throbber
Pharmaceutical Research, Vol. 22, No. 3, March 2005 (© 2005)
`DOI: 10.1007/s11095-004-1871-1
`
`Research Paper
`
`Intravenous Hydrophobic Drug Delivery: A Porous Particle Formulation of
`Paclitaxel (AI-850)
`
`Julie A. Straub,1,3 Donald E. Chickering,1 Jonathan C. Lovely,1 Huimin Zhang,1 Bhavdeep Shah,1
`William R. Waud,2 and Howard Bernstein1
`
`Received September 28, 2004; accepted December 13, 2004
`
`Purpose. To develop a rapidly dissolving porous particle formulation of paclitaxel without Cremophor
`EL that is appropriate for quick intravenous administration.
`Methods. A rapidly dissolving porous particle formulation of paclitaxel (AI-850) was created using spray
`drying. AI-850 was compared to Taxol following intravenous administration in a rat pharmacokinetic
`study, a rat tissue distribution study, and a human xenograft mammary tumor (MDA-MB-435) model in
`nude mice.
`Results. The volume of distribution and clearance for paclitaxel following intravenous bolus adminis-
`tration of AI-850 were 7-fold and 4-fold greater, respectively, than following intravenous bolus admin-
`istration of Taxol. There were no significant differences between AI-850 and Taxol in tissue concen-
`trations and tissue area under the curve (AUC) for the tissues examined. Nude mice implanted with
`mammary tumors showed improved tolerance of AI-850, enabling higher administrable does of pacli-
`taxel, which resulted in improved efficacy as compared to Taxol administered at its maximum tolerated
`dose (MTD).
`Conclusions. The pharmacokinetic data indicate that paclitaxel in AI-850 has more rapid partitioning
`from the bloodstream into the tissue compartments than paclitaxel in Taxol. AI-850, administered as an
`intravenous injection, has been shown to have improved tolerance in rats and mice and improved
`efficacy in a tumor model in mice when compared to Taxol.
`
`KEY WORDS: drug delivery; hydrophobic drugs; microparticles; paclitaxel; spray drying.
`
`INTRODUCTION
`
`The poor aqueous solubility of hydrophobic drugs is a
`challenging formulation problem, particularly for intravenous
`delivery (1). It is estimated that 40% of new chemical entities
`have poor aqueous solubility (2). Poor aqueous solubility is,
`in particular, a common property of compounds identified
`using combinatorial chemistry and high-throughput screening
`(3). Thus, creating intravenous formulations of such com-
`pounds is of significant value. Paclitaxel, the active ingredient
`in the commercially available anticancer agent Taxol (pacli-
`taxel injection, Bristol-Myers Squibb Oncology, Princeton,
`NJ, USA), has low aqueous solubility (4). Taxol consists of
`paclitaxel dissolved in Cremophor EL (BASF, Ludwigshafen,
`Germany; polyoxyethylated castor oil) and ethanol. Side ef-
`fects observed with the intravenous administration of Taxol
`such as hypersensitivity reactions, nephrotoxicity, and neuro-
`toxicity have been attributed to Cremophor (5). Due to such
`reactions, patients must be premedicated with corticosteroids
`and antihistamines, and Taxol must be administered as a 1–24
`h infusion (6–7). Taxol is an important treatment of breast,
`ovarian, and non-small cell lung carcinomas (8–9), and modi-
`fied formulations of paclitaxel that do not contain Cremophor
`
`1 TAcusphere, Inc., Watertown, Massachusetts 02472, USA.
`2 Southern Research Institute, Birmingham, Alabama 35205, USA.
`3 To whom correspondence should be addressed. (e-mail: julie.
`straub@acusphere.com)
`
`are currently of significant interest (10–12). A number of
`nanoparticle- and microparticle-based formulations of pacli-
`taxel, and related formulations such as liposomes and emul-
`sions, have been reported in the literature (13–21). In these
`formulations, the paclitaxel is encapsulated within another
`material such as a hydrophobic polymer like poly (lactide
`co-glycolide) (PLGA), a protein, or lipids. These types of
`materials create a sustained-release system for paclitaxel with
`drug release occurring over hours to months. The porous par-
`ticle formulation of paclitaxel (AI-850) discussed in this paper
`was developed with the goal of creating a rapidly dissolving
`formulation that could be administered as an i.v. bolus or a
`short infusion.
`Acusphere’s hydrophobic drug delivery system (HDDS),
`consisting of sub-micrometer to micrometer size porous par-
`ticles, can be used to create rapidly dissolving formulations of
`hydrophobic drugs (22–25). The porous particles consist of
`drug microparticles and sub-micrometer particles and a wa-
`ter-soluble excipient such as a sugar or amino acid. During
`reconstitution, the water-soluble excipient dissolves, leaving a
`suspension of drug microparticles and sub-micrometer par-
`ticles that dissolve upon further dilution in the plasma. The
`porous nature of the particles facilitates wetting and rapid
`dissolution of the encapsulated drug.
