Sie sind auf Seite 1von 21

Advanced Drug Delivery Reviews 58 (2006) 1009 – 1029

www.elsevier.com/locate/addr

Particle engineering techniques for inhaled biopharmaceuticals☆


Sunday A. Shoyele ⁎, Simon Cawthorne
3M Health Care Ltd, Morley Street, Loughborough, LE11 1EP, UK
Received 14 March 2006; accepted 25 July 2006
Available online 12 August 2006

Abstract

Formulation of biopharmaceuticals for pulmonary delivery is faced with the challenge of producing particles with the
optimal properties for deep lung deposition without altering the native conformation of these molecules. Traditional techniques
such as milling are continuously being improved while newer and more advanced techniques such as spray drying, spray freeze
drying and supercritical fluid technology are being developed so as to optimize pulmonary delivery of biopharmaceuticals.
While some of these techniques are quite promising, some are harsh and impracticable. Method scale up, cost-effectiveness and
safety issues are important factors to be considered in the choice of a technique. This paper reviews the presently developed
techniques for particle engineering biopharmaceuticals.
© 2006 Elsevier B.V. All rights reserved.

Keywords: Biopharmaceuticals; Protein/peptide; Particle engineering; Aerodynamic diameter; Spray drying; Supercritical fluid

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1010
2. Milling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1011
2.1. Jet milling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1011
2.2. Media milling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1012
3. Spray drying . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1013
3.1. CO2-assisted nebulization with a bubble dryer™ (CAN–BD) . . . . . . . . . . . . . . . . . . . . . . 1016
3.2. Supercritical fluid-assisted atomization (SAA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1016
3.3. Nebulization, air drying and electrostatic collection (NAE) . . . . . . . . . . . . . . . . . . . . . . . . 1016
4. Spray freeze-drying (SFD) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1017
4.1. Spray freezing into liquid (SFL). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1018
4.2. Spray-freezing with compressed CO2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1018


This review is part of the Advanced Drug Delivery Reviews theme issue on ʽʽChallenges & Innovations in Effective Pulmonary Systemic &
Macromolecular Drug Delivery", Vol. 58/9–10, 2006.
⁎ Corresponding author. Tel.: +44 1509613921; fax: +44 1509613944.
E-mail address: sashoyele@mmm.com (S.A. Shoyele).

0169-409X/$ - see front matter © 2006 Elsevier B.V. All rights reserved.
doi:10.1016/j.addr.2006.07.010
1010 S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029

5. Supercritical fluid technology (SF) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1018


5.1. RESS process . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1020
5.2. PGSS. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1020
5.3. GAS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1021
5.4. ASES. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1021
5.5. SEDS. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1022
5.6. PCA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1022
6. Microcrystallization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1023
6.1. Microcrystallization and sustained release . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1024
6.2. Polymorphism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1024
6.3. Controlling polymorphism with SF . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1024
7. Aerodynamic particle diameter . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1025
8. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1025
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1025

1. Introduction Although, the oral route has showed some promise


[6,7], delivering biopharmaceuticals via this route is
A biopharmaceutical is a protein or nucleic acid-based still quite challenging because of the vulnerability of
pharmaceutical substance used for therapeutic or in vivo these molecules to some gastro-intestinal enzymes and
diagnostic purposes, which is produced by means other reduced bioavailability caused by first pass metabo-
than direct extraction from a native (non-engineered) lism. Alternative routes such as transdermal, nasal,
biological source [1]. In other words, biopharmaceuticals buccal and ocular have been investigated [8–12].
include recombinant proteins, monoclonal antibodies Although, these routes showed little success, the search
and nucleic acid-based therapeutics. for a suitable alternative to injection still continues.
The biopharmaceutical industry started in the late Systemic delivery of biopharmaceuticals by inha-
1970s [2] with the development of the twin techniques lation is currently attracting considerable attention
of genetic engineering and hybridoma technology, but it because a number of these molecules are more
was not until 1982 that the first biopharmaceutical, efficiently absorbed from the lungs compared to oral,
‘Humulin™’ (recombinant human insulin, developed nasal or transdermal routes [13].
by Genentech and Eli Lilly) gained marketing approval The inhalation route offers an enormous absorptive
in the US [3]. Since this period, the market for surface area in the range 35–140 m2, thin (0.2 μm) and
biopharmaceuticals for use in human therapy has highly vascularized epithelium, which leads to high
continued to grow at a high rate with 270 new bioavailability [14]. Furthermore, in most societies oral
biopharmaceuticals being evaluated in clinical studies inhalation is well accepted by the general population
currently and estimated sales in excess of [15].
30 billion euros annually [4]. Presently, up to 160 bio- Formulating biopharmaceuticals for inhalation pre-
pharmaceuticals have gained medical approval and most sents new challenges to scientists. It is important that
are protein-based, although two nucleic acid-based these molecules be size reduced without altering their
products are now on the US/European market [5]. native conformations and also have optimal aerody-
Most of these approved biopharmaceuticals are namic properties for them to get to the deep lungs
being delivered via injection. Apart from the inconve- when inhaled [16].
nience this might cause to the patient (pain, abscess, Since the mucocilliary apparatus and alveolar
etc.), injections, in the majority of cases, cannot be macrophages are both involved in the clearance of
administered by patients themselves. There is a need drug molecules from the lung [17]; there is the
for some expertise of a healthcare professional. These possibility of producing particles to avoid these
factors do not help compliance and so there is a need mechanisms. For instance, uptake of particles by the
for alternative methods of administering these drugs to alveolar macrophages has been found to be size-
the patient. dependent [17]. Particles 6 μm are phagocytosed to a
S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029 1011

much smaller extent than those 3 μm in diameter [18], 2.1. Jet milling
while particles of diameter less than 0.26 μm are
minimally taken up by macrophages. However, if the Jet milling process involves micronization by inter-
objective of delivery were to target the alveolar particle collision and attrition [30]. The milling medium
macrophages as a medium for drug delivery [19,20], is a high velocity gas (air), which passes through the
particles of mass median diameter of around 3 μm would nozzles and carries along coarse particles into the jet mill
be the ideal particles to target. Depending on what the where they are suspended in the turbulent gas stream.
objectives of delivery are, particles of a specific size Coarse particles are fractured into smaller once in the gas
distribution will have to be produced for effective stream due to inter-particle collisions [31]. The spherical
delivery. and elliptical path taken by the gas stream moving at high
Biopharmaceuticals are relatively large molecules; speed, facilitates the exit of fine particles while larger
studies have shown that molecules with molecular particles remain in the mill for further micronization by
weight above 30 kDa may need an absorption enhancer more inter-particle collision. Jet mills can typically
to be absorbed in the alveoli [21]. Some of the absorption produce particles within the size range of 1–20 μm [25].
enhancers used involve entrapping or encapsulating Despite the possibility of having fine materials
these molecules within them [22]. This can potentially through jet milling, the disadvantages such as lack of
increase the size of the particles produced. There is control over parameters such as size, shape, morphol-
therefore a need to reduce these particles to within the ogy and surface properties of the milled particles
respirable range. coupled with the high energy input which can promote
Whilst it is crucial to control the aerodynamic particle chemical degradation [29], make this technique less
size, it is also important to control the surface properties promising for biopharmaceuticals. Furthermore, it has
of reduced particles as micronized particles tend to be been observed that the high energy required for such
highly charged leading to agglomeration. Furthermore, processes often damage the crystal surface, leading to
milling damages the crystal surface of particles leading highly charged and cohesive particles which result in
to partially amorphous parts, which leads to physical chemical and physical instability of the drug [23].
instability of the drug [23]. These limitations, however, have not dissuaded
Inhaled biopharmaceuticals therefore need to be scientists from exploring this cost-effective technique.
size reduced by suitable techniques that not only It has been studied as a potential particle engineering
produce particles within the target range of 1–5 μm but technique for various proteins.
also with optimal surface properties to aid pulmonary The model protein horseradish peroxidase has been
delivery without loss of biological activity. co-jet milled with poly(acrylate) to produce protein-
Because most biopharmaceuticals are quite labile loaded microparticles [32] after co-precipitating the two
[24], care should be exercised during their processing. compounds from demineralized water. Precipitating
Conventional micronization techniques such as milling with petroleum ether led to a remaining activity of 67±
have been used with limited success [25,26]. Alterna- 4.7% while isopropanol was the best precipitant leading
tive techniques are continuously evolving leading to to a remaining activity of approximately 90%. It was
the production of particles with the required optimal concluded that co-precipitation has a much greater
properties which include among others, narrow size impact on protein stability than the milling process itself.
distribution, optimal aerodynamic diameter and low Leuprolide acetate, a synthetic nonapeptide has been
surface energy. jet-milled and formulated into a metered dose inhaler
(MDI) [33,34]. It was observed that suspension MDI of
2. Milling leuprolide had a higher bioavailability (27.9% relative to
subcutaneous administration) compared to solution
Milling is the traditional method of drug powder MDI, using 20% ethanol as a co-solvent (6.6% relative
micronization [27]. Various mills like ball mills, to subcutaneous administration). This low bioavailabil-
colloid mills, hammer mills and jet or fluid energy ity could be attributed to increased chemical degradation
mills have been studied [28], however, the majority of of the protein facilitated by the ethanol or coarse aerosol
inhalation powders are prepared with a jet mill [29]. plume in the ethanol system leading to reduced
1012 S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029

