throbber
O R I G I N A L
`A R T I C L E
`
`P-glycoprotein is not involved in the
`differential oral potency of naloxone and
`naltrexone
`
`doi: 10.1111/j.1472-8206.2009.00724.x
`
`Mouna Kanaan, Youssef Daali, Pierre Dayer, Jules Desmeules*
`Clinical Pharmacology and Toxicology and Multidisciplinary Pain Center, Department of Anesthesiology, Pharmacology
`and Intensive care, Geneva University Hospitals, Faculty of Medicine, University of Geneva, CH-1211 Geneva 14,
`Switzerland
`
`A B S T R A C T
`
`The poor oral bioavailability of the opioid receptor antagonist naloxone (NA) when
`compared with naltrexone (NX) may be related to a greater interaction of naloxone
`with the efflux drug transporter P-glycoprotein (P-gp). We studied the involvement of
`P-gp in the transepithelial transport of the two opioid receptor antagonists, using a
`validated human in vitro Caco-2 cell monolayer model. The bidirectional transport
`of NA and NX (1, 50 and 100 lM) across the monolayers was investigated in the
`presence and absence of the specific P-gp inhibitor GF120918 (4 lM). NA and NX
`showed equal transport rates between the apical-to-basolateral (A–B) and the
`basolateral-to-apical (B–A) directions and neither the influx nor the efflux transport
`was affected by the P-gp inhibitor (P > 0.05). In conclusion, NA and NX are not P-gp
`substrates. The differential oral bioavailability of the two opioid antagonists is P-gp
`independent.
`
`Keywords
`absorption,
`Caco-2 cells,
`naloxone,
`naltrexone,
`oral bioavailability,
`P-glycoprotein
`
`Received 14 November 2008;
`revised 6 March 2009;
`accepted 13 March 2009
`
`*Correspondence and reprints:
`jules.desmeules@hcuge.ch
`
`I N T R O D U C T I O N
`
`Naloxone (NA), a synthetic N-allyl derivative of oxy-
`morphone, and its analogue naltrexone (NX) are both
`opioid antagonists at the mu, kappa and sigma opioid
`receptor sites [1] (Figure 1). Naloxone is the treatment of
`choice for opioid overdose, regardless of the type of
`opioid, as it permits a dose dependant reversal of opioid
`adverse events in the central nervous system. It is mostly
`administered by intravenous
`injection for complete
`bioavailability and rapid onset of action. Naloxone is
`mainly metabolized in the liver by glucuroconjugation
`into naloxone-3-glucuronide [2] and eliminated by the
`renal route [3]. Naltrexone is approximately twice as
`potent as naloxone [4] and is mostly used orally for the
`treatment of alcohol dependence. Naltrexone is metab-
`olized by dihydrodiol dehydrogenase to the main active
`metabolite 6-beta-naltrexol, via extensive hepatic and
`extra-hepatic routes [5]. Although a weaker opioid
`antagonist than naltrexone, 6-beta-naltrexol possibly
`contributes to the long duration of action observed with
`the parent compound, particularly because plasma
`
`concentrations of the metabolite are higher than those
`of the parent drug. Naltrexone and 6-beta-naltrexol are
`mainly excreted by the renal route [6].
`Naloxone has a low oral bioavailability when com-
`pared to naltrexone [7–10]. It is, however, unclear
`whether the very limited systemic availability of oral
`naloxone is due to a more extensive first pass meta-
`bolism, when compared with naltrexone, or
`to a
`poor gastrointestinal absorption. P-glycoprotein (P-gp)
`(ABCB1/MDR1) is expressed in the major organs asso-
`ciated with drug absorption, distribution and elimina-
`tion, therefore playing a major role in pharmacokinetics
`of several drugs.