`The production of the porous particles involves spray
`drying a solution containing the drug, the water-soluble ex-
`cipient, and a pore-forming agent (i.e., a volatile salt). The
`
`347
`
`0724-8741/05/0300-0347/0 © 2005 Springer Science+Business Media, Inc.
`
`AVENTIS EXHIBIT 2089
`Mylan v. Aventis, IPR2016-00712
`
`

`
`348
`
`Straub et al.
`
`pore-forming agent is volatilized along with the process sol-
`vent during the spray drying to produce a porous matrix com-
`prising drug microparticles and the excipient. The water-
`soluble excipient facilitates wetting of the drug particles dur-
`ing reconstitution, provides proper osmolality to the dosage
`form, and improves the storage stability of the dry powder.
`The spray-drying process can be performed aseptically, and
`the resulting particles are in the appropriate size range for i.v.
`administration following reconstitution, as well as appropri-
`ate for administration by other parenteral routes, oral admin-
`istration, and pulmonary administration.
`This paper discusses the application of the hydrophobic
`drug delivery system to a rapidly dissolving formulation of
`paclitaxel for intravenous administration as an i.v. bolus or a
`short infusion and which would not require premedication of
`the patients with corticosteroids and antihistamines.
`
`MATERIALS AND METHODS
`
`Production of Microparticles
`
`A paclitaxel-containing solution was prepared by dissolv-
`ing paclitaxel (6.25 mg/ml), polysorbate 80 (0.625 mg/ml), and
`polyvinylpyrrolidone C15 (0.625 mg/ml) in ethanol. An aque-
`ous solution was prepared containing the pore-forming agent
`(22), ammonium bicarbonate, at 2.5 mg/ml, and mannitol at
`25 mg/ml. The aqueous solution was added to the ethanol
`solution in a ratio of 1:4. The resulting solution was spray
`dried on a spray dryer custom-designed to operate aseptically.
`The spray dryer was made from 316 stainless steel compo-
`nents with tri-clamp connections. The system was run under
`positive pressure with sterile-filtered nitrogen gas for the dry-
`ing gas and atomization gas. A 0.22-␮m filter was installed on
`the exhaust to maintain sterility and to capture any paclitaxel
`particulates that escape the spray-dryer cyclone.
`The spray dryer used an internal mixing air-atomizing
`nozzle (the internal diameter of the fluid cap was 0.028⬙, and
`the internal diameter of the air cap was 0.060⬙) and nitrogen
`as the drying gas. Spray-drying conditions were as follows: 75
`ml/min solution flow rate, 154 L/min atomization gas rate, 120
`kg/h drying gas rate, 116°C inlet temperature, and 62°C outlet
`temperature. The resulting powder was collected, filled into
`glass vials, and stored at 2–8°C.
`
`Paclitaxel Potency and Purity Analysis
`
`For potency and purity analysis, AI-850 was dissolved in
`methanol:water:acetic acid (85:15:1) and analyzed by HPLC
`using an Agilent 1100 Series high pressure liquid chromotog-
`raphy (HPLC) (Agilent Technologies Inc., Palo Alto, CA,
`USA). Chromatographic conditions used for the analysis of
`paclitaxel used UV detection at 230 nm and a Phenomenex
`Curosil PFP column (5 ␮m, 250 × 4.6 mm) held at 35°C during
`analysis. The mobile phases used were A (acetonitrile:water,
`35:65) and B (acetonitrile:water, 75:25), with a gradient run-
`ning from 100% A to 85% A over 12 min, followed by a
`gradient running from 85% A to 0% A over the next 11.5
`min. A flow rate of 1.5 ml/min was used. Potency values were
`obtained via comparison to a reference standard obtained
`from Hauser Laboratories (Boulder, CO, USA) and are
`based on 100% potency being the theoretical loading of the
`AI-850 powder based on the composition spray dried.
`
`Differential Scanning Calorimetry (DSC)
`
`DSC analyses were carried out on a TA 2920 differential
`scanning calorimeter (TA Instruments, New Castle, DE,
`USA) using nitrogen as the purge gas. Indium metal was used
`as the calibration standard. The samples were heated on the
`DSC at a heating rate of 10°C/min to a final temperature of
`350°C.
`
`Dry Powder Particle Size Analysis
`
`Particle size analysis of AI-850 powder (prior to recon-
`stitution) was performed using a Malvern Mastersizer (Mal-
`vern Instruments Ltd., Worcester, England) fitted with a dry
`powder module at a pressure of 65 psi.
