Next Article in Journal
Melissa officinalis L. Aqueous Extract Exerts Antioxidant and Antiangiogenic Effects and Improves Physiological Skin Parameters
Previous Article in Journal
Synthesis and Single Crystal Structures of N-Substituted Benzamides and Their Chemoselective Selenation/Reduction Derivatives
Previous Article in Special Issue
Cyclooxygenase Inhibition Safety and Efficacy in Inflammation-Based Psychiatric Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anti-TNF-α Compounds as a Treatment for Depression

1
Department of Clinical Biochemistry and Pharmacology, School for Community Health Professions—Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel
2
Department of Nursing, School for Community Health Professions—Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel
*
Author to whom correspondence should be addressed.
Submission received: 22 March 2021 / Revised: 14 April 2021 / Accepted: 17 April 2021 / Published: 19 April 2021
(This article belongs to the Special Issue Anti-inflammatory Drugs as a Treatment for Psychiatric Disorders)

Abstract

:
Millions of people around the world suffer from psychiatric illnesses, causing unbearable burden and immense distress to patients and their families. Accumulating evidence suggests that inflammation may contribute to the pathophysiology of psychiatric disorders such as major depression and bipolar disorder. Copious studies have consistently shown that patients with mood disorders have increased levels of plasma tumor necrosis factor (TNF)-α. Given these findings, selective anti-TNF-α compounds were tested as a potential therapeutic strategy for mood disorders. This mini-review summarizes the results of studies that examined the mood-modulating effects of anti-TNF-α drugs.

1. Mood Disorders

Millions of people around the world suffer from psychiatric illnesses, causing unbearable burden and immense distress to patients and their families [1]. Moreover, psychiatric disorders are associated with extensive financial costs to patients, the health care system and society in general [2,3]. Patients with mood disorders such as bipolar disorder and depressive disorders are of a higher likelihood to suffer from suicidal death and various comorbidities, leading to increased mortality rates in comparison to matched-healthy subjects [4,5,6]. The lifetime prevalence of bipolar disorder in the general population is between 0.7–1.5% [7,8] and that of depressive disorders is between 10–20% [1,9]. These estimations likely depict only a fraction of the true numbers, suggesting that there are presumably myriads of concealed and undiagnosed cases, and acknowledges that there is societal and cultural variance in recognition and interpretation of psychiatric symptoms [1,10].
Bipolar disorder is recognized as one of the most complex and difficult-to-treat psychiatric illnesses. Patients with bipolar disorder suffer alternating periods of mania and depression [11,12]. Mania is characterized by euphoric mood, impaired judgment, hyperactivity and excitement, increased erotic thoughts and engagement in sexual activity, among other features [11,12].
Depression is a rampant and devastating mental disorder [1,9], and is more prevalent in women than in men [1]. Melancholy is the primary feature/manifestation of depression [13,14,15,16]. Patients with depression may have alternative or accompanying symptoms including anxiety, low self-esteem, changes in appetite, social isolation, diminished interest in hedonic activities, insomnia or hypersomnia, and suicidal thoughts and/or attempts, among others [13,14,15,16]. Expectedly, the severity of symptoms and duration of depressive episodes vary significantly and, understandably, depressive episodes can impact even the most basic aspects of patients’ lives. Occasionally, depression presents without a known triggering cause. However, sometimes a prominent emotional stimulus, such as a death of a close relative, precedes the inception of depression.
The most widely used treatment strategy for bipolar disorder is pharmacotherapy [11,12,17]. Other approaches include electroconvulsive therapy [18,19] and cognitive behavioral therapy [20]. Similarly, pharmacotherapy, psychotherapy and electroconvulsive therapy are the three most frequently used treatments for depressive disorders [17,18,21,22,23,24]. Among these, pharmacotherapy is the most common and it includes a wide variety of medications [23,24]. The treatment of depressive disorders is dictated by a number of factors including: (i) risk of suicide, (ii) the patient’s ability to understand and follow instructions (adherence to treatment), (iii) level of supportive resources, (iv) level of encountered stressors, and, (v) level of functional impairment [17,24].
The availability of abundant and diverse medication options available for the treatment of mood disorders notwithstanding, a high proportion of patients present a poor response to treatment [11,12,14,17,22,23,24]. Moreover, many patients suffer a plethora of unpleasant side effects (some of which may be severe and irreversible) further encouraging poor compliance to treatment [11,12,14,17,22,23,24,25,26,27]. These limitations accentuate the necessity for new treatment strategies for mood disorders in an effort to supply hope for additional sub-groups of patients.

2. Tumor Necrosis Factor (TNF)-α

TNF-α is a multi-functional cytokine which plays central roles in numerous physiological as well as pathological processes in mammals [28,29,30,31]. It was recognized early on for its ability to induce necrosis of tumor cells [32], but was subsequently associated with plentiful biological functions [28,29,30,31]. TNF-α is synthetized and secreted mainly by macrophages though several cell types (including glia cells and neurons in the brain) are capable of producing it [28,29,30,31,32,33,34,35]. Newly synthesized TNF-α localizes in cell membrane until it undergoes proteolytic cleavage by TNF-α-converting enzyme, which releases the soluble form of the protein [36,37] (see Figure 1 for illustration). Both the transmembrane and the soluble form of the protein are biologically active—binding to and activating TNF receptor 1 (TNFR1) as well as TNFR2 [30,31,38,39] (Figure 1). TNFR1 and TNFR2 share some similar functions (e.g., advancement of immune defense mechanisms, induction of inflammation, and promotion of cell proliferation and survival) but, they also have distinct, sometimes opposite, biological activities [30,31,38,39]. Principally, TNFR1 is connected to pathological processes such as inflammation, apoptosis and necrosis, while TNFR2 is mostly linked to physiological responses such as host defense, tissue repair and regeneration [30,31,38,39]. However, delineating these receptors with distinctive pathological versus physiological tasks would be an over-simplification of a more complex biological reality.
Thorough research has indicated TNF-α to be mostly linked to immune and inflammatory functions [30,31]. It has also been associated with cancer pathophysiology [29]. It is involved in various immune and inflammatory responses (usually acting as a pro-inflammatory mediator) contributing to host defense [30,31,38,39]. Under certain conditions, TNF-α facilitates apoptosis and cell death especially in cancer cells [29,30,31,38,39]. Nevertheless, and despite its common association with pathological conditions, TNF-α plays a crucial role in numerous physiological processes, particularly in the central nervous system (CNS—the brain and the spinal cord) [28,39]. For example, in the brain, TNF-α has a direct impact on neuronal function and survival, regulating production and secretion of neurotransmitters, controlling synaptic transmission, and contributing to myelin synthesis and preservation [28,39,40,41,42,43,44,45]. TNF-α was found to increase the permeability of the blood-brain barrier (BBB) which is accompanied by depressive behavior [46,47,48]. Dysfunction of the BBB hastens the penetration of inflammatory mediators and peripheral immune cells into the CNS leading to behavioral abnormalities and mood disorders [49,50]. Thus, taking into account the various crucial functions of TNF-α, it is expected that disruption of its activity would cause profound biological consequences, including alteration of neurological function.

3. Brain Inflammation, TNF-α and Mood Disorders

The CNS consists of two main types of cells: neurons and glia cells [33,34]. There are three types of glia cells: astrocytes, microglia, and oligodendrocytes [33,34]. The role of microglia cells in the CNS is comparable to that of macrophages in peripheral tissues. Astrocytes have important immune-inflammatory roles, and support the function and survival of neurons [33,34,51]. Oligodendrocytes produce myelin, the insulating substance that surrounds nerve cell axons. Microglia and astrocytes are involved in various neuro-inflammatory processes and are associated with numerous CNS pathologies [28,34,35,51,52,53,54]. Despite the presence of the BBB, the activity of the “peripheral” immune system still manages to impact the CNS. It has been consistently recognized that illnesses associated with systemic inflammation (e.g., rheumatoid arthritis and coronary artery disease) frequently present with behavioral abnormalities and symptoms of depression. Systemic inflammatory responses to infectious agents affect brain function and, in turn, evoke significant changes in behavior [54]. This association has revealed itself to be more than just a speculation, as even early studies suggested that dysregulation of the immune system may lead to depression [55,56]. Subsequently, many studies reported that immune-dysregulation and inflammation contribute to the pathophysiology of mood disorders. It was found that patients with depression had elevated levels of pro-inflammatory markers [57,58,59,60,61,62,63,64,65,66,67,68,69,70], while levels of anti-inflammatory mediators were either comparable [71,72] or lower [73] than those in control subjects. Bipolar patients were also reported to have abnormal levels of various inflammatory mediators [59,72,74,75,76,77,78,79,80,81,82,83,84,85]. In particular, numerous studies reported that TNF-α levels are elevated in patients with major depression [56,59,60,61,62,69,70,86] and bipolar disorder [59,72,74,75,76,78,79,80,81,82,83,84,85]. Abnormalities in TNF-α levels have been shown to influence the severity of psychiatric symptoms and response to treatment. For example, a recent study showed that elevated baseline plasma TNF-α levels in patients with major depression may predict a better improvement in intensity of suicidal thoughts [86]. Patients with bipolar disorder [87] and depression [88] were reported to have altered levels of TNFR1 and TNFR2, respectively. Interestingly, the latter two studies [87,88] did not demonstrate abnormal TNF-α levels among their population. However, despite the large body of data attesting for alterations in inflammatory mediator levels among patients with mood disorders, some studies reported opposite findings [80,85].
Furthermore, the “inflammation hypothesis” of mood disorders was strengthened by data that showed that psychotropic drugs possess anti-inflammatory effects. Antidepressants, mood stabilizers and antipsychotic drugs were reported to have anti-inflammatory effects which may contribute to their therapeutic efficacy [67,70,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107]. For example, Li et al. [98] reported that the mood stabilizer lithium reduced levels of TNF-α in patients with acute manic episodes. Valproate, another mood stabilizer, reduced the secretion of interleukin (IL)-6 and TNF-α production in vitro [108]. Similarly, various antidepressants were found to have potent anti-inflammatory effects [68,70,99,109,110]. This outcome is exemplified by the selective serotonin reuptake inhibitor fluoxetine which significantly decreased plasma IL-6 levels in patients with acute depression [111]. Antipsychotic drugs also exhibited anti-inflammatory effects [89,93,94,102,104,106]. This response can be seen in second generation antipsychotic drugs that decreased lipopolysaccharide-induced synthesis of IL-6 and TNF-α and increased the levels of the anti-inflammatory cytokine IL-10 in mice [102]. In contrast to these findings, some studies showed that psychotropic drugs exhibit pro-inflammatory effects in certain circumstances [64,89,104,107,112,113,114,115,116].
Additional support for the inflammation hypothesis of mood disorders came from studies that showed that treatment with various anti-inflammatory/immune-modulating drugs reduced symptom severity and improved conditions of patients with mood disorders [58,117,118,119,120,121,122,123]. Mainly, selective cyclooxygenase-2 inhibitors (e.g., celecoxib) were found beneficial as add-on therapy to psychotropic drugs in patients with mood disorders [58,120]. Nevertheless, here too, studies published negative findings regarding the effectiveness of anti-inflammatory/immune-modulating medications as a treatment for mood disorders [124,125]. Among the various anti-inflammatory drugs that have been explored as a potential treatment for mood disorders, selective TNF-α antagonists were given special attention. The following section summarizes the mood-modulating effects of clinically used anti-TNF-α compounds.

4. Anti-TNF-α as a Treatment for Mood Disorders

As summarized above, a large body of data suggested that out of the inflammatory mediators that have been linked to the pathophysiology of mood disorders, TNF-α in particular exhibited a seemingly significant role [56,57,58,59,69,70,71,78,80,84,86,90,126]. This was the basis for investigating the mood-modulating effects of selective anti-TNF-α compounds. Several selective anti-TNF-α compounds were developed and introduced for clinical use, typically for the treatment of immune-inflammation-related disorders such as rheumatoid arthritis, ankylosing spondylitis, psoriasis, inflammatory bowel diseases (e.g., Crohn’s disease), and hidradenitis suppurativa, among others [30,127,128,129,130,131,132,133,134,135,136,137,138,139,140]. The following paragraphs summarize the results of studies that tested the mood-modulating effects of anti-TNF-α compounds.

4.1. Search Strategy

The search strategy was based on surveying the following electronic databases for inclusive criteria: PubMed, Web of Science, and Google Scholar, for English language papers published in peer-reviewed journals reporting on the use of anti-TNF-a compounds in subjects with mood disorders. The customized search was restricted to the years 1990 (the year when the first report on the anti-TNF-a activity and beneficial therapeutic effects of infliximab was published [141]) to 2020. The search field contained the name of each compound, including: infliximab, etanercept, onercept, adalimumab, golimumab, humicade, certolizumab pegol, and pentoxifylline; together with each of the following keywords: depression, melancholia, depressive disorder, mania, bipolar disorder, manic-depressive illness. The search strategy resulted in many hits that were irrelevant to the purpose of the article. On the other hand, no relevant papers reporting on the effects of onercept, golimumab, humicade and certolizumab pegol in subjects with mood disorders were found. We included most relevant papers reporting on animal studies and almost all papers reporting on studies conducted in human subjects, because the latter were the main focus of the manuscript.

4.2. Infliximab

Infliximab is a chimeric TNF-α-specific neutralizing monoclonal antibody consisting of a human IgG Fc region and a murine Fv region (see Figure 2 for illustration). It is recognized as a potent selective TNF-α antagonist with powerful neutralizing effects against soluble TNF-α and, to a lesser extent, on transmembrane TNF-α [133,142,143,144]. Infliximab is capable of binding to both monomeric and trimeric forms of soluble TNF-α. Each infliximab molecule can bind to two TNF-α molecules, while a single TNF-α homotrimer can bind to up to three infliximab molecules [133,142,143,144]. Infliximab is administered intravenously and thus has a maximized (100%) bioavailability; it has a low clearance rate (~ 11 mL/hour) and a plasma half-life of nearly 8–10 days [133,143]. Infliximab has been used for the treatment of various rheumatoid and inflammatory-associated diseases such as rheumatoid arthritis, psoriasis, ankylosing spondylitis, and Crohn’s disease, among others [30,133]. Several studies examined the effects of infliximab on depressive symptoms among patients with Crohn’s disease [134,135] and ankylosing spondylitis [136,145,146] revealing encouraging results. Animal studies also demonstrated an antidepressant-like effect for infliximab [147,148]. Raison et al. [149] evaluated the antidepressant effect of infliximab in patients with treatment-resistant depression. Sixty patients were randomly allocated to receive either infliximab (n = 30) or a placebo (n = 30). Infliximab showed a significant therapeutic effect—mitigated depressive symptoms—but only in patients who had increased levels of inflammatory markers [149]. Consistent with these results, a recent meta-analysis study which evaluated the antidepressant efficacy of infliximab revealed that it was effective exclusively in patients with elevated levels of inflammatory markers such TNF-α and C-reactive protein [150]. The efficacy of infliximab was also tested in patients with bipolar depression [151,152,153,154]. McIntyre et al. [151] conducted a randomized, double-blind, placebo-controlled trial in which 29 patients were treated with infliximab and 31 patients with a placebo. Twelve weeks of infliximab treatment did not cause a significant reduction in severity of depressive symptoms. Only in a sub-group of patients with a history of childhood physical abuse infliximab (as compared to the placebo) led to a significant depletion in depressive symptoms [151]. Lee et al. [152] conducted a randomized, double-blind trial of adjunctive treatment with infliximab (together with standard pharmacotherapy) and a placebo for 12 weeks in patients with bipolar depression. They reported a significant improvement in a measure of anhedonia in infliximab-treated patients; however, the positive effect was short-lived and did not show sustainable positive results, dissipating within six weeks after the final infusion of the drug. Mansur et al. also reported positive therapeutic effects of infliximab on depressive symptoms [153] and cognitive function [154] in patients with bipolar depression. A recent study by the same group of investigators also demonstrated beneficial effects of infliximab on bipolar patients [155]. In a 12-week, randomized, double-blind trial, infliximab treatment was associated with a significant decrease in prefrontal levels of glutamate and a cognitive improvement in patients with bipolar depression [155]. Together, these findings (see summary of the findings in Table 1) suggest that infliximab produces antidepressant effects in particular sub-groups of depressive patients.

4.3. Etanercept

Etanercept is a human recombinant fusion protein of TNFR2 that neutralizes/inhibits TNF-α activity [30] (Figure 2). It is regarded as a less powerful TNF-α antagonist when compared to infliximab, but similarly to infliximab, it has a much stronger antagonizing effect against soluble TNF-α than transmembrane TNF-α [133,142,143,144]. Etanercept binds only to the trimeric form of soluble TNF-α and each etanercept molecule is capable of binding to one TNF-α molecule [133,142,143,144]. Etanercept is administered subcutaneously and has a bioavailability of nearly 75%; it has a relatively high but varying clearance rate (80–240 mL/hour) and a plasma half-life of 3–5.5 days [133,143]. Early pre-clinical studies showed that etanercept reduced depressive-like behavior in rats [156,157]. More recently, a study in rats showed that etanercept significantly decreased depressive-like behavior and improved cognitive function [158]. Similarly, a study in mice showed that etanercept exerted a potent antidepressant-like effect and an anxiolytic-like effect [159]. In line with these pre-clinical results, etanercept was found to significantly decrease the severity of fatigue, depression and anxiety symptoms among patients with psoriasis (Table 1) [137,138,160,161]. Moreover, non-randomized trials showed that addition of etanercept to standard therapy significantly reduced depressive and anxiety symptoms among patients with psoriasis [162,163,164] and rheumatoid arthritis [165,166]. For example, a prospective cohort study by Yang et al. [167] demonstrated that addition of etanercept to standard treatment was associated with a sustained significant reduction in depression and anxiety symptoms in psoriasis patients. In contrast to these findings, a study in patients with rheumatoid arthritis found that addition of etanercept to methotrexate (an immune-modulating drug) did not significantly improve depressive and anxiety symptoms [139]. Collectively, these results suggest that etanercept exhibits antidepressant and anxiolytic effects at least in some sub-groups of patients.

4.4. Adalimumab

Adalimumab is another human TNF-α-specific neutralizing monoclonal antibody (Figure 2). It has similar pharmacokinetic properties to infliximab. Each adalimumab molecule can bind to two TNF-α molecules, while a single TNF-α homotrimer can bind to up to three adalimumab molecules [133,142,143,144]. Adalimumab is administered subcutaneously and has a bioavailability of nearly 65%; it has a low clearance rate (~12 mL/hour) and a long but variable plasma half-life ranging from 10 to 20 days [133,143]. Randomized and non-randomized clinical trials showed that adalimumab exerts antidepressant and anxiolytic effects when administered to patients with chronic physical illnesses such as Crohn’s disease [140], psoriasis [128,129,168,169,170] and hidradenitis suppurativa [130] (Table 1). To the best of our knowledge, the mood-modulating effects of adalimumab have not been directly tested in psychiatric patients with mood disorders.

4.5. Pentoxifylline

Pentoxifylline is a methylxanthine drug (Figure 2) that for many years has been used for the treatment of different clinical conditions such as peripheral vascular disease [171,172], idiopathic and ischemic cardiomyopathy [173,174,175], coronary artery disease [176], chronic kidney disease [177], alcoholic hepatitis [178], among other illnesses [171,179,180]. Pentoxifylline is administered orally and has a relatively high bioavailability, depending on the used formulation [160]. It has a low binding rate to plasma proteins (minimizing the chance for drug-drug interactions) and distributes vastly throughout body tissues, extending to the brain. Pentoxifylline undergoes extensive metabolism (mainly through reduction and oxidation) and has a short plasma half-life ranging between 1 to 4 h, again, depending on the used formulation [160]. The therapeutic efficacy of pentoxifylline in the treatment of peripheral vascular disease seems to be derived from its ability to improve the deformability of red blood cells, decrease blood fibrinogen levels and inhibit platelet aggregation [172]. Moreover, pentoxifylline inhibits the enzyme phosphodiesterase [181]. In the context of the present article, pentoxifylline is recognized as a potent inhibitor of TNF-α [173,174,175,176,177,179,181,182,183,184,185,186]. Numerous studies showed that pentoxifylline inhibits the production of TNF-α in vitro and in vivo (in animals and humans) [173,174,175,176,177,179,181,182,183,184,185,186]. Thus, pentoxifylline is regarded as a strong non-selective TNF-α inhibitor (as it exerts other pharmacological properties).
Owing to the large body of data which linked TNF-α to the pathophysiology of depression, many pre-clinical studies have investigated the antidepressant potential of pentoxifylline [182,183,187]. Bah et al. [187] demonstrated that pentoxifylline exerted antidepressant-like effects in rats that were subjected to an experimental model of myocardial infarction. Pentoxifylline significantly increased sucrose preference and significantly decreased immobility time (both indicative of an antidepressant-like effect) in the forced swim test in post-infarction rats [187]. Mohamed et al. [182] observed that treatment with pentoxifylline for three weeks significantly increased sucrose preference in rats that were subjected to a chronic mild stress protocol. The chronic mild stress paradigm is used to induce depressive-like phenotypes in animals. Another study showed that pentoxifylline significantly decreased immobility time in rats that were exposed both to an inflammatory stimulus (lipopolysaccharide) and chronic mild stress [183]. Collectively, these studies [182,183,187] (among others) demonstrated that pentoxifylline has strong antidepressant-like effects in various behavioral models including the sucrose preference test and the forced swim test [182,183,187]. Consistent with these positive pre-clinical results, a randomized, double-blind, placebo-controlled clinical trial showed that adjunctive pentoxifylline treatment was associated with a significant anti-depressive effect [188]. Addition of pentoxifylline (400 mg/day) to escitalopram (20 mg/day) for 12 weeks significantly reduced depressive symptoms in patients with major depression [188]. Moreover, pentoxifylline caused a significant decrease in plasma TNF-α and IL-6 levels (suggestive of a potent anti-inflammatory effect) and a significant increase in plasma serotonin and brain-derived neurotrophic factor levels (suggestive of favorable behavioral/neuroprotective biochemical effects) [188]. These encouraging findings underscore the need for more randomized trials of pentoxifylline in patients with mood disorders.

5. Summary

Several clinical trials attested for the antidepressant efficacy of anti-TNF-α compounds (in patients with medical illnesses, major depression, or bipolar depression) [70]. Selective TNF-α antagonists such as infliximab and etanercept showed favorable neurological/antidepressant effects in specific sub-groups of patients. However, it is important to emphasize that most of the available data regarding the antidepressant effects of selective TNF-α antagonists is derived from studies in non-psychiatric patients (i.e., patients with inflammatory-associated diseases who presented depressive symptoms). Moreover, some evidence suggests that there is no connection between anti-TNF-α therapy and improvement in mood symptoms [139,150,151]. Therefore, new randomized, placebo-controlled clinical trials are necessary for direct examination of the mood-modulating effects of TNF-α antagonists in patients with mood disorders. In this regard, recently, concerns have been raised regarding the efficacy of selective TNF-α antagonists as a therapeutic strategy for mood disorders [139,151,189,190]. It is important to mention that most clinically available anti-TNF-α compounds possess low-to-null ability to cross the BBB, mainly due to their large molecular weight [191,192,193]. This suggests that the reported beneficial behavioral (antidepressant) effects of these compounds are derived from peripheral inhibition of TNF-α activity rather than a direct effect on the brain. Potent peripheral inhibition of TNF-α activity may be sufficient for diminishing brain inflammation. Therefore, it is important to continue studying the therapeutic mechanism of action and effectiveness of selective TNF-α antagonists as a treatment for mood disorders.

Author Contributions

S.U. read and summarized some of the papers cited in the manuscript, participated in the design and preparation of the figures, and participated in the writing of the manuscript. A.N.A. designed the manuscript, read and summarized some of the papers cited in the manuscript, participated in the design and preparation of the figures, and participated in the writing and submission of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Whiteford, H.A.; Degenhardt, L.; Rehm, J.; Baxter, A.J.; Ferrari, A.J.; Erskine, H.E.; Charlson, F.J.; Norman, R.E.; Flaxman, A.D.; Johns, N.; et al. Global burden of disease attributable to mental and substance use disorders: Findings from the Global Burden of Disease Study 2010. Lancet 2013, 382, 1575–1586. [Google Scholar] [CrossRef]
  2. Walker, E.R.; McGee, R.E.; Druss, B.G. Mortality in mental disorders and global disease burden implications a systematic review and meta-analysis. JAMA Psychiatry 2015, 72, 334–341. [Google Scholar] [CrossRef]
  3. Islek, D.; Kilic, B.; Akdede, B.B. Out-of-pocket health expenditures in patients with bipolar disorder, anxiety, schizophrenia and other psychotic disorders: Findings from a study in a psychiatry outpatient clinic in Turkey. Soc. Psychiatry Psychiatr. Epidemiol. 2018, 53, 151–160. [Google Scholar] [CrossRef] [PubMed]
  4. Bostwick, J.M.; Pankratz, V.S. Affective disorders and suicide risk: A reexamination. Am. J. Psychiatry 2000, 157, 1925–1932. [Google Scholar] [CrossRef]
  5. Crump, C.; Sundquist, K.; Winkleby, M.A.; Sundquist, J. Comorbidities and mortality in bipolar disorder: A Swedish national cohort study. JAMA Psychiatry 2013, 70, 931–939. [Google Scholar] [CrossRef]
  6. Baldessarini, R.J.; Tondo, L. Suicidal Risks in 12 DSM-5 Psychiatric Disorders. J. Affect. Disord. 2020, 271, 66–73. [Google Scholar] [CrossRef]
  7. Narrow, W.E.; Rae, D.S.; Robins, L.N.; Regier, D.A. Revised prevalence estimates of mental disorders in the United States: Using a clinical significance criterion to reconcile 2 surveys’ estimates. Arch. Gen. Psychiatry 2002, 59, 115–123. [Google Scholar] [CrossRef] [Green Version]
  8. Ferrari, A.J.; Stockings, E.; Khoo, J.P.; Erskine, H.E.; Degenhardt, L.; Vos, T.; Whiteford, H.A. The prevalence and burden of bipolar disorder: Findings from the Global Burden of Disease Study 2013. Bipolar Disord. 2016, 18, 440–450. [Google Scholar] [CrossRef] [PubMed]
  9. Hirschfeld, R.M.A. The epidemiology of depression and the evolution of treatment. J. Clin. Psychiatry 2012, 73, 5–9. [Google Scholar] [CrossRef] [PubMed]
  10. Ferrari, A.J.; Charlson, F.J.; Norman, R.E.; Flaxman, A.D.; Patten, S.B.; Vos, T.; Whiteford, H.A. The Epidemiological Modelling of Major Depressive Disorder: Application for the Global Burden of Disease Study 2010. PLoS ONE 2013, 8, e69637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Belmaker, R.H. Bipolar disorder. N. Engl. J. Med. 2004, 351, 476–486. [Google Scholar] [CrossRef]
  12. Grande, I.; Berk, M.; Birmaher, B.; Vieta, E. Bipolar disorder. Lancet 2016, 387, 1561–1572. [Google Scholar] [CrossRef]
  13. Baldwin, D.; Birtwistle, J. The Encyclopedia of Visual Medicine Series: An Atlas of Depression, 1st ed.; The Parthenon Publishing Group: London, UK, 2002; ISBN 1850709424. [Google Scholar]
  14. Belmaker, R.H.A.G. Major Depressive Disorder. N. Engl. J. Med. 2008, 358, 55–68. [Google Scholar] [CrossRef] [Green Version]
  15. Judd, L.L.; Akiskal, H.S.; Zeller, P.J.; Paulus, M.; Leon, A.C.; Maser, J.D.; Endicott, J.; Coryell, W.; Kunovac, J.L.; Mueller, T.I.; et al. Psychosocial disability during the long-term course of unipolar major depressive disorder. Arch. Gen. Psychiatry 2000, 57, 375–380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Wells, K.B.; Stewart, A.; Hays, R.D.; Burnam, M.A.; Daniels, M.; Berry, S.; Greenfield, S.; Ware, J. Functioning and Well-being Results from the Medical Outcomes. J. Am. Med. Assoc. 1989, 262, 914–919. [Google Scholar] [CrossRef]
  17. Kaplan, H.I.; Sadock, B.J.; Sadock, V.A. Synopsis of Psychiatry: Behavioral Sciences, Clinical Psychiatry, 10th ed.; Lippincott Williams and Wilkins: Philadelphia, PA, USA, 2007; ISBN 13:978-81-89960-37-7. [Google Scholar]
  18. Tess, A.V.; Smetana, G.W. Medical evaluation of patients undergoing electroconvulsive therapy. N. Engl. J. Med. 2009, 360, 1437–1444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Schoeyen, H.K.; Kessler, U.; Andreassen, O.A.; Auestad, B.H.; Bergsholm, P.; Malt, U.F.; Morken, G.; Oedegaard, K.J.; Vaaler, A. Treatment-resistant bipolar depression: A randomized controlled trial of electroconvulsive therapy versus algorithm-based pharmacological treatment. Am. J. Psychiatry 2015, 172, 41–51. [Google Scholar] [CrossRef]
  20. Chiang, K.-J.; Tsai, J.-C.; Liu, D.; Lin, C.-H.; Chiu, H.-L.; Chou, K.-R. Efficacy of cognitive-behavioral therapy in patients with bipolar disorder: A meta-analysis of randomized controlled trials. PLoS ONE 2017, 12, e0176849. [Google Scholar] [CrossRef] [Green Version]
  21. Mcallister-williams, R.H.; Arango, C.; Blier, P.; Demyttenaere, K.; Falkai, P.; Gorwood, P.; Hopwood, M.; Javed, A.; Kasper, S.; Malhi, G.S.; et al. Reconceptualising treatment-resistant depression as difficult- to-treat depression. Lancet Psychiatry 2021, 8, 14–15. [Google Scholar] [CrossRef]
  22. Lisanby, S.H. Electroconvulsive Therapy for Depression. N. Engl. J. Med. 2007, 1939–1945. [Google Scholar] [CrossRef] [Green Version]
  23. Cipriani, A.; Furukawa, T.A.; Salanti, G.; Chaimani, A.; Atkinson, L.Z.; Ogawa, Y.; Leucht, S.; Ruhe, H.G.; Turner, E.H.; Higgins, J.P.T.; et al. Comparative efficacy and acceptability of 21 antidepressant drugs for the acute treatment of adults with major depressive disorder: A systematic review and network meta-analysis. Lancet 2018, 391, 1357–1366. [Google Scholar] [CrossRef] [Green Version]
  24. Park, L.T.; Zarate, C.A. Clinical Practice Depression in the Primary Care Setting. N. Engl. J. Med. 2019, 380. [Google Scholar] [CrossRef]
  25. Shine, B.; Mcknight, R.F.; Leaver, L.; Geddes, J.R. Long-term effects of lithium on renal, thyroid, and parathyroid function: A retrospective analysis of laboratory data. Lancet 2015, 386, 461–468. [Google Scholar] [CrossRef] [Green Version]
  26. Azab, A.N.; Shnaider, A.; Osher, Y.; Wang, D.; Bersudsky, Y.; Belmaker, R.H. Lithium nephrotoxicity. Int. J. Bipolar Disord. 2015, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Akincigil, A.; Bowblis, J.R.; Levin, C.; Walkup, J.T.; Jan, S.; Crystal, S. Adherence to antidepressant treatment among privately insured patients diagnosed with depression. Med. Care 2007, 45, 363–369. [Google Scholar] [CrossRef] [Green Version]
  28. Montgomery, S.L.; Bowers, W.J. Tumor necrosis factor-alpha and the roles it plays in homeostatic and degenerative processes within the central nervous system. J. Neuroimmune Pharmacol. 2012, 7, 42–59. [Google Scholar] [CrossRef]
  29. Balkwill, F. Balkwill—2009—Tumour necrosis factor and cancer. Nat. Rev. Cancer 2009, 9, 361–371. [Google Scholar] [CrossRef] [PubMed]
  30. Sedger, L.M.; McDermott, M.F. TNF and TNF-receptors: From mediators of cell death and inflammation to therapeutic giants—Past, present and future. Cytokine Growth Factor Rev. 2014, 25, 453–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Kalliolias, G.D.; Ivashkiv, L.B. TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nat. Rev. Rheumatol. 2016, 12, 49–62. [Google Scholar] [CrossRef]
  32. Carswell, E.A.; Old, L.J.; Kassel, R.L.; Green, S.; Fiore, N.; Williamson, B. An endotoxin-induced serum factor that causes necrosis of tumors (activated macrophage). Immunology 1975, 72, 3666–3670. [Google Scholar]
  33. Volterra, A.; Meldolesi, J. Astrocytes, from brain glue to communication elements: The revolution continues. Nat. Rev. Neurosci. 2005, 6, 626–640. [Google Scholar] [CrossRef] [PubMed]
  34. Allen, N.J.; Barres, B.A. Neuroscience: Glia—More than just brain glue. Nature 2009, 457, 675–677. [Google Scholar] [CrossRef] [PubMed]
  35. Tenenbaum, M.; Azab, A.N.; Kaplanski, J. Effects of estrogen against LPS-induced inflammation and toxicity in primary rat glial and neuronal cultures. J. Endotoxin Res. 2007, 13, 158–166. [Google Scholar] [CrossRef] [PubMed]
  36. Blobel, C.P.; Takaishi, H.; Okada, Y.; Toyama, Y.; Horiuchi, K.; Kimura, T.; Miyamoto, T. Endotoxin Shock Myeloid Cells Prevents Lethality from (TACE/ADAM17) Inactivation in Mouse-Converting Enzyme α Cutting Edge: TNF. J. Immunol. Ref. 2007, 179, 2686–2689. [Google Scholar] [CrossRef] [Green Version]
  37. Darshinee, P.; Issuree, A.; Maretzky, T.; Mcilwain, D.R.; Monette, S.; Qing, X.; Lang, P.A.; Swendeman, S.L.; Park-Min, K.-H.; Binder, N.; et al. Brief report iRHOM2 is a critical pathogenic mediator of inflammatory arthritis. J. Clin. Investig. 2013, 123. [Google Scholar] [CrossRef]
  38. Locksley, R.M.; Killeen, N.; Lenardo, M.J. The TNF and TNF receptor superfamilies: Integrating mammalian biology. Cell 2001, 104, 487–501. [Google Scholar] [CrossRef] [Green Version]
  39. Probert, L. TNF and its receptors in the CNS: The essential, the desirable and the deleterious effects. Neuroscience 2015, 302, 2–22. [Google Scholar] [CrossRef] [Green Version]
  40. Floden, A.M.; Li, S.; Combs, C.K. β-Amyloid-stimulated microglia induce neuron death via synergistic stimulation of tumor necrosis factor α and NMDA receptors. J. Neurosci. 2005, 25, 2566–2575. [Google Scholar] [CrossRef]
  41. Zelová, H.; Hošek, J. TNF-α signalling and inflammation: Interactions between old acquaintances. Inflamm. Res. 2013, 62, 641–651. [Google Scholar] [CrossRef]
  42. Habbas, S.; Santello, M.; Becker, D.; Stubbe, H.; Zappia, G.; Liaudet, N.; Klaus, F.R.; Kollias, G.; Fontana, A.; Pryce, C.R.; et al. Neuroinflammatory TNFα Impairs Memory via Astrocyte Signaling. Cell 2015, 163, 1730–1741. [Google Scholar] [CrossRef] [Green Version]
  43. Jarskog, L.F.; Xiao, H.; Wilkie, M.B.; Lauder, J.M.; Gilmore, J.H. Cytokine regulation of embryonic rat dopamine and serotonin neuronal survival in vitro. Int. J. Dev. Neurosci. 1997, 15, 711–716. [Google Scholar] [CrossRef]
  44. Zhu, C.B.; Blakely, R.D.; Hewlett, W.A. The proinflammatory cytokines interleukin-1beta and tumor necrosis factor-alpha activate serotonin transporters. Neuropsychopharmacology 2006, 31, 2121–2131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Malynn, S.; Campos-Torres, A.; Moynagh, P.; Haase, J. The pro-inflammatory cytokine TNF-α regulates the activity and expression of the serotonin transporter (SERT) in astrocytes. Neurochem. Res. 2013, 38, 694–704. [Google Scholar] [CrossRef]
  46. Liu, H.; Luiten, P.G.M.; Eisel, U.L.M.; Dejongste, M.J.L.; Schoemaker, R.G. Depression after myocardial infarction: TNF-α-induced alterations of the blood-brain barrier and its putative therapeutic implications. Neurosci. Biobehav. Rev. 2013, 37, 561–572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Menard, C.; Pfau, M.L.; Hodes, G.E.; Kana, V.; Wang, V.X.; Bouchard, S.; Takahashi, A.; Flanigan, M.E.; Aleyasin, H.; Leclair, K.B.; et al. Social stress induces neurovascular pathology promoting depression. Nat. Neurosci. 2017, 20, 1752–1760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Cheng, Y.; Desse, S.; Martinez, A.; Worthen, R.J.; Jope, R.S.; Beurel, E. TNFα disrupts blood brain barrier integrity to maintain prolonged depressive-like behavior in mice. Brain Behav. Immun. 2018, 69, 556–567. [Google Scholar] [CrossRef]
  49. Wohleb, E.S.; Hanke, M.L.; Corona, A.W.; Powell, N.D.; Stiner, L.M.; Bailey, M.T.; Nelson, R.J.; Godbout, J.P.; Sheridan, J.F. β-Adrenergic receptor antagonism prevents anxiety-like behavior and microglial reactivity induced by repeated social defeat. J. Neurosci. 2011, 31, 6277–6288. [Google Scholar] [CrossRef] [Green Version]
  50. Wohleb, E.S.; Powell, N.D.; Godbout, J.P.; Sheridan, J.F. Stress-induced recruitment of bone marrow-derived monocytes to the brain promotes anxiety-like behavior. J. Neurosci. 2013, 33, 13820–13833. [Google Scholar] [CrossRef] [Green Version]
  51. Barreto, G.E.; Gonzalez, J.; Torres, Y.; Morales, L. Astrocytic-neuronal crosstalk: Implications for neuroprotection from brain injury. Neurosci. Res. 2011, 71, 107–113. [Google Scholar] [CrossRef]
  52. Pfau, M.L.; Ménard, C.; Russo, S.J. Inflammatory Mediators in Mood Disorders: Therapeutic Opportunities. Annu. Rev. Pharmacol. Toxicol. 2018, 58, 411–428. [Google Scholar] [CrossRef] [PubMed]
  53. Block, M.L.; Hong, J.S. Microglia and inflammation-mediated neurodegeneration: Multiple triggers with a common mechanism. Prog. Neurobiol. 2005, 76, 77–98. [Google Scholar] [CrossRef] [PubMed]
  54. Cunningham, C. Microglia and neurodegeneration: The role of systemic inflammation. Glia 2013, 61, 71–90. [Google Scholar] [CrossRef] [PubMed]
  55. Smith, R.S. The macrophage theory of depression. Med. Hypotheses 1991, 35, 298–306. [Google Scholar] [CrossRef]
  56. Maes, M.; Bosmans, E.; Suy, E.; Vandervorst, C.; De Jonckheere, C.; Raus, J. Immune disturbances during major depression: Upregulated expression of interleukin-2 receptors. Neuropsychobiology 1990, 24, 115–120. [Google Scholar] [CrossRef]
  57. Licinio, J.; Wong, M.L. The role of inflammatory mediators in the biology of major depression: Central nervous system cytokines modulate the biological substrate of depressive symptoms, regulate stress-responsive systems, and contribute to neurotoxicity and neuroprotection. Mol. Psychiatry 1999, 4, 317–327. [Google Scholar] [CrossRef] [Green Version]
  58. Müller, N.; Schwarz, M.J.; Dehning, S.; Douhe, A.; Cerovecki, A.; Goldstein-Müller, B.; Spellmann, I.; Hetzel, G.; Maino, K.; Kleindienst, N.; et al. The cyclooxygenase-2 inhibitor celecoxib has therapeutic effects in major depression: Results of a double-blind, randomized, placebo controlled, add-on pilot study to reboxetine. Mol. Psychiatry 2006, 11, 680–684. [Google Scholar] [CrossRef]
  59. Elhaik, E.; Zandi, P. Dysregulation of the NF-κB pathway as a potential inducer of bipolar disorder. J. Psychiatr. Res. 2015, 70, 18–27. [Google Scholar] [CrossRef]
  60. Liu, Y.; Ho, R.C.M.; Mak, A. Interleukin (IL)-6, tumour necrosis factor alpha (TNF-α) and soluble interleukin-2 receptors (sIL-2R) are elevated in patients with major depressive disorder: A meta-analysis and meta-regression. J. Affect. Disord. 2012, 139, 230–239. [Google Scholar] [CrossRef] [PubMed]
  61. Anderson, G. Editorial (Thematic Issue: The Kynurenine and Melatonergic Pathways in Psychiatric and CNS Disorders). Curr. Pharm. Des. 2016, 22, 947–948. [Google Scholar] [CrossRef] [PubMed]
  62. Kopschina Feltes, P.; Doorduin, J.; Klein, H.C.; Juárez-Orozco, L.E.; Dierckx, R.A.J.O.; Moriguchi-Jeckel, C.M.; De Vries, E.F.J. Anti-inflammatory treatment for major depressive disorder: Implications for patients with an elevated immune profile and non-responders to standard antidepressant therapy. J. Psychopharmacol. 2017, 31, 1149–1165. [Google Scholar] [CrossRef] [Green Version]
  63. Dimopoulos, N.; Piperi, C.; Psarra, V.; Lea, R.W.; Kalofoutis, A. Increased plasma levels of 8-iso-PGF2α and IL-6 in an elderly population with depression. Psychiatry Res. 2008, 161, 59–66. [Google Scholar] [CrossRef]
  64. Piletz, J.E.; Halaris, A.; Iqbal, O.; Hoppensteadt, D.; Fareed, J.; Zhu, H.; Sinacore, J.; Devane, C.L. Pro-inflammatory biomakers in depression: Treatment with venlafaxine. World J. Biol. Psychiatry 2009, 10, 313–323. [Google Scholar] [CrossRef] [PubMed]
  65. Zeugmann, S.; Quante, A.; Heuser, I.; Schwarzer, R.; Anghelescu, I. Inflammatory biomarkers in 70 depressed inpatients with and without the metabolic syndrome. J. Clin. Psychiatry 2010, 71, 1007–1016. [Google Scholar] [CrossRef] [PubMed]
  66. Raison, C.L.; Dantzer, R.; Kelley, K.W.; Lawson, M.A.; Woolwine, B.J.; Vogt, G.; Spivey, J.R.; Saito, K.; Miller, A.H. CSF concentrations of brain tryptophan and kynurenines during immune stimulation with IFN-α: Relationship to CNS immune responses and depression. Mol. Psychiatry 2010, 15, 393–403. [Google Scholar] [CrossRef] [Green Version]
  67. Hannestad, J.; Dellagioia, N.; Bloch, M. The effect of antidepressant medication treatment on serum levels of inflammatory cytokines: A meta-analysis. Neuropsychopharmacology 2011, 36, 2452–2459. [Google Scholar] [CrossRef] [PubMed]
  68. Shelton, R.C.; Claiborne, J.; Sidoryk-Wegrzynowicz, M.; Reddy, R.; Aschner, M.; Lewis, D.A.; Mirnics, K. Altered expression of genes involved in inflammation and apoptosis in frontal cortex in major depression. Mol. Psychiatry 2011, 16, 751–762. [Google Scholar] [CrossRef] [PubMed]
  69. Köhler, C.A.; Freitas, T.H.; Maes, M.; de Andrade, N.Q.; Liu, C.S.; Fernandes, B.S.; Stubbs, B.; Solmi, M.; Veronese, N.; Herrmann, N.; et al. Peripheral cytokine and chemokine alterations in depression: A meta-analysis of 82 studies. Acta Psychiatr. Scand. 2017, 135, 373–387. [Google Scholar] [CrossRef]
  70. Kappelmann, N.; Lewis, G.; Dantzer, R.; Jones, P.B.; Khandaker, G.M. Antidepressant activity of anti-cytokine treatment: A systematic review and meta-analysis of clinical trials of chronic inflammatory conditions. Mol. Psychiatry 2018, 23, 335–343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Liu, H.C.; Yang, Y.Y.; Chou, Y.M.; Chen, K.P.; Shen, W.W.; Leu, S.J. Immunologic variables in acute mania of bipolar disorder. J. Neuroimmunol. 2004, 150, 116–122. [Google Scholar] [CrossRef]
  72. Goldstein, B.I.; Kemp, D.E.; Soczynska, J.K.; McIntyre, R.S. Inflammation and the phenomenology, pathophysiology, comorbidity, and treatment of bipolar disorder: A systematic review of the literature. J. Clin. Psychiatry 2009, 70, 1078–1090. [Google Scholar] [CrossRef] [PubMed]
  73. Dhabhar, F.S.; Burke, H.M.; Epel, E.S.; Mellon, S.H.; Rosser, R.; Reus, V.I.; Wolkowitz, O.M. Low serum IL-10 concentrations and loss of regulatory association between IL-6 and IL-10 in adults with major depression. J. Psychiatr. Res. 2009, 43, 962–969. [Google Scholar] [CrossRef] [PubMed]
  74. Kim, H.W.; Rapoport, S.I.; Rao, J.S. Altered arachidonic acid cascade enzymes in postmortem brain from bipolar disorder patients. Mol. Psychiatry 2011, 16, 419–428. [Google Scholar] [CrossRef] [Green Version]
  75. Söderlund, J.; Olsson, S.K.; Samuelsson, M.; Walther-Jallow, L.; Johansson, C.; Erhardt, S.; Landén, M.; Engberg, G. Elevation of cerebrospinal fluid interleukin-1β in bipolar disorder. J. Psychiatry Neurosci. 2011, 36, 114–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Savas, H.A.; Gergerlioglu, H.S.; Armutcu, F.; Herken, H.; Yilmaz, H.R.; Kocoglu, E.; Selek, S.; Tutkun, H.; Zoroglu, S.S.; Akyol, O. Elevated serum nitric oxide and superoxide dismutase in euthymic bipolar patients: Impact of past episodes. World J. Biol. Psychiatry 2006, 7, 51–55. [Google Scholar] [CrossRef]
  77. Haack, M.; Hinze-Selch, D.; Fenzel, T.; Kraus, T.; Kühn, M.; Schuld, A.; Pollmächer, T. Plasma levels of cytokines and soluble cytokine receptors in psychiatric patients upon hospital admission: Effects of confounding factors and diagnosis. J. Psychiatr. Res. 1999, 33, 407–418. [Google Scholar] [CrossRef]
  78. O’Brien, S.M.; Scully, P.; Scott, L.V.; Dinan, T.G. Cytokine profiles in bipolar affective disorder: Focus on acutely ill patients. J. Affect. Disord. 2006, 90, 263–267. [Google Scholar] [CrossRef] [PubMed]
  79. Rao, J.S.; Harry, G.J.; Rapoport, S.I.; Kim, H.W. Increased excitotoxicity and neuroinflammatory markers in postmortem frontal cortex from bipolar disorder patients. Mol. Psychiatry 2010, 15, 384–392. [Google Scholar] [CrossRef]
  80. Modabbernia, A.; Taslimi, S.; Brietzke, E.; Ashrafi, M. Cytokine alterations in bipolar disorder: A meta-analysis of 30 studies. Biol. Psychiatry 2013, 74, 15–25. [Google Scholar] [CrossRef] [PubMed]
  81. Dickerson, F.; Stallings, C.; Origoni, A.; Boronow, J.; Yolken, R. Elevated serum levels of C-reactive protein are associated with mania symptoms in outpatients with bipolar disorder. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2007, 31, 952–955. [Google Scholar] [CrossRef] [PubMed]
  82. Kauer-Sant’Anna, M.; Kapczinski, F.; Andreazza, A.C.; Bond, D.J.; Lam, R.W.; Young, L.T.; Yatham, L.N. Brain-derived neurotrophic factor and inflammatory markers in patients with early- vs. late-stage bipolar disorder. Int. J. Neuropsychopharmacol. 2009, 12, 447–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Kapczinski, F.; Dal-Pizzol, F.; Teixeira, A.L.; Magalhaes, P.V.S.; Kauer-Sant’Anna, M.; Klamt, F.; Moreira, J.C.F.; Augusto de Bittencourt Pasquali, M.; Fries, G.R.; Quevedo, J.; et al. Peripheral biomarkers and illness activity in bipolar disorder. J. Psychiatr. Res. 2011, 45, 156–161. [Google Scholar] [CrossRef]
  84. Hope, S.; Dieset, I.; Agartz, I.; Steen, N.E.; Ueland, T.; Melle, I.; Aukrust, P.; Andreassen, O.A. Affective symptoms are associated with markers of inflammation and immune activation in bipolar disorders but not in schizophrenia. J. Psychiatr. Res. 2011, 45, 1608–1616. [Google Scholar] [CrossRef]
  85. Munkholm, K.; Braüner, J.V.; Kessing, L.V.; Vinberg, M. Cytokines in bipolar disorder vs. healthy control subjects: A systematic review and meta-analysis. J. Psychiatr. Res. 2013, 47, 1119–1133. [Google Scholar] [CrossRef] [PubMed]
  86. Choi, K.W.; Jang, E.H.; Kim, A.Y.; Kim, H.; Park, M.J.; Byun, S.; Fava, M.; Mischoulon, D.; Papakostas, G.I.; Yu, H.Y.; et al. Predictive inflammatory biomarkers for change in suicidal ideation in major depressive disorder and panic disorder: A 12-week follow-up study. J. Psychiatr. Res. 2021, 133, 73–81. [Google Scholar] [CrossRef] [PubMed]
  87. Millett, C.E.; Harder, J.; Locascio, J.J.; Shanahan, M.; Santone, G.; Fichorova, R.N.; Corrigan, A.; Baecher-Allan, C.; Burdick, K.E. TNF-α and its soluble receptors mediate the relationship between prior severe mood episodes and cognitive dysfunction in euthymic bipolar disorder. Brain Behav. Immun. 2020, 88, 403–410. [Google Scholar] [CrossRef] [PubMed]
  88. Diniz, B.S.; Teixeira, A.L.; Talib, L.L.; Mendonça, V.A.; Gattaz, W.F.; Forlenza, O.V. Increased soluble TNF receptor 2 in antidepressant-free patients with late-life depression. J. Psychiatr. Res. 2010, 44, 917–920. [Google Scholar] [CrossRef] [PubMed]
  89. Effects of Psychotropic Drugs on Nuclear Factor Kappa B. Available online: https://www.europeanreview.org/article/8738 (accessed on 23 November 2020).
  90. Bortolotto, V.; Cuccurazzu, B.; Canonico, P.L.; Grilli, M. NF-κB mediated regulation of adult hippocampal neurogenesis: Relevance to mood disorders and antidepressant activity. Biomed Res. Int. 2014, 2014, 612798. [Google Scholar] [CrossRef]
  91. Ichiyama, T.; Okada, K.; Lipton, J.M.; Matsubara, T.; Hayashi, T.; Furukawa, S. Sodium valproate inhibits production of TNF-α and IL-6 and activation of NF-κB. Brain Res. 2000, 857, 246–251. [Google Scholar] [CrossRef]
  92. Rao, J.S.; Bazinet, R.P.; Rapoport, S.I.; Lee, H.J. Chronic treatment of rats with sodium valproate downregulates frontal cortex NF-κB DNA binding activity and COX-2 mRNA 1. Bipolar Disord. 2007, 9, 513–520. [Google Scholar] [CrossRef] [PubMed]
  93. Jaehne, E.J.; Corrigan, F.; Toben, C.; Jawahar, M.C.; Baune, B.T. The effect of the antipsychotic drug quetiapine and its metabolite norquetiapine on acute inflammation, memory and anhedonia. Pharmacol. Biochem. Behav. 2015, 135, 136–144. [Google Scholar] [CrossRef] [PubMed]
  94. Faour-Nmarne, C.; Azab, A.N. Effects of olanzapine on LPS-induced inflammation in rat primary glia cells. Innate Immun. 2016, 22, 40–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Chang, M.C.J.; Contreras, M.A.; Rosenberger, T.A.; Rintala, J.J.O.; Bell, J.M.; Rapoport, S.I. Chronic valproate treatment decreases the in vivo turnover of arachidonic acid in brain phospholipids: A possible common effect of mood stabilizers. J. Neurochem. 2001, 77, 796–803. [Google Scholar] [CrossRef]
  96. Bazinet, R.P.; Rao, J.S.; Chang, L.; Rapoport, S.I.; Lee, H.J. Chronic carbamazepine decreases the incorporation rate and turnover of arachidonic acid but not docosahexaenoic acid in brain phospholipids of the unanesthetized rat: Relevance to bipolar disorder. Biol. Psychiatry 2006, 59, 401–407. [Google Scholar] [CrossRef]
  97. Lee, H.J.; Ertley, R.N.; Rapoport, S.I.; Bazinet, R.P.; Rao, J.S. Chronic administration of lamotrigine downregulates COX-2 mRNA and protein in rat frontal cortex. Neurochem. Res. 2008, 33, 861–866. [Google Scholar] [CrossRef]
  98. Li, H.; Hong, W.; Zhang, C.; Wu, Z.; Wang, Z.; Yuan, C.; Li, Z.; Huang, J.; Lin, Z.; Fang, Y. IL-23 and TGF-β1 levels as potential predictive biomarkers in treatment of bipolar i disorder with acute manic episode. J. Affect. Disord. 2015, 174, 361–366. [Google Scholar] [CrossRef]
  99. Castanon, N.; Leonard, B.E.; Neveu, P.J.; Yirmiya, R. Effects of antidepressants on cytokine production and actions. Brain Behav. Immun. 2002, 16, 569–574. [Google Scholar] [CrossRef]
  100. Nassar, A.; Sharon-Granit, Y.; Azab, A.N. Psychotropic drugs attenuate lipopolysaccharide-induced hypothermia by altering hypothalamic levels of inflammatory mediators in rats. Neurosci. Lett. 2016, 626, 59–67. [Google Scholar] [CrossRef]
  101. Rao, J.S.; Lee, H.J.; Rapoport, S.I.; Bazinet, R.P. Mode of action of mood stabilizers: Is the arachidonic acid cascade a common target? Mol. Psychiatry 2008, 13, 585–596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Sugino, H.; Futamura, T.; Mitsumoto, Y.; Maeda, K.; Marunaka, Y. Atypical antipsychotics suppress production of proinflammatory cytokines and up-regulate interleukin-10 in lipopolysaccharide-treated mice. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2009, 33, 303–307. [Google Scholar] [CrossRef] [PubMed]
  103. Nahman, S.; Belmaker, R.H.; Azab, A.N. Effects of lithium on lipopolysaccharide-induced inflammation in rat primary glia cells. Innate Immun. 2012, 18, 447–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Tourjman, V.; Kouassi, É.; Koué, M.È.; Rocchetti, M.; Fortin-Fournier, S.; Fusar-Poli, P.; Potvin, S. Antipsychotics’ effects on blood levels of cytokines in schizophrenia: A meta-analysis. Schizophr. Res. 2013, 151, 43–47. [Google Scholar] [CrossRef] [PubMed]
  105. Liu, Y.; Guo, W.; Zhang, Y.; Lv, L.; Hu, F.; Wu, R.; Zhao, J. Decreased Resting-State Interhemispheric Functional Connectivity Correlated with Neurocognitive Deficits in Drug-Naive First-Episode Adolescent-Onset Schizophrenia. Int. J. Neuropsychopharmacol. 2018, 21, 33–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Noto, C.; Ota, V.K.; Gouvea, E.S.; Rizzo, L.B.; Spindola, L.M.N.; Honda, P.H.S.; Cordeiro, Q.; Belangero, S.I.; Bressan, R.A.; Gadelha, A.; et al. Effects of risperidone on cytokine profile in drug-naïve first-episode psychosis. Int. J. Neuropsychopharmacol. 2014, 18, pyu042. [Google Scholar] [CrossRef] [PubMed]
  107. Nassar, A.; Azab, A.N. Effects of lithium on inflammation. ACS Chem. Neurosci. 2014, 5, 451–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Leu, S.J.; Yang, Y.Y.; Liu, H.C.; Cheng, C.Y.; Wu, Y.C.; Huang, M.C.; Lee, Y.L.; Chen, C.C.; Shen, W.W.; Liu, K.J. Valproic Acid and Lithium Meditate Anti-Inflammatory Effects by Differentially Modulating Dendritic Cell Differentiation and Function. J. Cell. Physiol. 2017, 232, 1176–1186. [Google Scholar] [CrossRef] [PubMed]
  109. Hwang, J.; Zheng, L.T.; Ock, J.; Lee, M.G.; Kim, S.H.; Lee, H.W.; Lee, W.H.; Park, H.C.; Suk, K. Inhibition of glial inflammatory activation and neurotoxicity by tricyclic antidepressants. Neuropharmacology 2008, 55, 826–834. [Google Scholar] [CrossRef]
  110. Lu, Y.; Ho, C.S.; Liu, X.; Chua, A.N.; Wang, W.; McIntyre, R.S.; Ho, R.C. Chronic administration of fluoxetine and pro-inflammatory cytokine change in a rat model of depression. PLoS ONE 2017, 12, e0186700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Sluzewska, A.; Rybakowski, J.K.; Laciak, M.; Mackiewicz, A.; Sobieska, M.; Wiktorowicz, K. Interleukin-6 Serum Levels in Depressed Patients before and after Treatment with Fluoxetine. Ann. N. Y. Acad. Sci. 1995, 762, 474–476. [Google Scholar] [CrossRef] [PubMed]
  112. Rao, R.; Hao, C.M.; Breyer, M.D. Hypertonic Stress Activates Glycogen Synthase Kinase 3β-mediated Apoptosis of Renal Medullary Interstitial Cells, Suppressing an NFκB-driven Cyclooxygenase-2-dependent Survival Pathway. J. Biol. Chem. 2004, 279, 3949–3955. [Google Scholar] [CrossRef] [Green Version]
  113. Meyer, J.M.; McEvoy, J.P.; Davis, V.G.; Goff, D.C.; Nasrallah, H.A.; Davis, S.M.; Hsiao, J.K.; Swartz, M.S.; Stroup, T.S.; Lieberman, J.A. Inflammatory Markers in Schizophrenia: Comparing Antipsychotic Effects in Phase 1 of the Clinical Antipsychotic Trials of Intervention Effectiveness Study. Biol. Psychiatry 2009, 66, 1013–1022. [Google Scholar] [CrossRef] [Green Version]
  114. Sárvári, A.K.; Veréb, Z.; Uray, I.P.; Fésüs, L.; Balajthy, Z. Atypical antipsychotics induce both proinflammatory and adipogenic gene expression in human adipocytes in vitro. Biochem. Biophys. Res. Commun. 2014, 450, 1383–1389. [Google Scholar] [CrossRef]
  115. Isgren, A.; Jakobsson, J.; Pålsson, E.; Ekman, C.J.; Johansson, A.G.M.; Sellgren, C.; Blennow, K.; Zetterberg, H.; Landén, M. Increased cerebrospinal fluid interleukin-8 in bipolar disorder patients associated with lithium and antipsychotic treatment. Brain Behav. Immun. 2015, 43, 198–204. [Google Scholar] [CrossRef]
  116. Martín-Hernández, D.; Caso, J.R.; Javier Meana, J.; Callado, L.F.; Madrigal, J.L.M.; García-Bueno, B.; Leza, J.C. Intracellular inflammatory and antioxidant pathways in postmortem frontal cortex of subjects with major depression: Effect of antidepressants. J. Neuroinflamm. 2018, 15. [Google Scholar] [CrossRef] [PubMed]
  117. Arana, G.W.; Forbes, R.A. Dexamethasone for the treatment of depression: A preliminary report. J. Clin. Psychiatry 1991, 52, 304–306. [Google Scholar] [PubMed]
  118. Wolkowitz, O.M.; Reus, V.I.; Keebler, A.; Nelson, N.; Friedland, M.; Brizendine, L.; Roberts, E. Double-blind treatment of major depression with dehydroepiandrosterone. Am. J. Psychiatry 1999, 156, 646–649. [Google Scholar] [CrossRef] [PubMed]
  119. DeBattista, C.; Posener, J.A.; Kalehzan, B.M.; Schatzberg, A.F. Acute antidepressant effects of intravenous hydrocortisone and CRH in depressed patients: A double-blind, placebo-controlled study. Am. J. Psychiatry 2000, 157, 1334–1337. [Google Scholar] [CrossRef] [PubMed]
  120. Nery, F.G.; Monkul, E.S.; Hatch, J.P.; Fonseca, M.; Zunta-Soares, G.B.; Frey, B.N.; Bowden, C.L.; Soares, J.C. Celecoxib as an adjunct in the treatment of depressive or mixed episodes of bipolar disorder: A double-blind, randomized, placebo-controlled study. Hum. Psychopharmacol. 2008, 23, 87–94. [Google Scholar] [CrossRef] [PubMed]
  121. Abbasi, S.H.; Hosseini, F.; Modabbernia, A.; Ashrafi, M.; Akhondzadeh, S. Effect of celecoxib add-on treatment on symptoms and serum IL-6 concentrations in patients with major depressive disorder: Randomized double-blind placebo-controlled study. J. Affect. Disord. 2012, 141, 308–314. [Google Scholar] [CrossRef]
  122. Arabzadeh, S.; Ameli, N.; Zeinoddini, A.; Rezaei, F.; Farokhnia, M.; Mohammadinejad, P.; Ghaleiha, A.; Akhondzadeh, S. Celecoxib adjunctive therapy for acute bipolar mania: A randomized, double-blind, placebo-controlled trial. Bipolar Disord. 2015, 17, 606–614. [Google Scholar] [CrossRef] [PubMed]
  123. Husain, M.I.; Chaudhry, I.B.; Husain, N.; Khoso, A.B.; Rahman, R.R.; Hamirani, M.M.; Hodsoll, J.; Qurashi, I.; Deakin, J.F.W.; Young, A.H. Minocycline as an adjunct for treatment-resistant depressive symptoms: A pilot randomised placebo-controlled trial. J. Psychopharmacol. 2017, 31, 1166–1175. [Google Scholar] [CrossRef]
  124. Fields, C.; Drye, L.; Vaidya, V.; Lyketsos, C. Celecoxib or naproxen treatment does not benefit depressive symptoms in persons age 70 and older: Findings from a randomized controlled trial. Am. J. Geriatr. Psychiatry 2012, 20, 505–513. [Google Scholar] [CrossRef] [Green Version]
  125. Husain, M.I.; Chaudhry, I.B.; Khoso, A.B.; Husain, M.O.; Hodsoll, J.; Ansari, M.A.; Naqvi, H.A.; Minhas, F.A.; Carvalho, A.F.; Meyer, J.H.; et al. Minocycline and celecoxib as adjunctive treatments for bipolar depression: A multicentre, factorial design randomised controlled trial. Lancet Psychiatry 2020, 7, 515–527. [Google Scholar] [CrossRef]
  126. Réus, G.Z.; Fries, G.R.; Stertz, L.; Badawy, M.; Passos, I.C.; Barichello, T.; Kapczinski, F.; Quevedo, J. The role of inflammation and microglial activation in the pathophysiology of psychiatric disorders. Neuroscience 2015, 300, 141–154. [Google Scholar] [CrossRef] [PubMed]
  127. Present Daniel, H.; Rutgeerts, P.; Targan, S.; Hanauer, S.B.; Mayer, L.; van Hogezand, R.A.; Podolsky, D.K.; Sands, B.E.; Braakman, T.; DeWoody, K.L.; et al. Infliximab for the treatment of fistulas in patients with Crohn’s disease. N. Engl. J. Med. 1999, 340, 1398–1404. [Google Scholar] [CrossRef] [Green Version]
  128. Menter, A.; Augustin, M.; Signorovitch, J.; Yu, A.P.; Wu, E.Q.; Gupta, S.R.; Bao, Y.; Mulani, P. The effect of adalimumab on reducing depression symptoms in patients with moderate to severe psoriasis: A randomized clinical trial. J. Am. Acad. Dermatol. 2010, 62, 812–818. [Google Scholar] [CrossRef] [PubMed]
  129. Leman, J.; Walton, S.; Layton, A.M.; Ward, K.A.; McBride, S.; Cliff, S.; Downs, A.; Landeira, M.; Bewley, A. The real world impact of adalimumab on quality of life and the physical and psychological effects of moderate-to-severe psoriasis: A UK prospective, multicenter, observational study. J. Dermatolog. Treat. 2020, 31, 213–221. [Google Scholar] [CrossRef] [PubMed]
  130. Scheinfeld, N.; Sundaram, M.; Teixeira, H.; Gu, Y.; Okun, M. Reduction in pain scores and improvement in depressive symptoms in patients with hidradenitis suppurativa treated with adalimumab in a phase 2, randomized, placebo-controlled trial. Dermatol. Online J. 2016, 22, 2. [Google Scholar]
  131. Roberti, R.; Iannone, L.F.; Palleria, C.; De Sarro, C.; Spagnuolo, R.; Barbieri, M.A.; Vero, A.; Manti, A.; Pisana, V.; Fries, W.; et al. Safety profiles of biologic agents for inflammatory bowel diseases: A prospective pharmacovigilance study in Southern Italy. Curr. Med. Res. Opin. 2020, 36, 1457–1463. [Google Scholar] [CrossRef]
  132. Iannone, L.F.; Bennardo, L.; Palleria, C.; Roberti, R.; de Sarro, C.; Naturale, M.D.; Dastoli, S.; Donato, L.; Manti, A.; Valenti, G.; et al. Safety profile of biologic drugs for psoriasis in clinical practice: An Italian prospective pharmacovigilance study. PLoS ONE 2020, 15, e0241575. [Google Scholar] [CrossRef]
  133. Tracey, D.; Klareskog, L.; Sasso, E.H.; Salfeld, J.G.; Tak, P.P. Tumor necrosis factor antagonist mechanisms of action: A comprehensive review. Pharmacol. Ther. 2008, 117, 244–279. [Google Scholar] [CrossRef]
  134. Persoons, P.; Vermeire, S.; Demyttenaere, K.; Fischler, B.; Vandenberghe, J.; Van Oudenhove, L.; Pierik, M.; Hlavaty, T.; Van Assche, G.; Noman, M.; et al. The impact of major depressive disorder on the short- and long-term outcome of Crohn’s disease treatment with infliximab. Aliment. Pharmacol. Ther. 2005, 22, 101–110. [Google Scholar] [CrossRef] [Green Version]
  135. Minderhoud, I.M.; Samsom, M.; Oldenburg, B. Crohn’s disease, fatigue, and infliximab: Is there a role for cytokines in the pathogenesis of fatigue? World J. Gastroenterol. 2007, 13, 2089–2093. [Google Scholar] [CrossRef] [PubMed]
  136. Ersözlü-Bozkirli, E.D.; Keşkek, Ş.Ö.; Bozkirli, E.; Yücel, A.E. The effect of infliximab on depressive symptoms in patients with ankylosing spondylitis. Acta Reumatol. Port. 2015, 2015, 262–267. [Google Scholar]
  137. Tyring, S.; Gottlieb, A.; Papp, K.; Gordon, K.; Leonardi, C.; Wang, A.; Lalla, D.; Woolley, M.; Jahreis, A.; Zitnik, R.; et al. Etanercept and clinical outcomes, fatigue, and depression in psoriasis: Double-blind placebo-controlled randomised phase III trial. Lancet 2006, 367, 29–35. [Google Scholar] [CrossRef]
  138. Krishnan, R.; Cella, D.; Leonardi, C.; Papp, K.; Gottlieb, A.B.; Dunn, M.; Chiou, C.F.; Patel, V.; Jahreis, A. Effects of etanercept therapy on fatigue and symptoms of depression in subjects treated for moderate to severe plaque psoriasis for up to 96 weeks. Br. J. Dermatol. 2007, 157, 1275–1277. [Google Scholar] [CrossRef] [PubMed]
  139. Kekow, J.; Moots, R.J.; Emery, P.; Durez, P.; Koenig, A.; Singh, A.; Pedersen, R.; Robertson, D.; Freundlich, B.; Sato, R.; et al. Patient-reported outcomes improve with etanercept plus methotrexate in active early rheumatoid arthritis and the improvement is strongly associated with remission: The COMET trial. Ann. Rheum. Dis. 2010, 69, 222–225. [Google Scholar] [CrossRef] [PubMed]
  140. Loftus, E.V.; Feagan, B.G.; Colombel, J.F.; Rubin, D.T.; Wu, E.Q.; Yu, A.P.; Pollack, P.F.; Chao, J.; Mulani, P. Effects of adalimumab maintenance therapy on health-related quality of life of patients with Crohn’s disease: Patient-reported outcomes of the CHARM trial. Am. J. Gastroenterol. 2008, 103, 3132–3141. [Google Scholar] [CrossRef]
  141. Exley, A.R.; Cohen, J.; Buurman, W.; Owen, R.; Lumley, J.; Hanson, G.; Aulakh, J.M.; Bodmer, M.; Stephens, S.; Riddell, A.; et al. Monoclonal antibody to TN F in severe septic shock. Lancet 1990, 335, 1275–1277. [Google Scholar] [CrossRef]
  142. Scallon, B.; Cai, A.; Solowski, N.; Rosenberg, A.; Song, X.Y.; Shealy, D.; Wagner, C. Binding and functional comparisons of two types of tumor necrosis factor antagonists. J. Pharmacol. Exp. Ther. 2002, 301, 418–426. [Google Scholar] [CrossRef] [Green Version]
  143. Furst, D.E.; Wallis, R.; Broder, M.; Beenhouwer, D.O. Tumor Necrosis Factor Antagonists: Different Kinetics and/or Mechanisms of Action May Explain Differences in the Risk for Developing Granulomatous Infection. Semin. Arthritis Rheum. 2006, 36, 159–167. [Google Scholar] [CrossRef]
  144. Mitoma, H.; Horiuchi, T.; Tsukamoto, H.; Ueda, N. Molecular mechanisms of action of anti-TNF-α agents—Comparison among therapeutic TNF-α antagonists. Cytokine 2018, 101, 56–63. [Google Scholar] [CrossRef]
  145. Ertenli, I.; Ozer, S.; Kiraz, S.; Apras, S.B.; Akdogan, A.; Karadag, O.; Calguneri, M.; Kalyoncu, U. Infliximab, a TNF-alpha antagonist treatment in patients with ankylosing spondylitis: The impact on depression, anxiety and quality of life level. Rheumatol. Int. 2012, 32, 323–330. [Google Scholar] [CrossRef]
  146. Webers, C.; Stolwijk, C.; Schiepers, O.; Schoonbrood, T.; Van Tubergen, A.; Landewé, R.; Van Der Heijde, D.; Boonen, A. Infliximab treatment reduces depressive symptoms in patients with ankylosing spondylitis: An ancillary study to a randomized controlled trial (ASSERT). Arthritis Res. Ther. 2020, 22. [Google Scholar] [CrossRef]
  147. Karson, A.; Demirtaş, T.; Bayramgürler, D.; Balci, F.; Utkan, T. Chronic administration of infliximab (TNF-α inhibitor) decreases depression and anxiety-like behaviour in rat model of chronic mild stress. Basic Clin. Pharmacol. Toxicol. 2013, 112, 335–340. [Google Scholar] [CrossRef]
  148. Fu, X.Y.; Li, H.Y.; Jiang, Q.S.; Cui, T.; Jiang, X.H.; Zhou, Q.X.; Qiu, H.M. Infliximab ameliorating depression-like behavior through inhibiting the activation of the IDO-HAAO pathway mediated by tumor necrosis factor-α in a rat model. Neuroreport 2016, 27, 953–959. [Google Scholar] [CrossRef]
  149. Raison, C.L.; Rutherford, R.E.; Woolwine, B.J.; Shuo, C.; Schettler, P.; Drake, D.F.; Haroon, E.; Miller, A.H. A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: The role of baseline inflammatory biomarkers. Arch. Gen. Psychiatry 2013, 70, 31–41. [Google Scholar] [CrossRef] [PubMed]
  150. Bavaresco, D.V.; Uggioni, M.L.R.; Ferraz, S.D.; Marques, R.M.M.; Simon, C.S.; Dagostin, V.S.; Grande, A.J.; da Rosa, M.I. Efficacy of infliximab in treatment-resistant depression: A systematic review and meta-analysis. Pharmacol. Biochem. Behav. 2020, 188. [Google Scholar] [CrossRef]
  151. McIntyre, R.S.; Subramaniapillai, M.; Lee, Y.; Pan, Z.; Carmona, N.E.; Shekotikhina, M.; Rosenblat, J.D.; Brietzke, E.; Soczynska, J.K.; Cosgrove, V.E.; et al. Efficacy of Adjunctive Infliximab vs Placebo in the Treatment of Adults with Bipolar I/II Depression: A Randomized Clinical Trial. JAMA Psychiatry 2019, 76, 783–790. [Google Scholar] [CrossRef] [PubMed]
  152. Lee, Y.; Mansur, R.B.; Brietzke, E.; Carmona, N.E.; Subramaniapillai, M.; Pan, Z.; Shekotikhina, M.; Rosenblat, J.D.; Suppes, T.; Cosgrove, V.E.; et al. Efficacy of adjunctive infliximab vs. placebo in the treatment of anhedonia in bipolar I/II depression. Brain Behav. Immun. 2020, 88, 631–639. [Google Scholar] [CrossRef] [PubMed]
  153. Mansur, R.B.; Delgado-Peraza, F.; Subramaniapillai, M.; Lee, Y.; Iacobucci, M.; Rodrigues, N.; Rosenblat, J.D.; Brietzke, E.; Cosgrove, V.E.; Kramer, N.E.; et al. Extracellular Vesicle Biomarkers Reveal Inhibition of Neuroinflammation by Infliximab in Association with Antidepressant Response in Adults with Bipolar Depression. Cells 2020, 9, 895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Mansur, R.B.; Subramaniapillai, M.; Lee, Y.; Pan, Z.; Carmona, N.E.; Shekotikhina, M.; Iacobucci, M.; Rodrigues, N.; Nasri, F.; Rashidian, H.; et al. Leptin mediates improvements in cognitive function following treatment with infliximab in adults with bipolar depression. Psychoneuroendocrinology 2020, 120, 104779. [Google Scholar] [CrossRef]
  155. Mansur, R.B.; Subramaniapillai, M.; Lee, Y.; Pan, Z.; Carmona, N.E.; Shekotikhina, M.; Iacobucci, M.; Rodrigues, N.; Nasri, F.; Rosenblat, J.D.; et al. Effects of infliximab on brain neurochemistry of adults with bipolar depression. J. Affect. Disord. 2021, 281, 61–66. [Google Scholar] [CrossRef]
  156. Bayramgürler, D.; Karson, A.; Özer, C.; Utkan, T. Effects of long-term etanercept treatment on anxiety- and depression-like neurobehaviors in rats. Physiol. Behav. 2013, 119, 145–148. [Google Scholar] [CrossRef] [PubMed]
  157. Krügel, U.; Fischer, J.; Radicke, S.; Sack, U.; Himmerich, H. Antidepressant effects of TNF-α blockade in an animal model of depression. J. Psychiatr. Res. 2013, 47, 611–616. [Google Scholar] [CrossRef]
  158. Brymer, K.J.; Fenton, E.Y.; Kalynchuk, L.E.; Caruncho, H.J. Peripheral etanercept administration normalizes behavior, hippocampal neurogenesis, and hippocampal reelin and GABAA receptor expression in a preclinical model of depression. Front. Pharmacol. 2018, 9. [Google Scholar] [CrossRef]
  159. Alshammari, M.A.; Khan, M.R.; Majid Mahmood, H.; Alshehri, A.O.; Alasmari, F.F.; Alqahtani, F.M.; Alasmari, A.F.; Alsharari, S.D.; Alhossan, A.; Ahmad, S.F.; et al. Systemic TNF-α blockade attenuates anxiety and depressive-like behaviors in db/db mice through downregulation of inflammatory signaling in peripheral immune cells. Saudi Pharm. J. 2020, 28, 621–629. [Google Scholar] [CrossRef]
  160. Tyring, S.; Bagel, J.; Lynde, C.; Klekotka, P.; Thompson, E.H.Z.; Gandra, S.R.; Shi, Y.; Kricorian, G. Patient-reported outcomes in moderate-to-severe plaque psoriasis with scalp involvement: Results from a randomized, double-blind, placebo-controlled study of etanercept. J. Eur. Acad. Dermatol. Venereol. 2013, 27, 125–128. [Google Scholar] [CrossRef]
  161. Jin, W.; Zhang, S.; Duan, Y. Depression symptoms predict worse clinical response to etanercept treatment in psoriasis patients. Dermatology 2018, 235, 55–64. [Google Scholar] [CrossRef] [PubMed]
  162. Gelfand, J.M.; Kimball, A.B.; Mostow, E.N.; Chiou, C.F.; Patel, V.; Xia, H.A.; Freundlich, B.; Stevens, S.R. Patient-reported outcomes and health-care resource utilization in patients with psoriasis treated with etanercept: Continuous versus interrupted treatment. Value Health 2008, 11, 400–407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Daudén, E.; Griffiths, C.E.M.; Ortonne, J.P.; Kragballe, K.; Molta, C.T.; Robertson, D.; Pedersen, R.; Estojak, J.; Boggs, R. Improvements in patient-reported outcomes in moderate-to-severe psoriasis patients receiving continuous or paused etanercept treatment over 54 weeks: The CRYSTEL study. J. Eur. Acad. Dermatol. Venereol. 2009, 23, 1374–1382. [Google Scholar] [CrossRef]
  164. Gniadecki, R.; Robertson, D.; Molta, C.T.; Freundlich, B.; Pedersen, R.; Li, W.; Boggs, R.; Zbrozek, A.S. Self-reported health outcomes in patients with psoriasis and psoriatic arthritis randomized to two etanercept regimens. J. Eur. Acad. Dermatol. Venereol. 2012, 26, 1436–1443. [Google Scholar] [CrossRef] [PubMed]
  165. Bae, S.C.; Gun, S.C.; Mok, C.C.; Khandker, R.; Nab, H.W.; Koenig, A.S.; Vlahos, B.; Pedersen, R.; Singh, A. Improved health outcomes with Etanercept versus usual DMARD therapy in an Asian population with established rheumatoid arthritis. BMC Musculoskelet. Disord. 2013, 14. [Google Scholar] [CrossRef] [Green Version]
  166. Machado, D.A.; Guzman, R.M.; Xavier, R.M.; Simon, J.A.; Mele, L.; Pedersen, R.; Ferdousi, T.; Koenig, A.S.; Kotak, S.; Vlahos, B. Open-label observation of addition of etanercept versus a conventional disease-modifying antirheumatic drug in subjects with active rheumatoid arthritis despite methotrexate therapy in the latin american region. J. Clin. Rheumatol. 2014, 20, 25–33. [Google Scholar] [CrossRef] [PubMed]
  167. Yang, A.; Xin, X.; Yang, W.; Li, M.; Li, L.; Liu, X. Etanercept reduces anxiety and depression in psoriasis patients, and sustained depression correlates with reduced therapeutic response to etanercept. Ann. Dermatol. Venereol. 2019, 146, 363–371. [Google Scholar] [CrossRef] [PubMed]
  168. Kimball, A.B.; Bensimon, A.G.; Guerin, A.; Yu, A.P.; Wu, E.Q.; Okun, M.M.; Bao, Y.; Gupta, S.R.; Mulani, P.M. Efficacy and Safety of Adalimumab among Patients with Moderate to Severe Psoriasis with Co-Morbidities. Am. J. Clin. Dermatol. 2011, 12, 51–62. [Google Scholar] [CrossRef]
  169. Bhutani, T.; Patel, T.; Koo, B.; Nguyen, T.; Hong, J.; Koo, J. A prospective, interventional assessment of psoriasis quality of life using a nonskin-specific validated instrument that allows comparison with other major medical conditions. J. Am. Acad. Dermatol. 2013, 69, e79–e88. [Google Scholar] [CrossRef]
  170. Gordon, K.B.; Armstrong, A.W.; Han, C.; Foley, P.; Song, M.; Wasfi, Y.; You, Y.; Shen, Y.K.; Reich, K. Anxiety and depression in patients with moderate-to-severe psoriasis and comparison of change from baseline after treatment with guselkumab vs. adalimumab: Results from the Phase 3 VOYAGE 2 study. J. Eur. Acad. Dermatol. Venereol. 2018, 32, 1940–1949. [Google Scholar] [CrossRef]
  171. Ward, A.; Clissold, S.P. Pentoxifylline. A Review of its Pharmacodynamic and Pharmacokinetic Properties, and its Therapeutic Efficacy. Drugs 1987, 34, 50–97. [Google Scholar] [CrossRef] [PubMed]
  172. Hiatt, W.R. Medical treatment of peripheral arterial disease and claudication. N. Engl. J. Med. 2001, 344, 1608–1621. [Google Scholar] [CrossRef]
  173. Sliwa, K.; Skudicky, D.; Candy, G.; Wisenbaugh, T.; Sareli, P. Randomised investigation of effects of pentoxifylline on left-ventricular performance in idiopathic dilated cardiomyopathy. Lancet 1998, 351, 1091–1093. [Google Scholar] [CrossRef]
  174. Sliwa, K.; Woodiwiss, A.; Candy, G.; Badenhorst, D.; Libhaber, C.; Norton, G.; Skudicky, D.; Sareli, P. Effects of pentoxifylline on cytokine profiles and left ventricular performance in patients with decompensated congestive heart failure secondary to idiopathic dilated cardiomyopathy. Am. J. Cardiol. 2002, 90, 1118–1122. [Google Scholar] [CrossRef]
  175. Sliwa, K.; Woodiwiss, A.; Kone, V.N.; Candy, G.; Badenhorst, D.; Norton, G.; Zambakides, C.; Peters, F.; Essop, R. Therapy of Ischemic Cardiomyopathy with the Immunomodulating Agent Pentoxifylline: Results of a Randomized Study. Circulation 2004, 109, 750–755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Fernandes, J.L.; de Oliveira, R.T.D.; Mamoni, R.L.; Coelho, O.R.; Nicolau, J.C.; Blotta, M.H.S.L.; Serrano, C.V. Pentoxifylline reduces pro-inflammatory and increases anti-inflammatory activity in patients with coronary artery disease-A randomized placebo-controlled study. Atherosclerosis 2008, 196, 434–442. [Google Scholar] [CrossRef]
  177. González-Espinoza, L.; Rojas-Campos, E.; Medina-Pérez, M.; Peña-Quintero, P.; Gómez-Navarro, B.; Cueto-Manzano, A.M. Pentoxifylline decreases serum levels of tumor necrosis factor alpha, interleukin 6 and C-reactive protein in hemodialysis patients: Results of a randomized double-blind, controlled clinical trial. Nephrol. Dial. Transpl. 2012, 27, 2023–2028. [Google Scholar] [CrossRef] [Green Version]
  178. De, B.K.; Gangopadhyay, S.; Dutta, D.; Baksi, S.D.; Pani, A.; Ghosh, P. Pentoxifylline versus prednisolone for severe alcoholic hepatitis: A randomized controlled trial. World J. Gastroenterol. 2009, 15, 1613–1619. [Google Scholar] [CrossRef]
  179. Zeni, F.; Pain, P.; Vindimian, M.; Gay, J.P.; Gery, P.; Bertrand, M.; Page, Y.; Page, D.; Vermesch, R.; Bertrand, J.C. Effects of pentoxifylline on circulating cytokine concentrations and hemodynamics in patients with septic shock: Results from a double-blind, randomized, placebo-controlled study. Crit. Care Med. 1996, 24, 207–214. [Google Scholar] [CrossRef]
  180. Harris, E.; Schulzke, S.M.; Patole, S.K. Pentoxifylline in preterm neonates: A systematic review. Pediatr. Drugs 2010, 12, 301–311. [Google Scholar] [CrossRef]
  181. Semmler, J.; Gebert, U.; Eisenhut, T.; Moeller, J.; Schonharting, M.M.; Allerat, A. Xanthine derivatives: Comparison between suppression of tumour necrosis factor-a production and inhibition of cAMP phosphodiesterase activity. Immunology 1993, 78, 520–525. [Google Scholar] [PubMed]
  182. Mohamed, B.; Aboul-Fotouh, S.; Ibrahim, E.A.; Shehata, H.; Mansour, A.A.; Az Yassin, N.; El-Eraky, W.; Abdel-Twab, A.M. Effects of pentoxifylline, 7-nitroindazole, and imipramine on tumor necrosis factor-α and indoleamine 2,3-dioxygenase enzyme activity in the hippocampus and frontal cortex of chronic mild-stress-exposed rats. Neuropsychiatr. Dis. Treat. 2013, 9, 697–708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Elgarf, A.S.A.; Aboul-Fotouh, S.; Abd-Alkhalek, H.A.; El Tabbal, M.; Hassan, A.N.; Kassim, S.K.; Hammouda, G.A.; Farrag, K.A.; Abdel-Tawab, A.M. Lipopolysaccharide repeated challenge followed by chronic mild stress protocol introduces a combined model of depression in rats: Reversibility by imipramine and pentoxifylline. Pharmacol. Biochem. Behav. 2014, 126, 152–162. [Google Scholar] [CrossRef]
  184. Duman, D.G.; Ozdemir, F.; Birben, E.; Keskin, O.; Ekşioğlu-Demiralp, E.; Celikel, C.; Kalayci, O.; Kalayci, C. Effects of pentoxifylline on TNF-α production by peripheral blood mononuclear cells in patients with nonalcoholic steatohepatitis. Dig. Dis. Sci. 2007, 52, 2520–2524. [Google Scholar] [CrossRef] [PubMed]
  185. El-Lakkany, N.; Seif el-Din, S.; Ebeid, F. The use of pentoxifylline as adjuvant therapy with praziquantel downregulates profibrogenic cytokines, collagen deposition and oxidative stress in experimental schistosomiasis mansoni. Exp. Parasitol. 2011, 129, 152–157. [Google Scholar] [CrossRef]
  186. Vakili, A.; Mojarrad, S.; Akhavan, M.M.; Rashidy-Pour, A. Pentoxifylline attenuates TNF-α protein levels and brain edema following temporary focal cerebral ischemia in rats. Brain Res. 2011, 1377, 119–125. [Google Scholar] [CrossRef]
  187. Bah, T.M.; Kaloustian, S.; Rousseau, G.; Godbout, R. Pretreatment with pentoxifylline has antidepressant-like effects in a rat model of acute myocardial infarction. Behav. Pharmacol. 2011, 22, 779–784. [Google Scholar] [CrossRef] [PubMed]
  188. El-Haggar, S.M.; Eissa, M.A.; Mostafa, T.M.; El-Attar, K.S.; Abdallah, M.S. The phosphodiesterase inhibitor pentoxifylline as a novel adjunct to antidepressants in major depressive disorder patients: A proof-of-concept, randomized, double-blind, placebo-controlled trial. Psychother. Psychosom. 2018, 87, 331–339. [Google Scholar] [CrossRef]
  189. Berk, M.; Walker, A.J.; Nierenberg, A.A. Biomarker-Guided Anti-inflammatory Therapies: From Promise to Reality Check. JAMA Psychiatry 2019, 76, 779–780. [Google Scholar] [CrossRef]
  190. Berk, M.; Vieta, E.; Dean, O.M. Anti-inflammatory treatment of bipolar depression: Promise and disappointment. Lancet Psychiatry 2020, 7, 467–468. [Google Scholar] [CrossRef]
  191. Boado, R.J.; Hui, E.K.W.; Lu, J.Z.; Zhou, Q.H.; Pardridge, W.M. Selective targeting of a TNFR decoy receptor pharmaceutical to the primate brain as a receptor-specific IgG fusion protein. J. Biotechnol. 2010, 146, 84–91. [Google Scholar] [CrossRef] [Green Version]
  192. Pardridge, W.M. Biologic TNFα-inhibitors that cross the human blood-brain barrier. Bioeng. Bugs 2010, 1, 233–236. [Google Scholar] [CrossRef] [Green Version]
  193. Chang, R.; Knox, J.; Chang, J.; Derbedrossian, A.; Vasilevko, V.; Cribbs, D.; Boado, R.J.; Pardridge, W.M.; Sumbria, R.K. Blood-Brain Barrier Penetrating Biologic TNF-α Inhibitor for Alzheimer’s Disease. Mol. Pharm. 2017, 14, 2340–2349. [Google Scholar] [CrossRef]
Figure 1. TNF-α and TNF-α Receptors. Transmembrane TNF-α (mTNF-α) undergoes proteolytic cleavage by TNF-α-converting enzyme (TACE) which generates the soluble form of the protein (sTNF-α). Both mTNF-α and sTNF-α are biologically active; they bind to and activate TNF receptor (TNFR) 1 and TNFR2. Arrows indicate that mTNF-α is also capable of activating TNFR1 and TNFR2.
Figure 1. TNF-α and TNF-α Receptors. Transmembrane TNF-α (mTNF-α) undergoes proteolytic cleavage by TNF-α-converting enzyme (TACE) which generates the soluble form of the protein (sTNF-α). Both mTNF-α and sTNF-α are biologically active; they bind to and activate TNF receptor (TNFR) 1 and TNFR2. Arrows indicate that mTNF-α is also capable of activating TNFR1 and TNFR2.
Molecules 26 02368 g001
Figure 2. TNF-α Antagonists. Clinically used selective TNF-α antagonists include recombinant TNF-α-specific monoclonal antibodies such as infliximab and adalimumab, and recombinant fusion proteins of TNFR such as etanercept which is a TNFR2 fusion protein. Pentoxifylline is a methylxanthine drug which exerts several pharmacological effects including potent inhibition of TNF-α activity (i.e., it is not a selective TNF-α antagonist). Abbreviations: ECD—extracellular domain, Fc—fragment crystallizable region, Fv—variable fragment, IgG—immunoglobulin G, TNFR2 – TNF-α receptor 2.
Figure 2. TNF-α Antagonists. Clinically used selective TNF-α antagonists include recombinant TNF-α-specific monoclonal antibodies such as infliximab and adalimumab, and recombinant fusion proteins of TNFR such as etanercept which is a TNFR2 fusion protein. Pentoxifylline is a methylxanthine drug which exerts several pharmacological effects including potent inhibition of TNF-α activity (i.e., it is not a selective TNF-α antagonist). Abbreviations: ECD—extracellular domain, Fc—fragment crystallizable region, Fv—variable fragment, IgG—immunoglobulin G, TNFR2 – TNF-α receptor 2.
Molecules 26 02368 g002
Table 1. Summary of clinical trials reporting on the mood-modulating effects of anti-TNF-α compounds in patients with various disease conditions.
Table 1. Summary of clinical trials reporting on the mood-modulating effects of anti-TNF-α compounds in patients with various disease conditions.
CompoundStudy DesignSample Size (Total)Disease ConditionType of Comparison (Follow-Up Duration) *Effect of TreatmentRef.
InfliximabProspective, non-randomized trialn = 100Crohn’s diseaseAll patients were treated with infliximab + standard therapy (4 weeks)Significant decrease in the proportion of depressed patients [134]
Prospective, non-randomized trialn = 14All patients were treated with infliximab + standard therapy (4 weeks)Significant reduction in depressive symptoms[135]
Prospective, non-randomized trialn = 29Ankylosing spondylitisAll patients were treated with three doses of infliximab + standard therapy (6 weeks)Significant reduction in depressive symptoms[136]
Randomized, placebo-controlled trialn = 23Standard therapy + placebo vs. standard therapy + infliximab, followed by infliximab-only treatment (54 weeks)Significant reduction in depressive symptoms[146]
Randomized, double-blind, placebo-controlled trialn = 60Major depressive disorder
(treatment-resistant)
Antidepressant(s) or medication free + placebo vs. antidepressant(s) or medication free + infliximab (12 weeks)Overall, no significant difference between groups. Infliximab significantly decreased depressive symptoms in a sub-group of patients with high baseline CRP levels [149]
Systematic review
and meta-analysis of four randomized controlled trials
n = 152Standard therapy + placebo vs. standard therapy + infliximabAdjunctive infliximab treatment did not have a significant effect on depressive symptoms [150]
Randomized, double-blind, placebo-controlled trialn = 60Bipolar depression with higher inflammatory activityStandard therapy + placebo vs. standard therapy + infliximab (12 weeks)No significant difference between groups. Infliximab significantly decreased depressive symptoms in a sub-group of patients with a history of childhood physical abuse[151]
Randomized, double-blind, placebo-controlled trialn = 60Standard therapy + placebo vs. standard therapy + infliximab (12 weeks)Adjunctive infliximab treatment led to a significant although transient anti-anhedonic effect[152]
Randomized, double-blind, placebo-controlled trialn = 55Bipolar depressionStandard therapy + placebo vs. standard therapy + infliximab (12 weeks)Significant reduction in depressive symptoms[153]
Randomized, double-blind, placebo-controlled trialn = 60Standard therapy + placebo vs. standard therapy + infliximab (12 weeks)Significant improvement in cognitive function (verbal memory)[154]
Randomized, double-blind, placebo-controlled trialn = 33Standard therapy + placebo vs. standard therapy + infliximab (12 weeks)Significant improvement in cognitive function but no significant effect on depressive symptoms[155]
EtanerceptRandomized, double-blind, placebo-controlled trial (phase 3)n = 618PsoriasisStandard therapy + placebo vs. standard therapy + etanercept (12 weeks)Significant decrease in depressive symptoms[137]
Prospective open-labeled trial (open-phase continuum of the study reported in reference # 137) n = 591Standard therapy + etanercept (84 weeks)A sustained significant decrease in depressive symptoms[138]
Randomized, double-blind, placebo-controlled trialn = 121Standard therapy + placebo vs. standard therapy + etanercept (24 weeks)Significant decrease in depressive symptoms[160]
Prospective, non-randomized trial n = 85Standard therapy + etanercept (24 weeks)Significant reduction in depression and anxiety symptoms[161]
Prospective, non-randomized (open-labeled) trialn = 2546Standard therapy + etanercept given in two regimens—continues vs. interrupted (24 weeks)Etanercept treatment (both regiments) led to a significant decrease in depressive symptoms[162]
Prospective, non-randomized (open-labeled) trialn = 711Standard therapy + etanercept given in two regimens—continues vs. interrupted (54 weeks)Etanercept treatment (both regiments) led to a significant improvement in depressive symptoms[163]
Part 1: A randomized, double-blind, dose-adjusted trial; Part 2: Open-labeled trial n = 752Standard therapy + etanercept given in various regimens (24 weeks)Significant reduction in depression and anxiety symptoms[164]
AdalimumabRandomized, double-blind, placebo-controlled trial (phase 3)n = 499Crohn’s diseaseStandard therapy + adalimumab given in various regimens (56 weeks)Significant decrease in depressive symptoms[140]
Randomized, double-blind, placebo-controlled trialn = 96PsoriasisStandard therapy + placebo vs. standard therapy + adalimumab (12 weeks)Significant decrease in depressive symptoms[128]
Prospective, non-randomized trialn = 143Standard therapy + adalimumab (24 weeks)Significant reduction in depression and anxiety symptoms[129]
Randomized, double-blind, placebo-controlled trialn = 828Standard therapy + placebo vs. standard therapy + adalimumab (16 weeks)Significant decrease in depressive symptoms[168]
Prospective, non-randomized trialn = 32Standard therapy + adalimumab (24 weeks)Significant decrease in depressive symptoms[169]
Randomized, double-blind, placebo-controlled trial (phase 3)n = 992Standard therapy + placebo vs. standard therapy + adalimumab (and vs. standard therapy + guselkumab) (24 weeks)Adalimumab significantly decreased depression and anxiety symptoms[170]
Randomized, double-blind, placebo-controlled trial (phase 2)n = 154Hidradenitis suppurativaStandard therapy + placebo vs. standard therapy + adalimumab (16 weeks)Adalimumab significantly decreased depressive symptoms in patients with high baseline pain score[130]
PentoxifyllineRandomized, double-blind, placebo-controlled trialn = 100Major depressive disorderEscitalopram + placebo vs. escitalopram + pentoxifylline (12 weeks)Significant decrease in depressive symptoms[188]
* Type of comparison and follow-up duration are indicated in the table only if they were clearly mentioned in the reporting article. CRP denotes C-reactive protein.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Uzzan, S.; Azab, A.N. Anti-TNF-α Compounds as a Treatment for Depression. Molecules 2021, 26, 2368. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26082368

AMA Style

Uzzan S, Azab AN. Anti-TNF-α Compounds as a Treatment for Depression. Molecules. 2021; 26(8):2368. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26082368

Chicago/Turabian Style

Uzzan, Sarit, and Abed N. Azab. 2021. "Anti-TNF-α Compounds as a Treatment for Depression" Molecules 26, no. 8: 2368. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26082368

Article Metrics

Back to TopTop