deposition in the deep lung, resulting in reduced uptake 2.2. Media milling
into the systemic circulation.
Insulin that is available as a crystalline powder [25] Milling as a technique has been improved and
has been jet-milled using nitrogen as the gas, to an adapted for the engineering of biopharmaceutical
average particle size of 2.4 μm [35]. The occurrence of molecules. Media milling is a concept that has found
insulin as crystals makes it amenable to jet milling due some application in biopharmaceutical processing. It
to good flow properties. involves using a liquid as a medium during milling and
The nature of the particles fed into the jet mill has a was first recommended by Adjei et al. using non-CFC
substantial effect on the efficiency of the mill [25]. If propellants for the preparation of peptide suspensions
particles fed in are too large, they can block the hopper for pressurized metered dose inhaler (pMDI) [42].
and pipe-work in the mill while too fine powder may Lizio et al., however, improved the technique using a
make it difficult to obtain a smooth feed rate into the mill, modified and abrasive resistant pearl mill [43]. They
with the optimal size of most materials for jet milling were able to produce fine protein particles with a
being in the 75–100 μm range. Brittle materials tend to volumetric mean diameter of 3.1 μm without gener-
fracture during inter-particle collision while ductile ating degradation products or contaminants from the
materials tend to undergo plastic deformation rather milling process. An important feature of this technique
than fracture [28]. However, Tomihisa [36] proposed that is the direct manufacturing of fine drug suspension for
chilling ductile materials before and during milling systemic aerosol delivery by pMDI.
makes them more brittle, which facilitate size reduction. Media milling has also been used by Zijlstra et al. to
Biopharmaceuticals that are not available as crystals produce protein with comparable qualities to particle
or forms suitable for jet milling can be lyophilized to engineering techniques such spray drying and spray
prepare coarse powders [25]. Kobayashi et al. jet-milled freeze drying [33]. They worked with cetrorelix
salmon calcitonin powders that had to be lyophilized acetate, a decapeptide used for the controlled ovarian
with lactose in order to improve the flow of the powder super stimulation for assisted reproductive technique.
prior to milling [37]. However, lyophilization followed Using a pearl mill equipped with a cryostat, the system
by milling can lead to loss of activity of the biomolecule was cooled to − 60 °C, cetrorelix lyophilisate in HFA
unless a suitable excipient is used as a lyoprotectants 227 was milled with ZrO2-Ir stabilized pearls for
during lyophilization. Lyoprotectants are important 15 min. It was observed that only minute amounts of
during lyophilization because at sub-zero temperatures, degradation products (b1.17%) were produced. This is
water crystallizes and leads to a strong increase in the indicative of the stability of the milled cetrorelix,
concentration of the molecules dispersed in it. This so- although minimal metal contaminants were observed.
called freeze concentration forces particles towards each Scanning electron micrograph (Fig. 1) showed that
other, which facilitates aggregation. [38]. Lyoprotectants milled cetrorelix consist of agglomerates of very small
prevent such aggregation. Preventing aggregation during primary particles which are mostly smaller than 1 μm.
lyophilization is further important because once in a Dynamic vapour sorption studies showed that the
stable dry state, increased shelf life can be expected even surface properties of milled cetrorelix was comparable
at ambient temperature [38,39]. For instance, a formu- to those from spray drying or spray freeze drying. The
lation of interferon-β containing sorbitol, human serum aerosol performance of the dry powder formulation
albumin and NaCl, did not significantly reduce its was assessed by comparing the fine particle fraction
activity after lyophilization and jet milling, while the (FPF) (b3.75 μm) of emitted powder from both the
same formulation without sorbitol showed significant ISF inhaler and the Novolizer. The ISF inhaler
loss of activity [40,41]. produced an FPF of 30% while the novolizer produced
Although the use of jet milling in size reduction of an FPF of 40%.
biopharmaceuticals has been relatively successful, the Irngartinger et al. also worked on media milling
two-step approach of either crystallizing or lyophilizing cetrorelix [44]. Using the same modified pearl mill as
before milling can make this technique more expensive described above, but incorporating 96% ethanol as a
and time-consuming. It also sometimes lead to degra- coolant, the cetrorelix was suspended within fluid
dation of the drug as shown above. propellant and after the milling, the suspension of
S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029 1013

a site for degradation of biopharmaceuticals. The use


of suitable excipients can reduce denaturation during
spray drying. For instance, polysorbate-20 has been
used to reduce spray drying induced denaturation of
human growth hormone [25].
Careful selection of operating parameters can play a
significant role in obtaining high quality product
during spray drying. Experimental design techniques
have been used to optimize the spray drying process
for β-galactosidase [46].
The yield obtained from spray drying also constitutes a
Fig. 1. SEM showing media-milled cetrorelix acetate. Reprinted limitation to the use of this technique. Spray drying has
from Zijlstra et al. [33] with permission from Elsevier. relatively low cyclone collection efficiency for particles
below 2 μm [25]. The typical yield from a spray dryer is
between 20% and 50% [47]. However, the introduction of
milled cetrorelix acetate in propellant was poured into high-performance cyclone by Büchi, Switzerland, has
a flask and rotated at room temperature until the improved the yield from spray drying to greater than 70%
propellant was completely evaporated. The dry [48].
powder obtained can be formulated as a dry powder Other important issues in spray drying are the lack of
inhaler, which makes this technique quite versatile. control over the mean droplet size, possibility of a broad
The use of media milling has improved the use of droplet distribution and the risk of clogging if pneumatic
milling for size reducing proteins and other biomole- nozzles are used [44]. The use of an ultrasonic nozzle,
cules as the low temperature of milling could reduce which is able to generate droplets with more uniform size,
deformation of these biomolecules and possibly reduce leading to a relatively homogenous size distribution of
the number of steps involved in milling. the produced powder, has been suggested [49]. The
problem envisaged here is that the energy needed for
3. Spray drying atomization may further contribute to the denaturation of
proteins.
In the spray drying process a feed solution containing Spray drying has however been recognized as a
the biopharmaceutical is atomized into droplets that dry successful process for generating protein-containing
rapidly due to their high surface area and intimate powder in a single step from solutions [50]. It is a
contact with the drying gas (compressed air) [45]. The technique that has found widespread use in particle
drying time for droplets depends on the process engineering of inhaled biopharmaceuticals, from pro-
conditions such as flow rate, pump rate, aspiration rate duction of aerodynamic particles to controlled release
and heat. This drying time can range from less than delivery systems. Particles from spray drying may be
100 min to a few seconds [25]. The temperature spherical, have convoluted surfaces, asperities, holes or
experienced by the droplets is considerably lower than voids [45,51].
the temperature of the drying air due to evaporative Patton et al. have been able to prepare respirable
cooling. The dried powder is protected from overheating insulin powders by spray drying a solution of the drug
by rapid removal of solvent from the drying zone. The with sodium citrate and mannitol [52]. A powder with
final product can be removed from the air stream by the particles typical of spray-dried particles, with a mass me-
use of cyclones or filters. dian diameter ranging from 2.0 to 2.8 μm was achieved.
Although thermal degradation can possibly be Recombinant human granulocyte colony-stimulating
avoided in spray drying, degradation during atomiza- factor (rhG-CSF), a protein with a molecular weight of
tion may constitute a problem to biopharmaceuticals. 18.8 kDa used in cancer chemotherapy has been
Atomization requires high shear rates, which can successfully spray-dried and tested for pulmonary
denature proteins [25]. The large surface area of the absorption in rabbits [53]. The product obtained had a
droplets improves mass transfer but can sometimes be mass median diameter of less than 3 μm with a
1014 S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029

corrugated surface. It was observed that the rhG-CSF acetate and although particles with median diameter of
was absorbed more rapidly from the lungs of the rabbit 3.5 μm with 83% of the particles smaller than 5 μm
than the subcutaneous dose. The inhalation powder also were produced, it was discovered that the outlet
had the desired pharmaceutical effect of increasing the temperature had an enormous effect on the rate of
white blood cell count, showing that the biological degradation of the peptide. A sample exposed to nearly
activity of the spray-dried powder was maintained. 60 °C at the product filter contained 1.3% of ornithin
Sutton et al. have succeeded in producing hollow derivatives while a sample exposed to nearly 50 °C
protein microcapsules using spray drying [54]. Al- generated a just 0.6% of ornithin derivative.
though the intended use for this fine powder is ultrasonic The effect of co-spraying recombinant human
imaging contrast enhancement, many of the particles DNase (rhDNase) with NaCl on the aerodynamic
prepared had dimensions in the respirable range. This properties of the protein has been studied [57]. Spray-
study was also able to show the effect of nozzle type on dried pure rhDNase powder was quite cohesive with a
the dimensions of particle obtained. When a two-fluid fine particle fraction of about 20% while powders
nozzle was used for atomization, the particle size ranged containing NaCl showed a linear relationship between
from 4 to 6 μm, while when a centrifugal nozzle was the NaCl content and FPF for a similar primary size
used, particles in the range 1–8 μm were obtained. (approximately 3 μm volume median diameter) of
Flake-like, corrugated aerodynamic, cetrorelix parti- particles. The particle morphology of these powders
cles (Fig. 2), have been produced by spray drying [33]. varied systematically with the salt content.
Lyophilized cetrorelix was dissolved in a mixture of The effect of various excipients on the biological
40 ml tert-butanol and 60 ml of distilled water. Tert- activity and aerosol performance of spray-dried
butanol was added because it improves dispersion lysozyme and catalase has also been studied [58].
characteristics of water [55]. The powder produced had Sucrose, trehalose and polyvinyl alcohol (PVA) were
a fine particle fraction (FPF) of 66%, which is consistent studied as potential stabilizers for proteins during
with the morphology of the particles produced. spray drying. It was observed that pure lysozyme, lost
Salmon calcitonin has also been spray-dried and the just over 10% of its original biological activity, while
effect of mannitol on aerosol performance of the powder the incorporation of sucrose, trehalose or a combina-
produced studied [56]. It was observed that surface tion of trehalose and PVA with lysozyme preserved
enrichment by mannitol increases surface energy approximately 100% of its original biological activity.
measured by inverse gas chromatography (iGC) and Pure catalase retained 55% of its biological activity
emitted dose was improved as the mannitol concentra- upon spray drying but this retention of activity was
tion decreased. increased to approximately 93% when either sucrose or
Irngartinger et al. [44], studied the effect of outlet trehalose was used to stabilize the enzyme during the
temperature on the stability of spray-dried cetrorelix manufacturing process. Catalase retained almost full
activity when it was stabilized using a mixture of PVA-
trehalose. SEM showed that the morphology of the
spray-dried powders appeared to be similar regardless of
the type and quantities of excipients used to stabilize the
protein.
Lactose, trehalose, dipamitoylphosphatidylcholine
(DPPC) and/or albumin have been studied as potential
excipients for spray drying proteins [59]. Lactose is of
particular interest because it had been established earlier
that lactose, being a reducing sugar, has the potential to
react with the primary amine of lysine residue and
generate Schiff-base by-product [60]. Using parathyroid
hormone (PTH) as model protein, after the spray drying
Fig. 2. SEM showing spray-dried cetrorelix acetate. Reprinted from process, the mass spectrum of the powder containing
Zijlstra et al. [33] with permission from Elsevier. lactose did not show any by-product resulting from a
S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029 1015

