Você está na página 1de 6

Fitoterapia 128 (2018) 142–147

Contents lists available at ScienceDirect

Fitoterapia
journal homepage: www.elsevier.com/locate/fitote

Pharmacokinetic profile and oral bioavailability of Kaurenoic acid from T


Copaifera spp. in rats
Dalyara Mendonça de Matosa, Milainy Rocha Vianab, Marcela Cristina de Oliveira Alvimb,
Lara Soares Aleixo de Carvalhoa, Laura Hora Rios Leitec, Ademar Alves Da Silva Filhoa,b,

Jorge Willian Leandro Nascimentoa,d,
a
Post graduation Program in Pharmaceutical Sciences, Federal University of Juiz de Fora, José Lourenço Kelmer Street, Juiz de Fora 36036-900, MG, Brazil
b
Department of Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Juiz de Fora, José Lourenço Kelmer Street, Juiz de Fora 36036-900, MG, Brazil
c
Department of Physiology, Institute of Biological Sciences, Federal University of Juiz de Fora, José Lourenço Kelmer Street, Juiz de Fora 36036-900, MG, Brazil
d
Department of Pharmacology, Institute of Biological Sciences, Federal University of Juiz de Fora, José Lourenço Kelmer Street, Juiz de Fora 36036-900, MG, Brazil

A R T I C LE I N FO A B S T R A C T

Keywords: Kaurenoic acid (KA) is a kaurane diterpene found in several medicinal plants that displays biological activities,
Kaurenoic acid such as anti-inflammatory, smooth muscle relaxant and hypotensive response. However, there are no phar-
Diterpene macokinetic data available about this molecule. The purpose of the study was to determine the pharmaco-
Pharmacokinetics kinetic profile and the oral bioavailability of KA in rats. Wistar rats submitted to jugular vein cannulation
Bioavailability
received 50 mg/kg of KA by intravenous or oral route. The implanted cannula allowed intravenous adminis-
Rat plasma
tration and serial blood collection along 10 h. Analytical quantification was performed by reversed phase
HPLC-UV and mobile phase composed by acetonitrile:acidified water (70:30 v/v). The validated analytical
method showed precision, accuracy, robustness, reliability and linearity between 0.75 and 100 μg/mL. KA
administered intravenously showed a linear and two-compartment kinetic behavior at the tested dose. The
following pharmacokinetic parameters were determined: Cmax = 22.17 ± 1.65 mg/L; V = 14.53 ± 1.47 L/
kg; CL = 17.67 ± 1.50 mL/min/kg; AUC0-∞ = 2859.65 ± 278.42 mg·min/L, K = 0.073 ± 0.005 h−1 and
t1/2β = 9.52 ± 0.61 h. Oral treatment did not provide detectable plasma levels of KA, avoiding the de-
termination of its bioavailability.

1. Introduction respiratory diseases and its pharmacological properties are mainly re-
lated to coumarin and kaurane-type diterpenes [2].
Kaurenoic acid (KA) (ent-kaur-16-en-19-oic acid) is a kaurane-type Kaurenoic acid is a bioactive compound that demonstrates anti-in-
diterpene found in plants commonly used in folk medicine, such as flammatory and analgesic properties associated with inhibition of cy-
copaiba oil (Copaifera langsdorffi) [1], guaco (Mikania glomerata and tokine production and activation of the NO-cyclic GMP-PKG-ATP-sen-
Mikania laevigata) [2] and yacon (Smallanthus sonchifolius) [3]. Copai- sitive K+ channel pathway [5]. Another important effect of this
fera spp. and Mikania spp., due to their well characterized use in tra- substance includes smooth muscle relaxation by blockage of extra-
ditional medicine, have been included among species of governmental cellular Ca2+ influx and opening of ATP-sensitive potassium channels
interest in Brazil to become available for population through the Uni- [6,7]. In fact, several reports indicate a wide spectrum of biological
fied Health System [4]. Copaiba is a large tree that commonly grows in effects of KA, which also includes antimicrobial [3], trypanocidal [8],
some states of Brazil, such as Amazonas, Pará and Ceará. Copaiba hypotensive [9], diuretic [10], antiasthmatic [11] and hypoglycemic
oleoresin is popularly used to treat a variety of conditions, for example, activities [12].
sore throat, urinary and pulmonary affections, and skin diseases [1]. Even presenting a promising therapeutic potential, there are only a
Guaco syrups are also widely applied in folk medicine for treatment of few studies about the cutaneous absorption of KA [13] and the

