You are on page 1of 14

nanomaterials

Article
Nanocrystal Suspensions for Enhancing the Oral Absorption
of Albendazole
Zhiwei Liang 1,2 , Min Chen 3 , Yuanyuan Yan 1,2 , Dongmei Chen 1,2,3, * and Shuyu Xie 1,2, *

1 National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan 430070, China
2 MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University,
Wuhan 430070, China
3 MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products,
Huazhong Agricultural University, Wuhan 430070, China
* Correspondence: chendongmei@mail.hzau.edu.cn (D.C.); snxsy1@126.com (S.X.);
Tel.: +86-13100721966 (D.C.); +86-13027163815 (S.X.)

Abstract: Albendazole (ABZ), an effective benzimidazole antiparasitic drug is limited by its poor
solubility and oral bioavailability. In order to overcome its disadvantages, ABZ nanocrystals were
prepared using a novel bottom-up method based on acid-base neutralization recrystallization with
high-speed mixing and dispersing. The cosolvent, stabilizer and preparation temperature were
optimized using single factor tests. The physicochemical properties, solubility and pharmacokinetics
of the optimal ABZ nanocrystals were evaluated. The high-performance liquid chromatography
(HPLC), differential scanning calorimetry (DSC) and X-ray powder diffraction (XRD) showed that
ABZ had no structural and crystal phase change after nanocrystallization. The saturated solubility of
ABZ nanocrystals in different solvents was increased by 2.2–118 fold. The oral bioavailability of the
total active ingredients (ABZ and its metabolites of albendazole sulfoxide (ABZSO) and albendazole
sulfone (ABZSO2)) of the nanocrystals in rats was enhanced by 1.40 times compared to the native
ABZ. These results suggest that nanocrystals might be a promising way to enhance the solubility and
Citation: Liang, Z.; Chen, M.; Yan, Y.; oral bioavailability of ABZ and other insoluble drugs.
Chen, D.; Xie, S. Nanocrystal
Suspensions for Enhancing the Oral Keywords: albendazole; nanocrystals; insolubility; bioavailability; oral absorption
Absorption of Albendazole.
Nanomaterials 2022, 12, 3032.
https://doi.org/10.3390/
nano12173032 1. Introduction
Academic Editor: Rosalia Bertorelli Albendazole (ABZ), a broad-spectrum, highly effective and low toxic benzimidazole
antiparasitic drug, is widely used for treating hydatid cysts and cerebral cysticercosis [1,2].
Received: 2 July 2022
It is the first choice for microsporidiosis in acquired immunodeficiency syndrome (AIDS),
Nanomaterials
Accepted: 2022,
25 August 12, x FOR PEER REVIEW
2022 2 of 15
ascariasis, enterobiasis, hookworm infections and neurocysticercosis [3,4]. It has also been
Published: 1 September 2022
widely used to treat various intestinal and systemic parasitosis in domestic animals and
Publisher’s Note: MDPI stays neutral companion animals [5,6]. After entering into the body, it can be oxidized to albendazole
with regard to jurisdictional claims in improve (ABZSO)
sulfoxide the oral absorption [12–15].oxidized
and then further Among to multiple methods
albendazole used(ABZSO2)
sulfone to improve the solu-
(Figure 1).
published maps and institutional affil- bility of insoluble drugs, nanocrystals has become the preferred strategy [12].
These two main metabolites have insect repellent activity [7]. However, its efficacy is often For exam-
iations. ple, theby
limited nanocrystals prepared by
poor oral absorption our group
mainly due tosignificantly improved
its low aqueous the oral
solubility, bioavailabil-
which severely
ity of its
limits cyadox and
clinical oxfendazole
usage. The oral[16,17]. Some innovative
bioavailability of ABZ in oral
dogsformulations
is only 12.57% based on nano-
and less than
crystals
5% have been
in humans come to market,
[8]. Therefore, and
it is very this technology
important offersthe
to improve more potential
solubility in the future
of albendazole
[18].
for its clinical application.
Copyright: © 2022 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/). Figure1.1.Albendazole
Figure Albendazolemetabolic
metabolicpathway.
pathway.

Some authors have used nanocrystals to increase the solubility, dissolution, oral bio-
availability and efficacy of albendazole and ricobendazole in infected mice, rats and dogs
[19–24]. For example, in a recent report, the nanocrystals
Nanomaterials 2022, 12, 3032. https://doi.org/10.3390/nano12173032 of ABZ enhanced its solubility
https://www.mdpi.com/journal/nanomaterials
by 8.9 fold [22]. A novel nanocrystalline formulation of ABZ obtained by spray drying
Nanomaterials 2022, 12, 3032 2 of 14

At present, many preparation technologies including liposomes [9], β-cyclodextrin


complex [10] and amorphous solid dispersion [11] have been reported to improve the
absorption of ABZ, while none of these ABZ preparations have entered the clinical research
stage. Nanoparticles with below 1000 nm in size could significantly increase the saturated
solubility and dissolution of insoluble drugs by enhancing their surface area and thus
improve the oral absorption [12–15]. Among multiple methods used to improve the solu-
bility of insoluble drugs, nanocrystals has become the preferred strategy [12]. For example,
the nanocrystals prepared by our group significantly improved the oral bioavailability of
cyadox and oxfendazole [16,17]. Some innovative oral formulations based on nanocrystals
have been come to market, and this technology offers more potential in the future [18].
Some authors have used nanocrystals to increase the solubility, dissolution, oral
bioavailability and efficacy of albendazole and ricobendazole in infected mice, rats and
dogs [19–24]. For example, in a recent report, the nanocrystals of ABZ enhanced its
solubility by 8.9 fold [22]. A novel nanocrystalline formulation of ABZ obtained by spray
drying with Poloxamer 188 as a stabilizer demonstrated a cyst inhibition effect of 3.7-fold
greater than that of the commercial ABZ oral product (Albenda) [23]. The nanocrystal-based
formulation also improved the pharmacokinetic performance and therapeutic effects of
ABZ in dogs [21]. It should be noted that nanocrystals cannot only be delivered by oral routs
but also by intravenous injection and other ways according to the clinical requirement [15].
For example, the nanocrystalline formulation of ABZ was intradermally delivered using
dissolving microneedles [25]. Lastly, ABZ nanocrystals were included within 3D printed
tablets or printlets using a melting solidification 3D printing technique, which allowed a
high drug nanocrystal content of up to 50% w/w [26]. In previous reports, the preparation
methods of ABZ nanocrystals included the antisolvent precipitation process [23], high
pressure homogenization (HPH), and nanomilling combing with spray drying [26], etc.
These methods face high production costs (1:1 for drugs and surfactants), organic solvents
(e.g., dichloromethane, dimethyl sulfoxide) and the strict equipment requirements of
HPH [21,27], which might be not easily suitable in industrial production for human and
veterinary clinics. Thus, there arises a need to find a simple, efficient and scalable method
to produce ABZ formulations.
In this study, ABZ nanocrystal suspension was prepared using a novel bottom-up
approach based on acid-base neutralization followed by high-speed mixing and dispers-
ing to easily achieve industrial production. The properties, solubility, dissolution and
pharmacokinetics of ABZ nanocrystal suspension in rats were evaluated.

2. Materials and Methods


2.1. Preparation of ABZ Nanocrystal Suspension
The ABZ nanocrystals suspension was prepared using acid-base neutralization com-
bined with high-speed mixing and dispersing. The schematic representation of the ABZ
nanocrystals preparation is shown in Figure 2. Three gram ABZ were dissolved in 18 mL
solution containing 15 g malic acid at 80 ◦ C under thermostatic magnetic stirrer (Shanghai
Respiratory Analytical Instrument Co., Ltd. 90-1, Shanghai, China). When the drug was
completely dissolved, the heating button of the thermostatic magnetic stirrer was turned off.
Subsequently, 15 mL 1.0 mol/L NaOH solution were slowly added into the acidic aqueous
solution containing ABZ under agitation, followed by adding 5% polyvinyl alcohol (PVA)
5 mL. The recrystallized ABZ was homodispersed using a high-speed dispersing device
(Laboratory digital display, high speed shear emulsion mixer, JRJ300-SH, Shanghai Hushi
Industrial Co., Ltd., Shanghai, China) at the rotational speed of 2000 rpm for 5 min. Finally,
the volume was fixed to 50 mL with sterilized water.
Nanomaterials 2022, 12,
Nanomaterials 2022, 12, 3032
x FOR PEER REVIEW 33 of
of 14
15

2. The schematic representation


Figure 2. representation of
of ABZ
ABZ nanocrystals
nanocrystals preparation.
preparation.

