<<

ACCEPTED MANUSCRIPT

DePierro et al. Supplement

Translating Molecular and Neuroendocrine Findings in Posttraumatic Stress Disorder and Resilience to Novel Therapies Supplemental Information

Table S1. Extended summary of candidate PTSD pharmacotherapies

Treatment Target Rationale for use in Effect Status Reverse Translation Engagement PTSD Size SSRIs (1, 2) Serotonergic M ainly to treat 0.48 A Perhaps PTSD involves (several 5-HT comorbid symptoms; diminished capacity to receptors) perhaps PTSD involves downregulate 5-HT1B receptors; diminished capacity to alterations of serotonergic downregulate 5-HT1B receptors in the amygdala; receptors connection of , trauma, MANUSCRIPT and hippocampal volume Venlafaxine (2) Monoaminergic Treatment of 0.48 B Not thoroughly investigated (serotonergic, symptoms overlapping norepinephrinergic) with depression; adrenergic involvement Trazodone (2) Monoaminergic I mprove sleep n/a B, but Not thoroughly investigated (5-HT2; alpha-1 disturbance may for antagonist) improve symptoms comorbid insomnia only Atypical Dopaminergic treat psychotic-like 0.1-0.41 C Not thoroughly investigated Antipsychotics (2) (D2 antagonism)ACCEPTED symptoms Benzodiazepines GABAergic treat anxiety 0.28 C Alterations of GABA (2) (GABA-A positive receptors allosteric modulator)

1 ACCEPTED MANUSCRIPT

DePierro et al. Supplement

Treatment Target Rationale for use in Effect Status Reverse Translation Engagement PTSD Size Clonidine/ Adrenergic Reduction of -0.08 C Modest effects suggest that Guanfacine (3-5) (alpha-2-agonist) hyperadrenergic PTSD symptom improvement activity may additionally require manipulation of other targets Prazosin (2, 6-8) Adrenergic Reduction of 0.78 C Specific effects on nightmares (alpha-1-antagonist) hyperadrenergic and sleep suggest these activity symptoms are underpinned by noradrenergic dysregulation Propranolol (9-11) Adrenergic Reduction of n/a C Modest effects in preventing (beta-2-antagonist) hyperadrenergic consolidation of aversive activity with the memories in PTSD compared to specific intent of robust effects in preclinical blocking studies highlights gap in reconsolidation of fear translatability of fear models to memory; possible PTSD treatment prophylactic MANUSCRIPT immediately after trauma Topiramate (12) GABAergic? Use of other 1.20 C Stress-activated limbic (voltage-gated anticonvulsants in kindling effect of PTSD sodium channels, refractory PTSD; avoid voltage-activated side effects seen with calcium channels, SSRIs GABA-A, AMPA) Typical Monoaminergic treat psychotic-like n/a C Not thoroughly investigated Antipsychotics (2) (5-HT2A symptoms antagonism, fast D2 dissociation, 5ACCEPTED- HT1A agonism)

2 ACCEPTED MANUSCRIPT

DePierro et al. Supplement

Treatment Target Rationale for use in Effect Status Reverse Translation Engagement PTSD Size Mirtazapine Monoaminergic successful treatment 0.27 C-D Not thoroughly investigated (13, 14) (Adrenergic, of other anxiety and antihistamine) sleep disorders Yohimbine (15) Adrenergic Since this n/a D Intrusive and hyperarousal (alpha-2-antagonist) medication induces symptoms may be underpinned flashbacks it may by noradrenergic dysregulation; enhance arousal during enhancing these symptoms in exposure therapy, psychotherapy do not promote facilitating extinction symptom reduction learning Bupropion (16) Monoaminergic SSRIs have been 0.21 D Not thoroughly investigated (dopaminergic, helpful but have more norepinephrinergic) side-effects than Bupropion Gabapentin Anxiety Reduction of anxiety n/a D Not thoroughly investigated symptoms, after trauma to prevent MANUSCRIPT mechanism PTSD; decrease unknown (voltage comorbid symptoms gated calcium such as alcohol use, channel) sleep disorder, which may contribute to PTSD severity Nefazodone (17) Monoaminergic Anxiolytic helpful n/a D1 Not thoroughly investigated (5-HT2; alpha-1 with sleep without side antagonist) effect burden of SSRIs MAOIs (2) Monoaminergic antidepressant 0.21 D1 Not thoroughly investigated ACCEPTEDeffects TCAs (2) Monoaminergic antidepressant 0.36 D1 Not thoroughly investigated effects

