Difference between revisions of "Ketamine"

From Self-sufficiency
Jump to: navigation, search
(History)
 
m (1 revision)
 
(4 intermediate revisions by 3 users not shown)
Line 1: Line 1:
 
{{For|the functional group referred to as ketimine|imine}}
 
{{For|the functional group referred to as ketimine|imine}}
 
{{drugbox
 
{{drugbox
| drug_name = Ketamine
+
| drug_name = ketamine
 
| Watchedfields = changed
 
| Watchedfields = changed
 
| verifiedrevid = 307101549
 
| verifiedrevid = 307101549
| IUPAC_name = (''RS'')-2-(2-Chlorophenyl)-2-(methylamino)-cyclohexan-1-one
+
| IUPAC_name = (''RS'')-2-(2-Chlorophenyl)-2-(methylamino)cyclohexan-1-one
 
| image = Ketamine.svg
 
| image = Ketamine.svg
 
| width = 150
 
| width = 150
Line 24: Line 24:
 
| melting_point = 262
 
| melting_point = 262
 
| melting_high = 263
 
| melting_high = 263
| bioavailability =  
+
| bioavailability =
| protein_bound =  
+
| protein_bound =
| metabolism = Hepatic, primarily by CYP3A4<ref>{{cite journal |author=Hijazi Y, Boulieu R |title=Contribution of CYP3A4, CYP2B6, and CYP2C9 isoforms to N-demethylation of ketamine in human liver microsomes |journal=Drug Metabolism and Disposition |volume=30 |issue=7 |pages=853–8 |year=2002 |month=July |pmid=12065445 |doi=10.1124/dmd.30.7.853}}</ref>
+
| metabolism = Hepatic, primarily by CYP3A4<ref>{{Cite journal|author=Hijazi Y, Boulieu R |title=Contribution of CYP3A4, CYP2B6, and CYP2C9 isoforms to N-demethylation of ketamine in human liver microsomes |journal=Drug Metabolism and Disposition |volume=30 |issue=7 |pages=853–8 |year=2002 |month=July |pmid=12065445 |doi=10.1124/dmd.30.7.853}}</ref>
 
| elimination_half-life = 2.5–3 hours.
 
| elimination_half-life = 2.5–3 hours.
 
| excretion = renal (>90%)
 
| excretion = renal (>90%)
Line 32: Line 32:
 
| pregnancy_US = <!-- A / B / C / D / X -->
 
| pregnancy_US = <!-- A / B / C / D / X -->
 
| pregnancy_category = B
 
| pregnancy_category = B
| legal_AU =  
+
| legal_AU = S8
 
| legal_CA = Schedule I
 
| legal_CA = Schedule I
 
| legal_UK = Class C
 
| legal_UK = Class C
| legal_US =  Schedule III  
+
| legal_US =  Schedule III
| legal_status =  
+
| legal_status =
 
| routes_of_administration =  [[Intravenous therapy|IV]], [[Intramuscular injection|IM]], [[Insufflation (medicine)|Insufflate]]d, oral, [[topical]]
 
| routes_of_administration =  [[Intravenous therapy|IV]], [[Intramuscular injection|IM]], [[Insufflation (medicine)|Insufflate]]d, oral, [[topical]]
 
}}
 
}}
 
{{Cleanup|date=March 2010}}
 
{{Cleanup|date=March 2010}}
  
'''Ketamine''' is a [[drug]] used in human and [[veterinary medicine]] developed by [[Parke-Davis]] (today a part of [[Pfizer]]) in 1962. Its [[hydrochloride]] [[salt (chemistry)|salt]] is sold as '''Ketanest''', '''Ketaset''', and '''Ketalar'''. [[pharmacology|Pharmacologically]], ketamine is classified as an [[NMDA receptor antagonist]].<ref name="harrison">{{cite journal |author=Harrison NL, Simmonds MA |title=Quantitative studies on some antagonists of N-methyl D-aspartate in slices of rat cerebral cortex |journal=British Journal of Pharmacology |volume=84 |issue=2 |pages=381–91 |year=1985 |month=February |pmid=2858237 |pmc=1987274}}</ref> At high, fully anesthetic level doses, ketamine has also been found to bind to [[opioid]] [[mu Opioid receptor|μ receptors]] and [[sigma receptor]]s.<ref>{{cite journal |author=Hirota K, Sikand KS, Lambert DG |title=Interaction of ketamine with mu2 opioid receptors in SH-SY5Y human neuroblastoma cells |journal=Journal of Anesthesia |volume=13 |issue=2 |pages=107–9 |year=1999 |pmid=14530949 |doi=10.1007/s005400050035}}</ref><ref>{{cite journal |author=Narita M, Yoshizawa K, Aoki K, Takagi M, Miyatake M, Suzuki T |title=A putative sigma1 receptor antagonist NE-100 attenuates the discriminative stimulus effects of ketamine in rats |journal=Addiction Biology |volume=6 |issue=4 |pages=373–376 |year=2001 |month=September |pmid=11900615 |doi=10.1080/13556210020077091}}</ref> Like other drugs of this class such as [[tiletamine]] and [[phencyclidine]] (PCP), it induces a state referred to as "[[dissociative drug|dissociative anesthesia]]"<ref>{{cite journal |author=Bergman SA |title=Ketamine: review of its pharmacology and its use in pediatric anesthesia |journal=Anesthesia Progress |volume=46 |issue=1 |pages=10–20 |year=1999 |pmid=10551055 |pmc=2148883}}</ref> and is used as a [[recreational drug]].
+
'''Ketamine''' is a [[drug]] used in human and [[veterinary medicine]] developed by [[Parke-Davis]] (today a part of [[Pfizer]]) in 1962. Its [[hydrochloride]] [[salt (chemistry)|salt]] is sold as '''Ketanest''', '''Ketaset''', and '''Ketalar'''. [[pharmacology|Pharmacologically]], ketamine is classified as an [[NMDA receptor antagonist]].<ref name="harrison">{{Cite journal|author=Harrison NL, Simmonds MA |title=Quantitative studies on some antagonists of N-methyl D-aspartate in slices of rat cerebral cortex |journal=British Journal of Pharmacology |volume=84 |issue=2 |pages=381–91 |year=1985 |month=February |pmid=2858237 |pmc=1987274}}</ref> At high, fully anesthetic level doses, ketamine has also been found to bind to [[opioid]] [[mu Opioid receptor|μ receptors]] and [[sigma receptor]]s.<ref>{{Cite journal|author=Hirota K, Sikand KS, Lambert DG |title=Interaction of ketamine with mu2 opioid receptors in SH-SY5Y human neuroblastoma cells |journal=Journal of Anesthesia |volume=13 |issue=2 |pages=107–9 |year=1999 |pmid=14530949 |doi=10.1007/s005400050035}}</ref><ref>{{Cite journal|author=Narita M, Yoshizawa K, Aoki K, Takagi M, Miyatake M, Suzuki T |title=A putative sigma1 receptor antagonist NE-100 attenuates the discriminative stimulus effects of ketamine in rats |journal=Addiction Biology |volume=6 |issue=4 |pages=373–376 |year=2001 |month=September |pmid=11900615 |doi=10.1080/13556210020077091}}</ref> Like other drugs of this class such as [[tiletamine]] and [[phencyclidine]] (PCP), it induces a state referred to as "[[dissociative drug|dissociative anesthesia]]"<ref>{{Cite journal|author=Bergman SA |title=Ketamine: review of its pharmacology and its use in pediatric anesthesia |journal=Anesthesia Progress |volume=46 |issue=1 |pages=10–20 |year=1999 |pmid=10551055 |pmc=2148883}}</ref> and is used as a [[recreational drug]].
  
Ketamine has a wide range of effects in humans, including [[analgesia]], [[anesthesia]], [[hallucination]]s, elevated [[blood pressure]], and [[bronchodilator|bronchodilation]].<ref>{{cite book |author=Peck TE, Hill SA, Williams M |year=2008 |page=111 |title=Pharmacology for anaesthesia and intensive care (3rd edition) |publisher=Cambridge university press |location=Cambridge |isbn=9780521704632}}</ref>  Ketamine is primarily used for the induction and maintenance of [[general anesthesia]], usually in combination with some sedative drug. Other uses include [[sedative|sedation]] in [[intensive care]], [[analgesia]] (particularly in emergency medicine), and treatment of [[bronchospasm]]. It is also a popular anesthetic in [[veterinary medicine]].
+
Ketamine has a wide range of effects in humans, including [[analgesia]], [[anesthesia]], [[hallucination]]s, elevated [[blood pressure]], and [[bronchodilator|bronchodilation]].<ref>{{Cite book|author=Peck TE, Hill SA, Williams M |year=2008 |page=111 |title=Pharmacology for anaesthesia and intensive care (3rd edition) |publisher=Cambridge university press |location=Cambridge |isbn=9780521704632}}</ref>  Ketamine is primarily used for the induction and maintenance of [[general anesthesia]], usually in combination with some sedative drug. Other uses include [[sedative|sedation]] in [[intensive care]], [[analgesia]] (particularly in emergency medicine), and treatment of [[bronchospasm]]. It has been shown to be effective in treating depression in patients with [[bipolar disorder]] who have not responded to other anti-depressants.<ref>{{Cite journal|author=Nancy Diazgranados et al |title= A Randomized Add-on Trial of an N-methyl-D-aspartate Antagonist in Treatment-Resistant Bipolar Depression|journal=Archives of General Psychiatry |volume=67 |issue=8 |pages=793–802 |year=2010 |month=August|pmid=20679587 |doi=10.1001/archgenpsychiatry.2010.90}}</ref> It is also a popular anesthetic in [[veterinary medicine]].
  
Ketamine is a [[chirality (chemistry)|chiral]] compound. Most pharmaceutical preparations of ketamine are [[racemic]]; however, some brands reportedly have (mostly undocumented) differences in [[enantiomer]]ic proportions. The more active enantiomer, (''S'')-ketamine, is also available for medical use under the brand name '''Ketanest S'''.<ref>{{cite journal |author=Krüger AD |title=[Current aspects of using ketamine in childhood] |language=German |journal=Anaesthesiologie Und Reanimation |volume=23 |issue=3 |pages=64–71 |year=1998 |pmid=9707751}}</ref> Ketamine is a ''core'' medicine in the [[World Health Organization]]'s "[[WHO Model List of Essential Medicines|Essential Drugs List]]", which is a list of minimum medical needs for a basic health care system.<ref name="essentialWHO">{{cite web
+
Ketamine is a [[chirality (chemistry)|chiral]] compound. Most pharmaceutical preparations of ketamine are [[racemic]]; however, some brands reportedly have (mostly undocumented) differences in [[enantiomer]]ic proportions. The more active enantiomer, [[Esketamine|(''S'')-ketamine]], is also available for medical use under the brand name '''Ketanest S'''.<ref>{{Cite journal|author=Krüger AD |title=[Current aspects of using ketamine in childhood] |language=German |journal=Anaesthesiologie Und Reanimation |volume=23 |issue=3 |pages=64–71 |year=1998 |pmid=9707751}}</ref> However (''R'')-ketamine is also active, and has differences in effects; the S enantiomer being a dissociative and the R enantiomer a hallucinogen. (''R'')-ketamine, (''S'')-ketamine & racemic (''R,S'')-ketamine all have qualitatively separate distinct effect profiles, although S has the most active potency. Ketamine is a ''core'' medicine in the [[World Health Organization]]'s "[[WHO Model List of Essential Medicines|Essential Drugs List]]", which is a list of minimum medical needs for a basic health care system.<ref name="essentialWHO">{{Cite web
 
  | month = March | year = 2005
 
  | month = March | year = 2005
 
  | url = http://whqlibdoc.who.int/hq/2005/a87017_eng.pdf
 
  | url = http://whqlibdoc.who.int/hq/2005/a87017_eng.pdf
Line 55: Line 55:
  
 
==History==
 
==History==
Ketamine was discovered by Dr. Craig Newlands of [[Wayne State University]]. It was then developed by Parke-Davis in 1962 as part of an effort to find a safer anesthetic alternative to [[phencyclidine]] (PCP), which was likely to cause [[hallucination]]s, [[neurotoxicity]] and [[seizure]]s. The drug was first given to [[United States|American]] soldiers during the [[Vietnam War]]. It is still widely used in humans. It is also used widely in [[veterinary medicine]], or as a [[battlefield medicine|battlefield]] anesthetic in developing nations.<ref name="bonanno">{{cite journal |author=Bonanno FG |title=Ketamine in war/tropical surgery (a final tribute to the racemic mixture) |journal=Injury |volume=33 |issue=4 |pages=323–7 |year=2002 |month=May |pmid=12091028 |doi=10.1016/S0020-1383(01)00209-1}}</ref>
+
Ketamine was discovered by Dr. Craig Newlands of [[Wayne State University]]. It was then developed by Parke-Davis in 1962 as part of an effort to find a safer anesthetic alternative to [[phencyclidine]] (PCP), which was likely to cause [[hallucination]]s, [[neurotoxicity]] and [[seizure]]s. The drug was first given to [[United States|American]] soldiers during the [[Vietnam War]]. It is still widely used in humans. It is also used widely in [[veterinary medicine]], or as a [[battlefield medicine|battlefield]] anesthetic in developing nations.<ref name="bonanno">{{Cite journal|author=Bonanno FG |title=Ketamine in war/tropical surgery (a final tribute to the racemic mixture) |journal=Injury |volume=33 |issue=4 |pages=323–7 |year=2002 |month=May |pmid=12091028 |doi=10.1016/S0020-1383(01)00209-1}}</ref>
The drug was used in [[psychiatry|psychiatric]] and other academic research through the 1970s, culminating in 1978 with the publishing of [[John C. Lilly|John Lilly]]'s ''The Scientist'' and [[Marcia Moore]] and Howard Alltounian's ''Journeys into the Bright World'', which documented the unusual phenomenology of ketamine intoxication.<ref>{{cite book |author=Alltounian, Howard Sunny; Marcia Moore |title=Journeys into the bright world |publisher=Para Research |location=Rockport, Mass |year=1978 |pages= |isbn=0-914918-12-5}}{{pn}}</ref>
+
The drug was used in [[psychiatry|psychiatric]] and other academic research through the 1970s, culminating in 1978 with the publishing of [[John C. Lilly|John Lilly]]'s ''The Scientist'' and [[Marcia Moore]] and Howard Alltounian's ''Journeys into the Bright World'', which documented the unusual phenomenology of ketamine intoxication.<ref>{{Cite book|author=Alltounian, Howard Sunny; Marcia Moore |title=Journeys into the bright world |publisher=Para Research |location=Rockport, Mass |year=1978 |pages= |isbn=0-914918-12-5}}{{Page needed|date=September 2010}}</ref>
  
The incidence of [[recreational drug use|recreational]] ketamine use increased through the end of the century, especially in the context of [[rave party|raves]] and other parties. The increase in illicit use prompted ketamine's placement in Schedule III of the [[United States]] [[Controlled Substance Act]] in August 1999.<ref name="csa">{{cite web | url=http://www.anestesia.com.mx/articulo/keta.html | title=Ketamine - Schedule III of The Controlled Substances Act (CSA) | author= |date= | publisher= Anestesiología Mexicana en Internet | accessdate=2006-12-22 }}</ref>
+
The incidence of [[recreational drug use|recreational]] ketamine use increased through the end of the century, especially in the context of [[rave party|raves]] and other parties. The increase in illicit use prompted ketamine's placement in Schedule III of the [[United States]] [[Controlled Substance Act]] in August 1999.<ref name="csa">{{Cite web| url=http://www.anestesia.com.mx/articulo/keta.html | title=Ketamine - Schedule III of The Controlled Substances Act (CSA) | author= |date= | publisher= Anestesiología Mexicana en Internet | accessdate=2006-12-22 }}</ref>
In the [[United Kingdom]], it became outlawed and labeled a [[Misuse of Drugs Act 1971|Class C drug]] on 1 January 2006.<ref>{{cite news| url=http://news.bbc.co.uk/2/hi/uk_news/4564606.stm | work=BBC News | title=Club 'horse' drug to be outlawed | date=28 December 2005 | accessdate=7 May 2010}}</ref> In [[Canada]] ketamine is classified as a Schedule I narcotic, as of August 2005.<ref>[http://laws.justice.gc.ca/en/C-38.8/229687.html#rid-229689 Controlled Drugs and Substances Act<!-- Bot generated title -->]</ref> In [[Hong Kong]], as of year 2000, ketamine is regulated under Schedule 1 of Hong Kong Chapter 134 ''Dangerous Drugs Ordinance''. It can only be used legally by health professionals, for university research purposes, or with a physician's prescription.<ref>[http://www.info.gov.hk/gia/general/200011/22/1122170.htm Government to tighten control on Ketamine<!-- Bot generated title -->]</ref> Article 332 Nicaraguan Law, "the use of the controlled analgesic substance Ketamine, may only be used by official veterinaries, selenisticans, paulinothlogists and jakfonries". Intriguingly, the law also states, that only in the province of "huesos perezosos" (literally translated as "Lazy Bones"), may this substance be used in the aforementioned context.
+
In the [[United Kingdom]], it became outlawed and labeled a [[Misuse of Drugs Act 1971|Class C drug]] on 1 January 2006.<ref>{{Cite news| url=http://news.bbc.co.uk/2/hi/uk_news/4564606.stm | work=BBC News | title=Club 'horse' drug to be outlawed | date=28 December 2005 | accessdate=7 May 2010}}</ref> In [[Canada]] ketamine is classified as a Schedule I narcotic, as of August 2005.<ref>[http://laws.justice.gc.ca/en/C-38.8/229687.html#rid-229689 Controlled Drugs and Substances Act<!-- Bot generated title -->]</ref> In [[Hong Kong]], as of year 2000, ketamine is regulated under Schedule 1 of Hong Kong Chapter 134 ''Dangerous Drugs Ordinance''. It can only be used legally by health professionals, for university research purposes, or with a physician's prescription.<ref>[http://www.info.gov.hk/gia/general/200011/22/1122170.htm Government to tighten control on Ketamine<!-- Bot generated title -->]</ref>
  
 
==Naming==
 
==Naming==
 
<!-- Please do not add names without a reliable source -->
 
<!-- Please do not add names without a reliable source -->
Ketamine was invented as CL369, and it was referred to as CI 581 (clinical investigation 581) during development at Parke-Davis.<ref name="Jansen23">{{cite book|title=Ketamine: Dreams and Realities|last=Jansen|first=Karl|publisher=Multidisciplinary Association for Psychedelic Studies|isbn=0966001931|year=2001|page=23}}</ref> Commercial brands of ketamine include "Ketalar, Ketaset, Ketmex, Ketotal, Ketamine-500 (Astrapin) and Imalgen".<ref name="Jansen38">{{cite book|title=Ketamine: Dreams and Realities|last=Jansen|first=Karl|publisher=Multidisciplinary Association for Psychedelic Studies|isbn=0966001931|year=2001|page=38}}</ref> Ketamine is a keto-amine, so named as it has a ketone C=O group bonded to carbons either side, as well as an amine group. As such it resembles NMDA.
+
Ketamine was invented as CL369, and it was referred to as CI 581 (clinical investigation 581) during development at Parke-Davis.<ref name="Jansen23">{{Cite book|title=Ketamine: Dreams and Realities|last=Jansen|first=Karl|publisher=Multidisciplinary Association for Psychedelic Studies|isbn=0966001931|year=2001|page=23}}</ref> Commercial brands of ketamine include "Ketalar, Ketaset, Ketmex, Ketotal, Ketamine-500 (Astrapin) and Imalgen".<ref name="Jansen38">{{Cite book|title=Ketamine: Dreams and Realities|last=Jansen|first=Karl|publisher=Multidisciplinary Association for Psychedelic Studies|isbn=0966001931|year=2001|page=38}}</ref> Ketamine is a keto-amine, so named as it has a ketone C=O group bonded to carbons either side, as well as an amine group. As such it resembles NMDA.
  
