Skip to main content
Chronic Stress logoLink to Chronic Stress
. 2025 Oct 24;9:24705470251392481. doi: 10.1177/24705470251392481

Repurposing Existing Drugs for the Treatment of Post-Traumatic Stress Disorder

Olha Strilbytska 1,, Oleh Lushchak 1,2,3,
PMCID: PMC12559684  PMID: 41164121

Abstract

Chronic stress and traumatic events affect the psychological and physiological state of a person. Post-traumatic stress disorder (PTSD) is a type of psychological stress that occurs in critical situations that pose a direct threat to the life of the person and/or his or her loved ones. The prevalence of PTSD is increasing every year due to growing exposure to traumatic factors such as wars, natural disasters, and other crises. These events lead to severe psychological consequences, affecting a rising number of people worldwide. Currently, PTSD can be challenging to treat due to the complex nature of the disorder, variability in individual responses to treatment, and limitations in available therapeutic options. Considering the emergency need for effective PTSD treatment, a drug repurposing strategy presents a promising avenue to accelerate the availability of therapies. Here, we summarized and described drugs that were repurposed for alleviating PTSD symptoms. Moreover, we discussed the potential of some drugs, including alpha-adrenergic modulators, cannabinoids, glutamatergic modulators, and antipsychotics, for being repurposed for PTSD treatment. Drug repurposing implies the rapid identification of compounds with an established safety profile and known therapeutic effects that may be effective in PTSD. Repurposing existing drugs with already established pharmacokinetics and pharmacodynamics may shorten development timelines, facilitate a direct transition to the second phase of clinical trials, and lower costs. However, potential drawbacks and negative aspects should be discussed comprehensively.

Keywords: post-traumatic stress disorder, traumatic factor, stress, treatment, drugs

Introduction

Post-traumatic stress disorder (PTSD) is a serious mental condition that may occur in individuals who have experienced traumatic events (TE), such as physical or sexual violence, war, accidents, natural disasters, or other extreme situations. 1 Diagnostic systems were developed to assess PTSD, including the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5) and the International Classification of Diseases, 11th Edition (ICD-11). 2 According to DSM-5, there are five specific criteria required for the diagnosis of PTSD, including A – trauma criterion, B – intrusive recollection criterion, C – avoidance criterion, D – negative cognitions and mood criterion, E – alterations in arousal or reactivity criterion. 3 The most significant criterion for PTSD diagnosis is experiencing TE.

In accordance with the World Health Organisation, the lifetime prevalence of PTSD is 3.9% of the world population. 4 Approximately 70% of people worldwide are being subjected to a potential TE at some point in their lives; however, only a small fraction (5.6%) develop PTSD.4,5 Influence of both external and internal risk factors, such as genetic predisposition, neurophysiological changes and social support, determines development and PTSD severity. 6 It is worth noting that the prevalence of PTSD increases up to 15.3% in countries with war conflicts. 7 A recent nationwide cross-sectional study examining stress, anxiety, and PTSD symptoms among Ukrainians revealed that 93% of respondents experienced at least one mental health issue at a moderate or severe level during the first year of the Russian invasion. 8

PTSD is a multifaceted disorder and includes psychological, neurobiological, and physiological aspects. Dysfunction of the hypothalamic-pituitary-adrenal (HPA) axis, hyper-activation of brain structures responsible for stress response, including the amygdala and hippocampus, and chronic inflammation are among the key physiological mechanisms associated with PTSD.6,911

Standard therapeutic approaches for the treatment of PTSD focus on psychotherapy (eg, cognitive behavioural therapy) and the use of pharmacological agents, including antidepressants. Psychotherapy is often unavailable due to high costs or a lack of qualified specialists, and the role of pharmacotherapy has been studied more extensively in recent years. However, many pharmaceuticals have undesirable side effects, which reduce their acceptability to patients. 12 Considering these challenges, recent research has focused on finding new therapeutic approaches based on a deeper understanding of the biological mechanisms of PTSD. In this context, there is a growing interest in repurposing drugs for the treatment of PTSD. Drug repurposing is based on the use of already approved drugs for the treatment of other conditions that have the potential to affect PTSD development.

Although progress has been made in elucidating the neurobiological mechanisms underlying PTSD, effective pharmacological options remain limited. Despite several drugs approved for other diseases showing potential for decreasing PTSD symptoms, the supporting evidence is fragmented and often limited. This highlights the need for a comprehensive review to summarize current knowledge that will assist rational selection of candidates for further preclinical and clinical investigation. This review represents the first effort to comprehensively discuss the effectiveness of some pharmaceutical compounds in alleviating PTSD symptoms. In this review, we summarised current approaches to repurpose medicines for the treatment of PTSD, analysed their potential advantages and limitations, and highlighted the prospects for developing this strategy. Current challenges in PTSD treatment and factors that complicate the search for effective therapeutic solutions are discussed.

Methodology

This narrative review includes summaries of 101 papers to describe compounds that are effective in decreasing PTSD symptoms. A comprehensive literature search was performed to identify relevant studies addressing the potential of some drugs for being repurposed for PTSD treatment. The search was performed in PubMed, Scopus, and Web of Science databases for the period from January 2000 to January 2025. The search strategy combined controlled vocabulary (MeSH terms in PubMed) and free-text keywords. The primary search terms included: “post-traumatic stress disorder” OR “PTSD” OR “treatment” OR “drugs” OR “compound”). Boolean operators (“AND,” “OR”) and truncations were used to maximize sensitivity and specificity. Additional articles were identified by manual screening of reference lists from selected papers. Only articles published in English were considered.

Only research, review articles, and chapters published were included in this study. Studies were included if they: (1) investigated compounds that show effectiveness to decrease PTSD symptoms in preclinical models or clinical trials; (2) provided primary data on molecular, physiological, or behavioral outcomes; and (3) were published in peer-reviewed journals. Exclusion criteria included: (1) conference abstracts, theses, or non-peer-reviewed reports; (2) studies not directly related to the topic of review; and (3) duplicate publications.

Common language editing tools were applied to improve grammar and readability in accordance with academic writing standards.

Drug Repurposing in Psychiatry

One of the promising topics in the current research is the repurposing of existing medicines, also known as drug repositioning, which involves identifying new therapeutic applications for already approved drugs.13,14 For example, during the COVID-19 pandemic, several drugs were repurposed, 15 including Remdesivir, an antiviral drug developed to treat Ebola, 16 as well as Dexamethasone, a widely used steroid that reduces mortality among hospitalised patients requiring ventilation. 17

The central nervous system (CNS) is a key focus of the research for drug repurposing due to the limited understanding of the pathophysiology and mechanisms of CNS disorders. Many approved drugs for CNS disorders have unclear modes of action, and disease characterisation relies heavily on clinical symptoms rather than biology. 18 Despite high prevalence and unmet needs, CNS drug development has low success rates and longer regulatory approval times. 19

Repurposing of existing pharmacological drugs is a promising area in the treatment of PTSD, as evidenced by the rapid growth in the number of studies in this area. Drug repurposing involves investigating the potential of existing drugs for new therapeutic uses. This strategy is particularly valuable in fields with a historically high risk of failure, such as psychiatry. 20 Established safety profiles, cost-effectiveness, accelerated timelines, and expanded treatment options are among the main benefits of a drug repurposing strategy. Already known safety and tolerability profiles entail significantly reduced risks associated with the development process compared to novel drug candidates. Based on publicly available data, it was defined that development costs for new therapeutic agents range from $314 million to $2.8 billion. 21 Moreover, drug development from original idea to implementation in clinical practice takes 12–15 years, 21 with an estimated success rate of only 2%. 22 Drug repurposing allows for bypassing early-stage development, which makes it a more cost-effective approach.

Drug repurposing is a complex process that includes several stages, including compound selection, compound acquisition, drug development and safety monitoring. 23 Drug-oriented, disease-oriented and target-oriented are three main approaches to drug repurposing. 23 A drug-oriented approach expands the application of an already approved drug. The disease-oriented approach is especially valuable for rare diseases and involves the identification of the diseases with homologous underlying biological mechanisms to the indication the original drug treats. Investigating the specific molecular targets implicated in a disease's pathology involves a target-oriented approach.

Many psychiatric disorders, such as depression, bipolar disorder, schizophrenia, and certain anxiety disorders, have limited effective pharmacological treatments. This raises an urgent need for innovative strategies to expand therapeutic options. Existing compounds were suggested to be effective in alleviating various CNS conditions. Ketamine, originally developed as an anaesthetic, has been successfully repurposed for treatment-resistant depression.24,25 Ketamine exerts rapid antidepressant effects through N-methyl-D-aspartate (NMDA) receptor antagonism and does not cause adverse effects. 26 Similarly, lithium has been used for gout and other medical conditions and was proven to be effective in the treatment of bipolar disorder. 27 The mood-stabilising properties of lithium are associated with its neurotrophic properties, which affect several molecular targets, including neurotrophins, glycogen synthase kinase 3 (GSK-3), and key proteins of the mitochondrial/endoplasmic reticulum. 28 Another example of drug repurposing in psychiatry is modafinil, which has been initially approved for narcolepsy, and recently investigated as an adjunctive treatment for major depressive disorder (MDD) 29 and schizophrenia.30,31 Antiepileptic drugs, lamotrigine and valproate, were effectively used for mood stabilisation in bipolar disorder.32,33 Atypical antipsychotics such as aripiprazole and quetiapine, initially used to treat schizophrenia, are now extensively used for depression and anxiety disorders.34,35

The drug repurposing approach presents several challenges, including the need to reevaluate dosage, long-term effects, and regulatory considerations for new indications. Nevertheless, the cost-effectiveness and faster development timelines make drug repurposing an attractive option, particularly in the context of increasing mental health issues globally. Psychological disorders often have complex and multifactorial mechanisms involving genetic, neurochemical, and environmental factors. To repurpose drugs for the treatment of psychological disorders, the detailed pathophysiology of these conditions should be studied.

Repurposing Existing Drugs for PTSD Therapy

Understanding the neurobiology of PTSD accelerates the development of effective pharmacotherapy. Post-traumatic stress disorder is a multidimensional disorder that combines a wide range of neurobiological changes, including disturbances of the hypothalamic-pituitary-adrenal gland axis, hyperactivation of the amygdala, and disruption of some hippocampal and cortical functions.36,37 Stress leads to an elevated release of stress hormones such as adrenaline and epinephrine into the bloodstream, initiating changes in the brain and triggering the stress response. 38 One of the key players in this stress response is corticotropin-releasing hormone (CRH), which is secreted by neurons in the paraventricular nucleus of the hypothalamus. CRH stimulates the release of adrenocorticotropic hormone (ACTH) from the anterior pituitary gland.39,40 ACTH moves through the bloodstream to the adrenal glands, prompting the release of cortisol. Cortisol plays a crucial role in regulating metabolic and immune functions, contributing to the overall stress response. Targeting cortisol levels and function could help alleviate the physiological and emotional symptoms of PTSD.

The renin-angiotensin-aldosterone (RAA) system is involved in the stress response and works in tandem with the HPA axis to regulate physiological and metabolic processes during stress. Stress-induced increase in the levels of angiotensin results in activation of the HPA axis and an increase in circulating cortisol. 41 Conversely, ACTH induces aldosterone secretion. 42 Overactivation of RAA system contributes to neuroinflammation and oxidative stress, which are associated with PTSD pathophysiology. 43 High aldosterone levels may cause depressive and anxiety-related behavior through direct impacts on brain regions, including the amygdala and prefrontal cortex. 44 Stress activates the sympathetic nervous system (SNS) and triggers the release of renin into the bloodstream. Renin converts angiotensinogen into angiotensin I, which is then converted into angiotensin II by angiotensin-converting enzyme (ACE). 45 Angiotensin II enhances the activity of the HPA axis, 46 playing a central role in the stress response in PTSD.

Neurotransmitter systems play a critical role in PTSD severity and represent key targets for drug therapy. Dysregulation of serotonin in PTSD is associated with heightened anxiety, aggression and depression. 47 Serotonin reuptake inhibitors (SSRIs) work by inhibiting the reuptake of serotonin and increasing serotonin levels in the brain, leading to a reduction of PTSD severity. Dopamine dysfunction is increasingly recognised as a contributing factor to the symptoms of anhedonia and emotional numbing commonly seen in PTSD. 48 Glutamate and glutamatergic systems are implicated in synaptic plasticity and fear learning. Increased blood glutamate levels are closely associated with mood disorders.4951 Drugs targeting the glutamatergic systems could be effective in alleviating PTSD symptoms.

SSRIs paroxetine and sertraline had been initially used to treat depression and anxiety disorders. 52 Currently, paroxetine and sertraline are the only drugs approved by the Food and Drug Administration as first-line treatments for PTSD. 53 The therapeutic actions of SSRIs are associated with the enhancement of serotonin levels that are decreased under stress or depression (Table 1). 54 SSRIs work by blocking the serotonin transporter (SERT) located at the presynaptic axon terminal. This inhibition prevents serotonin (5-hydroxytryptamine, or 5-HT) reabsorption by presynaptic neurons. As a result, serotonin accumulates in the synaptic cleft, allowing it to activate postsynaptic receptors for a more extended period. 55 Enhancement of the serotonergic neurotransmission causes improvement of mood, anxiety, and sleep. 56

Table 1.

Existed Drugs That Show Effectiveness in Alleviation PTSD Symptoms.

Drug Original Use Beneficial effects on PTSD symptoms Ref
Paroxetine Depression and anxiety decreases intrusive memories, flashbacks, and irritability, alleviates avoidance behaviors, decreases anxiety and depression [90,91]
Sertraline Depression and anxiety decreases intrusive memories, flashbacks, and irritability, alleviates avoidance behaviors, decreases anxiety and depression [ 92 ]
Dronabinol Nausea and vomiting caused by cancer treatment improves sleep quality and decreases hyperarousal [ 57 ]
Clonidine Hypertension alleviates sleep disturbances and nightmares; decreases overall PTSD symptoms [ 58 ]
Doxazosin Hypertension alleviates sleep disturbances and nightmares; decreases overall PTSD symptoms [6467]
Propranolol Hypertension, anxiety, and migraine decreases the intensity of intrusive memories, flashbacks, nightmares and hyperarousal [ 68 ]
Prazosin Hypertension alleviates sleep disturbances and nightmares; decreases overall PTSD symptoms [7274]
Ketamine Anesthetic decreases depression and overall PTSD symptoms [ 79 ]
Risperidone Schizophrenia and bipolar disorder improves the psychiatric symptoms of PTSD [ 81 ]
Quetiapine Schizophrenia and bipolar disorder treats hyperarousal and reexperience disorder; reduces flashbacks [8385]
Lamotrigine Epilepsy decreases reexperiencing and avoidance/numbing symptoms [ 86 ]
Valproate Epilepsy and bipolar disorder decreases hyperarousal, irritability, and mood disturbances [88,89]
Minocycline Antibiotic reduces mood symptoms of PTSD [ 93 ]
Metformin Type 2 diabetes reduces symptoms of PTSD [94,95]
Hydrocortisone Adrenocortical insufficiency reduces symptoms of depression and PTSD [96,97]

The study of existing cannabinoid drugs [NCT04448808] led by Charité University was focused on the effectiveness of dronabinol in overcoming nightmares caused by PTSD. 57 Dronabinol modulates the endocannabinoid system, which influences emotional regulation, fear extinction, and sleep. Preliminary evidence from open-label and small-scale trials suggested significant improvements in nightmare frequency, sleep quality, and hyperarousal symptoms. 57

A study [NCT04877093] conducted by the Minneapolis Veterans Affairs Healthcare System investigated the effectiveness of low-dose clonidine for the treatment of PTSD in veterans who often have PTSD.58,59 Clonidine, an alpha-2 adrenergic agonist, showed promise in improving PTSD symptoms, particularly sleep disturbances and nightmares. It was reported that significant improvements in symptoms occurred for doses above 0.1 mg/day, with 49–85% of patients experiencing partial or marked relief. 58 Mild side effects, including grogginess and dizziness, were reported in 23% of patients. 58 Clonidine acts centrally to inhibit norepinephrine release with subsequent inhibition of sympathetic overactivation associated with PTSD. 60 Moreover, clonidine can stabilise adrenergic activity and lead to improved mood and a reduction of aggressive behaviours. 61

Doxazosin was approved for the treatment of hypertension as well as urinary tract obstruction, and was shown to be effective in reducing PTSD symptoms 62 and may also be used for comorbid PTSD and alcohol use disorders. 63 Improved sleep quality and decreased frequency and intensity of trauma-related nightmares were documented in combat veterans with PTSD after 8 weeks of treatment with 4 mg/day of doxazosin. 64 The study by Rodgman and colleagues 65 showed alleviation of PTSD symptoms after four days of treatment with 16 mg/day of doxazosin. The efficiency of doxazosin for PTSD was also proven in several studies.66,67

Propranolol, used initially for the treatment of hypertension, anxiety, and migraine prevention, was repurposed for reducing the emotional intensity of traumatic memories associated with PTSD. 68 Propranolol acts as a non-selective beta-adrenergic blocker to reduce overactivation of the sympathetic nervous system caused by stress. 69 However, some controversial studies showed that posttrauma propranolol treatment had no efficacy for preventing subsequent PTSD 70 or did not reduce PTSD incidence. 71

A repurposed anti-hypertensive drug, prazosin, was shown to be beneficial for treating PTSD symptoms.7274 By blocking alpha-1 adrenergic receptors, prazosin improves sleep quality and reduces nightmares and PTSD symptoms. 75

Ketamine had been approved initially as a general anaesthetic and was repurposed for several other disorders, including depressive disorders, suicidal ideation, substance-use disorders, anxiety disorders, and bronchial asthma complications. 76 The beneficial effects of ketamine on mental health are realised via inhibition of NMDA receptors 77 and regulation of glutamate pathways. 78 A randomised controlled trial involving 30 individuals with chronic PTSD showed that the ketamine-treated group showed significant improvement in PTSD symptoms and depression. 79

Primarily used to treat schizophrenia and bipolar disorder, risperidone and quetiapine have shown potential in addressing PTSD symptoms.80,81 The effects of low-dose risperidone on aggression and PTSD symptoms in male combat veterans with PTSD were previously evaluated. 81 In this study, risperidone significantly reduced irritability and intrusive thoughts, suggesting its potential efficacy in managing these PTSD-related symptoms. 81 The beneficial effects of risperidone in PTSD are associated with the regulation of the dopaminergic system and HPA axis. 82 Quetiapine is recommended for persons with PTSD to decrease symptoms of hyperarousal and re-experience disorder.83,84 Moreover, the combination therapy with quetiapine, SSRI and gabapentin significantly reduced flashbacks. 85

Anticonvulsants, including lamotrigine and valproate, were found to be effective for the treatment of PTSD. Treatment with lamotrigine leads to improvement in reexperiencing and avoidance/numbing symptoms in both combat and civilian PTSD. 86 Lamotrigine inhibits voltage-gated sodium channels, reduces glutamate release in the brain, and inhibits overactivation of the amygdala. 87 Valproate shows potential for the treatment of PTSD by mitigating symptoms such as hyperarousal, irritability, and mood disturbances.88,89 It acts by increasing GABAergic activity and reducing glutamatergic excitotoxicity, addressing neural hyperactivity and stress-related neuroplastic changes observed in PTSD.88,89

Targeting biological pathways involved in PTSD with pharmaceutical components may be promising for investigating new approaches for its treatment. Inflammation was shown to play a key role in the pathogenesis and pathophysiology of PTSD.9,10 Anti-inflammatory drugs, including non-steroidal anti-inflammatory drugs (NSAIDs), corticosteroids, and cytokine inhibitors, could show promise in PTSD treatment based on their mechanisms of action. Statins may potentially help reduce neuroinflammation and cognitive decline associated with PTSD. To prevent or treat PTSD, common antibiotics with neuroprotective and anti-inflammatory effects may be effective. An example is minocycline, which passed phase IV of clinical trials, and its treatment is associated with reduced inflammation and reduced mood symptoms of PTSD. 93 Corticosteroid hydrocortisone, which is used for replacement therapy in primary, secondary, or acute adrenocortical insufficiency, showed promising effects in alleviating PTSD symptoms.94,95 Decreased intensity of chronic stress and PTSD symptoms at 6 months after traumatic event (cardiac surgery) was shown in patients exposed to hydrocortisone treatment. 96 A randomised double-blind trial showed that low-dose hydrocortisone treatment results in fewer PTSD and depression symptoms. 95 Moreover, the antibiotic rapamycin may be used as a treatment for anxiety disorders. 96

Metformin, initially approved for the treatment of Type 2 diabetes, was later shown to be effective in antiviral treatments.97,98 It was also able to decrease PTSD symptoms in both PTSD animal 99 and veterans. 100 Metformin activates the 5′-adenosine monophosphate-activated protein kinase (AMPK) pathway, attenuates oxidative stress and preserves mitochondrial function in the hippocampus of rat models of PTSD. 99

Challenges and Limitations of Drug Repurposing for PTSD

The process of drug repurposing is often considered to be a highly advantageous strategy; however, potential drawbacks and negative aspects should be discussed comprehensively (Figure 1). Within the area of PTSD, drug repurposing presents several challenges and limitations. The complexity of PTSD pathophysiology, with neurobiological and psychological aspects, significantly complicates the identification of repurposed drugs. PTSD pathophysiology is associated with alterations in multiple systems, including the HPA axis, the serotonergic and dopaminergic pathways, and inflammation-related processes.9,10 Considering the multifaceted nature of PTSD, identifying a drug that can comprehensively address these diverse mechanisms is challenging.

Figure 1.

Figure 1.

Representations of Benefits and Challenges of Drug Repurposing for PTSD Treatment.

High comorbidity of PTSD with other psychiatric and physical health conditions, such as depression, anxiety disorders, substance use disorders, and cardiovascular diseases, 101 makes drug repurposing challenging. Indeed, treatments for PTSD may interact with medications prescribed for co-occurring conditions.

Additional limitations for drug repurposing may be caused by the heterogeneity of PTSD symptoms across individuals. A repurposed drug may be effective for one subset of symptoms and may not work for others. Moreover, drugs used to treat a particular disorder may exhibit one set of side effects, but those used to treat another disease may show completely different side effects. Consequently, the potential safety risks and side effects when using drugs outside their approved indications, particularly in vulnerable PTSD populations, should be tested. Addressing all these limitations requires a multidisciplinary approach that integrates insights from neuroscience, pharmacology, and clinical practice.

Conclusion and Future Directions

Drug repurposing is a promising and cost-effective strategy for the development of new therapeutic approaches for PTSD. Applying a drug repurposing strategy allows for saving costs and expediting the drug development process. Using existing data on approved or investigational drugs within a drug repurposing strategy significantly reduces the time and costs associated with early-stage research and preclinical trials. Moreover, drug repurposing facilitates the development of combination therapies that address multiple aspects of PTSD pathophysiology, potentially providing more comprehensive and effective treatment options.

Despite these advantages, several challenges must be carefully addressed to maximise the success of this strategy. Potential obstacles include a limited understanding of PTSD heterogeneity, the need for robust clinical validation, possible off-target effects, and regulatory hurdles in repurposing drugs for new indications. Overcoming these challenges requires interdisciplinary collaboration and integration of advanced technologies and high-throughput screening methods.

Future research phase III clinical trials should prioritize alpha-adrenergic modulators, cannabinoids, glutamatergic modulators, and antipsychotics, given their demonstrated potential to alleviate core symptoms of PTSD. Furthermore, a key direction for future research is the application of precision medicine in PTSD treatment. Tailoring drug repurposing efforts based on individual patient profiles, including genetic factors and severity of symptoms, might be addressed by precision medicine in the context of PTSD. Genetic variations play a crucial role in shaping individual susceptibility to PTSD and treatment response. Additionally, considering the heterogeneity of PTSD symptoms, precision medicine approaches may help identify the most suitable repurposed drugs for different symptom clusters, ultimately leading to more personalised and targeted interventions. Consequently, further research is essential to optimise its implementation, address existing limitations, and ensure its successful integration into clinical practice.

As a narrative review, this work does not follow the complete set of guidelines of a systematic review, which may introduce limitations such as selection bias. Furthermore, the absence of meta-analytic synthesis restricts quantitative comparison across studies. Nonetheless, by critically integrating existing preclinical and clinical findings, the review provides an integrated overview and highlights key directions for future research.

List of abbreviations

PTSD

Post-traumatic stress disorder

TE

Traumatic events

DSM-5

Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition

ICD-11

International Classification of Diseases 11th Edition

HPA

Hypothalamic-pituitary-adrenal

CNS

Central nervous system

NMDA

N-methyl-D-aspartate

GSK-3

Glycogen synthase kinase 3

MDD

Major depressive disorder

CRH

Corticotropin-releasing hormone

ACTH

Adrenocorticotropic hormone

RAA

Renin-angiotensin-aldosterone

SNS

Sympathetic nervous system

ACE

Angiotensin-converting enzyme

SSRIs

Serotonin reuptake inhibitors

SERT

Serotonin transporter

NSAIDs

Non-steroidal anti-inflammatory drugs

AMPK

Monophosphate-activated protein kinase

Footnotes

Ethics Approval and Consent to Participate: This article is a literature review and does not involve human participants or primary data collection. Therefore, ethical approval and consent to participate declarations are not applicable.

Consent for Publication: Not applicable.

Author Contributions: OL led the development and writing of the manuscript. OS contributed to the literature search, data extraction for the table, and writing the manuscript. OL and OS reviewed and provided feedback. All authors approved the final manuscript.

Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by a grant from the Ministry of Education and Science of Ukraine [grant number 0125U000396].

The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

References

  • 1.Du J, Diao H, Zhou X, et al. Post-traumatic stress disorder: A psychiatric disorder requiring urgent attention. Med Rev. (2021). 2022;2(3):219-243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Hansen M, Armour C, McGlinchey E, et al. Investigating the DSM-5 and the ICD-11 PTSD symptoms using network analysis across two distinct samples. Psychol Trauma. 2023;15(5):757-766. [DOI] [PubMed] [Google Scholar]
  • 3.Pai A, Suris AM, North CS. Post-traumatic stress disorder in the DSM-5: Controversy, change, and conceptual considerations. Behav Sci (Basel). 2017;7(1):7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Koenen KC, Ratanatharathorn A, Ng L, et al. Post-traumatic stress disorder in the world mental health surveys. Psychol Med. 2017;47(13):2260-2274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kessler RC, Aguilar-Gaxiola S, Alonso J, et al. Trauma and PTSD in the WHO world mental health surveys. Eur J Psychotraumatol. 2017;8(sup5):1353383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Sherin JE, Nemeroff CB. Post-traumatic stress disorder: The neurobiological impact of psychological trauma. Dialogues Clin Neurosci. 2011;13(3):263-278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Charlson F, van Ommeren M, Flaxman A, et al. New WHO prevalence estimates of mental disorders in conflict settings: A systematic review and meta-analysis. Lancet. 2019;394(10194):240-248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lushchak O, Velykodna M, Bolman S, et al. Prevalence of stress, anxiety, and symptoms of post-traumatic stress disorder among Ukrainians after the first year of Russian invasion: A nationwide cross-sectional study. Lancet Reg Health Eur. 2023;36:100773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lushchak O, Orru M, Strilbytska O, et al. Metabolic and immune dysfunctions in post-traumatic stress disorder: What can we learn from animal models? EXCLI J. 2023;22:928-945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Dmytriv TR, Tsiumpala SA, Semchyshyn HM, et al. Mitochondrial dysfunction as a possible trigger of neuroinflammation at post-traumatic stress disorder (PTSD). Front Physiol. 2023;14:1222826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Serhiyenko V, Holzmann K, Holota S, et al. An exploratory study of physiological and biochemical parameters to identify simple, robust and relevant biomarkers for therapeutic interventions for ptsd: study rationale, key elements of design and a context of war in Ukraine. Proc Shevchenko Sci Soc Med Sci [Internet]. 2022;69(2). [Google Scholar]
  • 12.Pawar N, Wiegand TJ. Monoamine oxidase inhibitors. In: Wexler P, ed. Encyclopedia of Toxicology (Fourth Edition). Academic Press; 2024:499-502. [Google Scholar]
  • 13.Zhan P, Yu B, Ouyang L. Drug repurposing: An effective strategy to accelerate contemporary drug discovery. Drug Discov Today. 2022;27(7):1785-1788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Vaiserman A, Koliada A, Lushchak O, Castillo MJ. Repurposing drugs to fight aging: The difficult path from bench to bedside. Med Res Rev. 2021;41(3):1676-1700. [DOI] [PubMed] [Google Scholar]
  • 15.Bakowski MA, Beutler N, Wolff KC, et al. Drug repurposing screens identify chemical entities for the development of COVID-19 interventions. Nat Commun. 2021;12(1):3309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Beigel JH, Tomashek KM, Dodd LE, et al. Remdesivir for the treatment of COVID-19. Final Report. N Engl J Med. 2020;383(19):1813-1826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Martinez-Guerra BA, Gonzalez-Lara MF, Roman-Montes CM, et al. Outcomes of patients with severe and critical COVID-19 treated with dexamethasone: A prospective cohort study. Emerg Microbes Infect. 2022;11(1):50-59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hardman JG, Limbird LE, Molinoff PB. Goodman & Gilman’s the Pharmacological Basis of Therapeutics: Section III Chapter 12 Neurotransmission and the Central Nervous System. 11th. McGraw-Hill; 2008:265-578. [Google Scholar]
  • 19.Pangalos MN, Schechter LE, Hurko O. Drug development for CNS disorders: Strategies for balancing risk and reducing attrition. Nat Rev Drug Discov. 2007;6(7):521-532. [DOI] [PubMed] [Google Scholar]
  • 20.Fava M. The promise and challenges of drug repurposing in psychiatry. World Psychiatry. 2018;17(1):28-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Wouters OJ, McKee M, Luyten J. Estimated research and development investment needed to bring a new medicine to market, 2009–2018. JAMA. 2020;323(9):844-553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Yeu Y, Yoon Y, Park S. Protein localisation vector propagation: A method for improving the accuracy of drug repositioning. Mol BioSyst. 2015;11:2096-2102. [DOI] [PubMed] [Google Scholar]
  • 23.Xue H, Li J, Xie H, Wang Y. Review of drug repositioning approaches and resources. Int J Biol Sci. 2018;14(10):1232-1244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Carboni E, Carta AR, Carboni E, Novelli A. Repurposing ketamine in depression and related disorders: Can this enigmatic drug achieve success? Front Neurosci. 2021;15:657714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Serafini G, Howland RH, Rovedi F, et al. The role of ketamine in treatment-resistant depression: A systematic review. Curr Neuropharmacol. 2014;12(5):444-461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Strasburger SE, Bhimani PM, Kaabe JH, et al. What is the mechanism of ketamine's rapid-onset antidepressant effect? A concise overview of the surprisingly large number of possibilities. J Clin Pharm Ther. 2017;42(2):147-154. [DOI] [PubMed] [Google Scholar]
  • 27.O'Connell RA, Mayo JA, Flatow L, et al. Outcome of bipolar disorder on long-term treatment with lithium. Br J Psychiatry. 1991;159:123-129. [DOI] [PubMed] [Google Scholar]
  • 28.Machado-Vieira R, Manji HK, Zarate Jr CA. The role of lithium in the treatment of bipolar disorder: Convergent evidence for neurotrophic effects as a unifying hypothesis. Bipolar Disord. 2009;11(Suppl 2):92-109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Fava M, Thase ME, DeBattista C. A multicenter, placebo-controlled study of modafinil augmentation in partial responders to selective serotonin reuptake inhibitors with persistent fatigue and sleepiness. J Clin Psychiatry. 2005;66(1):85-93. [DOI] [PubMed] [Google Scholar]
  • 30.Scoriels L, Jones PB, Sahakian BJ. Modafinil effects on cognition and emotion in schizophrenia and its neurochemical modulation in the brain. Neuropharmacology. 2013;64:168-184. [DOI] [PubMed] [Google Scholar]
  • 31.Lees J, Michalopoulou PG, Lewis SW, et al. Modafinil and cognitive enhancement in schizophrenia and healthy volunteers: The effects of test battery in a randomised controlled trial. Psychol Med. 2017;47(13):2358-2368. [DOI] [PubMed] [Google Scholar]
  • 32.Ginsberg LD. Efficacy and safety of lamotrigine for adults with bipolar disorder in a private practice setting. CNS Spectr. 2006;11:376-382. [DOI] [PubMed] [Google Scholar]
  • 33.Manning JS. Valproate in bipolar disorder: Case examples from a family practice. Prim Care Companion J Clin Psychiatry. 1999;1(3):71-73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Shapiro DA, Renock S, Arrington E, et al. Aripiprazole, a novel atypical antipsychotic drug with a unique and robust pharmacology. Neuropsychopharmacology. 2003;28(8):1400-1411. [DOI] [PubMed] [Google Scholar]
  • 35.Chessick CA, Allen MH, Thase M, et al. Azapirones for generalised anxiety disorder. Cochrane Database Syst Rev. 2006;2006(3):CD006115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Malikowska-Racia N, Salat K. Recent advances in the neurobiology of post-traumatic stress disorder: A review of possible mechanisms underlying an effective pharmacotherapy. Pharmacol Res. 2019;142:30-49. [DOI] [PubMed] [Google Scholar]
  • 37.Heim C, Nemeroff CB. Neurobiology of post-traumatic stress disorder. CNS Spectr. 2009;14(1 Suppl 1):13-24. [PubMed] [Google Scholar]
  • 38.Goldstein DS. Adrenal responses to stress. Cell Mol Neurobiol. 2010;30(8):1433-1440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Hadad NA, Schwendt M, Knackstedt LA. Hypothalamic-pituitary-adrenal axis activity in post-traumatic stress disorder and cocaine use disorder. Stress. 2020;23(6):638-650. [DOI] [PubMed] [Google Scholar]
  • 40.Leistner C, Menke A. Hypothalamic-pituitary-adrenal axis and stress. Handb Clin Neurol. 2020;175:55-64. [DOI] [PubMed] [Google Scholar]
  • 41.Grippo A, Johnson A. Stress, depression and cardiovascular dysregulation: A review of neurobiological mechanisms and the integration of research from preclinical disease models. Stress Amst Neth. 2009;12:1-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.El Ghorayeb N, Bourdeau I, Lacroix A. Role of ACTH and other hormones in the regulation of aldosterone production in primary aldosteronism. Front Endocrinol. 2016;7:72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Mirzahosseini G, Ismael S, Ahmed HA, Ishrat T. Manifestation of renin angiotensin system modulation in traumatic brain injury. Metab Brain Dis. 2021;36(6):1079-1086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Murck H, Schlageter L, Schneider A, et al. The potential pathophysiological role of aldosterone and the mineralocorticoid receptor in anxiety and depression - lessons from primary aldosteronism. J Psychiatr Res. 2020;130:82-88. [DOI] [PubMed] [Google Scholar]
  • 45.Seligowski AV, Duffy LA, Merker JB, et al. The renin-angiotensin system in PTSD: A replication and extension. Neuropsychopharmacology. 2021;46(4):750-755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Calogero AE, Fornito MC, Aliffi A, et al. Role of peripherally infused angiotensin II on the human hypothalamic-pituitary-adrenal axis. Clin Endocrinol (Oxf). 1991;34(3):183-186. [DOI] [PubMed] [Google Scholar]
  • 47.Southwick SM, Paige S, Morgan 3rd CA, et al. Neurotransmitter alterations in PTSD: Catecholamines and serotonin. Semin Clin Neuropsychiatry. 1999;4(4):242-248. [DOI] [PubMed] [Google Scholar]
  • 48.Lokshina Y, Nickelsen T, Liberzon I. Reward processing and circuit dysregulation in post-traumatic stress disorder. Front Psychiatry. 2021;12:559401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Averill LA, Purohit P, Averill CL, et al. Glutamate dysregulation and glutamatergic therapeutics for PTSD: Evidence from human studies. Neurosci Lett. 2017;649:147-155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Shen J, Tomar JS. Elevated brain glutamate levels in bipolar disorder and pyruvate carboxylase-mediated anaplerosis. Front Psychiatry. 2021;12:640977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Inoshita M, Umehara H, Watanabe SY, et al. Elevated peripheral blood glutamate levels in major depressive disorder. Neuropsychiatr Dis Treat. 2018;14:945-953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kent JM, Coplan JD, Gorman JM. Clinical utility of the selective serotonin reuptake inhibitors in the spectrum of anxiety. Biol Psychiatry. 1998;44(9):812-824. [DOI] [PubMed] [Google Scholar]
  • 53.Alexander W. Pharmacotherapy for post-traumatic stress disorder in combat veterans: Focus on antidepressants and atypical antipsychotic agents. P T. 2012;37(1):32-38. [PMC free article] [PubMed] [Google Scholar]
  • 54.Stahl SM. Mechanism of action of serotonin selective reuptake inhibitors. Serotonin receptors and pathways mediate therapeutic effects and side effects. J Affect Disord. 1998;51(3):215-235. [DOI] [PubMed] [Google Scholar]
  • 55.Xue W, Wang P, Li B, et al. Identification of the inhibitory mechanism of FDA approved selective serotonin reuptake inhibitors: An insight from molecular dynamics simulation study. Phys Chem Chem Phys. 2016;18(4):3260-3271. [DOI] [PubMed] [Google Scholar]
  • 56.Lin J, Liu W, Guan J, et al. Latest updates on the serotonergic system in depression and anxiety. Front Synaptic Neurosci. 2023;15:1124112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Roepke S, Schoofs N, Priebe K, et al. Treating nightmares in post-traumatic stress disorder with dronabinol: Study protocol of a multicenter randomised controlled study (THC PTSD-trial). BMC Psychiatry. 2023;23(1):319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Burek GA, Waite MR, Heslin K, et al. Low-dose clonidine in veterans with post-traumatic stress disorder. J Psychiatr Res. 2021;137:480-485. [DOI] [PubMed] [Google Scholar]
  • 59.Detweiler MB, Pagadala B, Candelario J, et al. Treatment of post-traumatic stress disorder nightmares at a veterans affairs medical center. J Clin Med. 2016;5(12):117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Marazziti D, Carmassi C, Cappellato G, et al. Novel pharmacological targets of post-traumatic stress disorders. Life (Basel). 2023;13(8):1731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Giovannitti Jr JA, Thoms SM, Crawford JJ. Alpha-2 adrenergic receptor agonists: A review of current clinical applications. Anesth Prog. 2015;62(1):31-39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Richards A, Inslicht S, Ruoff LM, et al. An open-label study of doxazosin extended-release for PTSD: Findings and recommendations for future research on doxazosin. Focus (Am Psychiatr Publ). 2018;16(1):67-73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Back SE, Flanagan JC, Jones JL, et al. Doxazosin for the treatment of co-occurring PTSD and alcohol use disorder: Design and methodology of a randomised controlled trial in military veterans. Contemp Clin Trials. 2018;73:8-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Sethi R, Vasudeva S. Doxazosin for the treatment of nightmares: Does it really work? A case report. Prim Care Companion CNS Disord. 2012;14(5):PCC.12l01356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Rodgman C, Verrico CD, Holst M, et al. Doxazosin XL reduces symptoms of post-traumatic stress disorder in veterans with PTSD: A pilot clinical trial. J Clin Psychiatry. 2016;77(5):e561-e565. [DOI] [PubMed] [Google Scholar]
  • 66.De Jong J, Wauben P, Huijbrechts I, et al. Doxazosin treatment for post-traumatic stress disorder. J Clin Psychopharmacol. 2010;30(1):84-85. [DOI] [PubMed] [Google Scholar]
  • 67.Roepke S, Danker-Hopfe H, Repantis D, et al. Doxazosin, an α-1-adrenergic-receptor antagonist, for nightmares in patients with post-traumatic stress disorder and/or borderline personality disorder: A chart review. Pharmacopsychiatry. 2017;50(1):26-31. [DOI] [PubMed] [Google Scholar]
  • 68.Raut SB, Canales JJ, Ravindran M, et al. Effects of propranolol on the modification of trauma memory reconsolidation in PTSD patients: A systematic review and meta-analysis. J Psychiatr Res. 2022;150:246-256. [DOI] [PubMed] [Google Scholar]
  • 69.Mueller HS, Ayres SM. Propranolol decreases sympathetic nervous activity reflected by plasma catecholamines during evolution of myocardial infarction in man. J Clin Invest. 1980;65(2):338-346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Hoge EA, Worthington JJ, Nagurney JT, et al. Effect of acute posttrauma propranolol on PTSD outcome and physiological responses during script-driven imagery. CNS Neurosci Ther. 2012;18(1):21-27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Argolo FC, Cavalcanti-Ribeiro P, Netto LR, Quarantini LC. Prevention of post-traumatic stress disorder with propranolol: A meta-analytic review. J Psychosom Res. 2015;79(2):89-93. [DOI] [PubMed] [Google Scholar]
  • 72.Peskind ET, Bonner LT, Hoff DJ, Raskind MA. Prazosin reduces trauma-related nightmares in older men with chronic post-traumatic stress disorder. J Geriatr Psychiatry Neurol. 2003;16:165-171. [DOI] [PubMed] [Google Scholar]
  • 73.Raskind MA, Dobie DJ, Kanter ED, et al. The alpha1-adrenergic antagonist prazosin ameliorates combat trauma nightmares in veterans with post-traumatic stress disorder: A report of 4 cases. J Clin Psychiatry. 2000;61:129-134. [DOI] [PubMed] [Google Scholar]
  • 74.Ahmadpanaha M, Sabzeieea P, Hosseini SM, et al. Comparing the effect of prazosin and hydroxyzine on sleep quality in patients suffering from post-traumatic stress disorder. Neuropsychobiology. 2014;69:235-242. [DOI] [PubMed] [Google Scholar]
  • 75.Taylor F, Raskind MA. The alpha1-adrenergic antagonist prazosin improves sleep and nightmares in civilian trauma post-traumatic stress disorder. J Clin Psychopharmacol. 2002;22(1):82-85. [DOI] [PubMed] [Google Scholar]
  • 76.Gautam CS, Mahajan SS, Sharma J, et al. Repurposing potential of ketamine: Opportunities and challenges. Indian J Psychol Med. 2020;42(1):22-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Zorumski CF, Izumi Y, Mennerick S. Ketamine: NMDA receptors and beyond. J Neurosci. 2016;36(44):11158-11164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Chowdhury GM, Behar KL, Cho W, et al. ¹H-[¹³C]-nuclear magnetic resonance spectroscopy measures of ketamine's effect on amino acid neurotransmitter metabolism. Biol Psychiatry. 2012;71(11):1022-1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Feder A, Costi S, Rutter SB, et al. A randomised controlled trial of repeated ketamine administration for chronic post-traumatic stress disorder. Focus (Am Psychiatr Publ). 2023;21(3):296-305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Villarreal G, Hamner MB, Qualls C, Cañive JM. Characterising the effects of quetiapine in military post-traumatic stress disorder. Psychopharmacol Bull. 2018;48(2):8-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Monnelly EP, Ciraulo DA, Knapp C, Keane T. Low-dose risperidone as adjunctive therapy for irritable aggression in post-traumatic stress disorder. J Clin Psychopharmacol. 2003;23(2):193-196. [DOI] [PubMed] [Google Scholar]
  • 82.Krishnamurthy S, Garabadu D, Joy KP. Risperidone ameliorates post-traumatic stress disorder-like symptoms in modified stress re-stress model. Neuropharmacology. 2013;75:62-77. [DOI] [PubMed] [Google Scholar]
  • 83.Sattar SP, Ucci B, Grant K, et al. Quetiapine therapy for post-traumatic stress disorder. Ann Pharmacother. 2002;36(12):1875-1878. [DOI] [PubMed] [Google Scholar]
  • 84.Hamner MB, Deitsch SE, Brodrick PS, et al. Quetiapine treatment in patients with post-traumatic stress disorder: An open trial of adjunctive therapy. J Clin Psycho-Pharmacol. 2003;23:15-20. [DOI] [PubMed] [Google Scholar]
  • 85.Filteau MJ, Leblanc J, Bouchard RH. Quetiapine reduces flashbacks in chronic post-traumatic stress disorder. Can J Psychiatry. 2003;48:282-283. [DOI] [PubMed] [Google Scholar]
  • 86.Hertzberg MA, Butterfield MI, Feldman ME, et al. A preliminary study of lamotrigine for the treatment of post-traumatic stress disorder. Biol Psychiatry. 1999;45(9):1226-1229. [DOI] [PubMed] [Google Scholar]
  • 87.Costa B, Vale N. Understanding lamotrigine's role in the CNS and possible future evolution. Int J Mol Sci. 2023;24(7):6050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Petty F, Davis LL, Nugent AL, et al. Valproate therapy for chronic, combat-induced post-traumatic stress disorder. J Clin Psychopharmacol. 2002;22(1):100-101. [DOI] [PubMed] [Google Scholar]
  • 89.Otte C, Wiedemann K, Yassouridis A, Kellner M. Valproate monotherapy in the treatment of civilian patients with non-combat-related post-traumatic stress disorder: An open-label study. J Clin Psychopharmacol. 2004;24(1):106-108. [DOI] [PubMed] [Google Scholar]
  • 90.Marshall RD, Beebe KL, Oldham M, Zaninelli R. Efficacy and safety of paroxetine treatment for chronic PTSD: A fixed-dose, placebo-controlled study. Am J Psychiatry. 2001;158(12):1982-1988. [DOI] [PubMed] [Google Scholar]
  • 91.Kucukalić A, Bravo-Mehmedbasić A, Dzubur-Kulenović A. Paroxetine in the treatment of post traumatic stress disorder: Our experiences. Bosn J Basic Med Sci. 2008;8(1):76-79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Brady K, Pearlstein T, Asnis GM, et al. Efficacy and safety of sertraline treatment of post-traumatic stress disorder: A randomised controlled trial. JAMA. 2000;283(14):1837-1844. [DOI] [PubMed] [Google Scholar]
  • 93.Gerst A, Murthy T, Elson A, et al. Adjunctive minocycline for treatment of post-traumatic stress disorder. WMJ. 2021;120(4):321-324. [PubMed] [Google Scholar]
  • 94.Schelling G, Kilger E, Roozendaal B, et al. Stress doses of hydrocortisone, traumatic memories, and symptoms of post-traumatic stress disorder in patients after cardiac surgery: A randomised study. Biol Psychiatry. 2004;55(6):627-633. [DOI] [PubMed] [Google Scholar]
  • 95.Delahanty DL, Gabert-Quillen C, Ostrowski SA, et al. The efficacy of initial hydrocortisone administration at preventing post-traumatic distress in adult trauma patients: A randomised trial. CNS Spectr. 2013;18(2):103-111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.MacCallum PE, Cooze JB, Ward J, et al. Evaluating the effects of single, multiple, and delayed systemic rapamycin injections to contextual fear reconsolidation: Implications for the neurobiology of memory and the treatment of PTSD-like reexperiencing. Behav Brain Res. 2024;461:114855. [DOI] [PubMed] [Google Scholar]
  • 97.Halabitska I, Petakh P, Lushchak O, et al. Metformin in antiviral therapy: Evidence and perspectives. Viruses. 2024;16(12):1938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kamyshnyi O, Matskevych V, Lenchuk T, et al. Metformin to decrease COVID-19 severity and mortality: Molecular mechanisms and therapeutic potential. Biomed Pharmacother. 2021;144:112230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Wang J, Xiao B, Han F, Shi Y. Metformin alleviated the neuronal oxidative stress in hippocampus of rats under single prolonged stress. J Mol Neurosci. 2017;63(1):28-35. [DOI] [PubMed] [Google Scholar]
  • 100.Liu S, Raines AM, Yoshida Y, et al. 174-LB: Association between metformin treatment and improved symptoms of post-traumatic stress disorder. Diabetes. 2020;69:174-LB. [Google Scholar]
  • 101.Brady KT, Killeen TK, Brewerton T, Lucerini S. Comorbidity of psychiatric disorders and post-traumatic stress disorder. J Clin Psychiatry. 2000;61(Suppl 7):22-32. [PubMed] [Google Scholar]

Articles from Chronic Stress are provided here courtesy of SAGE Publications

RESOURCES