12.01.2013 Views

Nanocrystals: Current Strategies and Trends - International Journal ...

Nanocrystals: Current Strategies and Trends - International Journal ...

Nanocrystals: Current Strategies and Trends - International Journal ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

<strong>International</strong> <strong>Journal</strong> of Research in Pharmaceutical <strong>and</strong> Biomedical Sciences ISSN: 2229-3701<br />

_________________________________________Research Paper<br />

<strong>Nanocrystals</strong>: <strong>Current</strong> <strong>Strategies</strong> <strong>and</strong> <strong>Trends</strong><br />

Sanjay Bansal 1 *, Meena Bansal 1 <strong>and</strong> Rachna Kumria 2<br />

1 M.C.P. College, Jal<strong>and</strong>har, Punjab, India.<br />

2 MMCP, MMU, Mullana, Ambala, Haryana, India.<br />

___________________________________________________________________________<br />

ABSTRACT<br />

With the advent of modern technologies, a large number of drugs have been discovered which have a better<br />

efficiency but their clinical application is restricted due to poor water solubility. Nearly 40% of the drugs in the<br />

pipeline <strong>and</strong> around 60% of compounds coming directly from synthesis have poor solubility. Poor water<br />

solubility has become a leading challenge for the formulation of these compounds. Poor solubility is generally<br />

associated with poor bioavailability. <strong>Nanocrystals</strong> have the potential to overcome this issue. Change of<br />

materials into the nanodimension dramatically changes its physical properties. Drug nanocrystals are crystals<br />

with a size in the nanometer range (mean diameter < 1000nm). This review article outlines the various<br />

pharmaceutical advantages of nanosization, industrially relevant production technologies available currently<br />

with advantages <strong>and</strong> disadvantages of each <strong>and</strong> the various dosage forms developed using nanocrystals. The<br />

nanocrystal products in the market/pipeline will be briefly reviewed <strong>and</strong> the advantages of these as compared<br />

to traditional products would be highlighted.<br />

Key Words: <strong>Nanocrystals</strong>, high pressure homogenization, pearl milling, drug nanocrystals, dissocubes.<br />

1. INTRODUCTION<br />

With progress in high throughput screening<br />

methods <strong>and</strong> also with 60% of the drugs coming<br />

directly from synthesis the number of poorly<br />

soluble drugs is increasing 1 . Research efforts are<br />

being focused on the approaches to increase drug<br />

solubility e.g. solubilisation using surfactants,<br />

formation of micro emulsions , complex formation,<br />

formation of self emulsifying drug delivery system<br />

(SEDDS), solid dispersions, etc. Micronization of<br />

drug powders to size between 1-10 μm, in order to<br />

increase surface area <strong>and</strong> dissolution rate is not<br />

sufficient to overcome bioavailability of BCS Class<br />

II drugs. The next step towards solubilization is<br />

nanonization. <strong>Nanocrystals</strong> are crystals having size<br />

less than 1μm . As the particle size of a crystal is<br />

decreased to about 100 nm there is a drastic change<br />

in the properties of the material. The decreased size<br />

increases the surface area <strong>and</strong> solubility of drug<br />

manifolds <strong>and</strong> there is proportionate increase in the<br />

bioavailability of poorly soluble drugs.<br />

Nanonization has an additional effect when<br />

compared to micronisation. It increases not only<br />

the surface area, but also simultaneously the<br />

saturation solubility. The solubility of normally<br />

sized powders is a compound specific constant,<br />

depending only on the temperature <strong>and</strong> the solvent.<br />

However, when the particle size of a crystal is less<br />

than 1-2 μm, the saturation solubility is also a<br />

function of particle size. The dissolution pressure<br />

increases due to the strong curvature of the<br />

particles leading to an increase in saturation<br />

solubility. The theoretical background is provided<br />

by the Ostwald–Freundlich <strong>and</strong> the Kelvin<br />

equations 2 .<br />

The increase in saturation solubility has two<br />

effects:<br />

a) An increase in saturation solubility leading to an<br />

increase in dissolution rate.<br />

b) Formation of a supersaturated solution which<br />

in-turn increases the concentration gradient<br />

between the lumen of the gut <strong>and</strong> the blood. This<br />

would hasten drug diffusion promoting absorption.<br />

<strong>Nanocrystals</strong> offer a quick action onset due to<br />

faster dissolution <strong>and</strong> rapid absorption. This is<br />

advantageous particularly for drugs where a quick<br />

action is desired e.g. naproxen for relief of<br />

headache. The bio-availability of various drugs has<br />

been found to increase significantly when<br />

administered in the form of nanocrystals. A study<br />

was conducted by Liversidge <strong>and</strong> Conzentino<br />

(1995) 3 on the bioavailability of naproxen, when<br />

naproxen was administered as a conventional<br />

tablet, a suspension <strong>and</strong> as a nanosuspension. It<br />

was found that the area under the curve (AUC) of<br />

blood levels for analgesic naproxen was 32.7 mgh/l<br />

for conventional tablet, 44.7 mgh/l as Naprosyn ®<br />

suspension <strong>and</strong> the AUC increased to 79.5 mgh/l<br />

when naproxen was administered as<br />

nanosuspensions. Another study was conducted on<br />

the gonadotropin inhibitor danazol when<br />

administered as macrosuspension <strong>and</strong> as a<br />

Vol. 3 (1) Jan – Mar 2012 www.ijrpbsonline.com 406


<strong>International</strong> <strong>Journal</strong> of Research in Pharmaceutical <strong>and</strong> Biomedical Sciences ISSN: 2229-3701<br />

nanosuspension. The relative bio-availability was<br />

found to be 5.1% when danazol was administration<br />

as a microemulsion. Bioavailability of Danazol<br />

nanosuspension on the other side was found to be<br />

82.3% 4 . Due to their small size, nanosuspension<br />

can be injected intravenously leading to a 100%<br />

bio-availability. Thus any drug can be made 100%<br />

bio-available using the nanocrystal technology.<br />

<strong>Nanocrystals</strong> can show a strong adhesion because<br />

of the increased contact area for van der Waals<br />

attraction. The adhesiveness of the nanoparticles to<br />

the gut wall after oral administration enhances<br />

absorption <strong>and</strong> thereby increases the<br />

bioavailability. Lamprecht (2001) 5 observed<br />

differential uptake/ adhesion of polystyrene particle<br />

to inflamed colonic mucosa, with the deposition<br />

5.2%, 9.1%, <strong>and</strong> 14.5% for 10-mm, 1000-nm, <strong>and</strong><br />

100-nm particles, respectively. The behavior of<br />

polymeric nanoparticles in GIT is influenced by<br />

their bio-adhesive properties. Therefore, lectins<br />

have been shown to improve muco-adhesion of the<br />

drug 6 .<br />

<strong>Nanocrystals</strong> may be able to reduce the dose to be<br />

administered, provide a sustained drug release <strong>and</strong><br />

increase patient compliance. de Waard et al.<br />

(2010) 7 prepared nanocrystals of ibuprofen <strong>and</strong><br />

fenofibrate. He claimed that shape of the crystals<br />

increases the drug absorption to a great extent. So,<br />

Waard claims that doses of poorly soluble drugs<br />

such as ibuprofen <strong>and</strong> cholesterol reducing<br />

fenofibrate can be reduced if they are administered<br />

in nanocrystal form. The increased bio-availability<br />

leads to reduction in dosing frequency which may<br />

improve patient compliance. P<strong>and</strong>ey et al. (2003) 8<br />

demonstrated that the nanoparticles provided<br />

sustained release of the anti-tubercular drugs<br />

(rifampin, isoniazid <strong>and</strong> pyrazinamide) <strong>and</strong><br />

considerably enhanced their efficacy after oral<br />

administration. This solves the problem of patient<br />

non-compliance <strong>and</strong> thus reduces the incidence of<br />

relapse of the disease. Shegokar et al. (2010) 9<br />

prepared prolonged release formulation for dermal<br />

use incorporating nanocrystals of poorly water<br />

soluble lidocaine. <strong>Nanocrystals</strong> can be incorporated<br />

in various dosage forms which make administration<br />

by various routes feasible. Due to better solubility<br />

<strong>and</strong> bioavailability, nanocrystals can be supplied in<br />

patient friendly oral solid dosage forms such as<br />

tablets <strong>and</strong> capsules. <strong>Nanocrystals</strong> of poorly soluble<br />

drugs can also be incorporated in cosmetic products<br />

where they provide high penetration power through<br />

dermal application. A very small size of<br />

nanoparticles (200-400 nm) even smaller than the<br />

size of the smallest blood capillaries allows the<br />

nanosuspensions to be injected intravenously. This<br />

provides 100% bio-availability <strong>and</strong> simultaneously<br />

avoids the use of toxic surfactants or co solvents to<br />

dissolve the drug. Pulmonary <strong>and</strong> ophthalmic drug<br />

delivery of nanocrystals has also been achieved<br />

with better efficiency. Nanosuspensions can be<br />

used for targeted delivery because their surface<br />

properties <strong>and</strong> changing of the stabilizer can easily<br />

alter in-vivo behavior. Nanosuspensions afford a<br />

means of administering poorly soluble drugs to<br />

brain with decreased side effects. Different<br />

approaches can be used to target drugs to the site of<br />

action. One method is direct injection of<br />

nanoparticles at the target site e.g. injection into<br />

cancer tissue.<br />

The drug nanocrystals are a smart delivery system,<br />

a universal principle, which can be applied to any<br />

drug because any drug can be diminuted to<br />

nanocrystals. Furthermore, both lipophilic <strong>and</strong><br />

hydrophilic drugs can be incorporated as<br />

nanocrystals. Another essential prerequisite for<br />

entry to the pharmaceutical market is the<br />

availability of large scale production methods at<br />

sufficiently low cost <strong>and</strong> simultaneously meeting<br />

the regulatory requirements. The nanocrystals<br />

technology fulfills this criterion also. The pearl<br />

milling <strong>and</strong> high pressure homogenization can be<br />

extended for commercial production of<br />

nanocrystals <strong>and</strong> are accepted by the regulatory<br />

authorities.<br />

2. Nanocrystal technology<br />

Drug nanocrystals can be produced by bottom up<br />

techniques (precipitation methods) or top down<br />

techniques (size reduction by milling or high<br />

pressure homogenization). In case of bottom-up<br />

technologies, one starts with molecules in the<br />

solution <strong>and</strong> moves via association of these<br />

molecules to form solid particles, i.e. it is a<br />

classical precipitation process. The top down<br />

techniques are based on size reduction of relatively<br />

large particles into smaller particles by mechanical<br />

attrition. For industrial production, all products are<br />

prepared by top down technique. The basic<br />

techniques currently used by different companies<br />

are:<br />

2.1.Bottom-up technique (Precipitation method)<br />

2.2 Top down techniques<br />

2.2.1. Pearl/Ball milling (Nanosystems /Élan<br />

technology)<br />

Vol. 3 (1) Jan – Mar 2012 www.ijrpbsonline.com 407<br />

4, 10<br />

2.2.2 High Pressure Homogenization (HPH)<br />

2.2.2.1 Micro fluidizer technology (IDD-P TM<br />

technology)<br />

2.2.2.2 Piston gap homogenization in water<br />

(Dissocubes® technology)<br />

2.2.2.3 Piston gap homogenization in water<br />

mixtures or in non-aqueous medium<br />

(Nanopure® technology) 11-15<br />

2.2.3 Combination technology<br />

2.2.3.1. NANOEDGE® Technology<br />

2.2.3.2. SmartCrystal® Technology<br />

2.1 Bottom-up technique (Precipitation<br />

method): This is also known as hydrosol<br />

technology. This was developed by Sucker <strong>and</strong> the<br />

intellectual property is owned by S<strong>and</strong>oz


<strong>International</strong> <strong>Journal</strong> of Research in Pharmaceutical <strong>and</strong> Biomedical Sciences ISSN: 2229-3701<br />

