Cannabidiol for Pain Treatment: Focus on Pharmacology and Mechanism of Action
Abstract
:1. Introduction
2. Mechanism of Action
2.1. GPCRs
2.2. Ionotropic Receptors
2.3. Transporters
2.4. Enzymes
2.5. Nuclear Factors
2.6. Summary of Physiologically Relevant Pharmacological Targets
3. The Analgesic Potential of CBD
3.1. Animal Models of Neuropathic Pain
3.2. Inflammatory Pain
3.3. Arthritis-Related Pain
3.4. Other Pain Models
3.5. Clinical Studies
4. Summary
Supplementary Materials
Author Contributions
Funding
Conflicts of Interest
Abbreviations
Δ9-THC | Δ9tetrahydrocannabinol |
Δ9-THC-BDS | Δ9-tetrahydrocannabinol botanical drug substance |
σ1R | Sigma-1 receptor |
2-AG | 2-arachidonoylglycerol |
5HT1a | Serotonin 1A receptor |
AANAT | Aralkylamine N-acetyltransferase |
ABCC1 | Multidrug resistance-associated protein 1 |
ABCG2 | ATP-binding cassette super-family G member 2 |
ACAT | Acyl-CoA cholesterin acyltransferase |
AEA | Anandamide |
CAMP | Cyclic adenosine monophosphate |
CB | Cannabinoid receptor |
CBD | Cannabidiol |
CBDA | Cannabidiolic acid |
CBD–BDS | Cannabidiol-botanical drug substance |
CBC | Cannabichromene |
CBN | Cannabinol |
CCI | Chronic constriction injury |
CFA | Complete Freund’s adjuvant |
CHO | Chinese hamster ovary cells |
CYPs | Cytochrome P450 superfamily |
DOR | δ-opioid receptor |
EAE | Experimental autoimmune encephalitis |
ENT | Equilibrative nucleoside transporter |
EROD | Ethoxyresorufin-O-deethylase |
FAAH | Fatty acid amide hydrolase |
GCSF | Granulocyte colony stimulating factor |
GlyRs | Glycine receptors |
GPCR | G-protein coupled receptor |
GPR | Orphan G-protein coupled receptor |
GTP | Guanosine 5′-O-[gamma-thio]triphosphate |
HEK293 | Human embryonic kidney 293 cells |
HPLC | High-performance liquid chromatography |
HTRF | Homogeneous Time Resolved Fluorescence |
i.a. | Intra-articular |
IDO | Indoleamine-pyrrole 2,3-dioxygenase |
IFN-γ | Interferon- γ |
IFN-1β | Interferon-1β |
IL | Interleukin |
i.p. | Intra-peritoneal |
LOXs | Lipooxygenases |
LPS | Lipopolysaccharide |
MIA | Sodium monoiodoacetate |
MOR | μ-opioid receptor |
NAM | Negative allosteric modulator |
NBD-stearate | 12-N-methyl-(7-nitrobenz-2-oxa-1,3-diazo)aminostearic acid |
NMDA | N-methyl-d-aspartate receptor |
NSAIDs | Nonsteroidal anti-inflammatory drugs |
OA | Osteoarthritis |
PAC | Paclitaxel |
PAG | Periaqueductal grey |
PAM | Positive allosteric modulators |
PBMC | Peripheral blood mononuclear cell |
PLA2 | Phospholipase A2 |
PPRγ | Peroxisome proliferator-activated receptor gamma |
TENS | Transcutaneous electrical nerve stimulation |
TNFα | Tumor necrosis factor α |
TRP | Transient receptor potential channel |
VDAC1 | Voltage-dependent anion channel 1 |
References
- Russo, E.B. History of Cannabis and Its Preparations in Saga, Science, and Sobriquet. Chem. Biodivers. 2007, 4, 1614–1648. [Google Scholar] [CrossRef]
- Ladha, K.S.; Ajrawat, P.; Yang, Y.; Clarke, H. Understanding the Medical Chemistry of the Cannabis Plant is Critical to Guiding Real World Clinical Evidence. Molecules 2020, 25, 4042. [Google Scholar] [CrossRef] [PubMed]
- Toczek, M.; Malinowska, B. Enhanced endocannabinoid tone as a potential target of pharmacotherapy. Life Sci. 2018, 204, 20–45. [Google Scholar] [CrossRef] [PubMed]
- Adams, R.; Hunt, M. Structure of Cannabidiol, a Product Isolated from the Marihuana Extract of Minnesota Wild Hemp. I. J. Am. Chem. Soc. 1940, 62, 196–200. [Google Scholar] [CrossRef]
- Mechoulam, R.; Shani, A.; Edery, H.; Grunfeld, Y. Chemical basis of hashish activity. Science 1970, 169, 611–612. [Google Scholar] [CrossRef]
- Crippa, J.A.; Guimarães, F.S.; Campos, A.C.; Zuardi, A.W. Translational investigation of the therapeutic potential of cannabidiol (CBD): Toward a new age. Front. Immunol. 2018, 9. [Google Scholar] [CrossRef] [Green Version]
- Iffland, K.; Grotenhermen, F. An Update on Safety and Side Effects of Cannabidiol: A Review of Clinical Data and Relevant Animal Studies. Cannabis Cannabinoid Res. 2017, 2, 139–154. [Google Scholar] [CrossRef] [Green Version]
- Thapa, D.; Cairns, E.A.; Szczesniak, A.M.; Toguri, J.T.; Caldwell, M.D.; Kelly, M.E.M. The Cannabinoids Δ8THC, CBD, and HU-308 Act via Distinct Receptors to Reduce Corneal Pain and Inflammation. Cannabis Cannabinoid Res. 2018, 3, 11–20. [Google Scholar] [CrossRef] [Green Version]
- Hammell, D.C.; Zhang, L.P.; Ma, F.; Abshire, S.M.; McIlwrath, S.L.; Stinchcomb, A.L.; Westlund, K.N. Transdermal cannabidiol reduces inflammation and pain-related behaviours in a rat model of arthritis. Eur. J. Pain 2016, 20, 936–948. [Google Scholar] [CrossRef]
- Kozela, E.; Lev, N.; Kaushansky, N.; Eilam, R.; Rimmerman, N.; Levy, R.; Ben-Nun, A.; Juknat, A.; Vogel, Z. Cannabidiol inhibits pathogenic T cells, decreases spinal microglial activation and ameliorates multiple sclerosis-like disease in C57BL/6 mice. Br. J. Pharmacol. 2011, 163, 1507–1519. [Google Scholar] [CrossRef] [Green Version]
