throbber
Int. J. Cancer: 85, 131–141 (2000)
`娀 2000 Wiley-Liss, Inc.
`
`Publication of the International Union Against Cancer
`Publication de l’Union Internationale Contre le Cancer
`
`INCREASED INTRACELLULAR DRUG ACCUMULATION AND COMPLETE
`CHEMOSENSITIZATION ACHIEVED IN MULTIDRUG-RESISTANT SOLID
`TUMORS BY CO-ADMINISTERING VALSPODAR (PSC 833)
`WITH STERICALLY STABILIZED LIPOSOMAL DOXORUBICIN
`
`Rajesh KRISHNA1,2, Maryse ST-LOUIS3 and Lawrence D. MAYER1,2*
`1Department of Advanced Therapeutics, BC Cancer Agency, Vancouver, Canada
`2Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
`3Biotechnology Laboratory, University of British Columbia, Vancouver, Canada
`
`We have previously demonstrated that liposome encapsu-
`lation of doxorubicin (DOX) can alleviate adverse interac-
`tions with non-encapsulated DOX and the cyclosporine multi-
`drug-resistant (MDR) modulator Valspodar. We have now
`investigated the behavior of different liposomal DOX formu-
`lations in MDA435LCC6/MDR-1 human breast cancer solid
`tumor xenograft models to identify liposome characteristics
`associated with enhanced therapeutic activity and the mecha-
`nism whereby increased chemosensitization is achieved. Tox-
`icity studies incorporating conventional phosphatidylcholine
`(PC)/cholesterol (chol) and sterically stabilized (polyethylene
`glycol 2000 [PEG]-containing) formulations of DOX indi-
`cated that whereas PC/Chol DOX was approximately 3-fold
`more toxic in the presence of Valspodar, PEG containing
`distearoylglycerophosphocholine (DSPC)/Chol DOX was
`minimally affected.
`In mice bearing MDR tumors, co-
`administration of Valspodar and egg phosphocholine (EPC)/
`Chol DOX resulted in modest MDR modulation and efficacy,
`whereas the sterically stabilized formulation induced reduc-
`tions in tumor growth equivalent to that achieved for drug-
`sensitive tumors treated with non-encapsulated DOX. Phar-
`macokinetic studies revealed a 2.5-fold increase in plasma
`DOX area under the curve (AUC) upon co-administration of
`Valspodar with EPC/Chol DOX whereas no such alterations
`were observed with the sterically stabilized liposomes. Com-
`pared to non-encapsulated DOX combined with Valspodar,
`improvements in efficacy and toxicity correlated with the
`extent to which liposomal DOX formulations were able to
`circumvent pharmacokinetic interactions. Confocal micros-
`copy demonstrated that Valspodar increased cell-associated
`DOX which correlated with the level of anti-tumor efficacy.
`Int. J. Cancer 85:131–141, 2000.
`娀 2000 Wiley-Liss, Inc.
`
`The development of second generation multidrug-resistant (MDR)
`reversing agents alleviated many of the problems caused by earlier
`PGP blockers which were pharmacological agents with their own
`inherent toxicities. However, co-administration of conventional
`anticancer drugs with many of these newer MDR modulators has
`been shown to elicit drug-modulator interactions by virtue of PGP
`blockade in normal tissues such as liver, kidney, intestine and brain
`(Keller et al., 1992a; Gatmaitan and Arias, 1993). It may not be
`surprising then that clearance of several anticancer drugs is
`inhibited by MDR modulators such as cyclosporine (CsA) (Speeg
`et al., 1992), verapamil (Nooter et al., 1987), Valspodar (Speeg and
`Maldonado, 1994, previously named PSC 833) and GW918 (Booth
`et al., 1998).
`The effects of MDR modulators on drug transport proteins that
`cause alterations in anticancer drug excretion often lead to in-
`creased anticancer drug exposure of healthy tissues and have
`necessitated dose reduction in many preclinical (Krishna and
`Mayer, 1997; Keller et al., 1992a; Nooter et al., 1987) and clinical
`(Boote et al., 1996; Sarris et al., 1996) studies. Although doses can
`be adjusted to equal levels of toxicity, it is unclear how such
`pharmacokinetic (PK) changes may impact therapeutic activity.
`These interactions have been postulated to play a role in limiting
`the therapeutic outcome in some patients (Wishart et al., 1994;
`Miller et al., 1994). While changes in anticancer drug dose and/or
`
`schedule may be able to address toxicity or efficacy alterations
`brought about by MDR modulators,
`this clearly represents a
`significant complication in applying PGP blockade strategies to
`cancer chemotherapy. This is due to the fact that most chemother-
`apy regimens utilize drug combinations, of which more than one
`are often PGP substrates. Consequently, an ability to avoid such
`anticancer drug clearance alterations may be considered to be a
`significant advantage in MDR modulation strategies.
