throbber
Pharmaceutical Research, Vol. 16, No. 10, 1999
`
`Research Paper
`
`Inhibition of P-Glycoprotein by D-a-
`Tocophery! Polyethylene Glycol
`1000 Succinate (TPGS)
`
`Jay M. Dintaman! and Jeffrey A. Silverman!”
`
`Received March 5, 1999; accepted June 30, 1999
`
`Purpose. To investigate whether d-ca-tocopheryl polyethylene glycol
`1000 succinate (TPGS) functions as an inhibitor of P-glycoprotein (P-
`gp), the multidrug resistance transporter.
`Methods. Two assays were used to measure the function of TPGS on
`P-gp function. First, we examined the ability of TPGS to modulate
`the cytotoxicity of established, cytotoxic, P-glycoprotein substrates.
`Parental NIH 3T3 cells and NIH 3T3cells transfected with the human
`MDR1 cDNA (G185) were exposed to doxorubicin, paclitaxel, colchi-
`cine, vinblastine and 5-fluorouracil (SFU) in the presence or absence
`of TPGS. Cytotoxicity was assessed with the MTT assay. Second,
`polarized transport of the P-gp substrates rhodamine 123 (R123), pacli-
`taxel and vinblastine was measured using the humanintestinal HCT-
`8 and Caco-2 cell
`lines grown in Transwell dishes. Drug flux was
`measuredbyliquid scintillation counting or fluorescence spectroscopy
`of the media.
`Results. G185 cells were 27-135 fold moreresistant to the cytotoxic
`drugs doxorubicin,vinblastine, colchicine and paclitaxelthan the paren-
`tal NIH 373 cells. In contrast SFU, which is not a P-gp substrate, is
`equally cytotoxic to parental and G185 cells. Co-administration of
`TPGSenhanced the cytotoxicity of doxorubicin,vinblastine, paclitaxel,
`and colchicine in the G185 cells to levels comparable to the parental
`cells. TPGS did not increase the cytotoxicity of 5FU in the G185 cells.
`Using a polarized epithelial cell transport assay, TPGS blocked P-gp
`mediated transport of R123 andpaclitaxel in a dose responsive manner.
`Conclusions. These data demonstrate that TPGS acts as a reversal
`agent for P-glycoprotein mediated multidrug resistance and inhibits P-
`gp mediated drug transport. These results suggest that enhanced oral
`bioavailability of drugs co-administered with TPGS may,in part, be
`due to inhibition of P-glycoprotein in the intestine.
`KEY WORDS:P-glycoprotein; TPGS; drug transport; bioavailability.
`
`INTRODUCTION
`
`The multidrug transporter, P-glycoprotein (P-gp), is a 170
`kDa membraneprotein which functions as an ATP-dependent
`drug efflux pump. One activity of this protein is to lower
`the intracellular concentration of drugs thereby reducing the
`cytotoxic activity of anticancer drugs. Increased expression of
`this protein has been observed in human tumors andis often
`associated with failure of chemotherapy due to drug resistance
`(1-5). P-gp removes a large number of chemically unrelated
`drugs extending over many therapeutic indications such as anti-
`cancer drugs, steroids, antihistamines, antibiotics, calcium
`channel blockers and anti-HIV peptidomimetics (2,4,5).
`
`
`
`‘ Division of Drug Transport, AvMax,Inc. Berkeley, California 94710.
`7To whom correspondence
`should
`be
`addressed.
`(e-mail:
`jeffrey.silverman@ att.net)
`ABBREVIATIONS: P-gp, P-glycoprotein; R 123, Rhodamine 123;
`CsA, Cyclosporine A; TPGS, d-c-tocopheryl polyethylene glycol
`1000 succinate.
`
`The P-gp drug transporter is encoded by one gene, MDR1,
`in humans whereas in rodents two genes, mdria and mdrib
`encode highly similar drug transporters (6,7). P-gp is primarily
`expressed on the luminalsurface ofepithelial cells from several
`tissues including the intestine, liver, kidney, and the endothelial
`cells comprising the blood-brain and blood-testes barriers (8—
`10). The ability of this protein to export toxic compounds
`combined with this localization led to the hypothesis that a
`physiological function of the MDR1 encoded P-gp may be as a
`protective barrier or export mechanism for xenobiotics. Indeed,
`recent investigations with knockout mice in which the mdrla
`gene wasdisrupted have confirmed such a protective role for
`P-gp (11-14). Exposure of mdr/a deficient mice to vinblastine
`or ivermectin results in significantly higher tissue and plasma
`levels compared to wild-type animals. Moreover, these com-
`pounds are toxic in the knockout mice at doses which are
`innocuous to heterozygous and wild-type mice. These experi-
`ments further suggesteda role for P-gp in the blood brain barrier
`since the ivermectin accumulated in the brain of the mdrla
`deficient animals but not animals with an intact mdrla gene.