- Pan, H.; Mukhopadhyay, P.; Rajesh, M.; Patel, V.; Mukhopadhyay, B.; Gao, B.; Haskó, G.; Pacher, P. Cannabidiol attenuates cisplatin-Lnduced nephrotoxicity by decreasing oxidative/nitrosative stress, inflammation, and cell death. J. Pharmacol. Exp. Ther. 2009, 328, 708–714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Costa, B.; Trovato, A.E.; Comelli, F.; Giagnoni, G.; Colleoni, M. The non-psychoactive cannabis constituent cannabidiol is an orally effective therapeutic agent in rat chronic inflammatory and neuropathic pain. Eur. J. Pharmacol. 2007, 556, 75–83. [Google Scholar] [CrossRef] [PubMed]
- Thomas, B.F.; Gilliam, A.F.; Burch, D.F.; Roche, M.J.; Seltzman, H.H. Comparative receptor binding analyses of cannabinoid agonists and antagonists. J. Pharmacol. Exp. Ther. 1998, 285, 285–292. [Google Scholar] [PubMed]
- Tham, M.; Yilmaz, O.; Alaverdashvili, M.; Kelly, M.E.M.; Denovan-Wright, E.M.; Laprairie, R.B. Allosteric and orthosteric pharmacology of cannabidiol and cannabidiol-dimethylheptyl at the type 1 and type 2 cannabinoid receptors. Br. J. Pharmacol. 2019, 176, 1455–1469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Showalter, V.M.; Compton, D.R.; Martin, B.R.; Abood, M.E. Evaluation of binding in a transfected cell line expressing a peripheral cannabinoid receptor (CB2): Identification of cannabinoid receptor subtype selective ligands. J. Pharmacol. Exp. Ther. 1996, 278, 989–999. [Google Scholar]
- Thomas, A.; Ross, R.A.; Saha, B.; Mahadevan, A.; Razdan, R.K.; Pertwee, R.G. 6″-Azidohex-2″-yne-cannabidiol: A potential neutral, competitive cannabinoid CB1 receptor antagonist. Eur. J. Pharmacol. 2004, 487, 213–221. [Google Scholar] [CrossRef]
- Thomas, A.; Baillie, G.L.; Phillips, A.M.; Razdan, R.K.; Ross, R.A.; Pertwee, R.G. Cannabidiol displays unexpectedly high potency as an antagonist of CB 1 and CB 2 receptor agonists in vitro. Br. J. Pharmacol. 2007, 150, 613–623. [Google Scholar] [CrossRef] [Green Version]
- Martínez-Pinilla, E.; Varani, K.; Reyes-Resina, I.; Angelats, E.; Vincenzi, F.; Ferreiro-Vera, C.; Oyarzabal, J.; Canela, E.I.; Lanciego, J.L.; Nadal, X.; et al. Binding and signaling studies disclose a potential allosteric site for cannabidiol in cannabinoid CB2 receptors. Front. Pharmacol. 2017, 8, 744. [Google Scholar] [CrossRef]
- Laprairie, R.B.; Bagher, A.M.; Kelly, M.E.M.; Denovan-Wright, E.M. Biased type 1 cannabinoid receptor signaling influences neuronal viability in a cell culture model of huntington diseases. Mol. Pharmacol. 2016, 89, 364–375. [Google Scholar] [CrossRef] [Green Version]
- Navarro, G.; Reyes-Resina, I.; Rivas-Santisteban, R.; Sánchez de Medina, V.; Morales, P.; Casano, S.; Ferreiro-Vera, C.; Lillo, A.; Aguinaga, D.; Jagerovic, N.; et al. Cannabidiol skews biased agonism at cannabinoid CB1 and CB2 receptors with smaller effect in CB1-CB2 heteroreceptor complexes. Biochem. Pharmacol. 2018, 157, 148–158. [Google Scholar] [CrossRef]
- Russo, E.B.; Burnett, A.; Hall, B.; Parker, K.K. Agonistic properties of cannabidiol at 5-HT1a receptors. Neurochem. Res. 2005, 30, 1037–1043. [Google Scholar] [CrossRef] [PubMed]
- Rock, E.M.; Bolognini, D.; Limebeer, C.L.; Cascio, M.G.; Anavi-Goffer, S.; Fletcher, P.J.; Mechoulam, R.; Pertwee, R.G.; Parker, L.A. Cannabidiol, a nonpsychotropic component of cannabis, attenuates vomiting and nausea-like behaviour via indirect agonism of 5-HT 1A somatodendritic autoreceptors in the dorsal raphe nucleus. Br. J. Pharmacol. 2012, 165, 2620–2634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ryberg, E.; Larsson, N.; Sjögren, 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]
- 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] [Green Version]
- Henstidge, C.M.; Balenga, N.A.B.; Ford, L.A.; Ross, R.A.; Waldhoer, M.; Irving, A.J. The GPR55 ligand L-α-lysophosphatidylinositol promotes RhoA-dependent Ca 2+ signaling and NFAT activation. FASEB J. 2009, 23, 183–193. [Google Scholar] [CrossRef] [PubMed]
- Kapur, A.; Zhao, P.; Sharir, H.; Bai, Y.; Caron, M.G.; Barak, L.S.; Abood, M.E. Atypical responsiveness of the orphan receptor GPR55 to cannabinoid ligands. J. Biol. Chem. 2009, 284, 29817–29827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Laun, A.S.; Song, Z.H. GPR3 and GPR6, novel molecular targets for cannabidiol. Biochem. Biophys. Res. Commun. 2017, 490, 17–21. [Google Scholar] [CrossRef]
- Brown, K.J.; Laun, A.S.; Song, Z.H. Cannabidiol, a novel inverse agonist for GPR12. Biochem. Biophys. Res. Commun. 2017, 493, 451–454. [Google Scholar] [CrossRef]
- Laun, A.S.; Shrader, S.H.; Brown, K.J.; Song, Z.H. GPR3, GPR6, and GPR12 as novel molecular targets: Their biological functions and interaction with cannabidiol. Acta Pharmacol. Sin. 2019, 40, 300–308. [Google Scholar] [CrossRef] [Green Version]
