You are on page 1of 24

International Journal of Green Nanotechnology:

Biomedicine

ISSN: 1943-085x (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/ugnb20

Nanoparticles in Drug Delivery: Potential Green


Nanobiomedicine Applications

R. Ravichandran

To cite this article: R. Ravichandran (2009) Nanoparticles in Drug Delivery: Potential Green
Nanobiomedicine Applications, International Journal of Green Nanotechnology: Biomedicine, 1:2,
B108-B130, DOI: 10.1080/19430850903430427

To link to this article: https://doi.org/10.1080/19430850903430427

Published online: 09 Dec 2009.

Submit your article to this journal

Article views: 5175

View related articles

Citing articles: 1 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ugnb20
International Journal of Green Nanotechnology: Biomedicine, 1:B108–B130, 2009
Copyright c Taylor & Francis Group, LLC
ISSN: 1943-085x print / 1943-0906 online
DOI: 10.1080/19430850903430427

Nanoparticles in Drug Delivery: Potential Green


Nanobiomedicine Applications
R. Ravichandran

ABSTRACT. One of the most challenging tasks for formulators in the pharmaceutical industry is the
formulation of poorly soluble drugs. Conventional techniques employed for improving solubility of
these drugs have gained limited success. This holds true more often when dealing with drugs having
poor aqueous as well as organic solubility. Nanoparticles facilitates formulation of hydrophobic drugs
to improve solubility and efficacy mainly through nanosuspension approach. Nanosuspensions are sub-
micron colloidal dispersions of pure drug particles, stabilized by surfactants. This nanobiomedicine
delivery system is simple and advantageous compared to other strategies. Techniques such as media
milling, high-pressure homogenization, and use of microemulsion as a template have been used for
production of nanosuspensions. This green nanobiomedicine can be delivered by various routes, such
as oral, parenteral, pulmonary, and ocular systems. It is also possible to convert nanosuspensions to
patient-acceptable dosage forms like tablets, capsules, and lyophilized powder products. Nanosuspen-
sion technology has also been studied for active and passive targeted drug delivery systems. This review
article focuses on various manufacturing and formulation perspectives and applications of nanosuspen-
sions as a drug delivery system.

KEYWORDS. bioavailability, dissolution velocity, drug administration, drug formulation, green


nanobiomedicine, nanoparticles, nanosuspension, nanotechnology, solubility

INTRODUCTION addition, poorly water-soluble drugs are spe-


cially challenging, as they cannot achieve dis-
The automation of the drug discovery process solution and therefore they have a very difficult
by technologies such as high-throughput screen- pass through the dissolving fluid to contact
ing, combinatorial chemistry, and computer- the absorbing mucosa and to be absorbed.[3] If
aided drug design is leading to a vast num- the dissolution process of the drug molecule
ber of drug candidates possessing a very good is slow, due to the physicochemical proper-
efficacy.[1] Unfortunately, many of these drug ties of the drug molecules or formulation fac-
candidates (about 60%) are exhibiting poor tors, then dissolution may be the rate-limiting
aqueous and nonaqueous solubility and/or er- step in absorption and will influence drug
ratic absorption and hence require innovative bioavailability.[4] This is the case of Bio-
formulations and drug delivery systems.[2] In pharmaceutical Classification System (BCS)

Received September 14, 2009; accepted October 4, 2009.


R. Ravichandran is affiliated with the Regional Institute of Education (NCERT), Bhopal, India.
Address correspondence to R. Ravichandran, Regional Institute of Education (NCERT), Bhopal, 462 013,
India. E-mail: ravincert@gmail.com

B108
R. Ravichandran B109

Class II and Class IV drugs. For these kind TABLE 1. The descriptive terms of
of drugs, micronization,[5−8] nanonization,[9−11] approximate solubility of substances
complexation (e.g., cyclodextrins),[12−17] prepa-
ration of liposomes,[18−20] amorphous solid Parts of solvent needed
dispersions,[21−25] solubilization using cosol- Descriptive terms for 1 part solute
vents, use of permeation enhancers, oily so-
Very soluble <1
lutions, surfactant dispersions, salt formation, Freely soluble 1–10
precipitation techniques, etc., have been pro- Soluble 10–30
posed to increase the rate of dissolution and Sparingly soluble 30–100
especially the drug bioavailability after oral ad- Slightly soluble 100–1000
Very slightly soluble 1000–10,000
ministration for systemic drug absorption.[26] Practically insoluble or insoluble >10,000
These techniques have their own limitations and
hence have limited utility.[27] An alternative and
promising approach is the production of drug The United States Pharmacopeia–National For-
nanosuspensions that improves drug efficacy and mulary (USP-NF) generally expresses the solu-
pharmacoeconomics.[28] A nanosuspension con- bility in terms of the volume of solvent required
sists of drug nanocrystals, stabilizing agents, and to dissolve 1 g of the drug at a specified tem-
a liquid dispersion medium. The dispersion me- perature (e.g., 1 g acetylsalicylic acid in 300 mL
dia can be water, aqueous solutions, or non- H2 O, 5 mL ethanol at 25◦ C). Other references
aqueous media. The terms drugs nanocrystals may use more subjective terms to describe solu-
implies crystalline state of the discrete parti- bility, such as those given in Table 1.[32,33]
cles. The major advantages of nanosuspension Liquids that form a homogenous system when
technology are its general applicability to most mixed in any proportion are said to be miscible
drugs and its simplicity and are now the uni- (e.g., water and ethanol). Those in which only
versal formulation approach for most drugs.[29] certain volume ratios produce homogenous mix-
Nanosuspension technology offers solution not tures are said to be miscible in certain propor-
only to solubility of drug but also alters the phar- tions (e.g., water and chloroform). Immiscible
macokinetic of drug and thus improves drug liquids will not produce a homogenous solution
safety and efficacy.[30] The poorly soluble and in any proportions (e.g., water and olive oil). The
low-bioavailability drug, so called “brick dust” aqueous solubility of all drugs is of interest to
candidate once abandoned from formulation de- us, because it is only in the form of an aque-
velopment, can be rescued by formulating into ous solution that a drug can be absorbed into the
nanosuspension.[31] The aim is “maximizing dis- general circulation to exert a therapeutic effect.
solution rate and boosting bioavailability” of
drugs. Thermodynamic Solubility and Kinetic
Solubility
SOLUBILITY From a thermodynamic or chemical point of
view, each substance has only one solubility
One of the primary physicochemical consid- value at a specific temperature, pressure and vol-
erations in preparing pharmaceutical solutions ume. This value is defined as the saturated con-
is the solubility of the drug in a suitable sol- centration of the substance in solution when it
vent. Solubility may be defined as the maximum is in equilibrium with its most stable solid state
concentration of a substance that may be com- structure. In the literature, the term ‘solubility’
pletely dissolved in a given solvent at a given is often used loosely to mean the metastable or
temperature and pressure. When both solute and kinetic solubility, rather than the solubility at this
solvent are liquids, the term miscibility rather thermodynamically stable state. The apparent or
than solubility may be used to describe the affin- kinetic solubility, i.e., the concentration of the
ity between the liquids.[32] The solubility of a material in solution at apparent equilibrium (su-
substance may be described in a variety of ways. persaturation), decreases to the level of the true
B110 INTERNATIONAL JOURNAL OF NANOTECHNOLOGY: BIOMEDICINE

or thermodynamically stable solubility, after an correspondingly the lowest solubility. The crys-
infinite equilibrium time.[34,35] tal forms with higher free energy may exhibit an
It should be noted that the term ‘apparent sol- apparent solubility that is higher than the true
ubility’ has also been used in the literature to equilibrium solubility for the system. An ap-
discuss a change in solubility due to ionic inter- parent solubility increase can occur anytime the
action. The result of such interionic interactions starting solid material is not the most stable for
may be that the actual or effective number of the given system. However, a metastable crystal
ions is no longer the same as that calculated will produce only a transient increase in sol-
based on the concentration of the solution. This ubility. The most stable crystal will even-
is the reason why sometimes it is necessary to tually precipitate and the apparent solubility
replace concentration with activity, which is the gained will diminish until the thermodynamic
effective concentration. The term apparent sol- equilibrium solubility is reached. The degree
ubility used here refers to a deviation from the of supersaturation and of duration will de-
thermodynamic stability, which is caused by the pend on the characteristics of starting material
existence of disordered structure. On the other and on the nucleation rate and the growth kinet-
hand, the solubility of sparingly soluble salts is ics of the stable form. Consequently, an inherent
often expressed or discussed in terms of the sol- difficulty in working with metastable systems is
ubility product, which is an equilibrium constant that the kinetics of conversion often cannot be
describing the equilibrium between a sparingly predicted or controlled.[36−38]
soluble compound and a saturated solution of By virtue of having a higher apparent solubil-
its ions. The numerical value of the solubility ity, a metastable crystal will have an increased
product of a salt can thus be regarded as a quan- dissolution rate compared to the more stable
titative statement of the limit of solubility of the form. The change in mass, M, as a function of
salt.[34,35] time, t, for a solute is directly proportional to its
apparent solubility, Sapp , according to
Solubility Enhancement
dM/dt = K × A(Sapp − C) (1)
Poor water solubility of drugs is normally as-
sociated with low oral bioavailability in body.
where A is the solvent accessible surface area, C
Saturation solubility enhancement is the most
is the concentration of the solute in solution, and
interesting method to overcome this problem.
K is a constant that includes the diffusion coeffi-
In order to obtain increased solubility for drugs
cient of the solute and hydrodynamic properties
with poor water solubility, it is necessary to al-
of system.[36,37] This equation describes that the
ter the formulation to facilitate solubilization.
larger the apparent solubility, the greater is the
There are many classical approaches for solubi-
dissolution rate.
lization of poorly soluble drugs. These technolo-
It may be practical at time to use a solid mate-
gies are commonly used as primary strategies.
rial that gives a higher apparent solubility and/or
The choice conventional solubilization method
increase in the dissolution rate in order to en-
will depend upon how the drug can be solubi-
hance bioavailability. As an example, polymorph
lized, stability in the system, and biocompatibil-
B of chloramphenicol palmitate, which can pro-
ity of the vehicle for a given delivery route. For
duce a solubility that is roughly 2 times that of
solid dosage form, it may be possible to modify
polymorph A, gives higher blood levels in vivo.
the solid phase to enhance dissolution.
It showed that as the percentage of polymorph
Solid-Phase Modification B increases in the dosage form, there is a linear
increase in the blood concentration of chloram-
It is important to always consider that for phenicol palmitate.[38]
any raw drug substance or formulation, only Through alteration of the solid-state form, it
one solid phase is thermodynamically stable for is possible to increase the apparent solubility of
a given set environmental condition. The most a drug. However, the practical use of a higher
stable solid will have the lowest free energy and energy solid form is limited due to physical and
R. Ravichandran B111