`We earlier reported that liposome encapsulation of doxorubicin
`(DOX) can reduce non-encapsulated drug-Valspodar interactions,
`resulting in improved growth suppression of MDR murine solid
`tumors (Krishna and Mayer, 1997). The liposomal formulation
`used in these studies was composed of 120 nm diameter distearoyl-
`glycerophosphocholine (DSPC)/cholesterol (Chol) (55:45 molar
`ratio) that retains DOX for extended periods of time (Mayer et al.,
`1989). The enhanced antitumor activity observed in this study
`appeared to be a consequence of increased protection from
`Valspodar-mediated PK changes and toxicity exacerbation. How-
`ever, the mechanisms by which these effects were achieved are not
`fully understood, particularly since significant amounts of DOX are
`delivered to the liver by liposomes without notable toxicological
`consequences. In addition to alleviation of PK alterations, the
`increased delivery of DOX to MDR solid tumors using liposome
`delivery systems was associated with increased anti-tumor activity
`when co-administered with Valspodar compared with non-
`encapsulated drugs. These studies were unable, however,
`to
`distinguish the degree of DOX bioavailability (liposome entrapped
`vs. released drug) in the solid tumor. Therefore, the relative roles of
`PGP blockade and tumor drug levels remain unresolved.
`In order to address these questions, we compared here the
`toxicity, efficacy, pharmacokinetics, cellular distribution properties
`of egg phosphocholine (EPC)/Chol DOX (a system where over
`50% of the drug is released in the first hour) and a sterically
`stabilized PEG2000 (PEG) distearoylphosphoethanolamine (DSPE)/
`DSPC/Chol DOX formulation combined with the MDR modulator,
`Valspodar. The latter system was chosen on the basis of reports that
`incorporation of 5 mol% PEG-polymerized lipid in 100 nm
`DSPC/Chol vesicles results in increased circulation longevity,
`reduced liver uptake and increased tumor delivery (Papahadjopou-
`los et al., 1991). Therefore, the effects of altering the drug release,
`liver uptake and tumor accumulation properties of liposomal DOX
`on toxicological and therapeutic activity were compared to reveal
`the processes underlying the improvements in toxicity and efficacy
`achieved with liposomal systems in a human breast carcinoma
`
`Rajesh Krishna’s present address is: Department of Metabolism and
`Pharmacokinetics, Pharmaceutical Research Institute, Bristol-Myers Squibb
`Co, P.O. Box 4000, Princeton, NJ 08543, USA.
`
`*Correspondence to: Department of Advanced Therapeutics, BC Cancer
`Agency, 600 West 10th Avenue, Vancouver, BC, V5Z-4E6, Canada.
`Fax: ⫹1 604 877-6011.
`
`Received 10 May 1999; Revised 12 July 1999
`
`AMN1077
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`132
`
`KRISHNA ET AL.
`
`MDR solid tumor xenograft model. Furthermore, these formula-
`tions reflect the 2 liposomal DOX products that are either approved
`(sterically stabilized) or pending approval (EPC/Chol) for wide-
`spread clinical use. Consequently, determining their pharmacologi-
`cal properties in the presence of MDR modulators would be of
`considerable clinical interest.
`
`Material
`
`MATERIAL AND METHODS
`
`DOX hydrochloride for injection (U.S.P.) was purchased from
`David Bull (Vaudreil, Canada) and its purity affirmed by high-
`performance liquid chromatography (HPLC; see below). Valspodar
`was a generous gift from Novartis (Dorval, Canada) and its purity
`affirmed by LC/MS-MS (Varian LC-MS-MS system; Fisons,
`Altrincham, UK). DOX metabolite standards were generous gifts
`from Pharmacia Carlo Erba (Milan, Italy). PEG2000-DSPE (⬎99%
`purity), EPC (⬎99% purity) and DSPC (⬎99% purity) were
`obtained from Northern Lipids (Vancouver, Canada) and Chol from
`Sigma (St. Louis, MO). Cholesteryl hexadecyl ether (3H), a
`non-exchangeable, non-metabolizable lipid marker was purchased
`from Amersham (Oakville, Canada). HPLC grade solvents were
`obtained from BDH (Toronto, Canada) and used without further
`purification. Female BDF1 mice were obtained from Charles River
`(St. Constant, Canada). Female SCID/RAG2 mice were bred
`in-house at the BC Cancer Agency animal facility. The MDA435/
`LCC6 and its transfected MDR-1 line were generously provided by
`Dr. R. Clarke (Georgetown University, Washington, DC). These
`cells were maintained in tissue culture in Dulbeco’s modified Eagle
`medium (StemCell Technologies, Vancouver, Canada).
