Sie sind auf Seite 1von 18

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/269177119

Inhaled formulations and pulmonary drug


delivery systems for respiratory infections

Article in Advanced drug delivery reviews October 2014


DOI: 10.1016/j.addr.2014.10.022 Source: PubMed

CITATIONS READS

28 600

6 authors, including:

Qi Zhou Sharon Shui Yee Leung


Purdue University University of Sydney
48 PUBLICATIONS 601 CITATIONS 29 PUBLICATIONS 281 CITATIONS

SEE PROFILE SEE PROFILE

Thaigarajan Parumasivam Hak-Kim Chan


Universiti Sains Malaysia University of Sydney
21 PUBLICATIONS 82 CITATIONS 297 PUBLICATIONS 6,427 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Understanding Respiratory Pharmacology of Polymyxins View project

Antibiotics, Microbiology View project

All content following this page was uploaded by Sharon Shui Yee Leung on 15 February 2015.

The user has requested enhancement of the downloaded file.


ADR-12687; No of Pages 17
Advanced Drug Delivery Reviews xxx (2014) xxxxxx

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews


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

Inhaled formulations and pulmonary drug delivery systems for


respiratory infections
Qi (Tony) Zhou a, Sharon Shui Yee Leung a, Patricia Tang a, Thaigarajan Parumasivam a,
Zhi Hui Loh b, Hak-Kim Chan a,
a
Advanced Drug Delivery Group, Faculty of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia
b
GEA-NUS Pharmaceutical Processing Research Laboratory, Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore

a r t i c l e i n f o a b s t r a c t

Available online xxxx Respiratory infections represent a major global health problem. They are often treated by parenteral administra-
tions of antimicrobials. Unfortunately, systemic therapies of high-dose antimicrobials can lead to severe adverse
Keywords: effects and this calls for a need to develop inhaled formulations that enable targeted drug delivery to the airways
Inhaled antibiotics with minimal systemic drug exposure. Recent technological advances facilitate the development of inhaled
Pharmaceutical aerosol anti-microbial therapies. The newer mesh nebulisers have achieved minimal drug residue, higher aerosolisation
Nebulisation
efciencies and rapid administration compared to traditional jet nebulisers. Novel particle engineering and
Dry powder inhaler
Particle engineering
intelligent device design also make dry powder inhalers appealing for the delivery of high-dose antibiotics. In
Nanoparticles view of the fact that no new antibiotic entities against multi-drug resistant bacteria have come close to
Liposomes commercialisation, advanced formulation strategies are in high demand for combating respiratory super bugs.
Polymeric particles 2014 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2. Inhaled anti-bacterial formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.1. Wet aerosol formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.2. Dry powder formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.2.1. Powder production and particle engineering . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.2.2. Stability of spray-dried powders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.3. Liposomal formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.3.1. Amikacin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.3.2. Ciprooxacin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.3.3. Polymyxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.3.4. Proliposomes for anti-tuberculosis agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.4. Polymeric formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.4.1. Ciprooxacin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.4.2. Levooxacin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.4.3. Tobramycin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.4.4. Anti-tuberculosis agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.5. Nanoparticle formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.5.1. Amikacin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.5.2. Ciprooxacin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.5.3. Tobramycin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.5.4. Vancomycin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

This review is part of the Advanced Drug Delivery Reviews theme issue on Inhaled antimicrobial chemotherapy for respiratory tract infections: Successes, challenges and the road
ahead.
Corresponding author. Tel.: +61 2 9351 3054; fax: +61 2 9351 4391.
E-mail address: kim.chan@sydney.edu.au (H.-K. Chan).

http://dx.doi.org/10.1016/j.addr.2014.10.022
0169-409X/ 2014 Elsevier B.V. All rights reserved.

Please cite this article as: Q.(T.) Zhou, et al., Inhaled formulations and pulmonary drug delivery systems for respiratory infections, Adv. Drug Deliv.
Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.10.022
2 Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxxxxx

2.6. Combination formulations of antibiotics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0


2.6.1. Liquid formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.6.2. Powder formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.7. Combination formulations containing non-antibiotic adjuvants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.8. Bacteriophage formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3. Inhaled antiviral formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3.1. Zanamivir . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3.2. Laninamivir . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3.3. Anti-inuenza drugs under development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4. Inhaled anti-fungal formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.1. Amphotericin B . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.2. Itraconazole . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.3. Voriconazole . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

1. Introduction Most inhaled antibiotic products focus on the treatment of chronic


respiratory infections such as those in cystic brosis (CF) patients, an
Lower respiratory infections are particularly problematic [1], being autosomal recessive multi-system condition characterized by recurrent
the top leading cause of death in the developing countries and the respiratory infections [11]. Clinical in vivo data have shown that inhaled
third leading cause of death world-wide [2]. Apart from the high antibiotics can decrease the colonisation of Gram-negative bacteria such
mortality and morbidity rates, respiratory infections are also causing as Pseudomonas aeruginosa and delay the onset of infections [12]. Conse-
an escalating nancial burden to the global healthcare system. quently, improved lung function and quality of life can be achieved
In general, respiratory infections are difcult to treat because microbes along with reduced morbidities and hospital admissions. Nebulised
reside deep in the airways where only a small proportion of drug can ac- antibiotics can also be used for non-CF bronchiectasis although no
cess after traditional oral or parenteral administration. Consequently, high product has yet been approved for this indication. Compared to CF
doses of drugs are required to maintain drug levels above their minimum patients, the nebulisation of antibiotics may cause more severe local
inhibitory concentrations (MIC) at the infection sites [3]. It has been re- irritation, including bronchospasm, coughing and wheezing in patients
ported that less than 4% of colistin methanesulfonate, an inactive prodrug with non-CF bronchiectasis. Pre-treatment with bronchodilators or
of colistin, was converted to its active form after intravenous administra- encapsulation of drugs in liposomes may reduce the occurrences or
tion [4]. To achieve colistin concentrations above its MIC at the site of in- severity of these adverse events [10].
fection in airways, high doses of colistin methanesulfonate would have to
be administered parenterally to patients. Unfortunately, systemic expo-
2.1. Wet aerosol formulations
sure of high dose colistin can cause severe neurotoxicity and nephrotoxi-
city in up to 60% of treated patients [5]. Delivery of antibiotics directly to
Nebulisation of antibiotic solutions or suspensions is commonly
the respiratory tract provides an attractive solution in such situations be-
employed for the treatment of respiratory infections in both adult and
cause it allows higher drug concentrations to be achieved at the target
children [9,13]. During nebulisation, antibiotic liquid aerosols are
site with lower systemic exposures [6]. A pharmacokinetic study involv-
generated by mechanical or electrical mechanisms. Recent advances in
ing the nebulisation of colistin methanesulfonate solution (2 million IU)
nebuliser design have been reviewed elsewhere [9]. Briey, convention-
in cystic brosis patients clearly demonstrated that high drug concentra-
al jet nebulisers are bulky and generally have low drug delivery efcien-
tions could be achieved in the sputum (Cmax 6 mg/l) and maintained
cies [14], whilst vibrating mesh nebulisers exhibit minimal residual
above MIC90 for a prolonged period (e.g. 12 h) with negligible systemic
volume, rapid output and improved drug delivery efciencies despite
drug exposure (Cmax b0.3 mg/l) [7].
its higher purchase price. Other emerging technologies for nebulisation
In this review, innovative formulation strategies of inhaled drug ther-
include surface acoustic wave microuidic atomisation, which
apies for the treatment of respiratory infections are discussed. As devices
was shown to have a high delivery efciency, with FPF ranging from
for inhaled formulations have been reviewed previously [8] and clinical
7080% [15]. This new technology causes minimal damage to shear-
indications of inhaled anti-microbials are discussed in another article of
sensitive biomolecules of plasmid DNA and antibodies [16], thus solving
this issue [9], they are only briey described here wherein it is relevant
the problem of drug loss during high shear nebulisation. Today, drug
to the formulation design.
delivery by nebulisation can also be optimised by digital software
control and performance feedback systems. For example, the
nebulisation of a solution of diethylenetriaminepentaacetic acid in
2. Inhaled anti-bacterial formulations
saline using the I-neb nebuliser equipped with the Adaptive Aerosol
Delivery system resulted in a reasonably high FPF emitted (b4.6 m)
To date, only tobramycin and aztreonam solutions have been
of 56.8% [17].
approved for nebulisation in USA. Nebulised colistin methanesulfonate
solution (also known as colistimethate sodium, CMS) has been
approved only in some European countries. Other nebulised antibiotics 2.2. Dry powder formulations
include amikacin, ciprooxacin and levooxacin, all of which are under-
going clinical trials. For dry powder formulations, only tobramycin Dry powder inhaler (DPI) devices are generally small and portable.
(TOBI podhaler) and colistin methanesulfonate (Colobreathe) The duration of treatment for DPIs is also short [1820] and this results
have been approved in USA and Europe, respectively. Dry powder in improved patient compliance [21]. Dry powder mixtures of drugs
formulations of ciprooxacin and vancomycin are in the advanced and/or excipients may also be chemically more stable and less suscepti-
development stage [10]. ble to microbial contamination than corresponding liquid formulations

Please cite this article as: Q.(T.) Zhou, et al., Inhaled formulations and pulmonary drug delivery systems for respiratory infections, Adv. Drug Deliv.
Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.10.022
Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxxxxx 3

[2224]. These advantages contribute to the superior treatment ef- after spray drying or milling. McConville and Son developed a novel
ciencies and patient adherence of DPIs as exemplied by the colistin dry powder formulation of rifampicin by recrystallising the drug into a
methanesulfonate and tobramycin DPIs [19]. However, some powder ake-like crystal hydrate using anhydrous ethanol [41]. This formula-
formulations may be physically unstable at under conditions of high hu- tion was chemically stable for up to nine months under ambient condi-
midity, which is discussed in Section 2.2.2. tions, unlike amorphous rifampicin which underwent approximately
26% degradation during storage. Chan et al. also developed inhalable,
2.2.1. Powder production and particle engineering elongated crystals of rifapentine, a long-acting derivative of rifampicin
Drug particles with aerodynamic diameters ranging from 15 m are [42]. The crystalline particles exhibited a high FPF of 83.2% determined
optimal for lung deposition. To obtain ne drug powders in this size via the Aerolizer device at a ow rate of 100 l/min, and were also
range, jet milling is traditionally adopted by the pharmaceutical indus- chemically more stable than its amorphous counterpart [42].
try. However, jet-milled powder is notoriously cohesive because of the Surface coating of cohesive ne particles with force control agents
high surface energies of the milled particles [25]. For low-dose drugs (e.g. magnesium stearate or sodium stearate) can potentially improve
used for the treatment of asthma or chronic obstructive pulmonary dis- powder owability and aerosolisation [43,44]. Magnesium stearate
ease, coarse carrier particles are often added to improve powder ow. has been approved for inhalation by FDA (Foradil Certihaler,
However, this approach is unfavourable for high-dose antibiotics as ad- BREO ELLIPTA and ANORO ELLIPTA) and UK (Pulmicort CFC-
ditional excipients will increase powder volume which may necessitate free Inhaler). Qualitative and quantitative data from surface chemistry
an excessive number of inhalation manoeuvres or makes the multi-dose measurements have demonstrated that the deposition of an ultra-thin
inhaler bulky. Therefore, particle engineering becomes an attractive so- coating layer (in the nanometer-scale) of magnesium stearate on jet-
lution for the production of carrier-free (or with minimum carrier) milled drug powders by mechanical dry coating can bring a substantial
inhalable powders of antibiotics with good owability and superior reduction in their cohesiveness [4346]. Tobramycin powders coated
aerosolisation properties [26]. with 1% w/w of sodium stearate via spray drying also achieved a high
Spray drying is a prime example of a particle engineering technique FPF total (particles b5 m over the recovered dose) of 84.3 2.0%
suitable for the efcient production of inhalable particles with scale-up compared to 27.1 1.9% for pure spray-dried tobramycin, measured
capability. It has been used for the manufacturing of inhaled insulin via the Aerolizer device at a ow rate of 60 l/min [47]. It is noteworthy
(Exubera, Pzer), tobramycin (TOBI podhaler, Novartis) and man- that the choice of excipient is critical to the aerosolisation performance
nitol (Aridol, Pharmaxis). of coated powders. Belotti et al. showed that the addition of PEG-32
Leucine is a recognised dispersibility enhancer that is commonly stearate, a surface-active excipient, decreased the FPD of spray-dried
added into spray-drying feed solution or suspension [27,28]. Weaker amikacin powder [48].
particulate interactions are achieved with low surface-energy leucine Advanced particle engineering techniques and intelligent device
forming a rough shell or coating on the outer surface of spray-dried par- designs have contributed to much improved DPI therapies of antibiotics.
ticles [2931]. Porous particles of tobramycin and ciprooxacin pro- However, adverse events of cough and throat irritation are common for
duced by spray drying emulsion formulations of these drugs via the inhaled therapy of high-dose antibiotics in either wet or dry form. There
PulmoSphere technology have demonstrated satisfactory ow and is a long-term argument on whether wet form is more tolerable by the
aerosolisation performance [3234]. The PulmoSphere technology in- patients; however, to date there is no sufcient data from well-designed
volves the preparation of stable emulsions of peruorooctyl bromide clinical studies to conclude. On the other hand, the nature of drug plays
(Perubron) in water containing a pre-dissolved drug by high- a more important role in the severity of local side effects. This topic has
pressure homogenisation, followed by spray drying the emulsion. been discussed in detail in other articles of this issue [9,49].
Aerosolisation performances of PulmoSphere tobramycin formula-
tions were consistent at different temperatures (1040 C), relative hu- 2.2.2. Stability of spray-dried powders
midities (1065%) and airow rates (4085 l/min) [35]. Such high Amorphous or partly amorphous forms of many small molecule
aerosolisation efciencies translate to consistent drug deposition pat- drugs produced in the spray drying process may be unstable [5052].
terns in patients with varying inhalation abilities [36]. Additionally, Thereby one of concerns on spray-dried drug powders pertains to
the good powder owability of the spherical and porous spray-dried their physical and chemical instabilities, particularly for the reservoir-
particles reduced the need for carriers to aid manufacturing and type inhalers. In an early study, Adi et al. reported that spray-dried cip-
dispersion. rooxacin powders were amorphous and absorbed signicant amounts
Hollow particles of capreomycin (a second-line anti-tuberculosis of water of up to 7.9% w/w when exposed to a relative humidity of 50%
drug) with corrugated surfaces have been produced by spray drying a [53]. Re-crystallisation occurred at a higher relative humidity of 70%,
feed containing 80% capreomycin and 20% leucine in a co-solvent of demonstrating moisture-induced instability. Co-spray-dried combina-
ethanol and water (1:1) [37]. The powder formulation was stable at tion powders of ciprooxacin with 50% w/w mannitol reduced drug
4 C for three months, but physically and chemically unstable after hygroscopicity and prevented the re-crystallisation of amorphous
one month of storage under accelerated conditions of 40 C and 75% ciprooxacin [53]. However, the ip side of this is that the addition of
relative humidity. This was attributed to the absorption of signicant 50% excipients would signicantly increase the total amount of powder
amounts of water during storage [37]. In a Phase I clinical study, required for inhalation and thus may impact convenience.
administration of the highest dose (300 mg) of inhaled capreomycin Recently, Zhou et al. produced carrier-free colistin powders (a
was found to provide a serum concentration of drug above MIC against polypeptide) by spray drying. The powders exhibited high FPF total
Mycobacterium tuberculosis (2 mg/l) for only a short period of approxi- (b5 m over the recovered dose) values of N 80%, determined via the
mately 2 h, with a T1/2 of 4.8 h [38]. In this study, drug concentration in Aerolizer at a ow rate of 100 l/min [54]. The superior aerosolisation
local airways or in sputum was not measured. Inhaled therapy might performance of the spray-dried colistin powders is attributed to their
generate a high concentration of capreomycin in respiratory tracts rough particle surfaces (Fig. 1a) and low surface energies [55]. It is
although systemic drug concentration was low, which deserves noted that unlike many small molecule drugs that may re-crystallise
further investigation. Stable, inhalable porous para-aminosalicylic during storage, the spray-dried amorphous colistin powders remained
acid particles have also been produced by spray drying with 1,2- stable under ambient conditions, indicating that spray drying was suit-
dipalmitoyl-sn-glycero-3-phosphocholine in ethanol [39] or with able for the production of stable, inhalable colistin powder. It was also
ammonium carbonate [40]. found that both the supplied and spray-dried colistin powders absorbed
Re-crystallisation enables the production of inhalable crystalline up to 30% water when exposed to an elevated relative humidity of 90%.
drug powders that are otherwise unstable in their amorphous forms Airtight packaging for each individual dose is highly recommended to

Please cite this article as: Q.(T.) Zhou, et al., Inhaled formulations and pulmonary drug delivery systems for respiratory infections, Adv. Drug Deliv.
Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.10.022
4 Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxxxxx

Fig. 1. SEM micrographs of spray-dried antibiotic powder formulations of: (a) pure colistin (reprint from [54] with permission from John Wiley and Sons); (b) combination of colistin and
rifampicin (reprint from [56] with permission from Springer); (c) combination of ciprooxacin hydrochloride, gatioxacin hydrochloride and lysozyme (reprint from [137] with permis-
sion from Elsevier); (d) combination of moxioxacin, rifapentine, pyrazinamide and leucine (reprint from [144] with permission from Elsevier).

