abdominal cavity was opened after induction of anesthesia
SN-38 were performed at the final concentration corresponding to the with 10% chloral hydrate and the common bile duct was ligated and a
relative activity and the IC50 value for Ces2 and Ugt1a1 enzymes, re- catheter was inserted into the gallbladder. 30 min after pretreatment
spectively. with CAP, all rats were intravenously given with CPT-11. The bile
The RLM was prepared by the classic differential centrifugation samples were collected and weighted during 0–0.5, 0.5–1, 1–2, 2–3,
method, then the protein concentration of microsome was detected by 3–4 h after which the rats were sacrificed. Bile samples were analyzed
BCA Protein kit (Bradford, 1976). We had optimized the incubation as described below. An aliquot of 10 μL of bile samples was added with
system conditions, such as the concentration of microsome protein and 500 μL of extraction solvent (0.1% formic acid methanol), vortex mixed
the incubation time by univariate analysis. Then, we chose the best and centrifuged, and then the clear supernatant fluid was injected into
condition for the incubation system. The microsome protein and the the LC–MS/MS system.
substrates were pre-incubated separately for 5 min at 37 ± 1 °C. After
pre–incubation, 30 min incubation for samples were carried out in a

294
N. Wang et al. Journal of Functional Foods 40 (2018) 292–298

2.8. Statistical analysis

The pharmacokinetic parameters (AUC, CL, and T1/2) were calcu-


lated by the Drug and Statistics (DAS) 2.0.1 pharmacokinetics fitting
software. The inhibitory IC50 values were calculated and obtained by
fitting the data with nonlinear regression analysis using Prism 5.0
(Graph Pad Software Inc., San Diego, CA, USA). The results of control
and test groups were compared by one-way ANOVA followed by
Fisher’s Protected Least Significance Difference (PLSD) test. All ex-
perimental data were expressed by Mean ± SD. All statistical tests
were performed with SPSS Statistics version 17 (IBM Inc., Armonk,
USA).

3. Results

Fig. 2. PPB displacement of SN-38 in the presence of CAP. Data represent the mean and
3.1. The effect of CAP on the pharmacokinetics of CPT-11 and its main
standard error of mean of five independent experiments. **P < .01, compared with
metabolites
control group.

