SlideShare a Scribd company logo
1 of 59
P
a
N
A
D
a
A
R
R
A
A
K
C
L
B
N
1
r
C
1
C
m
s
[
i
p
a
r
i
0
h
Colloids and Surfaces B: Biointerfaces 101 (2013) 353– 360
Contents lists available at SciVerse ScienceDirect
Colloids and Surfaces B: Biointerfaces
j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c
a t e / c o l s u r f b
harmacokinetics of curcumin-loaded PLGA and PLGA–PEG
blend nanoparticles
fter oral administration in rats
ajeh Maissar Khalil , Thuane Castro Frabel do Nascimento
, Diani Meza Casa , Luciana Facco Dalmolin ,
na Cristina de Mattos, Ivonete Hoss, Marco Aurélio
Romano, Rubiana Mara Mainardes ∗
epartment of Pharmacy, Universidade Estadual do Centro-
Oeste/UNICENTRO, Rua Simeão Camargo Varela de Sá 03,
85040-080 Guarapuava, PR, Brazil
r t i c l e i n f o
rticle history:
eceived 2 March 2012
eceived in revised form 10 June 2012
ccepted 12 June 2012
vailable online 28 June 2012
eywords:
urcumin
C–MS/MS
ioavailability
anoparticles
a b s t r a c t
The aim of this study was to assess the potential of
nanoparticles to improve the pharmacokinetics of
curcumin, with a primary goal of enhancing its
bioavailability. Polylactic-co-glycolic acid (PLGA) and
PLGA–polyethylene glycol (PEG) (PLGA–PEG) blend
nanoparticles containing curcumin were obtained
by a single-emulsion solvent-evaporation technique,
resulting in particles size smaller than 200 nm. The
encapsulation efficiency was over 70% for both
formulations. The in vitro release study showed that cur-
cumin was released more slowly from the PLGA
nanoparticles than from the PLGA–PEG nanoparticles. A
LC–MS/MS method was developed and validated to quantify
curcumin in rat plasma. The nanoparticles
were orally administered at a single dose in rats, and the
pharmacokinetic parameters were evaluated
and compared with the curcumin aqueous suspension. It was
observed that both nanoparticles formu-
lations were able to sustain the curcumin delivery over
time, but greater efficiency was obtained with
the PLGA–PEG nanoparticles, which showed better results
in all of the pharmacokinetic parameters ana-
lyzed. The PLGA and PLGA–PEG nanoparticles increased
the curcumin mean half-life in approximately 4
and 6 h, respectively, and the Cmax of curcumin increased
2.9- and 7.4-fold, respectively. The distribution
and metabolism of curcumin decreased when it was carried
by nanoparticles, particularly PLGA–PEG
nanoparticles. The bioavailability of curcumin-loaded
PLGA–PEG nanoparticles was 3.5-fold greater than
the curcumin from PLGA nanoparticles. Compared to the
curcumin aqueous suspension, the PLGA and
PLGA–PEG nanoparticles increased the curcumin
bioavailability by 15.6- and 55.4-fold, respectively.
These results suggest that PLGA and, in particular,
PLGA–PEG blend nanoparticles are potential carriers
for the oral delivery of curcumin.
. Introduction
Curcumin is a polyphenol compound extracted from the
oot of Curcuma longa Linn, commonly known as turmeric.
hemically, curcumin is 1,7-bis(4-hydroxy-3-methoxyphenyl)-
,6-heptadiene-3,5-dione, commonly called diferuloylmethane.
urcumin has been used for centuries in Chinese and Indian
edicine to treat a variety of disorders [1]. Several studies have
hown that curcumin presents anti-inflammatory [2,3],
antioxidant
4–6] and antimicrobial activities [7], but the most important
effect
s its potential use against cancer due to its ability to suppresses
the
roliferation of a wide variety of tumor cells [8–10]. Curcumin is
ble to modulate numerous targets including transcription
factors,
eceptors, kinases, cytokines, enzymes and growth factors,
affect-
ng numerous molecular and biochemical cascades [1,11].
∗ Corresponding author. Tel.: +55 42 3629 8160; fax: +55 42
3629 8102.
E-mail address: [email protected] (R.M. Mainardes).
927-7765/$ – see front matter © 2012 Elsevier B.V. All rights
reserved.
ttp://dx.doi.org/10.1016/j.colsurfb.2012.06.024
© 2012 Elsevier B.V. All rights reserved.
The great pharmacological potential of curcumin and its ther-
apeutic applications are restricted because the molecule presents
some drawbacks, including low aqueous solubility at acidic and
physiological pH conditions, rapid hydrolysis in alkaline media
and light instability, inherent to its chemical composition. The
hydrophobic character of curcumin results in pharmacokinetic
restrictions such as low absorption and bioavailability by oral
route,
extensive metabolism and rapid elimination [12,13]. The main
strategies used to overcome the physicochemical limitations of
cur-
cumin and to increase its bioavailability are based on loading
the
compound in nanocarriers, such as liposomes [14],
cyclodextrins
[15], solid lipids [16,17] and polymeric nanoparticles [18–20].
Biodegradable polymeric nanoparticles are extensively used to
improve the therapeutic properties of various drugs and bioac-
tive compounds. Nanoencapsulation protects the molecules from
premature degradation, improves their solubility, and promotes
controlled drug release and drug targeting. Nanoparticles
present
low risk of toxicity, and the drug efficacy, specificity, tolerabil-
ity and the therapeutic index are enhanced with their use. The
pharmacokinetic parameters of the drug are modified when it
dx.doi.org/10.1016/j.colsurfb.2012.06.024
http://www.sciencedirect.com/science/journal/09277765
http://www.elsevier.com/locate/colsurfb
mailto:[email protected]
dx.doi.org/10.1016/j.colsurfb.2012.06.024
3 ces B:
i
i
r
h
s
b
b
m
t
p
n
p
t
t
h
b
a
r
c
i
m
r
a
i
b
A
t
[
a
a
l
f
o
L
r
d
a
2
2
4
p
f
(
c
A
a
c
2
n
s
B
m
w
p
(
U
54 N.M. Khalil et al. / Colloids and Surfa
s loaded in nanoparticles. Specifically, there are improvements
n absorption, bioavailability, and plasma circulation time, with
eduction of clearance, consequently increasing the drug’s mean
alf-life [21–25]. The physicochemical parameters of
nanoparticles
uch as particle size, surface modification charge, and hydropho-
icity influence the drug’s pharmacokinetics, impacting the
drug’s
ioavailability and biodistribution in particular. It is well docu-
ented that nanoparticles presenting a hydrophobic surface, like
he surface of poly (lactide-co-glycolide) (PLGA) nanoparticles,
resent short circulation times because they are rapidly recog-
ized by plasmatic opsonin and cleared by cells of the
mononuclear
hagocytic system (MPS). The process of opsonization is one of
he most important biological barriers to nanoparticle-based con-
rolled drug delivery. Coating of the surface of nanoparticles
with
ydrophilic polymers, such as polyethylene glycol (PEG),
polysor-
ates or poloxamers, sterically stabilizes the particles, i.e., they
re able to repel the absorption of opsonin proteins via steric
epulsion forces, and thus, the particles become “invisible” to
MPS
ells, increasing their plasmatic circulation time and resulting in
an
mprovement in drug bioavailability and half-life. Also, longer
plas-
atic circulation times increase the probability of the
nanoparticles
eaching their target [26–29].
Some recent works have demonstrated that PLGA nanoparticles
re able to improve the bioavailability of curcumin after oral
admin-
stration [18,19,30]. PLGA–PEG nanoparticles have been
developed
ecause of their great potential for having long circulation times.
lso, the potential advantage provided by the hydrophilic charac-
er of PEG can improve the biocompatibility of the delivery
system
31]. However, to the best of our knowledge, there has not yet
been
demonstrated report about the use of PLGA–PEG nanoparticles
s carriers for curcumin. Thus, polymeric nanoparticles,
especially
ong-circulating nanoparticles, were evaluated as potential
carriers
or curcumin oral delivery.
In this work, PLGA and PLGA–PEG blend nanoparticles were
btained for curcumin loading. An analytical method based on
C–MS/MS was developed and validated to quantify curcumin in
at plasma. The nanoparticles were orally administered at a
single
ose in rats, and the pharmacokinetic parameters were evaluated
nd compared with those of a curcumin aqueous suspension.
. Materials and methods
.1. Materials
Curcumin (code C1386), PLGA (Resomer RG 50:50 H; Mw
0–75 kDa, inherent viscosity 0.45–0.6 dl/g), PEG (Mw 10 kDa)
and
olyvinyl alcohol (PVA, 31 kDa, 88% hydrolyzed) were
purchased
rom Sigma–Aldrich (USA). The internal standard, salbutamol
99%), was obtained from European Pharmacopeia. Methylene
hloride and ethyl acetate were purchased from FMaia® (Brazil).
nalytical HPLC-grade ethanol, acetonitrile, methanol and acetic
cid were purchased from J.T. Baker (USA). All other solvents
and
hemicals were analytical or HPLC grade.
.2. Preparation of curcumin-loaded PLGA and PLGA–PEG
blend
anoparticles
The nanoparticles were obtained by the single-emulsion
olvent-evaporation technique, as previously described [32].
riefly, curcumin (5 mg) and PLGA (50 mg) were dissolved in a
ixture of ethyl acetate (1.5 mL) and methylene chloride (0.5
mL)
ith or without PEG (10 mg) at room temperature. This organic
hase was rapidly poured into 10 mL of PVA aqueous solution
0.5%, w/v) and emulsified by sonication for 5 min (35% of 500
W,
nique® Ultrasonic Mixing, Brazil), resulting in an oil-in-water
Biointerfaces 101 (2013) 353– 360
(O/W) emulsion. Next, the organic solvent was rapidly
eliminated
by evaporation under vacuum (20 min) at 37 ◦C. The particles
were
then recovered by centrifugation (19,975 × g, 30 min, 4 ◦C,
Cien-
tec CT-15000R centrifuge, Brazil) and washed twice with water
to
remove the surfactant. The nanoparticles were dispersed in the
cry-
oprotectant sucrose (5%, w/v), and the resulting nanosuspension
was cooled to −18 ◦C and freeze-dried (Terroni®, Brazil).
2.3. Particle size
The mean particle size and polydispersity index were deter-
mined by dynamic light scattering (BIC 90 plus – Brookhaven
Instruments Corp., USA). The analyses were performed at a
scat-
tering angle of 90◦ and a temperature of 25 ◦C. For each sample,
the
mean particle diameter, polydispersity and standard deviation of
ten measurements were calculated.
2.4. Drug entrapment efficiency
A Waters 2695 Alliance HPLC system (Milford, MA, USA) was
used for curcumin quantitation. The chromatographic analysis
was
performed in isocratic mode using a reverse phase C18 column
(VertiSep GES,Vertical Chromatography Co) with a 5 �m
particle
size, 4.6 mm internal diameter and 250 mm length. The mobile
phase consisted of a mixture of ethanol, acetonitrile and water
(80:10:10), pumped at a flow rate of 0.8 mL/min. The sample
injec-
tion volume was 20 �L, and the fluorescence detector was
operated
at an excitation wavelength of 365 nm and an emission
wavelength
of 512 nm.
The amount of curcumin incorporated into the nanoparticles
was determined directly after complete dissolution of nanoparti-
cles in acetonitrile. The solutions were centrifuged and
supernatant
was collected. After the appropriate dilutions in ethanol, 20 �L
of
the sample was injected into the HPLC system, and the drug
concen-
tration was obtained by comparison with a previously
constructed
analytical curve. Before injection, all of the solutions were
filtered
through a PVDF membrane filter (0.22-�m pore size,
Millipore). The
entrapment efficiency (%) was estimated by comparing the
amount
of curcumin extracted from nanoparticles with the initial
amount
used for the nanoparticles preparation.
2.5. In vitro release profile
The release of curcumin from nanoparticles was conducted by
suspending the nanoparticles (containing 1.5 mg of curcumin)
in
12 mL of phosphate saline buffer (PBS, 0.01 M, pH 7.4), and
the sus-
pension was divided in eight Eppendorf tubes. The experiments
were performed in triplicate and under sink conditions. The
tubes
were kept in a shaker at 37 ◦C at 150 rpm. At predetermined
time
intervals, the suspension was centrifuged at 19,975 × g for 15
min
to separate the released curcumin from the nanoparticles
[33,34].
The resulting precipitate in each tube was dispersed in 1.5 mL
of
phosphate saline buffer and incubated until the next sampling.
The
released curcumin present in the supernatant (1.5 mL in each
tube)
was diluted in ethanol, and 20 �L of this solution was injected
into
the HPLC to determine the amount of curcumin released at
different
time intervals.
2.6. Chromatograph system and conditions for curcumin
quantitation in plasma
The LC–MS/MS analysis was conducted in positive ion ESI
mode on a Quattro Micro API–Waters hexapole mass spectrom-
eter connected to a liquid chromatograph (Waters Alliance). The
analysis was conducted on a Phenomenex Luna C18(2) 100A
col-
umn (250 mm × 4.6 mm, 5 �m). The mobile phase consisted
of
ces B:
m
o
a
S
a
(
a
u
m
n
c
2
w
o
c
c
0
2
s
a
(
(
2
s
s
t
1
t
f
s
t
t
2
c
s
c
t
0
s
t
c
d
s
t
4
d
e
d
e
[
b
f
N.M. Khalil et al. / Colloids and Surfa
ethanol and 0.05% acetic acid solution (80:20, v/v) at a flow
rate
f 1.0 mL/min. The sheath gas and auxiliary gas were tuned to
give
n optimum response as necessary. The needle voltage was 4.5
kV.
albutamol was used as internal standard (IS) [35]. Argon was
used
s the collision gas at collision energy of 15 eV (curcumin) and
18 eV
salbutamol). The collision energy was individually tuned for
each
nalyte to obtain an optimum value. The analytes were quantified
sing selected ion reaction monitoring (SRM). The ion
transitions
/z 369.3→285.0 and m/z 240.0→147.7 were used for the
determi-
ation of curcumin and salbutamol, respectively. The
autosampler
ooler was maintained at 4 ◦C.
.7. Preparation of curcumin standards and quality control
A concentrated stock standard of curcumin and salbutamol (IS)
ere prepared by dissolving 4 mg of each compound in 20 mL
f methanol, generating a 200 �g/mL stock solution. Eight point
alibration curves were prepared by serial dilution of the cur-
umin stock solution (200 �g/mL in methanol) in the range of
.5–500 ng/mL. The calibration curve was prepared daily using
.45 mL of blank plasma with 50 �L of the appropriate working
olution, resulting in concentrations of 0.5, 10, 25, 50, 100, 200,
350
nd 500 ng/mL. Three quality controls (QC) were prepared at 1.5
low concentration), 225 (medium concentration) and 450 ng/mL
high concentration).
.8. Plasma sample preparation
To 100 �L of rat plasma sample (or a calibration standard or a
QC
ample) were added 100 �L of salbutamol (IS) and 100 �L of
0.5 M
odium hydroxide (to assist in the extraction of curcumin). The
mix-
ure was vortexed for 1 min. After the curcumin was extracted
with
300 �L of ethyl acetate (liquid–liquid extraction), followed by
agi-
ation in a shaker (10 min), it was centrifuged at 10,000 rpm at 4
◦C
or 10 min. The supernatant was evaporated using nitrogen gas in
a
ample concentrator. The obtained residue was reconstituted with
he mobile phase and vortexed for 20 s. The samples were
subjected
o LC–MS/MS analysis.
.9. Bioanalytical method validation
The specificity of the method was investigated by comparing
the
hromatogram of blank plasma with the blank plasma spiked with
tandard solutions and with the samples collected from rats after
urcumin administration.
The linearity of the bioanalytical assay was evaluated with a
otal of eight calibration standards over the concentration range
of
.5–500 ng/mL. Calibration curves were constructed by linear
least-
quares regression analysis by plotting the peak-area ratios
versus
he drug concentrations.
The limit of quantitation (LOQ) was defined as the lowest con-
entration of the analyte in the calibration curve that could be
etected with a variation of less than 15%.The intra-day preci-
ion and accuracy were determined within one day by analyzing
en replicates of the QC samples at concentrations of 1.5, 225
and
50 ng/mL of curcumin. The inter-day precision and accuracy
were
etermined on two separate occasions using replicates (n = 10) of
ach concentration used. The intra- and inter-day precision was
efined as the relative standard deviation (R.S.D.). The accuracy
was
xpressed using the following equation (1):
measured concentration
]
× 100 (1)
nominal spiked concentration
The freeze–thaw stability of the plasma samples was evaluated
y exposing QC samples at low and high concentrations to four
reeze–thaw (−20 ◦C to room temperature) cycles before sample
Biointerfaces 101 (2013) 353– 360 355
preparation. The stability of the samples in the autosampler was
evaluated by analyzing the extracted QC samples after being
placed
in the autosampler at 20 ◦C for 6 h, at which time the samples
were
analyzed. The long-term stability was verified by freezing (−20
◦C)
the QC samples for 250 days. Freshly processed standard
samples
were used to quantitate all of the QC samples. The analyses
were
performed in quintuplicate.
2.10. Pharmacokinetic study
Male adult Wistar rats with a mean body weight of 200–300 g
were fasted overnight prior to the experiments, with free access
to
water. The experimental protocol was approved by the
Institutional
Animal Ethics Committee of the Universidade Estadual de Ponta
Grossa, Brazil (Registration no. 06/2010). The rats were divided
randomly into three groups (n = 5). The formulations (curcumin
aqueous suspension, dispersion of the curcumin-loaded PLGA
nanoparticles and dispersion of the curcumin-loaded PLGA–
PEG
blend nanoparticles) were administered by oral gavage at a
single
dose of 50 mg/kg. The nanoparticles were dispersed in ultrapure
water.
Blood samples (500 �L) were withdrawn from the tail vein into
heparinized microtubes at the following times: 0.25, 0.5, 1, 1.5,
2,
4, 8, 12 and 24 h after dosing. The blood samples were
centrifuged
at 3020 × g for 10 min. The supernatant was collected,
transferred
to tightly sealed plastic tubes and stored at −20 ◦C until analysis
by LC–MS/MS. After each sampling the same volume removed
was
replaced with saline solution.
2.11. Data analysis and statistics
All of the in vitro results were expressed as the mean ± standard
deviation (S.D.) of three replicates. The in vivo results were
presented as the mean ± S.D. of five replicates. Pharmacokinetic
parameters were estimated using the model-independent method.
The terminal elimination rate constant (Ke) was estimated by
a linear regression analysis of the terminal portion of the log-
linear blood concentration–time profile of curcumin. The
terminal
elimination half-life (t1/2) was calculated from Ke using the
for-
mula t1/2 = 0.693/Ke. The maximum observed plasma
concentration
(Cmax) and the time taken to reach it (Tmax) were obtained
from
the curve plotting curcumin concentration vs. time. The area
under
each drug concentration time curve (AUC, ng/mL h) to the last
data
point was calculated by the linear trapezoidal rule and
extrapolated
to time infinity by the addition of CLast/Ke, where CLast is the
con-
centration of the last measured plasma sample. The apparent
body
clearance (Cl) was calculated using the equation Cl =
dose/AUC. The
apparent volume of distribution (Vd) was calculated by the
equation
Vd = dose/Ke AUC. Statistical analysis of the data was
performed via
one-way analysis of variance (ANOVA). The results were
considered
statistically significant if p < 0.05.
3. Results and discussion
3.1. Preparation of the curcumin-loaded PLGA and PLGA–PEG
blend nanoparticles
The nanoparticles containing curcumin were successfully
obtained by the single-emulsion solvent-evaporation method.
The
choice of a nanoencapsulation method is based on the drug
solubil-
ity, and because curcumin is hydrophobic, the method of
reducing
the size of the emulsion oil-in-water (O/W) is adequate for this
molecule. The ultrasonication was crucial to reduce the
emulsion
globules to nanometer size. Table 1 illustrates the size
characteris-
tics of the obtained nanoparticles. Both formulations, the PLGA
and
PLGA–PEG blend nanoparticles containing curcumin, presented
356 N.M. Khalil et al. / Colloids and Surfaces B: Biointerfaces
101 (2013) 353– 360
Table 1
Curcumin nanoparticle characteristics.
Polymer Particle size (nm)a Polydispersity indexa Size
distributiona Encapsulation efficiency (%)b
PLGA 161.93 ± 6.7 0.042 ± 0.01 146.2–200.7 nm (100%) 77.07
± 8.16
PLGA–PEG 152.37 ± 4.5 0.077 ± 0.01 109.9–185.1 nm (100%)
73.22 ± 9.77
Values reported as mean ± S.D.
m
o
p
n
r
l
w
e
d
d
e
3
a
T
i
t
T
t
n
l
b
o
s
n
2
n
d
c
m
F
t
a (n = 3).
b (n = 30).
onodisperse profiles and narrow size distributions. The presence
f PEG did not influence the mean particle size, but the polydis-
