Skip to main content
Erschienen in: Perioperative Medicine 1/2019

Open Access 01.12.2019 | Review

Cannabis-based medicines and the perioperative physician

verfasst von: Patrick Tapley, Suzanne Kellett

Erschienen in: Perioperative Medicine | Ausgabe 1/2019

Abstract

The increasing availability of cannabis for both recreational and medicinal purposes means that anaesthetists will encounter an increasing number of patients taking cannabis-based medications. The existing evidence base is conflicted and incomplete regarding the indications, interactions and long-term effects of these substances. Globally, most doctors have had little education regarding the pharmacology of cannabis-based medicines, despite the endocannabinoid system being one of the most widespread in the human body. Much is unknown, and much is to be decided, including clarifying definitions and nomenclature, and therapeutic indications and dosing. Anaesthetists, Intensivists, Pain and Perioperative physicians will want to contribute to this evidence base and attempt to harness such therapeutic benefits in terms of pain relief and opiate-avoidance, anti-emesis and seizure control. We present a summary of the pharmacology of cannabis-based medicines including anaesthetic interactions and implications, to assist colleagues encountering these medicines in clinical practice.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
BIS
Bispectral Index
CB
Cannabinoid
CBD
Cannabidiol
CBN
Cannabinol
CBM
Cannabis-based medicines
CYP
Cytochrome P450
D8THC
Delta-8-tetrahydrocannabinol
D9THC
Elta-9-tetrahydrocannabinol
EEG
Electroencephalogram
GABA
Gamma amino butyric acid
HPRA
Health Products Regulatory Authority
MA
Meta-analysis
MS
Multiple sclerosis
NASEM
National Academies of Science Engineering and Medicine
NNT
Number needed to treat
NNTH
Number needed to harm
PONV
Post-operative nausea and vomiting
RCT
Randomised controlled trial
SR
Systematic review
THC
Tetrahydrocannabinol
UK
United Kingdom
USA
United States of America
WHO
World Health Organisation

Introduction

Cannabis use for medicinal purposes was first documented in 2900 BC in China, when Emperor Shen Nong described benefit for rheumatism and malaria (Pertwee 2015) and later in Ancient Egyptian texts (Pertwee 2015; Zlas et al. 1993). In the United Kingdom (UK), Queen Victoria’s personal physician Sir John Russell Reynolds issued a tincture containing cannabis for her Majesty’s menstrual cramps (David 2017), subsequently publishing his 30 years’ worth of experience with the drug (Reynolds 1890).
Discussion in medical journals, the mainstream and social media around the use of cannabis for medicinal and non-medicinal purposes has increased recently, especially following the legalisation of cannabis for recreational use in Canada (Government of Canada 2018a) and the UK government’s decision to make cannabis-based medicines (CBMs) available for prescription by doctors on the specialist register (Department of Health and Social Care 2018).
The actual, social and economic legitimisation of cannabis and its medicinal derivatives makes it likely increasing numbers of patients will present on this class of medicines. Perioperative physicians will require a sound understanding of their pharmacology and evidence base, and may wish to exploit this group of compounds for therapeutic purposes in the perioperative period.
A search of Pubmed was conducted in February 2019 utilising the search terms cannab* and the AND function for the following search terms individually; anaes*, marijuana, pain, nausea, surgery and pharmaco*. Abstracts were then screened for their applicability, with full texts reviewed. This was supplemented by a review of recent publications from governmental and regulatory organisations relating to CBMs, with backward reference searching. A search of individual governmental websites looking for legislation around cannabis and cannabinoid use was also undertaken in September 2019.
The pharmacology of novel psychoactive compounds (“legal highs”) is outside the scope of this review.

The global position

The availability of CBMs varies geographically and there is no global consensus on how cannabis and CBMs should be regulated. International stakeholders and regulators, including the United Nations (International Drug Policy Consortium 2016; Transnational Institute and Global Drug Policy Observatory 2016) and World Health Organisation (WHO) have been inconsistent in their approach. The WHO’s Expert Committee on Drug Dependences’ recent review (Ghebreyesus 2019) recommended to the United Nations Office on Drugs and Crime (UNODC) that the rescheduling within the International Drug Control Conventions occurs for cannabis and cannabis resin, dronabinol, tetrahydrocannabinol and extracts and tinctures of cannabis. They also repeated their recommendation to remove cannabidiol (CBD) preparations (with not more than 0.2% delta-9-THC) from the International Drug Control Conventions. The UNODC subsequently delayed its vote on these recommendations, but despite this, many countries are proceeding to legalise or reschedule cannabis and/or CBMs, broadening public availability, with the UK the most recent country to reschedule CBMs. Table 1 details the current status of cannabis and CBMs in selected countries for medical use. The recreational use of cannabis is currently legalised in Uruguay, Canada and certain states within the USA (United Nations Office on Drugs and Crime 2019).
Table 1
Current legal status of Cannabis and CBMs in selected countries
Continent
Country
Year approved
Notes
North America
America (United Nations Office on Drugs and Crime 2019; Mead 2017; US Food and Drug Administration 2016; Aguilar et al. 2018)
1996-2017
Cannabis illegal under federal law, individual states have legalised to varying degrees
Medical use:
-Legalised by 33 states
-FDA has approved dronabinol (Marinol ®, Syndros® ), nabilone (Cesamet ®,) and cannabidiol (Epidiolex ®) (CBMs) for specific indications, but cannabis plant not FDA approved
Recreational use:
-Legalised by 10 states (Alaska, California, Colorado, Maine, Massachusetts, Michigan, Nevada, Oregon, Vermont, Washington) and District of Colombia for recreational use
-Illinois to be legalised January 1 2020
-Multiple states have bills in session to legalise recreational and medicinal cannabis and its products
-“Decriminalised” in numerous states (ie avoid jail term)
Canada (United Nations Office on Drugs and Crime 2019; Aguilar et al. 2018; Government of Canada 2018b; Health Canada 2018)
1999/2018
Medical use:
-Regulated by federal government
-Healthcare practitioner authorisation required
-Patients can access cannabis through federally licensed seller, designate someone to produce, or grow their own.
Recreational use:
-Cannabis for recreation legalised October 2018 under Cannabis Act
-Cannabis act permits sale of cannabis oil, fresh cannabis, dried cannabis, cannabis plants and seeds
-Retailers must be authorised to sell cannabis
Mexico (Aguilar et al. 2018; Government of Mexico 2018a; Government of Mexico 2018b; Government of Mexico 2018c)
2017
Medical use:
Cannabis use allowed for medical and scientific purposes
Application required for import and use of cannabis and CBMs for medical purposes
Products with < 1% THC recently marketed under Government authorisation with no prescription required
South America
Uruguay (United Nations Office on Drugs and Crime 2019; Aguilar et al. 2018; Uruguay Government 2018)
2013
Medical use:
-Prescription required for CBM for medicinal use
-Medicinal cannabis not authorised to be sold
-CMs accessible via specialist prescription
Recreational Use:
-Cannabis legalised and market regulated from December 2013 with strict controls
Chile (Aguilar et al. 2018)
2015
Medical use:
-Medical prescription required for medicinal cannabis
-CBM can be imported under special license
Brazil (Aguilar et al. 2018; Brazilian Government 2016)
2014
Medical use:
-Imports of medications based on CBD oil for medical and therapeutic use for patients with prescription allowed
-Nabiximols (Sativex ®) licensed for MS
-Cannabis plant prescription prohibited
Argentina (Aguilar et al. 2018; Argentinian government 2018; Government of Argentina 2019)
2017
Medical use:
-Prescription and government authorisation required through ministry of health
-State produced pharmaceuticals for domestic market
-Cannabis oils legal for import
Oceania
Australia (Aguilar et al. 2018; Australian Government Department of Health 2018; Therapeutic Goods Administration 2017)
2016
Medical use:
-cultivation, manufacture, prescribing and dispensing of medicinal cannabis products for patients now legal
-Access via authorised prescriber program, or special access scheme on individual patient basis. Must be prescribed
-Cannabis, THC, nabiximols (Sativex ®), dronabinol (Marinol ®), nabilone (Cesamet ®, Canemes ®) listed under schedule 8, require prescription
-Cannabidiol listed under schedule 4, requires prescription
New Zealand (Aguilar et al. 2018; New Zealand Parliament 2018; New Zealand Ministry of Health 2018)
1977
Medical use:
-Prescription required for most cannabis-based products, some require ministerial approval
-Nabiximols (Sativex ®)and CBD based products available without ministerial approval
-Bill currently passing through NZ parliament to improve access
Europe
Netherlands (Aguilar et al. 2018; Office of Medicinal Cannabis 2018; Government of the Netherlands 2019; Office of Medicinal Cannabis Government of the Netherlands 2019)
2000
Medical use:
-Medicinal use legalised in 2000
-Office of Medicinal Cannabis government agency responsible for supplying medicinal cannabis to pharmacies, prescription required
-5 compositions produced, with varying strengths of THC and CBD
Recreational use:
-Illegal, but smoking cannabis under strict conditions allowed
Germany (Aguilar et al. 2018; Broich 2018)
2017
Medical use:
-Medicinal cannabis products able to be prescribed by physician
-CBMs included in range of medications covered by public and private health insurance
Poland (Aguilar et al. 2018)
2017
Medical use:
-Medicinal cannabis sold in registered pharmacies.
-Patients need permission from pharmaceutical inspector and physician
France (Aguilar et al. 2018; National Agency for the Safety of Medicines and Health Products 2013)
2013
Medical use:
-Law changed to allow marketing authorisation of products containing cannabis or its derivatives
-Dronabinol (Marinol ®) approved and marketed
-Nabiximols (Sativex ®) approved but not marketed due to price disagreements
Ireland (Aguilar et al. 2018; Department of Health Ireland 2018; Health Products Regualtory Authority 2017)
 
Medical use:
-Cannabis for medical use access programme enables prescribing of Cannabis for medical use by Medical Consultant
-Authorised CBMs (nabiximols (Sativex ®) , dronabinol (Marinol ®), nabilone (Cesamet ®, Canemes ®) should be used in first instance with prescription
-Cannabis (plant or extracts not authorised as medicine) considered as treatment option
-Cannabidiol (Epidiolex ®) not currently authorised
 
UK (National Institute for Health and Care Excellence 2019; Dame Sally Davies 2018; National Health Service 2019)
2018
Medical use:
-CBMs recently rescheduled, only available via prescription from doctor on the specialist register
-Nabilone and Nabiximols (Sativex ®) currently licensed but the latter is not approved by NICE
-Dronabinol not available as a licensed medicine
-Cannabidiol not classed as CBM, and not controlled. Epidiolex® currently in licensing process.
-NICE draft guidance for prescription of CBMs currently open for public consultation
Middle East
Israel (Aguilar et al. 2018)
1992
Medical use:
-Medical Cannabis Unit established for the regulation of Cannabis
-Specialist physicians apply for Medical Cannabis permit on behalf of patient
- > 40,000 patients receiving medicinal cannabis
Asia
Japan (Aguilar et al. 2018)
 
Prohibited
Pakistan (Aguilar et al. 2018)
 
Prohibited
Philippines (Aguilar et al. 2018; Republic of Phillipines House of Representatives 2018)
2016
Medical use:
-Cannabis allowed with prior authorisation from a doctor and treatment delivered in dedicated centres
-Covers all forms of Cannabis, no specific mention CBMs
India (Aguilar et al. 2018; Indian Council of Medical Research 2018)
 
Medical use:
-Legal provisions for medicinal use yet to be implemented
-Council of Scientific and industrial research currently undertaking research into medical benefits
Africa
South Africa (Department of Agriculture, Forestry and Fisheries 2018; Constitutional Court of South Africa 2017; Medicines Control Council 2016)
2018
Medical use:
-Special authorisation from Medical Control Council by physician can be requested for prescription of medicinal cannabis products
-Cannabidiol preparations excluded from Medicines and related substances act

Definitions of cannabis and cannabinoids

Cannabis

The Cannabis genus encompasses three major species; Cannabis sativa, Cannabis indica and Cannabis ruderalis. The number of identifiable extractable compounds has increased dramatically from 60 (Ashton 1999) to over 500 in the last 20 years (Beaulieu et al. 2016), of which over 100 are cannabinoids (Bie et al. 2018).

