throbber
Development of Clinical Dosage Forms for a Poorly
`Water Soluble Drug I: Application of Polyethylene
`Glycol–Polysorbate 80 Solid Dispersion Carrier System
`
`ROSE-MARIE DANNENFELSER, HANDAN HE, YATINDRA JOSHI, SIMON BATEMAN, ABU T.M. SERAJUDDIN
`
`Novartis Pharmaceuticals Corporation, 1 Health Plaza, East Hanover, New Jersey 07936
`
`Received 11 September 2003; revised 23 November 2003; accepted 23 November 2003
`
`ABSTRACT: Different formulation approaches were evaluated to ensure that the formu-
`lation of a poorly water soluble compound chosen during early development achieves
`optimum bioavailability. The insoluble compound has an aqueous solubility of 0.17 mg/mL
`at 25  18C, a relatively high permeability (Caco2 Papp ¼ 6.1 104 cm/min), and poor
`bioavailability in dogs (dry blend formulation). Based on the prediction by GastroPlusTM,
`the oral absorption of this compound is sensitive to its apparent solubility and particle
`size. The oral bioavailability of three different formulations was compared in a dog model:
`a cosolvent-surfactant solution, a solid dispersion in a mixture of polyethylene glycol 3350
`and polysorbate 80, and a dry blend of micronized drug with microcrystalline cellulose.
`In absence of a parenteral injection, the bioavailability of the solution was considered to
`be 100%, and the relative oral bioavailability of the three formulations was 100, 99.1, 9.8,
`respectively. Comparable bioavailability was obtained with the solid dispersion and
`the cosolvent-surfactant solution, both of which showed a 10-fold higher bioavailability
`than the dry blend. Thus, a 20 mg dose strength capsule containing the solid dispersion
`formulation was selected for clinical development. The selected solid dispersion
`system was physically and chemically stable for at least 16 months at 258C/60% RH.
`In conclusion, the bioavailability of a poorly water soluble drug was greatly enhanced
`using the solid dispersion formulation containing a water soluble polymer with a surface
`active agent. ß 2004 Wiley-Liss, Inc. and the American Pharmacists Association J Pharm Sci
`93:1165–1175, 2004
`Keywords: poorly soluble; solid dispersion; PEG; bioavailability enhancement
`
`INTRODUCTION
`
`Due to the application of combinatorial chemistry
`and high-throughput screening during drug dis-
`covery in recent years, a majority of new drug
`candidates exhibit poor aqueous solubility, result-
`ing in the development of bioavailable dosage
`forms for such compounds to be very challenging
`for formulation scientists. A poorly water-soluble
`
`Correspondence to: Rose-Marie Dannenfelser (Telephone:
`862-778-2069; Fax: 973-781-8487;
`E-mail: rosemary.dannenfelser@pharma.novartis.com)
`
`Journal of Pharmaceutical Sciences, Vol. 93, 1165–1175 (2004)
`ß 2004 Wiley-Liss, Inc. and the American Pharmacists Association
`
`compound has classically been defined as one
`dissolving in less than 1 part per 10000 parts of
`water,1 which, in other words, is less than 100 mg/
`mL. A concentration of 100 mg/mL, however, may
`not appear to be very low in comparison with the
`aqueous solubility of <1 mg/mL (1 part per million)
`for compounds that in recent years are commonly
`emerging from drug discovery pipelines. A poorly
`water-soluble drug, more recently, has been
`defined in general terms to require more time to
`dissolve in the gastrointestinal fluid than it takes
`to be absorbed in the gastrointestinal tract.2 Thus,
`a greater understanding of dissolution and ab-
`sorption behaviors of drugs with low aqueous
`solubility is required to successfully formulate
`them into bioavailable drug products.
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 93, NO. 5, MAY 2004
`
`1165
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1040-0001
`
`

