Download Pharmacokinetics and bioavailability of three promising tilmicosin

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Particle-size distribution wikipedia , lookup

Transcript
Pharmacokinetics and bioavailability of three promising tilmicosin-loaded lipid
1
nanoparticles in comparison with tilmicosin phosphate following oral
2
administration in broiler chickens
3
4
Ali RASSOULI1,*, Alwan AL-QUSHAWI1, Fatemeh ATYABI2, Seyed Mostafa
5
PEIGHAMBARI3, Mehdi ESFANDYARI-MANESH4, Gholam Reza SHAMS1
6
7
1
Department of Pharmacology, Faculty of Veterinary Medicine, University of Tehran,
8
Tehran, Iran.
9
Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical
10
Sciences, Tehran, Iran.
11
Department of Avian Diseases, Faculty of Veterinary Medicine, University of Tehran,
12
Tehran, Iran.
13
Nanotechnology Research Center, Tehran University of Medical Sciences, Tehran,
14
Iran
15
2
3
4
16
17
*Correspondence: [email protected]
18
19
20
21
22
23
24
1
1
Abstract: Tilmicosin (TLM) is a semi-synthetic antimicrobial agent used mainly in
2
poultry and cattle but it has relatively poor oral bioavailability. This study was
3
conducted to compare the bioavailability (BA) and main pharmacokinetic (PK)
4
parameters of TLM after oral administration of tilmicosin phosphate (TLM-PH) and
5
three newly prepared lipid nanoparticles (LNPs) of TLM including solid lipid
6
nanoparticles (SLNs), nanostructured lipid carriers (NLCs) and lipid-core nanocapsules
7
(LNCs). Sixty broiler chickens were divided into eight groups. In four treatment groups
8
(n = 10), each bird was given a single oral dose (20 mg/kg) of a TLM formulation after
9
an overnight fasting and in four control groups (n = 5), the vehicles of those
10
formulations or distilled water were given. Plasma TLM concentrations were analyzed
11
using an HPLC method and the related PK parameters (Cmax, Tmax, AUC0–∞, t1/2, kel,
12
ClB/F, MRT and Vd/F) were obtained by non-compartmental analysis. The relative
13
bioavailability (Rel. BA) of TLM-SLNs, TLM-NLCs and TLM-LNCs were 1.7, 2.7
14
and 3.6 times, respectively, more than BA of TLM-PH. Mean Cmax values were 1.21,
15
1.58, 1.76 and 2.17 μg/ml for TLM-PH, TLM-SLN, TLM-NLC and TLM-LNC,
16
respectively. In conclusion, TLM-LNPs improved drug BA and PK parameters
17
especially TLM-LNC formulation which suggests an efficient delivery system for
18
TLM.
19
Key words: Tilmicosin, lipid nanoparticles, pharmacokinetics, bioavailability, chicken
20
21
22
23
24
2
1
1. Introduction
2
Tilmicosin (TLM) is a broad-spectrum macrolide antibiotic derived from tylosin which
3
is used in animals only. It has been approved for the treatment and control of
4
respiratory diseases associated with Mycoplasma gallisepticum, Mycoplasma synoviae,
5
and various bacteria such as Staphylococcus spp. and Pasteurella multocida in broiler
6
chickens. Generally, gram negative Enterobacteriaceae are resistant to TLM (1-4).
7
TLM is poorly water-soluble especially in basic mediums. At present, soluble TLM-PH
8
is used in veterinary medicine as oral solution but this form has problems of low
9
potency and low BA. High doses of TLM may enhance its efficacy but posing the risk
10
of acute cardiac toxicity since the severity of TLM toxic effects is dose-dependent (5,
11
6). Given these disadvantages, studies on new delivery systems for TLM are warranted.
12
Oral administration of drugs is considered as the easiest and the most practical route
13
but the gastrointestinal (GI) epithelium acts as a physical barrier and may reduce drug
14
absorption and produce poor oral BA. To overcome these problems, a number of new
15
delivery systems have been developed (7, 8). Oral BA of drugs is highly effected by
16
their solubility and permeability, the most important physicochemical parameters
17
which determine drug absorption (8). On the other hand, the BA of a drug usually
18
determines its therapeutic efficacy because it may affect the onset, intensity and
19
duration of action of the drug (1).
20
Lowering particle size has revealed promise for increasing the dissolution of drugs as
21
well as their BA since it can facilitate the delivery of drugs at the right place and time.
22
Nanoparticle (NP) delivery systems can be prepared by using biodegradable materials
23
such as lipids (5). These nanoparticles which should be stable and non-toxic can
24
3
improve the efficacy and safety of loaded drugs (8). The size and surface properties of
1
NPs highly affect their cellular internalization (9). In recent decades, the lipid-based
2
nanoparticles (LNPs) have got special interests due to the use of natural or synthetic
3
lipids in their formulation which demonstrate high drug biocompatibility and controlled
4
release characteristics.
