Vol 4, Issue 4, 2023 (307-332)
http://journal.unpad.ac.id/idjp
*Corresponding author,
e-mail : rizky17009@mail.unpad.ac.id (R. F. Pratama)
https://doi.org/10.24198/idjp.v4i3.40849
© 2023 R. F. Pratama et al
Review: Preparation of Flavonoid Nanoparticles using the Nanoprecipitation Method
Rizky Farhan Pratama*,1, Iyan Sopyan2, Taofik Rusdiana2
1Bachelor Program in Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran
2Department of Pharmaceutical and Formulation Technology, Faculty of Pharmacy,
Universitas Padjadjaran Jalan Raya Bandung-Sumedang KM 21, Jatinangor
45363, Indonesia
Submitted : 22/07/ 2022, Revised : 01/08/ 2022, Accepted : 06/06/2023, Published : 16/08/2023
Abstract
Flavonoids are polyphenolic compounds that have 15 carbon chains, 2 benzene
rings and a heterocyclic pyran ring. From the literature study, it is known that
flavonoids have various pharmacological activities such as anticancer,
antimicrobial, antiviral, antiangiogenic, antimalarial, antioxidant, neuroprotective,
antitumor, and antiproliferative agents. However, flavonoids have limited oral
bioavailability which may be due to their poor solubility, low permeability, and low
stability, which impair their effectiveness as therapeutic agents. One of the efforts
to increase solubility is nanoparticle technology where the active compound
particles are reduced to the nanometer scale, usually up to 100 nm.
Nanoprecipitation is a method of preparing nanoparticles by dissolving the active
drug substance and polymer into an organic solvent and then adding an anti-solvent
such as water. The advantages of this method are the production is relatively fast,
inexpensive, does not require a lot of energy, and does not require emulsion
precursors. The purpose of this literature review is to examine the technique of
making flavonoid nanoparticles using the nanoprecipitation method, the results of
their characterization and evaluation. Based on a literature review that has been
carried out on 30 journals, there are 20 flavonoid secondary metabolites that have
been prepared into nanoparticles using the nanoprecipitation method. Some of the
polymers used were effective in achieving satisfactory particle size, polydispersity
index (PDI), Zeta potential and Encapsulation Efficiency (EE%). Thus, the
nanoprecipitation method can be used to make flavonoid nanoparticles with optimal
formulations to improve the physicochemical properties of flavonoids for drug
development in the future.
Keywords: Flavonoid, Nanoparticles, Nanoprecipitation Method, Characterizatio
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
308
1. Introduction
Flavonoids are polyphenolic
compounds that have 15 carbon chains, 2
benzene rings and a heterocyclic pyran ring.
Based on existing literature studies,
flavonoids have various pharmacological
activities such as anticancer, antimicrobial,
antiviral, antiangiogenic, antimalarial,
antioxidant, neuroprotective, antitumor, and
antiproliferative agents. One example of a
compound that has pharmacological activity
is quercetin which has anticancer activity.
However, some flavonoid compounds,
especially flavonoids which are included in
the category of flavonoid aglycones have low
solubility, low absorption and short drug
residence time [1,2].
In order to overcome these problems,
physical and chemical modifications of drugs
and various methods were carried out. The
methods used were about reducing particle
size, formation of co-crystal, formation of
salt, solid dispersion, surfactants appliaction,
complexation methods, approaches with
nanotechnology and so on [3,4]. Several
studies have approached nanotechnology to
make stable formulas. Nano technology is
used to reduce the particle size of a particle
so that it will form nanoparticles.
Nanoparticles are substances that have a
diameter of less than 1000 nm [5]. Utilization
of nano technology or the manufacture of
nanoparticles can be used for flavonoid
modification. One example of the research
conducted by Telange et al. which proves that
the solubility of Apigenin can be increased by
the solubility value of Apigenin-phopolipid
Phytosome in water by 22.80 g/mL. The
formation of nanoparticles also serves to
increase anticancer activity, for example, the
formation of quercetin nanoparticles with
PLGA polymer which prevents the formation
of hepatocellular carcinoma through the
protection of the mitochondrial membrane in
the liver [6]. The simplest method of forming
nanoparticles is Nanoprecipitation.
Nanoprecipitation is a method of
preparing nanoparticles for hydrophobic
active substances developed by Fessi et al in
1989. The principle behind this method is
using the Marangoni effect. Overall this
method is carried out by making the active
drug substance and polymer dissolved in an
organic solvent which is then added to an
anti-solvent such as water. The advantages of
this method are that the production is
relatively fast, low cost, does not require a lot
of energy, and does not require emulsion
precursors. In addition, nanoprecipitation can
also produce nanoparticles with particle sizes
in the range of 50-300 nm [5,7,8,9].
Based on the literature search, the
preparation of nanoparticles with the
nanoprecipitation method has been widely
carried out on natural materials including
flavonoids. Therefore, a literature review was
carried out by collecting data on the
manufacture of flavonoid nanoparticles with
the nanoprecipitation method as a reference
for future research on the manufacture of
flavonoid nanoparticles.
2. Methods
This review was carried out by
conducting a literature search related to the
manufacture of flavonoid nanoparticles using
the nanoprecipitation method and their
characterization on the ScienceDirect,
Pubmed and Springer Link sites with specific
keywords used to search for qualified articles
such as "Flavonoid", "Nanoparticle",
"Nanoprecipitation". Method”, and
“Characterization”. The articles used are
articles from the last 10 years in English.
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
309
Figure 1. Methods performed for review
3. Results and Discussion
Table 1. Variety of the Nanoprecipitation method used in the preparation of Flavonoid
Flavonoid
Nanoparticle
Technologies
Nanoparticl
e Types
Carrier
Solubility/Bioavailability
/Dissolution
Reference
s
Apigenin
Apigenin-
Polimeric
PLGA
-
[27]
Search articles using keywords: “Flavonoid”, “Nanoparticle”, “Nanoprecipitation
Method”, and “Characterization” on ScienceDirect, Pubmed and Springer Link.
Initial search results (n = 301)
Reprocessed article search results
(n = 118)
Unprocessed article from search
results (n = 183)
Articles used in research (n = 30)
Exclusion critetia:
T
he article does not discuss the
manufacture of flavonoid
nanoparticles using the
nanoprecipitation method and
their characterization
Articles are not research
articles (such as reviews and
systematic reviews.)
Articles are not in English
language
Inclusion critetia:
T
he article discuss the
manufacture of flavonoid
nanoparticles using the
nanoprecipitation method and
their characterization
Articles are research articles
Articles published from the last
10 years
Articles are in English
language
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
310
loaded PLGA
nanoparticles
Nanoparticl
es
Apigenin-
loaded
galactose
tailored
PLGA
nanoparticles
Polimeric
Nanoparticl
es
Galaktosa dan
PLGA
-
[27]
Artocarpin
Artocarpin
Nanoparticle
system
(Artocarpin:
PVP)
Polimeric
Nanoparticl
es
PVP
Solubility: 1400-fold
[28]
Chrysin
Chrysin
nanocapsules
Lipid-
polymer
hybrid
Nanoparticl
es
PLGA,
Labrafac PG,
phosphatidylch
oline
-
[29]
Cirsiliol
Cirsiliol-
loaded
nanocapsules
Lipid-
polymer
hybrid
Nanoparticl
es
PEG-PCL, Span
80, Tween 80
Solubility: 24-fold
[30]
Curcumin
Curcumin
encapsulated
Chitosan
functionalized
PLGA Core
Shell
Nanoparticles
Organic-
Inorganic
hybrid
Nanoparticl
es
PLGA, Kitosan
-
[31]
PLGA-CTAB
curcumin
nanoparticles
Polimeric
Nanoparticl
es
PLGA, CTAB
-
[32]
Dihydromyricet
in
Nanocapsule
suspensions
containing
Dihydromyric
etin
Polimeric
Nanoparticl
es
Eudragit RS100
-
[33]
Diosmin
Polymer-
stabilized
diosmin
nanosuspensi
on
Polimeric
Nanoparticl
es
HPMC/MC
Dissolution: 2-fold
[34]
Eupafolin
Eupafolin
nanoparticles
Polimeric
Nanoparticl
es
PVA, Eudragit
E100
-
[35]
Fisetin
Fisetin-
loaded
nanoparticles
Polimeric
Nanoparticl
es
PCL, PLGA-
PEG-COOH,
Pluronic F127.
