throbber
Vol. 29 No. 3 March 2005
`
`Journal of Pain and Symptom Management 297
`
`Review Article
`
`Buprenorphine: Considerations
`for Pain Management
`Rolley E. Johnson, PharmD, Paul J. Fudala, PhD, and Richard Payne, MD
`Department of Psychiatry and Behavioral Sciences, Behavioral Pharmacology Research Unit (R.E.J.),
`Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Psychiatry
`(P.J.F.) and Behavioral Health Service (P.J.F.), University of Pennsylvania School of Medicine,
`VA Medical Center, Philadelphia, Pennsylvania; and Department of Neurology (R.P.), Memorial
`Sloan-Kettering Cancer Center, New York, New York, USA
`
`Abstract
`New effective analgesics are needed for the treatment of pain. Buprenorphine, a partial mu-
`opioid agonist which has been in clinical use for over 25 years, has been found to be
`amenable to new formulation technology based on its physiochemical and pharmacological
`profile. Buprenorphine is marketed as parenteral, sublingual, and transdermal
`formulations. Unlike full mu-opioid agonists, at higher doses, buprenorphine’s physiological
`and subjective effects, including euphoria, reach a plateau. This ceiling may limit the
`abuse potential and may result in a wider safety margin. Buprenorphine has been used for
`the treatment of acute and chronic pain, as a supplement to anesthesia, and for behavioral
`and psychiatric disorders including treatment for opioid addiction. Prolonged use of
`buprenorphine can result in physical dependence. However, withdrawal symptoms appear
`to be mild to moderate in intensity compared with those of full mu agonists. Overdoses
`have primarily involved buprenorphine taken in combination with other central nervous
`J Pain Symptom Manage 2005;29:297–326. 쑖 2005 U.S. Cancer
`system depressants.
`Pain Relief Committee. Published by Elsevier Inc. All rights reserved.
`
`Key Words
`Buprenorphine, pharmacology, pharmacodynamics, pharmacokinetics, pain management,
`partial agonists, formulations, opioids
`
`Introduction
`Buprenorphine has been available worldwide
`as a parenteral and sublingual analgesic since
`
`the 1970s. Parenteral buprenorphine has been
`approved for commercial marketing in the
`United States since December 1981. It is one of
`
`Address reprint requests to: Rolley E. Johnson, PharmD,
`Reckitt Benckiser Pharmaceuticals, Inc., 10710 Mid-
`lothian Pike, Suite 430, Richmond, VA 23235, USA.
`Accepted for publication: July 5, 2004.
`At the time this review was written, Dr. Johnson was
`Associate Professor, Department of Psychiatry and
`Behavioral Sciences,
`Johns Hopkins University
`School of Medicine. Currently, Dr. Johnson is Vice-
`
`President Clinical, Scientific and Regulatory Affairs,
`Reckitt Benckiser Pharmaceuticals and Adjunct Pro-
`fessor, Department of Psychiatry and Behavioral Sci-
`ences, Johns Hopkins University School of Medicine.
`Dr. Payne was Chief of the Pain and Palliative
`Care Service, Department of Neurology, Memorial
`Sloan-Kettering Cancer Center. Dr. Payne is now Di-
`rector, Institute on Care at the End of Life, Duke
`Divinity School, Duke University, Durham, NC.
`
`쑖 2005 U.S. Cancer Pain Relief Committee
`Published by Elsevier Inc. All rights reserved.
`
`0885-3924/05/$–see front matter
`doi:10.1016/j.jpainsymman.2004.07.005
`
`Page 1
`
`RB Ex. 2029
`BDSI v. RB PHARMACEUTICALS LTD
`IPR2014-00325
`
`

`

`298
`
`Johnson et al.
`
`Vol. 29 No. 3 March 2005
`
`a number of opioid partial agonists and mixed
`agonist-antagonists currently approved as anal-
`gesics by the Food and Drug Administration
`(Table 1).1
`Buprenorphine (Figure 1) is a derivative of
`the morphine alkaloid thebaine2,3 and is a
`member of
`the 6,14-endo-ethanotetrahydro-
`oripavine class of compounds that includes
`other potent analgesics such as diprenorphine
`and etorphine.4,5 Although buprenorphine has
`been shown to interact in vivo and in vitro with
`multiple opioid receptors, its primary activity
`in man is that of a partial agonist at the mu-
`opioid receptor and antagonist at the kappa
`receptor.6–10 The effects of binding at mu-
`opioid receptors include supraspinal analgesia,
`respiratory depression, and miosis. Buprenor-
`phine, being a partial mu-opioid agonist, may
`have a wider safety profile compared to full mu
`agonists, especially with regard to respiratory
`depression. Further, the slow dissociation of bu-
`prenorphine from the receptor may result in
`fewer signs and symptoms of opioid withdrawal
`upon termination of buprenorphine therapy
`than those which occur with full mu-opioid ago-
`nists, such as morphine, heroin, and metha-
`done. Buprenorphine’s antagonist effects at the
`kappa receptor are associated with limited
`spinal analgesia, and dysphoria and psychoto-
`mimetic effects.11
`Several delivery formulations of buprenor-
`phine have been investigated. Oral bioavailabil-
`ity of buprenorphine is low because of extensive
`first-pass hepatic metabolism.12,13 However, bu-
`prenorphine has certain physiochemical proper-
`ties (discussed later) that can allow for other drug
`delivery technologies to be utilized. The admin-
`istration of buprenorphine by the sublingual
`route allows for bypassing of the first-pass he-
`patic metabolism. Transdermal administration
`
`Fig. 1. Chemical structure of buprenorphine. The
`chemical name of buprenorphine is 6,14-ethenomor-
`phinan-7-methanol, 17-(cyclopropylmethyl)- α-(1,
`1-dimethylethyl)-4, 5-epoxy-18, 19-dihydro-3-hydroxy-
`6-methoxy-α-methyl-, [5α, 7α, (S)]. The structural
`formula is described in Reference 2.
`
`has proven clinical utility for numerous medica-
`tions and provides clinicians the opportunity to
`treat patients who cannot take oral medications,
`such as those with head, neck, mouth or bowel
`lesions, or persistent nausea and vomiting. Both
`the sublingual and transdermal analgesic
`dosage forms of buprenorphine are approved
`for use outside of the United States. In the
`United States, the sublingual formulation has
`been recently approved for the treatment of
`opioid addiction (but not as an analgesic)14 and
`a transdermal formulation is under develop-
`ment. Both are discussed in this review.
`The purpose of this review is to provide
`clinicians and researchers with information
`regarding the appropriate therapeutic use of
`buprenorphine for pain management, and an
`understanding of the mechanisms underlying
`its pharmacodynamic actions. Buprenorphine
`is approved for use as an analgesic for various
`
`Table 1
`Opioid Partial Agonist and Agonist/Antagonists Analgesics Commercially Available for
`Analgesia in the United States
`Activity at
`Dosage Forms
`Activity at
`Mu-opioid Receptor Kappa-opioid Receptor
`Available
`
`Usual Single
`Analgesic Dose (mg)
`
`Controlled Substances
`Act Schedule
`
`Medication
`
`Buprenorphine partial agonist
`Pentazocine
`partial agonist or
`weak antagonist
`partial agonist
`
`Butorphanol
`
`antagonist
`Nalbuphine
`Adapted from Gutstein and Akil.1
`
`antagonist
`agonist
`
`strong agonist
`
`agonist
`
`parenteral
`parenteral
`oral
`parenteral
`nasal
`parenteral
`
`0.3
`30
`50
`1–2
`1–2
`10
`
`III
`IV
`
`IV
`
`Unscheduled
`
`Page 2
`
`

`

`Vol. 29 No. 3 March 2005
`
`Buprenorphine for Pain Management
`
`299
`
`types of pain (e.g., acute, chronic, and neuro-
`pathic pain). It has also been used for treating
`various behavioral and psychiatric disorders
`(e.g., depression and opioid dependence).
`
`Preclinical Pharmacology
`Receptor Binding/Interactions Studies
`In vitro studies have shown that buprenor-
`phine binds with high affinity to mu- and kappa-
`opioid receptors and relatively lower affinity
`to delta-opioid receptors.15,16 Although most
`in vitro studies have shown buprenorphine to be
`relatively non-selective for these receptors,
`others have shown a selective potency of the (-)
`enantiomer of buprenorphine for kappa1 ⫽
`mu ⬎ delta ⬎ kappa2a ⬎ kappa2b,17 with a slow
`dissociation from all receptors.18
`In vivo studies have shown that buprenor-
`phine binds at the mu-opioid receptor,19 where
`it is believed that analgesic and other effects
`(e.g.,
`supraspinal analgesia, respiratory de-
`pression, miosis, decreased gastrointestinal
`motility, and euphoria) are mediated. Bupren-
`orphine is an antagonist at the kappa-opioid
`receptor; agonist activity at the kappa-opioid re-
`ceptor is thought to be associated with spinal
`analgesia, sedation, miosis, and psychotomi-
`metic (i.e., dysphoric) effects. Although bu-
`prenorphine binds with high affinity to the
`delta opioid receptor (but still lower than to
`the mu or kappa1 receptor), the functional sig-
`nificance of this interaction has not been fully
`elucidated.1 More recently, it has been proposed
`that partial agonist activity at the opioid-receptor-
`like 1 (ORL-1) receptor, with its endogenous
`ligand nociceptin or orphanin FQ (N/OFQ),
`may contribute to the analgesic effect of
`buprenorphine.20
`
`Buprenorphine Effects in Pain Models
`Buprenorphine has been shown to increase
`the nociceptive threshold to electrical stimula-
`tion in the tooth pulp assay in dogs.21,22 The
`antinociceptive potency of buprenorphine in
`the rat and guinea pig paw pressure tests was
`noted to be greater than morphine,23 and bu-
`prenorphine was shown to be 10 times more
`potent than morphine in the formalin test (a
`model of post-injury pain).24
`In addition to the biphasic dose-response
`curve observed for buprenorphine with regards
`to effects on respiration in mice and intestinal
`
`motility in rats,25 a bell-shaped dose-response
`curve for the antinociceptive action of bupren-
`orphine has been observed in certain preclini-
`cal pain models (e.g., mouse and rat hot plate,
`rat and monkey tail dip, and rat electrical
`stimulation of the tail and formalin-induced
`flinching),26–31 whereas a linear dose-response
`relationship has been observed in others (e.g.,
`rodent writhing and tail pressure).26 A curvilin-
`ear dose response for antinociceptive effects
`was first observed by Cowan and coworkers in
`the rodent tail dip/flick test,26 and later by Dum
`and Herz27 in in vivo binding studies in the rat.
`Explanations for this bell-shaped curve include
`a 2-receptor model and noncompetitive autoin-
`hibition.7,17,19,26 The peak of the dose-response
`curve occurred at a dose of approximately
`1 mg/kg. The entire curve shifted to the right
`following pretreatment with the opioid anta-
`gonists naloxone32 or naltrexone.27 Although
`readily demonstrated in preclinical analgesic
`studies, the bell-shaped dose-response curve has
`not been observed in clinical analgesic trials that
`have utilized much lower doses of buprenor-
`phine. A study (not an analgesic trial) designed
`to find the peak of this dose-response curve in
`human subjects used a maximum single dose of
`32 mg administered as a sublingual solution.33 A
`plateau of subjective and respiratory depressive
`effects was observed, consistent with the partial
`agonist classification of buprenorphine (Figure
`2); however, the effects were not biphasic even
`in this dose range.
`
`Distinguishing (Discriminative) Stimulus
`Properties and Self-Administration
`In studies where animals were trained to dis-
`tinguish between an opioid (e.g., morphine)
`and no drug (e.g., saline), buprenorphine gen-
`eralized to medications such as morphine and
`fentanyl.34,35 These results indicated that the
`internal drug cues produced by buprenorphine
`are similar to those of the other opioids. Ani-
`mals that have previously been made depen-
`dent on morphine will acquire drug-taking
`behavior (i.e., self-administration by pressing a
`lever that activates administration) when ex-
`posed to morphine-like drugs. Albeit some-
`times weakly, buprenorphine has been shown
`to support intravenous self-administration in
`animals under various conditions of reinforce-
`ment.36–40 Both drug-naı¨ve and drug-experienced
`
`Page 3
`
`

`

`300
`
`Johnson et al.
`
`Vol. 29 No. 3 March 2005
`
`precipitate an abstinence syndrome in rhesus
`monkeys maintained on morphine.43 In an-
`other study, no signs of opioid withdrawal were
`observed when saline was substituted for chroni-
`cally-administered buprenorphine in rhesus
`monkeys, and there were no signs of disrup-
`tions in other behaviors such as food intake.37
`Taken together, the ability of buprenorphine
`to generalize to morphine-like drugs along with
`its production of only relatively mild physical
`dependence indicates that buprenorphine’s
`potential for abuse is limited compared to many
`other opioids.
`Tolerance to the behavioral effects of bu-
`prenorphine has been reported in the rhesus
`monkey.38,44 Cross-tolerance of buprenorphine
`to morphine has been shown in the mouse25
`and rat.41
`
`Safety
`The LD50 values for buprenorphine, assessed
`in a number of animal species by various routes
`of administration, are shown in Table 2.45 Table
`3 shows the comparison of the ratio of the acute
`toxic doses to the antinociceptive doses yielding
`the therapeutic index for morphine and bu-
`prenorphine in rats. These data are consistent
`with a wide safety margin for buprenorphine.
`Studies in mice and rats have shown that bu-
`prenorphine is not a carcinogen at doses 1600
`times greater than the analgesic dose. From
`genetic toxicity studies, including the Ames test,
`the chromosomal aberration assay, and the
`mouse lymphoma forward mutation assay, it has
`been concluded that buprenorphine is not a
`mutagen and presents no genetic danger to
`man.
`
`Table 2
`Acute Toxicity (LD50) of Buprenorphine
`LD50 (mg/kg)
`Base
`HCI salt
`
`Route of Administration
`
`Species
`
`oral
`Mouse
`intravenous
`Mouse
`intramuscular
`Mouse
`intraperitoneal
`Mouse
`subcutaneous
`Mouse
`oral
`Rat
`intravenous
`Rat
`intramuscular
`Rat
`intraperitoneal
`Rat
`subcutaneous
`Rat
`intravenous
`Dog
`– -Data not available. Reference 45.
`
`260
`24
`–
`90
`–
`–
`31
`–
`197
`–
`–
`
`800
`72
`⬎600
`–
`⬎1000
`⬎1000
`62
`⬎600
`–
`⬎1000
`79
`
`Fig. 2. The effects of the partial-agonist buprenor-
`phine (closed circle) and the full-agonist methadone
`(open circle) on an opioid agonist scale. The scale
`contains 16 adjectives descriptive of opioid-like ago-
`nist effects rated on a 0–4 ordinal scale (maximum
`score ⫽ 64). Each vertical bar represents ⫾ 1 SEM.
`Reprinted from Walsh SL, Preston KL, Stitzer ML,
`Cone EJ, Bigelow GE, Clinical pharmacology of
`buprenorphine: ceiling effects at high doses. Clinical
`Pharmacology and Therapeutics, 1994, 55: 569–580,33
`with permission from the American Society for
`Clinical Pharmacology and Therapeutics.
`
`animals have been shown to self-administer
`buprenorphine.37,38
`
`Physical Dependence Liability
`Three primary preclinical experimental pro-
`cedures have been used to evaluate the mor-
`phine-like physical dependence potential of
`buprenorphine in animals. The first procedure
`is the substitution of buprenorphine for mor-
`phine in morphine-withdrawn animals. The
`second is the precipitation of an opioid absti-
`nence syndrome by buprenorphine in mor-
`phine-dependent animals. The third is the
`substitution of placebo (i.e., saline) to assess
`the presence of spontaneous withdrawal in bu-
`prenorphine-maintained animals.
`In studies of the above-described procedures,
`buprenorphine has been shown to produce
`either no, or a protracted but mild, opioid-
`like withdrawal syndrome in rats, dogs, and
`non-human primates.6,26,27,37,41,42 For example,
`Martin and coworkers showed that in dogs
`maintained on 125 mg/day morphine, at low
`doses, buprenorphine substituted for morphine
`(i.e., suppressed spontaneous withdrawal) and
`at higher doses, precipitated an abstinence syn-
`drome.6 Buprenorphine was also reported to
`
`Page 4
`
`

`

`Vol. 29 No. 3 March 2005
`
`Buprenorphine for Pain Management
`
`301
`
`Table 3
`Therapeutic Indices for Morphine
`and Buprenorphine
`ED50,
`LD50, Acute Tail Pressure
`(mg/kg)
`(mg/kg)
`
`Therapeutic
`Index
`LD50/ED50
`
`Opioid
`
`Morphine
`
`Buprenorphine
`
`306
`[237, 395]
`197
`[145, 277]
`
`0.66
`[0.26, 1.6]
`0.016
`[0.011, 0.024]
`
`464
`
`12,313
`
`References 25,26
`Numbers in brackets are 95% confidence limits.
`
`Although buprenorphine has been reported
`to be without teratogenic effects in rodents,46
`significant increases in skeletal abnormalities
`were noted in rats after subcutaneous admin-
`istration of 1 mg/kg/day and greater, but not
`at oral doses up to 160 mg/kg/day.14 Increases
`in skeletal abnormalities in rabbits after intra-
`muscular administration of 5 mg/kg/day, or
`1 mg/kg/day or more given orally were not
`statistically significant. Buprenorphine pro-
`duced statistically significant pre-implantation
`(oral doses of 1 mg/kg/day or more) and post-
`implantation (intravenous doses of 0.2 mg/kg/
`day) losses in rabbits.14
`Unlike effects observed from some other opi-
`oids, prenatal exposure in rats to buprenor-
`phine does not appear to affect activity, cycles of
`rest-activity, or developmental milestones.46–52
`The oral administration of buprenorphine to
`rats during gestation and lactation, at doses sev-
`eral hundred times greater than the analgesic
`dose, has been associated with delayed post-
`natal development of the righting reflex and
`startle response.14,53 It has been reported that
`buprenorphine reduces striatal nerve growth
`factor54 and produces toxic effects similar to
`methadone.52 Mixed effects of buprenorphine
`on maternal water intake, postnatal growth,
`maternal weight gain, frequency of resorption,
`or pup birth weights, number of stillbirths,
`and offspring mortality have also been re-
`ported.14,52,55–57 Physical dependence and toler-
`ance to the antinociceptive effects of morphine
`in pups exposed perinatally to buprenorphine
`and methadone have been demonstrated; gen-
`eralized neuromuscular development does not
`appear to be delayed by perinatal exposure to
`buprenorphine.57
`
`the mu-opioid receptor.18,58–60 This slow recep-
`tor dissociation has generally been regarded
`as
`the property responsible for buprenor-
`phine’s relatively long duration of action as an
`analgesic. Buprenorphine also has a high af-
`finity for the mu-opioid receptor, and is not
`displaced easily by antagonists, such as nalox-
`one, which have a lower receptor affinity.61
`The elimination half-life of buprenorphine
`in humans has been described as either bipha-
`sic62 or triphasic.63,64 Buprenorphine is highly
`bound (96%) to plasma proteins, primarily to
`α- and β-globulin fractions.65 Studies utilizing
`human liver microsomal preparations indicated
`that buprenorphine is demethylated to form
`norbuprenorphine, and is also metabolized
`to other compounds by cytochrome P-450
`3A4.66,67 Both buprenorphine and norbupren-
`orphine form conjugates with glucuronic
`acid.68,69 Studies in rats utilizing intraventricu-
`lar administration of norbuprenorphine and
`buprenorphine indicated that the intrinsic an-
`algesic activity of norbuprenorphine was 25%
`that of buprenorphine.70
`The oral bioavailability of buprenorphine
`is approximately 10%, secondary to extensive
`first-pass hepatic metabolism.12,71 Preclinical
`studies in rats indicate that buprenorphine
`distributes rapidly to the brain following intra-
`venous administration.70 Brain to plasma con-
`centration ratios of buprenorphine in rats
`following a single intravenous dose ranged from
`3.0 at 15 minutes to 10.5 at 6 hours post-drug
`administration.72 The more polar metabolite
`norbuprenorphine has an n-octanol:water par-
`tition coefficient about 10% that of bupren-
`orphine70 and penetrates
`into the central
`nervous system to a much lesser degree than
`the parent compound.73 In the rat, dog, monkey,
`and human, approximately 70% or more of an
`intravenous dose is recovered in the feces;74
`enterohepatic recycling is likely.75 A much
`lesser percentage of buprenorphine (10–30%)
`is found in the urine following administration
`by various other routes.65,75 Concentrations
`found in human red blood cells are comparable
`to those in the plasma.63
`
`Pharmacokinetics
`General Observations
`Buprenorphine is an extremely lipophilic
`compound58 that dissociates very slowly from
`
`Parenterally Administered Buprenorphine
`In the United States, buprenorphine, used
`as an analgesic, is only approved for parenteral
`administration, typically by the intramuscular
`
`Page 5
`
`

`

`302
`
`Johnson et al.
`
`Vol. 29 No. 3 March 2005
`
`or intravenous route. Peak plasma concentra-
`tions following intramuscular administration
`occurred, in general, 5 minutes after dosing,
`and in some patients, by 2 minutes.63 Mean
`plasma concentrations of buprenorphine in
`that study differed little after 5 minutes post-
`drug administration by either the intravenous
`or intramuscular routes; intramuscular bioavail-
`ability ranged from 40% to greater than 90%.
`The volume of distribution at steady state has
`usually been found to be between 200 and
`400 liters.76
`Following the administration of 0.3 mg of
`intravenous buprenorphine given intraopera-
`tively, the initial half-life was found to be about
`2 minutes,63 with a mean terminal half-life of 5
`hours.77 A study by Mendelson and coworkers78
`indicated that the mean terminal half-life of
`intravenously given buprenorphine (1 mg in-
`fused over 30 minutes) was about 6 hours.
`Kuhlman and colleagues79 reported a mean ter-
`minal half-life of 3.2 hours following single
`doses of 1.2 mg given intravenously.
`Buprenorphine clearance following intrave-
`nous administration has typically been reported
`to be between 70 and 80 liters/hour when doses
`in the analgesic range have been used.63,79 The
`clearance of buprenorphine in anesthetized
`patients was found to be lower than in the
`same individuals not under anesthesia secondary
`to reduced hepatic blood flow from the
`anesthetic.63
`
`Buprenorphine Sublingual Liquid/Buccal Strip
`The absorption of buprenorphine liquid
`from the sublingual mucosa is rapid, occurring
`within 5 minutes.80 In a study utilizing healthy
`volunteers,80,81 the bioavailability of buprenor-
`phine in a 30% ethanol solution administered
`sublingually was approximately 30%. Kuhlman
`and colleagues79 studied the pharmacokinetics
`of buprenorphine by various routes of admin-
`istration using a crossover design in healthy,
`non-dependent men who had a history of
`heroin abuse. Buprenorphine bioavailability by
`the sublingual and buccal routes was approxi-
`mately 51% and 28%, respectively, with much
`interindividual variability. The mean terminal
`half-lives were 28 hours following sublingual
`administration and 19 hours following buccal
`administration, compared with 3.2 hours
`following the intravenous route, perhaps re-
`lated to the sequestering of buprenorphine in
`
`the oral mucosa. Average clearances for the 3
`routes of administration were 210, 712, and 77
`liters/hour, respectively. In a study that evalu-
`ated sublingual dosages of buprenorphine up
`to 32 mg,33 peak plasma concentrations of bu-
`prenorphine were observed at 60 minutes fol-
`lowing doses of 2 and 4 mg, and at 30 minutes
`for doses of 8, 16, and 32 mg. Plasma concentra-
`tions after administration of the 32 mg dose
`were significantly elevated for up to 60 hours
`following medication administration. As noted
`previously, the oral bioavailability of buprenor-
`phine is very low (approximately 10%). Thus,
`the swallowing of buprenorphine that is not
`absorbed buccally or sublingually would con-
`tribute little to overall absorption.
`
`Buprenorphine Sublingual Tablets
`Following the sublingual administration of
`0.4 or 0.8 mg doses, there was no significant
`rise in buprenorphine plasma concentrations
`for 20 minutes; the time to maximum concen-
`tration was variable, ranging from 90 to 360
`minutes.76,77 The average systemic bioavailabil-
`ity was 55%, with large intersubject variability.
`A number of studies have assessed the phar-
`macokinetic profile of a buprenorphine tablet
`formulation. Bioavailability of the tablet was
`reported to be approximately 50–65% that
`of the sublingual solution, based on 48- and
`24-hour AUC measurements, respectively.82,83
`Results were generally comparable regardless
`of whether buprenorphine was administered
`as a single dose, or administered once daily
`over multiple days. When buprenorphine tab-
`lets were given over multiple days, average con-
`centrations peaked 2 hours after medication
`administration, in contrast to 1 hour as has
`been found for the solution.
`
`Buprenorphine for Intranasal Administration
`The bioavailability of intranasal buprenor-
`phine has been assessed in humans84 and
`sheep85 using a polyethylene glycol 300 (PEG)
`and a 5% dextrose vehicle. The buprenorphine
`formulation in humans was found to be approx-
`imately 50% bioavailable, with a time to maxi-
`mum concentration of 30 minutes. In sheep,
`the bioavailability of buprenorphine in PEG
`and dextrose was 70% and 89%, respectively;
`time to maximum concentration was 10 mi-
`nutes. From these data, it appears that an intra-
`nasal
`formulation of buprenorphine would
`
`Page 6
`
`

`

`Vol. 29 No. 3 March 2005
`
`Buprenorphine for Pain Management
`
`303
`
`Fig. 3. Approximate bioavailability of buprenorphine by route of administration. Reprinted from Methadone
`Treatment for Opioid Dependence [Figure 13.2 (c)]. Strain, Eric C., M.D., and Maxine L. Stitzer, Ph.D.,
`eds. The Johns Hopkins University Press. Baltimore, Maryland: The Johns Hopkins University Press, 1999: 300.
`Reprinted with permission from The Johns Hopkins University Press.
`
`provide a rapid onset of analgesic effect. The ap-
`proximate bioavailability of buprenorphine by vari-
`ous routes of administration is shown in Figure 3.
`Buprenorphine for Transdermal Administration
`The ideal medication for transdermal ad-
`ministration should be highly lipophilic and of
`low molecular weight (less than approximately
`1000) for ease of crossing the skin barrier.86 It
`should also be highly potent so that adequate
`doses could be delivered through the skin. Bu-
`prenorphine meets these requirements. It has
`an octanol-to-water partition coefficient of 1217
`(i.e., high lipophilicity),87 a molecular weight
`of 468, and is 25 to 50 times more potent as
`an analgesic, per mg, than morphine. Further,
`with a transdermal formulation, a therapeutic
`blood level could be maintained over an ex-
`tended period of time, thus improving compli-
`ance and effectiveness of the medication.
`Recently, a transdermal buprenorphine pro-
`duct has been approved and marketed in a
`number of European countries.88,89 This trans-
`dermal system is designed to continuously re-
`lease buprenorphine at one of three defined
`rates: 35, 52.5, or 70 µg/hr, corresponding to
`daily doses of 0.84, 1.26, and 1.68 mg/24 hr,
`respectively. Effective plasma levels are reached
`within 12 to 24 hours and are kept at a constant
`level for 72 hours. The buprenorphine is incor-
`porated into a polymer adhesive matrix.
`Three dosage strengths of a seven-day bu-
`prenorphine transdermal system are being de-
`veloped in the United States, which deliver 5,
`
`10, or 20 µg/hr buprenorphine, respectively.90
`The highest strength patch (20 µg/hr) will
`result in a dosage of 0.48 mg/day. Compared
`to the higher-strength European product de-
`scribed above, these three dosage strengths may
`be more useful for milder pain syndromes. The
`buprenorphine is dissolved in a polymer matrix
`and the rate of drug release is controlled by
`the diffusion of the buprenorphine in the adhe-
`sive matrix through the stratum corneum of
`the epidermis. The concentration of buprenor-
`phine mixed in the adhesive matrix is the same
`for each strength. After application of the trans-
`dermal system with release rates of 5, 10, and
`20 µg/hr to healthy subjects, mean (⫾SEM)
`peak buprenorphine plasma concentrations
`(Cmax) were 176 ⫾ 34, 191 ⫾ 19, and 471 ⫾
`77 pg/mL, respectively.90 The concentration of
`buprenorphine released from each system per
`hour is proportional to the surface area of the
`system. The time to reach steady-state plasma
`concentrations was approximately 24 to 48
`hours and the percentage of the total dose del-
`ivered in 7 days was 15%.90 Following system
`removal, concentrations decreased to about
`one-half in 12 hours, then declined more gradu-
`ally with an apparent
`terminal half-life of
`26 hours.90,91
`
`Special Considerations
`
`Buprenorphine in Renal Failure. The disposition
`of buprenorphine in patients with renal failure
`
`Page 7
`
`

`

`304
`
`Johnson et al.
`
`Vol. 29 No. 3 March 2005
`
`was examined in studies utilizing both single-
`and multiple-dosing.92 In the single-dose study
`using balanced anesthesia, buprenorphine was
`given intravenously at a dose of 0.3 mg. In the
`multiple-dose study, a variable-rate infusion was
`utilized with controlled ventilation to provide
`analgesia in the intensive care unit (median
`infusion rate of 161 µg/hr for a median of 30
`hours). In the first study, there were no differ-
`ences
`in buprenorphine kinetics between
`healthy patients and those with renal failure
`(all dialysis-dependent with creatinine clear-
`ances less than 5 mL/min). Buprenorphine
`clearances and dose-corrected plasma concen-
`trations were similar in the 2 groups of patients.
`However, in patients with renal failure (plasma
`creatinine concentration greater than 140
`µmol/liter), plasma concentrations of nor-
`buprenorphine were increased by a median of
`4 times, and buprenorphine-3-glucuronide by
`a median of 15 times.
`Another study, which measured only bupren-
`orphine (not metabolites) over a 3-hour sam-
`pling period, reported that the disposition of
`buprenorphine was similar in patients with end-
`stage renal failure compared to healthy con-
`trols.93 The renal failure patients did not show
`clinical evidence of sedation or respiratory
`depression.
`
`Buprenorphine in Hepatic Failure. Few data are
`available with regard to the use of buprenor-
`phine in patients with hepatic failure. A recent
`study evaluated the pharmacokinetic profile of
`buprenorphine (0.3 mg given intravenously) in
`subjects with mild to moderate chronic hepatic
`impairment and in healthy controls matched
`for age, weight, and sex.94 No differences be-
`tween the groups were observed for most
`pharmacokinetic parameters (e.g., steady-state
`volume of distribution, total clearance). How-
`ever, the maximum plasma concentrations of
`buprenorphine and norbuprenorphine were
`50% and 30% lower, respectively, in individuals
`with hepatic impairment. These subjects also
`had less nausea and vomiting than the controls.
`The results did not indicate the need for a
`buprenorphine dosage adjustment in individu-
`als with mild to moderate chronic hepatic
`impairment.
`
`Buprenorphine in Children and Infants. When
`buprenorphine (3 µg/kg) was given intrave-
`nously as premedication to children aged 4 to
`
`7 years, mean clearance was 3.6 liters/hr/kg
`and steady state volume of distribution varied
`from 1.2 to 8.3 liters/kg.95 None of the kinetic
`parameters correlated with age, body weight,
`or body surface area. Because buprenorphine
`plasma concentrations declined rapidly, termi-
`nal elimination half-life could not be estimated
`reliably. In a study of the pharmacokinetics of
`a buprenorphine infusion in premature neo-
`nates,96 the clearance of buprenorphine was
`lower than values previously reported for
`adults and children, probably related to im-
`maturity of
`the glucuronidation metabolic
`pathway.
`
`Clinical Pharmacology
`Analgesia and Anesthesia
`
`Pain Assessment and Treatment. Pain may be de-
`scribed as an unpleasant sensory and emotional
`experience associated with actual or potential
`tissue damage, or described in terms of such
`damage. It is typically categorized broadly as
`being either acute or chronic. Whereas acute
`pain is often associated with a particular injury
`or procedure, chronic pain is pain that has been
`present for more than three months, and which
`may be persistent or intermittent. In addition,
`chronic pain may persist after the disease itself
`has been effectively treated.97
`As noted by Bonica,98 few basic and clinical
`scientists had devoted their efforts to pain re-
`search prior to the 1960s. Differences between
`acute and chronic pain were not appreciated,
`and animal models, particularly for chronic
`pain, were not being developed. More recently,
`preclinical and clinical research studies have
`elucidated multiple mechanisms and sites asso-
`ciated with the production of pain.99 Pain itself
`is subject to much inter-individual variability
`with regard to threshold and tolerance, and has
`expectational and emotional components.100
`Thus, all clinical practice guidelines emphasize
`the need to use patient self-report as the gold
`standard for assessing pain rather than observ-
`ers’ reports because pain is such a personal
`experience.
`Numerous opioids and opioid-like medica-
`tions have been used to treat both acute and
`chronic pain. Chronic pain may involve pain
`related to cancer, as well as noncancer pain due
`
`Page 8
`
`

`

`Vol. 29 No. 3 March 2005
`
`Buprenorphine for Pain Management
`
`305
`
`to osteoarthritis, chronic back pain, and neural-
`gia. Although morphine is the prototypical
`agent, numerous other drugs such as hydroco-
`done, oxycodone, methadone, and others have
`been utilized effectively. The use of opioid anal-
`gesics for the treatment of chronic noncancer
`pain, however, still elicits controversy, much of
`it related to concerns regarding adverse effects
`and possible addiction.101 It is especially im-
`portant to differentiate between addiction to
`opioids and the a

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