5
Nanoparticles can augment drug absorption by enhancing its dissolution, decreasing
6
gastric emptying rate and improving drug intestinal permeability. Lipids are recognized
7
as agents which increase lymph formation and encourage lymph flow (10). Basically,
8
the body takes up the lipid and solubilized drug at the same time. Therefore, it can be
9
considered as a kind of “Trojan horse” effect (11).
10
Oral LNPs are able to assist drug dissolution and solubilization because of their ability
11
to protect drugs in a solubilized condition and facilitate their mixing with GI
12
solubilizers such as bile acids. Furthermore, the protective effect of LNPs along with
13
their sustained release properties save drugs from degrading conditions and improve
14
their stability in the gastrointestinal tract. The nanoscale range of LNPs facilitates their
15
absorption into microfold cells (M cells) of Peyer’s patches and eventually into the
16
lymphatic system, so contributes to bypass the first-pass metabolism (12).
17
Lipid nanoparticles including SLNs, NLCs and LNCs are colloidal carriers composed
18
of a lipid matrix that is solid or liquid at body temperature. The efficiency of LNPs is
19
highly influenced by their composition which are usually composed of lipids,
20
surfactants as well as their structures which affect the release properties (13).
21
Solid lipid nanoparticles were incorporated as a novel oral drug delivery system in the
22
1990s. Loading poorly water-soluble drugs into SLNs can improve their GI
23
4
solubilization, absorption, and BA and provide controlled release property as well (9,
1
14).
2
Nanostructured lipid carriers are considered as second generation of LNPs which
3
incorporate biocompatible solid lipid matrix and oily lipid (15). These carriers have
4
demonstrated high BA with various routes of administration such as oral route (7).
5
Lipid-core nanocapsules are a hybrid nanocarrier system with vesicular structure
6
composed of polymer and lipid. These nanocapsules have great qualities as drug
7
delivery systems for oral route and are able to increase solubility of lipophilic drugs,
8
shield them from enzymatic degradation, enhance drug BA and decrease their side
9
effects (16, 17). In addition, LNCs are more appropriate for prolonged release (18).
10
Lipid-core nanocapsules are different from other formulation by their composition
11
since LNCs have three main components including drug, lipid, and polymer. Proper
12
interactions between these components have a vital role in successful manufacturing
13
and efficacy of LNCs (17, 19).
14
The objective of the present study was to perform a PK analysis and to compare the BA
15
of newly designed oral TLM-LNP formulations as potential new delivery systems for
16
TLM in chickens. The main PK parameters of these LNP formulations were compared
17
with the conventional TLM-PH formulation and the possible mechanisms were
18
discussed.
19
2. Materials and Methods
20
2.1. Chemicals and drug formulations
21
Tilmosin® (tilmicosin phosphate aqueous solution, 250 mg/ml) were provided from
22
Rooyan Darou, Pharmaceutical Company (Semnan, Iran) and tilmicosin standard (TLM
23
content 97.3%) was kindly donated by Razak Pharmaceutical Company (Tehran, Iran).
24
5
TLM-SLN, TLM-NLC and TLM-LNC powders re-dispersed in distilled water to reach
1
TLM concentrations at 250 mg/ml of TLM. These TLM-LNP formulations were
2
prepared and their physiochemical properties and antibacterial activities were evaluated
3
at nanotechnology center in the Faculty of Pharmacy of Tehran University of Medical
4
Science (TUMS). In vitro antibacterial testing was carried out in Laboratory of Avian
5
Microbiology and Laboratory of Pharmacology in the Faculty of Veterinary Medicine
6
(FVM). The LNP preparations were in nanoscale range with suitable properties as
7
shown in Table 1 (the original article is in press).
8
TLM-PH and TLM-LNP formulations were diluted with distilled water to a final
9
concentration of 25 mg/ml prior to oral administration to chickens.
10
2.2. Experimental Animals
11
Sixty apparently healthy broiler chickens, aged 35 days and 1.0-1.2 kg B.W., were
12
obtained from the poultry farm of the Faculty of Veterinary Medicine (FVM),
13
University of Tehran. The broilers were housed in cages with at 12 h dark/light cycle.
14
Temperature was maintained at 25 ± 2 °C and humidity at 45 – 65% with free access to
15
a balanced feed and water. The birds were monitored for one week for any apparent
16
clinical signs and adaptation to study area before administration of the drugs. This
17
study was approved by the ethics committee of FVM, Project Ethics No. 7506006-6-10.
18
.
19
2.3. Drug Administration
20
The chickens were randomly divided into eight groups, four test groups (n = 10) and
21
four control groups (n = 5). Birds were deprived from feed for 12 h prior to drug
22
administrations and for 6 h after drug dosing but with free access to water. The test
23
groups were given a single oral dose of 20 mg/kg of TLM-PH and TLM-LNP
24
6
formulations equivalent to 20 mg/kg of TLM for TLM-SLN, TLM-NLC and TLM-
1
LNC, respectively. Meanwhile, the control groups received equal volumes of distilled
2
water or SLN, NLC and LNC vehicles (blanks). Oral administration was done directly
3
into the middle of esophagus using a gavage attached to a syringe following the zero
4
time-point blood sampling.