`

`1166
`
`DANNENFELSER ET AL.
`
`In efforts to enhance a drug’s dissolution rate
`and, in some cases, apparent aqueous solubility
`under gastrointestinal pH conditions, various
`techniques have been applied, including, but not
`limited to, particle size reduction. However, there
`is a practical limit with particle size reduction
`in how much can be achieved by conventional
`approaches.3 Therefore, formulation approaches
`are being explored to enhance bioavailability of
`poorly water-soluble drugs. One such formulation
`approach that has been shown to significantly
`enhance absorption of such drugs is the use of solid
`dispersions.3 – 18
`This article provides a case study leading to
`the development of a bioavailable dosage form for
`a poorly water-soluble drug, LAB687, for early
`phase clinical trials. The formulation approaches
`that have been investigated include a dry blend
`of micronized drug with excipients, a cosolvent-
`surfactant solution, and a solid dispersion system.
`Based on the relative oral bioavailability data in
`dog models, a solid dispersion formulation utiliz-
`ing a mixture of polyethylene glycol (PEG) 3350
`and polysorbate 80 as the drug carrier was chosen
`for Phase I clinical trials. Attempts were also
`made to correlate the drug’s pharmacokinetic data
`with the absorption predicted by GastroPlusTM
`(SimulationsPlus Inc., Lancaster, CA), a computer
`software that simulates and models gastroin-
`testinal absorption processes.
`
`MATERIALS AND METHODS
`
`Materials
`
`LAB687 (Fig. 1) is a neutral compound (MW 468.5)
`with a log P value of 4.7. It was obtained from the
`Chemical and Analytical Development depart-
`ment of Novartis Pharmaceuticals Corp. either in
`its polymorphic form A or as a mixture of poly-
`morphic forms A and C. The melting points for
`forms A and C are 131 and 1228C, respectively.
`The mixture of polymorphic forms A and C, which
`
`Figure 1. Structure of LAB687.
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 93, NO. 5, MAY 2004
`
`has an aqueous solubility of 0.17 mg/mL at room
`temperature, was used in the present study. PEG
`3350 (Van Waters & Rogers, Glen Rock, NJ),
`polysorbate 80 (Ruger Chemical, Irvington, NJ),
`microcrystalline cellulose (Avicel PH 101, FMC,
`Philadelphia, PA), fumed silica (Cab-o-sil, Cabot,
`Boyertown, PA), and other tablet excipients were
`obtained by Novartis from commercial sources
`and released for human use. All other chemicals
`were used as received.
`
`HPLC Analysis
`
`An isocratic HPLC assay method using a reverse-
`phase column (Waters Symmetry C18, 7.5 cm 
`4.6 mm, 3.5 mm) at 308C and an UV detector set at
`a wavelength of 235 nm was used to analyze
`concentrations of the drug and its degradation
`products. The mobile phase consisted of a 50:50 v/
`v mixture of acetonitrile and water. At a flow rate
`of 1.5 mL/min, the retention time of LAB687 was
`5 min.
`
`Solubility Studies
`
`The solubility of LAB687 in various pharmaceu-
`tically relevant solvents were studied at 25  18C.
`Its solubility in water as a function of PEG 3350
`and polysorbate 80 concentrations at 37  18C was
`also studied. For those vehicles that were solid at
`room temperature, solubility was determined at
`60  58C, where the solvents were molten. Each
`solution was equilibrated for at least 24 h on a
`bottle rotator set at 40 RPM. Aliquots of solutions
`in organic solvents and those in aqueous media
`were centrifuged and then filtered through Milli-
`pore filters of 0.22 mm pore size before analyzing
`by HPLC.
`
`Solubility in Bile Salt–Lecithin Solutions
`
`Different amounts of lecithin were dissolved in
`40 mM sodium glycocholate solutions. Excess
`drug was equilibrated with the bile salt–lecithin
`solutions at 37  18C for at least 24 h prior to
`HPLC analysis.
`
`Dosage Form Development
`
`formulation approaches were
`Three different
`investigated to obtain the optimal formulations
`for Phase I clinical trials: a dry blend consisting of
`micronized drug, a solid dispersion, and an oral
`cosolvent-surfactant solution.
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1040-0002
`
`