2.2.
2.2. Characterization
Characterization of
of the
the ABZ
ABZ Nanocrystals
Nanocrystals
2.2.1. Scanning Electron Microscopy (SEM)
2.2.1. Scanning Electron Microscopy (SEM)
The morphology of ABZ nanocrystals was determined using SEM to examine the
The morphology of ABZ nanocrystals was determined using SEM to examine the
morphology. The dried nanocrystals sample was placed on a silicon wafer and subsequently
morphology. The dried nanocrystals sample was placed on a silicon wafer and subse-
sprayed with gold via the sputter coater for 8 min. The sprayed thickness was about
quently sprayed with gold via the sputter coater for 8 min. The sprayed thickness was
50 nm, and it was then observed using a scanning electron microscope (High-Tech Co.,
about 50 nm, and it was then observed using a scanning electron microscope (High-Tech
Tokyo, Japan).
Co., Tokyo, Japan).
2.2.2. Determination of Size, Polydispersity Index (PDI) and Zeta Potential of Nanocrystals
2.2.2. Determination of Size, Polydispersity Index (PDI) and Zeta Potential of Nanocrys-
The size, PDI and zeta potential of the nanocrystals were measured using a laser
tals
particle size analyzer. The appropriate amount of ABZ nanosuspension was appropriately
Thewith
diluted size,double
PDI anddistilled
zeta potential
water ofandthemeasured
nanocrystals were
using measured
a laser using
particle a laser
size par-
analyzer
ticle size Malvern,
(ZX3600, analyzer. UK)
The at
appropriate
20 C. Theamount
◦ diameterofwas ABZ nanosuspension
calculated was appropriately
using volume distribution.
diluted
Three with double
repetitions weredistilled
determinedwaterbyand
usingmeasured using a laser
three independent particle size analyzer
batches.
(ZX3600, Malvern, UK) at 20 °C. The diameter was calculated using volume distribution.
2.2.3.
ThreeDifferential
repetitions Scanning Calorimetry
were determined (DSC)
by using three independent batches.
The dried ABZ nanocrystals were determined using DSC. The sample was scanned
2.2.3.
to 300Differential Scanning
◦ C at a scanning rate Calorimetry
of 10 ◦ C/min(DSC)using the 200 pc instrument (NETZSCH, Selbu,
Germany).
The driedThe ABZ
temperature rangewere
nanocrystals included the melting
determined usingpoint
DSC.ofThe
the sample
drug and excipients.
was scanned
Inert gas was introduced into the measurement to purify nitrogen at a pressure
to 300 °C at a scanning rate of 10 °C/min using the 200 pc instrument (NETZSCH, Selbu, of 20 psi
and a flow rate of 50 mL/min. Each experiment was performed in triplicate.
Germany). The temperature range included the melting point of the drug and excipients.
Inert gas was introduced into the measurement to purify nitrogen at a pressure of 20 psi
2.2.4.
and a X-ray Diffraction
flow rate (XRD) Study
of 50 mL/min. Each experiment was performed in triplicate.
X-ray diffraction analysis was carried out for the bulk drug and for the prepared
nanocrystals using X-ray
2.2.4. X-ray Diffraction diffractometer
(XRD) Study (D2 phaser Make: Bruker, Karlsruhe, Germany).
The powder was scanned from 5 to 40 ◦ (2θ).
X-ray diffraction analysis was carried out for the bulk drug and for the prepared
nanocrystals using X-ray diffractometer (D2 phaser Make: Bruker, Karlsruhe, Germany).
2.2.5. Sedimentation Rate, Redispersibility and pH
The powder was scanned from 5 to 40° (2θ).
To determine the sedimentation rate, the original height (H0) of 50 mL nanosuspension
in a 100 mL measuring
2.2.5. Sedimentation cylinder
Rate, was recorded
Redispersibility and pHafter shaking. After standing for 3 h, the
height (H) was measured again. The sedimentation rate was calculated according to the
To determine
following formula: the sedimentation rate, the original height (H0) of 50 mL nanosuspen-
sion in a 100 mL measuring cylinder was F = recorded
H/H0. after shaking. After standing for 3 h,
the height (H) was measured again. The sedimentation rate was calculated according to
The redispersibility
the following formula: of the nanosuspension was measured using redispersion time
after its sedimentation. Briefly, 50 mL nanosuspension was placed in a 100 mL measuring
F = H/H0.
cylinder for several days to produce sedimentation, then the layered nanosuspension was
shaken under a magnetic oscillator rotating at 20 r/min
The redispersibility of the nanosuspension was measured to calculate its redispersion
using redispersiontime.
time
After sample was prepared, the pH of ABZ nanosuspension was
after its sedimentation. Briefly, 50 mL nanosuspension was placed in a 100 mLtested withmeasuring
the aid of
acylinder
pH meter for (FE28,
severalShanghai Mettlersedimentation,
days to produce Toledo Co., Ltd., Shanghai,
then China)
the layered according towas
nanosuspension the
instructions. The probe was washed with distilled water before and after pH measurement,
shaken under a magnetic oscillator rotating at 20 r/min to calculate its redispersion time.
and the pHsample
After was measured three times
was prepared, to eliminate
the pH errors.
of ABZ nanosuspension was tested with the aid
of a pH meter (FE28, Shanghai Mettler Toledo Co., Ltd., Shanghai, China) according to

Nanomaterials 2022, 12, x. https://doi.org/10.3390/xxxxx www.mdpi.com/journal/nanomaterials


Nanomaterials 2022, 12, 3032 4 of 14

2.2.6. Content Determination


The 0.1 mL of three different batches of ABZ nanosuspension were accurately trans-
ferred, dissolved in N, N-Dimethylformamide (DMF) with the aid of ultrasound for 5 min
and diluted with mobile phase to prepare a test solution. The sample, after filtration through
a 0.22-micron filter, was determined using high performance liquid chromatography (HPLC,
Waters 2695 series), and the actual content was calculated using standard curve.

2.3. Solubility
The ABZ nanosuspension was lyophilized with a freeze dryer (Labconco, MO, USA)
for 48 h to determine the saturation solubility of nanocrystals. Excess lyophilized ABZ
nanocrystals and native ABZ were added into 20 mL of 25 ± 5 ◦ C double distilled water,
methanol, acetonitrile and DMF and then achieved dissolution equilibrium while oscillating
in the shaker at 25 ◦ C for 72 h (17). The supersaturated solution was centrifuged at
10,000 r/min (Hitachi Centrifugation CR21 GIII; Hitachi Koki Co., Ltd., Tokyo, Japan) for
10 min, and then the supernatant was collected for analysis using HPLC after filtration
through a 0.22-micron filter.

2.4. In Vitro Dissolution


According to the guidelines of Veterinary Pharmacopoeia of the People’s Republic of
China 2020, in vitro release of ABZ nanosuspensions was performed in PBS at pH = 2 and
pH = 8, respectively, using a paddle dissolution method (dissolution tester, RC806, Tianjin
Tianda Tianfu Technology Co., Ltd., Tianjin, China). The 50 µL of ABZ nanosuspension
(containing 5 mg of ABZ) and 5 mg of ABZ bulk drug were injected into a dissolution
vessel containing 1000 mL of PBS (pH 2.0, 8.0) solution preheated at 37 ◦ C, respectively.
The release medium after adding of drugs was placed at a temperature of (37 ± 0.5) ◦ C at a
speed of 60 r/min, and a certain volume (1 mL) of test solution was taken at 5, 15, 30, 60,
120, 240 and 360 min. The same volume of fresh PBS solution was re-added to maintain a
constant volume at each collect time point. The sample solution was filtered by 0.22 µM
filter and then injected into HPLC to determine the concentration of ABZ. Then cumulative
release percentage of ABZ nanosuspension and native ABZ vs. time was plotted.

2.5. Pharmacokinetics Study


The pharmacokinetics experiment was performed according to “Guidelines of the
Care and Use of Laboratory Animals” of Huazhong Agricultural University and approved
by the Ethics Committee of Huazhong Agricultural University. Twelve healthy SD rats
(half male and half female) with a uniform weight (300 ± 50 g) were used to evaluate the
pharmacokinetics of ABZ nanosuspension. The rats were randomly divided into 2 groups
with 6 rats in each group. The rats were deprived of food and water for 12 h before the
start of experiment. Each group was randomly orally administered the prepared ABZ
nanosuspension and bulk ABZ at the dose of 100 mg/kg, respectively. At the fixed time
points of 0.5, 1, 2, 4, 6, 8, 12, 24, 36, 48, 72 and 96 h after administration, blood samples
(0.3 mL) were taken from heart into 1.5 mL centrifuge tube. The plasma was obtained by
centrifugation at 4000 g/min for 2 min after standing for 30 s and then frozen in −20 ◦ C
refrigerator before usage.

2.6. HPLC Assay


All the samples were analyzed using HPLC (Waters 2695 series) combined with an
ultraviolet detector system with the wavelength set at 295 nm. For in vitro study, the
chromatographic separation was achieved with an analytical ZORBAX SB C18 column
(250 × 4.6 mm, i.d. 5 µm; Agilent Technology, Palo Alto, CA, USA) at 30 ◦ C. The mobile
phase was 0.5% acetic acid solution and acetonitrile. The flow rate and injection volume
were 1 mL/min and 50 µL, respectively. The chromatograms of ABZ, ABZSO and ABZSO2
are shown in Figure 3. Linearity of the standard curves for ABZ and its metabolites of
ABZSO and ABZSO2 ranged from 0.025 to 8.0 µg/mL, and the correlation coefficient (r)
4.6 mm, i.d. 5 μm; Agilent Technology,Palo Alto, USA) at 30 °C. The mobile phase was
0.5% acetic acid solution and acetonitrile. The flow rate and injection volume were 1
Nanomaterials 2022, 12, 3032 mL/min and 50 μL, respectively. The chromatograms of ABZ, ABZSO and ABZSO2 5 ofare
14
shown in Figure 3. Linearity of the standard curves for ABZ and its metabolites of ABZSO
and ABZSO2 ranged from 0.025 to 8.0 μg/mL, and the correlation coefficient (r) values
were >0.9994.
values The limit
were >0.9994. Theoflimit
detection (LOD)(LOD)
of detection and the
andlimit of quantitation
the limit (LOQ)
of quantitation werewere
(LOQ) 0.05
μg/mL
0.05 and 0.1
µg/mL andμg/mL, respectively.
0.1 µg/mL, Inter-Inter-
respectively. and intraday relative
and intraday standard
relative deviations
standard were
deviations
<3.0%.
were <3.0%.