Production for recreational use has been traced to 1967, when it was referred to as "mean green" and "rockmesc".<ref name="Jansen24">{{cite book|title=Ketamine: Dreams and Realities|last=Jansen|first=Karl|publisher=Multidisciplinary Association for Psychedelic Studies|isbn=0966001931|year=2001|page=24}}</ref> Recreational names for ketamine include "K",<ref name="Jansen26">{{cite book|title=Ketamine: Dreams and Realities|last=Jansen|first=Karl|publisher=Multidisciplinary Association for Psychedelic Studies|isbn=0966001931|year=2001|page=26}}</ref> "Ket",<ref name="Jansen55">{{cite book|title=Ketamine: Dreams and Realities|last=Jansen|first=Karl|publisher=Multidisciplinary Association for Psychedelic Studies|isbn=0966001931|year=2001|page=55}}</ref> "Special K"<ref name="Jansen26" />  and "Vitamin K".<ref name="Jansen55" /> and "Kandado" (adapted from the Spanish word, "candado" meaning padlock, used in South and Central America)
+
Production for recreational use has been traced to 1967, when it was referred to as "mean green" and "rockmesc".<ref name="Jansen24">{{Cite book|title=Ketamine: Dreams and Realities|last=Jansen|first=Karl|publisher=Multidisciplinary Association for Psychedelic Studies|isbn=0966001931|year=2001|page=24}}</ref> Recreational names for ketamine include "K",<ref name="Jansen26">{{Cite book|title=Ketamine: Dreams and Realities|last=Jansen|first=Karl|publisher=Multidisciplinary Association for Psychedelic Studies|isbn=0966001931|year=2001|page=26}}</ref> "Ket",<ref name="Jansen55">{{Cite book|title=Ketamine: Dreams and Realities|last=Jansen|first=Karl|publisher=Multidisciplinary Association for Psychedelic Studies|isbn=0966001931|year=2001|page=55}}</ref> "Special K"<ref name="Jansen26" />  and "Vitamin K".<ref name="Jansen55" />
  
 
==Medical use==
 
==Medical use==
Line 73: Line 73:
 
* Pediatric anesthesia (as the sole anesthetic for minor procedures or as an induction agent followed by muscle relaxant and endotracheal intubation)
 
* Pediatric anesthesia (as the sole anesthetic for minor procedures or as an induction agent followed by muscle relaxant and endotracheal intubation)
 
* Asthmatics or patients with chronic obstructive airway disease
 
* Asthmatics or patients with chronic obstructive airway disease
* In [[emergency medicine]] in entrapped patients suffering severe trauma<ref>{{cite journal |author=Cottingham R, Thomson K |title=Use of ketamine in prolonged entrapment |journal=Journal of Accident & Emergency Medicine |volume=11 |issue=3 |pages=189–91 |year=1994 |month=September |pmid=7804588 |pmc=1342430 |doi=10.1136/emj.11.3.189}}</ref>
+
* As part of a cream, gel, or liquid for topical application for nerve pain—the most common mixture is 10% [[ketoprofen]], 5% [[Lidocaine]], and 10% ketamine. Other ingredients found useful by pain specialists and their patients as well as the compounding pharmacists who make the topical mixtures include [[amitryptiline]], [[cyclobenzaprine]], [[clonidine]], [[tramadol]], and [[mepivicaine]] and other longer-acting local anaesthetics.
 +
* In [[emergency medicine]] in entrapped patients suffering severe trauma<ref>{{Cite journal|author=Cottingham R, Thomson K |title=Use of ketamine in prolonged entrapment |journal=Journal of Accident & Emergency Medicine |volume=11 |issue=3 |pages=189–91 |year=1994 |month=September |pmid=7804588 |pmc=1342430 |doi=10.1136/emj.11.3.189}}</ref>
 
* Emergency surgery in field conditions in war zones
 
* Emergency surgery in field conditions in war zones
 
* To supplement spinal / epidural anesthesia / analgesia utilizing low doses
 
* To supplement spinal / epidural anesthesia / analgesia utilizing low doses
Line 83: Line 84:
  
 
===Effects===
 
===Effects===
Impairs all senses, especially:  
+
Impairs all senses, especially:
 
*Sight
 
*Sight
 
*Balance
 
*Balance
Line 103: Line 104:
 
*Partial depressant/stimulant
 
*Partial depressant/stimulant
  
In medical settings, ketamine is usually injected intravenously or intramuscularly,<ref name="lankenau">{{cite journal |author=Lankenau SE, Sanders B, Bloom JJ, ''et al.'' |title=First injection of ketamine among young injection drug users (IDUs) in three U.S. cities |journal=Drug and Alcohol Dependence |volume=87 |issue=2-3 |pages=183–93 |year=2007 |month=March |pmid=16979848 |pmc=1852477 |doi=10.1016/j.drugalcdep.2006.08.015}}</ref> but it is also effective when [[insufflation (medicine)|insufflated]], smoked, or taken orally.<ref name="reboso">{{cite journal |author=Reboso Morales JA, González Miranda F |title=[Ketamine] |language=Spanish |journal=Revista Española de Anestesiología y Reanimación |volume=46 |issue=3 |pages=111–22 |year=1999 |month=March |pmid=10228376}}</ref>
+
In medical settings, ketamine is usually injected intravenously or intramuscularly,<ref name="lankenau">{{Cite journal|author=Lankenau SE, Sanders B, Bloom JJ, ''et al.'' |title=First injection of ketamine among young injection drug users (IDUs) in three U.S. cities |journal=Drug and Alcohol Dependence |volume=87 |issue=2-3 |pages=183–93 |year=2007 |month=March |pmid=16979848 |pmc=1852477 |doi=10.1016/j.drugalcdep.2006.08.015}}</ref> but it is also effective when [[insufflation (medicine)|insufflated]], smoked, or taken orally.<ref name="reboso">{{Cite journal|author=Reboso Morales JA, González Miranda F |title=[Ketamine] |language=Spanish |journal=Revista Española de Anestesiología y Reanimación |volume=46 |issue=3 |pages=111–22 |year=1999 |month=March |pmid=10228376}}</ref>
  
Since it suppresses breathing much less than most other available anaesthetics,<ref name="heshmati">{{cite journal |author=Heshmati F, Zeinali MB, Noroozinia H, Abbacivash R, Mahoori A |title=Use of ketamine in severe status asthmaticus in intensive care unit |journal=Iranian Journal of Allergy, Asthma, and Immunology |volume=2 |issue=4 |pages=175–80 |year=2003 |month=December |pmid=17301376 |doi=02.04/ijaai.175180 |doi_brokendate=2009-12-13}}</ref> ketamine is still used in human medicine as an anesthetic, however, due to the hallucinations which may be caused by ketamine, it is not typically used as a primary anesthetic, although it is the anaesthetic of choice when reliable ventilation equipment is not available.  Ketamine tends to increase heart rate and blood pressure.  Because ketamine tends to increase or maintain cardiac output, it is sometimes used in anesthesia for emergency surgery when the patient's fluid volume status is unknown (''e.g.'', from traffic accidents). Ketamine can be used in [[podiatry]] and other minor surgery, and occasionally for the treatment of migraine. There is ongoing research in France, the Netherlands, Russia, Australia and the US into the drug's usefulness in pain therapy, depression suppression, and for the treatment of [[alcoholism]]<ref>{{cite journal |author=Krystal JH, Madonick S, Perry E, ''et al.'' |title=Potentiation of low dose ketamine effects by naltrexone: potential implications for the pharmacotherapy of alcoholism |journal=Neuropsychopharmacology |volume=31 |issue=8 |pages=1793–800 |year=2006 |month=August |pmid=16395307 |doi=10.1038/sj.npp.1300994}}</ref> and [[heroin]] [[Substance use disorder|addiction]].<ref name="jovaisa">{{cite journal |author=Jovaisa T, Laurinenas G, Vosylius S, Sipylaite J, Badaras R, Ivaskevicius J |title=Effects of ketamine on precipitated opiate withdrawal |journal=Medicina |volume=42 |issue=8 |pages=625–34 |year=2006 |pmid=16963828 |url=http://medicina.kmu.lt/0608/0608-03e.htm}}</ref>
+
Since it suppresses breathing much less than most other available anaesthetics,<ref name="heshmati">{{Cite journal|author=Heshmati F, Zeinali MB, Noroozinia H, Abbacivash R, Mahoori A |title=Use of ketamine in severe status asthmaticus in intensive care unit |journal=Iranian Journal of Allergy, Asthma, and Immunology |volume=2 |issue=4 |pages=175–80 |year=2003 |month=December |pmid=17301376}}</ref> ketamine is still used in human medicine as an anesthetic, however, due to the hallucinations which may be caused by ketamine, it is not typically used as a primary anesthetic, although it is the anaesthetic of choice when reliable ventilation equipment is not available.  Ketamine tends to increase heart rate and blood pressure.  Because ketamine tends to increase or maintain cardiac output, it is sometimes used in anesthesia for emergency surgery when the patient's fluid volume status is unknown (''e.g.'', from traffic accidents). Ketamine can be used in [[podiatry]] and other minor surgery, and occasionally for the treatment of migraine. There is ongoing research in France, the Netherlands, Russia, Australia and the US into the drug's usefulness in pain therapy, depression suppression, and for the treatment of [[alcoholism]]<ref>{{Cite journal|author=Krystal JH, Madonick S, Perry E, ''et al.'' |title=Potentiation of low dose ketamine effects by naltrexone: potential implications for the pharmacotherapy of alcoholism |journal=Neuropsychopharmacology |volume=31 |issue=8 |pages=1793–800 |year=2006 |month=August |pmid=16395307 |doi=10.1038/sj.npp.1300994}}</ref> and [[heroin]] [[Substance use disorder|addiction]].<ref name="jovaisa">{{Cite journal|author=Jovaisa T, Laurinenas G, Vosylius S, Sipylaite J, Badaras R, Ivaskevicius J |title=Effects of ketamine on precipitated opiate withdrawal |journal=Medicina |volume=42 |issue=8 |pages=625–34 |year=2006 |pmid=16963828 |url=http://medicina.kmu.lt/0608/0608-03e.htm}}</ref>
  
 
In [[veterinary anesthesia]], ketamine is often used for its anesthetic and [[analgesic]] effects on cats, dogs, [[rabbit]]s, [[rat]]s, and other small animals. Veterinarians often use ketamine with sedative drugs to produce balanced anesthesia and analgesia, and as a constant rate infusion to help prevent [[pain wind-up]]. Ketamine is used to manage pain among large animals, though it has less effect on [[bovine]]s. It is the primary intravenous [[anesthetic]] agent used in equine surgery, often in conjunction with [[detomidine]] and [[thiopental]], or sometimes [[guaifenesin]].
 
In [[veterinary anesthesia]], ketamine is often used for its anesthetic and [[analgesic]] effects on cats, dogs, [[rabbit]]s, [[rat]]s, and other small animals. Veterinarians often use ketamine with sedative drugs to produce balanced anesthesia and analgesia, and as a constant rate infusion to help prevent [[pain wind-up]]. Ketamine is used to manage pain among large animals, though it has less effect on [[bovine]]s. It is the primary intravenous [[anesthetic]] agent used in equine surgery, often in conjunction with [[detomidine]] and [[thiopental]], or sometimes [[guaifenesin]].
  
Ketamine may be used in small doses (0.1–0.5&nbsp;mg/kg·h) as a local anesthetic, particularly for the treatment of pain associated with movement and [[neuropathic pain]].<ref>{{cite journal |author=Lynch ME, Clark AJ, Sawynok J, Sullivan MJ |title=Topical amitriptyline and ketamine in neuropathic pain syndromes: an open-label study |journal=The Journal of Pain |volume=6 |issue=10 |pages=644–9 |year=2005 |month=October |pmid=16202956 |doi=10.1016/j.jpain.2005.04.008}}</ref> It may also be used as an intravenous co-analgesic together with opiates to manage otherwise intractable pain, particularly if this pain is neuropathic (pain due to vascular insufficiency or shingles are good examples).  It has the added benefit of counter-acting [[spinal sensitization]] or wind-up phenomena experienced with [[chronic pain]]. At these doses, the [[psychoactive drug|psychotropic]] side effects are less apparent and well managed with [[benzodiazepine]]s.<ref name="elia">{{cite journal |author=Elia N, Tramèr MR |title=Ketamine and postoperative pain--a quantitative systematic review of randomised trials |journal=Pain |volume=113 |issue=1-2 |pages=61–70 |year=2005 |month=January |pmid=15621365 |doi=10.1016/j.pain.2004.09.036}}</ref> Ketamine is a co-analgesic, and so is most effective when used alongside a low-dose [[opioid]]; while it does have analgesic effects by itself, the higher doses required can cause disorienting side effects.<ref name="elia" /> The combination of ketamine with an opioid is, however, particularly useful for pain caused by cancer.<ref>{{cite journal |author=Saito O, Aoe T, Kozikowski A, Sarva J, Neale JH, Yamamoto T |title=Ketamine and N-acetylaspartylglutamate peptidase inhibitor exert analgesia in bone cancer pain |journal=Canadian Journal of Anaesthesia |volume=53 |issue=9 |pages=891–8 |year=2006 |month=September |pmid=16960267}}</ref>
+
Ketamine may be used in small doses (0.1–0.5&nbsp;mg/kg·h) as a local anesthetic, particularly for the treatment of pain associated with movement and [[neuropathic pain]].<ref>{{Cite journal|author=Lynch ME, Clark AJ, Sawynok J, Sullivan MJ |title=Topical amitriptyline and ketamine in neuropathic pain syndromes: an open-label study |journal=The Journal of Pain |volume=6 |issue=10 |pages=644–9 |year=2005 |month=October |pmid=16202956 |doi=10.1016/j.jpain.2005.04.008}}</ref> It may also be used as an intravenous co-analgesic together with opiates to manage otherwise intractable pain, particularly if this pain is neuropathic (pain due to vascular insufficiency or shingles are good examples).  It has the added benefit of counter-acting [[spinal sensitization]] or wind-up phenomena experienced with [[chronic pain]]. At these doses, the [[psychoactive drug|psychotropic]] side effects are less apparent and well managed with [[benzodiazepine]]s.<ref name="elia">{{Cite journal|author=Elia N, Tramèr MR |title=Ketamine and postoperative pain--a quantitative systematic review of randomised trials |journal=Pain |volume=113 |issue=1-2 |pages=61–70 |year=2005 |month=January |pmid=15621365 |doi=10.1016/j.pain.2004.09.036}}</ref> Ketamine is a co-analgesic, and so is most effective when used alongside a low-dose [[opioid]]; while it does have analgesic effects by itself, the higher doses required can cause disorienting side effects.<ref name="elia" /> The combination of ketamine with an opioid is, however, particularly useful for pain caused by cancer.<ref>{{Cite journal|author=Saito O, Aoe T, Kozikowski A, Sarva J, Neale JH, Yamamoto T |title=Ketamine and N-acetylaspartylglutamate peptidase inhibitor exert analgesia in bone cancer pain |journal=Canadian Journal of Anaesthesia |volume=53 |issue=9 |pages=891–8 |year=2006 |month=September |pmid=16960267}}</ref>
  
The effect of ketamine on the [[respiratory system|respiratory]] and [[circulatory system|circulatory]] systems is different from that of other anesthetics. When used at anesthetic doses, it will usually stimulate rather than depress the circulatory system.<ref>{{cite journal |author=Adams HA |title=[S-(+)-ketamine. Circulatory interactions during total intravenous anesthesia and analgesia-sedation] |language=German |journal=Der Anaesthesist |volume=46 |issue=12 |pages=1081–7 |year=1997 |month=December |pmid=9451493 |doi=10.1007/s001010050510}}</ref> It is sometimes possible to perform ketamine anesthesia without protective measures to the airways. Ketamine is also a potent [[analgesic]] and can be used in sub-anesthetic doses to relieve acute pain; however, its [[psychotropic]] properties must be taken into account. Patients have reported vivid hallucinations, "going into other worlds" or "seeing [[God]]" while anesthetized, and these unwanted psychological side-effects have reduced the use of ketamine in human medicine. They can, however, usually be avoided by concomitant application of a sedative such as a [[benzodiazepine]].<ref name="elia" />
+
The effect of ketamine on the [[respiratory system|respiratory]] and [[circulatory system|circulatory]] systems is different from that of other anesthetics. When used at anesthetic doses, it will usually stimulate rather than depress the circulatory system.<ref>{{Cite journal|author=Adams HA |title=[S-(+)-ketamine. Circulatory interactions during total intravenous anesthesia and analgesia-sedation] |language=German |journal=Der Anaesthesist |volume=46 |issue=12 |pages=1081–7 |year=1997 |month=December |pmid=9451493 |doi=10.1007/s001010050510}}</ref> It is sometimes possible to perform ketamine anesthesia without protective measures to the airways. Ketamine is also a potent [[analgesic]] and can be used in sub-anesthetic doses to relieve acute pain; however, its [[psychotropic]] properties must be taken into account. Patients have reported vivid hallucinations, "going into other worlds" or "seeing [[God]]" while anesthetized, and these unwanted psychological side-effects have reduced the use of ketamine in human medicine. They can, however, usually be avoided by concomitant application of a sedative such as a [[benzodiazepine]].<ref name="elia" />
  
Low-dose ketamine is recognized for its potential effectiveness in the treatment of [[complex regional pain syndrome]] ([[Complex regional pain syndrome|CRPS]]), according to a retrospective review published in the October 2004 issue of Pain Medicine.<ref>{{cite journal |author=Correll GE, Maleki J, Gracely EJ, Muir JJ, Harbut RE |title=Subanesthetic ketamine infusion therapy: a retrospective analysis of a novel therapeutic approach to complex regional pain syndrome |journal=Pain Medicine |volume=5 |issue=3 |pages=263–75 |year=2004 |month=September |pmid=15367304 |doi=10.1111/j.1526-4637.2004.04043.x}}</ref> Although low-dose ketamine therapy is established as a generally safe procedure, reported side effects in some patients have included hallucinations, dizziness, lightheadedness and nausea. Therefore nurses administering ketamine to patients with CRPS should only do so in a setting where a trained physician is available if needed to assess potential adverse effects on patients.<ref>{{cite press release |title=Severe Pain Relief may be Possible with a Common Anesthetic Drug |publisher=[[Blackwell Publishing]] |date=1 October 2004 |url=http://blackwellpublishing.com/press/pressitem.asp?ref=180 |accessdate=13 December 2009}}</ref>
+
Low-dose ketamine is recognized for its potential effectiveness in the treatment of [[complex regional pain syndrome]] ([[Complex regional pain syndrome|CRPS]]), according to a retrospective review published in the October 2004 issue of Pain Medicine.<ref>{{Cite journal|author=Correll GE, Maleki J, Gracely EJ, Muir JJ, Harbut RE |title=Subanesthetic ketamine infusion therapy: a retrospective analysis of a novel therapeutic approach to complex regional pain syndrome |journal=Pain Medicine |volume=5 |issue=3 |pages=263–75 |year=2004 |month=September |pmid=15367304 |doi=10.1111/j.1526-4637.2004.04043.x}}</ref> Although low-dose ketamine therapy is established as a generally safe procedure, reported side effects in some patients have included hallucinations, dizziness, lightheadedness and nausea. Therefore nurses administering ketamine to patients with CRPS should only do so in a setting where a trained physician is available if needed to assess potential adverse effects on patients.<ref>{{cite press release |title=Severe Pain Relief may be Possible with a Common Anesthetic Drug |publisher=[[Blackwell Publishing]] |date=1 October 2004 |url=http://blackwellpublishing.com/press/pressitem.asp?ref=180 |accessdate=13 December 2009}}</ref>
  
In some neurological ICUs, ketamine has been used in cases of prolonged [[status epilepticus]]. There has been some evidence that the NMDA-blocking effect of the drug protects neurons from glutamatergic damage during prolonged seizures.<ref>{{cite journal |author=Fujikawa DG |title=Neuroprotective Effect of Ketamine Administered After Status Epilepticus Onset |journal=Epilepsia |volume=36 |issue=2 |pages=186–195 |year=1995 |doi=10.1111/j.1528-1157.1995.tb00979.x}}</ref>
+
In some neurological ICUs, ketamine has been used in cases of prolonged [[status epilepticus]]. There has been some evidence that the NMDA-blocking effect of the drug protects neurons from glutamatergic damage during prolonged seizures.<ref>{{Cite journal|author=Fujikawa DG |title=Neuroprotective Effect of Ketamine Administered After Status Epilepticus Onset |journal=Epilepsia |volume=36 |issue=2 |pages=186–195 |year=1995 |pmid=7821277 |doi=10.1111/j.1528-1157.1995.tb00979.x}}</ref>
  
 
====Experimental antidepressant use====
 
====Experimental antidepressant use====
Line 121: Line 122:
 
When treating patients suffering from [[complex regional pain syndrome]] (CRPS) with a low-dose (subanesthetic) ketamine infusion, it was observed that some patients made a significant recovery from associated [[clinical depression|depression]]. This recovery was not formally documented, as the primary concern was the treatment of the patient's pain. It was not possible to quantify to what degree depression recovery was secondary to the patient's recovery from CRPS. Based on this result, it was thought that a low-dose (subanesthetic) infusion of ketamine was worth a trial in patients who were suffering from treatment-resistant depression without other physical or psychiatric illness.
 
When treating patients suffering from [[complex regional pain syndrome]] (CRPS) with a low-dose (subanesthetic) ketamine infusion, it was observed that some patients made a significant recovery from associated [[clinical depression|depression]]. This recovery was not formally documented, as the primary concern was the treatment of the patient's pain. It was not possible to quantify to what degree depression recovery was secondary to the patient's recovery from CRPS. Based on this result, it was thought that a low-dose (subanesthetic) infusion of ketamine was worth a trial in patients who were suffering from treatment-resistant depression without other physical or psychiatric illness.
  
