Open access peer-reviewed chapter

Perspective Chapter: New Use of the SSRI Fluvoxamine in the Treatment of COVID-19 Symptoms

Written By

Jawza F. Alsabhan and Tahani K. Alshammari

Submitted: 27 February 2022 Reviewed: 21 April 2022 Published: 04 July 2022

DOI: 10.5772/intechopen.105023

From the Edited Volume

COVID-19 Pandemic, Mental Health and Neuroscience - New Scenarios for Understanding and Treatment

Edited by Sara Palermo and Berend Olivier

Chapter metrics overview

105 Chapter Downloads

View Full Metrics

Abstract

From the perspective of repurposing medication, recent evidence suggests that the use of selective serotonin reuptake inhibitor antidepressants (SSRIs) can help reduce the severity of symptoms and death associated with SARS-CoV-2 infection. To focus more, COVID-19 is a viral disease with potentially high risk of symptoms. There is presently no cure. However, there are specific treatments that may help manage the condition. Since the SSRI fluvoxamine has a unique mechanism of action in reducing cytokine production, researchers have started to relate the antiviral effects via modulation of sigma-1 receptors with the vision of treatment options for COVID-19 patients. The scope of this chapter is to examine different mechanisms of fluvoxamine in relation to immune response, including both the serotonin and the sigma-1 receptor-related mechanisms. Addressing the impact of fluvoxamine in minimizing possible complications during COVID-19 infection.

Keywords

  • fluvoxamine
  • anti-inflammatory
  • coronavirus disease 2019
  • sigma-1 receptor
  • clinical studies
  • preclinical studies

1. Introduction – COVID-19 options for treatment

The coronavirus disease 2019 (COVID-19) is an acute respiratory infection related to the new RNA virus coronavirus 2, that produces severe acute respiratory syndrome (SARS-CoV-2) [1]. Since the first SARS-CoV-2-infected patients were reported in Wuhan, China, in December 2019, the number of individuals infected with COVID-19 has risen significantly around the world [2]. In January 2020, the World Health Organization (WHO) announced that COVID-19 had been labeled as a public health pandemic disease worldwide [3]. Likewise, during the last three years, individuals worldwide face a large wave of COVID-19 produced by SARS-CoV-2 variants (i.e., delta, lambda, mu, and omicron) on a widespread globe. In January 2022, the World Health Organization (WHO) published an updated report about enhancing the response to the new mutation called Omicron SARS-CoV-2 variant [4]. To focus more on COVID-19 symptoms, after the incubation period (2–14 days), three phases of COVID-19 symptoms appeared [1]. The first stage, known as the acute COVID-19 stage (stage I) characterized by mild to moderate symptoms including fever or chills, cough, shortness of breath fatigue, muscle or body aches, headache, new loss of taste or smell, sore throat, congestion or running nose, nausea, and diarrhea [2]. The prevalence of mild to moderate symptoms is significant in SARS-CoV-2-infected individuals [5]. Post-acute viral syndromes are acknowledged as prolonged and multi-organ effects [6]. The second stage (stage II) is less common and characterized by severe clinical stages such as dyspnea accompanied by hypoxemia [7, 8]. As a consequence of severe clinical symptoms, stage III can occur, including serious difficulties such as arrhythmias, septic shock, and multi-organ failures, ultimately resulting in intubation or death [2]. A high proportion of COVID-19 non survivors had pre-existing cardiovascular illness and multi organ dysfunction [9].

SARS-CoV-2 infection appears to have harmful effects on the central nervous system (CNS), leading to psychiatric and neurological symptoms [10]. Brain-related manifestations were reported in individuals with and without neuropsychiatric diseases [11]. Existing in vitro and in vivo studies support the notion of SARS-CoV-2-related neuroinvasive risks. These risks were transported mainly through olfaction and the trigeminal pathways [12]. According to national longitudinal research in China, depression, anxiety, and insomnia were the most common psychiatric disorders associated with the COVID-19 pandemic [13]. Another study indicated that the prevalence of neurological manifestations was reasonably noted, including encephalopathy, delirium, seizures, Guillain-Barré-syndrome, and motor dysfunctions [14]. Another review reported that the emergence of psychiatric disorders occurs within three months in COVID-19 survivors [6].