(nowadays Novartis) 16, 17 . In this technique the drug<br />

is dissolved in a solvent <strong>and</strong> then this solution is<br />

added to a non solvent leading to the precipitation<br />

of the finely dispersed drug nanocrystals. The<br />

precipitation technique is simple <strong>and</strong> requires low<br />

cost equipments. For example, the solvent can be<br />

poured into the non-solvent with a constant<br />

velocity in the presence of a high-speed stirrer.<br />

Main approaches include the use of static mixers or<br />

micro-mixers, which simulate the precipitation<br />

conditions in a small volume (i.e., simulating labscale<br />

conditions). In the case of micro-mixers,<br />

scaling up can be performed in a simple way by<br />

arranging many micro-mixers in parallel. This<br />

equipment is relatively simple <strong>and</strong> of relatively low<br />

cost (this is not necessarily valid for the micromixers).<br />

The drawbacks of this technique are that the drug<br />

needs to be soluble in at least one solvent. This<br />

however, is problematic for newly developed drugs<br />

which are generally insoluble in both aqueous <strong>and</strong><br />

organic media. Secondly, this solvent needs to be<br />

miscible with at least one non solvent. Solvent<br />

residues need to be removed, thus increasing<br />

production costs.<br />

In case of nanocrystals, care needs to be exercised<br />

to ensure that the crystals do not grow in size <strong>and</strong><br />

remain stabilized at the nanosize. Spray drying <strong>and</strong><br />

lyophilization are the techniques recommended to<br />

preserve the particle size in nano range 18 . Another<br />

alternative to preserve the size of nanocrystals is<br />

the use of polymeric growth inhibitors. Various<br />

stabilizers like sodium dodecyl sulfate (SDS),<br />

polyvinyl alcohol (PVA), tween® 80 <strong>and</strong><br />

polyxamer® 188 have been employed to prepare<br />

nanocrystals 19 .<br />

Nanomorphs: Another precipitation method has<br />

been developed by Soliqs/Abbott, to enhance<br />

dissolution rate <strong>and</strong> solubility. Carotene<br />

nanoparticles were developed for food industry<br />

e.g., Leucarotin® or Lucantin® (BASF) 20 . For<br />

preparation of these, a solution of the Carotenoid<br />

<strong>and</strong> surfactant in digestible oil was mixed with a<br />

suitable aqueous solvent. To this a protective<br />

colloid was added. Carotenoid was stabilized <strong>and</strong><br />

localized in the oily phase of this o/w emulsion.<br />

This emulsion was then lyophilized. X-ray analysis<br />

of the lyophilized product revealed that about 90%<br />

of the carotenoid was in the amorphous state. These<br />

particles were called nanomorphs (Nanomorph®).<br />

These were found to have higher saturation<br />

solubility when compared to crystalline material.<br />

At present, there is no pharmaceutical product in<br />

the market based on this technology.<br />

2.2 Top-down techniques<br />

2.2.1 Pearl/Ball milling: In this technique, the<br />

drug along with the milling media, dispersion<br />

media (generally water) <strong>and</strong> the stabilizer is fed<br />

into the milling chamber. Milling balls or small<br />

pearls are used as milling media. The movement of<br />

milling media generates high shear forces <strong>and</strong><br />

forces of impact which leads to particle size<br />

reduction. This technology was developed by<br />

Merisko-Liversidge et al. (2003) 21 . The pearls or<br />

balls comprise of ceramic (cerium or yttrium<br />

stabilized zirconium dioxide), glass, stainless steel<br />

or highly cross-linked polystyrene resin coated<br />

beads. The two basic principles of milling are<br />

employed. Either the milling material can be<br />

moved by an agitator or the complete container<br />

may be moved in a complex movement. In the<br />

latter method large batches are difficult to process,<br />

so mills using agitators are generally preferred for<br />

large batches. Milling time, however, depends upon<br />

various factors such as hardness of the drugs,<br />

surfactant contents, viscosity, temperature, energy<br />

input <strong>and</strong> size of the milling media. The milling<br />

time can last from 30 minutes to several hours 21 .<br />

Advantages of Pearl milling include low cost,<br />

simple technology <strong>and</strong> ability for large scale<br />

production. The disadvantages associated with this<br />

process are erosion from the milling material<br />

leading to product contamination, adherence of the<br />

product to the inner surface of the mill <strong>and</strong> to the<br />

surface of the milling pearls, long milling times(in<br />

case of hard drugs), potential growth of germs in<br />

the water phase (when milling for a longtime), time<br />

<strong>and</strong> costs associated with the separation procedure<br />

of the milling material from the drug nanoparticle<br />

suspension, especially when producing parenteral<br />

sterile products.<br />

Buchmann et al (1996) 22 reported the formation of<br />

glass micro particles when using glass beads as the<br />

milling media. The erosion from the glass beads<br />

could be reduced when these were coated with<br />

highly cross linked polystyrene resin 23 . The<br />

wastage of the drug due to adherence to milling<br />

surface is of significance in case of very expensive<br />

drugs, particularly when very small quantities are<br />

processed.<br />

The first four marketed products containing<br />

nanocrystals such as Rapamune®, Emend®,<br />

Tricor®, Megace ES® were prepared by Pearl mill<br />

technology by Elan nanosystems.<br />

2.2.2 High Pressure Homogenization Technique<br />

This Technique has been applied for many years<br />

for the production of emulsions <strong>and</strong> suspensions. A<br />

distinct advantage of this technology is its ease for<br />

scale up.<br />

There are three important technologies for<br />

producing nanocrystals using homogenization<br />

methods:-<br />

2.2.2.1. Microfluidizer technology (IDD-P TM<br />

technology)<br />

2.2.2.2. Piston gap homogenization in water<br />

(Dissocubes® technology)<br />

2.2.2.3. Piston gap homogenization in water<br />

mixtures or in non-aqueous medium<br />

(Nanopure® technology)<br />

Vol. 3 (1) Jan – Mar 2012 www.ijrpbsonline.com 408


<strong>International</strong> <strong>Journal</strong> of Research in Pharmaceutical <strong>and</strong> Biomedical Sciences ISSN: 2229-3701<br />

2.2.2.1 Microfluidizer technology:<br />

(Microfluidics TM Inc. USA) This technology is<br />

based on the jet-stream principle. Two streams of<br />

liquid with high velocity (upto 1000 m/sec) collide<br />

frontally under high pressures (upto 1700 bars) 24 .<br />

The particle size is reduced due to high shear force<br />

particle collision <strong>and</strong> cavitation 25 . The same can be<br />

achieved using jet stream homogenizers such as<br />

micro-fluidizer (Microfluidizer® Microfluidics<br />

Inc.). The collision chamber can be either Y-type<br />

or Z-type in shape. Surfactants or phospholipids are<br />

required to stabilize the desired particle size 26, 27 .<br />

Microfluidizer can be used for the production of<br />

drug nanosuspensions for soft drugs. However, this<br />

technique is not very convenient for large scale<br />

production as a large number of cycles (50 to 100<br />

passes) are required for sufficient particle size<br />

reduction 28, 29 . This technique is being utilized by<br />

SkyePharma Canada Inc. for production of<br />

submicron particles of poorly soluble drugs <strong>and</strong><br />

named it IDD-P TM (Insoluble Drug Delivery-<br />

Particle technology).<br />

2.2.2.2 Piston gap homogenization in water<br />

(Dissocubes® technology). Piston gap<br />

homogenization technology was developed by<br />

Müller et al., <strong>and</strong> acquired by SkyePharma in<br />

1999 14,30 . In this technique, powdered drug is<br />

dispersed in an aqueous surfactant solution which<br />

is then forced by a piston through tiny<br />

homogenization gap under high pressure. The gap<br />

width is adjusted according to the viscosity of the<br />

suspension <strong>and</strong> the applied pressure <strong>and</strong> is<br />