`The knockout mice displayed ivermectin toxicity at doses 50
`to 100 fold less than wild-type mice.
`Additional data have supported a role of P-gp in the intes-
`tine as both a barrier to absorption as well as a mechanism of
`disposition of drugs such as vinblastine, etoposide, paclitaxel
`and digoxin. For example, Su and Huang observed that inhibi-
`tion of P-gp increased bioavailability of digoxin by increasing
`absorption as well as reducing excretion (15). A similar phe-
`nomenonwas observed with etoposide (16). P-glycoprotein has
`recently been suggested to becritical in oral drug absorption
`(17-19). In concert with the drug metabolizing enzyme CYP3A,
`P-gp maylimit oral drug bioavailability in the gut by controlling
`drugtransport from the intestinal lumen and byaffecting access
`to CYP3A (19).
`Vitamin E TPGS,d-a-tocopheryl polyethylene glycol 1000
`succinate,is a derivative of vitamin E consisting of a hydrophilic
`polar head group (tocopherol succinate) and a lipophilic alkyl
`tail (polyethylene glycol) resulting in amphiphillic properties
`(Eastman Kodak,
`technical bulletin EFC-226). TPGS has a
`relatively low critical micelle concentration, 0.02 wt%, and acts
`to solubilize lipophilic compounds. Bordreaux et al. reported
`a two-fold increase in cyclosporine CsA area underthe plasma
`concentration-time-curve (AUC) when co-administered with
`LiquiE, a glycerol and water solution of TPGS (20). Sokol et
`al. similarly observed increases up to 71% in CsA AUC in
`subjects who received concomitant TPGS(21). Both Sokol and
`Bordreaux suggested that the increased drug absorption was
`due to enhanced micelle formation, resulting in improved CsA
`solubilization. Chang et al.
`later reported a 61% increase in
`CsA AUC when dosed with 20-25% of the TPGS previously
`used in the Sokol or Bordreaux studies (22). Chang etal. also
`suggested that TPGS may interact with P-gp in the intestine to
`increase CsA absorption.
`In the current investigation we examine the effect of TPGS
`on P-gp mediated drug resistance and transport of established
`P-gp substrates. If this agent functions as a P-gp reversal agent
`then perhapsits effect on drug absorptionis, in part, mediated
`by inhibition of active drug efflux in the intestine. Our data
`show TPGSto be an effective inhibitor of P-gp mediated drug
`
`0724-874 1/99/ 1000- 1550$16.00/0 © 1999 Plenum Publishing Corporation
`
`1550
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1020-0001
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1020-0001
`
`

`

`TPGSInhibition of P-Glycoprotein
`
`resistance and transport at concentrations well below the
`reported critical micelle concentration and suggests that its
`reversal activity is due to an effect on transport activity.
`
`MATERIALS AND METHODS
`
`Cell Culture
`
`The NIH3T3 Swiss mouse embryocell line was obtained
`from American Type Culture Collection (ATCC, Rockville,
`MD)and was grown in Dulbecco’s Modified Eagles Medium
`(Biowhittaker, Walkersville, MD) supplemented with 4.5 g/L
`glucose, 10% fetal bovine serum (Hyclone Laboratories, Logan,
`UT), 2 mM L-glutamine (Advanced Biotechnologies Incorpo-
`rated (ABI), Columbia, MD), and 0.01 mg/ml gentamicin (ABI).
`The drug resistant, NIH-MDR-G185, cell line, derived by trans-
`fection of the human MDRI gene into NIH3T3 cells (23),
`was obtained from M. M. Gottesman (NCI, NIH) and was
`maintained in similar medium supplemented with 60 ng/ml of
`colchicine (Sigma, St. Louis, MO). HCT-8 cells (ATCC), iso-
`lated from a human ileocecal adenocarcinoma cell line, were
`grown in RPMI-1640 medium (Biowhittaker) supplemented
`with 10% horse serum (Biowhittaker), 1 mM sodium pyruvate
`(Gibco BRL, Grand Island NY) and 0.01 mg/m! gentamicin.
`Caco-2 cells (ATCC), derived from a human colonic adenocarci-
`noma, were grown in Eagle’s MEM (Biowhittaker) supple-
`mented with 10% fetal bovine serum, and 0.01 mg/ml
`gentamicin. All cells were maintained in a humidified atmo-
`sphere with 5% CO, at 37°C.
`
`Cytotoxicity Assay
`
`Cells were plated at a density of 2.5-3.0 < 10° cells/well
`in 96-well microtiter plates (PGC, Gaithersburg, MD) and were
`exposed to 1-5000 nM of doxorubicin, vinblastine, colchicine,
`paclitaxel, 0.1-25 nM 5-fluorouracil (Sigma) and 0.00 1—.005%
`TPGS(Eastman, Kingsport, TN) for 72 hours. To ensure solubi-
`lization of the TPGS, a 1% solution of TPGSin ethanol was
`prepared fresh for each experiment and diluted further in cell
`culture medium to the indicated concentrations. Cell viability
`wasdetermined with the colorimetric MTT (3-(4,5-dimethy|Ithi-
`azol-2-yl)-2,5-dipheny] tetrazolium, Sigma) assay as previously
`described (24,25) and the absorbance was measured with a
`Dynex MRX Microplate Reader (Chantilly, VA) at 570 nm.