Cannabinoids

Cannabinoids are endogenous in humans, animals and plants, or synthetically produced, acting as ligands at the cannabinoid receptors. Cannabinoids can be psychoactive, for example delta-9-tetrahydrocannabinol (d9THC), delta-8-tetrahydrocannabinol (d8THC), cannabinol (CBN) or non-psychoactive, for example cannabidiol (CBD). Table 2 lists their classification, as well as examples of currently available CBMs (Pertwee 2015; Beaulieu et al. 2016; Zendulka et al. 2016; Yeon Kong et al. 2018; Hauser et al. 2018a; Barnes 2018; National Institute for Health and Care Excellence 2014; Rice and Cameron 2017; Krcevski-Skvarc et al. 2018).
Table 2
Classification of cannabinoids and some commercially available preparation
Group
Where found
Major known Compounds
Substance found in
Trade name
Route of administration
Current uses
Endo
-cannibinoid
Endogenous within body
-Arachidonoyl ethanolamide (Anandamide)
-2-arachidonylglycerol (2-AG)
-Docosatetraenoyl-ethanolamide (DEA),
-N-arachidonyldopamine (NADA),
-Virodhamine,
-2-arachidonylglyceryl,
-noladin ether
-dihomo- gamma-linolenoylethanolamide (HEA).
    
Synthetic cannabinoids
Chemically synthesised
Synthetic THC
-(-) trans isomer of delta 9 THC
Dronabinol
Marinol®
Oral capsule (formulated in sesame oil)
US; FDA approved;
-anorexia associated weight loss in AIDS patients
-chemo induced nausea and vomiting *
UK; not available
Syndros®
Liquid formulation
US; FDA approved
- anorexia associated weight loss in AIDS patients
-chemo induced nausea and vomiting *
Synthetic THC analogue (single molecule)
Nabilone
Cesamet®
Oral capsule
US; FDA approved chemo induced nausea and vomiting
UK; -chemo induced nausea and vomiting *
Variety; mainly aminoalkylindole derivatives ie JWH-018, UR-144, PB-22
Legal highs
Spice, K2, eclipse
Oral Smoked
 
Phyto
-cannibinoid (plant derived)
Cannabis genus;
-cannabis sativa,
-cannbis indica
-cannabis ruderalis
THC content varies 1-22%
CBD content varies 0.05-9%
Cannabis; Marijuana
 
Inhalation (commonly smoked)
UK; Illegal
US; legalised for medical use in 29 states, recreational use in 9 states
THC content varies 1-22%
CBD content varies 0.05-9%
Cananbis; Hashish
 
Oral
UK; Illegal
US; legalised for medical use in 29 states, recreational use in 9 states
Pure CBD (no psychoactive properties)
Cannabidiol
 
Oral capsule
Oromucosal spray
 
Epidiolex
Oil
US;FDA approved for treatment resistant epilepsy
2.7mg THC and 2.5mg CBD per spray
Nabiximols
Sativex®
Oromucosal spray
US; not approved. Approved in 30 other coutnrie
UK; MHRA but not NICE approved. MS induced spasticity
2.5mg THC, 1.25mg CBD
Cannador
 
Oral capsule
German based research only
*not responded to conventional antiemetics

Pharmacology of cannabinoids

Mode of action

The endocannabinoid system consists of both cannabinoid (CB) receptors and neurotransmitters, the plasma concentrations of which are normally at low levels. They are synthesised in the postsynaptic neurone (Hosking and Zajicek 2008) in response to stimuli including pain, stress, inflammation and are involved in the homeostasis of various body systems (Pertwee 2015). Antinociceptive effects occur via their actions as retrograde transmitters at presynaptic inhibitory CB1 receptors (Hauser et al. 2018a). Both CB1 and CB2 receptors are G protein coupled receptors (Gi,Go) with stimulation reducing cAMP production through the inhibition of adenylyl cyclase, resulting in an action on voltage gated calcium and potassium channels depressing neuronal excitability and reducing neurotransmitter release (Zendulka et al. 2016; Hauser et al. 2018a; Hosking and Zajicek 2008).
CB1 receptors are found in the cortex (thalamus, medulla, periaqueductal gray matter, descending pain pathways), spinal cord (descending pain pathways, dorsal horn) and peripherally on primary afferent sensory neurones where they outnumber the mu receptor, suggesting a potential mechanism for the modulation and treatment of neuropathic pain (Kumar et al. 2001).
CB2 receptors are involved in immunomodulation, with receptors distributed in the spleen, macrophages and Kupffer cells. It is increasingly recognised that the endocannabinoid system plays a crucial role in the maintenance of microglial activity through actions at CB1 and CB2 receptors, reducing neuro-inflammation (Bie et al. 2018; Bilkei-Gorzo et al. 2018). Relatively few CB2 receptors are found in the nervous system (Lucas et al. 2018), but they are inducible in the dorsal horn following inflammation or injury, with increased receptor concentration found in neuropathic pain models and receptor activation limiting the acute inflammatory process contributing to nociceptor sensitisation (Bie et al. 2018; Hosking and Zajicek 2008).
Exogenous and endogenous cannabinoids have differing effects at the CB1 and CB2 receptors. THC is an agonist at both, whilst CBD is a non-competitive antagonist at CB1 receptors at high concentrations, an inverse agonist at CB2 receptors and causes allosteric modulation of both CB receptors (Pertwee 2015; Lucas et al. 2018; Expert Committee on Drug Dependence 2018). The cannabinoid compounds, particularly CBD, have additional actions within the nervous system through signalling at a multitude of other receptors. This includes adenosine, serotonergic, adrenergic, nicotinic acetylcholine, glycine, nuclear peroxisome proliferator activated receptors (PPARs) and transient receptor potential (TPRV) ion channels (Capsaicin target). Anaesthetists should also note their actions at the opioid, NMDA and gamma amino butyric acid (GABA) receptors (Zendulka et al. 2016; Hauser et al. 2018a; Expert Committee on Drug Dependence 2018; Meng et al. 2017; Koppel et al. 2014).

Opioid system interaction

The cannabinoid and opioid systems are closely linked, with the activation of both opioid and cannabinoid receptors mediating common intracellular signalling mechanisms (Manzanares et al. 1999; Abrams et al. 2011; Scavone et al. 2013; Cohen et al. 2019; Pertwee et al. 2010). Opioid and cannabinoid receptors are found within the same cells and neurones in the central nervous system, with cannabinoids acting at kappa and delta receptors to increase endogenous opioid synthesis and release. Notably, the administration of opioid antagonists has been shown to block some of the effects of delta 9THC (Manzanares et al. 1999). The presence of opioid and cannabinoid receptors in noradrenergic pathways may have a role in the treatment of opiate withdrawal (Scavone et al. 2013).

NMDA system interaction

The NMDA receptor NR1 subunit is closely coupled to CB1 receptors, with the histidine triad nucleotide binding protein 1 (HINT 1) thought to be the pivotal modulator, exerting a negative control on NMDA receptors. HINT-1 gene deletion results in loss of CB1 inhibition of the NMDA receptor (Rodríguez-Muñoz et al. 2016). CB1 receptors have both presynaptic (reduced release of glutamate into synaptic cleft) and post-synaptic (intracellular NMDA signalling) effects (Rodríguez-Muñoz et al. 2016).
NMDA receptor activity stimulates the release of endocannabinoids, resulting in negative feedback reducing NMDA receptor numbers. It has been postulated that exo-cannabinoids are more potent inhibitors of the NMDA receptor than endocannabinoids (Pacheco et al. 2019; Ferreira et al. 2018), with exo-cannabinoids causing neural dysfunction and NMDA receptor hypofunction through alteration in the balance of NMDA-CB receptor regulation (Rodríguez-Muñoz et al. 2016).
The endocannabinoid system also regulates NMDA receptor activity by preventing over activation, neuroprotection from excitotoxicity and downregulating their activity (Rodríguez-Muñoz et al. 2016; Pacheco et al. 2019; Sánchez-Blázquez et al. 2014).

Gamma amino butyric acid

Gamma amino butyric acid (GABA) and CB1 receptors are closely localised in multiple cortical regions, including the hypothalamus, hippocampus and cortex (Cohen et al. 2019; Lotsch et al. 2018). CB1 receptors are expressed on GABAergic neurons, helping to regulate astrocyte and microglial activity, and hence neuroinflammation (Bilkei-Gorzo et al. 2018).
In preclinical studies, cannabinoids inhibit GABA release, through activation of CB1 receptors (Pertwee 2015; Laaris et al. 2011). They inhibit GABA uptake from the CNS extracellular space (Laaris et al. 2011), and cause allosteric modulation of GABA receptors (Bakas et al. 2017). Limited human studies show altered levels of GABAergic functions with chronic cannabis use, which may contribute to psychological effects (Cohen et al. 2019).

Pharmacokinetics

Absorption

The absorption of vaporised cannabinoids is rapid, with peak plasma concentrations observed within 10 min. THC’s bioavailability after inhalation ranges from 10 to 35%, and CBD 31% varying with device used and size of the particles (Kumar et al. 2001; Lucas et al. 2018; Karschner et al. 2011).
Oral bioavailability of CBM is low, at 2–20% for both CBD and THC (Lucas et al. 2018; Karschner et al. 2011; Anderson and Chan 2016) mainly due to extensive first pass metabolism (Lucas et al. 2018). Onset of action is 0.5–2 h due to slow intestinal absorption resulting in a longer duration of action (Kumar et al. 2001; Bridgeman and Abazia 2017).
An oromucosal spray preparation (nabiximols; Sativex ®) has a reported bioavailability similar to oral THC or intermediate between the oral and inhaled routes (Lucas et al. 2018; Expert Committee on Drug Dependence 2018; Karschner et al. 2011; Anderson and Chan 2016).
Transdermal administration is reported, with the permeability of CBD and CBN higher than d9THC and d8THC (Therapeutic Goods Administration 2017), but their hydrophobic nature means transdermal absorption is poor and requires permeation enhancement (Lucas et al. 2018).

