247
Views
1
CrossRef citations to date
0
Altmetric
Review

Cannabinoid treatment for the symptoms of autism spectrum disorder

&
Pages 65-79 | Received 01 Nov 2023, Accepted 12 Jan 2024, Published online: 23 Jan 2024

References

  • Lord C, Elsabbagh M, Baird G, et al. Autism spectrum disorder. Lancet (London, England). 2018 Aug 11;392(10146):508–520. doi: 10.1016/S0140-6736(18)31129-2
  • Hirota T, King BH. Autism Spectrum Disorder: A Review. JAMA. 2023 Jan 10;329(2):157–168. doi: 10.1001/jama.2022.23661
  • Maenner M, Warren Z, Robinson-Williams A, et al. Prevalence and characteristics of autism spectrum disorder among children aged 8 years — autism and developmental disabilities monitoring network, 11 Sites, United States, 2020. Cdc, editor. CDC: CDC; 2023.
  • Szkudlarek HJ, Desai SJ, Renard J, et al. Delta-9-tetrahydrocannabinol and cannabidiol produce dissociable effects on prefrontal cortical executive function and regulation of affective behaviors. Neuropsychopharmacology. 2019 Mar;44(4):817–825.
  • Castillo-Arellano J, Canseco-Alba A, Cutler SJ, et al. The polypharmacological effects of cannabidiol. Molecules. 2023 Apr 6;28(7):3271. doi: 10.3390/molecules28073271
  • Felberbaum M. 2018. https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm611046.htm
  • Devinsky O, Cross JH, Laux L, et al. Trial of cannabidiol for drug-resistant seizures in the Dravet syndrome. N Engl J Med. 2017 May 25;376(21):2011–2020. doi: 10.1056/NEJMoa1611618
  • Devinsky O, Patel AD, Cross JH, et al. Effect Of cannabidiol on drop seizures in the lennox-gastaut syndrome. N Engl J Med. 2018 May 17;378(20):1888–1897. doi: 10.1056/NEJMoa1714631
  • Thiele EA, Bebin EM, Bhathal H, et al. Add-on cannabidiol treatment for drug-resistant seizures in tuberous sclerosis complex: a placebo-controlled randomized clinical trial. JAMA Neurol. 2021 Mar 1;78(3):285–292. doi: 10.1001/jamaneurol.2020.4607
  • Aran A, Cassuto H, Lubotzky A, et al. Brief report: cannabidiol-rich cannabis in children with autism spectrum disorder and severe behavioral problems-a retrospective feasibility study. J Autism Dev Disord. 2019 Mar;49(3):1284–1288.
  • Barchel D, Stolar O, De-Haan T, et al. Oral cannabidiol use in children with autism spectrum disorder to treat related symptoms and Co-morbidities. Front Pharmacol. 2018;9:1521. doi: 10.3389/fphar.2018.01521
  • Fleury-Teixeira P, Caixeta FV, Ramires da Silva LC, et al. Effects of CBD-Enriched cannabis sativa extract on autism spectrum disorder symptoms: an observational study of 18 participants undergoing compassionate use. Front Neurol. 2019;10:1145. doi: 10.3389/fneur.2019.01145
  • Bar-Lev Schleider L, Mechoulam R, Saban N, et al. Real life experience of medical cannabis treatment in autism: analysis of safety and efficacy. Sci Rep. 2019 Jan 17;9(1):200.
  • Montagner PSS, Medeiros W, da Silva LCR, et al. Individually tailored dosage regimen of full-spectrum cannabis extracts for autistic core and comorbid symptoms: a real-life report of multi-symptomatic benefits. Front Psychiatry. 2023;14:1210155. doi: 10.3389/fpsyt.2023.1210155
  • Hacohen M, Stolar OE, Berkovitch M, et al. Children and adolescents with ASD treated with CBD-rich cannabis exhibit significant improvements particularly in social symptoms: an open label study. Transl Psychiatry. 2022 Sep 9;12(1):375. doi: 10.1038/s41398-022-02104-8
  • Stolar O, Hazan A, Vissoker RE, et al. Medical cannabis for the treatment of comorbid symptoms in children with autism spectrum disorder: an interim analysis of biochemical safety. Front Pharmacol. 2022;13:977484. doi: 10.3389/fphar.2022.977484
  • Bilge S, Ekici B. CBD-enriched cannabis for autism spectrum disorder: an experience of a single center in Turkey and reviews of the literature. J Cannabis Res. 2021 Dec 16;3(1):53. doi: 10.1186/s42238-021-00108-7
  • Buescher AV, Cidav Z, Knapp M, et al. Costs of autism spectrum disorders in the United Kingdom and the United States. JAMA Pediatr. 2014 Aug;168(8):721–728. doi: 10.1001/jamapediatrics.2014.210
  • Goel R, Hong JS, Findling RL, et al. An update on pharmacotherapy of autism spectrum disorder in children and adolescents. Inter Rev Psychiatry (Abingdon England). 2018 Feb;30(1):78–95. doi: 10.1080/09540261.2018.1458706
  • Iffland M, Livingstone N, Jorgensen M, et al. Pharmacological intervention for irritability, aggression, and self-injury in autism spectrum disorder (ASD). Cochrane Database Syst Rev. 2023 Oct 9;10(10):Cd011769. doi: 10.1002/14651858.CD011769.pub2
  • Fieiras C, Chen MH, Escobar Liquitay CM, et al. Risperidone and aripiprazole for autism spectrum disorder in children: an overview of systematic reviews. BMJ Evid Based Med. 2023 Feb;28(1):7–14.
  • Salazar de Pablo G, Pastor Jordá C, Vaquerizo-Serrano J, et al. Systematic review and meta-analysis: efficacy of pharmacological interventions for irritability and emotional dysregulation in autism spectrum disorder and predictors of response. Journal Of The American Academy Of Child & Adolescent Psychiatry. 2023 Feb;62(2):151–168.
  • Pruneti C, Coscioni G, Guidotti S. Evaluation of the effectiveness of behavioral interventions for autism spectrum disorders: a systematic review of randomized controlled trials and quasi-experimental studies. Clin Child Psychol Psychiatry. 2023 Oct 6;29(1):213–231. doi: 10.1177/13591045231205614
  • Sikich L, Kolevzon A, King BH, et al. Intranasal oxytocin in children and adolescents with autism spectrum disorder. N Engl J Med. 2021 Oct 14;385(16):1462–1473. doi: 10.1056/NEJMoa2103583
  • Guastella AJ, Boulton KA, Whitehouse AJO, et al. The effect of oxytocin nasal spray on social interaction in young children with autism: a randomized clinical trial. Mol Psychiatry. 2023 Feb;28(2):834–842.
  • Daniels N, Moerkerke M, Steyaert J, et al. Effects of multiple-dose intranasal oxytocin administration on social responsiveness in children with autism: a randomized, placebo-controlled trial. Mol Autism. 2023 Apr 20;14(1):16. doi: 10.1186/s13229-023-00546-5
  • Hollander E, Jacob S, Jou R, et al. Balovaptan vs placebo for social communication in childhood autism spectrum disorder: a randomized clinical trial. JAMA Psychiatry. 2022 Aug 1;79(8):760–769. doi: 10.1001/jamapsychiatry.2022.1717
  • Jacob S, Veenstra-VanderWeele J, Murphy D, et al. Efficacy and safety of balovaptan for socialisation and communication difficulties in autistic adults in North America and Europe: a phase 3, randomised, placebo-controlled trial. Lancet Psychiatry. 2022 Mar;9(3):199–210.
  • Fuentes J, Parellada M, Georgoula C, et al. Bumetanide oral solution for the treatment of children and adolescents with autism spectrum disorder: results from two randomized phase III studies. Autism Res. 2023 Oct 4;16(10):2021–2034. doi: 10.1002/aur.3005
  • Ou J, Smith RC, Tobe RH, et al. Efficacy of sulforaphane in treatment of children with autism spectrum disorder: a randomized double-blind placebo-controlled multi-center trial. J Autism Dev Disord. 2022 Nov 24. doi: 10.1007/s10803-022-05784-9
  • Dai Y, Zhang L, Yu J, et al. Improved symptoms following bumetanide treatment in children aged 3-6 years with autism spectrum disorder: a randomized, double-blind, placebo-controlled trial. Sci Bull (Beijing). 2021 Aug 15;66(15):1591–1598. doi: 10.1016/j.scib.2021.01.008
  • Yamasue H, Kojima M, Kuwabara H, et al. Effect of a novel nasal oxytocin spray with enhanced bioavailability on autism: a randomized trial. Brain. 2022 Apr 18;145(2):490–499. doi: 10.1093/brain/awab291
  • Bolognani F, Del Valle Rubido M, Squassante L, et al. A phase 2 clinical trial of a vasopressin V1a receptor antagonist shows improved adaptive behaviors in men with autism spectrum disorder. Sci Transl Med. 2019 May 8;11(491). doi: 10.1126/scitranslmed.aat7838
  • Kimi S, Maiti R, Srinivasan A, et al. Efficacy and safety of V(1a) receptor antagonists in autism spectrum disorder: a meta-analysis. Int J Dev Neurosci. 2023 Aug 28. doi: 10.1002/jdn.10297
  • Hu L, Du X, Jiang Z, et al. Oxytocin treatment for core symptoms in children with autism spectrum disorder: a systematic review and meta-analysis. Eur J Clin Pharmacol. 2023 Oct;79(10):1357–1363.
  • Lee TM, Lee KM, Lee CY, et al. Effectiveness of N-acetylcysteine in autism spectrum disorders: a meta-analysis of randomized controlled trials. Aust N Z J Psychiatry. 2021 Feb;55(2):196–206.
  • Rodrigues R, Lai MC, Beswick A, et al. Practitioner review: pharmacological treatment of attention-deficit/hyperactivity disorder symptoms in children and youth with autism spectrum disorder: a systematic review and meta-analysis. J Child Psychol Psychiatr. 2021 Jun;62(6):680–700.
  • Scahill L, McCracken JT, King BH, et al. Extended-release guanfacine for hyperactivity in children with autism spectrum disorder. Am J Psychiatry. 2015 Dec;172(12):1197–206.
  • Gringras P, Nir T, Breddy J, et al. Efficacy and safety of pediatric prolonged-release melatonin for insomnia in children with autism spectrum disorder. J Am Acad Child Adolesc Psychiatry. 2017 Nov;56(11):948–957.e4.
  • Siafis S, Çıray O, Wu H, et al. Pharmacological and dietary-supplement treatments for autism spectrum disorder: a systematic review and network meta-analysis. Mol Autism. 2022 Mar 4;13(1):10. doi: 10.1186/s13229-022-00488-4
  • Liang SC, Sun CK, Fan HY, et al. Therapeutic effects of antidepressants for global improvement and subdomain symptoms of autism spectrum disorder: a systematic review and meta-analysis. J Psychiatry Neurosci. 2022 Jul;47(4):E299–e310.
  • Limbu B, Deb S, Roy M, et al. Randomised controlled trials of mood stabilisers for people with autism spectrum disorder: systematic review and meta-analysis. BJPsych Open. 2022 Feb 24;8(2):e52. doi: 10.1192/bjo.2022.18
  • Ure A, Cox GR, Haslam R, et al. Acetylcholinesterase inhibitors for autistic spectrum disorders. Cochrane Database Syst Rev. 2023 Jun 1;6(6):Cd013851. doi: 10.1002/14651858.CD013851.pub2
  • Brignell A, Marraffa C, Williams K, et al. Memantine for autism spectrum disorder. Cochrane Database Syst Rev. 2022 Aug 25;8(8):Cd013845. doi: 10.1002/14651858.CD013845.pub2
  • de la Torre-Aguilar MJ, Gomez-Fernandez A, Flores-Rojas K, et al. Docosahexaenoic and eicosapentaenoic intervention modifies plasma and erythrocyte omega-3 fatty acid profiles but not the clinical course of children with autism spectrum disorder: a randomized control trial. Front Nutr. 2022;9:790250.
  • Jacob S, Anagnostou E, Hollander E, et al. Large multicenter randomized trials in autism: key insights gained from the balovaptan clinical development program. Mol Autism. 2022 Jun 11;13(1):25. doi: 10.1186/s13229-022-00505-6
  • McPartland JC, Bernier RA, Jeste SS, et al. ThE autism biomarkers consortium for clinical trials (abc-ct): scientific context, study design, and progress toward biomarker qualification. Front Integr Neurosci. 2020;14:16. doi: 10.3389/fnint.2020.00016
  • Siafis S, Çıray O, Schneider-Thoma J, et al. Placebo response in pharmacological and dietary supplement trials of autism spectrum disorder (ASD): systematic review and meta-regression analysis. Mol Autism. 2020 Aug 26;11(1):66. doi: 10.1186/s13229-020-00372-z
  • Faja S, Sabatos-DeVito M, Sridhar A, et al. Evaluation of clinical assessments of social abilities for use in autism clinical trials by the autism biomarkers consortium for clinical trials. Autism Res. 2023 May;16(5):981–996.
  • Satterstrom FK, Kosmicki JA, Wang J, et al. Large-scale exome sequencing study implicates both developmental and functional changes in the neurobiology of autism. Cell. 2020 Feb 6;180(3):568–584.e23. doi: 10.1016/j.cell.2019.12.036
  • Fadila S, Beucher B, Dopeso-Reyes IG, et al. Viral vector-mediated expression of NaV1.1, after seizure onset, reduces epilepsy in mice with Dravet syndrome. J Clin Invest. 2023 Jun 15;133(12). doi: 10.1172/JCI159316
  • Tamura S, Nelson A, Spratt P, et al. CRISPR activation rescues abnormalities in SCN2A haploinsufficiency-associated autism spectrum disorder. bioRxiv. 2022. https://www.biorxiv.org/content/10.1101/2022.03.30.486483v1
  • Jourdon A, Wu F, Mariani J, et al. Modeling idiopathic autism in forebrain organoids reveals an imbalance of excitatory cortical neuron subtypes during early neurogenesis. Nat Neurosci. 2023 Sep;26(9):1505–1515.
  • Modabbernia A, Velthorst E, Reichenberg A. Environmental risk factors for autism: an evidence-based review of systematic reviews and meta-analyses. Mol Autism. 2017;8(1):13. doi: 10.1186/s13229-017-0121-4
  • Guerrero-Alba R, Barragan-Iglesias P, Gonzalez-Hernandez A, et al. Some prospective alternatives for treating pain: the endocannabinoid system and its putative receptors GPR18 and GPR55. Front Pharmacol. 2018;9:1496. doi: 10.3389/fphar.2018.01496
  • Melancia F, Schiavi S, Servadio M, et al. Sex-specific autistic endophenotypes induced by prenatal exposure to valproic acid involve anandamide signalling. Br J Pharmacol. 2018 Sep;175(18):3699–3712.
  • Zamberletti E, Gabaglio M, Parolaro D. The endocannabinoid system and autism spectrum disorders: insights from animal models. Int J Mol Sci. 2017 Sep 7;18(9):1916. doi: 10.3390/ijms18091916
  • Wang W, Cox BM, Jia Y, et al. Treating a novel plasticity defect rescues episodic memory in Fragile X model mice. Mol Psychiatry. 2018 Aug;23(8):1798–1806.
  • Hosie S, Malone DT, Liu S, et al. Altered amygdala excitation and CB1 receptor modulation of aggressive behavior in the neuroligin-3(R451C) mouse model of autism. Front Cell Neurosci. 2018;12:234.
  • Poleg S, Kourieh E, Ruban A, et al. Behavioral aspects and neurobiological properties underlying medical cannabis treatment in Shank3 mouse model of autism spectrum disorder. Transl Psychiatry. 2021 Oct 13;11(1):524. doi: 10.1038/s41398-021-01612-3
  • Zou M, Liu Y, Xie S, et al. Alterations of the endocannabinoid system and its therapeutic potential in autism spectrum disorder. Open Biol. 2021 Feb;11(2):200306. doi: 10.1098/rsob.200306
  • Schiavi S, Manduca A, Carbone E, et al. Anandamide and 2-arachidonoylglycerol differentially modulate autistic-like traits in a genetic model of autism based on FMR1 deletion in rats. Neuropsychopharmacology. 2023 May;48(6):897–907.
  • Hurley MJ, Deacon RMJ, Chan AWE, et al. Reversal of behavioural phenotype by the cannabinoid-like compound VSN16R in fragile X syndrome mice. Brain. 2022 Mar 29;145(1):76–82. doi: 10.1093/brain/awab246
  • Karhson DS, Krasinska KM, Dallaire JA, et al. Plasma anandamide concentrations are lower in children with autism spectrum disorder. Mol Autism. 2018;9(1):18. doi: 10.1186/s13229-018-0203-y
  • Aran A, Eylon M, Harel M, et al. Lower circulating endocannabinoid levels in children with autism spectrum disorder. Mol Autism. 2019;10(1):2. doi: 10.1186/s13229-019-0256-6
  • Boggs DL, Nguyen JD, Morgenson D, et al. Clinical and preclinical evidence for functional interactions of cannabidiol and Δ(9)-Tetrahydrocannabinol. Neuropsychopharmacology. 2018 Jan;43(1):142–154.
  • Kozela E, Pietr M, Juknat A, et al. Cannabinoids Delta(9)-tetrahydrocannabinol and cannabidiol differentially inhibit the lipopolysaccharide-activated NF-kappaB and interferon-beta/STAT proinflammatory pathways in BV-2 microglial cells. J Biol Chem. 2010 Jan 15;285(3):1616–1626. doi: 10.1074/jbc.M109.069294
  • Juknat A, Pietr M, Kozela E, et al. Differential transcriptional profiles mediated by exposure to the cannabinoids cannabidiol and Δ9-tetrahydrocannabinol in BV-2 microglial cells. Br J Pharmacol. 2012 Apr;165(8):2512–28.
  • Juknat A, Pietr M, Kozela E, et al. Microarray and pathway analysis reveal distinct mechanisms underlying cannabinoid-mediated modulation of LPS-induced activation of BV-2 microglial cells. PloS One. 2013;8(4):e61462. doi: 10.1371/journal.pone.0061462
  • Juknat A, Gao F, Coppola G, et al. miRNA expression profiles and molecular networks in resting and LPS-activated BV-2 microglia-Effect of cannabinoids. PloS One. 2019;14(2):e0212039. doi: 10.1371/journal.pone.0212039
  • Silva-Cardoso GK, Lazarini-Lopes W, Primini EO, et al. Cannabidiol modulates chronic neuropathic pain aversion behavior by attenuation of neuroinflammation markers and neuronal activity in the corticolimbic circuit in male Wistar rats. Behav Brain Res. 2023 Aug 24;452:114588. doi: 10.1016/j.bbr.2023.114588
  • Cifelli P, Ruffolo G, De Felice E, et al. Phytocannabinoids in neurological diseases: could they restore a physiological GABAergic Transmission? Int J Mol Sci. 2020 Jan 22;21(3):723. doi: 10.3390/ijms21030723
  • Gonca E, Darıcı F. The effect of cannabidiol on ischemia/reperfusion-induced ventricular arrhythmias: the role of adenosine A1 receptors. J Cardiovasc Pharmacol Ther. 2015 Jan;20(1):76–83. doi: 10.1177/1074248414532013
  • Ribeiro A, Ferraz-de-Paula V, Pinheiro ML, et al. Cannabidiol, a non-psychotropic plant-derived cannabinoid, decreases inflammation in a murine model of acute lung injury: role for the adenosine A(2A) receptor. Eur J Pharmacol. 2012 Mar 5;678(1–3):78–85. doi: 10.1016/j.ejphar.2011.12.043
  • Campos AC, Guimarães FS. Involvement of 5HT1A receptors in the anxiolytic-like effects of cannabidiol injected into the dorsolateral periaqueductal gray of rats. Psychopharmacol (Berl). 2008 Aug;199(2):223–30. doi: 10.1007/s00213-008-1168-x
  • Hind WH, England TJ, O’Sullivan SE. Cannabidiol protects an in vitro model of the blood-brain barrier from oxygen-glucose deprivation via PPARγ and 5-HT1A receptors. Br J Pharmacol. 2016 Mar;173(5):815–825. doi: 10.1111/bph.13368
  • Iannotti FA, Hill CL, Leo A, et al. Nonpsychotropic plant cannabinoids, cannabidivarin (CBDV) and cannabidiol (CBD), activate and desensitize transient receptor potential vanilloid 1 (TRPV1) channels in vitro: potential for the treatment of neuronal hyperexcitability. ACS Chem Neurosci. 2014 Nov 19;5(11):1131–41. doi: 10.1021/cn5000524
  • Ryberg E, Larsson N, Sjögren S, et al. The orphan receptor GPR55 is a novel cannabinoid receptor. Br J Pharmacol. 2007 Dec;152(7):1092–101.
  • Huang J, Fan X, Jin X, et al. Cannabidiol inhibits Na(v) channels through two distinct binding sites. Nat Commun. 2023 Jun 17;14(1):3613. doi: 10.1038/s41467-023-39307-6
  • Keller R, Basta R, Salerno L, et al. Autism, epilepsy, and synaptopathies: a not rare association. Neurol Sci. 2017 Aug;38(8):1353–1361. doi: 10.1007/s10072-017-2974-x
  • Vargas DL, Nascimbene C, Krishnan C, et al. Neuroglial activation and neuroinflammation in the brain of patients with autism. Ann Neurol. 2005 Jan;57(1):67–81.
  • Morgan JT, Chana G, Pardo CA, et al. Microglial activation and increased microglial density observed in the dorsolateral prefrontal cortex in autism. Biol Psychiatry. 2010 Aug 15;68(4):368–76. doi: 10.1016/j.biopsych.2010.05.024
  • Suzuki K, Sugihara G, Ouchi Y, et al. Microglial activation in young adults with autism spectrum disorder. JAMA Psychiatry. 2013 Jan;70(1):49–58.
  • Salter MW, Beggs S. Sublime microglia: expanding roles for the guardians of the CNS. Cell. 2014 Jul 3;158(1):15–24. doi: 10.1016/j.cell.2014.06.008
  • Schafer DP, Heller CT, Gunner G, et al. Microglia contribute to circuit defects in Mecp2 null mice independent of microglia-specific loss of Mecp2 expression. Elife. 2016 Jul 26;5. doi: 10.7554/eLife.15224
  • Matcovitch-Natan O, Winter DR, Giladi A, et al. Microglia development follows a stepwise program to regulate brain homeostasis. Science. 2016 Aug 19;353(6301):aad8670. doi: 10.1126/science.aad8670
  • Salter MW, Stevens B. Microglia emerge as central players in brain disease. Nat Med. 2017 Sep 8;23(9):1018–1027. doi: 10.1038/nm.4397
  • Zhang L, Lin C, Zhu J, et al. Restoring prefrontal cortical excitation-inhibition balance with cannabidiol ameliorates neurobehavioral abnormalities in a mouse model of neurodevelopmental disorders. Neuropharmacology. 2023 Dec 1;240:109715. doi: 10.1016/j.neuropharm.2023.109715
  • Salzman C, Kochansky GE, Van Der Kolk BA, et al. The effect of marijuana on small group process. Am J Drug Alcohol Abuse. 1977;4(2):251–255. doi: 10.3109/00952997709002763
  • Salzman CV, Shader RI. Marijuana and hostility in a small-group setting. Am J Psychiatry. 1976;133(9):1029–1033.
  • Kamal BS, Kamal F, Lantela DE. Cannabis and the anxiety of fragmentation-a systems approach for finding an anxiolytic cannabis chemotype. Front Neurosci. 2018;12:730. doi: 10.3389/fnins.2018.00730
  • Ferber SG, Namdar D, Hen-Shoval D, et al. The “entourage effect”: terpenes coupled with Cannabinoids for the treatment of mood disorders and anxiety disorders. Curr Neuropharmacol. 2020;18(2):87–96. doi: 10.2174/1570159X17666190903103923
  • Staben J, Koch M, Reid K, et al. Cannabidiol and cannabis-inspired terpene blends have acute prosocial effects in the BTBR mouse model of autism spectrum disorder. Front Neurosci. 2023;17:1185737. doi: 10.3389/fnins.2023.1185737
  • Savino R, Medoro A, Ali S, et al. The emerging role of flavonoids in autism spectrum disorder: a systematic review. J Clin Med. 2023 May 17;12(10):3520. doi: 10.3390/jcm12103520
  • Russo EB. The case for the entourage effect and conventional breeding of clinical cannabis: no “strain,” no gain. Front Plant Sci. 2018;9:1969. doi: 10.3389/fpls.2018.01969
  • Aran A, Harel M, Cassuto H, et al. Cannabinoid treatment for autism: a proof-of-concept randomized trial. Mol Autism. 2021 Feb 3;12(1):6. doi: 10.1186/s13229-021-00420-2
  • Silva EAD Jr, Medeiros WMB, Santos JPMD, et al. Evaluation of the efficacy and safety of cannabidiol-rich cannabis extract in children with autism spectrum disorder: randomized, double-blind and controlled placebo clinical trial.Trends Psychiatry Psychother. 2022 May 26 ;44. Epub ahead of print. PMID: 35617670.
  • Bearss K, Johnson C, Smith T, et al. Effect of parent training vs parent education on behavioral problems in children with autism spectrum disorder: a randomized clinical trial. JAMA. 2015 Apr 21;313(15):1524–33. doi: 10.1001/jama.2015.3150
  • Berry-Kravis E, Hagerman R, Budimirovic D, et al. A randomized, controlled trial of ZYN002 cannabidiol transdermal gel in children and adolescents with fragile X syndrome (CONNECT-FX). J Neurodev Disord. 2022 Nov 25;14(1):56. doi: 10.1186/s11689-022-09466-6
  • Pietropaolo S, Marsicano G. The role of the endocannabinoid system as a therapeutic target for autism spectrum disorder: Lessons from behavioral studies on mouse models. Neurosci Biobehav Rev. 2022 Jan;132: 664–678. doi: 10.1016/j.neubiorev.2021.11.031
  • Cheroni C, Caporale N, Testa G. Autism spectrum disorder at the crossroad between genes and environment: contributions, convergences, and interactions in ASD developmental pathophysiology. Mol Autism. 2020 Sep 10;11(1):69. doi: 10.1186/s13229-020-00370-1
  • Aran A, Harel M, Ovadia A, et al. Mediators of placebo response to cannabinoid treatment in children with autism spectrum disorder. J Clin Med. 2023 Apr 24;12(9):3098. doi: 10.3390/jcm12093098

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.