- Bian, Y.M.; He, X.B.; Jing, Y.K.; Wang, L.R.; Wang, J.M.; Xie, X.Q. Computational systems pharmacology analysis of cannabidiol: A combination of chemogenomics-knowledgebase network analysis and integrated in silico modeling and simulation. Acta Pharmacol. Sin. 2019, 40, 374–386. [Google Scholar] [CrossRef]
- De Petrocellis, L.; Ligresti, A.; Moriello, A.S.; Allarà, M.; Bisogno, T.; Petrosino, S.; Stott, C.G.; Di Marzo, V. Effects of cannabinoids and cannabinoid-enriched Cannabis extracts on TRP channels and endocannabinoid metabolic enzymes. Br. J. Pharmacol. 2011, 163, 1479–1494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Petrocellis, L.; Vellani, V.; Schiano-Moriello, A.; Marini, P.; Magherini, P.C.; Orlando, P.; Di Marzo, V. Plant-derived cannabinoids modulate the activity of transient receptor potential channels of ankyrin type-1 and melastatin type-8. J. Pharmacol. Exp. Ther. 2008, 325, 1007–1015. [Google Scholar] [CrossRef] [PubMed]
- Bisogno, T.; Hanuš, L.; De Petrocellis, L.; Tchilibon, S.; Ponde, D.E.; Brandi, I.; Moriello, A.S.; Davis, J.B.; Mechoulam, R.; Di Marzo, V. Molecular targets for cannabidiol and its synthetic analogues: Effect on vanilloid VR1 receptors and on the cellular uptake and enzymatic hydrolysis of anandamide. Br. J. Pharmacol. 2001, 134, 845–852. [Google Scholar] [CrossRef] [PubMed]
- Qin, N.; Neeper, M.P.; Liu, Y.; Hutchinson, T.L.; Lubin, M.L.; Flores, C.M. TRPV2 is activated by cannabidiol and mediates CGRP release in cultured rat dorsal root ganglion neurons. J. Neurosci. 2008, 28, 6231–6238. [Google Scholar] [CrossRef] [Green Version]
- Nabissi, M.; Morelli, M.B.; Santoni, M.; Santoni, G. Triggering of the TRPV2 channel by cannabidiol sensitizes glioblastoma cells to cytotoxic chemotherapeutic agents. Carcinogenesis 2013, 34, 48–57. [Google Scholar] [CrossRef] [Green Version]
- Yang, K.H.; Galadari, S.; Isaev, D.; Petroianu, G.; Shippenberg, T.S.; Oz, M. The nonpsychoactive cannabinoid cannabidiol inhibits 5- hydroxytryptamine3A receptor-mediated currents in xenopus laevis oocytes. J. Pharmacol. Exp. Ther. 2010, 333, 547–554. [Google Scholar] [CrossRef] [Green Version]
- Mahgoub, M.; Keun-Hang, S.Y.; Sydorenko, V.; Ashoor, A.; Kabbani, N.; Al Kury, L.; Sadek, B.; Howarth, C.F.; Isaev, D.; Galadari, S.; et al. Effects of cannabidiol on the function of α7-nicotinic acetylcholine receptors. Eur. J. Pharmacol. 2013, 720, 310–319. [Google Scholar] [CrossRef]
- Bakas, T.; van Nieuwenhuijzen, P.S.; Devenish, S.O.; McGregor, I.S.; Arnold, J.C.; Chebib, M. The direct actions of cannabidiol and 2-arachidonoyl glycerol at GABAA receptors. Pharmacol. Res. 2017, 119, 358–370. [Google Scholar] [CrossRef]
- Ahrens, J.; Demir, R.; Leuwer, M.; De La Roche, J.; Krampfl, K.; Foadi, N.; Karst, M.; Haeseler, G. The nonpsychotropic cannabinoid cannabidiol modulates and directly activates alpha-1 and alpha-1-beta glycine receptor function. Pharmacology 2009, 83, 217–222. [Google Scholar] [CrossRef]
- Ross, H.R.; Napier, I.; Connor, M. Inhibition of recombinant human T-type calcium channels by Δ9-tetrahydrocannabinol and cannabidiol. J. Biol. Chem. 2008, 283, 16124–16134. [Google Scholar] [CrossRef] [Green Version]
- Ghovanloo, M.R.; Shuart, N.G.; Mezeyova, J.; Dean, R.A.; Ruben, P.C.; Goodchild, S.J. Inhibitory effects of cannabidiol on voltage-dependent sodium currents. J. Biol. Chem. 2019, 293, 16546–16558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rimmerman, N.; Ben-Hail, D.; Porat, Z.; Juknat, A.; Kozela, E.; Daniels, M.P.; Connelly, P.S.; Leishman, E.; Bradshaw, H.B.; Shoshan-Barmatz, V.; et al. Direct modulation of the outer mitochondrial membrane channel, voltage-dependent anion channel 1 (VDAC1) by cannabidiol: A novel mechanism for cannabinoid-induced cell death. Cell Death Dis. 2013, 4, e949. [Google Scholar] [CrossRef] [PubMed]
- Elmes, M.W.; Kaczocha, M.; Berger, W.T.; Leung, K.; Ralph, B.P.; Wang, L.; Sweeney, J.M.; Miyauchi, J.T.; Tsirka, S.E.; Ojima, I.; et al. Fatty Acid-binding Proteins (FABPs) Are Intracellular Carriers for Δ9-Tetrahydrocannabinol (THC) and Cannabidiol (CBD). J. Biol. Chem. 2015, 290, 8711–8721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, H.; McIntosh, A.L.; Martin, G.G.; Landrock, D.; Chung, S.; Landrock, K.K.; Dangott, L.J.; Li, S.; Kier, A.B.; Schroeder, F. FABP1: A Novel Hepatic Endocannabinoid and Cannabinoid Binding Protein. Biochemistry 2016, 55, 5243–5255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leishman, E.; Manchanda, M.; Thelen, R.; Miller, S.; Mackie, K.; Bradshaw, H.B. Cannabidiol’s Upregulation of N-acyl Ethanolamines in the Central Nervous System Requires N-acyl Phosphatidyl Ethanolamine-Specific Phospholipase D. Cannabis Cannabinoid Res. 2018, 3, 228–241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leweke, F.M.; Piomelli, D.; Pahlisch, F.; Muhl, D.; Gerth, C.W.; Hoyer, C.; Klosterkötter, J.; Hellmich, M.; Koethe, D. Cannabidiol