throbber
Dry Powder Inhaler Formulation
`
`Martin J Telko and Anthony J Hickey PhD DSc
`
`INTRODUCTION
`DRY POWDER INHALERS
`Development of the DPI
`Principles of Operation
`POWDER AND AEROSOL PHYSICS/PHYSICOCHEMICAL CHARACTERIZATION
`Crystallinity and Polymorphism
`Moisture Content and Hygroscopicity
`Particle Size
`Aerodynamic Diameter and Dynamic Shape Factor
`Fine-Particle Fraction
`Polydispersity
`Particle Sizing Techniques
`Surface Area and Morphology
`Forces of Interaction
`Surface Morphology
`Surface Area and Morphology Measurements
`DRUG PROPERTIES AND MANUFACTURE
`The Active Pharmaceutical Ingredient
`Active Pharmaceutical Ingredient Preparation
`FORMULATION
`Excipients
`Large Porous Particles
`Agglomerates
`PHARMACEUTICAL PROCESSING
`SUMMARY
`
`A drug product combines pharmacologic activity with pharmaceutical properties. Desirable per-
`formance characteristics are physical and chemical stability, ease of processing, accurate and
`reproducible delivery to the target organ, and availability at the site of action. For the dry powder
`inhaler (DPI), these goals can be met with a suitable powder formulation, an efficient metering
`system, and a carefully selected device. This review focuses on the DPI formulation and develop-
`ment process. Most DPI formulations consist of micronized drug blended with larger carrier
`particles, which enhance flow, reduce aggregation, and aid in dispersion. A combination of intrinsic
`physicochemical properties, particle size, shape, surface area, and morphology affects the forces of
`interaction and aerodynamic properties, which in turn determine fluidization, dispersion, delivery
`to the lungs, and deposition in the peripheral airways. When a DPI is actuated, the formulation is
`fluidized and enters the patient’s airways. Under the influence of inspiratory airflow, the drug
`particles separate from the carrier particles and are carried deep into the lungs, while the larger
`carrier particles impact on the oropharyngeal surfaces and are cleared. If the cohesive forces acting
`on the powder are too strong, the shear of the airflow may not be sufficient to separate the drug
`from the carrier particles, which results in low deposition efficiency. Advances in understanding of
`
`RESPIRATORY CARE • SEPTEMBER 2005 VOL 50 NO 9
`
`1209
`
`Liquidia's Exhibit 1056
`Page 1
`
`