Figure 3. The
Figure 3. The chromatogram
chromatogram of
of ABZ,
ABZ, ABZSO
ABZSO and
andABZSO2.
ABZSO2.

For
For plasma
plasma sample
sample treatment,
treatment, 100 μLof
100 µL of0.1
0.1mol/mL
mol/mL ammonia
ammonia solution
solution were
were added
added
into 100 µL serum sample under vortex. After shaking for 1 min, 2 mL
into 100 μL serum sample under vortex. After shaking for 1 min, 2 mL acetonitrileacetonitrile and and
2 mL2
ethyl acetate
mL ethyl were
acetate added
were addedandand
then further
then vortex
further forfor
vortex 1 1min
minfollowed
followedby byultrasonication
ultrasonication
for
for 5 min. The mixture was centrifuged for 10 min at 5000 r/min, and the precipitate
5 min. The mixture was centrifuged for 10 min at 5000 r/min, and the precipitate was
was
re-extracted
re-extracted byby adding
adding 22mL mLethyl
ethylacetate.
acetate.The
The mixed
mixed extraction
extraction solution
solution of two
of two times
times was
was dried under nitrogen at room temperature, and the residue was dissolved with 200 µL
dried under nitrogen at room temperature, and the residue was dissolved with 200 μL
methanol. The resuspension solution was centrifuged for 1 min at 4000 g (Hitachi Centrifu-
methanol. The resuspension solution was centrifuged for 1 min at 4000 g (Hitachi Centrif-
gation CR21 GIII; Hitachi Koki Co., Ltd., Tokyo, Japan), and the supernatant, after filtration
ugation CR21 GIII; Hitachi Koki Co., Ltd., Tokyo, Japan), and the supernatant, after filtra-
through a 0.22-micron filter, was taken for HPLC analysis. Separation of chromatographs
tion through a 0.22-micron filter, was taken for HPLC analysis. Separation of chromato-
was achieved using an Agilent Eclipse XDB-C18 column (250 mm × 4.6 mm i.d., 5 µm) at a
graphs was achieved using an Agilent Eclipse XDB-C18 column (250 mm × 4.6 mm i.d., 5
flow rate of 1 mL/min at 30 ◦ C. The gradient elution mode was set for separation of ABZ
μm) at a flow rate of 1 mL/min at 30 °C. The gradient elution mode was set for separation
and its main metabolites, with the mobile phase A containing 0.5% acetic acid solution and
of ABZ and its main metabolites, with the mobile phase A containing 0.5% acetic acid
mobile phase B containing pure acetonitrile with gradient elution. The gradient elution
solution and mobile phase B containing pure acetonitrile with gradient elution. The gra-
procedure was set as follows: 0 min, 90% A; 8 min, 25% A; 12 min, 90% A; and 15 min, 90%
dient elution procedure was set as follows: 0 min, 90% A; 8 min, 25% A; 12 min, 90% A;
A. The correlation coefficient (r) values of ABZ, metabolite ABZSO and ABZSO2 at different
and 15 min, 90% A. The correlation coefficient (r) values of ABZ, metabolite ABZSO and
concentrations (0.05 µg/mL, 0.1 µg/mL and 0.5 µg/mL) were >0.9994. The LOD and LOQ
ABZSO2
of at different
ABZ, ABZSO and concentrations
ABZSO2 were 0.05 (0.05µg/mL
μg/mL,and
0.1 μg/mL and 0.5
0.1 µg/mL, μg/mL) were
respectively. The>0.9994.
mean
The LOD
plasma and LOQ
recovery of ABZ,
rates ABZSO
of ABZ, andand
ABZSO ABZSO2
ABZSO2were 0.0597.89–112.09%,
were μg/mL and 0.1 μg/mL, respec-
98.06–119.11%
tively.
and The mean plasma
95.82–99.18%, recovery rates of ABZ, ABZSO and ABZSO2 were 97.89–112.09%,
respectively.
98.06–119.11% and 95.82–99.18%, respectively.
2.7. Statistical Analysis
2.7. Statistical Analysis
The data were analyzed via the one-way analysis of variance (ANOVA) using the SPSS
The data program
17.0 Windows were analyzed via Chicago,
(SPSS Co., the one-way analysis
IL, USA). of variance
Significant (ANOVA)
differences using the
and extremely
significant differences were determined as p values of 0.05 and 0.01, respectively. and ex-
SPSS 17.0 Windows program (SPSS Co., Chicago, USA). Significant differences
tremely significant differences were determined as p values of 0.05 and 0.01, respectively.
3. Results
3.1. Effect of Cosolvent on the Stability of ABZ Nanosuspension
3. Results
According
3.1. Effect to the on
of Cosolvent principle of neutralization
the Stability and solubility of ABZ in acid and alkali,
of ABZ Nanosuspension
saturated malic to
According acid,
thecitric acid,of
principle diethylene glycol
neutralization andether (DECS)ofand
solubility ABZ triethylamine were
in acid and alkali,
selected. It was found that it took a very high temperature (90 ◦ C) to completely dissolve
saturated malic acid, citric acid, diethylene glycol ether (DECS) and triethylamine were
ABZ when
selected. It DECS and triethylamine
was found were high
that it took a very usedtemperature
as cosolvents. (90At°C)
thetosame time, unknown
completely dissolve
impurities were detected using HPLC,
ABZ when DECS and triethylamine were used compared asto pure ABZ.
cosolvents. The
At theresults
same show
time, that ABZ
unknown
was not stable at the high temperature of 90 ◦ C (Figure 4). In comparison to citric acid, it
impurities were detected using HPLC, compared to pure ABZ. The results show that ABZ
took less time for ABZ to completely dissolve in malic acid at the temperature of 80 ◦ C.
was not stable at the high temperature of 90 °C (Figure 4). In comparison to citric acid, it
Therefore, malic acid was selected as the cosolvent.
took less time for ABZ to completely dissolve in malic acid at the temperature of 80 °C.
Therefore,
3.2. Effect ofmalic acidon
Stabilizer wastheselected
Stabilityas
of the
ABZcosolvent.
Nanosuspension
According to the early results of our data, polyvinyl pyrrolidone (PVPk30 ), polyvinyl
alcohol (PVA), Tween 80 and hydrogenated castor oil with polyoxymethylene (HEL-40)
were selected as stabilizers to prepare the nanosuspension.
Nanomaterials 2022, 12, x. https://doi.org/10.3390/xxxxx Four different surfactants
www.mdpi.com/journal/nanomaterials
with 5% concentration and saturated malic acid as cosolvent were used to prepare the
nanosuspension. It was found that the suspensions with PVPk30 as surfactant had obvious
Nanomaterials 2022, 12, 3032 6 of 14

stratification phenomenon, while the nanosuspension showed different degrees of agglom-


erations and poor mobility when using HEL-40. The nanosuspension prepared using PVA
and Tween 80 was a uniform milky white suspension and hardly produced sedimentation
(Figure 5). When sanding for (A)
1 week, the nanosuspension stabilized with PVA still did not
Nanomaterials 2022, 12, x FOR PEER REVIEW 6 of for
produce stratification and sedimentation. Therefore, PVA was selected as the stabilizer 15

this test.

(B)
Figure 4. The chromatogram of ABZ (A) and ABZ (B) under high temperature of 90 °C.

3.2. Effect of Stabilizer on the Stability of ABZ Nanosuspension


(A)
According to the early results of our data, polyvinyl pyrrolidone (PVPk30), polyvinyl
alcohol (PVA), Tween 80 and hydrogenated castor oil with polyoxymethylene (HEL-40)
were selected as stabilizers to prepare the nanosuspension. Four different surfactants with
5% concentration and saturated malic acid as cosolvent were used to prepare the nano-
suspension. It was found that the suspensions with PVPk30 as surfactant had obvious strat-
ification phenomenon, while the nanosuspension showed different degrees of agglomer-
ations and poor mobility when using HEL-40. The nanosuspension prepared using PVA
and Tween 80 was a uniform milky white suspension and hardly produced sedimentation
(Figure 5). When sanding for (B) 1 week, the nanosuspension stabilized with PVA still did
not produce stratification and sedimentation. Therefore, PVA was selected as the stabi-
Figure 4. The chromatogram of ABZ (A) and ABZ (B) under high temperature of 90 °C.
Figure 4. The
lizer for thischromatogram
test. of ABZ (A) and ABZ (B) under high temperature of 90 ◦ C.