Abbreviations: KA, kaurenoic acid; IS, internal standard; RT, retention time; HPLC, high-performance liquid chromatography; Cmax, maximum plasma concentration; Tmax, time to reach
Cmax; V, volume of distribution; CL, clearance; AUC, area under the curve; K, elimination rate constant; t1/2β, elimination half-life; CV, coefficient of variation; RE, relative error; SD,
standard deviation; i.v., intravenous; i.p., intraperitoneal

Corresponding author at: Universidade Federal de Juiz de Fora, Instituto de Ciências Biológicas, Laboratório de Farmacologia Clínica e Experimental (LaFaCE), Departamento de
Farmacologia, Rua José Lourenço Kelmer, s/n. 36036-900, Juiz de Fora, MG, Brazil.
E-mail address: jorge.willian@ufjf.edu.br (J.W.L. Nascimento).

https://doi.org/10.1016/j.fitote.2018.05.013
Received 1 December 2017; Received in revised form 4 May 2018; Accepted 13 May 2018
Available online 14 May 2018
0367-326X/ © 2018 Elsevier B.V. All rights reserved.
D.M.d. Matos et al. Fitoterapia 128 (2018) 142–147

bioavailability of guaco syrup metabolites after oral administration by successive dilutions, adding a known amount of KA into blank
[14]. The knowledge of pharmacokinetics and oral bioavailability is plasma. Quality control samples were prepared in plasma at low
crucial to select drug candidates to undergo clinical testing, besides (2.5 μg/mL), medium (50 μg/mL) and high (80 μg/mL) concentrations
providing basis for definition of an effective dosing regimen associated of the analyte. The working solution of internal standard (0.2 μg/mL)
with adequate plasma concentrations [15]. Considering that KA is one was obtained from dilution of stock solution in methanol. All plasma
of the major bioactive compounds found in guaco syrups and represents samples were stored at −20 °C.
a potential candidate for the development of new drugs, the aim of this
study was to determine the pharmacokinetic profile and oral bioavail- 2.4. Plasma samples pretreatment
ability of KA in rats following oral and intravenous administration.
Sample clean-up consisted of protein precipitation with acetonitrile
2. Materials and methods [20]. For each plasma sample (100 μL) it was added 50 μL of internal
standard (0.2 mg/mL) and 100 μL of acetic acid solution (1% v/v). After
2.1. Chemicals and reagents acidification, the sample was mixed for 1 min on a vortex mixer. Fol-
lowing this step, 200 μL of acetonitrile were added and, once more, the
KA was isolated from Copaifera spp. oleoresin following a previously sample was vigorously mixed for 1 min before centrifugation at 10000
described methodology [16]. This substance was kindly donated by Dr. RPM, 4 °C, for 10 min. The supernatant was analyzed by liquid chro-
Sergio Ricardo Ambrósio from The University of Franca (Unifran, matography.
Franca, São Paulo, Brazil). Chemical structure of KA was confirmed by
13
C and 1H NMR analysis. The internal standard (IS), α-bisabolol, was 2.5. Chromatographic analysis
purchased from Sigma Aldrich (St. Louis, USA). Ortho-phosphoric acid
was obtained from Merck (Darmstadt, Germany) and the glacial acetic Concentrations of KA in rat plasma were determined by HPLC-UV.
acid was purchased from Exodo Cientifica (Hortolândia, Brazil). Ul- The method was developed in a Waters system equipped with an
trapure water was obtained using the Purelab Option-Q purification Alliance e2695 Separation Module, quaternary pump, autosampler,
system from Elga (High Wycombe, UK). Methanol and acetonitrile degasser, column heater and double channel UV–Vis detector (Milford,
(HPLC grade) were purchased from J.T. Baker Chemicals (Xalostoc, USA). The Empower 3 software was used for system control, peak in-
Mexico). tegration and data analysis. Separation was carried out in a X-Bridge
C18 column (150 × 4.6 mm, 5 μm; Waters, Milford, USA) attached to a
2.2. Animal experiments C18 guard cartridge (20 × 4.6 mm, 5 μm; Waters, Milford, USA). The
isocratic mobile phase was composed by acetonitrile (A) and acidified
The animal experimental protocol was approved by the Ethical water (B) (ortho-phosphoric acid 0.1%), A:B (70:30, v/v), at a flow rate
