Sie sind auf Seite 1von 12

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/228373124

Formulation of Nanosuspensions as a New


Approach for the Delivery of Poorly Soluble Drugs

Article in Current Nanoscience · November 2009


DOI: 10.2174/157341309789378177

CITATIONS READS

52 559

4 authors, including:

Xiaohui Pu Jin Sun


Henan University Shenyang Pharmaceutical University
19 PUBLICATIONS 307 CITATIONS 189 PUBLICATIONS 3,200 CITATIONS

SEE PROFILE SEE PROFILE

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

I.V. administration View project

Polymer micelles View project

All content following this page was uploaded by Xiaohui Pu on 15 July 2015.

The user has requested enhancement of the downloaded file.


Current Nanoscience, 2009, 5, 417-427 417

Formulation of Nanosuspensions as a New Approach for the Delivery of Poorly


Soluble Drugs

Xiaohui Pu, Jin Sun*, Mo Li and Zhonggui He*

Department of Biopharmaceutics, School of Pharmacy, No. 103 Wenhua Road, Shenyang 110016, China

Abstract: Nanosuspensions have attracted much interest of many scientists as a new approach to deliver poorly soluble drugs because of
their process flexibility and special advantages such as improved oral bioavailability, high drug loading, targeting capabilities and so on.
Recently, the manufacture processes of nanosuspensions have developed quickly, and some new methods have emerged, for example,
controlled precipitation, emulsion method, microemulsion method and melt-emulsion method. Additionally, the post-process of nano-
suspension has also gained great progresses to expand the utility of nanosuspensions, including solidification technique and surface
modification technique. Moreover, the study on targeting delivery by nanosuspension following intravenous administration has become a
hot topic of many researches. In the article, we will highlight new developments in the production of nanosuspensions and summarize the
application of nanosuspensions via a variety of administration routes. In the end, we will overview the research and development
prospectives of nanosuspensions.

Keywords: Nanosuspension, poorly soluble drugs, bioavailability, layer-by-layer self-assembly, targeting.

1. INTRODUCTION
As shown in statistics, 1/3 of the drugs adopted by United
States Pharmacopoeia were classified as poorly soluble drugs.
Among the new chemical entities (NCE) discovered by pharmaceu-
tical chemist, 40% of NCE were poorly soluble compounds [1,2].
Also, 40% of the active pharmaceutical ingredients (API) obtained
by high-throughout screen were also poorly soluble molecules.
Lower bioavailability resulting from poor solubility and incomplete
dissolution in vivo, often holds back continuous development and
coming into the market of some promising NCE, or elicits insuffi-
cient therapeutic effects of certain drugs. The increasing number of
poorly soluble drugs requires innovative formulation approaches to
acquire a sufficient bioavailability after oral administration or at
least to make available intravenously injectable forms.
Poorly soluble compounds can be usually classified into two
types of molecules: “grease ball” and “brick dust” compounds [3].
Grease ball molecules represent highly lipophilic compounds with a
high logP, whereas brick dust molecules usually are compounds Fig. (1). The relationship between drug classification and the logP/melting
with a high melt point (m.p. >200) and a low logP. For example, point (modified after Wassvik, et. al. [4]).
78% of compounds with a ClogP of 6 and low melting point are
grease ball, whereas 52% of compounds with melting point of
potential drug expulsion during storage and complex manufacturing
250 and a ClogP of 2 belong to brick dust [4] (Fig. 1). The for-
method [21]. By contrast, brick dust molecules are poorly soluble
mer compounds can not form bonds with water molecules, so these
not only in water but also in oils, the above formulation strategies
compounds have poor aqueous solubility and their solubility is
do not work effectively due to low encapsulation efficiency and low
limited by solvation process. By contrast, the latter compounds
loading, and so the brick dust molecules are readily withdrawn
have high lattice energy and their solubility is limited by solid-state,
during the drug development stage.
in which the strong intermolecular bonds within the crystal struc-
ture restrict the solubility of the compound in water and in oils. For these reasons, Müller et al. [22, 23] firstly developed nano-
suspensions, a sub-micro colloidal dispersion system, to overcome
Grease ball molecules have easily passed through the drug de-
the above limitations in 1995. As for this system, pure drug parti-
velopment process pipeline to reach the market by adopting appro-
cles ranging in size from 10-1000 nm [24], were stabilized by sur-
priate formulation strategies, including incorporation of the proper
factants and polymeric materials. Drugs encapsulated within nano-
excipients (such as disintegrants, cosolvents, wetting agents, solubi-
suspensions exist in pharmaceutically acceptable crystalline or
lizers and cyclodextrins [5-11]) and application of micelles and
amorphous state. Nanosuspensions can successfully formulate the
lipid-based systems (emulsion [12,13], microemulsion [14], solid
brick dust molecules for improved dissolution and good absorption.
dispersion [15-17], liposome [18], solid lipid nanoparticle [19,20]
Apart from this, nanosuspensions have some following advantages:
and so on). But, there are still some deficiencies for the above for-
firstly, drugs no longer need to be in the soluble form. It is effective
mulations, such as low drug loading, high toxicity, poor stability,
for those molecules insoluble in oils [25]; secondly, the high drug
loading can be achieved as a drug exists in the form of pure solids,
and can significantly reduce the administration volume of high dose
*Address correspondence to these authors at the No. 59 Mailbox, Department of Bio- [25]; thirdly, nanosuspensions can increase the physical and chemi-
pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No. 103 cal stability of drugs as they are actually in the solid state [26, 27];
Wenhua Road, Shenyang 110016, China; Tel/Fax: +86-24-23986321;
E-mail: hezhonggui@gmail.com; sunjin66@21cn.com
finally, nanosuspensions can provide the passive targeting

1573-4137/09 $55.00+.00 © 2009 Bentham Science Publishers Ltd.


418 Current Nanoscience, 2009, Vol. 5, No. 4 Pu et al.

Fig. (2). Two differing manufacture processes of nanosuspensions. ‘bottom-up’ process,  ‘top-down’ process. I. The process includes dissolving drugs
in solvents or heating drug to fuse. II. The process is adding drug’s solution to surfactants’ solution. III. The technology used to this process can be microe-
mulsion, fusion-emulsion and supercritical fluid method. IV. The process is energy-addition step, which includes heat and/or mechanical energy. V. The drugs
are blended with surfactants and water by high speed dispersing homogenizer.VI. The process can be high-pressure homogenization and milling.

capabilities when injected intravenously, and represent a very increases with the simultaneous decrease in static pressure below
promising targeting carrier system for anti-cancer agents due to the liquid boiling point at room temperature according to the Ber-
enhanced permeability and retention effect [21, 28]. So the nano- noulli’s equation. In consequence, the liquid starts boiling and
suspensions are considered as the most promising system for the forms gas bubbles at room temperature, and the bubbles will im-
delivery of poorly soluble drug. plode when the suspension leaves the gap and normal air pressure is
This review focuses on the different aspects of nanosuspensions, reached again (called cavitation). The implosion forces are suffi-
including manufacturing process, characterization techniques, ciently high to break down the drug microparticles into nanoparti-
post-production processes, in vitro and in vivo properties, and ap- cles. As for this method, homogenization pressure and the number
plication in drug delivery via various administration routes. In the of homogenization cycles were the most important parameters af-
case of surface modification, we introduced the novel coating fecting particles size and size distribution. Additionally, percentage
strategy of layer-by-layer self-assembly technique for controlled of solid content, types and combination of surfactants and the
release and targeting capability. hardness of drugs influence the resulting nanoparticles properties.

2.2. Milling Method


2. MANUFACTURING PROCESSES
This patent-protected technology was developed by Liversidge
For the nanosuspensions manufacture, there are two converse et al. and can be divided into three steps: firstly, drug and surfac-
methods (Fig. 2): ‘bottom-up’ and the ‘top-down’ technologies [29]. tants were mixed to form a homogeneous mixture and inputted into
The bottom-up technology is an assembling method from molecules the milling chamber; secondly, the milling pearls or the milling ball
to nano-sized particles, including microprecipitation, microemul- was put into, and certain amount of water was or not added into the
sion, melt emulsification method and so on. The top-down tech- milling chamber; thirdly, the miller was turned on for about several
nology is a disintegration approach from large particles, microparti- to dozens of hours to prepare the nanosuspensions. This method
cles to nanoparticles, such as high-pressure homogenization and was called as wet-milling method if the water and other liquid was
media milling method. The processes and principles of these meth- used, otherwise called dry-milling method. Alternatively, it can be
ods will be described in detail and Table 1 showed their advantages further divided into jet-milling, pearl-milling and ball-milling
and disadvantages. method according to the types of the milling media used.
2.1. High-Pressure Homogenization In this method, the high energy and shear forces generated by
the breach and the collision between the milling media and the drug,
The process can be summarized into three steps: firstly, drug are the input energy to break the micro-sized into nano-sized parti-
powders are dispersed in a stabilizer solution to form cles. Particle size here is determined by stress intensity and the
pre-suspension; then pre-suspension was homogenized by the number of contact points. The milling media usually include glass,
high-pressure homogenizer at a low pressure for several times as a zirconium oxide or highly cross-linked polystyrene resin. A general
kind of premilling, and finally was homogenized at a high pressure problem of pearl mills is potential erosion from the milling pearls
for 10-25 cycles until the nanosuspensions with the desired size leading to product contamination [33]. If glass and highly
were prepared. According to the liquids used to suspend drug pow- cross-linked polystyrene resin are used as the pearl material, ero-
ders, the method is classified into homogenization in water (Dis- sion can be minimized.
soCubes), homogenization in water-free media and water mixtures
(Nanopure). 2.3. Microprecipitation or Controlled Precipitation
During homogenization, drug particles are fractured by cavita- Precipitation has been applied for years to prepare submicron
tion, high-shear force and the collision of the particles against each particles within the last decade [34-36], especially for the poorly
other. In the homogenization gap, the dynamic pressure of the fluid soluble drugs. Typically, the drug is firstly dissolved in a solvent.
Formulation of Nanosuspensions as a New Approach Current Nanoscience, 2009, Vol. 5, No. 4 419

