Next Article in Journal
Generalized Pustular Psoriasis and Systemic Organ Dysfunctions
Previous Article in Journal
A Comprehensive Analysis of Non-Desmosomal Rare Genetic Variants in Arrhythmogenic Cardiomyopathy: Integrating in Padua Cohort Literature-Derived Data
Previous Article in Special Issue
Peripheral Endocannabinoid Components and Lipid Plasma Levels in Patients with Resistant Migraine and Co-Morbid Personality and Psychological Disorders: A Cross-Sectional Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cannabinoid Analgesia in Postoperative Pain Management: From Molecular Mechanisms to Clinical Reality

by
Antonio J. Carrascosa
1,
Francisco Navarrete
2,3,4,
Raquel Saldaña
1,
María S. García-Gutiérrez
2,3,4,
Belinda Montalbán
1,
Daniela Navarro
2,3,4,
Fernando M. Gómez-Guijarro
1,
Ani Gasparyan
2,3,4,
Elena Murcia-Sánchez
1,
Abraham B. Torregrosa
2,3,4,
Paloma Pérez-Doblado
1,
Luisa Gutiérrez
2,3,4 and
Jorge Manzanares
2,3,4,*
1
Servicio de Anestesiologia y Reanimación, Hospital Universitario 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain
2
Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain
3
Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
4
Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(11), 6268; https://doi.org/10.3390/ijms25116268
Submission received: 26 April 2024 / Revised: 26 May 2024 / Accepted: 28 May 2024 / Published: 6 June 2024

Abstract

:
Postoperative pain (POP) is a challenging clinical phenomenon that affects the majority of surgical patients and demands effective management to mitigate adverse outcomes such as persistent pain. The primary goal of POP management is to alleviate suffering and facilitate a seamless return to normal function for the patient. Despite compelling evidence of its drawbacks, opioid analgesia remains the basis of POP treatment. Novel therapeutic approaches rely on multimodal analgesia, integrating different pharmacological strategies to optimize efficacy while minimizing adverse effects. The recognition of the imperative role of the endocannabinoid system in pain regulation has prompted the investigation of cannabinoid compounds as a new therapeutic avenue. Cannabinoids may serve as adjuvants, enhancing the analgesic effects of other drugs and potentially replacing or at least reducing the dependence on other long-term analgesics in pain management. This narrative review succinctly summarizes pertinent information on the molecular mechanisms, clinical therapeutic benefits, and considerations associated with the plausible use of various cannabinoid compounds in treating POP. According to the available evidence, cannabinoid compounds modulate specific molecular mechanisms intimately involved in POP. However, only two of the eleven clinical trials that evaluated the efficacy of different cannabinoid interventions showed positive results.

1. Introduction

Pain is an unpleasant sensory and emotional experience associated with, or similar to, actual or potential tissue damage [1]. It can be classified into two main categories based on the neurophysiological mechanisms underlying its origin: nociceptive pain, which arises from the activation of nociceptors in response to tissue damage caused by physical or chemical agents like trauma, chemical burns, or surgical procedures [2], and neuropathic pain, which results from direct damage or dysfunction of the sensory nerves [3]. Postoperative pain (POP) is a unique entity that necessitates prompt and effective alleviation to minimize suffering, facilitate the healing process and rehabilitation, and prevent complications. It is not solely attributable to inflammation or isolated nerve injury; its pathophysiology is unique, with specific consequences [4,5]. As a result, the response to analgesic treatment differs from other pain models.
To date, opioids remain a mainstay of perioperative pain management, employed for sedation during general anesthesia, supplementation during regional anesthesia, and the treatment of acute postoperative pain [6,7,8]. However, despite their numerous benefits, opioids are associated with a well-documented adverse effect profile that can impede recovery and the resumption of daily activities. Moreover, the escalating rates of opioid consumption and misuse in the United States have reached epidemic proportions over the past decade [9,10]. Additionally, balancing adequate postoperative pain relief while mitigating the risk of overdose or relapse is challenging, particularly as a growing number of surgical patients exhibit opioid tolerance, such as those with chronic pain conditions [11]. Therefore, the effectiveness of opioids for pain management during the perioperative period is undergoing reevaluation [12].
The search for drugs that offer advantages over existing prescriptions is a fundamental goal of pharmacological research in managing POP. Thus, it is interesting to study families of analgesics with new mechanisms of action, high potency, and minimal undesirable effects. The incorporation of non-opioid adjuvant medications into a perioperative pain management plan can not only potentially enhance patient outcomes but also serve as a critical component in minimizing opioid utilization and potentially mitigating the downstream risk of opioid misuse and dependence.
In this context, there has been a renewed interest in cannabinoids, a class of compounds with a historical precedent for pain management dating back to the 19th century [13]. This resurgence can be attributed to the discovery of cannabinoid receptors [8,14] and endogenous substances that modulate these receptors [15,16], despite the inherent challenges in studying substances prone to abuse which are thus subject to stringent legislative control [17]. Arguments supporting their use include the following: (1) there is evidence of activation of the ECS after surgery [18,19]; (2) cannabinoids are analgesics per se; and (3) they are substances that can act as adjuvants to facilitate the analgesic action of other drugs. Nevertheless, despite experimental evidence demonstrating their antinociceptive efficacy, clinical outcomes have been constrained by the short duration of studies, small sample sizes, absence of control groups, and biases prevalent in most conducted studies.
This review examines the current scientific evidence regarding the use of cannabis for the treatment of acute POP. Its interest lies in the fact that the etiology and treatment of pain produced by surgery are different from those of other painful conditions. Effective and rapid pain management is crucial to minimizing patient suffering and mitigating the complex physiological stress response triggered by surgery.

2. Cannabinoids and Therapeutic Potential in Pain Relief

Cannabinoids are a group of chemical substances derived from the cannabis plant, the endogenous cannabinoid system, or synthetic production that bind to varying degrees to cannabinoid receptors, including cannabinoid receptor type 1 (CB1r) [15,20,21,22,23] and type 2 (CB2r) [15,24,25,26,27,28,29,30,31,32]. This group of substances is extensive and diverse and can be classified in several ways. According to their origin, we distinguish three types of cannabinoids (Figure 1): (1) phytocannabinoids (naturally derived from plants); (2) endocannabinoids (endogenous cannabinoids); and (3) synthetic cannabinoids (artificially produced phytocannabinoids).

2.1. Phytocannabinoids

Phytocannabinoids are compounds characterized by a carbocyclic structure whose central ring is usually tetrahydropyran and by two chiral centers [33]. These compounds are produced naturally in the trichomes of the hemp plant (Cannabis sativa) to protect it against pests and the effects of the environment [34,35]. This plant is a tall annual shrub that grows naturally in temperate and tropical regions. It has been extensively utilized for medicinal purposes for millennia in various parts of Asia, particularly in India and China [36,37,38]. The resin extracted from the plant is known as hashish, while the name “marijuana” is attributed to the preparation of dried leaves and flowers of the plant.
More than 100 phytocannabinoids have been described, including their acid and neutral forms, analogs, and other transformation products [33,39]. The primary cannabinoids are ∆9-tetrahydrocannabinol (delta-9-THC or THC), 8-tetrahydrocannabinol (8-THC), cannabidiol (CBD), and cannabinol (CBN). Additionally, the plant contains other cannabinoids such as cannabichromene (CBC), cannabicyclol (CBL), cannabigerol (CBG), cannabigerol monomethyl ether (CBGM), cannabielsoin (CBE), cannabinodiol (CBND), cannabitriol (CBT), dehydrocannabifuran, and cannabicitran. The presence and quantities of these cannabinoids vary depending on the specific variety of Cannabis sativa being assessed [40]. It is interesting to note that Cannabis sativa synthesizes phytocannabinoids exclusively in their non-psychoactive acidic forms. Notably, the carboxyl group attached to these precursors is unstable and readily decarboxylates, releasing CO2 under heat or light exposure. This decarboxylation process transforms acidic cannabinoids into active neutral forms [41,42]. Furthermore, the relative abundance of each phytocannabinoid within the plant is significantly influenced by several factors, including growing conditions and extraction methods [19]. Additionally, cannabis has a complex botanical composition that encompasses over 200 terpenes and terpenoids. These terpenoids possess diverse pharmacological properties and have been linked to various therapeutic effects [43].

Phytocannabinoids and Pain Relief

THC remains the most extensively researched phytocannabinoid due to its potent psychoactive properties [44] and well-documented antinociceptive effects [45]. However, at higher doses, THC can induce intoxication. Despite this limitation, clinical trials generally support the efficacy of THC in managing chronic pain [46,47,48].
There are conflicting opinions in the literature regarding the use of phytocannabinoids for pain relief: (1) cannabis has been employed for medicinal purposes for millennia and the combination of its phytocannabinoids is more effective than currently available cannabinoid drugs. In this regard, emerging evidence suggests that herbal cannabis exhibits analgesic effects in both nociceptive and neuropathic pain. Notably, at least five high-quality randomized controlled clinical trials (RCTs) have demonstrated the efficacy of smoked cannabis in achieving pain relief [49,50,51,52,53,54]. (2) There is accumulating evidence and ongoing research for addressing common symptoms and conditions linked to pain, such as spasticity associated with multiple sclerosis or stroke [55,56], anxiety and posttraumatic stress disorder [57], migraine [58], nausea and vomiting [59], cachexia, inflammatory bowel diseases [60], and sleep disturbance [61,62]. (3) Cannabis has little capacity to cause overdose and is associated with lower rates of addiction compared to opioid analgesics. (4) Cannabis cultivation and production are relatively cost-effective. In contrast, there are detractors to the use of phytocannabinoids based on the following: (a) Herbal cannabis presents a complex chemical composition with significant variability and incomplete characterization, posing challenges for standardized dosing and consistent prediction of effects. Notably, cannabinoids can exhibit a spectrum of actions, sometimes even opposing effects, depending on several factors. These factors include the specific compound under investigation, its enantiomeric form, the plant species utilized in the study, and the patient’s overall health status [63,64,65]. As a result, it fails to meet the FDA criteria for drug approval. (b) Widespread recreational cannabis use raises concerns for potential individual and public health risks [66,67].
Consequently, there is an inherent risk that the availability of cannabis as a medicinal product will lead to increased accessibility and associated damage. (c) Only a small number of patients can achieve satisfactory clinical management; the advocacy for medical cannabis forms part of a well-structured and funded strategy to legalize cannabis for general use. (d) Inhaled cannabis via combustion methods, such as smoking, may present health risks due to the generation of harmful by-products. Thus, although inhaled cannabinoids produce bronchodilation, it is essential to note that the combustion of cannabis generates harmful by-products, including carbon monoxide, bronchial irritants, and potential carcinogens—the tar in a cannabis cigarette contains even higher concentrations of benzanthracenes and benzopyrenes [68]. Furthermore, some studies indicate that smoking cannabis, due to its consumption through deep and prolonged inhalations without a filter and higher combustion temperature than tobacco, may result in a fivefold elevation in carboxyhemoglobin levels and a threefold increase in tar intake compared to tobacco cigarettes [69].

2.2. Endocannabinoids

Endocannabinoids (eCBs) are endogenous signaling molecules naturally produced by all vertebrate animals, including humans. These lipid mediators are predominantly within the central nervous system (CNS) [70,71,72]. Additionally, eCBs are found in cells of the immune and reproductive systems, highlighting their diverse physiological roles [73,74]. These substances, consisting essentially of fatty acids derived from arachidonic acid (AA) metabolism, are critical regulators of various physiological processes, particularly within the CNS. They act as vital stress response regulators, aiding in adaptation or habituation to stress, guarding against the onset of stress-related illnesses and dysfunctions, and ultimately, promoting survival. They achieve this by interacting with autonomic, endocrine, and immune processes and sensory signaling mechanisms [75,76] (Figure 2). In addition, eCBs have been implicated in many behavioral processes, including memory [77,78], emotional state [79,80], feeding [81], inflammation [82,83], hemodynamic response [84,85], energy metabolism [86,87], pregnancy [88,89], and nociception [90,91,92]. Moreover, they also modulate the proliferation, motility, adhesion, and apoptosis of cells [93,94].
Our understanding of the endocannabinoid system (ECS) has grown significantly since the discovery of the first eCBs, anandamide (N-arachidonoylethanolamine, AEA) [15] followed by 2-arachidonoyl glycerol (2-AG) [16], both considered the primary players. These lipid mediators derived from arachidonic acid (AA) have cannabis-like effects. Further exploration has revealed additional eCBs within the brain, including the ether-linked 2-arachidonoyl-glyceryl ether (noladin ether), the AA ethanolamine derivative virodhamine, and N-arachidonoyldopamine (NADA). Notably, NADA acts primarily as a transient receptor potential vanilloid type-1 (TRPV1) agonist but also exhibits some activity at the CB1r. Additionally, structurally related compounds such as N-acylethanolamines (e.g., N-oleoyl ethanolamine (OEA) and N-palmitoyl ethanolamine (PEA)) and 2-oleoylglycerols (e.g., 2-oleoyl-glycerol and 2-linoleoyl-glycerol) are widely distributed in both the CNS and periphery, forming part of the expanded ECS. Nevertheless, their endocannabinoid classification remains contentious due to their lack of affinity for CB1r and CB2r [95,96,97].
eCBs are synthesized on demand, meaning they are not stored pre-formed within cells for later release. Following their release, their biological effects are rapidly terminated by cellular uptake and/or subsequent enzymatic degradation [98]. However, accumulated evidence mainly from pharmacological studies strongly suggests that there must be a maintained cannabinoid tone with a continuous release of endogenous ligands [99]. It is worth mentioning that eCBs are synthesized from membrane precursors, and the degradation products of eCBs serve as precursors for eicosanoids. Consequently, eCB signaling is integrated into a lipid metabolism and signaling network. Therefore, altering the activity of enzymes involved in eCB synthesis and degradation may also affect other lipid signaling systems [100].
Similarly, the distinct distribution patterns of enzymes responsible for eCBs synthesis and degradation throughout the cell and its compartments suggest diverse functional roles for these molecules [101]. Moreover, AEA and 2-AG exhibit distinct pharmacological profiles, interacting with CB1r and CB2r and other receptors like TRPV1 and GABAA [102,103]. Additionally, endogenous peptides known as pepcans or hemopressins can influence biological processes by acting on CB1r and CB2 [104,105,106].

Involvement of eCBs in the Regulation of Pain

Endocannabinoids (eCBs) modulate pain perception through a dual mechanism. The first involves the activity-dependent phasic release of eCBs triggered by neuronal activity. The second involves a sustained endogenous eCB tone, elevated in pathophysiological conditions like inflammation [99,107].
The diversity of cannabinoid-mediated signaling, the ligand concentration, the presence of other cannabinoid ligand molecules, and the different distributions of metabolic enzymes influence the response to specific eCBs. In this regard, the pathological state and tissue type significantly affect the levels of eCBs and related compounds [108]. These variations likely arise from the disease-specific alterations in enzymes responsible for eCB metabolism. These enzymes exhibit distinct functions, leading to variable effects on the metabolism of different eCBs and related lipids within the same family. Consequently, the levels of fatty acid amide hydrolase (FAAH), cyclooxygenase (COX), and lipoxygenase (LOX) may vary depending on the pathological condition [100]. Molecular studies have demonstrated modulation of the endocannabinoid system (ECS) following spinal cord injury (SCI), with changes observed during both the acute and chronic phases [108,109]. Specifically, AEA is upregulated during the first week after injury. Similarly, alterations in eCBs have been observed in neuropathic pain across various regions of the pain pathways of ascending and descending pain pathways [110,111].
Notably, CB1r is abundantly expressed in neurons and oligodendrocytes, being the AEA/CB1r system critical for neuronal survival [108,109]. Shifting to the chronic injury phase, two to three weeks after injury, there is an increase in 2-AG, a molecule that can activate both CB1r and CB2r. Furthermore, alongside these endocannabinoid changes, the chronic injury phase also witnesses increased CB2 receptor levels in macrophages and astrocyte-like cells [108,112]. CB2r was first considered a peripheral restricted cannabinoid receptor that could be present in the CNS only under certain pathological conditions [113]. However, since the publication of a study identifying the expression of CB2r in neurons of the brainstem of mice, rats, and ferrets under normal physiological conditions [114], much attention has been paid to the functional role it might play, particularly concerning neuroinflammatory processes [22,115].
At present, the diversity of ECS signaling molecules and their interactions with various receptors, together with the signaling complexity of receptor systems, makes the pharmacological intervention of the ECS a challenging task, containing a considerable degree of unpredictability in the outcome of the biological effects in a whole organism.

2.3. Synthetic Cannabinoids

Synthetic cannabinoids are molecules developed in laboratories that interact with cannabinoid receptors, thereby achieving a therapeutic effect [116]. Among these, the phytocannabinoids THC and CBD are available as synthetic compounds for a range of indications, as outlined below. Various pharmaceutical products are available in tablets, capsules, and sprays, which can only be obtained via prescription [117].
Some examples of synthetic cannabinoids are the following: (1) nabilone (Cesamet® or Canemes®) is a synthetic analog of delta-9-THC with a different molecular structure than THC, which gives it a slightly different interaction with cannabinoid receptors. It is marketed in capsules for oral administration. Cesamet is manufactured by Meda Pharmaceuticals Inc.(Somerset, NJ, USA) and Canemes by AOP Orphan Pharmaceuticals AG (Canonsburg, PA, USA). Its use is approved for the treatment of nausea and vomiting caused by chemotherapy as well as for pain control [118,119,120,121,122]. (2) Dronabinol (Marinol®, Adversa®, Syndros®, and Reduvo®) is an oral capsule or oral solution containing a synthetic analog of delta-9-THC prepared in 2.5 mg, 5 mg, or 10 mg. Marinol is produced by AbbVie Inc. (North Chicago, IL, USA) and Syndros by Insys Therapeutics Inc. (Chandler, AZ, USA). Its use has been approved for treating nausea, vomiting, loss of appetite, and weight loss [123,124]. (3) CBD (Epidiolex®) is a drug marketed as a viscous oral solution containing CBD as the main active ingredient (100 mg per mL). This medication is indicated for the treatment of seizures in patients with Lennox–Gastaut syndrome, Dravet syndrome, or tuberous sclerosis complex [125,126]. (4) Nabiximol (Sativex®) is an oromucosal spray containing approximately a 1:1 combination of THC and CBD extracted from the Cannabis plant, delivering 2.7 milligrams of THC and 2.5 milligrams of CBD per dose, manufactured by GW Pharmaceuticals Plc. This oromucosal spray is primarily indicated for managing spasticity associated with multiple sclerosis, particularly in patients who have not responded adequately to other therapies [127,128]. However, there is literature on treating neuropathic pain of different origins [129]. (5) Rimonabant (Acomplia™ and Zimulti™) is a synthetic cannabinoid compound characterized by a CB1r inverse agonism/antagonism mechanism of action and was designed to decrease appetite and promote weight loss in obese patients [130,131]. Despite its effectiveness in achieving the purposes for which it was intended, rimonabant was discontinued, and its sale is currently banned due to its association with depression and suicide attempts.

3. Molecular Mechanisms Underlying Analgesic Effects of Cannabinoids

Pain arises from a complex, multi-layered pathway within the nervous system. Sensory information travels from the site of injury through the dorsal horn of the spinal cord and relays to structures of the brainstem and diencephalon, including the thalamus, periaqueductal gray, parabrachial nucleus, reticular formation, amygdala, and hypothalamus, among others [132,133]. This intricate network integrates signals from both tissue damage (nociceptive pain) and dysfunctional brain processing (neuropathic pain) to create the final sensation of pain [134,135,136]. Likewise, the body activates the endocrine and immune systems to counter aversive stimuli and promote healing [137,138,139]. These integrated physiological processes culminate in a defensive biological response to injury [140,141].
Consequently, exposure to a repeated noxious stimulus, such as during tissue damage or exposure to too intense stimuli [142], triggers a neuroinflammatory phenomenon associated with an increased response of nociceptors (peripheral sensitization) [143] and increased excitability of the neurons in the spinal cord (central sensitization) [144,145,146] and in the cortical area (cortical sensitization) [147,148]. This response is mediated by several substances acting through cellular and molecular mechanisms. These mechanisms include the following: (1) significant cellular changes that result in ectopic and/or spontaneous nerve discharges, peripheral, and central hyperexcitability and phenotypic changes in conduction pathways, neurodegeneration, and reorganization of cell morphology; (2) molecular changes, highlighting the accumulation and increased expression of sodium channels in the periphery, increased activity of glutamate receptors, particularly the NMDA receptor, reduced GABAergic activity, changes in calcium penetration into neurons, and increased cytokines, chemotactic factors, growth factors, and ATP; and (3) changes in the structural and functional activity of neurons at the central and peripheral levels as well as neuroimmune interactions, which become more prominent during inflammatory reactions [135,149,150,151,152].
In the case of POP, current research on persistent pain management suggests that it may represent a distinct and common subtype of acute pain, differing from pain arising from antigens, chemical nociception, or neuropathic origin. This distinction in pathophysiology underscores the potential need for tailored treatment approaches for POP compared to other pain conditions. For instance, while spinal N-methyl-D-aspartate (NMDA) receptor antagonists effectively alleviate hypersensitivity in various pain models, they often lack efficacy in managing POP [153]. This evidence shows distinct molecular mechanisms at play in different types of pain. Conversely, intrathecal administration of non-NMDA receptor antagonists [154], NK-1 receptor antagonists [155], and cyclooxygenase-1 inhibitors [156] demonstrates promise for treating POP with minimal effects on nerve injury models.
Similarly, the descending facilitatory pathway originating from the rostral ventromedial medulla, known to contribute to behavioral hypersensitivity in inflammatory and neuropathic pain models, appears irrelevant in postincisional pain [157,158]. Interestingly, glial cell activation emerges as a potential factor in the development and persistence of pain after peripheral nerve injury [159]. This highlights the potential of targeting glial and neuronal–glial interactions for novel pain management strategies [160,161].
In this context, cannabinoids exert their antinociceptive effect through interaction with the ECS, a network of lipid-based signaling molecules that includes two G protein-coupled receptors (CB1r and CB2r), eCBs (AEA and 2-AG) which interact with these receptors [99,162], and two significant enzymes regulating the metabolism of eCBs (FAAH, which predominantly degrades AEA, and monoacylglycerol lipase (MAGL), which predominately degrades 2-AG) [110]. As represented in Figure 3, ECS components are present in neurons, astrocytes, oligodendrocytes, and microglia. CB1 receptors are predominantly localized to the plasma membrane of neurons, with a smaller population residing in the mitochondria (mCB1r). Presynaptic CB1 receptors regulate neurotransmitter release via a retrograde signaling mechanism. An increase in postsynaptic Ca2+ levels prompts the synthesis of eCBs within the postsynaptic neuron. Subsequently, these eCBs travel back (retrogradely) to the presynaptic terminal, activating CB1 receptors and inhibiting neurotransmitter release.
Therefore, the ECS is crucial in pain management as a critical modulator of synaptic function within the central nervous system (CNS) [99]. This extends their influence beyond pain perception, regulating various neural functions and behaviors within the immune and endocrine systems [75,76]. In this sense, glial cells influenced by the ECS release a spectrum of signaling molecules (chemokines and cytokines) within the CNS. This bidirectional communication between the nervous and immune systems facilitates adaptation or habituation to stress, protects against stress-induced pathologies, and ultimately promotes survival [163,164,165,166]. Additionally, the ECS exhibits potent antinociceptive and anti-inflammatory activities through interactions with diverse molecular targets, as demonstrated in in vivo studies (see [167,168] for reviews). Moreover, eCBs also significantly regulate hormone production, influencing hypothalamic-releasing factors, pituitary hormones, and peripheral steroidogenesis (see [169] for a review).
Extensive research is ongoing to elucidate the mechanisms and sites of action responsible for cannabinoid analgesia. CB1r and CB2r are prime candidates, with evidence pointing toward their involvement at the peripheral, spinal, and supraspinal levels [115,170]. Like many G protein-coupled receptors (GPCRs), these receptors exhibit remarkable versatility and adaptability. This is evident in their flexible ligand binding, diverse intracellular signaling pathways, ability to form homodimers and heterodimers, and varied subcellular localization throughout the body [171]. Similarly, evidence suggests that other receptors contribute to ECS signaling [171]. These include orphan G protein-coupled receptors GPR119 [172] and GPR55 [173,174,175,176], as well as peroxisome proliferator-activated receptors (PPARs) [8,175,177,178,179,180,181]. Moreover, cannabinoids can additionally activate ion channels, particularly TRPV1 receptors. The expression of TRPV1 on sensory nerves is known to mediate inflammatory pain, and TRPV1/CB1 receptor co-expression is enhanced in inflamed tissue [182,183].
Consequently, activating different cannabinoid receptor types and locations leads to varied responses to noxious stimuli [184]. Thus, metabotropic receptors, e.g., CB1r, CB2r, GPR55, and GPR119, are associated with a slower reaction but with a longer-lasting and more far-reaching action by allowing the opening of different channels for a longer time since second messengers can act in cascade (generating the activation of other proteins and substances). Typical intracellular events mediated by Gi/o proteins coupled with CB1r activation include inhibiting most voltage-dependent calcium channels and increasing potassium conductance [185,186,187,188,189,190]. They also stimulate the mitogen-activated protein kinase (MAPK) pathway to regulate proliferative and differentiative phenomena [72]. Both phenomena contribute to reducing neuronal excitability and suppressing neurotransmitter release. In this sense, activation of the CB1r inhibits the release of GABA or glutamate and neuropeptide by nerve terminals [191,192,193,194]. In the case of the CB2r, the transduction mechanisms coupled to the stimulus of this receptor are similar to those of the CB1r. Therefore, activation of the CB2r leads to inhibitory effects on the adenylate cyclase/AMPc system, as well as stimulation of the mitogen-activated kinase pathway (ERK, JNK, and p38) and the PI3KAkt pathway [195], pathways that are closely related to the processes of cell proliferation and survival and are therefore associated with their modification (Figure 4).
In contrast, interaction with ionotropic receptors such as TRPV1 generates rapid, short-lived responses. This suggests that cannabinoid modulation of inward currents through these receptors (ICRs) could activate sensory neurons, potentially leading to nociception (pain perception) [196,197,198]. However, behavioral studies contradict the potential nociceptive effects suggested by ionotropic receptor activation [196,199,200]. These studies report cannabinoid-induced antihyperalgesia and antinociception, signifying pain reduction at the periphery [168,201,202]. One possible explanation lies in the type of cannabinoid action on these receptors. Unlike full agonists, cannabinoids may only partially activate ICRs, which is insufficient to trigger nociceptor excitation [199,203,204]. Several studies show that cannabinoids evoke slow, small, inward currents and calcium accumulation, potentially falling below the threshold for pain activation [199,203,204]. Moreover, slow depolarization of nociceptor membrane potentials might lead to the inactivation of voltage-gated channels, which inhibits the generation of action potentials [205].
Notably, cannabinoids also can modulate the release of mediators involved in pain and inflammation. For instance, activation of CB1 receptors on neuronal presynapses reduces cellular activity and consequently diminishes the release of neurotransmitters like dopamine, noradrenaline, serotonin, GABA, and glutamate retrogradely [206,207,208,209,210]. Additionally, cannabinoids influence other biological systems through the control that second messengers may undergo and even through allosteric changes secondary to the insertion of cannabinoids in the cell membrane [211]. Thus, although classic descriptions of eCBs focus on interactions between the nervous and immune systems, recent research emphasizes their regulatory influence on endocrine function. This extends to various hormonal axes, including those controlling gonadal steroid, growth hormone, prolactin, thyroid hormone, and HPA axis activity [212,213].
Furthermore, although eCBs are known for localized action due to their rapid breakdown [164], the ability of cannabinoids to affect pain perception has supraspinal, spinal, and peripheral components within its general strategy of action of a local modulatory system [214]. Direct evidence for supraspinal cannabinoid antinociception has been substantiated by administration of cannabinoid agonists intracerebroventricularly and/or into encephalic structures at minimal doses [215,216,217,218], administration of supraspinally administered cannabinoid antagonists inducing pain [219,220], and electrical stimulation of the rat periaqueductal gray (PAG), as well as formalin injection into the hind paw and increased AEA release in the PAG as determined by microdialysis coupled to liquid chromatography/mass spectrometry [221], respectively. On the other hand, evidence of spinal cannabinoid antinociceptive effects has been obtained through behavioral, neurochemical, and electrophysiological studies using spinal cannabinoid agonists [222,223]. In this case, cannabinoids can act on spinal CB1rs to inhibit capsaicin-sensitive fibers in the dorsal horn and reduce the firing of wide dynamic range (WDR) neurons in response to noxious stimuli [222,224]. Additionally, activation of the spinal CB1r can decrease NMDA receptor activation by potentially inhibiting glutamate release into the spinal cord [225], and activation of CB2r suppresses activity in spinal nociceptive neurons, particularly under conditions of sensitization, and regulates the immune response, favoring the neuroprotective actions of neuroglia [226,227].
Furthermore, cannabinoids may modulate spinal noradrenergic and opioid systems [228,229]. Concerning the peripheral component, the antinociceptive action of CBr has been demonstrated in different pain models by peripherally administering CB1r [230] and CB2r [231,232,233] agonists. Likewise, it has been confirmed that endocannabinoid substances such as AEA also activate TRPV1 receptors [234]. For a more detailed review of the mechanisms involved in the analgesic effects of cannabinoids, please see [235,236].
In summary, the interaction of CBr agonists (either endogenous or exogenous) leads to a reduction in neuronal activity secondary to inhibition of bioelectrical activity by lowering the intracellular level of second messengers and loss of the ability to release their specific neurotransmitters (whatever they activate or inhibit). The cellular consequences are short-term modification of the permeability of membrane ion channels (mainly for K+ and Ca2+ [237,238]), decreasing neuronal excitability and long-term changes in gene expression that result in phenomena such as brain plasticity, dependence, and transformation of an acute response into a long-term adaptation or memory [189,239,240,241,242]. The overall result is decreased pain perception by modulating the nociceptive impulse at different levels and activating a descending inhibitory system acting on the spinal cord [92,243,244,245]. Multiple factors, such as the diversity of cannabinoid-mediated signaling [246,247,248,249,250], the ligand concentration [247,251,252], the presence of other cannabinoid ligand molecules [252], the exact localization of the cannabinoid receptors [184], and the different distributions of metabolic enzymes influence [110,253] the response to a specific cannabinoid.

