Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

REVIEWS

NANOSUSPENSIONS IN DRUG
DELIVERY
Barrett E. Rabinow
Abstract | A surprisingly large proportion of new drug candidates emerging from drug
discovery programmes are water insoluble, and therefore poorly bioavailable, leading to
abandoned development efforts. These so-called ‘brickdust’ candidates can now be rescued
by formulating them into crystalline nanosuspensions. In the process of overcoming issues
involving solubility, additional pharmacokinetic benefits of the drugs so formulated have come to
be appreciated. As such, insolubility issues of the past have provoked a paradigm change,
which now offers novel solutions for innovative drugs of the future.

SOLID LIPID NANOPARTICLES


Nanosuspensions of drugs are sub-micron colloidal dissolve them, and by maintaining the drug in a pre-
Sub-micron-sized spherical lipid dispersions of pure particles of drug, which are stabilized ferred crystalline state of size sufficiently small for
carriers of lipid-soluble drug by surfactants1. For the purpose of this review, they are pharmaceutical acceptability.
molecules. They are typically distinguished from nanoparticles, which are poly- In addition, utilization of the dense, solid state confers
formed by heating an aqueous
meric colloidal carriers of drugs2, and from SOLID LIPID an additional advantage of higher mass per volume
lipid mixture above the melting 3
point of the lipid, adding drug, NANOPARTICLES , which are lipidic carriers of drugs. loading. This is crucial when high dosing is required.
homogenizing, then cooling to Activity in this area has exploded recently because of the Such cases often fail with approaches involving molecular
freeze the drug within the the need for solubility enhancement. complexation (for example, CYCLODEXTRINS) because of
solid lipid spheres. This has become a problem during the past ten years the high molar ratio of complexing excipient that must
HIGH-THROUGHPUT
because of the new paradigm in HIGH-THROUGHPUT be used. A related benefit of high loading is reduced
SCREENING SCREENING of new drug candidates, which emphasizes the administration volume, which is crucial for low-volume
An automated instrumental importance of good fit with target-receptor geometry. intramuscular and ophthalmic applications6,7.
process for detecting the Candidates emerging from these screens have high Conventional approaches often attempt to solubilize
binding of tens of thousands of
molecular mass and high LOG P, both of which are factors insoluble drugs with the use of excessive amounts of co-
drug candidates to an isolated
protein receptor target that that contribute to insolubility4. solvents, but this poses toxicity problems. Subsequently,
mediates a disease process, a related issue arises during drug development when
thereby identifying worthwhile When can nanosuspensions help? the safety of the agent must be studied in animals at
leads for development. Nanosuspensions can be used for compounds that are many multiples of its intended dose in man. The need
WATER INSOLUBLE but which are soluble in oil (high log P), to administer very large doses of drugs must then be
although other lipidic systems, such as liposomes and accomplished without the interference of toxic side
emulsions, can be used to formulate these compounds effects caused by co-solvents8,9. Conducting these
as well. Nanosuspensions are the preferred solution, studies might now be possible by using nanosuspen-
however, when no other approach will work. In contrast sions that dispense with solvents. As a result of these
Senior Director, Strategic
Technical Development, with lipidic systems, nanosuspensions can be used to advantages, drug candidates once abandoned could
Baxter Healthcare successfully formulate compounds that are insoluble in now be rescued (FIG. 1).
Corporation, Baxter both water and oils. This can be the case when the crystal The above-described requirements have driven the
Technology Park, Round energy is high, indicated by a high melting point, which development of nanosuspension technology, which has
Lake, Illinois 60073, USA.
e-mail: barrett_rabinow@
reduces the tendency of the crystal to dissolve, regard- subsequently revealed secondary benefits that are now
baxter.com less of the solvent5. Nanosuspensions overcome delivery beginning to be realized (TABLE 1). For example, the par-
doi:10.1038/nrd1494 issues for these compounds by obviating the need to ticulate nature of the dosage form might offer alternative

NATURE REVIEWS | DRUG DISCOVERY VOLUME 3 | SEPTEMBER 2004 | 7 8 5


REVIEWS

Increasing drug polarity

Cosolvents
pH adjustment Micellar dispersions
Co-solvents Inclusion complexes Nanosuspensions
Salt formation Emulsions
Other lipid systems

Yes No
Suitable
molecular shape?

Dose?
Yes
High Low Low

Melting point?
High

Low High
Water soluble? Log P?
No

Can a salt be made?


Yes No

Start

Figure 1 | Decision tree for selection of formulation approach. The easiest applicable approaches are utilized. If a salt can be
made, or solubility increased by simple pH adjustment, these are done preferentially. If the drug is not particularly insoluble, co-solvents
are tried next. If there is adequate solubility in lipidic systems, then micelles, emulsions and so on are tried. Inclusion complexes, as
with cyclodextrins, can be considered. For the most intractable compounds — those with high Log P, high melting point and high
dose — nanosuspensions are used.

LOG P pharmacokinetic profiles in intravenous delivery that forces with other water molecules when located at a free
Log of the octanol–water can offer less toxic and more efficacious regimens. By surface. The system prefers to reduce this increase in
partition coefficient, which is a decreasing the particle size of the solid form of the drug, surface area by either dissolving incipient crystalline
measure of a drug’s lipophilicity.
the dissolution rate is increased, thereby addressing a nuclei, in the case of precipitation, or by agglomerating
Defined as the ratio of
un-ionized drug distributed number of issues related to poor oral BIOAVAILABILITY. The small particles, regardless of their formation mecha-
between the octanol and water solid state of the drug offers solutions to issues of chem- nism. This tendency is resisted by the formulator
phases at equilibrium. Higher ical stability, and the small size confers increased physical through the addition of surface-active agents, which
values imply greater lipophilicity. stability with respect to sedimentation. Although manu- reduce the γs/l and therefore the free energy of the system
WATER INSOLUBLE
facturing complexity is increased, it is offset by utilizing (FIG. 2). These agents confer immediate protection and
Less than 0.1 mg per ml unit operations that are already being deployed in other are more effective when present at the time of creation
solubility in water. commercialized products. To best achieve the end bene- of the new, fresh surface than if added afterwards.
fits of nanosuspension dosage forms, a firm understand- By virtue of their complementary properties, surfac-
CYCLODEXTRINS
5–8mer of cyclic linked amylose ing of formulation theory as well as manufacturing tants of two classes are utilized: charged or ionic sur-
or glucan molecules that forms practices is essential. factants, which effect an electrostatic repulsion among
a hydrophobic interior to the particles; and non-ionic polymers, which confer a
accommodate an insoluble Formulation theory steric repulsion — that is, they resist compression. The
compound, and a hydrophilic
exterior to solubilize in water.
Nanoparticles can be formed by building particles up mechanics of these surfactant-stabilized suspensions
from the molecular state, as in precipitation, or by can be considered from the perspective of potential
BIOAVAILABILITY breaking larger micron-sized particles down, as in energy (BOX 1).
A measure of the rate and milling. In either case, a new surface area, ∆A, is formed, If the particles approach each other too closely, they
extent of drug absorption from
an administered dose, expressed
which necessitates a free-energy (∆G) cost as defined by will agglomerate. This must be prevented to ensure a
as a ratio to an intravenously ∆G = γs/l •∆A, in which γs/l is the interfacial tension. This stable system. Energetically, this requires the placement
administered dose. arises because water molecules incur fewer attractive of a sufficiently high energy barrier at relatively long