avoid moisture induced instability [54]. In a subsequent study, by coat- (280 mg or 560 mg) was nebulised once-daily to cystic brosis patients
ing the hygroscopic colistin particle surfaces with hydrophobic rifampi- infected by P. aeruginosa for 28 days using the eFlow nebuliser [63].
cin, moisture-induced deterioration in aerosolisation performance of The outcomes of the study conrmed the acute tolerability, safety, biolog-
colistin could be circumvented [56]. Interestingly, data obtained from ical activity and efcacy of the inhaled therapy. Using the same nebuliser,
time-of-ight secondary ion mass spectrometry and X-ray photoelec- Li et al. reported a 3035% loss of entrapped amikacin with a FPF of 32.5%
tron spectroscopy showed that the surfaces of the composite drug par- upon nebulisation of Arikace [64]. The loss of encapsulated drug from
ticles were dominated by hydrophobic rifampicin although equivalent liposomes deserves further investigations with other nebuliser devices
ratios of two drugs were used [56]. in order to understand the role of nebuliser on the performance of liposo-
mal formulations.
2.3. Liposomal formulations
2.3.2. Ciprooxacin
Many inhaled antibiotics are water-soluble and can be absorbed Liposomal formulations enable sustained-release of ciprooxacin
readily into the systemic circulation, which are consequently cleared. [65,66] leading to reduced dosing frequencies and improved treatment
To maintain drug concentrations above their MICs in the respiratory convenience [67]. Clinical data have shown that a nebulised liposomal
tract, liposomal formulations of inhaled drugs are exploited to sustain formulation of ciprooxacin (Lipoquin) [68] and a combination of
drug release, prolong the action of drug in the infection sites and en- liposomal and free ciprooxacin (Pulmaquin) [67] were both efca-
hance bacterial killing. Sustained-release of antibiotics reduces dosing cious with well-accepted tolerability in CF and non-CF bronchitis pa-
frequency and improves patient compliance. In addition, liposomal for- tients. In vitro characterization of these liposomal ciprooxacin
mulations could potentially enable targeted delivery of antibiotics [57]. formulations has demonstrated liposome integrity following jet nebuliza-
When delivered to the respiratory tract, liposomes are engulfed and de- tion as measured by drug encapsulation, mean vesicle size distribution
graded by the macrophages with subsequent release of the antibiotics [69], and in vitro release prole [70]. The addition of surfactant (0.4%
within them [58] and this mechanism can be exploited to target intra- polysorbate 20) to these formulations resulted in a faster in vitro release
cellular infections [59]. Modication of liposomes with mannose could rate [66,71]. In contrast, other conventional liposomal preparations may
facilitate active targeting of macrophages with mannose receptors [60, have signicantly larger loss of drug encapsulation [64,72]. Additionally,
61]. Some liposomal formulations have been shown tolerated by inhala- the physicochemical properties of the liposomal formulations remained
tion and have a good safety prole, which has been discussed in a recent unchanged after storage at 28 C for up to two years [71].
review of the topic [62]. Table 1 summarises the key studies on inhaled A facile method was developed to produce inhaled liposomal formu-
liposomal formulations of anti-microbials. lations of ciprooxacin by dispersing a powder mixture of micronised
drug and excipients in saline [73,74]. Upon dispersed, over 90% of
2.3.1. Amikacin drug was encapsulated in the liposomes for the optimised formulations
Arikace is an inhaled liposomal formulation of amikacin, comprising but the encapsulation was reduced to approximately 50 to 55%
the drug encapsulated in neutral liposomes composed of dipalmitoyl- after nebulisation [73]. Such micronised powder exhibited relatively
phosphatidylcholine and cholesterol. In a Phase II clinical trial, Arikace high FPF of N50% when it was aerosolised by a deagglomeration rig at

Please cite this article as: Q.(T.) Zhou, et al., Inhaled formulations and pulmonary drug delivery systems for respiratory infections, Adv. Drug Deliv.
Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.10.022
Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxxxxx 5

60 l/min. The encapsulation efciency of aerosolised powder was

Drug encapsulation, vesicle size and in vitro [2,3]


Ref

[1]

[4]

[5]

[6]

[7]

[8]
approximately 35% for ciprooxacin [75]. In a subsequent study, the
encapsulation efciency of reconstituted formulation was improved to
93.5% in isotonic saline, 80% in bovine mucin, 75% in porcine mucus
and 73% in ve-fold-diluted ex vivo human cystic brosis patient

release are stable upon nebulization

24 times lower MICs against many


3035% loss of entrapped amikacin

Pseudomonas aeruginosa, Klebsiella


sputum [76].

Fast release with 50% of colistin


pneumoniae and Escherichia coli
reference and clinical strains of
In addition to sustained-drug release, liposomal ciprooxacin has
also demonstrated enhanced anti-microbial activity [77]. Gubernator
et al. reported that cationic liposomal ciprooxacin formulations

dissolved in 10 min
Faster release rate possessed 24 times lower MICs against most reference and clinical
after nebulisation

strains of P. aeruginosa, Klebsiella pneumoniae and Escherichia coli,


including a two-fold decrease in MIC against resistant strains of
Comments

High FPF
Low FPF
P. aeruginosa [78]. Enhanced anti-microbial effects of the cationic liposo-
mal formulation were attributed to the interaction of positively-charged
liposomes with the negatively-charged bacterial cell membrane [79].
Testing ow
rate (l/min)

Consequently, antibiotics in the liposomes accumulated in the


periplasm, leading to more anti-microbial molecules diffusing into the
cytoplasm [78,79].
12.5

12.5
N/A

N/A

N/A

60

60
63.5 (of the emitted

2.3.3. Polymyxins
aerosol, b4.95 m)

1535% (b4.4 m)
VMD of 3.74 m

Desai and co-workers have produced liposomal formulations of


polymyxin B by a thin-lm hydration technique, with dimyristoyl
phosphatidylglycerol and surfactants of Tween 80 and Span 20 [80].
32.5%

92.5%

The optimised formulation achieved an exceptionally high encapsula-


N/A

N/A
FPF

tion efciency of 100% even after nebulisation. However, it is interesting


to note the liposomal formulation had a much higher MIC (31.3 g/l)
Dispersion device
made in-house
Testing device

against P. aeruginosa (ATCC 27853) than pure polymyxin B (3.9


7.8 g/l) [80]. The possible reason for the reduced antimicrobial activity
Pari eFlow

Pari eFlow

Rotahaler

could be the negative particle surface charge of the liposomes. The


eFlow

N/A

N/A

consensus bactericidal mechanism of polymyxins is that: positively


charged drug molecules bind to negatively charged lipopolysaccharide
(DOTAP), 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine

of bacterial outer membrane and cause the rupture of bacterial cells


[81]. The binding to the lipopolysaccharide could be compromised for
negatively charged liposomes, leading to reduced antimicrobial activity.
1,2-Dioleoyloxy-3-trimethylammonium-propane

(DOPE), phosphatidyl-choline (PC), cholesterol

Further investigations are warranted to understand this issue by


Membrane extrusion Polysorbate 20, 0.4% (w/v), hydrogenated soy

comparing positively and negatively charged liposomes.


Membrane extrusion Hydrogenated soy phosphatidylcholine,

l- soybean phosphatidylcholine (SPC),

Using a dry lm method, Wallace et al. encapsulated colistin


sulphate and colistin methanesulfonate in dioleoylphosphatidylcholine
cholesterol from lanolin, mannitol
phosphatidylcholine, cholesterol

liposomes of approximately 180 nm in size and achieved encapsulation


Dioleoylphosphatidylcholine

efciencies of around 50% [82]. Contrary to expectations, the release of


Soya phosphatidylcholine

colistin from the liposomes was rapid, with 50% of colistin dissolved in
10 min and no further release was observed in 72 h. These ndings
differed from the sustained-release proles of colistin liposomes
Major excipient

reported by Wang et al. [83]. Wallace hypothesised that unlike typical


cholesterol

hydrophilic drugs, amphipathic colistin may have increased the


permeability of the liposomal bilayer, enabling more rapid release of
N/A

encapsulated colistin from the core of the liposomes. In a subsequent


study, incorporating colistin in liposomes of azithromycin also
Production method

enhanced the in vitro release of azithromycin and this was attributed


Thin lm method

Dry lm method
Summary of key liposomal formulations of inhaled antibiotics.

by spray drying

by spray drying

to the increased permeability of liposomes by colistin [84]. This study


Proliposomes

Proliposomes

opens up new research opportunities in the application of liposomal


formulations for the delivery of combination antibiotics to the lungs.
Nebulisation (Arikace) N/Aa

2.3.4. Proliposomes for anti-tuberculosis agents


Proliposomes are dry powders which can form liposomes upon
contact with water either before or after administration into the body
[85,86]. Rojanarat et al. developed spray-dried proliposomes of
isoniazid [87]. Data showed the proliposomes possessed a low FPF of
Ciprooxacin Nebulisation

Ciprooxacin Nebulisation
Formulation

Dry powder

Dry powder

1535% (b 4.4 m) due to particle agglomeration [87]. A recent study


showed that soya phosphatidylcholine proliposomes of rifapentine
N/A: not available.
Ciprooxacin Wet

Wet

produced using single-step spray drying, possessed mass median


aerodynamic diameters of 1.56 0.16 m and exhibited surprisingly
Rifapentine

good aerosolisation performance with FPF of 92.5 1.5% measured


Amikacin

Isoniazid
Colistin

via a low-efciency Rotahaler device at 60 l/min [88]. Hence, the choice


Table 1

Drug

of proliposome materials affects not just the drug release prole but also
a

aerosolisation performance.

Please cite this article as: Q.(T.) Zhou, et al., Inhaled formulations and pulmonary drug delivery systems for respiratory infections, Adv. Drug Deliv.
Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.10.022
6 Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxxxxx

One critical challenge in the assessment of inhaled sustained-release of many water-soluble antibiotics. Although polyvinyl alcohol (PVA)
formulations pertains to the selection of an appropriate in vitro and poly (lactide-co-glycolide) (PLGA) have been examined as
dissolution method. To date, there is neither regulatory guidelines on polymeric sustained-release agents in many studies in view of their
the dissolution testing of inhaled products nor any conclusive answers biocompatibilities and minimal toxicities in in vitro and in vivo studies
to what constitutes the best dissolution medium capable of mimicking [9395], the long-term persistence of these undissolved particles in
the pulmonary environment [89]. This creates a barrier for quick the lungs remains a concern as they may trigger pulmonary inamma-
prediction of formulation performance and development of generic tion and brosis [96]. Furthermore, as foreign particles in the lung
liposomal products. In May of 2014, FDA has called research grant would be removed either by mucociliary clearance or macrophage
applications to evaluate different in vitro release assays in terms of uptake, prolonged drug release exceeding i.e. 24 h by means of
their capacity in detecting formulation differences and predicting polymeric particles is extremely challenging in vivo; however, these
in vivo release of liposomal drug products. Recently, Cipolla developed may allow treatment that is targeting pulmonary macrophage (i.e. for
a dissolution method to evaluate the in vitro dissolution of inhaled treatment of latent tuberculosis). Hence, in vivo and clinical studies on
liposomal ciprooxacin formulations in the lungs [90]. Bovine serum the safety and efcacy of inhaled polymeric formulations are needed
was selected as the release agent that would trigger the release of to ll this knowledge gap. Key studies on polymeric formulations of
drug from the liposomes. The method was robust over a range of release inhaled antibiotics are listed in Table 2.
parameters including release agent, liposomal concentration, incuba-
tion temperature, and buffer pH [90]. An in vitro air-interface Calu-3 2.4.1. Ciprooxacin
cell model was also developed by Ong et al. to evaluate drug release Sustained-released powders of ciprooxacin and doxycycline have
and intracellular drug transport of liposomal ciprooxacin formulations been prepared by co-spray drying hydrochloride salts of both drugs
on the human epithelium cell lines [91]. Other challenges for the evalu- with PVA [97]. It was found that the incorporation of PVA slowed
ation of liposomal formulations include damage of liposome integrity down the release of both hydrophilic antibiotics, with less than 50% of
upon nebulisation [92] and differentiation of released and encapsulated the drugs dissolved in the pH 7.4 phosphate buffer for the PVA formula-
drug in vivo. Further fundamental studies are warranted to understand tion in 6 h compared to more than 90% dissolved for the pure drugs,
the drug delivery and therapeutic actions of liposomal formulations. measured by a modied Franz diffusion cell [97].

2.4. Polymeric formulations 2.4.2. Levooxacin


Sustained-release of levooxacin has also been achieved by encap-
Investigations into polymeric formulations of antibiotics for inhala- sulating the antibiotic in poly(caprolactone) and PLGA nanoparticles
tion have been conducted to address the rapid-release characteristics via an emulsicationsolvent-evaporation method [98]. Gradual release

Table 2
Summary of key polymeric formulations of inhaled antibiotics.

Drug Formulation Production method Major excipient Testing device FPF Testing Reference
ow rate
(l/min)

Ciprooxacin Dry powder Co-spray drying PVA Aerolizer 25.9% for 60 [9]
and ciprooxacin and
doxycycline 25.8% for
doxycycline
Levooxacin Dry powder Lipid-coated nanoparticles via an emulsication- PLGA, PVA, N/A N/A N/A [10,11]
solvent-evaporation method followed by spray drying phosphatidylcholine, L-
leucine
Tobramycin Dry powder Nanoparticle suspension by the emulsion/solvent PLGA, PVA, chitosan, Turbospin 3852% 60 [12]
diffusion method followed by spray drying alginate, lactose
Isoniazid Dry powder Chitosan microspheres followed by spray drying Chitosan, Cyclohaler 6070% 28.3 [13]
tripoliphosphate, lactose,
L-leucine
Isoniazid Dry powder Chitosan nanoparticles by ionic gelation method Chitosan, Cyclohaler 745% (b5.8 m), 60 [14]
followed by spray drying tripolyphosphate, lactose, 7.811%
mannitol or maldextrose, (b3.3 m)
L-leucine
Rifampicin Dry powder Recrystallization and coating with PLGA/PLA followed PLGA/PLA Aerolizer 2645% 60 [15]
by spray coating
Rifampicin Dry powder Amorphous matrix followed by spray drying PLGA/PLA Aerolizer 2333% 60 [15]
Rifampicin Dry powder Poly-(ethylene oxide)-block-distearoyl phoaphatidyl- mPEG-DSPE w/v) Pari LC Plus 40% 30 [16]
ethanolamine (mPEG-DSPE) nanoparticles followed by nebulizer
lyophilization for rehydration and nebulisation
Rifampicin Dry powder Solvent evaporation of single (w/o) and double PLGA, PVA A model DP-4 3370% 30 [17]
emulsion (w/o/w) with premix membrane dry powder
homogenization followed by freeze-drying insufator
Rifampicin Dry powder Microspheres using single emulsion (o/w) followed by PLGA, sucrose palmitate A model DP-4 52% 30 [18]
freeze-drying dry powder
insufator
Rifampicin Dry powder PLGA nanoparticle containing mannitol microspheres PLGA, mannitol Jethaler dual 35% 28.3 [19]
followed by four-uid nozzle spray drying chamber type
inhalation
device
Rifampicin Dry powder Spray drying PLGA-drug solution PLGA Cyclohaler 2232% 28.3 [20]
Rifampicin Dry powder Spray drying PLA-drug solution PLA Cyclohaler 5570% 28.3 [20]

Please cite this article as: Q.(T.) Zhou, et al., Inhaled formulations and pulmonary drug delivery systems for respiratory infections, Adv. Drug Deliv.
Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.10.022
Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxxxxx 7

of levooxacin-loaded poly(caprolactone) nanoparticles was main- nanoparticles [119,120]. Results showed that the MIC and minimum bac-
tained for 6 days whilst the PLGA nanoparticles released approximately teriostatic concentration of amikacin nanoparticles against P. aeruginosa
80% of the loaded drug in one day. Lipid-coated polymeric nanoparticles was less than half of the values for free amikacin, indicating enhanced
of levooxacin demonstrated improved biolm afnity and conse- anti-microbial activity [119,120].
quently, more superior anti-biolm activity than the pure drugs [99].
2.5.2. Ciprooxacin
2.4.3. Tobramycin Nanoparticle suspensions of ciprooxacin base were produced by
A chitosan-modied PLGA nanoparticle suspension of tobramycin sonicating a colloidal dispersion of drug in acetone with L-leucine
was prepared by the emulsion/solvent diffusion method [100]. added into the suspension as a matrix material. The inhalable aggre-
Sustained drug release was achieved in vitro in the pH 7.2 phosphate gates subsequently produced by freeze-drying were characterized by a
buffer for up to 30 days. The MIC of the nanoparticle suspension against high FPF total (b5.8 m) of 81% measured via a Monodose inhaler at a
P. aeruginosa was ten-fold higher than pure tobramycin, and severe ow rate of 30 l/min [121]. Furthermore, the nanoparticle agglomerates
in vitro cytotoxicity of the nanosuspension on human alveolar epithelial exhibited an accelerated dissolution prole compared to the supplied
cells (A549) was observed. Based on results obtained from the MTT ciprooxacin base powder [121].
assay, an approximate 82% reduction in cell viability was observed for
the blank chitosan-modied nanoparticle suspension compared to 2.5.3. Tobramycin
50% reduction for the tobramycin-loaded nanoparticles at a concentra- Nanoparticles of tobramycin were prepared by the conventional
tion of 1 mg/ml [100]. In the light of these ndings, caution should be high-pressure homogenisation approach in the presence of a surfactant
exercised when developing drug-loaded polymeric nanoparticles for (sodium glycocholate) to improve its dispersion [122]. The agglomer-
pulmonary delivery. ates formed by spray drying the drug nanosuspension, exhibited an
FPF (b 5 m relative to the loaded dose) of 61% when determined
2.4.4. Anti-tuberculosis agents using the Aerolizer at a ow rate of 100 l/min [122]. This was signi-
Micro- and nano-sized polymeric particles containing encapsulated ri- cantly higher than that of micronized tobramycin which was found to
fampicin, a rst-line anti-tuberculosis drug, have been investigated exten- be 36%.
sively. The ake-like dihydrate particles of rifampicin were coated with
PLGA or polylactide (PLA) using a spray dryer equipped with a multi- 2.5.4. Vancomycin
channel nozzle to reduce the initial drug release and protect the micro- Savara Pharmaceuticals has developed an inhalable dry powder
crystals from chemical decomposition [101]. The emulsionsolvent-evap- form of vancomycin hydrochloride (AeroVanc) [123125]. The
oration method has also been used to produce PLGA-rifampicin particles formulation of AeroVanc has not been disclosed but data from a
[102105]. Drug loading efciencies and particle size were found to be Phase I clinical study in healthy volunteers reported excellent tolerability
highly dependent on the shear forces and formulation parameters such and favourable pharmacokinetic proles [126].
as aqueous phase volume and polymer concentration [106].
Pre-mix membrane emulsication has also been employed [102, 2.6. Combination formulations of antibiotics
104,107]. In this technique, a polymer emulsion with narrow particle
distribution is generated through a Shirasu Porous Glass membrane Mono-therapy of antibiotics may lead to the development of
with known pore sizes into a continuous phase containing surfactants. antibiotic resistance [127]. Hence, combination therapies containing
Hu et al. showed that the initial burst release of rifampicin from PLGA different types of antibiotics have been extensively used clinically to
microparticles produced by this method could be avoided by coating prevent the emergence of drug resistance [128]. Drug combinations
with sodium alginate [108]. Other production methods include the should be carefully selected to exploit any potential synergism or even
electrostatic drop generation approach for the preparation of sodium antagonism in their anti-bacterial activities. An early study of the anti-
alginate microparticles loaded with 22% rifapentine [109] and an ionic microbial activity of a co-spray-dried combination of ciprooxacin and
gelation method for the production of isoniazid-loaded chitosan/ doxycycline hydrochloride (1:1) carried out based on the disc diffusion
tripolyphosphate nanoparticles [110]. method revealed minimal changes in the inhibition zone diameter
of the combination against Staphylococcus aureus, P. aeruginosa and
2.5. Nanoparticle formulations Streptococcus pyrogenes [129]. However, suppressive effects have been
reported for this combination against E. coli [130]. Therefore, both the
Inhalable nanoparticle formulations are advantageous for the measurement method and the target bacterial strain are critical in the
purposes of improving the solubility and dissolution rates of water- selection of synergistic combinations of antibiotics.
insoluble drugs as well as minimising premature mucociliary clearance
of drugs [111]. Inhaled nanoparticles could be exhaled during DPI 2.6.1. Liquid formulations
administration due to their extremely low mass. This problem has The combination of tobramycin with other antibiotics such as
been solved by formulating nanoparticles into inhalable micro- fosfomycin has resulted in synergistic anti-bacterial effects against
aggregates via spray drying [112114] or spray-freeze-drying [115, S. aureus and P. aeruginosa in both in vitro and in vivo models [131].
116] with mannitol [113,116], PVA, or leucine as matrix materials. The MacLeod et al. discovered that fosfomycin enhanced the uptake of
micro-sized aggregates generated by spray-freeze drying exhibited tobramycin in P. aeruginosa in a dose-dependent manner [132].
superior aerosolisation performance compared to those produced by Co-administration of tobramycin with clarithromycin [133,134] and
spray drying [112] and this was attributed to the porous structure and colistin [135] also exhibited enhanced anti-biolm activities. Results
low density of the particles [116,117]. However, the toxicities of inhaled of a Phase II clinical trial on nebulised tobramycin/fosfomycin in CF pa-
nanoparticles deserve further investigations. Key studies on nanoparti- tients conrmed the clinical anti-microbial efcacy against P. aeruginosa
cle formulations of inhaled antibiotics are listed in Table 3. [136].