4. Why Consider the Therapeutic Potential of Cannabinoids in Postoperative Pain Relief?

4.1. Activation of the ECS after Surgery

Surgical procedures invariably induce peripheral tissue trauma, initiating a well-orchestrated nociceptive response. The specific tissues implicated vary based on the surgical approach, encompassing skin, fascia, muscle, vasculature, viscera, and potentially neural structures. This damage directly activates nociceptive nerve fibers, particularly unmyelinated C-fibers, contributing to postsurgical pain’s dull, aching quality. Furthermore, tissue injury triggers a robust inflammatory response, a coordinated endeavor involving diverse cell types and releasing many mediators. These mediators are essential to clearing cellular debris, combating potential pathogens, and facilitating wound healing through scar tissue formation. However, the inflammatory process can also contribute to pain by sensitizing nearby nociceptors [254].
A significant challenge in POP arises from iatrogenic nerve injury. Specific surgical interventions, such as limb amputation or inguinal hernia repair, carry a higher risk of peripheral nerve damage [255]. This nerve injury can lead to the development of neuropathic pain [256], a chronic pain state characterized by burning, electrical, or dysesthetic sensations. Neuropathic pain is a prevalent condition, affecting at least 8% of individuals with chronic pain and significantly contributing to chronic postoperative pain (CPOP) [257,258]. It is noteworthy, however, that not all nerve injuries culminate in chronic pain; for instance, only approximately 80% of amputees develop CPOP [149,259].
In this context, preclinical POP studies have shown evidence of a crucial role for the ECS in resolving pain after surgery and preventing its transition into a chronic state by limiting pro-inflammatory responses within spinal cord glial cells [18,19]. Targeting eCBs offers several advantages: (1) localized and on-demand synthesis whereby eCBs are produced at the site of action, minimizing side effects associated with widespread cannabinoid receptor activation [247]; (2) endogenous production whereby the body naturally produces eCBs and the enzymes responsible for its breakdown, resulting in a shorter half-life and potentially lower toxicity compared to synthetic drug; and (3) synergy with existing treatments whereby the ECS can enhance the pain-relieving effects of common nonsteroidal anti-inflammatory drugs (NSAIDs), potentially improving pain management strategies [260].
However, despite evidence supporting eCBs’ role in pain relief [19,261,262,263,264] and the effectiveness of inhibiting FAAH and MAGL (enzymes degrading eCBs) in reducing pain [265], research also reveals complexities. Studies have shown increases and decreases in eCB levels depending on the pain-transmitting tissues analyzed [266,267]. Additionally, diverse studies suggest that blocking cannabinoids CB1r and CB2r can produce pain-relieving and anti-inflammatory effects [268,269,270,271]. This indicates that endocannabinoids can act as pro- and anti-inflammatory mediators [270,272], potentially making ECS activation detrimental in certain situations.
The current understanding suggests that the initial eCBs response to acute pain helps restore balance within the body, with precise timing and location being crucial. However, chronic pain states can dysregulate this system, leading to prolonged or inappropriate endocannabinoid activity. This complexity makes predicting the effectiveness of ECS-targeting drugs challenging for specific conditions.
At the clinical level, limited research exists. One study measured various endocannabinoid mediators in plasma, cerebrospinal fluid, and synovial fluid, correlating them with pain levels and opioid use after surgery [273]. The study found a significant correlation between synovial and cerebrospinal fluid 2-AG levels and both pain scores and opioid consumption. However, the potential confounding factor of pre-existing osteoarthritis and chronic pain in some participants needs to be addressed in future studies.

4.2. Cannabinoids Themselves Possess Analgesic Properties

The link between cannabinoid receptors and pain pathways has involved animal studies assessing cannabinoid antinociception [92,274]. Small-molecule CB1r and CB2r agonists and FAAH inhibitors were the most frequently evaluated. Preclinical data obtained with cannabinoid agonists after systemic [230,275] and/or perimedullary [225,230,276] administration in pain models of excess nociceptive, visceral, and neuropathic pain models [214,274,277] concluded that the antinociceptive effect is similar to opioids [278,279,280,281] and that it varies according to the substance chosen, the dose administered (a significant dose-dependent correlation exists between the administered cannabinoid dosage and the degree of antinociception observed) [282,283], and the route of administration (the doses required after administration by the perimedullary route are much lower and the duration of the effects longer than those used to achieve the same result by non-perimedullary routes) [229,284]. They also highlighted the greater antinociceptive power in inflammatory and neuropathic pain models [285,286,287,288]. In all cases, cannabinoids primarily act by inhibiting or releasing a series of modulators from neurons and/or non-neuronal tissues. This targeted action in inflamed tissues, where these modulators are present in relatively high amounts, is hypothesized to lead to a faster and more intense antinociceptive effect [83,277].
Furthermore, cannabinoids can act on various antinociceptive mechanisms, including the modulation of enzymes responsible for generating and releasing inflammatory agents and enhancing the abundance and diversity of cannabinoid receptors [274]. Regarding efficacy in neuropathic pain [289,290], it is relevant to highlight that the central cannabinoid CB2r is required for an anti-allodynic effect [22]. Cannabinoids also activate and desensitize another significant player in neuropathic pain (i.e., TRPV1) [291,292,293]. They reduce microglia and astrocyte activation as a mechanism related to the onset and maintenance of hypersensitivity in neuropathic pain [18,19].
However, human studies have yielded partially supportive findings from animal research regarding the use of cannabinoids for pain management [294,295]. Despite a long history of cannabis use for pain management [296], clinical trials have revealed limited efficacy for acute pain and even potential enhancement of specific pain responses [74,274,297,298,299,300,301,302]. Conversely, moderate-quality evidence suggests a small effect for chronic, non-cancer-related pain management with cannabinoids up to 6 months, potentially due to their influence on neuroplastic changes [303,304,305,306,307]. Therefore, based on the available literature, it is challenging to recommend cannabinoids as a general alternative to the currently marketed analgesic for pain management [302,308].
In this scenario, the complex nature of POP demands the development of more effective and better-tolerated therapeutic approaches. These options should target nociceptive and neuropathic pain pathways to relieve pain sensitization [140,309]. Ultimately, this could mitigate the reliance on opioid medications during POP and minimize the risk of long-term pain complications [11,310,311,312,313,314]. It is worth noting that the activation of the μ-opioid receptor is linked to numerous adverse effects, including respiratory depression [315,316], postoperative nausea and/or vomiting [317,318], constipation [319,320,321], urinary retention [318,322], mental clouding and somnolence [323,324], tolerance [325,326,327,328,329,330], hyperalgesia [313,331,332,333,334], dependence, and addiction [335,336,337,338,339]. Among these, a pivotal strategy is multimodal analgesia [340,341]. This involves combining various medications with different mechanisms of action to address pain from multiple angles. The aim is to minimize side effects from individual drugs and target different pain receptors for optimal pain control and improved recovery. Prioritizing non-opioid medications as the initial treatment for POP offers a safer and potentially more effective approach. These medications often have a wider therapeutic window, implying the dosage range for effective pain relief with minimal side effects.
To date, cannabinoids are emerging as promising candidates to replace or reduce reliance on other long-term pain medications. This potential stems from their ability to modulate the ECS, which in turn regulates the release of various neurotransmitters critical for pain perception, such as glutamate, GABA, serotonin, acetylcholine, dopamine, and norepinephrine [342,343,344]. However, research on the synergistic effects between existing analgesics and cannabinoid receptor agonists is limited. Nevertheless, several combinations promise enhanced pain management with reduced side effects. These include combinations with opioids, nonsteroidal anti-inflammatory drugs (NSAIDs), paracetamols, local anesthetics, and α2-adrenergic receptor agonists.

4.3. Opioids—Cannabinoids

The potential of cannabinoids to decrease opioid dosages and prolong the duration of adequate analgesia justifies further research. This is particularly intriguing considering their shared distribution within the descending pain inhibitory pathway and potential complementary cellular mechanisms of action compared to opioids [345] interacting synergistically in attenuating pain [346]. Thus, eCBs are produced and released within active neural circuits, where they play a critical role in mediating an adaptive response to mitigate pain and inflammation following injury and stress [247,347]. Pieces of evidence to date indicate that an ECS physiological tone mediates this regulation. In addition, ECS tone changes under pathophysiological situations such as inflammation, acting synergistically with the opioid system through different mechanisms [348,349]. A key consideration is the direct interaction between CB1r and mu-opioid receptors as functional heterodimers within the same neuron, as demonstrated when co-expressed [350]. Furthermore, cannabinoid administration can stimulate the synthesis and release of endogenous opioid peptides in the central nervous system and periphery [351].
While these properties imply a potential synergistic interaction between opioids and cannabinoids, the situation is not entirely straightforward due to the complexities of cannabinoid pharmacology. For example, the activation level of cannabinoid receptors (both basal and stimulated) by different agonists significantly influences the observed effects [352,353]. Moreover, partial agonists such as anandamide can act antagonistically in the presence of more efficacious agonists [354].
At present, preclinical studies support opioid sparing when co-administered systemically [355,356,357,358,359], intradurally, and/or intracerebroventricularly [229,360], or in a combination of routes [361,362].
However, clinical trials investigating nabilone and dronabinol in acute POP revealed no discernible benefits regarding opioid dose requirements or analgesic outcomes [363,364].

4.4. NSAIDs—Cannabinoids

NSAIDs primarily exert their antinociceptive effect by inhibiting the production of prostaglandins through COX enzyme blockade. COX-1 is constitutively expressed, while COX-2 is induced explicitly during inflammation. Notably, evidence suggests that NSAIDs may have additional pain-relieving mechanisms [365,366,367,368,369]. This has sparked research into potential interactions between cannabinoid agonists and NSAIDs for managing pain and inflammation. This interest stems from the convergence of pathways between endogenous cannabinoid receptor ligands and prostaglandins (molecules derived from arachidonic acid) [370]. Furthermore, evidence from cannabis signaling and the accumulation of arachidonic acid in brain slices exposed to cannabis derivatives supports this exploration [371].
Preclinical research suggests that eCBs and synthetic cannabinoids exhibit synergistic antinociceptive effects when combined with common NSAIDs [372,373,374,375,376]. This finding holds promise for the development of novel pain management strategies. However, further investigation is necessary to determine optimal dosing, safety profiles, and administration protocols for these combinations.
To our knowledge, there have been no clinical studies evaluating the analgesic efficacy of the combination of cannabinoids with NSAIDs.

4.5. Paracetamol—Cannabinoids

Several studies have revealed a surprising link between acetaminophen, the most commonly used pain medication, and the endocannabinoid system [377,378]. Research shows that blocking CB1r reduces the antinociceptive effects of paracetamol [379,380,381]. This effect is also observed in CB1 receptor-deficient (CB1–/–) mice [377,378] and those lacking FAAH (FAAH–/– mice) [382]. The link to the ECS becomes more evident when considering that acetaminophen can transform into N-arachidonoyl-phenolamine (AM404). This bioactive compound activates cannabinoid receptors and prevents the breakdown of natural pain-relieving endocannabinoids [383].
To our knowledge, there have been no clinical studies evaluating the analgesic efficacy of the combination of cannabinoids with paracetamol.

4.6. Local Anesthetics—Cannabinoids

Because endocannabinoid receptors are present in peripheral sensory afferents and spinal cord neurons [384,385,386,387], another strategy could be to develop synergistic interactions between cannabinoids and local anesthetics. Although the mechanisms for cannabinoid-induced antinociception are still unclear, there is literature that confirms a synergistic effect in experimental models [388].
To our knowledge, no clinical studies have evaluated the analgesic efficacy of combining cannabinoids with local anesthetics.

4.7. α2-Adrenergic Receptors—Cannabinoids

Alpha-2 agonists offer significant pain relief with the potential to decrease opioid consumption by attenuating nociceptive signaling throughout the nervous system, from peripheral sites to the brain [389,390,391]. Interestingly, they share similarities with CB1r agonists. Both belong to the G protein-coupled receptor family and can work together through various signaling pathways or by inhibiting adenylyl cyclase, resulting in decreased levels of cAMP and reduced activity of Ca2+ and K+ channels [392]. Synergistic analgesia can be achieved when a CB1r agonist is combined with an alpha-2 receptor agonist (such as clonidine, tizanidine, or guanfacine). Additionally, modulating the noradrenergic system, which regulates the activity of the hypothalamic–pituitary–adrenal axis (HPA), can be significant in treating anxiety and opioid withdrawal [393].
To our knowledge, no clinical studies have evaluated the analgesic efficacy of combining cannabinoids with alpha-2 agonists.

5. Cautions and Limitations of Using Cannabinoids for Postoperative Pain Relief

5.1. Which Product Is Suitable for Postoperative Pain Control?

The substantial heterogeneity among studies examining cannabinoids for POP pain management poses a significant challenge. These studies often utilize different cannabinoid medications with diverse mechanisms of action and varying activity levels of CB1r and CB2r, making it challenging to identify the most effective option. For example, THC [297,364,394], levonantradol [395,396], nabilone [363,397], and dronabinol [398] were used in POP. Additionally, CBD products have been employed in POP studies [399,400,401]. Two research articles used investigational compounds under development [402,403]. In this regard, it is not clear whether CBD is a better or worse analgesic than THC [404]. Nonetheless, patients typically perceive marijuana as potentially at least somewhat effective for pain management and are often open to using cannabinoid compounds for this purpose if recommended by a physician [405]. It is also known that nausea responds to THC, while anxiety responds better to CBD [406].
To date, unlike recreational users who prioritize psychoactive effects, medical cannabis patients often seek CBD-rich chemovars (strains) with minimal THC. These chemovars offer the potential for greater symptom control, improved functionality, and enhanced quality of life while minimizing unwanted side effects [407].
In the absence of evidence or clinical guidelines based on rigorous studies, it is advisable to use a cannabinoid product by assessing the effects obtained. A recommended strategy for cannabis initiation is “start low, go slow, and stay low”.

5.2. Pharmacokinetic Considerations and Routes of Administration of Cannabinoids in Postoperative Pain Relief

Pharmacokinetics, which involves a drug’s absorption, distribution, metabolism, and elimination, significantly influences its onset and duration of action. These are especially important for cannabinoids. Factors such as the route of administration—currently, cannabinoids have been marketed for systemic and topical use (Table 1)—and pharmacokinetic profile jointly dictate the clinical effects [408,409,410,411].
Absorption. Due to their limited aqueous solubility and lipophilic character, cannabinoids display significant variability in their effects depending on the chosen route of administration.
Pulmonary route: Inhaling cannabinoids brings on effects quickly, within 15 min, then levels off for 2–4 h before slowly wearing off. Their bioavailability varies considerably (between 10% and 85%), owing to various factors such as individual differences in inhalation techniques (number of puffs, duration and interval of puffs, breath hold time, and depth of inhalation), the device used, the size of inhaled particles, the temperature of the vaporizer, and the site of deposition within the respiratory system [410,411,412].
Oral route: THC and CBD formulations have a low bioavailability of around 6% due to their lipophilic structures, variable gut absorption, and extensive hepatic first-pass metabolism. Plasma concentrations for therapeutic effects remain within range for 2 to 6 h [409,411,413]. However, blood concentrations only reach 25–30% of those achieved through smoking the same dose. This is because first-pass metabolism by the liver reduces THC reaching circulation, although the resulting metabolite 11-hydroxy delta-9THC retains some psychoactive effects. The onset of the effect is delayed (0.5–2 h) and may be prolonged by continued slow absorption from the gut [414]. Notably, cannabinoids are best absorbed with fat, oils, or polar solvents like ethanol. Newer technologies, such as using nano- or ionized particles or incorporating omega fats into carrier oils, suggest a potential for increased absorption [415]. Additionally, the design of new water-soluble cannabinoid agonists opens up new possibilities for improved bioavailability [416].
Mucosal-related pathways: The sublingual and buccal regions of the oral cavity are lined with a non-keratinized, stratified, squamous epithelium. This specialized tissue is a selective barrier, allowing certain substances to pass through [417]. In this case, the formulation of THC and CBD (Sativex®) facilitates rapid absorptions and bypasses hepatic first-pass metabolism, resulting in higher plasma levels achieved through oral administration but lower than through inhalation administration [418]. However, sublingual and buccal routes have some drawbacks compared to oral administration. These include the following: (1) a shorter duration of action whereby the pain relief effects wear off quicker; (2) frequent dosing whereby maintaining stable pain control requires repeated administrations and this can increase the risk of side effects; and (3) potential for adverse reactions whereby rapid administration can lead to high drug concentrations in the bloodstream, raising the chance of severe reactions. Advances in nanoparticulate drug delivery represent a line of research aimed at enhancing the retention and absorption of drugs in the buccal and sublingual regions [419].
Skin-related pathways: By bypassing first-pass metabolism, transdermal cannabinoids can potentially offer a more consistent and controlled release of cannabinoids into the body compared to edibles [420]. However, their water-insoluble nature requires permeation enhancers to ensure they reach the bloodstream effectively [421,422]. Studies indicate that CBD exhibits ten times greater permeability in transcutaneous administration compared to THC. This finding suggests that CBD possesses a more polar structure than THC [410,423,424].
Intravenous route: Intravenous administration of cannabinoids presents a unique challenge due to their poor water solubility. Nonetheless, it remains the most reliable method for the administration of synthetic cannabinoids. This route bypasses first-pass metabolism, ensuring minimal variability in plasma concentrations and consistent results across patients. The resulting plasma profile following an intravenous dose closely mirrors that observed after inhalation. However, rapid redistribution within the body leads to a swift decline in plasma levels. Subsequently, drug metabolism contributes to a slower, sustained decrease in concentration [425]. It is important to note that the existing literature regarding the use of intravenous cannabinoids for postoperative pain management remains limited. Additionally, the research conducted thus far has primarily focused on the effects of tetrahydrocannabinol (THC) [426].
Distribution. Upon absorption, THC and other cannabinoids rapidly distribute to various tissues at rates influenced by blood flow [214,409,427]. Because they are extremely lipid-soluble, cannabinoids tend to accumulate in adipose tissues, reaching peak concentrations within 4–5 days. Subsequently, they undergo slow release into other body compartments, including the brain. Due to sequestration in fat, THC has a tissue elimination half-life of approximately 7 days, with complete elimination of a single dose potentially taking up to 30 days [428]. This accumulation phenomenon suggests that with repeated dosage, cannabinoids can persist in the body and continue to reach the brain. In the brain, THC and other cannabinoids exhibit differential distribution, with high concentrations observed in the neocortical, limbic, sensory, and motor areas. Notably, the volumes of distribution (Vd) for CBD and THC are notably high. Specifically, the volume of distribution at beta phase (Vdβ) is approximately 32 L/kg following intravenous administration for CBD [429], and the volume of distribution at steady state (Vdss) is approximately 3.4 L/kg following inhaled administration for THC [411].
Metabolism. The metabolism of THC primarily occurs in the liver, predominantly through cytochrome P450 (CYP450) isozymes such as CYP2C9, CYP2C19, and CYP3A4. THC is primarily metabolized into 11-hydroxy-THC (11-OH-THC) and 11-carboxy-THC (11-COOH-THC), which undergo glucuronidation and are subsequently excreted in the feces and urine [410]. Additionally, metabolism occurs in extrahepatic tissues expressing CYP450, such as the small intestine and brain [411]. It is worth noting that the metabolite 11-OH-THC is found in higher quantities in the brain compared to the unmetabolized THC compound, suggesting a potential role for 11-OH-THC in the effects experienced with THC [424,430,431]. The increased uptake of 11-OHTHC in the brain may be attributed to its lower plasma protein binding or the hydroxylated metabolite’s ability to pass through the blood–brain barrier [411]. In the case of CBD, it undergoes extensive hepatic metabolism, primarily by the cytochrome P450 (CYP) isozymes CYP2C19 and CYP3A4. Additional contribution comes from CYP1A1, CYP1A2, CYP2C9, and CYP2D6 [432]. Following hydroxylation to 7-OH-CBD, further hepatic metabolism primarily leads to fecal excretion, with a minor contribution to the urinary excretion of these metabolites. However, the pharmacological activity of CBD metabolites in humans remains largely unknown [433].
Elimination. The elimination half-life of THC demonstrates biphasic characteristics. A population pharmacokinetic model estimates a rapid initial half-life of approximately 6 min, followed by a slower terminal half-life of around 22 h [434]. This extended terminal phase is attributed to equilibration between THC stored in lipid compartments and its release back into the bloodstream [435]. Heavy cannabis users display an even longer terminal half-life due to the slow redistribution of THC from deep fatty tissues [436].
Consequently, blood THC concentrations exceeding 1 μg/L may persist for more than 24 h after their last use in heavy users [436,437]. Conversely, CBD also exhibits a prolonged terminal elimination half-life. Following intravenous administration, the average half-life is 24 ± 6 h, while inhalation results in a slightly longer value of 31 ± 4 h [429]. Notably, repeated daily oral administration of CBD leads to a significantly extended half-life, ranging from 2 to 5 days.

5.3. Potential Interactions of Cannabinoids in Postoperative Pain Relief

The metabolism of cannabinoids, particularly THC breakdown by cytochrome P450 (CYP) enzymes [438], suggests potential interactions with various drug classes [439,440,441]. While clinically significant interactions are rare [442], caution is advised, especially when combining cannabinoids with other central nervous system depressants (increased sedation), serotonin reuptake inhibitors (SSRIs)/antidepressants, sympathomimetics (potential for elevated heart rate and blood pressure), or pain medications [443,444,445].
Existing evidence has not demonstrated toxicity or loss of effect of concomitant medications, although such outcomes are theoretically possible. One exception is the interaction between high-dose CBD and clobazam, where elevated levels of a sedative metabolite, N-desmethyl clobazam, necessitate a dose reduction for that drug. Furthermore, the accumulating literature highlights the interaction of CBD with various catalytic activities of cytochrome P450 isoenzymes, demonstrating its potency as an inhibitor of CYP2C19 [446], CYP2D6 [447], or CYP3A4 [448], among others. Thus, it is crucial to consider potential interactions with other concomitant drugs metabolized by these isoenzymes [449].

5.4. Acute Adverse Effects of Cannabinoids

Our understanding of the effects of cannabinoid agonists in humans is predominantly derived from two sources: clinical observations and anecdotal reports from individuals consuming marijuana, as evidenced in Table 2. The pharmacokinetic profile, particularly the time course of action, exhibits significant variability depending on the administered dose and route. For instance, oral administration leads to a slower onset (30 min to 1 h) with longer-lasting effects (approximately 6 h) than inhalation or oral transmucosal routes. These latter routes offer a rapid onset with potent effects, but the duration is shorter. However, once established, the qualitative nature of the effects often displays a similar pattern across individuals [450].
Within the context of POP, the most commonly encountered acute adverse effects of cannabinoids are primarily attributed to their interactions with the central nervous system (CNS). Typically, the consumption of cannabinoids induces in humans an initial feeling of euphoria, well-being, and happiness, followed by a state of drowsiness. During this initial phase, one experiences excitation, dissociation of ideas, increased and distortion of extrasensory perception (increased visual and auditory perception), spatiotemporal errors of appreciation, alterations of emotions, and in some cases, fixed ideas, illusions, irresistible impulses, and hallucinations [428,451]. Other mental and behavioral effects observed in this phase are alterations in memory for recent events [20,452,453], alterations in motor coordination (e.g., driving vehicles), and other psychomotor abilities, difficulties in concentration, especially in complex tasks requiring divided attention, stuporous states (“hanging”), slowing of reactions, decreases in mental activity, and impairments in peripheral vision [454,455]. It is worth mentioning that the effects vary from one individual to another depending on the dose (they increase at higher doses), route of administration, individual vulnerability (personality, expectation, experience of the consumer), as well as the circumstances of consumption. In all cases, they are easily quantifiable, measurable over a few hours (generally no more than 4–6 h) [455,456], and difficult to correlate with plasma levels [457,458]. Dysphoric reactions such as panic and acute anxiety attacks, unpleasant somatic sensations, and paranoid feelings are dose-dependent and occur mostly during initial contact with cannabinoids or individuals with a history of psychosis [459].
Furthermore, their hemodynamic and digestive effects are other undesirable effects to consider during POP. Cannabinoids generally exhibit vasodilatory reflex properties when acting through CB1r [460,461]. This response is multifaceted and may involve three phases: vagal-mediated hypotension (Phase I), followed by a compensatory increase in blood pressure (Phase II), leading to prolonged hypotensive effects (Phase III) [462]. The most consistent cardiovascular effects of both marijuana smoking and i.v. administration of delta-9-THC are peripheral vasodilation and tachycardia (compared with bradycardia in animals), occurring within minutes to a quarter of an hour and lasting up to 3 h [451,463,464,465]. This increase in heart rate may elevate cardiac output and oxygen demand [466]. Blocking drugs can be used to mitigate this effect [467]. Additionally, inhibiting acetylcholine release from the autonomic nervous system fibers after interaction with intestinal CB1r should be considered a cause of intestinal ileus [468].
Nevertheless, cannabinoid receptor agonists offer a promising alternative for pain management compared to current medications. Opioids, while effective, can lead to life-threatening complications and contribute to the opioid crisis [10,469]. NSAIDs, such as ibuprofen and diclofenac, although widely used, can induce cardiovascular toxicity through mechanisms like prostaglandin inhibition. In contrast, the acute toxicity of cannabinoids is very low [67]. The dose of THC required to produce 50% mortality in rodents is extremely high compared with other commonly used drugs [470].

5.5. Patients with a History of Cannabinoid Use

One of the premises of treating POP is the individualization of the guidelines to achieving a satisfactory result and acceptable side effects. In this sense, taking a series of precautions in special risk groups is necessary to reduce undesirable effects.
Studies indicate that 10–20% of individuals aged 18–25 years may use cannabis weekly or more frequently. Due to its slow elimination from the body, these cannabinoids may persist in the tissues for weeks, potentially interacting with various anesthetic agents and affecting their efficacy [471,472]. Additionally, cannabis use may be associated with higher pain scores and poorer quality of sleep in the early postoperative period [410,412]. Therefore, a systematic preoperative inquiry regarding cannabis use is highly recommended. In cases of recent cannabis use, postponing elective surgery is advisable to minimize potential complications [471,472].
It is noteworthy that cannabinoids significantly enhance the hypnotic and sedative effects of CNS depressants commonly used in general anesthesia (barbiturates, opiates, benzodiazepines)—see above. This can lead to excessive sedation and potential respiratory depression. Additionally, cannabis use can also increase the risk of respiratory complications during general anesthesia. Smoking cannabis irritates the upper airway, causing inflammation (oropharyngitis) and swelling of the uvula (uvular edema) [473]. This swelling can obstruct the airway, especially during breathing tube insertion. In rare cases, cannabis use may contribute to isolated uvulitis, presenting with upper airway pain, fever, hypersalivation, dyspnea, and respiratory distress [474,475]. Furthermore, cannabis use has been linked to laryngospasm [476].
On the other hand, hemodynamic effects are also a concern. Low or moderate doses may cause an increase in sympathetic activity, leading to a faster heart rate (tachycardia) and increased cardiac output. Conversely, high doses can suppress sympathetic activity and stimulate parasympathetic activity, resulting in a slower heart rate (bradycardia) and low blood pressure (hypotension). Cannabis-induced hypotension usually responds well to intravenous fluids [477].
In cases of acute cannabis consumption, it is advisable to avoid medications likely to increase heart rate, such as ketamine, atropine, or epinephrine [478]. This is because cannabis can cause pronounced catecholamine release, potentially leading to tachycardia. Conversely, chronic use may cause catecholamine depletion, requiring lower anesthetic doses [471,472].

