Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Advanced Drug Delivery Reviews 47 (2001) 3–19

www.elsevier.com / locate / drugdeliv

Nanosuspensions as particulate drug formulations in therapy


Rationale for development and what we can expect for the
future
¨ *, C. Jacobs, O. Kayser
R.H. Muller
¨ Berlin, Institut f ur
Freie Universitat ¨ Pharmazie, Pharmazeutische Technologie, Biopharmazie and Biotechnologie, Kelchstraße 31,
D-12169 Berlin, Germany

Abstract

An increasing number of newly developed drugs are poorly soluble; in many cases drugs are poorly soluble in both
aqueous and organic media excluding the traditional approaches of overcoming such solubility factors and resulting in
bioavailability problems. An alternative and promising approach is the production of drug nanoparticles (i.e. nanosuspen-
sions) to overcome these problems. The major advantages of this technology are its general applicability to most drugs and
its simplicity. In this article, the production of nanoparticles on a laboratory scale is presented, special features such as
increased saturation solubility and dissolution velocity are discussed, and special applications are highlighted, for example,
mucoadhesive nanosuspensions for oral delivery and surface-modified drug nanoparticles for site-specific delivery to the
brain. The possibilities of large scale production — the prerequisite for the introduction of a delivery system to the market —
are also discussed.  2001 Elsevier Science B.V. All rights reserved.

Keywords: Nanosuspension; Homogenisation; Drug targeting; Drug delivery; Parasitic infection; Leishmania; Poor solubility

Contents

1. Introduction ............................................................................................................................................................................ 4
2. Production of nanosuspensions on a laboratory scale.................................................................................................................. 4
3. Physical and chemical properties of nanosuspensions ................................................................................................................. 6
3.1. Characterization of nanosuspensions.................................................................................................................................. 6
3.2. Physical and chemical properties ....................................................................................................................................... 8
4. Surface modification of nanosuspensions .................................................................................................................................. 9
5. Biological properties of nanosuspensions and medical indications............................................................................................... 11
5.1. Oral administration of nanosuspensions ............................................................................................................................. 11
5.2. Parenteral nanosuspensions — intravenous administration route .......................................................................................... 12
6. Large-scale production of nanosuspensions ............................................................................................................................... 15
7. Perspectives ............................................................................................................................................................................ 17
References .................................................................................................................................................................................. 17

*Corresponding author. Tel.: 149-30-838-50416; fax: 149-30-838-50478.


¨
E-mail address: mpharma@zedat.fu-berlin.de (R.H. Muller).

0169-409X / 01 / $ – see front matter  2001 Elsevier Science B.V. All rights reserved.
PII: S0169-409X( 00 )00118-6
4 ¨
R.H. Muller et al. / Advanced Drug Delivery Reviews 47 (2001) 3 – 19

1. Introduction ers [5–7] using a precipitation technique. Precipi-


tation was performed by dissolving the drug in a
Poorly soluble drugs are a general problem in solvent and adding this solvent to a non-solvent
pharmaceutical drug formulation [1]. Typical prob- (so-called ‘via humida paratum’). The basic chal-
lems associated with poorly soluble drugs are a too lenge of this technique is that during the precipitation
low bioavailability and / or erratic absorption. In case procedure the growing of the drug crystals needs to
of a too low bioavailability after oral administration, be limited by surfactant addition to avoid formation
parenteral administration cannot solve this problem of microparticles. The requisites limiting the ap-
in many cases. Due to the poor solubility, intraven- plicability of the precipitation technique are the
ous injection as a solution is not possible. Parenteral needs of the drug (a) to be soluble at least in one
administration as a micronized product (e.g. i.m. or solvent and (b) that this solvent needs to be miscible
i.p.) does not lead necessarily to sufficiently high with a non-solvent. These prerequisites exclude the
drug levels because the solute volume at the injection processing of drugs which are simultaneously poorly
site is too low. Low saturation solubility, generally soluble in aqueous and in non-aqueous media.
combined with a low dissolution velocity is the In the first generation applying disintegration
obstacle preventing sufficiently high blood levels. techniques, this was achieved by pearl milling lead-
Possible exceptions are only drugs being poorly ing to the product NanoCrystals  . This patent-
soluble but highly potent. protected technology was developed in 1990 by
Attempts to increase the saturation solubility, and Liversidge et al. [4] and formerly owned by the
thus solving the problem, are solubility enhancement company NanoSystems, recently acquired by Elan.
by using solubilization (e.g. mixed micelles as in To produce NanoCrystals  , the drug powder is
Valium MM  for i.v. injection), non-specific or dispersed in a surfactant solution and the obtained
specific complexation (e.g. addition of poly- suspension undergoes a pearl milling process for
ethylenglycol (PEG) or use of cyclodextrins) and hours up to several days.
solvent mixtures (e.g. ethanol–water, up to 20% The second generation products are drug
ethanol are possible). The limited success of these nanoparticles produced by high pressure homogeni-
attempts is documented by the low number of zation leading to the so called nanosuspensions
products on the pharmaceutical market based on (DissoCubes  ). The nanosuspensions were de-
these principles. The problems to find a suitable ¨
veloped by Muller et al. [3] in 1994 and the patent
formulation are even greater in case the drugs are rights owned by DDS (Drug Delivery Services)
poorly soluble in aqueous media and at the same GmbH in Germany, now owned by SkyePharma
time in organic media. This excludes the use of PLC. This chapter presents the production of
solvent mixtures a priori. nanosuspensions on a laboratory scale but also on
A general approach used for many years is the large industrial scale, discusses their special features,
micronization of poorly soluble drugs by colloid reviews present applications including bioavailabilty
mills or jet mills. The overall particle size dis- aspects, especially site-specific delivery and high-
tribution ranges from 0.1 mm to approximately 25 lights the future perspectives. Especially, the po-
mm, only negligible amount being below 1 mm in the tential for targeting to the mucosa of the gastroin-
nanometer range [2]. Micronization increases the testinal tract (GIT) after oral administration, and to
dissolution velocity of the drug due to the increase in the cells of the mononuclear phagocytic system
surface area but does not change the saturation (MPS) to treat MPS infections are discussed.
solubility. At very low saturation solubility, the
achieved increase in dissolution velocity does not
lead to a sufficiently high bioavailability. 2. Production of nanosuspensions on a
The next development step was transformation of laboratory scale
the micronized drug powder (i.e. drug microparti-
cles) to drug nanoparticles [3,4]. In the eighties, drug The disintegration principle for obtaining nanosus-
nanoparticles were produced by Sucker and co-work- pensions are the cavitation forces created in high
¨
R.H. Muller et al. / Advanced Drug Delivery Reviews 47 (2001) 3 – 19 5