2.5.1. Amikacin 2.6.2. Powder formulations


Varshosaz and co-workers optimised a solvent diffusion technique Lee et al. developed a binary formulation comprising hydrochloride
for the production of solid lipid nanoparticles of approximately salts of ciprooxacin and gatioxacin as well as a ternary (binary plus
150 nm in size with a high drug loading efciency of 88% [118]. Freeze lysozyme) combination of powders via spray drying (Fig. 1c) [137].
drying was employed to improve the long-term stability of the The binary combination showed a synergistic anti-microbial effect

Please cite this article as: Q.(T.) Zhou, et al., Inhaled formulations and pulmonary drug delivery systems for respiratory infections, Adv. Drug Deliv.
Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.10.022
8 Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxxxxx

against P. aeruginosa in a time-kill model [137]. The ternary combination [148]. Another ion chelator, lactoferrin, was reported to block biolm
resulted in an indifference interaction against P. aeruginosa, S. aureus, development of P. aeruginosa [149].
K. pneumoniae, and Acinetobacter baumannii but may have potential Osmotic agents such as mannitol may also be added as adjuvants. In
mucolytic effects [138] in CF patients which deserve further investigation. CF patients, pathogens can reside in the thick mucus which forms an
Combination powder formulations consisting of tobramycin and other impenetrable boundary preventing the entry of antibiotics. Using an
antibiotics including azithromycin, ceftazidime [139] or clarithromycin, in vitro articial mucus model, Yang et al. reported that the combination
were also produced by spray drying. Inhalable powders of tobramycin of mannitol with ciprooxacin enhanced its anti-microbial effects
and ceftazidime were spray-dried with dipalmitoylphosphatidylcholine, against P. aeruginosa [150]. Such effects were attributed to the presence
albumin, and lactose with the purpose of producing highly dispersible of mannitol which increased water inux to the articial mucus and
porous particles [139]. Contrary to expectations, the spray-dried subsequently improved ciprooxacin penetration through the mucus
combination particles were not porous, possessed relatively low FPF layer [150]. However, the dose of ciprooxacin in the proportion (5%
values (b 4.7 m over the nominal dose) of 2030% and displayed no w/w) that was investigated in combination with mannitol was low. If
synergistic anti-microbial action [139]. Spray drying suspended the actual dose of ciprooxacin as used in previous clinical trials
tobramycin in clarithromycin solution resulted in powders with (32.5 mg) [151] was combined with mannitol in a similar ratio, the
improved aerosolisation performance compared to the corresponding total powder mass of each dose could amount up to a total of 650 mg.
physical mixture [140]. The FPF (b4.7 m over the nominal dose), Further optimisation in the formulation may reduce the dose of total
measured via the Axahaler at a ow rate of 100 l/min, increased from powder. Other potential benets of adding mannitol to the inhaled
35% (tobramycin) and 31% (clarithromycin) for the physical mixture to antibiotic formulation include eradication of bacterial persistent cells
65% and 63% for tobramycin and clarithromycin, respectively, in the [152,153], improved airway clearance associated with CF [154] and
combination formulation [140]. non-CF bronchiectasis [155].
Selective combination powders of colistin and rifampicin were Other reported non-antibiotic components include deoxyribonu-
developed by co-spray drying based on apparent synergistic anti- clease (DNase, a mucolytic) with enhanced anti-microbial effects on
microbial activities against A. baumannii [56] and P. aeruginosa. A high P. aeruginosa in articial sputum [156] and beclomethasone dipropi-
aerosolisation efciency was achieved with an emitted dose of 96% onate for the treatment of respiratory infections in patients with
and FPF total of 92% measured via an Aerolizer device at 100 l/min chronic obstructive pulmonary diseases [157]. Nitric oxide donors
[56]. This was attributed to the wrinkled particle shape (Fig. 1b) and [158160] and D-amino acids [161] have also been shown in vitro
surface coating of rifampicin. The FPF total of pure colistin powder biolm-dispersal effects.
decreased from 80% to 63.2% when storage humidity increased from
60 to 75%. In strong contrast, the FPF total of the combination powder
was unchanged when relative humidity was increased. It was postulat- 2.8. Bacteriophage formulations
ed that the hydrophobic rifampicin precipitated earlier than colistin on
the particle surfaces during spray drying and this resulted in rifampicin Phage therapy represents a unique approach for the treatment of
dominating the surface composition of the particles and providing infections based on an anti-bacterial mechanism that is entirely
moisture protection for the combination powders. This was conrmed different from the traditional antibiotics. Bacteriophage is a type of
by surface chemistry measurements using X-ray photoelectron virus that infects and replicates itself inside bacterial cells. Phages
spectroscopy and time-of-ight secondary ion mass spectrometry [56]. have also acquired mechanisms to counter bacterial defences [162].
Inhalable formulations consisting of rst-line, second-line or a Furthermore, in many cases bacteria at the infection sites are protected
combination of both classes of anti-tuberculosis drugs have been well from antibiotic penetration by a biolm barrier. Phages are able to
studied to reect current treatment regimens [137,141144]. Advanced penetrate the biolm, replicate locally to kill bacteria and can also
formulations of polymeric particles have been extensively explored for potentially restore antibiotic efcacy [163165].
anti-tuberculosis drugs [141143,145]. In terms of maximising the Formulating bacteriophages as inhalable dry powders is generally
delivered dose to the patients, pure drug combinations are preferred. preferred to improve patient adherence and product stability. Making
A key concern in the development of combination formulations pertains biologically stable phages as dry powders is critical as stability impacts
to their physiochemical stabilities. For example, when rifampicin is on the cost and efcacy of treatment. Conventional powder technologies
combined with either isoniazid, pyrazinamide or both, degradation of such as mechanical milling are unsuitable as the energy and stresses
one or more drugs [146] was observed due to the formation of produced are known to cause damage to biopharmaceuticals such as
adduct-rifamycin isonicotinyl hydrazone [145]. A similar phenomenon proteins [166180]. A Canadian aerosol research group led by Finlay
was reported in the polymeric particles [145]. In a subsequent study and Vehring has undertaken pioneering studies on the formulation of
by Chan, isoniazid and rifampicin were replaced by moxioxacin and phages as powders for inhalation [181,182]. Inhalable powders of
rifapentine [137] and the powder formulation comprising this triple- bacteriophage KS4-M and KZ with lactose/lactoferrin (60:40, w/w)
combination of the anti-tuberculosis agents was prepared by co-spray were generated by lyophilisation followed by milling [182]. The
drying (Fig. 1d) [137]. Addition of 10% w/w leucine was found to be lyophilisation process exhibited an acceptable loss of titre with c. 2
essential to prevent overt crystallisation of pyrazinamide after log10s for KS4-M (from 2.3 0.5 1010 to 1.1 0.5 108) and c. 1
long-term storage [137]. log10 for KZ (from 1.7 0.4 109 to 7.8 3.3 108). However, the
FPF total measured at 60 l/min via the Aerolizer was relatively low, in
the range of 3234% over the loaded dose.
2.7. Combination formulations containing non-antibiotic adjuvants Improvements in the loss of titre and aerosolisation have been
achieved by low-temperature spray drying with protective excipients.
Besides the combination of various antibiotics, the incorporation of Dry powder formulations of bacteriophage KS4-M, KS14 and cocktails
non-antibiotic adjuvants in antibiotic formulations may potentially of phages KZ/D3 and KZ/D3/KS4-M, have been produced via spray
enhance their anti-microbial effects. Metallic cations (e.g. iron, calcium drying at an inlet temperature of 75 C and outlet temperatures ranging
and magnesium) are essential for bacterial growth, microbial adherence from 4045 C [181]. Casein sodium salt and -trehalose were added as
and biolm formation. Metal-ion chelators are hence useful for protective agents, surfactants like Tyloxapol and Pluronic F68 as wet
combating multi-drug resistant bacteria by binding to these ions [147] dispersing agents and L-leucine as a powder aerosolisation enhancer.
. A combination formulation consisting of an ion chelator, citrate and The optimised formulation had a total lung mass of approximately
taurolidine was shown to inhibit the growth of P. aeruginosa biolms 70% determined via the Aerolizer at 90 l/min and the titre loss was

Please cite this article as: Q.(T.) Zhou, et al., Inhaled formulations and pulmonary drug delivery systems for respiratory infections, Adv. Drug Deliv.
Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.10.022
Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxxxxx 9

Table 3
Key nanoparticle formulations for inhaled antibiotics.

Drug Formulation Production method Major Testing device FPF Testing Comments Reference
excipient ow rate
(l/min)

Amikacin Dry powder Lipid nanoparticles by solvent Sucrose, N/A N/A N/A Enhanced antimicrobial effects [2123]
diffusion technique followed by dextrose, against Pseudomonas aeruginosa
freeze-drying mannitol compared to pure drug
Ciprooxacin Dry powder Sonicating a colloidal dispersion of L-leucine Monodose inhaler 81% 30 Accelerated dissolution prole [24]
drug in acetone followed by freeze-
drying
Tobramycin Dry powder High-pressure homogenisation Sodium Aerolizer 61% 100 Improved dispersion compared to [25]
followed by spray drying glycocholate micronised drug
Vancomycin Dry powder Low density nanoparticle-clusters N/A A capsule-based 80% N/A N/A [2628]
with rough and porous surfaces device similar to
Aerolizer

less than 0.5 log. Future in vivo studies are needed to further conrm the Relenza is a formulation strictly for inhalation. One fatal incident
efcacies of these new inhaled anti-microbial therapies. occurred when Relenza was nebulised and given to a mechanically
ventilated adult patient suffering from severe H1N1 inuenza A. The lac-
3. Inhaled antiviral formulations tose in Relenza formulation triggered ventilator occlusion causing the
patient to have severe hypoxemia (SpO2 dropped to 7178%) and bilater-
3.1. Zanamivir al pneumothoraces [190]. The Relenza manufacturer, GlaxoSmithKline,
further claried that this formulation is not to be reconstituted in any
Only zanamivir is approved for inhalation use in US (Relenza, liquid formulation for nebulisation [191]. The aqueous saline solution of
GlaxoSmithKline, Middlesex, UK). In Relenza Rotadisk, each blister zanamivir that has been used for nebulisation does not contain lactose
contains a mixture of 5 mg zanamivir and 20 mg lactose [183]. Patients [192].
are to inhale the contents of two blisters, twice a day for ve days.
Signicant reductions in the median time to relieve the symptoms 3.2. Laninamivir
were reported to be 1.25 days in inuenza positive patients (512
years old) [184]. The Diskhaler is a low resistance device Laninamivir octanoate (LANI) is the newest neuraminidase inhibi-
(0.022 kPa1/2/(l/min)) which generates a ow rate of 90 l/min at a tors currently only approved in Japan (Inavir, Daiichi Sankyo Company
4 kPa pressure drop across the inhaler [185]. An in vitro study using Ltd, Tokyo, Japan and Biota Pharmaceuticals, Alpharetta, USA). It is an
the Next Generation Impactor revealed FPF less than 30% even at a antiviral drug that has long retention in the lungs and is able to remain
high ow rate of 90 l/min with most of powder retentions found in for more than 5 days and hence requires only a single inhalation as
the pre-separator (45.4 5.0%) and the device (20.9 5.2%) [185]. opposed to the twice daily ve days treatment course for zanamivir
Using a breath simulator, generating a peak inspiratory ow of 84 l/ [193]. In an animal study, t1/2 (time to half the concentration of LANI
min and 2.78 L of air and a realistic physical airway model instead of a soon after administration) was 41.4 h and even at 120 h post-dose,
USP throat, a similar lung dose was obtained [186]. Data obtained concentration of LANI in the lung was considerable higher than IC50
from atomic force microscopy has additionally uncovered doubling (50% inhibitory concentration) [194]. The median duration required
increase of adhesion force (240 nN to 582 nN) between spray-dried for fever and u symptoms to disappear was three to four days
lactose (dv, 0.5 = 150 m with geometric standard deviation of 1.8) (including days of administration) in patients having type A and B inu-
and micronized zanamivir (dv, 0.5 = 2.5 m with geometric standard enza, respectively [195].
deviation of 1.5) when the relative humidity was increased from 32% Patients need to inhale the required dose from a single-use disposable
to 85% [187]. This nding indicated that the already low FPF could be TwinCaps inhaler (Hovione Farmaciencia, SA, Portugal). TwinCaps is a
further decreased in high humidity. high resistance device (0.057 kPa1/2/(l/min)) which generates a ow rate
The low lung dose could be one of the reasons why Relenza gained of 35 l/min at a pressure drop of 4 kPa across the device [196]. A low
poor acceptance in the market. Relenza only managed to dominate ow of 20 l/min was claimed to be sufcient to achieve effective delivery
25% of anti-inuenza market in USA when it was rst marketed in [196]. Inavir manufacturer, Daiichi Sankyo, further claimed that the
1999/2000 [188]. In addition, worldwide safety warning involving risk FPF is consistent in the range of 1.2 kPa (corresponding to 19 l/min)
of bronchospasm was then issued which triggered GSK to reduce 4 kPa and therefore suitable for patients with weak inhalation strength
marketing for Relenza and further caused the sales to drop even [197]. However, the FPF values and device retention were not available
more [188]. Another reason for the poor acceptance is the difculty in in the open literature. Post-marketing surveillance conducted by the
using the Diskhaler. A survey, conducted in Okinawa during the company reported that more that 90% patients, ranging from 2 to
2010 u season, found the administration of zanamivir using 94 years old, were able to achieve sufcient inhalation required for
Diskhaler to be the most difcult compared with the administration therapeutic delivery. Children (b 10 years old) are required to inhale a
of oseltamivir (oral tablet) and laninamivir (powder inhaler) [188]. A total dose of 20 mg whilst 40 mg doses are to be inhaled for adults.
randomised controlled trial found that elderly people (N65 years old.), The single administration of LANI is commonly done under the supervi-
where 80% of inuenza related death occurs, cannot use the Diskhaler sion of health care professionals who ensure that the correct inhalation
correctly [189]. Even directly after training, 50% (19 out of 38 elderly technique is adhered by the patients. Laninamivir has proven to be
patients) of the subjects failed to load and prime the device correctly. effective to the H1N1/H275Y virus and shown similar clinical efcacy
This number increased to 65% after 24 h of training. Loading and to zanamivir and oseltamivir against H1N1, H3N2 and inuenza B
priming the device consist of too many steps, starting with the viruses [198,199]. Alleviation time for children was shorter with
squeezing of the side of the tray to remove it from the device, placing laninamivir compared to oseltamivir, whilst in adults the alleviation
the Rotadisk in the tray, inserting the tray back into the device and time is similar [200]. Moreover, animal studies have shown the superior
puncturing the blister. Unless the inhaler is improved, the delivery of efcacy of laninamivir against the highly pathogenic avian u virus
zanamivir to treat inuenza in elderly patients is unlikely to be efcient. H5N1 [201].

Please cite this article as: Q.(T.) Zhou, et al., Inhaled formulations and pulmonary drug delivery systems for respiratory infections, Adv. Drug Deliv.
Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.10.022
10 Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxxxxx

A rotating brush, with a precise speed rate, picks up the particles from the surface of compacted powder and release them at uniform dosing speed into air stream to form aerosol. Varying the reservoir diameter, where powder is loaded, and the
LANI has just completed a randomised, double-blind, placebo-

57 and 59 year old men and


controlled, parallel-arm Phase 2 clinical trial comparing the safety and

18 month old infant [29]

64 year old woman [29]


47 year old female [29]
Model and reference efcacy of a 40 mg and a 80 mg dose to placebo in 639 patients. Interest-
ingly, the study demonstrated a statistically signicant reduction in viral

Rabbit model [32].