786 | SEPTEMBER 2004 | VOLUME 3 www.nature.com/reviews/drugdisc


REVIEWS

Table 1 | Benefits of nanosuspensions


Physicochemical characteristic Potential benefits
Increased drug amount in dosage form Intravenous: reduced toxicity, increased efficacy
without harsh vehicles (extreme pH,
co-solvents)
Reduced particle size: increased drug Oral: increased rate and extent of absorption, increased bioavailability of drug:
dissolution rate area under plasma versus time curve, onset time, peak drug level, reduced
variability, reduced fed/fasted effects. Pulmonary: increased delivery to deep lung
Solid state: increased drug loading Reduced administration volumes; essential for intramuscular, subcutaneous,
ophthalmic use
Solid state: increased stability Increased resistance to hydrolysis and oxidation, increased physical stability
to settling
Particulate dosage form Intravenous: potential for intravenous sustained release via monocyte phagocytic
system targeting, reduced toxicity, increased efficacy. Oral: potential for reduced
first-pass hepatic metabolism

separation distances, to prevent the particles coming Manufacturing in practice


too close together. Therefore, a non-ionic polymeric For the manufacturing of nanosuspensions, precipitation
surfactant is also used that coats the surface with a conditions are chosen so as to minimize particle size.
hydrophobic chain, and permits a hydrophilic tail to There are two phases involved: the initial creation of
project into the water. Compression of the polymeric crystal nuclei, and their subsequent growth. Formation
coating, as by the approach of a similarly coated particle, of a stable suspension with the smallest particle size
causes loss of entropy and is therefore unfavourable. requires a high nucleation rate but low growth rate. Both
This provides the necessary repulsive barrier between process rates are dependent on temperature, described
two neighbouring particles. The polymeric coating by an ARRHENIUS type of expression, as well as on super-
performs a dual role: inhibiting crystal growth and saturation. The optimum temperature for nucleation
reducing particle size10. might lie below that for growth, which permits selective
Both electrostatic and steric mechanisms are enabled optimization for nucleation12. High-supersaturation
by combining polymers and ionic surfactants, which conditions are chosen for rapid nucleation by metering
therefore complement each other. Entropic steric small amounts of a water-miscible organic solution of
interactions are inherently more sensitive to tempera- the drug to the non-solvent water, under rapid mixing.
ture fluctuations than is electrostatic repulsion. This rapid dilution of solvent results in high-super-
Therefore, temperature cycling could disrupt a sus- saturation conditions, which causes spontaneous
pension stabilized only by polymer. To prevent this, nucleation and a subsequent reduction of the super-
ionic surfactants are used as well. There is a synergy saturation condition in the immediate vicinity of the
between the two, because adding a neutral polymer nucleating crystals; this reduces growth rates. This is
to a surface stabilized with an ionic surfactant permits the opposite of the conditions typically chosen for
greater coverage by the ionic surfactant. This occurs industrial crystallization. In the industrial situation, slow
because self-repulsion of the charged surfactant growth is chosen to generate relatively larger, purer
molecules is minimized, which therefore permits crystals, and the number of very fine particles is delib-
closer packing. erately reduced because of their interference with subse-
It is found for a variety of colloidal systems that the quent downstream processing. Slow growth is also more
attraction energy does not vary significantly, but that likely to directly result in the often preferred, thermo-
the repulsive energy fluctuates with surface potential. dynamically more stable, crystal form. By contrast amor-
Therefore, regardless of the nature of the bulk parti- phous or less stable forms are initially favoured by rapid
cles, colloidal stability is determined primarily by the crystallization, but can subsequently convert to more
choice of surfactants, which affects the repulsive stable forms, according to Ostwald’s ‘law of stages’13. This
potentials. This does not mean that there is no effect instability poses an obstacle unless secondary processing
of drug on the stability of suspensions; the drug is utilized to modify the crystal form (see below).
affects the nature of the surface to which the surfactant The rapid-nucleating conditions have two additional
must bind. consequences, which can be exploited to further reduce
The design of a stable formulation for nanosuspen- particle size. First, at high-supersaturation levels, the
ARRHENIUS EXPRESSION
sions can differ from that used for suspensions of larger crystal habit, or external appearance (as opposed to
A mathematical equation particle size. The latter often strives for weak flocculation, the internal, periodic crystal lattice structure), is changed
that relates the rate of a which corresponds to the secondary minimum, Vs, of to a needle-like or dendritic morphology. These crys-
chemical reaction, k, to change FIG. 3, and yields readily dispersible particles after agita- tals are easily broken, forming additional nuclei,
in temperature, T: tion. This is designed to prevent slow settling, which which catalyses the nucleation process at the expense
k = Ae–E /RT. E is the activation
energy or barrier for the crucial
promotes tight packing and consequent caking of the of growth12. Second, in comparison with crystals
step in the reaction, and R is the suspensions, which are then no longer dispersible11. (For grown more slowly, rapidly grown crystals tend to be
gas constant. a description of settling rate see BOX 2.) more imperfect, and often incorporate impurities and

NATURE REVIEWS | DRUG DISCOVERY VOLUME 3 | SEPTEMBER 2004 | 7 8 7


REVIEWS

Homogenization is advantageously utilized to exploit


Attrition —
for example, three consequences of rapid precipitation to either
milling Dissolution further reduce particle size or to resolve other potential
difficulties26. First, the crystal defects induced by rapid
precipitation render the crystal more susceptible to
Agglomeration Nucleation and
crystal growth rupture by the subsequent mechanical shock of homo-
genization. Second, fracture mechanics predict that the
dendritic morphology itself is more susceptible to
breakage because of the narrow dimension induced,
which must bear the full applied force (FIG. 4). Third, the
mechanical energy supplied by the homogenizer enables
initially formed, unstable amorphous particles that
a b result from rapid precipitation to undergo subsequent
crystallization to a stable state.

Wet milling. Another manufacturing technique for


Water molecule Water–surfactant interaction
preparing nanosuspensions is wet milling, in which the
Figure 2 | Creation and stabilization of nanoparticles, from the perspective of surface active agent, in the presence of surface stabilizer(s), is
energetics. a | In the case of unfavourable energetics, hydrophobic crystal surface directly comminuted by milling media. Particle size here is
contacting water molecules leads to crystal agglomeration, because water molecules are
determined by stress intensity and the number of con-
energetically driven to leave the surface, as shown by the arrows. b | A surfactant-stabilized
crystal surface reduces interfacial tension by allowing attractive water–surfactant interactions. tact points. The stress intensity is a function of the
The crystal surface is stabilized and shows reduced tendency to agglomerate. kinetic energy of the grinding beads, and the number of
contact points can be increased by utilizing smaller
grinding media. A drive shaft, attached to rotating
dislocations. This occurs because of the insufficient time disks, provides the energy to a charge of crosslinked
afforded for accurate incorporation of solute in to the polystyrene beads to comminute the drug crystals by a
growing crystal lattice. This effect is more pronounced compression–shear action27.
for flexible molecules that have many degrees of freedom.
Both the dendritic morphology, as well as the crystal Other techniques. Yet another technique involves the
defects, can be utilized in subsequent processing by spraying of a drug solution in a volatile organic solvent
homogenization to reduce particle size further. into a heated aqueous solution. Rapid solvent evapo-
ration produces drug precipitation in the presence of
Homogenization. Homogenization14,15 involves the surfactants28.
forcing of a suspension under pressure through a Lyophilization of a nanosuspension can be performed
valve16,17 that has a narrow aperture18,19. Bernoulli’s law to overcome either physical or chemical incompatibility,
requires that the high velocity of the suspension that permitting recovery of the original particle size after
results from flow past the constriction is compensated by reconstitution29. Sterilization can be performed by
a reduction in static pressure. This, in turn, causes bubbles aseptic processing of previously sterilized components30,
of water vapour to form, which then collapse as they exit membrane filtration for particles sufficiently small31,32,
the valve. These cause cavitation-induced shock waves, or, for drugs that will withstand it, steam sterilization33
which crack the particles. Turbulent flow and shear have or γ-irradiation.
also been investigated as mechanisms for reducing par- The manufacturing process is built around the unit
ticle size through homogenization20–22. Crystals are sus- operations of particle-size reduction and sterilization.
ceptible to such breakage to varying degrees. Pure crystals A schematic for an aseptic microprecipitation/homo-
have a theoretical tensile strength that is 100–1,000 times genization process is shown in FIG. 5. The process can be
stronger than what is observed in practice. However, the varied to permit the use of previously sterilized raw
inevitable crystal defects — dislocations and impurities material, sterile addition of excipients before filling, and
— weaken mechanical behaviour by migrating and accu- lyophilization. The unit operations of sterile crystal-
mulating at crystal-grain boundaries, which furnishes a lization, aseptic dispersion of previously sterilized drug
nidus of weakness23–25. and aseptic milling are all used in currently manufac-
tured microsuspensions, such as testosterone, insulin,
dexamethasone acetate and penicillin G34.
Box 1 | Potential energy of surfactant-stabilized suspensions
The repulsive energy of two similarly charged particles is given by the equation Characterization techniques
VR= (εaψ0/κ2)ln[1+exp(-κHo)], in which a is the particle radius, Ho is the distance of Tests for the quality of nanosuspensions are selected on
separation between the two particles, ε is the dielectric constant of the medium, ψ0 is the the basis of the performance required. For pharmaceuti-
electrostatic surface potential, and κ is related to the thickness of the diffuse electric cal use, a suspension must have a minimal tendency to
double layer114. The net repulsive energy decreases with separation of the particles (FIG. 3). agglomerate, which could lead to the formation of a
At shorter distances of separation, there is an attractive force between the two particles hard cake. A suspension with slow sedimentation rate is
due to van der Waals forces. The superposition of both these forces results in an attractive acceptable, perhaps even preferable, provided the prod-
potential well, provided the particles can overcome an energy barrier. uct is re-suspendable and homogeneous. In addition,