`
`Particle Size Analysis Post-Reconstitution
`
`Particle size analysis of paclitaxel particles in AI-850
`post-reconstitution was performed using a Multisizer II
`(Beckman Coulter Inc., Fullerton, CA, USA) equipped with
`a 50-␮m aperture. The electrolyte used for the analysis con-
`tained sodium chloride (9 mg/ml), monobasic sodium phos-
`phate monohydrate (0.19 mg/ml), anhydrous dibasic sodium
`phosphate (1.95 mg/ml), mannitol (55 mg/ml), polyvinylpyrol-
`lidone K15 (5.5 mg/ml), and was adjusted to pH 7.4 with 0.1
`N HCl. The electrolyte solution was presaturated with pacli-
`taxel immediately prior to use by addition of a solution of
`paclitaxel in methanol (35 mg paclitaxel/ml methanol; 4 ml to
`1 L of modified electrolyte), followed by filtration through an
`0.22-␮m cellulose acetate filter to remove excess (precipi-
`tated) paclitaxel. The electrolyte was saturated with paclitaxel
`to ensure the paclitaxel particles in AI-850 did not dissolve
`upon dilution into the electrolyte for analysis. The presence
`of mannitol and polyvinylpyrollidone K15 in the electrolyte
`was found to suppress crystallization of paclitaxel from the
`paclitaxel-saturated electrolyte. The paclitaxel-saturated elec-
`trolyte was analyzed by Coulter Multisizer analysis prior to
`use for analysis of AI-850 suspensions to ensure that no sig-
`nificant crystallization of paclitaxel had occurred in the elec-
`trolyte solution. Suspensions of AI-850 were added to the
`paclitaxel-saturated electrolyte for analysis.
`
`Microscopy
`
`For transmission electron microscopy (TEM), AI-850
`powder was embedded in a LR White Resin, which was then
`cut into thin (100–120 nm) sections on a diamond knife, and
`the sections were imaged on a Zeiss EM-10 transmission elec-
`tron microscope (Zeiss Group, Jena, Germany) at 60 kV.
`For scanning electron microscopy (SEM), the particles
`were sputter coated with platinum-palladium (80:20) and then
`imaged on a Hitachi S-4800 or a Hitachi S-2700 (Hitachi High
`Technologies America, Inc., San Jose, CA, USA).
`For scanning electron microscopy (SEM) of paclitaxel
`particles post-reconstitution, the suspension post-recon-
`stitution was filtered using an 0.22-␮m filter, and the pacli-
`taxel particles retained on the filter were rinsed with water
`and vacuum dried prior to sputter coating.
`
`In Vitro Dissolution Analysis
`
`Dissolution studies were conducted in PBS containing
`0.08% Tween 80 (T80/PBS). T80/PBS (10 ml) was added to
`
`

`
`Intravenous Hydrophobic Drug Delivery
`
`349
`
`an appropriate amount of material being tested to contain 5
`mg of paclitaxel in a 15 ml polypropylene conical tube, and
`the suspension was vortexed to create a suspension of micro-
`particles of paclitaxel. The suspension (0.25 ml) was then
`added to 250 ml of T80/PBS in a 600 ml glass beaker for
`dissolution analysis. Samples (1–2 ml) were removed and im-
`mediately filtered at each time-point. HPLC analysis was per-
`formed directly on the filtered aqueous solutions using an
`Agilent 1100 Series HPLC (Agilent Technologies Inc.). Chro-
`matographic conditions used for the analysis of dissolution of
`paclitaxel used a Nucleosil column (5 ␮m, C18, 100A, 250 ×
`4.6 mm), a mobile phase of 2 mM H3PO4/acetonitrile (2:3) at
`a flow rate of 1.5 ml/min, UV detection at 227 nm, and a run
`time of 25 min. The concentration of paclitaxel in the disso-
`lution media was 0.5 ␮g/ml. The saturation concentration ob-
`served for paclitaxel in T80/PBS was observed to be 1.5 ␮g/
`ml, and thus the dissolution assay was performed at a con-
`centration lower than the saturation concentration of
`paclitaxel in the dissolution media.
`
`Animal Care
`
`In the three animal studies described below, cage size
`and animal care conformed to the guidelines of the Guide for
`the Care and Use of Laboratory Animals, 7th Edition (Na-
`tional Research Council, National Academy Press, Washing-
`ton, DC, 1996), and the U.S. Department of Agriculture
`through the Animal Welfare Act (Public Law 99-198).
`
`Pharmacokinetic Study of Paclitaxel in Rats
`
`Male and female Sprague-Dawley rats (3/sex per group;
`animals were 8–11 weeks of age) were assigned to one of eight
`groups. Two groups received an i.v. bolus of Taxol (5 mg/kg
`or 10 mg/kg). Two groups received a 3 h i.v. infusion of Taxol
`(5 mg/kg or 10 mg/kg of paclitaxel). Four groups received an
`i.v. bolus of AI-850 (5 mg/kg, 10 mg/kg, 20 mg/kg, or 30 mg/kg
`of paclitaxel). AI-850 was reconstituted with water and di-
`luted with D5W for dosing at a paclitaxel concentration of 10
`mg/ml. Taxol was diluted with sterile saline for dosing at a
`paclitaxel concentration of 1 mg/ml.