generally in the size range of 5 to 20 μm 31 .<br />

According to Bernoulli equation the resulting high<br />

streaming velocity of the suspension causes an<br />

increase in the dynamic pressure which is<br />

compensated by a reduction in the static pressure.<br />

The static pressure in the gap falls below the<br />

vapour pressure of water at room temperature 32 . So<br />

water starts boiling in the gap at room temperature<br />

leading to the formation of gas bubbles. The<br />

formation of gas bubbles leads to pressure waves<br />

disintegrating the crystals. When the liquid leaves<br />

the homogenization gap, the static pressure<br />

increases to normal air pressure <strong>and</strong> gas bubbles<br />

collapse. This process of formation <strong>and</strong> implosion<br />

of gas bubbles is called cavitation. There is particle<br />

size diminution due to high shear forces, turbulent<br />

flow <strong>and</strong> the enormous power of these shock<br />

waves 33 . This technique has been used for<br />

production of nanosuspension of artemisinin <strong>and</strong><br />

quercetin using Tween 80 as a stabilizer (0.5- 2.5<br />

% w/w) 34, 35 .<br />

The use of water as dispersion medium has certain<br />

disadvantages such as hydrolysis of water sensitive<br />

drugs <strong>and</strong> problems during drying step. In case of<br />

thermolabile drugs or drugs having low melting<br />

point, removal of water necessitates the use of<br />

techniques such as lyophilization which are quite<br />

expensive. Dissocubes® technology therefore is<br />

most suitable when aqueous suspensions of<br />

nanocrystals are to be formulated for drugs that are<br />

poorly soluble in both aqueous <strong>and</strong> organic<br />

media 14 . Another advantage of this method is that it<br />

allows aseptic production of nanosuspensions for<br />

parenteral use 36 . Nanocrystal suspensions of<br />

cyclosporine, paclitaxel, amphidicolin,<br />

bupravaquone, azodyecarbonamide <strong>and</strong><br />

prednisolone have been prepared using this<br />

technique.<br />

The two main drawbacks associated with this<br />

method are high installation <strong>and</strong> maintenance cost<br />

of equipments <strong>and</strong> requirement of preprocessing of<br />

the drugs (e.g. micronization).<br />

2.2.2.3 Piston-gap homogenization in water<br />

reduced mixtures or non-aqueous medium<br />

(Nanopure® technology): Another approach using<br />

piston-gap homogenizer is the Nanopure®<br />

technology which is owned <strong>and</strong> developed by<br />

Pharmasol GmbH in Berlin. This technology uses<br />

non-aqueous phase or phases with reduced water<br />

content as dispersion media. Use of non aqueous<br />

media is advantageous for drugs which undergo<br />

hydrolysis in water. The different media used for<br />

homogenization include oils, water-glycerol<br />

mixtures, polyethylene glycols, water- alcohol<br />

mixture etc. These dispersion media have low<br />

vapor pressure. The static pressure in the<br />

homogenization gap does not fall below the vapor<br />

pressure of the liquid, so the liquid does not boil<br />

<strong>and</strong> cavitation does not occur. Even without<br />

cavitation, sufficient size reduction to nano range<br />

takes place 11, 12, 37 . The forces responsible for size<br />

diminution are particle collision <strong>and</strong> shear forces<br />

occurring in highly turbulent fluid in the gap 38 .<br />

Homogenization using Nanopure® technology is<br />

similar or more efficient at lower temperature, i.e.<br />

temperature below the freezing point of water.<br />

Melted non aqueous matrices such as PEG 6000<br />

that are solid at room temperature can also be used<br />

as a medium for homogenization. This leads to<br />

fixation of drug nanocrystals in the solid matrix<br />

<strong>and</strong> minimizes crystal contact <strong>and</strong> subsequent<br />

crystal growth. Drug nanocrystals dispersed in<br />

liquid PEG’s (such as Miglyol 812 or 829) or oils<br />

can be directly filled as drug nanosuspension into<br />

gelatin or HPMC capsules 39 . <strong>Nanocrystals</strong> have<br />

been used as powder for the production of solid<br />

dosage forms such as tablets <strong>and</strong> pellets.<br />

Preparation of solid oral dosage forms from the<br />

nanocrystal suspension requires the removal of<br />

dispersion media from the nanocrystals. Dispersion<br />

medium is removed by either freeze drying or<br />

spray drying. Nanopure Technology offers<br />

advantage in this case since evaporation is faster<br />

<strong>and</strong> takes place at lower temperature due to the use<br />

of non aqueous medium or water reduced mixtures.<br />

This is useful for thermolabile drugs.<br />

Vol. 3 (1) Jan – Mar 2012 www.ijrpbsonline.com 409


<strong>International</strong> <strong>Journal</strong> of Research in Pharmaceutical <strong>and</strong> Biomedical Sciences ISSN: 2229-3701<br />

Isotonic drug nanosuspensions for parenteral<br />

administration can be obtained by homogenization<br />

in water-glycerol mixtures (2.25 % of water free<br />

glycerol). Amphotericin-B powder was dispersed<br />

in liquid PEG-400 <strong>and</strong> in melted PEG 1000<br />

respectively <strong>and</strong> homogenized at 1500 bar up to 25<br />

homogenization cycles 38 .<br />

Thus, High Pressure Homogenization has a number<br />

of advantages to offer (a) it is a continuous process,<br />

(b) there is a limited contamination from the<br />

production equipment (e.g. contamination with iron<br />

was found to be less than 1 ppm ), (c) its ability for<br />

scaling up, even up to production level (d)<br />

possibility of production even without water.<br />

A comparative study on the performance of two<br />

different techniques of nanonization namely, HPH<br />

<strong>and</strong> milling was carried out on Ibuprofen by<br />

Mauludin et al. 19 . Particle size distribution of the<br />

nanocrystals formed was compared. It was found<br />

that the performance of the nanonization technique<br />

depends strongly on the drug properties. In case of<br />

Ibuprofen, which consists of hard crystals, HPH<br />

was superior to the ball milling technology.<br />

Increasing the milling time could not further reduce<br />

the particle size distribution.<br />

Advantages <strong>and</strong> disadvantages of different<br />

nanonization techniques have been listed in Table<br />

1.<br />

2.3. Combination Technologies<br />

The term combination technology has been used<br />

for technologies which combine a pre-treatment<br />

step followed by a high energy homogenization.<br />

2.3.1. NANOEDGE® Technology-<br />

(Microprecipitation <strong>and</strong> Homogenization).<br />

NANOEDGE® Technology was introduced by<br />

Baxter, <strong>and</strong> this involves a combination of<br />

precipitation followed by annealing process.<br />

Annealing process is carried out using high energy<br />

such as high shear forces <strong>and</strong>/or thermal energy 40 .<br />

When drug nanoparticles are produced by<br />

precipitation method alone, the precipitated<br />

nanoparticles have a tendency to grow. Also, the<br />

precipitated particles may be amorphous or<br />

partially amorphous. Upon keeping, the amorphous<br />

particles may re-crystallize <strong>and</strong> this may lead to a<br />

decreased bioavailability of the drug. Combination<br />

technology on the other h<strong>and</strong> has the potential to<br />

overcome these problems, firstly, by prevention of<br />

crystal growth <strong>and</strong> secondly by reducing the<br />

uncertainty of formation of either crystalline or<br />

amorphous state as the annealing process converts<br />

all precipitated particles to crystalline state.<br />

Nanoedge TM technology is particularly suitable for<br />

drugs that are soluble in non-aqueous media<br />

possessing low toxicity, such as N-methyl-2pyrrolidinone.<br />

But the drawback of this method is<br />

its cost especially in case of preparation of sterile<br />

parenteral products.<br />

2.3.2. SmartCrystal® technology This technology<br />

was first developed by PharmaSol GmbH <strong>and</strong> was<br />

later acquired by Abbott. It is a tool-box of<br />

different combination processes in which process<br />

variations can be chosen depending upon the<br />

physical characteristics of the drug (such as<br />

hardness). The process H42 involves a combination<br />

of spray-drying <strong>and</strong> HPH. Drug nanocrystals can<br />

be produced much faster in one to a few<br />

homogenization cycles. Process H69 (Precipitation<br />

<strong>and</strong> HPH) <strong>and</strong> H96 (lyophilization <strong>and</strong> HPH) yield<br />

nanocrystals of amphotericin B within a size range<br />

of about 50 nm 41 .<br />

S. Kobierski et al. (2008) 42 produced nanocrystals<br />

in a two-step process i.e. pre- milling followed by<br />

high pressure homogenization (HPH).<br />

Nanosuspensions of cosmetic active hesperidin<br />

were produced by ball-milling process <strong>and</strong> with<br />

combination process. Both the prepared<br />

nanosuspensions were kept for storage.<br />

Nanosuspension prepared using SmartCrystal®<br />

technology was found to be of a smaller size<br />

indicating better physical stability. Also<br />

combination technique is faster <strong>and</strong> more<br />

economical as compared to HPH alone.<br />

Möschwitzer <strong>and</strong> Müller 43 (2005) prepared spraydried<br />

hydrocortisone acetate powder from<br />

nanosuspension produced by HPH with a micron<br />

LAB 40 <strong>and</strong> planetary monomill “pulverisette 6”.<br />

The number of cycles required could be distinctly<br />

reduced. Additionally, a smaller particle size <strong>and</strong><br />

better particle size distribution could be obtained.<br />

Another finding of the study was that the<br />

application of different homogenization pressures<br />

(e.g. 300 <strong>and</strong> 500 bar) was equally efficient.<br />

Therefore, during large scale production, low<br />

homogenization pressures (300 bars) may be<br />

preferred to reduce wearing of the machine 44 .<br />

3. Processing of nanosuspension to form<br />

nanocrystals<br />

The nanonization of drugs by various techniques<br />

generally results in a liquid product called<br />

nanosuspension. But these nanosuspensions are<br />

directly used as a final product only in some special<br />

cases e.g. as pediatric or geriatric dosage forms. In<br />

most of the cases, a dry dosage form (particularly<br />

for oral administration) is preferred, may be a) for<br />

convenience, b) to achieve a controlled drug<br />

delivery, c) to prevent drug degradation, d) to<br />

enable better drug targeting, e) to increase the<br />

physical stability for long term storage <strong>and</strong> f) to<br />

obtain a fine non-aggregated suspension in the<br />

gastro-intestinal tract after oral administration. In<br />

such cases, the nanosuspension needs to be<br />

transformed into solid forms, which may be<br />

crystalline (<strong>Nanocrystals</strong>) or amorphous<br />

(Nanomorphs). Various techniques are used for this<br />

purpose like spray drying, freeze drying,<br />

pelletization or granulation.<br />

Vol. 3 (1) Jan – Mar 2012 www.ijrpbsonline.com 410


<strong>International</strong> <strong>Journal</strong> of Research in Pharmaceutical <strong>and</strong> Biomedical Sciences ISSN: 2229-3701<br />

