You are on page 1of 11

International Journal of Pharmaceutics 554 (2019) 224–234

Contents lists available at ScienceDirect

International Journal of Pharmaceutics


journal homepage: www.elsevier.com/locate/ijpharm

Review

Modified gelatin nanoparticles for gene delivery T


a,b,1 a,c,1 a,d,⁎,1
Osama Madkhali , George Mekhail , Shawn D. Wettig
a
School of Pharmacy, University of Waterloo, Waterloo ON, N2L 3G1, Canada
b
Department of Pharmaceutics, Faculty of Pharmacy, Jazan University, Jizan, Saudi Arabia
c
Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
d
Waterloo Institute of Nanotechnology, University of Waterloo, Waterloo ON, N2L 3G1, Canada

A R T I C LE I N FO A B S T R A C T

Keywords: Gelatin nanoparticles (GNPs) are one of the most extensively used natural polymers for gene therapy. With
Gelatin advantages of being biodegradable, biocompatible, low cost and easily modified, gelatin holds great promise as a
Gene delivery non-viral system for gene delivery. This review examines various methods of preparation of modified gelatin
Modification nanoparticles and considers how these modifications apply to gene delivery. The article discusses cationic ge-
Method
latin, PEGylated gelatin, thiolated gelatin, alendronate gelatin, and EGFR gelatin nanoparticles. This article also
considers several advantages of these modifications and their contribution to the improvement in the efficiency
of these systems, resulting in superior transfection and enhanced gene delivery in general.

1. Introduction require a delivery vehicle in order to efficiently travel through extra-


and intra-cellular barriers. Gene delivery vectors offer the nucleic acids
Gene therapy (GT) has received much attention due to its great the necessary protection against various nucleases encountered during
potential for the treatment of both acquired and inherited diseases such its travel to the target site. Moreover, delivery vectors might enhance
as cancer, cystic fibrosis (CF), acquired immunodeficiency syndrome the ability of the nanocomplexes to penetrate the cell membrane via
(AIDS), X-linked combined immune deficiency (X-linked SCID), em- various penetrating mechanisms. Gene carriers also play a crucial role
physema, retinitis pigmentosa, sickle cell anemia, hemophilia, in endosomal escape via different mechanisms (Cardoso et al., 2014;
Duchenne Muscular Dystrophy (DMD), some autosomal dominant dis- Karimi et al., 2015; Lacerda et al., 2012; Trabulo et al., 2010). Finally,
orders, vascular disease, neurodegenerative disorders, polygenic dis- the delivery vector enters the nucleus to express the required protein to
orders, inflammatory conditions, and other infectious diseases (Keeler correct or moderate specific diseases. Gene therapy is based on two
et al., 2017; Nayerossadat et al., 2012; Stone, 2010). A gene therapeutic types of delivery systems: viral and non-viral vectors. Viral vectors are
should fulfil two characteristics: (a) it should contain an active sub- more effective and generally have longer gene expression depending on
stance containing or consisting of a recombinant nucleic acid that is the nucleic acid type, virus type and its ability to combine the trans-
delivered to the nucleus in order to regulate, repair, replace, add, or gene with the host cell genome (Lundstrom and Boulikas, 2003). For
delete a defective gene; (b) its therapeutic, diagnostic, or prophylactic example, single stranded RNA viruses, such as alphaviruses exhibit a
effect relates to the recombinant nucleic acid it contains (Wirth et al., rapid but transient gene expression. Some of the DNA viruses, like
2013). When gene therapy is used in the treatment of genetic diseases, adenovirus, possess short term gene expression. However, adeno-asso-
it restricts these diseases through introducing genes coding for func- ciated viruses predictably engender long-term expression owing to their
tional proteins to cells; thus, it normalizes the cells and even organs in ability to integrate into the host genome. Finally, retroviruses, double-
question (Jin et al., 2014). Since the first attempt of gene therapy in stranded RNA viruses, could similarly induce long-term expression
1928 done by Fredrick Griffith (Griffith, 1928), enormous and sig- through chromosomal integration (Lundstrom and Boulikas, 2003;
nificant changes have been seen in the development and improvement Nayerossadat et al., 2012). Yet viral vectors have several concerns re-
of gene therapy. The most important change was the development of garding safety. As a result, an alternative method is the use of non-viral
delivery systems. Nucleic acids (such as plasmid DNA, messenger RNA vectors. They are safe, non-toxic, cheap, have low immunogenicity and
(mRNA), small interfering RNA (siRNA), and micro RNA (miRNA)) can be produced in scalable batches. Although they suffer from short-


Corresponding author at: School of Pharmacy, University of Waterloo, 200 University Ave. W., Waterloo, ON N2L 3G1, Canada.
E-mail address: wettig@uwaterloo.ca (S.D. Wettig).
1
All authors contributed equally to this work.

https://doi.org/10.1016/j.ijpharm.2018.11.001
Received 30 July 2018; Received in revised form 31 October 2018; Accepted 1 November 2018
Available online 05 November 2018
0378-5173/ © 2018 Elsevier B.V. All rights reserved.
O. Madkhali et al. International Journal of Pharmaceutics 554 (2019) 224–234

term gene expression yet, efforts are exerted to tailor suitable non-viral 2.5. Magnetofection
vectors with sufficient gene expression period (Jackson et al., 2006; Yin
et al., 2014). Two main categories of non-viral vectors have been used: The magnetofection technique is based on coupling therapeutic
physical and chemical methods. gene to magnetic nanoparticles, which are then introduced into the cell
culture (Jones et al., 2013). The field gradient is produced by adding
2. Physical methods of non-viral systems rare, earth electromagnets under the cell culture, which then result in
increasing transfection speed that arises from increasing the complex
These methods depend on using physical force in order to destabi- sedimentation. The therapeutic gene-magnetic particle complex is ad-
lize the cellular membrane, therefore facilitating the entry of gene ministered intravenously when it used in vivo. With the help of enzy-
therapeutic materials into the cells. These methods are simple and matic cleavage of cross linking molecules, charge interaction, or charge
straightforward. degradation, the genetic material is released (Plank et al., 2003). This
method is considered to be an alternative for certain primary cells, as
those transfections are difficult when using other techniques.
2.1. Electroporation