-
[12]
Genistein
Genistein-
loaded M-
PLGA-TPGS
Polimeric
Nanoparticl
es
M-PLGA-
TPGS
-
[26]
Luteolin
Hybrid
Polimeric
PLA, Eudragit
Bioavaialbility: 2.61-fold
[36]
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
311
PLA/Eudragit
100 Luteolin
nanoparticles
Nanoparticl
es
L100, Pluronic
F127
Naringenin
PVP-coated
naringenin
nanoparticles
Polimeric
Nanoparticl
es
PVP
-
[37]
Naringenin-
loaded
nanoparticles
Polimeric
Nanoparticl
es
Eudragit, PVA
-
[25]
Phloretin
Hydrogel
containing
polymeric
nanocapsules
loaded
with phloretin
Polimeric
Nanoparticl
es
PCL, Span 60,
Tween 80,
-
[38]
Poly-puerarin
Poly-puerarin
nanoparticles
with
Paclitaxel
Polimeric
Nanoparticl
es
Pluronic F127
-
[39]
Proantosianidin
PLGA
nanoparticles
loaded with
Proamthocian
idin-rich
Grapeseed
Extract,
Polimeric
Nanoparticl
es
PLGA
-
[40]
Quercetin
P. guajava
ethyl acetate
fraction
loaded
nanosuspensi
on
Polimeric
nanoparticle
s
PVA
Bioavailability: 6,28-fold
[15]
Starch
nanoparticles
loaded with
Quercetin
Starch
nanoparticle
s
Pati Kentang
-
[24]
MPEG-PLA
encapsulated
Quercetin
nanoparticle
Polimeric
nanoparticle
s
MPEG-PLA,
Pluronic F-68
Bioavaialability: 1.87-
fold
[16]
Quercetin
embedded
PLA
nanoparticles
Polimeric
nanoparticle
s
PLA, PVA
-
[41]
Zinc
phthalocyanin
e-Quercetin
loaded lipid-
polymer
hybrid
nanoparticle
Lipid-
polymer
hybrid
Nanoparticl
es
PLGA, soybean
lecithin, DSPE-
PEG2000
Bioavailability: 3.64-fold
[42]
Quercetin-
loaded
Eudragit®
Polimeric
nanoparticle
s
Eudragit S100
-
[43]
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
312
S100
Nanoparticles
Quercetin-
loaded PCL
based
nanoparticles
Polimeric
nanoparticle
s
PCL
-
[44]
Quercetin
conjugated
Fe3O4
nanoparticles
Magnetic
nanoparticle
s
Dextran, Fe3O4
-
[11]
Rutin
Rutin
nanospheres
Polimeric
Nanoparticl
es
Eudragit S100,
Poloxamer-188.
-
[45]
Salvigenin
Salvigenin-
loaded
mPEG-b-
PLGA with
Fe3O4
Magnetic
nanoparticle
s
mPEG-b-
PLGA, Fe3O4
-
[23]
Silibinin
Silibinin-
loaded
nanoparticles
Polimeric
Nanoparticl
es
PVA, Eudragit
E100
-
[46]
Silymarin
Silymarin-
loaded lipid
polymer
hybrid
nanoparticles
containing
chitosan
Lipid-
polymer
hybrid
Nanoparticl
es
PLGA, soybean
lecithin, DSPE-
PEG2000,
Kitosan
-
[47]
Silymarin-
Loaded
Eudragit
Polimeric
Nanoparticl
es
PVA, Eudragit
RS100 &
RL100
-
[14]
Table 2. Characterization of Flavonoid Nanoparticles Using Nanoprecipitation Method
Flavonoid
Nanoparticle
Technologies
Partikel
Size
(nm)
PDI
Zeta
Potential
(Mv)
EE (%)
Activity
References
Apigenin
Apigenin-loaded PLGA
nanoparticles
110,0
0,041 +
0,004
-25,0
70,3
Hepatocellular
Carcinoma Treatment
[27]
Apigenin-loaded
galactose tailored
PLGA nanoparticles
129,0
0,059 ±
0,007
-14,0
75,4
Hepatocellular
Carcinoma Treatment
[27]
Artocarpin
Artocarpin
Nanoparticle system
(Artocarpin: PVP)
128,4 ±
0,7
0,266 ±
0.024
-
>99
Hepatocellular
Carcinoma Treatment
[28]
Chrysin
Chrysin nanocapsules
176 ±
2,10
0,22 ±
0,01
-6,23 ±
0,18
87,10 ±
6,71
Antiglycemic,
Antihyperlipidemic
[29]
Cirsiliol
Cirsiliol-loaded
nanocapsules
158.1 ±
12,4
0,19 ±
0,01
−2,6 ± 5.1
53,5 ±
2,1
Anticancer
[30]
Curcumin
Curcumin encapsulated
Chitosan functionalized
207,6 ±
2,71
0,165 ±
0,075
+31,9 ±
1,03
75.53 ±
2,09
Alzheimer’s Disease
Treatment
[31]
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
313
PLGA Core Shell
Nanoparticles
PLGA-CTAB curcumin
nanoparticles
81,05 ±
3.85
0,107
+31,8
69,1
Breast Cancer
Treatment
[32]
Dihydromyricetin
Nanocapsule
suspensions containing
Dihydromyricetin
160 ± 5,0
0,120 ±
0,05
+8,5 ± 1,5
-
Photoprotection
[33]
Diosmin
Polymer-stabilized
diosmin
nanosuspension
316 ±
5,55
0,41 ±
0,04
-
-
-
[34]
Eupafolin
Eupafolin nanoparticles
90,8
-
-
-
Acute Renal Injury
Treatment
[35]
Fisetin
Fisetin-loaded
nanoparticles
198,7 ±
6,0
0,158 ±
0,02
-
74,78 ±
1,9
Antioxidant
[12]
Genistein
Genistein-loaded M-
PLGA-TPGS
225,7 ±
2,5
0,169
-14,2 ±
0,7
97,66
Liver Cancer
Treatment
[26]
Luteolin
Hybrid PLA/Eudragit
100 Luteolin
nanoparticles
452,23 ±
22,4
-
0,92 mV ±
0,04
71,02 ±
14,6%
-
[36]
Naringenin
PVP-coated naringenin
nanoparticles
110
-
-
99,93
Antioxidant
[37]
Naringenin-loaded
nanoparticles
90
-
-
-
Anticancer
[25]
Phloretin
Hydrogel containing
polymeric nanocapsules
loaded
with phloretin
252 ±
12,01
1,68 ±
0,11
>-1
>99
Antitumoral
[38]
Poly-puerarin
Poly-puerarin
nanoparticles with
Paclitaxel
70,26
0,15
−23,1
91,3
Anticancer
[39]
Proantosianidin
PLGA nanoparticles
loaded with
Proamthocianidin-rich
Grapeseed Extract,
132,5 ±
12,2
-
26,7 ±
3,8
65,2 ±
6,1
Dentin Degradation
Resistance
[40]
Quercetin
P. guajava ethyl acetate
fraction loaded
nanosuspension
241,32 ±
1,25
0,224 ±
0,011
-22 ± 0,4
92,85 ±
2,23
Antihyperglycemic
[15]
Starch nanoparticles
loaded with Quercetin
91,2
154,5
0,276
0,41
-
-
Antioxidant
[24]
MPEG-PLA
encapsulated Quercetin
nanoparticle
155,3 ±
3,2
0,2 ±
0,05
−3,14
-
Breast Cancer
Treatment
[16]
Quercetin embedded
PLA
nanoparticles
46 ± 4
-
-
62±3
Anticancer
[41]
Zinc phthalocyanine-
Quercetin loaded lipid-
polymer hybrid
nanoparticle
174,8
0,331
−30 ± 10
10 ± 4
Photodynamic
Anticancer
[42]
Quercetin-loaded
Eudragit® S100
Nanoparticles
66,8 ±
2,3
-
-5,2 ± 2,4
41,8 ±
9,1
Colon Cancer
Treatment
[43]
Quercetin-loaded PCL
based nanoparticles
215,9 ±
2,9
0,094
-12,9 ±
0,35
66,32 ±
0,4
-
[44]
Quercetin conjugated
Fe3O4 nanoparticles
72
-
+6,14
81,6
Breast Cancer
Treatment
[11]
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
314
Rutin
Rutin nanospheres
130,30 ±
35,29
0,29 ±
0,10
-22,90 ±
5,18
98,10 ±
0,50
Colon
Carcinoma Treatment
[45]
Salvigenin
Salvigenin-loaded
mPEG-b-PLGA with
Fe3O4
57 ± 2
0,168 ±
0,03
−33 ± 1,2
82 ± 1,6
Anticancer
[23]
Silibinin
Silibinin-loaded
nanoparticles
120
-
+4,6
79,0 ±
2,4
Oral Carcinoma
Treament
[46]
Silymarin
Silymarin-loaded
lipid polymer hybrid
nanoparticles
containing chitosan
286,5 ±
23,8
0,226 ±
0,008
45,3 ± 8,9
97,05 ±
0,01
Hepatoprotective
Agent
[47]
Silymarin-Loaded
Eudragit
84,70
0,38 ±
0,01
83,45
Hepatoprotective
Agent
[14]
The formation of flavonoid
nanoparticles increases the oral
bioavailability of insoluble active flavonoid
components via particle reduction which
resulting in increase of surface are.
Nanoparticles formation also led to the
increase of saturation solubility and
dissolution rate. [10,11,12,13].
Particle size and polydispersity
index (PDI) influence the physicochemical
properties of nanoparticles such as
dissolution speed, saturation solubility, and
physical stability. Polydispertity index also
describes the uniformity of particle shape.
The effective nanosuspension size for
absorption in the intestine ranged from 100 to
500 nm. The nanosuspension size is in the
range of 100 - 200 nm. The ideal particle size
for in-vivo treatment is less than 200 nm.
Nanoparticles are said to be homogeneous if
their polydispersity index is less than 0.4.
PDI values greater than 0.4 indicate particle
aggregation [14,15,16,17,18].
Zeta potential is related to the
electrostatic potential that develops at an
indistinct boundary between the
nanoparticles and the solution. Nanoparticles
have is stable and have positive or negative
charge if the zeta potential value is greater
than +30 mV or less than -30 mV for positive
charge and negative charge respectively
[15,19,20].