Table 1. The Advantages and Disadvantages of Different Nanosuspensions Manufacturing Processes

The Methods Advantages Disadvantages

widely applying regions, ease of scale-up and little batch-to-batch variation


pretreatment of micronized drug particles and
[30], narrow size distribution in the final product [31], allowing aseptic pro-
High-pressure homogenization presuspending materials before subjecting it to
duction of nanosuspensions for parenteral administration and flexibility in
homogenizeation
handling the drug quantity [32]

potential erosion of material from the milling


Milling the same as those for high-pressure homogenization
pearls

narrowly applying space, wide size distribution


Microprecipitation low need of energy, stable products and simple process
and potential toxicity of non-aqueous solvents

low need of energy, stable products, simple process, small size of particles and high concentration undesired surfactants and
Emulsion and microemulsion
uniform particle distribution residual solvents

much smaller, more uniform and more stable compared to that by the micro-
Microprecipitation-high pressure ho-
precipitation; less mechanical force and energy compared with the The manufacturing process is complicated.
mogenization
high-pressure homogenization

Few compliant objects, larger particles from it


Melt emulsification avoidance of organic solvents compared to the solvent diffusion
than solvent diffusion

Then this solution is mixed with a miscible antisolvent in the pres- The second method uses partially water-miscible solvents such
ence of surfactants. Rapid addition of a drug solution to the anti- as butyl lactate, benzyl alcohol, triacetin and ethyl acetate as the
solvent (usually water) leads to sudden supersaturation of drug in dispersed phase [41-43]. The emulsion or microemulsion is formed
the mixed solution, and generation of ultrafine crystalline or amor- by the conventional dispersion method and the drug nanosuspen-
phous drug solids. This process involves two phases: nuclei forma- sions are obtained by diluting the emulsion or microemulsion with
tion and crystal growth. When preparing a stable suspension with relatively large amount of water. The dilution causes complete dif-
the minimum particle size, a high nucleation rate but low growth fusion of the internal phase into the external phase, and leads to
rate is necessary. Both rates are dependent on temperature: the op- instantaneous formation of the nanosuspensions. The manner of
timum temperature for nucleation might lie below that for crystal diluting the emulsion included high-pressure homogenization or
growth, which permits temperature optimization [37]. magnetic stirring, the former much more efficient than the latter
High-supersaturation conditions are needed for rapid nucleation by [43].
adding drug in minimum volume of a water-miscible organic solu- The third method employs some organic solvents such as ethyl
tion into the water under rapid mixing at low temperature. This acetate (EA), toluene, or dichloromethane (DCM) to dissolve the
rapid dilution with water results in high- supersaturation conditions, drug as the internal phase. This solution was then dispersed into
and causes spontaneous nucleation and a subsequent reduction of solvent-saturated aqueous solution containing surfactants to form a
the supersaturation condition in the vicinity of the nucleating crys- crude emulsion and subjected to high-pressure homogenization to
tals, and reduction of crystal growth rates. In this process, the ratio form an ultrafine emulsion. The extraction of solvents and particle
of mixing time (mix) to precipitation time (precip) is a very impor- precipitation was carried out in an electrically heated stainless-steel
tant parameter influencing the resulting particle size. As mix is re- extraction column. SC CO2 was conversed from the bottom of the
duced relative to precip, the greater supersaturation (more rapid nu- extraction column at a certain flow rate, and the emulsion was de-
cleation) and longer time for precipitation (including condensation livered from the top countercurrently using a semi-preparative
and coagulation) may produce the minimum particle size until a HPLC pump at a constant flow rate. The ratio of the flow rates of
threshold value is reached. It is also pointed out that the stabilizers the SC CO2 to the emulsion was maintained constant. During this
should be added to aqueous phase, organic phase or both. process, the organic solvent was extracted quickly by SC CO2 and
the aqueous drug nanosuspensions were formed at the bottom of the
2.4. Emulsion and Microemulsion Method column simultaneously [44].
There are three methods to fabricate drug nanosuspensions by
the emulsification method: particles precipitation by evaporating 2.5. Microprecipitation-High Pressure Homogenization
low–medium boiling point solvents with negligible water solubility, (NANOEDGE)
by a quenching technique using partially water-miscible solvents, The NANOEDGE process (Baxter Healthcare Corporation)
such as benzyl alcohol and butyl lactate, or by a extracting tech- consisted of the precipitation of friable materials and subsequent
nique using supercritical CO2 (SC CO2) as extraction agent. Such fragmentation under high shear and/or thermal energy input, that is,
solvents are used as the dispersed phase of the emulsion to load the a combination of microprecipitation and high-pressure homogeni-
solute. zation [45, 46].
In the first method, an organic solvent or mixture of solvents The preparation process can be summarized into five steps [47].
loaded with drug is dispersed in the aqueous phase containing suit- Firstly, the drug was dissolved in a drug-soluble organic solvent to
able surfactants to form an emulsion or microemulsion. The organic form a solution. Secondly, the stabilizers were dissolved in the
phase is then evaporated under reduced pressure so that the drug second solvent in which drug was insoluble. Thirdly, the drug solu-
particles precipitate instantaneously to form a nanosuspensions tion was added to the second solvent under high-speed agitation.
stabilized by surfactants. Since a particle is formed in an emulsion Then, drug precipitated as microparticles during the diffusion from
droplet, it is flexible to control the nanoparticle size by controlling the miscible solvent to the second solvent. At the same time, the
the droplet size. Organic solvents usually included acetone, meth- surfactants in the solution were adsorbed into the surface of the
ylene chloride, chloroform [38, 39] and relatively safer ethyl acetate freshly formed microparticles rapidly to protect them from aggrega-
and ethyl formate etc [40]. tion or growth and then pre-suspension was prepared. Finally,
420 Current Nanoscience, 2009, Vol. 5, No. 4 Pu et al.

pre-suspension was homogenized under a suitable pressure for sev- toring of particle size by near-infrared spectrum [59] is also a good
eral times. This is a process of adding energy, and can further re- choice.
duce the size of particles and reinforce interactions between drug
nanoparticles and stabilizers. 3.2. Surface Charge (Zeta Potential)
Zeta potential gives certain information about the surface
2.6. Melt Emulsification Method charge properties and further the long-term physical stability of the
The melt emulsification method traditionally was used to pre- nanosuspensions. The value of particle surface charge indicates the
pare solid lipid nanoparticles, but Kocbek et al. [43] firstly used it strength of the interactive force between particle and particle at the
to prepare ibuprofen nanosuspensions. There are four steps in the nano-surfaces, which is the basis to nanosuspensions stability at the
production of nanosuspensions by melt emulsification method. macroscopic level. In order to obtain an electrostatically stabilized
Firstly, drug was added to aqueous solution containing stabilizer. nanosuspension, a minimum zeta potential of ± 30 mV is required
Secondly, the suspension was heated above the melting point of the [60, 61]. In the case of a combination of electrostatic and steric
drug and homogenized with high-speed homogenizer to form an stabilization, a minimum zeta potential of ±20 mV is desirable [62].
emulsion with melted liquid drug as the dispersed phase. Thirdly, it The zeta potentials values were commonly assessed by determining
was transferred to a high-pressure homogenizer for homogenization. the particle electrophoretic mobility using the Zetasizer (Malvern
During this process, the temperature was maintained above the Instruments Ltd., UK) and converting the electrophoretic mobility
melting point of the drug. Finally, the emulsion was then cooled at to the zeta potential via the Helmholtz-Smoluchowski equation [63].
a suitable temperature and the drug particles precipitated and even- In the field of materials science, an electroacoustic technique
tually formed the nanosuspensions. The particle size can be finely (AcoustoSizer, Matec Applied Sciences, USA) can also be used in
adjusted by the process parameters, such as drug concentration, the determination of the zeta potential [64].
type and concentration of stabilizers, cooling conditions, and ho-
mogenization procedure. 3.3. Crystalline State and Particle Morphology
The assessment of the crystalline state and particle morphology
3. CHARACTERIZATION TECHNIQUES together helps in understanding the polymorphic or morphological
The characteristics of nanoparticles exert an essential role in changes that a drug might undergo when subjected to nanosizing.
some in vitro and in vivo properties of nano-drug delivery system Additionally, during nanosuspensions preparation, drug crystalline
(Nano-DDS). For example, safety, efficacy and stability of state is likely to be partially converted to an amorphous form.
Nano-DDS are affected by the particle size, particle size distribu- Hence, it is essential to measure the extent of amorphous drug gen-
tion and zeta potential; solid state of nanoparticles influences the erated during the nanosuspensions production. The changes in the
dissolution performance by impact forces that shatter the lattice solid state of the drug particles as well as the extent of the amor-
along dislocation boundaries. So the nanoparticles characterization phous fraction can be determined by X-ray diffraction analysis [65,
is of a great importance to predict in vitro and in vivo Nano-DDS 66] and supplemented by differential scanning calorimetry [29, 67].
performance. Compared to the particle/microparticle, the in vivo In order to get an actual understanding of particle morphology, the
pharmacokinetic performance and biological function of nanoparti- techniques such as scanning electron microscopy (SEM) [29, 68,
cles strongly depends on its surface characteristics and solid state, 69], atomic force microscope [69, 70] or transmission electron mi-
including mean particle size and distribution, particle charge (zeta croscopy (TEM) [70, 71] are preferred.
potential), crystalline state and particle morphology.
4. IN VITRO AND IN VIVO PROPERTIES
3.1. Mean Particle Size and Particle Size Distribution 4.1. Saturation Solubility and Dissolution Rate
The mean particle size and the span of particle size distribution Dissolution rate is an important factor to assess the benefits
(polydispersity index, PI) are two important characteristic parame- from nanosuspensions in relation to the traditional meso- or mi-
ters because they affect the saturation solubility, dissolution rate, cro-formulations (e.g. coarse powder, micronized product), espe-
physical stability, even in-vivo behavior of nanosuspensions. The cially when designing the rapid-release dosage forms for poorly
Noyes–Whitney equation [48, 49], Ostwald–Freundlich equation soluble drugs. Paddle and basket methods (USP30) and
[50, 51], Kelvin equation [52-54] and the Prandtl equation [55] film-dialysis can be used to determine the dissolution rate. In addi-
could be used to explain the relationships among particle size, satu- tion to the adhesive properties [72, 73] of nanosuspensions, in-
ration solubility and dissolution rate of nano-preparations. Gao et al. creased dissolution rate and supersaturation solubility are the main
[56] had studied the effect of particle size on in-vivo pharmacoki- advantages of nanosuspensions compared to traditional dosage
netic behavior of oridonin (ORI) nanosuspensions following intra- forms. For example, the dissolution rate of ibuprofen nanosuspen-
venous administration in rats. ORI nanosuspensions with a mean sions was increased by more than three times compared to the mi-
particle size 103.315 nm was quickly removed from the circulat- cronized ibuprofen [43].
ing system. On the contrary, ORI nanosuspension with a mean par-
ticle size 897.214.2 nm exhibited a markedly delayed blood
4.2. In-Vivo Pharmacokinetic Performance
clearance and a high uptake in RES organs.
The establishment of relationship between in-vitro release and
At present, the mean particle size and PI could be measured by
in-vivo absorption and the monitoring of the in-vivo performance of
several techniques such as laser diffractometry (LD), photon corre-
the nanosuspensions are essential to a successful preparation, irre-
lation spectroscopy (PCS), microscope and coulter-counter. LD can
spective of the administration route and the delivery systems. For
detect and quantify the drug microparticles during the production
oral nanosuspensions, the drug dissolution rate can influence
process. LD yields a volume size distribution and can be used to
in-vivo biological performance of formulations to a larger extent.
measure particles ranging from 0.05 Gm up to 2000 m [57]. PCS
For instance, albendazole nanosuspensions enhanced the C max to
usually is used to determine the mean particle diameter and PI of
1.5-2 times than that of microsuspension, and increased the AUC0-
nanosuspensions. Measuring range of PCS is from 3 nm to 3 m
and the relative bioavailability (Fr) by 1-2 times than those of mi-
[58]. The coulter-counter gives the absolute number of particles per
crosuspension (Fig. 3) [74]. For intravenously injected nanosuspen-
volume unit for the different size classes, and it is a more efficient
sions, the organ distribution in part depends on the nanoparticle size
and appropriate technique than LD for quantifying the contamina-
and surface property. Surface hydrophilicity/hydrophobicity and
tion of nanosuspensions by microparticulate drugs. On-line moni-
Formulation of Nanosuspensions as a New Approach Current Nanoscience, 2009, Vol. 5, No. 4 421