ersity index was superior (p < 0.05) than that obtained from
PLGA
anoparticles, while maintaining a monomodal profile.
The encapsulation efficiency was determined directly, and the
esults are presented in Table 1. The method used for
nanoencapsu-
ation resulted in significant enclosure of curcumin, and the
process
as found to be highly reproducible. The PEG did not influence
the
ncapsulation, as the values between batches were not
significantly
ifferent (p > 0.05). Indeed, the hydrophilic character of PEG
kept it
irected to aqueous phase, while the hydrophobic core of PLGA
can
ntrap the hydrophobic drugs.
.2. In vitro curcumin release profile
The in vitro release of curcumin from nanoparticles was evalu-
ted simulating physiological conditions (37 ◦C, PBS buffer pH
7.4).
he in vitro release profiles of curcumin were obtained by graph-
ng the cumulative percentage of the drug released with respect
o the amount of curcumin encapsulated as a function of the
time.
he experiment was performed over nine days. Fig. 1 illustrates
he curcumin release profiles from the PLGA and PLGA–PEG
blend
anoparticles and indicates that there was a pronounced time pro-
ongation of the drug release.
It is evident that the PEG influenced the curcumin release
ecause there was a great difference between the release profiles
f curcumin from nanoparticles of different compositions. A
bipha-
ic release pattern of curcumin was observed from the PLGA–
PEG
anoparticles, where the initial 24 h period released
approximately
1% of drug, followed by a sustained release to a total of 56.9%
over
ine days of observation. This initial burst release may be due to
rug desorption from the particle surface, and the sustained
release
an be characterized by the drug diffusion through the polymeric
atrix and subsequent diffusion/erosion of the polymeric matrix.
ig. 1. In vitro release profile of curcumin from PLGA and
PLGA–PEG blend nanopar-
icles in PBS (0.01 M, pH 7.4) at 37 ◦ C. Values reported as the
mean ± S.D. (n = 3).
Curcumin release from the PLGA nanoparticles was slower than
from the PLGA–PEG blend nanoparticles (p < 0.05), and the
release
was progressive because it did not have a biphasic profile. After
24 h, only 5.8% of the drug had been released, and in nine days,
37%
of the curcumin had been released.
In general, it can be affirmed that the drug release depends upon
the solubility, diffusion and biodegradation of the matrix mate-
rials. Thus, the drug release mechanisms can be modified by the
choice of polymer matrices. Drug release also depends upon the
loading efficiency of the drug and the size of the nanoparticles
[24].
In our case, because the particle size and curcumin loading are
sim-
ilar for the PLGA and PLGA–PEG nanoparticles, we can attest
that
the difference between the amount of drug released from the
two
nanoparticles is due to the presence of PEG, as it has a
hydrophilic
character and can enhance the water permeation and drug diffu-
sion through the polymeric matrix [36]. It is possible that
curcumin
strongly interacts with the PLGA matrix, thus retarding the
release
capability, and that the PEG can increase the wettability of the
polymeric surface and matrix, contributing to the increase in
drug
release. The results show that the PLA-PEG nanoparticles
released
more curcumin than the PLGA nanoparticles (by approximately
1.5-fold; p < 0.05) during the period analyzed.
3.3. Bioanalytical method development and validation
A LC–MS/MS method for the determination and quantitation of
curcumin in rat plasma has been developed and validated. Initial
runs were conducted with mobile phases composed of acetoni-
trile:0.2% formic acid solution (40:60, v/v), acetonitrile:1%
formic
acid solution (70:30, v/v), acetonitrile:0.1% acetic acid solution
(70:30, v/v) and acetonitrile:0.005% acetic acid solution (70:30,
v/v). In all of these combinations, the curcumin peak resulted in
tailing, and the signal was slow.
Testing several ratios of methanol and acetic acid, the combina-
tion that resulted in a sharp peak with a sufficient response area
was using methanol and 0.05% acetic acid solution (80:20, v/v)
as
the mobile phase at a flow rate of 1 mL/min. The retention
times
were approximately 1.08 min and 0.82 min for curcumin and
IS,
respectively, and the total run time was 2 min.
The specificity of the method was evaluated by comparing the
chromatograms of curcumin in plasma (standard and sample)
and
those of potentially interfering plasma components. Representa-
tive chromatograms are shown in Fig. 2, including a blank
plasma
sample (Fig. 2A), plasma containing curcumin and salbutamol
stan-
dard (Fig. 2B) and a plasma sample obtained 30 min after the
oral
administration of 50 mg/kg of curcumin-loaded PLGA
nanoparticles
(Fig. 2C). The resulting chromatograms show the assay
specificity,
as there were no endogenous plasma components eluted at the
retention time of curcumin or IS.
The method was validated over a wide concentration range, and
the results were directly obtained and extrapolated on the
calibra-
tion curve. The calibration lines were shown to be linear from
0.5
to 500 ng/mL (r2 = 0.9941). The method was sensitive, and the
LOQ
was low (0.5 ng/mL). Other LC–MS/MS methods described in
the
N.M. Khalil et al. / Colloids and Surfaces B: Biointerfaces 101
(2013) 353– 360 357
F n stan
c
l
1
a
r
ig. 2. Representative chromatograms of (A) a blank plasma
sample, (B) a curcumi
urcumin, while that the lower are indicative of the peak of the
internal standard.
iterature for curcumin determination in plasma showed a LOQ
of
0 ng/mL [16] and 2.5 ng/mL [37].
Table 2 shows a summary of intra- and inter-day precision and
ccuracy for curcumin detection in rat plasma. The intra-day
accu-
acy of curcumin for rat plasma samples was 101.72–110.54%
for
dard, and (C) a curcumin sample. The upper peak of the figure
is representative of
QC samples with a R.S.D. of less than 6.70%. The inter-day
accu-
racy of curcumin for rat plasma samples ranged from 96.34% to
107.94% for QC samples with an R.S.D. of less than 4.34%.
These
results were within the limits established by the FDA guidelines
for the validation of bioanalytical methods [38].
358 N.M. Khalil et al. / Colloids and Surfaces B: Biointerfaces
101 (2013) 353– 360
Table 2
Intra-day and inter-day precision and accuracy of curcumin in
rat plasma (n = 10).
Nominal
concentration
(ng/mL)
Measured
concentration (ng/mL)
R.S.D. (%) Accuracy (%)
Intra-daya
1.5 1.66 ± 0.04 2.33 110.54
225 224.66 ± 8.00 3.56 99.85
450 457.76 ± 30.66 6.70 101.72
Inter-dayb
1.5 1.62 ± 0.05 3.39 107.94
225 216.77 ± 9.41 4.34 96.34
450 436.59 ± 18.74 4.29 97.02
a
t
o
r
s
s
c
p
a
w
o
o
s
f
3
o
m
n
c
p
s
c
r
t
m
1
A
w
a
c
f
c
i
T
S
The analyses were performed in the same day; R.S.D. = relative
standard devia-
ion.
b The analyses were performed in two different days within one
month.
Table 3 lists the data from the stability tests. No significant loss
f curcumin (≤1.4%) was observed after storage of the plasma at
oom temperature on the bench top for at least 6 h. The plasma
amples were stable over at least four freeze/thaw cycles and
were
table at −20 ◦C for at least 250 days, with no significant loss of
urcumin (≤1.8%). These results suggested that the plasma sam-
les could be stored at −20 ◦C for long periods, could be thawed
nd refrozen and could be maintained at room temperature for 6
h
ithout compromising the integrity and accuracy of the samples.
The sensitivity of this LC–MS/MS method offered advantages
ver other LC–MS/MS methods and conventional HPLC–UV
meth-
ds applied for curcumin pharmacokinetics. Also, the excellent
pecificity and short run time analysis make this method efficient
or curcumin pharmacokinetic applications.
.4. Pharmacokinetics study
The mean curcumin plasma concentration–time profiles after
ral administrations of 50 mg/kg of curcumin in different for-
ulations, curcumin aqueous suspension, curcumin-loaded PLGA
anoparticles and curcumin-loaded PLGA–PEG blend nanoparti-
les, are expressed in Fig. 3. The Table 4 summarizes the
relevant
harmacokinetic parameters.
After the oral administration of a curcumin aqueous suspen-
ion, the drug was absorbed quickly, and a maximum plasma
oncentration (Cmax) of approximately 4.066 ± 0.564 ng/mL was
eached in 30 min. Thereafter, the curcumin plasma concentra-
ion decreased abruptly, as the drug was distributed and rapidly
etabolized, resulting in a high Ke and short t1/2, approximately
.1 h. The curcumin was detected up to 8 h after administration.
sustained release of curcumin over 24 h was observed when it
as carried by the two nanoparticles formulations. Thirty minutes
fter oral administration of the curcumin-loaded PLGA
nanoparti-
les, the mean plasma concentration was 4.57 ± 0.35 ng/mL, and
or the curcumin-loaded PLGA–PEG nanoparticles, the plasma
oncentration was 9.1 ± 0.95 ng/mL. There was a significant
ncrease (p < 0.01) in curcumin absorption from the PLGA–PEG
able 3
tability of curcumin in rat plasma.
Sample condition Curcumin nominal concentration
1.5 ng/mL
Concentration measured
(ng/mL)a
R.S.D. (%)b Acc
6 h at room temperature 1.62 ± 0.1 1.42 108
Freeze–thaw four cycles 1.63 ± 0.04 0.7 109
250 days at −20 ◦ C 1.6 ± 0.05 1.1 107
a Values reported as mean ± S.D. (n = 5).
b Relative standard deviation, calculated comparing with CQ
freshly prepared.
Fig. 3. Comparison of in vivo plasma concentration vs. time
profiles of the different
curcumin formulations. All values reported are the mean ± S.D.
(n = 5).
nanoparticles in the first 30 min compared to free curcumin and
curcumin from the PLGA nanoparticles. The curcumin
concentra-
tion increased to 11.783 ± 0.454 ng/mL, Cmax, after 2 h
(Tmax), and
to 29.778 ± 4.632 ng/mL, Cmax, after 3 h (Tmax), with the
PLGA and
PLGA–PEG nanoparticles, respectively. Compared to free
curcumin,
the Cmax of curcumin from PLGA nanoparticles and PLGA–
PEG
nanoparticles was increased 2.9- and 7.4-fold, respectively.
The
increase in Cmax indicates that the nanoparticles were effective
in increasing drug absorption, and the delayed Tmax demon-
strates an obvious sustained release of curcumin. The
distribution
and metabolism of curcumin were decreased when it was car-
ried by nanoparticles (p < 0.01). The clearance of curcumin
from
the PLGA and PLGA–PEG nanoparticles was 16.3- and 61.6-
fold
lower than that of free curcumin, respectively. The PLGA–PEG
nanoparticles and PLGA nanoparticles decreased the curcumin
volume of distribution by 11.6- and 4.6-fold compared to free
curcumin. Thus, the t1/2 of curcumin from the PLGA increased
to 4 h, and that from the PLGA–PEG nanoparticles was
increased
to 6 h, while for free curcumin the t1/2 was 1 h. There was
a significant difference in the AUC0–inf between the curcumin
aqueous suspension, the curcumin–PLGA nanoparticles and the
curcumin–PLGA–PEG nanoparticles (p < 0.01). Between the
two
nanoparticle formulations, the curcumin from PLGA–PEG pre-
sented a relative bioavailability 3.5-fold superior to that of the
curcumin from PLGA nanoparticles. Compared to the curcumin
aqueous suspension, the PLGA and PLGA–PEG blend
nanoparticles
increased the curcumin bioavailability 15.6- and 55.4-fold,
respec-
tively.
Recently, Shaikh et al. [19] demonstrated that the PLGA
nanoparticles were able to increase the curcumin bioavailability
at least 9-fold when compared to curcumin administered with an
absorption enhancer. Tsai et al. [20] developed curcumin-loaded
450 ng/mL
uracy (%) Concentration measured
(ng/mL)a
R.S.D. (%)b Accuracy (%)
.31 474.50 ± 13.36 0.47 105.44
.00 481.07 ± 18.52 0.56 106.9
.25 483.99 ± 5.6 1.8 107.55
N.M. Khalil et al. / Colloids and Surfaces B: Biointerfaces 101
(2013) 353– 360 359
Table 4
Pharmacokinetic parameters of curcumin following single oral
administration of curcumin aqueous solution, curcumin-loaded
PLGA nanoparticles and curcumin-loaded
PLGA–PEG nanoparticles, in rats (n = 5).
Pharmacokinetic parameters Formulations
Curcumin aqueous suspension Curcumin PLGA nanoparticles
Curcumin PLGA–PEG nanoparticles
Dose (mg/kg) 50 50 50
AUC0–t (h ng/mL) 8.695 ± 1.872 134.251 ± 3.446* , # 447.80 ±
64.028*
AUC0–inf (h ng/mL) 8.762 ± 1.862 137.162 ± 3.694* , #
485.941 ± 54.663*
Cmax (ng/mL) 4.066 ± 0.564 11.783 ± 0.454* , # 29.778 ±
4.632*
Tmax (h) 0.5 2* , # 3*
Ke (1/h) 0.631 ± 0.072 0.178 ± 0.021* , # 0.119 ± 0.021*
t1/2 (h) 1.109 ± 0.124 3.929 ± 0.451* , # 5.979 ± 1.126*
Vd (L/kg) 9432.536 ± 2511.617 2073.664 ± 352.612* , #
900.544 ± 225.772*
Cl (L/h/kg) 5859.700 ± 1399.927 365.191 ± 37.351* , # 103.679
± 10.903*
Values reported as mean ± S.D. (n = 5). AUC: area under the
plasma concentration–time curve; Cmax : peak concentration;
Tmax : time to reach peak concentration; Ke : constant
of elimination; t1/2 : mean half-life; Vd : apparent volume of
distribution; Cl: clearance.
P
a
m
u
a
c
h
[
e
r
s
P
p
c
P
p
l
p
P
b
c
u
d
f
e
t
t
n
t
[
p
T
r
C
t
d
t
t
i
n
n
r
w
m
t
[
[
* Significantly different of free curcumin (p < 0.01).
# Significantly different of curcumin from PLGA–PEG
nanoparticles (p < 0.01).
LGA nanoparticles. When these particles were intravenously
dministrated in rats, a significant amount of curcumin was
found
ainly in the spleen due to phagocytic cell uptake in the retic-
loendothelial system. Xie et al. [30] showed that after the oral
dministration of curcumin-loaded PLGA nanoparticles, the cur-
umin had a 5.6-fold higher relative bioavailability and had a
longer
alf-life than that of native curcumin. In a similar work, Anand
et al.
18] demonstrated that curcumin-loaded PLGA nanoparticles
have
nhanced cellular uptake, increased bioactivity in vitro and supe-
ior bioavailability in vivo relative to free curcumin. To date, no
tudy has compared the pharmacokinetics of curcumin loaded in
LGA and PLGA–PEG blend nanoparticles.
In our study, the significant difference in pharmacokinetic
arameters, mainly bioavailability and half-life, between the free
urcumin aqueous suspension and the curcumin-loaded PLGA
and
LGA–PEG nanoparticle dispersions is explained by the inherent
roperties of colloidal nanoparticles in biological media, which
pro-
ong drug release and its in vivo trajectory. The in vitro release
rofile demonstrated that curcumin is released more rapidly from
LGA–PEG nanoparticles than from PLGA nanoparticles and
could
e more quickly available in blood. It is well supported that
pharma-
okinetic parameters are altered depending upon the
nanoparticles
sed, and their surface composition plays an important role in
rug bioavailability [39,40]. PEG is frequently used for the sur-
ace modification of various polymeric nanoparticles because it
xhibits excellent biocompatibility and is able to improve the
long-
erm systemic circulation of the nanoparticles. The PEG coating
on
he surface of the polymer reduces the interactions between the
anoparticles and the enzymes of the digestive fluids and
increases
he uptake of the drug in the blood stream and lymphatic tissue
41]. This effect can explain the difference between the
curcumin
harmacokinetics from PLGA and PLGA–PEG blend
nanoparticles.
he ability of the PEG to make the coated nanoparticles invisible
to
ecognition by MPS cells gives the particles long circulation
time.
onsequently, the drug half-life and bioavailability are higher
than
hose of a drug carried in uncoated nanoparticles [27]. We
recently
emonstrated that the presence of PEG in PLA nanoparticles con-
aining the antiretroviral zidovudine was essential in promoting
he increase in drug bioavailability after intranasal
administration
n rats [42].
The increased curcumin bioavailability obtained with the
anoparticulate systems confirms the excellent abilities of the
anoparticles to modulate the physicochemical properties of
drugs,
esulting in improved pharmacokinetics profiles. Because the
poor
ater solubility and low oral bioavailability of curcumin are the
ajor drawbacks in its medicinal application, the studied
nanopar-
icles represent an important initial step in the development of
[
[
[
a medicine containing curcumin, using the nanotechnology as
a tool.
4. Conclusions
In this study, nanoparticles coated or not with PEG were suc-
cessfully prepared by the emulsion solvent-evaporation method.
Also, an analytical method for determining curcumin in plasma
was
optimized. We demonstrated that all curcumin pharmacokinetic
parameters were improved by nanoparticles, especially PLGA–
PEG
nanoparticles. The curcumin Cmax, Tmax, t1/2 and AUC were
signifi-
cantly increased by nanoparticles, while distribution and
clearance
were decreased. The PLGA–PEG nanoparticles were able to
increase
the curcumin bioavailability in 3.5-fold compared to curcumin
from PLGA nanoparticles. Compared to curcumin aqueous
suspen-
sion, PLGA and PLGA–PEG nanoparticles increased the
curcumin
bioavailability in 15.6 and 55.4-fold, respectively. These results
demonstrate the great potential of PLGA and mainly PLGA–
PEG
blend nanoparticles as carriers for the oral delivery of
curcumin.
Acknowledgments
This study was supported by Conselho Nacional de Desenvolvi-
mento Científico Tecnológico (CNPq) (577183/2008-7), Fundaç
ão
Araucária (462/2010) and FINEP (01.08.0211.00).
References
[1] A. Goel, B.A. Kunnumakkara, B.B. Aggarwal, Biochem.
Pharmacol. 75 (2008) 787.
[2] I. Brouet, H. Ohshima, Biochem. Biophys. Res. Commun.
206 (1995) 533.
[3] X. Gao, J. Kuo, H. Jiang, D. Deeb, Y. Liu, G. Divine, R.A.
Chapman, S.A. Dulchavsky,
S.C. Gautam, Biochem. Pharmacol. 68 (2004) 51.
[4] T. Masuda, K. Hidaka, A. Shinohara, T. Maekawa, Y.
Takeda, H. Yamaguchi, J.
Agric. Food Chem. 47 (1999) 71.
[5] A.P. Lakshmanan, K. Watanabe, R.A. Thandavarayan, F.R.
Sari, H. Meilei, V. Soeti-
kno, S. Arumugam, V.V. Giridharan, K. Suzuki, M. Kodama,
Free Radic. Res. 45
(2011) 788.
[6] O.A.K. Khalil, O.M.M.F. Oliveira, J.C.R. Vellosa, A.U. de
Quadros, L.M. Dalposso,
T.K. Karam, R.M. Mainardes, N.M. Khalil, Food Chem. 133
(2012) 1001.
[7] A. Mazumder, K. Raghavan, J. Weinstein, K.W. Kohn, Y.
Pommier, Biochem.
Pharmacol. 49 (1995) 1165.
[8] B.B. Aggarwal, A. Kumar, A.C. Bharti, Anticancer Res. 23
(2003) 363.
[9] B.B. Aggarwal, K.B. Harikumar, Int. J. Biochem. Cell Biol.
41 (2009) 40.
10] Z.M. Shao, Z.Z. Shen, C.H. Liu, M.R. Sartippour, V.L. Go,
D. Heber, M. Nguyen, Int.
J. Cancer 98 (2002) 234.
11] P. Anand, C. Sundaram, S. Jhurani, A.B. Kunnumakkara,
B.B. Aggarwal, Cancer
Lett. 267 (2008) 133.
12] P. Anand, A.B. Kunnumakkara, R.A. Newman, B.B.
Aggarwal, Mol. Pharmacol. 4
(2007) 807.
13] R.A. Sharma, W.P. Steward, A.J. Gescher, Adv. Exp. Med.
Biol. 595 (2007) 453.
14] A. Kunwar, A. Barik, R. Pandey, K.I. Priyadarsini,
Biochim. Biophys. Acta 1760
(2006) 1513.
3 ces B:
[
[
[
[
[
[
[
[
[
[
[
[
[
[
[
[
[
[
[
[
[
[
[
[
[
60 N.M. Khalil et al. / Colloids and Surfa
15] V.R. Yadav, S. Prasad, R. Kannappan, J. Ravindran, M.M.
Chaturvedi, L.
Vaahtera, J. Parkkinen, B.B. Aggarwal, Biochem. Pharmacol. 80
(2010)
1021.
16] V. Kakkar, S. Singh, D. Singla, S. Sahwney, S.A. Chauhan,
G. Singh, I.P. Kaur, J.
Chromatogr. B: Analyt. Technol. Biomed. Life Sci. 878 (2010)
3427.
17] W. Tiyaboonchai, W. Tungpradit, P. Plianbangchang, Int.
J. Pharm. 337 (2007)
299.
18] P. Anand, H.B. Nair, B. Sung, A.B. Kunnumakkara, V.R.
Yadav, R.R. Tekmal, B.B.
Aggarwal, Biochem. Pharmacol. 79 (2010) 330.
19] J. Shaikh, D.D. Ankola, V. Beniwal, D. Singh, M.N.V. Ravi
Kumar, Eur. J. Pharm.
Sci. 37 (2009) 223.
20] Y.M. Tsai, C.F. Chien, L.C. Lina, T.H. Tsai, Int. J. Pharm.
416 (2011) 331.
21] A. Frank, E. Pridgen, L.K. Molnar, O.C. Farokhzad, Mol.
Pharmacol. 5 (2008) 505.
22] N.M. Khalil, E. Carraro, L.F. Cótica, R.M. Mainardes,
Expert. Opin. Drug Deliv. 8
(2011) 95.
23] N.M. Khalil, R.M. Mainardes, Curr. Drug Deliv. 6 (2009)
261.
24] A. Kumari, S.K. Yadav, S.C. Yadav, Colloids Surf. B
Biointerfaces 75 (2010) 1.
25] J.C. Leroux, E. Allemann, F. De Jaeghere, E. Doelker, R.L.
Gurny, J. Control. Release
39 (1996) 339.
26] R.M. Mainardes, M.P.D. Gremião, I.L. Brunetti, L.M. da
Fonseca, N.M. Khalil, J.
Pharm. Sci. 98 (2009) 257.
27] D.E. Owens, N.A. Peppas, Int. J. Pharm. 307 (2006) 93.
[
[
[
Biointerfaces 101 (2013) 353– 360
28] S. Stolnik, L. Illum, S.S. Davis, Adv. Drug Deliv. Rev. 16
(1995) 195.
29] V.P. Torchilin, V.S. Trubetskoy, Adv. Drug Deliv. Rev. 16
(1995) 141.
30] X. Xie, Q. Tao, Y. Zou, F. Zhang, M. Guo, Y. Wang, H.
Wang, Q. Zhou, S. Yu, J. Agric.
Food Chem. 59 (2011) 9280.