chemical issues. Significant investigations must When the concentrations of salt and acid are
be made in order to assure that a dosage form equal, the pH of the system equals the pKa of
using metastable crystals will maintain integrity the acid. As the pH decreases, the concentration
throughout the product shelf life.[38] of the molecular acid increases and that of the
salt decreases. This has some interesting impli-
Salt Formation and pH Control cations regarding the aqueous solubility of the
acid, because the undissociated form is much
Salt Formation. The aqueous solubility of
less soluble than its salt. Of further interest, ther-
nonelectrolytes is nearly always affected in some
apeutically, is the fact that it is the undissociated
way by the addition of an electrolyte. Salting-out
acid (HA) that more readily penetrates biological
is the precipitation of organic solutes from aque-
tissues to exert a therapeutic effect.[36−39] Thus,
ous solution by the addition of an electrolyte or
in formulating the product, some balance must
salt. This is attributed to competition between so-
be made between the more soluble salt form and
lute molecules for the solvent and is dependent
the biologically active acid and factors other than
upon the size and valence of the ions. Salting-in
pKa and pH must be considered (e.g., safety and
is the increase in solubility of an organic so-
comfort).
lute upon addition of an electrolyte.[36−38] The
By analogy to the weak acid, the total solu-
mechanism of this phenomenon is poorly under-
bility is described by
stood and it is rarely encountered. An example
is the group of proteins called globulins, which
are more soluble in dilute salt solutions than ST = SW (1 + 10(pKa −pH) ) (4)
in water.[36−39] Complex ion formation occurs
when an insoluble solute reacts with a soluble where the pKa refers to HA+ . In addition, one
substance to form a soluble complex. An ex- approach is to alter the polarity of the solute by
ample is the addition of the soluble potassium shifting it between its molecular (undissociated)
iodide (KI) to the insoluble iodine molecule (I2 ) and ionic (dissociated) states. A shift toward the
to form a soluble triiodide complex (KI3 ). ionic form improves solubility of the solute in
pH Control. According to the Henderson- water and other polar solvents. A shift toward the
Hasselbalch equation, the relationship between molecular species improves solute solubility in
pH, pKa , and relative concentrations of a weak nonpolar solvents. Such shifts may be produced
acid and its salt is by altering the pH of the solution (or using the
salt form of the compound).[36−39]
[A− ] = Ka [HA][H+ ] (2)
Cosolvents

where [A ] is the molar concentration of the salt Solute molecules are held together by cer-
(dissociated species) and [HA] is the concentra- tain intermolecular forces (dipole-dipole, in-
tion of the un dissociated acid. duced dipole-induced dipole, ion-ion, etc.), as
Changes in solubility brought about by alter- are the molecules of the solvent. In order for dis-
ations of the solvent pH can be predicted by the solution to occur, these cohesive forces of like
pHp equation. The pHp is the pH below which molecules must be broken and adhesive forces
an acid or above which a base will begin to pre- between solute and solvent must be formed.
cipitate. The solubility of a drug in a given solvent is
The Henderson-Hasselbalch equation can be largely a function of the polarity of the solvent.
simplified to Solvents may be considered polar, semipolar,
or nonpolar. Polar solvents will dissolve ionic
ST = SW (1 + 10(pH−pKa ) ) (3) and other polar solutes (i.e., those with an
asymmetric charge distribution or like dissolves
where SW = the molar solubility of the un disso- like), whereas nonpolar solvents will dissolve
ciated acid, ST = the total molar concentration nonpolar molecules. Semipolar solvents (e.g.,
of the salt form of the drug initially added. alcohols and ketones) may induce a certain
B112 INTERNATIONAL JOURNAL OF NANOTECHNOLOGY: BIOMEDICINE

degree of polarity in nonpolar molecules and long aliphatic chain segment) connected to a po-
may thus act to improve the miscibility of polar lar group. The polar group can be anionic (such
and nonpolar liquids. The relationship between as carboxylate, sulfate, or sulfonate), cationic
polarity and solubility may be used in practice to (such as ammonium, trialkylammonium, or pyri-
alter the solubility of a drug in a pharmaceutical dinium), zwitterionic (such as glycine or car-
solution.[36−39] nitine), or nonionic (such as polyethylene gly-
Another approach is to mix solvents of dif- col, glycerol or sugar). Due to the differences
ferent polarities to form a solvent system of op- in properties of the polar and nonpolar regions,
timum polarity to dissolve the solute. Such sol- surfactants tend to accumulate and orient at in-
vents must, obviously, be miscible. This method terfaces so that each region of the surfactant in-
is referred to as solvent blending or cosolvency teracts with a separate phase. The polar por-
and uses the dielectric constant as a guide to de- tion of the surfactant will associate with the
veloping the cosolvent system. Because many more polar phase (especially if it is water) and
solvents may be toxic when ingested, most sol- the nonpolar portion of the surfactant will re-
vent blends are limited to mixtures contain- main in the more nonpolar surfactant solvent.
ing water, ethanol, glycerin, propylene glycol, In water, as the concentration of surfactant in-
polyethylene glycol 400, or sorbitol solution. creases above a critical value, its molecules
The list is somewhat expanded for solutions for self-associate into soluble structures called mi-
external application. celles. The concentration at which they begin to
There are many pharmaceutical substances form is called the critical micelle concentration
that are nonpolar or are weak acids and bases (CMC).[36,37,39]
whose ionized salt forms are unstable in solu- These micelles are normally spherical, with
tion. In order to dispense solutions of these sub- the nonpolar regions of surfactant molecules
stances, we must derive a solvent of appropriate gathered in the center (core) and surrounded by a
polarity (or nonpolarity). Practically speaking, shell of the polar region, which is in contact with
this is a fairly simple problem to solve. Solu- the water. A nonpolar drug, which is squeezed
tions are prepared containing varying concen- out of water, can locate within the micelle core.
trations of ethanol or acetone in water, ranging A semipolar drug can locate between or partially
from 0% to 100%. The required concentration within the core and the polar shell. Because the
of drug is added to each solution and the solu- micelles are soluble in water, any drug that is
tions are refrigerated overnight, then viewed for incorporated into the micelle will also be solu-
precipitation.[38] ble in the aqueous system. If the monomers of
From this information it is possible to for- surfactant in solution do not affect the solubility
mulate a vehicle, substituting other solvents, of the solute, then the solute concentration will
which is of the necessary polarity and is phar- remain constant (at the intrinsic solubility, SW )
maceutically elegant. These calculated values until the CMC. After the CMC, the solute con-
of the dielectric constant are only approx- centration will increase with increasing surfac-
imate. Interactions between multiple solutes tant (micelle) concentration. A simple equation
and solvents may increase or decrease solu- representing a solute’s total solubility, ST , in a
bility. Nonetheless, the use of the dielectric surfactant system is
constant in formulating solvent systems gives
us a simple and scientific approach to esti-
mating our needs. It is, therefore, a useful ST = SW + κ(Csurf − CMC) (5)
tool.[38,39]

Solubilization by Micelles where Csurf is the total concentration of the sur-


factant and κ is the solubilization capacity. The
Surfactants are molecules with distinct polar solubilization capacity reflects the number of
and nonpolar regions. Most surfactant consists of surfactant molecule that are required to solu-
a hydrocarbon segment (usually in the form of bilize a single solute molecule.[36,37,39]
R. Ravichandran B113

Complexation initial segment of the curve in Figure 1 illus-


trates this. The linear region will continue until
Complexation is association between two the solubility of the complex itself is reached,
or more molecules to form a nonbonded entity at which point the total solubility of the solute
with well-defined stoichiometry. Two types of remains constant as indicated by the central seg-
complexation are most useful for increasing the ment of the curve.
solubility of drugs in aqueous media. Stacking A plateau is analogous to maximum solubility
complexes are formed by the overlap of the of a solute. Further addition of the complexing
planar regions of aromatic molecules, whereas agent can result in a reduction of the concentra-
the inclusion complexes are formed by the tion of the free solute and leveling of the curve at
insertion of the nonpolar region of one molecule the solubility of the pure complex, as illustrated
into the capacity of another molecule (or group by the final segment in the curve in Figure 1. An
of molecules). The mathematical description inclusion complex is produced by inclusion of a
for the equilibrium constant of a 1:1 complex, nonpolar molecule or the nonpolar region of a
K1:1 , is defined by molecule (known as the guest) into the nonpolar
cavity of another molecule or group of molecules
K1:1 = [SL]/[S][L] (6) (known as the host). When the guest molecule
enters the host molecule, the contact between
where [S] is the concentration of the free solute, water and the nonpolar region of the both is re-
[L] is the concentration of free ligand, and [SL] is duced. Thus inclusion phenomena are the result
the concentration of the solute/ligand complex. of the same driving force that produces micel-
The equilibrium constant is also commonly re- lization, self-association, and stacking, namely
ferred to as the stability constant or complexation the squeezing out from water of nonpolar moi-
constant.[36,37] eties.
From the above it can be seen that as the sta- The most commonly used host molecules
bility constant of a 1:1 complex increases, the are the cyclodextrins. These cyclic oligomers of
slope will increase until the value converges to glucose are relatively soluble in water and have
unity for a strong complex in which one ligand cavities large enough to accept nonpolar por-
molecule solubilizes one solute molecule. The tions of common drug molecules. The naturally

FIGURE 1. General solubilization profile for complexation.