3.2. Effect of Stabilizer on the Stability of ABZ Nanosuspension


According to the early results of our data, polyvinyl pyrrolidone (PVPk30), polyvinyl
alcohol (PVA), Tween 80 and hydrogenated castor oil with polyoxymethylene (HEL-40)
were selected as stabilizers to prepare the nanosuspension. Four different surfactants with
5% concentration and saturated malic acid as cosolvent were used to prepare the nano-
suspension. It was found that the suspensions with PVPk30 as surfactant had obvious strat-
ification phenomenon, while the nanosuspension showed different degrees of agglomer-
ations and poor mobility when using HEL-40. The nanosuspension prepared using PVA
and Tween 80 was a uniform milky white suspension and hardly produced sedimentation
(Figure 5). When sanding for 1 week, the nanosuspension stabilized with PVA still did
not produce stratification and sedimentation. Therefore, PVA was selected as the stabi-
lizer for this test.
Figure 5. The appearance of the nanosuspension prepared by different stabilizers. (A): PVP, (B): PVA,
Figure 5. The appearance of the nanosuspension prepared by different stabilizers. (A): PVP, (B):
(C):
PVA,Tween 80; (D):80;
(C): Tween HEL-40 as stabilizers.
(D): HEL-40 as stabilizers.
3.3. Properties of ABZ Nanosuspensions
3.3. Properties of ABZ Nanosuspensions
The optimization formulation condition and properties of ABZ nanocrystal is shown
The1.optimization
in Table formulation
ABZ nanosuspension wascondition
a uniformand propertiesofofa ABZ
suspension milkynanocrystal
white color.is Under
shown
SEM, the shape of the nanocrystals were elongated and evenly dispersed (Figure Under
in Table 1. ABZ nanosuspension was a uniform suspension of a milky white color. 6). In
SEM, thethe
addition, shape of thehydration
average nanocrystals
size were elongated was
of nanocrystals and 508
evenly dispersed
± 11.869 (Figure
nm with a PDI 6). of
In
addition, the average hydration size of nanocrystals was 508 ± 11.869 nm with
0.28 ± 0.01 and zeta potential of 2.30 ± 0.44 mV. ABZ contents and sedimentation rate as a PDI of
well as pH values of the nanosuspension were 10%, 1 and 3.5, respectively. The recovery
rates of ABZ from ABZ nanocrystals were 98.5–102.3%. The redispersion time was only
27 ± 1.0 s in the case of the layered ABZ suspension under the magnetic shaker at a rotating
rate of 20 r/min.
Figure 5. The appearance of the nanosuspension prepared by different stabilizers. (A): PVP, (B):
PVA, (C): Tween 80; (D): HEL-40 as stabilizers.

3.3. Properties of ABZ Nanosuspensions


The optimization formulation condition and properties of ABZ nanocrystal is shown
Nanomaterials 2022, 12, x FOR PEER REVIEW 7 of 15
Nanomaterials 2022, 12, 3032 7 of 14

0.28 ±1.0.01
Table and zeta formulation
Optimization potential ofcondition
2.30 ± 0.44
and mV. ABZ of
properties contents and sedimentation rate as
ABZ nanocrystal.
well as pH values of the nanosuspension were 10%, 1 and 3.5, respectively. The recovery
ABZ PVA rates
Malic of ABZ
Acid from ABZ nanocrystals
Temperature Speed were 98.5–102.3%.
Size (nm) The redispersion
ZP (mv) time PDI
was only
3g 0.25 g 27 ±g1.0 s in the80
15.0 case
◦ C of the layered ABZmin
2000 rpm/5 suspension
508 under
± 11.9 the magnetic shaker0.28
2.30 ± 0.44 at a±rotating
0.01
rate of 20 r/min.

Figure6.
Figure 6. SEM
SEM photomicrograph
photomicrographof
ofalbendazole
albendazolenanoparticles
nanoparticles((×100,000).
×100,000).

3.4.
TableIdentification
1. Optimization formulation condition and properties of ABZ nanocrystal.
ABZ
The retention
PVA
time of ABZ nanocrystals and
Malic Acid Temperature
ABZ standard
Speed
was determined
Size (nm) ZP (mv)
to PDI
be the
same 3g using HPLC.
0.25 g There
15.0 gwas no impurity
80 °C peak in the
2000 rpm/5 mindrug extracted
508 ± 11.9 from the nanocrystals.
2.30 ± 0.44 0.28 ± 0.01
It showed that the properties of ABZ nanocrystals prepared by this process were basically
unchanged. The DSC thermogram of ABZ showed that there was a unique endothermic
3.4. Identification
peak of ABZ at 213 ◦ C (Figure 7). The nanocrystals showed a characteristic melting peak
The retention time of ABZ nanocrystals and ABZ standard was determined to be the
of ABZ at 211 ◦ C, and the height of the endothermic peak did not change compared
same using HPLC. There was no impurity peak in the drug extracted from the nanocrys-
to the native drug, which indicated that the crystal form and crystallinity of ABZ in
tals. It showed that the properties of ABZ nanocrystals prepared by this process were ba-
the nanosuspensions did not change compared to the native ABZ. Malic acid showed a
sically unchanged.
characteristic peak at The110DSC
◦ C, thermogram of ABZ showed
while the characteristic peak that there was
vanished in the a unique endo-
nanocrystals.
thermic peak of ABZ at 213 °C (Figure 7). The nanocrystals showed a
This might be due to the fact that malic acid is present in molecular form in the nanocrystals.characteristic melt-
ing peak of ABZ at 211 °C, and the height of the endothermic
The XRD pattern of the native drug and prepared ABZ nanocrystals are shown in Figure peak did not change com-8.
pared
The XRD to the nativedecreased
intensity drug, which at 2indicated that and
theta of “11” the crystal
“18”, andformtheand crystallinity
sharp of ABZ in
peaks disappeared
the nanosuspensions
between 2 theta “20–22”. did not Thischange compared
difference might to be
thedue
native ABZ.
to the Malic acid
inference showed
of the malica
characteristic peak at 110 °C, while the characteristic peak vanished
acid and PVA since only 10% of the drug was present in the lyophilized powder of the in the nanocrystals.
This might beAdue
nanocrystals. to the fact
reduction that intensity
in peak malic acid is present
after in molecularwas
nanocrystallization form in the
also nanocrys-
reported [24].
tals. The XRD pattern of the native drug and prepared ABZ nanocrystals
The disappeared sharp peaks might be due to the relative crystallinity transformation are shown of in
Figure
ABZ 8. The XRD
nanocrystals intensity
covered by decreased at 2 theta in
PVA. As mentioned of the
“11” and “18”,
previous and theABZ
literature, sharphaspeaks
two
disappeared (Forms
polymorphs betweenI and2 theta
II). “20–22”.
Form II isThis difference might
enantiotropically be due
related toto the inference
Form I. As shown of the
in
malic acid and PVA since only 10% of the drug was present in the
Figure 8, ABZ might crystallize into the thermodynamically stable Form II in nanocrystals lyophilized powder of
the nanocrystals.
NC systems [23]. A reduction in peak intensity after nanocrystallization was also reported
[24]. The disappeared sharp peaks might be due to the relative crystallinity transformation
3.5. Equilibrium
of ABZ Solubility
nanocrystals of ABZ
covered byNanosuspension
PVA. As mentioned in the previous literature, ABZ has
two polymorphs
The saturated(Forms I andofII).
solubility ABZForm II is enantiotropically
nanocrystals in water and related to Form
different organic I. As shown
solvents
in Figure
was 8, ABZ
2.2~118.3 foldmight
highercrystallize
than thatinto the thermodynamically
of native ABZ, as shown instable TableForm2. The II solubility
in nanocrys- of
tals NC systems [23].
ABZ nanocrystals in water was 396.172 ± 0.053 µg/mL, which was enhanced 118.3 times
compared to the solubility of native ABZ in water (3.349 ± 0.098 µg/mL).

3.6. In Vitro Dissolution


In vitro dissolution of native ABZ and ABZ nanocrystals is shown in Figure 9. Com-
pared to native ABZ, the release rate and release degree of ABZ nanocrystals at pH = 2
and pH = 8 were significantly increased. When pH = 2, the ABZ nanocrystals released
98.1% of the drug within 5 min, while native ABZ only released 21.0%. When pH = 8, the
ABZ nanocrystals released 85.8% of the drug within 5 min and nearly completely released

Nanomaterials 2022, 12, x. https://doi.org/10.3390/xxxxx www.mdpi.com/journal/nanomaterials


Nanomaterials 2022, 12, 3032 8 of 14

the drug at 30 min, while native ABZ only released 20.5% at 5 min and was close to the
maximum value of 87.9% at 2 h.

Table 2. Solubility of native ABZ and ABZ nanocrystal in various solvents (25 ◦ C, standard atmo-
spheric pressure).

Solubility (µg/mL, Mean ± S.D.)


Solvents Enhanced Folds
Native ABZ ABZ Nanocrystal
Water 3.35 ± 0.10 3967 ± 0.05 b 1189
Ethanol 11.7 ± 0.00 226 ± 0.08 b 19.4
Acetonitrile 31.9 ± 0.30 282 ± 0.24 b 8.80
Nanomaterials
Nanomaterials 2022,
2022, 12,
12, x
x FOR
FOR PEER
PEER REVIEW
REVIEW DMF 7308 ± 0.34 16,078 ± 0.42 b 2.20 8
8 of
of 15
15
b Statistical significances compared to ABZ power are p < 0.01.

Figure
Figure 7.DSC
Figure7.7. DSCof
DSC ofbulk
of bulkdrug
bulk drugand
drug andalbendazole
and albendazolenanocrystals.
albendazole nanocrystals.
nanocrystals.

Figure
Figure8.8.
Figure XRD
8.XRD pattern
XRDpattern (B)
pattern(B) of
(B)of bulk
ofbulk drug
bulkdrug and
drugand albendazole
andalbendazole nanocrystals.
albendazolenanocrystals.
nanocrystals.