Distribution

Volume of distribution varies by cannabinoid studied, with a VD of 32 L/kg for CBD (intravenous administration), and 3.4 L/kg for THC (inhalation administration) (Lucas et al. 2018), which is said to follow a three-compartment model (Heuberger et al. 2015). Cannabinoids are highly lipid soluble (Kumar et al. 2001) with rapid penetration through the blood–brain barrier (Ashton 1999), the placenta and into breast milk (Lucas et al. 2018). This also leads to accumulation in fatty tissue, with continued activity following cessation.

Metabolism

The cannabinoids are mainly hydroxylated and glucuronidated in the liver by the cytochrome P450 family of isoenzymes (Kumar et al. 2001; Lucas et al. 2018; Karschner et al. 2011; Ujváry and Hanuš 2016). Some metabolites are equipotent to the parental compounds (Yeon Kong et al. 2018; Rong et al. 2017). THC is metabolised to over 80 metabolites by various isoenzymes, including CYP1A1, CYP1A2, CYP1B1, CYP2B6, CYP2A6, CYP2C9 and CYP3A4. Inhibition of CYP3A4 may result in clinically apparent interactions with oxycodone (Pertwee 2015; Zendulka et al. 2016; Hauser et al. 2018a; Lucas et al. 2018).
CBD is metabolised to over 100 metabolites by isoenzymes CYP1A1, CYP1A2, CYP3A4, CYP2C9 and CYP2D6, the most abundant metabolite being the hydroxylated 7-COOH CBD derivative (Lucas et al. 2018; Ujváry and Hanuš 2016). Inhibition of CYP2D6 and CYP3A4 results in interactions with oxycodone, benzodiazepines and haloperidol (Hauser et al. 2018a; Karschner et al. 2011; Ujváry and Hanuš 2016). Oral CBD increases clobazam (and active metabolite) plasma levels (CYP2C19 interaction) (Ujváry and Hanuš 2016), resulting in dose reductions of clobazam in recent randomised controlled trial (RCTs) (Thiele et al. 2018). Prolonged use of CBD results in CYP1A1 induction (Ujváry and Hanuš 2016).
Cannabinol is metabolised via CYP2C9 and CYP3A4, with no evidence cytochrome P450 interactions (Zendulka et al. 2016).
The significance of these interactions is uncertain as they have occurred either in vitro or in excess of clinically relevant concentrations.

Elimination/excretion

Clearance of cannabinoids is estimated to be 38.8 L/h to 53 L/h (Heuberger et al. 2015), with long terminal half-lives due to their lipophilicity. In regular users, this is extended, with measurable plasma concentrations of THC over 24 h after last administration. Fifteen percent of cannabinoid metabolites undergo enterohepatic recycling, prolonging their action (Ashton 1999).
THC and metabolites are mainly excreted in faeces (65–80%) and urine (20–35%) (Ashton 1999; WHO Expert Committee on Drug Dependence 2018). THC’s elimination half-life is 56 h in occasional and 28 h in chronic users (Ashton 1999) with urinary metabolites measurable 14 days post exposure (Vandrey et al. 2017). Nabilone’s (Cesamet ®; synthetic THC) elimination half-life is shorter than THC, at 2–4 h, yet 16% of a single dose is reportedly measurable at 7 days post administration (Ashton 1999).
CBD metabolites and unchanged drug are mainly excreted in the urine with an elimination half-life of 2–5 days (Lucas et al. 2018; Anderson and Chan 2016; Ujváry and Hanuš 2016).

Pharmacodynamics (of relevance to the perioperative physician)

Cardiovascular system

Tachycardia due to CB1 agonism in cardiac myocytes has been reported (Kumar et al. 2001; Lucas et al. 2018), but was not noted following intravenous administration of d9THC (Vandrey et al. 2017). Bradycardia, hypotension, an increased cardiac output and myocardial oxygen demand have been described (Dickerson 1980; Bryson and Frost 2011). These effects are potentially exacerbated by sympathomimetic agents although the mechanism of action is unclear (Lucas et al. 2018). Effects may be cannabinoid specific, with CBD (Expert Committee on Drug Dependence 2018) not reported to effect heart rate or blood pressure, and THC possibly having anticholinergic effects through depletion of acetylcholine stores (Dickerson 1980).

Central nervous system

Effects are well described largely in relation to their abuse as a recreational drug, including psychomotor impairment, sedation, dizziness, euphoria, disorientation and confusion. Effects may be enhanced if administered with other CNS depressant drugs, for example opioids or benzodiazepines, and have been observed in a clinical setting (Kumar et al. 2001; Lucas et al. 2018).
The behavioural and long-term psychological effects (including dependence) of cannabis are widely reported (Pertwee 2015; Kumar et al. 2001; Nugent et al. 2017), and not reiterated here. Some evidence suggests the abuse potential of CBMs, likelihood of withdrawal phenomena and mental health morbidity is low (Pertwee 2015; Aragona et al. 2009), but trials are of short duration and do not examine long term effects. Evidence suggests chronic cannabis use impairs learning, memory and attention, and causes complex mental health disorders (Pertwee 2015; Nugent et al. 2017; National Academies of Sciences Engineering and Medicine 2017; Campbell et al. 2018). Further research is needed to determine relevance to CBM use.

Respiratory system

There is no clear evidence of respiratory system effects when administered by routes other than smoking. This may be due to the absence of cannabinoid receptors in the brainstem (Kumar et al. 2001).
Perioperative practitioners should be alert to the recent warning from the FDA around the use of vaping THC oil (US Food and Drug Administration 2019). This followed on from a multitude of reports of severe pulmonary disease development associated with vaping of THC products (Layden et al. 2019). Any patient presenting in the perioperative period with new onset respiratory disease and a history of vaping THC should therefore be thoroughly evaluated with this kept in mind.

Immune system

Animal studies suggest that high-dose cannabinoids impair cell-mediated and humoral immunity (Kumar et al. 2001), and low-dose CBD causes immune stimulation (Expert Committee on Drug Dependence 2018). The clinical relevance in humans is unclear.

Interactions of note for the perioperative physician

Induction agents/volatiles

Effects of cannabinoids on dosing of volatile and intravenous anaesthetic agents is equivocal, with evidence limited to animal studies, case reports and two limited human studies.
Ether anaesthesia is prolonged in mice and rats by cannabidiol, d8THC and d9THC (Chesher et al. 1974). Halothane anaesthesia is prolonged and dose requirements reduced in dogs after THC administration (Stoelting et al. 1973), with similar effects noted in mice with isoflurane administration (Schuster et al. 2002). Little is known about the interaction between cannabinoids and modern inhalational anaesthetics.
Animal studies have shown cannabidiol, d8THC and d9THC prolong barbiturate anaesthesia in mice and rats (Chesher et al. 1974) and THC administration increases the doses of thiopentone and propofol required for sedation (Brand et al. 2008). A cannabis extract premedication in dogs resulted in quicker onset and longer lasting anaesthesia with propofol (Kumar et al. 2010). One postulated mechanism is the increased Andamide (endocannabinoid) levels in the brain with propofol, with the inhibition of the enzyme fatty acid amide hydrolyses (FAAH), which normally terminates Anandamides activity, thought to be key (Schelling et al. 2006).
There is limited evidence of the effect of cannabinoid exposure on anaesthesia in humans. Case reports suggest increased anaesthetic requirements with non-medicinal cannabis use (Richtig et al. 2015; Symons 2002). A prospective trial found significantly increased propofol dosing for induction and LMA insertion in cannabis users versus controls (Flisberg et al. 2009). Studies utilising bispectral index monitoring (BIS) found no differences between cannabis users and non-users with the bolus dose of propofol required to achieve a BIS of < 60 (Flisberg et al. 2009). Higher BIS values have been noted for patients under steady state volatile anaesthesia who were administered nabiximols (Sativex®) as a premedication versus controls (Ibera et al. 2018).
These results should be interpreted cautiously given the limited number of participants, the applicability of extrapolating animal studies to human practice, use of unknown quantities of non-prescribable CBMs (except one study) and uncertainties about prior cannabis consumption (Flisberg et al. 2009). Additionally in the electroencephalogram (EEG)/depth of anaesthesia studies, it is unclear if the effects are a result of cannabinoids on the EEG or the effect of cannabinoid-anaesthetic interaction.
In summary, there is minimal evidence base as to the effects of the current agents, with animal studies relating to older agents only (ether, halothane, isoflurane). The evidence for intravenous agents is conflicting and of poor quality, but propofol requirements may be higher. There is a current research opportunity for investigation into the interaction with newer agents in humans.

Opioids

Cannabinoid agonists facilitate endogenous opioid signalling and increase concentrations of endogenous opioids (Scavone et al. 2013; Abrams 2016).
In animal studies (Abrams 2016; Maguire and France 2018), cannabinoids and opioids are synergistic, with the analgesic efficacy of cannabinoids not reduced when opioid antagonists are administered. Human findings are variable; statistically significant reductions in pain scores, and similar opioid pharmacokinetics (with the exception of a reduced Cmax in the morphine group) pre and post vaporised cannabis use was found in chronic opioid users (morphine/oxycodone) (Abrams et al. 2011). In contrast, a small study found higher pain scores and greater rescue analgesia requirements post operatively in cannabis users, versus non-cannabis users (Jefferson et al. 2013). Chronic cannabinoid and cannabis users undergoing orthopaedic procedures showed higher post-operative pain scores without a significant increase in post-operative opioid consumption (Liu et al. 2018). All these studies have limited numbers of participants, and methodological issues that may confound the results.
In summary, cannabinoids and opioids are synergistic for both wanted and unwanted effects. Chronic cannabis users may have higher pain scores; it is unclear whether this is pathophysiological or a behavioral component of drug use.

Ketamine

Ketamine induces endogenous cannabinoid release (Pacheco et al. 2019; Ferreira et al. 2018), which may partially explain its role in anti-nociception. The psychomotor side effects of ketamine are enhanced with CBD administration, but no adverse behavioural or cardiovascular effects have been noted (Hallak et al. 2011).

Gabapentinoids

Gabapentin’s mechanism of action is via α2δ subunits on voltage-dependent calcium channels, with reduction in neural transmission. Similarly, activation of the CB receptor results in inhibition of the voltage dependent calcium channel (Pertwee 2015; Lile et al. 2016). Animal studies have shown the synergistic action of gabapentin and THC when used for the treatment of neuropathic pain, at the expense of increased side effects of THC (Atwal et al. 2017).
Human studies are limited; in multiple sclerosis, the combination of THC and gabapentin improved pain scores in neuropathic pain (Turcotte et al. 2015). High-dose gabapentin for management of cannabis tolerance produces THC like effects, and when gabapentin was used in combination with THC, these effects were seen to be increased, suggesting overlap of pharmacological actions (Lile et al. 2016).
In summary, the gabapentinoids and cannabinoids have overlapping pharmacological actions, with increased therapeutic and side effects when combination dosing is used.

