Naltrexone

From Wikipedia, the free encyclopedia
Naltrexone
Naltrexone skeletal.svg
Naltrexone-from-xtal-3D-bs-17.png
Clinical data
Pronunciation/ˌnælˈtrɛksn/
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
License data
Pregnancy
category
  • AU: B3
Routes of
administration
By mouth, intramuscular injection, subcutaneous implant
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-lifeOral (ReVia):[1]
• Naltrexone: 4 hours
6β-Naltrexol: 13 hours
Oral (Contrave):[2]
• Naltrexone: 5 hours
IM (Vivitrol):[3]
• Naltrexone: 5–10 days
• 6β-Naltrexol: 5–10 days
ExcretionUrine
Identifiers
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 mass341.407 g·mol−1
3D model (JSmol)
Melting point169 °C (336 °F)
SMILES
InChI
 ☒NcheckY (what is this?)  

Naltrexone, sold under the brand names ReVia and Vivitrol among others, is a medication primarily used to manage alcohol or opioid use disorder by reducing cravings and feelings of euphoria associated with substance use disorder.[4] It has also been found to be effective in the treatment of other addictions and may be used for them off-label.[5] An opioid-dependent person should not receive naltrexone before detoxification.[4] It is taken by mouth or by injection into a muscle.[4] Effects begin within 30 minutes.[4] A decreased desire for opioids, though, may take a few weeks.[4]

Side effects may include trouble sleeping, anxiety, nausea, and headaches.[4] In those still on opioids, opioid withdrawal may occur.[4] Use is not recommended in people with liver failure.[4] It is unclear if use is safe during pregnancy.[4][6] Naltrexone is an opioid antagonist and works by blocking the effects of opioids, both those from inside and outside the body.[4]

Naltrexone was first made in 1965 and was approved for medical use in the United States in 1984.[4][7] Naltrexone, as naltrexone/bupropion (brand name Contrave), is also used to treat obesity.[8]

Medical uses[]

Alcoholism[]

Naltrexone has been best studied as a treatment for alcoholism.[5] Naltrexone has been shown to decrease the amount and frequency of drinking.[9] It does not appear to change the percentage of people drinking.[10] Its overall benefit has been described as "modest".[11]

Acamprosate may work better than naltrexone for eliminating drinking, while naltrexone may decrease the desire for alcohol to a greater extent.[12]

The Sinclair method is a method of using opiate antagonists such as naltrexone to treat alcoholism. The person takes the medication once, about an hour before drinking, to avoid side effects that arise from chronic use.[13][14] The opioid antagonist is thought to block the positive-reinforcement effects of alcohol and may assist the person to stop or reduce drinking.[14]

Opioid use[]

Long-acting injectable naltrexone decreases heroin use more than placebo.[15] It has benefits over methadone and buprenorphine in that it is not a restricted medication.[15] It may decrease cravings for opioids after a number of weeks, and decreases the risk of overdose, at least during the time period that naltrexone is still active.[4][16] It is given once per month and has better compliance than the oral formulation.[17]

A 2011 review found insufficient evidence to determine the effect of naltrexone taken by mouth in opioid dependence.[18] 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.[19]

Others[]

Low dose Naltrexone (LDN) is a novel therapy for autoimmune conditions such as lupus, fibromyalgia, MS and usually does not exceed a 4.5 mg dose. Naltrexone is not useful for quitting smoking.[20]

Available forms[]

Naltrexone is available and most commonly used in the form of an oral tablet (50 mg).[21] Vivitrol, a naltrexone formulation for depot injection containing 380 mg of the medication per vial, is also available.[21][22] Additionally, naltrexone subcutaneous implants that are surgically implanted are available.[23] While these are manufactured in Australia, they are not authorized for use within Australia, but only for export.[24] By 2009, naltrexone implants showed superior efficacy in the treatment of heroin dependence when compared to the oral form.[25]

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 μ-opioid receptor blockade will increase gastrointestinal motility.