Fig. (3). Mean plasma concentration of albendazole sulfoxide-time curves after oral administration of albendazole formulations to rats at a dose of 50 mg/kg (n
= 6). ABZ-Sup: microsuspension including polysorbate 80 (0.5%); ABZ-T: nanosuspension stabilized by polysorbate 80 (0.5%); ABZ-P: nanosuspension
stabilized by poloxamer 188 (0.5%); ABZ-HT: nanosuspension stabilized by HPMC K4MCR P (0.5%) & Polysorbate 80 (0.5%) (Reprinted from Mittapalli, et
al. [74]).

interactions with plasma proteins are considered as important fac- freezing for lyophilization) that may destabilize the particles. Due
tors affecting the in-vivo organ distribution behavior after i.v. injec- to the above considerations, adding matrix-formers to the suspen-
tion of nanosuspensions [75-78]. With the quick development of sion prior to solidification is necessary. At present, except for tradi-
physical chemistry and biochemistry, many techniques to evaluate tional lyoprotectants (such as glucose, sucrose, trehalose, lactose,
the surface properties and protein interactions have emerged re- mannitol, sorbitol, maltose and dextran) [82-86]some new ma-
cently. For example, hydrophobic interaction chromatography, trix-formers have received significant interest. Van Eerdenbrugh et
previously employed to determine the surface hydrophobicity of al. had successfully used microcrystalline cellulose to displace su-
bacteria [79], can be used to determine surface hydrophobicity [80]. crose as a matrix former during freeze-drying of itraconazole
2-D PAGE [75] can be employed for the quantitative measurement nanosuspensions [87], and had again evaluated four alternative
of protein adsorption to nanoparticle surface after i.v. injection of matrix formers [Avicel®PH101, Fujicalin® (CaHPO4), Aerosil®200
drug nanosuspensions to animals. (SiO2) and Inutec®SP1] for their capability in preserving rapid
Although establishment of an in-vitro/in-vivo relationship is dissolution after spray-drying of nanosuspensions [88]. In addition,
extremely important to nanosuspensions, the biorelevant studies the effect of surface hydrophobicity on drug dissolution behavior
have not been reported by far. Especially, the correlation of phar- upon redispersion had been investigated, indicating the more in-
macokinetics and pharmacodynamics will be a further research tense hydrophobicity, the more aggregation of the nanoparticles and
topic. the slower the drug’s dissolution after solidification [89].

5.2. Surface Modification Techniques


5. POST-PRODUCTION PROCESSING
Nanosuspensions have the particular characteristics to increase
5.1. Solidification Techniques the saturation solubility and dissolution rate for the poorly soluble
Nanoparticles are usually produced in the liquid media, that is, drugs. But in some cases, the rapid or burst release of nanosuspen-
nanosuspensions. The nanosuspensions usually have the stability sions may result in the side effect and toxicity. As a colloid
issues involved in the physical (e.g. Ostwald ripening and agglom- nanoparticle system, nanosuspensions usually can target the mono-
eration) and chemical (e.g. hydrolysis) processes. In this case, solid cyte phagocytic system (MPS), which can aid in the treatment of
dosage forms are considered more attractive, due to their patient lymphatic-mediated diseases [90], like Mycobacterium tuberculosis,
convenience (marketing aspects) and good stability. Therefore, Listeria monogyna, Leishmania sp. The action is called as ‘passive
transformation of nanosuspensions into the solid dosage form is targeting’. However, the passive targeting process could pose an
desirable. Solidification methods of the nanosuspensions include obstacle when either macrophages are not the desired targets or
some unit-operations such as pelletization, granulation, spray dry- accumulated drug is toxic to MPS cells. Hence, in order to bypass
ing or lyophilization [81]. As the primary objective of the nanopar- the phagocytic uptake of the drug, its surface properties need to be
ticulate system is rapid dissolution, disintegration of the solid form tuned, just like stealth liposomes [91, 92] and nanoparticles [93,
and redispersion of the individual nanoparticles should be rather 94].
rapid, so that it does not impose a barrier on the integrated dissolu- Faced with the above problems, the surface modification of
tion process. Drying of nanoparticles can create stress on the parti- nanosuspensions will be very necessary. In the case of burst release
cles that can cause aggregation. For example, drying may lead to and passive targeting, the controlled release and long residence at
crystallization of the polymers such as poloxamers, thereby com- site of action may be effective. For example, Tan et al. had pre-
promising their ability to prevent aggregation. Drying can also cre- pared layer-by-layer self-assembly coated procaine hydrochloride
ate additional thermal stresses (due to heat for spray drying or
422 Current Nanoscience, 2009, Vol. 5, No. 4 Pu et al.

Table 2. The New Drug Application Based on Nanosuspensions Technique Reported and Marketed by Now

Drugs Indication Author or Company Route Status

Paclitaxel Anticancer American Bioscience Intravenous Marketed

Danazol Hormone Rogers T.L. Oral Reported [34]

Naproxen Anti-inflammatory Anchalee Ain-Ai Oral/parenteral Reported [104]

Probucol Lipid lowering Jyutaro Shudo Oral Reported [105]

Rapamune Immunosuppressant Elan Nanosystems Oral Marketed

Emend Anti-emetic Elan Nanosystems Oral Marketed

Cytokine inhibitor Crohn’s disease Elan Nanosystems Oral Phase II

Fenofibrate Lipid lowering SkyePharma Oral Marketed

Megestrol acetate Steroid hormone Par Pharmaceuticals Oral Marketed

paliperidone palmitate Anti-schizophrenia Johnson and Johnson Oral Phase III

Loviride Antivirotic B. Van Eerdenbrugh Intravenous Reported [28]

Busulfan Anticancer SkyePharma Intrathecal Undisclosed

Budesonide Asthma Jerry Z. Yang Pulmonary Reported [63]

Fluticasone Asthma Jerry Z. Yang Pulmonary Reported [63]

Insulin Diabetes BioSante Oral Undisclosed

Clofazimine Antimycobacterials K. Peters Intravenous Reported [106]

Buparvaquone Antibiotic Müller R. H. Oral Reported [91]

Oridonin Anticancer Lei Gao Intravenous Reported [55]

AZ68 Anticancer Kalle S. Oral/Intravenous Reported [107]

Ascorbyl palmitate Antioxidant Veerawat T. Intravenous Reported [68]