`
`Liposome and drug preparation
`Liposomes composed of EPC/Chol (55:45), PEG2000-DSPE/
`DSPC/Chol (5:50:45) and DSPC/Chol (55:45; mol:mol) were
`prepared by initially dissolving the lipid mixtures in chloroform
`(100 mg lipid per milliliter) and hydrating the dried lipid film in a
`300-mM citric acid, pH 4.00, buffer. The resulting multilamellar
`vesicles (MLVs) were subjected to 5 freeze-thaw cycles followed
`by a 10-cycle extrusion through 2 stacked 100-nm polycarbonate
`filters (Nuclepore, Pleasanton, CA) using a Lipex Extruder (Lipex
`Biomembranes, Vancouver, Canada). 3H-Cholesterylhexadecyl ether
`was used as a non-exchangeable, non-metabolizable lipid marker
`(Derksen et al., 1987). The resulting large unilammelar vesicles
`exhibited a mean diameter between 110–120 nm as determined
`using a Nicomp 270 submicron particle sizer (Particle Sizing
`Systems, Santa Barbara, CA).
`DOX was encapsulated in the liposomes using the transmem-
`brane pH gradient loading procedure (interior acidic) employing
`sodium carbonate as the alkalinizing agent and a drug-to-lipid
`weight ratio of 0.2:1.0 (Mayer et al., 1989). Liposomal DOX
`preparations were diluted with saline as necessary prior to in vivo
`administration. Valspodar (for animal studies) was dissolved in a
`10:1 mixture of ethanol (95%):Tween 80 and administered in a
`corn oil vehicle by oral gavage of a 200-µl volume (Keller et al.,
`1992b). Non-encapsulated DOX was administered in sterile saline.
`
`Toxicity evaluation studies
`Toxicity of the indicated DOX and Valspodar dose regimens was
`evaluated in dose range-finding studies using normal (non-tumor-
`bearing) female BDF1 mice. Toxic dose range-finding studies in
`tumor-free female mice were performed using 3 mice per group
`with appropriate group repetitions as described below. Briefly,
`mice were administered increasing (each dose in different groups)
`doses of i.v. DOX (free or liposomal) via the tail vein and oral
`Valspodar at a fixed dose of 100 mg/kg (4 hr before DOX) on days
`1, 5 and 9. Liposomal DOX doses were changed by increments of 5
`mg/kg on the day 1, 5, 9 injection schedule. DOX dose escalation
`was stopped when weight loss exceeded 30% or toxicity-related
`mortality was observed. Survival and the percent change in body
`weight was monitored over a 21-day period. Animals were
`
`monitored for other toxicity signs such as scruffy coat, dehydration,
`lethargy, ataxia or labored breathing. Animals which demonstrated
`significant physical manifestations of distress/toxicity or exhibited
`a body weight loss in excess of 30% were terminated. At the end of
`the 21-day study period, mice were terminated by carbon dioxide
`asphyxiation. Necropsies were performed to identify abnormalities
`in the major organs. The dose at which the body weight loss (group
`mean value) was ⱕ15% and all mice survived for the duration of
`study was established as the maximum tolerated dose (MTD). A
`deviation in MTD of ⫾1.5% was allowed in cases where body
`weight loss between 15–16.5% lasted only 1 day and where 100%
`recovery of body weight loss was observed, with 100% survival.
`The results were collated from at least 2 independent experiments.
`Select groups were repeated as a quality control measure to ensure
`reproducibility.
`
`In vivo anti-tumor activity
`MDA435LCC6 cells were cultured in DMEM and passaged at
`least 3 times in medium. A cell aliquot containing 5 ⫻ 106 cells in
`0.5 ml HBSS was injected i.p. into 2 female SCID/RAG2 mice.
`After 20–25 days, ascites (cells) were removed from the mouse via
`the peritoneal wall using a 20g needle and placed in sterile 15-ml
`conical tubes containing 5 ml of HBSS without Ca and Mg salts.
`The cell suspension was then centrifuged at 1000 g for 5 min and
`the supernatant discarded. Using a 27g needle fitted on a 1-ml
`syringe, 50 µl of the cell suspension was injected into 2 mammary
`fat pads on each mouse (2 ⫻ 106 cells per pad). A period of 18–21
`days was needed for the tumors to become palpable and measurable
`for drug treatments to begin. The MDR MDA435LCC6-MDR-1
`cells were maintained and passaged identically to the WT cells.