3. Chemical methods of non-viral systems


Electroporation is known as gene electro injection, gene electro
transfer, electrically mediated gene therapy, or electro gene transfer
Chemical methods of transfection are divided into two categories:
(Ramamoorth and Narvekar, 2015). It works by applying an electric
inorganic particles (such as calcium phosphate, silica, and gold parti-
field greater than the membrane capacitance into the targeted tissue
cles); and organic synthetic/natural materials (such as cationic lipid
cell membrane, resulting in a pore that allows the molecules to pass
and cationic polymers). Lipoplexes (cationic lipid and DNA) and poly-
through it. As a result, the previously injected DNA can enter into the
plexes (cationic polymer and DNA) are the most commonly used non-
cytoplasm and nucleoplasm of the cell (Nayerossadat et al., 2012). This
viral delivery vectors.
method is very effective and safe when it applied in vivo in comparison
to other non-viral methods. However, the complexity of surgical pro-
4. Gelatin
cedures, and high voltage [ > 700 V/cm] applied to the tissues, as well
as the difficulty of reaching some internal tissues makes this method
Gelatin, as a natural polymer, is one of the most effective non-viral
inappropriate for delivering DNA (Young and Dean, 2015).
vectors that has been used in the last two decades (Sabet et al., 2017).
Gelatin is extracted from animal collagen either through partial acid
2.2. Gene gun
(Type A) or alkaline (Type B) hydrolysis. Cationic gelatin (type A with
an isoelectric point of (IEP) of 7–9), is derived from partial acid hy-
The gene gun (also known as particle bombardment, micro pro-
drolysis of pig skin type 1 collagen, and anionic gelatin (type B, IEP
jectile gene transfer or ballistic DNA) delivers DNA coated heavy metal
4.8–5) is derived from alkaline bovine collagen (Fig. 1) (Patel et al.,
particles into the target tissue at a particular speed using high voltage
2008). Alkaline treatment causes respective hydrolysis of asparagine
electronic discharge, spark discharge, or high pressure inert gas, usually
and glutamine to aspartate and glutamate. Thus, the greater proportion
helium (Mali, 2013). The most common metal particles used in this
of carboxylic groups possessed by type B gelatin increases its negative
method are gold, tungsten, and silver, which all typically measure 1 µm
charge and lowers its IEP (Ninan et al., 2011).
in diameter. Gene transfer is affected by several parameters such as gas
Gelatin is distinguished from other polymers by having amino acid
pressure, particle size, and dose frequency. Precise delivery of DNA is
sequences such as Arg-Gly-Asp (RGD) in its structure. These amino acid
the most important advantage using the gene gun method, and it most
sequences modulate cell adhesion; consequently, they play a significant
commonly used in gene therapy that targets ovarian cancer cells in vitro
role in the final biological performance of gelatin in comparison to
(Ramamoorth and Narvekar, 2015).
synthetic polymers that lack these cell-recognition sites (Wang et al.,
2012). Gelatin is considered to be GRAS (generally regarded as safe)
2.3. Sonoporation according to the United States Food and Drug Administration (FDA)
(Kumar, 2005), and therefore it has been used in various pharmaceu-
Sonoporation is a noninvasive technique using ultrasound wave to tical, cosmetic, and food products for decades (Elzoghby et al., 2012;
permeabilize the cell membrane; thus, allowing the uptake of DNA. Kommareddy et al., 2005a; Lemieux et al., 2000). Gelatin is an am-
Genetic materials of interest are first administered into the circulation phiphilic polymer having both cationic and anionic charges along with
using microbubbles, followed by the application of the ultrasound hydrophobic groups present in the approximate ratio of 1:1:1 (Fig. 2).
waves. The ultrasound waves cavitate and break up the microbubbles Gelatin consists of eighteen non-uniformly distributed amino acids with
within the microcirculation of target tissue, leading to the disruption of both positive and negative charges (Samal et al., 2012). This compo-
the nearby cell membrane that results in targeted transfection of the sition gives rise to the special nature of this polypeptide. Lysine and
therapeutic gene (Omata et al., 2015). The major advantages of sono- arginine represent 13% of gelatin, and both possess a positive charge;
poration include safety, noninvasiveness, and the ability to reach in- 12% of the polymer is comprised of negatively charged glutamic and
ternal organs without the necessity of surgery; consequently, it is used aspartic acid groups. The hydrophobic group consists of leucine, iso-
in the brain, cornea, kidney, and peritoneal cavity, as well as in muscle leucine, methionine, and valine, representing 11% of the gelatin
and heart tissues (Ramamoorth and Narvekar, 2015; Ter Haar, 2007). structure. The rest of the chain includes glycine, proline, and hydro-
xyproline. During either acid or alkaline collagen hydrolysis, the posi-
2.4. Photoporation tions where the bonds break determine the molecular weight, the
number of polypeptide chains and the number of each kind of amino
This technique works by using a single laser pulse in order to gen- acid residue of the formed gelatin. However, there is no specific bond
erate a pore in the cell membrane allowing the DNA to enter into the proven to be more labile to break during collagen partial hydrolysis.
cells. The effectiveness of this method depends on the focal point and Multiple bond positions are vulnerable to breakage on a probability
pulse frequency of the laser. The major advantage of this approach is its basis, depending on pH and temperature rendering random bond hy-
safety, in which the pore that is formed by the laser can be healed in less drolysis which is the main reason of gelatin molecular heterogeneity.
than a second. However, the lack of documented evidence limits the use Moreover, the collagen source itself contributes to this variation, since
of this technique (Li and Huang, 2007). different animal species, tissue, age, and sex are all factors implicated in

225
O. Madkhali et al. International Journal of Pharmaceutics 554 (2019) 224–234

Fig. 1. Extraction of gelatin from collagen. The production of Type A (cationic) gelatin is shown on the right of the figure; the production of Type B (anionic) gelatin
is shown on the left of the figure. Reproduced with permission from (Hosseinkhani et al., 2015).

variable collagen amino acid composition. Accordingly, it is a challenge efficiency of plasmid DNA into the nucleus (Prabha et al., 2016). Par-
to control the chemical composition and the overall properties of ge- ticle size of gene therapy vectors is also an important factor that im-
latin materials. One of the most significant properties of gelatin is its pacts the access and passage of the vector to the targeting site (Jiang
versatile structure due to its protein nature that can be modified easily et al., 2007), and it also impacts the stability of colloidal particles in
by modifying its functional groups with different cross-linkers and solution. For efficient endocytosis and gene delivery, the particle size of
targeting-ligands. This property could be very beneficial to improving the complex should be below 200 nm and compact (Liu et al., 2007).
and developing potential gene delivery systems, with minimal toxic The particle size depends on many factors, including nucleic acid
effects on host cells (Busch et al., 2003; Wang et al., 2012). Forming concentration, and the order of addition of vector components during
gelatin nanoparticles for the delivery of nucleic acid to cells and the preparation. The particle sizes of polyplexes are closely related to the
nucleus is important for several reasons. Firstly, nanoparticles are taken overall surface charge of the particles. This review paper will focus on
up more easily and efficiently by cells than large particles (Panyam and several modifications that can be applied to gelatin nanoparticulate,
Labhasetwar, 2003). Secondly, nanoparticles have an ability to escape and their effects on gene delivery.
rapidly from the endosome; consequently, they are protected against
degradation (Labhasetwar, 2005). Additionally, the nano-size range of
4.1. Cationic gelatin nanoparticles
these delivery systems allows them to be injected directly into the
systemic circulation without the risk of blocking blood vessels. More-
Gelatin is a polyelectrolyte with a low-charge density that is ap-
over, nanoparticles have been proved to improve the transfection
preciably changed depending on the solution’s pH. As a result, the

A* B

Biodegradable

Low cost of production

Physiological tolerance

Easily modified

Biocompatible

Low antigenicity

Fig. 2. General chemical structure of gelatin (A), and features of gelatin (B).*Reproduced with permission from (Sahoo et al., 2015).

226
O. Madkhali et al. International Journal of Pharmaceutics 554 (2019) 224–234

Fig. 3. Chemical structures of molecules used to cationize gelatin.