5.6. Contraindications

Due to potential risks for fetal and neonatal health, including long-term neurodevelopmental effects, cannabis use is contraindicated during pregnancy and lactation [479,480,481]. Similarly, it is contraindicated in psychosis (except for CBD-predominant preparations) [482]. Cannabis should be used with caution in patients with conditions like unstable angina due to tachycardia and possible hypotension from THC, but it does not produce QTc issues [483]. The use of cannabis in children and teens requires further investigation due to potential impacts on cognitive development and academic performance [484]. Similarly, more research is needed to understand its role in addiction and dependency. Smoking cannabis should be avoided for chronic obstructive pulmonary disease (COPD) and asthma.

6. Clinical Trials Evaluating Cannabinoids for Postoperative Pain

Eight clinical trials investigating cannabinoids for POP have been conducted, involving a total of 924 patients and utilizing six different cannabinoid compounds, primarily THC or its analogs (e.g., dronabinol) [297,364,394,395,396,397,402,403] (Table 3). Overall, the trials predominantly reported negative findings, with only two studies demonstrating modest (e.g., slight reduction in pain scores) benefits [394,396]. Notably, six out of eight studies administered a single dose of cannabinoids, while in two other studies, administration was extended for 24 and 36 h, respectively.
These findings contradict some systematic reviews suggesting a potential role for cannabinoids in managing pain beyond acute scenarios [46,485]. Meta-analyses evaluating the analgesic efficacy of cannabinoids for acute POP [486,487] concluded that cannabinoids are not ideal for POP due to the following: (1) limited efficacy in that studies show limited pain reduction [394,396] or no effect [297] and (2) potential for hyperalgesia whereby high doses may even worsen pain [397]. To evaluate these findings, the best available qualitative evidence indicates no disparities in cumulative opioid consumption and no variances in the severity of rest pain at 24 h postoperatively.
Cannabinoids are generally well tolerated, with most adverse effects being mild to moderate [486,487]. Common side effects include blurred vision, hypotension, dizziness, drowsiness, dry mouth, hallucination, headache, and nausea. In fact, in five of eight studies [297,364,396,397,402], cannabinoids showed more frequent or severe adverse effects than the placebo for specific events or periods. Nevertheless, analyzing the adverse effect profile of cannabinoids is challenging due to variations in reporting and defining adverse effects among studies. Moreover, some studies failed to assess or report the statistical significance of group differences. For instance, one study noted that patients receiving a placebo were more likely to report postoperative nausea and vomiting compared to those receiving dronabinol, but the statistical significance of this finding was not provided [364]. Additionally, there is an observed increase in hypotension during the postoperative period, posing a risk factor for cerebrovascular disease [488]. A recent randomized controlled trial evaluating the efficacy of intravenous THC in preventing postoperative nausea and vomiting did not recommend its use due to an unacceptable side effect profile and limited efficacy [489].
It is worth noting that despite promising results in animal models, CBD appears ineffective for POP management. Three recent studies explored its therapeutic potential. In the first study, topical CBD administered to patients who had undergone total knee arthroplasty as a supplement to a standardized multimodal analgesic protocol did not reduce pain or opioid consumption [400]. In the second study, buccally absorbed CBD in arthroscopic rotator cuff repair patients showed a suitable safety profile and held promise in reducing pain in the immediate perioperative period [490]. However, a follow-up study indicated that CBD was not able to improve pain scores, patient satisfaction with pain control, or postoperative opioid consumption [399]. Finally, CBD was ineffective in reducing discomfort or opioid usage in patients undergoing ureteroscopy with stent placement for urinary stone disease [401].
The Clinical Trial as Proof of Principle of Analgesic Efficacy of Cannabinoids on Postoperative Pain (CANPOP) clinical trial, funded by the Medical Research Council, evaluated the analgesic efficacy of standardized cannabis plant extract (Cannador) administered by p.o. one hour before the intervention in patients with POP (tonsillectomized and patients undergoing abdominal surgery). This clinical study reported significant dose-related improvements in rescue analgesia requirements [394].
In conclusion, the use of cannabinoids for POP presents a mixed picture. While some studies show promise, the evidence suggests limited efficacy and potential side effects.

7. Possible Explanations for the Lack of Analgesic Efficacy in Postoperative Pain Relief

Unlike chronic pain, which is often dynamic and neuropathic, POP is acute, localized, and primarily driven by nociceptive mechanisms [491,492]. This difference in pain type might explain the limited effectiveness of cannabinoids for POP compared to chronic pain. Chronic pain is associated with complex changes in the ECS, including upregulation of cannabinoid receptors, altered receptor function, changes in eCB formation or release, and interactions with other pain mediators [393,493]. Supporting this hypothesis, are studies on healthy volunteers, suggesting limited effectiveness of cannabinoids for acute nociceptive pain [298].
Additional factors contributing to the lack of desired results are outlined in Table 4. Notably, the clinical dosage is constrained by the onset of THC-related side effects, being mainly psychotropic [464,494]. Furthermore, an analysis of studies revealed significant disparities in dosing regimens and administration patterns. Timing of administration varied across studies, with some administering cannabinoids preoperatively [363,396,402,403], postoperatively [297,394,395,398], and a few in both settings [412,495]. Moreover, a diverse range of analgesic protocols was employed, with some studies using unimodal opioid-based analgesia, others employing multimodal analgesia, and some using unspecified multimodal regimens [297,363,364,394,397,398,402,403,412,495]. Routes of administration also varied, with most studies investigating the oral route (PO). The heterogeneity of patient populations included in the studies, ranging from healthy individuals to those with various etiologies of diseases [86,495], further complicated the interpretation of the results.
Another factor contributing to the limited evidence for cannabinoids in POP relief might be the way surveys are worded. The phrasing of questions posed to patients could unintentionally influence their responses, hindering researchers’ ability to obtain accurate data relevant to their specific research goals. This is particularly noteworthy considering the findings of Khelemsky et al. [405]. Despite systematic reviews highlighting a lack of robust clinical evidence for cannabinoids in acute pain management, their study showed that patients generally perceive marijuana as at least somewhat effective for pain control. Additionally, patients expressed a willingness to use cannabinoid medications if prescribed by a healthcare provider. This discrepancy highlights the potential influence of question formulation on research outcomes.
Moreover, the selectivity of cannabinoid analogs at CB1 and CB2 receptors is crucial, which may be one of the leading causes of the therapeutic failure found in some studies, considering the opposite effects that may occur between CB1r and CB2r activation in some experimental contexts. These receptors have distinct locations and functions [97,184,496,497]. CB1 receptors are primarily located in the central nervous system, and their activation can produce psychotropic side effects [498,499,500]. CB2 receptors are abundant in peripheral tissues [25,501,502]. Their activation is associated with anti-inflammatory effects and does not show psychotropic actions [25,115,503]. These effects emphasize the importance of developing cannabinoid medications with targeted selectivity for CB2 receptors. By focusing on CB2 activation, researchers might achieve better pain relief with fewer side effects than medications that activate CB1 and CB2 receptors [18,22,227].
Finally, Pernia-Andrade et al. [504] provide compelling evidence supporting a pain-exacerbating mechanism of cannabinoid signaling in animals exposed to intense noxious stimuli. Their findings suggest that cannabinoid drugs and endocannabinoids (eCBs) produced in the spinal cord can disrupt the inhibitory regulation of pain-perceiving neurons, thereby facilitating the transmission of painful and non-painful mechanical stimuli along pain pathways to higher brain centers, effectively opening a “pain gate”.

8. Future Perspectives and Conclusions

The exploration of cannabinoid neuromodulation systems and the synthesis of CB1r and CB2r agonists present promising avenues for therapeutic use in pain management. Despite the demonstrated antinociceptive activity of cannabinoid agonists, particularly in preclinical studies, their clinical efficacy in the treatment of POP remains inconclusive. While some evidence suggests potential benefits, such as the avoidance of known adverse effects associated with traditional analgesics like opioids and nonsteroidal anti-inflammatory drugs (NSAIDs), the overall clinical data on cannabinoid effectiveness in perioperative and acute pain settings are limited and heterogeneous.
The current literature underscores the need for further well-designed clinical trials to elucidate the specific pathologies and conditions wherein cannabinoid agents might offer advantages over existing therapeutic options. Notably, such trials should adhere to rigorous methodological standards, as outlined by initiatives like the Initiative on Methods, Measurement, and Pain Assessment in Clinical Trials (IMMPACT), to ensure the validity and reliability of their findings [505]. Additionally, while new routes of cannabinoid administration, such as oral cannabis oil formulations, offer potential benefits in terms of safety and patient acceptability, their efficacy in the context of postoperative pain management requires robust investigation.
Furthermore, the potential long-term implications of cannabinoid therapy in reducing the incidence of chronic postsurgical pain and mitigating the need for prolonged opioid use remain largely unexplored. Large-scale, multicenter trials are warranted to comprehensively evaluate the role of cannabinoids in acute and postoperative pain relief, with careful consideration given to patient safety, optimal dosing regimens, and comparative effectiveness against established analgesic agents. Only through such concerted research efforts can we definitively determine the place of cannabinoids in the armamentarium of pain management strategies, thereby providing clinicians with evidence-based guidance for optimizing patient care in this challenging clinical domain.

Author Contributions

Conceptualization, A.J.C., F.N. and J.M.; methodology, A.J.C., F.N., R.S., M.S.G.-G., B.M., D.N., F.M.G.-G., A.G., E.M.-S., A.B.T., P.P.-D., L.G. and J.M.; figure design and creation, A.B.T. and L.G.; writing—original draft preparation, A.J.C., F.N. and J.M.; writing—review and editing, F.N. and J.M.; funding acquisition, J.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Mutua Madrileña Foundation, grant number UGP-22-010 to J.M.

