44
The new england journal of medicine n engl j med 374;13 nejm.org March 31, 2016 1232 From the Departments of Population Health (J.D.L., R.M., M.N.G.), Medicine, Division of General Internal Medicine and Clinical Innovation (J.D.L.), and Psy- chiatry (J.R.), New York University, and the New York State Psychiatric Institute, Columbia University College of Physi- cians and Surgeons (E.V.N.) — both in New York; the Division of General Inter- nal Medicine, the Department of Medi- cine, Rhode Island Hospital and Alpert Medical School of Brown University, Prov- idence (P.D.F., R.A.H., D.W.); Friends Re- search Institute (T.W.K., M.G., M.F.), the University of Baltimore, School of Crimi- nal Justice (T.W.K.), and Maryland Treat- ment Centers (M.F.) — all in Baltimore; the University of Pennsylvania (T.Y.B., J.W.C., C.P.O.) and the Philadelphia Vet- erans Affairs Medical Center (J.W.C.) — both in Philadelphia; the Center for Bio- medical Ethics and Humanities, School of Medicine (D.T.C.) and the School of Law (R.J.B.), University of Virginia, Char- lottesville; and Washington State Univer- sity, Spokane (S.M.M.). Address reprint requests to Dr. Lee at the Department of Population Health, New York University, 227 E. 30th St., New York, NY 10016, or at [email protected]. This article was updated on April 14, 2016, at NEJM.org. N Engl J Med 2016;374:1232-42. DOI: 10.1056/NEJMoa1505409 Copyright © 2016 Massachusetts Medical Society. BACKGROUND Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor antagonist, is effective for the prevention of relapse to opioid depen- dence. Data supporting its effectiveness in U.S. criminal justice populations are limited. METHODS In this five-site, open-label, randomized trial, we compared a 24-week course of extended- release naltrexone (Vivitrol) with usual treatment, consisting of brief counseling and refer- rals for community treatment programs, for the prevention of opioid relapse among adult criminal justice offenders (i.e., persons involved in the U.S. criminal justice system) who had a history of opioid dependence and a preference for opioid-free rather than opioid maintenance treatments and who were abstinent from opioids at the time of randomiza- tion. The primary outcome was the time to an opioid-relapse event, which was defined as 10 or more days of opioid use in a 28-day period as assessed by self-report or by testing of urine samples obtained every 2 weeks; a positive or missing sample was computed as 5 days of opioid use. Post-treatment follow-up occurred at weeks 27, 52, and 78. RESULTS A total of 153 participants were assigned to extended-release naltrexone and 155 to usual treatment. During the 24-week treatment phase, participants assigned to extended- release naltrexone had a longer median time to relapse than did those assigned to usual treatment (10.5 vs. 5.0 weeks, P<0.001; hazard ratio, 0.49; 95% confidence interval [CI], 0.36 to 0.68), a lower rate of relapse (43% vs. 64% of participants, P<0.001; odds ratio, 0.43; 95% CI, 0.28 to 0.65), and a higher rate of opioid-negative urine samples (74% vs. 56%, P<0.001; odds ratio, 2.30; 95% CI, 1.48 to 3.54). At week 78 (approximately 1 year after the end of the treatment phase), rates of opioid-negative urine samples were equal (46% in each group, P = 0.91). The rates of other prespecified secondary outcome mea- sures — self-reported cocaine, alcohol, and intravenous drug use, unsafe sex, and rein- carceration — were not significantly lower with extended-release naltrexone than with usual treatment. Over the total 78 weeks observed, there were no overdose events in the extended-release naltrexone group and seven in the usual-treatment group (P = 0.02). CONCLUSIONS In this trial involving criminal justice offenders, extended-release naltrexone was asso- ciated with a rate of opioid relapse that was lower than that with usual treatment. Opioid-use prevention effects waned after treatment discontinuation. (Funded by the National Institute on Drug Abuse; ClinicalTrials.gov number, NCT00781898.) ABSTRACT Extended-Release Naltrexone to Prevent Opioid Relapse in Criminal Justice Offenders Joshua D. Lee, M.D., Peter D. Friedmann, M.D., M.P.H., Timothy W. Kinlock, Ph.D., Edward V. Nunes, M.D., Tamara Y. Boney, M.S., C.C.R.C., Randall A. Hoskinson, Jr., Donna Wilson, M.S., Ryan McDonald, M.A., John Rotrosen, M.D., Marc N. Gourevitch, M.D., M.P.H., Michael Gordon, D.P.A., Marc Fishman, M.D., Donna T. Chen, M.D., M.P.H., Richard J. Bonnie, L.L.B., James W. Cornish, M.D., Sean M. Murphy, Ph.D., and Charles P. O’Brien, M.D., Ph.D. Original Article The New England Journal of Medicine Downloaded from nejm.org at University at Buffalo Libraries on March 29, 2017. For personal use only. No other uses without permission. Copyright © 2016 Massachusetts Medical Society. All rights reserved.

Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

  • Upload
    others

  • View
    10

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

T h e n e w e ngl a nd j o u r na l o f m e dic i n e

n engl j med 374;13 nejm.org March 31, 20161232

From the Departments of Population Health (J.D.L., R.M., M.N.G.), Medicine, Division of General Internal Medicine and Clinical Innovation (J.D.L.), and Psy-chiatry ( J.R.), New York University, and the New York State Psychiatric Institute, Columbia University College of Physi-cians and Surgeons (E.V.N.) — both in New York; the Division of General Inter-nal Medicine, the Department of Medi-cine, Rhode Island Hospital and Alpert Medical School of Brown University, Prov-idence (P.D.F., R.A.H., D.W.); Friends Re-search Institute (T.W.K., M.G., M.F.), the University of Baltimore, School of Crimi-nal Justice (T.W.K.), and Maryland Treat-ment Centers (M.F.) — all in Baltimore; the University of Pennsylvania (T.Y.B., J.W.C., C.P.O.) and the Philadelphia Vet-erans Affairs Medical Center ( J.W.C.) — both in Philadelphia; the Center for Bio-medical Ethics and Humanities, School of Medicine (D.T.C.) and the School of Law (R.J.B.), University of Virginia, Char-lottesville; and Washington State Univer-sity, Spokane (S.M.M.). Address reprint requests to Dr. Lee at the Department of Population Health, New York University, 227 E. 30th St., New York, NY 10016, or at joshua . lee@ nyumc . org.

This article was updated on April 14, 2016, at NEJM.org.

N Engl J Med 2016;374:1232-42.DOI: 10.1056/NEJMoa1505409Copyright © 2016 Massachusetts Medical Society.

BACKGROUNDExtended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor antagonist, is effective for the prevention of relapse to opioid depen-dence. Data supporting its effectiveness in U.S. criminal justice populations are limited.METHODSIn this five-site, open-label, randomized trial, we compared a 24-week course of extended-release naltrexone (Vivitrol) with usual treatment, consisting of brief counseling and refer-rals for community treatment programs, for the prevention of opioid relapse among adult criminal justice offenders (i.e., persons involved in the U.S. criminal justice system) who had a history of opioid dependence and a preference for opioid-free rather than opioid maintenance treatments and who were abstinent from opioids at the time of randomiza-tion. The primary outcome was the time to an opioid-relapse event, which was defined as 10 or more days of opioid use in a 28-day period as assessed by self-report or by testing of urine samples obtained every 2 weeks; a positive or missing sample was computed as 5 days of opioid use. Post-treatment follow-up occurred at weeks 27, 52, and 78.RESULTSA total of 153 participants were assigned to extended-release naltrexone and 155 to usual treatment. During the 24-week treatment phase, participants assigned to extended-release naltrexone had a longer median time to relapse than did those assigned to usual treatment (10.5 vs. 5.0 weeks, P<0.001; hazard ratio, 0.49; 95% confidence interval [CI], 0.36 to 0.68), a lower rate of relapse (43% vs. 64% of participants, P<0.001; odds ratio, 0.43; 95% CI, 0.28 to 0.65), and a higher rate of opioid-negative urine samples (74% vs. 56%, P<0.001; odds ratio, 2.30; 95% CI, 1.48 to 3.54). At week 78 (approximately 1 year after the end of the treatment phase), rates of opioid-negative urine samples were equal (46% in each group, P = 0.91). The rates of other prespecified secondary outcome mea-sures — self-reported cocaine, alcohol, and intravenous drug use, unsafe sex, and rein-carceration — were not significantly lower with extended-release naltrexone than with usual treatment. Over the total 78 weeks observed, there were no overdose events in the extended-release naltrexone group and seven in the usual-treatment group (P = 0.02).CONCLUSIONSIn this trial involving criminal justice offenders, extended-release naltrexone was asso-ciated with a rate of opioid relapse that was lower than that with usual treatment. Opioid-use prevention effects waned after treatment discontinuation. (Funded by the National Institute on Drug Abuse; ClinicalTrials.gov number, NCT00781898.)

A BS TR AC T

Extended-Release Naltrexone to Prevent Opioid Relapse in Criminal Justice Offenders

Joshua D. Lee, M.D., Peter D. Friedmann, M.D., M.P.H., Timothy W. Kinlock, Ph.D., Edward V. Nunes, M.D.,

Tamara Y. Boney, M.S., C.C.R.C., Randall A. Hoskinson, Jr., Donna Wilson, M.S., Ryan McDonald, M.A., John Rotrosen, M.D., Marc N. Gourevitch, M.D., M.P.H.,

Michael Gordon, D.P.A., Marc Fishman, M.D., Donna T. Chen, M.D., M.P.H., Richard J. Bonnie, L.L.B., James W. Cornish, M.D., Sean M. Murphy, Ph.D.,

and Charles P. O’Brien, M.D., Ph.D.

Original Article

The New England Journal of Medicine Downloaded from nejm.org at University at Buffalo Libraries on March 29, 2017. For personal use only. No other uses without permission.

Copyright © 2016 Massachusetts Medical Society. All rights reserved.

Page 2: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

n engl j med 374;13 nejm.org March 31, 2016 1233

Extended-Release Naltrexone in offenders

Opioid-use disorder is a chronic re-lapsing condition that has serious public health consequences. Opioid dependence

disproportionately affects U.S. criminal justice system populations, and relapse and overdose deaths occur at high rates after release from incarceration.1 Evidence-based opioid-agonist maintenance therapies for opioid dependence (methadone and buprenorphine) are effective in prison, jail, and community reentry (i.e., parole) settings2-5 but have historically been unavailable or discouraged among criminal justice clients.6-8 Extended-release naltrexone (Vivitrol, Alkermes), a sustained-release monthly injectable formula-tion of the full mu-opioid receptor antagonist, was approved by the Food and Drug Administra-tion in 2010 for the prevention of relapse to opioid dependence. Extended-release naltrexone may be particularly appealing and beneficial to patients and providers who are unlikely to access opioid-agonist maintenance treatment or who prefer a relapse-prevention medication. As a noncontrolled substance with no known abuse or diversion potential, extended-release naltrex-one has gained increasing acceptance in the criminal justice system despite limited data on effectiveness.

Extended-release naltrexone gradually releases sufficient naltrexone to block the euphoric ef-fects of opioids for approximately 1 month after injection and is efficacious as compared with placebo.9-12 A pilot study that was performed at the same five sites that participated in this trial used a single-group observational cohort design and showed the feasibility of using Depotrex, an alternative formulation of extended-release nal-trexone, as a treatment option for outpatient parolees and probationers.13 We conducted a large, multisite, randomized trial to examine the effectiveness of extended-release naltrexone among community-dwelling criminal justice of-fenders who were at high risk for opioid relapse and related adverse outcomes.

Me thods

Trial Design, Sites, and OversightThis open-label, randomized, controlled effec-tiveness trial compared six monthly injections of extended-release naltrexone (Vivitrol, Alkermes) with usual treatment (brief counseling and refer-rals for community treatment programs) for the prevention of opioid relapse among criminal

justice offenders. We hypothesized that the like-lihood of an opioid-relapse event would be low-er, the time to relapse longer, and overall rates of opioid use lower with extended-release nal-trexone than with usual treatment.

Five independently funded sites implemented a common collaborative protocol: University of Pennsylvania (Philadelphia), New York University School of Medicine and Bellevue Hospital Center (New York), Rhode Island Hospital and Brown University (Providence, Rhode Island), Columbia University Medical Center (New York), and Friends Research Institute (Baltimore). The University of Pennsylvania, which was the lead site, hosted the regulatory and data management cores and the data and safety monitoring board.

The rationale, protocol development, design, and methods of this trial are described in full elsewhere.14 All sites obtained approval from the local institutional review board and the U.S. Office for Human Research Protections for the com-mon trial protocol. The authors alone designed and implemented the trial; collected, accessed, and analyzed the data; and vouch for the com-pleteness and accuracy of the data and for the fidelity of the trial to the protocol, which is available with the full text of this article at NEJM.org. The first author wrote the initial draft of the manuscript, and all the authors partici-pated in revisions and approved the final draft. The sponsor (National Institute on Drug Abuse) and the manufacturer of extended-release nal-trexone (Alkermes) did not have editorial control or access to trial data. The manufacturer con-tributed Vivitrol in kind through an investigator-initiated trial contract, which allowed for review of and comment on the manuscript before sub-mission for publication.

ParticipantsWe recruited community-dwelling adult volunteers who were criminal justice offenders and who had a history of opioid dependence. Eligibility crite-ria were current (within the previous 12 months) or lifetime (any previous) opioid dependence (as defined by the Diagnostic and Statistical Manual of Mental Disorders, 4th edition [DSM-IV])15; a stated goal of opiate-free treatment rather than opioid-agonist or partial-agonist maintenance therapy; an opioid-free status as confirmed by negative urine toxicologic screening for all opioids before randomization; residence in the community and receipt of an adjudicated sentence that included

The New England Journal of Medicine Downloaded from nejm.org at University at Buffalo Libraries on March 29, 2017. For personal use only. No other uses without permission.

Copyright © 2016 Massachusetts Medical Society. All rights reserved.

Page 3: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

n engl j med 374;13 nejm.org March 31, 20161234

T h e n e w e ngl a nd j o u r na l o f m e dic i n e

supervision (e.g., parole, probation, outpatient drug-court programs, or other court-mandated treatment) or, in the previous 12 months, release from jail or prison, a plea-bargain arrangement, or any community supervision as above; general good health as determined by history and physi-cal examination; an age of 18 to 60 years; and the ability to provide written informed consent.

Exclusion criteria were other drug or alcohol dependence requiring a level of care that would interfere with trial participation; pregnancy or a plan to conceive during the 24-week treatment phase, lactation, or an inability to use adequate contraceptive methods; an untreated psychiatric disorder or medical condition that might make participation hazardous, including liver-enzyme levels more than three times the upper limit of the normal range and a body-mass index (BMI, the weight in kilograms divided by the square of the height in meters) of more than 40; allergy to nal-trexone, polylactide-co-glycolide, carboxymethyl-cellulose, or other components of the diluent; a current diagnosis of chronic pain for which opioids were prescribed; or a drug overdose in the previ-ous 3 years requiring inpatient hospitalization.

We recruited participants by standard out-reach to community-dwelling at-risk populations through print, radio, and online publicity and provider detailing (e.g., letters to clinic direc-tors); in an effort to minimize potential coer-cion, we did not recruit through direct referrals from criminal justice authorities, including de-partments of corrections, probation, or parole and drug courts or other diversion programs. Prescreening questionnaires were used to briefly evaluate potential participants and to schedule an in-person screening visit at which written informed consent was obtained; participants were required to establish their comprehension of consent information by passing an informed-consent quiz.

Randomization and Trial TreatmentsParticipants were randomly assigned, in a 1:1 ratio, to extended-release naltrexone or usual treatment for opioid-relapse prevention. An urn random-ization procedure ensured balance with respect to trial site, sex, and status regarding the need for opioid detoxification.16 An independent, cen-tralized, automated telephone system made the treatment assignments after eligibility of the participants was confirmed.17

Trial physicians or nurses administered extended-release naltrexone by injection and provided medication-management counseling. Extended-release naltrexone, at a dose of 380 mg, was administered by intramuscular injection once every 4 weeks during medical management visits, the first of which occurred at the time of ran-domization. A standard naloxone challenge (i.e., administration of >0.8 mg of naloxone intrave-nously, intramuscularly, or subcutaneously and assessment of opioid-withdrawal symptoms) had to be negative before the initial injection. At one site (Friends Research Institute), 12.5 mg of oral naltrexone was also administered as a low-dose challenge, followed by a 2-hour observation peri-od, before injection of extended-release nal-trexone; this additional challenge reflected the preferences of the local site and institutional review board. Medical management counseling focused on medication side effects, support for recovery and treatment participation, and coun-seling to reduce the risk of relapse and over-dose.18 Participants in the usual-treatment group received similar counseling that was focused on adverse events, the prevention of relapse and overdose, and support for community treatment involvement from the same trial personnel.

Incentives, follow-up visit schedules and pro-cedures, and community treatment referrals were the same in the two groups; all participants were encouraged by the same trial staff to access appropriate community treatment and relapse-prevention resources, including buprenorphine or methadone treatment if preferred or indicated during the trial and after the treatment phase (no extended-release naltrexone was provided after the end of the 24-week treatment phase). All participants were compensated for atten-dance at individual visits; total cash or voucher compensation across 17 visits varied according to site ($385 to $820).

Clinical AssessmentsFollow-up and assessment procedures were the same in the two groups. Visits occurred at screening, randomization, and then every 2 weeks for 24 weeks during the treatment phase. Post-treatment follow-up assessments occurred at weeks 27, 52, and 78 (three visits only). The visits occurring every 2 weeks and at weeks 27, 52, and 78 included urine toxicologic screening and self-report of opioid, cocaine, alcohol, and intrave-

The New England Journal of Medicine Downloaded from nejm.org at University at Buffalo Libraries on March 29, 2017. For personal use only. No other uses without permission.

Copyright © 2016 Massachusetts Medical Society. All rights reserved.

Page 4: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

n engl j med 374;13 nejm.org March 31, 2016 1235

Extended-Release Naltrexone in offenders

nous drug use with the use of the Timeline Followback calendar method for count data.19 Urine samples were tested for opiates (a level >300 ng per milliliter was considered to indicate a positive test), oxycodone, methadone, buprenor-phine, and cocaine metabolites. Data on unsafe sex were captured every 6 months with the use of the Sex Risk subscale of the Risk Assessment Battery (on which scores range from 0 to 18, with higher scores indicating a greater risk of contracting and spreading human immunodefi-ciency virus [HIV] infection through sexual be-haviors).20 Self-reported information about crim-inal activity, rearrests, and days of reincarceration was collected every 2 weeks with the use of the Timeline Followback method for days in con-trolled environments, monthly with the use of the Crime and Legal Activities Report,21 and every 6 months with the use of the legal-status items in the Addiction Severity Index Lite.22

OutcomesThe primary outcome was the time (in weeks) to an opioid-relapse event during the 24-week treat-ment phase. A relapse event was defined as 10 or more days of opioid use in a 28-day (4-week) period as assessed by self-report or by testing of urine samples obtained every 2 weeks; a positive or missing sample was computed as 5 days of opioid use. Relapse was considered to be a one-time event and corresponded to the loss of per-sistent opioid abstinence after randomization, when all participants had been opioid-free and had endorsed a goal of opioid abstinence. Re-lated opioid-use outcomes were rates of opioid-negative (vs. opioid-positive or missing) urine samples, the percentage of 2-week intervals with no opioid use as assessed by self-report or by testing of urine samples (confirmed abstinence), the percentage of days with self-reported opioid use, and post-treatment rates of opioid use as assessed by self-report (percentage of 2-week intervals with any opioid use vs. no opioid use) and by testing of single urine samples at weeks 52 and 78. The primary relapse outcome used during the treatment phase, defined by assess-ments performed every 2 weeks, self-report, and testing of urine samples, was not available dur-ing long-term follow-up because visits were scheduled only at weeks 52 and 78. Secondary outcomes of interest were rates of alcohol and nonopioid drug use, HIV risk behaviors, rearrests

and reincarcerations, and adverse events includ-ing opioid overdose.

Statistical AnalysisWe calculated that a sample size of 164 partici-pants per group, with an assumed loss to attri-tion of approximately 5% per month, would provide the trial with 80% power to detect a hazard ratio for relapse with usual treatment of 1.53 or higher, equivalent to an estimated differ-ence in relapse rates of 15 percentage points (45% vs. 30%).9,13 The primary outcome analysis tested whether extended-release naltrexone re-sulted in a longer time to relapse than that with usual treatment, with the use of Cox proportional-hazards regression models. We compared over-all relapse rates using intention-to-treat mixed-effects logistic-regression models and compared rates of positive urine tests and days with self-reported opioid use using a linear mixed-effects model for count data. Missed visits and missing data on urine samples were counted as positive for opioid use; thus, dropouts contributed to a relapse event. Missing data for secondary out-comes (cocaine, alcohol, and intravenous drug use; score on the Sex Risk subscale of the Risk Assessment Battery; and reincarceration) were estimated from available data only.

At the week 52 and week 78 visits, participants provided 6 months of self-reports on opioid use and a single urine sample. We analyzed the per-centage of opioid-negative (vs. opioid-positive or missing) urine samples at both visits using mixed-effects logistic models. We used a logistic mixed-effects model for repeated measures to analyze self-reported opioid use from week 1 to week 78 and to test for differences in the treat-ment groups over time with the use of an inter-action term between group and time.

R esult s

Screening and RandomizationRecruitment began in February 2009 and contin-ued through November 2013. The five sites ob-tained consent from and screened 437 persons, of whom 308 underwent randomization; 153 were assigned to extended-release naltrexone and 155 to usual treatment (Fig. 1). Common reasons for exclusion were an incomplete screen-ing visit, incomplete detoxification or a lack of opioid abstinence before randomization, and

The New England Journal of Medicine Downloaded from nejm.org at University at Buffalo Libraries on March 29, 2017. For personal use only. No other uses without permission.

Copyright © 2016 Massachusetts Medical Society. All rights reserved.

Page 5: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

n engl j med 374;13 nejm.org March 31, 20161236

T h e n e w e ngl a nd j o u r na l o f m e dic i n e

serious medical or psychiatric coexisting condi-tions. Two potential participants were excluded because of a BMI of more than 40 and therefore a potentially elevated risk of a severe injection-site reaction.

ParticipantsThe characteristics of the trial groups were similar at baseline. The mean age was 44 years; 85% of the participants were male, 77% were black or Hispanic, 74% were on parole or proba-tion, and 65% had not used heroin or other opioids in the previous 30 days (Table 1). All reported a history of DSM-IV opioid dependence. A total of 88% of the participants reported heroin use and 41% reported injection-drug use

during their lifetimes; 34% reported any opioid (heroin or other) use in the previous 30 days. A total of 9% of the participants required opioid detoxification to enter the trial.

Attendance at Scheduled Visits and Adherence to Medication

Most of the visits that were scheduled every 2 weeks (3096 of 4004, 77%) were attended; participants assigned to extended-release nal-trexone attended 79% of the scheduled visits, and those assigned to usual treatment attended 75%. A total of 75% of the participants com-pleted an end-of-treatment-phase visit at week 27. Overall, participants assigned to extended-release naltrexone completed 711 of the 918 planned monthly injections (77%). Seven participants (5%) declined any injections after randomiza-tion; 146 (95%) completed the first injection, 132 (86%) the second injection, 119 (78%) the third injection, 111 (73%) the fourth injection, 100 (65%) the fifth injection, and 93 (61%) the sixth injection.

Primary Outcome and Related Opioid-Use Outcomes

During the 24-week treatment phase, the time to relapse was significantly longer in the extended-release naltrexone group than in the usual-treatment group: 10.5 weeks versus 5.0 weeks (P<0.001; hazard ratio for relapse, 0.49; 95% confidence interval [CI], 0.36 to 0.68) (Fig. 2). A relapse event was detected in 66 participants assigned to extended-release naltrexone (43%) as compared with 99 assigned to usual treatment (64%) (P<0.001; odds ratio, 0.43; 95% CI, 0.28 to 0.65); this finding is consistent with the higher rate of opioid-negative urine samples, the lower percentage of days with self-reported opioid use, and the higher percentage of 2-week intervals with confirmed abstinence that we observed with extended-release naltrexone than with usual treatment (Table 2). An alternative analysis of missing urine data, in which only two consecutive confirmed positive urine screening results or self-reports of opioid use contributed to a “con-firmed relapse outcome,” also favored extended-release naltrexone (rate of relapse, 15% vs. 37%; P<0.001; hazard ratio, 0.33; 95% CI, 0.21 to 0.54). The treatment effect did not differ significantly according to site.

Figure 1. Screening, Randomization, and Follow-up.

308 Underwent randomization

437 Potential participants wereassessed for eligibility

129 Were excluded57 Had incomplete screening25 Were not abstinent from

opioids19 Had medical or psychiatric

reasons3 Had recent drug overdose2 Had a body-mass index >40

23 Had other reasons

153 Were assigned to receive extended-release naltrexone

146 Received intervention7 Declined to receive intervention

119 Completed 24-wk treatment phase follow-up

7 Were lost to follow-up8 Withdrew consent1 Had an adverse event1 Changed residence

13 Were incarcerated4 Had other reasons

155 Were assigned to receive usual treatment

155 Received intervention

126 Completed 24-wk treatment phasefollow-up

4 Were lost to follow-up1 Withdrew consent2 Died5 Changed residence

15 Were incarcerated2 Had other reasons

153 Were included in primary analysis 155 Were included in primary analysis

The New England Journal of Medicine Downloaded from nejm.org at University at Buffalo Libraries on March 29, 2017. For personal use only. No other uses without permission.

Copyright © 2016 Massachusetts Medical Society. All rights reserved.

Page 6: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

n engl j med 374;13 nejm.org March 31, 2016 1237

Extended-Release Naltrexone in offenders

Secondary OutcomesSeveral important secondary outcomes did not differ significantly between the groups: rates of cocaine, alcohol, and intravenous drug use; the score on the Sex Risk subscale of the Risk As-

sessment Battery; and self-reported reincarcera-tion (percentage of participants with any reincar-ceration and total days of incarceration) (Table 3). Participation in other, nontrial treatment in the community for opioid-use disorders did not dif-

Characteristic

Extended-Release Naltrexone (N = 153)

Usual Treatment (N = 155)

Age — yr 44.4±9.2 43.2±9.4

Male sex — no. (%) 129 (84.3) 132 (85.2)

Race or ethnic group — no./total no. (%)†

White 31/152 (20.4) 30/155 (19.4)

Black 81/152 (53.3) 74/155 (47.7)

Hispanic 37/152 (24.3) 45/155 (29.0)

Years of education 11.5±2.2 11.5±1.8

Current employment — no. (%) 26 (17.0) 29 (18.7)

Status with respect to supervision by criminal justice system — no. (%)

Current supervision‡ 121 (79.1) 124 (80.0)

Probation 55 (35.9) 62 (40.0)

Parole 57 (37.3) 54 (34.8)

Other 9 (5.9) 8 (5.2)

No supervision§ 32 (20.9) 31 (20.0)

Health insurance

Any 109 (71.2) 111 (71.6)

Medicaid 70 (45.8) 65 (41.9)

Opioid use during lifetime — no./total no. (%)

Opioid dependence¶ 153/153 (100) 155/155 (100)

Heroin use 135/152 (88.8) 137/155 (88.4)

Other, non-heroin, opioid use 77/152 (50.7) 74/155 (47.7)

Injection-drug use 64/152 (42.1) 62/155 (40.0)

Opioid use in past 30 days — no./total no. (%)

Heroin use 32/152 (21.1) 43/155 (27.7)

Other, non-heroin, opioid use 31/152 (20.4) 26/155 (16.8)

Any opioid use 47/152 (30.9) 59/155 (38.1)

Needed opioid detoxification to enter trial — no. (%) 13 (8.5) 14 (9.0)

Cocaine use in past 30 days — no./total no. (%) 30/152 (19.7) 29/155 (18.7)

Heavy alcohol use in past 30 days — no. (%) 18 (11.8) 19 (12.3)

* Plus–minus values are means ±SD. There were no significant differences between the two groups at baseline.† Race and ethnic group were self-reported.‡ Current supervision by the criminal justice system was defined as parole, probation, or other involvement (drug court,

diversion, or alternative sentencing program) at baseline.§ No supervision by the criminal justice system was defined as recent criminal justice involvement (i.e., arrest, incarcera-

tion, conviction, or plea bargain) 0 to 12 months before baseline, with no current supervision as above.¶ Opioid dependence was defined according to the Diagnostic and Statistical Manual of Mental Disorders, 4th edition.

Table 1. Baseline Characteristics of the Participants.*

The New England Journal of Medicine Downloaded from nejm.org at University at Buffalo Libraries on March 29, 2017. For personal use only. No other uses without permission.

Copyright © 2016 Massachusetts Medical Society. All rights reserved.

Page 7: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

n engl j med 374;13 nejm.org March 31, 20161238

T h e n e w e ngl a nd j o u r na l o f m e dic i n e

fer significantly between the groups (62% of the participants in the extended-release naltrexone group and 65% of the participants in the usual-treatment group, P = 0.63). More participants in the usual-treatment group than in the extended-release naltrexone group pursued opioid-agonist treatments during the trial (37% vs. 11%, P<0.001), primarily after resumed illicit opioid use and relapse. No participants reported con-tinuing extended-release naltrexone after the treatment phase; the drug was approved for the prevention of opioid relapse mid-trial and was

not widely available in public-sector facilities during the treatment phase.

Safety and Long-Term Opioid-Use OutcomesAdverse events, including medication-related ad-verse events, were more common among par-ticipants assigned to extended-release naltrexone than among those assigned to usual treatment (Table 4); however, significantly more serious adverse events occurred in the usual-treatment group than in the extended-release naltrexone group. All recorded overdose events, fatal or non-fatal, occurred among participants assigned to usual treatment (0 events in the extended-release naltrexone group vs. 5 in the usual-treatment group from week 0 to 25, P = 0.10; 0 vs. 7 events from week 0 to 78, P = 0.02); no overdoses oc-curred in the extended-release naltrexone group after discontinuation of the agent. The percent-age of participants with opioid-negative urine samples was similar in the two groups after completion of the treatment phase: 49% in the extended-release naltrexone group and 46% in the usual-treatment group at week 52 (P = 0.61), and 46% in both groups at week 78 (P = 0.91). Self-reported opioid use showed an interaction be-tween treatment and time: participants assigned to extended-release naltrexone reported less use than those assigned to usual treatment during the active treatment phase, but the increase after the active treatment phase was greater in the extended-release naltrexone group, with the re-sult that opioid use was similar in the two groups

Figure 2. Kaplan–Meier Curves for Relapse-free Survival.

Prob

abili

ty o

f Rel

apse

-free

Sur

viva

l 1.0

0.8

0.6

0.2

0.4

0.01 3 5 7 9 11 2519 21 23171513

Week

No. at RiskExtended-release

naltrexoneUsual treatment

153

155

144

116

139

104

129

96

121

84

117

76

112

72

110

67

104

65

100

61

92

59

87

56

87

56

Usual treatment

Extended-release naltrexone

Outcome

Extended-Release Naltrexone (N = 153)

Usual Treatment (N = 155) P Value

Hazard Ratio, Odds Ratio, or Incidence-Density Ratio

(95% CI)

Primary outcome: median time to relapse — wk† 10.5 5.0 <0.001 0.49 (0.36–0.68)‡

Opioid-relapse event — no. (%) 66 (43.1) 99 (63.9) <0.001 0.43 (0.28–0.65)§

Percentage of 2-wk intervals with confirmed abstinence 71.1 49.5 <0.001 2.50 (1.66–3.76)¶

Percentage of opioid-negative urine samples 74.1 55.7 <0.001 2.30 (1.48–3.54)¶

Percentage of days with self-reported opioid use 4.6 12.7 0.02 0.35 (0.21–0.59)∥

* CI denotes confidence interval.† Shown is the median time to relapse if a relapse event occurred.‡ Shown is the hazard ratio for relapse, calculated with a Cox proportional-hazards model.§ Shown is the odds ratio calculated with a generalized estimating equation (GEE) mixed-effects logistic-regression model, with trial site as

the repeated measure.¶ Shown is the odds ratio calculated with a GEE mixed-effects logistic-regression model for repeated measures, with participant as the repeat-

ed measure and with adjustment for trial site and week.∥ Shown is the incidence-density ratio calculated with a GEE mixed-effects Poisson regression model, with trial site as the repeated measure

and with adjustment for log days at risk.

Table 2. Opioid Relapse and Related Outcomes during the 24-Week Treatment Phase.*

The New England Journal of Medicine Downloaded from nejm.org at University at Buffalo Libraries on March 29, 2017. For personal use only. No other uses without permission.

Copyright © 2016 Massachusetts Medical Society. All rights reserved.

Page 8: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

n engl j med 374;13 nejm.org March 31, 2016 1239

Extended-Release Naltrexone in offenders

through week 52 and week 78. (Additional infor-mation is provided in Fig. S1A and S1B in the Supplementary Appendix, available at NEJM.org.)

Discussion

This U.S. multisite, randomized, controlled trial showed that extended-release naltrexone result-ed in a lower rate of opioid relapse than the rate with usual treatment in a predominantly male and minority population of outpatient, voluntary participants with criminal justice involvement and opioid abstinence at baseline. During a 24-week treatment phase, the percentage of partici-pants with a relapse event was lower among participants assigned to extended-release nal-trexone than among those assigned to usual treatment (43% vs. 64%), corresponding to an absolute difference in risk of 21 percentage points and a number needed to treat of 5. The

prevention of opioid use by extended-release naltrexone did not persist through follow-up at week 52 and week 78, approximately 6 months and 12 months, respectively, after the treatment phase had ended. In addition, we did not detect a benefit of extended-release naltrexone on sev-eral important secondary outcomes, including rates of cocaine, heavy alcohol, and injection-drug use. Rates of self-reported reincarceration and days of incarceration through week 27 were also not significantly lower in the extended-release naltrexone group than in the usual-treat-ment group.

There was not an increased risk of overdose events during treatment with extended-release naltrexone or immediately after its discontinua-tion; indeed, no overdose events were observed among participants assigned to extended-release naltrexone through 78 weeks, during which time seven overdose events occurred among partici-

Outcome

Extended-Release Naltrexone (N = 153)

Usual Treatment (N = 155) P Value

Odds Ratio or Incidence-Density Ratio

(95% CI)

Percentage of days with cocaine use† 3.9 4.3 0.71 0.91 (0.56–1.48)‡

Heavy drinking in past 30 days at wk 27 — no. (%)§

16 (13.9) 17 (14.9) 0.77 0.89 (0.43–1.87)¶

Any intravenous drug use — %∥ 7 (5.9) 10 (8.6) 0.43 0.67 (0.25–1.82)¶

Mean score on Sexual Risk subscale of RAB at wk 27**

2.75 2.86 0.68††

Any reincarceration — no. (%) 35 (22.9) 45 (29.0) 0.38 0.71 (0.33–1.52)‡‡

Total days of reincarceration 1651 2628 0.22 0.63 (0.32–1.23)‡

Days incarcerated

Among participants with any reincarceration

47.2±41.8 58.4±51.4 0.30††

Among all participants 11.1±28.4 17.6±38.9 0.10††

* Plus–minus values are means ±SD.† Data are based on self-report for weeks 1 to 25 with the use of the Timeline Followback calendar method. No

Timeline Followback data were available for 4 participants in the extended-release naltrexone group and 6 participants in the usual-treatment group.

‡ Shown is the incidence-density ratio calculated with a GEE mixed-effects Poisson regression model, with trial site as the repeated measure and adjustment for log days at risk.

§ Data on self-reported heavy drinking for weeks 1 to 25 were missing for 38 participants in the extended-release nal-trexone group and 41 participants in the usual-treatment group.

¶ Shown is the odds ratio calculated with a chi-square test.∥ Data on self-reported intravenous drug use for weeks 1 to 25 were missing for 35 participants in the extended-release

naltrexone group and 39 participants in the usual-treatment group.** Scores on the Sex Risk subscale of the Risk Assessment Battery (RAB) range from 0 to 18, with higher scores indicat-

ing a greater risk of contracting and spreading HIV infection through sexual behaviors.†† The P value was calculated with a t-test.‡‡ Shown is the odds ratio calculated with a GEE mixed-effects logistic-regression model, with trial site as the repeated

measure and adjustment for log days at risk.

Table 3. Secondary Outcomes.*

The New England Journal of Medicine Downloaded from nejm.org at University at Buffalo Libraries on March 29, 2017. For personal use only. No other uses without permission.

Copyright © 2016 Massachusetts Medical Society. All rights reserved.

Page 9: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

n engl j med 374;13 nejm.org March 31, 20161240

T h e n e w e ngl a nd j o u r na l o f m e dic i n e

Event

Extended-Release Naltrexone (N = 153)

Usual Treatment (N = 155) P Value

no. of participants (%)

≥1 Adverse event 119 (77.8) 90 (58.1) <0.001

Adverse event related to trial drug

Total 59 (38.6) NA

Leading to discontinuation of trial drug 5 (3.3) NA

Injection-site reaction

Mild or moderate 42 (27.5) NA

Severe 4 (2.6) NA

Headache 29 (19.0) 13 (8.4) 0.007

Gastrointestinal upset 28 (18.3) 3 (1.9) <0.001

Nasopharyngitis 15 (9.8) 17 (11.0) 0.74

Insomnia 11 (7.2) 8 (5.2) 0.46

Flulike symptoms 6 (3.9) 2 (1.3) 0.17

Depression 2 (1.3) 11 (7.1) 0.02

Serious adverse events† 16 (10.5)‡ 45 (29.0)§ 0.006

Depression or suicidality 3 (2.0) 6 (3.9)

Death 2 (1.3) 5 (3.2)

Chest pain 2 (1.3) 4 (2.6)

Musculoskeletal symptom 2 (1.3) 4 (2.6)

Stroke 2 (1.3) 0

COPD 0 5 (3.2)

Abscess 0 4 (2.6)

Nonfatal overdose 0 4 (2.6)

Exacerbation of preexisting condition 0 3 (1.9)

Surgery for colon cancer 0 2 (1.3)

Any overdose 0 7 (4.5) 0.02

Weeks 1–27 0 5 (3.2) 0.06

Weeks 28–78 0 2 (1.3) 0.50

Fatal overdose 0 3 (1.9) 0.25

Weeks 1–27 0 2 (1.3) 0.50

Weeks 28–78 0 1 (0.6) 1.00

Deaths 2 (1.3) 5 (3.2) 0.45

Weeks 1–27 0 2 (1.3) 0.50

Weeks 28–78 2 (1.3) 3 (1.9) 1.00

* COPD denotes chronic obstructive pulmonary disease, and NA not applicable.† A serious adverse event was defined as an adverse event that was life-threatening or that resulted in death, hospitaliza-

tion or prolongation of hospitalization, persistent or clinically significant disability or incapacity, a congenital anomaly or birth defect, or an important medical event. Included in the table are serious adverse events that affected two or more participants in a trial group.

‡ Serious adverse events in the extended-release naltrexone group also included one event each of abdominal pain, dehy-dration, acute renal failure, opioid withdrawal related to the trial drug, and hospitalization of unknown cause. Of the two deaths, one each was caused by homicide and seizure.

§ Serious adverse events in the usual-treatment group also included one event each of abdominal pain, asthma, cancer, influenza, hypertension, hypoglycemia, pancreatitis, and pneumonia. Of the five deaths, three were caused by overdose and one each was caused by homicide and cardiopulmonary arrest. Of the three events related to a preexisting condi-tion (i.e., resulting from exacerbation or treatment of a preexisting condition), one each was related to deep-vein thrombosis, diabetes, and musculoskeletal injury.

Table 4. Adverse Events and Serious Adverse Events.*

The New England Journal of Medicine Downloaded from nejm.org at University at Buffalo Libraries on March 29, 2017. For personal use only. No other uses without permission.

Copyright © 2016 Massachusetts Medical Society. All rights reserved.

Page 10: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

n engl j med 374;13 nejm.org March 31, 2016 1241

Extended-Release Naltrexone in offenders

pants assigned to usual treatment. Product label-ing and expert reviews describe an increased risk of opioid overdose among patients receiving extended-release naltrexone, both during treat-ment, when a patient may “super-dose” opioids to overcome the naltrexone blockade, or after therapy, when tolerance is low and resumption of opioid use is even more dangerous.23,24

The trial groups did not differ significantly at baseline in terms of demographic characteristics and criminal justice status, and results were not modified by site, missed visits, or missing data. Main relapse-prevention effects were similar to those in the placebo-controlled pivotal efficacy trial, in which the median percentage of weeks of confirmed abstinence was 90% in the extend-ed-release naltrexone group, as compared with 35% in the placebo group.12 On the basis of self-reports and a single urine sample at week 52 and week 78, months after extended-release naltrex-one therapy had ended, the relapse-prevention effects had waned. As is the case with any chronic illness, symptoms of opioid-use disorder are more likely to recur with the discontinuation of effective pharmacotherapy.25 Future research would be needed to determine whether long-term or continuous treatment with extended-release naltrexone — similar to buprenorphine or methadone maintenance therapy — could help maintain the short-term benefits observed in this trial and improve longer-term outcomes.