enhances anandamide signaling and alleviates psychotic symptoms of schizophrenia. Transl. Psychiatry 2012, 2, e94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carrier, E.J.; Auchampach, J.A.; Hillard, C.J. Inhibition of an equilibrative nucleoside transporter by cannabidiol: A mechanism of cannabinoid immunosuppression. Proc. Natl. Acad. Sci. USA 2006, 103, 7895–7900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mijangos-Moreno, S.; Poot-Aké, A.; Arankowsky-Sandoval, G.; Murillo-Rodríguez, E. Intrahypothalamic injection of cannabidiol increases the extracellular levels of adenosine in nucleus accumbens in rats. Neurosci. Res. 2014, 84, 60–63. [Google Scholar] [CrossRef]
- Takeda, S.; Usami, N.; Yamamoto, I.; Watanabe, K. Cannabidiol-2′,6′-dimethyl ether, a cannabidiol derivative, is a highly potent and selective 15-lipoxygenase inhibitor. Drug Metab. Dispos. 2009, 37, 1733–1737. [Google Scholar] [CrossRef] [Green Version]
- Wheal, A.J.; Cipriano, M.; Fowler, C.J.; Randall, M.D.; O’Sullivan, S.E. Cannabidiol improves vasorelaxation in Zucker diabetic fatty rats through cyclooxygenase activation. J. Pharmacol. Exp. Ther. 2014, 351, 457–466. [Google Scholar] [CrossRef] [Green Version]
- Evans, A.T.; Formukong, E.; Evans, F.J. Activation of phospholipase A2 by cannabinoids. Lack of correlation with CNS effects. FEBS Lett. 1987, 211, 119–122. [Google Scholar] [CrossRef] [Green Version]
- Fišar, Z.; Singh, N.; Hroudová, J. Cannabinoid-induced changes in respiration of brain mitochondria. Toxicol. Lett. 2014, 231, 62–71. [Google Scholar] [CrossRef] [PubMed]
- Koch, M.; Dehghani, F.; Habazettl, I.; Schomerus, C.; Korf, H.W. Cannabinoids attenuate norepinephrine-induced melatonin biosynthesis in the rat pineal gland by reducing arylalkylamine N-acetyltransferase activity without involvement of cannabinoid receptors. J. Neurochem. 2006, 98, 267–278. [Google Scholar] [CrossRef] [PubMed]
- Jenny, M.; Santer, E.; Pirich, E.; Schennach, H.; Fuchs, D. Δ9-Tetrahydrocannabinol and cannabidiol modulate mitogen-induced tryptophan degradation and neopterin formation in peripheral blood mononuclear cells in vitro. J. Neuroimmunol. 2009, 207, 75–82. [Google Scholar] [CrossRef]
- Cornicelli, J.A.; Gilman, S.R.; Krom, B.A.; Kottke, B.A. Cannabinoids impair the formation of cholesteryl ester in cultured human cells. Arteriosclerosis 1981, 1, 449–454. [Google Scholar] [CrossRef] [Green Version]
- Watanabe, K.; Motoya, E.; Matsuzawa, N.; Funahashi, T.; Kimura, T.; Matsunaga, T.; Arizono, K.; Yamamoto, I. Marijuana extracts possess the effects like the endocrine disrupting chemicals. Toxicology 2005, 206, 471–478. [Google Scholar] [CrossRef]
- Granja, A.G.; Carrillo-Salinas, F.; Pagani, A.; Gómez-Cañas, M.; Negri, R.; Navarrete, C.; Mecha, M.; Mestre, L.; Fiebich, B.L.; Cantarero, I.; et al. A cannabigerol quinone alleviates neuroinflammation in a chronic model of multiple sclerosis. J. Neuroimmune Pharmacol. 2012, 7, 1002–1016. [Google Scholar] [CrossRef]
- Hegde, V.L.; Singh, U.P.; Nagarkatti, P.S.; Nagarkatti, M. Critical Role of Mast Cells and Peroxisome Proliferator–Activated Receptor γ in the Induction of Myeloid-Derived Suppressor Cells by Marijuana Cannabidiol In Vivo. J. Immunol. 2015, 194, 5211–5222. [Google Scholar] [CrossRef] [Green Version]
- Juknat, A.; Pietr, M.; Kozela, E.; Rimmerman, N.; Levy, R.; Gao, F.; Coppola, G.; Geschwind, D.; Vogel, Z. Microarray and Pathway Analysis Reveal Distinct Mechanisms Underlying Cannabinoid-Mediated Modulation of LPS-Induced Activation of BV-2 Microglial Cells. PLoS ONE 2013, 8, e61462. [Google Scholar] [CrossRef] [Green Version]
- McPartland, J.M.; Duncan, M.; Di Marzo, V.; Pertwee, R.G. Are cannabidiol and Δ9-tetrahydrocannabivarin negative modulators of the endocannabinoid system? A systematic review. Br. J. Pharmacol. 2015, 172, 737–753. [Google Scholar] [CrossRef] [Green Version]
- Maione, S.; Piscitelli, F.; Gatta, L.; Vita, D.; De Petrocellis, L.; Palazzo, E.; De Novellis, V.; Di Marzo, V. Non-psychoactive cannabinoids modulate the descending pathway of antinociception in anaesthetized rats through several mechanisms of action. Br. J. Pharmacol. 2011, 162, 584–596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Britch, S.C.; Wiley, J.L.; Yu, Z.; Clowers, B.H.; Craft, R.M. Cannabidiol-Δ9-tetrahydrocannabinol interactions on acute pain and locomotor activity. Drug Alcohol Depend. 2017, 175, 187–197. [Google Scholar] [CrossRef] [PubMed]
- Greene, N.Z.; Wiley, J.L.; Yu, Z.; Clowers, B.H.; Craft, R.M. Cannabidiol modulation of antinociceptive tolerance to Δ9-tetrahydrocannabinol. Psychopharmacology 2018, 235, 3289–3302. [Google Scholar] [CrossRef] [PubMed]