COVID-19 outbreaks present challenges for therapeutic and drug management in clinical conditions and complications. Recently, several researchers emphasized medication repurposing as an approaching method to establish a fast-track line for discovering therapeutic treatments for COVID-19 patients worldwide [15, 16, 17, 18]. Drug repurposing, also known as drug reuse, is defined as exploring new indications for existing drugs. This approach limits the cost of both research and development significantly [19]. Serendipitously revealed a substantial number of current identified repurposed drugs [20]. The repurposing strategy provided promising treatment candidates in other viral pandemics outbreaks, including ZIKA, Ebola, and dengue [21].

Zhang et al summarized the current potential therapeutic approaches for diseases related to COVID-19 infection and explain their mechanisms of action, safety, and effectiveness such as antiviral drugs, convalescent plasma, spike Protein-Angiotensin-Converting enzyme 2 blockers, chloroquine, hydroxychloroquine, human monoclonal antibody, mesenchymal stem cells, inhaled nitric oxide (iNO), and corticosteroids [22]. Another review highlighted alternative treatment approaches such as mesenchymal stem cells, human monoclonal antibodies, and complementary Chinese medicine [20].

A comprehensive and systematic research about the repurposing of drugs in COVID-19 was conducted by Srivastava and Singh (2021), the results revealed that no drugs passed clinical trials or were approved by the FDA for COVID-19 [23]. In fact, various reports that clinical trials regarding the potential therapeutic possibilities, such as hydroxychloroquine, lopinavir-ritonavir, and ivermectin, showed that choices were unsuccessful in the treatment of COVID-19 patients [24, 25, 26, 27]. Although COVID-19 vaccines were successfully developed and massively distributed, there is a lack of effective treatment [28].

Most significantly, there are cumulative clinical data linked with the use of antidepressants, including the selective serotonin reuptake inhibitor (SSRI) and the serotonin-norepinephrine reuptake inhibitor (SNRI) with a reduced risk of clinical complications in SARS-CoV-2-infected patients [18]. In line with this, a randomized controlled trial showed that patients who received fluvoxamine had a decreased risk of clinical deterioration compared to those who received placebo during the early period of infection [29]. Following that, an observational study done by Hoertel et al. discovered that the antidepressants, such as SSRIs and SNRIs, may be associated with a lower risk of intubation or death in SARS-CoV-2-infected hospitalized patients [30]. This advantage was not reported with other antipsychotics. For example, a multicenter observational study indicated that administration of haloperidol was not associated with a reduced risk of intubation or death in hospitalized COVID-19 patients [31].

Fluvoxamine has advantages over other repurposing drugs such as positive safety profiles, widespread availability, inexpensive, accessible mode of administration as immediate-release tablets and controlled-release capsules, and use for children and adolescents [32, 33]. The available dosage form of fluvoxamine is both immediate-release tablets and controlled-release capsules. The main therapeutic indication of fluvoxamine is to treat patients with obsessive-compulsive disorder (OCD). The pharmacokinetic properties of fluvoxamine include long half-life of about 9–28 hours according to its dosage form. The clinical guideline recommended the therapeutic dose of fluvoxamine between 100–300 mg/day [34]. Fluvoxamine indication is related to COVID-19 was clarified on the US National Institutes of Health (NIH) COVID-19 Guidelines Panel on April 23, 2021, despite the fact that evidence for fluvoxamine effectiveness was lacking. Recently, a retrospective cohort study of COVID-19 patients treated with SSRIs using electronic health records of 87 health care centers across the US was done in November 2021 by Oskotsky et al. [35]. The study participants were COVID-19 patients receiving fluoxetine only (n = 470), COVID-19 patients receiving fluoxetine or fluvoxamine (n = 481), and COVID-19 patients receiving other SSRIs (n = 2898) compared with matched untreated control COVID-19 patients. The results showed that the patients who received any SSRI had a lower risk of death. In contrast, there was no significant relationship between SSRIs other than fluoxetine or fluvoxamine and the risk of mortality [35]. Finally, fluvoxamine might be used as a preventive medication for patients infected with SARS-CoV-2 in the early stages [16, 36]. In this chapter, we review the fluvoxamine mechanisms of action in COVID-19 and discuss the studies that focus on the impact of the use of fluvoxamine in minimizing the possible complication during COVID-19 infection. Here, we review two main mechanisms mediated by fluvoxamine, the serotonin and the sigma-1 receptor-related mechanisms.