Naltrexone has been reported to cause liver damage when given at doses higher than recommended.[26] 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 nonaddicted obese patients receiving 300 mg of naltrexone.[27] Subsequent studies have suggested limited or no toxicity in other patient populations and at typical recommended doses such as 50 to 100 mg/day.[26][5]

Naltrexone should not be started until several (typically 7–10) days of abstinence from opioids have 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.[28]

Whether or not naltrexone causes dysphoria, depression, anhedonia, or other aversive effects as side effects has been studied and reviewed.[29][30][31][32] Surprisingly, naltrexone seems to have minimal to no untoward influence in these areas.[29][30][31][32] According to one source:[30]

Naltrexone itself produces little or no psychoactive effect in normal research volunteers even at high doses, which is remarkable given that the endogenous opioid system is important in normal hedonic functioning. Because endogenous opioids are involved in the brain reward system, it would be reasonable to hypothesize that naltrexone might produce anhedonic or dysphoric effects. Although some evidence from small, early trials suggested that patients with a history of opiate dependence might be susceptible to dysphoric effects in response to naltrexone (Crowley et al. 1985; Hollister et al. 1981), reports of such effects have been inconsistent. Most large clinical studies of recovering opioid-dependent individuals have not found naltrexone to have an adverse effect on mood (Greenstein et al. 1984; Malcolm et al. 1987; Miotto et al. 2002; Shufman et al. 1994). Some studies have actually found improvements in mood during the course of treatment with naltrexone (Miotto et al. 1997; Rawlins and Randall 1976).

On the other hand, naltrexone has been found to reduce feelings of social connection.[33][34][35][36] Reports on whether naltrexone can decrease the pleasurable effects of listening to music are conflicting.[37][38][39]

Pharmacology[]

Pharmacodynamics[]

Naltrexone at opioid receptors
Affinities (Ki) Ratio Ref
MOR KOR DOR MOR:KOR:DOR
1.0 nM 3.9 nM 149 nM 1:4:149 [40]
0.0825 nM 0.509 nM 8.02 nM 1:6:97 [41]

Naltrexone and its active metabolite 6β-naltrexol are competitive antagonists of the μ-opioid receptor (MOR), the κ-opioid receptor (KOR) to a lesser extent, and the δ-opioid receptor (DOR) to a much lesser extent.[42] In one study, naltrexone was not a silent antagonist of these receptors but acted as a weak partial agonist, with Emax values of 29% at the MOR, 16% at the KOR, and 25% at the DOR.[43] In combination with agonists of these receptors however, naltrexone appears to become an inverse agonist of the MOR and a neutral antagonist of the KOR and DOR.[43] This may at least in part underlie its ability to precipitate withdrawal in opioid-dependent individuals.[43]

Occupancy of the opioid receptors by naltrexone has been studied using positron emission tomography (PET).[26][44] Naltrexone at a dose of 50 mg/day has been found to occupy approximately 90 to 95% of MORs and 20 to 35% of DORs.[26] Naltrexone at a dose of 100 mg/day with the last dose 2 hours before testing has been found to achieve 92.4 ± 1.3% occupancy of the KOR.[44] Per simulation, a lower dose of 25 mg/day would be expected to achieve around 60% occupancy of the KOR but still close to 90% occupancy of the MOR.[44]

In addition to the opioid receptors, naltrexone binds to and acts as an antagonist of the opioid growth factor receptor (OGFR) and toll-like receptor 4 (TLR4) and interacts with high- and low-affinity binding sites in filamin-A (FLNA).[45][46][47][48] It is said that very low doses of naltrexone (<0.001–1 mg/day) interact with FLNA, low doses (1 to 5 mg/day) produce TLR4 antagonism, and standard clinical doses (50 to 100 mg/day) exert opioid receptor and OGFR antagonism.[45][47]

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 thought to be generated via KOR antagonism,[49] which blocks the actions of the endogenous opioid peptide dynorphin.[50] Dynorphin typically instates drug-seeking behavior when it binds to the KOR, as well as decreasing dopaminergic signalling in the nucleus accumbens.[51]

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 glucuronic acid to form a glucuronide.[citation needed] The elimination half-life of naltrexone and its metabolite 6β-naltrexol are about 4 hours and 13 hours, respectively.[1] In Contrave oral tablets, which are extended-release, the half-life of naltrexone is 5 hours.[2] As Vivitrol, which is a suspension of microspheres for intramuscular injection, the half-life of both naltrexone and 6β-naltrexol is 5 to 10 days and is suitable for administration once every 4 weeks or once a month.[3] The half-life of occupancy of the MOR and duration of clinical effect of naltrexone appear to be longer than suggested by its plasma half-life.[52][53] A single 50 mg oral dose of naltrexone has been found to block MORs and opioid effects for 48 to 72 hours.[52][53][54]

Pharmacogenetics[]

Tentative evidence suggests that family history and presence of the Asn40Asp polymorphism predicts naltrexone being effective.[55][56]

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.

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 1963 by Metossian at Endo Laboratories, a small pharmaceutical company in New York City.[57] It was characterized by Blumberg, Dayton, and Wolf in 1965 and was found to be an orally active, long-acting, and very potent opioid antagonist.[57][58][59][7] The drug showed advantages over earlier opioid antagonists such as cyclazocine, nalorphine, and naloxone, including its oral activity, a long duration of action allowing for once-daily administration, and a lack of dysphoria, and was selected for further development.[7] It was patented by Endo Laboratories in 1967 under the developmental code name EN-1639A and Endo Laboratories was acquired by DuPont in 1969.[60][self-published source?] Clinical trials for opioid dependence began in 1973, and a developmental collaboration of DuPont with the National Institute on Drug Abuse for this indication started the next year in 1974.[60] The drug was approved by the FDA for the oral treatment of opioid dependence in 1984, with the brand name Trexan, and for the oral treatment of alcohol dependence in 1995, when the brand name was changed by DuPont to ReVia.[60][21] A depot formulation for intramuscular injection was approved by the FDA under the brand name Vivitrol for alcohol dependence in 2006 and opioid dependence in 2010.[22][21]

Society and culture[]

Generic names[]

Naltrexone is the generic name of the drug and its INN, USAN, BAN, DCF, and DCIT, while naltrexone hydrochloride is its USP and BANM.[61][62][63][64]

Brand names[]

Naltrexone is or has been marketed under a variety of brand names, including Adepend, Antaxone, Celupan, Depade, Nalorex, Narcoral, Nemexin, Nodict, Revia/ReVia, Trexan, and Vivitrol.[61][62][63][64] It is also marketed in combination with bupropion (naltrexone/bupropion) as Contrave,[65] and was marketed with morphine (morphine/naltrexone) as Embeda.[64][66] A combination of naltrexone with buprenorphine (buprenorphine/naltrexone) has been developed, but has not been marketed.[67]

Controversies[]

The FDA authorized use of injectable naltrexone for opioid addiction using a single study[68] that was led by Evgeny Krupitsky at Bekhterev Research Psychoneurological Institute, St Petersburg State Pavlov Medical University, St Petersburg, Russia,[69] 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.[70] A subsequent RCT in Norway did compare injectable naltrexone to buprenorphine and found them to be similar in outcomes.[71]

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.[72] His remarks set off sharp criticism with almost 700 experts in the field of substance use submitting a letter to Price cautioning him about Vivitrol's "marketing tactics" and warning him that his comment "ignore widely accepted science".[73] 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"[72] 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."[73]

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 use disorder. 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".[72]

Research[]

Depersonalization[]

Naltrexone is sometimes used in the treatment of dissociative symptoms such as depersonalization and derealization.[74][75] Some studies suggest it might help.[76] Other small, preliminary studies have also shown benefit.[74][75] Blockade of the KOR by naltrexone and naloxone is thought to be responsible for their effectiveness in ameliorating depersonalization and derealization.[74][75] Since these drugs are less efficacious in blocking the KOR relative to the MOR, higher doses than typically used seem to be necessary.[74][75]

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.[77] Evidence for recommending such use is lacking.[78][79]

This treatment has received attention on the Internet.[80]

Self-injury[]

One study suggests that self-injurious behaviors present in persons with developmental disabilities (including autism) can sometimes be remedied with naltrexone.[81] In these cases, the self-injury is believed to be done to release beta-endorphin, which binds to the same receptors as heroin and morphine.[82] If the "rush" generated by self-injury is removed, the behavior may stop.

Behavioral disorders[]

Some indications exist that naltrexone might be beneficial in the treatment of impulse-control disorders such as kleptomania, compulsive gambling, or trichotillomania (compulsive hair pulling), but evidence of its effectiveness for gambling is conflicting.[83][84][85] A 2008 case study reported successful use of naltrexone in suppressing and treating an internet pornography addiction.[86]

Interferon alpha[]

Naltrexone is effective in suppressing the cytokine-mediated adverse neuropsychiatric effects of interferon alpha therapy.[87][88]

Critical addiction studies[]

Some historians and sociologists have suggested that the meanings and uses attributed to anti-craving medicine, such as naltrexone, is context-dependent.[89] Studies have suggested the use of naltrexone in drug courts or healthcare rehabs is a form of "post-social control,"[90] or "post-disciplinary control,"[91] whereby control strategies for managing offenders and addicts shift from imprisonment and supervision toward more direct control over biological processes.

Sexual addiction[]

Small studies have shown a reduction of sexual addiction and problematic sexual behaviours from naltrexone.[92][93]

See also[]

  • Opioid antagonist § List of opioid antagonists
  • Buprenorphine/naltrexone
  • One Little Pill (2014 film) - documentary about using naltrexone to treat alcohol use disorder

References[]

  1. ^ Jump up to: a b http://www.accessdata.fda.gov/drugsatfda_docs/label/2013/018932s017lbl.pdf
  2. ^ Jump up to: a b https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/200063s020lbl.pdf
  3. ^ Jump up to: a b https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/021897s015lbl.pdf
  4. ^ Jump up to: 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. Archived from the original on 9 November 2017. Retrieved 9 November 2017.
  5. ^ Jump up to: a b c 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. S2CID 6372144.
  6. ^ 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. S2CID 13772333.
  7. ^ Jump up to: a b c Sadock BJ, Sadock VA, Sussman N (2012). Kaplan & Sadock's Pocket Handbook of Psychiatric Drug Treatment. Lippincott Williams & Wilkins. p. 265. ISBN 9781451154467. Archived from the original on 2017-12-05.
  8. ^ "Naltrexone/bupropion for obesity". Drug and Therapeutics Bulletin. 55 (11): 126–129. November 2017. doi:10.1136/dtb.2017.11.0550. PMID 29117992. S2CID 547660.
  9. ^ Rösner S, Hackl-Herrwerth A, Leucht S, Vecchi S, Srisurapanont M, Soyka M (December 2010). Srisurapanont M (ed.). "Opioid antagonists for alcohol dependence". The Cochrane Database of Systematic Reviews (12): CD001867. doi:10.1002/14651858.CD001867.pub2. PMID 21154349.
  10. ^ Donoghue K, Elzerbi C, Saunders R, Whittington C, Pilling S, Drummond C (June 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.
  11. ^ Garbutt JC (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.
  12. ^ Maisel NC, Blodgett JC, Wilbourne PL, Humphreys K, Finney JW (February 2013). "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.
  13. ^ Anderson K (Jul 28, 2013). "Drink Your Way Sober with Naltrexone". Psychology Today. Archived from the original on 16 September 2016. Retrieved 18 July 2016.
  14. ^ Jump up to: a b Sinclair JD (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.
  15. ^ Jump up to: 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. PMC 7075636. PMID 28526967.
  16. ^ 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. PMC 4920977. PMID 27338968.
  17. ^ Comer SD, Sullivan MA, Yu E, Rothenberg JL, Kleber HD, Kampman K, Dackis C, O'Brien CP (February 2006). "Injectable, sustained-release naltrexone for the treatment of opioid dependence: a randomized, placebo-controlled trial". Archives of General Psychiatry. 63 (2): 210–8. doi:10.1001/archpsyc.63.2.210. PMC 4200530. PMID 16461865.
  18. ^ Minozzi S, Amato L, Vecchi S, Davoli M, Kirchmayer U, Verster A (April 2011). Minozzi S (ed.). "Oral naltrexone maintenance treatment for opioid dependence". The Cochrane Database of Systematic Reviews (4): CD001333. doi:10.1002/14651858.CD001333.pub4. PMC 7045778. PMID 21491383.
  19. ^ Johansson BA, Berglund M, Lindgren A (April 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.
  20. ^ David SP, Lancaster T, Stead LF, Evins AE, Prochaska JJ (June 2013). "Opioid antagonists for smoking cessation". The Cochrane Database of Systematic Reviews (6): CD003086. doi:10.1002/14651858.CD003086.pub3. PMC 4038652. PMID 23744347.
  21. ^ Jump up to: a b c d 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.
  22. ^ Jump up to: a b "Alcoholism Once A Month Injectable Drug, Vivitrol, Approved By FDA Archived 2009-01-05 at the Wayback Machine," Medical News Today, April 16, 2006.
  23. ^ Therapeutic Goods Administration. "Australian Register of Therapeutic Goods Medicines" (Online database of approved medicines). Archived from the original on 2009-05-14. Retrieved 2009-03-22.
