Acessibilidade / Reportar erro

The endocannabinoid system in social anxiety disorder: from pathophysiology to novel therapeutics

Abstract

Social anxiety disorder (SAD) is a highly prevalent psychiatric disorder that presents with an early age of onset, chronic disease course, and increased risk of psychiatric comorbidity. Current treatment options for SAD are associated with low response rates, suboptimal efficacy, and possible risk of adverse effects. Investigation of new neurobiological mechanisms may aid in the identification of more specific therapeutic targets for the treatment of this disorder. Emerging evidence suggests that the endogenous cannabinoid system, also referred to as the endocannabinoid system (ECS), could play a potential role in the pathophysiology of SAD. This review discusses the known pathophysiological mechanisms of SAD, the potential role of the ECS in this disorder, current drugs targeting the ECS, and the potential of these novel compounds to enhance the therapeutic armamentarium for SAD. Further investigational efforts, specifically in human populations, are warranted to improve our knowledge of the ECS in SAD.

Social phobia; endocannabinoids; drug therapy; neurosciences; psychiatry


Introduction

Social anxiety disorder (SAD) is characterized by an excessive fear or anxiety of social situations wherein affected individuals worry they may behave in a manner that could lead to embarrassment, humiliation, or rejection by others.11. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5). Arlington: American Psychiatric Publishing; 2013. These fears may be restricted to performance-like situations, such as public speaking, but may also include more widespread social interactions, such as initiating a conversation or socializing at a gathering. As such, affected individuals typically avoid anxiety-provoking social stimuli.11. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5). Arlington: American Psychiatric Publishing; 2013. This enduring anxiety and avoidance often results in clinically significant psychosocial impairment, which interferes with the person’s daily routines, social engagements, relationships, occupation, and/or academic functioning.22. Leichsenring F, Leweke F. Social anxiety disorder. N Engl J Med. 2017;376:2255-64.,33. Schneier FR, Johnson J, Hornig CD, Liebowitz MR, Weissman MM. Social phobia. Comorbidity and morbidity in an epidemiologic sample. Arch Gen Psychiatry. 1992;49:282-8.

SAD is a highly prevalent, predominately youth-onset disorder which may affect 12.1% of individuals in the general population at least once in their lifetime.33. Schneier FR, Johnson J, Hornig CD, Liebowitz MR, Weissman MM. Social phobia. Comorbidity and morbidity in an epidemiologic sample. Arch Gen Psychiatry. 1992;49:282-8.,44. Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, Walters EE. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62:593-602. This disorder has a higher prevalence among females, often follows a chronic course, and has increased comorbidity with other psychiatric disorders.33. Schneier FR, Johnson J, Hornig CD, Liebowitz MR, Weissman MM. Social phobia. Comorbidity and morbidity in an epidemiologic sample. Arch Gen Psychiatry. 1992;49:282-8. Common coexisting health conditions include major depressive disorder, generalized anxiety disorder, agoraphobia, substance use disorders, and increased rates of suicidal ideation, as well as physical health concerns such as cardiovascular disease.33. Schneier FR, Johnson J, Hornig CD, Liebowitz MR, Weissman MM. Social phobia. Comorbidity and morbidity in an epidemiologic sample. Arch Gen Psychiatry. 1992;49:282-8.,55. Koyuncu A, İnce E, Ertekin E, Tükel R. Comorbidity in social anxiety disorder: diagnostic and therapeutic challenges. Drugs Context. 2019;8:212573. Individuals with SAD report higher unemployment rates, lower income, and greater financial dependency compared to those without SAD.33. Schneier FR, Johnson J, Hornig CD, Liebowitz MR, Weissman MM. Social phobia. Comorbidity and morbidity in an epidemiologic sample. Arch Gen Psychiatry. 1992;49:282-8.,66. Ruscio AM, Brown TA, Chiu WT, Sareen J, Stein MB, Kessler RC. Social fears and social phobia in the USA: results from the National Comorbidity Survey Replication. Psychol Med. 2008;38:15-28. Despite the significant socioeconomic costs and reduced quality of life associated with the disorder, only 35% of respondents with lifelong SAD reported seeking treatment specifically for social anxiety.66. Ruscio AM, Brown TA, Chiu WT, Sareen J, Stein MB, Kessler RC. Social fears and social phobia in the USA: results from the National Comorbidity Survey Replication. Psychol Med. 2008;38:15-28.

In terms of treatment, cognitive behavioral therapy (CBT) serves as a first-line psychotherapy approach for anxiety disorders.22. Leichsenring F, Leweke F. Social anxiety disorder. N Engl J Med. 2017;376:2255-64. A 2018 meta-analysis reported a pre-post CBT effect size of d = 1.37, compared to effect sizes of d = 1.33 for other psychotherapies and d = 0.88 for psychological or pill placebo.77. Bandelow B, Sagebiel A, Belz M, Görlich Y, Michaelis S, Wedekind D. Enduring effects of psychological treatments for anxiety disorders: meta-analysis of follow-up studies. Br J Psychiatry. 2018;212:333-8. Pharmacological treatment with selective serotonin reuptake inhibitors (SSRIs) and serotonin-noradrenaline reuptake inhibitors (SNRIs) has also been demonstrated to yield clinical improvement in SAD patients.88. Williams T, Hattingh CJ, Kariuki CM, Tromp SA, van Balkom AJ, Ipser JC, et al. Pharmacotherapy for social anxiety disorder (SAnD). Cochrane Database Syst Rev. 2017;10;CD001206.,99. Jakubovski E, Johnson JA, Nasir M, Müller-Vahl K, Bloch MH. Systematic review and meta-analysis: dose-response curve of SSRIs and SNRIs in anxiety disorders. Depress Anxiety. 2019;36:198-212. A 2019 meta-analysis examining 23 randomized-controlled SAD studies reported a standardized mean difference of 0.66 and 0.67 with respect to the measured treatment benefit of SSRIs and SNRIs compared to placebo.99. Jakubovski E, Johnson JA, Nasir M, Müller-Vahl K, Bloch MH. Systematic review and meta-analysis: dose-response curve of SSRIs and SNRIs in anxiety disorders. Depress Anxiety. 2019;36:198-212. A moderate response has been described for treatment with benzodiazepines, although these drugs have been associated with multiple adverse effects, including physiological dependence, withdrawal symptoms, and impaired cognition when used over longer periods of time.88. Williams T, Hattingh CJ, Kariuki CM, Tromp SA, van Balkom AJ, Ipser JC, et al. Pharmacotherapy for social anxiety disorder (SAnD). Cochrane Database Syst Rev. 2017;10;CD001206.,1010. Bystritsky A, Khalsa SS, Cameron ME, Schiffman J. Current diagnosis and treatment of anxiety disorders. P T. 2013;38:30-57. Anticonvulsants (pregabalin and gabapentin), monoamine oxidase inhibitors (MAOIs), reversible inhibitors of monoamine oxidase A, and tricyclic antidepressants (TCAs) have also demonstrated efficacy in the treatment of SAD.88. Williams T, Hattingh CJ, Kariuki CM, Tromp SA, van Balkom AJ, Ipser JC, et al. Pharmacotherapy for social anxiety disorder (SAnD). Cochrane Database Syst Rev. 2017;10;CD001206. MAOIs and TCAs carry a higher risk of severe adverse effects and interactions (for MAOIs, e.g., hypertension, serotonin syndrome, potentially dangerous interactions with other antidepressants or food requiring specific dietary restrictions; for TCAs, e.g., anticholinergic effects, cardiac adverse effects) compared to newer antidepressants such as SSRIs and SNRIs.88. Williams T, Hattingh CJ, Kariuki CM, Tromp SA, van Balkom AJ, Ipser JC, et al. Pharmacotherapy for social anxiety disorder (SAnD). Cochrane Database Syst Rev. 2017;10;CD001206.

Overall, current therapeutic options demonstrate suboptimal response rates, limited efficacy, and include risk for potentially severe side effects with certain medications, such as benzodiazepines or MAOIs. The relative paucity of effective treatments for this disorder may be partly due to our limited understanding of the pathophysiology of SAD, despite research efforts implicating several biological systems. Investigation of new neurobiological mechanisms may facilitate the discovery of more specific therapeutic targets for SAD and could aid in the future identification of biomarkers. One notable target includes the endogenous cannabinoid system (ECS), which emerging evidence suggests may mediate aspects of anxiety and social behavior.1111. Morena M, Patel S, Bains JS, Hill MN. Neurobiological interactions between stress and the endocannabinoid system. Neuropsychopharmacology. 2016;41:80-102.,1212. Wei D, Allsop S, Tye K, Piomelli D. Endocannabinoid signaling in the control of social behavior. Trends Neurosci. 2017;40:385-96.

This article aims to provide a narrative summary of the pathophysiological mechanisms involved in SAD, give an overview of the potential pathophysiological role of the ECS in SAD, and discuss future research directions and potential therapeutic agents for SAD targeting the ECS.

Known pathophysiological mechanisms of SAD

Behavioral inhibition and neurocircuitry in SAD

Behavioral inhibition (BI), described as a childhood tendency to withdraw from unfamiliar situations, people and environments,1313. Kagan J, Reznick JS, Clarke C, Snidman N, Garcia-Coll C. Behavioral inhibition to the unfamiliar. Child Dev. 1984;55:2212-25. has been associated with the development of SAD across several prospective studies.1414. Schwartz CE, Snidman N, Kagan J. Adolescent social anxiety as an outcome of inhibited temperament in childhood. J Am Acad Child Adolesc Psychiatry. 1999;38:1008-15.-15. Prior M, Smart D, Sanson A, Oberklaid F. Does shy-inhibited temperament in childhood lead to anxiety problems in adolescence? J Am Acad Child Adolesc Psychiatry. 2000;39:461-8. 16. Biederman J, Hirshfeld-Becker DR, Rosenbaum JF, Hérot C, Friedman D, Snidman N, et al. Further evidence of association between behavioral inhibition and social anxiety in children. Am J Psychiatry. 2001;158:1673-9. 1717. Hirshfeld-Becker DR, Biederman J, Henin A, Faraone SV, Davis S, Harrington K, et al. Behavioral inhibition in preschool children at risk is a specific predictor of middle childhood social anxiety: a five-year follow-up. J Dev Behav Pediatr. 2007;28:225-33. A 2020 meta-analysis found that behaviorally inhibited children had significantly increased odds of developing SAD (odds ratio = 5.84), indicating that childhood BI may be a risk factor/predictor of the disorder.1818. Sandstrom A, Uher R, Pavlova B. Prospective association between childhood behavioral inhibition and anxiety: a meta-analysis. J Abnorm Child Psychol. 2020;48:57-66.,1919. Clauss JA, Blackford JU. Behavioral inhibition and risk for developing social anxiety disorder: a meta-analytic study. J Am Acad Child Adolesc Psychiatry. 2012;51:1066-75.e1. One hypothesis suggests that BI is associated with either a lower threshold for or a stronger response to the detection of novel, salient, or threatening information, thereby implicating disruptions in certain neural circuitry.2020. Fox NA, Buzzell GA, Morales S, Valadez EA, Wilson M, Henderson HA. Understanding the emergence of social anxiety in children with behavioral inhibition. Biol Psychiatry. 2021;89:681-9. This is supported by studies which found that individuals with a childhood history of BI exhibit a greater striatal response to reward2121. Bar-Haim Y, Fox NA, Benson B, Guyer AE, Williams A, Nelson EE, et al. Neural correlates of reward processing in adolescents with a history of inhibited temperament. Psychol Sci. 2009;20:1009-18.-22. Guyer AE, Nelson EE, Perez-Edgar K, Hardin MG, Roberson-Nay R, Monk CS, et al. Striatal functional alteration in adolescents characterized by early childhood behavioral inhibition. J Neurosci. 2006;26:6399-405. 2323. Guyer AE, Benson B, Choate VR, Bar-Haim Y, Perez-Edgar K, Jarcho JM, et al. Lasting associations between early-childhood temperament and late-adolescent reward-circuitry response to peer feedback. Dev Psychopathol. 2014;26:229-43. and punishment2424. Helfinstein SM, Benson B, Perez-Edgar K, Bar-Haim Y, Detloff A, Pine DS, et al. Striatal responses to negative monetary outcomes differ between temperamentally inhibited and non-inhibited adolescents. Neuropsychologia. 2011;49:479-85. cues, as well as heightened activity in regions including the prefrontal cortex (PFC)2424. Helfinstein SM, Benson B, Perez-Edgar K, Bar-Haim Y, Detloff A, Pine DS, et al. Striatal responses to negative monetary outcomes differ between temperamentally inhibited and non-inhibited adolescents. Neuropsychologia. 2011;49:479-85.,2525. Jarcho JM, Fox NA, Pine DS, Etkin A, Leibenluft E, Shechner T, et al. The neural correlates of emotion-based cognitive control in adults with early childhood behavioral inhibition. Biol Psychol. 2013;92:306-14. and anterior cingulate cortex (ACC).2626. Lamm C, Walker OL, Degnan KA, Henderson HA, Pine DS, McDermott JM, et al. Cognitive control moderates early childhood temperament in predicting social behavior in 7‐year‐old children: an ERP study. Dev Sci. 2014;17:667-81. Greater amygdala reactivity to novel stimuli has also been reported in adolescents and adults who exhibited BI during childhood.2727. Pérez-Edgar K, Roberson-Nay R, Hardin MG, Poeth K, Guyer AE, Nelson EE, et al. Attention alters neural responses to evocative faces in behaviorally inhibited adolescents. Neuroimage. 2007;35:1538-46.-28. Schwartz CE, Wright CI, Shin LM, Kagan J, Rauch SL. Inhibited and uninhibited infants “grown up”: adult amygdalar response to novelty. Science. 2003;300:1952-3. 2929. Schwartz CE, Kunwar PS, Greve DN, Kagan J, Snidman NC, Bloch RB. A phenotype of early infancy predicts reactivity of the amygdala in male adults. Mol Psychiatry. 2012;17:1042-50.

These findings fall in line with those of functional imaging studies in persons with SAD. Results from such studies consistently demonstrate abnormal activity in regions including the amygdala, insula, PFC, and ACC, dubbed the “corticolimbic circuit.”3030. Kovner R, Oler JA, Kalin NH. Cortico-limbic interactions mediate adaptive and maladaptive responses relevant to psychopathology. Am J Psychiatry. 2019;176:987-99. Notably, heightened amygdalar and insular activities have been observed in response to performing stressful social tasks3131. Tillfors M, Furmark T, Marteinsdottir I, Fredrikson M. Cerebral blood flow during anticipation of public speaking in social phobia: a PET study. Biol Psychiatry. 2002;52:1113-9.-32. Lorberbaum JP, Kose S, Johnson MR, Arana GW, Sullivan LK, Hamner MB, et al. Neural correlates of speech anticipatory anxiety in generalized social phobia. Neuroreport. 2004;15:2701-5. 33. Guyer AE, Lau JY, McClure-Tone EB, Parrish J, Shiffrin ND, Reynolds RC, et al. Amygdala and ventrolateral prefrontal cortex function during anticipated peer evaluation in pediatric social anxiety. Arch Gen Psychiatry. 2008;65:1303-12. 3434. Blair K, Geraci M, Devido J, McCaffrey D, Chen G, Vythilingam M, et al. Neural response to self-and other referential praise and criticism in generalized social phobia. Arch Gen psychiatry. 2008;65:1176-84. or viewing negative facial expressions,3333. Guyer AE, Lau JY, McClure-Tone EB, Parrish J, Shiffrin ND, Reynolds RC, et al. Amygdala and ventrolateral prefrontal cortex function during anticipated peer evaluation in pediatric social anxiety. Arch Gen Psychiatry. 2008;65:1303-12.-34. Blair K, Geraci M, Devido J, McCaffrey D, Chen G, Vythilingam M, et al. Neural response to self-and other referential praise and criticism in generalized social phobia. Arch Gen psychiatry. 2008;65:1176-84. 35. Stein MB, Goldin PR, Sareen J, Zorrilla LT, Brown GG. Increased amygdala activation to angry and contemptuous faces in generalized social phobia. Arch Gen Psychiatry. 2002;59:1027-34. 3636. Yoon KL, Fitzgerald DA, Angstadt M, McCarron RA, Phan KL. Amygdala reactivity to emotional faces at high and low intensity in generalized social phobia: a 4-Tesla functional MRI study. Psychiatry Res. 2007;154:93-8. which in some studies has also correlated positively with the severity of SAD symptoms.3737. Phan KL, Fitzgerald DA, Nathan PJ, Tancer ME. Association between amygdala hyperactivity to harsh faces and severity of social anxiety in generalized social phobia. Biol Psychiatry. 2006;59:424-9.-38. Evans KC, Wright CI, Wedig MM, Gold AL, Pollack MH, Rauch SL. A functional MRI study of amygdala responses to angry schematic faces in social anxiety disorder. Depress Anxiety. 2008;25:496-505. 3939. Shah SG, Klumpp H, Angstadt M, Nathan PJ, Phan KL. Amygdala and insula response to emotional images in patients with generalized social anxiety disorder. J Psychiatry Neurosci. 2009;34:296-302. Although less consistent in direction, abnormal activity has also been reported in the PFC and ACC of subjects with SAD compared to controls.3232. Lorberbaum JP, Kose S, Johnson MR, Arana GW, Sullivan LK, Hamner MB, et al. Neural correlates of speech anticipatory anxiety in generalized social phobia. Neuroreport. 2004;15:2701-5.

33. Guyer AE, Lau JY, McClure-Tone EB, Parrish J, Shiffrin ND, Reynolds RC, et al. Amygdala and ventrolateral prefrontal cortex function during anticipated peer evaluation in pediatric social anxiety. Arch Gen Psychiatry. 2008;65:1303-12.
-3434. Blair K, Geraci M, Devido J, McCaffrey D, Chen G, Vythilingam M, et al. Neural response to self-and other referential praise and criticism in generalized social phobia. Arch Gen psychiatry. 2008;65:1176-84.,4040. Amir N, Klumpp H, Elias J, Bedwell JS, Yanasak N, Miller LS. Increased activation of the anterior cingulate cortex during processing of disgust faces in individuals with social phobia. Biol Psychiatry. 2005;57:975-81. Collectively, these findings suggest that the neurobiology of childhood BI may be associated with corticolimbic disruptions contributing to the development of SAD.