Acknowledgments

We thank BioRender for preparing the figures used in this manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Raja, S.N.; Carr, D.B.; Cohen, M.; Finnerup, N.B.; Flor, H.; Gibson, S.; Keefe, F.J.; Mogil, J.S.; Ringkamp, M.; Sluka, K.A.; et al. The revised International Association for the Study of Pain definition of pain: Concepts, challenges, and compromises. Pain 2020, 161, 1976–1982. [Google Scholar] [CrossRef] [PubMed]
  2. Armstrong, S.A.; Herr, M.J. Physiology, Nociception. In StatPearls; StatPearls Publishing LLC: Treasure Island, FL, USA, 2024. [Google Scholar]
  3. Attal, N.; Bouhassira, D.; Colvin, L. Advances and challenges in neuropathic pain: A narrative review and future directions. Br. J. Anaesth. 2023, 131, 79–92. [Google Scholar] [CrossRef] [PubMed]
  4. Brennan, T.J.; Zahn, P.K.; Pogatzki-Zahn, E.M. Mechanisms of incisional pain. Anesthesiol. Clin. N. Am. 2005, 23, 1–20. [Google Scholar] [CrossRef] [PubMed]
  5. Treede, R.D.; Meyer, R.A.; Raja, S.N.; Campbell, J.N. Peripheral and central mechanisms of cutaneous hyperalgesia. Prog. Neurobiol. 1992, 38, 397–421. [Google Scholar] [CrossRef] [PubMed]
  6. Hug, C.C., Jr. Opioids: Clinical use as anesthetic agents. J. Pain Symptom Manag. 1992, 7, 350–355. [Google Scholar] [CrossRef] [PubMed]
  7. Peponis, T.; Kaafarani, H.M.A. What Is the Proper Use of Opioids in the Postoperative Patient? Adv. Surg. 2017, 51, 77–87. [Google Scholar] [CrossRef]
  8. Matsuda, L.A.; Lolait, S.J.; Brownstein, M.J.; Young, A.C.; Bonner, T.I. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature 1990, 346, 561–564. [Google Scholar] [CrossRef] [PubMed]
  9. Manchikanti, L.; Kaye, A.M.; Knezevic, N.N.; McAnally, H.; Slavin, K.; Trescot, A.M.; Blank, S.; Pampati, V.; Abdi, S.; Grider, J.S.; et al. Responsible, Safe, and Effective Prescription of Opioids for Chronic Non-Cancer Pain: American Society of Interventional Pain Physicians (ASIPP) Guidelines. Pain Physician 2017, 20, S3–S92. [Google Scholar] [CrossRef]
  10. Alexander, J.C.; Patel, B.; Joshi, G.P. Perioperative use of opioids: Current controversies and concerns. Best Pract. Res. Clin. Anaesthesiol. 2019, 33, 341–351. [Google Scholar] [CrossRef] [PubMed]
  11. Hah, J.M.; Bateman, B.T.; Ratliff, J.; Curtin, C.; Sun, E. Chronic Opioid Use After Surgery: Implications for Perioperative Management in the Face of the Opioid Epidemic. Anesth. Analg. 2017, 125, 1733–1740. [Google Scholar] [CrossRef]
  12. Quinlan, J.; Lobo, D.N.; Levy, N. Postoperative pain management: Time to get back on track. Anaesthesia 2020, 75 (Suppl. S1), e10–e13. [Google Scholar] [CrossRef] [PubMed]
  13. Mechoulam, R.; Hanus, L. A historical overview of chemical research on cannabinoids. Chem. Phys. Lipids 2000, 108, 1–13. [Google Scholar] [CrossRef]
  14. Munro, S.; Thomas, K.L.; Abu-Shaar, M. Molecular characterization of a peripheral receptor for cannabinoids. Nature 1993, 365, 61–65. [Google Scholar] [CrossRef] [PubMed]
  15. Devane, W.A.; Hanus, L.; Breuer, A.; Pertwee, R.G.; Stevenson, L.A.; Griffin, G.; Gibson, D.; Mandelbaum, A.; Etinger, A.; Mechoulam, R. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science 1992, 258, 1946–1949. [Google Scholar] [CrossRef] [PubMed]
  16. Mechoulam, R.; Ben-Shabat, S.; Hanus, L.; Ligumsky, M.; Kaminski, N.E.; Schatz, A.R.; Gopher, A.; Almog, S.; Martin, B.R.; Compton, D.R.; et al. Identification of an endogenous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors. Biochem. Pharmacol. 1995, 50, 83–90. [Google Scholar] [CrossRef] [PubMed]
  17. Di, M.; Bisogno, T.; De Petrocellis, L. Endocannabinoids: New targets for drug development. Curr. Pharm. Des. 2000, 6, 1361–1380. [Google Scholar] [CrossRef] [PubMed]
  18. Romero-Sandoval, A.; Eisenach, J.C. Spinal cannabinoid receptor type 2 activation reduces hypersensitivity and spinal cord glial activation after paw incision. Anesthesiology 2007, 106, 787–794. [Google Scholar] [CrossRef] [PubMed]
  19. Alkaitis, M.S.; Solorzano, C.; Landry, R.P.; Piomelli, D.; DeLeo, J.A.; Romero-Sandoval, E.A. Evidence for a role of endocannabinoids, astrocytes and p38 phosphorylation in the resolution of postoperative pain. PLoS ONE 2010, 5, e10891. [Google Scholar] [CrossRef] [PubMed]
  20. Howlett, A.C.; Barth, F.; Bonner, T.I.; Cabral, G.; Casellas, P.; Devane, W.A.; Felder, C.C.; Herkenham, M.; Mackie, K.; Martin, B.R.; et al. International Union of Pharmacology. XXVII. Classification of cannabinoid receptors. Pharmacol. Rev. 2002, 54, 161–202. [Google Scholar] [CrossRef] [PubMed]
  21. Mukhopadhyay, S.; Shim, J.Y.; Assi, A.A.; Norford, D.; Howlett, A.C. CB1 cannabinoid receptor-G protein association: A possible mechanism for differential signaling. Chem. Phys. Lipids 2002, 121, 91–109. [Google Scholar] [CrossRef] [PubMed]
  22. Guindon, J.; Hohmann, A.G. Cannabinoid CB2 receptors: A therapeutic target for the treatment of inflammatory and neuropathic pain. Br. J. Pharmacol. 2008, 153, 319–334. [Google Scholar] [CrossRef] [PubMed]
  23. Walker, J.M.; Huang, S.M. Cannabinoid analgesia. Pharmacol. Ther. 2002, 95, 127–135. [Google Scholar] [CrossRef] [PubMed]
  24. Sugiura, T.; Kondo, S.; Sukagawa, A.; Nakane, S.; Shinoda, A.; Itoh, K.; Yamashita, A.; Waku, K. 2-Arachidonoylglycerol: A possible endogenous cannabinoid receptor ligand in brain. Biochem. Biophys. Res. Commun. 1995, 215, 89–97. [Google Scholar] [CrossRef] [PubMed]
  25. Malan, P.T., Jr.; Ibrahim, M.M.; Deng, H.; Liu, Q.; Mata, H.P.; Vanderah, T.; Porreca, F.; Makriyannis, A. CB2 cannabinoid receptor-mediated peripheral antinociception. Pain 2001, 93, 239–245. [Google Scholar] [CrossRef] [PubMed]
  26. Clayton, N.; Marshall, F.H.; Bountra, C.; O’Shaughnessy, C.T. CB1 and CB2 cannabinoid receptors are implicated in inflammatory pain. Pain 2002, 96, 253–260. [Google Scholar] [CrossRef] [PubMed]
  27. Ibrahim, M.M.; Deng, H.; Zvonok, A.; Cockayne, D.A.; Kwan, J.; Mata, H.P.; Vanderah, T.W.; Lai, J.; Porreca, F.; Makriyannis, A.; et al. Activation of CB2 cannabinoid receptors by AM1241 inhibits experimental neuropathic pain: Pain inhibition by receptors not present in the CNS. Proc. Natl. Acad. Sci. USA 2003, 100, 10529–10533. [Google Scholar] [CrossRef] [PubMed]
  28. Ibrahim, M.M.; Porreca, F.; Lai, J.; Albrecht, P.J.; Rice, F.L.; Khodorova, A.; Davar, G.; Makriyannis, A.; Vanderah, T.W.; Mata, H.P.; et al. CB2 cannabinoid receptor activation produces antinociception by stimulating peripheral release of endogenous opioids. Proc. Natl. Acad. Sci. USA 2005, 102, 3093–3098. [Google Scholar] [CrossRef] [PubMed]
  29. Nackley, A.G.; Makriyannis, A.; Hohmann, A.G. Selective activation of cannabinoid CB2 receptors suppresses spinal fos protein expression and pain behavior in a rat model of inflammation. Neuroscience 2003, 119, 747–757. [Google Scholar] [CrossRef] [PubMed]
  30. Hohmann, A.G.; Farthing, J.N.; Zvonok, A.M.; Makriyannis, A. Selective activation of cannabinoid CB2 receptors suppresses hyperalgesia evoked by intradermal capsaicin. J. Pharmacol. Exp. Ther. 2004, 308, 446–453. [Google Scholar] [CrossRef] [PubMed]
  31. Scott, D.A.; Wright, C.E.; Angus, J.A. Evidence that CB-1 and CB-2 cannabinoid receptors mediate antinociception in neuropathic pain in the rat. Pain 2004, 109, 124–131. [Google Scholar] [CrossRef]
  32. Valenzano, K.J.; Tafesse, L.; Lee, G.; Harrison, J.E.; Boulet, J.M.; Gottshall, S.L.; Mark, L.; Pearson, M.S.; Miller, W.; Shan, S.; et al. Pharmacological and pharmacokinetic characterization of the cannabinoid receptor 2 agonist, GW405833, utilizing rodent models of acute and chronic pain, anxiety, ataxia and catalepsy. Neuropharmacology 2005, 48, 658–672. [Google Scholar] [CrossRef] [PubMed]
  33. Sampson, P.B. Phytocannabinoid Pharmacology: Medicinal Properties of Cannabis sativa Constituents Aside from the “Big Two”. J. Nat. Prod. 2021, 84, 142–160. [Google Scholar] [CrossRef] [PubMed]
  34. Taura, F.; Sirikantaramas, S.; Shoyama, Y.; Yoshikai, K.; Shoyama, Y.; Morimoto, S. Cannabidiolic-acid synthase, the chemotype-determining enzyme in the fiber-type Cannabis sativa. FEBS Lett. 2007, 581, 2929–2934. [Google Scholar] [CrossRef] [PubMed]
  35. Radwan, M.M.; Chandra, S.; Gul, S.; ElSohly, M.A. Cannabinoids, Phenolics, Terpenes and Alkaloids of Cannabis. Molecules 2021, 26, 2774. [Google Scholar] [CrossRef] [PubMed]
  36. McPartland, J.M. Cannabis Systematics at the Levels of Family, Genus, and Species. Cannabis Cannabinoid Res. 2018, 3, 203–212. [Google Scholar] [CrossRef]
  37. Mechoulam, R. Cannabinoids as Therapeutic Agents, 1st ed.; Mechoulam, R., Ed.; Chapman and Hall/CRC: New York, NY, USA, 1986; p. 186. [Google Scholar] [CrossRef]
  38. Abel, E.L. Marihuana, the First Twelve Thousand Years; McGraw-Hill: New York, NY, USA, 1982; p. 304. [Google Scholar]
  39. Mehmedic, Z.; Chandra, S.; Slade, D.; Denham, H.; Foster, S.; Patel, A.S.; Ross, S.A.; Khan, I.A.; ElSohly, M.A. Potency trends of Delta9-THC and other cannabinoids in confiscated cannabis preparations from 1993 to 2008. J. Forensic Sci. 2010, 55, 1209–1217. [Google Scholar] [CrossRef] [PubMed]
  40. Al Ubeed, H.M.S.; Bhuyan, D.J.; Alsherbiny, M.A.; Basu, A.; Vuong, Q.V. A Comprehensive Review on the Techniques for Extraction of Bioactive Compounds from Medicinal Cannabis. Molecules 2022, 27, 604. [Google Scholar] [CrossRef]
  41. Abood, M.E.; Martin, B.R. Neurobiology of marijuana abuse. Trends Pharmacol. Sci. 1992, 13, 201–206. [Google Scholar] [CrossRef] [PubMed]
  42. Pertwee, R.G. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: Delta9-tetrahydrocannabinol, cannabidiol and delta9-tetrahydrocannabivarin. Br. J. Pharmacol. 2008, 153, 199–215. [Google Scholar] [CrossRef] [PubMed]
  43. Hanus, L.O.; Hod, Y. Terpenes/Terpenoids in Cannabis: Are They Important? Med. Cannabis Cannabinoids 2020, 3, 25–60. [Google Scholar] [CrossRef] [PubMed]
  44. Mechoulam, R. Marihuana chemistry. Science 1970, 168, 1159–1166. [Google Scholar] [CrossRef] [PubMed]
  45. Martin, B.R. Structural requirements for cannabinoid-induced antinociceptive activity in mice. Life Sci. 1985, 36, 1523–1530. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, L.; Hong, P.J.; May, C.; Rehman, Y.; Oparin, Y.; Hong, C.J.; Hong, B.Y.; AminiLari, M.; Gallo, L.; Kaushal, A.; et al. Medical cannabis or cannabinoids for chronic non-cancer and cancer related pain: A systematic review and meta-analysis of randomised clinical trials. BMJ 2021, 374, n1034. [Google Scholar] [CrossRef] [PubMed]
  47. Lynch, M.E.; Campbell, F. Cannabinoids for treatment of chronic non-cancer pain; a systematic review of randomized trials. Br. J. Clin. Pharmacol. 2011, 72, 735–744. [Google Scholar] [CrossRef] [PubMed]
  48. Strand, N.; D’Souza, R.S.; Karri, J.; Kalia, H.; Weisbein, J.; Kassa, B.J.; Hussain, N.; Chitneni, A.; Budwany, R.R.; Hagedorn, J.; et al. Medical Cannabis: A Review from the American Society of Pain and Neuroscience. J. Pain Res. 2023, 16, 4217–4228. [Google Scholar] [CrossRef] [PubMed]
  49. Abrams, D.I.; Jay, C.A.; Shade, S.B.; Vizoso, H.; Reda, H.; Press, S.; Kelly, M.E.; Rowbotham, M.C.; Petersen, K.L. Cannabis in painful HIV-associated sensory neuropathy: A randomized placebo-controlled trial. Neurology 2007, 68, 515–521. [Google Scholar] [CrossRef] [PubMed]
  50. Andreae, M.H.; Carter, G.M.; Shaparin, N.; Suslov, K.; Ellis, R.J.; Ware, M.A.; Abrams, D.I.; Prasad, H.; Wilsey, B.; Indyk, D.; et al. Inhaled Cannabis for Chronic Neuropathic Pain: A Meta-analysis of Individual Patient Data. J. Pain 2015, 16, 1221–1232. [Google Scholar] [CrossRef] [PubMed]
  51. Ellis, R.J.; Toperoff, W.; Vaida, F.; van den Brande, G.; Gonzales, J.; Gouaux, B.; Bentley, H.; Atkinson, J.H. Smoked medicinal cannabis for neuropathic pain in HIV: A randomized, crossover clinical trial. Neuropsychopharmacology 2009, 34, 672–680. [Google Scholar] [CrossRef] [PubMed]
  52. Wallace, M.S.; Marcotte, T.D.; Umlauf, A.; Gouaux, B.; Atkinson, J.H. Efficacy of Inhaled Cannabis on Painful Diabetic Neuropathy. J. Pain 2015, 16, 616–627. [Google Scholar] [CrossRef] [PubMed]
  53. Ware, M.A.; Wang, T.; Shapiro, S.; Robinson, A.; Ducruet, T.; Huynh, T.; Gamsa, A.; Bennett, G.J.; Collet, J.P. Smoked cannabis for chronic neuropathic pain: A randomized controlled trial. Can. Med. Assoc. J. 2010, 182, E694–E701. [Google Scholar] [CrossRef] [PubMed]
  54. Wilsey, B.; Marcotte, T.; Tsodikov, A.; Millman, J.; Bentley, H.; Gouaux, B.; Fishman, S. A randomized, placebo-controlled, crossover trial of cannabis cigarettes in neuropathic pain. J. Pain 2008, 9, 506–521. [Google Scholar] [CrossRef] [PubMed]
  55. Bakheit, A.M. The pharmacological management of post-stroke muscle spasticity. Drugs Aging 2012, 29, 941–947. [Google Scholar] [CrossRef] [PubMed]
  56. Koppel, B.S.; Brust, J.C.; Fife, T.; Bronstein, J.; Youssof, S.; Gronseth, G.; Gloss, D. 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, 1556–1563. [Google Scholar] [CrossRef] [PubMed]
  57. Passie, T.; Emrich, H.M.; Karst, M.; Brandt, S.D.; Halpern, J.H. Mitigation of post-traumatic stress symptoms by Cannabis resin: A review of the clinical and neurobiological evidence. Drug Test. Anal. 2012, 4, 649–659. [Google Scholar] [CrossRef] [PubMed]
  58. McGeeney, B.E. Cannabinoids and hallucinogens for headache. Headache 2013, 53, 447–458. [Google Scholar] [CrossRef] [PubMed]
  59. Chang, A.E.; Shiling, D.J.; Stillman, R.C.; Goldberg, N.H.; Seipp, C.A.; Barofsky, I.; Rosenberg, S.A. A prospective evaluation of delta-9-tetrahydrocannabinol as an antiemetic in patients receiving adriamycin and cytoxan chemotherapy. Cancer 1981, 47, 1746–1751. [Google Scholar] [CrossRef] [PubMed]
  60. Naftali, T.; Mechulam, R.; Lev, L.B.; Konikoff, F.M. Cannabis for inflammatory bowel disease. Dig. Dis. 2014, 32, 468–474. [Google Scholar] [CrossRef] [PubMed]
  61. Schierenbeck, T.; Riemann, D.; Berger, M.; Hornyak, M. Effect of illicit recreational drugs upon sleep: Cocaine, ecstasy and marijuana. Sleep Med. Rev. 2008, 12, 381–389. [Google Scholar] [CrossRef] [PubMed]
  62. Ware, M.A.; Fitzcharles, M.A.; Joseph, L.; Shir, Y. The effects of nabilone on sleep in fibromyalgia: Results of a randomized controlled trial. Anesth. Analg. 2010, 110, 604–610. [Google Scholar] [CrossRef] [PubMed]
  63. Cohen, P.J. Medical marijuana: The conflict between scientific evidence and political ideology. Part two of two. J. Pain Palliat. Care Pharmacother. 2009, 23, 120–140. [Google Scholar] [CrossRef] [PubMed]
  64. Hall, W.; Stjepanovic, D.; Caulkins, J.; Lynskey, M.; Leung, J.; Campbell, G.; Degenhardt, L. Public health implications of legalising the production and sale of cannabis for medicinal and recreational use. Lancet 2019, 394, 1580–1590. [Google Scholar] [CrossRef]
  65. Shover, C.L.; Humphreys, K. Six policy lessons relevant to cannabis legalization. Am. J. Drug Alcohol Abus. 2019, 45, 698–706. [Google Scholar] [CrossRef] [PubMed]
  66. Goodman, S.; Wadsworth, E.; Leos-Toro, C.; Hammond, D.; International Cannabis Policy Study Team. Prevalence and forms of cannabis use in legal vs. illegal recreational cannabis markets. Int. J. Drug Policy 2020, 76, 102658. [Google Scholar] [CrossRef] [PubMed]
  67. Hall, W.; Lynskey, M. Assessing the public health impacts of legalizing recreational cannabis use: The US experience. World Psychiatry 2020, 19, 179–186. [Google Scholar] [CrossRef] [PubMed]
  68. Tashkin, D.P. Effects of marijuana smoking on the lung. Ann. Am. Thorac. Soc. 2013, 10, 239–247. [Google Scholar] [CrossRef]
  69. Wu, T.C.; Tashkin, D.P.; Djahed, B.; Rose, J.E. Pulmonary hazards of smoking marijuana as compared with tobacco. N. Engl. J. Med. 1988, 318, 347–351. [Google Scholar] [CrossRef] [PubMed]
  70. Piomelli, D. The molecular logic of endocannabinoid signalling. Nat. Rev. Neurosci. 2003, 4, 873–884. [Google Scholar] [CrossRef] [PubMed]
  71. Mechoulam, R.; Parker, L.A. The endocannabinoid system and the brain. Annu. Rev. Psychol. 2013, 64, 21–47. [Google Scholar] [CrossRef] [PubMed]
  72. Zou, S.; Kumar, U. Cannabinoid Receptors and the Endocannabinoid System: Signaling and Function in the Central Nervous System. Int. J. Mol. Sci. 2018, 19, 833. [Google Scholar] [CrossRef]
  73. Hillard, C.J. The Endocannabinoid Signaling System in the CNS: A Primer. Int. Rev. Neurobiol. 2015, 125, 1–47. [Google Scholar] [CrossRef] [PubMed]
  74. Pacher, P.; Kogan, N.M.; Mechoulam, R. Beyond THC and Endocannabinoids. Annu. Rev. Pharmacol. Toxicol. 2020, 60, 637–659. [Google Scholar] [CrossRef] [PubMed]
  75. Finn, D.P. Endocannabinoid-mediated modulation of stress responses: Physiological and pathophysiological significance. Immunobiology 2010, 215, 629–646. [Google Scholar] [CrossRef] [PubMed]
  76. De Laurentiis, A.; Correa, F.; Fernandez Solari, J. Endocannabinoid System in the Neuroendocrine Response to Lipopolysaccharide-induced Immune Challenge. J. Endocr. Soc. 2022, 6, bvac120. [Google Scholar] [CrossRef] [PubMed]
  77. Varvel, S.A.; Lichtman, A.H. Evaluation of CB1 receptor knockout mice in the Morris water maze. J. Pharmacol. Exp. Ther. 2002, 301, 915–924. [Google Scholar] [CrossRef] [PubMed]
  78. Marsicano, G.; Wotjak, C.T.; Azad, S.C.; Bisogno, T.; Rammes, G.; Cascio, M.G.; Hermann, H.; Tang, J.; Hofmann, C.; Zieglgansberger, W.; et al. The endogenous cannabinoid system controls extinction of aversive memories. Nature 2002, 418, 530–534. [Google Scholar] [CrossRef] [PubMed]
  79. Martin, M.; Ledent, C.; Parmentier, M.; Maldonado, R.; Valverde, O. Involvement of CB1 cannabinoid receptors in emotional behaviour. Psychopharmacology 2002, 159, 379–387. [Google Scholar] [CrossRef] [PubMed]
  80. Tan, H.; Ahmad, T.; Loureiro, M.; Zunder, J.; Laviolette, S.R. The role of cannabinoid transmission in emotional memory formation: Implications for addiction and schizophrenia. Front. Psychiatry 2014, 5, 73. [Google Scholar] [CrossRef] [PubMed]
  81. Di Marzo, V.; Goparaju, S.K.; Wang, L.; Liu, J.; Batkai, S.; Jarai, Z.; Fezza, F.; Miura, G.I.; Palmiter, R.D.; Sugiura, T.; et al. Leptin-regulated endocannabinoids are involved in maintaining food intake. Nature 2001, 410, 822–825. [Google Scholar] [CrossRef] [PubMed]
  82. Maccarrone, M.; Bari, M.; Battista, N.; Finazzi-Agro, A. Endocannabinoid degradation, endotoxic shock and inflammation. Curr. Drug Targets Inflamm. Allergy 2002, 1, 53–63. [Google Scholar] [CrossRef] [PubMed]
  83. Nagarkatti, P.; Pandey, R.; Rieder, S.A.; Hegde, V.L.; Nagarkatti, M. Cannabinoids as novel anti-inflammatory drugs. Future Med. Chem. 2009, 1, 1333–1349. [Google Scholar] [CrossRef] [PubMed]
  84. Hogestatt, E.D.; Zygmunt, P.M. Cardiovascular pharmacology of anandamide. Prostaglandins Leukot. Essent. Fat. Acids 2002, 66, 343–351. [Google Scholar] [CrossRef] [PubMed]
  85. O’Sullivan, S.E. Endocannabinoids and the Cardiovascular System in Health and Disease. In Handbook of Experimental Pharmacology; Springer: Berlin/Heidelberg, Germany, 2015; Volume 231, pp. 393–422. [Google Scholar] [CrossRef]
  86. Li, C.; Jones, P.M.; Persaud, S.J. Role of the endocannabinoid system in food intake, energy homeostasis and regulation of the endocrine pancreas. Pharmacol. Ther. 2011, 129, 307–320. [Google Scholar] [CrossRef] [PubMed]
  87. Bellocchio, L.; Cervino, C.; Pasquali, R.; Pagotto, U. The endocannabinoid system and energy metabolism. J. Neuroendocrinol. 2008, 20, 850–857. [Google Scholar] [CrossRef] [PubMed]
  88. Taylor, A.H.; Amoako, A.A.; Bambang, K.; Karasu, T.; Gebeh, A.; Lam, P.M.; Marzcylo, T.H.; Konje, J.C. Endocannabinoids and pregnancy. Clin. Chim. Acta 2010, 411, 921–930. [Google Scholar] [CrossRef] [PubMed]
  89. Correa, F.; Wolfson, M.L.; Valchi, P.; Aisemberg, J.; Franchi, A.M. Endocannabinoid system and pregnancy. Reproduction 2016, 152, R191–R200. [Google Scholar] [CrossRef] [PubMed]
  90. Guindon, J.; Hohmann, A.G. The endocannabinoid system and pain. CNS Neurol. Disord. Drug Targets 2009, 8, 403–421. [Google Scholar] [CrossRef] [PubMed]
  91. Zogopoulos, P.; Vasileiou, I.; Patsouris, E.; Theocharis, S.E. The role of endocannabinoids in pain modulation. Fundam. Clin. Pharmacol. 2013, 27, 64–80. [Google Scholar] [CrossRef] [PubMed]
  92. Finn, D.P.; Haroutounian, S.; Hohmann, A.G.; Krane, E.; Soliman, N.; Rice, A.S.C. Cannabinoids, the endocannabinoid system, and pain: A review of preclinical studies. Pain 2021, 162, S5–S25. [Google Scholar] [CrossRef] [PubMed]
  93. Toth, B.I.; Dobrosi, N.; Dajnoki, A.; Czifra, G.; Olah, A.; Szollosi, A.G.; Juhasz, I.; Sugawara, K.; Paus, R.; Biro, T. Endocannabinoids modulate human epidermal keratinocyte proliferation and survival via the sequential engagement of cannabinoid receptor-1 and transient receptor potential vanilloid-1. J. Investig. Dermatol. 2011, 131, 1095–1104. [Google Scholar] [CrossRef] [PubMed]
  94. Costa, L.; Moreia-Pinto, B.; Felgueira, E.; Ribeiro, A.; Rebelo, I.; Fonseca, B.M. The major endocannabinoid anandamide (AEA) induces apoptosis of human granulosa cells. Prostaglandins Leukot. Essent. Fat. Acids 2021, 171, 102311. [Google Scholar] [CrossRef]
  95. De Petrocellis, L.; Cascio, M.G.; Di Marzo, V. The endocannabinoid system: A general view and latest additions. Br. J. Pharmacol. 2004, 141, 765–774. [Google Scholar] [CrossRef] [PubMed]
  96. Cristino, L.; Bisogno, T.; Di Marzo, V. Cannabinoids and the expanded endocannabinoid system in neurological disorders. Nat. Rev. Neurol. 2020, 16, 9–29. [Google Scholar] [CrossRef] [PubMed]
  97. Rezende, B.; Alencar, A.K.N.; de Bem, G.F.; Fontes-Dantas, F.L.; Montes, G.C. Endocannabinoid System: Chemical Characteristics and Biological Activity. Pharmaceuticals 2023, 16, 148. [Google Scholar] [CrossRef] [PubMed]
  98. Baker, D.; Pryce, G.; Giovannoni, G.; Thompson, A.J. The therapeutic potential of cannabis. Lancet Neurol. 2003, 2, 291–298. [Google Scholar] [CrossRef] [PubMed]
  99. Castillo, P.E.; Younts, T.J.; Chavez, A.E.; Hashimotodani, Y. Endocannabinoid signaling and synaptic function. Neuron 2012, 76, 70–81. [Google Scholar] [CrossRef] [PubMed]
  100. Nomura, D.K.; Morrison, B.E.; Blankman, J.L.; Long, J.Z.; Kinsey, S.G.; Marcondes, M.C.; Ward, A.M.; Hahn, Y.K.; Lichtman, A.H.; Conti, B.; et al. Endocannabinoid hydrolysis generates brain prostaglandins that promote neuroinflammation. Science 2011, 334, 809–813. [Google Scholar] [CrossRef] [PubMed]
  101. Katona, I.; Freund, T.F. Multiple functions of endocannabinoid signaling in the brain. Annu. Rev. Neurosci. 2012, 35, 529–558. [Google Scholar] [CrossRef] [PubMed]
  102. Ligresti, A.; De Petrocellis, L.; Di Marzo, V. From Phytocannabinoids to Cannabinoid Receptors and Endocannabinoids: Pleiotropic Physiological and Pathological Roles Through Complex Pharmacology. Physiol. Rev. 2016, 96, 1593–1659. [Google Scholar] [CrossRef] [PubMed]
  103. Pertwee, R.G.; Howlett, A.C.; Abood, M.E.; Alexander, S.P.; Di Marzo, V.; Elphick, M.R.; Greasley, P.J.; Hansen, H.S.; Kunos, G.; Mackie, K.; et al. International Union of Basic and Clinical Pharmacology. LXXIX. Cannabinoid receptors and their ligands: Beyond CB1 and CB2. Pharmacol. Rev. 2010, 62, 588–631. [Google Scholar] [CrossRef] [PubMed]
  104. Gomes, I.; Grushko, J.S.; Golebiewska, U.; Hoogendoorn, S.; Gupta, A.; Heimann, A.S.; Ferro, E.S.; Scarlata, S.; Fricker, L.D.; Devi, L.A. Novel endogenous peptide agonists of cannabinoid receptors. FASEB J. 2009, 23, 3020–3029. [Google Scholar] [CrossRef] [PubMed]
  105. Bauer, M.; Chicca, A.; Tamborrini, M.; Eisen, D.; Lerner, R.; Lutz, B.; Poetz, O.; Pluschke, G.; Gertsch, J. Identification and quantification of a new family of peptide endocannabinoids (Pepcans) showing negative allosteric modulation at CB1 receptors. J. Biol. Chem. 2012, 287, 36944–36967. [Google Scholar] [CrossRef] [PubMed]
  106. Wei, F.; Zhao, L.; Jing, Y. Signaling molecules targeting cannabinoid receptors: Hemopressin and related peptides. Neuropeptides 2020, 79, 101998. [Google Scholar] [CrossRef] [PubMed]
  107. Fine, P.G.; Rosenfeld, M.J. The endocannabinoid system, cannabinoids, and pain. Rambam Maimonides Med. J. 2013, 4, e0022. [Google Scholar] [CrossRef] [PubMed]
  108. Garcia-Ovejero, D.; Arevalo-Martin, A.; Petrosino, S.; Docagne, F.; Hagen, C.; Bisogno, T.; Watanabe, M.; Guaza, C.; Di Marzo, V.; Molina-Holgado, E. The endocannabinoid system is modulated in response to spinal cord injury in rats. Neurobiol. Dis. 2009, 33, 57–71. [Google Scholar] [CrossRef] [PubMed]
  109. Ahmed, M.M.; Rajpal, S.; Sweeney, C.; Gerovac, T.A.; Allcock, B.; McChesney, S.; Patel, A.U.; Tilghman, J.I.; Miranpuri, G.S.; Resnick, D.K. Cannabinoid subtype-2 receptors modulate the antihyperalgesic effect of WIN 55,212-2 in rats with neuropathic spinal cord injury pain. Spine J. 2010, 10, 1049–1054. [Google Scholar] [CrossRef] [PubMed]
  110. Sagar, D.R.; Gaw, A.G.; Okine, B.N.; Woodhams, S.G.; Wong, A.; Kendall, D.A.; Chapman, V. Dynamic regulation of the endocannabinoid system: Implications for analgesia. Mol. Pain 2009, 5, 59. [Google Scholar] [CrossRef]
  111. Svizenska, I.; Dubovy, P.; Sulcova, A. Cannabinoid receptors 1 and 2 (CB1 and CB2), their distribution, ligands and functional involvement in nervous system structures—A short review. Pharmacol. Biochem. Behav. 2008, 90, 501–511. [Google Scholar] [CrossRef] [PubMed]
  112. Racz, I.; Nadal, X.; Alferink, J.; Banos, J.E.; Rehnelt, J.; Martin, M.; Pintado, B.; Gutierrez-Adan, A.; Sanguino, E.; Manzanares, J.; et al. Crucial role of CB2 cannabinoid receptor in the regulation of central immune responses during neuropathic pain. J. Neurosci. 2008, 28, 12125–12135. [Google Scholar] [CrossRef] [PubMed]
  113. Benito, C.; Nunez, E.; Tolon, R.M.; Carrier, E.J.; Rabano, A.; Hillard, C.J.; Romero, J. Cannabinoid CB2 receptors and fatty acid amide hydrolase are selectively overexpressed in neuritic plaque-associated glia in Alzheimer’s disease brains. J. Neurosci. 2003, 23, 11136–11141. [Google Scholar] [CrossRef]
  114. Van Sickle, M.D.; Duncan, M.; Kingsley, P.J.; Mouihate, A.; Urbani, P.; Mackie, K.; Stella, N.; Makriyannis, A.; Piomelli, D.; Davison, J.S.; et al. Identification and functional characterization of brainstem cannabinoid CB2 receptors. Science 2005, 310, 329–332. [Google Scholar] [CrossRef]
  115. Kibret, B.G.; Ishiguro, H.; Horiuchi, Y.; Onaivi, E.S. New Insights and Potential Therapeutic Targeting of CB2 Cannabinoid Receptors in CNS Disorders. Int. J. Mol. Sci. 2022, 23, 975. [Google Scholar] [CrossRef] [PubMed]
  116. Abuhasira, R.; Shbiro, L.; Landschaft, Y. Medical use of cannabis and cannabinoids containing products—Regulations in Europe and North America. Eur. J. Intern. Med. 2018, 49, 2–6. [Google Scholar] [CrossRef] [PubMed]
  117. Arnold, J.C.; Nation, T.; McGregor, I.S. Prescribing medicinal cannabis. Aust. Prescr. 2020, 43, 152–159. [Google Scholar] [CrossRef] [PubMed]
  118. Ward, S.J.; Lichtman, A.H.; Piomelli, D.; Parker, L.A. Cannabinoids and Cancer Chemotherapy-Associated Adverse Effects. J. Natl. Cancer Inst. Monogr. 2021, 2021, 78–85. [Google Scholar] [CrossRef]
  119. Cunningham, D.; Bradley, C.J.; Forrest, G.J.; Hutcheon, A.W.; Adams, L.; Sneddon, M.; Harding, M.; Kerr, D.J.; Soukop, M.; Kaye, S.B. A randomized trial of oral nabilone and prochlorperazine compared to intravenous metoclopramide and dexamethasone in the treatment of nausea and vomiting induced by chemotherapy regimens containing cisplatin or cisplatin analogues. Eur. J. Cancer Clin. Oncol. 1988, 24, 685–689. [Google Scholar] [CrossRef] [PubMed]
  120. Tsang, C.C.; Giudice, M.G. Nabilone for the Management of Pain. Pharmacotherapy 2016, 36, 273–286. [Google Scholar] [CrossRef] [PubMed]
  121. Wissel, J.; Haydn, T.; Muller, J.; Brenneis, C.; Berger, T.; Poewe, W.; Schelosky, L.D. Low dose treatment with the synthetic cannabinoid Nabilone significantly reduces spasticity-related pain: A double-blind placebo-controlled cross-over trial. J. Neurol. 2006, 253, 1337–1341. [Google Scholar] [CrossRef]
  122. Walitt, B.; Klose, P.; Fitzcharles, M.A.; Phillips, T.; Hauser, W. Cannabinoids for fibromyalgia. Cochrane Database Syst. Rev. 2016, 7, CD011694. [Google Scholar] [CrossRef] [PubMed]
  123. Badowski, M.E.; Yanful, P.K. Dronabinol oral solution in the management of anorexia and weight loss in AIDS and cancer. Ther. Clin. Risk Manag. 2018, 14, 643–651. [Google Scholar] [CrossRef] [PubMed]
  124. Bar-Sela, G.; Zalman, D.; Semenysty, V.; Ballan, E. The Effects of Dosage-Controlled Cannabis Capsules on Cancer-Related Cachexia and Anorexia Syndrome in Advanced Cancer Patients: Pilot Study. Integr. Cancer Ther. 2019, 18, 1534735419881498. [Google Scholar] [CrossRef] [PubMed]
  125. Abu-Sawwa, R.; Stehling, C. Epidiolex (Cannabidiol) Primer: Frequently Asked Questions for Patients and Caregivers. J. Pediatr. Pharmacol. Ther. 2020, 25, 75–77. [Google Scholar] [CrossRef]
  126. Sekar, K.; Pack, A. Epidiolex as adjunct therapy for treatment of refractory epilepsy: A comprehensive review with a focus on adverse effects. F1000Res 2019, 8, F1000 Faculty Rev-234. [Google Scholar] [CrossRef] [PubMed]
  127. Conte, A.; Vila Silvan, C. Review of Available Data for the Efficacy and Effectiveness of Nabiximols Oromucosal Spray (Sativex(R)) in Multiple Sclerosis Patients with Moderate to Severe Spasticity. Neurodegener. Dis. 2021, 21, 55–62. [Google Scholar] [CrossRef] [PubMed]
  128. Meuth, S.G.; Henze, T.; Essner, U.; Trompke, C.; Vila Silvan, C. Tetrahydrocannabinol and cannabidiol oromucosal spray in resistant multiple sclerosis spasticity: Consistency of response across subgroups from the SAVANT randomized clinical trial. Int. J. Neurosci. 2020, 130, 1199–1205. [Google Scholar] [CrossRef]
  129. Perez, J.; Ribera, M.V. Managing neuropathic pain with Sativex: A review of its pros and cons. Expert Opin. Pharmacother. 2008, 9, 1189–1195. [Google Scholar] [CrossRef] [PubMed]
  130. Tonstad, S. Rimonabant: A cannabinoid receptor blocker for the treatment of metabolic and cardiovascular risk factors. Nutr. Metab. Cardiovasc. Dis. 2006, 16, 156–162. [Google Scholar] [CrossRef] [PubMed]
  131. Wierzbicki, A.S. Rimonabant: Endocannabinoid inhibition for the metabolic syndrome. Int. J. Clin. Pract. 2006, 60, 1697–1706. [Google Scholar] [CrossRef] [PubMed]
  132. Almeida, T.F.; Roizenblatt, S.; Tufik, S. Afferent pain pathways: A neuroanatomical review. Brain Res. 2004, 1000, 40–56. [Google Scholar] [CrossRef] [PubMed]
  133. D’Mello, R.; Dickenson, A.H. Spinal cord mechanisms of pain. Br. J. Anaesth. 2008, 101, 8–16. [Google Scholar] [CrossRef] [PubMed]
  134. Basbaum, A.I.; Bautista, D.M.; Scherrer, G.; Julius, D. Cellular and molecular mechanisms of pain. Cell 2009, 139, 267–284. [Google Scholar] [CrossRef] [PubMed]
  135. Ji, R.R.; Nackley, A.; Huh, Y.; Terrando, N.; Maixner, W. Neuroinflammation and Central Sensitization in Chronic and Widespread Pain. Anesthesiology 2018, 129, 343–366. [Google Scholar] [CrossRef] [PubMed]
  136. Millan, M.J. The induction of pain: An integrative review. Prog. Neurobiol. 1999, 57, 1–164. [Google Scholar] [CrossRef]
  137. Chapman, C.R.; Tuckett, R.P.; Song, C.W. Pain and stress in a systems perspective: Reciprocal neural, endocrine, and immune interactions. J. Pain 2008, 9, 122–145. [Google Scholar] [CrossRef] [PubMed]
  138. Rittner, H.L.; Brack, A.; Stein, C. Pain and the immune system. Br. J. Anaesth. 2008, 101, 40–44. [Google Scholar] [CrossRef] [PubMed]
  139. Ren, K.; Dubner, R. Interactions between the immune and nervous systems in pain. Nat. Med. 2010, 16, 1267–1276. [Google Scholar] [CrossRef] [PubMed]
  140. Pogatzki-Zahn, E.M.; Segelcke, D.; Schug, S.A. Postoperative pain-from mechanisms to treatment. Pain Rep. 2017, 2, e588. [Google Scholar] [CrossRef] [PubMed]
  141. Hillard, C.J.; Beatka, M.; Sarvaideo, J. Endocannabinoid Signaling and the Hypothalamic-Pituitary-Adrenal Axis. Compr. Physiol. 2016, 7, 1–15. [Google Scholar] [CrossRef]
  142. Imbe, H.; Iwai-Liao, Y.; Senba, E. Stress-induced hyperalgesia: Animal models and putative mechanisms. Front. Biosci. 2006, 11, 2179–2192. [Google Scholar] [CrossRef]
  143. Gangadharan, V.; Kuner, R. Pain hypersensitivity mechanisms at a glance. Dis. Models Mech. 2013, 6, 889–895. [Google Scholar] [CrossRef]
  144. Latremoliere, A.; Woolf, C.J. Central sensitization: A generator of pain hypersensitivity by central neural plasticity. J. Pain 2009, 10, 895–926. [Google Scholar] [CrossRef]
  145. Costigan, M.; Scholz, J.; Woolf, C.J. Neuropathic pain: A maladaptive response of the nervous system to damage. Annu. Rev. Neurosci. 2009, 32, 1–32. [Google Scholar] [CrossRef] [PubMed]
  146. Besson, J.M. The neurobiology of pain. Lancet 1999, 353, 1610–1615. [Google Scholar] [CrossRef] [PubMed]
  147. Levy, D. Endogenous mechanisms underlying the activation and sensitization of meningeal nociceptors: The role of immuno-vascular interactions and cortical spreading depression. Curr. Pain Headache Rep. 2012, 16, 270–277. [Google Scholar] [CrossRef] [PubMed]
  148. Torta, D.M.E.; Van Den Broeke, E.N.; Filbrich, L.; Jacob, B.; Lambert, J.; Mouraux, A. Intense pain influences the cortical processing of visual stimuli projected onto the sensitized skin. Pain 2017, 158, 691–697. [Google Scholar] [CrossRef] [PubMed]
  149. Chapman, C.R.; Vierck, C.J. The Transition of Acute Postoperative Pain to Chronic Pain: An Integrative Overview of Research on Mechanisms. J. Pain 2017, 18, 359.e1–359.e38. [Google Scholar] [CrossRef] [PubMed]
  150. Ellis, A.; Bennett, D.L. Neuroinflammation and the generation of neuropathic pain. Br. J. Anaesth. 2013, 111, 26–37. [Google Scholar] [CrossRef] [PubMed]
  151. Matsuda, M.; Huh, Y.; Ji, R.R. Roles of inflammation, neurogenic inflammation, and neuroinflammation in pain. J. Anesth. 2019, 33, 131–139. [Google Scholar] [CrossRef] [PubMed]
  152. Ho, I.H.T.; Chan, M.T.V.; Wu, W.K.K.; Liu, X. Spinal microglia-neuron interactions in chronic pain. J. Leukoc. Biol. 2020, 108, 1575–1592. [Google Scholar] [CrossRef]
  153. Zahn, P.K.; Brennan, T.J. Lack of effect of intrathecally administered N-methyl-D-aspartate receptor antagonists in a rat model for postoperative pain. Anesthesiology 1998, 88, 143–156. [Google Scholar] [CrossRef] [PubMed]
  154. Zahn, P.K.; Umali, E.; Brennan, T.J. Intrathecal non-NMDA excitatory amino acid receptor antagonists inhibit pain behaviors in a rat model of postoperative pain. Pain 1998, 74, 213–223. [Google Scholar] [CrossRef] [PubMed]
  155. Yamamoto, T.; Sakashita, Y. The role of the spinal opioid receptor like1 receptor, the NK-1 receptor, and cyclooxygenase-2 in maintaining postoperative pain in the rat. Anesth. Analg. 1999, 89, 1203–1208. [Google Scholar] [CrossRef] [PubMed]
  156. Zhu, X.; Conklin, D.; Eisenach, J.C. Cyclooxygenase-1 in the spinal cord plays an important role in postoperative pain. Pain 2003, 104, 15–23. [Google Scholar] [CrossRef] [PubMed]
  157. Porreca, F.; Ossipov, M.H.; Gebhart, G.F. Chronic pain and medullary descending facilitation. Trends Neurosci. 2002, 25, 319–325. [Google Scholar] [CrossRef] [PubMed]
  158. Pogatzki, E.M.; Urban, M.O.; Brennan, T.J.; Gebhart, G.F. Role of the rostral medial medulla in the development of primary and secondary hyperalgesia after incision in the rat. Anesthesiology 2002, 96, 1153–1160. [Google Scholar] [CrossRef] [PubMed]
  159. Obata, H.; Eisenach, J.C.; Hussain, H.; Bynum, T.; Vincler, M. Spinal glial activation contributes to postoperative mechanical hypersensitivity in the rat. J. Pain 2006, 7, 816–822. [Google Scholar] [CrossRef] [PubMed]
  160. Wen, Y.R.; Tan, P.H.; Cheng, J.K.; Liu, Y.C.; Ji, R.R. Microglia: A promising target for treating neuropathic and postoperative pain, and morphine tolerance. J. Formos. Med. Assoc. 2011, 110, 487–494. [Google Scholar] [CrossRef] [PubMed]
  161. Tang, J.; Bair, M.; Descalzi, G. Reactive Astrocytes: Critical Players in the Development of Chronic Pain. Front. Psychiatry 2021, 12, 682056. [Google Scholar] [CrossRef]
  162. Maldonado, R.; Banos, J.E.; Cabanero, D. The endocannabinoid system and neuropathic pain. Pain 2016, 157 (Suppl. S1), S23–S32. [Google Scholar] [CrossRef] [PubMed]
  163. Heifets, B.D.; Castillo, P.E. Endocannabinoid signaling and long-term synaptic plasticity. Annu. Rev. Physiol. 2009, 71, 283–306. [Google Scholar] [CrossRef] [PubMed]
  164. Di Marzo, V.; Petrosino, S. Endocannabinoids and the regulation of their levels in health and disease. Curr. Opin. Lipidol. 2007, 18, 129–140. [Google Scholar] [CrossRef] [PubMed]
  165. Hill, M.N.; McEwen, B.S. Involvement of the endocannabinoid system in the neurobehavioural effects of stress and glucocorticoids. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2010, 34, 791–797. [Google Scholar] [CrossRef] [PubMed]
  166. Gorzalka, B.B.; Hill, M.N.; Hillard, C.J. Regulation of endocannabinoid signaling by stress: Implications for stress-related affective disorders. Neurosci. Biobehav. Rev. 2008, 32, 1152–1160. [Google Scholar] [CrossRef] [PubMed]
  167. Bradshaw, H.B.; Walker, J.M. The expanding field of cannabimimetic and related lipid mediators. Br. J. Pharmacol. 2005, 144, 459–465. [Google Scholar] [CrossRef] [PubMed]
  168. Hohmann, A.G.; Suplita, R.L., 2nd. Endocannabinoid mechanisms of pain modulation. AAPS J. 2006, 8, E693–E708. [Google Scholar] [CrossRef] [PubMed]
  169. Murphy, L.L.; Munoz, R.M.; Adrian, B.A.; Villanua, M.A. Function of cannabinoid receptors in the neuroendocrine regulation of hormone secretion. Neurobiol. Dis. 1998, 5, 432–446. [Google Scholar] [CrossRef] [PubMed]
  170. Pertwee, R.G. Cannabinoid receptor ligands: Clinical and neuropharmacological considerations, relevant to future drug discovery and development. Expert Opin. Investig. Drugs 2000, 9, 1553–1571. [Google Scholar] [CrossRef] [PubMed]
  171. Wootten, D.; Christopoulos, A.; Marti-Solano, M.; Babu, M.M.; Sexton, P.M. Mechanisms of signalling and biased agonism in G protein-coupled receptors. Nat. Rev. Mol. Cell Biol. 2018, 19, 638–653. [Google Scholar] [CrossRef] [PubMed]
  172. Nevalainen, T.; Irving, A.J. GPR55, a lysophosphatidylinositol receptor with cannabinoid sensitivity? Curr. Top. Med. Chem. 2010, 10, 799–813. [Google Scholar] [CrossRef]
  173. Ross, R.A. The enigmatic pharmacology of GPR55. Trends Pharmacol. Sci. 2009, 30, 156–163. [Google Scholar] [CrossRef] [PubMed]
  174. Lauckner, J.E.; Jensen, J.B.; Chen, H.Y.; Lu, H.C.; Hille, B.; Mackie, K. GPR55 is a cannabinoid receptor that increases intracellular calcium and inhibits M current. Proc. Natl. Acad. Sci. USA 2008, 105, 2699–2704. [Google Scholar] [CrossRef] [PubMed]
  175. Ryberg, E.; Larsson, N.; Sjogren, S.; Hjorth, S.; Hermansson, N.O.; Leonova, J.; Elebring, T.; Nilsson, K.; Drmota, T.; Greasley, P.J. The orphan receptor GPR55 is a novel cannabinoid receptor. Br. J. Pharmacol. 2007, 152, 1092–1101. [Google Scholar] [CrossRef] [PubMed]
  176. Irving, A.; Abdulrazzaq, G.; Chan, S.L.F.; Penman, J.; Harvey, J.; Alexander, S.P.H. Cannabinoid Receptor-Related Orphan G Protein-Coupled Receptors. Adv. Pharmacol. 2017, 80, 223–247. [Google Scholar] [CrossRef] [PubMed]
  177. LoVerme, J.; Russo, R.; La Rana, G.; Fu, J.; Farthing, J.; Mattace-Raso, G.; Meli, R.; Hohmann, A.; Calignano, A.; Piomelli, D. Rapid broad-spectrum analgesia through activation of peroxisome proliferator-activated receptor-alpha. J. Pharmacol. Exp. Ther. 2006, 319, 1051–1061. [Google Scholar] [CrossRef] [PubMed]
  178. Guzman, M.; Lo Verme, J.; Fu, J.; Oveisi, F.; Blazquez, C.; Piomelli, D. Oleoylethanolamide stimulates lipolysis by activating the nuclear receptor peroxisome proliferator-activated receptor alpha (PPAR-alpha). J. Biol. Chem. 2004, 279, 27849–27854. [Google Scholar] [CrossRef] [PubMed]
  179. O’Sullivan, S.E.; Kendall, D.A. Cannabinoid activation of peroxisome proliferator-activated receptors: Potential for modulation of inflammatory disease. Immunobiology 2010, 215, 611–616. [Google Scholar] [CrossRef] [PubMed]
  180. Matsuda, L.A. Molecular aspects of cannabinoid receptors. Crit. Rev. Neurobiol. 1997, 11, 143–166. [Google Scholar] [CrossRef] [PubMed]
  181. Sun, Y.; Bennett, A. Cannabinoids: A new group of agonists of PPARs. PPAR Res. 2007, 2007, 23513. [Google Scholar] [CrossRef] [PubMed]
  182. Singh Tahim, A.; Santha, P.; Nagy, I. Inflammatory mediators convert anandamide into a potent activator of the vanilloid type 1 transient receptor potential receptor in nociceptive primary sensory neurons. Neuroscience 2005, 136, 539–548. [Google Scholar] [CrossRef] [PubMed]
  183. Holzer, P. The pharmacological challenge to tame the transient receptor potential vanilloid-1 (TRPV1) nocisensor. Br. J. Pharmacol. 2008, 155, 1145–1162. [Google Scholar] [CrossRef] [PubMed]
  184. Lutz, B. Neurobiology of cannabinoid receptor signaling. Dialogues Clin. Neurosci. 2020, 22, 207–222. [Google Scholar] [CrossRef] [PubMed]
  185. Deadwyler, S.A.; Hampson, R.E.; Bennett, B.A.; Edwards, T.A.; Mu, J.; Pacheco, M.A.; Ward, S.J.; Childers, S.R. Cannabinoids modulate potassium current in cultured hippocampal neurons. Recept. Channels 1993, 1, 121–134. [Google Scholar] [PubMed]
  186. Gomez del Pulgar, T.; Velasco, G.; Guzman, M. The CB1 cannabinoid receptor is coupled to the activation of protein kinase B/Akt. Biochem. J. 2000, 347, 369–373. [Google Scholar] [CrossRef] [PubMed]
  187. Galve-Roperh, I.; Rueda, D.; Gomez del Pulgar, T.; Velasco, G.; Guzman, M. Mechanism of extracellular signal-regulated kinase activation by the CB1 cannabinoid receptor. Mol. Pharmacol. 2002, 62, 1385–1392. [Google Scholar] [CrossRef] [PubMed]
  188. Molina-Holgado, F.; Pinteaux, E.; Heenan, L.; Moore, J.D.; Rothwell, N.J.; Gibson, R.M. Neuroprotective effects of the synthetic cannabinoid HU-210 in primary cortical neurons are mediated by phosphatidylinositol 3-kinase/AKT signaling. Mol. Cell. Neurosci. 2005, 28, 189–194. [Google Scholar] [CrossRef] [PubMed]
  189. Karanian, D.A.; Brown, Q.B.; Makriyannis, A.; Kosten, T.A.; Bahr, B.A. Dual modulation of endocannabinoid transport and fatty acid amide hydrolase protects against excitotoxicity. J. Neurosci. 2005, 25, 7813–7820. [Google Scholar] [CrossRef] [PubMed]
  190. Karanian, D.A.; Karim, S.L.; Wood, J.T.; Williams, J.S.; Lin, S.; Makriyannis, A.; Bahr, B.A. Endocannabinoid enhancement protects against kainic acid-induced seizures and associated brain damage. J. Pharmacol. Exp. Ther. 2007, 322, 1059–1066. [Google Scholar] [CrossRef] [PubMed]
  191. Hajos, N.; Katona, I.; Naiem, S.S.; MacKie, K.; Ledent, C.; Mody, I.; Freund, T.F. Cannabinoids inhibit hippocampal GABAergic transmission and network oscillations. Eur. J. Neurosci. 2000, 12, 3239–3249. [Google Scholar] [CrossRef] [PubMed]
  192. Kreitzer, A.C.; Regehr, W.G. Retrograde inhibition of presynaptic calcium influx by endogenous cannabinoids at excitatory synapses onto Purkinje cells. Neuron 2001, 29, 717–727. [Google Scholar] [CrossRef] [PubMed]
  193. Ohno-Shosaku, T.; Maejima, T.; Kano, M. Endogenous cannabinoids mediate retrograde signals from depolarized postsynaptic neurons to presynaptic terminals. Neuron 2001, 29, 729–738. [Google Scholar] [CrossRef] [PubMed]
  194. Wilson, R.I.; Kunos, G.; Nicoll, R.A. Presynaptic specificity of endocannabinoid signaling in the hippocampus. Neuron 2001, 31, 453–462. [Google Scholar] [CrossRef] [PubMed]
  195. Ozaita, A.; Puighermanal, E.; Maldonado, R. Regulation of PI3K/Akt/GSK-3 pathway by cannabinoids in the brain. J. Neurochem. 2007, 102, 1105–1114. [Google Scholar] [CrossRef] [PubMed]
  196. Price, T.J.; Patwardhan, A.; Akopian, A.N.; Hargreaves, K.M.; Flores, C.M. Modulation of trigeminal sensory neuron activity by the dual cannabinoid-vanilloid agonists anandamide, N-arachidonoyl-dopamine and arachidonyl-2-chloroethylamide. Br. J. Pharmacol. 2004, 141, 1118–1130. [Google Scholar] [CrossRef]
  197. Fischbach, T.; Greffrath, W.; Nawrath, H.; Treede, R.D. Effects of anandamide and noxious heat on intracellular calcium concentration in nociceptive drg neurons of rats. J. Neurophysiol. 2007, 98, 929–938. [Google Scholar] [CrossRef] [PubMed]
  198. Lee, M.G.; Weinreich, D.; Undem, B.J. Effect of olvanil and anandamide on vagal C-fiber subtypes in guinea pig lung. Br. J. Pharmacol. 2005, 146, 596–603. [Google Scholar] [CrossRef] [PubMed]
  199. Patwardhan, A.M.; Jeske, N.A.; Price, T.J.; Gamper, N.; Akopian, A.N.; Hargreaves, K.M. The cannabinoid WIN 55,212-2 inhibits transient receptor potential vanilloid 1 (TRPV1) and evokes peripheral antihyperalgesia via calcineurin. Proc. Natl. Acad. Sci. USA 2006, 103, 11393–11398. [Google Scholar] [CrossRef] [PubMed]
  200. Price, T.J.; Patwardhan, A.; Akopian, A.N.; Hargreaves, K.M.; Flores, C.M. Cannabinoid receptor-independent actions of the aminoalkylindole WIN 55,212-2 on trigeminal sensory neurons. Br. J. Pharmacol. 2004, 142, 257–266. [Google Scholar] [CrossRef] [PubMed]
  201. McCarberg, B.H.; Barkin, R.L. The future of cannabinoids as analgesic agents: A pharmacologic, pharmacokinetic, and pharmacodynamic overview. Am. J. Ther. 2007, 14, 475–483. [Google Scholar] [CrossRef] [PubMed]
  202. Mbvundula, E.C.; Rainsford, K.D.; Bunning, R.A. Cannabinoids in pain and inflammation. Inflammopharmacology 2004, 12, 99–114. [Google Scholar] [CrossRef] [PubMed]
  203. Jeske, N.A.; Patwardhan, A.M.; Gamper, N.; Price, T.J.; Akopian, A.N.; Hargreaves, K.M. Cannabinoid WIN 55,212-2 regulates TRPV1 phosphorylation in sensory neurons. J. Biol. Chem. 2006, 281, 32879–32890. [Google Scholar] [CrossRef] [PubMed]
  204. Jordt, S.E.; Bautista, D.M.; Chuang, H.H.; McKemy, D.D.; Zygmunt, P.M.; Hogestatt, E.D.; Meng, I.D.; Julius, D. Mustard oils and cannabinoids excite sensory nerve fibres through the TRP channel ANKTM1. Nature 2004, 427, 260–265. [Google Scholar] [CrossRef] [PubMed]
  205. Liu, L.; Lo, Y.; Chen, I.; Simon, S.A. The responses of rat trigeminal ganglion neurons to capsaicin and two nonpungent vanilloid receptor agonists, olvanil and glyceryl nonamide. J. Neurosci. 1997, 17, 4101–4111. [Google Scholar] [CrossRef] [PubMed]
  206. Ishac, E.J.; Jiang, L.; Lake, K.D.; Varga, K.; Abood, M.E.; Kunos, G. Inhibition of exocytotic noradrenaline release by presynaptic cannabinoid CB1 receptors on peripheral sympathetic nerves. Br. J. Pharmacol. 1996, 118, 2023–2028. [Google Scholar] [CrossRef] [PubMed]
  207. Kathmann, M.; Bauer, U.; Schlicker, E.; Gothert, M. Cannabinoid CB1 receptor-mediated inhibition of NMDA- and kainate-stimulated noradrenaline and dopamine release in the brain. Naunyn Schmiedeberg’s Arch. Pharmacol. 1999, 359, 466–470. [Google Scholar] [CrossRef] [PubMed]
  208. Nakazi, M.; Bauer, U.; Nickel, T.; Kathmann, M.; Schlicker, E. Inhibition of serotonin release in the mouse brain via presynaptic cannabinoid CB1 receptors. Naunyn Schmiedeberg’s Arch. Pharmacol. 2000, 361, 19–24. [Google Scholar] [CrossRef] [PubMed]
  209. Wilson, R.I.; Nicoll, R.A. Endogenous cannabinoids mediate retrograde signalling at hippocampal synapses. Nature 2001, 410, 588–592. [Google Scholar] [CrossRef] [PubMed]
  210. Shen, M.; Piser, T.M.; Seybold, V.S.; Thayer, S.A. Cannabinoid receptor agonists inhibit glutamatergic synaptic transmission in rat hippocampal cultures. J. Neurosci. 1996, 16, 4322–4334. [Google Scholar] [CrossRef] [PubMed]
  211. Barann, M.; Molderings, G.; Bruss, M.; Bonisch, H.; Urban, B.W.; Gothert, M. Direct inhibition by cannabinoids of human 5-HT3A receptors: Probable involvement of an allosteric modulatory site. Br. J. Pharmacol. 2002, 137, 589–596. [Google Scholar] [CrossRef] [PubMed]
  212. Weidenfeld, J.; Feldman, S.; Mechoulam, R. Effect of the brain constituent anandamide, a cannabinoid receptor agonist, on the hypothalamo-pituitary-adrenal axis in the rat. Neuroendocrinology 1994, 59, 110–112. [Google Scholar] [CrossRef] [PubMed]
  213. Steiner, M.A.; Wotjak, C.T. Role of the endocannabinoid system in regulation of the hypothalamic-pituitary-adrenocortical axis. Prog. Brain Res. 2008, 170, 397–432. [Google Scholar] [CrossRef]
  214. Iversen, L.; Chapman, V. Cannabinoids: A real prospect for pain relief? Curr. Opin. Pharmacol. 2002, 2, 50–55. [Google Scholar] [CrossRef] [PubMed]
  215. Welch, S.P.; Thomas, C.; Patrick, G.S. Modulation of cannabinoid-induced antinociception after intracerebroventricular versus intrathecal administration to mice: Possible mechanisms for interaction with morphine. J. Pharmacol. Exp. Ther. 1995, 272, 310–321. [Google Scholar] [PubMed]
  216. Martin, W.J.; Lai, N.K.; Patrick, S.L.; Tsou, K.; Walker, J.M. Antinociceptive actions of cannabinoids following intraventricular administration in rats. Brain Res. 1993, 629, 300–304. [Google Scholar] [CrossRef] [PubMed]
  217. Hohmann, A.G.; Tsou, K.; Walker, J.M. Cannabinoid suppression of noxious heat-evoked activity in wide dynamic range neurons in the lumbar dorsal horn of the rat. J. Neurophysiol. 1999, 81, 575–583. [Google Scholar] [CrossRef] [PubMed]
  218. Raffa, R.B.; Stone, D.J., Jr.; Hipp, S.J. Differential cholera-toxin sensitivity of supraspinal antinociception induced by the cannabinoid agonists delta9-THC, WIN 55,212-2 and anandamide in mice. Neurosci. Lett. 1999, 263, 29–32. [Google Scholar] [CrossRef] [PubMed]
  219. Strangman, N.M.; Patrick, S.L.; Hohmann, A.G.; Tsou, K.; Walker, J.M. Evidence for a role of endogenous cannabinoids in the modulation of acute and tonic pain sensitivity. Brain Res. 1998, 813, 323–328. [Google Scholar] [CrossRef] [PubMed]
  220. Welch, S.P.; Huffman, J.W.; Lowe, J. Differential blockade of the antinociceptive effects of centrally administered cannabinoids by SR141716A. J. Pharmacol. Exp. Ther. 1998, 286, 1301–1308. [Google Scholar] [PubMed]
  221. Walker, J.M.; Huang, S.M.; Strangman, N.M.; Tsou, K.; Sanudo-Pena, M.C. Pain modulation by release of the endogenous cannabinoid anandamide. Proc. Natl. Acad. Sci. USA 1999, 96, 12198–12203. [Google Scholar] [CrossRef] [PubMed]
  222. Richardson, J.D.; Aanonsen, L.; Hargreaves, K.M. Antihyperalgesic effects of spinal cannabinoids. Eur. J. Pharmacol. 1998, 345, 145–153. [Google Scholar] [CrossRef]
  223. Morisset, V.; Ahluwalia, J.; Nagy, I.; Urban, L. Possible mechanisms of cannabinoid-induced antinociception in the spinal cord. Eur. J. Pharmacol. 2001, 429, 93–100. [Google Scholar] [CrossRef]
  224. Hohmann, A.G.; Tsou, K.; Walker, J.M. Cannabinoid modulation of wide dynamic range neurons in the lumbar dorsal horn of the rat by spinally administered WIN55,212-2. Neurosci. Lett. 1998, 257, 119–122. [Google Scholar] [CrossRef] [PubMed]
  225. Richardson, J.D.; Aanonsen, L.; Hargreaves, K.M. Hypoactivity of the spinal cannabinoid system results in NMDA-dependent hyperalgesia. J. Neurosci. 1998, 18, 451–457. [Google Scholar] [CrossRef] [PubMed]
  226. Romero-Sandoval, A.; Nutile-McMenemy, N.; DeLeo, J.A. Spinal microglial and perivascular cell cannabinoid receptor type 2 activation reduces behavioral hypersensitivity without tolerance after peripheral nerve injury. Anesthesiology 2008, 108, 722–734. [Google Scholar] [CrossRef] [PubMed]
  227. Xu, K.; Wu, Y.; Tian, Z.; Xu, Y.; Wu, C.; Wang, Z. Microglial Cannabinoid CB2 Receptors in Pain Modulation. Int. J. Mol. Sci. 2023, 24, 2348. [Google Scholar] [CrossRef] [PubMed]
  228. Lichtman, A.H.; Martin, B.R. Cannabinoid-induced antinociception is mediated by a spinal alpha 2-noradrenergic mechanism. Brain Res. 1991, 559, 309–314. [Google Scholar] [CrossRef] [PubMed]
  229. Pugh, G., Jr.; Smith, P.B.; Dombrowski, D.S.; Welch, S.P. The role of endogenous opioids in enhancing the antinociception produced by the combination of delta9-tetrahydrocannabinol and morphine in the spinal cord. J. Pharmacol. Exp. Ther. 1996, 279, 608–616. [Google Scholar] [PubMed]
  230. Fox, A.; Kesingland, A.; Gentry, C.; McNair, K.; Patel, S.; Urban, L.; James, I. The role of central and peripheral Cannabinoid1 receptors in the antihyperalgesic activity of cannabinoids in a model of neuropathic pain. Pain 2001, 92, 91–100. [Google Scholar] [CrossRef] [PubMed]
  231. Malan, T.P., Jr.; Ibrahim, M.M.; Lai, J.; Vanderah, T.W.; Makriyannis, A.; Porreca, F. CB2 cannabinoid receptor agonists: Pain relief without psychoactive effects? Curr. Opin. Pharmacol. 2003, 3, 62–67. [Google Scholar] [CrossRef] [PubMed]
  232. Hanus, L.; Breuer, A.; Tchilibon, S.; Shiloah, S.; Goldenberg, D.; Horowitz, M.; Pertwee, R.G.; Ross, R.A.; Mechoulam, R.; Fride, E. HU-308: A specific agonist for CB2, a peripheral cannabinoid receptor. Proc. Natl. Acad. Sci. USA 1999, 96, 14228–14233. [Google Scholar] [CrossRef] [PubMed]
  233. Malan, T.P., Jr.; Ibrahim, M.M.; Vanderah, T.W.; Makriyannis, A.; Porreca, F. Inhibition of pain responses by activation of CB2 cannabinoid receptors. Chem. Phys. Lipids 2002, 121, 191–200. [Google Scholar] [CrossRef]
  234. Ross, R.A. Anandamide and vanilloid TRPV1 receptors. Br. J. Pharmacol. 2003, 140, 790–801. [Google Scholar] [CrossRef]
  235. Starowicz, K.; Finn, D.P. Cannabinoids and Pain: Sites and Mechanisms of Action. Adv. Pharmacol. 2017, 80, 437–475. [Google Scholar] [CrossRef] [PubMed]