`These MDR cells have had the MDR-1 cDNA introduced into the
`MDA435LCC6 cells (Leonessa et al., 1996). Cell passaging and
`inoculation were performed identical to the procedures used in the
`WT sensitive cell line.
`Tumor growth suppression experiments were conducted in
`SCID/RAG2 mice bearing orthotopic human breast carcinoma
`MDA435LCC6 (multidrug resistant, MDR-1 and sensitive, WT)
`solid tumors. After approximately 3 weeks, when the tumors
`(n ⫽ 8 per group) were established, treatment was initiated with
`dosage regimens incorporating i.v. non-encapsulated or liposomal
`DOX with or without p.o. Valspodar (given 4 hr before DOX) on
`days 1, 5 and 9. Experiments were repeated to ensure reproducibil-
`ity of tumor growth inhibition properties. Caliper measurements of
`the tumors were performed daily, and the tumor weights calculated
`according to the formula (Krishna and Mayer, 1997):
`
`Tumor weight (g) ⫽
`
`2
`
`length (cm) ⫻ [width (cm)]2
`
`This conversion formula was verified by comparing the calculation
`derived tumor weights to excised and weighed tumors. Animal
`weights and mortality were monitored daily. Animals bearing
`ulcerated tumors or where tumor weights exceeded 10% of the
`animals’ body weight were terminated. The weights of the bilateral
`tumors were averaged for each mouse and mean tumor weights for
`each treatment group ⫾ standard error of the mean were calculated.
`Statistical tests for these longitudinal data were performed using
`repeat measures ANOVA employing Statistica for Windows 4.0
`(StatSoft, Tulsa, OK) and statistical significance was set at p ⬍
`0.05.
`
`Pharmacokinetics and tissue distribution
`Female MDA435LCC6/MDR-1 solid tumor-bearing SCID/
`RAG2 mice received i.v. via the tail vein a single bolus of
`liposomal doxorubicin (3H EPC/Chol or PEG2000-DSPE/DSPC/
`Chol 0.2:1.0). Valspodar (100 mg/kg) was administered p.o. (in 0.2
`ml) and doxorubicin administered 4 hr later. After DOX dosing,
`groups of 3 mice per time point were anesthetized with 100 µl i.p.
`of ketamine/xylazine at 30 min, 1, 2, 4, 16, 24, 48 and 72 hr. Blood
`was collected by cardiac puncture and placed into EDTA-coated
`
`AMN1077
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`COMPLETE REVERSAL OF MDR IN SOLID TUMORS
`
`133
`
`microtainer tubes. Terminal blood and selected tissues were
`collected from all animals and were processed to determine lipid
`(using radioactivity) and DOX concentrations (using HPLC). After
`blood collection, the kidneys, livers and tumors were removed
`from each animal. Tissues were rinsed in PBS, pat dried on
`absorbent paper and weighed in pre-weighed 16 ⫻ 100-mm tubes.
`Samples were stored in ⫺20°C pending analysis. A 10–30%
`homogenate in distilled water was prepared using a Polytron
`homogenizer (Kinematica, Littau, Switzerland). A 0.2-ml aliquot of
`the homogenate was digested with a tissue solubilizer, decolorized
`with peroxide and analyzed for the lipid label by scintillation
`counting. The specimens obtained from the control animals were
`used as background samples.
`DOX and its metabolites in plasma and tissue extracts were
`determined using HPLC. The HPLC assay of Andersen et al.
`(1993) was used to analyze DOX and its metabolites with minor
`modification. Briefly, sample extraction with acetonitrile was
`followed by isocratic elution from a Nova-Pak C18 3.9 ⫻ 150 mm
`(Millipore, Bedford, MA) analytical reverse phase column and
`quantified by endogenous fluorescence (emission wavelength of
`515 nm). The mobile phase consisted of a 16-mM ammonium
`formate buffer (pH 3.5)/acetone/isopropanol mixture (75:20:5)
`delivered at a rate of 1.0 ml/min. The column was maintained at
`40°C. A NEC (Boxborough, MA) Powermate SX Plus Computer
`and a Systems Interface Module (Waters, Milford, MA) were used
`for data handling.
`Using this system, the retention times of DOXol, DOX, DOXone
`and 7-deoxyDOXone were 3.6, 5.8, 7.5 and 12.6 min, respectively.
`Recoveries, using acetonitrile as the extraction solvent, from
`plasma over a concentration range of 0.05–10 µg/ml of DOX,
`DOXone, 7-deoxyDOXone and DOXol, were between 80–110%.