  24. ^ 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.
  25. ^ Hulse GK, Morris N, Arnold-Reed D, Tait RJ (October 2009). "Improving clinical outcomes in treating heroin dependence: randomized, controlled trial of oral or implant naltrexone". Archives of General Psychiatry. 66 (10): 1108–15. doi:10.1001/archgenpsychiatry.2009.130. PMID 19805701.
  26. ^ Jump up to: a b c d Ray LA, Chin PF, Miotto K (March 2010). "Naltrexone for the treatment of alcoholism: clinical findings, mechanisms of action, and pharmacogenetics". CNS Neurol Disord Drug Targets. 9 (1): 13–22. doi:10.2174/187152710790966704. PMID 20201811.
  27. ^ Pfohl DN, Allen JI, Atkinson RL, Knopman DS, Malcolm RJ, Mitchell JE, Morley JE (1986). "Naltrexone hydrochloride (Trexan): a review of serum transaminase elevations at high dosage". NIDA Research Monograph. 67: 66–72. PMID 3092099. Archived from the original on 2017-01-21. Retrieved 2017-01-23.
  28. ^ Galanter, Marc; Kleber, Herbert. The American Psychiatric Publishing Textbook of Substance Abuse Treatment, 4th Edition. ISBN 1585622761[page needed]
  29. ^ Jump up to: a b Miotto K, McCann M, Basch J, Rawson R, Ling W (2002). "Naltrexone and dysphoria: fact or myth?". Am J Addict. 11 (2): 151–60. doi:10.1080/10550490290087929. PMID 12028745.
  30. ^ Jump up to: a b c Eric C. Strain; Maxine L. Stitzer (2006). The Treatment of Opioid Dependence. JHU Press. pp. 296–. ISBN 978-0-8018-8219-7.
  31. ^ Jump up to: a b Nathan PJ, O'Neill BV, Napolitano A, Bullmore ET (October 2011). "Neuropsychiatric adverse effects of centrally acting antiobesity drugs". CNS Neurosci Ther. 17 (5): 490–505. doi:10.1111/j.1755-5949.2010.00172.x. PMC 6493804. PMID 21951371.
  32. ^ Jump up to: a b Krupitsky E, Zvartau E, Blokhina E, Verbitskaya E, Wahlgren V, Tsoy-Podosenin M, Bushara N, Burakov A, Masalov D, Romanova T, Tyurina A, Palatkin V, Yaroslavtseva T, Pecoraro A, Woody G (September 2016). "Anhedonia, depression, anxiety, and craving in opiate dependent patients stabilized on oral naltrexone or an extended release naltrexone implant". Am J Drug Alcohol Abuse. 42 (5): 614–620. doi:10.1080/00952990.2016.1197231. PMC 5156574. PMID 27436632.
  33. ^ Inagaki TK, Hazlett LI, Andreescu C (April 2020). "Opioids and social bonding: Effect of naltrexone on feelings of social connection and ventral striatum activity to close others". J Exp Psychol Gen. 149 (4): 732–745. doi:10.1037/xge0000674. PMC 7021584. PMID 31414860.
  34. ^ Inagaki TK, Hazlett LI, Andreescu C (May 2019). "Naltrexone alters responses to social and physical warmth: implications for social bonding". Soc Cogn Affect Neurosci. 14 (5): 471–479. doi:10.1093/scan/nsz026. PMC 6545530. PMID 30976797.
  35. ^ Inagaki TK, Ray LA, Irwin MR, Way BM, Eisenberger NI (May 2016). "Opioids and social bonding: naltrexone reduces feelings of social connection". Soc Cogn Affect Neurosci. 11 (5): 728–35. doi:10.1093/scan/nsw006. PMC 4847702. PMID 26796966.
  36. ^ Meier IM, Bos PA, Hamilton K, Stein DJ, van Honk J, Malcolm-Smith S (December 2016). "Naltrexone increases negatively-valenced facial responses to happy faces in female participants". Psychoneuroendocrinology. 74: 65–68. doi:10.1016/j.psyneuen.2016.08.022. PMID 27588701. S2CID 40097592.
  37. ^ O'Brien CP, Gastfriend DR, Forman RF, Schweizer E, Pettinati HM (2011). "Long-term opioid blockade and hedonic response: preliminary data from two open-label extension studies with extended-release naltrexone". Am J Addict. 20 (2): 106–12. doi:10.1111/j.1521-0391.2010.00107.x. PMC 3895092. PMID 21314752.
  38. ^ Mallik A, Chanda ML, Levitin DJ (February 2017). "Anhedonia to music and mu-opioids: Evidence from the administration of naltrexone". Sci Rep. 7: 41952. doi:10.1038/srep41952. PMC 5296903. PMID 28176798.