Hydrocortisone Glucocorticoid M.A. Kassem Ophthalmic Reported [108]

Prednisolone Glucocorticoid M.A. Kassem Ophthalmic Reported [108]

Hexadecadrol Glucocorticoid M.A. Kassem Ophthalmic Reported [108]

Aphidicolin Antileishmanial O. Kayser Oral Reported [109]

Dihydroartemisinin Antimalarial Jiraporn C. Intravenous Reported [69]

Cilostazol Antiplatelet agent Jun-ichi Jinno Oral Reported [110]

Spironolactone Diuretics P. Langguth Intravenous Reported [111]

Carbamazepine Psychotolytic D. Douroumis Oral Reported [112]

Omeprazole Proton pump inhibitor Jan Möschwitzer Intravenous Reported [26]

Thymectacin Anticancer Elan Nanosystems Intravenous Undisclosed

Silver Eczema NUCRYST Topical Phase 

Mitotane Adrenal Cortex Hormones Michele Trotta Oral Reported [40]

Griseofulvin Antifungal Boris Y. Shekunov Oral Reported [41]

Tarazepide Selective CCKa-antagonist C. Jacobs Oral Reported [113]

Albendazole Anthelmintic drug Mittapalli P. K. Oral Reported [74]

Azithromycin Antimicrobial Dianrui Zhang Oral Reported [114]

Ketoprofen Analgesic Remon J.P. Oral Reported [115]

nanogels, which can effectively reduce and minimize the high burst the future direction of targeted drug delivery systems. For instance,
release of drug compared to uncoated nanogels [95]. Mucoadhesive modification of the surface of nanoparticles with phosphol-
bupravaquone nanosuspensions can increase the adhesion to the gut ipid-polyethylenegylcol (PEG) chains [98] could delay protein ad-
wall and prolong residence at gastrointestinal tract, and the results sorption binding and opsonization of the nanoparticles, thereby
of pharmacodynamic experiments revealed a 10-fold reduction in reducing macrophage uptake. This would increase circulation time,
the infective score of cryptosporidium parvum as compared to the and give the particles the opportunity to find, and leak out of dis-
bupravaquone nanosuspensions without mucoadhesive polymers continuities of vasculature in neoplasms, infections and inflamma-
after oral administration [96, 97]. In the case of the passive target, tions [99, 100]. Zahr et al. [101] modified surface of dexametha-
the engineering of stealth nanosuspensions (analogous to stealth sone nanoparticles via layer by layer assembly with PEGlyted
liposomes) by using various surface coatings to the desired site is polyelectrolytes to get long-circulating preparations. Turning to the
Formulation of Nanosuspensions as a New Approach Current Nanoscience, 2009, Vol. 5, No. 4 423

nanosuspensions, the dissolution rate was significantly enhanced in


the nanometer-sized system, more than 65% dissolved in 5 h, as
opposed to only 20% of the micronized drugs [114].
Apart from improved oral absorption, nanosuspensions offer the
following advantages: dose proportionality, low inter-subject vari-
ability. Drug nanosuspensions can be easily incorporated into vari-
ous dosage forms such as tablets, capsules and fast melts by means
of standard manufacturing techniques. Ketoprofen nanosuspensions
have been successfully incorporated into pellets for the sustained
release the drug over a period of 24 h [115].

6.2. Parenteral Drug Delivery


Injections provide fast onset of action, accurate dose, reliable
efficacy and avoidance of first-pass metabolism. Previously, injec-
tions of poorly water-soluble drugs were usually approached by
formulating drugs with excessive amounts of co-solvents, solubi-
lizers, surfactants and carrier materials, which may induce serious
toxicity. For instance, marketed paclitaxel injection containing
Cremophor EL often result in anaphylactoid reactions. To establish
a more acceptable formulation, an injectable nanosuspensions for-
mulation of poorly water-soluble drugs has emerged. Successful
formulation has been reported as applied to antineoplastic agents
[117], anaesthetic agents [118], antifungals and antibacterials [119],
as well as for agents for malignant hyperthermia [120] and cancer
pain [121].
Nanosuspensions can be administered via different parenteral
routes, such as intraarticular, intraperitoneal and intravenous injec-
tions. In some cases, their nanoparticulate nature dictates MPS tar-
geting. If intravenously administered nanoparticles do not dissolve
immediately, they will initially distribute to MPS organs [109], in
particular the Kupffer cells in the spleen and liver [122] (Fig. 4).
Fig. (4). Concentration of radioactivity in tissues post-dosing with IV Subsequent drug dissolution in MPS provides a depot effect of
itraconazole nanosuspensions as a single dose of 10 mg/kg (Reprinted from these organs. The finding of initial sequestration by the MPS, fol-
Rabinow, et al. [122]). lowed by slow release is generally found for intravenously admin-
istered itraconazole nanoparticulate dosage forms [123].
special properties of the functional excipient coating on the surface
of nanoparticles, it can be realized to actively target the desired 6.3. Ophthalmic Drug Delivery
disease sites. For example, Kreuter had successfully targeted the Most of ocular diseases are treated with topical application of
peptide dalargin to the brain by employing surface modified poly- drug solutions administered as eye drops. The present therapy with
isobutyl cyanoacrylate nanoparticles [102]. Albumin-bound pacli- conventional eye drops (solution or microsuspensions) requires
taxel nanosuspension (nab-paclitaxel) successfully accumulated frequent instillation due to the rapid and extensive pre-corneal loss
tumor tissue by binding of albumin to a cell surface, 60-kDa glyco- caused by drainage through the naso-lacrimal duct and the
protein (gp60) receptor (albondin) as well as via caveolar transport non-productive absorption after corneal permeation and blinking.
and binding of albumin to SPARC (secreted protein acid and rich in Frequent instillation leads to poor patient compliance, and admini-
cysteine) [103]. stration of a large dose induces glaucoma, cataract formation and
damage of optic nerve.
6. APPLICATIONS Ophthalmic drug delivery, more than any other route of ad-
Nanosuspensions have been used to prepare diverse dosage ministration, may benefit to a large extent from the characteristics
forms by post-production processing. The smaller particle size and of nano-sized drug particles. The nanometer size represents a state
larger surface area lead to increased dissolution rate and oral ab- of matter characterized by higher solubility, higher surface area
sorption. Additionally, the nanoparticulate nature might dictate available for high dissolution, better bioadhesion and corneal pene-
naturally targeting of the monocyte phagocytic system (MPS), and tration, and less frequent instillation. Therefore nanosuspensions
result in unusual pharmacokinetic consequences. At present, there can increase the local availability and biological activity of the drug
are many drugs in the form of nanosuspensions to be reported and compared with that of drugs administered in classical aqueous eye-
marketed (Table 2) via the various administration routes, including drops. Nanosuspensions manufactured by Kassem et al. [107] as an
oral, parenteral, ophthalmic and pulmonary routes. ophthalmic delivery system for certain glucocorticoid drugs can
6.1. Oral Drug Delivery illustrate this findings (Table 3).

In general, oral administration is first choice for various drugs Besides the natural bioadhesion of the nanoparticulate, surface
due to good patient compliance, readily transportation, simple charge and modified surface by suitable gel matrix and bioadhesive
manufacture process. The major problem associated with oral ad- matrix allow prolonged residence in the cul-de-sac and sustained
ministration is low bioavailability and finally its inadequate effi- release of the drug. If nanoparticles have a positive charge, signifi-
cacy due to poor solubility and incomplete dissolution. Oral nano- cant improvement in drug performance can be achieved. The posi-
suspensions have been specifically used to increase the absorption tive charge present in these nanoparticles could account for a strong
rate and bioavailability of drugs [116] due to much larger surface to interaction with the negatively charged mucosa of the conjunctiva,
volume ratio. For example, when formulating azithromycin as thus allowing for a sufficient residence time to permit an efficient
424 Current Nanoscience, 2009, Vol. 5, No. 4 Pu et al.

Table 3. Mean Values of Pharmacodynamic Parameters for Three Drugs Solution and Corresponding Nanosuspensions (Mean ± S.E.)

Preparations %IOPmax Tmax (h) AUC0–8 h HVD(h) HVDR (h) MRT (h)

(% increase in IOP. h)
Hydrocortisone 9.77 ± 0.37*** 1.35±0.11† 23.27 ± 1.08*** 2.36±0.28* 2.36±0.28*** 2.40±0.08***

solution

Hydrocortisone 17.22 ± 1.32 1.60±0.10 56.89 ± 4.60 3.13± 0.29 4.69 ± 0.26 3.47 ± 0.15

nanosuspension

Prednisolone 11.49±0.83*** 1.80± 0.08† 34.60 ± 2.64*** 3.11±0.31* 3.11±0.31*** 2.88±0.08***

solution

Prednisolone 25.70 ± 1.24 1.85 ± 0.07 103.18 ± 7.21 3.87 ± 0.28 5.82 ± 0.27 3.71 ± 0.07

nanosuspension

Dexamethasone 13.87±1.05*** 1.55± 0.09† 66.02 ± 5.85*** 4.82±0.33* 4.82±0.33*** 4.33 ± 0.11*

solution

Dexamethasone 24.97 ± 1.27 1.75 ± 0.08 148.05 ± 7.69 6.10 ± 0.29 8.39 ± 0.17 4.95 ± 0.30

nanosuspension

Statistical differences between the nanosuspensions of each drug and corresponding solution. % IOPmax: the maximum percentage increase in intraocular pressure (IOP); Tmax: the
time of maximum response; AUC0–8: the area under percentage increase in IOP versus time curve; HVD: half peak IOP response intensity prevails; HVDR: half value duration
relative to reference; MRT: the mean residence time. * p = 0.05 significant; ** p = 0.01 highly significant; *** p = 0.001 very highly significant; † Insignificant.