Encapsulation Efficiency (EE%) is
the percentage of drug trapped into the
nanocarrier matrix which refers to the total
drug input. A good Efficiency of
Encapsulation has a value of >80%
[21,22,23].
Preparation of nanoparticles with the
Nanoprecipitation method is carried out by
adding a dilute polymer solution to a non-
solvent or vice versa which is then followed
by polymer deposition at the nanoscale
[14,24,25,26].
The traditional Nanoprecipitation
method is prepared in large volumes of
solution by adding an anti-solvent to a
solvent containing hydrophobic molecules or
vice versa drop by drop under mixing. In
small-volume mixing processes, this process
produces particles instantaneously with a
narrow size distribution often at the
nanoscale. Anti-solvents must be miscible
with solvents containing polymers or drugs.
Solutions with large volumes are difficult to
control the results with the stirring process
[48]. The Heat induced evaporative
antisolvent nanoprecipitation (HIEAN)
method begins by dissolving the active
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
315
substance in an organic solvent at a low
boiling point. Then, the resulting solution is
added to the heated aqueous solution. The
increased temperature of the heated aqueous
solution ensures rapid evaporation of organic
solvents and results in a high degree of
saturation and rapid precipitation of the drug
in the form of suspended particles. Stabilizers
added to a heated aqueous solution adsorb
onto the surface of newly formed particles
and reduce the surface energy and prevent
particle growth [49, 50].
The nanoprecipitation method
approach with the help of sonication is
applied to reduce the particle size of the
active substance which is not soluble in
water. Ultrasonication into the carrier
containing solution is carried out to coat and
stabilize the nanoparticles. The propagation
of ultrasound into a liquid medium results in
alternating cycles of compression and
contraction resulting in vacuum bubbles
which accumulate energy and then release it
violently or it is also called cavitation.
Cavitation can trigger and accelerate various
chemical reactions including the formation of
nanoparticles [51].
The nanoparticles that are often
made by this method are polymeric
nanoparticles. Polymeric nanoparticles have
more stability in the gastrointestinal tract than
other colloidal carriers. Polymeric
nanoparticles have other advantages such as
maintaining effect on target tissues, solubility
for intravascular delivery and protection from
enzymatic degradation, especially in gastric
acid. The polymers that are often used are
polymers from the polyester family such as
PLGA, PLA and PCL [14,24,25,26].
Based on the results of the literature
research that has been carried out, 301
articles were obtained which were then
processed into 30 articles. The article
research process was carried out using
indexed databases such as ScienceDirect (n =
24), PubMed (n = 4) and SpringerLink (n =
2). The results of literature research can be
found in Table 1.
3.1 Apigenin
Apigenin was prepared into two
different nanoparticles, the first nanoparticle
using PLGA as a carrier while the second
nanoparticle using Galactose-PLGA as a
carrier. Both nanoparticles use the same
method. The manufacture of nanoparticles
starts from dissolving the polymer (PLGA or
Galactose-PLGA) and Apigenin in acetone.
The mixtures were poured into ultrapure
water containing poloxamer 188 [27].
Nanoparticles with PLGA produced
have particle size of 110 nm, polydispersity
index 0.041 ± 0.004, Zeta Potential -25.0 mV
and Efficiency of 70.3%. Nanoparticles with
Galactose-PLGA produced a particle size of
129.0 nm, polydispersity index 0.059 ±
0.007, Zeta Potential -14.0 mV and
encapsulation efficiency of 75.4%. The low
polydispersity index value indicates that the
nanoparticles show the same size between
each other. The zeta potential value of
nanoparticles made with Galactose-PLGA is
smaller than using only PLGA. This is due to
the PLGA galactosylation. The negative zeta
potential also aids the uptake of the
reticuloendothelial system (RES) [27].
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
316
3.2 Artocarpin
Artocarpine which has an autophagic
effect was made into naooparticles with an
artocarpine: PVP nanoparticle system with a
ratio of 1:10. The steps start from dissolving
artocarpine in ethanol to form organic phase.
The aqueous phase is prepared by adding
PVP in ultrapure water. The organic phase
was then injected into the aqueous phase. The
resulting nanoparticles had a particle size of
128.4 ± 0.7 nm, a polydispersity index of
0.266 ± 0.024, an encapsulation efficiency
above 99% and a water solubility of 1400
times that of free Artocarpin. The particle
size below 200 nm makes administration of
the drug in the body more effective. The PDI
value indicates the absence of agglomerates.
Formation of intermolecular hydrogen bonds
between the OH bonds of artocarpin and the
CO bonds of PVP increase solubility [28].
3.3 Chrysin
Chrysin nanocapsules were made
using Labrafac PG as oil, PLGA as polymer
and Phosphatidylcholine (PC) and Tween 80
as surfactants. The resulting nanocapsules
had a particle size of 176 ± 2.10 nm, a
polydispersity index of 0.22 ± 0.01, a Zeta
Potential of -6.23 ± 0.18 mV, and an
encapsulation efficiency of 87.10 ± 6.71%.
The obtained small particle size nanocapsules
provide a high mucosal adhesion ability on
the gastrointestinal surface, which ensures a
longer retention time. The PDI values of the
nanocapsules showed a good size
distribution. The increase of polymers and
surfactants caused an increase in the particle
size of the nanocapsules. This is due to an
increase in the viscosity of the polymer
solution, making it difficult for the
emulsification process to become smaller
droplets. Increasing the concentration of
tween 80 decrease particle size at a constant
amount of polymer and drug. This is due to a
reduction in the interfacial tension between
dispersed organic phase and dispersion
medium. The zeta potential value obtained is
too low but tween 80 which is reported to
stabilize nanoparticles with a steric effect will
provide protect nanocapsules stability even
though their zeta potential value is low [29].
3.4 Cirsiliol
Picture 1. Production of Cirsiliol nanocapsules [30]
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
317
Nanocapsules containing Cirsiliol
were prepared using the modified
nanoprecipitation method by adding oil and
lipophilic surfactants to the organic phase and
adding hydrophilic surfactants to the aqueous
phase. The method produces colloidal balls
with an oil-filled core. The organic phase
consisted of castor oil, cirsiliol, PEG-PCL
and Span 80 which were dissolved in a
mixture of acetone and ethanol while the
aqueous phase consisted of Tween 80
dissolved in water. PEG is used as a carrier
because PEG has a slow degradation due to
its high crystallinity level, thereby prolonging
drug release and maintaining drug stability.
To improve PEG performance, PCL and PEG
chains were combined by polymerizing CL
ring opening using mPEG as a macroinitiator
and stannous octoate as a catalyst. The
resulting nanocapsules have a particle size
characteristic of 158.1 ± 12.4 nm, a
polydispersity index of 0.19 ± 0.01, a Zeta
Potential of 2.6 ± 5.1 mV, an encapsulation
efficiency of 53.5 ± 2.1% and an increase in
solubility of 24 times that of ordinary
Cirsiliol [30].
3.5 Curcumin
1. Curcumin encapsulated Chitosan
functionalized PLGA Core Shell
Nanoparticles
Core/Shell nanoparticles are
suitable for drugs that target the brain. In
addition, this type of nanoparticles can
also protect the drug from P-gp efflux and
external degradation [31].
In this study, used the organic
phase consisting of curcumin and PLGA
dissolved in acetone and the aqueous
phase consisting of pluronic F127
dissolved in water. PLGA used as core
material because it is biocompatible,
biodegradable and non-toxic. This
polymer can also be used in conjunction
with other polymeric materials. After the
nanoparticle solution is formed, chitosan
is added to the solution so that
electrostatic interactions occur which will
produce Curcumin encapsulated Chitosan
functionalized PLGA Core Shell
Nanoparticles. Chitosan is able to
increase the residence time of intranasal
drugs. This is because the positive charge
interacts electrostatically with the mucin.
Chitosan also has the ability to form a gel
by absorbing water [31].
The characterization results
showed that the nanoparticles made had a
particle size of 207.6 ± 2.71 nm, a
polydispersity index of 0.165 ± 0.075, a
Zeta Potential of +31.9 ± 1.03 mV, and an
encapsulation efficiency of 75.53 ±
2.09%. Particle size and polydispersity
index increased with increasing
concentration of curcumin in the
formulation. Particle size also increased
by coating chitosan against PLGA [31].
2. PLGA-CTAB curcumin
nanoparticles
Curcumin will be used as an
anticancer by making nanoparticles by
applying PLGA as carrier and
hexadecyltrimethylammonium bromide
(CTAB) as a surfactant. PLGA and
curcumin were added to the mixture of
Acetone: Ethanol (17:3) to make an
organic phase while CTAB was dissolved
in water to make an aqueous phase.
PLGA is a polymer with negatively
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
318
charged molecules which is often used as
a carrier in the manufacture of
nanoparticles. However, positively
charged nanoparticles move more easily
from the endosome to the cytosol so that
their bioavailability is better than neutral
or negatively charged nanoparticles. To
produce positively charged nanoparticles,
CTAB which works as a surfactant as
well as a molecular charge modifier is
added during the nanoparticle
manufacturing process [32].
The characterization results
showed that the nanoparticles made had a
particle size of 81.05 ± 3.85 nm, a
polydispersity index of 0.107, a Zeta
Potential of +31.8 mV, and an
encapsulation efficiency of 69.1%. The
small particle size may be due to the
addition of CTAB. The results of the
Polydispersity Index stated that no
aggregates were formed. The zeta
potential obtained indicates that the
nanoparticles are stable enough so that
there is no aggregate formation. The
results of encapsulation efficiency are
influenced by the presence of PLGA
which has a low molecular weight [32].