Neurotransmitter systems in SAD

The monoamine hypothesis and pharmacological approaches suggest that the neurobiologies of depression and anxiety share imbalances in the monoaminergic neurotransmission system.4141. Schildkraut JJ. The catecholamine hypothesis of affective disorders: a review of supporting evidence. Am J Psychiatry. 1965;122:509-22. In this regard, neuro-molecular positron emission tomography (PET) and single photon emission computed tomography (SPECT) studies in SAD have largely focused on imaging serotonergic and dopaminergic neurotransmission, based on the reported efficacy of antidepressants.4141. Schildkraut JJ. The catecholamine hypothesis of affective disorders: a review of supporting evidence. Am J Psychiatry. 1965;122:509-22.,4242. Doruyter AG, Dupont P, Stein DJ, Lochner C, Warwick JM. Nuclear neuroimaging in social anxiety disorder: a review. J Nucl Med. 2018;59:1794-800.

Two PET studies examining presynaptic serotoninergic activity found elevated rates of serotonin synthesis in the hippocampus, basal ganglia, amygdala, and ACC of individuals with SAD.4343. Furmark T, Marteinsdottir I, Frick A, Heurling K, Tillfors M, Appel L, et al. Serotonin synthesis rate and the tryptophan hydroxylase-2: G-703T polymorphism in social anxiety disorder. J Psychopharmacol. 2016;30:1028-35.,4444. Frick A, Åhs F, Engman J, Jonasson M, Alaie I, Björkstrand J, et al. Serotonin synthesis and reuptake in social anxiety disorder: a positron emission tomography study. JAMA Psychiatry. 2015;72:794-802. Additionally, three studies employing two different tracers ([C-11]DASB PET4444. Frick A, Åhs F, Engman J, Jonasson M, Alaie I, Björkstrand J, et al. Serotonin synthesis and reuptake in social anxiety disorder: a positron emission tomography study. JAMA Psychiatry. 2015;72:794-802.,4545. Hjorth OR, Frick A, Gingnell M, Hoppe JM, Faria V, Hultberg S, et al. Expression and co-expression of serotonin and dopamine transporters in social anxiety disorder: a multitracer positron emission tomography study. Mol Psychiatry. 2019 Dec 10. doi: 10.1038/s41380-019-0618-7. Online ahead of print.
10.1038/s41380-019-0618-7...
and [I-123]-β-CIT SPECT4646. van der Wee NJ, van Veen JF, Stevens H, van Vliet IM, van Rijk PP, Westenberg HG. Increased serotonin and dopamine transporter binding in psychotropic medication-naive patients with generalized social anxiety disorder shown by 123I-β-(4-iodophenyl)-tropane SPECT. J Nucl Med. 2008;49:757-63.) reported higher serotonin receptor binding potential (a measure of receptor expression) within the raphe nuclei,4444. Frick A, Åhs F, Engman J, Jonasson M, Alaie I, Björkstrand J, et al. Serotonin synthesis and reuptake in social anxiety disorder: a positron emission tomography study. JAMA Psychiatry. 2015;72:794-802. caudate nucleus,4444. Frick A, Åhs F, Engman J, Jonasson M, Alaie I, Björkstrand J, et al. Serotonin synthesis and reuptake in social anxiety disorder: a positron emission tomography study. JAMA Psychiatry. 2015;72:794-802. insular cortex,4444. Frick A, Åhs F, Engman J, Jonasson M, Alaie I, Björkstrand J, et al. Serotonin synthesis and reuptake in social anxiety disorder: a positron emission tomography study. JAMA Psychiatry. 2015;72:794-802. nucleus accumbens,4545. Hjorth OR, Frick A, Gingnell M, Hoppe JM, Faria V, Hultberg S, et al. Expression and co-expression of serotonin and dopamine transporters in social anxiety disorder: a multitracer positron emission tomography study. Mol Psychiatry. 2019 Dec 10. doi: 10.1038/s41380-019-0618-7. Online ahead of print.
10.1038/s41380-019-0618-7...
and thalamus.4444. Frick A, Åhs F, Engman J, Jonasson M, Alaie I, Björkstrand J, et al. Serotonin synthesis and reuptake in social anxiety disorder: a positron emission tomography study. JAMA Psychiatry. 2015;72:794-802.,4646. van der Wee NJ, van Veen JF, Stevens H, van Vliet IM, van Rijk PP, Westenberg HG. Increased serotonin and dopamine transporter binding in psychotropic medication-naive patients with generalized social anxiety disorder shown by 123I-β-(4-iodophenyl)-tropane SPECT. J Nucl Med. 2008;49:757-63. Only one study investigated postsynaptic serotonergic function in SAD, in which PET imaging of [carbonyl-C-11]WAY-100635 demonstrated significantly reduced serotonin 1A receptor binding potential in the amygdala, insula, and ACC of patients with SAD.4747. Lanzenberger RR, Mitterhauser M, Spindelegger C, Wadsak W, Klein N, Mien LK, et al. Reduced serotonin-1A receptor binding in social anxiety disorder. Biol Psychiatry. 2007;61:1081-9.

Imaging of dopaminergic systems has produced less consistent results. Three studies employing [I-123]-β-CIT SPECT reported either an increase,4646. van der Wee NJ, van Veen JF, Stevens H, van Vliet IM, van Rijk PP, Westenberg HG. Increased serotonin and dopamine transporter binding in psychotropic medication-naive patients with generalized social anxiety disorder shown by 123I-β-(4-iodophenyl)-tropane SPECT. J Nucl Med. 2008;49:757-63. decrease,4848. Tiihonen J, Kuikka J, Bergstrom K, Lepola U, Koponen H, Leinonen E. Dopamine reuptake site densities in patients with social phobia. Am J Psychiatry. 1997;154:239-42. or no difference4949. Schneier FR, Abi‐Dargham A, Martinez D, Slifstein M, Hwang DR, Liebowitz MR, et al. Dopamine transporters, D2 receptors, and dopamine release in generalized social anxiety disorder. Depress Anxiety. 2009;26:411-8. in dopamine transporter binding in the striatum. As suggested by Schneier et al.,4949. Schneier FR, Abi‐Dargham A, Martinez D, Slifstein M, Hwang DR, Liebowitz MR, et al. Dopamine transporters, D2 receptors, and dopamine release in generalized social anxiety disorder. Depress Anxiety. 2009;26:411-8. these discrepancies could possibly be due to small sample sizes (n=12) or differences in SPECT assessment methods (receptor blocking compound, identification of volumes of interest or reference regions). Another SPECT study reported significantly reduced dopamine D2/D3 receptor binding potential in the striatum of patients with SAD and comorbid obsessive-compulsive disorder compared to healthy controls.5050. Schneier FR, Liebowitz MR, Abi-Dargham A, Zea-Ponce Y, Lin SH, Laruelle M. Low dopamine D2 receptor binding potential in social phobia. Am J Psychiatry. 2000;157:457-9. However, the same group found no difference in striatal dopamine D2/D3 receptor binding in a subsequent PET study, which employed a more reliable dopamine measurement technique and used a larger SAD sample size (n=17).4949. Schneier FR, Abi‐Dargham A, Martinez D, Slifstein M, Hwang DR, Liebowitz MR, et al. Dopamine transporters, D2 receptors, and dopamine release in generalized social anxiety disorder. Depress Anxiety. 2009;26:411-8. The latter study also reported a decrease in striatal dopamine D2/D3 receptor binding following an amphetamine challenge. Some of the observed differences may be due to variations in age range, symptom severity, treatments, and disease duration of the study cohorts, as well as varying strengths and limitations of the different imaging techniques and tracers employed in the aforementioned studies. While some of these findings suggest that serotonergic and dopaminergic activity may play a role in SAD, the limited and inconsistent results warrant further investigation to better understand the role of these neurotransmitter systems in SAD pathophysiology.

The hypothalamic-pituitary-adrenal axis in SAD

It has been well established that the hypothalamic-pituitary-adrenal (HPA) axis plays a major role in stress regulation.5151. Faravelli C, Sauroc CL, Lelli L, Pietrini F, Lazzeretti L, Godini L, et al. The role of life events and HPA axis in anxiety disorders: a review. Curr Pharm Des. 2012;18:5663-74.,5252. Hill MN, Tasker JG. Endocannabinoid signaling, glucocorticoid-mediated negative feedback, and regulation of the hypothalamic-pituitary-adrenal axis. Neuroscience. 2012;204:5-16. Activation of the HPA axis leads to increased corticotropin-releasing-hormone (CRH) signaling in the limbic forebrain, which, in turn, stimulates the downstream release of cortisol.5252. Hill MN, Tasker JG. Endocannabinoid signaling, glucocorticoid-mediated negative feedback, and regulation of the hypothalamic-pituitary-adrenal axis. Neuroscience. 2012;204:5-16. While acute stress leads to adaptive activation of the HPA axis with a transient increase in cortisol,5353. Soravia LM, Heinrichs M, Aerni A, Maroni C, Schelling G, Ehlert U, et al. Glucocorticoids reduce phobic fear in humans. Proc Natl Acad Sci U S A. 2006;103:5585-90. chronic stress can result in prolonged activation of the HPA axis via dysregulation of glucocorticoid-mediated feedback inhibition.5454. Radley J, Morilak D, Viau V, Campeau S. Chronic stress and brain plastistate: mechanisms underlying adaptive and maladaptive changes and implications for stress-related CNS disorders. Neurosci Biobehav Rev. 2015;58:79-91. These disruptive processes may affect the coping mechanisms established by CRH systems, leading to chronic symptoms of fear and anxiety.5151. Faravelli C, Sauroc CL, Lelli L, Pietrini F, Lazzeretti L, Godini L, et al. The role of life events and HPA axis in anxiety disorders: a review. Curr Pharm Des. 2012;18:5663-74.,5454. Radley J, Morilak D, Viau V, Campeau S. Chronic stress and brain plastistate: mechanisms underlying adaptive and maladaptive changes and implications for stress-related CNS disorders. Neurosci Biobehav Rev. 2015;58:79-91.

Previous studies have demonstrated that individuals with SAD had significantly elevated social stressor-induced cortisol levels compared to controls.5555. Condren RM, O’Neill A, Ryan MC, Barrett P, Thakore JH. HPA axis response to a psychological stressor in generalised social phobia. Psychoneuroendocrinology. 2002;27:693-703.-56. van West D, Claes S, Sulon J, Deboutte D. Hypothalamic-pituitary-adrenal reactivity in prepubertal children with social phobia. J Affect Disord. 2008;111:281-90. 5757. Yoon KL, Joormann J. Stress reactivity in social anxiety disorder with and without comorbid depression. J Abnorm Psychol. 2012;121:250-5. However, these findings are not consistent: other studies employing similar stress paradigms found no significant difference in cortisol levels.5858. Martel FL, Hayward C, Lyons DM, Sanborn K, Varady S, Schatzberg AF. Salivary cortisol levels in socially phobic adolescent girls. Depress Anxiety. 1999;10:25-7.-59. Klumbies E, Braeuer D, Hoyer J, Kirschbaum C. The reaction to social stress in social phobia: discordance between physiological and subjective parameters. PLoS One. 2014;9:e105670. 6060. Losiak W, Blaut A, Klosowska J, Slowik N. Social anxiety, affect, cortisol response and performance on a speech task. Psychopathology. 2016;49:24-30. These discrepancies may be due to variable length of stressor exposure, perception of risk posed by the stressor, comorbidities, or age, which are known to influence cortisol response.5757. Yoon KL, Joormann J. Stress reactivity in social anxiety disorder with and without comorbid depression. J Abnorm Psychol. 2012;121:250-5.,6161. Rose RM. Endocrine responses to stressful psychological events. Psychiatr Clin North Am. 1980;3:251-76. Interestingly, emerging evidence suggests that the ECS may contribute to the regulation of HPA axis activity.5252. Hill MN, Tasker JG. Endocannabinoid signaling, glucocorticoid-mediated negative feedback, and regulation of the hypothalamic-pituitary-adrenal axis. Neuroscience. 2012;204:5-16. As such, improved understanding of ECS processes may provide insight into the possible aberrations in the HPA axis stress response involved in the pathophysiology of SAD.

The endocannabinoid system

The ECS is a lipid-based signaling system of the central and peripheral nervous system (Figure 1).6262. Pertwee RG. Ligands that target cannabinoid receptors in the brain: from THC to anandamide and beyond. Addict Biol. 2008;13:147-59. The ECS is primarily composed of two G protein-coupled cannabinoid receptors, endogenous cannabinoid ligands, and enzymes responsible for ligand synthesis and degradation. Cannabinoid type 1 (CB1) receptors6363. Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bonner TI. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature. 1990;346:561-4. are the predominant cannabinoid receptors in the central nervous system (CNS) and mainly located on terminals of central and peripheral neurons.6464. Herkenham M, Lynn AB, Johnson MR, Melvin LS, de Costa BR, Rice KC. Characterization and localization of cannabinoid receptors in rat brain: a quantitative in vitro autoradiographic study. J Neurosci. 1991;11:563-83.,6565. Galiègue S, Mary S, Marchand J, Dussossoy D, Carrière D, Carayon P, et al. Expression of central and peripheral cannabinoid receptors in human immune tissues and leukocyte subpopulations. Eur J Biochem. 1995;232:54-61. Activation of these receptors inhibits neurotransmitter release, primarily from GABAergic and glutamatergic neurons as well as certain monoaminergic sites.6262. Pertwee RG. Ligands that target cannabinoid receptors in the brain: from THC to anandamide and beyond. Addict Biol. 2008;13:147-59. In comparison, cannabinoid type 2 (CB2) receptors6666. Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a peripheral receptor for cannabinoids. Nature. 1993;365:61-5. are primarily distributed in peripheral tissues and immune system cells, where they modulate cell migration and cytokine release.6565. Galiègue S, Mary S, Marchand J, Dussossoy D, Carrière D, Carayon P, et al. Expression of central and peripheral cannabinoid receptors in human immune tissues and leukocyte subpopulations. Eur J Biochem. 1995;232:54-61.

Figure 1
Illustration of endocannabinoid biosynthesis, binding, and metabolism. A) The biosynthesis of 2-AG is mediated by sequential hydrolysis, whereby a GPL is hydrolyzed by PLC or PLD to form DAG. DAG is then rapidly hydrolyzed by DAGL to form 2-AG. B) 2-AG is primarily broken down by MAGL to form arachidonic acid and glycerol. C) AEA is synthesized from phospholipid precursors PPEA which are initially converted to NAPE by NAT. NAPE is subsequently converted to AEA by NAPE-PLD activity. D) AEA is primarily metabolized by FAAH to form arachidonic acid and ethanolamine. E) Both 2-AG and AEA bind to CB1 on the presynaptic neuron. F) After receptor activation, AEA and 2-AG are transported back into the neuron by EMT. NAPE may be converted to AEA through alternative pathways.6767. Ahn K, McKinney MK, Cravatt BF. Enzymatic pathways that regulate endocannabinoid signaling in the nervous system. Chem Rev. 2008;108:1687-707. 2-AG = 2-arachidonoylglycerol; AEA = anandamide; CB1 = cannabinoid receptor 1; DAG = diacylglycerol; DAGL = DAG lipase; EMT = endocannabinoid membrane transporters; FAAH = fatty acid amide hydrolase; GPL = glycerophospholipid; MAGL = monoacylglycerol lipase; NAPE = N-arachidonoylphosphatidylethanolamine; NAPE-PLD = NAPE phospholipase D; NAT = N-acyltransferase; PLC = phospholipase C; PLD = phospholipase D; PPEA = phosphatidylethanolamine.

The two main endocannabinoids, N-arachidonoylethanolamine (anandamide; AEA)6868. Devane WA, Hanus L, Breuer A, Pertwee RG, Stevenson LA, Griffin G, et al. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science. 1992;258:1946-9. and 2-arachidonoylglycerol (2-AG),6969. Sugiura T, Kondo S, Sukagawa A, Nakane S, Shinoda A, Itoh K, et al. 2-arachidonoylgylcerol: a possible endogenous cannabinoid receptor ligand in brain. Biochem Biophys Res Commun. 1995;215:89-97. are lipid ligands that are synthesized and released on demand from the post-synaptic cell, leading to a retrograde suppression of neurotransmitter release.6262. Pertwee RG. Ligands that target cannabinoid receptors in the brain: from THC to anandamide and beyond. Addict Biol. 2008;13:147-59.,6767. Ahn K, McKinney MK, Cravatt BF. Enzymatic pathways that regulate endocannabinoid signaling in the nervous system. Chem Rev. 2008;108:1687-707. Other lipid ligands of cannabinoid receptors include 2-arachidonylglyceryl ether, N-arachidonoyl dopamine, N-oleoyl dopamine, O-arachidonoylethanolamine, and oleamide.7070. Pertwee RG. The therapeutic potential of drugs that target cannabinoid receptors or modulate the tissue levels or actions of endocannabinoids. AAPS J. 2005;7:E625-54. Signaling is terminated via metabolization of AEA and 2-AG through their catabolic enzymes, fatty acid amide hydrolase (FAAH)7171. Cravatt BF, Giang DK, Mayfield SP, Boger DL, Lerner RA, Gilula NB. Molecular characterization of an enzyme that degrades neuromodulatory fatty-acid amides. Nature. 1996;384:83-7. and monoacylglycerol lipase (MAGL),7272. Dinh TP, Carpenter D, Leslie FM, Freund TF, Katona I, Sensi SL, et al. Brain monoglyceride lipase participating in endocannabinoid inactivation. Proc Natl Acad Sci U S A. 2002;99:10819-24. respectively.

These endocannabinoids, collectively with their receptors, biosynthetic proteins, and degradative enzymes, are referred to as the ECS (Figure 1).

Role of the endocannabinoid system in the regulation of stress and anxiety

Over the last two decades, the ECS has emerged and been recognized as a potential regulator of stress and anxiety. Cannabis consumption studies investigating the exogenous cannabinoid delta-9-tetrahydrocannabinol (THC) found that, in addition to its addictive potential,7373. Connor JP, Stjepanović D, Le Foll B, Hoch E, Budney AJ, Hall WD. Cannabis use and cannabis use disorder. Nat Rev Dis Primers. 2021;7:16. THC consumption produces biphasic physiological effects depending on dose.7474. Crippa JA, Zuardi AW, Martín‐Santos R, Bhattacharyya S, Atakan Z, McGuire P, et al. Cannabis and anxiety: a critical review of the evidence. Hum Psychopharmacol. 2009;24:515-23.-75. D’Souza DC, Perry E, MacDougall L, Ammerman Y, Cooper T, Wu YT, et al. The psychotomimetic effects of intravenous delta-9-tetrahydrocannabinol in healthy individuals: implications for psychosis. Neuropsychopharmacology. 2004;29:1558-72. 7676. Hunault CC, Böcker KB, Stellato RK, Kenemans JL, de Vries I, Meulenbelt J. Acute subjective effects after smoking joints containing up to 69 mg Δ9-tetrahydrocannabinol in recreational users: a randomized, crossover clinical trial. Psychopharmacology (Berl). 2014;231:4723-33. Notably, low-dose THC consumption has been associated with anxiolytic effects, whereas high-dose consumption has been linked to anxiogenic effects.7777. Rubino T, Sala M, Vigano D, Braida D, Castiglioni C, Limonta V, et al. Cellular mechanisms underlying the anxiolytic effect of low doses of peripheral Δ 9-tetrahydrocannabinol in rats. Neuropsychopharmacology. 2007;32:2036-45.,7878. Rubino T, Guidali C, Vigano D, Realini N, Valenti M, Massi P, et al. CB1 receptor stimulation in specific brain areas differently modulate anxiety-related behaviour. Neuropharmacology. 2008;54:151-60. Given that the psychoactive effects of THC are mediated by CB1 receptor activation,6363. Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bonner TI. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature. 1990;346:561-4.,7979. Devane WA, Dysarz FA 3rd, Johnson MR, Melvin LS, Howlett AC. Determination and characterization of a cannabinoid receptor in rat brain. Mol Pharmacol. 1988;34:605-13. it is possible that ECS signaling may serve to buffer against anxiety and stress symptoms.

Animal studies have converged to show that increasing ECS activity via pharmacological stimulation of CB1 receptors (via CB1 agonism or via elevating AEA/2-AG through inhibition of their degradative enzymes) decreases behavioral measures of rodent anxiety.8080. Micale V, Di Marzo V, Sulcova A, Wotjak CT, Drago F. Endocannabinoid system and mood disorders: priming a target for new therapies. Pharmacol Ther. 2013;138:18-37. Conversely, decreasing ECS activity via antagonism or deletion of the CB1 receptor gene elicits an anxiogenic response.8080. Micale V, Di Marzo V, Sulcova A, Wotjak CT, Drago F. Endocannabinoid system and mood disorders: priming a target for new therapies. Pharmacol Ther. 2013;138:18-37. These anxiolytic effects seem to depend on CB1 activation in key structures implicated in the fear response, particularly those comprising the corticolimbic circuit.8181. Lin HC, Mao SC, Gean PW. Effects of intra-amygdala infusion of CB1 receptor agonists on the reconsolidation of fear-potentiated startle. Learn Mem. 2006;13:316-21.-82. Campolongo P, Roozendaal B, Trezza V, Hauer D, Schelling G, McGaugh JL, et al. Endocannabinoids in the rat basolateral amygdala enhance memory consolidation and enable glucocorticoid modulation of memory. Proc Natl Acad Sci U S A. 2009;106:4888-93. 83. Marsicano G, Wotjak CT, Azad SC, Bisogno T, Rammes G, Cascio MG, et al. The endogenous cannabinoid system controls extinction of aversive memories. Nature. 2002;418:530-4. 84. Tan H, Lauzon NM, Bishop SF, Bechard MA, Laviolette SR. Integrated cannabinoid CB1 receptor transmission within the amygdala-prefrontal cortical pathway modulates neuronal plastistate and emotional memory encoding. Cereb Cortex. 2010;20:1486-96. 8585. Tan H, Lauzon NM, Bishop SF, Chi N, Bechard M, Laviolette SR. Cannabinoid transmission in the basolateral amygdala modulates fear memory formation via functional inputs to the prelimbic cortex. J Neurosci. 2011;31:5300-12. In this regard, studies have shown that increased amygdala neuron excitability in rodents exposed to stress can be corrected by FAAH inhibition.8686. Hill MN, McLaughlin RJ, Bingham B, Shrestha L, Lee TT, Gray JM, et al. Endogenous cannabinoid signaling is essential for stress adaptation. Proc Natl Acad Sci U S A. 2010;107:9406-11.,8787. Bortolato M, Mangieri RA, Fu J, Kim JH, Arguello O, Duranti A, et al. Antidepressant-like activity of the fatty acid amide hydrolase inhibitor URB597 in a rat model of chronic mild stress. Biol Psychiatry. 2007;62:1103-10. Moreover, rodent models of anxiety, modeled via exposure to stress paradigms, consistently demonstrate elevated FAAH activity and reduced AEA levels in limbic areas, further supporting the anxiolytic potential of the ECS via FAAH inhibition.8686. Hill MN, McLaughlin RJ, Bingham B, Shrestha L, Lee TT, Gray JM, et al. Endogenous cannabinoid signaling is essential for stress adaptation. Proc Natl Acad Sci U S A. 2010;107:9406-11.,8888. Rademacher DJ, Meier SE, Shi L, Ho WS, Jarrahian A, Hillard CJ. Effects of acute and repeated restraint stress on endocannabinoid content in the amygdala, ventral striatum, and medial prefrontal cortex in mice. Neuropharmacology. 2008;54:108-16.-89. Hill MN, Kumar SA, Filipski SB, Iverson M, Stuhr KL, Keith JM, et al. Disruption of fatty acid amide hydrolase activity prevents the effects of chronic stress on anxiety and amygdalar microstructure. Mol Psychiatry. 2013;18:1125-35. 90. Gray JM, Vecchiarelli HA, Morena M, Lee TT, Hermanson DJ, Kim AB, et al. Corticotropin-releasing hormone drives anandamide hydrolysis in the amygdala to promote anxiety. J Neurosci. 2015;35:3879-92. 9191. Patel S, Roelke CT, Rademacher DJ, Hillard CJ. Inhibition of restraint stress‐induced neural and behavioural activation by endogenous cannabinoid signalling. Eur J Neurosci. 2005;21:1057-69. While the status of 2-AG and MAGL in anxiety is less studied, pharmacological and genetic investigations suggest that enhanced 2-AG signaling may play an important role in reducing anxiety-like behaviors and promoting adaptation under conditions of repeated stress exposure (reviewed by Bedse et al.9292. Bedse G, Hill MN, Patel S. 2-arachidonoylglycerol modulation of anxiety and stress adaptation: from grass roots to novel therapeutics. Biol Psychiatry. 2020;88:520-30.). Several studies have also shown that systemic MAGL inhibition reduces anxiety-like behaviors under basal and highly aversive conditions,9393. Bedse G, Hartley ND, Neale E, Gaulden AD, Patrick TA, Kingsley PJ, et al. Functional redundancy between canonical endocannabinoid signaling systems in the modulation of anxiety. Biol Psychiatry. 2017;82:488-99.-94. Busquets-Garcia A, Puighermanal E, Pastor A, de la Torre R, Maldonado R, Ozaita A. Differential role of anandamide and 2-arachidonoylglycerol in memory and anxiety-like responses. Biol Psychiatry. 2011;70:479-86. 95. Sciolino NR, Zhou W, Hohmann AG. Enhancement of endocannabinoid signaling with JZL184, an inhibitor of the 2-arachidonoylglycerol hydrolyzing enzyme monoacylglycerol lipase, produces anxiolytic effects under conditions of high environmental aversiveness in rats. Pharmacol Res. 2011;64:226-34. 96. Aliczki M, Zelena D, Mikics E, Varga ZK, Pinter O, Bakos NV, et al. Monoacylglycerol lipase inhibition-induced changes in plasma corticosterone levels, anxiety and locomotor activity in male CD1 mice. Horm Behav. 2013;63:752-8. 9797. Kinsey SG, O’Neal ST, Long JZ, Cravatt BF, Lichtman AH. Inhibition of endocannabinoid catabolic enzymes elicits anxiolytic-like effects in the marble burying assay. Pharmacol Biochem Behav. 2011;98:21-7. as well as acute and chronic stress-induced anxiety-like behaviors.9292. Bedse G, Hill MN, Patel S. 2-arachidonoylglycerol modulation of anxiety and stress adaptation: from grass roots to novel therapeutics. Biol Psychiatry. 2020;88:520-30.,9393. Bedse G, Hartley ND, Neale E, Gaulden AD, Patrick TA, Kingsley PJ, et al. Functional redundancy between canonical endocannabinoid signaling systems in the modulation of anxiety. Biol Psychiatry. 2017;82:488-99.,9898. Bedse G, Bluett RJ, Patrick TA, Romness NK, Gaulden AD, Kingsley PJ, et al. Therapeutic endocannabinoid augmentation for mood and anxiety disorders: comparative profiling of FAAH, MAGL and dual inhibitors. Transl Psychiatry. 2018;8:92.,9999. Lim J, Igarashi M, Jung KM, Butini S, Campiani G, Piomelli D. Endocannabinoid modulation of predator stress-induced long-term anxiety in rats. Neuropsychopharmacology. 2016;41:1329-39.

In line with the preclinical literature, human research has shown that treatment with the CB1 inverse agonist/receptor blocker rimonabant increases symptoms of anxiety and depression in some individuals.100100. Mitchell PB, Morris MJ. Depression and anxiety with rimonabant. Lancet. 2007;370:1671-2. Due to serious psychiatric adverse effects, including suicidal ideation, rimonabant was withdrawn worldwide, in an example of the potency and potential risks associated with manipulation of the ECS. Moreover, biochemical studies in humans have found that experimental exposure to the Trier Social Stress Test (TSST) increases serum concentrations of AEA, 2-AG and the other N-acylethanolamines immediately after the stress period, both in healthy participants (compared to unstressed controls)101101. Dlugos A, Childs E, Stuhr KL, Hillard CJ, de Wit H. Acute stress increases circulating anandamide and other N-acylethanolamines in healthy humans. Neuropsychopharmacology. 2012;37:2416-27. and in those diagnosed with major depression (compared to TSST-exposed controls).102102. Hill MN, Miller GE, Carrier EJ, Gorzalka BB, Hillard CJ. Circulating endocannabinoids and N-acyl ethanolamines are differentially regulated in major depression and following exposure to social stress. Psychoneuroendocrinology. 2009;34:1257-62. In the former study, baseline anxiety ratings also correlated negatively with baseline AEA concentrations.101101. Dlugos A, Childs E, Stuhr KL, Hillard CJ, de Wit H. Acute stress increases circulating anandamide and other N-acylethanolamines in healthy humans. Neuropsychopharmacology. 2012;37:2416-27. Similarly, among individuals with PTSD, those with lower peripheral AEA levels experienced more intrusive symptoms.103103. Hill MN, Bierer LM, Makotkine I, Golier JA, Galea S, McEwen BS, et al. Reductions in circulating endocannabinoid levels in individuals with post-traumatic stress disorder following exposure to the World Trade Center attacks. Psychoneuroendocrinology. 2013;38:2952-61. Clinical studies examining the FAAH C385A genetic polymorphism (rs324420) found that carriers of the A allele – which is associated with lower FAAH and higher AEA levels104104. Sipe JC, Chiang K, Gerber AL, Beutler E, Cravatt BF. A missense mutation in human fatty acid amide hydrolase associated with problem drug use. Proc Natl Acad Sci U S A. 2002;99:8394-9.-105. Chiang KP, Gerber AL, Sipe JC, Cravatt BF. Reduced cellular expression and activity of the P129T mutant of human fatty acid amide hydrolase: evidence for a link between defects in the endocannabinoid system and problem drug use. Hum Mol Genet. 2004;13:2113-9. 106106. Boileau I, Tyndale RF, Williams B, Mansouri E, Westwood DJ, Le Foll B, et al. The fatty acid amide hydrolase C385A variant affects brain binding of the positron emission tomography tracer [11C] CURB. J Cereb Blood Flow Metab. 2015;35:1237-40. – have a blunted amygdalar response to threat, greater ventral striatum response to reward, decreased correlation of amygdala reactivity and trait anxiety, enhanced fronto-amygdalar connectivity, and reduced stress reactivity.107107. Hariri AR, Gorka A, Hyde LW, Kimak M, Halder I, Ducci F, et al. Divergent effects of genetic variation in endocannabinoid signaling on human threat-and reward-related brain function. Biol Psychiatry. 2009;66:9-16.-108. Gunduz-Cinar O, MacPherson KP, Cinar R, Gamble-George J, Sugden K, Williams B, et al. Convergent translational evidence of a role for anandamide in amygdala-mediated fear extinction, threat processing and stress-reactivity. Mol Psychiatry. 2013;18:813-23. 109109. Dincheva I, Drysdale AT, Hartley CA, Johnson DC, Jing D, King EC, et al. FAAH genetic variation enhances fronto-amygdala function in mouse and human. Nat Commun. 2015;6:6395. Green et al.110110. Green DG, Kim J, Kish SJ, Tyndale RF, Hill MN, Strafella AP, et al. Fatty acid amide hydrolase binding is inversely correlated with amygdalar functional connectivity: a combined positron emission tomography and magnetic resonance imaging study in healthy individuals. J Psychiatry Neurosci. 2021;46:E238-46. failed to replicate the aforementioned relationship between the FAAH C385A polymorphism and amygdalar functional connectivity, possibly due to a small sample size. However, they did find that FAAH levels measured in vivo in the human brain are negatively correlated with fronto-amygdalar functional connectivity, suggesting that higher brain levels of AEA could increase coupling strength in fronto-amygdalar networks and affect stress response.110110. Green DG, Kim J, Kish SJ, Tyndale RF, Hill MN, Strafella AP, et al. Fatty acid amide hydrolase binding is inversely correlated with amygdalar functional connectivity: a combined positron emission tomography and magnetic resonance imaging study in healthy individuals. J Psychiatry Neurosci. 2021;46:E238-46. Another recent fMRI study in healthy males showed that neural activation of the anterior cingulate cortex and anterior insular cortex during extinction learning correlated positively with AEA baseline levels.111111. Spohrs J, Ulrich M, Grön G, Prost M, Plener PL, Fegert JM, et al. Fear extinction learning and anandamide: an fMRI study in healthy humans. Transl Psychiatry. 2021;11:161. Moreover, task-related changes in AEA were observed during fear extinction, suggesting that AEA may play a putative role in fear extinction learning.111111. Spohrs J, Ulrich M, Grön G, Prost M, Plener PL, Fegert JM, et al. Fear extinction learning and anandamide: an fMRI study in healthy humans. Transl Psychiatry. 2021;11:161. Finally, in the remarkable case report of a patient presenting with pain insensitivity and low fear and anxiety, the C385A polymorphism together with a microdeletion linked to decreased FAAH expression was detected as a possible causal factor. In addition, blood levels of AEA and other fatty-acid amides which are degraded by FAAH were unusually elevated in this individual.112112. Habib AM, Okorokov AL, Hill MN, Bras JT, Lee MC, Li S, et al. Microdeletion in a FAAH pseudogene identified in a patient with high anandamide concentrations and pain insensitivity. Br J Anaesth. 2019;123:e249-53.

The role of the ECS specifically in social anxiety is supported by various preclinical findings which demonstrate effects of ECS modulation, via either CB1 receptor activation or FAAH inhibition, on social interaction and social anxiety.113113. Trezza V, Vanderschuren LJ. Bidirectional cannabinoid modulation of social behavior in adolescent rats. Psychopharmacology (Berl). 2008;197:217-27.-114. Trezza V, Damsteegt R, Manduca A, Petrosino S, Van Kerkhof LW, Pasterkamp RJ, et al. Endocannabinoids in amygdala and nucleus accumbens mediate social play reward in adolescent rats. J Neurosci. 2012;32:14899-908. 115. Gonczarowska N, Tomaz C, Caixeta FV, Malcher-Lopes R, Barros M, Nishijo H, et al. CB1 receptor antagonism in capuchin monkeys alters social interaction and aversive memory extinction. Psychopharmacology (Berl). 2019;236:3413-9. 116. Manduca A, Servadio M, Campolongo P, Palmery M, Trabace L, Vanderschuren LJ, et al. Strain- and context-dependent effects of the anandamide hydrolysis inhibitor URB597 on social behavior in rats. Eur Neuropsychopharmacol. 2014;24:1337-48. 117. Matricon J, Seillier A, Giuffrida A. Distinct neuronal activation patterns are associated with PCP-induced social withdrawal and its reversal by the endocannabinoid-enhancing drug URB597. Neurosci Res. 2016;110:49-58. 118118. Schneider P, Bindila L, Schmahl C, Bohus M, Meyer-Lindenberg A, Lutz B, et al. Adverse social experiences in adolescent rats result in enduring effects on social competence, pain sensitivity and endocannabinoid signaling. Front Behav Neurosci. 2016;10:203. In comparison, clinical studies investigating this system in SAD are considerably limited. A recent clinical trial investigating the therapeutic effects of a FAAH inhibitor in SAD was negative. However, the authors observed a small to modest anxiolytic effect in patients with severe SAD and suggested that, based on the correlation between low trough concentrations of the inhibitor (i.e., the lowest concentration of the drug in the bloodstream) and low plasma AEA, future trials with a higher dose of the inhibitor may be warranted.119119. Schmidt ME, Liebowitz MR, Stein MB, Grunfeld J, Van Hove I, Simmons WK, et al. The effects of inhibition of fatty acid amide hydrolase (FAAH) by JNJ-42165279 in social anxiety disorder: a double-blind, randomized, placebo-controlled proof-of-concept study. Neuropsychopharmacology. 2021;46:1004-10. In addition, a recently published double-blind, placebo-controlled experimental study in healthy adults found that administration of the FAAH inhibitor PF-04457845 produced a 10-fold increase in peripheral AEA levels and decreased broad-spectrum fear-related phenotypes.120120. Mayo LM, Asratian A, Lindé J, Morena M, Haataja R, Hammar V, et al. Elevated anandamide, enhanced recall of fear extinction, and attenuated stress responses following inhibition of fatty acid amide hydrolase: a randomized, controlled experimental medicine trial. Biol Psychiatry. 2020;87:538-47. Furthermore, a 2021 double-blind, placebo-controlled clinical trial in healthy males employing the FAAH inhibitor JNJ-42165279 found that the drug attenuated activation in the amygdala, anterior cingulate, and bilateral insula during a face emotion processing task – effects which are consistent with those of previously observed anxiolytic agents.121121. Paulus MP, Stein MB, Simmons AN, Risbrough VB, Halter R, Chaplan SR. The effects of FAAH inhibition on the neural basis of anxiety-related processing in healthy male subjects: a randomized clinical trial. Neuropsychopharmacology. 2021;46:1011-9. Moreover, higher levels of plasma AEA were associated with greater attenuation in these brain regions.121121. Paulus MP, Stein MB, Simmons AN, Risbrough VB, Halter R, Chaplan SR. The effects of FAAH inhibition on the neural basis of anxiety-related processing in healthy male subjects: a randomized clinical trial. Neuropsychopharmacology. 2021;46:1011-9. While the latter two clinical studies were not conducted in a population with SAD, they suggest that FAAH inhibitors may have some potential in the treatment of fear-related disorders such as SAD.

Over the past two decades, cannabidiol (CBD), the primary non-psychotomimetic cannabinoid constituent of cannabis,122122. Boggs DL, Nguyen JD, Morgenson D, Taffe MA, Ranganathan M. Clinical and preclinical evidence for functional interactions of cannabidiol and Δ 9-tetrahydrocannabinol. Neuropsychopharmacology. 2018;43:142-54. has emerged as a drug with multiple potential therapeutic benefits including neuroprotective, anti-inflammatory, antioxidant, antipsychotic, and antianxiety effects (reviewed by McPartland et al.123123. McPartland JM, Duncan M, Di Marzo V, Pertwee RG. Are cannabidiol and Δ9‐tetrahydrocannabivarin negative modulators of the endocannabinoid system? A systematic review. Br J Pharmacol. 2015;172:737-53.). Although evidence is emerging that the anxiolytic effects of CBD may be mediated through serotonergic 5HT1a receptors124124. Russo EB, Burnett A, Hall B, Parker KK. Agonistic properties of cannabidiol at 5-HT1a receptors. Neurochem Res. 2005;30:1037-43.,125125. Fogaça MV, Reis FM, Campos AC, Guimaraes FS. Effects of intra-prelimbic prefrontal cortex injection of cannabidiol on anxiety-like behavior: involvement of 5HT1A receptors and previous stressful experience. Eur Neuropsychopharmacol. 2014;24:410-9. and vanilloid receptor type 1 (TRPV1)126126. Campos AC, Guimarães FS. Evidence for a potential role for TRPV1 receptors in the dorsolateral periaqueductal gray in the attenuation of the anxiolytic effects of cannabinoids. Prog Neuropsychopharmacol Biol Psychiatry. 2009;33:1517-21. receptors, some of these effects may also be exerted through modulation of the ECS. Specifically, mild agonistic effects on CB1 receptors and antagonistic effects on exogenous CB1 receptor agonists (THC) have been described.123123. McPartland JM, Duncan M, Di Marzo V, Pertwee RG. Are cannabidiol and Δ9‐tetrahydrocannabivarin negative modulators of the endocannabinoid system? A systematic review. Br J Pharmacol. 2015;172:737-53.,127127. Leweke FM, Piomelli D, Pahlisch F, Muhl D, Gerth CW, Hoyer C, et al. Cannabidiol enhances anandamide signaling and alleviates psychotic symptoms of schizophrenia. Transl Psychiatry. 2012;2:e94.,128128. Blessing EM, Steenkamp MM, Manzanares J, Marmar CR. Cannabidiol as a potential treatment for anxiety disorders. Neurotherapeutics. 2015;12:825-36. While the agonistic effects are still limited and controversial, emerging evidence suggests that CBD may act as a negative allosteric modulator of CB1 receptors, resulting in the aforementioned antagonistic effects on cannabinoid agonist activity.129129. Laprairie RB, Bagher AM, Kelly ME, Denovan-Wright EM. Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor. Br J Pharmacol. 2015;172:4790-805. Furthermore, there have been inconsistent findings on the effects of CBD on FAAH activity. Some animal studies found an inhibitory effect of CBD on FAAH activity in brain tissue of healthy mice,129129. Laprairie RB, Bagher AM, Kelly ME, Denovan-Wright EM. Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor. Br J Pharmacol. 2015;172:4790-805.,130130. Watanabe K, Ogi H, Nakamura S, Kayano Y, Matsunaga T, Yoshimura H, et al. Distribution and characterization of anandamide amidohydrolase in mouse brain and liver. Life Sci. 1998;62:1223-9. rats,131131. De Petrocellis L, Ligresti A, Moriello AS, Allarà M, Bisogno T, Petrosino S, et al. Effects of cannabinoids and cannabinoid‐enriched Cannabis extracts on TRP channels and endocannabinoid metabolic enzymes. Br J Pharmacol. 2011;163:1479-94. and cell membranes from mouse neuroblastoma,132132. Bisogno T, Hanuš L, De Petrocellis L, Tchilibon S, Ponde DE, Brandi I, et al. Molecular targets for cannabidiol and its synthetic analogues: effect on vanilloid VR1 receptors and on the cellular uptake and enzymatic hydrolysis of anandamide. Br J Pharmacol. 2001;134:845-52. whereas in vivo treatment of mouse glioma tissue with CBD increased FAAH activity.133133. Massi P, Valenti M, Vaccani A, Gasperi V, Perletti G, Marras E, et al. 5‐Lipoxygenase and anandamide hydrolase (FAAH) mediate the antitumor activity of cannabidiol, a non‐psychoactive cannabinoid. J Neurochem. 2008;104:1091-100. In addition, two studies reported a decrease in FAAH protein expression following CBD administration in chronically stressed mice134134. Fogaça MV, Campos AC, Coelho LD, Duman RS, Guimarães FS. The anxiolytic effects of cannabidiol in chronically stressed mice are mediated by the endocannabinoid system: role of neurogenesis and dendritic remodeling. Neuropharmacology. 2018;135:22-33. and LPS-treated mice.135135. de Filippis D, Iuvone T, D’amico A, Esposito G, Steardo L, Herman AG, et al. Effect of cannabidiol on sepsis‐induced motility disturbances in mice: involvement of CB1 receptors and fatty acid amide hydrolase. Neurogastroenterol Motil. 2008;20:919-27. Further investigation with more standardized methodologies may help elucidate the actions of CBD on FAAH.

Moreover, several preclinical studies136136. Almeida V, Levin R, Peres FF, Niigaki ST, Calzavara MB, Zuardi AW, et al. Cannabidiol exhibits anxiolytic but not antipsychotic property evaluated in the social interaction test. Prog Neuropsychopharmacol Biol Psychiatry. 2013;41:30-5.-137. Campos AC, de Paula Soares V, Carvalho MC, Ferreira FR, Vicente MA, Brandão ML, et al. Involvement of serotonin-mediated neurotransmission in the dorsal periaqueductal gray matter on cannabidiol chronic effects in panic-like responses in rats. Psychopharmacology (Berl). 2013;226:13-24. 138. Guimarães FS, Chiaretti TM, Graeff FG, Zuardi AW. Antianxiety effect of cannabidiol in the elevated plus-maze. Psychopharmacology (Berl). 1990;100:558-9. 139139. Resstel LB, Tavares RF, Lisboa SF, Joca SR, Corrêa FM, Guimarães FS. 5‐HT1A receptors are involved in the cannabidiol‐induced attenuation of behavioural and cardiovascular responses to acute restraint stress in rats. Br J Pharmacol. 2009;156:181-8. investigating effects of CBD in different animal models of anxiety have repeatedly demonstrated anxiolytic effects, as comprehensively reviewed by Blessing and colleagues.128128. Blessing EM, Steenkamp MM, Manzanares J, Marmar CR. Cannabidiol as a potential treatment for anxiety disorders. Neurotherapeutics. 2015;12:825-36. Among these, some animal studies have found that CBD produces anxiolytic effects following an inverted U-shaped dose-response curve.138138. Guimarães FS, Chiaretti TM, Graeff FG, Zuardi AW. Antianxiety effect of cannabidiol in the elevated plus-maze. Psychopharmacology (Berl). 1990;100:558-9.,140140. Linares IM, Zuardi AW, Pereira LC, Queiroz RH, Mechoulam R, Guimarães FS, et al. Cannabidiol presents an inverted U-shaped dose-response curve in a simulated public speaking test. Braz J Psychiatry. 2019;41:9-14.,141141. Nazario LR, Antonioli RJ, Capiotti KM, Hallak JE, Zuardi AW, Crippa JA, et al. Reprint of “caffeine protects against memory loss induced by high and non-anxiolytic dose of cannabidiol in adult zebrafish (Danio rerio). Pharmacol Biochem Behav. 2015;139 Pt B:134-40. Interestingly, this bell-shaped response is not exclusive to the anxiolytic effects of CBD, as it has also been observed in animal models of depression,142142. Zanelati TV, Biojone C, Moreira FA, Guimarães FS, Joca SR. Antidepressant-like effects of cannabidiol in mice: Possible involvement of 5-HT 1A receptors. Br J Pharmacol. 2010;159:122-8. compulsive behavior,143143. Casarotto PC, Gomes FV, Resstel LB, Guimarães FS. Cannabidiol inhibitory effect on marble-burying behaviour: Involvement of CB1 receptors. Behav Pharmacol. 2010;21:353-8. schizophrenia,144144. Levin R, Peres FF, Almeida V, Calzavara MB, Zuardi AW, Hallak JEC, et al. Effects of cannabinoid drugs on the deficit of prepulse inhibition of startle in an animal model of schizophrenia: the SHR strain. Front Pharmacol. 2014;5:10. cognitive impairment,145145. Peres FF, Levin R, Suiama MA, Diana MC, Gouvêa DA, Almeida V, et al. Cannabidiol prevents motor and cognitive impairments induced by reserpine in rats. Front Pharmacol. 2016;7:343. and other ailments.140140. Linares IM, Zuardi AW, Pereira LC, Queiroz RH, Mechoulam R, Guimarães FS, et al. Cannabidiol presents an inverted U-shaped dose-response curve in a simulated public speaking test. Braz J Psychiatry. 2019;41:9-14. These findings suggest that the optimal dose of CBD may depend on condition, indicating the need to test different doses in animals and humans in order to elucidate its therapeutic potential for the treatment of SAD and other anxiety disorders.

In terms of clinical findings, one study in human cells suggests that CBD increases AEA levels through binding to fatty acid-binding proteins and thereby prevents AEA uptake and catabolism by FAAH,146146. Elmes MW, Kaczocha M, Berger WT, Leung K, Ralph BP, Wang L, et al. Fatty acid-binding proteins (FABPs) are intracellular carriers for Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD). J Biol Chem. 2015;290:8711-21. rather than inhibiting FAAH directly. In addition, results from a clinical trial in schizophrenia indicate higher AEA levels in individuals receiving treatment with CBD.127127. Leweke FM, Piomelli D, Pahlisch F, Muhl D, Gerth CW, Hoyer C, et al. Cannabidiol enhances anandamide signaling and alleviates psychotic symptoms of schizophrenia. Transl Psychiatry. 2012;2:e94. While direct measurement of brain FAAH activity has been made possible by the novel PET radiotracer [C-11]CURB,147147. Wilson AA, Garcia A, Parkes J, Houle S, Tong J, Vasdev N. [11C]CURB: evaluation of a novel radiotracer for imaging fatty acid amide hydrolase by positron emission tomography. Nucl Med Biol. 2011;38:247-53. there have been to our knowledge no studies directly investigating brain FAAH activity before and after treatment with CBD. Finally, three randomized-controlled clinical trials in SAD have shown that CBD may improve SAD symptoms (Table 1).148148. Crippa JA, Derenusson GN, Ferrari TB, Wichert-Ana L, Duran FL, Martin-Santos R, et al. Neural basis of anxiolytic effects of cannabidiol (CBD) in generalized social anxiety disorder: a preliminary report. J Psychopharmacol. 2011;25:121-30.-149. Bergamaschi MM, Queiroz RH, Chagas MH, de Oliveira DC, De Martinis BS, Kapczinski F, et al. Cannabidiol reduces the anxiety induced by simulated public speaking in treatment-naive social phobia patients. Neuropsychopharmacology. 2011;36:1219-26. 150150. Masataka N. Anxiolytic effects of repeated cannabidiol treatment in teenagers with social anxiety disorders. Front Psychol. 2019;10:2466. Although these results appear to be promising, it should be noted that these trials were conducted in small samples (10 to 37 participants) and employed inconsistent assessment tools across studies. Such limitations warrant further investigation to produce robust findings and better understand the biological mechanisms of CBD action in SAD.

Table 1
Randomized, double-blind, placebo-controlled clinical trials of drugs targeting the ECS in SAD

Interaction of the endocannabinoid system with other systems and biological findings

As discussed, previous research has largely focused on establishing monoaminergic impairment affecting the limbic system22. Leichsenring F, Leweke F. Social anxiety disorder. N Engl J Med. 2017;376:2255-64.,152152. Marcin MS, Nemeroff CB. The neurobiology of social anxiety disorder: the relevance of fear and anxiety. Acta Psychiatr Scand Suppl. 2003;108:51-64. and dysfunction of the HPA axis153153. Faravelli C, Lo Sauro C, Godini L, Lelli L, Benni L, Pietrini F, et al. Childhood stressful events, HPA axis and anxiety disorders. World J psychiatry. 2012;2:13-25. as pathophysiological mechanisms of SAD. Accordingly, treatments developed and/or tested for SAD have mainly targeted serotonin (5-HT) and norepinephrine (NE) neurotransmission, specifically via drugs blocking their reuptake.88. Williams T, Hattingh CJ, Kariuki CM, Tromp SA, van Balkom AJ, Ipser JC, et al. Pharmacotherapy for social anxiety disorder (SAnD). Cochrane Database Syst Rev. 2017;10;CD001206. However, these drugs are ineffective in more than one-third of SAD patients.22. Leichsenring F, Leweke F. Social anxiety disorder. N Engl J Med. 2017;376:2255-64.

Recently, the ECS has been identified as a modulator of the aforementioned systems and incited interest in the development of new pharmacological treatments for mood and anxiety disorders. In particular, two preclinical studies found that synthetic CB1 receptor agonism was associated with increased NE efflux and anxiety-like behavior in healthy rodents.154154. Page ME, Oropeza VC, Sparks SE, Qian Y, Menko AS, Van Bockstaele EJ. Repeated cannabinoid administration increases indices of noradrenergic activity in rats. Pharmacol Biochem Behav. 2007;86:162-8.,155155. Page ME, Oropeza VC, Van Bockstaele EJ. Local administration of a cannabinoid agonist alters norepinephrine efflux in the rat frontal cortex. Neurosci Lett. 2008;431:1-5. The latter study also showed that local administration of a CB1 receptor antagonist (SR 141716A) followed by local administration of a CB1 receptor agonist (WIN55,212-2) produces a paradoxical inhibition of NE efflux.155155. Page ME, Oropeza VC, Van Bockstaele EJ. Local administration of a cannabinoid agonist alters norepinephrine efflux in the rat frontal cortex. Neurosci Lett. 2008;431:1-5. Together, these findings suggest that enhanced NE activity may be mediated, in part, by direct CB1 receptor agonism, localized at noradrenergic axon terminals. However, the paradoxical decrease in NE observed in the second study may also indicate that NE efflux is modulated indirectly via interneurons or other chemically distinct afferents.154154. Page ME, Oropeza VC, Sparks SE, Qian Y, Menko AS, Van Bockstaele EJ. Repeated cannabinoid administration increases indices of noradrenergic activity in rats. Pharmacol Biochem Behav. 2007;86:162-8. Similarly, direct or indirect stimulation of CB1 receptors has been linked to enhanced 5-HT neuronal activity.156156. Bambico FR, Katz N, Debonnel G, Gobbi G. Cannabinoids elicit antidepressant-like behavior and activate serotonergic neurons through the medial prefrontal cortex. J Neurosci. 2007;27:11700-11.

157. Gobbi G, Bambico FR, Mangieri R, Bortolato M, Campolongo P, Solinas M, et al. Antidepressant-like activity and modulation of brain monoaminergic transmission by blockade of anandamide hydrolysis. Proc Natl Acad Sci U S A. 2005;102:18620-5.
-158158. Cassano T, Gaetani S, Macheda T, Laconca L, Romano A, Morgese MG, et al. Evaluation of the emotional phenotype and serotonergic neurotransmission of fatty acid amide hydrolase-deficient mice. Psychopharmacology (Berl). 2011;214:465-76. However, these studies found that increased 5-HT neurotransmission following CB1 receptor agonism,156156. Bambico FR, Katz N, Debonnel G, Gobbi G. Cannabinoids elicit antidepressant-like behavior and activate serotonergic neurons through the medial prefrontal cortex. J Neurosci. 2007;27:11700-11. FAAH inhibition,157157. Gobbi G, Bambico FR, Mangieri R, Bortolato M, Campolongo P, Solinas M, et al. Antidepressant-like activity and modulation of brain monoaminergic transmission by blockade of anandamide hydrolysis. Proc Natl Acad Sci U S A. 2005;102:18620-5. or FAAH knockout158158. Cassano T, Gaetani S, Macheda T, Laconca L, Romano A, Morgese MG, et al. Evaluation of the emotional phenotype and serotonergic neurotransmission of fatty acid amide hydrolase-deficient mice. Psychopharmacology (Berl). 2011;214:465-76. was associated with enhanced stress-coping behaviors and antidepressant effects in otherwise healthy rodents.

Accumulating evidence also suggests that the ECS may act as a homeostatic regulator of HPA axis activity under basal and stress-related conditions.159159. Micale V, Drago F. Endocannabinoid system, stress and HPA axis. Eur J Pharmacol. 2018;834:230-9. In this regard, studies have found that chronic stress exposure or repeated corticosterone treatment activate the HPA axis response, which has been associated with changes in AEA levels, FAAH activity, and CB1 receptor expression.9090. Gray JM, Vecchiarelli HA, Morena M, Lee TT, Hermanson DJ, Kim AB, et al. Corticotropin-releasing hormone drives anandamide hydrolysis in the amygdala to promote anxiety. J Neurosci. 2015;35:3879-92.,159159. Micale V, Drago F. Endocannabinoid system, stress and HPA axis. Eur J Pharmacol. 2018;834:230-9.-160. Gray JM, Wilson CD, Lee TT, Pittman QJ, Deussing JM, Hillard CJ, et al. Sustained glucocorticoid exposure recruits cortico-limbic CRH signaling to modulate endocannabinoid function. Psychoneuroendocrinology. 2016;66:151-8. 161161. Hill MN, McLaughlin RJ, Morrish AC, Viau V, Floresco SB, Hillard CJ, et al. Suppression of amygdalar endocannabinoid signaling by stress contributes to activation of the hypothalamic-pituitary-adrenal axis. Neuropsychopharmacology. 2009;34:2733-45. Moreover, Di et al.162162. Di S, Malcher-Lopes R, Halmos KC, Tasker JG. Nongenomic glucocorticoid inhibition via endocannabinoid release in the hypothalamus: a fast feedback mechanism. J Neurosci. 2003;23:4850-7. showed that the paraventricular nucleus (PVN) participates in glucocorticoid-mediated feedback regulation of the HPA axis via endocannabinoid release. Specifically, this model suggests that, during the stress response, activation of hippocampal glucocorticoid receptors (via corticosterone) induces the synthesis of endocannabinoid ligands which bind to CB1 receptors on glutamatergic neurons.162162. Di S, Malcher-Lopes R, Halmos KC, Tasker JG. Nongenomic glucocorticoid inhibition via endocannabinoid release in the hypothalamus: a fast feedback mechanism. J Neurosci. 2003;23:4850-7. This binding activates a signaling cascade that leads to the inhibition of glutamate release onto the hypothalamic PVN, decreasing PVN neuronal activity and further hormone secretion.162162. Di S, Malcher-Lopes R, Halmos KC, Tasker JG. Nongenomic glucocorticoid inhibition via endocannabinoid release in the hypothalamus: a fast feedback mechanism. J Neurosci. 2003;23:4850-7. It is possible, then, that disruption of certain ECS processes may lead to dysregulation of HPA axis activity, which together may contribute to the development of sustained anxiety-related symptoms.

ECS interactions with oxytocin, the neuropeptide that reinforces social bonding and social reward, have also been observed. Specifically, Wei et al.163163. Wei D, Lee D, Cox CD, Karsten CA, Penagarikano O, Geschwind DH, et al. Endocannabinoid signaling mediates oxytocin-driven social reward. Proc Natl Acad Sci U S A. 2015;112:14084-9. showed that oxytocin drives AEA mobilization in the mouse nucleus accumbens, leading to reinforcement of social reward behavior. Pharmacological blockade of oxytocin receptors attenuated this response, whereas FAAH inhibition or gene deletion offset the behavioral effects of oxytocin receptor blockade.163163. Wei D, Lee D, Cox CD, Karsten CA, Penagarikano O, Geschwind DH, et al. Endocannabinoid signaling mediates oxytocin-driven social reward. Proc Natl Acad Sci U S A. 2015;112:14084-9. These findings suggest that the ECS may modulate the prosocial effects of oxytocin, which may open potential avenues of research in disorders of social impairment such as SAD.

Genetic studies examining ECS candidate genes relating to anxiety disorders have produced limited findings. Current evidence suggests that a single nucleotide polymorphism in the FAAH gene (rs324420; C385A)104104. Sipe JC, Chiang K, Gerber AL, Beutler E, Cravatt BF. A missense mutation in human fatty acid amide hydrolase associated with problem drug use. Proc Natl Acad Sci U S A. 2002;99:8394-9. influences FAAH protein expression,105105. Chiang KP, Gerber AL, Sipe JC, Cravatt BF. Reduced cellular expression and activity of the P129T mutant of human fatty acid amide hydrolase: evidence for a link between defects in the endocannabinoid system and problem drug use. Hum Mol Genet. 2004;13:2113-9.,106106. Boileau I, Tyndale RF, Williams B, Mansouri E, Westwood DJ, Le Foll B, et al. The fatty acid amide hydrolase C385A variant affects brain binding of the positron emission tomography tracer [11C] CURB. J Cereb Blood Flow Metab. 2015;35:1237-40. which has been found to correlate with trait anxiety.107107. Hariri AR, Gorka A, Hyde LW, Kimak M, Halder I, Ducci F, et al. Divergent effects of genetic variation in endocannabinoid signaling on human threat-and reward-related brain function. Biol Psychiatry. 2009;66:9-16. However, there is little to no evidence to support an association between genetic variants of other ECS genes and symptoms of SAD.

Thus, further investigation of ECS interactions with other biological systems and genetic variables may help improve our current knowledge and address the gap with respect to SAD pathophysiology, potentially allowing for the development of more targeted treatment options.

Summary of current drugs targeting the endocannabinoid system

Based on current evidence, it appears that the ECS may play a role in buffering against stress and anxiety. Thus, targeting components of this system could serve as a potentially valid strategy for the treatment of social anxiety symptoms. In this regard, several ECS-targeting drugs have been developed, with some demonstrating promising therapeutic potential.

Logically, CB1 agonists were considered promising, given their ability to enhance ECS activity and reduce anxiety-like behaviors in rodents.8080. Micale V, Di Marzo V, Sulcova A, Wotjak CT, Drago F. Endocannabinoid system and mood disorders: priming a target for new therapies. Pharmacol Ther. 2013;138:18-37.,164164. Haller J, Varga B, Ledent C, Freund TF. CB1 cannabinoid receptors mediate anxiolytic effects: convergent genetic and pharmacological evidence with CB1-specific agents. Behav Pharmacol. 2004;15:299-304.-165. Hill MN, Gorzalka BB. Enhancement of anxiety-like responsiveness to the cannabinoid CB1 receptor agonist HU-210 following chronic stress. Eur J Pharmacol. 2004;499:291-5. 166166. Patel S, Hillard CJ. Pharmacological evaluation of cannabinoid receptor ligands in a mouse model of anxiety: further evidence for an anxiolytic role for endogenous cannabinoid signaling. J Pharmacol Exp Ther. 2006;318:304-11. Notable synthetic CB1 agonists include WIN55,212-2, CP-55940, and HU-210. Although these compounds are generally well-tolerated, accompanying risk of other psychotropic adverse effects has been observed.167167. Moreira FA, Grieb M, Lutz B. Central side-effects of therapies based on CB1 cannabinoid receptor agonists and antagonists: focus on anxiety and depression. Best Pract Res Clin Endocrinol Metab. 2009;23:133-44. Direct CB1 agonism has also been associated with disturbances in social behavior and social play in some preclinical studies, prompting the exploration of other ECS targets.113113. Trezza V, Vanderschuren LJ. Bidirectional cannabinoid modulation of social behavior in adolescent rats. Psychopharmacology (Berl). 2008;197:217-27.,168168. Loureiro M, Kramar C, Renard J, Rosen LG, Laviolette SR. Cannabinoid transmission in the hippocampus activates nucleus accumbens neurons and modulates reward and aversion-related emotional salience. Biol Psychiatry. 2016;80:216-25.-169. Trezza V, Vanderschuren LJ. Cannabinoid and opioid modulation of social play behavior in adolescent rats: differential behavioral mechanisms. Eur Neuropsychopharmacol. 2008;18:519-30. 170170. Schneider M, Schömig E, Leweke FM. Acute and chronic cannabinoid treatment differentially affects recognition memory and social behavior in pubertal and adult rats. Addict Biol. 2008;13:345-57.

One of the most effective strategies to target anxiety through modulation of the ECS has been inhibition of the endocannabinoid degradation enzymes FAAH and MAGL. This allows for the indirect activation of CB1 receptors by increased levels of AEA and 2-AG. Accumulating evidence has demonstrated the anxiolytic effects of FAAH inhibitors (URB597, PF-3845, and PF-04457845), most prominently in rodents,8080. Micale V, Di Marzo V, Sulcova A, Wotjak CT, Drago F. Endocannabinoid system and mood disorders: priming a target for new therapies. Pharmacol Ther. 2013;138:18-37. as well as the aforementioned human experimental PF-04457845 trial.120120. Mayo LM, Asratian A, Lindé J, Morena M, Haataja R, Hammar V, et al. Elevated anandamide, enhanced recall of fear extinction, and attenuated stress responses following inhibition of fatty acid amide hydrolase: a randomized, controlled experimental medicine trial. Biol Psychiatry. 2020;87:538-47. Interestingly, prosocial effects of these compounds have also been consistently observed in rodent models of social impairment.117117. Matricon J, Seillier A, Giuffrida A. Distinct neuronal activation patterns are associated with PCP-induced social withdrawal and its reversal by the endocannabinoid-enhancing drug URB597. Neurosci Res. 2016;110:49-58.,171171. Realini N, Vigano’ D, Guidali C, Zamberletti E, Rubino T, Parolaro D. Chronic URB597 treatment at adulthood reverted most depressive-like symptoms induced by adolescent exposure to THC in female rats. Neuropharmacology. 2011;60:235-43.-172. Seillier A, Advani T, Cassano T, Hensler JG, Giuffrida T. Inhibition of fatty-acid amide hydrolase and CB1 receptor antagonism differentially affect behavioural responses in normal and PCP-treated rats. Int J Neuropsychopharmacol. 2010;13:373-86. 173. Seillier A, Martinez AA, Giuffrida A. Phencyclidine-induced social withdrawal results from deficient stimulation of cannabinoid CB1 receptors: implications for schizophrenia. Neuropsychopharmacology. 2013;38:1816-24. 174174. Melancia F, Schiavi S, Servadio M, Cartocci V, Campolongo P, Palmery M, et al. Sex-specific autistic endophenotypes induced by prenatal exposure to valproic acid involve anandamide signalling. Br J Pharmacol. 2018;175:3699-712. Although more limited in study, MAGL inhibitors (URB602, JZL184) have demonstrated similar results in terms of improved anxiety and social behavior.8080. Micale V, Di Marzo V, Sulcova A, Wotjak CT, Drago F. Endocannabinoid system and mood disorders: priming a target for new therapies. Pharmacol Ther. 2013;138:18-37.,175175. Manduca A, Morena M, Campolongo P, Servadio M, Palmery M, Trabace L, et al. Distinct roles of the endocannabinoids anandamide and 2-arachidonoylglycerol in social behavior and emotionality at different developmental ages in rats. Eur Neuropsychopharmacol. 2015;25:1362-74.,176176. Schiavi S, Manduca A, Segatto M, Campolongo P, Pallottini V, Vanderschuren LJ, et al. Unidirectional opioid-cannabinoid cross-tolerance in the modulation of social play behavior in rats. Psychopharmacology (Berl). 2019;236:2557-68. While a handful of inhibitor studies have reported insignificant results,177177. Moise AM, Eisenstein SA, Astarita G, Piomelli D, Hohmann AG. An endocannabinoid signaling system modulates anxiety-like behavior in male Syrian hamsters. Psychopharmacology (Berl). 2008;200:333-46.-178. Qin M, Zeidler Z, Moulton K, Krych L, Xia Z, Smith CB. Endocannabinoid-mediated improvement on a test of aversive memory in a mouse model of fragile X syndrome. Behav Brain Res. 2015;291:164-71. 179179. Seillier A, Giuffrida A. The cannabinoid transporter inhibitor OMDM-2 reduces social interaction: Further evidence for transporter-mediated endocannabinoid release. Neuropharmacology. 2018;130:1-9. the vast majority suggest that inhibition of endocannabinoid degradation enzymes may serve as a promising novel treatment strategy for anxiety-related disorders.

The opposing effects on anxiety and social behavior exerted through direct versus indirect CBR activation may be due to differences in the mechanism of action (e.g. partial vs full agonistic properties at CBR) and/or affinities of endogenous vs synthetic ligands for CB1R vs CB2R affinity.180180. Shahbazi F, Grandi V, Banerjee A, Trant JF. Cannabinoids and cannabinoid receptors: the story so far. iScience. 2020;23:101301. However, this variability may also be explained by existing differences in the underlying status of the ECS in animals with social impairments, such as ECS receptor expression, ligand levels, or enzyme activity.173173. Seillier A, Martinez AA, Giuffrida A. Phencyclidine-induced social withdrawal results from deficient stimulation of cannabinoid CB1 receptors: implications for schizophrenia. Neuropsychopharmacology. 2013;38:1816-24.,174174. Melancia F, Schiavi S, Servadio M, Cartocci V, Campolongo P, Palmery M, et al. Sex-specific autistic endophenotypes induced by prenatal exposure to valproic acid involve anandamide signalling. Br J Pharmacol. 2018;175:3699-712.,181181. Doenni VM, Gray JM, Song CM, Patel S, Hill MN, Pittman QJ. Deficient adolescent social behavior following earaly-life inflammation is ameliorated by augmentation of anandamide signaling. Brain Behav Immun. 2016;58:237-47. These aspects warrant further investigation of changes in ECS components in social impairment models as well as in human studies of disorders associated with social impairment in order to validate the therapeutic potential and potential risks of these ECS-targeting agents in clinical settings.

In contrast, CB1 inverse agonists/antagonists have been shown to further exacerbate anxiety-like behaviors. As mentioned above the inverse agonist Rimonabant, found to be effective for weight-loss and smoking cessation, was quickly withdrawn following the emergence of severe psychiatric side effects.182182. Sam AH, Salem V, Ghatei MA. Rimonabant: from RIO to Ban. J Obes. 2011;2011:432607. In line with this, CB1 inverse agonist, AM251 has been found to reduce social interactions in rodents183183. Litvin Y, Phan A, Hill MN, Pfaff DW, McEwen BS. CB1 receptor signaling regulates social anxiety and memory. Genes Brain Behav. 2013;12:479-89.,184184. Bogáthy E, Kostyalik D, Petschner P, Vas S, Bagdy G. Blockade of serotonin 2C receptors with SB-242084 moderates reduced locomotor activity and rearing by cannabinoid 1 receptor antagonist AM-251. Pharmacology. 2019;103:151-8. and non-human primates.115115. Gonczarowska N, Tomaz C, Caixeta FV, Malcher-Lopes R, Barros M, Nishijo H, et al. CB1 receptor antagonism in capuchin monkeys alters social interaction and aversive memory extinction. Psychopharmacology (Berl). 2019;236:3413-9. Interestingly and in contrast to CB1 inverse agonists, two preclinical studies found that administration of a CB1 neutral antagonist, AM4113, did not produce anxiogenic or depressive side effects when administered in rats.185185. Gueye AB, Pryslawsky Y, Trigo JM, Poulia N, Delis F, Antoniou K, et al. The CB1 neutral antagonist AM4113 retains the therapeutic efficacy of the inverse agonist rimonabant for nicotine dependence and weight loss with better psychiatric tolerability. Int J Neuropsychopharmacol. 2016;19:pyw068.,186186. Sink KS, Segovia KN, Sink J, Randall PA, Collins LE, Correa M, et al. Potential anxiogenic effects of cannabinoid CB1 receptor antagonists/inverse agonists in rats: comparisons between AM4113, AM251, and the benzodiazepine inverse agonist FG-7142. Eur Neuropsychopharmacol. 2010;20:112-22. Similarly, two other rodent studies found that Rimonabant, but not the CB1 neutral antagonist tetrahydrocannabivarin, produced depressive and anxiogenic-like behaviors following administration.187187. O’Brien LD, Wills KL, Segsworth B, Dashney B, Rock EM, Limebeer CL, et al. Effect of chronic exposure to rimonabant and phytocannabinoids on anxiety-like behavior and saccharin palatability. Pharmacol Biochem Behav. 2013;103:597-602.,188188. Jutkiewicz EM, Makriyannis A, Vemuri K, Bergman J. Pro-depressant-like effects of CB1 receptor inverse agonists/antagonists in male Sprague-Dawley rats. FASEB J. 2010;24:581.7. While inverse agonists induce a pharmacological response opposite to that of agonists, neutral antagonists do not activate the receptor – differences which may partly explain the absence of these behavioral side effects following administration of the latter group of agents.185185. Gueye AB, Pryslawsky Y, Trigo JM, Poulia N, Delis F, Antoniou K, et al. The CB1 neutral antagonist AM4113 retains the therapeutic efficacy of the inverse agonist rimonabant for nicotine dependence and weight loss with better psychiatric tolerability. Int J Neuropsychopharmacol. 2016;19:pyw068.

Overall, the development of ECS-targeting agents has produced some promising results with regards to modulation of anxiety-like symptoms and social behavior, particularly in the field of FAAH and MAGL inhibitors. However, current evidence largely focuses on measures of general anxiety in animal models, and lacks the support of clinical trials. Further investigation of the effect of ECS-targeting drugs, specifically in animal models of social impairment and human studies, may provide greater insight as to how these compounds effect other biological systems, the possibility of risks and adverse effects, as well as their therapeutic potential in SAD.

Future research and potential challenges

This article highlights some of the currently known pathophysiological mechanisms of SAD and the therapeutic potential of the ECS. Targeting specific components of the ECS which have been shown to play a role in anxiety-related symptoms and social behavior may serve as a novel and possibly more targeted treatment for such disorders. However, there are a number of challenges to consider with respect to this new approach.

Firstly, while CB1 activation has been hypothesized to improve social anxiety-like behaviors by enhancing ECS signaling, activation of this receptor has since also demonstrated increased risk of social113113. Trezza V, Vanderschuren LJ. Bidirectional cannabinoid modulation of social behavior in adolescent rats. Psychopharmacology (Berl). 2008;197:217-27.,168168. Loureiro M, Kramar C, Renard J, Rosen LG, Laviolette SR. Cannabinoid transmission in the hippocampus activates nucleus accumbens neurons and modulates reward and aversion-related emotional salience. Biol Psychiatry. 2016;80:216-25.-169. Trezza V, Vanderschuren LJ. Cannabinoid and opioid modulation of social play behavior in adolescent rats: differential behavioral mechanisms. Eur Neuropsychopharmacol. 2008;18:519-30. 170170. Schneider M, Schömig E, Leweke FM. Acute and chronic cannabinoid treatment differentially affects recognition memory and social behavior in pubertal and adult rats. Addict Biol. 2008;13:345-57. and cognitive189189. Puighermanal E, Busquets-Garcia A, Maldonado R, Ozaita A. Cellular and intracellular mechanisms involved in the cognitive impairment of cannabinoids. Philos Trans R Soc Lond B Biol Sci. 2012;367:3254-63. impairments, as well as disturbances in the reward system.190190. Wiskerke J, Pattij T, Schoffelmeer AN, De Vries TJ. The role of CB1 receptors in psychostimulant addiction. Addict Biol. 2008;13:225-38.,191191. Parsons LH, Hurd YL. Endocannabinoid signalling in reward and addiction. Nat Rev Neurosci. 2015;16:579-94. As previously discussed, indirect CB1 activation via FAAH or MAGL inhibition has been the most commonly proposed solution to possibly avoid these adverse effects. Additionally, peripheral CB1 agonists and CB1 positive allosteric modulators have also demonstrated the ability to avoid certain CB1-related side effects.192192. Mulpuri Y, Marty VN, Munier JJ, Mackie K, Schmidt BL, Seltzman HH, et al. Synthetic peripherally-restricted cannabinoid suppresses chemotherapy-induced peripheral neuropathy pain symptoms by CB1 receptor activation. Neuropharmacology. 2018;139:85-97.-193. Slivicki RA, Xu Z, Kulkarni PM, Pertwee RG, Mackie K, Thakur GA, et al. Positive allosteric modulation of cannabinoid receptor type 1 suppresses pathological pain without producing tolerance or dependence. Biol Psychiatry. 2018;84:722-33. 194194. Stasiulewicz A, Znajdek K, Grudzień M, Pawiński T, Sulkowska JI. A guide to targeting the endocannabinoid system in drug design. Int J Mol Sci. 2020;21:2778. Despite these alternative solutions, the vast majority of current supporting evidence is a result of animal experimentation. As such, the efficacy, safety, and tolerability of such drugs must be further validated through clinical studies to evaluate not only their therapeutic potential, but also their possible risks.

Secondly, it is important to consider that the ECS is a complex biological system that does not function in isolation, but rather through interactions with an array of other systems. In this regard, the ECS may interact with and modulate other systems including the endovanilloid,194194. Stasiulewicz A, Znajdek K, Grudzień M, Pawiński T, Sulkowska JI. A guide to targeting the endocannabinoid system in drug design. Int J Mol Sci. 2020;21:2778.,195195. Rubino T, Realini N, Castiglioni C, Guidali C, Vigano D, Marras E, et al. Role in anxiety behavior of the endocannabinoid system in the prefrontal cortex. Cereb Cortex. 2008;18:1292-301. opioid,196196. Befort K. Interactions of the opioid and cannabinoid systems in reward: Insights from knockout studies. Front Pharmacol. 2015;6:6. noradrenergic,197197. Carvalho AF, Van Bockstaele EJ. Cannabinoid modulation of noradrenergic circuits: implications for psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry. 2012;38:59-67. prostanoid,198198. Buisseret B, Alhouayek M, Guillemot-Legris O, Muccioli GG. Endocannabinoid and prostanoid crosstalk in pain. Trends Mol Med. 2019;25:882-96. serotonergic,199199. Haj-Dahmane S, Shen RY. Modulation of the serotonin system by endocannabinoid signaling. Neuropharmacology. 2011;61:414-20. dopaminergic,200200. Marcellino D, Carriba P, Filip M, Borgkvist A, Frankowska M, Bellido I, et al. Antagonistic cannabinoid CB1/dopamine D2 receptor interactions in striatal CB1/D2 heteromers. A combined neurochemical and behavioral analysis. Neuropharmacology. 2008;54:815-23. glutamatergic,201201. Rodríguez-Muñoz M, Sánchez-Blázquez P, Merlos M, Garzón-Niño J. Endocannabinoid control of glutamate NMDA receptors: the therapeutic potential and consequences of dysfunction. Oncotarget. 2016;7:55840. and GABAergic202202. Musella A, Fresegna D, Rizzo FR, Gentile A, Bullitta S, De Vito F, et al. A novel crosstalk within the endocannabinoid system controls GABA transmission in the striatum. Sci Rep. 2017;7:7363. systems, among other biological networks.194194. Stasiulewicz A, Znajdek K, Grudzień M, Pawiński T, Sulkowska JI. A guide to targeting the endocannabinoid system in drug design. Int J Mol Sci. 2020;21:2778. Accounting for these additional biological networks will be crucial in understanding the larger physiological context and environment in which the ECS functions, and, by extension, the biological pathways that pharmacological interventions in the ECS can potentially influence.

A third challenge to consider is the pathophysiological heterogeneity of individuals with SAD. While the pathophysiological mechanisms of SAD are not yet certain, a number of biological systems have been described above which may additionally interact with the ECS. In this regard, it is possible that SAD in some individuals may be due to greater dysregulation of the HPA axis,5151. Faravelli C, Sauroc CL, Lelli L, Pietrini F, Lazzeretti L, Godini L, et al. The role of life events and HPA axis in anxiety disorders: a review. Curr Pharm Des. 2012;18:5663-74. whereas others may have more severe disruptions in their serotonergic/dopaminergic systems4242. Doruyter AG, Dupont P, Stein DJ, Lochner C, Warwick JM. Nuclear neuroimaging in social anxiety disorder: a review. J Nucl Med. 2018;59:1794-800. or inflammatory processes.203203. Michopoulos V, Powers A, Gillespie CF, Ressler KJ, Jovanovic T. Inflammation in fear-and anxiety-based disorders: PTSD, GAD, and beyond. Neuropsychopharmacology. 2017;42:254-70.,204204. Uzun N, Akıncı MA. Hemogram parameters in childhood anxiety disorders: could anxiety disorders be related with inflammation? Med Hypotheses. 2021;146:110440. Accordingly, it is possible that ECS-targeted treatment may be beneficial to certain neurobiological subgroups of SAD patients, but potentially ineffective or even possibly harmful to others. As such, future research efforts should also focus on elucidating biological subpopulations in SAD and how ECS-targeted therapies could be individualized to maximize their therapeutic potential.

Conclusion

Overall, the ECS presents as a potential biological pathway in the pathophysiology of SAD and as promising avenue for developing novel therapeutic approaches. The lack of human ECS studies and clinical trials, combined with the complex nature of the ECS and the heterogenous pathophysiology of SAD, highlight significant gaps in our knowledge and possible challenges, though at the same time great potential for future research. Further investigational efforts – specifically in human populations – are warranted to improve our knowledge of the ECS in SAD and help clarify these emerging questions. In turn, such research may allow for the development of more targeted pharmacological treatment interventions for individuals with SAD.

Acknowledgements

SK is supported by the Academic Scholar Award and the Labatt Family Innovation Fund in Brain Health, Department of Psychiatry, University of Toronto, Toronto, ON, Canada.

References

  • 1
    American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5). Arlington: American Psychiatric Publishing; 2013.
  • 2
    Leichsenring F, Leweke F. Social anxiety disorder. N Engl J Med. 2017;376:2255-64.
  • 3
    Schneier FR, Johnson J, Hornig CD, Liebowitz MR, Weissman MM. Social phobia. Comorbidity and morbidity in an epidemiologic sample. Arch Gen Psychiatry. 1992;49:282-8.
  • 4
    Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, Walters EE. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62:593-602.
  • 5
    Koyuncu A, İnce E, Ertekin E, Tükel R. Comorbidity in social anxiety disorder: diagnostic and therapeutic challenges. Drugs Context. 2019;8:212573.
  • 6
    Ruscio AM, Brown TA, Chiu WT, Sareen J, Stein MB, Kessler RC. Social fears and social phobia in the USA: results from the National Comorbidity Survey Replication. Psychol Med. 2008;38:15-28.
  • 7
    Bandelow B, Sagebiel A, Belz M, Görlich Y, Michaelis S, Wedekind D. Enduring effects of psychological treatments for anxiety disorders: meta-analysis of follow-up studies. Br J Psychiatry. 2018;212:333-8.
  • 8
    Williams T, Hattingh CJ, Kariuki CM, Tromp SA, van Balkom AJ, Ipser JC, et al. Pharmacotherapy for social anxiety disorder (SAnD). Cochrane Database Syst Rev. 2017;10;CD001206.
  • 9
    Jakubovski E, Johnson JA, Nasir M, Müller-Vahl K, Bloch MH. Systematic review and meta-analysis: dose-response curve of SSRIs and SNRIs in anxiety disorders. Depress Anxiety. 2019;36:198-212.
  • 10
    Bystritsky A, Khalsa SS, Cameron ME, Schiffman J. Current diagnosis and treatment of anxiety disorders. P T. 2013;38:30-57.
  • 11
    Morena M, Patel S, Bains JS, Hill MN. Neurobiological interactions between stress and the endocannabinoid system. Neuropsychopharmacology. 2016;41:80-102.
  • 12
    Wei D, Allsop S, Tye K, Piomelli D. Endocannabinoid signaling in the control of social behavior. Trends Neurosci. 2017;40:385-96.
  • 13
    Kagan J, Reznick JS, Clarke C, Snidman N, Garcia-Coll C. Behavioral inhibition to the unfamiliar. Child Dev. 1984;55:2212-25.
  • 14
    Schwartz CE, Snidman N, Kagan J. Adolescent social anxiety as an outcome of inhibited temperament in childhood. J Am Acad Child Adolesc Psychiatry. 1999;38:1008-15.
  • 15
    Prior M, Smart D, Sanson A, Oberklaid F. Does shy-inhibited temperament in childhood lead to anxiety problems in adolescence? J Am Acad Child Adolesc Psychiatry. 2000;39:461-8.
  • 16
    Biederman J, Hirshfeld-Becker DR, Rosenbaum JF, Hérot C, Friedman D, Snidman N, et al. Further evidence of association between behavioral inhibition and social anxiety in children. Am J Psychiatry. 2001;158:1673-9.
  • 17
    Hirshfeld-Becker DR, Biederman J, Henin A, Faraone SV, Davis S, Harrington K, et al. Behavioral inhibition in preschool children at risk is a specific predictor of middle childhood social anxiety: a five-year follow-up. J Dev Behav Pediatr. 2007;28:225-33.
  • 18
    Sandstrom A, Uher R, Pavlova B. Prospective association between childhood behavioral inhibition and anxiety: a meta-analysis. J Abnorm Child Psychol. 2020;48:57-66.
  • 19
    Clauss JA, Blackford JU. Behavioral inhibition and risk for developing social anxiety disorder: a meta-analytic study. J Am Acad Child Adolesc Psychiatry. 2012;51:1066-75.e1.
  • 20
    Fox NA, Buzzell GA, Morales S, Valadez EA, Wilson M, Henderson HA. Understanding the emergence of social anxiety in children with behavioral inhibition. Biol Psychiatry. 2021;89:681-9.
  • 21
    Bar-Haim Y, Fox NA, Benson B, Guyer AE, Williams A, Nelson EE, et al. Neural correlates of reward processing in adolescents with a history of inhibited temperament. Psychol Sci. 2009;20:1009-18.
  • 22
    Guyer AE, Nelson EE, Perez-Edgar K, Hardin MG, Roberson-Nay R, Monk CS, et al. Striatal functional alteration in adolescents characterized by early childhood behavioral inhibition. J Neurosci. 2006;26:6399-405.
  • 23
    Guyer AE, Benson B, Choate VR, Bar-Haim Y, Perez-Edgar K, Jarcho JM, et al. Lasting associations between early-childhood temperament and late-adolescent reward-circuitry response to peer feedback. Dev Psychopathol. 2014;26:229-43.
  • 24
    Helfinstein SM, Benson B, Perez-Edgar K, Bar-Haim Y, Detloff A, Pine DS, et al. Striatal responses to negative monetary outcomes differ between temperamentally inhibited and non-inhibited adolescents. Neuropsychologia. 2011;49:479-85.
  • 25
    Jarcho JM, Fox NA, Pine DS, Etkin A, Leibenluft E, Shechner T, et al. The neural correlates of emotion-based cognitive control in adults with early childhood behavioral inhibition. Biol Psychol. 2013;92:306-14.
  • 26
    Lamm C, Walker OL, Degnan KA, Henderson HA, Pine DS, McDermott JM, et al. Cognitive control moderates early childhood temperament in predicting social behavior in 7‐year‐old children: an ERP study. Dev Sci. 2014;17:667-81.
  • 27
    Pérez-Edgar K, Roberson-Nay R, Hardin MG, Poeth K, Guyer AE, Nelson EE, et al. Attention alters neural responses to evocative faces in behaviorally inhibited adolescents. Neuroimage. 2007;35:1538-46.
  • 28
    Schwartz CE, Wright CI, Shin LM, Kagan J, Rauch SL. Inhibited and uninhibited infants “grown up”: adult amygdalar response to novelty. Science. 2003;300:1952-3.
  • 29
    Schwartz CE, Kunwar PS, Greve DN, Kagan J, Snidman NC, Bloch RB. A phenotype of early infancy predicts reactivity of the amygdala in male adults. Mol Psychiatry. 2012;17:1042-50.
  • 30
    Kovner R, Oler JA, Kalin NH. Cortico-limbic interactions mediate adaptive and maladaptive responses relevant to psychopathology. Am J Psychiatry. 2019;176:987-99.
  • 31
    Tillfors M, Furmark T, Marteinsdottir I, Fredrikson M. Cerebral blood flow during anticipation of public speaking in social phobia: a PET study. Biol Psychiatry. 2002;52:1113-9.
  • 32
    Lorberbaum JP, Kose S, Johnson MR, Arana GW, Sullivan LK, Hamner MB, et al. Neural correlates of speech anticipatory anxiety in generalized social phobia. Neuroreport. 2004;15:2701-5.
  • 33
    Guyer AE, Lau JY, McClure-Tone EB, Parrish J, Shiffrin ND, Reynolds RC, et al. Amygdala and ventrolateral prefrontal cortex function during anticipated peer evaluation in pediatric social anxiety. Arch Gen Psychiatry. 2008;65:1303-12.
  • 34
    Blair K, Geraci M, Devido J, McCaffrey D, Chen G, Vythilingam M, et al. Neural response to self-and other referential praise and criticism in generalized social phobia. Arch Gen psychiatry. 2008;65:1176-84.
  • 35
    Stein MB, Goldin PR, Sareen J, Zorrilla LT, Brown GG. Increased amygdala activation to angry and contemptuous faces in generalized social phobia. Arch Gen Psychiatry. 2002;59:1027-34.
  • 36
    Yoon KL, Fitzgerald DA, Angstadt M, McCarron RA, Phan KL. Amygdala reactivity to emotional faces at high and low intensity in generalized social phobia: a 4-Tesla functional MRI study. Psychiatry Res. 2007;154:93-8.
  • 37
    Phan KL, Fitzgerald DA, Nathan PJ, Tancer ME. Association between amygdala hyperactivity to harsh faces and severity of social anxiety in generalized social phobia. Biol Psychiatry. 2006;59:424-9.
  • 38
    Evans KC, Wright CI, Wedig MM, Gold AL, Pollack MH, Rauch SL. A functional MRI study of amygdala responses to angry schematic faces in social anxiety disorder. Depress Anxiety. 2008;25:496-505.
  • 39
    Shah SG, Klumpp H, Angstadt M, Nathan PJ, Phan KL. Amygdala and insula response to emotional images in patients with generalized social anxiety disorder. J Psychiatry Neurosci. 2009;34:296-302.
  • 40
    Amir N, Klumpp H, Elias J, Bedwell JS, Yanasak N, Miller LS. Increased activation of the anterior cingulate cortex during processing of disgust faces in individuals with social phobia. Biol Psychiatry. 2005;57:975-81.
  • 41
    Schildkraut JJ. The catecholamine hypothesis of affective disorders: a review of supporting evidence. Am J Psychiatry. 1965;122:509-22.
  • 42
    Doruyter AG, Dupont P, Stein DJ, Lochner C, Warwick JM. Nuclear neuroimaging in social anxiety disorder: a review. J Nucl Med. 2018;59:1794-800.
  • 43
    Furmark T, Marteinsdottir I, Frick A, Heurling K, Tillfors M, Appel L, et al. Serotonin synthesis rate and the tryptophan hydroxylase-2: G-703T polymorphism in social anxiety disorder. J Psychopharmacol. 2016;30:1028-35.
  • 44
    Frick A, Åhs F, Engman J, Jonasson M, Alaie I, Björkstrand J, et al. Serotonin synthesis and reuptake in social anxiety disorder: a positron emission tomography study. JAMA Psychiatry. 2015;72:794-802.
  • 45
    Hjorth OR, Frick A, Gingnell M, Hoppe JM, Faria V, Hultberg S, et al. Expression and co-expression of serotonin and dopamine transporters in social anxiety disorder: a multitracer positron emission tomography study. Mol Psychiatry. 2019 Dec 10. doi: 10.1038/s41380-019-0618-7 Online ahead of print.
    » 10.1038/s41380-019-0618-7
  • 46
    van der Wee NJ, van Veen JF, Stevens H, van Vliet IM, van Rijk PP, Westenberg HG. Increased serotonin and dopamine transporter binding in psychotropic medication-naive patients with generalized social anxiety disorder shown by 123I-β-(4-iodophenyl)-tropane SPECT. J Nucl Med. 2008;49:757-63.
  • 47
    Lanzenberger RR, Mitterhauser M, Spindelegger C, Wadsak W, Klein N, Mien LK, et al. Reduced serotonin-1A receptor binding in social anxiety disorder. Biol Psychiatry. 2007;61:1081-9.
  • 48
    Tiihonen J, Kuikka J, Bergstrom K, Lepola U, Koponen H, Leinonen E. Dopamine reuptake site densities in patients with social phobia. Am J Psychiatry. 1997;154:239-42.
  • 49
    Schneier FR, Abi‐Dargham A, Martinez D, Slifstein M, Hwang DR, Liebowitz MR, et al. Dopamine transporters, D2 receptors, and dopamine release in generalized social anxiety disorder. Depress Anxiety. 2009;26:411-8.
  • 50
    Schneier FR, Liebowitz MR, Abi-Dargham A, Zea-Ponce Y, Lin SH, Laruelle M. Low dopamine D2 receptor binding potential in social phobia. Am J Psychiatry. 2000;157:457-9.
  • 51
    Faravelli C, Sauroc CL, Lelli L, Pietrini F, Lazzeretti L, Godini L, et al. The role of life events and HPA axis in anxiety disorders: a review. Curr Pharm Des. 2012;18:5663-74.
  • 52
    Hill MN, Tasker JG. Endocannabinoid signaling, glucocorticoid-mediated negative feedback, and regulation of the hypothalamic-pituitary-adrenal axis. Neuroscience. 2012;204:5-16.
  • 53
    Soravia LM, Heinrichs M, Aerni A, Maroni C, Schelling G, Ehlert U, et al. Glucocorticoids reduce phobic fear in humans. Proc Natl Acad Sci U S A. 2006;103:5585-90.
  • 54
    Radley J, Morilak D, Viau V, Campeau S. Chronic stress and brain plastistate: mechanisms underlying adaptive and maladaptive changes and implications for stress-related CNS disorders. Neurosci Biobehav Rev. 2015;58:79-91.
  • 55
    Condren RM, O’Neill A, Ryan MC, Barrett P, Thakore JH. HPA axis response to a psychological stressor in generalised social phobia. Psychoneuroendocrinology. 2002;27:693-703.
  • 56
    van West D, Claes S, Sulon J, Deboutte D. Hypothalamic-pituitary-adrenal reactivity in prepubertal children with social phobia. J Affect Disord. 2008;111:281-90.
  • 57
    Yoon KL, Joormann J. Stress reactivity in social anxiety disorder with and without comorbid depression. J Abnorm Psychol. 2012;121:250-5.
  • 58
    Martel FL, Hayward C, Lyons DM, Sanborn K, Varady S, Schatzberg AF. Salivary cortisol levels in socially phobic adolescent girls. Depress Anxiety. 1999;10:25-7.
  • 59
    Klumbies E, Braeuer D, Hoyer J, Kirschbaum C. The reaction to social stress in social phobia: discordance between physiological and subjective parameters. PLoS One. 2014;9:e105670.
  • 60
    Losiak W, Blaut A, Klosowska J, Slowik N. Social anxiety, affect, cortisol response and performance on a speech task. Psychopathology. 2016;49:24-30.
  • 61
    Rose RM. Endocrine responses to stressful psychological events. Psychiatr Clin North Am. 1980;3:251-76.
  • 62
    Pertwee RG. Ligands that target cannabinoid receptors in the brain: from THC to anandamide and beyond. Addict Biol. 2008;13:147-59.
  • 63
    Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bonner TI. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature. 1990;346:561-4.
  • 64
    Herkenham M, Lynn AB, Johnson MR, Melvin LS, de Costa BR, Rice KC. Characterization and localization of cannabinoid receptors in rat brain: a quantitative in vitro autoradiographic study. J Neurosci. 1991;11:563-83.
  • 65
    Galiègue S, Mary S, Marchand J, Dussossoy D, Carrière D, Carayon P, et al. Expression of central and peripheral cannabinoid receptors in human immune tissues and leukocyte subpopulations. Eur J Biochem. 1995;232:54-61.
  • 66
    Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a peripheral receptor for cannabinoids. Nature. 1993;365:61-5.
  • 67
    Ahn K, McKinney MK, Cravatt BF. Enzymatic pathways that regulate endocannabinoid signaling in the nervous system. Chem Rev. 2008;108:1687-707.
  • 68
    Devane WA, Hanus L, Breuer A, Pertwee RG, Stevenson LA, Griffin G, et al. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science. 1992;258:1946-9.
  • 69
    Sugiura T, Kondo S, Sukagawa A, Nakane S, Shinoda A, Itoh K, et al. 2-arachidonoylgylcerol: a possible endogenous cannabinoid receptor ligand in brain. Biochem Biophys Res Commun. 1995;215:89-97.
  • 70
    Pertwee RG. The therapeutic potential of drugs that target cannabinoid receptors or modulate the tissue levels or actions of endocannabinoids. AAPS J. 2005;7:E625-54.
  • 71
    Cravatt BF, Giang DK, Mayfield SP, Boger DL, Lerner RA, Gilula NB. Molecular characterization of an enzyme that degrades neuromodulatory fatty-acid amides. Nature. 1996;384:83-7.
  • 72
    Dinh TP, Carpenter D, Leslie FM, Freund TF, Katona I, Sensi SL, et al. Brain monoglyceride lipase participating in endocannabinoid inactivation. Proc Natl Acad Sci U S A. 2002;99:10819-24.
  • 73
    Connor JP, Stjepanović D, Le Foll B, Hoch E, Budney AJ, Hall WD. Cannabis use and cannabis use disorder. Nat Rev Dis Primers. 2021;7:16.
  • 74
    Crippa JA, Zuardi AW, Martín‐Santos R, Bhattacharyya S, Atakan Z, McGuire P, et al. Cannabis and anxiety: a critical review of the evidence. Hum Psychopharmacol. 2009;24:515-23.
  • 75
    D’Souza DC, Perry E, MacDougall L, Ammerman Y, Cooper T, Wu YT, et al. The psychotomimetic effects of intravenous delta-9-tetrahydrocannabinol in healthy individuals: implications for psychosis. Neuropsychopharmacology. 2004;29:1558-72.
  • 76
    Hunault CC, Böcker KB, Stellato RK, Kenemans JL, de Vries I, Meulenbelt J. Acute subjective effects after smoking joints containing up to 69 mg Δ9-tetrahydrocannabinol in recreational users: a randomized, crossover clinical trial. Psychopharmacology (Berl). 2014;231:4723-33.
  • 77
    Rubino T, Sala M, Vigano D, Braida D, Castiglioni C, Limonta V, et al. Cellular mechanisms underlying the anxiolytic effect of low doses of peripheral Δ 9-tetrahydrocannabinol in rats. Neuropsychopharmacology. 2007;32:2036-45.
  • 78
    Rubino T, Guidali C, Vigano D, Realini N, Valenti M, Massi P, et al. CB1 receptor stimulation in specific brain areas differently modulate anxiety-related behaviour. Neuropharmacology. 2008;54:151-60.
  • 79
    Devane WA, Dysarz FA 3rd, Johnson MR, Melvin LS, Howlett AC. Determination and characterization of a cannabinoid receptor in rat brain. Mol Pharmacol. 1988;34:605-13.
  • 80
    Micale V, Di Marzo V, Sulcova A, Wotjak CT, Drago F. Endocannabinoid system and mood disorders: priming a target for new therapies. Pharmacol Ther. 2013;138:18-37.
  • 81
    Lin HC, Mao SC, Gean PW. Effects of intra-amygdala infusion of CB1 receptor agonists on the reconsolidation of fear-potentiated startle. Learn Mem. 2006;13:316-21.
  • 82
    Campolongo P, Roozendaal B, Trezza V, Hauer D, Schelling G, McGaugh JL, et al. Endocannabinoids in the rat basolateral amygdala enhance memory consolidation and enable glucocorticoid modulation of memory. Proc Natl Acad Sci U S A. 2009;106:4888-93.
  • 83
    Marsicano G, Wotjak CT, Azad SC, Bisogno T, Rammes G, Cascio MG, et al. The endogenous cannabinoid system controls extinction of aversive memories. Nature. 2002;418:530-4.
  • 84
    Tan H, Lauzon NM, Bishop SF, Bechard MA, Laviolette SR. Integrated cannabinoid CB1 receptor transmission within the amygdala-prefrontal cortical pathway modulates neuronal plastistate and emotional memory encoding. Cereb Cortex. 2010;20:1486-96.
  • 85
    Tan H, Lauzon NM, Bishop SF, Chi N, Bechard M, Laviolette SR. Cannabinoid transmission in the basolateral amygdala modulates fear memory formation via functional inputs to the prelimbic cortex. J Neurosci. 2011;31:5300-12.
  • 86
    Hill MN, McLaughlin RJ, Bingham B, Shrestha L, Lee TT, Gray JM, et al. Endogenous cannabinoid signaling is essential for stress adaptation. Proc Natl Acad Sci U S A. 2010;107:9406-11.
  • 87
    Bortolato M, Mangieri RA, Fu J, Kim JH, Arguello O, Duranti A, et al. Antidepressant-like activity of the fatty acid amide hydrolase inhibitor URB597 in a rat model of chronic mild stress. Biol Psychiatry. 2007;62:1103-10.
  • 88
    Rademacher DJ, Meier SE, Shi L, Ho WS, Jarrahian A, Hillard CJ. Effects of acute and repeated restraint stress on endocannabinoid content in the amygdala, ventral striatum, and medial prefrontal cortex in mice. Neuropharmacology. 2008;54:108-16.
  • 89
    Hill MN, Kumar SA, Filipski SB, Iverson M, Stuhr KL, Keith JM, et al. Disruption of fatty acid amide hydrolase activity prevents the effects of chronic stress on anxiety and amygdalar microstructure. Mol Psychiatry. 2013;18:1125-35.
  • 90
    Gray JM, Vecchiarelli HA, Morena M, Lee TT, Hermanson DJ, Kim AB, et al. Corticotropin-releasing hormone drives anandamide hydrolysis in the amygdala to promote anxiety. J Neurosci. 2015;35:3879-92.
  • 91
    Patel S, Roelke CT, Rademacher DJ, Hillard CJ. Inhibition of restraint stress‐induced neural and behavioural activation by endogenous cannabinoid signalling. Eur J Neurosci. 2005;21:1057-69.
  • 92
    Bedse G, Hill MN, Patel S. 2-arachidonoylglycerol modulation of anxiety and stress adaptation: from grass roots to novel therapeutics. Biol Psychiatry. 2020;88:520-30.
  • 93
    Bedse G, Hartley ND, Neale E, Gaulden AD, Patrick TA, Kingsley PJ, et al. Functional redundancy between canonical endocannabinoid signaling systems in the modulation of anxiety. Biol Psychiatry. 2017;82:488-99.
  • 94
    Busquets-Garcia A, Puighermanal E, Pastor A, de la Torre R, Maldonado R, Ozaita A. Differential role of anandamide and 2-arachidonoylglycerol in memory and anxiety-like responses. Biol Psychiatry. 2011;70:479-86.
  • 95
    Sciolino NR, Zhou W, Hohmann AG. Enhancement of endocannabinoid signaling with JZL184, an inhibitor of the 2-arachidonoylglycerol hydrolyzing enzyme monoacylglycerol lipase, produces anxiolytic effects under conditions of high environmental aversiveness in rats. Pharmacol Res. 2011;64:226-34.
  • 96
    Aliczki M, Zelena D, Mikics E, Varga ZK, Pinter O, Bakos NV, et al. Monoacylglycerol lipase inhibition-induced changes in plasma corticosterone levels, anxiety and locomotor activity in male CD1 mice. Horm Behav. 2013;63:752-8.
  • 97
    Kinsey SG, O’Neal ST, Long JZ, Cravatt BF, Lichtman AH. Inhibition of endocannabinoid catabolic enzymes elicits anxiolytic-like effects in the marble burying assay. Pharmacol Biochem Behav. 2011;98:21-7.
  • 98
    Bedse G, Bluett RJ, Patrick TA, Romness NK, Gaulden AD, Kingsley PJ, et al. Therapeutic endocannabinoid augmentation for mood and anxiety disorders: comparative profiling of FAAH, MAGL and dual inhibitors. Transl Psychiatry. 2018;8:92.
  • 99
    Lim J, Igarashi M, Jung KM, Butini S, Campiani G, Piomelli D. Endocannabinoid modulation of predator stress-induced long-term anxiety in rats. Neuropsychopharmacology. 2016;41:1329-39.
  • 100
    Mitchell PB, Morris MJ. Depression and anxiety with rimonabant. Lancet. 2007;370:1671-2.
  • 101
    Dlugos A, Childs E, Stuhr KL, Hillard CJ, de Wit H. Acute stress increases circulating anandamide and other N-acylethanolamines in healthy humans. Neuropsychopharmacology. 2012;37:2416-27.
  • 102
    Hill MN, Miller GE, Carrier EJ, Gorzalka BB, Hillard CJ. Circulating endocannabinoids and N-acyl ethanolamines are differentially regulated in major depression and following exposure to social stress. Psychoneuroendocrinology. 2009;34:1257-62.
  • 103
    Hill MN, Bierer LM, Makotkine I, Golier JA, Galea S, McEwen BS, et al. Reductions in circulating endocannabinoid levels in individuals with post-traumatic stress disorder following exposure to the World Trade Center attacks. Psychoneuroendocrinology. 2013;38:2952-61.
  • 104
    Sipe JC, Chiang K, Gerber AL, Beutler E, Cravatt BF. A missense mutation in human fatty acid amide hydrolase associated with problem drug use. Proc Natl Acad Sci U S A. 2002;99:8394-9.
  • 105
    Chiang KP, Gerber AL, Sipe JC, Cravatt BF. Reduced cellular expression and activity of the P129T mutant of human fatty acid amide hydrolase: evidence for a link between defects in the endocannabinoid system and problem drug use. Hum Mol Genet. 2004;13:2113-9.
  • 106
    Boileau I, Tyndale RF, Williams B, Mansouri E, Westwood DJ, Le Foll B, et al. The fatty acid amide hydrolase C385A variant affects brain binding of the positron emission tomography tracer [11C] CURB. J Cereb Blood Flow Metab. 2015;35:1237-40.
  • 107
    Hariri AR, Gorka A, Hyde LW, Kimak M, Halder I, Ducci F, et al. Divergent effects of genetic variation in endocannabinoid signaling on human threat-and reward-related brain function. Biol Psychiatry. 2009;66:9-16.
  • 108
    Gunduz-Cinar O, MacPherson KP, Cinar R, Gamble-George J, Sugden K, Williams B, et al. Convergent translational evidence of a role for anandamide in amygdala-mediated fear extinction, threat processing and stress-reactivity. Mol Psychiatry. 2013;18:813-23.
  • 109
    Dincheva I, Drysdale AT, Hartley CA, Johnson DC, Jing D, King EC, et al. FAAH genetic variation enhances fronto-amygdala function in mouse and human. Nat Commun. 2015;6:6395.
  • 110
    Green DG, Kim J, Kish SJ, Tyndale RF, Hill MN, Strafella AP, et al. Fatty acid amide hydrolase binding is inversely correlated with amygdalar functional connectivity: a combined positron emission tomography and magnetic resonance imaging study in healthy individuals. J Psychiatry Neurosci. 2021;46:E238-46.
  • 111
    Spohrs J, Ulrich M, Grön G, Prost M, Plener PL, Fegert JM, et al. Fear extinction learning and anandamide: an fMRI study in healthy humans. Transl Psychiatry. 2021;11:161.
  • 112
    Habib AM, Okorokov AL, Hill MN, Bras JT, Lee MC, Li S, et al. Microdeletion in a FAAH pseudogene identified in a patient with high anandamide concentrations and pain insensitivity. Br J Anaesth. 2019;123:e249-53.
  • 113
    Trezza V, Vanderschuren LJ. Bidirectional cannabinoid modulation of social behavior in adolescent rats. Psychopharmacology (Berl). 2008;197:217-27.
  • 114
    Trezza V, Damsteegt R, Manduca A, Petrosino S, Van Kerkhof LW, Pasterkamp RJ, et al. Endocannabinoids in amygdala and nucleus accumbens mediate social play reward in adolescent rats. J Neurosci. 2012;32:14899-908.
  • 115
    Gonczarowska N, Tomaz C, Caixeta FV, Malcher-Lopes R, Barros M, Nishijo H, et al. CB1 receptor antagonism in capuchin monkeys alters social interaction and aversive memory extinction. Psychopharmacology (Berl). 2019;236:3413-9.
  • 116
    Manduca A, Servadio M, Campolongo P, Palmery M, Trabace L, Vanderschuren LJ, et al. Strain- and context-dependent effects of the anandamide hydrolysis inhibitor URB597 on social behavior in rats. Eur Neuropsychopharmacol. 2014;24:1337-48.
  • 117
    Matricon J, Seillier A, Giuffrida A. Distinct neuronal activation patterns are associated with PCP-induced social withdrawal and its reversal by the endocannabinoid-enhancing drug URB597. Neurosci Res. 2016;110:49-58.
  • 118
    Schneider P, Bindila L, Schmahl C, Bohus M, Meyer-Lindenberg A, Lutz B, et al. Adverse social experiences in adolescent rats result in enduring effects on social competence, pain sensitivity and endocannabinoid signaling. Front Behav Neurosci. 2016;10:203.
  • 119
    Schmidt ME, Liebowitz MR, Stein MB, Grunfeld J, Van Hove I, Simmons WK, et al. The effects of inhibition of fatty acid amide hydrolase (FAAH) by JNJ-42165279 in social anxiety disorder: a double-blind, randomized, placebo-controlled proof-of-concept study. Neuropsychopharmacology. 2021;46:1004-10.
  • 120
    Mayo LM, Asratian A, Lindé J, Morena M, Haataja R, Hammar V, et al. Elevated anandamide, enhanced recall of fear extinction, and attenuated stress responses following inhibition of fatty acid amide hydrolase: a randomized, controlled experimental medicine trial. Biol Psychiatry. 2020;87:538-47.
  • 121
    Paulus MP, Stein MB, Simmons AN, Risbrough VB, Halter R, Chaplan SR. The effects of FAAH inhibition on the neural basis of anxiety-related processing in healthy male subjects: a randomized clinical trial. Neuropsychopharmacology. 2021;46:1011-9.
  • 122
    Boggs DL, Nguyen JD, Morgenson D, Taffe MA, Ranganathan M. Clinical and preclinical evidence for functional interactions of cannabidiol and Δ 9-tetrahydrocannabinol. Neuropsychopharmacology. 2018;43:142-54.
  • 123
    McPartland JM, Duncan M, Di Marzo V, Pertwee RG. Are cannabidiol and Δ9‐tetrahydrocannabivarin negative modulators of the endocannabinoid system? A systematic review. Br J Pharmacol. 2015;172:737-53.
  • 124
    Russo EB, Burnett A, Hall B, Parker KK. Agonistic properties of cannabidiol at 5-HT1a receptors. Neurochem Res. 2005;30:1037-43.
  • 125
    Fogaça MV, Reis FM, Campos AC, Guimaraes FS. Effects of intra-prelimbic prefrontal cortex injection of cannabidiol on anxiety-like behavior: involvement of 5HT1A receptors and previous stressful experience. Eur Neuropsychopharmacol. 2014;24:410-9.
  • 126
    Campos AC, Guimarães FS. Evidence for a potential role for TRPV1 receptors in the dorsolateral periaqueductal gray in the attenuation of the anxiolytic effects of cannabinoids. Prog Neuropsychopharmacol Biol Psychiatry. 2009;33:1517-21.
  • 127
    Leweke FM, Piomelli D, Pahlisch F, Muhl D, Gerth CW, Hoyer C, et al. Cannabidiol enhances anandamide signaling and alleviates psychotic symptoms of schizophrenia. Transl Psychiatry. 2012;2:e94.
  • 128
    Blessing EM, Steenkamp MM, Manzanares J, Marmar CR. Cannabidiol as a potential treatment for anxiety disorders. Neurotherapeutics. 2015;12:825-36.
  • 129
    Laprairie RB, Bagher AM, Kelly ME, Denovan-Wright EM. Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor. Br J Pharmacol. 2015;172:4790-805.
  • 130
    Watanabe K, Ogi H, Nakamura S, Kayano Y, Matsunaga T, Yoshimura H, et al. Distribution and characterization of anandamide amidohydrolase in mouse brain and liver. Life Sci. 1998;62:1223-9.
  • 131
    De Petrocellis L, Ligresti A, Moriello AS, Allarà M, Bisogno T, Petrosino S, et al. Effects of cannabinoids and cannabinoid‐enriched Cannabis extracts on TRP channels and endocannabinoid metabolic enzymes. Br J Pharmacol. 2011;163:1479-94.
  • 132
    Bisogno T, Hanuš L, De Petrocellis L, Tchilibon S, Ponde DE, Brandi I, et al. Molecular targets for cannabidiol and its synthetic analogues: effect on vanilloid VR1 receptors and on the cellular uptake and enzymatic hydrolysis of anandamide. Br J Pharmacol. 2001;134:845-52.
  • 133
    Massi P, Valenti M, Vaccani A, Gasperi V, Perletti G, Marras E, et al. 5‐Lipoxygenase and anandamide hydrolase (FAAH) mediate the antitumor activity of cannabidiol, a non‐psychoactive cannabinoid. J Neurochem. 2008;104:1091-100.
  • 134
    Fogaça MV, Campos AC, Coelho LD, Duman RS, Guimarães FS. The anxiolytic effects of cannabidiol in chronically stressed mice are mediated by the endocannabinoid system: role of neurogenesis and dendritic remodeling. Neuropharmacology. 2018;135:22-33.
  • 135
    de Filippis D, Iuvone T, D’amico A, Esposito G, Steardo L, Herman AG, et al. Effect of cannabidiol on sepsis‐induced motility disturbances in mice: involvement of CB1 receptors and fatty acid amide hydrolase. Neurogastroenterol Motil. 2008;20:919-27.
  • 136
    Almeida V, Levin R, Peres FF, Niigaki ST, Calzavara MB, Zuardi AW, et al. Cannabidiol exhibits anxiolytic but not antipsychotic property evaluated in the social interaction test. Prog Neuropsychopharmacol Biol Psychiatry. 2013;41:30-5.
  • 137
    Campos AC, de Paula Soares V, Carvalho MC, Ferreira FR, Vicente MA, Brandão ML, et al. Involvement of serotonin-mediated neurotransmission in the dorsal periaqueductal gray matter on cannabidiol chronic effects in panic-like responses in rats. Psychopharmacology (Berl). 2013;226:13-24.
  • 138
    Guimarães FS, Chiaretti TM, Graeff FG, Zuardi AW. Antianxiety effect of cannabidiol in the elevated plus-maze. Psychopharmacology (Berl). 1990;100:558-9.
  • 139
    Resstel LB, Tavares RF, Lisboa SF, Joca SR, Corrêa FM, Guimarães FS. 5‐HT1A receptors are involved in the cannabidiol‐induced attenuation of behavioural and cardiovascular responses to acute restraint stress in rats. Br J Pharmacol. 2009;156:181-8.
  • 140
    Linares IM, Zuardi AW, Pereira LC, Queiroz RH, Mechoulam R, Guimarães FS, et al. Cannabidiol presents an inverted U-shaped dose-response curve in a simulated public speaking test. Braz J Psychiatry. 2019;41:9-14.
  • 141
    Nazario LR, Antonioli RJ, Capiotti KM, Hallak JE, Zuardi AW, Crippa JA, et al. Reprint of “caffeine protects against memory loss induced by high and non-anxiolytic dose of cannabidiol in adult zebrafish (Danio rerio). Pharmacol Biochem Behav. 2015;139 Pt B:134-40.
  • 142
    Zanelati TV, Biojone C, Moreira FA, Guimarães FS, Joca SR. Antidepressant-like effects of cannabidiol in mice: Possible involvement of 5-HT 1A receptors. Br J Pharmacol. 2010;159:122-8.
  • 143
    Casarotto PC, Gomes FV, Resstel LB, Guimarães FS. Cannabidiol inhibitory effect on marble-burying behaviour: Involvement of CB1 receptors. Behav Pharmacol. 2010;21:353-8.
  • 144
    Levin R, Peres FF, Almeida V, Calzavara MB, Zuardi AW, Hallak JEC, et al. Effects of cannabinoid drugs on the deficit of prepulse inhibition of startle in an animal model of schizophrenia: the SHR strain. Front Pharmacol. 2014;5:10.
  • 145
    Peres FF, Levin R, Suiama MA, Diana MC, Gouvêa DA, Almeida V, et al. Cannabidiol prevents motor and cognitive impairments induced by reserpine in rats. Front Pharmacol. 2016;7:343.
  • 146
    Elmes MW, Kaczocha M, Berger WT, Leung K, Ralph BP, Wang L, et al. Fatty acid-binding proteins (FABPs) are intracellular carriers for Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD). J Biol Chem. 2015;290:8711-21.
  • 147
    Wilson AA, Garcia A, Parkes J, Houle S, Tong J, Vasdev N. [11C]CURB: evaluation of a novel radiotracer for imaging fatty acid amide hydrolase by positron emission tomography. Nucl Med Biol. 2011;38:247-53.
  • 148
    Crippa JA, Derenusson GN, Ferrari TB, Wichert-Ana L, Duran FL, Martin-Santos R, et al. Neural basis of anxiolytic effects of cannabidiol (CBD) in generalized social anxiety disorder: a preliminary report. J Psychopharmacol. 2011;25:121-30.
  • 149
    Bergamaschi MM, Queiroz RH, Chagas MH, de Oliveira DC, De Martinis BS, Kapczinski F, et al. Cannabidiol reduces the anxiety induced by simulated public speaking in treatment-naive social phobia patients. Neuropsychopharmacology. 2011;36:1219-26.
  • 150
    Masataka N. Anxiolytic effects of repeated cannabidiol treatment in teenagers with social anxiety disorders. Front Psychol. 2019;10:2466.
  • 151
    McMaster University. Cannabidiol for the treatment of anxiety disorders: an 8-week pilot study [Internet]. 2018 [cited 2021 20 Jul]. clinicaltrials.gov/ct2/show/study/NCT03549819
    » clinicaltrials.gov/ct2/show/study/NCT03549819
  • 152
    Marcin MS, Nemeroff CB. The neurobiology of social anxiety disorder: the relevance of fear and anxiety. Acta Psychiatr Scand Suppl. 2003;108:51-64.
  • 153
    Faravelli C, Lo Sauro C, Godini L, Lelli L, Benni L, Pietrini F, et al. Childhood stressful events, HPA axis and anxiety disorders. World J psychiatry. 2012;2:13-25.
  • 154
    Page ME, Oropeza VC, Sparks SE, Qian Y, Menko AS, Van Bockstaele EJ. Repeated cannabinoid administration increases indices of noradrenergic activity in rats. Pharmacol Biochem Behav. 2007;86:162-8.
  • 155
    Page ME, Oropeza VC, Van Bockstaele EJ. Local administration of a cannabinoid agonist alters norepinephrine efflux in the rat frontal cortex. Neurosci Lett. 2008;431:1-5.
  • 156
    Bambico FR, Katz N, Debonnel G, Gobbi G. Cannabinoids elicit antidepressant-like behavior and activate serotonergic neurons through the medial prefrontal cortex. J Neurosci. 2007;27:11700-11.
  • 157
    Gobbi G, Bambico FR, Mangieri R, Bortolato M, Campolongo P, Solinas M, et al. Antidepressant-like activity and modulation of brain monoaminergic transmission by blockade of anandamide hydrolysis. Proc Natl Acad Sci U S A. 2005;102:18620-5.
  • 158
    Cassano T, Gaetani S, Macheda T, Laconca L, Romano A, Morgese MG, et al. Evaluation of the emotional phenotype and serotonergic neurotransmission of fatty acid amide hydrolase-deficient mice. Psychopharmacology (Berl). 2011;214:465-76.
  • 159
    Micale V, Drago F. Endocannabinoid system, stress and HPA axis. Eur J Pharmacol. 2018;834:230-9.
  • 160
    Gray JM, Wilson CD, Lee TT, Pittman QJ, Deussing JM, Hillard CJ, et al. Sustained glucocorticoid exposure recruits cortico-limbic CRH signaling to modulate endocannabinoid function. Psychoneuroendocrinology. 2016;66:151-8.
  • 161
    Hill MN, McLaughlin RJ, Morrish AC, Viau V, Floresco SB, Hillard CJ, et al. Suppression of amygdalar endocannabinoid signaling by stress contributes to activation of the hypothalamic-pituitary-adrenal axis. Neuropsychopharmacology. 2009;34:2733-45.
  • 162
    Di S, Malcher-Lopes R, Halmos KC, Tasker JG. Nongenomic glucocorticoid inhibition via endocannabinoid release in the hypothalamus: a fast feedback mechanism. J Neurosci. 2003;23:4850-7.
  • 163
    Wei D, Lee D, Cox CD, Karsten CA, Penagarikano O, Geschwind DH, et al. Endocannabinoid signaling mediates oxytocin-driven social reward. Proc Natl Acad Sci U S A. 2015;112:14084-9.
  • 164
    Haller J, Varga B, Ledent C, Freund TF. CB1 cannabinoid receptors mediate anxiolytic effects: convergent genetic and pharmacological evidence with CB1-specific agents. Behav Pharmacol. 2004;15:299-304.
  • 165
    Hill MN, Gorzalka BB. Enhancement of anxiety-like responsiveness to the cannabinoid CB1 receptor agonist HU-210 following chronic stress. Eur J Pharmacol. 2004;499:291-5.
  • 166
    Patel S, Hillard CJ. Pharmacological evaluation of cannabinoid receptor ligands in a mouse model of anxiety: further evidence for an anxiolytic role for endogenous cannabinoid signaling. J Pharmacol Exp Ther. 2006;318:304-11.
  • 167
    Moreira FA, Grieb M, Lutz B. Central side-effects of therapies based on CB1 cannabinoid receptor agonists and antagonists: focus on anxiety and depression. Best Pract Res Clin Endocrinol Metab. 2009;23:133-44.
  • 168
    Loureiro M, Kramar C, Renard J, Rosen LG, Laviolette SR. Cannabinoid transmission in the hippocampus activates nucleus accumbens neurons and modulates reward and aversion-related emotional salience. Biol Psychiatry. 2016;80:216-25.
  • 169
    Trezza V, Vanderschuren LJ. Cannabinoid and opioid modulation of social play behavior in adolescent rats: differential behavioral mechanisms. Eur Neuropsychopharmacol. 2008;18:519-30.
  • 170
    Schneider M, Schömig E, Leweke FM. Acute and chronic cannabinoid treatment differentially affects recognition memory and social behavior in pubertal and adult rats. Addict Biol. 2008;13:345-57.
  • 171
    Realini N, Vigano’ D, Guidali C, Zamberletti E, Rubino T, Parolaro D. Chronic URB597 treatment at adulthood reverted most depressive-like symptoms induced by adolescent exposure to THC in female rats. Neuropharmacology. 2011;60:235-43.
  • 172
    Seillier A, Advani T, Cassano T, Hensler JG, Giuffrida T. Inhibition of fatty-acid amide hydrolase and CB1 receptor antagonism differentially affect behavioural responses in normal and PCP-treated rats. Int J Neuropsychopharmacol. 2010;13:373-86.
  • 173
    Seillier A, Martinez AA, Giuffrida A. Phencyclidine-induced social withdrawal results from deficient stimulation of cannabinoid CB1 receptors: implications for schizophrenia. Neuropsychopharmacology. 2013;38:1816-24.
  • 174
    Melancia F, Schiavi S, Servadio M, Cartocci V, Campolongo P, Palmery M, et al. Sex-specific autistic endophenotypes induced by prenatal exposure to valproic acid involve anandamide signalling. Br J Pharmacol. 2018;175:3699-712.
  • 175
    Manduca A, Morena M, Campolongo P, Servadio M, Palmery M, Trabace L, et al. Distinct roles of the endocannabinoids anandamide and 2-arachidonoylglycerol in social behavior and emotionality at different developmental ages in rats. Eur Neuropsychopharmacol. 2015;25:1362-74.
  • 176
    Schiavi S, Manduca A, Segatto M, Campolongo P, Pallottini V, Vanderschuren LJ, et al. Unidirectional opioid-cannabinoid cross-tolerance in the modulation of social play behavior in rats. Psychopharmacology (Berl). 2019;236:2557-68.
  • 177
    Moise AM, Eisenstein SA, Astarita G, Piomelli D, Hohmann AG. An endocannabinoid signaling system modulates anxiety-like behavior in male Syrian hamsters. Psychopharmacology (Berl). 2008;200:333-46.
  • 178
    Qin M, Zeidler Z, Moulton K, Krych L, Xia Z, Smith CB. Endocannabinoid-mediated improvement on a test of aversive memory in a mouse model of fragile X syndrome. Behav Brain Res. 2015;291:164-71.
  • 179
    Seillier A, Giuffrida A. The cannabinoid transporter inhibitor OMDM-2 reduces social interaction: Further evidence for transporter-mediated endocannabinoid release. Neuropharmacology. 2018;130:1-9.
  • 180
    Shahbazi F, Grandi V, Banerjee A, Trant JF. Cannabinoids and cannabinoid receptors: the story so far. iScience. 2020;23:101301.
  • 181
    Doenni VM, Gray JM, Song CM, Patel S, Hill MN, Pittman QJ. Deficient adolescent social behavior following earaly-life inflammation is ameliorated by augmentation of anandamide signaling. Brain Behav Immun. 2016;58:237-47.
  • 182
    Sam AH, Salem V, Ghatei MA. Rimonabant: from RIO to Ban. J Obes. 2011;2011:432607.
  • 183
    Litvin Y, Phan A, Hill MN, Pfaff DW, McEwen BS. CB1 receptor signaling regulates social anxiety and memory. Genes Brain Behav. 2013;12:479-89.
  • 184
    Bogáthy E, Kostyalik D, Petschner P, Vas S, Bagdy G. Blockade of serotonin 2C receptors with SB-242084 moderates reduced locomotor activity and rearing by cannabinoid 1 receptor antagonist AM-251. Pharmacology. 2019;103:151-8.
  • 185
    Gueye AB, Pryslawsky Y, Trigo JM, Poulia N, Delis F, Antoniou K, et al. The CB1 neutral antagonist AM4113 retains the therapeutic efficacy of the inverse agonist rimonabant for nicotine dependence and weight loss with better psychiatric tolerability. Int J Neuropsychopharmacol. 2016;19:pyw068.
  • 186
    Sink KS, Segovia KN, Sink J, Randall PA, Collins LE, Correa M, et al. Potential anxiogenic effects of cannabinoid CB1 receptor antagonists/inverse agonists in rats: comparisons between AM4113, AM251, and the benzodiazepine inverse agonist FG-7142. Eur Neuropsychopharmacol. 2010;20:112-22.
  • 187
    O’Brien LD, Wills KL, Segsworth B, Dashney B, Rock EM, Limebeer CL, et al. Effect of chronic exposure to rimonabant and phytocannabinoids on anxiety-like behavior and saccharin palatability. Pharmacol Biochem Behav. 2013;103:597-602.
  • 188
    Jutkiewicz EM, Makriyannis A, Vemuri K, Bergman J. Pro-depressant-like effects of CB1 receptor inverse agonists/antagonists in male Sprague-Dawley rats. FASEB J. 2010;24:581.7.
  • 189
    Puighermanal E, Busquets-Garcia A, Maldonado R, Ozaita A. Cellular and intracellular mechanisms involved in the cognitive impairment of cannabinoids. Philos Trans R Soc Lond B Biol Sci. 2012;367:3254-63.
  • 190
    Wiskerke J, Pattij T, Schoffelmeer AN, De Vries TJ. The role of CB1 receptors in psychostimulant addiction. Addict Biol. 2008;13:225-38.
  • 191
    Parsons LH, Hurd YL. Endocannabinoid signalling in reward and addiction. Nat Rev Neurosci. 2015;16:579-94.
  • 192
    Mulpuri Y, Marty VN, Munier JJ, Mackie K, Schmidt BL, Seltzman HH, et al. Synthetic peripherally-restricted cannabinoid suppresses chemotherapy-induced peripheral neuropathy pain symptoms by CB1 receptor activation. Neuropharmacology. 2018;139:85-97.
  • 193
    Slivicki RA, Xu Z, Kulkarni PM, Pertwee RG, Mackie K, Thakur GA, et al. Positive allosteric modulation of cannabinoid receptor type 1 suppresses pathological pain without producing tolerance or dependence. Biol Psychiatry. 2018;84:722-33.
  • 194
    Stasiulewicz A, Znajdek K, Grudzień M, Pawiński T, Sulkowska JI. A guide to targeting the endocannabinoid system in drug design. Int J Mol Sci. 2020;21:2778.
  • 195
    Rubino T, Realini N, Castiglioni C, Guidali C, Vigano D, Marras E, et al. Role in anxiety behavior of the endocannabinoid system in the prefrontal cortex. Cereb Cortex. 2008;18:1292-301.
  • 196
    Befort K. Interactions of the opioid and cannabinoid systems in reward: Insights from knockout studies. Front Pharmacol. 2015;6:6.
  • 197
    Carvalho AF, Van Bockstaele EJ. Cannabinoid modulation of noradrenergic circuits: implications for psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry. 2012;38:59-67.
  • 198
    Buisseret B, Alhouayek M, Guillemot-Legris O, Muccioli GG. Endocannabinoid and prostanoid crosstalk in pain. Trends Mol Med. 2019;25:882-96.
  • 199
    Haj-Dahmane S, Shen RY. Modulation of the serotonin system by endocannabinoid signaling. Neuropharmacology. 2011;61:414-20.
  • 200
    Marcellino D, Carriba P, Filip M, Borgkvist A, Frankowska M, Bellido I, et al. Antagonistic cannabinoid CB1/dopamine D2 receptor interactions in striatal CB1/D2 heteromers. A combined neurochemical and behavioral analysis. Neuropharmacology. 2008;54:815-23.
  • 201
    Rodríguez-Muñoz M, Sánchez-Blázquez P, Merlos M, Garzón-Niño J. Endocannabinoid control of glutamate NMDA receptors: the therapeutic potential and consequences of dysfunction. Oncotarget. 2016;7:55840.
  • 202
    Musella A, Fresegna D, Rizzo FR, Gentile A, Bullitta S, De Vito F, et al. A novel crosstalk within the endocannabinoid system controls GABA transmission in the striatum. Sci Rep. 2017;7:7363.
  • 203
    Michopoulos V, Powers A, Gillespie CF, Ressler KJ, Jovanovic T. Inflammation in fear-and anxiety-based disorders: PTSD, GAD, and beyond. Neuropsychopharmacology. 2017;42:254-70.
  • 204
    Uzun N, Akıncı MA. Hemogram parameters in childhood anxiety disorders: could anxiety disorders be related with inflammation? Med Hypotheses. 2021;146:110440.

Publication Dates

  • Publication in this collection
    30 Aug 2021
  • Date of issue
    Jan-Feb 2022

History

  • Received
    31 Mar 2021
  • Accepted
    15 June 2021
Associação Brasileira de Psiquiatria Rua Pedro de Toledo, 967 - casa 1, 04039-032 São Paulo SP Brazil, Tel.: +55 11 5081-6799, Fax: +55 11 3384-6799, Fax: +55 11 5579-6210 - São Paulo - SP - Brazil
E-mail: editorial@abp.org.br