Sie sind auf Seite 1von 21

International Journal of Pharmaceutics 546 (2018) 194–214

Contents lists available at ScienceDirect

International Journal of Pharmaceutics


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

Review

Nano spray drying for encapsulation of pharmaceuticals T


a b b c c,⁎
Cordin Arpagaus , Andreas Collenberg , David Rütti , Elham Assadpour , Seid Mahdi Jafari
a
NTB University of Applied Sciences of Technology Buchs, Institute for Energy Systems, Buchs, Switzerland
b
BÜCHI Labortechnik AG, Flawil, Switzerland
c
Department of Food Materials and Process Design Engineering, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran

A R T I C LE I N FO A B S T R A C T

Keywords: Many pharmaceuticals such as pills, capsules, or tablets are prepared in a dried and powdered form. In this field,
Nano spray drying spray drying plays a critical role to convert liquid pharmaceutical formulations into powders. In addition, in
Pharmaceuticals many cases it is necessary to encapsulate bioactive drugs into wall materials to protect them against harsh
Encapsulation process and environmental conditions, as well as to deliver the drug to the right place and at the correct time
Powder properties
within the body. Thus, spray drying is a common process used for encapsulation of pharmaceuticals. In view of
Nanoparticles
the rapid progress of nanoencapsulation techniques in pharmaceutics, nano spray drying is used to improve drug
formulation and delivery. The nano spray dryer developed in the recent years provides ultrafine powders at
nanoscale and high product yields. In this paper, after explaining the concept of nano spray drying and un-
derstanding the key elements of the equipment, the influence of the process parameters on the final powders
properties, like particle size, morphology, encapsulation efficiency, drug loading and release, will be discussed.
Then, numerous application examples are reviewed for nano spray drying and encapsulation of various drugs in
the early stages of product development along with a brief overview of the obtained results and characterization
techniques.

1. Introduction dryer. First, the liquid feed is atomized in a nozzle. The reduction in
droplet size leads to a large increase in the surface area. In the drying
Spray drying is a simple, fast, reproducible, and scalable drying chamber, the solvent in the sprayed droplets is quickly removed by the
technology (Arpagaus and Schwartzbach, 2008), allowing mild tem- continuous flow of a hot drying gas. Dry particles are formed and se-
perature conditions suited to heat-sensitive biopharmaceutical com- parated from the gas stream and are collected in a collection vessel.
pounds. It is well established in the food (Anandharamakrishnan, 2014; In the context of pharmaceuticals encapsulation applications, the
Celli et al., 2015; Esfanjani et al., 2015; Gharsallaoui et al., 2007; Jafari main benefits of spray drying are (Arpagaus et al., 2017, 2013; Wong
et al., 2008b, 2007; Mahdavi et al., 2014; Murugesan and Orsat, 2012; and John, 2015):
Rajabi et al., 2015), chemical (Masters, 1991; Okuyama and Lenggoro,
2003), and pharmaceutical industries (Bürki et al., 2011; Cal and • Control of particle size, shape, and morphology (amorphous/crys-
Sollohub, 2009; Vehring, 2008; Wang et al., 2005; Wendel and Celik, talline form, porosity)
2005). Compared to other drying technologies used in drug delivery • One-step process to directly convert various liquid feeds into dry
applications, spray drying is a continuous process to directly transform powders
various liquids (e.g. solutions, emulsions, dispersions, slurries, pastes or • Process simplicity and ease of operation
even melts) into solid particles with adjustable size, distribution, shape, • Low operating costs, energy efficient technology, and fast process
porosity, density, and chemical composition. Spray drying equipment is • Scale-up capability
commercially available and the production cost is typically lower • Open and closed cycle design for aqueous and organic solvents spray
compared to other drying technologies like for example freeze drying drying
(Arpagaus et al., 2017; Celli et al., 2015; Gharsallaoui et al., 2007). • Processing of heat-sensitive substances with low risk of degradation
The process steps of spray drying are basically (1) heating of the • Design of particles with controlled drug release properties
drying gas, (2) droplet generation, (3) drying of the droplets, and (4) • High encapsulation efficiency and extended shelf life
particle collection. Fig. 1 illustrates a principle flow diagram of a spray • Versatile technique for the formulation of nanocapsules with various

Corresponding author.
E-mail addresses: cordin.arpagaus@ntb.ch (C. Arpagaus), smjafari@gau.ac.ir (S.M. Jafari).

https://doi.org/10.1016/j.ijpharm.2018.05.037
Received 2 March 2018; Received in revised form 14 May 2018; Accepted 15 May 2018
Available online 17 May 2018
0378-5173/ © 2018 Elsevier B.V. All rights reserved.
C. Arpagaus et al. International Journal of Pharmaceutics 546 (2018) 194–214

Nomenclature SEM scanning electron microscopy


SLS static light scattering
AFM atomic force microscopy TEM transmission electron microscopy
ANOVA analysis of variance Tg glass transition temperature (°C)
C relative atomic concentration of carbon Tm melting point (°C)
CLSM confocal laser scanning microscopy Tm inlet drying temperature (°C]
Cp heat capacity (kJ/kg K) WPC whey protein concentrate
DLS dynamic light scattering XPS X-ray photoelectron spectroscopy
DOE design of experiments
DSC differential scanning calorimetry Subscripts
FTIR Fourier transform infrared spectroscopy
FESEM Field emission SEM E encapsulated in powder
LIBD Laser-induced breakdown detection S on the powder surface
NMR nuclear magnetic resonance O fish oil
N relative atomic concentration of nitrogen P protein
O relative atomic concentration of oxygen α, β, γ percentage (%)
PDI polydispersity index

encapsulating excipients various advantages, such as a higher surface-volume ratio, a higher


penetration rate into the cells, higher stability, higher possibility of
A spray dried powder form offers higher stability, better protection targeted release through surface decoration, etc.
from the environment (e.g. oxidation, light, and temperature), easier The definition of a nanoparticle in the pharmaceutical industry can
handling and storage, and redispersibility in aqueous solutions be described as solid colloidal particles with sizes below 1 µm (Kaialy
(Murugesan and Orsat, 2012; Okuyama et al., 2006). The produced and Al Shafiee, 2016; Lee et al., 2011; Li et al., 2010; Wong, 2015;
powders are high in quality and have typically low moisture contents Wong and John, 2015). Fig. 2 illustrates distinct application areas
(Schmid, 2011), resulting in high shelf life stability where nano spray drying plays a crucial role in the formulation of
(Anandharamakrishnan and Ishwarya, 2015). The cooling effect of the pharmaceuticals in powder form. The particle formation process during
evaporating solvent conserves the droplet temperature relatively low. nano spray drying can be described by solvent evaporation and diffu-
Therefore, heat-sensitive products can be dried with negligible de- sion of solutes in the droplet because of simultaneous heat and mass
gradation (Masters, 1991). In addition, spray drying offers high flex- transfer (Vehring, 2008). A solution of the product (A) is dispersed into
ibility to control particle size and morphology by optimizing the process fine droplets (B). The solvent evaporates immediately, the droplets
parameters and feed formulation (Nandiyanto and Okuyama, 2011). shrink, and a solid powder is formed. The final product (C) is a fine
Smooth and spherical, collapsed, dimpled, wrinkled, raisin-like, and powder that can be amorphous or crystalline. Feed dilution results in a
highly crumpled or folded particles can be obtained (Arpagaus et al., final product with smaller particle sizes. Practical applications include
2017; Nandiyanto and Okuyama, 2011). Various particle designs can be for example polymeric wall materials, like Arabic gum, whey protein,
prepared by spray drying encapsulation, such as single core, irregular, polyvinyl alcohol, modified starch, or maltodextrin (Li et al., 2015,
multi-wall, multi-cores, or composites. 2010), as will be discussed in more detail in Section 4. Nanonization or
The main advantage of nano spray drying is the production of drug- structural change is used to change the particle morphology from a
loaded nanoscale particles that can be used for drug delivery purposes. coarse grain to a very fine powder. This improves the solubility of the
Nano spray drying enables the generation of smaller particle sizes than final drug product due to the higher surface-to-volume ratio of nano
conventional spray dryers, which improves bioavailability and release particles and due to more amorphous structures, which the solvent (e.g.
of bioactive components and drugs. Drug-loaded nanoparticles offer water) can penetrate more efficiently. Typical examples are drugs with
poor solubility in water (Li et al., 2010; Martena et al., 2012a; Schmid
et al., 2011) and salts (Moncada et al., 2015). A main advantage of
nanonization is that particles of very uniform size are achieved. A
crystalline product (A) is dissolved in a solvent (B) and this solution (C)
is dispersed into tiny droplets (D). The result is a final product (E) si-
milar to the final product described in nano spray drying. A change of
crystalline drugs into more amorphous structures provides faster drug
release kinetics.
In nanoencapsulation, typically a liquid product is embedded inside
a solid matrix. Encapsulated oil-in-water nanoemulsion are common
examples, where the oil droplets serve as the reservoir for a lipophilic
drug product (A). Together with a carrier substance (B), and a filmogen
solution (C), an emulsion (D) is created. Organic binders like polymeric
wall materials (Ezhilarasi et al., 2013; Fathi et al., 2014; Gharsallaoui
et al., 2007; Mahdavi et al., 2014; Ngan et al., 2014; Rajabi et al., 2015;
Sabliov and Astete, 2015) are frequently used to reach high en-
capsulation efficiency and capsule stability (see more details on dif-
ferent wall materials in Table 2). The emulsion is then sprayed into
small droplets (E) and the solvent evaporates leaving a solid matrix
around the dispersed second phase (F). This results small droplets of the
Fig. 1. Principle flow diagram of a traditional spray dryer (Arpagaus et al.,
product (A) stored in the carrier substance (B) and embedded in the
2017). filmogen (C). Capsules smaller than 1 µm in diameter are known as

195
C. Arpagaus et al. International Journal of Pharmaceutics 546 (2018) 194–214

Fig. 2. Application areas for a nano spray dryer (Arpagaus et al., 2017).

nanocapsules (Arpagaus et al., 2017; Couvreur et al., 1995; Jafari et al., subject of nano spray drying technology have already been published
2008b). Jafari et al. (2007, 2008a,b) proposed the term “nanoparticle (Arpagaus, 2012, 2011; Arpagaus et al., 2017; Heng et al., 2011; Lee
encapsulation” for cases where the core material is within the nano size et al., 2011; Nandiyanto and Okuyama, 2011; Okuyama et al., 2006;
range and is encapsulated into a matrix of micron sized powder parti- Sosnik and Seremeta, 2015; Wang et al., 2005). The actual experiences
cles. In drug delivery, due to their small sizes and high specific surface gained from published works on nano spray drying for drug delivery
areas, nanocapsules offer improved dissolution rates and bioavailability applications are reviewed in this paper. After explaining the concept of
of the bioactive compounds compared to microcapsules (Lee et al., nano spray drying and understanding the key elements of the equip-
2011). Nanoenglobing is similar to nanoencapsulation, but a solid ment, the influence of the process parameters on the final powder
material is embedded in another solid or liquid product. Examples are properties, such as particle size, morphology, encapsulation efficiency,
carotenoids in gelatine or magnetic iron oxide nanoparticles in Eudragit drug loading and release, will be discussed. Then, numerous applica-
or maltodextrin (Perecin et al., 2014). A nanodispersion (D) is created tions of nano spray drying for the formulation and encapsulation of
from a solid product (A), a matrix material (B), water, and a filmogen different pharmaceuticals will be presented along with a brief overview
solution (C). This suspension is then sprayed into small droplets (E). of the obtained results and characterization techniques.
The matrix and the filmogen lead to an agglomeration or coating of the
suspended particles (A), resulting in nanocomposite particles (F).
2. Elements of a nano spray dryer
The recent advances in nanomedicine and nanotherapeutics have
increased the need for spray dryers that allow high yields of nanoscale
In order to produce nanoscale particles using spray drying tech-
particles with narrow size distribution. Several extensive reviews on the
nology, some modifications to the experimental setup of conventional

Fig. 3. Schematic of a nano spray dryer and its functional principles: Tin, Tout: inlet and outlet drying gas temperature (Arpagaus, 2012, 2011, Arpagaus et al., 2017,
2010a, 2010b, Büchi Labortechnik, 2013, 2010).

196
C. Arpagaus et al. International Journal of Pharmaceutics 546 (2018) 194–214

spray dryers are necessary. A significant constraint is the limited effi- (Arpagaus et al., 2017; Schmid, 2011; Schmid et al., 2011). The co-
ciency of the separation and collection of submicron particles by cy- current drying gas flow is heated up to the set inlet temperature (Tin)
clone separators. Typical cyclones are unable to collect particles below and directs the submicron particles to the electrostatic particle col-
2 µm (Bowen et al., 2013; Chan and Kwok, 2011; Heng et al., 2011; Maa lector. The dried particles are electrostatically charged and collected at
et al., 1998; Maltesen et al., 2008; Schmid et al., 2009). Even with high- the collecting electrode. At the same time, particle adhesion to the side
performance glass cyclones (Brandenberger, 2003), the median particle walls of the drying chamber is minimized, thus resulting in high particle
size cannot be reduced below 1.4 µm (Maltesen et al., 2008). In other collection efficiency (Arpagaus, 2012; Lee et al., 2011). The flow of the
words, particles of submicron size can only be collected to a very lim- drying gas is laminar, which makes the system suitable for gentle drying
ited extent with conventional spray dryers. A feasible way to collect heat-sensitive products with a low risk of degradation or loss of activity.
nanoscale particles is to use electrostatic particle collectors (Arpagaus, The drying gas exits the spray dryer in a purified form and the outlet
2011, 2010; Bürki et al., 2011; Heng et al., 2011; Li et al., 2010; Schmid temperature is measured.
et al., 2011, 2009; Schön and Baumgartner, 2009). In addition, tradi- A high voltage of 17 kV at the collecting electrode (electrical current
tional atomizers do not allow the generation of very fine droplets in of a few 10 mA) enables the efficient recovery of submicron particles at
order to achieve dried solid particles in the submicron range. Further- a separation efficiency greater than 99% for small solid batches of
more, a turbulent gas flow in the drying chamber may lead to increased 10 mg–2.7 g (Arpagaus et al., 2010b; Arpagaus and Meuri, 2010; Bürki
particle deposits on the chamber wall. Recent advances in nano spray et al., 2011; Lee et al., 2011; Li et al., 2010; Schmid, 2011; Schmid
dryer technology and the introduction of the Nano Spray Dryer B-90 by et al., 2011, 2009). The electrostatic particle collector can even collect
Büchi Labortechnik AG (Switzerland) in 2009 have made it possible to thin-walled particles without breaking (Feng et al., 2011; Sun et al.,
produce submicron particles (Arpagaus, 2011; Arpagaus et al., 2017, 2011). Finally, the particles are gently removed from the surface of the
2010a, 2010b, Büchi Labortechnik, 2013, 2010). Fig. 3 shows a sche- collecting electrode by using a scraper. Variations in the yield may
matic representation of the nano spray dryer and its functional princi- occur due to the manual collection of the powder with a scraper. It is
ples. recommended to recover the powder from the collecting electrode cy-
The droplet generation is based on vibrating mesh technology, linder in a controlled and dry atmosphere, and to store the samples
which has been adapted from nebulizers used in aerosol drug delivery afterward in glass vials inside a desiccator at room temperature for
(Dhand, 2002; Knoch and Keller, 2005; Lass et al., 2006; Smart et al., further characterization to prevent crystallization and moisture uptake
2002; Vecellio, 2006). A piezoelectric actuator vibrates a spray nozzle (Schmid, 2011).
at ultrasonic frequency (80–140 kHz). Attached to the nozzle is a small For aqueous feeds, the typical system configuration is an open mode
replaceable spray cap, which comprises a thin perforated metal plate, with air as drying gas. When using flammable organic solvents, a closed
containing a series of tiny laser drilled holes. The piezoelectric vibration mode operation with an Inert Loop B-295 (Büchi Labortechnik, 2010)
leads to a fast upward and downward movement of the spray mesh, and solvent resistant tubing is applied. The Inert Loop B-295 separates
thus ejecting millions of precisely sized droplets through the holes into the organic solvents from the drying gas and provides safe and clean
the drying chamber (glass cylinder, 80 cm height, 20 cm diameter). The handling of organic solvents during nano spray drying. Inert gases like
droplet size depends on the mesh size and the physicochemical prop- nitrogen and carbon dioxide are typically used as drying gases to pre-
erties of the fluid, such as viscosity and surface tension. Spray meshes vent the formation of an explosive gas mixture. The oxygen con-
are available with 4.0, 5.5, and 7.0 µm hole diameters. With a 4.0 µm centration is controlled below 4% and the inert gases are recirculated
spray mesh, water droplets of approximately 3–8 µm are produced throughout the entire circuit.

Fig. 4. Process parameters and formulation variables for a nano spray dryer (Arpagaus et al., 2017).

