You are on page 1of 11

Pharm Res (2019) 36:9

https://doi.org/10.1007/s11095-018-2538-7

RESEARCH PAPER

Physicochemical Characterization and Pharmacokinetics


of Agomelatine-Loaded PLGA Microspheres
for Intramuscular Injection
Hongjuan Zhang 1 & Chenguang Pu 1 & Qiao Wang 1 & Xinyi Tan 1 & Jingxin Gou 1 & Haibing He 1 &
Yu Zhang 1 & Tian Yin 1 & Yanjiao Wang 1 & Xing Tang 1

Received: 27 March 2018 / Accepted: 29 October 2018


# Springer Science+Business Media, LLC, part of Springer Nature 2018

ABSTRACT ABBREVIATIONS
Purpose The aim of this study was to design agomelatine AGM Agomelatine
loaded long acting injectable microspheres, with an eventual AUC Area under the curve
goal of reducing the frequency of administration and improv- Cmax Maximum concentration
ing patient compliance in treatment of depression. DALYs Disability adjusted life years
Methods AGM-loaded microspheres were prepared by an DAS Drug and statistics software
O/W emulsion solvent evaporation method. The physico- DSC Differential scanning calorimetry
chemical properties and in vitro performance of the micro- EMA European Medicines Agency
spheres were characterized. The pharmacokinetics of different FDA Food and drug administration
formulations with various particle sizes and drug loadings MDD Major depressive disorder
were evaluated. PLGA Poly (D,L-lactide- co-glycolide)
Results AGM-loaded microspheres with drug loading of PXRD Powder X-ray diffraction
23.7% and particle size of 60.2 μm were obtained. The SD Standard deviation
in vitro release profiles showed a small initial burst release SDS Sodium dodecyl sulfate
(7.36%) followed by a fast release, a period of lag time SEM Scanning electron microscopy
and a second accelerated release. Pore formation and T1/2 Plasma half-life
pore closure were observed in vitro, indicating that the Tmax Peak time
release of drug from microspheres is dominated by UPLC-MS/MS Ultra-performance liquid
water-filled pores. Pharmacokinetic studies showed that chromatography-tandem mass spectrometry
AGM microspheres could release up to 30 days in vivo at YLDs Years lived with disability
a steady plasma concentration. As well, particle size and
drug loading could significantly influence the in vivo re-
lease of AGM microspheres. INTRODUCTION
Conclusions AGM-loaded microspheres are a promising car-
rier for the treatment of major depressant disorder. Depression is a common heterogeneous disorder charac-
terized clinically by low spirit, mental retardation, and
exercise suppression. It has been estimated that depression
KEY WORDS agomelatine . PLGA microspheres . pore is expected to be the second highest cause of morbidity by
formation and pore closure . release in vitro . release in vivo 2020 (1). As well, patients with major depressive disorder
(MDD) in the risk of death from suicide, accident, coro-
nary heart disease, stroke and respiratory disease is in-
creasing (2–4). The estimated global calculated years lived
* Yanjiao Wang with disability (YLDs) and disability adjusted life years
TangLab@126.com (DALYs) of MDD are 8.2% (5.9-10.8%) and 2.5% (1.9-
3.2%) respectively (5). Furthermore, not only do patients
1
School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua with MDD suffer from increased serious physical illness,
Road, Shenyang 110016, China but they are also affected by decreased social function.
9 Page 2 of 11 Pharm Res (2019) 36:9

MDD has a high rate of relapse and high levels of dys- (2,7). Clinically, the use of agomelatine tablets requires
function, and so has serious consequences for both the liver function testing, which severely limits the use of the
depressed individuals and their families (6,7). At present, drug (9). Also, daily medication is particularly inconve-
the clinical treatment of depression is mainly drug based nient for people with MDD, as they often refuse to take
treatments, and the efficiency of the antidepressant drugs medicine because of psychological problems. Therefore, it
is only fully achieved when taken regularly and for a long is ideal to develop a depot-like system which can retain
period. It is recommended that the treatment cycle for the therapeutic concentration of agomelatine in plasma
depression is at least one year to minimize the risk of for a long time by releasing the drug in a controlled man-
recurrence (8,9). However, most studies have shown that ner for patients with MDD to ensure stable recovery and
more than 40% of patients discontinue the treatment to prevent relapse. Thus, promising polymeric micro-
within 1 month, and approximately 56% of patients dis- spheres can be applied to many hydrophobic drugs.
continue the treatment within 4 months (9). It is known With the development of drug delivery systems, depot-
that tolerability and psychological problems weaken the like sustained-release drug delivery systems including in-
capacity of antidepressants, and that a steady and jectable drug suspensions, oil-based injectable solutions,
sustained plasma concentration is critical for the treat- polymer-based in-situ formings and polymeric micro-
ment of depression (10,11). Hence, in order to achieve spheres that can release drugs longer than one week have
good clinical efficiency, it is of importance to develop a attracted much attention in the pharmaceutical field (14).
long-acting injection of antidepressants. These systems offer numerous advantages over traditional
Agomelatine (AGM, Fig. 1) is a novel small molecule drug delivery systems, including predictable drug release
antidepressant with melatonergic MT1/MT2 receptor ag- period, improved drug stability, reduced dosing frequen-
onist and 5-HT2C receptor antagonist activity. It was ap- cy, increased patient compliance, decreased side effects
proved by the EMA in 2009, and was thought to be able and an overall cost reduction of medical care (14).
to improve the sleep of people with MDD by synchroniz- Among various sustained-released drug delivery sys-
ing the circadian rhythms. It also showed improved toler- tems, polymeric microspheres, particularly PLGA–based
ability and a low propensity for drug withdrawl, sexual microspheres, are the most widely used and commercially
dysfunction and weight gain compared to other antide- successful due to their ability to encapsulate a variety of
pressants (3,6). According to its chemical structure, drugs, high bioavailability, biocompatibility and sustained
agomelatine is a water-insoluble small molecule drug. Its release (15). PLGA, certified by FDA to be used for drug
solubility increase with temperature and the effective con- delivery systems, not only has good biodegradability and
centration is very low due to its new mechanism of action biocompatibility, but can also tailor the release rate by
(12,13). At present, agomelatine is only commercially changing the molecular weight or the ratio of the two
available in oral tablets. However, various shortcomings monomers. Today, several commercial products are based
severely restrict clinical use of agomelatine tablets. First, on PLGA microspheres, including Risperdal Consta® (ris-
due to its short half-life (1–2 h) and severe first-pass effect peridone, Janssen, Inc.), Vivitrol® (naltrexone, Alkermes,
in the liver, orally administered agomelatine often shows a Inc.) and Leupron Depot® (leuprolide acetate, Abbot,
low absolute bioavailability (5% of oral dose), severe he- Inc.) and more (14,16). Many different types of processes
patic metabolism and further elevated aminotransferases are commonly employed for the preparation of polymer
microspheres, including emulsion-solvent extraction/evap-
oration, spray drying, interfacial polymerization and coac-
ervation (16,17). Until now, the preparation process and
many properties of PLGA microspheres including encap-
sulation efficiency, release and degradation rates have been
studied in detail (18–21). However, many obstacles hinder
the development of insoluble drug microsphere products.
On one hand, traditional emulsion solvent evaporation
techniques and spray drying techniques may result in pores
in the microspheres during the solvent removal process.
Too many drugs can then diffuse through the holes, lead-
ing to an unwanted burst release in the initial stage of
release. On the other hand, drug loading limitation is a
significant problem for insoluble drugs (22). Particularly
for hydrophobic drugs with relatively big water solubility,
Fig. 1 Chemical structure of agomelatine. it is difficult to achieve a good encapsulation. In addition,
Pharm Res (2019) 36:9 Page 3 of 11 9