`This assay is based on the reduction of MTT by mitochondria
`in viable cells to water insoluble formazan. The data presented
`are the mean SD ofat least 3 independent experiments, each
`performed in quadruplicate.
`
`Rhodamine 123 Transport
`
`Rhodamine 123 (R123; Sigma) transport was examined
`as previously described (26,27) using both HCT-8 and Caco-
`2 cells. Briefly, cells were grown in 6 well Corning Transwell
`dishes (HCT-8) or collagen coated Transwell dishes (Caco-2)
`until a tight monolayer was formed as measured by transepithe-
`lial electrical resistance or lucifer yellow impermeability. The
`integrity of the monolayers following the transport experiments
`was similarly evaluated. Typical TEER values were > 300
`Ohms/cm?. R123 was added ata final concentration of 13 4M
`to the basal or apical compartments and 200 wl samples were
`taken at the indicated times from the opposite chamber. TPGS
`
`1551
`
`was addedas an inhibitor to both compartments. Fluorescence
`of R123 in the media samples was measured using a Biotek
`FL500 Fluorescence Plate Reader (Winooski, VT) with an exci-
`tation wavelength of 485 nm and an emission wavelength of
`530 nm. All experiments were performedin triplicate; the data
`presented are the mean +SD andare representative of multi-
`ple experiments.
`
`Paclitaxel, Vinblastine and Cyclosporine Transport
`
`Inhibition of (H] paclitaxel (Moravek Biochemical, Brea,
`CA), PH] vinblastine (Amersham, Arlington Heights, IL), and
`[7H] cyclosporine (CsA; Amersham)efflux by TPGS was exam-
`ined in a manner similar to R123. The transported drug, 0.1
`wM (0.25 pCi/ml), was added to either the basal or apical
`compartment and 200 yl aliquots were taken at the indicated
`times from the opposite chamber. Radioactivity was measured
`by liquid scintillation counting.
`
`Western Blot Analysis
`
`Western blot analysis was performed as previously
`described 28. Briefly, crude cell membranes were isolated by
`lysing the cells in 10 mM Tris-HCl, pH 7.5; 10 mM NaCl;
`1
`mM MgCl, supplemented with pepstatin (1.5 wg/ml), leupeptin
`(1.5 jxg/ml) and 0.2 mM pefabloc. Cells were homogenized
`with 20 strokes of Dounce “B” (tight) pestle (Wheaton, Mill-
`ville, NJ), nuclei and cell debris were removed by centrifugation
`for 10 minutes at 400 X g. The supernatants were then centri-
`fuged at 100,000 X g for 30 minutes at 4°C andthepellets
`were resuspended in lysis buffer and stored at —80°C. 20 wg
`samples were fractionated in 8% polyacrylamide-SDSgel and
`transferred to 0.45 jxm nitrocellulose membrane. The mem-
`branes were blocked in PBS-T (0.1% Tween-20 in PBS) con-
`taining 5% skim milk for | hour and then probed with 1 wg/
`ml of C219 antibody (Signet Laboratories, Dedham, MA)in
`PBS overnight. The membranes were visualized by enhanced
`chemiluminescence according to the manufacturer’s instruc-
`tions (Pierce, Rockford, IL).
`
`RESULTS
`
`Western Blot Analysis
`
`Wefirst measured the relative levels of P-gp expression
`in the NIH3T3 and G185 cell lines by western blot analysis
`using the C219 antibody, which recognizes all P-gp isoforms
`(29). Consistent with previous data, high P-gp expression was
`observed in the G185 cells relative to that in the parental NIH
`3T3 cells (Fig. 1). We also examined P-gp expression in two
`humanintestinal carcinomacell lines, Caco-2 and HCT-8 which
`have been previously used for investigation of drug transport
`and to have polarized expression of P-gp (26). We observed
`that each of these intestinal cell
`lines have moderate P-gp
`expression albeit lower than the G185 cells (Fig. 1).
`
`Cytotoxicity Experiments
`
`The interaction of TPGS with P-gp wasinitially examined
`with cytotoxicity assays using parental NIH3T3 and MDRI
`transfected G185 cells to cytotoxic anticancer drugs. Consistent
`with previous reports (23,30), G185 cells were more resistant
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1020-0002
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1020-0002
`
`

`

`1552
`
`Dintaman and Silverman
`
` iene
`
`_ .