31] K. Avgoustakis, Curr. Drug Deliv. 1 (2004) 321.
32] M. Li, O. Rouaud, D. Poncelet, Int. J. Pharm. 363 (2008)
26.
33] C. Mohanty, S.K. Sahoo, Biomaterials 31 (2010) 6597.
34] K.R. Das, N. Kasoju, U. Bora, Nanomed. Nanotech. Biol.
Med. 6 (2010) 153.
35] A. Liu, H. Lou, L. Zhao, P. Fan, J. Pharm. Biomed. Anal.
40 (2006) 720.
36] M.T. Peracchia, R. Gref, Y. Minamitake, A. Domb, N.
Lotan, R. Langer, J. Control.
Release 46 (1997) 223.
37] K.Y. Yang, L.C. Lin, T.Y. Tseng, S.C. Wang, T.H. Tsai, J.
Chromatogr. B: Analyt.
Technol. Biomed. Life Sci. 853 (2007) 183.
38] US Food and Drug Administration, Guidance for Industry,
Bioanalytical Method
Validation, Centre for Drug Evaluation and Research (CDER),
Rockville, 2001.
39] V. Hoffart, A. Lamprecht, P. Maincent, T. Lecompte, C.
Vigneron, N. Ubrich, J.
Control. Release 113 (2006) 38.
40] N. Ubrich, C. Schmidt, R. Bodmeier, M. Hoffman, P.
Maincent, Int. J. Pharm. 288
(2005) 169.
41] M. Tobio, A. Sánchez, A. Vila, I.I. Soriano, C. Evora, J.L.
Vila-Jato, M.J. Alonso,
Colloids Surf. B Biointerfaces 18 (2000) 315.
42] R.M. Mainardes, N.M. Khalil, M.P.D. Gremião, Int. J.
Pharm. 395 (2010) 266.
Pharmacokinetics of curcumin-loaded PLGA and PLGA–PEG
blend nanoparticles after oral administration in rats1
Introduction2 Materials and methods2.1 Materials2.2
Preparation of curcumin-loaded PLGA and PLGA–PEG blend
nanoparticles2.3 Particle size2.4 Drug entrapment efficiency2.5
In vitro release profile2.6 Chromatograph system and conditions
for curcumin quantitation in plasma2.7 Preparation of curcumin
standards and quality control2.8 Plasma sample preparation2.9
Bioanalytical method validation2.10 Pharmacokinetic study2.11
Data analysis and statistics3 Results and discussion3.1
Preparation of the curcumin-loaded PLGA and PLGA–PEG
blend nanoparticles3.2 In vitro curcumin release profile3.3
Bioanalytical method development and validation3.4
Pharmacokinetics study4
ConclusionsAcknowledgmentsReferences
Abstract: The medicinal properties of Curcumin obtained
from Curcuma longa L. cannot be utilised because of poor
bioavailability due to its rapid metabolism in the lover and
intestinal wall. In this study, the effect of combining
piperine, a known inhibitor of hepatic and intestinal
glucuronidation, was evaluated on the bioavailability of
Curcumin in rats and healthy human volunteers. When
Curcumin was given alone, the dose 2g/kg to rats,
moderate serum concentrations were achieved over a
period of 4 h. Concomitant administration of piperine
20mg/kg increased (P < 0.02), and he bioavailability was
increased by 154%. On the other hand in humans after a
dose of 2g Curcumin alone, serum levels were either
undetectable or very low. Concomitant administration of
piperine 20mg produced much higher concentrations from
0.25 to 1h post drug (P < 0.01 at 0.25 and 0.5h; P < 0.001 at
1h), the increase in bioavailability was 2000%. The study
shows that in the dosages used, piperine enhances the
serum concentrations, extent of absorption and
bioavailability of curcumin in both rats and humans with no
adverse effects.
Key words: Curcumin, piperine, pharmacokinetics, Curcuma
longa, Zingiberaceae
Introduction
Curcumin in obtained from Curcuma longa L
(Zingiberaceae), a perennial herb widely cultivated in
tropical regions of Asian. Its rhizome is extensively used for
imparting colour and flavour to food. Current traditional
Indian medicine claims the use of its powder, turmeric,
against a wide variety of diseases (1). Extensive scientific
research (2) on curcumin had demonstrated a wide
spectrum of therapeutic effects which range from anti-
inflammatory, wound healing, antispasmodic,
anticoagulant, antitumor activities (3) and recently, with
potential utility in autoimmune deficiency syndrome (4).
Pharmacokinetic properties of curcumin indicate that
following oral administration, it is poorly absorbed (3) and
only traces of the compound appear in the blood, while
most of it is excreted in the faeces (5). The transformation
of curcumin into an unidentified compound during
absorption (6) and its glucuronidation in the liver (5, 7) are
probably responsible for its low concentration in the blood.
Planta Medica 64 (1998) 353 – 356
©Georg Thieme Veriag Stuttgart – New York
Black pepper (Piper nigrum L) and long pepper (Piper
longum L) have been in use as spices from ancient time’s
thoughout the world. A major component of the Piper
species id the alkaloid piperine (l-piperoylpiprtidine), which
had been reported to enhance the bioavailability of drugs
by inhibition of glucuronidation in the liver (8) and small
intestine (9).
In view of the potential therapeutic utility of curcumin it
appeared pertinent to examine the effect of piperine, a
known hepatic and intestinal metabolic inhibitor, on the
pharmacokinetic disposition of curcumin in animals and
man, to provide a scientific rationale of assigning it a
rightful place in the pharmacologist’s armamentarium.
Materials and Methods
Animal Studies
Albina Wistar rats (n=96) of both sexes (150-200g) were
chosen for the study. They were housed in well ventilated
cages, fed on commercial rat pellets supplied by Hind Lever,
Mumbai, with tap water as libitum. They were divided into
two sex and weight matched groups (n=6/group/time cut),
one group for administration of curcumin and the other for
concomitant curcumin and piperine. Curcumin and piperine
were supplied in pure powder for by Sami Chemicals and
Extracts, Bangalore, India. Both the compounds were
administered orally to fasted rats as an aqueous
suspension. The in group that received both drugs,
curcumin was administered first followed immediately by
piperine. Control rats received water only. Curcumin was
given in a dose of 2g/kg and piperine, 20mg/kg.
Under ether anaesthesia, pre and post drug jugular vein
blood samples were collected from both groups of rats into
centrifuge tubes at the time intervals – 0, 0.25, 0.50, 0.75,
1, 2, 3, 4, 5 and 6h. The blood was allowed to clot at room
temperature for about 1h and then centrifuged at 3000rpm
for 10min. The serum was separated out carefully using
Pasteur pipettes into storage tubes and frozen at -20°C
prior to analysis.
Influence of Piperine on the Pharmacokinetics of Curcumin in
Animals and Human Volunteers
Guido Shoba₁, David Joy₁, Thangam Joseph₁, M. Majeed₂. R.
Rajendran₂, and P.S.S.R. Srinivas₂
₁Department of Pharmacology, St John’s Medical College,
Bangalore, India
₂5AMI Chemicals & Extracts (P) Ltd., Banaglore, India
Received: August 1, 1997; Revision accepted: October 18, 1997
Human volunteer studies
Ten healthy male volunteers, 20 to 26 years, weighing 50 –
75kg (mean 60 ± 1.93) participated in a randomized cross
over trial, to determine the comparative bioavailability and
pharmacokinetic profile of curcumin when given alone and
with piperine. Complete physical examination and an
electrocardiogram were done. Laboratory tests comprising
complete blood counts and haemoglobin percentage, blood
biochemistry consisting of blood urea nitrogen (BUN) serum
creatinine, total and conjugated bilirubin, alkaline
phosphatase, aspartate transaminase (ASAT), alanine
transaminase (ALAT), urine albumin, and sugar were
preformed to confirm that the subjects included in the
study were normal. The study was formally approved by
the Institutional Ethical Committee and informed consent
was obtained for all subjects.
Subjects abstained from food since 10pm of the previous
evening and reported to the laboratory at 7am.
Venepuncture was done using a 20g scalp vein set with
heparin lock and left in situ. Blood samples (5ml) were
collected (without anticoagulant) at 0, 0.25, 0.50, 0.75, 1, 2,
3, 4, 5 and 6h post drug. Blood was allowed to clot at room
temperature for 1h. Serum separation and storage until
analysis was as explained earlier. Following basal blood
sample collection 2g of pure curcumin powder (4 capsules
of 500mg curcumin each) or 2g of pure curcumin powder
combined with 20mg of pure piperine powder (4 capsules
of 500mg curcumin + 5mg piperine each; identical capsules
prepared by Sami Chemicals and Extracts, Bangalore, India)
was given with 150ml of water. Blood sampling after
curcumin per se and curcumin with piperine was done two
occasions, separated by a two week wash out period on the
same volunteers. The following precautions were taken
during the trial; subjects refrained from smoking,
consuming alcohol, or beverages, and from taking drugs of
any kind 24h prior to and during the trial. Standard meals
were given to all the participants on the day of the test.
Analytical methods
Estimation of curcumin was done by reverse phase high
pressure liquid chromatography (HPLC) using modification
of the method described by Tannesen et al. (10). The
modification was done by Sami Chemicals and Extracts,
Bangalore, India, and is detailed below, the mobile phase
used was ethanol : methanol (60 : 40) instead of only
ethanol and the flow rate was changed from 1.2ml/min to
1ml/min. HPLC grade methanol and low actinic glassware
protected from light were used for the entire procedure.
Extraction and preparation of standard solution
Curcumin (25mg) was dissolved and diluted to 25ml with
methanol in a volumetric flask; 0.1ml (100 µm) of this was
transferred to a volumetric flask and diluted with methanol
up to 10ml making a 10ppm solution; 0.1ml of this 10ppm
solution was transferred to another 10ml volumetric flask
and the volume make up with methanol making a 0.1ppm
solution.
Extraction of curcumin from serum and preparation of
sample
Serum samples stored t -20°C were equilibrated to room
temperature before analysis. A portion of 1ml was
transferred into a 10ml volumetric flask and about 5ml of
methanol added. The mixture was shaken thoroughly and
heated at 80°C on a water bath for half an hour. After
cooling to room temperature, methanol was added to make
up the volume to 10ml and mixed well. The turbid solution
was transferred into a 15ml centrifuge tube and centrifuged
at 4000 RPM for 10 minutes. The supernatant was collected
by means of a 25ml syringe and 10cm needle (Luer lock)
and the clear solution filtered through a 0.45µm, 13mm
Millipore membrane filter, into a narrow end test tube.
20µl of the solution were injected into the chomatograph
for carry out the HPLC analysis.
Samples were read by UV absorbance of 254nM. The
recovery rate experiments were carried out by adding a
known amount of standard curcumin to the serum and the
added curcumin extracted as per procedure and quantified.
The recovery rate of curcumin from serum ranged from 87
– 89.9%. The minimum level of detection of curcumin was
0.001µg/ml.
Calculation
Content of curcumin in µg/ml in the test sample
Standard Reading x standard concentration
Standard Reading x standard concentration
Treatment of pharmacokinetic data
For calculation of pharmacokinetic parameters (PK), curve
fitting was carried out by a model independent method
with non-linear least-square regression analysis using a
computer designed programme “PHARMKIT”. This
programme uses an algorithm call “SIMPLEX” for calculating
non-linear least-squares. The various PK parameters
calculated were: absorption half-life (t½(a)), elimination half
live (t½el), volume of distribution (Vd); and clearance (CI).
Areas under the concentration time curve (AUCo-m) was
calculated using the trapezoidal method. Maximum
concentration (Cmax) and time to max (Tmax) are observed
values. Relative bioavailability (F) was calculated using
formula:
AUC Curcumin + piperine
AUC Curcumin
Statistical analysis
Serum concentration time curves and the PK parameter
from animal data were analysed using the Students ‘t’ test
while the paired ‘t’ test was used for comparing serum
concentration curves in humans. PK parameters of
curcumin when given alone in humans could not be
calculated as curcumin could not be detected in most of the
samples.
Planta Med. 64 (1998) Guido Shoba, David Joy, Thangam
Joseph, M. Majeed. R. Rajendran, and P.S.S.R. Srinivas
=
F = x 100
Results
Animal studies
Curcumin alone at 2g/kg or when combined with piperine,
20mg/kg, was well tolerated by the rats as they showed no
untoward effects for 48h. Yellow coloured faecal pellets
appeared at 30h post drug and continued up to 48h.
Perusal of Figure 1 indicates that when curcumin was given
alone, peak serum concentrations of 1.00 ≠ 0.26 µg/ml
were attained rapidly within 0.75h and plateaued till 1h.
Thereafter, the levels declined gradually reaching zero at
5h. The plasma concentration time curve of curcumin in
combination with piperine followed a similar pattern from 0
to 0.75h and 3 to 5h. However piperine produced higher
serum concentrations of curcumin at 1 and 2 h (1.55 ± 0.21
and 1.50 ± 0.25 µg/ml) respectively, being significantly
higher (P <0.02) at 2h. Thus piperine significantly enhanced
the serum concentration of curcumin, albeit for a limited
duration (although serum samples were collected up to 6h,
values are depicted till 5h only, since the 6h value was also
‘0’ in all animals).
Table 1 shows the values (mean ± SEM) of the
pharmacokinetic parameters if curcumin per se and when
combined with piperine. Cmax was increased from 1.35 ±
0.23 to 1.80 ± 0.16 µg/ml, but was not statistically
significant, while Tmax was significantly increased from 0.83
± 0.05 to 1.29 ± 0.23h (P <0.02). The t½(el) significantly
decreased from 1.70 ± 0.58 to 1.05 ± 0.18h (P<0.002).
Though t½(a) increased from 0.31 ± 0.07 to 0.47 ± 0.03h and
AUC increased from 2.36 ± 0.28 to 3.64 ± 0.31 µg/h/ml,
these increases were not statistically significant. CI
significantly decreased from 713.00 ± 12.00 to 495.00 ±
37.00:/h (P<0.02) but the decrease in the Vd from 1366.00
±248.70 to 782.60 ± 193.90L/kg was not significant. The
relative bioavailability of curcumin when combined with
Piperine is 154%.
Human volunteer studies
Curcumin alone or when combined with piperine was well
tolerated by all the subjects and there were no adverse or
untoward reactions; 2 subjects dropped out of the study for
non-medical reasons. Therefore all calculations presented
here are based on the data obtained from 8 subjects. In
figure 2 is shown the serum concentration if curcumin per
se and when given with piperine. Although serum samples
were collected up to 6h, we have depicted values till 3h,
since the 4, 5 and 6h values were also ‘0’ in all subjects.
Serum levels’ of curcumin when given alone were either
very low or undetectable at mot time points in most
subjects, explaining he almost flat serum concentration
curve (Fig. 2). However, when piperine was added the
serum concentrations of curcumin were significantly
increased at the time points up to 0.75h; P <0.01
Planta Med. 64 (1998) Guido Shoba, David Joy, Thangam
Joseph, M. Majeed. R. Rajendran, and P.S.S.R. Srinivas
Fig. 1 Serum concentrations µg/ml (mean ± SEM) of
curcumin 20g/kg oral alone and with piperine 20mg/kg in
rats (n= 6/group/time cut). Significance as compared to
curcumin alone; *P <0.02.
Fig. 2 Serum concentrations µg/ml (mean ± SEM) of
curcumin 2g oral alone and with piperine 20mg in humans
(n= 8). Significance as compared to curcumin alone;
*P <0.01 **P <0.001.
at 0.25h and 0.5h; P <0.001 and at 0.75h. Subsequently
there was a rapid decline up to 1h and thereafter a gradual
decline to zero by 3h.
In Table 2 are depicted the PL parameters (Mean ± SEM) of
curcumin when given alone and with piperine. Cmax
(observed values) when curcumin was given alone was only
0.006 ± 0.005 µg/ml at 1h whereas when piperine was
added the Cmax (observed value) was increased to 0.18 ±
0.16 µg/ml and was attained earlier, i.e. at 0.75h. Vd and CI
could not be calculated with curcumin alone as serum
levels were not detected at most time points in most
subjects. The mean AUC(0-tn) however, was calculated using
the trapezoidal method and was found to be 0.004 µg/ml,
the relative bioavailability of curcumin when given with
piperine was therefore 2000%.
Discussion
The results obtained in the study demonstrate that piperine
enhances the oral bioavailability of curcumin in both rats
and humans at does that we devoid of adverse side effects.
However, certain differences between rats and human with
respect to curcumin were evident. Curcumin per se attained
overall moderate serum concentrations over a 4h period in
rats with peak levels occurring between 0.75h to 1h. On the
other hand, in humans when curcumin was given alone only
negligible serum concentrations of curcumin were
detectable the serum concentration-time curve being
almost flat. This difference may be due to high oral dose
employed in the rat (2g/kg), whereas the human does was
about 60 times less, approximately 33mg/kg. Curcumin
serum concentrations reached zero at 5h in rats and 3h in
humans. Further in rats with the addition of piperine,
curcumin achieved high concentrations than in humans
albeit for a short period, took s longer time to peak and
declined slowly. Whereas in humans Tmax was attained
earlier and then declined rapidly. This rapidity in decline is
more apparent probably because of the high levels of
curcumin achieved with piperine as compared to curcumin
alone. There was an increase in the AUC though not
significant as an increase in bioavailability of curcumin by
about one and a half times as compared to curcumin given
alone in both rats and humans. In rats when piperine was
added to curcumin both Vd and CI decreased which may
have also contributed to the higher concentration, such a
comparison was not possible in humans for season
explained earlier. Our findings concerning absorption f
curcumin in rats are in agreement with data obtained by
Wahlstrom and Belnnow (11), who sowed that when
Sprague Dawley rats were given curcumin 1g/kg p.o.,
measurement of blood plasma levels and biliary excretion
indicated some absorption from the gut with no apparent
toxic effect unto 5g/kg p.o. Likewise, Khanna et al. (12);
found that after curcumin, 100mg/kg p.o., 74% was
absorbed from the gastrointestinal tract within the first 5h,
while complete elimination occurred within 48h. Our
results are, however, in conflict with studies by
Ravindranath and Chandrasekhara (6), who could not
detect curcumin 400mg/kg p.o. They, however, did report
60% absorption of curcumin as determined by the amount
excreted in the faeces.
There is evidence that piperine is a potent inhibitor of drug
metabolism, and glucuronidation altering the disposition
and bioavailability of a large number of drugs (8). Further
piperine at 20mg in humans has also been shown to
produce earlier Tmax higher Cmax and AUC of drugs like
propranolol and theophylline (13). This property of piperine
suggests that it may be involved in inhibiting the
metabolism of curcumin and enhancing bioavailability.
In conclusion, the study shows that piperine enhances the
serum concentration and bioavailability of curcumin in rats
and man probably due to increased absorption and reduced
metabolism.
References
1. Kuhup, P.N.V. (1977) Handbook of Medicinal Plants, Vol. 1.
Central Council for Research in Indians Medicine and
Homeopathy (CCRIMH), New Delhi.
2. Srimal, R.C., Dhawan, R.N. (1985) in: Development of
Unuani
drugs from herbal sources and the role of elements in their
mechanism of action. Hamdard National Foundation
Monograph. New Delhi.
3. Ammon, H.R.T., Wahi, M.A. (1991) Planta Med. 57, 1-7.
4. James, J. S. (1993) AIDS Treatment News 176, 1-3.
5. Ravindranath, V., Chandrasekhara, N. (1980) Toxicology
(Ireland) 16. 259-260.
6. Ravindranath, V., Chandrasekhara, N. (1981) Toxicology 20.
251-257.
7. Holder, G.M., Plummer, J.L., Ryan, A.J. (1978) Xenobiotica
8.
761-768.
8. Atal, C.K., Dubey, R.K., Singh, J.J. (1985) Pharmacol. Exp.
Ther.
232. 258-262.
9. Singh, J., Dubey, R.K., Atal, C.K. (1985) J. Pharmacol. Exp.
Ther
236. 488-493.
10. Tonnesen, H.H., Karlsen, J. (1993) J. Chromatography 259.
367-
369.
11. Wahlstrom, B., Blennow, G. (1978) Acta Pharmacol.
Toxxicol.
43. 86-92.
12. Khanna, M., Singh, S., Sarin, J.P.S. (1981) Indian Drugs 19.
31.
13. Bano, G., Raina, R.K., Zutshi, U., Bedi, K.L., Johri, R.K.,
Sharma,
S.C. (1991) Eur. J. Clin. Pharmacol. 14. 615-617.
Planta Med. 64 (1998) Guido Shoba, David Joy, Thangam
Joseph, M. Majeed. R. Rajendran, and P.S.S.R. Srinivas
Dr Guido Shoba
Department of Pharmacology
St. John’s Medical College
Bangalore 560 034
India
PaNADaARRAAKCLBN1.docx