cationization of gelatin is a significant factor in enhancing the ability of lower colloidal stability and electric binding efficiency (Chou et al.,
the polymer to interact with negatively charged cellular membranes or 2018; Kuo et al., 2011). PEI is a highly branched polymer with about
DNA, thereby obtaining an effective gene delivery vector (Zwiorek 25% primary amine groups, 50% secondary amine groups, and 25%
et al., 2004). Cationized gelatin is mainly prepared by introducing tertiary amine groups. It plays a crucial role in endosomal escape via
amine residues to the carboxyl groups of gelatin using poly- ‘‘proton-sponge mechanism’’ when grafted on gelatin. The proton-
ethyleneimine (Mimi et al., 2012), cholamine (Geh et al., 2016; sponge hypothesis presumes the ability of PEI (due to its secondary
Zwiorek et al., 2008), ethylenediamine (EDA) (Ishikawa et al., 2012; Xu amine groups) to buffer the pH, causing the ATPase enzyme in the
et al., 2014; Xu et al., 2008), putrescine, spermidine (Kushibiki et al., endosomal membrane to influx more protons to reach the desired pH,
2006b,c) or spermine (Konat Zorzi et al., 2011) (Fig. 3). Ethylenedia- resulting in subsequent influx of counter chloride ions which causes
mine cationized gelatin can condense plasmid DNA, expressing insulin- osmotic swelling and rupture of the endosomal membrane (Mimi et al.,
like growth factor (IGF)-1. It has shown a five-fold increase of IGF in 2012; Morille et al., 2008). In an interesting study conducted by Zorzi
adult articular chondrocytes compared with non-cationized gelatin. In et al (Zorzi et al., 2015), cationized gelatin showed lesser cytotoxicity in
addition, chondrocytes treated with pIGF using cationized gelatin were comparison with cationized atelocollagen and albumin. Moreover,
able to maintain stable IGF-1 overexpression when later grown in a among different amines used for cationization, gelatin cationized with
collagen (type II)-glycosaminoglycan (CG) scaffold for up two weeks spermine proved to be the optimum nanoparticulate system for pro-
and exhibited enhanced biosynthesis (Xu et al., 2008). tecting both pDNA and siRNA under the conditions endeavored. Simi-
Positive charges possessed by gelatin nanoparticles do not only in- larly, Kushibiki et al. (2006a) investigate the in vitro transfection effi-
teract electrostatically with the anionic terminal of phospholipid, pro- ciency of plasmid DNA for mouse fibroblasts using cationized gelatin
teins and glycans on the plasma membrane, but also promote nano- prepared from different types of amine compounds namely; ethylene-
particle association with the cells inducing cell uptake by cellular diamine, putrescine, spermidine and spermine. The highest level of
endocytic mechanisms (Bannunah et al., 2014). Among multiple en- gene expression was observed with spermine grafted gelatin owing to
docytosis pathways, clathrin-mediated (CME) (Fröhlich, 2012; Yameen two main reasons; a) the intrinsic ability of this natural polyamine to
et al., 2014) and caveolae mediated (Morille et al., 2008) endocytosis condense and pack DNA into small particles and b) it possesses the
are the major route for cellular uptake for positively charged poly- highest buffering effect among all other cationized gelatins used
plexes. Moreover, recent studies postulate that cellular uptake of posi- (Kushibiki et al., 2006c). Hence, it is currently used in commercial
tively charged NPs uptake is related to energy dependent processes like transfection agents like Lipofectamine®, which contains a lipid modified
proteins dynamin and F-actin (Dausend et al., 2008). However, highly with spermine (Hawley-Nelson et al., 2008). These results indicate that
positively-charged NPs could cause perforations in the cellular lipid cationic gelatin transfection efficiency and cytotoxicity depends on the
bilayer to enter the cells by-passing endocytic pathways (Sadat et al., type of gelatin modification and the molecular weight of the grafted
2016). Throughout gelatin cationization, a considerable challenge molecule.
aroused to balance between the high positive zeta potential required for Various methods have been used to prepare cationic gelatin nano-
both cell penetration and nanoparticles stability, and the low zeta po- particles. Those most commonly used are discussed in the next section.
tential required to avoid opsonization and cytotoxicity where cationic
NPs might interact with proteoglycans on the cell surface, triggering
5. Methods of preparations of cationic gelatin
cellular necrosis and apoptosis (Vega-Villa et al., 2008). For instance,
Chou et al. (Chou et al., 2018) revealed that gelatin nanoparticles with
5.1. Preparation of cationic gelatin using amination method
surface modification of 1.8-kD PEI showed the optimum protein de-
livery to cancer cells. However, 10-kD PEI grafted gelatin was more
Cationic gelatin is prepared by introducing a cationic agent directly
cytotoxic while PEI with molecular weight less than 1.8-kD unveiled
to the carboxylic group of gelatin (Fig. 4). Briefly, gelatin Type A (IEP

227
O. Madkhali et al. International Journal of Pharmaceutics 554 (2019) 224–234

Fig. 4. Schematic diagram of the preparation of cationic gelatin using EDA or spermine and EDC. Adapted with permission from (Samal et al., 2012).

7–9) is completely dissolved in 0.1 M phosphate buffer saline (PBS) robust in the preparation of gelatin nanoparticles (Geh et al., 2016).
containing potassium dihydrogen phosphate and disodium hydrogen
phosphate. The cationic agent is added, and the pH is subsequently 5.4. Preparation of cationic gelatin nanoparticles using ionic gelation
adjusted to 5. To this solution, 1-ethyl-3- (3-dimethylaminopropyl) technique
carbodiimide (EDC) is added, and the final volume is made up with
PBS. The solution is then agitated at 37 °C for 18 h and subsequently Zorzi et al. (Parraga et al., 2009; Zorzi et al., 2011) prepared ca-
dialyzed for two days with 16 changes of water. Following the dialysis tionic gelatin nanoparticles using the ionic gelation technique for an
process, the solution is freeze-dried for 4 to 7 days to afford cationized ocular surface (Fig. 6). In this technique, Zorzi and colleagues used
gelatin (Fukuyama et al., 2006). In this method, no nanoparticles are chondroitin sulfate (CS) and dextran sulfate (DS) to make cationic ge-
formed. There are several methods for preparing cationic gelatin na- latin. Briefly, gelatin was first cationized using spermine hydrochloride
noparticles, which will be described below. (SPM). The cationized gelatin was then added to triphosphate (TPP)
containing the plasmid and CS or DS. The advantage of this method
5.2. Preparation of cationic gelatin nanoparticles using two-step desolvation includes the fact that no organic solvents such as acetone and methanol
method are used, in comparison with one-step or two-step desolvation (Zorzi
et al., 2015). In addition, glutaraldehyde, which is reported to be toxic,
Two-step desolvation approach was adopted to limit the hetero- is not added to this method (Gough et al., 2002; Maitra et al., 2006).
geneity of gelatin structure and produce gelatin nanoparticles with a Table 1 summarized different methods of preparing cationic gelatin
reduced tendency for aggregation. Gelatin nanoparticles were prepared using different cationic agents and the effect of these methods on the
using a two-step desolvation method previously described by Coaster transfection efficiency and cell viability.
et al. (Coester et al., 2000). In this method, after the first desolvation
step, the low molecular gelatin fractions presented in the supernatant 6. PEGylated gelatin nanoparticles
were removed by decanting, and the high molecular fractions presented
in the sediment were re-desolved (Saxena et al., 2005). In brief, gelatin GNPs are mainly engulfed by the cells of the reticuloendothelial
type A is dissolved in distilled water under constant heating (45–50 °C). system (RES) upon systemic administration. This leads to weak trans-
Desolvating agents such as acetone or ethanol are added as to pre- fection and gene expression. However, coating the GNPs with poly
cipitate the high molecular weight (HMW) of gelatin. After the super- (ethylene glycol) (PEG) generates a dense hydrophilic shell of long
natant is discarded, the HMW gelatin is re-dissolved in distilled water chains that conserve the core of GNPs from non-specific hydrophobic
and stir at 600 RPM under a constant heating process. Subsequently, the interaction with serum protein. Consequently, this significantly reduces
pH is adjusted to 2.5 using HCl. Following this, acetone or ethanol is the effect of RES (Otsuka et al., 2003). Another advantage of PEGyla-
added drop-wise to form nanoparticles. Glutaraldehyde (GA) 25% (v/v) tion is that it may increase the hydrodynamic size of the particles (more
is added as a cross-linking agent, which is important to provide than 30 nm) which leads to a decrease in their clearance from the
homogenous, stable, and spherical shape nanoparticles, and to enhance kidney, in that renal filtration is dependent on the molecular mass and
circulation times in vivo (compared to uncrossed-linked nanoparticles volume. These advantages result in an increase in the circulation half-
(Elzoghby et al., 2012; Kommareddy et al., 2005a)), and stirred at life of the particles in vivo (Crawford, 2002; Kommareddy et al., 2005b).
600 rpm at room temperature overnight. The following day, the nano- PEG capacity for repelling proteins and avoiding macrophages depends
particles are purified using centrifugation or dialysis membrane prior to on different considerations such as PEG MW, density, the conformation
freeze drying. Following the freeze drying process, the gelatin is ca- and chain flexibility (Vonarbourg et al., 2006). According to multiple
tionized by introducing amino residues to the carboxyl group of gelatin studies, the optimum MW required for decreasing protein adsorption in
nanoparticles in a manner analogous to the cationization of molecular vitro is 1.5–3.5 KDa. However, regarding the macrophage uptake, very
gelatin. The gelatin nanoparticles are dispersed in highly purified long chains of approximately 20 KDa are essential (Mosqueira et al.,
water, and then the pH is adjusted between pH 4.5 and pH 5. To this 1999). In addition, the existence of PEGylation on the surface of the
dispersion 50 mg of amine residue (i.e. spermine, cholamine, etc.) is GNPs is beneficial in terms of the protection of the particles from di-
added and incubated for five minutes, the same amount of EDC is then gestion by proteinases (Xu et al., 2012).
added to the solution and the reaction is left for 1 h in the dark. The In vitro studies showed that PEGylated gelatin NPs were internalized
resulting cationized gelatin nanoparticles are purified using cen- by nonspecific endocytosis pathway where their cargo was delivered to
trifugation or dialysis membrane for two days prior to lyophilization. the peri-nuclear region within 12 h (Kaul and Amiji, 2004). Although
studies claimed that shielding of gelatin cationic groups via PEGylation
5.3. Preparation of cationic gelatin using one-step desolvation method decreases both cytotoxicity and efficacy in NPs, yet Hoskins et al.
proved that charge shielding via PEGylation had a small impact on
The one-step desolvation method is similar to the two-step method cellular uptake while greatly decreasing cytotoxicity, whereas obvious
described above. However, the first step which precipitates the high reduction in membrane damage, lipid peroxidation, and oxidative stress
molecular weight is omitted (Fig. 5). This method has proved to be were reported in neuroblastoma cells (Hoskins et al., 2012).

