Techniques to Improve Bioavailability of
Poorly Water Soluble Drugs – A review
Amit J. Raval1*
and Madhabhai M. Patel2
1Research Scholar, Jodhpur
National University, Jodhpur and Amika Pharma, Mehsana, Gujarat, India.
2Kalol Institute of Pharmacy,
Kalol-382721, Gujarat, India.
ABSTRACT:
The
mark of a successful formulation depends on how efficiently it makes the drug
available at the site of action. Therapeutic effectiveness of a drug depends
upon the bioavailability and ultimately upon the solubility of drug molecules
especially in oral formulation. Formulating an oral dosage form for a poorly water soluble drug is in itself a challenging task. A
number of ways are used to improve solubility of drug by like salt formation,
co-solvency, and addition of solublizing agent, micronization, solid dispersion and complexation and many
more. Although these techniques have been used to increase dissolution rate of
the drug, there are practical limitations with these techniques, the desired
bioavailability enhancement may not always be achieved. In this review we have
tried to discuss advanced techniques that have come up to solve these problems.
Here in, novel technologies, such as sonocrystallisation,
nanotechnologies such as nanosuspension, nanoemulsion, spray freezing in to liquid and some
commercialized technologies such as nanocrystal, nanopure, nanoedge, biorise®, diffucaps®
etc. which present novel methods of solubilisation,
that may allow for greater opportunities to deliver poorly soluble drugs have
been dealt in detail. These techniques may help a great in facing the
challenges in formulating drugs with poor water solubility.
KEYWORDS: Bioavailability,
techniques, sonocrystallisation, nanotechnology, poor
water solubility, drug delivery system.
INTRODUCTION:
The term Bioavailability, one of the
principal pharmacokinetic properties of drugs, is used to describe the fraction
of an administered dose of unchanged drug that reaches the systemic
circulation. By definition, when a medication is administered intravenously,
its bioavailability is 100%. However, when a medication is administered via
other routes (such as oral), its bioavailability decreases (due to incomplete
absorption or first-pass metabolism).
The measurement of the
amount of the drug in the plasma at periodic time intervals indirectly
indicates the rate and extent at which the active pharmaceutical ingredient is
absorbed from the drug product and becomes available at the site of action.
Bioavailability is one of the essential tools in pharmacokinetics, as it must
be considered when calculating dosages for non-intravenous routes of
administration. It is expressed as either absolute or relative bioavailability.
Absolute Bioavailability:
Absolute bioavailability
measures the availability of the active drug in systemic circulation after
non-intravenous administration (i.e., after oral, rectal, transdermal, and
subcutaneous administration).
In order to determine
absolute bioavailability of a drug, a pharmacokinetic study must be done to
obtain a plasma drug concentration vs time plot
for the drug after both intravenous (i.v.) and
non-intravenous administration. The absolute bioavailability is the
dose-corrected area under curve (AUC) non-intravenous divided by AUC
intravenous. Therefore, a drug given by the intravenous route will have an
absolute bioavailability of 1 (F=1) while drugs given by other routes usually
have an absolute bioavailability of less than one.
Relative bioavailability:
This measures the
bioavailability of a certain drug when compared with another formulation of the
same drug, usually an established standard, or through administration via a
different route. When the standard consists of intravenously administered drug,
this is known as absolute bioavailability.
REASONS OF POOR
BIOAVAILABILITY1:
1. Poor aqueous
solubility:
Poor solubility of a drug is
in most cases associated with poor bioavailability. The contents of
gastrointestinal tract are aqueous and hence a drug having poor aqueous
solubility has a low saturation solubility which is typically correlated with a
low dissolution velocity, resulting in poor oral bioavailability.
2. Inappropriate
partition coefficient:
Too hydrophilic drugs would
not be able to permeate through the gastrointestinal mucosa and too lipophilic drug will not dissolve in the aqueous
gastrointestinal contents. For optimum absorption, the drug should have
sufficient aqueous solubility to dissolve in the gastrointestinal contents and
also adequate lipid solubility to facilitate its partitioning into the lipoidal membrane and then into systemic circulation. Drugs
having partition coefficient (log P) value in the range of 1 to 3 shows good
passive absorption across lipid membranes, and those having log Ps greater than
3 or less than 1 have often poor transport characteristics.
3. First-pass metabolism:
Orally administered drugs
must pass through the intestinal wall and then through the portal circulation
to the liver; both are common sites of first pass metabolism (metabolism of a
drug before it reaches systemic circulation). Thus, many drugs may be
metabolized before adequate plasma concentrations are reached resulting in poor
bioavailability.
4. Degradation in the
gastrointestinal tract:
Drug substances used as
pharmaceuticals have diverse molecular structures and are, therefore, prone to
many and variable degradation pathways. Protein drugs, in particular are highly
susceptible to inactivation due to the pH and the enzymes present in
gastrointestinal tract. This degradation can be owing to the low pH in the
stomach or due to chemical reactions taking place in the gastrointestinal tract,
Enzymatic degradation of drug in gastrointestinal
Tract or Interaction with food.
Drug absorption, sufficient
and reproducible bioavailability and/or pharmacokinetic profile in humans are
recognized today as one of the major challenges in oral delivery of new drug
substances. With the progress in medicinal chemistry
and, especially due to the recent introduction of combinatorial chemistry and
high-throughput screening in identifying new chemical entities (NCE), the
solubility of new drug molecules has decreased sharply. While a value of less than 20 μg/mL for the solubility of a NCE was practically
unheard of until the 1980s, the situation has changed so much that in the
present day drug candidates with intrinsic solubilities
(solubility of neutral or unionized form) of less than 1 μg/mL are very common. More than 90%
of drugs approved since 1995 have poor solubility. It is estimated that 40% of active new chemical entities
(NCEs) identified in combinatorial screening programs employed by many
pharmaceutical companies are poorly water soluble2,5.