788 | SEPTEMBER 2004 | VOLUME 3 www.nature.com/reviews/drugdisc


REVIEWS

number of particles to achieve reasonable levels of


precision becomes tedious. Photon-correlation spec-
troscopy is rapid, but only covers the range of 0.02–3
µm38. Light-obscuration counters have a lower sensitivity
VR cut-off of 0.5 µm36. In addition to characterizing the
mean particle size and particle-size distribution, focus
VT
should also be directed to characterizing the very high-
end particle size, especially for injectables. Although
VR

not intended for this purpose, the United States


Pharmacopeia microscopic particle matter test, with
VM limits on number of particles greater than 10 µm and 25
Potential energy

Distance µm, is serviceable39. Finally, visualization of particle


H0 shape by atomic-force microscopy has been reported40.
0
In the course of the development of a nanosuspen-
sion formulation, one first screens various surfactant
packages to achieve the desired particle size, with a
VS
narrow distribution in size. Once achieved, accelerated
VA stability tests are performed, which challenges the
VA

system both thermally and mechanically. The rapid


assessment of sedimentation potential by near-infrared41
is possible. POLYMORPH stability to processing is assessed
by differential scanning calorimetry42–44 and X-ray
diffraction. On-line monitoring of particle size by
VD near-infrared45 is a possibility.
Figure 3 | Potential energy curve for approach of two nanoparticles. The total potential The bioavailability of a nanosuspension, by any
energy curve VT is a superposition of an attractive curve VA and a repulsive curve VR. As two route of administration, ultimately depends on the
particles approach, they can overcome the energy barrier VM, leading to attractive aggregation of dissolution of the drug. In vitro dissolution testing in
the closely packed particles. To prevent this for microparticulate suspensions, one can formulate a bio-relevant medium46,47 furnishes guidance as to
as a weak floc, making use of the secondary minimum VS114. the potential pharmacokinetics to be expected in vivo,
as explained in the applications section. Dissolution
rates can be affected by pH and the nature of the poly-
the product must be non-toxic and non-irritating. morph, which can therefore affect pharmacokinetics.
A suspension used for injectables must also be sterile, By analogy to recommended practice for liposomal
non-pyrogenic, syringeable, injectable, isotonic and non- dosage forms in clinical trials48, the drug levels in
haemolytic35. blood should be separated into a free component (mol-
Particle size growth is largely responsible for agglom- ecular and protein bound) and nanosuspension-bound
eration, as discussed earlier. A variety of techniques are fraction (BOX 3).
used to measure particle-size distribution, because of
their different operating principles and features. For Applications
example, laser light diffraction can be used because it is Nanosuspensions are used to advantage in diverse dosage
POLYMORPH fast and suitable for screening large numbers of samples, forms. In some cases, their small size and increased
One of several distinct crystal acquiring data in the useful range of 0.02–2,000 µm36. surface area leads to an increased dissolution rate and
packing arrangements,
However, because it is an ensemble37 rather than a single- increased bioavailability. In other cases, their particulate
potentially having a different
energy of formation and particle-counting technique, it should be calibrated nature dictates targeting of the MONOCYTE PHAGOCYTIC
melting point than other against a more accurate method. When the particles SYSTEM (MPS), with unusual pharmacokinetic conse-
polymorphic forms of the same are smaller than the wavelength of visible light, optical quences. Nanosuspension drugs that are marketed or
molecule. Its formation might microscopy cannot be used as a reference method. But currently in clinical trials are listed in TABLE 2.
be favored in preference to other
polymorphs by manipulation of
in this range, field emission low-voltage scanning elec-
temperature, solvent and degree tron microscopy can be used to image individual par- Oral. In general, oral suspensions are selected because of
of supersaturation. ticles. However, analysing a sufficiently large statistical the superior taste-masking of the particulate form, or
difficulty experienced by the very young and old in
swallowing tablets49, or to overcome solubility problems.
Box 2 | Settling rate Oral nanosuspensions have been specifically used to
Settling rate is given by the Stokes equation (D2/18η)(dp – dm)g, in which D is the particle increase the rate and extent of the absorption of drugs50
diameter, η is the viscosity of the medium, dp and dm are the densities of the particles and due to their solubility limitations. In comparison with a
the medium respectively and g is the gravitational constant. The critical size for settling can conventional 10-µm danazol suspension, a 169-nm
be calculated from the criterion of Overbeek114, which states that according to Stokes law nanoparticulate dispersion showed higher CMAX (3.01
colloidal particles that settle at a rate of only 1 mm in 24 hours, will never settle in actual versus 0.20 µg per ml) and AREA UNDER THE CURVE (AUC)
practice because of Brownian motion. For a density of particles of 1.15 and η of 1, the (16.5 versus 1.0 µg•h per ml) in a pharmacokinetic study
critical particle diameter is calculated at 300 nm. This can alter the stabilization strategy for in dogs. The bioavailability of the nanosuspension was
suspensions that have particle size constrained to this limit. equivalent to that of a cyclodextrin solution formulation,

NATURE REVIEWS | DRUG DISCOVERY VOLUME 3 | SEPTEMBER 2004 | 7 8 9


REVIEWS

b After precipitation
a Raw material Before homogenization c After homogenization

20 microns 20 microns 20 microns

Figure 4 | Engineering breakable crystals with a combination of microprecipitation and homogenization. Crystal
morphology of raw drug material is modified to facilitate breakage into smaller nanoparticles. a | Crystals of starting raw material are
too large and hard to run efficiently through a homogenizer. b | The raw material is solubilized, filter sterilized and precipitated, so as
to yield crystals of needle-like morphology, which are easily broken during homogenization c | Homogenization yields nanoparticles
suitable for parenteral injections.

indicating that the dissolution-rate-limited bioavailabilty the particles are shed from the mucus55,56. Perturbed
observed with the 10-µm suspension had been over- absorption resulting from an initial coating of salivary
come. The absolute bioavailability was 82.3 ± 10.1% of proteins on nanoparticles might be prevented by enteric
an intravenous control injection51. coating57. Targeting to macrophages in inflamed colon
Evidence for enhanced onset of action, reduction mucosa might also be possible58.
of FED/FASTED ratio (G. Liversidge, personal communi-
cation), and enhanced dose proportionality have also
been found. There is even evidence for decreased gas-
Solution of drug in
tric irritancy by nanosizing naproxen52. In this case, Aqueous buffer solution
water-miscible solvent with
with optional surfactant
reducing the particle size from 20–30 µm to 270 nm optional surfactant
led to faster absorption (tmax = 23.7 versus 33.5 min).
The implied decrease in gastric residence time, with
associated local high and prolonged concentration of
naproxen, was presumed to be responsible for the Sterilizing filter Sterilizing filter
reduced gastric irritancy scores. These effects are med-
iated primarily by an increase in dissolution rate sec-
ondary to increased surface area, as elaborated in the
Noyes–Whitney equation dC/dt = [DA/hV][Cs–C], in
MONOCYTE PHAGOCYTIC which dC/dt is the dissolution rate of a drug formula- Suspension of amorphous or semi-crystalline particles
SYSTEM (MPS)
The system of cells deployed
tion, D is the diffusion coefficient, h is the thickness of
throughout the body tissues, the diffusion layer at the solid–liquid interface, A is the
derived from monocyte surface area of drug exposed to the dissolution media,
precursor cells, which V is the volume of the dissolution media, Cs is the Homogenize
functionally police interstitial
concentration of a saturated solution of the solute in
fluid and blood for unwanted
particulate contamination, the dissolution medium at the experimental tempera-
removing them by phagocytosis. ture, and C is the concentration of drug in solution
at time t53. Stable crystalline product
CMAX Although reductions in particle size increase uptake
Highest drug concentration
by both dissolution and particulate pathways (see
in a plot of the plasma drug Remove solvent
concentration versus time. below), increased self-agglomeration of small particles
can, paradoxically, lead to reduced uptake, which must
AREA UNDER THE CURVE therefore be anticipated. Fill/finish
(AUC). Area under the curve
In addition, increased surface area and decreased
of a plot of plasma drug
particle size can lead to increased muco-adhesion, which Figure 5 | Schematic of aseptic microprecipitation/
concentration versus time,
measured after administration can increase gastrointestinal transit time and lead to homogenization process. Although the final dosage form is
particulate in nature, the drug can be filter sterilized by initially
of a drug. increased bioavailability. However, this result is compli-
dissolving it in a water-miscible organic solvent, and passing
cated by the effect of both surface charge54 and surfactant the resulting solution through a sterilizing filter. All subsequent
FED/FASTED EFFECTS
A difference in the absorption
type of the nanoparticles. These parameters determine steps are conducted in an aseptic environment (previously
of a drug depending on the fed the particles’ adsorption to, and penetration through, the sterilized and maintained as such). Following a solvent removal
or fasted state of the test animal. mucus layer to reach the underlying epithelial cells before step, the aqueous suspension is filled into vials.