Dexmedetomidine

There is limited evidence regarding potential interactions between cannabinoids and Dexmedetomidine. Animal studies have shown that a synthetic THC derivative (CP55,940) has additive or synergistic analgesic effects when administered with Dexmedetomidine, depending on the nociceptive stimulus utilised (Tham et al. 2005). The study failed to explain the mechanism of this apparent synergy; however, given the similar intracellular signalling mechanisms (calcium, potassium and cyclic AMP) activated by these medications and the close locality of the target receptors in the periaqueductal grey and substantia gelatinosa, receptor interaction is postulated (Tham et al. 2005).
Given the lack of current evidence around interactions in humans, further research should focus on this area.

Medical conditions where cannabinoids are recommended

A variety of National and Governmental organisations have provided reviews on the use of CBMs, producing recommendations with a varying hierarchy of evidence (Department of Health and Social Care 2018; Ghebreyesus 2019; Therapeutic Goods Administration 2017; Health Products Regualtory Authority 2017; National Academies of Sciences Engineering and Medicine 2017). Here, we review the commoner indications for CBMs.

Chronic pain

Information on the use of cannabinoids for chronic pain comes from trials, systematic reviews (SR), meta-analyses (MA) and organisational reports. The outcomes vary, and are limited by factors including study design, moderate to high risk of bias (Hauser et al. 2018a), limited participants (most recent SR/MA (Stockings et al. 2018a) identified 104 studies, 21 with > 100 participants), short duration of exposure to the cannabinoid (median eight weeks (Stockings et al. 2018a)) and varying definitions of “chronic pain”. Many studies within these systematic reviews are notable for high withdrawal rates in the treatment arms (Stockings et al. 2018a; Mucke et al. 2018).
The most recent SR concluded that the number needed to harm (NNTH) for cannabinoid use in chronic pain was 6 (opioids NNTH = 5) (Stockings et al. 2018a) with a number needed to treat (NNT) of 24 (30% reduction in pain). This compares unfavourably with opioids (NNT 4.3), pregabalin (7.7) and tricyclics (NNT 3.6) (Stockings et al. 2018a). When the pain intensity reduction (versus placebo) was pooled, it was equivalent to a 3 mm reduction on a 100 mm visual analogue scale. Taken with a higher risk of an adverse event and trial withdrawal (Stockings et al. 2018a), the authors suggested that whilst there was moderate evidence for pain reduction with cannabinoids compared with placebo (higher quality evidence for MS and neuropathic related pain), it seemed unlikely that cannabinoids are highly effective for chronic non-cancer pain.
Other SR/MAs make varying comments on the strength of the evidence, including weak recommendations (Meng et al. 2017), low strength (Rodríguez-Muñoz et al. 2016), moderate (Therapeutic Goods Administration 2017; Mucke et al. 2018; Whiting et al. 2015) (30% reduction pain relief), moderate to high (Aviram and Samuelly-Leichtag 2017), strong or “conclusive” (Abrams 2018) evidence for the use of cannabinoids in chronic pain. Others suggest a moderate to high risk of bias, concluding the evidence base is insufficient to make well found conclusions about the use of CBMs for cancer and non-cancer pain (Hauser et al. 2018b). Additionally, a large observational cohort study in Australia disputed cannabis use as an adjunct to reduce opiate consumption (Sánchez-Blázquez et al. 2014).
Globally, regulatory bodies have come to different conclusions. The Health Products Regulatory Authority (HPRA) of Ireland does not support CBMs as a treatment in chronic pain (Health Products Regualtory Authority 2017). The European Pain Federations recent position paper recommended CBMs be considered for chronic neuropathic pain, but as a third line agent, and stated there was insufficient evidence for CBMs for non-neuropathic chronic non-cancer pain (Hauser et al. 2018a). This is in direct contrast to the National Academies of Science Engineering and Medicine (NASEM) review on the health effects of cannabis and cannabinoids (National Academies of Sciences Engineering and Medicine 2017) which concluded that there was conclusive or substantial evidence for the use cannabis or cannabinoids for the treatment of pain in adults.
In summary, CBMs have a higher NNT than opioids, pregabalin or TCA, with a clinically insignificant pooled pain reduction of 3 mm on 100 mm VAS, and are thus unlikely to be effective in chronic, non-cancer pain, non-neuropathic pain. Additionally, other problems include study design and high withdrawal rates in intervention arms, with Cannabinoids demonstrating a higher risk of adverse events.

Nausea and vomiting secondary to chemotherapy

Nabilone (UK) and dronabinol (USA) are used to treat intractable post-chemotherapy nausea and vomiting (Abrams 2018), with the HPRA of Ireland recently permitting its use for this indication (Health Products Regualtory Authority 2017).
Reviews of cannabinoids for this indication have found them to be better than placebo (Whiting et al. 2015; Smith et al. 2015; Layeeque et al. 2006) of similar (Smith et al. 2015; Lewis et al. 1994) or better efficacy than antiemetics (dopamine antagonists) (Whiting et al. 2015), but with patients preferring CBMs (Smith et al. 2015). These reviews do not compare CBMs with steroids or serotonin (5HT3) antagonists. One randomised controlled trial utilising ondansetron as a comparator (Meiri et al. 2007) was stopped early due to recruitment difficulties, and had numerous methodological limitations including being underpowered for the authors conclusion that dronabinol was as efficacious as ondansetron.
The quality of evidence for the use of CBMs in preventing chemotherapy induced nausea and vomiting has been described as low (Whiting et al. 2015; Smith et al. 2015), “sometimes effective” (Therapeutic Goods Administration 2017) or conclusive/substantial evidence of benefit (Abrams 2018).
In summary, no completed studies have utilised modern antiemetics as a comparator, but cannabinoids are better than placebo, and display equivalent efficacy with dopamine antagonists. Further research will help determine the appropriate usage of CBM for nausea and vomiting.

Multiple sclerosis

Nabiximols (Sativex®) is licensed for multiple sclerosis (MS)-induced spasticity (Department of Health and Social Care 2018), which affects 17% of MS sufferers, with a similar proportion using cannabis for symptom control (Rice and Cameron 2017).
Previous MA/SR have produced various conclusions on the strength of the evidence of CBMs in MS-induced spasticity, ranging from low quality to conclusive evidence (Therapeutic Goods Administration 2017; Health Products Regualtory Authority 2017; Rice and Cameron 2017; Koppel et al. 2014; Whiting et al. 2015; Abrams 2018). A recent systematic review of reviews (Nielsen et al. 2018) for the use of cannabinoids in MS concluded that whilst the quality of the evidence from included studies was very low to low, five of the eleven reviews concluded that there was sufficient evidence for reduction in spasticity and/or pain in MS. However, the authors stated that despite the positive findings, the effect was small (Nielsen et al. 2018).
In summary, CBMs have a small positive effect on muscle spasticity, but the evidence quality is low.

Epilepsy

The United States Food and Drug Administration (FDA) (US Food and Drug Administration 2018) has recently approved cannabidiol oral solution (Epidiolex®) for the treatment of two forms of rare epilepsy in children aged over 2 years of age; Lennox-Gastaut syndrome and Dravet syndrome (Thiele et al. 2018; Devinsky et al. 2018; Devinsky et al. 2017). Most evidence is on the use of CBD, with the overall quality of the evidence in adults being limited (Koppel et al. 2014; Abrams 2018; Gloss and Vickrey 2014; Stockings et al. 2018b). Meta-analysis results pool effects in adults and children, with conclusions being influenced by the aforementioned paediatric studies (Sánchez-Blázquez et al. 2014; Devinsky et al. 2018; Devinsky et al. 2017; Gloss and Vickrey 2014; Stockings et al. 2018b). Outside of the USA, CBM use for epilepsy is not recommended in the UK (Department of Health and Social Care 2018), Ireland (Health Products Regualtory Authority 2017) and only once conventional treatments have failed in Australia (Therapeutic Goods Administration 2017).
In summary, the evidence base supports the use of CBD in children with certain neurological conditions, but not in adults.

Other

The NASEM review has considered CBMs for the treatment of other conditions, as detailed in Table 3 (Abrams 2018).
Table 3
Potential indications for CBM and evidence base
Evidence of benefit
Condition
Moderate
-Improving short-term sleep in sleep disturbance associated with OSA, Chronic pain, MS
Limited
-Improving symptoms of post-traumatic stress disorder, anxiety
-Improving appetite and decreasing weight loss associated with HIV/AIDS
-Tourette syndrome
None
-Treatment of cancer
-Irritable bowel syndrome symptoms
-Neurodegenerative conditions
-Addiction

Potential future uses in perioperative medicine

Nausea and vomiting prophylaxis and treatment

There is a paucity of evidence on the effects on post-operative nausea and vomiting (PONV). Nabilone (Cesamet®) (Lewis et al. 1994; Levin et al. 2017) and intravenous THC (Kleine-Brueggeney et al. 2015) have been shown to be ineffective for PONV. Nabilone premedication compared with placebo (Levin et al. 2017) or metoclopramide (Lewis et al. 1994) had no effect on PONV. Intravenous THC similarly showed a lack of effect, with early trial cessation due to an intolerable side effect profile (Kleine-Brueggeney et al. 2015).
Combination therapy (dronabinol and prochlorperazine) compared with routine care showed a reduction in the incidence of PONV, yet the retrospective nature and multiple confounders means the evidence has to be carefully interpreted (Layeeque et al. 2006).

Perioperative pain management

A systematic review looking at the efficacy of cannabinoids for acute pain management suggested no role for cannabinoids (Stevens and Higgins 2017).
For perioperative pain management, a small number of RCTs have been conducted with two studies suggesting benefit (Jain et al. 1981; Holdcroft et al. 2006). The first (Jain et al. 1981) showed significantly improved post-operative pain scores compared with placebo, but at the expense of increased side effects including drowsiness and dysphoria. The second (Holdcroft et al. 2006), a dose escalation study, with an oral capsule mixture of THC/cannabidiol (and other plant-based cannabinoid extracts) reported a similar NNT as other rescue analgesics, but with significantly increased side effects including sedation and nausea. The applicability of these results is limited by methodological issues and the small number of participants.
Six other studies (Ostenfeld et al. 2011; Beaulieu 2006; Buggy et al. 2003; Seeling et al. 2006; Kalliomäki et al. 2013; Guillaud et al. 1983) investigating the perioperative use of CBMs for analgesia showed no improvement in pain scores; one (Beaulieu 2006) showing significantly higher pain scores.
If CBMs are to be introduced into the clinical pharmacopoeia for perioperative analgesia, the potential for synergy with concurrently administered opioids (especially slow release formulations) in the perioperative period should be considered. One of the main concerns, and as recently highlighted by both the Anaesthesia Patient Safety Foundation (APSF) and ANZCAs faculty of pain medicine (Anaesthesia Patient Safety Foundation 2018; Australia and New Zealand College of Anaesthetists Faculty of Pain Medicine 2018) is the potential for opioid induced ventilatory impairment (OIVI) (Australia and New Zealand College of Anaesthetists Faculty of Pain Medicine 2018). We would recommend sedation soring be undertaken in these patients, as well as standardised order sets as recently recommended by the APSF (Anaesthesia Patient Safety Foundation 2018) and ANZCA (Australia and New Zealand College of Anaesthetists Faculty of Pain Medicine 2018).
In summary, cannabinoids may improve pain relief as part of multi-modal approach. There is an increased risk of adverse side effects including increased sedation and subsequent ventilatory impairment.

The future of CBMS

Further clarification on the role of non-CBD CBMs is expected later this year with the forthcoming UNODC vote on rescheduling as recommended by the WHO, increasing the focus on this group of medicines. With time, this may help to improve the evidence base, define clinical indications including potential therapeutic applications in perioperative medicine and provide outcome data from longer term use, which is currently lacking (Health Products Regualtory Authority 2017; Fitzcharles and Eisenberg 2018).
This latter point is arguably the most important, and whilst cannabis use per se has been associated with some cancers (prostate, glioma, cervical) and psychiatric morbidity, the quality of the evidence is limited, and it is uncertain if long-term effects of CBMs can be extrapolated from long term cannabis use (Nugent et al. 2017; National Academies of Sciences Engineering and Medicine 2017; Campbell et al. 2018).
Therefore, further research is required, and whilst one of the longest follow up studies of cannabis use in a medical setting (Ware et al. 2015) suggested no difference in serious adverse events between controls and cannabis users, the short duration of this and other studies involving CBMs limits conclusions on long-term safety (Hauser et al. 2018c). Long-term data on CBMs is now being collected through patient registries (national and pharmaceutical led) and observational studies providing reporting of adverse effects (Krcevski-Skvarc et al. 2018). Achieving greater clarity on the benefits and harms of CBMs may be affected by the legalisation of cannabis for recreational use in some territories (United Nations Office on Drugs and Crime 2018).

Conclusions

There are marked discrepancies in the literature regarding grading of the evidence base and the strength and quality of the resultant recommendations.
It is clear that with the increasing trend for legalisation of this class of medicines, and the large number of patients we as a specialty are involved with, the perioperative team need to have a broader understanding of the pharmacology interactions, and potential uses this group of drugs has.
As the evidence base increases, CBMs could become part of the perioperative teams’ armamentarium to help provide an opiate sparing multimodal analgesia regime as well as having a role in the management of common post-operative complications such as nausea and vomiting.

Acknowledgements

Not applicable.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
Zurück zum Zitat Abrams DI. The therapeutic effects of Cannabis and cannabinoids: an update from the National Academies of Sciences, Engineering and Medicine report. Eur J Intern Med. 2018;49:7–11.PubMedCrossRef Abrams DI. The therapeutic effects of Cannabis and cannabinoids: an update from the National Academies of Sciences, Engineering and Medicine report. Eur J Intern Med. 2018;49:7–11.PubMedCrossRef
Zurück zum Zitat Abrams DI, Couey P, Shade SB, Kelly ME, Benowitz NL. Cannabinoid–opioid interaction in chronic pain. Clin. Pharmacol Ther. 2011;90:844–51.PubMedCrossRef Abrams DI, Couey P, Shade SB, Kelly ME, Benowitz NL. Cannabinoid–opioid interaction in chronic pain. Clin. Pharmacol Ther. 2011;90:844–51.PubMedCrossRef
Zurück zum Zitat Anderson GD, Chan L. Pharmacokinetic drug interactions with tobacco, cannabinoids and smoking cessation products. Clin Pharmacokinet. 2016;55:1353–68.PubMedCrossRef Anderson GD, Chan L. Pharmacokinetic drug interactions with tobacco, cannabinoids and smoking cessation products. Clin Pharmacokinet. 2016;55:1353–68.PubMedCrossRef
Zurück zum Zitat Aragona M, Onesti E, Tomassini V, et al. Psychopathological and cognitive effects of therapeutic cannabinoids in multiple clerosis: A double-blind, placebo controlled, crossover study. Clin Neuropharmacol. 2009;32:41–7.PubMed Aragona M, Onesti E, Tomassini V, et al. Psychopathological and cognitive effects of therapeutic cannabinoids in multiple clerosis: A double-blind, placebo controlled, crossover study. Clin Neuropharmacol. 2009;32:41–7.PubMed
Zurück zum Zitat Atwal N, Casey SL, Mitchell VA, Vaughan CW. Neuropharmacology THC and gabapentin interactions in a mouse neuropathic pain model. Neuropharmacology. 2017;144:115–21.CrossRef Atwal N, Casey SL, Mitchell VA, Vaughan CW. Neuropharmacology THC and gabapentin interactions in a mouse neuropathic pain model. Neuropharmacology. 2017;144:115–21.CrossRef
Zurück zum Zitat Aviram J, Samuelly-Leichtag G. Efficacy of Cannabis-based medicines for pain management: a systematic review and meta-analysis of randomized controlled trials. Pain Physician. 2017;20:E755–96.PubMed Aviram J, Samuelly-Leichtag G. Efficacy of Cannabis-based medicines for pain management: a systematic review and meta-analysis of randomized controlled trials. Pain Physician. 2017;20:E755–96.PubMed
Zurück zum Zitat Bakas T, Van Nieuwenhuijzen PS, Devenish SO, Mcgregor IS, Arnold JC, Chebib M. The direct actions of cannabidiol and 2-arachidonoyl glycerol at GABA A receptors. Pharmacol. Res. 2017;119:358–70.PubMedCrossRef Bakas T, Van Nieuwenhuijzen PS, Devenish SO, Mcgregor IS, Arnold JC, Chebib M. The direct actions of cannabidiol and 2-arachidonoyl glycerol at GABA A receptors. Pharmacol. Res. 2017;119:358–70.PubMedCrossRef
Zurück zum Zitat Barnes MP. The case for medical cannabis—an essay. BMJ. 2018;362:k3230.CrossRef Barnes MP. The case for medical cannabis—an essay. BMJ. 2018;362:k3230.CrossRef
Zurück zum Zitat Beaulieu P. Effects of nabilone, a synthetic cannabinoid, on postoperative pain. Can J Anesth. 2006;53(8):769–75.PubMedCrossRef Beaulieu P. Effects of nabilone, a synthetic cannabinoid, on postoperative pain. Can J Anesth. 2006;53(8):769–75.PubMedCrossRef
Zurück zum Zitat Beaulieu P, Boulanger A, Desroches J, Clark AJ. Medical cannabis: considerations for the anesthesiologist and pain physician. Can J Anaesth. 2016;63:608–24.PubMedCrossRef Beaulieu P, Boulanger A, Desroches J, Clark AJ. Medical cannabis: considerations for the anesthesiologist and pain physician. Can J Anaesth. 2016;63:608–24.PubMedCrossRef
Zurück zum Zitat Bie B, Wu J, Foss JF, Naguib M. An overview of the cannabinoid type 2 receptor system and its therapeutic potential. Curr Opin Anesthesiol. 2018;31:407–14.CrossRef Bie B, Wu J, Foss JF, Naguib M. An overview of the cannabinoid type 2 receptor system and its therapeutic potential. Curr Opin Anesthesiol. 2018;31:407–14.CrossRef
Zurück zum Zitat Bilkei-Gorzo A, Albayram O, Ativie F, Chasan S, Zimmer T. Cannabinoid 1 receptor signaling on GABAergic neurons influences astrocytes in the ageing brain. PLoS One. 2018;13(8):1–21.CrossRef Bilkei-Gorzo A, Albayram O, Ativie F, Chasan S, Zimmer T. Cannabinoid 1 receptor signaling on GABAergic neurons influences astrocytes in the ageing brain. PLoS One. 2018;13(8):1–21.CrossRef
Zurück zum Zitat Brand PA, Paris A, Bein B, et al. Propofol sedation is reduced by δ9-tetrahydrocannabinol in mice. Anesth Analg. 2008;107:102–6.PubMedCrossRef Brand PA, Paris A, Bein B, et al. Propofol sedation is reduced by δ9-tetrahydrocannabinol in mice. Anesth Analg. 2008;107:102–6.PubMedCrossRef
Zurück zum Zitat Bridgeman MB, Abazia DT. Medicinal cannabis: history, pharmacology, and implications for the acute care setting. Pharm. Ther. 2017;42:180–8. Bridgeman MB, Abazia DT. Medicinal cannabis: history, pharmacology, and implications for the acute care setting. Pharm. Ther. 2017;42:180–8.
Zurück zum Zitat Bryson EO, Frost EAM. The perioperative implications of tobacco, marijuana, and other inhaled toxins. Int Anesthesiol Clin. 2011;49:103–18.PubMedCrossRef Bryson EO, Frost EAM. The perioperative implications of tobacco, marijuana, and other inhaled toxins. Int Anesthesiol Clin. 2011;49:103–18.PubMedCrossRef
Zurück zum Zitat Buggy DJ, Toogood L, Maric S, Sharpe P, Lambert DG, Rowbotham DJ. Lack of analgesic efficacy of oral δ-9-tetrahydrocannabinol in postoperative pain. Pain. 2003;106:169–72.PubMedCrossRef Buggy DJ, Toogood L, Maric S, Sharpe P, Lambert DG, Rowbotham DJ. Lack of analgesic efficacy of oral δ-9-tetrahydrocannabinol in postoperative pain. Pain. 2003;106:169–72.PubMedCrossRef
Zurück zum Zitat Campbell G, Hall WD, Peacock A, et al. Effect of cannabis use in people with chronic non-cancer pain prescribed opioids: findings from a 4-year prospective cohort study. Lancet Public Health. 2018;3:e341–50.PubMedPubMedCentralCrossRef Campbell G, Hall WD, Peacock A, et al. Effect of cannabis use in people with chronic non-cancer pain prescribed opioids: findings from a 4-year prospective cohort study. Lancet Public Health. 2018;3:e341–50.PubMedPubMedCentralCrossRef
Zurück zum Zitat Dame Sally Davies. The therapeutic and medicinal benefits of Cannabis based products-a review of recent evidence. London: Cannabis Scheduling Review Part 1; 2018. Dame Sally Davies. The therapeutic and medicinal benefits of Cannabis based products-a review of recent evidence. London: Cannabis Scheduling Review Part 1; 2018.
Zurück zum Zitat Devinsky O, Croos H, Laux L, et al. Trial of cannabidiol for drug-resistant seizures in the Dravet syndrome. N Engl J Med. 2017;376:2011–20.PubMedCrossRef Devinsky O, Croos H, Laux L, et al. Trial of cannabidiol for drug-resistant seizures in the Dravet syndrome. N Engl J Med. 2017;376:2011–20.PubMedCrossRef
Zurück zum Zitat Devinsky O, Patel AD, Cross JH, et al. Effect of cannabidiol on drop seizures in the Lennox–Gastaut syndrome. N Engl J Med. 2018;378:1888–97.PubMedCrossRef Devinsky O, Patel AD, Cross JH, et al. Effect of cannabidiol on drop seizures in the Lennox–Gastaut syndrome. N Engl J Med. 2018;378:1888–97.PubMedCrossRef
Zurück zum Zitat Dickerson SJ. Cannabis and its effect on anesthesia. J Am Assoc Nurse Anesth. 1980;48:526–8. Dickerson SJ. Cannabis and its effect on anesthesia. J Am Assoc Nurse Anesth. 1980;48:526–8.
Zurück zum Zitat Expert Committee on Drug Dependence. Cannabidiol (CBD) critical review report. Geneva: World Health Organization; 2018. Expert Committee on Drug Dependence. Cannabidiol (CBD) critical review report. Geneva: World Health Organization; 2018.
Zurück zum Zitat Ferreira RCM, Castor MGM, Piscitelli F, Di Marzo V, IDuarte IDG, Romero TRL. The involvement of the endocannabinoid system in the peripheral antinociceptive action of ketamine. J. Pain. 2018;19:487–95.PubMedCrossRef Ferreira RCM, Castor MGM, Piscitelli F, Di Marzo V, IDuarte IDG, Romero TRL. The involvement of the endocannabinoid system in the peripheral antinociceptive action of ketamine. J. Pain. 2018;19:487–95.PubMedCrossRef
Zurück zum Zitat Fitzcharles MA, Eisenberg E. Medical cannabis: a forward vision for the clinician. Eur J Pain. 2018;22:485–91.PubMedCrossRef Fitzcharles MA, Eisenberg E. Medical cannabis: a forward vision for the clinician. Eur J Pain. 2018;22:485–91.PubMedCrossRef
Zurück zum Zitat Flisberg P, Paech MJ, Shah T, Ledowski T, Kurowski I, Parsons R. Induction dose of propofol in patients using cannabis. Eur J Anaesthesiol. 2009;26:192–5.PubMedCrossRef Flisberg P, Paech MJ, Shah T, Ledowski T, Kurowski I, Parsons R. Induction dose of propofol in patients using cannabis. Eur J Anaesthesiol. 2009;26:192–5.PubMedCrossRef
Zurück zum Zitat Gloss D, Vickrey B. Cannabinoids for epilepsy (Review). Cochrane Database Syst Rev 2014; 3: Art. No.: CD009270. Gloss D, Vickrey B. Cannabinoids for epilepsy (Review). Cochrane Database Syst Rev 2014; 3: Art. No.: CD009270.
Zurück zum Zitat Guillaud J, Legagneux F, Paulet C, Leoni J, Lassner J. Mortality in Anaesthesia. European Acaemy of Aaesthesiology vol 3 Springer Berlin, Heidelberg. Comparison of Levonantradol 1 or 2mg IM with Pethdiine and Placebo for postoperative analgesia. 1983 .CrossRef Guillaud J, Legagneux F, Paulet C, Leoni J, Lassner J. Mortality in Anaesthesia. European Acaemy of Aaesthesiology vol 3 Springer Berlin, Heidelberg. Comparison of Levonantradol 1 or 2mg IM with Pethdiine and Placebo for postoperative analgesia. 1983 .CrossRef
Zurück zum Zitat Hallak JEC, Dursun SM, Bosi DC, et al. The interplay of cannabinoid and NMDA glutamate receptor systems in humans : preliminary evidence of interactive effects of cannabidiol and ketamine in healthy human subjects. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2011;35(1):198–202.PubMedCrossRef Hallak JEC, Dursun SM, Bosi DC, et al. The interplay of cannabinoid and NMDA glutamate receptor systems in humans : preliminary evidence of interactive effects of cannabidiol and ketamine in healthy human subjects. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2011;35(1):198–202.PubMedCrossRef
Zurück zum Zitat Hauser W, Finn DP, Kalso E, et al. European Pain Federation (EFIC) position paper on appropriate use of cannabis-based medicines and medical cannabis for chronic pain management. Eur J Pain. 2018a;22:1547–64.PubMedCrossRef Hauser W, Finn DP, Kalso E, et al. European Pain Federation (EFIC) position paper on appropriate use of cannabis-based medicines and medical cannabis for chronic pain management. Eur J Pain. 2018a;22:1547–64.PubMedCrossRef
Zurück zum Zitat Hauser W, Finnerup NB, Moore RA. Systematic reviews with meta-analysis on cannabis-based medicines for chronic pain: a methodological and political minefield. Pain. 2018c;159:1906–7.PubMedCrossRef Hauser W, Finnerup NB, Moore RA. Systematic reviews with meta-analysis on cannabis-based medicines for chronic pain: a methodological and political minefield. Pain. 2018c;159:1906–7.PubMedCrossRef
Zurück zum Zitat Hauser W, Petzke F, Fitzcharles MA. Efficacy, tolerability and safety of cannabis-based medicines for chronic pain management—an overview of systematic reviews. Eur J Pain. 2018b;22:455–70.PubMedCrossRef Hauser W, Petzke F, Fitzcharles MA. Efficacy, tolerability and safety of cannabis-based medicines for chronic pain management—an overview of systematic reviews. Eur J Pain. 2018b;22:455–70.PubMedCrossRef
Zurück zum Zitat Heuberger J, Guan Z, Oyetayo OO, et al. Population pharmacokinetic model of THC integrates oral, intravenous, and pulmonary dosing and characterizes short- and long-term pharmacokinetics. Clin Pharmacokinet. 2015;54:209–19.PubMedCrossRef Heuberger J, Guan Z, Oyetayo OO, et al. Population pharmacokinetic model of THC integrates oral, intravenous, and pulmonary dosing and characterizes short- and long-term pharmacokinetics. Clin Pharmacokinet. 2015;54:209–19.PubMedCrossRef
Zurück zum Zitat Holdcroft A, Maze M, Dore C, Tebbs S, Thompson S. A multicenter dose-escalation study of the analgesic and adverse effects of an oral cannabis extract (Cannador) for postoperative pain management. Anesthesiology. 2006;104(5):1040–6.PubMedCrossRef Holdcroft A, Maze M, Dore C, Tebbs S, Thompson S. A multicenter dose-escalation study of the analgesic and adverse effects of an oral cannabis extract (Cannador) for postoperative pain management. Anesthesiology. 2006;104(5):1040–6.PubMedCrossRef
Zurück zum Zitat Hosking RD, Zajicek JP. Therapeutic potential of cannabis in pain medicine. Br J Anaesth. 2008;101:59–68.PubMedCrossRef Hosking RD, Zajicek JP. Therapeutic potential of cannabis in pain medicine. Br J Anaesth. 2008;101:59–68.PubMedCrossRef
Zurück zum Zitat Ibera C, Shalom B, Saifi F, Shruder J, Davidson E. Effects of cannabis extract premedication on anesthetic depth. Harefuah. 2018;157:162–6.PubMed Ibera C, Shalom B, Saifi F, Shruder J, Davidson E. Effects of cannabis extract premedication on anesthetic depth. Harefuah. 2018;157:162–6.PubMed
Zurück zum Zitat International Drug Policy Consortium. The United Nations General Assembly Special Session (UNGASS) on the World Drug Problem. London: Report of Proceedings; 2016. International Drug Policy Consortium. The United Nations General Assembly Special Session (UNGASS) on the World Drug Problem. London: Report of Proceedings; 2016.
Zurück zum Zitat Jain A, Ryan J, McMahon G, Smith G. Evaluation of intramuscular levonantradol and placebo in acutepostoperative. J Clin Pharmacol. 1981;21:320–6.CrossRef Jain A, Ryan J, McMahon G, Smith G. Evaluation of intramuscular levonantradol and placebo in acutepostoperative. J Clin Pharmacol. 1981;21:320–6.CrossRef
Zurück zum Zitat Jefferson D, Harding H, Cawich S, Jackson-Gibson A. Postoperative analgesia in the Jamaican cannabis user. J Psychoactive Drugs. 2013;45:227–32.PubMedCrossRef Jefferson D, Harding H, Cawich S, Jackson-Gibson A. Postoperative analgesia in the Jamaican cannabis user. J Psychoactive Drugs. 2013;45:227–32.PubMedCrossRef
Zurück zum Zitat Kalliomäki J, Segerdahl M, Webster L, et al. Evaluation of the analgesic efficacy of AZD1940, a novel cannabinoid agonist, on post-operative pain after lower third molar surgical removal. Scand. J. Pain. 2013;4:17–22.PubMedCrossRef Kalliomäki J, Segerdahl M, Webster L, et al. Evaluation of the analgesic efficacy of AZD1940, a novel cannabinoid agonist, on post-operative pain after lower third molar surgical removal. Scand. J. Pain. 2013;4:17–22.PubMedCrossRef
Zurück zum Zitat Karschner EL, David Darwin W, Goodwin RS, Wright S, Huestis MA. Plasma cannabinoid pharmacokinetics following controlled oral Δ 9-tetrahydrocannabinol and oromucosal cannabis extract administration. Clin Chem. 2011;57:66–75.PubMedCrossRef Karschner EL, David Darwin W, Goodwin RS, Wright S, Huestis MA. Plasma cannabinoid pharmacokinetics following controlled oral Δ 9-tetrahydrocannabinol and oromucosal cannabis extract administration. Clin Chem. 2011;57:66–75.PubMedCrossRef
Zurück zum Zitat Kleine-Brueggeney M, Greif R, Brenneisen R, Urwyler N, Stueber F, Theiler LG. Intravenous Delta-9-tetrahydrocannabinol to prevent postoperative nausea and vomiting: A randomized controlled trial. Anesth Analg. 2015;121:1157–64.PubMedCrossRef Kleine-Brueggeney M, Greif R, Brenneisen R, Urwyler N, Stueber F, Theiler LG. Intravenous Delta-9-tetrahydrocannabinol to prevent postoperative nausea and vomiting: A randomized controlled trial. Anesth Analg. 2015;121:1157–64.PubMedCrossRef
Zurück zum Zitat Koppel BS, Brust JCM, Fife T, et al. Systematic review: efficacy and safety of medical marijuana in selected neurologic disorders: report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology 2014;82:1556–1563PubMedPubMedCentralCrossRef Koppel BS, Brust JCM, Fife T, et al. Systematic review: efficacy and safety of medical marijuana in selected neurologic disorders: report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology 2014;82:1556–1563PubMedPubMedCentralCrossRef
Zurück zum Zitat Krcevski-Skvarc N, Wells C, Hauser W. Availability and approval of cannabis-based medicines for chronic pain management and palliative / supportive care in Europe : A survey of the status in the chapters of the European Pain Federation. Eur J Pain. 2018;22:440–54.PubMedCrossRef Krcevski-Skvarc N, Wells C, Hauser W. Availability and approval of cannabis-based medicines for chronic pain management and palliative / supportive care in Europe : A survey of the status in the chapters of the European Pain Federation. Eur J Pain. 2018;22:440–54.PubMedCrossRef
Zurück zum Zitat Kumar RN, Chambers WA, Pertwee RG. Pharmacological actions and therapeutic uses of cannabis and cannabinoids. Anaesthesia. 2001;56:1059–68.PubMedCrossRef Kumar RN, Chambers WA, Pertwee RG. Pharmacological actions and therapeutic uses of cannabis and cannabinoids. Anaesthesia. 2001;56:1059–68.PubMedCrossRef
Zurück zum Zitat Kumar SSH, Dass LL, Sharma AK. Cannabis Indica (Bhang) extract as preanaesthetic to propofol anaesthesia in dogs. Journal of Applied Animal Research. 2010;37:125–7.CrossRef Kumar SSH, Dass LL, Sharma AK. Cannabis Indica (Bhang) extract as preanaesthetic to propofol anaesthesia in dogs. Journal of Applied Animal Research. 2010;37:125–7.CrossRef
Zurück zum Zitat Laaris N, Good CH, Lupica CR. Δ9 -tetrahydrocannabinol is a full agonist at CB1 receptors on GABA neuron axon terminals in the hippocampus. Neuropharmacology. 2011;59(443):121–7. Laaris N, Good CH, Lupica CR. Δ9 -tetrahydrocannabinol is a full agonist at CB1 receptors on GABA neuron axon terminals in the hippocampus. Neuropharmacology. 2011;59(443):121–7.
Zurück zum Zitat Layeeque R, Siegel E, Kass R, et al. Prevention of nausea and vomiting following breast surgery. Am J Surg. 2006;191:767–72.PubMedCrossRef Layeeque R, Siegel E, Kass R, et al. Prevention of nausea and vomiting following breast surgery. Am J Surg. 2006;191:767–72.PubMedCrossRef
Zurück zum Zitat Levin DN, Dulberg Z, Chan AW, Hare GM, Mazer D, Hong A. A randomized-controlled trial of nabilone for the prevention of acute postoperative nausea and vomiting in elective surgery. Can J Anesth. 2017;64:385–95.PubMedCrossRef Levin DN, Dulberg Z, Chan AW, Hare GM, Mazer D, Hong A. A randomized-controlled trial of nabilone for the prevention of acute postoperative nausea and vomiting in elective surgery. Can J Anesth. 2017;64:385–95.PubMedCrossRef
Zurück zum Zitat Lewis IH, Campbell DN, Barrowcliffe MP. Effect of nabilone on nausea and vomiting after total abdominal hysterectomy. Br J Anaesth. 1994;73:244–6.PubMedCrossRef Lewis IH, Campbell DN, Barrowcliffe MP. Effect of nabilone on nausea and vomiting after total abdominal hysterectomy. Br J Anaesth. 1994;73:244–6.PubMedCrossRef
Zurück zum Zitat Lile J, Wesley M, Kelly T, Hays R. Separate and combined effects of gabapentin and 9THC in humans discriminating 9THC. Behav Pharmacol. 2016;27:215–24.PubMedPubMedCentralCrossRef Lile J, Wesley M, Kelly T, Hays R. Separate and combined effects of gabapentin and 9THC in humans discriminating 9THC. Behav Pharmacol. 2016;27:215–24.PubMedPubMedCentralCrossRef
Zurück zum Zitat Lotsch J, Weyer-Menkhoff I, Tegeder I. Current evidence of cannabinoid-based analgesia obtained in preclinical and human experimental settings. Eur. J. Pain. 2018;22:471–84.PubMedCrossRef Lotsch J, Weyer-Menkhoff I, Tegeder I. Current evidence of cannabinoid-based analgesia obtained in preclinical and human experimental settings. Eur. J. Pain. 2018;22:471–84.PubMedCrossRef
Zurück zum Zitat Lucas CJ, Galettis P, Schneider J. The pharmacokinetics and the pharmacodynamics of cannabinoids. Br J Clin Pharm. 2018;84:2477–82.CrossRef Lucas CJ, Galettis P, Schneider J. The pharmacokinetics and the pharmacodynamics of cannabinoids. Br J Clin Pharm. 2018;84:2477–82.CrossRef
Zurück zum Zitat Maguire DR, France CP. Antinociceptive effects of mixtures of mu opioid receptor agonists and cannabinoid receptor agonists in rats : Impact of drug and fixed-dose ratio. Eur J Pharmacol. 2018;819:217–24.PubMedCrossRef Maguire DR, France CP. Antinociceptive effects of mixtures of mu opioid receptor agonists and cannabinoid receptor agonists in rats : Impact of drug and fixed-dose ratio. Eur J Pharmacol. 2018;819:217–24.PubMedCrossRef
Zurück zum Zitat Manzanares J, Corchero J, Romero J, Fernández-Ruiz JJ, Ramos J, Fuentes JA. Pharmacological and biochemical interactions between opioids and cannabinoids. Trends Pharmacol Sci. 1999;20:287–94.PubMedCrossRef Manzanares J, Corchero J, Romero J, Fernández-Ruiz JJ, Ramos J, Fuentes JA. Pharmacological and biochemical interactions between opioids and cannabinoids. Trends Pharmacol Sci. 1999;20:287–94.PubMedCrossRef
Zurück zum Zitat Mead A. The legal status of cannabis (marijuana) and cannabidiol (CBD) under U.S. law. Epilepsy Behav. 2017;70:288–91.PubMedCrossRef Mead A. The legal status of cannabis (marijuana) and cannabidiol (CBD) under U.S. law. Epilepsy Behav. 2017;70:288–91.PubMedCrossRef
Zurück zum Zitat Meiri E, Jhangiani H, Vredenburgh JJ, et al. Efficacy of dronabinol alone and in combination with ondansetron versus ondansetron alone for delayed chemotherapy-induced nausea and vomiting. Curr Med Res Opin. 2007;23:533–43.PubMedCrossRef Meiri E, Jhangiani H, Vredenburgh JJ, et al. Efficacy of dronabinol alone and in combination with ondansetron versus ondansetron alone for delayed chemotherapy-induced nausea and vomiting. Curr Med Res Opin. 2007;23:533–43.PubMedCrossRef
Zurück zum Zitat Meng H, Johnston B, Englesakis M, Moulin D, Bhatia A. Selective cannabinoids for chronic neuropathic pain : a systematic review and meta-analysis. Anesth Analg. 2017;125(5):1638–52.PubMedCrossRef Meng H, Johnston B, Englesakis M, Moulin D, Bhatia A. Selective cannabinoids for chronic neuropathic pain : a systematic review and meta-analysis. Anesth Analg. 2017;125(5):1638–52.PubMedCrossRef
Zurück zum Zitat Mucke M, Phillips T, Radbruch L, Petzke F, Hauser W. Cannabis-based medicines for chronic neuropathic pain in adults (review). Cochrane Database Syst. Rev. 2018;3:CD012182.PubMed Mucke M, Phillips T, Radbruch L, Petzke F, Hauser W. Cannabis-based medicines for chronic neuropathic pain in adults (review). Cochrane Database Syst. Rev. 2018;3:CD012182.PubMed
Zurück zum Zitat National Academies of Sciences Engineering and Medicine. The health effects of cannabis and cannabinoids: the current state of evidence nad recommendations for research. Washington, DC: The National Academies Press; 2017. National Academies of Sciences Engineering and Medicine. The health effects of cannabis and cannabinoids: the current state of evidence nad recommendations for research. Washington, DC: The National Academies Press; 2017.
Zurück zum Zitat Nielsen S, Germanos R, Weier M, et al. The use of cannabis and cannabinoids in treating symptoms of multiple sclerosis: a systematic review of reviews. Curr Neurol Neurosci Rep. 2018;18:8.PubMedCrossRef Nielsen S, Germanos R, Weier M, et al. The use of cannabis and cannabinoids in treating symptoms of multiple sclerosis: a systematic review of reviews. Curr Neurol Neurosci Rep. 2018;18:8.PubMedCrossRef
Zurück zum Zitat Nugent SM, Morasco BJ, O’Neil ME, et al. The effects of cannabis among adults with chronic pain and an overview of general harms a systematic review. Ann Intern Med. 2017;167:319–31.PubMedCrossRef Nugent SM, Morasco BJ, O’Neil ME, et al. The effects of cannabis among adults with chronic pain and an overview of general harms a systematic review. Ann Intern Med. 2017;167:319–31.PubMedCrossRef
Zurück zum Zitat Ostenfeld T, Price J, Albanese M, et al. A randomized, controlled study to investigate the analgesic efficacy of single doses of the molar tooth extraction. J. Clin. Pain. 2011;27(8):668–76.CrossRef Ostenfeld T, Price J, Albanese M, et al. A randomized, controlled study to investigate the analgesic efficacy of single doses of the molar tooth extraction. J. Clin. Pain. 2011;27(8):668–76.CrossRef
