TIBBİ KENEVİR VE SAĞLIK: FARMAKOLOJİK BİR DERLEME

Amaç: Kenevir antik çağlardan bugüne dek tedavi amaçlı kullanılmakta olan bir bitkidir. Her ne kadar kullanımı içerdiği psikoaktif bileşenler dolayısıyla sınırlandırılmış olsa da, kenevirin tıbbi yönüne dair araştırmalar literatürde oldukça yoğundur. Cannabis sativa L., içerdiği fitokannabinoidler ile endokannabinoid sistemde gen düzeyinde pek çok değişikliğe sebep olabilmektedir. Endokannabinoid sistemin pek çok patolojik durumda potansiyel bir terapötik hedef olabileceğine dair kanıtların derlenmesi hedefiyle bu çalışma ortaya koyulmuştur.

MEDICAL CANNABIS AND HEALTH: A PHARMACOLOGICAL REVIEW

Objective: Cannabis sativa L. is a plant that has been used for therapeutic purposes since ancient times. However, its use has been limited due to its content. Cannabis causes many changes at the gene level in the endocannabinoid system with the phytocannabinoids it contains. The aim of this study was to provide evidence that the endocannabinoid system could be a potential therapeutic target in many pathological conditions.

___

  • 1. Maule, W. J. (2015). Medical uses of marijuana (Cannabis sativa): fact or fallacy? British Journal of Biomedical Science, 72(2), 85-91.[CrossRef]
  • 2. Zuardi, A. W. (2006). History of cannabis as a medicine: a review. Brazilian Journal of Psychiatry, 28, 153-157. [CrossRef]
  • 3. Kalant, H. (2001). Medicinal use of cannabis: history and current status. Pain Res Manag, 6(2), 80-91. [CrossRef]
  • 4. Touw, M. (1981). The Religious and Medicinal Uses of Cannabis in China, India and Tibet. Journal of Psychoactive Drugs, 13(1), 23-34. [CrossRef]
  • 5. Pisanti, S., Bifulco, M. (2017). Modern History of Medical Cannabis: From Widespread Use to Prohibitionism and Back. Trends in Pharmacological Sciences, 38(3), 195-198. [CrossRef]
  • 6. Amin, M. R., Ali, D. W. (2019). Pharmacology of Medical Cannabis. Adv Exp Med Biol, 1162, 151-165. [CrossRef]
  • 7. Turan, M., (2000). Lif Bitkileri. Bursa: Uludağ Üniversitesi Ziraat Fakültesi Tarla Bitkilleri Bölümü,.
  • 8. Aydoğan, M., Terzı̇, Y. E., Gizlenci, Ş., Acar, M., Esen, A., Meral, H. (2020). Türkiye’de kenevir yetiştiriciliğinin ekonomik olarak yapılabilirliği: Samsun ili Vezirköprü ilçesi örneği. ANADOLU JOURNAL OF AGRICULTURAL SCIENCES, 35, 35-50. [CrossRef]
  • 9. Ağar, Osman T. (2019). "Cannabis’e Tarihsel Bir Bakış." Estüdam Halk Sağliği Dergisi , pp.7- 11.
  • 10. Koca Çalışkan, U, Yıldırım, S . (2020) "Kenevir Ve Sağlık Alanında Kullanımı" . Journal of Faculty of Pharmacy of Ankara University, 44: 112-136. [CrossRef]
  • 11. Onay, A., Yıldırım, H., Ekinci, R. (2020). Kenevir, Cannabis sativa L. Ankara: Palme Yayınevi.
  • 12. ElSohly, M. A., Radwan, M. M., Gul, W., Chandra, S., Galal, A. (2017). Phytochemistry of Cannabis sativa L. Prog Chem Org Nat Prod, 103, 1-36. [CrossRef]
  • 13. Pertwee, R. G. (2008). The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: delta9-tetrahydrocannabinol, cannabidiol and delta9-tetrahydrocannabivarin. British journal of pharmacology, 153(2), 199-215. [CrossRef]
  • 14. Crippa, J. A., Zuardi, A. W., Martín-Santos, R., Bhattacharyya, S., Atakan, Z., McGuire, P., Fusar-Poli, P. (2009). Cannabis and anxiety: a critical review of the evidence. Hum Psychopharmacol, 24(7), 515-523. [CrossRef]
  • 15. Zuardi, A. W., Morais, S. L., Guimarães, F. S., Mechoulam, R. (1995). Antipsychotic effect of cannabidiol. J Clin Psychiatry, 56(10), 485-486. [CrossRef]
  • 16. Mechoulam, R., Peters, M., Murillo-Rodriguez, E., Hanus, L. O. (2007). Cannabidiol--recent advances. Chem Biodivers, 4(8), 1678-1692. [CrossRef]
  • 17. Console-Bram, L., Marcu, J., Abood, M. E. (2012). Cannabinoid receptors: nomenclature and pharmacological principles. Progress in Neuro-Psychopharmacology and Biological Psychiatry, 38(1), 4-15. [CrossRef]
  • 18. Svíženská, I., Dubový, P., Šulcová, A. (2008). Cannabinoid receptors 1 and 2 (CB1 and CB2), their distribution, ligands and functional involvement in nervous system structures — A short review. Pharmacology Biochemistry and Behavior, 90(4), 501-511. [CrossRef]
  • 19. Kendall, D. A., Yudowski, G. A. (2017). Cannabinoid Receptors in the Central Nervous System: Their Signaling and Roles in Disease. Frontiers in Cellular Neuroscience, 10(294). [CrossRef]
  • 20. Szulakowska, A., Milnerowicz, H. (2007). Cannabis sativa in the Light of Scientific Research. Advances in Clinical and Experimental Medicine, 16, 807-815.
  • 21. Onaivi, E. S., Ishiguro, H., Gong, J.-P., Patel, S., Perchuk, A., Meozzi, P. A., Uhl, G. R. (2006). Discovery of the Presence and Functional Expression of Cannabinoid CB2 Receptors in Brain. Annals of the New York Academy of Sciences, 1074(1), 514-536. [CrossRef]
  • 22. Van Sickle, M. D., Duncan, M., Kingsley, P. J., Mouihate, A., Urbani, P., Mackie, K., Sharkey, K. A. (2005). Identification and Functional Characterization of Brainstem Cannabinoid CB2 Receptors. Science, 310(5746), 329-332. [CrossRef]
  • 23. Henstridge, C. M. (2012). Off-Target Cannabinoid Effects Mediated by GPR55. Pharmacology, 89(3-4), 179-187. [CrossRef]
  • 24. Katchan, V., David, P.,Shoenfeld, Y. (2016). Cannabinoids and autoimmune diseases: A systematic review. Autoimmunity Reviews, 15(6), 513-528. [CrossRef]
  • 25. Laprairie, R. B., Bagher, A. M., Denovan-Wright, E. M. (2017). Cannabinoid receptor ligand bias: implications in the central nervous system. Current Opinion in Pharmacology, 32, 32-43. [CrossRef]
  • 26. Cunha, P., Romão, A., Mascarenhas-Melo, F., Teixeira, H., Reis, F. (2011). Endocannabinoid system in cardiovascular disorders - new pharmacotherapeutic opportunities. Journal of Pharmacy And Bioallied Sciences, 3(3), 350-360. [CrossRef]
  • 27. Pagotto, U., Marsicano, G., Cota, D., Lutz, B., Pasquali, R. (2006). The Emerging Role of the Endocannabinoid System in Endocrine Regulation and Energy Balance. Endocrine Reviews, 27(1), 73-100. doi:10.1210/er.2005-0009
  • 28. Bridgeman, M. B., Abazia, D. T. (2017). Medicinal Cannabis: History, Pharmacology, And Implications for the Acute Care Setting. P & T : a peer-reviewed journal for formulary management, 42(3), 180-188. [CrossRef]
  • 29. Di Marzo, V. (2008). Targeting the endocannabinoid system: to enhance or reduce? Nat Rev Drug Discov, 7(5), 438-455. [CrossRef]
  • 30. Pacher, P., Mechoulam, R. (2011). Is lipid signaling through cannabinoid 2 receptors part of a protective system? Progress in Lipid Research, 50(2), 193-211. [CrossRef]
  • 31. Hasenoehrl, C., Taschler, U., Storr, M., Schicho, R. (2016). The gastrointestinal tract – a central organ of cannabinoid signaling in health and disease. Neurogastroenterology & Motility, 28(12), 1765-1780. [CrossRef]
  • 32. Ligresti, A., Petrocellis, L. D., Marzo, V. D. (2016). From Phytocannabinoids to Cannabinoid Receptors and Endocannabinoids: Pleiotropic Physiological and Pathological Roles Through Complex Pharmacology. Physiological Reviews, 96(4), 1593-1659. [CrossRef]
  • 33. Benito, C., Romero, J. P., Tolón, R. M., Clemente, D., Docagne, F., Hillard, C. J., Romero, J. (2007). Cannabinoid CB1 and CB2 Receptors and Fatty Acid Amide Hydrolase Are Specific Markers of Plaque Cell Subtypes in Human Multiple Sclerosis. The Journal of Neuroscience, 27(9), 2396. [CrossRef]
  • 34. Jean-Gilles, L., Feng, S., Tench, C. R., Chapman, V., Kendall, D. A., Barrett, D. A., Constantinescu, C. S. (2009). Plasma endocannabinoid levels in multiple sclerosis. Journal of the Neurological Sciences, 287(1), 212-215. [CrossRef]
  • 35. Killestein, J., Hoogervorst, E. L., Reif, M., Kalkers, N. F., Van Loenen, A. C., Staats, P. G., Polman, C. H. (2002). Safety, tolerability, and efficacy of orally administered cannabinoids in MS. Neurology, 58(9), 1404-1407. [CrossRef]
  • 36. Ungerleider, J. T., Andyrsiak, T., Fairbanks, L., Ellison, G. W., Myers, L. W. (1987). Delta-9- THC in the treatment of spasticity associated with multiple sclerosis. Adv Alcohol Subst Abuse, 7(1), 39-50. [CrossRef]
  • 37. Zajicek, J. P., Hobart, J. C., Slade, A., Barnes, D., Mattison, P. G. (2012). MUltiple Sclerosis and Extract of Cannabis: results of the MUSEC trial. Journal of Neurology, Neurosurgery & Psychiatry, 83(11), 1125. [CrossRef]
  • 38. Collin, C., Ehler, E., Waberzinek, G., Alsindi, Z., Davies, P., Powell, K., Ambler, Z. (2010). A double-blind, randomized, placebo-controlled, parallel-group study of Sativex, in subjects with symptoms of spasticity due to multiple sclerosis. Neurological Research, 32(5), 451-459. [CrossRef]
  • 39. Wade, D. T., Makela, P., Robson, P., House, H.,Bateman, C. (2004). 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. Multiple Sclerosis Journal, 10(4), 434-441. [CrossRef]
  • 40. Rog, D. J., Nurmikko, T. J., Young, C. A. (2007). Oromucosal Δ9- tetrahydrocannabinol/cannabidiol for neuropathic pain associated with multiple sclerosis: An uncontrolled, open-label, 2-year extension trial. Clinical Therapeutics, 29(9), 2068-2079. [CrossRef]
  • 41. Wallace, M. J., Blair, R. E., Falenski, K. W., Martin, B. R., DeLorenzo, R. J. (2003). The Endogenous Cannabinoid System Regulates Seizure Frequency and Duration in a Model of Temporal Lobe Epilepsy. Journal of Pharmacology and Experimental Therapeutics, 307(1), 129. [CrossRef]
  • 42. Ellison, J. M., Gelwan, E., Ogletree, J. (1990). Complex partial seizure symptoms affected by marijuana abuse. J Clin Psychiatry, 51(10), 439-440. [CrossRef]
  • 43. Gross, D. W., Hamm, J., Ashworth, N. L., Quigley, D. (2004). Marijuana use and epilepsy: prevalence in patients of a tertiary care epilepsy center. Neurology, 62(11), 2095-2097. [CrossRef]
  • 44. Mortati, K., Dworetzky, B., Devinsky, O. (2007). Marijuana: an effective antiepileptic treatment in partial epilepsy? A case report and review of the literature. Rev Neurol Dis, 4(2), 103-106.
  • 45. Hamerle, M., Ghaeni, L., Kowski, A., Weissinger, F., Holtkamp, M. (2014). Cannabis and other illicit drug use in epilepsy patients. European Journal of Neurology, 21(1), 167-170. [CrossRef]
  • 46. Hussain, S. A., Zhou, R., Jacobson, C., Weng, J., Cheng, E., Lay, J., Sankar, R. (2015). Perceived efficacy of cannabidiol-enriched cannabis extracts for treatment of pediatric epilepsy: A potential role for infantile spasms and Lennox–Gastaut syndrome. Epilepsy & Behavior, 47, 138-141. [CrossRef]
  • 47. Porter, B. E., Jacobson, C. (2013). Report of a parent survey of cannabidiol-enriched cannabis use in pediatric treatment-resistant epilepsy. Epilepsy & Behavior, 29(3), 574-577. [CrossRef]
  • 48. Gofshteyn, J. S., Wilfong, A., Devinsky, O., Bluvstein, J., Charuta, J., Ciliberto, M. A., Marsh, E. D. (2017). Cannabidiol as a Potential Treatment for Febrile Infection-Related Epilepsy Syndrome (FIRES) in the Acute and Chronic Phases. Journal of Child Neurology, 32(1), 35-40. [CrossRef]
  • 49. Suraev, A., Lintzeris, N., Stuart, J., Kevin, R. C., Blackburn, R., Richards, E., McGregor, I. S. (2018). Composition and Use of Cannabis Extracts for Childhood Epilepsy in the Australian Community. Scientific Reports, 8(1), 10154. [CrossRef]
  • 50. Stampanoni Bassi M., (2017). Cannabinoids in Parkinson's Disease.. Cannabis and Cannabinoid Research, 2(1), 21-29. [CrossRef]
  • 51. Basavarajappa, B. S., Shivakumar, M., Joshi, V., Subbanna, S. (2017). Endocannabinoid system in neurodegenerative disorders. J Neurochem, 142(5), 624-648. [CrossRef]
  • 52. Hurley, M. J., Mash, D. C., Jenner, P. (2003b). Expression of cannabinoid CB1 receptor mRNA in basal ganglia of normal and parkinsonian human brain. J Neural Transm (Vienna), 110(11), 1279-1288. [CrossRef]
  • 53. Hurley, M. J., Mash, D. C., Jenner, P. (2003a). Expression of cannabinoid CB1 receptor mRNA in basal ganglia of normal and parkinsonian human brain. Journal of Neural Transmission, 110(11), 1279-1288. [CrossRef]
  • 54. El Banoua, F., Caraballo, I., Flores, J. A., Galan-Rodriguez, B., Fernandez-Espejo, E. (2004). Effects on turning of microinjections into basal ganglia of D1 and D2 dopamine receptors agonists and the cannabinoid CB1 antagonist SR141716A in a rat Parkinson's model. Neurobiology of Disease, 16(2), 377-385. [CrossRef]
  • 55. Kelsey, J. E., Harris, O., Cassin, J. (2009). The CB1 antagonist rimonabant is adjunctively therapeutic as well as monotherapeutic in an animal model of Parkinson's disease. Behavioural Brain Research, 203(2), 304-307. [CrossRef]
  • 56. Meschler, J. P., Howlett, A. C., Madras, B. K. (2001). Cannabinoid receptor agonist and antagonist effects on motor function in normal and 1-methyl-4-phenyl-1,2,5,6- tetrahydropyridine (MPTP)-treated non-human primates. Psychopharmacology (Berl), 156(1), 79-85. [CrossRef]
  • 57. Benito, C., Núñez, E., Tolón, R. M., Carrier, E. J., Rábano, A., Hillard, C. J., Romero, J. (2003). Cannabinoid CB2 receptors and fatty acid amide hydrolase are selectively overexpressed in neuritic plaque-associated glia in Alzheimer's disease brains. J Neurosci, 23(35), 11136-11141. [CrossRef]
  • 58. Ramírez, B. G., Blázquez, C., del Pulgar, T. G., Guzmán, M., de Ceballos, M. L. (2005). Prevention of Alzheimer's Disease Pathology by Cannabinoids: Neuroprotection Mediated by Blockade of Microglial Activation. The Journal of Neuroscience, 25(8), 1904. [CrossRef]
  • 59. Koppel, J., Vingtdeux, V., Marambaud, P., d’Abramo, C., Jimenez, H., Stauber, M., Davies, P. (2014). CB2 Receptor Deficiency Increases Amyloid Pathology and Alters Tau Processing in a Transgenic Mouse Model of Alzheimer’s Disease. Molecular Medicine, 20(1), 29-36. [CrossRef]
  • 60. Solas, M., Francis, P. T., Franco, R., Ramirez, M. J. (2013). CB2 receptor and amyloid pathology in frontal cortex of Alzheimer's disease patients. Neurobiology of Aging, 34(3), 805- 808. [CrossRef]
  • 61. Tolón, R. M., Núñez, E., Pazos, M. R., Benito, C., Castillo, A. I., Martínez-Orgado, J. A., Romero, J. (2009). The activation of cannabinoid CB2 receptors stimulates in situ and in vitro beta-amyloid removal by human macrophages. Brain Research, 1283, 148-154. [CrossRe]
  • 62. Wu, J., Bie, B., Yang, H., Xu, J. J., Brown, D. L., Naguib, M. (2013). Activation of the CB2 receptor system reverses amyloid-induced memory deficiency. Neurobiology of Aging, 34(3), 791-804. [CrossRef]
  • 63. Esposito, G., Scuderi, C., Savani, C., Steardo Jr, L., De Filippis, D., Cottone, P.,Steardo, L. (2007). Cannabidiol in vivo blunts β-amyloid induced neuroinflammation by suppressing IL-1β and iNOS expression. British journal of pharmacology, 151(8), 1272-1279. [CrossRef]
  • 64. Glass, M., Faull, R. L., Dragunow, M. (1993). Loss of cannabinoid receptors in the substantia nigra in Huntington's disease. Neuroscience, 56(3), 523-527. [CrossRef]
  • 65. Richfield, E. K., Herkenham, M. (1994). Selective vulnerability in Huntington's disease: Preferential loss of cannabinoid receptors in lateral globus pallidus. Annals of Neurology, 36(4), 577-584. [CrossRef]
  • 66. Denovan-Wright, E. M., Robertson, H. A. (2000). Cannabinoid receptor messenger RNA levels decrease in a subset of neurons of the lateral striatum, cortex and hippocampus of transgenic Huntington's disease mice. Neuroscience, 98(4), 705-713. [CrossRef]
  • 67. Dowie, M. J., Bradshaw, H. B., Howard, M. L., Nicholson, L. F., Faull, R. L., Hannan, A. J., Glass, M. (2009). Altered CB1 receptor and endocannabinoid levels precede motor symptom onset in a transgenic mouse model of Huntington's disease. Neuroscience, 163(1), 456-465. [CrossRef]
  • 68. McCaw, E. A., Hu, H., Gomez, G. T., Hebb, A. L., Kelly, M. E., Denovan-Wright, E. M. (2004). Structure, expression and regulation of the cannabinoid receptor gene (CB1) in Huntington's disease transgenic mice. Eur J Biochem, 271(23-24), 4909-4920. [CrossRef]
  • 69. Consroe, P., Laguna, J., Allender, J., Snider, S., Stern, L., Sandyk, R., Schram, K. (1991). Controlled clinical trial of cannabidiol in Huntington's disease. Pharmacol Biochem Behav, 40(3), 701-708. [CrossRef]
  • 70. López-Sendón Moreno, J. L., García Caldentey, J., Trigo Cubillo, P., Ruiz Romero, C., García Ribas, G., Alonso Arias, M. A. A., García de Yébenes Prous, J. (2016). A double-blind, randomized, cross-over, placebo-controlled, pilot trial with Sativex in Huntington’s disease. Journal of Neurology, 263(7), 1390-1400. [CrossRef]
  • 71. Curtis, A., Mitchell, I., Patel, S., Ives, N., Rickards, H. (2009). A pilot study using nabilone for symptomatic treatment in Huntington's disease. Movement Disorders, 24(15), 2254-2259. [CrossRef]
  • 72. Alles, S. R. A., Smith, P. A. (2018). Etiology and Pharmacology of Neuropathic Pain. Pharmacol Rev, 70(2), 315-347. [CrossRef]
  • 73. Abrams, D. I., Jay, C. A., Shade, S. B., Vizoso, H., Reda, H., Press, S., . Petersen, K. L. (2007). Cannabis in painful HIV-associated sensory neuropathy: a randomized placebo-controlled trial. Neurology, 68(7), 515-521. [CrossRef]
  • 74. Ellis, R. J., Toperoff, W., Vaida, F., van den Brande, G., Gonzales, J., Gouaux, B., Atkinson, J. H. (2009). Smoked medicinal cannabis for neuropathic pain in HIV: a randomized, crossover clinical trial. Neuropsychopharmacology, 34(3), 672-680. [CrossRef]
  • 75. 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 : official journal of the American Pain Society, 9(6), 506-521. [CrossRef]
  • 76. Borgelt, L. M., Franson, K. L., Nussbaum, A. M., Wang, G. S. (2013). The pharmacologic and clinical effects of medical cannabis. Pharmacotherapy, 33(2), 195-209. [CrossRef]
  • 77. 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 trial. Pain, 133(1-3), 210-220. [CrossRef]
  • 78. Rog, D. J., Nurmikko, T. J.,Young, C. A. (2007). Oromucosal delta9- tetrahydrocannabinol/cannabidiol for neuropathic pain associated with multiple sclerosis: an uncontrolled, open-label, 2-year extension trial. Clin Ther, 29(9), 2068-2079. [CrossRef]
  • 79. Häuser, W., Fitzcharles, M. A., Radbruch, L., Petzke, F. (2017). Cannabinoids in Pain Management and Palliative Medicine. Dtsch Arztebl Int, 114(38), 627-634. [CrossRef]
  • 80. Fitzcharles, M. A., Baerwald, C., Ablin, J., Häuser, W. (2016). 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, 30(1), 47-61. [CrossRef]
  • 81. Fitzcharles, M. A., Ste-Marie, P. A., Häuser, W., Clauw, D. J., Jamal, S., Karsh, J.,Walsh, Z. (2016). Efficacy, Tolerability, and Safety of Cannabinoid Treatments in the Rheumatic Diseases: A Systematic Review of Randomized Controlled Trials. Arthritis Care Res (Hoboken), 68(5), 681-688. [CrossRef]
  • 82. Fraguas-Sánchez, A. I., Martín-Sabroso, C., Torres-Suárez, A. I. (2018). Insights into the effects of the endocannabinoid system in cancer: a review. British journal of pharmacology, 175(13), 2566-2580. [CrossRef]
  • 83. Sredni, S. T., Huang, C.-C., Suzuki, M., Pundy, T., Chou, P., Tomita, T. (2016). Spontaneous involution of pediatric low-grade gliomas: high expression of cannabinoid receptor 1 (CNR1) at the time of diagnosis may indicate involvement of the endocannabinoid system. Child's Nervous System, 32(11), 2061-2067. [CrossRef]
  • 84. Wu, X., Han, L., Zhang, X., Li, L., Jiang, C., Qiu, Y., Fu, J. (2012). Alteration of endocannabinoid system in human gliomas. Journal of Neurochemistry, 120(5), 842-849. [CrossRef]
  • 85. Ellert-Miklaszewska, A., Grajkowska, W., Gabrusiewicz, K., Kaminska, B., Konarska, L. (2007). Distinctive pattern of cannabinoid receptor type II (CB2) expression in adult and pediatric brain tumors. Brain Res, 1137(1), 161-169. [CrossRef]
  • 86. Sánchez, C., de Ceballos, M. L., Gomez del Pulgar, T., Rueda, D., Corbacho, C., Velasco, G.,Guzmán, M. (2001). Inhibition of glioma growth in vivo by selective activation of the CB(2) cannabinoid receptor. Cancer Res, 61(15), 5784-5789.
  • 87. Schley, M., Ständer, S., Kerner, J., Vajkoczy, P., Schüpfer, G., Dusch, M., Konrad, C. (2009). Predominant CB2 receptor expression in endothelial cells of glioblastoma in humans. Brain Res Bull, 79(5), 333-337. [CrossRef]
  • 88. Hohmann, T., Grabiec, U., Ghadban, C., Feese, K., Dehghani, F. (2017). The influence of biomechanical properties and cannabinoids on tumor invasion. Cell adhesion & migration, 11(1), 54-67. [CrossRef]
  • 89. Ma, C., Wu, T.-T., Jiang, P.-C., Li, Z.-Q., Chen, X.-J., Fu, K., Gong, R. (2016). Anti- carcinogenic activity of anandamide on human glioma in vitro and in vivo. Molecular medicine reports, 13(2), 1558-1562. [CrossRef]
  • 90. Fowler, C. J., Jonsson, K. O., Andersson, A., Juntunen, J., Järvinen, T., Vandevoorde, S., .Smart, D. (2003). Inhibition of C6 glioma cell proliferation by anandamide, 1-arachidonoylglycerol, and by a water soluble phosphate ester of anandamide: variability in response and involvement of arachidonic acid. Biochem Pharmacol, 66(5), 757-767. [CrossRef]
  • 91. Jacobsson, S. O., Wallin, T., Fowler, C. J. (2001). Inhibition of rat C6 glioma cell proliferation by endogenous and synthetic cannabinoids. Relative involvement of cannabinoid and vanilloid receptors. J Pharmacol Exp Ther, 299(3), 951-959. [CrossRef]
  • 92. Caffarel, M. M., Andradas, C., Mira, E., Pérez-Gómez, E., Cerutti, C., Moreno-Bueno, G., Sánchez, C. (2010). Cannabinoids reduce ErbB2-driven breast cancer progression through Akt inhibition. Mol Cancer, 9, 196. [CrossRef]
  • 93. Bisogno, T., Katayama, K., Melck, D., Ueda, N., De Petrocellis, L., Yamamoto, S., Di Marzo, V. (1998). Biosynthesis and degradation of bioactive fatty acid amides in human breast cancer and rat pheochromocytoma cells--implications for cell proliferation and differentiation. Eur J Biochem, 254(3), 634-642. [CrossRef]
  • 94. Melck, D., De Petrocellis, L., Orlando, P., Bisogno, T., Laezza, C., Bifulco, M., Di Marzo, V. (2000). Suppression of nerve growth factor Trk receptors and prolactin receptors by endocannabinoids leads to inhibition of human breast and prostate cancer cell proliferation. Endocrinology, 141(1), 118-126. [CrossRef]
  • 95. Ligresti, A., Moriello, A. S., Starowicz, K., Matias, I., Pisanti, S., De Petrocellis, L., Di Marzo, V. (2006). Antitumor activity of plant cannabinoids with emphasis on the effect of cannabidiol on human breast carcinoma. J Pharmacol Exp Ther, 318(3), 1375-1387. [CrossRef]
  • 96. Qamri, Z., Preet, A., Nasser, M. W., Bass, C. E., Leone, G., Barsky, S. H., Ganju, R. K. (2009). Synthetic cannabinoid receptor agonists inhibit tumor growth and metastasis of breast cancer. Mol Cancer Ther, 8(11), 3117-3129. [CrossRef]
  • 97. Chung, S. C., Hammarsten, P., Josefsson, A., Stattin, P., Granfors, T., Egevad, L., Fowler, C. J. (2009). A high cannabinoid CB(1) receptor immunoreactivity is associated with disease severity and outcome in prostate cancer. Eur J Cancer, 45(1), 174-182. [CrossRef]
  • 98. Cipriano, M., Häggström, J., Hammarsten, P., Fowler, C. J. (2013). Association between Cannabinoid CB1 Receptor Expression and Akt Signalling in Prostate Cancer. PLoS One, 8(6), e65798. [CrossRef]
  • 99. Orellana-Serradell, O., Poblete, C. E., Sanchez, C., Castellón, E. A., Gallegos, I., Huidobro, C., Contreras, H. R. (2015). Proapoptotic effect of endocannabinoids in prostate cancer cells. Oncol Rep, 33(4), 1599-1608. [CrossRef]
  • 100. Sarfaraz, S., Afaq, F., Adhami, V. M., Mukhtar, H. (2005). Cannabinoid receptor as a novel target for the treatment of prostate cancer. Cancer Res, 65(5), 1635-1641. [CrossRef]
  • 101. Mimeault, M., Pommery, N., Wattez, N., Bailly, C., Hénichart, J. P. (2003). Anti-proliferative and apoptotic effects of anandamide in human prostatic cancer cell lines: implication of epidermal growth factor receptor down-regulation and ceramide production. Prostate, 56(1), 1- 12. [CrossRef]
  • 102. Nithipatikom, K., Isbell, M. A., Endsley, M. P., Woodliff, J. E., Campbell, W. B. (2011). Anti- proliferative effect of a putative endocannabinoid, 2-arachidonylglyceryl ether in prostate carcinoma cells. Prostaglandins & other lipid mediators, 94(1-2), 34-43. [CrossRef]
  • 103. DeMorrow, S., Francis, H., Gaudio, E., Venter, J., Franchitto, A., Kopriva, S., Alpini, G. (2008). The endocannabinoid anandamide inhibits cholangiocarcinoma growth via activation of the noncanonical Wnt signaling pathway. American journal of physiology. Gastrointestinal and liver physiology, 295(6), G1150-G1158. [CrossRef]
  • 104. Frampton, G., Coufal, M., Li, H., Ramirez, J., DeMorrow, S. (2010). Opposing actions of endocannabinoids on cholangiocarcinoma growth is via the differential activation of Notch signaling. Experimental cell research, 316(9), 1465-1478. [CrossRef]
  • 105. Ramer, R., Schwarz, R., Hinz, B. (2019). Modulation of the Endocannabinoid System as a Potential Anticancer Strategy. Frontiers in pharmacology, 10, 430-430. [CrossRef]
  • 106. Vara, D., Salazar, M., Olea-Herrero, N., Guzmán, M., Velasco, G., Díaz-Laviada, I. (2011). Anti-tumoral action of cannabinoids on hepatocellular carcinoma: role of AMPK-dependent activation of autophagy. Cell Death Differ, 18(7), 1099-1111. [CrossRef]
  • 107. Xu, D., Wang, J., Zhou, Z., He, Z., Zhao, Q. (2015). Cannabinoid WIN55, 212-2 induces cell cycle arrest and inhibits the proliferation and migration of human BEL7402 hepatocellular carcinoma cells Corrigendum in /mmr/13/1/1054. Mol Med Rep, 12(6), 7963-7970. [CrossRef]
  • 108. Sallan, S. E., Zinberg, N. E., Frei, E., 3rd. (1975). Antiemetic effect of delta-9- tetrahydrocannabinol in patients receiving cancer chemotherapy. N Engl J Med, 293(16), 795- 797. [CrossRef]
  • 109. Darmani, N. A. (2001b). Delta(9)-tetrahydrocannabinol and synthetic cannabinoids prevent emesis produced by the cannabinoid CB(1) receptor antagonist/inverse agonist SR 141716A. Neuropsychopharmacology, 24(2), 198-203. [CrossRef]
  • 110. Darmani, N. A. (2001a). Delta-9-tetrahydrocannabinol differentially suppresses cisplatin- induced emesis and indices of motor function via cannabinoid CB(1) receptors in the least shrew. Pharmacol Biochem Behav, 69(1-2), 239-249. [CrossRef]
  • 111. Pomeroy, M., Fennelly, J. J., Towers, M. (1986). Prospective randomized double-blind trial of nabilone versus domperidone in the treatment of cytotoxic-induced emesis. Cancer Chemother Pharmacol, 17(3), 285-288. [CrossRef]
  • 112. Meiri, E., Jhangiani, H., Vredenburgh, J. J., Barbato, L. M., Carter, F. J., Yang, H. M., Baranowski, V. (2007). Efficacy of dronabinol alone and in combination with ondansetron versus ondansetron alone for delayed chemotherapy-induced nausea and vomiting. Curr Med Res Opin, 23(3), 533-543. [CrossRef]
  • 113. Polito, S., MacDonald, T., Romanick, M., Jupp, J., Wiernikowski, J., Vennettilli, A., Dupuis, L. L. (2018). Safety and efficacy of nabilone for acute chemotherapy-induced vomiting prophylaxis in pediatric patients: A multicenter, retrospective review. Pediatr Blood Cancer, 65(12), e27374. [CrossRef]
  • 114. Kleine-Brueggeney, M., Greif, R., Brenneisen, R., Urwyler, N., Stueber, F., Theiler, L. G. (2015). Intravenous Delta-9-Tetrahydrocannabinol to Prevent Postoperative Nausea and Vomiting: A Randomized Controlled Trial. Anesth Analg, 121(5), 1157-1164. [CrossRef]
  • 115. Pellati, F., Borgonetti, V., Brighenti, V., Biagi, M., Benvenuti, S., Corsi, L. (2018). Cannabis sativa L. and Nonpsychoactive Cannabinoids: Their Chemistry and Role against Oxidative Stress, Inflammation, and Cancer. BioMed research international, 2018, 1691428. [CrossRef]
  • 116. Costa, B., Colleoni, M., Conti, S., Parolaro, D., Franke, C., Trovato, A. E., Giagnoni, G. (2004). Oral anti-inflammatory activity of cannabidiol, a non-psychoactive constituent of cannabis, in acute carrageenan-induced inflammation in the rat paw. Naunyn-Schmiedeberg's archives of pharmacology, 369(3), 294–299. [CrossRef]
  • 117. Petrosino, S., Verde, R., Vaia, M., Allarà, M., Iuvone, T., Di Marzo, V. (2018). Anti- inflammatory Properties of Cannabidiol, a Nonpsychotropic Cannabinoid, in Experimental Allergic Contact Dermatitis. The Journal of pharmacology and experimental therapeutics, 365(3), 652–663. [CrossRef]
  • 118. Mackie, K., Stella, N. (2006). Cannabinoid receptors and endocannabinoids: evidence for new players. The AAPS journal, 8(2), E298–E306. [CrossRef]
  • 119. Parlakpınar, Hakan Celbis, Osman Ozhan, Onural Petekkaya, Semih Samdanci, Emine Ermis, Necip Koparir, Pelin Soylu, Ozcan Acet, Ahmet. (2016). Cardiovascular effects of JWH-018 from synthetic cannabinoids [Sentetik kannabinoidlerden JWH-018'in kardiyovaskuler etkileri], Medicine Science., 5. 1. [CrossRef]
  • 120. Köksoy S , Başalp A . (2021). Effect of synthetic cannabinoids (JWH-018) on antibody response to HBV vaccination, Mehmet Akif Ersoy Üniversitesi Sağlık Bilimleri Enstitüsü Dergisi, 2021; 9(1): 21-26. [CrossRef]
  • 121. Kogan, N. M., Mechoulam, R. (2007). Cannabinoids in health and disease. Dialogues in clinical neuroscience, 9(4), 413–430. [CrossRef]
  • 122. Wong, K. U., Baum, C. R. (2019). Acute Cannabis Toxicity. Pediatric emergency care, 35(11), 799–804. [CrossRef]
  • 123. Kuzucuoğlu, T., Uçkun, S., Temizel, F., Arslan, G., Çevik, B. (2018). Kannabinoid İntoksikasyonlarının Yoğun Bakım Sonuçları: Olgu Serisi. Dahili ve Cerrahi Bilimler Yoğun Bakım Dergisi.; 9(3): 84 - 87. [CrossRef]
Ankara Üniversitesi Eczacılık Fakültesi Dergisi-Cover
  • ISSN: 1015-3918
  • Yayın Aralığı: Yılda 3 Sayı
  • Başlangıç: 2016
  • Yayıncı: Ankara Üniversitesi Eczacılık Fakültesi
Sayıdaki Diğer Makaleler

YENİ İZOLE EDILEN VB_K1 BAKTERİYOFAJININ İZOLASYONU VE ESBL POZİTİF KLEBSİELLA TÜRLERİ ÜZERİNE DUYARLILIĞININ ARAŞTIRILMASI

Hilal Basak EROL, Banu KAŞKATEPE

ORGANİK REAKSİYONLARDA METAL İÇERMEYEN KATALİZÖR KULLANILARAK İNDOL TÜREVLERİNİN SENTEZİ

Ratnesh DAS, Hemlata DANGİ, Sushil KASHAW

GLYCYRRHIZA GLABRA'NIN BAZI TIBBİ ÖZELLİKLERİ (MEYAN KÖKÜ)

Falah Saleh MOHAMMED, Nuh KORKMAZ, Mehmet DOĞAN, Ali Erdem ŞABİK, Mustafa SEVİNDİK

TÜRKİYE’DE DOĞAL OLARAK YETİŞEN CELTIS AUSTRALIS L. VE C. TOURNEFORTII LAM. (CANNABACEAE) MEYVELERİNİN YAĞ ASİTİ BİLEŞİMLERİ VE ANTİMİKROBİYAL ETKİLERİNİN DEĞERLENDİRİLMESİ

Gülderen YILMAZ, Gözde ÖZTÜRK, Betül DEMİRCİ

BİR SPEKTROFOTOMETRİK İKİLİ KARIŞIM ANALİZİNİN DOĞRULAMA ÇALIŞMASI

Özgür ÜSTÜNDAĞ, Erdal DİNÇ

SİKLOOKSİJENAZ İNHİBİTÖRLERİ OLARAK BAZI 4,5-DİHİDRO-1H-PİRAZOL TÜREVLERİ ÜZERİNDE MOLEKÜLER DOKİNG ÇALIŞMALARI

Begüm EVRANOS AKSÖZ

2-((4-R-5-R1-4H-1,2,4-TRİAZOL-3-İL)TİYO)ASETALDEHİTLERİN İLİDENHİDRAZİDLERİNİN SENTEZİ, ANTİMİKROBİYAL VE ANTİFUNGAL AKTİVİTES

Olexandr PANASENKO, Roman SHCHERBYNA, Olha POLONETS, Natalia NEDOREZANIUK, Maryna DUCHENKO

3- VE 4-SÜBSTİTÜE-5-((3-FENİLPROPİL)TİYO)-4H-1,2,4-TRİAZOLLERİN MİKRODALGA SENTEZİ

Andrey SAFONOV, Alina NEVMYVAKA, Oleksandr PANASENKO, Yevgenii KNYSH

TIP VE ECZACILIK ALANINDAKİ 1,2,4-TRİAZOL TÜREVLERİ VE UYGULAMA ÖNERİLERİ

Yurii SAMELİUK, Tetyana KAPLAUSHENKO, Fadi AL ZEDAN

ALOE VERA (L.) BURM. F. (SARISABIR) BİTKİSİNİN FİTOTERAPİDE KULLANIMI

Hilal Ahsen TUNÇAY, Gülen İrem KAYA