228
O. Madkhali et al. International Journal of Pharmaceutics 554 (2019) 224–234

Fig. 5. Schematic illustration of the preparation of cationic gelatin nanoparticles using the one-step desolvation method.

Fig. 6. Schematic illustration of the preparation of cationic gelatin nanoparticles using the ionic gelatin technique.

Adding PEGylation to non-condensing type B GNPs has resulted in GNPs and PEGylated 125I-labeled GNPs after I.V. administration
an excellent system for effective distribution in solid tumors because of through the tail vein in LLC-bearing mice. They found that PEGylated
the presence of hyperpermeable angiogenic blood vessels in such tu- GNPs have longer circulating properties in the blood and remained in
mors and the enhanced permeability and retention (EPR) effect (Kaul the tumor for up to 24 h after administration. In another study thiolated
and Amiji, 2004, 2005). According to Amiji and Kaul (Kaul and Amiji, PEGylated GNPs exhibited prolonged circulation and enhanced tumor
2004), PEGylated GNPs favorably targeted tumor mass in Lewis lung extravasation in vivo in an orthotopic human breast adenocarcinoma
carcinoma (LLC) bearing female mice, and about 4–5% of the injected xenograft model (Kommareddy and Amiji, 2007a). In comparison with
dose remained in the tumor for approximately twelve hours after ad- the non-PEGylated GNPs, the PEGylated nanoparticles showed longer
ministration. Amiji and Kaul also stated that reporter pDNA encoding circulation with plasma and tumor half-lives of 15.3 and 37.8 h re-
for β-galactosidase (pCMV-β) was effectively encapsulated in PEGy- spectively. Generally, the advantages of the existence of PEG in com-
lated GNPs and showed significant expression in the tumor of LLC with bination with GNPs are summarized as follows: increase in the circu-
61% transfection efficiency (Kaul and Amiji, 2005). lation time in the plasma and tumor mass, stabilization of therapeutic
Kushibiki et al. (2004) studied the long-circulation property of PE- cargo during transportation, prevention of RES elimination, and the
Gylated gelatin using 125I-labeled gelatin. They compared unmodified provision of potential for the conjugation of targeting moieties (Kaul

229
O. Madkhali et al. International Journal of Pharmaceutics 554 (2019) 224–234

Table 1
Preparation of cationic gelatin for gene delivery using different cationic agents.
Method Gelatin Type Cationic agent NP* Gene used In vitro/ Cell type Outcomes Ref.
vivo

Two-step Type A Cholamine Yes pCMV-luc In vitro B16F10 Moderate transfection with high cell Zwiorek et al. (2004)
Type A Polyethyleneimine siRNA _ _ viability Zillies and Coester (2005)
pCMV-luc NIH 3 T3 High transfection with high cell Kuo et al. (2011)
In vitro viability
One step Type A and Cholamine Yes _ _ _ _ Geh et al. (2016), Zwiorek
Type B et al. (2008)
Ionic gelation Type A Spermine Yes pMUC5AC In vitro HCE Spermine showed high transfection Konat Zorzi et al. (2011)
Type B Spermine & pDNA & siRNA In vitro IOBA-NHC and gene silencing with adequate cell Zorzi et al. (2015)
Ethylenediamine HCE viability
IOBA-NHC
Aminization Type A Etylenediamine No pCMV-luc In vitro RGM-1 Spermine showed the highest Hosseinkhani et al. (2002a),
Spermine pGL3-Luc In vitro L929 transfection with low cell viability Hosseinkhani et al. (2002b)
Spermidine pGL3-Luc In vivo ddY mice† Spermine showed the highest Kushibiki et al. (2006b, c)
Putrescine transfection with low cell viability Kushibiki and Tabata (2005)

* Nanoparticles formed.

6–7 weeks old mice.

and Amiji, 2004, 2005). in anhydrous DMSO at room temperature. Methyl ether NHS-PEG with
(4.0 × 10−5 mol) previously dissolved DMSO is slowly added to the
6.1. Methods of preparations of PEGylated gelatin gelatin solution and left stirring at room temperature for 3 h, dialyzed
against deionized water, and then lyophilized.
Preparation of PEGylated gelatin is based on using amine reactive
PEG derivatives to form covalent bond with the free amino groups on 7. EGFR-targeted gelatin nanoparticles
gelatin (Fig. 8). Kyung and Youngro were able to synthesize carboxy-
lated derivative of PEG which was further activated using dicyclohex- One of the greatest challenges for gene delivery is the area of tar-
ylcarbodiimide (DCC) to couple the PEG to the amino group of gelatin geting. A delivery vector is required to distinguish host cells, evade
with a PEGyltion degree (mol/mol%) of 48% (Kim and Byun, 1999). nonspecific binding, and resist degradation in the systemic circulation.
According to another method described by Kaul and Amiji (Kaul and After reaching the target cells, the delivery vector should cross the cell
Amiji, 2002), PEG-epoxide is used as activated derivative to graft PEG membrane, facilitate the escape of the vector from the endosome, re-
as illustrated in Fig. 7 where higher PEGyltion degree (mol/mol%) were lease nucleic acid from the complex, which can then enter the nucleus
attained. Briefly, PEG-epoxide is dissolved in alkaline borate buffer (pH to express the required protein (Xu et al., 2013). Although tumor tar-
8.5), gelatin type B is then added, and the reaction is kept under stirring geting using PEG surface modified nanoparticles accomplishes some
for 14 h at 40 °C. The product obtained is precipitated with excess preferential accumulation in tumor cells and allows for intracellular
acetone to remove any unreacted PEG-epoxide, dialyzed against dis- delivery, some types of cancer do not have adequate vasculature, or the
tilled water, and then lyophilized. nanoparticles may not be able to penetrate deeply into the tumor mass
An alternative method relies on reacting gelatin with amine reactive (Xu et al., 2012). The mutation of the epidermal the growth factor re-
molecules like methoxy PEG-succinimidyl glutarate (Kommareddy and ceptor (EGFR) has been shown to be associated with poor prognosis in
Amiji, 2007b) and methoxy PEG-succinimidyl succinate (Kushibiki several types of cancers including ovarian cancer (Ciardiello and
et al., 2004; Kushibiki and Tabata, 2005) with a degree of PEGylation of Tortora, 2001). Between 33% and 75% of EGFR has reported to be
90% and 30–100% respectively. Briefly, gelatin (1.0 µmol) is dissolved overexpressed in ovarian cancer, and has been found in both the growth
and the progression of the disease (Sewell et al., 2002).
EGFR is a member of the ErbB/her family of ligand activated re-
ceptor tyrosine kinases (RTKs). It has been recognized as a molecular
target. EGFR consists of an extracellular ligand-binding domain like any
other receptor of tyrosine kinases which are involved in interactions
between receptors within a membrane, and a cytoplasmic domain with
tyrosine kinase activity (Schlessinger, 2002). Accordingly, receptor
mediated endocytosis is the main cellular uptake mechanism for gelatin
NPs decorated with EGFR (Mickler et al., 2012).
To conjugate EGFR targeting peptides to gelatin, either the gelatin
amino groups are converted to thiol groups via reacting with 2-imi-
nothiolane hydrochloride at room temperature for 15 h, (Kommareddy
and Amiji, 2005; Tseng et al., 2008) then left over night to react with
Sulfo-MBS activated EGFR at 4 °C (Tseng et al., 2008) or gelatin is firstly
modified with PEG maleimide which is then conjugated to the peptide
through a spacer containing a sulfhydryl group in its terminal cysteine
residue (Fig. 8) (Magadala and Amiji, 2008).
Consequently, the conjugation of gelatin with targeting EGFR pep-
tide has been shown to improve the transfection efficiency in several
types of cancer cells. EGFR-targeted GNPs carrying plasmid DNA en-
Fig. 7. Schematic illustration of the reaction between PEG-epoxide and gelatin coding for EGFp-N1 obtained the highest transfection efficiency in
nanoparticles. Reproduced from (Elzoghby, 2013). Panc-1 pancreatic adenocarcinoma cells in comparison with other

