You are on page 1of 19

bs_bs_banner

Journal of Pharmacy
Review
And Pharmacology

Engineered PLGA nano- and micro-carriers for pulmonary


delivery: challenges and promises
Francesca Ungaro, Ivana d’ Angelo, Agnese Miro, Maria I. La Rotonda and Fabiana Quaglia
Department of Pharmaceutical and Toxicological Chemistry, University of Naples Federico II, Via D. Montesano, Naples, Italy

Keywords Abstract
microparticle; nanoparticle;
poly(lactic-co-glycolic acid); pulmonary delivery Objectives The aim of this review is to summarize the current state-of-the-art in
poly(lactic-co-glycolic acid) (PLGA) carriers for inhalation. It presents the rational
Correspondence of use, the potential and the recent advances in developing PLGA microparticles and
Francesca Ungaro, Department of
nanoparticles for pulmonary delivery. The most promising particle engineering
Pharmaceutical and Toxicological Chemistry,
University of Naples Federico II, Via D.
strategies are discussed, highlighting the advantages along with the major challenges
Montesano 49, 80131 Naples, Italy. for researchers working in this field.
E-mail: ungaro@unina.it Key findings Biodegradable polymer carriers, such as PLGA particles, may permit
effective protection and long-term delivery of the inhaled drug and, when
Received October 15, 2011 adequately engineered, its efficient transport to the target. The carrier can be
Accepted January 23, 2012 designed for inhalation on the basis of several strategies through the adequate com-
bination of available particle technologies and excipients. In so doing, the properties
doi: 10.1111/j.2042-7158.2012.01486.x
of PLGA particles can be finely tuned at micro-size and nano-size level to fulfill spe-
cific therapeutic needs. This means not only to realize optimal in vitro/in vivo lung
deposition of the formulation, which is still crucial, but also to control the fate of the
drug in the lung after particle landing.
Summary Although many challenges still exist, PLGA carriers may be highly ben-
eficial and present a new scenario for patients suffering from chronic lung diseases
and for pharmaceutical companies working to develop novel inhaled products.

Introduction
Drug inhalation has a long and rich history that can be traced tant, as many inhaled drugs are developed either to be
back to the writings of many ancient civilizations, already absorbed systemically (e.g. peptides and proteins)[5] or
employing the pulmonary route of administration to deliver to exert their pharmacological effect in this region of
natural remedies either locally or to the body through the the lung (e.g. antitubercular drugs directed to alveolar
lung. If in the past inhaling ‘vapours’ (Egyptians in 1500) or macrophages).[6]
smoking cigarettes (Potter’s cigarettes in 1802) was sufficient, Although particle ‘landing’ at the site of interest remains
with the rapidly growing popularity and sophistication of crucial in determining the therapeutic efficacy of inhaled
therapies, there is an increasing demand for tailor-made therapeutics, lung retention and the ability to overcome
inhalable drug formulations able to yield the best therapeutic extracellular and cellular lung barriers is just as significant.[7]
outcomes via the most efficient delivery to the lungs.[1] Mucociliary clearance, designed by evolution to eliminate
It is well established that key features of the formulation inhaled and possibly noxious material from the airways, may
(e.g., aerodynamic size, flow and aerosolization properties) considerably limit the residence time of the deposited formu-
will affect the likelihood of it being deposited in the desired lation and, subsequently, the amount of drug reaching the
region of the lung.[2] In the case of inhaled drugs required target. Analogously, the nature and extent of the interactions
for the treatment of lung diseases, such as asthma, chronic of the drug with lung lining fluid (LLF), airway macrophages
obstructive pulmonary disease (COPD) and cystic fibrosis and lung epithelial cells will inevitably affect drug perma-
(CF), an effective ‘local’ delivery (i.e. centrally rather than at nence in situ and duration of the effect.
the periphery) of the drug is necessary.[3,4] Strategies for tar- To overcome these drawbacks, controlled and targeted
geting drugs to the alveoli have become increasingly impor- delivery of drugs to the lungs by means of polymeric drug

© 2012 The Authors. JPP © 2012


Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235 1217
PLGA carriers for inhalation Francesca Ungaro et al.

30 potential in the inhalation field. After a brief description of


Research papers (No. identified in PubMed)

the rationale behind the choice of polymer carriers for drug


inhalation, it describes the recent advances in developing
25
PLGA microparticles and nanoparticles for pulmonary deliv-
ery. The most promising particle engineering strategies are
20 discussed, highlighting the advantages along with the major
challenges for researchers working in this field.
15
Why a polymer carrier for inhalation?
10 Nowadays, drugs are administered via the pulmonary route
for two main purposes: (i) local therapies; (ii) systemic
5 absorption. It is currently believed that drug delivery into the
human lung represents the best way of treating pulmonary
diseases.[2] Inhalation allows direct delivery of the drug to the
0
site of action, with fewer systemic effects than oral therapy.
97
98
99
01
02
03
04
05
06
07
08
09
10
11 Meanwhile, the lungs may be used as a portal of entry to the
19
19
19
20
20
20
20
20
20
20
20
20
20
20

Year body, allowing systemic delivery of drugs via the airway sur-
faces into the bloodstream.[5] This is an attractive prospect
Figure 1 Research studies on poly(lactic-co-glycolic acid) carriers for owing to the large surface area of the alveolar region, thick-
pulmonary delivery identified in PubMed databases from 1997 to 2011.
ness of the epithelial barrier, extensive vascularization,
relatively low proteolytic activity in the alveolar space and
absence of first-pass metabolism.
delivery systems (DDS) is under investigation.[1,8] As a matter As can be seen in Table 1, locally acting drugs for inhalation
of fact, DDS may offer substantial advantages over conven- are mainly bronchodilators, corticosteroids and antibiotics,
tional dosage forms for inhalation, including long-term which can reverse constriction of bronchial airways and
maintenance of drug concentrations within a desired thera- control airway inflammation/infection. Inhaled products
peutic range, reduced doses, reduced dosing frequency, the administered for their systemic effects mainly include fast-
potential for limited side effects and, last but not least, onset analgesics, peptides and proteins, which would other-
improved patient adherence to the therapy. This would repre- wise need to be given by injection. In both cases, to yield the
sent a real benefit for those patients suffering from chronic best therapeutic outcomes, the drug has to effectively deposit
lung diseases. along the airways, remain in situ as long as possible, overcome
Despite encouraging premises, the development of safe extracellular and cellular airway barriers and, when needed,
and effective polymeric DDS for inhalation faces many chal- reach intracellular targets. This is the case of the emerging oli-
lenges, among which the choice of the right polymer excipi- gonucleotide therapies for chronic respiratory diseases, able
ent[9] and its adequate engineering in the form of inhalable to reach cell targets underlying disease pathogenesis.[18]
particles are primary.[10] In the 1990s, Edwards and colleagues Indeed, formulating biopharmaceuticals for inhalation pre-
developed a new type of inhalation aerosol based on sents new challenges to scientists. The structural complexity
poly(lactic-co-glycolic acid) (PLGA), characterized by par- of proteins and nucleic acids compared with conventional
ticles of small mass density and large size, which permitted drugs demands an efficient drug-delivery system that allows
efficient and prolonged delivery of insulin into the systemic the intact macromolecule to gain access to the target site at
circulation.[11] This was the first attempt to apply PLGA-based the right time and for proper duration.[38]
carriers to lung delivery. After almost 10 years, we are experi- Lung deposition results from the complex interplay of a
encing the revival of the research interest in biodegradable range of factors, which relate to the size of the particles, the
particles for drug inhalation (Figure 1). Nonetheless, it is inhalation device, the mode of inhalation or breathing
apparent from the current ‘manuscript boom’ that it is pattern and even to the lung anatomy of the individual.[2,39,40]
growing and presents a future challenge. In particular, the aerodynamic particle size, defined as the
Excellent reviews have recently summarized the new diameter of a sphere of unit density characterized by the same
concept of carrier-mediated lung targeting,[12,13] and the most settling velocity in air as the particle aerosol being measured,
interesting techniques to engineer particles for inhala- will critically influence the deposition of the inhaled formu-
tion.[10,14,15] Nonetheless, these works only mention biode- lation in the upper airways, rather than the conducting and
gradable PLGA particles among a huge range of technological the alveolated airways of the lungs.[2,40,41] It is well recognized
strategies. This review focuses on PLGA carriers and their that particles below 5 mm can be distributed deep into the

© 2012 The Authors. JPP © 2012


1218 Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235
Francesca Ungaro et al. PLGA carriers for inhalation

Table 1 Drugs marketed or undergoing preclinical/clinical studies by the pulmonary route

Current
Therapeutic development
application Drug class Drug Inhalation device* status**
[16,17]
Local delivery Asthma Short-acting b2 agonist Salbutamol (albuterol) Nebulizer, pMDI, DPI Marketed
Fenoterol pMDI
Pirbuterol Nebulizer, pMDI
Long-acting b2 agonist[16,17] Salmeterol pMDI, DPI Marketed
Formeterol DPI
Anticholinergic Ipratropium bromide Nebulizer, pMDI Marketed
compounds[16,17] Tiotropium bromide pMDI, DPI
Inhaled corticosteroids[16,17] Beclomethasone dipropionate Nebulizer, pMDI Marketed
Budesonide Nebulizer, DPI
Fluticasone propionate pMDI, DPI
COPD
Antisense EPI-2010 Nebulizer Terminated
oligonucleotides [18,19] AIR-645 Phase II
PXSTPI-1100 Preclinical
ATL-1102 Preclinical
CpG oligonucleotides [18,19] AVE-7279 Nebulizer Terminated
QAX-935 (IMO-2134) Phase I
siRNA[18,19] Excellair Nebulizer Phase II
Cystic fibrosis Antibiotics[20–23] Tobramycin Nebulizer, DPI Marketed
Aztreonam lisine Nebulizer Marketed
Colistimethate sodium Nebulizer Pilot trials
DPI Preclinical
Liposomal ciprofloxacin (ARD-3100) Nebulizer Phase II
Levofloxacin (Aeroquin, Nebulizer Phase III
formerly MP-376)
Mucolytics/mucous Dornase alfa Nebulizer Marketed
mobilizers[20,24] Lancovutide (Moli1901) Phase II
Antiproteases[25–27] a1-antitrypsin (AAT) Nebulizer Phase II
Respiratory Distress Pulmonary surfactant[28] Phospholipids/surfactant proteins Endo-tracheal tube Marketed
Syndrome
Systemic delivery Analgesia Opioids[29,30] Fentanyl (Staccato) Nebulizer Phase I
Liposomal fentanyl (AeroLEF) Phase II
Diabetes Peptides[31–37] Insulin (Affrezza) DPI Phase II
Glucagon-like peptide (MKC253) Phase I
Osteoporosis Calcitonin DPI Preclinical
Parathyroid hormone

*DPI, dry powder inhaler; pMDI, pressurized Metered Dose Inhaler. **More details on clinical studies can be found at http://clinicaltrials.gov/.