`
`M A T E R I A L S A N D M E T H O D S
`
`Materials
`Caco-2 cells (TC7 clone) were kindly provided by Tea
`Fevr, PhD (ISREC, Swiss Institute for Experimental
`Cancer Research, Lausanne, Switzerland). Penicillin–
`streptomycin was purchased from Sigma-Aldrich GmbH
`(Steinheim, Germany) and all other cell culture reagents
`
`ª 2009 The Authors Journal compilation ª 2009 Socie´ te´ Franc¸aise de Pharmacologie et de The´ rapeutique
`Fundamental & Clinical Pharmacology 23 (2009) 543–548
`
`543
`
`Nalox1222
`Nalox-1 Pharmaceuticals, LLC
`Page 1 of 6
`
`

`

`544
`
`M. Kanaan et al.
`
`Naloxone
`
`Naltrexone
`
`Figure 1 Chemical structures of naloxone and naltrexone.
`
`from Gibco BRL (Paisley, Scotland). Naloxone and
`naltrexone were purchased from Sigma-Aldrich GmbH
`and GF120918 was kindly provided by GlaxoSmithKline
`(Middlesex, UK).
`
`Cell culture
`Caco-2 cells (TC7 clone) were used at passages 21–25.
`Cells were cultured in Dulbecco’s modified Eagle’s
`medium (DMEM Glutamax, Gibco BRL) supplemented
`by 20% fetal bovine serum (FBS, Gibco BRL), 1%
`nonessential amino acids (NEA, Gibco BRL), 100 U/mL
`penicillin and 100 lg/mL streptomycin (Sigma-Aldrich),
`at 37 °C in a humidified atmosphere with 5% CO2.
`At 85–95% confluency, Caco-2 cells were treated with
`0.25% trypsin–EDTA (Gibco BRL) and seeded at a
`density of 65 000 cells/cm2 on polycarbonate mem-
`(12-mm diameter, 1.13 cm2,
`branes of Transwells
`0.4 lM pore size, 12-well plates; Costar, Cambridge,
`MA, USA), previously equilibrated for 1 h in the culture
`medium. Medium was changed the day after seeding and
`every other day thereafter [apical volume (A): 0.5 mL,
`basolateral volume (B): 1.5 mL]. Monolayers were used
`for transport studies 20–21 days post seeding to allow
`full maturation of the cells, including P-gp expression
`and appropriate tight junctions.
`
`Measurement of transepithelial electrical
`resistance
`Transepithelial electrical resistance (TEER) was checked
`every 5 days during the 21-day maturation of
`the
`monolayers. Prior to bidirectional
`transport studies,
`medium was removed from both apical and basolateral
`chambers and monolayers were rinsed three times with
`the transport buffer Hank’s balanced salt
`solution
`(HBSS),
`supplemented with 25 mM N-(2-hydroxyl-
`ethyl)piperazine-N¢-2ethane-sulfonic acid (HEPES) (Gib-
`co BRL) and pH adjusted to 7.4 with 0.5 M NaOH. Cells
`were equilibrated in the same buffer for 1 h and the
`integrity of each monolayer was checked by measuring
`
`its TEER with a Millicell-ERS ohmmeter (Millipore Corp.,
`Bedfort, MA, USA). Resistance was also checked imme-
`diately after the experiments.
`
`Transmission electron microscopy and Western
`blotting of P-glycoprotein
`Transmission electron microscopy and Western blotting
`of P-glycoprotein were performed in the context of
`the Caco-2 model validation in our previous study
`(see [11]).
`
`Transport studies
`After measurement of TEERs, HBSS buffer was removed
`from each chamber. Apical to basolateral (A–B) trans-
`port was initiated by replacing basolateral (B) buffer with
`1.5 mL of fresh HBSS supplemented with 25 mM HEPES,
`1% DMSO and pH adjusted to 7.4 with NaOH 0.5 M, and
`replacing apical (A) buffer with 0.5 mL of the drug
`solution in the same buffer (HBSS/HEPES pH 7.4). In
`other wells, B–A transport was initiated by replacing (A)
`buffer with 0.5 mL of fresh HBSS/HEPES pH 7.4 and (B)
`buffer with the drug solution in the same buffer
`(1.5 mL). For the P-gp inhibition studies, GF120918
`(4 lM) was present in both chambers. Samples (100 lL)
`were removed from each receiver chamber at various
`times (30, 60, 90, 120 and 180 min) and replaced with
`buffer to maintain constant volumes. The 3-h transport
`studies were performed at a constant agitation rate
`(50 rpm) using a circular shaker (type SSM1, StuartÒ) in
`an incubator (37 °C, 5% CO2 and humidified atmo-
`sphere). After the transport studies, all aliquots were
`stored at )20 °C until analysis.
