Trends in Analytical Chemistry: Shweta Sharma, Ankush Parmar, Shivpoojan Kori, Rajat Sandhir

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Trends in Analytical Chemistry 80 (2016) 30–40

Contents lists available at ScienceDirect

Trends in Analytical Chemistry


j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / t r a c

PLGA-based nanoparticles: A new paradigm in biomedical


applications
Shweta Sharma *, Ankush Parmar, Shivpoojan Kori, Rajat Sandhir
Institute of Forensic Science & Criminology, Panjab University, Chandigarh 160 014, India

A R T I C L E I N F O A B S T R A C T

Keywords:
Polymers were first introduced three decades ago as bioresorbable surgical devices. Since then, polymer-
Biodegradable
based nanoparticles have been extensively studied in a variety of fields. Nanocarriers formulated with
Poly(lactic-co-glycolic acid)
Nanoparticles
biocompatible and biodegradable polymers approved by the US FDA (Food and Drug Administration) and
Targeting EMA (European Medicines Agency) are being studied for the controlled delivery of various therapeutic
Sustained release agents. Amidst the various polymers synthesized for formulating polymeric nanoparticles, poly(lactic-
co-glycolic acid) (PLGA) is the most popular. PLGA has several interesting properties such as controlled
and sustained release, low cytotoxicity, long-standing biomedical applications, biocompatibility with tissues
and cells, prolonged residence time and targeted delivery. The main aim of this review was to compre-
hensively address the issues related to PLGA-based nanoparticles focusing on the methods of preparation,
characterization techniques, surface modification, mechanism of drug release and the drawbacks. The
review also critically addresses the developmental aspects of PLGA-based nanocarriers in terms of tar-
geted drug delivery, as well as exploring their efficacy in vitro and in vivo.
© 2016 Elsevier B.V. All rights reserved.

Contents

1. Introduction ........................................................................................................................................................................................................................................................... 31
2. Poly(lactic-co-glycolic acid) ............................................................................................................................................................................................................................. 31
2.1. Properties of PLGA ................................................................................................................................................................................................................................. 31
3. Methods of preparing PLGA nanoparticles ................................................................................................................................................................................................. 32
3.1. Emulsification–solvent evaporation method ................................................................................................................................................................................ 32
3.1.1. Single emulsion method ..................................................................................................................................................................................................... 32
3.1.2. Double emulsion method ................................................................................................................................................................................................... 32
3.2. Phase separation (coacervation) ....................................................................................................................................................................................................... 32
3.3. Emulsification–solvent diffusion method ...................................................................................................................................................................................... 32
3.4. Emulsification–reverse salting-out method ................................................................................................................................................................................. 32
3.5. Nanoprecipitation method (solvent displacement) ................................................................................................................................................................... 33
3.6. Dialysis ...................................................................................................................................................................................................................................................... 33
3.7. Spray drying ............................................................................................................................................................................................................................................ 33
3.8. Supercritical fluid technology ........................................................................................................................................................................................................... 33
3.8.1. Rapid expansion of supercritical solutions .................................................................................................................................................................. 33
3.8.2. Rapid expansion of supercritical solutions in liquid solvents .............................................................................................................................. 33
4. Characterization parameters for nanoparticles ......................................................................................................................................................................................... 33
5. Surface modification of PLGA nanoparticles .............................................................................................................................................................................................. 33

Abbreviations: DCC, N,N-Dicyclohexylcarbodiimide; DDS, Drug delivery system; DNA, Deoxyribose nucleic acid; EDC, 1-ethyl-3-(3-dimethylaminopropyl carbodiimide);
EGFR, Epidermal growth factor receptor; EMA, European Medicines Agency; EPR, Enhanced permeability and retention; FDA, Food and Drug Administration; FTIR, Fourier
transform infrared; GALT, Gut-associated lymphoid tissue; MDR, Multidrug resistance; MMPs, Matrix metalloproteinases; MPS, Mononuclear phagocyte system; MRI, Mag-
netic resonance imaging; NHS, N-hydroxysuccinimide; NMR, Nuclear magnetic resonance; O/W, Oil in water; PEG, Polyethylene glycol; PGA, Polyglycolic acid; P-gP, P-glycoprotein;
PLA, Polylactic acid; PLGA, Poly(lactic-co-glycolic acid); PNP, Polymeric nanoparticle; RES, Reticuloendothelial system; RNA, Ribose nucleic acid; SiRNA, Short interfering
ribose nucleic acid; 99mTC, Technetium 99m; Tg, Glass transition temperature; TPGS, d-α-tocopherol polyethylene glycol succinate; VCAM, Vascular cell adhesion mole-
cule; VEGFR, Vascular endothelial growth factor receptors; W/O/W, Water in oil–water; XPS, X-ray photoelectron spectroscopy.
* Corresponding author. Tel.: +91 172 2534121(O) +91 9872688577 (M).
E-mail address: 25shweta@pu.ac.in (S. Sharma).

http://dx.doi.org/10.1016/j.trac.2015.06.014
0165-9936/© 2016 Elsevier B.V. All rights reserved.
S. Sharma et al./Trends in Analytical Chemistry 80 (2016) 30–40 31

5.1. Polyethylene glycol ................................................................................................................................................................................................................................ 33


5.1.1. PEGylation strategies ............................................................................................................................................................................................................ 34
5.1.2. Polysorbate .............................................................................................................................................................................................................................. 34
5.1.3. Vitamin E TPGS ...................................................................................................................................................................................................................... 34
6. Mechanism of drug release from PLGA-based DDSs ............................................................................................................................................................................... 34
7. Physiochemical changes occurring in PLGA-based DDSs ...................................................................................................................................................................... 35
8. Drug release behaviour ..................................................................................................................................................................................................................................... 36
8.1. Phase I ........................................................................................................................................................................................................................................................ 36
8.2. Phase II ...................................................................................................................................................................................................................................................... 36
9. Factors affecting degradation .......................................................................................................................................................................................................................... 36
9.1. Polymer composition and molecular weight ............................................................................................................................................................................... 36
9.2. Drug type .................................................................................................................................................................................................................................................. 36
9.3. Size and shape of the matrix ............................................................................................................................................................................................................. 36
9.4. pH ................................................................................................................................................................................................................................................................ 36
9.5. Drug load .................................................................................................................................................................................................................................................. 37
10. PLGA-mediated drug delivery for cancer treatment ............................................................................................................................................................................... 37
11. Targeting strategies for efficient drug delivery ......................................................................................................................................................................................... 37
11.1. Passive targeting .................................................................................................................................................................................................................................. 37
11.2. Active targeting .................................................................................................................................................................................................................................... 37
11.2.1. Targeting of cancer cells ................................................................................................................................................................................................... 37
11.2.2. Targeting of tumour endothelium ................................................................................................................................................................................ 37
12. Ligand-anchored PLGA nanoparticles for cancer therapy ..................................................................................................................................................................... 38
13. PLGA nanoparticles as thriving mediators .................................................................................................................................................................................................. 38
13.1. Gene delivery for cancer treatment .............................................................................................................................................................................................. 38
13.2. Diagnosis and imaging of cancer ................................................................................................................................................................................................... 38
13.3. Theranostics of cancer ....................................................................................................................................................................................................................... 39
14. Drawbacks of using PLGA nanoparticle-based DDSs .............................................................................................................................................................................. 39
15. Conclusion .............................................................................................................................................................................................................................................................. 39
Acknowledgement ............................................................................................................................................................................................................................................... 39
References .............................................................................................................................................................................................................................................................. 39

1. Introduction nanoparticles [7]. The study by Joo et al. was comprehensive, de-
scribing various features of PLGA including surface modification,
In recent years, nanoparticles have been increasingly studied in targeting aspect and the intrinsic capacity with uptake of the cancer
the field of biomedical sciences. Depending upon the nature of the drug carrier [8]. The present review describes in detail the charac-
polymer used in their formulation, these particles may be catego- teristics of PLGA and its associated potentials in terms of the
rized into natural or synthetic. Delivery of various substances such structure–property relationship. The efficacy of PLGA nanoparticles
as vaccines, macromolecules and hydrophobic drugs to cells and was improved through ligand anchoring, which were applied as me-
organs such as the brain, liver and lungs can be achieved via these diators for gene delivery, in imaging of cancer and in the field of
nanoparticles, thus making them a multifaceted platform for tar- theranostics.
geted delivery [1]. However, for use as a vector for drug delivery
systems (DDSs), a nanoparticle must possess vital properties such 2. Poly(lactic-co-glycolic acid)
as biocompatibility, drug compatibility and proper biodegrada-
tion kinetics. A site-specific action of the drug at a therapeutically Polyester PLGA is a copolymer of polylactic acid (PLA) and
optimal rate and dose regimen can be achieved by controlling for polyglycolic acid (PGA). In terms of design and performance, PLGA
parameters such as particle size, surface properties and release rate is the best candidate as biomaterials for drug delivery. Here, PLGA
during the synthesis and design of the nanoparticles. Polymer- denotes poly-D, L-lactic-co-glycolic acid, where D- and L-lactic acid
based nanoparticles have been used for site-specific delivery, for forms are present in a fixed ratio [9]. The need for efficient and su-
cancer therapy, for clinical bioanalytical diagnostics, to design tissue- perior DDSs has led to the development of assorted block copolymers
engineered scaffolds and in devices [2]. For the synthesis of of polyesters with PEG. PLGA/PEG block copolymers are available
nanoparticles, a variety of polymers have been used, but the co- in two varieties: diblock (PLGA–PEG) or triblock with both ABA
polymer poly(lactic-co-glycolic acid) (PLGA) has been widely used (PLGA–PEG–PLGA) and BAB (PEG–PLGA–PEG).
in this context. PLGA is a biocompatible, biodegradable and safely
administrable polymer approved by the US FDA (Food and Drug Ad- 2.1. Properties of PLGA
ministration) and EMA (European Medicines Agency) [3]. Langer and
Folkman [4] were the first to report the controlled release of mac- PLGA is synthesized via random ring-opening copolymeriza-
romolecules via polymers. Thus, their pioneering work led to further tion of two different monomers, glycolic acid and lactic acid, in the
discoveries in the field of novel DDSs. This work led to the evolu- presence of tin (II) 2-ethylhexanote, tin (II) alkoxides or alu-
tion of anti-angiogenic DDSs to treat serious diseases such as cancer. minium isopropoxide as the catalyst. Glycolic and lactic acid units
Some reports have proved PLGA nanoparticles as an excellent vector are consecutively linked via ester linkages during polymerization,
for the transmission of biomolecules such as RNA, DNA, peptides, resulting in the formation of PLGA.
vitamins, proteins and drugs both in vivo and in vitro. Stevanovic Different forms of PLGA can be obtained by varying the ratio of
and Uskokovic highlighted the efficient delivery of vitamins using lactide to glycolide during the polymerization reaction, for example,
PLGA-based nano- and microparticles [5]. . In their review article, PLGA 50:50 (refers to a copolymer composed of 50% lactic acid and
Choi et al concisely described PLGA-aided tumour targeting [6]. In 50% glycolic acid), PLGA 75:25, PLGA 80:20, etc. Depending on the
another report, Locatelli and Franchini explained the synthesis and molecular weight and the lactide to glycolide copolymer ratio used,
applications of PLGA-b-polyethylene glycol (PEG) polymeric the deterioration time of the polymer may vary from several months
32 S. Sharma et al./Trends in Analytical Chemistry 80 (2016) 30–40

to several years [1,10]. Low molecular weight polymers with higher acetate, chloroform or dichloromethane). Thus, a water-in-oil primary
glycolide content are more hydrophilic and amorphous, and thus emulsion is formed. Further emulsification is achieved by adding
have a shorter deterioration time. This is because glycolic acid is the primary emulsion to an aqueous solution, followed by simul-
more hydrophilic and thus tends to absorb a large amount of water. taneous stirring and thereafter allowing the organic solvent to
Conversely, polymers with higher lactic acid content are more hy- evaporate [17]. Both of these techniques are optimal for laborato-
drophobic, absorb less amount of water and degrade in a more ry synthesis; on a large scale, however, they are applicable only to
gradual manner [11]. This phenomenon has been proven useful for liposoluble drugs and require excess energy during homogeniza-
controlled and sustained drug release varying from weeks to months. tion. However, it has been reported that any alteration in the process
The hydrolysis of PLGA yields two metabolic monomers: lactic acid parameters, for example, stirring speed and temperature, helps over-
and glycolic acid. Via the Krebs cycle, these monomers are endog- come the drawbacks of these techniques [8].
enously metabolized into the simpler by-products such as CO2 and
H2O [12]. PLGA can be processed into almost any kind of configu- 3.2. Phase separation (coacervation)
ration and can be used to encapsulate a variety of molecules. PLGA
polymers are miscible in various volatile organic solvents such as This process mainly leads to the formation of micrometre-
tetrahydrofuran, acetone, dichloromethane, chloroform and ethyl sized polymeric nanoparticles via liquid–liquid phase separation.
acetate. Physical properties such as molecular weight, polydisper- In this method, two liquid phases comprising a coacervate phase
sity index, Tg and degree of crystallinity are known to affect the and a supernatant phase depleted in the polymer are formed. The
swelling behaviour, biodegradation rate and mechanical strength coacervation process includes three major steps [9]:
of the polymer [13]. The type and molar ratio of the individual
monomer components in the polymer chain chiefly determine the (i) Phase separation of the coating polymer solution,
degree of crystallinity. (ii) Adsorption of the coacervate around the drug particles and
Makadia and Siegel [9] showed that a decrease in the degree of (iii) Quenching of the microspheres.
crystallinity along with an increase in the hydration rate (hydro-
lysis) is observed during copolymerization of a crystalline PGA with Polymers and solvent are mixed in an appropriate ratio, fol-
PLA. PLGA copolymers are generally glassy in nature and exhibit a lowed by the addition of the drug (i.e., hydrophilic drug in the
fairly rigid chain structure. This may be attributed to the Tg of PLGA W/O/W emulsion and hydrophobic drug in the O/W emulsion). A
exceeding its physiological temperature [14]. It has been demon- soft coacervate of the drug encapsulated in a droplet is extracted
strated that the dose and composition of PLGA nanocarriers has a as a result of phase separation of the polymer. The microdroplets
demarcating effect on blood clearance and uptake of these are quenched by dipping the coacervate quickly in an insoluble
nanocarriers by the mononuclear phagocyte system (MPS) [15]. medium. However, the morphology and size of the microspheres
can be controlled by altering the following process parameters [18]:
3. Methods of preparing PLGA nanoparticles
(a) Polymer concentration
Depending on the process of preparation, the structural orga- (b) Quenching temperature
nization of the nanoparticle may vary. Biodegradable PLGA (c) Quenching time
nanoparticles are generally prepared by dispersing the polymer. The (d) Solvent composition
nanoparticles are actually formed in the initial step, which is (e) Stirring rate
common for all techniques, wherein an emulsification system is pre-
pared [8]. The different methods of preparation are discussed in the 3.3. Emulsification–solvent diffusion method
following sections:
This technique is a modification of the salting-out process. An
3.1. Emulsification–solvent evaporation method initial thermodynamic equilibrium is attained by mutually satu-
rating the solvent and water at room temperature before use.
3.1.1. Single emulsion method Emulsification of the organic phase containing the polymer and drug
This is the most commonly used method for preparing PLGA in an aqueous surfactant solution is achieved via high-speed ho-
nanoparticles. O/W emulsification is generally used when the en- mogenization. In order to attain a phase transformation reaction and
capsulant is hydrophobic or poorly soluble in water [16]. To prepare outward diffusion of the solvent from the internal phase, water is
an organic phase, an appropriate amount of the polymer is initial- added while regularly stirring. Colloidal nanoparticles are formed
ly dissolved in a volatile organic solvent such as dichloromethane, as a result of nanoprecipitation. Solvent evaporation is then facili-
chloroform or ethyl acetate. Then, the drug or encapsulant is added tated by either evaporation or vacuum distillation [19]. This method
to this solution, resulting in dispersion. The dispersion containing offers several advantages such as enhanced encapsulation efficien-
the polymer and the drug is added to a continuously stirring aqueous cy, high batch-to-batch reproducibility, ease of scale-up, narrow size
solution with surfactants such as polyvinyl alcohol, polysorbate 80, distribution and simplicity. However, on the contrary, this tech-
poloxamer 188 and vitamin E TPGS (d-α-tocopherol polyethylene nique has limitations such as leakage of water-soluble drugs and
glycol succinate), thus generating a stable emulsion. The organic removal of high volume of water from the suspension.
solvent is then allowed to evaporate either by magnetic stirring or
by maintaining a reduced pressure [9]. 3.4. Emulsification–reverse salting-out method

3.1.2. Double emulsion method The organic phase containing the polymer and drug is firstly
The double emulsion method is also known as the W/O/W added to a water-miscible solvent. An O/W emulsion is formed by
method. The encapsulation efficiency and particle size are pre- forcefully magnetically stirring the preformed organic solvent with
dominantly affected by the type of solvent and stirring rate. An an aqueous solution containing the salting-out agent (e.g., magne-
appropriate amount of the drug is dissolved in an aqueous phase sium chloride or calcium chloride) and a colloidal stabilizer (e.g.,
(prepared in deionized water) followed by simultaneous addition polyvinyl pyrrolidone). An abrupt increase in the continuous phase
of the drug solution to a vigorously stirring organic phase (com- of the emulsion is facilitated by further diluting the O/W with a large
posed of PLGA dissolved in volatile organic solvents such as ethyl volume of water. The volatile organic solvent diffuses into the
S. Sharma et al./Trends in Analytical Chemistry 80 (2016) 30–40 33

aqueous phase starts, leading to the formation of nanoparticles [20]. profile and degradation pattern. Dynamic light scattering, scan-
The residual solvent and salting-out agents are removed by filtra- ning electron microscopy, transmission electron microscopy and
tion, leaving the nanoparticles behind. This technique offers the atomic force microscopy can be used to determine parameters such
advantage of encapsulating heat-sensitive agents such as pro- as size, distribution and morphology of the nanoparticle. It has also
teins, DNA and RNA. been revealed that the molecular weight of the polymer has an
adverse effect on the particle size, encapsulation efficiency and deg-
3.5. Nanoprecipitation method (solvent displacement) radation rate [23]. The chain length of the polymer represents the
molecular weight of the polymer, thus reflecting the chemical nature
It is a one-step process, generally used to entrap hydrophobic of the polymer, i.e. its hydrophobic or hydrophilic nature. It is well
drugs in the polymer matrix. The organic phase is formed by dis- known that polymers with shorter chain length are hydrophobic
solving the polymer and drug in a polar solvent (e.g., acetone, ethanol, and have a faster degradation rate. However, polymers with a longer
methanol and acetonitrile). This solution is added drop-wise to an chain length are generally hydrophilic in nature and have a shorter
aqueous solution containing an emulsifier or a surfactant. The rapid degradation rate. Thus, the molecular weight of the polymer plays
diffusion of the solvent takes place, resulting in the formation of a crucial role in deciding the release kinetics of the drug [29]. Size
nanoparticles [21–23]. exclusion chromatography is useful in determining the molecular
weight of the polymer [30]. It has also been observed that the in
3.6. Dialysis vitro and in vivo release characteristics of the drug are affected by
the physical state of both the drug and polymer. The muco-adhesion,
Dialysis is generally used to prepare small-sized nanoparticles nanoparticle constancy and intracellular trafficking are greatly de-
with a narrow distribution. The polymer is dissolved in a volatile pendent on the zeta potential [31]. The biodistribution of the
organic solvent and placed in a dialysis tube of appropriate pore nanoparticles is greatly dependent upon their hydrophobic nature.
size. Inside the dialysis bag, displacement of the solvent takes place, Various studies have corroborated that the retention time of hy-
along with a loss in the solubility, ultimately leading to the pro- drophilic particles is more that of hydrophobic particles [32].
gressive aggregation of polymer and formation of a homogeneous Techniques such as water contact angle measurement and hydro-
suspension of nanoparticles [21,24]. phobic interaction chromatography may be used to determine the
hydrophobicity and hydrophilicity of the nanoparticles, respective-
3.7. Spray drying ly [33]. The surface chemistry can be analysed with a variety of
techniques such as X-ray photoelectron spectroscopy (XPS), Fourier
Spray drying is an alternative to the conventional methods of transform infrared (FTIR) spectroscopy and nuclear magnetic res-
preparing polymer nanoparticles. This technique offers several ad- onance (NMR) spectroscopy [23,34].
vantages such as rapidity, convenience and implementation of fewer
processing parameters. In this process, a W/O dispersion is sprayed 5. Surface modification of PLGA nanoparticles
in a hot stream of air, leading to the formation of nanoparticles.
However, the adhesion of the nanoparticles to the inner walls of To serve as a targeted drug delivery vehicle, the nanoparticle must
the spray dryer hinders the effective collection of the formed par- be capable of persisting in the human systematic circulation. A pro-
ticles [25]. longed circulation time will help the nanoparticles reach the target
organ. On the contrary, these particles are removed from the blood-
3.8. Supercritical fluid technology stream by the reticuloendothelial system (RES). This phenomenon
has proven to be one of the foremost challenges in developing
Supercritical fluid technology has been proven to be a more nanoparticle-based DDS [1,35]. This may be attributed to the binding
environment-friendly approach to produce polymeric nanoparticles of nanoparticles with the opsonin proteins present in the blood
(PNPs). It can produce PNPs with high purity and fewer traces of serum when administered intravenously. Further, the opsonized par-
organic solvent [26]. Two principal methods are used to produce ticles become attached to the macrophages, where they are
nanoparticles [27]: ultimately internalized by phagocytosis. The final fate of such par-
ticles is their clearance from the body via the renal system. Therefore,
3.8.1. Rapid expansion of supercritical solutions despite their favourable biocompatibility and biodegradability, PLGA
A solution is formed by dissolving the solute in a supercritical nanoparticles are likely to be cleared rapidly from circulation by
fluid, accompanied by the rapid expansion of the solution in the macrophages of the MPS immediately after their intravenous ad-
ambient air. Homogeneous nucleation is achieved by reducing the ministration [36]. To overcome these limitations, the bare
pressure rapidly. Due to the rapid reduction in pressure, a high degree nanoparticles are subjected to surface modification. Surface modi-
of supersaturation is achieved, leading to expansion and ultimate- fication plays a crucial role in escaping the natural defence system
ly formation of nanoparticles [27]. of the body. Hydrophilic particles with size of about ≤100 nm have
the greatest survival rate in escaping the phagocytic system [23].
3.8.2. Rapid expansion of supercritical solutions in liquid solvents The retention time for hydrophilic nanoparticles is comparatively
It is a modified form of rapid expansion of supercritical solu- longer than that for hydrophobic nanoparticles. This is because hy-
tions, with expansion of the supercritical solution taking place in drophobic nanoparticles are preferably taken up by the RES and are
a liquid solvent instead of ambient air. Apparently, particle growth thus eliminated from the body [8]. To construct a hydrophilic core
is suppressed in the expansion jet, by the action of the liquid solvent, around PLGA nanoparticles, the nanoparticles are coated with surface
leading to the formation of nanoparticles [27,28]. derivatizers. The following chemical species act as good surface
modifiers:
4. Characterization parameters for nanoparticles
5.1. Polyethylene glycol
Characterization becomes a prerequisite for understanding the
properties of the nanoparticles. Several parameters can provide PEG is a nonionic, hydrophilic polymer with excellent biocom-
insight into the properties of the nanoparticles. First and fore- patibility. Coating of the nanoparticle surface with PEG, that is,
most, size helps determine the efficacy of the nanoparticles, release PEGylation, is the most commonly used technique for surface
34 S. Sharma et al./Trends in Analytical Chemistry 80 (2016) 30–40

Table 1
PLGA–PEG nanoparticles for anticancer drug delivery

Nanoparticles Drug loaded Target Inference Reference

PLGA–PEG Paclitaxel HeLa Entrapment of paclitaxel within a PEGylated PLGA nanoparticle resulted in higher tumour growth [40]
nanoparticles inhibition, followed by a significant augmentation in the tumour-specific localization of the drug
PLGA–mPEG Cisplatin BALB/c miceaa Higher survival rate and delayed tumour growth was observed for PLGA–mPEG nanoparticle-treated [41]
nanoparticles mice in comparison with animals treated with the free drug
PLGA–PEG Docetaxel Solid tumors Biological half-life of the drug is significantly augmented, while imparting considerable solid [42]
nanoparticles tumour accumulation

modification of the nanoparticles. The process allows the environment during the conjugation process for achieving maximum
nanoparticles to evade the MPS attack, thus concomitantly increas- efficiency.
ing their plasma half-life [37]. However, the mechanism underlying
this increase in the plasma half-life remains to be elucidated. It is 5.1.1.2. Activated conjugation. It is a two-step process where acti-
assumed that stearic repulsion as well as van der Waals forces vation is succeeded by conjugation. Minimal hydrolysis of the active
created by the hydrated barriers on the nanoparticle surface pre- intermediate occurs, and the undesirable formation of PEG–PEG con-
vents the nanoparticles from being opsonized. The high flexibility jugates is avoided [43]. Fig. 1b shows the synthetic route used for
of the polymer chain allows the free rotation of the polymer units, the conjugation of heterofunctional PEG to PLGA nanoparticles.
creating a highly hydrophilic stealth corona around the nanoparticles.
Thus, the nanoparticles are prevented from interacting with the mac- 5.1.1.3. Ring-opening polymerization. Among all the strategies used
romolecules present in the body. It has also been observed that PEG for the production of PEGylated PLGA nanoparticles, ring-opening
molecules with high molecular weight, high surface density and polymerization is the most commonly and widely used technique.
longer chain length are absorbed at a comparatively lower rate, thus The reaction is initiated by protic agents such as the hydroxyl group
increasing the residence time of the nanoparticle [38]. Moreover, of OH–PEG–COOH, leading to the formation of PLGA with hy-
PEGylation of PLGA nanoparticles has also been shown to enhance droxyl end groups, whereas the carboxyl end groups of PEG remain
the drug payload, solubility and kinetic stability, thereby improv- free. Fig. 1c shows the ring-opening polymerization of PEG to PLGA
ing the targeting index, therapeutic index as well as the accessibility nanoparticles.
of the nanoparticle to the target site. With the help of PEGylation,
the aqueous solubility and stability can be enhanced, intermolecu- 5.1.2. Polysorbate
lar aggregation can be reduced and immunogenicity can be Polysorbate 20, 40, 60 and 80 are the most commonly used non-
decreased. In their study, Danhier et al. showed that the entrap- ionic surfactants and emulsifiers in food and cosmetic products. Due
ment of paclitaxel within a PEGylated-PLGA-based nanoparticle to the surface coating of polymer nanoparticles with polysorbate,
altered its pharmacokinetics and biodistribution such that the their ability to cross the blood–brain barrier is enhanced. Because
tumour-specific localization of the drug was significantly aug- of the binding of the nanoparticles to the inner linings of the brain
mented, leading to higher tumour growth inhibition efficiency than capillaries, a large change in the concentration gradient is ob-
the free drug [39]. In another experiment, Gref et al. showed that served along with an increase in passive diffusion, resulting in
the PEGylated PLGA nanoparticles effect an increase in the circu- facilitated delivery of the drug to the brain.
lation time and decreased uptake by the liver; that is, the uptake
of nanoparticles was reduced from 66% within the first 5 min of ad- 5.1.3. Vitamin E TPGS
ministration, but the uptake was <30% (within 2 h) for non-coated Vitamin E TPGS is a synthetic water-soluble form of vitamin E.
nanoparticles [40]. Although long-circulating nanoparticles formu- TPGS is a PEG derivative of α-tocopherol that is water soluble. It is
lated with PEGylated PLGA possess many significant advantages, this commonly used as an emulsifier, a solubilizer and a vehicle in drug
strategy is also not free from limitations. Capping of PLGA with PEG delivery formulations. Vitamin E TPGS is also widely used as an emul-
prevent the interaction not only between the nanoparticles and the sifier for enhanced encapsulation efficiency, drug loading and release
opsonin but also between nanoparticles and the cell surface [36]. kinetics of hydrophobic drugs such as paclitaxel, doxorubicin and
Table 1 shows the effect of PEGylation on the uptake of PLGA 5-fluorouracil. The molecule has shown to improve the adhesion
nanoparticles in cancerous cells [40–42]. of nanoparticles to the cells and the haemodynamic properties of
the nanoparticles [33].
5.1.1. PEGylation strategies
5.1.1.1. Direct conjugation. Betancourt et al. reported three differ- 6. Mechanism of drug release from PLGA-based DDSs
ent methods for the covalent attachment of PEG to PLGA
nanoparticles [43]. Based on their study, varying the reaction con- The term ‘release mechanism’ may be defined as the way in which
ditions can help control the efficacy of conjugation as well as the drug molecules are transported or released and as an event of the
final copolymer composition. A non-significant incorporation of PEG process determining the release rate. Drug molecules are released
occurred when the PEG molecule was directly conjugated to the car- via three possible mechanisms [44]:
boxylic groups present on the surface of PLGA nanoparticles, because
of the inaccessibility of these groups. Moderate efficiency is ob- (i) Transport through water-filled pores
served, when the conjugation reactions are carried out in solution. (ii) Transport through the polymer
Direct conjugation of PLGA nanoparticles with PEG would also fa- (iii) Dissolution of the encapsulating polymer
cilitate the encapsulation of a desired agent with the nanoparticle,
as PEGylation would occur after the desired agent is encapsulated Large and highly hydrophilic molecules such as protein or DNA
within the solid polymer matrix core (Fig. 1a). However, the tech- are encapsulated agents that are generally transported through the
nique has certain limitations such as low yield because of the polymer phase and are released by transport through water-filled
purification steps. The nanoparticles must be exposed to an aqueous pores. Transport through water-filled pores is mainly achieved via
S. Sharma et al./Trends in Analytical Chemistry 80 (2016) 30–40 35

Fig. 1. (a). Direct conjugation of PEG to the surface of premade PLGA nanoparticles. (b) Activated conjugation of PLGA to heterofunctional PEG. (c) Ring-opening polymer-
ization of lactide and glycolide.

a process known as diffusion, where the molecules are driven by a 7. Physiochemical changes occurring in PLGA-based DDSs
chemical potential gradient. Another phenomenon of transport is
convection, where osmotic pressure is the driving force and hence When the DDS is administered in vitro or immersed in water,
the name osmotic pumping. This pressure may generate an influx the polymer quickly absorbs water. The water molecules occupy
of water into a non-swelling system. The phenomenon is most com- some volume in the polymer matrix, which is often regarded as pores
monly confronted in the case of cellulose acetate-derived DDSs [45]. and the process is said to be the pore-forming process. Due to the
PLGA possesses mobile polymer chains and can absorb a large small size of the pores, a minimal amount of drug is transported
amount of water, thus leading to a prominent swelling. Due to re- during the initial release phase. However, over time, the water-
arrangement of the polymer chain and swelling, the increase in the filled pores start to grow in size and number, thus creating a porous
volume of water is compensated. Sometimes, the encapsulated drug connected network. This network facilitates the drug release during
may be released as a result of dissolution of the polymer or erosion. the later phase. However, heterogeneous degradation via an auto-
Pores are created and an increase in the rate of diffusion is ob- catalytic phenomenon takes place when PLGA matrix comes in
served. The release rate is controlled by diffusion at the initial stages, contact with water. This phenomenon is termed as hydrolysis. During
whereas degradation or erosion plays a very vital role in the final hydrolysis, the ester bonds are broken, with a decrease in the mo-
stage of the release period. Thus, it can be concluded that drug lecular weight of the polymer and generation of acids [46]. With
release is controlled by more than one mechanism at once [44]. an increase in the dimension of the DDS, degradation is faster at
36 S. Sharma et al./Trends in Analytical Chemistry 80 (2016) 30–40

the centre of the PLGA matrix than at the surface [47]. Further- 8.2. Phase II
more, loss of polymer occurs when the dissolved polymer
degradation products diffuse into the release medium. As it can be In the second phase, the thicker drug layer is depleted, leading
hydrated rapidly, the PLGA polymer undergoes bulk erosion in spite to progressive release of the drug. Soluble oligomeric and mono-
of surface erosion. Pores are created by the dissolution of polymer meric products are formed due to the hydrolysis of the polymer
degradation and erosion processes. The generated small pores grow matrix. Hydrolysis creates a passage drug to be released by diffu-
in size by coalescing with the neighbouring pores, ultimately leading sion and erosion until complete polymer solubilization takes place.
to the formation of larger pores. The PLGA polymer can be rear- However, in the case of a triphasic release profile, phase I is usually
ranged by mobilizing its polymeric chains, leading to the formation referred to as burst release. This kind of behaviour is attributed mainly
of larger pores. The effects of dissolved degradation products on DDS to the hydration of non-encapsulated drug particles present on the
are as follows: surface or close to the surface of the DDS. As degradation and hy-
dration proceed, the polymer begins to grow dense, and consequently
(i) Due to their acidic nature, they catalyse hydrolysis a slow release or diffusion of the drug is observed in phase II. Burst
(ii) An increase in the rate of water absorption along with a de- release is sometimes followed by erosion, which is a comparative-
crease in the transport resistance of the polymer is observed ly fast release phase and sometimes called the second burst.
due to plasticization of the polymer by the dissolved degra-
dation product 9. Factors affecting degradation
(iii) The osmolality inside the polymer matrix is increased, thereby
enhancing the force of water absorption The following factors affect the degradation of PLGA
(iv) Due to the presence of many repeating monomeric units in nanoparticles:
a row, crystallization takes place, thus inhibiting the absorp-
tion of water
9.1. Polymer composition and molecular weight

The effect of dissolved degradation products on the release of


The composition of the polymer significantly determines the hy-
the polymer ceases with the onset of rapid erosion. The transport
drophilicity and rate of degradation of any delivery matrix [50]. A
resistance controls the release of the encapsulated moiety and
significant loss in the weight of the polymer has been observed with
polymer degradation kinetics. Transport resistance is found to be
a substantial increase in the polymer’s glycolic content. The deg-
affected by various processes such as pore formation, pore closure,
radation rate of the polymer is directly affected by the amount of
drug dissolution, polymer–drug interaction and drug–drug inter-
glycolic acid present, and the degradation rate increases with in-
action [44]. It has also been found that an increase in the density
crease in the glycolic acid proportion. Due to higher hydrophilicity,
of the particle and reduction in the porosity (structural relax-
preferential degradation of glycolic acid proportion is seen, leading
ation) might result in a decreased burst release of the drug molecule
to faster degradation of PLGA 50:50 (PLA:PGA) than PLGA 65:35.
from the PLGA nanoparticles. The magnitude of drug release in a
Subsequently, PLGA 65:35 shows faster degradation than PLGA 75:25
particular formulation is directly affected by the rate and extent of
and PLGA 85:15. Generally, lower degradation rates are exhibited
structural relaxation present in the formulated nanoparticle. Struc-
by high molecular weight polymers. Polymers of high molecular
tural relaxation in PLGA nanoparticles is dependent on various
weight possess longer polymeric chains and require longer dura-
properties such as molecular weight, fabrication methods, drug–
tions for degradation, whereas polymers of low molecular weight
polymer interactions, residual solvents and storage conditions.
possess smaller polymeric chains and are degraded in a shorter
Together, these factors contribute to structural relaxation. They may
duration.
also lead to a variability in burst release, limiting the develop-
ment of products using this type of drug delivery technology.
9.2. Drug type

8. Drug release behaviour Drug type also determines the mechanistic fate of polymer–
drug matrix degradation and drug release rate. The mechanism of
For mechanistic evaluation, the release profile can be used as a degradation, as well as the rate of matrix degradation, may be
basic parameter. However, the most commonly preferred release changed from bulk erosion to surface degradation depending on the
profile is zero order and the monophasic release is rarely ob- type of encapsulated drug.
served. As a result of heterogeneous degradation, a biphasic or
triphasic release profile may be observed [48]. In the case of surface- 9.3. Size and shape of the matrix
coated PLGA nanoparticles, a biphasic release profile with a relatively
rapid second phase is observed. The following patterns are ob- The degradation profile of large devices is shown to be signifi-
served in the case of biphasic drug release from PLGA nanoparticles cantly affected by the ratio of the surface area to volume. Faster and
[49]: greater degradation of the matrix takes place with a higher surface
area ratio, but the opposite effect is seen in matrices or devices with
8.1. Phase I a smaller surface area to volume ratio. Bulk degradation takes place
faster than pure surface degradation, leading to faster release of the
Drug type, drug concentration and polymer hydrophobicity are drug from devices with higher surface area to volume ratios.
some of the parameters associated with an initial burst of drug
release. As soon as the nanoparticle comes in contact with the dis- 9.4. pH
solution medium, the water penetrates the polymer matrix rapidly.
The drug present on the surface of the nanoparticle is released as In vitro studies have shown that both strongly alkaline and acidic
a function of solubility. Random scission of the PLGA molecule occurs, media tend to accelerate polymer degradation. However, due to the
significantly decreasing the molecular weight of the polymer. autocatalysis of the polymer by the carboxylic end groups, the dif-
However, there is no appreciable weight loss and formation of a ference between slightly acidic and neutral media becomes less
soluble monomer product during this phase. pronounced [51].
S. Sharma et al./Trends in Analytical Chemistry 80 (2016) 30–40 37

9.5. Drug load are damaged, limiting the maximal permissible dose of the drug.
A large amount of drug must be administered, as some of the drug
The rate and duration of drug release is significantly affected by is distributed into the non-targeted organs and tissues, ultimately
the amount of drug loading in the drug delivery matrix. A larger leading to non-specific toxicity. This is followed by the rapid removal
initial burst release is seen in matrices with higher drug content, of the drug, making the treatment process costly and uneconomi-
whereas a smaller burst release is observed in the case of matri- cal. With the implementation of nanoparticle-mediated DDSs, these
ces with lower drug content because of their smaller polymer to limitations can be overcome [1]. In general, nanoparticle-mediated
drug ratio. Depending on the type of drug, the effect is attenuated targeting can be achieved via two targeting strategies: passive and
when a certain drug concentration is reached. active targeting;

10. PLGA-mediated drug delivery for cancer treatment 11.1. Passive targeting

Cancer treatment via the oral route is preferred because of its The size of nanoparticles offers an additional advantage. They
non-invasive nature and better patient compliance. However, due have the ability to extravasate and accumulate inside the intersti-
to their poor oral availability, most anticancer drugs cannot be de- tial spaces, thus contributing to enhanced permeability. Enhanced
livered via the oral route. When administered orally, only a small retention is observed because of the ineffective lymphatic vessels,
fraction of the drug becomes available to the systematic circula- leading to inefficient drainage of the tumour tissue [56]. Altogeth-
tion. For example, the oral bioavailability of paclitaxel, docetaxel and er, these two phenomena constitute the enhanced permeability and
doxorubicin has been found to be 1%, < 10% and < 5%, respectively retention (EPR) effect, which is considered to be a gold standard in
[52]. The underlying reason for this poor availability is the exten- designing effective anticancer DDSs.
sive first-pass metabolic effect by cytochrome P-450 (liver microsomal
enzyme) [44], as well as their efflux by an overexpressed plasma 11.2. Active targeting
membrane transporter P-gp efflux pump [53]. The P-gp is encoded
via the gene MDR-1, which acts as an efflux pump. After a pro- In active targeting, the ligands are grafted at the surface of the
longed chemotherapeutic session, the efflux pump exports a wide nanoparticle. The tumour cells are found to possess overexpressed
range of chemo drugs and thus tends to decrease the accumula- receptors, to which these ligands bind specifically. Improved cel-
tion of functional drugs in MDR cells. Ultimately, the body gradually lular internalization rather than an increased tumour accumulation
stops responding, decreasing the drugs’ therapeutic efficacy simul- has been implicated in the enhanced antitumoural efficacy of ac-
taneously followed by treatment failure [54]. The administration of tively targeted nanoparticles. In the case of active targeting, the
P-gp inhibitors based on the drug of interest can be an alternative following two cellular targeting strategies are used:
for overcoming this limitation. On the contrary, P-gP administra-
tion is generally associated with aberrant toxicity, blocking of 11.2.1. Targeting of cancer cells
physiological anticancer drug efflux from the normal cells and in- Internalization-prone cell surface receptors such as transferrin,
terrupted efflux of toxins from the body via the P-gP efflux pump. folate, integrins or epidermal growth factor receptor (EGFR) are
The small intestine contains two types of cells: enterocytes and M mainly overexpressed in cancer cells. Thus, active targeting acts as
cells. Any liquid or soluble material is absorbed in the small intes- an alternative pathway to improve cellular uptake. The ligand-
tine via the enterocytes directly from the systematic circulation, mediated approach allows the cancer cells to be killed directly,
whereas particulate matters are absorbed by the M cells via the lym- followed by generating a cytotoxic effect on cells present at the pe-
phatic system. In the case of orally administered PLGA nanoparticles, riphery of the tumour [57].
the absorption predominantly occurs via the M cells present on
Peyer’s patches and via the isolated follicles of the gut-associated 11.2.2. Targeting of tumour endothelium
lymphoid tissue (GALT). The efflux of drug by the P-gp efflux trans- Recognition of specific receptors such as vascular endothelial
porter can be effectively overcome by entrapping the desired growth factor receptor (VEGFR)-1 and VEGFR-2, integrins (αvβ3,
molecules within the voids of the PLGA matrices. The drug ab- α5β), vascular cell adhesion molecule (VCAM)-1 or matrix
sorbed from the enterocytes in the systematic circulation tends to metalloproteinases (MMPs) by targeting ligands facilitates effec-
undergo first-pass metabolism, whereas the drug absorbed into the tive targeting of the tumour endothelium [1]. Folkman et al. were
lymphatic system tends to bypass this [36]. Thus, the incorpora- the first to propose the rationale behind this targeting. It has been
tion of active agents within the polymer matrix of the PLGA prevents suggested that tumour growth can be inhibited by preventing an-
depletion of the drug in the hostile environment of the gastroin- giogenesis [58]. Due to lack of oxygen, the endothelium in tumour
testinal lumen. The small size and unique surface chemistry of PLGA cells is destroyed, ultimately leading to the death of tumour cells.
nanoparticles afforded them improved adhesion, absorption and However, the size and metastatic capabilities of tumours can be con-
transport of the drug. The absorption pathway of a particulate de- trolled by stopping the surge of blood supply. The tumour core is
livery vehicle occurs via M cells in Peyer’s patches. Encapsulation composed of angiogenic blood vessels, which in turn support the
with PLGA nanoparticles enhanced the solubility, stability and phar- tumour cells. The nanoparticle binds with and kills these angio-
macokinetics of chemo drugs [55]. The drug concentration can be genic blood vessels, thus indirectly killing tumour cells. Nevertheless,
controlled, thereby reducing the risk of unwanted side effects and the nanoparticle-mediated approach has many advantages [8]:
maintaining useful treatment cycles, without damaging the healthy
cells. (i) In order to direct these nanoparticles to the target site, no
extravagation of the nanocarrier is required.
11. Targeting strategies for efficient drug delivery (ii) When administered intravenously, these particles tend to bind
quickly to their receptor sites.
Chemo drugs have many advantages, but when administered (iii) The possible risk of emerging resistance is prevented, as the
inside the biological milieu they pose many potential hazards such endothelial cells are genetically more stable than the tumour
as systemic toxicity, bone marrow suppression, cardiomyopathy and cells.
neurotoxicity. This is because chemo drugs cannot differentiate (iv) The majority of endothelial cells are expressed in almost all
between normal and cancerous cells, and the healthy cells or tissues types of tumour, hence making this approach ubiquitous.
38 S. Sharma et al./Trends in Analytical Chemistry 80 (2016) 30–40

12. Ligand-anchored PLGA nanoparticles for cancer therapy been proven to enhance the anticancer effect of the entrapped drug
by facilitating cellular uptake and intracellular retention of the
Cancer cells, unlike normal cells, have an innate tendency to pro- nanodrug carriers, in turn augmenting their antitumour efficacy [36].
liferate rapidly, supported by some overexposed receptors present A comprehensive overview of different ligands anchored or conju-
on the surface of the tumour cell. These receptors allow the uptake gated to PLGA nanoparticles is provided in Table 2 [60–62].
of growth factor via receptor-mediated endocytosis. This mecha-
nism can be used as a Trojan horse for site-specific delivery of 13. PLGA nanoparticles as thriving mediators
anticancer agents. The surface of the nanoparticles is decorated with
ligands such as antibodies, which tend to bind specifically with these 13.1. Gene delivery for cancer treatment
receptors [36]. The desired ligands can be attached to the surface
of the nanoparticles via simple physical associations or conjuga- Currently, gene therapy is considered one of the most promis-
tion reactions. ing tool for targeted drug delivery in cancer treatment. This technique
Carbodiimide chemistry is most commonly used for conjuga- can be used to treat a variety of infectious diseases such as mono-
tion reactions. Nanoparticles can be surface-derivatized via genic diseases and cancer [63]. When travelling from outside of the
conjugation reactions. A water-soluble carbodiimide reagent such cells to the cellular milieu, a gene or macromolecule faces many chal-
as 1-ethyl-3-(3-dimethylaminopropyl carbodiimide) (EDC)/N,N- lenges such as poor permeability, membrane non-selectivity and
dicyclohexylcarbodiimide (DCC) is allowed to react with a carboxyl degradation in the endo-lysosomal environment. To circumvent these
group present in the PLGA, leading to the formation of an amine- obstacles, non-viral novel vectors have been proposed.
reactive O-acylisourea intermediate. This intermediate reacts with PLGA nanoparticles have been explored as a delivery vehicle for
the amine group present in the ligand, resulting in the formation DNA, so that the target gene is effortlessly transfected with the cancer
of a PLGA–ligand conjugate. However, this reaction tends to lead cells. For this purpose, the surface properties of PLGA nanoparticles
to other unwanted secondary reactions. To avoid any further reac- such as surface charge or coating must be tailored as efficient car-
tion, N-hydroxysuccinimide (NHS) is added to the reaction mixture, riers for DNA transfer into cancer cells.36 Gene silencing via siRNA
and the amine-reactive intermediate thus formed early in the re- is currently the fastest growing sector of the antigene field for target
action is transformed into an NHS-ester derivative. The ester validation and therapeutic applications. However, the systematic
derivative immediately reacts with any primary amine group present deliverability of siRNA into target cells is often limited by their rapid
in the reaction mixture, along with the liberation of NHS, ultimate- degradation and poor cell penetration ability. To overcome these
ly leaving behind the PLGA–ligand conjugate. Importantly, the limitations, a broad spectrum of viral and non-viral vectors has been
conjugation reaction should proceed in an aqueous environment, successful in improving the targeted delivery of siRNA to cancer cells
as some portions of the free carboxylic end group remain embed- and protecting them from premature degradation in the biologi-
ded in the PLGA nanoparticle, which can limit availability for direct cal milieu. PLGA-based systems have also been investigated for the
conjugation. To overcome this situation, PLGA is dissolved in an targeted delivery of siRNA to cancer cells and for induction of gene
organic solvent such as dimethyl formamide, prior to the conjuga- silencing. Studies on the delivery of DNA or a specific gene via PLGA
tion reaction [59]. The strong conjugation reactions can be avoided nanoparticles are listed in Table 3 [64–66].
by using other ligands, for example, non-covalent binding of biotin–
PEG–NH2 with an avidin-functionalized PLGA nanoparticle. Ligands 13.2. Diagnosis and imaging of cancer
such as antibodies and Fab fragments can be attached to PLGA
nanoparticles. In the field of clinical oncology, tumour imaging plays a key role,
Substantial research has been conducted on ligand-mediated as it helps determine the recurrence of solid tumours along with
PLGA nanoparticles, but certain drawbacks have limited the use of monitoring the therapeutic responses. The currently available clin-
these engineered nanoparticles in practical situations. In vivo studies ical diagnostic methods are unable to detect cancer in the early stages
have revealed that a significant amount of the injected dose is ac- [1]. However, developing a non-invasive molecular imaging system
cumulated in various organs of the RES such as the liver and spleen. might allow the detection of tumours at an early stage. Recent ad-
This is undesirable as the anticancer drugs may damage the MPS vancements in nanoparticles have facilitated the use of contrast
organs. The clearance time is increased as these targeted agents for imaging [23]. Wang et al. prepared supramagnetic iron
nanoparticles are recognized by the MPS. Despite the limitations oxide-loaded PLGA nanoparticles for magnetic resonance imaging
of the ligand-targeted approach, ligand-targeted nanoparticles have (MRI) [67]. The imaging effects are enhanced along with an increase

Table 2
Ligand-targeted PLGA nanoparticles for cancer therapeutics

Nanoparticle Drug loaded Ligand Target site Inference Reference

PLGA Paclitaxel ScFv antibody Hepatic cancer Improved cellular cytotoxicity against Chepp-3 cells [60]
PLGA–PEG Cisplatin PSMA targeting aptamer Prostate The NPs were readily taken up by receptor mediated endocytosis [61]
PLGA–PEG Paclitaxel RGDp Human umbilical vein Enhanced uptake by integrin expressing malignant cells [62]
endothelial cells

Table 3
PLGA nanoparticles for gene delivery

Nanoparticles Gene Delivered Target Inference Reference


th
PLGA nanoparticles DNA COS-7 and Cf2 cells Localization into the endo-lysosomal compartment, [64]
250-fold protein expression in cells
Chitosan–PLGA nanoparticles Antisense oligonucleotides DNA/RNA Lung cancer cells Efficient delivery of antisense oligonucleotides [65]
DEPA–PVA–PLGA nanoparticles Anti-luciferase siRNA H1299 cell line 80–90% knockdown of the luciferase reporter gene [66]
S. Sharma et al./Trends in Analytical Chemistry 80 (2016) 30–40 39

in the half-life of nanoparticles in the bloodstream, with reduced 15. Conclusion


severity of side effects. In another experiment, Acharya et al. de-
tected sentinel lymph node of Wistar rats by encapsulating a Despite the several advantages of PLGA nanoparticle-based DDSs,
radiotracer, 99mTC, in PLGA nanoparticles [23]. these systems have their limitations as well. A relatively low drug
loading efficiency significantly limits the use of drug-loaded PLGA
13.3. Theranostics of cancer nanoparticles in clinical trials. The preparation of polymeric
nanoparticles is a state-of-the-art technology requiring a suitable
Theranostics collectively describes the therapeutic and diag- protocol, a thorough homogenization process, appropriate surfac-
nostic agents. It may combine passive and active targeting, envi- tants and creditable co-surfactant to obtain the desired polymeric
ronmentally responsive drug release, molecular imaging and other nanoparticles with optimal properties. Future research should be
therapeutic functions under a single platform [68]. For example, mag- directed towards developing techniques that can precisely control
netic nanoparticles containing doxorubicin were further encapsulated the particle size and morphology, which are considered factors de-
in PLGA nanoparticles via hydrophobic interactions. In another study, termining the applications of these nanoparticles. Further
PLGA-containing magnetic nanoparticles were designed with the investigation is needed to better control nanoparticle size, polydis-
dual function of drug delivery and imaging [69]. MRI scans have persity and charge surface, and to render these features easily
shown that these nanoparticles offer a better contrast than the com- reproducible during synthesis steps. The limitations of using PLGA
mercially available contrast agents. nanoparticles can be overcome through an extensive and thor-
ough evaluation of the pharmacokinetics, biodistribution and toxicity.
14. Drawbacks of using PLGA nanoparticle-based DDSs PLGA degradation and drug release rate can be increased with greater
hydrophilicity, increase in chemical interactions among the hydro-
Apart from developmental and nanoethical aspects, PLGA lytic groups, less crystallinity and larger volume to surface ratio of
nanoparticle-based DDSs have certain drawbacks: the device. All of these factors should be considered to tailor the
degradation and drug release mechanism as needed. Proper selec-
(i) One of the major drawbacks is that the EPR is often misun- tion of the preparation method, formulation/reaction variables and
derstood. It is a heterogeneous phenomenon that varies appropriate scale-up techniques/methods, combined with coordi-
substantially from model to model, and from patient to nated efforts from the industrial and academic sectors, can lead to
patient. Moreover, the potential efficacy of active drug tar- successful commercialization of engineered PLGA nanoparticles. Nev-
geting is overestimated sometimes. In theory, targeted ertheless, the prospects of putting drug-loaded PLGA NP technology
nanoparticles are retained more efficiently and rapidly than into practice are exciting as the next generation of DDSs.
non-targeted nanoparticles. However, an increase in immu-
nogenicity and protein adsorption is often noted with the Acknowledgement
introduction of targeting moieties [56].
(ii) Although PLGA-based nanoparticles offer advantages such as The authors are grateful to DST, Government of India, for pro-
high encapsulation efficiency in comparison to other viding financial support to IFSC through the PURSE grant.
nanoparticle-based DDS, they exhibit poor drug loading, which
might prove problematic for some drugs [8].
References
(iii) High burst release rate of the drug from nanoparticles is often
accompanied by the DDS. Thus, the efficacy of the DDS is lost,
[1] F. Danhier, E. Ansorena, J.M. Silva, R. Coco, A. Le, V. Breton, et al., based
followed by a non-targeted delivery of the payload or drug. nanoparticles: an overview of biomedical applications, J. Control. Release 161
(iv) The degradation of the PLGA polymer leads to the forma- (2012) 505–522.
tion of acids that limit the drug’s therapeutic efficacy. Drug [2] Y. Liu, H. Miyoshi, M. Nakamura, Nanomedicine for drug delivery and imaging:
a promising avenue for cancer therapy and diagnosis using targeted functional
delivery via PLGA nanoparticles as the encapsulant is not fea- nanoparticles, Int. J. Cancer 120 (2007) 2527–2537.
sible if they are pH sensitive. [3] S.A. Wickline, A.M. Neubauer, P.M. Winter, S.D. Caruthers, G.M. Lanza, Molecular
(v) The local tissue reactions occur at the site of application, which imaging and therapy of atherosclerosis with targeted nanoparticles, J. Magn.
Reson. Imaging 25 (2007) 667–680.
is another cause for concern. Studies have also suggested that
[4] R. Langer, J. Folkman, Polymers for the sustained release of proteins and other
specific biodistribution and toxicological profiles emerge when macromolecules, Nature 263 (1976) 797–800.
nanoparticles of any material are used for therapeutic ap- [5] D. Dragan, S. Magdalena, Poly(lactide-co-glycolide)-based micro and
plication [70]. nanoparticles for the controlled drug delivery of vitamins, Curr. Nanosci. 5
(2009) 1–14.
(vi) Relatively poor drug loading, higher cost of production and [6] J.-S. Choi, K. Seo, J.-W. Yoo, Recent advances in PLGA particulate systems for
difficulty in scaling up are some of the other limitations to drug delivery, J. Pharm. Investig. 42 (2012) 155–163.
the use of PLGA nanoparticles in clinical trials. [7] E. Locatelli, M.C. Franchini, Biodegradable PLGA-b-PEG polymeric nanoparticles:
synthesis, properties, and nanomedical applications as drug delivery systems,
(vii) Current knowledge on the effect of various process param- J. Nanoparticle Res. 14 (2012).
eters on the preparation of nanoparticles is still limited [27]. [8] F.S.T. Mirakabad, K. Nejati-Koshki, A. Akbarzadeh, M.R. Yamchi, M. Milani, N.
(viii) The particle size and morphology of the nanoparticles are af- Zarghami, et al., PLGA-based nanoparticles as cancer drug delivery systems,
Asian Pac. J. Cancer Prev. 15 (2014) 517–535.
fected by two major factors: homogenization conditions and [9] H.K. Makadia, S.J. Siegel, Poly Lactic-co-Glycolic acid (PLGA) as biodegradable
droplet size distribution. A thorough understanding of these controlled drug delivery carrier, Polymers (Basel) 3 (2011) 1377–1397.
parameters is still lacking. [10] A. Prokop, J.M. Davidson, Nanovehicular intracellular delivery systems, J. Pharm.
Sci. 97 (2008) 3518–3590.
(ix) During in vivo application, heterogeneous expression levels [11] G. Schliecker, C. Schmidt, S. Fuchs, T. Kissel, Characterization of a homologous
of the targeted receptor in cancer cells also pose a chal- series of D,L-lactic acid oligomers; a mechanistic study on the degradation
lenge. Various physiological barriers prevent these kinetics in vitro, Biomaterials 24 (2003) 3835–3844.
[12] S. Acharya, S.K. Sahoo, PLGA nanoparticles containing various anticancer agents
nanoparticles from reaching the target tissues.
and tumour delivery by EPR effect, Adv. Drug Deliv. Rev. 63 (2011) 170–183.
(x) The in vivo behaviour of these nanoparticles has not been [13] S.J. Siegel, J.B. Kahn, K. Metzger, K.I. Winey, K. Werner, N. Dan, Effect of drug
completely characterized [59]. type on the degradation rate of PLGA matrices, Eur. J. Pharm. Biopharm. 64
(xi) Due to the complexity of drug release from PLGA-based (2006) 287–293.
[14] N. Passerini, D.Q. Craig, An investigation into the effects of residual water on
DDSs, the results obtained with a specific DDS cannot be the glass transition temperature of polylactide microspheres using modulated
generalized [44]. temperature DSC, J. Control. Release 73 (2001) 111–115.
40 S. Sharma et al./Trends in Analytical Chemistry 80 (2016) 30–40

[15] Z. Panagi, A. Beletsi, G. Evangelatos, E. Livaniou, D.S. Ithakissios, K. Avgoustakis, [43] T. Betancourt, J.D. Byrne, N. Sunaryo, S.W. Crowder, M. Kadapakkam, S. Patel,
Effect of dose on the biodistribution and pharmacokinetics of PLGA and et al., PEGylation strategies for active targeting of PLA/PLGA nanoparticles,
PLGA-mPEG nanoparticles, Int. J. Pharm. 221 (2001) 143–152. J. Biomed. Mater. Res. A 91 (2009) 263–276.
[16] R.A. Jain, The manufacturing techniques of various drug loaded biodegradable [44] S. Fredenberg, M. Wahlgren, M. Reslow, A. Axelsson, The mechanisms of drug
poly(lactide-co-glycolide) (PLGA) devices, Biomaterials 21 (2000) 2475–2490. release in poly(lactic-co-glycolic acid)-based drug delivery systems–a review,
[17] S. Mao, J. Xu, C. Cai, O. Germershaus, A. Schaper, T. Kissel, Effect of WOW process Int. J. Pharm. 415 (2011) 34–52.
parameters on morphology and burst release of FITC-dextran loaded PLGA [45] M. Marucci, Characterization of the mechanisms of drug release from polymer-
microspheres, Int. J. Pharm. 334 (2007) 137–148. coated formulations using experiments and modelling., Doctoral Dissertation.
[18] F.J. Hua, T.G. Park, D.S. Lee, facile preparation of highly interconnected Lund University, Lund, Sweden, 2009.
macroporous PLGA scaffolds by liquid-liquid phase separation of a PLGA- [46] A. Shenderova, T.G. Burke, S.P. Schwendeman, The acidic microclimate in
dioxane-water ternary system, Polymer (Guildf.) 44 (2003) 1911–1920. poly(lactide-co-glycolide) microspheres stabilizes camptothecins, Pharm. Res.
[19] S.R. D’Mello, S.K. Das, N.G. Das, Polymeric nanoparticles for small-molecule 16 (1999) 241–248.
drugs: biodegradation of polymers and fabrication of nanoparticles. Drug [47] M. Dunne, I. Corrigan, Z. Ramtoola, Influence of particle size and dissolution
Delivery Nanoparticles Formulation and Characterization, 2006. conditions on the degradation properties of polylactide-co-glycolide particles,
[20] G. Lambert, E. Fattal, P. Couvreur, Nanoparticulate systems for the delivery of Biomaterials 21 (2000) 1659–1668.
antisense oligonucleotides, Adv. Drug Deliv. Rev. 47 (2001) 99–112. [48] N.S. Berchane, K.H. Carson, A.C. Rice-Ficht, M.J. Andrews, Effect of mean diameter
[21] H. Fessi, F. Puisieux, J.P. Devissaguet, N. Ammoury, S. Benita, Nanocapsule and polydispersity of PLG microspheres on drug release: experiment and theory,
formation by interfacial polymer deposition following solvent displacement, Int. J. Pharm. 337 (2007) 118–126.
Int. J. Pharm. 55 (1989) R1–R4. [49] L.C. Amann, M.J. Gandal, R. Lin, Y. Liang, S.J. Siegel, In vitro-in vivo correlations
[22] T. Govender, S. Stolnik, M.C. Garnett, L. Illum, S.S. Davis, PLGA nanoparticles of scalable PLGA-risperidone implants for the treatment of schizophrenia,
prepared by nanoprecipitation: drug loading and release studies of a water Pharm. Res. 27 (2010) 1730–1737.
soluble drug, J. Control. Release 57 (1999) 171–185. [50] L. Lu, S.J. Peter, M.D. Lyman, H.L. Lai, S.M. Leite, J.A. Tamada, et al., In vitro and
[23] Y.-I. Jeong, C.-S. Cho, S.-H. Kim, K.-S. Ko, S.-I. Kim, Y.-H. Shim, et al., Preparation in vivo degradation of porous poly(DL-lactic-co-glycolic acid) foams,
of poly(dl-lactide-co-glycolide) nanoparticles without surfactant, J. Appl. Polym. Biomaterials 21 (2000) 1837–1845.
Sci. 80 (2001) 2228–2236. [51] B.S. Zolnik, D.J. Burgess, Effect of acidic pH on PLGA microsphere degradation
[24] M. Kostog, S. Kohler, T. Liebert, T. Heinze, Pure cellulose nanoparticles from and release, J. Control. Release 122 (2007) 338–344.
trimethylsilyl cellulose, Macromol. Symp. 294 (2) (2010) 96–106. [52] I.E. Kuppens, T.M. Bosch, M.J. van Maanen, H. Rosing, A. Fitzpatrick, J.H. Beijnen,
[25] H. Nie, L.Y. Lee, H. Tong, C.H. Wang, PLGA/chitosan composites from a et al., Oral bioavailability of docetaxel in combination with OC144-093
combination of spray drying and supercritical fluid foaming techniques: new (ONT-093), Cancer Chemother. Pharmacol. 55 (2005) 72–78.
carriers for DNA delivery, J. Control. Release 129 (2008) 207–214. [53] M. Hennessy, J.P. Spiers, A primer on the mechanics of P-glycoprotein the
[26] K. Mishima, Biodegradable particle formation for drug and gene delivery using multidrug transporter, Pharmacol. Res. 55 (2007) 1–15.
supercritical fluid and dense gas, Adv. Drug Deliv. Rev. 60 (2008) 411–432. [54] B. Li, H. Xu, Z. Li, M. Yao, M. Xie, H. Shen, et al., Bypassing multidrug resistance
[27] J.P. Rao, K.E. Geckeler, Polymer nanoparticles: Preparation techniques and in human breast cancer cells with lipid/polymer particle assemblies, Int. J.
size-control parameters, Elsevier 36 (2011) 887–913. Nanomedicine 7 (2012) 187–197.
[28] Y.P. Sun, H.W. Rolling, J. Bandara, J.M. Meziani, C.E. Bunker, Preparation and [55] R. Langer, Drug delivery and targeting, Nature 392 (1998) 5–10.
processing of nanoscale materials by supercritical fluid technology, in: Y.P. Sun [56] F. Danhier, O. Feron, V. Preat, To exploit the tumor microenvironment: Passive
(Editor), Supercritical Fluid Technology in Materials Science and Engineering: and active tumor targeting of nanocarriers for anti-cancer drug delivery,
Synthesis, Properties, and Applications, Marcel Dekker, New York, 2002, pp. J. Control. Release 148 (2010) 135–146.
491–576. [57] F. Pastorino, C. Brignole, D. Di Paolo, B. Nico, A. Pezzolo, D. Marimpietri, et al.,
[29] G. Mittal, D.K. Sahana, V. Bhardwaj, M.N. Ravi Kumar, Estradiol loaded PLGA Targeting liposomal chemotherapy via both tumor cell-specific and tumor
nanoparticles for oral administration: effect of polymer molecular weight and vasculature-specific ligands potentiates therapeutic efficacy, Cancer Res. 66
copolymer composition on release behavior in vitro and in vivo, J. Control. (2006) 10073–10082.
Release 119 (2007) 77–85. [58] J. Folkman, Transplacental carcinogenesis by stilbestrol, N. Engl. J. Med. 285
[30] M. Garinot, V. Fievez, V. Pourcelle, F. Stoffelbach, A. des Rieux, L. Plapied, et al., (1971) 404–405.
PEGylated PLGA-based nanoparticles targeting M cells for oral vaccination, [59] H. Sah, L.A. Thoma, H.R. Desu, E. Sah, G.C. Wood, Concepts and practices used
J. Control. Release 120 (2007) 195–204. to develop functional PLGA-based nanoparticulate systems, Int. J. Nanomedicine
[31] F. Esmaeili, M.H. Ghahremani, B. Esmaeili, M.R. Khoshayand, F. Atyabi, R. 8 (2013) 747–765.
Dinarvand, PLGA nanoparticles of different surface properties: preparation and [60] G. Kou, J. Gao, H. Wang, H. Chen, B. Li, D. Zhang, et al., Preparation and
evaluation of their body distribution, Int. J. Pharm. 349 (2008) 249–255. characterization of paclitaxel-loaded PLGA nanoparticles coated with cationic
[32] I. Bala, S. Hariharan, M.N. Kumar, PLGA nanoparticles in drug delivery: the state SM5-1 single-chain antibody, J. Biochem. Mol. Biol. 40 (2007) 731–739.
of the art, Crit. Rev. Ther. Drug Carrier Syst. 21 (2004) 387–422. [61] S. Dhar, F.X. Gu, R. Langer, O.C. Farokhzad, S.J. Lippard, Targeted delivery
[33] Z. Zhang, S.S. Feng, The drug encapsulation efficiency, in vitro drug release, of cisplatin to prostate cancer cells by aptamer functionalized Pt(IV)
cellular uptake and cytotoxicity of paclitaxel-loaded poly(lactide)-tocopheryl prodrug-PLGA-PEG nanoparticles, Proc. Natl. Acad. Sci. U.S.A. 105 (2008)
polyethylene glycol succinate nanoparticles, Biomaterials 27 (2006) 4025–4033. 17356–17361.
[34] A. Yang, L. Yang, W. Liu, Z. Li, H. Xu, X. Yang, Tumor necrosis factor alpha blocking [62] F. Danhier, B. Vroman, N. Lecouturier, N. Crokart, V. Pourcelle, H. Freichels, et al.,
peptide loaded PEG-PLGA nanoparticles: preparation and in vitro evaluation, Targeting of tumor endothelium by RGD-grafted PLGA-nanoparticles loaded
Int. J. Pharm. 331 (2007) 123–132. with paclitaxel, J. Control. Release 140 (2009) 166–173.
[35] A. Kumari, S.K. Yadav, S.C. Yadav, Biodegradable polymeric nanoparticles based [63] S. Diez, G. Navarro, C.T. de ILarduya, In vivo targeted gene delivery by cationic
drug delivery systems, Colloids Surf. B. Biointerfaces 75 (2010) 1–18. nanoparticles for treatment of hepatocellular carcinoma, J. Gene Med. 11 (2009)
[36] A.K. Jain, M. Das, N.K. Swarnakar, S. Jain, Engineered PLGA nanoparticles: 38–45.
an emerging delivery tool in cancer therapeutics, Crit. Rev. Ther. Drug Carrier [64] K. Gvili, O. Benny, D. Danino, M. Machluf, Poly(D,L-lactide-co-glycolide acid)
Syst. 28 (2011) 1–45. nanoparticles for DNA delivery: waiving preparation complexity and increasing
[37] W. Lu, Y.Z. Tan, K.L. Hu, X.G. Jiang, Cationic albumin conjugated pegylated efficiency, Biopolymers 85 (2007) 379–391.
nanoparticle with its transcytosis ability and little toxicity against blood-brain [65] N. Nafee, S. Taetz, M. Schneider, U.F. Schaefer, C.M. Lehr, Chitosan-coated PLGA
barrier, Int. J. Pharm. 295 (2005) 247–260. nanoparticles for DNA/RNA delivery: effect of the formulation parameters on
[38] R. Gref, M. Luck, P. Quellec, M. Marchand, E. Dellacherie, S. Harnisch, et al., complexation and transfection of antisense oligonucleotides, Nanomedicine 3
‘Stealth’ corona-core nanoparticles surface modified by polyethylene glycol (2007) 173–183.
(PEG): influences of the corona (PEG chain length and surface density) and of [66] J. Nguyen, T.W. Steele, O. Merkel, R. Reul, T. Kissel, Fast degrading polyesters
the core composition on phagocytic uptake and plasma protein adsorption, as siRNA nano-carriers- for pulmonary gene therapy, J. Control Rel. 132 (2008)
Colloids Surf. B. Biointerfaces 18 (2000) 301–313. 243–251.
[39] F. Danhier, N. Lecouturier, B. Vroman, C. Jerome, J. Marchand-Brynaert, O. Feron, [67] Y.G.N.Y. Wang, Y. Chen, B. Shuter, J. Yi, J. Ding, S. Wang, et al., Formulation of
et al., Paclitaxel-loaded PEGylated PLGA-based nanoparticles: in vitro and in superparamagnetic iron oxides by nanoparticles of biodegradable polymers for
vivo evaluation, J. Control. Release 133 (2009) 11–17. magnetic resonance imaging, Adv. Funct. Mater. 18 (2008) 308–318.
[40] R. Gref, Y. Minamitake, M.T. Peracchia, V. Trubetskoy, V. Torchilin, R. Langer, [68] S.M. Janib, A.S. Moses, J.A. MacKay, Imaging and drug delivery using theranostic
Biodegradable long-circulating polymeric nanospheres, Science 263 (1994) nanoparticles, Adv. Drug Deliv. Rev. 62 (2010) 1052–1063.
1600–1603. [69] A. Singh, F. Dilnawaz, S. Mewar, U. Sharma, N.R. Jagannathan, S.K. Sahoo,
[41] E.C. Gryparis, M. Hatziapostolou, E. Papadimitriou, K. Avgoustakis, Anticancer Composite polymeric magnetic nanoparticles for co-delivery of hydrophobic
activity of cisplatin-loaded PLGA-mPEG nanoparticles on LNCaP prostate cancer and hydrophilic anticancer drugs and MRI imaging for cancer therapy, ACS Appl.
cells, Eur. J. Pharm. Biopharm. 67 (2007) 1–8. Mater. Interfaces 3 (2011) 842–856.
[42] M. Senthilkumar, P. Mishra, N.K. Jain, Long circulating PEGylated poly(D,L- [70] L.A. Dailey, N. Jekel, L. Fink, T. Gessler, T. Schmehl, M. Wittmar, et al.,
lactide-co-glycolide) nanoparticulate delivery of Docetaxel to solid tumors, Investigation of the proinflammatory potential of biodegradable nanoparticle
J. Drug Target. 16 (2008) 424–435. drug delivery systems in the lung, Toxicol. Appl. Pharmacol. 215 (2006) 100–108.

You might also like