3.5. Equilibrium
3.5. Equilibrium Solubility
Solubility of
of ABZ
ABZ Nanosuspension
Nanosuspension
The saturated
The saturated solubility
solubility of
of ABZ
ABZ nanocrystals
nanocrystals in
in water
water and
and different
different organic
organic solvents
solvents
was
was 2.2~118.3 fold higher than that of native ABZ, as shown in Table 2. The solubility of
2.2~118.3 fold higher than that of native ABZ, as shown in Table 2. The solubility of
ABZ
ABZ nanocrystals
nanocrystals inin water
water was
was 396.172
396.172 ±± 0.053
0.053 μg/mL,
μg/mL, which
which waswas enhanced
enhanced 118.3
118.3 times
times
compared
compared to to the
the solubility
solubility of
of native
native ABZ
ABZ inin water
water (3.349
(3.349 ±± 0.098
0.098 μg/mL).
μg/mL).

Table
Table 2.
2. Solubility
Solubility of
of native
native ABZ
ABZ and
and ABZ
ABZ nanocrystal
nanocrystal in
in various
various solvents (25 °C,
solvents (25 °C, standard
standard atmos-
atmos-
pheric pressure).
pheric pressure).
In vitro dissolution of native ABZ and ABZ nanocrystals is shown in Figure 9. Com-
pared to native ABZ, the release rate and release degree of ABZ nanocrystals at pH = 2
and pH = 8 were significantly increased. When pH = 2, the ABZ nanocrystals released
98.1% of the drug within 5 min, while native ABZ only released 21.0%. When pH = 8, the
Nanomaterials 2022, 12, 3032 ABZ nanocrystals released 85.8% of the drug within 5 min and nearly completely released
9 of 14
the drug at 30 min, while native ABZ only released 20.5% at 5 min and was close to the
maximum value of 87.9% at 2 h.

(A) (B)
Figure
Figure9.9.In vitro dissolution
In vitro curves
dissolution of native
curves albendazole
of native and albendazole.
albendazole Nanocrystals
and albendazole. in (A) pHin
Nanocrystals
=(A)
2.0 pH
PBS=and (B) pH = 8.0 PBS (Mean ± SD, n = 6).
2.0 PBS and (B) pH = 8.0 PBS (Mean ± SD, n = 6).

3.7.Pharmacokinetics
3.7. PharmacokineticsofofABZ
ABZNanocrystals
Nanocrystals
Theplasma
The plasmaconcentrations
concentrationsofofthe theABZ
ABZprototype
prototypeand anditsitsactive
activemetabolites
metabolites(ABZSO (ABZSO
andABZSO2)
and ABZSO2) vs. vs. time
time curves for for ABZ
ABZnanocrystals
nanocrystalsand andnative
native ABZ
ABZ after gastric
after administra-
gastric admin-
tion are are
istration shownshownin Figure 10. 10.
in Figure TheThepeak concentration
peak concentration of ABZ
of ABZprototype
prototypein nanocrystals
in nanocrystals and
the the
and native
nativeABZABZ group rapidly
group rapidlyreached
reached 0.66 ± 0.19
0.66 ± 0.19 μg/mLatat0.5
µg/mL 0.5hhand 0.61±
and0.61 0.06 μg/mL
± 0.06 µg/mL
0.60± ±
atat0.60 0.220.22
h, h, respectively.
respectively. TheyTheythenthen slowly
slowly decreased
decreased andandwerewere
lowerlower
thanthan the de-
the detec-
tection limit after staying in plasma for 96 and 72 h, respectively.
tion limit after staying in plasma for 96 and 72 h, respectively. The ABZSO, the first me- The ABZSO, the first
metabolite
tabolite of ABZ,
of ABZ, reached
reached the peak
the peak concentration
concentration of 2.08
of 2.08 ± 0.38
± 0.38 μg/mL µg/mL
at 4.67 1.03±h1.03
at ±4.67 andh
1.18 0.16±μg/mL
and±1.18 0.16 µg/mL
at 5.00 at 5.00h±in1.15
± 1.15 the hnanocrystals
in the nanocrystals
and nativeandABZnative ABZ groups,
groups, respec-
respectively.
tively.
The The second
second metabolitemetabolite
ABZSO2 ABZSO2 in the nanocrystals
in the nanocrystals and native
and native ABZ groups
ABZ groups reachedreached
the
the peak concentrations of 1.71 ± 0.30 µg/mL at 7.33 ±
peak concentrations of 1.71 ± 0.30 μg/mL at 7.33 ± 1.03 h and 1.03 ± 0.11 μg/mL at 8.00 h,at
1.03 h and 1.03 ± 0.11 µg/mL
8.00 h, respectively.
respectively. Both metabolites
Both metabolites in the native
in the native ABZ and ABZnanocrystals
and nanocrystalsgroups groups gradually
gradually de-
creased and they stayed in the plasma for more than 96 h. The total compounds ininnative
decreased and they stayed in the plasma for more than 96 h. The total compounds native
ABZand
ABZ andABZABZnanocrystals
nanocrystals groups
groups reached
reached the the peak
peak concentrations
concentrations of 2.08 ±
of 2.08 0.35 μg/mL
± 0.35 µg/mL
and 3.37±±0.82
and3.37 0.82μg/mL
µg/mL 5.60± ±
atat5.60 1.67
1.67 and
and 5.67
5.67 ±± 1.51
1.51 h, h, respectively.The
respectively. Thepeak
peakconcentration
concentration
of the total compounds of the ABZ nanocrystals was 1.62 fold
of the total compounds of the ABZ nanocrystals was 1.62 fold that of the native ABZ. that of the native ABZ.
The pharmacokinetic parameters are listed in Table 3. The
The pharmacokinetic parameters are listed in Table 3. The absorption of ABZ absorption of ABZ nanocrys-
nano-
tals was increased compared to that of the native ABZ. The area
crystals was increased compared to that of the native ABZ. The area under the concentra- under the concentration–
time curve (AUC0-∞), mean residence time (MRTlast) and elimination half-life (T1/2)
tion–time curve (AUC0-∞), mean residence time (MRTlast) and elimination half-life (T1/2)
of total compounds in the ABZ nanosuspension group were 101.72 ± 20.83 h*µg/mL,
of total compounds in the ABZ nanosuspension group were 101.72 ± 20.83 h*μg/mL, 32.98
32.98 ± 3.33 h and 83.73 ± 22.96 h, respectively, which were 1.40 fold (72.46 ± 6.18 h*µg/mL),
± 3.33 h and 83.73 ± 22.96 h, respectively, which were 1.40 fold (72.46 ± 6.18 h*μg/mL), 1.02
1.02 fold (32.02 ± 3.00 h), 1.04 fold (79.91 ± 12.65 h) compared to the native ABZ group,
fold (32.02 ± 3.00 h), 1.04 fold (79.91 ± 12.65 h) compared to the native ABZ group, respec-
respectively. The AUC0-∞, MRTlast and T1/2 of the ABZ prototype in nanocrystals group
tively. The AUC0-∞, MRTlast and T1/2 of the ABZ prototype in nanocrystals group were
were 16.38 ± 1.8 h*µg/mL, 43.24 ± 4.15 h and 66.78 ± 10.77 h, respectively, which were 1.05,
16.38 ± 1.8 h*μg/mL, 43.24 ± 4.15 h and 66.78 ± 10.77 h, respectively, which were 1.05, 1.23
1.23 and 2.02 times higher than those of the native ABZ group with 14.66 ± 4.04 h*µg/mL,
and 2.02 times higher than those of the native ABZ group with 14.66 ± 4.04 h*μg/mL, 35.17
35.17 ± 3.37 h and 62.40 ± 7.88 h, respectively. The AUC0-∞, MRTlast and T1/2 of ABZSO
± 3.37 h and 62.40 ± 7.88 h, respectively. The AUC0-∞, MRTlast and T1/2 of ABZSO in the
in the ABZ nanocrystals group (46.05 ± 11.08 h*µg/mL, 34.28 ± 2.10 h, 75.79 ± 21.68 h)
ABZ nanocrystals group (46.05 ± 11.08 h*μg/mL, 34.28 ± 2.10 h, 75.79 ± 21.68 h) increased
increased by 1.46 fold (31.58 ± 7.49 h*µg/mL), 1.11 fold (30.88 ± 4.37 h) and 1.13 fold
by 1.46 fold (31.58 ± 7.49 h*μg/mL), 1.11 fold (30.88 ± 4.37 h) and 1.13 fold (67.31 ± 8.24)
(67.31 ± 8.24) compared to the native ABZ group, respectively. The AUC0-∞, MRTlast and
T1/2 of ABZSO2 in the nanocrystals group were 1.17, 1.02 and 1.03 fold than those of the
native ABZ group, respectively.

Nanomaterials 2022, 12, x. https://doi.org/10.3390/xxxxx www.mdpi.com/journal/nanomaterials


Nanomaterials 2022, 12, x FOR PEER REVIEW 10 of 15

Nanomaterials 2022, 12, 3032 compared to the native ABZ group, respectively. The AUC0-∞, MRTlast and T1/2 of AB-
10 of 14
ZSO2 in the nanocrystals group were 1.17, 1.02 and 1.03 fold than those of the native ABZ
group, respectively.

(A) (B)

(C) (D)
Figure 10. The concentration vs. time curves of ABZ (A), ABZSO (B), ABZSO2 (C) and total active
Figure 10. The concentration vs. time curves of ABZ (A), ABZSO (B), ABZSO2 (C) and total active
ingredient (D) in SD rats after oral administration of nanocrystals and native ABZ at a dose of 100
ingredient (D) in SD rats after oral administration of nanocrystals and native ABZ at a dose of
mg/kg body weight.
100 mg/kg body weight.
Table 3. Pharmacokinetic parameters of native ABZ and nanocrystals in SD rats after an oral dose
Table 3. Pharmacokinetic parameters of native ABZ and nanocrystals in SD rats after an oral dose of
of 100 mg/kg (mean ± standard deviation, n = 6).
100 mg/kg (mean ± standard deviation, n = 6).
ABZ ABZSO ABZSO2 Total
Parameters Units ABZ ABZSO ABZSO2 Nanosuspen- Total Nanosuspen-
Parameters Units Native Nanosuspension Native Nanosuspension Native Native Native
Native Nanosuspension Native Nanosuspension sion
Nanosuspension Native sion
Nanosuspension

0.60±±0.22 ± b ± a ± b b
T
Tmaxmax hh 0.60 0.22 0.50
0.50 5.00 ± 1.15
5.00 1.15 4.67 ±a1.03
4.67 ± 1.03 b 8.00 8.00 7.33 ± 1.03
7.33 1.03 a 5.60
5.60 ± 1.67
1.67 5.67
5.67 ±±1.51
1.51
Cmax µg/mL 0.61 ± 0.06 0.66 ± 0.19 1.18 ± 0.16 2.08 ± 0.38 1.03 ± 0.11 1.71 ± 0.30 a 2.08 ± 0.35 3.37 ± 0.82 b
C
AUC0Ý max
∞ μg/mL
h*µg/mL 0.61 ± 0.06
14.7 ± 4.04 0.66 ±
16.4 ± 1.810.19
a 1.18 ±
31.6 ± 7.490.16 2.08
46.1 ± 11.1±a 0.38 a 1.03
30.7 ± 2.25 ± 0.11 1.71 ±
35.9 ± 3.59 0.30
a a 2.08 ± 0.35
725 ± 6.18 3.37 ± 0.82
102 ± 20.8 b b

341±±4.04 b a ± 14.2 ± 16.9±b 11.1 a ± 10.2 ± 2.25 b ±± bb


AUC
VZ 0-∞ h*μg/mL
mL/kg 14.7 85.5 16.4
340 ±±77.9
1.81 31.6
143 ± 7.49 10446.1 125 30.7 35.9
113 ±
± 21.6 3.59 a 725
89,468 6.18
34,099 102
41,904 ±±13,965
20.8
CL mL/h/kg 5.70 ± 1.81 3.42 ± 1.39 a 3.62 ± 0.22 2.22 ± 0.76 a 2.82 ± 0.52 2.35 ± 0.52 a 1101 ± 521 926 ± 223 b
VZ
MRTlast mL/kg
h 34135.2±±85.5
3.37 340
43.2 ± ±4.15
77.9a b 143± ±4.414.2
30.9 34.3104 ± b16.9 b
± 2.10 30.6 125
± 2.59± 10.2 113
31.3 ± 21.6
± 5.11 a b 89,468 ± 34099 41,904
32.0 ± 3.00 33.0 ±±3.33
13965
a b
a a a b
CL
Ke mL/h/kg
1/h 5.70
0.01± ± 1.81
0.001 3.42
0.01 ±±0.002
1.39 3.62
0.010 ± 0.001
± 0.22 0.0102.22 ± b0.76
± 0.003 a ± 0.002± 0.52
0.01 2.82 2.35
0.008 ±
± 0.002 b
0.52 a 1101
0.009 ± 0.001
± 521 926
0.009 ±±0.002
223
T1/2 h 62.4 ± 7.88 66.8 ± 10.8 b 67.3 ± 8.24 75.79 ± 21.68 a 87.82 ± 21.55 90.5 ± 26.4 b 79.9 ± 12.7 83.7 ± 23.0 b
MRTF last %h 35.2 ± 3.37 114.7 43.2 ± 4.15
a 30.9 ± 4.4172 34.3 ± 2.10 b 30.6 ± 2.59113 31.3 ± 5.11 a 32.0 ± 3.00 140 33.0 ± 3.33 a
Ke 1/h 0.01 ± 0.001 0.01 ±time
Tmax, 0.002 a 0.010
to reach ± 0.001 concentration;
maximum 0.010 ± 0.003 Cmax, b 0.01maximum
± 0.002 0.008 ± 0.002 of0.009
concentration
b
drug; ± 0.001
AUC0Ý∞ 0.009 ± 0.002
, area under
a

T1/2 h 62.4 ± 7.88 66.8 ±concentration–time


plasma 10.8 b 67.3 ± 8.24 curve;75.79 ± 21.68 of distribution;
VZ, volume
a 87.82 ± 21.55CL, 90.5 ± 26.4rate; MRTlast,
clearance
b 79.9 ± 12.7 83.7 ± 23.0
mean resident
b
time;
F % 114.7
Ke, elimination rate; T1/2, elimination 172 half-life; F, bioavailability. 113a Statistical significances compared 140 to native
ABZ aretimep < 0.05. b Statistical significances compared to native ABZ are p < 0.01.
Tmax, to reach maximum concentration; Cmax, maximum concentration of drug; AUC0-∞,
area under plasma concentration–time curve; VZ, volume of distribution; CL, clearance rate; MRT-
4. Discussion
last, mean resident time; Ke, elimination rate; T1/2, elimination half-life; F, bioavailability. a Statisti-
cal significances compared toand
Poor oral absorption native
lowABZ are p < 0.05.in
concentration
b Statistical significances compared to native
the blood and liver seriously affect the
ABZ are p < 0.01.
therapy efficacy of ABZ due to its insolubility [28,29]. It is reported that nanocrystalline
technology can reduce the drug particle size to the nanosized range and thus increase the
4. Discussion
dissolution and bioavailability of insoluble ABZ by increasing its specific surface area. Many
Poor oralwith
nanocrystals absorption and low concentration
unique physicochemical in thehave
properties blood andproduced
been liver seriously affect the
as a promising
therapy efficacy of ABZ due to its insolubility [28,29]. It is reported that nanocrystalline
drug delivery system for ABZ (Table 4). The reported production method mainly included
technology
HPH can reduce
combined the drug particle
with spray-drying size toand
processes theannanosized range
antisolvent and thus increase
precipitation the
technique
dissolutionwith
combined anddrying,
bioavailability
which areofnot
insoluble
suitableABZ by increasing
for scale production.its For
specific surface
the HPH area.
method,
Many nanocrystals
contamination due towith unique
the long physicochemical
duration properties
of homogenization haveenergy
and high been produced
productionasarea
the main disadvantages, which directly influence the crystallinity of nanocrystals, and it
is often used in the laboratory. The antisolvent precipitation method is a classical bottom-
up procedure to formulate nanocrystals, employing precipitation by solvent–antisolvent
Nanomaterials 2022, 12, 3032 11 of 14

addition and precipitation using solvent removal [18]. As an organic solvent is employed
during this process, it is not an eco-friendly fabrication method.

Table 4. The reported preparation methods of ABZ nanocrystal.

Method Solvent and Stabilizer Performance Reference


High-pressure homogenization Enhanced the chemoprophylactic and
Water, PVPk30 19
combined with spray drying clinical efficacy
High-pressure homogenization Water, ABZ and P188 AUC values of nano-sized ABZO was
21
combined with drying processes (1:1) enhanced by threefold
High-pressure homogenization AUC of ABZSO was almost twice than
Water, P188 22
combined with spray-drying processes that of the control
Approximately 4.2-fold higher AUC of
Spray drying Dichloromethane, P188 23
ABZSO in Beagle dogs
Antisolvent precipitation technique
Dimethyl sulfoxide Improved the dissolution 24
combined with drying
High pressure homogenization combined The dissolution rate noticeably
Water 25
with and spray-drying increased

In order to promote clinical application, the ABZ nanocrystal suspension was prepared
using a novel bottom-up approach based on acid-base neutralization followed by high-
speed mixing and dispersing to easily achieve industrial production. Firstly, the ABZ
was dissolved in malic acid solution at 80 ◦ C and then rapidly stirred. In the presence
of surfactant, the temperature was reduced, and the same molar amount of cold sodium
hydroxide solution was added at the same time. The neutralization of acid and alkali
resulted in the sudden supersaturation of ABZ in weakly acidic water and the formation
of fine grains. Then the ABZ nanocrystals was treated using high-speed mixing and
dispersing to get uniform nanocrystals. In this process, it is necessary to control the
dissolution temperature of ABZ to prevent its decomposition from producing impurities.
At the same time, as the total surface area of the prepared nanocrystals particles is several
orders of magnitude larger than that of the coarse particles, a large number of surfactant
molecules needed to be added to prevent the aggregation of drug crystals so as to ensure
the stability of nanocrystals [30,31]. It is important to select a suitable stabilizer because it
plays a key role in the behavior of NCs as well as their stability. Therefore, the selection of
cosolvent and surfactant was very important in the preparation of ABZ nanocrystals. The
choice of cosolvent was selected according to the solubility and stability of ABZ. ABZ was
dissolved quickly at 80 ◦ C, and no impurities were generated. Therefore, 80 ◦ C was chosen
as the dissolution temperature. Therefore, among the four selected cosolvents, the saturated
malic acid with the lower melting point was preferred. The selection of surfactants is mainly
influenced by the similar hydrophobicity between drugs and surfactants. It is reported
that similar hydrophobicity can provide better spatial stability [32–36]. In addition, the
changes in pH and surface charge will also destroy the stability of the nanocrystals, leading
to crystal aggregation. It is reported that ion stability with high zeta potential cannot
guarantee the stability of nanocrystals and that the surface neutral surfactant can enhance
the stability of drug nanocrystals. Among the four surfactants of PVPk30, PVA, Tween 80
and HEL-40 selected in this experiment [37], PVA has the longest stabilizing effect, and
thus it was selected as the best surfactant in this experiment.
A high-speed dispersing device can effectively reduce particle size and polydisper-
sity [38–41]. In this study, the best speed and stirring time were optimized. It was found
that the nanocrystal could obtain uniform nanoparticles without obvious agglomeration by
stirring for 5 min under 2000 rpm. It is reported that the small size of nanoparticles is more
conducive to drug absorption and stability [33]. The DSC thermogram showed that the
melting peak and the peak height of ABZ nanocrystals was the same as the endothermic
temperature of ABZ. The HPLC and XRD determination also showed that the molecular
structure was stable, and crystal transformation of the nanosuspension was observed after
Nanomaterials 2022, 12, 3032 12 of 14

