Naltrexone: Difference between revisions

From Wikipedia, the free encyclopedia
Content deleted Content added
WP:MEDMOS, other changes (e.g., history, names, analogues).
Line 4: Line 4:
| Watchedfields = changed
| Watchedfields = changed
| verifiedrevid = 464372546
| verifiedrevid = 464372546
| IUPAC_name = (4R,4aS,7aR,12bS)-3-(cyclopropylmethyl)-4a,9-dihydroxy-2,4,5,6,7a,13-hexahydro-1''H''-4,12-methanobenzofuro[3,2-e]isoquinoline-7-one
| drug_name =
| IUPAC_name = 17-(cyclopropylmethyl)-4,5α-epoxy- 3,14-dihydroxymorphinan-6-one| image = Naltrexone_skeletal.svg
| image = Naltrexone_skeletal.svg
| width = 200px
| image2 = Naltrexone-3D-balls.png
| image2 = Naltrexone-3D-balls.png
| width2 = 250px


<!--Clinical data-->
<!--Clinical data-->
| tradename = Revia, Vivitrol, other
| tradename = Revia, Vivitrol, others
| Drugs.com = {{drugs.com|monograph|naltrexone}}
| Drugs.com = {{drugs.com|monograph|naltrexone}}
| MedlinePlus = a685041
| MedlinePlus = a685041
Line 19: Line 21:
| legal_US = Rx-only
| legal_US = Rx-only
| legal_status = Rx-only
| legal_status = Rx-only
| routes_of_administration = By mouth, intramuscular
| routes_of_administration = [[Oral administration|By mouth]], [[intramuscular injection]]


<!--Pharmacokinetic data-->
<!--Pharmacokinetic data-->
| bioavailability = 5–40%
| bioavailability = 5–40%
| protein_bound = 21%
| protein_bound = 21%
| metabolism = hepatic
| metabolism = [[Liver]]
| elimination_half-life = 4 h (naltrexone),<br />13 h (6β-naltrexol)
| elimination_half-life = Naltrexone: 4 hours<br />[[6β-Naltrexol]]: 13 hours
| excretion = Urine
| excretion = [[Urine]]


<!--Identifiers-->
<!--Identifiers-->
Line 33: Line 35:
| ATC_prefix = N07
| ATC_prefix = N07
| ATC_suffix = BB04
| ATC_suffix = BB04
| ATC_supplemental =
| ATC_supplemental =
| PubChem = 5360515
| PubChem = 5360515
| IUPHAR_ligand = 1639
| IUPHAR_ligand = 1639
Line 48: Line 50:
| ChEMBL_Ref = {{ebicite|changed|EBI}}
| ChEMBL_Ref = {{ebicite|changed|EBI}}
| ChEMBL = 19019
| ChEMBL = 19019
| synonyms = EN-1639A; UM-792; ''N''-Cyclopropyl-methylnoroxymorphone; ''N''-Cyclopropylmethyl-14-hydroxydihydro-morphinone; 17-(Cyclopropylmethyl)-4,5α-epoxy-3,14-dihydroxymorphinan-6-one


<!--Chemical data-->
<!--Chemical data-->
| C=20 | H=23 | N=1 | O=4
| C=20 | H=23 | N=1 | O=4
| molecular_weight = 341.401 g/mol
| molecular_weight = 341.401 g/mol
| smiles = O=C4[C@@H]5Oc1c2c(ccc1O)C[C@H]3N(CC[C@]25[C@@]3(O)CC4)CC6CC6
| SMILES = O=C4[C@@H]5Oc1c2c(ccc1O)C[C@H]3N(CC[C@]25[C@@]3(O)CC4)CC6CC6
| StdInChI_Ref = {{stdinchicite|correct|chemspider}}
| StdInChI_Ref = {{stdinchicite|correct|chemspider}}
| StdInChI = 1S/C20H23NO4/c22-13-4-3-12-9-15-20(24)6-5-14(23)18-19(20,16(12)17(13)25-18)7-8-21(15)10-11-1-2-11/h3-4,11,15,18,22,24H,1-2,5-10H2/t15-,18+,19+,20-/m1/s1
| StdInChI = 1S/C20H23NO4/c22-13-4-3-12-9-15-20(24)6-5-14(23)18-19(20,16(12)17(13)25-18)7-8-21(15)10-11-1-2-11/h3-4,11,15,18,22,24H,1-2,5-10H2/t15-,18+,19+,20-/m1/s1
| StdInChIKey_Ref = {{stdinchicite|correct|chemspider}}
| StdInChIKey_Ref = {{stdinchicite|correct|chemspider}}
| StdInChIKey = DQCKKXVULJGBQN-XFWGSAIBSA-N
| StdInChIKey = DQCKKXVULJGBQN-XFWGSAIBSA-N

<!--Physical data-->
| melting_point = 169
| melting_point = 169
}}
}}
<!-- Definition and medical uses -->
<!-- Definition and medical uses -->
'''Naltrexone''', sold under the brand name '''Revia''' and '''Vivitrol''', is a [[medication]] primarily used to manage [[alcohol dependence]] and [[opioid dependence]].<ref name=AHFS2017/> In opioid dependence, it should not be started until people are detoxified.<ref name=AHFS2017/> It is taken by mouth or by [[intramuscular|injection into a muscle]].<ref name=AHFS2017/> Effects begin within 30 minutes.<ref name=AHFS2017/> A decreased desire for opioids however may take a few weeks.<ref name=AHFS2017/>
'''Naltrexone''', sold under the brand names '''Revia''' and '''Vivitrol''' among others, is a [[medication]] primarily used to manage [[alcohol dependence]] and [[opioid dependence]].<ref name=AHFS2017/> In opioid dependence, it should not be started until people are detoxified.<ref name=AHFS2017/> It is taken by mouth or by [[intramuscular|injection into a muscle]].<ref name=AHFS2017/> Effects begin within 30 minutes.<ref name=AHFS2017/> A decreased desire for opioids however may take a few weeks.<ref name=AHFS2017/>


<!-- Side effects and mechanism -->
<!-- Side effects and mechanism -->
Side effects may include [[trouble sleeping]], anxiety, [[nausea]], and headache.s<ref name=AHFS2017/> In those still on opioids, [[opioid withdrawal]] may occur.<ref name=AHFS2017/> Use is not recommended in people with [[liver failure]].<ref name=AHFS2017/> It is unclear if use is safe during [[pregnancy]].<ref name=AHFS2017/><ref>{{cite journal|last1=Tran|first1=TH|last2=Griffin|first2=BL|last3=Stone|first3=RH|last4=Vest|first4=KM|last5=Todd|first5=TJ|title=Methadone, Buprenorphine, and Naltrexone for the Treatment of Opioid Use Disorder in Pregnant Women.|journal=Pharmacotherapy|date=July 2017|volume=37|issue=7|pages=824-839|doi=10.1002/phar.1958|pmid=28543191}}</ref> Naltrexone is a pure [[opioid antagonist]] and works by blocking the activity of [[opioid]]s.<ref name=AHFS2017>{{cite web|title=Naltrexone Monograph for Professionals - Drugs.com|url=https://www.drugs.com/monograph/naltrexone.html|website=Drugs.com|publisher=American Society of Health-System Pharmacists|accessdate=9 November 2017}}</ref>
Side effects may include [[trouble sleeping]], [[anxiety]], [[nausea]], and [[headache]]s.<ref name=AHFS2017/> In those still on opioids, [[opioid withdrawal]] may occur.<ref name=AHFS2017/> Use is not recommended in people with [[liver failure]].<ref name=AHFS2017/> It is unclear if use is safe during [[pregnancy]].<ref name=AHFS2017/><ref>{{cite journal|last1=Tran|first1=TH|last2=Griffin|first2=BL|last3=Stone|first3=RH|last4=Vest|first4=KM|last5=Todd|first5=TJ|title=Methadone, Buprenorphine, and Naltrexone for the Treatment of Opioid Use Disorder in Pregnant Women.|journal=Pharmacotherapy|date=July 2017|volume=37|issue=7|pages=824-839|doi=10.1002/phar.1958|pmid=28543191}}</ref> Naltrexone is an [[opioid antagonist]] and works by blocking the effects of [[opioid]]s, both those in the body like [[endorphin]]s and those from outside of the body like [[morphine]] and [[heroin]].<ref name=AHFS2017>{{cite web|title=Naltrexone Monograph for Professionals - Drugs.com|url=https://www.drugs.com/monograph/naltrexone.html|website=Drugs.com|publisher=American Society of Health-System Pharmacists|accessdate=9 November 2017}}</ref>


<!-- History and culture -->
<!-- History and culture -->
Naltrexone was first made in 1965 and was approved for medical use in the United States in 1984.<ref name=AHFS2017/><ref>{{cite book|last1=Sadock|first1=Benjamin J.|last2=Sadock|first2=Virginia A.|last3=Sussman|first3=Norman|title=Kaplan & Sadock's Pocket Handbook of Psychiatric Drug Treatment|date=2012|publisher=Lippincott Williams & Wilkins|isbn=9781451154467|page=265|url=https://books.google.ca/books?id=qp-H3UruKvIC&pg=PT265|language=en}}</ref> As of 2017, the wholesale cost of tablets is about US$0.74 [[defined daily dose|per day]] in the US.<ref>{{cite web|url=http://www.whocc.no/atc_ddd_index/?code=N07BB04|title=Naltrexone|website=ATC/DDD Index|publisher=WHO Collaborating Centre for Drug Statistics Methodology|access-date=11 July 2016|quote="DDD ... 50 mg"}}</ref><ref name=Medi2017>{{cite web|title=NADAC as of 2017-11-29|url=https://data.medicaid.gov/Drug-Pricing-and-Payment/NADAC-as-of-2017-11-29/pmhw-skdi|website=Centers for Medicare and Medicaid Services|accessdate=3 December 2017|language=en}}</ref> The extended-release injections cost about $1,267 per month ($41.20 per day).<ref name=Medi2017/> Naltrexone, as [[bupropion/naltrexone]], is also used to treat [[obesity]].<ref>{{cite journal|title=Naltrexone/bupropion for obesity.|journal=Drug and therapeutics bulletin|date=November 2017|volume=55|issue=11|pages=126-129|doi=10.1136/dtb.2017.11.0550|pmid=29117992}}</ref>
Naltrexone was first made in 1965 and was approved for medical use in the United States in 1984.<ref name=AHFS2017/><ref name="SadockVirgina2012">{{cite book|last1=Sadock|first1=Benjamin J.|last2=Sadock|first2=Virginia A.|last3=Sussman|first3=Norman|title=Kaplan & Sadock's Pocket Handbook of Psychiatric Drug Treatment|date=2012|publisher=Lippincott Williams & Wilkins|isbn=9781451154467|page=265|url=https://books.google.ca/books?id=qp-H3UruKvIC&pg=PT265|language=en}}</ref> As of 2017, the wholesale cost of tablets is about US$0.74 [[defined daily dose|per day]] in the US.<ref>{{cite web|url=http://www.whocc.no/atc_ddd_index/?code=N07BB04|title=Naltrexone|website=ATC/DDD Index|publisher=WHO Collaborating Centre for Drug Statistics Methodology|access-date=11 July 2016|quote="DDD ... 50 mg"}}</ref><ref name=Medi2017>{{cite web|title=NADAC as of 2017-11-29|url=https://data.medicaid.gov/Drug-Pricing-and-Payment/NADAC-as-of-2017-11-29/pmhw-skdi|website=Centers for Medicare and Medicaid Services|accessdate=3 December 2017|language=en}}</ref> The extended-release injections cost about $1,267 per month ($41.20 per day).<ref name=Medi2017/> Naltrexone, as [[bupropion/naltrexone]], is also used to treat [[obesity]].<ref>{{cite journal|title=Naltrexone/bupropion for obesity.|journal=Drug and therapeutics bulletin|date=November 2017|volume=55|issue=11|pages=126-129|doi=10.1136/dtb.2017.11.0550|pmid=29117992}}</ref>

{{TOC limit|3}}


==Medical uses==
==Medical uses==
Line 82: Line 89:
A 2011 review found insufficient evidence to determine the effect of naltrexone take by mouth in opioid dependence.<ref>{{cite journal|last1=Minozzi|first1=S|last2=Amato|first2=L|last3=Vecchi|first3=S|last4=Davoli|first4=M|last5=Kirchmayer|first5=U|last6=Verster|first6=A|title=Oral naltrexone maintenance treatment for opioid dependence.|journal=The Cochrane database of systematic reviews|date=13 April 2011|issue=4|pages=CD001333|doi=10.1002/14651858.CD001333.pub4|pmid=21491383}}</ref> While some do well with this formulation, it must be taken daily, and a person whose cravings become overwhelming can obtain opioid intoxication simply by skipping a dose. Due to this issue, the usefulness of oral naltrexone in opioid use disorders is limited by the low retention in treatment. Naltrexone by mouth remains an ideal treatment for a small number of people with opioid use, usually those with a stable social situation and motivation. With additional [[contingency management]] support, naltrexone may be effective in a broader population.<ref>{{cite journal |doi=10.1111/j.1360-0443.2006.01369.x |pmid=16548929 |title=Efficacy of maintenance treatment with naltrexone for opioid dependence: A meta-analytical review |journal=Addiction |volume=101 |issue=4 |pages=491–503 |year=2006 |last1=Johansson |first1=Björn Axel |last2=Berglund |first2=Mats |last3=Lindgren |first3=Anna }}</ref>
A 2011 review found insufficient evidence to determine the effect of naltrexone take by mouth in opioid dependence.<ref>{{cite journal|last1=Minozzi|first1=S|last2=Amato|first2=L|last3=Vecchi|first3=S|last4=Davoli|first4=M|last5=Kirchmayer|first5=U|last6=Verster|first6=A|title=Oral naltrexone maintenance treatment for opioid dependence.|journal=The Cochrane database of systematic reviews|date=13 April 2011|issue=4|pages=CD001333|doi=10.1002/14651858.CD001333.pub4|pmid=21491383}}</ref> While some do well with this formulation, it must be taken daily, and a person whose cravings become overwhelming can obtain opioid intoxication simply by skipping a dose. Due to this issue, the usefulness of oral naltrexone in opioid use disorders is limited by the low retention in treatment. Naltrexone by mouth remains an ideal treatment for a small number of people with opioid use, usually those with a stable social situation and motivation. With additional [[contingency management]] support, naltrexone may be effective in a broader population.<ref>{{cite journal |doi=10.1111/j.1360-0443.2006.01369.x |pmid=16548929 |title=Efficacy of maintenance treatment with naltrexone for opioid dependence: A meta-analytical review |journal=Addiction |volume=101 |issue=4 |pages=491–503 |year=2006 |last1=Johansson |first1=Björn Axel |last2=Berglund |first2=Mats |last3=Lindgren |first3=Anna }}</ref>


===Other===
===Others===
It is not useful for quitting smoking.<ref>{{cite journal|first1=Sean P.|last1=David|first2=Tim|last2=Lancaster|first3=Lindsay F.|last3=Stead|first4=A. Eden|last4=Evins|title=Opioid antagonists for smoking cessation|journal=The Cochrane Database of Systematic Reviews|date=6 June 2013|issn=1469-493X|pages=CD003086|issue=6|pmid=23744347|pmc=4038652|doi=10.1002/14651858.CD003086.pub3|first5=Judith J.|last5=Prochaska}}</ref>
Naltrexone is not useful for [[smoking cessation|quitting smoking]].<ref>{{cite journal|first1=Sean P.|last1=David|first2=Tim|last2=Lancaster|first3=Lindsay F.|last3=Stead|first4=A. Eden|last4=Evins|title=Opioid antagonists for smoking cessation|journal=The Cochrane Database of Systematic Reviews|date=6 June 2013|issn=1469-493X|pages=CD003086|issue=6|pmid=23744347|pmc=4038652|doi=10.1002/14651858.CD003086.pub3|first5=Judith J.|last5=Prochaska}}</ref>


==Adverse effects==
===Available forms===
Vivitrol, a naltrexone formulation for [[Injection (medicine)#Depot injection|depot injection]], was approved by the FDA on April 13, 2006, for the treatment of alcohol dependence.<ref name="MedicalNewsToday2006">"[http://www.medicalnewstoday.com/articles/41707.php Alcoholism Once A Month Injectable Drug, Vivitrol, Approved By FDA]," Medical News Today, April 16, 2006.</ref>
The most common side effects reported with naltrexone are [[gastrointestinal]] complaints such as [[diarrhea]] and abdominal cramping. These adverse effects are analogous to the symptoms of [[opioid withdrawal]], as the mu receptor blockade will increase GI motility.


Additionally, naltrexone implants that are surgically implanted are available,<ref name=TGA>{{cite web| url = https://www.ebs.tga.gov.au/ebs/ANZTPAR/PublicWeb.nsf/cuMedicines?OpenView | title = Australian Register of Therapeutic Goods Medicines | accessdate = 2009-03-22 | author = Therapeutic Goods Administration| authorlink = | format = Online database of approved medicines }}</ref> While these are manufactured in Australia, they are not authorized for use within Australia, but only for export.<ref>{{cite web| url = https://www.ebs.tga.gov.au/servlet/xmlmillr6?dbid=ebs%2FPublicHTML%2FpdfStore.nsf&docid=12AA89E8C8E53B46CA257FA7004211EE&agid=(PrintDetailsPublic)&actionid=1 | title = Australian Register of Therapeutic Goods Medicines | accessdate = 2017-04-27 | author = Therapeutic Goods Administration| authorlink = | format = Online database of approved medicines, specific entry for "O'Neil Long Acting Naltrexone Implant" }}</ref> By 2009, naltrexone implants showed encouraging results.<ref>{{cite journal |doi=10.1001/archgenpsychiatry.2009.130 |pmid=19805701 |title=Improving Clinical Outcomes in Treating Heroin Dependence |journal=Archives of General Psychiatry |volume=66 |issue=10 |pages=1108–15 |year=2009 |last1=Hulse |first1=Gary K. |last2=Morris |first2=Noella |last3=Arnold-Reed |first3=Diane |last4=Tait |first4=Robert J. }}</ref>
Naltrexone has been reported to cause liver damage (when given at doses higher than recommended). It carries an FDA boxed warning for this rare side effect. Due to these reports, some physicians may check liver function tests prior to starting naltrexone, and periodically thereafter. Concerns for liver toxicity initially arose from a study of non-addicted obese patients receiving 300&nbsp;mg of naltrexone.<ref>{{cite journal |pmid=3092099 |url=https://archives.drugabuse.gov/pdf/monographs/download67.html |year=1986 |author1=Pfohl |first1=David N. |title=Naltrexone hydrochloride (Trexan): A review of serum transaminase elevations at high dosage |journal=NIDA research monograph |volume=67 |pages=66–72 |last2=Allen |first2=John I. |last3=Atkinson |first3=Richard L. |last4=Knopman |first4=David S. |last5=Malcolm |first5=Robert J. |last6=Mitchell |first6=James E. |last7=Morley |first7=John E. }}</ref> Subsequent studies have suggested limited toxicity in other patient populations.

Naltrexone should not be started until several (typically 7-10) days of abstinence from opioids has been achieved. This is due to the risk of acute opioid withdrawal if naltrexone is taken, as naltrexone will displace most opioids from their receptors. The time of abstinence may be shorter than 7 days, depending on the half-life of the specific opioid taken. Some physicians use a naloxone challenge to determine whether an individual has any opioids remaining. The challenge involves giving a test dose of naloxone and monitoring for opioid withdrawal. If withdrawal occurs, naltrexone should not be started.<ref name="galanter">Galanter, Marc; Kleber, Herbert. The American Psychiatric Publishing Textbook of Substance Abuse Treatment, 4th Edition. {{ISBN|1585622761}}{{page needed|date=January 2017}}</ref>


==Contraindications==
==Contraindications==
Naltrexone should not be used by persons with acute hepatitis or liver failure, or those with recent opioid use (typically 7–10 days).
Naltrexone should not be used by persons with acute hepatitis or liver failure, or those with recent opioid use (typically 7–10 days).


==Side effects==
==Pharmacogenetics==
The most common side effects reported with naltrexone are [[gastrointestinal]] complaints such as [[diarrhea]] and abdominal cramping. These adverse effects are analogous to the symptoms of [[opioid withdrawal]], as the mu receptor blockade will increase GI motility.

Naltrexone has been reported to cause liver damage (when given at doses higher than recommended). It carries an FDA boxed warning for this rare side effect. Due to these reports, some physicians may check liver function tests prior to starting naltrexone, and periodically thereafter. Concerns for liver toxicity initially arose from a study of non-addicted obese patients receiving 300&nbsp;mg of naltrexone.<ref>{{cite journal |pmid=3092099 |url=https://archives.drugabuse.gov/pdf/monographs/download67.html |year=1986 |author1=Pfohl |first1=David N. |title=Naltrexone hydrochloride (Trexan): A review of serum transaminase elevations at high dosage |journal=NIDA research monograph |volume=67 |pages=66–72 |last2=Allen |first2=John I. |last3=Atkinson |first3=Richard L. |last4=Knopman |first4=David S. |last5=Malcolm |first5=Robert J. |last6=Mitchell |first6=James E. |last7=Morley |first7=John E. }}</ref> Subsequent studies have suggested limited toxicity in other patient populations.

Naltrexone should not be started until several (typically 7-10) days of abstinence from opioids has been achieved. This is due to the risk of acute opioid withdrawal if naltrexone is taken, as naltrexone will displace most opioids from their receptors. The time of abstinence may be shorter than 7 days, depending on the half-life of the specific opioid taken. Some physicians use a naloxone challenge to determine whether an individual has any opioids remaining. The challenge involves giving a test dose of naloxone and monitoring for opioid withdrawal. If withdrawal occurs, naltrexone should not be started.<ref name="galanter">Galanter, Marc; Kleber, Herbert. The American Psychiatric Publishing Textbook of Substance Abuse Treatment, 4th Edition. {{ISBN|1585622761}}{{page needed|date=January 2017}}</ref>

==Pharmacology==

===Pharmacodynamics===
Naltrexone and its [[active metabolite]] [[6β-naltrexol]] are [[competitive antagonist]]s at the [[μ-opioid receptor]] (MOR), the [[κ-opioid receptor]] (KOR) to a lesser extent, and to a far lesser and possibly insignificant extent, at the [[δ-opioid receptor]] (DOR).<ref>{{cite journal |doi=10.1007/s40263-013-0096-4 |pmid=23881605 |pmc=4600601 |title=Targeted Opioid Receptor Antagonists in the Treatment of Alcohol Use Disorders |journal=CNS Drugs |volume=27 |issue=10 |pages=777–87 |year=2013 |last1=Niciu |first1=Mark J. |last2=Arias |first2=Albert J. }}</ref> The K<sub>i</sub> [[affinity (pharmacology)|affinity]] values of naltrexone at the MOR, KOR, and DOR have been reported as 0.0825 nM, 0.509 nM, and 8.02 nM, respectively, demonstrating a MOR/KOR binding ratio of 6.17 and a MOR/DOR binding ratio of 97.2.<ref name="pmid7562497">{{cite journal |pmid=7562497 |url=http://jpet.aspetjournals.org/content/274/3/1263.short |year=1995 |author1=Codd |first1=E. E. |title=Serotonin and norepinephrine uptake inhibiting activity of centrally acting analgesics: Structural determinants and role in antinociception |journal=The Journal of Pharmacology and Experimental Therapeutics |volume=274 |issue=3 |pages=1263–70 |last2=Shank |first2=R. P. |last3=Schupsky |first3=J. J. |last4=Raffa |first4=R. B. }}</ref>

====Mechanism of action====
The blockade of [[opioid receptor]]s is the basis behind naltrexone's action in the management of opioid dependence&mdash;it reversibly blocks or attenuates the effects of opioids. Its mechanism of action in alcohol dependence is not fully understood, but as an opioid receptor antagonist is likely to be due to the modulation of the [[dopaminergic]] [[mesolimbic pathway]] (one of the primary centers for risk-reward analysis in the brain, and a tertiary "pleasure center") which is hypothesized to be a major center of the reward associated with addiction that all major drugs of abuse are believed to activate. {{Citation needed|date=December 2009}} Mechanism of action may be antagonism to endogenous opioids such as [[tetrahydropapaveroline]], whose production is augmented in the presence of alcohol.<ref name="HaberRoske1996">{{cite journal |doi=10.1016/S0024-3205(96)00597-8 |pmid=9000113 |title=Alcohol induces formation of morphine precursors in the striatum of rats |journal=Life Sciences |volume=60 |issue=2 |pages=79–89 |year=1996 |last1=Haber |first1=Hanka |last2=Roske |first2=Irmgard |last3=Rottmann |first3=Matthias |last4=Georgi |first4=Monika |last5=Melzig |first5=Matthias F. }}</ref>

===Pharmacokinetics===
Naltrexone is [[metabolism|metabolized]] in the [[liver]] mainly to [[6β-naltrexol]] by the [[enzyme]] [[dihydrodiol dehydrogenase]]. Other [[metabolite]]s include 2-hydroxy-3-methoxy-6β-naltrexol and 2-hydroxy-3-methoxy-naltrexone. These are then further metabolized by [[conjugation (biochemistry)|conjugation]] with [[glucuronide]].{{Citation needed|date=June 2017}} The plasma [[biological half-life|half-life]] of naltrexone and its metabolite 6β-naltrexol are about 4 hours and 13 hours, respectively.{{Citation needed|date=June 2017}}

===Pharmacogenetics===
A naltrexone treatment study by Anton et al., released by the [[National Institutes of Health]] in February 2008 and published in the ''[[Archives of General Psychiatry]],'' has shown that alcoholics having a certain variant of the opioid receptor gene (G polymorphism of [[Single-nucleotide polymorphism|SNP]] Rs1799971 in the gene [[OPRM1]]), known as Asp40, demonstrated strong response to naltrexone and were far more likely to experience success at cutting back or discontinuing their alcohol intake altogether, while for those lacking the gene variant, naltrexone appeared to be no different from placebo.<ref name="ctidNCT00006206">{{cite journal |doi=10.1001/archpsyc.65.2.135 |pmid=18250251 |pmc=2666924 |title=An Evaluation of μ-Opioid Receptor (OPRM1) as a Predictor of Naltrexone Response in the Treatment of Alcohol Dependence |journal=Archives of General Psychiatry |volume=65 |issue=2 |pages=135–44 |year=2008 |last1=Anton |first1=Raymond F. |last2=Oroszi |first2=Gabor |last3=o'Malley |first3=Stephanie |last4=Couper |first4=David |last5=Swift |first5=Robert |last6=Pettinati |first6=Helen |last7=Goldman |first7=David }}</ref> The G allele of OPRM1 is most common in individuals of Asian descent, with 60% to 70% of people of Chinese, Japanese, and Indian ancestry having at least one copy, as opposed to 30% of Europeans and few Africans.<ref>{{cite web|url=http://www.snpedia.com/index.php/Rs1799971|title=Rs1799971 (SNPedia)}}</ref>
A naltrexone treatment study by Anton et al., released by the [[National Institutes of Health]] in February 2008 and published in the ''[[Archives of General Psychiatry]],'' has shown that alcoholics having a certain variant of the opioid receptor gene (G polymorphism of [[Single-nucleotide polymorphism|SNP]] Rs1799971 in the gene [[OPRM1]]), known as Asp40, demonstrated strong response to naltrexone and were far more likely to experience success at cutting back or discontinuing their alcohol intake altogether, while for those lacking the gene variant, naltrexone appeared to be no different from placebo.<ref name="ctidNCT00006206">{{cite journal |doi=10.1001/archpsyc.65.2.135 |pmid=18250251 |pmc=2666924 |title=An Evaluation of μ-Opioid Receptor (OPRM1) as a Predictor of Naltrexone Response in the Treatment of Alcohol Dependence |journal=Archives of General Psychiatry |volume=65 |issue=2 |pages=135–44 |year=2008 |last1=Anton |first1=Raymond F. |last2=Oroszi |first2=Gabor |last3=o'Malley |first3=Stephanie |last4=Couper |first4=David |last5=Swift |first5=Robert |last6=Pettinati |first6=Helen |last7=Goldman |first7=David }}</ref> The G allele of OPRM1 is most common in individuals of Asian descent, with 60% to 70% of people of Chinese, Japanese, and Indian ancestry having at least one copy, as opposed to 30% of Europeans and few Africans.<ref>{{cite web|url=http://www.snpedia.com/index.php/Rs1799971|title=Rs1799971 (SNPedia)}}</ref>


Line 105: Line 128:
Studies have found naltrexone to be more efficacious among certain white subjects, because of the genetic basis, than among black subjects, who generally do not carry the relevant gene variant.<ref>{{cite journal |doi=10.1111/j.1530-0277.2001.tb02356.x |pmid=11584154 |title=Efficacy of Naltrexone and Acamprosate for Alcoholism Treatment: A Meta-Analysis |journal=Alcoholism: Clinical and Experimental Research |volume=25 |issue=9 |pages=1335–41 |year=2001 |last1=Kranzler |first1=Henry R. |last2=Kirk |first2=Jeffrey }}</ref> A 2009 study of naltrexone as an alcohol dependence treatment among African Americans failed to find any statistically significant differences between naltrexone and a placebo.<ref>{{cite journal |doi=10.1016/j.drugalcdep.2009.07.006 |pmid=19717248 |pmc=3409877 |title=Naltrexone for the treatment of alcohol dependence among African Americans: Results from the COMBINE Study |journal=Drug and Alcohol Dependence |volume=105 |issue=3 |pages=256–8 |year=2009 |last1=Ray |first1=Lara A. |last2=Oslin |first2=David W. }}</ref> Studies have suggested that carriers of the G allele may experience higher levels of craving and stronger "high" upon alcohol consumption, compared to carriers of the dominant allele, and naltrexone somewhat blunts these responses, leading to a reduction in alcohol use in some studies.<ref>{{cite journal |doi=10.1111/j.1530-0277.2011.01633.x |pmid=21895723 |pmc=3249007 |title=The Role of the Asn40Asp Polymorphism of the Mu Opioid Receptor Gene (OPRM1) on Alcoholism Etiology and Treatment: A Critical Review |journal=Alcoholism: Clinical and Experimental Research |volume=36 |issue=3 |pages=385–94 |year=2012 |last1=Ray |first1=Lara A. |last2=Barr |first2=Christina S. |last3=Blendy |first3=Julie A. |last4=Oslin |first4=David |last5=Goldman |first5=David |last6=Anton |first6=Raymond F. }}</ref>
Studies have found naltrexone to be more efficacious among certain white subjects, because of the genetic basis, than among black subjects, who generally do not carry the relevant gene variant.<ref>{{cite journal |doi=10.1111/j.1530-0277.2001.tb02356.x |pmid=11584154 |title=Efficacy of Naltrexone and Acamprosate for Alcoholism Treatment: A Meta-Analysis |journal=Alcoholism: Clinical and Experimental Research |volume=25 |issue=9 |pages=1335–41 |year=2001 |last1=Kranzler |first1=Henry R. |last2=Kirk |first2=Jeffrey }}</ref> A 2009 study of naltrexone as an alcohol dependence treatment among African Americans failed to find any statistically significant differences between naltrexone and a placebo.<ref>{{cite journal |doi=10.1016/j.drugalcdep.2009.07.006 |pmid=19717248 |pmc=3409877 |title=Naltrexone for the treatment of alcohol dependence among African Americans: Results from the COMBINE Study |journal=Drug and Alcohol Dependence |volume=105 |issue=3 |pages=256–8 |year=2009 |last1=Ray |first1=Lara A. |last2=Oslin |first2=David W. }}</ref> Studies have suggested that carriers of the G allele may experience higher levels of craving and stronger "high" upon alcohol consumption, compared to carriers of the dominant allele, and naltrexone somewhat blunts these responses, leading to a reduction in alcohol use in some studies.<ref>{{cite journal |doi=10.1111/j.1530-0277.2011.01633.x |pmid=21895723 |pmc=3249007 |title=The Role of the Asn40Asp Polymorphism of the Mu Opioid Receptor Gene (OPRM1) on Alcoholism Etiology and Treatment: A Critical Review |journal=Alcoholism: Clinical and Experimental Research |volume=36 |issue=3 |pages=385–94 |year=2012 |last1=Ray |first1=Lara A. |last2=Barr |first2=Christina S. |last3=Blendy |first3=Julie A. |last4=Oslin |first4=David |last5=Goldman |first5=David |last6=Anton |first6=Raymond F. }}</ref>


==Chemistry==
==Mechanism of action==
Naltrexone can be described as a substituted [[oxymorphone]] &ndash; here the [[tertiary amine]] [[methyl]]-substituent is replaced with [[methylcyclopropane]]. Naltrexone is the N-cyclopropylmethyl derivative of oxymorphone.{{Citation needed|date=June 2017}}
Naltrexone and its [[active metabolite]] [[6β-naltrexol]] are [[competitive antagonist]]s at the [[μ-opioid receptor]] (MOR), the [[κ-opioid receptor]] (KOR) to a lesser extent, and to a far lesser and possibly insignificant extent, at the [[δ-opioid receptor]] (DOR).<ref>{{cite journal |doi=10.1007/s40263-013-0096-4 |pmid=23881605 |pmc=4600601 |title=Targeted Opioid Receptor Antagonists in the Treatment of Alcohol Use Disorders |journal=CNS Drugs |volume=27 |issue=10 |pages=777–87 |year=2013 |last1=Niciu |first1=Mark J. |last2=Arias |first2=Albert J. }}</ref> The K<sub>i</sub> [[affinity (pharmacology)|affinity]] values of naltrexone at the MOR, KOR, and DOR have been reported as 0.0825 nM, 0.509 nM, and 8.02 nM, respectively, demonstrating a MOR/KOR binding ratio of 6.17 and a MOR/DOR binding ratio of 97.2.<ref name="pmid7562497">{{cite journal |pmid=7562497 |url=http://jpet.aspetjournals.org/content/274/3/1263.short |year=1995 |author1=Codd |first1=E. E. |title=Serotonin and norepinephrine uptake inhibiting activity of centrally acting analgesics: Structural determinants and role in antinociception |journal=The Journal of Pharmacology and Experimental Therapeutics |volume=274 |issue=3 |pages=1263–70 |last2=Shank |first2=R. P. |last3=Schupsky |first3=J. J. |last4=Raffa |first4=R. B. }}</ref>


===Analogues===
The blockade of [[opioid receptor]]s is the basis behind naltrexone's action in the management of opioid dependence&mdash;it reversibly blocks or attenuates the effects of opioids. Its mechanism of action in alcohol dependence is not fully understood, but as an opioid receptor antagonist is likely to be due to the modulation of the [[dopaminergic]] [[mesolimbic pathway]] (one of the primary centers for risk-reward analysis in the brain, and a tertiary "pleasure center") which is hypothesized to be a major center of the reward associated with addiction that all major drugs of abuse are believed to activate. {{Citation needed|date=December 2009}} Mechanism of action may be antagonism to endogenous opioids such as [[tetrahydropapaveroline]], whose production is augmented in the presence of alcohol.<ref name="HaberRoske1996">{{cite journal |doi=10.1016/S0024-3205(96)00597-8 |pmid=9000113 |title=Alcohol induces formation of morphine precursors in the striatum of rats |journal=Life Sciences |volume=60 |issue=2 |pages=79–89 |year=1996 |last1=Haber |first1=Hanka |last2=Roske |first2=Irmgard |last3=Rottmann |first3=Matthias |last4=Georgi |first4=Monika |last5=Melzig |first5=Matthias F. }}</ref>
The closely related medication, [[methylnaltrexone]], is used to treat opioid-induced constipation but does not treat addiction as it does not cross the [[blood–brain barrier]]. [[Nalmefene]] is similar to naltrexone and is used for the same purposes as naltrexone. Naltrexone should not be confused with [[naloxone]], which is used in emergency cases of opioid [[overdose]]. Other related opioid antagonists include [[nalodeine]] and [[samidorphan]].


==History==
==Structure and pharmacokinetics==
Naltrexone was first [[chemical synthesis|synthesized]] in 1965 by Blumberg and Dayton and was found to be an [[oral administration|orally active]] and long-acting opioid antagonist.<ref name="PadwaCunningham2010">{{cite book|author1=Howard Padwa|author2=Jacob Cunningham|title=Addiction: A Reference Encyclopedia|url=https://books.google.com/books?id=hdMAnL5neAAC&pg=PA207|year=2010|publisher=ABC-CLIO|isbn=978-1-59884-229-6|pages=207–}}</ref><ref name="Bennett2004">{{cite book|author=G Bennett|title=Treating Drug Abusers|url=https://books.google.com/books?id=ft6IAgAAQBAJ&pg=PT112|date=14 January 2004|publisher=Routledge|isbn=978-1-134-93173-6|pages=112–}}</ref><ref name="SadockVirgina2012" /> It showed advantages over earlier opioid antagonists like [[cyclazocine]], [[nalorphine]], and naloxone including its oral activity, a long [[duration of action]] allowing for once-daily administration, and a lack of [[dysphoria]].<ref name="SadockVirgina2012" /> The drug was approved by the FDA for the oral treatment of heroin addiction in 1984 and for the oral treatment of alcohol dependence in 1995.<ref name="Milhorn2017">{{cite book|author=H. Thomas Milhorn|title=Substance Use Disorders: A Guide for the Primary Care Provider|url=https://books.google.com/books?id=wH86DwAAQBAJ&pg=PA88|date=17 October 2017|publisher=Springer International Publishing|isbn=978-3-319-63040-3|pages=88–}}</ref> A depot formulation for intramuscular injection was approved by the FDA for alcohol dependence in 2006.<ref name="MedicalNewsToday2006" />
Naltrexone can be described as a substituted [[oxymorphone]] &ndash; here the [[tertiary amine]] [[methyl]]-substituent is replaced with [[methylcyclopropane]]. Naltrexone is the N-cyclopropylmethyl derivative of oxymorphone.{{Citation needed|date=June 2017}}


==Society and culture==
Naltrexone is metabolized mainly to [[6β-naltrexol]] by the liver enzyme [[dihydrodiol dehydrogenase]]. Other metabolites include 2-hydroxy-3-methoxy-6β-naltrexol and 2-hydroxy-3-methoxy-naltrexone. These are then further metabolized by conjugation with glucuronide.{{Citation needed|date=June 2017}}


===Generic names===
The plasma [[Biological half-life|half-life]] of naltrexone and its metabolite 6β-naltrexol are about 4 hours and 13 hours, respectively.{{Citation needed|date=June 2017}}
''Naltrexone'' is the [[generic term|generic name]] of the drug and its {{abbrlink|INN|International Nonproprietary Name}}, {{abbrlink|BAN|British Approved Name}}, {{abbrlink|DCF|Dénomination Commune Française}}, and {{abbrlink|DCIT|Denominazione Comune Italiana}}, while ''naltrexone hydrochloride'' is its {{abbrlink|USP|United States Pharmacopeia}} and {{abbrlink|BANM|British Approved Name}}.<ref name="Elks2014" /><ref name="IndexNominum2000" /><ref name="MortonHall2012" /><ref name="Drugs.com" />


==Formulations==
===Brand names===
Naltrexone is marketed under a variety of brand names including Adepend, Antaxone, Celupan, Nalorex, Narcoral, Nemexin, Revia, ReVia, Trexan, and Vivitrol among others.<ref name="Elks2014">{{cite book|author=J. Elks|title=The Dictionary of Drugs: Chemical Data: Chemical Data, Structures and Bibliographies|url=https://books.google.com/books?id=0vXTBwAAQBAJ&pg=PA851|date=14 November 2014|publisher=Springer|isbn=978-1-4757-2085-3|pages=851–}}</ref><ref name="IndexNominum2000">{{cite book|title=Index Nominum 2000: International Drug Directory|url=https://books.google.com/books?id=5GpcTQD_L2oC&pg=PA715|year=2000|publisher=Taylor & Francis|isbn=978-3-88763-075-1|pages=715–}}</ref><ref name="MortonHall2012">{{cite book|author1=I.K. Morton|author2=Judith M. Hall|title=Concise Dictionary of Pharmacological Agents: Properties and Synonyms|url=https://books.google.com/books?id=tsjrCAAAQBAJ&pg=PA189|date=6 December 2012|publisher=Springer Science & Business Media|isbn=978-94-011-4439-1|pages=189–}}</ref><ref name="Drugs.com">https://www.drugs.com/international/naltrexone.html</ref> It is also marketed in combination with [[bupropion]] ([[bupropion/naltrexone]]) as Contrave and was marketed with [[morphine]] ([[morphine/naltrexone]]) as Embeda.<ref name="Drugs.com" /> A combination of naltrexone with [[buprenorphine]] ([[buprenorphine/naltrexone]]) has been developed but has not been marketed.<ref name="pmid18212797">{{cite journal | vauthors = McCann DJ | title = Potential of buprenorphine/naltrexone in treating polydrug addiction and co-occurring psychiatric disorders | journal = Clin. Pharmacol. Ther. | volume = 83 | issue = 4 | pages = 627–30 | year = 2008 | pmid = 18212797 | doi = 10.1038/sj.clpt.6100503 | url = }}</ref>
Vivitrol, a naltrexone formulation for [[Injection (medicine)#Depot injection|depot injection]], was approved by the FDA on April 13, 2006, for the treatment of alcohol dependence.<ref>"[http://www.medicalnewstoday.com/articles/41707.php Alcoholism Once A Month Injectable Drug, Vivitrol, Approved By FDA]," Medical News Today, April 16, 2006.</ref>


===Controversies===
Additionally, naltrexone implants that are surgically implanted are available,<ref name=TGA>{{cite web| url = https://www.ebs.tga.gov.au/ebs/ANZTPAR/PublicWeb.nsf/cuMedicines?OpenView | title = Australian Register of Therapeutic Goods Medicines | accessdate = 2009-03-22 | author = Therapeutic Goods Administration| authorlink = | format = Online database of approved medicines }}</ref> While these are manufactured in Australia, they are not authorized for use within Australia, but only for export.<ref>{{cite web| url = https://www.ebs.tga.gov.au/servlet/xmlmillr6?dbid=ebs%2FPublicHTML%2FpdfStore.nsf&docid=12AA89E8C8E53B46CA257FA7004211EE&agid=(PrintDetailsPublic)&actionid=1 | title = Australian Register of Therapeutic Goods Medicines | accessdate = 2017-04-27 | author = Therapeutic Goods Administration| authorlink = | format = Online database of approved medicines, specific entry for "O'Neil Long Acting Naltrexone Implant" }}</ref> By 2009, naltrexone implants showed encouraging results.<ref>{{cite journal |doi=10.1001/archgenpsychiatry.2009.130 |pmid=19805701 |title=Improving Clinical Outcomes in Treating Heroin Dependence |journal=Archives of General Psychiatry |volume=66 |issue=10 |pages=1108–15 |year=2009 |last1=Hulse |first1=Gary K. |last2=Morris |first2=Noella |last3=Arnold-Reed |first3=Diane |last4=Tait |first4=Robert J. }}</ref>


====2011 Krupitsky study====
==Similar medication==
The closely related medication, [[methylnaltrexone]], is used to treat opioid-induced constipation but does not treat addiction as it does not cross the [[blood brain barrier]]. [[Nalmefene]] is similar to naltrexone and is used for the same purposes as naltrexone. Naltrexone should not be confused with [[naloxone]], which is used in emergency cases of opioid [[overdose]].

==Controversies==
=== 2011 Krupitsky study===
The FDA authorized use of injectable naltrexone for opioid addiction using a single study<ref>{{cite web|url=http://www.psmag.com/health-and-behavior/vivitrol-help-control-addictions-57261|title=A Shot in the Dark: Can Vivitrol Help Us Control Our Addictions?|date=7 May 2013|publisher=}}</ref> that was led by Evgeny Krupitsky at Bekhterev Research Psychoneurological Institute, St Petersburg State Pavlov Medical University, St Petersburg, Russia,<ref name="Lancet_Krupitsky_2011_naltrexone">{{cite journal |title=Injectable extended-release naltrexone for opioid dependence: a double-blind, placebo-controlled, multicentre randomised trial |first1=Evgeny |last1=Krupitsky |first2=Edward V. |last2=Nunes |first3=Walter |last3=Ling |first4=Ari |last4=Illeperuma |first5=David R. |last5=Gastfriend |first6=Bernard L. |last6=Silverman |url=https://www.researchgate.net/profile/Evgeny_Krupitsky/publication/51086253_Krupitsky_E_Nunes_EV_Ling_W_Illeperuma_A_Gastfriend_DR_Silverman_BL_Injectable_extended-release_naltrexone_for_opioid_dependence_a_double-blind_placebo-controlled_multicentre_randomised_trial_Lancet_3/links/0deec521d0385a2aff000000/Krupitsky-E-Nunes-EV-Ling-W-Illeperuma-A-Gastfriend-DR-Silverman-BL-Injectable-extended-release-naltrexone-for-opioid-dependence-a-double-blind-placebo-controlled-multicentre-randomised-trial-L.pdf |format=PDF |journal=The Lancet|date=April 28, 2011 |volume=377 |pages=1506–13 |access-date=June 11, 2017 |doi=10.1016/S0140}}</ref> a country where opioid agonists such as methadone and buprenorphine are not available. The study was a "double-blind, placebo-controlled, randomized", 24-week trial running "from July 3, 2008 through October 5, 2009" with "250 patients with opioid dependence disorder" at "13 clinical sites in Russia" on the use of injectable naltrexone (XR-NTX) for opioid dependence. The study was funded by the Boston-based biotech [[Alkermes (company)|Alkermes]] firm which produces and markets naltrexone in the United States.
The FDA authorized use of injectable naltrexone for opioid addiction using a single study<ref>{{cite web|url=http://www.psmag.com/health-and-behavior/vivitrol-help-control-addictions-57261|title=A Shot in the Dark: Can Vivitrol Help Us Control Our Addictions?|date=7 May 2013|publisher=}}</ref> that was led by Evgeny Krupitsky at Bekhterev Research Psychoneurological Institute, St Petersburg State Pavlov Medical University, St Petersburg, Russia,<ref name="Lancet_Krupitsky_2011_naltrexone">{{cite journal |title=Injectable extended-release naltrexone for opioid dependence: a double-blind, placebo-controlled, multicentre randomised trial |first1=Evgeny |last1=Krupitsky |first2=Edward V. |last2=Nunes |first3=Walter |last3=Ling |first4=Ari |last4=Illeperuma |first5=David R. |last5=Gastfriend |first6=Bernard L. |last6=Silverman |url=https://www.researchgate.net/profile/Evgeny_Krupitsky/publication/51086253_Krupitsky_E_Nunes_EV_Ling_W_Illeperuma_A_Gastfriend_DR_Silverman_BL_Injectable_extended-release_naltrexone_for_opioid_dependence_a_double-blind_placebo-controlled_multicentre_randomised_trial_Lancet_3/links/0deec521d0385a2aff000000/Krupitsky-E-Nunes-EV-Ling-W-Illeperuma-A-Gastfriend-DR-Silverman-BL-Injectable-extended-release-naltrexone-for-opioid-dependence-a-double-blind-placebo-controlled-multicentre-randomised-trial-L.pdf |format=PDF |journal=The Lancet|date=April 28, 2011 |volume=377 |pages=1506–13 |access-date=June 11, 2017 |doi=10.1016/S0140}}</ref> a country where opioid agonists such as methadone and buprenorphine are not available. The study was a "double-blind, placebo-controlled, randomized", 24-week trial running "from July 3, 2008 through October 5, 2009" with "250 patients with opioid dependence disorder" at "13 clinical sites in Russia" on the use of injectable naltrexone (XR-NTX) for opioid dependence. The study was funded by the Boston-based biotech [[Alkermes (company)|Alkermes]] firm which produces and markets naltrexone in the United States.


A 2011 article reported that this single trial of naltrexone was performed not by comparing it to the best available, evidence-based treatment (methadone or buprenorphine) but by comparing it with a placebo.<ref>{{cite journal |doi=10.1016/S0140-6736(11)61333-0 |title=Injectable extended-release naltrexone for opioid dependence – Authors' reply |journal=The Lancet |volume=378 |issue=9792 |pages=666 |year=2011 |last1=Wolfe |first1=Daniel |last2=Carrieri |first2=MP |last3=Dasgupta |first3=Nabarun |last4=Bruce |first4=Douglas |last5=Wodak |first5=Alex }}</ref> A subsequent RCT in Norway did compare injectable naltrexone to buprenorphine and found it to be similar in outcomes.<ref>{{cite journal|last1=Tanum|first1=L|last2=Solli|first2=KK|last3=Latif|first3=ZE|last4=Benth|first4=JŠ|last5=Opheim|first5=A|last6=Sharma-Haase|first6=K|last7=Krajci|first7=P|last8=Kunøe|first8=N|title=The Effectiveness of Injectable Extended-Release Naltrexone vs Daily Buprenorphine-Naloxone for Opioid Dependence: A Randomized Clinical Noninferiority Trial.|journal=JAMA psychiatry|date=18 October 2017|doi=10.1001/jamapsychiatry.2017.3206|pmid=29049469}}</ref>
A 2011 article reported that this single trial of naltrexone was performed not by comparing it to the best available, evidence-based treatment (methadone or buprenorphine) but by comparing it with a placebo.<ref>{{cite journal |doi=10.1016/S0140-6736(11)61333-0 |title=Injectable extended-release naltrexone for opioid dependence – Authors' reply |journal=The Lancet |volume=378 |issue=9792 |pages=666 |year=2011 |last1=Wolfe |first1=Daniel |last2=Carrieri |first2=MP |last3=Dasgupta |first3=Nabarun |last4=Bruce |first4=Douglas |last5=Wodak |first5=Alex }}</ref> A subsequent RCT in Norway did compare injectable naltrexone to buprenorphine and found it to be similar in outcomes.<ref>{{cite journal|last1=Tanum|first1=L|last2=Solli|first2=KK|last3=Latif|first3=ZE|last4=Benth|first4=JŠ|last5=Opheim|first5=A|last6=Sharma-Haase|first6=K|last7=Krajci|first7=P|last8=Kunøe|first8=N|title=The Effectiveness of Injectable Extended-Release Naltrexone vs Daily Buprenorphine-Naloxone for Opioid Dependence: A Randomized Clinical Noninferiority Trial.|journal=JAMA psychiatry|date=18 October 2017|doi=10.1001/jamapsychiatry.2017.3206|pmid=29049469}}</ref>


===Tom Price's remarks 2017===
====Tom Price's remarks 2017====
In May 2017, [[United States Secretary of Health and Human Services]] [[Tom Price (American politician)|Tom Price]], praised [vivitrol] as the future of opioid addiction treatment after visiting the company’s plant in Ohio.<ref name="NYT_2017_Vivitrol"/> His remarks set off sharp criticism with almost 700 experts in the field of substance abuse submitting a letter to Price cautioning him about Vivitrol's "marketing tactics" and warning him that his comment "ignore widely accepted science".<ref name="Tom-Price-Letter-Re-MAT_2017">{{cite web |url=https://www.documentcloud.org/documents/3723472-Tom-Price-Letter-Re-MAT.html |title=Letter to Tom Price |date=May 2017 |access-date=June 11, 2017}}</ref> The experts pointed out that Vivitrol's competitors, [[buprenorphine]] and [[methadone]], are "less expensive", "more widely used" and have been "rigorously studied".
In May 2017, [[United States Secretary of Health and Human Services]] [[Tom Price (American politician)|Tom Price]], praised [vivitrol] as the future of opioid addiction treatment after visiting the company’s plant in Ohio.<ref name="NYT_2017_Vivitrol"/> His remarks set off sharp criticism with almost 700 experts in the field of substance abuse submitting a letter to Price cautioning him about Vivitrol's "marketing tactics" and warning him that his comment "ignore widely accepted science".<ref name="Tom-Price-Letter-Re-MAT_2017">{{cite web |url=https://www.documentcloud.org/documents/3723472-Tom-Price-Letter-Re-MAT.html |title=Letter to Tom Price |date=May 2017 |access-date=June 11, 2017}}</ref> The experts pointed out that Vivitrol's competitors, [[buprenorphine]] and [[methadone]], are "less expensive", "more widely used" and have been "rigorously studied".


Line 160: Line 181:


==See also==
==See also==
* [[Nalodeine]]
* [[One Little Pill (2014 film)|''One Little Pill'' (2014 film)]] - documentary about using naltrexone to treat alcohol use disorder
* [[Samidorphan]]
* [[Buprenorphine/naltrexone]]
* [[Opioid antagonist#List of opioid antagonists|Opioid antagonist § List of opioid antagonists]]
* [[Opioid antagonist#List of opioid antagonists|Opioid antagonist § List of opioid antagonists]]
* [[One Little Pill (2014 film)|''One Little Pill'' (2014 film)]] - documentary about using naltrexone to treat alcohol use disorder


==References==
==References==
{{Reflist|30em}}
{{Reflist|30em}}


==External links==


{{Antiaddictives}}
{{Antiaddictives}}
Line 175: Line 192:


[[Category:Alcohol abuse]]
[[Category:Alcohol abuse]]
[[Category:Alcohols]]
[[Category:Cyclopropanes]]
[[Category:Cyclopropanes]]
[[Category:Diols]]
[[Category:Ethers]]
[[Category:Ethers]]
[[Category:GABAA receptor negative allosteric modulators]]
[[Category:GABAA receptor negative allosteric modulators]]

Revision as of 01:50, 4 December 2017

Naltrexone
Clinical data
Trade namesRevia, Vivitrol, others
Other namesEN-1639A; UM-792; N-Cyclopropyl-methylnoroxymorphone; N-Cyclopropylmethyl-14-hydroxydihydro-morphinone; 17-(Cyclopropylmethyl)-4,5α-epoxy-3,14-dihydroxymorphinan-6-one
AHFS/Drugs.comMonograph
MedlinePlusa685041
Pregnancy
category
  • AU: B3
Routes of
administration
By mouth, intramuscular injection
ATC code
Legal status
Legal status
  • AU: S4 (Prescription only)
  • CA: ℞-only
  • UK: POM (Prescription only)
  • US: ℞-only
  • In general: ℞ (Prescription only)
Pharmacokinetic data
Bioavailability5–40%
Protein binding21%
MetabolismLiver
Elimination half-lifeNaltrexone: 4 hours
6β-Naltrexol: 13 hours
ExcretionUrine
Identifiers
  • (4R,4aS,7aR,12bS)-3-(cyclopropylmethyl)-4a,9-dihydroxy-2,4,5,6,7a,13-hexahydro-1H-4,12-methanobenzofuro[3,2-e]isoquinoline-7-one
CAS Number
PubChem CID
IUPHAR/BPS
DrugBank
ChemSpider
UNII
KEGG
ChEBI
ChEMBL
CompTox Dashboard (EPA)
ECHA InfoCard100.036.939 Edit this at Wikidata
Chemical and physical data
FormulaC20H23NO4
Molar mass341.401 g/mol g·mol−1
3D model (JSmol)
Melting point169 °C (336 °F)
  • O=C4[C@@H]5Oc1c2c(ccc1O)C[C@H]3N(CC[C@]25[C@@]3(O)CC4)CC6CC6
  • InChI=1S/C20H23NO4/c22-13-4-3-12-9-15-20(24)6-5-14(23)18-19(20,16(12)17(13)25-18)7-8-21(15)10-11-1-2-11/h3-4,11,15,18,22,24H,1-2,5-10H2/t15-,18+,19+,20-/m1/s1 checkY
  • Key:DQCKKXVULJGBQN-XFWGSAIBSA-N checkY
 ☒NcheckY (what is this?)  (verify)

Naltrexone, sold under the brand names Revia and Vivitrol among others, is a medication primarily used to manage alcohol dependence and opioid dependence.[1] In opioid dependence, it should not be started until people are detoxified.[1] It is taken by mouth or by injection into a muscle.[1] Effects begin within 30 minutes.[1] A decreased desire for opioids however may take a few weeks.[1]

Side effects may include trouble sleeping, anxiety, nausea, and headaches.[1] In those still on opioids, opioid withdrawal may occur.[1] Use is not recommended in people with liver failure.[1] It is unclear if use is safe during pregnancy.[1][2] Naltrexone is an opioid antagonist and works by blocking the effects of opioids, both those in the body like endorphins and those from outside of the body like morphine and heroin.[1]

Naltrexone was first made in 1965 and was approved for medical use in the United States in 1984.[1][3] As of 2017, the wholesale cost of tablets is about US$0.74 per day in the US.[4][5] The extended-release injections cost about $1,267 per month ($41.20 per day).[5] Naltrexone, as bupropion/naltrexone, is also used to treat obesity.[6]

Medical uses

Alcoholism

Naltrexone has been best studied as a treatment for alcoholism.[7] Naltrexone has been shown to decrease the amount and frequency of drinking.[8] It does not appear to change the percentage of people drinking.[9] Its overall benefit has been described as "modest".[10]

Acamprosate may work better than naltrexone for eliminating drinking, while naltrexone may decrease the desire for alcohol to a greater extent.[11]

The Sinclair method is a method of using opiate antagonists such as naltrexone to treat alcoholism. The person takes the medication about an hour (and only then) before drinking to avoid side effects that arise from chronic use.[12][13] The opioid antagonist blocks the positive reinforcement effects of alcohol and allows the person to stop or reduce drinking.[13]

Opioid use

Long acting injectable naltrexone decreases heroin use more than placebo.[14] It has benefits over methadone and buprenorphine in that it is not a restricted medication.[14] It may decrease cravings for opioids after a number of weeks and decreases the risk of overdose.[1][15] It is given once per month and has better compliance than the by mouth formulation.[16]

A 2011 review found insufficient evidence to determine the effect of naltrexone take by mouth in opioid dependence.[17] While some do well with this formulation, it must be taken daily, and a person whose cravings become overwhelming can obtain opioid intoxication simply by skipping a dose. Due to this issue, the usefulness of oral naltrexone in opioid use disorders is limited by the low retention in treatment. Naltrexone by mouth remains an ideal treatment for a small number of people with opioid use, usually those with a stable social situation and motivation. With additional contingency management support, naltrexone may be effective in a broader population.[18]

Others

Naltrexone is not useful for quitting smoking.[19]

Available forms

Vivitrol, a naltrexone formulation for depot injection, was approved by the FDA on April 13, 2006, for the treatment of alcohol dependence.[20]

Additionally, naltrexone implants that are surgically implanted are available,[21] While these are manufactured in Australia, they are not authorized for use within Australia, but only for export.[22] By 2009, naltrexone implants showed encouraging results.[23]

Contraindications

Naltrexone should not be used by persons with acute hepatitis or liver failure, or those with recent opioid use (typically 7–10 days).

Side effects

The most common side effects reported with naltrexone are gastrointestinal complaints such as diarrhea and abdominal cramping. These adverse effects are analogous to the symptoms of opioid withdrawal, as the mu receptor blockade will increase GI motility.

Naltrexone has been reported to cause liver damage (when given at doses higher than recommended). It carries an FDA boxed warning for this rare side effect. Due to these reports, some physicians may check liver function tests prior to starting naltrexone, and periodically thereafter. Concerns for liver toxicity initially arose from a study of non-addicted obese patients receiving 300 mg of naltrexone.[24] Subsequent studies have suggested limited toxicity in other patient populations.

Naltrexone should not be started until several (typically 7-10) days of abstinence from opioids has been achieved. This is due to the risk of acute opioid withdrawal if naltrexone is taken, as naltrexone will displace most opioids from their receptors. The time of abstinence may be shorter than 7 days, depending on the half-life of the specific opioid taken. Some physicians use a naloxone challenge to determine whether an individual has any opioids remaining. The challenge involves giving a test dose of naloxone and monitoring for opioid withdrawal. If withdrawal occurs, naltrexone should not be started.[25]

Pharmacology

Pharmacodynamics

Naltrexone and its active metabolite 6β-naltrexol are competitive antagonists at the μ-opioid receptor (MOR), the κ-opioid receptor (KOR) to a lesser extent, and to a far lesser and possibly insignificant extent, at the δ-opioid receptor (DOR).[26] The Ki affinity values of naltrexone at the MOR, KOR, and DOR have been reported as 0.0825 nM, 0.509 nM, and 8.02 nM, respectively, demonstrating a MOR/KOR binding ratio of 6.17 and a MOR/DOR binding ratio of 97.2.[27]

Mechanism of action

The blockade of opioid receptors is the basis behind naltrexone's action in the management of opioid dependence—it reversibly blocks or attenuates the effects of opioids. Its mechanism of action in alcohol dependence is not fully understood, but as an opioid receptor antagonist is likely to be due to the modulation of the dopaminergic mesolimbic pathway (one of the primary centers for risk-reward analysis in the brain, and a tertiary "pleasure center") which is hypothesized to be a major center of the reward associated with addiction that all major drugs of abuse are believed to activate. [citation needed] Mechanism of action may be antagonism to endogenous opioids such as tetrahydropapaveroline, whose production is augmented in the presence of alcohol.[28]

Pharmacokinetics

Naltrexone is metabolized in the liver mainly to 6β-naltrexol by the enzyme dihydrodiol dehydrogenase. Other metabolites include 2-hydroxy-3-methoxy-6β-naltrexol and 2-hydroxy-3-methoxy-naltrexone. These are then further metabolized by conjugation with glucuronide.[citation needed] The plasma half-life of naltrexone and its metabolite 6β-naltrexol are about 4 hours and 13 hours, respectively.[citation needed]

Pharmacogenetics

A naltrexone treatment study by Anton et al., released by the National Institutes of Health in February 2008 and published in the Archives of General Psychiatry, has shown that alcoholics having a certain variant of the opioid receptor gene (G polymorphism of SNP Rs1799971 in the gene OPRM1), known as Asp40, demonstrated strong response to naltrexone and were far more likely to experience success at cutting back or discontinuing their alcohol intake altogether, while for those lacking the gene variant, naltrexone appeared to be no different from placebo.[29] The G allele of OPRM1 is most common in individuals of Asian descent, with 60% to 70% of people of Chinese, Japanese, and Indian ancestry having at least one copy, as opposed to 30% of Europeans and few Africans.[30]

Because of the characteristics of the patient group in the US, the first study was done on white patients and the next without regard for ethnicity. Anton et al. found that patients of African descent did not have much success with naltrexone in treatment for alcohol dependence because of lacking the relevant gene.[29]

As white patients with the gene had a five times greater rate of success in reducing drinking when given naltrexone than did patients without the gene, when used in a protocol of Medical Management (MM), Anton et al. concluded,

"Because almost 25% of the treatment-seeking population carries the Asp40 allele, genetic testing of individuals before naltrexone treatment might be worth the cost and effort, especially if structured behavioral treatment were not being considered."[29] This would enable treatment to be targeted by genetics to patients for whom it would be most effective. They noted, "Naltrexone is relatively easy to administer and free of serious adverse effects and, as we observed in the Asp40 carriers we studied, it appears to be highly effective."[29]

Studies have found naltrexone to be more efficacious among certain white subjects, because of the genetic basis, than among black subjects, who generally do not carry the relevant gene variant.[31] A 2009 study of naltrexone as an alcohol dependence treatment among African Americans failed to find any statistically significant differences between naltrexone and a placebo.[32] Studies have suggested that carriers of the G allele may experience higher levels of craving and stronger "high" upon alcohol consumption, compared to carriers of the dominant allele, and naltrexone somewhat blunts these responses, leading to a reduction in alcohol use in some studies.[33]

Chemistry

Naltrexone can be described as a substituted oxymorphone – here the tertiary amine methyl-substituent is replaced with methylcyclopropane. Naltrexone is the N-cyclopropylmethyl derivative of oxymorphone.[citation needed]

Analogues

The closely related medication, methylnaltrexone, is used to treat opioid-induced constipation but does not treat addiction as it does not cross the blood–brain barrier. Nalmefene is similar to naltrexone and is used for the same purposes as naltrexone. Naltrexone should not be confused with naloxone, which is used in emergency cases of opioid overdose. Other related opioid antagonists include nalodeine and samidorphan.

History

Naltrexone was first synthesized in 1965 by Blumberg and Dayton and was found to be an orally active and long-acting opioid antagonist.[34][35][3] It showed advantages over earlier opioid antagonists like cyclazocine, nalorphine, and naloxone including its oral activity, a long duration of action allowing for once-daily administration, and a lack of dysphoria.[3] The drug was approved by the FDA for the oral treatment of heroin addiction in 1984 and for the oral treatment of alcohol dependence in 1995.[36] A depot formulation for intramuscular injection was approved by the FDA for alcohol dependence in 2006.[20]

Society and culture

Generic names

Naltrexone is the generic name of the drug and its INNTooltip International Nonproprietary Name, BANTooltip British Approved Name, DCFTooltip Dénomination Commune Française, and DCITTooltip Denominazione Comune Italiana, while naltrexone hydrochloride is its USPTooltip United States Pharmacopeia and BANMTooltip British Approved Name.[37][38][39][40]

Brand names

Naltrexone is marketed under a variety of brand names including Adepend, Antaxone, Celupan, Nalorex, Narcoral, Nemexin, Revia, ReVia, Trexan, and Vivitrol among others.[37][38][39][40] It is also marketed in combination with bupropion (bupropion/naltrexone) as Contrave and was marketed with morphine (morphine/naltrexone) as Embeda.[40] A combination of naltrexone with buprenorphine (buprenorphine/naltrexone) has been developed but has not been marketed.[41]

Controversies

2011 Krupitsky study

The FDA authorized use of injectable naltrexone for opioid addiction using a single study[42] that was led by Evgeny Krupitsky at Bekhterev Research Psychoneurological Institute, St Petersburg State Pavlov Medical University, St Petersburg, Russia,[43] a country where opioid agonists such as methadone and buprenorphine are not available. The study was a "double-blind, placebo-controlled, randomized", 24-week trial running "from July 3, 2008 through October 5, 2009" with "250 patients with opioid dependence disorder" at "13 clinical sites in Russia" on the use of injectable naltrexone (XR-NTX) for opioid dependence. The study was funded by the Boston-based biotech Alkermes firm which produces and markets naltrexone in the United States.

A 2011 article reported that this single trial of naltrexone was performed not by comparing it to the best available, evidence-based treatment (methadone or buprenorphine) but by comparing it with a placebo.[44] A subsequent RCT in Norway did compare injectable naltrexone to buprenorphine and found it to be similar in outcomes.[45]

Tom Price's remarks 2017

In May 2017, United States Secretary of Health and Human Services Tom Price, praised [vivitrol] as the future of opioid addiction treatment after visiting the company’s plant in Ohio.[46] His remarks set off sharp criticism with almost 700 experts in the field of substance abuse submitting a letter to Price cautioning him about Vivitrol's "marketing tactics" and warning him that his comment "ignore widely accepted science".[47] The experts pointed out that Vivitrol's competitors, buprenorphine and methadone, are "less expensive", "more widely used" and have been "rigorously studied".

Price had claimed that buprenorphine and methadone were "simply substitute"s for "illicit drugs"[46] whereas according to the letter, "the substantial body of research evidence supporting these treatments is summarized in guidance from within your own agency, including the Substance Abuse and Mental Health Services Administration, the US Surgeon General, the National Institute on Drug Abuse, and the Centers for Disease Control and Prevention. To briefly summarize, buprenorphine and methadone have been demonstrated to be highly effective in managing the core symptoms of opioid use disorder, reducing the risk of relapse and fatal overdose, and encouraging long-term recovery."[47]

According to a June 11, 2017 The New York Times article, Alkermes "has spent years coaxing, with a deft lobbying strategy that has targeted lawmakers and law enforcement officials. The company has spent millions of dollars on contributions to officials struggling to stem the epidemic of opioid abuse. It has also provided thousands of free doses to encourage the use of Vivitrol in jails and prisons, which have by default become major detox centers".[46]

Research

Depersonalization

Naltrexone is sometimes used in the treatment of dissociative symptoms such as depersonalization and derealization.[48][49] Some studies suggest it might help.[50] Other small, preliminary studies have also shown benefit.[48][49] It is thought that blockade of the KOR by naltrexone and naloxone is responsible for their effectiveness in ameliorating depersonalization and derealization.[48][49] Since these drugs are less efficacious in blocking the KOR relative to the MOR, higher dose than typically used seem to be necessary.[48][49]

Low-dose

"Low-dose naltrexone" (LDN) describes the "off-label" use of naltrexone at low doses for diseases not related to chemical dependency or intoxication, such as multiple sclerosis.[51] More research needs to be done before it can be recommended for clinical use.

Although there are scientific studies showing its efficacy in some conditions such as fibromyalgia,[52] other, more dramatic claims for its use in conditions like cancer and HIV have less scientific support.[51] This treatment has received significant attention on the Internet, especially through websites run by organizations promoting its use.[53]

Self-injury

Some studies suggest that self-injurious behaviors present in persons with developmental disabilities (including autism) can sometimes be remedied with naltrexone.[54] In these cases, it is believed that the self-injury is being done to release beta-endorphin, which binds to the same receptors as heroin and morphine.[55] If the "rush" generated by self-injury is removed, the behavior may stop.

Behavioral disorders

There are indications that naltrexone might be beneficial in the treatment of impulse control disorders such as kleptomania, compulsive gambling, or trichotillomania (compulsive hair pulling), but there is conflicting evidence of its effectiveness for gambling.[56][57][58] A 2008 case study reported successful use of naltrexone in suppressing and treating an internet pornography addiction.[59]

Interferon alpha

Naltrexone is effective in suppressing the cytokine-mediated adverse neuropsychiatric effects of interferon alpha therapy.[60][61]

See also

References

  1. ^ a b c d e f g h i j k l "Naltrexone Monograph for Professionals - Drugs.com". Drugs.com. American Society of Health-System Pharmacists. Retrieved 9 November 2017.
  2. ^ Tran, TH; Griffin, BL; Stone, RH; Vest, KM; Todd, TJ (July 2017). "Methadone, Buprenorphine, and Naltrexone for the Treatment of Opioid Use Disorder in Pregnant Women". Pharmacotherapy. 37 (7): 824–839. doi:10.1002/phar.1958. PMID 28543191.
  3. ^ a b c Sadock, Benjamin J.; Sadock, Virginia A.; Sussman, Norman (2012). Kaplan & Sadock's Pocket Handbook of Psychiatric Drug Treatment. Lippincott Williams & Wilkins. p. 265. ISBN 9781451154467.
  4. ^ "Naltrexone". ATC/DDD Index. WHO Collaborating Centre for Drug Statistics Methodology. Retrieved 11 July 2016. DDD ... 50 mg
  5. ^ a b "NADAC as of 2017-11-29". Centers for Medicare and Medicaid Services. Retrieved 3 December 2017.
  6. ^ "Naltrexone/bupropion for obesity". Drug and therapeutics bulletin. 55 (11): 126–129. November 2017. doi:10.1136/dtb.2017.11.0550. PMID 29117992.
  7. ^ Aboujaoude, E; Salame, WO (August 2016). "Naltrexone: A Pan-Addiction Treatment?". CNS drugs. 30 (8): 719–33. doi:10.1007/s40263-016-0373-0. PMID 27401883.
  8. ^ Rösner, S; Hackl-Herrwerth, A; Leucht, S; Vecchi, S; Srisurapanont, M; Soyka, M (8 December 2010). "Opioid antagonists for alcohol dependence". The Cochrane database of systematic reviews (12): CD001867. PMID 21154349.
  9. ^ Donoghue, Kim; Elzerbi, Catherine; Saunders, Rob; Whittington, Craig; Pilling, Stephen; Drummond, Colin (2015). "The efficacy of acamprosate and naltrexone in the treatment of alcohol dependence, Europe versus the rest of the world: A meta-analysis". Addiction. 110 (6): 920–30. doi:10.1111/add.12875. PMID 25664494.
  10. ^ c. Garbutt, James (2010). "Efficacy and Tolerability of Naltrexone in the Management of Alcohol Dependence". Current Pharmaceutical Design. 16 (19): 2091–7. doi:10.2174/138161210791516459. PMID 20482515.
  11. ^ Maisel, Natalya C.; Blodgett, Janet C.; Wilbourne, Paula L.; Humphreys, Keith; Finney, John W. (October 17, 2012). "Meta-analysis of naltrexone and acamprosate for treating alcohol use disorders: When are these medications most helpful?". Addiction. 108 (2): 275–93. doi:10.1111/j.1360-0443.2012.04054.x. PMC 3970823. PMID 23075288.
  12. ^ Anderson, Kenneth (Jul 28, 2013). "Drink Your Way Sober with Naltrexone". Psychology Today. Retrieved 18 July 2016.
  13. ^ a b Sinclair, J. D. (2001). "Evidence about the use of naltrexone and for different ways of using it in the treatment of alcoholism". Alcohol and Alcoholism. 36 (1): 2–10. doi:10.1093/alcalc/36.1.2. PMID 11139409.
  14. ^ a b Sharma, A; Kelly, SM; Mitchell, SG; Gryczynski, J; O'Grady, KE; Schwartz, RP (June 2017). "Update on Barriers to Pharmacotherapy for Opioid Use Disorders". Current psychiatry reports. 19 (6): 35. doi:10.1007/s11920-017-0783-9. PMID 28526967.
  15. ^ Sharma, B; Bruner, A; Barnett, G; Fishman, M (July 2016). "Opioid Use Disorders". Child and adolescent psychiatric clinics of North America. 25 (3): 473–87. doi:10.1016/j.chc.2016.03.002. PMID 27338968.
  16. ^ Comer, Sandra D.; Sullivan, Maria A.; Yu, Elmer; Rothenberg, Jami L.; Kleber, Herbert D.; Kampman, Kyle; Dackis, Charles; o'Brien, Charles P. (2006). "Injectable, Sustained-Release Naltrexone for the Treatment of Opioid Dependence". Archives of General Psychiatry. 63 (2): 210–8. doi:10.1001/archpsyc.63.2.210. PMC 4200530. PMID 16461865.
  17. ^ Minozzi, S; Amato, L; Vecchi, S; Davoli, M; Kirchmayer, U; Verster, A (13 April 2011). "Oral naltrexone maintenance treatment for opioid dependence". The Cochrane database of systematic reviews (4): CD001333. doi:10.1002/14651858.CD001333.pub4. PMID 21491383.
  18. ^ Johansson, Björn Axel; Berglund, Mats; Lindgren, Anna (2006). "Efficacy of maintenance treatment with naltrexone for opioid dependence: A meta-analytical review". Addiction. 101 (4): 491–503. doi:10.1111/j.1360-0443.2006.01369.x. PMID 16548929.
  19. ^ David, Sean P.; Lancaster, Tim; Stead, Lindsay F.; Evins, A. Eden; Prochaska, Judith J. (6 June 2013). "Opioid antagonists for smoking cessation". The Cochrane Database of Systematic Reviews (6): CD003086. doi:10.1002/14651858.CD003086.pub3. ISSN 1469-493X. PMC 4038652. PMID 23744347.
  20. ^ a b "Alcoholism Once A Month Injectable Drug, Vivitrol, Approved By FDA," Medical News Today, April 16, 2006.
  21. ^ Therapeutic Goods Administration. "Australian Register of Therapeutic Goods Medicines" (Online database of approved medicines). Retrieved 2009-03-22.
  22. ^ Therapeutic Goods Administration. "Australian Register of Therapeutic Goods Medicines" (Online database of approved medicines, specific entry for "O'Neil Long Acting Naltrexone Implant"). Retrieved 2017-04-27.
  23. ^ Hulse, Gary K.; Morris, Noella; Arnold-Reed, Diane; Tait, Robert J. (2009). "Improving Clinical Outcomes in Treating Heroin Dependence". Archives of General Psychiatry. 66 (10): 1108–15. doi:10.1001/archgenpsychiatry.2009.130. PMID 19805701.
  24. ^ Pfohl, David N.; Allen, John I.; Atkinson, Richard L.; Knopman, David S.; Malcolm, Robert J.; Mitchell, James E.; Morley, John E. (1986). "Naltrexone hydrochloride (Trexan): A review of serum transaminase elevations at high dosage". NIDA research monograph. 67: 66–72. PMID 3092099.
  25. ^ Galanter, Marc; Kleber, Herbert. The American Psychiatric Publishing Textbook of Substance Abuse Treatment, 4th Edition. ISBN 1585622761[page needed]
  26. ^ Niciu, Mark J.; Arias, Albert J. (2013). "Targeted Opioid Receptor Antagonists in the Treatment of Alcohol Use Disorders". CNS Drugs. 27 (10): 777–87. doi:10.1007/s40263-013-0096-4. PMC 4600601. PMID 23881605.
  27. ^ Codd, E. E.; Shank, R. P.; Schupsky, J. J.; Raffa, R. B. (1995). "Serotonin and norepinephrine uptake inhibiting activity of centrally acting analgesics: Structural determinants and role in antinociception". The Journal of Pharmacology and Experimental Therapeutics. 274 (3): 1263–70. PMID 7562497.
  28. ^ Haber, Hanka; Roske, Irmgard; Rottmann, Matthias; Georgi, Monika; Melzig, Matthias F. (1996). "Alcohol induces formation of morphine precursors in the striatum of rats". Life Sciences. 60 (2): 79–89. doi:10.1016/S0024-3205(96)00597-8. PMID 9000113.
  29. ^ a b c d Anton, Raymond F.; Oroszi, Gabor; o'Malley, Stephanie; Couper, David; Swift, Robert; Pettinati, Helen; Goldman, David (2008). "An Evaluation of μ-Opioid Receptor (OPRM1) as a Predictor of Naltrexone Response in the Treatment of Alcohol Dependence". Archives of General Psychiatry. 65 (2): 135–44. doi:10.1001/archpsyc.65.2.135. PMC 2666924. PMID 18250251.
  30. ^ "Rs1799971 (SNPedia)".
  31. ^ Kranzler, Henry R.; Kirk, Jeffrey (2001). "Efficacy of Naltrexone and Acamprosate for Alcoholism Treatment: A Meta-Analysis". Alcoholism: Clinical and Experimental Research. 25 (9): 1335–41. doi:10.1111/j.1530-0277.2001.tb02356.x. PMID 11584154.
  32. ^ Ray, Lara A.; Oslin, David W. (2009). "Naltrexone for the treatment of alcohol dependence among African Americans: Results from the COMBINE Study". Drug and Alcohol Dependence. 105 (3): 256–8. doi:10.1016/j.drugalcdep.2009.07.006. PMC 3409877. PMID 19717248.
  33. ^ Ray, Lara A.; Barr, Christina S.; Blendy, Julie A.; Oslin, David; Goldman, David; Anton, Raymond F. (2012). "The Role of the Asn40Asp Polymorphism of the Mu Opioid Receptor Gene (OPRM1) on Alcoholism Etiology and Treatment: A Critical Review". Alcoholism: Clinical and Experimental Research. 36 (3): 385–94. doi:10.1111/j.1530-0277.2011.01633.x. PMC 3249007. PMID 21895723.
  34. ^ Howard Padwa; Jacob Cunningham (2010). Addiction: A Reference Encyclopedia. ABC-CLIO. pp. 207–. ISBN 978-1-59884-229-6.
  35. ^ G Bennett (14 January 2004). Treating Drug Abusers. Routledge. pp. 112–. ISBN 978-1-134-93173-6.
  36. ^ H. Thomas Milhorn (17 October 2017). Substance Use Disorders: A Guide for the Primary Care Provider. Springer International Publishing. pp. 88–. ISBN 978-3-319-63040-3.
  37. ^ a b J. Elks (14 November 2014). The Dictionary of Drugs: Chemical Data: Chemical Data, Structures and Bibliographies. Springer. pp. 851–. ISBN 978-1-4757-2085-3.
  38. ^ a b Index Nominum 2000: International Drug Directory. Taylor & Francis. 2000. pp. 715–. ISBN 978-3-88763-075-1.
  39. ^ a b I.K. Morton; Judith M. Hall (6 December 2012). Concise Dictionary of Pharmacological Agents: Properties and Synonyms. Springer Science & Business Media. pp. 189–. ISBN 978-94-011-4439-1.
  40. ^ a b c https://www.drugs.com/international/naltrexone.html
  41. ^ McCann DJ (2008). "Potential of buprenorphine/naltrexone in treating polydrug addiction and co-occurring psychiatric disorders". Clin. Pharmacol. Ther. 83 (4): 627–30. doi:10.1038/sj.clpt.6100503. PMID 18212797.
  42. ^ "A Shot in the Dark: Can Vivitrol Help Us Control Our Addictions?". 7 May 2013.
  43. ^ Krupitsky, Evgeny; Nunes, Edward V.; Ling, Walter; Illeperuma, Ari; Gastfriend, David R.; Silverman, Bernard L. (April 28, 2011). "Injectable extended-release naltrexone for opioid dependence: a double-blind, placebo-controlled, multicentre randomised trial" (PDF). The Lancet. 377: 1506–13. doi:10.1016/S0140. Retrieved June 11, 2017.
  44. ^ Wolfe, Daniel; Carrieri, MP; Dasgupta, Nabarun; Bruce, Douglas; Wodak, Alex (2011). "Injectable extended-release naltrexone for opioid dependence – Authors' reply". The Lancet. 378 (9792): 666. doi:10.1016/S0140-6736(11)61333-0.
  45. ^ Tanum, L; Solli, KK; Latif, ZE; Benth, JŠ; Opheim, A; Sharma-Haase, K; Krajci, P; Kunøe, N (18 October 2017). "The Effectiveness of Injectable Extended-Release Naltrexone vs Daily Buprenorphine-Naloxone for Opioid Dependence: A Randomized Clinical Noninferiority Trial". JAMA psychiatry. doi:10.1001/jamapsychiatry.2017.3206. PMID 29049469.
  46. ^ a b c Goodnough, Abby; Zernike, Kate (June 11, 2017). "Seizing on Opioid Crisis, a Drug Maker Lobbies Hard for Its Product". The New York Times. Retrieved June 11, 2017. Advertising for Vivitrol on a subway car in Brooklyn last month. Marketing for the drug has shifted into high gear.
  47. ^ a b "Letter to Tom Price". May 2017. Retrieved June 11, 2017.
  48. ^ a b c d Daphne Simeon; Jeffrey Abugel (10 October 2008). Feeling Unreal: Depersonalization Disorder and the Loss of the Self. Oxford University Press. pp. 166–. ISBN 978-0-19-976635-2.
  49. ^ a b c d Ulrich F. Lanius, PhD; Sandra L. Paulsen, PhD; Frank M. Corrigan, MD (13 May 2014). Neurobiology and Treatment of Traumatic Dissociation: Towards an Embodied Self. Springer Publishing Company. pp. 489–. ISBN 978-0-8261-0632-2.
  50. ^ Sierra, Mauricio (2014). "Depersonalization disorder: Pharmacological approaches". Expert Review of Neurotherapeutics. 8 (1): 19–26. doi:10.1586/14737175.8.1.19. PMID 18088198.
  51. ^ a b Novella, Steven (5 May 2010). "Low Dose Naltrexone – Bogus or Cutting Edge Science?". Science-Based Medicine. Retrieved 5 July 2011.
  52. ^ Younger, Jarred; MacKey, Sean (2009). "Fibromyalgia Symptoms Are Reduced by Low-Dose Naltrexone: A Pilot Study". Pain Medicine. 10 (4): 663–72. doi:10.1111/j.1526-4637.2009.00613.x. PMC 2891387. PMID 19453963.
  53. ^ Bowling, Allen C. "Low-dose naltrexone (LDN) The "411" on LDN". National Multiple Sclerosis Society. Archived from the original on 22 December 2011. Retrieved 6 July 2011. {{cite web}}: Unknown parameter |deadurl= ignored (|url-status= suggested) (help)
  54. ^ Smith, Stanley G.; Gupta, Krishan K.; Smith, Sylvia H. (1995). "Effects of naltrexone on self-injury, stereotypy, and social behavior of adults with developmental disabilities". Journal of Developmental and Physical Disabilities. 7 (2): 137–46. doi:10.1007/BF02684958.
  55. ^ Manley, Cynthia (1998-03-20). "Self-injuries may have biochemical base: study". The Reporter.
  56. ^ Grant, Jon E.; Kim, Suck Won; Odlaug, Brian L. (2009). "A Double-Blind, Placebo-Controlled Study of the Opiate Antagonist, Naltrexone, in the Treatment of Kleptomania". Biological Psychiatry. 65 (7): 600–6. doi:10.1016/j.biopsych.2008.11.022. PMID 19217077. {{cite journal}}: Unknown parameter |laydate= ignored (help); Unknown parameter |laysource= ignored (help); Unknown parameter |laysummary= ignored (help)
  57. ^ "A Randomized, Double-Blind, Placebo-Controlled Trial of Naltrexone in the Treatment of Concurrent Alcohol Dependence and Pathological Gambling - ICH GCP - Clinical Trials Registry".
  58. ^ Kim, Suck Won; Grant, Jon E; Adson, David E; Shin, Young Chul (2001). "Double-blind naltrexone and placebo comparison study in the treatment of pathological gambling". Biological Psychiatry. 49 (11): 914–21. doi:10.1016/S0006-3223(01)01079-4. PMID 11377409.
  59. ^ Bostwick, J. Michael; Bucci, Jeffrey A. (2008). "Internet Sex Addiction Treated with Naltrexone". Mayo Clinic Proceedings. 83 (2): 226–30. doi:10.4065/83.2.226. PMID 18241634.
  60. ^ Vignau, J.; Karila, L.; Costisella, O.; Canva, V. (2005). "Hépatite C, Interféron α et dépression : Principales hypothèses physiopathologiques" [Hepatitis C, interferon a and depression: main physiopathologic hypothesis]. L'Encéphale (in French). 31 (3): 349–57. doi:10.1016/s0013-7006(05)82400-5. PMID 16142050. INIST 16920336.
  61. ^ Małyszczak, Krzysztof; Inglot, Małgorzata; Pawłowski, Tomasz; Czarnecki, Marcin; Rymer, Weronika; Kiejna, Andrzej (2006). "Objawy neuropsychiatryczne w trakcie leczenia interferonem alfa" [Neuropsychiatric symptoms related to interferon alpha]. Psychiatria polska (in Polish). 40 (4): 787–97. PMID 17068950.