throbber
Advanced Drug Delivery Reviews 59 (2007) 631 – 644
`
`www.elsevier.com/locate/addr
`
`Nanosizing — Oral formulation development and
`biopharmaceutical evaluation☆
`Filippos Kesisoglou, Santipharp Panmai, Yunhui Wu ⁎
`
`Department of Pharmaceutical Research, Merck & Co., Inc., West Point, PA, USA
`
`Received 21 March 2007; accepted 10 May 2007
`Available online 25 May 2007
`
`Abstract
`
`Poor aqueous solubility represents a major hurdle in achieving adequate oral bioavailability for a large percentage of drug compounds in drug
`development nowadays. Nanosizing refers to the reduction of the active pharmaceutical ingredient (API) particle size down to the sub-micron
`range, with the final particle size typically being 100–200 nm. The reduction of particle size leads to a significant increase in the dissolution rate of
`the API, which in turn can lead to substantial increases in bioavailability. This review describes the principles behind nanosizing, the production
`and characterization of nanoformulations as well as the current experience with utilization of such formulations in vivo.
`© 2007 Elsevier B.V. All rights reserved.
`
`Keywords: Nanosizing; Nanoparticle; Nanosuspension; Bioavailability enhancement; Dissolution; Formulation
`
`Contents
`
`1.
`2.
`3.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Introduction .
`Increasing dissolution rate through nanosization — theoretical aspects .
`.
`.
`.
`.
`.
`.
`Formulation development of nanoformulations
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.1.
`Selection of excipients
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.2.
`Characterization of nanoformulations .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.3.
`Process development at lab and commercial scales
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.3.1.
`Feasibility of nanosuspension .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.3.2. Nanosuspension for toxicology study .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3.3.3. Nanoformulation for clinical applications .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4. Biopharmaceutical evaluations of nanoformulations .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Computational approach — predicting performance of nanoformulations
`4.1.
`4.2. Nanoformulations for toxicology studies .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4.3. Nanoformulations for preclinical and clinical applications .
`.
`.
`.
`.
`.
`.
`4.3.1.
`In vitro studies
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4.3.2.
`Preclinical studies .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4.3.3.
`Clinical applications .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`5. Nanosuspensions for non-oral applications
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`632
`632
`633
`633
`633
`634
`634
`635
`636
`637
`637
`637
`639
`639
`639
`641
`641
`
`☆ This review is part of the Advanced Drug Delivery Reviews theme issue on “Drug solubility: How to measure it, how to improve it”.
`⁎ Corresponding author. Merck & Co., Inc., WP75B-310, 770 Sumneytown Pike, PO Box 4, West Point, PA 19486-0004, USA. Tel.: +1 215 652 6911; fax: +1 215
`993 2265.
`E-mail addresses: filippos_kesisoglou@merck.com (F. Kesisoglou), santipharp_panmai@merck.com (S. Panmai), yunhui_wu@merck.com (Y. Wu).
`
`0169-409X/$ - see front matter © 2007 Elsevier B.V. All rights reserved.
`doi:10.1016/j.addr.2007.05.003
`
`LUPIN EX. 1015
`Lupin v. iCeutica
`US Patent No. 8,999,387
`
`Page 1
`
`

`
`632
`
`F. Kesisoglou et al. / Advanced Drug Delivery Reviews 59 (2007) 631–644
`
`Future directions
`6.
`7. Conclusions .
`.
`References .
`.
`.
`.
`.
`
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`.
`.
`.
`
`. 642
`. 642
`. 643
`
`nanosized formulations through the different stages of drug
`development covering both formulation aspects such as excipient
`selection and assessment of bioperformance. Examples of
`utilization of nanosizing to increase oral absorption are provided
`and, when applicable, compared to other novel oral dosage forms.
`Finally, we discuss the advantages and limitations of nanosizing in
`terms of its applicability in the drug development process.
`
`2. Increasing dissolution rate through nanosization —
`theoretical aspects
`
`1. Introduction
`
`Advances in combinatorial chemistry, biology and genetics in
`the recent years have led to a steady increase in the number of
`drug candidates under development. Due to the phospholipidic
`nature of cell membranes, a certain degree of lipophilicity is
`oftentimes a requirement for the drug compound, not only to be
`absorbed through the intestinal wall following oral administra-
`tion but possibly also to exert its pharmacological action in the
`target tissue. While high lipophilicity is advantageous in terms of
`compound permeability,
`it
`intrinsically translates into poor
`aqueous solubility. Since the first step in the oral absorption
`process is dissolution of the drug compound in the gastrointes-
`tinal lumen contents, poor aqueous solubility is rapidly becom-
`ing the leading hurdle for formulation scientists working on oral
`delivery of drug compounds [1].