3.6 Dihydromyricetin
Dihydromyristetin nanocapsules are
made by preparing a solution of Eudragit first
and then adding Dihydromyristetin. To make
hydrogel, 1.5% Hydroxyethylcellulose,
methylparaben and propylene glycol were
added. Hydroxyethylcellulose works as a
thickening agent. The resulting nanocapsules
had a particle size of 160 ± 5.0 nm, a
polydispersity index of 0.120 ± 0.05 and a
Zeta Potential of 8.5 ± 1.5 mV. The
nanoparticles made have a narrow particle
size distribution but their stability is still
questionable because the zeta potential value
is close to zero. Zeta Potential with a positive
charge is needed in order to provide
electrostatic attraction with a negative zeta
potential on human skin, causing an
occlusive effect on the skin [33].
3.7 Diosmin
Diosmin made nanoparticles with two
nanoprecipitation methods, acid-base
neutralization method and anti-solvent
precipitation method. The polymers used as
stabilizers were
Hydroxypropylmethylcellulose (HPMC) and
methylcellulose (MC) [34].
The step of the acid-base
neutralization method in a nutshell starts with
dissolving Diosmin in 1 N NaOH. Diosmin
base solution was dropped into 0.1 N HCl
containing polymer stabilizer, HPMC or MC.
For certain formulations, it is necessary to
remove excess stabilizer and then remove it
by ultracentrifugation. The supernatant was
discarded and then re-dispersed in water [34].
For the antisolvent precipitation method step,
Diosmin was dissolved in DMSO then added
to an aqueous solution containing 0.15%
polymer, HPMC or MC. Stirring was
maintained until a homogeneous milk
suspension was obtained [34].
The nanoparticles with the best
characterization results had an MC
concentration of 14% with the addition of
53% mannitol accompanied by spray-drying
and using an acid-base neutralization method.
The best nanoparticles had a particle size of
316 ± 5.55 nm and a polydispersity index of
0.41 ± 0.04. The anisolven method produces
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
319
a higher PDI than the acid-base neutralization
method. The higher concentration of
stabilizer was reported not to significantly
affect the particle size, but the nanoparticle
formulation of the precipitation took longer
[34].
3.8 Eupafolin
In order to overcome the poor
solubility of Eupafolin, a research was
conducted on the manufacture of Eupafolin
nanoparticles. The reason why Eupafolin is
made into nanoparticles is because in
previous studies Eupafolin nanoparticles
have the potential to create new strategies in
dealing with acute kidney injury. In this
study, Eupafolin nanoparticles were prepared
using the nanoprecipitation method with
solvent evaporation. The organic phase
consisted of Eupafolin, Eudragit and 95%
alcohol while the aqueous phase consisted of
PVA and water. Eudragit E100 and PVA
were chosen as polymers because of their
non-toxic, water-soluble and often used for
oral treatment. The resulting nanoparticles
have a particle size of 90.8. Nanoparticles of
this size are effectively used for in-vivo
treatment [35].
3.9 Fisetin
In this study, fisetin nanoparticles
were made from an organic phase containing
Poly--caprolactone) (PCL), PLGA-PEG-
COOH block copolymer and fisetin in
acetonitrile and the aqueous phase only
consisted of water. Before fabricating
nanoparticles, Sechi et al. do the PLGA-PEG-
COOH conjugation first. PLGA polymer was
chosen to be a polymer because PLGA can
protect the active substance from
degradation, reduce side effects and form
sustained drug release types. To improve the
performance of PLGA, modification of the
PLGA surface was carried out with
poly(ethylene glycol) (PEG). PEG can
prevent the opsonins binding, prolongs
circulation time in blood, facilitate tissues
targeting and reduces PLGA’s absorption by
Rapid Reticuloendothelial System (RES).
PCL is suitable to be used as a PLGA partner,
this is because PLGA and PCL will form
hydrophobic core. In addition, PCL is
permeable and non-toxic [12].
The characterization results showed a
particle size of 198.7 ± 6.0 nm, a
polydispersity index of 0.158 ± 0.02, and an
encapsulation efficiency of 74.78 ± 1.9%.
The particle size of the PLGA-PEG-COOH
nanoparticles is larger than that of ordinary
PLGA, this is because of hydrophilic PEG
chains present in external aqueous phase. The
nanoparticles showed a narrow and unimodal
particle distribution based on the obtained
PDI values. PCL results in better
encapsulation due to its high affinity for
Fisetin. The affinity decreases with
increasing concentration of PLGA-PEG-
COOH which is more hydrophilic [12].
3.10 Genistein
Genistein nanoparticles are made
using PLGA, this is because PLGA is part of
the polyester family which has good
biocompatibility and biodegradability.
However, PLGA and other polyester families
has disadvantage. Liver and RES can easily
eliminate and absorbed PLGA. This can be
solved by the addition of d-a-tocopheryl
polyethylene glycol 1000 succinate (TPGS).
In order to obtain nanoparticles with higher
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
320
drug content and higher drug entrapment
efficiency, modification of the molecular
shape of the nanoparticles was carried out
into stars. Mannitol was used as the core of
the star polymer molecule in this study
because it has good chemical stability and is
suitable for use in the formulation of
anticancer tablets. The modified polymer was
named Mannitol-core PLGA-TPGS (M-
PLGA-TPGS) [26].
The organic phase consisted of M-
PLGA-TPGS alongside Genistein dissolved
by acetone while the aqueous phase consisted
of water. The characterization results showed
particle size of 225.7 ± 2.5 nm, polydispersity
index of 0.169, Zeta Potential of -14.2 ± 0.7
mV and encapsulation efficiency of 97.66 %.
The size of the nanoparticles is in the size
range that facilitates drug accumulation in
tumor blood vessels increased of drug
permeation and retention. Star-shaped
copolymers reduce particle size. The PDI
value indicates a relatively narrow size
distribution. Zeta potential is negative which
caused by ionized carboxyl groups from the
PLA and PGA. The encapsulation efficiency
is influenced by the binding affinity PLGA
which has star-shaped form and hydrophobic
Genistein [26].
3.11 Luteolin
In order to make biodegradable
luteolin nanoparticles, a biodegradable
polymer was chosen, polylactic acid (PLA).
Eudragit L100 is used in conjunction with
PLA as a polymer. This is because Eudragit
L100 is a pH-dependent polymer that is
resistant to gastric acidity and is soluble in
intestinal fluids. This polymer is suitable for
the active substance delivered to the large
intestine. In this research, two kinds of
organic phases were made. The first organic
phase has the composition Luteolin and
Eudragit L100 in Ethyl Alcohol. The second
organic phase is PLA in Dichloromethane.
The aqueous phase is Pluronic F127 which
acts as an emulsifier and is dissolved in HCl
pH 4 [36].
The resulting nanoparticles have a
particle size of 452.23 ± 22.4 nm, Zeta
Potential of 0.92 ± 0.04 mV, and
encapsulation efficiency of 71.02 ± 14.6%.
The higher concentration of total polymer
(combination of PLA and EUD100) led to the
production of larger nanoparticles. This is
due to an increase in the viscous force which
opposes the breakdown of the particles.
However, the viscous force increases
encapsulation efficiency. The presence of
PLA should make the zeta potential
negatively charged. The formation of a
PF127 layer on the particle surface makes the
surface charge a less negative value [36].
3.12 Naringenin
1. PVP-coated naringenin nanoparticles
Naringenin nanoparticles were
prepared by a simple nanoprecipitation
method using polyvinylpyrrolidone
(PVP) as a hydrophilic carrier. PVP is a
strong hydrophilic polymer which has
advantages such as delaying the
crystallization of compounds by forming
adduct molecules, non-toxic and
uncharged. PVP coating hydrophobic
surfaces increase biocompatibility and
reduce complement activation. The
manufacturing stage starts from
dissolving Naringenin in ethanol to make
the organic phase. The organic phase was
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
321
rapidly injected into the aqueous solution
containing the PVP. The injection process
was carried out alongside sonication of
mixture. The particle size obtained is 110
nm with encapsulation efficiency of
99.93%. The nitrogen atom coordinated
with naringenin provides stability and
prevents agglomeration [37].
2. Naringenin-loaded nanoparticles
In another study, naringenin
nanoparticles were fabricated using
Eudragit E and PVA. Eudragit E and
PVA were used as polymers/carriers
simultaneously. Eudragit E was chosen
because Eudragit has the function of
increasing the solubility of drugs that are
poorly soluble in water and have a
dimethylamino group base site that is
ionized in gastric juice. Naringenin
nanoparticles were made by dissolving
Naringenin and Eudragit® E in ethanol to
make an internal organic phase. The
phase is rapidly injected into the external
aqueous phase containing PVA. The
particle size obtained is 90 nm.
Nanoparticles do not form agglomerates
or adhesions. Judging from the particle
size, these nanoparticles can cross the
vascular endothelium and accumulate at
the tumor site through the EPR effect
because the nanoparticle size is less than
400 nm [25].
3.13 Phloretin
Phloretin is made into Hydrogel
preparations containing Floretin-charged
polymer nanocapsules. The core shell of the
nanocapsule can resist degradation caused by
ultraviolet light. PCL is used as a polymer
because it has advantages such as excellent
drug loading capacity, controlled release rate
and slower degradation compared to
poly(glycolic acid) and other polymers [38].