3.1 Spray Drying: Spray drying is a simple <strong>and</strong><br />

inexpensive method <strong>and</strong> is therefore suitable for<br />

industrial production. This method is used for drug<br />

nanosuspension produced by high pressure<br />

homogenization <strong>and</strong> is an aqueous solution of<br />

water-soluble matrix materials e.g. polymers (PVP,<br />

long chained PEG’s or polyvinyl alcohol), sugars<br />

(saccharose, lactose) or sugar alcohols like<br />

mannitol <strong>and</strong> sorbitol. In the subsequent step, the<br />

aqueous drug nanosuspension can be spray dried<br />

under adequate conditions. The resulting dry<br />

powder consists of drug nanocrystals embedded in<br />

a water- soluble matrix. The loading capacity of the<br />

solid powder with drug nanocrystals can be<br />

adjusted by varying the concentrations of<br />

surfactants in the original aqueous nanosuspension.<br />

Advantages of this method are that drug<br />

nanocrystals remain fixed within the matrix. Their<br />

physical contact is avoided so the chances of longterm<br />

physical instabilities like aggregation <strong>and</strong><br />

Ostwald ripening are minimized. Exceeding a<br />

certain maximum loading capacity of the matrix<br />

with drug nanocrystals has an increasing negative<br />

effect on crystal growth <strong>and</strong> release as fine<br />

dispersion. The spray dried nano sized powder can<br />

be filled into hard gelatin capsules or sachets or can<br />

used for making tablets. Drug nanoparticles<br />

produced in PEG 600 or Miglyol can directly be<br />

filled into soft gelatin capsules.<br />

3.2. Freeze drying: Another method for removing<br />

water from formulation is freeze drying. This<br />

however, is a complex <strong>and</strong> expensive process <strong>and</strong><br />

the product obtained is highly sensitive to process<br />

parameters. This method is not suitable for<br />

industrial production.<br />

A new technique based upon freeze drying was<br />

developed by de Waard (2010) [7] . In this technique<br />

a mixture of the drug, solvent, <strong>and</strong> mannitol is<br />

cooled rapidly, resulting in separation of drug in a<br />

nanocrystal form encased by a matrix of mannitol.<br />

This matrix increases the stability of the<br />

nanocrstallized drug without which the crystals<br />

may stick together <strong>and</strong> form one large crystal. de<br />

Waard also developed a spray-freeze-drying<br />

method which enables the process to be applied on<br />

an industrial scale. Another method developed by<br />

the same author was a spray-freeze-drying method<br />

which could make industrial application of this<br />

process simpler.<br />

Lyophilization of drug nanoparticles produced in<br />

water-reduced media can be used to produce FDDS<br />

(Fast Dissolving Drug Delivery Systems). For<br />

parenteral application Nanopure can be lyophilized<br />

<strong>and</strong> reconstituted prior to injection with isotonic<br />

media (e.g. water with glycerol).<br />

3.3. Pelletization: A number of pelletization<br />

techniques are known, but the most commonly<br />

used techniques are a) extrusion-spheronization <strong>and</strong><br />

b) drug coating onto sugar spheres. The<br />

pelletization technique is selected on the basis of<br />

the required drug content, properties of the drug<br />

<strong>and</strong> the available equipment. A multi-particulate<br />

dosage form such as coated pellet system is<br />

obtained irrespective of the pelletization technique<br />

applied. These multi-particulate dosage forms show<br />

distinct advantages over single unit dosage forms<br />

such as faster <strong>and</strong> more predictable gastric<br />

emptying <strong>and</strong> more uniform drug distribution in<br />

GIT within different individuals.<br />

3.3.1. Production of pellets containing drug<br />

nanocrystal-loaded matrix cores: The drug<br />

nanosuspension obtained by high-pressure<br />

homogenization is mixed with matrix material<br />

(fillers such as MCC, Lactose or Starch). Pellets<br />

are produced by extrusion-spheronization <strong>and</strong> can<br />

be subsequently coated with polymers to modify<br />

the drug release properties.<br />

Mucoadhesive budenoside nanocrystals were<br />

prepared using extrusion-spheronization. The<br />

obtained pellets were coated with Eudragit L 30 D-<br />

55 to obtain enteric coating <strong>and</strong> delayed drug<br />

release [44] . Another type of modified release pellets<br />

were prepared by Mauludin et al. [45] (2005). These<br />

effervescent pellet formulations containing<br />

ibuprofen drug nanocrystals were produced by<br />

HPH. These ibuprofen containing pellets loaded<br />

with nanocrystals of the drug dissolved completely<br />

within 30 minutes from both formulations.<br />

Spray coated pellets of hydrocortisone acetate were<br />

prepared (enteric coated) from mucoadhesive<br />

nanosuspension of this poorly soluble drug. The invitro<br />

dissolution tests showed an accelerated<br />

dissolution rate <strong>and</strong> an increased drug release for<br />

the pellets containing drug nanocrystals [46] .<br />

3.3.2. Production of pellets by Nanosuspension<br />

layering onto sugar cores: The drug<br />

nanosuspension obtained by HPH is directly<br />

layered onto sugar beads <strong>and</strong> subsequently coated<br />

with polymers using the same equipment to modify<br />

the drug release properties.<br />

4. Advanced Techniques for Production of Solid<br />

<strong>Nanocrystals</strong><br />

There are many alternative technologies that are<br />

industrially less relevant. These are discussed<br />

below.<br />

4.1. Solution Enhanced Dispersion by the<br />

Supercritical fluids (SEDS): The SEDS method<br />

was developed <strong>and</strong> patented by the University of<br />

Bradford [47] . This technique uses an antisolventbased<br />

recrystallization process. CO2 is used as<br />

antisolvent. The substances are atomized into a<br />

chamber containing compressed CO2. As two<br />

liquids collide, intense atomization into micronized<br />

droplets occurs, subsequently drying of micro<br />

Vol. 3 (1) Jan – Mar 2012 www.ijrpbsonline.com 411


<strong>International</strong> <strong>Journal</strong> of Research in Pharmaceutical <strong>and</strong> Biomedical Sciences ISSN: 2229-3701<br />

droplets occur as the solvent(s) <strong>and</strong> CO2 mix.<br />

Nanoparticles are formed because the two way<br />

mass transfer ie extraction of organic solvent <strong>and</strong><br />

CO2 diffusion into the droplets occurs [48] .<br />

It is important to enhance the mass transfer rate<br />

between the droplets <strong>and</strong> the anti solvent before the<br />

droplets coalesce to form large size drops.<br />

This technique has produced a new polymorph of<br />

flucticasone propionate. It also enabled control<br />

over the particle size <strong>and</strong> shape of formed particles.<br />

This polymorph exhibited improved drug delivery<br />

characteristics in a metered dose inhaler (MDI)<br />

formulation compared to conventional <strong>and</strong><br />

micronized drugs [49] .<br />

4.2. Spray Freezing into Liquid (SFL): The<br />

University of Texas (Austin) was the first to<br />

develop <strong>and</strong> patent the SFL method in 2003.This<br />

technique was first commercialized by Dow<br />

Chemical Company (Midl<strong>and</strong>, MI).The drug<br />

present in an aqueous /organic /aqueous organic cosolvent<br />

solution, aqueous organic emulsion or<br />

suspension is atomized directly into either a<br />

cryogenic liquid (e.g. argon, nitrogen or halocarbon<br />

refrigerants) or a compressed gas (e.g. CO2, ethane,<br />

propane or helium).<br />

The fed solution is atomized through a nozzle<br />

positioned at a distance above the boiling<br />

refrigerant. The droplets gradually solidify while<br />

passing through the cold halocarbon vapor, <strong>and</strong><br />

freeze completely as contact is made with the<br />

boiling refrigerant liquid. These frozen particles are<br />

then lyophilized to obtain free-flowing <strong>and</strong> dry<br />

micronized powder [50] . The SFL powder showed<br />

better results such as highly effective wettability,<br />

high surface area <strong>and</strong> enhanced dissolution rates.<br />

Unfortunately, this process may result in broad<br />

particle size distributions <strong>and</strong> non-micronized<br />

particles because agglomerates of the solution<br />

droplets are solidified while passing through the<br />

vapor phase <strong>and</strong> settle onto the surface of the<br />

cryogenic liquid [49] .<br />

4.3. Rapid Expansion of Supercritical Solution<br />

(RESS): The principle of the RESS process is to<br />

induce a fast nucleation of the supercritical fluid of<br />

dissolved drugs in the presence of surfactants. This<br />

results in particle formation with a desirable size<br />

distribution in a very short time. The surfactants<br />

serve to stabilize the formed small particles <strong>and</strong><br />

suppress any tendency towards particle<br />

agglomeration or particle growth while they are<br />

being formed. The rapid intimate contact with the<br />

surface modifier is achieved by having the surface<br />

modifiers dissolved in the supercritical fluid<br />

containing the dissolved drugs. A rapid intimate<br />

contact between the surfactant <strong>and</strong> the newly<br />

formed particles inhibits the crystal growth of the<br />

newly formed particle [49,51] .<br />

This technique successfully produced cyclosporine<br />

nanocrystals with a size of 500–700 nm.<br />

Cyclosporine could be stabilized at drug<br />

concentrations as high as 6.2 <strong>and</strong> 37.5 mg/mL in<br />

1.0% <strong>and</strong> 5.0% (w/w) Tween 80 solution,<br />

respectively [49,52] . The dissolution of griseofulvin<br />

was two times higher than conventional micronized<br />

preparation [53] .<br />

Combination of RESS technique followed by highpressure<br />

homogenization was used by Pace (2001)<br />

[51]<br />

to prepare stable nanosuspensions of poorly<br />

soluble drugs.<br />

Young (1999) [54] developed a process based on<br />

supercritical fluids, rapid expansion from the<br />

supercritical to the aqueous solution (RESAS).<br />

4.4. Rapid Expansion of Supercritical Solution<br />

into Aqueous Solution (RESSAS): In contrast to<br />

RESAS, the RESSAS process utilizes a<br />

supercritical fluid which is exp<strong>and</strong>ed into an<br />

aqueous solution containing a stabilizer. This<br />

technique was used by Turk et al. to produce<br />

phytosterol particles with a diameter less than 500<br />

nm. The surfactants or stabilizers are dissolved in<br />

the aqueous phase, not in the supercritical fluid [55] .<br />

4.5. Evaporative Precipitation into Aqueous<br />

Solution Process (EPAS): The evaporative<br />

precipitation into aqueous solution (EPAS) applies<br />

rapid phase separation to nucleate <strong>and</strong> grow<br />

nanoparticles <strong>and</strong> micro particles of poorly watersoluble<br />

drugs. The EPAS was developed <strong>and</strong><br />

patented by the University of Texas at Austin in<br />

2001 <strong>and</strong> commercialized by the Dow Chemical<br />

Company [49] . The drug is dissolved in a low<br />

boiling point organic solvent.<br />

The drug solution is pumped through a tube where<br />

it is heated under pressure at a temperature above<br />

the solvent’s boiling point. It is then sprayed<br />

through a fine atomizing nozzle into heated<br />

aqueous solution. This process results in an<br />

amorphous suspension. Surfactants are added for<br />

efficient particle formation <strong>and</strong> stabilization. The<br />

stable aqueous drug suspension is dried by<br />

lyophilization or spray drying. A variety of<br />

hydrophilic surfactants are added to the solution to<br />

diffuse to surface of the growing particles rapidly<br />

enough to prevent growth of particles. The EPAS<br />

technique has produced cyclosporine-A<br />

nanosuspension with particle size ranging from 130<br />

to 460 nm [56] .<br />

5. Applications of <strong>Nanocrystals</strong> by various<br />

routes of administration:-<br />

5.1 Oral administration: - Enhancement in<br />

bioavailability of poorly soluble drugs after oral<br />

administration is well documented in the literature<br />

[3, 10, 21 ] . Besides it has also been proved by various<br />

drug nanocrystal products placed in the market. A<br />

faster onset of action <strong>and</strong> decreased gastric<br />

Vol. 3 (1) Jan – Mar 2012 www.ijrpbsonline.com 412


<strong>International</strong> <strong>Journal</strong> of Research in Pharmaceutical <strong>and</strong> Biomedical Sciences ISSN: 2229-3701<br />