`

`DEVELOPMENT OF DOSAGE FORMS FOR A POORLY WATER SOLUBLE DRUG I
`
`1167
`
`Compatibility Screening
`
`A drug-excipient compatibility screening study
`was carried out to identify suitable excipients for
`the dry blend formulation. Mixtures of the drug
`with lactose, microcrystalline cellulose, stearic
`acid, magnesium stearate, and fumed silica in the
`presence of 20% added water were stressed at
`50  18C, as per the method described earlier.19
`Stability of LAB687 in different vehicles including
`solid dispersion carriers, such as PEG 400, PEG
`3350, propylene glycol, poloxamer 188, and poly-
`sorbate 80, was evaluated by storing the drug
`solutions at 50  58C for 3 weeks.
`
`Physical Stability
`
`In a preliminary study, it was established by
`spiking the solid dispersion matrices with crystal-
`line drug (1%) that the formation of a small
`number of drug crystals in the product during
`stability testing may not be detectable by powder
`X-ray diffraction and differential scanning calori-
`metry (DSC). However, an optical light micro-
`scope
`(Axioskop) with cross-polarized light
`connected to a hot stage (Mettler FP82) was used
`successfully to analyze the solid dispersions for
`the presence or absence of crystalline drug.
`Samples taken from the top, middle, and bottom
`of the solid dispersion plug were spread onto a
`glass slide and inserted into the hot stage.
`Observations were made initially, as is, at room
`temperature with cross-polarized light and as the
`samples were heated up to 708C at a heating rate
`of 58C/min on the hot stage. Crystalline material
`and all thermal events were noted.
`
`Prototype Formulations
`
`The dry blend formulation consisted of 50% micro-
`nized drug (mean particle size: 4.9 mm), 49.8%
`microcrystalline cellulose, and 0.2% fumed silica.
`Target fill weight for each of the size 0 hard
`gelatin capsules was 200 mg. The solid dispersion
`formulation consisted of 4% w/w LAB687 dis-
`persed in a 3:1 mixture of PEG 3350 and poly-
`sorbate 80. For this purpose, the drug was first
`dissolved in the molten carrier (PEG 3350/poly-
`sorbate 80) at 65  58C and a 500 mg aliquot of the
`hot molten solution was then manually filled with
`a positive displacement pipette into each size 0
`hard gelatin capsule. Each capsule thus contained
`20 mg of drug and 480 mg of carrier. The capsules
`were placed into high density polyethylene
`(HDPE) bottles and placed on stability.
`
`For bioavailability testing in animal models,
`50 mg of LAB687 was administered to each dog.
`Therefore, for ease of administration to dogs, sizes
`of powder-filled and solid dispersion-filled cap-
`sules were changed, although the compositions
`remained unchanged. Each size 000 capsule con-
`tained either 100 mg of powder blend or 1250 mg of
`solid dispersion.
`For the cosolvent-surfactant solution, 20 mg of
`drug was dissolved per milliliter of the cosolvent
`system consisting of 10% propylene glycol, 45%
`Cremophor RH40, 35% corn oil glycerides, and
`10% ethanol w/w/w/v. Each size 000 hard gelatin
`capsule was filled with 1.25 mL of the resulting
`solution and sealed with hot gelatin to eliminate
`leakage from the capsule.
`
`In Vitro Dissolution Testing
`
`Dissolution profiles of the capsule formulations
`containing micronized drug, solid dispersion, and
`cosolvent-surfactant solution were determined in
`0.01 N hydrochloric acid with 1% sodium lauryl
`sulfate (SLS) as the surfactant (pH 2), according
`to the USP apparatus I, basket method (100 RPM
`at 37  0.58C). Aliquots of dissolution medium
`collected at different time intervals were filtered
`through 0.45 mm filters and analyzed by HPLC.
`Additional dissolution studies with solid disper-
`sion capsules were also investigated using only
`water as the dissolution media with paddles at
`75 RPM, and in this case, aliquots were analyzed
`before and after filtration. Selected unfiltered
`aliquots were also analyzed by photon correla-
`tion spectroscopy using laser light scattering
`(Beckman Coulter N4 plus) to measure particle
`sizes of any phase-separated material in water.
`For this purpose, samples were loaded into 1 cm2
`cuvettes and placed in a thermostatic chamber.
`The sample viscosity and the water refractive
`index were factored in the particle size measure-
`ment by the instrument software. Light scattering
`was monitored at 908 angle and at a temperature
`of 258C.
`
`Absorption Prediction by GastroPlusTM
`
`LAB687 has a relatively high permeability (Caco2
`Papp ¼ 1.0  105 cm/s) and very low solubility
`(<1 mg/mL at pH 7.4). In Caco-2 cell incubations,
`the permeability of LAB687 from apical-to-baso-
`lateral cell surface was similar to that from
`basolateral-to-apical, suggesting that carrier-
`mediated efflux is not involved. Absorption of
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 93, NO. 5, MAY 2004
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1040-0003
`
`