recrystallization. These results showed that the preparation process had no adverse effect
on the drug structure. The saturated solubility of ABZ in different solvents significantly
increased when ABZ was prepared into nanocrystals. The increase in saturated solubility
may be due to the large increase in specific surface area and the decrease in particle size,
resulting in the large increase in dissolution rate [42]. The dissolution rate of the ABZ
nanocrystals also significantly increased. It is reported that the dissolution rate will be very
fast when the particle size is less than 1 µm [33].
ABZ can be rapidly metabolized into ABZSO in vivo, and then ABZSO can be me-
tabolized into ABZSO2, among which ABZSO has the highest blood concentration. It has
been reported that ABZSO2 has a similar effect to ABZSO on intestinal and nonintestinal
trichinella spiralis, but ABZSO2 has no effect on Echinococcus granulosus [43]. At the same
time, most of the literature only detected the metabolite of ABZSO and no ABZ prototype
and ABZSO2. In this study, ABZ and its two metabolites were all determined, and the
respective pharmacokinetic parameters of each active ingredient and the total of the three
active ingredients were calculated. The results show that the ABZSO concentration was
the highest in both the native ABZ and ABZ nanocrystals groups, which is consistent with
the previous literature [44,45]. The time to reach peak concentration of ABZ, ABZSO and
ABZSO2 in the nanocrystals group and the total compounds was earlier for all than that
of the native ABZ, and the maximum blood concentration was also higher than that of
the native ABZ group. Compared to the native ABZ group, the bioavailability of ABZ,
ABZSO, ABZSO2 and the totals of the three active compounds in the nanocrystals group
increased by 1.14, 172, 1.12 and 1.40 fold, respectively. The increased bioavailability and
absorption rate may be due to the decrease in particle size and the increase in saturated
solubility and dissolution rate. The huge specific surface area of nanocrystals with small
particle sizes will increase the adhesiveness of drugs to the gastrointestinal mucosa, thus
increasing their absorption and residence time in the gastrointestinal tract [46]. In future
research, the size of nanocrystals should be further reduced to more significantly enhance
oral absorption. All the above data show that the preparation of nanosuspension using
acid-base neutralization recrystallization combined with a high-speed dispersion method
might be a promising way to enhance the solubility and oral bioavailability of ABZ.

5. Conclusions
In this study, ABZ nanocrystals were prepared using a bottom-up method based
on acid-base neutralization recrystallization with high-speed mixing and dispersing. The
cosolvent, stabilizer and temperature were optimized using a single factor test. This process
is easy to scale up as it can be done in close proximity and could reduce the chances of
contamination. Additionally, as no organic solvent is employed during this process, it could
be an eco-friendly fabrication method. Simple instruments, low energy, less heat generation
and low cost are some characteristic features of this method. This technique could overcome
the problems associated with top-down and bottom-up approaches. The HPLC, DSC and
XRD demonstrated that the molecular structure and crystal form remain unchanged in the
preparation process. The saturation solubility and dissolution were significantly enhanced
by nanocrystallization, and the bioavailability of ABZ was significantly improved. The
nanosuspension prepared by acid-base neutralization recrystallization combined with
high-speed dispersion method might be a promising way to enhance the solubility and
oral bioavailability of ABZ and other insoluble drugs.

Author Contributions: D.C. and S.X. contributed to the study design and execution and gave final
approval of the manuscript. Z.L. was involved in the study execution, data analysis and interpretation,
and manuscript preparation. M.C. and Y.Y. contributed to animals experiments and data analysis.
All authors have read and agreed to the published version of the manuscript.
Funding: This work was supported by the Fundamental Research Funds for the Central Universities
(2662020DKPY008).
Nanomaterials 2022, 12, 3032 13 of 14

Institutional Review Board Statement: The pharmacokinetics experiment was performed according
to “Guidelines of the Care and Use of Laboratory Animals” of Huazhong Agricultural University
and approved by the Ethics Committee of Huazhong Agricultural University (HZAUCH-2020-0004).
Informed Consent Statement: Not applicable.
Data Availability Statement: Not applicable.
Conflicts of Interest: The authors declare no conflict of interest.

References
1. Morris, D.L. Preoperative albendazole therapy for hydatid cyst. Br. J. Surg. 1987, 74, 805–806. [CrossRef]
2. Gil-Grande, L.A.; Sánchez-Ruano, J.; García-Hoz, F.; Bárcena, R.; Rodriguez-Caabeiro, F.; Brasa, C.; Casado, N.; Prieto, J.G.;
Alvarez, A.I.; Aguilar, L.; et al. Randomised controlled trial of efficacy of albendazole in intra-abdominal hydatid disease. Lancet
1993, 342, 1269–1272. [CrossRef]
3. Zulu, I.; Veitch, A.; Sianongo, S.; McPhail, G.; Feakins, R.; Farthing, M.J.G.; Kelly, P. Albendazole chemotherapy for AIDS-related
diarrhoea in Zambia-clinical; parasitological and mucosal responses. Aliment. Pharm. Ther. 2002, 16, 595–601. [CrossRef]
4. Venkatesan, P. Albendazole. J. Antimicrob. Chemother. 1998, 41, 145–147. [CrossRef]
5. Del Brutto, O.H. Current approaches to cysticidal drug therapy for neurocysticercosis. Expert Rev. Anti. Infect. Ther. 2020, 18,
789–798. [CrossRef]
6. Nery, S.V.; McCarthy, J.S.; Traub, R.; Andrews, R.M.; Black, J.; Gray, D.; Weking, E.; Atkinson, J.-A.; Campbell, S.; Francis, N.;
et al. A cluster-randomised controlled trial integrating a community-based water; sanitation and hygiene programme; with mass
distribution of albendazole to reduce intestinal parasites in Timor-Leste, the WASH for WORMS research protocol. BMJ Open
2015, 5, e9293. [CrossRef]
7. Abate, E.; Elias, D.; Getachew, A.; Alemu, S.; Diro, E.; Britton, S.; Aseffa, A.; Stendahl, O.; Schön, T. Effects of albendazole
treatment on the clinical outcome and immunological responses in patients with helminth infection and pulmonary tuberculosis,
a randomized clinical trial. Med. Health Sci. 2013, 61, 91827.
8. Moro, P.; Schantz, P.M. Echinococcosis, a review. Int. J. Infect. Dis. 2009, 13, 125–133. [CrossRef]
9. Wen, H.; New, R.R.C.; Muhmut, M.; Wang, J.H.; Wang, Y.H.; Zhang, J.H.; Shao, Y.M.; Craig, P.S. Pharmacology and efficacy
of liposome-entrapped albendazole in experimental secondary alveolar echinococcosis and effect of co-administration with
cimetidine. Parasitology 1996, 113, 111–121. [CrossRef]
10. Castillo, J.; Palomo-Canales, J.; Garcia, J.; Lastres, J.; Bolas, F.; Torrado, J. Preparation and characterization of albendazole
β-cyclodextrin complexes. Drug Dev. Ind. Pharm. 1999, 25, 1241–1247. [CrossRef]
11. Martinez-Marcos, L.; Lamprou, D.A.; McBurney, R.T.; Halbert, G.W. A novel hot-melt extrusion formulation of albendazole for
increasing dissolution properties. Int. J. Pharm. 2016, 499, 175–185. [CrossRef] [PubMed]
12. McGuckin, M.B.; Wang, J.W.; Ghanma, R.; Qin, N.; Palma, S.D.; Donnelly, R.F.; Paredes, A.J. Nanocrystals as a master key to
deliver hydrophobic drugs via multiple administration routes. J. Control. Release 2022, 345, 334–353. [CrossRef] [PubMed]
13. Rabinow, B.E. Nanosuspensions in drug delivery. Nat. Rev. Drug Discov. 2004, 3, 785–796. [CrossRef] [PubMed]
14. Kesisoglou, F.; Panmai, S.; Wu, Y. Nanosizing-oral formulation development and biopharmaceutical evaluation. Adv. Drug
Deliver. Rev. 2007, 59, 631–644. [CrossRef]
15. Müller, R.; Jacobs, C.; Kayser, O. Nanosuspensions as particulate drug formulations in therapy, rationale for development and
what we can expect for the future. Adv. Drug Deliver. Rev. 2001, 47, 3–19. [CrossRef]
16. Sattar, A.; Chen, D.; Jiang, L.; Pan, Y.; Tao, Y.; Huang, L.; Liu, Z.; Xie, S.; Yuan, Z. Preparation; characterization and pharmacoki-
netics of cyadox nanosuspension. Sci. Rep. 2007, 7, 2289. [CrossRef]
17. Sun, Y.Z.; Chen, D.M.; Zhao, Y.; Zhou, K.X.; Zhang, B.; Wang, H.T.; Xie, S.Y. Exploitation of nanocrystal suspension as an effective
oral formulation for oxfendazole. Drug Deliv. Transl. Res. 2022, 12, 1219–1229. [CrossRef]
18. Mohammad, I.S.; Hu, H.Y.; Yin, L.F.; He, W. Drug nanocrystals, Fabrication methods and promising therapeutic applications. Int.
J. Pharm. 2019, 562, 187–202. [CrossRef]
19. Pensel, P.; Paredes, A.J.; Albani, C.M.; Allemandi, D.; Bruni, S.S.; Palma, S.D.; Elissondo, M.C. Albendazole nanocrystals in
experimental alveolar echinococcosis, Enhanced chemoprophylactic and clinical efficacy in infected mice. Vet. Parasitol. 2018, 251,
78–84. [CrossRef]
20. Paredes, A.J.; Camacho, N.M.; Schofs, L.; Dib, A.; Zarazaga, M.P.; Litterio, N.; Allemandi, D.A.; Bruni, S.S.; Lanusse, C.; Palma,
S.D. Ricobendazole nanocrystals obtained by media milling and spray drying, Pharmacokinetic comparison with the micronized
form of the drug. Int. J. Pharm. 2020, 585, 119501. [CrossRef]
21. Paredes, A.J.; Litterio, N.; Dib, A.; Allemandi, D.A.; Lanusse, C.; Bruni, S.S.; Palma, S.D. A nanocrystal-based formulation
improves the pharmacokinetic performance and therapeutic response of albendazole in dogs. J. Pharm. Pharmacol. 2018, 70, 51–58.
[CrossRef] [PubMed]
22. Paredes, A.J.; Bruni, S.S.; Allemandi, D.; Lanusse, C.; Palma, S.D. Albendazole nanocrystals with improved pharmacokinetic
performance in mice. Ther. Deliv. 2018, 9, 89–97. [CrossRef]
Nanomaterials 2022, 12, 3032 14 of 14