drug release [124,125]. Wrapping nanosuspensions in appropriate various poorly soluble drugs, in particular for the brick-dust mole-
gel matrix or bioadhesive matrix is another technique to prolong cules poorly soluble in both water and oils, and can reduce the so-
retention time. After surface modification by biodegradable hydro- cial investment and enhance the success rate in insoluble drug de-
philic polymer, flurbiprofen nanosuspensions demonstrated good velopment.
effect of sustained-release following ocular administration [125]. Altered pharmacokinetic profiles of drugs caused by nanosus-
pensions have become appreciable insofar as they improve safety
6.4. Pulmonary Drug Delivery and efficacy. So the study on in-vivo biological performance is
Pulmonary delivery is the preferred route of drug administration extremely important, and the establishment of an in-vitro/in-vivo
in the treatment of many respiratory diseases, such as asthma and relationship will become a hot research field in the further study of
chronic obstructive pulmonary disease [126,127]. nanosuspensions.
Over years, solution, microsuspension, and dry powder as con- The combination of nanosuspension solidification technique
ventional formulations were used in the preclinical studies with traditional dosage forms, e.g. incorporating drug nanoparticles
[127,128]. The utility of these formulations, however, has some into pellets, tablets or gels, will readily widen application of nano-
limitations in the preparation and delivery. For example, vehicle suspensions. Additionally their applications in buccal, nasal and
tolerability of solution formulations [129], limited diffusion and topical delivery will represent a more beautiful prospective.
dissolution of the drug, rapid clearance of the drug from the lungs Surface modification of the drug nanosuspensions can further
[130], less residence time, highly variable lung deposition and ex- increase the benefits, e.g. stabilizing blood level of drugs by con-
posure of microsuspensions [60,131-134] are formidable problems trolling drug release and targeting specific organ by using special
associated with pulmonary drug delivery. surface ligands in the production process or layer-by-layer
self-assembly technology. Controlled drug release and functional-
Nanosuspensions with a tailored particle size may overcome
ized surface coatings capable of eliciting passive or active targeting,
many challenges in the pulmonary drug delivery. Compared to the
will be regarded as the future promising step in the nanosuspen-
conventional formulations, nanosuspensions have several advan-
sions research.
tages: firstly, easy to dose with a syringe-type delivery device in
animal studies, and more consistent drug distribution in the lung
than dry powder formulations; secondly, the increased adhesiveness ACKNOWLEDGEMENT
of the drug in nanosuspensions to mucosal surfaces [135] offers a We are grateful for financial support from the National Basic
prolonged residence time for the drug at the absorption site. For Research Program of China (973 Program), No. 2009CB930300.
example, the plasma exposures for fluticasone and budesonide
nanosuspensions after the intratracheal administration were similar REFERENCES
to solutions and showed rapid absorption with first-order clearance
[1] Amidon, G.L.; Löbenberg, R. Modern bioavailability, bioequivalence and
kinetic as a result of the deep lung deposition and fast onset [105]. biopharmaceutics classification system. New scientific approaches to inter-
Additionally, fluticasone concentrations in the lung decreased national regulatory standards. Eur. J. Pharm. Biopharm., 2000, 50(1), 3-12.
gradually over 4 h after intratracheal administration [105]. [2] Martinez, M.; Augsburger, L.; Johnston, T.; Jones W.W. Applying the bio-
pharmaceutics classification system to veterinary pharmaceutical products.
7. CONCLUSIONS AND PROSPECTIVES Part I. Biopharmaceutics and formulation considerations. Adv. Drug Deliv.
Rev., 2002, 54, 805-824.
Nanosuspensions are considered as the most promising delivery [3] Bergström, C.A.; Wassvik, C.M.; Johansson, K.; Hubatsch, I. Poorly soluble
system for poorly soluble drugs, due to high bioavailability and less marketed drugs display solvation limited solubility. J. Med. Chem., 2007,
inter- and intra-subjects variances. It is proved suitable to formulate 50(23), 5858-5862.
Formulation of Nanosuspensions as a New Approach Current Nanoscience, 2009, Vol. 5, No. 4 425