`

`DRY POWDER INHALER FORMULATION
`
`aerosol and solid state physics and interfacial chemistry are moving formulation development from
`an empirical activity to a fundamental scientific foundation. Key words: dry powder inhaler, DPI,
`formulation development, particles, physico-chemical properties, drug delivery.
`[Respir Care 2005;
`50(9):1209–1227. © 2005 Daedalus Enterprises]
`
`INTRODUCTION
`
`Formulation development encompasses an array of pro-
`cesses in which an active pharmaceutical ingredient is in-
`corporated into a drug product. While biological activity is
`a prerequisite for a successful dosage form, it is not the
`sole determinant. Factors such as stability, processibility,
`delivery, and availability to the target organ contribute to
`an efficacious pharmaceutical system. Optimization of
`these factors is a key development task, and the final prod-
`uct is often a compromise between pharmaceutical and
`practical (ie, economic/engineering) considerations. For-
`mulation development is challenging because molecules
`with pharmacologic activity often display poor physico-
`chemical properties. In fact, the same molecular charac-
`teristics that confer pharmacologic activity (eg, high re-
`ceptor affinity)
`frequently limit a compound’s
`pharmaceutical utility, making it difficult or even unsuit-
`able for delivery.1,2 This is particularly true for many of
`the compounds that are identified by high-throughput
`screening methods.2,3
`Development of pharmaceuticals for inhalation is a par-
`ticular challenge, as it involves the preparation of a for-
`mulation and the selection of a device for aerosol disper-
`sion. The lungs have lower buffering capacity than other
`delivery sites (eg, the gastrointestinal tract or the blood),
`which limits the range of excipients that could enhance
`delivery outcomes. An additional variable, unique to pul-
`monary delivery, is the patient, both in terms of inhalation
`mode and respiratory-tract anatomy and physiology.4 There
`are many more ways to administer an inhaled aerosol than
`there are to swallow a tablet. Variability in delivered dose
`
`Martin J Telko and Anthony J Hickey PhD DSc are affiliated with the
`School of Pharmacy, University of North Carolina, Chapel Hill, North
`Carolina.
`
`Anthony J Hickey PhD DSc presented a version of this article at the 36th
`RESPIRATORY CARE Journal Conference, Metered-Dose Inhalers and Dry
`Powder Inhalers in Aerosol Therapy, held April 29 through May 1, 2005,
`in Los Cabos, Mexico.
`
`This research was partly supported by National Heart, Lung, and Blood
`Institute grant number HL67221. Martin J Telko is supported by a U.S.
`Pharmacopeia graduate fellowship.
`
`Correspondence: Anthony J Hickey PhD DSc, 1311 Kerr Hall, Dispersed
`Systems Laboratory, Division of Drug Delivery and Disposition, School
`of Pharmacy, CB 7360, University of North Carolina, Chapel Hill NC
`27599. E-mail: ahickey@unc.edu.
`
`to an individual or a population of patients can be sub-
`stantial.5,6 Consequently, reproducible therapeutic effect is
`difficult to assure.
`Treating respiratory diseases with inhalers requires de-
`livering sufficient drug to the lungs to bring about a ther-
`apeutic response. For optimal efficacy, drug administra-
`tion must be reliable, reproducible, and convenient. This
`goal can be achieved by a combination of formulation,
`metering, and inhaler design strategies.7 The technical and
`clinical aspects of device design and selection have been
`extensively reviewed elsewhere.8 –10 The following discus-
`sion outlines the design of dry powder inhaler (DPI) for-
`mulations to achieve the delivery goals. Formulation de-
`velopment and characterization strategies and processing
`methods will be discussed, with emphasis on their effect
`on stability, manufacturing feasibility, delivery, and bio-
`availability. To that end, an understanding of dry powder
`physics and surface chemistry is essential. The text fo-
`cuses on broad concepts and examples, with only sparing
`use of equations.
`
`DRY POWDER INHALERS
`
`Development of the DPI
`
`Inhaled drug delivery systems can be divided into 3
`principal categories: pressurized metered-dose inhalers
`(pMDIs), DPIs, and nebulizers, each class with its unique
`strengths and weaknesses. This classification is based on
`the physical states of dispersed-phase and continuous me-
`dium, and within each class further differentiation is based
`on metering, means of dispersion, or design. Nebulizers
`are distinctly different from both pMDIs and DPIs, in that
`the drug is dissolved or suspended in a polar liquid, usu-
`ally water. Nebulizers are used mostly in hospital and
`ambulatory care settings and are not typically used for
`chronic-disease management because they are larger and
`less convenient, and the aerosol is delivered continuously
`over an extended period of time. pMDIs and DPIs are
`bolus drug delivery devices that contain solid drug, sus-
`pended or dissolved in a nonpolar volatile propellant or in
`a dry powder mix (DPI) that is fluidized when the patient
`inhales. The clinical performance of the various types of
`inhalation devices has been thoroughly examined in many
`clinical trials, which have been reviewed by Barry and
`O’Callaghan,10 and more recently by Dolovich et al.8 Those
`authors concluded that none of the devices are clinically
`
`1210
`
`RESPIRATORY CARE • SEPTEMBER 2005 VOL 50 NO 9
`
`Liquidia's Exhibit 1056
`Page 2
`
`