`Fig. 1. Western blot analysis of P-glycoprotein expression. Twenty
`microgramsof total cell membrane proteins were separated by SDS-
`PAGE,transferred to PVDFfilters which were subsequently probed
`with the C219 antibody and visualized using chemiluminescence as
`described in Materials and Methods. Lane 1, NIH-3T3 G185; lane 2,
`NIH-3T3; lane 3, HCT-8; lane 4, Caco-2.
`
`Table 1. ECsg in NIH3T3 and NIH3T3-G185 Cells
`NIH3T3- NIH37T3-
`G185
`G185
`(1 uM (SpM_ NIH3T3-G185
`NIH3T3-
`NIH3T3 Gi85 Verapamil) CsA)
`(.0025% TPGS)
`
`35
`35
`35
`950
`35
`Doxorubicin
`40
`6
`20
`270
`2
`Vinblastine
`1070
`40
`100
`>5000
`60
`Paclitaxel
`45
`100
`ND
`1000
`30
`Colchicine
`
`Note: NIH 3T3 and G185cells were treated with 0-5000 nM doxorubi-
`
`cin, vinblastine, paclitaxel or colchicine in the absence or presence of
`1 wM verapamil, 5 4M CsA or 0.0025% TPGS. The concentration of
`the drug that reduces cell viability by 50% (ECs)) was determined
`using the MTT cytotoxicity assay as described in Materials and Meth-
`ods. Each experiment was performed in quadruplicate and repeated in
`at least 3 independent experiments. ND, not determined.
`
`to doxorubicin, paclitaxel, vinblastine and colchicine compared
`to parental NIH3T3 cells (Fig. 2). ECs) values were 27 to 135
`fold higher in G185 cells relative to the parental NIH3T3cells
`(Table 1). Established P-gp reversal agents, such as cyclosporine
`A (CsA) and verapamil, reduced the resistance to doxorubicin
`cytotoxicity in G185 cells to levels comparable to parental
`NIH3T3cells (Fig. 3). The reversal effect of CsA on doxorubi-
`cin, vinblastine, taxol and colchicine mediated toxicity in paren-
`tal NIH3T3 cells was modest as previously reported. This is
`
`consistent with their low level of P-gp expression (data not
`shown, (27,30). Co-administration of CsA or verapamil caused
`a similar reversal of G185 resistance to vinblastine, paclitaxel,
`and colchicine (Table 1 and data not shown).
`The effect of TPGS on P-gp mediated drug resistance was
`investigated by treating G185 cells with doxorubicin, vinblas-
`tine, paclitaxel, and colchicine concomitantly with varying
`doses of TPGS. The presence of TPGS increased drug sensitiv-
`ity of the G185 cells to doxorubicin in a dose dependent manner
`
`
`
`
`Colchicine
`
`+ ¢- 373 Control
`—«~G185 Control
`
`Colchicine (nM)
`
`Paclitaxel
`
`- *- 373 Control
`[6185Contra
`
`1.4
`5"
`
`23aSo
`
`s>
`B=
`&a
`4
`
`a
`12
`
`2f
`
`> 2
`
`os
`=a
`3 04
`a
`
`os
`
`- -373 Control.
`—»~—G185 Control
`
`1
`
`40
`
`100
`
`+000
`
`10000
`
`Doxorubicin (nM) =
`
`3S
`
`> £
`
`&ao
`a
`
`08
`
`06
`0.4
`02
`0
`
`Doxorubicin
`
`
`Vinblastine
`_——
`
`> o- 3T3 Control
`—2—G185 Control
`
`1
`
`10
`
`100
`
`4000
`
`10000
`
`1.4
`c
`z ,
`3S os
`
`5 2
`
`os
`a3 04
`a 0.2
`
`Vinblastine (nM)
`
`1
`
`10
`
`100
`
`4000
`
`10000
`
`Fig. 2. Cytotoxicity of doxorubicin (A), colchicine (B), vinblastine (C), and taxol (D) in parental NIH-3T3 cells (circles), and
`MDRI-transfected NIH-3T3 G185 cells (squares). Cells were treated with the indicated concentrations of drugs and the viability
`was measured by the MTT assay as described in Materials and Methods. Data are expressed relative to untreated controlcells.
`Each experiment was performed in quadruplicate and the data presented represent the mean + SD offour independent experiments.
`
`Paclitaxel (nM)
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1020-0003
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1020-0003
`
`

`

`TPGSInhibition of P-Glycoprotein
`
`1.6
`
`e+ 373 Control
`——G185 Coatrol
`= > Gt8S SO pM CoA
`—O—GI185 5.0 uM Verap
`
`Viability 1
`Relative
`
`10
`
`400
`
`1000
`
`10000
`
`Doxorubicin (nM)
`Fig. 3. Effect of CsA and Verapamil on the cytotoxicity of doxorubicin.
`Parental NIH-3T3 (diamonds) and G185 (squares) cells were exposed
`to the indicated concentrations of doxorubicin in the absence or pres-
`ence of CsA, 5.0 41M, or verapamil, 5.0 .M.. Data are expressed
`relative to untreated control cells. Each experiment was performed in
`quadruplicate and the data presented represent the mean + SDof four
`independent experiments.