Phloretin-charged polymer
nanocapsules were prepared by mixing an
organic phase consisting of copaiba oil,
sorbitan monostearate (Span 60), PCL and
Floretin in acetone:ethanol (8:1). The
hydrophilic phase contains polysorbate 80
(Tween 80) and water. Before making
hydrogel with the addition of Lecigel® 1%,
Nanocapsules were evaluated first. The
evaluation results showed a particle size of
252 ± 12.01 nm and a polydispersity index of
1.68 ± 0.11. The zeta potential is negatively
charged and close to zero, due to the presence
of polysorbate 80 at the particle/water
interface. The highest concentration of drug
that can be put into nanocapsules with
maximum encapsulation efficiency (>99%)
is phloretin 0.2 mg/Ml. The size distribution
is fairly narrow because of the polydispersity
index results obtained [38].
3.14 Poly-puerarin
Poly-puerarin nanoparticles with
Paclitaxel were prepared by using the
nanoprecipitation method. Poly-puerarin
serves as a carrier. Poly-puerarin is obtained
from the modification of Puerarin with
unsaturated olefins via acryloyl chloride
through free radical polymerization.
Azodiisobutyronitrile (AIBN) is used as an
initiator in the puerarin modification process.
Poly-puerarin nanoparticles were prepared by
nanoprecipitation and used as a carrier for
loading paclitaxel. Before making
nanoparticles, first Poly-puerarin was
dissolved in DMSO. The first oil phase was
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
322
prepared by dissolving Paclitaxel in DMSO
at concentration. The second oil phase was
prepared by dissolving Stabilizer F-127 in a
different DMSO. The added buffer is saline
phosphate buffer [39].
In this study, there were two
variations of the formula where there was a
difference in the percentage of paclitaxel
used. Satisfactory results were obtained from
10% paclitaxel where the particle size
characterization results were 70.26 nm,
polydispersity index 0.15, Zeta Potential 23.1
mV and encapsulation efficiency of 91.3%.
Due to the intermolecular forces, increasing
the concentration of paclitaxel and puerarin
did not increase the particle size. The
hydrophobic drug Paclitaxel increases the
amount of the hydrophobic compound of the
nanoparticle complex which helps the
formation of a dehydrated and compact core.
Zeta potential is negatively charged due to
the strong electronegativity of glucose
groups, ester bonds and other functional
groups of poly-puerarin molecules [39].
3.15 Proantosianidin
Proanthocyanidins in grape seed
extract bind to type I collagen, inhibit matrix
metalloproteinase (MMP) activity and
decrease the rate of dentin demineralization.
To make a drug that can reduce the level of
demineralization of tink with gradual drug
release, Proanthocyanidin in the extract was
prepared in the form of nanoparticles with
different compositions of the ratio of PLGA
nanoparticles loaded with grape seed, which
have PLGA/Grape Seed Extract ratios of
100:25, 100:50, and 100:50. 75 w/w,
synthesized by a modified nanoprecipitation
technique [40].
The characterization results showed a
particle size of 132.5 ± 12.2 nm, Zeta
Potential 26.7 ± 3.8 mV and encapsulation
efficiency of 65.2 ± 6.1%. The nanoparticles
are uniformly spherical. The resulting
particle size aids drug delivery through the
exposed dentinal tubules of the
demineralized dentin. The OH functional
group present in the extract structure makes
the zeta potential negatively charged [40].
3.16 Quercetin
1. Psidium guajava ethyl acetate fraction
loaded nanosuspension
The ethyl acetate fraction from
Psidium guajava was made into nanoparticles
because this fraction has antihyperglycemic
activity. The fraction contains Quercetin
which has low solubility. The organic phase
was made by dissolving the ethyl acetate
fraction in ethanol while PVA was dissolved
in water. PVA works as a surfactant. The
characterization results showed that the
nanoparticles had a particle size of 241.32 ±
1.25 nm, a polydispersity index of 0.224 ±
0.011, a Zeta Potential of -22 ± 0.4 mV, and
an encapsulation efficiency of 92.85 ±
2.23%. The particle size is in the range of
100-500 nm so that absorption in the intestine
will be more effective. The polydispersity
index obtained shows the uniformity of
particle shape in the nanoparticles. The
optimal amount of PVA provides protective,
electrostatic stability to the particles and
reduces agglomeration. Zeta potential in the
range of 20-40 mV indicates the presence of
electrostatic stability [15].
2. Starch nanoparticles loaded with
Quercetin
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
323
Quercetin nanoparticles are made by
utilizing starch made from corn, potatoes or
beans. Starch is a natural polymer that is
biodegradable, inexpensive and abundant.
However, starch has low solubility and is
sensitive to temperature and humidity. For
this reason, modifications are made with
nano technology. Starch nanoparticles
containing quercetin were prepared by
dissolving quercetin into a solution of
NaOH/urea/H2O (0.8 : 1: 98.2) containing
starch. After that, HCl was added.
NaOH/urea/H2O solution is used as a solvent
because it can dissolve cellulose. Urea has the
function of preventing the assemblage of
starch molecules [24].
The results showed that the suitable
starch to be used as a polymer was potato
starch. This is because corn starch and peanut
starch form aggregates after lyophilization,
while potato starch has a uniform nanofiber-
like structure. Potato starch nanoparticles
have a particle size of 91.2 154.5 nm and a
polydispersity index of 0.276 0.41 [24].
3. MPEG-PLA encapsulated Quercetin
nanoparticle
Methoxy poly(ethylene glycol)-
poly(lactide) (MPEG-PLA) polymer is used
as a carrier to make quercetin nanoparticles
which will be used as an agent to treat breast
cancer. The reason why a modified form of
PLA is used is because the nanoparticles with
the PLA polymer are rapidly cleared from the
circulatory system after systemic injection.
The copolymerization of PLA with
Polyethylene glycol (PEG) can improve these
limitations. In addition, the charge of the
nanoparticles will shift to a neutral charge,
the neutral charge helps the interaction of
polymer nanoparticles with the cell
membrane. MPEG provides a liquid
membrane on the outside of the nanoparticles
to resist hydrophobic drug release for drugs
that function as sustained-release doses [16].
The organic phase consisted of
MPEG-PLA and quercetin in acetonitrile.
The organic phase was added to a solution
containing pluronic F-68. The resulting
nanoparticles had a particle size of 155.3 ±
3.2 nm, a polydispersity index of 0.2 ± 0.05
and a Zeta Potential of 3.14 mV. Particle
sizes below 200 nm suggest that
nanoparticles can promote accumulation at
tumor sites. Polydispersity index below 0.4
indicates that the nanoparticles are in
homogeneous condition. The Zeta Potential
value obtained states that the nanoparticles
can avoid RES [16].
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
324
4. Quercetin embedded PLA nanoparticles
Picture 2. Production of Quercetin embedded PLA nanoparticles [41]
Quercetin nanoparticles with
continuous release drug function were
prepared using the emulsion (o/w)
nanoprecipitation method. PLA is used as a
carrier while PVA is used as a stabilizer.
Quercetin and PLA allow the formulation of
sustained-release drugs. This is due to the
delayed diffusion and strong interaction
between Quercetin and PLA. In the
manufacture of nanoparticles, Quercetin is
dissolved in ethanol and then added PLA
which has been dissolved in
dichloromethane. After that, the mixture was
added with PVA dissolved in water [41].
In this study, a variant of the
formulation was made with the concentration
of PLA, PVA and changing temperature. The
best nanoparticles were produced from the
formulation with 10% Quercetin, 2% PVA
and 20 mg/ml PLA and the manufacturing
temperature was at 25oC. The nanoparticles
had a particle size of 46 ± 4 nm and an
encapsulation efficiency of 62 ± 3 %. The
size of the nanoparticles increased with
increasing PLA concentration. The particle
size gradually decreased with increasing
PVA concentration while the encapsulation
efficiency did not change drastically. The
lower particle size at higher temperatures is
due to their high mobility [41].
5. Zinc phthalocyanine-Quercetin
loaded lipid-polymer hybrid nanoparticle
Quercetin can function as photodynamic
therapy by making Lipid-polymer hybrid
nanoparticle (LPN) which encapsulates Zinc
phthalocyanine and Quercetin. Zinc-
phthalocyanine works as a second generation
photosensitizer while Quercetin as an
anticancer agent. Both substances must
accumulate in the tumor site. The LPN
structure consists of a non-aqueous polymer
core, a single or multiple lipid layer, and a
polyethylene glycol (PEG) conjugated lipid
layer. The polymer core encapsulates most of
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
325
the Quercetin and Zinc-phthalocyanine. The
lipid layer facilitates absorption by cells. The
PEG-conjugated lipid layer increases the
circulation time of the active substance in the
body [42].
Picture 3. Production of Lipid-polymer hybrid nanoparticle [42]
LPN made a modified nanoprecipitation
method using PLGA as a carrier. In this
method PLGA diffusion occurs between two
different liquid phases. The organic phase
was prepared by dissolving Quercetin, Zinc-
phthalocyanine and PLGA in Acetone. The
aqueous phase was prepared by dissolving
PF-68, soy lecithin and DSPE-PEG in a
mixture of ethanol and water. The resulting
nanoparticles had a particle size of 155.3 ±
3.2 nm, polydispersity index 0.2 ± 0.05, Zeta
Potential 3.14 mV and encapsulation
efficiency of 10 ± 4 % for Quercetin and 55
± 5% for Zinc-phthalocyanine. Particle sizes
below 200 nm prove that the preparation is
effective for use by the intravenous route.