B114 INTERNATIONAL JOURNAL OF NANOTECHNOLOGY: BIOMEDICINE

occurring cyclodextrins contain 6, 7, and 8 enhanced surface area obtained in this way,
glucopyranose units and are termed α, β, and the dissolution rate and the bioavailability of
γ , respectively. The size of the cavacity in the poorly water-soluble drugs were expected to be
cyclodextrin is the major factor in determining high.[31,41]
which guest solute will be most acceptable for But from time to time it has been found that
complexation. In general, alkyl groups will fit there are principal limitations regarding the de-
well into the cavity of the α-cyclodextrin. The velopment of solid dispersion/solution methods.
β-cyclodextrin are most well suited for accept- Problems limiting the commercial application
ing single aromatic rings and the γ -cyclodextrin of solid dispersion involve its method of prepa-
has a large enough cavity to accommodate larger ration, reproducibility of its physicochemical
hydrocarbons such as pyrene.[37] The degree to properties, its formulation into dosage forms, the
which a solute molecule will be solubilized by scale up of the manufacturing processes, and the
a cyclodextrin molecule will depend on several physical and chemical stabilities of drug and ve-
properties. The solute molecule must have hicle.
a significant nonpolar portion in order to be
squeezed out of the water and into the cyclodex-
trin cavity. Because the interior dimensions of DISSOLUTION VELOCITY
a given cyclodextrin are fixed, a significant part
of molecule (or whole molecule) must then fit The dissolution process of solids in liquids has
inside the cyclodextrin. The intermolecular in- been described by Hildebrand and Scott[42,43]
teractions between the two molecules will deter- as involving three steps: (1) the removal of a
mine the strength of the complex. The aliphatic molecule from the solute; (2) creation of a hole
compounds are preferentially solubilized by in the solvent; and (3) insertion of the solute
α-cyclodextrin, whereas the aromatic ring com- molecule into the solvent (i.e., solute-solvent in-
pounds are best solubilized by β-cyclodextrin. teraction). This interaction between the solute
In addition, γ -cyclodextrins may increase and the solvent is obviously dependent on the
solubilization of the fused ring compounds.[37] physical and chemical nature of the two par-
ticipating molecules. The processes involved in
Solid Dispersions the dissolution of hydrophobic materials, which
have low aqueous solubility, will differ from
Solid dispersions have been developed five those affecting the dissolution of hydrophilic
decades ago. In 1961, Sekiguchi and Obi devel- substances. The amount of energy required to
oped a practical method whereby many of the remove a molecule of a sparingly soluble drug
limitations of the bioavailability enhancement from the solute particle is lower for a hydropho-
of poorly water-soluble drugs can be overcome. bic drug than for a hydrophilic drug (such as
This method, which was later termed solid dis- those composed of inorganic salts). The inter-
persion, involved the formation of eutectic mix- molecular bonds in inorganic salts are so strong
tures of drugs with water-soluble carriers by the that a large amount of energy is required to
melting of their physical mixtures. Sekiguchi disassociate the discrete molecules. However,
and Obi suggested that the drug was present in a when the individual solute molecules are liber-
eutectic mixture in a microcrystalline state.[31,40] ated, a hydrophilic salt molecule (or ion) is more
Later, Goldberg et al. demonstrated that the en- likely to interact with water than a hydrophobic
tire drug in a solid dispersion might not nec- drug molecule. Thus, the main factor affecting
essarily be present in a microcrystalline state; the solubility and dissolution of a hydropho-
a certain fraction of the drug might be molec- bic drug is the limited energy released when
ularly dispersed in the matrix, thereby forming a drug molecule is bonded to the solvent.[39]
a solid solution. In either case, once the solid In contrast, the main barrier to dissolution of a
dispersion was exposed to aqueous media and sparingly soluble hydrophilic substance appears
the carrier dissolved, the drug was released as to be disruption of the strong intermolecular
very fine, colloidal particles. Because of greatly forces.[34]
R. Ravichandran B115

FIGURE 2. Disintegration, deaggregation, and dissolution stages as a drug leaves a tablet or


granular matrix.

Theory of Dissolution or
When a solid dosage form is introduced into a
beaker of water or into the gastrointestinal tract, dC/dt = DS (Cs − C)/Vh (8)
the drugs begin to pass into solution from the
intact solid. Unless the solid dosage form is a where M is the mass of the solute dissolved
continguous polymeric device, the solid matrix in time t, dM/dt the mass rate of dissolution
also disintegrates into granules and these gran- (mass/time), D the diffusion coefficient of the
ules deaggregate in turn into fine particles. Dis- solute in solution, S the surface area of the ex-
integration, deaggregation, and dissolution may posed solid, h the thickness of the diffusion layer,
occur simultaneously with the release of a drug Cs the solubility of the solid (i.e., the concentra-
from its delivery form. These steps are separated tion of a saturated solution of the compound of
for clarification as depicted in Figure 2.[36] the solid and at the temperature of the experi-
The effectiveness of a solid dosage form in ment), and C the concentration of solute in the
releasing of its drug for systemic absorption de- bulk solution and at time t. The quantity dC/dt
pends somewhat on the rate of disintegration of is the dissolution rate and V the volume of solu-
the dosage form and deaggregation of the gran- tion.
ules. Ordinarily of more importance, however, In dissolution or mass transfer theory, it is
is the dissolution rate of the solid drugs. For assumed that an aqueous diffusion layer or stag-
class II drugs of the Biopharmaceutical Classi- nant liquid film of thickness h exist at the sur-
fication System (BCS), frequently dissolution is face of solid undergoing dissolution, as shown in
the limiting or rate-controlling step in absorp- Figure 3. This thickness h represents a station-
tion of poorly soluble drugs, because it is often ary layer of solvent in which the solute molecule
the slowest of the various step involved in re- exist in concentrations from Cs to C. Beyond the
lease of the drugs from its dosage forms and static diffusion layer, at x greater than h, mixing
passage into systemic circulation [36]. The rate occurs in the dissolution and the drug is found at
at which the solid dissolves in the solvent was a uniform concentration, C, throughout the bulk
proposed in quantitative terms by Noyes and phase.[36]
Whitney in 1897 and elaborated subsequently by At the solid surface-diffusion layer interface,
other coworkers.[44] The equation may be written x = 0, the drug in the solid is equilibrium with
as the drug in the diffusion layer. The gradient
or change in concentration with distance across
dM/dt = DS (Cs − C)/ h (7) the diffusion layer is constant as shown by the
B116 INTERNATIONAL JOURNAL OF NANOTECHNOLOGY: BIOMEDICINE

FIGURE 3. Dissolution of the drug from a solid matrix, showing the stagnant diffusion layer between
the dosage form surface and the bulk solution.

straight downward-sloping line. This is the gra- solubility and intestinal permeability. When
dient represented in Equations 7 and 8 by the combined with the dissolution of the drug prod-
term (Cs − C)/ h. When C is considerably less uct, the BCS takes into account three major
than the drug’s solubility, Cs , the system is rep- factors that govern the rate and extent of drug
resented by sink condition, and concentration C absorption from solid oral dosage forms: dis-
may eliminated from the equations. The equa- solution, solubility, and intestinal permeability.
tion then becomes According to the BCS, drug substances are clas-
sified as follows[46,47] :
dM/dt = DS Cs / h (9)
Class I: High Solubility–High Permeability
For a drug powder consisting of uniformly Class II: Low Solubility–High Permeability
sized particles, it is possible to derive an equa- Class III: High Solubility–Low Permeability
tion that expresses the rate of dissolution based Class IV: Low Solubility–Low Permeability
on the cube root of the weight of the parti-
cles. The radius of the particle is not assumed The Biopharmaceutical Classification Sys-
to be constant. The equation is known as the tem (BCS) groups poorly soluble compounds as
Hixson-Crowell cube root law,[45] and presented Class II and IV drugs, compounds that feature
as poor solubility and high permeability, and poor
solubility and poor permeability, respectively.
Mo1/3 − M 1/3 = κt (10) Drug substances are considered highly soluble
when the largest dose of a compound is soluble
in <250 mL water over a range of pH from 1.0 to
where Mo is the original mass of the drug parti-
7.5; highly permeable compounds are classified
cles, Mmass particles at time tand κ is the cube
as those compounds that demonstrate >90% ab-
root dissolution rate constant.
sorption of the administered dose. In contrast,
Biopharmaceutical Classification System compounds with solubilities below 0.1mg/mL
(BCS) face significant solubilization obstacles, and of-
ten even compounds with solubilities below
The BCS is scientific framework for classi- 10 mg/mL present difficulties related to sol-
fying drug substances based on their aqueous ubilisation during formulation.[48] Figure 4
R. Ravichandran B117

FIGURE 4. Possibilities of shifting the solubility-dissolution characteristics from a very poorly soluble
drug to dose:solubility ratio (D:S) within the range of values encountered in the human GI tract
(D:S > 250 mL).

gives a diagramic representation of solubility- ing,[9,62] high-pressure homogenization, either


dissolution characteristics. in water[10] or in water-free or water-reduced
media,[63] by combination technologies, e.g.,
precipitation and subsequent high-pressure
NANOSUSPENSIONS homogenization[64] or ball milling and subse-
quent high-pressure homogenization.[65] In the
Theoretical Overview literature, many data are available covering
the formulation and optimization of nanosus-
Recently, many poorly soluble drugs have pensions.[66−68] Liversidge and Cundy have
been nanonized to increase their dissolution rate, reported that if bioavailability is truly disso-
their saturation solubility, and in turn to en- lution rate limited, particle size reduction can
hance their oral bioavailability.[49−55] The con- significantly improve the oral bioavailability
cept of oral nanosuspensions has been specifi- of the drug.[62] It has been also investigated
cally used to increase the rate and extent of the that producing nanosuspensions for oral ad-
absorption of drugs that have poor and/or er- ministration leads to effective therapeutic
ratic dissolution. In the current decade, the con- concentrations in the blood because solubility
cept of nanosuspensions could be commercially and absorption problems in the gastrointestinal
exploited by pharmaceutical companies as mi- tract have been overcome by extensive size
cronization did in the last few decades. There- reduction.[10,49,56,62,69,70]
fore, drug nanocrystals represent an alternative An outstanding feature of nanosuspensions is
to existing drug delivery technologies for poorly the increase in saturation solubility and conse-
soluble compounds.[56,57] quently an increase in the dissolution velocity of
Drug nanonization can be achieved through the compound. The increase in saturation solu-
different techniques, where one distinguishes be- bility can be explained by the Kelvin–Gibbs and
tween the bottom-up and the top-down techno- the Ostwald–Freundlich equations. The Kelvin
logies. Bottom-up technologies are precipitation equation describes the vapor pressure over a
methods,[58−61] whereas top-down technologies curved surface of a liquid. It describes droplets
start from coarse drug macrosuspensions. The in a gas phase. The vapor pressure increases
diminution is achieved either by pearl/ball mill- with increasing curvature of the droplets, which
B118 INTERNATIONAL JOURNAL OF NANOTECHNOLOGY: BIOMEDICINE