`
`(Fig. 4). Treatment ofthe drug resistant G185 cells with TPGS
`lowered the ECs) concentrations for doxorubicin, vinblastine,
`paclitaxel and colchicine (Table 1). TPGS, 0.0025%, sensitized
`the G185 cells to all four of these P-gp substrate cytotoxic
`drugs to levels comparable to the parental NIH 3T3 cells. The
`highest dose of TPGS, 0.005%, resulted in decreased viability
`of both NIH3T3 and G185 cells and is likely due to toxicity
`associated with the high concentration of TPGS. At concentra-
`tions below 0.005% TPGSitself did not affect cell viability.
`These data suggest that TPGS modulates drug resistance by
`inhibiting P-gp activity in cells which over-express
`the
`MDRI1 gene.
`
`5-Fluorouracil Cytotoxicity
`
`Treatment of parental NIH 3T3 and G185 cells with 5-
`fluorouracil (SFU), a chemotherapeutic agent not transported
`
`1553
`
`by P-gp, results in a similar level of cytotoxicity in both cell
`lines (Fig. 5A) (1). Furthermore, co-incubation of SFU with
`CsA had no effect on the cytotoxicity of 5FU in either G185
`or NIH3T3 cells (Fig. 5B). Similarly, co-incubation of TPGS
`with 5FU did not increase the cytotoxicity of 5FU in either of
`these cell lines (Fig. SC).
`
`Rhodamine 123 Transport
`
`The fluorescent dye R123, an established substrate of P-
`glycoprotein (32,33), was used to examinethe ability of TPGS
`to block P-gp mediated transport. HCT-8 and Caco-2 cells have
`previously demonstrated directional transport of established P-
`gp substrates such as vinblastine, paclitaxel, CsA and R123 in
`the basolateral to apical direction (26,34—36). Expression of P-
`gp in these cells was confirmed by western blot analysis using
`the C219 antibody (Fig. 1). R123 was transported approximately
`7 and 9 fold greater flux in the basolateral to apical direction
`in HCT-8 and Caco-2 cells, respectively (Fig. 6). Consistent
`with this transport being mediated by P-gp, R123 flux was
`inhibited approximately 80% by co-incubation with 5 uM CsA.
`Similarly, 0.001—0.0025% TPGS blocked the basolateral
`to
`apical transport of R123 in a dose responsive manner further
`suggesting that TPGS inhibits transport mediated by P-gp
`(Fig. 6).
`
`Paclitaxel Transport
`
`The ability of TPGS to inhibit P-gp was confirmed by
`measuring polarized transport of paclitaxel. [7H] Paclitaxel is
`a good substrate for P-gp with approximately 14 and 40 fold
`greater transport from the basolateral to the apical compartment
`in HCT-8 and Caco-2 cells, respectively (Fig. 7). Addition of
`5 uM CsAblocked the polarized flux of paclitaxel by 80-90%.
`Similarly, co-incubation with TPGS resulted in a dose depen-
`dent decrease in paclitaxel transport (Fig. 7). The ICs) of TPGS
`for inhibition of paclitaxel transport is approximately 0.001%
`(v/v) in HCT-8 cells and 0.005% in Caco-2 cells. Polarized
`
`
`
`
`
`
`—e- -NIH3T3
`—s— G18S
`+-@- G185+0.001% TPGS
`—*- -G185+0.0025% TPGS
`—0— G185+0.005% TPGS
`
`
`
`
`
`
`
`RelativeViability
`
`~ oe _—
`
`1
`
`10
`
`100
`
`1000
`
`10000
`
`Doxorubicin (nM)
`
`Fig. 4. TPGS reversal of P-gp mediated resistance to doxorubicin. Parental NYH-3T3 (diamonds) and
`G185 cells were exposedto the indicated concentrations of doxorubicin with 0% TPGS(squares), 0.001%
`TPGS(circles), 0.0025% TPGS(triangles) or 0.005% TPGS (open circles, ©). Data are expressed relative
`to untreated control cells. Each experiment was performed in quadruplicate andthe data presented represent
`the mean +SD of four independent experiments.