There were no significant differences in mean body weight between


the control and CAP pretreatment groups after 7 days of treatment. 3.3. CPT-11 and SN-38 concentrations in the rat liver
After intravenous injection at the dose of 20 mg/kg, the plasma con-
centration–time curves of CPT-11 and its main metabolites with dif- To estimate distribution ratio between liver and plasma tissue (L/P
ferent pretreatments were shown in Fig. 1. ratio) and gain insight into the liver distribution, the concentration of
Upon seven consecutive days of pretreatment with CAP at the daily CPT-11 and SN-38 were quantified in rat liver and plasma samples. The
dosages of 3 mg/kg, the blood concentration of SN-38 had significantly liver tissue concentrations of SN-38 in the CAP pretreated group were a
increased (P < .05). Compared to the control group, the AUC0-∞ were little lower at the 1, 2 and 4 h, which were not significantly different
significantly increased to 1.48-fold in the CAP pretreated group, as between the different treatments. The further statistical study indicated
shown in Table 1. The difference in T1/2 did not quite reach statistical that the L/P ratios of SN-38 in the group pretreated with CAP were
significance, but the CL was significantly decreased by 29.92% lower than the control group. Moreover, the difference reach statistical
(P < .05). At the same time, it was interesting to note that the plasma significance at 2 h point (P < .05). For the CPT-11, there were not
concentration of SN-38 was higher than the group pretreated with ve- significant difference in liver distribution between CAP treatment group
hicle during 1 h to 4 h, and the difference was statistically significant and control group (P > .05), as shown in Fig. 3.
(P < .05). However, the main pharmacokinetic parameters of CPT-11,
SN-38G and APC didn’t hint statistically significant difference between 3.4. Effect of CAP on the key enzyme mediated CPT-11 metabolism in RLM
the group pretreated with vehicle and CAP (P > .05), as presented in
Supplementary Table 1. CPT-11 and SN-38 as the probe substrates, were mainly metabolized
by Ces2 enzyme and by Ugt1a1 enzymes, respectively (Zhang, Liu, &
Jeong, 2015). Therefore, the effect of CAP on Ces2 and Ugt1a1 activity
3.2. PPB displacement were further examined using CPT-11 and SN-38, respectively. The
conditions of incubation system in vitro have been optimized, and IC50
The time to reach PPB equilibrium of SN-38 with an oscillation values were estimated in RLM incubation system. It was found that the
frequency of 100 r/min was 60 min. Considering no leakage of protein IC50 of CAP for Ces2 and Ugt1a1 enzyme respectively were 60.74 μM
and proper ultrafiltration ratio, the sample was centrifuged at 7000g for and 408.2 μM respectively (Fig. 4). It demonstrated that CAP exhibited
15 min to obtain filtrate. The recovery rates at three concentrations weak inhibition on Ces2 enzyme and nearly no inhibition on Ugt1a1
(375, 40, 5 ng/mL) were (97.5 ± 1.70)%, (97.1 ± 0.61)% and enzyme. However, the Ces2 enzyme activity in RLM was not sig-
(97.3 ± 1.23)%, respectively, which indicating that SN-38 had no nificantly different after CAP long-term pretreatment. Meanwhile, the