Correll, ''et al.'' gave ketamine intravenously to patients commencing at 15–20&nbsp;mg/h (0.1–0.2&nbsp;mg/kg/h) and the dose increased until a maximum tolerated dose was achieved. This dose was assumed to be a therapeutic dose and was maintained for 5 days. Patients were able to eat, drink, watch television, or read. They could feel inebriated and/or unsteady when walking. If hallucinations occurred, the dose was to be reduced. The patients' normal medications were continued as it was feared that stopping them might result in severe depressive episodes. Before and following each treatment with ketamine, at patient clinic visits, the [[Beck Depression Inventory]] (BDI) and the [[Hamilton Rating Scale for Depression]] (HAMD-17) were obtained. Two of the patients were described with "impressive", as described by an attending doctor, improvement in depression being maintained for 12 months in patient A and recurrence at 2.5 months and 9 months in patient B.<ref name="pmid16533209">{{cite journal |author=Correll GE, Futter GE |title=Two case studies of patients with major depressive disorder given low-dose (subanesthetic) ketamine infusions |journal=Pain Medicine |volume=7 |issue=1 |pages=92–5 |year=2006 |pmid=16533209 |doi=10.1111/j.1526-4637.2006.00101.x}}</ref>
+
Correll, ''et al.'' gave ketamine intravenously to patients commencing at 15–20&nbsp;mg/h (0.1–0.2&nbsp;mg/kg/h) and the dose increased until a maximum tolerated dose was achieved. This dose was assumed to be a therapeutic dose and was maintained for 5 days. Patients were able to eat, drink, watch television, or read. They could feel inebriated and/or unsteady when walking. If hallucinations occurred, the dose was to be reduced. The patients' normal medications were continued as it was feared that stopping them might result in severe depressive episodes. Before and following each treatment with ketamine, at patient clinic visits, the [[Beck Depression Inventory]] (BDI) and the [[Hamilton Rating Scale for Depression]] (HAMD-17) were obtained. Two of the patients were described with "impressive", as described by an attending doctor, improvement in depression being maintained for 12 months in patient A and recurrence at 2.5 months and 9 months in patient B.<ref name="pmid16533209">{{Cite journal|author=Correll GE, Futter GE |title=Two case studies of patients with major depressive disorder given low-dose (subanesthetic) ketamine infusions |journal=Pain Medicine |volume=7 |issue=1 |pages=92–5 |year=2006 |pmid=16533209 |doi=10.1111/j.1526-4637.2006.00101.x}}</ref>
  
 
The [[National Institute of Health]] News reports that a study of 18 patients has found that ketamine significantly improved treatment-resistant major depression within hours of injection.<ref>[http://www.nih.gov/news/pr/aug2006/nimh-07b.htm NIH. "Experimental Medication Kicks Depression in Hours Instead of Weeks"] ''NIH News'', 7 August 2006</ref> The improvement lasted up to one week after the single dose.<ref>Khamsi, R. [http://www.newscientist.com/channel/health/dn9696.html "Ketamine relieves depression within hours"] ''[[New Scientist]]'', 8 August 2006.</ref>  The patients in the study were previously treatment resistant, having tried an average of six other treatments that failed. NIMH director Dr. Thomas Insel said in the paper:
 
The [[National Institute of Health]] News reports that a study of 18 patients has found that ketamine significantly improved treatment-resistant major depression within hours of injection.<ref>[http://www.nih.gov/news/pr/aug2006/nimh-07b.htm NIH. "Experimental Medication Kicks Depression in Hours Instead of Weeks"] ''NIH News'', 7 August 2006</ref> The improvement lasted up to one week after the single dose.<ref>Khamsi, R. [http://www.newscientist.com/channel/health/dn9696.html "Ketamine relieves depression within hours"] ''[[New Scientist]]'', 8 August 2006.</ref>  The patients in the study were previously treatment resistant, having tried an average of six other treatments that failed. NIMH director Dr. Thomas Insel said in the paper:
Line 128: Line 129:
 
</blockquote>
 
</blockquote>
  
The researchers apparently attribute the effect to ketamine being an [[N-methyl-D-aspartate receptor|NMDA]] [[receptor antagonist]].<ref>{{cite journal |author=Zarate CA, Singh JB, Carlson PJ, ''et al.'' |title=A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression |journal=Archives of General Psychiatry |volume=63 |issue=8 |pages=856–64 |year=2006 |month=August |pmid=16894061 |doi=10.1001/archpsyc.63.8.856}}</ref> Those findings of Zarate ''et al.'' corroborate earlier findings by Berman ''et al.''.<ref>{{cite journal |author=Berman RM, Cappiello A, Anand A, ''et al.'' |title=Antidepressant effects of ketamine in depressed patients |journal=Biological Psychiatry |volume=47 |issue=4 |pages=351–4 |year=2000 |month=February |pmid=10686270 |doi=10.1016/S0006-3223(99)00230-9}}</ref> However Zarate et al. do raise some concerns about their results due to a possible lack of blinding, because of the inebriating effects of low dose ketamine infusion, and it is recommended that future studies include an [[active placebo]].
+
The researchers apparently attribute the effect to ketamine being an [[N-methyl-D-aspartate receptor|NMDA]] [[receptor antagonist]].<ref>{{Cite journal|author=Zarate CA, Singh JB, Carlson PJ, ''et al.'' |title=A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression |journal=Archives of General Psychiatry |volume=63 |issue=8 |pages=856–64 |year=2006 |month=August |pmid=16894061 |doi=10.1001/archpsyc.63.8.856}}</ref> Those findings of Zarate ''et al.'' corroborate earlier findings by Berman ''et al.''.<ref>{{Cite journal|author=Berman RM, Cappiello A, Anand A, ''et al.'' |title=Antidepressant effects of ketamine in depressed patients |journal=Biological Psychiatry |volume=47 |issue=4 |pages=351–4 |year=2000 |month=February |pmid=10686270 |doi=10.1016/S0006-3223(99)00230-9}}</ref> However Zarate et al. do raise some concerns about their results due to a possible lack of blinding, because of the inebriating effects of low dose ketamine infusion, and it is recommended that future studies include an [[active placebo]].
  
The findings by Zarate et al. are confirmed by Liebrenz et al., who substantially, according to an attending doctor, helped a 55-year-old male subject with a treatment-resistant major depression and a co-occurring alcohol and [[benzodiazepine dependence]] by giving an intravenous infusion of 0.5&nbsp;mg/kg ketamine over a period of 50 minutes and Goforth et al. who helped a patient with severe, recurrent major depressive disorder that demonstrated marked improvement within 8 hours of receiving a preoperative dose of ketamine and one treatment of [[electroconvulsive therapy]] with bitemporal electrode placement.<ref>{{cite journal |author=Liebrenz M, Borgeat A, Leisinger R, Stohler R |title=Intravenous ketamine therapy in a patient with a treatment-resistant major depression |journal=Swiss Medical Weekly |volume=137 |issue=15-16 |pages=234–6 |year=2007 |month=April |pmid=17525879 |doi=2007/15/smw-11852}}</ref><ref name="pmid17435569">{{cite journal |author=Goforth HW, Holsinger T |title=Rapid relief of severe major depressive disorder by use of preoperative ketamine and electroconvulsive therapy |journal=The Journal of ECT |volume=23 |issue=1 |pages=23–5 |year=2007 |month=March |pmid=17435569 |doi=10.1097/01.yct.0000263257.44539.23}}</ref>
+
The findings by Zarate et al. are confirmed by Liebrenz et al., who substantially, according to an attending doctor, helped a 55-year-old male subject with a treatment-resistant major depression and a co-occurring alcohol and [[benzodiazepine dependence]] by giving an intravenous infusion of 0.5&nbsp;mg/kg ketamine over a period of 50 minutes and Goforth et al. who helped a patient with severe, recurrent major depressive disorder that demonstrated marked improvement within 8 hours of receiving a preoperative dose of ketamine and one treatment of [[electroconvulsive therapy]] with bitemporal electrode placement.<ref>{{Cite journal|author=Liebrenz M, Borgeat A, Leisinger R, Stohler R |title=Intravenous ketamine therapy in a patient with a treatment-resistant major depression |journal=Swiss Medical Weekly |volume=137 |issue=15-16 |pages=234–6 |year=2007 |month=April |pmid=17525879 }}</ref><ref name="pmid17435569">{{Cite journal|author=Goforth HW, Holsinger T |title=Rapid relief of severe major depressive disorder by use of preoperative ketamine and electroconvulsive therapy |journal=The Journal of ECT |volume=23 |issue=1 |pages=23–5 |year=2007 |month=March |pmid=17435569 |doi=10.1097/01.yct.0000263257.44539.23}}</ref>
  
However, a new study in mice by Zarate et al. shows that blocking the NMDA receptor is an intermediate step. According to this study, blocking NMDA increases the activity of another receptor, [[AMPA receptor|AMPA]], and this boost in AMPA activity is crucial for ketamine’s rapid antidepressant actions. NMDA and AMPA are receptors for the [[neurotransmitter]] [[Glutamic acid#Neurotransmitter|glutamate]]. The glutamate system has been implicated in depression recently. This is a departure from previous thinking, which had focused on [[serotonin]] and [[norepinephrine]]. The glutamate system may represent a new avenue for treatment and research.<ref>"NIMH. "Faster-Acting Antidepressants Closer to Becoming a Reality". 25 July 2007, http://www.nimh.nih.gov/science-news/2007/faster-acting-antidepressants-closer-to-becoming-a-reality.shtml</ref>
+
However, a new study in mice by Zarate et al. shows that blocking the NMDA receptor is an intermediate step. According to this study, blocking NMDA increases the activity of another receptor, [[AMPA receptor|AMPA]], and this boost in AMPA activity is crucial for ketamine’s rapid antidepressant actions. NMDA and AMPA are receptors for the [[neurotransmitter]] [[Glutamic acid#Neurotransmitter|glutamate]]. The glutamate system has been implicated in depression recently. This is a departure from previous thinking, which had focused on [[serotonin]] and [[norepinephrine]]. The glutamate system may represent a new avenue for treatment and research.<ref>[http://www.nimh.nih.gov/science-news/2007/faster-acting-antidepressants-closer-to-becoming-a-reality.shtml Faster-Acting Antidepressants Closer to Becoming a Reality]", [[National Institute of Mental Health|NIMH]], 25 July 2007</ref>
  
Krystal et al. retrospectively compared the seizure duration, ictal EEG, and cognitive side effects of ketamine and methohexital anesthesia with ECT in 36 patients.<ref name="pmid12556568">{{cite journal |author=Krystal AD, Weiner RD, Dean MD, ''et al.'' |title=Comparison of seizure duration, ictal EEG, and cognitive effects of ketamine and methohexital anesthesia with ECT |journal=The Journal of Neuropsychiatry and Clinical Neurosciences |volume=15 |issue=1 |pages=27–34 |year=2003 |pmid=12556568}}</ref> Ketamine was well tolerated and prolonged seizure duration overall, but particularly in those who had a seizure duration shorter than 25 seconds with methohexital at the maximum available stimulus intensity. Ketamine also increased midictal EEG slow-wave amplitude. Thus, a switch to ketamine may be useful when it is difficult to elicit a robust seizure. Faster post-treatment reorientation with ketamine may suggest a lower level of associated cognitive side effects.
+
Krystal et al. retrospectively compared the seizure duration, ictal EEG, and cognitive side effects of ketamine and methohexital anesthesia with ECT in 36 patients.<ref name="pmid12556568">{{Cite journal|author=Krystal AD, Weiner RD, Dean MD, ''et al.'' |title=Comparison of seizure duration, ictal EEG, and cognitive effects of ketamine and methohexital anesthesia with ECT |journal=The Journal of Neuropsychiatry and Clinical Neurosciences |volume=15 |issue=1 |pages=27–34 |year=2003 |pmid=12556568}}</ref> Ketamine was well tolerated and prolonged seizure duration overall, but particularly in those who had a seizure duration shorter than 25 seconds with methohexital at the maximum available stimulus intensity. Ketamine also increased midictal EEG slow-wave amplitude. Thus, a switch to ketamine may be useful when it is difficult to elicit a robust seizure. Faster post-treatment reorientation with ketamine may suggest a lower level of associated cognitive side effects.
  
Kudoh et al. investigated whether ketamine is suitable for depressed patients who had undergone orthopedic surgery.<ref name="pmid12088953">{{cite journal |author=Kudoh A, Takahira Y, Katagai H, Takazawa T |title=Small-dose ketamine improves the postoperative state of depressed patients |journal=Anesthesia and Analgesia |volume=95 |issue=1 |pages=114–8, table of contents |year=2002 |month=July |pmid=12088953 |doi=10.1097/00000539-200207000-00020 |url=http://www.anesthesia-analgesia.org/cgi/pmidlookup?view=long&pmid=12088953}}</ref> They studied 70 patients with major depression and 25 patients as the control (Group C). The depressed patients were divided randomly into two groups; patients in Group A, initial HAMD 12,7 (n = 35) were induced with propofol, fentanyl, and ketamine and patients in Group B, initial HAMD 12,3 (n = 35) were induced with propofol and fentanyl. Depressed mood, suicidal tendencies, somatic anxiety, and hypochondriasis significantly decreased in Group A as compared with Group B. The group receiving ketamine also had significantly lower postoperative pain.
+
Kudoh et al. investigated whether ketamine is suitable for depressed patients who had undergone orthopedic surgery.<ref name="pmid12088953">{{Cite journal|author=Kudoh A, Takahira Y, Katagai H, Takazawa T |title=Small-dose ketamine improves the postoperative state of depressed patients |journal=Anesthesia and Analgesia |volume=95 |issue=1 |pages=114–8, table of contents |year=2002 |month=July |pmid=12088953 |doi=10.1097/00000539-200207000-00020 |url=http://www.anesthesia-analgesia.org/cgi/pmidlookup?view=long&pmid=12088953}}</ref> They studied 70 patients with major depression and 25 patients as the control (Group C). The depressed patients were divided randomly into two groups; patients in Group A, initial HAMD 12,7 (n = 35) were induced with propofol, fentanyl, and ketamine and patients in Group B, initial HAMD 12,3 (n = 35) were induced with propofol and fentanyl. Depressed mood, suicidal tendencies, somatic anxiety, and hypochondriasis significantly decreased in Group A as compared with Group B. The group receiving ketamine also had significantly lower postoperative pain.
  
Acute administration of ketamine at the higher dose, but not [[imipramine]], increased BDNF protein levels in the rat [[hippocampus]]. The increase of hippocampal BDNF protein levels induced by ketamine might be necessary to produce a rapid onset of antidepressant action.<ref name="pmid17884272">{{cite journal |author=Garcia LS, Comim CM, Valvassori SS, ''et al.'' |title=Acute administration of ketamine induces antidepressant-like effects in the forced swimming test and increases BDNF levels in the rat hippocampus |journal=Progress in Neuro-psychopharmacology & Biological Psychiatry |volume=32 |issue=1 |pages=140–4 |year=2008 |month=January |pmid=17884272 |doi=10.1016/j.pnpbp.2007.07.027}}</ref>
+
Acute administration of ketamine at the higher dose, but not [[imipramine]], increased BDNF protein levels in the rat [[hippocampus]]. The increase of hippocampal BDNF protein levels induced by ketamine might be necessary to produce a rapid onset of antidepressant action.<ref name="pmid17884272">{{Cite journal|author=Garcia LS, Comim CM, Valvassori SS, ''et al.'' |title=Acute administration of ketamine induces antidepressant-like effects in the forced swimming test and increases BDNF levels in the rat hippocampus |journal=Progress in Neuro-psychopharmacology & Biological Psychiatry |volume=32 |issue=1 |pages=140–4 |year=2008 |month=January |pmid=17884272 |doi=10.1016/j.pnpbp.2007.07.027}}</ref>
  
 
====Treatment of addiction====
 
====Treatment of addiction====
The Russian doctor Evgeny Krupitsky (Clinical Director of Research for the Saint Petersburg Regional Center for Research in Addiction and Psychopharmacology) has claimed to have encouraging results by using ketamine as part of a treatment for alcohol addiction which combines psychedelic and aversive techniques.<ref>[http://www.eleusis.us/resource-center/references/acamethod.php The Combination of Psychedelic and Aversive Approaches in Alcoholism Treatment - Eleusis<!-- Bot generated title -->]</ref><ref>{{cite journal |author=Krupitsky EM, Grinenko AY |title=Ketamine psychedelic therapy (KPT): a review of the results of ten years of research |journal=Journal of Psychoactive Drugs |volume=29 |issue=2 |pages=165–83 |year=1997 |pmid=9250944}}</ref> This method involved psychotherapy, controlled ketamine use and group therapy, and resulted in 60 of the 86 alcoholic males selected for the study remaining fully abstinent through one year of treatment. He has also treated heroin addicts and reached the conclusion that one ketamine-assisted psychotherapy session was significantly more effective than [[active placebo]] in promoting abstinence from heroin during one year without any adverse reactions. In a recently published study 59 detoxified inpatients with heroin dependence received a ketamine-assisted psychotherapy (KPT) session prior to their discharge from an addiction treatment hospital, and were then randomized into two treatment groups.
+
The Russian doctor Evgeny Krupitsky (Clinical Director of Research for the Saint Petersburg Regional Center for Research in Addiction and Psychopharmacology) has claimed to have encouraging results by using ketamine as part of a treatment for alcohol addiction which combines psychedelic and aversive techniques.<ref>[http://www.eleusis.us/resource-center/references/acamethod.php The Combination of Psychedelic and Aversive Approaches in Alcoholism Treatment - Eleusis<!-- Bot generated title -->]</ref><ref>{{Cite journal|author=Krupitsky EM, Grinenko AY |title=Ketamine psychedelic therapy (KPT): a review of the results of ten years of research |journal=Journal of Psychoactive Drugs |volume=29 |issue=2 |pages=165–83 |year=1997 |pmid=9250944}}</ref> This method involved psychotherapy, controlled ketamine use and group therapy, and resulted in 60 of the 86 alcoholic males selected for the study remaining fully abstinent through one year of treatment. He has also treated heroin addicts and reached the conclusion that one ketamine-assisted psychotherapy session was significantly more effective than [[active placebo]] in promoting abstinence from heroin during one year without any adverse reactions. In a recently published study 59 detoxified inpatients with heroin dependence received a ketamine-assisted psychotherapy (KPT) session prior to their discharge from an addiction treatment hospital, and were then randomized into two treatment groups.
  
Participants in the first group received two addiction counseling sessions followed by two KPT sessions, (with a single im injection of 2&nbsp;mg/kg ketamine) with sessions scheduled on a monthly interval (multiple KPT group). Participants in the second group received two addiction counseling sessions on a monthly interval, but no additional ketamine therapy sessions (single KPT group). At one-year follow-up, survival analysis demonstrated a significantly higher rate of abstinence in the multiple KPT group. Thirteen out of 26 subjects (50%) in the multiple KPT group remained abstinent, compared to 6 out of 27 subjects (22.2%) in the single KPT group (p < 0.05). No differences between groups were found in depression, anxiety, craving for heroin, or their understanding of the meaning of their lives. It was concluded that three sessions of ketamine-assisted psychotherapy are more effective than a single session for the treatment of heroin addiction.<ref>http://www.eleusis.us/resource-center/references/ketamine-psychotherapy-heroin.pdf</ref><ref>{{cite journal |author=Krupitsky EM, Burakov AM, Dunaevsky IV, Romanova TN, Slavina TY, Grinenko AY |title=Single versus repeated sessions of ketamine-assisted psychotherapy for people with heroin dependence |journal=Journal of Psychoactive Drugs |volume=39 |issue=1 |pages=13–9 |year=2007 |month=March |pmid=17523581}}</ref>
+
Participants in the first group received two addiction counseling sessions followed by two KPT sessions, (with a single im injection of 2&nbsp;mg/kg ketamine) with sessions scheduled on a monthly interval (multiple KPT group). Participants in the second group received two addiction counseling sessions on a monthly interval, but no additional ketamine therapy sessions (single KPT group). At one-year follow-up, survival analysis demonstrated a significantly higher rate of abstinence in the multiple KPT group. Thirteen out of 26 subjects (50%) in the multiple KPT group remained abstinent, compared to 6 out of 27 subjects (22.2%) in the single KPT group (p < 0.05). No differences between groups were found in depression, anxiety, craving for heroin, or their understanding of the meaning of their lives. It was concluded that three sessions of ketamine-assisted psychotherapy are more effective than a single session for the treatment of heroin addiction.<ref>http://www.eleusis.us/resource-center/references/ketamine-psychotherapy-heroin.pdf</ref><ref>{{Cite journal|author=Krupitsky EM, Burakov AM, Dunaevsky IV, Romanova TN, Slavina TY, Grinenko AY |title=Single versus repeated sessions of ketamine-assisted psychotherapy for people with heroin dependence |journal=Journal of Psychoactive Drugs |volume=39 |issue=1 |pages=13–9 |year=2007 |month=March |pmid=17523581}}</ref>
  
Krupitsky and Kolp summarized their work to date in 2007.<ref>{{cite book |last1=Krupitsky |last2=Kolp |chapter=Ketamine Psychedelic Psychotherapy |title=Psychedelic Medicine: New Evidence for Hallucinogens as Treatments |editor=Michael J. Winkelman and Thomas B., Roberts |location=Westport, CT |publisher=Praeger/Greenwood}}{{pn}}</ref>
+
Krupitsky and Kolp summarized their work to date in 2007.<ref>{{Cite book|last1=Krupitsky |last2=Kolp |chapter=Ketamine Psychedelic Psychotherapy |title=Psychedelic Medicine: New Evidence for Hallucinogens as Treatments |editor=Michael J. Winkelman and Thomas B., Roberts |location=Westport, CT |publisher=Praeger/Greenwood}}{{Page needed|date=September 2010}}</ref>
  
 
Jovaisa et al. from Lithuania demonstrated attenuation of opiate withdrawal symptoms with ketamine. A total of 58 opiate-dependent patients were enrolled in a randomized, placebo-controlled, double-blind study. Patients underwent rapid opiate antagonist induction under general anesthesia. Prior to opiate antagonist induction patients were given either placebo (normal saline) or subanesthetic ketamine infusion of 0.5&nbsp;mg/kg·h. Ketamine group presented better control of withdrawal symptoms, which lasted beyond ketamine infusion itself. Significant differences between ketamine and Control groups were noted in anesthetic and early postanesthetic phases. There were no differences in effects on outcome after 4 months.<ref name="jovaisa"/>
 
Jovaisa et al. from Lithuania demonstrated attenuation of opiate withdrawal symptoms with ketamine. A total of 58 opiate-dependent patients were enrolled in a randomized, placebo-controlled, double-blind study. Patients underwent rapid opiate antagonist induction under general anesthesia. Prior to opiate antagonist induction patients were given either placebo (normal saline) or subanesthetic ketamine infusion of 0.5&nbsp;mg/kg·h. Ketamine group presented better control of withdrawal symptoms, which lasted beyond ketamine infusion itself. Significant differences between ketamine and Control groups were noted in anesthetic and early postanesthetic phases. There were no differences in effects on outcome after 4 months.<ref name="jovaisa"/>
Line 151: Line 152:
 
====Treatment of complex regional pain syndrome (reflex sympathetic dystrophy)====
 
====Treatment of complex regional pain syndrome (reflex sympathetic dystrophy)====
 
Ketamine is being used as an experimental and controversial treatment for [[Complex Regional Pain Syndrome]] (CRPS) also known as [[Complex regional pain syndrome|Reflex Sympathetic Dystrophy]] ([[Complex regional pain syndrome|RSD]]). CRPS/RSD is a severe chronic pain condition characterized by sensory, [[autonomic]], motor and dystrophic signs and symptoms. The pain in CRPS is continuous, it worsens over time, and it is usually disproportionate to the severity and duration of the inciting event. The hypothesis is that ketamine manipulates [[NMDA]] receptors which might reboot aberrant brain activity. There are two treatment modalities, the first consist of a low dose ketamine infusion of between 25–90&nbsp;mg per day, over five days either in hospital or as an outpatient. This is called the awake technique.
 