790 | SEPTEMBER 2004 | VOLUME 3 www.nature.com/reviews/drugdisc


REVIEWS

By reducing the size of particles to the sub-micron


Box 3 | Characterization tests
level, the uptake of intact gastrointestinal polymeric
particles has been shown to occur preclinically59,60, by Chemical
mechanisms involving M-cells in Peyer’s patches of the • Active ingredient
gastrointestinal lymphoid tissue61. This uptake pathway • Degradation products
communicates with the mesenteric lymph ducts, and • Moisture (for lyophilized and solid dosage forms)
empties via the thoracic duct into the systemic blood
• Preservatives
circulation. This approach therefore provides a route
for avoiding first-pass metabolism, as well as for target- • pH
ing sanctuaries of lymphatic-mediated diseases62. The Physical
low drug uptake by this pathway might be enhanced, as • Particle-size distribution
shown in cell-based studies, by coating drug particles
• Particle-size distribution in response to accelerated
with agents, such as vitamin B1263, that dock into trans-
ageing and shipping (freeze/thaw, mechanical agitation,
porter receptors on the intestinal epithelium. The use centrifugation)
of surfactants has also been found to have an impact on
• Drainability (from sides of container)
decoupling the intestinal P-glycoprotein drug-efflux
pump as well as interfering with lipidic chylomicron • Syringeability, injectability
transport systems. • Re-suspendability
• Dissolution in water or biorelevant medium
Injectable. Injection of poorly water-soluble drugs is • Compatibility after admixture
often approached by formulating drugs with excessive
• Zeta potential (electrostatic self-repulsion of particles)
amounts of co-solvents64, which provoke anaphylactoid
reactions65,66, pain on injection67 and precipitation of Biological
drug following dilution of the vehicle in the blood68,69. • Sterility
Lipids might work for drugs that are lipid-soluble70, • Pyrogenicity
which often is not the case for drugs having high crystal
• In vivo pharmacokinetics
energies71. To establish a more comprehensive approach,
therefore, the formulation of injectable drugs as
nanosuspensions has emerged. The approach is gener- solubility–pH profile might permit dissolution of the
ally applicable, provided that the aqueous solubility is compound. Their lipophilic character might permit
below several hundred µg per ml, to prevent OSTWALD passage through the phagolysosomal membrane, and
RIPENING. Successful formulation has been reported as as a result they could leave the cellular vesicle, enter
applied to antineoplastic agents72, anaesthetic agents73, the cytoplasm, and then exit the cell by diffusing
antifungals and antibacterials29, as well as for agents for down the drug concentration gradient as shown in
malignant hyperthermia74 and cancer pain75. FIG. 6. This intravenous depot effect will result in a
Several pharmacokinetic profiles can result following pharmacokinetic profile with significantly reduced
the injection of nanosuspensions. If the particles dissolve Cmax, but quite prolonged t1/2. This can be very advan-
in the blood readily, both the pharmacokinetics, and tageous for certain drug classes, for which toxicity is
therefore tissue distribution, will be equivalent to those mediated by peak plasma values, but for which efficacy
for the solution formulations76,77, affording a relatively is driven by AUC, as in the case of triazole antifungals84.
fast onset of action78. Alternatively, depot delivery via Formulation of the antifungal agent itraconazole as a
subcutaneous, intramuscular or intradermal routes nanosuspension resulted in significantly less toxicity
OSTWALD RIPENING offers prolonged drug release, because of the ability to after intravenous injection than the commercially
The tendency for a particle load more drug safely into a small injectable volume79. available cyclodextrin-solubilized drug itraconazole
dispersion to grow in diameter
The greater loading capacity (up to 30%) distinguishes (Sporanox; Janssen Pharmaceutica). This permitted a
over time, by a process in which
the smaller particles dissolve nanosuspensions from polymeric nanoparticulate much higher drug dose to be injected into rats and
preferentially, because of their vehicles. The small size of the particles results in sig- dogs, with a consequent increase in efficacy, as shown
higher solubility, with nificantly faster dissolution than microsuspensions, by mortality and colony counts in Candida albicans-
subsequent crystallization onto provided that post-injection aggregation does not challenged immunosuppressed rat models. The
larger particles, making them
even larger.
occur. Because drug dissolution is often the rate-limiting greater tissue drug loading that resulted enabled the
step in systemic drug uptake from a depot80,81, nano- successful treatment of fungal strains that are normally
PHAGOLYSOSOME suspensions often result in higher peak plasma levels as resistant to itraconazole in animal models treated
A cytoplasmic vesicular well. But because dissolution is not instantaneous, as it with nanosuspension85.
compartment consisting of the
would be for a solution dosage form, there is less toxicity The pharmacokinetic parameters of a nanocrystal
union of a phagosome with a
lysosome, containing enzymes associated with nanosuspensions, enabling high loading suspension of itraconazole, as determined in a clinical
and acid environment designed with safety73. trial86, can be compared with those of the commercial
to digest microbial particles. If intravenously administered particles do not dissolve Sporanox solution, as listed in the Physician’s Desk
immediately, they are subject to uptake by macrophages Reference 87 and other literature. The nanocrystal suspen-
t1/2
Half-life of plasma drug
of the MPS82, primarily the Kupffer cells in the liver and sion occupied a larger volume of distribution (1,677 ±
concentration, a measure of the those in the spleen. As the phagocytized drug particles 827 l versus 796 ± 185 l) and was cleared more slowly
duration of action of a drug. encounter the reduced pH of the PHAGOLYSOSOMES83, their (3.35 ± 1.8 l per h versus 22.9 ± 5.7 l per h) to give a

NATURE REVIEWS | DRUG DISCOVERY VOLUME 3 | SEPTEMBER 2004 | 7 9 1


REVIEWS

Table 2 | Solid-particulate-nanosuspension-based formulations in development and in the market


Drug Indication Drug delivery company Pharma company Route Status
Paclitaxel Anticancer American BioScience American Pharmaceutical Intravenous Phase III
Partners
Undisclosed Anti-infective Baxter NANOEDGE Undisclosed Oral/ Preclinical
multiple intravenous to Phase II
Undisclosed Anticancer Baxter NANOEDGE Undisclosed Intravenous/ Preclinical
oral to Phase I
Rapamune Immuno- Elan Nanosystems Wyeth Oral Marketed
suppressant
Emend Anti-emetic Elan Nanosystems Merck Oral Marketed
Cytokine Crohn’s Elan Nanosystems Cytokine Oral Phase II
inhibitor disease PharmaSciences
Diagnostic Imaging agent Elan Nanosystems Photogen Intravenous Phase I/II
Agent
Thymectacin Anticancer Elan Nanosystems NewBiotics./Ilex Oncology Intravenous Phase I/II
Fenofibrate Lipid lowering SkyePharma Undisclosed Oral Phase I
Busulfan Anticancer SkyePharma Supergen Intrathecal Phase I
Budesonide Asthma Elan Nanosystems Sheffield Pharmaceuticals Pulmonary Phase I
Silver Eczema, NUCRYST Self-developed Topical Phase I
atopic
dermatitis
Calcium Mucosal BioSante Self-developed Oral
phosphate vaccine adjuvant
for herpes Phase I
Insulin Diabetes BioSante Self-developed Oral Phase I