- Gonçalves, T.C.T.; Londe, A.K.; Albano, R.I.P.; De Araújo Júnior, A.T.; De Aguiar Azeredo, M.; Biagioni, A.F.; Vasconcellos, T.H.F.; Dos Reis Ferreira, C.M.; Teixeira, D.G.; De Souza Crippa, J.A.; et al. Cannabidiol and endogenous opioid peptide-mediated mechanisms modulate antinociception induced by transcutaneous electrostimulation of the peripheral nervous system. J. Neurol. Sci. 2014, 347, 82–89. [Google Scholar] [CrossRef] [PubMed]
- Abraham, A.D.; Leung, E.J.Y.; Wong, B.A.; Rivera, Z.M.G.; Kruse, L.C.; Clark, J.J.; Land, B.B. Orally consumed cannabinoids provide long-lasting relief of allodynia in a mouse model of chronic neuropathic pain. Neuropsychopharmacology 2020, 45, 1105–1114. [Google Scholar] [CrossRef] [PubMed]
- Xiong, W.; Cui, T.; Cheng, K.; Yang, F.; Chen, S.R.; Willenbring, D.; Guan, Y.; Pan, H.L.; Ren, K.; Xu, Y.; et al. Cannabinoids suppress inflammatory and neuropathic pain by targeting α3 glycine receptors. J. Exp. Med. 2012, 209, 1121–1134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vigil, J.M.; Montera, M.A.; Pentkowski, N.S.; Diviant, J.P.; Orozco, J.; Ortiz, A.L.; Rael, L.J.; Westlund, K.N. The therapeutic effectiveness of full spectrum hemp oil using a chronic neuropathic pain model. Life 2020, 10, 69. [Google Scholar] [CrossRef]
- Casey, S.L.; Atwal, N.; Vaughan, C.W. Cannabis constituent synergy in a mouse neuropathic pain model. Pain 2017, 158, 2452–2460. [Google Scholar] [CrossRef]
- Li, H.; Kong, W.; Chambers, C.R.; Yu, D.; Ganea, D.; Tuma, R.F.; Ward, S.J. The non-psychoactive phytocannabinoid cannabidiol (CBD) attenuates pro-inflammatory mediators, T cell infiltration, and thermal sensitivity following spinal cord injury in mice. Cell. Immunol. 2018, 329, 1–9. [Google Scholar] [CrossRef]
- Linher-Melville, K.; Zhu, Y.F.; Sidhu, J.; Parzei, N.; Shahid, A.; Seesankar, G.; Ma, D.; Wang, Z.; Zacal, N.; Sharma, M.; et al. Evaluation of the preclinical analgesic efficacy of naturally derived, orally administered oil forms of Δ9-tetrahydrocannabinol (THC), cannabidiol (CBD), and their 1:1 combination. PLoS ONE 2020, 15, e0234176. [Google Scholar] [CrossRef]
- De Gregorio, D.; McLaughlin, R.J.; Posa, L.; Ochoa-Sanchez, R.; Enns, J.; Lopez-Canul, M.; Aboud, M.; Maione, S.; Comai, S.; Gobbi, G. Cannabidiol modulates serotonergic transmission and reverses both allodynia and anxiety-like behavior in a model of neuropathic pain. Pain 2019, 160, 136–150. [Google Scholar] [CrossRef] [PubMed]
- Ward, S.J.; Ramirez, M.D.; Neelakantan, H.; Walker, E.A. Cannabidiol prevents the development of cold and mechanical allodynia in paclitaxel-treated female c57bl6 mice. Anesth. Analg. 2011, 113, 947–950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ward, S.J.; McAllister, S.D.; Kawamura, R.; Murase, R.; Neelakantan, H.; Walker, E.A. Cannabidiol inhibits paclitaxel-induced neuropathic pain through 5-HT 1A receptors without diminishing nervous system function or chemotherapy efficacy. Br. J. Pharmacol. 2014, 171, 636–645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- King, K.M.; Myers, A.M.; Soroka-Monzo, A.J.; Tuma, R.F.; Tallarida, R.J.; Walker, E.A.; Ward, S.J. Single and combined effects of Δ9-tetrahydrocannabinol and cannabidiol in a mouse model of chemotherapy-induced neuropathic pain. Br. J. Pharmacol. 2017, 174, 2832–2841. [Google Scholar] [CrossRef] [Green Version]
- Harris, H.M.; Sufka, K.J.; Gul, W.; Elsohly, M.A. Effects of Delta-9-Tetrahydrocannabinol and Cannabidiol on Cisplatin-Induced Neuropathy in Mice. Planta Med. 2016, 82, 1169–1172. [Google Scholar] [CrossRef] [Green Version]
- Lehmann, C.; Fisher, N.B.; Tugwell, B.; Szczesniak, A.; Kelly, M.; Zhou, J. Experimental cannabidiol treatment reduces early pancreatic inflammation in type 1 diabetes. Clin. Hemorheol. Microcirc. 2016, 64, 655–662. [Google Scholar] [CrossRef]
- Rajesh, M.; Mukhopadhyay, P.; Btkai, S.; Patel, V.; Saito, K.; Matsumoto, S.; Kashiwaya, Y.; Horvth, B.; Mukhopadhyay, B.; Becker, L.; et al. Cannabidiol attenuates cardiac dysfunction, oxidative stress, fibrosis, and inflammatory and cell death signaling pathways in diabetic cardiomyopathy. J. Am. Coll. Cardiol. 2010, 56, 2115–2125. [Google Scholar] [CrossRef] [Green Version]
- Toth, C.C.; Jedrzejewski, N.M.; Ellis, C.L.; Frey, W.H. Cannabinoid-Mediated Modulation of Neuropathic Pain and Microglial Accumulation in a Model of Murine Type I Diabetic Peripheral Neuropathic Pain. Mol. Pain 2010, 6. [Google Scholar] [CrossRef] [Green Version]
- Jesus, C.H.A.; Redivo, D.D.B.; Gasparin, A.T.; Sotomaior, B.B.; de Carvalho, M.C.; Genaro, K.; Zuardi, A.W.; Hallak, J.E.C.; Crippa, J.A.; Zanoveli, J.M.; et al. Cannabidiol attenuates mechanical allodynia in streptozotocin-induced diabetic rats via serotonergic system activation through 5-HT1A receptors. Brain Res. 2019, 1715, 156–164. [Google Scholar] [CrossRef]