  236. Durnett-Richardson, J. Cannabinoids Modulate Pain by Multiple Mechanisms of Action. J. Pain 2000, 1, 2–14. [Google Scholar] [CrossRef]
  237. Bahr, B.A.; Karanian, D.A.; Makanji, S.S.; Makriyannis, A. Targeting the endocannabinoid system in treating brain disorders. Expert Opin. Investig. Drugs 2006, 15, 351–365. [Google Scholar] [CrossRef] [PubMed]
  238. Galve-Roperh, I.; Aguado, T.; Palazuelos, J.; Guzman, M. Mechanisms of control of neuron survival by the endocannabinoid system. Curr. Pharm. Des. 2008, 14, 2279–2288. [Google Scholar] [CrossRef] [PubMed]
  239. Wallace, M.J.; Blair, R.E.; Falenski, K.W.; Martin, B.R.; DeLorenzo, R.J. The endogenous cannabinoid system regulates seizure frequency and duration in a model of temporal lobe epilepsy. J. Pharmacol. Exp. Ther. 2003, 307, 129–137. [Google Scholar] [CrossRef] [PubMed]
  240. Khaspekov, L.G.; Brenz Verca, M.S.; Frumkina, L.E.; Hermann, H.; Marsicano, G.; Lutz, B. Involvement of brain-derived neurotrophic factor in cannabinoid receptor-dependent protection against excitotoxicity. Eur. J. Neurosci. 2004, 19, 1691–1698. [Google Scholar] [CrossRef] [PubMed]
  241. Karanian, D.A.; Brown, Q.B.; Makriyannis, A.; Bahr, B.A. Blocking cannabinoid activation of FAK and ERK1/2 compromises synaptic integrity in hippocampus. Eur. J. Pharmacol. 2005, 508, 47–56. [Google Scholar] [CrossRef] [PubMed]
  242. Schomacher, M.; Muller, H.D.; Sommer, C.; Schwab, S.; Schabitz, W.R. Endocannabinoids mediate neuroprotection after transient focal cerebral ischemia. Brain Res. 2008, 1240, 213–220. [Google Scholar] [CrossRef] [PubMed]
  243. Hohmann, A.G.; Suplita, R.L.; Bolton, N.M.; Neely, M.H.; Fegley, D.; Mangieri, R.; Krey, J.F.; Walker, J.M.; Holmes, P.V.; Crystal, J.D.; et al. An endocannabinoid mechanism for stress-induced analgesia. Nature 2005, 435, 1108–1112. [Google Scholar] [CrossRef] [PubMed]
  244. Meng, I.D.; Johansen, J.P. Antinociception and modulation of rostral ventromedial medulla neuronal activity by local microinfusion of a cannabinoid receptor agonist. Neuroscience 2004, 124, 685–693. [Google Scholar] [CrossRef] [PubMed]
  245. Finn, D.P.; Jhaveri, M.D.; Beckett, S.R.; Roe, C.H.; Kendall, D.A.; Marsden, C.A.; Chapman, V. Effects of direct periaqueductal grey administration of a cannabinoid receptor agonist on nociceptive and aversive responses in rats. Neuropharmacology 2003, 45, 594–604. [Google Scholar] [CrossRef] [PubMed]
  246. Pertwee, R.G. Emerging strategies for exploiting cannabinoid receptor agonists as medicines. Br. J. Pharmacol. 2009, 156, 397–411. [Google Scholar] [CrossRef] [PubMed]
  247. Di Marzo, V. Targeting the endocannabinoid system: To enhance or reduce? Nat. Rev. Drug Discov. 2008, 7, 438–455. [Google Scholar] [CrossRef] [PubMed]
  248. Izzo, A.A.; Camilleri, M. Emerging role of cannabinoids in gastrointestinal and liver diseases: Basic and clinical aspects. Gut 2008, 57, 1140–1155. [Google Scholar] [CrossRef]
  249. Kearn, C.S.; Blake-Palmer, K.; Daniel, E.; Mackie, K.; Glass, M. Concurrent stimulation of cannabinoid CB1 and dopamine D2 receptors enhances heterodimer formation: A mechanism for receptor cross-talk? Mol. Pharmacol. 2005, 67, 1697–1704. [Google Scholar] [CrossRef] [PubMed]
  250. Carriba, P.; Ortiz, O.; Patkar, K.; Justinova, Z.; Stroik, J.; Themann, A.; Muller, C.; Woods, A.S.; Hope, B.T.; Ciruela, F.; et al. Striatal adenosine A2A and cannabinoid CB1 receptors form functional heteromeric complexes that mediate the motor effects of cannabinoids. Neuropsychopharmacology 2007, 32, 2249–2259. [Google Scholar] [CrossRef]
  251. Pertwee, R.G. The therapeutic potential of drugs that target cannabinoid receptors or modulate the tissue levels or actions of endocannabinoids. AAPS J. 2005, 7, E625–E654. [Google Scholar] [CrossRef] [PubMed]
  252. Di Marzo, V. Endocannabinoids: Synthesis and degradation. Rev. Physiol. Biochem. Pharmacol. 2008, 160, 1–24. [Google Scholar] [CrossRef] [PubMed]
  253. Katona, I.; Freund, T.F. Endocannabinoid signaling as a synaptic circuit breaker in neurological disease. Nat. Med. 2008, 14, 923–930. [Google Scholar] [CrossRef] [PubMed]
  254. Margraf, A.; Ludwig, N.; Zarbock, A.; Rossaint, J. Systemic Inflammatory Response Syndrome After Surgery: Mechanisms and Protection. Anesth. Analg. 2020, 131, 1693–1707. [Google Scholar] [CrossRef] [PubMed]
  255. Richebe, P.; Capdevila, X.; Rivat, C. Persistent Postsurgical Pain: Pathophysiology and Preventative Pharmacologic Considerations. Anesthesiology 2018, 129, 590–607. [Google Scholar] [CrossRef] [PubMed]
  256. Lavand’homme, P. The progression from acute to chronic pain. Curr. Opin. Anesthesiol. 2011, 24, 545–550. [Google Scholar] [CrossRef]
  257. Bouhassira, D.; Lanteri-Minet, M.; Attal, N.; Laurent, B.; Touboul, C. Prevalence of chronic pain with neuropathic characteristics in the general population. Pain 2008, 136, 380–387. [Google Scholar] [CrossRef] [PubMed]
  258. Rosenberger, D.C.; Pogatzki-Zahn, E.M. Chronic post-surgical pain-update on incidence, risk factors and preventive treatment options. BJA Educ. 2022, 22, 190–196. [Google Scholar] [CrossRef] [PubMed]
  259. Kehlet, H.; Jensen, T.S.; Woolf, C.J. Persistent postsurgical pain: Risk factors and prevention. Lancet 2006, 367, 1618–1625. [Google Scholar] [CrossRef]
  260. Fowler, C.J. Possible involvement of the endocannabinoid system in the actions of three clinically used drugs. Trends Pharmacol. Sci. 2004, 25, 59–61. [Google Scholar] [CrossRef] [PubMed]
  261. Smith, P.B.; Compton, D.R.; Welch, S.P.; Razdan, R.K.; Mechoulam, R.; Martin, B.R. The pharmacological activity of anandamide, a putative endogenous cannabinoid, in mice. J. Pharmacol. Exp. Ther. 1994, 270, 219–227. [Google Scholar] [PubMed]
  262. Calignano, A.; La Rana, G.; Giuffrida, A.; Piomelli, D. Control of pain initiation by endogenous cannabinoids. Nature 1998, 394, 277–281. [Google Scholar] [CrossRef] [PubMed]
  263. Lee, M.T.; Mackie, K.; Chiou, L.C. Alternative pain management via endocannabinoids in the time of the opioid epidemic: Peripheral neuromodulation and pharmacological interventions. Br. J. Pharmacol. 2023, 180, 894–909. [Google Scholar] [CrossRef] [PubMed]
  264. Wolf, J.; Urits, I.; Orhurhu, V.; Peck, J.; Orhurhu, M.S.; Giacomazzi, S.; Smoots, D.; Piermarini, C.; Manchikanti, L.; Kaye, A.D.; et al. The Role of the Cannabinoid System in Pain Control: Basic and Clinical Implications. Curr. Pain Headache Rep. 2020, 24, 35. [Google Scholar] [CrossRef] [PubMed]
  265. Haller, V.L.; Cichewicz, D.L.; Welch, S.P. Non-cannabinoid CB1, non-cannabinoid CB2 antinociceptive effects of several novel compounds in the PPQ stretch test in mice. Eur. J. Pharmacol. 2006, 546, 60–68. [Google Scholar] [CrossRef] [PubMed]
  266. Agarwal, N.; Pacher, P.; Tegeder, I.; Amaya, F.; Constantin, C.E.; Brenner, G.J.; Rubino, T.; Michalski, C.W.; Marsicano, G.; Monory, K.; et al. Cannabinoids mediate analgesia largely via peripheral type 1 cannabinoid receptors in nociceptors. Nat. Neurosci. 2007, 10, 870–879. [Google Scholar] [CrossRef] [PubMed]
  267. Maione, S.; De Petrocellis, L.; de Novellis, V.; Moriello, A.S.; Petrosino, S.; Palazzo, E.; Rossi, F.S.; Woodward, D.F.; Di Marzo, V. Analgesic actions of N-arachidonoyl-serotonin, a fatty acid amide hydrolase inhibitor with antagonistic activity at vanilloid TRPV1 receptors. Br. J. Pharmacol. 2007, 150, 766–781. [Google Scholar] [CrossRef] [PubMed]
  268. Costa, B.; Trovato, A.E.; Colleoni, M.; Giagnoni, G.; Zarini, E.; Croci, T. Effect of the cannabinoid CB1 receptor antagonist, SR141716, on nociceptive response and nerve demyelination in rodents with chronic constriction injury of the sciatic nerve. Pain 2005, 116, 52–61. [Google Scholar] [CrossRef] [PubMed]
  269. Saez-Cassanelli, J.L.; Fontanella, G.H.; Delgado-Garcia, J.M.; Carrion, A.M. Functional blockage of the cannabinoid receptor type 1 evokes a kappa-opiate-dependent analgesia. J. Neurochem. 2007, 103, 2629–2639. [Google Scholar] [CrossRef] [PubMed]
  270. Lunn, C.A.; Fine, J.S.; Rojas-Triana, A.; Jackson, J.V.; Fan, X.; Kung, T.T.; Gonsiorek, W.; Schwarz, M.A.; Lavey, B.; Kozlowski, J.A.; et al. A novel cannabinoid peripheral cannabinoid receptor-selective inverse agonist blocks leukocyte recruitment in vivo. J. Pharmacol. Exp. Ther. 2006, 316, 780–788. [Google Scholar] [CrossRef] [PubMed]
  271. Croci, T.; Zarini, E. Effect of the cannabinoid CB1 receptor antagonist rimonabant on nociceptive responses and adjuvant-induced arthritis in obese and lean rats. Br. J. Pharmacol. 2007, 150, 559–566. [Google Scholar] [CrossRef] [PubMed]
  272. Oka, S.; Wakui, J.; Ikeda, S.; Yanagimoto, S.; Kishimoto, S.; Gokoh, M.; Nasui, M.; Sugiura, T. Involvement of the cannabinoid CB2 receptor and its endogenous ligand 2-arachidonoylglycerol in oxazolone-induced contact dermatitis in mice. J. Immunol. 2006, 177, 8796–8805. [Google Scholar] [CrossRef] [PubMed]
  273. Azim, S.; Nicholson, J.; Rebecchi, M.J.; Galbavy, W.; Feng, T.; Reinsel, R.; Volkow, N.D.; Benveniste, H.; Kaczocha, M. Endocannabinoids and acute pain after total knee arthroplasty. Pain 2015, 156, 341–347. [Google Scholar] [CrossRef] [PubMed]
  274. Pertwee, R.G. Cannabinoid receptors and pain. Prog. Neurobiol. 2001, 63, 569–611. [Google Scholar] [CrossRef] [PubMed]
  275. Richardson, J.D.; Kilo, S.; Hargreaves, K.M. Cannabinoids reduce hyperalgesia and inflammation via interaction with peripheral CB1 receptors. Pain 1998, 75, 111–119. [Google Scholar] [CrossRef] [PubMed]
  276. Martin, W.J.; Loo, C.M.; Basbaum, A.I. Spinal cannabinoids are anti-allodynic in rats with persistent inflammation. Pain 1999, 82, 199–205. [Google Scholar] [CrossRef] [PubMed]
  277. Rahn, E.J.; Hohmann, A.G. Cannabinoids as pharmacotherapies for neuropathic pain: From the bench to the bedside. Neurotherapeutics 2009, 6, 713–737. [Google Scholar] [CrossRef] [PubMed]
  278. Bloom, A.S.; Dewey, W.L.; Harris, L.S.; Brosius, K.K. 9-nor-9beta-hydroxyhexahydrocannabinol, a cannabinoid with potent antinociceptive activity: Comparisons with morphine. J. Pharmacol. Exp. Ther. 1977, 200, 263–270. [Google Scholar] [PubMed]
  279. Buxbaum, D.M. Analgesic activity of 9 -tetrahydrocannabinol in the rat and mouse. Psychopharmacologia 1972, 25, 275–280. [Google Scholar] [CrossRef] [PubMed]
  280. Jacob, J.J.; Ramabadran, K.; Campos-Medeiros, M. A pharmacological analysis of levonantradol antinociception in mice. J. Clin. Pharmacol. 1981, 21, 327S–333S. [Google Scholar] [CrossRef] [PubMed]
  281. Walker, J.M.; Hohmann, A.G.; Martin, W.J.; Strangman, N.M.; Huang, S.M.; Tsou, K. The neurobiology of cannabinoid analgesia. Life Sci. 1999, 65, 665–673. [Google Scholar] [CrossRef] [PubMed]
  282. Alsalem, M.; Altarifi, A.; Haddad, M.; Aldossary, S.A.; Kalbouneh, H.; Aldaoud, N.; Saleh, T.; El-Salem, K. Antinociceptive and Abuse Potential Effects of Cannabinoid/Opioid Combinations in a Chronic Pain Model in Rats. Brain Sci. 2019, 9, 328. [Google Scholar] [CrossRef] [PubMed]
  283. Maguire, D.R.; France, C.P. 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, 217–224. [Google Scholar] [CrossRef] [PubMed]
  284. Welch, S.P.; Stevens, D.L. Antinociceptive activity of intrathecally administered cannabinoids alone, and in combination with morphine, in mice. J. Pharmacol. Exp. Ther. 1992, 262, 10–18. [Google Scholar] [PubMed]
  285. Bloom, A.S.; Dewey, W.L. A comparison of some pharmacological actions of morphine and delta9-tetrahydrocannabinol in the mouse. Psychopharmacology 1978, 57, 243–248. [Google Scholar] [CrossRef] [PubMed]
  286. Lichtman, A.H.; Smith, F.L.; Martin, B.R. Evidence that the antinociceptive tail-flick response is produced independently from changes in either tail-skin temperature or core temperature. Pain 1993, 55, 283–295. [Google Scholar] [CrossRef] [PubMed]
  287. Smith, P.B.; Martin, B.R. Spinal mechanisms of delta9-tetrahydrocannabinol-induced analgesia. Brain Res. 1992, 578, 8–12. [Google Scholar] [CrossRef] [PubMed]
  288. Thorat, S.N.; Bhargava, H.N. Evidence for a bidirectional cross-tolerance between morphine and delta9-tetrahydrocannabinol in mice. Eur. J. Pharmacol. 1994, 260, 5–13. [Google Scholar] [CrossRef] [PubMed]
  289. Cravatt, B.F.; Lichtman, A.H. The endogenous cannabinoid system and its role in nociceptive behavior. J. Neurobiol. 2004, 61, 149–160. [Google Scholar] [CrossRef] [PubMed]
  290. Goya, P.; Jagerovic, N.; Hernandez-Folgado, L.; Martin, M.I. Cannabinoids and neuropathic pain. Mini Rev. Med. Chem. 2003, 3, 765–772. [Google Scholar] [CrossRef] [PubMed]
  291. Bisogno, T.; Maurelli, S.; Melck, D.; De Petrocellis, L.; Di Marzo, V. Biosynthesis, uptake, and degradation of anandamide and palmitoylethanolamide in leukocytes. J. Biol. Chem. 1997, 272, 3315–3323. [Google Scholar] [CrossRef] [PubMed]
  292. Di Marzo, V.; Blumberg, P.M.; Szallasi, A. Endovanilloid signaling in pain. Curr. Opin. Neurobiol. 2002, 12, 372–379. [Google Scholar] [CrossRef]
  293. Anand, U.; Otto, W.R.; Sanchez-Herrera, D.; Facer, P.; Yiangou, Y.; Korchev, Y.; Birch, R.; Benham, C.; Bountra, C.; Chessell, I.P.; et al. Cannabinoid receptor CB2 localisation and agonist-mediated inhibition of capsaicin responses in human sensory neurons. Pain 2008, 138, 667–680. [Google Scholar] [CrossRef] [PubMed]
  294. 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–E796. [Google Scholar] [CrossRef] [PubMed]
  295. Petzke, F.; Tolle, T.; Fitzcharles, M.A.; Hauser, W. Cannabis-Based Medicines and Medical Cannabis for Chronic Neuropathic Pain. CNS Drugs 2022, 36, 31–44. [Google Scholar] [CrossRef] [PubMed]
  296. Russo, E.B. History of cannabis and its preparations in saga, science, and sobriquet. Chem. Biodivers. 2007, 4, 1614–1648. [Google Scholar] [CrossRef] [PubMed]
  297. Buggy, D.J.; Toogood, L.; Maric, S.; Sharpe, P.; Lambert, D.G.; Rowbotham, D.J. Lack of analgesic efficacy of oral delta-9-tetrahydrocannabinol in postoperative pain. Pain 2003, 106, 169–172. [Google Scholar] [CrossRef] [PubMed]
  298. Kraft, B.; Frickey, N.A.; Kaufmann, R.M.; Reif, M.; Frey, R.; Gustorff, B.; Kress, H.G. Lack of analgesia by oral standardized cannabis extract on acute inflammatory pain and hyperalgesia in volunteers. Anesthesiology 2008, 109, 101–110. [Google Scholar] [CrossRef] [PubMed]
  299. Naef, M.; Curatolo, M.; Petersen-Felix, S.; Arendt-Nielsen, L.; Zbinden, A.; Brenneisen, R. The analgesic effect of oral delta-9-tetrahydrocannabinol (THC), morphine, and a THC-morphine combination in healthy subjects under experimental pain conditions. Pain 2003, 105, 79–88. [Google Scholar] [CrossRef] [PubMed]
  300. Naef, M.; Russmann, S.; Petersen-Felix, S.; Brenneisen, R. Development and pharmacokinetic characterization of pulmonal and intravenous delta-9-tetrahydrocannabinol (THC) in humans. J. Pharm. Sci. 2004, 93, 1176–1184. [Google Scholar] [CrossRef] [PubMed]
  301. Wallace, M.; Schulteis, G.; Atkinson, J.H.; Wolfson, T.; Lazzaretto, D.; Bentley, H.; Gouaux, B.; Abramson, I. Dose-dependent effects of smoked cannabis on capsaicin-induced pain and hyperalgesia in healthy volunteers. Anesthesiology 2007, 107, 785–796. [Google Scholar] [CrossRef] [PubMed]
  302. Campbell, F.A.; Tramer, M.R.; Carroll, D.; Reynolds, D.J.; Moore, R.A.; McQuay, H.J. Are cannabinoids an effective and safe treatment option in the management of pain? A qualitative systematic review. BMJ 2001, 323, 13–16. [Google Scholar] [CrossRef] [PubMed]
  303. Fitzcharles, M.A.; Baerwald, C.; Ablin, J.; Hauser, W. Efficacy, tolerability and safety of cannabinoids in chronic pain associated with rheumatic diseases (fibromyalgia syndrome, back pain, osteoarthritis, rheumatoid arthritis): A systematic review of randomized controlled trials. Schmerz 2016, 30, 47–61. [Google Scholar] [CrossRef] [PubMed]
  304. Mucke, M.; Phillips, T.; Radbruch, L.; Petzke, F.; Hauser, W. Cannabis-based medicines for chronic neuropathic pain in adults. Cochrane Database Syst. Rev. 2018, 3, CD012182. [Google Scholar] [CrossRef] [PubMed]
  305. Phillips, T.J.; Cherry, C.L.; Cox, S.; Marshall, S.J.; Rice, A.S. Pharmacological treatment of painful HIV-associated sensory neuropathy: A systematic review and meta-analysis of randomised controlled trials. PLoS ONE 2010, 5, e14433. [Google Scholar] [CrossRef] [PubMed]
  306. Stockings, E.; Campbell, G.; Hall, W.D.; Nielsen, S.; Zagic, D.; Rahman, R.; Murnion, B.; Farrell, M.; Weier, M.; Degenhardt, L. Cannabis and cannabinoids for the treatment of people with chronic noncancer pain conditions: A systematic review and meta-analysis of controlled and observational studies. Pain 2018, 159, 1932–1954. [Google Scholar] [CrossRef] [PubMed]
  307. Johal, H.; Devji, T.; Chang, Y.; Simone, J.; Vannabouathong, C.; Bhandari, M. Cannabinoids in Chronic Non-Cancer Pain: A Systematic Review and Meta-Analysis. Clin. Med. Insights Arthritis Musculoskelet. Disord. 2020, 13, 1179544120906461. [Google Scholar] [CrossRef] [PubMed]
  308. Fisher, E.; Moore, R.A.; Fogarty, A.E.; Finn, D.P.; Finnerup, N.B.; Gilron, I.; Haroutounian, S.; Krane, E.; Rice, A.S.C.; Rowbotham, M.; et al. Cannabinoids, cannabis, and cannabis-based medicine for pain management: A systematic review of randomised controlled trials. Pain 2021, 162, S45–S66. [Google Scholar] [CrossRef] [PubMed]
  309. Chou, R.; Gordon, D.B.; de Leon-Casasola, O.A.; Rosenberg, J.M.; Bickler, S.; Brennan, T.; Carter, T.; Cassidy, C.L.; Chittenden, E.H.; Degenhardt, E.; et al. Management of Postoperative Pain: A Clinical Practice Guideline From the American Pain Society, the American Society of Regional Anesthesia and Pain Medicine, and the American Society of Anesthesiologists’ Committee on Regional Anesthesia, Executive Committee, and Administrative Council. J. Pain 2016, 17, 131–157. [Google Scholar] [CrossRef] [PubMed]
  310. Inturrisi, C.E. Clinical pharmacology of opioids for pain. Clin. J. Pain 2002, 18, S3–S13. [Google Scholar] [CrossRef] [PubMed]
  311. Oderda, G.M.; Gan, T.J.; Johnson, B.H.; Robinson, S.B. Effect of opioid-related adverse events on outcomes in selected surgical patients. J. Pain Palliat. Care Pharmacother. 2013, 27, 62–70. [Google Scholar] [CrossRef] [PubMed]
  312. Kumar, K.; Kirksey, M.A.; Duong, S.; Wu, C.L. A Review of Opioid-Sparing Modalities in Perioperative Pain Management: Methods to Decrease Opioid Use Postoperatively. Anesth. Analg. 2017, 125, 1749–1760. [Google Scholar] [CrossRef] [PubMed]
  313. Weber, L.; Yeomans, D.C.; Tzabazis, A. Opioid-induced hyperalgesia in clinical anesthesia practice: What has remained from theoretical concepts and experimental studies? Curr. Opin. Anesthesiol. 2017, 30, 458–465. [Google Scholar] [CrossRef] [PubMed]
  314. Edwards, D.A.; Hedrick, T.L.; Jayaram, J.; Argoff, C.; Gulur, P.; Holubar, S.D.; Gan, T.J.; Mythen, M.G.; Miller, T.E.; Shaw, A.D.; et al. American Society for Enhanced Recovery and Perioperative Quality Initiative Joint Consensus Statement on Perioperative Management of Patients on Preoperative Opioid Therapy. Anesth. Analg. 2019, 129, 553–566. [Google Scholar] [CrossRef] [PubMed]
  315. Lee, L.A.; Caplan, R.A.; Stephens, L.S.; Posner, K.L.; Terman, G.W.; Voepel-Lewis, T.; Domino, K.B. Postoperative opioid-induced respiratory depression: A closed claims analysis. Anesthesiology 2015, 122, 659–665. [Google Scholar] [CrossRef] [PubMed]
  316. Nagappa, M.; Weingarten, T.N.; Montandon, G.; Sprung, J.; Chung, F. Opioids, respiratory depression, and sleep-disordered breathing. Best Pract. Res. Clin. Anaesthesiol. 2017, 31, 469–485. [Google Scholar] [CrossRef] [PubMed]
  317. Shaikh, S.I.; Nagarekha, D.; Hegade, G.; Marutheesh, M. Postoperative nausea and vomiting: A simple yet complex problem. Anesth. Essays Res. 2016, 10, 388–396. [Google Scholar] [CrossRef] [PubMed]
  318. de Boer, H.D.; Detriche, O.; Forget, P. Opioid-related side effects: Postoperative ileus, urinary retention, nausea and vomiting, and shivering. A review of the literature. Best Pract. Res. Clin. Anaesthesiol. 2017, 31, 499–504. [Google Scholar] [CrossRef] [PubMed]
  319. Dorn, S.; Lembo, A.; Cremonini, F. Opioid-induced bowel dysfunction: Epidemiology, pathophysiology, diagnosis, and initial therapeutic approach. Am. J. Gastroenterol. Suppl. 2014, 2, 31–37. [Google Scholar] [CrossRef] [PubMed]
  320. Farmer, A.D.; Holt, C.B.; Downes, T.J.; Ruggeri, E.; Del Vecchio, S.; De Giorgio, R. Pathophysiology, diagnosis, and management of opioid-induced constipation. Lancet Gastroenterol. Hepatol. 2018, 3, 203–212. [Google Scholar] [CrossRef] [PubMed]
  321. Kurz, A.; Sessler, D.I. Opioid-induced bowel dysfunction: Pathophysiology and potential new therapies. Drugs 2003, 63, 649–671. [Google Scholar] [CrossRef] [PubMed]
  322. Verhamme, K.M.; Sturkenboom, M.C.; Stricker, B.H.; Bosch, R. Drug-induced urinary retention: Incidence, management and prevention. Drug Saf. 2008, 31, 373–388. [Google Scholar] [CrossRef] [PubMed]
  323. Devlin, J.W.; Roberts, R.J. Pharmacology of commonly used analgesics and sedatives in the ICU: Benzodiazepines, propofol, and opioids. Crit. Care Clin. 2009, 25, 431–449, vii. [Google Scholar] [CrossRef] [PubMed]
  324. Wheeler, M.; Oderda, G.M.; Ashburn, M.A.; Lipman, A.G. Adverse events associated with postoperative opioid analgesia: A systematic review. J. Pain 2002, 3, 159–180. [Google Scholar] [CrossRef] [PubMed]
  325. Athanasos, P.; Smith, C.S.; White, J.M.; Somogyi, A.A.; Bochner, F.; Ling, W. Methadone maintenance patients are cross-tolerant to the antinociceptive effects of very high plasma morphine concentrations. Pain 2006, 120, 267–275. [Google Scholar] [CrossRef] [PubMed]
  326. Chia, Y.Y.; Liu, K.; Wang, J.J.; Kuo, M.C.; Ho, S.T. Intraoperative high dose fentanyl induces postoperative fentanyl tolerance. Can. J. Anesth. 1999, 46, 872–877. [Google Scholar] [CrossRef] [PubMed]
  327. Guignard, B.; Bossard, A.E.; Coste, C.; Sessler, D.I.; Lebrault, C.; Alfonsi, P.; Fletcher, D.; Chauvin, M. Acute opioid tolerance: Intraoperative remifentanil increases postoperative pain and morphine requirement. Anesthesiology 2000, 93, 409–417. [Google Scholar] [CrossRef] [PubMed]
  328. Joseph, E.K.; Reichling, D.B.; Levine, J.D. Shared mechanisms for opioid tolerance and a transition to chronic pain. J. Neurosci. 2010, 30, 4660–4666. [Google Scholar] [CrossRef] [PubMed]
  329. Vinik, H.R.; Kissin, I. Rapid development of tolerance to analgesia during remifentanil infusion in humans. Anesth. Analg. 1998, 86, 1307–1311. [Google Scholar] [CrossRef] [PubMed]
  330. Wright, C.; Bigelow, G.E.; Stitzer, M.L.; Liebson, I.A. Acute physical dependence in humans: Repeated naloxone-precipitated withdrawal after a single dose of methadone. Drug Alcohol Depend. 1991, 27, 139–148. [Google Scholar] [CrossRef] [PubMed]
  331. Angst, M.S.; Clark, J.D. Opioid-induced hyperalgesia: A qualitative systematic review. Anesthesiology 2006, 104, 570–587. [Google Scholar] [CrossRef] [PubMed]
  332. Angst, M.S.; Koppert, W.; Pahl, I.; Clark, D.J.; Schmelz, M. Short-term infusion of the mu-opioid agonist remifentanil in humans causes hyperalgesia during withdrawal. Pain 2003, 106, 49–57. [Google Scholar] [CrossRef] [PubMed]
  333. Chu, L.F.; Clark, D.J.; Angst, M.S. Opioid tolerance and hyperalgesia in chronic pain patients after one month of oral morphine therapy: A preliminary prospective study. J. Pain 2006, 7, 43–48. [Google Scholar] [CrossRef] [PubMed]
  334. Fletcher, D.; Martinez, V. Opioid-induced hyperalgesia in patients after surgery: A systematic review and a meta-analysis. Br. J. Anaesth. 2014, 112, 991–1004. [Google Scholar] [CrossRef] [PubMed]
  335. Alam, A.; Gomes, T.; Zheng, H.; Mamdani, M.M.; Juurlink, D.N.; Bell, C.M. Long-term analgesic use after low-risk surgery: A retrospective cohort study. Arch. Intern. Med. 2012, 172, 425–430. [Google Scholar] [CrossRef] [PubMed]
  336. Bateman, B.T.; Franklin, J.M.; Bykov, K.; Avorn, J.; Shrank, W.H.; Brennan, T.A.; Landon, J.E.; Rathmell, J.P.; Huybrechts, K.F.; Fischer, M.A.; et al. Persistent opioid use following cesarean delivery: Patterns and predictors among opioid-naive women. Am. J. Obstet. Gynecol. 2016, 215, 353.e1–353.e18. [Google Scholar] [CrossRef] [PubMed]
  337. Bates, C.; Laciak, R.; Southwick, A.; Bishoff, J. Overprescription of postoperative narcotics: A look at postoperative pain medication delivery, consumption and disposal in urological practice. J. Urol. 2011, 185, 551–555. [Google Scholar] [CrossRef]
  338. Dowell, D.; Haegerich, T.M.; Chou, R. CDC Guideline for Prescribing Opioids for Chronic Pain—United States, 2016. JAMA 2016, 315, 1624–1645. [Google Scholar] [CrossRef] [PubMed]
  339. Rodgers, J.; Cunningham, K.; Fitzgerald, K.; Finnerty, E. Opioid consumption following outpatient upper extremity surgery. J. Hand Surg. 2012, 37, 645–650. [Google Scholar] [CrossRef] [PubMed]
  340. Curatolo, M.; Sveticic, G. Drug combinations in pain treatment: A review of the published evidence and a method for finding the optimal combination. Best Pract. Res. Clin. Anaesthesiol. 2002, 16, 507–519. [Google Scholar] [CrossRef] [PubMed]
  341. O’Neill, A.; Lirk, P. Multimodal Analgesia. Anesthesiol. Clin. 2022, 40, 455–468. [Google Scholar] [CrossRef] [PubMed]
  342. Alger, B.E. Retrograde signaling in the regulation of synaptic transmission: Focus on endocannabinoids. Prog. Neurobiol. 2002, 68, 247–286. [Google Scholar] [CrossRef] [PubMed]
  343. Kano, M.; Ohno-Shosaku, T.; Hashimotodani, Y.; Uchigashima, M.; Watanabe, M. Endocannabinoid-mediated control of synaptic transmission. Physiol. Rev. 2009, 89, 309–380. [Google Scholar] [CrossRef] [PubMed]
  344. Schlicker, E.; Kathmann, M. Modulation of transmitter release via presynaptic cannabinoid receptors. Trends Pharmacol. Sci. 2001, 22, 565–572. [Google Scholar] [CrossRef] [PubMed]
  345. Mao, J.; Price, D.D.; Lu, J.; Keniston, L.; Mayer, D.J. Two distinctive antinociceptive systems in rats with pathological pain. Neurosci. Lett. 2000, 280, 13–16. [Google Scholar] [CrossRef] [PubMed]
  346. Manzanares, J.; Corchero, J.; Romero, J.; Fernandez-Ruiz, J.J.; Ramos, J.A.; Fuentes, J.A. Pharmacological and biochemical interactions between opioids and cannabinoids. Trends Pharmacol. Sci. 1999, 20, 287–294. [Google Scholar] [CrossRef] [PubMed]
  347. Di Marzo, V.; Fontana, A.; Cadas, H.; Schinelli, S.; Cimino, G.; Schwartz, J.C.; Piomelli, D. Formation and inactivation of endogenous cannabinoid anandamide in central neurons. Nature 1994, 372, 686–691. [Google Scholar] [CrossRef] [PubMed]