significant adsorption to the ultrafiltration apparatus in the experi- amount of SN-38G was decreased by 8.41%, which indicated the
mental concentration range. PPB displacement studies clarified that SN- Ugt1a1 enzyme activity was decreased by 8.41%, but the difference still
38 in the presence of CAP resulted in significant decrease of the bound didn’t reach significant level (P > .05), as shown in Fig. 4.
ratios of SN-38 in rat plasma. The binding rates of SN-38 (0.2, 0.5, 1 μg/
mL) were (95.7 ± 0.72)%, (94.1 ± 0.97)%, (94.4 ± 0.49)% in rat
3.5. The effect of CAP on bile excretion of the CPT-11 and its main
plasma, respectively. In the presence of CAP (0.5 μg/mL), the plasma
metabolites in rat
protein binding rates of SN-38 were significantly decreased to
(90.7 ± 1.83)%, (88.5 ± 1.91)%, (86.2 ± 2.89)%. Accordingly, the
The cumulative bile excretion of CPT-11, SN-38 and SN-38G in the
unbound SN-38 concentrations were significantly increased to 2.15,
positive control group pretreated with CsA, were much lower than the
1.95 and 2.48-fold (P < .01), shown in Fig. 2.
control group due to the inhibition of the BCRP and P-gp transporters

Table 1
The main pharmacokinetic parameters of SN-38 in rat plasma (Mean ± SD, n = 6).

Analyte Parameter 0.5% CMC-Na CsA 3 mg/kg CAP

* ^3 ^3**
SN-38 AUC(0-∞) (μg h/L) 0.91 ± 0.17 × 10 1.55 ± 0.31 × 10 1.35 ± 0.35 × 10^3*
t1/2 (h) 2.72 ± 1.41 3.11 ± 0.35 3.21 ± 1.42

*Indicates P < .05 and ** Indicates P < .01 compared to the 0.5% CMC-Na (control group).

295
N. Wang et al. Journal of Functional Foods 40 (2018) 292–298

Fig. 3. The distribution concentrations and L/P


ratio of CPT-11 and SN-38 after intravenous ad-
ministration of CPT-11 (20 mg/kg) to rats given
oral 0.5% CMC-Na (control group) and CAP (3
mg/kg) for seven consecutive days (n = 5).
*
P < .05, compared with control group.

(Li et al., 2014). Those target analytes were significantly decreased to (Lin et al., 2017). In the present study, the potential CAP–CPT-11
53.4%, 53.5%, 62.9%, respectively (P < .05). Meanwhile, the cumu- pharmacokinetic interactions were evaluated, which would foster the
lative bile excretion of CPT-11, SN-38 and SN-38G were slightly re- hope of innovative applications of CAP in human cancer. This study
duced in CAP pretreated group (Fig. 5), and the detail cumulative bile investigated the effects of CAP on the pharmacokinetics of CPT-11 and
excretions of CPT-11, SN-38 and SN-38G data were shown in Table 2. its main metabolites in rats and preliminary examine the multiple
mechanisms underlying the drug interactions, including PPB displace-
ment and liver distribution of CPT-11 and SN-38, role of metabolism
4. Discussion
and biliary excretion in vitro and in situ.
In the pharmacokinetic study, the rats were pretreated with CAP at
Currently, CAP has potential antitumor effects and induces apop-
3 mg/kg CAP for 7 day based on the average daily intake of CAP (Zhai
tosis in many types of malignant cell lines, including colon adeno-
et al., 2013) and the pretreatment period reported in several published
carcinoma, hepatocellular carcinoma, breast cancer and many others,
literatures (Chen et al., 2015; Duan et al., 2013). The pharmacokinetic
suggesting that CAP would be a promising therapeutic agent for cancer