means decreasing particle size. It has also been crease in the surface area consequently increases
postulated that the surface curvature of the dis- the dissolution velocity, e.g., exploited in mi-
solving solid particles will influence solubility in cronized or nanosized products. In addition, drug
water. The basic theory derives from the classical nanoparticles are characterized by an increase
Gibbs–Kelvin equation, which, when adapted to in saturation solubility Cs . According to Noyes
the solubility of solids, is known as the Ostwald– and Whitney, the increase in Cs —in addition to
Freundlich equation: enlargement surface area—further increases the
dissolution velocity. The final saturation solu-
−(G) = RT ln(Sr /S∞ ) = (2γ V )/r bility achieved is, of course, compound-specific
(11) based on the differences in compound-specific
or it can be simplicity written as dissolution pressures. The dissolution velocity is
reversely proportional to the diffusional distance
ln(Sr /S∞ ) = 2γ M/ρrRT (12) h, which means that reducing h leads to a further
increase in dissolution velocity. According to the
where (G) is the difference in the free en- Prandtl equation,[52,74,75] the diffusional distance
ergy of a solution of small and large particles; h decreases for very small particles. The simul-
Sr and S∞ are the solubility of a spherical parti- taneous increase in saturation solubility Cs and
cle of radius r and the solubility of a noncurved decrease in h leads to an increased concentra-
solute surface (r → ∞), respectively; V is the tion gradient (Cs − Cx )/ h, thus enhancing the
molar volume of the solute; M is molarities of dissolution velocity in addition to the surface
the solute; ρ is density; γ is the solid–liquid in- effect.
terfacial tension; R is the universal gas constant;
T is the absolute temperature; and r is the parti- Production Process
cle radius. Equation 11 is valid for particles that
There are several production techniques to
have a very large surface-to-volume ratio and is
produce drug nanocrystals. Basically, one can
of practical importance only for particles smaller
differentiate between top-down and bottom-up
than 1.0 µm in diameter.[39,71−73] It has also been
technologies. Typically, the drug nanocrystals
claimed that the surface of finely divided solids
are generated in a liquid dispersion medium
may be less regularly crystalline and more amor-
(e.g., by precipitation or a disintegration pro-
phous than that of well-grown crystals.[73] There
cess). The obtained product from this process
are thus different reasons behind deviations from
is a suspension of drug nanocrystals in a liquid
the thermodynamically stable solubility. Factors
stabilized by a surfactant or polymer (so-called
such as impurities, ion effect, particle size, and
“nanosuspensions”). In contrast to micronized
crystal structure are some of the factors that may
powders, the drug nanocrystals can be adminis-
lead to such deviations.
tered using very different administration routes.
The most important feature of nanocrys-
Oral administration is possible as a suspension.
tals is the increase in saturation solubility and
More patient-convenient dosage forms can be
surface area, and consequently an increase in
produced by transferring the liquid nanosuspen-
the dissolution velocity of the compound. The
sions to solid dosage forms, i.e., tablets or pellets
law of Noyes–Whitney describes the dissolution
or granulate-containing capsules. In addition,
velocity[44] :
because of their small size, the nanosuspen-
sions can be injected parenteraly, especially
dC/dt = DA(Cs − Cx )/ h (13)
intravenously. Intravenous injection leads ‘per
definition’ to a 100% bioavailability.[50]
D is the diffusion coefficient, A is the surface
area, Cs is the saturation solubility, Cx is the bulk Bottom-up Technologies
concentration, and h is the so-called “diffusional
distance” over which the concentration gradient The term “bottom-up technology” means that
occurs (note: division of the equation by the vol- one starts from the molecular level, and goes via
ume v leads to dC/dt). It is obvious that an in- molecular association to the formation of a solid
R. Ravichandran B119

particle. That means that we are discussing clas- r The drug needs to be soluble in at least
sical precipitation techniques by reducing the one solvent (thus excluding all new drugs
solvent quality, for example, by pouring the sol- that are simultaneously poorly soluble in
vent into a nonsolvent or changing the temper- aqueous and inorganic media).
ature or a combination of both. Precipitation is r The solvent needs to be miscible with at
a classical technique in pharmaceutical chem- least one nonsolvent.
istry and technology. The Latin terminology is r Solvent residues need to be removed, thus
via humida paratum (v.h.p.), which means be- increasing production costs.
ing prepared via a liquid process (solutions are r It is a little bit tricky to preserve the particle
made to obtain a fine powder [precipitate] dis- character (i.e., size, especially the amor-
persed in a ‘wet’ environment). Because it is a phous fraction).
long known process in pharmacy, a clear outline
is necessary to document the innovative height In general, it is recommended that a sec-
of patent applications based on this process.[54] ond consecutive process be performed for par-
The particles generated by precipitation, as ticle preservation, that is, spray drying or
for example by Sucker, are in most cases crys- lyophilization.[54]
talline in nature; in contrast to this, the com-
pany Knoll (nowadays owned by Abbott, the new Top-down Technologies
name is “Soliqs”) created amorphous particles There are basically two top-down techniques,
by a precipitation technique.[23] The product is which means starting from a large-sized powder
NanoMorphTM . As outlined above, an additional and performing a size diminution.[50,54]
positive feature is a further increase in the disso- Pearl Milling. Liversidge and co-workers de-
lution velocity due to the amorphous character veloped the milling process to yield the so-called
of the product. The precipitation in the amor- 
R
NanoCrystals . The mills used by the Elan Com-
phous form is achieved by an aqueous polymer
pany are basically containers with small pearls,
solution. However, for a commercial product, it
beads, or balls that can have different sizes, a
is necessary to preserve the amorphous character
typical size being 1–2 mm. The drug powder is
during the shelf life to avoid changes in bioavail-
dispersed in a surfactant solution, and the ob-
ability caused by a reduction in the dissolution
tained suspension is poured into the mill. In the
velocity due to the transfer of the amorphous
milling process, the pearls are moved, either by
drug to a crystalline drug.[54]
using a stirrer or by moving the milling container
The advantage of the precipitation techniques
itself. The drug particles are disintegrated be-
is that relatively simple equipment can be used.
tween the moving pearls. This process is accom-
For example, the solvent can be poured into
panied by the erosion of milling material from
the nonsolvent with a constant velocity in the
the pearls, a phenomenon well known for these
presence of a high-speed stirrer. Very elegant
mills and documented in the literature.[54] The
approaches are the use of static mixers or mi-
milling technique clearly has some advantages:
cromixers, which simulates the precipitation
conditions in a small volume (i.e., simulating r Simple technology
lab-scale conditions), but being simultaneously r Low-cost process regarding the milling it-
a continuous process. In the case of micromix- self
ers, scaling up can be performed in a simple way r Large-scale production possible to some
just by so-called numbering up, which means extent (batch process)
arranging many micromixers in parallel. The
equipment is relatively simple, and in the case As disadvantages, we can see:
of large conventional static mixers, of relatively
low cost (this is not necessarily valid for the r Potential erosion from the milling material
micromixers). However, there are some major leading to product contamination
general disadvantages of the precipitation tech- r Duration of the process not being very pro-
niques. duction friendly
B120 INTERNATIONAL JOURNAL OF NANOTECHNOLOGY: BIOMEDICINE

FIGURE 5. Change of the diameter of the streaming dispersion in a piston-gap homogenizer from
the cylinder containing the bulk suspension to then narrow homogenization gap. The actual range
of the static pressure as a function of the location inside the homogenizer is given in the diagrams
below (left: situation for homogenization in water; right: homogenization in water-mixtures or water-
free media).

r Potential growth of germs in the water very tiny gap with an extremely high velocity.
phase when milling for a longtime Prior to entering the gap, the suspension is
r Time and costs associated with the sep- contained in a cylinder with a relatively large
aration procedure of the milling material diameter compared to the width of the following
from the drug nanoparticle suspension, es- gap. In the APV LAB 40, the diameter of the
pecially when producing parenteral sterile cylinder is about 3 cm, it narrows to about
products roughly 3–25 µm (varies with applied pressure)
when the suspension enters the homogenization
Homogenization in Water. Homogenization gap. The cavitation and shear forces in the
in water is performed either using microflu- gap were sufficiently high to break particles
idizers as high-pressure homogenizers (Skye that were distinctly larger than the gap width.
Pharma Canada Inc.) or piston-gap homogeniz- Therefore it is possible to disrupt relatively
ers. In microfluidizer, a frontally collision of large sized powders (up to 200 µm).[50,54,77]
two fluid streams under pressure up to 1700 According to the law of Bernoulli, the flow
bar leads to particle collision, shear forces, and volume of a liquid in a closed system per cross-
cavitation forces. The collision chambers are section is constant. That means the reduction
designed as Y-type or Z-type. A major dis- in the diameter leads to a tremendous increase
advantage of microfluidizers is the required in the dynamic pressure and simultaneously a
long production time. In many cases, time- decrease of the static pressure when the liquid
consuming 50–100 passes are necessary for suf- is in the homogenizer gap. A liquid boils when
ficient particle reduction.[76] The company Skye its vapor pressure is equal to the air/static
Pharma Canada Inc. applies this technology to pressure of the environment. In the gap, the
produce submicron particles of poorly soluble static pressure drops below the vapor pressure
drugs.[50,76] of the liquid at room temperature (Figure 5).
Cavitation was employed as the most impor- Consequently, the liquid starts boiling, forms
tant effect to diminute particles in a piston-gap gas bubbles that implode after leaving the
homogenizer. In this homogenizer types, the homogenization gap, and being again under
dispersion (emulsion or suspension) passes a normal air pressure conditions.[50,54]
R. Ravichandran B121

There are advantages of the piston-gap ho- (HPMC) capsules. Nanosuspensions in mix-
mogenization technique: tures of water with water-miscible liquids (e.g.,
ethanol, isopropanol) can be used in the gran-
r The effective particle diminution ulation process for tablet production; the sol-
r Production lines can be qualified and vali- vent mixture evaporates much better than wa-
dated ter. Drugs that are susceptible to hydrolysis in
r Production lines already exist in industry, water can be prepared as nonaqueous suspen-
for example, production of parenterals, and sions, for example ethanol, glycerol, or propy-
so on lene glycol.[80] Prior to intravenous injection, the
r No contamination, for example, iron con- glycerol drug nanosuspensions can be diluted
tent is below 1 ppm[78] with water for injection to yield an isotonic sus-
r Off-shelf equipment pension. Alternatively, isotonic water glycerol
r Simple process mixtures can be directly homogenized. Previ-
r Low-cost process/low-cost equipment ously cavitation was considered as the domi-
nating factor for particle diminution; therefore,
To summarize: homogenization with piston- homogenization was only performed in pure wa-
gap homogenizers has good potential to be used ter, and glycerol was added afterwards to avoid
as a production technique because it fulfills the an impairment of particle diminution efficiency.
key industrial features such as large-scale pro- 
R
Another option of the Nanopure technology is
duction and various regulatory aspects.[50,54] the production of aqueous polymer particle dis-
Homogenization in Water–Liquid Mixtures persions with a diameter of a few micrometers

R
and Nonaqueous Media. The DissoCubes or in the nanometer range. This could be an al-
patent describes homogenization in pure water ternative to polymer dispersions (e.g., surelease,
as a dispersion medium; the rationale behind— ethyl cellulose) for the coating of tablets that are
as explained in detail in the patent—is that the now produced by using solvents. Aqueous shel-
cavitation is seen as being responsible for the 
R
lac dispersions produced with Nanopure tech-
diminution of the particles. To obtain cavita- [54]
nology have already been described. Table 2
tion in the dispersion medium, one needs to gives a comparison of the three techniques.
have a high vapor pressure of the dispersion
fluid at room temperature. This is fulfilled with Combination Technology
water, but not with other liquids such as oils
or liquid polyethyleneglycol (PEG) 600. In ad- In 2001, the Baxter Healthcare Company pre-

R
dition, homogenization at higher temperatures sented their NANOEDGE technology at the
(e.g., 80◦ C) should be much more efficient than annual meeting of the American Association
at room temperature because the vapor pressure of Pharmaceutical Scientists (AAPS) in Denver.

of water is much higher at 80◦ C, thus leading
R
NANOEDGE is an aqueous suspension of drug
to more extensive cavitation.[79] The basic of the nanoparticles suitable for intravenous injection.
R
Nanopure technology is that homogenization The particles are prepared basically by a high-
was performed against these teachings, leading pressure homogenization process, which means
to a comparable or even improved product. Un- either by microfluidization or homogeniza-