  348. Cadas, H.; Gaillet, S.; Beltramo, M.; Venance, L.; Piomelli, D. Biosynthesis of an endogenous cannabinoid precursor in neurons and its control by calcium and cAMP. J. Neurosci. 1996, 16, 3934–3942. [Google Scholar] [CrossRef] [PubMed]
  349. Costa, B.; Siniscalco, D.; Trovato, A.E.; Comelli, F.; Sotgiu, M.L.; Colleoni, M.; Maione, S.; Rossi, F.; Giagnoni, G. AM404, an inhibitor of anandamide uptake, prevents pain behaviour and modulates cytokine and apoptotic pathways in a rat model of neuropathic pain. Br. J. Pharmacol. 2006, 148, 1022–1032. [Google Scholar] [CrossRef]
  350. Hojo, M.; Sudo, Y.; Ando, Y.; Minami, K.; Takada, M.; Matsubara, T.; Kanaide, M.; Taniyama, K.; Sumikawa, K.; Uezono, Y. mu-Opioid receptor forms a functional heterodimer with cannabinoid CB1 receptor: Electrophysiological and FRET assay analysis. J. Pharmacol. Sci. 2008, 108, 308–319. [Google Scholar] [CrossRef] [PubMed]
  351. Babalonis, S.; Walsh, S.L. Therapeutic potential of opioid/cannabinoid combinations in humans: Review of the evidence. Eur. Neuropsychopharmacol. 2020, 36, 206–216. [Google Scholar] [CrossRef] [PubMed]
  352. An, D.; Peigneur, S.; Hendrickx, L.A.; Tytgat, J. Targeting Cannabinoid Receptors: Current Status and Prospects of Natural Products. Int. J. Mol. Sci. 2020, 21, 5064. [Google Scholar] [CrossRef] [PubMed]
  353. Yao, B.B.; Mukherjee, S.; Fan, Y.; Garrison, T.R.; Daza, A.V.; Grayson, G.K.; Hooker, B.A.; Dart, M.J.; Sullivan, J.P.; Meyer, M.D. In vitro pharmacological characterization of AM1241: A protean agonist at the cannabinoid CB2 receptor? Br. J. Pharmacol. 2006, 149, 145–154. [Google Scholar] [CrossRef] [PubMed]
  354. Lu, H.C.; Mackie, K. An Introduction to the Endogenous Cannabinoid System. Biol. Psychiatry 2016, 79, 516–525. [Google Scholar] [CrossRef] [PubMed]
  355. Cichewicz, D.L.; Martin, Z.L.; Smith, F.L.; Welch, S.P. Enhancement mu opioid antinociception by oral delta9-tetrahydrocannabinol: Dose-response analysis and receptor identification. J. Pharmacol. Exp. Ther. 1999, 289, 859–867. [Google Scholar] [PubMed]
  356. Fuentes, J.A.; Ruiz-Gayo, M.; Manzanares, J.; Vela, G.; Reche, I.; Corchero, J. Cannabinoids as potential new analgesics. Life Sci. 1999, 65, 675–685. [Google Scholar] [CrossRef] [PubMed]
  357. Ghosh, P.; Bhattacharya, S.K. Cannabis-induced potentiation of morphine analgesia in rat--role of brain monoamines. Indian J. Med. Res. 1979, 70, 275–280. [Google Scholar] [PubMed]
  358. Reche, I.; Fuentes, J.A.; Ruiz-Gayo, M. Potentiation of delta 9-tetrahydrocannabinol-induced analgesia by morphine in mice: Involvement of mu- and kappa-opioid receptors. Eur. J. Pharmacol. 1996, 318, 11–16. [Google Scholar] [CrossRef] [PubMed]
  359. Smith, F.L.; Cichewicz, D.; Martin, Z.L.; Welch, S.P. The enhancement of morphine antinociception in mice by delta9-tetrahydrocannabinol. Pharmacol. Biochem. Behav. 1998, 60, 559–566. [Google Scholar] [CrossRef] [PubMed]
  360. Cichewicz, D.L.; Haller, V.L.; Welch, S.P. Changes in opioid and cannabinoid receptor protein following short-term combination treatment with delta9-tetrahydrocannabinol and morphine. J. Pharmacol. Exp. Ther. 2001, 297, 121–127. [Google Scholar] [PubMed]
  361. Reche, I.; Ruiz-Gayo, M.; Fuentes, J.A. Inhibition of opioid-degrading enzymes potentiates delta9-tetrahydrocannabinol-induced antinociception in mice. Neuropharmacology 1998, 37, 215–222. [Google Scholar] [CrossRef] [PubMed]
  362. Yesilyurt, O.; Dogrul, A.; Gul, H.; Seyrek, M.; Kusmez, O.; Ozkan, Y.; Yildiz, O. Topical cannabinoid enhances topical morphine antinociception. Pain 2003, 105, 303–308. [Google Scholar] [CrossRef] [PubMed]
  363. Levin, D.N.; Dulberg, Z.; Chan, A.W.; Hare, G.M.; Mazer, C.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, 385–395. [Google Scholar] [CrossRef] [PubMed]
  364. Seeling, W.; Kneer, L.; Buchele, B.; Gschwend, J.E.; Maier, L.; Nett, C.; Simmet, T.; Steffen, P.; Schneider, M.; Rockemann, M. [Delta9-tetrahydrocannabinol and the opioid receptor agonist piritramide do not act synergistically in postoperative pain]. Anaesthesist 2006, 55, 391–400. [Google Scholar] [CrossRef] [PubMed]
  365. Miranda, H.F.; Sierralta, F.; Pinardi, G. Neostigmine interactions with non steroidal anti-inflammatory drugs. Br. J. Pharmacol. 2002, 135, 1591–1597. [Google Scholar] [CrossRef] [PubMed]
  366. Mitchell, J.A.; Warner, T.D. Cyclo-oxygenase-2: Pharmacology, physiology, biochemistry and relevance to NSAID therapy. Br. J. Pharmacol. 1999, 128, 1121–1132. [Google Scholar] [CrossRef] [PubMed]
  367. Smith, W.L.; DeWitt, D.L.; Garavito, R.M. Cyclooxygenases: Structural, cellular, and molecular biology. Annu. Rev. Biochem. 2000, 69, 145–182. [Google Scholar] [CrossRef] [PubMed]
  368. Warner, T.D.; Mitchell, J.A. Cyclooxygenases: New forms, new inhibitors, and lessons from the clinic. FASEB J. 2004, 18, 790–804. [Google Scholar] [CrossRef] [PubMed]
  369. Miranda, H.F.; Puig, M.M.; Prieto, J.C.; Pinardi, G. Synergism between paracetamol and nonsteroidal anti-inflammatory drugs in experimental acute pain. Pain 2006, 121, 22–28. [Google Scholar] [CrossRef] [PubMed]
  370. Fimiani, C.; Liberty, T.; Aquirre, A.J.; Amin, I.; Ali, N.; Stefano, G.B. Opiate, cannabinoid, and eicosanoid signaling converges on common intracellular pathways nitric oxide coupling. Prostaglandins Other Lipid Mediat. 1999, 57, 23–34. [Google Scholar] [CrossRef] [PubMed]
  371. Reichman, M.; Nen, W.; Hokin, L.E. Delta9-tetrahydrocannabinol increases arachidonic acid levels in guinea pig cerebral cortex slices. Mol. Pharmacol. 1988, 34, 823–828. [Google Scholar] [PubMed]
  372. Guindon, J.; Beaulieu, P. Antihyperalgesic effects of local injections of anandamide, ibuprofen, rofecoxib and their combinations in a model of neuropathic pain. Neuropharmacology 2006, 50, 814–823. [Google Scholar] [CrossRef] [PubMed]
  373. Guindon, J.; De Lean, A.; Beaulieu, P. Local interactions between anandamide, an endocannabinoid, and ibuprofen, a nonsteroidal anti-inflammatory drug, in acute and inflammatory pain. Pain 2006, 121, 85–93. [Google Scholar] [CrossRef] [PubMed]
  374. Ulugol, A.; Ozyigit, F.; Yesilyurt, O.; Dogrul, A. The additive antinociceptive interaction between WIN 55,212-2, a cannabinoid agonist, and ketorolac. Anesth. Analg. 2006, 102, 443–447. [Google Scholar] [CrossRef]
  375. Diaz-Reval, M.I.; Cardenas, Y.; Huerta, M.; Trujillo, X.; Sanchez-Pastor, E.A.; Gonzalez-Trujano, M.E.; Virgen-Ortiz, A.; Perez-Hernandez, M.G. Activation of Peripheral Cannabinoid Receptors Synergizes the Effect of Systemic Ibuprofen in a Pain Model in Rat. Pharmaceuticals 2022, 15, 910. [Google Scholar] [CrossRef] [PubMed]
  376. Paunescu, H.; Coman, O.A.; Coman, L.; Ghita, I.; Georgescu, S.R.; Draghia, F.; Fulga, I. Cannabinoid system and cyclooxygenases inhibitors. J. Med. Life 2011, 4, 11–20. [Google Scholar] [PubMed]
  377. Klinger-Gratz, P.P.; Ralvenius, W.T.; Neumann, E.; Kato, A.; Nyilas, R.; Lele, Z.; Katona, I.; Zeilhofer, H.U. Acetaminophen Relieves Inflammatory Pain through CB1 Cannabinoid Receptors in the Rostral Ventromedial Medulla. J. Neurosci. 2018, 38, 322–334. [Google Scholar] [CrossRef] [PubMed]
  378. Mallet, C.; Daulhac, L.; Bonnefont, J.; Ledent, C.; Etienne, M.; Chapuy, E.; Libert, F.; Eschalier, A. Endocannabinoid and serotonergic systems are needed for acetaminophen-induced analgesia. Pain 2008, 139, 190–200. [Google Scholar] [CrossRef] [PubMed]
  379. Hama, A.T.; Sagen, J. Cannabinoid receptor-mediated antinociception with acetaminophen drug combinations in rats with neuropathic spinal cord injury pain. Neuropharmacology 2010, 58, 758–766. [Google Scholar] [CrossRef] [PubMed]
  380. Ottani, A.; Leone, S.; Sandrini, M.; Ferrari, A.; Bertolini, A. The analgesic activity of paracetamol is prevented by the blockade of cannabinoid CB1 receptors. Eur. J. Pharmacol. 2006, 531, 280–281. [Google Scholar] [CrossRef] [PubMed]
  381. Dani, M.; Guindon, J.; Lambert, C.; Beaulieu, P. The local antinociceptive effects of paracetamol in neuropathic pain are mediated by cannabinoid receptors. Eur. J. Pharmacol. 2007, 573, 214–215. [Google Scholar] [CrossRef] [PubMed]
  382. Mallet, C.; Barriere, D.A.; Ermund, A.; Jonsson, B.A.; Eschalier, A.; Zygmunt, P.M.; Hogestatt, E.D. TRPV1 in brain is involved in acetaminophen-induced antinociception. PLoS ONE 2010, 5, e12748. [Google Scholar] [CrossRef]
  383. Hogestatt, E.D.; Jonsson, B.A.; Ermund, A.; Andersson, D.A.; Bjork, H.; Alexander, J.P.; Cravatt, B.F.; Basbaum, A.I.; Zygmunt, P.M. Conversion of acetaminophen to the bioactive N-acylphenolamine AM404 via fatty acid amide hydrolase-dependent arachidonic acid conjugation in the nervous system. J. Biol. Chem. 2005, 280, 31405–31412. [Google Scholar] [CrossRef] [PubMed]
  384. Ahluwalia, J.; Urban, L.; Capogna, M.; Bevan, S.; Nagy, I. Cannabinoid 1 receptors are expressed in nociceptive primary sensory neurons. Neuroscience 2000, 100, 685–688. [Google Scholar] [CrossRef] [PubMed]
  385. Farquhar-Smith, W.P.; Egertova, M.; Bradbury, E.J.; McMahon, S.B.; Rice, A.S.; Elphick, M.R. Cannabinoid CB1 receptor expression in rat spinal cord. Mol. Cell. Neurosci. 2000, 15, 510–521. [Google Scholar] [CrossRef] [PubMed]
  386. Salio, C.; Doly, S.; Fischer, J.; Franzoni, M.F.; Conrath, M. Neuronal and astrocytic localization of the cannabinoid receptor-1 in the dorsal horn of the rat spinal cord. Neurosci. Lett. 2002, 329, 13–16. [Google Scholar] [CrossRef] [PubMed]
  387. Szallasi, A.; Blumberg, P.M. Vanilloid (Capsaicin) receptors and mechanisms. Pharmacol. Rev. 1999, 51, 159–212. [Google Scholar] [PubMed]
  388. Kang, S.; Kim, C.H.; Lee, H.; Kim, D.Y.; Han, J.I.; Chung, R.K.; Lee, G.Y. Antinociceptive synergy between the cannabinoid receptor agonist WIN 55,212-2 and bupivacaine in the rat formalin test. Anesth. Analg. 2007, 104, 719–725. [Google Scholar] [CrossRef] [PubMed]
  389. Tonner, P.H. Additives used to reduce perioperative opioid consumption 1: Alpha2-agonists. Best Pract. Res. Clin. Anaesthesiol. 2017, 31, 505–512. [Google Scholar] [CrossRef] [PubMed]
  390. Boyd, R.E. Alpha2-adrenergic receptor agonists as analgesics. Curr. Top. Med. Chem. 2001, 1, 193–197. [Google Scholar] [CrossRef] [PubMed]
  391. Giovannitti, J.A., Jr.; Thoms, S.M.; Crawford, J.J. Alpha-2 adrenergic receptor agonists: A review of current clinical applications. Anesth. Prog. 2015, 62, 31–39. [Google Scholar] [CrossRef] [PubMed]
  392. Pan, H.L.; Wu, Z.Z.; Zhou, H.Y.; Chen, S.R.; Zhang, H.M.; Li, D.P. Modulation of pain transmission by G-protein-coupled receptors. Pharmacol. Ther. 2008, 117, 141–161. [Google Scholar] [CrossRef] [PubMed]
  393. Manzanares, J.; Julian, M.; Carrascosa, A. Role of the cannabinoid system in pain control and therapeutic implications for the management of acute and chronic pain episodes. Curr. Neuropharmacol. 2006, 4, 239–257. [Google Scholar] [CrossRef] [PubMed]
  394. 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, 1040–1046. [Google Scholar] [CrossRef]
  395. Guillaud, J.; Legagneux, F.; Paulet, C.; Leoni, J.; Lassner, J. Essai du lvonantradol pour l’analgsie postopratoire. Cah. D’anesthsiologie 1983, 31, 243–248. [Google Scholar]
  396. Jain, A.K.; Ryan, J.R.; McMahon, F.G.; Smith, G. Evaluation of intramuscular levonantradol and placebo in acute postoperative pain. J. Clin. Pharmacol. 1981, 21, 320S–326S. [Google Scholar] [CrossRef] [PubMed]
  397. Beaulieu, P. Effects of nabilone, a synthetic cannabinoid, on postoperative pain. Can. J. Anesth. 2006, 53, 769–775. [Google Scholar] [CrossRef] [PubMed]
  398. Hickernell, T.R.; Lakra, A.; Berg, A.; Cooper, H.J.; Geller, J.A.; Shah, R.P. Should Cannabinoids Be Added to Multimodal Pain Regimens After Total Hip and Knee Arthroplasty? J. Arthroplast. 2018, 33, 3637–3641. [Google Scholar] [CrossRef] [PubMed]
  399. Alaia, M.J.; Li, Z.I.; Chalem, I.; Hurley, E.T.; Vasavada, K.; Gonzalez-Lomas, G.; Rokito, A.S.; Jazrawi, L.M.; Kaplan, K. Cannabidiol for Postoperative Pain Control After Arthroscopic Rotator Cuff Repair Demonstrates No Deficits in Patient-Reported Outcomes Versus Placebo: 1-Year Follow-up of a Randomized Controlled Trial. Orthop. J. Sports Med. 2024, 12, 23259671231222265. [Google Scholar] [CrossRef] [PubMed]
  400. Haffar, A.; Khan, I.A.; Abdelaal, M.S.; Banerjee, S.; Sharkey, P.F.; Lonner, J.H. Topical Cannabidiol (CBD) After Total Knee Arthroplasty Does Not Decrease Pain or Opioid Use: A Prospective Randomized Double-Blinded Placebo-Controlled Trial. J. Arthroplast. 2022, 37, 1763–1770. [Google Scholar] [CrossRef] [PubMed]
  401. Narang, G.; Moore, J.; Wymer, K.; Chang, Y.H.; Lim, E.; Adeleye, O.; Humphreys, M.R.; Stern, K.L. Effect of Cannabidiol Oil on Post-ureteroscopy Pain for Urinary Calculi: A Randomized, Double-blind, Placebo-controlled Trial. J. Urol. 2023, 209, 726–733. [Google Scholar] [CrossRef] [PubMed]
  402. Kalliomaki, J.; Segerdahl, M.; Webster, L.; Reimfelt, A.; Huizar, K.; Annas, P.; Karlsten, R.; Quiding, H. 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. [Google Scholar] [CrossRef] [PubMed]
  403. Ostenfeld, T.; Price, J.; Albanese, M.; Bullman, J.; Guillard, F.; Meyer, I.; Leeson, R.; Costantin, C.; Ziviani, L.; Nocini, P.F.; et al. A randomized, controlled study to investigate the analgesic efficacy of single doses of the cannabinoid receptor-2 agonist GW842166, ibuprofen or placebo in patients with acute pain following third molar tooth extraction. Clin. J. Pain 2011, 27, 668–676. [Google Scholar] [CrossRef] [PubMed]
  404. Moore, A.; Straube, S.; Fisher, E.; Eccleston, C. Cannabidiol (CBD) Products for Pain: Ineffective, Expensive, and With Potential Harms. J. Pain 2024, 25, 833–842. [Google Scholar] [CrossRef] [PubMed]
  405. Khelemsky, Y.; Goldberg, A.T.; Hurd, Y.L.; Winkel, G.; Ninh, A.; Qian, L.; Oprescu, A.; Ciccone, J.; Katz, D.J. Perioperative Patient Beliefs Regarding Potential Effectiveness of Marijuana (Cannabinoids) for Treatment of Pain: A Prospective Population Survey. Reg. Anesth. Pain Med. 2017, 42, 652–659. [Google Scholar] [CrossRef] [PubMed]
  406. Garcia-Gutierrez, M.S.; Navarrete, F.; Gasparyan, A.; Austrich-Olivares, A.; Sala, F.; Manzanares, J. Cannabidiol: A Potential New Alternative for the Treatment of Anxiety, Depression, and Psychotic Disorders. Biomolecules 2020, 10, 1575. [Google Scholar] [CrossRef] [PubMed]
  407. Vigil, J.M.; Stith, S.S.; Brockelman, F.; Keeling, K.; Hall, B. Systematic combinations of major cannabinoid and terpene contents in Cannabis flower and patient outcomes: A proof-of-concept assessment of the Vigil Index of Cannabis Chemovars. J. Cannabis Res. 2023, 5, 4. [Google Scholar] [CrossRef] [PubMed]
  408. Gaston, T.E.; Friedman, D. Pharmacology of cannabinoids in the treatment of epilepsy. Epilepsy Behav. 2017, 70, 313–318. [Google Scholar] [CrossRef] [PubMed]
  409. Grotenhermen, F. Pharmacokinetics and pharmacodynamics of cannabinoids. Clin. Pharmacokinet. 2003, 42, 327–360. [Google Scholar] [CrossRef] [PubMed]
  410. Lucas, C.J.; Galettis, P.; Schneider, J. The pharmacokinetics and the pharmacodynamics of cannabinoids. Br. J. Clin. Pharmacol. 2018, 84, 2477–2482. [Google Scholar] [CrossRef] [PubMed]
  411. Huestis, M.A. Human cannabinoid pharmacokinetics. Chem. Biodivers. 2007, 4, 1770–1804. [Google Scholar] [CrossRef] [PubMed]
  412. Liu, C.W.; Bhatia, A.; Buzon-Tan, A.; Walker, S.; Ilangomaran, D.; Kara, J.; Venkatraghavan, L.; Prabhu, A.J. Weeding Out the Problem: The Impact of Preoperative Cannabinoid Use on Pain in the Perioperative Period. Anesth. Analg. 2019, 129, 874–881. [Google Scholar] [CrossRef] [PubMed]
  413. Mlost, J.; Bryk, M.; Starowicz, K. Cannabidiol for Pain Treatment: Focus on Pharmacology and Mechanism of Action. Int. J. Mol. Sci. 2020, 21, 8870. [Google Scholar] [CrossRef] [PubMed]
  414. British Medical Association. Therapeutic Uses of Cannabis; Harwood Academic Publishers: Amsterdam, The Netherlands, 1997; 142p. [Google Scholar]
  415. Zieglgansberger, W.; Brenneisen, R.; Berthele, A.; Wotjak, C.T.; Bandelow, B.; Tolle, T.R.; Lutz, B. Chronic Pain and the Endocannabinoid System: Smart Lipids—A Novel Therapeutic Option? Med. Cannabis Cannabinoids 2022, 5, 61–75. [Google Scholar] [CrossRef] [PubMed]
  416. Pertwee, R.G.; Gibson, T.M.; Stevenson, L.A.; Ross, R.A.; Banner, W.K.; Saha, B.; Razdan, R.K.; Martin, B.R. O-1057, a potent water-soluble cannabinoid receptor agonist with antinociceptive properties. Br. J. Pharmacol. 2000, 129, 1577–1584. [Google Scholar] [CrossRef] [PubMed]
  417. Rajesh, B. Gandhi, J.R.R. Oral cavity as a site for bioadhesive drug delivery. Adv. Drug Deliv. Rev. 1994, 13, 43–74. [Google Scholar] [CrossRef]
  418. Martin, J.H.; Schneider, J.; Lucas, C.J.; Galettis, P. Exogenous Cannabinoid Efficacy: Merely a Pharmacokinetic Interaction? Clin. Pharmacokinet. 2018, 57, 539–545. [Google Scholar] [CrossRef] [PubMed]
  419. Hua, S. Advances in Nanoparticulate Drug Delivery Approaches for Sublingual and Buccal Administration. Front. Pharmacol. 2019, 10, 1328. [Google Scholar] [CrossRef] [PubMed]
  420. Mahmoudinoodezh, H.; Telukutla, S.R.; Bhangu, S.K.; Bachari, A.; Cavalieri, F.; Mantri, N. The Transdermal Delivery of Therapeutic Cannabinoids. Pharmaceutics 2022, 14, 438. [Google Scholar] [CrossRef] [PubMed]
  421. Challapalli, P.V.; Stinchcomb, A.L. In vitro experiment optimization for measuring tetrahydrocannabinol skin permeation. Int. J. Pharm. 2002, 241, 329–339. [Google Scholar] [CrossRef] [PubMed]
  422. Stinchcomb, A.L.; Valiveti, S.; Hammell, D.C.; Ramsey, D.R. Human skin permeation of Delta8-tetrahydrocannabinol, cannabidiol and cannabinol. J. Pharm. Pharmacol. 2004, 56, 291–297. [Google Scholar] [CrossRef] [PubMed]
  423. Fantegrossi, W.E.; Moran, J.H.; Radominska-Pandya, A.; Prather, P.L. Distinct pharmacology and metabolism of K2 synthetic cannabinoids compared to Delta9-THC: Mechanism underlying greater toxicity? Life Sci. 2014, 97, 45–54. [Google Scholar] [CrossRef] [PubMed]
  424. Ng, T.; Gupta, V.; Keshock, M.C. Tetrahydrocannabinol (THC). In StatPearls; StatPearls Publishing LLC: Treasure Island, FL, USA, 2023. [Google Scholar]
  425. Englund, A.; Stone, J.M.; Morrison, P.D. Cannabis in the arm: What can we learn from intravenous cannabinoid studies? Curr. Pharm. Des. 2012, 18, 4906–4914. [Google Scholar] [CrossRef] [PubMed]
  426. Raft, D.; Gregg, J.; Ghia, J.; Harris, L. Effects of intravenous tetrahydrocannabinol on experimental and surgical pain. Psychological correlates of the analgesic response. Clin. Pharmacol. Ther. 1977, 21, 26–33. [Google Scholar] [CrossRef] [PubMed]
  427. Lindgren, J.E.; Ohlsson, A.; Agurell, S.; Hollister, L.; Gillespie, H. Clinical effects and plasma levels of delta9-tetrahydrocannabinol (delta9-THC) in heavy and light users of cannabis. Psychopharmacology 1981, 74, 208–212. [Google Scholar] [CrossRef] [PubMed]
  428. Maykut, M.O. Health consequences of acute and chronic marihuana use. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 1985, 9, 209–238. [Google Scholar] [CrossRef] [PubMed]
  429. Ohlsson, A.; Lindgren, J.E.; Andersson, S.; Agurell, S.; Gillespie, H.; Hollister, L.E. Single-dose kinetics of deuterium-labelled cannabidiol in man after smoking and intravenous administration. Biomed. Environ. Mass Spectrom. 1986, 13, 77–83. [Google Scholar] [CrossRef] [PubMed]
  430. Casati, S.; Angeli, I.; Ravelli, A.; Del Fabbro, M.; Minoli, M.; Orioli, M. 11-OH-THC in hair as marker of active cannabis consumption: Estimating a reliable cut-off by evaluation of 672 THC-positive hair samples. Forensic. Sci. Int. 2019, 304, 109951. [Google Scholar] [CrossRef] [PubMed]
  431. Martin-Santos, R.; Crippa, J.A.; Batalla, A.; Bhattacharyya, S.; Atakan, Z.; Borgwardt, S.; Allen, P.; Seal, M.; Langohr, K.; Farre, M.; et al. Acute effects of a single, oral dose of d9-tetrahydrocannabinol (THC) and cannabidiol (CBD) administration in healthy volunteers. Curr. Pharm. Des. 2012, 18, 4966–4979. [Google Scholar] [CrossRef] [PubMed]
  432. Zendulka, O.; Dovrtelova, G.; Noskova, K.; Turjap, M.; Sulcova, A.; Hanus, L.; Jurica, J. Cannabinoids and Cytochrome P450 Interactions. Curr. Drug Metab. 2016, 17, 206–226. [Google Scholar] [CrossRef] [PubMed]
  433. Ujvary, I.; Hanus, L. Human Metabolites of Cannabidiol: A Review on Their Formation, Biological Activity, and Relevance in Therapy. Cannabis Cannabinoid Res. 2016, 1, 90–101. [Google Scholar] [CrossRef] [PubMed]
  434. Heuberger, J.A.; Guan, Z.; Oyetayo, O.O.; Klumpers, L.; Morrison, P.D.; Beumer, T.L.; van Gerven, J.M.; Cohen, A.F.; Freijer, J. Population pharmacokinetic model of THC integrates oral, intravenous, and pulmonary dosing and characterizes short- and long-term pharmacokinetics. Clin. Pharmacokinet. 2015, 54, 209–219. [Google Scholar] [CrossRef] [PubMed]
  435. Lucas, C.J.; Galettis, P.; Song, S.; Solowij, N.; Reuter, S.E.; Schneider, J.; Martin, J.H. Cannabinoid Disposition After Human Intraperitoneal Use: AnInsight Into Intraperitoneal Pharmacokinetic Properties in Metastatic Cancer. Clin. Ther. 2018, 40, 1442–1447. [Google Scholar] [CrossRef]
  436. Toennes, S.W.; Ramaekers, J.G.; Theunissen, E.L.; Moeller, M.R.; Kauert, G.F. Comparison of cannabinoid pharmacokinetic properties in occasional and heavy users smoking a marijuana or placebo joint. J. Anal. Toxicol. 2008, 32, 470–477. [Google Scholar] [CrossRef] [PubMed]
  437. Skopp, G.; Potsch, L. Cannabinoid concentrations in spot serum samples 24–48 hours after discontinuation of cannabis smoking. J. Anal. Toxicol. 2008, 32, 160–164. [Google Scholar] [CrossRef] [PubMed]
  438. Doohan, P.T.; Oldfield, L.D.; Arnold, J.C.; Anderson, L.L. Cannabinoid Interactions with Cytochrome P450 Drug Metabolism: A Full-Spectrum Characterization. AAPS J. 2021, 23, 91. [Google Scholar] [CrossRef] [PubMed]
  439. Lopera, V.; Rodriguez, A.; Amariles, P. Clinical Relevance of Drug Interactions with Cannabis: A Systematic Review. J. Clin. Med. 2022, 11, 1154. [Google Scholar] [CrossRef] [PubMed]
  440. Maldonado, C.; Peyraube, R.; Fagiolino, P.; Oricchio, F.; Cunetti, L.; Vazquez, M. Human Data on Pharmacokinetic Interactions of Cannabinoids: A Narrative Review. Curr. Pharm. Des. 2024, 30, 241–254. [Google Scholar] [CrossRef] [PubMed]
  441. Kocis, P.T.; Wadrose, S.; Wakefield, R.L.; Ahmed, A.; Calle, R.; Gajjar, R.; Vrana, K.E. CANNabinoid Drug Interaction Review (CANN-DIR). Med. Cannabis Cannabinoids 2023, 6, 1–7. [Google Scholar] [CrossRef]
  442. Russo, E.B. Current Therapeutic Cannabis Controversies and Clinical Trial Design Issues. Front. Pharmacol. 2016, 7, 309. [Google Scholar] [CrossRef] [PubMed]
  443. Vazquez, M.; Guevara, N.; Maldonado, C.; Guido, P.C.; Schaiquevich, P. Potential Pharmacokinetic Drug-Drug Interactions between Cannabinoids and Drugs Used for Chronic Pain. Biomed. Res. Int. 2020, 2020, 3902740. [Google Scholar] [CrossRef] [PubMed]
  444. Arellano, A.L.; Papaseit, E.; Romaguera, A.; Torrens, M.; Farre, M. Neuropsychiatric and General Interactions of Natural and Synthetic Cannabinoids with Drugs of Abuse and Medicines. CNS Neurol. Disord. Drug Targets 2017, 16, 554–566. [Google Scholar] [CrossRef] [PubMed]
  445. Vaughn, S.E.; Strawn, J.R.; Poweleit, E.A.; Sarangdhar, M.; Ramsey, L.B. The Impact of Marijuana on Antidepressant Treatment in Adolescents: Clinical and Pharmacologic Considerations. J. Pers. Med. 2021, 11, 615. [Google Scholar] [CrossRef] [PubMed]
  446. Jiang, R.; Yamaori, S.; Okamoto, Y.; Yamamoto, I.; Watanabe, K. Cannabidiol is a potent inhibitor of the catalytic activity of cytochrome P450 2C19. Drug Metab. Pharmacokinet. 2013, 28, 332–338. [Google Scholar] [CrossRef] [PubMed]
  447. Yamaori, S.; Okamoto, Y.; Yamamoto, I.; Watanabe, K. Cannabidiol, a major phytocannabinoid, as a potent atypical inhibitor for CYP2D6. Drug Metab. Pharmacokinet. 2011, 39, 2049–2056. [Google Scholar] [CrossRef] [PubMed]
  448. Yamaori, S.; Ebisawa, J.; Okushima, Y.; Yamamoto, I.; Watanabe, K. Potent inhibition of human cytochrome P450 3A isoforms by cannabidiol: Role of phenolic hydroxyl groups in the resorcinol moiety. Life Sci. 2011, 88, 730–736. [Google Scholar] [CrossRef]
  449. Balachandran, P.; Elsohly, M.; Hill, K.P. Cannabidiol Interactions with Medications, Illicit Substances, and Alcohol: A Comprehensive Review. J. Gen. Intern. Med. 2021, 36, 2074–2084. [Google Scholar] [CrossRef] [PubMed]
  450. MacCallum, C.A.; Russo, E.B. Practical considerations in medical cannabis administration and dosing. Eur. J. Intern. Med. 2018, 49, 12–19. [Google Scholar] [CrossRef] [PubMed]
  451. Dewey, W.L. Cannabinoid pharmacology. Pharmacol. Rev. 1986, 38, 151–178. [Google Scholar] [CrossRef] [PubMed]
  452. Hampson, R.E.; Deadwyler, S.A. Cannabinoids, hippocampal function and memory. Life Sci. 1999, 65, 715–723. [Google Scholar] [CrossRef] [PubMed]
  453. Heyser, C.J.; Hampson, R.E.; Deadwyler, S.A. Effects of delta-9-tetrahydrocannabinol on delayed match to sample performance in rats: Alterations in short-term memory associated with changes in task specific firing of hippocampal cells. J. Pharmacol. Exp. Ther. 1993, 264, 294–307. [Google Scholar] [PubMed]
  454. Adams, I.B.; Martin, B.R. Cannabis: Pharmacology and toxicology in animals and humans. Addiction 1996, 91, 1585–1614. [Google Scholar] [CrossRef] [PubMed]
  455. Hollister, L.E. Cannabis—1988. Acta Psychiatr. Scand. Suppl. 1988, 345, 108–118. [Google Scholar] [CrossRef] [PubMed]
  456. Hollister, L.E. Health aspects of cannabis. Pharmacol. Rev. 1986, 38, 1–20. [Google Scholar] [CrossRef]
  457. Barnett, G.; Licko, V.; Thompson, T. Behavioral pharmacokinetics of marijuana. Psychopharmacology 1985, 85, 51–56. [Google Scholar] [CrossRef] [PubMed]
  458. Huestis, M.A.; Sampson, A.H.; Holicky, B.J.; Henningfield, J.E.; Cone, E.J. Characterization of the absorption phase of marijuana smoking. Clin. Pharmacol. Ther. 1992, 52, 31–41. [Google Scholar] [CrossRef] [PubMed]
  459. Johns, A. Psychiatric effects of cannabis. Br. J. Psychiatry 2001, 178, 116–122. [Google Scholar] [CrossRef] [PubMed]
  460. Stanley, C.P.; Hind, W.H.; Tufarelli, C.; O’Sullivan, S.E. Cannabidiol causes endothelium-dependent vasorelaxation of human mesenteric arteries via CB1 activation. Cardiovasc. Res. 2015, 107, 568–578. [Google Scholar] [CrossRef] [PubMed]
  461. Stanley, C.P.; Hind, W.H.; Tufarelli, C.; O’Sullivan, S.E. The endocannabinoid anandamide causes endothelium-dependent vasorelaxation in human mesenteric arteries. Pharmacol. Res. 2016, 113, 356–363. [Google Scholar] [CrossRef] [PubMed]