Fig. 4. Inhibitory effect of CAP on Ces2 and


Ugt1a1 enzyme. For short-term, reaction velo-
cities are expressed as a ratio (in percentage) re-
lative to the velocity without CAP in RLM. For
long-term, reaction velocities are expressed as
generation amount of SN-38 and SN-38G.

296
N. Wang et al. Journal of Functional Foods 40 (2018) 292–298

Fig. 5. Biliary excretion of CPT-11, SN-38 and SN-38G over 4 h after CPT-11 intravenous injection to rats given oral 0.5% CMC-Na (control group), CsA (10 mg/kg, positive control group)
and CAP (3 mg/kg) for seven consecutive days (n = 6). Data are expressed as mean ± S.D.

Table 2 displacement of SN-38 ex vivo was not the main reason for the in-
Effect of CAP on the biliary excretion cumulative of CPT-11, SN-38 and SN-38G creased unbound concentration of SN-38 in vivo, which wouldn't result
(Mean ± SD, n = 6).
in clinically significant interactions. The findings from this study need
Experimental groups Cumulative excretion of bile (μg/kg/4h) to be understood in conjunction with DMEs inhibition, as well as dis-
position mediated by DTs.
CPT-11 SN-38 SN-38G In the positive control group, CsA as the reported inhibitor of P–gp
^3
and BCRP transporter, had increased systemic availability of CPT-11,
Control group 1.36 ± 0.36 × 10 69.3 ± 14.1 76.5 ± 17.3
CsA pretreated group 0.73 ± 0.14 × 10^3* 37.1 ± 4.09* 48.1 ± 7.88*
SN-38 and SN-38G in rat. Our previous studies also found CAP could
CAP pretreated group 1.02 ± 0.32 × 10^3 60.8 ± 19.9 70.8 ± 15.4 increase systemic exposure of other substrate drugs of P–gp and BCRP
transporters that mediated the biliary excretion of CPT-11, SN-38 and
* Significantly different with P < .05 versus 0.5% CMC-Na (control group). SN-38G. However, through observing the serum profiles, it was inter-
esting to note that there were difference of effect period on pharma-
results illustrated that co-administration of 3 mg/kg CAP increased the cokinetic behavior of SN-38 between CAP and CsA treatments. The
plasma concentration and the systemic exposure of SN-38. Meanwhile, former affected pharmacokinetics mainly during 1–4 h (Fig. 1b) and the
the AUC0-∞ were significantly increased to 1.48-fold (P < .05), after latter increased concentration of SN-38 during 4–8 h. Moreover, the
the rats had ingested a CAP-supplemented diet for 7 days. As we all plasma concentrations of SN-38 were obviously higher than the group
know, SN-38 is the active metabolite of CPT-11 and exert the phar- pretreated with vehicle during 1–4 h, and the difference were still sta-
macological effect. The increased bioavailability of SN-38 was pro- tistically significant (P < .05). It was illustrated that the biochemical
mising to achieve a higher anticancer efficacy in colon carcinoma mechanism may be different in some aspects between the effects of CsA
treatment (Chu et al., 2014). However, SN-38 has a narrow therapeutic and CAP on the pharmacokinetic of SN-38. To test this hypothesis, we
index, and its cytotoxicity is much greater than the parent drug. Higher investigated the biliary excretion outputs of CPT-11 and it metabolites.
SN-38 accumulation level may lead to acute diarrhea and other choli- Compared to the control group, the positive control group pretreated
nergic syndrome and fatal hematological toxicity. Therefore, CAP pre- with CsA, significantly decreased the biliary excretion, which was
administered in a relevant supplementary concentration would enhance consistent with the pharmacokinetic results. The accumulation bile
the pharmacological efficacy and increase the supra-therapeutic level excretion of SN-38 had also decreased in the group pretreated with
risk of SN-38 by heightening plasma concentration levels after intake. CAP, but it didn’t reach the statistical significance (p > .05). The re-
Then, if a patient on CPT-11 treatment intakes excessive CAP-con- sults explained satisfactorily the above different pharmacokinetic be-
taining food/drug, it’s better to reduce the dose of CPT-11 accordingly. havior, and indicated that CAP had a slight regulation in bile elimina-
It was generally accepted that an increase in free fraction of the drug tion of SN-38.
would have effect on its disposition process in vivo, and significantly According to the aforementioned knowledge, there were still several
alter AUC of the drug. For further research, we conducted the PPB DMEs and DTs in the distribution and metabolism system that would
displacement experiment to investigate the unbound plasma con- influence the disposition behavior of SN-38, which were also key
centration changes of SN-38 in rat plasma. Our findings suggested that pathways for SN-38 disposition. Hence, the findings from this study
CAP significantly enhanced the unbound fractions of SN-38 and in- need to be understood in conjunction with tissue distribution, as well as
creased its unbound concentrations in vitro (P < .01). However, other the role of several DMEs.
published literatures (Benet & Hoener, 2002; Shaik, Bohnert, Williams, Distribution refers to the drug within the blood circulation to reach
Gan, & LeDuc, 2016) indicated that the changes in plasma protein the body's tissues and organs process, which has close relationship with
binding in vitro rarely leaded to significant pharmacokinetic interac- the tissue storage, efficacy and adverse effects. Many studies have
tion, as increased free drug concentration would lead to rapid elim- shown Oatp1b2 transporter in rat (OATP1B1 in human) may mediate
ination of the drug until steady state is obtained. Besides, some in- hepatic uptake of SN-38 from plasma in rat. Based on the pharmaco-
vestigators (Fujita et al., 2016) have shown that the significant PPB kinetic results, we investigated the effect of CAP on the distribution