230
O. Madkhali et al. International Journal of Pharmaceutics 554 (2019) 224–234

Fig. 8. Schematic illustration of grafting different ligands to gelatin nanoparticles. Gelatin nanoparticles can be chemically modified by adding -thiol groups, cationic
agents, alendronate, PEG, and conjugating targeting EGFR peptide to PEGylated gelatin nanoparticles.

controls, particularly 48 h after transfection (Magadala and Amiji, quaternary protein structure of gelatin (Bacalocostantis et al., 2013). In
2008). The intravenous injection of EGFR-targeted GNPs to mice addition, disulfide bonds can stabilize the nanoparticles during sys-
bearing Panc-1 pancreatic adenocarcinoma showed almost a double temic circulation and release the encapsulated payload when they are
efficiency with regard to targeting, in comparison with PEG-GNPs and broken inside the cell (Kommareddy and Amiji, 2005). Groups of thiols
unmodified GNPs. Additionally, it accumulated and was sustained for a are easily and rapidly crosslinked; thus, they can be used for the
longer period in the tumor mass (Xu et al., 2013). Another study by Xu synthesis of polymeric delivery vectors (Bacalocostantis et al., 2013).
and Amiji (Xu and Amiji, 2012) used EGFR-targeted thiolated gelatin Glutathione (GSH) is a dipeptide, used as an antioxidant to prevent
nanoparticles to deliver plasmid DNA into Panc-1 pancreatic adeno- damage caused by an oxygen species. GSH and peroxide exist in high
carcinoma cells. The EGFR improved the targeting, and the thiol group concentration inside the cells to a greater extent than they do outside
improved the stability of GNPs. The results showed that EGFR-targeted (100-fold higher), and their concentration is much higher in the cyto-
thiolated GNPs had a small nanoparticle size (150–200 nm) with high plasm of tumor cells. As a result, Kommareddy and Amiji (Kommareddy
GFP expression, even higher than the positive control lipofectin-com- and Amiji, 2005) introduced a thiol (SH) group into gelatin using a 2-
plexed DNA, and they obtained high cell viability as well (Xu and Amiji, iminothilane reagent and prepared the nanoparticles by desolvation
2012). Using targeting-ligands with GNPs facilitates the delivery system using ethanol under adjusted and controlled pH and temperature con-
‘s recognition of the host cell; as a result, transfection is improved, and ditions. The plasmid DNA was then incorporated into the thiolated
cytotoxicity is reduced, thereby achieving the optimal goal for gene gelatin nanoparticles. The thiolated GNP encapsulated DNA showed
therapy delivery systems. high transfection efficiency in NIH-3T3 murine fibroblast cells in con-
trast to unmodified gelatin and lipofectin®-complexed DNA
(Kommareddy and Amiji, 2005). Six hours after transfection, the ex-
8. Thiolated gelatin nanoparticles pression of the green fluorescent protein was observed. These results
can be interpreted as the disulfide bonds increasing the stability of the
The thiol group (-SH) has been considered as a potential addition to nanoparticles and indicated that thiolated GNPs have the rapid release
GNPs due to its ability to respond to environmental changes, either of their contents into a highly reducing environment inside the cell
inside or outside the cell. A thiol group is similar to alcohol in its where the high concentration of GSH (Kommareddy and Amiji, 2005,
chemical structure but differs in terms of its chemical properties; thiols 2007b). Furthermore, the same group evaluated three modifications of
are more nucleophilic, more acidic, and more readily oxidized than gelatin: PEG-GNPs, thiolated-GNPs, and PEG-modified thiolated GNPs
alcohol (Senning, 1997). Adding thiol groups to gelatin leads to the (Fig. 8) in NIH-3 T3 to deliver plasmid DNA. Of the three formulations
formation of disulfide bonds (S-S) in an oxidation reaction within the tested, PEG-thiolated GNPs indicated the highest GFP expression to
polymer. This can be beneficial in strengthening the tertiary and