`Serial blood samples (0.3 ml) were taken during 24 h and
`processed into plasma. For the bolus dose groups, blood
`samples were collected at 0 min (pre-dose), 5 and 30 min, and
`at 1, 2, 3, 5, 8, 12, and 24 h after dosing. For the infusion
`groups, blood samples were collected at 0 min (pre-infusion)
`and at the following time-points after the start of the infusion:
`30 min, and at 1, 2, 3, 4, 5, 8, 12, and 24 h. The plasma samples
`were analyzed for paclitaxel using a validated liquid chroma-
`tography/mass spectrometry/mass spectrometry (LC/MS/MS)
`assay at Southern Research Institute. The pharmacokinetic
`parameters were calculated assuming a two-compartment
`analysis using WinNonlin software.
`
`Tissue Distribution Study of Paclitaxel in Rats
`Single doses of either AI-850 or Taxol were administered
`intravenously via tail vein to groups of 18 male Sprague-
`Dawley rats/group. Animals were 8–9 weeks of age. The pac-
`litaxel dose was 5.7 mg/kg for both formulations. AI-850 was
`reconstituted with water and diluted into D5W to a paclitaxel
`concentration of 6.9 mg/ml for administration. Taxol Injection
`was diluted with saline to a final paclitaxel concentration of
`1.0 mg/ml for administration. Samples of plasma and selected
`
`tissues were collected from three rats/group at 0.5, 1, 2, 5, 12,
`and 24 h post-dose. The following tissues were collected and
`flash-frozen in liquid nitrogen: adrenal glands, brain, bone
`marrow, heart, kidney, liver, lung, muscle (back), small intes-
`tine (duodenum), sciatic nerve, spleen, testes, and thymus.
`Plasma and tissue samples were analyzed for concentrations
`of paclitaxel using a validated LC/MS/MS method (tissue
`samples were homogenized prior to analysis) at Southern Re-
`search Institute. Mean tissue paclitaxel concentration data
`were subjected to pharmacokinetic analyses using WinNonlin
`software. Values of AUC (area under the curve) were calcu-
`lated using the trapezoidal rule. Plasma paclitaxel concentra-
`tion data were fit to a compartmental model.
`
`In Vivo Antitumor Efficacy Study in Mice
`
`Mice were young (approximately 23 g), adult, female
`athymic NCr-Nu (14 mice/group). Mice were implanted sub-
`cutaneously with 30–40 mg fragments of the human mammary
`tumor MDA-MB-435 on day 0 (26). Treatment was scheduled
`to begin when the tumors ranged in size from 75 to 150 mg.
`Three groups were treated with AI-850 (15 mg/kg, 30 mg/kg,
`or 40 mg/kg of paclitaxel), one group was treated with the
`maximum tolerated dose (MTD) of Taxol (27) in this model
`(15 mg/kg of paclitaxel), and one control group was treated
`with D5W. AI-850 was reconstituted with 5% dextrose
`(D5W) to paclitaxel concentrations of 1.5 mg/ml, 3 mg/ml,
`and 4 mg/ml. Taxol was diluted with saline to a paclitaxel
`concentration of 1.5 mg/ml. Agents were administered via i.v.
`injection into the tail vein, once a day for 5 days. Mice were
`observed for survival, tumor size, and body weight. Tumors
`were measured by caliper in two dimensions and converted to
`tumor mass using the formula for a prolate elipsoid (l × w2/2).
`Antitumor activity was assessed by the delay in tumor
`growth of the treated groups in comparison to the vehicle-
`treated control. The values for the time required for two tu-
`mor mass doublings (four times the original size) and t-c were
`calculated. The value of t-c (days) is the difference in the
`median of times post-implant for tumors of the treated groups
`to attain an evaluation size compared to the median of the
`control group. The time required for tumor mass doubling is
`calculated based on the initial tumor weight at the beginning
`of the treatment period, and values between measurements
`are calculated by exponential extrapolation.
`
`RESULTS
`
`Analysis of Paclitaxel Within AI-850
`
`AI-850 was made via spray drying an ethanol-water so-
`lution containing paclitaxel, ammonium bicarbonate (the
`pore-forming agent), mannitol, polysorbate 80, and polyvinyl-
`pyrrolidone C15. As expected for a spray-drying process, pac-
`litaxel encapsulation efficiency was essentially 100%, with ob-
`served values of chromatographic potency of 99.8% ± 1.5%
`(n ⳱ 6 lots). The chromatographic purity of paclitaxel in
`AI-850 was comparable to the purity of the starting bulk pac-
`litaxel, indicating that the paclitaxel molecule was stable to
`the processing conditions. For example, the chromatographic
`purity of the paclitaxel for both the starting raw material and
`the lot of AI-850 used in the tissue distribution study were
`observed to be 99.5%. DSC analysis indicated that the pacli-
`taxel within AI-850 was amorphous.