R
der the Nanopure technology, not only drugs, tion in water using piston-gap homogenizers,
but also polymers can be processed, leading to a or alternatively, by a new production process
product with a mean diameter in the nanometer developed by Baxter.[54] This process is called
range (nanoparticles) or a size of a few microm- the “microprecipitation method,” and is a com-
eters (both particle ranges are covered). bination of precipitation followed by a process
Nonaqueous dispersion media can be used to with high-energy input (e.g., homogenization).
produce oral formulations, for example, drug The process includes three steps:
nanocrystals dispersed in liquid PEG 600 or
Miglyol (MCT) oil, that can be directly filled r dissolving the organic compound in a
into soft gelatin capsules, sealed hard gela- water-miscible first solvent to form a so-
tine capsules, or hydroxypropylmethyl cellulose lution;
B122 INTERNATIONAL JOURNAL OF NANOTECHNOLOGY: BIOMEDICINE

TABLE 2. Advantages and disadvantages of effective diminution in the subsequent high-


different methods for the production of pressure homogenization step. This is of special
nanocrystals importance when the available jet milled drug is
highly crystalline and simultaneously possesses
Technology Advantages Disadvantages a very hard crystal structure (high Mohs degree).
rFinely dispersed rNeeds to be In such cases, the direct homogenization of
Precipitation
the jet-milled powder can require a higher
drug stabilized
rGood control of rOrganic solvent number of homogenization cycles, which means
desired size residue longer production times. In the case of very
rNot universally hard crystals, the maximum homogenization
applicable, only
pressure applicable during production (e.g.,
drugs with certain
properties are 2000 bar) might potentially lead to a maximum
possible (e.g., dispersitivity being rather close to 1 µm, and not
soluble in at least in the preferred nanometer range.[50,54] Another
one solvent)
Milling rLow-energy rResidue from combination technology has been developed
technique milling media by Petersen using ball milling and subsequent
rProven by 4 fda rCan be a slow high-pressure homogenization (CT).[65] With
approved drugs process (several the CT technology, the performance (e.g., phys-
days)
rNeeds to be ical stability) of nanocrystals could be distinctly
stabilized improved, as for example for dermal applica-
rLarge batches tion. Nanocrystals can increase the penetration
difficult to produce of poorly soluble cosmetic and pharmaceutical
due to size of
actives into the skin. The increased saturation
milling chamber
Homogenization rUniversally rHigh-energy solubility leads to an increased concentration
applicable technique ingredient, thus promoting passive penetration.
rNo problem with rGreat experience Molecules penetrated into the skin are very
large batches needed
rFast method fast replaced by new molecules dissolving from
(several minutes the nanocrystal depot in the cream. The first
possibly) four cosmetic nanocrystal products with rutin
rWater free were launched by Juvena. Compared to the
production
water-soluble rutin glucoside, the original rutin
possible
molecule as smartCrystal formulation possesses
a 500 times higher bioactivity (as measured by
sun protection factor [SPF]).[65,81] Of course the
r mixing this solution with the second sol- same principle can be applied to poorly soluble
vent to obtain a “presuspension” (= pre- pharmaceutical drugs of interest for dermal
cipitate); and application. Dermal application of nanocrystals
r adding energy to this “presuspension” to is protected by a US and Patent Cooperation
form particles having an average effective Treaty (PCT) patent application.[65,81]
particle size of 400 nm to 2.0 µm.

The advantages of the Baxter method Other Techniques for Production of Drug
are that the problem of the precipitation Nanoparticles
technique—continuing crystal growth after
precipitation—has been overcome because the The intensive researches of new technolo-
particles are only an intermediate product to gies lead to many other approaches to produce
undergo subsequent diminution/annealing. In nanocrystals. There are many alternative tech-
addition, by optimal choice of precipitation con- nologies, as discussed below.
ditions, more friable particles (semicrystalline, Precipitation with Compressed Fluid Antisol-
amorphous) can be produced which allow more vent. This technique uses an antisolvent-based
R. Ravichandran B123

recrystallization process. CO2 is used as antisol- Rapid Expansion of Supercritical Solution


vent. The substances are atomized into a cham- into Aqueous Solution. In contrast to RESAS,
ber containing compressed CO2 . As two liq- the rapid expansion of supercritical solution
uids collide, intense atomization into micronized into aqueous solution (RESSAS) process uti-
droplets occurs, subsequently drying of micro- lizes a supercritical fluid that is expanded into
droplets occurs as the solvent(s) and CO2 mix. an aqueous solution containing a stabilizer. This
Nanoparticles are formed because the two way technique was used by Turk et al. to produce
mass transfer: extraction of organic solvent and phytosterol particles with a diameter less than
CO2 diffusion into the droplets.[82] 500 nm. The surfactants or stabilizers are dis-
High mass transfers rates have been success- solved in the aqueous phase, not in the super-
fully achieved in the solution enhanced disper- critical fluid.[88]
sion by the supercritical fluids (SEDS). The Cryogenic Spray Processes. The cryogenic
SEDS process was patented by Hanna and York spray process is an attractive alternative to obtain
and owned by University of Bradford.[83] This microparticles and nanoparticles of poorly sol-
technique has produced, e.g., a new polymorph uble drugs. Halocarbon refrigerants and liquid
of flucticasonepropionate, it also enabled con- nitrogen have been used as cryogenic media in
trol over the particle size and shape of formed conventional spray-freezing into the vapor pro-
particles. This polymorph exhibited improved cesses. The feed solution is atomized through a
drug delivery characteristics in a metered dose nozzle positioned at a distance above the boil-
inhaler (MDI) formulation compared to conven- ing refrigerant. The droplets gradually solidify
tional and micronized drugs.[83,84] while passing through the cold halocarbon vapor,
Rapid Expansion from Liquified-Gas and freeze completely as contact is made with
Solution. Young et al. developed a process the boiling refrigerant liquid. Unfortunately, this
based on supercritical fluids, rapid expansion process may result in broad particle size distri-
from the supercritical to the aqueous solution butions and nonmicronized particles because ag-
(RESAS). This technique[85] could produce glomerates of the solution droplets are solidified
stable nanosuspensions of poorly water-soluble while passing through the vapor phase and settle
drugs. The principle of this process is to onto the surface of the cryogenic liquid.[84,89]
induce rapid nucleation of in supercritical Spray-Freezing into Liquid (SFL) Process.
fluids dissolved drugs in the presence of To overcome agglomerated particles, spray-
surface-modifying agents resulting in particles freezing into liquid was developed and patented
formulation with a desirable size distribution in by The University of Texas at Austin in 2003[90]
a very short time. The surface-modifying agents and commercialized by the Dow Chemical Com-
serve to stabilize the formed small particles pany. The SFL process utilizes the atomiza-
and suppress any tendency towards particle tion of feed solution containing drugs directly
agglomeration or particle growth while they are into a cryogenic liquid to produce frozen nano-
being formed. The rapid intimate contact with structured particles. The frozen particles are then
the surface modifier is achieved by having the lyophilized to obtain dry, freely flowing mi-
surface modifiers dissolved in the supercritical cronized powders. The advantages of the SFL
fluid containing the dissolved drugs. A rapid in- process result from intense atomization and
timate contact between the surface modifier and rapid freezing rates. A high degree of atom-
the newly formed particles inhibits the crystal ization is achieved by spraying directly into
growth of the newly formed particle.[84,86] cryogenic liquid and the ultrarapid freezing
This technique successfully produced cy- rates prevent the phase separation of solutes
closporine with a size of 500–700 nm. Cy- within the feed solution and induce formation of
closporine nanocrystals could be stabilized amorphous structures. The intense atomization
at drug concentrations as high as 6.2 and and ultrarapid freezing rates lead to amorphous
37.5 mg/mL in 1.0% and 5.0% (w/w) Tween nanostructures with enhanced wetting and sig-
80 solution, respectively.[84,87] nificantly enhanced dissolution rates.[84,90]
B124 INTERNATIONAL JOURNAL OF NANOTECHNOLOGY: BIOMEDICINE

Solvent Evaporation Process-Evaporative tency of the nanosuspension technique, reducing


R
Precipitation into Aqueous Solution Process. drug load from 22.5 to 7.5 mg/kg (Wellvone ),
The evaporative precipitation into aqueous so- but increasing activity 2.5-fold at the same
lution (EPAS) applies rapid phase separation to time.[72] Buparvaquone nanosuspensions pro-
nucleate and grow nanoparticles and micropar- vide some advantages, and in T-cell receptor
ticles of poorly water-soluble drugs. The EPAS (TCR)-α–deficient mice infected with Cryp-
was developed and patented by the University tosporidium parvum oocysts, the significance
of Texas at Austin in 2001 and commercialized of the nanosuspensions is documented. In com-
by the Dow Chemical Company.[84] A drug so- parison to micronized drug powder, the infectiv-
lution in organic solvent is pumped through a ity score was reduced from 2.0 (negative con-
tube where it is heated under pressure and then trol group, untreated) to 1.47 for micronized
spraying through a nozzle into heated aqueous buparvaquone and even to 1.02 for equivalent
solution. This process results in an amorphous nanosuspensions.[72,94] Despite the substantial
suspension. The stable aqueous drug suspension amount of literature data in support of nanopar-
is dried by lyophilization or spray drying. A ticles formulations, especially for BCS Class II
variety of hydrophilic surfactants are added to compounds, there are relatively limited number
the solution to diffuse to surface of the growing of current drug products in the market that utilize
particles rapidly enough to prevent growth of nanoparticles for oral application (Table 3).
particles. The EPAS technique has produced cy- Furthermore, the nanosuspension was able to
closporin A nanosupensions with particle a size overcome the significant food effect (∼3.2×)
ranging from 130 to 460 nm.[84,91] observed with the microsuspension formulation.
It is worth mentioning that, in the dog model,
Drug Nanosuspension Administration 
R
the Nanocrystal suspension (120 nm) signifi-
Oral Administration of the Nanosuspensions cantly outperformed a suspension of wet-milled
drugs with a size of 480 nm in terms of fasted
First choice of application is oral admin- state exposure. The results from the dog model
istration. When a drug is given orally, the were confirmed in the clinic, with the EMEND
bioavailability and finally its efficacy depends formulation exhibiting no food effect. It is also
on the solubility and absorption in the gastroin- worth mentioning that a prepitant is classified
testinal tract. In vitro, active compounds have as a BCS Class II and IV compound, empha-
failed in the past because their poor solubility sizing that nanonizing is not only confined to
has limited in vivo absorption and did not high-permeability compounds but may also be
lead to effective therapeutic concentrations. applicable to low-permeability drugs, provided
Simple examples reflecting this problem of poor dissolution is the slower step in the absorption
solubility combined with low absorption are process.[93,95]
the experimental compounds buparvaquone and
atovaquone. A way to improve bioavailability Parenteral Nanosuspension—Intravenous
of atovaquone or buparvaquone would be to Administration Route
increase the absorption rate by formulating
them as a nanosuspension. Oral administration Administration of drugs via the parenteral
of nanosuspensions can overcome this problem route is critical, i.e., usually accompanied by
because of the high adhesiveness of drug parti- physical and biological problems, such as pro-
cles on biological surfaces—here the epithelial duction under aseptic conditions, a sophisticated
gut wall—and prolonging the absorption time. protocol for safety issues, last but not least,
In comparison to Wellvone-treated mice, con- biological problems, such as endotoxins, aller-
taining a micronized drug, nanosuspensions of gic reactions, and inconvenience for the patient.
atovaquone at equivalent doses reduced infec- Formulation of atovaquone as a nanosuspen-
tivity from 40% to 15% at a reduced drug sion for intravenous (i.v.) use to treat murine
concentration of only 7.5 mg/kg after oral toxoplasmosis showed a significant reduction
administration.[72] These results reflects the po- of Toxoplasma gondii at a dose of 0.3 mg/kg
R. Ravichandran B125

