You are on page 1of 15

ADR-12595; No of Pages 15

Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews


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

Emerging inhalation aerosol devices and strategies: Where are


we headed?☆
Qi (Tony) Zhou a, Patricia Tang a, Sharon Shui Yee Leung a, John Gar Yan Chan a,b, Hak-Kim Chan a,⁎
a
Advanced Drug Delivery Group, Faculty of Pharmacy, The University of Sydney, Sydney, 2006 NSW, Australia
b
Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, The University of Sydney, 2037 NSW. Australia

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

Article history: Novel inhaled therapeutics including antibiotics, vaccines and anti-hypertensives, have led to innovations in de-
Accepted 24 March 2014 signing suitable delivery systems. These emerging design technologies are in urgent demand to ensure high
Available online xxxx aerosolisation performance, consistent efficacy and satisfactory patient adherence. Recent vibrating-mesh and
software technologies have resulted in nebulisers that have remarkably accurate dosing and portability. Alterna-
Keywords:
tively, dry powder inhalers (DPIs) have become highly favourable for delivering high-dose and single-dose drugs
Inhalation therapy
Pulmonary drug delivery
with the aid of advanced particle engineering. In contrast, innovations are needed to overcome the technical con-
Nebuliser strains in drug–propellant incompatibility and delivering high-dose drugs with pressurised metered dose in-
Pressurised metered dose inhaler halers (pMDIs). This review discusses recent and emerging trends in pulmonary drug delivery systems.
Dry powder inhaler © 2014 Elsevier B.V. All rights reserved.
Computational modelling
Particle engineering
Patient adherence

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2. Nebulisers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2.1. Integration of software control . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3. Pressurised metered dose inhalers (pMDIs) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3.1. Addressing the poor patient adherence problem . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3.1.1. Breath-actuated devices . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3.1.2. Add-on devices . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3.1.3. Dose counter . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3.2. Exploring new therapeutics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3.2.1. Combination formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3.2.2. High dose antibiotics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4. Dry powder inhalers (DPIs) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.1. Innovation in DPI devices . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.1.1. Design modification and innovative concepts for passive devices . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.1.2. Active devices . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.1.3. Future directions in DPI design . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.2. Computational modelling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.3. Emerging DPI approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.3.1. Reusable DPIs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.3.2. Single-use DPIs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.3.3. Formulation considerations for device-formulation compatibility . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.4. Device material . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

☆ This review is part of the Advanced Drug Delivery Reviews theme issue on “Improving the efficacy of inhaled drugs for severe lung diseases: Emerging pulmonary delivery strategies”.
⁎ Corresponding author.
E-mail address: kim.chan@sydney.edu.au (H.-K. Chan).

http://dx.doi.org/10.1016/j.addr.2014.03.006
0169-409X/© 2014 Elsevier B.V. All rights reserved.

Please cite this article as: Q.(T.) Zhou, et al., Emerging inhalation aerosol devices and strategies: Where are we headed?, Adv. Drug Deliv. Rev.
(2014), http://dx.doi.org/10.1016/j.addr.2014.03.006
2 Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

5. Emerging inhalation therapies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0


5.1. Inhaled anti-pulmonary arterial hypertension agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.2. Inhaled antibiotics for respiratory bacterial infections . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.2.1. Pneumonia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.2.2. Tuberculosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.3. Inhaled neuraminidase inhibitor for influenza . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.4. Inhaled cyclosporine for transplant rejection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.5. Inhaled alpha 1 antitrypsin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.6. Inhaled cytokines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.7. Inhaled antibodies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
6. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

1. Introduction on the Activaero AKITA® and Philips Respironics I-Neb® adaptive aero-
sol delivery (AAD) systems.
Therapeutic applications of respiratory drugs have rapidly expanded
beyond conventional indications such as asthma and chronic obstruc-
tive pulmonary diseases (COPDs), to include inhaled antimicrobials, 2.1. Integration of software control
vaccines, and anti-hypertensives. The physico-chemical properties and
dose regimen of these newer therapies may vary significantly, thereby Originally designed to be coupled with a jet nebuliser (AKITA® JET),
necessitating new formulation techniques and device designs. As a the AKITA® system – a stand-alone SmartCard electronic control unit
result, innovative technologies in aerosol medicine are emerging in with an air compressor – can now also coordinate with newer mesh
parallel with these new therapeutic indications to ensure optimal nebulisers (AKITA® APIXNEB and AKITA2® APIXNEB) [4]. The software
aerosolisation performance, therapeutic efficacy and patient adherence. directs the compressor unit which utilises positive air pressure to con-
This review covers emerging aerosol device innovations and the evolv- trol the patient's entire inhalation manoeuvre [2]. By controlling the
ing roles of nebulisers, dry powder inhalers (DPIs) and pressurised aerosol flow rate, delivery volume and timing, the AKITA® system can
metered dose inhalers (pMDIs), as well as their impact on patient optimise aerosol release at specific periods of a patient's inspiratory
adherence. phase to target certain regions of the lungs and maximise aerosol deliv-
ery [11].
2. Nebulisers The AKITA® system has been particularly useful in gaining valuable
lung deposition and clinical trial data. To refine imaging techniques
Nebulisers generate an inhalable drug aerosol from a solution or sus- used to assess lung deposition, Fleming et al. [12] used AKITA®
pension. They are useful for treatment of respiratory diseases as asthma, system-controlled mesh nebulisation to generate radiolabelled human
COPD and cystic fibrosis (CF) [1]. The most common nebuliser type is serum albumin with a variety of different lung deposition patterns.
the jet nebuliser, which generates aerosols from the liquid medicament These were used to show that the combination of single photon emis-
using a source of compressed gas. Although relatively inexpensive, sion computed tomography and X-ray computed tomography was su-
treatment with jet nebulisers has long treatment time, the air compres- perior to planar imaging, for the analysis of aerosol lung deposition.
sors are bulky and noisy, and expensive medications are wasted in con- With application to clinical efficacy, Bakkar et al. [13] demonstrated
siderable residual volumes [2]. Many of the pitfalls are addressed by that the AKITA® system can aid in determining optimal regional deposi-
more recent vibrating-mesh nebulisers which have much greater porta- tion profiles of inhaled drugs in specific disease states. Specifically,
bility and operate silently. The low velocity plume and minimal residual nebulised dornase alpha was directed to either the smaller or larger air-
volume greatly enhance drug delivery to the lungs [3,4]. However, these ways of patients with CF. Deposition in the smaller airways demonstrat-
new devices may have a much higher upfront cost and there is a lack of ed greater improvement in therapeutic efficacy. Such data can allow
open literature demonstrating equivalent dosing of existing products to formulation and device improvements to enhance clinical response to
the more commonly used jet nebulisers. Other types of liquid aerosol inhaled medications, and minimise unwanted side effects. The technol-
generation systems include the Respimat® SoftMist™ Inhaler and ogy also opens up new opportunities for efficacy testing and clinical tri-
AERx® systems, which generate liquid aerosol by mechanically forcing als. For example, Moller et al. [14] showed that lung deposition of
a drug solution through a nozzle array [5,6]. lipopolysaccharide, an endotoxin that can trigger airway inflammation
Optimisation of this existing nebuliser technology has focused on and exacerbations in patients with COPD, can be well-controlled using
maximising aerosol lung deposition with each breath. Nebulisers typi- an AKITA® system coupled with jet nebulisation to limit the risk of ad-
cally generate aerosols throughout the entire respiratory cycle of the pa- verse effects. This could allow safe testing of new treatments for COPD
tients, leading to a significant loss during expiration. Thus mechanical by using an AKITA® system controlled lipopolysaccharide challenge
regulation of aerosol generation has been implemented in breath- test. Therefore, the AKITA® system has established new avenues in in-
enhanced (Pari LC® Star) or breath-actuated (e.g. Trudell AeroEclipse® haled delivery such as regional targeting of nebulised drugs and in refin-
II) jet nebulisers. These limit aerosol loss by mechanically restricting ing clinical trials.
the majority of aerosolisation to the inspiratory phase [1,7,8] and have Similarly, the I-Neb® consists of an AAD software control unit and a
been shown to shorten treatment time whilst minimising wastage of mesh nebuliser but combined into a single handheld device [15]. The
expensive medications [9,10]. However, these are relatively crude com- I-Neb® software has two modes of operation — a tidal breathing mode
pared to the more recent integration of digital control systems with and a targeted-inhalation mode. Whilst tidal breathing mode adapts
nebulisers, which provide much more precise regulation of aerosol de- device aerosolisation to a patient's regular tidal breathing patterns, the
livery. These electronic systems further reduce treatment time by latter uses a unique vibratory feedback mechanism to guide the patient
personalising aerosolisation to an individual patient's breathing pattern, towards an optimal breathing pattern that maximises aerosol deposi-
thereby maximising lung drug deposition. Here we provide an update tion for a shorter treatment time.

Please cite this article as: Q.(T.) Zhou, et al., Emerging inhalation aerosol devices and strategies: Where are we headed?, Adv. Drug Deliv. Rev.
(2014), http://dx.doi.org/10.1016/j.addr.2014.03.006
Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx 3

Accurate and efficient dosing with the I-Neb® has made it increas- continuous efforts are required to advance pMDI technology in order
ingly favourable for use with novel experimental therapies. Diaz et al. to retain its relevance in inhalation therapies. Thus the question is: in
[16] examined inhaled interferon gamma in the treatment of idiopathic shaping the next generation of pMDI use, what can we expect?
pulmonary fibrosis and found that total in vivo lung deposition was 65%
with the I-Neb®, compared to just 7% with a conventional jet nebuliser. 3.1. Addressing the poor patient adherence problem
Importantly, interferon gamma was aerosolised without affecting the
efficacy of the protein. Patients used the I-Neb® three times a week, Many patients experience difficulty in coordinating the actuation
for eight weeks, with adherence of approximately 97%, demonstrating and inhalation, leading to sub-optimal disease control and increased
feasibility of the device for long term treatment. In a novel preliminary risk of hospitalisation [21]. Two approaches, use of breath-actuated
trial of inhaled iloprost in patients with precapillary pulmonary hyper- pMDIs and add-on devices like spacers and valved holding chambers
tension and right-side heart failure, Martischnig et al. [17] cited accurate have been implemented to overcome this problem. Another frequent
dosing and short treatment time for choosing the I-Neb® over jet and ul- complaint from pMDI users is that it is very difficult to determine
trasonic nebulisers. Finally, the I-Neb® might also be used to improve when their pMDIs will be empty [22]. This has been addressed by the in-
clinical efficacy of existing treatments. In a group of patients with corporation of a dose counter in the pMDI device.
COPD, Goodman et al. [18] asked patients to use the I-Neb® in lieu of
their existing nebuliser system for administration of bronchodilator 3.1.1. Breath-actuated devices
and/or corticosteroid therapy. This led to significant improvements in The first breath-actuated pMDI was introduced by 3M in the
dyspnea and fatigue, which may also have been associated with better early 1970s (Autohaler™), and many other designs have been
adherence or more correct use of the I-Neb® device. Thus the I-Neb® marketed since [20]. Though improved treatment outcomes with
system is establishing its advantage both for clinical trials, as well as en- breath-actuated pMDI systems were reported for patients with
hanced delivery of current and future inhaled therapeutics. poor coordination [23,24], the increased device cost and complexity
The AKITA® and I-Neb® systems have potential to considerably im- have hindered a wider market adoption. Therefore, simpler and more
prove treatment times, aerosol deposition and patient adherence. In ad- cost-effective breath-actuation and breath-coordination technologies
dition, they augment the scope and consistency of clinical trials with are necessary for realising their benefits to larger patient populations.
more accurate dosing and adherence logging systems. Such digital Recent development of the I-Breath coordination cap [25] is a good ex-
logging systems may allow telecommunication between patients and ample of attempts to improve breath-coordination. It contains a flexible
healthcare professionals regarding treatment parameters and feed- coordination cap designed to fit onto a conventional pMDI. In the rest-
backs, which have a potential to increase treatment adherence and ing mode, multiple airtight sealing slits made of deformable materials
reduce hospital visits [6,19]. Hence, we expect to see growing software are closed to prevent a patient from substantial inhalation prior to actu-
integration for nebulisers that evolve with future technological ation. When the canister is pressed, the slits in the coordination cap are
advances. However, increased adoption of these newer technologies forced to open by compression, allowing airflow during patient inhala-
desperately requires economic evaluation of these devices and im- tion. The clinical study conducted by Azouz et al. [26] showed that the
plementation of electronic technology advances to improve device patients' coordination between inhalation and actuation was signifi-
portability. Long-term clinical trials are also needed to determine cantly improved when using the I-breath cap together with simple
patient acceptance, optimal dosing regimens and to assess patient instructions.
outcomes [3].
The newer vibrating-mesh nebulisers have addressed many of the 3.1.2. Add-on devices
concerns associated with previous nebuliser types and are unlikely to Spacers and valved holding chambers are considered as add-on de-
be superseded soon. If cost-effectiveness of these devices can be demon- vices for pMDIs. They increase the delay time between firing and inha-
strated in future studies, we anticipate an increased uptake of these lation, and allow patients to inhale the medication slowly and deeply,
mesh devices by patients, particularly in the domiciliary setting. Whilst to increase lung deposition [27,28]. They also help reduce the fraction
in general, mesh devices will replace older nebulisers, the unique fea- of large particles deposited in the oropharynx and hence minimise
tures of the I-Neb® such as the individualised programming of device local adverse effects [29,30]. The spacers and valved holding chambers
settings to each drug, may continue to restrict its application to a limited will continue its popularity among the young children and elderly pa-
number of formulations. We anticipate growth in the number of clinical tient groups who cannot coordinate their breathing with actuation
studies with both the AKITA® and I-Neb® units, particularly those inves- [31]. A drawback associated with the use of plastic is the build-up of
tigating regional lung deposition. Increased understanding of lung de- static charges on the spacer wall, decreasing the drug delivery efficien-
position will see revisions and optimisation of their existing software, cy. This issue could be largely resolved by antistatic plastic spacers
and potentially an advent of other competing software-integrated (AeroChamber Plus®, Trudell) and metal valved holding chambers
nebuliser devices. As electronic technology evolves, a reduction in the (Vortex®, Pari) [32]. One direction for continuing spacer development
physical size and weight of the software-controlled AKITA® and will be optimising spacer geometry and valve design [33], and applying
I-Neb® units is envisaged, which should further aid acceptance by anti-static materials [34] to improve their performance.
patients.
3.1.3. Dose counter
3. Pressurised metered dose inhalers (pMDIs) The importance of integrated dose counters on the new pMDIs in
development was emphasised in guidelines (but not mandatory) issued
The pMDI has been favoured by patients and healthcare profes- by the US Food and Drug Administration (FDA) [35]. Mechanical dose
sionals for the treatment of asthma and COPD since its introduction counters are designed to rely on an active event of firing such as
(Medihaler Epi™) in 1956 [20]. Whilst the basic designs remain rela- sound, temperature or pressure change, with their reliability proven
tively unchanged, significant improvements in the sub-components of clinically [36,37]. Electronic dose counters have also been developed
pMDI were achieved in the past two decades, driven mainly by the [38,39], but at a relatively higher cost and concerns remain regarding
transition of the chlorofluorocarbons (CFCs) to the hydrofluoroalkanes the reliability of battery life. Nonetheless, the incorporation of dose
(HFAs) propellants (or the even more environmentally-friendly counters will become essential for pMDI development to improve
hydrofluoroolefins (HFOs), although this is unlikely to occur in the disease management, by preventing patients from using their inhalers
near future) and stringent regulations on dosing consistency. Facing beyond the recommended number of doses and thus receiving subopti-
increasing competition from DPIs and novel soft-mist inhalers, mal treatment [40].