pressure homogenizers, e.g. piston-gap homogenizers temperature [8]. In consequence, water starts boiling
like APV Gaulin types (Fig. 1). The drug powder is at room temperature due to the high pressure, gas
dispersed in an aqueous surfactant solution by high bubbles are formed which implode ( 5 cavitation)
speed stirring. The obtained ‘macro’-suspension is when the fluid leaves the homogenisation gap. These
passed through a high pressure homogenizer apply- cavitation forces are strong enough to break the drug
ing typically 1500 bar and three to ten up to a microparticles to drug nanoparticles.
maximum of 20 passes ( 5 homogenisation cycles). The mean particle size in the nanometer range
Generally it is recommended to start with a powder obtained by this procedure depends on the pressure
as fine as possible, that means a jet milled product. and number of cycles applied, in addition it is
The suspension passes a very small homogenization affected by the hardness of the drug itself. For
gap in the homogenizer, typically having a width of, example, for paclitaxel nanosuspension a mean di-
for example, approximately 25 mm at 1500 bar (Fig. ameter of 330 nm was reported [9], for clofazemine
2). Due to the narrowness of the gap the streaming 600 nm [10,11], and for the drug RMKP22 540 nm
velocity of the suspension increases tremendously, [3,11]. Of course, these diameters depend also on the
that means the dynamic fluid pressure increases. production conditions chosen. For example, for
Simultaneously the static pressure on the fluid de- budesonide a pressure of 1500 bar and ten cycles
creases below the boiling point of water at room lead to a mean PCS diameter of 511 nm, increasing
the cycle numbers to 15 reduces the size to 462 nm,
and increasing the pressure to 2500 bar and ten
cycles leads to particles with a diameter of 363 nm
[12]. In summary, a certain size can be produced in a
controlled way by adjusting the production parame-
ters pressure and cycle number accordingly. For each
drug, a minimum size exists which can be achieved
by applying a certain pressure, this minimum size
depends on the power density in the homogenizer
and the hardness of the drug itself.
Homogenizers to be used on a laboratory scale
are, for example, the APV Micron LAB 40 (APV
Deutschland GmbH, Lubeck, ¨ Germany) but also
other piston-gap homogenizers from Avestin (Aves-
tin Inc., Ottawa, Canada) and Stansted (Stansted
Fluid Power Ltd., Stansted, UK). The LAB 40 has a
minimum batch volume of 20 ml and a maximum of
40 ml, thus allowing the cost-effective processing of
even expensive drug materials. Even smaller vol-
umes can be prepared by using, for example, the
Avestin EmulsiFlex-B3 (volume 3.5 ml). The op-
timum particle size of nanosuspension depends on
the nature of the drug (therapeutic field) and the
desired biopharmaceutical properties. In cases where
a very fast dissolution is required, a size of approxi-
mately 100–200 nm is preferred. Where prolonged
dissolution is desired (e.g. mucoadhesive nanosus-
pension for treatment of Cryptosporidium infec-
tions), the mean particle diameter might be placed in
¨
Fig. 1. APV LAB 40 (APV Deutschland GmbH, Lubeck, Ger- the upper nanometer range, e.g. 800–1000 nm. For
many). intravenous administration, larger nanosuspensions
6 ¨
R.H. Muller et al. / Advanced Drug Delivery Reviews 47 (2001) 3 – 19

Fig. 2. Production of nanosuspensions by high pressure homogenization (I: implosion area, II: boiling area).

are preferred for targeting to the MPS cells (to avoid 3. Physical and chemical properties of
complete dissolution of the particles before they nanosuspensions
reach the macrophages). For a fast dissolution of the
drug in the blood, small nanosuspensions are more 3.1. Characterization of nanosuspensions
suited. It should be pointed out, that a relationship
such as ‘the smaller the drug nanoparticles, the better Essential characterization parameters for nanosus-
the product’ does not exist. The size needs to be pensions are:
tailor-made specific for each therapeutic purpose.
Typical concentrations of solid in the suspensions (1) Size and size distribution
processed are 10 or 20% with Stanstead homogeniz- (2) Particle charge (zeta potential)
ers; however 40% suspensions were also processed (3) Crystalline status
[13]. The solid concentration chosen depends on the (4) Dissolution velocity and saturation solubility.
final application of the nanosuspension. For intraven-
ous injection 10% or even less are sufficient for For surface-modified nanosuspensions a number of
further processing of the nanosuspension to a dry additional parameters have to be investigated to
product (e.g. by granulation to tablets); a higher solid obtain a complete picture, especially with relevance
content is desirable to remove less water. for the in vivo behavior:
¨
R.H. Muller et al. / Advanced Drug Delivery Reviews 47 (2001) 3 – 19 7

(5) Adhesion properties (in case of mucoadhesive electrophoresis and typically expressed as electro-
particles) phoretic mobility [(mm / S) /(V/ cm)] or converted to
(6) Surface hydrophilicity / hydrophobicity the zeta potential (mV). For a physically stable
(7) Interaction with body proteins. nanosuspension solely stabilized by electrostatic
repulsion, a zeta potential of 630 mV is required as
The mean size and width of distribution (polydis- a minimum [15,16]. In the case of a combined
persity index) is typically determined by photon electrostatic and steric stabilization, as a rough guide
correlation spectroscopy (PCS) [14]. The measuring line of 20 mV is sufficient.
range of PCS is limited to approximately 3 nm–3 Determination of dissolution velocity is important
mm. Therefore additionally laser diffractometry (LD) to assess the benefits compared to the traditional
is used to detect any content of particles in the drug formulation (e.g. coarse powder, micronized
micrometer range or aggregates of drug nanoparti- product). To determine the dissolution velocity, the
cles. Depending on the type of equipment employed, methods described in the Pharmacopoeia can be used
the measuring range is approximately 0.05–80 mm (e.g. for determination of the saturation solubility
up to maximum of 2000 mm. The LD data are shaking experiments at different temperatures (4, 20,
volume data, typical characterization parameters are 408C) need to be performed until equilibrium has
the diameters 50, 90, 95, and 99% (i.e. a diameter of been reached). Special feature of the nanosuspen-
99% means that 99% of the volume of the particles sions is an increased saturation solubility. Increased
is below the given size in micrometers). dissolution velocity and increased saturation solu-
For nanosuspensions to be administered intraven- bility — apart from the adhesive properties of
ously, an additional analysis by Coulter counter nanosuspensions — are the basic benefit of nanosus-
technique is essential. In contrast to the LD pro- pensions compared to traditional dosage forms. To a
viding only a relative size distribution, the Coulter large extent, these two parameters allow estimation
counter gives absolute data (i.e. absolute number of of the change in in vivo performance (blood profiles,
particles per volume unit for the different size plasma peaks, bioavailability).
classes). For intravenous injection the content of For intravenously injected nanosuspensions, addi-
particles larger 5 mm is critical and has to be under a tional parameters need to be determined which affect
certain limit. Bearing in mind that the smallest size the in vivo fate of the drug nanoparticles. Surface
of blood capillaries is 5–6 mm, too high for a hydrophilicity / hydrophobicity is considered as one
number of such particles that would lead to capillary of the important parameters affecting the in vivo
blockade and embolism. Therefore, this factor needs organ distribution after i.v. injection. The surface
to be controlled by Coulter counter analysis. hydrophobicity determines the interaction with cells
The crystalline structure of the nanosuspensions prior to phagocytosis [17,18]; in addition, it is a
can be assessed by differential scanning calorimetry relevant parameter for the adsorption of plasma
(DSC) [12]. This is especially important when a drug proteins — the key factor for organ distribution
exists in different polymorphic forms. In addition, [19,21–23]. To avoid artefacts, the surface hydro-
the high pressure homogenization technique for phobicity needs to be determined in the original
nanosuspensions can be used to particles with an environment of the drug nanoparticles, that means in
amorphous fraction, thus leading to an enhancement aqueous dispersion medium. A suitable technique is
of the saturation solubility. The extend of such a hydrophobic interaction chromatography (HIC), pre-
transform can be quantified and its stability during viously employed to determine the surface hydro-
storage monitored by DSC — preferentially in phobicity of bacteria [24], and then transferred to the
combination with X-ray analysis. In general, wide characterization of nanoparticulate drug carriers. For
angle X-ray analysis (WAXS) is employed because it details, we refer to Refs. [18,25,26].
is easily accessible (in contrast to SAXS). The qualitative and quantitative composition of the
Particle charge is a stability determining parameter protein absorption pattern, observed after i.v. in-
in aqueous nanosuspensions. It is measured by jection of the particles, is today generally recognized
8 ¨
R.H. Muller et al. / Advanced Drug Delivery Reviews 47 (2001) 3 – 19