TABLE 3. Overview of current state of development of drugs using nanotechnology[3,50,92,93]

Tradename Drug Indication Applied technology Company Status


R 
R
Rapamune Rapamycin Immunesuppressive Elan Nanocrystal Wyeth Marketed

R 
R
Emend Aprepitant Antiemetic Elan Nanocrystal Merck Marketed

R 
R
Tricor Fenofibrate Hypercholesterolemia Elan Nanocrystal Abbott Marketed

R 
R
Megace ES Megestrol Antianorexic Elan Nanocrystal Par Pharmaceutical Marketed
Companies

R 
R
Avinza Morphine sulfate Pain relief Elan NanoCrystals King Marketed
Pharmaceutical

R 
R
Focalin XR Dexmethylphenidate Immediate and a Elan NanoCrystals Novartis Marketed
hydrochloride second delayed
release CNS
stimulant

R 
R
Ritalin LA Methylphenidate Extended-release CNS Elan NanoCrystals Novartis Marketed
hydrochloride stimulant

R
Zanaflex Tizanidine centrally acting Elan NanoCrystals Acorda Marketed
CapsulesTM hydrochloride α2 -adrenergic
agonist

R 
R
Triglide Fenofibrate Hypercholesterolemia IDD-P Skyepharma Sciele Pharma Inc. Marketed

R
Semapimod Guanylhydrazone TNF-α inhibitor Own Cytokine Phase II
Pharmasciences

R
Paxceed Paclitaxel Anti-inflammatory Unknown Angiotech Phase III
Pharmaceuticals
Inc

R 
R
Theralux Thymectacin Anticancer Nanocrystal élan Celmed Phase II
BioSciences Inc

R
Nucryst Silver Antibacterial Own Nucryst Phase II
Pharmaceuticals

in comparison to 30 mg/kg when given EL was not used in the formulation).[96] Other
orally.[72] investigators reported that particle size played

R
The nanoparticle-based product Abraxane an important role in pharmacokinetics and tissue
was approved by the Food and Drug Adminis- distribution of oridonin nanosuspension.[97] The
tration (FDA) in 2006 for intravenous adminis- pharmacokinetics and tissue distribution of ori-

R
tration. Abraxane is a novel formulation con- donin nanosuspensions A (particle size of about
sisting of lyophilized particles with 10% (w/w) 100 nm) and nanosuspension B (particle size of
paclitaxel and 90% (w/w) albumin. The particle about 900 nm) were studied after intravenous
size of the suspension is about 130 nm. In a Phase administration using New Zealand rabbits and
I trial, 39 patients with advanced nonhemato- Kunming mice as experimental animals, respec-
logic malignancies were treated at dose levels tively. An Oridonin control solution was studied
from 80 to 200 mg/mL in multiple cycles of in parallel. The results showed that oridonin
weekly 30-min intravenous infusions. In contrast nanosuspension A exhibited pharmacokinetic
to Taxol treatment, no premedication was used and biodistribution properties similar to the
in this study. The maximum tolerated dose ob- solution due to its rapid dissolution in the
served from this study was higher than the com- blood circulation. Oridonin nanosuspension B,

R
mercial Taxol formulation. Furthermore, this however, showed a high uptake in reticuloen-
study confirmed that the nanoparticle formula- dothelial system (RES) organs, thus exhibited
tion of paclitaxel eliminates the need for premed- a markedly different pharmacokinetic property
ication (because the toxic excipient Cremophor compared to nanosuspension A.[97]
B126 INTERNATIONAL JOURNAL OF NANOTECHNOLOGY: BIOMEDICINE

Pulmonary Drug Delivery via complex formation with sucroseerucate. The


resultant nanosuspension increased the perme-
Many water-insoluble drugs were delivered to ability flux of diclofenac sodium across the skin
the respiratory tract for local or systemic treat- by up to 3.8-fold compared to the control. The
ment of diseases. Unfortunately, many of these optimal weight ratio for the highest diclofenac
drugs are poorly soluble in aqueous solution sodium permeation was 8.8, at which point
and simultaneously insoluble in nonaqueous me- the mean diameter of the nanosuspension was
dia. A number of years ago chlorofluorocarbon 14.4 nm.[100]
(CFC) aerosols was the best choice as drugs de-
livery for respiratory tract. But in compliance
with the Montreal protocol of 1987, the use of Ocular Delivery of Nanosuspensions
chlorofluorocarbons (CFCs) must be avoided. Drug nanosuspensions for ocular drug deliv-
Therefore, as alternative drugs delivery without ery system has been developed by Pignatello
CFCs, metered dose inhalers (MDIs) or dry pow- et al.[101−104] The codispersion of cloricromene-
der inhalers (DPIs) were developed. The nebu- hydrochloride (AD6) in Eudragit RS or RL poly-
lized nanosuspensions produce aerosol droplets mers resulted in nanosuspensions that showed
loaded with a large number of drugs nanocrys- good mean sizes for ophthalmic applications and
tals. The respirable fraction is distinctly in- a positive surface charge. The suspensions al-
creased using nebulized nanosuspensions com- lowed for improved corneal adhesion and sta-
pared to nebulized microcrystals (conventional bility upon storage, particularly at low temper-
MDIs). The smaller particle the size of the drugs atures. When preparation was performed in an
nanocrystals, the more droplets are loaded with isotonic saline solution, the dispersion of AD6
drug. In addition, the muco-adhesive property in the polymer network protected the ester drug
of nanoparticles leads to a prolonged residence from the hydrolytic cleavage into the inactive
time at mucosal surface of the lung.[76] and insoluble acid form. According to prelim-
Yang et al. reported that nanosuspensions inary biological evaluation of the nanosuspen-
of fluctasone exhibited good physical/chemical sions that showed a higher drug availability in
properties for pulmonary delivery. The phar- the rabbit aqueous humor after the drug’s ad-
macokinetic studies after the intra tracheal ministration in Eudragit RL nanosuspensions,
administration of nanosuspensions showed deep AD6-loaded Eudragit Retard nanosuspensions
lung deposition and fast lung absorption, with appear to offer a promising means of improving
solubility playing an important role in lung re- the shelf life and bioavailability of this drug after
tention and duration of action. Overall, these ophthalmic application.[101]
studies have demonstrated that nanosuspen- Nanosuspension as an ophthalmic delivery
sions can be used for pulmonary drug deliv- system has been also investigated by Kassem
ery in preclinical animal studies.[98] Superi- et al.[105] The effect of particle size in the mi-
ority of the drug nanosusupension was also cron and nano size ranges as well as the effect
shown by Hernandez-Kirstein using bupar- of viscosity of the nanosuspensions on the oc-
vaquone nanosuspensions for pulmonary deliv- ular bioavailability was studied by measuring
ery system.[99] the intraocular pressure of normotensive Albino
rabbits using a tonometer. The results show that
Transdermal Delivery of Nanosuspensions compared to solution and microcrystalline sus-
pensions, it is a common feature of the three
It has been reported to use diclofenac sodium drugs that the nanosuspensions always enhance
nanosuspension for trans dermal delivery. The the rate and extent of ophthalmic drug absorp-
basic trans dermal characteristics of the nanosus- tion as well as the intensity of drug action. In the
pension were evaluated using a Yucatan mi- majority of cases, nanosuspensions extend the
cropig (YMP) skin model. Diclofenac sodium duration of the drug effect to a significant extent.
nanosuspension was successfully dispersed into The data presented confirm that nanosuspen-
isopropylmyristate as a nanosized suspension sions differ from microcrystalline suspensions
R. Ravichandran B127

and solutions as ophthalmic drug delivery sys- drugs in—preferentially—one single tablet. In
tems and that the differences are statistically the future, more drugs will be poorly soluble
highly to very highly significant. The results con- and thus require smart formulation technologies
firm also the importance of viscosity of nanosus- to make them soluble and bioavailable. An in-
pensions especially in increasing the duration of creased awareness in patients not willing to suf-
drug action.[105] fer from unnecessary side effects will lead to
an increased number of products using nanosus-
pensions to reduce these risks. By modifying
CONCLUSION the nanocrystal surface it is possible to achieve
a prolonged or a targeted release. This will be an
As the examples have shown, nanosuspen- important part of the work for the nanoparticles
sion technology offers great benefits with only a in future.
few minor drawbacks. It is clearly ideally suited
for drugs with solubility problems. Particle size
diminution and the resulting increase in parti- REFERENCES
cle surface, curvature, saturation solubility, and
consequently the increased dissolution velocity 1. Nielloud, F.; Marti-Mestres, G. Drugs and the Phar-
are important factors. Solubility enhancement maceutical Sciences, 2nd ed.; Informa Healthcare: New
alone is not the only important factor. It be- York, 2008.
comes even more important when a drug has 2. Gao, L.; Zhang, D.; Chen, M. Drug nanocrystals for
a narrow therapeutic window where it can be the formulation of poorly soluble drugs and its application
absorbed. In these cases, the increased solubil- as a potential drug delivery system. J. Nanoparticle Res.
2008, 10, 845–862.
ity and dissolution velocity lead to an acceptable
3. Ravichandran, R. Nanotechnology based drug de-
bioavailability. In addition, the nanosuspension livery systems, NanoBiotechnology 2009; in press. DOI:
technology enables formulations to be devel- 10.1007/s12030-009-9028-2. http://www.springerlink.com/
oped without the need of problematic surfac- content/j6440717w6232125.
tants (e.g., Cremophor EL), which may cause 4. Liu, R. Water-Insoluble Drug Formulation: Pharma-
enhanced side effects or adverse reactions. Fur- ceutical Emulsions and Suspensions, 2nd ed.; CRC Press:
thermore, nanosuspensions allow for a fast ac- Baco Raton, FL, 2008.
5. Rasenack, N.; Steckel, H.; Muller, B. W. Microniza-
tion onset, as the drug is absorbed quickly due
tion of anti-inflammatory drugs for pulmonary delivery by
to the fast dissolution of the nanoparticles. This a controlled crystallization process. J. Pharm. Sci. 2003,
is an advantage, especially for drugs that need 92, 35–44.
to work fast (e.g., naproxen for headache re- 6. Steckel, H., Rasenack, N.; Muller, B. W. In-situ-
lief). The enhanced solubility also leads to an micronization of disodium cromoglycate for pulmonary
identical or very similar absorption in fed and delivery. Eur. J. Pharm. Biopharm. 2003, 55, 173–180.
fasted conditions. Drugs that normally require 7. Steckel, H.; et al. In vitro characterization of
food to become soluble will be bioequivalent as jet-milled and in-situmicronizedfluticasone-17-propionate.
Int. J. Pharm. 2003, 258, 65–75.
nanosuspensions in fed and fasted states. If it 8. Rasenack, N.; Muller, B. W. Micron-size drug parti-
is necessary to give a large dosage in order to cles: common and novel micronization techniques. Pharm.
achieve reasonable blood levels for poorly solu- Dev. Technol. 2004, 9, 1–13.
ble drugs resulting in increased side effects, the 9. Liversidge, G. G.; et al. Surface modified drugs
nanosuspension technology allows for smaller nanoparticles; US Patent 5,145,684; Sterling Drug, New
doses and thus decreased side effects. Disadvan- York; USA, 1992.
tages are the sometimes long production times, 10. Muller, R. H.; et al. Pharmaceutical nanosuspensions
for medical administration as systems with increased satu-
e.g., in pearl milling. However, new patents to
ration solubility and rate of solution; US Patent 5,858,410;
accelerate large scale production have just been USA, 1999.
filed (e.g., H42 Müller and Moeschwitzer 2005). 11. Ostrander, K. D., Bosch, H. W.; Bondanza, D. M.
In addition, improved technologies are required An in-vitro assessment of a NanoCrystal beclomethasone
to be able to produce tablets with high drug dipropionate colloidal dispersion via ultrasonic nebuliza-
nanosuspension loads to formulate high-dose tion. Eur. J. Pharm. Biopharm. 1999, 48, 207–215.
B128 INTERNATIONAL JOURNAL OF NANOTECHNOLOGY: BIOMEDICINE