`Nanosizing refers to the reduction of the active pharmaceutical
`ingredient (API) particle size down to the sub-micron range.
`While reduction of particle size has been employed in pharma-
`ceutical industry for several decades, recent advances in milling
`technology and our understanding of such colloidal systems have
`enabled the production of API particles of 100–200 nm size in a
`reproducible manner. The sub-micron particles are stabilized with
`surfactants or polymers in nanosuspensions which can be further
`processed into standard dosage forms, such as capsules or tablets,
`suitable for oral administration. These nanoformulations offer
`increased dissolution rates for drug compounds and complement
`other technologies used to enhance bioavailability of insoluble
`compounds (BCS Class II and IV) such as solubility enhancers
`(i.e. surfactants), liquid-filled capsules or solid dispersions of
`drugs in their amorphous state.
`The advantages of nanoformulations in oral drug delivery have
`been demonstrated in vitro in dissolution testing and in vivo in both
`preclinical studies as well as clinical trials. Nanocrystalline API has
`been shown to dramatically increase the rate of dissolution in vitro,
`improve bioavailability, reduce variability and alleviate positive
`food effects for orally administered drug molecules. As seen in
`Table 1, there are currently five pharmaceutical products that utilize
`nanocrystalline API to achieve their drug delivery goals. The goal
`of this review is to cover the theoretical background and practical
`aspects behind utilization of nanosizing as a means to improve oral
`bioavailability of drug compounds. We discuss development of
`
`Table 1
`Current marketed pharmaceutical products utilizing nanocrystalline API
`
`The solid API dissolution rate is proportional to the surface
`area available for dissolution as described by the Nernst–
`Brunner/Noyes–Whitney equation [2–4]:
`
`
`¼ A D
`Cs− Xd
`V
`
`dX
`dt
`
`h
`
`ð1Þ
`
`where dX/dt = dissolution rate, Xd = amount dissolved, A = par-
`ticle surface area, D = diffusion coefficient, V = volume of fluid
`available for dissolution, Cs = saturation solubility, h = effective
`boundary layer thickness.
`Based on this principle, API micronization has been
`extensively used in the pharmaceutical industry to improve
`oral bioavailability of drug compounds. It is evident that a
`further decrease of the particle size down to the sub-micron
`range will further increase dissolution rate due to the increase of
`the effective particle surface area [5]. For example in the case of
`aprepitant, the nanocrystal dispersion of 120-nm particle size
`exhibits a 41.5-fold increase in surface area over the standard
`5 μm suspension [6]. Furthermore, as described by the Prandtl
`equation, the diffusion layer thickness (h) will also be decreased
`thus resulting in an even faster dissolution rate [7].
`In addition to the dissolution rate enhancement described above,
`an increase in the saturation solubility of the nanosized API is also
`expected [8], as described by the Freundlich–Ostwald equation:
`
`
`ð2Þ
`S ¼ Sl exp
`
`2gM
`rqRT
`
`where S=saturation solubility of the nanosized API, S∞=satu-
`ration solubility of an infinitely large API crystal, γ is the crystal-
`
`Product
`
`Drug compound
`
`Indication
`
`Company
`
`Nanoparticle technology
`
`RAPAMUNE®
`EMEND®
`TriCor®
`MEGACE® ES
`Triglide™
`
`Sirolimus
`Aprepitant
`Fenofibrate
`Megestrol acetate
`Fenofibrate
`
`Immunosuppressant
`Antiemetic
`Treatment of hypercholesterolemia
`Appetite stimulant
`Treatment of hypercholesterolemia
`
`Wyeth
`Merck
`Abbott
`PAR Pharmaceutical
`First Horizon Pharmaceutical
`
`Elan Drug Delivery Nanocrystals®
`Elan Drug Delivery Nanocrystals®
`Elan Drug Delivery Nanocrystals®
`Elan Drug Delivery Nanocrystals®
`SkyePharma IDD®-P technology
`
`Page 2
`
`

`
`F. Kesisoglou et al. / Advanced Drug Delivery Reviews 59 (2007) 631–644
`
`633
`
`medium interfacial tension, M is the compound molecular weight,
`r is the particle radius, ρ is the density, R is a gas constant and T is
`the temperature.