smaller airways and this penetration correlates well with a formulations (mainly suspensions) in polluting propel-
good clinical response to local treatment. Differently, a par- lants (chlorofluorocarbon and hydrofluoroalkane are gases
ticle fraction with an aerodynamic diameter in the 1–2 mm causing ozone depletion and greenhouse effects) and delivery
range is probably the most efficient for deposition into the efficiency strongly depends on the actuation-breath coordi-
capillary-rich alveolar airspaces, the target for the systemic nation of the user.[44] DPIs, on the other hand, are breath-
delivery of drugs. Finally, submicron particles can be exhaled, actuated and do not cause any coordination problem and,
if they are not aggregated and/or if insufficient time is avail- thus, have become accepted increasingly by both patients and
able for their migration to the lung walls. formulators.[45,46] In particular, DPIs are currently regarded as
A number of inhalation devices are available on the market the device of choice for macromolecule delivery to the lungs,
to effectively deliver inhaled drugs to the lungs, such as nebu- due to their potential to overcome solubility, bioavailability
lizers, pressurized metered dose inhalers (pMDIs) and dry and stability issues encountered by nebulizers and pMDIs.[14]
powder inhalers (DPIs).[42] Since they have superior handling, From a technological point of view, the handling, process-
metering and reliability as compared with nebulizers, pMDIs ing and inhalation of excipient-free dried drug particles
are the most commonly used inhalers to treat asthma and are challenging. Novel strategies have been developed and
COPD.[43] Nonetheless, pMDIs require the use of liquid drug investigated for drug inhalation via DPIs, including

© 2012 The Authors. JPP © 2012


Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235 1219
PLGA carriers for inhalation Francesca Ungaro et al.

Table 2 Main advantages of polymer carriers in pulmonary delivery Although controversial, literature data highlight that LLFs,
Microcarriers Protection of the entrapped drug especially lung mucus, along with resident macrophages, rep-
Sustained drug release resent a critical barrier to carrier-based therapies. By virtue of
Reduction of daily drug doses (i.e. side effects) its peculiar size and surface properties, a polymer carrier may
Combined therapy allow easy diffusion of a drug through the LLF,[52,53] its prefer-
Dry formulation
ential uptake from lung epithelial cells or improved escape
Macrophage escape (>10 mm)
from locally circulating macrophages.[11,54] It has been dem-
Nanocarriers Protection of the entrapped drug
Sustained drug release onstrated that the potential for access of particles to airway
Reduction of daily drug doses (i.e. side effects) mucus can be tuned at size level.[53] While large nanoparticles
Combined therapy with a mean size around 560 nm were almost completely
Efficient transport through lung lining fluid blocked by a 220 mm-thick CF sputum layer, small nanopar-
Cell targeting (e.g. macrophages) ticles were retarded only by a factor of 1.3 as compared with
Mucosal vaccination
buffer.[53] It is worth noting that particle size plays a crucial
role also in determining its preferential uptake by resident
microparticles.[8,9] Advanced products include Pulmo- macrophages, which can rapidly engulf the foreign particles
Sphere™ particles, engineered tobramycin-containing as a defence mechanism. A size of 1–2 mm for inhaled par-
hollow porous microparticles made of dipalmitoyl- ticles is ideal for macrophages to phagocytose (cell diameters
phosphatidylcholine (DPPC), the principal component of ~15–22 mm), with smaller (below 200 nm) size being taken
endogenous lung surfactants.[23] Technosphere™ Insulin up less efficiently.[5] In contrast, large particles with geometric
(MannKind Corp., Valencia, USA), based on the intermo- diameters of 10–20 mm, able to penetrate deep into lungs and
lecular self-assembly of fumaryl diketopiperazine (FDKP), avoid macrophage engulfment, are considered ideal for sys-
forming a three-dimensional highly porous sphere, is also temic delivery of protein drugs.[11] Nonetheless, the reduction
facing the market.[31,33] Nonetheless, these systems may suffer of the particle size to a nanometric range has been suggested
from the short duration of their therapeutic effect (i.e. fre- to further enhance the potential of biodegradable polymer
quent administrations). In the same way, lipid-based vesicles carriers for mucosal delivery of vaccines.[55]
(e.g. liposomes, lipoplexes, solid lipid nanoparticles), the Beside particle dimensions, the role played by the surface
most extensively investigated carriers for controlled delivery chemistry of the carrier in its interactions with, and adhe-
to the lung, do not allow sustained drug release and are sion to, lung cellular and extracellular barriers should also be
mainly delivered by nebulization.[8,9] considered.[52] It has now become clear that slight differences
In this scenario, biodegradable polymer carriers have in surface properties may have significant implications in the
become an increasingly attractive option for inhalation cellular uptake and the interactions of the carrier with the
therapies (Table 2).[12,15,47] Although calling into question biological environment. Particular attention must be paid to
safety issues, which are far to be fully settled, the use of imparting a charge or adhesiveness to the particle. When
polymer carriers may allow the reduction of daily drug doses, modified on the surface with carboxyl groups, particles as
and this could contribute to better control of local chronic small as 59 nm may be completely immobilized by human
infections while diminishing the rate of drug appearance in mucus.[56] On the other hand, large (200–500 nm) non-
the bloodstream (i.e. nonspecific distribution to non-target adhesive nanoparticles achieved by covalent modification of
tissues) and subsequent side effects. The carrier can also offer polystyrene particles with polyethylenglycol (PEG) have
the potential to combine different drugs[48] or a drug with been demonstrated to rapidly penetrate human mucus.[57]
helper excipients (e.g. absorption promoters, mucolytic More recently, hydrophilic poloxamer-coated biodegradable
agents).[49] This can be very important in the case of complex nanoparticles, displaying a negative charge, have been dem-
therapeutic regimens, such as those currently required to onstrated to more easily diffuse across the mucus barrier
treat or, better, to eradicate chronic pulmonary diseases. Fur- leading to a higher intracellular accumulation as compared
thermore, polymer particles can play a crucial role in improv- with positively charged nanoparticles modified with chito-
ing the therapeutic index of macromolecular drugs by: (i) san.[58] Thus, a thorough understanding of carrier interac-
increasing the stability of the drug both ‘in the bottle’ and tions with LLFs is a condition sine qua non to achieve
in vivo; (ii) increasing the amount of drug that reaches the site particles engineered to cross mucus barriers and, thus, for
of action; (iii) prolonging the residence time of the drug lung delivery.
in situ (i.e. sustained pharmacological effect).[50,51] Assuming the carrier successfully overcomes extracellular
Another important issue that can be potentially addressed barriers, the drug has still to interact with the target cells of
by polymer carrier-based systems intended for inhalation is the respiratory tract to be effective. The interaction of the
overcoming extracellular and cellular barriers imposed by the carrier with cells is critical in many applications such as
lungs (Figure 2).[7] mucosal vaccination or novel oligodeoxynucleotide (ODN)-

© 2012 The Authors. JPP © 2012


1220 Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235
Francesca Ungaro et al. PLGA carriers for inhalation

Cellular barriers Extracellular barriers

Trachea

Mucus

Luminal mucus layer

Bronchi Periciliary layer

Bronchioles Alveolar lining fluid

Pulmonary surfactant
Alveoli
Aqueous subphase

Figure 2 Schematic representation of cellular and extra-cellular barriers imposed by the lung to inhaled drugs. The trachea and bronchi epithelia are
made up of basal, ciliated, brush and goblet cells (~60 mm-height). The same cells are present in bronchiolar epithelium but are not as tall (~10 mm). In the
alveolar region, type-I and secretor type-II cells build up the epithelium. The conducting airways are lined with airway surface liquid, a mucus gel-aqueous
sol complex composed by the periciliary and the luminal mucus layers, which get thinner from the level of the trachea (up to 100 mm) to the bronchioles
(~3 mm). The alveoli are lined with alveolar subphase fluid and pulmonary surfactant, which is composed of approximately 80–85% phospholipids,
5–10%proteins and 5–10% other lipids. All the lung surfaces are supervised by resident mononuclear phagocytes (i.e. airway macrophages). They
are highly represented in the alveolar lining fluid (~80% of the bronchoalveolar lavage cellular components) and so are often referred to as alveolar
macrophages.[5,30,31]

based therapies. These applications require a firm control Poly(lactic-co-glycolic acid) carriers
over particle–cell interactions, which are mainly dictated by in drug delivery and their potential
size and surface properties of the delivery system. The induc- for inhalation
tion of humoral immune responses at the mucosal lymphoid
tissue (MALT) of the respiratory tract requires the transport Among synthetic biodegradable polymers, thermoplastic ali-
of particulate antigens by M-cells and their delivery to the phatic poly(ester)s, PLGAs, have generated tremendous
sub-mucosa, where dendritic cells and macrophages process research interest due to their excellent biocompatibility as
and present the antigen to naïve T-cells in the adjacent well as the possibility of tailoring their biodegradability by
mucosal lymph nodes. Similarly, at the alveoli, particulate varying composition (lactide/glycolide ratio), molecular
antigens are taken up and processed by circulating antigen- weight and chemical structure (i.e. capped and uncapped
presenting cells. Thus, when designing new inhalable end-groups).[51,65] PLGAs characterized by very different in
vaccines, surface-modified polymer carriers, especially nano- vivo life-times, ranging from three weeks to over a year, are
particles, may represent a paramount tool not only to protect available and approved for human use. Drug encapsulation
the protein-based antigen, but also to tune its interaction within PLGA copolymers is regarded as a powerful means to
with immune cells.[59,60] The challenges of nucleic acid deliv- achieve its sustained release for long time-frames and, in the
ery via the pulmonary route are possibly greater, since the case of labile drugs, effectively protect the molecule from in
drug has to cross the cellular membrane and gain access into vivo degradation occurring at the administration site. Thus,
the cytoplasm/nuclei, where the final targets are located.[19,61] injectable PLGA microparticles (< 250 mm – ideally
Therefore, the main function of the carrier, along with mac- < 125 mm) with different formulation design have been
romolecule protection against enzymatic attack, is to facili- developed for the delivery of small drugs as well as protein
tate ODN cellular uptake, promote its endosomal escape and therapeutics and nucleic acids,[50,51] resulting in several mar-
release it at the final intracellular target. This can be achieved keted products. On the other hand, PLGA nanoparticles
by biodegradable polymer carriers.[52,62–64] (< 1 mm) represent a well-established tool to achieve

© 2012 The Authors. JPP © 2012


Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235 1221
PLGA carriers for inhalation Francesca Ungaro et al.