Advertisement

2. Mechanism of fluvoxamine

2.1 Inhibition of serotonin transporter

The main mechanism of action for fluvoxamine, as a selective serotonin-reuptake inhibitor (SSRIs), in the brain is to inhibit the serotonin transporter (SERT) on the 5-HT neuron in the synaptic cleft leading to an increase in the level of the neurotransmitter serotonin [33, 37].

The serotonin transporter is also located on the platelet membranes [38] and in the gut. The serotonin plasma-platelet regulation is a complex biphasic mechanism. Specifically, an elevation in plasma serotonin is associated with reducing platelet SERT surface expression [39]. As a consequence of serotonin transporter inhibition in the platelets by fluvoxamine, this could reduce platelet aggregation [40]. In support of this, elevated circulating serotonin was reported in hypertensive patients [41]. In fact, previously proven that fluvoxamine should be used with caution in patients receiving NSAIDs, aspirin, or other drugs that may impair coagulation and peptic ulcer patients due to the antiplatelet activity mechanism SSRIs that can increase the risk of bleeding [42, 43]. Interestingly, a previous report indicated that transgenic mice harboring SERT construct mutation exhibited altered platelet aggregation [44]. Overall, the physiological role of SERT in Platelets includes maintaining system homeostasis, regulation of drugs’ concentration, and function [39, 45].

Besides, the antiplatelet activity mechanism of fluvoxamine has a protective effect against myocardial infarction (MI) and a promising role for patients with thrombotic risk [46]. Clearly, fluvoxamine has anti-inflammatory properties in several animal and clinical studies through the mechanism of serotonin transporter inhibition [47, 48]. For instance, in a Parkinson’s disease model, fluvoxamine has been shown to reduce inflammation in injured striatal neurons by elevating anti-inflammatory cytokines and lowering inflammatory cytokines and lipid peroxidation in a 6-hydroxydopamine (6-OHDA) lesion-induced rat model [49]. In line with this, chronic administration of fluvoxamine to the 6-OHDA Parkinsonism model improves the Parkinson’s-like symptoms, including a reduction in the dopaminergic neuronal degeneration, improvement in motor dysfunction, and normalization of corticosterone levels in the circuitry [50]. Another preclinical setting utilized the same model, the 6-OHDA Parkinsonism model, which indicated that chronic fluvoxamine treatment alters inflammatory hallmarks of Parkinsonism centrally using both the mRNA and the protein level analysis. The level of IL-1β, IL-6, and TNF-α was reduced following one month of Fluvoxamine treatment [49]. Highlighting the fluvoxamine-mediated pharmacological link between serotonin – anti-inflammatory mechanisms at central and peripheral levels.

Likewise, another study revealed that fluvoxamine inhibits inflammation genes in in vitro settings [32]. In multiple sclerosis, fluvoxamine reduced the lymphocyte infiltration and the circulating IFN-γ in experimental autoimmune encephalomyelitis. It further affects the pathological demyelination of lumbar spinal cord in multiple sclerosis animal models [51]. In the context of dementia, a previous review highlighted the anti-inflammatory effects of SSRIs, and the evidence provided indicated that introducing SSRIs in neurodegenerative dementias has beneficial effects [52]. Previous studies have proven that fluvoxamine has potent anti-inflammatory properties in vitro and in vivo models. Besides being an effective anti-inflammatory agent, fluvoxamine was found to have antioxidant effects. In a rat model of ulcers, the stress-induced peptic ulcer model, compared to the control group, the fluvoxamine-treated group exhibited reduced biochemical measurements representing oxidative stress [53]. Most significantly, the serotonin transporter in rodents and humans is highly expressed by the lung, demonstrating that the serotonin transporter in lung endothelium controls the bioavailability of the potent vasoconstrictor serotonin [54, 55]. It was reported that inhibiting gut- and lung-serotonin modulates pulmonary hypertension [56]. Additionally, fluvoxamine can positively improve the lung function of chronic obstructive pulmonary disease (COPD) [57]. For depressed COPD patients, SSRIs are the first-line treatment [58]. However, there is a need for a further clinical study to clarify more about the role of fluvoxamine in the treatment of COVID-19 patients. Although other antidepressants can act on serotonin transporter as an inhibitor, they did not have the same favorable impact on COVID-19 patients [57]. As a result, it is unclear whether fluvoxamine’s blockage of serotonin transporters plays a substantial role in its positive benefits for COVID-19 patients. The anti-inflammatory effects of serotonin transporter inhibition, on the other hand, may play a role in its positive benefits [55]. To support the potential use of SSRIs in the treatment of post-COVID-19 depression, a recent observational study showed that treatment with different SSRIs antidepressants has positive effects in patients with post-COVID-19 depression [59]. In support of this, another report highlighted that SSRIs might decrease the rate of mortality in COVID-19 patients [35]. However, different mechanism of action needs to be clarified more about the role of fluvoxamine in the treatment of COVID-19 patients. These mechanisms might be acid sphingomyelinase mediated [60], or melatonin receptor-mediated [61].