5
2.4. Blood sampling
6
Blood samples (about 1.5 ml) were collected from the brachial or jugular vein into
7
sterile heparinized tubes prior to administration of different formulations (0 h) and at 1,
8
2, 4, 8, 12, 24, 48, 72, 96, and 120 h post administration. Within 1 h after sample
9
collection, the blood samples were centrifuged at ∼3500 rpm for 5 min. The harvested
10
plasma samples were stored at − 20°C until further use.
11
2.5. Sample preparation
12
To prepare plasma sample, 50 μl of perchloric acid was added to 950 μl of each chicken
13
plasma, vortexed for 30 seconds and centrifuged at ∼3500 rpm (Eppendorf Centrifuge,
14
Model 5810 R, Germany) for 5 min. The supernatant was transferred into special glass
15
tube and 20 𝜇l of each sample was injected into the HPLC system for analysis (1).
16
2.6. HPLC analysis
17
TLM concentrations in plasma were measured by using an HPLC system (Waters,
18
USA) which consisted of a multi-solvent pump, solvent degasser, UV detector,
19
autosampler, interface and Chromate software. The HPLC column was C18 (5 μm
20
particle size, 125× 4.6 mm). The modified methods of Clark et al. (2009) and Eraslan
21
(2007) were used for determination of TLM concentrations in plasma. HPLC analysis
22
was conducted using a mobile phase consisting of 0.2 M ammonium acetate (pH 5),
23
water, acetonitrile, and methanol (20:32:24:24). Mobile phase were filtered under
24
7
vacuum through a 0.45 μm membrane filter. Chromatographic separation was achieved
1
at a flow rate of 1ml/min using UV detection at 291 nm (3, 20).
2
TLM stock solution of 1.0 mg⁄ml was prepared by adding 10 mg of TLM standard in 10
3
ml of acetonitrile: water (1:1, v/v). Then it was further diluted in chicken plasma to
4
yield 0.01, 0.05, 0.1, 0.5, 1 and 5 μg/ml.
5
The HPLC method for TLM in chicken plasma was validated by assessing the linearity,
6
accuracy, precision, recovery, selectivity and sensitivity according to performance
7
criteria for method validation (3). Standard calibration curve was provided by using six
8
concentrations of TLM (0.01 - 5 μg/ml) and it was used for calculation of TLM levels
9
in plasma samples.
10
2.7. Pharmacokinetic Analysis
11
TLM plasma concentration data of each bird were used to depict its concentration-time
12
profile. The maximal plasma concentration of drug (𝐶max) and the time to reach 𝐶max
13
(𝑇max) were directly obtained from the observed concentration versus time profiles.
14
Non-compartmental analysis was used to estimate the PK parameters (AUC0–∞, t1/2,
15
Vd/F, ClB/F, kel, and MRT). The linear trapezoidal rule was used to calculate areas
16
under concentration-time curves from 0-120 h (AUC0-120) and from 120 h to infinity
17
(AUC120–∞) using the following equation: AUC120–∞ = last Cp/ kel. The Rel. BA
18
calculated by using the following equation and PK parameters obtained using Excel
19
2013.
20
𝑅𝑒𝑙. 𝐵𝐴 =
AUC (0 − ∞) of TLM − LNP formulation
AUC (0 − ∞) of TLM − PH formulation
21
22
2.8. Statistical Analysis
8
Data were expressed as mean±SD and analyzed with SPSS software (Ver. 19). The
1
differences in PK parameters were analyzed by using one-way ANOVA and Tukey’s
2
test (P < 0.05).
3
3. Results
4
The calibration curve for HPLC analysis of TLM was linear over the range of 0.01– 5
5
μg/ml as indicated by R² = 0.999. The calculated limit of detection (LOD) in chicken
6
plasma was 0.005 μg/ml and the limit of quantification (LOQ) was 0.015 μg/ml. At
7
TLM concentrations of 0.1, 1 and 5 μg/ml, the recovery rates were 99.6 ± 9.8%, 101 ±
8
7.5% and 100 ± 3.7%, respectively, and the precision of the method as expressed by
9
RSD% of inter-day and intra-day assays were (4.08, 3.30 and 3.46) and (3.06, 4.05 and
10
3.74), respectively.
11
TLM was well tolerated by the chickens without any noticeable event. The major PK
12
parameters and mean concentration-time profiles for TLM-PH, TLM-SLN, TLM-NLC
13
and TLM-LNC formulations were shown in Table 2 and Figure, respectively. There
14
was no detectable peak corresponding TLM retention time in the plasma samples of
15
control groups.
16
The Cmax mean value of TLM-LNC and other TLM-LNPs formulation was significantly
17
higher than that of TLM-PH (P < 0.05). In general, the mean values of AUC0–∞, t1/2, kel,
18
ClB/F, Vd/F and MRT for various TLM-LNP formulations were significantly different
19
from those of TLM-PH (P < 0.05). The AUC0–∞ values after oral administration of
20
TLM-LNPs were significantly higher than TLM-PH (P < 0.05).