as the essential key factor for organ distribution 3.2. Physical and chemical properties
[20–22,27,28]. The protein analysis by 2-D PAGE
was modified and especially adapted to the analysis An outstanding feature of nanosuspensions is the
of protein adsorption patterns on nanoparticles [20]. increase in saturation solubility and consequently an
Time-dependent adsorption patterns of proteins on increase in the dissolution velocity of the compound.
nanoparticles were determined after incubation of This increase in the dissolution velocity takes place
polymeric particles with plasma or serum [20,29,30], in addition to the increase caused by the enlargement
but also after collection of i.v. injected particles in of the surface area, e.g. exploited in micronized
animals [31,32]. products. In general, in textbooks, the saturation
Basic correlations could be established between solubility is defined as being a compound-specific
the protein adsorption patterns and the organ dis- constant depending only on the temperature. In case
tribution, here reflected by MPS cell uptake, certain of polymorphism, the saturation solubility depends
particles circulating in the blood [29], particles taken also on the crystalline structure (i.e. the inner
up by the bone marrow, and particles delivering energy) — this means solubility is best for the
drugs to the brain [30,33]. These established correla- polymorphic modification that is characterised by
tions can now be fully exploited to produce in a highest energy and lowest melting point. However
controlled way target-specific nanosuspensions. For — apart from temperature and crystalline / amor-
detailed information about this concept of ‘differen- phous status — the saturation solubility is also a
tial protein adsorption’, we refer to the relevant function of the particle size. This size-dependency
literature and patents [19–22,26,27,32]. In this chap- comes only into effect for particles having a size
ter we only discuss targeting to the MPS cells. below approximately 1 mm — a phenomenon ob-
The uptake of i.v. injected nanoparticles by MPS served in tabletting which leads to an increase of the
cells is a natural phenomenon. The particles are intrinsic dissolution rate of such fine drug particles
recognized as being foreign and phagocytosed by the [34,35]
macrophages, mainly in the liver (60–90%), the The increase in saturation solubility can be ex-
spleen (approximately 1–5%) and to a very small plained by the Kelvin and the Ostwald–Freundlich
extend by the lung macrophages [18]. Because it is a equation [35]. The Kelvin equation describes the
natural process, this targeting has been called ‘natu- vapor pressure over a curved surface of a liquid
ral targeting’ in the literature. Because it happens droplet in gas. The vapor pressure increases with
automatically, this ‘natural targeting’ is considered to increasing curvature, which means decreasing par-
be achievable very easily. Despite this ‘easiness’, no ticle size. This equation, describing the transition of
commercial products are on the market exploiting molecules from a liquid phase (droplet) to a gas
this effect. In addition, it has to be realized that phase, can also be applied to the transition of
uptake of the drug nanoparticles has to be extremely molecules from a solid phase (drug particle) to a
fast to avoid dissolution of the particles. In addition, liquid phase. The vapor pressure is then replaced by
it should be as complete as possible, that means the dissolution pressure. The equilibrium between
preferably 90–100%. Depending on the nature of the dissolving molecules and molecules recrystallizing
surfactants used, sometimes it takes 15–30 min [18]. on particle surfaces (determining the extend of
This might be too long for many nanosuspensions of saturation solubility) is shifted in favour of the
drugs. Therefore, the surface properties of the dissolution process. As a result of this increased
nanosuspensions need to be adjusted this way, so dissolution tendency, the saturation solubility in-
that fast recognition and uptake takes place. This creases. The dependency of the saturation solubility
means the natural uptake by the MPS cells needs to is also described in the Ostwald–Freundlich equation
be further accelerated and enhanced. The surface comparing the solubilities of two particles with
properties need to be changed in a controlled way different diameters [35].
such that preferentially adsorbed proteins can medi- The Noyes–Whitney equation describes the disso-
ate the uptake process by macrophages. This can be lution velocity dc / dt which depends on the surface
done by using HIC in combination with 2-D PAGE. area A and difference (cs 2 cx) /h (cx — equilibrium
¨
R.H. Muller et al. / Advanced Drug Delivery Reviews 47 (2001) 3 – 19 9

concentration in bulk phase, h — diffusional dis- tion process depends on the production parameters
tance). According to the Prandtl equation, the diffu- (homogenization pressure, number of cycles) but is
sional distance h is reduced for small particles [35]. also affected by the hardness of the drug. Of course,
The simultaneous increase in saturation solubility cs the generated structure of the particles needs to be
and decrease in h leads to an increased concentration preserved in case it is not a priori stable, e.g. by
gradient (cs 2 cx) /h, thus enhancing the dissolution lyophilisation or spray-drying. The increase in the
velocity in addition to the surface effect. amorphous fraction leads to a further increase of the
A third pronounced property is the adhesiveness saturation solubility. It is generally known that
generally described for nanoparticles [36]. There is a amorphous drugs have a higher saturation solubility
distinct increase in adhesiveness of ultra fine pow- then the crystalline form.
ders compared to coarse powders. A nice example Another special feature of nanosuspensions is the
from daily life is differently sized sugar. Coarse absence of Ostwald ripening [42], meaning physical
sugar crystals adhere less effectively to a cake or long-term stability as an aqueous suspension [11,43].
cookie compared to iced sugar. This adhesiveness of Ostwald ripening has been described for highly
small drug nanoparticles can be exploited for im- dispersed systems, which means a reduction in size
proved oral delivery of poorly soluble drugs. The of the finest particle fraction and their final dis-
oral administration of drugs in form of drug appearance combined with simultaneously growth of
nanoparticles (NanoCrystals  ) has been reported to the larger particles. Reasons for the Ostwald ripening
have a full range of positive effects [37]: are the different saturation solubilities in the vicinity
of differently sized particles and the concentration
(1) Improving bioavailability gradient existing between them. Molecules from the
(2) Improving dose proportionality higher concentrated solution around very small par-
(3) Reducing fed / fasted variability ticles diffuse to the vicinity of larger particles where
(4) Reducing inter-subject variability a lower concentration is present. This leads to
(5) Enhancing absorbtion rate (both human and supersaturation and drug crystallization, which
animal data) means growth of the larger particles. Simultaneously
the vicinity of smaller particles will be below the
These data reported by the company NanoSystems saturation concentration, thus the new drug will be
have been acquired in vivo in animals but also in dissolved — the fine particles are getting smaller.
humans. Reported increases in bioavailability are This is a continuous process finally leading to the
quite impressive, e.g. for danazole nanosuspension disappearance of the fine particles. The lack of
being 82% compared to only 5% for the commercial Ostwald ripening in nanosuspensions is attributed to
danazole macrosuspension [37,38]. the uniform particle size created by the homogeniza-
An additional feature of nanosuspensions is a tion process. The differences in saturation solubility
potential change in the crystalline structure, which in combination with the a priori low solubility of the
means increasing the amorphous fraction in the poorly soluble drug keeps the concentration differ-
particle or even creating completely amorphous ences sufficiently low to avoid the ripening effect.
particles. It has been described for tabletting where a
sufficiently high punching pressure can transform a
polymorphic compound to a higher energy modi- 4. Surface modification of nanosuspensions
fication. The application of high pressures during the
production of nanosuspensions was found to promote The in vivo performance of nanosuspensions can
the amorphous state, e.g. the drug RMKP22 was be improved further above the effects described in
found amorphous in the nanosuspension [39]. For the previous chapter by controlled surface modi-
NanoCrystals  prepared by a low energy milling fication of the drug nanoparticles. The adhesiveness
process, a completely crystalline structure was re- of the drug nanoparticles is considered to be a major
ported [4,40,41]. The extend of the amorphous factor increasing the bioavailability and reducing
fraction produced in the high pressure homogeniza- variability of absorption and erratic absorption.
10 ¨
R.H. Muller et al. / Advanced Drug Delivery Reviews 47 (2001) 3 – 19