Committee for Animal Handling (CEUA number: 040/2014) of the of 1 mL/min. The column heater was maintained at 40 °C and the UV
Federal University of Juiz de Fora and was in agreement with the detector was set at 200 nm. An injection volume of 80 μL was estab-
Ethical Principles in Animal Research adopted by the Brazilian Council lished.
for Control of Animal Experimentation (CONCEA).
Male Wistar rats (200-250 g, n = 6) were used to perform the pre- 2.6. Validation of the analytical method
clinical pharmacokinetic study of KA. The animals were housed in
propylene cages, at 22 ± 2 °C, in groups of four, under a 12 h light- Method validation followed orientations of the guidance for bioa-
dark cycle and with free access to food and water. Following anesthesia nalytical method validation from Food and Drug Admnistration [21].
achieved using a mixture of ketamine (72 mg/kg body weight i.p.) and Selectivity was assessed comparing plasma samples spiked with KA and
xylazine (3 mg/kg body weight i.p.), a silastic catheter was inserted into internal standard with blank samples. Linearity was evaluated over the
the right jugular vein of all animals [17,18]. Immediately after surgery, range of 0.75–100 μg/mL, using the internal standardization method.
the rats received an intramuscular prophylactic dose of antibiotics The calibration curves were constructed plotting peak area ratios (KA
(Pentabiotic, 24,000 IU/kg body wt, Fort Dodge) and a subcutaneous peak area/IS peak area) versus plasma concentration, at seven con-
injection of analgesic medication (Flunixin Meglumine, 1.1 mg/kg body centrations levels of KA (n = 3). A new calibration curve was prepared
wt, Schering-Plogh) [19]. The animals were kept in cages individually for each day of analysis. Lower limit of quantification was investigated
and were allowed to recover for at least 2 days before being submitted by analyzing five replicates of the lowest concentration of the analyte
to the experiments. The implanted cannula was used for intravenous with an acceptable accuracy and precision (< 20%), and limit of de-
administration of the drug, while gavage was used for oral adminis- tection was determined as a concentration with a signal-to-noise
tration. Both intravenous and oral groups received a 50 mg/Kg dose of ratio > 5.
KA prepared in saline with 10% of Tween 80. The jugular cannula also Precision and accuracy (intra-day and inter-day) were assessed by
allowed repeated blood sampling from the right atrium in both ex- quantification of five replicates prepared at low (2.5 μg/mL), medium
perimental groups. Blood samples (200 μL) were collected at the fol- (50 μg/mL) and high (80 μg/mL) concentrations of analyte. Method
lowing intervals: 0 (previous to drug administration), 0.166, 0.333, 0.5, precision and accuracy were expressed as coefficient of variation (CV%)
0.75, 1, 2, 4, 6, 8 and 10 h after administration. The blood volume and relative error (RE%), respectively. Carryover test consisted of in-
collected in each sample was replaced by donated blood from healthy jection of a sample processed at the upper limit of quantification
donor rats to avoid reduction in the animal's blood volume. The samples (100 μg/mL) followed by injections of blank samples. Recovery was
were collected with heparinized syringes and were centrifuged at determined comparing peak area ratio of extracted quality control
10000 RPM, 25 °C, for 10 min. The separated plasma was kept in freezer samples with standard samples of same concentrations prepared in
(−20 °C) until HPLC analysis was carried out. aqueous solution. Stability study evaluated three replicates of quality
control samples (2.5, 50 and 80 μg/mL) at room temperature, auto-
2.3. Preparation of standard solutions, calibration and quality control sampler and after three successive freeze and thaw cycles.
samples
2.7. Data analysis
Stock solutions of KA (1 mg/mL) and internal standard (α-bisabolol,
1 mg/mL) were prepared in methanol and stocked at 4 °C. The cali- Results from the HPLC analysis of the plasma samples were plotted
bration standards (0.75, 1, 2.5, 5, 10, 50, 100, μg/mL) were prepared as drug concentration vs. time to obtain the plasma decay curves of KA