21
4. Discussion
22
9
In the present study, we compared TLM-loaded NPLs with its conventional
1
formulation, TLM-PH. It was found that all TLM-loaded NPLs had significantly higher
2
systemic BA than the TLM-PH.
3
There are many possible mechanisms for increased oral BA of TLM by using LNPs
4
formulations. In general, LNPs are incorporated solid or liquid lipids similar to the fat
5
existing in food. Lipids can stimulate secretion of gastric-pancreatic lipases and co-
6
lipases. Consequently, according to their residence time, a large amount of ingested
7
lipids is already analyzed in GI (15). The absorption of fatty acids or mono and
8
diacylglycerides which are available in LNP formulations or made following digestion
9
by GI lipase may facilitate oral absorption of drugs (21). Besides, the release of biliary
10
lipids and salts is stimulated, which in turn enhance the production of mixed micelles,
11
which include solubilized drug molecules. As a result, by participation in mixed
12
micelles, TLM-LPNs could be better absorbed through lymphatic system (15). The
13
relatively lower absorption of TLM-SLN and TLM-NLC formulations in comparison
14
with TLM-LNCs may be referred to lower ability of young chickens to digest long
15
chain fatty acid (C14 to C24) existing in hydrogenated castor oil and Compritol 888
16
ATO. In contrast, the higher BA of TLM-LNC formulation may be due to higher
17
affinity of GI lipases in broilers for short and medium-chain fatty acids (C6 to C12),
18
which are the main components of its lipid, coconut oil (22).
19
The oral BA of TLM increased from 1.66 folds in TLM-SLNs to 3.61 folds in TLM-
20
LNCs, which correlated inversely with particle size of LNPs as it decreased from 193.0
21
± 2.64 nm (SLNs) to 116.6 ± 7.63 nm (LNCs). The nanoscale range of LNPs
22
formulations leads to decrease in particle size and highly increases their surface area
23
(15). In addition, the reduction in particle size is positively related to an adequate and
24
10
steady absorption of TLM in GI. In general, the increase in the bio-adhesion of these
1
LNPs formulations to the GI wall leads to prolong their residence time and their contact
2
with epithelial membranes, which improve their absorption (21).The reduction in
3
particles size, may also lead GI system to uptake them by other routes such as entry to
4
sub-mucosal tissues through intracellular pathways. The process of GI uptake may
5
include diffusion of particles through mucus and being more accessible to enterocyte
6
surface, epithelial interaction and cellular trafficking (7).
7
Negative surface charges (zeta potential) of TLM-LNP formulations can be considered
8
as another possible mechanism for enhancement of TLM oral bioavailability. In
9
general, glycocalix renders the intestinal mucosa a negative charge, which it will attract
10
positively charged nanoparticles. Therefore, the intestinal mobility of particles seems to
11
be highly related to their surface charges which is inversely related to particle surface
12
charge potentials. With negatively charged particles, it can be expected a higher
13
transport rates in comparison to near neutral or positively charged particles, whose
14
transport will be highly limited because of particle aggregation and electrostatic
15
adhesive interactions with intestinal mucin fibers. Increasing the efficiency of the
16
penetration through intestinal mucosa is important to improve oral delivery system.
17
Nanoparticles should be sufficiently small to keep away from severely steric inhibition
18
and adhesion to the intestinal fiber mesh. On the other hand, NPLs should have some
19
mucoadhesion to prolong their retention time and contact with intestinal mucosa (8). In
20
addition, surface charge of NPs also plays an important role in M cells uptake. For
21
instance, it has been reported that negatively charged NPs had higher M cell uptake
22
than positive charged NPs (23). However, the TLM-NLC formulation did not achieve
23
higher AUC values compared to TLM-LNC formulation in spite of showing higher ZP
24
11
(zeta potential) value (- 23.5 ± 1.13 mv), because the particles size and their surface
1
charge are working together in intestinal absorption process, and TLM-LNC had more
2
ideal characteristics with regard to its smaller particles size (116.6 ± 7.63 nm) with
3
optimal ZP value (- 16.3 ± 2.51 mv).
4
LNPs can also enhance absorption of TLM through lymphatic flow. They can induce
5
lipoprotein production and lymphatic lipid flux to augment the level of lymphatic TLM
6
transportation, which is significantly affected by the lipid and surfactants type (21). In
7
addition, the absorption by M-cells of Peyer’s patches is an additional way for
8
lymphatic transport of LNPs (12). The small size of LNPs allows more efficient
9
absorption particularly in the lymphoid system; consequently bypass the possible liver
10
first-pass metabolism (19). Although highly lipophilic compounds such as long chain
11
triglyceride lipids can easily reach systemic circulation by the lymphatic vessels,
12
nevertheless, particles size persists as the most vital factor in lymphatic absorption (24).
13
Particles with larger sizes may last longer in the Peyer’s patches, whereas smaller NPs
14
are transported directly to the thoracic duct, especially when the NPs coated by
15
polymers leading to be easily captured by lymphatic vessels (24). Therefore, it seems
16
that small particles size of TLM-LNCs with their polymeric structure are responsible
17
for enhancing lymphatic uptake and increasing their BA.