This trial had important limitations. First, the trial was not blinded. Although an open-label effectiveness design may increase generalizabil-ity, it may also increase attention, recall, and assessment biases. The agreement between the results of this trial and those of other recent trials is reassuring with respect to validity.12,26 Second, sites were in the U.S. Northeast corridor, and participants were former or current heroin users; implications for other U.S. regions and prescrip-tion-opioid disorders are unclear. Third, eligibility criteria did not distinguish among levels of pre-vious opioid use or grades of severity of opioid-use disorder; randomization was stratified ac-

cording to whether or not detoxification was required before randomization. The majority of participants enrolled in this trial had not used opioids in the previous 30 days and did not re-quire detoxification. Fourth, intensity and meth-ods of community supervision varied according to state and site (e.g., parolees in home confine-ment vs. mandated residential treatment), though the main effects did not vary according to site. Finally, this trial did not directly compare extended-release naltrexone with the standard of opioid-agonist maintenance treatment, a com-parison that is being assessed in an ongoing trial (ClinicalTrials.gov number, NCT02032433).

In summary, this U.S. multisite, open-label, randomized effectiveness trial showed that among adult offenders who had a history of opioid dependence, the rate of relapse was lower among participants assigned to extended-release naltrexone than among those assigned to usual treatment.

Supported by the National Institute on Drug Abuse (NIDA) through a collaborative clinical trial mechanism, PAR-07-232 (R01DA024549, to Dr. Friedmann; R01DA024550, to Dr. Kinlock; R01DA024553, to Dr. O’Brien; R01DA024554, to Dr. Nunes; and R01DA024555, to Dr. Lee), and additional support (K24DA022412, to Dr. Nunes). Trial medication was provided in kind from an investigator-initiated grant from Alkermes. Funding from the Dana Foundation to Dr. O’Brien supported the conduct of a five-site pilot study.

Dr. Lee reports receiving grant support and study medication from Alkermes and study medication from Indivior (formerly Reckitt Benckiser). Dr. Friedmann reports receiving fees for serv-ing on an advisory board and travel support from Indivior and an honorarium for leading a roundtable discussion from Orexo. Dr. Kinlock reports receiving grant support and study medication from Alkermes. Dr. Nunes reports serving on an advisory board for Alkermes, receiving study medication from Reckitt Benckiser and Duramed Pharmaceuticals, being lead investiga-tor for a NIDA-funded study of a computer-delivered behavioral intervention supplied by HealthSim, and being site principal in-vestigator for a study funded by, and receiving travel support from, Brainsway. Dr. Rotrosen reports receiving study medica-tion from Alkermes and Indivior. Dr. Gordon reports receiving grant support and study medication from Alkermes. Dr. Fishman reports receiving travel support from Alkermes. Dr. O’Brien re-ports receiving consulting fees from Alkermes. No other poten-tial conflict of interest relevant to this article was reported.

Disclosure forms provided by the authors are available with the full text of this article at NEJM.org.

We thank the trial participants and combined research staff across all sites.

References1. Binswanger IA, Blatchford PJ, Mueller SR, Stern MF. Mortality after prison re-lease: opioid overdose and other causes of death, risk factors, and time trends from 1999 to 2009. Ann Intern Med 2013; 159: 592-600.

2. Magura S, Lee JD, Hershberger J, et al. Buprenorphine and methadone mainte-nance in jail and post-release: a random-ized clinical trial. Drug Alcohol Depend 2009; 99: 222-30.3. Gordon MS, Kinlock TW, Schwartz

RP, et al. Buprenorphine treatment for probationers and parolees. Subst Abus 2015; 36: 217-25.4. Gordon MS, Kinlock TW, Schwartz RP, Fitzgerald TT, O’Grady KE, Vocci FJ. A randomized controlled trial of prison-

The New England Journal of Medicine Downloaded from nejm.org at University at Buffalo Libraries on March 29, 2017. For personal use only. No other uses without permission.

Copyright © 2016 Massachusetts Medical Society. All rights reserved.

Page 11: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

n engl j med 374;13 nejm.org March 31, 20161242

Extended-Release Naltrexone in offenders

initiated buprenorphine: prison outcomes and community treatment entry. Drug Alcohol Depend 2014; 142: 33-40. 5. Gordon MS, Kinlock TW, Schwartz RP, O’Grady KE. A randomized clinical trial of methadone maintenance for pris-oners: findings at 6 months post-release. Addiction 2008; 103: 1333-42.6. Lee JD, Rich JD. Opioid pharmaco-therapy in criminal justice settings: now is the time. Subst Abus 2012; 33: 1-4.7. Chandler RK, Fletcher BW, Volkow ND. Treating drug abuse and addiction in the criminal justice system: improving public health and safety. JAMA 2009; 301: 183-90.8. Nunn A, Zaller N, Dickman S, Trim-bur C, Nijhawan A, Rich JD. Methadone and buprenorphine prescribing and refer-ral practices in US prison systems: results from a nationwide survey. Drug Alcohol Depend 2009; 105: 83-8.9. Comer SD, Collins ED, Kleber HD, Nuwayser ES, Kerrigan JH, Fischman MW. Depot naltrexone: long-lasting antagonism of the effects of heroin in humans. Psy-chopharmacology (Berl) 2002; 159: 351-60.10. Bigelow GE, Preston KL, Schmittner J, Dong Q, Gastfriend DR. Opioid challenge evaluation of blockade by extended-release naltrexone in opioid-abusing adults: dose-effects and time-course. Drug Alcohol Depend 2012; 123: 57-65.11. Comer SD, Sullivan MA, Yu E, et al. Injectable, sustained-release naltrexone for the treatment of opioid dependence: a randomized, placebo-controlled trial. Arch Gen Psychiatry 2006; 63: 210-8.

12. Krupitsky E, Nunes EV, Ling W, Illepe-ruma A, Gastfriend DR, Silverman BL. Injectable extended-release naltrexone for opioid dependence: a double-blind, place-bo-controlled, multicentre randomised trial. Lancet 2011; 377: 1506-13.13. Coviello DM, Cornish JW, Lynch KG, et al. A multisite pilot study of extended-release injectable naltrexone treatment for previously opioid-dependent parolees and probationers. Subst Abus 2012; 33: 48-59.14. Lee JD, Friedmann PD, Boney TY, et al. Extended-release naltrexone to prevent relapse among opioid dependent, criminal justice system involved adults: rationale and design of a randomized controlled effectiveness trial. Contemp Clin Trials 2015; 41: 110-7.15. Diagnostic and statistical manual of mental disorders: DSM-IV. 4th ed. Wash-ington, DC: American Psychiatric Associ-ation, 2000.16. Stout RL, Wirtz PW, Carbonari JP, Del Boca FK. Ensuring balanced distribution of prognostic factors in treatment out-come research. J Stud Alcohol Suppl 1994; 12: 70-5.17. U.S. Department of Veterans Affairs Office of Research and Development, CSP Coordinating Center — Perry Point, MD (http://www .research .va .gov/ programs/ csp/ perrypoint .cfm).18. Lee JD, Grossman E, DiRocco D, et al. Extended-release naltrexone for treatment of alcohol dependence in primary care. J Subst Abuse Treat 2010; 39: 14-21.19. Sobell LC, Sobell MB. Alcohol time-

line followback users’ manual. Toronto: Addiction Research Foundation, 1995.20. Watkins KE, Metzger D, Woody G, McLellan AT. High-risk sexual behaviors of intravenous drug users in- and out-of-treatment: implications for the spread of HIV infection. Am J Drug Alcohol Abuse 1992; 18: 389-98.21. Nurco DN, Kinlock T, Balter MB. The severity of preaddiction criminal behavior among urban, male narcotic addicts and two nonaddicted control groups. J Res Crime Delinq 1993; 30: 293-316.22. McLellan AT, Kushner H, Metzger D, et al. The fifth edition of the Addiction Severity Index. J Subst Abuse Treat 1992; 9: 199-213.23. Alkermes. Full prescribing informa-tion: VIVITROL (naltrexone for extended-release injectable suspension), revised July 2013 (http://www .accessdata .fda .gov/ drugsatfda_docs/ label/ 2013/ 021897s020s023lbl .pdf).24. Kjome KL, Moeller FG. Long-acting injectable naltrexone for the management of patients with opioid dependence. Subst Abuse 2011; 5: 1-9.25. McLellan AT, Lewis DC, O’Brien CP, Kleber HD. Drug dependence, a chronic medical illness: implications for treatment, insurance, and outcomes evaluation. JAMA 2000; 284: 1689-95.26. Lee JD, McDonald R, Grossman E, et al. Opioid treatment at release from jail us-ing extended-release naltrexone: a pilot proof-of-concept randomized effective-ness trial. Addiction 2015; 110: 1008-14.Copyright © 2016 Massachusetts Medical Society.

SHARING DATA IN A PUBLIC HEALTH EMERGENCYThe case for sharing data, and the consequences of not doing so, have been brought into stark relief by the Ebola and Zika

outbreaks. In response, the New England Journal of Medicine has become a journal signatory to the following statement.

“In the context of a public health emergency of international concern, it is imperative that all parties make available any information that might have value in combatting the crisis. As research funders and journals, we are committed to working in partnership to

ensure that the global response to public health emergencies is informed by the best available research evidence and data.

Journal signatories will make all content concerning the Zika virus free access. Any data or preprint deposited for unrestricted dissemination ahead of submission of any paper will not preempt later publication in these journals.

Funder signatories will require researchers undertaking work relevant to public health emergencies to establish mechanisms to share quality-assured interim and final data as rapidly and widely as possible, including with public health and research

communities and the World Health Organization.

We urge other journals and research funders to make the same commitments.”

The New England Journal of Medicine Downloaded from nejm.org at University at Buffalo Libraries on March 29, 2017. For personal use only. No other uses without permission.

Copyright © 2016 Massachusetts Medical Society. All rights reserved.

Page 12: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

Injectable extended-release naltrexone (XR-NTX) foropioid dependence: long-term safety and effectiveness

Evgeny Krupitsky1, Edward V. Nunes2, Walter Ling3, David R. Gastfriend4, Asli Memisoglu4 &Bernard L. Silverman4

Bekhterev Research Psychoneurological Institute and St. Petersburg State Pavlov Medical University, St. Petersburg, Russian Federation,1 NewYork State PsychiatricInstitute and Department of Psychiatry, Columbia University, New York City, NY, USA,2 Department of Psychiatry and Biobehavioral Sciences, University ofCalifornia Los Angeles, Los Angeles, CA, USA3 and Alkermes, Inc., Waltham, MA, USA4

ABSTRACT

Aims To describe drug use and safety with intramuscular injectable extended-release naltrexone (XR-NTX) in opioiddependence during a 1-year open-label extension phase. Design Following 6 months of randomized, double-blind,placebo (PBO)-controlled injections given every 28 days, patients receiving XR-NTX 380 mg continued andPBO patients were switched to open-label XR-NTX, with monthly individual drug counseling, for a furtheryear. Setting Thirteen clinical sites in Russia. Participants Adult opioid-dependent outpatients. MeasurementsMonthly urine samples; reports of craving and functioning; adverse events. Findings For the open-label extension(n = 114), 67 continued on XR-NTX and 47 switched from PBO during the double-blind phase to XR-NTX during theopen-label phase. Overall, 62.3% (95% CI: 52.7%, 71.2%) completed the extension. Discontinuation occurred mostcommonly because of withdrawal of consent (18.4%) and loss to follow-up (11.4%); two patients discontinued as aresult of lack of efficacy and one because of adverse events. Urine testing revealed that 50.9% (41.5%, 60.4%) wereabstinent from opioids at all assessments during the 1-year open-label phase. Adverse events reported by 21.1%of patients were judged to be study drug-related. Injection site reactions were infrequent (6.1%) and the majoritywere mild. Elevations in liver function tests occurred for 16.7% of patients, but none of these elevations wasjudged to be clinically significant. No patients died, overdosed or discontinued as a result of severe adverse events.Conclusions During a 1-year open-label extension phase of injectable XR-NTX for the prevention of relapse in opioiddependence, 62.3% of patients completed the phase and 50.9% were abstinent from opioids. No new safety concernswere evident.

Keywords Craving, depot naltrexone, extended-release naltrexone, heroin dependence, injectable naltrexone,opioid dependence, long-term safety, naltrexone, sustained release formulations.

Correspondence to: Evgeny Krupitsky, Department of Addictions, St.-Petersburg Bekhterev Psychoneurological Research Institute, Bekhtereva Street, 3,St. Petersburg 192019, Russia. E-mail: [email protected] 26 September 2012; initial review completed 20 November 2012; final version accepted 27 March 2013

INTRODUCTION

The 2009 National Survey on Drug Use and Health esti-mated that approximately 1.5 million Americans aged 18years or older were dependent on opioids in the prior year,including 345 000 dependent on heroin and 1 255 000on prescription opioid medications used non-medically[1]. Rates of opioid dependence throughout the restof the world have been on the increase [2]. Opioiddependence is a major public health concern because ofincreased morbidity and mortality, poor social function-ing, unemployment, and crime associated with thisdisorder [3–5].

Opioid dependence is a chronic disorder requir-ing long-term treatment [6,7]. Effective options formanaging the disorder include several pharmaco-therapy agents (methadone, buprenorphine, naltrexone)and psychosocial interventions [8–13]. However,relapse following cessation of treatment is high, withonly an estimated 25% of heroin-dependent indivi-duals remaining abstinent after receiving methadonetreatment [14]. Relapse following non-compliancewith oral naltrexone is a particular concern [9].Episodes of opioid use during non-compliance havebeen associated with relapse to full opioid dependence[15].

RESEARCH REPORT

bs_bs_banner

doi:10.1111/add.12208

© 2013 Society for the Study of Addiction Addiction, 108, 1628–1637

Page 13: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

Concerns about compliance with oral naltrexone ledto the development of a once-monthly extended-releaseformulation of injectable naltrexone (XR-NTX; Vivitrol®;Alkermes, Inc., Waltham, MA, USA). In this formulation,naltrexone is gradually released from microspheres com-posed of poly-(d,l-lactide-co-glycolide), a polymer used indissolvable surgical sutures. The efficacy of XR-NTX forthe prevention of relapse to opioid dependence followingdetoxification was recently demonstrated in a multi-center, placebo (PBO)-controlled, randomized clinicaltrial [16]. This study reported a median of 90% confirmedabstinent weeks for XR-NTX versus 35% for PBO overthe course of 6 months of treatment (P = 0.0002) with57.9% (73/126) of XR-NTX patients versus 41.9% (52/124) of PBO patients receiving all six double-blind doses.XR-NTX also has demonstrated efficacy in the treatmentof alcohol dependence [17], and is now approved in theUSA and Russia for both dependencies.

Although XR-NTX has shown efficacy for opioiddependence in the context of a 6-month study, thechronic, relapsing nature of this disorder has led toquestions regarding long-term treatment, specifically:Are initial treatment gains from baseline to end of thedouble-blind phase maintained over time during a1-year open-label extension?; What proportion ofpatients continue?; Do any new safety concerns becomeevident? This study reports descriptively on the resultsof a 1-year open-label treatment phase that followedthe initial 6-month double-blind phase in terms of dura-bility of improvements seen in the initial 6-monthperiod, patient retention and safety of XR-NTX for thetreatment of opioid dependence.

METHODS

Overview

The current study reports the results from a 52-weekextension study that followed the initial 24-weekrandomized, double-blind, PBO-controlled, multi-siteinvestigation of XR-NTX as a treatment for opioiddependence [16]. In the extension phase, patients whohad received XR-NTX during the initial 24-week periodcontinued on open-label XR-NTX for an additional52 weeks. Patients receiving PBO during the initial24-week treatment period were switched to open-labelXR-NTX for the next 52 weeks. The study was conductedbetween July 2008 and November 2010 at 13 clinicalsites in Russia. At each of the participating sites, an inde-pendent ethics committee/institutional review boardapproved the protocol and participants gave written,informed consent in accordance with the HelsinkiAccords. The open-label extension study was conductedfrom June 2008 to November 2012.

Participants

In the initial 6-month double-blind phase the studyrecruited males and females (!18 years) meeting Diag-nostic and Statistical Manual of Mental Disorders (fourthedition) [18] criteria for opioid (primarily heroin)dependence disorder who were voluntarily seeking treat-ment and had completed inpatient opioid detoxification("30 days). Patients were excluded if they had takenany opioids for !7 days prior to screening or if they wereunder justice system coercion (i.e. parole or probation, orpending legal proceedings with potential for incarcera-tion). To participate, it was required that patients involvea significant other (e.g. spouse, relative) who wouldsupervise the patient’s compliance with the visit sched-ule and study procedures. Women of childbearingpotential agreed to use contraception while participatingin the study. Patients did not receive reimbursements forparticipating in the study, but did receive reimburse-ments for transportation. Patients were excluded if theywere pregnant or breastfeeding, or had any of the fol-lowing: significant medical conditions; positive naloxonechallenge (appearance of vital sign elevations or opioidwithdrawal symptoms); hepatic failure, past/presenthistory of an AIDS-indicator disease, or active hepatitisand/or aspartate aminotransferase (AST) or alanineaminotransferase (ALT) >3¥ the upper limit of normal;known intolerance and/or hypersensitivity to naltrex-one, carboxymethylcellulose or polylactide-co-glycolide;psychosis, bipolar disorder, major depressive disorderwith suicidal ideation, current substance dependenceother than opioids or heroin, including alcohol; positiveurine test for cocaine/amphetamines; or naltrexone usewithin the last 6 months.

Study intervention

For the initial 6-month double-blind phase patients wererandomized to either XR-NTX 380 mg or PBO in a 1:1ratio, stratifying by site and gender. The study investigatoror a designated staff member injected XR-NTX within aweek of detoxification (!7 days following last opioiddose) and then every four weeks, for a total of six injec-tions. Patients who completed the initial 6-month studywere offered the open-label, 1-year extension study,which provided open-label XR-NTX 380 mg injectionsevery four weeks for up to 13 additional doses (total of 19injections over 18 months) at no expense to patients.

Throughout the 1.5-year study, participants wereoffered sessions of manualized Individual Drug Coun-seling (IDC), adapted for opioid dependence [19]. IDC-trained psychologists or psychiatrists reviewed patients’substance use, recovery efforts, functioning and adverseevents, providing support and advice. Sessions werebiweekly during the initial 6-month double-blind phase

Extended-release naltrexone 1629

© 2013 Society for the Study of Addiction Addiction, 108, 1628–1637

Page 14: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

and monthly during the 12-month open-label phase.Counseling sessions occurred when injections occurred,although not necessarily with the same clinician.

Patients were advised to not use the following medica-tions at any time during the 18-month protocol: oral nal-trexone, buprenorphine, levomethadyl acetate/LAAM,methadone, other prescription opioids, antipsychotics,anticonvulsants, antidepressants and anxiolytics. Per-mitted medications included anticonvulsants if dosingwas stable and short-acting PRN (as needed) insomniamedications, for example zopiclone.

Efficacy and safety assessments

Urine drug testing for opioids (immunochromatography-based one-step in vitro tests) was performed at scheduledvisits, weekly for 6 months during the double-blind phaseand monthly during the 1-year extension phase, detect-ing urine morphine and methadone concentrations at300 ng/mL. Urine results for weeks 1–4 were prospec-tively omitted because participants might challenge theblockade during this period. Self-report of drug use, usingthe Timeline Follow-back (TLFB) method [20], was usedto confirm negative urine results. The TLFB method usescalendars and daily recall of substance use on specificdays to record opioid quantity/frequency. If use of opioidsfor a given week was evident from the TLFB, the week wascoded as ‘not abstinent’. In addition, the Addiction Sever-ity Index (ASI) [21] was administered at baseline and themonthly visits during the open-label phase. From the ASI,days in the past 30 using individual types of drugs andalcohol were examined.

Also included to assess the durability of effects weremeasures of retention, opioid craving, functioning andglobal improvement. Craving was assessed weekly duringthe first 6 months and monthly during the 1-year exten-sion phase with a self-report Visual Analogue Scale of‘need for opioids’ (scale: 0–100, i.e. ‘not at all’ to ‘verymuch so’) [11]. Health functioning was measured withthe SF-36v2™ Health Survey [22] and the EQ-5D [23].The SF-36 and EQ-5D were obtained at baseline, endof the double-blind phase (month 6), and months 9, 12,16 and last visit (month 19, which occurred 1 monthafter the last injection at month 18). Global improve-ment was measured with the Clinical Global Impres-sion Improvement (CGI-I) scale [24]. ‘Responders’ weredefined a priori as having a CGI-I score of 1 (very much) or2 (much) improved. The CGI-I was obtained at baselineand months 6, 12 and 19.

Safety was assessed during the 1-year extension phasethrough monthly monitoring of treatment-emergentadverse events, vital signs, biochemistry and hematologyurine/blood tests (including liver function tests), andphysical examination of injection sites. Laboratory tests

were evaluated relative to established norms and changesfrom baseline. Determinations of severity and clinical sig-nificance were made by investigators at each site. Electro-cardiograms (ECGs) were obtained at baseline, month 6,month 12 and month 19.

Statistical analysis

Missing urine drug test results were imputed as positivefor opioids; retention was censored upon discontinuation;craving, SF-36, EQ-5D, ASI and CGI-I scores wereimputed using last post-dose observation carried forward.

Retention was examined through a Kaplan–Meiertime-to-discontinuation survival analysis, using thesample of patients who entered the open-label phase.Safety results are presented descriptively in terms of thenumber and percent of patients displaying any adverseevents or other safety concerns.

To allow descriptive comparisons with the results fromthe double-blind phase, we present here data for thosepatients (n = 114) who completed the double-blind phaseand then entered the open-label phase. Statistical analy-ses were performed using SAS (v. 9.1).

RESULTS

Patient characteristics and disposition

There were 335 individuals screened for the initial double-blind phase, and 250 of these (74.6%) were randomizedto XR-NTX or PBO (Fig. 1). Of these, 57.9% (73 of 126)XR-NTX patients versus 41.9% (52/124) PBO patientsreceived all six double-blind doses. Of the initial 250randomized patients, 53.2% (67/126) continued withXR-NTX into the 1-year open label phase versus 37.9%(47/124; P = 0.017) who were randomized to PBO, butwere switched to XR-NTX for the open label phase. Theprimary reasons for attrition during the 1-year open-labelphase were withdrawal of consent (18.4%; 21/114) andbecoming lost to follow-up (11.4%; 13/114).

In general, patients who continued into the 1-yearopen-label phase were similar to the subset that did notcomplete the preceding double-blind phase and did notenter the open-label extension phase (Table 1). Thesample was predominantly young, male, white, addictedto heroin for about 10 years, and had high rates of HIVand hepatitis C infection. In the sample entering the1-year continuation phase, 89.5% (102/114) were usingheroin at baseline (prior to entering the double-blindstudy), 8.8% (10/113) were using methadone and 9.8%(11/112) were using other opioids/analgesics.

Retention and durability of effects

Of the group that began the extension phase, 62.3% (71/114; 95% CI: 52.7%, 71.2%) completed the full 1-year of

1630 Evgeny Krupitsky et al.

© 2013 Society for the Study of Addiction Addiction, 108, 1628–1637

Page 15: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

treatment. This included 58.2% (39/67; 45.4%, 70.2%)of those continuing on XR-NTX and 68.1% (32/47;52.9%, 80.9%) of those who switched from PBO toXR-NTX. During the double-blind phase, significantlymore XR-NTX patients were retained. However, once thePBO patients switched to XR-NTX during the open-labelphase, their rate of attrition over time leveled off (Fig. 2).Of the original sample randomized to XR-NTX at theoutset of the double-blind study, 31% (39/126; 23.0%,39.8%) persisted with 18 months of treatment (24 weeksof double-blind plus 52-week extension).

Overall, 50.9% (58/114; 95% CI: 41.5%, 60.4%) ofpatients were abstinent from opioids at all scheduledmonthly assessments during the open-label phase withsimilar results in both groups: 49.3% of those continu-ing with XR-NTX and 53.2% of those who switchedfrom PBO. Of the 13 scheduled monthly urine drug tests,

an average of 76.7% (SD = 31.5) of tests were negativefor opioids (Fig. 3). Among open-label patients whoreceived XR-NTX or PBO during the double-blind phase,an average of 73.7% (SD = 33.2) and 81.0% (SD =28.6), respectively, of the tests were negative for opioids.Across the 1-year open-label phase, the percent ofopioid-free days was, on average, 83.4% (SD = 27.5). Forthose who received XR-NTX or PBO during the double-blind phase, there were an average of 80.6% (SD = 29.7)and 87.4% (SD = 23.8) opioid-free days. Three patients(of 47) who received PBO during the double-blind phasehad a positive urine test for opioids at the start of theopen-label phase.

Self-reported use of opioids, other drugs and alcohol isshown in Table 2. For all drugs, mean use in the past 30days at the end of the open-label phase remained ata similar low level, as was evident at the end of the

Figure 1 Flow diagram and subjectdispositionPBO = placebo; XR-NTX = extendedrelease naltrexone.

Table 1 Patient demographic and baseline clinical characteristics.

Characteristic

6-month double-blind phase 1-year open-label phase

XR-NTX 380 mg PBO XR-NTX→XR-NTX PBO→XR-NTX

n = 126 n = 124 n = 67 n = 47

Age, mean years (SD) 29.4 (# 4.8) 29.7 (# 3.6) 29.5 (# 5.0) 29.4 (# 3.8)Sex, n (%) male 113 (89.7%) 107 (86.3%) 62 (92.5%) 40 (85.1%)Race, n (%) white 124 (98.4%) 124 (100%) 67 (100%) 47 (100%)Duration of opioid dependence (years), mean (SD) 9.1 (# 4.5) 10.0 (# 3.9) 9.0 (# 4.2) 9.4 (# 4.0)Days of pre-study inpatient detoxification, mean (SD) 18 (# 9) 18 (# 7) 15.9 (# 8.2) 15.5 (# 6.8)Opioid Craving Scale, mean (SD) 18 (# 23) 22 (# 24) 20.7 (# 22.5) 18.6 (# 23.5)HIV serology, n (%) positive 51 (40.5%) 52 (41.9%) 31 (46.3%) 15 (31.9%)Hepatitis C, n (%) positive 111 (88.1%) 117 (94.4%) 58 (86.6%) 42 (89.3%)

PBO = placebo; XR-NTX = extended release naltrexone.

Extended-release naltrexone 1631

© 2013 Society for the Study of Addiction Addiction, 108, 1628–1637

Page 16: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

double-blind phase. Alcohol use was not highly elevatedat baseline (mean of approximately 5 days per month),and showed little change over the double-blind and open-label phases. For patients using any opioids at the end ofthe open-label phase, the median was 7.5 days of opioiduse in the past 30 days. As previously reported, in thedouble-blind phase XR-NTX patients had significantreductions in craving for opioids compared with PBO[16]. When PBO patients were switched to XR-NTX,craving for opioids was also reduced over time within this

group (Fig. 4). For those continuing on XR-NTX, meancraving for opioids remained low into and throughoutthe 1-year extension phase.

In the double-blind phase, the percentage of patientswho achieved responder status on the CGI-I, and meanchanges in the SF-36 mental components scores andEQ-5D were significantly greater for XR-NTX versus PBO[16]. In the XR-NTX group, 91.0% of those whocompleted (n = 67) the double-blind phase and began theopen-label phase were rated CGI-I responders; after the

* Ongoing patients, and those who completed the study, were censored at the day of the last visit.** Received XR-NTX during the open-label period

Figure 2 Time to discontinuation from extended release naltrexone (XR-NTX) for two cohorts: patients initially randomized to XR-NTXversus subsequently switched to XR-NTX from placebo*

Table 2 Mean (SD) values for Addiction Severity Index self-reported drug and alcohol days used in past 30 days over the course of thedouble-blind and open-label phases.

BaselineBeginning of open-label phase(6 months)

End of open-label phase(18 months)

PBO, XR-NTX, PBO, XR-NTX, PBO, XR-NTX,XR-NTX XR-NTX XR-NTX XR-NTX XR-NTX XR-NTX

OpioidsHeroin 18.4 (#11.8) 20.9 (#10.1) 0.3 (#1.5) 0.0 (#0.2) 0.1 (#0.5) 0.8 (#3.8)Methadone 1.2 (# 4.5) 0.2 (# 0.9) 0.0 (#0.2) 0.0 (#0.0) 0.0 (#0.0) 0.0 (#0.0)Other opiates 0.7 (# 3.0) 0.5 (# 1.4) 0.1 (#0.7) 0.0 (#0.3) 0.1 (#0.4) 0.0 (#0.0)

Alcohol 6.4 (# 8.1) 4.3 (# 5.9) 7.1 (#7.1) 5.9 (#6.5) 6.5 (#7.9) 5.5 (#6.6)Cocaine 0.3 (# 1.9) 0.0 (# 0.0) 0.0 (#0.3) 0.0 (#0.0) 0.0 (#0.0) 0.0 (#0.0)Cannabis 0.7 (# 2.4) 1.5 (# 4.7) 0.6 (#1.4) 0.5 (#1.7) 0.6 (#2.6) 0.3 (#1.0)Hypnotics/tranquilizers 1.3 (# 3.2) 1.6 (# 4.7) 0.2 (#0.8) 0.1 (#0.5) 0.0 (#0.0) 0.0 (#0.0)

PBO = placebo; PBO→XR-NTX = subgroup of patients who were randomized to PBO during the double-blind (initial 6-month) phase and then switchedto XR-NTX for the open-label phase; XR-NTX→XR-NTX = subgroup of patients who were randomized to XR-NTX during the double-blind phase andcontinued on XR-NTX for the open-label phase; XR-NTX = extended-release naltrexone.

1632 Evgeny Krupitsky et al.

© 2013 Society for the Study of Addiction Addiction, 108, 1628–1637

Page 17: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

switch, by the end of the 1-year open-label extensionphase, PBO→XR-NTX patients had a similar percent ofresponders, 89.4% (n = 47), while the XR-NTX→XR-NTX responder rate remained high (97.0%, n = 67).Changes in the SF-36 through the 1-year open-labelphase indicated that, for patients continuing on XR-NTX,overall patient health functioning gains evident over timefrom baseline to the end of the double-blind phase weremaintained over the course of the open-label phase.Mean # SD scores on the SF-36 Physical and MentalComponent scores, respectively, were 55.3 # 3.8 and50.6 # 9.2 for the XR-NTX group at end of the double-blind phase for those continuing into the open-labelphase (n = 67), and 56.3 # 4.2 and 50.2 # 8.9 withcontinuation on XR-NTX at the end of the 1-year openlabel phase (n = 62). On the SF-36 Mental Component

score, scores for PBO patients were stable: 49.4.1 # 8.7(end of double-blind) (n = 47) to 50.1 # 7.3 after switch-ing to XR-NTX (end of open-label) (n = 46). The SF-36Physical Component score for this group also remainedstable (54.4 # 6.2 to 56.6 # 4.0 from end of double-blind to end of open-label phases). EQ-5D scoresshowed continued improvement over the course of theopen-label phase in both groups [XR-NTX in bothphases: 81.6 # 12.4 (n = 67) to 83.8 # 12.7 (n = 67);PBO→XR-NTX: 77.9 # 18.10 (n = 47) to 82.7 # 15.1(n = 47)].

Safety

During the 1-year extension, overall, 21.1% (24/114) ofpatients reported an adverse event that was judged to bestudy drug related (Table 3). No specific type of adverse

Cohort: XR-NTX !XR-NTX

Cohort: PBO!XR-NTX

Figure 3 Urine opioid drug testing—complete results for individual patients by monthPBO = placebo; XR-NTX = extended release naltrexone.

Extended-release naltrexone 1633

© 2013 Society for the Study of Addiction Addiction, 108, 1628–1637

Page 18: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

event predominated. Injection site reactions were infre-quent (6.1%; 7/114) and the majority were mild (3 pain;2 extravasation; 1 induration; 1 swelling). One patientdiscontinued treatment during the 1-year extensionphase owing to a non-serious adverse event. This patient,who had ongoing hepatitis B and C infections, hadelevated liver enzymes at baseline (ALT 136 IU/L, AST87 IU/L, gamma-glutamyl transferase [GGT] 523 IU/L)and while receiving PBO (after three injections: ALT420 IU/L, AST 448 IU/L, GGT 1510 IU/L) during the6-month double-blind phase. These elevations continued

during the extension phase and the patient was discon-tinued (6 weeks after last dose of XR-NTX: ALT 553 IU/L,AST 615 IU/L, GGT 754 IU/L). Three patients experi-enced a total of four serious adverse events (SAEs) duringthe 1-year extension phase. No individual SAE wasreported by more than one patient. The SAEs were acutepancreatitis, cardiomyopathy, hepatitis A and pulmonarytuberculosis (the latter two occurring in the samepatient). The pancreatitis was judged as possibly relatedto XR-NTX and the cardiomyopathy was judged as prob-ably not related to XR-NTX. No deaths or overdoses

* Last Observation Carried Forward (Subjects censored at last visit).** Received XR-NTX during the open-label period (part B)

Figure 4 Mean changes in opioid craving over the course of double-blind and open-label phases for cohorts of patients that enteredopen-label treatment*XR-NTX = extended release naltrexone.

Table 3 Adverse events during 1-year open label treatment with extended release naltrexone (XR-NTX).

EventsOverall XR-NTXa,XR-NTX PBOb,XR-NTXn = 114 n = 67 n = 47

Any adverse event 48 (42.1%) 29 (43.3%) 19 (40.4%)Discontinued owing to non-serious adverse event 1 0 1 (2.1%)Toothache 7 (6.1%) 3 (4.5%) 4 (8.5%)Influenza 6 (5.3%) 4 (6.0%) 2 (4.3%)Bacteriuria 3 (2.6%) 2 (3.0%) 1 (2.1%)Injection site pain 3 (2.6%) 1 (1.5%) 2 (4.3%)Deaths 0 0 0Serious adverse events 3 (2.6%) 3 (4.5%) 0Study drug-related adverse eventsc 24 (21.1%) 14 (20.9%) 10 (21.3%)

PBO = placebo. aPatients who received XR-NTX in the 6-month double-blind phase and remained on XR-NTX for the 1-year open-label phase. bPatientswho received placebo in the 6-month double-blind phase and were switched to XR-NTX for the 1-year open-label phase. cOnly adverse events that werecoded by investigators as study drug-related are included here.

1634 Evgeny Krupitsky et al.

© 2013 Society for the Study of Addiction Addiction, 108, 1628–1637

Page 19: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

occurred during either the 6-month double-blind phaseor the 1-year extension phase.

During the open-label phase, 22 patients [7 (14.9%)who switched and 15 (22.4%) who were continuingon XR-NTX] had laboratory abnormalities. Of these,17 were considered to be related to XR-NTX. Specificincreases in liver enzymes were experienced by 13(19.4%) of patients who continued on XR-NTX, and 6(12.8%) of those who switched from PBO to XR-NTXduring the open-label phase [overall: 19 (16.7%)]. Alllaboratory abnormalities were judged mild or moderatein severity. None of the laboratory abnormalities wereviewed as clinically meaningful by the investigators’judgment.

There were no clinically significant abnormalitiesdetected through measurement of vital signs or throughphysical examinations. An abnormality of mild severitywas evident on an ECG recording for one patient (short-ened PR).

DISCUSSION

In this long-term study of patients who receivedXR-NTX during an open-label, 1-year extension phasefollowing 6 months of double-blind treatment withXR-NTX or PBO, XR-NTX patients maintained theirimprovements over time in regard to abstinence fromopioids, craving for opioids and overall health function-ing. Patients who switched from PBO treatmentduring the double-blind phase to 1 year of open-labelXR-NTX treatment were a select subpopulation of thoseinitially randomized to PBO (with only 3 of 47 ofthese patients testing positive for opioids at the begin-ning of the open-label phase). However, even this self-selected subgroup appeared to improve further incraving for opioids over time once these patients beganreceiving XR-NTX during the open-label extensionphase. About half of all patients who began the exten-sion phase were completely abstinent from opioidsacross the additional year of assessment. Opioid use wasrare during the follow up, and episodes of use, whichmay have represented testing the blockade, did notappear to result in dropout and relapse. Because of theclinical importance of retention and abstinence, opioid-negative urine was analyzed imputing missing urine aspositive—a conservative approach to describing thepattern of results. There was no evidence that patientsincreased their use of other drugs and alcohol afterdecreasing their use of opioids over the course of thedouble-blind and open-label phases.

No new safety concerns were observed for XR-NTXduring the open-label extension. Long-term treatmentwith XR-NTX showed a low rate of adverse events, theabsence of severe adverse events, and a low overall rate

(2.6%) of injection site pain, with no serious injection sitereactions. No patients discontinued the open-label exten-sion owing to serious adverse events. In this sample, inwhich 88% had chronic hepatitis C at baseline, elevationsin liver function tests occurred in about 10% of patients,and were not clinically meaningful. These results extendthe analyses of liver function tests conducted on the6-month double-blind phase in the treatment of opioiddependence [16], as well as a 6-month study of hepaticsafety for XR-NTX in the treatment of alcohol depend-ence [25], which concluded there was no evidencefor hepatotoxicity with XR-NTX taken in the approveddosage.

Retention rates over 18 months of XR-NTX treat-ment were encouraging. Of those initially randomizedto XR-NTX in the double-blind phase, 31% completed18 months of treatment, and of those who began the1-year extension phase, 62.2% completed it. Systematiclong-term studies of opioid dependence treatment arerare, and it is difficult to compare the retention ratesfound here to other studies because retention willvary depending on the design of the initial treatmentphase, length of treatment, setting, country wherestudy was conducted, and other study and patientcharacteristics.

Several limitations of this study should be noted.Long-term efficacy of XR-NTX with individual drugcounseling was based on open-label treatment, withoutrandomization. In the course of long-term studies,differential attrition may be expected. Because of thedouble-blind phase preceding this extension study,opioid-dependent patients who survived in treatmentwith PBO and counseling for 6 months and then soughtto enter the open-label extension study may haverepresented a subgroup with higher motivation, result-ing in more favorable outcomes once switched to activeXR-NTX during the open-label phase. A potential limita-tion is that this study was conducted in Russia. The gen-eralizability of these results to other countries that havedifferent systems for providing services to addicted indi-viduals is not known. Further research is needed toconfirm these findings in other settings. However, a largeretrospective analysis of US insurance claims across allapproved treatments reported favorable total health-carecost findings and rates of re-hospitalization in XR-NTX-treated patients [17]. An important limitation is thatpatients were not tracked after dropout from treatmentin either the acute trial [16] or the long-term extensionreported here. Dropout from treatment for opioiddependence and relapse is, unfortunately, a commonoutcome [9,14,15]. Risks after dropout include relapseand death from opioid overdose, and future researchon treatments for opioid dependence should trackdropouts to better understand relapse rates, how to

Extended-release naltrexone 1635

© 2013 Society for the Study of Addiction Addiction, 108, 1628–1637

Page 20: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

further reduce attrition (e.g. with behavioral interven-tions and comorbidity measures), safety and what pro-portion may, in fact, sustain abstinence even afterXR-NTX is discontinued.

In summary, improvements over time following a6-month double-blind phase were maintained during1 year of long-term treatment with XR-NTX and nonew safety concerns were evident.

Trial registration

Clinicaltrials.gov Identifier: NCT00678418.

Declaration of interests

This study was funded by Alkermes, Inc. The Medisorbpreparation used in XR-NTX was developed with supportfrom the National Institute on Drug Abuse (grantR43DA013531) and the National Institute on AlcoholAbuse and Alcoholism (grant N43AA001002).

Evgeny Krupitsky, MD, PhD, is a consultant forAlkermes and received research funding for this studyfrom Alkermes.

Edward V. Nunes, MD, was a member of the AdvisoryBoard to Alkermes that designed this trial and served asan unpaid consultant to an expert panel convened byAlkermes.

Walter Ling, MD, has been an Advisory Board memberfor Alkermes and US World Med; has received researchfunding from Titan Pharmaceuticals and investigator-initiated research funding from Hythiam; and researchsupport, an unrestricted educational grant and speakersupport from Reckitt Benckiser.

David R. Gastfriend, MD, Asli Memisoglu, ScD, andBernard L. Silverman, MD, are employees of Alkermes,Inc.

Acknowledgement

We would like to acknowledge the staff at Alkermes, PSIand Cytel for their work in the implementation andbiostatistical analysis of this study, especially ElliotEhrich, Doreen Campbell, Handong Zhang, AnnaO’Rourke and Stanislav Nikitin. We thank Dr EdwardSchweizer of Paladin Consulting Group, Inc. (Princeton,NJ, USA), who provided medical writing assistance withthe manuscript, and was compensated by Alkermes, Inc.,for his assistance.

References

1. Substance Abuse and Mental Health Services Administra-tion. Results from the 2009 National Survey on Drug Use andHealth: Volume I. Summary of National Findings (Office ofApplied Studies, NSDUH Series H-38A, HHS Publication No.

SMA 10-4586 Findings). Rockville, MD: Substance Abuse &Mental Health Services Administration–the US Departmentof Health and Human Services; 2010.

2. UNODC. World Drug Report 2010 (United Nations Publica-tion, Sales No. E.10.XI.13). 2010. Available at: http://www.unodc.org/documents/wdr/WDR_2010/World_Drug_Report_2010_lo-res.pdf (accessed 22 August 2012).(Archived by WebCite® at http://www.webcitation.org/60NzBlRJC).

3. Hser Y., Anglin M. D., Powers K. A 24-year follow-up ofCalifornia narcotics addicts. Arch Gen Psychiatry 1993; 50:577–84.

4. Wilcox H. C., Conner K. R., Caine E. D. Association ofalcohol and drug use disorders and completed suicide: anempirical review of cohort studies. Drug Alcohol Depend2004; 76S: S11–S19.

5. National Institute of Drug Abuse. NIDA Research Report-Heroin Abuse and Addiction. NIH Publication No. 05–4165,2005.

6. Leshner A. I. Drug addiction research: moving toward the21st century. Drug Alcohol Depend 1998; 51: 5–7.

7. McLellan A. T., McKay J. R. Components of successful addic-tion treatment. In: Graham A. W., Schultz T. K., Mayo-SmithM. F., Ries R. K., Wilford B. B., editors. Principles of AddictionMedicine, 3rd edn. Chevy Chase: American Society ofAddiction Medicine; 2003, p. 429–42.

8. Mattick R. P., Kimber J., Breen C., Davoli M. Buprenorphinemaintenance versus placebo or methadone maintenancefor opioid dependence. Cochrane Database Syst Rev 2008; 2:CD002207.

9. Minozzi S., Amato L., Vecchi S., Davoli M., Kirchmayer U.,Verster A. Oral naltrexone maintenance treatment foropioid dependence. Cochrane Database Syst Rev 2011; 4:CD001333.

10. Mattick R. P., Breen C., Kimber J., Davoli M. Methadonemaintenance therapy versus no opioid replacement therapyfor opioid dependence. Cochrane Database Syst Rev 2009; 3:CD002209.

11. Krupitsky E. M., Zvartau E. E., Masalov D. V., Tsoi M. V.,Burakov A. M., Egorova V. Y. et al. Naltrexone for heroindependence treatment in St. Petersburg, Russia. J SubstAbuse Treat 2004; 26: 285–94.

12. Fudala P. J., Bridge T. P., Herbert S., Williford W. O., Chiang C.N., Jones K. et al. Office-based treatment of opiate addictionwith a sublingual-tablet formulation of buprenorphine andnaloxone. N Engl J Med 2003; 349: 949–58.

13. Newman R., Whitehill W. Double-blind comparisonof methadone and placebo maintenance treatments ofnarcotic addicts in Hong Kong. Lancet 1979; 8: 485–8.

14. Dekimpe M. G., Van De Gucht L. M. V., Hanssens D. M.,Powers K. I. Long run abstinence after narcoticsabuse: what are the odds? Manage Sci 1998; 44: 1478–92.

15. Sullivan M., Garawi F., Bisaga A., Comer S. D., Carpenter K.,Raby W. N. et al. Management of relapse in naltrexonemaintenance for heroin dependence. Drug Alcohol Depend2007; 91: 289–92.

16. Krupitsky E., Nunes E. V., Ling W., Illeperuma A., GastfriendD. R., Silverman B. L. Injectable extended-release naltrexonefor opioid dependence: a double-blind, placebo-controlled,multicentre randomised trial. Lancet 2011; 377: 1506–13.

17. Garbutt J. C., Kranzler H. R., O’Malley S. S., Gastfriend D. R.,Pettinati H. M., Silverman B. L. et al. Efficacy and tolerability

1636 Evgeny Krupitsky et al.

© 2013 Society for the Study of Addiction Addiction, 108, 1628–1637

Page 21: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

of long-acting injectable naltrexone for alcohol dependence:a randomized controlled trial. JAMA 2005; 293: 1617–25.

18. American Psychiatric Association. Diagnostic and StatisticalManual of Mental Disorders, 4th edn. text revision (DSM-IV-TR). Washington, DC: American Psychiatric Association;2000.

19. Mercer D., Woody G. E. Individual Drug Counseling. NIH Pub-lication Number 99-4380; Rockville, MD: National Instituteon Drug Abuse; 1999.

20. Sobell L. C., Sobell M. B. Timeline follow-back: a techniquefor assessing self-reported alcohol consumption. In: LittenR. Z., Allen J. P., editors. Measuring Alcohol Consumption:Psychosocial and Biological Methods, Totowa, NJ: TheHumana Press; 1992, p. 41–72.

21. McLellan A. T., Kushner H., Metzger D., Peters R., Smith I.et al. The fifth edition of the addiction severity index. J SubstAbuse Treat 1992; 9: 199–213.

22. Ware J. E., Kosinski M., Dewey J. E. How to Score VersionTwo of the SF-36 Health Survey. Lincoln RI: QualityMetricIncorporated; 2000.

23. Euroqol Group. Euroqol—A new facility for the measure-ment of health-related quality of life. Health Policy (NewYork) 1990; 16: 199–208.

24. Guy W. ECDEU Assessment for Psychopharmacology,Revised Edition. Rockville, MD: NIMH Publication; 1976,p. 76–338.

25. Lucey M. R., Silverman B. L., Illeperuma A., O’Brien C. P.Hepatic safety of once-monthly injectable extended-releasenaltrexone administered to actively drinking alcoholics.Alcohol Clin Exp Res 2008; 32: 498–504.

26. Baser O., Chalk M., Fiellin D. A., Gastfriend D. R. Cost andutilization outcomes of opioid-dependence treatments. Am JManag Care 2011; 17: S235–48.

Extended-release naltrexone 1637

© 2013 Society for the Study of Addiction Addiction, 108, 1628–1637

Page 22: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

This document is a scanned copy of a printed document. No warranty is given about theaccuracy of the copy. Users should refer to the original published version of the material.

Page 23: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

Articles

1506 www.thelancet.com Vol 377 April 30, 2011

Lancet 2011; 377: 1506–13

Published OnlineApril 28, 2011

DOI:10.1016/S0140-6736(11)60358-9

See Comment page 1468

Bekhterev Research Psychoneurological Institute,

St Petersburg State Pavlov Medical University,

St Petersburg, Russia (Prof E Krupitsky MD); New York State Psychiatric Institute and

Department of Psychiatry, Columbia University, New York,

NY, USA (Prof E V Nunes MD); Department of Psychiatry and

Biobehavioral Sciences, University of California

Los Angeles, Los Angeles, CA, USA (Prof W Ling MD); and

Alkermes, Waltham, MA, USA (A Illeperuma MA,

D R Gastfriend MD, B L Silverman MD)

Correspondence to:Prof Evgeny Krupitsky,

Department of Addictions, St Petersburg Bekhterev

Psychoneurological Research Institute, Bekhtereva Street 3, St Petersburg 192019, Russia

[email protected]

Injectable extended-release naltrexone for opioid dependence: a double-blind, placebo-controlled, multicentre randomised trial Evgeny Krupitsky, Edward V Nunes, Walter Ling, Ari Illeperuma, David R Gastfriend, Bernard L Silverman

SummaryBackground Opioid dependence is associated with low rates of treatment-seeking, poor adherence to treatment, frequent relapse, and major societal consequences. We aimed to assess the effi cacy, safety, and patient-reported outcomes of an injectable, once monthly extended-release formulation of the opioid antagonist naltrexone (XR-NTX) for treatment of patients with opioid dependence after detoxifi cation.

Methods We did a double-blind, placebo-controlled, randomised, 24-week trial of patients with opioid dependence disorder. Patients aged 18 years or over who had 30 days or less of inpatient detoxifi cation and 7 days or more off all opioids were enrolled at 13 clinical sites in Russia. We randomly assigned patients (1:1) to either 380 mg XR-NTX or placebo by an interactive voice response system, stratifi ed by site and gender in a centralised, permuted-block method. Participants also received 12 biweekly counselling sessions. Participants, investigators, staff , and the sponsor were masked to treatment allocation. The primary endpoint was the response profi le for confi rmed abstinence during weeks 5–24, assessed by urine drug tests and self report of non-use. Secondary endpoints were self-reported opioid-free days, opioid craving scores, number of days of retention, and relapse to physiological opioid dependence. Analyses were by intention to treat. This trial is registered at ClinicalTrials.gov, NCT00678418.

Findings Between July 3, 2008, and Oct 5, 2009, 250 patients were randomly assigned to XR-NTX (n=126) or placebo (n=124). The median proportion of weeks of confi rmed abstinence was 90·0% (95% CI 69·9–92·4) in the XR-NTX group compared with 35·0% (11·4–63·8) in the placebo group (p=0·0002). Patients in the XR-NTX group self-reported a median of 99·2% (range 89·1–99·4) opioid-free days compared with 60·4% (46·2–94·0) for the placebo group (p=0·0004). The mean change in craving was –10·1 (95% CI –12·3 to –7·8) in the XR-NTX group compared with 0·7 (–3·1 to 4·4) in the placebo group (p<0·0001). Median retention was over 168 days in the XR-NTX group compared with 96 days (95% CI 63–165) in the placebo group (p=0·0042). Naloxone challenge confi rmed relapse to physiological opioid dependence in 17 patients in the placebo group compared with one in the XR-NTX group (p<0·0001). XR-NTX was well tolerated. Two patients in each group discontinued owing to adverse events. No XR-NTX-treated patients died, overdosed, or discontinued owing to severe adverse events.

Interpretation XR-NTX represents a new treatment option that is distinct from opioid agonist maintenance treatment. XR-NTX in conjunction with psychosocial treatment might improve acceptance of opioid dependence pharmacotherapy and provide a useful treatment option for many patients.

Funding Alkermes.

IntroductionOpioid dependence is a potentially life-threatening illness1 associated with adverse societal eff ects including increased morbidity and mortality, poor social functioning, economic dependence, and crime.2–4 The worldwide incidence of opioid dependence has increased during the past decade, and many patients are not receiving treatment for the disorder, although rates of treatment are increasing in many countries.1,5,6 The main treatments consist of either maintenance pharmaco-therapy with counselling or drug-free psychosocial treatment. Although abstinence is the primary goal, drug-free treatment is associated with high rates of relapse.7 Agonist maintenance, such as with the μ-opioid receptor agonist methadone or the partial agonist buprenorphine, has an established role in the management of opioid

dependence, with studies, reviews, and meta-analyses reporting a variety of public-health and safety benefi ts. These benefi ts include decreases in illicit drug use; reduced rates of HIV seroconversion, and improved morbidity, mortality, HIV risk behaviours, and patient functioning.5,7–10 However, in 122 of 192 UN member states, agonist therapy is restricted or unavailable because of philosophical preferences for opioid-free treatment or policy concerns about physiological dependence or abuse and illegal drug diversion.5,6 Furthermore, agonist therapy might be less suitable for certain subgroups of patients, particularly young people, patients with a brief history of addiction or who are new to treatment, and patients whose employment might prohibit opioid use (eg, health-care providers, pilots, and police, fi re, emergency and military personnel).

Downloaded from ClinicalKey.com at University at Buffalo The State University of New York March 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 24: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

Articles

www.thelancet.com Vol 377 April 30, 2011 1507

An alternative pharmacotherapy that supports abstinence is naltrexone, a µ-opioid receptor antagonist that does not have opioid agonist eff ects, produces no euphoria or sedation, and is not addictive. Antagonist pharmacotherapy is particularly appropriate for patients who have achieved abstinence during inpatient treatment or incarceration and are at risk of relapse after discharge. Naltrexone cessation causes no symptoms of withdrawal because patients are not physically opioid dependent. However, apart from when dosing is supervised, such as for recovering physicians11 or in the context of intensive behavioural treatments,12 oral naltrexone has generally been ineff ective because of poor adherence.13

In 1976, the US National Institute on Drug Abuse requested development of a long-acting opioid antagonist. Responses to this request consisted of subcutaneous naltrexone implants, which have shown effi cacy14,15 but are associated with adverse events related to surgical insertion; and a long-acting injectable naltrexone formulation, which was eff ective in a small, 2-month long controlled trial.16 A once-monthly extended-release formulation of injectable naltrexone (XR-NTX, Vivitrol, Alkermes, Waltham MA, USA) has been approved in the USA and Russia for treatment of alcohol dependence. This formulation, administered via intramuscular injection by a health-care provider, gradually releases naltrexone from microspheres composed of medical-grade poly-(d,l-lactide-co-glycolide)—a polymer used in dissolvable surgical sutures. In patients with alcohol dependence, XR-NTX reduced the incidence of heavy drinking17 and increased the rate of total abstinence over 6 months in those with initial abstinence compared with placebo,18 with associated improvements in health and social functioning.19

We did a multicentre, randomised, placebo-controlled 24-week trial to assess the effi cacy, safety, and patient-reported outcomes of once-monthly XR-NTX for the treatment of opioid dependence.

MethodsPatientsMen and women aged 18 years or over who met the Diagnostic and Statistical Manual of Mental Disorders 4th edition20 criteria for opioid dependence disorder, who were completing inpatient opioid detoxifi cation (≤30 days), and who were off opioids for at least 7 days were enrolled at 13 clinical sites in Russia. Patients were voluntarily seeking treatment and were excluded if they were under justice system coercion—ie, parole or probation, or pending legal proceedings with potential for incarceration. Every patient also had a signifi cant other (eg, spouse or relative) who supervised their compliance with the visit schedule and study procedures. Women of childbearing potential agreed to use contraception during the study.

Exclusion criteria were pregnancy or breastfeeding; signifi cant medical conditions (eg, acute renal failure,

endocarditis, and tuberculosis); positive naloxone challenge (increases in vital signs or opioid withdrawal symptoms); hepatic failure; past or present history of an AIDS-indicator disease; active hepatitis or aspartate amino transferase or alanine aminotransferase more than three times the upper limit of normal; known intolerance or hypersensitivity to naltrexone, carmellose, or polylactide-co-glycolide; psychosis, bipolar disorder, major depressive disorder with suicidal ideation, or present dependence on substances other than opioids or heroin, including alcohol; positive urine test for cocaine or amphetamines; and naltrexone use within the past 6 months.

Each site’s independent ethics committee or institu-tional review board approved the protocol and participants gave written, informed consent in accordance with the Declaration of Helsinki.

Randomisation and maskingWe randomly assigned patients (1:1) to either 380 mg XR-NTX or placebo by an interactive voice response system, stratifi ed by site and sex with a centralised, permuted-block method with a block size of four. This system was also used to manage the supply of masked study drugs. Participants, investigators, staff , and the sponsor were masked to treatment allocation. To ensure masking, amber vials and syringes were used, and diff erent personnel did counselling and data collection.

ProceduresPatients received an injection of XR-NTX or placebo within 1 week after detoxifi cation and then every 4 weeks thereafter, for a total of six injections over 24 weeks. Participants were also off ered 12 biweekly sessions of individual drug counselling, adapted for opioid dependence.21 Psychologists or psychiatrists who were trained in individual drug counselling reviewed patients’ substance use, recovery eff orts, functioning, and adverse events, and provided support and advice to patients. Upon completion of the 24-week treatment period, all patients were off ered open-label XR-NTX treatment for an additional year. All treatment was off ered at no expense to patients. Urine drug testing for opioids (immunochromatography-based one-step in-vitro tests) was done weekly for 24 weeks and detected urine morphine and methadone at concentrations greater than 300 ng/mL.

The following drugs were prohibited during the study: naltrexone, buprenorphine, levacetylmethadol, metha done, other prescription opioids, antipsychotics, anticonvulsants, antidepressants, and anxiolytics. Permit-ted drugs were anticonvulsants if dosing was stable and short-acting insomnia drugs, such as zopiclone, as required.

The primary endpoint was the response profi le for confi rmed abstinence during weeks 5–24. We prospectively omitted weeks 1–4 from this endpoint because participants might challenge the blockade during this period, after which abstinence should stabilise.

Downloaded from ClinicalKey.com at University at Buffalo The State University of New York March 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 25: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

Articles

1508 www.thelancet.com Vol 377 April 30, 2011

Confi rmed abstinence was defi ned as a negative urine drug test and no self-reported opioid use on the timeline follow-back (TLFB) survey.22 The TLFB survey uses calendars and daily recall of substance use on specifi c days to record quantity or frequency of opioid use. Omission of any of these criteria resulted in failure to confi rm abstinence for the week.

Secondary a-priori endpoints were self-reported opioid-free days according to the TLFB, opioid craving scores, number of days of retention, and relapse to physiological opioid dependence. Because use of opioids might produce relapse to physiological opioid dependence, measurement of both opioid use and physiological dependence was important. Craving was assessed with a weekly self-report visual analogue scale (VAS) of need for opioids (scale 0–100, 0=not at all; 100=very much so).23 Physiological dependence was assessed via naloxone challenge at baseline, upon any positive urine drug screen, at treatment discontinuation, and at week 24. Patients were removed from the study if the naloxone challenge test was positive, to protect the patient from the possibility of a prolonged precipitated withdrawal with XR-NTX. Other health outcomes that were also assessed included the HIV risk assessment battery,24 the 36-item short form health survey (version 2),25 patients’ VAS assessments of their general health on the EuroQol-5 dimensions questionnaire,26 and investigators’ revised clinical global impression ratings.27

Safety was assessed by weekly monitoring of treatment-emergent adverse events, vital signs, biochemistry and haematology on urine and blood samples, including liver function tests, monthly physical examination of injection sites, and baseline and endpoint electrocardiographs.

Statistical analysisBefore the trial, we calculated that a sample size of 125 patients per treatment group would provide 85% and 96% power to detect an eff ect size of Cohen’s d 0·4 and 0·5, respectively, by a Wilcoxon rank-sum test at a two-sided signifi cance level of 0·05. Intent-to-treat analyses of effi cacy endpoints were done with all randomised patients. We created response profi les by calculating the number of confi rmed abstinence weeks for weeks 5–24 for each patient and then dividing by the number of scheduled tests (20). The response profi le for each treatment group is the cumulative distribution function of percent of opioid-free weeks. For between-group comparisons we used a two-sided Van der Waerden test28—a non-parametric test of whether k population distributions are equal. To assess the eff ect of baseline characteristics, the rate of opioid-negative urine drug tests were analysed with ANCOVA, containing factors for treatment group, sex, and sex-by-treatment interaction, and with age, duration of opioid dependence, and duration of last pre-study inpatient detoxifi cation as covariates. Consistency of the eff ects of treatment on opioid-free weeks across subgroups

335 screened for eligibility 85 excluded31 did not meet inclusion

criteria8 lost to follow-up

36 withdrew consent10 other

250 randomised

124 assigned to and received placebo126 assigned to and received XR-NTX

47 completed trial67 completed trial

124 included in primary analysis126 included in primary analysis

77 did not complete trial6 lost to follow-up

34 lack of efficacy12 non-compliance

2 adverse events23 other reasons

59 did not complete trial6 lost to follow-up

22 lack of efficacy17 withdrew consent 2 adverse events12 other reasons

Figure 1: Trial profi leXR-NTX=extended-release naltrexone.

XR-NTX (n=126)

Placebo (n=124)

Age (years) 29·4 (4·8) 29·7 (3·6)

Men 113 (90%) 107 (86%)

White 124 (98%) 124 (100%)

Duration of opioid dependence (years) 9·1 (4·5) 10·0 (3·9)

Days of pre-study inpatient detoxifi cation 18 (9) 18 (7)

Opioid craving scale 18 (23) 22 (24)

HIV serology positive 51 (40%) 52 (42%)

Hepatitis C positive 111 (88%) 117 (94%)

Data are mean (SD) or number (%). XR-NTX=extended-release naltrexone.

Table 1: Demographics and baseline clinical characteristics

0 10 20 30 40 50 60 70 80 90 1000

10

20

30

40

50

60

70

80

90

100

Opio

id-fr

ee w

eeks

(%)

Participants (%)

XR-NTX (n=126)Placebo (n=124)

Figure 2: Percent of confi rmed opioid-free weeks (cumulative) among participants treated with XR-NTX compared with placeboXR-NTX=extended-release naltrexone.

Downloaded from ClinicalKey.com at University at Buffalo The State University of New York March 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 26: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

Articles

www.thelancet.com Vol 377 April 30, 2011 1509

defi ned by baseline characteristics (sex, age, duration of opioid dependence, and duration of pre-study detoxifi cation) and site was measured with ANCOVA models. Retention was assessed with Kaplan-Meier curves and a log-rank test. Changes from baseline in weekly craving scores were analysed with a generalised estimation equation model, assuming normal distri-bution and autoregressive correlation structure, with baseline craving as a covariate. For secondary endpoints, group diff erences were tested with the Van de Waerden test for continuous endpoints and χ² tests or Fisher’s exact test for categorical endpoints. Adverse events were compared by Fisher’s exact test.

Missing urine drug test results were imputed as positive for opioids; retention was censored upon discontinuation, craving was imputed using last observation carried forward, and missing TLFB data were imputed using patients’ rates of opioid-free days during the 30 pre-detoxifi cation days. For all other endpoints, all available data were included in analyses.

The primary endpoint was tested with a two-sided α=0·05. For craving and retention outcomes p values were adjusted for multiplicity using the Bonferroni-Holm method29 to preserve family-wise type 1 error at 0·05.

A full statistical analysis was also done by an independent academic statistician who came to the same conclusions.

Role of the funding sourceThe sponsor designed the protocol in collaboration with participating investigators. The sponsor had the overall responsibility for the conduct of the study. Data were collected and monitored by Alkermes and PSI (Zug, Switzerland), a contract research organisation. Data were managed and analysed by Alkermes clinical and regulatory personnel, and staff at Cytel (Cambridge, MA, USA), and were interpreted by the authors with input

from Alkermes clinical and statistical staff . The fi rst author had full access to all study data and had fi nal responsibility for the decision to submit for publication.

ResultsBetween July 3, 2008, and Oct 5, 2009, 335 candidates were screened, 250 of whom were randomly assigned to XR-NTX or placebo (fi gure 1). Participants were predominantly young, white men (table 1) who had been addicted to heroin for about 10 years. High rates of HIV and hepatitis C infection were reported in the study population (table 1). In the 30 days before the fi rst injection, heroin was used by 221 (88%) of 250 participants, methadone by 29 (12%), and other opioids or analgesics by 33 (13%). Demographic and baseline clinical charac teristics showed no substantial inter-group diff erences (table 1).

Of 4285 urine drug tests and TLFB responses obtained, 4178 (97·5%) were in agreement. On 53 (1·2%) of 4285 occasions, participants self-reported using opioids despite opioid-negative urine tests. During weeks 5–24, there were 2098 of 5000 (42·0%) missing urine samples, 1255 (50·6%) of 2480 with placebo and 833 (33·1%) of 2520 with XR-NTX; 2096 of 2098 missing samples were because of early termination. Patients in the XR-NTX group received 1191 (99·7%) of 1194 scheduled counselling sessions (median 12; range 1–13) versus 922 (99·6%) of 926 for the placebo group (median 8; range 1–13).

The percentage of opioid-free weeks was signifi cantly higher in the XR-NTX group than the placebo group (p=0·0002), with substantial separation between groups across all measured values of opioid-free weeks (fi gure 2). The median proportion of patients who had confi rmed abstinence was higher in the XR-NTX group than the placebo group (p=0·0002; table 2). Total

XR-NTX (n=126) Placebo (n=124) Treatment eff ect* p value

Primary endpoint

Proportion of weeks of confi rmed abstinence 90·0% (69·9 to 92·4) 35·0% (11·4 to 63·8) 55·0 (15·9 to 76·1) 0·0002

Patients with total confi rmed abstinence 45 (35·7%, 27·4 to 44·1) 28 (22·6%, 15·2 to 29·9) 1·58 (1·06 to 2·36) 0·0224

Secondary endpoint

Proportion of self-reported opioid-free days over 24 weeks 99·2% (89·1 to 99·4) 60·4% (46·2 to 94·0) 38·7 (3·3 to 52·5) 0·0004

Craving: mean change in VAS score from baseline –10·1 (–12·3 to –7·8) 0·7 (–3·1 to 4·4) –10·7 (–15·0 to 6·4) <0·0001†

Number of days of retention >168‡ 96 (63 to 165) 0·61 (0·44 to 0·86) 0·0042†

Participants with positive naloxone challenge test 1 (0·8%, 0·0 to 2·3) 17 (13·7%, 7·7 to 19·8) 17·3 (2·3 to 127·8) <0·0001

Other outcomes

Patients who completed double-blind treatment period 67 (53·2%, 44·5 to 61·9) 47 (37·9%, 29·4 to 46·4) 1·40 (1·06 to 1·85) 0·0171

Risk for HIV: mean change in behaviour scores from baseline –0·187 (–0·224 to –0·150) –0·130 (–0·173 to –0·087) –0·057 (–0·113 to –0·001) 0·0212

Mean change from baseline in VAS self-ratings on EQ-5D 14·1 (9·6 to 18·7) 2·7 (–1·9 to 7·8) 11·4 (5·0 to 17·8) 0·0005

Proportion rated as much or very much improved on CGI 85·9% (77·8 to 94·0) 57·5% (45·7 to 69·5) 1·49 (1·19 to 1·87) 0·0002

Data are median (95% CI) or number (%, 95% CI), unless otherwise stated. XR-NTX=extended release naltrexone. VAS=visual analogue scale. EQ-5D=EuroQol-5 dimensions questionnaire. CGI=clinical global impression. *Diff erence between XR-NTX and placebo for location parameters and relative risk for proportions. Hazard ratio of early termination (Cox model) is shown for retention. †Adjusted for multiplicity by the Bonferroni-Holm method29 to preserve family-wise type 1 error at 0·05. ‡95% CI cannot be calculated because median exceeds the study duration.

Table 2: Clinical outcomes

Downloaded from ClinicalKey.com at University at Buffalo The State University of New York March 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 27: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

Articles

1510 www.thelancet.com Vol 377 April 30, 2011

abstinence was reported in 36% of patients in the XR-NTX group compared with 23% in the placebo group (p=0·0224; table 2). When effi cacy was analysed on the basis of the full 24-week period, including weeks 1–4, results were still signifi cant (p=0·0001). 119 (94%) of 126 patients in the XR-NTX group were opioid free compared with 96 (77%) of 124 in the placebo group by week 2, and this separation persisted through to the end of the trial (fi gure 3). No signifi cant relation was noted between age, sex, or duration of opioid dependence and the rate of opioid-free urine tests (data not shown). The treatment eff ect was consistent across baseline variables and study sites (data not shown).

All four secondary endpoints also showed signifi cant diff erences between the treatment groups (table 2). Median self-report of opioid-free days over 24 weeks was 99% for the XR-NTX compared with 60% for the placebo group (p=0·0004; table 2; fi gure 3). There was a statistically and clinically signifi cantly greater reduction in opioid craving in the XR-NTX group than the placebo group by week 8 (p=0·0048), which persisted every week through to week 24 (baseline to week 24: XR-NTX 18·2–8·8 vs placebo 21·8–22·5; p<0·0001, adjusted for multiplicity; table 2; fi gure 3). Median number of days of retention was 168 days (ie, still retained at the end of the study) in the XR-NTX group compared with 96 days for the placebo group (p=0·0042, adjusted for multiplicity; table 2; fi gure 3). All six injections were received by 73 (57·9%) of patients in the XR-NTX group compared with 52 (41·9%) of the placebo group (XR-NTX:placebo ratio 1·37, 95% CI 1·06–1·78; p=0·0171). Relapse to physiogical opioid dependence was identifi ed in one patient (who had missed two previous injections) in the XR-NTX group compared with 17 on placebo (p<0·0001; table 2).

Health outcome measures were similar between groups at baseline; however, the XR-NTX group had signifi cantly greater improvement from baseline than placebo in reduction of HIV risk, increased general health, and investigators’ clinical global impression improvement ratings. Baseline and post-treatment 36-item short form physical component summary scores were normal for both groups. The mental component score was well below US population norms (ie, score of 50) for both groups at baseline, but at study end the XR-NTX group (but not the placebo group) had normalised and was signifi cantly better than placebo by 0·5 SD (mean 50·37 [SD 9·18] vs 45·28 [10·47]; diff erence 5·09, 95% CI 2·09–8·09; p=0·0043). Similar results were found on all four subscales, including vitality (58·13 [8·43]) and were similar to Russian normative population scores.30

XR-NTX was generally well tolerated; two patients in each group discontinued owing to adverse events (table 3). 103 (41%) of 250 patients experienced at least one adverse event; a higher proportion of patients in the XR-NTX group than the placebo group had at least one

0 4 8 12 16 20 24

0 4 8 12 16 20 24

–12

–10

–8

–6

–4

–2

0

2

4

6

Mea

n ch

ange

in cr

avin

g sc

ore

Number at riskXR-NTXPlacebo

0 30 60 90 120 150 180

11082

9975

9164

8458

7553

2320

Treatment days

0

10

20

30

40

50

60

70

80

90

100

0

10

20

30

40

50

60

70

80

90

100Pa

tient

s with

full r

eten

tion

(%)

Log-rank p=0·0042 (adjusted)

XR-NTX (n=126)Placebo (n=124)

C

B

p<0·0001 (adjusted)

Opio

id-fr

ee p

atie

nts (

%)

Treatment week

XR-NTX (n=126)Placebo (n=124)

A

Figure 3: Key secondary effi cacy outcomes(A) Proportion of opioid-free patients by timeline follow-back self-report. (B) Mean change from baseline in craving. p value is based on a generalised estimating equation model assuming normal distribution and autoregressive correlation structure. (C) Time-to-discontinuation of study treatment. p values for analyses of craving (B) and retention (C) are adjusted for multiplicity. XR-NTX=extended-release naltrexone.

Downloaded from ClinicalKey.com at University at Buffalo The State University of New York March 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 28: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

Articles

www.thelancet.com Vol 377 April 30, 2011 1511

adverse event (p=0·005). All non-serious adverse events were deemed mild or moderate by investigators and most were judged to be unrelated to the study drug. Serious adverse events were uncommon and no episodes of intractable pain management were reported. No overdose events, suicide attempts, or deaths, or other severe adverse events were reported.

The mean increase from baseline of alanine aminotransferase was 6·9 IU/L in the XR-NTX group and 5·6 IU/L in the placebo group, and for aspartate aminotransferase the mean increase from baseline was 3·8 IU/L in the XR-NTX group and 6·7 IU/L for placebo. Hepatic enzyme abnormalities were more common with XR-NTX (data not shown).

DiscussionDetoxifi ed, opioid-dependent adults voluntarily seeking treatment who received XR-NTX had more opioid-free weeks than those who received placebo. Effi cacy did not vary by age, sex, or duration of opioid dependence. There was a persistent anti-craving eff ect over weeks 8–24, 94% fewer naloxone-confi rmed relapses to dependence, and nearly double the median length of retention in treatment in patients who received XR-NTX than those on placebo. Onset was rapid, with an anti-craving eff ect at week 1, an increase in abstinent days within 2 weeks, and improved retention at 1 month.

Although this study did not include a comparison with oral naltrexone, a meta-analysis of ten studies of oral naltrexone compared with placebo in multiple countries with 696 participants in total and a mean duration of 6 months did not fi nd benefi ts for retention or prevention of relapse (panel).13 Similarly, a study of oral naltrexone compared with treatment without naltrexone did not report an anti-craving eff ect,31 whereas in the present study

treatment with XR-NTX resulted in a rapid progressive decline in craving to 50% of baseline compared with no change with placebo. These diff erences might have been because oral naltrexone was self-administered daily and because XR-NTX has diff erent release kinetics, which, compared with daily oral naltrexone, yields about four times the area-under-the-curve plasma concentration of naltrexone and reduced exposure to 6β-naltrexol.32 Comparison of the present results with a small study of an injectable formulation of naltrexone are diffi cult because the previous study was only 8 weeks long, used a diff erent psychosocial intervention, and was done in the USA.16 However, both studies reported that extended-release, injectable naltrexone was superior to placebo for the outcome of opioid-negative urine.

XR-NTX was generally well tolerated and no new safety fi ndings were reported. Adverse events of any kind were reported by half of patients in the XR-NTX group compared with a third of those in the placebo group; however, rates of discontinuations owing to adverse events and serious adverse events were similar in both groups. High baseline incidence of opioid dependence-related medical comorbidity, including hepatitis C and HIV infection, might have aff ected liver enzyme measurements. Abnormal liver function tests occurred only in patients with existing hepatitis C infection (data not shown). An FDA warning previously advised US providers of the occurrence of injection site reactions and the importance of proper injection technique; injection site pain was more prevalent in the XR-NTX group compared with the placebo group, although no severe adverse reactions were reported. No instances of intractable pain were reported, although patients with acute or chronic pain or anticipated pain episodes (eg, elective surgery) were excluded and study investigators were instructed in pain management alternatives to opioid analgesics. Previous studies have shown that the competitive blockade of naltrexone can be overcome: rats given XR-NTX, and then either hydrocodone or fentanyl at 10–20 times the usual doses achieved an analgesia response and did not have signifi cant respiratory depression or sedation.33

A strength of this study was its geographic setting in Russia—one of the many countries where opioid agonist therapy is unavailable,6 but where there is an alarming growth in availability of heroin and the fastest-growing HIV infection rate in the world.34 The report of effi cacy in these seriously ill patients is important both in Russia and as a model for the rest of the world. Patients included in this study share similarities with the opioid-dependent population in other countries, including relatively young age, predominantly male sex, and high rates of infection with HIV and hepatitis C. Nevertheless, given the population and treatment system diff erences, general-isability of these results beyond Russia is a topic for further research. However, in countries with a viable system of opioid agonist maintenance treatment, patient resistance

XR-NTX (n=126)

Placebo (n=124)

p value

Nasopharyngitis 9 (7%) 3 (2%) 0·14

Insomnia 8 (6%) 1 (1%) 0·036

Hypertension 6 (5%) 4 (3%) 0·75

Infl uenza 6 (5%) 5 (4%) >0·99

Injection site pain 6 (5%) 1 (1%) 0·12

Toothache 5 (4%) 2 (2%) 0·45

Headache 4 (3%) 3 (2%) >0·99

≥1 adverse event 63 (50%) 40 (32%) 0·005

≥1 drug-related adverse event 33 (26%) 12 (10%) 0·001

≥1 serious adverse event* 3 (2%) 4 (3%) 0·72

Discontinued owing to adverse events 2 (2%) 2 (2%) ··

Data are number (%). XR-NTX=extended-release naltrexone. *Three patients in the XR-NTX group reported four serious adverse events (infectious processes, eg, AIDS or HIV) and four patients in the placebo group reported fi ve serious adverse events (two infectious, one drug dependence, one psychotic disorder, and one peptic ulcer).

Table 3: Clinical adverse events

Downloaded from ClinicalKey.com at University at Buffalo The State University of New York March 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 29: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

Articles

1512 www.thelancet.com Vol 377 April 30, 2011

to placebo treatment or ethical considerations might make it diffi cult to do a placebo-controlled trial. The extent of patient interest in XR-NTX when opioid substitution treatments are available remains a topic for future health services research; however, there might be interest among those whose employment prohibits opioid use, those with a relatively recent addiction to opioids, and those who wish to secure their recovery after a successful course of agonist therapy. In countries where both XR-NTX and opioid substitution treatments are available, the relative costs of such treatments might be an important factor in their clinical use and accessibility. Another strength of this study was the rigorous defi nition used for opioid abstinence, which included both self-report and urine testing. Furthermore, the imputation that patients who were lost to treatment represented treatment failures was a conservative interpretation that is consistent with the importance of treatment retention and abstinence.

There are several limitations of this study. There was a substantial clinical response to placebo; however, the treatment group still showed greater benefi ts than those in the placebo group. Retention in the placebo group might have been reduced by recognition upon opioid use that one was on placebo or—among patients in the placebo group who had relapsed to regular opioid use—by reluctance to return to the clinic and face a withdrawal reaction from a naloxone challenge test. Despite these possibilities, the placebo group showed a substantial retention and response profi le, and a markedly higher rate of positive naloxone challenge tests. Drug use might have been under-reported on self-report; however, there was a high degree of agreement between results from urine tests and self-report and the urine data was a required confi rmatory element of the primary effi cacy measure. The high retention rate might have been

infl uenced by the inclusion criterion that patients have someone available to supervise attendance, the provision of individual counselling, the absence of alternative treatments (eg, methadone or buprenorphine) in Russia, and the promise of active XR-NTX treatment for all patients after 6 months in the subsequent open-label extension safety study.

Additional research on the practical aspects of opioid antagonist treatment might support further improvement of patient outcomes.35 Patients must be fully detoxifi ed before receiving opioid antagonists to avoid precipitation of opioid withdrawal; thus, methods for antagonist induction and treatment transition need to be optimised. Studies are needed on the diff erential roles of agonist and antagonist maintenance therapies—eg, in early versus late stage illness, in the context of chaotic versus structured social supports, in patients with versus those without chronic pain, or in judicial or employment settings. The worldwide societal eff ects of this disease lend an urgency to the replication of these results and call for research into this treatment approach in diff erent countries and settings, such as primary-care offi ces; in diff erent populations, including those that might be less compliant than the patients included in this study; and on the appropriate duration of treatment, long-term benefi ts and safety, and the health economic and policy aspects.

The results of this study suggest that XR-NTX off ers a new approach—distinct from opioid-agonist mainten-ance—that assists patients in abstaining from opioids and prevents relapse to opioid dependence. Given the heterogeneity of patient needs, to provide optimum care for patients who are opioid dependent, a comprehensive set of treatment options is needed, including existing agonist maintenance treatments, which are well validated both in effi cacy and eff ectiveness research7–10 and psycho-social management. The fi ndings of the present study suggest that antagonist therapy could also play a part. A once-monthly supervised pharmacological treat ment with proven effi cacy that is free of physical dependence and is not subject to illegal diversion might aid com-munity and cultural acceptance of opioid depen dence pharmacotherapy and provide a useful treatment option for many patients.ContributorsEK had full access to the original data, reviewed the data analyses, contributed to data interpretation, wrote the fi rst draft of the manuscript, made fi nal decisions on all parts of the report, and approved the fi nal version of the submitted report. All other authors had access to the data used in the paper and additional data when requests were made and wrote the fi nal draft. EK, EVN, AI, DRG, and BLS designed the study. EK enrolled patients. AI did the statistical analyses and generated tables and fi gures. EK, DRG, and BLS provided study supervision and administrative support.

Confl icts of interestThe Medisorb preparation used in XR-NTX was developed with support from the National Institute on Drug Abuse (grant R43DA013531) and National Institute on Alcohol Abuse and Alcoholism (grant N43AA001002). EK is a consultant for Alkermes and received research funding for this study from Alkermes. EVN was a member of the

Panel: Research in context

Systematic reviewIn systematic reviews, opioid substitution treatment (buprenorphine and methadone) was eff ective in the treatment of opioid dependence,8,9 but such agonist treatments are restricted or unavailable in many countries and might not be suitable for all patients. Systematic reviews of antagonist maintenance with oral naltrexone have generally reported the treatment to be ineff ective because of poor adherence.13

InterpretationIn this study, once-monthly extended release naltrexone (XR-NTX) was superior to placebo with respect to the endpoints of confi rmed abstinence, craving for opioids, retention, and prevention of relapse to opioid dependence. XR-NTX off ers a new treatment option without risk of physical dependence or illegal diversion. This approach might aid community and cultural acceptance of opioid dependence pharmacotherapy.

Downloaded from ClinicalKey.com at University at Buffalo The State University of New York March 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 30: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

Articles

www.thelancet.com Vol 377 April 30, 2011 1513

Alkermes advisory board that designed this trial and was an unpaid consultant to an expert panel convened by Alkermes, with approval from the Columbia University Department of Psychiatry. WL has been an advisory board member for Alkermes and US World Med, has received research funding from Titan Pharmaceuticals, investigator initiated research funding from Hythiam, and research support, an unrestricted educational grant, and speaker support from Reckitt Benckiser. AI, DRG, and BLS are full-time employees of Alkermes.

AcknowledgmentsWe thank the staff at Alkermes, PSI, and Cytel for their work in the implementation and biostatistical analysis of this study, especially Elliot Ehrich, Doreen Campbell, Handong Zhang, Anna O’Rourke, and Stanislav Nikitin. We thank Edward Schweizer, who provided medical writing assistance with the manuscript and was compensated by Alkermes for his assistance. We also thank Shenghan Lai for doing an independent analysis of the data (based on the entire raw data set and assessment of the study protocol and prespecifi ed plan for data analysis), the results of which are the results reported in the manuscript. Johns Hopkins University School of Medicine was paid for Shenghan Lai’s time by Alkermes.

References1 UNODC, World Drug Report 2010. United Nations Publication,

New York, USA. http://www.unodc.org/documents/wdr/WDR_2010/World_Drug_Report_2010_lo-res.pdf (accessed Oct 20, 2010).

2 Hser Y, Anglin MD, Powers K. A 24-year follow-up of California narcotics addicts. Arch Gen Psychiatry 1993; 50: 577–84.

3 Wilcox HC, Conner KR, Caine ED. Association of alcohol and drug use disorders and completed suicide: an empirical review of cohort studies. Drug Alcohol Depend 2004; 76S: S11–19.

4 National Institute of Drug Abuse. NIDA research report: heroin abuse and addiction. 2005. http://www.unodc.org/documents/wdr/WDR_2010/World_Drug_Report_2010_lo-res.pdf (accessed March 24, 2011).

5 Cook C. The global state of harm reduction 2010: key issues for broadening the response. International Harm Reduction Association, London 2010. http://www.ihra.net/fi les/2010/06/29/GlobalState2010_Web.pdf (accessed Nov 11, 2010).

6 Mathers B, Degenhardt L, Ali H, et al, for the 2009 Reference Group to the UN on HIV and Injecting Drug Use. HIV prevention, treatment, and care services for people who inject drugs: a systematic review of global, regional, and national coverage. Lancet 2010; 375: 1014–28.

7 Ball JC, Ross A. The eff ectiveness of methadone maintenance treatment. New York, NY, USA: Springer-Verlag, 1991.

8 Mattick RP, Kimber J, Breen C, Davoli M. Buprenorphine maintenance versus placebo or methadone maintenance for opioid dependence. Cochrane Database Syst Rev 2008; 2: CD002207.

9 Marsch LA. The effi cacy of methadone maintenance interventions in reducing illicit opiate use, HIV risk behavior and criminality: a meta-analysis. Addiction 1988; 93: 515–32.

10 Kleber H. Pharmacologic treatments for opioid dependence: detoxifi cation and maintenance options. Dialogues Clin Neurosci 2007; 9: 455–70.

11 McLellan AT, McKay JR. Components of successful addiction treatment. In: Graham AW, Schultz TK, Mayo-Smith MF, Ries RK, Wilford BB, eds. Principles of addiction medicine, 3rd edn. Chevy Chase, MD, USA: American Society of Addiction Medicine, 2003: 429–42.

12 Nunes EV, Rothenberg JL, Sullivan MA, Carpenter KM, Kleber HD. Behavioral therapy to augment oral naltrexone for opioid dependence: a ceiling on eff ectiveness? Am J Drug Alcohol Depend 2006; 32: 503–17.

13 Minozzi S, Amato L, Vecchi S, Davoli M, Kirchmayer U, Verster A. Oral naltrexone maintenance treatment for opioid dependence. Cochrane Database Syst Rev 2006; 1: CD001333.

14 Hulse GK, Morris N, Arnold-Reed D, et al. Improving clinical outcomes in treating heroin dependence: randomized, controlled trial of oral or implant naltrexone. Arch Gen Psychiatry 2009; 66: 1108–15.

15 Kunøe N, Lobmaier P, Vederhus JK, et al. Naltrexone implants after in-patient treatment for opioid dependence: randomized controlled trial. Br J Psychiatry 2009; 194: 541–46.

16 Comer SD, Sullivan MA, Yu E, et al. Injectable, sustained-release naltrexone for the treatment of opioid dependence. Arch Gen Psychiatry 2006; 63: 210–18.

17 Garbutt JC, Kranzler HR, O’Malley SS, et al. Effi cacy and tolerability of long-acting injectable naltrexone for alcohol dependence: a randomized controlled trial. JAMA 2005; 293: 1617–25.

18 O’Malley SS, Garbutt JC, Gastfriend DR, Dong Q, Kranzler HR. Effi cacy of extended-release naltrexone in alcohol-dependent patients who are abstinent before treatment. J Clin Psychopharmacol 2007; 27: 507–12.

19 Gastfriend DR. Intramuscular extended-release naltrexone: current evidence. Ann NY Acad Sci 2011; 1216: 144–66.

20 American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 4th edn, text revision (DSM-IV-TR). Washington, DC: American Psychiatric Association, 2000.

21 Mercer DE, Woody GE. Therapy manuals for drug addiction series. Individual Drug Counseling. Rockville, MD: National Institutes of Health, US Department of Health and Human Services, 1999.

22 Sobell LC, Sobell MB. Timeline follow-back: a technique for assessing self-reported alcohol consumption. In: Litten RZ, Allen JP, eds. Measuring alcohol consumption: psychosocial and biological methods. Totowa, NJ: The Humana Press, 1992: 41–72.

23 Krupitsky EM, Zvartau EE, Masalov DV, et al. Naltrexone for heroin dependence treatment in St. Petersburg, Russia. J Subst Abuse Treat 2004; 26: 285–94.

24 Metzger DS, Navaline HA, Woody GE. Assessment of substance abuse: HIV risk assessment battery (RAB). In: Carson-DeWitt R, Macmillan-Thomson G, eds. Encyclopedia of drugs, alcohol, and addictive behavior. New York: Macmillan Reference, 2001.

25 Ware JE, Kosinski M, Dewey JE. How to score version two of the SF-36 Health Survey. Lincoln, RI: QualityMetric, 2000.

26 Euroqol Group. Euroqol—a new facility for the measurement of health-related quality of life. Health Policy 1990; 16: 199–208.

27 Guy W, ed. ECDEU Assessment for psychopharmacology, revised edition. Rockville, MD, USA: NIMH Publications, 1976.

28 van der Waerden BL. Order tests for the two-sample problem. II, III. Proc K Ned Akad Wet A 1953; 564: 303–16.

29 Holm S. A simple sequentially rejective multiple test procedure. Scand J Statist 1979; 6: 65–70.

30 Amirdjanova VN, Goryachev DV, Korshunov NI, Rebrov AP, Sorotskaya VN. Population indices of quality of life on SF-36: results of the multicentral study of quality of life “MIRAZH”. Rheumatol Sci Pract 2008; 1: 36–48.

31 Dijkstra BA, De Jong CA, Bluschke SM, Krabbe PF, van der Staak CP. Does naltrexone aff ect craving in abstinent opioid-dependent patients? Addict Biol 2007; 12: 176–82.

32 Dunbar JL, Turncliff RZ, Dong Q, Silverman BL, Ehrich EW, Lasseter KC. Single- and multiple-dose pharmacokinetics of long-acting injectable naltrexone. Alcohol Clin Exp Res 2006; 30: 480–90.

33 Dean RL, Todtenkopf MS, Deaver DR, et al. Overriding the blockade of antinociceptive actions of opioids in rats treated with extended-release naltrexone. Pharmacol Biochem Behav 2008; 89: 515–22.

34 The Paris Pact Initiative. Illicit drug trends in the Russian Federation. United Nations Offi ce on Drugs and Crime Regional Offi ce for Central Asia. Moscow, April 2008. http://www.unodc.org/documents/regional/central-asia/Illicit%20Drug%20Trends% 20Report_Russia.pdf (accessed April 25, 2010).

35 Lobmaier P, Gossop M, Waal H, Bramness J. The pharmacological treatment of opioid addiction—a clinical perspective. Eur J Clin Pharmacol 2010; 66: 537–45.

Downloaded from ClinicalKey.com at University at Buffalo The State University of New York March 29, 2017.For personal use only. No other uses without permission. Copyright ©2017. Elsevier Inc. All rights reserved.

Page 31: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

Injectable extended-release naltrexone for the prevention of relapse to opioid dependence following opioid detoxification Evgeny Krupitsky Neuropsychiatry. 2.4 (Aug. 2012): p355. DOI: http://dx.doi.org.gate.lib.buffalo.edu/10.2217/npy.12.34 Copyright: COPYRIGHT 2012 Future Medicine Ltd. http://www.futuremedicine.com.gate.lib.buffalo.edu/page/journal/npy/aims.jsp

Author(s): Evgeny Krupitsky 1

Opioid dependence continues to be a worldwide problem [101,102] . Prevalence rates for heroin and other opiates range between 0.3 and 0.5% of the world's population aged between 15 and 64 years [101] . A dramatic rise in the non-medical use of opioid pain medications has also been occurring, particularly in the USA, with an estimated 1.9 million Americans abusing or dependent on prescription pain medications in 2010 [1] . Dependence on opioids is associated with increased morbidity and mortality, poor social functioning, economic dependence and crime [2-4] . The economic burden to society of opioid-use disorders is large, with total US societal costs of prescription opioid abuse estimated at US$55.7 billion in 2007 [5] and total US costs of heroin addiction estimated at US$21.9 billion in 1996 [6] .

Available treatment modalities vary across different countries, with the most common approaches consisting of either agonist maintenance pharmacotherapy or drug-free psychosocial treatment. Maintenance pharmacotherapy options include methadone (a µ-opioid receptor agonist) or buprenorphine (a partial agonist). The efficacy and safety of buprenorphine and methadone are documented by numerous studies [7,8] . In the majority of UN member countries (122 of 192), however, agonist therapy is unavailable or restricted owing to concerns about physiological dependence or abuse and illegal diversion [2] . In addition, agonist therapy is sometimes not the preferred treatment for specific types of patients. This includes young people, those with a brief history of addiction or who are new to treatment, and those whose employment may prohibit opioid use (e.g., healthcare providers, pilots and police, fire, emergency and military personnel). Drug-free psychosocial treatment is an option for these and other patients, but is associated with high rates of relapse [9] .

Opioid dependence can also be treated with naltrexone, a µ-opioid receptor antagonist. However, in general, problems with adherence to oral naltrexone have undermined its efficacy in the treatment of opioid dependence [10] . This problem with adherence was anticipated by the US National Institute on Drug Abuse as early as 1976 and led to requests for the development of a long-acting opioid antagonist. Following this request, Alkermes, Inc. (MA, USA) developed a once-monthly extended-release formulation of injectable naltrexone (XR-NTX, Vivitrol® ) [11] . XR-NTX gradually releases naltrexone from microspheres composed of medical-grade polylactide-co -glycolide, a polymer used in dissolvable surgical sutures. This article will review the clinical trial data on which approval of XR-NTX for opioid dependence was based, and present information

Page 32: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

on its administration, clinical pharmacology, mechanism of action, pharmacodynamics, pharmacokinetics and its associated adverse events and labeled warnings.

Indications & usage

In its oral form, naltrexone was approved by the US FDA for treatment of opioid dependence in 1984. The extended-release formulation was approved more recently (October 2010) in the USA for prevention of relapse to opioid dependence among detoxified individuals as part of a comprehensive management program that includes psychosocial support. Prior to its approval for opioid dependence, XR-NTX was approved for use in the treatment of alcohol dependence in both the USA and Russia.

XR-NTX is contraindicated in patients with acute hepatitis or liver failure, patients receiving opioid analgesics, patients with current physiologic opioid dependence, patients in acute opioid withdrawal, any individual who has failed a naloxone challenge test or has a positive urine screen for opioids, and patients who have previously exhibited hypersensitivity to naltrexone, polylactide-co -glycolide, carboxymethyl cellulose or any other components of the diluent.

Dosage & administration

The standard dosage of XR-NTX is 380 mg delivered as an intramuscular gluteal injection. Injections are delivered every 4 weeks (or once a month) by a healthcare professional. It is recommended that injections be administered in alternating buttocks over the course of treatment. If a dose is missed, the next dose should be administered as soon as possible. To assure proper release kinetics and avoid microsphere particle entry into the circulatory system, XR-NTX should never be administered intravenously.

It is not required that patients be pretreated with oral naltrexone before beginning XR-NTX injections, however, patients should be opioid free for 7-10 days prior to treatment with XR-NTX.

Clinical pharmacology

* Mechanism of action

Naltrexone, and its active metabolite 6-[beta]-naltrexol, are antagonists with a high affinity for the µ-opioid receptor. The clinical efficacy of naltrexone results from blocking the effects of opioids through competitive binding at these receptors. The ability of XR-NTX to sustain a blockade of opioid receptors in opioid abusers over the month following an injection was recently demonstrated in an opioid challenge experiment [12] . In this study, low subjective ratings of 'any drug effect', indicating blockade, were maintained for a full 28-day period for each of three dosage levels that were tested in the experiment (75, 150 and 300 mg) of XR-NTX, and the FDA-approved formulation is marketed at a higher naltrexone dose (380 mg).

Page 33: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

With XR-NTX, extended release is achieved through the embedding of naltrexone within a matrix of microspheres (<100 µm diameter) made of polylactide- co -glycolide. Polylactide-co -glycolide is a common biodegradable copolymer that has been used safely in various human applications, including sutures, orthopedics, bone plates and other extended-release medications.

* Pharmacodynamics

Beyond its opioid-blocking properties, naltrexone has few effects on the human body. Some pupillary constriction is evident with naltrexone, but the mechanism of this effect is unknown. XR-NTX is not associated with the development of tolerance or dependence on naltrexone. However, in individuals who are physiologically dependent on opioids, naltrexone and XR-NTX will precipitate acute withdrawal. This is why individuals must be detoxified from opioids before initiating XR-NTX.

Clinical concerns about the effects of sustained blockade of the µ-opioid receptor on experienced pleasure have led to research investigating whether or not XR-NTX reduces pleasure from activities such as sex, exercise, food and other daily activities. In one report, alcohol-dependent patients (n = 74), at the end of receiving XR-NTX injections nearly continuously for 3-5 years were asked to rate how pleasurable a number of daily activities were on a 1 ('not at all') to 5 ('very much') scale in the prior 90 days [13] . A minority of patients rated drinking alcohol as 'moderately', 'quite a bit' or 'very much' pleasurable, whereas 60-92% rated exercise, sex, eating good food and six other common activities in these categories [13] . Although the study did not assess baseline pleasure ratings or outcomes with opioid-dependent patients, it suggests that the effect of long-term XR-NTX on hedonic response may operate on a gradient, with greater attenuation for alcohol reward than for other rewarding and more healthy stimuli.

Enhanced reactivity to conditioned cues is believed to play an important role in relapse with substance-use disorders. The impact of XR-NTX on such conditioned cues as measured by a blood-oxygen-level-dependent/functional MRI cue-reactivity procedure has been investigated with alcohol-dependent subjects [14] . In this study, the blood-oxygen-level-dependent functional MRI cue-reactivity procedure was conducted immediately before, and 2 weeks after, an XR-NTX or placebo injection. Results indicated that XR-NTX attenuates the salience of cues that have been associated with alcohol. This effect of XR-NTX on brain function may interrupt the process through which conditioned cues can trigger 'slips' and relapse. However, the extent to which these results generalized to opioid-dependent individuals is not known.

* Pharmacokinetics

Following an injection with XR-NTX, there is an initial peak in naltrexone plasma concentrations after approximately 2 h [15] . A second peak occurs approximately 2-3 days later. Concentrations slowly decline 14 days postinjection, but measurable levels persist for more than 1 month. The overall median peak concentration obtained in the pharmacokinetic study was 12.9 ng/ml [15] . Other investigations have indicated that

Page 34: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

plasma concentrations of naltrexone of less than 1 ng/ml are sufficient to antagonize heroin-induced effects [16,17] .

Maximum plasma concentration and the area under the curve for naltrexone and 6-[beta]-naltrexol (the major metabolite of naltrexone) after administration of XR-NTX are proportional to dose [15] . Total naltrexone dose is lower with a single dose of 380 mg XR-NTX compared with oral dosing with 50 mg naltrexone over 28 days (i.e., 1400 mg), but the area under the curve is three- to fourfold higher [15] . Following the first injection, a steady state is reached at the end of the 1-month dosing interval. Repeat injections of XR-NTX show minimal accumulation (<15%) of naltrexone or 6-[beta]-naltrexol.

Naltrexone is extensively metabolized in humans. Production of the metabolite 6-[beta]-naltrexol is mediated by dihydrodiol dehydrogenase. The cytochrome P450 system is not involved in naltrexone metabolism. Two other minor metabolites are 2-hydroxy-3-methoxy-6-[beta]-naltrexol and 2-hydroxy-3-methoxy-naltrexone.

Significantly less 6-[beta]-naltrexol is generated with injection of XR-NTX compared with oral administration of naltrexone owing to a reduction in first-pass hepatic metabolism with XR-NTX [15] . Elimination of naltrexone and its metabolites occurs primarily via urine; there is minimal excretion of unchanged naltrexone. The elimination half-life of naltrexone and 6-[beta]-naltrexol following administration of XR-NTX is 5-10 days [15] .

Clinical evidence

* Overview of randomized, placebo-controlled clinical trials

The first evidence of efficacy for a long-acting injectable naltrexone formulation came from a small, short-term (2 month) controlled trial using a product that was not submitted for approval in the USA [18] . This study compared placebo with 192 and 384 mg of long-acting injectable naltrexone to placebo. Retention in treatment was dose related, with 39, 60 and 68% of patients, respectively, remaining in treatment at the end of 2 months. The percentage of urine samples negative for opioids, methadone, cocaine, benzodiazepines and amphetamines was significantly higher for the 384-mg group versus placebo (p < 0.001) and for 192 mg versus placebo (p = 0.046) when missing urines were considered positive. However, when missing urines were not considered positive, these group comparisons were no longer significant.

The primary efficacy study for the marketed version of XR-NTX was a 6-month placebo-controlled study conducted at 13 clinical sites in Russia [19] . This study is the only published controlled trial of XR-NTX for opioid dependence. The current review therefore focuses on the results of this trial.

Opioid-dependent (primarily heroin) individuals who had completed inpatient opioid detoxification participated. Following detoxification, XR-NTX was injected every 4 weeks, for a total of six injections over 24 weeks. Patients were randomized to XR-NTX (n = 126) or placebo (n = 124), with all participants also receiving up to 12 biweekly

Page 35: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

sessions of individual drug counseling in conjunction with injection visits. The primary efficacy measure was a response profile, defined as the proportion of patients at each possible response level of confirmed opioid-free weeks who achieved that amount (or greater) of opioid-free weeks (using only data from weeks 5 to 24). Weekly confirmed abstinence was defined as the following: the patient provided urine for drug testing, the testing was negative and the patient reported no opioid use. Thus, missing urines were coded as positive for opioids. Patients who used illicit opioids during the trial continued on treatment.

The results of the trial indicated that XR-NTX was statistically and clinically superior to placebo on all a priori primary (p = 0.0002) (Figure 1) and secondary efficacy measures (Table 1). The median XR-NTX patient had confirmed abstinence for [greater than or equal]90% of weeks versus 35% for placebo and the mean total of confirmed abstinence was 35.7% weeks with XR-NTX versus 22.6% with placebo. There was a greater reduction in opioid craving in the XR-NTX group compared with placebo by week 8 and that difference persisted every week through week 24 (p 168 days for the XR-NTX group versus 96 days for the placebo group, with 67 in the XR-NTX group and 47 in the placebo group completing all six injections (p = 0.017). XR-NTX patients attended a median of 12 counseling sessions versus eight for placebo patients. The XR-NTX group also improved more than the placebo group with regard to relapse to physiological opioid dependence (p < 0.0001), HIV-risk behaviors (p = 0.025), self-reported health status (p = 0.0005), clinician ratings of global improvement (p = 0.0002), and health-related quality of life (mental component; p = 0.0043). It should be noted that the absolute levels of improvement in both the XR-NTX and placebo groups occurred in conjunction with the provision of individual drug counseling. Further research is needed to determine the contribution of counseling to the overall degree of improvement evident with XR-NTX.

* Health economic outcomes

The healthcare costs associated with treatment of opioid dependence with psychosocial treatment alone, methadone, buprenorphine, oral naltrexone or XR-NTX have been examined using claims data from a large US health plan [20] . In this study, analyses focused on 6-month medication persistence, healthcare utilization, direct paid claims for opioid-dependence medications, detoxification and rehabilitation, opioid-related and nonrelated inpatient admissions, outpatient services and total costs. Although the pharmacy costs for XR-NTX are more than other treatments, total healthcare costs (combining inpatient, outpatient and pharmacy) were found to be greatest with psychosocial treatment alone, and XR-NTX total costs were not significantly different from oral naltrexone or buprenorphine and were 49% lower than with methadone. Although study limitations include retrospective design using case-mix adjustment, lack of indirect costs (e.g., job absenteeism or criminal justice costs) and a focus only on individuals with commercial insurance, XR-NTX-treated patients had fewer opioid-related and nonopioid-related hospitalizations than patients receiving any of the approved oral medications for opioid dependence.

Page 36: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

* Adverse reactions

XR-NTX is generally well tolerated. In the published Phase III clinical trial, two patients in both groups discontinued treatment due to adverse events (drug-dependence relapse, psychotic disorder, hepatitis C and nausea) [19] . Moreover, no overdose events, suicide attempts, deaths or other severe adverse events were reported in the trial. Overall, more patients in the XR-NTX group reported an adverse event than in the placebo group (50 vs 32.3%), but no adverse events were judged to be severe.

Rates of specific adverse events with the use of XR-NTX in an opioid-dependent population were low (Table 2) [21] . Only one adverse event (insomnia) showed a significantly greater incidence for the XR-NTX group compared with placebo (6 vs 1%). Injection-site pain was more prevalent in XR-NTX versus placebo patients (5 vs 1%), and a FDA warning has advised US providers of the occurrence of injection-site reactions and the importance of proper injection technique.

Hepatic enzyme abnormalities were more common with XR-NTX compared with placebo and XR-NTX has a boxed warning regarding naltrexone hepatotoxicity [16] . The Phase III trial sample had a high baseline incidence of hepatitis C (89%) and HIV infection (41%); however, abnormal liver function tests over the course of treatment occurred only in patients with existing hepatitis C infections, were transient and not clinically meaningful.

A clinical concern with the use of naltrexone is whether the blockade of µ-opioid receptors is potentially surmountable. There has been one published case report of an individual who overcame the blockade from a surgically implanted version of long-acting naltrexone (approved for use in Russia) by using very large amounts of heroin [22] . In an animal study, rats administered XR-NTX showed no significant respiratory depression or sedation when given hydrocodone or fentanyl at ten- to 20-times the usual doses to achieve an analgesic response [23] . There have been published reports of death from opioid overdose with the implanted version of long-acting naltrexone both during treatment and following removal of the implant [24,25] .

* Use in special populations

The use of XR-NTX in patients with hepatic impairment deserves comment. A boxed warning in the package insert for oral naltrexone, and subsequently XR-NTX, in relation to hepatotoxicity was prompted by early studies reporting hepatotoxicity at very high dosages of oral naltrexone (350 mg/day) in obese patients and those with senile dementia [26] . To address this concern, a study examined the pharmacokinetics of XR-NTX at the 190-mg dose (although not at the full 380-mg marketed dose) among a small sample of individuals with mild-to-moderate hepatic impairment [27] . Results of the study indicated no difference in pharmacokinetic parameters between those with mild-to-moderate hepatic impairment compared with controls following administration of XR-NTX. Similarly, transient and clinically insignificant enzyme elevations were found but

Page 37: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

no evidence for hepatotoxicity in a detailed analysis of hepatic safety in the use of XR-NTX for alcohol dependence [28] .

The safety, efficacy and pharmacokinetics of XR-NTX in a pediatric population has not been examined to date. The use of XR-NTX during pregnancy, labor and delivery and with nursing mothers, has also not been investigated.

XR-NTX is contraindicated in patients known or expected to have a need for opioid analgesia. Administration of XR-NTX in conjunction with other medications has not been explored in clinical drug interaction studies to date. Prescribers should therefore weigh the risks and benefits of concomitant drug use when considering XR-NTX. However, in the Phase III study of XR-NTX for treatment of alcohol dependence, almost 30% of patients were taking concomitant antidepressants without any evident safety concerns [29] .

* Clinical & practice issues

Although XR-NTX has demonstrated efficacy in a 6-month trial, clinicians will need guidance on how long to continue the injections. This issue remains to be addressed in future studies.

Another key issue for clinical practice is how best to rapidly and safely transition a patient from agonist use to XR-NTX antagonist therapy. Recommendations have been provided for such a transition that tailor the detoxification strategy to the severity of physiological opioid dependence [30] . The suggested approach typically begins with a 4-8 mg dose of buprenorphine, particularly for moderate-to-severely dependent individuals; no buprenorphine may be needed for mildly dependent individuals. This is combined with clonidine and other ancillary medications, followed by 1-2 days of progressive oral naltrexone doses before initiating XR-NTX. Moderately dependent individuals may require partial hospitalization for this regimen and severely dependent individuals may require an inpatient setting. One limitation of this buprenorphine-clonidine-naltrexone procedure is that precipitated withdrawal must be anticipated and actively managed. Furthermore, the efficacy and safety of this approach needs to be investigated in controlled trials. As mentioned, the product labeling for XR-NTX requires that a patient be opioid free for a minimum of 7-10 days.

An alternative method of detoxification involves a gradual taper, first substituting 2-4 mg buprenorphine when withdrawal symptoms emerge, usually 12-18 h after the last dose of heroin or other short-acting opioid, and then titration up to 4-16 mg of buprenorphine per day until withdrawal symptoms are suppressed, then tapering to 0 mg over the next 7-14 days. This would then be followed by the appropriate opioid-free period before initiating XR-NTX [30] .

A further consideration in the use of XR-NTX is pain management. Two options are regional analgesia and use of nonopioid analgesics. If possible, a patient can schedule surgery after discontinuing XR-NTX. Because the blockade of µ-opioid receptors by

Page 38: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

naltrexone is competitive, it is surmountable. Thus, opioid pain therapy can be implemented as part of anesthesia or analgesia. However, if opioids are used, the patient needs to be continuously monitored in an anesthesia care setting by persons not involved in the conduct of the surgical or diagnostic procedure. The healthcare professional providing the opioid therapy should be trained in the management of the respiratory effects of potent opioids, specifically the establishment and maintenance of a patent airway and assisted ventilation and the setting equipped and staffed for cardiopulmonary resuscitation.

Although the challenges of successful transition from agonist use and pain management are important clinical issues, the approaches to these challenges described above have been met with success in routine practice [30,31] . In addition, these challenges are not dissimilar to those evident with the use of short-acting opioid antagonists. These challenges also have to be weighed in the context of a once-monthly treatment for prevention of relapse in opioid-dependent individuals that is free of physical dependence, addresses the compliance of oral medications and is not subject to illegal diversion. XR-NTX represents a distinct alternative to previously existing treatment options for eligible patients.

Table 1. Efficacy results from a Phase III clinical trial of extended-release naltrexone versus placebo.

Efficacy measure XR-NTX

(n = 126)

Placebo (n

= 124) p-value

Response profile, median proportion (95%

CI) of weeks of confirmed abstinence for

weeks 5-24

90.0%

(69.9-92.4)

35.0%

(11.4-63.8) 0.0002

Median (95% CI) self-report of opioid-free

days over 24 weeks

99.2%

(89.1-99.4)

60.4%

(46.2-94.0) 0.0004

Total confirmed abstinence over weeks 5-

24 (percent; 95% CI)

45 (35.7%;

27.4-44.1)

28 (22.6%;

15.2-29.9) 0.0224

Craving, VAS score, mean change from

baseline (95% CI; scale: 0-100; baseline:

XR-NTX 18.2; PBO 21.8)

-10.1 (-

12.3 to -

7.8)

+0.7 (-3.1-

4.4)

168[double

dagger]

96

(63-

165)

0.0042[dagger]

Treatment completion, number (percent;

95% CI) of patients who completed

double-blind treatment period

67 (53.2%;

44.5- 61.9)

47 (37.9%;

29.4-46.4) 0.0171

Relapse to physical opioid dependence;

number (percent; 95% CI) of participants

with positive naloxone challenge test

1 (0.8%;

0.0-2.3)

17 (13.7%;

7.7-19.8) <0.0001

Risk for AIDS behavior scores; change

from baseline, mean (95% CI; scale: 0-1;

baseline: XR-NTX 0.300; PBO 0.279)

-0.187 (-

0.224 to -

0.150)

-0.130 (-

0.173 to -

0.087)

0.0212

EQ-5D[trademark] General Health State

VAS self-ratings; change from baseline,

+14.1 (9.6-

18.7)

+2.7 (-1.9-

7.8) 0.0005

Page 39: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

Efficacy measure XR-NTX

(n = 126)

Placebo (n

= 124) p-value

mean (95% CI; scale: 0-100; baseline: XR-

NTX 68.7; PBO 69.9)

CGI improvement ratings; percentage of

patients rated by investigators as much or

very much improved (95% CI)

85.9%

(77.8-94.0)

57.5%

(45.7-69.5) 0.0002

[dagger] p-value adjusted for multiplicity.

[double dagger] Confidence interval could not be calculated because median exceeds study duration.

CGI: Clinical Global Impression; PBO: Placebo; VAS: Visual analog scale; XR-NTX: Extended-release naltrexone.

Data taken from [14] .

Table 2. Adverse events in the extended-release naltrexone Phase III trial.

Adverse events XR-NTX 380 mg; n = 126; n (%) Placebo; n = 124; n (%)

Nasopharyngitis 9 (7) 3 (2)

Insomnia 8 (6)[dagger] 1 (1)

Hypertension 6 (5) 4 (3)

Influenza 6 (5) 5 (4)

Injection-site pain 6 (5) 1 (1)

Toothache 5 (4) 2 (2)

Headache 4 (3) 3 (2)

[dagger] p < 0.05 different from placebo.

Data taken from [16] .

Practice points

* Injectable extended-release naltrexone (XR-NTX), approved in the USA for the prevention of relapse among detoxified opioid-dependent individuals, was formulated to address the adherence problem that limited the clinical usefulness of oral naltrexone.

* XR-NTX is administered monthly by intramuscular injection by a healthcare professional and should be accompanied by psychosocial counseling.

* A Phase III multicenter, placebo-controlled, randomized clinical trial found XR-NTX to be superior to placebo on all primary and secondary end points.

Page 40: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

* Naturalistic analysis of a large healthcare claims database found that total healthcare costs following treatment with XR-NTX were not significantly different from oral naltrexone or buprenorphine, and were 49% lower than with methadone.

* In the Phase III trial, adverse events were mild-to-moderate in severity, with nasopharyngitis, insomnia and injection-site pain occurring more frequently with XR-NTX than placebo. Discontinuations due to adverse events were similar in both groups.

* Unanswered questions include the efficacy of XR-NTX therapy in different settings, such as primary care offices, and the appropriate duration and long-term safety of XR-NTX treatment.

CAPTION(S):

Figure 1. Percentage of confirmed opioid-free weeks (cumulative) among participants treated with extended-release naltrexone versus placebo in the Phase III trial.

On this graph, the y-axis represents each decile of possible opioid-free weeks (for the 20-week period of weeks 5-24; the first 4 weeks were excluded to take into account patient testing/opioid use extinction). The x-axis is the percentage of participants who achieved each amount (or greater) of aggregated opioid-free weeks. For example, the percentage of patients who achieved 100% confirmed opioid-free weeks was 23% for placebo versus 36% for XR-NTX; the median placebo patient (vertical dashed line) achieved 35% of opioid-free weeks versus 90% in the XR-NTX group.

XR-NTX: Extended-release naltrexone.

Adapted with permission from [19] © 2011 Elsevier Ltd.

References

Papers of special note have been highlighted as: * of interest ** of considerable interest

1 Substance Abuse and Mental Health Services Administration. Results from the 2010 National Survey on Drug Use and Health: Summary of National Findings , NSDUH Series H-41, HHS Publication No. (SMA) 11-4658. Substance Abuse and Mental Health Services Administration, Rockville, MD, USA (2011).

2 Mathers B, Degenhardt L, Ali H et al. HIV prevention, treatment, and care services for people who inject drugs: a systematic review of global, regional, and national coverage. Lancet 375(9719), 1014-1028 (2010).

3 Degenhardt L, Bucello C, Mathers B et al. Mortality among regular or dependent users of heroin and other opioids: a systematic review and meta-analysis of cohort studies. Addiction 106, 32-51 (2011).

Page 41: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

4 Wall R, Rehm J, Fischer B et al. Social costs of untreated opioid dependence. J. Urb. Health 77, 688-722 (2000).

5 Birnbaum HG, White AG, Schiller M, Waldman T, Cleveland JM, Roland CL. Societal costs of prescription opioid abuse, dependence, and misuse in the United States. Pain Med. 12, 657-667 (2011).

6 Mark TL, Woody GE, Juday T, Kleber HD. The economic costs of heroin addiction in the United States. Drug Alcohol Depend. 61, 195-206 (2001).

7 Mattick RP, Kimber J, Breen C, Davoli M. Buprenorphine maintenance versus placebo or methadone maintenance for opioid dependence. Cochrane Database Syst. Rev. 2, CD002207 (2008).

8 Kleber H. Pharmacologic treatments for opioid dependence: detoxification and maintenance options. Dialogs Clin. Neurosci. 9, 455-470 (2007).

9 Ball JC, Ross A. The Effectiveness of Methadone Maintenance Treatment . Springer-Verlag, NY, USA (1991).

10 Gueorguieva R, Wu R, Pittman B et al. New insights into the efficacy of naltrexone based on trajectory-based reanalyses of two negative clinical trials. Biol. Psychiatry 61, 1290-1295 (2007).

11 Gastfriend DR. Intramuscular extended-release naltrexone: current evidence. Ann. NY Acad. Sci. 1216, 144-166 (2011).

* Comprehensive review of all studies that have focused on the use of extended-release naltrexone (XR-NTX) in a variety of populations.

12 Bigelow GE, Preston KL, Schmittner J, Dong Q, Gastfriend DR. Opioid challenge evaluation of blockade by extended-release naltrexone in opioid-abusing adults: dose-effects and time-course. Drug Alcohol Depend. 123(1-3), 57-65 (2012).

13 O'Brien CP, Gastfriend DR, Forman RF, Schweizer E, Pettinati HM. Long-term opioid blockade and hedonic response: preliminary data from two open-label extension studies with extended-release naltrexone. Am. J. Addict. 20, 106-112 (2011).

14 Lukas SE. Extended-release injectable naltrexone (XR-NTX) treatment of alcohol dependence: reduction in cue-induced craving and CNS activation on fMRI. Presented at: The Research Society on Alcoholism . San Antonio, TX, USA, 26-30 June 2010.

15 Dunbar JL, Turncliff RZ, Dong Q, Silverman BL, Ehrich EW, Lasseter KC. Single and multiple dose pharmacokinetics of long-acting naltrexone. Alcohol Clin. Exp. Res. 30, 480-490 (2006).

Page 42: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

16 Chiang CN, Hollister LE, Kishimoto A, Barnett G. Kinetics of analtrexone sustained-release preparation. Clin. Pharmacol. Ther. 36, 704-708 (1984).

17 Comer SD, Collins ED, Kleber HD, Nuwayser ES, Kerrigan JH, Fischman MW. Depot naltrexone: long-lasting antagonism of the effects of heroin in humans.Psychopharmacology 159, 351-360 (2002).

18 Comer SD, Sullivan MA, Yu E et al. Injectable, sustained-release naltrexone for the treatment of opioid dependence. Arch. Gen. Psychiatry 63, 210-218 (2006).

19 Krupitsky E, Nunes EV, Ling W, Illeperuma A, Gastfriend DR, Silverman BL. Injectable extended-release naltrexone for opioid dependence: a double-blind, placebo-controlled, multicenter randomised trial. Lancet 377, 1506-1513 (2011).

** Phase III randomized trial conducted in Russia that was the primary study confirming the safety and efficacy of 6-months of XR-NTX for opioid dependence.

20 Baser O, Chalk M, Fiellin DA, Gastfriend DR. Cost and utilization outcomes of opioid dependence treatments. Am. J. Manag. Care 17, S235-S248 (2011).

* Analysis of a large claims database that found that healthcare costs following treatment with XR-NTX were not significantly different from oral naltrexone or buprenorphine, and were 49% lower than with methadone.

21 VIVITROL® , package insert. Alkermes Inc., Dublin, Ireland.

22 Krupitsky EM, Burakov AM, Tsoy MV et al. Overcoming opioid blockade from depot naltrexone. Addiction 102, 1164-1165 (2007).

23 Dean RL, Todtenkopf MS, Deaver DR et al. Overriding the blockade of antinociceptive actions of opioids in rats treated with extended-release naltrexone. Pharmacol. Biochem. Behav. 89, 515-522 (2008).

24 Oliver P, Horspool M, Keen J. Fatal opiate overdose following regimen changes in naltrexone treatment. Addiction 100, 560-563 (2005).

25 Gibson AE, Degenhardt LJ, Hall WD. Opioid overdose deaths can occur in patients with naltrexone implants. Med. J. Aust. 186, 152-153 (2007).

26 Pfohl DN, Allen JI, Atkinson RL et al. Naltrexone hydrochloride (trexan): a review of serum transaminase elevations at high dosage. NIDA Res. Monogr. 67, 66-72 (1986).

27 Turncliff RZ, Dunbar JL, Dong Q et al. Pharmacokinetics of long-acting naltrexone in subjects with mild-to-moderate hepatic impairment. J. Clin. Pharmacol. 45, 1259-1267 (2005).

Page 43: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

28 Lucey MR, Silverman BL, Illeperuma A, O'Brien CP. Hepatic safety of once-monthly injectable extended-release naltrexone administered to actively drinking alcoholics. Alcohol Clin. Exp. Res. 32, 498-504 (2008).

29 Garbutt JC, Kranzler HR, O'Malley SS et al. Efficacy and tolerability of long-acting injectable naltrexone for alcohol dependence: a randomized controlled trial. JAMA 293, 1617-1625 (2005).

* Phase III randomized, placebo-controlled, multicenter trial demonstrating the efficacy of XR-NTX with alcohol-dependent patients.

30 Sigmon SC, Bisaga A, Nunes EV, O'Connor PG, Kosten T, Woody G. Opioid detoxification and naltrexone induction strategies: recommendations for clinical practice. Am. J. Drug Alcohol Abuse 38(3), 187-199 (2012).

31 Ockert DM, Volpicelli JR, Baier AR, Coons EE, Fingesten A. A nonopioid procedure for outpatient opioid detoxification. J. Addict. Med. 5, 110-114 (2010).

* Websites

101 UNODC: World Drug Report 2010 (2010). (United Nations Publication, Sales No. E.10.XI.13). www.unodc.org/documents/wdr/WDR_2010/World_Drug_Report_2010_lo-res.pdf (Accessed 20 October 2010)

102 Cook C. The Global State of Harm Reduction 2010: Key issues for broadening the response. International Harm Reduction Association, London (2010). www.ihra.net/files/2010/06/29/GlobalState2010_Web.pdf (Accessed 11 November 2010)

Author Affiliation(s):

[*] Department of Addictions, Bekhterev Research Psychoneurological Institute & St Petersburg State Pavlov Medical University, 3 Bekhtereva Street, St Petersburg 192019, Russia. [email protected]

Financial & competing interests disclosure

The author was a consultant for Alkermes, Inc. and received research funding from Alkermes in 2007-2010. The author has no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Editorial support was provided by Edward Schweizer of Paladin Consulting Group, Inc. (Princeton, NJ, USA) through a contract from Alkermes, Inc.

Page 44: Extended-Release Naltrexone to Prevent Opioid Relapse in ... Article...Extended-release naltrexone, a sustained-release monthly injectable formulation of the full mu-opioid receptor

Evgeny Krupitsky

Source Citation (MLA 8th Edition) Krupitsky, Evgeny. "Injectable extended-release naltrexone for the prevention of relapse to

opioid dependence following opioid detoxification." Neuropsychiatry, vol. 2, no. 4, 2012, p. 355+. Academic OneFile, go.galegroup.com/ps/i.do?p=AONE&sw=w&u=sunybuff_main&v=2.1&id=GALE%7CA323966333&it=r&asid=0b954241993c2914994b6d8f7a9ebe8d. Accessed 29 Mar. 2017.

Gale Document Number: GALE|A323966333