Zurück zum Zitat Pacheco F, Roberto T, Romero L, Dimitri I, Duarte G. Ketamine induces central antinociception mediated by endogenous cannabinoids and activation of CB 1 receptors. Neurosci. Lett. 2019;699:140–4.PubMedCrossRef Pacheco F, Roberto T, Romero L, Dimitri I, Duarte G. Ketamine induces central antinociception mediated by endogenous cannabinoids and activation of CB 1 receptors. Neurosci. Lett. 2019;699:140–4.PubMedCrossRef
Zurück zum Zitat Pertwee RG, Howlett AC, Abood ME, et al. International union of basic and clinical pharmacology . LXXIX . Cannabinoid receptors and their ligands: beyond CB 1 and CB 2. Pharmacol Rev. 2010;62:588–631.PubMedPubMedCentralCrossRef Pertwee RG, Howlett AC, Abood ME, et al. International union of basic and clinical pharmacology . LXXIX . Cannabinoid receptors and their ligands: beyond CB 1 and CB 2. Pharmacol Rev. 2010;62:588–631.PubMedPubMedCentralCrossRef
Zurück zum Zitat Reynolds JR. Therapeutical uses and toxic effects of cannabis indica. Lancet. 1890;135:637–8.CrossRef Reynolds JR. Therapeutical uses and toxic effects of cannabis indica. Lancet. 1890;135:637–8.CrossRef
Zurück zum Zitat Rice J, Cameron M. Cannabinoids for treatment of MS symptoms: state of the evidence. Curr Neurol Neurosci Rep. 2017;18:1–10. Rice J, Cameron M. Cannabinoids for treatment of MS symptoms: state of the evidence. Curr Neurol Neurosci Rep. 2017;18:1–10.
Zurück zum Zitat Richtig G, Bosse G, Arlt F, Heymann CV. Cannabis consumption before surgery may be associated with increased tolerance of anesthetic drugs: A case report. Int J Case Rep. 2015;6:436–8. Richtig G, Bosse G, Arlt F, Heymann CV. Cannabis consumption before surgery may be associated with increased tolerance of anesthetic drugs: A case report. Int J Case Rep. 2015;6:436–8.
Zurück zum Zitat Rodríguez-Muñoz M, Sánchez-Blázquez P, Merlos M, Garzon-Nino J. Endocannabinoid control of glutamate NMDA receptors : the therapeutic potential and consequences of dysfunction. Oncotarget. 2016;34:55840–62. Rodríguez-Muñoz M, Sánchez-Blázquez P, Merlos M, Garzon-Nino J. Endocannabinoid control of glutamate NMDA receptors : the therapeutic potential and consequences of dysfunction. Oncotarget. 2016;34:55840–62.
Zurück zum Zitat Rong C, Carmona NE, Lee YL, et al. Drug-drug interactions as a result of co-administering ∆ 9-THC and CBD with other psychotropic agents. Expert Opin Drug Saf. 2017;17:51–4.PubMedCrossRef Rong C, Carmona NE, Lee YL, et al. Drug-drug interactions as a result of co-administering ∆ 9-THC and CBD with other psychotropic agents. Expert Opin Drug Saf. 2017;17:51–4.PubMedCrossRef
Zurück zum Zitat Sánchez-Blázquez P, Rodríguez-Muñoz M, Garzón J. The cannabinoid receptor 1 associates with NMDA receptors to produce glutamatergic hypofunction : implications in psychosis and schizophrenia. Front. Pharmacol. 2014;4:1–10.CrossRef Sánchez-Blázquez P, Rodríguez-Muñoz M, Garzón J. The cannabinoid receptor 1 associates with NMDA receptors to produce glutamatergic hypofunction : implications in psychosis and schizophrenia. Front. Pharmacol. 2014;4:1–10.CrossRef
Zurück zum Zitat Scavone J, Sterling R, Van Bockstaele E. Cannabinoid and opioid interactions: implications for opiate dependence and withdrawal. Neuroscience. 2013;248:637–54.PubMedCrossRef Scavone J, Sterling R, Van Bockstaele E. Cannabinoid and opioid interactions: implications for opiate dependence and withdrawal. Neuroscience. 2013;248:637–54.PubMedCrossRef
Zurück zum Zitat Schelling G, Hauer D, Azad SC, et al. Effects of general anesthesia on anandamide blood levels in humans. Anesthesiology. 2006;104(2):273–7.PubMedCrossRef Schelling G, Hauer D, Azad SC, et al. Effects of general anesthesia on anandamide blood levels in humans. Anesthesiology. 2006;104(2):273–7.PubMedCrossRef
Zurück zum Zitat Schuster J, Ates M, Brune K, Gühring H. The cannabinoids R(−)-7-hydroxy-delta-6-tetra-hydrocannabinol-dimethylheptyl (HU-210), 2-O-arachidonoylglycerylether (HU-310) and arachidonyl-2-chloroethylamide (ACEA) increase isoflurane provoked sleep duration by activation of cannabinoids 1 (CB1)-recep. Neuroscience Letters. 2002;326:196–200.PubMedCrossRef Schuster J, Ates M, Brune K, Gühring H. The cannabinoids R(−)-7-hydroxy-delta-6-tetra-hydrocannabinol-dimethylheptyl (HU-210), 2-O-arachidonoylglycerylether (HU-310) and arachidonyl-2-chloroethylamide (ACEA) increase isoflurane provoked sleep duration by activation of cannabinoids 1 (CB1)-recep. Neuroscience Letters. 2002;326:196–200.PubMedCrossRef
Zurück zum Zitat Seeling W, Kneer L, Buchele B, et al. Delta(9)-tetrahydrocannabinol and the opioid receptor agonist piritramide do not act synergistically in postoperative pain. Der Anästhesist. 2006;55:391–400.CrossRef Seeling W, Kneer L, Buchele B, et al. Delta(9)-tetrahydrocannabinol and the opioid receptor agonist piritramide do not act synergistically in postoperative pain. Der Anästhesist. 2006;55:391–400.CrossRef
Zurück zum Zitat Smith L, Azariah F, Lavender V, Stoner N, Bettiol S. Cannabinoids for nausea and vomiting in adults with cancer receiving chemotherapy (Review). Cochrane Database Syst Rev. 2015; 11: Art. No. CD009464 Smith L, Azariah F, Lavender V, Stoner N, Bettiol S. Cannabinoids for nausea and vomiting in adults with cancer receiving chemotherapy (Review). Cochrane Database Syst Rev. 2015; 11: Art. No. CD009464
Zurück zum Zitat Stevens AJ, Higgins MD. A systematic review of the analgesic efficacy of cannabinoid medications in the management of acute pain. Acta Anaesth Scand. 2017;61:268–80.PubMedCrossRef Stevens AJ, Higgins MD. A systematic review of the analgesic efficacy of cannabinoid medications in the management of acute pain. Acta Anaesth Scand. 2017;61:268–80.PubMedCrossRef
Zurück zum Zitat Stockings E, Campbell G, Hall WD, et al. Cannabis and cannabinoids for the treatment of people with chronic non-cancer pain conditions. Pain. 2018a;159:1932–54.PubMedCrossRef Stockings E, Campbell G, Hall WD, et al. Cannabis and cannabinoids for the treatment of people with chronic non-cancer pain conditions. Pain. 2018a;159:1932–54.PubMedCrossRef
Zurück zum Zitat Stockings E, Zagic D, Campbell G, Weier M, Hall WD, Nielsen S, Herkes GK, et al. Evidence for cannabis and cannabinoids for epilepsy: a systematic review of controlled and observational evidence. J Neurol Neurosurg Psychiatr. 2018b;89:741–53.CrossRef Stockings E, Zagic D, Campbell G, Weier M, Hall WD, Nielsen S, Herkes GK, et al. Evidence for cannabis and cannabinoids for epilepsy: a systematic review of controlled and observational evidence. J Neurol Neurosurg Psychiatr. 2018b;89:741–53.CrossRef
Zurück zum Zitat Stoelting R, Martz R, Gartner J, Creasser C, Brown D, Forney R. Effects of delta-9-tetrahydrocannibinol on Halothane MAC in dogs. Anesthesiology. 1973;35:521–4.CrossRef Stoelting R, Martz R, Gartner J, Creasser C, Brown D, Forney R. Effects of delta-9-tetrahydrocannibinol on Halothane MAC in dogs. Anesthesiology. 1973;35:521–4.CrossRef
Zurück zum Zitat Tham SM, Angus JA, Tudor EM, Wright CE. Synergistic and additive interactions of the cannabinoid agonist CP55,940 with μ opioid receptor and α 2-adrenoceptor agonists in acute pain models in mice. Br. J. Pharmacol. 2005;144(6):875–84.PubMedCrossRef Tham SM, Angus JA, Tudor EM, Wright CE. Synergistic and additive interactions of the cannabinoid agonist CP55,940 with μ opioid receptor and α 2-adrenoceptor agonists in acute pain models in mice. Br. J. Pharmacol. 2005;144(6):875–84.PubMedCrossRef
Zurück zum Zitat Therapeutic Goods Administration. Guidance for the use of medicinal cannabis in Australia Overview. Woden ACT: Australian Government Department of Health; 2017. Version 1 Therapeutic Goods Administration. Guidance for the use of medicinal cannabis in Australia Overview. Woden ACT: Australian Government Department of Health; 2017. Version 1
Zurück zum Zitat Thiele EA, Marsh ED, French JA, et al. Cannabidiol in patients with seizures associated with Lennox-Gastaut syndrome (GWPCARE4): a randomised, double-blind, placebo-controlled phase 3 trial. The Lancet. 2018;391:1085–96.CrossRef Thiele EA, Marsh ED, French JA, et al. Cannabidiol in patients with seizures associated with Lennox-Gastaut syndrome (GWPCARE4): a randomised, double-blind, placebo-controlled phase 3 trial. The Lancet. 2018;391:1085–96.CrossRef
Zurück zum Zitat Transnational Institute, Global Drug Policy Observatory. UNGASS 2016: a broken or broad consensus? UN summit cannot hide growing divergence in the global drug policy landscape. Amsterdam: United Nations; 2016. Transnational Institute, Global Drug Policy Observatory. UNGASS 2016: a broken or broad consensus? UN summit cannot hide growing divergence in the global drug policy landscape. Amsterdam: United Nations; 2016.
Zurück zum Zitat Turcotte D, Doupe M, Torabi M, et al. Nabilone as an adjunctive to gabapentin for multiple sclerosis-induced neuropathic pain : a randomized controlled trial. Pain Med. 2015;16:149–59.PubMedCrossRef Turcotte D, Doupe M, Torabi M, et al. Nabilone as an adjunctive to gabapentin for multiple sclerosis-induced neuropathic pain : a randomized controlled trial. Pain Med. 2015;16:149–59.PubMedCrossRef
Zurück zum Zitat Ujváry I, Hanuš L. Human metabolites of cannabidiol: a review on their formation, biological activity, and relevance in therapy. Cannabis Cannabinoid Res. 2016;1(1):90–101.PubMedPubMedCentralCrossRef Ujváry I, Hanuš L. Human metabolites of cannabidiol: a review on their formation, biological activity, and relevance in therapy. Cannabis Cannabinoid Res. 2016;1(1):90–101.PubMedPubMedCentralCrossRef
Zurück zum Zitat Vandrey R, Herrmann ES, Mitchell JM, et al. Pharmacokinetic profile of oral cannabis in humans: blood and oral fluid disposition and relation to pharmacodynamic outcomes. J Anal Toxicol. 2017;41:83–99.PubMedPubMedCentralCrossRef Vandrey R, Herrmann ES, Mitchell JM, et al. Pharmacokinetic profile of oral cannabis in humans: blood and oral fluid disposition and relation to pharmacodynamic outcomes. J Anal Toxicol. 2017;41:83–99.PubMedPubMedCentralCrossRef
Zurück zum Zitat Ware MA, Wang T, Shapiro S, Collet J. Cannabis for the management of pain: assessment of safety study(COMPASS). J. Pain. 2015;16:1233–42.PubMedCrossRef Ware MA, Wang T, Shapiro S, Collet J. Cannabis for the management of pain: assessment of safety study(COMPASS). J. Pain. 2015;16:1233–42.PubMedCrossRef
Zurück zum Zitat Whiting PF, Wolff RF, Deshpande S, et al. Cannabinoids for medical use: a systematic review and meta-analysis. JAMA. 2015;313:2456–73.PubMedCrossRef Whiting PF, Wolff RF, Deshpande S, et al. Cannabinoids for medical use: a systematic review and meta-analysis. JAMA. 2015;313:2456–73.PubMedCrossRef
Zurück zum Zitat Yeon Kong T, Kim JH, Kyun Kim D, Suk LH. Synthetic cannabinoids are substrates and inhibitors of multiple drug-metabolizing enzymes. Arch Pharm Res. 2018;41:691–710.CrossRef Yeon Kong T, Kim JH, Kyun Kim D, Suk LH. Synthetic cannabinoids are substrates and inhibitors of multiple drug-metabolizing enzymes. Arch Pharm Res. 2018;41:691–710.CrossRef
Zurück zum Zitat Zendulka O, Dovrtělová G, Nosková K, et al. Cannabinoids and cytochrome P450 interactions. Curr Drug Metab. 2016;17:206–26.PubMedCrossRef Zendulka O, Dovrtělová G, Nosková K, et al. Cannabinoids and cytochrome P450 interactions. Curr Drug Metab. 2016;17:206–26.PubMedCrossRef
Zurück zum Zitat Zlas J, Stark H, Seligman J. Early medical use of cannabis. Nature. 1993;363:215.CrossRef Zlas J, Stark H, Seligman J. Early medical use of cannabis. Nature. 1993;363:215.CrossRef
Metadaten
Titel
Cannabis-based medicines and the perioperative physician
verfasst von
Patrick Tapley
Suzanne Kellett
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Perioperative Medicine / Ausgabe 1/2019
Elektronische ISSN: 2047-0525
DOI
https://doi.org/10.1186/s13741-019-0127-x

Weitere Artikel der Ausgabe 1/2019

Perioperative Medicine 1/2019 Zur Ausgabe

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.