Table 3 Examples of poly(lactic-co-glycolic acid) (PLGA)-based carriers tested for pulmonary delivery

Type Drug Formulation Key in vitro/in vivo findings


[67,68]
PLGA Conventional Insulin Microparticles/ Prolonged hypoglycaemic effect in vivo
microparticles microparticles mannitol blends Minimal signs of lung toxicity
Interleukin-2[69,70] Microparticles/ Preservation of in vitro biological activity (T-cells)
mannitol blends
Large porous particles Anthocyanin[71] Dry powders Prolonged inhibition of A549 cells death induced by
cigarette smoke
Persistence in mouse lung (20 days)
No signs of lung inflammation (10 days)
Deslorelin[72] Dry powders High drug concentrations in plasma up to 7 days
Heparin[73] Dry powders Increased plasma half-life of the drug
No cytotoxicity (Calu-3)
Insulin[49] Dry powders Preservation of protein structural integrity
In vivo alveolar deposition
Hypoglycaemic activity at low doses in diabetic rats
Prolonged hypoglycaemic effect in diabetic rats
Prostaglandin E1[74,75] Dry powders Increased stability in rat lung homogenates
Increased plasma half-life of the drug
No cytotoxicity (Calu-3)
No in vivo acute toxicity
Hollow porous particles Palmytil-acylated- Dry powders In vivo deposition in the central lung
exendin-4[76] Prolonged hypoglycaemic effect in diabetic rats
No signs of lung inflammation (2 weeks)
PLGA Nanoparticle dispersions Combined Dispersion for Sustained plasma therapeutic drug levels
nanoparticles antitubercular nebulizer Increased plasma half-life
therapy [77] Enhanced drug bioavailability
Hepatitis B vaccine[78] Dispersion for Enhanced mucosal and humoral responses to the vaccine
nebulizer Efficient uptake of nanoparticles by rat alveolar
macrophages
No cytotoxicity (Calu-3)
Insulin[79] Dispersion for Prolonged hypoglycaemic effect in diabetic
nebulizer
Trojan particles (porous Hepatitis B surface Dry powders High mucosal immune response in the lungs
nanoparticle-aggregate antigen[80]
particles) Rifampicin[81] Dry powders Sustained plasma therapeutic drug levels
Drug persistence in lung tissue and cells
Nano-embedded siRNA[64] Dry powders Preservation of siRNA structural integrity
microparticles Efficient in vitro gene silencing
No cytotoxicity (H1299 cells)
TAS-103 Dry powders Enhanced cytotoxicity against A549 cells
anticancer drug[82] High local drug concentration
Tobramycin[83] Dry powders In vitro antimicrobial activity against P. aeruginosa
Nanoparticle composition affects in vivo deposition

targeted drug or gene delivery, with special emphasis on Among the techniques developed for the production
cancer treatment.[66] of PLGA microparticles, solvent evaporation/extraction
In the attempt to discover novel strategies for the pro- methods are the most widely employed for the microencap-
longed release of drugs in the lung, PLGA nanoparticles and sulation of therapeutic macromolecules.[51] The quality of the
microparticles have been studied also as carriers for inhala- final product is related to the preservation of protein integrity
tion. As summarized in Table 3, studies with PLGA micropar- along manufacturing and in vivo application.[86,87] Excellent
ticles for inhalation have been carried out mostly with the reviews have summarized all the possible causes of protein
intention of developing inhalable formulations of proteins degradation during the microencapsulation process high-
and other macromolecules.[49,67–70,72–74,84] Currently, protein lighting the strategies efficient in preventing the phenom-
encapsulation within PLGA copolymers is considered a enon.[88,89] As a function of the critical steps affecting the
very useful strategy to entrap and deliver the native macro- quality of the final product, it is useful to single out a class of
molecule in a sustained manner.[50,51,85] excipients and select, on the basis of the literature reports,

© 2012 The Authors. JPP © 2012


1222 Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235
Francesca Ungaro et al. PLGA carriers for inhalation

those more suitable to the specific design.[65] For inhalable 20 days.[71] If this feature is desirable to prolong the therapeu-
microparticles, the microencapsulation technique and the tic effect, its influence on the safety of respirable PLGA
selected excipients should also contribute to the correct aero- particles has not been elucidated yet. Anyway, in vitro cyto-
solization properties of the particles. The technological toxicity studies have shown that native PLGA has no manifest
paradox is that respirable particles have to be small for depo- toxicity against healthy lung macrophages,[99] CF bronchial
sition, but also large enough to allow metering during manu- cells[100] or lung carcinoma cell lines.[64] Furthermore, the pro-
facturing and delivery of the dosage form. inflammatory potential of PLGA nanoparticles has been
Thus, microparticle porosity, which is generally considered recently investigated in vivo and results suggest that biode-
a microsphere defect to be overcome,[90] may oddly become a gradable nanoparticles are safer than non-biodegradable
desired feature for inhalation.[91,92] Conversely, conventional polystyrene particles[101] and do not induce lung tissue dam-
microparticles may require the use of a inert carrier (e.g. age,[102] also up to 10 days.[71] Nonetheless, chronic toxicologi-
mannitol) to become respirable.[69,70] cal data on PLGA particles are still poor and demand for
Beside macromolecule inhalation, the potential of PLGA further studies.
particles as carriers for lung delivery of antibiotics has been As polyesters, PLGAs undergo hydrolysis in vivo, forming
also deeply investigated (Table 3).[77,81,93–97] Here, the primary biologically compatible moieties that should be cleared from
goal is to increase the local concentration of the antibiotic the lung. With respect to biodegradation, among all commer-
within the macrophages, the host cells for some bacteria, such cially available PLGA types, those characterized by a rapid in
as Mycobacterium tuberculosis. Although some successful vitro degradation are undoubtedly preferred for inhalation.
attempts have been made to deliver antibiotics to the lungs The creation of synthetic PLGA derivatives more hydrophilic
via PLGA microparticles, nanoparticles seem to better fit than native PLGAs has been attempted and novel poly(vinyl
the purpose. When encapsulated into PLGA nanoparticles, alcohol) (PVA)-based branched polyesters with PLGA side
the elimination half-life and mean residence time of the chains, characterized by faster degradation rates compared
entrapped antibiotics are significantly prolonged as com- with linear PLGA of similar molecular weight, have been
pared with the orally-administered parent drugs, resulting in employed for pulmonary delivery [62,103,104].
an enhanced drug bioavailability.[77] Although few experimental studies have been performed
Despite the advantage of reduced doses and dosing fre- so far to fully explore the potential and drawbacks of PLGA in
quency, the first-developed PLGA nanoparticle-based ‘liquid’ pulmonary delivery, respirable dry powders based on PLGA
formulations pose several challenges to practical implementa- particles appear an enticing technological approach for inha-
tion. Long delivery times, low delivery efficiencies, stability lation. However, depositing drug doses reliably into the lungs
of sustained-release formulations in aqueous solution and through the newest DPIs is straightforward in principle, but
access to clean water in developing countries are only a few challenging in reality. The peculiar properties of the drug par-
of the challenges highlighted by researchers in this field. ticles, such as size, shape, density and surface properties, will
Novel formulation approaches for antibiotic delivery have affect handling, processing, and inhalation of the formula-
involved the creation of micron-scale dry powders based on tion and, therefore, its likelihood of being deposited in the
PLGA nanoparticles.[81,83,97] In fact, dry powders based on self- desired region of the lung [10,105]. In the following, a critical
assembled rifampicin-loaded PLGA nanoparticles (‘porous account of the most promising production and formulation
nanoparticle-aggregate particles’ or PNAPs) were devel- techniques employed to engineer PLGA microparticles and
oped.[81] Alternatively, mannitol microparticles containing nanoparticles for inhalation is provided, highlighting the
rifampicin-loaded PLGA nanoparticles for inhalation therapy goals already achieved and those to be gained.
of tuberculosis were prepared in one step using a four-fluid
nozzle spray drier.[97] These technologies have demonstrated a
Engineering poly(lactic-co-glycolic
great potential also in the development of dry powders for
acid) microparticles for inhalation
protein delivery, with particular regard to mucosal vaccina-
tion,[80,98] and short interference RNA (siRNA) delivery.[64] Respirable PLGA microparticles have been generated,
Although current literature data suggest the potential of making use of several techniques, that feature partly compet-
PLGA particles for prolonged drug delivery in the lung, their ing, partly complementary characteristics. Great efforts have
use for inhalation is still embryonic and presents some short- been made to improve lung deposition of the particle and
comings. In particular, a huge amount of work has still to be overcoming intracellular and extracellular barriers imposed
produced on clearance mechanisms and persistence in vivo by lungs. Besides the PLGA polymeric platform, excipients
that may considerably limit the benefit of sustained-release are sometimes needed to optimize the final particle proper-
inhalation therapy. The persistence of PLGA particles in vivo ties as a function of the preparation technique. The desirable
has been only indirectly demonstrated by some authors, who product characteristics firstly include the correct aerosoliza-
showed the presence of the released drug in the lung for up to tion properties (i.e. low mass mean aerodynamic diameter

© 2012 The Authors. JPP © 2012


Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235 1223
PLGA carriers for inhalation Francesca Ungaro et al.

