throbber
HHS Public Access
`Author manuscript
`Eur J Phann Sci. Author manuscript; available in PMC 2015 September 25.
`
`Published in final edited form as:
`Eur J Pharm Sci. 2011November20; 44(4): 522-533. doi:l0.1016/j.ejps.2011.09.014 .
`
`~ s=
`0 ...,
`
`~ -~
`
`0 ...,
`s:::
`m
`::::s
`c: en
`
`(")
`:::::!.
`
`"O -
`
`)>
`
`c: -~
`0 ...,
`s: m
`::::s
`c: cn
`"O -
`
`(")
`:::::!.
`
`~ -~
`
`0 ...,
`
`Nanoparticle agglomerates of fluticasone propionate in
`combination with albuterol sulfate as dry powder aerosols
`
`Nashwa El-Gendy1·3, Warangkana Pomputtapitak1, and Cory Berkland 1•2·*
`1Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS 66047.
`
`2Department of Chemical and Petroleum Engineering, The University of Kansas, Lawrence, KS
`66047.
`
`3Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-suef
`University, Egypt.
`
`Abstract
`Particle engineering strategies remain at the forefront of aerosol research for localized treatment of
`lung diseases and represent an alternative for systemic drug therapy. With the hastily growing
`popularity and complexity of inhalation therapy, there is a rising demand for tailor-made inhalable
`drug particles capable of affording the most proficient delivery to the lungs and the most
`advantageous therapeutic outcomes. To address this formulation demand, nanoparticle
`agglomeration was used to develop aerosols of the asthma therapeutics, fluticasone or albuterol. In
`addition, a combination aerosol was formed by drying agglomerates of fluticasone nanoparticles in
`the presence of albuterol in solution. Powders of the single drug nanoparticle agglomerates or of
`the combined therapeutics possessed desirable aerodynamic properties for inhalation. Powders
`were efficiently aerosolized (-75% deposition determined by cascade impaction) with high fine
`particle fraction and rapid dissolution. Nanoparticle agglomeration offers a unique approach to
`obtain high performance aerosols from combinations of asthma therapeutics.
`
`Keywords
`
`Fluticasone; albuterol; combination therapy; dry powder; aerosols
`
`1. Introduction
`
`Asthma and chronic obstructive pulmonary disease (COPD) are currently treated using
`either nebulizers, pressurized metered dose inhalers or dry powder inhalers (Dalby and
`Suman, 2003; Murnane et al., 2008b; Yang et al., 2008a). A major determinant of aerosol
`deposition in the respiratory tract is the aerodynamic size of particles and the polydispersity
`
`'To whom correspondence should be addressed. The University of Kansas, 2030 Becker Drive, Lawrence, KS 66047. Phone: (001)
`785- 864-1455. Fax: (001) 785- 864- 1454.berkland@ku.edu.
`Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. AB a service to our
`customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of
`the resulting proof before it is published in its final citable form. Please note that during the production process errors may be
`discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
`
`