143
D.M.d. Matos et al. Fitoterapia 128 (2018) 142–147

Fig. 1. (A) Selectivity analysis: comparison of blank plasma chromatogram (blue line) with a plasma sample spiked with α-bisabolol (1) and kaurenoic acid (2). (B)
Calibration curve of kaurenoic acid. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

[22]. Two-compartmental pharmacokinetic analysis was performed by KA demonstrated satisfactory stability after short-term analysis
using PK Solutions 2.0 (Summit Research Services, Ashland, USA). The (room temperature), freeze and thaw cycles and post-preparative ana-
area under the concentration-time curve was calculated from the time lysis (in autosampler). The deviation ranges are also presented in
zero to the last data point using trapezoidal method (AUC0-t) and with Table 1. Precision and accuracy results were expressed as coefficient of
extrapolation to infinity (AUC0-∞). The maximum plasma concentration variation (CV%) and relative error (RE%), respectively. Intraday and
(Cmax) and the time to reach Cmax (Tmax) values were observed from the interday analysis of both parameters showed values of CV and
experimental data. The elimination rate constant K was determinate RE < 15%, proving the reliability of the method to quantify KA in
from the slope of the regression line, and the elimination half-life (t1/2β) plasma samples.
obtained by 0.693/K. All data were presented as mean and standard The chosen method to clean-up plasma samples was protein pre-
deviation (mean ± SD). cipitation with acetonitrile. This simple, rapid and low cost technique
results in adequate purification, allowing direct injection in HPLC [23].
Several extraction methods for KA in plasma were tested before, such as
3. Results and discussion solid-phase extraction, protein precipitation with acetonitrile, and li-
quid-liquid extraction applying a variety of solvents. Comparison of
The analytical method proved to be simple, low cost and reliable, these techniques pointed out that protein precipitation was the most
with good performance, linearity, stability, accuracy, precision and efficient for recovery of KA from plasma with high reproducibility [14].
robustness. The addition of the internal standard, α-bisabolol, increased However, when applying this extraction, we observed the phenomenon
the method confidence, facilitating relative quantification of KA. of co-precipitation of the analyte with proteins, one of the dis-
No interfering peaks were detected at the retention times of KA advantages of this clean-up method. To improve KA recovery, an
(RT = 13.2 min) and α-bisabolol (RT = 7.5 min) (Fig. 1A). The cali- acidification step with acetic acid was included before precipitation
bration curve (Fig. 1B) showed good linearity within the range of with acetonitrile, leading to a successful extraction of KA from plasma
0.75–100 μg/mL, providing the mean linear equation y = 0.0135× - matrix.
0.017 (r = 0.999). As mentioned before, despite being known for its great potential as
The results obtained in the validation process are presented in antispasmodic, anti-inflammatory and antihypertensive, no pharmaco-
Table 1. Limit of detection and the low limit of quantification were kinetic study of this substance has been reported following oral or i.v.
0.5 μg/mL and 0.75 μg/mL, respectively. Carryover test proved that no administration.
residual from previous elutions produced interfering peaks at the re- Pharmacokinetic characterizations of natural products are im-
tention times of analyte and internal standard. Therefore, there was no portant to provide useful data to predict biological events and sub-
risk of contamination between injections. stantiate clinical studies [24]. The kinetics of promising molecules have
been described through administration of isolated compounds like the
Table 1 clinical study of the diterpenoid glycosides, stevioside and rebaudioside
Results from analytical method validation. A, two natural sweeteners [25]. As well investigations following oral
Parameter Result administration of plant extracts have been carried out, for example the
preclinical evaluation of Rosmarinus officinalis L. extract, known for its
Linearity 0.75–100 μg/mL
antibacterial, antioxidant, antitumor, anti-inflammatory and anti-
Correlation Coefficient r = 0.999
Limit of Quantification 0.75 μg/mL
nociceptive activities. After oral administration of R. officinalis extract,
Limit of Detection 0.5 μg/mL carnosol, rosmanol, and carnosic acid were found in rat plasma, al-
Estability: 88.92–100.80% lowing the pharmacokinetic study of these diterpenes [26].
- Short term 94.46–105.94% An investigation about the dermal absorption of KA have been
- Freeze and thaw cycles 98.89–102.42%
performed through in vitro Franz-cells' system. Application of KA at 1%
- Autosampler
Precision (CV%) in an oily formulation, after a 24 h exposure time, indicated that
- Intraday 7.2 clinically relevant skin concentrations of KA were achieved – about
- Interday 9.3 10% had been absorbed and found in the skin layers [13].
Accuracy (RE%) 4.9
Another study attempted to determine the kinetic disposition of
- Intraday 6.5
- Interday
guaco metabolites by HPLC-MS/MS in human volunteers that received
Recovery (%) 85 an oral dose of 60 mL of guaco syrup (17.6 mg of coumarin, 1.1 mg of o-