Mice model [31].
Mice model [30]
shedding on day 3 for those patients in both the 40 mg and 80 mg
cohorts compared to placebo; however, neither two doses achieved a
statistically signicant reduction in the median time to alleviation of
inuenza symptoms as measured by the Flu-iiQ patient-recorded
outcome questionnaire [202]. The median time to alleviation of inuenza

Corresponding survival rate: 50, 83, 100%


symptoms was 102.3 h for the 40 mg cohort and 103.2 h for the 80 mg
Once daily for 5 days. Nebulisation time:

Once daily for 5 days. Nebulisation time:

cohort, as compared to 104.1 h for the placebo cohort [202]. The reason

Once daily for 3 days. Three different


One capsule daily for 5 days. 10 mg

Once daily for 4 days. Nebulisation


4 min (1st day), 7 min (2nd day),

is unclear that virus shedding did not translate into the alleviation of

doses: 0.5, 1.0, and 1.75 mg/kg.


inuenza symptoms. Therefore, full analysis and understanding of all
the clinical data are essential prior to the further development of inhaled
714 min (3rd5th days)

DAS181 in each capsule

LANI.
Twice a day for 4 days

3.3. Anti-inuenza drugs under development


Dose 1.5 ml/kg
Dosing regime

time: 30 min

It is only a matter of time before a mutated strain(s) emerged


8 min

triggering resistance to the available antiviral drugs that targets the


viral genes which encode the proteins that are susceptible to mutation
deposited = 6.1 mg

deposited = 7.9 mg
Deposited lung dose

[203,204]. Broad spectrum antiviral drugs that do not target these


20.4 mg ribavirin/

viral proteins will then be more valid since it will still be efcacious
kg body weight
5 m) = 68.2%

5 m) = 68.2%
Total DAS181

Total DAS181

despite gene mutations of these viruses. Inhaled anti-inuenza drugs


under development but yet approved are listed in Table 4 including
6570%
FPF (1

FPF (1

DAS181 [205], ribavirin [206], recombinant human catalase [207] and


N/A

N/A

Cidovir [208].
Nebulised (type of nebuliser is not

Rotating brush powder generator

4. Inhaled anti-fungal formulations


Nebulised with Aerotech II (CIS-
Nebulised with Aerogen Pro X

(Palas, Karlsruhe, Germany)a

Immunocompromised patients with malignancy, hematologic


disease, HIV, cancer and organ transplantation are highly susceptible
USA, Bedford, MA)

to invasive pulmonary fungal infections. These infections cause


Delivery method

unacceptably high mortality and morbidity rates (4090%) and become


an alarming healthcare problem [209,210]. A vast majority of the
Aerolizer

specied)

infected cases are caused by Candida spp. and Aspergillus spp. [211].
Oral and/or intravenous administrations of anti-fungal drugs, such as
amphotericin B, ucytosine and a handful of clinically available azole
Stock solution of 10.0 mg DAS181/ml water is

agents, are the mainstay in the treatment of pulmonary fungal


infections [212]. However, these traditional treatments are unavoidably
Dry powder composition is not specied.

associated with different degree of systemic toxicities which often lead


diluted in saline to give 1.3 mg/ml

to early discontinuation of treatments, and hence have poor therapeutic


outcomes. In the last two decades, increasing attention has been drawn
RHC in saline (33,000 U/ml).

to pulmonary delivery of anti-fungal agents for the prevention of inva-


100 mg ribavirin/ml water
1.0 mg DAS181/ml water

piston speed, to change the compaction strength, will give different aerosol sizes.

sive pulmonary fungal infections [213]. Yet, the administration of


Inhaled anti-viral drugs that showed efcacy against different types of virus.

aerosolised antifungal agents is limited to case studies due to the lack


Micronised Cidovir

of sufcient clinical data on their efcacy and safety proles [213,214].


Whilst further well-designed clinical trials are required to establish
Formulation

their therapeutic roles and risks, here we will provide an update on


the recent developments of anti-fungals designed for aerosol delivery
(Table 5).
H3N2 inuenza

H1N1 inuenza

Smallpox virus
parainuenza

4.1. Amphotericin B
Target virus

Human

virus 3

Amphotericin B, which is a membrane-active polyene macrolide


virus

virus

antifungal agent, has been the most commonly used therapy for the
treatment of life-threatening pulmonary fungal infections and the
protein with sialidase activity)

High dose ribavirin to shorten

Recombinant human catalase

most thoroughly studied agent for inhalation administration [215].


Liposomal amphotericin B dry powder inhaler formulation was
DAS181 (a recombinant

developed by Shah and Misra [216]. However, majority of the studies


are intended to demonstrate the feasibility of nebulising commercially
treatment time

available formulations, including Fungizone (a deoxycholate formula-


tion), Ambisome (liposome-based formulation), Abelect and
Amphotec (lipid-based formulations). The corresponding pharmaco-
Cidovir
(RHC)
Table 4

kinetics properties and safety proles have been summarised in Kuiper


API

and Ruijgrok [217]. In brief, the liposomal and lipid-based formulations

Please cite this article as: Q.(T.) Zhou, et al., Inhaled formulations and pulmonary drug delivery systems for respiratory infections, Adv. Drug Deliv.
Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.10.022
Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxxxxx 11

Table 5
Formulations of inhaled anti-fungal drugs.

Drug Formulation Production method Major excipient FPF Device Test Reference
ow
rate (l/
min)

Amphotericin Liposomal Commercially available as Hydrogenated soy 18.5% Atomiser NL9M 15 [33]
B formulation for AmBisome for injection phosphatidylcholine
nebulisation (48%), cholesterol (24%),
distearoylphosphatidylglycerol
(19%)
Suspension for Chitosanstearic acid conjugate Chitosanstearic acid with 17 4052% Air-jet nebuliser 60 [34]
nebulisation nanomicelles by solvent evaporation or 60% substitution degree
(6095% w/w)
Suspension for Non-ionic surfactant vesicles Hydroxypropyl-- 421% Buxco nebulisation 60 [35]
nebulization cyclodextrin: system
amphotericin B = 100:1 w/w,
lipids including
Mono-n-hexadecyl
ether tetraethylene glycol,
cholesterol, and dicetyl
phosphate (30 and 150 mM)
Itraconazole Suspension for Crystalline nanoparticles Mannitol (30%), leucine (12%) 4754% Aeroneb 28.3 [36]
nebulisation by wet-milling and
amorphous nanostructured
aggregated by ultra-rapid freezing
Suspension for Crystalline nanoparticles Polysorbate 80 (14%) N/A Pari LC Plus, Medel N/A [37]
nebulisation by wet-milling Jet Basic, Multisonic
and Pari eFlow
Dry powder Chitosan-based nanoparticle Lactose (2.520% w/w), 1643% Cyclohaler 60 [38]
by ionic gelation, then mannitol (2.520% w/w),
spray drying nanosuspension leucine (010% w/w)
Dry powder Crystalline nanoparticles by TPGS (3.2%), mannitol (64.5%), 4663% Axahaler 100 [39]
high-pressure homogenization, sodium taurocholate
then spray drying nanosuspension (0.30.9%)
Dry powder Spray drying drug solution with excipients Mannitol (6190%), 4767% Axahaler 100 [40]
phospholipid (0.363.47%)
Voriconazole Dry powder by spray Crystalline microparticles in the PVP K12 (33%), PVP K30 543% Handihaler 60 [41]
drying micro-nano- absence of excipients and amorphous (2533%)
suspensions nanostructured aggregated in the presence of
excipients by thin lm freezing

were nebulised better with higher local drug concentration and no stability after nebulisation. Alsaadi et al. [220] formulated amphotericin
negative effect on pulmonary surfactant when compared with the B-hydroxypropyl-g-cyclodextrin (1:100 w/w with 1 mg/ml amphotericin
deoxycholate formulation. It is believed that the detrimental effect on B) complex into non-ionic surfactant vesicles which was composed of
lung function may be related to the deoxycholate component. mono-n-hexadecyl ether tetraethylene glycol, cholesterol, and dicetyl
Therefore, the recent work has been focused on studying the liposomal phosphate in a 3:3:1 molar ratio (30 or 150 mM lipid). The formulations
or other encapsulated formulations. were freeze-dried to extend the shelf-life and then rehydrated with water
Fauvel et al. [218] investigated the in vitro aerosol performance of for nebulisation using a Buxco nebulisation system to treat leishmania-
liposomal amphotericin B delivered by a Atomiser NL9M nebuliser sis. A treatment with 5 doses of aerosolised non-ionic surfactant vesicle
with an AOBOX compressor and conrmed the ability to deliver the formulation (once daily for 5 consecutive days with a total dose of
liposomal formulation to the alveolar compartment. They also tested 12.65 mg drug/kg) exhibited enhanced anti-fungal performance com-
the drug uptake and toxicity of drug at concentrations achievable by pared with treatment using amphotericin B-dextrin solution in a mice
nebulisation on the A549 human lung epithelial cell line. Drug model.
concentrations above the MIC of most Aspergillus species were reported,
but amphotericin B approached levels potentially being cytotoxic to the 4.2. Itraconazole
cells. Their ndings could possibly explain the inconsistency in the
safety prole observed in various clinical studies [217] when different Itraconazole is a highly hydrophobic and weakly basic broad spec-
nebulisation set-ups and/or protocol were used. trum anti-fungal azole available as oral and intravenous formulations
Gilani et al. [219] specically formulated amphotericin B for jet (e.g. Sporanox). There are strong interests to develop inhalable formu-
nebulisation using chitosanstearic acid conjugate nanomicelles as the lations of itraconazole because of its low aqueous solubility (1 ng/ml at
carrier. A solvent evaporation technique was used to incorporate the pH 7), coupled with the requirement of a high therapeutic concentra-
drug into nanomicelles in a size range of 101248 nm with encapsula- tion (0.5 g/g lung tissue or 0.5 g/ml blood), as well as its poor and
tion efciency up to 97%. They found that the micelle size was decreased erratic absorption characteristics [221]. Even so, improving drug disso-
with increasing drug loading, which was attributed to the enhanced lution and/or solubility remains a challenge for pulmonary delivery of
hydrophobic interaction between adjacent drug and stearic acid itraconazole to treat invasive pulmonary aspergillosis. A common
molecules. Whilst comparable antifungal activities were obtained strategy to enhance the dissolution rate of poorly water-soluble drugs
between the micelle formulations and Fungizone, the micelles is to reduce the drug particle size to the nanometer scale, which results
possessed less aggregated amphotericin B which is responsible for in very large specic surface area for dissolution [222,223]. Particle en-
systemic toxicity. The air-jet nebulised nanomicelle formulation gineering techniques, including evaporative precipitation of aqueous
showed a FPF (b6.4 m) of 52% without compromising on drug solution (100500 nm), spray freezing into liquid (2050 nm) and

Please cite this article as: Q.(T.) Zhou, et al., Inhaled formulations and pulmonary drug delivery systems for respiratory infections, Adv. Drug Deliv.
Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.10.022
12 Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxxxxx

ultra-rapid freezing (2050 nm), have been used in the past decade to 4.3. Voriconazole
prepare nanostructured itraconazole dispersions for nebulisation.
Major ndings are summarised in Yang et al. [224]. Briey, nebulised Voriconazole is a broad-spectrum anti-fungal agent and available as
itraconazole nanoparticle formulations were able to deliver the drug oral and intravenous products (e.g. Vfend and Captisol). Though it is
to the alveoli; improved bioavailability and in-vivo prophylactic effects 700 times more soluble than itraconazole, its low aqueous solubility
of inhaled itraconazole formulations were achieved compared with the (0.61 mg/ml at pH 7) remains a challenge in formulation design. The
Sporanox oral formulation [224]. efcacy of inhaled voriconazole therapy was demonstrated in three life-
Whilst particle size reduction signicantly enhances the dissolution threatening invasive aspergillosis cases when systemic voriconazole
rate of itraconazole, the potential impact of polymorphs on the solubil- treatments were withdrawn due to severe adverse effects [214].
ity of drug was also recognised. Yang et al. [225] compared the bioavail- Intravenous formulation was nebulised to introduce 40 mg voriconazole
ability of nano-crystalline and amorphous itraconazole formulations, to patients without attempts to control their respiratory pattern. After a
which were prepared by wet-milling and ultra-rapid freezing 3 month treatment, all patients were reported to return to good
techniques, respectively. Single-dose (20 mg/ml in 5 ml suspension) conditions with minimal or no adverse effects reported during and
was nebulised to SpragueDawley rats using a nose-only dosing 6 months after the therapy.
apparatus. They found that the systemic bioavailability of the amorphous Dry powder-based inhalation formulations of voriconazole were
formulation (peak concentration of 180 ng/ml at 4 h after dosing) was 3.8 developed by Beinborn and co-workers using a thin lm freezing
times higher than that of the crystalline counterpart, despite similar aero- technique [229]. Various process parameters, type/grade of stabilising
dynamic performance and lung deposition of the two formulations. This excipient, drug to excipient ratio, solvents and solid contents were
was attributed to the rapid dissolution of the amorphous nanoparticles examined [229]. Whilst microstructure low density crystalline aggre-
and higher levels of supersaturation which was dened as the ratio of gates (specic surface area = 10 m2/g) were produced in the absence
drug concentration to the equilibrium solubility (4.7 times of the crystal- of excipients, addition of PVP as a stabilising agent produced low density
line formulation). A wet-milled itraconazole nanosuspension containing amorphous nanostructure voriconazole aggregates (specic surface
polysorbate 80 (itraconazole: polysorbate 80 = 86:14, 10% total solid area = 15180 m2/g). When the powders were assessed using a
content) was shown to improve bioavailability in a rat model [221]. A Handihaler at 60 l/min, the highest ne particle fraction was obtained
dose of 22.5 mg/kg was nebulised and a systemic peak concentration of in an excipient-free formulation (42.4%) and a formulation stabilised by
104 ng/ml 4 h after dosing was reached. This formulation could result PVP K12 at a drug to excipient ratio of 1:2 (43.1%). In a follow up study
in longer exposure of the lung tissue to itraconazole (14.3 g/g lung tissue [229], the microstructure crystalline voriconazole gave more favourable
24 h after dosing). Such features are important for a poorly water-soluble pharmacokinetics in lung tissues (AUC024 h = 452.6 g h/g) compared
drug to enhance its efcacy, but drug dosing should be carefully consid- with the amorphous nanostructure aggregates formulation (AUC0
ered to avoid systemic toxicity. 24 h = 232.1 g h/g) after a 10 mg/kg insufated dose. The low retention
For the development of DPI formulations, Jafarinejad et al. [226] of voriconazole in the lung tissue for the amorphous nanostructure ag-
employed a modied ionic gelation approach to encapsulate itraconazole gregates was attributed to its faster dissolution rate, similar to the nd-
into chitosan-based nanoparticle (190240 nm) with an encapsulation ing of adding phospholipid to the amorphous microparticle itraconazole
efciency of up to 55%. The nanosuspension was then spray dried with formulation [228]. In such case, the benets brought by faster dissolu-
and without excipients (lactose, mannitol and leucine) to produce tion were out-weighed by the rapid elimination of drugs from the
inhalable microparticles. The highest FPF obtained was 43% for a formula- lungs as well as the potential risk of getting plasma peak
tion containing 10% mannitol and 10% leucine using a Cyclohaler at concentration related side effects.
60 l/min for 4 s. A nanoparticle-based dry powder formulation was also
proposed by Duret et al. [227]. Itraconazole suspensions were produced 5. Conclusions
by high-pressure homogenization and stabilised by 10% tocopherol
polyethylene 1000 (10% w/w), then co-spray dried with mannitol and/ Inhaled anti-microbial therapy is an emerging and rapidly expanding
or sodium taurocholate. They found that the nanoparticle formulation area. As systemic exposure of many high-dose anti-microbials can cause
has a much higher FPF (46.263.2%) and solubility (5896 ng/ml) severe adverse effects, inhalation therapy provides an attractive solution
compared to the microparticle-based itraconazole formulations that allows the establishment of high localized drug concentrations in the
(23.1% and b10 ng/ml). Amorphous dry powder formulations were respiratory tract with minimal systemic drug exposure. In reality, it is
prepared by spray drying itraconazole solution with mannitol and/ challenging to formulate high-dose anti-microbial agents into inhaled
or hydrogenated soy-lecithin (phospholipid) as excipients. High dosage forms as the dose, dosing frequency and site of action of inhaled
FPF (46.967.0%) was obtained and the addition of phospholipid antibiotics are substantially different from those for asthma and chronic
(10%) resulted in a signicantly higher dissolution rate [227]. Duret obstructive pulmonary disease. Delivery of high-dose drugs via tradition-
et al. [228] compared the bioavailabilities of three itraconazole- al nebulisation may require lengthy treatment durations. High drug doses
containing dry powder formulations: micronized crystalline powder, in a DPI also preclude or limit the use of carriers or other functional
amorphous powders with and without phospholipid. A single dose excipients as these would signicantly increase the bulk volume of total
(0.5 mg/kg) of each of the powders was endotracheally insufated dose and necessitate a bulkier device or a larger number of doses. Techno-
into mice. The amorphous formulations had a higher systemic logical advances are essential to maximise drug delivery efciencies and
bioavailability (plasmatic AUC 024 h of 491.5 ng/ml without convenience of use.
phospholipid and 376.8 ng/ml with phospholipid) compared with Nebulisation is still the mainstay of inhaled therapy for the treat-
the micronized crystalline itraconazole formulation (182 ng/ml). ment of respiratory infections. However, traditional jet nebulisers,
Though the addition of phospholipid increased the wetting and with their long administration times and low delivery efciencies, do
absorption of the itraconazole powders, a faster elimination rate from not meet the stringent requirements of inhaled high-dose antibiotics.
the lung tissue was also observed (elimination half-life of 4.1 h). On These problems have been largely solved by the development of mesh
the other hand, amorphous powders without phospholipid had an nebulisers. Apart from nebulisers, DPIs are also becoming popular and
extended pulmonary retention (elimination half-life of 14.7 h), even have attracted rising interest in recent years for the delivery of inhaled
higher than the crystalline formulation (elimination half-life of 6.5 h). high-dose antibiotics. Their popularity stems from their enhanced sta-
With the improved local bioavailability, careful dosing of the dry bilities, the ability to administer high drug doses within relatively
powder formulations may achieve sufcient anti-fungal activity with shorter durations of time and ease of use. Cough and throat irritation
minimum systemic toxicity. are some common problems caused by the inhalation of high-dose of