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1020-0004
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1020-0004
`
`

`

`Dintaman and Silverman
`
`—8— Control B:A
`—O— Control A:B
`++ de CSA BIA
`—— 0.001% TPGS B:A
`— + ~ 0.0025% TPGS B:A
`
`A Caco-2
`$
`8
`7
`
`3a
`
`£5
`
`6
`ow
`&
`Cc
`© 4
`Fe
`ao
`3
`
`Sm
`
`A
`
`“
`
`5-Flucrouracil
`
`bn G85
`
`[Te NIH373
`
`1554
`
`£a3> >c
`
`s
`xc]oe
`x
`
`
`
`
`
`
`
`2
`S14
`
`
`
`0
`5-Fluorouracil (nM)
`
`0
`
`1
`
`4
`
`5
`
`6
`
`2 3
`Time, hrs
`
`—#— Control B:A
`~—~&— Control! A:B
`— @- 0.005% TPGS B:A
`
`B Het-8
`
`:-@--0.001%TPGS B:A
`
`~#-- 0.0025%TPGS B:A
`
`Time, hrs
`Fig. 6. Rhodamine 123 transport in Caco-2 and HCT-8 cells. Caco-2
`(A) and HCT-8 (B) were grown on Transwell dishes as described in
`Materials and Methods. Rhodamine 123, 13 «1M, was added to the
`apical or basolateral compartment in the absence or presence of CsA,
`5 uM, or 0.0025, 0.005, 0.001% TPGS and media aliquots were taken
`from the opposite chamber at the indicated times. The data presented
`are the mean +SD oftriplicate wells and are representative of at least
`three independent experiments.
`
`10 wM 5Fiuorouracil with CsA
`
`ONIH3T3
`mG185
`
`
`
`
`06
`6
`
`0
`
`CsA (4M)
`
`7.5 1M 5-Fluorouracil with TPGS
`
`ae
`wGI85s
`|
`ee
`
`0.001
`
`9.0025
`wt% TPGS
`
`0.006
`
` o
`
`oo B
`OB
`
`2”
`Bos
`2 06
`£ o4
`Sos o2
`oA
`Qa
`
`09 c
`os
`07
`
`2
`3z
`>oe>
`
`=2o~
`
`Fig. 5. Cytotoxicity of 5-fluorouracil to parental NIH-3T3, and drug
`resistent NIH-3T3 G185cell lines in the presence and absence of CsA
`and TPGS. A. NIH-3T3 (diamonds) and NIH-3T3 G185 (squares)cells
`were exposedto the indicated concentrations of 5-fluorouracil. Viability
`was measured by the MTTassayas described in Materials and Methods.
`Data are expressed relative to untreated control cells. Each experiment
`was performed in quadruplicate and the data presented represent the
`mean +SD of three independent experiments. B. Parental NIH-3T3
`(open bars) and G185 (closed bars) cells were exposed to 10 4M of
`SFU with the indicated concentrations of CsA. C. Parental NIH-3T3
`(open bars) and G185 (closed bars) cells were exposed to 7.5 1M of
`SFU and the indicated concentrations of TPGS.
`
`transport of [3H] vinblastine and [7H] CsA were also inhibited
`by addition of TPGS (data not shown). These data, combined
`with the cytotoxicity and R123 transport data suggest that TPGS
`is an effective P-gp reversal agent.
`
`DISCUSSION
`
`A majoreffort has been undertaken by many laboratories
`to identify inhibitors of P-glycoprotein to increase the efficacy
`of cancer treatment and to enhance the absorption of orally
`administered drugs. The data presented here support the hypoth-
`esis that TPGS functions as one such P-gp inhibitor. TPGS
`
`increased the sensitivity of P-gp expressing cells to several
`widely used cytotoxic drugs which are well established P-gp
`substrates. TPGS also effectively blocked polarized transport
`of R123 and paclitaxel in an epithelial cel! transport assay. The
`reduction of directional transport provides strong evidence for
`TPGS functioning as an inhibitor of P-gp. Conversely, no effect
`was observed with SFU, a cytotoxic drug not associated with
`P-gp mediated drug resistance or transport. 5FU is not transpor-
`ted by the P-gp pumpthus, its cytotoxicity is unaffected by the
`addition of established P-gp inhibitors such as quinine, quini-
`dine or verapamil (31,37). In the experiments presented here
`neither TPGS nor CsA impacted the cytotoxicity of SFU in
`either the NIH 3T3 or G185cells.
`Previously it has been suggested that co-administration of
`TPGSwith CsA enhancedabsorption of the immunosuppressant
`due to micelle formation (21). Concentrations of TPGS adminis-
`tered in the current work are well below the critical micelle
`concentration, 0.02 wt% in waterat 37°C,thereforeit is unlikely
`that micelle formation is responsible for the observed effects.
`In fact, the ICsrequired to inhibit R123 and paclitaxel transport
`across HCT-8 or Caco-2 cell monolayers is 20 fold less than.