More Related Content

Similar to PaNADaARRAAKCLBN1.docx

Ukicrs poster 2015
Ukicrs poster 2015Ukicrs poster 2015
Ukicrs poster 2015Nashwa Osman
 
Formulation and evaluation of Muco adhesive Buccal Tablets of Rampril
Formulation and evaluation of Muco adhesive Buccal Tablets of RamprilFormulation and evaluation of Muco adhesive Buccal Tablets of Rampril
Formulation and evaluation of Muco adhesive Buccal Tablets of Ramprildoddaapurupa
 
Healing Effects of Hydroalcoholic Extract of Guava (Psidium guajava) Leaf on ...
Healing Effects of Hydroalcoholic Extract of Guava (Psidium guajava) Leaf on ...Healing Effects of Hydroalcoholic Extract of Guava (Psidium guajava) Leaf on ...
Healing Effects of Hydroalcoholic Extract of Guava (Psidium guajava) Leaf on ...Dr. Anuj S Parihar
 
Design of gastroretentive bilayer floating films of propranolol hydrochloride...
Design of gastroretentive bilayer floating films of propranolol hydrochloride...Design of gastroretentive bilayer floating films of propranolol hydrochloride...
Design of gastroretentive bilayer floating films of propranolol hydrochloride...Namdeo Shinde
 
Combined effects of PEGylation and particle size on uptake of PLGA particles ...
Combined effects of PEGylation and particle size on uptake of PLGA particles ...Combined effects of PEGylation and particle size on uptake of PLGA particles ...
Combined effects of PEGylation and particle size on uptake of PLGA particles ...Nanomedicine Journal (NMJ)
 
In vitro and in vivo evaluation of positively charged liposaccharide derivati...
In vitro and in vivo evaluation of positively charged liposaccharide derivati...In vitro and in vivo evaluation of positively charged liposaccharide derivati...
In vitro and in vivo evaluation of positively charged liposaccharide derivati...Adel Abdelrahim, PhD
 
Oral drug delivery proteins
Oral drug delivery proteinsOral drug delivery proteins
Oral drug delivery proteinsswati gadekar
 
ppt on published research paper
ppt on published research paperppt on published research paper
ppt on published research paperNITIN KANWALE
 
Evaluating the Effects of Different Molecular Weights of Polymers in Stabiliz...
Evaluating the Effects of Different Molecular Weights of Polymers in Stabiliz...Evaluating the Effects of Different Molecular Weights of Polymers in Stabiliz...
Evaluating the Effects of Different Molecular Weights of Polymers in Stabiliz...Smruti Chaudhari, Ph.D.
 
PLGA (poly lactic co glycolic acid )
PLGA (poly lactic co glycolic acid ) PLGA (poly lactic co glycolic acid )
PLGA (poly lactic co glycolic acid ) MaryamHesham Mahmoud
 
Intranasal delivery of drug loaded thiolated co-polymeric microparticles for...
Intranasal delivery of drug loaded thiolated co-polymeric microparticles for...Intranasal delivery of drug loaded thiolated co-polymeric microparticles for...
Intranasal delivery of drug loaded thiolated co-polymeric microparticles for...Gaurav Patil
 
Formulation and characterization of epigallocatechin gallate nanoparticles
Formulation and characterization of epigallocatechin gallate nanoparticlesFormulation and characterization of epigallocatechin gallate nanoparticles
Formulation and characterization of epigallocatechin gallate nanoparticlesRamkumar Ponnuraj
 
Comparative study on the effect of hydrophilic and hydrophobic polymers on th...
Comparative study on the effect of hydrophilic and hydrophobic polymers on th...Comparative study on the effect of hydrophilic and hydrophobic polymers on th...
Comparative study on the effect of hydrophilic and hydrophobic polymers on th...pharmaindexing
 
ppt on published research paper
ppt on published research paperppt on published research paper
ppt on published research paperNITIN KANWALE
 
DESIGN AND ASSESSMENT OF COLON SPECIFIC DRUG DELIVERY OF CELECOXIB USING PU...
DESIGN AND ASSESSMENT OF COLON SPECIFIC DRUG  DELIVERY OF CELECOXIB USING  PU...DESIGN AND ASSESSMENT OF COLON SPECIFIC DRUG  DELIVERY OF CELECOXIB USING  PU...
DESIGN AND ASSESSMENT OF COLON SPECIFIC DRUG DELIVERY OF CELECOXIB USING PU...Lakshmi
 
2011 enteric microdphere formultions of papain
2011 enteric microdphere formultions of papain2011 enteric microdphere formultions of papain
2011 enteric microdphere formultions of papainRocioChong
 

Similar to PaNADaARRAAKCLBN1.docx (20)

Ukicrs poster 2015
Ukicrs poster 2015Ukicrs poster 2015
Ukicrs poster 2015
 
Formulation and evaluation of Muco adhesive Buccal Tablets of Rampril
Formulation and evaluation of Muco adhesive Buccal Tablets of RamprilFormulation and evaluation of Muco adhesive Buccal Tablets of Rampril
Formulation and evaluation of Muco adhesive Buccal Tablets of Rampril
 
Healing Effects of Hydroalcoholic Extract of Guava (Psidium guajava) Leaf on ...
Healing Effects of Hydroalcoholic Extract of Guava (Psidium guajava) Leaf on ...Healing Effects of Hydroalcoholic Extract of Guava (Psidium guajava) Leaf on ...
Healing Effects of Hydroalcoholic Extract of Guava (Psidium guajava) Leaf on ...
 
Design of gastroretentive bilayer floating films of propranolol hydrochloride...
Design of gastroretentive bilayer floating films of propranolol hydrochloride...Design of gastroretentive bilayer floating films of propranolol hydrochloride...
Design of gastroretentive bilayer floating films of propranolol hydrochloride...
 
H045047053
H045047053H045047053
H045047053
 
Combined effects of PEGylation and particle size on uptake of PLGA particles ...
Combined effects of PEGylation and particle size on uptake of PLGA particles ...Combined effects of PEGylation and particle size on uptake of PLGA particles ...
Combined effects of PEGylation and particle size on uptake of PLGA particles ...
 
Bioactive peptides
Bioactive peptidesBioactive peptides
Bioactive peptides
 
In vitro and in vivo evaluation of positively charged liposaccharide derivati...
In vitro and in vivo evaluation of positively charged liposaccharide derivati...In vitro and in vivo evaluation of positively charged liposaccharide derivati...
In vitro and in vivo evaluation of positively charged liposaccharide derivati...
 
Oral drug delivery proteins
Oral drug delivery proteinsOral drug delivery proteins
Oral drug delivery proteins
 
ppt on published research paper
ppt on published research paperppt on published research paper
ppt on published research paper
 
Pullulan
PullulanPullulan
Pullulan
 
Evaluating the Effects of Different Molecular Weights of Polymers in Stabiliz...
Evaluating the Effects of Different Molecular Weights of Polymers in Stabiliz...Evaluating the Effects of Different Molecular Weights of Polymers in Stabiliz...
Evaluating the Effects of Different Molecular Weights of Polymers in Stabiliz...
 
PLGA (poly lactic co glycolic acid )
PLGA (poly lactic co glycolic acid ) PLGA (poly lactic co glycolic acid )
PLGA (poly lactic co glycolic acid )
 
Intranasal delivery of drug loaded thiolated co-polymeric microparticles for...
Intranasal delivery of drug loaded thiolated co-polymeric microparticles for...Intranasal delivery of drug loaded thiolated co-polymeric microparticles for...
Intranasal delivery of drug loaded thiolated co-polymeric microparticles for...
 
Formulation and characterization of epigallocatechin gallate nanoparticles
Formulation and characterization of epigallocatechin gallate nanoparticlesFormulation and characterization of epigallocatechin gallate nanoparticles
Formulation and characterization of epigallocatechin gallate nanoparticles
 
Comparative study on the effect of hydrophilic and hydrophobic polymers on th...
Comparative study on the effect of hydrophilic and hydrophobic polymers on th...Comparative study on the effect of hydrophilic and hydrophobic polymers on th...
Comparative study on the effect of hydrophilic and hydrophobic polymers on th...
 
Polycarbonate_2013
Polycarbonate_2013Polycarbonate_2013
Polycarbonate_2013
 
ppt on published research paper
ppt on published research paperppt on published research paper
ppt on published research paper
 
DESIGN AND ASSESSMENT OF COLON SPECIFIC DRUG DELIVERY OF CELECOXIB USING PU...
DESIGN AND ASSESSMENT OF COLON SPECIFIC DRUG  DELIVERY OF CELECOXIB USING  PU...DESIGN AND ASSESSMENT OF COLON SPECIFIC DRUG  DELIVERY OF CELECOXIB USING  PU...
DESIGN AND ASSESSMENT OF COLON SPECIFIC DRUG DELIVERY OF CELECOXIB USING PU...
 
2011 enteric microdphere formultions of papain
2011 enteric microdphere formultions of papain2011 enteric microdphere formultions of papain
2011 enteric microdphere formultions of papain
 

More from bunyansaturnina

Your tasks will be to answer questions based on the indicators that .docx
Your tasks will be to answer questions based on the indicators that .docxYour tasks will be to answer questions based on the indicators that .docx
Your tasks will be to answer questions based on the indicators that .docxbunyansaturnina
 
Your taskYou must identify a specific, local problem (eithe.docx
Your taskYou must identify a specific, local problem (eithe.docxYour taskYou must identify a specific, local problem (eithe.docx
Your taskYou must identify a specific, local problem (eithe.docxbunyansaturnina
 
Your taskis to analyze and evaluate how various types of medi.docx
Your taskis to analyze and evaluate how various types of medi.docxYour taskis to analyze and evaluate how various types of medi.docx
Your taskis to analyze and evaluate how various types of medi.docxbunyansaturnina
 
Your task this week is to check the internet and the Common Vulner.docx
Your task this week is to check the internet and the Common Vulner.docxYour task this week is to check the internet and the Common Vulner.docx
Your task this week is to check the internet and the Common Vulner.docxbunyansaturnina
 
Your task is to take unit I Will Survive Ecosystems and Adaptations.docx
Your task is to take unit I Will Survive Ecosystems and Adaptations.docxYour task is to take unit I Will Survive Ecosystems and Adaptations.docx
Your task is to take unit I Will Survive Ecosystems and Adaptations.docxbunyansaturnina
 
Your task is to perform and document encryption of Thunderbird Email.docx
Your task is to perform and document encryption of Thunderbird Email.docxYour task is to perform and document encryption of Thunderbird Email.docx
Your task is to perform and document encryption of Thunderbird Email.docxbunyansaturnina
 
Your task is to explain the process of the juvenile justice system a.docx
Your task is to explain the process of the juvenile justice system a.docxYour task is to explain the process of the juvenile justice system a.docx
Your task is to explain the process of the juvenile justice system a.docxbunyansaturnina
 
Your task is to create a journalistic profile that focuses on a .docx
Your task is to create a journalistic profile that focuses on a .docxYour task is to create a journalistic profile that focuses on a .docx
Your task is to create a journalistic profile that focuses on a .docxbunyansaturnina
 
Your task is to evaluate the available evidence on the social, emoti.docx
Your task is to evaluate the available evidence on the social, emoti.docxYour task is to evaluate the available evidence on the social, emoti.docx
Your task is to evaluate the available evidence on the social, emoti.docxbunyansaturnina
 
Your task is to conduct research on the ways that universities p.docx
Your task is to conduct research on the ways that universities p.docxYour task is to conduct research on the ways that universities p.docx
Your task is to conduct research on the ways that universities p.docxbunyansaturnina
 
Your task is to compare and contrast two artworks  given Below.docx
Your task is to compare and contrast two artworks  given Below.docxYour task is to compare and contrast two artworks  given Below.docx
Your task is to compare and contrast two artworks  given Below.docxbunyansaturnina
 
Your task is to create a personal essay that focuses on your per.docx
Your task is to create a personal essay that focuses on your per.docxYour task is to create a personal essay that focuses on your per.docx
Your task is to create a personal essay that focuses on your per.docxbunyansaturnina
 
Your Task is to Carry out an independent research study (.docx
Your Task is to Carry out an independent research study (.docxYour Task is to Carry out an independent research study (.docx
Your Task is to Carry out an independent research study (.docxbunyansaturnina
 
Your Research Project is due this week. It must consist of1. 5 .docx
Your Research Project is due this week. It must consist of1. 5 .docxYour Research Project is due this week. It must consist of1. 5 .docx
Your Research Project is due this week. It must consist of1. 5 .docxbunyansaturnina
 
Your supervisor wants the staff to understand the importance of.docx
Your supervisor wants the staff to understand the importance of.docxYour supervisor wants the staff to understand the importance of.docx
Your supervisor wants the staff to understand the importance of.docxbunyansaturnina
 
Your supervisor has asked you to create a new entity-relationship di.docx
Your supervisor has asked you to create a new entity-relationship di.docxYour supervisor has asked you to create a new entity-relationship di.docx
Your supervisor has asked you to create a new entity-relationship di.docxbunyansaturnina
 
Your supervisor asks you to lead a team of paralegals in the office .docx
Your supervisor asks you to lead a team of paralegals in the office .docxYour supervisor asks you to lead a team of paralegals in the office .docx
Your supervisor asks you to lead a team of paralegals in the office .docxbunyansaturnina
 
Your research paper final must be written using APA style and incl.docx
Your research paper final must be written using APA style and incl.docxYour research paper final must be written using APA style and incl.docx
Your research paper final must be written using APA style and incl.docxbunyansaturnina
 
Your submission should be a PowerPoint slide presentation with 1.docx
Your submission should be a PowerPoint slide presentation with 1.docxYour submission should be a PowerPoint slide presentation with 1.docx
Your submission should be a PowerPoint slide presentation with 1.docxbunyansaturnina
 
your research must includeExecutive summaryAbstractP.docx
your research must includeExecutive summaryAbstractP.docxyour research must includeExecutive summaryAbstractP.docx
your research must includeExecutive summaryAbstractP.docxbunyansaturnina
 

More from bunyansaturnina (20)

Your tasks will be to answer questions based on the indicators that .docx
Your tasks will be to answer questions based on the indicators that .docxYour tasks will be to answer questions based on the indicators that .docx
Your tasks will be to answer questions based on the indicators that .docx
 
Your taskYou must identify a specific, local problem (eithe.docx
Your taskYou must identify a specific, local problem (eithe.docxYour taskYou must identify a specific, local problem (eithe.docx
Your taskYou must identify a specific, local problem (eithe.docx
 
Your taskis to analyze and evaluate how various types of medi.docx
Your taskis to analyze and evaluate how various types of medi.docxYour taskis to analyze and evaluate how various types of medi.docx
Your taskis to analyze and evaluate how various types of medi.docx
 
Your task this week is to check the internet and the Common Vulner.docx
Your task this week is to check the internet and the Common Vulner.docxYour task this week is to check the internet and the Common Vulner.docx
Your task this week is to check the internet and the Common Vulner.docx
 
Your task is to take unit I Will Survive Ecosystems and Adaptations.docx
Your task is to take unit I Will Survive Ecosystems and Adaptations.docxYour task is to take unit I Will Survive Ecosystems and Adaptations.docx
Your task is to take unit I Will Survive Ecosystems and Adaptations.docx
 
Your task is to perform and document encryption of Thunderbird Email.docx
Your task is to perform and document encryption of Thunderbird Email.docxYour task is to perform and document encryption of Thunderbird Email.docx
Your task is to perform and document encryption of Thunderbird Email.docx
 
Your task is to explain the process of the juvenile justice system a.docx
Your task is to explain the process of the juvenile justice system a.docxYour task is to explain the process of the juvenile justice system a.docx
Your task is to explain the process of the juvenile justice system a.docx
 
Your task is to create a journalistic profile that focuses on a .docx
Your task is to create a journalistic profile that focuses on a .docxYour task is to create a journalistic profile that focuses on a .docx
Your task is to create a journalistic profile that focuses on a .docx
 
Your task is to evaluate the available evidence on the social, emoti.docx
Your task is to evaluate the available evidence on the social, emoti.docxYour task is to evaluate the available evidence on the social, emoti.docx
Your task is to evaluate the available evidence on the social, emoti.docx
 
Your task is to conduct research on the ways that universities p.docx
Your task is to conduct research on the ways that universities p.docxYour task is to conduct research on the ways that universities p.docx
Your task is to conduct research on the ways that universities p.docx
 
Your task is to compare and contrast two artworks  given Below.docx
Your task is to compare and contrast two artworks  given Below.docxYour task is to compare and contrast two artworks  given Below.docx
Your task is to compare and contrast two artworks  given Below.docx
 
Your task is to create a personal essay that focuses on your per.docx
Your task is to create a personal essay that focuses on your per.docxYour task is to create a personal essay that focuses on your per.docx
Your task is to create a personal essay that focuses on your per.docx
 
Your Task is to Carry out an independent research study (.docx
Your Task is to Carry out an independent research study (.docxYour Task is to Carry out an independent research study (.docx
Your Task is to Carry out an independent research study (.docx
 
Your Research Project is due this week. It must consist of1. 5 .docx
Your Research Project is due this week. It must consist of1. 5 .docxYour Research Project is due this week. It must consist of1. 5 .docx
Your Research Project is due this week. It must consist of1. 5 .docx
 
Your supervisor wants the staff to understand the importance of.docx
Your supervisor wants the staff to understand the importance of.docxYour supervisor wants the staff to understand the importance of.docx
Your supervisor wants the staff to understand the importance of.docx
 
Your supervisor has asked you to create a new entity-relationship di.docx
Your supervisor has asked you to create a new entity-relationship di.docxYour supervisor has asked you to create a new entity-relationship di.docx
Your supervisor has asked you to create a new entity-relationship di.docx
 
Your supervisor asks you to lead a team of paralegals in the office .docx
Your supervisor asks you to lead a team of paralegals in the office .docxYour supervisor asks you to lead a team of paralegals in the office .docx
Your supervisor asks you to lead a team of paralegals in the office .docx
 
Your research paper final must be written using APA style and incl.docx
Your research paper final must be written using APA style and incl.docxYour research paper final must be written using APA style and incl.docx
Your research paper final must be written using APA style and incl.docx
 
Your submission should be a PowerPoint slide presentation with 1.docx
Your submission should be a PowerPoint slide presentation with 1.docxYour submission should be a PowerPoint slide presentation with 1.docx
Your submission should be a PowerPoint slide presentation with 1.docx
 
your research must includeExecutive summaryAbstractP.docx
your research must includeExecutive summaryAbstractP.docxyour research must includeExecutive summaryAbstractP.docx
your research must includeExecutive summaryAbstractP.docx
 

Recently uploaded

Employee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxEmployee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxNirmalaLoungPoorunde1
 
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdf
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdfFraming an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdf
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdfUjwalaBharambe
 
CELL CYCLE Division Science 8 quarter IV.pptx
CELL CYCLE Division Science 8 quarter IV.pptxCELL CYCLE Division Science 8 quarter IV.pptx
CELL CYCLE Division Science 8 quarter IV.pptxJiesonDelaCerna
 
Like-prefer-love -hate+verb+ing & silent letters & citizenship text.pdf
Like-prefer-love -hate+verb+ing & silent letters & citizenship text.pdfLike-prefer-love -hate+verb+ing & silent letters & citizenship text.pdf
Like-prefer-love -hate+verb+ing & silent letters & citizenship text.pdfMr Bounab Samir
 
Difference Between Search & Browse Methods in Odoo 17
Difference Between Search & Browse Methods in Odoo 17Difference Between Search & Browse Methods in Odoo 17
Difference Between Search & Browse Methods in Odoo 17Celine George
 
How to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptxHow to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptxmanuelaromero2013
 
Capitol Tech U Doctoral Presentation - April 2024.pptx
Capitol Tech U Doctoral Presentation - April 2024.pptxCapitol Tech U Doctoral Presentation - April 2024.pptx
Capitol Tech U Doctoral Presentation - April 2024.pptxCapitolTechU
 
MARGINALIZATION (Different learners in Marginalized Group
MARGINALIZATION (Different learners in Marginalized GroupMARGINALIZATION (Different learners in Marginalized Group
MARGINALIZATION (Different learners in Marginalized GroupJonathanParaisoCruz
 
ENGLISH 7_Q4_LESSON 2_ Employing a Variety of Strategies for Effective Interp...
ENGLISH 7_Q4_LESSON 2_ Employing a Variety of Strategies for Effective Interp...ENGLISH 7_Q4_LESSON 2_ Employing a Variety of Strategies for Effective Interp...
ENGLISH 7_Q4_LESSON 2_ Employing a Variety of Strategies for Effective Interp...JhezDiaz1
 
What is Model Inheritance in Odoo 17 ERP
What is Model Inheritance in Odoo 17 ERPWhat is Model Inheritance in Odoo 17 ERP
What is Model Inheritance in Odoo 17 ERPCeline George
 
Types of Journalistic Writing Grade 8.pptx
Types of Journalistic Writing Grade 8.pptxTypes of Journalistic Writing Grade 8.pptx
Types of Journalistic Writing Grade 8.pptxEyham Joco
 
Blooming Together_ Growing a Community Garden Worksheet.docx
Blooming Together_ Growing a Community Garden Worksheet.docxBlooming Together_ Growing a Community Garden Worksheet.docx
Blooming Together_ Growing a Community Garden Worksheet.docxUnboundStockton
 
DATA STRUCTURE AND ALGORITHM for beginners
DATA STRUCTURE AND ALGORITHM for beginnersDATA STRUCTURE AND ALGORITHM for beginners
DATA STRUCTURE AND ALGORITHM for beginnersSabitha Banu
 
Solving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptxSolving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptxOH TEIK BIN
 
AmericanHighSchoolsprezentacijaoskolama.
AmericanHighSchoolsprezentacijaoskolama.AmericanHighSchoolsprezentacijaoskolama.
AmericanHighSchoolsprezentacijaoskolama.arsicmarija21
 
ECONOMIC CONTEXT - PAPER 1 Q3: NEWSPAPERS.pptx
ECONOMIC CONTEXT - PAPER 1 Q3: NEWSPAPERS.pptxECONOMIC CONTEXT - PAPER 1 Q3: NEWSPAPERS.pptx
ECONOMIC CONTEXT - PAPER 1 Q3: NEWSPAPERS.pptxiammrhaywood
 
Pharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdfPharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdfMahmoud M. Sallam
 
Presiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha electionsPresiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha electionsanshu789521
 
Roles & Responsibilities in Pharmacovigilance
Roles & Responsibilities in PharmacovigilanceRoles & Responsibilities in Pharmacovigilance
Roles & Responsibilities in PharmacovigilanceSamikshaHamane
 

Recently uploaded (20)

TataKelola dan KamSiber Kecerdasan Buatan v022.pdf
TataKelola dan KamSiber Kecerdasan Buatan v022.pdfTataKelola dan KamSiber Kecerdasan Buatan v022.pdf
TataKelola dan KamSiber Kecerdasan Buatan v022.pdf
 
Employee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxEmployee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptx
 
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdf
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdfFraming an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdf
Framing an Appropriate Research Question 6b9b26d93da94caf993c038d9efcdedb.pdf
 
CELL CYCLE Division Science 8 quarter IV.pptx
CELL CYCLE Division Science 8 quarter IV.pptxCELL CYCLE Division Science 8 quarter IV.pptx
CELL CYCLE Division Science 8 quarter IV.pptx
 
Like-prefer-love -hate+verb+ing & silent letters & citizenship text.pdf
Like-prefer-love -hate+verb+ing & silent letters & citizenship text.pdfLike-prefer-love -hate+verb+ing & silent letters & citizenship text.pdf
Like-prefer-love -hate+verb+ing & silent letters & citizenship text.pdf
 
Difference Between Search & Browse Methods in Odoo 17
Difference Between Search & Browse Methods in Odoo 17Difference Between Search & Browse Methods in Odoo 17
Difference Between Search & Browse Methods in Odoo 17
 
How to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptxHow to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptx
 
Capitol Tech U Doctoral Presentation - April 2024.pptx
Capitol Tech U Doctoral Presentation - April 2024.pptxCapitol Tech U Doctoral Presentation - April 2024.pptx
Capitol Tech U Doctoral Presentation - April 2024.pptx
 
MARGINALIZATION (Different learners in Marginalized Group
MARGINALIZATION (Different learners in Marginalized GroupMARGINALIZATION (Different learners in Marginalized Group
MARGINALIZATION (Different learners in Marginalized Group
 
ENGLISH 7_Q4_LESSON 2_ Employing a Variety of Strategies for Effective Interp...
ENGLISH 7_Q4_LESSON 2_ Employing a Variety of Strategies for Effective Interp...ENGLISH 7_Q4_LESSON 2_ Employing a Variety of Strategies for Effective Interp...
ENGLISH 7_Q4_LESSON 2_ Employing a Variety of Strategies for Effective Interp...
 
What is Model Inheritance in Odoo 17 ERP
What is Model Inheritance in Odoo 17 ERPWhat is Model Inheritance in Odoo 17 ERP
What is Model Inheritance in Odoo 17 ERP
 
Types of Journalistic Writing Grade 8.pptx
Types of Journalistic Writing Grade 8.pptxTypes of Journalistic Writing Grade 8.pptx
Types of Journalistic Writing Grade 8.pptx
 
Blooming Together_ Growing a Community Garden Worksheet.docx
Blooming Together_ Growing a Community Garden Worksheet.docxBlooming Together_ Growing a Community Garden Worksheet.docx
Blooming Together_ Growing a Community Garden Worksheet.docx
 
DATA STRUCTURE AND ALGORITHM for beginners
DATA STRUCTURE AND ALGORITHM for beginnersDATA STRUCTURE AND ALGORITHM for beginners
DATA STRUCTURE AND ALGORITHM for beginners
 
Solving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptxSolving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptx
 
AmericanHighSchoolsprezentacijaoskolama.
AmericanHighSchoolsprezentacijaoskolama.AmericanHighSchoolsprezentacijaoskolama.
AmericanHighSchoolsprezentacijaoskolama.
 
ECONOMIC CONTEXT - PAPER 1 Q3: NEWSPAPERS.pptx
ECONOMIC CONTEXT - PAPER 1 Q3: NEWSPAPERS.pptxECONOMIC CONTEXT - PAPER 1 Q3: NEWSPAPERS.pptx
ECONOMIC CONTEXT - PAPER 1 Q3: NEWSPAPERS.pptx
 
Pharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdfPharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdf
 
Presiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha electionsPresiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha elections
 
Roles & Responsibilities in Pharmacovigilance
Roles & Responsibilities in PharmacovigilanceRoles & Responsibilities in Pharmacovigilance
Roles & Responsibilities in Pharmacovigilance
 