Therefore, the approach was taken to further improve To achieve the desired effect, the surface must not be
the bioavailability performance by modifying the completely covered by the surface modifier. To
particle surface using mucoadhesive polymers. judge if the adsorbed amount is sufficient, the newly
In the case of intravenous administration for created surface needs to be characterized in their
targeting the drug nanoparticles to the MPS, clear- relevant parameters for the modification purpose, e.g.
ance by the macrophages could be further enhance surface hydrophobicity to enhance adsorption of
above the natural clearance effect by modifying the MPS uptake-promoting proteins (e.g. by HIC and
particle surface with surfactants or polymers thereby 2-D PAGE) (Fig. 3).
increasing the adsorption of blood proteins which To increase the adhesion to the gut wall of orally
promote recognition by the MPS. administered nanosuspensions, the particles were
To create a certain surface property, the drug surface modified with polymers, like chitosan and
nanoparticles can be produced using special surfac- carbopol. The particles were directly produced using
tants or polymers as stabilizers in the production high pressure homogenization, afterwards the
process. Where this is not possible (e.g. lack of nanosuspension was incorporated into the polymer
sufficient stabilization effect), the surface modifiers solution.
can be admixed to the nanosuspension. According to The surface-properties of i.v. injected particles
their affinity to the particle surface, they will adsorb determine protein adsorption pattern and organ dis-
and lead to a modification. The degree of modi- tribution resulting from the physical properties of the
fication can be followed by physical measurements, used surfactants. There is a correlation surface
e.g. charge (zeta potential) measurements or hydro- hydrophobicity–protein adsorption pattern in the
phobic interaction chromatography (HIC) analysis. blood–organ distribution [26,27]. Nanosuspensions

Fig. 3. Cuboid nanosuspension particle coated with stabilizer, e.g. surfactant or polymer. The protein adsorption is a function of the
properties of the stabilizer layer (e.g. charge, hydrophobicity). Depending on the composition of the protein adsorption pattern, MPS uptake
can be enhanced or reduced.
¨
R.H. Muller et al. / Advanced Drug Delivery Reviews 47 (2001) 3 – 19 11

for i.v. administration were produced using Tween lem of poor solubility combined with low absorption
80  and poloxamer 188 as stabilizer [44]. Com- are the experimental compounds, bupravaquone and
position of the nanosuspension was tarazepide 1.0%, the recently approved drug, atovaquone
poloxamer 188 1.0% and Tween 80  0.5%, while (Wellvone  ), used for the treatment of opportunistic
ten homogenization cycles with 1500 bar were Pneumocystis carinii infections in HIV patients.
applied. The mean particle diameter was 400 nm, the Besides this indication, both drugs are also very
LD 100% was 3.5 mm. effective for the treatment of leishmaniasis, but
require an extremely high dose. Furthermore, ato-
vaquone was recently approved in combination with
5. Biological properties of nanosuspensions and proguanil (Malarone  ) for therapy of non-compli-
medical indications cated P. falciparum malaria [45].
Atovaquone is given orally three times 750 mg
5.1. Oral administration of nanosuspensions daily, reflecting a high dose as an antibiotic. The
main reason for the high dose is the low absorption
First choice of application is oral administration. — only 10–15% of this poorly soluble drug. A way
When a drug is given orally, the bioavailability and to improve bioavailability of atovaquone or bup-
finally its efficacy depends on the solubility and ravaquone would be to increase the absorption rate
absorption in the gastrointestinal tract. In vitro, by formulating as a nanosuspension. Oral administra-
highly active compounds have failed in the past tion of nanosuspensions can overcome this problem
because their poor solubility has limited in vivo because of the high adhesiveness of drug particles
absorption and did not lead to effective therapeutic sticking on biological surfaces — here the epithelial
concentrations. Simple examples reflecting this prob- gut wall — and prolonging the absorption time.
When formulating atovaquone as a nanosuspension
(Fig. 4) and given orally to Leishmania-infected
mice, the parameter for increased absorption is
related to the infectivity score of each animal or it is
related to the reduction of the parasite load in the
liver. In comparison to Wellvone  -treated mice,
containing a micronized drug, nanosuspensions of
atovaquone at equivalent doses reduced infectivity
from 40 to 15% at a reduced concentration of only
7.5 mg / kg [46]. These results reflect the potency of
this technique, reducing drug load from 22.5 mg / kg
(Wellvone  ) to 7.5 mg / kg, but increasing activity
2.5-fold at the same time. Another example for the
effectiveness of nanosuspensions for improving bio-
availability is the application of amphotericin B.
When amphotericin B is given orally in approved
drugs for the treatment of GIT mycosis, the drug is
not or only to a minor extent absorbed. Formulating
this antimycotic and leishmanicidal drug as a
nanosuspension and administering it orally, the num-
ber of L. donovani parasites in vivo was reduced
significantly, indicating a high uptake of the drug in
the gastrointestinal tract. When given amphotericin B
nanosuspensions orally, the infectivity score (IS) was
Fig. 4. Transmission electron micrographs of a atovaquone determined at 25% (control5100% parasite growth).
nanosuspension (medium size: 468 nm). In comparison to i.v. (!) administered liposomes
12 ¨
R.H. Muller et al. / Advanced Drug Delivery Reviews 47 (2001) 3 – 19