Ketamine is being used as an experimental and controversial treatment for [[Complex Regional Pain Syndrome]] (CRPS) also known as [[Complex regional pain syndrome|Reflex Sympathetic Dystrophy]] ([[Complex regional pain syndrome|RSD]]). CRPS/RSD is a severe chronic pain condition characterized by sensory, [[autonomic]], motor and dystrophic signs and symptoms. The pain in CRPS is continuous, it worsens over time, and it is usually disproportionate to the severity and duration of the inciting event. The hypothesis is that ketamine manipulates [[NMDA]] receptors which might reboot aberrant brain activity. There are two treatment modalities, the first consist of a low dose ketamine infusion of between 25–90&nbsp;mg per day, over five days either in hospital or as an outpatient. This is called the awake technique.
Open label, prospective, pain journal evaluation of a 10-day infusion of intravenous ketamine (awake technique) in the CRPS patient concluded that "A four-hour ketamine infusion escalated from 40–80 mg over a 10-day period can result in a significant reduction of pain with increased mobility and a tendency to decreased autonomic dysregulation".<ref name=pubmed16850072>{{cite journal |author=Goldberg ME, Domsky R, Scaringe D, ''et al.'' |title=Multi-day low dose ketamine infusion for the treatment of complex regional pain syndrome |journal=Pain Physician |volume=8 |issue=2 |pages=175–9 |year=2005 |month=April |pmid=16850072 |url=http://www.painphysicianjournal.com/linkout_vw.php?issn=1533-3159&vol=8&page=175}}</ref>
+
Open label, prospective, pain journal evaluation of a 10-day infusion of intravenous ketamine (awake technique) in the CRPS patient concluded that "A four-hour ketamine infusion escalated from 40–80 mg over a 10-day period can result in a significant reduction of pain with increased mobility and a tendency to decreased autonomic dysregulation".<ref name=pubmed16850072>{{Cite journal|author=Goldberg ME, Domsky R, Scaringe D, ''et al.'' |title=Multi-day low dose ketamine infusion for the treatment of complex regional pain syndrome |journal=Pain Physician |volume=8 |issue=2 |pages=175–9 |year=2005 |month=April |pmid=16850072 |url=http://www.painphysicianjournal.com/linkout_vw.php?issn=1533-3159&vol=8&page=175}}</ref>
  
 
Case notes of 33 patients whose CRPS pain was treated by the inpatient administration of a continuous subanesthetic intravenous infusion of ketamine were reviewed at Mackay Base Hospital, Queensland, Australia. A total of 33 patients with diagnoses of CRPS who had undergone ketamine treatment at least once were identified. Due to relapse, 12 of 33 patients received a second course of therapy, and two of 33 patients received a third. There was complete pain relief in 25 (76%), partial relief in six (18%), and no relief in two (6%) patients.
 
Case notes of 33 patients whose CRPS pain was treated by the inpatient administration of a continuous subanesthetic intravenous infusion of ketamine were reviewed at Mackay Base Hospital, Queensland, Australia. A total of 33 patients with diagnoses of CRPS who had undergone ketamine treatment at least once were identified. Due to relapse, 12 of 33 patients received a second course of therapy, and two of 33 patients received a third. There was complete pain relief in 25 (76%), partial relief in six (18%), and no relief in two (6%) patients.
Line 159: Line 160:
 
The second treatment modality consists of putting the patient into a medically-induced coma and given an extremely high dosage of ketamine; typically between 600–900&nbsp;mg.<ref name=cnn_rsd_coma>[http://www.rsdfoundation.org/en/CNN_Transcript_RSD.htm CNN report on ketamine therapy for CRPS/RSD] 1 September 2006</ref> This version, currently not allowed in the United States, is most commonly done in Germany but some treatments are now also taking place in Monterrey, Mexico. According to Dr Schwartzman, 14 cases out of 41 patients in the coma induced ketamine experiments were completely cured. "We haven't cured the original injury", he says, "but we have cured the RSD or kept it in remission. The RSD pain is gone." He added that "No one ever cured it before... In 40 years, I have never seen anything like it. These are people who were disabled and in horrible pain. Most were completely incapacitated. They go back to work, back to school, and are doing everything they used to do. Most are on no medications at all. I have taken morphine pumps out of people. You turn off the pain and reset the whole system."<ref name=cnn_rsd_coma/>
 
The second treatment modality consists of putting the patient into a medically-induced coma and given an extremely high dosage of ketamine; typically between 600–900&nbsp;mg.<ref name=cnn_rsd_coma>[http://www.rsdfoundation.org/en/CNN_Transcript_RSD.htm CNN report on ketamine therapy for CRPS/RSD] 1 September 2006</ref> This version, currently not allowed in the United States, is most commonly done in Germany but some treatments are now also taking place in Monterrey, Mexico. According to Dr Schwartzman, 14 cases out of 41 patients in the coma induced ketamine experiments were completely cured. "We haven't cured the original injury", he says, "but we have cured the RSD or kept it in remission. The RSD pain is gone." He added that "No one ever cured it before... In 40 years, I have never seen anything like it. These are people who were disabled and in horrible pain. Most were completely incapacitated. They go back to work, back to school, and are doing everything they used to do. Most are on no medications at all. I have taken morphine pumps out of people. You turn off the pain and reset the whole system."<ref name=cnn_rsd_coma/>
  
In Tuebingen, Germany Dr Kiefer treated a patient presented with a rapidly progressing contiguous spread of CRPS from a severe ligamentous wrist injury. Standard pharmacological and interventional therapy successively failed to halt the spread of CRPS from the wrist to the entire right arm. Her pain was unmanageable with all standard therapy. As a last treatment option, the patient was transferred to the intensive care unit and treated on a compassionate care basis with anesthetic doses of ketamine in gradually increasing (3–5&nbsp;mg/kg·h) doses in conjunction with [[midazolam]] over a period of 5 days. On the second day, [[edema]], and discoloration began to resolve and increased spontaneous movement was noted. On day 6, symptoms completely resolved and infusions were tapered.  The patient emerged from anesthesia completely free of pain and associated CRPS signs and symptoms. The patient has maintained this complete remission from CRPS for 8 years now. The psychiatric side effects of ketamine were successfully managed with the concomitant use of midazolam and resolved within 1 month of treatment.<ref>{{cite journal |author=Kiefer RT, Rohr P, Ploppa A, Altemeyer KH, Schwartzman RJ |title=Complete recovery from intractable complex regional pain syndrome, CRPS-type I, following anesthetic ketamine and midazolam |journal=Pain Practice |volume=7 |issue=2 |pages=147–50 |year=2007 |month=June |pmid=17559485 |doi=10.1111/j.1533-2500.2007.00123.x}}</ref>
+
In Tuebingen, Germany Dr Kiefer treated a patient presented with a rapidly progressing contiguous spread of CRPS from a severe ligamentous wrist injury. Standard pharmacological and interventional therapy successively failed to halt the spread of CRPS from the wrist to the entire right arm. Her pain was unmanageable with all standard therapy. As a last treatment option, the patient was transferred to the intensive care unit and treated on a compassionate care basis with anesthetic doses of ketamine in gradually increasing (3–5&nbsp;mg/kg·h) doses in conjunction with [[midazolam]] over a period of 5 days. On the second day, [[edema]], and discoloration began to resolve and increased spontaneous movement was noted. On day 6, symptoms completely resolved and infusions were tapered.  The patient emerged from anesthesia completely free of pain and associated CRPS signs and symptoms. The patient has maintained this complete remission from CRPS for 8 years now. The psychiatric side effects of ketamine were successfully managed with the concomitant use of midazolam and resolved within 1 month of treatment.<ref>{{Cite journal|author=Kiefer RT, Rohr P, Ploppa A, Altemeyer KH, Schwartzman RJ |title=Complete recovery from intractable complex regional pain syndrome, CRPS-type I, following anesthetic ketamine and midazolam |journal=Pain Practice |volume=7 |issue=2 |pages=147–50 |year=2007 |month=June |pmid=17559485 |doi=10.1111/j.1533-2500.2007.00123.x}}</ref>
  
 
====Postoperative pain====
 
====Postoperative pain====
The dissociative anesthetic effects of ketamine have also been applied within the realm of postoperative pain management. Low doses of ketamine have been found to significantly reduce morphine consumption as well as reports of nausea following abdominal surgery.<ref name="postopms">{{cite journal |author=Lundberg GD |title=Postoperative Ketamine Can Reduce Morphine Consumption and Nausea |journal=The Medscape Journal of Medicine |volume=10 |issue=10 |page=234 |year=2008 |pmc=2605142}}</ref>
+
The dissociative anesthetic effects of ketamine have also been applied within the realm of postoperative pain management. Low doses of ketamine have been found to significantly reduce morphine consumption as well as reports of nausea following abdominal surgery.<ref name="postopms">{{Cite journal|author=Lundberg GD |title=Postoperative Ketamine Can Reduce Morphine Consumption and Nausea |journal=The Medscape Journal of Medicine |volume=10 |issue=10 |page=234 |year=2008 |pmc=2605142}}</ref>
  
 
====Contents of various brands of ketamine====
 
====Contents of various brands of ketamine====
Ketanest S  Parke Davis (Pfizer) - S enantiomer only, no preservatives<ref name="PMID15055891">{{cite journal |author=De Reuck J, Decoo D, Strijckmans K, Lemahieu I |title=Does the severity of leukoaraiosis contribute to senile dementia? A comparative computerized and positron emission tomographic study |journal=European Neurology |volume=32 |issue=4 |pages=199–205 |year=1992 |pmid=1505589 |doi=10.1017/S0265021504003059}}</ref>
+
Ketanest S  Parke Davis (Pfizer) - S enantiomer only, no preservatives<ref name="PMID15055891">{{Cite journal|author=De Reuck J, Decoo D, Strijckmans K, Lemahieu I |title=Does the severity of leukoaraiosis contribute to senile dementia? A comparative computerized and positron emission tomographic study |journal=European Neurology |volume=32 |issue=4 |pages=199–205 |year=1992 |pmid=1505589 |doi=10.1017/S0265021504003059}}</ref>
  
 
Ketaset (Wyeth) - R/S ketamine and benzethonium chloride as a preservative.<ref>[http://ftdodge.naccvp.com/view.php?u=country&p=msds&prodnum=1003101.HTM Fort Dodge (division of Wyeth) Ketamine Lable]. Article retrieved on 1 November 2008</ref>
 
Ketaset (Wyeth) - R/S ketamine and benzethonium chloride as a preservative.<ref>[http://ftdodge.naccvp.com/view.php?u=country&p=msds&prodnum=1003101.HTM Fort Dodge (division of Wyeth) Ketamine Lable]. Article retrieved on 1 November 2008</ref>
Line 174: Line 175:
  
 
==Neuropharmacology==
 
==Neuropharmacology==
Ketamine was long thought to act primarily by inhibiting NMDA receptors.<ref name="pmid14740534">{{cite journal |author=Radovanović D, Pjević M |title=[Ketamine: the past 30 years and its future] |journal=Medicinski Pregled |volume=56 |issue=9-10 |pages=439–45 |year=2003 |pmid=14740534 |doi=10.2298/MPNS0310439R}}</ref><ref name="pmid18175098">{{cite journal |author=Sinner B, Graf BM |title=Ketamine |journal=Handbook of Experimental Pharmacology |volume= 182|issue=182 |pages=313–33 |year=2008 |pmid=18175098 |doi=10.1007/978-3-540-74806-9_15}}</ref> But another NMDA receptor antagonist, MK-801, does not exert the same hypnotic effects.<ref>{{cite journal |author=Kelland MD, Soltis RP, Boldry RC, Walters JR |title=Behavioral and electrophysiological comparison of ketamine with dizocilpine in the rat |journal=Physiology & Behavior |volume=54 |issue=3 |pages=547–54 |year=1993 |month=September |pmid=8415949 |doi=10.1016/0031-9384(93)90248-E}}</ref> It appears more likely that the hypnotic effects of ketamine are produced by inhibiting hyperpolarization-activated cyclic nucleotide-modulated ([[HCN1]]) cation channels, which mediate the "sag" current (''I<sub>h </sub>'') in neurons <ref name="pmid19158287">{{cite journal |author=Chen X, Shu S, Bayliss DA |title=HCN1 channel subunits are a molecular substrate for hypnotic actions of ketamine |journal=J. Neurosci. |volume=29 |issue=3 |pages=600–9 |year=2009 |month=January |pmid=19158287 |pmc=2744993 |doi=10.1523/JNEUROSCI.3481-08.2009 |url=}}</ref>. Inhibition of ''I<sub>h</sub>'' by ketamine in cultured neurons causes a hyperpolarizing shift in resting membrane potential and enhances summation of excitatory currents.<ref name="PMID 15958747">{{cite journal |author=Chen X, Sirois JE, Lei Q, Talley EM, Lynch C, Bayliss DA |title=HCN subunit-specific and cAMP-modulated effects of anesthetics on neuronal pacemaker currents |journal=The Journal of Neuroscience |volume=25 |issue=24 |pages=5803–14 |year=2005 |month=June |pmid=15958747 |doi=10.1523/JNEUROSCI.1153-05.2005}}</ref> Such effects, if induced ''in vivo'', would likely induce cortical oscillations reminiscent of sleep.<ref>{{cite journal |author=Carr DB, Andrews GD, Glen WB, Lavin A |title=alpha2-Noradrenergic receptors activation enhances excitability and synaptic integration in rat prefrontal cortex pyramidal neurons via inhibition of HCN currents |journal=The Journal of Physiology |volume=584 |issue=2 |pages=437–50 |year=2007 |month=October |pmid=17702809 |pmc=2277172 |doi=10.1113/jphysiol.2007.141671}}</ref> Most importantly, knockout of HCN1 channels in mice eliminates the hypnotic actions of ketamine.<ref name="PMID 15958747"/>  
+
Ketamine was long thought to act primarily by inhibiting NMDA receptors.<ref name="pmid14740534">{{Cite journal|author=Radovanović D, Pjević M |title=[Ketamine: the past 30 years and its future] |journal=Medicinski Pregled |volume=56 |issue=9-10 |pages=439–45 |year=2003 |pmid=14740534 |doi=10.2298/MPNS0310439R}}</ref><ref name="pmid18175098">{{Cite journal|author=Sinner B, Graf BM |title=Ketamine |journal=Handbook of Experimental Pharmacology |volume= 182|issue=182 |pages=313–33 |year=2008 |pmid=18175098 |doi=10.1007/978-3-540-74806-9_15}}</ref> But another NMDA receptor antagonist, MK-801, does not exert the same hypnotic effects.<ref>{{Cite journal|author=Kelland MD, Soltis RP, Boldry RC, Walters JR |title=Behavioral and electrophysiological comparison of ketamine with dizocilpine in the rat |journal=Physiology & Behavior |volume=54 |issue=3 |pages=547–54 |year=1993 |month=September |pmid=8415949 |doi=10.1016/0031-9384(93)90248-E}}</ref> It appears more likely that the hypnotic effects of ketamine are produced by inhibiting hyperpolarization-activated cyclic nucleotide-modulated ([[HCN1]]) cation channels, which mediate the "sag" current (''I<sub>h </sub>'') in neurons.<ref name="pmid19158287">{{Cite journal|author=Chen X, Shu S, Bayliss DA |title=HCN1 channel subunits are a molecular substrate for hypnotic actions of ketamine |journal=J. Neurosci. |volume=29 |issue=3 |pages=600–9 |year=2009 |month=January |pmid=19158287 |pmc=2744993 |doi=10.1523/JNEUROSCI.3481-08.2009 |url=}}</ref> Inhibition of ''I<sub>h</sub>'' by ketamine in cultured neurons causes a hyperpolarizing shift in resting membrane potential and enhances summation of excitatory currents.<ref name="PMID 15958747">{{Cite journal|author=Chen X, Sirois JE, Lei Q, Talley EM, Lynch C, Bayliss DA |title=HCN subunit-specific and cAMP-modulated effects of anesthetics on neuronal pacemaker currents |journal=The Journal of Neuroscience |volume=25 |issue=24 |pages=5803–14 |year=2005 |month=June |pmid=15958747 |doi=10.1523/JNEUROSCI.1153-05.2005}}</ref> Such effects, if induced ''in vivo'', would likely induce cortical oscillations reminiscent of sleep.<ref>{{Cite journal|author=Carr DB, Andrews GD, Glen WB, Lavin A |title=alpha2-Noradrenergic receptors activation enhances excitability and synaptic integration in rat prefrontal cortex pyramidal neurons via inhibition of HCN currents |journal=The Journal of Physiology |volume=584 |issue=2 |pages=437–50 |year=2007 |month=October |pmid=17702809 |pmc=2277172 |doi=10.1113/jphysiol.2007.141671}}</ref> Most importantly, knockout of HCN1 channels in mice eliminates the hypnotic actions of ketamine.<ref name="PMID 15958747"/>
  
Ketamine is an [[uncompetitive inhibition|uncompetitive]] [[NMDA receptor antagonist]].<ref name="pmid 16424917">{{cite journal |author=Lipton SA |title=Paradigm shift in neuroprotection by NMDA receptor blockade: memantine and beyond |journal=Nature Reviews. Drug Discovery |volume=5 |issue=2 |pages=160–70 |year=2006 |month=February |pmid=16424917 |doi=10.1038/nrd1958}}</ref> This [[receptor (biochemistry)|receptor]] opens in response to binding of the [[neurotransmitter]] [[glutamate]], and blockade of this receptors is believed to mediate the [[analgesia|analgesic]] (reduction of pain) effects of ketamine at low doses <ref name="pmid18175098">{{cite journal |author=Sinner B, Graf BM |title=Ketamine |journal=Handb Exp Pharmacol |volume= |issue=182 |pages=313–33 |year=2008 |pmid=18175098 |doi=10.1007/978-3-540-74806-9_15 |url=}}</ref>. Evidence for this is reinforced by the fact that [[naloxone]], an [[opioid]] antagonist, does not reverse the analgesia. Studies also seem to indicate that ketamine is "use dependent" meaning it only initiates its blocking action once a glutamate binds to the NMDA receptor.{{Citation needed|date=December 2008}}
+
Ketamine is an [[uncompetitive inhibition|uncompetitive]] [[NMDA receptor antagonist]].<ref name="pmid 16424917">{{Cite journal|author=Lipton SA |title=Paradigm shift in neuroprotection by NMDA receptor blockade: memantine and beyond |journal=Nature Reviews. Drug Discovery |volume=5 |issue=2 |pages=160–70 |year=2006 |month=February |pmid=16424917 |doi=10.1038/nrd1958}}</ref> This [[receptor (biochemistry)|receptor]] opens in response to binding of the [[neurotransmitter]] [[glutamate]], and blockade of this receptors is believed to mediate the [[analgesia|analgesic]] (reduction of pain) effects of ketamine at low doses.<ref name="pmid18175098">{{Cite journal|author=Sinner B, Graf BM |title=Ketamine |journal=Handb Exp Pharmacol |volume= 182|issue=182 |pages=313–33 |year=2008 |pmid=18175098 |doi=10.1007/978-3-540-74806-9_15 |url=}}</ref> Evidence for this is reinforced by the fact that [[naloxone]], an [[opioid]] antagonist, does not reverse the analgesia. Studies also seem to indicate that ketamine is "use dependent" meaning it only initiates its blocking action once a glutamate binds to the NMDA receptor.{{Citation needed|date=December 2008}}
  
At high, fully anesthetic level doses, ketamine has also been found to bind to [[opioid]] mu receptors and sigma receptors.{{Citation needed|date=December 2008}}  Thus, loss of consciousness that occurs at high doses may be partially due to binding at the opioid mu and sigma receptors.{{Citation needed|date=December 2008}} It has also been shown to act as a [[potency (pharmacology)|potent]] [[D2 receptor|D<sub>2</sub> receptor]] [[partial agonist]],<ref name="pmid19391150">{{cite journal | author = Seeman P, Guan HC, Hirbec H | title = Dopamine D2High receptors stimulated by phencyclidines, lysergic acid diethylamide, salvinorin A, and modafinil | journal = Synapse (New York, N.Y.) | volume = 63 | issue = 8 | pages = 698–704 | year = 2009 | month = August | pmid = 19391150 | doi = 10.1002/syn.20647 | url = http://dx.doi.org/10.1002/syn.20647}}</ref> as well as a [[dopamine reuptake inhibitor]].{{Citation needed|date=April 2010}}
+
At high, fully anesthetic level doses, ketamine has also been found to bind to [[opioid]] mu receptors and sigma receptors.{{Citation needed|date=December 2008}}  Thus, loss of consciousness that occurs at high doses may be partially due to binding at the opioid mu and sigma receptors.{{Citation needed|date=December 2008}} It has also been shown to act as a [[potency (pharmacology)|potent]] [[D2 receptor|D<sub>2</sub> receptor]] [[partial agonist]],<ref name="pmid19391150">{{Cite journal | unused_data = DUPLICATE DATA: doi = 10.1002/syn.20647 | author = Seeman P, Guan HC, Hirbec H | title = Dopamine D2High receptors stimulated by phencyclidines, lysergic acid diethylamide, salvinorin A, and modafinil | journal = Synapse (New York, N.Y.) | volume = 63 | issue = 8 | pages = 698–704 | year = 2009 | month = August | pmid = 19391150 | doi = 10.1002/syn.20647}}</ref> as well as a [[dopamine reuptake inhibitor]].{{Citation needed|date=April 2010}}
  