197
C. Arpagaus et al. International Journal of Pharmaceutics 546 (2018) 194–214

3. Formulation of drugs for nano spray drying and process morphology of nano spray dried bovine serum albumin particles.
efficiency As shown in Table 1, the key parameters controlling the final par-
ticle size are the spray mesh size (Baba and Nishida, 2013, 2012; Lee
Fig. 4 illustrates the main adjustable process parameters and for- et al., 2011) and the solid concentration (Lee et al., 2011; Li et al.,
mulation variables for a nano spray dryer (marked black), as well as the 2010; Schmid, 2011; Schmid et al., 2011, 2009). Smaller droplets are
output parameters (marked grey). The most important input parameters favored by a higher viscosity, a lower surface tension, and a smaller
identified are the spray mesh size, the spray rate intensity, the solid spray mesh (Beck-Broichsitter et al., 2012; Bürki et al., 2011; De Cicco
concentration, the polymer and surfactant concentrations, the drying et al., 2014; Lee et al., 2011; Littringer et al., 2013). The submicron
gas inlet temperature, the solvent type, and the drying gas flow rate. particle size region is typically reached when using a 4.0 µm spray mesh
Table 1 gives an overview of the main process parameters and their and diluted solutions of 0.1–1% (w/v). Further reduction of particle
influence on the output parameters (i.e. outlet temperature, droplet size is possible by further dilution, as demonstrated in several studies
size, feed rate) and final product properties (i.e. particle size, moisture (Baba and Nishida, 2013; Beck-Broichsitter et al., 2015a, 2012; Lee
content, yield, stability). The thickness of each arrow illustrates the et al., 2011; Li et al., 2010; Nandiyanto and Okuyama, 2011; Ngan
strength of the related influence. et al., 2014; Pérez-Masiá et al., 2015). The typical organic solvents used
Of primary interest for pharmaceuticals are the effects on the par- in nano spray drying of pharmaceuticals are:
ticle size, morphology, product yield, productivity, encapsulation effi-
ciency, drug loading, drug release profile, and stability. Depending on • dichloromethane (DCM) (Beck-Broichsitter et al., 2012; Bege et al.,
the application, an optimized set of process parameters is found. The 2013; Dahili et al., 2015; Dahili and Feczkó, 2015; Draheim et al.,
optimization of the process parameters is usually done by trial and 2015; Panda et al., 2014; Schafroth et al., 2012),
error. However, design of experiment (DOE) studies help to optimize • acetone (Beber et al., 2014; Beck-Broichsitter et al., 2015b; Draheim
the nano spray drying process, as shown by several authors (Arpagaus et al., 2015; Durli et al., 2014; Fontana et al., 2014; Gu et al., 2015;
et al., 2017; Bürki et al., 2011; Draheim et al., 2015; Durli et al., 2014; Li et al., 2010; Perecin et al., 2015),
Gu et al., 2015; Harsha et al., 2017, 2013b; Lee et al., 2011; Li et al., • ethanol (Baba and Nishida, 2013, 2012; Bürki et al., 2011; Maged
2010; Littringer et al., 2013; Schafroth et al., 2012; Schoubben et al., et al., 2017; Zellnitz et al., 2015a),
2014). • methanol (Basran, 2017; Öztürk et al., 2017, 2015; Schmid, 2011;
DOE studies enable to determine the optimal process conditions Suryaprakash et al., 2014),
with fewer experiments, thereby reducing the cost of experiments and • acetonitrile (Amsalem et al., 2017; Denora et al., 2016),
material usage. Factorial experimental designs and the Taguchi method • ethylacetate (Draheim et al., 2015), and
are the main used statistical approaches. Bürki et al. (2011) in- • mixtures thereof with water (Aquino et al., 2014; Dimer et al.,
vestigated the effects of the inlet temperature of the drying air, the 2015a; Kaewjan and Srichana, 2016; Martena et al., 2012a;
ethanol content of the spray solution and the spray mesh on particle Schoubben et al., 2015, 2013a; Son et al., 2013b).
size by a 33 full factorial design, focusing on nano spray drying a pro-
tein. For salbutamol, Littringer et al. (2013) studied the influence of The selection of the organic solvent is based on the solubilization of
mesh size, feed concentration and drying air temperature on particle the drug and the encapsulating wall materials. The mixing ratio is ad-
size, product quantity per time and product yield with a 33 full factorial justed to allow the complete dissolution of the compounds (Schoubben
design. Schoubben et al. (2014) developed inhalable capreomycin et al., 2013a). For example, acetone–water mixtures dissolve steroidal
powders with a desired particle diameter and highest yield using a 23 dexamethasone well, and the low viscosity of the acetone allows higher
factorial design. Durli et al. (2014) evaluated the influence of the type flow rates through the vibrating spray mesh, which shortens the pro-
of organic solvent and surfactant on powder properties of anti-in- cessing time (Fontana et al., 2014). Compared to water, organic sol-
flammatory dexamethasone using a 32 full factorial analysis design vents generate slightly smaller droplets due to their lower surface
composed of two variables at three levels. Gu et al. (2015) applied a tension, viscosity, and density (Suryaprakash et al., 2014). In addition,
central 3-factor 5-level composite design to study the effects of polymer organic solvents enable lower drying temperatures because of the lower
concentration, inlet temperature and air flow on the yield, spray rate boiling points. Dichloromethane (40 °C) or acetone (56 °C) lead to fast
and drying efficiency for the preparation of solid dispersions. More drying and prevent particles from sticking to the walls or agglomer-
recently, Harsha et al. (2017) optimized the nano spray drying process ating. The evaporation temperature is lower than the melting tem-
for albumin particles loaded with salbutamol using a central composite perature or the glass transition temperature of certain polymers. A
design. Analysis of variance (ANOVA) is typically used to determine the drawback of toxic solvents (such as methanol or chloroform) is the
statistical significance of each factor on individual responses. Lee et al. potential residual content in the final product as an impurity. Poly
(2011) applied the Taguchi design method (based on five variables and (lactic-co-glycolic acid) (PLGA) is one of the most commonly used
three levels) to identify the dominant factors affecting the size and materials for spray dried biomedical applications due to its well

Table 1
Influence of the main process parameters in nano spray drying ( / strong increasing/decreasing influence, ↑/↓ weak increasing/decreasing influence, – minimal or
no influence) ().
adapted from Arpagaus et al. (2017)
Process parameter Outlet temperature Droplet size Particle size Feed rate Moisture content Yield Stability

Drying gas flow rate↑ – – – – –


Drying gas humidity↑ ↑ – – – ↓ –
Inlet temperature↑ – ↑ – ↓ ↑
Spray mesh size↑ ↓ – – ↑
Spray rate intensity↑ ↑ ↑ ↑ –
Circulation pump rate↑ – ↑ ↑ ↑ – – ↑
Solid concentration (viscosity)↑ ↑ – ↓ ↑ –
Surfactant/stabilizer in feed↑ – ↓ ↓ ↑ – ↑
Solvent instead of water ↓ ↓ ↑ ↑ –

198
C. Arpagaus et al. International Journal of Pharmaceutics 546 (2018) 194–214

established biocompatibility and biodegradability (Arpagaus and particles have been optimized by several researchers (Bürki et al., 2011;
Schafroth, 2009, 2007). As an example, the optimum inlet drying Li et al., 2010; Schmid, 2011; Schmid et al., 2011, 2009) through
temperature of PLGA dissolved in dichloromethane is in the range of adding surfactants, which balance the surface-to-viscous forces inside
29–32 °C (Schafroth et al., 2012). For water applications, the outlet the drying droplet and enable the formation of a smooth spherical
temperatures range between 28 and 59 °C (Arpagaus et al., 2017), surface on the dry particle. More corrugated surfaces are observed for
which makes nano spray drying a very suitable process for heat-sensi- example for pure dexamethasone without an encapsulating polymer
tive drugs (Amsalem et al., 2017; Aquino et al., 2014; Heng et al., (Fontana et al., 2014), or levofloxacin encapsulated in chitosan
2011). The outlet gas temperature is mainly controlled by the inlet (Merchant et al., 2014). Shriveled (Harsha, 2015) and wrinkled (Torge
drying gas temperature, the drying gas flow rate, and the fluid feed rate et al., 2017) particles are obtained from surfactant-free formulations
(Arpagaus et al., 2017). In a spray dryer, the droplet temperature rises (Lee et al., 2011) and are more likely for larger particles (Bürki et al.,
initially to the saturated wet-bulb surface temperature, remains con- 2011). Doughnut-like particles are the result of hydrodynamic effects
stant during evaporation, and approaches the temperature of the sur- and the loss of structural stability of the sprayed droplets (Dahili et al.,
rounding gas at the dryer outlet. A lower drying gas flow rate leads to a 2015; Lee et al., 2011). Composite particles, formed from suspensions
lower outlet temperature. A smaller feed rate increases the outlet prior to the nano spray drying, consist of agglomerated nanoparticles,
temperature because less fluid is evaporated over the same period of which give rise to a high specific surface area (Beck-Broichsitter et al.,
time (Beck-Broichsitter et al., 2015b; Littringer et al., 2013). A higher 2012). Torge et al. (2017) embedded PLGA nanoparticles in a mannitol
inlet temperature reduces the relative humidity of the drying gas and matrix. The encapsulated nanoparticles may preserve their integrity,
forms particles with less moisture content leading to a drier powder specific properties, redispersibility, enhanced solubility, and targeting
with less stickiness onto the drying chamber. Considering electrostatic capacity (Sarma et al., 2014).
interactions, at higher zeta potentials, more repulsion of particles will As shown in several studies, the nano spray drying process itself has
occur resulting in less stickiness. Residual moisture contents of lower a negligible or only a marginal impact on the product degradation or
than 0.5% in mannitol and 2–5% in trehalose powders have been ob- aggregation. Schultz et al. (2015) showed that the activity of insulin-
served (Schmid, 2011). To achieve a final product with low residual like growth factor encapsulated in trehalose remained unaffected.
moisture contents, the inlet temperature needs to be set as high as Schmid (2011) detected a slight reduction in L-lactic dehydrogenase
possible and the difference in temperatures of the inlet and the outlet enzyme activity during pump circulation. An option to minimize the
has to be as small as possible (Arpagaus et al., 2013). The drying times risk of activity loss during the nano spray drying process is to cool the
for water droplets in a nano spray dryer are in the order of 10 ms, as- feed vessel in ice (Amsalem et al., 2017; Schoubben et al., 2013a; Torge
suming 7 µm droplets, 75 °C drying temperature and 100 L/min drying et al., 2017) or to use a larger spray mesh to reduce the mechanical
air flow rate (Feng et al., 2011). shear of atomization, as shown by Bürki et al. (2011) with β-galacto-
The feed rate increases primarily with the spray mesh size, the sidase enzyme. After nano spray drying, the activity of the encapsulated
setting of the relative spray rate intensity, the recirculation pump rate, product is preserved if the powder is stored under controlled conditions
and it depends on the feed formulation (i.e. the substance, the solid and if a stabilizer is added into the feed formulation. Most nano spray
concentration, the solvent type, and the addition of surfactant). For dried powders tend to be amorphous due to the too short drying time to
pure water, the specified feed rates are in the ranges of 10–20, 25–50, form crystalline structures. Examples include salbutamol (Chan and
and 80–150 mL/h for a 4.0, 5.5, and 7.0 µm spray mesh, respectively Kwok, 2011; Littringer et al., 2013), albuterol in mannitol (Son et al.,
(Büchi Labortechnik, 2010). When using an aqueous substance and a 2013a) and trehalose (Schmid, 2011; Schmid et al., 2011), levofloxacin
4.0 µm spray mesh, the feed rates may vary in a range of about in chitosan (Merchant et al., 2014), or clozapine-risperidone in PLGA
3–25 mL/h (Arpagaus et al., 2010b; Li et al., 2010; Schmid, 2011; (Panda et al., 2014). To prevent recrystallization of amorphous drugs,
Schmid et al., 2011; Sun et al., 2011). With a 5.5 µm spray mesh and the powders are stored under dry conditions (Littringer et al., 2013;
0.1% (w/v) chitosan in 1% (v/v) acetic acid, feed rates up to about Pinto et al., 2017; Zellnitz et al., 2015a). Bürki et al. (2011) showed that
50 mL/h were measured by Gautier et al. (2010). Generally, a higher nano spray dried β-galactosidase enzyme in trehalose lost only 3% of its
solid concentration results in a lower feed rate (Beck-Broichsitter et al., activity during three weeks of storage at 70 °C. Dahili and Feczkó
2015b; Gu et al., 2015; Schmid, 2011; Schoubben et al., 2013a). The (2015) enhanced the activity of peroxidase enzyme by immobilization
addition of organic solvents to the water tends to increase the feed in ethylcellulose and PLGA. Pérez-Masiá et al. (2015) observed bioac-
rates, which can be attributed to the lower surface tensions. The further tive stability of folic acid (vitamin B9) in whey protein after 60 days
admixture of a small amount of surfactant (e.g. Tween) into the feed under dry storage conditions and in darkness. Merchant et al. (2014)
may also improve the throughput, as reported for example by Schmid found no evidence of changes in the crystallinity of chitosan powder
et al. (2011) for trehalose solutions. The added surfactant leads to a after storage for 60 days at room temperature and ambient humidity.
reduction of the surface tension and the formation of a more uniform Martena et al. (2012a) did not observe any changes in the physical
spray. This is also a strategy to reduce the stickiness of atomized dro- properties of amorphous nano spray dried blood pressure regulation
plets and to produce of a spray with smaller droplets. A lower relative nicergoline at accelerated storage conditions (60% RH, 25 °C) during
spray rate intensity reduces the feed rate almost linearly (Beck- the testing period of 6 months. The amorphous form was stable for at
Broichsitter et al., 2015b). Moreover, a faster circulation rate of the least a period of 1 year when stored at 3 °C. Also, De Cicco et al. (2014)
peristaltic pump increases the fluid pressure on the spray mesh, thus conducted accelerated storage stability tests of antibacterial gentamicin
increasing the initial droplet size and consequently the feed rate in alginate-pectin powders at 40 °C and 75% relative humidity. The
(Suryaprakash et al., 2014). drug was preserved for 6 months. Finally, Harsha (2013) kept nano-
The morphology of particles prepared by nano spray drying depends particle formulations of antibiotic amoxicillin stable for 24 months at
on the drying conditions and the feed properties. Dense, hollow, 25 °C and 60% RH.
porous, and encapsulated structures with spherical, wrinkled, shriveled,
or even doughnut-like shapes are possible (Arpagaus et al., 2017). In 4. Nanoencapsulation of various drugs by nano spray drying
general, slow drying yields more compact particles, while fast and high
temperature drying leads to the formation of more hollow particles with Nano spray drying has received a widespread application in the
a thin shell (Feng et al., 2011; Kaewjan and Srichana, 2016; Nandiyanto formulation of pharmaceutical dosage forms to transform solutions,
and Okuyama, 2011; Sun et al., 2011). Surface-active compounds in the suspensions, emulsions, liposomes, and others liquid feeds into a dry
formulations, such as leucine also promote the formation of hollow solid submicron powder. Since the market introduction of the Nano
particles (Feng et al., 2011). The smoothness and sphericity of the Spray Dryer B-90 by Büchi Labortechnik AG (Switzerland) in 2009,

199
C. Arpagaus et al. International Journal of Pharmaceutics 546 (2018) 194–214

there have been a great number of research papers published (at least
over 160) in the field of nano spray drying of pharmaceuticals. This

Beck-Broichsitter et al. (2012)


Beck-Broichsitter et al. (2012)
Schmid (2011), Schmid et al.
section reviews the accumulated application experience. Different wall
materials (excipients) applied in nano spray drying of pharmaceuticals

Draheim et al. (2015)


has been listed in Table 2. Table 2 also summarizes the process para-

Gautier et al. (2010)

Dahili et al. (2017)


Torge et al. (2017)
Feng et al. (2011)
Blasi et al. (2010)
meters for pure excipients used as dispersing agents, binders and sta-

Gu et al. (2015)
Li et al. (2010)
bilizers (Beck-Broichsitter et al., 2012; Blasi et al., 2010; Dahili et al.,

(2011, 2009)
References
2017; Draheim et al., 2015; Feng et al., 2011; Gautier et al., 2010; Gu
et al., 2015; Li et al., 2010; Schmid et al., 2009; Schmid, 2011; Schmid
et al., 2011; Torge et al., 2017). This includes numerous compounds,
such as:

Product yield
• water-soluble carbohydrates (i.e. Arabic gum, alginate, chitosan,
cyclodextrin, cellulose derivatives, modified starch, maltodextrin,

43–95

50–78

60–85

75–91
> 90

n.a.

n.a.

n.a.
n.a.
n.a.
n.a.
(%)
pectin, mannitol, lactose trehalose),
• proteins (i.e. gelatin, serum albumin, whey protein, sodium case-
inate, silk fibroin, leucine),

Solid sample amount


water-soluble synthetic polymers (i.e. poly(vinyl alcohol), poly
(ethylene glycol) or poly(acrylic acid) (Carbopol)),
• hydrophobic synthetic polymers (i.e. poly(lactic-co-glycolic acid),

1’080–1’350
poly(ε-caprolactone), poly(vinyl pyrrolidone), Kollidon, Eudragit),

30–300

10–50
and

(mg)

800
n.a.

n.a.

n.a.

n.a.
n.a.
n.a.
n.a.

Different wall materials (excipients) which have been applied in encapsulation of pharmaceuticals by nano spray drying (n.a. = not available).
fats (i.e. stearic acid and glyceryl behenate (Compritol)).

In this paper, published studies on nano spray dried pharmaceu-

Particle size
ticals have been structured according to the following routes of ad-

1.0–16.0

2.0–10.0
0.2–1.1

0.4–1.2
0.3–3.0

2.1–5.4

1.1–7.2

0.6–1.6
2.8–4.4
0.3–1.7

2.4–8.0
ministration:
(µm)
• Pulmonary
• Oral
Drying gas (L/

100 (N2/CO2)
115 (N2/CO2)
• Intravenous
• Topical

90–120
• Ophthalmic
min)

100

100
115

100

140

130
100
100
• Intraperitoneal
• Intravesical
T out (°C)

• Cerebral
38–60

30–45

32–39

20–30
20–30

29–36
n.a.

n.a.

n.a.
45

55
The experimental process parameters for nano spray drying (i.e.
T in (°C)

60–100

30–50
30–50

50–90

36–68
inlet and outlet drying temperatures, drying gas flow) have been
100

110

120

n.a.
75

80

summarize in the relevant Tables for the following sections which can
be used as first guess values for applying the nano spray dryer to
Dichloro-methane (DCM)

identical or similar substances. In addition, there are many applications


Acetone, Ethylacetate,

of nano spray dried particles containing drugs and excipients for the
Water, Acetic acid

treatment of various diseases, including:

• tuberculosis,
Acetone

• asthma,
Solvent

Water

Water
Water

Water

Water

DCM,
DCM
DCM

• inflammation,
• cystic fibrosis,
Nanosuspension of mannitol and poly(lactic-co-glycolic acid)
Leucine (Aminoacid, dispersing agent), Trehalose (Stabilizer,

• diabetes,
Arabic gum, whey protein, maltodextrin, polyvinyl alcohol

• Alzheimer’s and Parkinson’s diseases,


Hypromellose (Hydroxypropyl-methylcellulose, for oral
Trehalose, mannitol or disodium phosphate surfactant,

• fungal infection,
Encapsulation wall materials (excipient), application

• ophthalmic disorders,
• cancer,
PLGA and stabilizers (Poloxamer, surfactant)

• high blood pressure,


• congestive heart failure,
• edema
Chitosan (Biopolymer, fat blocker)
Sodium alginate (Polysaccharide)

PLGA (Biodegradable polymer)

PLGA (Fine carrier supports)

and others, thus underlining the versatility of the nano spray drying
and modified starch

increases shelf life)

technology to develop nanomedicines.