entrapping amphothericin B, like Ambisome  (IS5 technology is considered as a suitable new colon
40%) and commercial crude amphotericin B (mi- delivery systems for the treatment of colon cancer,
cronized drug particles) with no leishmanicidal ef- helminth infections, gastrointestinal inflammation or
fect, these results confirm the potency for nanosus- GIT associated diseases like sprue (zoeliaki).
pensions to increase bioavailability. From the back-
ground of finding an oral formulation for Leish- 5.2. Parenteral nanosuspensions — intravenous
mania-infected patients, nanosuspensions demon- administration route
strate a new pathway for future therapy.
The adhesiveness of nanosuspensions can not only Infections like tuberculosis, listeriosis, leish-
be used to improve bioavailability, it is also a smart maniasis, and toxoplasmosis are caused by parasites
way for drug targeting of parasites persisting in the residing the macrophages of the MPS, thus being
gastrointestinal tract. Cryptosporidium parvum has relatively easily accessible by i.v. injected particles.
attracted renewed interest, since it has been identified The i.v. injected particles are heavily and quickly
as the main pathogen causing severe diarrhoea in taken up by the MPS cells in case they absorb uptake
immunosuppressant HIV patients. In the US and promoting proteins like apolipoproteins. Fig. 5 shows
Europe, approximately 10–15% of AIDS patients are a protein adsorption pattern of hydrophobic poly-
suffering from this opportunistic pathogen, and today styrene particles which are being cleared in vivo very
there is still no effective therapy or clinically useful fast by liver and spleen macrophages.
drug. Formulating drugs to combat C. parvum must However, some parasite do also reside in the brain
cover two main aspects. First, targeting the drug to (CNS). Especially, in trypanosomiasis and toxoplas-
the pathogen which is located in the epithelial mosis, the brain-localized parasite mostly leads to
membranes of the gut wall, and secondly, increasing relapsing infections if not cured. Therefore, it would
the time for the drug in the GIT to prolong the be of importance to target drug nanoparticles via
pharmacological window with regard to the fast surface modification to the brain.
washing-out during diarrhoea. Bupravaquone Kreuter et al. [49] reported successful targeting of
nanosuspensions provide some advantages, and in the peptide, dalargin, to the brain using Tween 80 
TCR-alpha-deficient mice infected with C. parvum surface modified polyisobutylcyanoacrylates
oocysts, the significance of the nanosuspensions is nanoparticles [49]. The mechanism was identified as
documented. In comparison to micronized drug being ApoE-mediated and was confirmed for the
powder, the infectivity score was reduced from 2.0 nanosuspensions discussed here. Fig. 5 shows a 2-D
(negative control group, untreated) to 1.47 for mi- PAGE gel of particles having an affinity to the brain
cronized bupravaquone and even to 1.02 for equiva- and transporting the drug. The Tween surface-modi-
lent nanosuspensions [47]. With respect to the fied nanoparticles where analyzed by 2-D PAGE and
pathophysiological situation in the infected GIT, showed also the characteristic ApoE spot, that is
nanosuspensions were incorporated in a mucoadhe- clearly visible (arrow). At present, surface-modified
sive chitosan matrix (0.5%) and the effectiveness melarsenoxide particles (Tween 80  coated) are
was retested. Infectivity scores of 0.68 in the cecum under investigation in vivo to treat murine
and 0.49 in the ileum documented the improved trypanosomiasis phase II, also called sleeping sick-
action of the mucoadhesive nanosuspension delivery ness. The clinically used drug, melarsoprol
system [48]. Formulating poorly soluble drugs for (Arsobal  ), has only 3–5% of the administered dose
the treatment of Cryptosporidium infections or other detectable in the brain fluid. An improved brain
GIT infections, like helminths, will be an important delivery formulation would allow to reduce the
strategy in the future. therapeutic dose and toxic side-effects in parallel.
In summary, oral administration of nanosuspen- These results would also have an impact on other
sions is a drug delivery strategy, not only to improve CNS-associated diseases, like trypanosomiasis, but
bioavailability, but also to target gastrointestinal also toxoplasmosis and cerebral malaria.
bacterial and parasitic infections. We did not discuss Administration of drugs via the parenteral route is
other fields of interest here, but the nanosuspension critical, i.e. usually accompanied by physical and
¨
R.H. Muller et al. / Advanced Drug Delivery Reviews 47 (2001) 3 – 19 13

Fig. 5. (a) 2-D PAGE gel of Tween 80 modified polyisobutylcyanoacrylate nanoparticles (right) versus unmodified nanoparticles (left)
¨ [30]). Tween 80 coated PBCA particles showing preferentially adsorption of ApoE (arrow). (b) Semi-quantitative 2-D
(modified after Luck
¨ [30]).
PAGE analysis of polybutylcyanoacrylate nanoparticles after incubation in citrate plasma (gels from (a), modified after Luck

biological problems, like production under aseptic to target drugs to the desired site of action, even
conditions, a sophisticated protocol for safety issues, when they show physical problems like poor solu-
and, last but not least, biological problems, such as bility. Paclitaxel reflects such a problem [6]. Because
endotoxins, allergic reactions, and inconvenience for of low solubility, it has been formulated for i.v. use
the patient. with Cremophor EL. The main disadvantage is the
On the other hand, parental administration can not high risk of allergic reaction during infusion, which
be avoided in every cases. Sometimes it is favoured has been often reported in the past. However,
14 ¨
R.H. Muller et al. / Advanced Drug Delivery Reviews 47 (2001) 3 – 19

parenteral administration also has its advantages. A pora sphaerica, which has been used in the past in
simple but effective drug targeting principle is to experimental biology for cell cycle synchronization.
deliver the drug to infected macrophages. A whole Distinct differences in the inhibition of DNA-poly-
range of human pathogenic microorganisms persist merase type I between mammalian and parasite cells,
intracellularly in macrophages, like Mycobacterium revealed new interest in this unique structure as a
tuberculosis, Listeria monogyna, Leishmania sp. new antiparasitic lead. Besides its activity, the use of
Application of drugs as particles will exploit the aphidicolin is limited, because it is extremely fast
natural endocytotic mechanism of these mononuclear cleaved when given orally and its poor solubility has
cells of grabbing all particular objects, mostly cell stopped research in the past. By formulating aphidi-
fragments, macromolecular proteins, or microorga- colin as nanosuspension, these main disadvantages
nisms invading the body. Colloidal drug carriers can be minimized. In in vitro test systems with
systems have attracted interest for many years in infected RAW macrophages, the uptake of the
their ability to incorporate different drugs in mi- nanosuspensions improved the EC 50 value from 200
croparticles, liposomes, polymer particles, mi- to 40 ng / ml in comparison to DMSO-dissolved drug.
croemulsions thereby exploiting this natural uptake The endocytic uptake of aphidicolin particles is
mechanism (Fig. 6). Even for the treatment of demonstrated microscopically (Fig. 7). The dark
Leishmania infections these above-mentioned deliv- spots display macrophages filled completely with
ery systems have been investigate intensively. How- nanosuspensions (Fig. 7, middle), as visualised by
ever, even today no formulation technique has been transmission electron microscopy (TEM) (Fig. 7,
approved and launched to the market. For liposomes, right). In comparison, Fig. 7 (left) shows untreated
the main problems are high costs in production or the control of RAW macrophages seeded on Petri plates.
absence of a cost-effective and simple scaling-up Compounds also approved for peroral use only
technology (see Section 6). Nanosuspensions may show a broad potential for i.v. or i.m. administration,
contribute to solve these problems. when formulated as nanosuspension. For example,
Aphidicolin is a fungal metabolite from Nigros- atovaquone is only for oral use, due to the fact that

