throbber
1521-009X/47/7/690–698$35.00
`DRUG METABOLISM AND DISPOSITION
`U.S. Government work not protected by U.S. copyright
`
`https://doi.org/10.1124/dmd.118.085977
`Drug Metab Dispos 47:690–698, July 2019
`
`Pharmacokinetic Interaction between Naloxone and Naltrexone
`Following Intranasal Administration to Healthy Subjects
`
`Philip Krieter, C. Nora Chiang, Shwe Gyaw, Phil Skolnick, and Rebekah Snyder
`
`National Institutes of Health, National Institute on Drug Abuse, Bethesda, Maryland (P.K., C.N.C., S.G., P.S.); and Sekisui XenoTech,
`LLC, Kansas City, Kansas (R.S.)
`
`Received December 18, 2018; accepted April 11, 2019
`
`ABSTRACT
`
`Naloxone (17-allyl-4,5a-epoxy-3,14-dihydroxymorphinan-6-one HCl),
`a m-opioid receptor antagonist, is administered intranasally to re-
`verse an opioid overdose but its short half-life may necessitate
`subsequent doses. The addition of naltrexone [17-(cyclopropylmethyl)-
`4,5a-epoxy-3,14-dihydroxymorphinan-6-one], another m-receptor
`antagonist, which has a reported half-life of 3 1/2 hours, may extend
`the available time to receive medical treatment. In a phase 1 phar-
`macokinetic study, healthy adults were administered naloxone and
`naltrexone intranasally, separately and in combination. When ad-
`ministered with naloxone, the Cmax value of naltrexone decreased
`62% and the area under the concentration-time curve from time zero
`to infinity (AUC0–inf) decreased 38% compared with when it was
`given separately; lower concentrations of naltrexone were observed
`as early as 5 minutes postdose. In contrast, the Cmax and AUC0–inf
`values of naloxone decreased only 18% and 16%, respectively, when
`
`given with naltrexone. This apparent interaction was investigated
`further to determine if naloxone and naltrexone shared a transporter.
`Neither compound was a substrate for organic cation transporter
`(OCT) 1, OCT2, OCT3, OCTN1, or OCTN2. There was no evidence of
`the involvement of a transmembrane transporter when they were
`tested separately or in combination at concentrations of 10 and
`500 mM using Madin-Darby canine kidney II cell monolayers at
`pH 7.4. The efflux ratios of naloxone and naltrexone increased to six
`or greater when the apical solution was pH 5.5, the approximate
`pH of the nasal cavity; there was no apparent interaction when
`the two were coincubated. The importance of understanding how
`opioid antagonists are absorbed by the nasal epithelium is magnified
`by the rise in overdose deaths attributed to long-lived synthetic
`opioids and the realization that better strategies are needed to treat
`opioid overdoses.
`
`Introduction
`
`Opioid overdose in the United States led to 750,000 emergency
`department visits and more than 49,000 opioid-related deaths in 2017
`(https://www.drugabuse.gov/related-topics/trends-statistics/overdose-
`death-rates). The use of the antagonist naloxone (17-allyl-4,5a-epoxy-
`3,14-dihydroxymorphinan-6-one HCl) has been endorsed by multiple
`government agencies to limit opioid-induced fatalities (https://
`www.surgeongeneral.gov/priorities/opioid-overdose-prevention/
`naloxone-advisory.html). Improvised naloxone kits for intrana-
`sal administration have been promoted for use by nonmedical
`personnel and the general public to counteract opioid overdoses
`(Carpenter et al., 2016); however, approximately one-half of subjects
`in a human use study could not assemble and use the device without
`proper training (Edwards et al., 2015). In 2015, the U.S. Food and
`Drug Administration approved Narcan, an intranasal device that
`
`This work was supported by the National Institutes of Health National Institute
`on Drug Abuse [Contracts N01DA-12-8905, N01DA-13-8920, and N01DA-14-
`8914].
`P.K. and S.G. are employees of the National Institutes of Health (NIH). C.N.C. is
`a former NIH employee. P.S. is an employee of Opiant Pharmaceuticals; he was
`employed by the NIH when this study was designed and executed. R.S. is an
`employee of Sekisui XenoTech LLC.
`https://doi.org/10.1124/dmd.118.085977.
`
`delivers 4 mg naloxone in a volume of 0.1 ml. Ninety percent of
`subjects were able to use it correctly without any training; it also
`produces plasma concentrations as rapidly as an intramuscular
`injection (Krieter et al., 2016). Due to its short half-life, naloxone
`may be effective for 1 hour or less, and depending on the quantity and
`nature of the opioid ingested the person could relapse into respiratory
`depression (Li et al., 2018) before trained medical personnel respond.
`Naltrexone [17-(cyclopropylmethyl)-4,5 a-epoxy-3,14-
`dihydroxymorphinan-6-one], a m-opioid receptor antagonist, has a
`reported half-life of approximately 3 1/2 hours (Yuen et al., 1999) and
`has a 5-fold higher affinity for the receptor compared with naloxone
`(Cassel et al., 2005). While the duration of occupancy of naloxone on the
`m-receptor has a half-life of 2 hours (Kim et al., 1997), naltrexone has a
`half-life duration of 72 hours (Lee et al., 1988). This is longer than the
`plasma half-life of naltrexone and its major metabolite 6b-naltrexol
`(Meyer et al., 1984), suggesting it remains on the receptor longer than
`indicated by the plasma concentrations. Combining it with naloxone
`may increase the window for response to an opioid overdose.
`An initial study demonstrated that naltrexone can be absorbed after
`nasal administration of 2 mg in 1 ml of water; the Cmax value was
`3.86 ng/ml at 0.38 hours (Brown et al., 2014). When a crushed extended-
`release oxycodone tablet containing 3.6 mg naltrexone was administered
`intranasally, the maximum concentration of the antagonist was 4.4 ng/ml
`at 0.3 hours (Setnik et al., 2015).
`
`ABBREVIATIONS: AUC0–inf, area under the concentration-time curve from time zero to infinity; D-PBS, PBS containing 0.2% bovine serum
`albumin; HEK293, human embryonic kidney 293; LC-MS/MS, liquid chromatography–tandem mass spectrometry; MDCKII, Madin-Darby canine
`kidney II; m/z, mass-to-charge ratio; naloxone, 17-allyl-4,5a-epoxy-3,14-dihydroxymorphinan-6-one HCl; naltrexone, 17-(cyclopropylmethyl)-4,5a-
`epoxy-3,14-dihydroxymorphinan-6-one; OCT, organic cation transporter; Papp, apparent permeability; PK, pharmacokinetic.
`
`690
`
`Nalox1225
`Nalox-1 Pharmaceuticals, LLC
`Page 1 of 9
`
`

`

`PK of Naloxone and Naltrexone with Intranasal Administration
`
`691
`
`While naltrexone is indicated for the prevention of relapse to opioid
`dependence, it has not been evaluated for the reversal of an opioid
`overdose. A pilot study was conducted to assess the feasibility of
`combining naltrexone with naloxone as an intranasal formulation to
`lengthen the time for the reversal of opioid-induced respiratory de-
`pression in emergency situations. The results demonstrate reductions in
`plasma concentrations when the two drugs were combined relative to
`when they were given separately; the reduction in naltrexone concen-
`trations was more pronounced than for naloxone. In vitro studies were
`conducted to determine the nature of the interaction. This report details
`the results from both the clinical and in vitro studies.
`
`Pharmacokinetic Study
`
`Materials and Methods
`
`Study Participants. The clinical study was conducted by Vince & Associates
`Clinical Research (Overland Park, KS). The study was approved by the MidLands
`Independent Review Board (Overland Park, KS); all subjects gave written,
`informed consent before participation. The study was carried out in accordance
`with the 1996 International Conference on Harmonization for Good Clinical
`Practices Guidelines (http://www.ich.org/fileadmin/Public_Web_Site/
`ICH_Products/Guidelines/Efficacy/E6/E6_R1_Guideline.pdf). The study was
`registered on ClinicalTrials.gov as NCT03851731.
`Healthy male and female volunteers aged 18–55 years, with body mass index
`of 18–30 kg/m2, participated in the pharmacokinetic (PK) study. Participants were
`currently not taking either prescription or over-the-counter medications, and
`nonsmokers or those who smoked 20 or fewer cigarettes per day were enrolled.
`Screening procedures conducted within 21 days of study initiation included the
`following: medical history, physical examination, evidence of nasal irritation,
`12-lead ECG, complete blood count, clinical chemistry, coagulation markers,
`hepatitis and human immunodeficiency screening, urinalysis, and urine drug
`screen. Female participants were tested for pregnancy at screening and
`admission to the clinic. Participants were excluded if they had abnormal nasal
`anatomy or symptoms (e.g., runny nose, nasal polyps), an upper respiratory
`tract infection, used opioid analgesics for pain relief within the previous
`14 days, or in the judgment of the investigator had significant acute or chronic
`medical conditions. Participants were required to abstain from grapefruit juice
`and alcohol 72 hours prior to admission to the end of the last blood draw of the
`study. On days of dosing, a participant’s vital signs were required to be within the
`acceptable range before receiving naloxone, defined as: systolic blood pres-
`sure .90 and #140 mm Hg; diastolic blood pressure .55 and #90 mm Hg;
`resting heart rate .40 and #100 beats per minute; and respiratory rate .8
`and #20 respirations per minute.
`Study Design. This was an inpatient, double-blind, randomized, three-period,
`three-treatment, six-sequence, crossover study. Participants were randomly
`assigned to one of six possible sequences. On the day after clinic admission,
`participants were administered the study drugs in randomized order with a 4-day
`washout period between doses. Participants remained in the clinic for 13 days
`until all three treatments were administered; they received a follow-up phone call
`3–5 days after discharge. They fasted from midnight before each dosing day until
`1 hour after dose administration. Participants refrained from smoking and
`caffeine-containing drinks for 1 hour before until 2 hours after dosing. They
`received one of the following three treatments in one nostril:
`
`Treatment 1: Naltrexone (2 mg), intranasally; (one 0.1 ml spray of a
`20 mg/ml formulation);
`Treatment 2: Naloxone (4 mg), intranasally; (one 0.1 ml spray of a
`40 mg/ml formulation); or
`intranasally
`Treatment 3: Naltrexone (2 mg) and naloxone (4 mg),
`(one 0.1 ml spray of the 20 mg/ml naltrexone plus 40 mg/ml naloxone
`formulation).
`
`The study drugs were administered in the supine position, and subjects remained
`in this position for approximately 1 hour after dosing. Participants were instructed
`not to breathe when the drug was administered to simulate an opioid overdose
`with a patient in respiratory arrest. Twelve-lead ECGs were collected predose and
`at 1 and 6 hours postdose. Venous blood samples (4 ml) were collected for the
`analyses of plasma naloxone and naltrexone concentrations at predose; 2.5, 5, 10,
`
`15, 20, 30, 45, and 60 minutes; and 2, 3, 4, 6, 8, 12, 24, 30, 36, 48, 60, and 72 hours
`postdose using Vacutainer tubes containing sodium heparin. The plasma was
`stored at , 260°C until analyzed.
`Study Drugs. Naltrexone HCl and naloxone HCl powders were purchased
`from Mallinckrodt, Inc. (St. Louis, MO) and were current Good Manufacturing
`Practices grade (https://www.fda.gov/drugs/pharmaceutical-quality-resources/
`current-good-manufacturing-practice-cgmp-regulations). The formulations were
`made by the pharmacists at Vince & Associates Clinical Research; sterile water
`for injection was the vehicle for both compounds. The study drugs were
`administered using a LMA mucosal atomization device (Teleflex Medical Europe
`Ltd., Athione, Ireland) and a 1-ml disposable syringe. The syringes and devices
`were weighed before and after dose administration. Based on the dose analysis
`and weight of the dose administered, the mean 6 S.D. of milligrams administered
`were the following: treatment A, 2.24 6 0.03 mg naltrexone HCl; treatment B,
`4.58 6 0.05 mg naloxone HCl; and treatment C, 2.27 6 0.07 mg naltrexone HCl
`plus 4.62 6 0.15 mg naloxone HCl.
`Analytical Methods. Plasma naloxone concentrations were assayed as
`described previously (Krieter et al., 2016); the lower limit of quantitation was
`0.01 ng/ml. The interday precision of the calibration curves and quality control
`samples ranged from 3.22% to 9.05% and the accuracy ranged between 23.14%
`and 5.33% during the analysis of the samples.
`Plasma naltrexone and 6b-naltrexol concentrations were determined using a
`validated liquid chromatography–tandem mass spectrometry (LC-MS/MS) assay
`by XenoBiotic Laboratories (Plainsboro, NJ). Plasma samples (0.15 ml) were
`mixed with 0.1 ml of 1% formic acid in water and 0.05 ml of acetonitrile:water
`(2:8) containing the internal standards (0.5 ng naltrexone-d3 and 0.25 ng
`6b-naltrexol-d3) and added to individual wells of a preconditioned 96-well plate.
`The plate was washed sequentially with 1% formic acid in water, water, methanol:
`water (1:1), and methanol. The analytes were eluted using 4% ammonium
`hydroxide in methanol. After evaporation, the residue was dissolved in 0.15 ml
`methanol:0.1% formic acid (8:92) and submitted to LC-MS/MS analysis. The AB
`MDS Sciex API-5000 LC-MS/MS system (Framingham, MA) with an atmo-
`spheric pressure chemical ionization source was operated in the positive ion
`detection mode. The mobile phase consisted of a gradient from 93% mobile phase
`A (10 mM ammonium formate, pH 4.0)/7% mobile phase B [acetonitrile:
`methanol (2:8)] to 80% mobile phase A/20% mobile phase B over 1.7 minutes at a
`flow rate of 0.5 ml/min through a 2.1  50 mm Kinetex EVO C18 2.6 mm column
`(Phenomenex, Torrance, CA). Naltrexone eluted at approximately 1.45 minutes;
`ions monitored had mass-to-charge ratios (m/z) 342.2 and 324.2 for naltrexone
`and 345.2 and 327.3 for its internal standard. 6b-Naltrexol eluted at approx-
`imately 1.60 minutes; ions monitored had m/z 344.2 and 326.2 for 6b-naltrexol
`and 347.1 and 329.3 for its internal standard. The interday precision of the
`calibration curves and quality control samples for naltrexone ranged from 2.92%
`to 7.87%, and the accuracy ranged between 23.50% and 0.75% during the
`analysis of the samples. The interday precision of the calibration curves and
`quality control samples of 6b-naltrexol ranged from 2.89% to 7.38%, and the
`accuracy ranged between 27.13% and 2.00% during the analysis of the samples.
`The lower limit of quantification for both naltrexone and 6b-naltrexol was
`0.02 ng/ml.
`Data Analyses. The safety population included all subjects who received at
`least one intranasal dose; the PK population included all participants who received
`at least one dose with sufficient data to calculate meaningful PK parameters. The
`PK parameters were calculated using standard noncompartmental methods and
`a validated installation of WinNonlin Phoenix, version 6.3 (Cetera, Princeton,
`NJ). Values of peak plasma concentrations (Cmax) and the time to reach Cmax were
`the observed values obtained directly from the concentration-time data. The
`terminal elimination half-life was estimated by linear regression analysis. The area
`under the concentration-time curve from time zero to the last quantifiable
`concentration was determined by the linear-up/log-down trapezoidal method
`and extrapolated to the area under the concentration-time curve from time zero to
`infinity (AUC0–inf) by adding the value of the last quantifiable concentration
`divided by the terminal rate constant. Since the extrapolated area under the plasma
`concentration-time curve was less than 20% for all participants, only AUC0–inf is
`reported. The apparent total body clearance was calculated as the dose divided by
`AUC0–inf. Within an ANOVA framework, comparisons of ln-transformed PK
`parameters were performed using a mixed-effects model, where sequence, period,
`and treatment were the independent factors. The 90% confidence intervals for the
`ratio of the geometric least-squares mean values of Cmax and AUC0–inf were
`
`Nalox1225
`Nalox-1 Pharmaceuticals, LLC
`Page 2 of 9
`
`

`

`692
`
`Krieter et al.
`
`constructed for comparison of the two drugs administered in combination versus
`separately. The 90% confidence intervals were obtained by exponentiation of the
`90% confidence intervals for the differences between the least-squares mean values
`based on a ln scale. All analyses of demographic and safety data were performed
`using SAS statistical software, version 9.3 (SAS Institute, Inc., Cary, NC).
`
`In Vitro Transporter Studies
`
`Methods. Transporter studies with organic cation transporter (OCT) 1, OCT2,
`and OCT3 were conducted using human embryonic kidney 293 (HEK293) cells
`(ATCC, Manassas, VA) that had been transfected with vectors containing human
`transporter cDNA; control cells were transfected with the vector only. The culture
`medium was Dulbecco’s modified Eagle’s medium supplemented with 8.9%
`FBS, 0.89% antibiotic, and 1.79 mM L-glutamine. The incubation medium for the
`HEK293 cells was Hanks’ balanced salt solution containing 10 mM HEPES,
`pH 7.4. OCTN1 and OCTN2 transporter studies used S2 cells established by
`culturing microdissected S2 segments derived from transgenic mice harboring the
`temperature-sensitive simian virus 40 large T-antigen gene. Cells were transfected
`with vectors containing the human transporter cDNA. Control cells for all five
`transporters contained only the vector cDNA. The culture medium for the S2 cells
`was RITC80-7 supplemented with 4.7% FBS, 9.3 mg/ml epidermal growth factor,
`0.08 U insulin/ml, and 9.5 mg/l transferrin. PBS containing 0.2% bovine serum
`albumin (D-PBS), pH 7.4, was used as the incubation medium for the S2 cells. All
`cells were cultured with 5% CO2 and 95% relative humidity at 33°C (S2 cells) or
`37°C (HEK293 cells).
`The medium was removed by aspiration, the cells were rinsed with either 1 ml
`of Hanks’ balanced salt solution or D-PBS (this was replaced with medium
`containing the control inhibitor or solvent control), and then the cells were
`preincubated. After preincubation, the medium was removed and replaced with
`0.3 ml buffer containing either the test article or the positive control. The final
`concentration of naloxone and naltrexone was 1 mM in 0.2% DMSO, the solvent
`for the test articles and controls. Cells were incubated for 2, 5, 10, or 20 minutes at
`37°C, at which time the solutions were removed. Positive controls were 10 mM
`[14C]metformin (OCT1 and OCT2), 5 mM [14C]triethylamine (OCT3 and
`OCTN1), and 0.03 mM [3H]carnitine (OCTN2). All incubations were done in
`triplicate. HEK293 cells were washed one time with 1 ml of ice-cold D-PBS and
`twice with 1 ml of ice-cold PBS; S2 cells were washed three times with ice-cold
`D-PBS. After removal of the medium, 0.5 ml of purified water was added to each
`well to lyse the cells and samples were collected for analysis of naloxone and
`naltrexone and then analyzed by LC-MS/MS. Samples were mixed with 20 ml
`water with the internal standard (nalmefene). An AB Sciex API-4000 mass
`spectrometer was operated in the positive mode with an Acquity UPLC BEH C18
`analytical column (2.1  50 mm, 1.7 mm). The mobile phase consisted of a
`gradient from 95% mobile phase A (10 mM ammonium acetate)/5% mobile phase
`B (10 mM ammonium acetate in acetonitrile with 0.1% ammonium hydroxide)
`to 10% mobile phase A/90% mobile phase B in 2 minutes. The flow rate was
`0.5 ml/min. The ions monitored had m/z 328.2 and 310.0 for naloxone, m/z 342.2
`and 324.1 for naltrexone, and m/z 340.3 and 322.3 for the internal standard.
`Concentrations were calculated as the area under the curve compared with that of
`the internal standard using known concentrations of naloxone and naltrexone. For
`the positive controls, a 0.3 ml aliquot of the cell lysate was mixed with 5 ml of
`scintillation cocktail, and radioactivity was measured by liquid scintillation
`
`counting. Samples were collected for protein content using the BCA-Protein
`Assay Kit (Thermo Fisher Scientific, Waltham, MA). The uptake amount and
`cleared volume of naloxone and naltrexone were calculated as follows:
`Uptake  amount  into  cells  ðpmol=wellÞ ¼ pmol  in  cell  lysate
` 500  ml=1000  ml
`
`Cleared  volume  ðml=mg  proteinÞ ¼
`uptake  amount  into  cells  ðpmol=wellÞ
`mg  protein=well  initial  concentration  ðpmol=mlÞ
`
`For positive controls, disintegrations per minute were substituted for picomoles
`in the previous equations.
`Transport studies were also conducted using wild-type Madin-Darby canine
`kidney II (MDCKII) cells that had been transfected with vectors containing
`human transporter cDNA (Netherlands Cancer Institute, Amsterdam, Netherlands).
`Cells were plated and maintained on 24-well Transwell plates (Corning,
`Corning, NY) for 3–5 days prior to the experiment. Culture medium was
`removed and incubation medium (Hanks’ balanced salt solution supplemented
`with 25 mM HEPES and 25 mM glucose) was added to the cells. The pH of
`the basolateral buffer was 7.4 and that of the apical buffer was either pH 7.4
`or 5.5. Approximately 10 minutes after incubation medium was added, the
`transepithelial electrical resistance was recorded and cells were preincubated at
`37°C for 30–60 minutes. After preincubation, the medium containing naloxone,
`naltrexone, or control compounds ([3H]mannitol and [14C]caffeine) was added to
`the donor chamber. Samples were collected from the receiver side at 15, 60, and
`120 minutes and replaced by 0.1 ml of incubation medium. The transepithelial
`electrical resistance was also measured at the end of the incubation to determine if
`the cells were still confluent. Samples were mixed with 25 ml of methanol:water
`(1:1 v/v) and 75 ml of the internal standard (hydroxybuproprion-d6) in methanol:
`water (1:1 v/v) and analyzed by LC-MS/MS using an AB Sciex API-5500 mass
`spectrometer as described previously, except that the gradient changed from 70%
`mobile phase A/30% mobile phase B to 5% mobile phase A/95% mobile phase B
`over 3 minutes. Ions for the internal standard were monitored at m/z 262.0 and
`244.0. Concentrations of radioactivity were determined as detailed previously.
`The apparent permeability (Papp) was calculated as follows:
`
`
`1
`A0  C0
`where dQ is the amount of test drug transported (in picomoles); dT is the
`incubation time (in seconds); A0 is the surface area of the membrane (in squared
`centimeters); and C0 is the initial concentration of the test drug in the donor
`chamber (in picomoles per cubic centimeter). The efflux ratio was calculated as
`the Papp basal-to-apical/Papp apical-to-basal ratio. The sex of the cell lines used in
`the experiments is unknown.
`
`dQ
`dT
`
`Results
`
`Pharmacokinetic Study
`Subject Characteristics. All subjects initiating the study (Table 1)
`received at least one dose of naloxone and/or naltrexone; 11 subjects
`
`TABLE 1
`Pharmacokinetics of naloxone: subject demographics
`
`Demographics
`
`All
`
`Male
`
`Number
`Mean age, y (range)
`Race
`White
`Black/African American
`Ethnicity
`Hispanic or Latino
`Not Hispanic or Latino
`Mean Weight, kg (range)
`Mean BMI, kg/m2 (range)
`
`BMI, body mass index.
`
`12
`36.0 (22.0–48.0)
`
`6
`39.2 (26.0–48.0)
`
`4
`8
`
`1
`11
`74.7 (49.4–99.2)
`25.0 (19.2–29.3)
`
`1
`5
`
`0
`6
`82.7 (71.8–99.2)
`25.3 (23.6–27.2)
`
`Female
`
`6
`32.8 (22.0–48.0)
`
`3
`3
`
`1
`5
`66.6 (49.4–84.8)
`24.6 (19.2–29.3)
`
`Nalox1225
`Nalox-1 Pharmaceuticals, LLC
`Page 3 of 9
`
`

`

`PK of Naloxone and Naltrexone with Intranasal Administration
`
`693
`
`When naloxone was added to the naltrexone intranasal formulation,
`the Cmax value decreased from 4.55 to 1.71 ng/ml, a decline of
`approximately 62% (Fig. 2; Table 2), and the AUC0–inf value decreased
`approximately 38%. The median time to reach Cmax value increased
`from 0.33 to 0.75 hours when the combination formulation was
`administered compared with naltrexone alone. However, the half-life
`of naltrexone was unchanged.
`The decreased concentration of naltrexone concentrations in treatment
`C was evident even at 5 and 10 minutes after dose administration; the
`concentrations were 83%–86% lower than when naltrexone was dosed
`alone. The concentrations of naltrexone continued to be considerably
`lower even at 6 hours postdose. In contrast, there was no change in the
`PK values of 6b-naltrexol when naltrexone was administered with or
`without naloxone (Fig. 3; Tables 2 and 3). The two formulations were
`bioequivalent for 6b-naltrexol, based on the 90% confidence intervals of
`Cmax and AUC0–inf (Table 3).
`There were minor differences in the PK parameters between males
`and females (Table 4). However, the small sample size of this pilot study
`precludes any definitive conclusions regarding sex-related differences
`following intranasal administration of either drug.
`Safety. Five subjects experienced at least one adverse event of any
`grade or attribution that was judged to be related to the test drugs; all
`were mild in severity. Headache was the single most frequent adverse
`event (three events in each of three subjects). There was one drug-related
`incident of mild inflamed mucosa (score of 1) that occurred 24 hours
`after dosing with 2 mg naltrexone. Vital signs, ECG, and clinical
`laboratory parameters did not reveal any clinically significant changes
`after any of the doses.
`
`In Vitro Transporters
`The ratios of naloxone and naltrexone uptake by the five transporter-
`expressing cell lines compared with the control cells were all less than 2,
`indicating that neither compound was a substrate for OCT1, OCT2,
`OCT3, OCTN1, or OCTN2 (Table 5). Positive controls had ratios of
`uptake that ranged from 6.4 for OCT3 to 75.4 for OCTN2 and
`demonstrated inhibition of uptake by their respective inhibitor.
`Permeability of naloxone and naltrexone across a polarized cell layer
`was tested using control MDCKII cells. Concentrations on the donor
`side were either 10 or 500 mM. Transporter studies normally use buffers
`that are pH 7.4 on both the apical and basolateral sides. Since the pH of
`the nasal passage is approximately pH 5.5–6.5,
`the studies were
`conducted also with the apical buffer at pH 5.5, while the basolateral
`buffer remained at pH 7.4.
`When the pH of the buffer was pH 7.4 on both sides, the efflux ratios
`of both naloxone and naltrexone were less than 2 at concentrations of
`
`Fig. 1. Mean (S.D.) concentrations of naloxone in healthy participants following
`intranasal administration of 4 mg naloxone alone and in combination with 2 mg
`naltrexone. Upper graph: concentrations to 12 hours postdose; Lower graph:
`concentrations to 1 hour postdose.
`
`completed the study. One female subject withdrew during the first period
`due to a moderate headache with mild nausea that occurred 28 hours
`after administration of 2 mg naltrexone.
`Pharmacokinetics. The geometric mean Cmax and AUC0–inf values
`of naloxone following a 4-mg intranasal dose were 4.30 ng/ml and
`8.13 ng·h/ml, respectively (Fig. 1; Table 2). The Cmax and AUC0–inf
`values decreased by approximately 18% and 16 %, respectively, when
`naloxone was administered in combination with 2 mg naltrexone
`(Table 3). The median time to reach Cmax value remained unchanged
`at 30 minutes, and the elimination half-life was also unchanged.
`
`TABLE 2
`Geometric mean pharmacokinetic parameters (%CV) of naloxone, naltrexone, and 6b-naltrexol
`
`Variablea (U)
`
`Naloxone
`
`Naltrexone
`
`6b-Naltrexol
`
`Alone (Treatment B)
`
`Plus Naltrexone (Treatment C) Alone (Treatment A)
`
`Plus Naloxone (Treatment C) Alone (Treatment A)
`
`Plus Naloxone (Treatment C)
`
`N
`Cmax (ng/ml)
`Tmax (h)
`AUC0–inf (ng·h/ml)
`lz (h21)
`t1/2 (h)
`CL/F (l/min)
`
`11
`4.30 (47.5)
`0.50 (0.25–0.75)
`8.13 (38.2)
`0.380 (32.1)
`1.83 (32.1)
`6.71 (38.2)
`
`11
`3.60 (36.5)
`0.50 (0.25–0.75)
`7.00 (32.5)
`0.355 (43.9)
`1.95 (45.7)
`7.79 (43.9)
`
`12
`4.55 (80.0)
`0.33 (0.17–1.0)
`9.61 (39.1)
`0.319 (18.2)
`2.17 (18.2)
`3.12 (39.1)
`
`11
`1.71 (35.1)
`0.75 (0.25–2.0)
`5.88 (25.2)b
`0.322 (10.3)b
`2.15 (10.3)b
`5.10 (25.2)b
`
`12
`2.09 (32.5)
`2.00 (0.75–4.0)
`30.8 (32.7)
`0.0433 (31.6)
`16.0 (31.6)
`NC
`
`11
`2.09 (26.2)
`2.00 (0.75–3.0)
`28.3 (28.4)
`0.0430 (24.5)
`16.1 (24.5)
`NC
`
`AUC0–inf, area under the plasma concentration-time curve from time zero to infinity; CL/F, apparent clearance; lz, terminal phase rate constant; NC, not calculated; t1/2, terminal phase half-life;
`Tmax, time to reach Cmax; Treatment A, 2 mg naltrexone, intranasally; Treatment B, 4 mg naloxone, intranasally; Treatment C, 2 mg naltrexone plus 4 mg naloxone, intranasally.
`aGeometric mean (%CV) for all except median (range) for Tmax.
`bN = 10.
`
`Nalox1225
`Nalox-1 Pharmaceuticals, LLC
`Page 4 of 9
`
`

`

`694
`
`Krieter et al.
`
`TABLE 3
`Statistical summary of treatment comparisons
`
`Variable
`
`Analyte
`
`Comparison (Treatment
`C vs. Reference)
`
`Geometric Mean Ratio
`(Treatment C/Reference)
`
`Cmax (ng/ml)
`
`AUC0–inf (ng·h/ml)
`
`Naloxone
`Naltrexone
`6b-Naltrexol
`Naloxone
`Naltrexone
`6b-Naltrexol
`
`C vs. B
`C vs. A
`C vs. A
`C vs. B
`C vs. A
`C vs. A
`
`81.5
`38.4
`101
`84.6
`61.6
`94.4
`
`90% CI
`
`63.6–105
`25.7–57.3
`92.7–110
`70.3–102
`50.9–74.6
`89.3–99.8
`
`AUC0–inf, area under the plasma concentration-time curve from time zero to infinity; CI, confidence interval; Treatment A, 2 mg naltrexone, intranasally; Treatment
`B, 4 mg naloxone, intranasally; Treatment C, 2 mg naltrexone plus 4 mg naloxone, intranasally.
`
`10 and 500 mM (Tables 6 and 7). The addition of 50-fold higher
`concentration of naloxone to both the lower and higher naltrexone
`donor solutions did not reduce the efflux ratio to an appreciable
`amount. Similar results were observed when the higher concentration
`of naltrexone was added to the naloxone solutions.
`Lowering the pH of the apical buffer to pH 5.5, while maintaining the
`basolateral at pH 7.4, caused a 3- to 5-fold decrease in Papp values of
`naltrexone in the A-to-B ratio and 2- to 4-fold increase in the efflux
`direction (Table 6). The efflux ratios increased to between 12.1 and 18.2.
`Similar results were observed using naloxone (Table 7). The Papp values
`changed considerably regardless of whether naloxone and naltrexone
`were tested separately or in combination.
`The mean transepithelial electrical resistance values were above
`100 V  cm2 both pre- and postdose in all of the MDCKII studies. The
`Papp values of [3H]mannitol were in the range of 0.41–1.37  10–6 cm/s
`in the A-to-B and B-to-A directions, while they ranged between 15.5 and
`53.5  10–6 cm/s for [14C]caffeine in all of the incubations.
`
`Discussion
`
`Delivery of naloxone by the nasal route has been recognized by the
`medical community and public officials as an effective way to reverse
`opioid overdoses (Ryan and Dunne, 2018). However, the short half-life
`of naloxone and the increased incidence of overdoses linked to synthetic
`opioids with a longer duration of action may require more than one dose
`to be administered to prevent renarcotization (Klebacher et al., 2017).
`Therefore, the addition of a longer-acting opioid antagonist to the
`naloxone formulation was initially hypothesized as a means to increase
`the time to obtain proper medical attention.
`The large decrease in the nasal absorption of naltrexone in the
`presence of naloxone was unexpected. It was observed as early as
`5 minutes following administration of both opioid antagonists but was
`far more pronounced for naltrexone. The Cmax value of naltrexone was
`reduced to 1.7 ng/ml when the drugs were combined, i.e., less than
`2 ng/ml, which is generally regarded as a concentration that
`is
`sufficient to adequately block the effects of opioid agonists (Comer
`et al., 2006). The combination product of naloxone and naltrexone
`was not pursued further.
`Although the concentration of naltrexone was below the target of
`2 ng/ml, direct absorption into the brain via the olfactory nerves that
`protrude through the cribiform plate in the olfactory epithelium may lead
`to a higher concentration at the site of action (Illum, 2000). The cerebral
`spinal fluid/plasma ratio of zidovudine was higher after intranasal
`administration compared with an intravenous infusion at 15 minutes
`postdose in rats (Seki et al., 1994). Similar results were observed in the
`rat using cephalexin (Sakane et al., 1994). However, the olfactory
`epithelium accounts for only 3%–5% of the nasal cavity’s total surface
`area, which may limit its role in the efficient transfer of drugs directly to
`the central nervous system (Grassin-Delyle et al., 2012).
`
`Rabiner et al. (2011) hypothesized that 6b-naltrexol contributed to
`the long occupancy of the m-opioid receptor due to its long plasma
`half-life. Due to this possibility, the Food and Drug Administration
`draft guidance on new formulations of naltrexone hydrochloride requires
`the analysis of 6b-naltrexol (https://www.fda.gov/downloads/Drugs/
`GuidanceComplianceRegulatoryInformation/Guidances/UCM194641.pdf).
`This metabolite, while approximately 2-fold less potent than the parent
`compound on the m-receptor, is 100-fold less potent than naltrexone
`in vivo in nonhuman primates (Ko et al., 2006) and has no effect on
`either morphine-induced analgesia or pupil constriction in humans
`(Yancey-Wrona et al., 2011). Thus, 6b-naltrexol
`is peripherally
`restricted; therefore, its involvement in the central nervous system
`effects of naltrexone is minimal.
`
`Fig. 2. Mean (S.D.) conce

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