`
`

`
`350
`
`Straub et al.
`
`Fig. 1. Dry powder particle size distribution of AI-850 as analyzed on
`the Malvern Mastersizer.
`
`Particle Characteristics
`
`The mean particle size for AI-850 prior to reconstitution
`was 1.53 ␮m ± 0.07 ␮m (n ⳱ 6 lots). A dry powder particle
`size distribution from a representative lot is shown in Fig. 1.
`The volume mean diameter is 1.52 ␮m, and 40% of the vol-
`ume is less than 1.0 ␮m. A scanning electron micrograph of
`AI-850 particles is shown in Fig. 2, in which the surface of the
`microparticles appears relatively smooth, and the micropar-
`ticles appear to be sphere-shaped. A transmission electron
`micrograph of cross-sections of AI-850 particles is shown in
`Fig. 3, where the bright white areas are the locations of the
`water-soluble excipients, the light gray areas are the locations
`of the paclitaxel, and the darker gray areas are the locations
`of the pores. Prior to reconstitution, the porosity of the AI-
`850 particles was predominantly internal.
`Multiple analytical methods were evaluated as options
`for quantitative analysis of the particle size distribution of the
`paclitaxel particles within a reconstituted AI-850 suspension.
`Most of these methods (e.g., laser sizing methods) require
`large dilutions to obtain particle concentrations with the op-
`erating range of the respective detector, and as a result the
`AI-850–derived paclitaxel particles dissolved before analyses
`of such dilutions could be performed. A method for quanti-
`tative analysis was developed using a Coulter Multisizer II
`
`Fig. 3. TEM of cross section of AI-850 dry powder particles.
`
`run using electrolyte saturated with paclitaxel, which prevents
`the rapid dissolution of the AI-850–derived paclitaxel par-
`ticles. The particle size distribution of the paclitaxel particles
`from a reconstituted AI-850 suspension analyzed on a Coulter
`Multisizer is shown in Fig. 4, with an observed volume mean
`particle size of 2.0 ␮m. The Coulter Multisizer method used
`an aperture that could only detect particles down to 1 ␮m.
`Thus, unlike the Malvern Mastersizer analysis of the AI-850
`microparticles, which had a lower limit of detection of 0.5 ␮m,
`the sub-micrometer particles were not detected with the
`Coulter Multisizer method, and the mean particle size value
`reported is an overestimation of the actual value. Attempts to
`perform suspension particle size analysis using a laser-based
`method on a Coulter LS230, which would have allowed for
`analysis of nano- and microparticles, using paclitaxel satu-
`rated media, were not successful due to problems attributed
`to the crystallization of paclitaxel within the paclitaxel-
`saturated media itself, affecting the LS230 analysis. An SEM
`image of paclitaxel particles isolated from a suspension post-
`reconstitution is shown in Fig. 5, which not only demonstrates
`the presence of microparticles of paclitaxel but also demon-
`strates that the microparticles themselves are internally po-
`rous, with significant surface porosity.
`Paclitaxel dissolution for the parent bulk paclitaxel lot
`and for the average of 17 lots of AI-850 is shown in Fig. 6.
`These data illustrate that the process consistently produces a
`rapidly dissolving paclitaxel product.
`
`Fig. 2. SEM of AI-850 dry powder particles.
`
`Fig. 4. Coulter Multisizer particle size distribution of reconsti-
`tuted AI-850.
`
`

`
`Intravenous Hydrophobic Drug Delivery
`
`351
`
`MB-435. Treatment began on day 14, when the median tu-
`mor size ranged from 108 to 144 mg (individual tumors rang-
`ing from 100 mg to 172 mg). The control tumors grew well,
`with a doubling time of approximately 10 days (200–400 mg).
`Agents were administered i.v. once a day for 5 days. Maxi-
`mum weight losses observed were 4% for 15 mg/kg Taxol, and
`0%, 9%, and 21% for AI-850 at 15 mg/kg, 30 mg/kg, and 40
`mg/kg, respectively. Tumor weight data over time is shown in
`Fig. 8. At the 15 mg/kg dose, the time required for two tumor
`mass doublings was 59.6 days for Taxol and 56.9 days for
`AI-850, and the value of t-c was 44.4 days for Taxol and 41.7
`days AI-850. For 30 mg/kg dose of AI-850, the time required
`for two tumor mass doublings was 66.4 days, and the value of
`
`Fig. 5. SEM of AI-850 particles post-reconstitution.
`
`Rat Pharmacokinetic Study
`
`Plasma pharmacokinetic data for rats dosed with AI-850
`and Taxol are shown in Fig. 7 and Table I. AI-850 was well
`tolerated at all doses (5 mg/kg, 10 mg/kg, 20 mg/kg, and 30
`mg/kg of paclitaxel). Taxol administered as a 3-h infusion was
`also well tolerated at both dose levels (5 mg/kg and 10 mg/kg
`of paclitaxel). However, all the rats administered an i.v. bolus
`dose of Taxol at 10 mg/kg of paclitaxel died within 2 h.
`
`Rat Tissue Distribution Study
`
`Groups of rats were given an i.v. bolus dose of AI-850
`(5.7 mg/kg of paclitaxel) or an i.v. bolus dose of Taxol (5.7
`mg/kg of paclitaxel). Both an i.v. bolus dose of AI-850 (5.7
`mg/kg of paclitaxel) and an i.v. bolus dose of Taxol (5.7 mg/kg
`of paclitaxel) were well tolerated. Data from selected time-
`points are shown in Table II, and tissue paclitaxel AUC val-
`ues are shown in Table III.
`
`In Vivo Antitumor Efficacy
`
`To assess the efficacy of AI-850 in a tumor model, mice
`(young, adult female athymic NCr-Nu) were implanted sub-
`cutaneously (s.c.) with the human mammary tumor MDA-
`
`Fig. 6. In vitro dissolution of paclitaxel from bulk paclitaxel (䊐) and
`AI-850 (䉱). For AI-850, the error bars represent the standard devia-
`tion for 17 lots.
`
`Fig. 7. Plasma paclitaxel concentrations in Sprague-Dawley rats. All
`values are means ± standard deviations for the combined data for
`male and female rats. (a) Treatment groups were Taxol Bolus at 5
`mg/kg (䉱) and 10 mg/kg (䊏). (b) Treatment groups were Taxol In-
`fusion at 5 mg/kg (䊉) and 10 mg/kg (䊏). (c) Treatment groups
`were AI-850 Bolus at 5 mg//kg (䊊), 10 mg/kg (䊏), 20 mg/kg (䉱), and
`30 mg/kg (×).
`
`

`
`352
`
`Straub et al.
`
`Table I. Pharmacokinetic Analysis of Paclitaxel Formulations
`
`PK parameter
`
`Taxol i.v.
`bolus 5
`mg/kg
`
`Taxol i.v.
`bolus 10
`mg/kg
`
`Taxol 3-h
`infusion
`5 mg/kg
`
`Taxol 3-h
`infusion
`10 mg/kg
`
`AI-850
`i.v. bolus
`5 mg/kg
`
`AI-850
`i.v. bolus
`10 mg/kg
`
`AI-850
`i.v. bolus
`20 mg/kg
`
`AI-850
`i.v. bolus
`30 mg/kg
`
`AUCa (ng ⭈ h/ml)
`NAb
`2601 ± 386
`4427 ± 994
`NAb
`0.53 ± 0.11
`0.31 ± 0.09
`T1/2␣ (h)
`NAb
`5.98 ± 1.03
`2.80 ± 0.25
`T1/2␤ (h)
`C1 (ml h−1 kg−1)
`NAb
`2249 ± 306
`1255 ± 256
`NAb
`8363 ± 2063
`2289 ± 487
`Vdss (ml/kg)
`Cmax measured (ng/ml) 7742 ± 1763 35,338 ± 15,541 1170 ± 359
`
`12,670 ± 5741
`1.04 ± 0.25
`5.96 ± 0.83
`761 ± 231
`2085 ± 617
`6725 ± 2745
`
`6524 ± 388
`2095 ± 213
`854 ± 49
`0.77 ± 0.32
`0.40 ± 0.12
`0.24 ± 0.07
`3.28 ± 1.37
`3.18 ± 0.56
`3.49 ± 0.72
`2460 ± 154
`4388 ± 465
`5309 ± 318
`17,011 ± 2458 10,847 ± 1860 5747 ± 1094
`912 ± 116
`2140 ± 320
`4980 ± 998
`
`12,434 ± 1558
`0.62 ± 0.34
`2.50 ± 0.68
`2070 ± 259
`4778 ± 690
`8162 ± 1210
`
`a AUC for the 10 mg/kg 3-h infusion of Taxol was calculated over 0 to 24 h, whereas all the other AUC values were calculated over 0 to 12 h.
`b These data are not available for the group receiving a 10 mg/kg i.v. bolus of Taxol, as the rats died shortly after dosing.
`The values shown are the mean ± SD for combined data for male and female rats (n ⳱ 6).
`
`t-c was 51.2 days. For 40 mg/kg dose of AI-850, the time
`required for two tumor mass doublings was >76 days, and the
`value of t-c was >61 days.