Fig. 6. Uptake of drugs and intracellular trafficking: free drugs (h) versus nanosuspensions (j). (I) Diffusion, (II) endocytosis, (IIa)
releases of nanoparticles and movement from vacuoles to infected lysosomes (parasite indicated by oval shapes), (IIb) fusion of vacuole with
lysosome and intracellular particle realease.
¨
R.H. Muller et al. / Advanced Drug Delivery Reviews 47 (2001) 3 – 19 15

Fig. 7. Targeting of nanosuspensions to RAW-macrophages. Left: untreated RAW macrophages (TEM); middle: RAW-macrophages
incubated with 5% (V/ V) aphidicolin nanosuspension (circles) (light microscope); right: Leishmania-infected RAW-macrophages incubated
with 5% (v / v) aphidicolin nanosuspension (TEM). LM, lysosome membrane; NP, nanoparticle; P, parasite; VNP, nanoparticles in vacuole.

the poor solubility of micronized particles does not replacement for one patient resulting in a limited
allow i.v. application (capillary blocking). Formula- quality of life compared to many low-cost opera-
tion of atovaquone as a nanosuspension for i.v. use tions, such as hip replacements, resulting in a high
to treat murine toxoplasmosis showed a significant number of survival years with a high quality of life.
reduction of Toxoplasma gondii at a dose of 0.3 Therefore, the cost factor for a technology is getting
mg / kg in comparison to 30 mg / kg when given more and more important.
orally. The importance of the availability of large scale
production facility becomes evident when looking at
polymeric drug nanoparticles. For more than 30
6. Large-scale production of nanosuspensions years, intensive research has been performed in this
area, but this delivery system does not really exist on
The possibility and ability for large-scale pro- the market. There is one product, Abdoscan  by
duction of a delivery system or a dosage form is the Nycomed; however, this is not a product for chronic
essential prerequisite for its introduction to the treatment of diseases but a diagnostic agent for
pharmaceutical market. It is pointless to have a very single administration (meaning a product with less
neat delivery system but no possibility to produce it problems regarding regulatory acceptance). Depend-
on a large scale to supply the market. In addition, the ing on the type of nanoparticles, there are many
production technology needs to be low-cost to different reasons for the lack of products on the
consider the financial restrictions of the health market. However, from our point of view, a general
systems, even in relatively rich countries. A technol- reason is the lack of a production method on a large
ogy which is feasible but extremely expensive will scale yielding a product of acceptable quality by the
not find its way to the market, especially when regulatory authorities. An important point is that it is
considering the recent discussions on the cost-to- not sufficient at all to have available a production
patient benefit ratios. In the mid-1980s in the UK, line which can produce on large scale. As a pre-
calculations were made to multiply the medium requisite for acceptance by the regulatory authorities,
survival time of patients in years with a life quality this production line needs to be suitable for qualifica-
parameter. The resulting figure was compared to the tion and validation. In addition, the technology needs
cost per operation. This means it was not considered to be cost-effective. There are still problems to meet
economical to perform a very cost-intensive heart all these requirements for the production of poly-
16 ¨
R.H. Muller et al. / Advanced Drug Delivery Reviews 47 (2001) 3 – 19

meric nanoparticles. In contrast to this drug, Instead of passing, say three times through one
nanoparticles can easily be produced on a large scale homogenizer, three homogenizers can be placed in
using the high pressure homogenization technique series to produce the product in a continuous mode
[50,35] already applied in various areas for large- (Fig. 8, upper). Using three homogenizers is possible
scale production. because the homogenizers are off-the-shelf equip-
High pressure homogenizers are available with ment and are in addition of low-cost. A suitable
different capacities from a few hundred to a few homogenizer is the Rannie having a capacity of 1000
thousand liter per hour. The high pressure homogeni- l at a maximum pressure of 1500 bar. To place
zation technology is well established in many differ- homogenizers in series is less economical where a
ent areas of applications ranging from food to higher number of cycles is required. For example,
lubricant production. In pharmaceutics, emulsions for with five cycles and simultaneously a relatively small
parenteral nutrition are produced by high pressure batch size of, say 1000 l, a low-cost solution would
homogenisation, which means production lines ac- be to apply a discontinuous mode of one homogen-
cepted for parenteral products are available. izer. The suspension will be passed from the feeding
For the production of a nanosuspension, the container through the homogenizer to the product
macrosuspension needs to be passed somewhere container, after this first cycle the suspension will be
between three times up to five / ten or a maximum of passed back and so on, so that five passes could be
20 times through a high pressure homogeniser. performed within 5 h of production (Fig. 8, middle).

Fig. 8. Scheme of production lines. Production of nanosuspensions applying three passes (cycles) by using three homogenizers (H) in series
(5discontinous production, upper), applying five to ten cycles by passing the product multiple times through one or two homogenizers
(5discontinous production, middle and lower; FC, feeding container; PC, product container).
¨
R.H. Muller et al. / Advanced Drug Delivery Reviews 47 (2001) 3 – 19 17