passage of water across the organic phase. In particular, high


osmotic pressures in wi generate an influx of water from the
external aqueous phase (we) to wi. The occurrence of this
process during particle hardening results in the formation of
Conventional Large porous Hollow porous
a typically porous structure.
microparticles particles microparticles
In this sense, to control PLGA particle porosity and, conse-
Figure 3 Typical architectures of respirable poly(lactic-co-glycolic acid) quently, flow and aerosolization properties of the developed
microparticles. dry powders, hydroxypropyl-b-cyclodextrin (HPbCD) has
been recently tested as aid-excipient in insulin-loaded PLGA
microparticles intended for pulmonary delivery.[49,91] The use
(MMAD), high fine particle fraction (FPF) and emitted
of HPbCD in LPP production is very intriguing not only in
dose). To this end, PLGA particles have been engineered in
the light of cyclodextrin’s osmotic properties but also for its
very different ways, taking into account that the particles
potential to enhance macromolecule adsorption through
should also comply with the requirements of drug stability
respiratory epithelium. Several formulations, differing in
and prolonged release. (Figure 3)
HPbCD and insulin loadings, were produced by the wi/o/we
Among the parameters that can be adjusted to optimize the
technique and their properties compared.[91] Insulin and,
efficiency of dry powders for inhalation, particle mass density
when added, HPbCD were solubilized in wi. The technologi-
and size have recently drawn researchers’ attention to limit
cal results show that the combination of appropriate amounts
loss of drug owing to PLGA particle aggregation in the inhaler
of insulin and HPbCD plays a crucial role in achieving PLGA/
and macrophage-mediated clearance of the particles from the
HPbCD/insulin LPPs with the desired bulk and aerodynamic
lungs.[5,10] The idea is that conventional PLGA particles
properties (i.e. a highly porous structure, a very low density
achieved by micronization (i.e. 1–3 mm) are both prone to
(0.1 g/ml) and a theoretical MMAD (MMADt) lower than
aggregation and can be rapidly phagocytosed in the deep lung
10 mm).[91] The good aerosolization behaviour of the devel-
by alveolar macrophages. On the contrary, large porous par-
oped LPPs was confirmed by in vitro aerosolization tests,
ticles (LPPs) with low mass density (< 0.4 g/cm3) and high
showing that optimized PLGA/HPbCD/insulin particles had
geometric diameter (> 5 mm) can be used to enhance both
an experimental MMAD (MMADexp) ranging from 4.01 to
particle aerosolization behaviour and residence time in the
7.00 and an FPF estimated to be 26.9–89.6% at the different
lung.[106] Thus, working on size, shape and density of the par-
airflow rates tested (i.e. 30–90 l/min).[49] Confocal micro-
ticles, the first PLGA-based LPPs for insulin delivery were
scopy studies, performed after in vivo administration of
achieved.[11] More recently, great efforts have been made to
labelled PLGA/HPbCD/insulin LPPs to the rat lung by means
achieve ‘regularly shaped’ porous particles by operating selec-
of a low-scale DPI, suggest that particles reach the alveoli and
tively on size and density, thus achieving large porous ‘micro-
remain in situ after delivery. Finally, the therapeutic potential
spheres’. In so doing, PLGA-based LPPs have been engineered
of optimized PLGA/HPbCD/insulin LPPs was confirmed by
to sustain deslorelin delivery via the deep lungs,[72] allow effi-
dose–response studies performed in both normoglycaemic
cient pulmonary adsorption of low-molecular-weight hep-
and streptozotocin-induced diabetic rats.[49] In vivo data show
arin[73] and increase insulin systemic availability.[49]
that PLGA/HPbCD/insulin LPPs are able to reach the alveoli
and release insulin, which is absorbed in its bioactive form.
Large porous particles
While insulin solutions administered via pulmonary route
From a technological standpoint, PLGA-based porous micro- are unable to cause a significant hypoglycaemic effect, insulin
spheres for inhalation have been typically obtained by the delivered through PLGA/HPbCD/insulin LPPs at the same
double emulsion (wi/o/we) technique in the presence of a doses (0.5–4.0 IU/kg) significantly reduces blood glucose
porogen (Table 4). level as a function of the administered dose in both animal
When microspheres are prepared by the wi/o/we technique, models. The developed LPPs, tested in hyperglycaemic rats in
the presence of osmotically active drugs and/or excipients in evident pathological conditions, exert a very significant and
the internal aqueous phase (wi), as well as their concentra- longer hypoglycaemic effect even at insulin doses as low as
tion, generates an osmotic pressure gradient which can 0.5 IU/kg (about 0.5 mg of PLGA/HPbCD/insulin LPP per
strongly affect particle porosity (Figure 4a).[65] Thus, LPPs rat) as compared with an insulin solution. The duration of
can be achieved adding osmogens in the internal aqueous the effect was consistent with previous results achieved by
phase of the double emulsion, thus causing water influx from Edwards and colleagues,[11] who showed falling glucose levels
the external to the internal aqueous phase during solvent for the first 10 h after inhalation of PLGA/insulin LPPs, fol-
evaporation (i.e. particle hardening). Under the influence of lowed by relatively constant low glucose levels for the remain-
an osmotic gradient, the organic phase of the double emul- der of the 96-h period. It is worth noting that these effects
sion acts as a semi-permeable membrane allowing the were observed for PLGA/insulin LPPs at much higher insulin

© 2012 The Authors. JPP © 2012


1224 Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235
Francesca Ungaro et al. PLGA carriers for inhalation

Table 4 Preparation techniques employed to develop porous poly(lactic-co-glycolic acid) (PLGA) microparticles for inhalation

Preparation technique Porosigen agent Delivered drug


[72]
Supercritical fluids (CO2SC) Cyclodextrins Deslorelin
Precision particle Oils[107] Ciprofloxacin
Fabrication (PPF)*
Single emulsion (o/w) Poloxamer (Pluronic F68 and F127) [84,108] Human Growth Hormone
Palmytil-acylated-exendin-4
Double emulsion (wi/o/we) Cyclodextrins[49,76,91,109] Insulin
Bovine Serum Albumin
Palmytil-acylated-exendin-4
Sodium chloride[76] Palmytil-acylated-exendin-4
Poly(ethylenimine)[73,74] LMW Heparin
Prostaglandin E1
Ammonium bicarbonate[92,100,110,111] Budesonide
Decoy oligonucleotide
Doxorubicin
Lysozyme
Rhodamine-dextran
Poly(vinyl alcohol)[112] Hepatitis B surface antigen
None[113]** Capreomycin sulfate

*Patented technology that uses acoustic excitation and/or flow-limited field-injection electrostatic spraying to fabricate uniformly sized particles. **Par-
ticle porosity has been achieved by rapid evaporation of the organic solvent.

(a)
wi
Osmogen o we

Osmogen
Water we
wi o
influx

Osmogen

(b)
wi/o/we

w1

o
w2

Figure 4 Production of large porous particles by the double-emulsion technique. (a) Generation of an osmotic pressure gradient between wi and we.
(b) Addition of an effervescent salt (e.g. ammonium bicarbonate) in wi.

doses (about 9 mg of LPPs containing 20% of insulin by nary delivery of palmityl-acylated exendin-4 (Pal-Ex4), a
weight were administered per rat) as compared with PLGA/ potent glucagon-like peptide-1 agonist with great potential
HPbCD/insulin LPPs. for the treatment of diabetes.[76] The contemporary addition
HPbCD has been recently employed to produce also of HPbCD and sodium chloride within wi, along with
albumin-coated hollow porous PLGA particles for pulmo- controlled solvent evaporation, allowed the achievement of

© 2012 The Authors. JPP © 2012


Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235 1225
PLGA carriers for inhalation Francesca Ungaro et al.

hollow LPPs. Plain particles were further processed with optimal aerodynamic properties. Although the authors suc-
human serum albumin (HSA), which was conjugated with ceeded in improving drug encapsulation efficiency, poor
the carboxylate groups of poly(ethylene-alt-maleic anhy- control over release properties was achieved without further
dride), to achieve HSA-coated hollow LPPs based on PLGA. modification of formulation conditions. To further engineer
The developed Pal-Ex4 loaded particles demonstrated great the system, lipid-engineered gas-foamed LPPs were recently
potential for inhalation, in terms of drug loading, release rate, developed in our laboratory.[92] To control LPP release prop-
in vitro/in vivo aerosolization efficiency and duration of the erties, two lipid aid excipients were tested. Our first choice
hypoglycemic effects in vivo. On the basis of the results, the was DPPC, the major component of human lung surfactant,
authors ascribe several benefits to HSA coating: prevention of which is gaining increasing research interest in the develop-
particle aggregation, improvement of LPP aerosolization ment of respirable dry powders for a number of reasons,
properties and, last but not least, potential for tight binding including its biocompatibility.[9] As an alternative lipid
of the fatty-acid conjugated peptide, so as to control its release excipient, we investigated the potential of 1,2-dioleoyl-3-
in vivo.[76] trimethylammonium-propane (DOTAP), a cationic lipid
Despite encouraging results, severe limitation of the extensively studied as transfection agent for nucleic acids.[117]
osmogen-based technological approach is represented by the The effect of the presence of DPPC or DOTAP upon the prop-
poor control of drug encapsulation efficiency, ascribable to erties of gas-foamed LPPs containing a model hydrophilic
mass exchanges and consequent drug loss occurring between macromolecule, rhodamine B isothiocyanate–dextran
the two phases during particle hardening. This phenomenon, (Rhod-dex), was assessed. We found that in the case of hydro-
as well as rapid drug release due to the macroporous structure philic macromolecules unable to interact with PLGA end-
of the system, can be particularly dramatic in the case of groups, such as Rhod-dex, excipient addition was essential
highly hydrophilic macromolecules, such as nucleic acids.[114] to increase the amount of drug entrapped within LPPs, being
Establishment of interactions of the drug with helper excipi- as high as 80% only for DPPC-engineered or DOTAP-
ents may limit macromolecule loss. Along this line, an alter- engineered gas-foamed LPPs. The Rhod-dex release profile
native osmotic agent that has been tested to produce LPPs is from LPPs was also strongly affected by excipient addition in
polyethylenimine (PEI), a hydrophilic polycation that forms the initial formulation, with lipid-engineered LPPs allowing
complexes by electrostatic interactions with different anionic for a more prolonged release of Rhod-dex as compared with
macromolecules (e.g. nucleic acids, heparin).[115] A excipient-free LPPs. Conceiving the developed LPPs for drug
concentration-dependent increase in particle porosity was inhalation, DPPC-engineered and DOTAP-engineered LPPs
experienced after PEI addition in wi during preparation of displayed optimal FPF and MMADexp. In vivo deposition
PLGA microparticles for the delivery of oligonucle- studies performed after intratracheal administration of LPPs
otides.[114,116] The use of PEI as core-modifying agent has been in rats confirmed the ability of the developed dry powders
re-examined to achieve respirable PLGA LPPs of low- to deposit along bronchi and bronchioles. On the basis of
molecular-weight heparin[73] and, more recently, prostaglan- encouraging technological results, we are currently develop-
din E1 (PGE1).[74] Despite particle porosity, LPPs showed high ing respirable DPPC-engineered gas-foamed LPPs for pro-
entrapment efficiency, ascribed to drug–polycation inter- longed pulmonary delivery of a decoy oligodeoxynucleotide
actions. Furthermore, in both cases, a remarkable extension to nuclear factor-kB (NF-kB) (dec-ODN) for the treatment
of the plasma half-life of the drug (from 6 h to 24 h) was of chronic lung inflammation (e.g. CF).[100] Dec-ODN LPPs
observed. represent the first attempt to delivery ODN-based therapeu-
An alternative formulation strategy to achieve PLGA- tics to the lung in the form of dry powders. Dec-ODN was
based LPPs for the prolonged release of hydrophilic macro- effectively encapsulated (~80%) within biodegradable par-
molecules relies on the use of an effervescent agent, namely ticles with controlled porosity, confirming that the use of
ammonium bicarbonate, which decomposes into ammonia lipid excipients and gas foaming may result in efficient
and carbon dioxide during emulsification, forming a porous microencapsulation of a highly hydrophilic macromolecule
matrix as the gas products escape (i.e. gas-foamed LPPs)[110] (i.e. dec-ODN) within highly porous PLGA particles. Fur-
(Figure 4b). Since pore formation depends on effervescence thermore, a sustained release of dec-ODN from LPPs was
rather than on diffusional mass exchanges between aqueous achieved. Particle aserosol performance, as evaluated by
phases, this technique allows efficient encapsulation of mac- multi-stage liquid impinger, demonstrated that the devel-
romolecules in highly porous PLGA particles.[92,100,110] oped dec-ODN LPPs have very good flow properties, with
The new process was firstly applied by Yang et al. for the more than 95% of the capsule content being emitted during
encapsulation of two model molecules, lysozyme and doxo- aerosolization. The MMADexp of < 6 mm, along with findings
rubicin hydrochloride.[110] The addition of an appropriate of previous studies, strongly supports a preferential depo-
amount of ammonium bicarbonate was essential to achieve a sition of dec-ODN LPPs in the distal airways.[63] In vitro
homogeneous population of highly porous particles with pharmacological studies performed in CF human bronchial