144
D.M.d. Matos et al. Fitoterapia 128 (2018) 142–147

Fig. 2. Typical chromatograms of plasma samples after kaurenoic acid administration in rat. (A) Sample collected 10 min after intravenous administration. (B)
Sample collected 1 h after oral administration.

coumaric acid and 8.9 mg of KA). However, at this dose, no quantifiable significant levels of KA were detected in plasma along 10 h (Fig. 3B),
levels of KA were found in blood samples collected until 10 h after suggesting poor absorption or an extensive pre-systemic elimination of
administration [14]. the drug through this route. Thus, determination of its oral bioavail-
The pharmacokinetic study presented herein was conducted in rats ability was impaired.
treated with 50 mg/kg of the drug through intravenous and oral routes. Following intravenous administration, the KA concentration-time
The technique of jugular vein cannulation, applied for serial blood curve can be best described as a two-compartment model, with two
collection, consists of an efficient vascular access technique suited to distinct phases (Fig. 3A). This information was considered to calculate
monitor temporal changes in blood constituents with little animal the pharmacokinetic parameters of the drug (Table 2). Thus, the dis-
manipulation and distress [18]. In fact, it proved to be ideal for phar- tribution phase (α-phase) was delineated from time zero to 60 min after
macokinetic studies such as this, since it guaranteed optimum re- administration of the drug, and the subsequent period was defined as
producibility among the experimental animals, generating plasma the elimination phase (β-phase). This model assumes the existence of a
decay curves with similar profiles and small deviations. Several studies central compartment, composed by blood and well-perfused tissues that
applying this technique use saline to replace the volume of blood col- easily equilibrate with blood, and another peripheral, which involves
lected. However, considering that a relevant amount of blood is with- tissues where the drug distributes slowly [27]. Once the equilibrium
drawn from animals during the experiment (11 × 200 μL), the blood between central and peripheral compartments is achieved, the plasma
replacement using donor rats instead of saline replacement prevents concentrations decline less rapidly [28].
hemodilution, reducing interferences on drug's pharmacokinetics. The maximum concentration (Cmax) estimated after a single dose of
A typical representative chromatogram obtained from an animal KA was 22.17 ± 1.65 mg/L. The estimated volume of distribution (V)
sample collected 10 min after i.v. administration is shown in Fig. 2A was 14.53 ± 1.47 L/kg. This high value of V reflects the liposolubility
(KA retention time = 13.2 min). On the other hand, Fig. 2B exemplifies of KA, indicating that this molecule is widely distributed in body tissues
a chromatogram of a sample collected 1 h after oral treatment with KA and fluids. The elimination half-life (t1/2β) is considered a dependent
(KA not detected). parameter, since its value depends on clearance (CL) and volume of
Serial blood collections performed in intravenous group furnished distribution: t1/2β = (0.693 V)/CL [27]. The t1/2β of KA was
data to describe the plasma decay of KA (Fig. 3A) and determine its 9.52 ± 0.61 h, while the clearance was 17.67 ± 1.50 mL/min/kg
pharmacokinetics parameters. However, after oral administration, no (Table 2).