231
O. Madkhali et al. International Journal of Pharmaceutics 554 (2019) 224–234

even a greater degree than the positive control lipofectin-complexed chains of the gelatin matrix molecule which facilitate further mod-
DNA (Kommareddy and Amiji, 2007b). Generally, both PEG-GNPs and ifications. This may be useful for coupling of ligands for improving the
PEG-modified thiolated GNPs demonstrated longer circulation in the target diseased tissues and to enhance particular cellular uptake or af-
blood and higher accumulation in the tumor cells, in contrast with fect intracellular distribution. Gelatin nanoparticles are promising non-
unmodified GNPs (Xu et al., 2012). viral vectors for gene delivery applications. The modifications im-
A new tumor-targeted siRNA delivery system using polymerized proved the delivery system and led to more effective transfection effi-
siRNA (poly-siRNA) and thiol-modified gelatin nanoparticles was de- ciency and cell viability. Depending on the final outcome/objective of
veloped by Lee et al. (Lee et al., 2013). The poly-siRNA was prepared by the gelatin nanoparticle, various manufacturing methods may be uti-
self-polymerization of the thiol group and was encapsulated in the self- lized such as cationic, PEGylated, thiolated, EGFR-targeted, and alen-
assembled thiolated-GNPs using chemical cross-linking. The results dronate gelatin nanoparticles. One of the biggest challenges for using
showed that the siRNA was protected from enzymatic degradation; the gelatin as a gene delivery system is scaling up, particularly for gelatin
siRNA molecules were released effectively in a reductive condition. nanoparticles. With scaling up, gelatin loses its properties such as bio-
Also, poly-siRNA-thiolated –GNPs demonstrated an excellent accumu- compatibility and biodegradability and results in more aggregates and
lation in tumor cells, induced effective target gene-silencing in tumors less homogenous and stable nanoparticles, which is more noticeable
after intravenous injection, and demonstrated high cell viability, closer with modified gelatin. This problem can be overcome by using cross-
to 100% compared with lipofectamine and non-thiolated siRNA-GNPs linkers such as glutaraldehyde, which should be removed after pre-
(Lee et al., 2013). It is apparent that the disulfide bonds formed by the paring nanoparticles due to the risk of toxicity.
(thiol) group could play a significant role in the stability of nano-
particles, thereby resulting in effective gene expression and high cell Acknowledgements
viability.
Funding from the School of Pharmacy, University of Waterloo,
9. Alendronate gelatin nanoparticles Canada and the Faculty of Pharmacy, Jazan University, Kingdom of
Saudi Arabia is gratefully acknowledged. SDW is funded by the Natural
Bisphosphonates, like alendronate, are characterized by their high Sciences and Engineering Research Council of Canada (RGPIN-2016-
affinity to hydroxyapatite. Moreover, they maintain their binding cap- 04009).
ability even after conjugation to macromolecules (Low and Kopecek,
2012). For targeted gene delivery, Alendronate (ALN) modified gelatin References
biopolymers, which could be promising for bone targeting, were syn-
thesized and characterized. An innovative four-component system Bacalocostantis, I., Mane, V.P., Goodley, A.S., Bentley, W.E., Muro, S., Kofinas, P., 2013.
composed of alendronate sodium trihydrate (ALN), gelatin, gemini Investigating polymer thiolation in gene delivery. J. Biomater. Sci. Polym. Ed. 24,
912–926.
16–3-16 surfactant and DNA was prepared. For ALN conjugation, ge- Bannunah, A.M., Vllasaliu, D., Lord, J., Stolnik, S., 2014. Mechanisms of nanoparticle
latin is dissolved in a suitable volume of phosphate buffer (pH 4.5 and internalization and transport across an intestinal epithelial cell model: effect of size
heated to 50 °C) and then ALN is added at the required molar ratios. and surface charge. Mol. Pharm. 11, 4363–4373.
Busch, S., Schwarz, U., Kniep, R., 2003. Chemical and structural investigations of bio-
EDC is added as a last step to the gelatin-ALN solution to limit self- mimetically grown fluorapatite–gelatin composite aggregates. Adv. Funct. Mater. 13,
cross-linking of gelatin molecules (Fig. 8). ALN-gelatin biopolymers 189–198.
prepared at various alendronate/gelatin ratios were utilized to prepare Cardoso, A.M., Morais, C.M., Silva, S.G., Marques, E.F., de Lima, M.C.P., Jurado, M.A.S.,
2014. Bis-quaternary gemini surfactants as components of nonviral gene delivery
nanoparticles and were optimized in combination with DNA and gemini systems: a comprehensive study from physicochemical properties to membrane in-
surfactant for transfecting both HEK-293 and MG-63 cell lines (Mekhail teractions. Int. J. Pharm. 474, 57–69.
et al., 2016). The study revealed that not only did the degree of ALN Chou, M.-J., Yu, H.-Y., Hsia, J.-C., Chen, Y.-H., Hung, T.-T., Chao, H.-M., Chern, E.,
Huang, Y.-Y., 2018. Highly efficient intracellular protein delivery by cationic poly-
substitution on gelatin significantly affected the transfection efficiency
ethyleneimine-modified gelatin nanoparticles. Materials 11, 301.
of the gelatin based nanocomplexes but also the type of grafted gelatin. Ciardiello, F., Tortora, G., 2001. A novel approach in the treatment of cancer: targeting
In particular, nanocomplexes formulated using the Type A functiona- the epidermal growth factor receptor. Clin. Cancer Res. 7, 2958–2970.
lized gelatin prepared at a 1:4 gelatin: ALN ratio had a higher zeta Coester, C., Langer, K., Von Briesen, H., Kreuter, J., 2000. Gelatin nanoparticles by two
step desolvation a new preparation method, surface modifications and cell uptake. J.
potential than those obtained from Type B functionalized gelatin and Microencapsul. 17, 187–193.
thus a higher bone selective transfection efficiency (comparing MG-63 Crawford, J., 2002. Clinical uses of PEGylated pharmaceuticals in oncology. Cancer Treat.
to HEK-293), which was approximately three times that of Lipofecta- Rev. 28, 7–11.
Dausend, J., Musyanovych, A., Dass, M., Walther, P., Schrezenmeier, H., Landfester, K.,
mine 2000® and 1.5 times the transfection efficiency of the gemini/ Mailänder, V., 2008. Uptake mechanism of oppositely charged fluorescent nano-
DNA complexes (Mekhail et al., 2016). The presence of ALN as a tar- particles in HeLa cells. Macromol. Biosci. 8, 1135–1143.
geting moiety facilitated receptor mediated endocytosis for cellular Elzoghby, A.O., 2013. Gelatin-based nanoparticles as drug and gene delivery systems:
reviewing three decades of research. J. Control. Release 172, 1075–1091.
uptake. Those results revealed that ALN-modified gelatin established a Elzoghby, A.O., Samy, W.M., Elgindy, N.A., 2012. Protein-based nanocarriers as pro-
platform for both drug delivery and a promising vector for the for- mising drug and gene delivery systems. J. Controlled Release : Off. J. Controlled
mulation of targeted cationic nanocomplexes for enhanced gene de- Release Soc. 161, 38–49.
Fröhlich, E., 2012. The role of surface charge in cellular uptake and cytotoxicity of
livery to bone tissues.
medical nanoparticles. Int. J. Nanomed. 7, 5577.
Fukuyama, N., Onuma, T., Jujo, S., Tamai, Y., Suzuki, T., Myojin, K., Tabata, Y., Ishihara,
10. Conclusion Y., Takano, J., Mori, H., 2006. Efficient preparation of cationized gelatin for gene
transduction. Tokai J. Exp. Clin. Med. 31, 49–52.
Geh, K.J., Hubert, M., Winter, G., 2016. Optimisation of one-step desolvation and scale-up
There are numerous advantages of using gelatin as polymer for of gelatine nanoparticle production. J. Microencapsul. 1–10.
fabrication of nanoparticles. Gelatin possesses low cellular toxicity be- Gough, J.E., Scotchford, C.A., Downes, S., 2002. Cytotoxicity of glutaraldehyde cross-
cause it is derived from animal protein and also producing very low- linked collagen/poly (vinyl alcohol) films is by the mechanism of apoptosis. J.
Biomed. Mater. Res. Part A 61, 121–130.
level allergic reaction. Gelatin can be easily polymerized to form na- Griffith, F., 1928. The significance of pneumococcal types. Epidemiol. Infect. 27,
noparticles which are relatively stable and reproducible. Gelatin also 113–159.
facilitates for future up scaling. Due to low cost of gelatin and its bio- Hawley‐Nelson, P., Ciccarone, V., Moore, M.L., 2008. Transfection of cultured eukaryotic
cells using cationic lipid reagents. Curr. Protoc. Mol. Biol. 81 9.4. 1-9.4. 17.
degradable nature gelatin gain popularity among researcher specially Hoskins, C., Wang, L., Cheng, W.P., Cuschieri, A., 2012. Dilemmas in the reliable esti-
for commercial scaling. The major issues related with nanomaterials are mation of the in-vitro cell viability in magnetic nanoparticle engineering: which tests
about their termination from the body after targeting to the specific and what protocols? Nanoscale Res. Lett. 7, 77.
Hosseinkhani, H., Abedini, F., Ou, K.L., Domb, A.J., 2015. Polymers in gene therapy
organ, tissue or cell. Another advantage of gelatin is amino acid side-

232
O. Madkhali et al. International Journal of Pharmaceutics 554 (2019) 224–234

technology. Polym. Adv. Technol. 26, 198–211. biodegradable amphiphilic PCL-Pluronic (L35)-PCL block copolymers. Colloids Surf.,
Hosseinkhani, H., Aoyama, T., Ogawa, O., Tabata, Y., 2002a. Ultrasound enhancement of A 302, 430–438.
in vitro transfection of plasmid DNA by a cationized gelatin. J. Drug Target. 10, Low, S.A., Kopecek, J., 2012. Targeting polymer therapeutics to bone. Adv. Drug Deliv.
193–204. Rev. 64, 1189–1204.
Hosseinkhani, H., Aoyama, T., Yamamoto, S., Ogawa, O., Tabata, Y., 2002b. In vitro Lundstrom, K., Boulikas, T., 2003. Viral and non-viral vectors in gene therapy: technology
transfection of plasmid DNA by amine derivatives of gelatin accompanied with ul- development and clinical trials. Technol. Cancer Res. Treat. 2, 471–485.
trasound irradiation. Pharm. Res. 19, 1471–1479. Magadala, P., Amiji, M., 2008. Epidermal growth factor receptor-targeted gelatin-based
Ishikawa, H., Nakamura, Y., Jo, J.-I., Tabata, Y., 2012. Gelatin nanospheres incorporating engineered nanocarriers for DNA delivery and transfection in human pancreatic
siRNA for controlled intracellular release. Biomaterials 33, 9097–9104. cancer cells. Am. Assoc. Pharm. Sci. J. 10, 565–576.
Jackson, D.A., Juranek, S., Lipps, H.J., 2006. Designing nonviral vectors for efficient gene Maitra, A., De, T.K., Mitra, S., 2006. Hydrogel nanoparticles: applications in drug de-
transfer and long-term gene expression. Mol. Ther. 14, 613–626. livery. second ed. pp. 2821–2837.
Jiang, H.-L., Kim, Y.-K., Arote, R., Nah, J.-W., Cho, M.-H., Choi, Y.-J., Akaike, T., Cho, C.- Mali, S., 2013. Delivery systems for gene therapy. Indian J. Human Genet. 19, 3.
S., 2007. Chitosan-graft-polyethylenimine as a gene carrier. J. Control. Release 117, Mekhail, G.M., Kamel, A.O., Awad, G.A., Mortada, N.D., Rodrigo, R.L., Spagnuolo, P.A.,
273–280. Wettig, S.D., 2016. Synthesis and evaluation of alendronate-modified gelatin biopo-
Jin, L., Zeng, X., Liu, M., Deng, Y., He, N., 2014. Current progress in gene delivery lymer as a novel osteotropic nanocarrier for gene therapy. Nanomed. Nanotechnol.
technology based on chemical methods and nano-carriers. Theranostics 4, 240. Biol. Med. 11, 2251–2273.
Jones, C.H., Chen, C.-K., Ravikrishnan, A., Rane, S., Pfeifer, B.A., 2013. Overcoming Mickler, F.M., Möckl, L., Ruthardt, N., Ogris, M., Wagner, E., Bräuchle, C., 2012. Tuning
nonviral gene delivery barriers: perspective and future. Mol. Pharm. 10, 4082–4098. nanoparticle uptake: live-cell imaging reveals two distinct endocytosis mechanisms
Karimi, M., Solati, N., Ghasemi, A., Estiar, M.A., Hashemkhani, M., Kiani, P., Mohamed, mediated by natural and artificial EGFR targeting ligand. Nano Lett. 12, 3417–3423.
E., Saeidi, A., Taheri, M., Avci, P., 2015. Carbon nanotubes part II: a remarkable Mimi, H., Ho, K.M., Siu, Y.S., Wu, A., Li, P., 2012. Polyethyleneimine-based core-shell
carrier for drug and gene delivery. Exp. Opin. Drug Delivery 12, 1089–1105. nanogels: a promising siRNA carrier for argininosuccinate synthetase mRNA knock-
Kaul, G., Amiji, M., 2002. Long-circulating poly (ethylene glycol)-modified gelatin na- down in HeLa cells. J. Control. Release 158, 123–130.
noparticles for intracellular delivery. Pharm. Res. 19, 1061–1067. Morille, M., Passirani, C., Vonarbourg, A., Clavreul, A., Benoit, J.-P., 2008. Progress in
Kaul, G., Amiji, M., 2004. Biodistribution and targeting potential of poly(ethylene glycol)- developing cationic vectors for non-viral systemic gene therapy against cancer.
modified gelatin nanoparticles in subcutaneous murine tumor model. J. Drug Target. Biomaterials 29, 3477–3496.
12, 585–591. Mosqueira, V.C.F., Legrand, P., Gref, R., Heurtault, B., Appel, M., Barratt, G., 1999.
Kaul, G., Amiji, M., 2005. Tumor-targeted gene delivery using poly(ethylene glycol)- Interactions between a macrophage cell line (J774A1) and surface-modified poly (D,
modified gelatin nanoparticles: in vitro and in vivo studies. Pharm. Res. 22, 951–961. L-lactide) nanocapsules bearing poly (ethylene glycol). J. Drug Target. 7, 65–78.
Keeler, A.M., ElMallah, M.K., Flotte, T.R., 2017. Gene therapy 2017: progress and future Nayerossadat, N., Maedeh, T., Ali, P.A., 2012. Viral and nonviral delivery systems for
directions. Clin. Trans. Sci. gene delivery. Adv. Biomed. Res. 1.
Kim, K.J., Byun, Y., 1999. Preparation and characterizations of self-assembled PEGylated Ninan, G., Jose, J., Abubacker, Z., 2011. Preparation and characterization of gelatin ex-
gelatin nanoparticles. Biotechnol. Bioprocess Eng. 4, 210–214. tracted from the skins of rohu (Labeo rohita) and common carp (Cyprinus carpio). J.
Kommareddy, S., Amiji, M., 2005. Preparation and evaluation of thiol-modified gelatin Food Process. Preserv. 35, 143–162.
nanoparticles for intracellular DNA delivery in response to glutathione. Bioconjug. Omata, D., Negishi, Y., Suzuki, R., Oda, Y., Endo-Takahashi, Y., Maruyama, K., 2015.
Chem. 16, 1423–1432. Nonviral Gene Delivery Systems by the Combination of Bubble Liposomes and
Kommareddy, S., Amiji, M., 2007a. Biodistribution and pharmacokinetic analysis of long- Ultrasound. Adv. Genet. 89, 25–48.
circulating thiolated gelatin nanoparticles following systemic administration in Otsuka, H., Nagasaki, Y., Kataoka, K., 2003. PEGylated nanoparticles for biological and
breast cancer-bearing mice. J. Pharm. Sci. 96, 397–407. pharmaceutical applications. Adv. Drug Deliv. Rev. 55, 403–419.
Kommareddy, S., Amiji, M., 2007b. Poly(ethylene glycol)-modified thiolated gelatin na- Panyam, J., Labhasetwar, V., 2003. Biodegradable nanoparticles for drug and gene de-
noparticles for glutathione-responsive intracellular DNA delivery. Nanomed. livery to cells and tissue. Adv. Drug Deliv. Rev. 55, 329–347.
Nanotechnol. Biol. Med. 3, 32–42. Parraga, J.E., Zorzi, G.K., Sanchez, A., Seijo, B., 2009. Gelatin nanoparticles for ocular
Kommareddy, S., Shenoy, D.B., Amiji, M.M., 2005a. Gelatin nanoparticles and their gene delivery, Human gene therapy. Mary Ann Liebert Inc 140 Huguenot Street, 3rd
biofunctionalization. Nanotechnol. Life Sci Online. FL, New Rochelle, NY 10801 USA. pp. 1545–1545.
Kommareddy, S., Tiwari, S.B., Amiji, M.M., 2005b. Long-circulating polymeric nano- Patel, Z.S., Yamamoto, M., Ueda, H., Tabata, Y., Mikos, A.G., 2008. Biodegradable gelatin
vectors for tumor-selective gene delivery. Technol. Cancer Res. Treat. 4, 615–625. microparticles as delivery systems for the controlled release of bone morphogenetic
Konat Zorzi, G., Contreras-Ruiz, L., Parraga, J.E., Lopez-Garcia, A., Romero Bello, R., protein-2. Acta Biomater. 4, 1126–1138.
Diebold, Y., Seijo, B., Sanchez, A., 2011. Expression of MUC5AC in ocular surface Plank, C., Schillinger, U., Scherer, F., Bergemann, C., Rémy, J.-S., Krötz, F., Anton, M.,
epithelial cells using cationized gelatin nanoparticles. Mol. Pharm. 8, 1783–1788. Lausier, J., Rosenecker, J., 2003. The magnetofection method: using magnetic force
Kumar, C.S., 2005. Biofunctionalization of nanomaterials. In: Kumar, Challa S.S.R. (Ed.), to enhance gene delivery. Biol. Chem. 384, 737–747.
Biofunctionalization of Nanomaterials. Wiley-VCH, pp. 377. Prabha, S., Arya, G., Chandra, R., Ahmed, B., Nimesh, S., 2016. Effect of size on biological
Kuo, W.-T., Huang, H.-Y., Chou, M.-J., Wu, M.-C., Huang, Y.-Y., 2011. Surface mod- properties of nanoparticles employed in gene delivery. Artif. Cells Nanomed.
ification of gelatin nanoparticles with polyethylenimine as gene vector. J. Biotechnol. 44, 83–91.
Nanomater. 2011, 28. Ramamoorth, M., Narvekar, A., 2015. Non viral vectors in gene therapy-an overview. J.
Kushibiki, T., Matsuoka, H., Tabata, Y., 2004. Synthesis and physical characterization of Clin. Diagn. Res.: JCDR 9, GE01.
poly (ethylene glycol)-gelatin conjugates. Biomacromolecules 5, 202–208. Sabet, S., George, M.A., El-Shorbagy, H.M., Bassiony, H., Farroh, K.Y., Youssef, T.,
Kushibiki, T., Nagata-Nakajima, N., Sugai, M., Shimizu, A., Tabata, Y., 2006a. Enhanced Salaheldin, T.A., 2017. Gelatin nanoparticles enhance delivery of hepatitis C virus
anti-fibrotic activity of plasmid DNA expressing small interference RNA for TGF-β recombinant NS2 gene. PLoS One 12, e0181723.
type II receptor for a mouse model of obstructive nephropathy by cationized gelatin Sadat, S.M., Jahan, S.T., Haddadi, A., 2016. Effects of size and surface charge of poly-
prepared from different amine compounds. J. Control. Release 110, 610–617. meric nanoparticles on in vitro and in vivo applications. J. Biomater.
Kushibiki, T., Tabata, Y., 2005. Preparation of poly(ethylene glycol)-introduced catio- Nanobiotechnol. 7, 91.
nized gelatin as a non-viral gene carrier. J. Biomater. Sci. Polym. Ed. 16, 1447–1461. Sahoo, N., Sahoo, R.K., Biswas, N., Guha, A., Kuotsu, K., 2015. Recent advancement of
Kushibiki, T., Tomoshige, R., Iwanaga, K., Kakemi, M., Tabata, Y., 2006b. Controlled gelatin nanoparticles in drug and vaccine delivery. Int. J. Biol. Macromol. 81,
release of plasmid DNA from hydrogels prepared from gelatin cationized by different 317–331.
amine compounds. J. Controlled Release: Off. J. Controlled Release Soc. 112, Samal, S.K., Dash, M., Van Vlierberghe, S., Kaplan, D.L., Chiellini, E., Van Blitterswijk, C.,
249–256. Moroni, L., Dubruel, P., 2012. Cationic polymers and their therapeutic potential.
Kushibiki, T., Tomoshige, R., Iwanaga, K., Kakemi, M., Tabata, Y., 2006c. In vitro Chem. Soc. Rev. 41, 7147–7194.
transfection of plasmid DNA by cationized gelatin prepared from different amine Saxena, A., Sachin, K., Bohidar, H., Verma, A.K., 2005. Effect of molecular weight het-
compounds. J. Biomater. Sci. Polym. Ed. 17, 645–658. erogeneity on drug encapsulation efficiency of gelatin nano-particles. Colloids Surf.,
Labhasetwar, V., 2005. Nanotechnology for drug and gene therapy: the importance of B 45, 42–48.
understanding molecular mechanisms of delivery. Curr. Opin. Biotechnol. 16, Schlessinger, J., 2002. Ligand-induced, receptor-mediated dimerization and activation of
674–680. EGF receptor. Cell 110, 669–672.
Lacerda, L., Russier, J., Pastorin, G., Herrero, M.A., Venturelli, E., Dumortier, H., Al- Senning, A., 1997. A review of:“An Introduction to Organosulfur Chemistry.
Jamal, K.T., Prato, M., Kostarelos, K., Bianco, A., 2012. Translocation mechanisms of Sewell, J., Macleod, K., Ritchie, A., Smyth, J., Langdon, S., 2002. Targeting the EGF re-
chemically functionalised carbon nanotubes across plasma membranes. Biomaterials ceptor in ovarian cancer with the tyrosine kinase inhibitor ZD 1839 (‘Iressa’). Br. J.
33, 3334–3343. Cancer 86, 456–462.
Lee, S.J., Yhee, J.Y., Kim, S.H., Kwon, I.C., Kim, K., 2013. Biocompatible gelatin nano- Stone, D., 2010. Novel viral vector systems for gene therapy. Mol. Divers. Preserv. Int.
particles for tumor-targeted delivery of polymerized siRNA in tumor-bearing mice. J. 1002–1007.
Control. Release 172, 358–366. Ter Haar, G., 2007. Therapeutic applications of ultrasound. Prog. Biophys. Mol. Biol. 93,
Lemieux, P., Vinogradov, S.V., Gebhart, C.L., Guerin, N., Paradis, G., Nguyen, H.K., 111–129.
Ochietti, B., Suzdaltseva, Y.G., Bartakova, E.V., Bronich, T.K., St-Pierre, Y., Alakhov, Trabulo, S., Cardoso, A.L., Mano, M., De Lima, M.C.P., 2010. Cell-penetrating pepti-
V.Y., Kabanov, A.V., 2000. Block and graft copolymers and NanoGel copolymer des—mechanisms of cellular uptake and generation of delivery systems.
networks for DNA delivery into cell. J. Drug Target. 8, 91–105. Pharmaceuticals 3, 961–993.
Li, S.-D., Huang, L., 2007. Non-viral is superior to viral gene delivery. J. Controlled Tseng, C.-L., Wu, S.Y.-H., Wang, W.-H., Peng, C.-L., Lin, F.-H., Lin, C.-C., Young, T.-H.,
Release: Official J. Controlled Release Soc. 123, 181. Shieh, M.-J., 2008. Targeting efficiency and biodistribution of biotinylated-EGF-
Liu, C., Gong, C., Pan, Y., Zhang, Y., Wang, J., Huang, M., Wang, Y., Wang, K., Gou, M., conjugated gelatin nanoparticles administered via aerosol delivery in nude mice with
Tu, M., 2007. Synthesis and characterization of a thermosensitive hydrogel based on lung cancer. Biomaterials 29, 3014–3022.