297
N. Wang et al. Journal of Functional Foods 40 (2018) 292–298

from blood to liver during 1–4 h. Interestingly, CAP could reduce the Appendix A. Supplementary material
distribution of SN-38 in liver, and the L/P ratio had a significantly
difference at 2 h (P < .05). Meanwhile, other published literature Supplementary data associated with this article can be found, in the
(Suresh & Srinivasan, 2010) showed that the concentration of CAP in online version, at http://dx.doi.org/10.1016/j.jff.2017.11.014.
liver was highest at 3 h after oral CAP, and then the concentrations of
CAP in liver were decreased rapidly, which suggested the CAP in- References
hibitory concentration may be effective only during 1–4 h, thereby
leading to transient inhibition of Oatp1b2 transporter. Besides, the Babbar, S., Chanda, S., & Bley, K. (2010). Inhibition and induction of human cytochrome
P450 enzymes in vitro by capsaicin. Xenobiotica, 40, 807–816.
tissue distribution results were consistent with our previous study
Benet, L. Z., & Hoener, B. (2002). Changes in plasma protein binding have little clinical
finding that CAP could increase the bioavailability of pitavastatin, the relevance. Clinical Pharmacology and Therapeutics, 71, 115–121.
substrate of Oatp1b2 transporter in rat (Chen et al., 2015). The results Bode, A. M., & Dong, Z. (2011). The two faces of capsaicin. Cancer Research, 71,
2809–2814.
indicated that CAP may inhibit the Oatp1b2 mediated SN-38 uptake to Bradford, M. M. (1976). A rapid and sensitive method for the quantitation of microgram
some extent, which would mainly contribute in this DDIs. quantities of protein utilizing the principle of protein-dye binding. Analytical
Metabolism is structural change process for drugs in the body, Biochemistry, 72, 248–254.
Chapa-Oliver, A. M., & Mejia-Teniente, L. (2016). Capsaicin: From Plants to a Cancer-
which impact on the pharmacokinetics of the drug greatly. Liver mi- Suppressing Agent. Molecules, 21, 931. http://doi.org/10.3390/molecules21080931.
crosomal incubation is the commonly used method for the drug inter- Chen, F., Zhai, X., Zhu, C., & Lu, Y. (2015). Effects of capsaicin on pharmacokinetics of
pitavastatin in rats. Xenobiotica, 45, 171–176.
action study in vitro, because it’s easy to operate and has good re- Chu, C., Abbara, C., Tandia, M., Polrot, M., Gonin, P., Farinotti, R., & Bonhomme-Faivre,
producibility. We evaluated the activity of the key enzymes including L. (2014). Cetuximab increases concentrations of irinotecan and of its active meta-
Ces2 and Ugt1a1 by RLM incubation system. Our findings indicated bolite SN-38 in plasma and tumour of human colorectal carcinoma-bearing mice.
Fundamental & Clinical Pharmacology, 28, 652–660.
that CAP had a weak inhibitory effect on Ces2 (IC50 = 60.74 μM) in Duan, L., Yan, Y., Sun, Y., Zhao, B., Hu, W., & Li, G. (2013). Contribution of TRPV1 and
RLM. The results in vitro appeared to be inconsistent with the phar- multidrug resistance proteins in the permeation of capsaicin across different in-
macokinetic result in vivo, as SN-38 exposure amount was increased in testinal regions. International Journal of Pharmaceutics, 445, 134–140.
Fujita, K., Masuo, Y., Okumura, H., Watanabe, Y., Suzuki, H., Sunakawa, Y., ... Kato, Y.
the CAP pretreated group. However, other study indicated that systemic (2016). Increased Plasma Concentrations of Unbound SN-38, the Active Metabolite of
exposure to CAP which follows from either the ingestion of CAP-con- Irinotecan, in Cancer Patients with Severe Renal Failure. Pharmaceutical Research, 33,
269–282.
taining foods or topical administration of a high-concentration cuta- Gooding, S. M. D., Canter, P. H., Coelho, H. F., Boddy, K., & Ernst, E. (2010). Systematic
neous patch is highly unlikely to inhibit or induce the activities of review of topical capsaicin in the treatment of pruritus. International Journal of
metabolic enzymes (Babbar et al., 2010). Besides, upon seven con- Dermatology, 49, 858–865.
Hernandez-Ortega, M., Ortiz-Moreno, A., Hernandez-Navarro, M. D., Chamorro-Cevallos,
secutive days of pretreatment with CAP, the Ces2 enzyme activity in the G., Dorantes-Alvarez, L., & Necoechea-Mondragon, H. (2012). Antioxidant, anti-
CAP pretreated group was not significantly different compared to the nociceptive, and anti-inflammatory effects of carotenoids extracted from dried
pepper (Capsicum annuum L.). Journal of Biomedicine & Biotechnology, 2012, 524019.
control group. Therefore, the inhibition enzyme activity of CAP on Ces2
http://dx.doi.org/10.1155/2012/524019.
is negligible in vivo; it might be masked or reversed by other me- Iusuf, D., Ludwig, M., Elbatsh, A., van Esch, A., van de Steeg, E., Wagenaar, E., ...
chanism interventions. Schinkel, A. H. (2014). OATP1A/1B transporters affect irinotecan and SN-38 phar-
macokinetics and carboxylesterase expression in knockout and humanized transgenic
Based on our findings, we could draw a conclusion that CAP cer- mice. Molecular Cancer Therapeutics, 13, 492–503.