`Assuming a molecular weight of 500, ρ = 1 g/mL and a γ
`value of 15–20 mN m- 1 for the crystal-intestinal fluid inter-
`facial tension, the above equation would predict an approxi-
`mately 10–15% increase in solubility at a particle size of
`100 nm. However a more significant increase in solubility ap-
`pears to occur in reality e.g. Muller and Peters reported an
`increase of 50% in the solubility of an insoluble antimicrobial
`compound when the particle size was reduced from 2.4 μm to
`800 or 300 nm [8]. This increase in solubility leads to a further
`increase in dissolution rate and, as a result, nanosuspensions
`often achieve significantly higher exposure levels compared to
`suspensions of micronized API, even when the same surfactants
`are used. Finally, the increase in surface wetting by the sur-
`factants in the nanosuspension formulations most likely results
`in a further enhancement of the dissolution rates compared to
`micronized suspensions.
`
`3. Formulation development of nanoformulations
`
`Compared with formulation efforts using traditional processes
`such as wet-granulation (WG), roller-compaction (RC), or direct
`compression (DC), development of nanoformulations is one of
`the more complex formulation works. Not only must the drug
`particles be rendered into nanosized domains via technically
`demanding processes, but
`they must also be stabilized and
`formulated rigorously to retain the nature and properties of the
`nanoparticles. This review will focus on Elan's nanomilling
`technology for oral formulation applications. Before delving in, a
`snapshot of other nanoparticle technologies is provided.
`For the purposes of this discussion,
`the definition of
`“nanoparticles” will be confined to crystalline particles with a
`monolithic core. There are two main approaches to making
`nanoparticles: ‘top down’ and ‘bottom up’ technologies [9,10].
`The ‘top down’ approach is by far the more popular; it will be
`referred to as ‘nanosizing’. The approach basically relies on
`mechanical attrition to render large crystalline particles into
`nanoparticles. Examples of the ‘top down’ approach include
`Elan's NanoCrystal® wet-milling technology [11] and Sky-
`ePharma's Dissocubes® high-pressure homogenization technol-
`ogy [9,12]. The ‘bottom up’ approach relies on controlled
`precipitation/crystallization [10]. The process involves dissolving
`the drug in a solvent and precipitating it in a controlled manner to
`nanoparticles through addition of an anti-solvent (usually, water).
`This technology is available from DowPharma (Midland, MI,
`USA) and BASF Pharma Solutions (Florham Park, NJ, USA). A
`hybrid approach is also feasible. Baxter's NANOEDGE®
`technology employs both ‘bottom up’ and ‘top down’ approaches
`through microprecipitation and homogenization [9].
`The focus and examples of this review will be based on the
`application of the NanoCrystal® technology to the development
`of nanoformulations. However, most of the discussion on
`properties and characterization of nanoparticles, selection of
`stabilizers, and considerations in nanoformulation development
`is relevant to the other technologies.
`
`3.1. Selection of excipients
`
`Formulation of nanosuspension requires a careful selection of
`stabilizers. Stabilizers are needed to stabilize the nanoparticles
`against inter-particle forces and prevent them from aggregating. At
`the nanometer domain, attractive forces between particles, due to
`dispersion or van der Waals forces, come into play [13]. This
`attractive force increases dramatically as the particles approach
`each other, ultimately resulting in an irreversible aggregation. To
`overcome the attractive interaction, repulsive forces are needed.
`There are two modes of imparting repulsive forces or energetic
`barriers to a colloidal system — steric stabilization and electrostatic
`stabilization. Steric stabilization is achieved by adsorbing polymers
`onto the particle surface. As the particles approach each other, the
`osmotic stress created by the encroaching steric layers acts to keep
`the particles separate. Electrostatic stabilization is obtained by
`adsorbing charged molecules, which can be ionic surfactants or
`charged polymers, onto the particle surface. Charge repulsion
`provides an electrostatic potential barrier to particle aggregation.
`Typically, the use of steric stabilization alone is sufficient to
`stabilize the nanoparticles and prevent irreversible aggregation.
`However, enough attractive force between particles may still
`remain to cause a loose and reversible flocculation. To circumvent
`flocculation, steric stabilization is often combined with electrostatic
`stabilization for additional repulsive contribution.
`Common pharmaceutical excipients that are suitable for use as
`polymeric stabilizers include the cellulosics, such as hydroxy-
`propylcellulose (HPC) and hydroxypropylmethylcellulose
`(HPMC), povidone (PVP K30), and pluronics (F68 and F127)
`[11,14–16]. The molecular weights of these polymers are usually
`between 50 k and 100 kDa. The chains should be long enough to
`provide a steric layer, but not too big to slow down dissolution.
`The surfactant stabilizers can be non-ionic, such as polysorbate
`(Tween 80), or anionic, such as sodium laurylsulfate (SLS) and
`docusate sodium (DOSS). Cationic surfactants are typically not
`used as stabilizers for oral formulation due to their antiseptic
`properties. Smaller surfactant molecules can also stabilize
`nanoparticles, but are usually more prone to Ostwald ripening
`and particle growth. Several groups have reported the use of the
`above stabilizers in their work [11]. Also, surfactants often help in
`the wetting and dispersion of the drug particles which are usually
`very hydrophobic. In marketed products based on Elan's
`NanoCrystal® technology, stabilizers such as HPMC E3,
`Povidone, HPC-SL, DOSS, and SLS have been used.
`Nanosuspensions are typically converted to a solid dosage
`form for clinical formulations. Prior to drying, redispersants need
`to be added to the nanosuspension to ensure complete redisper-
`sion of nanoparticles into their primary, pre-drying state [17].
`Sugars, such as sucrose, lactose, and mannitol, are commonly
`used as redispersants in oral formulations. The sugar molecules
`serve as “protectants” and prevent nanoparticles from aggregating
`as they are concentrated during drying [17].
`
`3.2. Characterization of nanoformulations
`
`A broad range of characterization tools and techniques exists for
`nanosuspensions. Muller et al. [12] have provided a comprehensive
`
`Page 3
`
`

`
`634
`
`F. Kesisoglou et al. / Advanced Drug Delivery Reviews 59 (2007) 631–644
`
`coverage of these characterization tools. Techniques for charac-
`terizing nanoparticles can be generalized into two sub-groups.
`The first group deals with attributes and properties of single
`nanoparticles, such as their particle size and surface charge (zeta
`potential). Particle crystallinity, dissolution, and surface coverage
`also fall in this category. The second group measures bulk
`properties, such as the viscosity. Redispersibility testing is
`additionally used to evaluate the redispersion of solid nanofor-
`mulations in relevant media, such as water and gastric fluid.
`The most basic property of a nanoparticle is its size. Various
`methods are available for particle size measurement [18]. A
`popular technique is laser light scattering, which allows quick
`determination of the particle size and distribution. Many models
`are available from Horiba [Irvine, CA, USA], Malvern [Worcester-
`shire, UK], and Microtrac [Montgomeryville, PA, USA] etc. One
`model that has been frequently used is the Horiba LA-910, which
`can measure from 50 nm to 1000 μm. Typically, only a few drops
`of nanosuspensions are required for a measurement (note that
`dilution into an aqueous medium is necessary). Useful information
`includes the mean values, along with the D10, D50, and D90 (D90
`means that 90% of the particles, by volume, are below the given
`size). The particle size distribution of milled nanosuspensions is
`typically narrow, with a coefficient of variation (CV) of about 25 to
`40%. For comparison, the CVof latex standard particles, which are
`relatively monodisperse, is generally 5 to 10% whereas the CV of
`un-milled APIs, which are more polydisperse, is typically N100%.
`Another fundamental property of nanoparticles is their surface
`charge. Surface charges can arise from (i) ionization of the particle
`surface or (ii) adsorption of ions (such as surfactants) onto the
`surface. Typically, the surface charge is assessed through measure-
`ments of the zeta potential. Zeta potential is the potential at the
`hydrodynamic shear plane and can be determined from the particle
`mobility under an applied electric field [13]. The mobility will
`depend on the effective charge on the surface. Zeta potential is also
`a function of electrolyte concentration. Examples of dilution media
`− 4 M. Various models are
`are aqueous KCl solutions, e.g., 10
`available from Brookhaven [Worcestershire, UK], Horiba [Irvine,
`CA, USA], Malvern [Worcestershire, UK], Matec [Northborough,
`MA, USA] etc. The addition of anionic surfactants typically leads
`to a more negative zeta potential value. Zeta potential values in the
`−15 mV to −30 mVare common for well-stabilized nanoparticles.
`Viscosity is one of the more prominent bulk properties. For a
`nanosuspension, whose viscosity can vary dramatically, depend-
`ing on the extent of flocculation, it is helpful to determine the
`viscosity as a function of shear rate. Either a controlled-stress or
`a controlled-strain rheometer can be used (note that the yield
`stress can only be determined with the former design). Several
`models are available from TA Instruments [New Castle, DE,
`USA], Malvern [Worcestershire, UK], and Brookfield [Mid-
`dleboro, MA b USA]. Typically, measurements can be made
`using the cone-and-plate geometry. The working shear rate range
`is from 0.01 to 1000 s- 1. The viscosity ranges from 1 cP for water
`or dilute nanosuspensions to 1000 cP or greater for concentrated
`nanosuspensions. Newtonian behavior (constant viscosity
`across the usual range of shear rates) is typical of well-stabilized
`nanosuspensions while shear-thinning (decreasing viscosity
`with increasing shear rate) is inherent to flocculating systems.
`
`3.3. Process development at lab and commercial scales
`
`3.3.1. Feasibility of nanosuspension
`At the early stage in development, the API is usually in tight
`supply, e.g., even an amount of 100 mg may be hard to come by.
`Therefore, it is crucial that
`the feasibility of a nanomilled
`suspension can be assessed with as little API as possible.
`Typically, the feasibility work can be carried out at the small
`scale using 100 mg or less of API. The Nanomill® System [Elan
`Drug Discovery, King of Prussia, PA] can be employed. The
`working capacity of the smallest chamber is 10 mL, and a very
`small suspension volume can be evaluated. The nanomilling
`process involves the high shearing of drug suspensions in the
`presence of grinding media, as described by Merisko-Liversidge
`et al. [11]. The milling media is a highly cross-linked polystyrene
`resin (500-μm beads). Selected stabilizers can be screened, such
`as the cellulosics and pluronics. The batch milling time is
`normally within a few hours at a mill speed of 5000 rpm. The
`particle size of the milled suspension can be checked at the
`completion of milling. Drug suspensions with terminal mean
`particle size in the 100- to 250-nm range are generally deemed
`feasible and can be considered for preclinical pharmacokinetic
`evaluation (the ‘success rate’ in reaching the described mean
`particle size range, based on oral bioavailability enhancement is
`around 80% to 90% in our experience). The milled material can
`be recovered from the suspension + media mixture. At
`this
`juncture, only short-term physical stability (e.g. 24 to 48 h)
`needs to be demonstrated, mainly to cover the duration of the
`animal study. If oral bioavailability enhancement is achieved and
`further development is warranted, then additional formulation
`development and optimization work can be conducted.
`While particle size and morphology of the starting API are of
`less concern if nanosizing is to be employed in formulation
`development, the chemical form of the API needs be considered
`prior to laboratory testing. Typically, the neutral form is the
`preferred starting form. While there has been an example of a
`salt form-containing nanoformulation, such as Par Pharmaceu-
`tical's MEGACE® ES with its acetate salt, pharmaceutical salts
`are generally not preferred. Possible liabilities of the salt form
`are (i) risk of disproportionation, e.g., an HCl salt disproportio-
`nating to the free base form during nanomilling, (ii) risk of
`aggregation due to charge-based interactions in the small
`intestine, such as those with bile salts, and (iii)
`rapid
`solubilization and turnover of nanoparticles of a salt form into
`larger particles of the neutral form due to the pH changes in the
`gastrointestinal (GI) tract. Furthermore, the API's solubility
`should be low to minimize the potential for Ostwald ripening,
`and the most stable form in water should be used. APIs with
`ionizable groups and pKa between 2 and 7 (e.g. physiological
`pH range) run the risk of charge-based interactions even if not
`presented as a salt. The typical starting particle size of the API is
`between a few microns and a hundred microns. Larger starting
`materials are acceptable at the feasibility stage, but run the risk
`of clogging the nanomill at the larger processing scale (in which
`case, an API pre-milling step is usually employed).
`Another consideration is the possibility of shear-induced API
`form conversion or amorphous drug formation. Milling speed is
`
`Page 4
`
`

`
`F. Kesisoglou et al. / Advanced Drug Delivery Reviews 59 (2007) 631–644
`
`635
`
`often the cause of formation of amorphous form of the API,
`which can lead to enhanced solubility and Ostwald ripening.
`Oftentimes, poor physical stability can be attributed to
`amorphous form formation. One remedy would be to mill at a
`lower speed. Generation of heat by the milling process can also
`result in form conversion. The mill is typically jacketed to
`minimize the temperature rise. The crystallinity (and form) of
`the milled API can be checked by XRPD. This is accomplished
`by spinning down the nanoparticles via ultracentrifugation and
`performing a measurement on the moist sediment. Crystalline
`peaks of the API should be identifiable on top of the broad
`amorphous band of the polymeric stabilizer.
`
`3.3.2. Nanosuspension for toxicology study
`The next step in formulation development is geared toward
`supporting toxicology studies. An early-phase toxicology study
`typically spans 2 weeks to 3 months. The API requirement could
`range from a few hundred grams to a few kilograms. It is usually
`acceptable to formulate as a liquid nanosuspension at this stage
`[as opposed to a solid nanoformulation, which is preferred in a
`clinical setting (see Section 3.3.3)]. In the following sub-
`sections, various aspects in formulating and manufacturing
`nanosuspensions for toxicology studies are discussed: from
`composition and process to storage and manufacturing logistics.
`
`3.3.2.1. Stabilizer selection. A nanosuspension formulation
`for toxicology studies needs to be stable as well as be
`processable at the target drug concentration. Hence, formulation
`screening work should continue until a sufficiently stable
`nanosuspension can be identified. The work can be conducted at
`the small scale, for example, with the Nanomill® system and at
`low drug concentrations. A reasonable stability protocol would
`be to mill various nanosuspensions and monitor the particle size
`of these suspensions at 5 °C and ambient, starting from a day to
`a few weeks. A two- to three-week time is adequate to identify a
`sufficiently stable nanosuspension. Typically, the search should
`produce a few feasible polymeric stabilizers, such as HPMC or
`HPC. A suitable working polymer:drug ratio is from 0.05:1 to
`0.5:1. After a stable composition has been identified, the next
`step is to scale-up the drug loading to the target concentration.
`Normally, the target will be at least 100 mg/mL to meet the
`needs for toxicology studies. At these higher concentrations, the
`main issue is usually flocculation. Physical stability of more
`concentrated suspensions generally falls in line with those of
`dilute suspensions. The major consequences of flocculation are
`two-fold. The first is the larger effective particle size with
`reduced surface area for dissolution. The second is the viscosity
`increase (but this is generally minor and not problematic). In
`many cases, flocculation can be minimized by raising the level
`of anionic surfactant, such as SLS or DOSS, which helps
`improve wetting and electrostatic stabilization. Care should be
`taken not to add excessive surfactant as this can result in
`enhanced solubility and Ostwald ripening. To characterize a
`nanosuspension formulation, only a small quantity is needed, on
`the order of tens of grams of API. This low API requirement is
`advantageous, given the limited supply of API available at this
`point in the development process.
`
`One concept that could prove useful in the selection of
`polymeric stabilizer is that of surface coverage. In principle, to
`fully provide steric stabilization, the polymeric stabilizers must
`fully adsorb onto the surfaces of the nanoparticles. While
`nanosuspensions can and have been formulated successfully,
`little attention is paid to the whereabouts of the stabilizer.
`Knowledge of the adsorption isotherm may help provide
`additional insights into the formulation efforts. Panmai and
`Deshpande [19] described a convenient method for determining
`the adsorption isotherm of a nanosuspension, which involves
`the determination of the fractions of the stabilizer that are bound
`to the drug surface and unbound in solution for a given polymer
`concentration. A drug example was given using HPC-SL and
`PVP K29/32 as stabilizers (mean particle size = 100 nm). First, a
`series of nanosuspensions were prepared for different stabilizers
`and at different amounts (ranging from 0.05:1 to 0.5:1 stabilizer:
`drug). Then, the nanosuspensions were ultracentrifuged to settle
`the nanoparticles,
`leaving a clear supernatant, which was
`assayed for the concentration of the unbound polymer. Through
`mass balance, the fractions of bound and unbound polymers
`were calculated. The resulting adsorption isotherm clearly
`showed a monotonic adsorption and surface saturation for HPC-
`SL. On the other hand, there was a virtual lack of surface
`adsorption for PVP K29/32. The greater affinity of HPC-SL is
`likely due to its greater hydrophobicity than that of PVP.
`Furthermore, the minimum ratio of HPC-SL to drug to ensure
`surface coverage is around 0.12 to 1. This result is very much in
`line with the common working ratios of 0.1:1 to 0.2:1 for
`stabilizer:drug. The value is expected to change with the
`stabilizer and the particle size. Hence, this approach may be
`used to select a polymeric stabilizer on a more rational basis.
`
`3.3.2.2. Milling process. A conventional media mill would be
`needed to process the amount of API and suspension volume
`required for toxicology studies. Assuming a drug concentration
`of 100 to 200 mg/mL for a typical toxicology study (in dogs or
`rats) the required suspension volume is often greater than 5 L.
`One example of a suitable media mill is the Dynomill [Glen
`Mills, Inc., Clifton, NJ], with chamber sizes of 300 mL and
`600 mL. Inside the milling chamber is a shaft with a series of
`impellers, which provide the high-shear agitation (up to
`4000 rpm). Larger mills, such as the Netzsch mills [Netzsch
`Inc., Exton, PA], which come in 2-L, 10-L, and 60-L chamber
`sizes, also exist to handle even larger volume requirements. To
`supply a large volume, the mill is configured in the recirculation
`mode. For example, a 600-mL chamber, charged with milling
`media and suspension, can be connected to a vessel of 5 to 10 L.
`The suspension then flows into the milling chamber, undergoes
`intense media grinding, and exits the mill through a small gap.
`The milling media are strained by the gap and retained within
`the mill. The mill and the vessel are jacketed to control the
`temperature. The inlet suspension is around 5 to 10 °C while the
`outlet product can be anywhere from 15 to 30 °C, or even
`higher, depending on the mill speed and product viscosity. The
`overall milling time scales according to the residence time in the
`chamber. The required supply can typically be prepared within a
`12- to 48-h total run time. Once milling is complete, the product
`
`Page 5
`
`

`
`636
`
`F. Kesisoglou et al. / Advanced Drug Delivery Reviews 59 (2007) 631–644
`
`is harvested. The process yield is generally very high, being
`greater than 95% yield.
`It is perhaps worthwhile noting some of the key variables in
`media milling. Milling speed is one of the most important
`operating parameters. Typically,
`the milling tip speed falls
`between 5 and 15 m/s [20]; the actual impeller or shaft speed (in
`the thousands of rpms) of a particular mill will vary with its
`design and size. The media loading is also important and is
`generally around 80% to 90% of the maximum fill volume,
`which permits milling to the 100- to 200-nm range. As for the
`suspension properties, the milling time generally decreases with
`increasing suspension viscosity, due to higher shear forces and
`energy input, and with increasing drug loading, due to higher
`particle densities in the grinding zone [20].
`
`3.3.2.3. Manufacturing logistics. The use of nanosuspensions
`in toxicology studies can be complicated by the manufacturing
`logistics involved, which if not well managed, may result in a
`delay in the development program. Since the duration of the
`study can be quite long, e.g., up to 3 months, API and
`suspension volume requirements for the studies are large. Also,
`the doses are normally high, from 100 to 1000 mg/kg. These
`two factors, not to mention the non-routine processing, virtually
`rule out the option of a daily preparation. As a result, the
`nanosuspension supply needs to be prepared in bulk (or at least
`roll-outs) and analytically tested prior to the initiation of the
`toxicology studies. With careful planning of the manufacture
`well in advance, the supply can typically be turned around in a
`few weeks — a time-span that should be acceptable to the
`Project Team.
`
`In addition to supply,
`3.3.2.4. Nanosuspension storage.
`stability and storage are additional considerations for long-
`term usage of nanosuspensions. As mentioned above,
`the
`toxicology studies can be up to 3 months long, or perhaps
`longer, a time-frame which can pose concerns with respect to
`both

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