Please cite this article as: Q.(T.) Zhou, et al., Inhaled formulations and pulmonary drug delivery systems for respiratory infections, Adv. Drug Deliv.
Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.10.022
Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxxxxx 13

antibiotics in both liquid and powder forms, which can be minimised. [18] D. Cipolla, H.K. Chan, J. Schuster, D. Farina, Personalizing aerosol medicine:
development of delivery systems tailored to the individual, Ther. Deliv. 1 (2010)
With advanced particle engineering techniques and intelligent inhaler 667682.
designs, it is expected that the delivery performance and patient adher- [19] D.E. Geller, M.W. Konstan, S.B. Noonberg, C. Conrad, Novel tobramycin inhalation
ence of antibiotic DPIs would improve further. powder in cystic brosis subjects: pharmacokinetics and safety, Pediatr. Pulmonol.
42 (2007) 307313.
Liposomal, polymeric and nanoparticle formulations of inhaled [20] M.W. Konstan, P.A. Flume, M. Kappler, R. Chiron, M. Higgins, F. Brockhaus, J. Zhang,
antibiotics are advanced drug delivery systems designed for the G. Angyalosi, E. He, and D.E. Geller, Safety, efcacy and convenience of tobramycin
purposes of targeting drug delivery, sustaining drug release and inhalation powder in cystic brosis patients: the EAGER trial, J. Cyst. Fibros. 10
(2011) 5461.
enhancing the anti-microbial activities of antibiotics in the lungs. In [21] M.J. Harrison, M. McCarthy, C. Fleming, C. Hickey, C. Shortt, J.A. Eustace, D.M.
vivo safety and efcacy evaluations particularly clinical studies are Murphy, B.J. Plant, Inhaled versus nebulised tobramycin: a real world comparison
essential to the further development of such advanced formulations. in adult cystic brosis (CF), Journal of Cystic Fibrosis.
[22] H. Blau, H. Mussaf, M.M. Zahav, D. Prais, M. Livne, B.M. Czitron, H.A. Cohen,
Given that no new antibiotics are in the advanced development stage
Microbial contamination of nebulizers in the home treatment of cystic brosis,
against Gram-negative multi-drug resistant bacteria, innovative Child Care Health Dev. 33 (2007) 491495.
formulations of existing antibiotics are of paramount importance to [23] H.A. Cohen, Z. Cohen, A.S. Pomeranz, B. Czitron, E. Kahan, Bacterial contamination
win the long-term war against the respiratory super bugs. of spacer devices used by asthmatic children, J. Asthma 42 (2005) 169172.
[24] T.W. De Vries, S.R. Rienstra, E.R. Van Der Vorm, Bacterial contamination of
inhalation chambers: results of a pilot study, J. Aerosol Med. 17 (2004) 354356.
[25] Q. Zhou, D. Morton, Druglactose binding aspects in adhesive mixtures: controlling
Acknowledgement performance in dry powder inhaler formulations by altering lactose carrier
surfaces, Adv. Drug Deliv. Rev. 64 (2012) 275284.
The authors acknowledge the nancial support from the Australian [26] J.G. Weers, J. Bell, H.K. Chan, D. Cipolla, C. Dunbar, A.J. Hickey, I.J. Smith, Pulmonary
formulations: what remains to be done? J. Aerosol Med. Pulm. Drug Deliv. 23
Research Councils Discovery Projects funding scheme (DP120102778) (Suppl. 2) (2010) S5S23.
and the National Health and Medical Research Council's (NHMRC) [27] T. Sou, L.M. Kaminskas, T.H. Nguyen, R. Carlberg, M.P. McIntosh, D.A.V. Morton, The ef-
Project Grant funding scheme (APP1065046). Qi Tony Zhou is an fect of amino acid excipients on morphology and solid-state properties of multi-
component spray-dried formulations for pulmonary delivery of biomacromolecules,
NHMRC Early Career Fellow (APP1053528). Thaigarajan Parumasivam
Eur. J. Pharm. Biopharm. 83 (2013) 234243.
is a recipient of Malaysian Government scholarship. [28] T. Sou, L. Orlando, M.P. McIntosh, L.M. Kaminskas, D.A.V. Morton, Investigating the
interactions of amino acid components on a mannitol-based spray-dried powder
formulation for pulmonary delivery: a design of experiment approach, Int. J.
References Pharm. 421 (2011) 220229.
[29] S. Hoe, J.W. Ivey, M.A. Boraey, A. Shamsaddini-Shahrbabak, E. Javaheri, S.
[1] S.A. Antoniu, I. Cojocaru, Inhaled colistin for lower respiratory tract infections, Matinkhoo, W.H. Finlay, R. Vehring, Use of a fundamental approach to spray-
Expert Opin. Drug Deliv. 9 (2012) 333342. drying formulation design to facilitate the development of multi-component dry
[2] WHO, The Global Burden of Disease2004. powder aerosols for respiratory drug delivery, Pharm. Res. 31 (2014) 449465.
[3] D. Traini, P.M. Young, Delivery of antibiotics to the respiratory tract: an update, [30] M. Paajanen, J. Katainen, J. Raula, E.I. Kauppinen, J. Lahtinen, Direct evidence on
Expert Opin. Drug Deliv. 6 (2009) 897905. reduced adhesion of Salbutamol sulphate particles due to L-leucine coating,
[4] S.W.S. Yapa, J. Li, C.J.H. Porter, R.L. Nation, K. Patel, M.P. McIntosh, Population Powder Technol. 192 (2009) 611.
pharmacokinetics of colistin methanesulfonate in rats: achieving sustained lung [31] J. Raula, F. Thielmann, M. Naderi, V.P. Lehto, E.I. Kauppinen, Investigations on par-
concentrations of colistin for targeting respiratory infections, Antimicrob. Agents ticle surface characteristics vs. dispersion behaviour of L-leucine coated carrier-free
Chemother. 57 (2013) 50875095. inhalable powders, Int. J. Pharm. 385 (2010) 7985.
[5] S.M. Garonzik, J. Li, V. Thamlikitkul, D.L. Paterson, S. Shoham, J. Jacob, F.P. Silveira, A. [32] D.E. Geller, J. Weers, S. Heuerding, Development of an inhaled dry-powder formu-
Forrest, R.L. Nation, Population pharmacokinetics of colistin methanesulfonate and lation of tobramycin using PulmoSphere (TM) technology, J. Aerosol Med. Pulm.
formed colistin in critically ill patients from a multicenter study provide dosing Drug Deliv. 24 (2011) 175182.
suggestions for various categories of patients, Antimicrob. Agents Chemother. 55 [33] H. Stass, J. Nagelschmitz, S. Willmann, H. Delesen, A. Gupta, S. Baumann, Inhalation
(2011) 32843294. of a dry powder ciprooxacin formulation in healthy subjects: a Phase I study, Clin.
[6] A. Schuster, C. Haliburn, G. Dring, M.H. Goldman, Freedom Study Group, Safety, Drug Invest. 33 (2013) 419427.
efcacy and convenience of colistimethate sodium dry powder for inhalation [34] J. Weers, T. Tarara, The PulmoSphere platform for pulmonary drug delivery, Ther.
(Colobreathe DPI) in patients with cystic brosis: a randomised study, Thorax 68 Deliv. 5 (2014) 277295.
(2013) 344350. [35] A. Haynes, J. Nakamura, C. Heng, S. Heuerding, G. Thompson, R. Malcolmson,
[7] S.W.S. Yapa, J. Li, K. Patel, J.W. Wilson, M.J. Dooley, J. George, D. Clark, S. Poole, E. Aerosol performance of tobramycin inhalation powder, Respir. Drug Deliv., 2010,
Williams, C.J.H. Porter, R.L. Nation, M.P. McIntosh, Pulmonary and systemic pp. 701706.
pharmacokinetics of inhaled and intravenous colistin methanesulfonate in cystic [36] H.A. Tiddens, D.E. Geller, P. Challoner, R.J. Speirs, K.C. Kesser, S.E. Overbeek, D.
brosis patients: targeting advantage of inhalational administration, Antimicrob. Humble, S.B. Shrewsbury, T.A. Standaert, Effect of dry powder inhaler resistance
Agents Chemother. 58 (2014) 25702579. on the inspiratory ow rates and volumes of cystic brosis patients of six years
[8] Q. Zhou, P. Tang, S.S.Y. Leung, J.G.Y. Chan, H.-K. Chan, Emerging inhalation aerosol and older, J. Aerosol Med. 19 (2006) 456465.
devices and strategies: where are we headed? Adv. Drug Deliv. Rev. 75 (2014) [37] J. Fiegel, L. Garcia-Contreras, M. Thomas, J. VerBerkmoes, K. Elbert, A. Hickey, D.
317. Edwards, Preparation and in vivo evaluation of a dry powder for inhalation of
[9] T. Velkov, A.R. Nusaibah, Q. Zhou, H.-K. Chan, J. Li, Inhaled antimicrobial capreomycin, Pharm. Res. 25 (2008) 805811.
chemotherapy for respiratory tract infections: successes, challenges and the road [38] A.S. Dharmadhikari, M. Kabadi, B. Gerety, A.J. Hickey, P.B. Fourie, E. Nardell, Phase I,
ahead, Adv. Drug Deliv. Rev. (2014). single-dose, dose-escalating study of inhaled dry powder capreomycin: a new ap-
[10] D. Cipolla, H.-K. Chan, Inhaled antibiotics to treat lung infection, Pharm. Patent proach to therapy of drug-resistant tuberculosis, Antimicrob. Agents Chemother.
Anal. 2 (2013) 647663. 57 (2013) 26132619.
[11] A.M. Jones, J.M. Helm, Emerging treatments in cystic brosis, Drugs 69 (2009) [39] N. Tsapis, D. Bennett, K. ODriscoll, K. Shea, M.M. Lipp, K. Fu, R.W. Clarke, D.
19031910. Deaver, D. Yamins, J. Wright, C.A. Peloquin, D.A. Weitz, D.A. Edwards, Direct
[12] N. Hoiby, Recent advances in the treatment of Pseudomonas aeruginosa infections lung delivery of para-aminosalicylic acid by aerosol particles, Tuberculosis
in cystic brosis, BMC Med. 9 (2011). 83 (2003) 379385.
[13] P.C.L. Kwok, H.-K. Chan, Delivery of inhalation drugs to children for asthma and [40] S. Gad, L. Tajber, O.I. Corrigan, A.M. Healy, Preparation and characterisation of novel
other respiratory diseases, Adv. Drug Deliv. Rev. 73 (2014) 8388. spray-dried nano-structured para-aminosalicylic acid particulates for pulmonary
[14] G. Reychler, A. Keyeux, C. Cremers, C. Veriter, D.O. Rodenstein, G. Liistro, Comparison delivery: impact of ammonium carbonate on morphology, chemical composition
of lung deposition in two types of nebulization intrapulmonary percussive ventila- and solid state, J. Pharm. Pharmacol. 64 (2012) 12641274.
tion vs jet nebulization, Chest 125 (2004) 502508. [41] Y.-J. Son, J.T. McConville, A new respirable form of rifampicin, Eur. J. Pharm.
[15] A.S. Qi, J.R. Friend, L.Y. Yeo, D.A.V. Morton, M.P. McIntosh, L. Spiccia, Miniature in- Biopharm. 78 (2011) 366376.
halation therapy platform using surface acoustic wave microuidic atomization, [42] J.G.Y. Chan, C.C. Duke, H.X. Ong, J.C.Y. Chan, A.S. Tyne, H.K. Chan, W.J. Britton, P.M.
Lab Chip 9 (2009) 21842193. Young, D. Traini, A novel inhalable form of rifapentine, J. Pharm. Sci. 103 (2014)
[16] A.E. Rajapaksa, J.J. Ho, A. Qi, R. Bischof, T.H. Nguyen, M. Tate, D. Piedrata, M.P. 14111421.
McIntosh, L.Y. Yeo, E. Meeusen, R.L. Coppel, J.R. Friend, Effective pulmonary [43] Q.T. Zhou, L. Qu, I. Larson, P.J. Stewart, D.A.V. Morton, Improving aerosolization of
delivery of an aerosolized plasmid DNA vaccine via surface acoustic wave nebuli- drug powders by reducing powder intrinsic cohesion via a mechanical dry coating
zation, Respir. Res. 15 (2014) 60. approach, Int. J. Pharm. 394 (2010) 5059.
[17] K. Nikander, I. Prince, S. Coughlin, S. Warren, G. Taylor, Mode of breathing-tidal or [44] Q. Zhou, L. Qu, T. Gengenbach, I. Larson, P.J. Stewart, D.A.V. Morton, Effect of surface
slow and deep-through the I-neb adaptive aerosol delivery (AAD) system affects coating with magnesium stearate via mechanical dry powder coating approach on
lung deposition of (99 m) Tc-DTPA, J. Aerosol Med. Pulm. Drug Deliv. 23 (Suppl. 1) the aerosol performance of micronized drug powders from dry powder inhalers,
(2010) S37S43. AAPS Pharmscitech 14 (2013) 3844.

Please cite this article as: Q.(T.) Zhou, et al., Inhaled formulations and pulmonary drug delivery systems for respiratory infections, Adv. Drug Deliv.
Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.10.022
14 Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxxxxx

[45] Q. Zhou, J.A. Denman, T. Gengenbach, S. Das, L. Qu, H.L. Zhang, I. Larson, P.J. Stewart, [72] A.M.A. Elhissi, J. Giebultowicz, A.A. Stec, P. Wroczynski, W. Ahmed, M.A. Alhnan, D.
D.A.V. Morton, Characterization of the surface properties of a model pharmaceutical Phoenix, K.M.G. Taylor, Nebulization of ultradeformable liposomes: the inuence
ne powder modied with a pharmaceutical lubricant to improve ow via a of aerosolization mechanism and formulation excipients, Int. J. Pharm. 436
mechanical dry coating approach, J. Pharm. Sci. 100 (2011) 34213430. (2012) 519526.
[46] Q.T. Zhou, B. Armstrong, I. Larson, P.J. Stewart, D. Morton, Understanding the [73] T.R. Desai, R.E.W. Hancock, W.H. Finlay, A facile method of delivery of liposomes by
inuence of powder owability, uidization and de-agglomeration characteristics nebulization, J. Control. Release 84 (2002) 6978.
on the aerosolization of pharmaceutical model powders, Eur. J. Pharm. Sci. 40 [74] T.R. Desai, J.P. Wong, R.E. Hancock, W.H. Finlay, A novel approach to the pulmonary
(2010) 412421. delivery of liposomes in dry powder form to eliminate the deleterious effects of
[47] C. Parlati, P. Colombo, F. Buttini, P.M. Young, H. Adi, A.J. Ammit, D. Traini, milling, J. Pharm. Sci. 91 (2002) 482491.
Pulmonary spray dried powders of tobramycin containing sodium stearate to [75] T.R. Desai, R.E.W. Hancock, W.H. Finlay, Delivery of liposomes in dry powder form:
improve aerosolization efciency, Pharm. Res. 26 (2009) 10841092. aerodynamic dispersion properties, Eur. J. Pharm. Sci. 20 (2003) 459467.
[48] S. Belotti, A. Rossi, P. Colombo, R. Bettini, D. Rekkas, S. Politis, G. Colombo, A.G. [76] L.G. Sweeney, Z. Wang, R. Loebenberg, J.P. Wong, C.F. Lange, W.H. Finlay, Spray-
Balducci, F. Buttini, Spray dried amikacin powder for inhalation in cystic brosis freeze-dried liposomal ciprooxacin powder for inhaled aerosol drug delivery,
patients: a quality by design approach for product construction, Int. J. Pharm. Int. J. Pharm. 305 (2005) 180185.
471 (2014) 507515. [77] M. Alhariri, A. Azghani, A. Omri, Liposomal antibiotics for the treatment of
[49] J. Weers, Inhaled antimicrobial therapy barriers to effective treatment, Adv. Drug infectious diseases, Expert Opin. Drug Deliv. 10 (2013) 15151532.
Deliv. Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.08.013 (In press). [78] J. Gubernator, Z. Druis-Kawa, A. Dorotkiewicz-Jach, W. Doroszkiewicz, A. Kozubek,
[50] A. Columbano, G. Buckton, P. Wikeley, A study of the crystallisation of amorphous In vitro antimicrobial activity of liposomes containing ciprooxacin, meropenem
salbutamol sulphate using water vapour sorption and near infrared spectroscopy, and gentamicin against gram-negative clinical bacterial strains, Lett. Drug Des.
Int. J. Pharm. 237 (2002) 171178. Discov. 4 (2007) 297304.
[51] L.C. Grisedale, M.J. Jamieson, P.S. Belton, S.A. Barker, D.Q.M. Craig, Characterization [79] Z. Drulis-Kawa, A. Dorotkiewicz-Jach, J. Gubernator, G. Gula, T. Bocer, W.
and quantication of amorphous material in milled and spray-dried salbutamol Doroszkiewicz, The interaction between Pseudomonas aeruginosa cells and cationic
sulfate: a comparison of thermal, spectroscopic, and water vapor sorption PC:Chol:DOTAP liposomal vesicles versus outer-membrane structure and
approaches, J. Pharm. Sci. 100 (2011) 31143129. envelope properties of bacterial cell, Int. J. Pharm. 367 (2009) 211219.
[52] S. Gaisford, Isothermal microcalorimetry for quantifying amorphous content in [80] T. Desai, G. Tyrrell, T. Ng, W. Finlay, In vitro evaluation of nebulization properties,
processed pharmaceuticals, Adv. Drug Deliv. Rev. 64 (2012) 431439. antimicrobial activity, and regional airway surface liquid concentration of
[53] H. Adi, P.M. Young, H.-K. Chan, H. Agus, D. Traini, Co-spray-dried mannitolcipro- liposomal polymyxin B sulfate, Pharm. Res. 20 (2003) 442447.
oxacin dry powder inhaler formulation for cystic brosis and chronic obstructive [81] T. Velkov, K.D. Roberts, R.L. Nation, P.E. Thompson, J. Li, Pharmacology of
pulmonary disease, Eur. J. Pharm. Sci. 40 (2010) 239247. polymyxins: new insights into an old class of antibiotics, Future Microbiol 8
[54] Q. Zhou, D. Morton, H.H. Yu, J. Jacob, J.P. Wang, J. Li, H.K. Chan, Colistin powders (2013) 711724.
with high aerosolisation efciency for respiratory infection: preparation and [82] S.J. Wallace, J. Li, R.L. Nation, R.J. Prankerd, B.J. Boyd, Interaction of colistin and
in vitro evaluation, J. Pharm. Sci. 102 (2013) 37363747. colistin methanesulfonate with liposomes: colloidal aspects and implications for
[55] D.A.V. Morton, T. Jong, J. Li, J.D. Hale, R.L. Nation, I. Larson, Investigating the particle formulation, J. Pharm. Sci. 101 (2012) 33473359.
engineering of colistin for pulmonary delivery, Respiratory Drug Delivery Europe [83] D. Wang, L. Kong, J. Wang, X. He, X. Li, Y. Xiao, Polymyxin E sulfate-loaded liposome
2011, 2011, pp. 367372. for intravenous use: preparation, lyophilization, and toxicity assessment in vivo,
[56] Q. Zhou, T. Gengenbach, J. Denman, H. Yu, J. Li, H. Chan, Synergistic antibiotic com- PDA J. Pharm. Sci. Technol. 63 (2009) 159167.
bination powders of colistin and rifampicin provide high aerosolization efciency [84] S.J. Wallace, R.L. Nation, J. Li, B. Boyd, Physicochemical aspects of the coformulation
and moisture protection, AAPS J. 16 (2014) 3747. of colistin and azithromycin using liposomes for combination antibiotic therapies,
[57] S.P. Vyas, K. Khatri, Liposome-based drug delivery to alveolar macrophages, Expert J. Pharm. Sci. 102 (2013) 15781587.
Opin. Drug Deliv. 4 (2007) 9599. [85] A.M.A. Elhissi, K.M.G. Taylor, Delivery of liposomes generated from proliposomes
[58] C. Kelly, C. Jefferies, S.-A. Cryan, Targeted liposomal drug delivery to monocytes using air-jet, ultrasonic, and vibrating-mesh nebulisers, J. Drug Deliv. Sci. Technol.
and macrophages, J. Drug Deliv. 727241 (2011). 15 (2005) 261265.
[59] J. Blanchard, L. Danelishvili, I. Gonda, L. Bermudez, Liposomal ciprooxacin [86] K.-H. Song, S.-J. Chung, C.-K. Shim, Preparation and evaluation of proliposomes
preparation is active against Mycobacterium avium subsp hominissuis and containing salmon calcitonin, J. Control. Release 84 (2002) 2737.
Mycobacterium abscessus in macrophages and in biolm, 2014 American Thoracic [87] W. Rojanarat, N. Changsan, E. Tawithong, S. Pinsuwan, H.K. Chan, T. Srichana,
Society Conference, A6677, San Diego, 2014. Isoniazid proliposome powders for inhalationpreparation, characterization and
[60] S. Chono, K. Kaneko, E. Yamamoto, K. Togami, K. Morimoto, Effect of surface- cell culture studies, Int. J. Mol. Sci. 12 (2011) 44144434.
mannose modication on aerosolized liposomal delivery to alveolar macrophages, [88] A. Patil-Gadhe, V. Pokharkar, Single step spray drying method to develop
Drug Dev. Ind. Pharm. 36 (2010) 102107. proliposomes for inhalation: a systematic study based on quality by design
[61] S. Chono, T. Tanino, T. Seki, K. Morimoto, Efcient drug targeting to rat alveolar approach, Pulm. Pharmacol. Ther. 27 (2014) 197207.
macrophages by pulmonary administration of ciprooxacin incorporated into [89] T. Riley, D. Christopher, J. Arp, A. Casazza, A. Colombani, A. Cooper, M. Dey, J. Maas, J.
mannosylated liposomes for treatment of respiratory intracellular parasitic Mitchell, M. Reiners, N. Sigari, T. Tougas, S. Lyapustina, Challenges with developing
infections, J. Control. Release 127 (2008) 5058. in vitro dissolution tests for orally inhaled products (OIPs), AAPS Pharmscitech 13
[62] D. Cipolla, B. Shekunov, J. Blanchard, A. Hickey, Lipid-based carriers for pulmonary (2012) 978989.
products: preclinical development and case studies in humans, Adv. Drug Deliv. [90] D. Cipolla, H. Wu, S. Eastman, T. Redelmeier, I. Gonda, H.-K. Chan, Development
Rev. 75 (2014) 5380. and characterization of an in vitro release assay for liposomal ciprooxacin for
[63] J.P. Clancy, L. Dupont, M.W. Konstan, J. Billings, S. Fustik, C.H. Goss, J. Lymp, P. Minic, inhalation, J. Pharm. Sci. 103 (2014) 314327.
A.L. Quittner, R.C. Rubenstein, K.R. Young, L. Saiman, J.L. Burns, J.R.W. Govan, B. [91] H.X. Ong, D. Traini, D. Cipolla, I. Gonda, M. Bebawy, H. Agus, P.M. Young, Liposomal
Ramsey, R. Gupta, G. Arikace Study, Phase II studies of nebulised Arikace in CF nanoparticles control the uptake of ciprooxacin across respiratory epithelia,
patients with Pseudomonas aeruginosa infection, Thorax 68 (2013) 818825. Pharm. Res. 29 (2012) 33353346.
[64] Z.L. Li, Y.L. Zhang, W. Wurtz, J.K. Lee, V.S. Malinin, S. Durwas-Krishnan, P. Meers, [92] D. Cipolla, I. Gonda, H.K. Chan, Liposomal formulations for inhalation, Ther. Deliv. 4
W.R. Perkins, Characterization of nebulized liposomal amikacin (Arikace (TM)) (2013) 10471072.
as a function of droplet size, J. Aerosol Med. Pulm. Drug Deliv. 21 (2008) 245253. [93] R.O. Salama, D. Traini, H.K. Chan, A. Sung, A.J. Ammit, P.M. Young, Preparation and
[65] R. Bhavane, E. Karathanasis, A.V. Annapragada, Triggered release of ciprooxacin evaluation of controlled release microparticles for respiratory protein therapy, J.
from nanostructured agglomerated vesicles, Int. J. Nanomedicine 2 (2007) Pharm. Sci. 98 (2009) 27092717.
407418. [94] R.A. Roberts, T. Shen, I.C. Allen, W. Hasan, J.M. DeSimone, J.P.Y. Ting, Analysis of the
[66] D. Cipolla, H.Y. Wu, I. Gonda, S. Eastman, T. Redelmeier, H.K. Chan, Modifying the murine immune response to pulmonary delivery of precisely fabricated nano- and
release properties of liposomes toward personalized medicine, J. Pharm. Sci. 103 microscale particles, PLoS One 8 (2013) e62115.
(2014) 18511862. [95] L.A. Dailey, N. Jekel, L. Fink, T. Gessler, T. Schmehl, M. Wittmar, T. Kissel, W.
[67] D.J. Serisier, D. Bilton, A. De Soyza, P.J. Thompson, J. Kolbe, H.W. Greville, D. Cipolla, Seeger, Investigation of the proinammatory potential of biodegradable
P. Bruinenberg, I. Gonda, Inhaled, dual release liposomal ciprooxacin in non-cystic nanoparticle drug delivery systems in the lung, Toxicol. Appl. Pharmacol. 215
brosis bronchiectasis (ORBIT-2): a randomised, double-blind, placebo-controlled (2006) 100108.
trial, Thorax 68 (2013) 812817. [96] J.F. Tomashefski Jr., A.M. Cohen, C.F. Doershuk, Long term histopathologic
[68] P. Bruinenberg, J. Blanchard, D. Cipolla, F. Dayton, S. Mudumba, I. Gonda, Inhaled follow-up of bronchial arteries after therapeutic embolization with polyvinyl
liposomal ciprooxacin: once a day management of respiratory infections, Respira- alcohol (Ivalon) in patients with cystic brosis, Hum. Pathol. 19 (1988) 555561.
tory, Drug Delivery, 2010, pp. 7382. [97] H. Adi, P.M. Young, H.K. Chan, R. Salama, D. Traini, Controlled release antibiotics for
[69] D. Cipolla, F. Dayton, S. Fulzele, E. Gabatan, S. Mudumba, H. Wu, D. Yim, R. dry powder lung delivery, Drug Dev. Ind. Pharm. 36 (2010) 119126.
Zwolinski, Inhaled liposomal ciprooxacin: in vitro properties and aerosol perfor- [98] W.S. Cheow, M.W. Chang, K. Hadinoto, Antibacterial efcacy of inhalable
mance, Respiratory Drug Delivery Orlando, FL, 2010. levooxacin-loaded polymeric nanoparticles against E. coli biolm cells: the effect
[70] D. Cipolla, H. Wu, J. Chan, H.-K. Chan, I. Gonda, Liposomal ciprooxacin for of antibiotic release prole, Pharm. Res. 27 (2010) 15971609.
inhalation retains integrity following nebulization, Respiratory Drug Delivery [99] W.S. Cheow, M.W. Chang, K. Hadinoto, The roles of lipid in anti-biolm efcacy of
Europe 2013, Berlin, Germany, 2013. lipid-polymer hybrid nanoparticles encapsulating antibiotics, Colloids Surf. A
[71] D. Cipolla, H. Wu, I. Gonda, H.-K. Chan, Aerosol performance and long-term Physicochem. Eng. Asp. 389 (2011) 158165.
stability of surfactant-associated liposomal ciprooxacin formulations with [100] F. Ungaro, I. d'Angelo, C. Coletta, R.D.D. Bianca, R. Sorrentino, B. Perfetto, M.A.
modied encapsulation and release properties, AAPS Pharmscitech (2014) Tufano, A. Miro, M.I. La Rotonda, F. Quaglia, Dry powders based on PLGA
110. nanoparticles for pulmonary delivery of antibiotics: modulation of encapsulation

Please cite this article as: Q.(T.) Zhou, et al., Inhaled formulations and pulmonary drug delivery systems for respiratory infections, Adv. Drug Deliv.
Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.10.022
Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxxxxx 15

efciency, release rate and lung deposition pattern by hydrophilic polymers, J. [128] J. Woodcock, J.P. Grifn, R.E. Behrman, Development of novel combination
Control. Release 157 (2012) 149159. therapies, N. Engl. J. Med. 364 (2011) 985987.
[101] Y.-J. Son, J.T. McConville, Preparation of sustained release rifampicin microparticles [129] H. Adi, P.M. Young, H.-K. Chan, P. Stewart, H. Agus, D. Traini, Cospray dried
for inhalation, J. Pharm. Pharmacol. 64 (2012) 12911302. antibiotics for dry powder lung delivery, J. Pharm. Sci. 97 (2008) 33563366.
[102] T.V.P. Doan, J.C. Olivier, Preparation of rifampicin-loaded PLGA microspheres for [130] P. Yeh, A.I. Tschumi, R. Kishony, Functional classication of drugs by properties of
lung delivery as aerosol by premix membrane homogenization, Int. J. Pharm. 382 their pairwise interactions, Nat. Genet. 38 (2006) 489494.
(2009) 6166. [131] D.L. MacLeod, L.M. Barker, J.L. Sutherland, S.C. Moss, J.L. Gurgel, T.F. Kenney, J.L.
[103] M.L. Manca, G. Loy, M. Zaru, A.M. Fadda, S.G. Antimisiaris, Release of rifampicin Burns, W.R. Baker, Antibacterial activities of a fosfomycin/tobramycin combina-
from chitosan, PLGA and chitosan-coated PLGA microparticles, Colloids Surf. B: tion: a novel inhaled antibiotic for bronchiectasis, J. Antimicrob. Chemother. 64
Biointerfaces 67 (2008) 166170. (2009) 829836.
[104] T.V.P. Doan, W. Couet, J.C. Olivier, Formulation and in vitro characterization of [132] D.L. MacLeod, J. Velayudhan, T.F. Kenney, J.H. Therrien, J.L. Sutherland, L.M. Barker,
inhalable rifampicin-loaded PLGA microspheres for sustained lung delivery, Int. J. W.R. Baker, Fosfomycin enhances the active transport of tobramycin in Pseudomo-
Pharm. 414 (2011) 112117. nas aeruginosa, Antimicrob. Agents Chemother. 56 (2012) 15291538.
[105] R. Diab, J. Brillault, A. Bardy, A.V.L. Gontijo, J.C. Olivier, Formulation and in vitro [133] M. Tre-Hardy, H. Traore, N. El Manssouri, F. Vanderbist, M. Vaneechoutte, M.J.
characterization of inhalable polyvinyl alcohol-free rifampicin-loaded PLGA Devleeschouwer, Evaluation of long-term co-administration of tobramycin and
microspheres prepared with sucrose palmitate as stabilizer: efciency for ex vivo clarithromycin in a mature biolm model of cystic brosis clinical isolates of Pseu-
alveolar macrophage targeting, Int. J. Pharm. 436 (2012) 833839. domonas aeruginosa, Int. J. Antimicrob. Agents 34 (2009) 370374.
[106] A. Budhian, S.J. Siegel, K.I. Winey, Haloperidol-loaded PLGA nanoparticles: [134] M. Tre-Hardy, C. Nagant, N. El Manssouri, F. Vanderbist, H. Traore, M.
systematic study of particle size and drug content, Int. J. Pharm. 336 (2007) Vaneechoutte, J.-P. Dehaye, Efcacy of the combination of tobramycin and a
367375. macrolide in an in vitro Pseudomonas aeruginosa mature biolm model,
[107] F. Ito, H. Fujimori, H. Honnami, H. Kawakami, K. Kanamura, K. Makino, Effect of Antimicrob. Agents Chemother. 54 (2010) 44094415.
polyethylene glycol on preparation of rifampicin-loaded PLGA microspheres with [135] G. Herrmann, L. Yang, H. Wu, Z. Song, H. Wang, N. Hoiby, M. Ulrich, S. Molin, J.
membrane emulsication technique, Colloids Surf. B: Biointerfaces 66 (2008) Riethmueller, G. Doering, Colistintobramycin combinations are superior to mono-
6570. therapy concerning the killing of biolm Pseudomonas aeruginosa, J. Infect. Dis. 202
[108] C. Hu, H. Feng, C. Zhu, Preparation and characterization of rifampicin-PLGA (2010) 15851592.
microspheres/sodium alginate in situ gel combination delivery system, Colloids [136] S.A. McColley, B. Trapnell, D. Kissner, M. McKevitt, B. Montgomery, and J. Rosen,
Surf. B: Biointerfaces 95 (2012) 162169. Fosfomycin/tobramycin for inhalation (FTI): microbiological results of a phase 2
[109] N. Yang, Study on Rifapentine-sodium Alginate Microspheres for Lung Targeting placebo-controlled trial in patients with cystic brosis and Pseudomonas
Drug Delivery(PhD thesis) Beijing Tuberculosis and Thoracic Tumor Research aeruginosa, Pediatr. Pulmonol. (2010) 338-338.
Institute, Beijing, China, 2011. [137] S.H. Lee, J. Teo, D. Heng, W.K. Ng, H.-K. Chan, R.B.H. Tan, Synergistic combination
[110] P.S. Pourshahab, K. Gilani, E. Moazeni, H. Eslahi, M.R. Fazeli, H. Jamalifar, dry powders for inhaled antimicrobial therapy: formulation, characterization and
Preparation and characterization of spray dried inhalable powders containing in vitro evaluation, Eur. J. Pharm. Biopharm. 83 (2013) 275284.
chitosan nanoparticles for pulmonary delivery of isoniazid, J. Microencapsul. 28 [138] G.R. Cannefax, A.W. Hanson, Ineffectiveness of lysozyme therapy in experimental
(2011) 605613. syphilis in the rabbit, Br. J. Venereal Dis. 39 (1963) 192194.
[111] J. Zhang, L. Wu, H.-K. Chan, W. Watanabe, Formation, characterization, and fate of [139] M.D. Tsifansky, Y. Yeo, O.V. Evgenov, E. Bellas, J. Benjamin, D.S. Kohane, Micropar-
inhaled drug nanoparticles, Adv. Drug Deliv. Rev. 63 (2011) 441455. ticles for inhalational delivery of antipseudomonal antibiotics, AAPS J. 10 (2008)
[112] Y.J. Wang, K. Kho, W.S. Cheow, K. Hadinoto, A comparison between spray drying 254260.
and spray freeze drying for dry powder inhaler formulation of drug-loaded lipid- [140] G. Pilcer, V. De Bueger, K. Traina, H. Traore, T. Sebti, F. Vanderbist, K. Amighi,
polymer hybrid nanoparticles, Int. J. Pharm. 424 (2012) 98106. Carrier-free combination for dry powder inhalation of antibiotics in the treatment
[113] K. Yamasaki, P.C.L. Kwok, K. Fukushige, R.K. Prud'homme, H.K. Chan, Enhanced dis- of lung infections in cystic brosis, Int. J. Pharm. 451 (2013) 112120.
solution of inhalable cyclosporine nano-matrix particles with mannitol as matrix [141] D.V. Gowda, V. Datta, S. Meenakshi, V.K. Gupta, Formulation and evaluation of dry
former, Int. J. Pharm. 420 (2011) 3442. powders containing anti tuberculosis drugs for pulmonary delivery, Indo Am. J.
[114] N. Tsapis, D. Bennett, B. Jackson, D.A. Weitz, D.A. Edwards, Trojan particles: large Pharm. Res. 3 (2013) 12391248.
porous carriers of nanoparticles for drug delivery, Proc. Natl. Acad. Sci. 99 (2002) [142] P. Muttil, J. Kaur, K. Kumar, A.B. Yadav, R. Sharma, A. Misra, Inhalable microparticles
1200112005. containing large payload of anti-tuberculosis drugs, Eur. J. Pharm. Sci. 32 (2007)
[115] W.S. Cheow, M.L.L. Ng, K. Kho, K. Hadinoto, Spray-freeze-drying production of 140150.
thermally sensitive polymeric nanoparticle aggregates for inhaled drug delivery: [143] L.L.I.J. Booysen, L. Kalombo, E. Brooks, R. Hansen, J. Gilliland, V. Gruppo, P.
effect of freeze-drying adjuvants, Int. J. Pharm. 404 (2011) 289300. Lungenhofer, B. Semete-Makokotlela, H.S. Swai, A.F. Kotze, A. Lenaerts, L.H. du
[116] S.M. D'Addio, J.G.Y. Chan, P.C.L. Kwok, B.R. Benson, R.K. Prud'homme, H.-K. Chan, Plessis, In vivo/in vitro pharmacokinetic and pharmacodynamic study of spray-
Aerosol delivery of nanoparticles in uniform mannitol carriers formulated by dried poly-(dl-lactic-co-glycolic) acid nanoparticles encapsulating rifampicin and
ultrasonic spray freeze drying, Pharm. Res. 30 (2013) 28912901. isoniazid, Int. J. Pharm. 444 (2013) 1017.
[117] S.M. D'Addio, J.G.Y. Chan, P.C.L. Kwok, R.K. Prud'homme, H.-K. Chan, Constant size, [144] J.G.Y. Chan, H.-K. Chan, C.A. Prestidge, J.A. Denman, P.M. Young, D. Traini, A novel
variable density aerosol particles by ultrasonic spray freeze drying, Int. J. Pharm. dry powder inhalable formulation incorporating three rst-line anti-tubercular
427 (2012) 185191. antibiotics, Eur. J. Pharm. Biopharm. 83 (2013) 285292.
[118] J. Varshosaz, S. Ghaffari, M.R. Khoshayand, F. Atyabi, S. Azarmi, F. Kobarfard, [145] R. Sharma, D. Saxena, A. Dwivedi, A. Misra, Inhalable microparticles containing
Development and optimization of solid lipid nanoparticles of amikacin by central drug combinations to target alveolar macrophages for treatment of pulmonary
composite design, J. Liposome Res. 20 (2010) 97104. tuberculosis, Pharm. Res. 18 (2001) 14051410.
[119] J. Varshosaz, S. Ghaffari, M.R. Khoshayand, F. Atyabi, A.J. Dehkordi, F. Kobarfard, [146] H.I. Seifart, D.P. Parkin, and P.R. Donald, Stability of isoniazid, rifampin and
Optimization of freeze-drying condition of amikacin solid lipid nanoparticles pyrazinamide in suspensions used for the treatment of tuberculosis in children,
using D-optimal experimental design, Pharm. Dev. Technol. 17 (2012) Pediatr. Infect. Dis. J. 10 (1991) 827831.
187194. [147] I.I. Raad, X. Fang, X.M. Keutgen, Y. Jiang, R. Sherertz, and R. Hachem, The role of
[120] S. Ghaffari, J. Varshosaz, A. Saadat, F. Atyabi, Stability and antimicrobial effect chelators in preventing biolm formation and catheter-related bloodstream
of amikacin-loaded solid lipid nanoparticles, Int. J. Nanomedicine 6 (2010) infections, Curr. Opin. Infect. Dis. 21 (2008) 385392.
3543. [148] A.I. Shaikh, M. Musaddiq, Antibiotic-EDTA combination induced dispersal of
[121] N. El-Gendy, V. Desai, C. Berkland, Agglomerates of ciprooxacin nanoparticles Pseudomonas aeruginosa biolm, J. Pure Appl. Microbiol. 6 (2012) 363367.
yield ne dry powder aerosols, J. Pharm. Innov. 5 (2010) 7987. [149] P.K. Singh, M.R. Parsek, E.P. Greenberg, M.J. Welsh, A component of innate
[122] G. Pilcer, F. Vanderbist, K. Amighi, Preparation and characterization of spray-dried immunity prevents bacterial biolm development, Nature 417 (2002) 552555.
tobramycin powders containing nanoparticles for pulmonary delivery, Int. J. [150] Y. Yang, M.D. Tsifansky, S. Shin, Q. Lin, Y. Yeo, Mannitol-guided delivery of
Pharm. 365 (2009) 162169. ciprooxacin in articial cystic brosis mucus model, Biotechnol. Bioeng. 108
[123] Savara, Inc., Development of inhaled vancomycin for treatment of MRSA infections (2011) 14411449.
in CF, http://sbirsource.com/sbir/awards/141627-development-of-inhaled-vanco- [151] R. Wilson, T. Welte, E. Polverino, A. De Soyza, H. Greville, A. O'Donnell, J. Alder, P.
mycin-for-treatment-of-mrsa-infections-in-cf2014. Reimnitz, B. Hampel, Ciprooxacin dry powder for inhalation in non-cystic brosis
[124] N. El-Gendy, P. Selvam, P. Soni, C. Berkland, Development of budesonide bronchiectasis: a phase II randomised study, Eur. Respir. J. 41 (2013) 11071115.
nanocluster dry powder aerosols: processing, J. Pharm. Sci. 101 (2012) 34253433. [152] K.R. Allison, M.P. Brynildsen, J.J. Collins, Metabolite-enabled eradication of bacterial
[125] P. Backman, H. Adelmann, G. Petersson, C.B. Jones, Advances in inhaled technolo- persisters by aminoglycosides, Nature 473 (2011) 216220.
gies: understanding the therapeutic challenge, predicting clinical performance, [153] N. Barraud, A. Buson, W. Jarolimek, S.A. Rice, Mannitol enhances antibiotic
and designing the optimal inhaled product, Clin. Pharmacol. Ther. 95 (2014) sensitivity of persister bacteria in Pseudomonas aeruginosa biolms, PLoS One 8
509520. (2013).
[126] Savara, Inc., Savara Pharmaceuticals' AeroVanc granted U.S. orphan drug designa- [154] M.L. Aitken, G. Bellon, K. De Boeck, P.A. Flume, H.G. Fox, D.E. Geller, E.G. Haarman,
tion for the treatment of MRSA lung infection in cystic brosis patients, http:// H.U. Hebestreit, A. Lapey, I.M. Schou, J.B. Zuckerman, B. Charlton, C.F. Investigators,
savarapharma.com/savara-pharmaceuticals-aerovanc-granted-u-s-orphan-drug- Long-term inhaled dry powder mannitol in cystic brosis an international
designation-for-the-treatment-of-mrsa-lung-infection-in-cystic-brosis-patients/ randomized study, Am. J. Respir. Crit. Care Med. 185 (2012) 645652.
2012. [155] D. Bilton, E. Daviskas, S.D. Anderson, J. Kolbe, G. King, R.G. Stirling, B.R. Thompson,
[127] J. Li, C.R. Rayner, R.L. Nation, R.J. Owen, D. Spelman, K.E. Tan, L. Liojios, D. Milne, B. Charlton, B. Investigators, Phase 3 randomized study of the efcacy and
Heteroresistance to colistin in multidrug-resistant Acinetobacter baumannii, safety of inhaled dry powder mannitol for the symptomatic treatment of
Antimicrob. Agents Chemother. 50 (2006) 29462950. non-cystic brosis bronchiectasis, Chest 144 (2013) 215225.

Please cite this article as: Q.(T.) Zhou, et al., Inhaled formulations and pulmonary drug delivery systems for respiratory infections, Adv. Drug Deliv.
Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.10.022
16 Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxxxxx

[156] Y. Yang, M.D. Tsifansky, C.J. Wu, H.I. Yang, G. Schmidt, Y. Yeo, Inhalable antibiotic [186] R. Delvadia, M. Hindle, P. Worth Longest, P.R. Byron, In vitro tests for aerosol
delivery using a dry powder co-delivering recombinant deoxyribonuclease and deposition II: IVIVCs for different dry powder inhalers in normal adults, J. Aerosol
ciprooxacin for treatment of cystic brosis, Pharm. Res. 27 (2010) 151160. Med. 26 (2013) 138144.
[157] S.H. Lee, J. Teo, D. Heng, Y. Zhao, W.K. Ng, H.-K. Chan, R.B.H. Tan, Steroid-decorated [187] V. Brard, E. Lesniewska, C. Andrs, D. Pertuy, C. Laroche, Y. Pourcelot, Dry powder
antibiotic microparticles for inhaled anti-infective therapy, J. Pharm. Sci. 103 inhaler: inuence of humidity on topology and adhesion studied by AFM, Int. J.
(2014) 11151125. Pharm. 232 (2002) 213224.
[158] N. Barraud, D.J. Hassett, S.H. Hwang, S.A. Rice, S. Kjelleberg, J.S. Webb, Involvement [188] S. Sunagawa, F. Higa, H.L. Cash, M. Tateyama, T. Uno, J. Fujita, Single-dose inhaled
of nitric oxide in biolm dispersal of Pseudomonas aeruginosa, J. Bacteriol. 188 laninamivir: registered in Japan and its potential role in control of inuenza
(2006) 73447353. epidemics, Inuenza Other Respi. Viruses 7 (2013) 13.
[159] N. Barraud, D. Schleheck, J. Klebensberger, J.S. Webb, D.J. Hassett, S.A. Rice, S. [189] P. Diggory, C. Fernandez, A. Humphrey, V. Jones, M. Murphy, Comparison of elderly
Kjelleberg, Nitric oxide signaling in pseudomonas aeruginosa biolms mediates peoples technique in using two dry powder inhalers to deliver zanamivir:
phosphodiesterase activity, decreased cyclic Di-GMP levels, and enhanced randomised controlled trial, BMJ 322 (2001) 577579.
dispersal, J. Bacteriol. 191 (2009) 73337342. [190] S. Kiatboonsri, C. Kiatboonsri, P. Theerawit, Fatal respiratory events caused by
[160] N. Barraud, M.V. Storey, Z.P. Moore, J.S. Webb, S.A. Rice, S. Kjelleberg, Nitric oxide- zanamivir nebulization, Clin. Infect. Dis. 50 (2010) 620.
mediated dispersal in single- and multi-species biolms of clinically and industri- [191] H.M. Steel, A.F. Peppercorn, Fatal respiratory events caused by zanamivir nebuliza-
ally relevant microorganisms, Microb. Biotechnol. 2 (2009) 370378. tion, Clin. Infect. Dis. 51 (2010) 121.
[161] I. Kolodkin-Gal, D. Romero, S.G. Cao, J. Clardy, R. Kolter, R. Losick, D-amino acids [192] M.G. Ison, J.W. Gnann, S. Nagy-Agren, J. Treannor, C. Pava, R. Steigbigel, M.
trigger biolm disassembly, Science 328 (2010) 627629. Elliott, H.L. Weiss, F.G. Hayden, N.C.A.S. Group, Safety and efcacy of nebulised
[162] B. Burrowes, D.R. Harper, J. Anderson, M. McConville, M.C. Enright, Bacteriophage zanamivir in hospitalized patients with serious inuenza, Antivir. Ther. 8
therapy: potential uses in the control of antibiotic-resistant pathogens, Expert (2003) 183190.
Rev. Anti-Infect. Ther. 9 (2011) 775785. [193] S. Sunagawa, F. Higa, H.L. Cash, M. Tateyama, T. Uno, J. Fujita, Single-dose inhaled
[163] R.M. Donlan, Preventing biolms of clinically relevant organisms using bacterio- laninamivir: registered in Japan and its potential role in control of inuenza
phage, Trends Microbiol. 17 (2009) 6672. epidemics, Inuenza Other Respir. Viruses 7 (2013) 13.
[164] I.W. Sutherland, K.A. Hughes, L.C. Skillman, K. Tait, The interaction of phage and [194] M. Yamashita, Laninamivir and its prodrug, CS-8958: long-acting neuraminidase
biolms, FEMS Microbiol. Lett. 232 (2004) 16. inhibitors for the treatment of inuenza: a review, Antivir. Chem. Chemother. 21
[165] T.K. Lu, J.J. Collins, Dispersing biolms with engineered enzymatic bacteriophage, (2010) 7184.
Proc. Natl. Acad. Sci. U. S. A. 104 (2007) 1119711202. [195] S. Kashiwagi, S. Yoshida, H. Yamaguchi, N. Mitsui, M. Tanigawa, K. Shiosakai, N.
[166] H.-K. Chan, Formulation challenges: protein powders for inhalation, in: M. Rathbone, Yamanouchi, T. Shiozawa, F. Yamaguchi, Clinical efcacy of long-acting neuramini-
J. Hadgraft, M. Roberts (Eds.), Modied-release Drug Delivery Technology, Informa dase inhibitor laninamivir octanoate hydrate in postmarketing surveillance, J. Infect.
Healthcare, New York, 2008, pp. 631643. Chemother. 19 (2013) 223232.
[167] A. Clark, N. Dasovich, I. Gonda, H.-K. Chan, The balance between biochemical and [196] Hovione, http://www.hovione.com/twincaps/twincaps.asp.
physical stability for inhalation protein powders: rhDNase as an example, in: R. [197] K. Inoue, The Formulation Development and Device Designing of Inavir Dry
Dalby, P. Byron, S. Farr (Eds.), Respiratory Drug Delivery V, Interpharm Press, Powder Inhaler, Inhalation Asia, Hong Kong, 2013.
Buffalo Grove, IL, 1996, pp. 167174. [198] M. Yamashita, T. Tomozawa, M. Kakuta, et al., CS-8958, a prodrug of the new
[168] P. Kwok, H.-K. Chan, Pulmonary delivery of peptides and proteins, in: C.v.d. Walle neuraminidase inhibitor R-125489, shows long-acting anti-inuenza virus activity,
(Ed.), Peptide and Protein Delivery, Academic Press, London, 2011, pp. 2346. Antimicrob. Agents Chemother. 53 (2009) 186192.
[169] P. Kwok, R. Salama, H.-K. Chan, Proteins, peptides, and controlled-release formula- [199] S. Kubo, M. Kakuta, M. Yamashita, In vitro and in vivo effects of a long-acting
tions for inhalation, in: P. Colombo, D. Traini, F. Buttini (Eds.), Inhalation Drug De- anti-inuenza agent CS-8958 (laninamivir octanoate, Inavir) against pandemic
livery: Techniques and Products, John Wiley & Sons, 2012, pp. 121144. (H1N1) 2009 inuenza viruses, Jpn. J. Antibiot. 63 (2010) 337346.
[170] H.K. Chan, A. Clark, I. Gonda, M. Mumenthaler, C. Hsu, Spray dried powders and [200] N. Kawai, H. Ikematsu, N. Iwaki, et al., Clinical effectiveness of oseltamivir for
powder blends of recombinant human deoxyribonuclease (rhDNase) for aerosol inuenza A(H1N1) virus with H274Y neuraminidase mutation, J. Infect. 59
delivery, Pharm. Res. 14 (1997) 431437. (2009) 207212.
[171] H.K. Chan, I. Gonda, Solid state characterization of spray-dried powders of recom- [201] Y. Itoh, K. Shinya, M. Kiso, et al., In vitro and in vivo characterization of new swine-
binant human deoxyribonuclease (RhDNase), J. Pharm. Sci. 87 (1998) 647654. origin H1N1 inuenza viruses, Nature 460 (2009) 10211025.
[172] F. Separovic, Y.H. Lam, X. Ke, H.K. Chan, A solid-state NMR study of protein hydra- [202] Biota, http://investors.biotapharma.com/releasedetail.cfm?ReleaseID=8636262014.
tion and stability, Pharm. Res. 15 (1998) 18161821. [203] J.P. Wong, M.E. Christopher, X. Dai, G. Schnell, E.R. Stephen, D. Van Loon, S.
[173] Y.H. Lam, R. Bustami, T. Phan, H.K. Chan, F. Separovic, A solid-state NMR study of pro- Viswanathan, Aerosol and nasal delivery of vaccines and antiviral drugs
tein mobility in lyophilized protein-sugar powders, J. Pharm. Sci. 91 (2002) 943951. against seasonal and pandemic inuenza, Expert Rev. Respir. Med. 4 (2010)
[174] I. Gonda, D. Cipolla, S. Shire, K. Meserve, S. Weck, A. Clark, H.-K. Chan, A case study 171184.
in aerosol protein drug development: aqueous solution aerosols of rhDNase, in: P. [204] F. Hayden, Developing new antiviral agents for inuenza treatment: what does the
Byron, R. Dalby, S. Farr (Eds.), Respiratory Drug Delivery IV, Interpharm Press, Buf- future hold, Clin. Infect. Dis. 48 (2009) S3S13.
falo Grove, IL, 1994, pp. 4754. [205] R. Moss, T. Li, Methods, Compounds and Compositions for Treatment of
[175] R.T. Bustami, H.K. Chan, F. Dehghani, N.R. Foster, Generation of micro-particles of Parainuenza Virus in Immunocompromised Patients, 2013. (PCT/US2013/
proteins for aerosol delivery using high pressure modied carbon dioxide, 026754, WO2013123521 A1).
Pharm. Res. 17 (2000) 13601366. [206] B.E. Gilbert, M.T. McLeay, Megaribavirin aerosol for the treatment of inuenza A
[176] S. Goodall, N. Chew, K. Chan, D. Auriac, M.J. Waters, Aerosolization of protein solu- virus infections in mice, Antivir. Res. 78 (2008) 223229.
tions using thermal inkjet technology, J. Aerosol Med. 15 (2002) 351357. [207] X.L. Shi, Z.H. Shi, H. Huang, H.G. Zhu, P. Zhou, D. Ju, Therapeutic effect of recombi-
[177] R.O. Salama, D. Traini, H.K. Chan, A. Sung, A.J. Ammit, P.M. Young, Preparation and nant human catalase on H1N1 inuenza-induced pneumonia in mice, Inamma-
evaluation of controlled release microparticles for respiratory protein therapy, J. tion 33 (2009) 166172.
Pharm. Sci. 98 (2009) 27092717. [208] D. Verreault, K.S. Sivasubramani, J.D. Talton, A.L. Doyle, J.D. Reddy, S.Z. Killeen, P.J.
[178] S.H. Lee, D. Heng, W.K. Ng, H.K. Chan, R.B.H. Tan, Nano spray drying: a novel meth- Didier, P.A. Marx, C.J. Roy, Evaluation of inhaled cidofovir as postexposure prophy-
od for preparing protein nanoparticles for protein therapy, Int. J. Pharm. 403 lactic in an aerosol rabbitpox model, Antivir. Res. 93 (2012) 204208.
(2011) 192200. [209] S.K. Pitman, R.H. Drew, J.R. Perfect, Addressing current medical needs in invasive
[179] P.C.L. Kwok, A. Tunsirikongkon, W. Glover, H.K. Chan, Formation of protein nano- fungal infection prevention and treatment with new antifungal agents, strategies
matrix particles with controlled surface architecture for respiratory drug delivery, and formulations, Expert Opin. Emerg. Drugs 16 (2011) 559586.
Pharm. Res. 28 (2011) 788796. [210] J.A. Smith, C.A. Kauffman, Pulmonary fungal infections, Respirology 17 (2012)
[180] A.S. Tyne, J.G.Y. Chan, E.R. Shanahan, I. Atmosukarto, H.-K. Chan, W.J. Britton, N.P. 913926.
West, TLR2-targeted secreted proteins from Mycobacterium tuberculosis are pro- [211] M.A. Pfaller, D.J. Diekema, Epidemiology of invasive mycoses in North America,
tective as powdered pulmonary vaccines, Vaccine 31 (2013) 43224329. Crit. Rev. Microbiol. 36 (2010) 153.
[181] S. Matinkhoo, K.H. Lynch, J.J. Dennis, W.H. Finlay, R. Vehring, Spray-dried respirable [212] A.H. Limper, K.S. Knox, G.A. Sarosi, N.M. Ampel, J.E. Bennett, A. Catanzaro, S.F.
powders containing bacteriophages for the treatment of pulmonary infections, J. Davies, W.E. Dismukes, C.A. Hage, K.A. Marr, C.H. Mody, J.R. Perfect, D.A. Stevens,
Pharm. Sci. 100 (2011) 51975205. G. Amer, Thoracic soc fungal working, an ofcial American Thoracic Society
[182] L. Golshahi, K.H. Lynch, J.J. Dennis, W.H. Finlay, In vitro lung delivery of bacterio- statement: treatment of fungal infections in adult pulmonary and critical care
phages KS4-M and KZ using dry powder inhalers for treatment of Burkholderia patients, Am. J. Respir. Crit. Care Med. 183 (2011) 96128.
cepacia complex and Pseudomonas aeruginosa infections in cystic brosis, J. Appl. [213] J. Le, D.S. Schiller, Aerosolised delivery of antifungal agents, Curr. Fungal Infect. Rep.
Microbiol. 110 (2011) 106117. 4 (2010) 96102.
[183] GlaxoSmithKline, Relenza Rotadisk, (Zanamivir for inhalation) with the [214] O. Hilberg, C.U. Andersen, O. Henning, T. Lundby, J. Mortensen, E. Bendstrup,
Diskhaler inhalation device, prescription information, Research Triangle Park, Remarkably efcient inhaled antifungal monotherapy for invasive pulmonary
NCavailable at http://us.gsk.com/products/assets/us_relenza.pdf. aspergillosis, Eur. Respir. J. 40 (2012) 271273.
[184] J.A. Hedrick, A. Barzilai, U. Behre, F.W. Henderson, J. Hammond, L. Reilly, O. Keene, [215] M. Klepser, Use of aerosolised antifungals for the prophylaxis against and
Zanamivir for treatment of symptomatic inuenza A and B infection in children treatment of invasive pulmonary aspergillosis: climbing it because it is there?
ve to twelve years of age: a randomized controlled trial, Pediatr. Infect. Dis. J. Pharm. Anal. Acta S1 (2011) e001.
19 (2000) 410417. [216] S.P. Shah, A. Misra, Development of liposomal amphotericin B dry powder inhaler
[185] A. Kamiya, M. Sakagami, P.R. Byron, Cascade impactor practice for a high dose dry formulation, Drug Deliv. 11 (2004) 247253.
powder inhaler at 90 L/min: NGI versus modied 6-stage and 8-stage ACI, J. [217] L. Kuiper, E.J. Ruijgrok, A review on the clinical use of inhaled amphotericin B, J.
Pharm. Sci. 98 (2009) 10281039. Aerosol Med. Pulm. Drug Deliv. 22 (2009) 213227.