2.2 The Sigma-1 receptor-mediated mechanism

Several studies established the role of the sigma-1 receptor in the replication of SARS-CoV-2. Initially, it was recognized by its action as a cellular factor known as endoplasmic reticulum (ER) protein sigma-1 receptor that mediates the early steps of viral RNA replication but not the persistent hepatitis C virus (HCV) RNA replication [62]. The ER chaperones are required to control the production of glycosylated proteins in the cell, including those required for viral infection phases and those involved in immune escape [63]. Therefore, they play various important roles at various stages of the infectious cycle. A recent study recognized protein-protein interactions between SARS-CoV-2 and human proteins including the sigma-1 and sigma-2 receptors [64]. In line with this, a recent report linked this molecular pathway and SARS-CoV-2 replication [65]. Moreover, sigma-1 receptor agonists may protect against mitochondrial damage and ER stress in response to SARS-CoV-2 infection [66]. For that reason, the use of drugs with sigma-1 receptor ligand properties may enhance additive value in early intervention for COVID-19 patients [67].

Previous reports have linked sigma-1 receptor and psychiatric disorders, including anxiety [68], depression [69], and schizophrenia [70].

Cognitive deficit is a core feature of psychiatric disorders, including depression. Reports highlighted the role of the sigma-1 receptor in the fluvoxamine antidepressant mediated mechanism [71]. First, Sigma-1 receptors are expressed abundantly in cortical and motor brain regions [72], indicating functional involvement in the pathology of depression. Additionally, electrophysiological studies indicated that sigma-1 receptor modulated glutamatergic transmission in primary hippocampal neurons. Besides, they were found to be highly expressed in the olfactory bulb [72], a brain region involved in adult neurogenesis. Adult neurogenesis is involved in modulating synaptic plasticity [73] and disrupted in preclinical models of depression [74]. A previous study reported sigma-1 receptor’s functional involvement in modulating adult neurogenesis [75]. In a model of Alzheimer’s with cognitive impairment, sigma-1 receptors were found to promote spines’ maintenance and maturation [76]. Recent studies highlight the role of spine modulation in promoting fast-acting antidepressant effects [77], signifying the role of Sigma-1 receptors in tacking pathological mechanisms of depression.

Besides, sigma-1 receptors were reported to modulate protein kinase signaling (pERK) molecularly in a neuropathic pain animal model [78], and this kinase pathway was found to be involved in depression [79].

In addition, a recent review found that the significant potentiation of the nerve growth factor (NGF) by SSRIs including fluvoxamine leading to induced neurite outgrowth in cell culture and brain plasticity via selective sigma-1 receptor agonistic activity [80]. In clinical settings, NGF was reported to be reduced in depressed patients [81], this reduction was correlated with suicidal tendencies [82]. Moreover, preclinical studies in animal models of depression reported reduced NGF levels [83, 84, 85].

Additionally, modulation of the sigma-1 receptor pathway positively impacted inflammatory pathways such as IL-6 tumor necrosis factor-α (TNFα) [86]. In line with this, sigma-1 receptor agonist (the pRE-084) presented neuroprotective effects measured by a decreased infarct volume in an embolic stroke model. This was mostly regraded y a reduction in inflammatory cytokines, including TNF-alpha and multiple isoforms of IL in cortical tissues [87]. In another study utilizing a stroke model in rats, the sigma-1 receptor was reported to exert neuroprotective effects, and these modulations were mediated via targeting glutamate-induced excitotoxicity [88]. Also, in a rat model of renal ischemia, the fluvoxamine mediated sigma-1 receptor mechanism was reported to enhance the survival rate, renal function, and histological characteristics [89]. All these evidence suggest that molecular modulation of the sigma-1 receptor has beneficial effects. It further confirms that the neuroprotective effects of fluvoxamine render it to be considered a prophylactic therapeutic candidate to prevent COVID-19 risks. However, further studies are required to answer whether the anti-inflammatory and other fluvoxamine neuroprotective effects of fluvoxamine are clinically relevant for COVID-19 patients.

For the most important part, which is the relation between the use of antidepressants and sigma-1 receptor, a study examined in an animal model the SSRIs binding affinity to the sigma-1 receptor [71]. As a result, the study revealed that different SSRIs, including sertraline, fluoxetine, citalopram, and fluvoxamine, have a significant functional impact ranging from high to moderate affinity for sigma-1 receptors in the rat brain (Table 1).

AntidepressantsAffinity to sigma 1 receptorAction at sigma receptorAnti-inflammatory effectReference
Fluvoxamine (SSRI)PotentAgonistBeneficial effect in both preclinical and clinical models of inflammation.[40, 62, 86, 90]
Sertraline (SSRI)High to moderateAntagonistStrong anti-inflammatory effects by lowering and controlling pro-inflammatory cytokines.[18, 71]
Fluoxetine (SSRI)High to moderateAgonistAnti-inflammatory effects of fluoxetine in lipopolysaccharide (LPS)-stimulated microglial cells.[91, 92]
Escitalopram (SSRI)High to moderateAgonistBeneficial effect as an anti-inflammatory in the preclinical module.[66, 93]
Citalopram(SSRI)High to moderateAgonistBeneficial effect in both preclinical and clinical models of inflammation.[48, 92]
Paroxetine (SSRI)Very weakPartial anti-inflammatory effect.[48]
Imipramine (TCA)Very weakAgonistWeak inflammatory effect.[48, 94]
Venlafaxine (SNRI)Very weakWeak inflammatory effect.[94]
Mirtazapine (NaSSA)Very weakNo sufficient evidence in clinical studies.[95]

Table 1.

Affinity of the antidepressants for sigma-1 receptor.

TCA: Tricyclic antidepressants; SSRI: Selective serotonin reuptake inhibitor; SNRI: Serotonin norepinephrine reuptake inhibitor; NaSSA: Noradrenaline specific serotonergic antidepressant. The measure for sigma affinity indicates that potent = 17 nM, high to moderate = 30–400 nM, very week > 10,000 nM.

Advertisement

3. Conclusion

In this chapter, we reviewed the mechanisms of action of fluvoxamine, including the serotonergic and the sigma-1 mediated mechanisms with regard to COVID-19 infection. Fluvoxamine is not FDA-approved for the treatment of any infection-related disorder. However, accumulated evidence highlighted potential beneficial effects.

To sum up, fluvoxamine could help to improve the clinical deterioration associated with COVID-19 symptoms. However, it is for future studies to mechanistically examine the potential mechanisms and profile consequences related to preventive measures of fluvoxamine in treating COVID-19 patients.

Advertisement

Conflicts of interest

The authors declare that there is no conflict of interest.

