دورية أكاديمية

Cannabis constituents for chronic neuropathic pain; reconciling the clinical and animal evidence.

التفاصيل البيبلوغرافية
العنوان: Cannabis constituents for chronic neuropathic pain; reconciling the clinical and animal evidence.
المؤلفون: Sokolaj E; Pain Management Research Institute, Kolling Institute of Medical Research, Northern Clinical School, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales, Australia., Assareh N; Pain Management Research Institute, Kolling Institute of Medical Research, Northern Clinical School, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales, Australia., Anderson K; Pain Management Research Institute, Kolling Institute of Medical Research, Northern Clinical School, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales, Australia., Aubrey KR; Pain Management Research Institute, Kolling Institute of Medical Research, Northern Clinical School, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales, Australia., Vaughan CW; Pain Management Research Institute, Kolling Institute of Medical Research, Northern Clinical School, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales, Australia.
المصدر: Journal of neurochemistry [J Neurochem] 2023 Sep 25. Date of Electronic Publication: 2023 Sep 25.
Publication Model: Ahead of Print
نوع المنشور: Journal Article; Review
اللغة: English
بيانات الدورية: Publisher: Wiley on behalf of the International Society for Neurochemistry Country of Publication: England NLM ID: 2985190R Publication Model: Print-Electronic Cited Medium: Internet ISSN: 1471-4159 (Electronic) Linking ISSN: 00223042 NLM ISO Abbreviation: J Neurochem Subsets: MEDLINE
أسماء مطبوعة: Publication: 2001- : Oxford, UK : Wiley on behalf of the International Society for Neurochemistry
Original Publication: New York : Raven Press
مستخلص: Chronic neuropathic pain is a debilitating pain syndrome caused by damage to the nervous system that is poorly served by current medications. Given these problems, clinical studies have pursued extracts of the plant Cannabis sativa as alternative treatments for this condition. The vast majority of these studies have examined cannabinoids which contain the psychoactive constituent delta-9-tetrahydrocannabinol (THC). While there have been some positive findings, meta-analyses of this clinical work indicates that this effectiveness is limited and hampered by side-effects. This review focuses on how recent preclinical studies have predicted the clinical limitations of THC-containing cannabis extracts, and importantly, point to how they might be improved. This work highlights the importance of targeting channels and receptors other than cannabinoid CB1 receptors which mediate many of the side-effects of cannabis.
(© 2023 International Society for Neurochemistry.)
References: Abrams, D. I., Jay, C. A., Shade, S. B., Vizoso, H., Reda, H., Press, S., Kelly, M. E., Rowbotham, M. C., & Petersen, K. L. (2007). Cannabis in painful HIV-associated sensory neuropathy: A randomized placebo-controlled trial. Neurology, 68, 515-521.
Adamson Barnes, N. S., Mitchell, V. A., Kazantzis, N. P., & Vaughan, C. W. (2016). Actions of the dual FAAH/MAGL inhibitor JZL195 in a murine neuropathic pain model. British Journal of Pharmacology, 173, 77-87.
Ahn, K., Johnson, D. S., Mileni, M., Beidler, D., Long, J. Z., McKinney, M. K., Weerapana, E., Sadagopan, N., Liimatta, M., Smith, S. E., Lazerwith, S., Stiff, C., Kamtekar, S., Bhattacharya, K., Zhang, Y., Swaney, S., van Becelaere, K., Stevens, R. C., & Cravatt, B. F. (2009). Discovery and characterization of a highly selective FAAH inhibitor that reduces inflammatory pain. Chemistry & Biology, 16, 411-420.
Anavi-Goffer, S., Baillie, G., Irving, A. J., Gertsch, J., Greig, I. R., Pertwee, R. G., & Ross, R. A. (2012). Modulation of L-α-lysophosphatidylinositol/GPR55 mitogen-activated protein kinase (MAPK) signaling by cannabinoids. The Journal of Biological Chemistry, 287, 91-104.
Arora, V., Li, T., Kumari, S., Wang, S., Asgar, J., & Chung, M. K. (2022). Capsaicin-induced depolymerization of axonal microtubules mediates analgesia for trigeminal neuropathic pain. Pain, 163, 1479-1488.
Aviram, J., & Samuelly-Leichtag, G. (2017). Efficacy of cannabis-based medicines for pain management: A systematic review and meta-analysis of randomized controlled trials. Pain Physician, 20, E755-E796.
Bakas, T., van Nieuwenhuijzen, P. S., Devenish, S. O., McGregor, I. S., Arnold, J. C., & Chebib, M. (2017). The direct actions of cannabidiol and 2-arachidonoyl glycerol at GABA(a) receptors. Pharmacological Research: The Official Journal of the Italian Pharmacological Society, 119, 358-370.
Berman, J. S., Symonds, C., & Birch, R. (2004). Efficacy of two cannabis based medicinal extracts for relief of central neuropathic pain from brachial plexus avulsion: Results of a randomised controlled trial. Pain, 112, 299-306.
Bialas, P., Fitzcharles, M. A., Klose, P., & Hauser, W. (2022). Long-term observational studies with cannabis-based medicines for chronic non-cancer pain: A systematic review and meta-analysis of effectiveness and safety. European Journal of Pain, 26, 1221-1233.
Blankman, J. L., & Cravatt, B. F. (2013). Chemical probes of endocannabinoid metabolism. Pharmacological Reviews, 65, 849-871.
Boggs, D. L., Nguyen, J. D., Morgenson, D., Taffe, M. A., & Ranganathan, M. (2018). Clinical and preclinical evidence for functional interactions of Cannabidiol and Δ9-tetrahydrocannabinol. Neuropsychopharmacology, 43, 142-154.
Borgelt, L. M., Franson, K. L., Nussbaum, A. M., & Wang, G. S. (2013). The pharmacologic and clinical effects of medical cannabis. Pharmacotherapy, 33, 195-209.
Buggy, D. J., Toogood, L., Maric, S., Sharpe, P., Lambert, D. G., & Rowbotham, D. J. (2003). Lack of analgesic efficacy of oral delta-9-tetrahydrocannabinol in postoperative pain. Pain, 106, 169-172.
Cai, S., Gomez, K., Moutal, A., & Khanna, R. (2021). Targeting T-type/CaV3.2 channels for chronic pain. Translational Research, 234, 20-30.
Canavan, C., Inoue, T., McMahon, S., Doody, C., Blake, C., & Fullen, B. M. (2022). The efficacy, adverse events, and withdrawal rates of the pharmacological management of chronic spinal cord injury pain: A systematic review and meta-analysis. Pain Medicine, 23, 375-395.
Carey, L. M., Gutierrez, T., Deng, L., Lee, W. H., Mackie, K., & Hohmann, A. G. (2017). Inflammatory and neuropathic nociception is preserved in GPR55 knockout mice. Scientific Reports, 7, 944.
Casey, S. L., Atwal, N., & Vaughan, C. W. (2017). Cannabis constituent synergy in a mouse neuropathic pain model. Pain, 158, 2452-2460.
Casey, S. L., Mitchell, V. A., Sokolaj, E. E., Winters, B. L., & Vaughan, C. W. (2022). Intrathecal actions of the cannabis constituents delta(9)-tetrahydrocannabinol and cannabidiol in a mouse neuropathic pain model. International Journal of Molecular Sciences, 23, 3.
Castillo, P. E., Younts, T. J., Chavez, A. E., & Hashimotodani, Y. (2012). Endocannabinoid signaling and synaptic function. Neuron, 76, 70-81.
Castillo-Arellano, J., Canseco-Alba, A., Cutler, S. J., & Leon, F. (2023). The polypharmacological effects of cannabidiol. Molecules, 28, 3271-3297.
Chaves, C., Bittencourt, P. C. T., & Pelegrini, A. (2020). Ingestion of a THC-rich cannabis oil in people with fibromyalgia: A randomized, double-blind, placebo-controlled clinical trial. Pain Medicine, 21, 2212-2218.
Choi, S., Na, H. S., Kim, J., Lee, J., Lee, S., Kim, D., Park, J., Chen, C. C., Campbell, K. P., & Shin, H. S. (2007). Attenuated pain responses in mice lacking Ca(V)3.2 T-type channels. Genes, Brain, and Behavior, 6, 425-431.
Clapper, J. R., Moreno-Sanz, G., Russo, R., Guijarro, A., Vacondio, F., Duranti, A., Tontini, A., Sanchini, S., Sciolino, N. R., Spradley, J. M., Hohmann, A. G., Calignano, A., Mor, M., Tarzia, G., & Piomelli, D. (2010). Anandamide suppresses pain initiation through a peripheral endocannabinoid mechanism. Nature Neuroscience, 13, 1265-1270.
Cohen, S. P., Vase, L., & Hooten, W. M. (2021). Chronic pain: An update on burden, best practices, and new advances. Lancet, 397, 2082-2097.
Colloca, L., Ludman, T., Bouhassira, D., Baron, R., Dickenson, A. H., Yarnitsky, D., Freeman, R., Truini, A., Attal, N., Finnerup, N. B., Eccleston, C., Kalso, E., Bennett, D. L., Dworkin, R. H., & Raja, S. N. (2017). Neuropathic pain. Nature Reviews Disease Primers, 3, 17002.
Corey-Bloom, J., Wolfson, T., Gamst, A., Jin, S., Marcotte, T. D., Bentley, H., & Gouaux, B. (2012). Smoked cannabis for spasticity in multiple sclerosis: A randomized, placebo-controlled trial. CMAJ, 184, 1143-1150.
Costa, B., Trovato, A. E., Comelli, F., Giagnoni, G., & Colleoni, M. (2007). The non-psychoactive cannabis constituent cannabidiol is an orally effective therapeutic agent in rat chronic inflammatory and neuropathic pain. European Journal of Pharmacology, 556, 75-83.
Dahlhamer, J., Lucas, J., Zelaya, C., Nahin, R., Mackey, S., DeBar, L., Kerns, R., Von Korff, M., Porter, L., & Helmick, C. (2018). Prevalence of chronic pain and high-impact chronic pain among adults-United States, 2016. MMWR. Morbidity and Mortality Weekly Report, 67, 1001-1006.
De Gregorio, D., McLaughlin, R. J., Posa, L., Ochoa - Sanchez, R., Enns, J., Lopez-Canul, M., Aboud, M., Maione, S., Comai, S., & Gobbi, G. (2019). Cannabidiol modulates serotonergic transmission and reverses both allodynia and anxiety-like behavior in a model of neuropathic pain. Pain, 160, 136-150.
de Melo Reis, R. A., Isaac, A. R., Freitas, H. R., de Almeida, M. M., Schuck, P. F., Ferreira, G. C., Andrade-da-Costa, B., & Trevenzoli, I. H. (2021). Quality of life and a surveillant endocannabinoid system. Frontiers in Neuroscience, 15, 747229.
De Petrocellis, L., Ligresti, A., Moriello, A. S., Allarà, M., Bisogno, T., Petrosino, S., Stott, C. G., & Di Marzo, V. (2011). Effects of cannabinoids and cannabinoid-enriched cannabis extracts on TRP channels and endocannabinoid metabolic enzymes. British Journal of Pharmacology, 163, 1479-1494.
De Petrocellis, L., Nabissi, M., Santoni, G., & Ligresti, A. (2017). Actions and regulation of ionotropic cannabinoid receptors. Advances in Pharmacology, 80, 249-289.
De Vry, J., Kuhl, E., Franken-Kunkel, P., & Eckel, G. (2004). Pharmacological characterization of the chronic constriction injury model of neuropathic pain. European Journal of Pharmacology, 491, 137-148.
Devinsky, O., Nabbout, R., Miller, I., Laux, L., Zolnowska, M., Wright, S., & Roberts, C. (2019). Long-term cannabidiol treatment in patients with Dravet syndrome: An open-label extension trial. Epilepsia, 60, 294-302.
Dogrul, A., Gardell, L. R., Ossipov, M. H., Tulunay, F. C., Lai, J., & Porreca, F. (2003). Reversal of experimental neuropathic pain by T-type calcium channel blockers. Pain, 105, 159-168.
Dos Santos, R., Veras, F., Netto, G., Elisei, L., Sorgi, C., Faccioli, L., & Galdino, G. (2023). Cannabidiol prevents chemotherapy-induced neuropathic pain by modulating spinal TLR4 via endocannabinoid system activation. The Journal of Pharmacy and Pharmacology, 75, 655-665.
Dumbraveanu, C., Strommer, K., Wonnemann, M., Choconta, J. L., Neumann, A., Kress, M., Kalpachidou, T., & Kummer, K. K. (2023). Pharmacokinetics of orally applied cannabinoids and medical marijuana extracts in mouse nervous tissue and plasma: Relevance for pain treatment. Pharmaceutics, 15, 853-867.
Dykukha, I., Malessa, R., Essner, U., & Uberall, M. A. (2021). Nabiximols in chronic neuropathic pain: A meta-analysis of randomized placebo-controlled trials. Pain Medicine, 22, 861-874.
Eisenberg, E., Ogintz, M., & Almog, S. (2014). The pharmacokinetics, efficacy, safety, and ease of use of a novel portable metered-dose cannabis inhaler in patients with chronic neuropathic pain: A phase 1a study. Journal of Pain & Palliative Care Pharmacotherapy, 28, 216-225.
Ellis, R. J., Toperoff, W., Vaida, F., van den Brande, G., Gonzales, J., Gouaux, B., Bentley, H., & Atkinson, J. H. (2009). Smoked medicinal cannabis for neuropathic pain in HIV: A randomized, crossover clinical trial. Neuropsychopharmacology, 34, 672-680.
Englund, A., Oliver, D., Chesney, E., Chester, L., Wilson, J., Sovi, S., de Micheli, A., Hodsoll, J., Fusar-Poli, P., Strang, J., Murray, R. M., Freeman, T. P., & McGuire, P. (2023). Does cannabidiol make cannabis safer? A randomised, double-blind, cross-over trial of cannabis with four different CBD:THC ratios. Neuropsychopharmacology, 48, 869-876.
Finnerup, N. B., Attal, N., Haroutounian, S., McNicol, E., Baron, R., Dworkin, R. H., Gilron, I., Haanpää, M., Hansson, P., Jensen, T. S., Kamerman, P. R., Lund, K., Moore, A., Raja, S. N., Rice, A. S. C., Rowbotham, M., Sena, E., Siddall, P., Smith, B. H., & Wallace, M. (2015). Pharmacotherapy for neuropathic pain in adults: A systematic review and meta-analysis. Lancet Neurology, 14, 162-173.
Fowler, C. J. (2015). The potential of inhibitors of endocannabinoid metabolism for drug development: A critical review. Handbook of Experimental Pharmacology, 231, 95-128.
Fox, A., Kesingland, A., Gentry, C., McNair, K., Patel, S., Urban, L., & James, I. (2001). The role of central and peripheral Cannabinoid1 receptors in the antihyperalgesic activity of cannabinoids in a model of neuropathic pain. Pain, 92, 91-100.
Gedin, F., Blome, S., Ponten, M., Lalouni, M., Fust, J., Raquette, A., Vadenmark Lundquist, V., Thompson, W. H., & Jensen, K. (2022). Placebo response and media attention in randomized clinical trials assessing cannabis-based therapies for pain: A systematic review and meta-analysis. JAMA Network Open, 5, e2243848.
Ghovanloo, M. R., Shuart, N. G., Mezeyova, J., Dean, R. A., Ruben, P. C., & Goodchild, S. J. (2018). Inhibitory effects of cannabidiol on voltage-dependent sodium currents. The Journal of Biological Chemistry, 293, 16546-16558.
Giossi, R., Carrara, F., Padroni, M., Bilancio, M. C., Mazzari, M., Enisci, S., Romio, M. S., Boni, G., Corrù, F., Fittipaldo, V. A., Tramacere, I., Pani, A., Scaglione, F., & Fornasari, D. (2022). Systematic review and meta-analysis seem to indicate that cannabinoids for chronic primary pain treatment have limited benefit. Pain and Therapy, 11, 1341-1358.
Giuffrida, A., Beltramo, M., & Piomelli, D. (2001). Mechanisms of endocannabinoid inactivation: Biochemistry and pharmacology. The Journal of Pharmacology and Experimental Therapeutics, 298, 7-14.
Guindon, J., & Hohmann, A. G. (2009). The endocannabinoid system and pain. CNS & Neurological Disorders Drug Targets, 8, 403-421.
Hama, A., & Sagen, J. (2011). Activation of spinal and supraspinal cannabinoid-1 receptors leads to antinociception in a rat model of neuropathic spinal cord injury pain. Brain Research, 1412, 44-54.
Hameed, S. (2019). Na(v)1.7 and Na(v)1.8: Role in the pathophysiology of pain. Molecular Pain, 15, 1744806919858801.
Harding, E. K., Souza, I. A., Gandini, M. A., Gadotti, V. M., Ali, M. Y., Huang, S., Antunes, F. T. T., Trang, T., & Zamponi, G. W. (2023). Differential regulation of Ca(v) 3.2 and Ca(v) 2.2 calcium channels by CB(1) receptors and cannabidiol. British Journal of Pharmacology, 180, 1616-1633.
Harris, H. M., Sufka, K. J., Gul, W., & ElSohly, M. A. (2016). Effects of Delta-9-tetrahydrocannabinol and Cannabidiol on cisplatin-induced neuropathy in mice. Planta Medica, 82, 1169-1172.
Hayakawa, K., Mishima, K., Hazekawa, M., Sano, K., Irie, K., Orito, K., Egawa, T., Kitamura, Y., Uchida, N., Nishimura, R., Egashira, N., Iwasaki, K., & Fujiwara, M. (2008). Cannabidiol potentiates pharmacological effects of Delta(9)-tetrahydrocannabinol via CB(1) receptor-dependent mechanism. Brain Research, 1188, 157-164.
Hind, W. H., England, T. J., & O'Sullivan, S. E. (2016). Cannabidiol protects an in vitro model of the blood-brain barrier from oxygen-glucose deprivation via PPARγ and 5-HT1A receptors. British Journal of Pharmacology, 173, 815-825.
Hlozek, T., Uttl, L., Kaderabek, L., et al. (2017). Pharmacokinetic and behavioural profile of THC, CBD, and THC+CBD combination after pulmonary, oral, and subcutaneous administration in rats and confirmation of conversion in vivo of CBD to THC. European Neuropsychopharmacology, 27, 1223-1237.
Howlett, A. C. (2002). The cannabinoid receptors. Prostaglandins & Other Lipid Mediators, 68-69, 619-631.
Howlett, A. C. (2005). Cannabinoid receptor signaling. Handbook of Experimental Pharmacology, 168, 53-79.
Huang, J., Fan, X., Jin, X., Jo, S., Zhang, H. B., Fujita, A., Bean, B. P., & Yan, N. (2023). Cannabidiol inhibits Na(v) channels through two distinct binding sites. Nature Communications, 14, 3613.
Ignatowska-Jankowska, B. M., Ghosh, S., Crowe, M. S., Kinsey, S. G., Niphakis, M. J., Abdullah, R. A., Tao, Q., O' Neal, S. T., Walentiny, D. M., Wiley, J. L., Cravatt, B. F., & Lichtman, A. H. (2014). In vivo characterization of the highly selective monoacylglycerol lipase inhibitor KML29: Antinociceptive activity without cannabimimetic side effects. British Journal of Pharmacology, 171, 1392-1407.
Jagodic, M. M., Pathirathna, S., Joksovic, P. M., Lee, W., Nelson, M. T., Naik, A. K., Su, P., Jevtovic-Todorovic, V., & Todorovic, S. M. (2008). Upregulation of the T-type calcium current in small rat sensory neurons after chronic constrictive injury of the sciatic nerve. Journal of Neurophysiology, 99, 3151-3156.
Jayamanne, A., Greenwood, R., Mitchell, V. A., Aslan, S., Piomelli, D., & Vaughan, C. W. (2006). Actions of the FAAH inhibitor URB597 in neuropathic and inflammatory chronic pain models. British Journal of Pharmacology, 147, 281-288.
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., & da Cunha, J. M. (2019). Cannabidiol attenuates mechanical allodynia in streptozotocin-induced diabetic rats via serotonergic system activation through 5-HT1A receptors. Brain Research, 1715, 156-164.
Jhaveri, M. D., Richardson, D., Kendall, D. A., Barrett, D. A., & Chapman, V. (2006). Analgesic effects of fatty acid amide hydrolase inhibition in a rat model of neuropathic pain. The Journal of Neuroscience, 26, 13318-13327.
Jhaveri, M. D., Richardson, D., Robinson, I., Garle, M. J., Patel, A., Sun, Y., Sagar, D. R., Bennett, A. J., Alexander, S. P. H., Kendall, D. A., Barrett, D. A., & Chapman, V. (2008). Inhibition of fatty acid amide hydrolase and cyclooxygenase-2 increases levels of endocannabinoid related molecules and produces analgesia via peroxisome proliferator-activated receptor-alpha in a model of inflammatory pain. Neuropharmacology, 55, 85-93.
Johal, H., Devji, T., Chang, Y., Simone, J., Vannabouathong, C., & Bhandari, M. (2020). Cannabinoids in chronic non-cancer pain: A systematic review and meta-analysis. Clinical Medicine Insights. Arthritis and Musculoskeletal Disorders, 13, 1179544120906461.
Jordan, C. J., & Xi, Z. X. (2019). Progress in brain cannabinoid CB(2) receptor research: From genes to behavior. Neuroscience and Biobehavioral Reviews, 98, 208-220.
Kano, M., Ohno-Shosaku, T., Hashimotodani, Y., Uchigashima, M., & Watanabe, M. (2009). Endocannabinoid-mediated control of synaptic transmission. Physiological Reviews, 89, 309-380.
Kazantzis, N. P., Casey, S. L., Seow, P. W., Mitchell, V. A., & Vaughan, C. W. (2016). Opioid and cannabinoid synergy in a mouse neuropathic pain model. British Journal of Pharmacology, 173, 2521-2531.
King, K. M., Myers, A. M., Soroka-Monzo, A. J., Tuma, R. F., Tallarida, R. J., Walker, E. A., & Ward, S. J. (2017). Single and combined effects of Delta(9) -tetrahydrocannabinol and cannabidiol in a mouse model of chemotherapy-induced neuropathic pain. British Journal of Pharmacology, 174, 2832-2841.
Kinsey, S. G., Long, J. Z., Cravatt, B. F., & Lichtman, A. H. (2010). Fatty acid amide hydrolase and monoacylglycerol lipase inhibitors produce anti-allodynic effects in mice through distinct cannabinoid receptor mechanisms. The Journal of Pain, 11, 1420-1428.
Kinsey, S. G., Long, J. Z., O'Neal, S. T., Abdullah, R. A., Poklis, J. L., Boger, D. L., Cravatt, B. F., & Lichtman, A. H. (2009). Blockade of endocannabinoid-degrading enzymes attenuates neuropathic pain. The Journal of Pharmacology and Experimental Therapeutics, 330, 902-910.
Kinsey, S. G., Wise, L. E., Ramesh, D., Abdullah, R., Selley, D. E., Cravatt, B. F., & Lichtman, A. H. (2013). Repeated low-dose administration of the monoacylglycerol lipase inhibitor JZL184 retains cannabinoid receptor type 1-mediated antinociceptive and gastroprotective effects. The Journal of Pharmacology and Experimental Therapeutics, 345, 492-501.
Kumar Kalvala, A., Bagde, A., Arthur, P., Kumar Surapaneni, S., Ramesh, N., Nathani, A., & Singh, M. (2022). Role of Cannabidiol and Tetrahydrocannabivarin on paclitaxel-induced neuropathic pain in rodents. International Immunopharmacology, 107, 108693.
Li, H., Kong, W., Chambers, C. R., Yu, D., Ganea, D., Tuma, R. F., & Ward, S. J. (2018). The non-psychoactive phytocannabinoid cannabidiol (CBD) attenuates pro-inflammatory mediators, T cell infiltration, and thermal sensitivity following spinal cord injury in mice. Cellular Immunology, 329, 1-9.
Lim, G., Sung, B., Ji, R. R., & Mao, J. (2003). Upregulation of spinal cannabinoid-1-receptors following nerve injury enhances the effects of win 55,212-2 on neuropathic pain behaviors in rats. Pain, 105, 275-283.
Linher-Melville, K., Zhu, Y. F., Sidhu, J., Parzei, N., Shahid, A., Seesankar, G., Ma, D., Wang, Z., Zacal, N., Sharma, M., Parihar, V., Zacharias, R., & Singh, G. (2020). Evaluation of the preclinical analgesic efficacy of naturally derived, orally administered oil forms of Delta9-tetrahydrocannabinol (THC), cannabidiol (CBD), and their 1:1 combination. PLoS One, 15, e0234176.
Long, J. Z., Li, W., Booker, L., Burston, J. J., Kinsey, S. G., Schlosburg, J. E., Pavón, F. J., Serrano, A. M., Selley, D. E., Parsons, L. H., Lichtman, A. H., & Cravatt, B. F. (2009). Selective blockade of 2-arachidonoylglycerol hydrolysis produces cannabinoid behavioral effects. Nature Chemical Biology, 5, 37-44.
Long, J. Z., Nomura, D. K., Vann, R. E., Walentiny, D. M., Booker, L., Jin, X., Burston, J. J., Sim-Selley, L. J., Lichtman, A. H., Wiley, J. L., & Cravatt, B. F. (2009). Dual blockade of FAAH and MAGL identifies behavioral processes regulated by endocannabinoid crosstalk in vivo. Proceedings of the National Academy of Sciences of the United States of America, 106, 20270-20275.
Lu, H. C., & Mackie, K. (2016). An introduction to the endogenous cannabinoid system. Biological Psychiatry, 79, 516-525.
Lu, H. C., & Mackie, K. (2021). Review of the endocannabinoid system. Biological Psychiatry: Cognitive Neuroscience and Neuroimaging, 6, 607-615.
Lucas, C. J., Galettis, P., & Schneider, J. (2018). The pharmacokinetics and the pharmacodynamics of cannabinoids. British Journal of Clinical Pharmacology, 84, 2477-2482.
Mackie, K., & Hille, B. (1992). Cannabinoids inhibit N-type calcium channels in neuroblastoma-glioma cells. Proceedings of the National Academy of Sciences of the United States of America, 89, 3825-3829.
Maione, S., Costa, B., & Di Marzo, V. (2013). Endocannabinoids: A unique opportunity to develop multitarget analgesics. Pain, 154(Suppl 1), S87-S93.
Malvestio, R., Medeiros, P., Negrini-Ferrari, S., et al. (2021). Cannabidiol in the prelimbic cortex modulates the comorbid condition between the chronic neuropathic pain and depression-like behaviour in rats: The role of medial prefrontal cortex 5-HT1A and CB1 receptors. Brain Research Bulletin, 174, 323-338.
Martin, W. J., Coffin, P. O., Attias, E., Balinsky, M., Tsou, K., & Walker, J. M. (1999). Anatomical basis for cannabinoid-induced antinociception as revealed by intracerebral microinjections. Brain Research, 822, 237-242.
Matarazzo, A. P., Elisei, L. M. S., Carvalho, F. C., Bonfilio, R., Ruela, A. L. M., Galdino, G., & Pereira, G. R. (2021). Mucoadhesive nanostructured lipid carriers as a cannabidiol nasal delivery system for the treatment of neuropathic pain. European Journal of Pharmaceutical Sciences, 159, 105698.
McPartland, J. M., Duncan, M., Di Marzo, V., & Pertwee, R. G. (2015). Are cannabidiol and Delta(9) -tetrahydrocannabivarin negative modulators of the endocannabinoid system? A systematic review. British Journal of Pharmacology, 172, 737-753.
Meng, H., Johnston, B., Englesakis, M., Moulin, D. E., & Bhatia, A. (2017). Selective cannabinoids for chronic neuropathic pain: A systematic review and meta-analysis. Anesthesia and Analgesia, 125, 1638-1652.
Mirlohi, S., Bladen, C., Santiago, M. J., Arnold, J. C., McGregor, I., & Connor, M. (2022). Inhibition of human recombinant T-type calcium channels by phytocannabinoids in vitro. British Journal of Pharmacology, 179, 4031-4043.
Mitchell, V. A., Harley, J., Casey, S. L., Vaughan, A. C., Winters, B. L., & Vaughan, C. W. (2021). Oral efficacy of Delta(9)-tetrahydrocannabinol and cannabidiol in a mouse neuropathic pain model. Neuropharmacology, 189, 108529.
Morgan, C. J., Schafer, G., Freeman, T. P., & Curran, H. V. (2010). Impact of cannabidiol on the acute memory and psychotomimetic effects of smoked cannabis: Naturalistic study: Naturalistic study [corrected]. The British Journal of Psychiatry, 197, 285-290.
Mucke, M., Phillips, T., Radbruch, L., Petzke, F., & Hauser, W. (2018). Cannabis-based medicines for chronic neuropathic pain in adults. Cochrane Database of Systematic Reviews, 3, CD012182.
Muller, C., Morales, P., & Reggio, P. H. (2018). Cannabinoid ligands targeting TRP channels. Frontiers in Molecular Neuroscience, 11, 487.
Na, H. S., Choi, S., Kim, J., Park, J., & Shin, H. S. (2008). Attenuated neuropathic pain in Cav3.1 null mice. Molecules and Cells, 25, 242-246.
Notcutt, W., Price, M., Miller, R., Newport, S., Phillips, C., Simmons, S., & Sansom, C. (2004). Initial experiences with medicinal extracts of cannabis for chronic pain: Results from 34 ‘N of 1’ studies. Anaesthesia, 59, 440-452.
Nurmikko, T. J., Serpell, M. G., Hoggart, B., Toomey, P. J., Morlion, B. J., & Haines, D. (2007). Sativex successfully treats neuropathic pain characterised by allodynia: A randomised, double-blind, placebo-controlled clinical trials. Pain, 133, 210-220.
Nutt, D. J., Phillips, L. D., Barnes, M. P., Brander, B., Curran, H. V., Fayaz, A., Finn, D. P., Horsted, T., Moltke, J., Sakal, C., Sharon, H., O'Sullivan, S. E., Williams, T., Zorn, G., & Schlag, A. K. (2022). A multicriteria decision analysis comparing pharmacotherapy for chronic neuropathic pain, including cannabinoids and cannabis-based medical products. Cannabis and Cannabinoid Research, 7, 482-500.
Okine, B. N., Gaspar, J. C., & Finn, D. P. (2019). PPARs and pain. British Journal de Pharmacologie, 176, 1421-1442.
O'Sullivan, S. E. (2007). Cannabinoids go nuclear: Evidence for activation of peroxisome proliferator-activated receptors. British Journal of Pharmacology, 152, 576-582.
O'Sullivan, S. E. (2016). An update on PPAR activation by cannabinoids. British Journal of Pharmacology, 173, 1899-1910.
Pertwee, R. G. (2005). Pharmacological actions of cannabinoids. Handbook of Experimental Pharmacology, 168, 1-51.
Pertwee, R. G. (2008). Ligands that target cannabinoid receptors in the brain: From THC to anandamide and beyond. Addiction Biology, 13, 147-159.
Pertwee, R. G., Rock, E. M., Guenther, K., Limebeer, C. L., Stevenson, L. A., Haj, C., Smoum, R., Parker, L. A., & Mechoulam, R. (2018). Cannabidiolic acid methyl ester, a stable synthetic analogue of cannabidiolic acid, can produce 5-HT(1A) receptor-mediated suppression of nausea and anxiety in rats. British Journal of Pharmacology, 175, 100-112.
Rahn, E. J., & Hohmann, A. G. (2009). Cannabinoids as pharmacotherapies for neuropathic pain: From the bench to the bedside. Neurotherapeutics, 6, 713-737.
Rahn, E. J., Makriyannis, A., & Hohmann, A. G. (2007). Activation of cannabinoid CB1 and CB2 receptors suppresses neuropathic nociception evoked by the chemotherapeutic agent vincristine in rats. British Journal of Pharmacology, 152, 765-777.
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., Song, X. J., Stevens, B., Sullivan, M. D., Tutelman, P. R., Ushida, T., & Vader, K. (2020). The revised International Association for the Study of Pain definition of pain: Concepts, challenges, and compromises. Pain, 161, 1976-1982.
Riedel, G., & Davies, S. N. (2005). Cannabinoid function in learning, memory and plasticity. Handbook of Experimental Pharmacology, 168, 445-477.
Robson, P. J. (2014). Therapeutic potential of cannabinoid medicines. Drug Testing and Analysis, 6, 24-30.
Rog, D. J., Nurmikko, T. J., Friede, T., & Young, C. A. (2005). Randomized, controlled trial of cannabis-based medicine in central pain in multiple sclerosis. Neurology, 65, 812-819.
Ross, H. R., Napier, I., & Connor, M. (2008). Inhibition of recombinant human T-type calcium channels by Delta9-tetrahydrocannabinol and cannabidiol. The Journal of Biological Chemistry, 283, 16124-16134.
Russo, E. B. (2011). Taming THC: Potential cannabis synergy and phytocannabinoid-terpenoid entourage effects. British Journal of Pharmacology, 163, 1344-1364.
Russo, E. B., Burnett, A., Hall, B., & Parker, K. K. (2005). Agonistic properties of cannabidiol at 5-HT1a receptors. Neurochemical Research, 30, 1037-1043.
Russo, R., Loverme, J., La Rana, G., et al. (2007). The fatty acid amide hydrolase inhibitor URB597 (cyclohexylcarbamic acid 3′-carbamoylbiphenyl-3-yl ester) reduces neuropathic pain after oral administration in mice. The Journal of Pharmacology and Experimental Therapeutics, 322, 236-242.
Ryberg, E., Larsson, N., Sjögren, S., Hjorth, S., Hermansson, N. O., Leonova, J., Elebring, T., Nilsson, K., Drmota, T., & Greasley, P. J. (2007). The orphan receptor GPR55 is a novel cannabinoid receptor. British Journal of Pharmacology, 152, 1092-1101.
Sainsbury, B., Bloxham, J., Pour, M. H., Padilla, M., & Enciso, R. (2021). Efficacy of cannabis-based medications compared to placebo for the treatment of chronic neuropathic pain: A systematic review with meta-analysis. Journal of Dental Anesthesia and Pain Medicine, 21, 479-506.
Schimrigk, S., Marziniak, M., Neubauer, C., Kugler, E. M., Werner, G., & Abramov-Sommariva, D. (2017). Dronabinol is a safe Long-term treatment option for neuropathic pain patients. European Neurology, 78, 320-329.
Scholz, J., Finnerup, N. B., Attal, N., Aziz, Q., Baron, R., Bennett, M. I., Benoliel, R., Cohen, M., Cruccu, G., Davis, K. D., Evers, S., First, M., Giamberardino, M. A., Hansson, P., Kaasa, S., Korwisi, B., Kosek, E., Lavand'homme, P., Nicholas, M., … Classification Committee of the Neuropathic Pain Special Interest Group (NeuPSIG). (2019). The IASP classification of chronic pain for ICD-11: Chronic neuropathic pain. Pain, 160, 53-59.
Scott, D. A., Wright, C. E., & Angus, J. A. (2004). Evidence that CB-1 and CB-2 cannabinoid receptors mediate antinociception in neuropathic pain in the rat. Pain, 109, 124-131.
Selvarajah, D., Gandhi, R., Emery, C. J., & Tesfaye, S. (2010). Randomized placebo-controlled double-blind clinical trial of cannabis-based medicinal product (Sativex) in painful diabetic neuropathy: Depression is a major confounding factor. Diabetes Care, 33, 128-130.
Serpell, M., Ratcliffe, S., Hovorka, J., Schofield, M., Taylor, L., Lauder, H., & Ehler, E. (2014). A double-blind, randomized, placebo-controlled, parallel group study of THC/CBD spray in peripheral neuropathic pain treatment. European Journal of Pain, 18, 999-1012.
Silva, N. R., Gomes, F. I. F., Lopes, A. H. P., Cortez, I. L., dos Santos, J. C., Silva, C. E. A., Mechoulam, R., Gomes, F. V., Cunha, T. M., & Guimarães, F. S. (2022). The cannabidiol analog PECS-101 prevents chemotherapy-induced neuropathic pain via PPARgamma receptors. Neurotherapeutics, 19, 434-449.
Silva-Cardoso, G. K., Lazarini-Lopes, W., Hallak, J. E., Crippa, J. A., Zuardi, A. W., Garcia-Cairasco, N., & Leite-Panissi, C. R. A. (2021). Cannabidiol effectively reverses mechanical and thermal allodynia, hyperalgesia, and anxious behaviors in a neuropathic pain model: Possible role of CB1 and TRPV1 receptors. Neuropharmacology, 197, 108712.
Skrabek, R. Q., Galimova, L., Ethans, K., & Perry, D. (2008). Nabilone for the treatment of pain in fibromyalgia. The Journal of Pain, 9, 164-173.
Smith, P. B., & Martin, B. R. (1992). Spinal mechanisms of delta 9-tetrahydrocannabinol-induced analgesia. Brain Research, 578, 8-12.
Starowicz, K., & Finn, D. P. (2017). Cannabinoids and pain: Sites and mechanisms of action. Advances in Pharmacology, 80, 437-475.
Staton, P. C., Hatcher, J. P., Walker, D. J., Morrison, A. D., Shapland, E. M., Hughes, J. P., Chong, E., Mander, P. K., Green, P. J., Billinton, A., Fulleylove, M., Lancaster, H. C., Smith, J. C., Bailey, L. T., Wise, A., Brown, A. J., Richardson, J. C., & Chessell, I. P. (2008). The putative cannabinoid receptor GPR55 plays a role in mechanical hyperalgesia associated with inflammatory and neuropathic pain. Pain, 139, 225-236.
Stockings, E., Campbell, G., Hall, W. D., Nielsen, S., Zagic, D., Rahman, R., Murnion, B., Farrell, M., Weier, M., & Degenhardt, L. (2018). 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, 159, 1932-1954.
Taffe, M. A., Creehan, K. M., & Vandewater, S. A. (2015). Cannabidiol fails to reverse hypothermia or locomotor suppression induced by Delta(9) -tetrahydrocannabinol in Sprague-Dawley rats. British Journal of Pharmacology, 172, 1783-1791.
Torrance, N., Smith, B. H., Bennett, M. I., & Lee, A. J. (2006). The epidemiology of chronic pain of predominantly neuropathic origin. Results from a general population survey. The Journal of Pain, 7, 281-289.
Toth, C., Mawani, S., Brady, S., Chan, C., Liu, C., Mehina, E., Garven, A., Bestard, J., & Korngut, L. (2012). An enriched-enrolment, randomized withdrawal, flexible-dose, double-blind, placebo-controlled, parallel assignment efficacy study of nabilone as adjuvant in the treatment of diabetic peripheral neuropathic pain. Pain, 153, 2073-2082.
Toth, C. C., Jedrzejewski, N. M., Ellis, C. L., & Frey, W. H., 2nd. (2010). Cannabinoid-mediated modulation of neuropathic pain and microglial accumulation in a model of murine type I diabetic peripheral neuropathic pain. Molecular Pain, 6, 16.
Tyree, G. A., Sarkar, R., Bellows, B. K., Ellis, R. J., Atkinson, J. H., Marcotte, T. D., Wallace, M. S., Grant, I., Shi, Y., Murphy, J. D., & Grelotti, D. J. (2019). A cost-effectiveness model for adjunctive smoked cannabis in the treatment of chronic neuropathic pain. Cannabis and Cannabinoid Research, 4, 62-72.
Ueberall, M. A., Essner, U., Vila Silvan, C., & Mueller-Schwefe, G. H. H. (2022). Comparison of the effectiveness and tolerability of Nabiximols (THC:CBD) Oromucosal spray versus Oral Dronabinol (THC) as add-on treatment for severe neuropathic pain in real-world clinical practice: Retrospective analysis of the German pain e-registry. Journal of Pain Research, 15, 267-286.
Varvel, S. A., Wiley, J. L., Yang, R., Bridgen, D. T., Long, K., Lichtman, A. H., & Martin, B. R. (2006). Interactions between THC and cannabidiol in mouse models of cannabinoid activity. Psychopharmacology, 186, 226-234.
Vuong, L. A., Mitchell, V. A., & Vaughan, C. W. (2008). Actions of N-arachidonyl-glycine in a rat neuropathic pain model. Neuropharmacology, 54, 189-193.
Wade, D. T., Robson, P., House, H., Makela, P., & Aram, J. (2003). A preliminary controlled study to determine whether whole-plant cannabis extracts can improve intractable neurogenic symptoms. Clinical Rehabilitation, 17, 21-29.
Walitt, B., Klose, P., Fitzcharles, M. A., Phillips, T., & Hauser, W. (2016). Cannabinoids for fibromyalgia. Cochrane Database of Systematic Reviews, 7, CD011694.
Wang, L., Hong, P. J., May, C., Rehman, Y., Oparin, Y., Hong, C. J., Hong, B. Y., AminiLari, M., Gallo, L., Kaushal, A., Craigie, S., Couban, R. J., Kum, E., Shanthanna, H., Price, I., Upadhye, S., Ware, M. A., Campbell, F., Buchbinder, R., … Busse, J. W. (2021). Medical cannabis or cannabinoids for chronic non-cancer and cancer related pain: A systematic review and meta-analysis of randomised clinical trials. BMJ, 374, n1034.
Wang, X. F., Galaj, E., Bi, G. H., Zhang, C., He, Y., Zhan, J., Bauman, M. H., Gardner, E. L., & Xi, Z. X. (2020). Different receptor mechanisms underlying phytocannabinoid- versus synthetic cannabinoid-induced tetrad effects: Opposite roles of CB(1) /CB(2) versus GPR55 receptors. British Journal of Pharmacology, 177, 1865-1880.
Ward, S. J., McAllister, S. D., Kawamura, R., Murase, R., Neelakantan, H., & Walker, E. A. (2014). Cannabidiol inhibits paclitaxel-induced neuropathic pain through 5-HT(1A) receptors without diminishing nervous system function or chemotherapy efficacy. British Journal of Pharmacology, 171, 636-645.
Ward, S. J., Ramirez, M. D., Neelakantan, H., & Walker, E. A. (2011). Cannabidiol prevents the development of cold and mechanical allodynia in paclitaxel-treated female C57Bl6 mice. Anesthesia and Analgesia, 113, 947-950.
Ware, M. A., Adams, H., & Guy, G. W. (2005). The medicinal use of cannabis in the UK: Results of a nationwide survey. International Journal of Clinical Practice, 59, 291-295.
Ware, M. A., Wang, T., Shapiro, S., Robinson, A., Ducruet, T., Huynh, T., Gamsa, A., Bennett, G. J., & Collet, J. P. (2010). Smoked cannabis for chronic neuropathic pain: A randomized controlled trial. CMAJ, 182, E694-E701.
Watkins, A. R. (2019). Cannabinoid interactions with ion channels and receptors. Channels (Austin, Tex.), 13, 162-167.
Weizman, L., Dayan, L., Brill, S., Nahman-Averbuch, H., Hendler, T., Jacob, G., & Sharon, H. (2018). Cannabis analgesia in chronic neuropathic pain is associated with altered brain connectivity. Neurology, 91, e1285-e1294.
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., Schmidlkofer, S., Westwood, M., & Kleijnen, J. (2015). Cannabinoids for medical use: A systematic review and meta-analysis. JAMA, 313, 2456-2473.
Williams, A. C. C., Fisher, E., Hearn, L., & Eccleston, C. (2020). Psychological therapies for the management of chronic pain (excluding headache) in adults. Cochrane Database of Systematic Reviews, 8, CD007407.
Williams, J., Haller, V. L., Stevens, D. L., & Welch, S. P. (2008). Decreased basal endogenous opioid levels in diabetic rodents: Effects on morphine and delta-9-tetrahydrocannabinoid-induced antinociception. European Journal of Pharmacology, 584, 78-86.
Wilsey, B., Marcotte, T., Deutsch, R., Gouaux, B., Sakai, S., & Donaghe, H. (2013). Low-dose vaporized cannabis significantly improves neuropathic pain. The Journal of Pain, 14, 136-148.
Wilsey, B., Marcotte, T., Tsodikov, A., Millman, J., Bentley, H., Gouaux, B., & Fishman, S. (2008). A randomized, placebo-controlled, crossover trial of cannabis cigarettes in neuropathic pain. The Journal of Pain, 9, 506-521.
Wilsey, B., Marcotte, T. D., Deutsch, R., Zhao, H., Prasad, H., & Phan, A. (2016). An exploratory human laboratory experiment evaluating vaporized cannabis in the treatment of neuropathic pain from spinal cord injury and disease. The Journal of Pain, 17, 982-1000.
Winters, B. L., & Vaughan, C. W. (2021). Mechanisms of endocannabinoid control of synaptic plasticity. Neuropharmacology, 197, 108736.
Wolf, J., Urits, I., Orhurhu, V., Peck, J., Orhurhu, M. S., Giacomazzi, S., Smoots, D., Piermarini, C., Manchikanti, L., Kaye, A. D., Kaye, R. J., & Viswanath, O. (2020). The role of the cannabinoid system in pain control: Basic and clinical implications. Current Pain and Headache Reports, 24, 35.
Wong, G. Y., & Gavva, N. R. (2009). Therapeutic potential of vanilloid receptor TRPV1 agonists and antagonists as analgesics: Recent advances and setbacks. Brain Research Reviews, 60, 267-277.
Wong, S. S. C., Chan, W. S., & Cheung, C. W. (2020). Analgesic effects of cannabinoids for chronic non-cancer pain: A systematic review and meta-analysis with meta-regression. Journal of Neuroimmune Pharmacology, 15, 801-829.
Woolf, C. J., & American College of Physicians and American Physiological Society. (2004). Pain: Moving from symptom control toward mechanism-specific pharmacologic management. Annals of Internal Medicine, 140, 441-451.
Xiong, W., Cui, T., Cheng, K., Yang, F., Chen, S. R., Willenbring, D., Guan, Y., Pan, H. L., Ren, K., Xu, Y., & Zhang, L. (2012). Cannabinoids suppress inflammatory and neuropathic pain by targeting alpha3 glycine receptors. The Journal of Experimental Medicine, 209, 1121-1134.
Xu, D. H., Cullen, B. D., Tang, M., & Fang, Y. (2020). The effectiveness of topical Cannabidiol oil in symptomatic relief of peripheral neuropathy of the lower extremities. Current Pharmaceutical Biotechnology, 21, 390-402.
Xue, Y., Chidiac, C., Herault, Y., & Gaveriaux-Ruff, C. (2021). Pain behavior in SCN9A (Nav1.7) and SCN10A (Nav1.8) mutant rodent models. Neuroscience Letters, 753, 135844.
Yevenes, G. E., & Zeilhofer, H. U. (2011). Allosteric modulation of glycine receptors. British Journal of Pharmacology, 164, 224-236.
Zadikoff, C., Wadia, P. M., Miyasaki, J., Chen, R., Lang, A. E., So, J., & Fox, S. H. (2011). Cannabinoid, CB1 agonists in cervical dystonia: Failure in a phase IIa randomized controlled trial. Basal Ganglia, 1, 91-95.
Zajicek, J. P., Hobart, J. C., Slade, A., Barnes, D., Mattison, P. G., & Group, M. R. (2012). Multiple sclerosis and extract of cannabis: Results of the MUSEC trial. Journal of Neurology, Neurosurgery, and Psychiatry, 83, 1125-1132.
Zhang, H. B., & Bean, B. P. (2021). Cannabidiol inhibition of murine primary nociceptors: Tight binding to slow inactivated states of Na(v)1.8 channels. The Journal of Neuroscience, 41, 6371-6387.
Zhu, C. Z., Mikusa, J. P., Fan, Y., Hollingsworth, P. R., Pai, M., Chandran, P., Daza, A. V., Yao, B. B., Dart, M. J., Meyer, M. D., Decker, M. W., Hsieh, G. C., & Honore, P. (2009). Peripheral and central sites of action for the non-selective cannabinoid agonist WIN 55,212-2 in a rat model of post-operative pain. British Journal of Pharmacology, 157, 645-655.
Zubcevic, K., Petersen, M., Bach, F. W., Heinesen, A., Enggaard, T. P., Almdal, T. P., Holbech, J. V., Vase, L., Jensen, T. S., Hansen, C. S., Finnerup, N. B., & Sindrup, S. H. (2023). Oral capsules of tetra-hydro-cannabinol (THC), cannabidiol (CBD) and their combination in peripheral neuropathic pain treatment. European Journal of Pain, 27, 492-506.
فهرسة مساهمة: Keywords: cannabidiol; cannabis; delta-9-tetrahydrocannibinol; ion channel; neuropathic pain; receptor
تواريخ الأحداث: Date Created: 20230925 Latest Revision: 20230925
رمز التحديث: 20231215
DOI: 10.1111/jnc.15964
PMID: 37747128
قاعدة البيانات: MEDLINE
الوصف
تدمد:1471-4159
DOI:10.1111/jnc.15964