Where ten cycles are required, one could place two soluble drugs. Any drug can be transformed to drug
homogenizers between the containers to keep the nanoparticles leading to an increase in saturation
production time constant at 5 h (Fig. 8, lower). solubility, dissolution velocity, and providing the
Applying pearl milling for the production of drug general feature of an increased adhesiveness to
nanoparticles requires milling times from hours to a surfaces. Surface modification of the drug nanocryst-
few days. Milling over a few days bears the risk of als can further increase the benefits, e.g. by produc-
microbiological problems, especially when perform- ing mucoadhesive nanosuspensions for oral applica-
ing the milling at room temperature or having tion or surface-modified site-specific nanoparticles
dispersion media providing nutrition to bacteria. for intravenous injection (e. g. targeting to the brain,
Production times of generally a few hours are, bone marrow etc.). A further advantage is that this
therefore, a basic advantage of producing drug innovative nanosuspension technology can be com-
nanoparticles by high pressure homogenization. In bined with traditional dosage forms, e.g. incorporat-
addition, high pressure homogenization has a steriliz- ing drug nanoparticles into pellets or tablets for oral
ing effect. It is a technique applied, for example, for delivery. An outstanding feature of the technology is
the rupture of cells including bacteria, thus increas- its simplicity. The more simple the system is, the
ing the microbiological product quality. higher are the chances to launch products on the
Another important point is the contamination of market. The success of drug nanoparticles, especially
the product by erosion from the production equip- the third generation product nanosuspensions, will be
ment. Pearl mills bear the risk of erosion of material demonstrated by the number of products on the
from the pearls — contamination of the product by market in the near future.
pearl material has been reported [51]. Even for oral
administration, eroded particulate material might
represent a problem. The formation of microparticles
References
from the pearls could occur, such microparticles
might interact with the M cells of the Peyer’s patches
¨
[1] R.H. Muller, ¨
S. Benita, B. Bohm (Eds.), Emulsions and
with unknown consequences on the immune system. Nanosuspensions for the Formulation of Poorly Soluble
Particle uptake by the M cells is also considered as Drugs, Medpharm Scientific, Stuttgart, 1998.
one approach for oral vaccination, therefore, distur- ¨
[2] R.H. Muller, K. Peters, R. Becker, B. Kruss, Nanosuspen-
bances of the immune system by continuous ad- sions for the i.v. administration of poorly soluble drugs —
ministration of non-biodegradable microparticles stability during sterilization and long-term storage, Int.
Symp. Control. Release Bioact. Mater. 22 (1995) 574–575.
cannot be excluded. Such contamination does not ¨
[3] R.H. Muller, R. Becker, B, Kruss, K. Peters, 1998. Pharma-
exist in products produced by high pressure homoge- ceutical nanosuspensions for medicament administration as
nization. Possible contamination of the nanosuspen- system of increased saturation solubility and rate of solution,
sions could occur from metal ions coming off from US Patent No. 5858410.
the wall of the homogenizer. The metal contamina- [4] G.G. Liversidge, K.C. Cundy, J. Bishop, D. Czekai, 1991.
Surface modified drug nanoparticles, US Patent No.
tion was analyzed in nanosuspensions produced 5145684.
under extreme conditions, i.e. 20 cycles at a maxi- [5] M. List, 1987. Hydrosole, eine intravenose¨ Arzneiform zur
mum pressure of 1500 bar. The most dominant ion in Herstellung von Injektionen und Infusionen in Wasser
steel is iron, thus this metal was analyzed in the ¨
schwer loslicher Wirkstoffe. Dissertation, University of
nanosuspensions and was found to be below 1 ppm Basel, Switzerland.
[6] P. Gaßmann, 1990. Herstellen und Stabilisieren von Hydro-
[52], i.e. an uncritical level. ¨
solen zur intravenosen Applikation, PhD thesis, University of
Basel, Switzerland.
[7] P. Gaßmann, M. List, A. Schweitzer, H. Sucker, Hydrosols
7. Perspectives — Alternatives for the parenteral application of poorly water
soluble drugs, Eur. J. Pharm. Biopharm. 40 (1994) 64–72.
[8] P. Bernoulli, in: S. Sucker, H. Fuchs, P. Speiser (Eds.),
Drug nanocrystals — independent of their way of
Pharmazeutische Technologie, Georg Thieme, Stuttgart,
production — represent a technology to overcome 1978, pp. 101–102.
solubility problems and bioavailability problems of ¨
[9] B. Bohm, 1999. Herstellung und Charakerisierung von
drugs which can be generally applied to all poorly Nanosuspensionen als neue Arzneiform fur ¨ Arzneistoffe mit
18 ¨
R.H. Muller et al. / Advanced Drug Delivery Reviews 47 (2001) 3 – 19

¨
geringer Bioverfugbarkeit, PhD thesis, Freie Universitat ¨ [25] T. Blunk, E. Mak, R.H. Muller, ¨ Characterization of colloidal
Berlin, Germany drug carriers: determination of surface hydrophobicity by
[10] K. Peters, S. Leitzke, J.E. Diederichs, K. Borner, H. Hahn, hydrophobic interaction chromatography, Pharm. Ind. 55
¨
R.H. Muller, S. Ehlers, 2000. Preparation of a clofazimine (1993) 612–615.
nanosuspension for intravenous use and evaluation of its [26] K.H. Wallis, R.H. Muller, ¨ Determination of the surface
therapeutic efficacy in murine Mycobacterium avium in- hydrophobicity of colloidal dispersions by mini-hydrophobic
fection, J. Antimicrob. Chemother., submitted. interaction chromatography, Pharm. Ind. 55 (1993) 1124–
[11] K. Peters, 1999. Nanosuspensionen-ein neues For- 1128.
mulierungsprinzip fur ¨ schwerlosliche
¨ Arzneistoffe, PhD ¨
[27] R.H. Muller, Differential opsonization: A new approach for
thesis, Freie Universitat ¨ Berlin, Germany. the targeting of colloidal drug carriers, Arch. Pharm. 322
[12] C. Jacobs, 2000. PhD thesis, Freie Universitat ¨ Berlin, (1989) 700.
Germany (in preparation). ¨
[28] R.H. Muller, ¨
M. Luck, K. Seehof, M. Kresse, J. Kreuter, S.
[13] K.P. Krause, 2000. PhD thesis, Freie Universitat ¨ Berlin, Harnisch, 2000. Adsorbed plasma proteins as a mean for site
Germany (in preparation). specific delivery of colloidal carriers, Adv. Drug Deliv. Rev.
[14] ¨
B.W. Muller, ¨
R.H. Muller, Particle size analysis of latex (submitted).
suspensions and microemulsions by photon correlation spec- [29] T. Blunk, 1994. Plasmaproteinadsorption auf kolloidalen
troscopy, J. Pharm. Sci. 73 (1984) 915–918. ¨
Arzneistofftragern, PhD thesis, Christian-Albrechts-Univer-
[15] P. Ney, Zetapotentiale und Flotierbarkeit von Mineralien, ¨ zu Kiel, Germany.
sitat
Springer, Wien, 1974. [30] M. Luck,¨ 1997. Plasmaproteinadsorption als moglicher ¨
[16] R.H. Muller, ¨ Zetapotential und Partikelladung-Kurze ¨
Schlusselfaktor ¨ eine kontrollierte Arzneistoffapplikation
fur
Theorie, praktische Meßdurchfuhrung, ¨ Dateninterpretation, ¨
mit partikularen ¨
Tragern, PhD thesis, Freie Universitat ¨
Wissenschaftliche, Stuttgart, 1996. Berlin, Germany.
[17] C.J. Van Oss, D.R. Absolom, H.W. Neumann, Interaction of [31] K. Seehof, M. Kresse, R.H. Muller, ¨ Plasma protein ad-
phagocytes with other blood cells and with pathogenic and sorption onto model magnetite dispersions: comparison
nonpathogenic microbes, Ann. NY Acad. Sci. 416 (1984) between in vitro and ex vivo results by 2D-PAGE, in:
332–350. Proceedings of the 3rd World Meeting of APV/APGI, Berlin,
[18] ¨
R.H. Muller, Colloidal Carriers for Controlled Drug Delivery 2000, submitted.
and Targeting. Modification, Characterization and in vivo [32] K. Seehof, PhD thesis, Freie Universitat ¨ Berlin (in prepara-
Distribution, CRC Press, Boca Raton, FL, 1991. tion).
[19] R.H. Muller,¨ S. Heinemann, Surface modelling of mi- ¨
[33] R.H. Muller, ¨
M. Luck, ¨
J.Kreuter, 1997. Arzneistofftragerpar-
croparticles as parenteral systems with high tissue affinity, ¨ die gewebsspezifische Arzneistoffapplikation, Ger-
tikel fur
in: R. Gurny, H.E. Junginger (Eds.), Bioadhesion. Possi- man Patent Application No. 197 45 950.1.
bilities and Future Trends, Wissenschaftliche, Stuttgart, 1989, [34] C. Nystrom, ¨ Dissolution properties of soluble drugs: Theo-
pp. 202–214. retical background and possibilities to improve the dissolu-
[20] T. Blunk, D.F. Hochstrasser, J.-C. Sanchez, B.W. Muller, ¨ tion behavior, in: R.H. Muller, ¨ ¨
S. Benita, B. Bohm (Eds.),
¨
R.H. Muller, Colloidal carriers for intravenous drug target- Emulsions and Nanosuspensions for the Formulation of
ing: Plasma protein adsorption patterns on surface-modified Poorly Soluble Drugs, Medpharm Scientific, Stuttgart, 1998,
latex particles evaluated by two-dimensional polyacrylamide pp. 143–147.
gel electrophoresis, Electrophoresis 14 (1993) 1382–1387. [35] R.H. Muller,¨ B.H.L. Bohm, ¨ Nanosuspensions, in: R.H.
[21] T. Blunk, M. Luck, ¨ ¨ D.F. Hochstrasser, J.-C.
A. Calvor, ¨
Muller, S. Benita, B. Bohm¨ (Eds.), Emulsions and Nanosus-
Sanchez, B.W. Muller, ¨ ¨
R.H. Muller, Kinetics of plasma pensions for the Formulation of Poorly Soluble Drugs,
protein adsorption on model particles for controlled drug Medpharm Scientific, Stuttgart, 1998, pp. 149–173.
delivery and drug targeting, Eur. J. Pharm. Biopharm. 42 [36] D. Duchene, G. Ponchel, Bioadhesion of solid oral dosage
(1996) 262–268. forms, why and how?, Eur. J. Pharm. Biopharm. 44 (1997)
[22] M. Luck,¨ ¨
W. Schroder, S. Harnisch, K. Thode, T. Blunk, 15–23.
B.-R. Paulke, M. Kresse, R.H. Muller, ¨ Identification of [37] G.C. Liversidge, Drug nanocrystals for improved drug
plasma proteins facilitated by enrichment on particulate delivery, in: Int. Symp. Control. Release Bioact. Mater,
surfaces: Analysis by two-dimensional electrophoresis and Workshop on Particulate Drug Delivery Systems, Vol. 23,
N-terminal microsequencing, Electrophoresis 18 (1997) 1996.
2961–2967. ¨
[38] R.H. Muller, Nanosuspensionen-eine neue Formulierung fur ¨
[23] ¨
M. Luck, ¨
B.-R. Paulke, W. Schroder, ¨
T. Blunk, R.H. Muller, ¨
schwerlosliche Arzneistoffe, in: R.H. Muller,¨ G.E. Hilde-
Analysis of plasma protein adsorption on polymeric brand (Eds.), Pharmazeutische Technologie: Moderne Ar-
nanoparticles with different surface characteristics, J. zneiformen, Lehrbuch fur ¨ Studierende der Pharmazie-Nach-
Biomed. Mater. Res. 1 (1997) 478–485. schlagewerk fur ¨ Apotheker in Offizin, Krankenhaus und
[24] C.J. Van Oss, C.F. Gillman, A.W. Neumann (Eds.), Forschung, 2nd Edition, Wissenschaftliche, Stuttgart, 1998,
Phagocytotic Engulfment and Cell Adhesiveness as Cellular pp. 393–399.
Surface Phenomenon, Marcel Dekker, New York, 1975. ¨
[39] M.J. Grau, R.H. Muller, Increase of dissolution velocity and
¨
R.H. Muller et al. / Advanced Drug Delivery Reviews 47 (2001) 3 – 19 19