3 ACCEPTED MANUSCRIPT

DePierro et al. Supplement

Treatment Target Rationale for use in Effect Status Reverse Translation Engagement PTSD Size Riluzole (16, 18) Glutamatergic; because of anti- n/a E Not specific to PTSD, but in GABAergic; glutamatergic (NMDA) GAD, correlation between excitotoxic target, trialed in other improvement in anxiety pathways, anxiety disorders with symptoms and increase in mitochondrial some signal hippocampal volume function, fat , sodium channel function, calcium-dependent potassium currents (VGNC, kainate and NMDAR, GABA-A receptors) buprenex/vivitrol Opiate (kappa Observation that n/a E1 Development of KOR PET (19, 20) opioid receptor patients self-medicate radiotracer which lead to finding antagonist) with opioids to MANUSCRIPT that KOR in ventral striatal alleviate PTSD circuit involved in trauma- symptoms; preclinical related dysphoria, perhaps data demonstrating mediated by HPA improved behavioral responses to stress Hydrocortisone HPA axis enhancement of n/a E1 Elevated or diminished levels (21) synthetic hormone reduced glucocorticoid of glucocorticoids during a replacement signaling to improve critical window immediately stress response following trauma may cause immediately following permanent cytoarchitectural trauma to prevent changes which contribute to ACCEPTEDPTSD; may have pathophysiology effects on memory reconsolidation

4 ACCEPTED MANUSCRIPT

DePierro et al. Supplement

Treatment Target Rationale for use in Effect Status Reverse Translation Engagement PTSD Size Hydrocortisone- HPA axis facilitation of n/a E1 Hydrocortisone may diminish augmented exposure- synthetic hormone extinction, therapeutic fear during reactivation of therapy (22) administration prior reconsolidation memory which allows patient to to exposure sessions more meaningfully engage material Pregnenolone (23) GABAergic, Low levels of the n/a E1 Trials still underway unknown (GABA- hormone observed in A; downstream PTSD; GABA-A targets of steroid receptors metabolites) downregulated in chronic stress Neuropeptide Y HPA axis; regulates HPA axis, n/a E1 Trials still underway (24) neurogenesis may contribute resilience during uncontrollable stress; levels found to be low MANUSCRIPT in PTSD Ketamine Glutamatergic observational studies n/a E1 Glutamatergic pathway in (25, 26) system; dendritic that patients who PTSD still under investigation, spine? (NMDA received ketamine after but likely related to the effect of receptor antagonist) trauma may have a chronic stress on learning and lower incidence of memory; ketamine may rapidly PTSD promote neuroplasticity in PTSD

ACCEPTED

5 ACCEPTED MANUSCRIPT

DePierro et al. Supplement

Treatment Target Rationale for use in Effect Status Reverse Translation Engagement PTSD Size MDMA-assisted Creation of ideal Fostering trust in the 0.8-2.8 E1 Probing the pathway of psychotherapy setting for a therapeutic alliance, opening/closing of critical (27, 28) psychotherapeutic maintaining optimal periods of plasticity experience that leads arousal during memory to meaningful re- retrieval and appraisal of trauma reconsolidation, feelings of self- compassion 7-Keto DHEA HPA axis/GABA- DHEA may promote n/a E1 Results of trial not yet (29) A antagonist, resilience after trauma published positive NMDA modulation, metabolizes cortisol Pregabalin Glutamatergic Has been helpful in n/a E1 Plan is to probe (voltage-gated alcohol use disorder endophenotype with genetic calcium channel that plus GAD in the past MANUSCRIPT variant glutamate transporter modulates glutamate so may be helpful for transporter 1) GAD plus PTSD; case reports of adjunctive pregabalin ameliorating PTSD symptoms GSK561679 HPA axis PTSD 0.277 in E1 Towards development of (Verucerfont) (CRF-1 antagonist) endophenotype with child abuse endophenotypes and (30) exposure to child abuse NR3C1 personalized medicine and also have a SNP of methyl- the receptor gene have ation group CRF1 receptor ACCEPTEDhyperactivity/ suppresses ACTH response to stress