Please cite this article as: Q.(T.) Zhou, et al., Emerging inhalation aerosol devices and strategies: Where are we headed?, Adv. Drug Deliv. Rev.
(2014), http://dx.doi.org/10.1016/j.addr.2014.03.006
4 Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

3.2. Exploring new therapeutics Typical pMDI valves are designed to deliver a volume ranging from
25 to 100 μL per actuation. A direct approach to deliver high doses of
3.2.1. Combination formulations drug is to utilise larger metering valve sizes. However, Stein et al. [56]
Clinical outcomes have proven the benefits of combination therapies showed only a modest increase in respirable dose with increasing
of long-acting β2 agonists (LABAs) and corticosteroids [41,42], as well valve size because of the decreased drug delivery efficiency. The pres-
as LABAs and long-acting muscarinic antagonists (LAMAs) [43,44] for sure generated from the propellant in the metering chamber decreases
the management of asthma or COPD. These findings have led to the as the formulation is being released, and eventually the pressure be-
commercialisation of a number of combination formulations and more comes too low to effectively drive out the residual contents, reducing
are under development [45]. Recently, triple therapies combining the overall drug delivery efficiency. This phenomenon becomes more
LBAB/LAMA/corticosteroids have also been proposed for the COPD pa- obvious for a larger valve size. To address this problem, a built-in
tients with concomitant asthma and frequent exacerbations [46,47]. pressuriser was proposed by Alston et al. [57] to maintain the pressure
With the inhalation therapy moving towards combination products, of the metering chamber at a certain level throughout the whole actua-
continual efforts are required to address the various technical chal- tion process. A metering valve size larger than 100 μL was shown to be
lenges presented in combining multiple drug components in a single feasible with this design and the highest delivered dose achieved was
pMDI canister. 5 mg of insulin using a formulation described by Dellamary et al. [58].
Drugs in pMDIs can exist in either solution or suspension form. The Though this technique has been introduced for almost a decade, a
limitation of developing solution pMDI combination formulation falls wide adoption in field has not been seen.
in the dissimilar solubilities of drug components in the HFA propellants. For solution formulation, the solubility of drugs in HFA is a major
Foster® (beclomethasone/formoterol fumarate) is the only pMDI of limiting factor. Ethanol is generally employed as a co-solvent to improve
combination solution in the market. The can-in-can pMDI design pro- the solubility of polar drugs, but negative impacts on the aerosol perfor-
posed by Chiesi Farmaceutici S.p.A. [48], in which drugs are stored sep- mance were reported [59]. It is not an easy task to develop high dose
arately in two reservoirs (one within the other), offers a promising suspension formulations either. The drug delivery efficiency was
approach for combination solution formulations. In such design, combi- found to decrease with increasing drug levels [56]. Stronger particle–
nation drugs are not dispensed concomitantly but sequentially upon particle interactions are expected for formulations with high drug con-
two actuations. centrations, leading to drug flocculation. Further advancements in for-
The majority of the commercial combination pMDI products are in mulation technologies are essential for the realisation of high-dose
the suspension form [45]. Drug deposition on the canister and metering pMDIs.
valve surfaces used to be a problem for many suspension formulations.
The situation has been resolved to a large extent with advanced surface 4. Dry powder inhalers (DPIs)
coating technologies [49] and fast-fill fast-empty metering valves where
the metering chamber is open to the bulk formulation [50]. Therefore, 4.1. Innovation in DPI devices
the biggest challenge for developing a combination product lies in en-
suring the suspension stability of individual drugs in the same canister. The amount of fine drug particles delivered to the lungs is governed
One way to improve suspension stability is using HFA soluble polymers by the balance of varying forces including interparticulate forces within
(e.g. PEG and PVP) and ethanol-dissolved oleic acid, as suspension powders, dispersion forces by airflow through an inhaler device and
stabilizers. This approach has been adopted in two commercial prod- deposition forces in airways. Fine powders generated through micro-
ucts Symbicort® (budesonide/formoterol fumarate) and Dulera® nisation generally have high surface energy hence are notoriously cohe-
(formoterol fumarate/mometasone furoate). However, the co- sive. An inhaler device is necessary to disperse such powders into
formulation effect – apparent as individual drug components give inhalable aerosols. Aerosolisation behaviour of a DPI thus depends on
different aerosol performance due to drug–drug interactions and/ both the powder formulations and the inhaler devices. Whilst much ef-
or different particle size distributions – has been reported for most forts have been focused on formulations in the past [60], there is a grow-
existing combination formulations [45,51], and will remain a key ing need to understand and improve the device design. Modification
technical challenge. and innovation on inhaler design can provide improvements in both
The established spray-drying-based PulmoSphere™ technology has powder aerosolisation and patient adherence.
offered a promising potential to develop combination pMDI formula-
tions with minimum co-formulation effect [52]. Recently, Lechuga- 4.1.1. Design modification and innovative concepts for passive devices
Ballesteros and co-workers proposed a novel extension of this approach The powder dispersion mechanisms in DPIs are complex, depending
to minimise the co-formulation effect, by co-suspending drug micro- on the specific device, and involving air turbulence [61,62] and impac-
crystals with spray dried porous phospholipid particles (PulmoSphere tion [60,63]. Recent inhaler designs attempt to enhance powder de-
™) [45,53]. Single, dual and triple formulations of formoterol fumarate, agglomeration or drug-carrier detachment by increasing air turbulence
glycopyrrolate and mometasone furoate were successfully produced, and particulate collisions.
with the aerosol performance (fine particle fraction (FPF) of 55%) de- Addition of collision or turbulence enhancers in the airflowpath can
pending solely on the properties of porous particles, regardless of the be a simple way to improve de-agglomeration. Such enhancers could be
drugs and dose levels. This co-suspension technique provides a platform in the form of a 3D array of rods [64], or an oscillating bead [65].
to formulate a wide range of respiratory drugs, as well as using the same However, not all the modified designs can substantially improve
device configuration to deliver different formulations. aerosolisation efficiency. For the Clickhaler®, the grid mesh did not sig-
nificantly increase the respirable doses of salbutamol sulphate or
beclometasone dipropionate [66]. This is because modifying grid affects
3.2.2. High dose antibiotics not only the impaction or turbulence, but also the flow trajectory [67,
There is a clear trend towards the use of antiobiotics for respiratory 68]. In contrast, a modified Handihaler® equipped with a 3D array of
infections [54,55], with administration currently limited to DPIs and rods in the mouthpiece improved the FPF5μm/ED (FPF of b5 μm relative
nebulizers. The absence of antibiotic formulations in the pMDI form is to the emitted dose) from 87.6 to 97.3% at a flow rate of 45 L/min for
primarily due to the small dose range that can be afforded by pMDI a submicron excipient enhanced growth formulation [64]. Computa-
per actuation, generally 1 mg or less. To maintain the competitiveness tional fluid dynamics (CFD) analysis revealed that a much higher non-
of pMDIs for pulmonary delivery, technologies to expand their dosing dimensional specific dissipation factor (a proposed parameter combin-
range are necessary. ing the effects of turbulence, eddy length scale, and particle exposure

Please cite this article as: Q.(T.) Zhou, et al., Emerging inhalation aerosol devices and strategies: Where are we headed?, Adv. Drug Deliv. Rev.
(2014), http://dx.doi.org/10.1016/j.addr.2014.03.006
Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx 5

time) contributed to the improved aerosolisation behaviour compared 45 L/min, demonstrating high aerosolisation efficiency. Surprisingly,
to the 2D mesh device and unmodified device [64,69]. the drug degradation is less than 5% for the aerosols of 175 medications
Conix™ is a passive DPI device utilising a reverse-flow cyclone tech- [78], due to the extremely short exposure time of the drug to the hot
nology [70] to generate a high velocity energised vortex to provide substrates [76].
shear and collision for de-agglomerating powders. An interesting device It is worth noting that although the dispersion energy of active de-
feature is the retention of large carrier particles in the inhaler as a colli- vices does not rely on inspiration flow, throat deposition of drug aero-
sion media to promote de-agglomeration [70]. sols could be dependent on the patient's inhalation pattern because
Other innovative design concepts go beyond the conventional ap- the main mechanism of throat deposition is inertial impaction. Thus,
proaches of generating shear forces and/or impactions by inhalation to drug deposition in the oropharynx may vary substantially for patients
disperse the powder. In a prototype flutter-aerodynamic device, an with different inhalation capacities [79].
aeroelastic film coated with drug particles was fluttered by the inspira-
tory flow and drug particles were dislodged from the film during oper- 4.1.3. Future directions in DPI design
ation [71]. For high dose drugs, it is crucial to ensure sufficient loading to Passive devices based on traditional dispersion mechanisms will
and consistent detachment of the particles from the film. A single-use continue to dominate the DPI markets in the near future. In past de-
DPI based on the ActiveMesh™ technology was developed with a cades, device efficiency has been less significant as the majority of med-
mesh-sieving dispersion principle. Powder formulation is packed in a ications, such as those for asthma, only require low drug doses (b1 mg).
mesh compartment inside the inhaler. Driven by the airflow during in- However, this has become a crucial aspect with the increasing interests
halation, the mesh package vibrates and forces the powder agglomer- of delivering high dose active agents (N100 mg) such as antibiotics. It is
ates through the mesh holes to promote de-agglomeration [72]. Since a challenge to deliver such large amounts of powder without
the mesh vibration is a function of airflow rate, device emptying and compromising the device portability and patient adherence. Further-
de-agglomeration may be a concern at low flow rates for cohesive pow- more, deposition of a high-dose powder in the oropharynx by a low-
ders which are difficult to be released from the small holes [72]. A 45 μm efficiency inhaler could cause severe side effects such as coughing or
size mesh plus a turbulence-generating insert were shown to signifi- throat irritation, resulting in withdrawal of inhalation therapy.
cantly improve the device performance [73]. In both the fluttering and Most passive DPIs are only prescribed to patients aged ≥ 6 years,
mesh-sieving designs, airflow is not the direct source for powder dis- leaving nebulisation the only choice for the younger children. Active
persion; whilst both de-agglomeration processes are still driven and af- DPIs can open a new window for the inhaled paediatric therapies. An-
fected by the airflow rate. However, they provide new concepts for other opportunity for active DPIs is in the delivery of dry powder aerosol
dispersing drug powders, which could circumvent the patent issues of to the critically ill patients. In the past, DPIs were unsuitable for the in-
traditional inhalers. cubated patients because patients cannot generate the required airflow
to disperse the powder from passive inhalers. However, in a recent
4.1.2. Active devices study, Tang et al. [80] has developed a novel powder delivery system
Passive devices have been the mainstream for aerosolising powder suitable for incubated patients with the aid of a ventilation bag. This sys-
formulations. However, the dispersion performance of many passive tem has been applied to deliver the mannitol powder to the distal end of
devices suffers from being airflow dependent. This problem is eliminat- tracheal tubes in the critically ill patients (Fig. 1) [81].
ed in the active devices which utilise the energy provided externally
such as compressed air (e.g. Exubera® and Aspirair®), electrical vibra- 4.2. Computational modelling
tion (e.g. MicroDose), and heat (e.g. Staccato®). So far, active devices
are not as popular as passive ones due to higher cost and reduced With the rapid advancements of the computational technologies,
portability. CFD has become a useful tool for inhaler designs in providing informa-
A multi-dose (up to 64) active device using compressed air has been tion on the air flow patterns, turbulence levels and particle trajectories
recently developed to deliver micro-doses of drug (b20 μg) [74]. Drug within and post device [82,83]. Different modelling approaches used
powders are pre-filled in small pocket holes drilled through a rotating for pharmaceutical aerosol dispersion with corresponding advantages
disc, with only one drug pocket positioned in the air passage at a time. and limitations were reviewed by Wong et al. [82] and Ruzycki et al.
Two different compressed airflows were applied with the primary [83]. Whilst few studies focusing on optimising the design variables
flow dispersing the drug powder from the pocket and the secondary for pMDIs [84,85] and nebulizers [86,87], CFD modelling has been
flow fluidising and pushing the powder into the primary flow path. most prevalent for DPIs. However, the complicated processes involved
The FPF values of four tested powders (spray dried insulin, phenylala- in DPI dispersion have made it challenging to elucidate the underlying
nine, nitrendipine and phenylalanine labelled fluorescein isothiocynate) powder de-agglomeration mechanisms by pure numerical analysis.
ranged from 50 to 70% measured by an Anderson Cascade Impactor at Therefore, using a combination of CFD simulations and in vitro ex-
28.3 L/min [74]. This active device has a potential to deliver expensive ac- periments is common for providing insights into the relative contribu-
tive ingredients, although filling such small amount of cohesive drug tions of the various dispersion mechanisms on the aerosolisation
powders into the disc can be challenging. performance.
A piezoelectric system has also been employed to actively aerosolise Coates et al. [61,67,88–90] were among the first to use coupled CFD
drug powders [75]. In the MicroDose DPI, powders are stored in a blis- and in vitro experiments to study the Aerolizer® for a carrier-free
ter. When the inhaler is activated by inspiration, the blister is opened formulation. Results on the effects of the grid design, air inlet di-
and powders are in contact with a piezo vibrator. In vitro results of an mension, capsule size, mouthpiece dimension and air flow have
atropine sulphate and lactose powder formulation aerosolised via the been summarised by Wong et al. [82] and de Boer et al. [91]. Jiang
MicroDose DPI showed a FPFtotal (b5.8 μm) of 57% at a flow rate of et al. [92] further investigated the effect of extending the height of
28.3 L/min [75]. the swirling chamber of the Aerolizer® and found improved aero-
Instead of mechanically dispersing the powder, the Staccato® device solisation performance which was ascribed to a lengthened powder–
uses heat to vaporise drugs followed by condensation into inhalable air interaction time.
aerosols [76]. Drugs are coated on a metallic strip and vaporised with The performance of the Aerolizer® and HandiHaler® for carrier-
a high temperature up to 400 °C within 0.2 s [77]. The drug vapour con- based formulations was investigated by Donovan et al. [93]. CFD analy-
denses quickly and generates solid aerosols. The emitted doses were sis showed that the carrier particle–inhaler collision frequency was
above 89% and FPFemitted (FPF b 6 μm relative to the emitted drug increased with the carrier particle size in the Aerolizer®, but not in the
mass) values were in the range of 80–93% at flow rates of 15, 30 and HandiHaler®. Therefore, they attributed the improved aerosol