233
O. Madkhali et al. International Journal of Pharmaceutics 554 (2019) 224–234

Vega-Villa, K.R., Takemoto, J.K., Yáñez, J.A., Remsberg, C.M., Forrest, M.L., Davies, N.M., Yameen, B., Choi, W.I., Vilos, C., Swami, A., Shi, J., Farokhzad, O.C., 2014. Insight into
2008. Clinical toxicities of nanocarrier systems. Adv. Drug Deliv. Rev. 60, 929–938. nanoparticle cellular uptake and intracellular targeting. J. Control. Release 190,
Vonarbourg, A., Passirani, C., Saulnier, P., Benoit, J.-P., 2006. Parameters influencing the 485–499.
stealthiness of colloidal drug delivery systems. Biomaterials 27, 4356–4373. Yin, H., Kanasty, R.L., Eltoukhy, A.A., Vegas, A.J., Dorkin, J.R., Anderson, D.G., 2014.
Wang, H., Boerman, O.C., Sariibrahimoglu, K., Li, Y., Jansen, J.A., Leeuwenburgh, S.C., Non-viral vectors for gene-based therapy. Nat. Rev. Genet. 15, 541.
2012. Comparison of micro-vs. nanostructured colloidal gelatin gels for sustained Young, J.L., Dean, D.A., 2015. Chapter three-electroporation-mediated gene delivery.
delivery of osteogenic proteins: bone morphogenetic protein-2 and alkaline phos- Adv. Genet. 89, 49–88.
phatase. Biomaterials 33, 8695–8703. Zillies, J., Coester, C., 2005. Evaluating gelatin based nanoparticles as a carrier system for
Wirth, T., Parker, N., Ylä-Herttuala, S., 2013. History of gene therapy. Gene 525, double stranded oligonucleotides. J. Pharm. Pharm. Sci.: Publ. Can. Soc. Pharm. Sci.
162–169. Societe canadienne des Sci. Pharm. 7, 17–21.
Xu, J., Amiji, M., 2012. Therapeutic gene delivery and transfection in human pancreatic Zorzi, G.K., Parraga, J.E., Seijo, B., Sanchez, A., 2015. Comparison of different cationized
cancer cells using epidermal growth factor receptor-targeted gelatin nanoparticles. J. proteins as biomaterials for nanoparticle-based ocular gene delivery. Colloids Surf. B
Visualized Exp: JoVE, e3612. Biointerfaces 135, 533–541.
Xu, J., Ganesh, S., Amiji, M., 2012. Non-condensing polymeric nanoparticles for targeted Zorzi, G.K., Párraga, J.E., Seijo, B., Sánchez, A., 2011. Hybrid nanoparticle design based
gene and siRNA delivery. Int. J. Pharm. 427, 21–34. on cationized gelatin and the polyanions dextran sulfate and chondroitin sulfate for
Xu, J., Gattacceca, F., Amiji, M., 2013. Biodistribution and pharmacokinetics of EGFR- ocular gene therapy. Macromol. Biosci. 11, 905–913.
targeted thiolated gelatin nanoparticles following systemic administration in pan- Zwiorek, K., Bourquin, C., Battiany, J., Winter, G., Endres, S., Hartmann, G., Coester, C.,
creatic tumor-bearing mice. Mol. Pharm. 10, 2031–2044. 2008. Delivery by cationic gelatin nanoparticles strongly increases the im-
Xu, J., Singh, A., Amiji, M.M., 2014. Redox-responsive targeted gelatin nanoparticles for munostimulatory effects of CpG oligonucleotides. Pharm. Res. 25, 551–562.
delivery of combination wt-p53 expressing plasmid DNA and gemcitabine in the Zwiorek, K., Kloeckner, J., Wagner, E., Coester, C., 2004. Gelatin nanoparticles as a new
treatment of pancreatic cancer. BMC Cancer 14, 75. and simple gene delivery system. J. Pharm. Pharm. Sci.:Publ. Can. Soc. Pharm. Sci.
Xu, X., Capito, R.M., Spector, M., 2008. Delivery of plasmid IGF-1 to chondrocytes via Societe canadienne des Sciences Pharm. 7, 22–28.
cationized gelatin nanoparticles. J. Biomed. Mater. Res. Part A 84, 73–83.

234

You might also like