18
Furthermore, non-ionic surfactants such as Poloxamer 407 and Tween 80 are the other
19
factors which may increase the BA of TLM-LNC and TLM-NLC formulations due to
20
the ability of these surfactants to improve their intestinal permeability by disturbing the
21
cellular membranes and opening the tight junctions of intestinal epithelial cell (15) and
22
facilitating paracellular transportation of LNPs (10). Surfactants can also contribute to
23
the improvement of the affinity between LNPs and the intestinal epithelial membranes,
24
12
and enhance their bio-adhesion to the GI wall (19). These surfactants are favored for
1
oral formulations and efficiently reduce the degradation of LNPs in GI. The
2
polyethylene oxide (PEO) chains in these surfactants hamper the anchoring of the
3
lipase/co-lipase complex that is in charge of lipid degradation. By providing sterically
4
stabilizing layers with different thicknesses of PEO chains on LNPs surfaces, the in
5
vivo degradation rate of lipid matrix can be adjusted and slowed down and given time
6
for the LNPs to be absorbed (13). In addition, the Tween 80 can increase the intestinal
7
uptake due to its ability to inhibit the p-glycoprotein efflux pump (25). The results of
8
the present study regarding the higher efficiency of LNCs in improvement PK
9
properties are in line with findings of Bendera et al. (2012) who reported that Tween 80
10
can be used to deliver drugs efficiently to the brain and to inflamed tissues (16).
11
The presence of both surfactants, Poloxamer 407 and Tween 80 as ingredients of TLM-
12
LNCs and TLM-NLCs may have contributed to the quality of these formulations, and
13
as a result, increased their AUC and Cmax values much more than those of TLM-SLNs
14
and TLM-PH. These surfactants may be acted by decreasing the degradation of these
15
formulations, prolonging TLM release and enhancing their crossing through the
16
intestinal barrier.
17
Indeed, Eudragit S 100, which was used to coat TLM-LNCs, is a polymer with a pH-
18
dependent solubility. It releases the drug in the regions of GI with pH > 7 like large
19
intestine and colon (8), where it gets gradually soluble. Eudragit S 100 has been used to
20
entrap insulin to protect it from degradation by GI juices and to permit it to be released
21
in regions of the GI with pH > 7, where proteolytic enzymes are in low levels (26).
22
Therefore, Eudragit S 100 served as a potential oral carrier in TLM-LNC formulations
23
in the present study. In addition, Mohammadzadeh et al. (2014) demonstrated the
24
13
efficiency of Eudragit S 100 in decreasing P-glycoprotein activity and efflux process. It
1
seems that, this polymer improved the BA of TLM by dual actions (27). Generally, the
2
presence of a polymeric coating wall provides a protective layer against harsh
3
environment of the GI such as proteolytic enzymes and may prolong the exposure of
4
TLM-LNCs with intestinal epithelial cells, and consequently it may enhance BA of
5
TLM (17).
6
The relatively high BA of TLM-NLCs in the present study is in accordance with
7
findings of Aburahma and Badr-Eldin (2014), since a sustained release with little
8
degradation/aggregation behavior had been demonstrated by using Compritol 888 ATO
9
because of its long-chain fatty acids (23, 28). On the other hand, Severino et al. (2012)
10
reported that medium-chain triglycerides lipids are more effective than long-chain
11
triglycerides with regard to sustained release (11), which it seems more closely to the
12
results of the present study especially with regard to TLM-LNCs, in which coconut oil
13
comprising its oily core.
14
Although hydrogenated castor oil in TLM-SLNs was an effective nanoparticle system
15
for controlled release and improvement of PK characteristics of loaded drugs (14), it
16
achieved the least optimal PK parameters values. This may be due to its more rapid
17
degradation in GI which leads to an increased rate of TLM release (6). Many other
18
factors may have also contributed to decreased TLM-SLNs absorption such as their
19
relatively higher particles size (193.0 ± 2.64 nm), and less zeta potential (- 15.6 ± 3.21
20
mv). However, Han et al. (2008) suggested that the high initial release rate of TLM
21
from SLN might be helpful because it gets therapeutic level soon.
22
The TLM release by TLM-LNPs was slowed down just to reach therapeutic serum
23
levels so that the blood concentrations did not reach toxic levels and obviated adverse
24
14
effects such as cardio-toxicity induced by high doses of conventional TLM (6). Using
1
TLM-LNPs decreased TLM plasma elimination rate as indicated by higher elimination
2
of t1/2 and increased MRT, which caused a longer stay for TLM in blood circulation.
3
Abu-Basha et al. (2007) also studied the BA and PK parameters of TLM in chickens
4
using Provitil orally at 30 mg/kg B.W. The average AUC0−72 was 24.2 ± 3.9 (μg.h/ml),
5
Cmax (2.09 ± 0.37 μg/ml), Tmax (3.99 ± 0.84 h). In spite of using 1.5 fold higher dose,
6
the values reported for PK parameters are much lower than those of TLM-LNC
7
formulation especially with regard to AUC values (1).