Advertisement

Funding

Not funded

References

  1. 1. Wiersinga WJ, Rhodes A, Cheng AC, Peacock SJ, Prescott HC. Pathophysiology, transmission, diagnosis, and treatment of coronavirus disease 2019 (COVID-19): A review. JAMA – Journal of American Medical Association. 2020;324(8):782-793
  2. 2. Guan W, Ni Z, Hu Y, Liang W, Ou C, He J, et al. Clinical characteristics of coronavirus. The New England Journal of Medicine. 2020;382(18):1708-1720
  3. 3. Novel Coronavirus (2019-nCoV). In: WHO. 2020. pp. 1-5
  4. 4. WHO. Enhancing response to Omicron SARS-CoV-2 Variant: Technical brief and priority actions for Member States. WHO; 2022. pp. 1-28
  5. 5. Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: A descriptive study. Lancet. 2020;395(10223):507-513
  6. 6. Nalbandian A, Sehgal K, Gupta A, Madhavan MV, McGroder C, Stevens JS, et al. Post-acute COVID-19 syndrome. Nature Medicine. 2021;27(4):601-615
  7. 7. Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan , China: A retrospective cohort study. Lancet. 2020;395(10229):1054-1062
  8. 8. Chaudhary S, Benzaquen S, Woo JG, Rubinstein J, Matta A, Albano J, et al. Clinical characteristics, respiratory mechanics, and outcomes in critically ill individuals with COVID-19 infection in an underserved urban population. Respiratory Care. 2021;66(6):897-907
  9. 9. Zaim S, Chong JH, Sankaranarayanan V, Harky A. COVID-19 and multiorgan response. Current Problems in Cardiology. 2020;45(8):100618
  10. 10. Guerrero JI, Barragán LA, Martínez JD, Montoya JP, Peña A, Sobrino FE, et al. Central and peripheral nervous system involvement by COVID-19: A systematic review of the pathophysiology, clinical manifestations, neuropathology, neuroimaging, electrophysiology, and cerebrospinal fluid findings. BMC Infectious Diseases. 2021;21(1):1-16
  11. 11. Panariello F, Cellini L, Speciani M, De Ronchi D, Atti AR. How does SARS-CoV-2 affect the central nervous system? A working hypothesis. Frontiers in Psychiatry. 2020;19(11):582345
  12. 12. Liu JM, Tan BH, Wu S, Gui Y, Le SJ, Li YC. Evidence of central nervous system infection and neuroinvasive routes, as well as neurological involvement, in the lethality of SARS-CoV-2 infection. Journal of Medical Virology. 2021;93(3):1304-1313
  13. 13. Shi L, Lu ZA, Que JY, Huang XL, Lu QD, Liu L, et al. Long-term impact of covid-19 on mental health among the general public: A nationwide longitudinal study in China. International Journal of Environmental Research and Public Health. 2021;18(16):8790
  14. 14. Najjar S, Najjar A, Chong DJ, Pramanik BK, Kirsch C, Kuzniecky RI, et al. Central nervous system complications associated with SARS-CoV-2 infection: Integrative concepts of pathophysiology and case reports. Journal of Neuroinflammation. 2020;17(1):1-14
  15. 15. Hsieh K, Wang Y, Chen L, Zhao Z, Savitz S. Drug repurposing for COVID - 19 using graph neural network and harmonizing multiple evidence. 2021;1-14
  16. 16. Hashimoto K. Repurposing of CNS drugs to treat COVID-19 infection: Targeting the sigma-1 receptor. European Archives of Psychiatry and Clinical Neuroscience. 2021;271(2):249-258
  17. 17. Barghash RF, Fawzy IM, Chandrasekar V, Singh AV, Katha U, Mandour AA. In silico modeling as a perspective in developing potential vaccine candidates and therapeutics for COVID-19. Coatings. 2021;11(1273):1-26
  18. 18. Gaber M, Hamed M, Samir R. The possible immunoregulatory and anti-inflammatory effects of selective serotonin reuptake inhibitors in coronavirus disease patients. Medical Hypotheses. 2020;144(110140):1-5
  19. 19. Pushpakom S, Iorio F, Eyers PA, Escott KJ, Hopper S, Wells A, et al. Drug repurposing: Progress, challenges and recommendations. Nature Reviews. Drug Discovery. 2018;18(1):41-58
  20. 20. Parvathaneni V, Gupta V. Utilizing drug repurposing against COVID-19 – Efficacy, limitations, and challenges. Life Sciences. 2020;15(259):118275
  21. 21. Mani D, Wadhwani A, Krishnamurthy PT. Drug repurposing in antiviral research: A current scenario. Journal of Young Pharmacists. 2019;11(2):117-121
  22. 22. Zhang J, Xie B, Hashimoto K. Brain, behavior , and immunity current status of potential therapeutic candidates for the COVID-19 crisis. Brain, Behavior, and Immunity. 2020;87(April):59-73