Please cite this article as: Q.(T.) Zhou, et al., Emerging inhalation aerosol devices and strategies: Where are we headed?, Adv. Drug Deliv. Rev.
(2014), http://dx.doi.org/10.1016/j.addr.2014.03.006
6 Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

Fig. 1. A novel system delivering powders to the incubated patients. The schematic shows how to determine the dose and particle size distribution of mannitol aerosol coming out from this
system.
Reprint from Ref. [80] with kind permission from Mary Ann Liebert, Inc.

performance of the larger lactose carrier formulations in the Aerolizer® de-agglomeration on mechanical impaction [96]. They found that the
to the higher carrier–inhaler collision, and suggested impaction was a first impaction only broke the agglomerate into small fragments with
major de-agglomeration mechanism. Shur et al. [94] also computation- a weakened structure whilst the subsequent impaction disintegrated
ally studied the flow characteristics within the HandiHaler® and them into fine particles. They further proposed that the fine particle
Cyclohaler (same design as the Aerolizer®) to identify the key design fraction could be expressed as a unified error function of the ratio be-
features of inhalers. They fabricated two modified versions of the tween the impaction energy and agglomerate strength (Fig. 2).
Cyclohaler with the same specific resistances as the HandiHaler® to In their follow-up study [63], a fully coupled CFD–DEM model was
match the aerosol performance for the carrier-based Spiriva® formula- applied to study the dispersion of multiple agglomerates in the
tion. The two modified designs exhibited very different cyclonic flow Aerolizer® and identified the particle–wall impaction being the primary
behaviours and only the one with similar flow properties as that of de-agglomeration mechanism instead of the turbulence flow or
the HandiHaler® attained comparable performance. Longest et al. [64] particle–particle collisions. Sequential impactions of agglomerates
later utilised CFD to investigate the mouthpiece designs of HandiHaler®, with the inhaler base and grids led to a significant increase in the FPF.
as discussed in Section 4.1.1. Though the dispersion mechanisms were elucidated, Tong et al. [63]
With the increasing interests in developing high-dose DPI, de Boer emphasised that the information collected were specific for the formu-
et al. [91] evaluated computationally and experimentally the per- lation and device studied. Recently, they applied CFD–DEM analysis
formance of the high-dose disposable Twincer™ DPI to seek product
improvement. CFD results revealed that the flow split was independent
of pressure drop across the inhaler and the bypass channel had little
contribution to the swirl in the classifier. Therefore, they proposed
blocking the bypass channel to reduce the total flow rate to minimise
the mouth and throat deposition without affecting the dispersion
efficiency.
Whilst the aforementioned studies have provided considerable
insights in the mechanisms of powder de-agglomeration, the collected
data can only be treated qualitatively due to the lack of information
on particle interactions. To achieve quantitative analysis of the de-
agglomeration mechanisms, specific interests have been placed to cou-
ple the CFD with discrete element method (DEM) modelling which can
take into account the mechanics of particle–particle, particle–fluid and
particle–device interactions during powder dispersion.
A one-way coupled CFD–DEM approach was employed by Tong et al.
[95] to study the de-agglomeration of a single agglomerate comprising
particles of various sizes and poly-dispersities in a cyclonic flow model
similar to the Aerolizer®. They found that the breakup of agglomerate
into smaller fragments was governed by the particle–particle tensile Fig. 2. Correlation between FPF and the ratio between the impact energy and agglomerate
strength and particle–wall impact energy. Later, they used a two-way cohesion energy (ϕ).
coupled CFD–DEM model to investigate the mechanism of powder Reprint from Ref. [96] with kind permission from Elsevier.

Please cite this article as: Q.(T.) Zhou, et al., Emerging inhalation aerosol devices and strategies: Where are we headed?, Adv. Drug Deliv. Rev.
(2014), http://dx.doi.org/10.1016/j.addr.2014.03.006
Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx 7

coupled with in vitro experiments to identify the critical design features dose is not inhaled [97]. In a randomised cross-over comparison clinical
of the Aerolizer® for carrier based formulations (Foradile®). CFD–DEM study of NEXThaler®, Diskus®, and Turbuhaler® in 66 subjects, the
analysis in the inhaler and USP induction port showed that high- NEXThaler was rated as the easiest to use and most preferred inhaler
velocity particle–wall impactions in the inhaler dispersion chamber with a significantly lower number of using error [98]. Reloadable or re-
contributed mainly to the detachment of fine drug particles from the usable inhalers are favourable to administer frequently used medica-
coarse carriers (Fig. 3) [68]. tions. Most capsule-based DPIs belong to this group and are still active
CFD–DEM modelling is more computationally time-consuming in the DPI market. In the last five years, there is an increasing trend to
compared with CFD alone. However, with the increase in the power of explore new applications of DPI to deliver antimicrobials, vaccines or
advanced processors, it is envisaged that advanced computations will other single-dose active ingredients. As a result, single-use devices are
play a more pivotal role in inhaler design in the near future. specially designed for the transient DPI therapies.

4.3. Emerging DPI approaches 4.3.1. Reusable DPIs


In reusable DPIs, the powder formulation is prefilled in a capsule or
DPIs can be grouped into four classes: multi-unit, multi-dose reser- cartridge, which is then loaded into the inhaler device for use. With
voir, reusable single dose, and single-use devices. The choice of device the dose storage compartment being detached from the inhaler, reus-
is dependent on dose, dosing frequency, and powder properties. able inhalers can come with smaller dimensions. Since the device can
Multi-dose devices are popular for the frequently used medicines with be reused many times, the cost of medication per dose is significantly
low doses such as anti-asthma drugs. There has been an increasing reduced. In some cases, more than one dosing unit can be given, making
emphasis on simplifying the operation procedure to reduce incorrect dosing more flexible.
device operation and improve patient adherence. For example, Reusable devices are currently used for high dose antibiotics be-
NEXThaler® is a multi-dose inhaler designed with a simple operation cause a multi-dose inhaler will be too bulky for the many high-dose
procedure: open the lid, inhale from the mouthpiece and close the lid. units required. Both TOBI® Podhaler® (tobramycin) and Colobreathe
A full dose feedback system was built into the NEXThaler® using a Turbospin® (colistimethate sodium) are capsule-based reusable DPI.
breath-actuated mechanism [97]. The inhaler is activated only when a Passive reloadable devices can achieve satisfactory aerosol performance
certain inspiration flow rate is reached to avoid miscounting if the with minimum flow rate dependence when drug powders are engi-
neered, which will be discussed in Section 4.3.3.

4.3.2. Single-use DPIs


Single-use DPIs provide an economical means for the pulmonary
drug delivery of less-frequent or single-dose applications, e.g. inhaled
vaccination. Design of disposable DPIs is quite different from that of
the multi-dose counterparts. Besides the general requirements of dose
consistency and dispersion efficiency, the single-use DPI products
should also incorporate the features of simple operation procedures
and low-costs. The TwinCaps® is a disposable passive DPI device for
delivery of laninamivir octanoate (Inavir®), long-acting neuraminidase
inhibitor prodrug, for the treatment of influenza. Unlike its short-
acting predecessor, zanamivir that requires administration twice daily
for five days, a single dose of 40 mg of inhaled laninamivir can be effec-
tive for a week, making it a perfect candidate for a disposable DPI. The
design of TwinCaps® is simple, with only two assembled plastic parts
consisting of a main inhaler body and a pre-filled powder reservoir
chamber. Since the Inavir® TwinCaps® has been approved for children
under 10 years old, a sufficient aerosolisation performance at lower
flow rates will be necessary [68].
Inhaler device contamination is a serious concern for nebulisation
treatments. It has been reported up to 65% of nebulisers were contami-
nated by microbes including pathogenic Pseudomonas aeruginosa after
home use by CF patients [99–101]. The use of DPIs as an alternative
may reduce the risk of retaining pathogenic bacteria because of dry con-
ditions in both the drug powder and inhaler. However, in a previous
study, Staphylcoccus aureus was still found in patient-returned DPI
devices (2 out of 11 DPIs) [102]. Therefore disposable devices go a
step further to mitigate the potential contamination due to inappropri-
ate cleaning and storage.
The Twincer™ is a disposable device used to deliver antibiotics of co-
listin for treatment of chronic infections in CF patients. It has three plas-
tic plate-like parts which can be easily assembled by stacking and
clicking three pieces together [103]. The powder formulation is packed
in a blister-like compartment [91]. The powders are dispersed through a
multiple air classifier technology [103]. Unlike Inavir® which requires
only a single administration, inhaled colistin therapy for CF patients
Fig. 3. Spatial distribution of total carrier particle–wall collision in the inhaler and USP in-
duction port by CFD–DEM analysis of the (a) original Aerolizer; and (b) cross-gird
can be administered twice daily for at least 28 days, which makes the
Aerolizer (colours represent impaction velocity between the particle and wall). cost issue more prominent. However, the disposable design is advanta-
Reprint from Ref. [68] with kind permission from Springer. geous for reducing the potential risk of bacterial contamination, an issue

Please cite this article as: Q.(T.) Zhou, et al., Emerging inhalation aerosol devices and strategies: Where are we headed?, Adv. Drug Deliv. Rev.
(2014), http://dx.doi.org/10.1016/j.addr.2014.03.006
8 Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

which is also critical for the treatment of highly contagious diseases 4.4. Device material
such as tuberculosis [104].
Aespira's resQhaler™ is a recently developed single-use disposable Powder retention in inhaler devices may be significant, and can
DPI based on the ActiveMesh™ technology, as discussed in Section 4.1.1 reach 20%–50% of the loaded doses for cohesive powders [114–116].
[73]. The testing doses of resQhaler™ are up to 3 mg, which may limit This can be due to mechanical impaction of the powder on the device
its use for high-dose drugs until further design improvements can accom- surface and/or electrostatic charge generated during powder dispersion.
modate more powder [72]. Triboelectrification is an inevitable consequence of particle–particle or
particle–device contact or friction during the processing dispersion
4.3.3. Formulation considerations for device-formulation compatibility [117]. As pharmaceutical solids and most DPI devices are insulative,
The compatibility between device design and formulation properties the generated electrostatic charge is difficult to dissipate [118]. Both im-
should be considered carefully. There is no device which can fit all paction and charge depend on the surface roughness [119], contact area
formulations; in turn, no formulation is suitable for all devices. For [120] and particulate physico-chemical properties [121,122]. Hence,
most DPIs, good flow and fluidisation properties are required for the modification of device surface characteristics may effectively reduce
powder formulation with regard to manufacturing and drug release the device retention by minimising attractive forces.
[105]. In the traditional DPIs of asthma or COPD therapies, coarse carrier Pharmaceutical lubricants such as MgSt and leucine have a low sur-
(mostly lactose) is a common excipient to improve the flow and face energy, which can reduce adhesion and friction between particle–
fluidisation behaviour of powder formulations. Because of the low particle or particle–inhaler surface [105]. MgSt is a widely used lubri-
doses of bronchodilator and corticosteroids (typically b1 mg), lactose cant for tabletting, and is an approved excipient for inhalation products
carriers are also functioning as diluents to facilitate the manufacturing. by the US FDA (Foradil® Certihaler®) [123], UK Medicines and
However, for the high-dose antibiotic DPI formulations (N 100 mg), ad- Healthcare products Regulatory Agency (MHRA) (Pulmicort® CFC-free
dition of lactose will increase the powder mass and volume excessively. Inhaler) [124] and many other countries. Given its established safety
Therefore, particle engineering has been extensively explored to pro- profiles, coating of MgSt [110,125,126] and leucine [127–129] on
duce drug particles with satisfactory flow and fluidisation properties inhalable particles has been investigated extensively to improve device
without adding a carrier. Powders with superior flow and aerosolisation retention and aerosolisation by decreasing particle surface energy and
performance will not only facilitate dose preparation during powder cohesion [130]. Recently, Heng et al. have proposed a new
manufacturing, but also reduce the airflow dependence of aerosol per- alternative strategy of coating the interior surfaces of capsules and DPI
formance. Porous tobramycin particles with demonstrated flowability devices (Rotahaler® and Aerolizer®) with MgSt, to reduce drug reten-
and aerosol performance in the TOBI® Podhaler® are produced by emul- tion [115]. For the Aerolizer®, coating of capsules and inhalers at the
sion spray drying via the PulmoSphere™ technology [106]. Aerosol per- MgSt concentration of 0.3 mg/mL, significantly reduced capsule and in-
formance of the PulmoSphere tobramycin formulations was consistent haler retentions by 79 and 69%, respectively, compared to the uncoated
in different temperatures (10–40 °C), relative humidities (10–65%) counterparts. The Rotahaler®, which has a lower dispersion efficiency
and airflow rates (40–85 L/min) [107]. Colistin powders with corrugat- than the Aerolizer®, also showed a substantial reduction in capsule
ed surfaces and high aerosol performance (FPFtotal (b5 μm) N 80% via (by 73%) and device retention (by 53%). For both inhalers, reduced
the Aerolizer at 100 L/min and a pressure drop of 4 kPa) were prepared capsule and device retentions translated to a significantly higher fine
by Zhou et al. using spray drying without adding any external excipient particle doses [115].
[108]. Co-spray drying has also been employed to produce wrinkled In another study, the surface of a newly designed CC-3D resin inhaler
particles of a combination powder containing colistin and rifampicin and HPMC capsules were coated with a low surface energy polytetra-
(Fig. 4), with combined characteristics of synergistic antimicrobial ac- fluoroethylene suspension [131]. The capsule, capsule chamber and
tivity, high aerosol efficiency (FPFtotal (b5 μm) N 90% via the Aerolizer mouthpiece retention were significantly reduced from 14.1 to 7.3%,
at 100 L/min and a pressure drop of 4 kPa) and moisture protection 8.8 to 2.2% and 13.2 to 9.1%, respectively.
[109]. Dry coating of drug particle surface with magnesium stearate The above studies have shown the effectiveness of surface coating to
(MgSt) was also shown to be an effective approach to improve the reduce powder retention in capsule and inhaler devices. Addition of an-
flowability and aerosolisation of cohesive jet-milled drug powders tistatic additives into these materials may further reduce the adhesive
[110]. Nanoparticle delivery for aerosol formulation is also an emerging forces. The lowest pull-off forces between insulin particles and polymer
area [111]. It has been reviewed previously [112,113] and elsewhere in surfaces measured by atomic force microscopy were achieved for the
other articles of this special issue. material of polypropylene with addition of an antistatic additive (Dow

Fig. 4. Scanning electron micrographs of the spray dried antibiotics particles of: (a) colistin sulphate; and (b) combination of colistin sulphate and rifampicin (with a high emitted dose of
96% and high FPFtotal of 92%). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
Reprint from Ref. [109] with kind permission from Springer.