`To protect DOX and its metabolites from photodegradation, all
`procedures were shielded from direct exposure to light. In addition,
`DOX was found to be stable in the mobile-phase solvent mixture
`for at least 96 hr (40°C), on the HPLC autosampler tray for at least
`96 hr, in reconstituted form at 4°C for at least 10 days and for at
`least 4 freeze-thaw cycles.
`The plasma data were modeled using WinNONLIN Version 1.5
`PK software (Pharsight, Mountain View, CA) to calculate area
`under the curve (AUC), half-life (T1/2) and plasma clearance (CLp)
`according to standard equations (Gibaldi and Perrier, 1982). The
`trapezoidal rule was used to calculate the AUCs in tissue concentra-
`tion-time profiles employing a computer software AUC (program
`provided by Dr. W. Riggs, Faculty of Pharmaceutical Sciences,
`University of British Columbia). Tissue DOX levels were corrected
`for blood volume to account for material residing in the vasculature
`of the tissues, using previously published values (Bally et al.,
`1993).
`Select samples were assayed for free and liposome-associated
`DOX using Microcon-30 filters (Amicon, Oakville, Canada) using
`the equilibrium filtration method of Mayer and St. Onge (1995).
`Separation of free from liposomal and protein-bound drug was
`performed using Microcon-30 (0.5-ml capacity, Amicon) ultrafiltra-
`tion devices with a m.w. cut-off of 30 kDa. Microcon-30 samples
`were centrifuged at 4°C, 8,000 g for 20 min in a microcentrifuge
`(IEC Micromax Centrifuge; International Equipment, Needham
`Heights, MA). The ultrafiltrate was processed for DOX by HPLC
`(see above).
`
`Confocal microscopy and imaging studies
`For confocal
`imaging studies, SCID/RAG2 mice bearing
`MDA435LCC6/MDR-1 tumors were treated with non-encapsu-
`lated DOX, EPC/Chol DOX or PEG-DSPE/DSPC/Chol DOX (5
`mg/kg) in the presence and absence of Valspodar 100 mg/kg (4 hr
`before DOX). At the indicated times following DOX administra-
`tion, tissues were aseptically dissected, bathed in PBS and imaged
`fresh. Before imaging, thin pieces of tumors were placed on
`concave slides and observed under a 60⫻ oil immersion lens.
`These were then viewed under the confocal microscope to deter-
`
`mine DOX distribution characteristics. As controls, known amounts
`of non-encapsulated DOX or PEG-DSPE/DSPC/Chol DOX lipo-
`somes were infused into freshly isolated muscle tissues and viewed
`for DOX fluorescence.
`Confocal images were collected on an Optiphot 2 research
`microscope (Nikon, Tokyo, Japan) attached to a confocal laser
`scanning microscope (MRC-600, BioRad, Hercules, CA) using
`COMOS software (BioRad). The laser line on the krypton/argon
`laser was 488 nm. Filterblock BHS was used to detect DOX (488
`nm excitation, 515 nm emission). The numerical aperture was 0.75
`on the ⫻20 air objective and 1.2 on the ⫻60 oil objective. The
`images were captured such that the xyz dimensions were 0.4 µm
`cubed (⫻20) and 0.2 µm pixel (⫻60). NIH Image version 1.61 was
`used for image analysis, and all images were based on maximum
`intensity projection. Projections made in the NIH Image were
`saved in TIFF format, then imported to Adobe Photoshop version
`4.0 where the different fluorophore images were assigned to
`individual RGB channels and subsequently merged to provide the
`final image of the single or multiple sections.
`
`Toxicity
`
`RESULTS
`
`In order to identify the MTD for the day 1, 5 and 9 dosage
`regimen used in therapeutic experiments as well as to investigate
`the mechanisms of Valspodar-mediated increases in DOX toxicity,
`21-day dose range-finding toxicity studies were conducted with
`EPC/Chol, DSPC/Chol and PEG-DSPE/DSPC/Chol
`liposomal
`formulations of DOX as well as non-encapsulated drug in the
`presence and absence of p.o. Valspodar in non-tumor-bearing
`healthy mice. The results of this study are shown in Table I, where
`the body weight loss is presented, along with the MTDs (as defined
`in Material and Methods). The 3 liposomal DOX formulations
`yielded variable toxicity characteristics that depended on their
`respective abilities to retain DOX.