[4] Wassvik, C.M.; Holmén, A.G.; Draheim, R.; Artursson, P.; Bergström, C.A. [29] Grau, M.J.; Kayser, O.; Müller, R.H. Nanosuspensions of poorly soluble
Molecular characteristics for solid-state limited solubility. J. Med. Chem., drugs - reproducibility of small-scale production. Int. J. Pharm., 2000, 196,
2008, 51(10), 3035-3039. 155-157.
[5] Arias, M.J.; Moyano, J.R.; Gines, J.M. Investigation of the triamterene-- [30] Müller, R.H.; Böhm, B.H.L. Nanosuspensions. Müller, R.H., Benita, S.,
cyclodextrin system prepared by co-grinding. Int. J. Pharm., 1997, 153, Böhm, B.H.L. Eds.: In: Emulsions and Nanosuspensions for the Formulation
181-189. of Poorly Soluble Drugs. Medpharm Scientific Publishers: Stuttgart, 1998, pp.
[6] Badawy, S.I.F.; Ghorab, M.M.; Adeyeye, C.M. Characterization and 149-174.
bioavailability of danazol-hydroxypropyl--cyclodextrin coprecipitates. Int. J. [31] Krause, K.; Müller, R.H. Production and characterization of highly concen-
Pharm., 1996a, 128, 45-54. trated nanosuspensions by high pressure homogenization. Int. J. Pharm.,
[7] Badawy, S.I.F.; Marshall, A.L.; Ghorab, M.M.; Adeyeye, C.M. A study of 2001, 214, 21-24.
the complexation between danazol and hydrophilic cyclodextrin derivatives. [32] Rogers, T.L.; Gillespie, I.B.; Hitt, J.E.; Fransen, K.L.; Crowl, C.A.; Tucker,
Drug Dev. Ind. Pharm., 1996b, 22, 959-966. C.J.; Kupperblatt, G.B.; Becker, J.N.; Wilson, D.L.; Todd, C.; Broomall,
[8] Esclusa-Diaz, M.T.; Guimaraens-Mendez, M.; Perez-Marcos, M.B.; C.F.; Evans, J.C.; Elder E.J. Development and characterization of a scalable
Vila-Jato, J.L.; Torres-Labandeira, J. Characterization and in vitro dissolu- controlled precipitation process to enhance the dissolution of poorly wa-
tion behaviour of ketoconazole/- and 2-hydroxypropyl--cyclodextrin in- ter-soluble drugs. Pharm. Res., 2004, 22(11), 2048-2057.
clusion compounds. Int. J. Pharm., 1996, 143, 203-210. [33] Matteucci, M.E.; Hotze, M.A.; Johnston, K.P.; Williams, R.O. 3rd. Drug
[9] Jarvinen, T.; Jarvinen, K.; Schwarting, N.; Stella, V.J. -cyclodextrin deriva- nanoparticles by antisolvent precipitation: mixing energy versus surfactant
tives, SBE--CD and HP--CD, increase the oral bioavailability of cinnariz- stabilization. Langmuir, 2006, 22(21), 8951-8959.
ine in beagle dogs. J. Pharm. Sci., 1995, 84, 295-299. [34] Matteucci, M.E.; Brettmann, B.K.; Rogers, T.L.; Elder, E.J.; Williams, R.O.
[10] Rogers, T.L.; Nelsen, A.C.; Brown, J.N.; Sarkari, M.; Young, T.J.; Johnston, 3rd; Johnston, K.P. Design of potent amorphous drug nanoparticles for rapid
K.P.; Williams, R.O. A novel particle engineering technology to enhance generation of highly supersaturated media. Mol. Pharm., 2007, 4(5),
dissolution of poorly water soluble drugs: spray-freezing into liquid. Eur. J. 782-793.
Pharm. Biopharm., 2002, 54, 271-280. [35] Gassmann, P.; List, M.; Schweitzer, A.; Sucker, H. Hydrosols — alternatives
[11] Ventura, C. A.; Tirendi, S.; Puglisi, G.; Bousquet, E.; Panza, L. Improvement for the parenteral application of poorly watersoluble drugs. Eur. J. Pharm.
of water solubility and dissolution rate of ursodeoxycholic acid and che- Biopharm., 1994, 40, 64-72.
nodeoxy- cholic acid by complexation with natural and modified [36] Myerson, A.S.; Ginde, R. Handbook of Industrial Crystallization. Butter-
-cyclodextrins. Int. J. Pharm., 1997, 149, 1-13. worth- Heinemann, 1992, pp. 45-46.
[12] Floyd, A.G. Top ten considerations in the development of parenteral emul- [37] Bodmeier, R.; McGinity, J.M. Solvent selection in the preparation of poly
sions. Pharm. Sci. Technol., 1999, 4, 134-143. (DL-lactide) microspheres prepared by solvent evaporation method. Int. J.
[13] Nakano, M. Places of emulsions in drug delivery. Adv. Drug Deliv. Rev., Pharm., 1998, 43, 179-186.
2000, 45, 1-4. [38] Sah, H. Ethyl formate-alternative dispersed solvent useful in preparing
[14] Williams, R.O.; Mahaguna, V.; Sriwongjanya, M. Characterization of an PLGA microspheres. Int. J. Pharm., 2000, 195, 103-113.
inclusion complex of cholesterol and hydroxypropyl--cyclodextrin. Eur. J. [39] Trotta, M.; Gallarate, M.; Pattarino, F.; Morel, S. Emulsions containing
Pharm. Biopharm., 1998, 46, 355-360. partially water miscible solvents for the preparation of drug nanosuspensions.
[15] Serajuddin, A.T. Solid dispersion of poorly water soluble drugs: early prom- J. Control. Release., 2001, 76, 119-128.
ises, subsequent problems, and recent breakthroughs. J. Pharm. Sci., 1999, [40] Trotta, M.; Gallarate, M.; Carlotti, M.E.; Morel, S. Preparation of griseo-
88, 1058-1066. fulvin nanoparticles from water-dilutable microemulsions. Int. J. Pharm.,
[16] Moneghini, M.; Kikic, I.; Voinovich, D.; Perissutti, B.; Filipovic-Grcic, J. 2003, 254, 235-242.
Processing of carbamazepine-PEG 4000 solid dispersions with supercritical [41] Kocbek, P.; Baumgartner, S.; Kristl, J. Preparation and evaluation of nano-
carbon dioxide: preparation, characterization, and in vitro dissolution. Int. J. suspensions for enhancing the dissolution of poorly soluble drugs. Int. J.
Pharm., 2001, 222, 129-138. Pharm., 2006, 312, 179-186.
[15] Zerrouk, N.; Chemtob, C.; Arnaud, P.; Toscani, S.; Dugue, J. In vitro and in [42] Shekunov, B.Y.; Chattopadhyay, P.; Seitzinger, J.; Huff, R. Nanoparticles of
vivo evaluation of carbamazepine-PEG 6000 solid dispersions. Int. J. Pharm., poorly water-soluble drugs prepared by supercritical fluid extraction of e-
2001, 225, 49-62. mulsions. Pharm. Res., 2006, 23, 196-204.
[16] Lawrence, M.J.; Rees, G.D. Microemulsion-based media as novel drug [43] Kipp, J.E.; Wong, J.; Joseph, C.T.; Doty, M.; Mark, J.; Rebbeck, C.; Chris-
delivery systems. Adv. Drug Deliv. Rev., 2000, 45, 89-121. tine, L. Microprecipitation method for preparing submicron suspensions. US
[17] Schwendener, R.A.; Schott, H. Lipophilic 1-beta-D-arabinofuranosyl cyto- Patent, 6607784, 2003.
sine derivatives in liposomal formulations for oral and parenteral antileuke- [44] Chaubal, M.; Doty, M.; Kipp, J.; Papadopoulos, P.; Rebbeck, C.; Werling, J.;
mic therapy in the murine L1210 leukemia model. J. Cancer Res. Clin. On- Wong, J. Preparation of stable, sterile nanosuspensions for intravenous ad-
col., 1996, 122, 723-726. ministration. Proceedings of the Controlled Release Society Meeting, Salt
[18] Pereira de Oliveira, M.; Garcion, E.; Venisse, N.; Benoit, J.P.; Couet, W.; Lake City: UT, March 2003.
Olivier, J.C. Tissue Distribution of Indinavir Administered as Solid Lipid [45] Kipp, J.E.; Wong, J.; Doty, M.; Werling, J.; Rebbeck, C.; Brynjelsen S.
Nanocapsule Formulation in mdr1a (+/+) and mdr1a (-/-) CF-1 Mice. Pharm. Method for preparing submicron particle suspensions. US Patent, 0031719
Res., 2005, 2211, 1898-1905. A1, 2003.
[19] Rao, G.C.; Kumar, M.S.; Mathivanan, N.; Rao, M.E. Nanosuspensions as the [46] Noyes, A.A.; Whitney, W.R. The rate of solution of solid substances in their
most promising approach in nanoparticulate drug delivery systems. Phar- own solutions. J. Am. Chem. Soc., 1897, 19, 930-934.
mazie, 2004, 59(1), 5-9. [47] Hintz, R.J.; Johnson, K.C. The effect of particle size distribution on dissolu-
[20] Müller, R.H.; Peters, K.; Becker, R.; Kruss, B. Nanosuspensions — a novel tion rate and oral absorption. Int. J. Pharm., 1989, 51, 9-17.
formulation for the i.v. administration of poorly soluble drugs. 7th Int. Conf. [48] Abdou, H.M. Dissolution. Gennaro, A.R., Schwartz, J.B., Eds.: In: Rem-
On Pharm. Technol. (APV:APGI), Budapest, 1995a, pp. 491-492. ington’s Pharm. Sci., Mack Publishing Company: Easton, PA, 1990, vol. 18,
[21] Müller, R.H., Peters, K., Becker, R., Kruss, B. Nanosuspensions for the i.v. pp. 589-602.
administration of poorly soluble drugs — stability during sterilization and [49] Higuchi, W.I.; Ho, N.F.H.; Goldberg, A.H. Theories of dispersion techniques.
long-term storage. 22nd Int. Symp. Control. Release Bioact. Mater., Seattle, The Theory and Particle of Industrial Pharmacy, Lachman, L.; Lieberman,
1995b, pp. 574-575. H.A.; Kaning Eds.: J.L. Lea and Febiger: Philadelphia, 1970, 120-136.
[22] Müller, R.H.; Becker, R.; Kruss, B.; Peters, K. Pharmaceutical nanosuspen- [50] Nyström, C. Dissolution properties of soluble drugs: Theoretical background
sions for medicament administration as system of increased saturation solu- and possibilities to improve the dissolution behaviour. In: Emulsions and
bility and rate of solution. US Patent, No. 5858410, 1998. Nanosuspensions for the Formulation of Poorly Soluble Drugs. Müller, R.H.;
[23] Na, G.C.; Stevens, H.J. Jr.; Yuan, B.O.; Rajagopalan, N. Physical stability of Benita, S.; Böhm, B. Eds.: Medpharm: Stuttgart, 1998, 143-148.
ethyl diatrizoate nanocrystalline suspension in steam sterilization. Pharm. [51] Evans, D.F.; Wennerström, H. The Colloidal Domain: Where Physics,
Res., 1999, 16, 569-574. Chemistry, Biology and Technology Meet, 2nd ed.; VCH Publishers Inc.:
[24] Möschwitzer, J.; Achleitner, G.; Pomper, H.; Müller, R.H. Development of NewYork, 1994.
an intravenously injectable chemically stable aqueous omeprazole formula- [52] Grant, D.J.W; Brittain, H.G. In Physical Characterisation of Pharmaceutical
tion using nanosuspension technology. Eur. J. Pharm. Biopharm., 2004, 58, Solids. Brittain, H.G., Eds.: Marcel Dekker: New York, NY, USA, 1995,
615-619. 322-386.
[25] Merisko-Liversidge, E.; Linden, W. Stabilization of active agents by formu- [53] Mosharraf, M.N. The effect of particle size and shape on the surface specific
lation into nanoparticulate form. World Patent, WO03024424, 2003. dissolution rate of microsized practically insoluble drugs. Int. J. Pharm.,
[26] Rabinow, B. Nanosuspensions in drug delivery. Nat. Rev. Drug Discov., 1995, 122, 35-47.
2004, 3, 785-796. [54] Gao, L.; Zhang, D.; Chen M. Studies on pharmacokinetics and tissue distri-
[27] Van Eerdenbrugh, B.; Froyen, L.; Martens, J.A.; Blaton, N.; Augustijns, P.; bution of oridonin nanosuspensions. Int. J. Pharm., 2008, 355, 321-327.
Brewster, M.; Van den Mooter, G. Characterization of physico-chemical [55] Barber, T.A. In: Pharmaceutical Particulate Matter: Analysis and Control
properties and pharmaceutical performance of sucrose co-freeze-dried solid Interpharm: Buffalo Grove, 1993, Ch. 8.
nanoparticulate powders of the anti-HIV agent loviride prepared by media [56] Lines, R.W. In Liquid- and Surface-Borne Particle Measurement Handbook,
milling. Int. J. Pharm., 2007, 338, 198-206. Knapp, J.Z.; Barber, T.A.; Lieberman, A.; Eds.: Marcel Dekker: New York,
[28] Mittapalli, P.K.; Yamasani, M.R.; Shashank, A. Formulation of nanosuspen- 1996, Ch. 4.
sions of albendazole for oral administration. Curr. Nanosci., 2008, 4, 53-58. [57] Higgins, J.P. Spectroscopic approach for on-line monitoring of particle size
426 Current Nanoscience, 2009, Vol. 5, No. 4 Pu et al.

during the processing of pharmaceutical nanoparticles. Anal. Chem., 2003, delivery to cells and tissue. Adv. Drug Deliv. Rev., 2003, 55(3), 329-347.
75, 1777-1785. [85] Van Eerdenbrugh, B.; Vercruysse, S.; Martens, J.A.; Vermant, J.; Froyen, L.;
[58] Müller, R.H. Zetapotential und partikelladung—kurze theorie, praktische Van Humbeeck, J.; Van den Mooter, G.; Augustijns, P. Microcrystalline cel-
meßdurchführung, dateninterpretation. Wissenschaftliche Verlagsgesellschaft lulose, a useful alternative for sucrose as a matrix former during
Stuttgart, 1996. freeze-drying of drug nanosuspensions – a case study with itraconazole. Eur.
[59] Riddick, T.M. Zeta-meter manual. Zeta Meter Inc., New York, 1968. J. Pharm. Biopharm., 2008b, 70(2), 590-596.
[60] Müller, R. H.; Jacobs, C. Production and characterization of a budesonide [86] Van Eerdenbrugh, B.; Froyen, L.; Van Humbeeck, J.; Martens, J.A.;
nanosuspension for pulmonary administration. Pharm. Res., 2002b, 19, Augustijns, P.; Van Den Mooter, G. Aternative matrix formers for nanosus-
189-194. pension solidification Dissolution performance and X-ray microanalysis as
[61] Yang, J.Z.; Young, A.L.; Chiang, P.C.; Thurston, A.; Pretzer, D.K. Flutica- an evaluation tool for powder dispersion. Eur. J. Pharm. Sci., 2008c, 35(4),
sone and budesonide nanosuspensions for pulmonary delivery: preparation, 344-353.
characterization, and pharmacokinetic studies. J. Pharm. Sci., 2008, 97(11), [87] Van Eerdenbrugh, B.; Froyen, L.; Van Humbeeck, J.; Martens, J.A.;
4869-4878. Augustijns, P.; Van Den Mooter, G. Drying of crystalline drug nanosuspen-
[62] Liang, Y.C.; Binner, J.G.P. Effect of triblock copolymer non-ionic surfac- sions – the importance of surface hydrophobicity on dissolution behavior
tants on the rheology of 3 mol% yttria stabilised zirconia nanosuspensions. upon redispersion. Eur. J. Pharm. Sci., 2008a, 35(1-2), 127-135.
Ceram. Int., 2008, 34, 293-297. [88] O’Driscoll, C.M. In: Lymphatic Transport of Drugs, Charman, W. N.; Stella,
[63] Müller, R.H.; Grau, M.J. Increase of dissolution rate and solubility of poorly V. J. Eds. CRC Press Inc., Boca Raton, 1992, 1-35.
water soluble drugs as nanosuspension. Proceedings. World Meeting [89] Yang, T.; Choi, M.K.; Cui, F.D.; Kim, J.S.; Chung, S.J.; Shim, C.K.; Kim,
APGI/APV, Paris, 1998, 2, 62-624. D.D. Preparation and evaluation of paclitaxel-loaded PEGylated immu-
[64] Scholer, N.; Krause, K.; Kayser, O.; Müller, R.H.; Borner, K.; Hahn, H.; noliposome. J. Control. Release, 2007, 120, 169-177.
Liesenfeld, O. Atovaquone nanosuspensions show excellent therapeutic ef- [90] Lu, Y.; Li, J.; Wang, G. In vitro and in vivo evaluation of mPEG-PLA modi-
fect in a new murine model of reactivated toxoplasmosis. Antimicrob. Agents fied liposomes loaded glycyrrhetinic acid. Int. J. Pharm., 2008, 356,
Chemother., 2001, 45, 1771-1779. 274-281.
[65] Bond, L.; Allen, S.; Davies, M.C.; Roberts, C.J.; Shivji, A.P.; Tendler, S.J.; [91] Nakada, Y.; Tudomi, R.; Sakurai, K.; Takahashi, Y. Evaluation of
Williams, P.M.; Zhang, J. Differential scanning calorimetry and scanning long-circulating nanoparticles using biodegradable ABA triblock copolymers
thermal microscopy analysis of pharmaceutical materials. Int. J. Pharm., containing of poly(L-lactic acid) A-blocks attached to central
2002, 243, 71-82. poly(oxyethylene) B-blocks in vivo. Int. J. Pharm., 1998, 175, 109-117.
[66] Chen, Y.; Liu, J.; Yang, X.; Zhao, X.; Xu, H. Oleanolic acid nanosuspen- [92] Chen, J.; Tian, B.; Yin, X.; Zhang, Y.; Hu, D.; Hu, Z.; Liu, M.; Pan, Y.; Zhao,
sions: preparation, in-vitro characterization and enhanced hepatoprotective J.; Li, H.; Hou, C.; Wang, J.; Zhang, Y. Preparation, characterization and
effect. J. Pharm. Pharmacol., 2005, 57, 259-264. transfection efficiency of cationic PEGylated PLA nanoparticles as gene de-
[67] Teeranachaideekul, V.; Junyaprasert, V.B.; Souto, E.B.; Müller, R.H. De- livery systems. J. Biotechnol., 2007, 130(2), 107-113.
velopment of ascorbyl palmitate nanocrystals applying the nanosuspension [93] Tan, J.P.; Wang, Q; Tam, K.C. Control of burst release from nanogels via
technology. Int. J. Pharm., 2008, 354, 227-234. layer by layer assembly. J. Control. Release, 2008, 1283, 248-254.
[68] Chingunpitak, J.; Puttipipatkhachorn, S.; Chavalitshewinkoon-Petmitr, P.; [94] Kayser, O. A new approach for targeting to Cryptosporidium parvum using
Tozuka, Y.; Moribe, K.; Yamamoto, K. Formation, Physical stability and in mucoadhesive nanosuspensions: research and applications. Int. J. Pharm.,
vitro antimalarial activity of dihydroartemisinin nanosuspensions obtained by 2001, 214, 83-85.
co-grinding method. Drug Dev. Ind. Pharm., 2008, 34, 314-322. [95] Müller, R.H.; Jacobs, C. Buparvaquone mucoadhesive nanosuspension:
[69] Lindfors, L.; Skantze, P.; Skantze, U.; Westergren, J.; Olsson, U. Amorphous preparation, optimisation and long-term stability. Int. J. Pharm., 2002a, 237,
drug nanosuspensions. 3. particle dissolution and crystal growth. Langmuir, 151-161.
2007, 23, 9866-9874. [96] Onyuksel, H.; Rubinstein, I. Materials and methods for making improved
[70] Duchêne, D.; Ponchel G. Bioadhesion of solid oral dosage forms, why and micelle compositions; lipid bonded to water soluble polymer. US Patent,
how? Eur. J. Pharm. Biopharm., 1997, 44, 15-23. 6217886, 2001.
[71] Yoncheva, K.; Lizarraga, E.; Irache, J.M. Pegylated nanoparticles based on [97] Wu, N.Z.; Da, D.; Rudoll, T.L.; Needham, D.; Whorton, A.R.; Dewhirst,
poly (methyl vinyl ether-co-maleic anhydride): preparation and evaluation of M.W. Increased microvascular permeability contributes to preferential ac-
their bioadhesive properties. Eur. J. Pharm. Sci., 2005, 24, 411-419. cumulation of stealth liposomes in tumor tissue. Cancer Res., 1993, 53,
[72] Mittapalli, P.K.; Yamasani, M.R.; Shashank, A. Improved bioavailability of 3765-3770.
albendazole following oral administration of nanosuspension in rats. Curr. [98] Lode, J.; Fichtne,r I.; Kreuter, J.; Berndt, A.; Diederichs, J.E.; Reszka, R.
Nanosci., 2007, 3, 191-194. Influence of surface-modifying surfactants on the pharmacokinetic behavior
[73] Blunk, T.; Hochstrasser, D.F.; Sanchez, J.-C.; Müller, B.W.; Müller, R.H. of 14C-Poly (methylmethacrylate) nanparticles in experimental tumor mod-
Colloidal carriers for intravenous drug targeting: Plasma protein adsorption els. Pharm. Res., 2001, 18, 1613-1619.
patterns on surface-modified latex particles evaluated by two-dimensional [99] Zahr, A.S.; de Villiers, M; Pishko, M.V. Encapsulation of drug nanoparticles
polyacrylamide gel electrophoresis. Electrophoresis, 1993, 14, 1382-1387. in self-assembled macromolecular nanoshells. Langmuir, 2005, 21, 403-410.
[74] Blunk, T.; Lück, M.; Calvör, A.; Hochstrasser, D.F.; Sanchez, J.-C.; [100] Kreuter, J.; Petrov, V.E.; Kharkevich, D.A.; Alyautdin, R.N. Influence of the
Müller, B.W.; Müller, R.H. Kinetics of plasma protein adsorption on model type of surfactant on the analgesic effects induced by the peptide dalargin
particles for controlled drug delivery and drug targeting. Eur. J. Pharm. after 1st delivery across the blood-brain barrier using surfactant coated
Biopharm., 1996, 42, 262-268. nanoparticles. J. Control. Release, 1997, 49, 81-87.
[75] Lück, M.; Schroder, W.; Harnisch, S.; The, K.; Blunk, T.; Paulke, B.R.; [101] Desai, N.; Trieu, V.; Yao, Z.; Louie, L.; Ci, S.; Yang, A.; Tao, C.; De, T.;
Kresse, M.; Müller R.H. Identification of plasma proteins facilitated by en- Beals, B.; Dykes, D.; Noker, P.; Yao, R.; Labao, E.; Hawkins, M.;
richment on particulate surfaces: Analysis by two-dimensional electrophore- Soon-Shiong, P. Increased antitumor activity, intratumor paclitaxel concen-
sis and N-terminal microsequencing. Electrophoresis, 1997a, 18, 2961-2967. trations, and endothelial cell transport of cremophor-free, albumin-bound pa-
[76] Lück, M.; Paulke, B.R.; Schroder, W.; Blunk, T.; Müller, R.H. Analysis of clitaxel, ABI-007, compared with cremophor-based paclitaxel. Clin. Cancer
plasma protein adsorption on polymeric nanoparticles with different surface Res., 2006, 12, 1317-1324.
characteristics. J. Biomed. Mater. Res., 1997b, 1, 478-485. [102] Ain-Ai, A.; Gupta, P.K. Effect of arginine hydrochloride and hydroxypropyl
[77] Van Oss, C.J.; Absolom, D.R.; Neumann, H.W. Interaction of phagocytes cellulose as stabilizers on the physical stability of high drug loading nano-
with other blood cells and with pathogenic and nonpathogenic microbes. Ann. suspensions of a poorly soluble compound. Int. J. Pharm., 2008, 351,
NY Acad. Sci., 1984, 416, 332-350. 282-288,
[78] Wallis, K.H.; Müller, R.H. Determination of the surface hydrophobicity of [103] Shudo, J.; Pongpeerapa,t A.; Wanawongthai, C.; Moribe, K.; Yamamoto, K.
colloidal dispersions by mini-hydrophobic interaction chromatography. In vivo assessment of oral administration of probucol nanoparticles in rats.
Pharm. Ind., 1993, 55, 1124-1128. Biol. Pharm. Bull., 2008, 31, 321-325.
[79] Müller, R.H. Manufacturing of nanoparticles by milling and homogenization [104] Peters, K.; Leitzke, S.; Diederichs, J.E.; Borner, K.; Hahn, H.; Müller, R.H.;
techniques, Gupta, R.B.; Kompella, U.B., Eds. Nanoparticle Technology for Ehlers, S. Preparation of a clofazimine nanosuspension for intravenous use
Drug Delivery, Drugs and the Pharmaceutical Sciences, Taylor & Francis and evaluation of its therapeutic efficacy in murine Mycobacterium avium
Group, LLC: NewYork, USA, 2006, vol. 159, pp. 21-51. infection. J. Antimcrob. Chem., 2000, 45, 77-83.
[80] Abdelwahed, W.; Degobert, G.; Fessi, H. Investigation of nanocapsules [105] Sigfridsson, K.; Forssén, S.; Holländer, P.; Skantze, U.; de Verdier, J. A
stabilization by amorphous excipients during freeze-drying and storage. Eur. formulation comparison, using a solution and different nanosuspensions of a
J. Pharm. Biopharm., 2006, 63, 87-94. poorly soluble compound. Eur. J. Pharm. Biopharm., 2007, 67, 540-547.
[81] Abdelwahed, W.; Degobert, G.; Fessi, H. A pilot study of freeze drying of [106] Kassem, M.A.; Abdel Rahman, A.A.; Ghorab, M.M.; Ahmed, M.B.; Khalil,
poly(epsiloncaprolactone) nanocapsules stabilized by poly(vinyl alcohol): R.M. Nanosuspension as an ophthalmic delivery system for certain glucocor-
formulation and process optimization. Int. J. Pharm., 2006, 17, 178-188. ticoid drugs. Int. J. Pharm., 2007, 340, 126-133.
[82] Konan, Y.N.; Gurny, R.; Allémann, E. Preparation and characterization of [107] Kayser, O. Nanosuspensions for the formulation of aphidicolin to improve
sterile and freeze-dried sub-200 nm nanoparticles. Int. J. Pharm., 2002, 233, drug targeting effects against Leishmania infected macrophages. Int. J.
239-252. Pharm., 2000, 196, 253-256.
[83] Chaubal, M.V.; Popescu, C. Conversion of nanosuspensions into dry pow- [108] Jinno, J.; Kamada, N.; Miyake, M.; Yamada, K.; Mukai, T.; Odomi, M.;
ders by spray drying: A case study. Pharm. Res., 2008, 25(10), 2302-2308. Toguchi, H.; Liversidge, G.G.; Higaki, K.; Kimura, T. Effect of particle size
[84] Panyam, J.; Labhasetwar, V. Biodegradable nanoparticles for drug and gene reduction on dissolution and oral absorption of a poorly water-soluble drug,
Formulation of Nanosuspensions as a New Approach Current Nanoscience, 2009, Vol. 5, No. 4 427