The concentration of Lecithin and Zinc-
phthalocyanine did not affect the particle size
while the increase in the concentration of
Quercetin and PLGA increased the particle
size. The presence of a single layer of DSPE-
PEG increased the stability of the preparation
[42].
6. Quercetin-loaded Eudragit® S100
Nanoparticles
Quercetin which will be used for colon
cancer drug manufacture of Quercetin non-
particles with Eudragit S100 polymer as a
carrier because other studies have proven an
increase in the bioavailability and stability of
the drug [43].
The material used consists of an organic
phase and an aqueous phase. The organic
phase consisted of Eudragit S100 and
Quercetin dissolved in ethanol. Eudragit
S100 is suitable for drugs targeted against the
colon. This is due to the carboxyl group of the
methacrylic acid moiety contained in
Eudragit S100. The carboxyl group will
ionize upon contact with neutral or alkaline
pH. This results in a negative repulsion
between the carboxylate side groups causing
the polymer to lose its charge. This carboxyl
group also makes the polymer insoluble in
the stomach [43].
The resulting nanoparticles are colloidal
dispersion like milk which is stable. This
nanoparticle has a particle size of 66.8 ± 2.3
nm, Zeta Potential -5.2 ± 2.4 mV and
Encapsulation Efficiency of 41.8 ± 9.1%. A
negatively charged zeta potential is generated
due to the presence of sodium lauryl sulfate
added by to Eudragit S100. The
encapsulation efficiency obtained indicates
that some of the quercetin may have been
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
326
partitioned into the aqueous phase during the
nanoparticle formation process [43].
7. Quercetin-loaded PCL based
nanoparticles
Polycaprolactone (PCL) is used as a carrier
for the manufacture of quercetin
nanoparticles with various formulation
variants. PCL was chosen because of its high
permeability, biodegradability and
biocompatibility, non-toxic, non-mutagenic
and suitable for controlled release drugs. The
organic phase contains PCL and quercetin
dissolved in acetone. The aqueous phase
contains Pluronic F-127 as a stabilizer which
is dissolved in water. The resulting
nanoparticles have a particle size of 215.9 ±
2.9 nm, polydispersity index 0.094, Zeta
Potential 12.9 ± 0.35 mV and encapsulation
efficiency of 66.32 ± 0.4%. Increasing the
concentration of Pluronic F-127 increased the
particle size. Increasing the concentration of
PCL increases the viscosity of the organic
phase. The increase in viscosity increases the
diffusion resistance of the drug into the
aqueous phase and enhances the
encapsulation of the drug into nanoparticles
[44].
8. Quercetin conjugated Fe3O4
nanoparticles
The Nanoprecipitation method controls the
particle diameter and the unique magnetic
properties of the magnetite nanoparticles.
However, this method has limitations in the
manufacture of magnetic nanoparticles where
the agglomeration and distribution of
particles is not uniform, these limitations can
be overcome by the addition of urea. The
manufacturing process begins with
dissolving quercetin in acetone and
dissolving magnetite nanoparticles coated
with dextran in dimethyl sulfoxide (DMSO)
and deionized water. After that, the magnetite
nanoparticle solution was added with N-
hydroxy succinimide (NHS) and 1-ethyl-3-
(3-dimethylaminopropyl) carbodiimide
(EDC). Then the mixture was added to the
quercetin solution and the pH was adjusted
with KOH. Urea functions as an aggregation
controller and stabilizer to produce prism-
shaped particles. Dextran is a surfactant
which is used to control the shape of spherical
aggregates and to protect the phase change of
nanoparticles [11].
The best nanoparticles have a particle size of
72 nm, Zeta Potential 6.14 mV and
Efficiency of 81.6%. Particle size occurs due
to the conjugation of quercetin to Fe3O4
through the EDC/NHS reaction. The zeta
potential is positively charged due to the
strong electrostatic interaction between the
drug and the protonation of magnetite
through the carboxyl molecule and the
presence of anionic ions from the drug
molecule. Positive charge can increase drug
penetration into tumor cells [11].
3.17 Rutin
Rutin was made into nanospheres which were
prepared by the nanoprecipitation method.
Eudragit S100 functions as a polymer and
Poloxamer-188 as a stabilizer. Nanosphere is
a particle matrix in which the entire mass of
solid and drug particles is physically and
uniformly dispersed. Eudragit S100 was used
as a polymer because it does not degrade
below pH 7. In other words, this polymer is
insoluble in the pH of the stomach and
intestines, but soluble in the pH of the large
intestine (pH > 7) [45].
Nanosphere was prepared by dissolving
Eudragit S100 and rutin in a sealed bottle
containing methanol to make the organic
phase and dissolving Poloxamer-188 in
distilled water. The characterization results
show that the nanosphere has a particle size
of 130.30 ± 35.29 nm, polydispersity index
0.29 ± 0.10, Zeta Potential -22.90 ± 5.18 mV
and encapsulation efficiency of 98.10 ±
0.50%. The high encapsulation efficiency is
due to rutin has a high affinity for organic
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
327
solvents that dissolve the dissolved polymer
and the interaction between rutin and
Eudragit S100 which indicates the formation
of intermolecular hydrogen bonds. The
polydispersity index value obtained indicates
that the nanoparticles are stable without any
agglomerates. The negative charge on the
zeta potential measurement was attributed to
the free acrylic acid group of Eudragit S100
as an anionic polymer [45].
3.18 Salvigenin
In order to overcome its poor bioavailability,
a study was conducted on the preparation of
Salvigenin loaded mPEG-b-PLGA coated
with iron oxide (Fe3O4) nanoparticles.
PLGA is used as a polymer because PLGA
has various intrinsic physical and chemical
properties such as monomer ratio (PL/GA),
drug loading potential, and controllable size
and shape of drug polymer particles. The
addition of iron oxide particles into the
polymer nanocarrier is very important
because it allows the nanoparticles to be
delivered by a magnetic field to the tumor site
[23].
Before making Salvigenin nanoparticles,
Fe3O4 nanoparticles must be made first with
the main ingredients FeCl3.6H2O and
FeCl2.4H2O with a molar ratio of 2:1,
ammonia solution as a precipitation agent and
oleic acid as a surfactant. In the process of
making salvigenin nanoparticles, the organic
phase was prepared by dissolving mPEG-b-
PLGA and Salvigenin in acetone. Fe3O4
solution was dissolved in different acetone.
Then the two solutions are mixed. After that
it was added to 20 mL of distilled water drop
by drop. After the addition of the complete
mixture, the acetone was removed under
vacuum with a rotary evaporator [23].
The characterization results showed that
Salvigenin loaded with mPEG-b-PLGA
coated with iron oxide nanoparticles had a
particle size of 57 ± 2 nm, a polydispersity
index of 0.168 ± 0.03, Zeta Potential 33 ± 1.2
mV and an encapsulation efficiency of 82 ±
1.6%. Nanoparticles are said to be stable and
positively charged based on the
characterization results [23].
3.19 Silibinin
Silibinin nanoparticles are prepared using the
nanoprecipitation method and utilizing
Eudragit and PVA. Eudragit was used as a
polymer and PVA was used as a stabilizer.
The reason for using Eudragit is because
Eudragit is a positive copolymer which has a
base site for ionized tertiary amine groups in
gastric juice which can make nanoparticles
easily soluble in the stomach [46].
Silibinin nanoparticles have the composition
Silibinin:Eudragit:PVA (1:10:10;w/w/w).
The organic phase was prepared by
dissolving Silibinin into ethanol in a closed
glass bottle. The aqueous phase contains
PVA. The characterization results showed
that Silibinin nanoparticles had a particle size
of 120 nm, Zeta Potential of 4.6 mV and
encapsulation efficiency of 79.0 ± 2.4%.
Small particle size < 200 nm so that drug
accumulation in tumor cells increases. The
positive charge is suitable for drugs that are
absorbed by charged cellular membranes
[46].
3.20 Silymarin
1. Silymarin-loaded lipid polymer
hybrid nanoparticles containing
chitosan
Silymarin was prepared into lipid-polymer
hybrid nanoparticles. This type of
nanoparticle utilizes liposomes and polymer
nanoparticles. These nanoparticles are
shaped like a shell core consisting of a
polymer core and a phospholipid wall. The
polymer used is chitosan. This is because
Chitosan has an amino group that can change
the charge to be positive when in a weak acid
state. The positive charge allows the
compound to adsorb mucin on the intestinal
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
328
mucosa through electrostatic interactions
[47].
The lipid-polymer hybrid of Silymarin
nanoparticles was prepared using a two-step
nanoprecipitation method. In this method,
lipid-polymer hybrid nanoparticles were
obtained by incubating the nanoparticle
dispersion with chitosan solution after
evaporation of the organic solvent. The
aqueous phase was prepared by dissolving
soybean lecithin and DSPE-PEG 2000 in
ethanol. The Organic Phase was prepared by
dissolving PLGA and silymarin in a mixture
of acetonitrile-methanol. Chitosan solution
was prepared by dissolving chitosan in 0.5%
acetic acid and then filtering through a 0.8 m
membrane [47].