`

`DRY POWDER INHALER FORMULATION
`
`Table 1. Dry Powder Inhalers Versus Metered-Dose Inhalers
`
`Advantages of the Dry Powder Inhaler
`Environmental sustainability, propellant-free design
`Little or no patient coordination required
`Formulation stability
`Disadvantages of the Dry Powder Inhaler
`Deposition efficiency dependent on patient’s inspiratory airflow
`Potential for dose uniformity problems
`Development and manufacture more complex/expensive
`
`(Adapted from Reference 18.)
`
`superior and that device selection should be guided by
`other factors, such as convenience, cost, and patient pref-
`erence.
`First approved in 1956, the pMDI was the first modern
`inhaler device.11 With a global market share of about 80%,
`the pMDI remains the most widely used device.12 The
`development of DPIs has been motivated by the desire for
`alternatives to pMDIs, to reduce emission of ozone-deplet-
`ing and greenhouse gases (chlorofluorocarbons and hy-
`drofluoroalkanes, respectively) that are used as propel-
`lants, and to facilitate the delivery of macromolecules and
`products of biotechnology. Concurrently, DPIs proved suc-
`cessful in addressing other device and formulation-related
`shortcomings of the pMDI. DPIs are easier to use, more
`stable and efficient systems. Because a pMDI is pressur-
`ized, it emits the dose at high velocity, which makes pre-
`mature deposition in the oropharynx more likely.13,14 Thus,
`pMDIs require careful coordination of actuation and inha-
`lation. Despite enhancements to their design (eg, use of
`spacers),15 incorrect use of pMDIs is still a prevalent prob-
`lem; Giraud and Roche found that poor coordination of
`actuation and inhalation caused decreased asthma control
`in a substantial proportion of patients treated with corti-
`costeroid pMDIs.16 Since DPIs are activated by the pa-
`tient’s inspiratory airflow, they require little or no coordi-
`nation of actuation and inhalation. This has frequently
`resulted in better lung delivery than was achieved with
`comparable pMDIs.17
`Since DPIs are typically formulated as one-phase, solid-
`particle blends, they are also preferred from a stability and
`processing standpoint.18 Dry powders are at a lower en-
`ergy state, which reduces the rate of chemical degradation
`and the likelihood of reaction with contact surfaces. By
`contrast, pMDI formulations, which include propellant and
`cosolvents, may extract organic compounds from the de-
`vice components.19 Table 1 summarizes the main advan-
`tages and disadvantages of the DPI (versus the pMDI). For
`more detail on the evolution of aerosol delivery devices,
`excellent reviews are available.11,20
`The development of several new DPI devices, which
`have been reviewed elsewhere,18,21–23 and the commercial
`
`success of the bronchodilator-corticosteroid combination
`product Advair (GlaxoSmithKline, Research Triangle Park,
`North Carolina) have further stimulated interest in and
`development of DPIs.7
`
`Principles of Operation
`
`Figure 1 shows the principles of DPI design. Most DPIs
`contain micronized drug blended with larger carrier parti-
`cles, which prevents aggregation and helps flow. The im-
`portant role these carrier particles play is discussed later in
`this article. The dispersion of a dry powder aerosol is
`conducted from a static powder bed. To generate the aero-
`sol, the particles have to be moved. Movement can be
`brought about by several mechanisms. Passive inhalers
`employ the patient’s inspiratory flow. When the patient
`activates the DPI and inhales, airflow through the device
`creates shear and turbulence; air is introduced into the
`powder bed and the static powder blend is fluidized and
`enters the patient’s airways. There, the drug particles sep-
`arate from the carrier particles and are carried deep into
`the lungs, while the larger carrier particles impact in the
`oropharynx and are cleared. Thus, deposition into the lungs
`is determined by the patient’s variable inspiratory air-
`flow.24 –26 Inadequate drug/carrier separation is one of the
`main explanations for the low deposition efficiency en-
`countered with DPIs.27 Dose uniformity is a challenge in
`the performance of DPIs. This is a greater concern with
`powders than with liquids because of the size and discrete
`nature of the particulates.
`Various dispersion mechanisms have been adopted for
`DPIs.22 While most DPIs are breath-activated, relying on
`inhalation for aerosol generation, several power-assisted
`devices (pneumatic, impact force, and vibratory) have been
`developed or are currently under development. These de-
`vices are being considered for the delivery of systemically
`active drugs that have narrow therapeutic windows.28 It is
`important to note that these “active” inhalers are not sub-
`ject to the same limitations as passive inhalers and have a
`different advantage/disadvantage profile. Moreover, it has
`been suggested that if shear and turbulence could be stan-
`dardized by using a dispersion mechanism that is indepen-
`dent of the patient’s breath, high delivery efficiency and
`reproducibility might be achieved. Thus, an active inhaler
`might provide formulation-independent delivery.29 There
`are no commercially available active-dispersion DPIs.
`Therefore, in the interest of brevity, these devices are not
`discussed here; the reader is instead referred to other lit-
`erature.28 –30
`
`POWDER AND AEROSOL PHYSICS/PHYSICOCHEMICAL
`CHARACTERIZATION
`
`The character of particulate systems is central to the
`performance of DPIs. Powders present unique design chal-
`
`RESPIRATORY CARE • SEPTEMBER 2005 VOL 50 NO 9
`
`1211
`
`Liquidia's Exhibit 1056
`Page 3
`
`

`

`DRY POWDER INHALER FORMULATION
`
`Fig. 1. Principle of dry powder inhaler design. The formulation typically consists of micronized drug blended with larger carrier particles,
`dispensed by a metering system. An active or passive dispersion system entrains the particles into the patient’s airways, where drug
`particles separate from the carrier particles and are carried into the lung.
`
`lenges. Powders are 2-phase gas-solid systems. When pow-
`ders are static, they behave as solids; when they flow, they
`resemble liquids, easily assuming the shape of the con-
`taining vessel.31 When a powder is dispersed in air, as is
`the case after actuation of a DPI, in many ways it conforms
`to its carrier gas (unlike gases or vapors, pharmaceutical
`powders are nonequilibrium systems). Whereas gas and
`liquid behavior is understood and accurately predicted by
`equations derived from first principles, physical equations
`governing powders are often empirical or rely on assump-
`tions that are only approximations to real systems, such as
`homogeneity in size and shape of particles. As a conse-
`quence, equations describing the behavior of solids are
`less predictive than their fluid counterparts. The reader is
`referred to texts on multiphase flow phenomena.32–36
`Powder properties can vary widely. Powder features,
`such as the physicochemical properties and morphology of
`its constituent particles and the distribution of particle sizes,
`contribute to variability. Unlike liquid solutions or gas
`mixtures, powders are never completely homogeneous (at
`primary particulate scale) and segregation by size, which
`is a function of external forces, is always a potential prob-
`lem. The aerodynamic behavior, which has a profound
`effect on the disposition of drug from a DPI, is particularly
`sensitive to powder properties.
`
`Crystallinity and Polymorphism
`
`Many pure organic substances, including most drugs,
`are crystalline. A crystal is a solid in which the molecules
`
`or ions are arranged in an ordered, repeating pattern (the
`unit cell) extending in 3 spatial dimensions. Crystalline
`systems are defined by the intermolecular spacing (ie, bond
`lengths and bond angles) of the unit cell, which can be
`determined by x-ray diffraction.37 There are 7 crystal
`classes, which yield 14 distinct lattice structures.38 The
`arrangement of molecules into crystals is governed by non-
`covalent interactions, including hydrogen bonding, van der
`Waals forces, ␲-␲stacking, and electrostatic interactions.39
`Nearly one third of all drugs are known to display poly-
`morphism,40 which is the ability of a solid to exist in more
`than one crystal from. A prominent example of a poly-
`morphic pharmaceutical is carbamazepine, which has 4
`known polymorphs, one of which was discovered almost
`30 years after identification of the first polymorphs.41 De-
`termination of the polymorphic forms of a drug is an im-
`portant part of the formulation-development process, be-
`cause polymorphic forms are not equivalent. Different
`polymorphs are at different energy states and thus have
`different properties, including stability, solubility, and even
`bioavailability.38 Identification of all polymorphs of a drug
`also has important economic implications, because a sep-
`arate patent can be granted for each polymorph.40
`It is also possible to generate a noncrystalline solid. In
`most cases this involves cooling a fluid so rapidly that its
`molecules lose mobility before assuming their lattice po-
`sitions. A noncrystalline material is considered amorphous
`because it lacks long-range order. Amorphous materials
`have higher Gibbs free energies than crystals; thermody-
`namic laws predict that, in the long-term, materials seek to
`
`1212
`
`RESPIRATORY CARE • SEPTEMBER 2005 VOL 50 NO 9
`
`Liquidia's Exhibit 1056
`Page 4
`
`

`

`DRY POWDER INHALER FORMULATION
`
`Fig. 3. Hygroscopic growth. Particles absorb moisture as they
`traverse the humid environment of the airways, resulting in in-
`creased particle size.
`
`Knowledge of crystallization and polymorphism is still
`unfolding, and the ability to predict polymorphism remains
`imperfect. In most solids, a large number of different in-
`termolecular interactions are possible, but few are actually
`observed.48 The difficulties involved in crystallization are
`illustrated by several reported cases of “disappearing poly-
`morphs.” These cases were characterized by difficulty in
`resynthesis of a polymorph after initial synthesis, despite
`seemingly identical procedure and conditions.49 Control-
`ling crystallization is at the heart of “particle engineering,”
`which is a term that is used with increasing frequency in
`the pharmaceutical and chemical literature. Control over
`the crystallization process could yield particles with pre-
`cisely engineered morphology; co-crystallization (inclu-
`sion of functional impurities into the crystal) could then
`become a formulation strategy, resulting in “supramolecu-
`lar pharmaceutics.”47
`
`Moisture Content and Hygroscopicity
`
`Hygroscopicity is the intrinsic tendency of a material to
`take on moisture from its surroundings. The hygroscopic-
`ity is affected by the crystallinity of the material and the
`morphology of the particles. Hygroscopic drugs present a
`greater risk of physical and chemical instability. Moisture
`uptake and loss due to changes in relative humidity can
`result in local dissolution and recrystallization, leading to
`irreversible aggregation through solid bridge formation,22
`which can adversely affect aerosol generation and lung
`deposition.50 Hygroscopicity can also alter the adhesive
`and cohesive properties, or, in more extreme situations,
`substantially increase particle size.51 Hygroscopic growth
`(Fig. 3) involves the uptake of moisture, which will reach
`equilibrium in droplets as a function of the water activity
`of the solution formed and the surrounding atmosphere of
`water vapor; the Kelvin-Gibbs equation describes the phe-
`nomenon involved.52 Hygroscopic growth has implications
`for the equilibrium moisture content of the particles in the
`dosage form prior to aerosol generation; it can cause chem-
`ical or physical instability of the product. For aerosols, the
`physical instability is more important, because agglomer-
`ation may be irreversible and lead to an inability to gen-
`erate aerosol particles of respirable size. As aerosol parti-
`cles enter the lungs, they experience a high-humidity
`
`Fig. 2. Crystal habit. Inhibition of growth in one of more spatial
`directions (ka, kb, and kc) results in particles with plate or needle
`morphology.
`
`minimize their free energies by transitioning to lower en-
`ergy states (eg, crystallization). Whether this will occur at
`a timescale that need be of concern to the pharmaceutical
`scientist is governed by the chemical kinetics of the sys-
`tem.
`Different polymorphs can be discerned in terms of var-
`ious physicochemical properties. Polymorphs usually dif-
`fer in density, melting point, solubility, and hygroscopic-
`ity. The most stable polymorph frequently has the highest
`density, highest melting point, and lowest solubility. Dis-
`criminating analytical methods to characterize polymorphs
`include x-ray diffraction and thermal analysis, such as
`differential scanning calorimetry.38 To reduce the risk of
`transformation during processing or storage, the most sta-
`ble polymorph is typically selected for development, pro-
`vided its other properties are manageable.
`While crystallinity refers to the geometry of the unit
`cell, crystal habit describes the morphology of particles,
`which can vary independently of the crystal lattice struc-
`ture if crystal growth rates (during precipitation) vary in
`some dimension (Fig. 2).42 Crystal habit is important be-
`cause particle shape affects aerodynamic behavior and,
`thus, lung deposition. Crystallization and crystal habit are
`influenced by various factors, including identity of sol-
`vent,43,44 impurities present during crystallization,45 and
`processing variables such as temperature, pH, solution vol-
`ume, and viscosity.46
`Some compounds will spontaneously incorporate sol-
`vent molecules into the lattice structure upon crystalliza-
`tion or storage at certain conditions. This phenomenon has
`been referred to as pseudopolymorphism, and is relevant
`for many drugs that exist as solvates or hydrates.47 It is
`important to understand the conditions that will result in
`hydration, because, as with true polymorphs, hydrates dif-
`fer in their physicochemical properties.
`
`RESPIRATORY CARE • SEPTEMBER 2005 VOL 50 NO 9
`
`1213
`
`Liquidia's Exhibit 1056
`Page 5
`
`

`

`DRY POWDER INHALER FORMULATION
`
`Fig. 4. Strategies for altering the aerodynamic diameter. A: Aero-
`dynamic diameter equation. B: Large, low-density porous parti-
`cles. C: Needle-shaped particles. Particles in both B and C are
`expected to have aerodynamic diameters smaller than their size
`would suggest. Dae ⫽ aerodynamic diameter. Deq ⫽ unit density of
`equivalent volume sphere. ␳p ⫽ particle density. ␳o ⫽ unit density.
`X ⫽ dynamic shape factor.
`
`environment (99.5% relative humidity at 37°C). Although
`they may not reach equilibrium during transit, susceptible
`aerosol particles may be subject to hygroscopic growth,
`which increases particle dimensions and affects lung dep-
`osition.53 Hygroscopic growth can be prevented by coating
`the drug particles with hydrophobic films.52 However, no
`such approach has been successfully implemented in a
`marketed formulation.
`The equilibrium moisture content of a drug and excip-
`ient must be determined over a range of relative humidi-
`ties, so that storage conditions can be defined and other
`protective measures considered. Excipients that modify
`the hygroscopic properties of a drug may need to be con-
`sidered.
`
`Particle Size
`
`Particle size is the single most important design variable
`of a DPI formulation. Methods for determining particle
`size and distribution use various geometric features or phys-
`icochemical properties.54 Among these, aerodynamic di-
`ameter is the most relevant to lung delivery and ultimately
`to therapeutic effect. There is substantial literature from
`the fields of industrial hygiene, environmental and occu-
`pational medicine, and pharmaceutical sciences that links
`aerodynamic size and size distribution to the probability of
`deposition in specific lung sites. The statistical basis for
`these relationships in terms of variability in airways ge-
`ometry and lung physiology, both between individuals and
`within an individual, has been sufficient to allow the de-
`velopment of semi-empirical models correlating particle
`size with lung deposition.55
`
`Aerodynamic Diameter and Dynamic Shape Factor
`
`Aerodynamic diameter is the most appropriate measure
`of aerosol particle size, because it relates to particle dy-
`namic behavior and describes the main mechanisms of
`aerosol deposition; both gravitational settling and inertial
`impaction depend on aerodynamic diameter. To reach the
`peripheral airways, where drug is most efficiently absorbed,
`particles need to be in the 1–5 ␮m aerodynamic diameter
`range.56 Particles larger than 5 ␮m usually deposit in the
`oral cavity or pharynx, from which they are easily cleared.
`In contrast, particles smaller than 0.5 ␮m may not deposit
`at all, since they move by Brownian motion and settle very
`slowly. Moreover, they are inefficient, as a 0.5-␮m sphere
`delivers only 0.1% of the mass that a 5-␮m sphere carries
`into the lungs. In a series of studies, the optimal particle
`size of aerosol particles was examined for several different
`therapeutic agents in patients with different disease states.
`Although some differences due to patient lung function
`were noted, the optimal size was always in this 1–5 ␮m
`range.57– 60
`
`The aerodynamic diameter, Dae, is defined by the diam-
`eter of an equivalent volume sphere of unit density Deq
`with the same terminal settling velocity as the actual par-
`ticle. For particles larger than 1 ␮m, the following expres-
`sion describes the relationship between these dimensions.
`
`Dae ⫽ Deq冑冉 ␳p
`
`␳oX
`
`冊
`
`where ␳
`p and ␳o are particle and unit densities, and ␹is the
`
`dynamic shape factor. Pharmaceutical powders are rarely
`spherical, and shape factors are dimensionless measures of
`the deviation from sphericity. The dynamic shape factor is
`the ratio of the actual resistance force experienced by the
`nonspherical falling particle to the resistance force expe-
`rienced by a sphere having the same volume.61 Dynamic
`shape factors are determined either experimentally or us-
`ing more complex models that are beyond the scope of this
`paper. A very thorough review of this concept, with values
`for common shapes, is provided by Hinds.61
`The above equation merits closer examination. As dis-
`cussed, it is the aerodynamic diameter that determines lung
`disposition, irrespective of geometric particle size (to a cer-
`tain point). The aerodynamic diameter can be decreased by
`decreasing the particle size, decreasing particle density, or
`increasing the dynamic shape factor. This concept is shown
`graphically in Figure 4, and is discussed in more detail below.
`All 3 of these approaches have been applied.
`
`Fine-Particle Fraction
`
`“Fine-particle fraction” is the percentage of particles in
`the fine-particle range (1–5 ␮m). “Fine-particle mass” is
`
`1214
`
`RESPIRATORY CARE • SEPTEMBER 2005 VOL 50 NO 9
`
`Liquidia's Exhibit 1056
`Page 6
`
`

`

`DRY POWDER INHALER FORMULATION
`
`the total mass of the particles that are in the fine-particle
`range.62 The fine-particle component of aerosols is usually
`defined as the percentage of particles that are smaller than
`5 ␮m aerodynamic diameter, or, in the case of certain
`particle-sizing instruments, a cut-off diameter that is close
`to 5 ␮m. Quite often this may be in the 6 –7 ␮m range. The
`danger of adopting these values as definitive measures of
`equivalency is associated with the effect of particle size on
`deposition.63 This is considered more later in this article.
`
`Polydispersity
`
`For drug delivery it is the convention to consider the
`mass associated with each particle size as the frequency
`term in the distribution, since this relates directly to dose.
`Conventional statistical properties apply to populations of
`particles (ie, mode, mean, and median). It is usual to de-
`fine the central tendency of numbers of aerosol particles
`by the mass median aerodynamic diameter, which reflects
`the particle size that divides the distribution in half as a
`function of mass. Monomodal distributions may conform
`to log-normal mathematical interpretation, in which case
`the breadth of the distribution can be expressed in terms of
`the geometric standard deviation, which is usually derived
`by dividing the particle size at the 84th percentile by the
`median size, to achieve a dimensionless number greater
`than 1.
`When considering particle size, the degree of polydis-
`persity (ie, the range of particle sizes around the mode) is
`also important. The simplest and preferred system exhibits
`a single mode. However, many pharmaceutical aerosols
`will exhibit more than one mode. It is conceivable that 2
`completely different aerosol distributions (eg, small me-
`dian size with narrow distribution or large median size
`with broad distribution) could give exactly the same fine-
`particle fraction. However, within the fine-particle frac-
`tion, the aerosol would exhibit different sizes, leading to
`differences in regional lung deposition, resulting in vari-
`ations in therapeutic effect. Thus, degree of dispersity is an
`important consideration for both quality and efficacy of
`pharmaceutical aerosols.64 The nature of the aerosol dis-
`tribution must be established accurately if its implications
`for deposition and efficacy are to be understood.
`Another consideration relates to the standard DPI for-
`mulation, which is frequently bimodal, because it contains
`micronized drug and substantially larger carrier particles.
`Recognizing the potential for multimodal distributions is
`important to the application of statistical methods to the
`interpretation of the data. Traditional methods of data in-
`terpretation (eg, log-normal mathematical fits to distribu-
`tions65) may be superseded by other mathematical ap-
`proaches66 or nonlinear curve-fitting using calibration
`data.67
`
`Fig. 5. The electrical low-pressure impactor. (Courtesy of Dekati
`Ltd, Tampere, Finland.)
`
`Particle Sizing Techniques
`
`Several techniques are available for determining parti-
`cle size distributions; they have been described in depth
`elsewhere65 and will be covered briefly here. The aerosol
`sizing techniques can be classified as (1) inertial methods,
`(2) light-scattering methods, or (3) imaging methods.
`
`Cascade Impactor. Cascade impactors,68,69 including
`multi-stage liquid impingers, are the most widely used
`instruments for sizing aerosols; they are recommended by
`both the United States and the European pharmacopeias.
`Their utility stems from the fact that they directly measure
`aerodynamic size, rather than equivalent volume diameter
`(based on cross-sectional area) like the other methods. The
`theory of cascade impactor operation has been described
`in depth elsewhere.70 Briefly, cascade impactors contain
`several stages, with orifices of decreasing size, stacked on
`top of each other. When the aerosol is drawn through the
`impactor, the particles deposit on different stages, based
`on their inertia. After each run, the impactor is disassem-
`bled and the mass of particles deposited on each stage is
`determined, mostly via analytical methods (dissolution in
`solvent, followed by chromatography or ultraviolet absor-
`bance). A cut-off diameter is associated with each stage of
`the impactor. This diameter varies with airflow, so the
`impactor must be calibrated for different flow rates. This
`airflow dependence allows investigation of the effect of
`different inspiratory flow rates on deposition.
`The electrical low-pressure impactor71 (Figure 5) is a
`rather recent modification of the cascade impactor. Parti-
`cles passing through the electrical low-pressure impactor
`are charged before traversing the cascade of stages. Their
`impact on the stages produces an electrical current that is
`detected and converted into particle-size data that can be
`interpreted immediately. The utility of the electrical low-
`pressure impactor has been demonstrated in the sizing of
`particles in diesel engine exhaust71–76 and other combus-
`
`RESPIRATORY CARE • SEPTEMBER 2005 VOL 50 NO 9
`
`1215
`
`Liquidia's Exhibit 1056
`Page 7
`
`

`

`DRY POWDER INHALER FORMULATION
`
`Fig. 6. Particle sizing via laser-light scattering.
`
`Fig. 7. Particle sizing via digital image analysis.
`
`tion processes.77 A limitation of the electrical low-pressure
`impactor is that it is not suitable for particles larger than 20
`␮m, so it cannot be used to size carrier particles, which
`limits its utility for sizing pharmaceutical aerosols. Based
`on a PubMed search, reference to electrical low-pressure
`impactors in medical/pharmaceutical journals is limited to
`a single publication, in which an electrical low-pressure
`impactor was used for sizing sub-micron size pMDI par-
`ticles.78 However, the electrical low-pressure impactor has
`great potential to simplify the aerosol sizing process and is
`likely to make an impact in the field in the future.
`
`Light Scattering and Laser Diffraction. Light-scatter-
`ing methods, especially laser-light-scattering, are quite
`commonplace in formulation development. The operating
`principle of laser-light scattering is depicted in Figure 6.
`An expanded laser beam is passed through a sample that is
`being drawn through a measuring zone. Different size par-
`ticles diffract the light at d

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