cilostazol, in beagle dogs. J. Control. Release, 2006, 111, 56-64. the rat. Int. J. Pharm., 2007, 339, 251-260.
[109] Langguth, P.; Hanafy, A.; Frenzel, D.; Grenier, P.; Nhamias, A.; Ohlig, T.; [121] Moghimi, S.M.; Hunter, A.C.; Murray, J.C. Long-circulating and tar-
Vergnault, G.; Spahn-Langguth, H. Nanosuspension formulations for get-specific nanoparticles: theory to practice. Pharmacol. Rev., 2001, 53,
low-soluble drugs: pharmacokinetic evaluation using spironolactone as 283-318.
model compound. Drug Dev. Ind. Pharm., 2005, 31, 319-329. [122] Pignatello, R.; Bucolo, C.; Ferrara, P.; Maltese, A.; Puleo, A.; Puglisi, G.
[110] Douroumis, D.; Fahr, A. Stable carbamazepine colloidal systems using the Eudragit RS100® nanosuspensions for the ophthalmic controlled delivery of
cosolvent technique. Eur. J. Pharm. Sci., 2007, 30, 367-374. ibuprofen. Eur. J. Pharm. Sci., 2002a, 16, 53-61.
[111] Jacobs, C.; Kayser, O.; Müller R.H. Nanosuspensions as a new approach for [123] Pignatello, R.; Bucolo, C.; Spedalieri, G.; Maltese, A.; Puglisi, G. Flur-
the formulation for the poorly soluble drug tarazepide. Int. J. Pharm., 2000, biprofen-loaded acrylate polymer nanosuspensions for ophthalmic applica-
196, 161-164. tion. Biomaterial, 2002b, 23, 3247-3255.
[112] Zhang, D.; Tan, T.; Gao, L.; Zhao, W.; Wang, P. Preparation of azithromycin [124] Borgstrom, L. The importance of the device in asthma therapy. Respir. Med.,
nanosuspensions by high pressure homogenization and its physicochemical 2001, 95, S26-S29.
characteristics studies. Drug Dev. Ind. Pharm., 2007, 33, 569-575. [125] Courrier, H.M.; Butz, N; Vandamme, T.F. Pulmonary drug delivery systems:
[113] Vergote, G.J.; Vervaet, C.; Van Driessche, I.; Hoste, S.; De Smedt, S.; Recent developments and prospects. Crit. Rev. Ther. Drug Carrier Syst.,
Demeester, J.; Jain, R.A.; Ruddy, S.; Remon, J.P. An oral controlled release 2002, 19, 425-498.
matrix pellet formulation containing nanocrystalline ketoprofen. Int. J. [126] Sakagami, M. In vivo, in vitro and ex vivo models to assess pulmonary ab-
Pharm., 2001, 219, 81-87. sorption and disposition of inhaled therapeutics for systemic delivery. Adv.
[114] Jia, L.; Wong, H.; Cerna, C.; Weitman, S.D. Effect of nanonization on ab- Drug Deliv. Rev., 2006, 58(9-10), 1030-1060.
sorption of 301029: ex vivo and in vivo pharmacokinetic correlations deter- [127] Liao, X.; Wiedmann, T.S. Solubilization of cationic drugs in lung surfactant.
mined by liquid chromatography/mass spectrometry. Pharm. Res., 2002, 19, Pharm. Res., 2003, 20(11), 1858-1863.
1091-1096. [128] Berry, J.; Kline, L.C.; Sherwood, J.K.; Chaudhry, S.; Obenauer-Kutner, L.;
[115] Merisko-Liversidge, E. Formulation and antitumor activity evaluation of Hart, J.L.; Sequeira, J. Influence of the size of micronized active pharmaceu-
nanocrystalline suspensions of poorly soluble anticancer drugs. Pharm. Res., tical ingredient on the aerodynamic particle size and stability of a metered
1996, 13, 272-278. dose inhaler. Drug Dev. Ind. Pharm., 2004, 30(7), 705-714.
[116] Boedeker, B.H.; Lojeski, E.W.; Kline, M.D.; Haynes, D.H. Ultra-long dura- [129] Leong, B.K.; Coombs, J.K.; Sabaitis, C.P.; Rop, D.A.; Aaron, C.S. Quantita-
tion local anesthesia produced by injection of lecithin-coated tetracaine mi- tive morphometric analysis of pulmonary deposition of aerosol particles in-
crocrystals. J. Clin. Pharmacol., 1994, 34, 699-702. haled via intratracheal nebulization, intratracheal instillation or nose-only
[117] Karan, S.M.; Lojeski, E.W.; Haynes, D.H.; Bina, S.; Wesche, D.L.; Boedeker, inhalation in rats. J. Appl. Toxicol., 1998, 18(2), 149-160.
B.H.; Muldoon, S.M. Intravenous lecithin-coated microcrystals of dantrolene [130] Snipes, M.B.; Chavez, G.T.; Muggenburg, B.A. Disposition of 3-, 7-, and
are effective in the treatment of malignant hyperthermia:an investigation in 13-microns microspheres instilled into lungs of dogs. Environ. Res., 1984,
rats, dogs, and swine. Anesth. Analg., 1996, 82, 796-802. 33(2), 333-342.
[118] Shulman, M. Treatment of cancer pain with epidural butylamino benzoate [131] Telko, M.J.; Hickey A.J. Dry powder inhaler formulation. Respir. Care.,
suspension. Reg. Anesth., 1987, 12, 1-4. 2005, 50(9), 1209-1227.
[119] Moghimi, S.M. Hunter, A.C.; Murray, J.C. Longcirculating and tar- [132] Buckton, G. Characterisation of small changes in the physical properties of
get-specific nanoparticles: theory to practice. Pharmacol. Rev., 2001, 53(2), powders of significance for dry powder inhaler formulations. Adv. Drug De-
283-318. liv. Rev., 1997, 26(1), 17-27.
[120] Rabinow, B.; Kipp, J.; Papadopoulos, P.; Wong, J.; Glosson, J.; Gass, J.; Sun, [133] Ponchel, G.; Montisci, M.-J.; Dembri, A.; Durrer, C.; Duchene, D. Mucoad-
C.S.; Wielgos, T.; White, R.; Cook, C.; Barker, K.; Wood, K. Itraconazole hesion of colloidal particulate systems in the gastrointestinal tract. Eur. J.
IV nanosuspension enhances efficacy through altered pharmacokinetics in Pharm. Biopharm., 1997, 4(1), 25-31.

Received: November 21, 2008 Revised: May 27, 2009 Accepted: June 03, 2009

View publication stats

Das könnte Ihnen auch gefallen