As can be seen in the following sections, the organic solvents used to
medicaments)
PLGA suspensions
polysorbate

dissolve poorly water soluble drugs are mainly ethanol, methanol,


(PLGA)

acetone, ethylacetate, and dichloromethane. With highly diluted solu-


tions containing 0.1–1% (w/v) solids concentrations, finest solid par-
Table 2

ticles down to 100 nm can be obtained by nano spray drying. Pure drug
particles in the nanosize dimensions and the amorphous state offer

200
Table 3
Published studies on nano spray dried drug delivery applications by pulmonary (lung) route (Drug loading (%) = Amount of drug in particles/Mass of particles; Encapsulation efficiency (%) = Amount of drug in
particles/Initial drug amount; n.a. = not available; – = not relevant).
Type of drug, application, wall material (excipient) Solvent T in (°C) T out (°C) Drying gas Particle size Solid sample Product Drug Encap-sulation References
C. Arpagaus et al.

(L/min) (µm) amount (mg) yield (%) loadi- efficiency (%)


ng
(%)

Bacitracin (Polypeptide antibiotic) Water 100–120 n.a. n.a. 3.1–6.6 20–200 18–82 – Schoubben et al. (2014)
Ciprofloxacin and gatifloxacin (Antiinflammatory, antibacterial) Water 120 40–45 100 (N2) 0.8–3.6 n.a. 70–80 Lee et al. (2013)
Ethambutol dihydrochloride (Antituberculosis drug for inhalation) Water 110 n.a. n.a. 0.2 n.a. n.a. Ahmad et al. (2014)
mixed with chitosan carrier particles
Salbutamol sulfate (Asthma) Water, 80–120 40–50 110 1.0–6.4 25–2’500 41–90 Faulhammer et al. (2018,
Ethanol 2015), Littringer et al. (2013),
Zellnitz et al. (2015a,b, 2014)
Salbutamol sulphate (Asthma) Water 120 49 110 0.6–7.6 n.a. n.a. Pinto et al. (2017)
Alkaline phosphatase (Model protein for β-cyclodextrin, L-leucine Water n.a. n.a. n.a. ∼1 n.a. n.a. n.a. n.a. Guo and Li (2013)
pulmonary delivery)
Amoxicillin (Antibiotic to treat Chitosan nanosuspension Water, Acetic 120 80 120 0.2–1.2 n.a. n.a. 90 n.a. Nguyen et al. (2017)
pneumococcal disease) by ionic gelation acid
Azithromycin (Antibiotic to treat cystic Mannitol, Leucine, Sodium Water 80 45 120 1.0–5.0 500 n.a. 30–40 n.a. Hindle et al. (2015)
fibrosis) chloride
Bovine serum albumin (Model protein Surfactant, Water 80–120 36–55 150 0.5–2.6 n.a. 68–76 n.a. n.a. Lee et al. (2011)
derived from cows) Polyoxyethylene, Sorbitan
monoleate
Capreomycin (Pulmonary tuberculosis) Leucine Ethanol, 80–120 n.a. 100 1.0–5.0 20–200 71–82 10–21 n.a. Schoubben et al. (2015, 2014,
Acetone, 2013a,b)
Water
Ciprofloxacin (Antibiotics) Mannitol, L-leucine, Water, 70 40 120 0.5–5.0 n.a. 50–60 n.a. n.a. Farkas et al. (2015), Longest

201
Poloxamer Ethanol et al. (2015)
Clarithromycin (Antibacterial to treat Leucine, Chitosan Water, 80 n.a. 120 0.3–2.5 150 58–63 35–37 n.a. Dimer et al. (2015a)
lung infections) Ethanol
Cyclosporin (Immune system PLGA DCM, Ethanol 29–32 28–32 102–132 0.9–2.2 600 20–56 13–14 38–41 Schafroth et al. (2012)
suppressant) (N2/CO2)
Dexamethasone (Steroid, anti- PLGA DCM, Ethanol 29–30 30–32 111–132 0.9–1.7 600 32–54 21–27 62–81 Schafroth et al. (2012)
inflammatory) (N2/CO2)
Dexamethasone (Steroid, anti- Poly(ε-caprolactone), Acetone 55 34 110 0.6–1.6 210 65–80 43 93–97 Fontana et al. (2014)
inflammatory) Deoxycholate Water
Fluticasone propionate (Asthma) Lactose Water 120 45–56 130 0.5–3.0 2’700 77 10 n.a. Arpagaus et al. (2010b)
β-galactosidase (Enzyme catalyzing Trehalose Water 80 36–53 100–110 1.0–5.0 500 60–94 n.a. n.a. Bürki et al. (2011)
hydrolysis of saccharides)
Insulin-like growth factor I (Anabolic Trehalose, Silk-fibroin, Water 70 n.a. 115–130 0.3–3.2 n.a. n.a. 74 93 Schultz et al. (2015)
peptide) Polysorbate
Ketoprofen lysinate (Anti-inflammatory Leucine Water, 60–110 n.a. 100 2.4–6.6 n.a. 21–90 n.a. n.a. Aquino et al. (2014)
to treat cystic fibrosis) Isopropyl-
alcohol
Levofloxacin (Antibiotic to treat lung Chitosan, L-leucine Water 120 n.a. 133 2.5–4.6 525 51–79 77–93 n.a. Merchant et al. (2014)
infections)
Methotrexate (Lung cancer) Carbopol (Poly(acrylic Water 85–115 30–50 n.a. 6.8 n.a. 77 89 n.a. Harsha et al. (2015a)
acid))
Phospholipid dispersions (Liposome) Trehalose, Egg, Lecitin Water 60–120 44–47 90–140 0.7–9.1 4’000–50’000 70 n.a. n.a. Brinkmann-Trettenes et al.
(2014)
Pyrazinamide (Anti-tuberculosis) L-leucine Water Ethanol 90–115 n.a. 100 2.8–3.8 n.a. n.a. n.a. n.a. Kaewjan and Srichana (2016)
Resveratrol (Pulmonary arterial Poly(ε-caprolactone), Acetone, 55 n.a. 110 (N2, 1.0–5.0 300 71–88 31 99 Dimer et al. (2015b)
hypertension) Sodium deoxycholate, Water CO2)
Trehalose
Sildenafil (Pulmonary arterial PLGA Acetone 45 25–30 100 (N2, 4.0–11.0 400–1’600 n.a. n.a. n.a. Beck-Broichsitter et al.
hypertension) CO2) (2015a)
(continued on next page)
International Journal of Pharmaceutics 546 (2018) 194–214
C. Arpagaus et al. International Journal of Pharmaceutics 546 (2018) 194–214

better absorption rates and higher bioavailability (Lee et al., 2011;

Behara et al. (2014a,b,c), Son


Martena et al., 2012a; Rascioni et al., 2016), and encourage future

Arpagaus et al. (2010b)


developments in this research field. Nanocapsules, with their reduced

Behara et al. (2014b)


size and large specific surface area provide pronounced improvement in

Yang et al. (2015)


et al. (2013a,b)
controlled drug release and bioavailability (Sithole et al., 2017). This
enables the generation of target drug delivery systems (Arpagaus, 2011;
References

Harsha et al., 2015a; Li et al., 2010). Under optimized conditions, un-


iquely high product yields of about 76–96% can be achieved to process
small sample amounts in the range of 10 mg–2.5 g. Variations in the
yield may occur due to particle depositions around the spray cap
Encap-sulation
efficiency (%)

(Amsalem et al., 2017; Brinkmann-Trettenes et al., 2014; Schmid et al.,


2011) and the chamber walls (Brinkmann-Trettenes et al., 2014; Bürki
et al., 2011; Lee et al., 2011), nozzle blockage (Schmid, 2011; Schmid
n.a.
n.a.

n.a.
n.a.

et al., 2011), or due to losses during the manual collection of the


loadi-

powder with a rubber spatula (Al-Dhubiab, 2013; Harsha et al., 2017).


Drug

12.5

n.a.
(%)

30

30
ng

However, the ability to process small sample amounts makes a nano


spray dryer very suitable for testing valuable biological materials such
yield (%)

as for example monoclonal antibodies, recombinant proteins, or siRNA-


Product

50–60

50–60

based therapeutics (Amsalem et al., 2017; Heng et al., 2011; Schmid,


n.a.
70

2011; Schoubben et al., 2014). Moreover, nano spray drying enables


the encapsulation of drugs in polymers with a high efficiency of over
95% and adjustable drug loading, as shown by several studies in the
amount (mg)
Solid sample

next titles.
2’000
n.a.

n.a.
n.a.

4.1. Pulmonary (lung)


Particle size

A major advantage of nano spray drying is the optimization of the


particle size and morphology for pulmonary drug delivery, as demon-
0.5–3.0
0.5–4.0

0.5–4.0
(µm)

strated by many studies listed in Table 3. There has been significant


0.9

research activity in the formulation of dry powder aerosols for the


treatment of lung diseases, such as:
Drying gas
(L/min)

• asthma with salbutamol (Arpagaus et al., 2010b; Behara et al.,


130
120

120
n.a.

2014a,b,c; Faulhammer et al., 2018, 2015; Littringer et al., 2013;


Pinto et al., 2017; Son et al., 2013a; Yang et al., 2015; Zellnitz et al.,
T out (°C)

2015a,b, 2014), terbutaline (Behara et al., 2014b), or fluticasone


52–56
40–45

n.a.

(Arpagaus et al., 2010b)


40

• inflammation by dexamethasone (Schafroth et al., 2012),



T in (°C)

pulmonary arterial hypertension with resveratrol (Dimer et al.,


70–85

2015b) or sildenafil (Beck-Broichsitter et al., 2015a),


120

n.a.


70

cystic fibrosis with ketoprofen (Aquino et al., 2014) or azithromycin


(Hindle et al., 2015),
• lung cancer with methotrexate (Harsha et al., 2015a),

Ethanol

Ethanol
Solvent

tuberculosis with capreomycin (Schoubben et al., 2015, 2014,


Water,

Water,
Water

n.a.

2013a, b), ethambutol (Ahmad et al., 2014), or pyrazinamide


(Kaewjan and Srichana, 2016), and
• bacterial infections with amoxicillin (Nguyen et al., 2017), cipro-
floxacin (Farkas et al., 2015; Lee et al., 2013; Longest et al., 2015),
Mannitol, L-leucine,

Mannitol, L-leucine,

gatifloxacin (Lee et al., 2013), clarithromycin (Dimer et al., 2015a),


Lactose, Leucine

or levofloxacin (Merchant et al., 2014).


Poloxamer

Poloxamer
Type of drug, application, wall material (excipient)

The selection of a suitable matrix excipient is essential for the en-


Lactose

capsulation of drugs by nano spray drying to achieve the desired de-


composition of the particles and the drug release in the lungs. Mannitol,
chitosan, leucine, lactose, and trehalose are widely used due to their
high aqueous solubility and low toxicity. The hygroscopic excipient
mannitol is especially advantageous for the treatment of bacterial in-
Salbutamol (Albuterol) (Asthma)

fections in cystic fibrosis (Hindle et al., 2015; Torge et al., 2017).


Terbutaline sulfate (Asthma)
Salbutamol sulfate (Asthma)

Salbutamol sulfate (Asthma)

Chitosan offers several advantages for mucosal delivery, such as low


toxicity and good biodegradability as well as antibacterial activity
Table 3 (continued)

(Cerchiara et al., 2015; Dimer et al., 2015a; Merchant et al., 2014; Ngan
et al., 2014; Nguyen et al., 2017; Rampino et al., 2013). Leucine is a
very popular dispersion enhancer to increase the flowability of nano
spray dried particles, as shown in various pulmonary drug delivery
studies (Aquino et al., 2014; Behara et al., 2014a, 2014b, 2014c; Dimer
et al., 2015a; Feng et al., 2011; Guo and Li, 2013; Kaewjan and

202
Table 4
C. Arpagaus et al.

Different studies on nano spray dried drug delivery applications by oral (gastrointestinal) route (Drug loading (%) = Amount of drug in particles/Mass of particles; Encapsulation efficiency (%) = Amount of drug in
particles/Initial drug amount; n.a. = not available; – = not relevant).
Type of drug, application, wall material (excipient) Solvent T in (°C) T out (°C) Drying gas Particle size Solid sample Product Drug Encap-sulation References
(L/min) (µm) amount (mg) yield (%) loading (%) efficiency (%)

Dexamethasone (Steroid, anti-inflammatory) with poloxamer and Ethanol Acetone 55 n.a. 110 0.4–1.2 500 4–74 – Durli et al. (2014)
surfactants Propanol
Furosemide (Diuretic to treat congestive heart failure and edema) Acetone 100 n.a. 100 (N2/ 0.7–1.7 n.a. 69 Li et al. (2010)
CO2)
Griseofulvin (Antifungal) Methanol, 70 40–45 120 (N2) 3.4–6.5 n.a. n.a. Schmid (2011),
Acetone Schmid et al. (2011)
Indomethacin (Anti-inflammatory, reducing pain and fever) Water, Ethanol 50 n.a. 90 0.6 n.a. n.a. Martena et al.
(2012b)
Nimesulide (Anti-inflammatory, reducing pain and fever) Ammonia 90 n.a. 85 0.2–2.5 n.a. ∼90 Rascioni et al. (2016)
Mecigestone (Steroidal progestin, hormone therapy, treatment of DCM 40 34 135 (N2/ < 10 500 n.a. Nazarov et al. (2016)
gynecological disorders) CO2)
Nicergoline (Blood pressure regulation, blood vessels dilating) Water, Ethanol 50 n.a. 90 0.5–1.6 ∼1’000 91–93 Martena et al.
(2012a)
Dextrans (Model to regulate release Silica particles formed via sol–gel Water 120 58 120 ∼2 n.a. n.a. n.a. n.a. Wang and Friess
kinetics) processing (2017)
Acyclovir (Antiviral and antiherpes Hyaluronic acid, poly(acrylic Water, DCM 85 n.a. n.a. < 0.5 n.a. 75 22 85 Sithole et al. (2017)
agent) acid), hydroxypropyl-cyclodextrin
Amoxicillin (Antibacterial, stomach Gelatine, Carbopol Water 80–120 n.a. n.a. 0.1–0.8 500 64–93 89–93 n.a. Harsha (2013, 2012)
infections, Helicobacter pylori)

203
Curcumin (Antioxidant) loaded solid Compritol ATO 888, Sodium Water 100 n.a. 120 0.5–1.0 n.a. n.a. n.a. n.a. Wang et al. (2016c)
lipid nanoparticles (SLNs) caseinate (Emulsifier), Pectin
(Stabilizer)
Curcumin (food additive, skincare, Stearic acid, Sodium caseinate, Water 100 n.a. 120 0.5–1.0 n.a. n.a. n.a. n.a. Xue et al. (2017b)
natural dye) loaded SLNs Pectin
Curcumin in egg yolk low density Pectin Water 70–120 50–60 120 0.5–1.5 n.a. n.a. n.a. n.a. Zhou et al. (2016)
lipoprotein nanogels
Curcumin in cross-linked egg yolk low Pectin, Carboxymethyl cellulose Water 100 n.a. 120 ∼1 n.a. n.a. n.a. n.a. Zhou et al. (2018)
density lipoprotein nanogels (CMC)
Dexketoprofen Trometamol (Anti- Kollidon (polyvinyl Methanol 120 54 n.a. 0.1–0.7 1’000 n.a. n.a. 36–51 Öztürk et al. (2017,
inflammatory, analgesic delivery) polypyrrolidone), Eudragit 2015)
Eugenol oil nanoemulsion Gum arabic, Lecithin Water, Ethanol 100 n.a. 100–110 0.2–0.5 n.a. n.a. n.a. n.a. Hu et al. (2016)
(Antimicrobial)
Metformin hydrochloride Carbopol Water 100 35 100–110 0.2–0.4 500 74 89 n.a. Harsha (2015)
(Antidiabetic, mucoadhesive)
Nanosuspensions of crosslinked Poly(vinyl alcohol) (PVA) Water 100 n.a. 100 0.1–0.3 n.a. n.a. n.a. n.a. Moshe et al. (2017)
polymeric micelles (Lipophilic
drugs)
Nanoemulsion of peppermint oil Sodium caseinate, Propylene Water 100 n.a. 120 1.0–2.0 n.a. n.a. n.a. n.a. Wang et al. (2016d)
(Antimicrobial), vitamins, glycol, Pectin
minerals, amino acids
Selegiline (Parkinson's disease, Gelatin Water 120 27 100–110 0.1–0.2 n.a. 86–93 98 97–99 Al-Dhubiab (2013)
depression)
Sitagliptin (Antidiabetic, Carbopol Water 80–120 n.a. n.a. 2.0–8.0 500–2’000 64–92 73 n.a. Harsha et al. (2013b)
mucoadhesive)
(continued on next page)
International Journal of Pharmaceutics 546 (2018) 194–214
C. Arpagaus et al. International Journal of Pharmaceutics 546 (2018) 194–214

Srichana, 2016; Merchant et al., 2014; Schoubben et al., 2014, 2013a,

Harsha et al. (2015b,


Wang et al. (2016b)
Wang et al. (2016a)
Xue et al. (2017a)
2013b, 2015; Son et al., 2013a; Yang et al., 2015). The ideal aero-

Cerchiara et al.

Harsha (2015),
dynamic particle diameter for efficient delivery into the deep lung is
about 1–5 µm, which allows penetration and deposition in the alveolar
References

2013a)
regions. Too large particles deposit in the throat and excessively small

(2015)
particles are exhaled. Compared to oral administration, the high bioa-
vailability in the lungs enables a reduction of drug doses, which leads to
a decrease in systemic side effects and benefits patient compliance
Encap-sulation
efficiency (%)

(Kaewjan and Srichana, 2016).


To improve the flowability and the dosing accuracy of nano spray
68–76

dried drug particles in dry powder inhalers, glass beads in the size range
n.a.

n.a.

n.a.

n.a.

of 400–600 µm (Zellnitz et al., 2015b, 2014) or coarse lactose


loading (%)

(60–240 µm) (Faulhammer et al., 2018, 2015) and mannitol (160 µm)
(Pinto et al., 2017) powders are often used as larger carrier particles.
52–64

71–89
Drug

During inhalation, the smaller drug particles separate from the carrier
n.a.

n.a.

n.a.

particles and are deposited in the alveoli (Arpagaus et al., 2010b).