longer half-life (346 ± 225 h terminal versus 35.4 ± 29.4 inflammation. In effect, this would result in passive target-
h mean and 30 h terminal88) and larger area under the ing by the enhanced permeation and retention effect for
plasma concentration curve for the first 24 hours, tumours, and sites of infection and inflammation91,92.
AUC24, (51,558 ± 10,635 µg•h per l versus 30,605 ±
8,961 µg•h per l). This behaviour is consistent with Pulmonary. To target the deep lung, respirable aerosols
MPS depot behaviour, resulting in prolonged delivery should have mean aerodynamic diameters of 1–5 µm93.
for the nanosuspension. The nanocrystal suspension In conventional solid-in-liquid dispersions, the solid drug
was well tolerated. has a particle size that is comparable to that of the aerosol.
A macrophage uptake mechanism also explained There is therefore statistical inhomogeneity in the par-
the behaviour of a clofazimine nanosuspension. After titioning of drug particles among the carrier droplets94.
intravenous administration, both a liposomal and a Nanosuspensions would ameliorate this by increasing the
nanosuspension formulation of this drug had similar number of particles per droplet95. In fact, aerosol cascade
drug distribution to the liver, spleen and lung, and impactor studies have shown significantly higher res-
achieved effective control of a macrophage-inhabiting pirable fractions and lower unwanted systemic uptake
mycobacterium29. via throat deposition for nanosuspensions compared
The particles will remain in the macrophages if they with micronized formulations96. Respirable dose was
are too insoluble, or if they cannot be metabolized to increased from 227 µg to 421 µg by decreasing particle
soluble compounds. To remedy this problem, modifica- size from 4.4 µm to 0.73 µm, prepared by homo-
tion of molecules has been used to increase clearance genization97. The use of already approved surfactants
following phagocytic sequestration. In this case, can be used for drug nanosuspensions98, to avoid safety
nanoparticulate X-ray contrast agents were injected sub- problems posed by polymeric nanoparticulates99.
cutaneously, such that their subsequent cellular uptake In a Phase I clinical trial, nebulized nanocrystal
and delivery to lymph nodes89 resulted in opacification (75–300 nm) budesonide suspension had double the
of these organs. Modification of the molecules enabled Cmax, and almost half the tmax, of larger, 4,400-nm sized
PHOSPHOLIPID-PEG
their subsequent clearance and removal. Pulmicort Respules. The total amount absorbed, as
A molecule consisting of
phospholipid covalently Modification of the surface of slowly dissolving drug indicated by AUC, was comparable. The faster absorp-
bonded to a polyethylene glycol crystals with PHOSPHOLIPID-POLYETHYLENEGYLCOL (PEG) tion, which resulted in higher peak plasma levels,
polymer, used as a reagent to chains90 could delay protein adsorption and opsonization could have been attributable either to more rapid dis-
coat a drug or drug delivery of the particles, thereby reducing macrophage uptake. solution or faster entry to the blood via access to the
vehicle with a polyethylene
glycol (PEG) surface, thereby
This would increase circulation time, and give the particles additional vasculature of the peripheral lung100.
conferring longer blood the opportunity to find, and leak out of, discontinuities of Nanosuspension targeting to the alveolar macro-
circulation time. vasculature, such as occurs in neoplasms, infections and phages is an attractive option for diseases mediated by

792 | SEPTEMBER 2004 | VOLUME 3 www.nature.com/reviews/drugdisc


REVIEWS

these cells, both to elevate drug concentration within the secondary lymphoid organs — mimicking the migration
target cell, and to decrease concentration systemically, path of the mycobacteria — as a kind of Trojan
thereby reducing potential toxicity. With consequently Horse101,102. Enhanced uptake of 0.5-µm versus 3-µm
prolonged regional drug levels, the minimization of sized particles by non-macrophage respiratory epithelium
dose frequency might also be anticipated. This could cell lines has also been demonstrated103.
be useful in tuberculosis, in which the mycobacteria For the efficient delivery of particles to the lungs,
reside in macrophages. Efficacy would occur both by clearance mechanisms must be overcome. These consist
enhanced killing of the intracellular parasites as well as of the muco-ciliary escalator in the upper airways, and
by utilizing the trafficking of alveolar macrophages to alveolar macrophage uptake deep in the lung. As shown

Opsonins
complement,
immunoglobulins

Drug/ Opsonized
nanoparticle nanoparticle
Pseudopod extension Pseudopodia fusion
occurs as surface receptors dock leads to topological
onto opsonizing ligands internalization

Receptor-
mediated
recognition

Crosslinking of cell-
surface receptors
Cytoplasmic
generates intracellular
membrane
signals, reorganizing actin-
based cytoskeleton
to extend pseudopodia Budding,
recycling
of membrane
Phagosome
Recycling compartment
pH 6

pH 5–6
Nucleus
1 µm

Enzyme 500 nm
vesicle
TEM of drug
Trans-Golgi network Lysosome crystals in phagosomes

Phagolysosome
pH 4–5.5

Ideal pH–solubility curve of


drug for phagolysosomal escape

Drug
Solubility

Cytoplasm
pH 7.4

pH 5.0 pH 7.4 Extracellular drug release


Figure 6 | Trafficking of drug nanoparticles by macrophages, monocytes and neutrophils. Nanosuspension drug particles,
opsonized by proteins, dock onto receptors on the cell surface. This initiates phagocytosis or internalization of the particle. Membrane
material is recycled back to cell surface via the recycling compartment, as enzyme vesicles from the trans-Golgi network fuse with
the phagosomes, while the pH is progressively decreased. Fusion with lysosomes lowers pH further. Depending on the pH–solubility
curve of drug, the drug can escape from depots in intracytoplasmic compartments, entering first the cytoplasm then extracellularly,
providing sustained systemic drug release83. TEM, transmission electron micrograph.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 3 | SEPTEMBER 2004 | 7 9 3


REVIEWS

above, nanosuspensions can result in reduced deposition latter publications, it was found that use of the agent
in the former, but enhanced deposition in the latter. Polysorbate 80 in the formulation led to deposition of
The consequences of this for nanoparticles of insulin apolipoprotein E on the nanoparticles, which facilitated
have been noted104, for which phagolysosomal degrada- brain uptake by receptors on the brain endothelial cells.
tion of the protein drug subsequent to macrophage
uptake occurs. However, for a molecule with appro- Concluding remarks and predictions
priate solubility-versus-pH profile, and stability in an Nanosuspension technology initially arose to address
acid environment, macrophage uptake simply pro- solubility needs of drug discovery pipelines. As experi-
PHAGOCYTOSIS
vides a mechanism for sustained release. Additionally, ence was acquired through applications, however, more
Process of removal of modification of the surfactant coating can decrease subtle problems began to be viewed as becoming solvable
105
particulate matter, bacteria, PHAGOCYTOSIS . An intriguing system incorporating through this new technology. Altered pharmacokinetic
viruses, degraded cell debris, nanoparticles, held together by van der Waals forces, to profiles have become appreciated insofar as they improve
and so on by any of several
form large porous particles has been proposed to offer safety and efficacy.
mechanisms (for example,
complement activation, the advantages of both minimal phagocytosis as well as The future growth of nanosuspension technology
opsonization or receptor tenacious adsorption, thereby ensuring maximal disso- will mirror that of drug delivery in general, borrowing
mediation) involving extension lution106. The particles are larger than the 3-µm limit concepts with appropriate adaptation for particulates:
of the cell membrane to for effective phagocytosis, therefore ensuring longer applications to protein delivery will be established;
surround and engulf the object,
followed by severance of the
lifetime within the lung for sustained release. But controlled stability of the amorphous phase will permit
membrane leading to a distinct because they are porous, their density is low, which even faster dissolution of drug nanosuspensions; ver-
vesicle within the cytoplasm of results in an optimal aerodynamic diameter. satility in coating technology will be explored. In intra-
the cell, termed the phagosome. venous delivery, control over endosomal escape following
Central nervous system. Nanosuspensions afford a means phagocytosis of drugs will become improved, and target-
OMMAYA RESERVOIR
A system for the delivery of a of administering increased concentrations of poorly ing will become more widely accepted as its value is
flowable drug formulation to the water-soluble drugs to the brain with decreased systemic confirmed. Passive targeting will adapt what has been
ventricles of the brain, involving effects107. Significant efficacy has been shown with learned from long-circulating liposomes. Blood–brain
a compartment for containment microparticulate busulfan in mice administered intra- barrier targeting will perhaps benefit from the experi-
implanted in a proximal
subcutaneous location.
thecally108. The work has advanced to Phase I in patients ence with polymeric nanoparticles, and oral drug
afflicted with neoplastic meningitis, administered via an delivery could combine nanosuspensions with receptor-
PEGYLATION OMMAYA RESERVOIR for intraventricular delivery, and via mediated particulate uptake. Such a combination,
Coating, either covalently or lumbar puncture. The drug was well tolerated and now that dissolution problems have been largely
by physical adsorption, a
resulted in delayed progression of disease (H. Friedman, solved, would address the last great barrier to drug
polyetheylene glycol polymer
onto a drug molecule, surface personal communication). Epidural injection of a 10% absorption — that of permeability-limited uptake.
or particle, rendering it less butamben suspension for cancer pain was well tolerated The solution to this problem would simultaneously
hospitable to the deposition in dogs and humans75,109.Future work will probably also avoid first-pass hepatic metabolism as well. We predict
of opsonizing proteins which involve less invasive routes, utilizing either passive target- that an era will emerge in which soluble drugs will be
mediate phagocytosis. Such
coated drugs and particles
ing (via PEGYLATION, as has been done for liposomes)110 intentionally converted to insoluble complexes to take
typically manifest extended or active111–113 targeting to the brain following intra- advantage of the benefits conferred by nanosuspension
circulation times in blood. venous administration of nanosuspensions. In these drug delivery.