8
Keles et al. (2001) also investigated the PK and tissue concentrations of TLM after oral
9
administration of a single dose (50 mg/kg, B.W.) in fowl. TLM was slowly eliminated
10
from the serum and lung with t1/2 of 30.2 ± 2.4 and 75.7 ± 3.7 h, respectively. The mean
11
Cmax was 6.2 times greater in the lung (7.96 ±0.30 μg/ml) than that in serum (1.28 ±
12
0.04 μg/ml) with Tmax at 4.66 ± 2.0 and 17.78 ± 7.51 h, respectively (29)
13
It is expected that TLM-LNCs followed by TLM-NLCs can demonstrate better in vivo
14
antibacterial activity due to their higher AUC, Cmax and sustained release properties
15
(14). In spite of their sub-MIC plasma concentrations against E. coli, it seems that
16
TLM-LNPs may be more active than TLM-PH, since clinical efficacy of TLM
17
formulations not only was affected by plasma drug levels but also it was related to
18
intracellular TLM penetration which usually tends to be more accumulated within avian
19
phagocytic cells (30), like macrophages, monocytes, and heterophils. Furthermore,
20
TLM has a high post-antibiotic and sub-MIC effects, which could slow disease
21
development by allowing immune system to eliminate bacterial infection (30). In
22
general, TLM Cmax values of all tested formulations which were detected in chicken
23
plasma after oral administration were higher than the MIC for M. gallisepticum and M.
24
15
synoviae (0.0125-0.1 μg/ml). These values were also higher than the MIC of plasma
1
TLM in cattle against Corynebacterium pyogenes (0.04 μg/ml) and S. aureus (0.78
2
μg/ml) (1). Consequently, the clinical efficacy of tested TLM-LNP formulations is
3
expected to be highly satisfactory with TLM-LNC formulation due to their better PK
4
parameters followed by TLM-NLC and TLM-SLN formulations.
5
In conclusion, systemic BA of TLM was significantly enhanced by using oral LNPs
6
(TLM-SLNs, TLM-NLCs and TLM-LNCs) in comparison to conventional TLM-PH in
7
broiler chickens. Newly formulated TLM-LNPs increased the AUC0–∞, Cmax, Tmax and
8
MRT values depending on their particle size, particle surface charges, as well as lipid
9
and surfactant compositions. The best BA results achieved by LNC formulation (3.6
10
fold) followed by NLCs (2.71 fold) compared to TLM-PH. It seems that the hybrid
11
delivery system (LNCs) is more promising to achieve a sustained release TLM
12
formulation with a higher antibacterial activity and lower drug toxicity.
13
14
15
16
Acknowledgement
17
The authors wish to thank the Nanotechnology Research Center of TUMS; A. Yazdani
18
in Avian Microbiology Laboratory of FVM, University of Tehran for her technical
19
assistance. They are also grateful to Razak Pharmaceutical Co. and Bachter
20
Pharmaceutical Co. for donating TLM standard and Eudragit S 100, respectively.
21
This study was partially supported by the Research Council of University of Tehran
22
(Grant No., 7506006-06-10).
23
24
16
References:
1
1. Abu-Basha EA, Idkaidek NM, Al-Shunnaq AF. Pharmacokinetics of tilmicosin
2
(provitil powder and pulmotil liquid AC) oral formulations in chickens. Vet Res
3
Commun 2007; 31: 477-485.
4
2. Avci T and Elmas M. Milk and blood pharmacokinetics of tylosin and tilmicosin
5
following parenteral administrations to cows. Sci World J 2014; Article ID 869096.
6
3. Eraslan G. Effects of different doses of tilmicosin on erythrocyte catalase activity
7
and plasma malondialdehyde levels in chicks. Bull Vet Inst Pulawy 2007; 51: 145-
8
147.
9
4. Jordan FTW and Horrocks BK. The minimum inhibitory concentration of
10
tilmicosin and tylosin for Mycoplasma Synoviae and a comparison of their efficacy
11
in the control of Mycoplasma gallisepticum infection in broiler chicks. Avian Dis
12
1996; 40: 326-334.
13
5. Chen X, Wang T, Lu M, Zhu L, Wang Y, Zhou W. Preparation and evaluation of
14
tilmicosin-loaded hydrogenated castor oil nanoparticle suspensions of different
15
particle sizes. Intl J Nanomed 2014; 9: 2655-2664.
16
6. Han C, Qi CM, Zhao BK, Cao J, Xie SY, Wang SL, Zhou WZ. Hydrogenated
17
castor oil nanoparticles as carriers for the subcutaneous administration of
18
tilmicosin: in vitro and in vivo studies. J Vet Pharmacol Therap 2008; 32: 116-123.
19
7. Zhang T, Chen J, Zhang Y, Shen Q, Pan W. Characterization and evaluation of
20
nanostructured lipid carrier as a vehicle for oral delivery of etoposide. Eur J
21
Pharmaceut Sci 2011; 43: 174-179.