Another recently proposed strategy to achieve almost complete
yield (%)

pulmonary deposition utilizes the hygroscopic growth of inhaled par-


Product

74–88

71–96

ticles in the airways. Nano spray dried powders made of albuterol,


n.a.

n.a.

n.a.

mannitol, L-leucine and poloxamer with an aerodynamic diameter of


about 1.5 μm are considered optimal for delivering a high drug payload
amount (mg)
Solid sample

(Son et al., 2013a,b). Inside the humid airways, the particles take up
100 - 2′000

water and become larger. With this approach, fine particle fractions
(< 5 μm) of 95% are reached. Other studies with asthma drug terbu-
400
n.a.

n.a.

n.a.

taline sulfate (Behara et al., 2014b), antibiotics such as ciprofloxacin


(Farkas et al., 2015; Longest et al., 2015), and anti-inflammatory azi-
Particle size

1.0–10.0

thromycin (Hindle et al., 2015) further demonstrate the feasibility of


0.5–0.8

3.0–5.0

0.4–0.5

0.1–0.6

this enhanced excipient-growth approach.


(µm)

4.2. Oral (gastrointestinal)


Drying gas

100–110
(L/min)

In view of the increasing number of poorly water-soluble drugs,


120

120

120

n.a.

optimizing oral bioavailability remains a major challenge for the


pharmaceutical industry. The formulation of poorly soluble active
T out (°C)

pharmaceutical ingredients as nanosized amorphous solids or en-


27–35
n.a.

n.a.

n.a.

capsulated in a water-soluble polymer matrix has become preferred


40

approaches in recent years.


T in (°C)

Several studies have been conducted to generate nano spray dried


100

100

100

120

oral dosage forms for immediate and sustained drug release, as shown
80

in Table 4. Nano spray drying is suitable for the direct conversion of


pure, poorly water-soluble drugs into dry submicron powders from
organic solutions without wall materials, as shown for example in the
case of diuretic furosemide (Li et al., 2010), pain reducing nimesulide
Solvent

Water

Water

Water

Water

Water

(Rascioni et al., 2016), blood vessels dilating nicergoline (Martena


et al., 2012a), fever reducing indomethacin (Martena et al., 2012b),
anti-inflammatory dexamethasone (Durli et al., 2014), or steroidal
Stearic acid, Sodium Caseinate,

Pectin, Gum Arabic, Alginate,

hormone mecigestone (Nazarov et al., 2016). By nano spray drying,


Compritol ATO 888, Sodium

Rascioni et al. (2016) decreased the particles of nimesulide from 2.9 to


1.4 µm and the degree of crystallinity to 67%. Martena et al. (2012b)
Gelatine, Carbopol

reduced the mean particle diameter of indometacin from 9.2 to 0.6 µm


Caseinate, Pectin

with a crystallinity degree of about 45%, which led to an increase in


Type of drug, application, wall material (excipient)

drug dissolution. The nano spray dried mecigestone particles prepared


Chitosan

by Nazarov et al. (2016) were 7.5 times more bioavailable after oral
Pectin

CMC

administration to laboratory white rats than the starting 200 μm sized


crystalline drug. Durli et al. (2014) showed for dexamethasone dis-
Solid lipid nanoparticles (SLNs) layer-

solved in organic solution that the addition of surfactants allowed an


Vancomycin (Antibiotic, colon drug

increase in process yield during the nano economy drying process.


Solid lipid nanoparticles (SLNs)

Solid lipid nanoparticles (SLNs)

Particle analysis revealed that more than 90% of the nano spray dried
polysaccharide coated

Vildagliptin (Antidiabetic,

particles were submicron.


The wall materials used for the oral administration of encapsulated
Table 4 (continued)

by-layer coated

mucoadhesive)

pharmaceuticals prepared by means of nano spray drying are mainly


water-soluble polymers, such as gelatin, chitosan, compritol, sodium
delivery)

caseinate, pectin, carbopol, carboxymethyl cellulose, gum arabic, algi-


nate, Kollidon and Eudragit. The selection of a suitable wall material for
nanoencapsulation is based on similar factors as for microencapsulation
(Gharsallaoui et al., 2007). The relevant criteria are manifold, including

204
C. Arpagaus et al. International Journal of Pharmaceutics 546 (2018) 194–214

compatibility with the encapsulated active ingredient, suitable release protein-pectin and protein-carboxymethyl cellulose nanogels in order to
properties, high encapsulation efficiency, mechanical strength, storage improve the gastrointestinal stability.
stability, easy emulsification, water solubility and edibility. Self- assembled polymeric micelles are also very popular and a va-
Nano spray drying has also been utilized as a process to dry pre- luable nanotechnology platform for encapsulation and administration
formed nanoparticles, namely, solid lipid nanoparticles (SLNs) (Wang of lipophilic drugs, especially for oral mucosal administration. Moshe
et al., 2016a,b,c, Xue et al., 2017a, 2017b), nanoemulsions (Hu et al., et al. (2017) demonstrated the feasibility of nano spray drying for the
2016; Wang et al., 2016d), nanogels (Zhou et al., 2018, 2016), self- production of stable powders from cross-linked micelles of poly(vinyl
assembled polymeric micelles (Moshe et al., 2017), lipid-polymer hy- alcohol)-g-poly(N-isopropyl acrylamide) in a size range of 100–250 nm,
brid nanoparticles (Sithole et al., 2017), or silica particles formed via which could be redispersed in water in any desired concentration at the
sol–gel processing (Wang and Friess, 2017). In most cases, the pre- time of administration. The production method was described as very
formed nanoparticles were mixed with additional excipients and dis- versatile and could open up new horizons in the application of poly-
persing agents prior to nano spray drying to fabricate fine powders. meric micelles and other self-organized polymeric nanomaterials in
SLNs are regarded as promising nanoscale delivery systems for diagnostics and therapies.
highly lipophilic drugs with limited bioavailability and biological effi- Sithole et al. (2017) encapsulated the antiherpes drug acyclovir in
cacy. SLNs offer a high loading capacity in their lipid core compared to hybrid biopolymeric nanoparticles, which were prepared from cross-
other colloidal delivery systems. Several studies have shown that SLNs linking hyaluronic acid with poly(acrylic acid). The spray dried nano-
can be successfully formulated using nano spray drying technology. particles were smaller than 500 nm in average and the solubility of
Wang et al. (2016a) prepared pectin (a polysaccharide from citrus peel) acyclovir could be improved by 30% compared to the conventional
coated SLNs made of Compritol ATO 888 (glyceryl behenate) and so- formulation, consequentially enhancing its bioavailability as an oral
dium caseinate (milk protein). The nano spray dried SLNs powders drug delivery system.
were in the range of 1–3 µm and showed excellent stability. Wang et al. Silica-based sol–gel materials are another promising delivery form
(2016b) coated SLNs made from Compritol 888 ATO with various for controlled drug release purpose. Its main advantage is that drugs
natural polysaccharides by nano spray drying. Wang et al. (2016c) can be entrapped in a porous glass network without the formation of
further tested the feasibility of cross-linking the polymeric double layer covalent bonds between the active ingredient and the matrix. Wang and
made of sodium caseinate and pectin for oral delivery of curcumin, a Friess (2017) used nano spray drying to produce water-soluble silica gel
lipophilic bioactive studied as a model drug and nutrient. The cross- carrier particles based on a new silicate-based precursor. Dextrans of
linking significantly improved the physico-chemical properties of the different molecular weights (1, 5 and 60 kDa) were incorporated as
SLNs, resulting in higher encapsulation efficiency and loading capacity, model compounds to regulate the release kinetics. A sustained release
better stability and slower release profile in simulated gastrointestinal over 35 days could be achieved for 60 kDa dextran.
conditions. The cross-linking further helped forming homogenous nano
spray dried powders of 0.5–1 µm size. Encapsulation of curcumin in 4.3. Intravenous (blood circulation)
SLNs also significantly improved its antioxidant activity in aqueous
condition compared to free curcumin. Wang et al. (2016d) applied this Site-specific drug delivery is very important to prevent undesired
encapsulation technology to develop powder formulations of instant side effects. Thereby, the particle size plays a crucial role in target drug
functional drinks from a pectin-casein with incorporated peppermint oil distribution, as shown in Table 5. In this context, Harsha et al. (2014)
as a potential antimicrobial agent. The formulation allowed the si- prepared 5–15 µm sized gelatin microspheres containing the anticancer
multaneous encapsulation of hydrophilic and hydrophobic vitamins as drug carboplatin using nano spray drying for treating lung cancer. After
well as mineral salts and showed a prolonged shelf life. Xue et al. intravenous injection to albino mice, the drug concentration in the
(2017a) fabricated submicron SLNs by homogenizing melted stearic lungs was significantly higher than in any other tissues e.g. liver,
acid directly in an aqueous phase containing sodium caseinate and spleen, or in the blood. It was stated that the particles could be used not
pectin. After nano spray drying, the obtained dry SLN powders could be only for sustained drug release, but also for targeted delivery of ther-
effectively redispersed in water without variation of dimension, shape apeutic agents to specific organs in the body. The development of new
and morphology. In another study, Xue et al. (2017b) encapsulated cancer chemotherapeutics is a constant need. In this application area,
curcumin in SLNs and produced nano spray dried powders of 0.5–1 μm Anzar et al. (2018) explored the nano spray drying method for the
size with uniform distribution and no visible aggregation. The pre- encapsulation of simvastatin in PLGA. Simvastatin, which is typically
sented manufacturing method of nanoparticles is solvent-free and used to lower cholesterol levels, has been investigated for the effective
promises to encapsulate heat-sensitive lipophilic drugs that are stable in treatment of breast cancer. The process parameters were optimized to
SLNs for oral administration. Hu et al. (2016) produced spherical dry obtain particles of about 260 nm. In vitro cytotoxicity studies revealed
powders (200–500 nm) from eugenol oil nanoemulsions with natural an enhanced anticancer activity with increasing drug concentration and
lecithin and gum arabic. Ethanol was used as a co-surfactant, which exposure time. Overall, the nanoparticles were found to be appropriate
improved the morphology and homogeneity of powders during nano for the treatment of solid tumor, e.g. breast cancer via intravenous
spray drying. The excellent redispersibility of the nanoemulsion pow- administration. Recently, Harsha et al. (2017) encapsulated salbutamol
ders and the strong antimicrobial effect was seen as an incentive for in albumin microspheres. After fast intravenous injection in rabbits, the
other potential applications of nanoemulsions with other ingredients. particles with about 8 µm mean size were widely deposit in the pul-
Nanogels made of egg yolk low density lipoprotein and poly- monary capillaries of the lungs. The systemic administration of opti-
saccharide are also being explored as oral application systems with mally sized particles is seen as an efficient alternative to currently used
promising encapsulation and release potentials. Zhou et al. (2016) re- inhalation-based delivery to the lung. In another application, Panda
ported on a pH- and heat-induced method for the fabrication of such et al. (2014) co-entrapped the antipsychotic drugs clozapine and ris-
nanogels with a diameter of only 58 nm as nutrient delivery systems. peridone in 250 nm PLGA nanoparticles by nano spray drying and ob-
The nano spray dried powders produced from the nanogels exhibited an served sustained drug release profiles in vitro over several days. The
average particle size of about 1 µm and could be redispersed in water drugs were evenly dispersed within the PLGA particles without any
and maintained the nanoscale size. Encapsulation of curcumin into the crystalline content. The prepared particles could be used as an in-
nanogels was effectively achieved. The nanogels showed a pH-depen- jectable formulation to cross the blood–brain barrier for the treatment
dent controlled release profile in gastric and intestinal conditions, de- of psychotic disorders, such as Schizophrenia. Last but not least, small
monstrating potential as oral delivery systems for lipophilic nutrients interfering RNAs (siRNAs) are promising therapeutics for the treatment
and drugs. Furthermore, Zhou et al. (2018) chemically cross-linked of a wide variety of genetic disorders and for the silencing of

205
Table 5
Published studies on nano spray dried drug delivery applications by different routes other than pulmonary and oral routes (Drug loading (%) = Amount of drug in particles/Mass of particles; Encapsulation efficiency
(%) = Amount of drug in particles/Initial drug amount; n.a. = not available; – = not relevant).
C. Arpagaus et al.

Type of drug, application, wall material (excipient) Solvent T in (°C) T out (°C) Drying gas Particle size Solid sample Product Drug Encap-sulation References
(L/min) (µm) amount (mg) yield (%) loading (%) efficiency (%)

Intravenous (injection) route


Carboplatin (Drug targeting lung Gelatin Water 85–115 30–50 n.a. 5–15 n.a. 88 72 n.a. Harsha et al. (2014)
cancer passively after intravenous
injection)
Clozapine and risperidone PLGA, Polyvinylalcohol (PVA) DCM Water 60–80 31–38 112–118 (N2) 0.2–0.4 100 48–64 6–13 89–98 Panda et al. (2014)
(Antipsychotic drugs to treat
Schizophrenia)
Salbutamol sulphate (Asthma, Bovine serum albumin (BSA) Water 80–120 20 90–95 1.2–10.0 n.a. 86 n.a. 72 Harsha et al. (2017)
targeting lungs passively after
intravenous injection)
Simvastatin (Breast cancer) PLGA, PVA DCM, Water n.a. n.a. n.a. 0.1–0.7 115 n.a. 9 63 Anzar et al. (2018)
siRNA-loaded human serum albumin PLGA, PEG-PLGA DCM, Chloroform, 50 < 39 118–121 0.3–1.0 150–200 30–60 <1 n.a. Amsalem et al.
nanoparticles (Silencing gene Acetonnitrile (N2/CO2) (2017)
expression)

Topical (skin or wound) route


Amoxicillin (Antibacterial to inhibit Chitosan Water, Acetic acid 120 80 130 0.1–0.4 n.a. 60–63 82 n.a. Ngan et al. (2014)
bacterial growth)
Dexamethasone (Anti-inflammatory Poly(ε-caprolactone), Acetone Water 55 n.a. 110 0.4–2.3 100 72–90 47 94 Beber et al. (2014)
drug to treat skin diseases) Deoxycholate
Econazole nitrate (Antifungal drug) Hydroxypropyl-β- Ethanol 95 45 115 0.1–0.3 n.a. 78–87 48–55 n.a. Maged et al. (2017)
cyclodextrin, Stabilizer Tween

206
Gentamicin sulfate (Antibacterial drug Alginate, Pectin Water 90 n.a. 100 0.3–1.0 n.a. 65–91 23–28 70–83 De Cicco et al.
for wound dressing) (2014)
Soy bean isoflavones (Skin cancer Hypromellose acetate Water, Ethanol 60 29 100 0.3–1.3 n.a. 60–88 n.a. 62–86 Del Gaudio et al.
treatment, anti-ageing agent, succinate (2017)
chemopreventive)
Soy bean isoflavone extract CMC Water n.a. n.a. n.a. ∼0.65 n.a. n.a. n.a. 78–89 Del Gaudio et al.
(2016)

Ophtalmic (eye) route


Dexamethasone (Anti-inflammatory steroid, eye drop solutions to cure Ethanol 50 35 100 (N2/ 0.4–2.2 10–125 n.a. – Baba and Nishida
ophthalmic disorders) CO2) (2013)
Fluorometholone (Anti-inflammatory steroid, eye drop solutions to cure Ethanol 50 35 100 (N2/ 0.4–1.2 10–125 n.a. Baba and Nishida
ophthalmic disorders) CO2) (2013)
Calpain inhibitors (Drugs to cure Alzheimer’s and Parkinson’s diseases) Ethanol 50–100 35 100–150 0.1–1.3 n.a. n.a. Baba and Nishida
(N2/CO2) (2012)
Dirithromycin (Ocular bacterial Kollidon SR (Polyvinyl Methanol 90 30–40 n.a. 0.3–0.5 n.a. n.a. n.a. 25–44 (Basran, 2017)
infections) alcohol and polyviniyl
pyrolidone)
Intravesical (bladder) route
Thiolated glycol chitosan conjugates N-acetylcysteine, Glutathione, Water, Acetonitrile 40 30–32 100 (N2/ 0.2–1.4 n.a. 15–25 36–73 64–100 Denora et al. (2016)
(Mucoadhesive, local bladder Glycol, Chitosan CO2)
diseases)

Intraperitoneal route
Paclitaxel (Anticancer therapy, Low molecular weight poly(3- Chloroform 61 n.a. 120 (N2/ 0.7–3.7 2’500 n.a. 13 n.a. Bonartsev et al.
cytostatic) hydroxybutyrate) CO2) (2017), Zernov et al.
(2017)

Cerebral (brain) route


Nimodipine (Dilatation regulator in PLGA DCM 45 n.a. 120 0.7–3.7 n.a. n.a. 10–40 93–98 Bege et al. (2013)
brain arterioles)
International Journal of Pharmaceutics 546 (2018) 194–214
C. Arpagaus et al. International Journal of Pharmaceutics 546 (2018) 194–214

transcription during gene expression. Amsalem et al. (2017) loaded enhanced therapeutic efficacy of the prepared mucoadhesive formula-
solid cross-linked human serum albumin primary nanoparticles tions suitable for the efficient treatment of severe ocular bacterial in-
(∼100 nm) with siRNA, and encapsulated them in PLGA or a mixture of fections. Baba and Nishida (2012) prepared calpain inhibitor steroid
PEG and PLGA by nano spray drying. The drying temperature was nanocrystals using nano spray drying. The particle size was controlled
around 50 °C to prevent damage to the heat-sensitive siRNA. The pre- by means of selecting the spray mesh sizes and the concentration of the
pared solid nano-in-nanoparticles had a median size of about ethanol-dissolved drug solution. The average particle sizes were 378,
580–770 nm and exhibited stable spherical shape with a unique inner 527, and 813 nm in comparison to the spray mesh sizes of 4.0, 5.5, and
morphology. Freeze-fracture SEM revealed many encapsulated siRNA- 7.0 μm. It was stated that the formed calpain inhibitors might be a
loaded nanoparticles embedded within a PLGA polymeric fiber matrix promising drug for curing intractable diseases such as Alzheimer’s
surrounded by a dense polymeric wall. The structural integrity and the disease and Parkinson’s disease. In a subsequent study, Baba and
gene silencing activity of the double encapsulated siRNA were fully Nishida (2013) prepared submicron steroid nanocrystals of fluor-
preserved after encapsulation. The nanoscale powder form enabled ometholone and dexamethasone with the potential use as eye drop
systemic administration, e. g. by intravenous injection, and thus the solutions the treatment of ophthalmic disorders. More details have been
controlled release of siRNA-based therapeutics and other sensitive nu- provided in Table 5.
cleic acids.
4.6. Intraperitoneal
4.4. Topically (skin or wounds)
In cancer therapy, paclitaxel is one of the most widely applied an-
In addition to oral, pulmonary, and intravenous administration titumor cytostatics. Zernov et al. (2017) encapsulated paclitaxel in low
pathways, researchers have also produced nanocapsulated drugs molecular weight poly(3-hydroxybutyrate) by nano spray drying (see
through nano spray drying that can be applied to the skin or wounds Table 5). The synthesized particles had a mean diameter of 2.7 µm and
during surgery (Table 5). De Cicco et al. (2014) loaded alginate-pectin a regular spherical shape with a smooth surface. The crystallinity of the
nanopowders with 25% antibacterial gentamicin. This formulation polymeric microparticles was about 65%. The study of Bonartsev et al.
turns into a gel when administered locally on a wound. The produced (2017) demonstrated a significant antitumor activity of the paclitaxel
particles with a 4.0 µm spray mesh ranged between 310 and 405 nm. All loaded microparticles both in vitro on murine hepatoma cells and in vivo
formulations were obtained with good production yield ranging from by intraperitoneal administration against transplanted tumor models in
65 to 91%. Sustained drug release was obtained, achieving a total drug mice, i.e. murine Lewis lung carcinoma and xenografts of human breast
permeation period of 3–6 days. Ngan et al. (2014) prepared amoxicillin cancer. The sustained release of paclitaxel over 2 months makes the
loaded chitosan nanoparticles by nano spray drying with improved developed biodegradable particles suitable for application in long-term
antibacterial activity. With a highly diluted chitosan solution of 0.025% anticancer therapy.
(w/v) and a 4.0 µm spray mesh the particle sizes could be adjusted by
the molecular weight of the chitosan in a range of 95–336 nm. The 4.7. Intravesical (bladder)
antibacterial activity against Staphylococcus aureus bacteria was two
times more effective than that of pure amoxicillin. The study demon- Mucoadhesive drug delivery systems offer the advantages of ex-
strated that the prepared nanoparticles strongly inhibited bacterial tending the residence time at the site of interest. Denora et al. (2016)
growth. Beber et al. (2014) dried submicron polymeric particles con- nano spray dried mucoadhesives conjugates of N-acetylcysteine- and
taining anti-inflammatory dexamethasone and prepared oil-in-water glutathione-glycol chitosan polymer for intravesical drug delivery (see
emulsion creams for local administration to the skin. By optimizing the Table 5). In particular, nano spray drying provided spherical particles
powder properties, the creams showed controlled drug release kinetics with an average size of 0.8 µm suitable for vesical administration by a
and a significant increase in retention of drug in the epidermis com- catheter. The prepared particles exhibited good mucoadhesive proper-
pared to formulations containing free drug. Maged et al. (2017) for- ties in a wide range of urine pH (5.0–7.0) and are seen as effective
mulated cyclodextrin nanoparticles incorporating antifungal econazole, formulation for intravesical drug delivery and treatment of local
which is used to treat skin infections. The nano spray dried particles bladder diseases. The drug residence time in the bladder could be in-
ranged between 185 and 325 nm and revealed high yields of 79–85% creased, and subsequently the efficacy of therapy. In this perspective,
and drug loadings of about 50%. Del Gaudio et al. (2017) produced further studies are in progress.
hypromellose acetate succinate particles loaded with a soy isoflavones
extract by nano spray drying. Particles in submicronic range (mean size 4.8. Cerebral (brain)
around 550 nm) and narrow size distribution with high encapsulation
efficiency (up to 86%) were obtained with enhanced skin penetration Nimodipine is a substance that regulates the dilatation of blood
performance. In a previous study, Del Gaudio et al. (2016) stabilized vessels, particularly the cerebral vasculature in the brain. Originally it
the isoflavone extract in carboxymethyl cellulose and enhanced its af- was developed for the treatment of high blood pressure. Bege et al.
finity with aqueous media. The permeation through biological mem- (2013) encapsulated nimodipine in PLGA particles by nano spray
branes increased up to 4.5 times compared to raw soy isoflavone ex- drying. The drying conditions are shown in Table 5. The particles
tract. In a nutshell, the nano spray dried formulations were able to ranged from 1.9 to 2.4 µm and were suspended in a fibrin sealant as an
increase the bioavailability of soy isoflavones and could be used as in situ depot system for treatments related to brain surgery. The fibrin
topical delivery systems for skin cancer treatment or as anti-ageing glue is biodegradable and stable in the cerebrospinal fluid inside the
agent enclosed in cosmeceutical products. brain and the spine. The PLGA particles were also colored with en-
capsulated fluorescent coumarin dye to visualize the homogeneity of
4.5. Ophthalmic (eye) the suspended drug particles in the adhesive. Nimodipine loads of
10–40% and high encapsulation efficiency could be achieved, which
Colloidal drug delivery systems play an important role in the en- make the depot forming device promising for the local treatment of
hancement of the ocular bioavailability of active pharmaceutical in- vasospasm after subarachnoid haemorrhage (bleeding).
gredients, especially for topical instillations. In this application field,
Basran (2017) incorporated the antibacterial agent dirithromycin into 5. Characterization and analysis of nano spray dried drugs
Kollidon-based polymeric nanoparticles (0.3–0.5 µm) by nano spray
drying. The encapsulation enabled to extend the release up to 6 h with Characterization of nano spray dried drugs is very important to

207
C. Arpagaus et al. International Journal of Pharmaceutics 546 (2018) 194–214

provide in-depth information about their quantitative and qualitative sharp tip without any contacts. In fact, topography study of these
parameters, so that these nanocarriers can be formulated appropriately powder particles is possible through AFM. Finally, CLSM can capture
for different applications and administration routes. Properties such as fluorescent-enriched images of the samples revealing the position of the
morphology, particle size and size distribution, surface properties and core/wall materials and their distribution within the powder particles.
physicochemical attributes can be analyzed in nanoencapsulated drugs
obtained from a nano spray drier (Faridi Esfanjani et al., 2017). These 5.2. Size and surface analysis of powder particles
properties affect the storage, stability, functional characteristics, release
and bioavailability of the final pharmaceutical products (Jafari et al., Many parameters of the drug-loaded carriers including stability,
2017a). On the other hand, a deep knowledge in this area could help appearance, rheology, release profile, etc., are influenced by their size
researchers to better design a drug delivery system without any side (Faridi Esfanjani and Jafari, 2016). The size analysis results of nano
effects and with minimum doses for controlled and target delivery. In spray dried powder particles can be presented as particle size dis-
this section, a brief overview of common and most important analytical tribution (PDS), polydispersity index (PDI), Sauter or Volume mean
methods for the characterization of nano spray dried drugs will be diameter, Span, and Z-average (based on intensity, number, or volume
provided. of particles). Also, the size distribution of powder particles can have a
single peak (optimum distribution) or multiple peaks, e.g., two peaks
5.1. Morphology of powder particles (bimodal distribution) as a result of secondary droplet breakup or the
generation of satellite droplets during atomization (Al-Dhubiab, 2013;
One of the important properties of nano spray dried powders is their Harsha et al., 2013a; Littringer et al., 2013; Rampino et al., 2013).
morphology, which plays an important role on the functional and re- There are different analytical instruments to measure the size of nano
lease characteristics of nanoencapsulated products (Jafari and spray dried particles, including Statical Light Scattering (SLS), Dynamic
Esfanjani, 2017). Analyzing the morphology of powder nanoparticles Light Scattering (DLS), Gravitational Settling and Centrifugation, and
can provide useful information about their structure, shape, and size Laser-Induced Breakdown Detection (LIBD).
through capturing high quality images by advanced microscopic ana- Due to engagement of different ionic materials such as phospholi-
lysis like Electron Microscopy, including Scanning Electron Microscopy pids, phenolic compounds, proteins, polysaccharides, etc. in na-
(SEM), Field Emission SEM (FESEM), and Transmission Electron Mi- noencapsulated powders produced by nano spray drying, there are
croscopy (TEM); Atomic Force Microscopy (AFM); and Confocal Laser some electric charges at the surface of these nanocarrier particles,
Scanning Microscopy (CLSM). Explaining the principles of these tech- which could strongly affect the stability, release rate, bioavailability,
niques is outside the scope of this paper and readers are referred to and physicochemical properties of the final products (Assadpour and
study specialized literature in this field. High-resolution images at a Jafari, 2017). The most common way to express these surface electric
magnification of ×103–×106 and a resolution smaller than 1 nm can be charges is the measurement of the zeta-potential, which typically could
obtained by Electron Microscopy techniques from nano spray dried be in the range of +100 to −100 mV. Briefly (Basran,
powders as shown in Fig. 5. 2017): ± 0–10 mV (highly unstable), ± 10–20 mV (relatively
By applying AFM, it is possible to visualize nano spray dried par- stable), ± 20–30 mV (moderately stable), and > ± 30 mV (highly
ticles in three dimensions (Ghasemi et al., 2017). This technique pro- stable); in other words, those particles with a zeta-potential > +30 mV
vides useful information about the structure of nanoparticles at the or < −30 mV are considered strongly cationic and anionic, respec-
atomic and molecular level by scanning the surface of samples with a tively with enough stability against attraction forces and minimum

Fig. 5. Examples of the SEM characterization results for nano spray dried powder particles containing: A: Salbutamol sulfate particles (1% (w/w) solid concentration,
nano spray dried at 100 °C); reprinted with permission from Littringer et al. (2013); B: Powders of albuterol sulfate in mannitol, L-leucine and poloxamer 188
(30:48:20:2 mixing ratio, 0.5% (w/w) solid concentration in water–ethanol solution (80:20), nano spray dried at 70 °C); reprinted with permission from Son et al.
(2013a,b); C: Aggregated cyclosporin nanoparticles in PLGA (50:50, 15 kDa, scale bar 300 nm) (dissolved in DCM and nano spray dried at 29 °C); reprinted with
permission from Schafroth et al. (2012); D: Uniform trehalose particles prepared with the addition of 0.005% (w/w) polysorbate 20 (0.1% (w/w) solid concentration,
nano spray dried at 120 °C); reprinted with permission from Schmid et al. (2011); E: Particles of β-galactosidase encapsulated in trehalose (1:2 (w/w) mixing ratio,
5% (w/w) solid concentration, nano spray dried at 80 °C); reprinted with permission from Bürki et al. (2011); F: Bovine serum albumin particles prepared with 0.05%
(w/v) Tween 80 (0.5% (w/w) solid concentration, nano spray dried at 120 °C); reprinted with permission from Lee et al. (2011).

208
C. Arpagaus et al. International Journal of Pharmaceutics 546 (2018) 194–214

aggregation due to strongly repulsive forces. It is possible to measure 6. Bioavailability and release of nano spray dried drugs
the zeta-potential through some lab instruments, including electro-
phoresis, laser Doppler velocimetry, and zeta sizer (Malvern, UK). 6.1. Entrance of nano spray dried drugs into the blood stream
Another important characteristic is the surface composition analysis
of powder particles produced by nano spray drying, which can be There are many routes for the administration of drugs such as pul-
achieved by X-ray photoelectron spectroscopy (XPS). By this technique, monary, oral, topical, and injection procedures. In general, spray dried
it is feasible to determine what elements/compounds are existent at the nanoparticles containing encapsulated drugs are in the form of pow-
surface layer (a few nm information depth) of dried powder particles, so ders, which are converted into pills for oral consumption by the pa-
that an in-depth analysis of the particle surfaces could help researchers tients. After oral administration, the entry of nano spray dried drugs
to evaluate the successfulness of process and predict the behavior of into systematic circulation is restricted by several hurdles including
these nanocarriers in real situations. Jafari et al. (2008a) identified the acidic conditions of the stomach, brush border membrane and proteases
unloaded fish oils in nano-encapsulated powders by analysing the sur- in the gut lumen, metabolism by the liver enzymes, and tightly-bound
face composition through XPS. The relative atomic concentration (%) of intestinal epithelial cells (Jafari and McClements, 2017). Nano spray
carbon, oxygen, and nitrogen in the surface layer of the powder sample dried particles loaded with drugs can pass the intestinal epithelium by
was explored. As a consequence, the percent composition (relative diffusing through the cells (transcellular route) (Harsha, 2015; Harsha
coverage) of the different components, such as fish oil in the surface et al., 2013a). In fact, nanocarriers produced by nano spray drying can
layer of powder prepared from whey protein concentrate (WPC) and have a higher bioavailability; i.e., higher availability of the drugs for
maltodextrin was calculated by using the relation in a matrix formula the human body.
as: Nano spray dried drugs that are consumed via the oral route for
reaching the target tissue must adhere to the mucus and cross the
CE = αCS + βCP + γ CO mucosal layer as the first barrier (Jafari et al., 2017b). This layer, which
covers the epithelium, is retarding the transport of drugs by inhibiting
OE = αOS + βOP + γ OO the direct adhesion to the epithelial cells. The nanoencapsulated drugs
should be able to enter the blood stream to trigger the targeted release.
Viscoelasticity is a key factor for the mucus layer, because based on this
NE = α NS + β NP + γ NO parameter, there are two types of loosely and firmly adherent mucus
layers. pH also varies throughout the gut; in the stomach, the pH of the
where C, O, and N are the relative atomic concentrations of carbon, mucus is about 1–2 on the luminal surface to nearly neutral (pH = 7) at
oxygen and nitrogen in encapsulated powder (CE, OE and NE), mal- the epithelial surface. This pH will be partially neutralized in the
todextrin (CS, OS and NS), protein, i.e., WPC (CP, OP and NP), and fish duodenum and steadily increases through the rest of the gastro-
oil (CO, OO, NO); which are values obtained from the areas of the C 1s, O intestinal tract, so that the pH reaches 7–8 in the colon and rectum
1s, and N 1s at the XPS peaks (Fig. 6). For instance, spray dried powder (Jafari et al., 2017a). It should also be noted that the mucus layer has a
particles made with whey protein concentrate and maltodextrin in the hydrophobic nature due to presence of lipids, glucolipids, and cer-
proportion of 1:3 and containing 20% nanodroplets of fish oil had a amides. Nano spray dried drugs need to be mucoadhesive to pass
surface elemental composition of 26% carbon, 4.5% nitrogen, and through this layer and then, penetrate and move from the epithelial
69.5% oxygen. It was calculated that these powder particles have a cells into the blood stream. There are many theories and mechanisms
surface oil coverage of about 15 to 35% based on the initial emulsifi- for this process, which however, is outside the scope of this paper.
cation technique and the emulsion droplet size for the spray dryer feed
(Jafari et al., 2008a).
6.2. Release of drugs from nano spray dried particles

5.3. Physicochemical analysis of nano spray dried powders Another important factor for nano spray dried drugs is the release
rate and process within the body; many release mechanisms have been
Beside morphological, size, and surface analysis of powder particles, developed to achieve both temporal and sustained controlled release of
it is important to measure some other properties such as formation of nanocarriers (Assadpour et al., 2017). These release theories can be
chemical bonds, crystalline and amorphous nature, thermal properties, classified into partitioning, dissolution, osmosis, diffusion, swelling,
magnetic characteristics, etc. in nanocarriers loaded with bioactive and erosion processes. There are some analytical techniques including
compounds (Jafari et al., 2017a). Characterization of crystalline or
amorphous powders and crystallographic structures of particles con-
taining various drugs can be performed by X-ray diffraction (XRD)
(Fontana et al., 2014; Merchant et al., 2014; Panda et al., 2014;
Rascioni et al., 2016).
Another useful technique is Differential Scanning Calorimetry
(DSC), which can provide some details about thermal properties of
powder particles including glass transition temperature (Tg), heat ca-
pacity (Cp), melting point (Tm), and recrystallization time of nano-
carriers (Baba and Nishida, 2013; Martena et al., 2012a; Son et al.,
2013a). On the other hand, identification of different materials and
compounds within the powder and their chemical interactions is
achieved through Fourier Transform Infrared Spectroscopy (FTIR)
(Basran, 2017; Cerchiara et al., 2015; Fontana et al., 2014; Merchant
et al., 2014; Wang et al., 2016c). Finally, the data obtained by Nuclear
Magnetic Resonance (NMR) can reflect simultaneously the chemical
nature and the molecular mobility of individual components (Ahmad Fig. 6. An example of surface composition analysis of powder particles loaded
et al., 2014; Basran, 2017; Merchant et al., 2014; Öztürk et al., 2017). with nanoencapsulated bioactives and produced by spray drying; reprinted with
permission from Jafari et al. (2008a,b).

209
C. Arpagaus et al.

Table 6
Common mathematical models for the analysis of drug release from nano spray dried particles (reprinted with permission from Jafari et al., 2017b).
Type of model Model Equation Model parameters

Zero-Order C = C 0 − K0 t • CC ==Amount of drug release or dissolved


0
• K = Zero Initial amount of drug in solution
0
• t = time order rate constant

First Order Kinetic Log C = Log C0 − Kt/2.303 0
• CK ==Initial concentration of drug
First order constant
• t = time

Higuchi C = [D (2qt − Cs) Cst]1/2 • CD == total amount of drug release per unit area of the matrix
• qt =diffusion coefficient for the drug in the matrix
• C = total amount of drug in a unit volume of matrix
s
• t = timedimensional solubility of drug in the polymer matrix
• Ct
Korsemeyer-Peppas* Ct
= kt n C∞
• = fraction of drug release at time t
C∞
• kn == rate constant
• release exponent
Hixson-Crowell 1 1 t of drug released in time t
Ct3 −Ct3 = KHC t
• CC =amount
0 amount of drug in the tablet
• K =initial
HC

210
• =rate constant for Hixson-Crowell equation
Weibull −(t − T ) ⎤ b of dissolved drug as a function of time t
C = C0 [1−exp ⎡
a
• CC0==amount of drug being released.
⎣ ⎦ • T = lagtotaltimeamount
measured as a result of dissolution process parameters
• a = scale parameter
• b = shape of dissolution that describes the time dependence
• curve.

Hopfenberg n M is the amount of drug in time t


Mt k0 . t t
= 1− 1−
M∞ C0 . a
( ) • M is the total amount ofdissolved
∞ drug dissolved when the pharmaceutical dosage form is exhausted
• k is the erosion rate constant
o
• C is the initial concentration of drug in the matrix
o
• a is the initial radius for a sphere or cylinder or the half thickness for a slab
• n is 1, 2 and 3 for a slab, cylinder and sphere respectively

Baker–Lonsdale 2 is the drug release amount at time t M∞ is the amount of drug released at an infinite time
3 Mt 3 Mt • Mt
f = 1− 1− = kt
2 ( M∞ ) M∞ • K is the release constant which corresponds to the slope of the graph when plotted as [d(Mt/M∞)]/dt with respect to the root of time inverse
Release exponent (n) Nutrient transport mechanism Rate as a function of time

n = 0.5 Fickian diffusion t −0.5


0.5 < n < 1 Non Fickian diffusion t n−1
n=1 Case II transport Zero order release
n Higher than 1 Super Case II transport t n−1

* n values and related release mechanisms.