Please cite this article as: Q.(T.) Zhou, et al., Inhaled formulations and pulmonary drug delivery systems for respiratory infections, Adv. Drug Deliv.
Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.10.022
Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxxxxx 17

[218] M. Fauvel, C. Farrugia, N. Tsapis, C. Gueutin, O. Cabaret, C. Bories, S. Bretagne, G. [14] P.S. Pourshahab, K. Gilani, E. Moazeni, H. Eslahi, M.R. Fazeli, H. Jamalifar, Preparation
Barratt, Aerosolized liposomal amphotericin B: prediction of lung deposition, and characterization of spray dried inhalable powders containing chitosan nanopar-
in vitro uptake and cytotoxicity, Int. J. Pharm. 436 (2012) 106110. ticles for pulmonary delivery of isoniazid, J. Microencapsul. 28 (2011) 605613.
[219] K. Gilani, E. Moazeni, T. Ramezanli, M. Amini, M.R. Fazeli, H. Jamalifar, Development [15] Y.-J. Son, J.T. McConville, Preparation of sustained release rifampicin microparticles
of respirable nanomicelle carriers for delivery of amphotericin B by jet for inhalation, J. Pharm. Pharmacol. 64 (2012) 12911302.
nebulisation, J. Pharm. Sci. 100 (2011) 252259. [16] J. Abdulla, Y.-F. Tan, Y. Darwis, Rehydrated lyophilized rifampicin-loaded mPEG
[220] M. Alsaadi, J.L. Italia, A.B. Mullen, M. Kumar, A.A. Candlish, R.A.M. Williams, C.D. DSPE formulations for nebulization, AAPS Pharmscitech 11 (2010) 663671.
Shaw, F. Al Gawhari, G.H. Coombs, M. Wiese, A.H. Thomson, M. Puig-Sellart, J. [17] T.V.P. Doan, W. Couet, J.C. Olivier, Formulation and in vitro characterization of
Wallace, A. Sharp, L. Wheeler, P. Warn, K.C. Carter, The efcacy of aerosol treat- inhalable rifampicin-loaded PLGA microspheres for sustained lung delivery, Int. J.
ment with non-ionic surfactant vesicles containing amphotericin B in rodent Pharm. 414 (2011) 112117.
models of leishmaniasis and pulmonary aspergillosis infection, J. Control. Release [18] R. Diab, J. Brillault, A. Bardy, A.V.L. Gontijo, J.C. Olivier, Formulation and in vitro
160 (2012) 685691. characterization of inhalable polyvinyl alcohol-free rifampicin-loaded PLGA
[221] C. Rundfeldt, H. Steckel, H. Scherliess, E. Wyska, P. Wlaz, Inhalable highly concen- microspheres prepared with sucrose palmitate as stabilizer: efciency for ex vivo
trated itraconazole nanosuspension for the treatment of bronchopulmonary asper- alveolar macrophage targeting, Int. J. Pharm. 436 (2012) 833839.
gillosis, Eur. J. Pharm. Biopharm. 83 (2013) 4453. [19] K. Ohashi, T. Kabasawa, T. Ozeki, H. Okada, One-step preparation of rifampicin/
[222] F. Andrade, D. Rafael, M. Videira, D. Ferreira, A. Sosnik, B. Sarmento, Nanotechnol- poly(lactic-co-glycolic acid) nanoparticle-containing mannitol microspheres using
ogy and pulmonary delivery to overcome resistance in infectious diseases, Adv. a four-uid nozzle spray drier for inhalation therapy of tuberculosis, J. Control.
Drug Deliv. Rev. 65 (2013) 18161827. Release 135 (2009) 1924.
[223] H.-K. Chan, P.C.L. Kwok, Production methods for nanodrug particles using the [20] I. Coowanitwong, V. Arya, P. Kulvanich, G. Hochhaus, Slow release formulations of
bottom-up approach, Adv. Drug Deliv. Rev. 63 (2011) 406416. inhaled rifampin, AAPS J. 10 (2008) 342348.
[224] W. Yang, N.P. Wiederhold, R.O. Williams 3rd, Drug delivery strategies for improved [21] J. Varshosaz, S. Ghaffari, M.R. Khoshayand, F. Atyabi, S. Azarmi, F. Kobarfard,
azole antifungal action, Expert Opin. Drug Deliv. 5 (2008) 11991216. 3Development and optimization of solid lipid nanoparticles of amikacin by central
[225] W. Yang, K.P. Johnston, R.O. Williams III, Comparison of bioavailability of amor- composite design, J. Liposome Res. 20 (2010) 97104.
phous versus crystalline itraconazole nanoparticles via pulmonary administration [22] J. Varshosaz, S. Ghaffari, M.R. Khoshayand, F. Atyabi, A.J. Dehkordi, F. Kobarfard,
in rats, Eur. J. Pharm. Biopharm. 75 (2010) 3341. Optimization of freeze-drying condition of amikacin solid lipid nanoparticles using
[226] S. Jafarinejad, K. Gilani, E. Moazeni, M. Ghazi-Khansari, A.R. Najafabadi, N. Mohajel, D-optimal experimental design, Pharm. Dev. Technol. 17 (2012) 187194.
Development of chitosan-based nanoparticles for pulmonary delivery of [23] S. Ghaffari, J. Varshosaz, A. Saadat, F. Atyabi, Stability and antimicrobial effect of
itraconazole as dry powder formulation, Powder Technol. 222 (2012) 6570. amikacin-loaded solid lipid nanoparticles, Int. J. Nanomedicine 6 (2010) 3543.
[227] C. Duret, N. Wauthoz, T. Sebti, F. Vanderbist, K. Amighi, New inhalation-optimised [24] N. El-Gendy, V. Desai, C. Berkland, Agglomerates of ciprooxacin nanoparticles yield
itraconazole nanoparticle-based dry powders for the treatment of invasive pulmo- ne dry powder aerosols, J. Pharm. Innov. 5 (2010) 7987.
nary aspergillosis, Int. J. Nanomedicine 7 (2012) 54755489. [25] G. Pilcer, F. Vanderbist, K. Amighi, Preparation and characterization of spray-dried
[228] C. Duret, R. Merlos, N. Wauthoz, T. Sebti, F. Vanderbist, K. Amighi, Pharmacokinetic tobramycin powders containing nanoparticles for pulmonary delivery, Int. J.
evaluation in mice of amorphous itraconazole-based dry powder formulations for Pharm. 365 (2009) 162169.
inhalation with high bioavailability and extended lung retention, Eur. J. Pharm. [26] Savara, Inc., Development of inhaled vancomycin for treatment of MRSA infections
Biopharm. 86 (2014) 4654. in CF, http://sbirsource.com/sbir/awards/141627-development-of-inhaled-vanco-
[229] N.A. Beinborn, H.L. Lirola, R.O. Williams III, Effect of process variables on morphol- mycin-for-treatment-of-mrsa-infections-in-cf2014.
ogy and aerodynamic properties of voriconazole formulations produced by thin [27] N. El-Gendy, P. Selvam, P. Soni, C. Berkland, Development of budesonide nanocluster
lm freezing, Int. J. Pharm. 429 (2012) 4657. dry powder aerosols: processing, J. Pharm. Sci. 101 (2012) 34253433.
[28] P. Backman, H. Adelmann, G. Petersson, C.B. Jones, Advances in inhaled technolo-
gies: understanding the therapeutic challenge, predicting clinical performance,
References cited in the Tables and designing the optimal inhaled product, Clin. Pharmacol. Ther. 95 (2014)
509520.
[1] Z.L. Li, Y.L. Zhang, W. Wurtz, J.K. Lee, V.S. Malinin, S. Durwas-Krishnan, P. Meers, [29] R. Moss, T. Li, Methods, Compounds and Compositions for Treatment of
W.R. Perkins, Characterization of nebulized liposomal amikacin (Arikace (TM)) as Parainuenza Virus in Immunocompromised Patients, Nexbio, Inc., 2013
a function of droplet size, J. Aerosol Med. Pulm. Drug Deliv. 21 (2008) 245253. [30] B.E. Gilbert, M.T. McLeay, Megaribavirin aerosol for the treatment of inuenza A
[2] D. Cipolla, F. Dayton, S. Fulzele, E. Gabatan, S. Mudumba, D. Yim, H. Wu, Z. R., Inhaled virus infections in mice, Antivir. Res. 78 (2008) 223229.
liposomal ciprooxacin: in vitro properties and aerosol performance, Respiratory [31] X.L. Shi, Z.H. Shi, H. Huang, H.G. Zhu, P. Zhou, D. Ju, Therapeutic effect of recombinant
Drug Delivery Orlando, FL, 2010. human catalase on H1N1 inuenza-induced pneumonia in mice, Inammation 33
[3] D. Cipolla, H. Wu, J. Chan, H.-K. Chan, I. Gonda, Liposomal ciprooxacin for inhala- (2009) 166172.
tion retains integrity following nebulization, Respiratory Drug Delivery Europe [32] D. Verreault, K.S. Sivasubramani, J.D. Talton, A.L. Doyle, J.D. Reddy, S.Z. Killeen, P.J.
2013, Berlin, Germany, 2013. Didier, P.A. Marx, C.J. Roy, Evaluation of inhaled cidofovir as postexposure prophy-
[4] D. Cipolla, H. Wu, I. Gonda, H.-K. Chan, Aerosol performance and long-term stability lactic in an aerosol rabbitpox model, Antivir. Res. 93 (2012) 204208.
of surfactant-associated liposomal ciprooxacin formulations with modied encap- [33] M. Fauvel, C. Farrugia, N. Tsapis, C. Gueutin, O. Cabaret, C. Bories, S. Bretagne, G.
sulation and release properties, AAPS Pharmscitech (2014) 110. Barratt, Aerosolized liposomal amphotericin B: prediction of lung deposition,
[5] J. Gubernator, Z. Druis-Kawa, A. Dorotkiewicz-Jach, W. Doroszkiewicz, A. Kozubek, in vitro uptake and cytotoxicity, Int. J. Pharm. 436 (2012) 106110.
In vitro antimicrobial activity of liposomes containing ciprooxacin, meropenem [34] K. Gilani, E. Moazeni, T. Ramezanli, M. Amini, M.R. Fazeli, H. Jamalifar, Development
and gentamicin against gram-negative clinical bacterial strains, Lett. Drug Des. of respirable nanomicelle carriers for delivery of amphotericin B by jet nebulisation,
Discov. 4 (2007) 297304. J. Pharm. Sci. 100 (2011) 252259.
[6] S.J. Wallace, J. Li, R.L. Nation, R.J. Prankerd, B.J. Boyd, Interaction of colistin and [35] M. Alsaadi, J.L. Italia, A.B. Mullen, M. Kumar, A.A. Candlish, R.A.M. Williams, C.D.
colistin methanesulfonate with liposomes: colloidal aspects and implications for Shaw, F. Al Gawhari, G.H. Coombs, M. Wiese, A.H. Thomson, M. Puig-Sellart, J.
formulation, J. Pharm. Sci. 101 (2012) 33473359. Wallace, A. Sharp, L. Wheeler, P. Warn, K.C. Carter, The efcacy of aerosol treatment
[7] W. Rojanarat, N. Changsan, E. Tawithong, S. Pinsuwan, H.K. Chan, T. Srichana, with non-ionic surfactant vesicles containing amphotericin B in rodent models of
Isoniazid proliposome powders for inhalationpreparation, characterization and leishmaniasis and pulmonary aspergillosis infection, J. Control. Release 160 (2012)
cell culture studies, Int. J. Mol. Sci. 12 (2011) 44144434. 685691.
[8] A. Patil-Gadhe, V. Pokharkar, Single step spray drying method to develop [36] W. Yang, K.P. Johnston, R.O. Williams III, Comparison of bioavailability of amorphous
proliposomes for inhalation: a systematic study based on quality by design versus crystalline itraconazole nanoparticles via pulmonary administration in rats,
approach, Pulm. Pharmacol. Ther. 27 (2014) 197207. Eur. J. Pharm. Biopharm. 75 (2010) 3341.
[9] H. Adi, P.M. Young, H.-K. Chan, R. Salama, D. Traini, Controlled release antibiotics for [37] C. Rundfeldt, H. Steckel, H. Scherliess, E. Wyska, P. Wlaz, Inhalable highly
dry powder lung delivery, Drug Dev. Ind. Pharm. 36 (2010) 119126. concentrated itraconazole nanosuspension for the treatment of bronchopulmonary
[10] W.S. Cheow, M.W. Chang, K. Hadinoto, Antibacterial efcacy of inhalable aspergillosis, Eur. J. Pharm. Biopharm. 83 (2013) 4453.
levooxacin-loaded polymeric nanoparticles against E. coli biolm cells: the effect [38] S. Jafarinejad, K. Gilani, E. Moazeni, M. Ghazi-Khansari, A.R. Najafabadi, N. Mohajel,
of antibiotic release prole, Pharm. Res. 27 (2010) 15971609. Development of chitosan-based nanoparticles for pulmonary delivery of
[11] W.S. Cheow, M.W. Chang, K. Hadinoto, The roles of lipid in anti-biolm efcacy of itraconazole as dry powder formulation, Powder Technol. 222 (2012) 6570.
lipid-polymer hybrid nanoparticles encapsulating antibiotics, Colloids Surf. A [39] C. Duret, N. Wauthoz, T. Sebti, F. Vanderbist, K. Amighi, New inhalation-optimised
Physicochem. Eng. Asp. 389 (2011) 158165. itraconazole nanoparticle-based dry powders for the treatment of invasive
[12] F. Ungaro, I. d'Angelo, C. Coletta, R.D.D. Bianca, R. Sorrentino, B. Perfetto, M.A. Tufano, pulmonary aspergillosis, Int. J. Nanomedicine 7 (2012) 54755489.
A. Miro, M.I. La Rotonda, F. Quaglia, Dry powders based on PLGA nanoparticles for [40] C. Duret, N. Wauthoz, T. Sebti, F. Vanderbist, K. Amighi, New respirable and fast
pulmonary delivery of antibiotics: modulation of encapsulation efciency, release dissolving itraconazole dry powder composition for the treatment of invasive
rate and lung deposition pattern by hydrophilic polymers, J. Control. Release 157 pulmonary aspergillosis, Pharm. Res. 29 (2012) 28452859.
(2012) 149159. [41] N.A. Beinborn, H.L. Lirola, R.O. Williams III, Effect of process variables on morphology
[13] A.J. Kundawala, V.A. Patel, H.V. Patel, D. Choudhary, Isoniazid loaded chitosan micro- and aerodynamic properties of voriconazole formulations produced by thin lm
spheres for pulmonary delivery: preparation and characterization, Der. Pharmacia freezing, Int. J. Pharm. 429 (2012) 4657.
Sinica 2 (2011) 8897.

Please cite this article as: Q.(T.) Zhou, et al., Inhaled formulations and pulmonary drug delivery systems for respiratory infections, Adv. Drug Deliv.
Rev. (2014), http://dx.doi.org/10.1016/j.addr.2014.10.022
View publication stats

Das könnte Ihnen auch gefallen