- Vasko, M.R. I Ia Inhibitory Interneuron Definition. In Inflammatory Pain; Binder, M.D., Hirokawa, N., Windhorst, U., Eds.; Encyclopedia of Neuroscience; Springer: Berlin/Heidelberg, Germany, 2009. [Google Scholar]
- Verrico, C.D.; Wesson, S.; Konduri, V.; Hofferek, C.J.; Vazquez-Perez, J.; Blair, E.; Dunner, K.; Salimpour, P.; Decker, W.K.; Halpert, M.M. A randomized, double-blind, placebo-controlled study of daily cannabidiol for the treatment of canine osteoarthritis pain. Pain 2020, 161, 2191–2202. [Google Scholar] [CrossRef]
- Britch, S.C.; Goodman, A.G.; Wiley, J.L.; Pondelick, A.M.; Craft, R.M. Antinociceptive and Immune Effects of Delta-9-Tetrahydrocannabinol or Cannabidiol in Male Versus Female Rats with Persistent Inflammatory Pain. J. Pharmacol. Exp. Ther. 2020, 373, 416–428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karmaus, P.W.F.; Wagner, J.G.; Harkema, J.R.; Kaminski, N.E.; Kaplan, B.L.F. Cannabidiol (CBD) enhances lipopolysaccharide (LPS)-induced pulmonary inflammation in C57BL/6 mice. J. Immunotoxicol. 2013, 10, 321–328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rock, E.M.; Limebeer, C.L.; Parker, L.A. Effect of cannabidiolic acid and ∆9-tetrahydrocannabinol on carrageenan-induced hyperalgesia and edema in a rodent model of inflammatory pain. Psychopharmacology 2018, 235, 3259–3271. [Google Scholar] [CrossRef] [PubMed]
- Moreno-Martet, M.; Feliú, A.; Espejo-Porras, F.; Mecha, M.; Carrillo-Salinas, F.J.; Fernández-Ruiz, J.; Guaza, C.; De Lago, E. The disease-modifying effects of a Sativex-like combination of phytocannabinoids in mice with experimental autoimmune encephalomyelitis are preferentially due to Δ9-tetrahydrocannabinol acting through CB1 receptors. Mult. Scler. Relat. Disord. 2015, 4, 505–511. [Google Scholar] [CrossRef] [Green Version]
- Gamble, L.J.; Boesch, J.M.; Frye, C.W.; Schwark, W.S.; Mann, S.; Wolfe, L.; Brown, H.; Berthelsen, E.S.; Wakshlag, J.J. Pharmacokinetics, Safety, and Clinical Efficacy of Cannabidiol Treatment in Osteoarthritic Dogs. Front. Vet. Sci. 2018, 5, 165. [Google Scholar] [CrossRef] [Green Version]
- Philpott, H.T.; O’Brien, M.; McDougall, J.J. Attenuation of early phase inflammation by cannabidiol prevents pain and nerve damage in rat osteoarthritis. Pain 2017, 158, 2442–2451. [Google Scholar] [CrossRef]
- Malfait, A.M.; Gallily, R.; Sumariwalla, P.F.; Malik, A.S.; Andreakos, E.; Mechoulam, R.; Feldmann, M. The nonpsychoactive cannabis constituent cannabidiol is an oral anti-arthritic therapeutic in murine collagen-induced arthritis. Proc. Natl. Acad. Sci. USA 2000, 97, 9561–9566. [Google Scholar] [CrossRef] [Green Version]
- Lowin, T.; Tingting, R.; Zurmahr, J.; Classen, T.; Schneider, M.; Pongratz, G. Cannabidiol (CBD): A killer for inflammatory rheumatoid arthritis synovial fibroblasts. Cell Death Dis. 2020, 11, 1–11. [Google Scholar] [CrossRef]
- Winklmayr, M.; Gaisberger, M.; Kittl, M.; Fuchs, J.; Ritter, M.; Jakab, M. Dose-Dependent Cannabidiol-Induced Elevation of Intracellular Calcium and Apoptosis in Human Articular Chondrocytes. J. Orthop. Res. 2019, 37, 2540–2549. [Google Scholar] [CrossRef] [Green Version]
- Genaro, K.; Fabris, D.; Arantes, A.L.F.; Zuardi, A.W.; Crippa, J.A.S.; Prado, W.A. Cannabidiol is a potential therapeutic for the affective-motivational dimension of incision pain in rats. Front. Pharmacol. 2017, 8, 391. [Google Scholar] [CrossRef] [Green Version]
- Wong, H.; Cairns, B.E. Cannabidiol, cannabinol and their combinations act as peripheral analgesics in a rat model of myofascial pain. Arch. Oral Biol. 2019, 104, 33–39. [Google Scholar] [CrossRef] [PubMed]
- Rodríguez-Muñoz, M.; Onetti, Y.; Cortés-Montero, E.; Garzón, J.; Sánchez-Blázquez, P. Cannabidiol enhances morphine antinociception, diminishes NMDA-mediated seizures and reduces stroke damage via the sigma 1 receptor 11 Medical and Health Sciences 1109 Neurosciences 11 Medical and Health Sciences 1115 Pharmacology and Pharmaceutical Scien. Mol. Brain 2018, 11, 1–12. [Google Scholar]
- Neelakantan, H.; Tallarida, R.J.; Reichenbach, Z.W.; Tuma, R.F.; Ward, S.J.; Walker, E.A. Distinct interactions of cannabidiol and morphine in three nociceptive behavioral models in mice. Behav. Pharmacol. 2015, 26, 304–314. [Google Scholar] [CrossRef]
- Crivelaro do Nascimento, G.; Ferrari, D.P.; Guimaraes, F.S.; Del Bel, E.A.; Bortolanza, M.; Ferreira, N.C., Jr. Cannabidiol increases the nociceptive threshold in a preclinical model of Parkinson’s disease. Neuropharmacology 2020, 163, 107808. [Google Scholar] [CrossRef] [PubMed]
- Finn, D.P.; Beckett, S.R.G.; Roe, C.H.; Madjd, A.; Fone, K.C.F.; Kendall, D.A.; Marsden, C.A.; Chapman, V. Effects of coadministration of cannabinoids and morphine on nociceptive behaviour, brain monoamines and HPA axis activity in a rat model of persistent pain. Eur. J. Neurosci. 2004, 19, 678–686. [Google Scholar] [CrossRef]
- Cunha, J.M.; Carlini, E.A.; Pereira, A.E.; Ramos, O.L.; Pimentel, C.; Gagliardi, R.; Sanvito, W.L.; Lander, N.; Mechoulam, R. Chronic administration of cannabidiol to healthy volunteers and epileptic patients. Pharmacology 1980, 21, 175–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Consroe, P.; Laguna, J.; Allender, J.; Snider, S.; Stern, L.; Sandyk, R.; Kennedy, K.; Schram, K. Controlled clinical trial of cannabidiol in Huntington’s disease. Pharmacol. Biochem. Behav. 1991, 40, 701–708. [Google Scholar] [CrossRef]
- Wade, D.T.; Makela, P.; Robson, P.; House, H.; Bateman, C. Do cannabis-based medicinal extracts have general or specific effects on symptoms in multiple sclerosis? A double-blind, randomized, placebo-controlled study on 160 patients. Mult. Scler. 2004, 10, 434–441. [Google Scholar] [CrossRef]
- Cuñetti, L.; Manzo, L.; Peyraube, R.; Arnaiz, J.; Curi, L.; Orihuela, S. Chronic Pain Treatment With Cannabidiol in Kidney Transplant Patients in Uruguay. Transplant. Proc. 2018, 50, 461–464. [Google Scholar] [CrossRef]
- Taylor, L.; Gidal, B.; Blakey, G.; Tayo, B.; Morrison, G. A Phase I, Randomized, Double-Blind, Placebo-Controlled, Single Ascending Dose, Multiple Dose, and Food Effect Trial of the Safety, Tolerability and Pharmacokinetics of Highly Purified Cannabidiol in Healthy Subjects. CNS Drugs 2018, 32, 1053–1067. [Google Scholar] [CrossRef] [Green Version]
- Devinsky, O.; Patel, A.D.; Thiele, E.A.; Wong, M.H.; Appleton, R.; Harden, C.L.; Greenwood, S.; Morrison, G.; Sommerville, K. Randomized, dose-ranging safety trial of cannabidiol in Dravet syndrome. Neurology 2018, 90, e1204–e1211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Manini, A.F.; Yiannoulos, G.; Bergamaschi, M.M.; Hernandez, S.; Olmedo, R.; Barnes, A.J.; Winkel, G.; Sinha, R.; Jutras-Aswad, D.; Huestis, M.A.; et al. Safety and pharmacokinetics of oral Cannabidiol when administered concomitantly with intravenous Fentanyl in humans. J. Addict. Med. 2015, 9, 204. [Google Scholar] [CrossRef] [PubMed]
- Xu, D.H.; Cullen, B.D.; Tang, M.; Fang, Y. The Effectiveness of Topical Cannabidiol Oil in Symptomatic Relief of Peripheral Neuropathy of the Lower Extremities. Curr. Pharm. Biotechnol. 2019, 21, 390–402. [Google Scholar] [CrossRef] [PubMed]
- Szaflarski, J.P.; Bebin, E.M.; Cutter, G.; DeWolfe, J.; Dure, L.S.; Gaston, T.E.; Kankirawatana, P.; Liu, Y.; Singh, R.; Standaert, D.G.; et al. Cannabidiol improves frequency and severity of seizures and reduces adverse events in an open-label add-on prospective study. Epilepsy Behav. 2018, 87, 131–136. [Google Scholar] [CrossRef] [Green Version]
- Devinsky, O.; Patel, A.D.; Cross, J.H.; Villanueva, V.; Wirrell, E.C.; Privitera, M.; Greenwood, S.M.; Roberts, C.; Checketts, D.; VanLandingham, K.E.; et al. Effect of cannabidiol on drop seizures in the lennox–gastaut syndrome. N. Engl. J. Med. 2018, 378, 1888–1897. [Google Scholar] [CrossRef] [Green Version]
- Thiele, E.A.; Marsh, E.D.; French, J.A.; Mazurkiewicz, M.B.; Benbadis, S.R.; Joshi, C.; Lyons, P.D.; Taylor, A.; Roberts, C.; Sommerville, K.; et al. Cannabidiol in patients with seizures associated with Lennox-Gastaut syndrome (GWPCARE4): A randomised, double-blind, placebo-controlled phase 3 trial. Lancet 2018, 391, 1085–1096. [Google Scholar] [CrossRef]
- Laux, L.C.; Bebin, E.M.; Checketts, D.; Chez, M.; Flamini, R.; Marsh, E.D.; Miller, I.; Nichol, K.; Park, Y.; Segal, E.; et al. Long-term safety and efficacy of cannabidiol in children and adults with treatmentresistant Lennox-Gastaut syndrome or Dravet syndrome: Expanded access program results. Epilepsy Res. 2019, 154, 13–20. [Google Scholar] [CrossRef]
- Devinsky, O.; Cross, J.H.; Laux, L.; Marsh, E.; Miller, I.; Nabbout, R.; Scheffer, I.E.; Thiele, E.A.; Wright, S. Trial of cannabidiol for drug-resistant seizures in the dravet syndrome. N. Engl. J. Med. 2017, 376, 2011–2020. [Google Scholar] [CrossRef] [Green Version]
- Capano, A.; Weaver, R.; Burkman, E. Evaluation of the effects of CBD hemp extract on opioid use and quality of life indicators in chronic pain patients: A prospective cohort study. Postgrad. Med. 2020, 132, 56–61. [Google Scholar] [CrossRef] [Green Version]
- Van De Donk, T.; Niesters, M.; Kowal, M.A.; Olofsen, E.; Dahan, A.; Van Velzen, M. An experimental randomized study on the analgesic effects of pharmaceutical-grade cannabis in chronic pain patients with fibromyalgia. Pain 2019, 160, 860–869. [Google Scholar] [CrossRef]
- Hoggart, B.; Ratcliffe, S.; Ehler, E.; Simpson, K.H.; Hovorka, J.; Lejčko, J.; Taylor, L.; Lauder, H.; Serpell, M. A multicentre, open-label, follow-on study to assess the long-term maintenance of effect, tolerance and safety of THC/CBD oromucosal spray in the management of neuropathic pain. J. Neurol. 2015, 262, 27–40. [Google Scholar] [CrossRef] [PubMed]
- Serpell, M.; Ratcliffe, S.; Hovorka, J.; Schofield, M.; Taylor, L.; Lauder, H.; Ehler, E. A double-blind, randomized, placebo-controlled, parallel group study of THC/CBD spray in peripheral neuropathic pain treatment. Eur. J. Pain 2014, 18, 999–1012. [Google Scholar] [CrossRef] [PubMed]
- Johnson, J.R.; Burnell-Nugent, M.; Lossignol, D.; Ganae-Motan, E.D.; Potts, R.; Fallon, M.T. Multicenter, Double-Blind, Randomized, Placebo-Controlled, Parallel-Group Study of the Efficacy, Safety, and Tolerability of THC:CBD Extract and THC Extract in Patients with Intractable Cancer-Related Pain. J. Pain Symptom Manag. 2010, 39, 167–179. [Google Scholar] [CrossRef] [PubMed]
- Johnson, J.R.; Lossignol, D.; Burnell-Nugent, M.; Fallon, M.T. An open-label extension study to investigate the long-term safety and tolerability of THC/CBD oromucosal spray and oromucosal THC spray in patients with terminal cancer-related pain refractory to strong opioid analgesics. J. Pain Symptom Manag. 2013, 46, 207–218. [Google Scholar] [CrossRef]
- Russo, M.; Naro, A.; Leo, A.; Sessa, E.; D’Aleo, G.; Bramanti, P.; Calabrò, R.S. Evaluating sativex® in neuropathic pain management: A clinical and neurophysiological assessment in multiple sclerosis. Pain Med. 2016, 17, 1145–1154. [Google Scholar] [CrossRef] [Green Version]
- Portenoy, R.K.; Ganae-Motan, E.D.; Allende, S.; Yanagihara, R.; Shaiova, L.; Weinstein, S.; McQuade, R.; Wright, S.; Fallon, M.T. Nabiximols for opioid-treated cancer patients with poorly-controlled chronic pain: A randomized, placebo-controlled, graded-dose trial. J. Pain 2012, 13, 438–449. [Google Scholar] [CrossRef]
- Nurmikko, T.J.; Serpell, M.G.; Hoggart, B.; Toomey, P.J.; Morlion, B.J.; Haines, D. Sativex successfully treats neuropathic pain characterised by allodynia: A randomised, double-blind, placebo-controlled clinical trial. Pain 2007, 133, 210–220. [Google Scholar] [CrossRef]
- Blake, D.R.; Robson, P.; Ho, M.; Jubb, R.W.; McCabe, C.S. Preliminary assessment of the efficacy, tolerability and safety of a cannabis-based medicine (Sativex) in the treatment of pain caused by rheumatoid arthritis. Rheumatology 2006, 45, 50–52. [Google Scholar] [CrossRef] [Green Version]
- Lichtman, A.H.; Lux, E.A.; McQuade, R.; Rossetti, S.; Sanchez, R.; Sun, W.; Wright, S.; Kornyeyeva, E.; Fallon, M.T. Results of a Double-Blind, Randomized, Placebo-Controlled Study of Nabiximols Oromucosal Spray as an Adjunctive Therapy in Advanced Cancer Patients with Chronic Uncontrolled Pain. J. Pain Symptom Manag. 2018, 55, 179–188. [Google Scholar] [CrossRef] [Green Version]
- Lynch, M.E.; Cesar-Rittenberg, P.; Hohmann, A.G. A double-blind, placebo-controlled, crossover pilot trial with extension using an oral mucosal cannabinoid extract for treatment of chemotherapy-induced neuropathic pain. J. Pain Symptom Manag. 2014, 47, 166–173. [Google Scholar] [CrossRef]
- Santoro, M.; Mirabella, M.; De Fino, C.; Bianco, A.; Lucchini, M.; Losavio, F.; Sabino, A.; Nociti, V. Sativex® effects on promoter methylation and on CNR1/CNR2 expression in peripheral blood mononuclear cells of progressive multiple sclerosis patients. J. Neurol. Sci. 2017, 379, 298–303. [Google Scholar] [CrossRef] [PubMed]
- Martin-Santos, R.A.; Crippa, J.; 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]
- Schoedel, K.A.; Szeto, I.; Setnik, B.; Sellers, E.M.; Levy-Cooperman, N.; Mills, C.; Etges, T.; Sommerville, K. Abuse potential assessment of cannabidiol (CBD) in recreational polydrug users: A randomized, double-blind, controlled trial. Epilepsy Behav. 2018, 88, 162–171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ignatowska-Jankowska, B.; Jankowski, M.; Glac, W.; Swiergiel, A.H. Cannabidiol-induced lymphopenia does not involve NKT and NK cells. J. Physiol. Pharmacol. 2009, 60, 99–103. [Google Scholar] [PubMed]
- Calvey, T.N.; Williams, N.E. Principles and Practice of Pharmacology for Anaesthetists; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2008. [Google Scholar]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Mlost, J.; Bryk, M.; Starowicz, K. Cannabidiol for Pain Treatment: Focus on Pharmacology and Mechanism of Action. Int. J. Mol. Sci. 2020, 21, 8870. https://doi.org/10.3390/ijms21228870
Mlost J, Bryk M, Starowicz K. Cannabidiol for Pain Treatment: Focus on Pharmacology and Mechanism of Action. International Journal of Molecular Sciences. 2020; 21(22):8870. https://doi.org/10.3390/ijms21228870
Chicago/Turabian StyleMlost, Jakub, Marta Bryk, and Katarzyna Starowicz. 2020. "Cannabidiol for Pain Treatment: Focus on Pharmacology and Mechanism of Action" International Journal of Molecular Sciences 21, no. 22: 8870. https://doi.org/10.3390/ijms21228870
APA StyleMlost, J., Bryk, M., & Starowicz, K. (2020). Cannabidiol for Pain Treatment: Focus on Pharmacology and Mechanism of Action. International Journal of Molecular Sciences, 21(22), 8870. https://doi.org/10.3390/ijms21228870