International Journal of Pharmaceutics 546 (2018) 194–214
C. Arpagaus et al. International Journal of Pharmaceutics 546 (2018) 194–214

destructive and non-destructive methods to measure the release of ultrafine spray and a highly efficient electrostatic powder collector to
drugs from nanoencapsulation systems, so that it can be predicted what extend the size spectrum of separable particles to the nanoscale. The
happens to encapsulated drugs when administered into the human fabrication of nanoparticles by nano spray drying is relatively simple
body. and fast, which make the technology suitable for processing smallest
Characteristics of the nano spray dried drugs such as size, shape, sample amounts ranging from 10 mg to 2.5 g with uniquely high yields
surface charge, wall composition, molecular weight, etc. play a major of over 95%. The high yields enable the economical use of valuable
role on the release and bioavailability of drugs. For example, prolonged active pharmaceutical ingredients, like pure drugs and drug loaded
circulation and accumulation in the targeted tissue and enhanced dif- polymeric particles. Tiny spray dried particles can be formed down to a
fusion in the tissue can be obtained by applying carriers with a particle size of only 100 nm with diluted solutions of 0.1–1% (w/v) solids
size of 20–100 nm. Beck-Broichsitter et al. (2015a) reported for ex- concentration. The drying process is gentle and contributes to main-
ample that smaller particles exhibit a higher speed of active compound taining the stability and activity of heat-sensitive materials, such as
release compared to larger ones, due to the difference in exposed sur- peptides, proteins, hormones and amino acids. Different particle
face area, as evidenced by sildenafil-loaded polymeric particles. Re- morphologies can be created, including dense, hollow and porous
garding the influence of molecular weight, it has been shown that the particles with spherical, wrinkled, shrinked or donut shapes. The effects
release kinetics of active compounds encapsulated in PLGA polymers on the powder properties, like particle size, morphology, yield, pro-
with a low molecular weight are faster compared to those with a high ductivity, encapsulation efficiency, drug loading, release profile and
molecular weight, due to the higher hydrolysis and degradation rate stability depend on the selected process parameters. The most im-
(Panda et al., 2014). Also, a PLGA polymer with low molecular weight portant adjustable parameters are the inlet temperature, drying gas
can provide sustained release for four to six weeks (Schafroth et al., flow rate, spray mesh size, solvent, solids concentration, and selection
2012). Release profiles typically exhibit two phases. In the first burst of the corresponding excipients, stabilizers and surfactants. Depending
phase, about 10–30% of the active compound is released due to free on the application, an optimized set of process parameters can be
binding at the surface of the particles (Harsha et al., 2015b, 2013a). The found. Nano spray drying has been successfully applied for a wide
rest of the active compound is released in a second phase for a longer variety of pharmaceutical applications, such as increasing the bioa-
duration, known as sustained release (Harsha, 2015, 2013; Harsha vailability of poorly soluble drugs by nanoisation and structural mod-
et al., 2013a). ification, as well as encapsulation of nanoparticles, nanoemulsions or
Al-Dhubiab (2013) developed selegiline loaded gelatin nanospheres nanosuspensions in biocompatible polymeric wall materials for sus-
(∼100 nm) by nano spray drying and evaluated the drug release in tained drug release and high bioavailability. High encapsulation effi-
vitro. The encapsulation provided a controlled release of the drug over a ciencies of over 95% are achieved by adjustable drug loadings. The
period of 10 h. The prepared nanospheres were considered as an effi- prepared drug loaded particles are administered in various ways, in-
cient oral formulation of selegiline for the treatment of Parkinson's cluding pulmonary, oral, intravenous, topically, ophthalmic, in-
disease. In vivo studies need to be carried out to substantiate these traperitoneal, intravesical, or even cerebral, which underlines the ver-
findings. Harsha et al. (2015a,b, 2013a) encapsulated the antidiabetic satility of the nano spray drying technology. To further explore the
drugs vildagliptin and metformin in gelatin and carbopol for mu- potentials of nano spray drying technology future research should focus
coadhesive oral administration with sustained release. Spherical parti- on the further commercialization of this technology on an industrial
cles in the range of 0.1–0.6 µm with smooth surface were obtained by scale. The applications discussed so far are restricted to the laboratory
nano spray drying with up to 90% drug loading and high product scale. There is an increasing need for scale-up to pave the way for
yields. Complete drug release could be delayed by up to 12 h. The same translating nano spray dried pharmaceutical products from bench-to-
research group also designed nano spray dried oral dosage forms of bedside. The application trends are in the areas of pulmonary drug
amoxicillin in gelatin nanoparticles to treat Helicobacter pylori infection delivery, nanotherapeutics, the encapsulation of nanoemulsions with
in stomach (Harsha, 2013, 2012) or antidiabetic sitagliptin in mu- poorly water-soluble active ingredients and the formulation of nano-
coadhesive carbapol microspheres (Harsha et al., 2013b). Öztürk et al. crystals for a higher bioavailability.
(2017, 2015) loaded dexketoprofen in Kollidon and Eudragit nano-
particles (0.7–0.7 µm) by nano spray drying and extended the drug References
release and the analgesic efficacy following oral application to mice.
Cerchiara et al. (2015) encapsulated the antibiotic vancomycin in Ahmad, M.I., Ungphaiboon, S., Srichana, T., 2014. The development of dimple-shaped
chitosan particles to treat serious colon infections. The nanoparticles chitosan carrier for ethambutol dihydrochloride dry powder inhaler. Drug Dev. Ind.
Pharm. 41, 791–800. http://dx.doi.org/10.3109/03639045.2014.903493.
showed prolonged in vitro drug release at low pH, similar to the gastric Al-Dhubiab, B.E., 2013. Formulation and in vitro evaluation of gelatin nanospheres for
environment. In addition, the particles showed a uniform bactericidal the oral delivery of selegiline. Curr. Nanosci. 9, 21–25. http://dx.doi.org/10.2174/
activity. 157341313805117866.
Amsalem, O., Nassar, T., Benhamron, S., Lazarovici, P., Benita, S., Yavin, E., 2017. Solid
Considering the modeling of release data and its kinetics for nano nano-in-nanoparticles for potential delivery of siRNA. J. Control. Release 257,
spray dried particles loaded with drugs, there are common mathema- 144–155. http://dx.doi.org/10.1016/j.jconrel.2016.05.043.
tical and experimental models which may lead to the accurate predic- Anandharamakrishnan, C., 2014. Drying Techniques for Nanoencapsulation, in:
Techniques for Nanoencapsulation of Food Ingredients. Springer, New York, pp.
tion in terms of delivery performance and design of nanoparticles. In
51–60. https://doi.org/10.1007/978-1-4614-9387-7.
fact, these models describe the concentration of the released drug as a Anandharamakrishnan, C., Ishwarya, P.S., 2015. Chapter 8: Spray drying for na-
function of time. The most applied models have been summarized in noencapsulation of food components. In: Spray Drying Techniques for Food
Ingredient Encapsulation. John Wiley & Sons Ltd, Chichester, UK, pp. 180–197.
Table 6 along with their relevant parameters and predicted release
Anzar, N., Mirza, M.A., Anwer, K., Khuroo, T., Alshetaili, A.S., Alshahrani, S.M., Meena,
mechanisms. J., Hasan, N., Talegaonkar, S., Panda, A.K., Iqbal, Z., 2018. Preparation, evaluation
and pharmacokinetic studies of spray dried PLGA polymeric submicron particles of
7. Conclusions and further remarks simvastatin for the effective treatment of breast cancer. J. Mol. Liq. 249, 609–616.
http://dx.doi.org/10.1016/j.molliq.2017.11.081.
Aquino, R.P., Stigliani, M., Del Gaudio, P., Mencherini, T., Sansone, F., Russo, P., 2014.
The number of publications in the field of nano spray drying for Nanospray drying as a novel technique for the manufacturing of inhalable NSAID
pharmaceuticals has increased rapidly since the market introduction of powders. Hindawi Publ. Corp. Sci. World J. 1–7.
Arpagaus, C., 2012. A novel laboratory-scale spray dryer to produce nanoparticles. Dry.
the Nano Spray Dryer B-90 in 2009 by Büchi Labortechnik AG Technol. 30, 1113–1121. http://dx.doi.org/10.1080/07373937.2012.686949.
(Switzerland). More than 160 papers have been identified and reviewed Arpagaus, C., 2011. Nano Spray Dryer B-90: Literature review and applications, best@
in this paper. Compared to conventional spray drying processes, nano buchi Information Bulletin, Number 63/2011.
Arpagaus, C., 2010. Spray Drying R&D Solutions – BÜCHI’s Nano Spray Dryer: A world
spray drying relies on a vibrating mesh technology to produce an

211
C. Arpagaus et al. International Journal of Pharmaceutics 546 (2018) 194–214

novelty in laboratory scale. ONdrugDelivery, March 2010, page 30. Available from: J. Pharm. Sci. 1–12. http://dx.doi.org/10.1002/jps.21886.
http://www.ondrugdelivery.com 40–41. Celli, G.B., Ghanem, A., Brooks, M.S.-L., 2015. Bioactive encapsulated powders for
Arpagaus, C., John, P., Collenberg, A., Rütti, D., 2017. Chapter 10: nanocapsules for- functional foods – a review of methods and current limitations. Food Bioprocess
mation by nano spray drying. In: Jafari, S.M. (Ed.), Nanoencapsulation Technologies Technol. 1825–1837. http://dx.doi.org/10.1007/s11947-015-1559-z.
for the Food and Nutraceutical Industries. Elsevier Inc., pp. 346–401. Cerchiara, T., Abruzzo, A., di Cagno, M., Bigucci, F., Bauer-Brandl, A., Parolin, C., Vitali,
Arpagaus, C., Meuri, M., 2010. Laboratory scale spray drying of inhalable particles: a B., Gallucci, M.C., Luppi, B., 2015. Chitosan based micro- and nanoparticles for
review. Respir. Drug Deliv. 469–476. colon-targeted delivery of vancomycin prepared by alternative processing methods.
Arpagaus, C., Rütti, D., Meuri, M., 2013. Chapter 18: enhanced solubility of poorly so- Eur. J. Pharm. Biopharm. 92, 112–119. http://dx.doi.org/10.1016/j.ejpb.2015.03.
luble drugs via spray drying. In: Douroumis, D., Fahr, A. (Eds.), Drug Delivery 004.
Strategies for Poorly Water-Soluble Drugs. John Wiley & Sons, Ltd., pp. 551–585. Chan, H.-K., Kwok, P.C.L., 2011. Production methods for nanodrug particles using the
Arpagaus, C., Schafroth, N., 2009. Laboratory scale spray drying of biodegradable poly- bottom-up approach. Adv. Drug Deliv. Rev. 63, 406–416. http://dx.doi.org/10.1016/
mers. Respir. Drug Deliv. Eur. 269–274. j.addr.2011.03.011.
Arpagaus, C., Schafroth, N., 2007. Spray Dried Biodegradable Polymers as target material Couvreur, P., Dubernet, C., Puisieux, F., 1995. Controlled drug delivery with nano-
for Controlled Drug Delivery, best@buchi Information Bulletin, Number 46/2007. particles – current possibilities and future trends.pdf. Eur. J. Pharm. Biopharm. 41,
Arpagaus, C., Schafroth, N., Meuri, M., 2010a. Laboratory Scale Spray Drying of Inhalable 2–13.
Drugs: A Review, best@buchi Information Bulletin, Number 59/2010. Dahili, L.A., Feczkó, T., 2015. Cross-linking of horseradish peroxidase enzyme to fine
Arpagaus, C., Schafroth, N., Meuri, M., 2010b. Laboratory Scale Spray Drying of Lactose: particles generated by nano spray dryer B-90. Period Polytech. Chem. Eng. 59,
A Review, best@buchi Information Bulletin, Number 57/2010. 209–214. http://dx.doi.org/10.3311/PPch. 7590.
Arpagaus, C., Schwartzbach, H., 2008. Scale-up from the Büchi Mini Spray Dryer B-290 to Dahili, L.A., Kelemen-Horváth, I., Feczkó, T., 2015. 2,4-Dichlorophenol removal by
the Niro MOBILE MINOR, best@buchi Information Bulletin, Number 52/2008. purified horseradish peroxidase enzyme and crude extract from horseradish im-
Assadpour, E., Jafari, S.M., 2017. Spray drying of folic acid within nano-emulsions: op- mobilized to nano spray dried ethyl cellulose particles. Process Biochem. 50,
timization by Taguchi approach. Dry Technol. 35, 1152–1160. http://dx.doi.org/10. 1835–1842. http://dx.doi.org/10.1016/j.procbio.2015.08.008.
1080/07373937.2016.1242016. Dahili, L.A., Nagy, E., Feczko, T., 2017. 2,4-dichlorophenol enzymatic removal and its
Assadpour, E., Jafari, S.M., Maghsoudlou, Y., 2017. Evaluation of folic acid release from kinetic study using horseradish peroxidase crosslinked to nano spray-dried poly
spray dried powder particles of pectin-whey protein nano-capsules. Int. J. Biol. (Lactic-Co-Glycolic Acid) fine particles. J. Microbiol. Biotechnol. 27, 768–774.
Macromol. 95, 238–247. http://dx.doi.org/10.1016/j.ijbiomac.2016.11.023. http://dx.doi.org/10.4014/jmb.1606.06002.
Baba, K., Nishida, K., 2013. Steroid nanocrystals prepared using the nano spray dryer B- De Cicco, F., Porta, A., Sansone, F., Aquino, R.P., Del Gaudio, P., 2014. Nanospray
90. Pharmaceutics 5, 107–114. http://dx.doi.org/10.3390/pharmaceutics5010107. technology for an in situ gelling nanoparticulate powder as a wound dressing. Int. J.
Baba, K., Nishida, K., 2012. Calpain inhibitor nanocrystals prepared using Nano Spray Pharm. 473, 30–37. http://dx.doi.org/10.1016/j.ijpharm.2014.06.049.
Dryer B-90. Nanoscale Res. Lett. 7, 1–9. Del Gaudio, P., Russo, P., Rodriguez Dorado, R., Sansone, F., Mencherini, T., Gasparri, F.,
Basran, E., 2017. Ocular application of dirithromycin incorporated polymeric nano- Aquino, R.P., 2017. Submicrometric hypromellose acetate succinate particles as
particles: an in vitro evaluation. Turkish J. Pharm. Sci. 14, 191–200. http://dx.doi. carrier for soy isoflavones extract with improved skin penetration performance.
org/10.4274/tjps.69855. Carbohydr. Polym. 165, 22–29. http://dx.doi.org/10.1016/j.carbpol.2017.02.025.
Beber, T.C., Andrade, D.F., Kann, B., Fontana, M.C., Coradini, K., Windbergs, M., Beck, Del Gaudio, P., Sansone, F., Mencherini, T., De Cicco, F., Russo, P., Aquino, R., Dumoulin,
R.C.R., 2014. Submicron polymeric particles prepared by vibrational spray-drying: E., Puglisi, G., Aquino, R.P., 2016. Nanospray drying as a novel tool to improve
semisolid formulation and skin penetration/permeation studies. Eur. J. Pharm. technological properties of soy isoflavone extracts. Planta Med. 83, 426–433. http://
Biopharm. 88, 602–613. http://dx.doi.org/10.1016/j.ejpb.2014.07.008. dx.doi.org/10.1055/s-0042-110179.
Beck-Broichsitter, M., Paulus, I.E., Greiner, A., Kissel, T., 2015a. Modified vibrating-mesh Denora, N., Lopedota, A., Perrone, M., Laquintana, V., Iacobazzi, R.M., Milella, A.,
nozzles for advanced spray-drying applications. Eur. J. Pharm. Biopharm. 92, Fanizza, E., Depalo, N., Cutrignelli, A., Lopalco, A., Franco, M., 2016. Spray-dried
96–101. http://dx.doi.org/10.1016/j.ejpb.2015.03.001. mucoadhesives for intravesical drug delivery using N-acetylcysteine- and glu-
Beck-Broichsitter, M., Schweiger, C., Schmehl, T., Gessler, T., Seeger, W., Kissel, T., 2012. tathione-glycol chitosan conjugates. Acta Biomater. 43, 170–184. http://dx.doi.org/
Characterization of novel spray-dried polymeric particles for controlled pulmonary 10.1016/j.actbio.2016.07.025.
drug delivery. J. Control. Release 158, 329–335. http://dx.doi.org/10.1016/j.jconrel. Dhand, R., 2002. Nebulizers that use a vibrating mesh or plate with multiple apertures to
2011.10.030. generate aerosol. Respir. Care 47, 406–416.
Beck-Broichsitter, M., Strehlow, B., Kissel, T., 2015b. Direct fractionation of spray-dried Dimer, F., de Souza Carvalho-Wodarz, C., Haupenthal, J., Hartmann, R., Lehr, C.-M.,
polymeric microparticles by inertial impaction. Powder Technol. 286, 311–317. 2015a. Inhalable clarithromycin microparticles for treatment of respiratory infec-
http://dx.doi.org/10.1016/j.powtec.2015.08.033. tions. Pharm. Res. 32, 3850–3861. http://dx.doi.org/10.1007/s11095-015-1745-8.
Bege, N., Renette, T., Endres, T., Beck-Broichsitter, M., Hänggi, D., Kissel, T., 2013. In situ Dimer, F., Ortiz, M., Pohlmann, A.R., Guterres, S.S., 2015b. Inhalable resveratrol mi-
forming nimodipine depot system based on microparticles for the treatment of croparticles produced by vibrational atomization spray drying for treating pulmonary
posthemorrhagic cerebral vasospasm. Eur. J. Pharm. Biopharm. 84, 99–105. http:// arterial hypertension. J. Drug Deliv. Sci. Technol. 29, 152–158. http://dx.doi.org/10.
dx.doi.org/10.1016/j.ejpb.2012.12.016. 1016/j.jddst.2015.07.008.
Behara, S.R.B., Farkas, D.R., Hindle, M., Longest, P.W., 2014a. Development of a high Draheim, C., de Crécy, F., Hansen, S., Collnot, E.-M., Lehr, C.-M., 2015. A design of ex-
efficiency dry powder inhaler: effects of capsule chamber design and inhaler surface periment study of nanoprecipitation and nano spray drying as processes to prepare
modifications. Pharm. Res. 31, 360–372. http://dx.doi.org/10.1007/s11095-013- PLGA nano- and microparticles with defined sizes and size distributions. Pharm. Res.
1165-6. 32, 2609–2624. http://dx.doi.org/10.1007/s11095-015-1647-9.
Behara, S.R.B., Longest, P.W., Farkas, D.R., Hindle, M., 2014b. Development and com- Durli, T.L., Dimer, F.A., Fontana, M.C., Pohlmann, A.R., Beck, R.C.R., Guterres, S.S., 2014.
parison of new high-efficiency dry powder inhalers for carrier-free formulations. J. Innovative approach to produce submicron drug particles by vibrational atomization
Pharm. Sci. 103, 465–477. http://dx.doi.org/10.1002/jps.23775. spray drying: influence of the type of solvent and surfactant. Drug Dev. Ind. Pharm.
Behara, S.R.B., Longest, P.W., Farkas, D.R., Hindle, M., 2014c. Development of high ef- 40, 1011–1020. http://dx.doi.org/10.3109/03639045.2013.798804.
ficiency ventilation bag actuated dry powder inhalers. Int. J. Pharm. 465, 52–62. Esfanjani, A.F., Jafari, S.M., Assadpoor, E., Mohammadi, A., 2015. Nano-encapsulation of
http://dx.doi.org/10.1016/j.ijpharm.2014.01.043. saffron extract through double-layered multiple emulsions of pectin and whey protein
Blasi, P., Schoubben, A., Giovagnoli, S., Rossi, C., Ricci, M., 2010. Alginate micro- and concentrate. J. Food Eng. 165, 149–155. http://dx.doi.org/10.1016/j.jfoodeng.2015.
nanoparticle production by spray drying. In: Proc. Meet. Lact. as a Carr. Inhal. Prod. 06.022.
Parma, Italy, Sept. 26-28, 2010. pp. 137–138. Ezhilarasi, P.N., Karthik, P., Chhanwal, N., Anandharamakrishnan, C., 2013.
Bonartsev, A., Zernov, A., Yakovlev, S., Zharkova, I., Myshkina, V., Mahina, T., Nanoencapsulation techniques for food bioactive components: a review. Food
Bonartseva, G., Andronova, N., Smirnova, G., Borisova, J., Kalishjan, M., Shaitan, K., Bioprocess Technol. 6, 628–647. http://dx.doi.org/10.1007/s11947-012-0944-0.
Treshalina, H., 2017. New Poly(3-hydroxybutyrate) microparticles with paclitaxel Faridi Esfanjani, A., Jafari, S.M., 2016. Biopolymer nano-particles and natural nano-
sustained release for intraperitoneal administration. Anticancer Agents Med. Chem. carriers for nano-encapsulation of phenolic compounds. Colloids Surf., B 146,
17, 434–441. http://dx.doi.org/10.2174/1871520615666160504095433. 532–543. http://dx.doi.org/10.1016/j.colsurfb.2016.06.053.
Bowen, M., Turok, R., Maa, Y.-F., 2013. Spray drying of monoclonal antibodies: in- Faridi Esfanjani, A., Jafari, S.M., Assadpour, E., 2017. Preparation of a multiple emulsion
vestigating powder-based biologic drug substance bulk storage. Dry Technol. 31, based on pectin-whey protein complex for encapsulation of saffron extract nano-
1441–1450. http://dx.doi.org/10.1080/07373937.2013.796968. droplets. Food Chem. 221, 1962–1969. http://dx.doi.org/10.1016/j.foodchem.2016.
Brandenberger, H., 2003. Development of a novel high-performacne cyclone to increase 11.149.
the yield in a Mini Spray Dryer, best@buchi Information Bulletin, Number 27/2003. Farkas, D.R., Hindle, M., Longest, P.W., 2015. Characterization of a new high-dose dry
Brinkmann-Trettenes, U., Barnert, S., Bauer-Brandl, A., 2014. Single step bottom-up powder inhaler (DPI) based on a fluidized bed design. Ann. Biomed. Eng. 43,
process to generate solid phospholipid nano-particles. Pharm. Dev. Technol. 19, 2804–2815. http://dx.doi.org/10.1007/s10439-015-1335-2.
326–332. http://dx.doi.org/10.3109/10837450.2013.778875. Fathi, M., Martin, A., McClements, D.J., 2014. Nanoencapsulation of food ingredients
Büchi Labortechnik AG, 2013. Nano Spray Dryer B-90 Technical data sheet, en 1311 A. using carbohydrate based delivery systems. Trends Food Sci. Technol. 39, 18–39.
Available from: http://static2.buchi.com/sites/default/files/technical-data-pdf/B- http://dx.doi.org/10.1016/j.tifs.2011.08.003.
90_Data_Sheet_en_A_0.pdf. Faulhammer, E., Wahl, V., Zellnitz, S., Khinast, J.G., Paudel, A., 2015. Carrier-based dry
Büchi Labortechnik AG, 2010. Nano Spray Dryer B-90 Brochure, Flawil, Switzerland. powder inhalation: Impact of carrier modification on capsule filling processability
11592236 en 1012. Available from: http://www.buchi.com. and in vitro aerodynamic performance. Int. J. Pharm. 491, 231–242. http://dx.doi.
Bürki, K., Jeon, I., Arpagaus, C., Betz, G., 2011. New insights into respirable protein org/10.1016/j.ijpharm.2015.06.044.
powder preparation using a nano spray dryer. Int. J. Pharm. 408, 248–256. http://dx. Faulhammer, E., Zellnitz, S., Wutscher, T., Stranzinger, S., Zimmer, A., Paudel, A., 2018.
doi.org/10.1016/j.ijpharm.2011.02.012. Performanc indicators for carrier based DPIs: carrier surface properties for capsule
Cal, K., Sollohub, K., 2009. Spray drying technique. I: hardware and process parameters. filling and API properties for in vitro aerosolisation. Int. J. Pharm. 536. http://dx.doi.