`In the absence of Valspodar, EPC/Chol DOX exhibited an MTD
`of 15 mg/kg, where the body weight loss was 1.7% with 100%
`survival (Table I). For the 2 saturated lipid formulations, DSPC/
`Chol and PEG-DSPE/DSPC/Chol DOX, the MTD was identical at
`25 mg/kg, representing a 1.7-fold increase in MTD compared with
`the more leaky EPC/Chol formulation. When EPC/Chol DOX was
`
`TABLE I – TOXICITY AS A FUNCTION OF DOX DOSE FOR NON-ENCAPSULATED
`AND LIPOSOMAL FORMULATIONS1
`
`Group
`
`Non-encapsulated
`DOX
`
`MTD
`
`EPC/Chol
`DOX
`
`MTD
`
`DSPC/Chol
`DOX
`
`MTD
`
`PEG-DSPE/DSPC/Chol
`DOX
`
`MTD
`
`Dose
`(mg/kg)
`
`2.5
`5
`7.5
`10
`
`5
`10
`15
`20
`
`15
`20
`25
`
`15
`20
`25
`30
`
`Day 10 weight loss (% survival)
`
`⫺PSC 833
`
`⫹PSC 833
`
`0 (100)
`3.5 (100)
`10.7 (100)
`29.2 (0)
`7.5 mg/kg
`
`2.8 (100)
`1.7 (100)
`20.4 (0)
`15 mg/kg
`
`10.8 (100)
`3.1 (100)
`16.0 (100)
`25 mg/kg
`
`7.3 (100)
`6.5 (100)
`9.3 (100)
`4.4 (67)2
`25 mg/kg
`
`10.2 (100)
`18.5 (0)
`28.2 (0)
`
`2.5 mg/kg
`
`0.9 (100)
`23.1 (0)
`
`24.4 (0)
`5 mg/kg
`
`12.9 (100)
`15.0 (100)
`18.5 (67)
`20 mg/kg
`
`9.8 (100)
`14.8 (100)
`14.8 (100)
`25.3 (0)
`25 mg/kg
`
`1Data are group mean values (n ⫽ 3 mice per group treated i.v. on
`days 1, 5 and 9).–2Body weight loss nadir is on day 19 (⫺10.2%) with
`67% survival.
`
`AMN1077
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`134
`
`KRISHNA ET AL.
`
`combined with Valspodar, significant toxicity resulted, necessitat-
`ing a dose reduction by 3-fold, to 5 mg/kg. This is comparable to
`the 3-fold decrease in MTD caused by Valspodar for non-
`encapsulated drug administration (Table I). In contrast, Valspodar
`necessitated a small 1.2-fold decrease in DOX encapsulated in
`DSPC/Chol liposomes and no dose reduction was required for the
`PEG-containing sterically stabilized liposomes (Table I).
`
`Effıcacy
`The anti-tumor activity of the 3 types of liposomal DOX
`formulations was evaluated in vivo in the absence and presence of
`Valspodar using the MDA435LCC6 and PGP-overexpressing
`
`MDA435LCC6/MDR-1 human breast carcinoma xenograft solid
`tumor models. Figure 1 presents the tumor growth curves for mice
`treated with non-encapsulated drug (a), EPC/Chol DOX (b),
`DSPC/Chol DOX (c) and sterically stabilized PEG-DSPE/DSPC/
`Chol DOX (d ) in the presence and absence of Valspodar.
`When MDA435LCC6 cells (sensitive, WT or resistant, MDR)
`are inoculated in the mammary fat pads of SCID/RAG2 mice, solid
`tumors readily establish (tumor take rates ⬎95%). Figure 1 shows
`that both MDR and WT-untreated controls exhibit comparable
`tumor growth rates, with the exponential growth phase occurring
`between days 5 and 14 (slope of linear regression line ⫽ 0.067 for
`
`FIGURE 1 – Anti-tumor efficacy of free (a), EPC/Chol DOX (b), DSPC/Chol (c) and PEG-DSPE/DSPC/Chol (d) against MDA435LCC6 WT or
`MDR1 human xenograft solid tumors in the absence and presence of co-administered Valspodar. MDA435LCC6 tumors were grown on mammary
`fat pads of female SCID/RAG2 mice. Oral Valspodar (100 mg/kg) and i.v. DOX treatments were initiated once tumors were established (20–100
`mg) and were given on days 1, 5 and 9 at the indicated doses of free and liposomal DOX. Valspodar was administered 4 hr prior to DOX injection.
`Data are expressed as mean ⫾ standard error of the mean. For legends, see individual panels.