`
`DISCUSSION
`AI-850 is produced via spray drying a solution containing
`paclitaxel, mannitol, polyvinylpyrrolidone C15, polysorbate
`80, and ammonium bicarbonate (the pore-forming agent).
`The pore-forming agent, ammonium bicarbonate, is removed
`during processing. Although spray drying has been described
`in the literature for a paclitaxel formulation, it has been for
`the production of a poly (lactic-co-glycolic acid)–based sus-
`tained-release microsphere formulation of paclitaxel (21),
`
`whereas AI-850 is an immediate-release formulation of pacli-
`taxel.
`The level of mannitol present in AI-850 is such that,
`post-reconstitution, AI-850 is iso-osmotic. The level of poly-
`sorbate 80 present in AI-850 (10% of the paclitaxel level)
`would result in a dose of 0.47 mg/kg polysorbate based on the
`standard dose of paclitaxel (175 mg/m2; 4.7mg/kg), which is
`well below doses of polysorbate 80 in commercial intra-
`venously administered products. For example, Taxotere
`(Aventis Pharmaceuticals, Inc., Bridgewater, NJ, USA),
`which is a similar chemotherapeutic to Taxol, is formulated with
`polysorbate 80. A standard dose of docetaxol from Taxotere
`(75 mg/m2; 2 mg/kg) would contain 52 mg/kg of polysorbate 80.
`
`Table II. Rat Tissue Distribution of Paclitaxel
`
`Organ
`
`Adrenal gland
`
`Bone marrow
`
`Brain
`
`Heart
`
`Kidney
`
`Liver
`
`Lung
`
`Muscle
`
`Plasma
`
`Sciatic nerve
`
`Small intestine
`
`Spleen
`
`Testes
`
`Thymus
`
`Agent
`
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`
`Mean (±SD) paclitaxel concentration (ng/g or ng/ml)
`
`0.5 h
`
`2 h
`
`5 h
`
`12 h
`
`13,637 ± 2068
`17,455 ± 1391
`2424a
`3034a
`112 ± 18
`161 ± 18
`8897 ± 570
`11,073 ± 2175
`14,123 ± 1086
`17,650 ± 1477
`19,102 ± 3998
`22,392 ± 5630
`14,047 ± 11,160
`11,467 ± 1396
`2867 ± 411
`3449 ± 341
`564 ± 14
`4780 ± 886
`1226 ± 333
`1860 ± 579
`11,323 ± 949
`14,698 ± 3536
`11,385 ± 1644
`13,392 ± 611
`221 ± 27
`323 ± 65
`2245 ± 733
`2552 ± 138
`
`6048 ± 1439
`6317 ± 455
`2953a
`3171a
`101 ± 32
`75 ± 12
`3767 ± 1060
`4604 ± 816
`6585 ± 1664
`7820 ± 1147
`10,630 ± 5856
`10,107 ± 2160
`9743 ± 5332
`5916 ± 1023
`2330 ± 426
`3477 ± 323
`185 ± 122
`792 ± 149
`802 ± 105
`1787 ± 347
`6270 ± 3107
`8957 ± 2562
`6960 ± 826
`8757 ± 196
`285 ± 86
`243 ± 42
`2288 ± 866
`3477 ± 323
`
`3298 ± 493
`4136 ± 1031
`2510a
`2778a
`49 ± 11
`57 ± 12
`1693 ± 226
`2499 ± 636
`3113 ± 487
`4282 ± 1697
`2887 ± 549
`7701 ± 3470
`3839 ± 3344
`2895 ± 476
`1675 ± 149
`2132 ± 154
`42 ± 5
`343 ± 170
`581 ± 61
`989 ± 306
`3003 ± 709
`5008 ± 1087
`4554 ± 811
`6174 ± 1252
`229 ± 47
`377 ± 61
`2106 ± 393
`2132 ± 154
`
`1659 ± 214
`996 ± 23
`1245a
`1440a
`36 ± 9
`BLOQ
`710 ± 40
`603 ± 10
`1414 ± 239
`847 ± 25
`1757 ± 144
`2057 ± 547
`1842 ± 390
`842 ± 86
`1024 ± 87
`905 ± 143
`18 ± 4
`43 ± 5
`345 ± 33
`311 ± 33
`1567 ± 63
`1924 ± 182
`2090 ± 86
`1578 ± 217
`181 ± 24
`237 ± 45
`2560 ± 445
`905 ± 143
`
`a Sample from all animals was pooled at each time-point due to small tissue sample size.