  39. ^ Laeng B, Garvija L, Løseth G, Eikemo M, Ernst G, Leknes S (April 2021). "'Defrosting' music chills with naltrexone: The role of endogenous opioids for the intensity of musical pleasure". Conscious Cogn. 90: 103105. doi:10.1016/j.concog.2021.103105. PMID 33711654. S2CID 232163311.
  40. ^ Raynor K, Kong H, Chen Y, Yasuda K, Yu L, Bell GI, Reisine T (February 1994). "Pharmacological characterization of the cloned kappa-, delta-, and mu-opioid receptors". Molecular Pharmacology. 45 (2): 330–4. PMID 8114680.
  41. ^ Codd EE, Shank RP, Schupsky JJ, Raffa RB (September 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.
  42. ^ Niciu MJ, Arias AJ (October 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.
  43. ^ Jump up to: a b c Wang D, Sun X, Sadee W (May 2007). "Different effects of opioid antagonists on mu-, delta-, and kappa-opioid receptors with and without agonist pretreatment". J Pharmacol Exp Ther. 321 (2): 544–52. doi:10.1124/jpet.106.118810. PMID 17267582. S2CID 28500012.
  44. ^ Jump up to: a b c de Laat B, Nabulsi N, Huang Y, O'Malley SS, Froehlich JC, Morris ED, Krishnan-Sarin S (June 2020). "Occupancy of the kappa opioid receptor by naltrexone predicts reduction in drinking and craving". Mol Psychiatry. doi:10.1038/s41380-020-0811-8. PMID 32541931. S2CID 219692020.
  45. ^ Jump up to: a b Toljan K, Vrooman B (September 2018). "Low-Dose Naltrexone (LDN)-Review of Therapeutic Utilization". Med Sci (Basel). 6 (4): 82. doi:10.3390/medsci6040082. PMC 6313374. PMID 30248938.
  46. ^ Bachtell R, Hutchinson MR, Wang X, Rice KC, Maier SF, Watkins LR (2015). "Targeting the Toll of Drug Abuse: The Translational Potential of Toll-Like Receptor 4". CNS Neurol Disord Drug Targets. 14 (6): 692–9. doi:10.2174/1871527314666150529132503. PMC 5548122. PMID 26022268.
  47. ^ Jump up to: a b Lee B, Elston DM (June 2019). "The uses of naltrexone in dermatologic conditions". J Am Acad Dermatol. 80 (6): 1746–1752. doi:10.1016/j.jaad.2018.12.031. PMID 30582992.
  48. ^ Zagon IS, Verderame MF, McLaughlin PJ (February 2002). "The biology of the opioid growth factor receptor (OGFr)". Brain Res Brain Res Rev. 38 (3): 351–76. doi:10.1016/s0165-0173(01)00160-6. PMID 11890982. S2CID 37812525.
  49. ^ Walker BM, Koob GF (February 2008). "Pharmacological evidence for a motivational role of kappa-opioid systems in ethanol dependence". Neuropsychopharmacology. 33 (3): 643–52. doi:10.1038/sj.npp.1301438. PMC 2739278. PMID 17473837.
  50. ^ Koob GF, Volkow ND (August 2016). "Neurobiology of addiction: a neurocircuitry analysis". The Lancet. Psychiatry. 3 (8): 760–73. doi:10.1016/S2215-0366(16)00104-8. PMC 6135092. PMID 27475769.
  51. ^ Koob GF, Buck CL, Cohen A, Edwards S, Park PE, Schlosburg JE, Schmeichel B, Vendruscolo LF, Wade CL, Whitfield TW, George O (January 2014). "Addiction as a stress surfeit disorder". Neuropharmacology. 76 Pt B: 370–82. doi:10.1016/j.neuropharm.2013.05.024. PMC 3830720. PMID 23747571.
  52. ^ Jump up to: a b Mannelli P, Peindl KS, Wu LT (June 2011). "Pharmacological enhancement of naltrexone treatment for opioid dependence: a review". Subst Abuse Rehabil. 2011 (2): 113–123. doi:10.2147/SAR.S15853. PMC 3128868. PMID 21731898.
  53. ^ Jump up to: a b Lee MC, Wagner HN, Tanada S, Frost JJ, Bice AN, Dannals RF (July 1988). "Duration of occupancy of opiate receptors by naltrexone". J Nucl Med. 29 (7): 1207–11. PMID 2839637.
  54. ^ Narcotic Antagonists: Naltrexone Pharmacochemistry and Sustained-release Preparations. Department of Health and Human Services, Public Health Service, Alcohol, Drug Abuse, and Mental Health Administration, National Institute on Drug Abuse, Division of Research. 1981. pp. 148–.