`
`AMN1077
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`COMPLETE REVERSAL OF MDR IN SOLID TUMORS
`
`135
`
`WT vs. 0.056 for MDR tumors). Both groups were terminated on
`day 18, when tumor weights reached 0.84 ⫾ 0.05 and 0.76 ⫾ 0.05 g
`for the WT and MDR tumors, respectively. Treatment of WT
`tumors with non-encapsulated DOX at 7.5 mg/kg on days 1, 5 and 9
`(Fig. 1a) resulted in significant tumor growth suppression until day
`20, when tumors weighed 0.088 ⫾ 0.01 g. These tumors eventually
`grew to 0.68 ⫾ 0.09 g by day 40. However, administration of
`non-encapsulated DOX at 7.5 mg/kg on days 1, 5 and 9 in mice
`bearing MDR tumors did not cause any tumor growth suppression
`(Fig. 1a). When MDR tumor-bearing mice were treated with the
`MTD of non-encapsulated DOX combined with Valspodar (DOX
`dose of 3 mg/kg), there was partial tumor growth inhibition until
`day 11. After this time,
`tumor growth rates were similar to
`untreated MDR controls until day 18 when mice were terminated
`(Fig. 1a). Growth of MDR tumors treated with non-encapsulated
`DOX plus Valspodar was significantly different from both un-
`treated MDR tumors as well as MDR tumors treated with
`non-encapsulated drug alone between days 7 and 12 post-DOX
`administration. However, in comparison to WT tumors treated with
`non-encapsulated DOX 7.5 mg/kg, the MDR tumor growth inhibi-
`tion caused by non-encapsulated drug and Valspodar was transient
`(Fig. 1a).
`When MDR tumors were treated with liposomal DOX formula-
`tions in the presence and absence of Valspodar (Fig. 1b–d ), varying
`degrees of tumor growth suppression were observed. As seen in
`Figure 1b, EPC/Chol DOX alone (5 mg/kg) was unable to induce
`substantial inhibition of MDR tumor growth, with tumors weighing
`0.86 ⫾ 0.2 g on day 18. In the presence of Valspodar, however,
`EPC/Chol DOX at 3 mg/kg treatment closely resembled EPC/Chol
`DOX alone until day 12, after which tumor growth was decreased
`between days 12 and 20 (Fig. 1b). This indicated modest delayed
`anti-tumor activity. This MDR modulation caused by EPC/Chol
`DOX and Valspodar was significantly better than that caused by
`non-encapsulated DOX and Valspodar ( p ⬍ 0.05).
`Figure 1c illustrates the tumor growth inhibition of DSPC/Chol
`liposomal DOX in the presence and absence of Valspodar. In the
`absence of Valspodar, DSPC/Chol liposomal DOX caused a modest
`reduction in tumor growth. The tumor growth inhibition caused by
`DSPC/Chol DOX was significantly different from untreated con-
`trols until day 12, after which the tumor growth rate increased.
`Tumor weight for MDR tumors treated with DSPC/Chol DOX
`alone was 0.51 ⫾ 0.1 g on day 20. In comparison, Valspodar caused
`a significant increase in DSPC/Chol DOX tumor growth suppres-
`sion of the MDR solid tumors, where tumor weight was 0.25 ⫾
`0.05 g on day 20. The tumor growth inhibition resulting from
`DSPC/Chol DOX and Valspodar
`treatment was significantly
`( p ⬍ 0.05) greater than that observed for EPC/Chol DOX and
`Valspodar.
`PEG-DSPE/DSPC/Chol DOX formulations displayed superior
`anti-tumor activity in the presence and absence of Valspodar when
`compared with the 2 liposomal DOX formulations described above
`as well as non-encapsulated drug (Fig. 1d ). In the absence of
`Valspodar, PEG-DSPE/DSPC/Chol DOX caused a significant
`reduction in tumor growth until day 11, when tumor weight was
`0.1 ⫾ 0.01 g (compared with a tumor weight of 0.05 ⫾ 0.01 g on
`day 1), after which tumor growth rates increased. Tumor weight for
`this group was 0.39 ⫾ 0.02 g on day 20. In the presence of
`Valspodar, PEG-DSPE/DSPC/Chol DOX significantly inhibited
`tumor growth and this growth suppression was not significantly
`different from WT tumors treated with non-encapsulated DOX at
`its MTD. Specifically, the tumor weights on day 20 for PEG-DSPE/
`DSPC/Chol DOX in presence of Valspodar were 0.1 ⫾ 0.02 g
`compared with 0.088 ⫾ 0.01 g for WT tumors treated with
`non-encapsulated DOX. This suppression of tumor growth for
`PEG-DSPE/DSPC/Chol DOX and Valspodar was significantly
`different ( p ⬍ 0.05) from all other treatment groups for mice
`bearing MDR solid tumors.