  23. 23. Srivastava K, Singh MK. Drug repurposing in COVID-19: A review with past , present and future. Metab Open. 2021;12(August):100121
  24. 24. Self WH, Semler MW, Leither LM, Casey JD, Angus DC, Brower RG, et al. Effect of hydroxychloroquine on clinical status at 14 days in hospitalized patients with COVID-19: A Randomized Clinical Trial. JAMA – Journal of American Medical Association. 2020;324(21):2165-2176
  25. 25. López-Medina E, López P, Hurtado IC, et al. Effect of ivermectin on time to resolution of symptoms among adults with mild COVID-19: A Randomized Clinical Trial. 2021;325(14):1426-1435
  26. 26. Abayomi A, Osibogun A, Ezechi O, Wright K, Ola B, Ojo O, et al. A multi-centre, randomized, double-blind, placebo-controlled clinical trial of the efficacy and safety of chloroquine phosphate, hydroxychloroquine sulphate and lopinavir/ritonavir for the treatment of COVID-19 in Lagos State: Study protocol for a randomi. Trials. 2021;22(869):1-19
  27. 27. Asili P, Mirahmad M, Tabatabaei-Malazy O, Manayi A, Haghighat E, Mahdavi M, et al. Characteristics of published/registered clinical trials on COVID-19 treatment: A systematic review. Journal of Pharmaceutical Sciences. 2021;29:449-467
  28. 28. Khataniar A, Pathak U, Rajkhowa S, Jha AN. A comprehensive review of drug repurposing strategies against known drug targets of COVID-19. Covid. 2022;2(2):148-167
  29. 29. Lenze EJ, Mattar C, Zorumski CF, Stevens A, Schweiger J, Nicol GE, et al. Fluvoxamine vs Placebo and clinical deterioration in outpatients with symptomatic COVID-19: A Randomized Clinical Trial. JAMA – Journal of American Medical Association. 2020;324(22):2292-2300
  30. 30. Hoertel N, Sánchez-Rico M, Vernet R, Beeker N, Jannot AS, Neuraz A, et al. Association between antidepressant use and reduced risk of intubation or death in hospitalized patients with COVID-19: Results from an observational study. Molecular Psychiatry. 2021;26(9):5199-5212
  31. 31. Hoertel N, Sa M, Neuraz A, Blanco C, Jannot S, Paris N, et al. Observational study of haloperidol in hospitalized patients with COVID-19. PLoS One. 2021;16(2):e0247122
  32. 32. Hashimoto Y, Suzuki T, Hashimoto K. Old drug fluvoxamine, new hope for COVID-19. European Archives of Psychiatry and Clinical Neuroscience. 2022;272(1):161-163
  33. 33. Cheer SM, Figgitt DP. A review of its therapeutic potential in the management of anxiety disorders in children and adolescents. Paediatric Drugs. 2001;3(10):763-781
  34. 34. FDA. Fluvoxamine Maleate Tablets Label. 2012. pp. 1-35
  35. 35. Oskotsky T, Marić I, Tang A, Oskotsky B, Wong RJ, Aghaeepour N, et al. Mortality risk among patients with COVID-19 prescribed selective serotonin reuptake inhibitor antidepressants. JAMA Neurology. 2021;4(11):1-11
  36. 36. Gordon DE, Jang GM, Bouhaddou M, Xu J, Obernier K, White KM, et al. A SARS-CoV-2 protein interaction map reveals targets for drug repurposing. Nature. 2020;583(7816):459-468
  37. 37. Al-timimi SJ, Jamil NF. Current pharmacological treatment of obsessive-compulsive disorder. Arabian Journal of Psychiatry. 2020;31(2):98-104
  38. 38. De Abajo FJ. Effects of selective serotonin reuptake inhibitors on platelet function: Mechanisms, clinical outcomes and implications for use in elderly patients. Drugs & Aging. 2011;28(5):345-367
  39. 39. Mercado CP, Kilic F. Molecular mechanisms of sert in platelets: Regulation of plasma serotonin levels. Molecular Interventions. 2010;10(4):231-241
  40. 40. Sukhatme VP, Reiersen AM, Vayttaden SJ, Sukhatme VV. Fluvoxamine: A review of its mechanism of action and its role in COVID-19. Frontiers in Pharmacology. 2021;12(April):1-9
  41. 41. Brenner B, Harney JT, Ahmed BA, Jeffus BC, Unal R, Mehta JL, et al. Plasma serotonin levels and the platelet serotonin transporter. Journal of Neurochemistry. 2007;102(1):206-215
  42. 42. Howland RH. Exploring psychotherapeutic issues and agents in clinical practice. Journal of Psychosocial Nursing. 2004;45(4):15-22
  43. 43. Laursen SB, Leontiadis GI, Stanley AJ, Hallas J, Schaffalitzky de Muckadell OB. The use of selective serotonin receptor inhibitors (SSRIs) is not associated with increased risk of endoscopy-refractory bleeding, rebleeding or mortality in peptic ulcer bleeding. Alimentary Pharmacology & Therapeutics. 2017;46(3):355-363