Fig. 3. Comparison of kaurenoic acid plasma decay (dose = 50 mg/kg). (A) Intravenous route. (B) Oral route.

145
D.M.d. Matos et al. Fitoterapia 128 (2018) 142–147

Table 2
Pharmacokinetics parameters of kaurenoic acid determined after intravenous administration (dose = 50 mg/kg).
Rat Cmax (mg/L) ASC0-t ASC0-∞ V (L/kg) CL K t1/2β (h)
(mg.min/L) (mg.min/L) (mL/min/kg) (h−1)

1 19.6 1764.1 3368.3 12.2 15 0.073 9.5


2 21.1 1412.8 2583.7 15.5 19 0.075 9.2
3 23.4 1530.9 2785.1 15.6 18 0.069 10.0
4 24.0 1702,9 2953.9 15.0 17 0.068 10.2
5 21.8 1028.3 2676.8 15.7 19 0.071 9.7
6 23.1 1685.5 2790.1 13.2 18 0.081 8.5
Mean ± SD 22.17 ± 1.65 1520.75 ± 273.42 2859.65 ± 278.42 14.53 ± 1.47 17.67 ± 1.50 0.073 ± 0.005 9.52 ± 0.61

The process from plant to medicine – which comprehends the dis- [3] E.P. Padla, L.T. Solis, C.Y. Ragasa, Antibacterial and antifungal properties of ent-
covery, development and launch of a new drug into the market – in- kaurenoic acid from Smallanthus sonchifolius, Chin. J. Nat. Med. 10 (2012) 408–414.
[4] D.J. Marmitt, S. Bitencourt, A.C. Silva, C. Rempel, M.I. Goettert, Scientific pro-
volves high investments, a long period of time and multi-disciplinary duction of plant species included in the Brazilian national list of medicinal plants of
approaches [29]. As discussed before, the knowledge about kinetics interest to the unified health system (RENISUS) from 2010 to 2013, J. Chem.
disposition of drugs in the body brings evidence of the efficacy and Pharm. Res. 8 (2016) 123–132.
[5] S.S. Mizokami, N.S. Arakawa, S.R. Ambrosio, A.C. Zarpelon, R. Casagrande,
safety of active compounds, contributing to the definition of dosing T.M. Cunha, S.H. Ferreira, F.Q. Cunha, W.A. Verri Jr., Kaurenoic acid from
regimens [30]. The lack of preclinical pharmacokinetics studies re- Sphagneticola trilobata inhibits inflammatory pain: effect on cytokine production
garding metabolites from natural products still represents a limitation and activation of the NO-cyclic GMP-protein kinase G-ATP-sensitive potassium
channel signaling pathway, J. Nat. Prod. 75 (2012) 896–904.
in the process of development of new drugs using plant active com- [6] K.M. de Alencar Cunha, L.A.F. Paiva, F.A. Santos, N.V. Gramosa, E.R. Silveira,
pounds, therefore, the kinetic evaluation of KA performed can support V.S.N. Rao, Smooth muscle relaxant effect of kaurenoic acid, a diterpene from
future clinical trials with this bioactive molecule. Copaifera langsdorffii on rat uterus in vitro, Phytother. Res. 17 (2003) 320–324.
[7] C.R. Tirapelli, S.R. Ambrosio, F.B. Costa, S.T. Coutinho, D.C.R. Oliveira,
A.M. Oliveira, Analysis of the mechanisms underlying the vasorelaxant action of
4. Conclusion kaurenoic acid in the isolated rat aorta, Eur. J. Pharmacol. 492 (2004) 233–241.
[8] H.S. Vieira, J.A. Takahashi, A.B. Oliveira, E. Chiari, M.A.D. Boaventura, Novel
Intravenous administration of KA follows a linear and two-com- derivatives of kaurenoic acid: preparation and evaluation of their trypanocidal
activity, J. Braz. Chem. Soc. 13 (2002) 151–157.
partment kinetic behavior at the tested dose. Oral treatment did not [9] C.R. Tirapelli, S.R. Ambrosio, F.B. Costa, A.M. Oliveira, Diterpenes: a therapeutic
provide detectable levels of KA, suggesting poor absorption or an ex- promise for cardiovascular diseases, Recent Pat. Cardiovasc. Drug Discov. 3
tensive pre-systemic elimination through this route. This is the first (2008) 1–8.
[10] L.I. Somova, F.O. Shode, K. Moodley, Y. Govender, Cardiovascular and diuretic
pharmacokinetic study of this molecule. The data presented here elu- activity of kaurene derivatives of Xylopia aethiopica and Alepidea amatymbica, J.
cidates the pharmacokinetic of KA, contributing to future clinical stu- Ethnopharmacol. 77 (2001) 165–174.
dies of this compound. [11] J.H. Cho, J.Y. Lee, S.S. Sim, W.K. Whang, C.J. Kim, Inhibitory effects of diterpene
acids from root of Aralia cordata on IgE-mediated asthma in guinea pigs, Pulm.
Pharmacol. Ther. 23 (2010) 190–199.
Author contributions [12] X. Deng, Y. Shen, J. Yang, J. He, Y. Zhao, L. Peng, Y. Leng, Q. Zhao, Discovery and