(''rac'')-ketamine is a noncompetitive inhibitor of the α7 nAChR at clinically relevant concentrations. The preservative benzethonium chloride competitively inhibits α7 and α4β2 nAChRs at concentrations present in the clinical formulation of Ketalar.<ref name="pmid11606328">{{cite journal |author=Coates KM, Flood P |title=Ketamine and its preservative, benzethonium chloride, both inhibit human recombinant alpha7 and alpha4beta2 neuronal nicotinic acetylcholine receptors in Xenopus oocytes |journal=British Journal of Pharmacology |volume=134 |issue=4 |pages=871–9 |year=2001 |month=October |pmid=11606328 |pmc=1573008 |doi=10.1038/sj.bjp.0704315}}</ref>
+
(''rac'')-ketamine is a noncompetitive inhibitor of the α7 nAChR at clinically relevant concentrations. The preservative benzethonium chloride competitively inhibits α7 and α4β2 nAChRs at concentrations present in the clinical formulation of Ketalar.<ref name="pmid11606328">{{Cite journal|author=Coates KM, Flood P |title=Ketamine and its preservative, benzethonium chloride, both inhibit human recombinant alpha7 and alpha4beta2 neuronal nicotinic acetylcholine receptors in Xenopus oocytes |journal=British Journal of Pharmacology |volume=134 |issue=4 |pages=871–9 |year=2001 |month=October |pmid=11606328 |pmc=1573008 |doi=10.1038/sj.bjp.0704315}}</ref>
  
Ketamine is [[racemic]], and its R and S [[stereoisomers]] have different binding affinities: (S)-ketamine has about four times greater affinity for the PCP site of the NMDA receptor than does (R)-ketamine (in guinea pig brain).{{Citation needed|date=December 2008}} [[Esketamine|(S)-ketamine]] seems to induce drowsiness more strongly than the (R) enantiomer; it is probable that (R)-ketamine is the stronger sigma agonist and so this enantiomer is likely to be responsible for the lowering of the seizure threshold that can occur with ketamine.{{Citation needed|date=December 2008}} Since (S)-ketamine has greater analgesic effects and less hallucinogenic side effects than (R)-ketamine, the pure (S) enantiomer is sometimes preferred to the racemic mix for use in medical procedures, especially when lower doses are used for minor surgical procedures where the patient remains conscious during the operation.<ref>{{cite journal |author=Togal T, Demirbilek S, Koroglu A, Yapici E, Ersoy O |title=Effects of S(+) ketamine added to bupivacaine for spinal anaesthesia for prostate surgery in elderly patients |journal=European Journal of Anaesthesiology |volume=21 |issue=3 |pages=193–7 |year=2004 |month=March |pmid=15055891}}</ref>
+
Ketamine is [[racemic]], and its R and S [[stereoisomers]] have different binding affinities: (S)-ketamine has about four times greater affinity for the PCP site of the NMDA receptor than does (R)-ketamine (in guinea pig brain).{{Citation needed|date=December 2008}} [[Esketamine|(S)-ketamine]] seems to induce drowsiness more strongly than the (R) enantiomer; it is probable that (R)-ketamine is the stronger sigma agonist and so this enantiomer is likely to be responsible for the lowering of the seizure threshold that can occur with ketamine.{{Citation needed|date=December 2008}} Since (S)-ketamine has greater analgesic effects and less hallucinogenic side effects than (R)-ketamine, the pure (S) enantiomer is sometimes preferred to the racemic mix for use in medical procedures, especially when lower doses are used for minor surgical procedures where the patient remains conscious during the operation.<ref>{{Cite journal|author=Togal T, Demirbilek S, Koroglu A, Yapici E, Ersoy O |title=Effects of S(+) ketamine added to bupivacaine for spinal anaesthesia for prostate surgery in elderly patients |journal=European Journal of Anaesthesiology |volume=21 |issue=3 |pages=193–7 |year=2004 |month=March |pmid=15055891}}</ref>
  
 
<div class="center">
 
<div class="center">
Line 195: Line 196:
 
The effects seem to take place mainly in the [[hippocampus|hippocampal formation]] and in the [[prefrontal cortex]]. This evidence, along with the NMDA receptor's connection with the memory formation process, explains ketamine's profound effects on [[memory]] and thought. These effects inhibit the filtering function of the brain and may mirror the sensory overload associated with [[schizophrenia]] and [[near death experience]]s.{{Citation needed|date=February 2007}}
 
The effects seem to take place mainly in the [[hippocampus|hippocampal formation]] and in the [[prefrontal cortex]]. This evidence, along with the NMDA receptor's connection with the memory formation process, explains ketamine's profound effects on [[memory]] and thought. These effects inhibit the filtering function of the brain and may mirror the sensory overload associated with [[schizophrenia]] and [[near death experience]]s.{{Citation needed|date=February 2007}}
  
The local anesthetic effects are likely from the blocking action of ketamine on [[sodium channel]]s.<ref>{{cite journal |author=Benoit E |title=Effects of intravenous anaesthetics on nerve axons |journal=European Journal of Anaesthesiology |volume=12 |issue=1 |pages=59–70 |year=1995 |month=January |pmid=7705328}}</ref>  Its ''in vitro'' blocking potency of sodium channels in the resting state is similar to that of [[lidocaine]].<ref>{{cite journal |author=Wagner LE, Gingrich KJ, Kulli JC, Yang J |title=Ketamine blockade of voltage-gated sodium channels: evidence for a shared receptor site with local anesthetics |journal=Anesthesiology |volume=95 |issue=6 |pages=1406–13 |year=2001 |month=December |pmid=11748399 |url=http://meta.wkhealth.com/pt/pt-core/template-journal/lwwgateway/media/landingpage.htm?issn=0003-3022&volume=95&issue=6&spage=1406}}</ref>
+
The local anesthetic effects are likely from the blocking action of ketamine on [[sodium channel]]s.<ref>{{Cite journal|author=Benoit E |title=Effects of intravenous anaesthetics on nerve axons |journal=European Journal of Anaesthesiology |volume=12 |issue=1 |pages=59–70 |year=1995 |month=January |pmid=7705328}}</ref>  Its ''in vitro'' blocking potency of sodium channels in the resting state is similar to that of [[lidocaine]].<ref>{{Cite journal|author=Wagner LE, Gingrich KJ, Kulli JC, Yang J |title=Ketamine blockade of voltage-gated sodium channels: evidence for a shared receptor site with local anesthetics |journal=Anesthesiology |volume=95 |issue=6 |pages=1406–13 |year=2001 |month=December |pmid=11748399 |url=http://meta.wkhealth.com/pt/pt-core/template-journal/lwwgateway/media/landingpage.htm?issn=0003-3022&volume=95&issue=6&spage=1406}}</ref>
  
Ketamine has a well-documented neuroprotective effect against ischemic brain-injury and glutamate induced brain injury.<ref>{{cite journal |author=Shibuta S, Varathan S, Mashimo T |title=Ketamine and thiopental sodium: individual and combined neuroprotective effects on cortical cultures exposed to NMDA or nitric oxide |journal=British Journal of Anaesthesia |volume=97 |issue=4 |pages=517–24 |year=2006 |month=October |pmid=16928698 |doi=10.1093/bja/ael192}}</ref>
+
Ketamine has a well-documented neuroprotective effect against ischemic brain-injury and glutamate induced brain injury.<ref>{{Cite journal|author=Shibuta S, Varathan S, Mashimo T |title=Ketamine and thiopental sodium: individual and combined neuroprotective effects on cortical cultures exposed to NMDA or nitric oxide |journal=British Journal of Anaesthesia |volume=97 |issue=4 |pages=517–24 |year=2006 |month=October |pmid=16928698 |doi=10.1093/bja/ael192}}</ref>
 
One hypothesis of its working mechanism in case of chronic pain management and depression is that it works as an antidote to an overactivity in glutamergic brain circuits.
 
One hypothesis of its working mechanism in case of chronic pain management and depression is that it works as an antidote to an overactivity in glutamergic brain circuits.
  
Line 204: Line 205:
 
{{Refimprove|section|date=December 2008}}
 
{{Refimprove|section|date=December 2008}}
 
[[Image:Ketamine Crystals.jpg|thumb|left|10&nbsp;ml bottle of liquid ketamine drying into crystals]]
 
[[Image:Ketamine Crystals.jpg|thumb|left|10&nbsp;ml bottle of liquid ketamine drying into crystals]]
[[Image:Ketamine.JPG|thumb|right|500&nbsp;mg ketamine hydrochloride]]  
+
[[Image:Ketamine.JPG|thumb|right|500&nbsp;mg ketamine hydrochloride]]
 
[[Image:Ketaminehydrochloride.jpg|thumb|right|Ketamine dried and scraped]]
 
[[Image:Ketaminehydrochloride.jpg|thumb|right|Ketamine dried and scraped]]
Ketamine sold illicitly comes from diverted legitimate supplies and semi-legitimate suppliers, theft from legitimate suppliers, and is imported in varying amounts from countries in which it is less strictly regulated.
+
Ketamine sold illicitly comes either from diverted legitimate supplies and semi-legitimate suppliers, or from theft from legitimate suppliers. It is imported in varying amounts from countries in which it is less strictly regulated.
  
In 2003, the US [[Drug Enforcement Agency]] conducted [[Operation TKO]], a probe into the quality of ketamine being imported from [[Mexico]].<ref name="SI-TKO">{{cite news| url=http://sportsillustrated.cnn.com/2007/more/07/18/steroids0424/1.html | work=CNN | title=SI.com - The Mexican Connection - Jul 18, 2007 | accessdate=7 May 2010}}</ref> As a result of operation TKO, US and Mexican authorities shut down the Mexico City company Laboratorios Ttokkyo, which was the biggest producer of ketamine in Mexico. According to the DEA, over 80% of ketamine seized in the US is of Mexican origin.<ref name="SI-TKO"/> The [[World Health Organization]] Expert Committee on Drug Dependence, in its thirty-third report (2003),<ref>[http://whqlibdoc.who.int/trs/WHO_TRS_915.pdf Untitled-59<!-- Bot generated title -->]</ref> recommended research into its recreational use/misuse due to growing concerns about its rising popularity in Europe, Asia and North America. This is due in part to its prevention of depression.<ref>[http://abcnews.go.com/Health/Depression/story?id=2282814&page=1 ABC News: Ketamine, a Tranquilizer and Popular Club Drug, May Work for Depression<!-- Bot generated title -->]</ref>
+
In 2003, the US [[Drug Enforcement Agency]] conducted [[Operation TKO]], a probe into the quality of ketamine being imported from [[Mexico]].<ref name="SI-TKO">{{Cite news| url=http://sportsillustrated.cnn.com/2007/more/07/18/steroids0424/1.html | work=CNN | title=SI.com - The Mexican Connection - Jul 18, 2007 | accessdate=7 May 2010}}</ref> As a result of operation TKO, US and Mexican authorities shut down the Mexico City company Laboratorios Ttokkyo, which was the biggest producer of ketamine in Mexico. According to the DEA, over 80% of ketamine seized in the US is of Mexican origin.<ref name="SI-TKO"/> The [[World Health Organization]] Expert Committee on Drug Dependence, in its thirty-third report (2003),<ref>[http://whqlibdoc.who.int/trs/WHO_TRS_915.pdf Untitled-59<!-- Bot generated title -->]</ref> recommended research into its recreational use/misuse due to growing concerns about its rising popularity in Europe, Asia and North America. This is due in part to its prevention of depression.<ref>[http://abcnews.go.com/Health/Depression/story?id=2282814&page=1 ABC News: Ketamine, a Tranquilizer and Popular Club Drug, May Work for Depression<!-- Bot generated title -->]</ref>
  
In ''[[E for Ecstasy]]''<ref>{{cite book |author=Saunders N, Heron L |year=1993 |title=E for Ecstasy |publisher=N. Saunders |location=London |isbn=0950162884}}{{pn}}</ref> (a book examining the uses of the street drug [[Methylenedioxymethamphetamine|Ecstasy]] in the UK)  the writer, activist and Ecstasy advocate [[Nicholas Saunders (activist)|Nicholas Saunders]] highlighted test results showing that certain consignments of the drug also contained ketamine. Consignments of Ecstasy known as "Strawberry" contained what Saunders described as a "potentially dangerous combination of ketamine, [[ephedrine]] and [[selegiline]]", as did a consignment of "Sitting Duck" Ecstasy tablets.<ref>See: [http://ecstasy.org/testing/pillstilJuly.html] for details online.</ref>  
+
In the 1993 book ''[[E for Ecstasy]]''<ref>{{Cite book|author=Saunders N, Heron L |year=1993 |title=E for Ecstasy |publisher=N. Saunders |location=London |isbn=0950162884}}{{Page needed|date=September 2010}}</ref> (about the uses of the street drug [[Methylenedioxymethamphetamine|Ecstasy]] in the UK)  the writer, activist and Ecstasy advocate [[Nicholas Saunders (activist)|Nicholas Saunders]] highlighted test results showing that certain consignments of the drug also contained ketamine. Consignments of Ecstasy known as "Strawberry" contained what Saunders described as a "potentially dangerous combination of ketamine, [[ephedrine]] and [[selegiline]]", as did a consignment of "Sitting Duck" Ecstasy tablets.<ref>See: [http://ecstasy.org/testing/pillstilJuly.html] for details online.</ref>
  
The former chairman of the British [[Advisory Council on the Misuse of Drugs]], [[David Nutt]], suggested that Ketamine should be upgraded from a class C drug due to the harm it can cause to users.<ref>{{cite news|url=http://www.guardian.co.uk/politics/2009/nov/01/david-nutt-gordon-brown-drugs |title=Ministers face rebellion over drug tsar's sacking &#124; Politics &#124; The Observer |publisher=Guardian |date= 1 November 2009|accessdate=2010-01-22 | location=London | first=Jamie | last=Doward}}</ref>
+
The former chairman of the British [[Advisory Council on the Misuse of Drugs]], [[David Nutt]], suggested that Ketamine should be upgraded from a class C drug due to the harm it can cause to users.<ref>{{Cite news|url=http://www.guardian.co.uk/politics/2009/nov/01/david-nutt-gordon-brown-drugs |title=Ministers face rebellion over drug tsar's sacking &#124; Politics &#124; The Observer |publisher=Guardian |date= 1 November 2009|accessdate=2010-01-22 | location=London | first=Jamie | last=Doward}}</ref>
  
 
===Methods of use===
 
===Methods of use===
Line 220: Line 221:
  
 
===Detection of use===
 
===Detection of use===
Ketamine may be quantitated in blood or plasma to confirm a diagnosis of poisoning in hospitalized patients, provide evidence in an impaired driving arrest or to assist in a medicolegal death investigation. Blood or plasma ketamine concentrations are usually in a range of 0.5-5.0 mg/L in persons receiving the drug therapeutically (during general anesthesia), 1-2 mg/L in those arrested for impaired driving and 3-20 mg/L in victims of acute fatal overdosage. Urine is often the preferred specimen for routine drug abuse monitoring purposes. The presence of norketamine, a pharmacologically-active metabolite, is useful for confirmation of ketamine ingestion.<ref>Feng N, Vollenweider FX, Minder EI, Rentsch K, Grampp T, Vonderschmitt DJ. Development of a gas chromatography-mass spectrometry method for determination of ketamine in plasma and its application to human samples. Ther. Drug Monit. 17: 95-100, 1995.</ref><ref>Parkin MC, Turfus SC, Smith NW, Halket JM, Braithwaite RA, Elliott SP, Osselton MD, Cowan DA, Kicman AT. Detection of ketamine and its metabolites in urine by ultra high pressure liquid chromatography-tandem mass spectrometry. J. Chrom. B 876: 137-142, 2008.</ref><ref>R. Baselt, ''Disposition of Toxic Drugs and Chemicals in Man'', 8th edition, Biomedical Publications, Foster City, CA, 2008, pp. 806-808.</ref>
+
Ketamine may be quantitated in blood or plasma to confirm a diagnosis of poisoning in hospitalized patients, provide evidence in an impaired driving arrest or to assist in a medicolegal death investigation. Blood or plasma ketamine concentrations are usually in a range of 0.5-5.0&nbsp;mg/L in persons receiving the drug therapeutically (during general anesthesia), 1–2&nbsp;mg/L in those arrested for impaired driving and 3–20&nbsp;mg/L in victims of acute fatal overdosage. Urine is often the preferred specimen for routine drug abuse monitoring purposes. The presence of norketamine, a pharmacologically-active metabolite, is useful for confirmation of ketamine ingestion.<ref>Feng N, Vollenweider FX, Minder EI, Rentsch K, Grampp T, Vonderschmitt DJ. Development of a gas chromatography-mass spectrometry method for determination of ketamine in plasma and its application to human samples. Ther. Drug Monit. 17: 95-100, 1995.</ref><ref>Parkin MC, Turfus SC, Smith NW, Halket JM, Braithwaite RA, Elliott SP, Osselton MD, Cowan DA, Kicman AT. Detection of ketamine and its metabolites in urine by ultra high pressure liquid chromatography-tandem mass spectrometry. J. Chrom. B 876: 137-142, 2008.</ref><ref>R. Baselt, ''Disposition of Toxic Drugs and Chemicals in Man'', 8th edition, Biomedical Publications, Foster City, CA, 2008, pp. 806-808.</ref>
  
 
===Psychological effects===
 
===Psychological effects===
Ketamine produces effects similar to [[phencyclidine]] (PCP) and [[dextromethorphan]] (DXM). Unlike the other well known dissociatives PCP and DXM, ketamine is very short acting, its [[Psychedelics, dissociatives and deliriants|hallucinatory]] effects lasting sixty minutes when insufflated or injected and up to two hours when ingested, the total experience lasting no more than a couple of hours.<ref>{{cite journal |author=Giannini AJ, Loiselle RH, Giannini MC, Price WA |title=Phencyclidine and the dissociatives |journal=Psychiatric Medicine |volume=3 |issue=3 |pages=197–217 |year=1985 |pmid=2893430}}</ref> Like other dissociative anaesthetics, hallucinations caused by ketamine are fundamentally different from those caused by [[tryptamine]]s and [[phenethylamine]]s. At low doses, hallucinations are only seen when one is in a dark room with one's eyes closed, while at medium to high doses the effects are far more intense and obvious.<ref>AJ Giannini,RH Loisellle, MC Giannini, WA Price, 1987, op. cit. The dissociatives. Medical Psychiatry. 3(3):197-205, 1987{{Verify source|what's the title or other bibliographic info?|date=December 2009}}</ref>
+
Ketamine produces effects similar to [[phencyclidine]] (PCP) and [[dextromethorphan]] (DXM). Unlike the other well known dissociatives PCP and DXM, ketamine is very short acting, its [[Psychedelics, dissociatives and deliriants|hallucinatory]] effects lasting sixty minutes when insufflated or injected and up to two hours when ingested, the total experience lasting no more than a couple of hours.<ref>{{Cite journal|author=Giannini AJ, Loiselle RH, Giannini MC, Price WA |title=Phencyclidine and the dissociatives |journal=Psychiatric Medicine |volume=3 |issue=3 |pages=197–217 |year=1985 |pmid=2893430}}</ref> Like other dissociative anaesthetics, hallucinations caused by ketamine are fundamentally different from those caused by [[tryptamine]]s and [[phenethylamine]]s. At low doses, hallucinations are only seen when one is in a dark room with one's eyes closed, while at medium to high doses the effects are far more intense and obvious.<ref>AJ Giannini,RH Loisellle, MC Giannini, WA Price, 1987, op. cit. The dissociatives. Medical Psychiatry. 3(3):197-205, 1987{{Verify source|what's the title or other bibliographic info?|date=December 2009}}</ref>
  
Ketamine produces a dissociative state, characterised by a sense of detachment from one's physical body and the external world which is known as [[depersonalization]] and [[derealization]].<ref>{{cite journal |author=Giannini AJ, Underwood NA, Condon M |title=Acute ketamine intoxication treated by haloperidol: a preliminary study |journal=American Journal of Therapeutics |volume=7 |issue=6 |pages=389–91 |year=2000 |month=November |pmid=11304647 |doi=10.1097/00045391-200007060-00008}}</ref> At sufficiently high doses (e.g. 150&nbsp;mg intramuscular), users may experience what is coined the "[[K-hole]]", a state of dissociation whose effects are thought to mimic the phenomenology of schizophrenia.<ref>{{cite book |author=Giannini AJ |title=Drug Abuse |publisher=Health Information Press|location= Los Angeles|year=1999 |pages=104 |isbn=1-885987-11-0}}{{pn}}</ref> Users may experience worlds or dimensions that are ineffable, all the while being completely unaware of their individual identities or the external world. Users have reported intense hallucinations including visual hallucinations, perceptions of falling, fast and gradual movement and flying, 'seeing God', feeling connected to other users, objects and the cosmos, experiencing psychotic reactions, and shared hallucinations, and thoughts with adjacent users. [[John C. Lilly]],<ref>{{cite book |author=Lilly, John Cunningham |title=The scientist: a metaphysical autobiography |publisher=Ronin Pub |location=Berkeley, CA |year=1997 |pages= |isbn=0-914171-72-0}}{{pn}}</ref> [[Marcia Moore]]<ref>{{cite book |author=Alltounian, Howard Sunny; Marcia Moore |title=Journeys into the bright world |publisher=Para Research |location=Rockport, Mass |year=1978 |pages= |isbn=978-0-914918-12-7}}{{pn}}</ref> and [[D. M. Turner]]<ref>{{cite book |author=Turner, D. M. |title=The essential psychedelic guide |publisher=Panther Press |location=San Francisco, CA |year=1994 |pages= |isbn=0-9642636-1-0}}{{pn}}</ref> (among others) have written extensively about their own [[spirituality|spiritual]]/[[psychonaut]]ic use of ketamine. (Both Moore<ref>{{cite book|title=Ketamine: Dreams and Realities|last=Jansen|first=Karl|publisher=Multidisciplinary Association for Psychedelic Studies|isbn=0966001931|year=2001|page=50}}</ref> and Turner<ref>{{cite book|title=Ketamine: Dreams and Realities|last=Jansen|first=Karl|publisher=Multidisciplinary Association for Psychedelic Studies|isbn=0966001931|year=2001|page=89}}</ref> died prematurely in a way that has been linked to their ketamine use.)
+
Ketamine produces a dissociative state, characterised by a sense of detachment from one's physical body and the external world which is known as [[depersonalization]] and [[derealization]].<ref>{{Cite journal|author=Giannini AJ, Underwood NA, Condon M |title=Acute ketamine intoxication treated by haloperidol: a preliminary study |journal=American Journal of Therapeutics |volume=7 |issue=6 |pages=389–91 |year=2000 |month=November |pmid=11304647 |doi=10.1097/00045391-200007060-00008}}</ref> At sufficiently high doses (e.g. 150&nbsp;mg intramuscular), users may experience what is coined the "[[K-hole]]", a state of dissociation whose effects are thought to mimic the phenomenology of schizophrenia.<ref>{{Cite book|author=Giannini AJ |title=Drug Abuse |publisher=Health Information Press|location= Los Angeles|year=1999 |pages=104 |isbn=1-885987-11-0}}{{Page needed|date=September 2010}}</ref> Users may experience worlds or dimensions that are ineffable, all the while being completely unaware of their individual identities or the external world. Users have reported intense hallucinations including visual hallucinations, perceptions of falling, fast and gradual movement and flying, 'seeing God', feeling connected to other users, objects and the cosmos, experiencing psychotic reactions, and shared hallucinations, and thoughts with adjacent users. [[John C. Lilly]],<ref>{{Cite book|author=Lilly, John Cunningham |title=The scientist: a metaphysical autobiography |publisher=Ronin Pub |location=Berkeley, CA |year=1997 |pages= |isbn=0-914171-72-0}}{{Page needed|date=September 2010}}</ref> [[Marcia Moore]]<ref>{{Cite book|author=Alltounian, Howard Sunny; Marcia Moore |title=Journeys into the bright world |publisher=Para Research |location=Rockport, Mass |year=1978 |pages= |isbn=978-0-914918-12-7}}{{Page needed|date=September 2010}}</ref> and [[D. M. Turner]]<ref>{{Cite book|author=Turner, D. M. |title=The essential psychedelic guide |publisher=Panther Press |location=San Francisco, CA |year=1994 |pages= |isbn=0-9642636-1-0}}{{Page needed|date=September 2010}}</ref> (among others) have written extensively about their own [[spirituality|spiritual]]/[[psychonaut]]ic use of ketamine. (Both Moore<ref>{{Cite book|title=Ketamine: Dreams and Realities|last=Jansen|first=Karl|publisher=Multidisciplinary Association for Psychedelic Studies|isbn=0966001931|year=2001|page=50}}</ref> and Turner<ref>{{Cite book|title=Ketamine: Dreams and Realities|last=Jansen|first=Karl|publisher=Multidisciplinary Association for Psychedelic Studies|isbn=0966001931|year=2001|page=89}}</ref> died prematurely in a way that has been linked to their ketamine use.)
  