`
`Analytical method
`Naloxone and naltrexone analysis for the 50 and 100 lM
`assays was performed by LC-UV at 230 nm. Separation
`was carried out on a Zorbax eclipse XDB-C8 (150 ·
`4.6 mm i.d., particle size: 5 lM) from Agilent, coupled
`with a guard column with the same stationary phase. The
`mobile phase consisted of a mixture of acetonitrile and
`orthophosphoric acid (50 mM) adjusted to pH 4.2 with
`sodium hydroxide 4N and was delivered at 0.9 mL/min. A
`gradient elution was used in which the mobile phase
`composition was changed from 12–88% to 30–70%
`(ACN-Orthophosphoric acid) within 5 min, maintained
`at 30–70% until t = 8 min, and returned to the initial
`composition from t = 8 to t = 9 min. Naloxone and
`naltrexone analysis for 1 and 10 lM assays was achieved
`using an LC–MS system (Esquire 3000 + Ion-Trap) from
`Bruker daltonics (Billerica, MA, USA) equipped with an
`
`ª 2009 The Authors Journal compilation ª 2009 Socie´ te´ Franc¸aise de Pharmacologie et de The´ rapeutique
`Fundamental & Clinical Pharmacology 23 (2009) 543–548
`
`Nalox1222
`Nalox-1 Pharmaceuticals, LLC
`Page 2 of 6
`
`

`

`Naloxone, naltrexone and P-glycoprotein
`
`545
`
`electrospray source working in positive ion mode. The ion
`transitions monitored in multiple reaction-monitoring
`modes were m/z 342 fi 324, and 328 fi 310 for
`naltrexone and naloxone, respectively. Optimized ESI
`source voltages were as
`follows:
`spray needle at
`+4.3 kV, end plate offset at )500 V, capillary exit offset
`at )200 V, skimmer 1 at )107.4 V. Further ion source
`parameters were 70 psi nebulizer gas and 11 L/min of
`drying gas with a temperature of 350 °C. Separation was
`achieved with an XTerraÒ MS C18 column (100 mm ·
`2.1 mm i.d., particle size: 3.5 lM) from Waters (Milford,
`MA, USA) at 0.3 mL/min. Mobile phase consisted of
`ammonium formate 20 mM and acetonitrile (20–80%).
`In all cases, the standard curves were obtained by
`linear regression of measured peak area vs. concentra-
`tion and used to calculate concentrations of the analytes
`in unknown and QC samples. Samples consisted of
`aliquots removed from the receiver chambers (drug
`solution in the aqueous buffer HBSS). No additional
`treatment was needed. No extraction was required and
`the samples were directly injected into the HPLC system
`without need for an internal standard. The performance
`of both methods, in terms of reproducibility, repeatability
`and linearity were assessed before analysis (data not
`shown).
`
`Calculations
`TEER was calculated from the following equation [12]:
`
`TEER ¼ ðTEERmono TEERblankÞ  A
`
`where TEERmono is the cell monolayer and polycarbonate
`porous membrane resistance, TEERblank the polycarbon-
`ate porous membrane resistance and A the polycarbonate
`porous membrane surface area (1.13 cm2).
`Apical to basolateral (Papp(A–B)) and basolateral to
`(Papp(B–A)) apparent permeability coefficients
`apical
`were calculated according to Artursson [13] using the
`following equation:
`
`Pappðcm/sÞ ¼ ðdQ=dtÞ=ðA  C0  60Þ
`
`where dQ/dt (lg/min) is the permeability rate of the
`drug, calculated from the regression line of the time
`points of sampling, A is the surface area of the monolayer
`(cm2) and C0 the initial drug concentration in the donor
`chamber (lg/L).
`Karlsson et al. [14] suggested that there is involve-
`ment of a drug efflux transporter in Caco-2 cells if the
`efflux ratio (TR = Papp(B–A)/Papp(A–B)) is >2 and if a
`
`decreased secretory transport rate (Papp(B–A)) is ob-
`served in the presence of an inhibitor of the transporter
`in question. For a compound with an efflux ratio of 1.5–
`2.0, a positive effect of
`the inhibitor confirms the
`implication of the efflux transporter [15].
`
`Statistics
`The unpaired bilateral Student’s t test was used for
`statistical comparison of the transport rates in each
`direction and the transport rates in the presence or
`absence of the P-gp inhibitor for a particular direction
`(Xlstat version 5.0; Addinsoft, New York, NY, USA).
`A P value of <0.05 was considered significant.
`
`R E S U L T S
`
`Differentiation and integrity of Caco-2 cell
`monolayers
`Transmission electron microscopy (TEM)
`Histological examination showed a continuous differen-
`tiated cell monolayer presenting microvilli on the apical
`cell surface, interdigitations, numerous desmosomes, and
`tight junctions (see [11]).
`
`Transepithelial electrical resistance
`Caco-2 cell monolayers with TEER values between 300
`and 400 W.cm2 were used in the study. Measurements
`conducted after the experiments displayed similar values
`and confirmed the integrity of the monolayers during all
`of the experiments. No tendency towards an effect on
`TEER was observed under the various experimental
`conditions (pH, substrates and inhibitors).
`
`P-glycoprotein expression and activity
`Immunoblotting and transepithelial transport of P-gp probe
`The Western blot analysis revealed a C219 antibody-
`reactive band of 170 kDa corresponding to P-gp expres-
`sion (see [11]). The P-gp probe rhodamine 123 (5 lM)
`[16] showed a positive interaction with P-glycoprotein in
`our Caco-2 cell monolayers, with a secretory transport
`markedly inhibited by GF120918 (4 lM) ((Papp(B–A):
`
`7.1 ± 0.5.10)6 vs. 1.3 ± 1.4.10
`)6, P < 0.05).
`
`Transepithelial transport of naloxone and
`naltrexone
`The bidirectional transport of naloxone and naltrexone
`(1, 50 and 100 lM) was investigated at pH 7.4/7.4 in
`the presence and absence of 4 lM GF120918. The lowest
`naloxone and naltrexone concentrations tested (1 lM)
`correspond,
`respectively,
`to six times
`the plasma
`
`ª 2009 The Authors Journal compilation ª 2009 Socie´ te´ Franc¸aise de Pharmacologie et de The´ rapeutique
`Fundamental & Clinical Pharmacology 23 (2009) 543–548
`
`Nalox1222
`Nalox-1 Pharmaceuticals, LLC
`Page 3 of 6
`
`

`

`546
`
`M. Kanaan et al.
`
`Control
`
`+ GF120918
`
`(I)
`
`0.12
`
`0.1
`
`0.08
`
`0.06
`
`0.04
`
`0.02
`
`0
`
`Transported quantity (µg/cm2/3 h)
`
`(II)
`
`0.08
`
`0.07
`
`0.06
`
`0.05
`
`0.04
`
`0.03
`
`0.02
`
`0.01
`
`0
`
`Transported quantity (µg/cm2/3 h)
`
`Control
`
`+ GF120918
`
`Figure 2 Transepithelial transport of naloxone (I) and naltrexone
`(II) across Caco-2 cell monolayers in the presence and absence of
`GF120918.
`Naloxone (I) and naltrexone (II) (1 lM) were added to the apical
`side (open columns) or the basolateral side (solid columns) of the
`monolayers in the presence (+GF120918) or absence (control) of
`4 lM of GF120918 at pH 7.4/7.4. Data are mean ± SD of three
`experiments. P < 0.05.
`
`antagonists are well absorbed compounds [19]. Our
`transepithelial bidirectional study indicates that passive
`diffusion seems to be the major mechanism of naloxone
`and naltrexone transmembrane transit.
`Experimental animal data suggest that naloxone may
`interact with P-glycoprotein. Naloxone has been shown
`to stimulate ATPase activity in the plasma membranes of
`the multidrug-resistant Chinese hamster ovary (CHO)
`cell line CR1R12, as well as in purified reconstituted P-gp
`liposomes, although only at very high concentrations
`
`concentration measured 2 min after an injection of
`0.4 mg naloxone (0.01 lg/mL) [17] and 45 times the
`peak plasma concentration (Cmax = 13.7 ng/mL) mea-
`sured after multiple oral doses of 50mg naltrexone [18].
`Naloxone and naltrexone showed no statistical differ-
`ence between apical-to-basolateral
`(Papp(A–B)) and
`basolateral-to-apical (Papp(B–A)) transport. The efflux
`ratio (TR = Papp(B–A)/Papp(A–B)) of the two molecules
`was between 1.1 and 1.3, indicating that naloxone and
`naltrexone are not actively transported by an efflux
`transporter. This was confirmed by the use of the P-gp
`inhibitor, GF120918, which failed to alter the observed
`ratio (Table I, Figure 2).
`
`D I S C U S S I O N
`
`The results of our study clearly show that neither
`naloxone nor naltrexone are P-gp substrates. Indeed, no
`significant efflux ratio was observed for the two mole-
`cules across the P-gp-overexpressing Caco-2 cell mono-
`layers, as their respective fluxes in the absorptive and
`secretory directions were equivalent at all concentrations
`tested. Moreover, a potent P-gp inhibitor, GF120918,
`had no effect on bidirectional flux, either by the
`inhibition of efflux transport or by the enhancement of
`absorptive flux (P > 0.05). The absorptive apparent
`permeability coefficients of naloxone and naltrexone
`)6) show otherwise that the two opioid
`(Papps > 10 · 10
`
`Table I Apparent permeability coefficients Papp (cm/s) of naloxone
`and naltrexone in the presence and absence of GF120918 at pH
`7.4/7.4.
`
`Drug
`
`concentration
`
`± P-gp inhibitor
`
`Papp (A–B)
`)6)
`(cm/s) (10
`pH 7.4/7.4
`
`Papp (B–A)
`
`(cm/s) (10)6)
`pH 7.4/7.4
`
`Naloxone
`1 lM
`+ 4 lM GF120918
`50 lM
`+ 4 lM GF120918
`100 lM
`+ 4 lM GF120918
`Naltrexone
`1 lM
`+ 4 lM GF120918
`50 lM
`+ 4 lM GF120918
`100 lM
`+ 4 lM GF120918
`
`25.4 ± 0.8
`
`22.2 ± 2.6
`
`28.0 ± 0.5
`
`28.7 ± 1.3
`
`28.4 ± 0.3
`
`26.6 ± 1.4
`
`15.1 ± 0.6
`
`13.7 ± 0.8
`
`14.6 ± 0.2
`
`14.7 ± 0.3
`
`13.9 ± 0.4
`
`19.8 ± 5.2
`
`29.2 ± 1.6
`
`30.7 ± 2.0
`
`34.1 ± 0.5
`
`33.8 ± 2.9
`
`31.4 ± 2.1
`
`33.0 ± 1.8
`
`19.7 ± 0.5
`
`19.9 ± 0.9
`
`20.0 ± 1.1
`
`20.1 ± 1.0
`
`17.5 ± 0.6
`
`17.7 ± 0.2
`
`Efflux ratio (TR)
`
`1.1 (no net efflux)
`
`1.3
`
`1.2 (no net efflux)
`
`1.1
`
`1.1 (no net efflux)
`
`1.2
`
`1.3 (no net efflux)
`
`1.4
`
`1.3 (no net efflux)
`
`1.3
`
`1.2 (no net efflux)
`
`0.8
`
`Values are the mean ± SD of three experiments. P < 0.05.
`
`ª 2009 The Authors Journal compilation ª 2009 Socie´ te´ Franc¸aise de Pharmacologie et de The´ rapeutique
`Fundamental & Clinical Pharmacology 23 (2009) 543–548
`
`Nalox1222
`Nalox-1 Pharmaceuticals, LLC
`Page 4 of 6
`
`

`

`Naloxone, naltrexone and P-glycoprotein
`
`547
`
`(>100 lM) [20]. Our assays at lower concentrations
`(1 and 50 lM) do not confirm these results. Naloxone also
`weakly inhibited vinblastine binding to the plasma
`membranes of multidrug-resistant CHO (B30) cells at
`high concentrations (100 lM) [21]. In another experi-
`mental study in P-gp expressing CHO cells [22], naloxone
`caused a decrease in P-glycoprotein phosphorylation.
`Naloxone seems indeed to interact with animal P-gp, but
`with very low affinity, mainly resulting in weak inhibi-
`tion of the efflux transporter. No data concerning the
`interaction of naltrexone with P-glycoprotein or any
`other ‘‘ATP-binding cassette’’ (ABC) or ‘‘solute-linked
`carriers’’ (SLC) transporter are available in the literature.
`Naloxone and naltrexone are two structurally related
`opioid antagonists, mainly metabolized in the liver into
`naloxone-3-glucuronide and 6-beta-naltrexol, respec-
`tively [2,5]. Naloxone and naltrexone show similar
`potencies when administered parenterally. However,
`after oral administration, naltrexone is thought to be
`significantly more potent and long-lasting than naloxone
`in antagonizing the central effects of opioids [23]. Indeed,
`after oral administration, naloxone bioavailability is less
`than 2% [7,8,24,25], whereas naltrexone is between 5
`and 40% [9,10]. The huge difference observed in
`bioavailability after oral administration renders oral
`naloxone useless in opioid and alcohol addiction treat-
`ment. It has, however,
`lead to the development and
`commercialization of combinations of naloxone and
`opioid agonists, such as pentazocine and buprenorphine,
`to avoid the illicit parenteral use of the opioid agonist
`[26,27], as well as oxycodone to prevent peripherally
`mediated constipation without
`interfering with the
`central analgesic effect [28].
`Naloxone and naltrexone show very similar liposolu-
`bility (LogPs: 1.5 and 1.4,
`respectively), however,
`naltrexone has a higher pKa than naloxone (8.4 vs.
`7.9) [29,30]. Hence, at gastrointestinal pH conditions
`(pH 5–8)
`[31] as well as at physiological pH,
`the
`percentage of the ionized form of naltrexone is greater
`than that of naloxone. Consequently,
`the physico-
`chemical properties of the two opioid antagonists do
`not favour a greater gastrointestinal permeability of
`naltrexone as compared to naloxone. Additionally, our
`experimental study showed an almost twofold greater
`naloxone permeability across Caco-2 cells compared to
`)6 vs.
`naltrexone (ex, at 1 lM; Papp(A–B): 25.4 ± 0.8.10
`)6, P < 0.05).
`15.1 ± 0.6.10
`Evidence largely implicates hepatic first-pass metabo-
`lism in the poor oral bioavailability of naloxone, based
`on studies investigating the systemic biodisposition of
`
`the drug after oral and intravenous administration
`[7,8,24,25,32]. However, other mechanisms such as
`P-glycoprotein or other efflux drug transporters may be
`involved in limiting its gastrointestinal absorption and
`thus oral bioavailability. Our results show that the poor
`oral bioavailability of naloxone is not linked to a limited
`gastrointestinal absorption of the drug.
`In conclusion, our study excludes the involvement of
`P-glycoprotein in the poor oral bioavailability of nalox-
`one as compared to naltrexone. Naloxone gastrointesti-
`nal absorption does not seem to be the major cause of its
`poor oral bioavailability. In fact, rapid and extensive
`metabolism of the opioid antagonist appears as the more
`plausible factor to be considered.
`
`C O N F L I C T S O F I N T E R E S T
`
`The authors state no conflict of interest.
`
`A C K N O W L E D G E M E N T
`
`This work was supported by the Department of Anes-
`thesiology, Pharmacology and Intensive care, Geneva
`University Hospitals, CH-1211 Geneva 14, Switzerland.
`
`R E F E R E N C E S
`
`1 Helm S., Trescot A.M., Colson J., Sehgal N., Silverman S. Opioid
`antagonists, partial agonists, and agonists/antagonists: the role
`of office-based detoxification. Pain Physician (2008) 11
`225–235.
`2 Weinstein S.H., Pfeffer M., Schor J.M., Indindoli L., Mintz M.
`Metabolites of naloxone in human urine. J. Pharm. Sci. (1971)
`60 1567–1568.
`3 Fishman J., Roffwarg H., Hellman L. Disposition of naloxone-
`7,8,3H in normal and narcotic-dependent men. J. Pharmacol.
`Exp. Ther. (1973) 187 575–580.
`4 Crabtree B.L. Review of naltrexone, a long-acting opiate
`antagonist. Clin. Pharm. (1984) 3 273–280.
`5 Porter S.J., Somogyi A.A., White J.M. Kinetics and inhibition of
`the formation of 6beta-naltrexol from naltrexone in human
`liver cytosol. Br. J. Clin. Pharmacol. (2000) 50 465–471.
`6 Bullingham R.E., McQuay H.J., Moore R.A. Clinical pharmaco-
`kinetics of narcotic agonist-antagonist drugs. Clin. Pharmaco-
`kinet. (1983) 8 332–343.
`7 Kreek M.J. Opioid antagonists and partial antagonists, in:
`Schuster C.R., Kuhar M.J. (Eds), Pharmacological aspects of drug
`dependence. Towards an integrated neurobehavioral approach,
`Springer-Verlag, Berlin Heidelberg, New York, 1996, p. 565.
`8 De Schepper H.U., Cremonini F., Park M.I., Camilleri M.
`Opioids and the gut: pharmacology and current clinical
`experience. Neurogastroenterol. Motil. (2004) 16 383–394.
`Review.
`
`ª 2009 The Authors Journal compilation ª 2009 Socie´ te´ Franc¸aise de Pharmacologie et de The´ rapeutique
`Fundamental & Clinical Pharmacology 23 (2009) 543–548
`
`Nalox1222
`Nalox-1 Pharmaceuticals, LLC
`Page 5 of 6
`
`

`

`548
`
`M. Kanaan et al.
`
`9 Meyer M.C., Straughn A.B., Lo M.W., Schary W.L., Whitney
`C.C. Bioequivalence, dose-proportionality, and pharmacokinet-
`ics of naltrexone after oral administration. J. Clin. Psychiatry
`(1984) 45 15–19.
`10 Wall M.E., Brine D.R., Perez-Reyes M. Metabolism and dispo-
`sition of naltrexone in man after oral and intravenous admin-
`istration. Drug Metab. Dispos. (1981) 9 369–375.
`11 Kanaan M., Daali Y., Dayer P., Desmeules J. Lack of interaction
`of the NMDA receptor antagonists dextromethorphan and
`dextrorphan with P-glycoprotein. Curr. Drug Metab. (2008) 9
`144–151.
`12 Amidon G.L., Lennernas H., Shah V.P., Crison J.R. A theoretical
`basis for a biopharmaceutic drug classification: the correlation
`of in vitro drug product dissolution and in vivo bioavailability.
`Pharm. Res. (1995) 12 413–420.
`13 Artursson P. Epithelial transport of drugs in cell culture. I: A
`model for studying the passive diffusion of drugs over intestinal
`absorptive (Caco-2) cells. J. Pharm. Sci. (1990) 79 476–482.
`14 Karlsson J., Kuo S.M., Ziemniak J., Artursson P. Transport of
`celiprolol across human intestinal epithelial (Caco-2) cells:
`mediation of secretion by multiple transporters including
`P-glycoprotein. Br. J. Pharmacol. (1993) 110 1009–1016.
`15 Polli J.W., Wring S.A., Humphreys J.E., Huang L., Morgan J.B.,
`Webster L.O., Serabjit-Singh C.S. Rational use of in vitro P-
`glycoprotein assays in drug discovery. J. Pharmacol. Exp. Ther.
`(2001) 299 620–628.
`16 Takano M., Hasegawa R., Fukuda T., Yumoto R., Nagai J.,
`Murakami T. Interaction with P-glycoprotein and transport of
`erythromycin, midazolam and ketoconazole in Caco-2 cells.
`Eur. J. Pharmacol. (1998) 358 289–294.
`17 Naloxone product information. http://www.documed.ch.
`18 Dunbar J.L., Turncliff R.Z., Dong Q., Silverman B.L., Ehrich
`E.W., Lasseter K.C. Single- and multiple-dose pharmacokinetics
`of long-acting injectable naltrexone. Alcohol Clin. Exp. Res.
`(2006) 30 480–490.
`19 Yee S. In vitro permeability across Caco-2 cells (colonic) can
`predict in vivo (small intestinal) absorption in man – fact or
`myth. Pharm. Res. (1997) 14 763–766.
`20 Rebbeor J.F., Senior A.E. Effects of cardiovascular drugs on
`ATPase activity of P-glycoprotein in plasma membranes and in
`purified reconstituted form. Biochim. Biophys. Acta. (1998)
`1369 85–93.
`
`21 Callaghan R., Riordan J.R. Synthetic and natural opiates
`interact with P-glycoprotein in multidrug-resistant cells. J. Biol.
`Chem. (1993) 268 16059–16064.
`22 Callaghan R., Riordan J.R. Collateral sensitivity of multidrug
`resistant cells to narcotic analgesics is due to effects on the
`plasma membrane. Biochim. Biophys. Acta. (1995) 1236
`155–162.
`23 Gonzalez J.P., Brogden R.N. Naltrexone. A review of its
`pharmacodynamic and pharmacokinetic properties and thera-
`peutic efficacy in the management of opioid dependence. Drugs
`(1988) 35 192–213.
`24 Fishman J., Roffwarg H., Hellman L. Disposition of naloxone-
`7,8,3H in normal and narcotic-dependent men. J. Pharmacol.
`Exp. Ther. (1973) 187 575–580.
`25 Foss J.F. A review of the potential role of methylnaltrexone in
`opioid bowel dysfunction. Am. J. Surg. (2001) 182 19S–26S.
`26 Baum C., Hsu J.P., Nelson R.C. The impact of the addition of
`naloxone on the use and abuse of pentazocine. Public Health
`Rep. (1987) 102 426–429.
`27 Simojoki K., Vorma H., Alho H. A retrospective evaluation of
`patients switched from buprenorphine (Subutex) to the bupr-
`enorphine/naloxone combination (Suboxone). Subst. Abuse
`Treat Prev. Policy (2008) 17 16.
`28 Meissner W., Leyendecker P., Mueller-Lissner S., et al. A
`randomised controlled trial with prolonged-release oral oxyco-
`done and naloxone to prevent and reverse opioid-induced
`constipation. Eur. J. Pain (2009) 13 56–64.
`29 Clark’s. Isolation and Identification of Drugs in pharmaceuti-
`cals, body fluids, and post-mortem material, The Parmaceutical
`Press, London, UK, 1986.
`30 Gerebtzoff G., Seelig A. In silico prediction of blood-brain
`barrier permeation using the calculated molecular cross-
`sectional area as main parameter. J. Chem. Inf. Model. (2006)
`46 2638–2650.
`31 Neuhoff S., Ungell A.L., Zamora I., Artursson P. pH-dependent
`bidirectional transport of weakly basic drugs across Caco-2
`monolayers: implications for drug-drug interactions. Pharm.
`Res. (2003) 20 1141–1148.
`32 Weinstein S.H., Pfeffer M., Schor J.M., Franklin L., Mintz M.,
`Tutko E.R. Absorption and distribution of naloxone in rats after
`oral and intravenous administration. J. Pharm. Sci. (1973) 62
`1416–1419.
`
`ª 2009 The Authors Journal compilation ª 2009 Socie´ te´ Franc¸aise de Pharmacologie et de The´ rapeutique
`Fundamental & Clinical Pharmacology 23 (2009) 543–548
`
`Nalox1222
`Nalox-1 Pharmaceuticals, LLC
`Page 6 of 6
`
`

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