1. Na, G. C. et al. Physical stability of ethyl diatrizoate 10. Ziller, K. H & Rupprecht, H. H. Control of crystal growth in 18. Kinney, R. R. et al. Homogenization valve. US Patent
nanocrystalline suspension in steam sterilization. Pharm. drug suspensions. Part II: influence of polymers on 5,749,650 (1998).
Res. 16, 569–574 (1999). dissolution and crystallization during temperature cycling. 19. Wilbey, A. Homogenization. J. Soc Dairy Tech. 45, 31–32
2. Horn, D. & Rieger, J. Organic nanoparticles in the aqueous Pharm Ind. 52, 1017–1022 (1990). (1992).
phase-theory, experiment, and use. Angew. Chem. Int. Ed. 11. Tabibi, S. E. & Rhodes, C. T. in Modern Pharmaceutics 3rd 20. Jahnke, S. in Emulsions and Nanosuspensions for the
40, 4330–4361 (2001). edn (eds Banker, G. S. & Rhodes C. T.) Ch. 9 (Marcel Formulation of Poorly Soluble Drugs (eds Muller, R. H.,
Comprehensive, schematically illustrated analysis of Dekker, New York, 1996). Benita, S. & Bohm, B.) 177–200 (Medpharm Scientific,
the various methods of forming nanoparticles. 12. Myerson, A. S. & Ginde, R. in Handbook of Industrial Stuttgart, 1998).
3. Muller, R. H. et al. Solid lipid nanoparticles (SLN) for Crystallization (ed. Myerson, A. S.) 45–46 (Butterworth– 21. Mohr, K.-H. High-pressure homogenization. Part I. Liquid-
controlled drug delivery — a review of the state of the art. Heinemann, Boston, 1992). liquid dispersion in turbulence fields of high energy density.
Eur. J. Pharm. and Biopharm. 50, 161–177 (2000). 13. Garside, J. in Particle Design Via Crystallization (eds J. Food Engineer. 6, 177–186 (1987).
4. Lipinski, C. A., Lombardo, F., Dominy, B. & Feeney, P. Ramanarayanan, R. et al.) 16–25 (AIChE Symposium Series, 22. Pandolfe, W. D. Effect of dispersed and continuous phase
Experimental and computational approaches to estimate Vol. 87, New York, 1991). viscosity on droplet size of emulsions generated by
solubility and permeability in drug discovery and 14. Muller, R. H. & Peters, K. Nanosuspensions for the formulation homogenization. J. Dispersion Sci. Tech. 2, 459–474 (1981).
development settings. Adv. Drug Deliv. Reviews 23, 3–25 of poorly water soluble drugs. I. Preparation by a size 23. Huttenrauch, R. Fundamentals of pharmaceutics. Acta
(1997). reduction technique. Int. J. Pharm. 160, 229–237 (1998). Pharm. Technol. 34, 1–10 (1988).
Provides and explanation and characterization of 15. Liedtke, S. , Wissing, S., Muller, R. H. & Mader, K. Influence Overview of the types of imperfections in crystals and
the increase of solubility and permeability problems of high pressure homogenisation equipment on their impact on subsequent processing.
observed with new drug compounds. nanodispersions characteristics. Int. J. Pharm. 196, 24. Grant, D. & York, P. Entropy of processing: a new quantity
5. Yalkowsky, S. H. Techniques of Solubilization of Drugs 1–14 183–185 (2000). for comparing the solid state disorder of pharmaceutical
(Marcel Dekker, New York, 1981). 16. Pandolfe, W. D. Development of the new Gaulin Micro- materials. Int. J. Pharmaceutics 30, 161–180 (1986).
6. Joshi, A. Microparticulates for ophthalmic drug delivery. GapTM homogenizing valve. J. Dairy Sci. 65, 2035–2044 25. Duddu, S. P. & Grant, D. The use of thermal analysis in the
J. Ocular Pharm. 10, 29–45 (1994). (1982). assessment of crystal disruption. Thermochimica Acta.
7. LeBourlais, C. et al. Ophthalmic drug delivery systems-recent References 16 and 20 explain the theory and operating 248, 131–145 (1995)
advances. Prog. Retinal and Eye Res. 17, 33–58 (1998). principles of the homogenization process quite well. 26. Kipp, J. E. et al. Microprecipitation method for preparing
8. Weiner, M. and Bernstein, I. L. Adverse Reactions to Drug 17. Schultz, S., Wagner, G. & Ulrich, J. On the influence of submicron suspensions. US Patent 6,607,784 B2 (2003).
Formulation Agents (Marcel Dekker, New York, 1989). geometric parameters of a combined orifice valve on the 27. Merisko-Liversidge, E., Liversidge, G. G. & Cooper, E.
9. Gad, S. C. Drug Safety Evaluation Ch. 13.8 (John Wiley and attainable mean droplet diameter during high-pressure Nanosizing: a formulation approach for poorly-water-soluble
Sons, New York, 2002). homogenization. Eng. Life Sci. 2, 337–340 (2002). compounds. Eur. J. Pharm Sci. 18, 113–120 (2003).