tainly alters the pharmacokinetic of SN-38 and increased the bioavail- Kato, R., Higashitani, A., Irie, T., Kusukawa, Y., Yamamoto, Y., Nakagawa, M., ... Tanaka,
ability of SN-38 to a significant extent in the rat. The complex DDIs K. (2012). Influence of capsaicin on fluctuation of digoxin pharmacokinetics in li-
popolysaccharide-treated rats. Xenobiotica, 42, 798–807.
mechanisms were originated from the net–effect, which integrated in Li, L., Yao, Q., Xu, S., Hu, H., Shen, Q., Tian, Y., ... Zeng, S. (2014). Cyclosporin A affects
significant plasma protein binding displacement of SN-38, lower L/P the bioavailability of ginkgolic acids via inhibition of P-gp and BCRP. European
Journal of Pharmaceutics and Biopharmaceutics, 88, 759–767.
ratio in distribution phase and slight reduction of biliary excretion in Lin, Y., Wang, H., Hsu, Y., Cho, C., Yang, M., & Chien, C. (2017). Capsaicin induces
elimination phase after CAP treatment. The net–effects have been de- autophagy and apoptosis in human nasopharyngeal carcinoma cells by down-
scribed as a “see–saw” phenomenon (Wu, 2012), and the importance of regulating the PI3K/AKT/mTOR pathway. International Journal of Molecular Sciences,
18, 1343. http://dx.doi.org/10.3390/ijms18071343.
this effect has been elucidated via in vitro, in situ and in animal studies, Marangon, E., Posocco, B., Mazzega, E., & Toffoli, G. (2015). Development and validation
which would be helpful for the rational prediction of possible interac- of a high-performance liquid chromatography-tandem mass spectrometry method for
tion. Meanwhile, for many drugs, such as CPT-11 that modulate by a the simultaneous determination of irinotecan and its main metabolites in human
plasma and its application in a clinical pharmacokinetic study. PLoS ONE, 10,
variety of enzymes and transporters in vivo, it’s not suitable to study e118194. http://dx.doi.org/10.1371/journal.pone.0118194.
and evaluate this series of complex food–drug interactions associated Newton, K. F., Newman, W., & Hill, J. (2012). Review of biomarkers in colorectal cancer.
Colorectal Disease, 14, 3–17.
with multiple mechanisms by only considering single DME and DT. Reyes-Escogido, M. D. L., Gonzalez-Mondragon, E. G., & Vazquez-Tzompantzi, E. (2011).
Otherwise, it could lead to ignorance or wrongly prediction of severe Chemical and pharmacological aspects of capsaicin. Molecules, 16, 1253–1270.
adverse reactions and/or pharmacological effects reduction. Shaik, A. N., Bohnert, T., Williams, D. A., Gan, L. L., & LeDuc, B. W. (2016). Mechanism of
drug-drug interactions between warfarin and statins. Journal of Pharmaceutical
In summary, since the SN-38 disposition was altered when CAP co- Sciences, 105, 1976–1986.
administered to rats, we suggest that care should be taken of the pos- Smith, N. F., Figg, W. D., & Sparreboom, A. (2006). Pharmacogenetics of irinotecan
metabolism and transport: An update. Toxicology in vitro, 20, 163–175.
sible CAP–drug interactions when patient’s concurrent intake of CAP.
Suresh, D., & Srinivasan, K. (2010). Tissue distribution & elimination of capsaicin, pi-
With regard to potential pharmacokinetic drug–CAP interactions, it is perine & curcumin following oral intake in rats. The Indian Journal of Medical
necessary to confirm and clarify these effects of CAP to gain insight into Research, 131, 682–691.
Takanohashi, T., Isaka, M., Ubukata, K., Mihara, R., & Bernard, B. K. (2010). Studies of
any possible interactions between CAP and CPT-11 or other drugs in the toxicological potential of capsinoids, XIII: Inhibitory effects of capsaicin and
further clinical use. capsinoids on cytochrome P450 3A4 in human liver microsomes. International Journal
of Toxicology, 29, 22S–26S.
Vanhoefer, U., Harstrick, A., Achterrath, W., Cao, S., Seeber, S., & Rustum, Y. M. (2001).
Irinotecan in the treatment of colorectal cancer: Clinical overview. Journal of Clinical
Conflict of interest Oncology, 19, 1501–1518.
Wu, B. (2012). Pharmacokinetic interplay of phase II metabolism and transport: A the-
oretical study. Journal of Pharmaceutical Sciences, 101, 381–393.
The authors declare that there are no conflicts of interest. Zhai, X., Chen, J., Liu, J., Shi, F., & Lu, Y. (2013). Food-drug interactions: Effect of
capsaicin on the pharmacokinetics of simvastatin and its active metabolite in rats.
Food and Chemical Toxicology, 53, 168–173.
Zhai, X., Shi, F., Chen, F., & Lu, Y. (2013). Capsaicin pretreatment increased the bioa-
Acknowledgement vailability of cyclosporin in rats: Involvement of P-glycoprotein and CYP 3A inhibi-
tion. Food and Chemical Toxicology, 62, 323–328.
Zhang, N., Liu, Y., & Jeong, H. (2015). Drug-Drug Interaction Potentials of Tyrosine
This work was supported by the National Natural Science Kinase Inhibitors via Inhibition of UDP-Glucuronosyltransferases. Scientific Reports, 5,
Foundation of China (Grants 81473287, 81673715 and 81403012). 17778. http://dx.doi.org/10.1038/srep17778.

298

You might also like