solubility of poorly soluble drugs by formulation as nanosus- [47] C. Jacobs, O. Kayser, Mucoadhesive Nanosuspension for the
pension, in: Proceedings 2nd World Meeting APGI /APV, formulation of Bupraquone, in: Proceedings of the 3rd World
Paris, 1998, pp. 623–624. Meeting of APV/APGI, Berlin, 2000, in press.
[40] G.C. Liversidge, P. Conzentino, Drug particle size reduction [48] O. Kayser, C. Jacobs, J.S. Keithly, 2000. Anticryptosporidial
for decreasing gastric irritancy and enhancing absorption of activity of bupravaquone and improving its in vivo efficacy
naproxen in rats, Int. J. Pharm. 125 (1995) 309–313. by the formulation as a mucoadhesive nanosuspension, Int.
[41] G.C. Liversidge, K.C. Cundy, Particle size reduction for Symp. Control. Release Bioact. Mater. (in press).
improvement of oral bioavailability of hydrophobic drugs. I. [49] J. Kreuter, V.E. Petrov, D.A. Kharkevich, R.N. Alyautdin,
Absolute oral bioavailability of nanocrystalline danazole in Influence of the type of surfactant on the analgesic effects
beagle dogs, Int. J. Pharm. 127 (1995) 91–97. induced by the peptide dalargin after 1st delivery across the
[42] E.A. Rawlins, Bentley’s Textbook of Pharmaceutics, 8th blood–brain barrier using surfactant coated nanoparticles, J.
Edition, Bailliere Tindall, London, 1982. Control. Release 49 (1997) 81–87.
[43] ¨
K. Peters, R.H. Muller, Nanosuspensions for the oral appli- ¨
[50] R.H. Muller, S. Gohla, A. Dingler, T. Schneppe, Large scale
cation of poorly soluble drugs, in: Proceeding European production of solid lipid nanoparticles (SLNE) and nanosus-
Symposium on Formulation of Poorly-available Drugs for pensions (DissoCubesE), in: D. Wise (Ed.), Handbook of
Oral Administration, APGI, Paris, 1996, pp. 330–333. Pharmaceutical Controlled Release Technology, 2000, in
[44] ¨
C. Jacobs, O. Kayser, R.H. Muller, Nanosuspensions as a press.
new approach for the formulation for the poorly soluble drug [51] S. Buchmann, W. Fischli, F.P. Thiel, R. Alex, Aqueous
tarazepide, Int. J. Pharm. 196 (2000) 161–164. microsuspension, an alternative intravenous formulation for
[45] S. Looareesuwan, J.D. Chulay, C.J. Canfield, D.B. Hutchin- animals studies, in: Proceedings 42nd Annual Congress APV,
son, Atovaquone and proguanil hydrochloride followed by Mainz, Germany, 1996, p. 124.
primaquine for treatment of Plasmodium vivax malaria in [52] K.P. Krause, O. Kayser, K. Mader,¨ ¨
R. Gust, R.H. Muller,
Thailand, Trans. R. Soc. Trop. Med. Hyg. 93 (1999) 637– Heavy metal contamination of nanosuspensions produced by
640. high-pressure homogenisation, Int. J. Pharm. 196 (2000)
[46] O. Kayser, R.W. Water, S.J. Upton, C. Jacobs, J.S. Keithly, 169–172.
2000. Anticryptosporidial activity of bupravaquone and
development of an mucoadhesive drug delivery system for
improving its in vivo efficacy, J. Drug Target. (submitted).

You might also like