`

`1168
`
`DANNENFELSER ET AL.
`
`LAB687 in the capsule formulations containing
`micronized drug, solid dispersion, and cosolvent-
`surfactant solution in dogs was predicted by
`GastroPlusTM (SimulationsPlus Inc.), a computer
`software that simulates and models the gastro-
`intestinal absorption processes based on the
`Advanced Compartmental Absorption and Transit
`(ACAT) model. Sensitivity analysis was also
`conducted to evaluate the relationships among
`absorption, solubility, particle size and dissolu-
`tion profiles.
`
`Bioavailability Study
`
`LAB687 was administered orally to three beagle
`dogs at a dose of 50 mg with one size 000 capsule
`containing micronized drug with excipients,
`the PEG 3350/polysorbate 80 solid dispersion, or
`two size 000 capsules of 20 mg/mL cosolvent-
`surfactant solution. A washout period of 5 days
`was used between dosing. The dogs were fasted
`overnight before dosing and allowed free access to
`water throughout the day. Plasma samples were
`analyzed for drug assay by LC/MS/MS method.
`The relative oral bioavailability (Frel) of each
`formulation was compared to that achieved from
`the reference, cosolvent-surfactant solution, and
`appropriate statistical analysis was conducted.
`
`RESULTS AND DISCUSSION
`
`Dosage Form Development
`
`LAB687 was found to be compatible with the
`evaluated tablet and solution excipients with no
`significant degradation products seen at 508C
`with 20% water for 1 week and 508C for 3 weeks,
`respectively. LAB687 was practically insoluble
`in water (solubility 0.17 mg/mL at 25  18C) and
`highly soluble in many pharmaceutically accep-
`table organic solvents. The solubility of the drug
`in PEG 400 was greater than 60 mg/mL at 258C.
`In a 3:1 mixture of PEG 3350 and polysorbate 80
`the solubility at 608C was greater than 100 mg/g
`(>10% w/w). To minimize any potential for the
`crystallization of drug at room temperature, a
`4% w/w solid dispersion was selected for further
`evaluation. Advantages of such a PEG-polysor-
`bate based solid dispersion carrier was previously
`reported in the literature.3,18,20,21
`
`In Vitro Dissolution
`
`Dissolution studies were conducted for purposes
`of quality control as well as for understanding
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 93, NO. 5, MAY 2004
`
`the mechanism of release of drug from the dosage
`forms. Since the drug was insoluble in water
`throughout the gastrointestinal pH range, the
`dissolution profiles of prototype capsules contain-
`ing the micronized drug and the solid dispersion
`were determined in an acidic medium (0.01 N
`HCl) containing 1% SLS at 378C, and the results
`are shown in Figure 2a. Complete release of
`drug was observed in less than 45 min for the
`solid dispersion while only 80% in 45 min was
`released with the dry blend capsules. With the
`presence of a high concentration of SLS in the
`medium which is required for sink conditions,
`the dissolution method was developed for the
`batch-to-batch quality control of formulations and
`does not necessarily reflect in vivo performance
`of the dosage forms.
`For a better understanding of the release
`behavior of the drug in gastrointestinal fluid in
`absence of a high concentration of surfactant, the
`dissolution of the above-mentioned solid disper-
`sion formulation containing 25% w/w polysorbate
`80 in the matrix was studied in 250 mL of pure
`water at 378C, where the aliquots were analyzed
`before and after filtration through 0.22 mm filters,
`and the results are shown in Figure 2b. The
`medium (water) turned milky white in color,
`indicating that the solid dispersion mixed well
`with water even in the absence of added surfactant
`in the dissolution medium. Although PEG 3350
`and polysorbate 80 may not be miscible in all
`proportions,22 the amount of water used as the
`dissolution medium was sufficient to dissolve all
`of the PEG 3350 and polysorbate 80 in the formu-
`lation to create a clear solution.23 The milky color
`in the dissolution medium can thus be attributed
`to the active material being dispersed into very
`fine particles. The filtration of the dissolution
`medium revealed that approximately 20% of the
`drug passed through the 0.22 mm filter, and the
`concentration of drug in the filtrate remained
`practically the same whether a 0.22 or a 0.45 mm
`filter was used. As per the photon correlation
`spectroscopy using laser light scattering, the
`average sizes of the particulates in the unfiltered
`medium at 10, 30, and 60 min were 0.6  0.08,
`0.9  0.13, and 1.0  0.07 mm, respectively, while
`there were no measurable particles in the filtrate.
`It may be concluded from this experiment that
`under physiological conditions where approxi-
`mately 250 mL of gastrointestinal fluid might
`be present, about 20% of drug from the prototype
`solid dispersion capsule would dissolve in the
`medium and the excess would precipitate out as
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1040-0004
`
`