potential toxicity caused by residual solvent inside the mi- Characterization of AGM-Loaded PLGA Microspheres
crospheres can be a concern (22).
In this work, in order to improve the bioavailability, reduce Particle Size and Morphology
the side effects caused by the first pass effect of the oral tablets
and achieve sustained release of AGM for the treatment of Particle size and distribution of AGM-loaded PLGA micro-
MDD, AGM-loaded PLGA microspheres were first devel- spheres were investigated by a laser diffraction particle size
oped using the traditional emulsion solvent evaporation meth- analyzer (BT-9300S, Bettersize Co., Ltd., Dandong, China).
od. Preparation, characterization, in vitro and in vivo release The microspheres were re-dispersed in distilled water prior to
properties of AGM-loaded PLGA microspheres were subse- analysis. The mean size and size distribution were expressed
quently evaluated in detail. by D50 (volume weighted mean diameter) and Span (Span
value = (D90-D10)/D50) respectively.
The surface morphology of the microspheres was exam-
MATERIALS AND METHODS ined using scanning electron microcopy (SEM) (SU8010,
Hitachi., Ltd., Japan). Freeze-dried microspheres were spread
Materials and Animals on double-sided conductive adhesive tape, which was previ-
ously attached to a copper stub, and then the sample was
Poly (D, L-lactide-co-glycolide) (PLGA) (LA/GA: 75:25, Mw: coated with a thin layer of gold under an argon atmosphere
70 kDa, carboxylic acid end group) was purchased from and observed by SEM. SEM photos of the in vitro release study
Jinandaigang Biomaterials (Jinan, China). Poly vinyl alcohol were also obtained.
(KURARAY POVAL 22–88, 87.0-89.0% hydrolyzed) was
purchased from Kuraray Co., Ltd. (Osaka, Japan). X-Ray Diffraction Analysis
Agomelatine (AGM) was provided by Benyuan Pharmacy
Co., Ltd. (Benxi, China). Dichloromethane (DCM), acetoni- To investigate the physical state of AGM within the micro-
trile and all the other reagents were of chromatographic spheres, AGM, blank microspheres, physical mixture of AGM
grade. and blank microspheres and AGM-loaded microspheres were
Male Sprague-Dawley rats, with the average weight of studied by powder X-ray diffraction (PXRD) (PANanalyical,
180–220 g, were obtained from the Experiment Animal Almelo, The Netherlands) using Nifiltered Cu Kα radiation.
Center of Shenyang Pharmaceutical University (Shenyang, The experiment was conducted at a voltage of 40 KV and
China). All procedures were performed according to the 30 mA. Samples were analyzed in a 2θ range of 5–60°.
guidelines issued by the Ethical Committee of Shenyang
Pharmaceutical University (SYPU-IACUC-C2017–0512- Differential Scanning Calorimetry
001). All efforts were made to minimize animal suffering and
to limit the number of animals used with the approval of the AGM, blank microspheres, physical mixture of AGM and
Animal Ethics Committee of Shenyang Pharmaceutical blank microspheres and microsphere products were studied
University. by differential scanning calorimetry (DSC) technique using a
METTLER TOLEDO DSC1 (METTLER TOLEDO,
Preparation of AGM-Loaded PLGA Microspheres Switzerland). 5 mg samples were placed in an aluminum
pan and heated at a rate of 10°C/min, using dry atmosphere
An O/W emulsion solvent evaporation technique was used to of nitrogen as carrier gas, across a temperature range of 30 to
prepare AGM-loaded PLGA microspheres (23,24). Briefly, 150°C.
300 mg PLGA and 129 mg AGM were dissolved in 1.5 mL
DCM to obtain an organic phase. The organic phase was HPLC-UV Analysis and Drug Loading
cooled to 4°C, slowly added to 30 mL pre-cooled 1% (w/v)
PVA aqueous solution and then stirred (Ultra-Turrax Quantitative analysis of agomelatine in various samples was
TP18/10, IKA, Germany) at a set speed for 2 min to form performed using a Hitachi high pressure liquid chromatogra-
the O/W emulsion. The obtained emulsion was added to phy (HPLC) system consisting of a Hitachi Chromaster 5110
120 mL distilled water to dilute the emulsion, and stirred at HPLC pump, a Hitachi Chromaster 5210 autosampler, a
300 rpm for 4 h to evaporate the solvent. The resulting mi- Hitachi Chromaster 5310 column oven and a Hitachi
crospheres were collected by centrifugation at 3000 rpm for Chromaster 5410 UV detector. For analysis, a C18
5 min, washed three times with distilled water to remove re- reversed-phase column (Haito Pack ODS, 250 × 4.6 mm,
sidual organic solvent and PVA, lyophilized (VirTis 5 μm) was used, with the mobile phase of acetonitrile:water
AdVantage Plus Bench Top Freeze Dryer, SP industries, 50:50 (v/v) and the flow rate was of 1 mL/min. UV detection
Inc., USA) and finally stored at 4°C. was performed at 230 nm.
9 Page 4 of 11 Pharm Res (2019) 36:9