794 | SEPTEMBER 2004 | VOLUME 3 www.nature.com/reviews/drugdisc


REVIEWS

28. Sarkari, M. et al. Enhanced drug dissolution using 52. Liversidge, G. G. & Conzentino, P. Drug particle size reduction 80. Zuidema, J., Pieters. F. A. & Duchateau, G. Release and
evaporative precipitation into aqueous solution. Int. J. Pharm. for decreasing gastric irritancy and enhancing absorption of absorption rate aspects of intramuscularly injected
243, 17–31 (2002). naproxen in rats. Int. J. Pharm. 125, 309–313 (1995). pharmaceuticals. Int. J. Pharm. 47, 1–12 (1988).
29. Peters, K. et al. Preparation of a clofazimine nano- 53. Tong, W-Q. in Water–Insoluble Drug Formulation (ed. Liu, R.) Excellent analysis of the issues involved with
suspension for intravenous use and evaluation of its Ch. 4 (Interpharm, Denver, 2000). intramuscular injectables, and strategies for success.
therapeutic efficacy in murine Mycobacterium avium 54. Desai, M. P. Gastrointestinal uptake of biodegradable 81. Stout, P. J. M. et al. Dissolution performance related to
infection. J. Antimcrob. Chem. 45, 77–83 (2000). microparticles: effect of particle size. Pharm. Res. 13, particle size distribution for commercially available
30. Toguchi, H. Sterility assurance of microspheres. J. Contr. Rel. 1838–1845 (1996). prednisolone acetate suspensions. Drug Development Ind.
62, 51–55 (1999) 55. Duchene, D. & Ponchel, G. Bioadhesion of solid oral dosage Pharm. 18, 395–408 (1992).
31. Zheng, J. Y. & Bosch, J. W. Sterile filtration of NanoCrystalTM forms, why and how? Eur. J. Pharm. and Biopharm. 44, 82. Moghimi, S. M., Hunter, A. C. & Murray, J. C. Long-
drug formulations. Drug Develop. Ind. Pharm. 23, 15–23 (1997). circulating and target-specific nanoparticles: theory to
1087–1093 (1997). 56. Behrens, I., Pena, A. I, Alonso, M. J. & Kissel, T. practice. Pharmacol. Rev. 53, 283–318 (2001).
32. Konan, Y. N., Gurny, R. & Allemann, E. Preparation and Comparative uptake studies of bioadhesive and non- Comprehensive analysis of the factors involved in
characterization of sterile and freeze-dried sub-200 nm bioadhesive nanoparticles in human intestinal cell lines and pharmacokinetic fate of nanoparticles, subject to
nanoparticles. Int. J. Pharm. 233, 239–252 (2002). rats: the effect of mucus on particle adsorption and uptake by the MPS or its avoidance, thereby leading
33. Na, G. C., Stevens, H. J., Yuan, B. & Rajagopalan, N. transport. Pharm. Res. 19, 1185–1193 (2002). to long circulation.
Physical stability of ethyl diatrizoate nanocrystalline 57. Florence, A. T. & Hussain, N. Transcytosis of nanoparticle 83. Mukherjee, S., Ghosh, R. N. & Maxfield, F. R. Endocytosis.
suspension in steam sterilization. Pharm. Res. 16, and dendrimer delivery systems: evolving vistas. Adv. Drug Physiol. Rev. 77, 759–803 (1997).
569–574 (1999). Del. Rev. 50, S69–S89 (2001). Thorough review of intracellular redistribution
34. Floyd, A. G. & Jain, S. in Pharmaceutical Dosage Forms, 58. Lamprecht, A., Schafer, U. & Lehr, C-M. Size dependent following phagocytosis of nanoparticles.
Disperse Systems 2nd edn Vol. 2 (eds Lieberman, H. A., bioadhesion of micro-and nanoparticulate carriers to the 84. Andes, D. et al. In vivo pharmacodynamics of antifungal
Rieger, M. M.. & Banker, G. S.) 295–298 (Marcel Dekker, inflamed colonic mucosa. Pharm Res. 18, 788–794 drugs in treatment of candidiasis, Antimicrob. Agents
New York, 1996). (2001). Chemother. 47, 1179–1186 (2003).
35. Akers, M. J., Fites, A. L. & Robison, R. L. Formulation design 59. Florence, A. T. & Hussain, N. Transcytosis of nanoparticle 85. Rabinow, B. E. et al. Enhanced efficacy of NANOEDGE
and development of parenteral suspensions. J. Parenter. and dendrimer delivery systems: evolving vistas. Adv. Drug itraconazole nanosuspension in an immunosuppressed rat
Sci. Tech. 41, 88–96 (1987). Del. Rev. 50, S69–S89 (2001). model infected with an itraconazole-resistant C. albicans
Excellent overview of the issues associated with the 60. Jani, P., Halbert, G., Langridge, J. & Florence, A. strain. Am. Assoc. Pharma. Scientists AR6184 (2003).
production of parenteral suspensions, and strategies Nanoparticle uptake by the rat gastrointestinal mucosa: 86. Donnelly, J. P. et al. Pharmacokinetics of a 14 day course
for resolving them. quantitation and particle size dependency. J. Pharm. of itraconazole nanocrystals given intravenously to
36. Barber, T. A. in Pharmaceutical Particulate Matter: Analysis Pharmacol. 42, 821–826 (1990). allogeneic haematopoietic stem cell transplant (HCST)
and Control Ch. 8 (Interpharm, Buffalo Grove, 1993). 61. Clark, M., Jepson, M. & Hirst, B. Exploiting M cells for drug recipients. 41st Intersci. Conf. Antimicrob. Agents
37. Weiner, B. B. in Liquid- and Surface-Borne Particle and vaccine delivery. Adv. Drug Deliv. Rev. 50, 81–106 Chemother. A32 (2001).
Measurement Handbook (eds Knapp, J. Z., Barber, T. A. & (2001). 87. Sporanox (Itraconazole) injection: approved labeling.
Lieberman, A) Ch. 5 (Marcel Dekker, New York, 1996). 62. O’Driscoll, C. M. in Lymphatic Transport of Drugs (eds Physicians’ Desk Reference 1772–1776 (Thomson PDR,
38. Lines, R. W. in Liquid- and Surface-Borne Particle Charman, W. N. & Stella, V. J.) 1–35 (CRC, Florida, 1992). Montvale, 2004).
Measurement Handbook (eds Knapp, J. Z., Barber, T. A. 63. Russell-Jones, G. J. et al. Vitamin B12-mediated transport of 88. Willems, L., van der Geest, R. & de Beule, K. Itraconazole
& Lieberman, A) Ch. 4 (Marcel Dekker, New York, nanoparticles across Caco-2 cells. Int. J. Pharm. 179, oral solution and intravenous formulations: a review of
1996). 247–255 (1999). pharmacokinetics and pharmacodynamics. J. Clin. Pharm.
39. <788> Particulate Matter in Injections, Microscopic Particle 64. Bittner, B. & Mountfield, R. J., Intravenous administration Ther. 26, 159–169 (2001).
Count Test. USP 27 The United States Pharmacopeia, of poorly soluble new drug entities in early durg discovery: 89. McIntire, G. L. et al. Time course of nodal enhancement
United States Pharmacopeial Convention Inc. Rockville, Md. the potential impact of formulatin on pharmacokinetic with CT X-ray nanoparticle contrast agents: effect of
(2004). parameters. Curr. Opinion Drug Discov. Dev. 5, 59–71 (2002). particle size and chemical structure. Investigative Radiol.
40. Shi, H. G. et al. Characterization of crystalline drug 65. Theis, J. G. et al. Anaphylactoid reactions in children 35, 91–96 (2000).
nanoparticles using atomic force microscopy and receiving high-dose intravenous cyclosporine for reversal of 90. Onyuksel, H. & Rubinstein, I. Materials and methods for
complementary techniques. Pharm. Res. 20, 479–484 tumor resistance:the causative role of improper dissolution making improved micelle compositions; lipid bonded to
(2003). of cremophor EL. J. Clin. Oncol. 13, 2508–2516 (1995). water soluble polymer. US Patent 6,217,886 (2001).
41. Kuentz, M. and Rothlisberger, D. Rapid assessment of 66. O’Dwyer, P. J. & Weiss, R. B. Hypersensitivity reactions 91. Wu, N. et al. Increased microvascular permeability
sedimentation stability in dispersions using near infrared induced by etoposide. Cancer Res. 68, 959–961 (1984). contributes to preferential accumulation of stealth
transmission measurements during centrifugation and 67. Duma, R. J., Akers, M. J. & Turco, S. J. in Pharmaceutical liposomes in tumor tissue. Cancer Res. 53, 3765–3770
oscillatory rheology. Eur. J. Pharm. Biopharm. 56, 355–361 Dosage Forms: Parenteral Medications 2nd edn Vol. 1 (eds (1993).
(2003). Avis, K. E., Lieberman, H. A. & Lachman, L.) 17–58 (Marcel Classic paper, providing fascinating in vivo
42. Giron, D. Thermal analysis and calorimetric methods in the Dekker, New York, 1992). fluorescence video microscopic analysis of the
characterisation of polymorphs and solvates. Thermochim. 68. Ward, G. H. & Yalkowsky, S. H. Studies in Phlebitis. accumulation of liposomes in tumour tissue due to
Acta 248, 1–59 (1995). VI. Dilution-induced precipitation of amiodarone HCl. vascular permeability.
43. Yoshii, K. Application of differential scanning calorimetry to J. Parenter. Sci. Technol. 47, 161–165 (1993). 92. Lode, J., Fichtner, I., Kreuter, J., Berndt, A., Diederichs, J. &
the estimation of drug purity: various problems and their 69. Davio, S. R. et al. Precipitation of the renin inhibitor ditekiren Reszka, R. Influence of surface-modifying surfactants on the
solutions in purity analysis. Chem. Pharm. Bull. 45, 338–343 upon iv infusion; in vitro studies and their relationship to in pharmacokinetic behavior of 14C-Poly (methylmethacrylate)
(1997). vivo precipitation in the cynomolgus monkey. Pharm. Res. nanparticles in experimental tumor models. Pharm. Res. 18,
44. Barnes, A. F. et al. A review of the applications of thermal 8, 80–83 (1991). 1613–1619 (2001).
methods within the pharmaceutical industry. J. Thermal 70. Davis, S. S. et al. Lipid emulsions as drug delivery systems. 93. Martonen, T. B. & Katz, I. M. Deposition patterns of
Anal. 40, 499–509 (1993). Ann. NY Acad. Sci. 507, 76–78 (1987b). aerosolized drugs within the human lungs. Pharm. Res. 10,
45. Higgins, J. P. Spectroscopic approach for on-line monitoring 71. Yalkowsky, S. H. Techniques of Solubilization of Drugs 1–14 871–878 (1993).
of particle size during the processing of pharmaceutical (Marcel Dekker, New York, 1981). 94. Chan, H.-K. & Gonda, I. Development of a systematic theory
nanoparticles. Anal. Chem. 75, 1777–1785 (2003). 72. Merisko-Liversidge, E. Formulation and antitumor activity of suspension inhalation aerosols. II. Aggregates of
46. Nicolaides, E. et al. Biorelevant dissolution testing to predict evaluation of nanocrystalline suspensions of poorly soluble monodisperse particles nebulized in polydisperse droplets.
the plasma profile of lipophilic drugs after oral administration. anticancer drugs. Pharm. Res. 13, 272–278 (1996). Int. J. Pharm. 41, 147–157 (1988).
Pharm. Res. 18, 380–388 (2001). 73. Boedeker, B. H., Logeski, E., Kline, M. & Haynes, D. 95. Wiedmann, T. S., DeCastro, L. & Wood, R. W. Nebulization
47. Dressman, J. B. & Reppas, C. In vitro–in vivo correlations for Ultra-long duration local anesthesia produced by injection of of NanoCrystalsTM: Production of a respirable solid-in-liquid
lipophilic, poorly water-soluble drugs. Euro. J. Pharm. Sci. lecithin-coated tetracaine microcrystals. J. Clin Pharmacol. –in-air colloidal dispersion. Pharm. Res. 14, 112–116 (1997).
11 (Suppl. 2), S73–S80 (2000). 34, 699–702 (1994). 96. Ostrander, K. D., Bosch, H. & Bondanza, D. An in vitro
48. U. S. Dept. of Health and Human Services, Food and Drug 74. Karan, S. M. et al. Intravenous lecithin-coated microcrystals assessment of a NanoCrystalTM beclomethasone
Administration. Guidance for Industry: Liposome Drug of dantrolene are effective in the treatment of malignant dipropionate colloidal dispersion via ultrasonic nebulization.
Products; Chemistry, manufacturing, and controls; human hyperthermia:an investigation in rats, dogs, and swine. Eur. J. Pharm. and Biopharm. 48, 207–215 (1999).
pharmacokinetics and bioavailability; and labeling Anesth. Analg. 82, 796–802 (1996). 97. Keller, M., Jauernig, J., Lintz, F-C. & Knoch, M. Nebulizer
documentation, August 2002. 75. Shulman, M. Treatment of cancer pain with epidural butyl- nanosuspensions: important device and formulation
49. Donovan, M. & Flanagan, D. in Pharmaceutical Dosage amino benzoate suspension. Reg. Anesth. 12, 1–4 (1987). interactions. Resp. Drug Delivery VIII, 197–206 (2002).
Forms, Disperse Systems 2nd edn Vol. 2. (eds Lieberman, 76. Clement, M. A. et al. Tissue distribution and plasma 98. Jacobs, C. & Muller, R. H. Production and
H. A., Rieger, M. M. & Banker, G. S.) Ch. 8 (Marcel Dekker. clearance of a novel microcrystal-encapsulated flurbiprofen characterization of a budesonide nanosuspension for
New York, 1996). formulation. The Pharmacologist 34, 204 (1992). pulmonary administration. Pharm. Res. 19, 189–194
50. Jia, L., Wong, J., Cerna, C. & Weitman, S. Effect of 77. Gassmann, P. et al. Hydrosols: Alternatives for the parenteral (2002).
nanonization on absorption of 301029: ex vivo and in vivo application of poorly water soluble drugs. Eur. J. Pharm. 99. Dailey, L. A. et al. Nebulization of biodegradable nanoparticles:
pharmacokinetic correlations determined by liquid Biopharm. 40, 64–72 (1994). impact of nebulizer technology and nanoparticle
chromatography/mass spectrometry. Pharm. Res. 19, 78. Viernstein, J. & Stumpf, C. Similar central actions of characteristics on aerosol features. J. Control. Rel. 86,
1091–1096(2002). intravenous methohexitone suspension and solution in the 131–144 (2003).
51. Liversidge, G. & Cundy, K. Particle size reduction for rabbit. J. Pharm. Pharmacol. 44, 66–68 (1992). 100. Kraft, W. K. et al. The pharmacokinetics of nebulized
improvement of oral bioavailability of hydrophobic drugs: The lack of agreement of the in vitro dissolution work nanocrystal budesonide suspension in healthy volunteers.
I. Absolute oral bioavailability of nanocrystalline danazol in in the Viernstein work can probably be attributed to J. Clin. Pharmacol. 44, 67–72 (2004).
beagle dogs. Int. J. Pharm. 125, 91–97 (1995). not using a plasma-simulating solvent. 101. Sharma, R., Saxena, D., Dwivedi, A. & Misra, A. Inhalable
Early paper on the improvement in oral 79. Parikh, I. et al. Microcrystal technology-based sustained microparticles containing drug combinations to target
bioavailability resulting from reducing particle size delivery of nifedipine in rats. The Pharmacologist 33, 197 alveolar macrophages for treatment of pulmonary
to the nano-domain. (1991). tuberculosis. Pharm. Res. 18, 1405–1410 (2001).