structure-activity relationships of ent-Kaurene diterpenoids as potent and selective
11β-HSD1 inhibitors: potential impact in diabetes, Eur. J. Med. Chem. 65 (2013)
A. A. Da Silva-Filho and L. S. A. Carvalho contributed for the partial 403–414.
obtainment of KA and its chemical characterization. L. H. R. Leite and [13] J.P. Mestres, A. Usubillaga, L. Diaz, M. Larroque, L. Vian, G. Marti-Mestres,
M. R. Viana performed animal experiments. Assessment of in vitro dermal absorption of the kaurenic acid from Coespeletia
Moriziana extracts, Fitoterapia 82 (2011) 585–590.
M. R. Viana and M. C. O. Alvim performed HPLC analysis. D. M.
[14] J.C. Gasparetto, R.G. Peccinini, T.M.G. Francisco, L.B. Cerqueira, F.R. Campos,
Matos and J. W. L. Nascimento performed pharmacokinetic studies, R. Pontarolo, A kinetic study of the main guaco metabolites using syrup formulation
analyzed the data and drafted the manuscript. All authors revised the and the identification of an alternative route of coumarin metabolism in humans,
manuscript. PLoS One 10 (2015) 1–22.
[15] B.M. Amore, J.P. Gibbs, M.G. Emery, Application of in vivo animal models to
characterize the pharmacokinetic and pharmacodynamic properties of drug can-
Conflicts of interest didates in discovery settings, Comb. Chem. High Throughput Screen. 13 (2010)
207–218.
[16] M.R. Simão, L.J. Carneiro, R.A. dos Santos, J.K. Bastos, R.C.S. Veneziani,
The authors declare no conflict of interest. S.R. Ambrosio, C.S. Mizuno, In vitro cytotoxicity study of ent-kaurenoic acid deri-
vatives against human breast carcinoma cell line, Med. Chem. Res. 25 (2016)
Acknowledgements 303–309.
[17] L.H.R. Leite, A.C.R. Lacerda, C.H. Balthazar, U. Marubayashi, C.C. Coimbra, Central
angiotensin AT1 receptors are involved in metabolic adjustments in response to
Dr. Sérgio Ricardo Ambrósio, from the University of Franca (Franca, graded exercise in rats, Peptides 30 (2009) 1931–1935.
São Paulo, Brazil) is gratefully acknowledged for providing part of the [18] K.V. Thrivikraman, R.L. Huot, P.M. Plotsky, Jugular vein catheterization for re-
peated blood sampling in the unrestrained conscious rat, Brain Res. Protocol. 10
substance evaluated in this work. The authors are grateful to CAPES,
(2002) 84–94.
PIBIC/CNPq/UFJF and CNPq for the fellowships. This work was sup- [19] C.H. Balthazar, L.H.R. Leite, A.G. Rodrigues, C.C. Coimbra, Performance-enhancing
ported by FAPEMIG (Grant numbers #APQ 0171/11; APQ 02015/14; and thermoregulatory effects of intracerebroventricular dopamine in running rats,
Pharmacol. Biochem. Behav. 93 (2009) 465–469.
PPM-00296-16) and CNPq (Grant number #487221/2012-5).
[20] J.W.L. Nascimento, M.J.C. Carmona, T.M.V. Strabelli, J.O.C. Auler Jr.,
S.R.C.J. Santos, Penetration of cefuroxime in subcutaneous tissue during coronary
References artery bypass grafting surgery, J. Chromatogr. B 877 (2009) 3960–3964.
[21] FDA, Guidance for Industry: Bioanalytical Method Validation, United States
Department of Health and Human Service, http://www.fda.gov/downloads/Drugs/
[1] L.A.F. Paiva, L.A. Gurgel, R.M. Silva, A.R. Tomé, N.V. Gramosa, E.R. Silveira, Guidances/ucm070107.pdf, (2001) (accessed 29.07.2017).
F.A. Santos, V.S.N. Rao, Anti-inflammatory effect of kaurenoic acid, a diterpene [22] W.A. Ritschel, G.L. Kearns, Handbook of Basic Pharmacokinetics: Including Clinical
from Copaifera langsdorffii on acetic acid-induced colitis in rats, Vasc. Pharmacol. 39 Applications, seventh ed., American Pharmacists Association, Washington, 2009.
(2002) 303–307. [23] J. Ma, J. Shi, H. Le, R. Cho, J.C. Huang, S. Miao, B.K. Wong, A fully automated
[2] S.K.V. Bertolucci, A.B.D. Pereira, J.E.B.P. Pinto, J.A.A. Ribeiro, A.B. Oliveira, plasma protein precipitation sample preparation method for LC–MS/MS bioana-
F.C. Braga, Development and validation of an RP-HPLC method for quantification of lysis, J. of Chromatogr. B. 862 (2008) 219–226.
cinnamic acid derivatives and kaurane-type diterpenes in Mikania laevigata and [24] W. Chen, Y. Liang, L. Xie, T. Lu, X. Liu, G. Wang, Pharmacokintics of the Ginkgo B
Mikania glomerata, Planta Med. 75 (2009) 280–285. following intravenous Administration of Ginkgo B emulsion in rats, Biol. Pharm.