covalent bond between the sugar and PTH; this was also Nektar Pulmonary Delivery System was approximately
the case for the spectrum of powder containing trehalose 90% and a fine particle mass, defined as particles
a non-reducing sugar. No peaks from other degradation b3.3 μm, was 56%.
products were observed by either HPLC or mass Spray drying has also been used to prepare encapsu-
spectrometry. It was further demonstrated that albumin lated bovine serum albumin (BSA) from aqueous two-
as an excipient could markedly decrease systemic phase systems (ATPs) containing polyvinyl alcohol and
absorption of the hormone through binding. However, dextran solutions in different proportions [64]. Using a
all the powders produced presented an average primary laboratory-scale spray dryer, the dryer was operated in
particle diameter between 3.9 and 5.9 μm and achieved co-current mode, with a jacketed two-fluid nozzle. The
up to 98% emitted dose and up to 61% FPF using a DPI emulsion produced from the ATPs was continuously
device. stirred as it was fed into the spray dryer. Particles were
Further studies have been performed on the use of separated from the dry air by a membrane filter. SEM
lactose and DPPC as excipients for spray drying micrograph of the powders produced showed that the
proteins. Bosquillon et al. [61] prepared a dry powder shape and surface morphology of particles were strongly
by spray drying human growth hormone (hGH) with affected by the composition of the two-phase system. As
lactose and DPPC in proportion 20:20:60 w/w/w in a the PVA content increased, the particles turned from
0.1% total excipient concentration. highly wrinkled to smooth spheres. Thread-like struc-
The proportion of soluble hGH dimers after powder tures and small doughnut shaped particles were also
production was evaluated by size-exclusion chroma- observes as well as a slight increase of the particle size
tography. The dimer content was b1% in native hGH as when the PVA content increased.
well as in hGH powder, indicating that incorporation of Controlled release anti-influenza antibody has been
hGH in the a solution containing lactose and DPPC did produced for inhalation using immunoglobulin (Ig)
not induce dimerization. In contrast, 3.9% of hGH incorporated spray-dried lipid micro-particles (SDLM)
dimers were obtained following spray drying with [65]. DPPC was used as the major excipient because of
lactose in the absence of DPPC. This shows that DPPC its biocompatibility and being a component of normal
has a stabilizing effect on proteins when co-spray-dried lung surfactant. Either lactose or hydroxyethylstarch
with lactose. This assumption was further demostrated (HES) was used as an additional excipient. Prior to
by the fact that when a potential shift in iso-electric spray drying, the lipid was complexed with Ca2+ in a
point (pI) was analysed by iso-electric focussing, a dry 1:1 molar ratio, yielding SDLM of increased stability.
powder containing trehalose, a non-reducing sugar, as a Two categories of particles were generated:
negative control, no modification to the protein pI was
observed after formulation of both lactose/DPPC • Retentive powder, containing 10% (w/w) HES in
powder and trehalose/DPPC powder and up to 5 months addition to 60% (w/w) DPPC-Ca2+/DPPE-Texas
of storage at 4 °C for the powder containing lactose and Red and 30% (w/w) of human IgG
DPPC. • Non-retentive particles that were constructed by
Stabilizing proteins with a surface acting agent replacing HES with lactose plus 1% tyloxapol, a
(surfactant) is a crucial step during spray drying of biocompatible surfactant detergent approved for
proteins because proteins being amphipathic tend to human use in surfactant formulations [65]
adsorb at the air–liquid interface of droplets in spray,
unfold and aggregate at the droplet surface. Excipients Co-spray drying proteins with biocompatible sur-
have been found to exclude proteins from this interface factant lipid, such as DPPC (with or without additives
and promote their stability [62]. such as tyloxapol) dramatically reduced the particle
Spray drying as a technique has also found great use size and improved the aerodynamic characteristics and
in controlled release of biopharmaceuticals. homogeneity of aerosols (MMAD of 3.42 μm with
The PEG–insulin covalent complex developed for FPF in excess of 70%). In addition, such surfactant–
controlled released insulin by Nektar has been prepared lipid excipients make the microparticles compatible
for pulmonary delivery by spray drying [63]. Using a with non-aqueous propellants, offering a degree of
Büchi 190 mini spray dryer, the ED of the aerosol from flexibility in the strategy of delivery [53].
1016 S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029

It was concluded that controlled spray drying of IgG, to assist the nebulization of the solution [68].
with DPPC-Ca2+ as a major excipient, results in the Advantages attributed to this technique include: the
generation of microparticles with good aerodynamic minimal decomposition of thermally labile drugs such
properties. as proteins, the absence of a high pressure vessel as
Microencapsulated chitosan nanoparticles for lung opposed to normal supercritical fluid technologies, and
delivery of proteins have been produced by spray the small size of particle produced (below 3 μm) [68].
drying [66]. The process involves, firstly, preparing the Two variants of this technique have been reported.
protein loaded nanoparticles by a process described by
Calvo et al. [67] and then encapsulating them in lactose 3.1. CO2-assisted nebulization with a bubble dryer™
and mannitol. The encapsulation was performed by (CAN–BD)
spray drying. Aqueous solutions of the excipients,
lactose and mannitol, were prepared by dissolution in In this method, the drug is dissolved in water, alcohol
purified water to obtain concentration of 7.5%–10%. or a mixture of both. The solution produced is mixed with
Chitosan nanoparticles prepared were resuspended in CO2 in either a sub-critical or supercritical state by
purified water and mixed with the excipients solution in pumping both fluids through a low volume tee to generate
order to achieve excipient/nanoparticle ratio of 95:5, an emulsion [69]. The emulsion produced expands
90:10, 80:20, and 50:50 (w/w). The final suspension through a nozzle into the drying chamber, which is held
concentration ranged from 0.84% to 8.4% (w/v). Dry at atmospheric pressure to generate aerosol microbubbles,
powders were obtained by spray drying aqueous and microdroplets that are dried by a heated nitrogen gas
solutions of the excipients and suspension of chitosan source.
nanoparticles in the excipients using Büchi mini spray Sievers et al. have applied this technique to prepare
dryer, B290 model with a feed rate of 2.5 ml/min. rhDNase aerosol for nebulization [70]. Buffered
The morphology of the particles produced was aqueous solution of rhDNase was mixed with SFCO2
dependent on powder composition. Mannitol solution in a low dead-volume tee. The mixture formed was
without nanoparticles produced mostly spherical but expanded through a restrictor to form aerosol particles
aggregated microparticles while lactose solution pro- with diameter less than 5 μm. The aerosol formed
duced well-defined, non-aggregated spherical micro- could be diluted with air for inhalation or collected for
spheres. Combination of mannitol (90%) and lactose formulation in other pharmaceutical products such as
(10%) also produced spherical particles which were pMDI. Following the processing, 95% of the respira-
less aggregated than the mannitol particles with better ble protein was in an active unagglomerated form.
defined limits. Spherical microparticles with more
defined limits were produced upon incorporation of 3.2. Supercritical fluid-assisted atomization (SAA)
the protein-containing chitosan nanoparticles. This
might suggest that the incorporation of nanoparticles Reverchon et al. [71] described a process where
as a solid structure enhances the morphology of the supercritical CO2 and a solution of the drug (in water
microspheres. In the formulation comprising mannitol or organic solvent) are mixed and sprayed into a vessel
with 10% lactose, no improvement in particle morpho- at conditions near atmospheric pressure and a flow of
logy was observed. In addition, no differences in hot nitrogen.
morphology were found between microparticles made This technique has been used in the processing of
of mannitol-containing nanoparticles and microparti- protein powders such as recombinant human deoxy-
cles of both mannitol and lactose with nanoparticles. ribonuclease (rhDNase), lysozyme, lactate dehydroge-
Despite the seeming success of spray drying as a nase (LDH), ovalbumin, and trypsogen [72–75].
technique for particle engineering of biopharmaceu-
ticals, further modifications have been performed on it 3.3. Nebulization, air drying and electrostatic
to further produce respirable particles with total collection (NAE)
retention of biological activity.
Supercritical carbon dioxide (CO2)-assisted spray Dalby et al. have also described a process similar to
drying is a technique where supercritical CO2 is used spray drying [76]. Fine protein particles were produced
S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029 1017

by combining nebulization, air drying and electrostatic is used makes this technique hugely impractical.
collection. Using alkaline phosphatase as a model Combining a spray dryer with a lyophilizer makes
protein, a solution of this protein was prepared with the technique quite expensive.
lactose and nebulized in a small-particle aerosol Using cetrorelix as a model peptide, Zijlstra et al.
generator (SPAG-2). This aerosol entered a secondary [33] were able to produce particles of large surface
drying chamber followed by an electrostatic collector, area and high porosity which were comparable to those
which could be operated with a potential difference produced by Costantino et al. [79] and Maa at al. [78]
ranging from 0 to 20 kV. Particles were recovered from (as seen in Fig. 3), although degradation of the drug
the precipitator by rinsing with a propellant. Although did not occur, the aerosol performance of the powder
this technique produced fine particles, there was some produced in an adhesive mixture was poor. This could
loss in the activity of the alkaline phosphatase. be attributed to the fact that SFD produced very fragile
particles, which cannot withstand the production
4. Spray freeze-drying (SFD) process of an adhesive mixture. During the mixing
process, the fragile particles of SFD processed cetro-
SFD was first introduced in 1994 [77]. It was relix powder were exposed to impact forces from
classified as a variant of dry milling. It involves spraying colliding lactose particles, which crushed the particles
a solution containing the macromolecule into a vessel to smaller fragments. These very small fragments,
containing a cryogenic liquid such as nitrogen, oxygen or once attached to the lactose carrier, are more difficult
argon. Since the normal boiling point for such a liquid is to separate again from the carrier during inhalation
very low, the droplets are quickly frozen. Lyophilizing because of their small diameter and low density (small
these frozen droplets results in porous spherical particles amount of mass per particle).
suitable for inhalation [27]. It has been established that Maa et al. compared particles produced from spray
these droplets may begin to freeze during the time of and spray freeze drying of rhDNase and anti-IgE
flight through the cold vapour phase and then completely monoclonal antibody (anti-IgEMAb) [79].
freeze upon contact with cryogenic liquid [78–81]. Using the same atomization procedure, spray
Although this technique has been used for producing drying produced small (approximately 3 μm) dense
protein particles, it is faced with the limitations of stresses particles, but SFD resulted in large (approximately 8–
associated with freezing and drying, which may cause 10 μm), porous particles. The FPF of the spray freeze-
irreversible damage to the protein. This is manifested as dried powder was significantly better than that of the
structural denaturation, aggregation and loss of biological spray-dried powder, which confirms the concept of
activity upon rehydration [82,83]. Similar to spray porous particles giving low aerodynamic size.
drying, loss of stability due to unfolding and aggregation The enzyme lysozyme has been spray freeze-dried
remains a major challenge. [84]. Using an aqueous feed containing lysozyme
Webb et al. were able to show that adsorption of
recombinant human interferon-γ (rhIFN-γ) at the air–
liquid interface produced by the atomization resulted
in significant aggregation [81]. This confirms that the
majority of protein activity loss in the SFD process is
due to protein adsorption at the air–liquid interface in
the atomization step and during the freezing of the
droplet in the cold vapour phase rather than during the
lyophilization process.
Further possible limitations to this technique
include the fact that it is time consuming, the nature
of the process has safety issues involved with spraying
into cryogenic fluids and it is expensive. A full SFD
process may take as long as 3 days and the fact that Fig. 3. SEM showing spray freeze-dried cetrorelix acetate. Reprinted
liquid nitrogen with a boiling point below − 195.8 °C from Zijlstra et al. [33] with permission from Elsevier.
1018 S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029

powder in 10 mM phosphate buffer (pH 7.4). The 4.2. Spray-freezing with compressed CO2
aqueous feed solution was sprayed through a two-fluid
nozzle. The frozen particles were collected and lyoph- In this process, aqueous solution of the biopharma-
ilized. It was discovered that the rapid freezing rates ceutical and excipients is simultaneously atomized and
obtained in the SFD process generate significant super- frozen by dispersing supercritical CO2 in it [88]. The
saturation and thus rapid nucleation rates of dissolved solution containing the biopharmaceutical is mixed
substances. The rapid nucleation and restricted growth with compressed CO2 in a static mixer to form droplets
that occurs after the fine droplets are formed and frozen of a CO2-saturated aqueous solution dispersed in CO2.
leads to extremely small primary particles with a high Joule–Thomson expansion cooling brings about the
surface area following lyophilization [84]. rapid freezing of the droplets when injected into the
Having confirmed that adsorption of protein at the spray tower through a nozzle [88]. Once the spraying
air–liquid interface during atomization is mainly process is completed, the frozen particles are lyoph-
responsible for loss of activity of proteins during spray ilized to obtain dried particles. The main goal of this
drying and SFD, techniques have been developed to method is to obtain stable, porous or hollow protein
limit the time of exposure to the air–liquid interface particles with a narrow size distribution suitable for
during atomization. Two techniques have been devel- inhalation.
oped and they include: This method has been successfully used in the
processing of trypsinogen [88]. Using different
• Spray freezing into liquid mathematical models, it was discovered that the flow
• Spray freezing with compressed CO2 rates of the solution and CO2 as well as excipient
concentration could influence the size of solid particles
4.1. Spray freezing into liquid (SFL) of protein produced.

SFL is a novel particle engineering technique which 5. Supercritical fluid technology (SF)
was initially developed for particle engineering poorly
water soluble or insoluble drugs by atomizing a feed Supercritical fluids (SFs) are gases and liquids at
liquid containing a poorly water soluble drug and temperatures and pressures above their critical points (Tc
solubility enhancing excipients below the surface of a – critical temperature; Pc – critical pressure) [89]. The
cryogenic liquid to produce rapidly frozen particles ranges, 1 b T/Tc b 1.1 and 1 b P/Pc b 2, have been estab-
that are subsequently dried [85]. The ultra rapid lished as of particular interest for supercritical fluid
freezing prevents phase separation of the drug and application because in this region, the SF exists as a
excipient and also prevents crystalline growth in single phase with several advantageous properties of
frozen water. These two factors could result in non- both liquids and gases. They have density values that
homogeneous powder aggregates consisting of crys- enable appreciable solvation power, whilst the viscosity
talline drug domains. Ultra-rapid freezing rates of solutes in SF is lower than in liquids. The diffusivity of
produce a porous micronized flowable powder with a solutes is higher, which facilitates mass transfer [89].
high surface area [85]. Most importantly, SFs are highly compressible, partic-
This technique has successfully been adapted for ularly near the critical point and so their densities and
proteins. The liquid solution of protein is sprayed thus solvation power can be controlled by variation in
directly into the liquid nitrogen through an insulated temperature and pressure [90].
nozzle, rather than into cold vapour as compared to SFD. For pharmaceutical applications, the most widely
Insulin particles produced by SFL had a low bulk used SF is CO2 (SFCO2) because of its low critical
density, a high surface area, narrow particle size temperature (31.2 °C) and pressure (7.4 MPa) [66], it is
distribution and no loss in stability as characterized by non-flammable, non-toxic and inexpensive. The only
absence of covalent dimer [86]. Bovine serum albumin limitation facing the use of SFCO2 is its limited
(BSA) was also studied using this technique [87] and solvation power for compounds of interest although
degradation was lower for SFL than in the case of SFD this can be turned into an advantage in a situation
reported in an earlier study [79]. where the SFCO2 is used as an anti solvent [89].
S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029 1019

It is possible to modify the solvation power of a The second method is a relatively harsh technique
SFCO2 by incorporating a small amount of volatile co- but it is similar to the first one because they both
solvent, such as acetone or ethanol (i.e. organic involve using SFCO2 as a solvent rather than an anti
modifier) [91]. Although, the critical properties of the solvent. This has been named ‘precipitation from gas-
resulting supercritical solutions are changed, the saturated solution’ (PGSS) [92]. In this process the SF
addition can dramatically improve the loading capacity is dissolved in molten solute and the resulting
or selectivity of the modified SF [89]. supercritical solution fed via an orifice into a chamber
Particle engineering by SFs can be divided into to allow a rapid expansion under ambient conditions
three broad groups (see Fig. 4) [89]. [93].
The third method is the most common method and
• Precipitation from supercritical solutions composed different variants of the technique have been devel-
of SF and solutions oped. This technique utilizes a similar concept to the
• Precipitation from gas saturated solutions use of anti-solvent in solvent-based crystallization
• Precipitation from saturated solutions using SF as processes [89]. The high solubility of SFCO2 in
anti-solvent. organic solvents leads to volume expansion when the
fluids make contact. This leads to reduction in solvent
The first method involves dissolving or solubilizing density and parallel fall in solvation capacity. Such
the drug in a SF followed by rapid expansion of the SF reduction causes increased level of supersaturation,
solution across a heated orifice to cause a reduction in solute nucleation and particle formation [25].
the density of the solution, thereby decreasing the Acronyms such as GAS (gaseous anti-solvent),
solvation power of the SF which leads to precipitation ASES (aerosol solvent extraction system), SEDS
of the drug [68]. This process has been christened (solution enhanced dispersion by SF), and PCA
‘rapid expansion of supercritical solution’ (RESS) (precipitation by compressed anti-solvent) have been
[89]. assigned to different applications of this technique.

Fig. 4. Schematics of various supercritical fluid techniques. Reprinted from Vemavarapu et al. [134] with permission from Elsevier.
1020 S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029

5.1. RESS process triblock co-polymer. The objective of the study was to
develop a micro-encapsulation process whereby the
Krukonis first applied SFs to produce particles of particle size distribution could be controlled whilst
drugs with a narrow size distribution [94]. Sufficient avoiding the use of a toxic solvent. Ethanol was used as
solubility of the material to be processed in the SF is the a co-solvent. The polymer and co-solvent were charged
major prerequisite for the use of RESS process. The into a pressure vessel. SFCO2 was added to the vessel via
solubility of a drug powder in SF will depend on the a vessel containing the protein of interest. The resultant
density of the SF, the drug's chemical structure and the mixture was stirred for about 4 h and kept for 1 h without
SF-drug contact time [68]. The morphology and size agitation to ensure the polymer was dissolved and the
distribution of the particles formed are dependent on the protein suspended. The resultant solution/suspension was
pre-expansion concentration of the solute in SF and the sprayed through a capillary nozzle held at 313.15 K. The
expansion conditions (e.g. temperature, pressure) [95]. yield of the processed microparticles was found to be
The higher the pre-expansion concentration, the narrower dependent on the solubility of the polymer in the SF/co-
the particle size distribution [68]. The expansion solvent. It was observed that ethanol used as the co-
conditions depend on the nozzle temperature, geometry solvent in this study produced particles that were less
and size [68]. The nozzle has to be maintained at a adhesive. This is thought to be due to the low solubility of
suitable pre-expansion temperature to prevent the high molecular weight PEGs in absolute ethanol.
premature precipitation of the drug.
The limitations facing the use of RESS for biophar- 5.2. PGSS
maceutical include: high temperature needed for the rapid
expansion (typical temperature of 313 K) which can In the PGSS process, the viscosity of the molten
destroy proteins, poor predictive control of particle size compound can be decreased due to the dissolved gas [93].
and morphology [25]. Scale up of this technique is limited The gas saturated liquid phase is expanded to generate
by particle aggregation and nozzle blockage caused particles from materials that are not necessarily soluble in
expansion cooling [25]. It is also limited to molecules that SF and so avoid the limitations of the RESS process.
are soluble in SFCO2. Compounds can also absorb some of the CO2 and con-
Despite these limitations RESS has been studied for sequently swell (polymers) or melt at temperature signi-
the particle engineering of proteins by some authors. ficantly lower than the normal melting or glass transition
Cyclosporin A, an immunosuppressant drug that has temperature.
been used for the treatment of respiratory tract infections, Cyclosporine has been processed using this technique
has been processed by RESS [93]. Cyclosporine was [93]. Melting point depression study was performed on
dissolved in a near critical or SFCO2 and the resulting the cyclosporine by loading 10 mg of cyclosporine in a
solution was depressurized rapidly by spraying the solu- glass tube (i.d. = 5.8 mm) and placed inside a view cell in
tion at atmospheric conditions through a 50 μm nozzle temperature-controlled bath. CO2 was gradually intro-
placed inside the expansion chamber. Cyclosporine duced into the view cell at 5 bar increments. The system
particles precipitated using this technique were spherical was isolated and equilibrated for at least 30 min at each
with the primary particle size of 150 nm that were highly pressure interval. The melting point of cyclosporine was
aggregated. The effect of the pre-expansion temperature depressed to 25, 35, 40, 45, and 50 °C from 150 °C
and pressure on the characteristics of the precipitates was (normal melting point of cyclosporine) when exposed to
also investigated. The morphology and particle size of the CO2 at 53, 58, 60, 65 and 77 bar, respectively.
precipitates were not significantly affected, however, the The PGSS was performed by dissolving compressed
amount of cyclosporine precipitation was observed to CO2 in cyclosporine, an equilibrium cell containing the
increase as the system pressure was increased. cyclosporine was heated up to the required temperature
Lysozyme and lipase have been microencapsulated in and allowed to equilibrate. The system was then pres-
polymers by modifying RESS in a technique called ‘rapid surized with CO2 to melt the cyclosporine. The resultant
expansion of supercritical solution with a non-solvent solution was then expanded through a nozzle and the CO2
(RESS-N) [96]. The polymers studied included PEG, evaporated in the expansion chamber; cyclosporine
PLA, PLGA and PEG–poly(propylene glycol)–PEG particles were then collected for analysis. At all
S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029 1021

conditions of micronization ranging from 160 to 200 bar GAS has been used to process biomolecules such as
and 25 to 45 °C, cyclosporine was precipitated as lysozyme, insulin and myoglobin from organic
microspheres as small as 150 nm and was highly solvents [101–104]. The proteins were dissolved in
aggregated as observed by SEM. The average particle DMSO, MeOH, EtOH or DMF and the effect of the
size of the precipitates was not significantly affected as the solvents on particle size, morphology and biological
pre-expansion pressure increased from 180 to 200 bar. activity studied. For insulin, methanol produced the
The effect of nozzle diameter on the micronization of smallest particles (0.05–0.45 μm) while DMSO
cyclosporine was also assessed. With a 100-μm nozzle, produced the largest particles (1.4–8 μm). DMSO
the primary particle size increased from 150 nm to 1 μm. was used as the main solvent for lysozyme and the
This is believed to be due to a greater reduction in pressure effect of operating temperature studied. The highest
and density of the fluid at the exit of the nozzle [97]. It was activity retention was observed at 18–35 °C (92–
observed that the particles formed with the 100-μm 100%) while the lowest activity retention was
nozzle became porous upon the release of CO2 from the observed at 45 °C (60%). The particle size range was
cyclosporine-saturated phase during depressurization. similar for all temperatures (0.05–0.3 μm).
However, the particle size distribution measured by The principal disadvantage of this process is the
coulter and Andersen Cascade Impactor (ACI) was higher lack of control of particle formation. This has been
with median diameter of 4.5 μm and FPF of 59% observed to be true in batch operating conditions
(b 4.7 μm), respectively. because the level of saturation is not maintained [98].

5.3. GAS 5.4. ASES

In this process, the SF is added to the particle forma- This process involves pumping CO2 into a high-
tion vessel that contains the protein solution of interest pressure vessel until the system reaches the desired
[98]. The protein precipitates during the dissolution of the fixed conditions (pressure and temperature) [68]. The
SF in the solvent. The SF is generally introduced through protein solution is sprayed via an atomization device
the bottom of the vessel and bubbled through the protein into the vessel containing SFCO2. Particles are then
solution to achieve better mixing of the solvent and anti- collected on a filter at the bottom of the vessel [98]. In
solvent [68]. Once the protein has precipitated out, the this process, the anti-solvent concept of GAS remains
solvent–SF can be removed. The protein particles are but happens at the droplet level. This offers a
then washed with subsequent SF washes. Finally, the favourable higher anti-solvent to solvent ratio, an
pressure in the vessel is released and the protein powder increased surface area and mass transfer rate and hence,
recovered. an acceleration of the drying process. The rate is
The solvent in which the protein is dissolved should particularly fast when the operating pressure reaches
have a high solvent power for the protein, preferably be the mixture critical pressure. The process is then
soluble in the SF of choice and be compatible with the controlled by mixing of miscible fluids rather than
protein. Dimethyl sulfoxide (DMSO) and N,N-dimethyl mass transfer over interface of the droplets in the spray
formamide (DMFA) have been used since they satisfy [98]. When the miscibility of the fluids is poor,
the criteria of solvation of the protein and solubility in especially in systems containing water and CO2, the
SFCO2. However, there are concerns over the toxicity mass transfer requires special attention. The mass
and compatibility with protein molecules [99,100]. transfer can be improved by increasing the drying
Alternatives to SFCO2 have also been studied. medium to solvent ratio, decreasing the droplet size or
Supercritical ammonia and ethane have been used, the relative velocity between the solvent and drying
while ammonia produced completely denatured pro- medium [98]. This process enables production of
teins [102], ethane produced results comparable to protein particles with narrow size distribution, uniform
SFCO2 in terms of particle size and improved shape and desired physico-chemical characteristics
biological activity of insulin [102]. Whilst the perfor- [105,106].
mance of SF-ethane may be promising, its high ASES has been applied in producing insulin loaded
flammability will limit its use. PEG/PLA nanoparticles [107]. PLA and varying levels
1022 S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029

of 1900 Da PEG were used to prepare nanoparticles technique compared to other anti-solvent-based SCF
from DMSO. The organic solution of PEG, PLA and process. It is suitable for scaling up and manufacturing
bovine insulin was atomized through a 50-μm nozzle according to GMP requirements [25]. Furthermore, the
at a rate of 1 ml/min into a vessel containing the use of destructive organic solvents such as DMSO can
SFCO2. SEM of the particles indicated that lower PEG be avoided.
compositions produced well-separated nanoparticles. Lysozyme has been processed by SEDS with a
In contrast higher PEG compositions formed a micro-fine product showing a spherical morphology
continuous network structure. Light scattering analysis with free flowing powder handling properties [107].
showed that particles below 1 μm were obtained under When the retention of enzymatic activity of SEDS-
the adopted preparation condition. However, it was processed lysozyme was compared to those processed
observed that the addition of high amounts of low MW by spray drying and SFD, the SEDS-processed
PEG was essential to preparing products with high enzyme retained the highest enzymatic activity [107].
drug loading, high protein stability, effective drug Supercoiled plasma DNA (pDNA) has been pro-
release and solvent free product [107]. cessed by SEDS from aqueous solutions [108].
Increased yields pDNAwere obtained when the aqueous
5.5. SEDS feed containing mannitol was buffered to counteract the
acidic nature of the SFCO2 [107]. This indicates that by
In the earlier discussed ASES process, the mass using a buffered aqueous solution, the SEDS process
transfer of the SF into the sprayed droplet determines can be used to control the particle formation and protect
the rate of particle formation, while particle agglom- labile biological materials during processing.
eration and aggregation phenomena are influenced by Okamoto et al. [108] recently studied the stability of
the rate of solvent mass transfer into the SF from the a chitosan–pDNA complex powder prepared by a
droplet [89]. Mass transfer of SF into droplet is SEDS-type process for pulmonary delivery. An
dependent upon atomization efficiency while mass aqueous chitosan–pDNA complex solution containing
transfer of solvent into SF is dependent on dispersing mannitol was injected into the stream of a SFCO2/
and mixing phenomena between the solution droplet ethanol admixture to precipitate a gene powder. The
and the SF [107]. To minimize particle agglomeration obtained gene powder and gene solution (0.06 μg
often observed in ASES and other anti-solvent-based pDNA and 0.33% sodium dodecylsulfate aqueous
techniques, and to reduce drying times, increased mass solution) were placed in stability chambers at 25 or
transfer rates are required [89]. This has been 40 °C for 4 weeks. The integrity and transfection
successfully achieved by the SEDS process. In this potency of the gene were examined by electrophoresis
process, the drug solution and the SF are introduced and in vivo pulmonary transfection study in mice. The
into the particle formation vessel (where temperature process decreased the supercoiled DNA during the
and pressure are controlled) through a co-axial nozzle manufacturing; however, the decrease in the remaining
with a mixing chamber [68]. The high velocity of the supercoiled and open circular DNA in the powders
SF allows the production of droplets of very small during storage was much slower than in solutions. The
sizes, while the mixing of solvent with the SF inside powders had higher transfection potency than the
the mixing chamber leads to an increase of mass solutions containing the same amount of DNA. The
transfer of SF into the solvent and vice versa [68]. A effect of chitosan on the stability of DNA in solution
high mass transfer allows a faster nucleation and a was not obvious but it improved the stability of DNA
smaller particle size with less agglomeration [107]. in powders during manufacturing and storage.
SEDS has been further optimized to process water-
soluble compounds such as peptides and proteins by 5.6. PCA
introducing an organic solvent, the SCF, and the
aqueous solution as separate streams into a co-axial The PCA is a slight modification of the ASES
three-component nozzle. This helps in overcoming process. The process has been reported to be a one-step
problems associated with limited solubility of water in technique for effectively producing solvent free
SFCO2. SEDS is a more controllable and reproducible particles with a narrow size distribution at mild
S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029 1023

operating conditions [109,110]. It involves feeding and polymer. To start precipitation, the solvent flow
SFCO2 into a precipitator so as to pressurize the was stopped and the protein/polymer DMSO–water
precipitator to a desired value. Then the CO2 flow rate solution fed through the nozzle at the same flow rate as
into the precipitator is fixed. The solution of protein is the solvent. The mixture solvent–CO2 continuously
fed into the precipitator through a nozzle. The protein left the precipitator and the precipitated protein-loaded
particles are precipitated due to the anti-solvent effect particles collected on a 0.2 μm membrane filter. Once
of the SFCO2 [111]. the entire solution had been injected, the SFCO2
Fine protein particles of catalase and insulin have continued to flow through the vessel to wash out the
been prepared using the PCA process [112,113]. solvent. It was observed that working at 80 bar
Insulin was dissolved in DMSO or DMF and these (pressure in precipitation), a wet product was always
solutions were sprayed into a crystallizer with a observed over the filter plate so it was necessary to
continuous feed of SFCO2 [113]. The insulin powder increase the pressure to 90 bar to obtain a dry product.
formed had 90% particles with diameter less than 4 μm As pressure increased, the extraction of the liquid
and 10% less than 1 μm. it was also observed that the solvent to the supercritical phase takes place faster due
processed insulin maintained its biological activity. to a higher solubility of the solvent in the CO2. As
Protein-loaded N-trimethyl chitosan (TMC) micro- pressure increased, the atomization of the solution
spheres have been prepared by the PCA process [113]. produced smaller droplets and the drying time de-
It had been observed that using the PCA process to creased. The influence of water content of the DMSO–
precipitate proteins and polar polymers from DMSO water solution was also studied and it was observed
using CO2 as solvent has some difficulties [114]. that, as the water content of the solution increased, the
When operating below the mixture critical pressure of morphology changed from nanoparticles to a mixture
the DMSO–CO2, solution droplets formed by atom- of nanoparticles and microspheres. Water concentra-
ization in the nozzle dry slowly due to the low tions in the DMSO solution above 3.5% w/w produced
solubility of DMSO in the surrounding CO2 gas phase a wet product. The presence of water enhances the
[113]. A wet product or a film over the filter plate is mass transfer of SFCO2 through the droplet interface
often obtained when operating at these conditions due to the high solubility of the solution in the SFCO2.
[114]. On the other hand, proteins and polar polymers However, there seems to be an optimal water content
have an extremely low solubility in CO2 so when for the DMSO solution as above a certain water
working at conditions above the critical pressure of the concentration, solubility of the solvent mixture in the
mixture, high supersaturations are induced as soon as vapour phase decreases and the drying is no longer
the proteins or polymers come into contact with the efficient leading to a wet product [111].
CO2, in which case agglomerated nanoparticles are
formed [113]. It has been suggested that the addition of 6. Microcrystallization
a third component to the system solvent–CO2 modifies
its phase behaviour [115,116]. Perez de Diego et al. Crystalline protein particles have been found to be
used water to modify the phase behaviour of the more active and stable than their amorphous counter-
system DMSO–CO2 and solved the limitation of the parts [117,118]. The higher stability arises from the
PCA at conditions below the mixture critical pressure fact that, unlike crystalline phases, amorphous materi-
of the system solvent–CO2. This approach made it als consist of disordered arrangement of molecules and
possible to develop a process to produce protein- therefore possess no distinguishable crystal lattice
loaded TMC particles suitable for inhalation. SFCO2 [68]. Thermodynamically, the absence of crystallinity
was fed using into the precipitator to the desired vol- causes energy content higher than the crystalline state,
ume and pressure. The CO2 flow rate was then fixed at leading to lower stability and higher reactivity [68].
20 kg/h and the pressure in the precipitator automat- Amorphous protein particles are cleared rapidly from
ically regulated. DMSO was filtered and fed into the systemic circulation and are more susceptible to
precipitator using a Gilson piston pump to avoid the hydrolytic and enzymatic degradation because of
blockage of the nozzle at the beginning of the injection their higher reactivity [119]. These factors make
of the aqueous DMSO solution of protein (lysozyme) crystalline protein desirable as a fine pharmaceutical
1024 S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029

ingredient. Crystallization of proteins involves a one- microcrystals with a mean diameter of 3 μm were
step process and results in high purity. Crystallization prepared using this seed zone method. SEM micrograph
can improve protein handling during processing, showed the microcrystals to be of a homogenous
storage and delivery. It can also offer sustained release rhombohedral shape with some rhombus forms, without
of the therapeutic agent for an effective duration by aggregates. Following the administration of 32 U/kg of
changing the dissolution characteristics [120]. the microcrystal suspension to streptozotocin-induced
Despite these advantages, apart from insulin, few diabetic rats by intratracheal instillation, the blood glucose
crystalline forms of proteins have been used as levels were reduced and hypoglycemia was prolonged
pharmaceutical active ingredient, even though over over 13 h as compared to the normal insulin solution
90% of pharmaceutical products that contain a drug in [123]. It was suggested that the sustained release effect
particulate form, do so as a crystalline form [121]. This is was due to the decreased solubility of the microcrystals.
because proteins being large and flexible are difficult to
crystallize [119]. Furthermore, crystallization can lead to 6.2. Polymorphism
particles with a wide size distribution. Milling has been
applied to size reduce crystallized protein to respirable Polymorphism is the propensity of a substance to
size, but milling may lead to a high energy input leading crystallize into more than one crystal structure [68]. This
to particles of reduced crystallinity and may contain is a potential problem for the crystallization of
disordered regions, thereby introducing regions of biopharmaceuticals for pulmonary delivery as regulatory
reduced stability [122]. bodies only approve a specific crystal structure or
The concept of microcrystallization has been used polymorph [128]. During crystallization, different poly-
to overcome milling-induced disorder in crystalline morphs of a compound may be formed [68]. The more
powders. stable polymorph has the higher molecular packing
Lee et al. [119] have succeeded in producing density and has the lower values for: Gibb's free energy,
microcrystals of α-lactalbumin, a 16 kDa glycoprotein. fugacity, vapour pressure, thermodynamic activity,
α-Lactalbumin was dissolved in 0.1 N acetic acid dissolution rate per unit area in any solvent and rate of
containing PEG-8000 as a stabilizer. The pH of the reaction, including decomposition rate [68]. For thermo-
protein solution was adjusted to about 3.0. 10 N, NaOH dynamic reasons, the less stable polymorph would want
solution was added rapidly to the protein solution to to convert to the stable one. The phase transition can
adjust pH to about 4.0 (at this pH, the protein has lowest occur through for different mechanisms: solid–solid
solubility). The supersaturated protein solution was transition, melting, solution and solution mediated [129].
stored at 16 °C. The microcrystals formed were roughly The kinetics of the phase transitions can be influenced by
spherical and about 1–2 μm in diameter. These spherical typical environmental parameters (temperature, pressure,
crystals have been applied for pulmonary delivery [123]. relative humidity), presence of crystalline defect, impu-
rities and mechanical stress [68].
6.1. Microcrystallization and sustained release Polymorph purity is therefore an important parameter
to consider in a drug product since the presence of
The challenge presently confronting pulmonary differing crystal phases can accelerate the conversion
delivery of protein is the short duration of action obtained process by lowering the relevant activation energy
via this route due to fast rate of clearance via the barrier [130]. The phase transformation could lead to a
mucocilliary apparatus and alveolar macrophages different polymorph with unwanted physico-chemical
[124,125]. Different methods of prolonging insulin properties. For this reason, regulatory authorities have
absorption in the lung have been proposed such as long recognized the need to limit polymorphic impuri-
microencapsulation using a biodegradable polymer, but ties in pharmaceutical materials [130].
problem is perceived with the accumulation of these
polymers in the lung [126] and loss of insulin activity 6.3. Controlling polymorphism with SF
during the preparation of microspheres [127].
Recently, a unique insulin microcrystallization process The identification of stable polymorphs with desired
using a seed zone method was developed [123]. Insulin physical properties is very important for product
S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029 1025

development. Before a drug is submitted for review to the the aspect ratio will only increase the dae very small
regulatory authorities, it is important to identify the most amounts [49]. This means that dae can be made small as
stable or suitably stable polymorph of such a drug and long as the short axis remains small (e.g. ≤1 μm) [49].
work with it. The solid phase must be monitored, es-
pecially after processes that are potentially able to modify 8. Conclusion
the solid-state properties of the components [68]. For
instance, during micronization processes such as milling, The emergence of advanced particle engineering
spray drying and spray freeze drying, the substance is techniques coupled with the modification of the
usually exposed to mechanical stress, contact with sol- traditional methods such as milling has contributed
vents, heating–cooling cycles that can often lead to to the increased possibility of formulating biopharma-
alteration of the solid phase, such as new polymorph ceutical for pulmonary delivery. Particles with aerosol
formation, dehydration or melting mechanism [68]. properties suited for deep lung delivery have been
The SEDS process has proved to be an effective engineered without destroying the biological activity
technique in obtaining pure polymorphs of different of these sensitive molecules. The wide range of
drugs based on different operating conditions and techniques currently available or being developed
crystallization kinetics [131]. The rapid drying and coupled with the increasing knowledge of excipient
cooling experienced in SEDS exclusively gives rise to use for the protection of biopharmaceuticals now
a polymorph of a drug [68]. allows a diverse range of biopharmaceuticals to be
processed for use in inhalation delivery devices. The
7. Aerodynamic particle diameter challenge to its development is that scientist may now
shift from “can the molecule be processed” to “which
Aerodynamic diameter (dae) particle size is a key process should be used” since it is yet to be shown if
parameter determining the aerosol performance of an any one technique can provide the solution to all types
inhaled powder [49]. Unlike the physical particle of molecule.
diameter, dae is a concept incorporating the size, shape,
and density of particle [27]. For a solid spherical particle, References
dae is simply equal to the physical diameter (d) multiplied
by the square root of the particle density (ρ) [49]. [1] G. Walsh, Biopharmaceuticals and biotechnology medicines: an
issue of nomenclature, Eur. J. Pharm. Sci. 15 (2002) 135–138.
pffiffiffi [2] G. Walsh, Second generation biopharmaceuticals, Eur. J.
dae ¼ ð q Þd ð1Þ Pharm. Biopharm. 58 (2004) 185–196.
[3] I. Johnson, Human insulin from recombinant DNA technol-
This means for a given density, small particles have ogy, Science 291 (2004) 632–637.
lower aerodynamic diameter compared to larger parti- [4] J. Fielder, N. Wagner, S. Grammaticos, H. Hoffman, H.
cles. However porous particles because of their low Kaufmann, R. Otto, Use of flow-cytometric analysis to optimize
density would have low dae based on Eq. (1). cell banking strategies for production of biopharmaceuticals from
mammalian cells, J. Biotechnol. 120 (2005) 111–120.
Nektar Therapeutics has adopted this porous [5] G. Walsh, Biopharmaceuticals: recent approvals and likely
particle approach in the formation of their pulmo- directions, Trends Biotechnol. 23 (2005) 553–558.
sphere® [132]. The physical diameter (d) is mostly [6] Y. Bagger, L.B. Tanko, P. Alexadersen, M.A. Karsdal, M.
measured using laser diffraction technique [133]. Olson, L. Mindeholm, M. Azria, C. Christiansen, Oral salmon
calcitonin induced suppression of urinary collagen type II
Characterizing the dae for non-spherical particles such
degradation in post menopausal women: a new potential
as elongated or needle-like object is quite challenging as treatment of osteoarthritis, Bone 33 (2005) 425–430.
Eq. (1) cannot be used. It has been discovered that the dae [7] Y.J. Zheng, M. Fulu, Decrease of genital organ weights and
of elongated particles is controlled by the short axis rather plasma testosterone in rat following oral administration of
than the long axis [47]. It was found that dae increases leuprolide microemulsion, Int. J. Pharm. 307 (2006) 209–215.
with the aspect ratio (i.e. ratio of the long axis to the short [8] C. Cullander, R.H. Guy, Routes of delivery: case studies (6),
transdermal delivery of peptides and proteins, Adv. Drug
axis) of an elongated particle. When the aspect ratio is Deliv. Rev. 8 (1992) 291–329.
greater than 15 or 20, dae will be about two or three times [9] P. Edman, E. Bjork, Routes of delivery: case studies (1), nasal
the short axis (physical diameter). Any further increase in deliveryofpeptidedrugs,Adv.DrugDeliv.Rev. 8(1992)165–177.
1026 S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029

[10] D. Harris, J.H. Liaw, J.R. Robinson, Routes of delivery: case [29] R.H. Snow, B.H. Kaye, C.E. Capes, G.C. Sresty, Size
studies (7) ocular delivery of peptide and protein drugs, Adv. reduction and size enlargement, in: R.H. Perry, D. Green
Drug Deliv. Rev. 8 (1992) 331–339. (Eds.), Perry's Chemical Engineer's Handbook, McGraw
[11] N.F.N. Ho, C.L. Barsutin, P.S. Burton, H.P. Merckle, Routes Hill, New York, 1984, pp. 1–59.
of delivery: case studies (3), mechanistic insights to buccal [30] L.H. Van Vlack, Elements of Materials Science and Engineer-
delivery of proteinaceous substance, Adv. Drud Deliv. Rev. ing, Addison Wesley Publishing, Reading, 1980, pp. 185–208.
8 (1992) 197–235. [31] A. Adjei, Pulmonary bioavailability of peptide drugs: animal
[12] P.L. Smith, D.A. Wall, C.H. Gochoco, G. Wilson, Routes of models, preclinical studies and clinical studies, Presented at
delivery: case studies (5), oral absorption of peptides and Respiratory Drug Delivery II, Keystone, Co, 1990.
proteins, Adv. Drug Deliv. Rev. 8 (1992) 253–290. [32] W. Schlocker, S. GschlieBer, A. Bernkop-Schniirch, Evalu-
[13] V. Codrons, F. Vanderbist, R.K. Vanbeeck, M. Arras, D. ation of the potential of air jet milling of solid protein–poly
Lison, V. Preat, R. Vanbever, Systemic delivery of parathyroid (acrylate) complexes for microparticle preparation, Euro
hormone (1–34) using inhalation dry powder in rats, J. Pharm. J. Pharm. Biopharm. 62 (2006) 260–266.
Sci. 92 (2003) 938–948. [33] G.S. Zijlstra, W.L.J. Hinrichs, A.H. deBoer, H.W. Frijlink,
[14] M.A. Hollinger, Respiratory pharmacology and toxicology, The role of particle engineering in relation to formulation and
W. B., Saunders, Philadelphia, 1994, pp. 1–20. de-agglomeration principle in the development of a dry
[15] P.R. Byron, Determinants of drug and polypeptide bioavail- powder formulation for inhalation of cetrorelix, Eur. J. Pharm.
ability from aerosols delivered to the lung, Adv. Drug Deliv. Sci 23 (2004) 139–149.
Rev. 5 (1990) 107–132. [34] A. Adjei, D. Sundberg, J. Miller, A. Chin, Bioavailability
[16] T.S. Purewal, Formulation of metered dose inhalers, in: T.S. of leuprolide acetate following nasal and inhalation deli-
Purewal, D.J.W. Grant (Eds.), Metered Dose Inhaler Tech- very to the rats and healthy humans, Pharm. Res. 9 (1992)
nology, Interpharm, Illinois, 1998, pp. 9–68. 244–249.
[17] U.B. Kompella, V.H. Lee, Delivery systems for the [35] K.G. Backstrom, C.M. Dahlback, P. Edman, A.C.B. Johans-
penetration enhancement of peptide and protein drugs: design son, Therapeutic preparation for inhalation. International
consideration, Adv. Drug Deliv. Rev. 46 (2001) 211–245. Patent Application (1995) WO 95/00127.
[18] S.P. Sorokin, A morphological and cytochemical study of the [36] K. Tomihisa, Grinding of rubber below the brittle temperature
great alveolar cell, J. Histochem. Cytochem. 14 (1961) 884–897. with supersonic jet air. Japanese Patent (1973) JP73103614.
[19] L. Garcia-Conteras, L.D. Jones, A.J. Hickey, Alveolar [37] S. Kobayashi, S. Kondo, K. Juni, Pulmonary delivery of
macrophages as a target for drug delivery, Res. Drug Deliv. salmon calcitonin dry powders containing absorption
IX (2004) (California). enhances in rats, Pharm. Res. (1996) 80–83.
[20] J. Liu, H. Wong, J. Moselly, B. Bowen, X.Y. Wu, M.R. [38] W.L.J. Hinrichs, N.N. De Smelt, J. Demeester, H.W. Frijlink,
Johnston, Targeting colloidal particulates to thoracic lymph Insulin is a promising cryo- and lyoprotectant for PEGylated
nodes, Lung Cancer 51 (2006) 377–386. lipoplexes, J. Control. Release 103 (2005) 465–479.
[21] K. Corkery, Inhalable drugs for systemic therapy, Respir. Care [39] S. Ohtake, C. Schebor, S.P. Palecek, J.J. de Pablo, Phase
45 (2000) 831–835. behaviour of freeze-dried phospholipid–cholesterol mixtures
[22] A. Hussain, J.J. Arnold, M.A. Khan, F. Ahsan, Absorption stabilized with trehalose, Biochim. Biophys. Acta, Biomembr.
enhancers in pulmonary delivery, J. Control. Release 94 1713 (2005) 57–64.
(2004) 15–24. [40] R.M. Platz, J. Utsum, Y. Satoh, N. Naruse, Pharmaceutical
[23] M. Hanna, P. York. Method and apparatus for the formation of aerosol formulation of solid polypeptide microparticles and
particles. European Patent 0706421B1, 1998. method for the preparation thereof. International Patent
[24] E.A.Quinn, R.T.Forbes,A.C.Williams,M.J.Oliver, L.McKenzie, Application (1991) WO 91/16038.
T. Purewal, Protein conformational stability in the hydro- [41] R.M. Platz, A. Ip, C.L. Whitham, Process for preparing
fluoroalkane propellants tetraflouroethane and heptafluoropro- micronized polypeptide drugs. US Patent (1994) 5354562.
pane analysed by Fourier transform Raman spectroscopy, Int. [42] A.L. Adjei, E.S. Johnson, J.W. Kesterson, LHRH analog
J. Pharm. 186 (1999) 31–41. formulations, US Patent (1990) 4897256.
[25] K.A. Johnson, Preparation of peptide and protein powders for [43] R. Lizio, M. Damm, A.W. Sarlikiotis, H.H. Bauer, C.M. Lehr,
inhalation, Adv. Drug Deliv. Rev. 26 (1997) 3–15. Low temperature micronization of a peptide drug in a fluid
[26] E. Phillips, E. Allsopp, T. Christensen, M. Fitzgerald, L. Zhao, propellant: case study cetrorelix, AAPS PharmSciTech 2
Size reduction of peptides and proteins by jet milling, in: R.N. (2001) 1–7.
Dalby, P.R. Byron, S.J. Farr (Eds.), Proceeding of RDD, vol. VI, [44] M. Irngartinger, V. Camuglia, M. Damm, J. Goede, J. Frijlink,
Interpharm, Buffalo, 1998, pp. 161–168. Pulmonary delivery of therapeutic peptides via dry powder
[27] R.C. Willis, Crystallize to deliver, Mod. Drug Discov. (2003) inhalation: effects of micronisation and manufacturing, Eur. J.
12 (March). Pharm. Biopharm. 58 (2004) 7–14.
[28] G.W. Hallworth, The formulation and evaluation of pressur- [45] K. Masters, Spray Drying Handbook, Longman Scientific and
ized metered dose inhalers, in: D. Ganderton, T.M. Jones Technical, Essex, England, 1991.
(Eds.), Drug Delivery to the Respiratory Tract, Ellis Hor- [46] J. Broadhead, S.K.F. Rouan, I. Hau, C.T. Rhodes, The effect
wood, Chistester, 1987, pp. 87–118. of process and formulation variable on the properties of spray
S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029 1027

dried β-galactosidase, J. Pharm. Pharmacol. 46 (1994) [62] M. Adler, M. Unger, G. Lee, Surface composition of spray
458–467. dried particles of bovine serum albumin/trehalose/surface,
[47] P. Labrude, M. Rasolomanana, C. Vigneron, C. Thiron, B. Pharm. Res. 17 (2000) 863–870.
Chailott, Protective effect of sucrose on spray drying of [63] C.L. Leach, J.S. Patton, K.M. Perkins, M.C. Kwo, B. Bueche,
oxyhemoglobin, J. Pharm. Sci. 78 (1989) 223–229. L. Guo, M.D. Bentley, Dry powder formulation of inhaled
[48] H. Brandenberger, Best@buchi evaporation, Inf. Bull. 27 PEG-Insulin yields prolonged systemic activity in dogs,
(2003). Presented at the American Diabetes Association 63rd
[49] H.K. Chan, Dry powder aerosol drug delivery – opportunities Scientific Session, June, Louisiana, New Orleans, 2003.
for colloid and surface scientists, Colloids and Surfaces A: [64] J. Elversson, A. Millqvist-Fureby, Aqueous two-phase system
Physicochem. Eng. Aspects 284–285 (2006) 50–55. as a formulation concept for spray-dried protein, Int. J. Pharm.
[50] B. Bittner, T. Kissel, Ultra atomization for spray drying: a versatile 294 (2005) 73–87.
technique for the preparation of protein loaded biodegradable [65] L. Dellamary, D.J. Smith, A. Bloom, S. Bot, G.–R. Guo, M.
microspheres, J. Microencapsul 16 (1999) 325–341. Costello, A. Bot, Rational design of solid aerosols for immu-
[51] Y.F. Maa, P.A. Nguyen, J.D. Andya, T.D. Sweeney, S.J. Shire, noglobulin delivery by modulation of aerodynamic and release
C.C. Hsu, Effects of spray drying and subsequent processing on characteristics, J. Control. Release 95 (2004) 489–500.
residual moisture content and physical/biochemical stability of [66] A. Grenha, B. Seijo, C. Remunan-Lopez, Microencapsulated
protein inhalation powders, Pharm. Res. 15 (1998) 768–775. chitosan nanoparticles for lung protein delivery, Eur. J. Pharm.
[52] J.S. Patton, L.C. Foster, R.M. Platz, Methods and composition Sci. 25 (2005) 427–437.
for pulmonary delivery of insulin. International patent [67] P. Calvo, C. Remunan-Lopez, J.L. Vita-Jato, M.J. Alonso,
application, WO95/ 24183. Novel hydrophilic chitosan–polyethylene oxide nanoparti-
[53] R.W. Niven, F.D. Lott, A.Y. Ip, J.M. Cribbs, Pulmonary cles as protein carriers, J. Appl. Polym. Sci. 63 (1997)
delivery of powders and solutions containing recombinant 125–132.
human granulocyte colony-stimulating factor, Pharm. Res 11 [68] I. Pasquali, R. Bettini, F. Gordono, Solid-state chemistry and
(1994) 1101–1109. particle engineering with supercritical fluids in pharmaceu-
[54] A.D. Sutton, R.A. Johnson, P.J. Senior, D. Heath, Preparation tics, Eur. J. Pharm. Sci. 27 (2006) 299–310.
of diagnostic agents. European patent application (2000) [69] R.E. Sievers, U. Karst, P.D. Milewski, S.P. Sellers, B.A.
0681843A2. Miles, J.D. Schaefer, C.R. Stoldt, C.Y. Xu, Formation of
[55] S. Willaya-Areekul, G.F. Needham, N. Milton, M.L. Roy, S.L. aqueous small droplet aerosol assisted by supercritical carbon
Nail, Freeze-drying of tert-butanol/water co-solvent system: a dioxide, Aerosol Sci. Technol. 30 (1999) 3–15.
case report on formation of a friable freeze dried powder of [70] R.E. Sievers, U. Karst, J.D. Schaefer, C.R. Stoldt, B.A.
tobramycin sulphate, J. Pharm. Sci. 91 (2002) 1147–1155. Watkins, Supercritical CO2-assisted nebulization for the
[56] H. Chan, D. Lechuga-Ballesteros, R. Vehring, W.R. Foss, J.C. production and administration of drugs, J. Aerosol Sci. 27
Feeley, A. Clark, Effect of bulk and surface properties on the (1996) S497–S498.
aerosol performance of dry powders for inhalation, Presented [71] E. Reverchon, I. De Marco, G. Della Porta, Rifampicin
at 2002 AAPS, Annual Meeting and Exposition, Nov. 10–14, microparticles production by supercritical anti-solvent pre-
Canada, Toronto, Ontario, 2002. cipitation, Int. J. Pharm. 243 (2002) 83–91.
[57] H.K. Chan, A. Clark, I. Gonda, M. Mumenthaler, C. Hsu, [72] R.E. Sievers, B.A. Miles, S.P. Sellers, P.D. Milewski, K.D.
Spray dried powders and powder blends of recombinant Kusek, New process for manufacture of 1-micron spherical
human deoxyribonuclease (rhDNase) for aerosol delivery, drug particles by CO2-assisted nebulization of aqueous
Pharm. Res. 14 (1997) 431–437. solutions, Proceeding of Resp. Drug Deliv., VI, South
[58] Y.–H. Liao, M.B. Brown, S.A. Jones, T. Nazir, G.P. Martin, Carolina, 1998, pp. 417–419.
The effect of polyvinyl alcohol on the in-vitro stability and [73] S.P. Sellers, G.S. Clark, R.E. Sievers, J.F. Carpenter, Dry pow-
delivery of spray-dried protein particles from surfactant-free ders of stable protein formulations from aqueous solutions
HFA 134a-based pressurised metered dose inhalers, Int. J. prepared using supercritical CO2-assisted aerosolization,
Pharm. 304 (2005) 29–39. J. Pharm. Sci. 90 (2001) 785–797.
[59] V. Codrons, F. Vanderbist, R. Verbeeck, A. Mohammed, D. [74] R.E. Sievers, E.T.S. Huang, J.A. Villa, T.R. Walsh, H.V.
Lison, V. Preat, R. Vanbever, Systemic delivery of parathyroid Meresman, C.D. Liang, S.P. Cape, Rapid gentle drying using
hormone (1–34) using inhalation dry powder in rats, J. Pharm. dense carbon dioxide to form fine dry powders, Proceed-
Sci. 92 (2003) 938–950. ings of Resp Drug Deliv: VIII. Tucson, Arizona, 2002,
[60] J.D. Andya, Y.F. Maa, H.R. Costantino, P.A. Nguyen, N. pp. 675–677.
Dasovich, T.D. Sweeney, C.C. Hsu, S.J. Shire, The effect of [75] R.E. Sievers, E.T.S. Huang, J.A. Villa, J.K. Kawamoto, M.M.
formulation excipients on protein stability and aerosol per- Evans, P.R. Brauner, Low temperature manufacturing of fine
formance of spray dried powders of a recombinant humanized pharmaceutical powders with supercritical fluid aerosolization
anti-IgE monoclonal antibody, Pharm. Res. 16 (3) 350–358. in bubble dryer, Pure Appl. Chem. 73 (2001) 1299–1303.
[61] C. Bosquillon, V. Preat, R. Vanbever, Pulmonary delivery of [76] R.N. Dalby, V. Naini, P.R. Byron, Droplet drying and electro-
growth hormone using dry powders and visualization of its static collection, a novel alternative to conventional commu-
local fate in rats, J. Control. Release 96 (2004) 233–244. nition techniques, J. Biopharm. Sci. 3 (1992) 91–99.
1028 S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029

[77] W.R. Gombotz, M.S. Healy, L.R. Brown, H.E. Auer, Process for [94] V. Krukonis, Supercritical fluid nucleation of difficult to
producing small particles of biologically active molecules, comminute solids, Proceedings of the Annual Meeting
European Patent Application (1994) EP0432232B1. AlChE, vol. 140 F, 1984, p. T-214.
[78] Y.F. Maa, P.A. Nguyen, T. Sweeny, S.J. Shire, C.C. Hsu, [95] E.M. Phillips, V.J. Stella, Rapid expansion from supercritical
Protein inhalation powders. Spray drying vs spray freeze solutions: applications to pharmaceutical processes, Int. J.
drying, Pharm. Res. 16 (1999) 249–254. Pharm. 94 (1993) 1–10.
[79] H.R. Constantino, L. Firouzabadian, K. Hogeland, C. Wu, C. [96] K. Mishinma, K. Matsuyama, D. Tanabe, S. Yamauchi, T.J.
Banganski, C.M. Cordova, K.G. Carrasquillo, K. Griebenow, Young, K.P. Johnston, Microencapsulation of protein by rapid
S.E. Zale, M.A. Tracy, Protein spray freeze-drying. Effect of expansion of supercritical solution with a non solvent,
atomization conditions on particle size and stability, Pharm. Material, Interfaces and Electrochemical Phenomena,
Res. 17 (2000) 1374–1384. AlChE Journal, vol. 46, 2000, pp. 857–865.
[80] H.R. Constantino, L. Firouzabadian, C. Wu, K.G. Carras- [97] T.J. Young, S. Mawson, K.P. Johnstone, I.B. Henriksen, G.W.
quillo, K. Griebenwo, S.E. Zale, M.A. Tracy, Protein spray Pace, A.K. Mishra, Rapid expansion from supercritical to
freeze-drying: 2. Effect of formulation variables on particle aqueous solution to produce submicron suspensions of water-
size and stability, J. Pharm. Sci. 91 (2002) 388–395. insoluble drugs, Biotechnol. Prog. 16 (2000) 402–407.
[81] S.D. Webb, S.T. Golledge, J.T. Cleland, J.F. Carpenter, T.W. [98] N. Jovanovic, A. Bouchard, G.W. Hofland, G. Witkamp, D.J.
Randolph, Surface adsorption of recombinant human interferon- Crommelin, W. Jiskoot, Stabilization of proteins in dry
ã in lyophilized and spray-lyophilized formulations, J. Pharm. powder formulations using supercritical fluid technology,
Sci. 91 (2002) 1474–1487. Pharm. Res. 21 (2004) 1955–1969.
[82] M.C. Heller, J.F. Carpenter, T.W. Randolph, Protein formu- [99] S.D. Yeo, G.B. Lim, P.G. Debenedetti, H. Bernstein, Formation
lation and lyophilization cycle design: prevention of damage of microparticulate protein powders using a supercritical fluid
due to freeze-concentration induced phase separation, Bio- anti-solvent, Biotechnol. Bioeng. 41 (1993) 341–346.
technol. Bioeng. 63 (1999) 166–174. [100] M. Jackson, H.H. Mantsch, Beware of proteins in DMSO,
[83] R.A. DePaz, D.A. Dale, C.C. Barnett, J.F. Carpenter, A.C. Biochim. Biophys. Acta 1078 (1991) 231–235.
Gaertner, W. Randolph, Effects of drying methods and [101] R. Thiering, F. Dehghani, N.R. Foster, Micronization of
additives on the structure, function and storage stability of model proteins using compressed CO2, Proceedings of the 5th
subtilisin: role of protein conformation and molecular International Symposium on Supercritical Fluids, Atlanta,
mobility, Enzyme Microb. Technol. 31 (2002) 765–774. 2000.
[84] Z. Yu, K.P. Johnston, R.O. Williams III, Spray freezing into [102] N.R. Foster, H.L. Regtop, F. Dehghani, R.T. Bustami, H.–K.
liquid versus spray-freeze drying: influence of atomization on Chan, Synthesis of small particles, WO Patent No: 0245690.
protein aggregation and biological activity, Eur. J. Pharm. Sci. [103] R. Thiering, F. Dehghani, A. Dillow, N.R. Foster, The
27 (2006) 9–18. influence of operating conditions on the dense gas precipi-
[85] T.L. Roger, A.C. Nelsen, J. Hu, J.N. Brown, M. Sarkari, T.J. tation of model proteins, J. Chem. Technol. Biotechnol. 75
Young, K.P. Johnston, R.O. William III, A novel particle (2000) 29–41.
engineering technology to enhance dissolution of poorly [104] S.D. Yeo, P.G. Debenechi, M. Radosz, H.W. Schmidt,
water soluble drugs: spray-freezing into liquid, Eur. J. Pharm. Supercritical anti-solvent process for substituted para-linked
Biopharm. 54 (2002) 271–280. aromatic polyamides: phase equilibrium and morphology
[86] Z. Yu, T.L. Rogers, J. Hu, K.P. Johnston, R.O. Williams III, study, Macromolecules 26 (1993) 6207–6210.
Preparation and characterization of microparticles containing [105] S.D. Yeo, G.B. Lim, P.G. Debenedetti, H. Bernstein, Formation
peptide produced by a novel process: spray freezing into of microparticulate protein powders using a supercritical fluid
liquid, Eur. J. Pharm. Biopharm. 54 (2002) 221–228. anti-solvent, Biotechnol. Bioeng. 41 (1993) 341–346.
[87] Z. Yu, A.S. Garcia, K.P. Johnston, R.O. Williams, Spray [106] S. Palakodaty, P. York, Phase behaviour effects on particle
freezing into liquid for highly stable protein nanostructured formation process using supercritical fluids, Pharm. Res. 16
microparticles, Eur. J. Pharm. Biopharm. 58 (2004) (1999) 976–985.
529–537. [107] P. Caliceti, S. Salmaso, N. Elvassore, A. Bertucco, Effective
[88] M. Henczka, J. Baldyga, B.Y. Shekunov, Modelling of spray protein release from PEG/PLA nanoparticles produced by
freezing with compressed CO2, Chem. Eng. Sci. 61 (2006) compressed anti-solvent precipitation techniques, J. Control.
2880–2887. Release 94 (2004) 195–205.
[89] P. York, Strategies for particle design using supercritical fluid [108] H. Okamoto, Y. Sakamura, K. Shiraki, K. Oka, S. Nishida, H.
technologies, PSST 2 (1999) 430–440. Todo, K. Lida, K. Danjo, Stability of chitosan–pDNA
[90] J.W. Tom, P.G. Debenedetti, J. Aerosol Sci. 22 (1999) 555–584. complex powder prepared by supercritical CO2 process, Int.
[91] W.J. Schmitt, R.C. Reid, Fluid Phase Equilib. 32 (1986) 77–99. J. Pharm 290 (2005) 73–81.
[92] E. Weidner, High-pressure chemical engineering, Process [109] R. Bodmeier, H. Wang, D.J. Dixon, S. Mawson, K.P.
Technol, Proceedings, vol. 12, Elservier, 1996. Johnston, Polymeric microspheres prepared by spraying into
[93] A. Tandya, F. Dehghani, N.R. Foster, Micronization of compressed CO2, Pharm. Res. 12 (1995) 1211–1217.
cyclosporine using dense gas technique, J. Supercrit. Fluids [110] R.F. Falk, W. Th. Randolph, Process variable implications for
37 (2006) 272–278. residual solvent removal and polymer morphology in the
S.A. Shoyele, S. Cawthorne / Advanced Drug Delivery Reviews 58 (2006) 1009–1029 1029

formation of gentamycin-loaded poly(L-lactide) microparti- [124] J.S. Skyler, R.A. Gelfand, I.A. Koirides, Treatment of type 2
cles, Pharm. Res. 15 (1998) 1233–1237. diabetes mellitus with inhaled human insulin: a 3 month,
[111] Y. Perez deDiego, H.C. Pellikaan, F.E. Wubbolts, G. multicenter trial (abstract), Diabetes 47 (1998) A61 (suppl.).
Borchard, G.J. Witkamp, P.J. Jansens, Opening new operating [125] W.T. Cefalu, R.A. Gelfand, I.A. Koirides, Treatment of type 2
windows for protein micronization using the PCA process, diabetes mellitus with inhaled insulin: a 3 month, multicenter
J. Supercrit. Fluids 36 (2006) 216–224. trial (abstract), Diabetes 47 (suppl.) (1998) A61.
[112] P.G. Debenedetti, G. Lim, R.K. PrudHomme, Formation of [126] J.S. Patton, J. Bukar, S. Nagarajan, Inhaled insulin, Adv. Drug
protein microparticles by antisolvent precipitation. European Deliv. Rev. 35 (1999) 235–247.
patent application EP052314A1, 1993. [127] A. Sanchez, B. Villamayor, Y. Guo, J. Mclver, M.J. Alonso,
[113] S. Yeo, G. Lim, P.G. Debenedetti, H. Bernestein, Formulation Formulation strategies for the stabilization of tetanus toxoid in
of microparticulate protein powders using a supercritical fluid poly(lactide co-glycolide) microspheres, Int. J. Pharm. 185
antisolvent, Biotechnol. Bioeng. 41 (1993) 341–346. (1999) 255–266.
[114] Y. Perez de Diego, M.B.P. van Rooijen, F.E. Wubbolts, P.J., [128] R.J. Davey, N. Blagden, G.D. Potts, R. Docherty, Polymor-
Effect of process parameters on microparticle formation by phism in molecular crystals: stabilization of a metastable form
precipitation with compressed anti-solvent (PCA), Proceed- by conformational mimicry, J. Am. Chem. Soc. 199 (2000)
ings of Eighth International Workshop on Industrial Crystal- 1767–1772.
lization, 2001, pp. 98–104. [129] G.G.Z. Zhang, D. Law, E.A. Schmilt, Y. Qiu, Phase
[115] E. Reverchon, I. DeMarco, Supercritical antisolvent micro- transformation considerations during process development
nization of cefonicid, thermodynamic interpretation of results, and manufacture of solid oral dosage forms, Adv. Drug Deliv.
J. Supercrit. Fluids 31 (2004) 207–215. Rev. 56 (2004) 371–390.
[116] A. Weber, I.V. Yelash, T. Kraska, Effect of the phase [130] P. York, Particle design, a ‘supercritical’ issue for the pharma
behaviour of the solvent–anti-solvent systems on the gas industry, Pharm. Vision, (200) 28–30.
anti-solvent crystallization of paracetamol, J. Supercrit. Fluids [131] A.D. Edward, B.Y. Shekunov, A.I. Kordikowski, R.T. Forbes,
33 (2005) 107–113. P. York, Crystallization of pure anhydrous polymorphs of
[117] A.A. Elkordy, R.T. Forbes, B.W. Barry, Integrity of crystalline carbamazepine by solution enhanced dispersion with super-
lysozyme exceeds that of a spray dried form, Int. J. Pharm. critical fluids (SEDS), J. Pharm. Sci. 90 (2001) 1115–1124.
247 (2002) 79–90. [132] S.P. Duddu, S.A. Sick, Y.H. Walter, T.E. Tarara, T.R.,
[118] A.A. Elkordy, R.T. Forbes, B.W. Barry, Stability of crystallised Improved lung delivery from a passive dry powder inhaler
and spray dried lysozyme, Int. J. Pharm. 278 (2004) 209–219. using an engineered pulmosphere powder, Pharm. Res. 19
[119] M.J. Lee, J.–H. Kwon, J.–S. Shin, C.–W. Kim, Microcrys- (2002) 689–695.
tallization of α-lactalbumin, J. Crystal Growth 282 (2005) [133] A.H. de Boer, D. Gjaltema, P. Hagedoom, M. Schaller, W.
434–437. Witt, H.W. Frijlink, Design and application of a new modular
[120] A. Jen, H.P. Merkle, Pharm. Res. 18 (2001) 1483. adapter laser diffraction characterisation of inhalation aero-
[121] C. Valder, D. Merrifield, Pharmaceutical Technology, Smithk- sols, Int. J. Pharm. 249 (2002) 233–245.
line Beecham R&D News, vol. 32, 1996, p. 1. [134] C. Vemavarapu, M.J. Mollan, M. Lodaya, T.E. Needham,
[122] I. Krycer, J.A. Hersey, A comparative study of communica- Design and process aspect of laboratory scale SCF particle
tion in rotatory and vibratory ballmills, Powder Technol. 27 formation systems, Int. J. Pharm. 292 (2005) 3.
(1980) 137–141.
[123] J.–H. Kwon, B.–H. Lee, J.–J. Lee, C.W. Kim, Insulin
microcrystal suspension as a long acting formulation for
pulmonary delivery, Eur. J. Pharm. Sci. 22 (2004) 107–116.

Das könnte Ihnen auch gefallen