A direct extraction method was used to determine the suspension) as a single dose of 2.6 mg/kg by mouth to
loading content and encapsulation efficiency of AGM in investigate the oral bioavailability and the minimum ef-
microspheres accurately. In brief, approximately 10 mg fective blood concentration of AGM in rats. Group 2,
microspheres were dissolved in acetonitrile and sonicated Group 3 and Group 4 received different AGM micro-
for 1 min. Then the samples were filtered through a spheres formulations (formulation MS1, MS2 and MS3
0.22 μm membrane and determined by HPLC. Drug respectively) by intramuscular injection at the right
loading and encapsulation efficiency were calculated using hind-leg muscle as a single dose of 10 mg/kg. At
the following equations: predetermined intervals, approximately 0.3 mL blood
samples were collected via the orbital vein with hepa-
weight of AGM in microspheres rinized tubes. The sampling time points for AGM solu-
Drug loadingð%Þ ¼
weight of microspheres tion was 10, 20, 30, 45, 60, 90, 120, 240, 360, 480,
 100 ð1Þ 720, 1440 and 2160 min after administration by mouth
and 0.5, 1, 2, 4, 8, 12 h and 1, 2, 3, 5,7, 9, 12, 15, 18,
21, 24, 27, 30 and 35 days for the three AGM micro-
sphere groups. The heparinized blood samples were
Encapsulation efficiencyð%Þ separated immediately by centrifugation at 4000 rpm
Actual AGM loading for 10 min and stored at −80°C until subsequent ex-
¼  100 ð2Þ traction and analysis.
Theoretical AGM loading
Plasma samples were processed as follows. First,
100 μL of plasma samples, 20 μL of indapamide
(500 ng/mL, internal standard) and 20 μL of acetonitrile
In Vitro Release and Degradation Evaluation
were mixed by vortex for 3 min. Next, 2 mL of ethyl
acetate were added to the mixture and vortexed for
The optimal formulation performance in vitro was investi-
10 min to extract the mixture. After centrifugation at
gated as follows. Briefly, 10 mg AGM-loaded micro-
10,000 rpm for 10 min, 1.5 mL of the organic layer were
spheres were suspended in 10 mL release medium (0.5%
removed and dried with nitrogen. Finally, after removal
SDS, 0.02% NaN3) (25,26). All the samples were incubat-
of the solvent, the residue was reconstituted with 100 μL
ed in a shaking water bath (Zhicheng Inc., China) at 37 ±
of mobile phase, and the plasma concentration of AGM
1°C and 100 rpm. At predetermined sampling points,
was d etermined u sing ultra p erformance liqu id
samples were centrifuged at 3000 rpm for 10 min
chromatography-tandem mass spectrometry (UPLC-MS/
(USTC chuangxing Co. Ltd. Zonkia Branch, China).
MS, Water Corp., Milford, MA).
9 mL supernatant were separated for the HPLC assay,
The obtained data were analyzed using a non-
and fresh medium with equal volume was added in the
compartmental method with drug and statistics (DAS) soft-
meantime. All release tests were conducted in triplicate.
ware (version 2.0, Mathematical Pharmacology Professional
The amount of released drug during the first 24 h was
Committee of China, Shanghai, China).
designated as Bburst release^. In order to evaluate its re-
lease mechanism and degradation, SEM was used to ob-
serve the microspheres at different time points during the
Storage Stability
release test.
A preliminary stability study of the optimized formulation
In Vivo Pharmacokinetic Study was carried out. Freeze-dried microspheres were kept at
4°C and room temperature, respectively. Samples were
The in vivo performance of AGM-loaded microspheres taken on 0 d, 7 d and 28 d to observe its fluidity, and
was investigated, to further examine the influence of par- their in vitro release were determined.
ticle size and drug loading on the in vivo release of AGM-
loaded microspheres. The pharmacokinetics of three
AGM-loaded microsphere formulations and AGM solu- Statistical Analysis
tion were investigated. The dosing of rats in the study
was calculated based on the oral dose of humans. A paired student’s t test was used for statistical analysis,
After 7 days acclimatization, rats were randomly di- with p < 0.05 considered the minimum level of signifi-
vided into four groups (n = 6). All animals were fasted cance. Pharmacokinetic parameters were obtained using
for 12 h with access to water prior to dosing. Group 1 a non-compartmental method with drug and statistics
received AGM solution (AGM carboxymethylcellulose (DAS) software.
Pharm Res (2019) 36:9 Page 5 of 11 9