 
Users may feel as though their perceptions are located so deep inside the mind that the real world seems distant (hence the use of a "hole" to describe the experience). Some users may not remember this part of the experience after regaining consciousness, in the same way that a person may forget a dream. Owing to the role of the NMDA receptor in long-term potentiation, this may be due to disturbances in memory formation. The "re-integration" process is slow, and the user gradually becomes aware of surroundings. At first, users may not remember their own names, or even know that they are human, or what that means. Movement is extremely difficult, and a user may not be aware that he or she has a body at all.
 
Users may feel as though their perceptions are located so deep inside the mind that the real world seems distant (hence the use of a "hole" to describe the experience). Some users may not remember this part of the experience after regaining consciousness, in the same way that a person may forget a dream. Owing to the role of the NMDA receptor in long-term potentiation, this may be due to disturbances in memory formation. The "re-integration" process is slow, and the user gradually becomes aware of surroundings. At first, users may not remember their own names, or even know that they are human, or what that means. Movement is extremely difficult, and a user may not be aware that he or she has a body at all.
  
 
==Long-term side effects==
 
==Long-term side effects==
 +
=== Neurological Effects ===
 
{{Main|Olney's lesions}}
 
{{Main|Olney's lesions}}
Chronic use of ketamine may lead to cognitive impairments including memory problems.<ref>{{cite journal |author=Okon T |title=Ketamine: an introduction for the pain and palliative medicine physician |journal=Pain Physician |volume=10 |issue=3 |pages=493–500 |year=2007 |month=May |pmid=17525784 |url=http://www.painphysicianjournal.com/linkout_vw.php?issn=1533-3159&vol=10&page=493}}</ref> In 1989, psychiatry professor [[John Olney]] reported that ketamine caused reversible changes in two small areas of the rat brain. 40&nbsp;mg/kg resulted in fluid-filled cavities ("vacuoles") appearing inside cells. The cavities disappeared after several days, unless high doses of the far more toxic PCP or close relative MK801 were repeatedly given, in which case some cell death was seen. Roland Auer injected monkeys with MK801 and was unable to produce any vacuoles. When Auer was asked in 1998 whether persons undergoing anesthesia with Ketalar were at risk of these changes, his reply was that he doubted that it was even a remote possibility because of fundamental differences in metabolism between the rat and human brain. Ketamine can block excitotoxicity (brain damage due to low oxygen, low sugar, epilepsy, trauma, etc.) but it can also excite the brain at low doses by switching off the inhibitory system. Why this is not damaging in monkeys and humans probably lies in the fact that ketamine binds to an increasingly wide range of different receptors as the dose level rises, and some of these receptors act to shut down the excitement. In humans, by the time a potentially toxic dose is reached, the "excitement window" has been passed and the drug is starting to activate other systems that switch cells off again, a result of ketamine's promiscuity that improves its safety relative to MK801. MK801 binds very specifically to N-P receptors. The other part of the explanation is that rats have rates of brain metabolism that are almost twice as high as those in humans to start with. It is because of this higher base rate of metabolism that ketamine causes over-excitement in rats at doses below those at which it activates shutdown systems.<ref>{{cite journal |author=Olney JW, Labruyere J, Price MT |title=Pathological changes induced in cerebrocortical neurons by phencyclidine and related drugs |journal=Science |volume=244 |issue=4910 |pages=1360–2 |year=1989 |month=June |pmid=2660263 |doi=10.1126/science.2660263}}</ref><ref name=dxm_health>[http://www.erowid.org/chemicals/dxm/dxm_health2.shtml Erowid DXM Vaults: Health: The Bad News Isn't In: A Look at Dissociative-Induced Brain Damage, by Anderson C<!-- Bot generated title -->]</ref><ref name=Tryba_et_al>{{cite journal |author=Tryba M, Gehling M |title=Clonidine--a potent analgesic adjuvant |journal=Current Opinion in Anaesthesiology |volume=15 |issue=5 |pages=511–7 |year=2002 |month=October |pmid=17019247 |url=http://meta.wkhealth.com/pt/pt-core/template-journal/lwwgateway/media/landingpage.htm?issn=0952-7907&volume=15&issue=5&spage=511 |doi=10.1097/00001503-200210000-00007}}</ref>
+
Chronic use of ketamine may lead to cognitive impairments including memory problems.<ref>{{Cite journal|author=Okon T |title=Ketamine: an introduction for the pain and palliative medicine physician |journal=Pain Physician |volume=10 |issue=3 |pages=493–500 |year=2007 |month=May |pmid=17525784 |url=http://www.painphysicianjournal.com/linkout_vw.php?issn=1533-3159&vol=10&page=493}}</ref> In 1989, psychiatry professor [[John Olney]] reported that ketamine caused reversible changes in two small areas of the rat brain. 40&nbsp;mg/kg resulted in fluid-filled cavities ("vacuoles") appearing inside cells. The cavities disappeared after several days, unless high doses of the far more toxic PCP or close relative MK801 were repeatedly given, in which case some cell death was seen. Roland Auer injected monkeys with MK801 and was unable to produce any vacuoles. When Auer was asked in 1998 whether persons undergoing anesthesia with Ketalar were at risk of these changes, his reply was that he doubted that it was even a remote possibility because of fundamental differences in metabolism between the rat and human brain. Ketamine can block [[excitotoxicity]] (brain damage due to low oxygen, low sugar, epilepsy, trauma, etc.) but it can also excite the brain at low doses by switching off the inhibitory system. Why this is not damaging in monkeys and humans probably lies in the fact that ketamine binds to an increasingly wide range of different receptors as the dose level rises, and some of these receptors act to shut down the excitement. In humans, by the time a potentially toxic dose is reached, the "excitement window" has been passed and the drug is starting to activate other systems that switch cells off again, a result of ketamine's promiscuity that improves its safety relative to MK801. MK801 binds very specifically to N-P receptors. The other part of the explanation is that rats have rates of brain metabolism that are almost twice as high as those in humans to start with. It is because of this higher base rate of metabolism that ketamine causes over-excitement in rats at doses below those at which it activates shutdown systems.<ref>{{Cite journal|author=Olney JW, Labruyere J, Price MT |title=Pathological changes induced in cerebrocortical neurons by phencyclidine and related drugs |journal=Science |volume=244 |issue=4910 |pages=1360–2 |year=1989 |month=June |pmid=2660263 |doi=10.1126/science.2660263}}</ref><ref name=dxm_health>[http://www.erowid.org/chemicals/dxm/dxm_health2.shtml Erowid DXM Vaults: Health: The Bad News Isn't In: A Look at Dissociative-Induced Brain Damage, by Anderson C<!-- Bot generated title -->]</ref><ref name=Tryba_et_al>{{Cite journal|author=Tryba M, Gehling M |title=Clonidine--a potent analgesic adjuvant |journal=Current Opinion in Anaesthesiology |volume=15 |issue=5 |pages=511–7 |year=2002 |month=October |pmid=17019247 |url=http://meta.wkhealth.com/pt/pt-core/template-journal/lwwgateway/media/landingpage.htm?issn=0952-7907&volume=15&issue=5&spage=511 |doi=10.1097/00001503-200210000-00007}}</ref>
  
The first large-scale, longitudinal study of ketamine users found that heavy ketamine users had impaired memory by several measures, including verbal, short-term memory and visual memory. However occasional (1-2 times per month) ketamine users and ex ketamine users were not found to differ from controls in memory, attention and psychological well-being tests. This suggests that occasional use of ketamine does not lead to prolonged harm and that any damage that might occur may be reversible when ketamine use is stopped.<ref name="Addiction Users Study">{{cite journal |doi=10.1111/j.1360-0443.2009.02761.x |title=Consequences of chronic ketamine self-administration upon neurocognitive function and psychological wellbeing: a 1-year longitudinal study |year=2009 |last1=Morgan |first1=Celia J. A. |last2=Muetzelfeldt |first2=Leslie |last3=Curran |first3=H. Valerie |journal=Addiction |volume=105 |pages=121}}</ref>
+
The first large-scale, longitudinal study of ketamine users found that heavy ketamine users had impaired memory by several measures, including verbal, short-term memory and visual memory. However occasional (1-2 times per month) ketamine users and ex ketamine users were not found to differ from controls in memory, attention and psychological well-being tests. This suggests that occasional use of ketamine does not lead to prolonged harm and that any damage that might occur may be reversible when ketamine use is stopped.<ref name="Addiction Users Study">{{Cite journal|doi=10.1111/j.1360-0443.2009.02761.x |pmid=19919593 |title=Consequences of chronic ketamine self-administration upon neurocognitive function and psychological wellbeing: a 1-year longitudinal study |year=2009 |last1=Morgan |first1=Celia J. A. |last2=Muetzelfeldt |first2=Leslie |last3=Curran |first3=H. Valerie |journal=Addiction |volume=105 |issue=1 |pages=121}}</ref>
  
Vutskits et al. from Geneva showed that short-term exposure of cultures to ketamine at concentrations of ≥ 20 μg/mL leads to a significant loss of differentiated cells and that non-cell death-inducing concentrations of ketamine (10 μg/mL) can still initiate long-term alterations of dendritic arbor in differentiated neurons, including dendritic retraction and branching point elimination. They also demonstrated that chronic (>24 h) administration of ketamine at concentrations as low as 0.01 μg/mL can interfere with the maintenance of dendritic arbor architecture. These results raise the possibility that chronic exposure to low, subanesthetic concentrations of ketamine, while not affecting cell survival, could still impair neuronal morphology and thus might lead to dysfunctions of neural networks.<ref>{{cite journal |author=Hargreaves RJ, Hill RG, Iversen LL |title=Neuroprotective NMDA antagonists: the controversy over their potential for adverse effects on cortical neuronal morphology |journal=Acta Neurochirurgica. Supplementum |volume=60 |issue= |pages=15–9 |year=1994 |pmid=7976530}}</ref>
+
Vutskits et al. from Geneva showed that short-term exposure of cultures to ketamine at concentrations of ≥ 20 μg/mL leads to a significant loss of differentiated cells and that non-cell death-inducing concentrations of ketamine (10 μg/mL) can still initiate long-term alterations of dendritic arbor in differentiated neurons, including dendritic retraction and branching point elimination. They also demonstrated that chronic (>24 h) administration of ketamine at concentrations as low as 0.01 μg/mL can interfere with the maintenance of dendritic arbor architecture. These results raise the possibility that chronic exposure to low, subanesthetic concentrations of ketamine, while not affecting cell survival, could still impair neuronal morphology and thus might lead to dysfunctions of neural networks.<ref>{{Cite journal|author=Hargreaves RJ, Hill RG, Iversen LL |title=Neuroprotective NMDA antagonists: the controversy over their potential for adverse effects on cortical neuronal morphology |journal=Acta Neurochirurgica. Supplementum |volume=60 |issue= |pages=15–9 |year=1994 |pmid=7976530}}</ref>
  
 
There is a long list of medicines that could counteract these potential toxic effects, including [[clonidine]], [[anticholinergics]], [[benzodiazepines]], [[barbiturates]] and [[risperidone]].<ref name=dxm_health /><ref name=Tryba_et_al /><ref>AJ Giannini, N Underwood, M Condon op. cit.{{Verify source|date=December 2009}}</ref>
 
There is a long list of medicines that could counteract these potential toxic effects, including [[clonidine]], [[anticholinergics]], [[benzodiazepines]], [[barbiturates]] and [[risperidone]].<ref name=dxm_health /><ref name=Tryba_et_al /><ref>AJ Giannini, N Underwood, M Condon op. cit.{{Verify source|date=December 2009}}</ref>
  
A study in [[Bristol]] reported in the British Medical Journal on 3 May 2008 linked urinary tract disease with ketamine use. Symptoms reported by users include an increased need to urinate, passing blood in urine, leakage of urine and pain on urination. These symptoms are associated with the scarification of the bladder lining, which leads to a shrunken bladder, [[erythema]], and contact bleeding, and can then move to the ureters and damage the kidneys.<ref>{{cite journal |doi=10.1136/bmj.39562.711713.80 |title=Urinary tract disease associated with chronic ketamine use |year=2008 |last1=Cottrell |first1=A. M |last2=Athreeres |first2=R. |last3=Weinstock |first3=P. |last4=Warren |first4=K. |last5=Gillatt |first5=D. |journal=BMJ |volume=336 |page=973}}</ref>
+
===Urinary Tract Effects===
 +
A study in [[Bristol]] reported in the British Medical Journal on 3 May 2008 linked urinary tract disease with ketamine use. Symptoms reported by users include an increased need to urinate, passing blood in urine, leakage of urine and pain on urination. These symptoms are associated with the scarification of the bladder lining, which leads to a shrunken bladder, [[erythema]], and contact bleeding, and can then move to the ureters and damage the kidneys.<ref>{{Cite journal|doi=10.1136/bmj.39562.711713.80 |title=Urinary tract disease associated with chronic ketamine use |year=2008 |last1=Cottrell |first1=A. M |last2=Athreeres |first2=R. |last3=Weinstock |first3=P. |last4=Warren |first4=K. |last5=Gillatt |first5=D. |journal=BMJ |volume=336 |page=973}}</ref>
  
In a study of 9 daily ketamine users, Shahani et al. found "marked thickening of the bladder wall, a small capacity, and perivesicular stranding, consistent with severe inflammation. At cystoscopy, all patients had severe ulcerative cystitis. Biopsies in 4 patients revealed epithelial denudation and inflammation with a mild eosinophilic infiltrate. Cessation of ketamine use, with the addition of [[pentosan polysulfate]], appeared to provide some symptomatic relief."<ref>{{cite journal |author=Shahani R, Streutker C, Dickson B, Stewart RJ |title=Ketamine-associated ulcerative cystitis: a new clinical entity |journal=Urology |volume=69 |issue=5 |pages=810–2 |year=2007 |month=May |pmid=17482909 |doi=10.1016/j.urology.2007.01.038}}</ref>
+
In a study of 9 daily ketamine users, Shahani et al. found "marked thickening of the bladder wall, a small capacity, and perivesicular stranding, consistent with severe inflammation. At cystoscopy, all patients had severe ulcerative cystitis. Biopsies in 4 patients revealed epithelial denudation and inflammation with a mild eosinophilic infiltrate. Cessation of ketamine use, with the addition of [[pentosan polysulfate]], appeared to provide some symptomatic relief."<ref>{{Cite journal|author=Shahani R, Streutker C, Dickson B, Stewart RJ |title=Ketamine-associated ulcerative cystitis: a new clinical entity |journal=Urology |volume=69 |issue=5 |pages=810–2 |year=2007 |month=May |pmid=17482909 |doi=10.1016/j.urology.2007.01.038}}</ref>
  
Many long term users report "K-pains" or "ketamine cramps"<ref>{{cite news|url=http://news.bbc.co.uk/newsbeat/hi/health/newsid_7831000/7831301.stm|title=Warning over rising ketamine use|last=Middleton|first=Duncan|date=15 January 2009|publisher=BBC|accessdate=19 January 2010}}</ref>. The exact cause of these pains are unknown. The Ketamine induced abdominal pain is primarily limited to users injecting more than a gram or more of ketamine a day. It has been suggested that the amino acid Tyrosine may help alleviate the pain.<ref>{{cite book|title=Ketamine: Dreams and Realities|last=Jansen|first=Karl|publisher=Multidisciplinary Association for Psychedelic Studies|isbn=0966001931|year=2001|page=271</ref> 
+
The bladder symptoms are so severe amongst some recreational users that their bladders have had to be removed. In these cases neobladders have been created using sections of gut but there is a significant risk of similar damage occurring to the neobladder if ketamine use is not stopped.
  
In October 2009, Professor [[David Nutt]], formerly of the British Home Office's [[Advisory Council on the Misuse of Drugs]], recommended that ketamine be re-classed in the UK, as several patients who have used the drug recreationally had experienced bladder spasms and some have "irreversible bladder damage". Professor Nutt also reported that a small number of patients have had their bladder completely removed through ketamine abuse. The recommendation and case studies were featured heavily in the British press.<ref>{{cite news|url=http://news.bbc.co.uk/1/hi/england/bristol/7867449.stm|title= Ketamine link to bladder failure|first=BBC News|date=4 February 2009|publisher=BBC News|accessdate=15 January 2010}}</ref>
+
Many long term users report "K-pains" or "ketamine cramps".<ref>{{Cite news|url=http://news.bbc.co.uk/newsbeat/hi/health/newsid_7831000/7831301.stm|title=Warning over rising ketamine use|last=Middleton|first=Duncan|date=15 January 2009|publisher=BBC|accessdate=19 January 2010}}</ref> The exact cause of these pains are unknown. The Ketamine induced abdominal pain is primarily limited to users of a gram or more of ketamine a day (route of administration does not seem to affect this symptom). It has been suggested that the amino acid Tyrosine may help alleviate the pain.<ref>{{Cite book|title=Ketamine: Dreams and Realities|last=Jansen|first=Karl|publisher=Multidisciplinary Association for Psychedelic Studies|isbn=0966001931|year=2001|page=271}}</ref>
  
== See also ==
+
In October 2009, Professor [[David Nutt]], formerly of the British Home Office's [[Advisory Council on the Misuse of Drugs]], recommended that ketamine be re-classed in the UK, as several patients who have used the drug recreationally had experienced bladder spasms and some have "irreversible bladder damage". Professor Nutt also reported that a small number of patients have had their bladder completely removed through ketamine abuse. The recommendation and case studies were featured heavily in the British press.<ref>{{Cite news|url=http://news.bbc.co.uk/1/hi/england/bristol/7867449.stm|title= Ketamine link to bladder failure|first=BBC News|date=4 February 2009|publisher=BBC News|accessdate=15 January 2010}}</ref>
 +
 
 +
==See also==
 
* [[Arylcyclohexylamine]]
 
* [[Arylcyclohexylamine]]
 +
* [[Esketamine]]
 
* [[Hallucinogen]]
 
* [[Hallucinogen]]
 
* [[Dissociation]]
 
* [[Dissociation]]
Line 267: Line 273:
 
{{Dopaminergics}}
 
{{Dopaminergics}}
 
{{Glutamatergics}}
 
{{Glutamatergics}}
 +
{{Use dmy dates|date=September 2010}}
  
 
[[Category:General anesthetics]]
 
[[Category:General anesthetics]]
Line 294: Line 301:
 
[[no:Ketamin]]
 
[[no:Ketamin]]
 
[[pl:Ketamina]]
 
[[pl:Ketamina]]
[[pt:Cloridrato de cetamina]]
+
[[pt:Ketamina]]
 
[[ru:Кетамин]]
 
[[ru:Кетамин]]
 
[[simple:Ketamine]]
 
[[simple:Ketamine]]

Latest revision as of 21:01, 24 September 2010

ketamine
150px
200px
Systematic (IUPAC) name
(RS)-2-(2-Chlorophenyl)-2-(methylamino)cyclohexan-1-one
Clinical data
Pregnancy
category
  • B
Routes of
administration
IV, IM, Insufflated, oral, topical
Legal status
Legal status
Pharmacokinetic data
Metabolism Hepatic, primarily by CYP3A4[1]
Biological half-life 2.5–3 hours.
Excretion renal (>90%)
Identifiers
CAS Number 6740-88-1
ATC code N01AX03 (WHO) N01AX14
PubChem CID 3821
DrugBank DB01221
ChemSpider 3689
Chemical data
Formula C13H16ClNO
Molar mass 237.725 g/mol[[Script error: No such module "String".]]
Script error: No such module "collapsible list".
Physical data
Melting point 262 to 263 °C (504 to 505 °F) (HCl)
  (verify)
Script error: No such module "TemplatePar".Expression error: Unexpected < operator.