Molecules of this type can
provide a number of challenges in pharmaceutical development and may
potentially lead to slow dissolution in biological fluids, insufficient and
inconsistent systemic exposure and consequent sub-optimal efficacy in patients,
particularly when delivered via the oral route of administration. Approximately
one-third of new compounds synthesized in medicinal chemistry laboratories have
an aqueous solubility less than 10 μg/mL, another one-third have a solubility from 10 to 100 μg/mL, and the solubility of
the remaining third is >100 μg/mL .
Advances in the
pharmaceutical sciences have led to the establishment of a number of approaches
for addressing the issues of low aqueous solubility. These strategies for
improving and maximizing dissolution rate include micronisation
to produce increased surface area for dissolution, the use of salt forms with
enhanced dissolution profiles, solubilisation of
drugs in co-solvents and micellar
solutions, complexation with cyclodextrins and the use of lipidic systems for the
delivery of lipophilic drugs. Although these techniques have been shown to be
effective at enhancing oral bioavailability, the success of these approaches is
dependent at times on the specific physicochemical nature of the molecules
being studied3,4.
A variety of formulation
strategies have been developed to improve the solubility and, thus, the
bioavailability of such compounds. Formulation strategies include
self-dispersing and self-emulsifying formulations, solid solutions and
dispersions, ionic inclusion and lipid-based complexation, formation of salts,
polymorphs, cocrystals, and prodrugs,
and particle size reduction techniques such as micronization
or nanomilling. The techniques that are used to
overcome poor drug solubility and hence bioavailability in turn can be broadly
categorized under two major headings: chemical modifications and Physical
modifications. Chemical modifications include techniques like – Salt formation,
co-crystallization, Co-solvency, Hydrotropy, Use of solubilizing agent, Nanotechnology etc. whereas Physical
modifications include techniques like-Particle size reduction (Micronization, nanosuspensions),
Modification of Crystal habit (Polymorphs and Pseudopolymorphs),
Complexation, Solubilization by surfactants (Microemulsions, SMEDDS), Drug dispersion in carriers etc5.
Solubilisation technologies such as micellar
systems are reliant on the acceptable solubility and compatibility of
therapeutic molecules in a limited range of pharmaceutically acceptable excipients, whilst the increasing number of weakly ionisable and neutral molecules entering development
constrains the opportunities for salt formation as a method of improving
dissolution rate. Furthermore, whilst micronisation
increases the dissolution rate of drugs through increased surface area, it does
not increase equilibrium solubility. Often for drugs with very low aqueous
solubility, the achieved increase in dissolution rate is insufficient to
provide adequate enhancement of bioavailability. The salt formation is not
feasible for neutral compounds and the synthesis of appropriate salt forms of
drugs that are weakly acidic or weakly basic may often not be practical4.
Much of the research
that has been reported on solid dispersion technologies involves drugs that are
poorly water-soluble and highly permeable to biological membranes as with these
drugs dissolution is the rate limiting step to absorption. Hence, the
hypothesis has been that the rate of absorption in vivo will be concurrently
accelerated with an increase in the rate of drug dissolution. Physical
modification has also been utilized to improve the solubility of poorly water soluble drugs. For such physical modifications,
various excipients such as cyclodextrins,
carbohydrates, hydro tropes, polyglycolized glycerides, and dendrimers are
utilized6.
All the approaches used
to counteract the solubility and bioavailability challenges, discussed above
have some limitations. Nowadays numbers of advanced techniques have come up to
solve these problems and the commercialization and scale-up has also been done
in few of the cases.
Few such techniques
have been discussed below:
1. Melt
Sonocrystallization7:
Melt sonocrystallization a novel particle engineering technique
to enhance dissolution of hydrophobic drugs and to study its effect on crystal
properties of drug. Particle engineering techniques are developing to modify
the physicochemical, micromeritic and
biopharmaceutical properties of the drug. Number of particle
design techniques are reported, such as spherical crystallization,
extrusion spheronization, melt solidification, spray
drying, pastillation, solution atomization and
crystallization by sonication (SAXS), where simultaneous crystallization and
agglomeration occur.
Ultrasound (US) was
introduced in the traditional process of pharmaceutical technology of few years
ago. For instance, several workers reported US assisted compaction and US spray
congealing of variety of systems where physical modification of structure of
drug or excipients was done to improve drug release
and compaction properties of drug. Besides these effects on solid, US may also
act on a liquid or melt mixtures causing cavitation
and extreme molecular motion, which divides the drop of material into number of
microdrops of narrow size range. One of the
mechanical effects cause by ultrasonification is
disaggregation or deagglomeration of the particle
assembling. Cavitation is an important phenomenon of ultrasonication. The energy produced due to the collapse of
bubbles at very high temperature was responsible for breaking of particles.
The so generated shock
waves can cause the particle collide into one another with great force since
these are similar charge particles problem of agglomeration is greatly reduced
There are reports on application of ultrasonic (US) energy during
crystallization, i.e. sonocrystallization. US energy
has been used to achieve nucleation at moderate super saturation during the
crystallization process or terminal treatment to achieve deagglomeration
and to obtain desired crystal habit.
The drug with poor
solubility is melted in a paraffin bath and the molten mass is poured in a
vessel containing deionized water maintained in
heated condition using thermostatic water bath and sonicated
using probe ultrasonicator. The product obtained
after solidification of dispersed droplet is separated by filtration and dried
at room temperature.
Melt sonocrystallization, which is combination of melt
solidification and ultrasonication, has advantages of
melt solidified bonds and hard surface. This hardened surface has enabled the
particles to withstand high sonication shear and maintain integrity even in
highly porous form. Ultrasonication has been reported
to cause spontaneous nucleation at relatively low degrees of super saturation,
due to increase in number of collisions. Similarly, ultrasonication
enhanced collisions in molecules of the melt favors nucleation rather than
crystallization. Processing temperature is an important factor in the design of
the technique.
Application of
ultrasonic energy to the molten mass significantly increases kinetic energy of
the molecules and number of collisions causing faster crystallization of the
drug. Chaudhari P.D. et.al. have prepared and
studied Valdecoxib agglomerates comprising of
irregular in shape having rough surface with pores obtained by MSC technique,
The MSC Valdecoxib agglomerate has shown some number
of shallow circular pits on the surface, cracks in the crystals of the drug and
has shown significantly higher specific surface area and thereby increase in
saturated solubility.
2. Precipitation
Ultrasonication Method8,9:
The bioavailability of
the solid API having poor aqueous solubility can be improved by formulating
them as nanosuspensions. The dissolution rate of API
is proportional to the surface area available for dissolution as described by
the Noyes–Whitney equation and, in addition to the dissolution rate enhancement, an increase in the solubility of nanosized API is also expected. The nanocrystals
can be obtained either by particle size reduction of larger crystals (top-down
approach) such as high-pressure homogenization and media milling or by building
up crystals by precipitation of dissolved molecules (bottom-up approach).
Regarding the bottom-up method, in the last decade, supercritical fluid-based
techniques have been widely investigated to obtain nanosized
drug particles. In the last decade, ultrasound has received much attention and
has been used as an effective method of controlling the nucleation and
crystallization process. Ultrasound irradiation has been proved to be a
feasible mixing method and it can intensify mass transfer and accelerate
molecular diffusion.
In order to obtain
crystals with an increased dissolution rate and, thus, to enhance the
bioavailability, the particle size has to be reduced. Stable nanosuspensions were prepared by the precipitation–ultrasonication method. The process parameters that affect
the process are concentration of API in the organic phase, the power input and
the time length of ultrasonication.
For the preparation of nanosuspension of the API with poor aqueous solubility, the
API is dissolved in organic solvent. PVA dissolved in water is used to obtain antisolvent as given in figure 1. Both the solutions are
passed through a 0.45µm filter. The anti-solvent is cooled to below 3 ºC in an ice-water bath. Then, the organic solution is
quickly introduced into the precooled anti-solvent
with stirring at high speed. After the anti-solvent precipitation, the samples
are immediately transferred to a test tube and treated with an ultrasonic probe
at specific ultrasonic power input for specific time length.
Figure 1: Flow chart of the precipitation-ultrasonication procedure for the preparation of nanosuspension.
During the process, the
temperature is controlled using an ice-water bath. The obtained nanosuspensions are concentrated by centrifugation at very
high speeds using an ultracentrifuge. After the centrifugation, the supernatant
is replaced with 0.2% PVA solution. The solid residue is redispersed
using a bath sonicator and the final drug content is
adjusted to required strength using an appropriate volume of 0.2% PVA solution.
Dengning Xia et.al.
have prepared Nitrendipine nanosuspensions using the precipitation ultrasonication
method. They found that particle size of nanocrystals
was highly dependent on process parameters. With the optimized process
parameters, nanosuspensions with diameter of about
209nm (±9 nm) could be obtained. No substantial crystalline
change was found in nanocrystals compared with raw
crystals. Marked enhancement of dissolution rate was achieved by the reduction
in particle size. The oral bioavailability of Nitrendipine
in Wistar rats resulted from nanosuspension was found
to be increased by 5.0-fold compared with the commercial tablets. The advantage
of this method being it is easy to apply and needs only simple equipment and,
thus, is a promising method for preparing drug nanosuspensions.
3. LIQUISOLID
Technique10,11,12:
A liquisolid
system has the ability to improve the dissolution properties of poorly water soluble drugs. Liquisolid
compacts are flowing and compactable powdered forms of liquid medications. Several
studies have shown that the liquisolid technique is a
promising method for promoting dissolution rate of poorly
water soluble drugs. In liquisolid compact, a liquid
medication is converted into acceptably flowing and compactible
powder forms. The term ‘liquid medication’ implies liquid lipophilic
(oily) drug and solution or suspension of poorly water soluble drugs carried in
suitable water miscible non-volatile liquid systems termed the liquid vehicle.
By simple blending with suitable excipients ‘carrier
and coating materials’, the liquid medication may be converted into a
dry-looking, non-adherent, free flowing and readily compactible
powder.
Since
drug dissolution is often the rate limiting step in gastrointestinal
absorption, the significant increase in wetting properties and surface area of
drug particles available for dissolution from liquisolid
compacts may be expected to display enhanced drug release characteristics and,
consequently, improved oral bioavailability. The technique of liquisolid compacts has been successfully employed to improve the in vitro release of
poorly water soluble drugs such as Carbamazepine, Famotidine, Piroxicam, Indomethacin, Hydrocortisone, Naproxen and Prednisolone etc.
The drugs which are
water insoluble or liquid lipophilic are dissolved in
a suitable selected non-volatile solvent. This non-volatile solvent with drug
dissolved may exist either in solution or else suspension form known as “liquid
medicament”. The liquid medicament is converted into free flowing, non-adherent,
dry form and readily compressible powders with the help of different
compressible carriers like (Starch, cellulose and lactose etc.) and other
coating materials like (Colloidal silica and Talc etc.). Because of drug being
present in the liquid medicament as solubilized or
molecularly dispersed state, the dissolution is enhanced due to increased
surface area as well as wetting area. There by the Liquisolid
technique is applied for water insoluble drugs to enhance dissolution rate, it
may also increase bioavailability in turn.
4. Nanocrystal technology:
The pharmaceutical
benefits of nanocrystals include improvement in
formulation performance, such as enhanced dissolution velocity and saturation
solubility, reproducibility of oral absorption, improved dose-bioavailability,
proportionality and increased patient compliance via reduction of number of
oral units to be taken. Nanocrystal serves as ideal
delivery system for oral drugs having the dissolution velocity as rate limiting
step for absorption, i.e. drugs of the biopharmaceutical classification system
(BCS) class II and IV. In addition, nanocrystals can
be injected intravenously as aqueous nanosuspensions.
Two basic approaches
are involved in production of nanocrystals, the
bottom–up technologies (controlled precipitation/ crystallization) and the
top–down technologies, nanonizing (large-size drug
powder to be reduced in size, e.g. by mechanical attrition).
4.1 Bottom-up techniques13,14,15:
Around 1980
Sucker developed the so called “hydrosols”, the intellectual property acquired
by Sandoz (nowadays Novartis). This technology is basically a classical
precipitation process where drug is
dissolved in a solvent and subsequently precipitated by mixing with a
non-solvent. It yields crystalline drug nanoparticles.
This method requires strict control of the process, avoidance of crystal growth
(to the micrometer range), drug solubility in at least one solvent, and of
course has the problem of solvent residues.
Another
precipitation process was developed by Auweter and
Horn, leading to amorphous nanoparticles of the active. The particles are spherical due to
precipitation process. This process is used by BASF for products developed in
the food sector (e.g. Lucarotin® or Lucantin® which is a solution of the carotenoid,
together with a surfactant in a digestible oil), and for pharmaceuticals by Soliqs® (AbbottGmbHandCo. KG, Ludwigshafen) previously Knoll/BASF. The Soliqs trade name is NanoMorph®.
4.2 Top-down techniques:
(a) NanoCrystals® technology[16,17]:
The NanoCrystals® technology by élan (prev. Nanosystems)
uses a bead/pearl mill to achieve particle size diminution. It was developed by
Liversidge et al. Milling
media, dispersion medium (generally water) containing stabilizer along with
drug are charged into a milling chamber. Shear forces generated by the movement
of the milling media lead to particle size reduction. Smaller or larger coated
milling pearls of ceramics (cerium or yttrium stabilized zirconium dioxide), stainless steel, glass or highly crosslinked
polystyrene resin-coated beads can be used. Erosion from the milling material
during the milling process is a common problem of this technology. The milling
time mainly depends on the hardness of the drug, viscosity, temperature, energy
input, size of the milling media and surfactant concentration used. The milling
time can last from about 30 min to hours or several days. This is an important
industrially used technology for particle size reduction, proven by the
FDA-approved products.
Lab scale production
can be carried out at using 100mg or less of API by using the Nanomill® system (élan Drug Discovery, PA, USA). The
chemical form of API needs to be considered for laboratory testing, typically
the neutral form is preferred. Production volumes of more than 5 L (flow
through mode) can be produced using the Dynomill
(Glen Mills, Inc. Cliffton, NJ, USA) with chamber
size of 300 and 600 mL. Also larger sized mills are
available (e.g. Netzsch mills (Netzsch
Inc., Exton, PA, USA)), e.g. in 2, 10 and 60 L chamber size.
(b) Microfluidizer technology (IDD-PTM technology)18:
The Microfluidizer technology is based on the jet stream
principle and can generate small particles by a frontal collision of two fluid
streams in a Y-type or Z-type chamber under pressures up to 1700 bar.
Figure 2: Working
of Microfluidizer processor
The jet
streams lead to particle collision, shear forces and cavitation
forces (Microfluidizer®, Microfluidics
Inc.). working of Microfluidizer
processor is shown in figure 2. Often a relatively high number of cycles
(50–100 passes) are necessary to obtain sufficient particle size reduction. SkyePharma Canada Inc. (formerly Canadian company Research
Triangle pharmaceuticals (RTP)) uses this principle for their Insoluble Drug
Delivery-Particles (IDD-PTM) technology to achieve the production of submicron
particles of poorly soluble drugs. Product on the market is Triglide®,
fenofibrate.
(c) Dissocubes® technology19-28:
The Dissocubes® technology was developed by Müller
and co-workers by employing piston-gap homogenizers (e.g. APV Gaulin/Rannie homogenizers). The
technology was acquired by SkyePharma PLC in 1999. A
drug dispersed in an aqueous surfactant solution (macrosuspension)
is forced by a piston under pressure (up to 4000 bar, typically 1500–2000 bars)
through a tiny gap (e.g.5–20µm). The resulting high streaming velocity of the
suspension causes an increase in the dynamic pressure. This is compensated by a
reduction in the static pressure below the vapor pressure of the aqueous phase;
hence, water starts boiling forming gas bubbles. These gas bubbles collapse
immediately when the liquid leaves the homogenization gap (=cavitation).
The drug particles are reduced in size due the high power of the shockwaves
caused by cavitation. The mean size of bulk population
obtained for the high pressure homogenization process depends on the power
density of the homogenizer (homogenizer pressure), number of homogenization
cycles and hardness of drug. Because of crystalline nature they appeared in cuboid or irregular shape, in contrast to spherical amorphous drug nanoparticles.
(d) Nanopure® technology21:
The Nanopure®
technology is another approach using the piston-gap homogenizer (prev. PharmaSol GmbH, now Abbott). This technology uses a primary
dispersion medium, non-aqueous liquids, e.g. oils, liquid and solid (melted)
PEG, or water reduced media (e.g.
glycerol–water, ethanol–water mixtures), and optionally homogenization at low temperatures. These media have low vapor pressure,
cavitation takes place very limited or not at all. At
homogenization at room temperature, the water starts boiling, i.e. the static
pressure on the water is reduced to the vapor pressure of water at 20 ◦C, being 23.4. For example, the vapor
pressure of Miglyol 812 oil is only 0.01 hPa (=0.01 mbar) at 20 ◦C, i.e. more than 2000 fold lower. Therefore when water
shows cavitation, the oil will not. Even without cavitation, the size diminution is sufficient because of
shear forces, particle collisions and turbulences. The optional low
temperatures allow the processing of temperature sensitive drugs, in addition
at lower temperatures materials, are more fragile. Final nanosuspensions
product in oil or PEG can be directly filled into gelatin or HPMC capsules.
With nanopure technology, homogenization can be performed in a nonaqueous phase or phases with reduced water content. And,
in contrast to more pronounced cavitation at higher
temperatures, homogenization is similar or more efficient at lower
temperatures, even below the freezing point of water.
(e) NANOEDGETM
technology3,21:
This is a
type of combination technology where a combination of a pre-treatment step with
a subsequent high energy steps, for
example – but not necessarily – high pressure homogenization is done. The NANOEDGETM technology by Baxter uses a first
classical precipitation step with a subsequent annealing step by applying high
energy, e.g. high pressure homogenization. According to the patent claims, the
annealing step prevents the growth of the precipitated nanocrystals.
Annealing is defined in this invention as the process of converting matter that
is thermodynamically unstable into a more stable form by single or repeated
application of energy (direct heat or mechanical stress), followed by thermal
relaxation. This lowering of energy may be achieved by conversion of the solid
form from a less ordered to a more ordered lattice structure. Alternatively,
this stabilization may occur by a reordering of the surfactant molecules at the
solid–liquid interface.
The technique is based
on direct homogenization, microprecipitation, and
lipid emulsions. In microprecipitation, the drug
first is dissolved in a water-miscible solvent to form a solution. Then, the
solution is mixed with a second solvent to form a presuspension
and energy is added to the presuspension to form
particles having an average effective particle size of 400 nm to 2 μ. The
energy-addition step involves adding energy through sonication, homogenization,
countercurrent flow homogenization, microfluidization,
or other methods of providing impact, shear, or cavitation
forces. A drug suspension resulting from these processes may be administered
directly as an injectable solution, provided
water-for-injection is used in the formulation and an appropriate means for
solution sterilization is applied. Nanoedge
technology facilitates small particle sizes (<1000 nm [volume weighted
mean]), high drug loading (10–200 mg/mL), long-term
stability (up to 2 years at room temperature or temperatures as low as 5 °C),
the elimination of co solvents, reduced levels of surfactants, and the use of
safe, well-tolerated surfactants.
(f) SmartCrystal® technology:
The smartCrystal®
technology is owned by Abbott and marketed by its drug delivery company Soliqs in Ludwigshafen/Germany. It is a family of various
combination processes, a kind of tool boxes to
tailor-make the nanocrystals for each specific
application, and considering the physical properties of the drug (e.g.
hardness). It was acquired from PharmaSol GmbH in
2007, and these crystalline nanoparticles complement
to the amorphous nanoparticles by Soliqs,
NanoMorph®. The smartCrystal
technology is considered as the second generation of drug nanocrystals.
Injecting nanosuspensions with very small nanocrystals can permit fast dissolution and mimic
injection of a solution.
5. Biorise®
Technology[29]:
Eurand’s Biorise technology exploits
the much faster dissolution rate of nanocrystalline
and amorphous forms of drugs when compared to unmodified or even micronized
forms. As shown in figure 3 Biorise creates new physical
entities (NPEs) by physically breaking down a drug’s crystal lattice, resulting
in drug nanocrystals or amorphous drug with enhanced solubilization properties, faster absorption and
ultimately, increased absolute bioavailability. These NPEs are then stabilized
in an inert biological carrier, generally a polymer, to prevent the processed
drug from agglomerating or reverting back to its crystalline form.
Figure 3: Biorise
Technology
Eurand has two alternative activation systems available that
are used to convert a drug into its thermodynamically activated state. These
systems provide flexibility and allow the technology to be applied to a range
of compounds with differing characteristics:
1.
High Energy
Mechanochemical Activation (HEMA):
This involves
the application of friction and impact energy to the drug, thereby increasing
its entropy and transforming the drug into its activated state. This is a dry
system, which maintains the drug/carrier matrix in a powder form at all times.
2.
Solvent
Induced Activation (SIA):
This technique is particularly suitable for thermolabile
compounds and compounds with a low melting point. With this system, a drug can
be solubilized in an appropriate solvent and layered
on to swellable, crosslinked
carriers. Controlled evaporation of the solvent and drying of the material
creates nanoparticle and amorphous drug that are
stabilized in a carrier. Compared to other solubility enhancing technologies, Eurands’ Biorise system offers
faster and more efficient processing times, is cost effective, and produces a
stable product without the use of surfactants. The finished product is a drug
powder, which can be incorporate into a variety of dosage forms including
tablets and capsules.
6. Diffucaps® Technology29:
Eurand’s Diffucaps technology (figure
4) is a flexible multiparticulate system that
provides optimal release profiles for single drugs and drug combinations to
reliably overcome the problems associated with pH-dependent insolubility
encountered in the GI tract. This proprietary technology has been developed
specifically for weak, basic drugs and involves the incorporation of
pharmaceutically acceptable organic acid or a crystallization-inhibiting
polymer onto inert cores and coating the drug-layered beads with proprietary functional
polymers.
Figure 4: Diffucaps
Technology
Formulations using an acid
core ensure an acidic environment surrounds the drug at all times, thereby
producing a soluble drug in an in vivo environment where it would
otherwise be insoluble. Further, Diffucaps
incorporates release-controlling polymers, a functional drug layer, core
granules or crystals, and a protective coating in one technology, providing
sophisticated control of drug delivery timing that goes beyond what a single
technology system can provide. Drug beads are typically created by layering
active drug onto a neutral core (such as sugar spheres or cellulose spheres)
and applying one or more rate-controlling, functional membranes. The drug
layering process can be conducted either from aqueous or organic solvent-based
drug solutions/suspensions and results in a small (approximately 1.5 mm or less
in diameter), spherical, and multilayered bead particle. The beads may then be
filled into capsules or compressed into orally disintegrating tablets to create
the final dosage form. The inherent flexibility of the Diffucaps
system permits the easy adjustment of both dosage strength and pharmacokinetic
profile to achieve the required in vivo results. Beads can have
different release profiles, different active ingredients, or both – all in one
product.
7. Oral synchronous drug delivery system [30] :
Delivery Therapeutics Ltd
(DT), an Israeli biotechnology
company, has developed a proprietary technology enabling an efficient oral delivery of poorly absorbed drugs, mainly protein drugs and poorly-water soluble drugs.
It is a novel approach to increase the bioavailability of protein drugs, poorly
absorbed drugs and drugs susceptible to efflux pumping incidents. This approach
has been tested over 4 years in a variety of biological models and resulted in
the design of the oral
synchronous delivery system (OSD).
The OSD proprietary (patent
allowed in Israel, patent pending in the USA) matrix is a tailor-made
platform, capable of synchronizing
the release process of drug(s) together with functional additives such as
absorption enhancers (AE); enzyme inhibitors (PI), and efflux pump inhibitors
[e.g. p-glycoprotein (pGP)], over a predetermined
time interval along specific locations in the intestine. In other words: the
matrix is designed to cause a concomitant release of two ingredients or more
and “forces” them to stay together, while traveling down the gut. The
concomitant release provided by the OSD allows continuous action of the
functional adjuvant (absorption enhancer, enzyme inhibitor, or solubilizing agent) throughout the whole length of the
intestine, or at pre-designed intestinal segments over predetermined time
slots. The outcome is an improved
bioavailability of the drug of interest compared with the bioavailability
accomplished when administered in non-synchronous carriers (Figure 5 below).
Figure 5: concept of Oral
synchronous drug delivery system
8. Cryogenic processing techniques:
8.1 Spray freezing into liquid(SFL)31-34:
Cryogenic processing techniques
have been developed to enhance the dissolution rate by creating nanostructured amorphous particles with high degrees of
porosity. The cryogenic spray-freezing into liquid (SFL) process produces
amorphous solid solutions of drug and excipients. The
formation of metastable amorphous solid solutions
yields higher energy states for the drug and thus a greater thermodynamic
driving force for dissolution.
Figure 6: Schematic diagram of
the SFL apparatus illustrating the solution cell (A), high pressure pump (B),
atomizing nozzle (C), and the cryogenic liquid cell (D).
SFL technology is a
cryogenic particle engineering process that utilizes the atomization of a feed
solution containing APIs and/or excipient(s) directly
into a cryogenic liquid to produce frozen nanostructured
particles. The frozen particles are then lyophilized to obtain dry, free
flowing micronized powders. Advantages of the SFL process result from intense
atomization in conjunction with rapid freezing rates. Because liquid–liquid
impingement occurs between the pressurized feed solution exiting the nozzle and
the cryogenic liquid, such as liquid nitrogen, the highest degree of
atomization is achieved by spraying directly into the cryogenic liquid as
opposed to spraying into the vapor phase above the cryogenic liquid.
SFL particles have been
shown to have a large specific surface area, producing powders with rapid
dissolution. Additionally, the SFL process produces powders that are consistent
with a solid solution. SFL powders are formulated with small amounts
of surfactant to achieve high drug loadings (50%-86% drug/total solids) while
maintaining high dissolution rates. SFL powders require smaller amounts of
surfactant to achieve high dissolution rates. These high-drug-loaded
SFL powders contain amorphous nanostructured
aggregates with high surface area and excellent wettability.
A schematic diagram of the SFL apparatus is shown in figure 6.
8.2 Spray freeze drying:
Another cryogenic process,
the spray-freeze-drying (SFD) method, typically involves the atomization of a
drug-containing solution in gaseous nitrogen above a pool of liquid nitrogen.
The fine droplets of drug/solvent are frozen, then lyophilized to remove the
solvent. The rapid freezing rates in the cryogenic liquid substrate do not
allow for molecular arrangement into crystalline domains, so SFD processing
produces amorphous drug nanoparticle aggregates with
improved dissolution rates. The scalability of this type of
technology, however, has limited its widespread industrial use.
8.3 Ultra-rapid Freezing (URF)35:
Ultra-rapid freezing (URF)
technology involves the use of a solid cryogenic substrate with a thermal
conductivity between 10 and 20 W/m degrees K. A solution of the drug is applied
to the solid surface of the substrate, where instantaneous freezing takes
place. Brownian motion of the particles in solution is slowed significantly, so
reactive species have little time to react before being frozen into the solid
state. Removal of the frozen particles and lyophilization
of the solvent produces stable amorphous drug particles. URF technology has
been shown to produce uniform, amorphous, drug particle/excipient
aggregates. Additionally, the process is continuous, allowing for
improved scale-up applications. A reservoir of boiling cryogenic liquid is not
required, allowing for lower operating costs and more convenient operation.
Numerous citations report solvent/drug/excipient compositions being frozen in liquid or gaseous
nitrogen or other cryogenic fluids. All of these approaches face the same
challenge in transferring the heat necessary to cool and freeze the solution
forming the drug particle domains. The heat transfer is forced to pass through
a gas film at the surface of the particle. This imparts a rate-limiting step in
the heat transfer and defines the maximum freeze rate.
TABLE 1: Marketed preparations of Nanocrystal
technology
|
Trade name |
Therapeutic
use |
Applied
technology |
Pharma company |
|
Rapamune® (Rapamycin, Sirolimus) |
Immunosuppressive |
élan nanosystems |
Wyeth Pharmaceuticals |
|
Emend® (Aprepitant) |
Antiemetic |
élan nanosystems |
Merck and Co. |
|
Tricor® (Fenofibrate) |
Hypercholesterolemia |
élan nanosystems |
Abbott Laboratories |
|
Triglide® (Fenofibrate) |
Hypercholesterolemia |
IDD-P® technology |
Produced by SkyePharma
marketed by Sciele Pharma
Inc. (Atlanta, CA, USA). |
|
Megace ES® (Megestrol acetate) |
Antianorexic |
élan nanosystems |
Par Pharmaceutical Companies Inc. (Spring Valley, NY, USA) |
|
Avinza® (Morphine sulfate) |
Psychostimulant |
élan nanosystems |
King Pharmaceuticals |
|
Focalin® XR (Dexmethyl-phenidate HCl) |
Psychostimulant |
élan nanosystems |
Novartis |
|
Ritalin® LA (Methylphenidate HCl) |
Psychostimulant |
élan nanosystems |
Novartis |
|
Zanaflex CapsulesTM (Tizanidine HCl) |
Muscle relaxant |
élan nanosystems |
Acorda |
URF technology overcomes the limitation of transferring
heat through a gas film by eliminating the gas interface element and using
direct contact with the cryogenic substrate. Drug solutions that come into
direct contact with a boiling liquid cryogenic substrate transfer heat through
a gas bubble film until the temperature of the particle comes into thermal
equilibrium with the liquid at its boiling point. Note that
conduction is improved by using a cryogenic material with a high thermal
conductivity, density, and mass relative to the solution being frozen so as to
maintain the surface temperature and heat transfer rate while the solution is
being frozen. With URF technology, the thickness of the freezing solution may
be controlled to fix its minimum freezing rate since the freezing rate drives
the particle formation and determines the freezing solution's characteristics
and, hence, drug particle formation.
Purvis T et.al. prepared rapidly dissolving
formulations of the poorly water-soluble drug Repaglinide
using ultra-rapid freezing (URF). URF processing created nanostructured
drug/excipient particles with higher dissolution
rates than were achieved for unprocessed drug. It was found that URF
processing, yielded fast-dissolving formulation that was physically and
chemically stable, resistant to alkali degradation or spontaneous recrystallization in the formulation.
CONCLUSION:
Poor bioavailability is a major limitation in
successful drug delivery by oral route. It is rapidly becoming the leading
hurdle for formulation scientists working on oral delivery of drugs. Lot of
research work is focused on oral bioavailability enhancement of the poorly
absorbed drugs. Growing percentage of NCEs displaying solubility issues demands
that technologies for enhancing drug solubility be developed to reduce the
percentage of poorly soluble drug candidates eliminated from development as a
result. Drug cyclodextrin inclusion complexes,
surfactant addition and particle size reduction via comminution,
spray drying and solvent recrystallisation, possess
significant limitations on the extent to which they may solubilise
insoluble and nearly insoluble compounds. Novel technologies, such as sonocrystallisation, nanotechnologies such as nanosuspension, nanoemulsion,
spray freezing in to liquid and some commercialized technologies such as nanocrystal, nanopure, nanoedge, biorise®, diffucaps® etc. present novel methods of solubilisation that may allow for greater opportunities to
deliver poorly soluble drugs.
REFERENCES:
1.
Thakkar H,
Patel B and Thakkar S. A Review on techniques for
oral bioavailability enhancement of drugs. International Journal of Pharmaceutical Sciences Review and Research. 4(3); 2010: 33.
2.
Abu TS. Salt formation to improve drug solubility. Advanced Drug Delivery
Reviews. 59; 2007: 603–616.
3.
Sharma S and Aggarwal
G. Novel technologies for oral delivery of poorly soluble drugs. Research
Journal of Pharmaceutical, Biological and Chemical Sciences. 2010: 292-305.
4.
Blagden N, de Matas M, Gavan PT
and York P. Crystal engineering of active pharmaceutical ingredients to improve
solubility and dissolution rates. Advanced
Drug Delivery Reviews. 59; 2007: 617–630.
5.
Singh MC, Sayyad AB
and Sawant SD. Review on various techniques of
solubility enhancement of poorly soluble drugs with special emphasis on solid
dispersion. Journal of Pharmacy
Research. 3(10); 2010.
6.
Tiwari R, Tiwari G, Srivastava B and Rai
AK. Solid Dispersions: An Overview To Modify
Bioavailability Of Poorly Water Soluble Drugs. International Journal of PharmTech Research. 1(4); 2009: 1338-1349.
7.
Chaudhari PD and Uttekar PS. Melt-Sonocrystallization: A Novel Particle Engineering Technique
for Solubility Enhancement. International
Journal of PharmTech Research. 1(1); 2009:
111-120.
8.
Xia D, Quan P, Piao
H, Piao H, Sun S, Yin Y and Cui F. Preparation of stable Nitrendipine Nanosuspensions
using the Precipitation-Ultrasonication method for
enhancement of dissolution and oral bioavailability. European Journal of Pharmaceutical Sciences. 40; 2010: 325–334.
9.
de Castro ML and Priego-Capote
F. Ultrasound-assisted crystallization (sonocrystallization).
Ultrason. Sonochem.
14; 2007: 717–724.
10.
Kumar SK, Suria PK, Satish K, Satyanarayana K and
Kumar RH. Solubility Enhancement of a Drug By LIQUISOLID Technique. International Journal of Pharma and Bio Sciences. 1(3); 2010.
11.
Akinlade B, Elkordy AA, Essa EA and Elhagar S. Liquisolid Systems
to Improve the Dissolution of Furosemide. Scientia Pharmaceutica; Sci Pharm. 78; 2010: 325–344.
12.
Fahmy RH and Kassem MA.
Enhancement of famotidine dissolution rate through liquisolid tablets formulation: In vitro and in vivo
evaluation. European Journal of
Pharmaceutics and Biopharmaceutics. 69; 2008: 993–1003.
13.
Sucker H and Gassmann P. Improvements in
pharmaceutical compositions. GB Patent
2269536A. 1994.
14.
Auweter H, Andre V, Horn D and Luddecke
E. The function of gelatin in controlled precipitation processes of nanosize particles. Journal
of Dispersion Science Technology. 1998: 163–184.
15.
Auweter H, Bohn H, Heger R, Horn
D, Siegel B and Siemensmeyer K. Precipitated
water-insoluble colorants in colloid disperse form. US Patent 6,494,924. BASF AG. 2002.
16.
Liversidge GG and Cundy KC. Particle
size reduction for improvement of oral bioavailability of hydrophobic drugs: I.
Absolute oral bioavailability of nanocrystalline danazol
in beagle dogs. International Journal of
Pharmaceutics. 125; 1995: 91–97.
17.
Liversidge GG, Cundy KC, Bishop J and
Czekai D. Surface modified drug nanoparticles.
US Patent 5145684. 1992.
18.
Haynes DH. Phospholipid-coated microcrystals: Injectable
formulations of water insoluble drugs. US
Patent 5091187. 1992.
19.
Insoluble Drug
Delivery Platform. Available from: URL: http://www.skyepharma.com.
20.
Müller RH and Akkar A. Drug nanocrystals of poorly soluble drugs. In Encyclopedia of Nanoscience and Nanotechnology. Edited by: Nalwa HS. American Scientific Publishers. 2004; 627–638.
21.
Shegokar R and Müller RH. Nanocrystals: Industrially feasible multifunctional
formulation technology for poorly soluble actives. International Journal of Pharmaceutics. 399; 2010: 129–139.
22.
Müller RH, Becker R and Kruss KP.
Pharmaceutical nanosuspensions for medicament
administration as systems with increased saturation solubility and rate of
solution. US Patent 5858410. 1992.
23.
Müller RH, Böhm BHL, Grau MJ. Nanosuspensions—a formulation approach
for poorly soluble and poorly bioavailable drugs. In:
Handbook of Pharmaceutical Controlled Release. Edited by: Wise DL. Marcel
Dekker, New York. 2000; 345–357.
24.
Müller RH, Jacobs C and Kayser O.
Nanosuspensions as particulate drug formulations in
therapy: rationale for development and what we can expect for the future. Advance in Drug Delivery Review. 47;
2001a: 3–19.
25.
Müller RH, Krause K and Mäder K.
Method for controlled production of ultrafine microparticles and nanoparticles.
WO/2001/003670. 2001b.
26.
Müller RH and Jacobs C. Buparvaquone
mucoadhesive nanosuspension:
preparation, optimisation and long-term stability. International Journal of Pharmaceutics. 237; 2002: 151–161.
27.
Müller RH, Pardeike J and Hommoss A. Nanoparticles in
therapeutics: drug nanocrystals and lipid nanoparticles. NanoTrends, Potsdam,
Germany. 2006.
28.
Müller RH, Shegokar R and Singh
KK. Positively charged nanostructured lipid carriers (NLC) for drug delivery by adhesion. Ninth
International Symposium on ‘Advances in Technology and Business Potential of
New Drug Delivery Systems’ Controlled Release Society-Indian Local Chapter, Mumbai, India. 2009: 36.
29.
Eurand - Drug Development and Delivery Technologies
Platforms. Available from: URL: http://www.eurand.com.
30.
The OSD: Oral Synchronous Drug
Delivery System, Delivery Therapeutics Ltd (DT), Executive summary march 2003. Available from: URL:
http://www.bzbgroup.com.
31.
Hu J, Johnston KP and Williams RO
III; Spray freezing into liquid (SFL) particle engineering technology to enhance
dissolution of poorly water soluble drugs: organic solvent versus
organic/aqueous co-solvent systems. European Journal of
Pharmaceutical Sciences. 20; 2003: 295–303.
32.
Hu J, Rogers TL, Brown J, Young T, Johnston KP and
Williams RO. Improvement of dissolution rates of poorly
water soluble APIs using novel spray freezing into liquid technology. Pharmaceutical
Research. 19; 2002:1278–1284.
33.
Rogers TL, Nelsen AC, Sarkari M, Young T,
Johnston KP and Williams RO. Enhanced aqueous dissolution of a poorly water soluble drug by novel particle engineering
technology: spray-freezing into liquid with atmospheric freeze-drying. Pharmaceutical Research. 20; 2003:
485–493.
34.
Yu Z, Rogers TL, Hu J, Johnston KP and
Williams RO. Preparation and characterization of microparticles
containing peptide produced by a novel process: spray freezing into liquid. European
Journal of Pharmaceutics and Biopharm. 54; 2002:
221–228.
35. Purvis T, Mattucci
ME, Crisp MT, Johnston KP and Williams RO III. Rapidly Dissolving Repaglinide Powders Produced by the Ultra-Rapid Freezing
Process. American Association of Pharmaceutical Science Technology.
8(3); 2007: Article 58.
Received on 17.08.2011
Accepted
on 15.09.2011
©
A&V Publication all right reserved
Research Journal of
Pharmaceutical Dosage Forms and Technology. 3(5): Sept.-Oct. 2011, 183-192