irritancy has been reported when naproxen was<br />

formulated as nanosuspension [3, 57] . Due to fast<br />

dissolution of nanocrystals, the drug solubility is<br />

enhanced, making it bioequivalent in fed <strong>and</strong><br />

fasting conditions. The bioadhesive nature of<br />

nanocrystals offers additional advantage of<br />

increased stay in the gastro-intestinal tract which<br />

enhances bioavailability [58, 59] . The nano size can<br />

be exploited for better drug targeting as reported<br />

for lymphatic drug uptake [60] or for inflammatory<br />

tissues [5] . Nanosuspensions can be formulated as<br />

more concentrated <strong>and</strong> less viscous. Patient has to<br />

take lesser dose of easily swallowed formulation<br />

(e.g. Megace ES). Nanoparticles provided<br />

sustained release of anti-tubercular drugs –<br />

rifampin, isoniazid <strong>and</strong> pyrazinamide <strong>and</strong><br />

considerably improved their efficacy after oral<br />

administration [8] . Amphotericin B administered<br />

orally as a nanosuspension showed dramatically<br />

improved bioavailability as compared to its<br />

conventional oral commercial products such as<br />

Fungizone, AmBisome <strong>and</strong> micronized<br />

Amphotericin B [33, 62] . Aqueous nanosuspensions<br />

can be used directly in a liquid dosage form or<br />

incorporated in a dry dosage form such as tablets,<br />

capsules <strong>and</strong> fast melts by means of st<strong>and</strong>ard<br />

manufacturing technologies. Ketoprofen<br />

nanosuspensions have been successfully<br />

incorporated into pellets to release the drug over a<br />

period of 24 hrs.<br />

[63] . Oral Fenofibrate<br />

nanosuspensions showed better bioavailability as<br />

compared to conventional micronized drug<br />

suspensions [64] .<br />

5.2 Parenteral administration:- The conventional<br />

parenteral preparations (particularly for intravenous<br />

administration) of poorly soluble drugs are<br />

formulated by dissolving the drug with the help of<br />

cosolvents, surfactants, liposomes or cyclodextrins<br />

which is often associated with toxic effects <strong>and</strong><br />

large injection volumes. Aqueous nanosuspensions<br />

are an ideal formulation to overcome these<br />

problems e.g. Paclitaxel nanosuspensions cause<br />

less toxicity as compared to Taxol with<br />

Chremophor EL [65] . The parenteral administration<br />

of Clofazimine, an anti-tubercular drug was<br />

restricted due to its poor solubility. Clofazimine<br />

nanosuspension consisting only of the drug <strong>and</strong> a<br />

minimum amount of surfactant was injected<br />

intravenously, which showed better stability <strong>and</strong><br />

efficacy over liposomal Clofazimine in M. avium<br />

infected mice [66] . An intravenous nanosuspension<br />

of Itraconazole was reported to enhance the anti<br />

fungal effect in comparison to its solution dosage<br />

form in rats [67] . Due to higher loading capacity<br />

with nanosuspensions, the injectable dose can be<br />

distinctly reduced, compared to solutions [36] . Drug<br />

nanosuspensions can be sterilized by autoclaving,<br />

using gamma radiations or by sterile filtration using<br />

0.22 mm filter [68, 69] . <strong>Nanocrystals</strong> above 200nm<br />

dissolve slowly <strong>and</strong> are taken up by macrophages<br />

of the liver. This causes potentially targeted<br />

toxicity to the liver. <strong>Nanocrystals</strong> with size well<br />

below 100 nm dissolve much faster. The injection<br />

of nanosuspensions containing smaller particles<br />

resembles the intravenous injection of solutions<br />

<strong>and</strong> reduces the uptake by the liver. The targeting<br />

of clofazimine nanosuspension to the lung, liver,<br />

spleen <strong>and</strong> reticulo-endothelial system was<br />

comparable to the liposomal formulation [66] .<br />

Intravenous administration of nanosuspensions has<br />

further advantage of passive drug delivery to<br />

inflammatory sites where endothelium becomes<br />

permeable due to pathological processes. The<br />

passive accumulation in such sites with leaky<br />

vasculature was found to be more effective with<br />

long circulating nanoparticles [70] . Flexibility of<br />

nanosuspensions was further demonstrated by<br />

effective subcutaneous treatment of mice infected<br />

with M. tuberculosis [71] .<br />

5.3 Pulmonary drug delivery: - Poorly soluble<br />

drugs can be delivered directly to the lungs by<br />

nebulizing the aqueous nanosuspensions using<br />

mechanical or ultrasonic nebulizers. Using<br />

nanoparticles, drug is more evenly distributed in<br />

droplets. All aerosol droplets are likely to contain<br />

drug nanocrystals. Budenoside, poorly water<br />

soluble corticosteroid, has been successfully<br />

prepared as a nanosuspension for pulmonary<br />

delivery. It showed long term stability. No particle<br />

growth <strong>and</strong> aggregates formed over a period of one<br />

year [72] . In addition, Buparvaquone<br />

nanosuspension was formulated for an alternative<br />

treatment of lung infection (pneumonia) to deliver<br />

the drug at the site of lung infection using<br />

nebulization [73] . Administration to infected guinea<br />

pigs of nebulized rifampin, isoniazid <strong>and</strong><br />

pyrazinamide encapsulated in wheat germ<br />

agglutinin-functionalized PLG nanoparticles was<br />

much more effective. Three doses administered<br />

fortnightly for 45 days were sufficient to produce a<br />

sterilizing effect in lungs <strong>and</strong> spleen [74] . Drug<br />

nanocrystals showed an increased mucoadhesiveness<br />

leading to a prolonged residence time<br />

at the lung mucosa [73] .<br />

5.4 Dermal application: - Dermal<br />

nanosuspensions are mainly of interest if<br />

conventional approaches fail. <strong>Nanocrystals</strong> can<br />

increase the penetration of poorly soluble cosmetic<br />

<strong>and</strong> pharmaceutical substances into skin. This<br />

happens because increased saturation solubility<br />

increases the concentration gradient. Juvena<br />

launched first four Nanocrystal cosmetic products<br />

with rutin. Petersen (2006) [75] reported that rutin<br />

Nanocrystal formulation possesses 500 times<br />

higher bioactivity (measured as Sun Protection<br />

Factor,SPF) compared to water-soluble rutin-<br />

Vol. 3 (1) Jan – Mar 2012 www.ijrpbsonline.com 413


<strong>International</strong> <strong>Journal</strong> of Research in Pharmaceutical <strong>and</strong> Biomedical Sciences ISSN: 2229-3701<br />