PaNADaARRAAKCLBN1.docx

  • 2. a r i 0 h Colloids and Surfaces B: Biointerfaces 101 (2013) 353– 360 Contents lists available at SciVerse ScienceDirect Colloids and Surfaces B: Biointerfaces j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / c o l s u r f b harmacokinetics of curcumin-loaded PLGA and PLGA–PEG blend nanoparticles fter oral administration in rats ajeh Maissar Khalil , Thuane Castro Frabel do Nascimento , Diani Meza Casa , Luciana Facco Dalmolin , na Cristina de Mattos, Ivonete Hoss, Marco Aurélio Romano, Rubiana Mara Mainardes ∗ epartment of Pharmacy, Universidade Estadual do Centro- Oeste/UNICENTRO, Rua Simeão Camargo Varela de Sá 03, 85040-080 Guarapuava, PR, Brazil r t i c l e i n f o rticle history: eceived 2 March 2012 eceived in revised form 10 June 2012 ccepted 12 June 2012 vailable online 28 June 2012
  • 3. eywords: urcumin C–MS/MS ioavailability anoparticles a b s t r a c t The aim of this study was to assess the potential of nanoparticles to improve the pharmacokinetics of curcumin, with a primary goal of enhancing its bioavailability. Polylactic-co-glycolic acid (PLGA) and PLGA–polyethylene glycol (PEG) (PLGA–PEG) blend nanoparticles containing curcumin were obtained by a single-emulsion solvent-evaporation technique, resulting in particles size smaller than 200 nm. The encapsulation efficiency was over 70% for both formulations. The in vitro release study showed that cur- cumin was released more slowly from the PLGA nanoparticles than from the PLGA–PEG nanoparticles. A LC–MS/MS method was developed and validated to quantify curcumin in rat plasma. The nanoparticles were orally administered at a single dose in rats, and the pharmacokinetic parameters were evaluated and compared with the curcumin aqueous suspension. It was observed that both nanoparticles formu- lations were able to sustain the curcumin delivery over time, but greater efficiency was obtained with the PLGA–PEG nanoparticles, which showed better results in all of the pharmacokinetic parameters ana- lyzed. The PLGA and PLGA–PEG nanoparticles increased the curcumin mean half-life in approximately 4 and 6 h, respectively, and the Cmax of curcumin increased 2.9- and 7.4-fold, respectively. The distribution and metabolism of curcumin decreased when it was carried
  • 4. by nanoparticles, particularly PLGA–PEG nanoparticles. The bioavailability of curcumin-loaded PLGA–PEG nanoparticles was 3.5-fold greater than the curcumin from PLGA nanoparticles. Compared to the curcumin aqueous suspension, the PLGA and PLGA–PEG nanoparticles increased the curcumin bioavailability by 15.6- and 55.4-fold, respectively. These results suggest that PLGA and, in particular, PLGA–PEG blend nanoparticles are potential carriers for the oral delivery of curcumin. . Introduction Curcumin is a polyphenol compound extracted from the oot of Curcuma longa Linn, commonly known as turmeric. hemically, curcumin is 1,7-bis(4-hydroxy-3-methoxyphenyl)- ,6-heptadiene-3,5-dione, commonly called diferuloylmethane. urcumin has been used for centuries in Chinese and Indian edicine to treat a variety of disorders [1]. Several studies have hown that curcumin presents anti-inflammatory [2,3], antioxidant 4–6] and antimicrobial activities [7], but the most important effect s its potential use against cancer due to its ability to suppresses the roliferation of a wide variety of tumor cells [8–10]. Curcumin is ble to modulate numerous targets including transcription factors, eceptors, kinases, cytokines, enzymes and growth factors, affect- ng numerous molecular and biochemical cascades [1,11]. ∗ Corresponding author. Tel.: +55 42 3629 8160; fax: +55 42
  • 5. 3629 8102. E-mail address: [email protected] (R.M. Mainardes). 927-7765/$ – see front matter © 2012 Elsevier B.V. All rights reserved. ttp://dx.doi.org/10.1016/j.colsurfb.2012.06.024 © 2012 Elsevier B.V. All rights reserved. The great pharmacological potential of curcumin and its ther- apeutic applications are restricted because the molecule presents some drawbacks, including low aqueous solubility at acidic and physiological pH conditions, rapid hydrolysis in alkaline media and light instability, inherent to its chemical composition. The hydrophobic character of curcumin results in pharmacokinetic restrictions such as low absorption and bioavailability by oral route, extensive metabolism and rapid elimination [12,13]. The main strategies used to overcome the physicochemical limitations of cur- cumin and to increase its bioavailability are based on loading the compound in nanocarriers, such as liposomes [14], cyclodextrins [15], solid lipids [16,17] and polymeric nanoparticles [18–20]. Biodegradable polymeric nanoparticles are extensively used to improve the therapeutic properties of various drugs and bioac- tive compounds. Nanoencapsulation protects the molecules from premature degradation, improves their solubility, and promotes controlled drug release and drug targeting. Nanoparticles present low risk of toxicity, and the drug efficacy, specificity, tolerabil- ity and the therapeutic index are enhanced with their use. The pharmacokinetic parameters of the drug are modified when it
  • 8. U 54 N.M. Khalil et al. / Colloids and Surfa s loaded in nanoparticles. Specifically, there are improvements n absorption, bioavailability, and plasma circulation time, with eduction of clearance, consequently increasing the drug’s mean alf-life [21–25]. The physicochemical parameters of nanoparticles uch as particle size, surface modification charge, and hydropho- icity influence the drug’s pharmacokinetics, impacting the drug’s ioavailability and biodistribution in particular. It is well docu- ented that nanoparticles presenting a hydrophobic surface, like he surface of poly (lactide-co-glycolide) (PLGA) nanoparticles, resent short circulation times because they are rapidly recog- ized by plasmatic opsonin and cleared by cells of the mononuclear hagocytic system (MPS). The process of opsonization is one of he most important biological barriers to nanoparticle-based con- rolled drug delivery. Coating of the surface of nanoparticles with ydrophilic polymers, such as polyethylene glycol (PEG), polysor- ates or poloxamers, sterically stabilizes the particles, i.e., they re able to repel the absorption of opsonin proteins via steric epulsion forces, and thus, the particles become “invisible” to MPS ells, increasing their plasmatic circulation time and resulting in an mprovement in drug bioavailability and half-life. Also, longer plas- atic circulation times increase the probability of the nanoparticles eaching their target [26–29].
  • 9. Some recent works have demonstrated that PLGA nanoparticles re able to improve the bioavailability of curcumin after oral admin- stration [18,19,30]. PLGA–PEG nanoparticles have been developed ecause of their great potential for having long circulation times. lso, the potential advantage provided by the hydrophilic charac- er of PEG can improve the biocompatibility of the delivery system 31]. However, to the best of our knowledge, there has not yet been demonstrated report about the use of PLGA–PEG nanoparticles s carriers for curcumin. Thus, polymeric nanoparticles, especially ong-circulating nanoparticles, were evaluated as potential carriers or curcumin oral delivery. In this work, PLGA and PLGA–PEG blend nanoparticles were btained for curcumin loading. An analytical method based on C–MS/MS was developed and validated to quantify curcumin in at plasma. The nanoparticles were orally administered at a single ose in rats, and the pharmacokinetic parameters were evaluated nd compared with those of a curcumin aqueous suspension. . Materials and methods .1. Materials Curcumin (code C1386), PLGA (Resomer RG 50:50 H; Mw 0–75 kDa, inherent viscosity 0.45–0.6 dl/g), PEG (Mw 10 kDa) and
  • 10. olyvinyl alcohol (PVA, 31 kDa, 88% hydrolyzed) were purchased rom Sigma–Aldrich (USA). The internal standard, salbutamol 99%), was obtained from European Pharmacopeia. Methylene hloride and ethyl acetate were purchased from FMaia® (Brazil). nalytical HPLC-grade ethanol, acetonitrile, methanol and acetic cid were purchased from J.T. Baker (USA). All other solvents and hemicals were analytical or HPLC grade. .2. Preparation of curcumin-loaded PLGA and PLGA–PEG blend anoparticles The nanoparticles were obtained by the single-emulsion olvent-evaporation technique, as previously described [32]. riefly, curcumin (5 mg) and PLGA (50 mg) were dissolved in a ixture of ethyl acetate (1.5 mL) and methylene chloride (0.5 mL) ith or without PEG (10 mg) at room temperature. This organic hase was rapidly poured into 10 mL of PVA aqueous solution 0.5%, w/v) and emulsified by sonication for 5 min (35% of 500 W, nique® Ultrasonic Mixing, Brazil), resulting in an oil-in-water Biointerfaces 101 (2013) 353– 360 (O/W) emulsion. Next, the organic solvent was rapidly eliminated by evaporation under vacuum (20 min) at 37 ◦C. The particles were then recovered by centrifugation (19,975 × g, 30 min, 4 ◦C, Cien- tec CT-15000R centrifuge, Brazil) and washed twice with water to
  • 11. remove the surfactant. The nanoparticles were dispersed in the cry- oprotectant sucrose (5%, w/v), and the resulting nanosuspension was cooled to −18 ◦C and freeze-dried (Terroni®, Brazil). 2.3. Particle size The mean particle size and polydispersity index were deter- mined by dynamic light scattering (BIC 90 plus – Brookhaven Instruments Corp., USA). The analyses were performed at a scat- tering angle of 90◦ and a temperature of 25 ◦C. For each sample, the mean particle diameter, polydispersity and standard deviation of ten measurements were calculated. 2.4. Drug entrapment efficiency A Waters 2695 Alliance HPLC system (Milford, MA, USA) was used for curcumin quantitation. The chromatographic analysis was performed in isocratic mode using a reverse phase C18 column (VertiSep GES,Vertical Chromatography Co) with a 5 �m particle size, 4.6 mm internal diameter and 250 mm length. The mobile phase consisted of a mixture of ethanol, acetonitrile and water (80:10:10), pumped at a flow rate of 0.8 mL/min. The sample injec- tion volume was 20 �L, and the fluorescence detector was operated at an excitation wavelength of 365 nm and an emission wavelength of 512 nm. The amount of curcumin incorporated into the nanoparticles was determined directly after complete dissolution of nanoparti-
  • 12. cles in acetonitrile. The solutions were centrifuged and supernatant was collected. After the appropriate dilutions in ethanol, 20 �L of the sample was injected into the HPLC system, and the drug concen- tration was obtained by comparison with a previously constructed analytical curve. Before injection, all of the solutions were filtered through a PVDF membrane filter (0.22-�m pore size, Millipore). The entrapment efficiency (%) was estimated by comparing the amount of curcumin extracted from nanoparticles with the initial amount used for the nanoparticles preparation. 2.5. In vitro release profile The release of curcumin from nanoparticles was conducted by suspending the nanoparticles (containing 1.5 mg of curcumin) in 12 mL of phosphate saline buffer (PBS, 0.01 M, pH 7.4), and the sus- pension was divided in eight Eppendorf tubes. The experiments were performed in triplicate and under sink conditions. The tubes were kept in a shaker at 37 ◦C at 150 rpm. At predetermined time intervals, the suspension was centrifuged at 19,975 × g for 15 min to separate the released curcumin from the nanoparticles [33,34]. The resulting precipitate in each tube was dispersed in 1.5 mL of
  • 13. phosphate saline buffer and incubated until the next sampling. The released curcumin present in the supernatant (1.5 mL in each tube) was diluted in ethanol, and 20 �L of this solution was injected into the HPLC to determine the amount of curcumin released at different time intervals. 2.6. Chromatograph system and conditions for curcumin quantitation in plasma The LC–MS/MS analysis was conducted in positive ion ESI mode on a Quattro Micro API–Waters hexapole mass spectrom- eter connected to a liquid chromatograph (Waters Alliance). The analysis was conducted on a Phenomenex Luna C18(2) 100A col- umn (250 mm × 4.6 mm, 5 �m). The mobile phase consisted of ces B: m o a S a ( a u m
  • 15. s t c d s t 4 d e d e [ b f N.M. Khalil et al. / Colloids and Surfa ethanol and 0.05% acetic acid solution (80:20, v/v) at a flow rate f 1.0 mL/min. The sheath gas and auxiliary gas were tuned to give n optimum response as necessary. The needle voltage was 4.5 kV. albutamol was used as internal standard (IS) [35]. Argon was used s the collision gas at collision energy of 15 eV (curcumin) and 18 eV salbutamol). The collision energy was individually tuned for each nalyte to obtain an optimum value. The analytes were quantified sing selected ion reaction monitoring (SRM). The ion transitions /z 369.3→285.0 and m/z 240.0→147.7 were used for the determi- ation of curcumin and salbutamol, respectively. The
  • 16. autosampler ooler was maintained at 4 ◦C. .7. Preparation of curcumin standards and quality control A concentrated stock standard of curcumin and salbutamol (IS) ere prepared by dissolving 4 mg of each compound in 20 mL f methanol, generating a 200 �g/mL stock solution. Eight point alibration curves were prepared by serial dilution of the cur- umin stock solution (200 �g/mL in methanol) in the range of .5–500 ng/mL. The calibration curve was prepared daily using .45 mL of blank plasma with 50 �L of the appropriate working olution, resulting in concentrations of 0.5, 10, 25, 50, 100, 200, 350 nd 500 ng/mL. Three quality controls (QC) were prepared at 1.5 low concentration), 225 (medium concentration) and 450 ng/mL high concentration). .8. Plasma sample preparation To 100 �L of rat plasma sample (or a calibration standard or a QC ample) were added 100 �L of salbutamol (IS) and 100 �L of 0.5 M odium hydroxide (to assist in the extraction of curcumin). The mix- ure was vortexed for 1 min. After the curcumin was extracted with 300 �L of ethyl acetate (liquid–liquid extraction), followed by agi- ation in a shaker (10 min), it was centrifuged at 10,000 rpm at 4 ◦C or 10 min. The supernatant was evaporated using nitrogen gas in a ample concentrator. The obtained residue was reconstituted with
  • 17. he mobile phase and vortexed for 20 s. The samples were subjected o LC–MS/MS analysis. .9. Bioanalytical method validation The specificity of the method was investigated by comparing the hromatogram of blank plasma with the blank plasma spiked with tandard solutions and with the samples collected from rats after urcumin administration. The linearity of the bioanalytical assay was evaluated with a otal of eight calibration standards over the concentration range of .5–500 ng/mL. Calibration curves were constructed by linear least- quares regression analysis by plotting the peak-area ratios versus he drug concentrations. The limit of quantitation (LOQ) was defined as the lowest con- entration of the analyte in the calibration curve that could be etected with a variation of less than 15%.The intra-day preci- ion and accuracy were determined within one day by analyzing en replicates of the QC samples at concentrations of 1.5, 225 and 50 ng/mL of curcumin. The inter-day precision and accuracy were etermined on two separate occasions using replicates (n = 10) of ach concentration used. The intra- and inter-day precision was efined as the relative standard deviation (R.S.D.). The accuracy was xpressed using the following equation (1): measured concentration
  • 18. ] × 100 (1) nominal spiked concentration The freeze–thaw stability of the plasma samples was evaluated y exposing QC samples at low and high concentrations to four reeze–thaw (−20 ◦C to room temperature) cycles before sample Biointerfaces 101 (2013) 353– 360 355 preparation. The stability of the samples in the autosampler was evaluated by analyzing the extracted QC samples after being placed in the autosampler at 20 ◦C for 6 h, at which time the samples were analyzed. The long-term stability was verified by freezing (−20 ◦C) the QC samples for 250 days. Freshly processed standard samples were used to quantitate all of the QC samples. The analyses were performed in quintuplicate. 2.10. Pharmacokinetic study Male adult Wistar rats with a mean body weight of 200–300 g were fasted overnight prior to the experiments, with free access to water. The experimental protocol was approved by the Institutional Animal Ethics Committee of the Universidade Estadual de Ponta Grossa, Brazil (Registration no. 06/2010). The rats were divided randomly into three groups (n = 5). The formulations (curcumin aqueous suspension, dispersion of the curcumin-loaded PLGA
  • 19. nanoparticles and dispersion of the curcumin-loaded PLGA– PEG blend nanoparticles) were administered by oral gavage at a single dose of 50 mg/kg. The nanoparticles were dispersed in ultrapure water. Blood samples (500 �L) were withdrawn from the tail vein into heparinized microtubes at the following times: 0.25, 0.5, 1, 1.5, 2, 4, 8, 12 and 24 h after dosing. The blood samples were centrifuged at 3020 × g for 10 min. The supernatant was collected, transferred to tightly sealed plastic tubes and stored at −20 ◦C until analysis by LC–MS/MS. After each sampling the same volume removed was replaced with saline solution. 2.11. Data analysis and statistics All of the in vitro results were expressed as the mean ± standard deviation (S.D.) of three replicates. The in vivo results were presented as the mean ± S.D. of five replicates. Pharmacokinetic parameters were estimated using the model-independent method. The terminal elimination rate constant (Ke) was estimated by a linear regression analysis of the terminal portion of the log- linear blood concentration–time profile of curcumin. The terminal elimination half-life (t1/2) was calculated from Ke using the for- mula t1/2 = 0.693/Ke. The maximum observed plasma concentration (Cmax) and the time taken to reach it (Tmax) were obtained from the curve plotting curcumin concentration vs. time. The area
  • 20. under each drug concentration time curve (AUC, ng/mL h) to the last data point was calculated by the linear trapezoidal rule and extrapolated to time infinity by the addition of CLast/Ke, where CLast is the con- centration of the last measured plasma sample. The apparent body clearance (Cl) was calculated using the equation Cl = dose/AUC. The apparent volume of distribution (Vd) was calculated by the equation Vd = dose/Ke AUC. Statistical analysis of the data was performed via one-way analysis of variance (ANOVA). The results were considered statistically significant if p < 0.05. 3. Results and discussion 3.1. Preparation of the curcumin-loaded PLGA and PLGA–PEG blend nanoparticles The nanoparticles containing curcumin were successfully obtained by the single-emulsion solvent-evaporation method. The choice of a nanoencapsulation method is based on the drug solubil- ity, and because curcumin is hydrophobic, the method of reducing the size of the emulsion oil-in-water (O/W) is adequate for this molecule. The ultrasonication was crucial to reduce the emulsion globules to nanometer size. Table 1 illustrates the size
  • 21. characteris- tics of the obtained nanoparticles. Both formulations, the PLGA and PLGA–PEG blend nanoparticles containing curcumin, presented 356 N.M. Khalil et al. / Colloids and Surfaces B: Biointerfaces 101 (2013) 353– 360 Table 1 Curcumin nanoparticle characteristics. Polymer Particle size (nm)a Polydispersity indexa Size distributiona Encapsulation efficiency (%)b PLGA 161.93 ± 6.7 0.042 ± 0.01 146.2–200.7 nm (100%) 77.07 ± 8.16 PLGA–PEG 152.37 ± 4.5 0.077 ± 0.01 109.9–185.1 nm (100%) 73.22 ± 9.77 Values reported as mean ± S.D. m o p n r l w e d d e
  • 22. 3 a T i t T t n l b o s n 2 n d c m F t a (n = 3). b (n = 30). onodisperse profiles and narrow size distributions. The presence f PEG did not influence the mean particle size, but the polydis- ersity index was superior (p < 0.05) than that obtained from PLGA anoparticles, while maintaining a monomodal profile. The encapsulation efficiency was determined directly, and the esults are presented in Table 1. The method used for nanoencapsu-