212
C. Arpagaus et al. International Journal of Pharmaceutics 546 (2018) 194–214

org/10.1016/j.ijpharm.2017.12.004. Kaewjan, K., Srichana, T., 2016. Nano spray-dried pyrazinamide-L-leucine dry powders,
Feng, A.L., Boraey, M.A., Gwin, M.A., Finlay, P.R., Kuehl, P.J., Vehring, R., 2011. physical properties and feasibility used as dry powder aerosols. Pharm. Dev. Technol.
Mechanistic models facilitate efficient development of leucine containing micro- 21, 68–75. http://dx.doi.org/10.3109/10837450.2014.971373.
particles for pulmonary drug delivery. Int. J. Pharm. 409, 156–163. http://dx.doi. Kaialy, W., Al Shafiee, M., 2016. Recent advances in the engineering of nanosized active
org/10.1016/j.ijpharm.2011.02.049. pharmaceutical ingredients: promises and challenges. Adv. Colloid Interface Sci. 228,
Fontana, M.C., Durli, T.L., Pohlmann, A.R., Guterres, S.S., Beck, R.C.R., 2014. Polymeric 71–91. http://dx.doi.org/10.1016/j.cis.2015.11.010.
controlled release inhalable powder produced by vibrational spray-drying: one-step Knoch, M., Keller, M., 2005. The customised electronic nebuliser: a new category of liquid
preparation and in vitro lung deposition. Powder Technol. 258, 49–59. http://dx.doi. aerosol drug delivery systems. Expert Opin. Drug Deliv. 2, 377–390. http://dx.doi.
org/10.1016/j.powtec.2014.03.011. org/10.1517/17425247.2.2.377.
Gautier, S., Arpagaus, C., Schafroth, N., Meuri, M., Deschamps, A., Maquet, V., 2010. Very Lass, J.S., Sant, A., Knoch, M., 2006. New advances in aerosolised drug delivery: vibrating
fine chitosan microparticles with narrow & controlled size distribution using spray- membrane nebuliser technology. Expert Opin. Drug Deliv. 3, 693–702. http://dx.doi.
drying technologies. Drug Deliv. Technol. 10, 30–37. org/10.1517/17425247.3.5.693.
Gharsallaoui, A., Roudaut, G., Chambin, O., Voilley, A., Saurel, R., 2007. Applications of Lee, S.H., Heng, D., Ng, W.K., Chan, H.-K., Tan, R.B.H., 2011. Nano spray drying: A novel
spray-drying in microencapsulation of food ingredients: an overview. Food Res. Int. method for preparing protein nanoparticles for protein therapy. Int. J. Pharm. 403,
40, 1107–1121. http://dx.doi.org/10.1016/j.foodres.2007.07.004. 192–200. http://dx.doi.org/10.1016/j.ijpharm.2010.10.012.
Ghasemi, S., Jafari, S.M., Assadpour, E., Khomeiri, M., 2017. Production of pectin-whey Lee, S.H., Teo, J., Heng, D., Ng, W.K., Chan, H.K., Tan, R.B.H., 2013. Synergistic com-
protein nano-complexes as carriers of orange peel oil. Carbohydr. Polym. 177, bination dry powders for inhaled antimicrobial therapy: Formulation, characteriza-
369–377. http://dx.doi.org/10.1016/j.carbpol.2017.09.009. tion and in vitro evaluation. Eur. J. Pharm. Biopharm. 83, 275–284.
Gu, B., Linehan, B., Tseng, Y.-C., 2015. Optimization of the Büchi B-90 spray drying Li, X., Anton, N., Arpagaus, C., Belleteix, F., Vandamme, T.F., 2010. Nanoparticles by
process using central composite design for preparation of solid dispersions. Int. J. spray drying using innovative new technology: the Büchi Nano Spray Dryer B-90. J.
Pharm. 491, 208–217. http://dx.doi.org/10.1016/j.ijpharm.2015.06.006. Control. Release 147, 304–310. http://dx.doi.org/10.1016/j.jconrel.2010.07.113.
Guo, J., Li, H., 2013. Nano spray drying of alkaline phosphatase with β-cyclodextrin and Li, X., Anton, N., Vandamme, T.F., 2015. Spray-drying of nano- and microcapsules of
l-leucine for inhalation. J. Control. Release 172, e107. http://dx.doi.org/10.1016/j. nutraceuticals. In: Shi, J. (Ed.), Functional Food Ingredients and Neutraceuticals –
jconrel.2013.08.261. Processing Technologies. CRC Press, Taylor & Francis Group, Boca Raton, pp.
Harsha, S., 2015. In vitro and in vivo evaluation of nanoparticles prepared by nano spray 455–480.
drying for stomach mucoadhesive drug delivery. Dry Technol. 33, 1199–1209. Littringer, E.M., Zellnitz, S., Hammernik, K., Adamer, V., Friedl, H., Urbanetz, N., a.,
http://dx.doi.org/10.1080/07373937.2014.995305. 2013. Spray drying of aqueous salbutamol sulfate solutions using the nano spray
Harsha, S., 2013. Pharmaceutical suspension containing both immediate/sustained-re- dryer B-90 – the impact of process parameters on particle size. Dry. Technol. 31,
lease amoxicillin-loaded gelatin nanoparticles: preparation and in vitro character- 1346–1353. http://dx.doi.org/10.1080/07373937.2013.793701.
ization. Drug Des. Dev. Ther. 7, 1027–1033. http://dx.doi.org/10.2147/DDDT. Longest, P.W., Golshahi, L., Behara, S.R.B., Tian, G., Farkas, D.R., Hindle, M., 2015.
S39956. Efficient nose-to-lung (N2L) aerosol delivery with a dry powder inhaler. J. Aerosol
Harsha, S., 2012. Dual drug delivery system for targeting H. pylori in the stomach: pre- Med. Pulm. Drug Deliv. 28, 189–201. http://dx.doi.org/10.1089/jamp.2014.1158.
paration and in vitro characterization of amoxicillin-loaded Carbopol® nanospheres. Maa, Y.F., Nguyen, P., a., Sit, K., Hsu, C.C.,, 1998. Spray-drying performance of a bench-
Int. J. Nanomed. 7, 4787–4796. http://dx.doi.org/10.2147/IJN.S34312. top spray dryer for protein aerosol powder preparation. Biotechnol. Bioeng. 60,
Harsha, S., Al-Dhubiab, B.E., Nair, A.B., Al-Khars, M., Al-Hassan, M., Rajan, R., 301–309. http://dx.doi.org/10.1002/(SICI)1097-0290(19981105)60:3<301::AID-
Attimarad, M., Venugopala, K.N., Asif, A.H., 2015a. Novel drying technology of BIT5>3.0.CO;2-L.
microsphere and its evaluation for targeted drug delivery for lungs. Dry Technol. 33, Maged, A., Mahmoud, A.A., Ghorab, M.M., 2017. Hydroxypropyl-beta-cyclodextrin as
502–512. http://dx.doi.org/10.1080/07373937.2014.963202. cryoprotectant in nanoparticles prepared by nano-spray drying technique. J. Pharm.
Harsha, S., Al-Dhubiab, B.E., Nair, A.B., Attimarad, M., Venugopala, K.N., Sa, K., 2017. Sci. Emerg. Drugs 5, 1–5. http://dx.doi.org/10.4172/2380-9477.1000121.
Pharmacokinetics and tissue distribution of microspheres prepared by spray drying Mahdavi, S.A., Jafari, S.M., Ghorbani, M., Assadpoor, E., 2014. Spray-drying micro-
technique: Targeted drug delivery. Biomed. Res. 28, 3387–3396. encapsulation of anthocyanins by natural biopolymers : a review. Dry. Technol. 32,
Harsha, S., Al-Khars, M., Al-Hassan, M., Kumar, N.P., Nair, A.B., Attimarad, M., Al- 509–518. http://dx.doi.org/10.1080/07373937.2013.839562.
Dhubiab, B.E., 2014. Pharmacokinetics and tissue distribution of spray-dried carbo- Maltesen, M.J., Bjerregaard, S., Hovgaard, L., Havelund, S., van de Weert, M., 2008.
platin microspheres: lung targeting via intravenous route. Arch. Pharm. Res. 37, Quality by design – spray drying of insulin intended for inhalation. Eur. J. Pharm.
352–360. http://dx.doi.org/10.1007/s12272-013-0151-1. Biopharm. 70, 828–838. http://dx.doi.org/10.1016/j.ejpb.2008.07.015.
Harsha, S., Aldhubiab, B.E., Alhaider, I.A., Attimarad, M., Nair, A., 2013a. Carbopol 934- Martena, V., Censi, R., Hoti, E., Malaj, L., Di Martino, P., 2012a. A new nanospray drying
P loaded with vildagliptin for diabetic delivery. In vitro and in vivo evaluation of method for the preparation of nicergoline pure nanoparticles. J. Nanopart. Res. 14,
nanoparticles. Curr. Nanosci. 9, 642–647. 1–10. http://dx.doi.org/10.1007/s11051-012-0934-1.
Harsha, S., Aldhubiab, B.E., Nair, A.B., Alhaider, I.A., Attimarad, M., Venugopala, K.N., Martena, V., Censi, R., Hoti, E., Malaj, L., Di Martino, P., 2012b. Indomethacin nano-
Srinivasan, S., Gangadhar, N., Asif, A.H., 2015b. Nanoparticle formulation by Büchi crystals prepared by different laboratory scale methods: effect on crystalline form and
B-90 nano spray dryer for oral mucoadhesion. Drug Des. Dev. Ther. 9, 273–282. dissolution behavior. J. Nanopart. Res. 14. http://dx.doi.org/10.1007/s11051-012-
http://dx.doi.org/10.2147/DDDT.S66654. 1275-9.
Harsha, S., Attimard, M., Khan, T.A., Nair, A.B., Aldhubiab, B.E., Sangi, S., Shariff, A., Masters, K., 1991. Spray Drying Handbook, fifth ed. Longman Scientific & Technical,
2013b. Design and formulation of mucoadhesive microspheres of sitagliptin. J. Harlow, USA.
Microencapsul. 30, 257–264. http://dx.doi.org/10.3109/02652048.2012.720722. Merchant, Z., Taylor, K.M.G., Stapleton, P., Razak, S.A., Kunda, N., Alfagih, I., Sheikh, K.,
Heng, D., Lee, S.H., Ng, W.K., Tan, R.B.H., 2011. The nano spray dryer B-90. Expert Opin. Saleem, I.Y., Somavarapu, S., 2014. Engineering hydrophobically modified chitosan
Drug Deliv. 8, 965–972. http://dx.doi.org/10.1517/17425247.2011.588206. for enhancing the dispersion of respirable microparticles of levofloxacin. Eur. J.
Hindle, M., Kaviratna, A., Yamarthi, D., Longest, P.W., 2015. Characterization of spray Pharm. Biopharm. 88, 816–829. http://dx.doi.org/10.1016/j.ejpb.2014.09.005.
dried azithromycin dry powders for inhalation. J. Aerosol Med. Pulmonary Drug Moncada, M., Astete, C., Sabliov, C., Olson, D., Boeneke, C., Aryana, K.J., 2015. Nano
Deliv. 4 A9-A9. spray-dried sodium chloride and its effects on the microbiological and sensory
Hu, Q., Gerhard, H., Upadhyaya, I., Venkitanarayanan, K., Luo, Y., 2016. Antimicrobial characteristics of surface-salted cheese crackers. J. Dairy Sci. 98, 5946–5954. http://
eugenol nanoemulsion prepared by gum arabic and lecithin and evaluation of drying dx.doi.org/10.3168/jds.2015-9658.
technologies. Int. J. Biol. Macromol. 87, 130–140. http://dx.doi.org/10.1016/j. Moshe, H., Davizon, Y., Menaker Raskin, M., Sosnik, A., 2017. Novel poly(vinyl alcohol)-
ijbiomac.2016.02.051. based amphiphilic nanogels by non-covalent boric acid crosslinking of polymeric
Jafari, S.M., Assadpoor, E., Bhandari, B., He, Y., 2008a. Nano-particle encapsulation of micelles. Biomater. Sci. 5, 2295–2309. http://dx.doi.org/10.1039/C7BM00675F.
fish oil by spray drying. Food Res. Int. 41, 172–183. http://dx.doi.org/10.1016/j. Murugesan, R., Orsat, V., 2012. Spray drying for the production of nutraceutical in-
foodres.2007.11.002. gredients – a review. Food Bioprocess Technol. 5, 3–14. http://dx.doi.org/10.1007/
Jafari, S.M., Assadpoor, E., He, Y., Bhandari, B., 2008b. Encapsulation efficiency of food s11947-011-0638-z.
flavours and oils during spray drying. Dry Technol. 26, 816–835. http://dx.doi.org/ Nandiyanto, A.B.D., Okuyama, K., 2011. Progress in developing spray-drying methods for
10.1080/07373930802135972. the production of controlled morphology particles: from the nanometer to sub-
Jafari, S.M., Esfanjani, A.F., 2017. Chapter 14: instrumental analysis and characterization micrometer size ranges. Adv. Powder Technol. 22, 1–19. http://dx.doi.org/10.1016/
of nanocapsules. In: Nanoencapsulation Technologies for the Food and Nutraceutical j.apt.2010.09.011.
Industries. Academic Press, pp. 524–544. Nazarov, A.K., Zavarzin, I.V., Nazarov, G.V., Aksenov, A.V., Levina, I.S., 2016. Drug
Jafari, S.M., Esfanjani, A.F., Katouzian, I., Assadpour, E., 2017a. Chapter 10: release, synthesis methods and manufacturing technology: preparation and bioavailability
characterization, and safety of nanoencapsulated food ingredients. In: evaluation of micronized steroidal mecigestone drug substance. Pharm. Chem. J. 49,
Nanoencapsulation of Food Bioactive Ingredients. Academic Press, pp. 401–453. 706–710. http://dx.doi.org/10.1007/s11094-016-1357-6.
Jafari, S.M., He, Y., Bhandari, B., 2007. Encapsulation of nanoparticles of d-limonene by Ngan, L.T.K., Wang, S.-L., Hiep, Đ.M., Luong, P.M., Vui, N.T., Đinh, T.M., Dzung, N.A.,
spray drying: role of emulsifiers and emulsifying techniques. Dry Technol. 25, 2014. Preparation of chitosan nanoparticles by spray drying, and their antibacterial
1069–1079. http://dx.doi.org/10.1080/07373930701396758. activity. Res. Chem. Intermed. 40, 2165–2175. http://dx.doi.org/10.1007/s11164-
Jafari, S.M., Katouzian, I., Rajabi, H., Ganje, M., 2017b. Chapter 13: bioavailability and 014-1594-9.
release of bioactive components from nanocapsules. In: Nanoencapsulation Nguyen, T.V., Nguyen, T.T.H., Wang, S.L., Vo, T.P.K., Nguyen, A.D., 2017. Preparation of
Technologies for the Food and Nutraceutical Industries. Academic Press, pp. chitosan nanoparticles by TPP ionic gelation combined with spray drying, and the
494–523. antibacterial activity of chitosan nanoparticles and a chitosan nanoparticle–amox-
Jafari, S.M., McClements, D.J., 2017. Nanotechnology approaches for increasing nutrient icillin complex. Res. Chem. Intermed. 43, 3527–3537. http://dx.doi.org/10.1007/
bioavailability. Adv. Food Nutr. Res. 81, 1–30. http://dx.doi.org/10.1016/BS.AFNR. s11164-016-2428-8.
2016.12.008. Okuyama, K., Abdullah, M., Lenggoro, I.W., Iskandar, F., 2006. Preparation of functional