  462. Malinowska, B.; Baranowska-Kuczko, M.; Schlicker, E. Triphasic blood pressure responses to cannabinoids: Do we understand the mechanism? Br. J. Pharmacol. 2012, 165, 2073–2088. [Google Scholar] [CrossRef] [PubMed]
  463. Gorelick, D.A.; Heishman, S.J.; Preston, K.L.; Nelson, R.A.; Moolchan, E.T.; Huestis, M.A. The cannabinoid CB1 receptor antagonist rimonabant attenuates the hypotensive effect of smoked marijuana in male smokers. Am. Heart J. 2006, 151, 754.e751–754.e755. [Google Scholar] [CrossRef] [PubMed]
  464. Huestis, M.A.; Gorelick, D.A.; Heishman, S.J.; Preston, K.L.; Nelson, R.A.; Moolchan, E.T.; Frank, R.A. Blockade of effects of smoked marijuana by the CB1-selective cannabinoid receptor antagonist SR141716. Arch. Gen. Psychiatry 2001, 58, 322–328. [Google Scholar] [CrossRef] [PubMed]
  465. Jones, R.T. Cardiovascular system effects of marijuana. J. Clin. Pharmacol. 2002, 42, 58S–63S. [Google Scholar] [CrossRef] [PubMed]
  466. Tashkin, D.P.; Levisman, J.A.; Abbasi, A.S.; Shapiro, B.J.; Ellis, N.M. Short-term effects of smoked marihuana on left ventricular function in man. Chest 1977, 72, 20–26. [Google Scholar] [CrossRef] [PubMed]
  467. Perez-Reyes, M. he Psychologic and Physiologic Effects of Active Cannabinoids. In Marihuana and Medicine; Humana Press: Totowa, NJ, USA, 1999. [Google Scholar]
  468. Shook, J.E.; Burks, T.F. Psychoactive cannabinoids reduce gastrointestinal propulsion and motility in rodents. J. Pharmacol. Exp. Ther. 1989, 249, 444–449. [Google Scholar] [PubMed]
  469. Hollmann, M.W.; Rathmell, J.P.; Lirk, P. Optimal postoperative pain management: Redefining the role for opioids. Lancet 2019, 393, 1483–1485. [Google Scholar] [CrossRef]
  470. Hall, W.; Solowij, N. Adverse effects of cannabis. Lancet 1998, 352, 1611–1616. [Google Scholar] [CrossRef]
  471. Beaulieu, P.; Boulanger, A.; Desroches, J.; Clark, A.J. Medical cannabis: Considerations for the anesthesiologist and pain physician. Can. J. Anesth. 2016, 63, 608–624. [Google Scholar] [CrossRef] [PubMed]
  472. Laudanski, K.; Wain, J. Considerations for Cannabinoids in Perioperative Care by Anesthesiologists. J. Clin. Med. 2022, 11, 558. [Google Scholar] [CrossRef] [PubMed]
  473. Mallat, A.; Roberson, J.; Brock-Utne, J.G. Preoperative marijuana inhalation--an airway concern. Can. J. Anesth. 1996, 43, 691–693. [Google Scholar] [CrossRef] [PubMed]
  474. Boyce, S.H.; Quigley, M.A. Uvulitis and partial upper airway obstruction following cannabis inhalation. Emerg. Med. 2002, 14, 106–108. [Google Scholar] [CrossRef] [PubMed]
  475. Guarisco, J.L.; Cheney, M.L.; LeJeune, F.E., Jr.; Reed, H.T. Isolated uvulitis secondary to marijuana use. Laryngoscope 1988, 98, 1309–1312. [Google Scholar] [CrossRef] [PubMed]
  476. White, S.M. Cannabis abuse and laryngospasm. Anaesthesia 2002, 57, 622–623. [Google Scholar] [CrossRef] [PubMed]
  477. Ghuran, A.; Nolan, J. Recreational drug misuse: Issues for the cardiologist. Heart 2000, 83, 627–633. [Google Scholar] [CrossRef] [PubMed]
  478. Kuczkowski, K.M. Anesthetic implications of drug abuse in pregnancy. J. Clin. Anesth. 2003, 15, 382–394. [Google Scholar] [CrossRef] [PubMed]
  479. Badowski, S.; Smith, G. Cannabis use during pregnancy and postpartum. Can. Fam. Physician 2020, 66, 98–103. [Google Scholar]
  480. Conner, S.N.; Bedell, V.; Lipsey, K.; Macones, G.A.; Cahill, A.G.; Tuuli, M.G. Maternal Marijuana Use and Adverse Neonatal Outcomes: A Systematic Review and Meta-analysis. Obstet. Gynecol. 2016, 128, 713–723. [Google Scholar] [CrossRef] [PubMed]
  481. Grant, K.S.; Petroff, R.; Isoherranen, N.; Stella, N.; Burbacher, T.M. Cannabis use during pregnancy: Pharmacokinetics and effects on child development. Pharmacol. Ther. 2018, 182, 133–151. [Google Scholar] [CrossRef] [PubMed]
  482. Ortiz-Medina, M.B.; Perea, M.; Torales, J.; Ventriglio, A.; Vitrani, G.; Aguilar, L.; Roncero, C. Cannabis consumption and psychosis or schizophrenia development. Int. J. Soc. Psychiatry 2018, 64, 690–704. [Google Scholar] [CrossRef] [PubMed]
  483. Sellers, E.M.; Schoedel, K.; Bartlett, C.; Romach, M.; Russo, E.B.; Stott, C.G.; Wright, S.; White, L.; Duncombe, P.; Chen, C.F. A Multiple-Dose, Randomized, Double-Blind, Placebo-Controlled, Parallel-Group QT/QTc Study to Evaluate the Electrophysiologic Effects of THC/CBD Spray. Clin. Pharmacol. Drug Dev. 2013, 2, 285–294. [Google Scholar] [CrossRef] [PubMed]
  484. Hurd, Y.L.; Manzoni, O.J.; Pletnikov, M.V.; Lee, F.S.; Bhattacharyya, S.; Melis, M. Cannabis and the Developing Brain: Insights into Its Long-Lasting Effects. J. Neurosci. 2019, 39, 8250–8258. [Google Scholar] [CrossRef] [PubMed]
  485. Whiting, P.F.; Wolff, R.F.; Deshpande, S.; Di Nisio, M.; Duffy, S.; Hernandez, A.V.; Keurentjes, J.C.; Lang, S.; Misso, K.; Ryder, S.; et al. Cannabinoids for Medical Use: A Systematic Review and Meta-analysis. JAMA 2015, 313, 2456–2473. [Google Scholar] [CrossRef] [PubMed]
  486. Abdallah, F.W.; Hussain, N.; Weaver, T.; Brull, R. Analgesic efficacy of cannabinoids for acute pain management after surgery: A systematic review and meta-analysis. Reg. Anesth. Pain Med. 2020, 45, 509–519. [Google Scholar] [CrossRef] [PubMed]
  487. Stevens, A.J.; Higgins, M.D. A systematic review of the analgesic efficacy of cannabinoid medications in the management of acute pain. Acta Anaesthesiol. Scand. 2017, 61, 268–280. [Google Scholar] [CrossRef] [PubMed]
  488. Fisher, B.A.; Ghuran, A.; Vadamalai, V.; Antonios, T.F. Cardiovascular complications induced by cannabis smoking: A case report and review of the literature. J. Emerg. Med. 2005, 22, 679–680. [Google Scholar] [CrossRef] [PubMed]
  489. Kleine-Brueggeney, M.; Greif, R.; Brenneisen, R.; Urwyler, N.; Stueber, F.; Theiler, L.G. Intravenous Delta-9-Tetrahydrocannabinol to Prevent Postoperative Nausea and Vomiting: A Randomized Controlled Trial. Anesth. Analg. 2015, 121, 1157–1164. [Google Scholar] [CrossRef] [PubMed]
  490. Alaia, M.J.; Hurley, E.T.; Vasavada, K.; Markus, D.H.; Britton, B.; Gonzalez-Lomas, G.; Rokito, A.S.; Jazrawi, L.M.; Kaplan, K. Buccally Absorbed Cannabidiol Shows Significantly Superior Pain Control and Improved Satisfaction Immediately After Arthroscopic Rotator Cuff Repair: A Placebo-Controlled, Double-Blinded, Randomized Trial. Am. J. Sports Med. 2022, 50, 3056–3063. [Google Scholar] [CrossRef] [PubMed]
  491. Allegri, M.; Clark, M.R.; De Andres, J.; Jensen, T.S. Acute and chronic pain: Where we are and where we have to go. Minerva Anestesiol. 2012, 78, 222–235. [Google Scholar] [PubMed]
  492. Gupta, A.; Kaur, K.; Sharma, S.; Goyal, S.; Arora, S.; Murthy, R.S. Clinical aspects of acute post-operative pain management & its assessment. J. Adv. Pharm. Technol. Res. 2010, 1, 97–108. [Google Scholar] [PubMed]
  493. Miller, L.K.; Devi, L.A. The highs and lows of cannabinoid receptor expression in disease: Mechanisms and their therapeutic implications. Pharmacol. Rev. 2011, 63, 461–470. [Google Scholar] [CrossRef]
  494. D’Souza, D.C.; Perry, E.; MacDougall, L.; Ammerman, Y.; Cooper, T.; Wu, Y.T.; Braley, G.; Gueorguieva, R.; Krystal, J.H. The psychotomimetic effects of intravenous delta-9-tetrahydrocannabinol in healthy individuals: Implications for psychosis. Neuropsychopharmacology 2004, 29, 1558–1572. [Google Scholar] [CrossRef] [PubMed]
  495. Jennings, J.M.; Angerame, M.R.; Eschen, C.L.; Phocas, A.J.; Dennis, D.A. Cannabis Use Does Not Affect Outcomes After Total Knee Arthroplasty. J. Arthroplast. 2019, 34, 1667–1669. [Google Scholar] [CrossRef] [PubMed]
  496. Mackie, K. Distribution of cannabinoid receptors in the central and peripheral nervous system. In Handbook of Experimental Pharmacology; Springer: Berlin/Heidelberg, Germany, 2005. [Google Scholar] [CrossRef]
  497. Pazos, M.R.; Nunez, E.; Benito, C.; Tolon, R.M.; Romero, J. Functional neuroanatomy of the endocannabinoid system. Pharmacol. Biochem. Behav. 2005, 81, 239–247. [Google Scholar] [CrossRef] [PubMed]
  498. Hu, S.S.; Mackie, K. Distribution of the Endocannabinoid System in the Central Nervous System. In Handbook of Experimental Pharmacology; Springer: Berlin/Heidelberg, Germany, 2015; Volume 231, pp. 59–93. [Google Scholar] [CrossRef]
  499. Tao, R.; Li, C.; Jaffe, A.E.; Shin, J.H.; Deep-Soboslay, A.; Yamin, R.; Weinberger, D.R.; Hyde, T.M.; Kleinman, J.E. Cannabinoid receptor CNR1 expression and DNA methylation in human prefrontal cortex, hippocampus and caudate in brain development and schizophrenia. Transl. Psychiatry 2020, 10, 158. [Google Scholar] [CrossRef] [PubMed]
  500. Kunos, G.; Osei-Hyiaman, D.; Batkai, S.; Sharkey, K.A.; Makriyannis, A. Should peripheral CB1 cannabinoid receptors be selectively targeted for therapeutic gain? Trends Pharmacol. Sci. 2009, 30, 1–7. [Google Scholar] [CrossRef]
  501. Atwood, B.K.; Mackie, K. CB2: A cannabinoid receptor with an identity crisis. Br. J. Pharmacol. 2010, 160, 467–479. [Google Scholar] [CrossRef] [PubMed]
  502. Maccarrone, M.; Bab, I.; Biro, T.; Cabral, G.A.; Dey, S.K.; Di Marzo, V.; Konje, J.C.; Kunos, G.; Mechoulam, R.; Pacher, P.; et al. Endocannabinoid signaling at the periphery: 50 years after THC. Trends Pharmacol. Sci. 2015, 36, 277–296. [Google Scholar] [CrossRef] [PubMed]
  503. Dhopeshwarkar, A.; Mackie, K. CB2 Cannabinoid receptors as a therapeutic target-what does the future hold? Mol. Pharmacol. 2014, 86, 430–437. [Google Scholar] [CrossRef] [PubMed]
  504. Pernia-Andrade, A.J.; Kato, A.; Witschi, R.; Nyilas, R.; Katona, I.; Freund, T.F.; Watanabe, M.; Filitz, J.; Koppert, W.; Schuttler, J.; et al. Spinal endocannabinoids and CB1 receptors mediate C-fiber-induced heterosynaptic pain sensitization. Science 2009, 325, 760–764. [Google Scholar] [CrossRef] [PubMed]
  505. Cooper, S.A.; Desjardins, P.J.; Turk, D.C.; Dworkin, R.H.; Katz, N.P.; Kehlet, H.; Ballantyne, J.C.; Burke, L.B.; Carragee, E.; Cowan, P.; et al. Research design considerations for single-dose analgesic clinical trials in acute pain: IMMPACT recommendations. Pain 2016, 157, 288–301. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Classification of cannabinoid compounds according to their origin. THCV: Δ9-tetrahydrocannabivarin; CBN: cannabinol; THC: Δ9-tetrahydrocannabinol; CBC: cannabichromene; CBG: cannabigerol; CBD: cannabidiol.
Figure 1. Classification of cannabinoid compounds according to their origin. THCV: Δ9-tetrahydrocannabivarin; CBN: cannabinol; THC: Δ9-tetrahydrocannabinol; CBC: cannabichromene; CBG: cannabigerol; CBD: cannabidiol.
Ijms 25 06268 g001
Figure 2. Schematic representation of the connectivity between the nervous, endocrine, and immune systems through the autonomous nervous system and systemic circulation.
Figure 2. Schematic representation of the connectivity between the nervous, endocrine, and immune systems through the autonomous nervous system and systemic circulation.
Ijms 25 06268 g002
Figure 3. Schematic representation of the main endocannabinoid system components, including the expression of CB1r and CB2r in the CNS. One of the significant properties of Go protein-coupled CB1r/CB2r is that they inhibit the release of a series of transmitters (e.g., glutamate) from the presynaptic terminal. Abbreviations: AEA: anandamide; 2-AG: 2-arachidonylglycerol; CB1R/CB2R: cannabinoid receptor type 1/type 2; DAGLα: diacylglycerol lipase alpha; eCB: endocannabinoid; FAAH: fatty acid amide hydrolase; Go and Gq: different sets of G proteins; MAGL: monoacylglycerol lipase; mCB1R: mitochondrial CB1 receptor; NAPE-PLD: N-acyl phosphatidylethanolamine phospholipase D; P2 × 7: P2 × 7 purinergic receptor; TRPV1: transient receptor potential cation channel subfamily V, member 1.
Figure 3. Schematic representation of the main endocannabinoid system components, including the expression of CB1r and CB2r in the CNS. One of the significant properties of Go protein-coupled CB1r/CB2r is that they inhibit the release of a series of transmitters (e.g., glutamate) from the presynaptic terminal. Abbreviations: AEA: anandamide; 2-AG: 2-arachidonylglycerol; CB1R/CB2R: cannabinoid receptor type 1/type 2; DAGLα: diacylglycerol lipase alpha; eCB: endocannabinoid; FAAH: fatty acid amide hydrolase; Go and Gq: different sets of G proteins; MAGL: monoacylglycerol lipase; mCB1R: mitochondrial CB1 receptor; NAPE-PLD: N-acyl phosphatidylethanolamine phospholipase D; P2 × 7: P2 × 7 purinergic receptor; TRPV1: transient receptor potential cation channel subfamily V, member 1.
Ijms 25 06268 g003
Figure 4. Diagram illustrating the critical signaling pathways triggered by the activation of cannabinoid receptors. CB1r activation triggers Gi/o coupling, leading to inhibition of adenylyl cyclase, modulation of membrane ion channels, and activation of MAPK/ERK and transcription factor signaling pathways. This ultimately results in membrane hyperpolarization. Activation of the CB2 receptor leads to Gi/o coupling, inhibiting adenylyl cyclase and activating MAPK/ERK signaling and transcription factors. Activation of the GPR119 receptor leads to Gs coupling, stimulating adenylyl cyclase, increasing cAMP levels, and activating PKA. Activation of the GPR55 receptor leads to G13 coupling, activating the phospholipase C and RhoA pathway, resulting in increased intracellular calcium levels, activation of MAPK/ERK signaling, and translocation of transcription factors. Activation of the TRPV1 receptor causes a non-selective influx of cations and membrane depolarization. Finally, CB1 and CB2 receptor activation leads to KROX-24 activation through the MAPK/ERK signaling cascade. KROX-24 is a regulatory nuclear transcription factor closely associated with critical biological functions such as stabilizing long-lasting, long-term potentiation, cell differentiation, survival, or death signaling in neuronal cells or regulation of specific neurotransmitters and receptors.
Figure 4. Diagram illustrating the critical signaling pathways triggered by the activation of cannabinoid receptors. CB1r activation triggers Gi/o coupling, leading to inhibition of adenylyl cyclase, modulation of membrane ion channels, and activation of MAPK/ERK and transcription factor signaling pathways. This ultimately results in membrane hyperpolarization. Activation of the CB2 receptor leads to Gi/o coupling, inhibiting adenylyl cyclase and activating MAPK/ERK signaling and transcription factors. Activation of the GPR119 receptor leads to Gs coupling, stimulating adenylyl cyclase, increasing cAMP levels, and activating PKA. Activation of the GPR55 receptor leads to G13 coupling, activating the phospholipase C and RhoA pathway, resulting in increased intracellular calcium levels, activation of MAPK/ERK signaling, and translocation of transcription factors. Activation of the TRPV1 receptor causes a non-selective influx of cations and membrane depolarization. Finally, CB1 and CB2 receptor activation leads to KROX-24 activation through the MAPK/ERK signaling cascade. KROX-24 is a regulatory nuclear transcription factor closely associated with critical biological functions such as stabilizing long-lasting, long-term potentiation, cell differentiation, survival, or death signaling in neuronal cells or regulation of specific neurotransmitters and receptors.
Ijms 25 06268 g004
Table 1. Characteristics, clinical effects, and advantages and disadvantages of different routes of administration of cannabinoids.
Table 1. Characteristics, clinical effects, and advantages and disadvantages of different routes of administration of cannabinoids.
Pulmonary RouteOralOther Routes
SmokingVaporizationOral TransmucosalTopical
Characteristics
  • It is the most common route of administration but not recommended (joints, bongs, pipes, etc.).
  • Combustion at 600–900 °C produces toxic byproducts: tar, PAHs (polycyclic aromatic hydrocarbons), carbon monoxide (CO), and ammonia (NH3).
  • Chronic use is associated with respiratory symptoms (bronchitis, cough, phlegm) but not with lung cancer or COPD (if cannabis only).
  • Patients may mix with tobacco, increasing respiratory/cancer risk.
  • 30–50% of cannabis is lost in ‘side-stream’ smoke.
  • Heat cannabis at 160–230 °C reduces CO, but PAHs are not eliminated.
  • Vaporization produces significantly fewer harmful byproducts than smoking.
  • Reduced pulmonary symptoms were reported compared to smoking.
  • Oils, capsules and other oral routes are becoming increasingly popular due to the convenience and accuracy of dosing.
  • Edibles (brownies/cookies) can be more difficult to dose.
  • Juicing and cannabis teas do not allow for adequate decarboxylation of the raw plant.
  • Tinctures and lozenges with limited research.
  • Intermediate onset
Nabiximols oral spray is currently the only cannabis-based prescription that delivers a standardized dosage of CBD/THC in a 1:1 ratio with extensive research.Topical is ideal for localized symptoms (dermatological conditions, arthritis), with limited research evidence.
Clinical effectsOnset (min)5–10 min60–180 min15–45 minVariable
Duration (h)2–4 h6–8 h6–8 hVariable
AdvantagesRapid action benefits acute or episodic symptoms (nausea/pain).Less odor, convenient and discreet, prolonged effect. Advantage for chronic disease/symptoms.Pharmaceutical form (nabiximols) available, with documented efficacy and safety.Less systemic effect, suitable for localized symptoms.
DisadvantagesDexterity is required, vaporizers may be expensive, and not all are portable.Titration challenges due to delayed onset.Expensive, spotty availability.Only local effects.
Table 2. Adverse events mainly associated with THC content in cannabis-based medicines.
Table 2. Adverse events mainly associated with THC content in cannabis-based medicines.
Side EffectMost CommonCommonRare
Drowsiness/fatigue×
Dizziness×
Cough, phlegm, bronchitis (smoking only)×
Anxiety×
Nausea×
Cognitive effects×
Euphoria ×
Blurred vision ×
Headache ×
Orthostatic hypotension ×
Toxic psychosis/paranoia ×
Depression ×
Ataxia/dyscoordination ×
Tachycardia (after titration) ×
Cannabis hyperemesis ×
Diarrhea ×
Table 3. Summary of clinical trials evaluating cannabinoids in postoperative pain. AZD1940, peripherally restricted CB1r/CB12r agonist; Cannador, mixture of cannabinoid plant extracts containing predominantly THC and CBD (ratio 1:0.3–0.5); CBD, cannabidiol; GW842166, selective CB2r agonist; I.M.IMtramuscular; P.O.POr os; PCA, patient-controlled analgesia; RCT: randomized controlled trial; THC, tetrahydrocannabinol.
Table 3. Summary of clinical trials evaluating cannabinoids in postoperative pain. AZD1940, peripherally restricted CB1r/CB12r agonist; Cannador, mixture of cannabinoid plant extracts containing predominantly THC and CBD (ratio 1:0.3–0.5); CBD, cannabidiol; GW842166, selective CB2r agonist; I.M.IMtramuscular; P.O.POr os; PCA, patient-controlled analgesia; RCT: randomized controlled trial; THC, tetrahydrocannabinol.
Type and Design of the StudySubjectsSurgical ProcedurePrimary OutcomePostoperative AnalgesiaCannabinoid InterventionMain ResultsReference
RCT (double blind, placebo-controlled, crossover design)56
patients
Acute Fracture or traumaN/SN/SLevonantradol I.M. Preoperative regimen. Single injection: 1, 1.5, 2, 2.5, and 3 mg.Pain relief with the four doses; analgesia persisted for more than 6 h with the 2.5 and 3 mg doses. Jain et al., 1981 [349]
Placebo-controlled, single-dose100
patients
Renal surgery with lumbar incisionN/SNoramidopyrine (metamizol), Camylofine (anti-cholinergic drug)Levonantradol I.M. Postoperative regimen. Single injection: 1 and 2 mg.No significant difference compared with placebo.Guillaud et al., 1983 [348]
RCT (double blind, placebo-controlled, single dose, parallel)40
patients
Elective abdominal hysterectomyPain scoresMorphine PCATHC P.O. Postoperative regimen. Single dose: 5 mg.No significant difference compared with placebo.
Increased awareness of surroundings is more frequently reported with THC
Buggy
et al., 2003 [259]
RCT (double-blind, placebo-controlled)41
patients
Orthopedic, gynecology, urology, and
plastic or general surgery
Opioid
consumption
at 24 h
Morphine PCANabilone P.O. Preoperative and postoperative regimen. 1 and 2 mg.No significant difference compared with placebo.Beaulieu et al., 2006 [350]
RCT (double-blind, placebo-controlled)100
patients
Radical retropubic prostatectomyOpioid
consumption
at 48 h
Piritramide PCADronabinol P.O. Preoperative (evening before operation) and postoperative until the morning of the 2nd postoperative day) regimen. 8 doses of 5 mg.No significant difference between dronabinol and placebo groups in the self-administration of post-operative piritramide.Seeling et al., 2006 [322]
Dose escalating study65
patients
Various major
Surgeries (included orthopedic, gynecologic, urology, plastics, and general)
N/SMorphine PCACannador P.O. Postoperative regimen. Single dose: 5, 10, 15 and 24 mg.Significant dose-related improvements in rescue analgesia requirements in the 10 and 15 mg groups.
Study ended because of a serious vasovagal adverse event in a patient receiving 15 mg.
Holdcroft
et al., 2006 [347]
CT (double-blind, placebo-controlled)112
patients
Third molar tooth extractionPain scores up to 10 h postsurgery500 mg acetaminophen,
15 mg codeine
phosphate
GW842166 P.O. Preoperative regimen. Single dose: 100 and 800 mg.In comparison to ibuprofen, single doses of GW842166 (100 and 800 mg) failed to demonstrate clinically meaningful analgesia in the setting of acute dental pain.Ostenfeld
et al., 2011 [353]
RCT (double-blind, placebo-controlled)150
patients
Removal of impacted lower third molar toothArea under
the curve pain scores
1000 mg acetaminophenAZD1940 P.O. Preoperative regimen. Single dose: 800 µg.No significant differences compared with placebo.Kalliomaki
et al., 2013 [352]
RCT (double-blind, placebo-controlled)99 patientsArthroscopic rotator cuff repairPain scores
Patient satisfaction with pain control
Opioid consumption at days 1, 2, 7, and 14
5 mg oxycodone
325 mg acetaminophen
CBD P.O. Postoperative regimen. Repeated dose (three times a day, 14 days): 25 and 50 mgDays 1 and 2:
Lower pain score in the CBD group.
Higher patient satisfaction with pain control.
No statistical difference between groups in opioid consumption
Days 7 and 14:
No significant differences compared with placebo.
Alaia et al., 2022 [422]
RCT (double-blind, placebo-controlled)80 patientsTotal knee arthroplastyPain and sleep scores
Cumulative postoperative opioid use
1000 mg acetaminophen
300 mg gabapentin
15 mg meloxicam
5 mg oxycodone
CBD topical. Postoperative regimen. Repeated dose (three times a day, 14 days): 120 mg/ounceNo significant differences compared with placebo.Haffar et al., 2022 [421]
RCT (double-blind, placebo-controlled)94 patientsUreteroscopy with stent placement for urinary stone diseasePain scores
Postoperative opioid use
5 mg oxycodone CBD P.O. Postoperative regimen. Repeated dose (3 days): 20 mgNo significant differences compared with placebo.Narang et al., 2023 [424]
RCT (double-blind, placebo-controlled)83 patients (follow-up)Arthroscopic rotator cuff repairPain scores
Patient satisfaction with pain control
Opioid consumption at 7 and 14 days
5 mg oxycodone
325 mg acetaminophen
CBD P.O. Postoperative regimen. Repeated dose (three times a day, 14 days): 25 and 50 mgNo significant differences compared with placebo.Alaia et al., 2024 [423]
Table 4. Factors related to the lack of evidence regarding analgesia for postsurgical pain.
Table 4. Factors related to the lack of evidence regarding analgesia for postsurgical pain.
Related to the symptomsPostoperative pain is usually localized. In this context, the activation of the endocannabinoid system is minor compared to that in chronic pain, and cannabinoids are mainly associated with the relief of neuropathic pain.
Pain assessment is highly subjective, and the quantification and comparison between study groups are generally inconclusive.
Related to the productOn the one hand, the composition of phytocannabinoids is very heterogeneous. On the other hand, studies of its components separately do not provide the same results as when the whole plant is analyzed.
Human clinical trials on postoperative pain used almost exclusively THC or an analog.
Few studies are testing a mixture of THC/CBD or CBD without THC in humans for the treatment of postoperative pain.
Lack of knowledge about cannabinoid interaction (synergic, antagonistic, entourage effect).
Related to the administration routeIn the systemic routes, the clinical doses seem limited by the appearance of side effects, primarily psychotropic.
There is no record of perimedullary administration at the clinical level of cannabinoid compounds, with robust and proven analgesic preclinical evidence for this route of administration.
Related to the studiesShort-term duration (mostly single-dose administration and a short follow-up period).
There is considerable variation in the doses and therapeutic guidelines employed.
There are no 100% effective standard treatments against which to compare the effect of cannabinoids.
A small number of patients in multiple operative settings.
No homogenous groups (healthy patients, patients with diseases of different etiologies).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Carrascosa, A.J.; Navarrete, F.; Saldaña, R.; García-Gutiérrez, M.S.; Montalbán, B.; Navarro, D.; Gómez-Guijarro, F.M.; Gasparyan, A.; Murcia-Sánchez, E.; Torregrosa, A.B.; et al. Cannabinoid Analgesia in Postoperative Pain Management: From Molecular Mechanisms to Clinical Reality. Int. J. Mol. Sci. 2024, 25, 6268. https://doi.org/10.3390/ijms25116268

AMA Style

Carrascosa AJ, Navarrete F, Saldaña R, García-Gutiérrez MS, Montalbán B, Navarro D, Gómez-Guijarro FM, Gasparyan A, Murcia-Sánchez E, Torregrosa AB, et al. Cannabinoid Analgesia in Postoperative Pain Management: From Molecular Mechanisms to Clinical Reality. International Journal of Molecular Sciences. 2024; 25(11):6268. https://doi.org/10.3390/ijms25116268

Chicago/Turabian Style

Carrascosa, Antonio J., Francisco Navarrete, Raquel Saldaña, María S. García-Gutiérrez, Belinda Montalbán, Daniela Navarro, Fernando M. Gómez-Guijarro, Ani Gasparyan, Elena Murcia-Sánchez, Abraham B. Torregrosa, and et al. 2024. "Cannabinoid Analgesia in Postoperative Pain Management: From Molecular Mechanisms to Clinical Reality" International Journal of Molecular Sciences 25, no. 11: 6268. https://doi.org/10.3390/ijms25116268

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop