throbber
European Journal of Pharmaceutical Sciences 18 (2003) 113–120
`
`www.elsevier.com / locate / ejps
`
`Review
`N anosizing: a formulation approach for poorly-water-soluble compounds
`*
`Elaine Merisko-Liversidge , Gary G. Liversidge, Eugene R. Cooper
`Elan Drug Delivery,3500 Horizon Dr., King of Prussia, PA 19406, USA
`
`Received 21 August 2002; received in revised form 15 November 2002; accepted 18 November 2002
`
`Abstract
`
`Poorly-water-soluble compounds are difficult to develop as drug products using conventional formulation techniques and are frequently
`abandoned early in discovery. The use of media milling technology to formulate poorly-water-soluble drugs as nanocrystalline particles
`
`offers the opportunity to address many of the deficiencies associated with this class of molecules. NanoCrystal Technology is an attrition
`process wherein large micron size drug crystals are media milled in a water-based stabilizer solution. The process generates physically
`stable dispersions consisting of nanometer-sized drug crystals. Nanocrystalline particles are a suitable delivery system for all commonly
`used routes of administration, i.e. oral, injectable (IV, SC, and IM) and topical applications. In addition, aqueous dispersions of
`nanoparticles can be post-processed into tablets, capsules, fast-melts and lyophilized for sterile product applications. The technology has
`been successfully incorporated into all phases of the drug development cycle from identification of new chemical entities to refurbishing
`marketed products for improving their performance and value.
` 2002 Elsevier Science B.V. All rights reserved.
`
`Keywords: Poorly-water-soluble compounds; Nanoparticles and drug delivery
`
`1 . Introduction
`
`It is estimated that 40% or more of active substances
`being identified through combinatorial screening programs
`are poorly soluble in water (Lipinski, 2001, 2002). When
`these molecules are formulated using conventional meth-
`ods, the performance of the drug in preclinical screens is
`oftentimes erratic and highly variable.
`In the clinic,
`conventional formulations of poorly-water-soluble drugs
`are frequently plagued with problems such as poor and
`highly variable bioavailability. The dosage form is often-
`times affected by the fed–fasted state of the patient and its
`onset of action is slower than anticipated. All of these
`issues lead to sub-optimal dosing and poor performance.
`Generally,
`it
`is more expeditious and cost effective to
`chemically re-design the molecule,
`than to move a
`blemished molecule through the development process
`(Lipinski et al., 1997; Lipper, 1999; Venkatesh and Lipper,
`2000; Veber et al., 2002).
`Currently, there are a limited number of formulation
`approaches available for compounds that are poorly solu-
`ble in water. The most direct approach for enhancing
`solubility is to generate a salt. If, however, the compound
`
`*Corresponding author. Tel.: 11-610-313-5130.
`E-mail address: elaine.liversidge@elan.com (E. Merisko-Liversidge).
`
`is non-ionizable, solubility concerns are generally ad-
`dressed by micronization and / or the development of oil-
`based solutions in gelatin capsules, i.e. soft-gel technology.
`In addition, co-solvents, surfactants or complexing agents
`such as cyclodextrins (Stella and Rajewski, 1997; Loftsson
`and Brewster, 1996; Akers, 2002) have been employed.
`Reasonable success has also been met
`in formulating
`water-insoluble drugs using emulsion (Nakano, 2000;
`Floyd, 1999), microemulsion (Lawrence and Rees, 2000)
`and solid dispersion technology (Leuner and Dressman,
`2000; Serajuddin, 1999; Breitenbach, 2002). Although
`some of these approaches have been successfully utilized,
`especially for highly potent compounds with low dose
`requirements, there is a growing need for more effective
`and versatile ways to handle formulation issues associated
`with poorly-water-soluble molecules. A broadly based
`technology applicable to this class of molecule could have
`a tremendous impact on discovery effectiveness and
`improve the performance of products suffering from
`formulation-related issues.
`A newer drug delivery approach for poorly-water-solu-
`ble compounds has come about in the last few years. In
`this approach, poorly-water-soluble compounds are formu-
`lated as nanometer-sized drug particles. There are various
`methodologies for generating drug nanoparticle formula-
`tions (Tom and Debenedetti, 1991; Pace et al., 1999;
`
`0928-0987 / 02 / $ – see front matter  2002 Elsevier Science B.V. All rights reserved.
`doi:10.1016 / S0928-0987(02)00251-8
`
`0001
`
`PSG2011
`Catalent Pharma Solutions v. Patheon Softgels
`IPR2018-00422
`
`

`

`114
`
`E. Merisko-Liversidge et al. / European Journal of Pharmaceutical Sciences 18 (2003) 113–120
`
`Muller et al., 2001; Rogers et al., 2001). This review,
`
`however, will focus on NanoCrystal Technology whereby
`poorly-water-soluble drugs are formulated as nanometer-
`sized drug crystals using high-shear media mills. Since this
`approach has been adapted to handle milligram quantities
`of drug substance, for the research scientist this technology
`provides an avenue to improve screening efforts without
`having to deal with solubility-related performance issues.
`For the pharmaceutical scientist, this approach provides a
`universal process suitable for formulation development and
`commercialization of various dosage forms. The first
`
`product to incorporate this technology is Rapamune , an
`immune suppressant agent, marketed by Wyeth Research
`Laboratories.
`
`2 . Nanocrystalline particles
`
`Nanocrystalline particles are nanometer-sized drug par-
`ticles of a poorly-water-soluble compound. Fig. 1 shows an
`electron micrograph and a diagrammatic representation of
`drug particles formulated as a colloidal dispersion. The
`electron-dense nanometer-sized particles exhibit a defined
`geometrical shape which is dictated by the morphology of
`the unprocessed crystalline powder, fracture plane of the
`crystals and drug / stabilizer interactions. The effects of
`shape on the properties and biological performance of the
`formulations have not been fully evaluated.
`Nanocrystalline dispersions consist of water, drug and
`stabilizer. If required, other excipients such as buffer, salts
`
`Fig. 1. Nanocrystalline drug particles. The transmission electron micro-
`
`graph of a NanoCrystal Colloidal dispersion magnified 35,0003. The
`insert provides a visual description of
`the crystalline nanoparticles
`generated using wet milling technology. The nanoparticles are typically
`less than 400 nm and are physically stabilized with a polymeric excipient.
`
`and sugars can be added to the dispersion. For most
`nanocrystalline formulations, drug concentration is 400
`mg / ml or less. The choice and concentration of stabilizer
`are selected to promote the particle size reduction process
`and generate physically stable formulations. To be effec-
`tive, the stabilizer must be capable of wetting the surface
`of the drug crystals and providing a steric or ionic barrier.
`In the absence of the appropriate stabilizer,
`the high
`surface energy of nanometer-sized particles would tend to
`agglomerate or aggregate the drug crystals. Physically
`stable nanocrystalline formulations are obtained when the
`weight ratio of drug to stabilizer is 20:1 to 2:1. Too little
`stabilizer induces agglomeration or aggregation and too
`much stabilizer promotes Ostwald ripening. The process of
`identifying an appropriate stabilizer(s) for a drug candidate
`is empirical and can be accomplished using milligram
`quantities of drug. Many commonly used pharmaceutical
`excipients such as the cellulosics, pluronics, polysorbates
`and povidones are acceptable stabilizers for generating
`physically stable nanoparticle dispersions (Liversidge et
`al., 1992; Kibbe, 2000; Schott, 1955). Oftentimes, stabili-
`zation is achieved using a combination on a non-ionic plus
`ionic stabilizer.
`Nanocrystalline dispersions are prepared using media
`milling processes (Liversidge et al., 1992). The milling
`chamber is charged with milling media, water, drug and
`stabilizer as depicted in Fig. 2. Drug concentration general-
`ly ranges from 1 to 400 mg / ml. High-energy-generated
`shear forces and / or the forces generated during impaction
`of the milling media with the drug provide the energy
`input to fracture drug crystals into nanometer-sized par-
`ticles. The process can be performed in either a batch or
`re-circulation mode. In batch mode, the time required to
`obtain dispersions with unimodal distribution profiles and
`mean diameters ,200 nm is 30–60 min. A comparison of
`the size of naproxen crystals before and after media
`milling is shown in Fig. 3. The media milling process
`readily fractures micron-sized drug crystals into a homoge-
`neous nanoparticle dispersion. The media milling process
`can successfully process micronized and non-micronized
`drug crystals. Once the formulation and process are
`optimized,
`there is very little batch-to-batch variation
`detected in the quality of the dispersion and processing
`time. A concern raised regarding the media milling pro-
`cesses for pharmaceutical applications is the quality and
`durability of the milling media used in manufacturing.
`
`Currently, the milling media used for the NanoCrystal
`Technology is a proprietary highly cross-linked poly-
`styrene resin. In addition, during process development and
`validation potential in-process impurities are monitored.
`Residual monomers are typically ,50 ppb and insolubles
`generated during processing are no more than 0.005%
`w / w based on the drug concentration of the dispersion or
`resulting solid dosage form.
`It is well documented that various processes can ad-
`versely induce polymorphic transitions (Byrn et al., 1995).
`
`0002
`
`

`

`E. Merisko-Liversidge et al. / European Journal of Pharmaceutical Sciences 18 (2003) 113–120
`
`115
`
`Fig. 2. The Media Milling Process is shown in a schematic representation. The milling chamber charged with polymeric media is the active component of
`the mill. The mill can be operated in a batch or re-circulation mode. A crude slurry consisting of drug, water and stabilizer is fed into the milling chamber
`and processed into a nanocrystalline dispersion. The typical residence time required to generate a nanometer-sized dispersion with a mean diameter ,200
`nm is 30–60 min.
`
`Fig. 3. The particle size distribution profile of naproxen crystals before (m) and after milling (d). Before milling, the drug crystals had a mean particle
`size of 24.2 microns. After being processed for 30 min in a media mill, the mean particle size of the nanocrystalline dispersion was 0.147 microns with
`D 5 0.205 microns. The particle size measurements were generated using laser light diffraction in a Horiba LA-910 using polystyrene nanospheres
`90
`ranging in size from 0.1 to 10 microns as standards.
`
`0003
`
`

`

`116
`
`E. Merisko-Liversidge et al. / European Journal of Pharmaceutical Sciences 18 (2003) 113–120
`
`For nanoparticles prepared by wet milling technology, this
`has not been an issue (Liversidge and Cundy, 1995). The
`milling process is performed under controlled temperature
`conditions and the aqueous phase effectively dissipates the
`heat generated during processing. In addition, basic testing
`methods performed before and after milling can be readily
`implemented to ensure the chemical
`integrity of
`the
`compound.
`Finally, a few comments should be made regarding the
`physical stability of the colloidal dispersions resulting from
`media milling processes. In the liquid state, the dispersions
`are very stable, especially if the solubility of the drug is
`less than 1 mg / ml and the drug, using wet-milling
`technology, can be formulated at concentrations greater
`than 100 mg / ml. For drug candidates that are slightly
`soluble, with time and under accelerated temperatures,
`evidence of Ostwald ripening may be observed (Schott,
`1955). However, dispersions of nanoparticles can be post-
`processed as a dry powder for solid dosage development or
`lyophilized for injectable products. These dried powders
`are designed to re-disperse into nanometer-sized particles
`when placed in water or an alternate water-based environ-
`ment. The ability of the dried powder to re-disperse into a
`non-aggregated / non-agglomerated nanoparticulate disper-
`sion is critical to the development of a solid dosage form
`that maintains the benefits of this enabling drug delivery
`technology.
`
`3 . Applications for oral delivery
`
`Dissolution kinetics is the primary driving force behind
`the improved pharmacokinetic properties of nanoparticle
`formulations of poorly-water-soluble compounds (Liver-
`sidge and Cundy, 1995). The dissolution rate of a drug is a
`function of its intrinsic solubility and particle size. For
`poorly-water-soluble drugs, surface area of the drug par-
`ticles drives dissolution. As described by the Nernst–
`Brunner and Levich modification of the Noyes–Whitney
`model of dissolution (Dressman et al., 1998; Horter and
`Dressman, 2001), surface area of the drug is directly
`proportional to its rate of dissolution:
`
`dX / dt 5 (A 3 D/d) 3 (C 2 X /V )
`
`where X is the amount of drug in solution, t is time, A is
`the effective surface area, D is the diffusion coefficient of
`the drug, d is the effective diffusion boundary layer, C is
`the saturation solubility of the drug, and V is the volume of
`the dissolution medium.
`The ability to formulate poorly-water-soluble molecules
`as nanometer-sized crystals can have a dramatic effect on
`bioavailability. If the drug particles are near spherical in
`shape, nanoparticles reduced in size from 10 microns to
`200 nm generate a 50-fold increase in surface area to
`volume ratio. This increase in surface area can have a
`
`major impact on drug absorption, provided the formulation
`retains or disperses into discrete particles after dosing. If
`bioavailability is truly dissolution rate limited, particle size
`reduction can significantly improve the performance of the
`drug (Liversidge and Cundy, 1995). The effects of particle
`size on the bioavailability of a poorly-water-soluble drug
`are demonstrated in Fig. 4 (Loper, 1999). In this study, the
`bioavailability of a poorly-water-soluble drug candidate
`was improved as the particle size of the preparation was
`reduced from 5 to 0.1 micron using wet media milling
`technology. This pattern of results is routinely observed,
`provided the primary factor effecting the bioavailability of
`the drug is rate and extent of dissolution. As currently
`practiced, formulating compounds as nanocrystalline dis-
`persions will not be of value when bioavailability is
`affected by metabolic- and / or permeation-related issues. In
`the future, it should be possible to combine this technology
`with agents that enhance permeation (Aungst, 2000) and /
`or minimize gut-related metabolic issues (Benet et al.,
`1999; Kusuhara et al., 1998). This combination of ap-
`proaches would lend itself to molecules referred to in the
`biopharmaceutics classification system as class four mole-
`cules. These molecules are poorly water soluble and have
`limited membrane permeability.
`For poorly-water-soluble molecules, there are benefits to
`be gained when dissolution is no longer a limiting factor.
`As shown in Fig. 5, a nanocrystalline formulation of
`danazol, whether administered as a liquid or a capsule,
`effectively reduced the variable performance observed with
`
`the marketed product, Danocrine . For poorly-water-solu-
`ble compounds, food plus the physiological environment
`induced by the fed state oftentimes is conducive for
`improving the bioavailability of the drug. In contrast, there
`
`Fig. 4. For poorly-water-soluble compounds, the particle size of the drug
`crystals can effect bioavailability. The graph shows the plasma con-
`centration time curve of a poorly-water-soluble discovery compound. The
`compound was administered orally at an identical dose to fasted dogs as
`nanoparticles or as cruder dispersions. The only variable was the mean
`particle size of each dispersion: 100 nm (o); 500 nm (d); 2 microns (j);
`and 5 microns (,).
`
`0004
`
`

`

`E. Merisko-Liversidge et al. / European Journal of Pharmaceutical Sciences 18 (2003) 113–120
`
`117
`
`fold increase in the AUCs during the first hour of the
`study. In addition to this need for improving the rate of
`absorption, for various therapeutic classes there is also the
`need for the medication to be effective over a prolonged
`period of time. For poorly-water-soluble drugs, this need is
`being met by using nanoparticles in combination with
`well-established controlled release, delayed release and
`pulsatile release technology. In addition, since the surface
`properties of the nanoparticles can be readily altered,
`particles can be generated with bioadhesive properties that
`provide an opportunity to impact the biological perform-
`ance of a drug (Lee et al., 2000). Finally, it should be
`stressed that, if properly formulated, the dispersions can be
`processed, using standard equipment, into tablets, capsules
`and fast melts. Nanocrystalline particles provide a phar-
`maceutics approach that is highly versatile while eliminat-
`ing or minimizing the development issues associated with
`this class of molecule.
`
`4 . Applications for injectable products
`
`Nanocrystalline formulations are a suitable dosage form
`for poorly-water-soluble injectable products (Merisko-
`Liversidge et al., 1996; Cooper, 2000; Bittner and Mount-
`field, 2002). The question that is oftentimes raised is why
`and when would it be advantageous to use nanoparticles as
`opposed to a more traditional formulation approach. One
`of the major advantages is that drug nanoparticles provide
`an opportunity for dosing at significantly higher levels than
`what can be accomplished using a more traditional ap-
`proach. Harsh solvents or co-solvents present in a tradi-
`tional formulation for this class of molecules are, often-
`times, dose limiting as a result of the toxicity of the
`excipients. Nanocrystalline formulations do not contain
`solvents and the stabilizers present in the formulation are
`chosen based on their safe use in injectable products. The
`ability to safely dose at higher levels provides an oppor-
`tunity for dose escalation that can impact performance. For
`instance, Fig. 6 compares the performance of paclitaxel, a
`poorly-water-soluble anticancer agent, formulated as the
`
`marketed product (Taxol ) using Cremophor EL / ethanol
`mixture to solubilize the drug and prepared as a nanoparti-
`
`Fig. 5. Nanocrystalline particles can reduce absorption variability re-
`sulting from the presence or absence of food. The data compare the
`performance of various dosage forms of danazol administered to vol-
`unteers in the fed and fasted state at a 200 mg dose. The variability
`observed in the commercial product was significantly lowered when
`danazol was formulated using nanoparticles and administered as a liquid
`dispersion or a dry-filled capsule.
`
`is often a loss in bioavailability in the fasted state, due to
`the absence of solubility enhancing agents present in food
`and bile. However, when poorly-water-soluble drugs are
`formulated as a uniform dispersion of nanometer-sized
`particles, variation in bioavailability, resulting from the fed
`vs. fasted state, can be minimized.
`Improving the dissolution rate of a poorly-water-soluble
`compound generally correlates with faster absorption rates
`(Liversidge and Conzentino, 1995). This can be a par-
`ticularly desirable feature for therapeutics, such as naprox-
`en and other poorly-water-soluble compounds, targeting
`pain control. For naproxen, it would be highly beneficial to
`have a dosage form with a fast onset of action. Table 1
`shows the results of a human pharmacokinetic study. The
`bioavailability of nanocrystalline naproxen is compared to
`
`that of the marketed products: Naprosyn (suspension) and
`
`Anaprox
`(tablet). Though many of the pharmacokinetic
`parameters remain unchanged, the time to reach maximal
`drug concentrations is approximately 50% less for the
`nanocrystalline dispersion, while maintaining a 2.5–4.5-
`
`Table 1
`Human pharmacokinetics of naproxen following oral administration of a nanocrystalline naproxen suspension, a Naprosyn
`tablets in the fed state. Dose: 500 mg / volunteer and N523
`
`
`
`suspension and Anaprox
`
`
`
`PK parameter
`
`T
`max
`AUC
`
`(h)
`
`(0 – 1 h)
`
`(mg h / l)
`
`(mg / l)
`(mg h / l)
`
`C
`max
`AUC
`(0 – a)
`(h)
`t
`
`1 / 2
`
`Formulation
`
`Nanocrystalline
`naproxen
`
`1.6961.05
`33.4969.12
`(27.3% CV)
`50.8966.03
`810.246166.858
`16.1864.42
`
`Naprosyn
`
`
`
`3.3361.32
`13.3868.1
`(61.1% CV)
`49.0966.44
`49.656188.12
`16.1662.81
`
`Anaprox
`
`
`
`3.2061.68
`7.4168.94
`(120.7% CV)
`53.4567.16
`863.976231.28
`14.8263.55
`
`0005
`
`

`

`118
`
`E. Merisko-Liversidge et al. / European Journal of Pharmaceutical Sciences 18 (2003) 113–120
`
`cles is determined by a multitude of factors, including the
`physical properties of the particles, the dose, infusion time
`and the dissolution of the drug particles in the hemo-
`dynamic pool of blood. For drug candidates that have
`solubility in the mg / ml range or are ionizable, following
`intravenous administration the nanoparticles will dissolve.
`In this situation, the biodistribution profile of the drug in a
`conventional formulation and a nanoparticle formulation
`will be equivalent. However, if the drug candidate has an
`intrinsic solubility in the mg / ml range, depending on the
`dose, the particles may not dissolve. Comparing the plasma
`profiles of paclitaxel and other poorly-water-soluble drugs
`formulated in solution, nanoparticles can generally be
`administered intravenously at higher doses (Table 2). In
`addition, the differences observed in the pharmacokinetic
`properties are in line with the differences that are antici-
`pated when comparing the distributional properties follow-
`ing IV injection of a drug in solution versus in a nanopar-
`ticulate platform. The decrease in clearance rate and
`limited volume distribution are currently explained by the
`sequestration of the nanoparticles primarily by the Kupffer
`cells of the liver. The interesting observation that has been
`repeatedly demonstrated is that entrapment of nanoparti-
`cles by the Kupffer cells does not adversely effect the
`safety profile of the drug. In actuality, for intravenous
`delivery, maximum tolerated dose can be increased by
`five- to 10-fold in comparison to solutions formulated with
`the aid of solvents, surfactants or cyclodextrins. In addi-
`tion, as shown in Table 3, sequestration by the phagocytic
`cells of the liver does not adversely effect efficacy. This
`can be explained by either of two working hypotheses. The
`first scenario is that the population of particles that avoids
`sequestration by the mononuclear phagocytic system
`(MPS) delivers a sufficiency of drug to the diseased site
`(Nouchi et al., 1998; Jain, 1996; Kong et al., 2000; Hobbs
`et al., 1998). Alternatively, once sequestered by the
`Kupffer cells, the cells act as a controlled release vehicle
`for the drug (Martin et al., 1982; Adachi et al., 1992; Soma
`
`Fig. 6. The efficacy of nanoparticle paclitaxel in the MV-522 human lung
`xenograft murine tumor model. The efficacy of the marketed product
`
`(Taxol ) and nanocrystalline paclitaxel was compared following in-
`travenous administration. Both formulations were administered at
`the
`maximum tolerated dose, i.e. the marketed product was administered at
`30 mg / kg (qd31). At this dose, a 22% death rate was observed for the
`marketed product. In comparison, nanocrystalline paclitaxel was well
`tolerated at 90 mg / kg (qd31). In this study, N59 for each experimental
`group. Data show the change in tumor weight for Controls (no treatment),
`Marketed Product (30 mg / kg) and Nanocrystalline Paclitaxel (90 mg /
`kg).
`
`the
`cle dispersion. The maximum tolerated dose of
`nanoparticle paclitaxel formulation is greater than that of
`the commercial product. This translates into an improved
`efficacy profile for the drug.
`In using a nanometer-sized particle formulation as an
`injectable, the particle nature of the formulation can effect
`its pharmacokinetic properties. Following intravenous in-
`jection, particles are sequestered by MPS-enriched organs,
`such as the liver and spleen (Illum et al., 1982; Juliano,
`1988; Toster et al., 1990; Stolnik et al., 1995; Neal et al.,
`1998; Liu et al., 2000; Moghimi et al., 2001). The
`biodistribution profile of intravenously injected nanoparti-
`
`Table 2
`Comparison of the maximum tolerated dose of IV administered anticancer agents formulated as nanocrystalline particles or using a marketed product, if
`available, or a more conventional formulation approach
`
`Compound and
`formulations tested
`
`Camptothecin
`(a) Nanocrystalline formulation
`(b) Solubilized (ethanol / Tween 80)
`
`Etoposide
`(a) Nanocrystalline formulation
`(b) Marketed product
`
`Paclitaxel
`(a) Nanocrystalline formulation
`
`(b) Marketed product
`
`Murine
`tumor
`model
`
`Panc03
`Panc03
`
`Panc03
`Panc03
`
`MV-522
`
`MV-522
`
`Injection
`schedule
`
`Bolus
`Bolus
`
`d3,5,7
`d3,5,7
`
`d35
`d31
`d35
`d31
`
`Total
`dose
`(mg / kg)
`
`48.6
`18.0
`
`69
`69
`
`100
`90
`100
`30
`
`No.
`toxic
`deaths
`
`0 / 4
`1 / 4
`
`0 / 5
`2 / 5
`
`0 / 9
`0 / 9
`2 / 9
`2 / 9
`
`0006
`
`

`

`E. Merisko-Liversidge et al. / European Journal of Pharmaceutical Sciences 18 (2003) 113–120
`
`119
`
`Table 3
`Comparison of the efficacy of nanocrystalline paclitaxel with the conventional formulation using the mammary 16C murine tumor model (N55 for each
`experimental group)
`
`Treatment
`
`No treatment
`Nanopaclitaxel
`
`
`Taxol conventional
`formulation,
`cremophor / ethanol
`
`Total
`a
`dose
`(mg / kg)
`
`d
`NA
`87.5
`44
`87.5
`44
`
`Median tumor
`burden on
`day 11 (mg)
`
`2550 (763–3924)
`0 (0–0)
`0 (0–64)
`76 (0–256)
`417 (196–661)
`
`b
`
`T / C
`(%)
`
`NA
`0
`0
`3
`16
`
`Log
`10
`tumor
`cell kill
`
`c
`
`NA
`5.9
`2.8
`2.2
`1.2
`
`Cures
`tumor free
`day 162
`
`0 / 5
`3 / 5
`0 / 5
`0 / 5
`0 / 5
`
`a Dose administered intravenously on a daily 35 dosing schedule.
`b T / C (%): tumor wt. in Treated animals / tumor wt. in untreated Controls3100.
`]
`]
`c Log
`cell kill5tumor growth delay / 3.323tumor volume doubling time.
`10
`d NA, not applicable.
`
`et al., 2000). Currently, both scenarios are being further
`explored. In addition, studies are in progress to identify
`how best to manipulate the surface properties, size and
`shape of the nanoparticles to eliminate, when desirable,
`uptake by the phagocytic cells of MPS-enriched organs.
`This is the approach that has been successfully im-
`plemented in the work that has produced the ‘‘stealth’’
`liposome (Woodle, 1998; Gregoriades, 1995; Allen, 1997;
`Papisov, 1998) and has been utilized to demonstrate the
`passive targeting capability of other types of microcarriers
`(Couvreur et al., 1990; Gref et al., 1994; Stolnik et al.,
`1995; Moghimi et al., 2001). Future studies will focus on
`the second generation of nanocrystalline particles to be
`laced with functionalized surface coatings that are capable
`of eliciting passive or active targeting.
`
`5 . Conclusions
`
`The use of wet milling technology to formulate poorly-
`water-soluble compounds is a viable approach capable of
`resolving many of
`the current
`issues associated with
`developing
`and
`commercializing
`poorly-water-soluble
`molecules. The successful implementation of nanocrystal
`technology to a chemical entity is primarily driven by
`solubility properties of the drug and hence can be readily
`applied to various classes of compounds. Nanocrystalline
`formulations can be dried and post-processed into capsules
`and tablets. However, care must be taken to identify a
`process and formulation that yields a re-dispersible sold
`dosage form. For
`injectables,
`the dispersions can be
`sterilized by terminal heat (Na et al., 1999), filtration
`(Zheng and Bosch, 1997) or gamma irradiation and stored
`as a liquid or lyophilized and re-dispersed at the time of
`injection. Though this review is focused on oral and
`injectable applications of nanocrystalline formulations, the
`approach is a very versatile drug delivery platform and is
`suitable for other commonly used routes of administration
`such as nasal and pulmonary delivery (Wiedmann et al.,
`1997). In the last 10 years, nanoparticle technology has
`
`evolved from a conception to a viable commercializable
`drug delivery platform whose versatility and applicability
`are just beginning to be realized. The percentage of poorly-
`water-soluble molecules being identified as actives is
`increasing. New approaches for bringing these molecules
`to the market place in a timely fashion are a necessity for
`the success of the pharmaceutical industry and for addres-
`sing the unmet medical needs these therapeutics may
`provide.
`
`R eferences
`
`Adachi, Y., Arii, S., Funaki, N., Higashitsuji, H., Fijita, S., Furutani, M.,
`Mise, M., Zhang, W., Tobe, T., 1992. Tumoricidal activity of Kupffer
`cells augmented by anticancer drugs. Life Sci. 51, 177–183.
`Akers, M.J., 2002. Excipient–drug interactions in parenteral formulations.
`J. Pharm. Sci. 91, 2283–2300.
`Allen, T.M., 1997. Liposomes: opportunities in drug delivery. Drugs 54,
`8–14.
`Aungst, B., 2000. Intestinal permeation enhancers. J. Pharm. Sci. 89,
`429–442.
`Benet, L.Z., Izumi, T., Zhang, Y., Silverman, J.A., Wacher, V.J., 1999.
`Intestinal MDR transport proteins and P-450 enzymes as barriers to
`oral drug delivery. J. Control. Release 62, 25–31.
`Bittner, B., Mountfield, R.J., 2002. Intravenous administration of poorly
`soluble new drug entities in early drug discovery: The potential impact
`of
`formulation on pharmacokinetic parameters. Cur. Opin. Drug
`Discovery and Dev., 59–71.
`Breitenbach, J., 2002. Melt extrusion: from process to drug delivery
`technology. EJPB 54, 107–117.
`Byrn, S., Pfeiffer, R., Ganey, M., Hoiberg, C., Poochikian, G., 1995.
`Pharmaceutical solids: a strategic approach to regulatory considera-
`tions. Pharm. Res. 12, 945–954.
`Cooper, E., 2000. Nanoparticles in drug delivery. In: NanoTechnology
`Symposium, NIH, September.
`Couvreur, P., Roblot-Treuple, L., Poupon, M.F., Brasseur, F., Puisieux,
`F., 1990. Nanoparticles as microcarriers for anticancer drugs. Adv.
`Drug Deliv. Rev. 5, 209–230.
`Dressman, J.B., Amidon, G.L., Reppas, C., Shah, V.P., 1998. Dissolution
`testing as a prognostic tool for oral drug adsorption: immediate release
`dosage forms. Pharm. Res. 15, 11–22.
`Floyd, A.G., 1999. Top ten considerations in the development of
`parenteral emulsions. Pharm. Sci. Tech. 4, 134–143.
`Gref, R., Minamitake, Y., Peracchia, M.T., Trubetskoy, V., Torchilin, V.,
`
`0007
`
`

`

`120
`
`E. Merisko-Liversidge et al. / European Journal of Pharmaceutical Sciences 18 (2003) 113–120
`
`Langer, R., 1994. Biodegradable long-circulating polymeric nanos-
`pheres. Science 263, 1600–1603.
`Gregoriades, G., 1995. Engineering liposomes for drug delivery: progress
`and problems. TIBTECH 13, 527–537.
`Hobbs, S.K., Monsdky, W.L., Yuan, F., Roberts, G.W., Griffith, L.,
`Torchilin, V., Jain, R.K., 1998. Regulation of transport pathways in
`tumor vessels: role of tumor type and microenvironment. Proc. Natl.
`Acad. Sci. 95, 4607–4612.
`Horter, D., Dressman, J.B., 2001. Influence of physicochemical properties
`on dissolution of drugs in the gastrointestinal tract. Adv. Drug Deliv.
`Rev. 46, 75–87.
`Illum, L., Davis, S.S., Wilson, C.G., Thomas, N.W., Frier, M., Hardy,
`J.G., 1982. Blood clearance and organ disposition of intravenously
`administered colloidal particles. Effect of particle size, nature, and
`shape. Int. J. Pharm. 2, 135–136.
`Jain, R.K., 1996. Delivery of molecular medicine to solid tumors. Science
`271, 1079–1080.
`Juliano, R.L., 1988. Factors affecting the clearance kinetics and tissue
`distribution of liposomes, microspheres and emulsions. Adv. Drug
`Deliv. Rev. 2, 31–54.
`Kibbe, A.H., 2000. Handbook of Pharmaceutical Excipients, 3rd Edition.
`AphA and PhP, Washington, DC.
`Kong, G., Braun, R.D., Dewhirst, M.W., 2000. Hypothermia enables
`tumor-specific nanoparticle delivery: effect of particle size. Cancer
`Res. 60, 4440–4445.
`Kusuhara, H., Suzuki, H., Sugiyama, Y., 1998. The role of p-glycoprotein
`and canalicular multispecific organic anion transporter in the hepato-
`biliary excretion of drugs. J. Pharm. Sci. 87, 1025–1040.
`Lawrence, M.J., Rees, G.D., 2000. Microemulsion-based media as novel
`drug delivery systems. Adv. Drug Deliv. Rev. 45, 89–121.
`Lee, J.W., Park, J.H., Robinson, J.R., 2000. Bioadhesive-based dosage
`forms: the next generation. J. Pharm. Sci. 89, 850–866.
`Leuner, C., Dressman, J., 2000. Improving drug solubility for oral
`delivery using solid dispersions. Eur. J. Pharm. Biopharm. 50, 47–60.
`Lipinski, C.A., Lombardo, F., Dominy, B.W., Feeney, P.J., 1997. Ex-
`perimental and computational approaches to estimate solubility and
`permeability in drug discovery and development setting. Adv. Drug
`Deliv. Rev. 23, 3–25.
`Lipinski, C.A., 2001. Avoiding investment in doomed drugs, is poor
`solubility an industry wide problem? Curr. Drug Dis., 17–19.
`Lipinski, C., 2002. Poor aqueous solubility—an industry wide problem in
`drug discovery. Am. Pharm. Rev. 5, 82–85.
`Lipper, R.A., 1999. E pluribus product. Mod. Drug Dis. 2, 55–60.
`Liversidge, G.G., Cundy, K.C., Bishop, J.F., Czekai, D.A., 1992. Sur

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket