throbber
Naltrexone long-acting formulation
`in the treatment of alcohol dependence
`
`R E V I E W
`
`Bankole A Johnson
`Department of Psychiatry and
`Neurobehavioral Sciences, University
`of Virginia, Charlottesville, VA, USA
`
`Correspondence: Bankole A Johnson
`Alumni Professor and Chairman,
`Department of Psychiatry and
`Neurobehavioral Sciences,
`University of Virginia, PO Box 800623,
`Charlottesville, VA 22908-0623, USA
`Tel +1 434 924 5457
`Fax +1 434 244 7565
`Email bankolejohnson@virginia.edu
`
`Abstract: While oral naltrexone has a demonstrated ability to decrease alcohol reinforcement,
`it also has pharmacotherapeutic limitations, such as a small treatment effect size, adverse events,
`and plasma level fl uctuations. The pharmacokinetic profi le of naltrexone could be enhanced
`by intramuscular administration, which would sustain its release over several weeks and keep
`plasma levels relatively constant, ie, low enough to minimize side effects but high enough to
`reduce drinking. Vivitrex®/Vivitrol® and Naltrel® are injectable naltrexone depot formulations
`that have been tested as possible medications for treating alcohol dependence. Their adverse-
`event profi les appear to be less severe than that of oral naltrexone. Vivitrex®/Vivitrol® has
`demonstrated effi cacy at decreasing heavy drinking among alcohol-dependent males. Naltrel®
`helped to promote abstinence and decrease the incidence of relapse in two samples of alco-
`hol-dependent subjects. The data on a third formulation, Depotrex®, are still limited. All three
`formulations require further study of their effi cacy.
`Keywords: alcohol dependence, depot, Depotrex®, Naltrel®, naltrexone, Vivitrex®, Vivitrol®
`
`Introduction
`The reinforcing effects of alcohol associated with its abuse liability are mediated
`by dopaminergic pathways that originate in the ventral tegmental area, relay to the
`nucleus accumbens with neuronal inputs from other limbic regions, and progress to
`the cortex (Wise and Bozarth 1987; Weiss and Porrino 2002; Koob 2003). Naltrexone,
`a mu-opioid receptor antagonist, decreases alcohol reinforcement via two mecha-
`nisms: (1) suppression of alcohol-mediated beta-endorphin stimulation of dopamine
`neurons directly in the nucleus accumbens, and (2) reduction of beta-endorphin
`disinhibition of the tonic inhibition of dopamine cells by gamma-aminobutyric acid
`neurons in the ventral tegmental area (Spanagel and Zieglgansberger 1997; Johnson
`and Ait-Daoud 2000).
`Srisurapanont and Jarusuraisin (2005), in a review of 27 randomized controlled
`clinical trials, reported that oral naltrexone was effi cacious at decreasing relapse and
`a return to heavy drinking among recently abstinent alcohol-dependent individuals,
`which is consistent with the above hypothesis. Yet, since the pharmacokinetic proper-
`ties of oral naltrexone lead to signifi cant fl uctuations in plasma levels with oral daily
`dosing, its general effectiveness has been limited by two consequential factors. First,
`the low plasma trough level of oral naltrexone diminishes its effi cacy, which could
`explain why medication adherence above 85% is required in order for there to be a
`therapeutic response (Volpicelli et al 1997). Second, high peak levels are deemed
`responsible for adverse events (Croop et al 1997; King et al 1997), and up to 15% of
`oral naltrexone recipients drop out of treatment because of adverse events, especially
`nausea (Croop et al 1997).
`The effectiveness of naltrexone also is limited by its small treatment effect
`size (Johnson and Ait-Daoud 2000; Feinn and Kranzler 2005), especially in newer
`
`Therapeutics and Clinical Risk Management 2007:3(5) 741–749
`© 2007 Dove Medical Press Limited. All rights reserved
`
`741
`
`AMN1050
`IPR of Patent No. 7,919,499
`
`

`

`Johnson
`
`and multi-site trials; the number needed to treat (ie, to see
`a difference from placebo) is 7 for decreasing the likelihood
`of relapse and 12 for decreasing the likelihood of returning
`to drinking (Srisurapanont and Jarusuraisin 2005). Never-
`theless, subjects with the Asp40 allele of the mu-opioid
`receptor, as opposed to those with the Asn40 allelic type,
`might derive greater therapeutic benefi t than is seen in
`the averaged response (Oslin et al 2003). Further study is
`needed to confi rm these results.
`Optimizing the pharmacokinetic profi le of naltrexone by
`developing a deep intramuscular injection that would release
`naltrexone over several weeks would, therefore, enhance its
`overall effectiveness. Consequently, plasma levels would
`remain relatively constant and low enough to reduce the
`incidence of adverse events yet high enough for the desired
`anti-drinking effects (Bartus et al 2003). In other words,
`while the effect size of naltrexone’s long-acting, intramus-
`cular formulation would not be expected to exceed the effect
`size of oral naltrexone, the overall outcome would probably
`be enhanced by the increased compliance and longer exposure
`to a therapeutic dose. This review focuses on the therapeutic
`effects and pharmacological properties of two long-acting,
`injectable depot preparations of naltrexone – Vivitrex®,
`recently renamed Vivitrol® (Alkermes, Inc., Cambridge,
`MA, USA), and Naltrel® (DrugAbuse Sciences, Inc., Paris,
`France) – for treating alcohol dependence. Another depot
`formulation, Depotrex® (Biotek, Inc., Woburn, MA, USA),
`for which published data are limited, is also mentioned.
`Table 1 provides a summary of the advantages and dis-
`advantages of depot naltrexone preparations compared with
`oral naltrexone in alcohol-dependent individuals.
`
`Currently available preparations
`Properly formulated depot preparations can maintain
`relatively constant plasma levels for days or weeks because
`of the slow, timed release of the compound. Long-acting
`naltrexone depot formulations also are designed to minimize
`the high plasma peaks and exposure of the gastrointestinal
`tract to naltrexone that occur with the oral formulation.
`Thus, there is a reduction in nausea, the main adverse event
`associated with discontinuation of naltrexone treatment.
`Also, the relatively stable plasma levels of a naltrexone depot
`formulation help to maintain constant levels of mu-opioid
`receptor occupancy, and, importantly, this facilitates a
`linear pharmacodynamic response. Since alcohol-dependent
`individuals often are relatively non-compliant with regard to
`medication taking (Rohsenow et al 2000), spacing naltrexone
`injections at intervals of up to 4 weeks, thereby keeping
`plasma levels constant, should enhance compliance and
`promote greater effi cacy.
`Vivitrex®/Vivitrol® is naltrexone formulated into
`poly-(lactide-co-glycolide) (Shive and Anderson 1997),
`small-diameter (⬍100 µm), injectable microspheres, which
`contain other proprietary active moieties that lead to its
`extended-release properties lasting for several weeks (Lewis
`1990). In animal studies, these microspheres were suspended
`in 1 mL of an aqueous solution (3.0% low-viscosity
`carboxymethylcellulose, 0.9% saline, and 0.1% Tween-20),
`enabling injection of a 50 mg/kg dose of naltrexone (Bartus
`et al 2003). The plasma naltrexone level reached its peak
`at approximately 15 ng/mL by the third day post-injection,
`was sustained at approximately 12 ng/mL for another
`18 days, and then tapered off until it dipped below 1 ng/mL
`
`Table 1 Advantages and disadvantages of depot naltrexone preparations compared with oral naltrexone in alcohol-dependent
`individuals
`Advantages of depot naltrexone preparations compared with
`oral naltrexone
`• Effi cacy is not compromised since there are
`
`not signifi cant fl uctuations in plasma levels
`
`causing low trough levels
`• Adverse events, particularly nausea, are not
`
`increased by high peak levels that would result from the
`
`plasma level fl uctuations
`•
`Since injections are spaced 4-weeks apart,
`
`problems with compliance are minimized
`• The simplicity of supervision and administration
` might make the depot formulations suitable for
`
`forensic settings
`• Patients who will be in situations where oral
`
`naltrexone is unavailable can receive treatment
`
`
`
`
`Disadvantages of depot naltrexone preparations compared
`with oral naltrexone
`• An apparent gender disparity in effi cacy
`
`(with men receiving the greater benefi t)
`
`requires further exploration
`• Certain adverse events, such as erythema,
`
`induration, and injection site reactions, are unique to
`
`the depot formulations
`• Vivitrol® is contraindicated in patients
`
`receiving opioid analgesics
`• More health care providers must be involved to
`
`ensure proper administration
`• Depot formulations could be cost prohibitive for many
`
`patients
`• Delivery of psychosocial support might be
`
`needed more often than the monthly injections
`
`742
`
`Therapeutics and Clinical Risk Management 2007:3(5)
`
`AMN1050
`IPR of Patent No. 7,919,499
`
`

`

`14 days after that (Bartus et al 2003). Vivitrex® resulted in
`an approximate 70% reduction, compared with placebo, of
`morphine-induced analgesia in the hot-plate test for approxi-
`mately 3 weeks – an effect that disappeared by 4 weeks after
`injection. The expected rise in mu-receptor density, caused by
`Vivitrex®-induced antagonist blockade, was evaluated using
`[D-ala2, N-methyl-phe4, glycol5] enkephalin ([3H]DAMGO).
`This revealed that there was a 110% increase, compared with
`placebo, in mu-receptor density, from 5 days after the injec-
`tion until 33 days later, most prominently in the thalamus,
`nucleus accumbens, dorsal raphe nucleus, and striatum.
`Vivitrex®, therefore, appears to block effectively the central
`mu-opioid receptors for a period of approximately 4 weeks
`after the injection (Bartus et al 2003).
`Fewer data on Naltrel® than on Vivitrex®/Vivitrol® exist
`in the public domain. Naltrel® consists of naltrexone incor-
`porated within microspheres of poly-(DL-lactide) polymer.
`These microspheres are contained in single-dose vials and
`suspended in a diluent comprising mannitol, carboxymeth-
`ylcellulose, polysorbate 80, and water for injection. When
`metabolized, the polylactide polymer produces water and
`carbon dioxide. Degradation of the microspheres causes
`naltrexone to be released (Kranzler et al 2004).
`A lesser-known third formulation, Depotrex®, is dis-
`cussed briefl y in the Clinical Results section below.
`
`Pharmacodynamics
`and pharmacokinetics
`The marked analgesic response to morphine in the hot-plate
`paradigm in rats was blocked by Vivitrex® (50 mg/kg) from
`the fi rst day of injection until 4 weeks later. An injection
`of Vivitrex® 5 weeks after the fi rst injection led to suppres-
`sion of morphine analgesia for another 4 weeks (Bartus
`et al 2003). When Vivitrex® was injected subcutaneously,
`plasma naltrexone peaked at approximately 15 ng/mL after
`approximately 3 days; following intramuscular injection, it
`peaked at 19 ng/mL, also after approximately 3 days. Mean
`plasma naltrexone levels were 12 to 14 ng/mL for the next
`3 weeks regardless of the route of administration, and they
`were detectable until 5 weeks after the injection. After the
`administration of a competitive mu-receptor antagonist, there
`usually is a neuroadaptive upregulation of these receptors
`(Lahti and Collins 1978; Zukin et al 1982). This pharmacody-
`namic response was quantifi ed by measuring the mu-receptor
`density with [3H]DAMGO radioligand autoradiography fol-
`lowing the administration of Vivitrex®. After a single injec-
`tion, signifi cant increases in mu-receptor density occurred,
`especially in the midbrain and striatum a week later and in the
`
`Naltrexone long-acting formulation for alcohol dependence
`
`neocortex a month later; these were sustained for 2–4 weeks.
`Similar results were seen in immunochemistry studies, but
`with relatively smaller increases, which ranged from 10%
`to 40% (Bartus et al 2003). Importantly, the amount of mu-
`receptor upregulation after injection of Vivitrex® appears
`similar to the amount after at least 4 weeks of oral naltrexone
`administration (Giordano et al 1990). In view of the fact
`that suppression of morphine analgesia also occurred in the
`hot-plate paradigm for 5 weeks after the administration of
`a single Vivitrex® injection, it is reasonable to suggest that
`a pharmacologically relevant dose of Vivitrex®/Vivitrol®
`continues its pharmacodynamic effect of blocking central
`mu-receptors for up to 1 month post-injection.
`Johnson et al (2004) showed, in a double-blind,
`placebo-controlled, randomized, multi-site, 16 week study
`of 30 alcohol-dependent individuals, that the 25 subjects
`receiving an intramuscular injection of Vivitrex® (400 mg)
`every 4 weeks for 4 months had a mean plasma 6-beta-
`naltrexol (naltrexone’s major metabolite) trough level of
`3.0 ng/mL and a mean naltrexone trough level of 1.3 ng/mL.
`In contrast, an earlier study found that – 16 hours after
`administration of oral naltrexone (50 mg) – subjects had a
`mean serum 6-beta-naltrexol level of 24.9 ng/mL (McCaul
`et al 2000). The fi ndings of King et al (1997) showed mean
`urinary concentrations of 29.0 µg/mg for 6-beta-naltrexol
`and 2.9 µg/mg for naltrexone, 3 hours after oral administra-
`tion of naltrexone (50 mg) in 24 male moderate-to-heavy
`social drinkers.
`Galloway et al (2005) demonstrated, in an open-label,
`single-site, 6 week study of 16 alcohol-dependent indi-
`viduals receiving just one intramuscular injection of Naltrel®
`(300 mg), that serum naltrexone levels increased to a peak of
`approximately 2.04 ng/mL at 2 weeks and dissipated slowly
`to 0.58 ng/mL over the next 4 weeks. Plasma naltrexone and
`6-beta-naltrexol levels at week 4 were approximately 0.75
`and 2.2 ng/mL, respectively. These levels were proportion-
`ately (ie, to dose) less than those found in the Vivitrex® study
`by Johnson et al (2004).
`In humans, the peak plasma concentration of long-acting
`naltrexone depot formulations is greater than that of oral nal-
`trexone during the days immediately after the injection. The
`advantage of these formulations with respect to tolerability,
`therefore, may be that such peaks just occur early in treatment
`with the depot preparations whereas they occur daily with
`oral naltrexone. The lack of fi rst-pass metabolism with the
`long-acting preparations, with diminished 6-beta-naltrexol
`levels, also might lead to an improved adverse-event profi le
`as increased levels of beta-naltrexol have been associated
`
`Therapeutics and Clinical Risk Management 2007:3(5)
`
`743
`
`AMN1050
`IPR of Patent No. 7,919,499
`
`

`

`Johnson
`
`with a greater severity and frequency of naltrexone-related
`adverse events (King et al 1997).
`Thus, preclinical and human studies provide a phar-
`macodynamic and pharmacokinetic basis for the monthly
`injection of a long-acting naltrexone depot formulation as
`treatment for alcohol dependence through the blockade of
`mu-opioid receptors.
`
`Clinical results
`Clinical trials involving alcohol-dependent individuals have
`examined the effi cacy, safety, and tolerability of Naltrel® and
`Vivitrex®/Vivitrol®.
`
`Naltrel®
`The fi rst published study on the effi cacy, safety, and toler-
`ability of Naltrel® for treating alcohol dependence comprised
`a multi-site, double-blind, 12 week clinical trial. One hun-
`dred fi fty-eight alcohol-dependent men and women were
`assigned to receive Naltrel® and 157 received placebo, both
`accompanied by motivation enhancement-based psycho-
`social support, every 4 weeks (Kranzler et al 2004). The
`fi rst Naltrel® dose consisted of one injection of 150 mg in
`each buttock, and each dose thereafter was just 150 mg.
`Placebo was identical in number and volume of injections
`but did not contain the active compound. Generally, Naltrel®
`appeared to be well tolerated and safe. Side effects that were
`reported signifi cantly more frequently in the Naltrel® group
`than in the placebo group included injection site reactions,
`chest pain, and upper abdominal pain. Irritability, however,
`was more common after placebo than after injection of
`Naltrel®. There were 13 dropouts (8.2%) in the Naltrel®
`group and only 6 dropouts (3.8%) in the placebo group;
`the subjects’ reasons for discontinuing treatment, however,
`were similar between the groups. Naltrel® recipients were
`more likely than placebo recipients to have a higher mean
`number of cumulative abstinent days (52.8 days, 95%
`CI 48.5–57.2 days, vs 45.6 days, 95% CI 41.1–50.0 days,
`respectively; p = 0.018) and a longer median time to fi rst
`drink (5 days, 95% CI 3–9 days, vs 3 days, 95% CI 2–4
`days, respectively; p = 0.003). The effects of gender on treat-
`ment outcome were not examined, probably because of the
`relatively small sample size (Kranzler et al 2004).
`A single-site, 6 week, open-label trial studied 16 alcohol-
`dependent individuals who were given a single intramuscular
`dose of Naltrel® (300 mg) (Galloway et al 2005). Of the
`198 adverse events that were reported, 17 were rated as
`severe, including fatigue, gastrointestinal pain, irritability,
`nausea, somnolence (2 reports), headache (4 reports from
`
`3 subjects), injection site pain, injection site mass, lethargy,
`depression, increased gamma-glutamyl transferase (GGT)
`level (an index of heavy drinking) (Conigrave et al 2002),
`back pain, and fl atulence. There were no serious adverse
`events. Also, the trend was for participants’ drinking out-
`comes to improve between enrollment and the end of the
`trial (Galloway et al 2005).
`Since the Naltrel® formulation has shown promise as an
`effi cacious medication for treating alcohol dependence, it
`deserves further study. Early fi ndings indicate that Naltrel® is
`safe and well tolerated, and its adverse-event profi le appears
`to be milder than that reported for oral naltrexone. Additional
`data are needed regarding the effects of gender on treatment
`outcome. Future studies also should show whether Naltrel® is
`likely to cause injection site-related allergic-type reactions.
`
`Vivitrex®/Vivitrol®
`The fi rst published study on the initial effi cacy, safety, and
`tolerability of Vivitrex® for treating alcohol dependence was
`a double-blind, placebo-controlled, randomized, multi-site,
`16 week clinical trial (Johnson et al 2004). Twenty-fi ve
`alcohol-dependent individuals were assigned to receive intra-
`muscular injections of Vivitrex® (400 mg) every 4 weeks,
`while fi ve participants received placebo via the same route
`of administration every 4 weeks. Vivitrex® appeared to be
`relatively safe and well tolerated; the most common adverse
`events were non-specifi c abdominal pain, nausea, pain at
`the injection site, and headaches. Two Vivitrex® recipients
`and zero placebo recipients discontinued treatment because
`of side effects. One participant dropped out due to indura-
`tion at the injection site, and one was discontinued by the
`research staff because of an allergic reaction that resulted
`in angioedema, which resolved soon after the participant
`stopped taking the medication. Even though any conclu-
`sions regarding effi cacy must take into consideration the
`study’s unbalanced cell design, it did appear that Vivitrex®
`was more likely than placebo to lead to a lower percentage
`of heavy drinking days (ie, 11.7% vs 25.3%, respectively).
`In the exercise of scientifi c caution, no inferential statistical
`testing was conducted on these descriptive values. Addition-
`ally, participants in both the Vivitrex® and placebo groups
`demonstrated improved drinking outcomes between enroll-
`ment and study end (Johnson et al 2004).
`The effi cacy, safety, and tolerability of Vivitrex® were
`later studied in a placebo-controlled, double-blind, ran-
`domized, multi-site, 24 week clinical trial (Garbutt et al
`2005). Intramuscular injections of high-dose Vivitrex®
`(380 mg) (n = 205), low-dose Vivitrex® (190 mg) (n = 210),
`
`744
`
`Therapeutics and Clinical Risk Management 2007:3(5)
`
`AMN1050
`IPR of Patent No. 7,919,499
`
`

`

`or matching placebo (n = 209), along with low-intensity
`psychosocial support, were administered to alcohol-
`dependent men and women every 4 weeks. Participants
`who received high-dose Vivitrex® were signifi cantly more
`likely than placebo recipients to report the adverse events of
`decreased appetite, nausea, pain at the injection site, dizzi-
`ness, and fatigue. The low-dose Vivitrex® and placebo groups
`experienced adverse events at a similar frequency. Although
`14.1% of the high-dose Vivitrex® recipients dropped out of
`treatment, only 6.7% of the low-dose Vivitrex® and placebo
`groups did so. Injection site reactions, headaches, and nausea
`were the most common reasons given for discontinuing treat-
`ment. Two high-dose Vivitrex® recipients had serious adverse
`events caused by an interstitial pneumonia and allergic-type
`eosinophilic pneumonia, both of which resolved after medical
`treatment. The high-dose Vivitrex® group, averaged between
`men and women, had a signifi cantly lower percentage of
`heavy drinking days than did placebo recipients (hazard
`ratio [HR] 0.75, 95% CI 0.60–0.94; p = 0.02). An analysis
`by gender, however, demonstrated that the only improvement
`in drinking outcomes among high-dose Vivitrex® recipients
`was in men (HR 0.56, 95% CI 0.41–0.77; p ⬍ 0.001) and
`not women (HR 1.23, 95% CI 0.85–1.78; p = 0.28). These
`fi ndings demonstrate that although women in the high-dose
`Vivitrex® group versus the placebo group reported a 23%
`relative increase in percentage of heavy drinking, men in
`the high-dose Vivitrex® group reported a relative decrease of
`44% in the same variable. High-dose Vivitrex® and placebo
`recipients did not differ signifi cantly in GGT level, and low-
`dose Vivitrex® and placebo recipients did not experience a
`signifi cant difference in GGT level or drinking outcomes
`(Garbutt et al 2005).
`At least four points need to be made concerning the evi-
`dence that Vivitrex®/Vivitrol® can decrease heavy drinking in
`men but not women (Johnson 2006). First, since individuals
`with alcohol dependence in their family history have reportedly
`experienced the best results with oral naltrexone (Monterosso
`et al 2001), it is tempting to speculate that male subjects in the
`Garbutt et al (2005) trial may have responded to Vivitrex® for
`the same reason. Comparative rates of family history of alco-
`holism between men and women, however, were not given.
`Hence, future studies testing the effi cacy of Vivitrex®/Vivitrol®
`should investigate any potential interaction between familial
`alcoholism (or related variables including age of alcoholism
`onset) and treatment outcome.
`Second, Vivitrex® injections might have been more likely
`in women than in men to be delivered subcutaneously instead
`of intramuscularly, thereby slowing absorption, since women
`
`Naltrexone long-acting formulation for alcohol dependence
`
`tend to have a relatively higher percentage of body fat (Blaak
`2001). Indeed, in a study by Kiefer et al (2005), drinking
`outcomes appeared to be better for women than for men
`receiving oral naltrexone. Since Garbutt et al (2005) did not
`study pharmacokinetic data, a report comparing the kinetic
`profi le of Vivitrex®/Vivitrol® between women and men
`would be required to exclude this possibility.
`Third, alcohol-dependent men and women enrolled
`in clinical trials perhaps cannot be compared directly as
`they might differ on non-drinking outcomes, including
`familial pressure to change, rates of affective disorder,
`or individual motivation to achieve treatment objectives.
`There is no evidence, however, to suggest that the women
`enrolled in this trial were atypical of women participat-
`ing in pharmacotherapy trials for the treatment of alcohol
`dependence. Moreover, among the enrolled men, there was
`probably heterogeneity on these same factors. Attempts
`to match women and men who are enrolled in pharmaco-
`therapy trials for treating alcohol dependence on multiple
`non-drinking-related factors would not be practical and
`would lead to the same conclusion, ie, that the therapeutic
`effect of Vivitrex®/Vivitrol® to diminish heavy drinking
`among alcohol-dependent men does not translate to alcohol-
`dependent women. Subjects who participate in pharmaco-
`therapy trials for treating alcohol dependence are mostly
`men, and the relatively small sample sizes of single-site
`studies do not allow meaningful statistical comparisons of
`drinking outcomes between women and men. Of the two
`important trials that resulted in US Food and Drug Admin-
`istration approval of oral naltrexone for treating alcohol
`dependence (O’Malley et al 1992; Volpicelli et al 1992),
`only the O’Malley et al (1992) study included women, but
`not in large enough numbers to permit gender comparisons.
`Given the multitude of published studies testing oral naltrex-
`one for the treatment of alcohol dependence (Srisurapanont
`and Jarusuraisin 2005), a meta-analytic approach to
`examining for a gender effect on treatment outcome would
`be of scientifi c interest. If oral naltrexone has demonstrated
`similar effi cacy between women and men, then the absence
`of an effect for Vivitrex® in women might be a result of
`the fact that oral naltrexone and Vivitrex®/Vivitrol® are
`prepared and administered differently. If, on the other
`hand, meta-analytic studies reveal that oral naltrexone, like
`Vivitrex®/Vivitrol®, exhibits greater effi cacy for men than
`for women, then it is plausible that such fi ndings would be
`related to common pharmacodynamic interaction factors.
`A greater understanding of such factors is necessary for
`optimization of treatment delivery.
`
`Therapeutics and Clinical Risk Management 2007:3(5)
`
`745
`
`AMN1050
`IPR of Patent No. 7,919,499
`
`

`

`Johnson
`
`Fourth, pharmacotherapy studies of naltrexone or its
`analogues for treatment of alcohol dependence usually
`reveal a small to medium effect size. Thus, the differential
`effi cacy for Vivitrex® between men and women might have
`happened by chance.
`The importance of the fi ndings of allergic-type interactions
`with Vivitrex® is uncertain. Based upon the two cases of
`pneumonia reported by Garbutt et al (2005) and the one case
`of angioedema reported by Johnson et al (2004), the allergic-
`type reaction rate for Vivitrex®/Vivitrol® would be 1 per 218
`study subjects. Additional investigation of oral naltrexone
`and naloxone (a structurally similar medication designed for
`intravenous injection) is needed to determine their allergic-
`type reaction rates. Comparing the Vivitrex®/Vivitrol®
`fi ndings directly with any such results, however, would be
`complicated by differences in study population size, disease
`states, and length of exposure, among other factors. A prudent
`analysis would require extensive monitoring of the potential
`for allergic-type reactions after Vivitrex®/Vivitrol® admin-
`istration in future clinical trials. Since Vivitrex®/Vivitrol®
`cannot be removed from a subject’s body after it is injected,
`and any allergic-type reactions would be prolonged as a
`result of the formulation’s long duration of action, a practical
`approach to naltrexone treatment might (depending upon the
`allergic-type reaction rates for oral naltrexone and naloxone)
`be to use a small “test dose” of Vivitrex®/Vivitrol® before
`delivering the full therapeutic dose a few days later.
`Clinical evidence suggests that Vivitrex®/Vivitrol® can
`diminish heavy drinking among men but not women. The
`reason for this difference in effi cacy is still unclear, as is the
`pathophysiological signifi cance of the potential for allergic-
`type reactions with Vivitrex®/Vivitrol®; thus, further investi-
`gation is warranted. Overall, Vivitrex®/Vivitrol® appears to
`be safe and well tolerated, with a milder adverse-event profi le
`than oral naltrexone. Future studies should compare directly
`the side-effect profi les of Vivitrex®/Vivitrol® and Naltrel®.
`
`Depotrex®
`Published data on another depot naltrexone formulation,
`Depotrex®, are limited. Depotrex® appears to cause a
`stable and sustained increase in plasma naltrexone levels.
`It antagonizes mu-opioid receptors with few side effects
`(Heishman et al 1994; Alim et al 1995). Comparative dose-
`ranging pharmacokinetic data on Depotrex® have been
`reported in a study of 12 heroin-dependent individuals
`(Comer et al 2002). Depotrex® (low and high doses of 192
`and 384 mg, respectively) kept plasma naltrexone levels
`above 1 ng/mL for 3 and 4 weeks, respectively. Mean peak
`
`levels for the low and high Depotrex® doses were 3.8 and
`8.9 ng/mL, respectively. Plasma beta-naltrexol levels were
`proportionately higher but were undetectable 5 weeks
`following administration. Both the low and high doses
`antagonized heroin-induced positive subjective effects. The
`primary adverse event reported was mild discomfort at the
`injection site, with no irritation or erythema (Comer et al
`2002). Previously, Kranzler et al (1998) reported promis-
`ing fi ndings showing that the administration of Depotrex®
`(206 mg) was associated with a prolonged increase in plasma
`naltrexone, similar to the Comer et al (2002) study in heroin
`addicts. Moreover, their study highlighted the effi cacy of
`Depotrex®, compared with placebo, at reducing heavy drink-
`ing among alcohol-dependent individuals; nevertheless,
`injection site reactions including induration were observed
`in some participants (Kranzler et al 1998). Additional studies
`on this promising formulation are needed.
`
`Discussion
`While the naltrexone long-acting formulations discussed
`herein have the benefi t of lower adverse-event profi les and
`necessitate fewer visits to a treatment center than would be
`needed for the administration of oral naltrexone, their use
`does require that more health care providers be trained. For
`instance, injections must be administered properly to decrease
`the possibility of local site reactions, which could, in turn,
`diminish compliance. Moreover, the number of physicians
`or nurses might have to be increased so that providers are on
`hand to administer the injections and to assess and triage any
`medical complications that may occur (Johnson 2006).
`Although depression and other psychiatric problems are
`not listed among the contraindications for injectable naltrex-
`one in the Vivitrol® package insert (Alkermes, Inc. 2005),
`adverse events of a suicidal nature were reported infrequently
`in controlled trials among Vivitrol®-treated patients (1% vs 0
`in the placebo group), and depressed mood was twice as likely
`(10% vs 5%) for Vivitrol® (380 mg) recipients vs placebo
`recipients in a 24 week controlled trial. Hence, alcohol-
`dependent patients taking Vivitrol® should be monitored for
`depression or suicidal ideation (Alkermes, Inc. 2005).
`Vivitrol® is contraindicated in patients who are receiving
`opioid analgesics. If pain management becomes necessary
`in an emergency situation, Vivitrol® recipients should be
`given regional analgesia, conscious sedation with a benzo-
`diazepine, and non-opioid analgesics or general anesthesia.
`In situations requiring opioid analgesia, administration of a
`rapidly acting opioid analgesic that minimizes the duration
`of respiratory depression is recommended, with the amount
`
`746
`
`Therapeutics and Clinical Risk Management 2007:3(5)
`
`AMN1050
`IPR of Patent No. 7,919,499
`
`

`

`of analgesic titrated to the patient’s needs. These patients
`should be closely monitored by personnel who are trained in
`cardiopulmonary resuscitation (Alkermes, Inc. 2005).
`Thus far, no precedent exists in the psychiatric fi eld for
`administering a medication intramuscularly instead of orally.
`Practitioners, therefore, might use these long-acting depot
`preparations only if a “trial” of oral naltrexone has failed
`because of low compliance. Furthermore, it also is possible
`that, in real-world generic clinics as opposed to clinical trial
`settings in a research facility, patients might be less likely to
`consent to injections for the treatment of alcohol dependence
`due to injection phobia, relatively less individual attention
`paid by medical staff, or a lower intensity of psychosocial
`support provided by health professionals. Providers might
`also consider a “trial” of oral naltrexone to guarantee early
`detection of any adverse events (Johnson 2006).
`If long-acting depot formulations of naltrexone are
`approved by the US Food and Drug Administration, their
`widespread use might be limited because of cost. Uneven
`insurance coverage has hampered the use of oral naltrexone
`in many parts of the US. While the prices of the depot formu-
`lations have not yet been announced, their daily cost might
`exceed that of oral naltrexone. Hence, these preparations
`might be less accessible to uninsured patients who cannot
`pay on their own (Johnson 2006).
`Treatment providers should not avoid delivering regular,
`adequate psychosocia

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