After being characterized, the lipid-polymer
hybrid nanoparticles Silymarin with Chitosan
had a particle size of 286.5 ± 23.8 nm, a
polydispersity index of 0.226 ± 0.008, a Zeta
Potential of 45.3 ± 8.9 mV and an
encapsulation efficiency of 97.05 ± 0.01%.
The addition of chitosan increases the particle
size and changes the zeta potential charge
from negative to positive. In addition, the oral
bioavailability of silymarin in this form was
14.38 times higher than that of nanoparticles
without chitosan [47].
2. Silymarin-Loaded Eudragit
Silymarin is manufactured in the form of
Silymarin Loaded Eudragit Nanoparticles.
This is because Eudragit Polymer has the
ability to form positively charged
nanodispersions, moderate bio-adhesive
strength, and has no irritating effect on the
mucosal surface. Silymarin Loaded Eudragit
Nanoparticles were prepared by the
nanoprecipitation method. First, an organic
phase was made with a polymer composition
of silymarin and Eudragit with a constant
ratio of Eudragit RS 100 & Eudragit LS 100
(1:1 w/w) dissolved in acetone. The aqueous
phase having the composition of deionized
water containing PVA. PVA is a synthetic
polymer that is soluble in water and functions
as a stabilizer. Eudragit RS100 has very low
water permeability, while Eudragit RL100
has high water permeability. To improve the
quality of the drug release rate, these multiple
polymers were used concurrently [14].
In this study, nine formulations were made
with three different concentrations of PVA
(1, 2, 3% w/v) and three different
organic/water phase ratios (1:6, 1:10, 1:20
v/v). The optimal formulation chosen was a
drug/polymer ratio of 1:1 and stirring for 5
minutes at 480 rpm on a magnetic stirrer
followed by homogenization for 30 minutes
at 23.5 krpm. The optimal formulation has
characterization results with a particle size of
84.70 nm, a polydispersity index of 0.38 ±
0.01, and an encapsulation efficiency of
84.35 %. In addition, the oral bioavailability
of silymarin in this form was 14.38 times
higher than that of nanoparticles without
chitosan. When the PVA concentration was
increased, the particle size and polydispersity
index increased at low organic phase ratios to
water (1:6) while decreasing at high organic
phase ratios to water (1:10 and 1:20).
Moreover, at higher organic to water phase
ratio and as PVA concentration increased,
particle size and entrapment efficiency
decreased [14].
4. Conclusion
Based on the literature review that has been
done, there are various nanoprecipitation
methods that have been applied to the
preparation of flavonoid nanoparticles from
20 prepared secondary metabolites of
flavonoids. These methods consist of
Traditional Nanoprecipitation, Heat induced
evaporative antisolvent nanoprecipitation,
and Sonication-assisted nanoprecipitation.
The most frequently used method is
Traditional Nanoprecipitation.
The types of flavonoid nanoparticles which
were produced are polymeric nanoparticles,
lipid-polymer hybrid nanoparticles, magnetic
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
329
nanoparticles, organic-inorganic hybrid
nanoparticles and starch nanoparticles.
Polymeric nanoparticles are a type of
nanoparticle that is often produced.
Nanoparticles with the smallest particle size
were obtained from research on production of
Quercetin nanoparticles using PLA as a
carrier and PVA as a stabilizer using the
Sonication-assisted nanoprecipitation
method. PLGA in the manufacture of
Apigenin nanoparticles using the Traditional
Nanoprecipitation method produced the
lowest polydispersity index value. The use of
PVP and PCL in research on the manufacture
of Artocarpin, Naringenin and Floretin
nanoparticles resulted in an encapsulation
efficiency of greater than 99%.
Acknowledgements
Author of this article is grateful to the lecturer
who has been supportive and helpful in
providing data and information for the
purpose of this study.
A. K. Umar et al / Indo J Pharm 3 (2021) 57-70
330
References
[1] Kumar S, Pandey A. Chemistry and
Biological Activities of Flavonoids: An
Overview. The Scientific World Journal.
2013;2013:1-16.
[2] Ullah A, Munir S, Badshah S, Khan
N, Ghani L, Poulson B et al. Important
Flavonoids and Their Role as a Therapeutic
Agent. Molecules. 2020;25(22):5243.
[3] Modasiya MK, Patel JN, Rathod
DM, Patel NA. Techniques to improve the
solubility of poorly soluble drugs.
International Journal of Pharmacy and Life
Sciences. 2012;3(2):1459-1469
[4] Savjani KT, Gajjar AK, Savjani JK.
Drug solubility: importance and
enhancement techniques. ISRN Pharm.
2012;2012:195727.
doi:10.5402/2012/195727
[5] Barreras-Urbina CG, Ramírez-
Wong B, López-Ahumada GA, et al. Nano-
and Micro-Particles by Nanoprecipitation:
Possible Application in the Food and
Agricultural Industries, International
Journal of Food Properties. 2016;19:1912-
1923.
[6] Dobrzynska M, Napierala M,
Florek E. Flavonoid Nanoparticles: A
Promising Approach for Cancer Therapy.
Biomolecules. 2020;10(9):1268.
[7] Ansari MJ. Factors Affecting
Preparation and Properties gf Nanoparticles
by Nanoprecipitation Method. IAJPS.
2018;4(12):4854-4858.
[8] Massella D, Celasco E, Salaün F,
Ferri A, Barresi AA. Overcoming the
Limits of Flash Nanoprecipitation:
Effective Loading of Hydrophilic Drug into
Polymeric Nanoparticles with Controlled
Structure. Polymers (Basel).
2018;10(10):1092.
[9] Wang Y, Li P, Truong-Dinh Tran T,
Zhang J, Kong L. Manufacturing
Techniques and Surface Engineering of
Polymer Based Nanoparticles for Targeted
Drug Delivery to Cancer. Nanomaterials
(Basel). 2016;6(2):26.
[10] Awouafack MD, Tane P, Morita H.
Isolation and Structure Characterization of
Flavonoids. In: Justino, G. C. , editor.
Flavonoids - From Biosynthesis to Human
Health [Internet]. London: IntechOpen;
2017 [cited 2022 Jul 19]. Available from:
https://www.intechopen.com/chapters/545
24 doi: 10.5772/67881
[11] Kumar SR, Priyatharshni S, Babu
VN, et al. Quercetin conjugated
superparamagnetic magnetite nanoparticles
for in-vitro analysis of breast cancer cell
lines for chemotherapy applications. J
Colloid Interface Sci. 2014;436:234-242.
[12] Sechi M, Syed DN, Pala N, et al.
Nanoencapsulation of dietary flavonoid
fisetin: Formulation and in vitro antioxidant
and α-glucosidase inhibition activities.
Mater Sci Eng C Mater Biol Appl.
2016;68:594-602
[13] Zhao J, Yang J, Xie Y.
Improvement strategies for the oral
bioavailability of poorly water-soluble
flavonoids: An overview. Int J Pharm.
2019;570:118642.
doi:10.1016/j.ijpharm.2019.118642
[14] El-Nahas AE, Allam AN,
Abdelmonsif DA, El-Kamel AH.
Silymarin-Loaded Eudragit Nanoparticles:
Formulation, Characterization, and
Hepatoprotective and Toxicity Evaluation.
AAPS PharmSciTech. 2017;18(8):3076-
3086.
[15] Gaur PK. Nanosuspension of
flavonoid-rich fraction from Psidium
guajava Linn for improved type 2-diabetes
potential. Journal of Drug Delivery Science
and Technology. 2021;62:102358.
[16] Sharma G, Park J, Sharma AR, et al.
Methoxy poly(ethylene glycol)-
poly(lactide) nanoparticles encapsulating
quercetin act as an effective anticancer
agent by inducing apoptosis in breast
cancer. Pharm Res. 2015;32(2):723-735.
[17] Wathoni N, Nguyen AN, Rusdin A,
et al. Enteric-Coated Strategies in
Colorectal Cancer Nanoparticle Drug
Delivery System. Drug Des Devel Ther.
2020;14:4387-4405.
[18] Yao Y, Zhou Y, Liu L, et al.
Nanoparticle-Based Drug Delivery in
Cancer Therapy and Its Role in
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
331
Overcoming Drug Resistance. Frontiers in
Molecular Biosciences. 2020;7.
[19] Biriukov D, Fibich P, Předota M.
Zeta Potential Determination from
Molecular Simulations. The Journal of
Physical Chemistry C. 2020;124(5):3159-
3170
[20] Clogston JD, Patri AK. Zeta
potential measurement. Methods Mol Biol.
2011;697:63-70.
[21] Kharia A, Singhai A, Verma R.
Formulation and Evaluation of Polymeric
Nanoparticles of an Antiviral Drug for
Gastroretention. International Journal of
Pharmaceutical Sciences and
Nanotechnology. 2012;4:1557-1562.
[22] Lv Y, He H, Qi J, et al. Visual
validation of the measurement of
entrapment efficiency of drug nanocarriers.
Int J Pharm. 2018;547(1-2):395-403.
[23] Mansourizadeh F, Sepehri H,
Khoee S, Farimani M, Delphi L, Tousi M.
Designing Salvigenin loaded mPEG-b-
PLGA @Fe3O4 nanoparticles system for
improvement of Salvigenin anti-cancer
effects on the breast cancer cells, an in vitro
study. Journal of Drug Delivery Science
and Technology. 2020;57:101619.