glycoside . Dermal application of nanocrystals is<br />

protected by a US <strong>and</strong> PCT patent application.<br />

Shegokar et al. (2011) [76] reported that both rutin<br />

<strong>and</strong> hesperidin nanocrystals increased the SPF by<br />

36% <strong>and</strong> 59% respectively. This proves that<br />

nanocrystals increase the penetration into the skin.<br />

Shaal et al. (2010) [77] prepared apigenin<br />

nanocrystals <strong>and</strong> reported that UV skin protective<br />

potential can be significantly increased by<br />

decreasing the particle size from micrometer to the<br />

nanometer range.<br />

5.5. Ophthalmic drug delivery: -<br />

Nanosuspensions can prove beneficial for drugs<br />

that have poor solubility in lachrymal fluids.<br />

Nanosuspensions offer advantage of prolonged<br />

retention time in the eye, most likely due to their<br />

adhesive properties. Another advantage of<br />

nanosuspensions is high drug loading which avoids<br />

high tonicity created by water soluble drugs.<br />

Pignatello (2002) [78] developed a polymeric<br />

nanosuspension of ibuprofen for ophthalmic drug<br />

delivery. Ophthalmic nanosuspensions of glucocorticoid<br />

drugs; hydrocortisone, prednisolone <strong>and</strong><br />

dexamethasone show increased rate <strong>and</strong> extent of<br />

drug absorption <strong>and</strong> also prolonged duration [79] .<br />

5.6 Targeted drug delivery: - <strong>Nanocrystals</strong> can<br />

have deep excess to the human body because of<br />

particle size <strong>and</strong> control of surface properties. So<br />

they can also be used for targeted drug delivery.<br />

Kayser (2000) [80] developed a nanosuspention of<br />

aphidicolin to improve drug targeting against<br />

Leishmania-infected macrophages. He<br />

demonstrated that aphidicolin was highly active at<br />

a concentration in the microgram range. Similarly<br />

peptide dalargin was successfully targeted to the<br />

brain by employing surface modified poly(butyl )<br />

cyanoacrylate nanoparticles [81] .<br />

Nanoparticles offer a promising new cancer<br />

treatment that may one day replace radiation <strong>and</strong><br />

chemotherapy. Kangius RF therapy attaches<br />

microscopic nanoparticles to cancer cells <strong>and</strong> then<br />

cooks tumors inside the body with radio waves that<br />

heat only the nanoparticles <strong>and</strong> the adjacent<br />

cancerous cells.<br />

Muco-adhesive pellets or nanoparticles have been<br />

used as specific carrier systems for oral<br />

administration [82-84] . Cevc et al. (1998) [85] used<br />

transferosomes for targeted topical delivery.<br />

Bupravaquone nanosuspension was successfully<br />

used for targeting of Cryptosporidium parvum, (the<br />

organism responsible for cryptosporidium) by<br />

altering the mucoadhesive properties [86] .<br />

Amphoterecin B as pulmonary nanosuspension was<br />

used to target conditions such as pulmonary<br />

asperigillosis [87] .<br />

6. Marketed products <strong>and</strong> products in the<br />

pipeline<br />

The first four marketed products containing<br />

nanocrystals were prepared by Pearl mill<br />

technology by Elan nanosystems. Rapamune® was<br />

the first marketed product containing sirolimus<br />

nanocrystals introduced in 2000 by Wyeth. The<br />

main advantages of nanocrystal technology in the<br />

coated tablets of Rapamune® nanocrystals are<br />

more convenient dosage form <strong>and</strong> a 21% higher<br />

bioavailability as compared to the Rapamune<br />

solution. A smaller particle size leads to greater<br />

solubility <strong>and</strong> larger surface area consequently<br />

increased dissolution velocity <strong>and</strong> thus greater<br />

bioavailability<br />

Emend®, the second product incorporating<br />

nanocrystal technology was introduced in 2003 by<br />

Merck. Emend® is a capsule containing pellets of<br />

Aprepitant, drug used for the treatment of emesis.<br />

Aprepitant shows a narrow absorption window i.e.<br />

absorbed in the upper gastrointestinal tract only. In<br />

this case, large increase in surface area due to<br />

nanonisation leads to rapid in-vivo dissolution, fast<br />

absorption <strong>and</strong> increased bioavailability.<br />

Tricor® was launched in December 2004 by<br />

Abbott Laboratory. The active ingredient is<br />

fenofibrate. The normal size fenofibrate is absorbed<br />

when taken with a meal. The nanocrystal<br />

technology makes the bioavailability of fenofibrate<br />

independent of meals.<br />

Megace ES® (Megestrol acetate) (ES st<strong>and</strong>s for<br />

Enhanced Stability).<br />

Megace ES® was introduced by Par<br />

Pharmaceutical companies, Inc. The nanocrystal<br />

technology leads to several advantages. The<br />

<strong>Nanocrystals</strong> offer enhanced bioavailability which<br />

reduces the amount of single dose. The patient has<br />

to take less volume. Another advantage is that there<br />

is no need of increasing viscosity to prevent<br />

sedimentation. Decreased dose <strong>and</strong> reduced<br />

viscosity lead to better patient compliance.<br />

Products already in the market or in the pipeline are<br />

listed in table 2 <strong>and</strong> table 3 respectively.<br />

7. Limitations of drug nanocrystal technology:-<br />

Many nanoparticulate delivery systems are under<br />

academic investigation. But only few made it to the<br />

market. This may be due to missing nanotoxicity<br />

<strong>and</strong> cytotoxicity data, lack of regulatory accepted<br />

status of the excipients, lack of large scale<br />

production lines which can be validated <strong>and</strong><br />

acceptable by the regulatory authorities.<br />

Nanotoxicity may be attributed to the small size<br />

(below about 150 nm) of nanocrystals, due to<br />

which they can have access to any cell of the body<br />

via pinocytosis. This increases the risk of<br />

cytotoxicity.<br />

Moreover, this technology requires expensive<br />

equipments which increase the cost of the final<br />

product. The use of this technique is restricted to<br />

Vol. 3 (1) Jan – Mar 2012 www.ijrpbsonline.com 414


<strong>International</strong> <strong>Journal</strong> of Research in Pharmaceutical <strong>and</strong> Biomedical Sciences ISSN: 2229-3701<br />

BCS class II drugs only. Furthermore, the<br />

production of nanocrystals <strong>and</strong> their stability is<br />

dependent on the molecular structure of the drug.<br />

Due to this, only certain categories of drugs will be<br />

suitable c<strong>and</strong>idates for this technique.<br />

8. Conclusion<br />

Nanocrystal technology seems to be a promising<br />

tool for the formulation of poorly soluble drugs.<br />

The nanocrystals despite being poorly soluble just<br />

dissolve <strong>and</strong> disappear in the presence of large<br />

amount of water. They have been successfully<br />

employed to improve the bioavailability, better<br />

drug targeting with minimum side effects, reduced<br />

drug dosage <strong>and</strong> hence better patient compliance.<br />

They can be incorporated in solid dosage forms<br />

like tablets <strong>and</strong> capsules which are more patient<br />

friendly. Nevertheless their nanotoxicity needs to<br />

be assessed <strong>and</strong> more authenticated therapeutic<br />

data is awaited.<br />

However, nanocrystals hold promise to appear in<br />

many future products, not only pharmaceutical but<br />

also cosmetic. They can be seen as a ray of hope in<br />

the targeted treatment of cancer.<br />

Declaration of interest<br />

The authors state no conflict of interests <strong>and</strong> have<br />

received no payment in the preparation of this<br />

manuscript.<br />

Acknowledgement<br />

The authors are thankful to Dr. R.H. Müller for<br />

providing the requisite reference material.<br />

Table 1: Advantages <strong>and</strong> disadvantages of different methods for the production of nanocrystals<br />

Technology Advantages Disadvantages<br />

Precipitation a. Finely dispersed drug<br />

b. Better control of desired size<br />

c. Low-energy technique<br />

Milling a. Low-energy technique<br />

b. Proven by first four marketed<br />

products<br />

High Pressure<br />

Homogenization<br />

a. Universally applicable<br />

b. Large scale production possible<br />

c. Fast method (several minutes<br />

possibly<br />

d. Continuous process<br />

a. Particle growth on keeping<br />

b. Organic solvent residue<br />

c. Not universally applicable, only drugs with<br />

certain properties are possible (e.g., soluble in at least one<br />

solvent)<br />

d. Needs to be stabilized<br />

a. Impurity due to erosion from milling material<br />

b. Can be a slow process (several days)<br />

c. Wastage of the drug due to adherence to the<br />

pearls<br />

d. Large scale production difficult due to size of<br />

milling chamber<br />

a. High-energy technique<br />

b. Great experience required<br />

Table 2: List of marketed products containing nanocrystals<br />

Trade name Drug Company Applied technology<br />

Rapamune® Rapamycin Wyeth Ball milling<br />

Emend® Aprepitant Merck Ball milling<br />

Tricor® Fenofibrate Abbott Ball milling<br />

Megace ES® Megestrol Par Pharmaceutical Companies Ball milling<br />

Avinza® Morphine sulfate King Pharmaceutical Ball milling<br />

Focalin® XR Dexmethylphenidate hydrochloride Novartis Ball milling<br />

Zanaflex CapsulesTM Tizanidine hydrochloride Acorda Ball milling<br />

Triglide® Fenofibrate Sciele Pharma Inc. HPH (Microfluidizer)<br />

Ritalin® LA Methylphenidate hydrochloride Accorda Ball milling<br />

Table 3: Products under clinical trial<br />

Trade name Drug Company<br />

Semapimod® Guanylhydrazone<br />

Cytokine Pharma<br />

sciences<br />

Paxceed® Paclitaxel<br />

Angiotech<br />

Pharmaceuticals Inc<br />

Theralux® Thymectacin Celmed BioSciences Inc<br />

Nucryst® Silver Nucryst Pharmaceuticals<br />

Vol. 3 (1) Jan – Mar 2012 www.ijrpbsonline.com 415


<strong>International</strong> <strong>Journal</strong> of Research in Pharmaceutical <strong>and</strong> Biomedical Sciences ISSN: 2229-3701<br />