22
8. Plapied L, Duhem N, Des Rieux A and Préat V. Fate of polymeric nanocarriers for
oral drug delivery. Curr Opin Colloid Interface Sci 2011; 16: 228-237.
17
23
24
9. Beloqui A, Solinís MA, Gascón AR, Del Pozo-Rodríguez A, Des Rieux A, Préa
1
VT. Mechanism of transport of saquinavir-loaded nanostructured lipid carriers
2
across the intestinal barrier. J Controlled Release 2013; 166: 115-123.
3
10. Tiwari R and Pathak K. Nanostructured lipid carrier versus solid lipid nanoparticles
4
of simvastatin: Comparative analysis of characteristics, pharmacokinetics and tissue
5
uptake. Intl J Pharmaceut 2011; 415: 232- 243.
6
11. Severino P, Andreani T, Macedo AS, Fangueiro JF, Santana MHA, Silva AM,
7
Souto EB. Current state-of-art and new trends on lipid nanoparticles (SLN and
8
NLC) for oral drug delivery. J Drug Delivery 2012; Article ID 750891.
9
12. Battaglia L and Gallarate M. Lipid nanoparticles: state of the art, new preparation
10
methods and challenges in drug delivery. Expert Opin Drug Deliv 2012; 9(5): 497-
11
508.
12
13. Shah R, Eldrige D, Palombo E, Harding I. Composition and Structure. In: Lipid
13
nanoparticles: Production, Characterization and Stability. Springer Briefs in
14
Pharmaceut Sci Drug Develop 2015; 319-10710-3.
15
14. Xie S, Zhu L, Dong Z, Wang Y, Xiaofang Wang X, WenZhong Zhou WZ.
16
Preparation and evaluation of ofloxacin-loaded palmitic acid solid lipid
17
nanoparticles. Intl J Nanomed 2011; 6: 547-555.
18
15. Zhuanga CY, Li N, Wang M, Zhang XN, Pana WS,Peng JJ, Pana YS, Tang X.
19
Preparation and characterization of vinpocetine loaded nanostructured lipid carriers
20
(NLC) for improved oral bioavailability. Intl J Pharmaceut 2010; 394: 179-185.
21
16. Bendera EA, Adornec MD, Coloméa LM, Abdallab DSP, Guterresa SS, Pohlmann
22
AR. Hemocompatibility of poly(-caprolactone) lipid-core nanocapsules stabilized
23
18
with polysorbate 80-lecithin and uncoated or coated with chitosan. Intl J
1
Pharmaceut 2012; 426: 271- 279.
2
17. Cattania VB, Fiel LA, Jager A, Jager E, Colome LM, Uchoa F. Lipid-core
3
nanocapsules restrained the indomethacin ethyl ester hydrolysis in the
4
gastrointestinal lumen and wall acting as mucoadhesive reservoirs. European J
5
Pharmaceut Sci 2010; 39: 116-124.
6
18. Fiel LA, Contri RV, Bica JF, Figueiró F, Battastini AMO, Guterres SS. Labeling
7
the oily core of nanocapsules and lipid-core nanocapsules with a triglyceride
8
conjugated to a fluorescent dye as a strategy to particle tracking in biological
9
studies. Nanoscale Res Letters 2014; 9: 233.
10
19. Li H, Zhao XB, Ma YK, Zhai GX, Li LB, Lou HX. Enhancement of
11
gastrointestinal absorption of quercetin by solid lipid nanoparticles. Journal of
12
Controlled Release 2009; 133: 238-244.
13
20. Clark CR, Dowling PM, Boison JO. Development and validation of a method for
14
determination of tilmicosin residues in equine plasma and tissues using HPLC. J
15
Liquid Chromatogr Related Tech 2009; 32: 2839-2856.
16
21. Qi J, Lu Y, Wu W. Absorption, disposition and pharmacokinetics of solid lipid
nanoparticles. Curr Drug Metab 2012; 13: 418-428.
22. Smink W. Fatty acid digestion, synthesis and metabolism in broiler chickens and
pigs. PhD, Wageningen University, Wageningen, Netherlands, 2012.
23. Harde H, Das M, Jain S. Solid lipid nanoparticles: an oral bioavailability enhancer
vehicle. Expert Opinion on Drug Delivery 2011; 8: 1407-1424.
19
17
18
19
20
21
22
24. Manjunath K and Venkateswarlu V. Pharmacokinetics, tissue distribution and
1
bioavailability of clozapine solid lipid nanoparticles after intravenous and
2
intraduodenal administration. J Controlled Release 2005; 107: 215-228.
3
25. Rege BD, Kao JP, Polli JE: Effects of nonionic surfactants on membrane
transporters in Caco-2 cell monolayers. Eur J Pharm Sci 2002; 16: 237-246.
26. Jain D, Panda AK, Majumdar DK. Eudragit S 100 entrapped insulin microspheres
for oral delivery. AAPS Pharm Sci Tech 2005; 6: 100-107.