`the critical micelle concentration. Further, 0.001 wt% TPGS
`also significantly reversed the multidrug resistant phenotype of
`the NIH3T3-G185 cell line to doxorubicin, vinblastine, taxol
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1020-0005
`
`
`
`0.00 «45.00=17.80«626006=— 6.00 7.60 «010.00 12.50 20.00 «2250 26.00
`
`
`
`
`
`
`
`
`Transported
`%R123
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1020-0005
`
`

`

`TPGSInhibition of P-Glycoprotein
`
`1555
`
`--@--.005% TPGSBtoA
`
`1.87(aPedtawiAtwe A
`
`receiving TPGShad previously experienced chronic cholestasis
`1.6||—-t—Paclitaxel B to A
`—~@—CSABtOA
`resulting in decreased bile flow suggesting poor solubilization
`1.4
`— dh— .0025% TPGS BtoA
`of the lipophilic CsA. It was hypothesized that TPGS functioned
`as a bile substitute and solubilized the CsA through micelle
`formation, thus facilitating the absorption of the drug through
`the intestinal lumen. Similarly, Pan et al. reported a 28 and
`32% decrease in CsA daily dose when co-administered with
`Liqui-E, a water soluble form of TPGS and a 26% decrease in
`daily CsA cost (44). Using normal healthy volunteers, Chang
`et al. observed a 60% rise in CsA area under the curve (AUC)
`in subjects receiving a TPGS-CsA cocktail. Decreased oral
`clearance and volume of distribution were also observed in
`those subjects. These authors proposed that the large, amphi-
`pathic TPGS mayalso be acting as an inhibitor of P-glycopro-
`tein to enhance absorption and decrease transport back into the
`intestinal lumen. The current data support the hypothesis that
`one mechanism through which TPGS may enhance oral bio-
`availability is via inhibition of P-gp. Clearly further study on
`the effect of TPGS on oral drug delivery is required to confirm
`sucharole.
`
`Time, hrs
`
`B
`
`[—€3-—-Pacitaxal Ato B
`—t—Pacitaxel BtoA
`—-*—CsABIOA
`— A— 0025 % TPGS BtoA
`= + @ + +.005 % TPGS Bto A
`— HE - 01% TPGS BtoA
`
`
`
`35
`
`Paclitaxel,
`
`pmol
`Paclitaxel,
`pmol
`
`Time, hrs
`
`REFERENCES
`
`2.
`
`7.
`
`1. L. J. Goldstein. MDR/ gene expression in solid tumors. Eur. J.
`Cancer. 32A:1039—1050 (1996).
`J. A. Endicott and V. Ling. The biochemistry of P-glycoprotein
`mediated multidrug resistance. Ann. Rev. Biochem. 58:137-171
`(1989).
`3. M. M. Gottesman. How cancer cells evade chemotherapy: Six-
`Fig. 7. Paclitaxel transport in HCT-8 and Caco-2 cells. Cells were
`teenth Richard and Hinda Rosenthal foundation award lecture.
`grown on in Transwell dishes as described in Materials and Methods.
`Cancer Res. 53:747-754 (1993).
`(7HjPaclitaxel, 0.1 41M (0.2 4.Ci/ml) wasplacedin the basolateral (B)
`4. M. M. Gottesman and I. Pastan. The multidrug transporter, a
`or apical (A) chamber; media aliquots were takenat the indicated times
`double edged sword. J. Biol. Chem. 263:12163—12166 (1988).
`and radioactivity was measuredby liquidscintillation counting. Control
`5. M.M. Gottesman andI. Pastan. Biochemistry of multidrug resis-
`Caco-2 (A) or HCT-8 (B) cells B to A (closed squares), control A to
`tance mediated by the multidrug transporter. Ann. Rev. Biochem.
`62:385-427 (1993).
`B (open squares), 5 ~M CsA B to A (diamonds), 0.0025% TPGS B
`6. S.S. Thorgeirsson, J. A. Silverman, T. W. Gant, and P. A. Marino.
`to A (triangles), 0.005% TPGSBto A (circles).
`Multidrug resistance gene family and chemical carcinogens. Phar-
`macol. Ther. 49:283-292 (1991).
`P. Borst, A. H. Schinkel, J. J. M. Smit, E. Wagenaar, L. Van
`Deemter, A. J. Smith, W. H. M. Eijdems, F. Baas, and G. J. R.
`Zaman. Classical and novel forms of multidrug resistance and
`the physiological functions of P-glycoproteins in mammals. Phar-
`macol. Ther. 60:289-—299 (1993).
`8. F. Thiebaut, T. Tsuruo, H. Hamada, M. M. Gottesman,I. Pastan,
`and M. C. Willingham. Cellular localization of the multidrug
`resistance gene product P-glycoprotein in normal humantissues.
`Proc. Natl. Acad. Sci. USA 84:7735-7738 (1987).
`9. F. Thiebaut, T. Tsuruo, H. Hamada, M. M. Gottesman,I. Pastan,
`and M. C. Willingham. Immunohistochemicallocalization in nor-
`mal tissues of different epitopes in the multidrug transport protein
`P170: Evidencefor localization in brain capillaries and crossreac-
`tivity on one antibody with a muscle protein. J. Histochem. Cyto-
`chem. 37:159-164 (1989).
`10. B.L. Lum and M. P. Gosland. MDRexpression in normal tissues.
`Hematol. Oncol. North Am. 9:319-336 (1995).