REFERENCES<br />

1. Merisko-Liversidge E. 2002. <strong>Nanocrystals</strong>:<br />

resolving pharmaceutical formulation issues<br />

associated with poorly water-soluble<br />

compounds. Particles; April 20–23; Orl<strong>and</strong>o,<br />

Florida, USA.<br />

2. Grant, D.J.W., Brittian, H.G., 1995. In:<br />

Brittian, H.G. (Ed.), Physical Characterisation<br />

of Pharmaceutical Solids. Marcel Dekker, New<br />

York.<br />

3. Liversidge, G.G., Conzentino, P. 1995. Drug<br />

particle size reduction for decreasing gastric<br />

irritancy <strong>and</strong> enhancing absorption of<br />

naproxen in rats. Int. J. Pharm. 125,309–313.<br />

4. Liversidge G.G., Cundy K.C.1995. Particle<br />

size reduction for improvement of oral<br />

bioavailability of hydrophobic drugs: Absolute<br />

oral bioavailabilty of nanocrystalline danazol<br />

in beagle dogs. Int. J. Pharm. 125, 91-97.<br />

5. Lamprecht, A., Ubrich, N., Yamamoto, H., et<br />

al. 2001. Biodegradable nanoparticles for<br />

targeted drug delivery in treatment of<br />

inflammatory bowel disease. J. Pharmacol.<br />

Exp. Ther. 299 (2), 775–781.<br />

6. Gabor, F., Bogner, E., Weissenboeck, A.,<br />

Wirth, M. 2004. The lectin-cell interaction <strong>and</strong><br />

its implications to intestinal lectin- mediated<br />

drug delivery. Adv. Drug. Deliv. Rev. 56, 459–<br />

480.<br />

7. de Waard, H., De Beer, T., Hinrichs, W.L.J.,<br />

Vervaet, C., Remon, J.P.,Frijlink, H.W., 2010.<br />

Controlled crystallization of the lipophilic drug<br />

Fenofibrate during freeze-drying: Elucidation<br />

of the mechanism by in- line Raman<br />

spectroscopy. The AAPS <strong>Journal</strong> 12(4), 569-<br />

575.<br />

8. P<strong>and</strong>ey R., Zahoor A., Sharma S., Khuller<br />

G.K. 2003. Nanoparticle encapsulated<br />

antitubercular drugs as a potential oral drug<br />

delivery system against murine tuberculosis.<br />

Tuberculosis (Edinb); 83, 373–378.<br />

9. Shegokar,R., Al Shaal,L., Müller, R.H., 2010.<br />

Local Anaesthetic nanocrystals as prolonged<br />

release formulation. First Combined Annual<br />

Meeting of <strong>International</strong> Pharmaceutical<br />

Federation, PSWC <strong>and</strong> AAPS New Orleans<br />

Louisiana. USA. Abstract no. 2203.<br />

10. Liversidge G.G., Cundy K.C., Bishop J.F.,<br />

Czekai D.A. 1992. NANOSYSTEMS LLC.<br />

Surface modified drug nanoparticles. US Pat.:<br />

5,145,684.<br />

11. Müller, R.H., Mäder K., Krause K., 2002.<br />

PharmaSol GmbH, Dispersions for<br />

formulation slightly or poorly soluble active<br />

ingredients. Pat.: CA0002388550A1, Feb. 7.<br />

12. Radtke, M., 2001. Nanopure TM: pure drug<br />

nanoparticles for the formulation of poorly<br />

soluble drugs. New Drugs 3,62-68<br />

13. Müller, R.H. 2002. Nanopure technology for<br />

the production of drug nanocrystals <strong>and</strong><br />

polymeric particles. 4 th World Meeting<br />

ADRITELF/APV/APGI. Florence<br />

14. Müller, R.H., Jacobs, C., Kayser, O. 2003.<br />

DissoCubes - a novel formulation for poorly<br />

soluble <strong>and</strong> poorly bioavailable drugs, in<br />

Modified-Release Drug Delivery Systems,<br />

M.J. Rathbone, Hadgraft, J., Roberts, M. S.,<br />

Editor. Marcel Dekker. 135-149<br />

15. Akkar, A., Müller, R. H. 2003. <strong>Nanocrystals</strong><br />

of Itraconazole <strong>and</strong> amphotericin B produced<br />

by high pressure homogenisation. Annual<br />

Meeting of the American Association of<br />

Pharmaceutical Scientists, Salt Lake City.<br />

16. Gassmann, P., List, M., Schweitzer, A. et al.<br />

1994. Hydrosols – alternatives for the<br />

parenteral application of poorly water soluble<br />

drugs. Eur. J. Pharm. Biopharm. 40, 64–72.<br />

17. List, M.A., Sucker, H. 1988. Pat No. GB<br />

2200048. Great Britian.<br />

18. Speiser, P.P. 1998. Poorly soluble drugs, a<br />

challenge in drug delivery, In Müller, R.H.,<br />

Benita, S., Bohm, B. (eds.), Emulsions <strong>and</strong><br />

Nanosuspensions for the Formulation of<br />

Poorly Soluble Drugs, Medpharm Stuttgart.<br />

Scientific Publishers: 15-28<br />

19. Mauludin, R., Möschwitzer, J., Müller, R. H.<br />

2005. Comparison of Ibuprofen Drug<br />

<strong>Nanocrystals</strong> Produced by High Pressure<br />

Homogenization (HPH) versus Ball Milling,<br />

AAPS Annual Meeting, Nashville, T2217.<br />

20. Shackleford, D.M., Faassen, W.A., Houwing,<br />

N., et al.2003. Contribution of lymphatically<br />

transported testosterone undecanoate to the<br />

systemic exposure of testosterone after oral<br />

administration of two <strong>and</strong>riol formulations in<br />

conscious lymph duct-cannulated dogs. J.<br />

Pharmacol. Exp. Ther. 3063, 925–33.<br />

21. Merisko-Liversidge, E., Liversidge, G.G.,<br />

Copper, E.R. 2003. Nanosizing: a formulation<br />

approach for poorly-water-soluble compounds.<br />

Eur. J. Pharma. Sci. 18, 113–20.<br />

22. Buchmann, S., Fischli, W., Thiel, F.P., Alex,<br />

R. 1996. Aqueous microsuspension, an<br />

alternative intravenous formulation for animal<br />

studies. In: 42nd annual congress of the<br />

<strong>International</strong> Association for Pharmaceutical<br />

Technology (APV), Mainz.<br />

23. Bruno, J.A.D., Brian, D., Gustow, Evan, Illig,<br />

Kathleen J, Rajagopalan, Nats, Sarpotdar.<br />

1992. Method of grinding pharmaceutical<br />

substances. US 5,518,187.<br />

24. Bruno, R.P., McIlwrick, R.1999.<br />

Microfluidizer processor technology for high<br />

performance particle size reduction, mixing<br />

<strong>and</strong> dispersion. Eur. J. Pharm. Biopharm. 56,<br />

29–36.<br />

25. Tunick, M.H., Van Hecken, D.L., Cooke, P.H.,<br />

et al. 2002 Transmission electron microscopy<br />

of mozzarella cheeses made from<br />

Vol. 3 (1) Jan – Mar 2012 www.ijrpbsonline.com 416


<strong>International</strong> <strong>Journal</strong> of Research in Pharmaceutical <strong>and</strong> Biomedical Sciences ISSN: 2229-3701<br />

microfluidized milk. J. Agric. Food Chem. 50,<br />

99–103.<br />

26. Sunstrom, J.E. IV.,William, R., Marshik-<br />

Guerts, B. 1996. General Route to<br />

Nanocrystalline Oxides by Hydrodynamic<br />

Cavitation. Chem. Mater. 8(8), 2061-2067.<br />

27. Gruverman, I.J., Thum, J.R., Production of<br />

Nanostructures Under Turbulent Collision<br />

Reaction Conditions - Application to Catalysts,<br />

Superconductors, CMP Abrasives, Ceramics<br />

<strong>and</strong> other Nanoparticles. Microfluidics<br />

Research.<br />

28. Parikh, I.S., Ulagaraj.1997. Composition <strong>and</strong><br />

method of preparing microparticles of water<br />

insoluble substances 93969997.<br />

29. Dearn, A.R. 2000. Atovaquone<br />

pharmaceutical compositions. US, 08/974248,<br />

6018080.<br />

30. Rainbow, B.E. 2004. Nanosuspensions in<br />

Drug Delivery. Nat. Rev. 3, 785-796.<br />

31. Möschwitzer, J., Müller, R.H. 2005.<br />

Application. Germany: 2005. Method for the<br />

production of ultrafine submicron<br />

nanosuspensions. DE 10 2005 011 786.4.<br />

32. Müller, R.H., Peters, K., Becker, R., et al.<br />

1995. Nanosuspensions – a novel formulation<br />

for the IV administration of poorly soluble<br />

drugs. 1st World Meeting of the <strong>International</strong><br />

Meeting on Pharmaceutics, Biopharmaceutics<br />

<strong>and</strong> Pharmaceutical Technology; Budapest.<br />

33. Müller, R.H., Jacobs, C., Kayser, O. 2001.<br />

Nanosuspensions as particulate drug<br />

formulations in therapy: Rationale for<br />

development <strong>and</strong> what we can expect for the<br />

future. Adv. Drug Deliv. Rev. 471, 3–19.<br />

34. Sahoo, N. G., Al Shaal, L., Kakran, M., Li, L.,<br />

Müller, R. H. 2010a. Artimisinin nanocrystals<br />

for improved oral bioavailability in malaria<br />

treatment, AAPS Annual Meeting, T2154,<br />

New Orleans.<br />

35. Sahoo, N. G., Al Shaal, L., Kakran, M., Li, L.,<br />

Müller, R. H. 2010. Antioxident quercetin:<br />

preparation <strong>and</strong> characterization of<br />

nanocrystals, AAPS Annual Meeting, W4134,<br />

New Orleans.<br />

36. Möschwitzer, J., Achleitner, G., Pomper, H.,<br />

Müller, R.H. 2004. Development of an<br />

intravenously injectable chemically stable<br />

aqueous omeprazole formulation using<br />

nanosuspension technology. Eur. J. Pharm.<br />

Biopharm. 58 (3), 615–619.<br />

37. Fichera, M.A., Keck, C.M. Müller, R.H. 2004.<br />

Nanopure Technology - Drug <strong>Nanocrystals</strong> for<br />

the Delivery of Poorly Soluble Drugs, in<br />

Particles. Orl<strong>and</strong>o.<br />

38. Bushrab, N.F., Müller, R.H. 2003.<br />

<strong>Nanocrystals</strong> of Poorly Soluble Drags for Oral<br />

Administration. New Drugs 5, 20-22.<br />

39. Keck, C.M., Bushrab, N.F. Müller, R.H. 2004.<br />

Nanopure® <strong>Nanocrystals</strong> for Oral Delivery of<br />

Poorly Soluble Drugs, in Particles. Orl<strong>and</strong>o.<br />

40. Kipp, J.E., Wong, J.C.T., Doty, M.J., et al.<br />

2003. Microprecipitation method for<br />

preparing submicron suspensions. US Patent<br />

6607784; USA.<br />

41. Fichera, M.A., Wissing, S.A., Müller, R.H.<br />

2004. Effect of 4000 Bar Homogenisation<br />

Pressure on Particle Diminution in Drug<br />

Suspensions, in APV. Niirnberg.<br />

42. Kobierski, S., Hanisch, J., Mauludin, R.,<br />

Müller, R. H., Keck, C. 2008. Nanocrystal<br />

production by smartCrystal combination<br />

technology, Int. Symp. Control. Rel. Bioact.<br />

Mater. 35, #3239, New York City.<br />

43. Möschwitzer, J., Müller, R. H. 2005.<br />

Development of a New Two-Step Process for<br />

the Effective Production Drug <strong>Nanocrystals</strong><br />

By, AAPS Annual Meeting, Nashville,<br />

W5124.<br />

44. Möschwitzer, J., Müller, R. H. 2004. Final<br />

Formulations for Drug <strong>Nanocrystals</strong>: Pellets,<br />

AAPS Pharmaceutics <strong>and</strong> Drug Delivery<br />

Conference, Philadelphia, PA.<br />

45. Mauludin, R., Möschwitzer, J., Müller, R. H.<br />

2005. Development of Effervescent <strong>and</strong> Pellet<br />

Formulations Containing Ibuprofen Drug<br />

<strong>Nanocrystals</strong> Produced by High Pressure<br />

Homogenization, AAPS Annual Meeting,<br />

Nashville, W5115.<br />

46. Möschwitzer, J., Müller, R. H. 2006. Spray<br />

coated pellets as carrier system for<br />

mucoadhesive drug nanocrystals, Eur. J.<br />

Pharm. Biopharm. 62, 282-287.<br />

47. Hanna, M. H., York, P. 1998. Method <strong>and</strong><br />

apparatus for the formulation of particles. US<br />

Patent 5,851,453.1998. USA.<br />

48. Hu, J. 2002. Improvement of dissolution rates<br />

of poorly water soluble APIs using novel spray<br />

freezing into liquid technology. Pharm. Res.,<br />

19, 1278–1284.<br />

49. Hu, J., Johnston, K. P., Williams, R. O. 3rd.<br />

2004. Nanoparticle engineering processes for<br />

enhancing the dissolution rates of poorly water<br />

soluble drugs. Drug. Dev. Ind. Pharm., 30,<br />

233–245.<br />

50. Rogers, T. L., Johnston, K. P., Williams, R. O.<br />

3rd. 2001. Solution-based particle formation of<br />

pharmaceutical powders by supercritical or<br />

compressed fluid CO2 <strong>and</strong> cryogenic sprayfreezing<br />

technologies. Drug Dev. Ind. Pharm.,<br />

27, 1003–1015.<br />

51. Pace, G. W. 2001. Process to generate<br />

submicron particles of water insoluble<br />

compounds. US Patent 6, 103. 2001. USA<br />

52. Young, T. J. 2000. Rapid expansion from<br />

supercritical to aqueous solution to produce<br />

submicron suspensions of water-insoluble<br />

drugs. Biotechnol. Prog. 16, 402–407.<br />

Vol. 3 (1) Jan – Mar 2012 www.ijrpbsonline.com 417


<strong>International</strong> <strong>Journal</strong> of Research in Pharmaceutical <strong>and</strong> Biomedical Sciences ISSN: 2229-3701<br />

53. Li, Z. 2008. Preparation of griseofulvin<br />

microparticles by supercritical fluid expansion<br />

depressurization process. Powder Technology.<br />

182, 459-465.<br />

54. Young, T. J. 1999. Encapsulation of lysozyme<br />

in a biodegradable polymer by precipitation<br />

with a vapor-over-liquid antisolvent. J. Pharm.<br />

Sci. , 88, 640–650<br />

55. Turk, M., Lietzow, R. 2004. Stabilized<br />

nanoparticles of phytosterol by rapid<br />

expansion from supercritical solution into<br />

aqueous solution. AAPS PharmSciTech. 2004 ,<br />

5(4). Article 56, 1-10.<br />

56. Chen, X. 2002. Preparation of cyclosporine A<br />

nanoparticles by evaporative precipitation into<br />

aqueous solution. Int. J. Pharm., 242, 3–14.<br />

57. Eickhoff, W.M., Engers, D.A., Mueller, K.R.<br />

1996. Nanoparticulate NSAID compositions,<br />

Application. US 95-385614, 5518738.<br />

58. Jacobs, C., Kayser, O., Müller, R.H. 2001.<br />

Production <strong>and</strong> characterisation of<br />

mucoadhesive nanosuspensions for the<br />

formulation of bupravaquone. Int. J. Pharm.<br />

214 (1-2), 3–7.<br />

59. Müller, R.H., Jacobs, C. 2002. Buparvaquone<br />

mucoadhesive nanosuspension: preparation,<br />

optimisation <strong>and</strong> long-term stability. Int. J.<br />

Pharm. 237 (1-2), 151–161.<br />

60. Hussain, N., Jaitley, V., Florence, A.T. 2001.<br />

Recent advances in the underst<strong>and</strong>ing of<br />

uptake of microparticulates across the<br />

gastrointestinal lymphatics. Adv. Drug Deliv.<br />

Rev. 50 (12), 107–142.<br />

61. Mauludin, R., Müller, R. H. 2006.<br />

Investigation of crystalline state of dry powder<br />

ibuprofen nanocrystals produced by high<br />

pressure homogenization technique, AAPS<br />

Annual Meeting, San Antonio.<br />

62. Kayser, O., Olbrich, C., Yardley, V., Kiderlen,<br />

A.F., Croft, S.L. 2003. Formulation of<br />

Amphotericin B as nanosuspension for oral<br />

administration. Int. J Pharm 254, 73-75.<br />

63. Vergote, G.J. 2001. An oral controlled release<br />

matrix pellet formulation containing<br />

nanocrystalline ketoprofen. Int. J. Pharm. 21;<br />

219(1-2), 81-7.<br />

64. Hanafy, A., Spahn, H., Vergnault, G., Grenier,<br />

P., Grozdanis, M.T., Lenhardt, T. 2007.<br />

Pharmacokinetic evaluation of oral Fenofibrate<br />

nanosuspension <strong>and</strong> SLN in comparison to<br />

conventional suspensions of micronized drug.<br />

Adv. Drug Del. Rev. 59, 419-426 .<br />

65. Singla, A.K., Garg, A., Aggarwal, D. 2002.<br />

Paclitaxel <strong>and</strong> its formulations. Int. J Pharm.<br />

235(1–2), 179–192.<br />

66. Peters, K., Leitjke, S., Diederichs, J.E.,<br />

Vorner, K., Hahn, H., Müller, R. H., Ehlers, S.<br />

2000. Preparation of a clofazimine<br />

nanosuspension for intravenous use <strong>and</strong><br />

evaluation of its therapeutic efficacy in murine<br />

Mycobacterium avium infection. J.<br />

Antimicrob. Chemother. 45, 77–83.<br />

67. Rainbow, B., Kipp, J., Papadopoulos, P.,<br />

Wong, J., Gass, J., Sun, C.S., Wielgos, T.,<br />

White, R., Cook, C., Barker, K., Wood, K.<br />

2007. Itraconazole IV nanosuspension<br />

enhances efficacy through altered<br />

pharmacokinetic in the rat. Int. J. Pharm. 339,<br />

251-260.<br />

68. Na, G.C., Stevens, Jr. J., Yuan, B.O.,<br />

Rajagopalan, N. 1999. Physical stability of<br />

ethyl diatrizoate nanocrystalline suspension in<br />

steam sterilization. Pharm. Res. 16(4), 569–<br />

574.<br />

69. Müller, R.H., Jacobs, C., Kayser, O. 2000.<br />

Nanosuspensions for the formulation of poorly<br />

soluble drugs. In: Nielloud F, Marti-Mestres<br />

G, eds. Pharmaceutical Emulsions <strong>and</strong><br />

Suspensions. New York: Marcel Dekker,<br />

S383–407.<br />

70. Moghimi, S.M., Hunter, A.C., Murray, J.C.<br />

2001. Long-circulating <strong>and</strong> target-specific<br />

nanoparticles: theory to practice. Pharmacol.<br />

Rev. 53, 283–318.<br />

71. P<strong>and</strong>ey, R., Khuller, G.K. 2004. Subcutaneous<br />

nanoparticle-based antitubercular<br />

chemotherapy in an experimental model. J.<br />

Antimicrob. Chemother. 54, 266–268.<br />

72. Müller, R.H., Jacobs, C. 2002. Production <strong>and</strong><br />

characterization of a budenoside<br />

nanosuspension for pulmonary administration.<br />

Pharm. Res. 19, 189-94.<br />

73. Hern<strong>and</strong>ez-Trejo, N., Kayser, O., Steckel, H.,<br />

Müller, R.H. 2005. Characterization of<br />

nebulised buparvaquone nanosuspension –<br />

effect of nebulization technology. J. Drug<br />

Target. 13(8-9), 499-507.<br />

74. Sharma, A., Sharma, S., Khuller, G.K. 2004.<br />

Lectin-functionalized poly (lactide-coglycolide)<br />

nanoparticles as oral/aerosolized<br />

antitubercular drug carriers for treatment of<br />

tuberculosis. J. Antimicrob. Chemother. 54,<br />

761–766.<br />

75. Peterson, R. 2006. <strong>Nanocrystals</strong> for use in<br />

topical cosmetic formulations <strong>and</strong> method of<br />

production thereof. US Patent Application No.<br />

60/8866, 233.<br />

76. Shegokar, R., Keck, C. M., Müller, R. H.,<br />

Gohla, S., 2011. Cosmetic nanocrystals:<br />

products <strong>and</strong> dermal effects, P 123, 6th Polish-<br />

German Symposium on Pharmaceutical<br />

Sciences: Perspectives for a new decade,<br />

Düsseldorf, 20-21 May 2011.<br />

77. Al Shaal, L., Shegokar, R., Müller, R. H.,<br />

2010. Novel UV skin protective antioxidant<br />

nanocrystals, AAPS Annual Meeting, W4089,<br />

New Orleans.<br />

78. Pignatello, R. 2002. Eudragit RS100<br />

nanosuspensions for the ophthalmic controlled<br />

Vol. 3 (1) Jan – Mar 2012 www.ijrpbsonline.com 418


<strong>International</strong> <strong>Journal</strong> of Research in Pharmaceutical <strong>and</strong> Biomedical Sciences ISSN: 2229-3701<br />

delivery of ibuprofen. Eur. J. Pharm. Sci., 16:<br />

53–61.<br />

79. Kassem, M. A. 2007. Nanosuspension as an<br />

ophthalmic delivery system for certain<br />

glucocorticoid drugs. Int. J. Pharm., 340: 126–<br />

133.<br />

80. Kayser, O. 2000. Nanosuspensions for the<br />

formulation of aphidicolin to improve drug<br />

targeting effects against Leishmania infected<br />

macrophages. Int. J. Pharm. 196, 253-256.<br />

81. Schroeder, U., Sommerfeld, P., Sabel, B.<br />

1998. Efficiency of oral dalargin-loaded<br />

nanoparticle delivery across the blood brain<br />

barrier. Peptides 19(4), 777-780.<br />

82. Lamprecht, A., Schafer, U., Lehr, C.M., 2001.<br />

Pharm. Res. 18, 788–793.<br />

83. Takeuchi, H.,Yamamoto, H., Kawashima,Y.,<br />

2001. Adv. Drug Deliv. Rev. 47, 39–54<br />

84. Ponchel, G., Montisci, M.-J., Dembri, A.,<br />

Durrer, C., Duchene, D., 1997. Eur. J. Pharm.<br />

Biopharm. 44, 25–31.<br />

85. Cevc, G., Gebauer, D., Stieber, J., Schatzlein,<br />

A., Blume, G., 1998. Biochim. Biophys. Acta<br />

1368, 201–215.<br />

86. Kayser, O. 2001. A new approach for<br />

targeting to Cryptosporidium parvum using<br />

mucoadhesive nanosuspensions: research <strong>and</strong><br />

applications. Int. J. Pharm. 214, 83-85.<br />

87. Kohno, S., Otsubo, T., Tanaka, E., Maruyama,<br />

K., Hara, K. 1997. Amphotericin B<br />

encapsulated in polyethylene glycol<br />

immunoliposomes for infectious diseases.<br />

Adv. Drug Del. Rev. 24, 325-329.<br />

Vol. 3 (1) Jan – Mar 2012 www.ijrpbsonline.com 419

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!