Please cite this article as: Q.(T.) Zhou, et al., Emerging inhalation aerosol devices and strategies: Where are we headed?, Adv. Drug Deliv. Rev.
(2014), http://dx.doi.org/10.1016/j.addr.2014.03.006
Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx 9

Entira™ AS) [132]. Future aerosolisation tests from the inhaler devices fosfomycin/amikacin (completed phase I for ventilated-associated
made of these materials are necessary to confirm the correlation be- pneumonia), fosfomycin/tobramycin (completed phase II for cystic fi-
tween the atomic force microscopy force measurement and device brosis patients), liposomal ciprofloxacin (Lipoquin™ which has com-
retention. pleted phase IIa for CF, Pulmaquin™ in phase III for non-cystic fibrosis
bronchiectasis), levofloxacin (Aeroquin™, completed phase III for CF).
5. Emerging inhalation therapies The DPI is now another viable option to deliver these high dose antibi-
otics. For example, Bayer is developing a DPI of ciprofloxacin (1 capsule,
5.1. Inhaled anti-pulmonary arterial hypertension agents twice daily) for CF, which is also currently in phase III for non-CF bron-
chiectasis. Additionally, dry powder vancomycin (AeroVanc™, 2 or 4
Nebulised iloprost is advantageous over the intravenous administra- capsules twice daily) is in phase IIa trial for CF.
tion of epoprostenol as it avoids the occurrence of tachyphylaxis and
prevents complications caused by the use of a central-venous route 5.2.1. Pneumonia
[133]. However, inhaled iloprost has a short half-life (approximately Oral and intravenous administered antibiotics commonly used in the
2 h) which means that patients suffering from pulmonary arterial hy- treatment of community-acquired and ventilated associated pneumo-
pertension have to administer the drug up to nine times a day. The com- nia, respectively. However, these routes could produce systemic side ef-
bination of inhaled iloprost and oral sildenafil was shown to be more fects since the drugs are distributed to various tissues via the blood
effective, and allowed reduction in the number of dose administrations circulation following the administration [143]. The systemic side effects
of iloprost alone, by prolonging the vasodilatory effect [133]. In a prom- can reduce patient adherence and therefore local pulmonary delivery is
ising study, treprostinil, which has a long plasma half-life than iloprost advantageous. Furthermore, pulmonary drug delivery will result in high
and is only administered up to a maximum of four times a day, was antibiotic concentrations in the lungs and hence minimise the bacterial
prepared as a pMDI formulation [134]. Doses of 1 mg/mL (15 μg resistance caused by insufficient local drug concentrations via systemic
treprostinil/puff) and 2 mg/mL (30 μ g treprostinil/puff) of treprostinil delivery systems.
sodium were prepared and 39 patients received 30, 45 or 60 μg of In a separate study, 400 mg of nebulised BAY41-6551 (Bayer
treprostinil. Patients who received 45 μg (3 puffs of 1 mg/mL Schering Pharma/Nektar Therapeutics) containing amikacin, was ad-
treprostinil) or 60 μg (2 puffs of 2 mg/mL treprostinil) showed pulmo- ministered every 12 h using a proprietary gasless vibrating-mesh
nary vasodilation surpassing the 120 min observation time which are nebuliser to the ventilated associated pneumonia patients. The patients
similar to the nebulised treprostinil (administered for 1 min). Further- showed positive results with the maximum amikacin concentration in
more, no side effects (e.g. nauseousness or dizziness) were observed, tracheal aspirate found to be 25 times higher than the MIC (256 μg/mL)
proving that administering treprostinil using a pMDI was well- in 50% of patients (6 out of 12 patients) [144]. In another study, amikacin
tolerated by the patients. concentration in epithelial lining fluid (which reflects the condition of the
Oral administration of nifedipine was shown to reduce pulmonary infection site more accurately than the amikacin concentration in the
arterial pressure [135,136] but no clinical study of inhaled nifedipine suctioned tracheal aspirates) was found to be four times higher than the
has been reported. Several reports have shown success in preparing MIC of 256 μg/mL threshold [145]. These studies indicate that local deliv-
spray-dried polymeric microparticles to control the release of sildenafil ery to the lung is more efficacious than systemic administrations. The
[137] and nifedipine [138], aiming to further reduce the dosing frequen- BAY41-6551 is currently in phase III trials for approval in ventilated
cy. However, the benefits of controlled-release characteristics of inhaled associated pneumonia patients.
nifedipine may need verification by clinical trials. By administering aminoglycosides locally to the lung, the risks
of neuro-, nephro- and ototoxicity, which can be induced when the
5.2. Inhaled antibiotics for respiratory bacterial infections drugs are administered through intravenous routes, can be minimised
[146]. Several in vitro works have reported successful preparation of
The TOBI® Podhaler® approved by FDA for cystic fibrosis treatment dry powder suitable for pulmonary delivery for treatment of pneumo-
may have higher treatment satisfaction contributing to ease of use and nia, for example, DNase1 [147], fosfomycin [148], colistin [91,108] and
treatment convenience [139]. TOBI® Podhaler® (Novartis AG, Basel, combination of ciprofloxacin hydrochloride plus gatifloxacin hydro-
Switzerland) delivers 112 mg tobramycin (from 4 capsules) as powder chloride [149], and colistin plus rifampicin [150].
aerosols twice a day. However, the repeat operation procedure “load,
pierce, inhale, check capsule if it has emptied properly and repeat 5.2.2. Tuberculosis
steps if not” for four capsules might be a tedious process. As shown above, inhaled antibiotics (particularly DPIs) have been
The Turbospin® delivers 125 mg colistimethate sodium used extensively and have shown great success in treating pulmonary
(Colobreathe® , Forest Laboratories, Inc.) from a single capsule, infections by increasing local concentration, improving patient adher-
twice a day. Although dose preparation time is reduced, multiple inha- ence and decrease systemic effects. This has opened a window of oppor-
lations may be necessary as a single inspiration may be insufficient to tunity to treat other infectious diseases, especially tuberculosis, where
empty the entire dose. With regard to operation convenience, multiple multidrug resistance is common due to poor treatment efficacy and pa-
inhalations of a single capsule have less repeated steps. However, when tient adherence [151].
a patient inhales too much powder from a single inhalation, side effects Three first line antibiotics for tuberculosis, pyrazinamide, rifampicin
such as coughing and chest tightness often occur [140]. A new high- and isoniazid, were successfully combined as a single DPI formulation
dose DPI Orbital®, which is able to deliver up to 400 mg of powder, with a FPFloaded (particlesb5.0 μm over the loaded dose) of 46 ± 3% pro-
has a mechanism to control the dose of each inhalation. The dose con- duced by spray drying [152]. Delivering these three antibiotics in a sin-
tainment chamber has an orifice that controls the release of the powder, gle administration may provide a more efficient and rapid approach to
making it possible for patient to inhale a set dose per inhalation over treat local and systemic infections, which could potentially translate to
multiple inhalations [141]. improvement in patient adherence. There are a number other studies
In a thorough review by Cipolla and Chan on inhaled antibiotics which have successfully prepared inhaled anti-tubercular formulations,
[142], most inhaled antibiotics have been prepared as nebulised formu- including a single powder formulation containing rifapentine,
lations — colistin (Colomycin® and Promixin®, approved in Europe for moxifloxacin, and pyrainzamide (Fig. 5) [151] and a capreomycinoleate
CF); aztreonam (Cayston®, FDA approved for CF), liposomal amikacin powder having high efficacy against Mycobacterium tuberculosis and
(ARIKACE™, phase III for CF in Europe, phase II for nontuberculosis low toxicity [153]. A recent novel approach to transport anti-tubercular
mycobacteria in USA, and completed phase II for non CF bronchiectasis), drugs into infected macrophages involves hydrolysed galactomannan-

Please cite this article as: Q.(T.) Zhou, et al., Emerging inhalation aerosol devices and strategies: Where are we headed?, Adv. Drug Deliv. Rev.
(2014), http://dx.doi.org/10.1016/j.addr.2014.03.006
10 Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

Fig. 5. Scanning electron micrographs of the spray dried particles containing rifapentine, moxifloxacin and pyrazinamide, (a) with and (b) without leucine. The powder formulation with
and without leucine had similar median aerodynamic diameters of 2.58 ± 0.08 μm and 2.51 ± 0.06 μm, with a relatively high FPF of 55.5 ± 1.9% and 63.6 ± 2.0%, respectively.
Reprint from ref. [151] with kind permission from Springer.

modified nanoparticles and flower-like polymeric micelles with high 5.4. Inhaled cyclosporine for transplant rejection
loading of rifampicin [154].
Cyclosporine A is a potent immunosuppressant drug used in long-
term to prevent allograft rejection in the lung transplant patients. How-
5.3. Inhaled neuraminidase inhibitor for influenza ever, oral and parenteral administrations induce substantial systemic
side effects which include renal toxicity and hypertension [162]. Provid-
Laninamivir octanoate is the newest neuraminidase inhibitor for in- ing high therapeutic concentration of cyclosporine A locally to the lungs
fluenza treatment administrated by a DPI following the use of may reduce the drug dose and hence lower the incidence of the system-
oseltamivir (Tamiflu®, administered orally), zanamivir (Relenza®, by in- ic side effects. In a single centre, double-blinded, placebo-controlled trial
halation) and peramivir (Rapiacta®, by single or multiple intravenous comparing the nebulised cyclosporine A in propylene glycol to the aero-
administrations). Although the efficacy of oseltamivir decreased against sol placebo (three times a week for two years), the incidence of bronchi-
the oseltamivir-resistant H1N1/H275Y virus, it was still predominantly olitis obliterans syndrome in the patients receiving cyclosporine A was
prescribed by physicians during the 2008–2009 flu season in Japan reduced and positive improvements in pulmonary lung functions
over zanamivir because of the simple oral administration proce- were observed [163]. Unfortunately, the multi-centre phase III trial
dure [155]. In a survey (conducted in Okinawa during the 2010 flu sea- (CYCLIST) showed no significant difference in delaying or preventing
son) comparing the ease of administration between oseltamivir, the occurrence of bronchiolitis obliterans syndrome in lung transplanted
zanamivir and laninamivir, the administration of zanamivir using patients when they received nebulised cyclosporine A formulation
Diskhaler® was found to be the most difficult [156]. Laninamivir in propylene glycol (62.5 mg/mL cyclosporine A in propylene
(Inavir®) has been approved and prescribed more often than glycol) [164].
oseltamivir and zanamivir in Japan since 2010 [156]. Biota Pharmaceu- Administration of nebulised cyclosporine A to these patients was
ticals is currently conducting a multinational phase II clinical trials of found to be challenging and claimed to influence the study outcome.
laninamivir in USA [157] which likely to be followed by approval soon. Various works have produced cyclosporine A formulations without
As a long-acting neuraminidase inhibitor, laninamivir remains in the the inclusion of irritants such as ethanol and propylene glycol, which
lungs for more than 5 days. Following a single dose of laninamivir, the are commonly used solvents to increase the dissolution of hydrophobic
median duration for fever and flu symptoms to subside was three and cyclosporine A. Nebulised cyclosporine A encapsulated in liposomes
four days (including days of administration), respectively, in patients [165], as well as dry powders prepared by dry-emulsion [166], anti-
having type A and B influenza. Thus only a single dose is necessary to solvent precipitation [167] and spray drying [168,169] have successfully
complete the treatment [157]. In contrast, oseltamivir and zanamivir been prepared. By removing the unsaturated fatty acids in the surfac-
have to be administered twice daily for five days [155]. Absolute ad- tant of glycerol monooleate, the dissolution of cyclosporine A formed
herence in these five-day therapies has to be ensured for complete clear- by dry-emulsion was significantly enhanced and stable micelles (ap-
ance of the virus. However, patients tend to stop taking the medications proximately 320 nm) were formed when resuspended in water [166].
once the flu symptoms have improved [157]. Thus the convenience of sin- With the absence of the unsaturated fatty acids, its oxidation to toxic
gle dose administration by laninamivir should considerable increase pa- lipoperoxide during storage was avoided and greater chemical stability
tient adherence. was achieved. Animal studies showed that the powder could potentially
Detailed information of the TwinCaps® device, which is used for pul- be used to treat respiratory inflammation.
monary delivery of laninamivir, is described in Section 4.3.2. Two dosing The use of lung-surfactant mimic phospholipids 1,2-dipalmitoyl-
chambers, each contains 10 mg of laninamivir octanoate powder, are sn-glycero-3-phosphocholine and 1,2-dipalmitoyl-sn-glycero-3-
present in each TwinCaps® inhaler (Hovione FarmaCiencia, SA). Chil- (phosphor-rac-1-glycerol) in cyclosporine A powder was found to in-
dren (b10 years old) are required to inhale from one device (total crease the powder dispersability, with the FPFTotal (b4.46 μm) of approx-
dose of 20 mg) and adults from two devices (total dose of 40 mg). The imately 50% measured by the next generation impactor at 60 L/min [168].
single administration of laninamivir is commonly done in front of the The bioavailability of hydrophobic cyclosporine A was increased by the in-
health care professionals to ensure correct inhalation technique. clusion of mannitol [167]. Porous microparticles (tap density of ≤about
Laninamivir has proven to be effective to the H1N1/H275Y virus and 0.3 g/cm3), suitable for nasal and pulmonary inhalation, containing an
shown similar clinical efficacy to oseltamivir and zanamivir against immunosuppressant drug (e.g. cyclosporine A, everolimus, tacrolimus,
H1N1, H3N2 and influenza B viruses [158,159]. Alleviation time for chil- sirolimus and pimecrolimus) and excipient (e.g. saccharide, amino acid,
dren was shorter with laninamivir compared to oseltamivir, whilst in a sugar alcohol) without the use of surfactant have been reported [169].
adults the alleviation time is similar [160]. Animal studies have further The porous particles were prepared using spray drying and a blowing
shown the superior efficacy of laninamivir against the highly pathogenic agent such as ammonium carbonate and ammonium bicarbonate. Ad-
avian flu virus H5N1 [161]. ministration of cyclosporine A using DPIs offers benefits which include

Please cite this article as: Q.(T.) Zhou, et al., Emerging inhalation aerosol devices and strategies: Where are we headed?, Adv. Drug Deliv. Rev.
(2014), http://dx.doi.org/10.1016/j.addr.2014.03.006
Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx 11

higher dose delivery with better control of aerosol properties and stability No adverse effects were found in terms of lung function and inflamma-
[170]. Active DPIs, instead of passive DPIs, may be more suitable for these tion in asthmatic patients. Subsequently, a Phase II multi-centre,
patients and can increase adherence since they often have limited lung randomised, double-blind, placebo-controlled trial was conducted on
function after the transplantation. 147 adult asthmatic patients, who were taking inhaled corticosteroids
and had a history of worsening symptoms after contracting a common
5.5. Inhaled alpha 1 antitrypsin cold [178]. The patients received either inhaled IFN-β or placebo for
14 days when cold symptoms appeared. Outcomes of this clinical trials
The current administration of alpha 1 antitrypsin (A1AT) in patients indicate that the severity of asthma symptoms were significantly re-
is by intravenous infusion (60 mg/kg per dose [171]). Although inhala- duced in patients who received inhaled IFN-β (patients on treatment
tion of A1AT has been shown to restore the protease–antiprotease im- had a 65% reduction in moderate exacerbations). These patients also
balance and anti-elastase activity in A1AT deficiency and CF patients demonstrated improved lung function as indicated by the steady in-
[172], no approved products are commercially available. When crease of morning peak expiratory flow. A constant increase of a bio-
radiolabelled A1AT (Prolastin®; α1-proteinase inhibitor (human); marker indicator for anti-viral activity was observed for the IFN-β
TalecrisBiotherapeutics Inc., Research Triangle Park, NC, USA) was patients group, whilst a rapid decrease was observed in patients treated
nebulised with AKITA2® APIXNEB® (Activaero GmbH, Gemünden, with placebo. Due to the positive outcome of IFN-β, a Phase II proof-of-
Germany) to the CF and A1AT deficiency patients, high lung deposition concept trial in COPD patients [179] and the preclinical development of
(≥ 70% of loaded drugs) and high peripheral deposition (N 40%) were IFN-β for patients with other severe viral lung infection including pneu-
achieved [173]. The lung and peripheral depositions were found to be monia [180] are currently underway.
similar in the healthy patients, which showed that this nebuliser per- Interferon-γ (IFN-γ) is an immune-regulatory cytokine responsible
formed similarly in patients despite the disease severity. Furthermore, for the fibroblast proliferation and therefore has attracted interest for
AKITA2® APIXNEB® nebuliser was well received by patients. The rela- treatment of idiopathic pulmonary fibrosis [181]. Subcutaneous injec-
tively short time (7–13 min to administer 100 mg of A1AT) and ease tion of IFN-γ to 826 patients with idiopathic pulmonary fibrosis failed
of administration were beneficial to improve patient acceptance. Anoth- to show a significant benefit in a randomised, double-blinded,
er smart nebuliser system, I-Neb® Adaptive Aerosol Delivery (I-Neb® placebo-controlled trial [182]. However, it was suggested that local de-
AAD, Philips Respironics, Parsippany, NJ) described in Section 2 has livery of IFN-γ to the epithelial lining of lung might be more efficacious
shown high lung depositions in a relatively short administration time [181]. Aerosol IFN-γ delivered by I-Neb® Adaptive Aerosol Delivery over
to deliver 25 mg A1AT: 56.6% in 7.5 min (Tidal Breathing Mode) and 80 weeks (100 μg, 3 times/week) to 10 patients with idiopathic pulmo-
59.9% in 4.4 min (Target Inhalation Mode with inspiratory time set for nary fibrosis showed efficient in vivo delivery to the lungs, with 65.4 ±
6 s) [174]. When the inspiratory time was increased to 9 s, 64.5% of 4.8% deposited in the parenchyma section determined by the scintigra-
the total dose was successfully deposited in the lungs in 2.5 min. This phy method [16]. In this study, inhaled IFN-γ was well-tolerated with
showed that slow and deep inspiration by patients can significantly re- no significant systemic side effects and patient adherence was as high
duce the administration time using the I-Neb® AAD system. This direct as 96.7 ± 4.81%. Further studies on inhaled IFN-γ for idiopathic pulmo-
administration of A1AT to the lungs will be more effective and reduce nary fibrosis are anticipated.
the systematic effect than the intravenous administration. Additionally, Inhaled IFN-γ has also been investigated for the treatment of pulmo-
the low residual volume in the AKITA2® APIXNEB® and I-Neb® AAD sys- nary tuberculosis [183]. However, the verdict on treatment efficacy
tems means a more cost effective treatment compared to the intrave- from various studies is still inconclusive [184]. In an early open-label
nous administration due to the high cost of A1AT. clinical trial, 5 tuberculosis patients received aerosol interferon-γ
500 μg 3 times a week for 1 month. Results were promising, with spu-
5.6. Inhaled cytokines tum smears returning negative [185]. However, these positive results
were not reproduced in the two subsequent clinical studies [186,187].
Inhaled human granulocyte macrophage colony stimulating factor Another recent randomised, controlled 4-month trial of adjunctive in-
(GM-CSF) is a promising therapy for pulmonary alveolar proteinosis haled IFN-γ only showed a transient beneficial effect at the 4-week
[175]. This rare lung disease is associated with high levels of neutralising timepoint [188]. Therefore, more rigorous clinical studies are necessary
antibodies against GM-CSF, causing accumulation of lipoproteinaceous to establish any potential clinical benefits of inhaled IFN-γ therapy
material in the alveoli and consequent deterioration in gas exchange against pulmonary tuberculosis.
[176]. Standard treatment involves whole-lung lavage, an invasive pro- Inhaled Interleukin-2 alone or as combination therapies have
cedure that is repeated every 6–12 months [176]. Subcutaneous admin- attracted interest for the treatment of pulmonary metastases of renal
istration of GM-CSF has been reported as a safe and effective treatment. cell carcinoma in the last two decades [189]. Previous clinical studies
However, exceptionally high incidences (in up to 85% of patients) of have shown acceptable patient tolerance and proven efficacy, the
systemic side effects such as rash, fever and bone pain have been report- results of which have been reviewed previously [189,190]. A recent
ed [177]. Aerosolised GM-CSF is proposed to mitigate these systemic ad- clinical study in 20 patients has reported inhalation of low-dose
verse effects. In a single-patient case study, a patient received nebulised interleukin-2 (3 × 3 million IU once daily) in conjunction with monthly
GM-CSF (sargramostim) treatment when marked deterioration in gas dacarbazine bolus injections offered an effective and safe treatment
exchange was observed after two lavages. A positive clinical response option for lung metastases in melanoma. In the study, the treatment
and improvements in gas exchange were exhibited with no adverse ef- (inhaled interleukin-2 plus dacarbazine) exhibited clinical efficacy in 9
fects after 1 year of treatment [176]. A multicentre, self-controlled phase patients with partial regression (27%) or stable disease (33%). In the
II trial conducted in Japan also found that nebulised GM-CSF (leukine) prophylaxis group, newly developed lung metastases were not ob-
therapy was safe and effective in treating pulmonary alveolar pro- served during the inhaled interleukin-2 therapy [191]. These promising
teinosis [175]. Additionally, the treatment provided a sustainable thera- results demonstrate the potential of inhaled cytokines for lung cancer-
peutic effect, with more than 80% of the subjects remaining stable related treatments.
without the need for further therapy for more than a year. Therefore
inhaled GM-CSF may be a viable therapeutic alternative to the current 5.7. Inhaled antibodies
invasive whole-lung lavage treatment.
Inhaled interferon beta (IFN-β) has been investigated for treatment Monoclonal antibody treatments may be effective in the manage-
of asthmatic symptoms, COPD and infectious diseases. A Phase I study, ment and control of severe asthma [192]. However, clinical reports on
completed in 2009, demonstrated that inhaled IFN-β was well tolerated. inhaled therapy of monoclonal antibodies are scarce. An early clinical

Please cite this article as: Q.(T.) Zhou, et al., Emerging inhalation aerosol devices and strategies: Where are we headed?, Adv. Drug Deliv. Rev.
(2014), http://dx.doi.org/10.1016/j.addr.2014.03.006
12 Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

study examined the attenuation efficacy of inhaled recombinant [8] V. Arunthari, R.S. Bruinsma, A.S. Lee, M.M. Johnson, A prospective, comparative trial
of standard and breath-actuated nebulizer: efficacy, safety, and satisfaction, Respir.
humanised monoclonal antibody-E25 on the airway response to the al- Care 57 (2012) 1242–1247.
lergen in asthmatic patients [193]. In this randomised, double-blind, [9] K. Leung, E. Louca, A.L. Coates, Comparison of breath-enhanced to breath-actuated
three-group study in 33 subjects with mild allergic asthma, no signifi- nebulizers for rate, consistency, and efficiency, Chest 126 (2004) 1619–1627.
[10] J.L. Rau, A. Ari, R.D. Restrepo, Performance comparison of nebulizer designs:
cant efficacy was found for the inhaled therapy compared to the placebo constant-output, breath-enhanced, and dosimetric, Respir. Care 49 (2004)
groups. Recently, GSK has completed Phase I clinical trials on safety, tol- 174–179.
erability, pharmacokinetics and pharmacodynamics of an inhaled fully [11] P. Kroneberg, W. Zimlich, B. Muellinger, A. Wenker, G. Scheuch, Device output is
not a predictor of lung dose, Drug Delivery to the Lung 19, Edinbugh, United
human, single domain antibody directed against the TNFR1 receptor Kingdom, 2008.
(GSK1995057) for the treatment of respiratory disorders. The results [12] J. Fleming, J. Conway, C. Majoral, L. Tossici-Bolt, I. Katz, G. Caillibotte, D. Perchet, M.
showed significantly reduced pulmonary and systemic markers of in- Pichelin, B. Muellinger, T. Martonen, P. Kroneberg, G. Apiou-Sbirlea, The use of
combined single photon emission computed tomography and X-ray computed to-
flammation and LPS-induced clinical symptoms (i.e. fever, nausea) for
mography to assess the fate of inhaled aerosol, J. Aerosol Med. Pulm. Drug Deliv. 24
the GSK1995057 treatment group compared to placebo group, with (2011) 49–60.
no observed severe side events [194]. Further clinical studies are neces- [13] E.M. Bakker, S. Volpi, E. Salonini, E.C. van der Wiel-Kooij, C.J.J.C.M. Sintnicolaas, W.
sary to elucidate the future role of monoclonal antibodies in pulmonary C.J. Hop, B.M. Assael, P.J.F.M. Merkus, H.A.W.M. Tiddens, Improved treatment re-
sponse to dornase alfa in cystic fibrosis patients using controlled inhalation, Eur.
diseases. Respir. J. 38 (2011) 1328–1335.
[14] W. Moller, I. Heimbeck, T.P. Hofer, G. Khadem Saba, M. Neiswirth, M.
Frankenberger, L. Ziegler-Heitbrock, Differential inflammatory response to inhaled
6. Conclusions lipopolysaccharide targeted either to the airways or the alveoli in man, PLoS One 7
(2012) e33505.
Inhaled pharmaceuticals have seen a number of innovations as they [15] K.C. Kesser, D.E. Geller, New aerosol delivery devices for cystic fibrosis, Respir. Care
54 (2009) 754–768.
evolve to accommodate novel medications and improve the efficacy of [16] K.T. Diaz, S. Skaria, K. Harris, M. Solomita, S. Lau, K. Bauer, G.C. Smaldone, R. Condos,
existing treatments. In particular, targeted treatment and vaccination Delivery and safety of inhaled interferon-gamma in idiopathic pulmonary fibrosis,
of pulmonary infectious diseases are a growing area of interest, and in J. Aerosol Med. Pulm. Drug Deliv. 25 (2012) 79–87.
[17] A.M. Martischnig, A. Tichy, M. Nikfardjam, G. Heinz, I.M. Lang, D. Bonderman, In-
turn the high-dose and disposable dry powder inhalers become increas- haled iloprost for patients with precapillary pulmonary hypertension and right-
ingly popular. pMDIs continue to encounter formulation challenges, al- side heart failure, J. Card. Fail. 17 (2011) 813–818.
though device improvements, such as the relatively discrete electronic [18] N. Goodman, M. Morgan, K. Nikander, S. Hinch, S. Coughlin, Evaluation of patient-
reported outcomes and quality of life with the I-neb AAD system in patients with
dose counters, are more apparent. Meanwhile, software control units
chronic obstructive pulmonary disease, J. Aerosol Med. Pulm. Drug Deliv. 23
for nebulisers have advanced treatment shortening and dosing accuracy (Suppl. 1) (2010) S61–S70.
to a new level, and have appealing implications for enhancing clinical [19] K. Nikander, J. Denyer, M. Dodd, T. Dyche, K. Webb, P. Weller, D. Stableforth, The
Adaptive Aerosol Delivery system in a telehealth setting: patient acceptance, per-
trials. Whilst the DPI, pMDI and nebulizer fields are nearing maturity,
formance and feasibility, J. Aerosol Med. Pulm. Drug Deliv. 23 (2010) S21–S27.
computational modelling of aerosol kinetics is a budding area. As the [20] S.W. Stein, P. Sheth, P.D. Hodson, P.B. Myrdal, Advances in metered dose inhaler
cost of computing power reduces, new approaches such as CFD–DEM technology: hardware development, AAPS PharmSciTech (2013) 1–13.
promise to optimise device-formulation coupling, and will become inte- [21] A.S. Melani, M. Bonavia, V. Cilenti, C. Cinti, M. Lodi, P. Martucci, M. Serra, N. Scichilone,
P. Sestini, M. Aliani, M. Neri, Inhaler mishandling remains common in real life and is
gral in the future. Indeed, it appears that the explosive growth of global associated with reduced disease control, Respir. Med. 105 (2012) 930–938.
digital technology has finally caught up with inhaled therapeutics. We [22] J.B. Fink, B.K. Rubin, Problems with inhaler use: a call for improved clinician and
anticipate digital technology will define the next era of novel inhalation patient education, Respir. Care 50 (2005) 1360–1375.
[23] G. Gross, R.M. Cohen, H. Guy, Efficacy response of inhaled HFA-albuterol delivered
devices and formulations, as well as delving into less traditional roles in- via the breath-actuated Autohaler inhalation device is comparable to dose in pa-
volving improved patient interaction and adherence. Patient adherence tients with asthma, J. Asthma 40 (2003) 487–495.
and treatment outcomes might also be improved by the application of [24] H. Worth, J.F. Muir, W.R. Pieters, Comparison of hydrofluoroalkane-beclomethasone
dipropionate autohaler (TM) with budesonide turbuhaler (TM) in asthma control,
aerosol delivery to an increasing variety of pharmaceuticals. Respiration 68 (2001) 517–526.
[25] K.F.M. Zuyderhoudt, Inhalers and Housing Caps for Inhalers, 2012. (PCT/EP2011/
006000).
Acknowledgement [26] W. Azouz, J. Campbell, J. Stephenson, D. Saralaya, H. Chrystyn, Improved metered
dose inhaler technique when a coordination cap is used, J. Aerosol Med. Pulm.
The authors acknowledge the financial support from the Australian Drug Deliv. (2013), http://dx.doi.org/10.1089/jamp.2012.1031 (ahead of print).
[27] C.L. Leach, G.L. Colice, A pilot study to assess lung deposition of HFA-
Research Council's Discovery Projects funding scheme (DP120102778
beclomethasone and CFC-beclomethasone from a pressurized metered dose inhal-
& DP110105161) and the National Health and Medical Research er with and without add-on spacers and using varying breathhold times, J. Aerosol
Council's (NHMRC) Project Grant funding scheme (APP1065046). Qi Med. Pulm. Drug Deliv. 23 (2010) 355–361.
Tony Zhou is a recipient of Early Career Fellowship from NHMRC [28] L.E. Mansfield, R. Maynes, A valved holding chamber does not decrease the bron-
chodilator activity of budesonide-formoterol combination metered dose inhaler,
(APP1053528). John Chan is the recipient of the Australian Postgraduate J. Asthma 50 (2013) 71–74.
Award from the Australian Federal Government. [29] C. Crim, M. Holmes, B. Lee, R. Cavanaugh, W. Lincourt, Evaluation of particle size
distribution of albuterol sulfate hydrofluoroalkane administered via metered-
dose inhaler with and without valved holding chambers, Ann. Allergy Asthma
References Immunol. 94 (2005) 80–85.
[30] M.W. Nagel, K.J. Wiersema, S.L. Bates, J.P. Mitchell, Performance of large- and
[1] M. Knoch, M. Keller, The customised electronic nebuliser: A new category of liquid small-volume valved holding chambers with a new combination long-term
aerosol drug delivery systems, Expert Opin. Drug Deliv. 2 (2005) 377–390. bronchodilator/anti-inflammatory formulation delivered by pressurized metered
[2] A. Fischer, J. Stegemann, G. Scheuch, R. Siekmeier, Novel devices for individualized dose inhaler, J. Aerosol Med. 15 (2002) 427–433.
controlled inhalation can optimize aerosol therapy in efficacy, patient care and [31] W.H.O. Ten Have, N.J.V. De Berg, P.J.E. Bindels, W.M.C. Van Aalderen, J. Van Der
power of clinical trials, Eur. J. Med. Res. 14 (2009) 71–77. Palen, Assessment of inhalation technique in children in general practice: in-
[3] T. Daniels, N. Mills, P. Whitaker, Nebuliser systems for drug delivery in cystic fibro- creased risk of incorrect performance with new device, J. Asthma 45 (2008) 67–71.
sis, Cochrane Database Syst. Rev. 4 (2013) CD007639. [32] S.P. Newman, Principles of metered-dose inhaler design, Respir. Care 50 (2005)
[4] B.K. Rubin, Pediatric aerosol therapy: new devices and new drugs, Respir. Care 56 1177–1190.
(2011) 1411–1423. [33] R.F. Oliveira, S.F.C.F. Teixeira, L.F. Silva, J.C.F. Teixeira, H. Antunes, Development of
[5] R. Dalby, M. Spallek, T. Voshaar, A review of the development of Respimat® Soft new spacer device geometry: a CFD study (part I), Comput. Methods Biomech.
Mist™ Inhaler, Int. J. Pharm. 283 (2004) 1–9. Biomed. Eng. 15 (2012) 825–833.
[6] D. Cipolla, H.-K. Chan, J. Schuster, D. Farina, Personalizing aerosol medicine: devel- [34] T.M. Goncalves, K. Alhanout, A. Nicolay, J.C. Dubus, D.P. de Santana, V. Andrieu,
opment of delivery systems tailored to the individual, Ther. Deliv. 1 (2010) Comparative in vitro performance of three small-volume valved holding chambers
667–682. with beclomethasone/formoterol pressurized metered dose inhaler, J. Aerosol
[7] A.B. Watts, J.T. McConville, R.O. Williams, Current therapies and technological Med. Pulm. Drug Deliv. 26 (2013) 223–227.
advances in aqueous aerosol drug delivery, Drug Dev. Ind. Pharm. 34 (2008) [35] US Food and Drug Administration, Guidance for Industry. Integration of Dose-
913–922. counting Mechanisms into MDI Drug Products, 2003.

Please cite this article as: Q.(T.) Zhou, et al., Emerging inhalation aerosol devices and strategies: Where are we headed?, Adv. Drug Deliv. Rev.
(2014), http://dx.doi.org/10.1016/j.addr.2014.03.006
Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx 13

[36] C. Weinstein, H. Staudinger, I. Scott, N.J. Amar, C. LaForce, Dose counter perfor- [66] T. Ehtezazi, D.R. Allanson, I.D. Jenkinson, I. Shrubb, C. O'Callaghan, Investigating im-
mance of mometasone furoate/formoterol inhalers in subjects with asthma or proving powder deagglomeration via dry powder inhalers at a low inspiratory
COPD, Respir. Med. 105 (2011) 979–988. flow rate by employing add-on spacers, J. Pharm. Sci. 97 (2008) 5212–5221.
[37] R.L. Wasserman, K. Sheth, W.R. Lincourt, N.W. Locantore, J.C. Rosenzweig, C. Crim, [67] M.S. Coates, D.F. Fletcher, H.K. Chan, J.A. Raper, Effect of design on the performance
Real-world assessment of a metered-dose inhaler with integrated dose counter, of a dry powder inhaler using computational fluid dynamics. Part 1: grid structure
Allergy Asthma Proc. 27 (2006) 486–492. and mouthpiece length, J. Pharm. Sci. 93 (2004) 2863–2876.
[38] P. Allsop, M.C. Bainton, S.A. Day, C. Dickens, D.J. Glenton, S.G.D. Kelly, R. Perkins, R.J. [68] Q.T. Zhou, Z. Tong, P. Tang, M. Citterio, R. Yang, H.K. Chan, Effect of device design on
Warby, Metered Dose Inhaler Comprising a Dose Counter, 2009. (WO2009022139). the aerosolization of a carrier-based dry powder inhaler—a case study on Aerolizer
[39] M. Patel, J. Pilcher, A. Chan, K. Perrin, P. Black, R. Beasley, Six-month in vitro valida- Foradile, AAPS J. 15 (2013) 511–522.
tion of a metered-dose inhaler electronic monitoring device: implications for asth- [69] P.W. Longest, Y.J. Son, L. Holbrook, M. Hindle, Aerodynamic factors responsible for
ma clinical trial use, J. Allergy Clin. Immunol. 130 (2012) 1420–1422. the deaggregation of carrier-free drug powders to form micrometer and
[40] J.B. Conner, P.O. Buck, Improving asthma management: the case for mandatory in- submicrometer aerosols, Pharm. Res. 30 (2013) 1608–1627.
clusion of dose counters on all rescue bronchodilators, J. Asthma 50 (2013) [70] L.I. Harrison, C.C. Novak, M.J. Needham, P. Ratner, Comparative pulmonary function
658–663. and pharmacokinetics of fluticasone propionate and salmeterol xinafoate delivered
[41] P.J. Barnes, Scientific rationale for using a single inhaler for asthma control, Eur. by two dry powder inhalers to patients with asthma, J. Aerosol Med. Pulm. Drug
Respir. J. 29 (2007) 587–595. Deliv. 24 (2011) 245–252.
[42] A. Bodzenta-Lukaszyk, G. Pulka, A. Dymek, D. Bumbacea, T. McIver, B. Schwab, H. [71] P. Selvam, D. McNair, R. Truman, H.D.C. Smyth, A novel dry powder inhaler: effect
Mansikka, Efficacy and safety of fluticasone and formoterol in a single pressurized of device design on dispersion performance, Int. J. Pharm. 401 (2010) 1–6.
metered dose inhaler, Respir. Med. 105 (2011) 674–682. [72] C. Friebel, S. Stolte, H. Steckel, I. Solomon, Performance of a new single-use dispos-
[43] D.A. Mahler, A. D'Urzo, E.D. Bateman, S.A. Ozkan, T. White, C. Peckitt, C. Lassen, B. able dry powder inhaler prototype, Respiratory, Drug Delivery, 2010, pp. 517–522.
Kramer, Concurrent use of indacaterol plus tiotropium in patients with COPD pro- [73] L. Wagenseil, A.K. Menge, I. Solomon, H. Steckel, Optimization and performance of
vides superior bronchodilation compared with tiotropium alone: a randomised, the ResQhaler — a single-use disposable dry powder inhaler, J. Aerosol Med. Pulm.
double-blind comparison, Thorax 67 (2012) 781–788. Drug Deliv. 26 (2013) A240.
[44] D.P. Tashkin, S.T. Varghese, Combined treatment with formoterol and tiotropium is [74] X. Zhang, Y. Ma, L. Zhang, J. Zhu, F. Jin, The development of a novel dry powder in-
more efficacious than treatment with tiotropium alone in patients with chronic haler, Int. J. Pharm. 431 (2012) 45–52.
obstructive pulmonary disease, regardless of smoking status, inhaled corticoste- [75] T.E. Corcoran, R. Venkataramanan, R.M. Hoffman, M.P. George, A. Petrov, T.
roid use, baseline severity, or gender, Pulm. Pharmacol. Ther. 24 (2011) 147–152. Richards, S. Zhang, J. Choi, Y.Y. Gao, C.D. Oakum, R.O. Cook, M. Donahoe, Systemic
[45] D. Lechuga-Ballesteros, B. Noga, R. Vehring, R.H. Cummings, S.K. Dwivedi, Novel delivery of atropine sulfate by the MicroDose dry-powder inhaler, J. Aerosol Med.
cosuspension metered-dose inhalers for the combination therapy of chronic ob- Pulm. Drug Deliv. 26 (2013) 46–55.
structive pulmonary disease and asthma, Future Med. Chem. 3 (2011) 1703–1718. [76] J.D. Rabinowitz, M. Wensley, P. Lloyd, D. Myers, W. Shen, A. Lu, C. Hodges, R. Hale,
[46] P.J. Barnes, Triple inhalers for obstructive airways disease: will they be useful? Ex- D. Mufson, A. Zaffaroni, Fast onset medications through thermally generated aero-
pert Rev. Respir. Med. 5 (2011) 297–300. sols, J. Pharmacol. Exp. Ther. 309 (2004) 769–775.
[47] A. Chatterjee, M. Shah, A.O. D'Souza, B. Bechtel, G. Crater, A.A. Dalal, Observational [77] K. Dinh, D.J. Myers, M. Glazer, T. Shmidt, C. Devereaux, K. Simis, P.D. Noymer, M.
study on the impact of initiating tiotropium alone versus tiotropium with He, C. Choosakul, Q. Chen, J.V. Cassella, In vitro aerosol characterization of Staccato®
fluticasone propionate/salmeterol combination therapy on outcomes and costs in Loxapine, Int. J. Pharm. 403 (2011) 101–108.
chronic obstructive pulmonary disease, Respir. Res. 13 (2012) 15. [78] J.D. Rabinowitz, P.M. Lloyd, P. Munzar, D.J. Myers, S. Cross, R. Damani, R. Quintana,
[48] G. Brambilla, D. Howlett, R. Johnson, D.A. Lewis, Metered-dose Inhaler and Method D.A. Spyker, P. Soni, J.V. Cassella, Ultra-fast absorption of amorphous pure drug
of Using the Same, 2012. (WO 2012032010 A1). aerosols via deep lung inhalation, J. Pharm. Sci. 95 (2006) 2438–2451.
[49] D. Dohmeier, D. Heyworth, T. Wilde, The application of a new high performance [79] L. Borgstrom, B. Olsson, L. Thorsson, Degree of throat deposition can explain the
dual-layer coating to pressurized metered dose inhaler hardware, Respir Drug variability in lung deposition of inhaled drugs, J. Aerosol Med. 19 (2006) 473–483.
Deliv Eur, 2009, pp. 209–212. [80] P. Tang, H.K. Chan, D. Rajbhandari, P. Phipps, Method to introduce mannitol pow-
[50] G. Fradley, D. Hodson, Optimization of fluid flow in pMDI valves, Respiratory, Drug der to intubated patients to improve sputum clearance, J. Aerosol Med. Pulm. Drug
Delivery, 2008, pp. 329–332. Deliv. 24 (2011) 1–9.
[51] P.G.A. Rogueda, R. Price, T. Smith, P.M. Young, D. Traini, Particle synergy and aero- [81] H.-K. Chan, D. Rajbhandari, P. Tang, J.D. Brannan, P. Phipps, Safety of administering dry
sol performance in non-aqueous liquid of two combinations metered dose inhala- powder mannitol to stimulate sputum clearance in intubated intensive care patients
tion formulations: an AFM and Raman investigation, J. Colloid Interface Sci. 361 with sputum retention: a pilot study, Am. J. Respir. Crit. Care Med. 185 (2012) A6809.
(2011) 649–655. [82] W. Wong, D.F. Fletcher, D. Traini, H.K. Chan, P.M. Young, The use of computational
[52] J.G. Weers, E.G. Schutt, L.A. Dellamary, T.E. Tarara, A. Kabalnov, Stabilized prepara- approaches in inhaler development, Adv. Drug Deliv. Rev. 64 (2012) 312–322.
tions for use in metered dose inhalers. US Patent 6,309,623., (2001). [83] C.A. Ruzycki, E. Javaheri, W.H. Finlay, The use of computational fluid dynamics in
[53] R. Vehring, D. Lechuga-Ballesteros, V. Joshi, B. Noga, S.K. Dwivedi, Cosuspensions of inhaler design, Expert Opin. Drug Deliv. 10 (2013) 307–323.
microcrystals and engineered microparticles for uniform and efficient delivery of [84] C. Kleinstreuer, H.W. Shi, Z. Zhang, Computational analyses of a pressurized
respiratory therapeutics from pressurized metered dose inhalers, Langmuir 28 metered dose inhaler and a new drug-aerosol targeting methodology, J. Aerosol
(2012) 15015–15023. Med. 20 (2007) 294–309.
[54] M. Miravitlles, A. Anzueto, Antibiotics for acute and chronic respiratory infection in [85] P.W. Longest, M. Hindle, Evaluation of the respimat soft mist inhaler using a concur-
patients with chronic obstructive pulmonary disease, Am. J. Respir. Crit. Care Med. rent CFD and in vitro approach, J. Aerosol Med. Pulm. Drug Deliv. 22 (2009) 99–112.
188 (2013) 1052–1057. [86] E.R. Arulmuthu, D.J. Williams, H. Baldascini, H.K. Versteeg, M. Hoare, Studies on
[55] R. Hudson, B.O. Blair, Inhaled antibiotics for gram-negative respiratory infections, aerosol delivery of plasmid DNA using a mesh nebulizer, Biotechnol. Bioeng. 98
Future Med. Chem. 3 (2011) 1663–1677. (2007) 939–955.
[56] S.W. Stein, B.R. Forsyth, J.S. Stefely, J.D. Christensen, T.D. Alband, P.A. Jinks, [87] S.-C. Shen, Y.-J. Wang, Y.-Y. Chen, Design and fabrication of medical micro-
Expanding the dosing range of metered dose inhalers through formulation and nebulizer, Sensors Actuators A Phys. 144 (2008) 135–143.
hardware optimization, Respiratory, Drug Delivery, 2004, pp. 125–134. [88] M.S. Coates, H.K. Chan, D.F. Fletcher, J.A. Raper, Influence of air flow on the perfor-
[57] W. Alston, A.R. Clark, C. Schuler, K.R. Walsh, Increased Dosage Metered Dose Inhal- mance of a dry powder inhaler using computational and experimental analyses,
er, 2004. (PCT/US2003/034473). Pharm. Res. 22 (2005) 1445–1453.
[58] L.A. Dellamary, T.E. Tarara, A. Kabalnov, J.G. Weers, E.G. Schutt, Stabilized Bioactive [89] M.S. Coates, H.-K. Chan, D.F. Fletcher, H. Chiou, Influence of mouthpiece geometry
Preparations and Methods of Use, 2002. (PCT/US1998/020613). on the aerosol delivery performance of a dry powder inhaler, Pharm. Res. 24
[59] B. Zhu, D. Traini, H.K. Chan, P.M. Young, The effect of ethanol on the formation and (2007) 1450–1456.
physico-chemical properties of particles generated from budesonide solution- [90] M.S. Coates, D.F. Fletcher, H.K. Chan, J.A. Raper, The role of capsule on the perfor-
based pressurized metered-dose inhalers, Drug Dev. Ind. Pharm. 39 (2013) mance of a dry powder inhaler using computational and experimental analyses,
1625–1637. Pharm. Res. 22 (2005) 923–932.
[60] A.H. de Boer, H.K. Chan, R. Price, A critical view on lactose-based drug formulation [91] A.H. de Boer, P. Hagedoorn, R. Woolhouse, E. Wynn, Computational fluid dynamics
and device studies for dry powder inhalation: which are relevant and what inter- (CFD) assisted performance evaluation of the Twincer disposable high-dose dry
actions to expect? Adv. Drug Deliv. Rev. 64 (2012) 257–274. powder inhaler, J. Pharm. Pharmacol. 64 (2012) 1316–1325.
[61] M.S. Coates, H.K. Chan, D.F. Fletcher, J.A. Raper, Effect of design on the performance [92] L.Q. Jiang, Y. Tang, H.J. Zhang, X.F. Lu, X.J. Chen, J.B. Zhu, Importance of powder
of a dry powder inhaler using computational fluid dynamics. Part 2: air inlet residence time for the aerosol delivery performance of a commercial dry powder
size, J. Pharm. Sci. 95 (2006) 1382–1392. inhaler Aerolizer, J. Aerosol Med. Pulm. Drug Deliv. 25 (2012) 265–279.
[62] A. Voss, W.H. Finlay, Deagglomeration of dry powder pharmaceutical aerosols, Int. [93] M.J. Donovan, S.H. Kim, V. Raman, H.D. Smyth, Dry powder inhaler device influence
J. Pharm. 248 (2002) 39–50. on carrier particle performance, J. Pharm. Sci. 101 (2012) 1097–1107.
[63] Z.B. Tong, B. Zheng, R.Y. Yang, A.B. Yu, H.K. Chan, CFD–DEM investigation of the dis- [94] J. Shur, S. Lee, W. Adams, R. Lionberger, J. Tibbatts, R. Price, Effect of device design
persion mechanisms in commercial dry powder inhalers, Powder Technol. 240 on the in vitro performance and comparability for capsule-based dry powder in-
(2013) 19–24. halers, AAPS J. 14 (2012) 667–676.
[64] Y.-J. Son, P.W. Longest, G. Tian, M. Hindle, Evaluation and modification of commer- [95] Z.B. Tong, R.Y. Yang, K.W. Chu, A.B. Yu, S. Adi, H.K. Chan, Numerical study of the ef-
cial dry powder inhalers for the aerosolization of a submicrometer excipient en- fects of particle size and polydispersity on the agglomerate dispersion in a cyclonic
hanced growth (EEG) formulation, Eur. J. Pharm. Sci. 49 (2013) 390–399. flow, Chem. Eng. J. 164 (2010) 432–441.
[65] R.M. Curtis, M.J. Donovan, H. Smyth, Inhaler to Deliver Substances for Prophy- [96] Z.B. Tong, S. Adi, R.Y. Yang, H.K. Chan, A.B. Yu, Numerical investigation of the de-
laxis or Prevention of Disease or Injury Caused by the Chemical Agents, 2013. agglomeration mechanisms of fine powders on mechanical impaction, J. Aerosol
(US2013/0213397 A1). Sci. 42 (2011) 811–819.

Please cite this article as: Q.(T.) Zhou, et al., Emerging inhalation aerosol devices and strategies: Where are we headed?, Adv. Drug Deliv. Rev.
(2014), http://dx.doi.org/10.1016/j.addr.2014.03.006
14 Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx

[97] P. Irene, B. Gaetano, Effect of the inhalation volume on drug delivery performance [128] M. Paajanen, J. Katainen, J. Raula, E.I. Kauppinen, J. Lahtinen, Direct evidence on re-
of NEXThaler®, in: Respiratory Drug Delivery Europe, 2013. 273–278. duced adhesion of Salbutamol sulphate particles due to L-leucine coating, Powder
[98] T. Voshaar, M. Spinola, P. Linnane, A. Campanini, D. Lock, A. Lafratta, M. Scuri, B. Technol. 192 (2009) 6–11.
Ronca, A. Melani, Comparing usability of NEXThaler® with other inhaled [129] T. Sou, L. Orlando, M.P. McIntosh, L.M. Kaminskas, D.A.V. Morton, Investigating the
corticosteroid/long-acting β2-agonist fixed combination dry powder inhalers in interactions of amino acid components on a mannitol-based spray-dried powder
asthma patients, J. Aerosol Med. Pulm. Drug Deliv. (2013), http://dx.doi.org/10. formulation for pulmonary delivery: a design of experiment approach, Int. J.
1089/jamp.2013.1086 (Ahead of print). Pharm. 421 (2011) 220–229.
[99] H. Blau, H. Mussaffi, M.M. Zahav, D. Prais, M. Livne, B.M. Czitron, H.A. Cohen, Micro- [130] Q. Zhou, J.A. Denman, T. Gengenbach, S. Das, L. Qu, H.L. Zhang, I. Larson, P.J. Stewart,
bial contamination of nebulizers in the home treatment of cystic fibrosis, Child D.A.V. Morton, Characterization of the surface properties of a model pharmaceuti-
Care Health Dev. 33 (2007) 491–495. cal fine powder modified with a pharmaceutical lubricant to improve flow via a
[100] H.A. Cohen, Z. Cohen, A.S. Pomeranz, B. Czitron, E. Kahan, Bacterial contamination mechanical dry coating approach, J. Pharm. Sci. 100 (2011) 3421–3430.
of spacer devices used by asthmatic children, J. Asthma 42 (2005) 169–172. [131] S.B. Behara, D. Farkas, M. Hindle, P.W. Longest, Development of a high efficiency
[101] T.W. De Vries, S.R. Rienstra, E.R. Van Der Vorm, Bacterial contamination of inhala- dry powder inhaler: effects of capsule chamber design and inhaler surface modifi-
tion chambers: results of a pilot study, J. Aerosol Med. 17 (2004) 354–356. cations, Pharm. Res. (2013) 1–13.
[102] F.A. Mabbott, A.B. Mullen, A. Smith, G. Smith, A.C. Boyter, Qualitative analysis of mi- [132] S.C. Strathmann, M.A. Murphy, B.A. Goeckner, P.W. Carter, J.-B.D. Green, Forces
crobial contamination of inhalers, J. Hosp. Infect. 77 (2011) 277–278. between insulin microspheres and polymers surfaces for a dry powder inhaler,
[103] A.H. de Boer, P. Hagedoorn, E.M. Westerman, P.P.H. Le Brun, H.G.M. Heijerman, H. Int. J. Pharm. 372 (2009) 147–153.
W. Frijlink, Design and in vitro performance testing of multiple air classifier tech- [133] M. Lopez-Meseguer, C. Berastegui, V. Monforte, C. Bravo, E. Domingo, A. Roman,
nology in a new disposable inhaler concept (Twincer®) for high powder doses, Inhaled Iloprost plus oral sildenafil in patients with severe pulmonary arterial
Eur. J. Pharm. Sci. 28 (2006) 171–178. hypertension delays the need for lung transplantation, Transplant. Proc. 45
[104] E.A. Nardell, Inhaled colistin: a novel approach for reducing drug-resistant TB (2013) 2347–2350.
transmission, 17th Annual Conference of the International Union Against Tubercu- [134] R. Voswinckel, F. Reichenberger, H. Gall, T. Schmehl, T. Gessler, R.T. Schermuly, F.
losis and Lung Disease — North American Region, Vancouver, BC, Canada, 2013. Grimminger, L.J. Rubin, W. Seeger, H.A. Ghofrani, H. Olschewski, Metered dose in-
[105] Q. Zhou, D. Morton, Drug-lactose binding aspects in adhesive mixtures: controlling haler delivery of treprostinil for the treatment of pulmonary hypertension, Pulm.
performance in dry powder inhaler formulations by altering lactose carrier sur- Pharmacol. Ther. 22 (2009) 50–56.
faces, Adv. Drug Deliv. Rev. 64 (2012) 275–284. [135] B.G. Phillips, J.L. Bauman, M.D. Schoen, T.J. Hoon, B.J. Schrader, S. Rich, Serum nifed-
[106] D.E. Geller, J. Weers, S. Heuerding, Development of an inhaled dry-powder formu- ipine concentrations and response of patients with pulmonary hypertension, Am. J.
lation of tobramycin using PulmoSphere (TM) technology, J. Aerosol Med. Pulm. Cardiol. 77 (1996) 996–999.
Drug Deliv. 24 (2011) 175–182. [136] A.S. Malik, S. Warshafsky, S. Lehrman, Meta-analysis of the long-term effect of
[107] A. Haynes, J. Nakamura, C. Heng, S. Heuerding, G. Thompson, R. Malcolmson, Aero- nifedipine for pulmonary hypertension, Arch. Intern. Med. 157 (1997) 621–625.
sol performance of tobramycin inhalation powder, Respiratory, Drug Delivery, [137] B.-B. Moritz, C. Schweiger, T. Schmehl, T. Gessler, W. Seeger, T. Kissel, Characterization
2010, pp. 701–706. of novel spray-dried polymeric particles for controlled pulmonary drug deliv-
[108] Q. Zhou, D. Morton, H.H. Yu, J. Jacob, J.P. Wang, J. Li, H.K. Chan, Colistin powders ery, J. Control. Release 158 (2012) 329–335.
with high aerosolisation efficiency for respiratory infection: preparation and [138] A. Saigal, W.K. Ng, R.B.H. Tan, S.Y. Chan, Development of controlled release
in vitro evaluation, J. Pharm. Sci. 102 (2013) 3736–3747. inhalable polymeric microspheres for treatment of pulmonary hypertension, Int.
[109] Q.T. Zhou, T. Gengenbach, J.A. Denman, H.H. Yu, J. Li, H.K. Chan, Synergistic antibi- J. Pharm. 450 (2013) 114–122.
otic combination powders of colistin and rifampicin provide high aerosolization ef- [139] M.W. Konstan, P.A. Flume, M. Kappler, R. Chiron, M. Higgins, F. Brockhaus, J. Zhang,
ficiency and moisture protection, AAPS J. 16 (2014) 37–47. G. Angyalosi, E. He, D.E. Geller, Safety, efficacy and convenience of tobramycin
[110] Q.T. Zhou, L. Qu, I. Larson, P.J. Stewart, D. Morton, Improving aerosolization of drug inhalation powder in cystic fibrosis patients: The EAGER trial, J. Cyst. Fibros. 10
powders by reducing powder intrinsic cohesion via a mechanical dry coating ap- (2011) 54–61.
proach, Int. J. Pharm. 394 (2010) 50–59. [140] M. Hoppentocht, P. Hagedoorn, H.W. Frijlink, A.H. de Boer, Developments and
[111] H.-K. Chan, Nanodrug particles and nanoformulations for drug delivery, Adv. Drug strategies for inhaled antibiotic drugs in tuberculosis therapy: a critical evaluation,
Deliv. Rev. 63 (2011) 405. Eur. J. Pharm. Biopharm. (2013), http://dx.doi.org/10.1016/j.ejpb.2013.1010.1019
[112] H.-K. Chan, P.C.L. Kwok, Production methods for nanodrug particles using the (e-pub).
bottom-up approach, Adv. Drug Deliv. Rev. 63 (2011) 406–416. [141] P.M. Young, J. Crapper, G. Philips, K. Sharma, H.-K. Chan, D. Traini, Overcoming dose
[113] J. Zhang, L. Wu, H.-K. Chan, W. Watanabe, Formation, characterization, and fate of limitations using the Orbital® multi-breath dry powder inhaler, J. Aerosol Med.
inhaled drug nanoparticles, Adv. Drug Deliv. Rev. 63 (2011) 441–455. Pulm. Drug Deliv. (2013), http://dx.doi.org/10.1089/jamp.2013.1080 (ahead of
[114] P.C.L. Kwok, A. Tunsirikongkon, W. Glover, H.K. Chan, Formation of protein nano- print).
matrix particles with controlled surface architecture for respiratory drug delivery, [142] D. Cipolla, H.-K. Chan, Inhaled antibiotics to treat lung infections, Pharm. Pat. Anal.
Pharm. Res. 28 (2011) 788–796. 2 (2013) 647–663.
[115] D. Heng, S.H. Lee, W.K. Ng, H.K. Chan, J.W. Kwek, R.B.H. Tan, Novel alternatives to [143] K. Togami, S. Chono, K. Morimoto, Aerosol-based efficient delivery of
reduce powder retention in the dry powder inhaler during aerosolization, Int. J. clarithromycin, a macrolide antimicrobial agent, to lung epithelial lining fluid
Pharm. 452 (2013) 194–200. and alveolar macrophages for treatment of respiratory infections, J. Aerosol Med.
[116] L. Tajber, O.I. Corrigan, A.M. Healy, Spray drying of budesonide, formoterol Pulm. Drug Deliv. 25 (2012) 110–115.
furnarate and their composites—II. Statistical factorial design and in vitro deposi- [144] M.S. Niederman, J. Chastre, K. Corkery, J.B. Fink, C.-E. Luyt, M.S. García, BAY41-6551
tion properties, Int. J. Pharm. 367 (2009) 86–96. achieves bactericidal tracheal aspirate amikacin concentrations in mechanically
[117] P.C.L. Kwok, H.K. Chan, Effect of relative humidity on the electrostatic charge prop- ventilated patients with Gram-negative pneumonia, Intensive Care Med. 38
erties of dry powder inhaler aerosols, Pharm. Res. 25 (2008) 277–288. (2012) 263–271.
[118] S. Karner, N. Anne Urbanetz, The impact of electrostatic charge in pharmaceutical [145] C.-E. Luyt, M. Clavel, K. Guntupalli, J. Johannigman, J.I. Kennedy, C. Wood, K.
powders with specific focus on inhalation-powders, J. Aerosol Sci. 42 (2011) Corkery, D. Gribben, J. Chastre, Pharmacokinetics and lung delivery of PDDS-
428–445. aerosolized amikacin (NKTR-061) in intubated and mechanically ventilated pa-
[119] T.N. Vladykina, B.V. Deryagin, Y.P. Toporov, The effect of surface roughness on tients with nosocomial pneumonia, Crit. Care 13 (2009).
triboelectrification of insulators, Phys. Chem. Mech. Surf. 3 (1985) 2817–2821. [146] G. Mombelli, L. Coppens, J.P. Thys, J. Klastersky, Anti-Pseudomonas activity in bron-
[120] H. Watanabe, M. Ghadiri, T. Matsuyama, Y.L. Ding, K.G. Pitt, H. Maruyama, S. chial secretions of patients receiving amikacin or tobramycin as a continuous infu-
Matsusaka, H. Masuda, Triboelectrification of pharmaceutical powders by particle sion, Antimicrob. Agents Chemother. 19 (1981) 72–75.
impact, Int. J. Pharm. 334 (2007) 149–155. [147] Y. Shaaltiel, U. Hanania, Y. Matiuhin, L. Fux, A. Shulman, T. Kizhner, Dry Powder
[121] P.A. Carter, O.E. Cassidy, G. Rowley, D.R. Merrifield, Triboelectrification of fractionated Formulation of DNase 1, 2013. (WO2013114371 (A1)).
crystalline and spray-dried lactose, Pharm. Pharmacol. Commun. 4 (1998) 111–115. [148] F. Kaare, Methods for Treating Cystic Fibrosis or Pneumonia with Bacterial
[122] M. Murtomaa, P. Harjunen, V. Mellin, V.P. Lehto, E. Laine, Effect of amorphicity on Infection via Pulmonary Administration of Fosfomycin, 2008. (WO2008071197
the triboelectrification of lactose powder, J. Electrost. 56 (2002) 103–110. (A1)).
[123] US Food and Drug Administration, Medication guide. Foradil Certihaler (formoterol [149] D. Heng, S.H. Lee, J. Teo, W.K. Ng, H.-K. Chan, R.B.H. Tan, Synergistic combination dry
fumarate inhalation powder), http://www.fda.gov/downloads/Drugs/DrugSafety/ powders for inhaled antimicrobial therapy, AIP Conf. Proc. 1542 (2013) 113–116.
ucm088603.pdf 2010. [150] Q. Zhou, T. Gengenbach, J.A. Denman, H.H. Yu, J. Li, H.K. Chan, Inhalable powder
[124] UK Medicines and Healthcare Products Regulatory Agency, Public Assessment Re- formulations of combination antibiotics with enhanced antimicrobial activities,
port. Pulmicort CFC-free Inhaler (Budesonide), 2008. 2014, Respiratory Drug Delivery 2014, Fajardo, Puerto Rico, May 4–8, (2014).
[125] Q. Zhou, L. Qu, T. Gengenbach, I. Larson, P.J. Stewart, D. Morton, Effect of surface [151] J.G.Y. Chan, A.S. Tyne, A. Pang, H.-K. Chan, P.M. Young, W.J. Britton, C.C. Duke, D.
coating with magnesium stearate via mechanical dry powder coating approach Traini, A rifapentine-containing inhaled triple antibiotic formulation for rapid
on the aerosol performance of micronized drug powders from dry powder in- treatment of tubercular infection, Pharm. Res. (2013), http://dx.doi.org/10.1007/
halers, AAPS PharmSciTech 14 (2013) 38–44. s11095-013-1245-7 (Published online 16 Nov 2013).
[126] Q.T. Zhou, B. Armstrong, I. Larson, P.J. Stewart, D. Morton, Understanding the [152] J.G.Y. Chan, H.-K. Chan, C.A. Prestidge, J.A. Denman, P.M. Young, D. Traini, A novel
influence of powder flowability, fluidization and de-agglomeration characteristics dry powder inhalable formulation incorporating three first-line anti-tubercular an-
on the aerosolization of pharmaceutical model powders, Eur. J. Pharm. Sci. 40 tibiotics, Eur. J. Pharm. Biopharm. 83 (2013) 285–292.
(2010) 412–421. [153] A. Schoubben, P. Blasi, M.L. Marenzoni, L. Barberini, S. Giovagnoli, C. Cirotto, M.
[127] N.Y.K. Chew, B.Y. Shekunov, H.H.Y. Tong, A.H.L. Chow, C. Savage, J. Wu, H.K. Chan, Ricci, Capreomycin supergenerics for pulmonary tuberculosis treatment: prepara-
Effect of amino acids on the dispersion of disodium cromoglycate powders, J. tion, in vitro, and in vivo characterization, Eur. J. Pharm. Biopharm. 83 (2013)
Pharm. Sci. 94 (2005) 2289–2300. 388–395.

Please cite this article as: Q.(T.) Zhou, et al., Emerging inhalation aerosol devices and strategies: Where are we headed?, Adv. Drug Deliv. Rev.
(2014), http://dx.doi.org/10.1016/j.addr.2014.03.006
Q.(T.) Zhou et al. / Advanced Drug Delivery Reviews xxx (2014) xxx–xxx 15

[154] M.A. Moretton, D.A. Chiappetta, F. Andrade, J. das Neves, D. Ferreira, B. Sarmento, A. Eda, Y. Tsuchihashi, K. Morimoto, M. Akira, M. Terada, J. Otsuka, M. Ebina, C.
Sosnik, Hydrolyzed galactomannan-modified nanoparticles and flower-like poly- Kaneko, T. Nukiwa, J.P. Krischer, K. Akazawa, K. Nakata, Inhaled granulocyte/
meric micelles for the active targeting of rifampicin to macrophages, J. Biomed. macrophage-colony stimulating factor as therapy for pulmonary alveolar
Nanotechnol. 9 (2013) 1076–1087. proteinosis, Am. J. Respir. Crit. Care Med. 181 (2010) 1345–1354.
[155] H. Ikematsu, N. Kawai, Laninamivir octanoate: a new long-acting neuraminidase [176] J.A.R. Portal, E.R. Becerra, A.S. Garrido, Response to inhaled granulocyte-
inhibitor for the treatment of influenza, Expert Rev. Anti-Infect. Ther. 9 (2011) macrophage colony-stimulating factor in a patient with alveolar proteinosis,
851–857. Arch. Bronconeumol. 45 (2009) 150–152.
[156] S. Sunagawa, F. Higa, H.L. Cash, M. Tateyama, T. Uno, J. Fujita, Single-dose inhaled [177] S.B. Venkateshiah, T.D. Yan, T.L. Bonfield, M.J. Thomassen, M. Meziane, C. Czich, M.
laninamivir: registered in Japan and its potential role in control of influenza epi- S. Kavuru, An open-label trial of granulocyte macrophage colony stimulating factor
demics, Influenza Other Respi. Viruses 7 (2013) 1–3. therapy for moderate symptomatic pulmonary alveolar proteinosis, Chest 130
[157] S. Kashiwagi, S. Yoshida, H. Yamaguchi, N. Mitsui, M. Tanigawa, K. Shiosakai, N. (2006) 227–237.
Yamanouchi, T. Shiozawa, F. Yamaguchi, Clinical efficacy of long-acting neura- [178] Comparing efficacy and safety of inhaled SNG001 to placebo, http://clinicaltrials.
minidase inhibitor laninamivir octanoate hydrate in postmarketing surveillance, gov/ct2/show/NCT01126177 .
J. Infect. Chemother. 19 (2013) 223–232. [179] http://www.synairgen.com/programmes/ifn-β-in-copd.aspx .
[158] S. Kubo, M. Kakuta, M. Yamashita, In vitro and in vivo effects of a long-acting anti- [180] http://www.synairgen.com/programmes/ifn-β-in-severe-viral-lung-infection.aspx .
influenza agent CS-8958 (laninamivir octanoate, Inavir) against pandemic (H1N1) [181] R. Rafii, M.M. Juarez, T.E. Albertson, A.L. Chan, A review of current and novel ther-
2009 influenza viruses, Jpn. J. Antibiot. 63 (2010) 337–346. apies for idiopathic pulmonary fibrosis, J. Thorac. Dis. 5 (2013) 48–73.
[159] M. Yamashita, T. Tomozawa, M. Kakuta, et al., CS-8958, a prodrug of the new neur- [182] T.E. King Jr., C. Albera, W.Z. Bradford, U. Costabel, P. Hormel, L. Lancaster, P.W.
aminidase inhibitor R-125489, shows long-acting anti-influenza virus activity, Noble, S.A. Sahn, J. Szwarcberg, M. Thomeer, D. Valeyre, R.M. du Bois, I.S. Grp, Effect
Antimicrob. Agents Chemother. 53 (2009) 186–192. of interferon gamma-1b on survival in patients with idiopathic pulmonary fibrosis
[160] N. Kawai, H. Ikematsu, N. Iwaki, et al., Clinical effectiveness of oseltamivir for influenza (INSPIRE): a multicentre, randomised, placebo-controlled trial, Lancet 374 (2009)
A (H1N1) virus with H274Y neuraminidase mutation, J. Infect. 59 (2009) 207–212. 222–228.
[161] Y. Itoh, K. Shinya, M. Kiso, et al., In vitro and in vivo characterization of new swine- [183] A. Misra, A.J. Hickey, C. Rossi, G. Borchard, H. Terada, K. Makino, P.B. Fourie, P.
origin H1N1 influenza viruses, Nature 460 (2009) 1021–1025. Colombo, Inhaled drug therapy for treatment of tuberculosis, Tuberculosis 91
[162] B.D. Kahan, Cyclosporine, N. Engl. J. Med. 321 (1989) 1725–1738. (2011) 71–81.
[163] S. Groves, M. Galazka, B. Johnson, T. Corcoran, A. Verceles, E. Britt, N. Todd, B. [184] R.S. Wallis, Lack of a therapeutic role for interferon gamma in patients with tuber-
Griffith, G.C. Smaldone, A. Iacono, Inhaled cyclosporine and pulmonary function culosis, J. Infect. Dis. 209 (2014) 627–U6628.
in lung transplant recipients, J. Aerosol Med. Pulm. Drug Deliv. 23 (2010) 31–39. [185] R. Condos, W.N. Rom, N.W. Schluger, Treatment of multidrug-resistant pul-
[164] R.W. Nieven, V. Verret, H. Raff, T. Corcoran, S.G. Dilly, The challenges of developing monary tuberculosis with interferon-gamma via aerosol, Lancet 349 (1997)
an inhaled cyclosporine product for lung transplant patient, Respiratory Drug De- 1513–1515.
livery, 2012, pp. 51–60. [186] R. Suarez-Mendez, I. Garcia-Garcia, N. Fernandez-Olivera, M. Valdes-Quintana, M.T.
[165] J. Behr, G. Zimmermann, R. Baumgartner, et al., Lung deposition of a liposomal cy- Milanes-Virelles, D. Carbonell, D. Machado-Molina, C.M. Valenzuela-Silva, P.A.
closporine A inhalation solution in patients after lung transplantation, J. Aerosol Lopez-Saura, Adjuvant interferon gamma in patients with drug-resistant pulmo-
Med. Pulm. Drug Deliv. 22 (2009) 121–130. nary tuberculosis: a pilot study, BMC Infect. Dis. 4 (2004).
[166] H. Sato, K. Ogawa, Y. Kojo, Y. Kawabata, T. Mizumoto, S. Yamada, S. Onoue, Devel- [187] W.J. Koh, O.J. Kwon, G.Y. Suh, M.P. Chung, H. Kim, N.Y. Lee, T.S. Kim, K.S. Lee, Six-
opment of cyclosporine A-loaded dry-emulsion formulation using highly purified month therapy with aerosolized interferon-gamma for refractory multidrug-
glycerol monooleate for safe inhalation therapy, Int. J. Pharm. 448 (2013) 282–289. resistant pulmonary tuberculosis, J. Korean Med. Sci. 19 (2004) 167–171.
[167] K. Yamasaki, P.C.L. Kwok, K. Fukushige, R.K. Prud'homme, H.-K. Chan, Enhanced [188] R. Dawson, R. Condos, D. Tse, M.L. Huie, S. Ress, C.H. Tseng, C. Brauns, M. Weiden, Y.
dissolution of inhalable cyclosporine nano-matrix particles with mannitol as ma- Hoshino, E. Bateman, W.N. Rom, Immunomodulation with recombinant
trix former, Int. J. Pharm. 420 (2011) 34–42. interferon-gamma 1b in pulmonary tuberculosis, PLoS One 4 (2009).
[168] X. Wu, W. Zhang, D. Jr Hayes, H.M. Mansour, Physicochemical characterization and [189] J. Thipphawong, Inhaled cytokines and cytokine antagonists, Adv. Drug Deliv. Rev.
aerosol dispersion performance of organic solution advanced spray-dried cyclo- 58 (2006) 1089–1105.
sporine A multifunctional particles for dry powder inhalation aerosol delivery, [190] E. Huland, H. Heinzer, H. Huland, Overview of interleukin-2 inhalation therapy,
Int. J. Nanomedicine 8 (2013) 1269–1283. Cancer J. Sci. Am. 6 (2000) S104–S112.
[169] N.K. Whitefield, G. Martyn, F. Dey, P. Crew, Immunosuppressant Microparticles [191] C. Posch, F. Weihsengruber, K. Bartsch, V. Feichtenschlager, M. Sanlorenzo, I. Vujic,
Composition, 2012. (WO2010130982 (A2)). B. Monshi, S. Ortiz-Urda, K. Rappersberger, Low-dose inhalation of interleukin-2
[170] L. Willis, D. Hayes Jr., H.M. Mansour, Therapeutic liposomal dry powder inhalation bio-chemotherapy for the treatment of pulmonary metastases in melanoma
aerosols for targeted lung delivery, Lung 190 (2012) 251–262. patients, Br. J. Cancer 110 (2014) 1427–1432, http://dx.doi.org/10.1038/bjc.2014.
[171] M. Mohanka, D. Khemasuwan, J.K. Stoller, A review of augmentation therapy for 62.
alpha-1 antitrypsin deficiency, Expert Opin. Biol. Ther. 12 (2012) 685–700. [192] P.M. O'Byrne, Role of monoclonal antibodies in the treatment of asthma, Can.
[172] R.C. Hubbard, M.L. Brantly, S.E. Sellers, et al., Anti-neutrophil-elastase defenses of Respir. J. 20 (2013) 23–25.
the lower respiratory tract in α1-antitrypsin deficiency directly augmented with [193] J.V. Fahy, D.W. Cockcroft, L.P. Boulet, H.H. Wong, F. Deschesnes, E.E. Davis, J. Ruppel,
an aerosol of α1-antitrypsin, Ann. Intern. Med. 111 (1989) 206–212. J.Q. Su, D.C. Adelman, Effect of aerosolized anti-IgE (E25) on airway responses to
[173] P. Brand, M. Schulte, M. Wencker, C.H. Herpich, G. Klein, K. Hanna, T. Meyer, Lung inhaled allergen in asthmatic subjects, Am. J. Respir. Crit. Care Med. 160 (1999)
deposition of inhaled α1-proteinase inhibitor in cystic fibrosis and α1-antitrypsin 1023–1027.
deficiency, Eur. Respir. J. 34 (2009) 354–360. [194] C. O'Kane, A. Bayliffe, A. Serone, P. Bareille, S. Smith, U. Hamid, V. Brown, T. Wright,
[174] D.E. Geller, K.C. Kesser, The I-neb adaptive aerosol delivery system enhances deliv- Y. Chen, R. Wilson, J.S. Elborn, D.F. McAuley, Tumour Necrosis Factor receptor 1 in-
ery of a1-antitrypsin with controlled inhalation, J. Aerosol Med. Pulm. Drug Deliv. hibition using a novel inhaled human antibody reduces inflammation in a human
23 (2010) S55–S59. model of lung injury induced by inhaled lipopolysaccharide; a randomised
[175] R. Tazawa, B.C. Trapnell, Y. Inoue, T. Arai, T. Takada, Y. Nasuhara, N. Hizawa, Y. placebo-controlled clinical trial, Thorax 68 (2013) A51.
Kasahara, K. Tatsumi, M. Hojo, H. Ishii, M. Yokoba, N. Tanaka, E. Yamaguchi, R.

Please cite this article as: Q.(T.) Zhou, et al., Emerging inhalation aerosol devices and strategies: Where are we headed?, Adv. Drug Deliv. Rev.
(2014), http://dx.doi.org/10.1016/j.addr.2014.03.006

You might also like