23. Hu, C.H.; Liu, Z.S.; Liu, C.Y.; Zhang, Y.G.; Fan, H.N.; Qian, F. Improvement of Antialveolar Echinococcosis Efficacy of Albendazole
by a Novel Nanocrystalline Formulation with Enhanced Oral Bioavailability. ACS Infect. Dis. 2019, 6, 802–810. [CrossRef]
[PubMed]
24. Koradia, D.K.; Parikh, H.R. Dissolution enhancement of albendazole through nanocrystal formulation. J. Pharm. Bioall. Sci. 2012,
4, 62–63. [CrossRef] [PubMed]
25. Permana, A.D.; Paredes, A.J.; Zanutto, F.V.; Amir, M.N.; Ismail, I.; Bahar, M.A.; Sumarheni, P.S.D.; Donnelly, R.F. Albendazole
Nanocrystal-Based Dissolving Microneedles with Improved Pharmacokinetic Performance for Enhanced Treatment of Cystic
Echinococcosis. ACS App. Mater. Inter. 2021, 13, 38745–38760. [CrossRef]
26. Lopez-Vidal, L.; Real, J.P.; Real, D.A.; Camacho, N.; Kogan, M.J.; Paredes, A.J.; Palma, S.D. Nanocrystal-based 3D-printed tablets,
Semi-solid extrusion using melting solidification printing process (MESO-PP) for oral administration of poorly soluble drugs. Int.
J. Pharm. 2022, 611, 121311. [CrossRef]
27. Paredes, A.J.; Llabot, J.M.; Bruni, S.S.; Allemandi, D.; Palma, S.D. Self-dispersible nanocrystals of albendazole produced by high
pressure homogenization and spray-drying. Drug Dev. Ind. Pharm. 2016, 42, 1564–1570. [CrossRef]
28. Craing, P.S. Current research in eahinococcosis. Parasitol. Today 1994, 10, 209–210. [CrossRef]
29. Morris, D.L.; Clarkson, M.J.; Stallbaumer, M.F.; Pritchard, J.; Jones, R.S.; Chinnery, J.B. Albendazole treatment os pulmonary
hydatid cysts in naturally infected sheep: A study with relevance to the treatment of hydatid cysts in man. Thorax 1985, 40,
453–458. [CrossRef]
30. Rowe, R.C.; Sheskey, P.J.; Weller, P.J.; Rowe, R.; Sheskey, P.; Weller, P. Handbook of Pharmaceutical Excipients, 4th ed.; Pharmaceutical
Press and American Pharmaceutical Association: London, UK, 2003; pp. 271–273.
31. Shegokar, R.; Müller, R.H. Nanocrystals, industrially feasible multifunctional formulation technology for poorly soluble actives.
Int. J. Pharmaceut. 2010, 399, 129–139. [CrossRef]
32. Lee, J.; Lee, S.J.; Choi, J.Y.; Yoo, J.Y.; Ahn, C.-H. Amphiphilic amino acid copolymers as stabilizers for the preparation of
nanocrystal dispersion. Eur. J. Pharm. Sci. 2005, 24, 441–449. [CrossRef] [PubMed]
33. Rainer, H.M.; Katrin, P. Nanosuspensions for the formulation of poorly soluble drugs, I. Preparation by a size-reduction technique.
Int. J. Pharmaceut. 1998, 160, 229–237.
34. Ji, Y.B.; Gao, Y.X.; Hao, X.; Zhang, X.; Wang, X. Preparation of annonaceous acetogenins nanosuspensions using poloxamer 188 as
a stabilizer and their in vitro and in vivo investigation. Acta Pharm. Sin. B 2018, 53, 2113–2121.
35. Zhang, J.; Bunker, M.; Parker, A.; Madden-Smith, C.E.; Patel, N.; Roberts, C.J. The stability of solid dispersions of felodipine in
polyvinylpyrrolidone characterized by nanothermal analysis. Int. J. Pharmaceut. 2011, 414, 210–217. [CrossRef]
36. Hilbig, J.; Ma, Q.; Davidson, P.M.; Weiss, J.; Zhong, Q. Physical and antimicrobial properties of cinnamon bark oil co-
nanoemulsified by lauric arginate and Tween 80. Int. J. Food Microbiol. 2016, 233, 52–59. [CrossRef]
37. Kuskov, A.N.; Voskresenskaya, A.A.; Goryachay, A.V. Preparation and characterization of amphiphilic poly-N-vinylpyrrolidone
nanoparticles containing indomethacin. J. Mater. Sci. Mater. Med. 2010, 21, 1521–1530. [CrossRef]
38. Lopes, M.A.; Abrahim-Vieira, B.; Oliveira, C. Probing insulin bioactivity in oral nanoparticles produced by ultrasonication-assisted
emulsification/internal gelation. Int. J. Nanomed. 2015, 10, 5865–5880.
39. Santos, A.C.; Cunha, J.; Veiga, F. Ultrasonication of insulin-loaded microgel particles produced by internal gelation, Impact on
particle’s size and insulin bioactivity. Carbohyd. Polym. 2013, 98, 1397–1408. [CrossRef]
40. Kim, H.Y.; Han, J.A.; Kweon, D.K. Effect of ultrasonic treatments on nanoparticle preparation of acid-hydrolyzed waxy maize
starch. Carbohyd. Polym. 2012, 93, 582–588. [CrossRef]
41. Adhiyaman, R.; Basu, S.K. Modification of dipyridamole using different solvents and crystallisation conditions. Int. J. Pharmaceut.
2006, 321, 27–34. [CrossRef]
42. Böhm, B.H.; Müller, R.H. Lab-scale production unit design for nanosuspensions of sparingly soluble cytotoxic drugs. Pharm. Sci.
Technol. Today 1999, 2, 336–339. [CrossRef]
43. Lubega, G.W.; Prichard, R.K. Interaction of benzimidazole anthelmintics with Haemonchus contortus tubulin: Binding affinity
and anthelmintic efficacy. Exp. Parasitol. 1999, 73, 203–213. [CrossRef]
44. Zhang, H.; Zhao, J.; Chen, B.; Ma, Y.; Li, Z.; Shou, X.; Wen, L.; Yuan, Y.; Gao, H.; Ruan, J.; et al. Pharmacokinetics and tissue
distribution study of liposomal albendazole in naturally Echinococcus granulosus infected sheep by a validated UPLC-Q-TOF-MS
method. J. Chromatog. B 2020, 1141, 122016. [CrossRef] [PubMed]
45. Fabbri, J.; Espinosa, J.P.; Pensel, P.E.; Medici, S.K.; Gamboa, G.U.; Benoit, J.P.; Elissondo, M.C. Do albendazole-loaded lipid
nanocapsules enhance the bioavailability of albendazole in the brain of healthy mice? Acta Trop. 2020, 201, 105215. [CrossRef]
46. Ban, C.; Jo, M.; Park, Y.H.; Kim, J.H.; Han, J.Y.; Lee, K.W.; Kweon, D.-H.; Choi, Y.J. Enhancing the oral bioavailability of curcumin
using solid lipid nanoparticles. Food Chem. 2020, 302, 125328. [CrossRef]

You might also like