4
5
6
7
27. Mohammadzadeh R, Baradaran B, Valizadeh H, Yousefi B, Zakeri-Milani P.
8
Reduced ABCB1 expression and activity in the presence of acrylic copolymers.
9
Adv Pharmaceut Bulletin 2014; 4: 219-224
10
28. Aburahma MH and Badr-Eldin SM. Compritol 888 ATO: a multifunctional lipid
11
excipient in drug delivery systems and nano-pharmaceuticals. Expert Opin Drug
12
Deliv 2014; 11: 1865-1883.
13
29. Keles O, Bakirel T, Sener S. Pharmacokinetics and tissue levels of tilmicosin in
fowls. Turk J Vet Anim Sci 2001; 25: 629-634.
14
15
30. Fricke JA, Clark CR, Boison JO, Chirino-Trejo M, Inglis TES, Dowling PM.
16
Pharmacokinetics and tissue depletion of tilmicosin in turkeys. J Vet Pharmacol
17
Therap 2008; 31: 591-594.
18
20
1
TLM
TLM-SLN
TLM-NLC
TLM-LNC
standard
Composition
TLM
Hydrogenated
Compritol 888 ATO
Eudragit S 100 as
(97.3%)
castor oil as a solid
as a solid lipid
polymer, Coconut
lipid matrix and
matrix, sesame oil as
oil as oil core lipid
Poly Vinyl
a liquid oil and
and Span 80,
Alcohol 5% (PVA) Poloxamer 407,
Tween 80 as
as a surfactant
surfactants
Tween 80 as a
surfactant
66.3 ± 2.67
86.5 ± 2.17
94.0 ± 3.60
Zeta potential mv
- 15.6 ± 3.21
- 23.5 ± 1.13
- 16.3 ± 2.51
Particle
193.0 ± 2.64
156.6 ± 7.63
116.6 ± 7.63
Entrapment
efficiency (%)
size
(mm)
In vitro release 97% drug
Initial burst
Initial burst release
Initial burst release
(h)
release
release (18%)
(15%) within first 2 h
(13%) within first 2
at pH 7.4
within 12 h
within first 2 h
followed by a
h followed by a
followed by a
constant sustained
constant sustained
constant sustained
release for 200 h
release for 200 h
release for 120 h
MIC
S. aureus
μg/ml E. coli
1
1
0.5
0.5
4
4
2
2
21
Table 1: Preparation, physicochemical properties and MIC against S. aureus and E.
1
coli of TLM-SLN, TLM-NLC, TLM-LNC formulations and TLM standard.
2
lipid
3
nanoparticles; NLC: nanostructured lipid carriers and LNC: lipid-core nanocapsule.
4
TLM-PH:
conventional
tilmicosin
phosphate
solution;
SLN:
solid
5
6
7
8
22
PK parameter
AUC0–∞(μg.h/ml)
TLM-PH
TLM-SLN
TLM-NLC
TLM-LNC
43.0 ± 2.5 d
71.5 ± 5.8 c
116.4 ± 17.8
155.1 ± 8.6 a
b
t1/2 β(h)
29.3 ± 2.6 c
39.8 ± 3.9 b
46.5 ± 5.0 a
41.3 ± 3.4 b
Vd/F (L/kg)
20.4 ± 2.0 a
10.3 ± 2.7 b
11.9 ± 1.5 b
7.84 ± 2.6 b
MRT (h)
42.0 ± 3.8 b
50.3 ± 5.8 a
67.2 ± 8.8 a
59.6 ± 0.1a
ClB/F (ml/(min.kg))
0.48 ± 0.10 a
0.29 ± 0.04 b
0.19 ± 0.05 c
0.14 ± 0.04 d
kel(h−1)
0.03 ± 0.01a
0.03 ± 0.01a
0.01 ± 0.01 c
0.02 ± 0.01 b
Tmax(h)
2.40 ± 0.24 c
5.60 ± 0.67 b
4.80 ± 0.80 b
12.00 ± 0.00 a
Cmax(μg/ml)
1.21 ± 0.09 b
1.58 ± 0.22 a
1.76 ± 0.38 a
2.17 ± 0.30 a
100
166.2
270.7
360.7
Rel.BA (F) %
1
2
3
Table 2: Pharmacokinetic parameters of TLM in chickens after oral administration of a
4
single dose of TLM-PH, TLM-SLN, TLM-NLC and TLM-LNC formulations (20
5
mg/kg body weight). Data are expressed as mean ± SD (n = 10). Data with different
6
letters (a, b, c, d) differ significantly (P < 0.05).
7
8
9
10
11
23
1
3
plasma concentration (μg/ml)
TLM-PH
2,5
TLM-SLN
TLM-NLC
2
TLM-LNC
1,5
1
0,5
0
0
20
40
60
80
100
120
140
Time (h)
2
Figure. Plasma concentration vs. time curves of TLM after a single oral administration
3
of TLM-PH, TLM-SLN, TLM-NLC and TLM-LNC formulations (20 mg/kg B.W) in
4
broiler chickens. Each value represents the mean and SD (n = 10)
5
6
7
8
24