  • 23. ation resulted in significant enclosure of curcumin, and the process as found to be highly reproducible. The PEG did not influence the ncapsulation, as the values between batches were not significantly ifferent (p > 0.05). Indeed, the hydrophilic character of PEG kept it irected to aqueous phase, while the hydrophobic core of PLGA can ntrap the hydrophobic drugs. .2. In vitro curcumin release profile The in vitro release of curcumin from nanoparticles was evalu- ted simulating physiological conditions (37 ◦C, PBS buffer pH 7.4). he in vitro release profiles of curcumin were obtained by graph- ng the cumulative percentage of the drug released with respect o the amount of curcumin encapsulated as a function of the time. he experiment was performed over nine days. Fig. 1 illustrates he curcumin release profiles from the PLGA and PLGA–PEG blend anoparticles and indicates that there was a pronounced time pro- ongation of the drug release. It is evident that the PEG influenced the curcumin release ecause there was a great difference between the release profiles f curcumin from nanoparticles of different compositions. A bipha- ic release pattern of curcumin was observed from the PLGA– PEG anoparticles, where the initial 24 h period released
  • 24. approximately 1% of drug, followed by a sustained release to a total of 56.9% over ine days of observation. This initial burst release may be due to rug desorption from the particle surface, and the sustained release an be characterized by the drug diffusion through the polymeric atrix and subsequent diffusion/erosion of the polymeric matrix. ig. 1. In vitro release profile of curcumin from PLGA and PLGA–PEG blend nanopar- icles in PBS (0.01 M, pH 7.4) at 37 ◦ C. Values reported as the mean ± S.D. (n = 3). Curcumin release from the PLGA nanoparticles was slower than from the PLGA–PEG blend nanoparticles (p < 0.05), and the release was progressive because it did not have a biphasic profile. After 24 h, only 5.8% of the drug had been released, and in nine days, 37% of the curcumin had been released. In general, it can be affirmed that the drug release depends upon the solubility, diffusion and biodegradation of the matrix mate- rials. Thus, the drug release mechanisms can be modified by the choice of polymer matrices. Drug release also depends upon the loading efficiency of the drug and the size of the nanoparticles [24]. In our case, because the particle size and curcumin loading are sim- ilar for the PLGA and PLGA–PEG nanoparticles, we can attest that the difference between the amount of drug released from the two nanoparticles is due to the presence of PEG, as it has a
  • 25. hydrophilic character and can enhance the water permeation and drug diffu- sion through the polymeric matrix [36]. It is possible that curcumin strongly interacts with the PLGA matrix, thus retarding the release capability, and that the PEG can increase the wettability of the polymeric surface and matrix, contributing to the increase in drug release. The results show that the PLA-PEG nanoparticles released more curcumin than the PLGA nanoparticles (by approximately 1.5-fold; p < 0.05) during the period analyzed. 3.3. Bioanalytical method development and validation A LC–MS/MS method for the determination and quantitation of curcumin in rat plasma has been developed and validated. Initial runs were conducted with mobile phases composed of acetoni- trile:0.2% formic acid solution (40:60, v/v), acetonitrile:1% formic acid solution (70:30, v/v), acetonitrile:0.1% acetic acid solution (70:30, v/v) and acetonitrile:0.005% acetic acid solution (70:30, v/v). In all of these combinations, the curcumin peak resulted in tailing, and the signal was slow. Testing several ratios of methanol and acetic acid, the combina- tion that resulted in a sharp peak with a sufficient response area was using methanol and 0.05% acetic acid solution (80:20, v/v) as the mobile phase at a flow rate of 1 mL/min. The retention times were approximately 1.08 min and 0.82 min for curcumin and IS, respectively, and the total run time was 2 min.
  • 26. The specificity of the method was evaluated by comparing the chromatograms of curcumin in plasma (standard and sample) and those of potentially interfering plasma components. Representa- tive chromatograms are shown in Fig. 2, including a blank plasma sample (Fig. 2A), plasma containing curcumin and salbutamol stan- dard (Fig. 2B) and a plasma sample obtained 30 min after the oral administration of 50 mg/kg of curcumin-loaded PLGA nanoparticles (Fig. 2C). The resulting chromatograms show the assay specificity, as there were no endogenous plasma components eluted at the retention time of curcumin or IS. The method was validated over a wide concentration range, and the results were directly obtained and extrapolated on the calibra- tion curve. The calibration lines were shown to be linear from 0.5 to 500 ng/mL (r2 = 0.9941). The method was sensitive, and the LOQ was low (0.5 ng/mL). Other LC–MS/MS methods described in the N.M. Khalil et al. / Colloids and Surfaces B: Biointerfaces 101 (2013) 353– 360 357 F n stan c
  • 27. l 1 a r ig. 2. Representative chromatograms of (A) a blank plasma sample, (B) a curcumi urcumin, while that the lower are indicative of the peak of the internal standard. iterature for curcumin determination in plasma showed a LOQ of 0 ng/mL [16] and 2.5 ng/mL [37]. Table 2 shows a summary of intra- and inter-day precision and ccuracy for curcumin detection in rat plasma. The intra-day accu- acy of curcumin for rat plasma samples was 101.72–110.54% for dard, and (C) a curcumin sample. The upper peak of the figure is representative of QC samples with a R.S.D. of less than 6.70%. The inter-day accu- racy of curcumin for rat plasma samples ranged from 96.34% to 107.94% for QC samples with an R.S.D. of less than 4.34%. These results were within the limits established by the FDA guidelines for the validation of bioanalytical methods [38].
  • 28. 358 N.M. Khalil et al. / Colloids and Surfaces B: Biointerfaces 101 (2013) 353– 360 Table 2 Intra-day and inter-day precision and accuracy of curcumin in rat plasma (n = 10). Nominal concentration (ng/mL) Measured concentration (ng/mL) R.S.D. (%) Accuracy (%) Intra-daya 1.5 1.66 ± 0.04 2.33 110.54 225 224.66 ± 8.00 3.56 99.85 450 457.76 ± 30.66 6.70 101.72 Inter-dayb 1.5 1.62 ± 0.05 3.39 107.94 225 216.77 ± 9.41 4.34 96.34 450 436.59 ± 18.74 4.29 97.02 a t o r s s c
  • 29. p a w o o s f 3 o m n c p s c r t m 1 A w a c f c i T S The analyses were performed in the same day; R.S.D. = relative standard devia- ion.
  • 30. b The analyses were performed in two different days within one month. Table 3 lists the data from the stability tests. No significant loss f curcumin (≤1.4%) was observed after storage of the plasma at oom temperature on the bench top for at least 6 h. The plasma amples were stable over at least four freeze/thaw cycles and were table at −20 ◦C for at least 250 days, with no significant loss of urcumin (≤1.8%). These results suggested that the plasma sam- les could be stored at −20 ◦C for long periods, could be thawed nd refrozen and could be maintained at room temperature for 6 h ithout compromising the integrity and accuracy of the samples. The sensitivity of this LC–MS/MS method offered advantages ver other LC–MS/MS methods and conventional HPLC–UV meth- ds applied for curcumin pharmacokinetics. Also, the excellent pecificity and short run time analysis make this method efficient or curcumin pharmacokinetic applications. .4. Pharmacokinetics study The mean curcumin plasma concentration–time profiles after ral administrations of 50 mg/kg of curcumin in different for- ulations, curcumin aqueous suspension, curcumin-loaded PLGA anoparticles and curcumin-loaded PLGA–PEG blend nanoparti- les, are expressed in Fig. 3. The Table 4 summarizes the relevant harmacokinetic parameters. After the oral administration of a curcumin aqueous suspen- ion, the drug was absorbed quickly, and a maximum plasma
  • 31. oncentration (Cmax) of approximately 4.066 ± 0.564 ng/mL was eached in 30 min. Thereafter, the curcumin plasma concentra- ion decreased abruptly, as the drug was distributed and rapidly etabolized, resulting in a high Ke and short t1/2, approximately .1 h. The curcumin was detected up to 8 h after administration. sustained release of curcumin over 24 h was observed when it as carried by the two nanoparticles formulations. Thirty minutes fter oral administration of the curcumin-loaded PLGA nanoparti- les, the mean plasma concentration was 4.57 ± 0.35 ng/mL, and or the curcumin-loaded PLGA–PEG nanoparticles, the plasma oncentration was 9.1 ± 0.95 ng/mL. There was a significant ncrease (p < 0.01) in curcumin absorption from the PLGA–PEG able 3 tability of curcumin in rat plasma. Sample condition Curcumin nominal concentration 1.5 ng/mL Concentration measured (ng/mL)a R.S.D. (%)b Acc 6 h at room temperature 1.62 ± 0.1 1.42 108 Freeze–thaw four cycles 1.63 ± 0.04 0.7 109 250 days at −20 ◦ C 1.6 ± 0.05 1.1 107 a Values reported as mean ± S.D. (n = 5). b Relative standard deviation, calculated comparing with CQ
  • 32. freshly prepared. Fig. 3. Comparison of in vivo plasma concentration vs. time profiles of the different curcumin formulations. All values reported are the mean ± S.D. (n = 5). nanoparticles in the first 30 min compared to free curcumin and curcumin from the PLGA nanoparticles. The curcumin concentra- tion increased to 11.783 ± 0.454 ng/mL, Cmax, after 2 h (Tmax), and to 29.778 ± 4.632 ng/mL, Cmax, after 3 h (Tmax), with the PLGA and PLGA–PEG nanoparticles, respectively. Compared to free curcumin, the Cmax of curcumin from PLGA nanoparticles and PLGA– PEG nanoparticles was increased 2.9- and 7.4-fold, respectively. The increase in Cmax indicates that the nanoparticles were effective in increasing drug absorption, and the delayed Tmax demon- strates an obvious sustained release of curcumin. The distribution and metabolism of curcumin were decreased when it was car- ried by nanoparticles (p < 0.01). The clearance of curcumin from the PLGA and PLGA–PEG nanoparticles was 16.3- and 61.6- fold lower than that of free curcumin, respectively. The PLGA–PEG nanoparticles and PLGA nanoparticles decreased the curcumin volume of distribution by 11.6- and 4.6-fold compared to free curcumin. Thus, the t1/2 of curcumin from the PLGA increased to 4 h, and that from the PLGA–PEG nanoparticles was increased to 6 h, while for free curcumin the t1/2 was 1 h. There was a significant difference in the AUC0–inf between the curcumin
  • 33. aqueous suspension, the curcumin–PLGA nanoparticles and the curcumin–PLGA–PEG nanoparticles (p < 0.01). Between the two nanoparticle formulations, the curcumin from PLGA–PEG pre- sented a relative bioavailability 3.5-fold superior to that of the curcumin from PLGA nanoparticles. Compared to the curcumin aqueous suspension, the PLGA and PLGA–PEG blend nanoparticles increased the curcumin bioavailability 15.6- and 55.4-fold, respec- tively. Recently, Shaikh et al. [19] demonstrated that the PLGA nanoparticles were able to increase the curcumin bioavailability at least 9-fold when compared to curcumin administered with an absorption enhancer. Tsai et al. [20] developed curcumin-loaded 450 ng/mL uracy (%) Concentration measured (ng/mL)a R.S.D. (%)b Accuracy (%) .31 474.50 ± 13.36 0.47 105.44 .00 481.07 ± 18.52 0.56 106.9 .25 483.99 ± 5.6 1.8 107.55 N.M. Khalil et al. / Colloids and Surfaces B: Biointerfaces 101 (2013) 353– 360 359 Table 4 Pharmacokinetic parameters of curcumin following single oral
  • 34. administration of curcumin aqueous solution, curcumin-loaded PLGA nanoparticles and curcumin-loaded PLGA–PEG nanoparticles, in rats (n = 5). Pharmacokinetic parameters Formulations Curcumin aqueous suspension Curcumin PLGA nanoparticles Curcumin PLGA–PEG nanoparticles Dose (mg/kg) 50 50 50 AUC0–t (h ng/mL) 8.695 ± 1.872 134.251 ± 3.446* , # 447.80 ± 64.028* AUC0–inf (h ng/mL) 8.762 ± 1.862 137.162 ± 3.694* , # 485.941 ± 54.663* Cmax (ng/mL) 4.066 ± 0.564 11.783 ± 0.454* , # 29.778 ± 4.632* Tmax (h) 0.5 2* , # 3* Ke (1/h) 0.631 ± 0.072 0.178 ± 0.021* , # 0.119 ± 0.021* t1/2 (h) 1.109 ± 0.124 3.929 ± 0.451* , # 5.979 ± 1.126* Vd (L/kg) 9432.536 ± 2511.617 2073.664 ± 352.612* , # 900.544 ± 225.772* Cl (L/h/kg) 5859.700 ± 1399.927 365.191 ± 37.351* , # 103.679 ± 10.903* Values reported as mean ± S.D. (n = 5). AUC: area under the plasma concentration–time curve; Cmax : peak concentration; Tmax : time to reach peak concentration; Ke : constant of elimination; t1/2 : mean half-life; Vd : apparent volume of distribution; Cl: clearance. P a m u a
  • 36. n n r w m t [ [ * Significantly different of free curcumin (p < 0.01). # Significantly different of curcumin from PLGA–PEG nanoparticles (p < 0.01). LGA nanoparticles. When these particles were intravenously dministrated in rats, a significant amount of curcumin was found ainly in the spleen due to phagocytic cell uptake in the retic- loendothelial system. Xie et al. [30] showed that after the oral dministration of curcumin-loaded PLGA nanoparticles, the cur- umin had a 5.6-fold higher relative bioavailability and had a longer alf-life than that of native curcumin. In a similar work, Anand et al. 18] demonstrated that curcumin-loaded PLGA nanoparticles have nhanced cellular uptake, increased bioactivity in vitro and supe- ior bioavailability in vivo relative to free curcumin. To date, no tudy has compared the pharmacokinetics of curcumin loaded in LGA and PLGA–PEG blend nanoparticles. In our study, the significant difference in pharmacokinetic arameters, mainly bioavailability and half-life, between the free urcumin aqueous suspension and the curcumin-loaded PLGA and
  • 37. LGA–PEG nanoparticle dispersions is explained by the inherent roperties of colloidal nanoparticles in biological media, which pro- ong drug release and its in vivo trajectory. The in vitro release rofile demonstrated that curcumin is released more rapidly from LGA–PEG nanoparticles than from PLGA nanoparticles and could e more quickly available in blood. It is well supported that pharma- okinetic parameters are altered depending upon the nanoparticles sed, and their surface composition plays an important role in rug bioavailability [39,40]. PEG is frequently used for the sur- ace modification of various polymeric nanoparticles because it xhibits excellent biocompatibility and is able to improve the long- erm systemic circulation of the nanoparticles. The PEG coating on he surface of the polymer reduces the interactions between the anoparticles and the enzymes of the digestive fluids and increases he uptake of the drug in the blood stream and lymphatic tissue 41]. This effect can explain the difference between the curcumin harmacokinetics from PLGA and PLGA–PEG blend nanoparticles. he ability of the PEG to make the coated nanoparticles invisible to ecognition by MPS cells gives the particles long circulation time. onsequently, the drug half-life and bioavailability are higher than hose of a drug carried in uncoated nanoparticles [27]. We recently emonstrated that the presence of PEG in PLA nanoparticles con-
  • 38. aining the antiretroviral zidovudine was essential in promoting he increase in drug bioavailability after intranasal administration n rats [42]. The increased curcumin bioavailability obtained with the anoparticulate systems confirms the excellent abilities of the anoparticles to modulate the physicochemical properties of drugs, esulting in improved pharmacokinetics profiles. Because the poor ater solubility and low oral bioavailability of curcumin are the ajor drawbacks in its medicinal application, the studied nanopar- icles represent an important initial step in the development of [ [ [ a medicine containing curcumin, using the nanotechnology as a tool. 4. Conclusions In this study, nanoparticles coated or not with PEG were suc- cessfully prepared by the emulsion solvent-evaporation method. Also, an analytical method for determining curcumin in plasma was optimized. We demonstrated that all curcumin pharmacokinetic parameters were improved by nanoparticles, especially PLGA– PEG nanoparticles. The curcumin Cmax, Tmax, t1/2 and AUC were
  • 39. signifi- cantly increased by nanoparticles, while distribution and clearance were decreased. The PLGA–PEG nanoparticles were able to increase the curcumin bioavailability in 3.5-fold compared to curcumin from PLGA nanoparticles. Compared to curcumin aqueous suspen- sion, PLGA and PLGA–PEG nanoparticles increased the curcumin bioavailability in 15.6 and 55.4-fold, respectively. These results demonstrate the great potential of PLGA and mainly PLGA– PEG blend nanoparticles as carriers for the oral delivery of curcumin. Acknowledgments This study was supported by Conselho Nacional de Desenvolvi- mento Científico Tecnológico (CNPq) (577183/2008-7), Fundaç ão Araucária (462/2010) and FINEP (01.08.0211.00). References [1] A. Goel, B.A. Kunnumakkara, B.B. Aggarwal, Biochem. Pharmacol. 75 (2008) 787. [2] I. Brouet, H. Ohshima, Biochem. Biophys. Res. Commun. 206 (1995) 533. [3] X. Gao, J. Kuo, H. Jiang, D. Deeb, Y. Liu, G. Divine, R.A. Chapman, S.A. Dulchavsky, S.C. Gautam, Biochem. Pharmacol. 68 (2004) 51. [4] T. Masuda, K. Hidaka, A. Shinohara, T. Maekawa, Y. Takeda, H. Yamaguchi, J.
  • 40. Agric. Food Chem. 47 (1999) 71. [5] A.P. Lakshmanan, K. Watanabe, R.A. Thandavarayan, F.R. Sari, H. Meilei, V. Soeti- kno, S. Arumugam, V.V. Giridharan, K. Suzuki, M. Kodama, Free Radic. Res. 45 (2011) 788. [6] O.A.K. Khalil, O.M.M.F. Oliveira, J.C.R. Vellosa, A.U. de Quadros, L.M. Dalposso, T.K. Karam, R.M. Mainardes, N.M. Khalil, Food Chem. 133 (2012) 1001. [7] A. Mazumder, K. Raghavan, J. Weinstein, K.W. Kohn, Y. Pommier, Biochem. Pharmacol. 49 (1995) 1165. [8] B.B. Aggarwal, A. Kumar, A.C. Bharti, Anticancer Res. 23 (2003) 363. [9] B.B. Aggarwal, K.B. Harikumar, Int. J. Biochem. Cell Biol. 41 (2009) 40. 10] Z.M. Shao, Z.Z. Shen, C.H. Liu, M.R. Sartippour, V.L. Go, D. Heber, M. Nguyen, Int. J. Cancer 98 (2002) 234. 11] P. Anand, C. Sundaram, S. Jhurani, A.B. Kunnumakkara, B.B. Aggarwal, Cancer Lett. 267 (2008) 133. 12] P. Anand, A.B. Kunnumakkara, R.A. Newman, B.B. Aggarwal, Mol. Pharmacol. 4 (2007) 807. 13] R.A. Sharma, W.P. Steward, A.J. Gescher, Adv. Exp. Med.
  • 41. Biol. 595 (2007) 453. 14] A. Kunwar, A. Barik, R. Pandey, K.I. Priyadarsini, Biochim. Biophys. Acta 1760 (2006) 1513. 3 ces B: [ [ [ [ [ [ [ [ [ [ [ [ [ [ [ [
  • 42. [ [ [ [ [ [ [ [ [ 60 N.M. Khalil et al. / Colloids and Surfa 15] V.R. Yadav, S. Prasad, R. Kannappan, J. Ravindran, M.M. Chaturvedi, L. Vaahtera, J. Parkkinen, B.B. Aggarwal, Biochem. Pharmacol. 80 (2010) 1021. 16] V. Kakkar, S. Singh, D. Singla, S. Sahwney, S.A. Chauhan, G. Singh, I.P. Kaur, J. Chromatogr. B: Analyt. Technol. Biomed. Life Sci. 878 (2010) 3427. 17] W. Tiyaboonchai, W. Tungpradit, P. Plianbangchang, Int. J. Pharm. 337 (2007) 299. 18] P. Anand, H.B. Nair, B. Sung, A.B. Kunnumakkara, V.R. Yadav, R.R. Tekmal, B.B. Aggarwal, Biochem. Pharmacol. 79 (2010) 330. 19] J. Shaikh, D.D. Ankola, V. Beniwal, D. Singh, M.N.V. Ravi Kumar, Eur. J. Pharm. Sci. 37 (2009) 223.
  • 43. 20] Y.M. Tsai, C.F. Chien, L.C. Lina, T.H. Tsai, Int. J. Pharm. 416 (2011) 331. 21] A. Frank, E. Pridgen, L.K. Molnar, O.C. Farokhzad, Mol. Pharmacol. 5 (2008) 505. 22] N.M. Khalil, E. Carraro, L.F. Cótica, R.M. Mainardes, Expert. Opin. Drug Deliv. 8 (2011) 95. 23] N.M. Khalil, R.M. Mainardes, Curr. Drug Deliv. 6 (2009) 261. 24] A. Kumari, S.K. Yadav, S.C. Yadav, Colloids Surf. B Biointerfaces 75 (2010) 1. 25] J.C. Leroux, E. Allemann, F. De Jaeghere, E. Doelker, R.L. Gurny, J. Control. Release 39 (1996) 339. 26] R.M. Mainardes, M.P.D. Gremião, I.L. Brunetti, L.M. da Fonseca, N.M. Khalil, J. Pharm. Sci. 98 (2009) 257. 27] D.E. Owens, N.A. Peppas, Int. J. Pharm. 307 (2006) 93. [ [ [ Biointerfaces 101 (2013) 353– 360 28] S. Stolnik, L. Illum, S.S. Davis, Adv. Drug Deliv. Rev. 16 (1995) 195. 29] V.P. Torchilin, V.S. Trubetskoy, Adv. Drug Deliv. Rev. 16 (1995) 141.