`

`DEVELOPMENT OF DOSAGE FORMS FOR A POORLY WATER SOLUBLE DRUG I
`
`1169
`
`Figure 2.
`(a) Dissolution profiles for 50 mg/capsule dry blend and solid dispersion
`prototype formulations in 1000 mL of 0.01 N HCl/1% SLS using USP apparatus I, basket
`method at 100 RPMs and at 378C. (^: solid dispersion, &: dry blend); (b) dispersion study
`of the solid dispersion formulation in 250 mL water at 378C before and after filtration
`through 0.22 mm filter (^: 0.22 mm, &: nonfiltered); effect of polysorbate 80 concentration
`on the dispersibility of LAB687 in 1000 mL of water at 378C—percent recovery (c) after
`filtration through 0.45 mm filter and (d) before filtration. (^: 0% polysorbate 80, &: 5%,
`~: 10%, &: 15%, *: 20%, *: 25%).
`
`fine particles. The sizes of precipitated particles
`which were initially 0.6 mm (10-min point) in-
`creased with time (30 and 60 min points).
`To further elucidate the role of polysorbate 80
`in the solid dispersion carrier, the dissolution or
`dispersion of this formulation with different ratios
`of PEG 3350/polysorbate 80 (100:0 to 75:25) was
`studied in water (1000 mL) at 378C. Figure 2c
`shows the dissolution profiles (fraction passing
`through 0.45 mm filters) of LAB687 as a function of
`polysorbate 80 concentration in the matrix, while
`Figure 2d shows the dissolution/dispersion profiles
`in water when the aliquots were not filtered.
`The fraction passing through the filter, that is,
`the drug dissolved in the dissolution medium
`(water), increased gradually with an increase in
`polysorbate 80 concentration in the solid disper-
`sion matrix, and the total amount dispersed in
`water (unfiltered concentration) also increased
`with the increase in the surfactant concentration
`in the matrix. In a separate experiment, the solid
`dispersion formulation in neat PEG 3350 (no
`surfactant present in the matrix) was dispersed
`in 250 mL of water, and the particle size of the
`precipitated material was measured. Unlike the
`
`solid dispersion containing 25% w/w polysor-
`bate 80, the average size of the precipitated drug
`particles was 6–7 mm. Thus, the presence of sur-
`factant in the solid dispersion matrix is not only
`helpful in dissolving the drug in the dissolution
`medium, it also reduces the size of precipitated
`particles and thereby facilitates their redissolu-
`tion rate as a result of higher surface area.
`
`Stability
`
`The solid dispersion formulation (3:1 PEG 3350/
`polysorbate 80) showed excellent chemical and
`physical stability with minor changes in dissolu-
`tion (Table 1). In the solid dispersion formulation,
`the drug was either in a molecularly dispersed
`state or in a phase-separated amorphous state,
`since no crystals were detected after its prepara-
`tion. If it is in the amorphous state, there is a
`potential for crystallization of the drug substance
`to occur with time. Therefore, the solid dispersion
`formulation was monitored closely for any occur-
`rence of crystallization during the accelerated
`stability testing period. Although a formal stabi-
`lity study was conducted for 3 months only,
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 93, NO. 5, MAY 2004
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1040-0005
`
`

`

`1170
`
`DANNENFELSER ET AL.
`
`Table 1. Stability of the Solid Dispersion Capsules of LAB687 in a 3:1 Mixture of PEG 3350 and Polysorbate
`80 Stored in HDPE Bottles
`
`Time
`Point
`
`Initial
`1 M
`2 M
`3 M
`
`16 M
`
`Storage Condition
`
`Compound
`Potency (mg/cap)
`
`Total
`Impurity %
`
`Microscopic
`Analysis
`
`—
`408C/75% RH
`408C/75% RH
`258C/60% RH
`408C/75% RH
`258C/60% RH
`408C/75% RH
`
`20.1
`20.4
`20.6
`20.5
`20.5
`ND
`ND
`
`0.1
`0.1
`0.1
`0.1
`0.1
`ND
`ND
`
`No crystals
`No crystals
`No crystals
`No crystals
`No crystals
`No crystals
`Crystals
`
`% Dissolved
`
`15
`
`17.9
`ND
`ND
`ND
`
`37.6
`18.4
`
`30
`
`78.9
`ND
`ND
`ND
`
`100.2
`49.8
`
`45
`
`60
`
`103.2
`ND
`ND
`ND
`
`103.1
`74.2
`
`104.5
`103.1
`101.1
`99.3
`
`103.4
`84.8
`
`ND, not determined.
`
`subsequent visual examination of retained sam-
`ples did not show any crystallization of drug in the
`matrix even after 16 months of storage at 258C/
`60% RH. This indicates that the drug appears to
`have a low crystallization potential in the solid
`dispersion formulation. Only a small decrease in
`the dissolution rate of the capsules stored at 408C/
`75% RH was observed after 16 months (Table 1).
`It is a common phenomenon for hard gelatin
`capsules to show a decrease in dissolution rates
`when stored at 408C/75% RH.24 This phenomenon
`can be the main contributor to the decreased
`dissolution seen with the solid dispersion formu-
`lation, since only a few crystals were observed in
`the samples stored at 408C/75% RH for 16 months.
`
`GastroPlusTM Simulation
`The absorption in dogs predicted by GastroPlusTM
`for the 50 mg capsule formulation containing
`micronized drug utilizing an aqueous solubility of
`1 mg/mL and the drug particle size of 5 mm was low
`(8.6%) as shown in Figure 3 and the total fraction
`of absorption in the small intestine and colon
`was estimated at 1.5 and 7.1%, respectively. The
`absorption values were reflective of the drug
`transit time in the small intestine (1.5–1.8 h)
`and in the colon (10–12 h) in dogs, indicating
`that the drug was absorbed uniformly throughout
`the entire intestinal tract. As reported earlier, the
`drug solubility in water is 0.17 mg/mL, and,
`therefore, the use of a higher value of 1 mg/mL in
`the GastroPlusTM calculation was based on the
`consideration that the solubility of LAB687 in the
`gastrointestinal fluids would be higher in pre-
`sence of bile salts, lecithin, lipolytic products,
`etc.25 For example, the solubility of LAB687 was
`shown to be five times greater in 40 mM sodium
`glycocholate (0.8 mg/mL) than in water and almost
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 93, NO. 5, MAY 2004
`
`10 times greater in the presence of 40 mM sodium
`glycocholate with 4 mM lecithin (1.9 mg/mL).
`The selection of the drug solubility value for the
`simulation could, however, be somewhat arbi-
`trary in nature since concentrations of such phy-
`siological
`solubilizing agents may vary in
`different regions of the gastrointestinal tract
`and from person to person. As shown later in the
`paper, a sensitivity analysis for drug solubility
`might present a better picture of the overall effect.
`The extent of absorption in the GastroPlusTM
`simulation was low primarily due to the poor
`solubility of the drug. A sensitivity analysis using
`the software showed that the oral absorption of
`LAB687 is very sensitive to solubility changes, as
`shown in Figure 4a. Though the actual solubility
`of the drug in the intestinal tract containing the
`mixture of the vehicle and intestinal fluid is
`unknown, the simulation suggested that different
`vehicles might have a significant impact on gastro-
`intestinal absorption of the drug. For example, if
`the solubility changes from 1 to 10 or 100 mg/mL,
`the absorption could increase from 8.6 to 50 or 95%,
`respectively. Based on the in vitro dissolution test,
`
`Figure 3. Predicted oral absorption of 50 mg LAB687
`in a dry blend with micronized drug by GastroPlusTM in
`dogs. Parameters used in the prediction include particle
`size of 5 mm and solubility value of 1 mg/mL.
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1040-0006
`
`

`

`DEVELOPMENT OF DOSAGE FORMS FOR A POORLY WATER SOLUBLE DRUG I
`
`1171
`
`Figure 4. Simulated correlation for LAB687 in dogs between (a) absorption and
`solubility using a solubility range of 1–100 mg/mL, particle size of 4.9 mm, dose of 50 mg/
`dog, high permeability; (b) absorption and particle size using a particle size range of
`1–100 mm, solubility of 1 mg/mL, dose of 50 mg/dog, high permeability; (c) absorption and
`dose using a dose range of 5–500 mg/dog, solubility of 1 mg/mL, particle size of 4.9 mm, high
`permeability; (d) absorption and permeability using a dose of 50 mg/dog, particle size of
`4.9 mm, solubility of 1 mg/mL.
`
`it was found that the kinetic solubility and possibly
`even the equilibrium solubility of the drug in
`simulated intestinal fluid from a solid dispersion
`formulation could be increased as compared to that
`of the dry blend with micronized drug. Therefore,
`it is likely that the high bioavailability of the drug
`could be increased as compared to a dry blend by
`such formulation approaches as solid dispersion
`and cosolvent-surfactant solubilization. Similarly,
`the absorption is also sensitive to particle size
`changes, Figure 4b. On the other hand, due to the
`extremely low solubility of the drug, the absorption
`is not very sensitive to dose changes with a dose
`range between 5 and 500 mg/dog, Figure 4c. As
`expected, the oral absorption is relatively in-
`sensitive to permeability since the drug is highly
`permeable, Figure 4d.
`
`Dog Bioavailability Results
`
`The individual and mean serum concentrations
`and pharmacokinetic parameters of LAB687 in
`dogs are listed in Tables 2–5.
`The administration of the cosolvent-surfactant
`solution to the dogs resulted in a low inter-animal
`variability in concentrations of LAB687 as shown
`
`in Table 2. The absorption was rapid with a tmax of
`1–2 h. The Cmax and AUC0– 48h values ranged
`between 989 and 1320 ng/mL and between 6320
`and 7540 ng h/mL, respectively. When the cosol-
`vent-surfactant solution was diluted with water,
`the particle size measured by laser light scattering
`was 125 mm, possibly representing a micellar
`system or microemulsion. The AUC0 – 48h (6960 ng
`h/mL) of this formulation was, therefore, used as
`the reference for calculating the relative bioavail-
`ability (Frel) of the other two formulations since
`the drug could not be given for an intravenous
`dose due to lack of a biocompatible intravenous
`formulation.
`The administration of solid dispersion to dogs
`also resulted in a low inter-animal variability
`in the concentrations of LAB687 (Table 3) except
`that the concentration at 12 h in Dog 2 was higher
`than that in the other dogs. The drug was rapidly
`absorbed with the time to peak concentration at
`1–2 h. The Cmax and AUC0– 48h values ranged
`between 770 and 853 ng/mL and between 5890 and
`8900 ng h/mL, respectively. The mean AUC0– 48h of
`6900 ng h/mL showed an equal relative bioavail-
`ability of LAB687 (99.1%) as compared to the
`cosolvent-surfactant solution.
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 93, NO. 5, MAY 2004
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1040-0007
`
`

`

`1172
`
`DANNENFELSER ET AL.
`
`Table 2. Serum Concentrations and Relevant Pharmacokinetic Parameters
`Following Single Oral Doses of 50 mg/dog LAB687 in Cosolvent-Surfactant Solution
`
`Concentration (ng/mL)
`
`Time (h)
`
`Dog 1
`
`Dog 2
`
`Dog 3
`
`Mean
`
`0
`0.5
`1
`2
`4
`8
`12
`24
`48
`Tmax (h)
`Cmax (ng/mL)
`AUC0– 48h (ng : h/mL)
`
`BLQa
`279
`989
`820
`240
`169
`137
`112
`32.4
`1
`989
`7010
`
`BLQ
`615
`1320
`574
`296
`231
`167
`42.0
`21.8
`1
`1320
`6320
`
`BLQ
`35.3
`602
`1170
`383
`249
`189
`78.0
`21.3
`2
`1170
`7540
`
`BLQ
`310
`970
`855
`306
`216
`164
`77.3
`25.2
`1–2b
`1160
`6960
`
`aBLQ indicates concentration below limit of quantification (<21.2 ng/mL).
`bA range was given.
`cNot applicable.
`
`SD
`
`BLQ
`291
`359
`300
`72.1
`42.0
`26.1
`35.0
`6.27
`n/ac
`166
`610
`
`In contrast to the formulations described above,
`the administration of dry blend with micronized
`drug to dogs resulted in a high inter-animal
`variability in concentrations of LAB687 as shown
`in Table 4. It was found that the concentrations
`of the parent compound in one dog (Dog 1) were
`significantly higher than those in the other dogs.
`The absorption showed a rapid onset after the
`administration with a tmax of 1 h. However, the
`Cmax and AUC0 – 48h were low with values ranging
`between 43.7 and 245 ng/mL and between 184 and
`1650 ng h/mL, respectively. The mean Frel value
`
`of the formulation showed a low relative bio-
`availability of LAB687 (9.8%) as compared to the
`solid dispersion or cosolvent-surfactant solution
`(Table 5).
`Although the absolute bioavailability of LAB687
`in these formulations could not be determined, the
`predicted oral bioavailability from the dry blend
`with micronized drug (8.6%) correlated well with
`the in vivo relative oral bioavailability (9.8%),
`suggesting that the drug in the cosolvent-surfac-
`tant solution or solid dispersion possibly provided
`almost complete bioavailability, which was tenfold
`
`Table 3. Serum Concentrations and Relevant Pharmacokinetic Parameters
`Following Single Oral Doses of 50 mg/dog LAB687 in Solid Dispersion Matrix
`
`Concentration (ng/mL)
`
`Time (h)
`
`Dog 1
`
`Dog 2
`
`Dog 3
`
`Mean
`
`0
`0.5
`1
`2
`4
`8
`12
`24
`48
`Tmax (h)
`Cmax (ng/mL)
`AUC0– 48h (ng : h/mL)
`
`BLQa
`BLQ
`430
`787
`277
`196
`133
`74.2
`33.0
`2
`787
`5910
`
`BLQ
`179
`527
`853
`292
`167
`523
`62.6
`23.6
`2
`853
`8900
`
`BLQ
`61.8
`770
`718
`365
`242
`221
`62.0
`BLQ
`1
`770
`5890d
`
`BLQ
`80.3
`576
`786
`311
`202
`292
`66.3
`18.9
`1–2b
`803
`6900
`
`SD
`
`BLQ
`90.9
`175
`67.5
`47.1
`37.8
`205
`6.88
`17.0
`n/ac
`43.8
`1730
`
`aBLQ indicates concentration below limit of quantification (<21.2 ng/mL).
`bA range was given.
`cNot applicable.
`dArea under serum concentration–time curve from 0 to last measurable time point.
`
`JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 93, NO. 5, MAY 2004
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1040-0008
`
`