6 ACCEPTED MANUSCRIPT

DePierro et al. Supplement

Treatment Target Rationale for use in Effect Status Reverse Translation Engagement PTSD Size Stimulates enhance extinction n/a E1 Trials still underway (+imaginal exposure) mitochondrial learning (31) oxidative metabolism and cerebral oxygen consumption; enhances memory consolidation/ extinction D-cycloserine- Glutamatergic? Induce LTP during 0.61 E1 Relevance of NMDA- assisted exposure (NDMA agonist) therapy and enhance mediated learning and plasticity therapy extinction of memory during exposure therapy (32) Oxytocin Glucocorticoid; reduced intensity of n/a E1 Oxytocin receptor methylation (33) social bonding fear memory by may be implicated in (oxytocin receptors; decreasing amygdala MANUSCRIPT pathophysiology; Oxytocin’s increase social activation; increased role in critical period plasticity, cognition/feelings of pro-social behavior regulation of HPA system in closeness) lacking in PTSD PTSD; may mediate social cognition deficits in PTSD Cannabidiol Endocannabinoid patients have n/a E1 Involvement of the ECB (34, 35) (inhibition of the increased CB1 system in the expression of uptake or of the receptors which may be contextual fear conditioning; a enzymatic caused by a chronic potential new target in the degradation of deficiency of the adenosinergic system endocannabinoids) endocannabinoid AEA CB1 agonist Endocannabinoid enhance fear n/a E1 Trials still underway (Nabilone) ACCEPTEDextinction via endocannabinoid system

7 ACCEPTED MANUSCRIPT

DePierro et al. Supplement

Treatment Target Rationale for use in Effect Status Reverse Translation Engagement PTSD Size Nitrous Oxide Consolidation of may block n/a E1 Research still preliminary (36, 37) memories; NMDA; consolidation of kindling (? NMDA) memories after trauma; some preclinical evidence of role of NO as mediator of stress and hippocampal degeneration and cognitive deficits SRX246 HPA axis/parvo- preclinical and n/a E1 Involvement of AVPR1a (38) cellular preliminary clinical signaling involvement in circuits vasopressinergic data suggesting relevant to processing aversive (vasopressin V1A antagonist may correct social and emotional receptor antagonist) a dysregulation of information vasopressinergic signaling in stress -MANUSCRIPT related illnesses Doxazosin Adrenergic same mechanism as n/a E1 Still in trials (39) (alpha-1 antagonist) prazosin but once daily dosing, less hypotension TNX-102/Tonmya Monoaminergic sleep disturbance n/a E1/ E2 Meaningful improvement at (Cyclobenzaprine) (5-HT2A may lead to week 4, but this signal never (40) antagonism, alpha 1 development and further examined to investigate antagonist, maintenance of other underlying mechanism PTSD symptoms antagonism) ACCEPTED

8 ACCEPTED MANUSCRIPT

DePierro et al. Supplement

Treatment Target Rationale for use in Effect Status Reverse Translation Engagement PTSD Size Mifepristone HPA axis may restore n/a E2 Modest effects in relation to (41, 42) GR type-2 receptor homeostasis to cognition suggest HPA antagonist alterations in HPA axis involvement in cognitive aspects seen in PTSD through of PTSD elevation in cortisol and ACTH, resulting in recalibration of axis Tiagabine GABAergic perhaps correct n/a E2 Some signal in symptom (43, 44) (selective GABA PTSD deficits in reduction perhaps helped reuptake inhibitor) GABA signaling confirm GABA involvement Orvepitant Substance P/ substance P- n/a E2 Potential to dissect and clarify (GW823296) tachykinin pathway neurokinin1 involved role of substance P/tachykinin (45) (tachykinin receptor in experimental models pathway in PTSD 1) of stress, fear, reward; substance P elevated in PTSD CSF MANUSCRIPT Ganaxolone GABAergic previous data n/a E2 Likely role for neuro-steroids (synthetic analog of (GABA-A positive indicating neuroactive in pathophysiology, but still allopregnanolone) allosteric modulator) steroids with GABA-A unclear; perhaps endophenotype (46) activity involved in pathophysiology of PTSD BNC210 Nicotinic anti - inflammatory n/a E2 Anxiolytic without cognitive (47) (α7-nicotinic or sedating effects of acetylcholine benzodiazepines receptor negative allosteric modulator)ACCEPTED

9 ACCEPTED MANUSCRIPT

DePierro et al. Supplement

Treatment Target Rationale for use in Effect Status Reverse Translation Engagement PTSD Size Nepicastat (48) Adrenergic r educe the elevated n/a E2 Clarifying an endophenotype (inhibitor of NE activity in PTSD and its treatment response -β- and therefore reduce hydroxylase) PTSD hyper-arousal symptom Sirolimus Protein Synthesis i nhibit mTOR to n/a E2 Traumatic memories that are (49) (mTOR inhibitor) alter amygdala and more recent may be less hippocampal dendritic entrenched and therefore arborization during possibly more possible to change exposure to modify reconsolidation of traumatic memory Neurokinin 1 Substance substance P- 0.22 E2 Signal for substance P- antagonist P/tachykinin neurokinin1 involved neurokinin system involvement (GR205171) pathway (tachykinin in experimental models in at least hyperarousal (50) receptor 1) of stress, fear, reward; MANUSCRIPT symptoms substance P elevated in PTSD CSF SNC-102 (51, 52) Glutamatergic, n/a n/a E2 None-data never published GABAergic (co-agonist of NMDA receptors)

Note: Applying VA/DoD guidelines (53) and clinical usage data from a recent review (54): A=FDA Approved; B = Not approved but VA/DoD guidelines support use; C = Used clinically but not recommended in guidelines; D = Use is not common practice; D1 = Guidelines recommend, but not commonly used; E1 = Experimental use/Active trials; E2 = Experimental use/Failed trials. ACCEPTED

10 ACCEPTED MANUSCRIPT

DePierro et al. Supplement

Supplemental References

1. MacNamara A, Rabinak CA, Kennedy AE, Fitzgerald DA, Liberzon I, Stein MB, et al. (2016): Emotion regulatory brain function and SSRI treatment in PTSD: neural correlates and predictors of change. Neuropsychopharmacology. 41:611-618.

2. Stahl SM (2013): Stahl's essential psychopharmacology: neuroscientific basis and practical applications. 3rd ed. Cambridge, UK: Cambridge University Press.

3. Horrigan JP (1996): Guanfacine for PTSD nightmares. Journal of the American Academy of Child and Adolescent Psychiatry. 35:975.

4. Harmon RJ, Riggs PD (1996): Clonidine for posttraumatic stress disorder in preschool children. Journal of the American Academy of Child & Adolescent Psychiatry.

5. Connor DF, Grasso DJ, Slivinsky MD, Pearson GS, Banga A (2013): An open-label study of guanfacine extended release for traumatic stress related symptoms in children and adolescents. Journal of child and adolescent psychopharmacology. 23:244-251.

6. Raskind MA, Peskind ER, Chow B, Harris C, Davis-Karim A, Holmes HA, et al. (2018): Trial of prazosin for post-traumatic stress disorder in military veterans. New England Journal of Medicine. 378:507-517. MANUSCRIPT 7. Berardis DD, Marini S, Serroni N, Iasevoli F, Tomasetti C, de Bartolomeis A, et al. (2015): Targeting the noradrenergic system in posttraumatic stress disorder: a systematic review and meta-analysis of prazosin trials. Current drug targets. 16:1094-1106.

8. Singh B, Hughes AJ, Mehta G, Erwin PJ, Parsaik AK (2016): Efficacy of prazosin in posttraumatic stress disorder: a systematic review and meta-analysis. Prim Care Companion CNS Disord. 18.

9. Argolo FC, Cavalcanti-Ribeiro P, Netto LR, Quarantini LC (2015): Prevention of posttraumatic stress disorder with propranolol: a meta-analytic review. Journal of psychosomatic research. 79:89-93.

10. Brunet A, Saumier D, Liu A, Streiner DL, Tremblay J, Pitman RK (2018): Reduction of PTSD symptomsACCEPTED with pre-reactivation propranolol therapy: a randomized controlled trial. American Journal of Psychiatry. 175:427-433.

11. Taylor F, Cahill L (2002): Propranolol for reemergent posttraumatic stress disorder following an event of retraumatization: a case study. Journal of Traumatic Stress. 15:433-437.

11 ACCEPTED MANUSCRIPT

DePierro et al. Supplement

12. Berlin HA (2007): Antiepileptic drugs for the treatment of post-traumatic stress disorder. Current psychiatry reports. 9:291-300.

13. Alderman CP, Condon JT, Gilbert AL (2009): An open-label study of mirtazapine as treatment for combat-related PTSD. Annals of Pharmacotherapy. 43:1220-1226.

14. Davidson JR, Weisler RH, Butterfield MI, Casat CD, Connor KM, Barnett S, et al. (2003): Mirtazapine vs. placebo in posttraumatic stress disorder: a pilot trial. Biological psychiatry. 53:188-191.

15. Southwick SM, Krystal JH, Morgan CA, Johnson D, Nagy LM, Nicolaou A, et al. (1993): Abnormal noradrenergic function in posttraumatic stress disorder. Archives of general psychiatry. 50:266-274.

16. Averill LA, Purohit P, Averill CL, Boesl MA, Krystal JH, Abdallah CG (2017): Glutamate dysregulation and glutamatergic therapeutics for PTSD: evidence from human studies. Neuroscience letters. 649:147-155.

17. Hidalgo R, Hertzberg M, Mellman T, Petty F, Tucker P, Weisler R, et al. (1999): Nefazodone in post-traumatic stress disorder: Results from six open-label trials. International Clinical Psychopharmacology. 14:61-68. 18. Grant P, Song JY, Swedo SE (2010): ReviewMANUSCRIPT of the use of the glutamate antagonist riluzole in psychiatric disorders and a description of recent use in childhood obsessive- compulsive disorder. Journal of child and adolescent psychopharmacology. 20:309-315.

19. Pietrzak RH, Naganawa M, Huang Y, Corsi-Travali S, Zheng M-Q, Stein MB, et al. (2014): Association of in vivo κ-opioid receptor availability and the transdiagnostic dimensional expression of trauma-related psychopathology. JAMA psychiatry. 71:1262-1270.

20. Lake EP, Mitchell BG, Shorter DI, Kosten T, Domingo CB, Walder AM (2019): Buprenorphine for the treatment of posttraumatic stress disorder. The American journal on addictions. 28:86-91.

21. Zohar J, Yahalom H, Kozlovsky N, Cwikel-Hamzany S, Matar MA, Kaplan Z, et al. (2011): High dose hydrocortisone immediately after trauma may alter the trajectory of PTSD: interplay between clinical and animal studies. European Neuropsychopharmacology. 21:796- 809. ACCEPTED

22. Yehuda R, Bierer LM, Pratchett LC, Lehrner A, Koch EC, Van Manen JA, et al. (2015): Cortisol augmentation of a psychological treatment for warfighters with posttraumatic stress disorder: randomized trial showing improved treatment retention and outcome. Psychoneuroendocrinology. 51:589-597.

12 ACCEPTED MANUSCRIPT

DePierro et al. Supplement

23. Locci A, Pinna G (2017): Neurosteroid biosynthesis down‐regulation and changes in GABAA receptor subunit composition: a biomarker axis in stress‐induced cognitive and emotional impairment. British journal of pharmacology. 174:3226-3241.