146
D.M.d. Matos et al. Fitoterapia 128 (2018) 142–147

Bull. 30 (2007) 1–5. [27] L.A. Bauer (Ed.), Applied clinical pharmacokinetics, second ed., McGraw-Hill, New
[25] A. Wheeler, A.C. Boileau, P.C. Winkler, J.C. Compton, I. Prakash, X. Jiang, York, 2001.
D.A. Mandarino, Pharmacokinetics of rebaudioside A and stevioside after single [28] J.T. DiPiro, W.J. Spruill, R.A. Blouin, J.M. Pruemer (Eds.), Concepts in clinical
oral doses in healthy men, Food Chem. Toxicol. 46 (2008) 54–60. pharmacokinetics, fourth ed., American Society of Health-System Pharmacists,
[26] L. Wang, C. Gan, Z. Wang, L. Liu, M. Gao, Q. Li, C. Yang, Determination and Bethesda, 2005.
pharmacokinetic study of three diterpenes in rat plasma by UHPLC-ESI-MS/MS [29] S.M.K. Rates, Plants as source of drugs, Toxicon 39 (2001) 603–613.
after oral administration of Rosmarinus officinalis L. extract, Molecules 22 (2017) [30] S.S. Singh, Preclinical pharmacokinetics: an approach towards safer and efficacious
1–12. drugs, Curr. Drug Metab. 7 (2006) 165–182.

147

Você também pode gostar