`

`El-Gendy et al.
`
`Page2
`
`(Louey et al., 2004; Pilcer and Amighi, 2010; Pritchard, 2001). Inhaled drugs should ideally
`possess an aerodynamic diameter less than 5 µm. for delivery into the 'deep' lung for local
`therapy or systemic absorption (Weers et al., 2010). Nanoparticles (<0.5 µm.) are more likely
`to be exhaled, which may lead to dose variability (Shi et al., 2007). If delivered as a
`suspension, such small particles are also prone to particle growth due to Ostwald ripening
`and can suffer from uncontrolled agglomeration (Berkland, 2010). A major obstacle to
`inhaled therapeutics is the inability to efficiently deliver large quantities of a drug to the
`deep lung (Gillian, 2010).
`
`Natural aerosols, in particular, spores from molds and fungi as well as soot and asbestos,
`have a size and structure that allows them to aerosolize efficiently into the lungs. They are
`composed of underlying nanostructures that join together to form microparticles. Following
`this rationale, nanoparticle agglomerates were designed by formulating nanometer-sized
`drug particles, then assembling them to micron-sized clusters with the desired aerodynamic
`diameter (e.g., 1 µm. for treating distal airways or 3-5 µ m for treating upper airways)
`(Bailey et al., 2008; El Gendy et al., 2009; Plumley et al., 2009). By agglomerating
`nanoparticles under controlled process conditions, nanoparticle agglomerate dry powders
`can be tailored to the desired physical and chemical characteristics for aerosol delivery and
`dissolution (Aillon et al., 2010; El-Gendy and Berkland, 2009).
`
`Asthma is a disease that is commonly treated with two types of aerosolized agents;
`bronchodilators (fu agonists) and anti-inflammatory agents (steroidal compounds). Apart
`from acute asthma attacks, which are primarily treated with short acting ~2 agonists, there is
`a strong need for chronic therapy to reduce inflammation and to avoid asthma exacerbations
`(Barnes, 2002). Therapeutic interventions using combinations of a ~ agonist and a
`glucocorticoid have emerged as an effective asthma management strategy to control
`persistent asthma (Rajeswari et al., 2006). The use of~ agonists to prevent bronchial spasm
`and glucocorticoids to decrease inflammation is widely accepted (W estmeier and Steckel,
`2008). Combination formulations have also been suggested to be more effective than a
`single drug due to synergistic effects in the same target cell in the lung epithelia. It appears
`rational, therefore, to combine both substances in one particle instead of formulating a
`combination product containing both drugs in a physical mixture (Adi et al., 2008; Nelson et
`al., 2003; Papi et al., 2007).
`
`Combination products such as Advair and Symbicort are currently marketed. Advair
`combines fluticasone propionate and salmeterol xinafoate into one inhaler (Michael et al.,
`2000). Salmeterol (long acting~ agonists) does not replace the need for rescue inhalers,
`such as albuterol, which are still necessary for immediate relief of asthma symptoms (Kamin
`et al., 2007; Salpeter et al., 2006). Symbicort is another combination product containing
`budesonide and fonnoterol. Fluticasone propionate is a synthetic corticosteroid used to treat
`asthma, allergic rhinitis (hay fever) and eosinophilic esophagitis (Murnane et al., 2008a;
`Rehman et al., 2004; Vatanara et al., 2009). Albuterol sulfate is a short-acting P2
`adrenoreceptor agonist used for the relief ofbronchospasm in conditions such as asthma and
`COPD, and is currently one of the most prescribed bronchodilators for the treatment of
`bronchial asthma (Ahmad et al., 2009; Xu et al., 2010).
`
`Eur J Phann Sci. Author manuscript; available in PMC 2015 September 25.
`
`)>
`c: .....
`:J"
`0 ...,
`s:::
`m
`::J c:
`UJ
`0
`::::J.
`.....
`"'O
`
`~
`:T
`0 ...,
`s::: m
`::J c:
`UJ
`0
`::::J.
`~
`
`:J"
`
`)>
`c: .....
`0 ...,
`s::: m
`::J c:
`UJ
`0
`:::!.
`.....
`"'O
`
`

`

`El-Gendy et al.
`
`Page3
`
`Development of dry powder aerosols for delivering fluticasone and/or albuterol nanoparticle
`agglomerates as single anti-asthmatic therapies or in combination to achieve synergistic
`effect is herein described. The study illustrates the formulation of fluticasone nanoparticles
`using potentially acceptable surfactants that control the size and surface charge of the
`prepared nanoparticles. Also, albuterol nanoparticles free of excipients were engineered
`using different techniques. The nanoparticle suspensions were destabilized via ionic charge
`interactions using L-leucine. Combination drug formulations were prepared by adding
`albuterol aqueous solution to the fluticasone nanoparticle suspension followed by addition of
`L-leucine. The aerosol performance of these nanoparticles agglomerate formulations were
`fully characterized and compared to micronized stock drug.
`
`2. Materials and methods
`
`2.1. Materials
`
`Fluticasone propionate (Flu) and albuterol sulfate (Albu) were generously provided by 3M .
`L-a-phosphatidylcholine (lecithin; Lee), cetyl alcohol (CA), L-leucine (Leu) and
`polyvinylpyrrolidone K90 (PVP) were purchased from Sigma Chemical Co., USA. Pluronic
`F-127 (PL, Mw-12,220) was purchased from BASF, USA. Ethanol, acetone, potassium
`dihydrogen phosphate (KH2P04), disodium hydrogen phosphate (Na2HP04) and sodium
`chloride (NaCl) were purchased through Fisher Scientific, USA. Floatable dialysis
`membrane units (MWCO=lO kDa) were obtained from Spectrum Laboratories Inc., USA.
`Amicon Ultra Centrifugal filter units (MWC0=5 kDa) used for dissolution were purchased
`from Millipore, Co (Billerica, MA). Double-distilled water was used throughout the study,
`provided by an EASYpure® RODI (Barnstead International, USA).
`
`2.2. Nanoparticle formulation
`
`2.2.1. Preparation of fluticasone nanoparticle suspension-Nanoparticle
`suspensions of fluticasone propionate were prepared using antisolvent precipitation .
`Solutions of the drug in organic solvent (acetone or ethanol) were prepared at different
`concentrations and directly injected into water at a rate of 2.5 rnUmin. A variety of solvent/
`non-solvent ratios were precipitated under ultrasonication (probe-type sonicator, Fisher
`Scientific, Sonic Dismembrator) operating with an amplitude of 48% in an ice bath or under
`homogenization (probe-type homogenizer, Tissue tearor, Biospec Products, Inc.).
`Hydrophobic surfactants (cetyl alcohol and lecithin) were added to the drug solution while
`hydrophilic surfactants (PL F 127, PV A and PVP K90) were dissolved in the aqueous phase.
`
`2.2.2. Formulation of combination therapy-The combined formulation was prepared
`by adding albuterol sulfate dissolved in water to the precipitated fluticasone propionate
`nanosuspension during homogenization at 25,000 rpm. The two drugs were combined, at a
`ratio of2: 1 w/w, fluticasone propionate: albuterol sulfate (Papi et al., 2007; Westmeier and
`Steckel, 2008).
`
`2.2.3. Fabrication of albuterol nanoparticle suspension-Albuterol sulfate
`nanoparticles were prepared by precipitation or by a top-down (attrition) method.
`Concerning the precipitation technique, solutions of albuterol in water were prepared and
`
`Eur J Phann Sci. Author manuscript; available in PMC 2015 September 25.
`
`)>
`c: .....
`:J"
`0 ...,
`s::: m
`::J c:
`UJ
`0
`::::J.
`.....
`"'O
`
`~
`:T
`0 ...,
`s:::
`m
`::J c:
`UJ
`0
`::::J.
`~
`
`)>
`c: .....
`:J"
`0 ...,
`s::: m
`::J c:
`UJ
`0
`:::!.
`.....
`"'O
`
`

`

`El-Gendy et al.
`
`Page4
`
`directly injected into ethanol or acetone at a rate of2.5 mUmin. Various solvent/ non(cid:173)
`solvent ratios were used under ultrasonication operating with an amplitude of 48% or under
`homogenization. In the top-down method, albuterol nanoparticles were prepared by
`ultrasonicating or homogenizing a suspension of albuterol in acetone or ethanol. The
`concentration of the drug in the anti-solvent was varied between 0.2 and 1 mg/mL. The
`ultrasonication or homogenization time was also varied.
`
`2.3. Characterization of nanopartlcle suspensions
`
`The average size and polydispersities of the nanoparticle suspensions were determined by
`dynamic light scattering (Brookhaven, ZetaP ALS, SA). The same instrument was used to
`determine the zeta potential of the nanoparticles in 1 mM potassium chloride solution. Three
`runs of 15 cycles were acquired, and the mean zeta potential was recorded. Measurements
`were taken at an angle of 90° to the incident light source. Some samples were frozen at -80
`°C and lyophilized (FreeZone 1) for -36 hat a temperature of-50 °C under vacuum (-0.02
`millibar). Lyophilized powder was stored at room temperature for further characterization.
`
`2.4. Agglomeration of nanoparticle suspensions
`
`Nanoparticles were agglomerated via addition of an agglomerating agent. L-Leucine solution
`in water (2.5 mg/mL) was slowly injected into nanoparticle colloids during homogenization
`at 25,000 rpm for 30 s. The amount of L-leucine added was adjusted to a fluticasone: L(cid:173)
`leucine ratio equal to 1 : 1 for agglomerating the fluticasone suspension and the combination
`suspension. An albuterol: L-leucine ratio of 1: 1.5 was used for agglomerating the albuterol
`suspension.
`
`The agglomerated suspensions were incubated with the agglomerating agent for three hours.
`Then, the size of the prepared nanoparticle agglomerates was measured in Isoton diluent
`using a Coulter Multisizer 3 (Beckman Coulter Inc.) equipped with a 100 )JIIl aperture. The
`suspensions were kept overnight at room temperature to allow evaporation of organic
`solvent and then frozen at -80 °C. The frozen suspensions were transferred to the freeze
`dryer where drying lasted for -3 days. Lyophilized powder was stored at room temperature
`for further characterization.
`
`2.5. Particle size and morphology by transmission electron microscopy {TEM)
`
`Lyophilized powders were resuspended in Isotonic solution and the particle size and size
`distribution was detected using a Coulter Multisizer 3. In addition, the size and morphology
`of the lyophilized nanoparticles and nanoparticle agglomerate powders were evaluated using
`JEOL 1200 EXII transmission electron microscope. Prior to imaging, carbon-coated grids
`(Electron Microscopy Sciences) were placed on a droplet of the suspensions on a glass
`microscope slide to permit the adsorption of the particles onto the grid. After this, the grid
`was blotted with a filter paper and air dried for 1 h .
`
`2.6. Powder flow characteristics
`
`Nanoparticle agglomerate dry powders were poured through a glass funnel from a height of
`4 cm onto a level bench top. The angle that the side of the conical heap made with the
`horizontal plane was recorded as the angle of repose (tan e =height I radius). In addition,
`
`Eur J Phann Sci. Author manuscript; available in PMC 2015 September 25.
`
`)>
`c: .....
`:J"
`0 ...,
`s:::
`m
`::J c:
`UJ
`0
`::::J.
`.....
`"'O
`
`~
`:T
`0 ...,
`s::: m
`::J c:
`UJ
`0
`::::J.
`~
`
`:J"
`
`)>
`c: .....
`0 ...,
`s::: m
`::J c:
`UJ
`0
`:::!.
`.....
`"'O
`
`

`

`El-Gendy et al.
`
`Page5
`
`bulk and tapped densities were determined. Then, the Hausner ratio (tapped density I bulk
`density) and Carr's compressibility index (CJ [(tapped density- bulk density)/ tapped
`density X 100%] were calculated (Kumar et al., 2001; Louey et al., 2004).
`
`2.7. Evaluation of Aerosol performance of nanoparticle agglomerate dry powders
`
`2.7.1. Measurement of theoretical mass mean aerodynamic diameter-The
`geometric particle size and tap density measurements were used for calculating the
`theoretical mass mean aerodynamic diameter (dae) of the nanoparticle agglomerates (El(cid:173)
`Gendy et al., 201 Ob; Fiegel et al., 2008).
`
`2.7.2. Aerodynamic size distribution by time-of-flight analysis-The aerodynamic
`diameter and size distributions of the nanoparticle agglomerate powders were determined by
`time-of-flight measurement (TOP) using an Aerosizer LD (Amherst Instruments, Hadely,
`MA, USA) equipped with a 700 µm aperture operating at 6 psi. For these studies, -1 mg of
`the powder was added to the instrument disperser and data were collected for -60 s under
`high shear force (-3.4 kPa). The instrument size limits were 0.10--200 µm and particle
`counts were above 100,000 for all measurements.
`
`2.7.3. In vitro aerosol deposition of nanoparticle agglomerates by cascade
`impactor-An eight-stage Mark II Andersen Cascade Impactor (ACI, Tisch
`Environmental, Inc.) had stages with particle aerodynamic diameter specifications at a flow
`rate of28.3 Umin as follows: pre-separator (10.00 µm), stage 0 (9.00 µm), stage 1 (5.80
`µm), stage 2 (4.70 µm), stage 3 (3.30 µm), stage 4 (2.10 µm), stage 5 {l.10 µm), stage 6
`(0.70 µm), stage 7 (0.40 µm) and the final filter(< 0.40 µm). Aerodynamic behavior of
`nanoparticle agglomerate dry powders was assessed using the ACI and compared with that
`of the two drugs as received.
`
`The powder was delivered into the cascade impactor by placing capsules (gelatin type, size
`3, generously provided from Capsugel®, NJ, USA) containing 5 ± 0.5 mg of powder into a
`Plastiape Monodose Inhaler RSOl Model 7. The capsule was punctured and the powder was
`drawn through the cascade impactor which was operated at a flow rate of 28.3 Umin for 4 s.
`Dry powder aerosols deposited on each of the nine stages of the impactor were quantified by
`HPLC. After actuation, the device, capsule, adapter, throat, all plates, stages and filter were
`washed into separate volumetric flasks using ethanol (for fluticasone alone or Flu/Alu
`combination) or phosphate buffered saline (pH 7.4 for albuterol). Appropriate sample
`dilutions were made prior to testing by HPLC. Each sample was tested in triplicate.
`
`Concerning the combination formula, the powder deposited on stages was suspended in
`ethanol and was ultrasonicated in a bath-type sonicator (Branson 3510) for 30 min. Then,
`the solution was centrifuged (Beckman, Avanti) at-15,000 rpm for 30 min and the amount
`of fluticasone in the supernatant was determined using a reversed-phase HPLC method. As
`albuterol has a very slightly solubility in ethanol, the drug content in both supernatant and
`precipitate were detected by HPLC.
`
`All ACI experiments were performed under controlled conditions (21 ± 2 °C, 50 ± 5% RH)
`in triplicate. The emitted dose (ED) was defmed as the mass of drug delivered from the
`
`Eur J Phann Sci. Author manuscript; available in PMC 2015 September 25.
`
`)>
`c: .....
`:J"
`0 ...,
`s::: m
`::J c:
`UJ
`0
`::::J.
`.....
`"'O
`
`~
`:T
`0 ...,
`s:::
`m
`::J c:
`UJ
`0
`::::J.
`~
`
`)>
`c: .....
`:J"
`0 ...,
`s::: m
`::J c:
`UJ
`0
`:::!.
`.....
`"'O
`
`

`

`El-Gendy et al.
`
`Page6
`
`inhaler (i.e., total amount excluding the inhaler device and capsule) (Xu et al., 2010). The
`emitted fraction was determined as the percent of the emitted dose divided by the initial
`mass delivered into the impactor (Lechuga-Ballesteros et al., 2008; Shur et al., 2008; Yang
`et al., 2008b ).
`
`The fine particle fraction of the total dose (FPFm) was calculated as the percentage of
`aerosolized particles that reached the lower seven stages of the impactor (corresponding to
`aerodynamic diameters below 5 µm; stage 2-filter), or the lower five stages (corresponding
`to aerodynamic diameters below 3 µm; stage 4-filter) (El-Gendy et al., 2010a).
`
`The fine particle fraction of the emitted dose (FPFED) was determined from the cumulative
`mass distribution curve at 5 µm and 3 µm and was calculated as a function of the emitted
`dose. Additionally, mass median aerodynamic diameter (MMAD) and geometric standard
`deviation (GSD) were determined from the cumulative mass distribution curve (Pham and
`Wiedmann, 2001; Vanbever et al., 1999; Xu et al., 2010).
`
`2.8. Solid-state characterization
`
`2.8.1. Power X-Ray Diffraction (PXRD)-For analysis of crystallinity, X-ray diffraction
`analysis was performed using an XGEN-4000 (Scintag, Inc.). The powders were analyzed
`over the range of5° to 45° (20) at 45.0 kV and 35.0 mA.
`
`2.8.2. Differential scanning calorimetry (DSC)-DSC data of materials as received,
`nanoparticles and nanoparticle agglomerates were collected using a Q 100 DSC from TA
`Instruments. For thermogram acquisition, sample sizes of 1 to 5 mg were weighed into
`aluminum hermetic pans. Measurement was carried out under inert conditions (nitrogen
`flow of 50 mL/min) with a scan rate of 10 °C/min from 25 to 350 °C.
`
`2.8.3. Thermogravimetric analysis (TGA)-TGA was also performed using a Q50
`TGA from TA Instruments. A platinum sample pan was loaded with 5 ± 0.5 mg of sample
`and heated from 25 to 350 °C at a rate of 10 °C/min under dry nitrogen flowing at rate of 40
`mL/min. Data analysis was completed using Universal Analysis 2000 (Version 4.3A)
`software that was provided by TA Instruments.
`
`2.9. Determination of process yield
`
`The weight of dry powder for the prepared nanoparticle agglomerates was measured and the
`yield was calculated (El Gendy et al., 2009).
`
`2.10. Determination of drug content uniformity of nanoparticle agglomerates
`
`Fluticasone content in the dry powders was assessed by dispersing 1 mg of the lyophilized
`powder in 10 mL of ethanol. The dispersion was ultrasonicated in a bath-type sonicator for
`30 min. Then the solution was centrifuged at -15,000 rpm for 30 min to remove insoluble
`ingredients and the amount of fluticasone in the supernatant was determined using a
`reversed-phase HPLC method. For the combination formula, albuterol content was detected
`in both the pellet after dissolving in 10 mL PBS and in the supernatant using HPLC. Drug
`content in albuterol nanoparticle agglomerates was determined by dispersing 1 mg of the
`
`Eur J Phann Sci. Author manuscript; available in PMC 2015 September 25.
`
`)>
`c: .....
`:J"
`0 ...,
`s::: m
`::J c:
`UJ
`0
`::::J.
`.....
`"'O
`
`~
`:T
`0 ...,
`s:::
`m
`::J c:
`UJ
`0
`::::J.
`~
`
`)>
`c: .....
`:J"
`0 ...,
`s::: m
`::J c:
`UJ
`0
`:::!.
`.....
`"'O
`
`

`

`El-Gendy et al.
`
`Page?
`
`lyophilized powder in PBS. The dispersion was ultrasonicated in a bath-type sonicator for
`15 min. Then the amount of albuterol was determined using a reversed-phase HPLC method.
`All experiments were performed in triplicate and drug content was calculated (El Gendy et
`al., 2009).
`
`2.11. Dissolution studies
`
`The dissolution of the prepared nanoparticles and nanoparticle agglomerates was determined
`under sink condition and compared with the dissolution of the drugs as received. The
`dissolution offluticasone was carried out at 37 ± 0.5 °C in a 1 liter beaker. Lyophilized
`powder (-10 mg) was dispersed in 10 mL PBS (pH 7 .4) and was suspended in a floatable
`dialysis membrane unit (Mw cut-off= 10 kDa). The unit was allowed to float in 500 mL of
`PBS and the whole assembly was stirred at a constant speed (100 rpm) using a magnetic
`stirrer (Barnstead, Thermolyne MIRAK™). At predetermined time intervals for a total
`period of8 h, samples (1 mL) of the medium were withdrawn from the dialysis bag and
`replaced with fresh medium. Then, the samples were centrifuged for 30 min at -13,000 rpm.
`The supernatant was removed and the pellet was dissolved in 1 mL of ethanol. Fluticasone
`content was determined using reversed-phase HPLC. In the case of the combination
`formula, both pellet and supernatant were analyzed for albuterol using a reversed-phase
`HPLC method. Studies were conducted in triplicate.
`
`For albuterol dry powders, 5 mg was dispersed in 0.6 mL phosphate buffered saline (PBS,
`pH 7.4) and placed in a 5 kDa Ultra Centrifugal filter unit which was immersed inside a 10
`mL centrifugal tube containing PBS solution to a final volume of7 mL. All samples were
`incubated at 37 ± 0.5 °C and shaken at 100 rpm. One mL aliquots were taken at various time
`points up to 8 hours from the bulk solution and replaced with 1 mL of fresh PBS. The drug
`concentration was measured using reversed-phase HPLC. All experiments were performed
`in triplicate.
`
`2.12. Quantitative analysis of the drug concentrations by HPLC
`
`Drug content, dissolution, and ACI concentration on stages were carried out using reversed(cid:173)
`phase HPLC methods. A Shimadzu HPLC system including a solvent delivery pump
`(Shimadzu LC-lOAT), a controller (Shimadzu SCL-lOA), SIL-lOAxL autoinjector, and a
`SPD-lOA UV detector was used in this study. Chromatograms were acquired and analyzed
`using Shimadzu Class VP 4.3 software. A long Zorbax SB C-18 column (Agilent C; 4.6 mm
`x 100 mm) with a particle diameter of3.5 µm was used for separation. During the assay of
`fluticasone, the drug was eluted isocratically at a mobile phase flow rate of 1.2 mL/min and
`monitored with a UV detector operating at 238 nm. The mobile phase for the assay consisted
`of an acetonitrile and water mixture (65 :35 v/v) (Asmus et al., 2004; Steckel and Muller,
`1998). The run time for the assay was 10 minutes, and the retention time for fluticasone was
`3.9 ± 0.2 min. For analyzing albuterol samples, an isocratic system was used with mobile
`phase of 90: 10 v/v phosphate buffer (10 mM, pH3 .5): acetonitrile at a flow rate of 0.3
`mUmin and detection was performed at 225 nm (Kamin et al., 2007). The run time for the
`assay was 10 min, and the retention time for albuterol was 3 .85 ± 0.4 min.
`
`Eur J Phann Sci. Author manuscript; available in PMC 2015 September 25.
`
`)>
`c: .....
`:J"
`0 ...,
`s:::
`m
`::J c:
`UJ
`0
`::::J.
`.....
`"'O
`
`~
`:T
`0 ...,
`s::: m
`::J c:
`UJ
`0
`::::J.
`~
`
`:J"
`
`)>
`c: .....
`0 ...,
`s::: m
`::J c:
`UJ
`0
`:::!.
`.....
`"'O
`
`

`

`El-Gendy et al.
`
`Page 8
`
`3. Results and discussion
`
`3.1. Fluticasone nanopaticle suspensions made by precipitation
`
`To obtain small nanoparticles from poorly water soluble fluticasone propionate, a non(cid:173)
`solvent precipitation method was employed (Bilati et al., 2005). Selected surfactants were
`chosen from a list of excipients that may be appropriate for inhalation (Chougule et al.,
`2007; Pilcer and Amighi, 2010). Different suspensions were produced using water as anti(cid:173)
`solvent and a drug concentration ofO.l % or 0.2% w/w dissolved in ethanol or acetone.
`Fluticasone particles prepared without surfactants were very large with high polydispersity.
`Mean diameters of fluticasone nanoparticles made with individual surfactants were larger
`than those with combined surfactants. Particle size tended to increase and colloidal stability
`was difficult to maintain as fluticasone concentration was increased. Nanoparticles
`precipitated from acetone were larger than those precipitated from ethanol. Nanoparticle size
`appeared to decease slightly when using ultrasonication rather than homogenization during
`the precipitation process (Supplementary Table 1).
`
`The most successful fluticasone nanosuspension was prepared by precipitation from ethanol
`underultrasonication (0.1% w/vFlu + 0.01% w/v PVP + 0.005% w/v Lee). The surfactant
`combination employed yielded a small drug particle size (-400 nm) and low polydispersity
`(0.132). The charged surface of the nanoparticles (-12 m V) allowed the potential to
`destabilize this colloid via interaction with an agglomerating agent (Table 1 ). This formula
`was chosen for the preparation of the fluticasone nanoparticle agglomerates and for the
`combination formulation with albuterol sulfate in solution.
`
`3.2. Albuterol nanoparticle suspensions made by two approaches
`
`3.2.1. Production of nanoparticles by precipitation-Albuterol nanoparticles were
`first prepared by precipitation. A smaller nanoparticle size was produced when using
`acetone as non-solvent as opposed to ethanol. A decrease in particle diameter followed from
`a decrease of the drug concentration. The smallest nanoparticle size was obtained at a 2.5/25
`water/acetone ratio at a drug concentration of0.1 % w/v; however, the polydispersity was
`high with low yields for all precipitation trials (Supplementary Table 2).
`
`3.2.2. Production of nanoparticles by attrition-Nanoparticles were also produced
`by fragmenting micronized drug particles using homogenization or ultrasonication. The
`homogenizer was superior to ultrasonication in the preparation of albuterol nanoparticles
`causing a significant decrease in size with low polydispersity (Supplementary Table 3). With
`these considerations in mind, the A 7 nanoparticle formulation was selected which was
`prepared by homogenizing a suspension of the drug in acetone in a concentration of 1
`mg/mL for 15 min (Table 1).
`
`3.3. Agglomeration of the formulated nanoparticles
`
`Colloidal suspensions offluticasone nanoparticles, albuterol nanoparticles (A7) and
`fluticasone nanoparticles combined with albuterol in solution were destabilized using L(cid:173)
`leucine to disrupt the electrostatic repulsion between particles (Young et al., 2002). The
`resulting nanoparticle agglomerates had a geometric size of-3-5 }Utl (Table 2). After
`
`Eur J Phann Sci. Author manuscript; available in PMC 2015 September 25.
`
`)>
`c: .....
`:J"
`0 ...,
`s:::
`m
`::J c:
`UJ
`0
`::::J.
`.....
`"'O
`
`~
`:T
`0 ...,
`s::: m
`::J c:
`UJ
`0
`::::J.
`~
`
`:J"
`
`)>
`c: .....
`0 ...,
`s::: m
`::J c:
`UJ
`0
`:::!.
`.....
`"'O
`
`

`

`El-Gendy et al.
`
`Page9
`
`drying, powders showed a slightly broader size distribution (Fig. 1 ). In addition, the
`combination powders had a wider distribution compared to the single-drug formulations,
`perhaps due to large albuterol particles formed during freeze drying.
`
`The particle size of the nanoparticles and nanoparticle agglomerates was congruent with the
`structures observed in TEM micrographs. Fluticasone nanoparticles were slightly elongated
`with smooth surfaces and a particle size of -400 run (Fig. 2A). Nanoparticle agglomerates
`appeared as elongated nanoparticles that were agglomerated together into micron-sized
`clusters with a somewhat porous structure (Fig. 2B). The combination powders of
`fluticasone nanoparticles dried with albuterol in solution generally exhibited slightly larger
`particles with a rough surface. Presumably, albuterol in solution deposited on the rod-shaped
`fluticasone particles during drying (Fig. 2C). Small albuterol nanoparticles with a particle
`diameter less than 100 run were (Fig. 3A) [/agglomerated into micron-sized particles (Fig.
`3B).
`
`Powder properties for micronized drugs as received and nanoparticle agglomerates were also
`studied. Flowability and density characterization helped elucidate any differences in bulk
`powder properties (Table 3). Flowability indices were calculated from density differences
`and the angle of repose. The micronized drugs showed a larger angle of repose, greater tap
`density and higher values of the Hausner ratio and Carr's index compared to the
`nanoparticle agglomerates (Fig. 4). This was probably the result of a reduction of cohesive
`forces in nanoparticle agglomerates compared to drug powders as received. L-Leucine may
`have also reduced surface energy in nanoparticle agglomerate dry powders (Shur et al.,
`2008).
`
`3.4. Nanoparticle agglomerates yielded desirable aerosol characteristics
`Theoretical mass mean aerodynamic diameters ( daem) of the prepared nanoparticle
`agglomerates were calculated from the geometric particle size and tap density. The
`calculated daero (0.8--1. l µm) was appropriate for increasing the probability of aerosol
`deposition in the alveolar region of the lungs. Aerosizer LD time-of-fight (TOF)
`measurements of nanoparticle agglomerate dry powders also showed particles in the
`respirable size range (2 - 3.2 µm) with relatively narrow size distribution (Table 2 and Fig.
`5). The MAD value of the combination formula obtained by Aerosizer appeared to be close
`to that of the pure fluticasone nanoparticle agglomerates.
`
`Cascade impactor analysis is the standard technique for in vitro characterization of dry
`powder aerosols. Fluticasone nanoparticle agglomerates mainly deposited on stages 3 and 4,
`while albuterol nanoparticle agglomerates favored deposition on stages 4 and 5 of the
`impactor. The combination formula exhibited size distribution similar to fluticasone
`nanoparticle agglomerates with some additional powder deposition on upper stages. This
`may be explained by the fact that once fluticasone nanoparticle agglomerates were dried in
`the presence of albuterol in solution, forces such as van Der Waals are sufficient to hold all
`particles together. This may suggest that fluticasone nanoparticle agglomerates were indeed
`a 'carrier' for much of the albuterol. Albuterol in the combination showed a different
`deposition profile than the fluticasone, although the albuterol deposition was improved
`
`Eur J Phann Sci. Author manuscript; available in PMC 2015 September 25.
`
`)>
`c: .....
`:J"
`0 ...,
`s::: m
`::J c:
`UJ
`0
`::::J.
`.....
`"'O
`
`~
`:T
`0 ...,
`s:::
`m
`::J c:
`UJ
`0
`::::J.
`~
`
`)>
`c: .....
`:J"
`0 ...,
`s::: m
`::J c:
`UJ
`0
`:::!.
`.....
`"'O
`
`

`

`El-Gendy et al.
`
`Page 10
`
`compared to the albuterol as received. This may be due to the segregation of some larger
`albuterol particles from the fluticasone nanoparticle agglomerates.
`
`Conversely, drug powders as received largely deposited on the mouthpiece, throat and the
`upper stages (Fig. 6). A larger percentage of fluticasone (27%) and albuterol (28%) powder
`as received remained in the capsule shell and device when compared to the nanoparticle
`agglomerates (9% for Flu NA, 11 % for Albu NA and 18% for the combined formulation).
`This reflected the efficient aerosolization and high fine particle fraction of the nanoparticle
`agglomerates. In addition, the irregular shape of the nanoparticle agglomerates may decrease
`their contact area with device su

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