24. Schmeltzer SN, Herman JP, Sah R (2016): Neuropeptide Y (NPY) and posttraumatic stress disorder (PTSD): a translational update. Experimental neurology. 284:196-210.

25. Krystal JH, Abdallah CG, Averill LA, Kelmendi B, Harpaz-Rotem I, Sanacora G, et al. (2017): Synaptic loss and the pathophysiology of PTSD: implications for ketamine as a prototype novel therapeutic. Current psychiatry reports. 19:74.

26. Feder A, Parides MK, Murrough JW, Perez AM, Morgan JE, Saxena S, et al. (2014): Efficacy of intravenous ketamine for treatment of chronic posttraumatic stress disorder: a randomized clinical trial. JAMA psychiatry. 71:681-688.

27. Amoroso T, Workman M (2016): Treating posttraumatic stress disorder with MDMA- assisted psychotherapy: a preliminary meta-analysis and comparison to prolonged exposure therapy. Journal of Psychopharmacology. 30:595-600.

28. Mithoefer MC, Feduccia AA, Jerome L, Mithoefer A, Wagner M, Walsh Z, et al. (2019): MDMA-assisted psychotherapy for treatment of PTSD: study design and rationale for phase 3 trials based on pooled analysis of six phase 2 randomizedMANUSCRIPT controlled trials. Psychopharmacology. 29. Yehuda R, Brand S, Golier J, Yang RK (2 006): Clinical correlates of DHEA associated with post‐traumatic stress disorder. Acta Psychiatrica Scandinavica. 114:187-193.

30. Pape JC, Carrillo-Roa T, Rothbaum BO, Nemeroff CB, Czamara D, Zannas AS, et al. (2018): DNA methylation levels are associated with CRF1 receptor antagonist treatment outcome in women with post-traumatic stress disorder. Clinical Epigenetics. 10:136.

31. Zoellner LA, Telch M, Foa EB, Farach FJ, McLean CP, Gallop R, et al. (2017): Enhancing extinction learning in posttraumatic stress disorder with brief daily imaginal exposure and methylene blue: a randomized controlled trial. The Journal of clinical psychiatry. 78:e782- e789.

32. Rothbaum BO, Price M, Jovanovic T, Norrholm SD, Gerardi M, Dunlop B, et al. (2014): A randomized, double-blind evaluation of D-cycloserine or alprazolam combined with virtual reality exposure ACCEPTEDtherapy for posttraumatic stress disorder in Iraq and Afghanistan War veterans. American Journal of Psychiatry. 171:640-648.

33. Flanagan JC, Sippel LM, Wahlquist A, Moran-Santa Maria MM, Back SE (2018): Augmenting Prolonged Exposure therapy for PTSD with intranasal oxytocin: A randomized, placebo-controlled pilot trial. Journal of psychiatric research. 98:64-69.

13 ACCEPTED MANUSCRIPT

DePierro et al. Supplement

34. Kansagara D, O’Neil M, Nugent S, Freeman M, Low A, Kondo K (2017): Benefits and Harms of Cannabis in Chronic Pain or Post-Traumatic Stress Disorder: A Systematic Review. VA ESP Project no. 05-225. Washington, DC: US Department of Veterans Affairs; 2016.

35. Bitencourt RM, Takahashi RN (2018): Cannabidiol as a Therapeutic alternative for post- traumatic stress disorder: from bench research to confirmation in human trials. Frontiers in neuroscience. 12.

36. Nagele P, Duma A, Kopec M, Gebara MA, Parsoei A, Walker M, et al. (2015): Nitrous oxide for treatment-resistant major depression: a proof-of-concept trial. Biological psychiatry. 78:10-18.

37. Oosthuizen F, Wegener G, Harvey BH (2005): Nitric oxide as inflammatory mediator in post-traumatic stress disorder (PTSD): evidence from an animal model. Neuropsychiatric Disease and Treatment. 1:109-123.

38. Lee RJ, Coccaro EF, Cremers H, McCarron R, Lu S-F, Brownstein M, et al. (2013): A novel V1a receptor antagonist blocks vasopressin-induced changes in the CNS response to emotional stimuli: an fMRI study. Frontiers in systems neuroscience. 7:100.

39. Smith C, Koola MM (2016): Evidence for using doxazosin in the treatment of posttraumatic stress disorder. Psychiatric annals. 46:553MANUSCRIPT-555. 40. Tonix Pharmaceuticals Holding Corp (2019): Tonmya for PTSD.

41. Golier JA, Caramanica K, DeMaria R, Yehuda R (2012): A pilot study of mifepristone in combat-related PTSD. Depression Research and Treatment. 2012:393251.

42. Golier JA, Caramanica K, Michaelides AC, Makotkine I, Schmeidler J, Harvey PD, et al. (2016): A randomized, double-blind, placebo-controlled, crossover trial of mifepristone in Gulf War veterans with chronic multisymptom illness. Psychoneuroendocrinology. 64:22-30.

43. Connor KM, Davidson JR, Weisler RH, Zhang W, Abraham K (2006): Tiagabine for posttraumatic stress disorder: effects of open-label and double-blind discontinuation treatment. Psychopharmacology. 184:21-25.

44. Davidson JR, Brady K, Mellman TA, Stein MB, Pollack MH (2007): The efficacy and tolerability of tiagabineACCEPTED in adult patients with post-traumatic stress disorder. Journal of clinical psychopharmacology. 27:85-88.

45. Ebner K, Sartori SB, Singewald N (2009): Tachykinin receptors as therapeutic targets in stress-related disorders. Current pharmaceutical design. 15:1647-1674.

14 ACCEPTED MANUSCRIPT

DePierro et al. Supplement

46. Rasmusson AM, Marx CE, Jain S, Farfel GM, Tsai J, Sun X, et al. (2017): A randomized controlled trial of ganaxolone in posttraumatic stress disorder. Psychopharmacology. 234:2245- 2257.

47. Garakani A, Murrough JW, Iosifescu DV (2014): Advances in psychopharmacology for anxiety disorders. Focus. 12:152-162.

48. Graham DP, Nielsen DA, Kosten TR, Davis LL, Hamner MB, Makotkine I, et al. (2014): Examining the Utility of Using Genotype and Functional Biology in a Clinical Pharmacology Trial: Pilot Testing Dopamine β-Hydroxylase, , and PTSD. Psychiatric genetics. 24:181-182.

49. Surís A, Smith J, Powell C, North CS (2013): Interfering with the reconsolidation of traumatic memory: sirolimus as a novel agent for treating veterans with posttraumatic stress disorder. Annals of Clinical Psychiatry. 25:33-40.

50. Mathew SJ, Vythilingam M, Murrough JW, Zarate Jr CA, Feder A, Luckenbaugh DA, et al. (2011): A selective neurokinin-1 receptor antagonist in chronic PTSD: a randomized, double- blind, placebo-controlled, proof-of-concept trial. European Neuropsychopharmacology. 21:221- 229.

51. Boothby LA, Doering PL (2005): Acamprosate for the treatment of alcohol dependence. Clinical therapeutics. 27:695-714. MANUSCRIPT 52. Witkiewitz K, Saville K, Hamreus K (2012): Acamprosate for treatment of alcohol dependence: mechanisms, efficacy, and clinical utility. Therapeutics and clinical risk management. 8:45-53.

53. VA/DOD (2017): VA/DOD Clinical Practice Guideline for the Management of Posttraumatic Stress Disorder and Acute Stress Disorder. Washington, DC: Author.

54. Krystal JH, Davis LL, Neylan TC, Raskind MA, Schnurr PP, Stein MB, et al. (2017): It is time to address the crisis in the pharmacotherapy of posttraumatic stress disorder: a consensus statement of the PTSD Psychopharmacology Working Group. Biological psychiatry. 82:e51-e59.

ACCEPTED

15