`

`DEVELOPMENT OF DOSAGE FORMS FOR A POORLY WATER SOLUBLE DRUG I
`
`1173
`
`Table 4. Serum Concentrations and Relevant Pharmacokinetic Parameters
`Following Single Oral Doses of 50 mg/dog LAB687 in Dry Blend Containing
`Micronized Drug
`
`Concentration (ng/mL)
`
`Time (h)
`
`Dog 1
`
`Dog 2
`
`Dog 3
`
`Mean
`
`0
`0.5
`1
`2
`4
`8
`12
`24
`48
`Tmax (h)
`Cmax (ng/mL)
`AUC0 –48h (ng : h/mL)
`
`BLQa
`BLQ
`245
`146
`127
`48.3
`45.7
`51.9
`BLQ
`1
`245
`1650
`
`BLQ
`BLQ
`94.8
`41.4
`BLQ
`BLQ
`25.6
`BLQ
`BLQ
`1
`94.8
`184
`
`BLQ
`21.5
`43.7
`21.3
`27.4
`BLQ
`24.3
`BLQ
`BLQ
`1
`43.7
`205c
`
`BLQ
`7.20
`128
`70.0
`51.0
`16.0
`32.0
`17.3
`BLQ
`1
`128
`681
`
`SD
`
`BLQ
`12.4
`105
`67.0
`66.8
`27.9
`12.0
`30.0
`BLQ
`n/ab
`105
`842
`
`aBLQ indicates concentration below limit of quantification (<21.2 ng/mL).
`bA range was given.
`cArea under the serum concentration–time curve from 0 to last measurable time point.
`
`higher than that of the dry blend with micronized
`drug. Furthermore, the solid dispersion or cosol-
`vent-surfactant solution showed much less varia-
`bility in terms of the pharmacokinetic parameters.
`It appeared that the rate, extent, and gastrointest-
`inal location of oral absorption of the drug could be
`predicted well by GastroPlusTM a physiological
`based simulation software. The simulation pro-
`gram allowed us to incorporate the experimental
`data of the drug into the prediction (e.g., Caco-2
`permeability, solubility) along with the built-in
`physiological values (such as pH and transit time
`of each intestinal segment in dogs) to increase
`the reliability of the absorption prediction. Other
`absorption related factors including the time for
`the rate of precipitation, water volume after the
`oral dose in capsule, particle size were also
`considered in the prediction. The good Gastro-
`PlusTM prediction for LAB687 could also be factors
`of its minimal first pass metabolism and that it
`is not involved in any active transporters. Most
`importantly, the software provided a sensitivity
`analysis, that allowed users to understand the
`
`correlations between absorption and associated
`parameters, such as solubility, dose, particle size,
`permeability, and pharmacokinetic parameters.
`Thus, the software indeed provided an insight
`into the formulation development processes and
`allowed foresight to potential issues prior to for-
`mulation investigation.
`It should also be mentioned that according
`to Chiou and Buehler,26 drugs are often absorbed
`more completely in dogs than in humans, and,
`therefore, the oral absorption in dogs might not
`be a true reflection of hum

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