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

Andrographolide solid dispersions formulated by Soluplus to enhance

interface wetting, dissolution, and absorption


Bing Song,1 Jian Wang,2 Si-Jing Lu,3 Li-Na Shan 3
1
Department of Endocrinology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
2
Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Shenyang, 110016, China
3
Department of Respiratory, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
Correspondence to: L. N. Shan (E-mail: shanlina123@163.com) and S. J. Lu (E-mail: sxbsln@163.com)

ABSTRACT: Andrographolide (ADG) had profound functions as drug health-care product. To improve ADG application in medical care, solid
dispersion technique was introduced. Initially, several polymers were chosen to prepare ADG solid dispersion using hot-melt extrusion. Work-
ing mechanisms of ADG solid dispersion, which include molecular docking and wetting property, were studied. The strongest molecular bind-
ing energy of ADG with Soluplus contributed to carrier-controlled mechanism, leading to its highest drug dissolution. Soluplus as solid
dispersion excipient could significantly improve drug absorption. In addition, ADG solid dispersions formulated by Soluplus were prepared
and studied, and the ratios of drug to polymer were 1:5, 1:7, 1:9, and 1:12, respectively. It demonstrated that hydrogen bond interactions were
formed between ADG and Soluplus based on infrared (IR) spectroscopy analysis. The characteristic peaks of crystal state of ADG at 11.97 ,
14.95 , 15.86 , and 9.78 disappeared in ADG solid dispersion, demonstrating that excipient was efficient in transforming drug crystal state to
amorphous phase by interacting with ADG. ADG-Soluplus solid dispersion of 1:7 turned out to be the best with maximum cumulative release
amount of more than 80%, whereas other ADG solid dispersions were in the range of 60–70%. The cumulative release amount of pure
ADG was below 20%. Comparing with the oral administration of pure ADG, we found that the mean Cmax and AUC values of ADG for ADG-
Soluplus solid dispersion were approximately increased 1.96- to 2.53-fold. © 2019 Wiley Periodicals, Inc. J. Appl. Polym. Sci. 2019, 136, 48354.

KEYWORDS: andrographolide; hot-melt extrusion; solid dispersion; Soluplus

Received 23 March 2019; accepted 13 July 2019


DOI: 10.1002/app.48354

INTRODUCTION surface and interacting with drug molecules.23 Many polymers, such
Andrographolide (ADG) is one of the main active components as polyoxyethylene pyrrolidone K30,24 polyethylene glycol,25,26
isolated from the Chinese herb Andrographis paniculata. ADG Eudragit series,27,28 hydroxy propyl cellulose,29 poloxamer,30 and
has many pharmacological actions, including anti-inflammatory,1,2 Soluplus,31,32 have been considered as excipients for forming solid
anti-infection,3–5 anticancer,6–8 anti-hyperglycemia,9 anti-angio- dispersion.
genesis,10,11 hepato-protection,12,13 antifertility,14 and anti-HIV15,16 In this article, ADG solid dispersion was prepared by hot-melt
effects. However, its high lipophilicity (log P = 2.63), low aqueous sol- extrusion. Hot-melt extrusion method was first used in the field
ubility (74 μg mL−1), and poor stability seriously limit its applica- of pharmaceutical preparations in 1970s.33 It requires no solvent
tion.17 ADG can be normally made into tablet, capsule, and dropping participation, has few operation steps, and is suitable for large-
pill, whereas its oral bioavailability is low due to its low solubility scale industrial production.34,35 Since the application of various
(74 μg mL−1).18 In order to tackle this problem, soluble salt and new auxiliary materials and pharmaceutical equipment, the appli-
superfine grinding have been used. Various formulation techniques, cation frequency of this method has been increasing in the past
including nanoemulsion,19 cyclodextrin inclusion complexes,20 and 40 years. However, it is difficult to apply this method for ther-
liposomes,21,22 have been also applied. Among them, solid dispersion mally unstable excipients and drugs, typically as albendazole.36,37
turns out to be an outstanding strategy to improve dissolution and In the process of research work, we screened out the suitable
bioavailability of ADG. Solid dispersion is designed as drug dispersion polymer and drug/polymer ratio for preparing ADG solid disper-
system that drug is highly dispersed in a suitable solid carrier, which sion, analyzed their molecular interaction through IR spectros-
can effectively change drug physical state from crystalline to amor- copy, and studied crystal form using X-ray diffraction (XRD).
phous phase owing to the advantages of carriers in enlarging area Further, the current study focused on interface wetting property

Additional Supporting Information may be found in the online version of this article.
© 2019 Wiley Periodicals, Inc.

48354 (1 of 7) J. APPL. POLYM. SCI. 2019, DOI: 10.1002/APP.48354


ARTICLE WILEYONLINELIBRARY.COM/APP

of as-synthesized ADG solid dispersion through measuring Shimadzu Inertsil ODS-3 column (4.6 mm × 150 mm, 5 μm) at a
dynamic contact angle. wavelength of 220 nm. The HPLC working conditions for ADG
analysis were (1) methanol:water (0.1% formic acid) = 51:49, v/v;
EXPERIMENTAL (2) flow rate of 1.0 mL min−1; and (3) column temperature of
Materials 30  C  1  C.
ADG with purity above 99% was purchased from Wuhan Dong-
Infrared Spectroscopy
kangyuan Technology Co., Ltd. (Wuhan, China). Polyethylene glycol
The IR spectroscopy (Spectrum 1000; Perkin Elmer) spectra of
6000 (PEG 6000), polyvinylpyrrolidone VA64 (PVP VA64), and pol-
10 scans, including ADG, Soluplus, physical mixtures (ADG and
oxamer 188 (F68) were purchased from Sigma-Aldrich Chemical
Soluplus with weight ratio of 1:5, 1:7, 1:9, and 1:12) and solid dis-
Co. (St. Louis, MO). Soluplus was provided by BASF Company
persions (ADG:Soluplus = 1:5, 1:7, 1:9, and 1:12, respectively)
(Ludwigshafen, Germany). Other chemical agents were obtained
were obtained over the spectral region of 400–4000 cm−1. All the
from Tianjin Bodi Chemical Holding Co., Ltd. (Tianjin, China).
samples were made with KBr.
Preparation Process and Working Mechanism
X-ray Diffraction
ADG was physically mixed with different polymers. All chemical
XRD of ADG, Soluplus, and ADG solid dispersion formed by
structures were shown in the Supporting Information Figure S1.
Soluplus with different drug/polymer ratios was analyzed using a
The ratios of ADG/polymer (w/w) and temperature settings were
Shimadzu XRD-6000 diffractometer (Shimadzu Corporation)
listed in Table I. The weight ratios of ADG and carrier were set
equipped with a Cu-Ka source and set in Bragg–Brentano geome-
at 1:5, 1:7, 1:9, and 1:12, respectively. Extrusion process of all
try, scanning between 5  C and 50  C 2θ at 2  C min−1 with a
ADG and polymer physical mixtures was conducted using a
0.04 step size.
Thermo Scientific Haake MiniLab II conical, co-rotating, twin-
screw microcompounder (Thermo Electron, Karlsruhe, Ger- Contact Angle Measurement
many). The screw speed was set at 50 rpm. The interface wetting property of ADG solid dispersion formu-
The 3D structures of macromolecules were optimized using the lated by Soluplus was studied by dynamic contact angle measured
Sybyl 6.9.1 software package. The parameters that included energy using an automatic contact angle meter model JCY series
change (0.005 kcal mol−1) and max iterations (10 000) were also (Shanghai, China) with operation manual.39 Briefly, 200 mg sam-
optimized. All other parameters were kept at the default values. ple was weighed and compressed using a circular stainless steel
Molecular docking was studied using the AutoDock 4.0 software. punch and die to get tablet with flat surface. A drop of dissolu-
Energy evaluations were set at 25 000 000 per run. Discovery Studio tion medium (2 μL) was placed on tablet as the start of measure-
4.0 was used to understand the binding interactions between the ment. Tangent method was explored to calculate the contact
drug and polymers as well as molecular binding energy. angle of tested sample.40

In vitro Dissolution Pharmacokinetics Study


In vitro dissolution tests for screening suitable polymer and optimal Thirty male Wister rats (200–250 g) were obtained from the Labora-
ADG/polymer ratio for ADG solid dispersion were performed using tory Animal Center of Liaoning Medical University. The study pro-
stirring paddle method (50 rpm, 37  C) with a RCZ-6C3 tester tocols were approved by the Institutional Animal Ethics Committee.
(Huanghai, China) according to 2015 Chinese pharmacopeia.38 The rats were divided randomly into five groups (n = 6 per group)
Since small intestine was the main absorption site for ADG, samples for the administrations of a single dose of ADG, ADG-F68 1/7 solid
were exposed to pH 6.8 phosphate buffer solutions (250 mL). At pre- dispersion, ADG-Soluplus 1/7 solid dispersion, ADG-PEG 6000 1/7
determined time intervals, 5 mL of sample medium was withdrawn solid dispersion, and ADG-PVP AV64 solid dispersion. The ADG or
and an equivalent amount of fresh medium was added into the disso- ADG solid dispersions were dissolved in physiological saline solu-
lution cup. Sample medium administered through a 0.45 μm micro- tions to form suspensions. Each group of rats was orally dosed at
porous membrane was determined by a Shimadzu LC-20AT HPLC 50 mg kg−1 of ADG. The blood samples (0.3 mL) were collected
system (Shimadzu Corporation, Kyoto, Japan) equipped with a from orbital venous plexus into dried heparinized tubes at 5, 15,
30, 45, 60, 90, 120, 180, 240, 360, 480, and 720 min and were cen-
trifuged at 4000 rpm for 10 min at 4  C to obtain the plasma samples.
Table I. The Ratios of ADG/Polymer (w/w) and Temperature Settings The plasma (0.1 mL) was mixed with 200 μL of acetonitrile and cen-
trifuged at 12 000 rpm for 20 min at 4  C. The supernatant was col-
Drug–polymer Process lected and an aliquot of 20 μL of the solution was analyzed by
Sample ID system w:w temperature ( C) LC/MS/MS system.
1 ADG:F68 1:7 150 The LC–MS/MS system (Waters Corporation, Milford, MA) was
2 ADG:PEG 6000 1:7 130 fitted with an Acquity UPLC BEH C18 column (2.1 mm × 100 mm,
3 ADG:PVP VA64 1:7 170 1.7 μm; Waters Co. Ltd., Milford, MA). The mobile phases consisted
4 ADG:Soluplus 1:7 150 of LC grade water with 0.1% formic acid (A) and LC grade methanol
5 ADG: Soluplus 1:5 150 (B) with the following gradient profile: 0.0–1.5 min from 60 to
6 ADG: Soluplus 1:9 150
90% B, 1.5–2.5 min from 90 to 90% B, 2.5–3.0 min 90 to 60% B, and
3.0–5.0 min at 60% B. Mass detection of ADG was performed in neg-
7 ADG: Soluplus 1:12 150
ative, and quantification was performed using the multiple reaction

48354 (2 of 7) J. APPL. POLYM. SCI. 2019, DOI: 10.1002/APP.48354


ARTICLE WILEYONLINELIBRARY.COM/APP

monitoring (MRM) method with the transitions of m/z 349.1 ! m/ on Pluronic F68. Figure 3(d) showed the molecular docking results
z 287.1 for ADG. According to preliminary experiments (data not of ADG with PVP-VA64, and the corresponding binding affinity
shown), the performance parameters that include specificity, linear- was calculated as −4.37 kcal mol−1. As for their binding mode, a
ity, accuracy, precision, decision limit, quantification limit, and sta- hydrogen bond formed by a hydroxyl group of ADG with PVP
bility proved that the validated method was qualified for the VA64 was observed, and the hydroxyl group also formed an intra-
determination of ADG. molecular hydrogen bond with another hydroxyl group at its ortho-
position. In a summary, it was found that ADG can interact with the
RESULTS AND DISCUSSION four types of material mainly through hydrogen bond formed by the
Superior Polymer for ADG Solid Dispersion hydroxyls of the small molecule. Besides, several hydrogen bonds
The photograph of the prepared ADG-Soluplus solid dispersion was were also formed with Soluplus, the binding affinity of which was
shown in Figure 1. ADG-Soluplus solid dispersion was uniformly proved to be the highest, followed by PVP VA64, F68, and PEG 6000
powder particles, which was favorable for obtaining solid dispersion according to their binding affinities. Therefore, the stronger binding
with high drug dispersity. According to drug dissolution result affinity between ADG and PVP VA64 contributed to its obvious
[Figure 2(a)], it was obvious that Soluplus was the best choice among higher drug dissolution.
these excipients with highest cumulative drug release amount of The wetting property of ADG and ADG solid dispersions prepared
more than 80% while others were lower than 70% (the dissolution of by these excipients was studied to elucidate their working mecha-
ADG solid dispersion made by PVP VA64, F68 and PEG 6000 were nism. It is well known that drug release mechanisms of solid disper-
almost 60, 20, and 20%, respectively). The cumulative release amount sion divide into drug-controlled and carrier-controlled mechanism.
of pure ADG was below 20%. In order to illustrate this result, molec- Carrier becomes the controller when drug molecularly disperses
ular docking strategy was applied to calculate the binding energy within polymer layer and releases along with carrier. On the con-
between drug and excipient. The binding modes of the four com- trary, when drug releases faster than polymer diffusion layer, drug
plexes that were proved to be the most stable one with the lowest manages dissolution process and is mainly influenced by its charac-
binding energy were analyzed further. Figure 3(a) described the teristics (size, physical form, etc.). Carrier-controlled mechanism is
molecular docking result of Soluplus with ADG, and the binding superior to drug-controlled mechanism. As shown in Figure 2(b),
affinity of the complex was calculated as −4.73 kcal mol−1, which the initial time period of contact angle of ADG solid dispersions for-
was the lowest among the four complexes. It observed that there were mulated by PEG 6000 and F68 was close to ADG (contact angle
two hydrogen bonds formed between the dihydroxyl group of ADG values of ADG, ADG-PEG solid dispersion, and ADG-F68 solid dis-
with the ether oxygen atoms on ester group of Soluplus, additionally, persion were in the range of 75 –88 ), while ADG solid dispersion
and hydrophobic contacts were found between the met- formulated by Soluplus and PVP VA64 became easier to be wetted
hylenecyclohexyl group of ADG and caprolactam group on Soluplus. (contact angle values of ADG-Soluplus solid dispersion and ADG-
Figure 3(b) showed the molecular interaction between ADG and PVP VA64 solid dispersion were 48.88 and 66.23 , respectively). It
PEG 6000, the binding affinity of which was calculated as demonstrated that ADG controlled its release in the former two solid
−4.18 kcal mol−1, and the hydrogen bonds formed by three hydroxyl dispersions and led to their low drug cumulative release. Polymer
group of ADG with the oxygen atoms on PEG 6000 were the major of Soluplus and PVP VA64 exerted functions in bringing out
contacts in this complex. Figure 3(c) represented the molecular inter- ADG along with its dissolution. The obtained wetting property
action of Pluronic F68 with ADG generated by molecular docking result confirmed in vitro dissolution result, which was in agreement
studies, the binding affinity of them was calculated as with the conclusions from related literature that carrier-controlled
−4.28 kcal mol−1, and two hydrogen bonds can be found in this mechanism is superior to drug-controlled mechanism.41 The disso-
complex were formed by the dihydroxyl group with oxygen atoms lution mechanism was related with molecular binding energy mea-
sured using molecular docking technique. The molecular binding
energies of Soluplus and PVP VA64 with ADG were −4.73 and
−4.37 kcal mol−1, respectively, and they were higher than PEG 6000
and F68 with ADG (−4.18 and −4.28 kcal mol−1). The contact angle
result was in agreement with in vitro drug release result, demonstrat-
ing that the stronger binding affinity between ADG and carrier con-
tributed to its better wetting ability and therefore higher dissolution.
The result further confirmed the reason why Soluplus can be chosen
as the final excipient for preparing ADG solid dispersion. The eluci-
dation of working mechanism of ADG solid dispersion provided
valuable instruction when screening proper excipient and clearly
presented the important information of drug and excipient in solid
dispersion.

Pharmacokinetic Study
The concentration–time curves of ADG and ADG solid dispersion
Figure 1. The photograph of the prepared ADG-Soluplus solid dispersion. were shown in Figure 4 and the pharmacokinetics parameters were
[Color figure can be viewed at wileyonlinelibrary.com] shown in Table II. The area under the plasma concentration–time

48354 (3 of 7) J. APPL. POLYM. SCI. 2019, DOI: 10.1002/APP.48354


ARTICLE WILEYONLINELIBRARY.COM/APP

Figure 2. The in vitro dissolution (a) and contact angle (b) of ADG and ADG solid dispersion. [Color figure can be viewed at wileyonlinelibrary.com]

Figure 3. Molecular docking of ADG with Soluplus (a), PEG 6000 (b), F68 (c), and PVP-AV64 (d). [Color figure can be viewed at wileyonlinelibrary.com]

curve (AUC) and the ADG peak plasma concentration (Cmax) characteristic bands that belonged to ─OH, ester, carbanyl group,
differed markedly between different samples (the mean AUC values CH2 at 3399.0, 2928.8, 1727.6, 1673.7, 1408.5, and 1033.0 cm−1,
of ADG, ADG-PEG 6000, ADG-F68, ADG-PVP AV64, ADG- respectively.42 It also presented C─O─C band at 1010.0 cm−1,
Soluplus were 2.72, 3.36, 3.41, 4.76, and 8.72 mg L−1*h, respectively, C─H stretching vibration band at 981.2 cm−1, and C C rocking
and their mean Cmax values were 0.86, 0.91, 0.96, 1.59, and vibration band at 702.8 cm−1.43 The characteristic bands OH group
2.91 mg L−1 respectively). It should be first noted that the oral bio- from soluplus showed the characteristic band at 3445.9 cm−1 and
availability of these four ADG solid dispersions were higher than ester group exhibited the characteristic band at 2930.4 cm−1.44
ADG evidenced by their higher AUC compared with ADG
according to Table II, demonstrating that solid dispersion was an
effective strategy to improve ADG bioavailability. Comparing with
the oral administration of pure ADG, we found that the mean Cmax
and AUC values of ADG for ADG-Soluplus solid dispersion were
approximately increased 1.96- to 2.53-fold, confirming the superior-
ity of Soluplus as ADG solid dispersion carrier owing to the strong
binding affinity between ADG and Soluplus. Compared to other
ADG solid dispersions, the ADG-Soluplus solid dispersion 1:7 had
significant high oral bioavailability.18,29 Other solid dispersion,
including ADG-PEG 6000, ADG-PVP VA64, and ADG-F68,
showed little effect on AGD absorption. Thus, Soluplus is a good
solid dispersion excipient to increase drug absorption. Good drug
absorption of ADG-Soluplus solid dispersion may be related to the
highest cumulative drug release in media.

Infrared Figure 4. Plasma concentration–time curves of ADG after oral administra-


The interaction between ADG and excipient was studied through IR tion of ADG and ADG solid dispersions. Data are expressed as mean  SD
analysis. The IR spectra are presented in Figure 5. ADG showed its (n = 6). [Color figure can be viewed at wileyonlinelibrary.com]

48354 (4 of 7) J. APPL. POLYM. SCI. 2019, DOI: 10.1002/APP.48354


ARTICLE WILEYONLINELIBRARY.COM/APP

Table II. The Main Pharmacokinetic Parameters of ADG Solid Dispersions in Rats

Solid dispersions

ADG ADG-PEG 6000 ADG-F68 ADG-PVP AV64 ADG-Soluplus


AUC(0-∞) (mg/L*h) 2.72  0.15 3.36  0.23 3.41  0.36 4.76  0.28 8.72  1.36
t1/2 (h) 2.33  0.59 2.38  0.36 2.43  0.21 2.52  0.32 2.23  0.21
Tmax (h) 0.75  0.24 1.03  0.25 1.23  0.24 1.13  0.68 1.02  0.11
Cmax (mg L−1) 0.86  0.12 0.91  0.13 0.96  0.26 1.59  0.32 2.91  0.12

Carbanyl group exhibited two characteristic bands at 1739.3 cm−1 X-ray Diffraction
and 1636.7 cm−1. In addition, its CH2 presented two bands at The XRD spectra of ADG, excipient, physical mixtures and ADG
1442.4 cm−1 and 1241.1 cm−1.45 Whereas Soluplus had aliphatic solid dispersions formulated by Soluplus were shown in Figure 6.
ether (─O─) stretching band at 973.4 cm−1. After mixing ADG with Crystal ADG displayed its characteristic peaks at 9.78 , 11.97 ,
Soluplus or preparing ADG solid dispersion, the stretching vibration 14.95 , and 15.86 2θ. After mixing with Soluplus, intensity of
of C─H from ADG cannot be observed while the band of aliphatic drug peaks was weakened because the excipient was able to cover
ether from Soluplus still presented in IR spectra, indicating that these peaks by interacting with ADG according to the wavelength
the C─H of ADG was covered by aliphatic ether band of Soluplus. movement of physical mixture of IR result. However, the pres-
According to molecular docking result of ADG with Soluplus, hydro- ence of many characteristic peaks illustrated that crystal ADG
gen bonding interaction was mainly formed. ADG showed its char- was remained in mixture samples. The situation was changed
acteristic bands at 3399.0, 1727.6—, and 1631.5 cm−1, which can be when ADG was loaded into Soluplus using solid dispersion strat-
assigned to ─OH and C O groups, respectively. The spectrum of egy. The characteristic peaks of crystal state of ADG at 11.97 ,
Soluplus displayed classical bands at 3445.9, 1739.3, and 14.95 , 15.86 , and 9.78 disappeared in ADG solid dispersion,
1636.7 cm−1, which belonged to ─OH and C O groups. After demonstrating that excipient was efficient in transforming drug
mixing ADG with Soluplus, C O groups of Soluplus can be crystal state to amorphous phase by interacting with ADG. The
observed while ─OH groups were assigned to ADG, indicating that amorphous state of ADG with stronger energy than crystal ADG
the ─OH of ADG interacted with C O group of Soluplus. As for can be better wetted and released from solid dispersion carrier.
ADG solid dispersion, both ─OH and C O groups originated from Further, compared to physical mixing ADG with Soluplus, solid
Soluplus, which possibly demonstrated that ─OH group of Soluplus dispersion technique was effectively in enhancing molecular
had molecular interaction with C O groups of ADG apart from the interaction and performing functions.
hydrogen bonding formed between ─OH group of ADG with C O
group of Soluplus. In this case, stronger molecular interaction can be
Drug Dissolution and Wetting Property
formed for ADG solid dispersion.
The drug dissolution result was shown in Figure 7(a). It was clear
that ADG solid dispersion formulated by different amount of Sol-
uplus was able to improve drug release owing to the drug amorphous
phase in solid dispersion evidenced by XRD analysis. The amor-
phous ADG had stronger energy to be dissolved than raw ADG
because crystal ADG was harder to be wetted and dissolved, leading

Figure 5. IR spectra of raw ADG, Soluplus, ADG solid dispersions and ADG Figure 6. XRD patterns of raw ADG, Soluplus, ADG solid dispersions and
physical mixtures. [Color figure can be viewed at wileyonlinelibrary.com] ADG physical mixtures. [Color figure can be viewed at wileyonlinelibrary.com]

48354 (5 of 7) J. APPL. POLYM. SCI. 2019, DOI: 10.1002/APP.48354


ARTICLE WILEYONLINELIBRARY.COM/APP

Figure 7. The results of drug release (a) and contact angle (b,c) of samples. (c) The magnifying line chart of contact angle of ADG solid dispersions. [Color
figure can be viewed at wileyonlinelibrary.com]

to enhanced drug dissolution of ADG solid dispersion (the drug property of ADG solid dispersion of 1:7 was the best among these
release of ADG solid dispersion was at least three times higher than formulations [Figure 7(c)], giving evidence for its highest drug
ADG). Among all these formulations, ADG-Soluplus solid disper- dissolution. After fitting the dynamic wetting profiles (Table III),
sion of 1:7 turned out to be the best with maximum cumulative the fitting equations of ADG solid dispersions were all binomial
release amount of more than 80%, while others (ADG-Soluplus solid expressions, which turned to be different from fitting equations
dispersion with weight ratio between drug and carrier of 1:5, 1:9, and of ADG (y = −1.561ln(x) + 72.19) and Soluplus
1:12) were in the range of 60–70%. The wetting property study will (y = 22.754x−0.117). It demonstrated that ADG solid dispersion
address the further reason why ADG solid dispersion of 1:7 was the was able to improve wetting property by lowing contact angle of
best formulation. ADG and changing fitting equation type. All these contributions
were made due to strong hydrogen bonding formed by ADG and
Figure 7(b) described dynamic wetting process of ADG, Soluplus,
Soluplus with capacity to convert crystal ADG to amorphous
mixture of ADG and Soluplus, and ADG solid dispersions. Over-
ADG. Furthermore, the dynamic contact angle measurement
all, the contact angle of dissolution medium on tablets gradually
turned out to be an effective method to reflect the binding affin-
decreased with time. The extreme high contact angle of ADG
ity between drug and carrier and to elucidate the dissolution
reflected that it was hard to be wetted. On the contrary, Soluplus
mechanism of solid dispersion. The final results demonstrated
can be easily wetted using dissolution medium. The contact angle
that Soluplus was the best choice for preparing ADG solid disper-
profiles of physical mixture of ADG and Soluplus were lower
sion. The best drug/polymer ratio can be determined to 1:7. The
than ADG but higher than ADG-Soluplus solid dispersions, dem-
interface wetting and dissolution of this formulation turned out
onstrating that (1) the adding of Soluplus was favorable in
to be optimal ADG solid dispersion, which is certain to have
enhancing wetting property of ADG and (2) solid dispersion was
huge value for industrialization development of ADG. The eluci-
an effective method to get better ADG wetting property than
dation of working mechanism of ADG solid dispersion provided
physical mixture. After forming solid dispersion, the contact
valuable instruction when screening proper excipient.
angle profiles were between ADG and physical mixtures, showing
that the wetting property was enhanced compared with ADG and
so did its drug dissolution. According to the previous discussion CONCLUSIONS
in the section “Superior polymer for ADG solid dispersion,” the
In summary, ADG solid dispersion was prepared using hot-melt
dissolution mechanism of ADG solid dispersion formulated by
extrusion method. PEG 6000, F68, Soluplus, and PVP VA64 were
Soluplus belonged to carrier-controlled mechanism. It was obvi-
initially screened as excipient for ADG solid dispersion. These
ous that the contact angles of ADG solid dispersions prepared
results confirmed that the strongest molecular binding energy of
using different ratios of Soluplus were significantly lower than
ADG with Soluplus contributed to carrier-controlled mechanism
ADG and physical mixtures, demonstrating that these solid dis-
and led to its highest drug dissolution. Soluplus as solid disper-
persions all fitted to carrier-controlled mechanism. The wetting
sion excipient could significantly improve drug absorption. ADG
solid dispersion formulated by Soluplus with various weight
ratios was further focused. According to XRD measurement,
Table III. The Fitting Wquations of ADG, Soluplus, and ADG Solid
amorphous ADG was remained in solid dispersion and led to the
Dispersions
enhancement of drug release. ADG solid dispersion of 1:7 with
best ability to be wetted turned out to be the best ADG solid dis-
Sample Fitting curve R2
persion. It is believed that the elucidation of working mechanism
ADG y = −1.561ln(x) + 72.19 0.9479 of ADG solid dispersion provided valuable instruction for the
Soluplus y = 22.754x−0.117 0.8299 development of solid dispersion technology.
1:5 y = 0.0804x2–1.5167x + 51.847 0.9895
1:7 y = 0.1241x2–2.1005x + 50.525 0.9757
ACKNOWLEDGMENTS
2
1:9 y = 0.0091x –0.4847x + 47.667 0.9785
This research was supported by Youth Found of National Natural
1:12 y = 0.0189x2–0.6238x + 47.744 0.8911
Science Foundation (81700050).

48354 (6 of 7) J. APPL. POLYM. SCI. 2019, DOI: 10.1002/APP.48354


ARTICLE WILEYONLINELIBRARY.COM/APP

REFERENCES 24. Bothiraja, C.; Shinde, M. B.; Rajalakshmi, S.; Pawar, A. P.


1. Liu, L.; Yan, Y.; Zheng, L.; Jia, H.; Han, G. Nat. Prod. Res. J. Pharm. Pharmacol. 2009, 61, 1465.
2019, 1, https://doi.org/10.1080/14786419.2018.1501689. 25. Alagdar, G.; Oo, M. K.; Sengupta, P.; Mandal, U. K.;
2. Tao, L.; Zhang, L.; Gao, R.; Jiang, F.; Cao, J.; Liu, H. Front. Jaffri, J. M.; Chatterjee, B. Int. J. Pharm. Invest. 2017,
Neurosci. 2018, 12, 657. 7, 142.
3. Ekalaksananan, T.; Sookmai, W.; Fangkham, S.; 26. Ogawa, N.; Hiramatsu, T.; Suzuki, R.; Okamoto, R.;
Pientong, C.; Aromdee, C.; Seubsasana, S.; Kongyingyoes, B. Shibagaki, K.; Fujita, K.; Takahashi, C.; Kawashima, Y.;
Yamamoto, H. Eur. J. Pharm. Sci. 2018, 111, 205.
Nutr. Cancer. 2015, 67, 687.
27. Higashi, K.; Seo, A.; Egami, K.; Otsuka, N.; Limwikrant, W.;
4. Wintachai, P.; Kaur, P.; Lee, R. C.; Ramphan, S.;
Yamamoto, K.; Moribe, K. J. Pharm. Pharmacol. 2016,
Kuadkitkan, A.; Wikan, N.; Ubol, S.; Roytrakul, S.; Chu, J. J.;
68, 655.
Smith, D. R. Sci. Rep. 2015, 5, 14179.
28. Ueda, K.; Kanaya, H.; Higashi, K.; Yamamoto, K.;
5. Yan, Y. Y.; Shi, G. X.; Shao, J.; Wang, T. M.; Wang, C. Z.
Moribe, K. Int. J. Pharm. 2018, 538, 57.
Zhongguo Zhong Yao Za Zhi. 2013, 38, 3819.
29. Ma, Y.; Yang, Y.; Xie, J.; Xu, J.; Yue, P.; Yang, M. Int.
6. Khan, I.; Khan, F.; Farooqui, A.; Ansari, I. A. Nutr. Cancer.
J. Nanomed. 2018, 13, 3763.
2018, 70, 787.
30. Wu, Q.; Kennedy, M. T.; Nagapudi, K.; Kiang, Y. H. Int.
7. Peng, Y.; Wang, Y.; Tang, N.; Sun, D.; Lan, Y.; Yu, Z.;
J. Pharm. 2017, 521, 1.
Zhao, X.; Feng, L.; Zhang, B.; Jin, L.; Yu, F.; Ma, X.; Lv, C.
J. Exp. Clin. Cancer Res. 2018, 37, 248. 31. Lavra, Z. M.; Pereira, D. S. D.; Re, M. I. Drug Dev. Ind.
Pharm. 2017, 43, 42.
8. Zhai, Z.; Qu, X.; Li, H.; Ouyang, Z.; Yan, W.; Liu, G.;
Liu, X.; Fan, Q.; Tang, T.; Dai, K.; Qin, A. Mol. Med. Rep. 32. Penumetcha, S. S.; Gutta, L. N.; Dhanala, H.; Yamili, S.;
2015, 11, 1139. Challa, S.; Rudraraju, S.; Rudraraju, S.; Rudraraju, V. Drug
Dev. Ind. Pharm. 2016, 42, 1609.
9. Ji, X.; Li, C.; Ou, Y.; Li, N.; Yuan, K.; Yang, G.; Chen, X.;
Yang, Z.; Liu, B.; Cheung, W. W.; Wang, L.; Huang, R.; 33. Patil, H.; Tiwari, R. V.; Repka, M. A. AAPS PharmSciTech.
Lan, T. Mol. Cell. Endocrinol. 2016, 437, 268. 2016, 17, 20.
10. Dai, J.; Lin, Y.; Duan, Y.; Li, Z.; Zhou, D.; Chen, W.; 34. Agrawal, A.; Dudhedia, M.; Deng, W.; Shepard, K.;
Wang, L.; Zhang, Q. Q. Int. J. Biol. Sci. 2017, 13, 660. Zhong, L.; Povilaitis, E.; Zimny, E. AAPS PharmSciTech.
2016, 17, 214.
11. Guo, X.; Zhao, M.; Lin, Y.; Chen, W.; Wang, S.; Dai, J.
Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2018, 43, 821. 35. Barea, S. A.; Mattos, C. B.; Cruz, A. C.; Chaves, V. C.;
Pereira, R. N.; Simoes, C. M.; Kratz, J. M.; Koester, L. S.
12. Das, S.; Pradhan, G. K.; Das, S.; Nath, D.; Das, S. K. Chem.
Drug Dev. Ind. Pharm. 2017, 43, 511.
Biol. Interact. 2015, 242, 281.
36. Dinunzio, J. C.; Brough, C.; Hughey, J. R.; Miller, D. A.;
13. Mishra, N.; Yadav, K. S.; Rai, V. K.; Yadav, N. P. AAPS
Williams, R. R.; McGinity, J. W. Eur. J. Pharm. Biopharm.
PharmSciTech. 2017, 18, 381.
2010, 74, 340.
14. Akbarsha, M. A.; Murugaian, P. Phytother. Res. 2000, 14, 432.
37. Hengsawas, S. S.; Keen, J. M.; Huang, S.; Zhang, F.;
15. Tang, C.; Liu, Y.; Wang, B.; Gu, G.; Yang, L.; Zheng, Y.; McGinity, J. W.; Williams, R. R. Drug Dev. Ind. Pharm.
Qian, H.; Huang, W. Arch. Pharm. (Weinheim). 2012, 345, 647. 2017, 43, 797.
16. Uttekar, M. M.; Das, T.; Pawar, R. S.; Bhandari, B.; 38. Zeng, C.; Jiang, W.; Tan, M.; Yang, X.; He, C.; Huang, W.;
Menon, V.; Nutan; Gupta, S. K.; Bhat, S. V. Eur. J. Med. Xing, J. Eur. J. Pharm. Sci. 2016, 85, 123.
Chem. 2012, 56, 368. 39. Li, J.; Fan, N.; Wang, X.; Li, C.; Sun, M.; Wang, J.; Fu, Q.;
17. Parveen, R.; Ahmad, F. J.; Iqbal, Z.; Samim, M.; Ahmad, S. He, Z. Eur. J. Pharm. Sci. 2017, 106, 244.
Drug Dev. Ind. Pharm. 2014, 40, 1206. 40. Li, J.; Guo, Y. Mater. Sci. Eng. C Mater. Biol. Appl. 2017,
18. Zhang, Y.; Hu, X.; Liu, X.; Dandan, Y.; Di, D.; Yin, T.; 73, 670.
Zhang, S.; Tang, X. Int. J. Pharm. 2015, 493, 214. 41. Li, J.; Wang, X.; Li, C.; Fan, N.; Wang, J.; He, Z.; Sun, J.
19. Yen, C. C.; Chen, Y. C.; Wu, M. T.; Wang, C. C.; Wu, Y. T. Mol. Pharm. 2017, 14, 2781.
Int. J. Nanomed. 2018, 13, 669. 42. Guo, L.; Kang, L.; Liu, X.; Lin, X.; Di, D.; Wu, Y.; Kong, D.;
20. Zhang, T.; Zhu, L.; Li, M.; Hu, Y.; Zhang, E.; Jiang, Q.; Deng, Y.; Song, Y. Eur. J. Pharm. Sci. 2017, 104, 13.
Han, G.; Jin, Y. Mol. Pharm. 2017, 14, 1718. 43. Ansari, M. T.; Pervez, H.; Shehzad, M. T.; Mahmood, Z.;
21. Li, M.; Zhang, T.; Zhu, L.; Wang, R.; Jin, Y. Int. J. Pharm. Razi, M. T.; Ranjha, N. M.; Khanum, N. Pak. J. Pharm. Sci.
2017, 528, 163. 2012, 25, 447.
22. Suo, X. B.; Zhang, H.; Wang, Y. Q. Biomed. Chromatogr. 44. Obaidat, R.; Alnaief, M.; Jaeger, P. Pharm. Dev. Technol.
2007, 21, 730. 2018, 23, 697.
23. Fan, N.; Ma, P.; Wang, X.; Li, C.; Zhang, X.; Zhang, K.; 45. Zi, P.; Zhang, C.; Ju, C.; Su, Z.; Bao, Y.; Gao, J.; Sun, J.;
Li, J.; He, Z. Carbohydr. Polym. 2018, 199, 492. Lu, J.; Zhang, C. Eur. J. Pharm. Sci. 2019, 134, 233.

48354 (7 of 7) J. APPL. POLYM. SCI. 2019, DOI: 10.1002/APP.48354

You might also like