NATURE REVIEWS | DRUG DISCOVERY VOLUME 3 | SEPTEMBER 2004 | 7 9 5


REVIEWS

102. Vyas, S. P., Kannan, M., Jain, S., Mishra, V. & Singh, P. 109. Shulman, M., Hoseph, N. J. & Haller, C. A. Effect of 114. Vanderhoff, J. W. in Pharmaceutical Dosage Forms,
Design of liposomal aerosols for improved delivery of epidural and subarachnoid injections of a 10% Disperse Systems 2nd edn Vol. 2 (Lieberman, H. A., Rieger,
rifampicin to alveolar macrophages. Int. J. Pharm. 269, butamben suspension. Reg. Anesth. 15, 142–146 M. M. & Banker, G. S.) Ch. 4 (Marcel Dekker, New York,
37–49 (2004). (1990). 1996).
103. Foster, K. A., Yazdanian, M. & Audus, K. Microparticulate Perhaps the first paper on application of epidural
uptake mechanims of in-vitro cell culture models of the injection of microsuspensions in humans. Acknowledgement
respiratory epithelium. J. Pharm. Pharmacol. 53, 57–66 110. Koukourakis, M. et al. High intratumoural accumulation of Thanks to J. Kipp (Baxter Healthcare Corp.) for his input to figures 1
(2001). stealth® liposomal doxorubicin (Caelyx®) in glioblastomas and 4.
104. Edwards, D. A. Large porous particles for pulmonary drug and in metastatic brain tumours. Brit. J. Cancer 83,
delivery. Science 276, 1868–1871(1997). 1281–1286 (2000). Competing financial interest
105. Evora, C. et al. Relating the phagocytosis of microparticles 111. Kreuter, J. Nanoparticulate systems for brain delivery The author declares competing financial interests: see Web version
by alveolar macrophages to surface chemistry: the effect of of drugs. Adv. Drug Deliv. Reviews 47, 65–81 for details.
1,2-dipalmitoylphosphatidylcholine. J. Contr. Rel. 51, (2001).
143–152 (1998). 112. Gessner, A., Olbrich, C., Schroder, W., Kayser, O. & Muller,
106. Tsapis, N., Bennett, D., Jackson, B., Weitz, D. & Edwards, D. R. H. The role of plasma proteins in brain targeting: species Online links
Trojan particles: large porous carriers of nanoparticles for drug dependent protein adsorption patterns on brain-specific
delivery. Proc. Natl Acad. Sci. USA. 99, 12001–12005 (2002). lipid drug conjugate (LDC) nanoparticles. Int. J. Pharm. 214, DATABASES
107. Grossman, S. A. & Krabak, M. J. Leptomeningeal carcino- 87–91 (2001). The following terms in this article are linked online to:
matosis. Cancer treatment reviews 25, 103–119 (1999). 113. Schroder, U. & Sabel, B. A. Nanoparticles, a drug carrier EntrezGene:
108. Archer, G. E. et al. Intrathecal busulfan treatment of system to pass the blood-brain barrier, permit central http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=gene
human neoplastic meningitis in athymic nude rats. analgesic effects of i.v. dalargin injections. Brain Res. 710, P-glycoprotein
J. Neuro-Oncology. 44, 233–241 (1999). 121–124 (1996). Access to this interactive links box is free online.

796 | SEPTEMBER 2004 | VOLUME 3 www.nature.com/reviews/drugdisc

You might also like