[24] Farrag Y, Ide W, Montero B, et al.
Preparation of starch nanoparticles loaded
with quercetin using nanoprecipitation
technique. Int J Biol Macromol.
2018;114:426-433.
[25] Krishnakumar N, Sulfikkaralia N,
RajendraPrasad N, Karthikeyan S.
Enhanced anticancer activity of naringenin-
loaded nanoparticles in human cervical
(HeLa) cancer cells. Biomedicine and
perventive nutrition. 2011.
[26] Wu B, Liang Y, Tan Y, et al.
Genistein-loaded nanoparticles of star-
shaped diblock copolymer mannitol-core
PLGA-TPGS for the treatment of liver
cancer. Mater Sci Eng C Mater Biol Appl.
2016;59:792-800.
[27] Ganguly S, Dewanjee S, Sen R, et
al. Apigenin-loaded galactose tailored
PLGA nanoparticles: A possible strategy
for liver targeting to treat hepatocellular
carcinoma. Colloids Surf B Biointerfaces.
2021;204:111778.
[28] Tzeng CW, Tzeng WS, Lin LT, Lee
CW, Yen FL, Lin CC. Enhanced
autophagic activity of artocarpin in human
hepatocellular carcinoma cells through
improving its solubility by a nanoparticle
system. Phytomedicine. 2016;23(5):528-
540.
[29] El-Hussien D, El-Zaafarany GM,
Nasr M, Sammour O. Chrysin
nanocapsules with dual anti-glycemic and
anti-hyperlipidemic effects: Chemometric
optimization, physicochemical
characterization and pharmacodynamic
assessment. Int J Pharm. 2021;592:120044.
[30] Al-Shalabi E, Alkhaldi M, Sunoqrot
S. Development and evaluation of
polymeric nanocapsules for cirsiliol
isolated from Jordanian Teucrium polium
L. as a potential anticancer nanomedicine.
Journal of Drug Delivery Science and
Technology. 2020;56.
[31] Dhas N, Mehta, TA. Intranasal
delivery of Chitosan decorated PLGA core
/shell nanoparticles containing flavonoid to
reduce oxidative stress in the treatment of
Alzheimer’s disease. Journal of Drug
Delivery Science and Technology.
2020;61.
[32] Meena R, Kumar S, Kumar R,
Gaharwar US, Rajamani P. PLGA-CTAB
curcumin nanoparticles: Fabrication,
characterization and molecular basis of
anticancer activity in triple negative breast
cancer cell lines (MDA-MB-231 cells).
Biomed Pharmacother. 2017;94:944-954.
[33] Dalcin A, Roggia I, Felin S,
Vizzotto B, Mitjans M, Vinardell M, et al.
UVB photoprotective capacity of hydrogels
containing dihydromyricetin nanocapsules
to UV-induced DNA damage. Colloids and
Surfaces B: Biointerfaces.
2021;197:11431.
[34] Freag M, Elnaggar Y, Abdallah O.
Development of novel polymer-stabilized
diosmin nanosuspensions: In vitro appraisal
and ex vivo permeation. International
journal of pharmaceutics. 2013; 454.
R F. Pratama et al / Indo J Pharm 4 (2023) 307-332
332
[35] Zhang H, Chen MK, Li K, Hu C, Lu
MH, Situ J. Eupafolin nanoparticle
improves acute renal injury induced by LPS
through inhibiting ROS and inflammation.
Biomed Pharmacother. 2017;85:704-711.
[36] Elmowafy M, Alhakimy NA,
Shalaby K, et al. Hybrid polylactic
acid/Eudragit L100 nanoparticles: A
promising system for enhancement of
bioavailability and pharmacodynamic
efficacy of luteolin. 2021;65.
[37] Kumar RP, Abraham A. PVP-
coated naringenin nanoparticles for
biomedical applications - In vivo
toxicological evaluations. Chem Biol
Interact. 2016;257:110-118.
[38] Casarini TPA, Frank LA, Benin T,
Onzi G, Pohlmann AR, Guterres SS.
Innovative hydrogel containing polymeric
nanocapsules loaded with phloretin:
Enhanced skin penetration and adhesion.
Mater Sci Eng C Mater Biol Appl.
2021;120:111681.
[39] Yi T, Huang J, Chen X, Xiong H,
Kang Y, Wu J. Synthesis, characterization,
and formulation of poly-puerarin as a
biodegradable and biosafe drug delivery
platform for anti-cancer therapy.
Biomaterials Science. 2019;7.
[40] Fawzy AS, Priyadarshini BM,
Selvan ST, Lu TB, Neo J.
Proanthocyanidins-Loaded Nanoparticles
Enhance Dentin Degradation Resistance. J
Dent Res. 2017;96(7):780-789.
[41] Pandey SK, Patel DK, Thakur R,
Mishra DP, Maiti P, Haldar C. Anti-cancer
evaluation of quercetin embedded PLA
nanoparticles synthesized by emulsified
nanoprecipitation. Int J Biol Macromol.
2015;75:521-529.
[42] Thakur NS, Mandal N, Patel G, et
al. Co-administration of zinc
phthalocyanine and quercetin via hybrid
nanoparticles for augmented photodynamic
therapy. Nanomedicine. 2021;33:102368.
[43] Sunoqrot S, Abujamous L. pH-
sensitive polymeric nanoparticles of
quercetin as a potential colon cancer-
targeted nanomedicine. Journal of Drug
Delivery Science and Technology.
2019;52:670-676.
[44] Kumar V. Verma P, Singh S.
Development and evaluation of
biodegradable polymeric nanoparticles for
the effective delivery of quercetin using a
quality by design approach. LWT - Food
Science and Technology. 2015;61.
[45] Asfour MH, Mohsen AM.
Formulation and evaluation of pH-sensitive
rutin nanospheres against colon carcinoma
using HCT-116 cell line. J Adv Res.
2017;9:17-26.
[46] Gohulkumar M, Gurushankar K,
Rajendra Prasad N, Krishnakumar N.
Enhanced cytotoxicity and apoptosis-
induced anticancer effect of silibinin-
loaded nanoparticles in oral carcinoma
(KB) cells. Mater Sci Eng C Mater Biol
Appl. 2014;41:274-282.
[47] Liang J, Liu Y, Liu J, et al.
Chitosan-functionalized lipid-polymer
hybrid nanoparticles for oral delivery of
silymarin and enhanced lipid-lowering
effect in NAFLD. Journal of
Nanobiotechnology. 2018;16.
[48] Liu Y, et al. Formulation of
Nanoparticles Using Mixing-Induced
Nanoprecipitation for Drug Delivery. Ind.
Eng. Chem. Res. 2020;59(9): 41344149.
[49] Bosselmann S, Nagao M, Chow K
T, et al. Influence of formulation and
processing variables on properties of
itraconazole nanoparticles made by
advanced evaporative precipitation into
aqueous solution. AAPS PharmSciTech.
2012;13(3):949960.
[50] Mugheirbi N, Paluch K, Tajber L.
Heat induced evaporative antisolvent
nanoprecipitation (HIEAN) of
itraconazole. International journal of
pharmaceutics. 2014.
[51] Baldassarre F, et al. Sonication-
Assisted Production of Fosetyl-Al
Nanocrystals: Investigation of Human
Toxicity and In Vitro Antibacterial
Efficacy against Xylella Fastidiosa.
Nanomaterials. 2020;10(6):1174.

Refbacks

  • There are currently no refbacks.


Copyright (c) 2023 Rizky Farhan Pratama, Iyan Sopyan, Taofik Rusdiana

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.

 site stats View My Stats 

Licensed :

Creative Commons Attribution license icon
IdJP is licensed under a Creative Commons Attribution-4.0 International (CC BY 4.0) License

  Universitas Padjadjaran. Back to Top
 
https://international.ui.ac.id/wp-content/uploads/2023/01/ https://dispendukcapil.kedirikota.go.id/gambar/raffi/ https://bahkapulajjah.pematangsiantar.go.id/xdemo/ https://dispendukcapil.kedirikota.go.id/gambar/demo/ sule slot toto slot https://sieni.dinamika.ac.id/codebase/ https://gcrf.architecture.ui.ac.id/wp-content/slothoki/ https://gcrf.architecture.ui.ac.id/wp-content/usutoto/ https://gcrf.architecture.ui.ac.id/wp-content/slot88/ https://gcrf.architecture.ui.ac.id/wp-content/akurat77/ https://appem.kuningankab.go.id/toto-slot/ https://appem.kuningankab.go.id/toto4d/ https://appem.kuningankab.go.id/slot777/ https://appem.kuningankab.go.id/slot88/ https://appem.kuningankab.go.id/slothoki/
https://www.store.smkind-mm2100.sch.id/ sule slot situs slot toto slot https://edatabase.dpmptsp.riau.go.id/demo/ https://www.kideco.co.id/wp-content/slot88/ https://appem.kuningankab.go.id/slot-gacor/index.html slot online https://appem.kuningankab.go.id/public/s-demo/ https://igtc-indo.or.id/xgaming/ slot online gacor https://simanis.jayawijayakab.go.id/vendor/ https://international.ui.ac.id/wp-content/uploads/2019/02/-/xgaming/ https://international.ui.ac.id/wp-content/uploads/2019/02/-/raffi/ https://international.ui.ac.id/wp-content/uploads/2019/02/-/sule/