213
C. Arpagaus et al. International Journal of Pharmaceutics 546 (2018) 194–214

nanostructured particles by spray drying. Adv. Powder Technol. 17, 587–611. http:// laser diffraction. Respir. Drug Deliv. VIII 2, 525–528.
dx.doi.org/10.1163/156855206778917733. Son, Y.-J., Longest, P.W., Hindle, M., 2013a. Aerosolization characteristics of dry powder
Okuyama, K., Lenggoro, I.W., 2003. Preparation of nanoparticles via spray route. Chem. inhaler formulations for the excipient enhanced growth (EEG) application: effect of
Eng. Sci. 58, 537–547. http://dx.doi.org/10.1016/S0009-2509(02)00578-X. spray drying process conditions on aerosol performance. Int. J. Pharm. 443, 137–145.
Öztürk, A.A., Yenilmez, E., Arslan, R., Şenel, B., Yazan, Y., 2017. Dexketoprofen http://dx.doi.org/10.1016/j.ijpharm.2013.01.003.Aerosolization.
Trometamol-Loaded Kollidon ® SR and Eudragit ® RS 100 polymeric nanoparticles: Son, Y.-J., Longest, P.W., Tian, G., Hindle, M., 2013b. Evaluation and modification of
formulation and in vitro-in vivo evaluation. Lat. Am. J. Pharm. Am. J. Pharm 36, commercial dry powder inhalers for the aerosolization of a submicrometer excipient
2153–2165. enhanced growth (EEG) formulation. Eur. J. Pharm. Sci. 49, 390–399. http://dx.doi.
Öztürk, A.A., Yenilmez, E., Yazan, Y., 2015. Preparation and Characterization of org/10.1016/j.ejps.2013.04.011.
Dexketoprofen Tromethamol Loaded Kollidon-SR Nanoparticles. In: ISOPS 11th Sosnik, A., Seremeta, K.P., 2015. Advantages and challenges of the spray-drying tech-
International Symposium on Pharmaceutical Sciences, June 9–12, 2015, Ankara, nology for the production of pure drug particles and drug-loaded polymeric carriers.
Turkey. Adv. Colloid Interface Sci. 223, 40–54. http://dx.doi.org/10.1016/j.cis.2015.05.003.
Panda, A., Meena, J., Katara, R., Majumdar, D.K., 2014. Formulation and characterization Sun, Y., Song, X., Wang, J., Yu, J., 2011. Preparation of lithium carbonate hollow spheres
of clozapine and risperidone co-entrapped spray-dried PLGA nanoparticles. Pharm. by spray pyrolysis. Cryst. Res. Technol. 46, 173–177. http://dx.doi.org/10.1002/
Dev. Technol. Early Online 1–11. http://dx.doi.org/10.3109/10837450.2014. crat.201000532.
965324. Suryaprakash, R.C., Lohmann, F.P., Wagner, M., Abel, B., Varga, A., 2014. Spray drying as
Perecin, C.J., Yoshioka, S.A., De Oliveira, A.M., Chitta, V.A., Cerize, N.N.P., 2014. a novel and scalable fabrication method for nanostructured CsH2PO4, Pt-thin-film
Nanopartículas Superparamagnéticas Encapsuladas com Polímeros Para Aplicação composite electrodes for solid acid fuel cells. RSC Adv. 4, 60429–60436. http://dx.
No Tratamento De Câncer Por Hipertermia. In: XXIV Congresso Brasiliero de doi.org/10.1039/C4RA10259B.
Engenharia Biomédica – CBEB 2014. pp. 2703–2706. Torge, A., Grützmacher, P., Mücklich, F., Schneider, M., 2017. The influence of mannitol
Perecin, C.J., Yoshioka, S., Chitta, V., Gratens, X., Léo, P., De Oliveira, A., Cerize, N., on morphology and disintegration of spray-dried nano-embedded microparticles. Eur.
2015. Magnetite Nanoparticles Encapsulated with PCL and Poloxamer by Nano Spray J. Pharm. Sci. 104, 171–179. http://dx.doi.org/10.1016/j.ejps.2017.04.003.
Drying technique. In: XIV Brazil MRS (Materials Research Society) Meeting, Rio, Vecellio, L., 2006. The mesh nebulizer: a recent technical innovation for aerosol delivery.
September 27 to October 01, 2015, Awarded Abstract: ASEE, Symposium B. Breathe 2, 252–260.
Pérez-Masiá, R., López-Nicolás, R., Periago, M.J., Ros, G., Lagaron, J.M., López-Rubio, A., Vehring, R., 2008. Pharmaceutical particle engineering via spray drying. Pharm. Res. 25,
2015. Encapsulation of folic acid in food hydrocolloids through nanospray drying and 999–1022. http://dx.doi.org/10.1007/s11095-007-9475-1.
electrospraying for nutraceutical applications. Food Chem. 168, 124–133. http://dx. Wang, B., Friess, W., 2017. Spray drying of silica microparticles for sustained release
doi.org/10.1016/j.foodchem.2014.07.051. application with a new sol-gel precursor. Int. J. Pharm. 532, 281–288. http://dx.doi.
Pinto, J.T., Radivojev, S., Zellnitz, S., Roblegg, E., Paudel, A., 2017. How does secondary org/10.1016/j.ijpharm.2017.09.016.
processing affect the physicochemical properties of inhalable salbutamol sulphate Wang, B., Zhang, W., Zhang, W., Mujumdar, A.S., Huang, L., 2005. Progress in drying
particles? A temporal investigation. Int. J. Pharm. 528, 416–428. http://dx.doi.org/ technology for nanomaterials. Dry. Technol. 23, 7–32.
10.1016/j.ijpharm.2017.06.027. Wang, T., Hu, Q., Zhou, M., Xia, Y., Nieh, M.P., Luo, Y., 2016a. Development of “all
Rajabi, H., Ghorbani, M., Jafari, S.M., Mahoonak, S.A., Rajabzadeh, G., 2015. Retention natural” layer-by-layer redispersible solid lipid nanoparticles by nano spray drying
of saffron bioactive components by spray drying encapsulation using maltodextrin, technology. Eur. J. Pharm. Biopharm. 107, 273–285. http://dx.doi.org/10.1016/j.
gum Arabic and gelatin as wall materials. Food Hydrocoll. 51, 327–337. http://dx. ejpb.2016.07.022.
doi.org/10.1016/j.foodhyd.2015.05.033. Wang, T., Hu, Q., Zhou, M., Xue, J., Luo, Y., 2016b. Preparation of ultra-fine powders
Rampino, A., Borgogna, M., Blasi, P., Bellich, B., Cesàro, A., 2013. Chitosan nanoparticles: from polysaccharide-coated solid lipid nanoparticles and nanostructured lipid car-
preparation, size evolution and stability. Int. J. Pharm. 455, 219–228. http://dx.doi. riers by innovative nano spray drying technology. Int. J. Pharm. 511, 219–222.
org/10.1016/j.ijpharm.2013.07.034. http://dx.doi.org/10.1016/j.ijpharm.2016.07.005.
Rascioni, R., Censi, R., Malaj, L., Di Martino, P., 2016. Effect of particle size reduction and Wang, T., Ma, X., Lei, Y., Luo, Y., 2016c. Solid lipid nanoparticles coated with cross-linked
crystalline form on dissolution behaviour of nimesulide. J. Therm. Anal. Calorim. polymeric double layer for oral delivery of curcumin. Colloids Surf., B 148, 1–11.
123, 2213–2223. http://dx.doi.org/10.1007/s10973-015-4874-8. http://dx.doi.org/10.1016/j.colsurfb.2016.08.047.
Sabliov, C.M., Astete, C.E., 2015. Chapter 17 – polymeric nanoparticles for food appli- Wang, T., Soyama, S., Luo, Y., 2016d. Development of a novel functional drink from all
cations. In: Sabliov, C.M., Chen, H., Yada, R. (Eds.), Nanotechnology and Functional natural ingredients using nanotechnology. LWT – Food Sci. Technol. 73, 458–466.
Foods: Effective Delivery of Bioactive Ingredients. John Wiley & Sons, Ltd., pp. http://dx.doi.org/10.1016/j.lwt.2016.06.050.
272–296. Wendel, S.C., Celik, M., 2005. Chapter 5: Spray Drying and Pharmaceutical Applications,
Sarma, S.J., Das, R.K., Brar, S.K., Le Bihan, Y., Buelna, G., Verma, M., Soccol, C.R., 2014. in: Handbook of Pharmaceutical Granulation Technology. pp. 129–157.
Application of magnesium sulfate and its nanoparticles for enhanced lipid production Wong, T.W., 2015. Nanospray drying technology: existing limitations and future chal-
by mixotrophic cultivation of algae using biodiesel waste. Energy 78, 16–22. http:// lenges. Recent Pat. Drug Deliv. Formul. 9, 185–186. http://dx.doi.org/10.2174/
dx.doi.org/10.1016/j.energy.2014.04.112. 1872211309666150513111150.
Schafroth, N., Arpagaus, C., Jadhav, U.Y., Makne, S., Douroumis, D., 2012. Nano and Wong, T.W., John, P., 2015. Advances in spray drying technology for nanoparticle for-
microparticle engineering of water insoluble drugs using a novel spray-drying pro- mation. In: Aliofkhazraei, M. (Ed.), Handbook of Nanoparticles. Springer
cess. Colloids Surf., B 90, 8–15. http://dx.doi.org/10.1016/j.colsurfb.2011.09.038. International Publishing, pp. 1–16.
Schmid, K., 2011. Spray drying of protein precipitates and evaluation of the nano spray Xue, J., Wang, T., Hu, Q., Zhou, M., Luo, Y., 2017a. Insight into natural biopolymer-
dryer B-90. PhD Thesis. Ludwig-Maximilians-University, Munich. emulsified solid lipid nanoparticles for encapsulation of curcumin: effect of loading
Schmid, K., Arpagaus, C., Friess, W., 2011. Evaluation of the nano spray dryer B-90 for methods. Food Hydrocoll. http://dx.doi.org/10.1016/j.foodhyd.2017.12.018.
pharmaceutical applications. Pharm. Dev. Technol. 16, 287–294. http://dx.doi.org/ Xue, J., Wang, T., Hu, Q., Zhou, M., Luo, Y., 2017b. A novel and organic solvent-free
10.3109/10837450.2010.485320. preparation of solid lipid nanoparticles using natural biopolymers as emulsifier and
Schmid, K., Arpagaus, C., Friess, W., 2009. Evaluation of a vibrating mesh spray dryer for stabilizer. Int. J. Pharm. 531, 59–66. http://dx.doi.org/10.1016/j.ijpharm.2017.08.
preparation of submicron particles. Respir. Drug Deliv. 323–326. 066.
Schön, M., Baumgartner, R., 2009. Method for heating and laminating, electrostatic se- Yang, X., Xu, Y., Cai, Y., Li, H., 2015. Novel nano-spray-dried powders for efficient pul-
parator, spray drier, separating device and method for separating particles. Patent EP monary drug delivery. Abstr./J. Control. Release 213, e117–e118. http://dx.doi.org/
2056037 (A1), priority date: 2007-10-30. 10.1016/j.jconrel.2015.05.197.
Schoubben, A., Blasi, P., Giontella, A., Giovagnoli, S., Ricci, M., 2015. Powder, capsule Zellnitz, S., Narygina, O., Resch, C., Schroettner, H., Urbanetz, N.A., 2015a.
and device: an imperative ménage à trois for respirable dry powders. Int. J. Pharm. Crystallization speed of salbutamol as a function of relative humidity and tempera-
494, 40–48. http://dx.doi.org/10.1016/j.ijpharm.2015.08.012. ture. Int. J. Pharm. 489, 170–176. http://dx.doi.org/10.1016/j.ijpharm.2015.04.
Schoubben, A., Blasi, P., Marenzoni, M.L., Barberini, L., Giovagnoli, S., Cirotto, C., Ricci, 079.
M., 2013a. Capreomycin supergenerics for pulmonary tuberculosis treatment: pre- Zellnitz, S., Schroettner, H., Urbanetz, N.A., 2015b. Influence of surface characteristics of
paration, in vitro, and in vivo characterization. Eur. J. Pharm. Biopharm. 83, modified glass beads as model carriers in dry powder inhalers (DPIs) on the aero-
388–395. http://dx.doi.org/10.1016/j.ejpb.2012.11.005. solization performance. Drug Dev. Ind. Pharm. 41, 1710–1717. http://dx.doi.org/10.
Schoubben, A., Giovagnoli, S., Blasi, P., Ricci, M., 2013b. Production of a capreomycin 3109/03639045.2014.997246.
sulfate inhalable powder by nano spray-drying. CRS Abstr. B. – Poster Index 27, Univ. Zellnitz, S., Schroettner, H., Urbanetz, N.A., 2014. Surface modified glass beads as model
Pavia 37. carriers in dry powder inhalers—influence of drug load on the fine particle fraction.
Schoubben, A., Giovagnoli, S., Tiralti, M.C., Blasi, P., Ricci, M., 2014. Capreomycin in- Powder Technol. 268, 377–386. http://dx.doi.org/10.1016/j.powtec.2014.08.059.
halable powders prepared with an innovative spray-drying technique. Int. J. Pharm. Zernov, A.L., Bonartsev, A.P., Yakovlev, S.G., Myshkina, V.L., Makhina, T.K., Parshina,
469, 132–139. http://dx.doi.org/10.1016/j.ijpharm.2014.04.042. E.S., Kharitonova, E.P., Bonartseva, G.A., Shaitan, K.V., 2017. Low molecular weight
Schultz, I., Vollmers, F., Lühmann, T., Rybak, J.-C., Wittmann, R., Stank, K., Steckel, H., poly(3-hydroxybutyrate) microparticles synthesized by piezoelectric spray drying for
Kardziev, B., Schmidt, M., Högger, P., Meinel, L., 2015. Pulmonary insulin-like the sustained release of paclitaxel. Nanotechnologies Russ. 12, 218–225. http://dx.
growth factor I delivery from trehalose and silk-fibroin microparticles. ACS Biomater. doi.org/10.1134/S1995078017020136.
Sci. Eng. 1, 119–129. http://dx.doi.org/10.1021/ab500101c. Zhou, M., Khen, K., Wang, T., Hu, Q., Xue, J., Luo, Y., 2018. Chemical crosslinking im-
Sithole, M.N., Choonara, Y.E., du Toit, L.C., Kumar, P., Marimuthu, T., Kondiah, P.P.D., proves the gastrointestinal stability and enhances nutrient delivery potentials of egg
Pillay, V., 2017. Development of a novel polymeric nanocomposite complex for drugs yolk LDL/polysaccharide nanogels. Food Chem. 239, 840–847. http://dx.doi.org/10.
with low bioavailability. AAPS PharmSciTech 6, 1–12. http://dx.doi.org/10.1208/ 1016/j.foodchem.2017.07.019.
s12249-017-0796-z. Zhou, M., Wang, T., Hu, Q., Luo, Y., 2016. Low density lipoprotein/pectin complex na-
Smart, J., Berg, E., Nerbrink, O., Zuban, R., Blakey, D., New, M., Mastersizer, M., 2002. nogels as potential oral delivery vehicles for curcumin. Food Hydrocoll. 57, 20–29.
TouchSpray technology: comparison of the droplet size with cascade impaction and http://dx.doi.org/10.1016/j.foodhyd.2016.01.010.

214

Das könnte Ihnen auch gefallen