`11. A. H. Schinkel, J. J. M. Smit, O. van Tellingen, J. H. Beijnen,
`E. Wagenaar, L. van Deemter, C. A. A. M. Mol, M. A. van der
`Valk, E. C. Robanus-Maandag,H. P. J. te Riele, A. J. M. Berns,
`and P. Borst. Disruption of the mouse mdr/a P-glycoprotein gene
`leads to a deficiency in the blood-brain barrier and to increased
`sensitivity to drugs. Cell 77:491~502 (1994).
`J. van Asperen, O. van Tellingen, A. Sparreboom, A. H. Schinkel,
`P. Borst, W. J. Nooijen, and J. H. Beijnen. Enhancedoral bioavail-
`ability of paclitaxel in mice treated with the P-glycoprotein blocker
`SDZ PSC 833. Br J. Cancer 76:1181—1183 (1997).
`J. van Asperen, A. H. Schinkel, J. H. Beijnen, W. J. Nooijen, P.
`Borst, and O. van Tellingen. Altered pharmacokinetics of vinblas-
`tine in mdrla P-glycoprotein-deficient mice. J. Natl. Cancer Inst.
`88:994—999 (1996).
`
`and colchicine,all established P-gp substrates. These data sug-
`gest that TPGS micelle formation in the intestinal lumen may
`not be the sole factor behind the increase in CsA absorption
`previously observed (20-22).
`Several other surfactants, e.g. polysorbates, Cremophor
`EL, and Solutol 15, have been observed to be inhibitors of P-
`gp (38-41). These compoundsare frequently added to pharma-
`ceutical formulations to enhance solubility. These agents may
`also function to inhibit P-gp to addto their effect of enhancing
`drug absorption. Indeed the plasma concentrations of Cremo-
`phor ELin patients administered paclitaxel, which is formulated
`with this surfactant, reach levels sufficient to inhibit P-gp in
`vitro (42). The efficacy of this drug may, in part, be due to the
`activity of the Cremophor EL. Pluronic P85 has also recently
`been observed to block P-gp mediated rhodamine 123 efflux
`in Caco-2 and bovine brain microvessel endothelial cells (43).
`These data suggested that this agent may be useful for formula-
`tions to enhance brain and oral absorption.
`TPGS has been used to enhance the bioavailability of
`CsAin liver transplant patients with the effects of significantly
`improving absorption and reducing daily drug cost. Sokol et
`al. and Boudreauxet al. reported increases in CsA absorption
`in pediatric transplant recipients treated with oral TPGS, 12.5
`IU/kg and 10 U/kg, respectively. The majority of patients
`
`12.
`
`13.
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1020-0006
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1020-0006
`
`

`

`1556
`
`Dintaman and Silverman
`
`14.
`
`A. H. Schinkel, U. Mayer, E. Wagenaar, C. A. A. M. Mol, L.
`van Deemter, J. J. M. Smit, M. A. van der Valk, A. C. Voordouw,
`H. Spits, O. van Tellingen, J. M. J. M. Zijlmans, W. E. Fibbe,
`and P. Borst. Normal viability and altered pharmacokinetics in
`mice lacking mdr -type (drug transporting) P-glycoproteins. Proc.
`Natl. Acad. Sci. USA 94:4028—4033 (1997).
`. §S.-F Su and J. -D. Huang. Inhibition of the intestinal digoxin
`absorption and exsorption by quinidine. Drug Metab. Dispos.
`24:142-147 (1996).
`B. -L. Leu and J. -ding Huang. Inhibition of intestinal P-glycopro-
`tein and effects on etoposide absorption. Cancer Chemother. Phar-
`macol. 35:432—436 (1995).
`V. J. Wacher, C. -Y. Wu, and L. Z. Benet. Overlapping substrate
`specificities and tissue distribution of cytochrome P450 3A and
`P-glycoprotein: Implications for drug delivery and activity in
`cancer chemotherapy. Mol. Carcin. 13:129-134 (1995).
`. V.J. Wacher, L. Salphati, and L. Z. Benet. Active secretion and
`enterocytic drug metabolism barriers to drug absorption. Adv.
`Drug Del. Rev. 20:99-112 (1996).
`V. J. Wacher, J. A. Silverman, Y. Zhang, and L. Z. Benet. Role
`of P-glycoprotein and cytochrome P450 3A in limiting oral
`absorption of peptides and peptidomimetics. J. Pharm. Sci.
`87:1322—1330 (1998).
`J. P. Boudreaux, D. H. Hayes, S. Mizrahi, P. Maggiore, J. Blazek,
`and D. Dick. Use of water-soluble liquid vitamin E to enhance
`cyclosporine absorption in children after liver transplant. Trans-
`plant. Proc. 25:1875 (1993).
`R. J. Sokol, K. E. Johnson, F. M. Karrer, M. R. Narkewicz, D.
`Smith, and I. Kam. Improvement of cyclosporin absorption in
`children after liver transplantation by means of water-soluble
`vitamin E. Lancet. 338:212-215 (19

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