`
`Pharmacokinetics and tissue distribution
`A comprehensive PK evaluation was performed to correlate
`toxicity and efficacy data with plasma and tissue (tumor and liver)
`DOX and DOX metabolite distribution properties determined using
`HPLC analysis. The comparison of DSPC/Chol DOX and non-
`encapsulated (free) DOX pharmacokinetics and tissue distribution
`properties in the presence and absence of Valspodar has been
`extensively characterized (Krishna and Mayer, 1997). Since the
`toxicity and efficacy properties of these formulations were very
`comparable in the current models,
`the results presented here
`focused on comparisons between a liposomal system which leaks a
`significant portion of entrapped drug into the circulation (EPC/
`Chol DOX), and a sterically stabilized system that exhibits
`negligible drug release in plasma compartment and increased
`circulation lifetimes (PEG-DSPE/DSPC/Chol DOX). As a refer-
`ence comparison, non-encapsulated DOX treatments (with or
`without Valspodar) were evaluated for DOX concentrations in
`plasma and the tumor. The goal of these comparisons was to reveal
`important DOX distribution and metabolism properties that dictate
`toxicity and efficacy behavior.
`
`Plasma drug kinetics
`Figure 2a presents the DOX plasma concentrations after i.v.
`administration of non-encapsulated DOX and the 2 liposomal DOX
`formulations at a DOX dose of 5 mg/kg. Following administration
`of non-encapsulated drug, DOX is rapidly eliminated from the
`circulation. Concentrations of DOX beyond 4 hr were below assay
`detection limits. The concentration-time profile was characterized
`by a Cmax of 1.5 ⫾ 0.1 µg/ml and an AUC of 4.4 µg.hr/ml (Fig. 2a).
`However, when Valspodar was co-administered with non-
`encapsulated DOX at 5 mg/kg, DOX elimination from plasma was
`characterized by a prolonged terminal elimination phase (Fig. 2a).
`Valspodar caused significant ( p ⬍ 0.05) increases in Cmax (3.9 ⫾ 0.5
`µg/ml) and AUC (48.1 µg.hr/ml) of non-encapsulated DOX com-
`pared to data obtained in the absence of the MDR modulator. This
`approximately 11-fold increase in DOX AUC caused by Valspodar
`is consistent with the 10-fold increase in DOX AUC for the
`non-encapsulated DOX-Valspodar combination observed in the
`P388/ADR solid tumor model described earlier (Krishna and
`Mayer, 1997).
`As shown in Figure 2a, DOX elimination from plasma for both
`EPC/Chol and PEG-DSPE/DSPC/Chol DOX systems exhibits a
`monophasic elimination profile characterized by a 1-compartment
`model with first-order elimination. While EPC/Chol DOX plasma
`concentration exhibits rapid elimination of the drug within 24 hr,
`PEG-DSPE/DSPC/Chol DOX displays a prolonged circulation
`life-time with over 20% remaining at 24 hr. In all cases, parent
`DOX was the only detectable entity with no indication of any
`metabolites present. In the presence of Valspodar, the elimination
`profile remained monophasic, however, DOX concentrations at
`earlier time points were significantly ( p ⬍ 0.05) increased for
`EPC/Chol liposomes whereas no such Valspodar effect was ob-
`served for PEG containing DSPC/Chol liposomes. This is contrast
`to observations for non-encapsulated DOX which demonstrated
`increases in the terminal elimination phase in the presence of
`Valspodar (Fig. 2a). Co-administration of Valspodar and EPC/Chol
`DOX increased the AUC of DOX by 2.6-fold and Cmax by 2.3-fold,
`accounting for the 40% reduction in plasma clearance (Table II;
`significant at p ⬍ 0.05). In contrast, Valspodar caused minor
`changes in the pharmacokinetics of DOX encapsulated in PEG-
`DSPE/DSPC/Chol liposomes, with small 36% and 25% increases
`in Cmax and AUC, respectively (Table II).
`Figure 2b shows the elimination of liposomal lipid from plasma.
`These data demonstrate that, similar to DOX pharmacokinetics,
`liposomal lipid elimination is monophasic, characterized by a
`1-compartment model with first-order elimination. The plasma
`clearance (CLp) for PEG-DSPE/DSPC/Chol liposomes was 2.4-
`fold lower than EPC/Chol, and the half-life (T1/2) was 2.2-fold
`higher for PEG-DSPE/DSPC/Chol liposomes compared to EPC/
`
`AMN1077
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`136
`
`KRISHNA ET AL.
`
`Chol (Table II). Valspodar did not significantly alter the liposomal
`lipid elimination kinetics for either liposomal formulation, as
`demonstrated by the small changes in lipid ha

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