Ketamine is a drug used in human and veterinary medicine developed by Parke-Davis (today a part of Pfizer) in 1962. Its hydrochloride salt is sold as Ketanest, Ketaset, and Ketalar. Pharmacologically, ketamine is classified as an NMDA receptor antagonist.[2] At high, fully anesthetic level doses, ketamine has also been found to bind to opioid μ receptors and sigma receptors.[3][4] Like other drugs of this class such as tiletamine and phencyclidine (PCP), it induces a state referred to as "dissociative anesthesia"[5] and is used as a recreational drug.

Ketamine has a wide range of effects in humans, including analgesia, anesthesia, hallucinations, elevated blood pressure, and bronchodilation.[6] Ketamine is primarily used for the induction and maintenance of general anesthesia, usually in combination with some sedative drug. Other uses include sedation in intensive care, analgesia (particularly in emergency medicine), and treatment of bronchospasm. It has been shown to be effective in treating depression in patients with bipolar disorder who have not responded to other anti-depressants.[7] It is also a popular anesthetic in veterinary medicine.

Ketamine is a chiral compound. Most pharmaceutical preparations of ketamine are racemic; however, some brands reportedly have (mostly undocumented) differences in enantiomeric proportions. The more active enantiomer, (S)-ketamine, is also available for medical use under the brand name Ketanest S.[8] However (R)-ketamine is also active, and has differences in effects; the S enantiomer being a dissociative and the R enantiomer a hallucinogen. (R)-ketamine, (S)-ketamine & racemic (R,S)-ketamine all have qualitatively separate distinct effect profiles, although S has the most active potency. Ketamine is a core medicine in the World Health Organization's "Essential Drugs List", which is a list of minimum medical needs for a basic health care system.[9]

History

Ketamine was discovered by Dr. Craig Newlands of Wayne State University. It was then developed by Parke-Davis in 1962 as part of an effort to find a safer anesthetic alternative to phencyclidine (PCP), which was likely to cause hallucinations, neurotoxicity and seizures. The drug was first given to American soldiers during the Vietnam War. It is still widely used in humans. It is also used widely in veterinary medicine, or as a battlefield anesthetic in developing nations.[10] The drug was used in psychiatric and other academic research through the 1970s, culminating in 1978 with the publishing of John Lilly's The Scientist and Marcia Moore and Howard Alltounian's Journeys into the Bright World, which documented the unusual phenomenology of ketamine intoxication.[11]

The incidence of recreational ketamine use increased through the end of the century, especially in the context of raves and other parties. The increase in illicit use prompted ketamine's placement in Schedule III of the United States Controlled Substance Act in August 1999.[12] In the United Kingdom, it became outlawed and labeled a Class C drug on 1 January 2006.[13] In Canada ketamine is classified as a Schedule I narcotic, as of August 2005.[14] In Hong Kong, as of year 2000, ketamine is regulated under Schedule 1 of Hong Kong Chapter 134 Dangerous Drugs Ordinance. It can only be used legally by health professionals, for university research purposes, or with a physician's prescription.[15]

Naming

Ketamine was invented as CL369, and it was referred to as CI 581 (clinical investigation 581) during development at Parke-Davis.[16] Commercial brands of ketamine include "Ketalar, Ketaset, Ketmex, Ketotal, Ketamine-500 (Astrapin) and Imalgen".[17] Ketamine is a keto-amine, so named as it has a ketone C=O group bonded to carbons either side, as well as an amine group. As such it resembles NMDA.

Production for recreational use has been traced to 1967, when it was referred to as "mean green" and "rockmesc".[18] Recreational names for ketamine include "K",[19] "Ket",[20] "Special K"[19] and "Vitamin K".[20]

Medical use

File:Ketamine 10ml bottle.jpg
10 ml bottles of ketamine (veterinary use)

Indications for use as an anaesthetic:

  • Pediatric anesthesia (as the sole anesthetic for minor procedures or as an induction agent followed by muscle relaxant and endotracheal intubation)
  • Asthmatics or patients with chronic obstructive airway disease
  • As part of a cream, gel, or liquid for topical application for nerve pain—the most common mixture is 10% ketoprofen, 5% Lidocaine, and 10% ketamine. Other ingredients found useful by pain specialists and their patients as well as the compounding pharmacists who make the topical mixtures include amitryptiline, cyclobenzaprine, clonidine, tramadol, and mepivicaine and other longer-acting local anaesthetics.
  • In emergency medicine in entrapped patients suffering severe trauma[21]
  • Emergency surgery in field conditions in war zones
  • To supplement spinal / epidural anesthesia / analgesia utilizing low doses

Contraindications

Effects

Impairs all senses, especially:

  • Sight
  • Balance
  • Sense of time

Cardiovascular:

  • Partial depressant

Gastrointestinal:

Musculoskeletal:

Neurological:

Respiratory:

  • Partial depressant/stimulant

In medical settings, ketamine is usually injected intravenously or intramuscularly,[22] but it is also effective when insufflated, smoked, or taken orally.[23]

Since it suppresses breathing much less than most other available anaesthetics,[24] ketamine is still used in human medicine as an anesthetic, however, due to the hallucinations which may be caused by ketamine, it is not typically used as a primary anesthetic, although it is the anaesthetic of choice when reliable ventilation equipment is not available. Ketamine tends to increase heart rate and blood pressure. Because ketamine tends to increase or maintain cardiac output, it is sometimes used in anesthesia for emergency surgery when the patient's fluid volume status is unknown (e.g., from traffic accidents). Ketamine can be used in podiatry and other minor surgery, and occasionally for the treatment of migraine. There is ongoing research in France, the Netherlands, Russia, Australia and the US into the drug's usefulness in pain therapy, depression suppression, and for the treatment of alcoholism[25] and heroin addiction.[26]

In veterinary anesthesia, ketamine is often used for its anesthetic and analgesic effects on cats, dogs, rabbits, rats, and other small animals. Veterinarians often use ketamine with sedative drugs to produce balanced anesthesia and analgesia, and as a constant rate infusion to help prevent pain wind-up. Ketamine is used to manage pain among large animals, though it has less effect on bovines. It is the primary intravenous anesthetic agent used in equine surgery, often in conjunction with detomidine and thiopental, or sometimes guaifenesin.

Ketamine may be used in small doses (0.1–0.5 mg/kg·h) as a local anesthetic, particularly for the treatment of pain associated with movement and neuropathic pain.[27] It may also be used as an intravenous co-analgesic together with opiates to manage otherwise intractable pain, particularly if this pain is neuropathic (pain due to vascular insufficiency or shingles are good examples). It has the added benefit of counter-acting spinal sensitization or wind-up phenomena experienced with chronic pain. At these doses, the psychotropic side effects are less apparent and well managed with benzodiazepines.[28] Ketamine is a co-analgesic, and so is most effective when used alongside a low-dose opioid; while it does have analgesic effects by itself, the higher doses required can cause disorienting side effects.[28] The combination of ketamine with an opioid is, however, particularly useful for pain caused by cancer.[29]

The effect of ketamine on the respiratory and circulatory systems is different from that of other anesthetics. When used at anesthetic doses, it will usually stimulate rather than depress the circulatory system.[30] It is sometimes possible to perform ketamine anesthesia without protective measures to the airways. Ketamine is also a potent analgesic and can be used in sub-anesthetic doses to relieve acute pain; however, its psychotropic properties must be taken into account. Patients have reported vivid hallucinations, "going into other worlds" or "seeing God" while anesthetized, and these unwanted psychological side-effects have reduced the use of ketamine in human medicine. They can, however, usually be avoided by concomitant application of a sedative such as a benzodiazepine.[28]

Low-dose ketamine is recognized for its potential effectiveness in the treatment of complex regional pain syndrome (CRPS), according to a retrospective review published in the October 2004 issue of Pain Medicine.[31] Although low-dose ketamine therapy is established as a generally safe procedure, reported side effects in some patients have included hallucinations, dizziness, lightheadedness and nausea. Therefore nurses administering ketamine to patients with CRPS should only do so in a setting where a trained physician is available if needed to assess potential adverse effects on patients.[32]

In some neurological ICUs, ketamine has been used in cases of prolonged status epilepticus. There has been some evidence that the NMDA-blocking effect of the drug protects neurons from glutamatergic damage during prolonged seizures.[33]

Experimental antidepressant use

When treating patients suffering from complex regional pain syndrome (CRPS) with a low-dose (subanesthetic) ketamine infusion, it was observed that some patients made a significant recovery from associated depression. This recovery was not formally documented, as the primary concern was the treatment of the patient's pain. It was not possible to quantify to what degree depression recovery was secondary to the patient's recovery from CRPS. Based on this result, it was thought that a low-dose (subanesthetic) infusion of ketamine was worth a trial in patients who were suffering from treatment-resistant depression without other physical or psychiatric illness.

Correll, et al. gave ketamine intravenously to patients commencing at 15–20 mg/h (0.1–0.2 mg/kg/h) and the dose increased until a maximum tolerated dose was achieved. This dose was assumed to be a therapeutic dose and was maintained for 5 days. Patients were able to eat, drink, watch television, or read. They could feel inebriated and/or unsteady when walking. If hallucinations occurred, the dose was to be reduced. The patients' normal medications were continued as it was feared that stopping them might result in severe depressive episodes. Before and following each treatment with ketamine, at patient clinic visits, the Beck Depression Inventory (BDI) and the Hamilton Rating Scale for Depression (HAMD-17) were obtained. Two of the patients were described with "impressive", as described by an attending doctor, improvement in depression being maintained for 12 months in patient A and recurrence at 2.5 months and 9 months in patient B.[34]

The National Institute of Health News reports that a study of 18 patients has found that ketamine significantly improved treatment-resistant major depression within hours of injection.[35] The improvement lasted up to one week after the single dose.[36] The patients in the study were previously treatment resistant, having tried an average of six other treatments that failed. NIMH director Dr. Thomas Insel said in the paper:

"To my knowledge, this is the first report of any medication or other treatment that results in such a pronounced, rapid, prolonged response with a single dose. These were very treatment-resistant patients."

The researchers apparently attribute the effect to ketamine being an NMDA receptor antagonist.[37] Those findings of Zarate et al. corroborate earlier findings by Berman et al..[38] However Zarate et al. do raise some concerns about their results due to a possible lack of blinding, because of the inebriating effects of low dose ketamine infusion, and it is recommended that future studies include an active placebo.

The findings by Zarate et al. are confirmed by Liebrenz et al., who substantially, according to an attending doctor, helped a 55-year-old male subject with a treatment-resistant major depression and a co-occurring alcohol and benzodiazepine dependence by giving an intravenous infusion of 0.5 mg/kg ketamine over a period of 50 minutes and Goforth et al. who helped a patient with severe, recurrent major depressive disorder that demonstrated marked improvement within 8 hours of receiving a preoperative dose of ketamine and one treatment of electroconvulsive therapy with bitemporal electrode placement.[39][40]

However, a new study in mice by Zarate et al. shows that blocking the NMDA receptor is an intermediate step. According to this study, blocking NMDA increases the activity of another receptor, AMPA, and this boost in AMPA activity is crucial for ketamine’s rapid antidepressant actions. NMDA and AMPA are receptors for the neurotransmitter glutamate. The glutamate system has been implicated in depression recently. This is a departure from previous thinking, which had focused on serotonin and norepinephrine. The glutamate system may represent a new avenue for treatment and research.[41]

Krystal et al. retrospectively compared the seizure duration, ictal EEG, and cognitive side effects of ketamine and methohexital anesthesia with ECT in 36 patients.[42] Ketamine was well tolerated and prolonged seizure duration overall, but particularly in those who had a seizure duration shorter than 25 seconds with methohexital at the maximum available stimulus intensity. Ketamine also increased midictal EEG slow-wave amplitude. Thus, a switch to ketamine may be useful when it is difficult to elicit a robust seizure. Faster post-treatment reorientation with ketamine may suggest a lower level of associated cognitive side effects.

Kudoh et al. investigated whether ketamine is suitable for depressed patients who had undergone orthopedic surgery.[43] They studied 70 patients with major depression and 25 patients as the control (Group C). The depressed patients were divided randomly into two groups; patients in Group A, initial HAMD 12,7 (n = 35) were induced with propofol, fentanyl, and ketamine and patients in Group B, initial HAMD 12,3 (n = 35) were induced with propofol and fentanyl. Depressed mood, suicidal tendencies, somatic anxiety, and hypochondriasis significantly decreased in Group A as compared with Group B. The group receiving ketamine also had significantly lower postoperative pain.

Acute administration of ketamine at the higher dose, but not imipramine, increased BDNF protein levels in the rat hippocampus. The increase of hippocampal BDNF protein levels induced by ketamine might be necessary to produce a rapid onset of antidepressant action.[44]

Treatment of addiction

The Russian doctor Evgeny Krupitsky (Clinical Director of Research for the Saint Petersburg Regional Center for Research in Addiction and Psychopharmacology) has claimed to have encouraging results by using ketamine as part of a treatment for alcohol addiction which combines psychedelic and aversive techniques.[45][46] This method involved psychotherapy, controlled ketamine use and group therapy, and resulted in 60 of the 86 alcoholic males selected for the study remaining fully abstinent through one year of treatment. He has also treated heroin addicts and reached the conclusion that one ketamine-assisted psychotherapy session was significantly more effective than active placebo in promoting abstinence from heroin during one year without any adverse reactions. In a recently published study 59 detoxified inpatients with heroin dependence received a ketamine-assisted psychotherapy (KPT) session prior to their discharge from an addiction treatment hospital, and were then randomized into two treatment groups.

Participants in the first group received two addiction counseling sessions followed by two KPT sessions, (with a single im injection of 2 mg/kg ketamine) with sessions scheduled on a monthly interval (multiple KPT group). Participants in the second group received two addiction counseling sessions on a monthly interval, but no additional ketamine therapy sessions (single KPT group). At one-year follow-up, survival analysis demonstrated a significantly higher rate of abstinence in the multiple KPT group. Thirteen out of 26 subjects (50%) in the multiple KPT group remained abstinent, compared to 6 out of 27 subjects (22.2%) in the single KPT group (p < 0.05). No differences between groups were found in depression, anxiety, craving for heroin, or their understanding of the meaning of their lives. It was concluded that three sessions of ketamine-assisted psychotherapy are more effective than a single session for the treatment of heroin addiction.[47][48]

Krupitsky and Kolp summarized their work to date in 2007.[49]

Jovaisa et al. from Lithuania demonstrated attenuation of opiate withdrawal symptoms with ketamine. A total of 58 opiate-dependent patients were enrolled in a randomized, placebo-controlled, double-blind study. Patients underwent rapid opiate antagonist induction under general anesthesia. Prior to opiate antagonist induction patients were given either placebo (normal saline) or subanesthetic ketamine infusion of 0.5 mg/kg·h. Ketamine group presented better control of withdrawal symptoms, which lasted beyond ketamine infusion itself. Significant differences between ketamine and Control groups were noted in anesthetic and early postanesthetic phases. There were no differences in effects on outcome after 4 months.[26]

Treatment of complex regional pain syndrome (reflex sympathetic dystrophy)

Ketamine is being used as an experimental and controversial treatment for Complex Regional Pain Syndrome (CRPS) also known as Reflex Sympathetic Dystrophy (RSD). CRPS/RSD is a severe chronic pain condition characterized by sensory, autonomic, motor and dystrophic signs and symptoms. The pain in CRPS is continuous, it worsens over time, and it is usually disproportionate to the severity and duration of the inciting event. The hypothesis is that ketamine manipulates NMDA receptors which might reboot aberrant brain activity. There are two treatment modalities, the first consist of a low dose ketamine infusion of between 25–90 mg per day, over five days either in hospital or as an outpatient. This is called the awake technique. Open label, prospective, pain journal evaluation of a 10-day infusion of intravenous ketamine (awake technique) in the CRPS patient concluded that "A four-hour ketamine infusion escalated from 40–80 mg over a 10-day period can result in a significant reduction of pain with increased mobility and a tendency to decreased autonomic dysregulation".[50]

Case notes of 33 patients whose CRPS pain was treated by the inpatient administration of a continuous subanesthetic intravenous infusion of ketamine were reviewed at Mackay Base Hospital, Queensland, Australia. A total of 33 patients with diagnoses of CRPS who had undergone ketamine treatment at least once were identified. Due to relapse, 12 of 33 patients received a second course of therapy, and two of 33 patients received a third. There was complete pain relief in 25 (76%), partial relief in six (18%), and no relief in two (6%) patients.

The degree of relief obtained following repeat therapy (N=12) appeared even better, as all 12 patients who received second courses of treatment experienced complete relief of their CRPS pain. The duration of relief was also impressive, as was the difference between the duration of relief obtained after the first and after the second courses of therapy. In this respect, following the first course of therapy, 54% of 33 individuals remained pain free for 3 months or more and 31% remained pain free for 6 months or more. After the second infusion, 58% of 12 patients experienced relief for a year or more, while almost 33% remained pain free for over 3 years. The most frequent side effect observed in patients receiving this treatment was a feeling of inebriation. Hallucinations occurred in six patients. Less frequent side effects also included complaints of light-headedness, dizziness, and nausea. In four patients, an alteration in hepatic enzyme profile was noted; the infusion was terminated and the abnormality resolved thereafter. No long-term side-effects were noted.[50]

The second treatment modality consists of putting the patient into a medically-induced coma and given an extremely high dosage of ketamine; typically between 600–900 mg.[51] This version, currently not allowed in the United States, is most commonly done in Germany but some treatments are now also taking place in Monterrey, Mexico. According to Dr Schwartzman, 14 cases out of 41 patients in the coma induced ketamine experiments were completely cured. "We haven't cured the original injury", he says, "but we have cured the RSD or kept it in remission. The RSD pain is gone." He added that "No one ever cured it before... In 40 years, I have never seen anything like it. These are people who were disabled and in horrible pain. Most were completely incapacitated. They go back to work, back to school, and are doing everything they used to do. Most are on no medications at all. I have taken morphine pumps out of people. You turn off the pain and reset the whole system."[51]

In Tuebingen, Germany Dr Kiefer treated a patient presented with a rapidly progressing contiguous spread of CRPS from a severe ligamentous wrist injury. Standard pharmacological and interventional therapy successively failed to halt the spread of CRPS from the wrist to the entire right arm. Her pain was unmanageable with all standard therapy. As a last treatment option, the patient was transferred to the intensive care unit and treated on a compassionate care basis with anesthetic doses of ketamine in gradually increasing (3–5 mg/kg·h) doses in conjunction with midazolam over a period of 5 days. On the second day, edema, and discoloration began to resolve and increased spontaneous movement was noted. On day 6, symptoms completely resolved and infusions were tapered. The patient emerged from anesthesia completely free of pain and associated CRPS signs and symptoms. The patient has maintained this complete remission from CRPS for 8 years now. The psychiatric side effects of ketamine were successfully managed with the concomitant use of midazolam and resolved within 1 month of treatment.[52]

Postoperative pain

The dissociative anesthetic effects of ketamine have also been applied within the realm of postoperative pain management. Low doses of ketamine have been found to significantly reduce morphine consumption as well as reports of nausea following abdominal surgery.[53]

Contents of various brands of ketamine

Ketanest S Parke Davis (Pfizer) - S enantiomer only, no preservatives[54]

Ketaset (Wyeth) - R/S ketamine and benzethonium chloride as a preservative.[55]

Ketanest - S ketamine only

Ketalar - R/S ketamine + Phermerol (benzethonium chloride) added as a preservative

Neuropharmacology

Ketamine was long thought to act primarily by inhibiting NMDA receptors.[56][57] But another NMDA receptor antagonist, MK-801, does not exert the same hypnotic effects.[58] It appears more likely that the hypnotic effects of ketamine are produced by inhibiting hyperpolarization-activated cyclic nucleotide-modulated (HCN1) cation channels, which mediate the "sag" current (Ih ) in neurons.[59] Inhibition of Ih by ketamine in cultured neurons causes a hyperpolarizing shift in resting membrane potential and enhances summation of excitatory currents.[60] Such effects, if induced in vivo, would likely induce cortical oscillations reminiscent of sleep.[61] Most importantly, knockout of HCN1 channels in mice eliminates the hypnotic actions of ketamine.[60]

Ketamine is an uncompetitive NMDA receptor antagonist.[62] This receptor opens in response to binding of the neurotransmitter glutamate, and blockade of this receptors is believed to mediate the analgesic (reduction of pain) effects of ketamine at low doses.[57] Evidence for this is reinforced by the fact that naloxone, an opioid antagonist, does not reverse the analgesia. Studies also seem to indicate that ketamine is "use dependent" meaning it only initiates its blocking action once a glutamate binds to the NMDA receptor.[citation needed]

At high, fully anesthetic level doses, ketamine has also been found to bind to opioid mu receptors and sigma receptors.[citation needed] Thus, loss of consciousness that occurs at high doses may be partially due to binding at the opioid mu and sigma receptors.[citation needed] It has also been shown to act as a potent D2 receptor partial agonist,[63] as well as a dopamine reuptake inhibitor.[citation needed]

(rac)-ketamine is a noncompetitive inhibitor of the α7 nAChR at clinically relevant concentrations. The preservative benzethonium chloride competitively inhibits α7 and α4β2 nAChRs at concentrations present in the clinical formulation of Ketalar.[64]

Ketamine is racemic, and its R and S stereoisomers have different binding affinities: (S)-ketamine has about four times greater affinity for the PCP site of the NMDA receptor than does (R)-ketamine (in guinea pig brain).[citation needed] (S)-ketamine seems to induce drowsiness more strongly than the (R) enantiomer; it is probable that (R)-ketamine is the stronger sigma agonist and so this enantiomer is likely to be responsible for the lowering of the seizure threshold that can occur with ketamine.[citation needed] Since (S)-ketamine has greater analgesic effects and less hallucinogenic side effects than (R)-ketamine, the pure (S) enantiomer is sometimes preferred to the racemic mix for use in medical procedures, especially when lower doses are used for minor surgical procedures where the patient remains conscious during the operation.[65]

The effects seem to take place mainly in the hippocampal formation and in the prefrontal cortex. This evidence, along with the NMDA receptor's connection with the memory formation process, explains ketamine's profound effects on memory and thought. These effects inhibit the filtering function of the brain and may mirror the sensory overload associated with schizophrenia and near death experiences.[citation needed]

The local anesthetic effects are likely from the blocking action of ketamine on sodium channels.[66] Its in vitro blocking potency of sodium channels in the resting state is similar to that of lidocaine.[67]

Ketamine has a well-documented neuroprotective effect against ischemic brain-injury and glutamate induced brain injury.[68] One hypothesis of its working mechanism in case of chronic pain management and depression is that it works as an antidote to an overactivity in glutamergic brain circuits.

Recreational use

Illicit sale

File:Ketamine Crystals.jpg
10 ml bottle of liquid ketamine drying into crystals
File:Ketamine.JPG
500 mg ketamine hydrochloride
File:Ketaminehydrochloride.jpg
Ketamine dried and scraped

Ketamine sold illicitly comes either from diverted legitimate supplies and semi-legitimate suppliers, or from theft from legitimate suppliers. It is imported in varying amounts from countries in which it is less strictly regulated.

In 2003, the US Drug Enforcement Agency conducted Operation TKO, a probe into the quality of ketamine being imported from Mexico.[69] As a result of operation TKO, US and Mexican authorities shut down the Mexico City company Laboratorios Ttokkyo, which was the biggest producer of ketamine in Mexico. According to the DEA, over 80% of ketamine seized in the US is of Mexican origin.[69] The World Health Organization Expert Committee on Drug Dependence, in its thirty-third report (2003),[70] recommended research into its recreational use/misuse due to growing concerns about its rising popularity in Europe, Asia and North America. This is due in part to its prevention of depression.[71]

In the 1993 book E for Ecstasy[72] (about the uses of the street drug Ecstasy in the UK) the writer, activist and Ecstasy advocate Nicholas Saunders highlighted test results showing that certain consignments of the drug also contained ketamine. Consignments of Ecstasy known as "Strawberry" contained what Saunders described as a "potentially dangerous combination of ketamine, ephedrine and selegiline", as did a consignment of "Sitting Duck" Ecstasy tablets.[73]

The former chairman of the British Advisory Council on the Misuse of Drugs, David Nutt, suggested that Ketamine should be upgraded from a class C drug due to the harm it can cause to users.[74]

Methods of use

The main form in which Ketamine is sold is in powder form, to be insufflated (inhaled).

Ketamine is sold in either powdered or liquid form. In its powdered form it can be insufflated, injected, or taken orally. It is also possible to smoke the drug in a joint or pipe, usually mixed with marijuana and tobacco. The smoke has a distinctive bitter taste but the effects of the high hit much faster than when insufflated, ingested or injected intramuscularly. Ketamine is typically injected into the leg, the onset for IM is about one minute. Heavy ketamine users solely use IM as their primary method of administration due to the bypassing of the liver, its increased efficiency, and smoother high. Oral use usually requires more material, but results in a longer trip. However, when administered orally, ketamine is rapidly metabolised to norketamine, which possesses sedating effects; this route of administration is unlikely to produce a dissociative state characteristic of ketamine unless very high doses (500 mg+) are ingested.[75]

Detection of use

Ketamine may be quantitated in blood or plasma to confirm a diagnosis of poisoning in hospitalized patients, provide evidence in an impaired driving arrest or to assist in a medicolegal death investigation. Blood or plasma ketamine concentrations are usually in a range of 0.5-5.0 mg/L in persons receiving the drug therapeutically (during general anesthesia), 1–2 mg/L in those arrested for impaired driving and 3–20 mg/L in victims of acute fatal overdosage. Urine is often the preferred specimen for routine drug abuse monitoring purposes. The presence of norketamine, a pharmacologically-active metabolite, is useful for confirmation of ketamine ingestion.[76][77][78]

Psychological effects

Ketamine produces effects similar to phencyclidine (PCP) and dextromethorphan (DXM). Unlike the other well known dissociatives PCP and DXM, ketamine is very short acting, its hallucinatory effects lasting sixty minutes when insufflated or injected and up to two hours when ingested, the total experience lasting no more than a couple of hours.[79] Like other dissociative anaesthetics, hallucinations caused by ketamine are fundamentally different from those caused by tryptamines and phenethylamines. At low doses, hallucinations are only seen when one is in a dark room with one's eyes closed, while at medium to high doses the effects are far more intense and obvious.[80]

Ketamine produces a dissociative state, characterised by a sense of detachment from one's physical body and the external world which is known as depersonalization and derealization.[81] At sufficiently high doses (e.g. 150 mg intramuscular), users may experience what is coined the "K-hole", a state of dissociation whose effects are thought to mimic the phenomenology of schizophrenia.[82] Users may experience worlds or dimensions that are ineffable, all the while being completely unaware of their individual identities or the external world. Users have reported intense hallucinations including visual hallucinations, perceptions of falling, fast and gradual movement and flying, 'seeing God', feeling connected to other users, objects and the cosmos, experiencing psychotic reactions, and shared hallucinations, and thoughts with adjacent users. John C. Lilly,[83] Marcia Moore[84] and D. M. Turner[85] (among others) have written extensively about their own spiritual/psychonautic use of ketamine. (Both Moore[86] and Turner[87] died prematurely in a way that has been linked to their ketamine use.)

Users may feel as though their perceptions are located so deep inside the mind that the real world seems distant (hence the use of a "hole" to describe the experience). Some users may not remember this part of the experience after regaining consciousness, in the same way that a person may forget a dream. Owing to the role of the NMDA receptor in long-term potentiation, this may be due to disturbances in memory formation. The "re-integration" process is slow, and the user gradually becomes aware of surroundings. At first, users may not remember their own names, or even know that they are human, or what that means. Movement is extremely difficult, and a user may not be aware that he or she has a body at all.

Long-term side effects

Neurological Effects

Chronic use of ketamine may lead to cognitive impairments including memory problems.[88] In 1989, psychiatry professor John Olney reported that ketamine caused reversible changes in two small areas of the rat brain. 40 mg/kg resulted in fluid-filled cavities ("vacuoles") appearing inside cells. The cavities disappeared after several days, unless high doses of the far more toxic PCP or close relative MK801 were repeatedly given, in which case some cell death was seen. Roland Auer injected monkeys with MK801 and was unable to produce any vacuoles. When Auer was asked in 1998 whether persons undergoing anesthesia with Ketalar were at risk of these changes, his reply was that he doubted that it was even a remote possibility because of fundamental differences in metabolism between the rat and human brain. Ketamine can block excitotoxicity (brain damage due to low oxygen, low sugar, epilepsy, trauma, etc.) but it can also excite the brain at low doses by switching off the inhibitory system. Why this is not damaging in monkeys and humans probably lies in the fact that ketamine binds to an increasingly wide range of different receptors as the dose level rises, and some of these receptors act to shut down the excitement. In humans, by the time a potentially toxic dose is reached, the "excitement window" has been passed and the drug is starting to activate other systems that switch cells off again, a result of ketamine's promiscuity that improves its safety relative to MK801. MK801 binds very specifically to N-P receptors. The other part of the explanation is that rats have rates of brain metabolism that are almost twice as high as those in humans to start with. It is because of this higher base rate of metabolism that ketamine causes over-excitement in rats at doses below those at which it activates shutdown systems.[89][90][91]

The first large-scale, longitudinal study of ketamine users found that heavy ketamine users had impaired memory by several measures, including verbal, short-term memory and visual memory. However occasional (1-2 times per month) ketamine users and ex ketamine users were not found to differ from controls in memory, attention and psychological well-being tests. This suggests that occasional use of ketamine does not lead to prolonged harm and that any damage that might occur may be reversible when ketamine use is stopped.[92]

Vutskits et al. from Geneva showed that short-term exposure of cultures to ketamine at concentrations of ≥ 20 μg/mL leads to a significant loss of differentiated cells and that non-cell death-inducing concentrations of ketamine (10 μg/mL) can still initiate long-term alterations of dendritic arbor in differentiated neurons, including dendritic retraction and branching point elimination. They also demonstrated that chronic (>24 h) administration of ketamine at concentrations as low as 0.01 μg/mL can interfere with the maintenance of dendritic arbor architecture. These results raise the possibility that chronic exposure to low, subanesthetic concentrations of ketamine, while not affecting cell survival, could still impair neuronal morphology and thus might lead to dysfunctions of neural networks.[93]

There is a long list of medicines that could counteract these potential toxic effects, including clonidine, anticholinergics, benzodiazepines, barbiturates and risperidone.[90][91][94]

Urinary Tract Effects

A study in Bristol reported in the British Medical Journal on 3 May 2008 linked urinary tract disease with ketamine use. Symptoms reported by users include an increased need to urinate, passing blood in urine, leakage of urine and pain on urination. These symptoms are associated with the scarification of the bladder lining, which leads to a shrunken bladder, erythema, and contact bleeding, and can then move to the ureters and damage the kidneys.[95]

In a study of 9 daily ketamine users, Shahani et al. found "marked thickening of the bladder wall, a small capacity, and perivesicular stranding, consistent with severe inflammation. At cystoscopy, all patients had severe ulcerative cystitis. Biopsies in 4 patients revealed epithelial denudation and inflammation with a mild eosinophilic infiltrate. Cessation of ketamine use, with the addition of pentosan polysulfate, appeared to provide some symptomatic relief."[96]

The bladder symptoms are so severe amongst some recreational users that their bladders have had to be removed. In these cases neobladders have been created using sections of gut but there is a significant risk of similar damage occurring to the neobladder if ketamine use is not stopped.

Many long term users report "K-pains" or "ketamine cramps".[97] The exact cause of these pains are unknown. The Ketamine induced abdominal pain is primarily limited to users of a gram or more of ketamine a day (route of administration does not seem to affect this symptom). It has been suggested that the amino acid Tyrosine may help alleviate the pain.[98]

In October 2009, Professor David Nutt, formerly of the British Home Office's Advisory Council on the Misuse of Drugs, recommended that ketamine be re-classed in the UK, as several patients who have used the drug recreationally had experienced bladder spasms and some have "irreversible bladder damage". Professor Nutt also reported that a small number of patients have had their bladder completely removed through ketamine abuse. The recommendation and case studies were featured heavily in the British press.[99]

See also

References

Cite error: Invalid <references> tag; parameter "group" is allowed only.

Use <references />, or <references group="..." />

zh-min-nan:Ketamine ca:Ketamina cs:Ketamin da:Ketamin de:Ketamin et:Ketamiin es:Ketamina eu:Ketamina fr:Kétamine it:Ketamina he:קטמין hu:Ketamin nl:Ketamine ja:ケタミン no:Ketamin pl:Ketamina pt:Ketamina ru:Кетамин simple:Ketamine sk:Ketamín sr:Кетамин fi:Ketamiini sv:Ketamin th:คีตามีน uk:Кетамін ur:Ketamine

zh:氯胺酮
  1. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  2. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  3. Hirota K, Sikand KS, Lambert DG (1999). "Interaction of ketamine with mu2 opioid receptors in SH-SY5Y human neuroblastoma cells". Journal of Anesthesia. 13 (2): 107–9. doi:10.1007/s005400050035. PMID 14530949. 
  4. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  5. Bergman SA (1999). "Ketamine: review of its pharmacology and its use in pediatric anesthesia". Anesthesia Progress. 46 (1): 10–20. PMC 2148883Freely accessible. PMID 10551055. 
  6. Peck TE, Hill SA, Williams M (2008). Pharmacology for anaesthesia and intensive care (3rd edition). Cambridge: Cambridge university press. p. 111. ISBN 9780521704632. 
  7. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  8. Krüger AD (1998). "[Current aspects of using ketamine in childhood]". Anaesthesiologie Und Reanimation (in German). 23 (3): 64–71. PMID 9707751. 
  9. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  10. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  11. Alltounian, Howard Sunny; Marcia Moore (1978). Journeys into the bright world. Rockport, Mass: Para Research. ISBN 0-914918-12-5. [page needed]
  12. "Ketamine - Schedule III of The Controlled Substances Act (CSA)". Anestesiología Mexicana en Internet. Retrieved 2006-12-22. 
  13. "Club 'horse' drug to be outlawed". BBC News. 28 December 2005. Retrieved 7 May 2010. 
  14. Controlled Drugs and Substances Act
  15. Government to tighten control on Ketamine
  16. Jansen, Karl (2001). Ketamine: Dreams and Realities. Multidisciplinary Association for Psychedelic Studies. p. 23. ISBN 0966001931. 
  17. Jansen, Karl (2001). Ketamine: Dreams and Realities. Multidisciplinary Association for Psychedelic Studies. p. 38. ISBN 0966001931. 
  18. Jansen, Karl (2001). Ketamine: Dreams and Realities. Multidisciplinary Association for Psychedelic Studies. p. 24. ISBN 0966001931. 
  19. 19.0 19.1 Jansen, Karl (2001). Ketamine: Dreams and Realities. Multidisciplinary Association for Psychedelic Studies. p. 26. ISBN 0966001931. 
  20. 20.0 20.1 Jansen, Karl (2001). Ketamine: Dreams and Realities. Multidisciplinary Association for Psychedelic Studies. p. 55. ISBN 0966001931. 
  21. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  22. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  23. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  24. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  25. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  26. 26.0 26.1 Jovaisa T, Laurinenas G, Vosylius S, Sipylaite J, Badaras R, Ivaskevicius J (2006). "Effects of ketamine on precipitated opiate withdrawal". Medicina. 42 (8): 625–34. PMID 16963828. 
  27. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  28. 28.0 28.1 28.2 Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  29. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  30. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  31. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  32. "Severe Pain Relief may be Possible with a Common Anesthetic Drug" (Press release). Blackwell Publishing. 1 October 2004. http://blackwellpublishing.com/press/pressitem.asp?ref=180. Retrieved 13 December 2009. 
  33. Fujikawa DG (1995). "Neuroprotective Effect of Ketamine Administered After Status Epilepticus Onset". Epilepsia. 36 (2): 186–195. doi:10.1111/j.1528-1157.1995.tb00979.x. PMID 7821277. 
  34. Correll GE, Futter GE (2006). "Two case studies of patients with major depressive disorder given low-dose (subanesthetic) ketamine infusions". Pain Medicine. 7 (1): 92–5. doi:10.1111/j.1526-4637.2006.00101.x. PMID 16533209. 
  35. NIH. "Experimental Medication Kicks Depression in Hours Instead of Weeks" NIH News, 7 August 2006
  36. Khamsi, R. "Ketamine relieves depression within hours" New Scientist, 8 August 2006.
  37. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  38. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  39. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  40. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  41. Faster-Acting Antidepressants Closer to Becoming a Reality", NIMH, 25 July 2007
  42. Krystal AD, Weiner RD, Dean MD; et al. (2003). "Comparison of seizure duration, ictal EEG, and cognitive effects of ketamine and methohexital anesthesia with ECT". The Journal of Neuropsychiatry and Clinical Neurosciences. 15 (1): 27–34. PMID 12556568. 
  43. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  44. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  45. The Combination of Psychedelic and Aversive Approaches in Alcoholism Treatment - Eleusis
  46. Krupitsky EM, Grinenko AY (1997). "Ketamine psychedelic therapy (KPT): a review of the results of ten years of research". Journal of Psychoactive Drugs. 29 (2): 165–83. PMID 9250944. 
  47. http://www.eleusis.us/resource-center/references/ketamine-psychotherapy-heroin.pdf
  48. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  49. Krupitsky; Kolp. "Ketamine Psychedelic Psychotherapy". In Michael J. Winkelman and Thomas B., Roberts. Psychedelic Medicine: New Evidence for Hallucinogens as Treatments. Westport, CT: Praeger/Greenwood. [page needed]
  50. 50.0 50.1 Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  51. 51.0 51.1 CNN report on ketamine therapy for CRPS/RSD 1 September 2006
  52. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  53. Lundberg GD (2008). "Postoperative Ketamine Can Reduce Morphine Consumption and Nausea". The Medscape Journal of Medicine. 10 (10): 234. PMC 2605142Freely accessible. 
  54. De Reuck J, Decoo D, Strijckmans K, Lemahieu I (1992). "Does the severity of leukoaraiosis contribute to senile dementia? A comparative computerized and positron emission tomographic study". European Neurology. 32 (4): 199–205. doi:10.1017/S0265021504003059. PMID 1505589. 
  55. Fort Dodge (division of Wyeth) Ketamine Lable. Article retrieved on 1 November 2008
  56. Radovanović D, Pjević M (2003). "[Ketamine: the past 30 years and its future]". Medicinski Pregled. 56 (9-10): 439–45. doi:10.2298/MPNS0310439R. PMID 14740534. 
  57. 57.0 57.1 Sinner B, Graf BM (2008). "Ketamine". Handbook of Experimental Pharmacology. 182 (182): 313–33. doi:10.1007/978-3-540-74806-9_15. PMID 18175098. 
  58. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  59. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  60. 60.0 60.1 Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  61. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  62. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  63. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  64. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  65. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  66. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  67. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  68. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  69. 69.0 69.1 "SI.com - The Mexican Connection - Jul 18, 2007". CNN. Retrieved 7 May 2010. 
  70. Untitled-59
  71. ABC News: Ketamine, a Tranquilizer and Popular Club Drug, May Work for Depression
  72. Saunders N, Heron L (1993). E for Ecstasy. London: N. Saunders. ISBN 0950162884. [page needed]
  73. See: [1] for details online.
  74. Doward, Jamie (1 November 2009). "Ministers face rebellion over drug tsar's sacking | Politics | The Observer". London: Guardian. Retrieved 2010-01-22. 
  75. Erowid Ketamine Vault: Dosage
  76. Feng N, Vollenweider FX, Minder EI, Rentsch K, Grampp T, Vonderschmitt DJ. Development of a gas chromatography-mass spectrometry method for determination of ketamine in plasma and its application to human samples. Ther. Drug Monit. 17: 95-100, 1995.
  77. Parkin MC, Turfus SC, Smith NW, Halket JM, Braithwaite RA, Elliott SP, Osselton MD, Cowan DA, Kicman AT. Detection of ketamine and its metabolites in urine by ultra high pressure liquid chromatography-tandem mass spectrometry. J. Chrom. B 876: 137-142, 2008.
  78. R. Baselt, Disposition of Toxic Drugs and Chemicals in Man, 8th edition, Biomedical Publications, Foster City, CA, 2008, pp. 806-808.
  79. Giannini AJ, Loiselle RH, Giannini MC, Price WA (1985). "Phencyclidine and the dissociatives". Psychiatric Medicine. 3 (3): 197–217. PMID 2893430. 
  80. AJ Giannini,RH Loisellle, MC Giannini, WA Price, 1987, op. cit. The dissociatives. Medical Psychiatry. 3(3):197-205, 1987[verification needed]
  81. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  82. Giannini AJ (1999). Drug Abuse. Los Angeles: Health Information Press. p. 104. ISBN 1-885987-11-0. [page needed]
  83. Lilly, John Cunningham (1997). The scientist: a metaphysical autobiography. Berkeley, CA: Ronin Pub. ISBN 0-914171-72-0. [page needed]
  84. Alltounian, Howard Sunny; Marcia Moore (1978). Journeys into the bright world. Rockport, Mass: Para Research. ISBN 978-0-914918-12-7. [page needed]
  85. Turner, D. M. (1994). The essential psychedelic guide. San Francisco, CA: Panther Press. ISBN 0-9642636-1-0. [page needed]
  86. Jansen, Karl (2001). Ketamine: Dreams and Realities. Multidisciplinary Association for Psychedelic Studies. p. 50. ISBN 0966001931. 
  87. Jansen, Karl (2001). Ketamine: Dreams and Realities. Multidisciplinary Association for Psychedelic Studies. p. 89. ISBN 0966001931. 
  88. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  89. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  90. 90.0 90.1 Erowid DXM Vaults: Health: The Bad News Isn't In: A Look at Dissociative-Induced Brain Damage, by Anderson C
  91. 91.0 91.1 Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  92. Morgan, Celia J. A.; Muetzelfeldt, Leslie; Curran, H. Valerie (2009). "Consequences of chronic ketamine self-administration upon neurocognitive function and psychological wellbeing: a 1-year longitudinal study". Addiction. 105 (1): 121. doi:10.1111/j.1360-0443.2009.02761.x. PMID 19919593. 
  93. Hargreaves RJ, Hill RG, Iversen LL (1994). "Neuroprotective NMDA antagonists: the controversy over their potential for adverse effects on cortical neuronal morphology". Acta Neurochirurgica. Supplementum. 60: 15–9. PMID 7976530. 
  94. AJ Giannini, N Underwood, M Condon op. cit.[verification needed]
  95. Cottrell, A. M; Athreeres, R.; Weinstock, P.; Warren, K.; Gillatt, D. (2008). "Urinary tract disease associated with chronic ketamine use". BMJ. 336: 973. doi:10.1136/bmj.39562.711713.80. 
  96. Lua error in package.lua at line 80: module 'Module:Citation/CS1/Suggestions' not found.
  97. Middleton, Duncan (15 January 2009). "Warning over rising ketamine use". BBC. Retrieved 19 January 2010. 
  98. Jansen, Karl (2001). Ketamine: Dreams and Realities. Multidisciplinary Association for Psychedelic Studies. p. 271. ISBN 0966001931. 
  99. "Ketamine link to bladder failure". BBC News. 4 February 2009. Retrieved 15 January 2010.  |first1= missing |last1= in Authors list (help)