  • 44. 30] X. Xie, Q. Tao, Y. Zou, F. Zhang, M. Guo, Y. Wang, H. Wang, Q. Zhou, S. Yu, J. Agric. Food Chem. 59 (2011) 9280. 31] K. Avgoustakis, Curr. Drug Deliv. 1 (2004) 321. 32] M. Li, O. Rouaud, D. Poncelet, Int. J. Pharm. 363 (2008) 26. 33] C. Mohanty, S.K. Sahoo, Biomaterials 31 (2010) 6597. 34] K.R. Das, N. Kasoju, U. Bora, Nanomed. Nanotech. Biol. Med. 6 (2010) 153. 35] A. Liu, H. Lou, L. Zhao, P. Fan, J. Pharm. Biomed. Anal. 40 (2006) 720. 36] M.T. Peracchia, R. Gref, Y. Minamitake, A. Domb, N. Lotan, R. Langer, J. Control. Release 46 (1997) 223. 37] K.Y. Yang, L.C. Lin, T.Y. Tseng, S.C. Wang, T.H. Tsai, J. Chromatogr. B: Analyt. Technol. Biomed. Life Sci. 853 (2007) 183. 38] US Food and Drug Administration, Guidance for Industry, Bioanalytical Method Validation, Centre for Drug Evaluation and Research (CDER), Rockville, 2001. 39] V. Hoffart, A. Lamprecht, P. Maincent, T. Lecompte, C. Vigneron, N. Ubrich, J. Control. Release 113 (2006) 38. 40] N. Ubrich, C. Schmidt, R. Bodmeier, M. Hoffman, P. Maincent, Int. J. Pharm. 288 (2005) 169. 41] M. Tobio, A. Sánchez, A. Vila, I.I. Soriano, C. Evora, J.L.
  • 45. Vila-Jato, M.J. Alonso, Colloids Surf. B Biointerfaces 18 (2000) 315. 42] R.M. Mainardes, N.M. Khalil, M.P.D. Gremião, Int. J. Pharm. 395 (2010) 266. Pharmacokinetics of curcumin-loaded PLGA and PLGA–PEG blend nanoparticles after oral administration in rats1 Introduction2 Materials and methods2.1 Materials2.2 Preparation of curcumin-loaded PLGA and PLGA–PEG blend nanoparticles2.3 Particle size2.4 Drug entrapment efficiency2.5 In vitro release profile2.6 Chromatograph system and conditions for curcumin quantitation in plasma2.7 Preparation of curcumin standards and quality control2.8 Plasma sample preparation2.9 Bioanalytical method validation2.10 Pharmacokinetic study2.11 Data analysis and statistics3 Results and discussion3.1 Preparation of the curcumin-loaded PLGA and PLGA–PEG blend nanoparticles3.2 In vitro curcumin release profile3.3 Bioanalytical method development and validation3.4 Pharmacokinetics study4 ConclusionsAcknowledgmentsReferences Abstract: The medicinal properties of Curcumin obtained from Curcuma longa L. cannot be utilised because of poor bioavailability due to its rapid metabolism in the lover and intestinal wall. In this study, the effect of combining piperine, a known inhibitor of hepatic and intestinal glucuronidation, was evaluated on the bioavailability of Curcumin in rats and healthy human volunteers. When Curcumin was given alone, the dose 2g/kg to rats, moderate serum concentrations were achieved over a period of 4 h. Concomitant administration of piperine 20mg/kg increased (P < 0.02), and he bioavailability was increased by 154%. On the other hand in humans after a dose of 2g Curcumin alone, serum levels were either
  • 46. undetectable or very low. Concomitant administration of piperine 20mg produced much higher concentrations from 0.25 to 1h post drug (P < 0.01 at 0.25 and 0.5h; P < 0.001 at 1h), the increase in bioavailability was 2000%. The study shows that in the dosages used, piperine enhances the serum concentrations, extent of absorption and bioavailability of curcumin in both rats and humans with no adverse effects. Key words: Curcumin, piperine, pharmacokinetics, Curcuma longa, Zingiberaceae Introduction Curcumin in obtained from Curcuma longa L (Zingiberaceae), a perennial herb widely cultivated in tropical regions of Asian. Its rhizome is extensively used for imparting colour and flavour to food. Current traditional Indian medicine claims the use of its powder, turmeric, against a wide variety of diseases (1). Extensive scientific research (2) on curcumin had demonstrated a wide spectrum of therapeutic effects which range from anti- inflammatory, wound healing, antispasmodic, anticoagulant, antitumor activities (3) and recently, with potential utility in autoimmune deficiency syndrome (4). Pharmacokinetic properties of curcumin indicate that following oral administration, it is poorly absorbed (3) and only traces of the compound appear in the blood, while most of it is excreted in the faeces (5). The transformation of curcumin into an unidentified compound during absorption (6) and its glucuronidation in the liver (5, 7) are probably responsible for its low concentration in the blood.
  • 47. Planta Medica 64 (1998) 353 – 356 ©Georg Thieme Veriag Stuttgart – New York Black pepper (Piper nigrum L) and long pepper (Piper longum L) have been in use as spices from ancient time’s thoughout the world. A major component of the Piper species id the alkaloid piperine (l-piperoylpiprtidine), which had been reported to enhance the bioavailability of drugs by inhibition of glucuronidation in the liver (8) and small intestine (9). In view of the potential therapeutic utility of curcumin it appeared pertinent to examine the effect of piperine, a known hepatic and intestinal metabolic inhibitor, on the pharmacokinetic disposition of curcumin in animals and man, to provide a scientific rationale of assigning it a rightful place in the pharmacologist’s armamentarium. Materials and Methods Animal Studies Albina Wistar rats (n=96) of both sexes (150-200g) were chosen for the study. They were housed in well ventilated cages, fed on commercial rat pellets supplied by Hind Lever, Mumbai, with tap water as libitum. They were divided into two sex and weight matched groups (n=6/group/time cut), one group for administration of curcumin and the other for concomitant curcumin and piperine. Curcumin and piperine were supplied in pure powder for by Sami Chemicals and Extracts, Bangalore, India. Both the compounds were administered orally to fasted rats as an aqueous suspension. The in group that received both drugs, curcumin was administered first followed immediately by piperine. Control rats received water only. Curcumin was given in a dose of 2g/kg and piperine, 20mg/kg.
  • 48. Under ether anaesthesia, pre and post drug jugular vein blood samples were collected from both groups of rats into centrifuge tubes at the time intervals – 0, 0.25, 0.50, 0.75, 1, 2, 3, 4, 5 and 6h. The blood was allowed to clot at room temperature for about 1h and then centrifuged at 3000rpm for 10min. The serum was separated out carefully using Pasteur pipettes into storage tubes and frozen at -20°C prior to analysis. Influence of Piperine on the Pharmacokinetics of Curcumin in Animals and Human Volunteers Guido Shoba₁, David Joy₁, Thangam Joseph₁, M. Majeed₂. R. Rajendran₂, and P.S.S.R. Srinivas₂ ₁Department of Pharmacology, St John’s Medical College, Bangalore, India ₂5AMI Chemicals & Extracts (P) Ltd., Banaglore, India Received: August 1, 1997; Revision accepted: October 18, 1997
  • 49. Human volunteer studies Ten healthy male volunteers, 20 to 26 years, weighing 50 – 75kg (mean 60 ± 1.93) participated in a randomized cross over trial, to determine the comparative bioavailability and pharmacokinetic profile of curcumin when given alone and with piperine. Complete physical examination and an electrocardiogram were done. Laboratory tests comprising complete blood counts and haemoglobin percentage, blood biochemistry consisting of blood urea nitrogen (BUN) serum creatinine, total and conjugated bilirubin, alkaline phosphatase, aspartate transaminase (ASAT), alanine transaminase (ALAT), urine albumin, and sugar were preformed to confirm that the subjects included in the study were normal. The study was formally approved by the Institutional Ethical Committee and informed consent was obtained for all subjects. Subjects abstained from food since 10pm of the previous evening and reported to the laboratory at 7am. Venepuncture was done using a 20g scalp vein set with heparin lock and left in situ. Blood samples (5ml) were collected (without anticoagulant) at 0, 0.25, 0.50, 0.75, 1, 2, 3, 4, 5 and 6h post drug. Blood was allowed to clot at room temperature for 1h. Serum separation and storage until analysis was as explained earlier. Following basal blood sample collection 2g of pure curcumin powder (4 capsules of 500mg curcumin each) or 2g of pure curcumin powder combined with 20mg of pure piperine powder (4 capsules of 500mg curcumin + 5mg piperine each; identical capsules prepared by Sami Chemicals and Extracts, Bangalore, India) was given with 150ml of water. Blood sampling after curcumin per se and curcumin with piperine was done two occasions, separated by a two week wash out period on the same volunteers. The following precautions were taken
  • 50. during the trial; subjects refrained from smoking, consuming alcohol, or beverages, and from taking drugs of any kind 24h prior to and during the trial. Standard meals were given to all the participants on the day of the test. Analytical methods Estimation of curcumin was done by reverse phase high pressure liquid chromatography (HPLC) using modification of the method described by Tannesen et al. (10). The modification was done by Sami Chemicals and Extracts, Bangalore, India, and is detailed below, the mobile phase used was ethanol : methanol (60 : 40) instead of only ethanol and the flow rate was changed from 1.2ml/min to 1ml/min. HPLC grade methanol and low actinic glassware protected from light were used for the entire procedure. Extraction and preparation of standard solution Curcumin (25mg) was dissolved and diluted to 25ml with methanol in a volumetric flask; 0.1ml (100 µm) of this was transferred to a volumetric flask and diluted with methanol up to 10ml making a 10ppm solution; 0.1ml of this 10ppm solution was transferred to another 10ml volumetric flask and the volume make up with methanol making a 0.1ppm solution. Extraction of curcumin from serum and preparation of sample Serum samples stored t -20°C were equilibrated to room temperature before analysis. A portion of 1ml was transferred into a 10ml volumetric flask and about 5ml of methanol added. The mixture was shaken thoroughly and heated at 80°C on a water bath for half an hour. After cooling to room temperature, methanol was added to make
  • 51. up the volume to 10ml and mixed well. The turbid solution was transferred into a 15ml centrifuge tube and centrifuged at 4000 RPM for 10 minutes. The supernatant was collected by means of a 25ml syringe and 10cm needle (Luer lock) and the clear solution filtered through a 0.45µm, 13mm Millipore membrane filter, into a narrow end test tube. 20µl of the solution were injected into the chomatograph for carry out the HPLC analysis. Samples were read by UV absorbance of 254nM. The recovery rate experiments were carried out by adding a known amount of standard curcumin to the serum and the added curcumin extracted as per procedure and quantified. The recovery rate of curcumin from serum ranged from 87 – 89.9%. The minimum level of detection of curcumin was 0.001µg/ml. Calculation Content of curcumin in µg/ml in the test sample Standard Reading x standard concentration Standard Reading x standard concentration Treatment of pharmacokinetic data For calculation of pharmacokinetic parameters (PK), curve fitting was carried out by a model independent method with non-linear least-square regression analysis using a computer designed programme “PHARMKIT”. This programme uses an algorithm call “SIMPLEX” for calculating non-linear least-squares. The various PK parameters calculated were: absorption half-life (t½(a)), elimination half live (t½el), volume of distribution (Vd); and clearance (CI). Areas under the concentration time curve (AUCo-m) was
  • 52. calculated using the trapezoidal method. Maximum concentration (Cmax) and time to max (Tmax) are observed values. Relative bioavailability (F) was calculated using formula: AUC Curcumin + piperine AUC Curcumin Statistical analysis Serum concentration time curves and the PK parameter from animal data were analysed using the Students ‘t’ test while the paired ‘t’ test was used for comparing serum concentration curves in humans. PK parameters of curcumin when given alone in humans could not be calculated as curcumin could not be detected in most of the samples. Planta Med. 64 (1998) Guido Shoba, David Joy, Thangam Joseph, M. Majeed. R. Rajendran, and P.S.S.R. Srinivas = F = x 100 Results Animal studies Curcumin alone at 2g/kg or when combined with piperine,
  • 53. 20mg/kg, was well tolerated by the rats as they showed no untoward effects for 48h. Yellow coloured faecal pellets appeared at 30h post drug and continued up to 48h. Perusal of Figure 1 indicates that when curcumin was given alone, peak serum concentrations of 1.00 ≠ 0.26 µg/ml were attained rapidly within 0.75h and plateaued till 1h. Thereafter, the levels declined gradually reaching zero at 5h. The plasma concentration time curve of curcumin in combination with piperine followed a similar pattern from 0 to 0.75h and 3 to 5h. However piperine produced higher serum concentrations of curcumin at 1 and 2 h (1.55 ± 0.21 and 1.50 ± 0.25 µg/ml) respectively, being significantly higher (P <0.02) at 2h. Thus piperine significantly enhanced the serum concentration of curcumin, albeit for a limited duration (although serum samples were collected up to 6h, values are depicted till 5h only, since the 6h value was also ‘0’ in all animals). Table 1 shows the values (mean ± SEM) of the pharmacokinetic parameters if curcumin per se and when combined with piperine. Cmax was increased from 1.35 ± 0.23 to 1.80 ± 0.16 µg/ml, but was not statistically significant, while Tmax was significantly increased from 0.83 ± 0.05 to 1.29 ± 0.23h (P <0.02). The t½(el) significantly decreased from 1.70 ± 0.58 to 1.05 ± 0.18h (P<0.002). Though t½(a) increased from 0.31 ± 0.07 to 0.47 ± 0.03h and AUC increased from 2.36 ± 0.28 to 3.64 ± 0.31 µg/h/ml, these increases were not statistically significant. CI significantly decreased from 713.00 ± 12.00 to 495.00 ± 37.00:/h (P<0.02) but the decrease in the Vd from 1366.00 ±248.70 to 782.60 ± 193.90L/kg was not significant. The relative bioavailability of curcumin when combined with
  • 54. Piperine is 154%. Human volunteer studies Curcumin alone or when combined with piperine was well tolerated by all the subjects and there were no adverse or untoward reactions; 2 subjects dropped out of the study for non-medical reasons. Therefore all calculations presented here are based on the data obtained from 8 subjects. In figure 2 is shown the serum concentration if curcumin per se and when given with piperine. Although serum samples were collected up to 6h, we have depicted values till 3h, since the 4, 5 and 6h values were also ‘0’ in all subjects. Serum levels’ of curcumin when given alone were either very low or undetectable at mot time points in most subjects, explaining he almost flat serum concentration curve (Fig. 2). However, when piperine was added the serum concentrations of curcumin were significantly increased at the time points up to 0.75h; P <0.01 Planta Med. 64 (1998) Guido Shoba, David Joy, Thangam Joseph, M. Majeed. R. Rajendran, and P.S.S.R. Srinivas Fig. 1 Serum concentrations µg/ml (mean ± SEM) of curcumin 20g/kg oral alone and with piperine 20mg/kg in rats (n= 6/group/time cut). Significance as compared to curcumin alone; *P <0.02. Fig. 2 Serum concentrations µg/ml (mean ± SEM) of curcumin 2g oral alone and with piperine 20mg in humans
  • 55. (n= 8). Significance as compared to curcumin alone; *P <0.01 **P <0.001. at 0.25h and 0.5h; P <0.001 and at 0.75h. Subsequently there was a rapid decline up to 1h and thereafter a gradual decline to zero by 3h. In Table 2 are depicted the PL parameters (Mean ± SEM) of curcumin when given alone and with piperine. Cmax (observed values) when curcumin was given alone was only 0.006 ± 0.005 µg/ml at 1h whereas when piperine was added the Cmax (observed value) was increased to 0.18 ± 0.16 µg/ml and was attained earlier, i.e. at 0.75h. Vd and CI could not be calculated with curcumin alone as serum levels were not detected at most time points in most subjects. The mean AUC(0-tn) however, was calculated using the trapezoidal method and was found to be 0.004 µg/ml, the relative bioavailability of curcumin when given with piperine was therefore 2000%. Discussion The results obtained in the study demonstrate that piperine enhances the oral bioavailability of curcumin in both rats and humans at does that we devoid of adverse side effects. However, certain differences between rats and human with respect to curcumin were evident. Curcumin per se attained overall moderate serum concentrations over a 4h period in rats with peak levels occurring between 0.75h to 1h. On the other hand, in humans when curcumin was given alone only negligible serum concentrations of curcumin were detectable the serum concentration-time curve being
  • 56. almost flat. This difference may be due to high oral dose employed in the rat (2g/kg), whereas the human does was about 60 times less, approximately 33mg/kg. Curcumin serum concentrations reached zero at 5h in rats and 3h in humans. Further in rats with the addition of piperine, curcumin achieved high concentrations than in humans albeit for a short period, took s longer time to peak and declined slowly. Whereas in humans Tmax was attained earlier and then declined rapidly. This rapidity in decline is more apparent probably because of the high levels of curcumin achieved with piperine as compared to curcumin alone. There was an increase in the AUC though not significant as an increase in bioavailability of curcumin by about one and a half times as compared to curcumin given alone in both rats and humans. In rats when piperine was added to curcumin both Vd and CI decreased which may have also contributed to the higher concentration, such a comparison was not possible in humans for season explained earlier. Our findings concerning absorption f curcumin in rats are in agreement with data obtained by Wahlstrom and Belnnow (11), who sowed that when Sprague Dawley rats were given curcumin 1g/kg p.o., measurement of blood plasma levels and biliary excretion indicated some absorption from the gut with no apparent toxic effect unto 5g/kg p.o. Likewise, Khanna et al. (12); found that after curcumin, 100mg/kg p.o., 74% was absorbed from the gastrointestinal tract within the first 5h, while complete elimination occurred within 48h. Our results are, however, in conflict with studies by Ravindranath and Chandrasekhara (6), who could not detect curcumin 400mg/kg p.o. They, however, did report 60% absorption of curcumin as determined by the amount excreted in the faeces. There is evidence that piperine is a potent inhibitor of drug metabolism, and glucuronidation altering the disposition
  • 57. and bioavailability of a large number of drugs (8). Further piperine at 20mg in humans has also been shown to produce earlier Tmax higher Cmax and AUC of drugs like propranolol and theophylline (13). This property of piperine suggests that it may be involved in inhibiting the metabolism of curcumin and enhancing bioavailability. In conclusion, the study shows that piperine enhances the serum concentration and bioavailability of curcumin in rats and man probably due to increased absorption and reduced metabolism. References 1. Kuhup, P.N.V. (1977) Handbook of Medicinal Plants, Vol. 1. Central Council for Research in Indians Medicine and Homeopathy (CCRIMH), New Delhi. 2. Srimal, R.C., Dhawan, R.N. (1985) in: Development of Unuani drugs from herbal sources and the role of elements in their mechanism of action. Hamdard National Foundation Monograph. New Delhi. 3. Ammon, H.R.T., Wahi, M.A. (1991) Planta Med. 57, 1-7. 4. James, J. S. (1993) AIDS Treatment News 176, 1-3. 5. Ravindranath, V., Chandrasekhara, N. (1980) Toxicology (Ireland) 16. 259-260. 6. Ravindranath, V., Chandrasekhara, N. (1981) Toxicology 20. 251-257. 7. Holder, G.M., Plummer, J.L., Ryan, A.J. (1978) Xenobiotica 8.
  • 58. 761-768. 8. Atal, C.K., Dubey, R.K., Singh, J.J. (1985) Pharmacol. Exp. Ther. 232. 258-262. 9. Singh, J., Dubey, R.K., Atal, C.K. (1985) J. Pharmacol. Exp. Ther 236. 488-493. 10. Tonnesen, H.H., Karlsen, J. (1993) J. Chromatography 259. 367- 369. 11. Wahlstrom, B., Blennow, G. (1978) Acta Pharmacol. Toxxicol. 43. 86-92. 12. Khanna, M., Singh, S., Sarin, J.P.S. (1981) Indian Drugs 19. 31. 13. Bano, G., Raina, R.K., Zutshi, U., Bedi, K.L., Johri, R.K., Sharma, S.C. (1991) Eur. J. Clin. Pharmacol. 14. 615-617. Planta Med. 64 (1998) Guido Shoba, David Joy, Thangam Joseph, M. Majeed. R. Rajendran, and P.S.S.R. Srinivas Dr Guido Shoba Department of Pharmacology St. John’s Medical College Bangalore 560 034 India