© 2012 The Authors. JPP © 2012


1226 Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235
Francesca Ungaro et al. PLGA carriers for inhalation

o/w o
o
w w

Figure 5 Production of large porous particles by the single-emulsion technique. Interconnected pores are generated by addition of extractable poro-
gens in the organic phase.

epithelial IB3-1 cells showed that lipopolysaccharide (LPS) the deep lungs for up to seven days as compared with plain
challenge caused an increase of NF-kB/DNA binding activity, deslorelin (plasma concentrations sustained up to three days)
which was significantly inhibited by DPPC-engineered and conventional deslorelin PLGA particles (plasma concen-
dec-ODN LPPs at 24 and 72 h.[100] This inhibitory effect on trations 2-fold lower at day 7). More recently, the patented
NF-kB/DNA binding activity was correlated with decreased PPF technology was used to create monodisperse PLGA-
interleukin (IL)-6 and IL-8 secretion and mRNA levels.In con- based LPPs for pulmonary delivery of nano-precipitated
trast, naked dec-ODN exhibited these effects only at 24 h. ciprofloxacin (NanoCipro dry powders) using oils as extract-
These results were supported by an enhanced structural stabil- able porogens.[107] Notably, particle morphology strictly
ity of dec-ODN in cultured cells due to entrapment in LPPs.[63] depended upon the nature and concentration of the extract-
PLGA-based LPPs have been also achieved by the single able oil – canola oil led to PLGA particles with a porous web-
emulsion technique (Table 1)[84,108]. Porous biodegradable like internal structure and with silicon oil hollow porous
PLGA microparticles with interconnected pores have been particles were achieved. The main limitation of the technique
fabricated using poloxamers as extractable porogens was the low ciprofloxacin encapsulation efficiency.
(Figure 5). Pores are generated by the time difference between Despite the peculiar limitations, all the advanced particle-
PLGA hardening and the extraction of the hydrophilic surfac- engineering techniques, along with the traditional methods of
tants from the organic phase using water. Nonetheless, the microencapsulation, have contributed to the increased possi-
drug should be loaded by adsorption onto LPPs. Thus, it is bility of formulating LPPs for pulmonary delivery. Notably,
very important to favour drug interactions with PLGA. This PLGA particles have been effectively engineered into LPPs
is the case of the fatty acid-conjugated peptide Pal-Ex4, the with aerosol and release properties suitable for the prolonged
palmitic acid moieties of which were demonstrated to delivery of drugs deep in the lungs. The challenge for research-
improve Ex4 interactions with the hydrophobic PLGA and, ers working in this field is that none of these techniques can
consequently, to extend its in vitro release (completed after really provide the solution to all types of molecules.
five days).[76] In turn, an extended therapeutic duration in vivo
was observed, though ascribable not only to sustained drug
Engineering poly(lactic-co-glycolic
delivery but also to the increased plasma half-life of Pal-Ex4
acid) nanoparticles for inhalation
due to the induction of albumin binding, typical of the con-
jugated peptide itself. In the current era of nanotechnology,biodegradable nanopar-
Novel technologies, such as the supercritical fluid process ticles are gaining momentum as carriers for the inhalation
and the patented precision particle fabrication (PPF), have of drugs.[12,118] Indeed, nanoparticle delivery to the lungs is an
been also tested for the achievement of PLGA-based LPPs attractive concept because it can cause retention of the carrier
(Table 1). Supercritical CO2 processing was applied to in the lungs (particles of a few hundred nanometers represent a
prepare deslorelin-loaded LPPs with the aim of reducing the tenacious resident of the lungs) accompanied by a prolonged
residual solvent content of the particles, retain protein integ- drug release,meaning improved lung bioavailability over con-
rity and sustain its release in vivo.[72] The developed particles ventional PLGA microparticles. Furthermore, nanoparticles
effectively sustained the systemic delivery of deslorelin via represent a promising tool to more simply penetrate airway

© 2012 The Authors. JPP © 2012


Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235 1227
PLGA carriers for inhalation Francesca Ungaro et al.

(a)

Nanoparticle on Nano-embedded
Trojan particles
coarse inert carrier microparticles

Figure 6 Inhalable poly(lactic-co-glycolic acid) nanoparticles.


Nanoparticle-based dry powders for inhalation can be achieved by:
adsorption on coarse inert carriers, nanoparticle self-assembling (Trojan
particles or porous nanoparticle-aggregate particles) or embedding
nanoparticles into an inert microparticle.

barriers, which can be better overcome at nanosize level.[52]


Finally, carriers of nanometric size offer potential for drug
×10000 2 μm 2.00 kV 5 mm
targeting to specific lung tissue and cell populations.[119]
From a technological point of view, the development of (b)
effective PLGA nanocarriers for inhalation first requires
adequate engineering of the particles at the nanosize level. On
this matter, PLGA nanoparticles with different features
(size, morphology, zeta-potential) may be achieved by con-
trolling the parameters specific to the production method
employed.[120,121] Since the potential of nanoparticles to inter-
act with lung tissue strongly depends on nanoparticle size and
surface charge,[52] this is an important aspect to be taken into
account for excipient selection and formulation design.
Nonetheless, another great challenge of using nanoparticles
for inhalation is the low inertia of dry powders with a mean
size lower than 1 mm, which are generally exhaled upon inha-
lation.[5,12] Thus, the most widely employed nanoparticle-
×30000 1 μm 2.00 kV 5 mm
based inhalation approach relies on the nebulization of
nanoparticulate aqueous dispersions.
Figure 7 Scanning electron micrographs of spray dried rifampicin
Different strategies to engineer the system at the microsize porous nanoparticle-aggregate particles (PNAPs) containing 80% nano-
level and achieve ‘micrometric’ nanoparticle-based dry particles by weight (PNAP80). A magnification of the surface of the PNAP
powders have recently been explored (Figure 6).[98,122] Among indicates a shell of aggregated nanoparticles with intact structure (scale
them, self-assembly of nanoparticles or their embedding bars represent: 2 mm (a), 1 mm (b)).[81]
within an inert ‘microcarrier’ have been extensively investi-
gated and, probably, represent the most promising techno-
rophage targeting) could be combined with the ease of flow,
logical approach.
processing and aerosolization potential of LPPs.
Formation of the large porous nanoparticle aggregates
The ‘Trojan’ approach
occurs via a spray-drying process that ensures the drying time
Since nanoparticle drying to develop dry powders for inhala- of the sprayed droplet (i.e. time required for a droplet to dry)
tion is very difficult to achieve, given that nanoparticles is sufficiently shorter than the characteristic time for redi-
aggregate excessively in the dry state, large porous carriers stribution of nanoparticles by diffusion within the drying
have been used to deliver nanoparticles deep in the lung (i.e. droplet (i.e. time required for nanoparticles to diffuse from
Trojan particles) in the form of dry powders.[123] As in Vir- the edge of the droplet to its centre). The proof of principle of
gilio’s epic poem, which tells of the Greek army entering and using Trojan particles for inhalation was given on polystyrene
destroying the city of Troy staying hidden in a horse, PNAPs nanoparticles. This approach has been found useful for
have been developed to act as carrier particles that release achieving hybrid DPPC and hyaluronic acid large porous
nanoparticles once delivered into the body, thereby acting as a carriers for PLGA nanoparticles releasing dexamethasone
‘Trojan’ delivery systems for nanoparticles (Figure 7).[80,81,123] acetate.[124] Scanning electron microscopy and confocal laser
In so doing, all the advantages of nanoparticles for inhalation scanning microscopy analysis showed that dexamethasone-
(e.g. long-term residence in situ, prolonged drug release, mac- loaded Trojan particles are spherical, hollow and possess an

© 2012 The Authors. JPP © 2012


1228 Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235
Francesca Ungaro et al. PLGA carriers for inhalation

irregular surface ascribable to nanoparticles, likely held


together by DPPC and hyaluronic acid.
More recently, rifampicin-loaded PLGA nanoparticles have 2~10 μm
been dispersed throughout a matrix of an inert excipient,
l-leucine, a hydrophobic amino acid demonstrated to
improve powder dispersibility and aerosolization proper-
ties.[81] Rifampicin-loaded PNAPs possessed properties suit- Lung lining fluid
able for efficient deposition in the lungs. In vitro release Lung epithelium
showed an initial burst of rifampicin, with the remainder
available for release beyond 8 h. Analogously, systemic levels
of rifampicin were detected for 6–8 h after delivery of PNAPs
to guinea-pigs by insufflation. A prolonged permanence of
rifampicin both in bronco-alveolar lavage (BAL) and lung Figure 8 Proposed mechanism of nanoparticle release from nano-
embedded microparticles into lung lining fluid.
tissue was observed as compared with porous particles con-
taining free rifampicin.
A recent paper described dry powders intended for vaccine
delivery were based on PNAP technology.[80] In particular, Among the inert excipients available for NEM production,
PLGA/PEG nanoparticles composed of a PLGA core and a lactose is the first choice and the most commonly used in
PEG hydrophilic shell, containing recombinant hepatitis B marketed DPIs. It has an established safety and stability
surface antigen (rHBsAg), were prepared by the double emul- profile, may be processed by different manufacturing tech-
sion method and subsequently spray-dried with l-leucine. niques with tight controls over purity and physical proper-
Again, hollow and low-density particles with a rough inner ties, is easily available at different grades and is inexpensive.[9]
surface, ascribed to intact PLGA/PEG nanoparticles, were It is conceived that the fine particles of lactose reaching the
achieved. Various PNAP formulations containing rHBsAg lungs are rapidly absorbed and metabolized by the intestinal
were administered to guinea-pigs by the pulmonary route epithelium and are principally excreted in urine. Further-
and the immune response elicited in the systemic circulation more, in contrast to oral administration, lactose swallowed at
and in the lungs assessed. The IgG titres were measured in the the levels present in inhaled preparations (up to 25 mg) is
serum for 24 weeks after the initial immunization; whereas unlikely to present problems even in patients with lactose
the titres of IgA, an indicator of mucosal immunity, were intolerance.[129]
measured in BAL. Results showed that rHBsAg PNAPs have Other safe inert excipients, such as mannitol and trehalose,
the advantage of eliciting a high mucosal immune response in have been tested as potential carriers in naoparticle-based dry
the lungs without traditional adjuvants. Nonetheless, no for- powders for inhalation and have found their way into mar-
mulation was able to elicit a systemic response comparable keted products for inhalation.[9] Research interest in mannitol
with that of a control antigen adsorbed on alum administered is increased by its ability to act as an airway rehydrating agent,
via the parenteral route. inducing water flux into the bronchial lumen.[130] Indeed, the
first attempts to produce NEMs for inhalation have been
made with mannitol particles loaded with insulin-loaded
Nano-embedded microparticles
PLGA nanoparticles by a spray-drying fluidized bed granula-
Although direct drying of nanoparticle suspensions has dem- tor.[79] This resulted in soft matrix composite granules dis-
onstrated some success in the creation of dry powders com- playing aerosolization properties superior to those of freeze-
posed of agglomerated PLGA nanoparticles, this technique dried nanoparticles. In vivo deposition studies showed that
requires great attention to preserve nanoparticle integrity more than 50% of the composite granules were able to reach
upon drying and may not always offer the desired control over rat bronchioles and alveoli. As a result, a prolonged pharma-
their flow and aerosolization properties. An alternative early- cological effect lasting more than 12 h was achieved as
investigated approach consists of nanocomposite particles compared with insulin solutions administered either intrave-
obtained by inclusion of drug-loaded PLGA nanoparticles nously or intratracheally.
within sugar micoparticles, used as inert carriers.[83,125–128] Recently, inhalable composite particles made of sugars and
The nanoparticle-containing micron-sized particles, also PLGA have been achieved by a classical spray-drying tech-
termed nano-embedded microparticles (NEMs), are nique.[82,83,125,126] Trehalose and lactose were tested as inert car-
designed to release primary nanoparticles after reaching riers for rifampicin-loaded PLGA nanoparticles.[125,126]
deep lung, upon the dissolution of the inert carrier in LLF Formulation studies were useful to assess the effect of the
(Figure 8). preparation conditions – inlet temperature, size and weight
ratio of primary NPs – on the properties of NEMs. Notably,

© 2012 The Authors. JPP © 2012


Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235 1229
PLGA carriers for inhalation Francesca Ungaro et al.

the authors found that the inlet temperature of the spray- Thus, PLGA nanoparticle composition appears to play a
drier was crucial to allow the decomposition of NEMs into crucial role in determining not only the technological fea-
intact nanoparticles and its adjustment was necessary when tures of nanoparticles but also, once processed in the form of
the size of the primary nanoparticles or the sugar carrier were NEMs, their in vitro/in vivo deposition pattern.
changed. Contrariwise, once achieved at optimum tempera- A crucial issue pertaining to NEMs is related to the drug-
tures, the ratio by weight of nanoparticles did not affect the to-powder ratio, which still needs to be improved to achieve
inhalation performance of NEMs. On the basis of encourag- safe and effective NEMs for in vivo inhalation of antibiot-
ing preliminary data, PLGA nanoparticles were loaded with ics.[83] Actually, the addition of an inert ingredient in elevated
an anti-cancer drug, TAS-103, and spray-dried with threal- ranges decreases the amount of PLGA nanoparticles and, in
ose.[82] The cytotoxicity of the developed NEMs against A549 turn, the dose of active drug administered. Nonetheless, this
cells was higher than that of the free drug. Furthermore, when is not a challenge for drugs that are therapeutically active at
NEMs were administered to rats by inhalation, the cytotoxic very low doses, such as biotech molecules (i.e. proteins rather
agent reached higher concentrations in the lung than in the than oligonucleotides). On this matter, it should be under-
plasma or those achieved after intravenous administration of lined that spray-drying has been demonstrated to be an excel-
free drug. lent technique also for engineering of siRNA-loaded PLGA
In a recent work, we tried to shed light on the role played by nanoparticles.[64,127] Mannitol NEMs with aerosol properties
nanoparticle composition on the interactions of NEMs with suited for deep lung delivery have been engineered without
the lung environment.[83] To this purpose, we designed and destroying the biological activity of sensitive molecules.
developed a pulmonary delivery system for antibiotics, such In the light of current literature data it can be concluded
as tobramycin, based on NEMs consisting of PLGA nanopar- that PNAPs and NEMs are very promising systems for sus-
ticles embedded in an inert microcarrier made of lactose. At tained delivery of conventional and biotech drugs in the
nanosize level, helper hydrophilic polymers were used to lungs. In perspective, they may represent important tools for
impart the desired surface, bulk and release properties to mucosal vaccination as well as for efficient treatment of local
PLGA nanoparticles prepared by a modified emulsion- lung diseases, which demand nanosized carriers.
solvent diffusion technique.[83] Results showed that PVA and
chitosan are essential to optimize the size and modulate the
Conclusions
surface properties of tobramycin-loaded PLGA nanopar-
ticles, whereas the use of alginate allows efficient tobramycin Over the last 20 years, drug delivery to the respiratory tract
entrapment within nanoparticles and its release up to one has become of choice for the treatment of asthma, COPD and
month. Optimized formulations displayed good in vitro respiratory infections. The main objective is to confine the
antimicrobial activity against Pseudomonas aeruginosa plank- therapeutic agent to the airways so as to maximize drug con-
tonic cells. Nonetheless, since nanoparticles will be embed- centration at the site of action and, in so doing, maximize
ded in vivo in the lung mucus, which will prevent them from drug efficacy. In addition, the lung is under investigation as a
reaching the target, selected nanoparticle formulations were portal of entry to the body, permitting delivery of drugs, espe-
subjected to further in vitro studies to assess their ability to cially peptides and proteins, via the airway surfaces into the
interact with mucus environment. The rough determination bloodstream. Literature review shows that advanced particle
of nanoparticle–mucin interactions by the mucin-particle engineering techniques have contributed to the increased
method suggest that chitosan-engineered nanoparticles have possibility of formulating nano-carriers and micro-carriers
a higher tendency to interact with mucin as compared with for both local and systemic pulmonary delivery of conven-
PVA-engineered ones. The strength of interaction was con- tional drugs and biotech macromolecules, such as emerging
firmed by the z-potential of the mucin–nanoparticle disper- inhaled proteins and oligonucleotides.
sion, indicating a strong adsorption of the polymer onto the Although carrier-free formulations for inhalation are
surface of chitosan-engineered nanoparticles. This phenom- somewhat advisable due to their low toxicological profile,
enon will likely affect nanoparticle diffusion through mucus. biodegradable and biocompatible polymer particles may
Fluorescence-assisted transport studies performed on a arti- permit efficient protection and long-term delivery of the
ficial mucus model,[131] showed that nanoparticle transport inhaled drug. On this matter, PLGA particles for inhalation
through mucus is facilitated when mucin shields the nano- have been produced on the basis of numerous strategies that
particle surface charge (i.e. chitosan-engineered nanopar- use a combination of available particle technologies and
ticles). Notably, in vivo biodistribution studies of NEMs excipients to develop the desired dosage form. Particle prop-
engineered by spray-drying showed that PVA-engineered erties can be finely tuned at micro-size and nano-size level to
alginate/PLGA nanoparticles reached the deep lung, while fulfill specific therapeutic needs. The challenge to carrier
chitosan-engineered nanoparticles were found in great development is that none of the technological approaches
amounts in the upper airways, lining lung epithelial surfaces. reported in the literature is universal and it is very difficult

© 2012 The Authors. JPP © 2012


1230 Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235
Francesca Ungaro et al. PLGA carriers for inhalation

to choose a priori the one that best fits the purpose. In vitro/in still exist, PLGA carriers represent a real benefit for both phar-
vivo studies represent a critical step before selection of the maceutical companies working to develop novel inhaled
best formulations as candidates for use in humans. Although products and patients suffering from chronic lung diseases.
lung deposition studies are still crucial, there is an impelling
need for in vitro/in vivo models to investigate what happens Declarations
after the particle has landed. Special attention must be paid to
the evaluation of the safety of PLGA in the lung. In particular, Conflict of interest
the shortage of chronic toxicological data on engineered The Author(s) declare(s) that they have no conflicts of inter-
PLGA particles must be balanced with studies to definitively est to disclose.
assess the toxicological potential of such particles in the lung,
before their in vivo use.
Funding
In the light of these considerations, nano-carriers and
micro-carriers of PLGA seem an emerging formulation strat- The authors wish to thank Fondazione per la Ricerca sulla
egy in pulmonary delivery. When adequately engineered, Fibrosi Cistica-Onlus – Delegazione di Torino (FFC#5/2007)
PLGA particles may provide sustained drug delivery to the and – Delegazione del Lago di Garda con i GdS dell’Isola Berga-
lungs, extend duration of action, reduce the therapeutic dose masca e di Chiasso (FFC#23/2011) for supporting their
and reduce the adverse effects of highly toxic drugs, thus research on the development of respirable PLGA carriers for
improving patient compliance. Although many challenges cystic fibrosis therapy.

References 9. Pilcer G, Amighi K. Formulation 17. Global Strategy for Asthma Manage-
strategy and use of excipients in pul- ment and Prevention. [Global Initia-
1. Weers JG et al. Pulmonary formula- monary drug delivery. Int J Pharm tive for Asthma (GINA) web site].
tions: what remains to be done? J 2010; 392: 1–19. 2010. Available at: http://www.
Aerosol Med Pulm Drug Deliv 2010; 10. Chow AH et al. Particle engineering ginasthma.org/.
23(Suppl. 2): S5–23. S5–23. for pulmonary drug delivery. Pharm 18. Seguin RM, Ferrari N. Emerging oli-
2. Newman S et al. Respiratory Drug Res 2007; 24: 411–437. gonucleotide therapies for asthma
Delivery: Essential Theory & Practice. 11. Edwards DA et al. Large porous and chronic obstructive pulmonary
Richmond, VA: RDD Online, 2009. particles for pulmonary drug deli- disease. Expert Opin Investig Drugs
3. Onoue S et al. New treatments for very. Science 1997; 276: 1868– 2009; 18: 1505–1517.
chronic obstructive pulmonary 1871. 19. Parry-Billings M et al. Oligo-
disease and viable formulation/device 12. Rytting E et al. Biodegradable poly- nucleotides: new therapeutic appr-
options for inhalation therapy. Expert meric nanocarriers for pulmonary oaches for asthma and chronic
Opin Drug Deliv 2009; 6: 793–811. drug delivery. Expert Opin Drug Deliv obstructive pulmonary disease. Curr
4. Ibrahim BM et al. Challenges and 2008; 5: 629–639. Opin Investig Drugs 2010; 11: 1276–
advances in the development of inhal- 13. Kurmi BD et al. Micro- and 1285.
able drug formulations for cystic nanocarrier-mediated lung targeting. 20. Heijerman H et al. Inhaled medica-
fibrosis lung disease. Expert Opin Expert Opin Drug Deliv 2010; 7: 781– tion and inhalation devices for lung
Drug Deliv 2011; 8: 451–466. 794. disease in patients with cystic fibrosis:
5. Patton JS, Byron PR. Inhaling medi- 14. Shoyele SA, Cawthorne S. Particle a European consensus. J Cyst Fibros
cines: delivering drugs to the body engineering techniques for inhaled 2009; 8: 295–315.
through the lungs. Nat Rev Drug biopharmaceuticals. Adv Drug Deliv 21. Geller DE et al. Levofloxacin inhala-
Discov 2007; 6: 67–74. Rev 2006; 58: 1009–1029. tion solution (MP-376) in patients
6. Misra A et al. Inhaled drug therapy for 15. Mohamed F, van der Walle CF. Engi- with cystic fibrosis with Pseudomonas
treatment of tuberculosis. Tuberculo- neering biodegradable polyester par- aeruginosa. Am J Respir Crit Care Med
sis (Edinb) 2011; 91: 71–81. ticles with specific drug targeting and 2011; 183: 1510–1516.
7. Patton JS et al. The particle has drug release properties. J Pharm Sci 22. Geller DE et al. Pharmacokinetics and
landed–characterizing the fate of 2008; 97: 71–87. safety of MP-376 (levofloxacin inhala-
inhaled pharmaceuticals. J Aerosol 16. Global Strategy for the Diagnosis, tion solution) in cystic fibrosis sub-
Med Pulm Drug Deliv 2010; 23(Suppl. Management and Prevention of jects. Antimicrob Agents Chemother
2): S71–S87. S71–S87. COPD. [Global Initiative for Chronic 2011; 55: 2636–2640.
8. Salama R et al.Recent advances in con- Obstructive Lung Disease (GOLD) 23. Geller DE et al. Development of an
trolled release pulmonary therapy. web site]. 2010. Available at: http:// inhaled dry-powder formulation of
Curr Drug Deliv 2009; 6: 404–414. www.goldcopd.org/. tobramycin using PulmoSphere

© 2012 The Authors. JPP © 2012


Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235 1231
PLGA carriers for inhalation Francesca Ungaro et al.

technology. J Aerosol Med Pulm Drug Technol Ther 2011; 13: 1201– 47. Cryan SA. Carrier-based strategies for
Deliv 2011; 24: 175–182. 1206. targeting protein and peptide drugs to
24. Pettit RS, Johnson CE. Airway- 34. Tewes F et al. Development and the lungs. AAPS J 2005; 7: E20–E41.
rehydrating agents for the treatment characterisation of soluble polymeric 48. Adi H et al. Controlled release anti-
of cystic fibrosis: past, present, and particles for pulmonary peptide deliv- biotics for dry powder lung delivery.
future. Ann Pharmacother 2011; 45: ery. Eur J Pharm Sci 2010; 41: 337– Drug Dev Ind Pharm 2010; 36: 119–
49–59. 352. 126.
25. Martin SL et al. Safety and efficacy 35. Baginski L et al. Investigations into 49. Ungaro F et al. Insulin-loaded PLGA/
of recombinant alpha(1)-antitrypsin the fate of inhaled salmon calcitonin cyclodextrin large porous particles
therapy in cystic fibrosis. Pediatr at the respiratory epithelial barrier. with improved aerosolization proper-
Pulmonol 2006; 41: 177–183. Pharm Res 2011; 29: 332–341. ties: in vivo deposition and hypogly-
26. Griese M et al. alpha1-Antitrypsin 36. Codrons V et al. Systemic delivery caemic activity after delivery to rat
inhalation reduces airway inflamma- of parathyroid hormone (1-34) using lungs. J Control Release 2009; 135:
tion in cystic fibrosis patients. Eur inhalation dry powders in rats. J 25–34.
Respir J 2007; 29: 240–250. Pharm Sci 2003; 92: 938–950. 50. Sinha VR, Trehan A. Biodegradable
27. Brand P et al. Lung deposition of 37. Shoyele SA et al. The effects of excipi- microspheres for protein delivery.
inhaled alpha1-proteinase inhibitor ents and particle engineering on the J Control Release 2003; 90: 261–280.
in cystic fibrosis and alpha1- biophysical stability and aerosol per- 51. Mundargi RC et al. Nano/micro
antitrypsin deficiency. Eur Respir J formance of parathyroid hormone technologies for delivering macro-
2009; 34: 354–360. (1-34) prepared as a dry powder for molecular therapeutics using poly
28. Raghavendran K et al. Surfactant inhalation. AAPS PharmSciTech 2011; (D,L-lactide-co-glycolide) and its
therapy for acute lung injury and 12: 304–311. derivatives. J Control Release 2008;
acute respiratory distress syndrome. 38. Shoyele SA. Controlling the release of 125: 193–209.
Crit Care Clin 2011; 27: 525–559. proteins/peptides via the pulmonary 52. Lai SK et al. Mucus-penetrating nano-
29. Alexza Pharmaceuticals. A Growing route. Methods Mol Biol 2008; 437: particles for drug and gene delivery to
Pipeline of Promising Staccato® 141–148. 141–148. mucosal tissues. Adv Drug Deliv Rev
Products [Alexza Pharmaceuticals 39. Rubin BK. Air and soul: the science 2009; 61: 158–171.
web site]. 2011. Available at: http:// and application of aerosol therapy. 53. Sanders NN et al. Cystic fibrosis
www.alexza.com/products. Respir Care 2010; 55: 911–921. sputum: a barrier to the transport of
30. YMBiosciences Inc. Free and 40. Carvalho TC et al. Influence of par- nanospheres. Am J Respir Crit Care
liposome-encapsulated fentanyl: ticle size on regional lung deposition– Med 2000; 162: 1905–1911.
leveraging pulmonary delivery for what evidence is there? Int J Pharm 54. Evora C et al. Relating the phago-
personalized pain management [Pul- 2011; 406: 1–10. cytosis of microparticles by
monary drug delivery:new perspec- 41. Bisgaard H et al. Drug Delivery to the alveolar macrophages to surface
tives on inhalers and inhalables web Lung. New York: Marcel Dekker, 2002. chemistry: the effect of 1,2-
site]. 2007. Available at: http://www. 42. Dolovich MB, Dhand R. Aerosol drug dipalmitoylphosphatidylcholine. J
ondrugdelivery.com/publications/ delivery: developments in device Control Release 1998; 51: 143–152.
Pulmonary_June_07.pdf. design and clinical use. Lancet 2011; 55. Csaba N et al. Nanoparticles for nasal
31. Cassidy JP et al. Insulin lung deposi- 377: 1032–1045. vaccination. Adv Drug Deliv Rev 2009;
tion and clearance following Techno- 43. Hess DR. Metered-dose inhalers and 61: 140–157.
sphere(R) insulin inhalation powder dry powder inhalers in aerosol 56. Olmsted SS et al. Diffusion of macro-
administration. Pharm Res 2011; 28: therapy. Respir Care 2005; 50: 1376– molecules and virus-like particles in
2157–2164. 1383. human cervical mucus. Biophys J
32. Marino MT et al. Pharmacokinetics 44. Geller DE. Comparing clinical fea- 2001; 81: 1930–1937.
and pharmacodynamics of inhaled tures of the nebulizer, metered-dose 57. Tang BC et al. Biodegradable polymer
GLP-1 (MKC253): proof-of-concept inhaler, and dry powder inhaler. nanoparticles that rapidly penetrate
studies in healthy normal volunteers Respir Care 2005; 50: 1313–1321. the human mucus barrier. Proc Natl
and in patients with type 2 diabetes. 45. Selroos O et al. Use of dry powder Acad Sci USA 2009; 106: 19268–
Clin Pharmacol Ther 2010; 88: 243– inhalers in acute exacerbations of 19273.
250. asthma and COPD. Ther Adv Respir 58. Mura S et al. Biodegradable nanopar-
33. Peyrot M, Rubin RR. Patient-reported Dis 2009; 3: 81–91. ticles meet the bronchial airway
outcomes in adults with type 2 diabe- 46. Lareau SC, Yawn BP. Improving barrier: how surface properties affect
tes using mealtime inhaled techno- adherence with inhaler therapy in their interaction with mucus and epi-
sphere insulin and Basal insulin COPD. Int J Chron Obstruct Pulmon thelial cells. Biomacromolecules 2011;
versus premixed insulin. Diabetes Dis 2010; 5: 401–406. 401–406. 12: 4136–4143.

© 2012 The Authors. JPP © 2012


1232 Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235
Francesca Ungaro et al. PLGA carriers for inhalation

59. Chadwick S et al. Delivery strategies studies on aerodynamic properties of DL-lactide/glycolide copolymer
to enhance mucosal vaccination. dry powder inhaler formulations. (PLGA) nanospheres to prolong
Expert Opin Biol Ther 2009; 9: 427– Drug Dev Ind Pharm 2011; 37: 1376– hypoglycemic effect. J Control Release
440. 1386. 1999; 62: 279–287.
60. Chadwick S et al. Nanotechnology 70. Devrim B et al. Preparation and 80. Muttil P et al. Immunization of
solutions for mucosal immunization. evaluation of PLGA microparticles as guinea pigs with novel hepatitis B
Adv Drug Deliv Rev 2010; 62: 394–407. carrier for the pulmonary delivery of antigen as nanoparticle aggregate
61. Lam JK et al. Pulmonary delivery of rhIL-2: I. Effects of some formulation powders administered by the pulmo-
therapeutic siRNA. Adv Drug Deliv parameters on microparticle charac- nary route. AAPS J 2010; 12: 330–337.
Rev. Epub 2011 February 26. teristics. J Microencapsul 2011; 28: 81. Sung JC et al. Formulation and
62. Nguyen J et al. Fast degrading polyes- 582–594. pharmacokinetics of self-assembled
ters as siRNA nano-carriers for pul- 71. Yoo NY et al. Antioxidant encapsu- rifampicin nanoparticle systems for
monary gene therapy. J Control lated porous poly(lactide-co- pulmonary delivery. Pharm Res 2009;
Release 2008; 132: 243–251. glycolide) microparticles for 26: 1847–1855.
63. Ungaro F et al. Biodegradable par- developing long acting inhalation 82. Tomoda K et al. Preparation and
ticles for local and prolonged delivery system. Colloids Surf B Biointerfaces properties of inhalable nanocompos-
of an oligonucleotide decoy to nuclear 2011; 88: 419–424. ite particles for treatment of lung
factor-kB in the lung. In: Dalby RN 72. Koushik K et al. Pulmonary delivery cancer. Colloids Surf B Biointerfaces
et al, ed. RDD Europe 2011. Rich- of deslorelin: large-porous PLGA par- 2009; 71: 177–182.
mond, VA: RDD Online, 2011: 511– ticles and HPbetaCD complexes. 83. Ungaro F et al. Dry powders based on
513. Pharm Res 2004; 21: 1119–1126. PLGA nanoparticles for pulmonary
64. Jensen DK et al. Design of an inhal- 73. Rawat A et al. Inhalable large porous delivery of antibiotics: modulation of
able dry powder formulation of microspheres of low molecular weight encapsulation efficiency, release rate
DOTAP-modified PLGA nanopar- heparin: in vitro and in vivo evalua- and lung deposition pattern by hydro-
ticles loaded with siRNA. J Control tion. J Control Release 2008; 128: 224– philic polymers. J Control Release
Release 2012; 157: 141–148. 232. 2012; 157: 149–159.
65. De Rosa G et al. Use of additives in the 74. Gupta V, Ahsan F. Influence of PEI as 84. Kim H et al. Highly porous large
design of Poly(Lactide-Co-Glycolide) a core modifying agent on PLGA poly(lactic-co-glycolic acid) micro-
microspheres for drug delivery. In: microspheres of PGE1, a pulmonary spheres adsorbed with palmityl-
Ravi Kumar MNV, ed. Handbook of selective vasodilator. Int J Pharm acylated exendin-4 as a long-acting
Particulate Drug Delivery. Stevenson 2011; 413: 51–62. inhalation system for treating diabe-
Ranch, CA: American Scientific Pub- 75. Gupta V et al. Feasibility study of tes. Biomaterials 2011; 32: 1685–1693.
lisher, 2008: 61–91. aerosolized prostaglandin E1 micro- 85. Malik DK et al. Recent advances in
66. Jain AK et al. Engineered PLGA nano- spheres as a noninvasive therapy for protein and peptide drug delivery
particles: an emerging delivery tool in pulmonary arterial hypertension. J systems. Curr Drug Deliv 2007; 4: 141–
cancer therapeutics. Crit Rev Ther Pharm Sci 2010; 99: 1774–1789. 151.
Drug Carrier Syst 2011; 28: 1–45. 76. Kim H et al. Albumin-coated porous 86. Alonso MJ et al. Determinants of
67. Hamishehkar H et al. Pharmacoki- hollow poly(lactic-co-glycolic acid) release rate of tetanus vaccine from
netics and pharmacodynamics of microparticles bound with palmityl- polyester microspheres. Pharm Res
controlled release insulin loaded acylated exendin-4 as a long-acting 1993; 10: 945–953.
PLGA microcapsules using dry inhalation delivery system for the 87. Zhu G et al. Stabilization of proteins
powder inhaler in diabetic rats. treatment of diabetes. Pharm Res encapsulated in injectable poly
Biopharm Drug Dispos 2010; 31: 189– 2011; 28: 2008–2019. (lactide- co-glycolide). Nat Biotechnol
201. 77. Pandey R et al. Poly (DL-lactide-co- 2000; 18: 52–57.
68. Hamishehkar H et al. Effect of carrier glycolide) nanoparticle-based inhal- 88. van de WM et al. Protein instability in
morphology and surface characteris- able sustained drug delivery system poly(lactic-co-glycolic acid) micro-
tics on the development of respirable for experimental tuberculosis. J Anti- particles. Pharm Res 2000; 17: 1159–
PLGA microcapsules for sustained- microb Chemother 2003; 52: 981–986. 1167.
release pulmonary delivery of insulin. 78. Thomas C et al. Aerosolized PLA and 89. van der Walle CF et al. Current
Int J Pharm 2010; 389: 74–85. PLGA nanoparticles enhance approaches to stabilising and analys-
69. Devrim B et al. Preparation and humoral, mucosal and cytokine ing proteins during microencapsula-
evaluation of poly(lactic-co-glycolic responses to hepatitis B vaccine. Mol tion in PLGA. Expert Opin Drug Deliv
acid) microparticles as a carrier for Pharm 2011; 8: 405–415. 2009; 6: 177–186.
pulmonary delivery of recombinant 79. Kawashima Y et al. Pulmonary deliv- 90. Pistel KF, Kissel T. Effects of salt
human interleukin-2: II. In vitro ery of insulin with nebulized addition on the microencapsulation

© 2012 The Authors. JPP © 2012


Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235 1233
PLGA carriers for inhalation Francesca Ungaro et al.

of proteins using W/O/W double 101. Dailey LA et al. Investigation of the 113. Giovagnoli S et al. Preparation of
emulsion technique. J Microencapsul proinflammatory potential of biode- large porous biodegradable micro-
2000; 17: 467–483. gradable nanoparticle drug delivery spheres by using a simple double-
91. Ungaro F et al. Cyclodextrins in the systems in the lung. Toxicol Appl Phar- emulsion method for capreomycin
production of large porous particles: macol 2006; 215: 100–108. sulfate pulmonary delivery. Int J
development of dry powders for the 102. Hara K et al. Histological examina- Pharm 2007; 333: 103–111.
sustained release of insulin to the tion of PLGA nanospheres for 114. De Rosa G et al. Long-term
lungs. Eur J Pharm Sci 2006; 28: 423– intratracheal drug administration. Int release and improved intracellular
432. J Pharm 2008; 356: 267–273. penetration of oligonucleotide-
92. Ungaro F et al. Engineering gas- 103. Rytting E et al. In vitro and in vivo polyethylenimine complexes entrap-
foamed large porous particles for performance of biocompatible ped in biodegradable microspheres.
efficient local delivery of macro- negatively-charged salbutamol- Biomacromolecules 2003; 4: 529–536.
molecules to the lung. Eur J Pharm Sci loaded nanoparticles. J Control 115. Vicennati P et al. Polyethylenimine in
2010; 41: 60–70. Release 2010; 141: 101–107. medicinal chemistry. Curr Med Chem
93. O’Hara P, Hickey AJ. Respirable 104. Beck-Broichsitter M et al. Pulmo- 2008; 15: 2826–2839.
PLGA microspheres containing nary targeting with biodegradable 116. De Rosa G et al. Poly(lactide-co-
rifampicin for the treatment of tuber- salbutamol-loaded nanoparticles. J glycolide) microspheres for the con-
culosis: manufacture and character- Aerosol Med Pulm Drug Deliv 2010; trolled release of oligonucleotide/
ization. Pharm Res 2000; 17: 955–961. 23: 47–57. polyethylenimine complexes. J Pharm
94. Suarez S et al. Respirable PLGA micro- 105. Hassan MS, Lau R. Effect of particle Sci 2002; 91: 790–799.
spheres containing rifampicin for the formulation on dry powder inhala- 117. Simberg D et al. DOTAP (and other
treatment of tuberculosis: screening tion efficiency. Curr Pharm Des 2010; cationic lipids): chemistry, biophysics,
in an infectious disease model. Pharm 16: 2377–2387. and transfection. Crit Rev Ther Drug
Res 2001; 18: 1315–1319. 106. Edwards DA et al. Recent advances Carrier Syst 2004; 21: 257–317.
95. Hirota K et al. Delivery of rifampicin- in pulmonary drug delivery using 118. Andrade F et al. Nanocarriers for pul-
PLGA microspheres into alveolar large, porous inhaled particles. J Appl monary administration of peptides
macrophages is promising for treat- Physiol 1998; 85: 379–385. and therapeutic proteins. Nanomedi-
ment of tuberculosis. J Control Release 107. Arnold MM et al. NanoCipro cine (Lond) 2011; 6: 123–141.
2010; 142: 339–346. encapsulation in monodisperse 119. Hillaireau H, Couvreur P. Nanocarri-
96. Doan TV, Olivier JC. Preparation of large porous PLGA microparticles. ers’ entry into the cell: relevance to
rifampicin-loaded PLGA micro- J Control Release 2007; 121: 100–109. drug delivery. Cell Mol Life Sci 2009;
spheres for lung delivery as aerosol by 108. Kim HK et al. Biodegradable poly- 66: 2873–2896.
premix membrane homogenization. meric microspheres with ‘open/ 120. Astete CE, Sabliov CM. Synthesis and
Int J Pharm 2009; 382: 61–66. closed’ pores for sustained release of characterization of PLGA nanopar-
97. Ohashi K et al. One-step preparation human growth hormone. J Control ticles. J Biomater Sci Polym Ed 2006;
of rifampicin/poly(lactic-co-glycolic Release 2006; 112: 167–174. 17: 247–289.
acid) nanoparticle-containing man- 109. Kwon MJ et al. Long acting porous 121. Mora-Huertas CE et al. Polymer-
nitol microspheres using a four-fluid microparticle for pulmonary pro- based nanocapsules for drug delivery.
nozzle spray drier for inhalation tein delivery. Int J Pharm 2007; 333: Int J Pharm 2010; 385: 113–142.
therapy of tuberculosis. J Control 5–9. 122. Sung JC et al. Nanoparticles for drug
Release 2009; 135: 19–24. 110. Yang Y et al. Development of highly delivery to the lungs. Trends Biotech-
98. Pulliam B et al. Design of porous large PLGA microparticles for nol 2007; 25: 563–570.
nanoparticle-based dry powder pul- pulmonary drug delivery. Biomateri- 123. Tsapis N et al. Trojan particles: large
monary vaccines. Expert Opin Drug als 2009; 30: 1947–1953. porous carriers of nanoparticles for
Deliv 2007; 4: 651–663. 111. Oh YJ et al. Preparation of drug delivery. Proc Natl Acad Sci USA
99. Coowanitwong I et al. Slow release budesonide-loaded porous PLGA 2002; 99: 12001–12005.
formulations of inhaled rifampin. microparticles and their thera- 124. Gomez-Gaete C et al. Dexamethasone
AAPS J 2008; 10: 342–348. peutic efficacy in a murine asthma acetate encapsulation into Trojan par-
100. De Stefano D et al. Sustained inhibi- model. ticles. J Control Release 2008; 128:
tion of IL-6 and IL-8 expression by J Control Release 2011; 150: 56–62. 41–49.
decoy ODN to NF-kappaB delivered 112. Thomas C et al. Particle size influ- 125. Tomoda K et al. Preparation and
through respirable large porous par- ences the immune response produced properties of inhalable nanocompos-
ticles in LPS-stimulated cystic fibrosis by hepatitis B vaccine formulated in ite particles: effects of the size, weight
bronchial cells. J Gene Med 2011; 13: inhalable particles. Pharm Res 2010; ratio of the primary nanoparticles
200–208. 27: 905–919. in nanocomposite particles and

© 2012 The Authors. JPP © 2012


1234 Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235
Francesca Ungaro et al. PLGA carriers for inhalation

temperature at a spray-dryer inlet siRNA-containing PLGA nanopar- logical review of lactose to support
upon properties of nanocomposite ticles intended for inhalation. J clinical administration by inhalation.
particles. Colloids Surf B Biointerfaces Control Release 2010; 142: 138–145. Food Chem Toxicol 1997; 35: 719–
2008; 64: 70–76. 128. Yamamoto H et al. Engineering 733.
126. Tomoda K et al. Preparation and of poly(DL-lactic-co-glycolic acid) 130. Wills PJ. Inhaled mannitol in cystic
properties of inhalable nanocompos- nanocomposite particles for dry fibrosis. Expert Opin Investig Drugs
ite particles: effects of the temperature powder inhalation dosage forms of 2007; 16: 1121–1126.
at a spray-dryer inlet upon the pro- insulin with the spray-fluidized bed 131. Yang Y et al. Mannitol-guided deliv-
perties of particles. Colloids Surf B granulating system. Advanced Powder ery of Ciprofloxacin in artificial cystic
Biointerfaces 2008; 61: 138–144. Technology 2007; 18: 215–228. fibrosis mucus model. Biotechnol
127. Jensen DM et al. Spray drying of 129. Baldrick P, Bamford DG. A toxico- Bioeng 2011; 108: 1441–1449.

© 2012 The Authors. JPP © 2012


Royal Pharmaceutical Society 2012 Journal of Pharmacy and Pharmacology, 64, pp. 1217–1235 1235

You might also like