`
`

`
`Intravenous Hydrophobic Drug Delivery
`
`353
`
`Table III. Paclitaxel AUC in Rat Tissues
`
`Organ
`
`Adrenal gland
`
`Bone marrow
`
`Brain
`
`Heart
`
`Kidney
`
`Liver
`
`Lung
`
`Muscle
`
`Plasma
`
`Sciatic nerve
`
`Small intestine
`
`Spleen
`
`Testes
`
`Thymus
`
`Agent
`
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`AI-850
`Taxol
`
`AUC0-inf (ng⭈h/ml)
`
`66,466
`64,120
`45,817
`40,060
`1146
`1167
`34,572
`40,777
`66,516
`66,359
`80,642
`107,218
`126,782
`49,870
`38,759
`37,385
`1409
`7541
`13,016
`16,647
`67,659
`85,043
`86,871
`84,820
`19,680
`22,750
`NA
`1,432,935
`
`Two key parameters for intravenously administered sus-
`pensions of drug particles are particle size and dissolution
`rate. The particle size of reconstituted AI-850 was shown to
`be appropriate for intravenous administration, with a volume
`mean suspension particle size of less than 2.0 ␮m. The TEM
`of sections of AI-850 particles and the SEM image of the
`paclitaxel particles demonstrate the porous nature of the par-
`ticles.
`Lot to lot reproducibility of rapid in vitro dissolution of
`paclitaxel in AI-850 was observed (Fig. 6). Paclitaxel in AI-
`850 dissolved in vitro significantly more rapidly than the par-
`
`Fig. 8. Mouse MDA-MB-435 xenograft mammary tumor efficacy
`data (median tumor weight for n ⳱ 14) for the 5% dextrose control
`(⽧), 15 mg/kg Taxol (䊏), 15 mg/kg AI-850 (䉱), 30 mg/kg AI-850 (×),
`and 40 mg/kg AI-850 (䊊).
`
`ent bulk drug, with 95% of the paclitaxel in AI-850 dissolved
`in 5 min.
`Following a single i.v. bolus dose of AI-850 ranging from
`5 to 30 mg/kg of paclitaxel, plasma concentrations of pacli-
`taxel increased with increasing dose (Fig. 7c). The increases in
`Cmax were disproportionately greater than the increases in
`AI-850 doses, however, the ratio of mean measured Cmax was
`never more than 2.5 for a 2-fold increase in dose (Table I).
`AUC0-12 also increased disproportionately to dose. Both
`clearance (Cl) and steady-state volume of distribution (Vdss)
`decreased with increasing dose of AI-850, suggesting satura-
`tion of the elimination and/or distribution mechanism. Values
`for initial half-life (t1/2␣) and terminal half-life (t1/2␤) were
`consistent across the AI-850 doses tested. Overall, no differ-
`ences between the sexes were observed in the plasma phar-
`macokinetics of paclitaxel following i.v. bolus administration
`of AI-850.
`For rats receiving Taxol at 5 or 10 mg/kg of paclitaxel by
`either i.v. bolus or 3-h infusion, plasma concentrations of pac-
`litaxel increased more than proportionately with increasing
`dose (Fig. 7a,b). Doubling the i.v. bolus dose of Taxol from 5
`to 10 mg/kg of paclitaxel resulted in a 4.5-fold increase in
`mean measured Cmax at 5 min post-dose (Table I). Likewise,
`at the end of the 3-h infusion period in the 5 mg/kg and the 10
`mg/kg paclitaxel groups, plasma concentrations of paclitaxel
`were 6-fold higher for animals receiving Taxol at 10 mg/kg of
`paclitaxel than for animals receiving Taxol at 5 mg/kg of pac-
`litaxel. The Cl and Vdss of paclitaxel were lower in the 10
`mg/kg Taxol infusion group as compared to the 5 mg/kg Taxol
`infusion group. Values for terminal half-life (t1/2␤) were
`longer when Taxol was administered as a 3-h infusion than
`when administered by i.v. bolus. This difference may be due
`to either true differences in pharmacokinetics with i.v. Taxol
`bolus vs. Taxol infusion dosing or an inability to determine a
`true terminal half-life within the limitations of the sampling
`times and the assay sensitivity. Overall, no differences be-
`tween the sexes were observed in the plasma pharmacokinet-
`ics of paclitaxel following i.v. bolus or i.v. infusion adminis-
`tration of Taxol.
`A comparison of the pharmacokinetic results for AI-850
`and Taxol administered as i.v. bolus doses (Table I) demon-
`strates that at the 5 mg/kg dose, the Cl and Vdss of paclitaxel
`were 4- and 7-fold higher in those animals receiving AI-850 as
`compared to those receiving Taxol. Both t1/2␣ and t1/2␤ were
`similar across all i.v. bolus dose groups of AI-850 and Taxol.
`The paclitaxel Cmax achieved following i.v. bolus administra-
`tion of Taxol 5 mg/kg was 8-fold higher than that achieved
`with i.v. bolus administration of the same dose of AI-850. The
`data indicate that paclitaxel in AI-850 partitions more rapidly
`from the bloodstream into the tissue compartments than the
`paclitaxel in Taxol.
`Pharmacokinetic studies of Taxol in animals (28) and i

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