12. Sri, K. V.; et al. Preparation and characterization of 29. Suri, S. S.; Fenniri, H.; Singh, B. Nanotechnology-
quercetin and rutin cyclodextrin inclusion complexes. Drug based drug delivery systems. J Occup. Med. Toxicol. 2007,
Dev. Ind. Pharm. 2007, 33, 245–253. 1, 2–16.
13. Calabro, M. L.; et al. The rutin/beta-cyclodextrin 30. Kirupakar, B. R.; Nanosuspension drug delivery:
interactions in fully aqueous solution: spectroscopic studies Technology and application.Express Pharma Pulse, 2009,
and biological assays. J. Pharm. Biomed. Anal. 2005, 36, pp. 1–6. http://www.expresspharmaonline.com./20050224/
1019–1027. nanotechol.shtml
14. Brewster, M. E.; Loftsson, T. Cyclodextrins as phar- 31. Chingunpituk, J. Nanosuspension technology for
maceutical solubilizers. Adv. Drug Deliv. Rev. 2007, 59, drug delivery. Walailak. J. Sci, Tech. 2007, 4, 139–153.
645–666. 32. US Pharmacopoeia Conventional. The United States
15. Loftsson, T.; et al. Cyclodextrins in drug delivery. Pharmacopoeia, 24th rev. ed; Rockville: The United States
Expert Opin. Drug Deliv. 2005, 2, 335–351. Pharmacopoeia Conventional Inc.: Rockville, MD, 2000,
16. Loftsson, T.; Brewster, M. E. Pharmaceutical appli- p. 8.
cations of cyclodextrins. 1. Drug solubilization and stabi- 33. Remington, J. P. Remington’s Pharmaceutical Sci-
lization. J. Pharm. Sci. 1996, 85, 1017–1025. ences, 16th ed.; Mack: Easton, PA, 1980, pp. 176–178.
17. Loftsson, T.; Brewster, M. E.; Másson, M. Role of 34. Mosharraf, M.; Sebhatu, T.; Nystrom, C. The effects
cyclodextrins in improving oral drug delivery. Am. J. Drug. of disordered structure on the solubility and dissolution
Deliv. 2004, 2, 1–15. rates of some hydrophilic, sparingly soluble drugs. Int. J.
18. Mu, X.; Zhong, Z. Preparation and properties of Pharm. 1999, 177, 29–51.
poly(vinyl alcohol)-stabilized liposomes. Int. J. Pharm. 35. Mosharraf, M.; Nystrom, C. The effect of dry mixing
2006, 318, 55–61. on the apparent solubility of hydrophobic, sparingly soluble
19. Johnston, M. J.; et al. Characterization of the drug drugs. Eur. J. Pharm. Sci. 1999, 9, 145–156.
retention and pharmacokinetic properties of liposomal 36. Martin, A.; J. Swarbrick, Cammarata, A. Physi-
nanoparticles containing dihydrosphingomyelin. Biochim. cal Pharmacy: Physical Chemical Principles in the Phar-
Biophys. Acta 2007, 1768, 1121–1127. maceutical Sciences, 4th subed.; Philadelphia: Lippincott
20. Kreuter, A.; et al. Liposomal pegylated doxorubicin Williams & Wilkins: Philadelphia, 1993, pp. 125–142,
versus low-dose recombinant interferon Alfa-2a in the treat- 212–250, 329–334.
ment of advanced classic Kaposi’s sarcoma; retrospective 37. Myrdal, P. B.; Yalkowsky, S. H. Solubilization of
analysis of three German centers. Cancer Invest. 2005, 23, Drugs in Aqueous Media, 2nd ed., Encyclopedia of Phar-
653–659. maceutical Technology, Vol. 3; Swarbick, J.; Boylan, J. C.,
21. Dannenfelser, R. M.; et al. Development of clinical Eds.; Marcel Dekker: New York, 2002, pp. 2458–2580.
dosage forms for a poorly water soluble drug I: Applica- 38. Wells, J. I. Pharmaceutical Preformulation: The
tion of polyethylene glycolpolysorbate 80 solid dispersion Physicochemical Properties of Drug Substances, 1st ed.;
carrier system. J. Pharm. Sci. 2004, 93, 1165–1175. Chichester: Ellis Horwood: Chichester, 1988, pp. 21–85,
22. Joshi, H. N.; et al. Bioavailability enhancement 94–100.
of a poorly water-soluble drug by solid dispersion in 39. Florence, A. T.; Attwood, D. Physicochemical Prin-
polyethylene glycol-polysorbate 80 mixture. Int. J. Pharm. ciples of Pharmacy, 2nd ed.; Macmillan Publishers: Lon-
2004, 269, 251–258. don, 1981, pp. 89–90,131–172, 199–208.
23. Karavas, E.; et al. Investigation of the release mech- 40. Sekiguchi, K.; Obi, N. Studies on absorption of eu-
anism of a sparingly water-soluble drug from solid dis- tectic mixture. I. A comparison of the behavior of eutectic
persions in hydrophilic carriers based on physical state of mixture of sulfathiazole and that of ordinary sulfathiazole
drug, particle size distribution and drug-polymer interac- in man. Chem. Pharm. Bull. 1961, 9, 866–872.
tions. Eur. J. Pharm. Biopharm. 2007, 66, 334–347. 41. Goldberg, A. H.; Gibaldi, M.; Kanig, J. L. Increasing
24. Overhoff, K. A.; et al. Solid dispersions of itracona- dissolution rates and gastrointestinal absorption of drugs
zole and enteric polymers made by ultra-rapid freezing. Int. via solid solutions and eutectic mixtures II. Experimental
J. Pharm. 2007, 336, 122–132. evaluation of eutectic mixture: urea acetaminophen system.
25. Serajuddin, A. T. Solid dispersion of poorly water- J. Pharm. Sci. 1966, 55, pp. 482–487.
soluble drugs: early promises, subsequent problems, and 42. Hildebrand, J. H.; Scott, R. L. Solubility of Nonelec-
recent breakthroughs. J. Pharm. Sci. 1999, 88, 1058–1066. trolytes, 3rd ed.; Reinhold: New York, 1950, pp. 11–13, 47,
26. Torchilin, V. P. Nanotechnology in Drugs, 2nd ed.; 160, 175–197.
Imperial College Press: London, 2008. 43. Scatchard, G. Equilibria in non-electrolyte solutions
27. Sahoo, S. K.; Labhasetwar, V. Nanotech approaches in relation to the vapor pressure and densities of the com-
to drug delivery and imaging. Drug Discov. Today 2008, 8, ponents. Chem. Rev. 1931, 8, 321–333.
1112–1120. 44. Noyes, A. A.; Whitney, W. R. The rate of solution of
28. Salata, O. V. Applications of nanoparticles in biol- solid substances in their own solutions. J. Am. Chem. Soc.
ogy and medicine. J Nanobiotechnol. 2004, 2, 1–6. 1897, 19, 930–934.
R. Ravichandran B129

45. Hixon, A. W.; Crowell, J. H. Dependence of reaction 61. Sucker, H.; Gassmann, P. Improvements in pharma-
velocity upon surface and agitation. I. Theoretical consid- ceutical compositions; GB Patent 2269536A; Sandoz Ltd.,
eration. Ind. Eng. Chem. 1931, 23, 923–931. Switzerland; UK, 1994.
46. Amidon, G. L.; et al. A theoretical basis for a bio- 62. Liversidge, G. G.; Cundy, K. C. Particle size reduc-
pharmaceutic drug classification: the correlation of in vitro tion for improvement of oral bioavailability of hydropho-
drug product dissolution and in vivo bioavailability. Pharm. bic drugs: I. Absolute oral bioavailability of nanocrys-
Res. 1995, 12, 413–420. talline danazol in beagle dogs. Int. J.Pharm. 1995, 125, 91–
47. http://en.wikipedia.org/wiki/Biopharmaceutics 97.
Classification System 63. Muller, R. H.; Mäder, K.; Krause, K. Verfahren
48. Hite, M.; Turner, S.; Cathy Federici, R. C. Part 1: zur schonenden Herstellung von hochfeinen Micro-
Oral Delivery of Poorly Soluble Drugs. Research and Prod- /Nanopartikeln; PCT Application PCT/EP00/06535; Ger-
R
uct Development Group at SCOLR Pharma Inc.: Both- many, 2000.
well, WA, 2002, pp. 1–3. 64. Kipp, J. E.; et al. Microprecipitation method for
49. Hecq, J.; et al. Preparation and characterization of preparing submicron suspensions; US Patent 6,607,784;
nanocrystals for solubility and dissolution rate enhance- Baxter International Inc., Deerfield, IL; USA, 2003.
ment of nifedipine. Int. J. Pharm. 2005, 299, 167–177. 65. Petersen, R. D. Nanocrystals for use in top-
50. Keck, C. M.; Muller, R. H. Drug nanocrystals of ical fomulations and method of production thereof;
poorly soluble drugs produced by high pressure homogeni- PCT/EP2007/009943; Abbott GmbH; Germany, 2006.
sation. Eur. J. Pharm. Biopharm. 2006, 62, 3–16. 66. Weder, H. G.; Hoogevest, V. Nanosuspensions for
51. Merisko-Liversidge, E.; Liversidge, G. G.; Cooper, intravenous administration; US Patent 5,726,164; USA,
E. R. Nanosizing: a formulation approach for poorly- 1998.
water-soluble compounds. Eur. J. Pharm. Sci. 2003, 18, 67. Muller, R. H.; Benita, S.; Bohm, B. Emulsions
113–120. and Nanosuspensions for the Formulation of Poorly Sol-
52. Muller, R. H.; Böhm, B. H. L.; Grau, M. J. uble Drugs; Stuttgart.: Medpharm Scientific Publishers:
Nanosuspensions—a formulation approach for poorly sol- Stuttgart, 1998, 396 S.
uble and poorly bioavailable drugs. In Handbook of Phar- 68. Muller, R. H.; Möschwitzer, J.; Bushrab, F. N. Man-
maceutical Controlled Release; Wise, D. L., Ed., Marcel ufacturing of nanoparticles by milling and homogenisation
Dekker: New York, 2000, pp. 345–357. techniques. In Nanoparticle Technology for Drug Delivery;
53. Muller, R. H.; Jacobs, C.; Kayser, O. DissoCubes—a Gupta, R. B.; Kompella, U. B., Eds.; Tayler & Francis: New
novel formulation for poorly soluble and poorly bioavail- York, 2006, pp. 21–52.
able drugs. In Modified-Release Drug Delivery Systems; 69. Jinno, J.; et al. Effect of particle size reduction on
Rathbone, M. J.; Hadgraft, J.; Roberts, M. S., Eds.; Marcel dissolution and oral absorption of a poorly water-soluble
Dekker: New York, 2003, pp. 135–149. drug, cilostazol, in beagle dogs. J Control Release, 2006,
54. Muller, R. H.; Akkar, A. Drug nanocrystals of poorly 111, 56–64.
soluble drugs. In Encyclopedia of Nanoscience and Nan- 70. Rao, Y. M., Pavan Kumar, M.; Apte, S. Formulation
otechnology; Nalwa, H. S., Ed. American Scientific Pub- of nanosuspensions of albendazole for oral administration.
lishers: Valencia, CA 2004, pp. 627–638. Curr. Nanosci. 2008, 4, 53–58.
55. Muller, R. H.; Keck, C. M. Improvement of delivery 71. Buckton, G.; Beezer, A. E. The relationship between
& solubility of poorly soluble drugs by nanonisation. In particle size and solubility. Int. J. Pharm. 1992, 82, R7–
Konferenzdokumentation LTS Academy, 2007. R10.
56. Liversidge, G. G.; Conzentino, P. Drug particle size 72. Muller, R. H.; Jacobs, C.; Kayser, O. Nanosuspen-
reduction for decreasing gastric irritancy and enhancing sions as particulate drug formulations in therapy. Rationale
absorption of naproxen in rats. Int. J. Pharm., 1995, 125(2), for development and what we can expect for the future.
309–313. Adv. Drug Deliv. Rev. 2001, 47, 3–19.
57. Muller, R. H.; Jacobs, C.; Kayser, O. Nanosuspen- 73. Mukerjee, P. Thermodynamic aspects of solubil-
sions for the Formulation of Poorly Soluble Drugs, Phar- ity of small particles. J. Pharm. Sci. 1972, 61, 478–
maceutical Emulsions and Suspensions, ed. F. Nielloud and 479.
G. Marti-Mestres, Marcel Dekker, 2000, pp 383–407. 74. Muller, R. H. Nanosuspensionen—eine neue For-
58. Wu, T. H.; et al., Preparation, physicochemical char- mulierung fürschwerlösliche Arzneistoffe. In Phar-
acterization, and antioxidant effects of quercetin nanopar- mazeutische Technologie: Moderne Arzneiformen, 2nd ed.;
ticles. Int. J. Pharm. 2008, 346, 160–168. Müller, R. H.; G. E. Hildebrand, G. E., Eds.; Stuttgart:
59. Auweter, H.; et al., Precipitated water-insoluble col- Wissenschaftliche Verlagsgesellschaft: Stuttgart, 1998, pp.
orants in colloid disperse form; US Patent 6,494,924; BASF 393–400.
Aktiengesellschaft; USA, 2002. 75. Mosharraf, M.; Nystrom, C. The effect of particle
60. List, M.; Sucker, H. Pharmaceutical colloidal hy- size and shape on the surface specific dissolution rate of
drosols for injection; GB Patent 2200048; Sandoz Ltd., microsized practically insoluble drugs. Int. J. Pharm., 1995,
Switzerland; UK, 1988. 122, 35–47.
B130 INTERNATIONAL JOURNAL OF NANOTECHNOLOGY: BIOMEDICINE

76. Moschwitzer, J. Drug Nanocrystals Prepared by 90. Williams, R. O.; et al. Process for production of
High Pressure Homogenisation—The Universal Formula- nanoparticles and microparticles by spray freezing into liq-
tion Approach for Poorly Soluble Drugs; Dissertation; In- uid; US Patent 20030041602; USA, 2003.
stitut für Pharmazeutische Technologie, Freie Universität 91. Chen, X.; et al. Preparation of cyclosporine A
Berlin, 2006. nanoparticles by evaporative precipitation into aqueous so-
77. Muller, R. H.; Peters, K. Nanosuspensions for the lution. Int. J. Pharm. 2002, 242, 3–14.
formulation of poorly soluble drugs: I. Preparation by a 92. Elan, P. Drug technologies nanocrystals—
size-reduction technique. Int. J. Pharmaceut. 1998, 160, technology overview. Presented at the Annual Meeting
229–237. of Controlled Release Sociaty (CRS); CRS: Long Beach,
78. Krause, K. P.; et al. Heavy metal contamination of CA, 2007.
nanosuspensions produced by high-pressure homogenisa- 93. Kesisoglou, F.; Panmai, S.; Wu, Y. Nanosizing–oral
tion. Int. J. Pharm. 2000, 196, 169–172. formulation development and biopharmaceutical evalua-
79. Muller, R. H., Krause, K.; Mader, K. Method for con- tion. Adv. Drug Deliv. Rev., 2007, 59, 631–644.
trolled production of ultrafine microparticles and nanopar- 94. Jacobs, C.; Kayser, O.; Muller, R. H. Production and
ticles; PCT Application PCT/EP2000/006535; Germany, characterization of mucoadhesive nanosuspensions for the
2000. formulation of bupravaquone. Int. J. Pharm. 2001, 214,
80. Bushrab, N., F.; Muller, R. H. Nanocrystals of poorly 3–7.
soluble drugs for oral administration. New Drugs, 2003, 5, 95. Scholer, N.; et al. Atovaquone nanosuspensions
20–22. show excellent therapeutic effect in a new murine
81. Keck, C. M.; Kobierski, S.; Mauludin, R.; Muller, model of reactivated toxoplasmosis. Antimicrobial. Agents
R. H. Second generation of drug nanocrystals for delivery Chemother. 2001, 45, 1771–1779.
of poorly soluble drugs: smartCrystals technology. DOSIS, 96. Wong, J.; et al. Suspensions for intravenous (IV)
2008, 24, 125–130. injection: a review of development, preclinical and clinical
82. Hu, J.; et al. Improvement of dissolution rates aspects. Adv. Drug Deliv. Rev. 2008, 60, 939–954.
of poorly water soluble APIs using novel spray freez- 97. Gao, L.; et al. Studies on pharmacokinetics and tis-
ing into liquid technology. Pharm. Res. 2002, 19, 1278– sue distribution of oridonin nanosuspensions. Int. J. Pharm.
1284. 2008, 355, 321–327.
83. Hanna, M. H.; York, P. Method and apparatus for the 98. Yang, J. Z.; et al. Fluticasone and budesonide
formulation of particles; US Patent 5,851,453; USA, 1998. nanosuspensions for pulmonary delivery: preparation, char-
84. Hu, J., Johnston, K. P.; Williams, R. O., 3rd. acterization, and pharmacokinetic studies. J. Pharm. Sci.
Nanoparticle engineering processes for enhancing the dis- 2008, 97, 4869–4878.
solution rates of poorly water soluble drugs. Drug. Dev. 99. Hernandez-Trejo, N.; et al. Characterization of neb-
Ind. Pharm. 2004, 30, 233–245. ulized buparvaquonenano suspensions—effect of nebuliza-
85. Young, T. J.; et al. Encapsulation of lysozyme in tion technology. J. Drug Target 2005, 13, 499–507.
a biodegradable polymer by precipitation with a vapor- 100. Piao, H.; et al. A novel solid-in-oil nanosuspension
over-liquid antisolvent. J. Pharm. Sci. 1999, 88, 640– for transdermal delivery of diclofenac sodium. Pharm. Res.
650. 2008, 25, 896–901.
86. Pace, G. W.; et al. Process to generate submicron 101. Pignatello, R.; et al. Preparation and characterization
particles of water insoluble compounds; US Patent 6, 103; of eudragit retard nanosuspensions for the ocular delivery of
USA, 2001. cloricromene. AAPS Pharm. Sci. Techol. 2006, 7(1), article
87. Young, T. J.; et al. Rapid expansion from supercriti- 27. http://www.aapspharmscitech.org
cal to aqueous solution to produce submicron suspensions 102. Pignatello, R.; Bucolo, C.; Puglisi, G. Ocular toler-
of water-insoluble drugs. Biotechnol. Prog. 2000, 16, 402– ability of EudragitRS100 and RL100 nanosuspensions as
407. carriers for ophthalmic controlled drug delivery. J. Pharm.
88. Turk, M.; Lietzow, R. Stabilized nanoparticles of Sci. 2002, 91, 2636–2641.
phytosterol by rapid expansion from supercritical solution 103. Pignatello, R.; et al. Eudragit RS100 nanosuspen-
into aqueous solution. AAPS Pharm. Sci. Techol. 2004, 5(4), sions for the ophthalmic controlled delivery of ibuprofen.
article 56. http://www.aapspharmscitech.org/view.asp? Eur. J. Pharm. Sci. 2002, 16, 53–61.
art=pt050456 104. Pignatello, R.; et al. Flurbiprofen-loaded acrylate
89. Rogers, T. L., Johnston, K. P.; Williams, R. O., 3rd. polymer nanosuspensions for ophthalmic application. Bio-
Solution-based particle formation of pharmaceutical pow- materials 2002, 23, 3247–3255.
ders by supercritical or compressed fluid CO2 and cryo- 105. Kassem, M. A.; et al. Nanosuspension as an oph-
genic spray-freezing technologies. Drug Dev. Ind. Pharm. thalmic delivery system for certain glucocorticoid drugs.
2001, 27, 1003–1015. Int. J. Pharm. 2007, 340, 126–133.

You might also like