RESULTS AND DISCUSSION prevent the drug from migrating to the water phase by
decreasing the solubility of AGM in water and increasing
Optimizing the Formulation of Microspheres the viscosity of the organic phase. In addition, it was re-
ported that increasing the viscosity of the organic phase
The traditional emulsification solvent evaporation method by increasing the polymer concentration or molecular
was employed to prepare AGM-loaded microspheres. The weight could increase the drug encapsulation (28). Thus,
parameters of the preparation process and formulation PLGA with a relatively high molecular weight and a high
were optimized by assessing major factors including drug concentration of 200 mg/mL was used to achieve high
loading, encapsulation efficiency, particle size distribution drug encapsulation and long duration of drug release.
and burst release (in terms of the cumulative drug release
after 1 day). In the preliminary study, it was found that a Characterization of AGM Microspheres
high encapsulation efficiency could be achieved by in-
creasing the concentration of PLGA, but the burst release The optimized AGM microspheres (Formulation E) were
of AGM-loaded PLGA microspheres was mainly related characterized in terms of particle size, size distribution, mor-
to the particle size. In order to obtain microspheres with phology, drug loading and encapsulation efficiency.
low burst release, the formulation was optimized by pre- As shown in Fig. 2, AGM microspheres have a mean
paring microspheres of different particle sizes through diameter of 60.2 μm and a narrow particle size distribution
varying the stirring speed. As shown in Table I, the par- (Span = 0.2). It has been reported that larger particle size of
ticle size of the microspheres could be adjusted by chang- microspheres could lead to a higher the encapsulation effi-
ing the stirring speed of emulsification, with a higher stir- ciency. (28). As well, the particle size and its distribution can
ring speed resulting in smaller microspheres. It was also significantly affect the in vitro release in the microspheres
found that as the particle size increased, burst release system. Therefore, microspheres with a relatively big parti-
from the microspheres decreased significantly (27). cle size are beneficial for obtaining a high drug encapsula-
Finally, considering that a very large particle size could tion and slow drug release.
restrict the syringeability of the formulation, formulation The SEM images (Fig. 3a and b) revealed smooth surfaces
E was chosen as the optimal formulation. structure with no pores and drug crystals. They confirmed
A simple direct extract method was used to determine that the particle size of AGM microspheres was evenly distrib-
the loading content and encapsulation efficiency of AGM uted, which is consistent with that obtained from the laser
in the microspheres accurately. The determined drug diffraction particle size analyzer.
loading of the optimized formulation was 23.7% and the The physical state of AGM within the microspheres
average encapsulation efficiency was 78.5%. It is well was investigated using the PXRD technique. As the phys-
known that the solubility of the drug in water is one of ical properties of high molecular weight PLGA are not
the major factors determining the encapsulation efficiency suitable for PXRD testing, blank microspheres instead of
of drugs in an emulsion solvent evaporation process PLGA was used in this study. Figure 4 showed the PXRD
(23,27). Drugs with high water solubility always tend to patterns of AGM, blank microspheres, microspheres with
leak into the outer water phase, causing a low encapsula- 20% AGM loading, the physical mixture of AGM and
tion efficiency (27). However, AGM is an insoluble drug blank microspheres (AGM: blank microspheres = 20: 80).
with a slightly higher solubility, and the solubility of AGM Several distinct peaks in the PXRD of AGM were consis-
in water increases with temperature, which is not condu- tent with reports indicating that AGM is present in a
cive to encapsulation of the drug. In order to improve the crystalline form II (29). However, they showed that only
encapsulation efficiency, the organic phase and the water a faint crystal diffraction peak appeared in microspheres
phase was precooled to a low temperature, which could when the drug loading was 20%, and thus it can be con-
cluded that the drug in microspheres with 20% AGM
Table I Relationship of Particle Size and Burst Release of AGM-Loaded loading mainly existed in the amorphous state, and a
PLGA Microspheres small part of AGM existed in a crystalline state.
Further investigation of the physical state of AGM within
Formulation A B C D E F
the microspheres was performed using DSC. Figure 5 showed
Stirring speed (rpm) 12,000 10,000 8000 6000 4000 2000 the DSC pattern of AGM, blank microspheres, physical mix-
Mean particle size (μm) 23.0 31.0 36.5 50.4 60.2 100.9 ture of AGM and blank microspheres and AGM-loaded mi-
Drug loading (%) 21.8 22.9 22.5 23.4 23.7 22.5 crospheres (20% drug loading). The AGM powder and phys-
Encapsulation efficiency (%) 72.5 75.9 74.5 77.8 78.5 77.4 ical mixtures showed obvious endothermic peaks at approxi-
Burst release (%) 25.6 22.6 16.4 11.4 7.3 7.1 mately 109°C. By contrast, there were two small endothermic
peaks at near 78°C and 97°Cin the microspheres with drug
9 Page 6 of 11 Pharm Res (2019) 36:9

Fig. 2 Particle size distribution of


AGM microspheres (75/25 PLGA
70 kDa; stirring speed: 4000 rpm/
min; drug loading = 20.31%;
encapsulation efficiency =
70.23%).

loading of 20%. This confirms that a small portion of AGM initially, and degradation/erosion controlled during the
exists in a crystalline state, but the majority of drug was dis- final stages of the release period (32) .
persed in the polymer matrix in an amorphous state, which is Figure 6 showed the in vitro release profile of AGM-loaded
consistent with the PXRD data. The endothermic peaks near microspheres. Only about 7.36% AGM was released from
78°C and 97°C were below the melting point of the drug, PLGA microspheres in the first day in vitro, indicating a small
which inferred that a small part of AGM and PLGA form burst release of AGM-loaded PLGA microspheres. From day
cocrystals, as AGM contains a secondary amide group which 2 to day 14, drug was released quickly from the microspheres
can be involved in supramolecular heterosynthons with the at a nearly constant rate of about 1.45% per day. However,
carboxyl end of PLGA (30). from day 14 to day 70, a lag time occurred and the drug was
It is well known that hydrophobic drugs tend to crystallize released at a rate of approximately 0.45% per day. In the next
inside the microspheres, and phase separation occurs within 35 days, the second accelerated release occurred and drug was
microspheres at high drug loadings, whereas the drug was released quickly at a nearly constant rate of 1.06% per day in
dispersed in the polymer in an amorphous state at lower drug this stage. The above results indicated that the pattern of drug
loading (17,31). In our study, both PXRD and DSC results release in vitro was different from the typical triphasic release
confirmed that there was a very small amount of AGM dis- profile, which can be divided into three stages: an initial burst
persed in a crystal state in the microspheres, and thus it could release, lag time depending on the Mw and end-capping of
be concluded that the maximal drug embedding capacity in the polymer with a slow or absent diffusion-controlled release,
the PLGA was less than 20%. and finally a second erosion-accelerated release (17).
However, in this study it showed that a rapid burst release
In Vitro Release Behavior of AGM Microspheres phase was followed by a period of near zero-order quick re-
lease, a lag phase and a final second accelerated release.
Drug release from PLGA-based drug delivery systems can Except for the first day’s burst release, the drug release
follow a combination of several mechanisms. In general, profiles of different release durations of AGM-loaded micro-
diffusion and degradation/erosion are two main release spheres could all be fitted to a zero order model, and accept-
mechanisms of microspheres, It is commonly believed that able regression coefficients were achieved for all release phases
the release rate of microspheres is diffusion-controlled (Table II). These results indicated that the drug release from

Fig. 3 SEM images of AGM


microspheres (75/25 PLGA
70 kDa; stirring speed: 4000 rpm/
min; D50 = 61.33; drug loading =
20.07%; encapsulation
efficiency = 71.20%).
Pharm Res (2019) 36:9 Page 7 of 11 9

Fig. 4 The PXRD curve of AGM


(a), blank microspheres (b), AGM-
loaded microspheres (c), physical
mixture of AGM and blank
microspheres (d).

PLGA microspheres was predominately governed by diffusion the diffusion of the drug. In other words, the initial burst
of drug through connected channels in the microspheres, release was attributed to the diffusion of non-encapsulated
which are formed by the presence of drug. drug particles on the surface or drug molecules close to the
To further investigate the release mechanism of AGM- surface. As the release continued, the surface of the micro-
loaded microspheres in vitro, SEM images of AGM-loaded spheres became uneven and the hydrophilic pores became
microspheres in different release stages were obtained larger and more numerous. In this phase, a fast release oc-
(Fig. 7). It can be observed that the microspheres kept their curred at a nearly constant rate of 1.45% per day and this is
shape and integrity over 10 weeks, until they disintegrated consistent with SEM results. However, after 2 weeks, the SEM
completely, suggesting that the slow-degradation region was images clearly showed the closure of the big pores at the sur-
at the surface, that is, that the internal degradation of the face, and the pores seemed to be still closed after 10 weeks,
microspheres was faster than at the surface. At the same time, which caused the lag time of drug from day 14 to day 70.
the surface of the microspheres began to show a small quantity Next, many detectable small pores began to form after
of big hydrophilic pores after 1 day, which was formed due to 10 weeks which can be seen in Fig. 7. This suggested that

Fig. 5 The DSC curves of AGM


(a), blank microspheres (b), AGM-
loaded microspheres (c), physical
mixture of AGM and blank
microspheres (d).
9 Page 8 of 11 Pharm Res (2019) 36:9

Fig. 6 In vitro release profiles of AGM-loaded microspheres (the optimal


formulation: drug loading = 20%, D50 = 60 μm, containing 2.0 mg AGM)
in release medium (0.5% SDS solution, containing 0.02% NaN3). Each point
represents the mean ± SD; n = 3.

the second accelerated release was attributed to a large num-


ber of newly formed small pores.
The above in vitro results indicated that the release of
drug from the microspheres was mainly through water-
filled pores. The initial burst release was attributed to
the diffusion of the drug in the surface of the micro-
spheres. However, the microsphere release in the later
phase was mostly the result of diffusion through water-
filled pores. Figure 8 showed the in vitro release process
of AGM-loaded microspheres. In general, the overall re-
lease mechanism of microspheres can be explained as fol-
lows. First, when the microspheres are immersed in an
aqueous release medium, the drug from the surface
spreads to the medium and hydration occurs, which is
faster than erosion (33,34). PLGA absorbs a large amount
of water and temporarily increases the pressure inside the
microspheres, which leads to the formation of big pores in
the low density area on the surface of the microspheres.
As the amount of the water increases inside the micro- Fig. 7 SEM images of AGM microspheres in the different release stages.
spheres, and the auto-catalytic phenomenon within the Each arrowhead represents a pore.
microspheres means the internal degradation of the mi-
crospheres is faster than at the surface, the pores become rearrangement of the PLGA chains. This means that
bigger and more numerous (32,34). Drug is mainly re- any significant increase in pressure can be compensated
leased from the big pores in this phase. Then, as the by swelling and arrangement of the polymer chains. The
release progresses, the volume of the water inside in- big pores in the surface of the microspheres later started
creases, as well as the mobility and the ability for to close in the release medium (35–37), but with further
hydrolysis of the PLGA and internal self-catalysis, a lot of
Table II Evaluation of Drug Release Kinetics of AGM-Loaded new pores formed and the drug tended to release from the
Microspheres newly formed pores. As well, a small amount of crystal-
lized or aggregated hydrophobic drug may reduce the
Phase Burst release(%) 2–14 day 14–70 day 70–105 day
release rate from another aspect. In general, the release
Slope 7.36 1.45 0.448 1.056 rate of microspheres is governed by the pore formation
R2 0.9806 0.9878 0.9989 and pore closure which occur simultaneously. The release
rate of microspheres in different release phases are kept
Pharm Res (2019) 36:9 Page 9 of 11 9

Fig. 8 The in vitro drug release


process of AGM-loaded
microspheres.

constant, which is common for drugs encapsulated in 30 days with almost no burst release in vivo, and the bioavail-
PLGA drug delivery systems. Studies showed that small ability of AGM was dramatically increased when it was pre-
porous microspheres prepared by high-Mw, hydrophobic pared into sustained release microspheres. Three different
PLGA with low polymer chain mobility is more likely to AGM microspheres were released in vitro for 3 months, with
release drugs in a constant rate (34). a burst release of 25%, 7.0% and 7.0% respectively, but when
released in vivo for 30 days demonstrated a burst release of
In Vivo Pharmacokinetics of AGM Microspheres 10.0%, 9.0% and 6.0% (calculated by the AUC (0-1 day)
versus AUC (0-∞)), respectively. This indicated that in vitro re-
The in vitro release studies showed that the AGM-loaded mi- lease test could predict the burst release of AGM microspheres
crospheres exhibited a promising drug release behavior, as
reflected by a small burst release followed by a slow release.
To further evaluate the in vivo release behavior of different
AGM microspheres, the pharmacokinetics of oral AGM solu-
tion, which could evaluate the oral bioavailability and the
minimum effective blood concentration of AGM in rats, and
three different formulations (MS1, MS2 and MS3) of AGM
microspheres were investigated.
The mean plasma concentration-time profiles of AGM in
rats after oral administration of AGM solution and intramus-
cular administration of AGM microspheres are shown in
Fig. 9. The main calculated pharmacokinetic parameters were
listed in Table III.
As can be seen in Fig. 9a and Table III, the AGM solution
was absorbed by the mouth quickly and the plasma concen-
tration reached the peak concentration (Cmax) 32.6 ng/mL in
approximately 1 h. The plasma concentration decreased be-
low 5 ng/mL after a single oral dose of 2.6 mg/mL AGM
solution to the rats after 24 h, suggesting that the minimun
effective plasma concentration was about 5 ng/mL.
As seen in Fig. 9b, after intramuscular injection of three
different AGM microspheres, the plasma concentration of
AGM reached a peak concentration (109.5, 37.4 and
20.3 ng/mL for MS1, MS2 and MS3 respectively), and then
the concentration declined gradually. The blood concentra-
tion dropped below 5 ng/mL in 30 days. Compared to AGM
solution administrated intragastrically, the T1/2 value of
AGM microspheres was prolonged to 96.5 h, 99.6 h and
178.4 h, respectively, which was approximately 6.3, 6.6 and
11.8-fold longer than that of oral AGM solution.
Furthermore, the AUC of three different AGM microspheres Fig. 9 Pharmacokinetic results of AGM. (a) mean concentration-time profile
was approximately 4.8, 3.3 and 2.5-fold more than the calcu- of AGM after a single oral dose of 2.6 mg/mL AGM solution to the rats, (b)
plasma concentration- time profile of AGM after intramuscular injection of
lated AUC of AGM oral solution of the equivalent dose, re- AGM-loaded microspheres (10 mg/kg). MS1: drug loading = 20%, D50 =
spectively. These results further confirmed that AGM micro- 30 μm, MS2: drug loading = 7%, D50 = 30 μm, MS3: drug loading = 20%,
spheres could provide a sustained release of approximately D50 = 60 μm.
9 Page 10 of 11 Pharm Res (2019) 36:9

Table III The Non-


Compartmental Model Parameters Formulations AGM solution MS1 MS2 MS3
of AGM after i.g. Administration of
2.6 mg/mL AGM Solution and AUC(0-t) (mg/L·h) 330.9 ± 52.5 6779.0 ± 1106.3 5025.2 ± 841.6 3709.2 ± 778.7
Intramuscular Administration of AUC(0-∞) (mg/L·h) 461.3 ± 106.5 7285.6 ± 1149.6 7564.1 ± 3940.2 5560.0 ± 996.3
AGM Microspheres (10 mg/mL) AUC(0-1 day) (mg/L·h) – 680.6 ± 111.1 451.6 ± 75.6 224.4 ± 47.1
(Mean ± SD; n = 6)
T1/2 (h) 15.2 ± 7.3 96.5 ± 16.6 99.6 ± 33.3 178.4 ± 27.0
Cmax(mg/L) 32.6 ± 12.5 109.5 ± 17.6 37.4 ± 1.4 20.3 ± 4.7
Tmax (h) 1.0 ± 0.0 1.0 ± 0.9 3.3 ± 4.0 –
Burst release (%) – 10.0 9.0 6.0

in vivo. It is significant to develop an in vitro release assay with to agglomerate and its burst release became slowly. However,
good in vitro and in vivo correlation (IVIVC), which can be used there was no significant change in fluidity and the burst release
not only to predict the in vivo behavior of the formulation and when the microspheres were stored at 4°C.
optimize the formation during formulation development, but
also be used to control the quality of the product in the prep-
aration of the product process, and so further work will focus
on establishing a new in vitro release assay with a good in vitro CONCLUSION
and in vivo relationship.
Comparing MS1 with MS2, it can be inferred that drug AGM-loaded PLGA microspheres with smooth surface
loading could significantly influence the burst release of the and narrow particle size distribution were prepared by
microsphere in vivo. The lower the drug loading could result in the emulsion solvent evaporation method. The drug load-
the smaller the burst release in vivo. However, the AUC of ing content and encapsulation efficiency of AGM micro-
MS1 was bigger than that of MS2, indicating that AGM spheres were 23.7% and 78.5% respectively. DSC and
was eliminated quickly in the blood and the rate of elimination PXRD analysis demonstrated that the majority of drug
of AGM is faster than absorption. Comparing MS1 with was dispersed in the polymer matrix in an amorphous
MS3, it was apparent that particle size had a significant effect state, and only a small part of AGM existed in a crystal-
on the release of microspheres in vivo. It can be seen that the line state.
bigger the particle size, the smaller the burst release in vivo, In vitro release studies showed that AGM microspheres
suggesting that a relatively big particle is beneficial for achiev- released slowly with a small burst release of 7.0% followed
ing a sustained release in vivo with a small burst release. As is by a fast release, a lag time and a second accelerated
shown in Table III, T1/2: MS1 < MS2 < MS3, Cmax: MS1 > release in vitro. SEM images showed pore formation and
MS2 > MS3, burst release in vivo: MS1 > MS2 > MS3. The pore closure in different release stages, indicating that the
results concluded that MS3 showed the best sustained release main release mechanism of the microspheres was diffusion
characteristics and minimal burst release, which was very im- through water filled pores in vitro.
portant for reducing the side effects caused by a too high Over 30 days, a sustained and steady plasma concen-
concentration in the blood in vivo. tration of AGM could be achieved after a single intramus-
cular injection of AGM microspheres. In vivo release stud-
Storage Stability ies also showed that particle size and drug loading had
significant effects on the release of AGM-loaded micro-
The stability of the optimized formulation was measured at spheres. These results suggested the potential use of
4°C and room temperature for 30 days, respectively. As AGM-loaded PLGA microspheres for treatment of de-
shown in Table IV, microspheres at room temperature tended pression over a long time period.

Table IV The Stability Results of


the Optimized Formulation Condition 0 day 7 days 28 days

4 °C 25 °C 4 °C 25 °C 4 °C 25 °C

Fluidity good good good Slightly agglomeration good agglomeration


Burst release in vitro 8.96 8.49 8.18 6.99 8.88 6.94
Pharm Res (2019) 36:9 Page 11 of 11 9

REFERENCES 21. Rodríguez VJ, Bravo-Osuna I, Herrero-Vanrell R, Molina


Martínez IT, Guzmán NM. Optimising the controlled release of
dexamethasone from a new generation of PLGA-based micro-
1. Kessler RC, Chiu WT, Demler O, Walters EE. Prevalence, spheres intended for intravitreal administration. Eur J Pharm Sci.
Severity, and Comorbidity of twelve-month DSM-IV disorders in 2016;92:287–97.
the National Comorbidity Survey Replication (NCS-R). Arch Gen 22. Guo Y, Yang Y, He L, Rong S, Pu C, Xie B, et al. Injectable
Psychiatry. 2005;62:617–27. sustained-release depots of PLGA microspheres for insoluble drugs
2. Dolder CR, Nelson M, Snider M. Agomelatine treatment of major prepared by hot-melt extrusion. Pharm Res. 2017;34:2211–22.
depressive disorder. Ann Pharmacother. 2008;42:1822–31.
23. Ramazani F, Chen W, van Nostrum CF, Storm G, Kiessling F,
3. Sanand L, Arranzb B. Agomelatine: a novel mechanism of antide-
Lammers T, et al. Strategies for encapsulation of small hydrophilic
pressant action involving the melatonergic and the serotonergic
and amphiphilic drugs in PLGA microspheres: state-of-the-art and
system. Eur Psychiatry. 2008;23(6):396–402.
challenges. Int J Pharm. 2016;499:358–67.
4. Garcia-Cebrianabcde A, Grassifgahai L, et al. Factors influencing
24. Alaina FJ, Galperin A, Ratner BD. Drug encapsulated polymeric
depression endpoints research (FINDER): study design and popu-
microspheres for intracranial tumor therapy: a review of the litera-
lation characteristics. Eur Psychiatry. 2008;23(1):57–65.
ture ☆ ☆☆. Adv Drug Deliv Rev. 2015;91:23–37.
5. Ferrari AJ, Charlson FJ, Norman RE, Patten SB, Freedman G,
Murray CJ, et al. Burden of depressive disorders by country, sex, 25. Hirota K, Doty AC, Ackermann R, et al. Characterizing release
age, and year: findings from the global burden of disease study mechanisms of leuprolide acetateloaded PLGA microspheres for
2010. PLoS Med. 2013;10:e1001547. IVIVC development I: In vitro evaluation. J Control Release.
6. Dubovsky SL, Warren C. Agomelatine, a melatonin agonist with 2016;244:302–313.
antidepressant properties. Expert Opin Investig Drugs. 2009;18: 26. Pu C, Wang Q, Zhang H, et al. In Vitro-In Vivo Relationship of
1533–40. Amorphous Insoluble API (Progesterone) in PLGA Microspheres.
7. De BC, Guardiolalemaitre B, Mocaër E, Renard P, Muñoz C, Pharm Res. 2017;34(12):2787–2797.
Millan MJ. Agomelatine, the first melatonergic antidepressant: dis- 27. Guo W, Quan P, Fang L, et al. Sustained release donepezil loaded
covery, characterization and development. Nat Rev Drug Discov. PLGA microspheres for injection: Preparation, in vitro, and in vivo,
2010;9:628–42. study. Asian J Pharm Sci. 2015;10(5):405–414.
8. Kennedy SH. Agomelatine: efficacy at each phase of antidepressant 28. Ogawa Y, Yamamoto M, Okada H, Yashiki T, Shimamoto T. A
treatment. Cns Drugs. 2009;23(Suppl 2):41–7. new technique to efficiently entrap leuprolide acetate into micro-
9. Demyttenaere K. Agomelatine: a narrative review. Eur capsules of polylactic acid or copoly(lactic/glycolic) acid. Chem
Neuropsychopharmacol. 2011;21(Suppl 4):S703–S709. Pharm Bull. 1988;36:1095–103.
10. Serna MC, Cruz I, Real J, et al. Duration and adherence of anti- 29. Souvie J C, Gonzalez-Blanco I, Thominot G, et al. Process for the
depressant treatment (2003 to 2007) based on prescription data- synthesis and crystalline form agomelatine: U.S. Patent 7,544,
base. Eur Psychiatry. 2010;25(4):206–213. 839[P]. 2009-6-9.
11. Kessler RC, Coccaro EF, Fava M, Jaeger S, Jin R, Walters E. The 30. Prohens R, Barbas R, Portell A, Fontbardia M, Alcobé X,
prevalence and correlates of DSM-IV intermittent explosive disor- Puigjaner C. Polymorphism of Cocrystals: the promiscuous behav-
der in the National Comorbidity Survey Replication. Arch Gen ior of Agomelatine. Cryst Growth Des. 2016;16:1063–70.
Psychiatry. 2006;63:669–78. 31. Mao S, Shi Y, Li L, Xu J, Schaper A, Kissel T. Effects of process
12. Du W, Zhou Y, Gong Y, Zhao C, et al. Investigation of physico- and formulation parameters on characteristics and internal mor-
chemical properties and in-vitro in-vivo evaluation of agomelatine phology of poly(d,l-lactide-co-glycolide) microspheres formed by
polymorphs. Asian J Pharm Sci. 2013;8(3):181–190. the solvent evaporation method. Eur J Pharm Biopharm.
13. Yan L, Gao H, Ren F, Ren G. Solubility of Agomelatine crystal 2008;68:214–23.
form I and form II in pure solvents and (isopropanol + water) 32. Pu C, Wang Q, Zhang H, Gou J, Guo Y, Tan X, et al. In vitro-in
mixtures. J Chem Eng Data. 2015;60:151019103620003. vivo relationship of amorphous insoluble API (Progesterone) in
14. Rhee YS, Park CM, DeLuca PP, Mansour HM. Sustained-release PLGA microspheres. Pharm Res. 2017:1–11.
injectable drug delivery. Pharm Tech. 2010:34;s6–s13
33. Gu B, Sun X, Papadimitrakopoulos F, et al. Seeing is believing,
15. Freiberg S, Zhu XX. Polymer microspheres for controlled drug
PLGA microsphere degradation revealed in PLGA microsphere/
release. Int J Pharm. 2004;282:1–18.
PVA hydrogel composites. J Control Release. 2016;228:170–178.
16. Varde NK, Pack DW. Microspheres for controlled release drug
34. Fredenberg S, Wahlgren M, Reslow M, Axelsson A. The mecha-
delivery. Expert Opin Biol Ther. 2004;4:35.
nisms of drug release in poly(lactic-co-glycolic acid)-based drug de-
17. Wischke C, Schwendeman SP. Principles of encapsulating hydro-
livery systems—a review. Int J Pharm. 2011;415:34–52.
phobic drugs in PLA/PLGA microparticles. Int J Pharm. 2008;364:
298–327. 35. Fredenberg S, Jönsson M, Laakso T, Wahlgren M, Reslow M,
18. Zhang Z, Bi X, Li H, et al. Enhanced targeting efficiency of PLGA Axelsson A. Development of mass transport resistance in
microspheres loaded with Lornoxicam for intra-articular adminis- poly(lactide-co-glycolide) films and particles – a mechanistic study.
tration. Drug deliv. 2011;18(7):536–544. Int J Pharm. 2011;409:194–202.
19. Wang W, Cai Y, Zhang G, Liu Y, Sui H, Park K, et al. 36. Kang J, Schwendeman SP. Pore closing and opening in biodegrad-
Sophoridine-loaded PLGA microspheres for lung targeting: prep- able polymers and their effect on the controlled release of proteins.
aration, in vitro, and in vivo evaluation. Drug Delivery. 2016;23: Mol Pharm. 2007;4:104.
3674–80. 37. Fredenberg S, Wahlgren M, Reslow M, Axelsson A. Pore forma-
20. Zhang Y, Wischke C, Mittal S, Mitra A, Schwendeman SP. Design tion and pore closure in poly(D,L-lactide-co-glycolide) films. J
of controlled release PLGA microspheres for hydrophobic Control Release. 2011;150:142–9.
fenretinide. Mol Pharm. 2016;13:2622–30.

You might also like