  55. ^ Ray LA, Barr CS, Blendy JA, Oslin D, Goldman D, Anton RF (March 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.
  56. ^ Garbutt JC, Greenblatt AM, West SL, Morgan LC, Kampov-Polevoy A, Jordan HS, Bobashev GV (August 2014). "Clinical and biological moderators of response to naltrexone in alcohol dependence: a systematic review of the evidence". Addiction. 109 (8): 1274–84. doi:10.1111/add.12557. PMID 24661324.
  57. ^ Jump up to: a b National Research Council (U.S.). Committee on Problems of Drug Dependence (1974). Report of the Thirty-sixth Annual Scientific Meeting: Committee on Problems of Drug Dependence, Mexico City, March 10-14, 1974. National Academies. pp. 265–. ISBN 9780309022446. NAP:13963.
  58. ^ Padwa H, Cunningham J (2010). Addiction: A Reference Encyclopedia. ABC-CLIO. pp. 207–. ISBN 978-1-59884-229-6.
  59. ^ G Bennett (14 January 2004). Treating Drug Abusers. Routledge. pp. 112–. ISBN 978-1-134-93173-6.
  60. ^ Jump up to: a b c Joseph Wouk (1 March 2009). Google Ldn !. Lulu.com. pp. 78–88. ISBN 978-0-578-00439-6.
  61. ^ Jump up to: a b Elks J (14 November 2014). The Dictionary of Drugs: Chemical Data: Chemical Data, Structures and Bibliographies. Springer. pp. 851–. ISBN 978-1-4757-2085-3.
  62. ^ Jump up to: a b Index Nominum 2000: International Drug Directory. Taylor & Francis. 2000. pp. 715–. ISBN 978-3-88763-075-1.
  63. ^ Jump up to: a b Morton IK, Hall JM (6 December 2012). Concise Dictionary of Pharmacological Agents: Properties and Synonyms. Springer Science & Business Media. pp. 189–. ISBN 978-94-011-4439-1.
  64. ^ Jump up to: a b c "Naltrexone". Archived from the original on 2017-12-04. Retrieved 2017-12-04.
  65. ^ "Bupropion and naltrexone Uses, Side Effects & Warnings". Drugs.com. 8 June 2020. Retrieved 16 September 2020.
  66. ^ "Morphine and naltrexone Uses, Side Effects & Warnings". Drugs.com. 14 October 2019. Retrieved 16 September 2020.
  67. ^ McCann DJ (April 2008). "Potential of buprenorphine/naltrexone in treating polydrug addiction and co-occurring psychiatric disorders". Clinical Pharmacology and Therapeutics. 83 (4): 627–30. doi:10.1038/sj.clpt.6100503. PMID 18212797. S2CID 21165673.
  68. ^ Armstrong W (7 May 2013). "A Shot in the Dark: Can Vivitrol Help Us Control Our Addictions?". Pacific Standard. Archived from the original on 13 September 2017.
  69. ^ Krupitsky E, Nunes EV, Ling W, Illeperuma A, Gastfriend DR, Silverman BL (April 2011). "Injectable extended-release naltrexone for opioid dependence: a double-blind, placebo-controlled, multicentre randomised trial". Lancet. 377 (9776): 1506–13. doi:10.1016/s0140-6736(11)60358-9. PMID 21529928. S2CID 16690413.
  70. ^ Wolfe D, Carrieri MP, Dasgupta N, Bruce D, Wodak A (2011). "Injectable extended-release naltrexone for opioid dependence – Authors' reply". The Lancet. 378 (9792): 666. doi:10.1016/S0140-6736(11)61333-0. S2CID 205963967.
  71. ^ Tanum L, Solli KK, Latif ZE, Benth JŠ, Opheim A, Sharma-Haase K, Krajci P, Kunøe N (December 2017). "Effectiveness of Injectable Extended-Release Naltrexone vs Daily Buprenorphine-Naloxone for Opioid Dependence: A Randomized Clinical Noninferiority Trial". JAMA Psychiatry. 74 (12): 1197–1205. doi:10.1001/jamapsychiatry.2017.3206. PMC 6583381. PMID 29049469.
  72. ^ Jump up to: a b c Goodnough A, Zernike K (June 11, 2017). "Seizing on Opioid Crisis, a Drug Maker Lobbies Hard for Its Product". The New York Times. Archived from the original on June 11, 2017. Retrieved June 11, 2017. Advertising for Vivitrol on a subway car in Brooklyn last month. Marketing for the drug has shifted into high gear.
  73. ^ Jump up to: a b "Letter to Tom Price". May 2017. Archived from the original on June 25, 2017. Retrieved June 11, 2017.
  74. ^ Jump up to: a b c d Simeon D, Abugel J (10 October 2008). Feeling Unreal: Depersonalization Disorder and the Loss of the Self. Oxford University Press. pp. 166–. ISBN 978-0-19-976635-2.
  75. ^ Jump up to: a b c d Lanius UF, Paulsen SL, Corrigan FM (13 May 2014). Neurobiology and Treatment of Traumatic Dissociation: Towards an Embodied Self. Springer Publishing Company. pp. 489–. ISBN 978-0-8261-0632-2.
  76. ^ Sierra M (January 2008). "Depersonalization disorder: pharmacological approaches". Expert Review of Neurotherapeutics. 8 (1): 19–26. doi:10.1586/14737175.8.1.19. PMID 18088198. S2CID 22180718.
  77. ^ Novella S (5 May 2010). "Low Dose Naltrexone – Bogus or Cutting Edge Science?". Science-Based Medicine. Archived from the original on 8 July 2011. Retrieved 5 July 2011.
  78. ^ "Low-Dose Naltrexone". National MS Society. Retrieved 12 May 2014.
  79. ^ Novella, Steven (5 May 2010). "Low Dose Naltrexone – Bogus or Cutting Edge Science?". Retrieved 5 July 2011.
  80. ^ Bowling AC. "Low-dose naltrexone (LDN) The "411" on LDN". National Multiple Sclerosis Society. Archived from the original on 22 December 2011. Retrieved 6 July 2011.
  81. ^ Smith SG, Gupta KK, Smith SH (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. S2CID 144215400.
  82. ^ Manley C (1998-03-20). "Self-injuries may have biochemical base: study". The Reporter. Archived from the original on 2009-01-05.
  83. ^ Grant JE, Kim SW, Odlaug BL (April 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. S2CID 22992128. Lay summaryScience Daily (April 3, 2009).
  84. ^ Clinical trial number NCT00326807 for "A Randomized, Double-Blind, Placebo-Controlled Trial of Naltrexone in the Treatment of Concurrent Alcohol Dependence and Pathological Gambling" at ClinicalTrials.gov
  85. ^ Kim SW, Grant JE, Adson DE, Shin YC (June 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. S2CID 22134798.
  86. ^ Bostwick JM, Bucci JA (February 2008). "Internet sex addiction treated with naltrexone". Mayo Clinic Proceedings. 83 (2): 226–30. doi:10.4065/83.2.226. PMID 18241634.
  87. ^ Vignau J, Karila L, Costisella O, Canva V (2005). "[Hepatitis C, interferon a and depression: main physiopathologic hypothesis]" [Hepatitis C, interferon a and depression: main physiopathologic hypothesis]. L'Encephale (in French). 31 (3): 349–57. doi:10.1016/s0013-7006(05)82400-5. PMID 16142050. INIST:16920336.
  88. ^ Małyszczak K, Inglot M, Pawłowski T, Czarnecki M, Rymer W, Kiejna A (2006). "[Neuropsychiatric symptoms related to interferon alpha]" [Neuropsychiatric symptoms related to interferon alpha]. Psychiatria Polska (in Polish). 40 (4): 787–97. PMID 17068950. Archived from the original on 2017-02-02.
  89. ^ Campbell ND (2013-03-28). "Why Can't They Stop?" A Highly Public Misunderstanding of Science. pp. 238–262. doi:10.1215/9780822395874-010. ISBN 978-0-8223-5350-8.
  90. ^ Vrecko S (2009-06-01). "Therapeutic Justice in Drug Courts: Crime, Punishment and Societies of Control". Science as Culture. 18 (2): 217–232. doi:10.1080/09505430902885623. S2CID 144197523.
  91. ^ Aleksanyan J (2020-05-09). "Governing beyond the closet: Remaking stigma, identity, and sexual behavior in a post-disciplinary rehab". Ethnography: 1466138120923702. doi:10.1177/1466138120923702.
  92. ^ Malandain, Leo; Blanc, Jean-Victor; Ferreri, Florian; Thibaut, Florence (June 2020). "Pharmacotherapy of Sexual Addiction". Current Psychiatry Reports. 22 (6): 30. doi:10.1007/s11920-020-01153-4. ISSN 1523-3812. PMID 32377953. S2CID 218527367.
  93. ^ Herron, Abigail J., Brennan, Tim K. eds. ASAM Essentials of Addiction Medicine, The. 3rd Edition. Two Commerce Square, 2001 Market Street, Philadelphia, PA 19103 USA:Lippincott Williams & Wilkins; 2020.

External links[]

  • "Naltrexone". Drug Information Portal. U.S. National Library of Medicine.
Retrieved from ""