  44. 44. Carneiro AMD, Cook EH, Murphy DL, Blakely RD. Interactions between integrin αIIbβ3 and the serotonin transporter regulate serotonin transport and platelet aggregation in mice and humans. The Journal of Clinical Investigation. 2008;118(4):1544-1552
  45. 45. Jedlitschky G, Greinacher A, Kroemer HK. Transporters in human platelets: Physiologic function and impact for pharmacotherapy. Blood. 2012;119(15):3394-3402
  46. 46. Yang Y, Li X, Chen S, Xiao M, Liu Z, Li J, et al. Mechanism and therapeutic strategies of depression after myocardial infarction. Psychopharmacology. 2021;238(6):1401-1415
  47. 47. Rafiee L, Hajhashemi V, Javanmard SH. Fluvoxamine inhibits some inflammatory genes expression in LPS / stimulated human endothelial cells, U937 macrophages , and carrageenan-induced paw edema in rat. Iranian Journal of Basic Medical Sciences. 2016;19:977-984
  48. 48. Arteaga-henríquez G, Simon MS, Burger B, Weidinger E, Wijkhuijs A, Arolt V, et al. Low-grade inflammation as a predictor of antidepressant and anti- inflammatory therapy response in MDD Patients: A systematic review of the literature in combination with an analysis of experimental data collected in the EU-MOODINFLAME consortium. Frontiers in Psychiatry. 2019;10(458):1-15
  49. 49. Dallé E, Daniels WMU, Mabandla MV. Fluvoxamine maleate normalizes striatal neuronal inflammatory cytokine activity in a Parkinsonian rat model associated with depression. Behavioural Brain Research. 2017;316:189-196
  50. 50. Dallé E, Daniels WMU, Mabandla MV. Long-term treatment with fluvoxamine decreases nonmotor symptoms and dopamine depletion in a postnatal stress rat model of Parkinson’s disease. Oxidative Medicine and Cellular Longevity. 2020;20:1-15
  51. 51. Gharghani M. Fluvoxamine-An antidepressant drug with anti-inflammatory and oligodendrogenesis effects in rat model of monophasic multiple sclerosis: Studies in vivo and in vitro. Journal of Neurology and Neurophysiology. 2016;2016:9562
  52. 52. Abanmy N, Alsabhan J, Gard P, Scutt G. Association between the Val66Met polymorphism (rs6265 / G196A) of the BDNF gene and cognitive performance with SSRI use in Arab Alzheimer’s disease patients. Saudi Pharmaceutical Journal. 2021;29(12):1392-1398
  53. 53. Elsaed WM, Alahmadi AM, Al-Ahmadi BT, Taha JA, Tarabishi RM. Gastroprotective and antioxidant effects of fluvoxamine on stress-induced peptic ulcer in rats. Journal of Taibah University Medical Science. 2018;13(5):422-431
  54. 54. Castro ECC, Sen P, Parks WT, Langston C, Galambos C. The role of serotonin transporter in human lung development and in neonatal lung disorders. Canadian Respiratory Journal. 2017;2017:1-10
  55. 55. Meikle C, Creeden J, McCullumsmith C, Worth R. SSRIs : Applications in inflammatory lung disease and implications for COVID-19. Neuropsychopharmacol Reports. 2021;41(3):325-335
  56. 56. Abid S, Houssaini A, Chevarin C, Marcos E, Tissot CM, Gary-Bobo G, et al. Inhibition of gut- and lung-derived serotonin attenuates pulmonary hypertension in mice. American Journal of Physiology. Lung Cellular and Molecular Physiology. 2012;303(6):500-508
  57. 57. Pollok J, JEM VA, Carson-Chahhoud KV. Pharmacological interventions for the treatment of depression in chronic obstructive pulmonary disease. Cochrane Database of Systematic Reviews. 2018;2018:1-53
  58. 58. Cafarella PA, Effing TW, Usmani ZA, Frith PA. Treatments for anxiety and depression in patients with chronic obstructive pulmonary disease: A literature review. Respirology. 2012;17(4):627-638
  59. 59. Mazza MG, Zanardi R, Palladini M, Rovere-Querini P, Benedetti F. Rapid response to selective serotonin reuptake inhibitors in post-COVID depression. European Neuropsychopharmacology. 2022;54:1-6
  60. 60. Schloer S, Brunotte L, Goretzko J, Mecate-Zambrano A, Korthals N, Gerke V, et al. Targeting the endolysosomal host-SARS-CoV-2 interface by clinically licensed functional inhibitors of acid sphingomyelinase (FIASMA) including the antidepressant fluoxetine. Emerging Microbes and Infections. 2020;9(1):2245-2255
  61. 61. Kuykendall J. Fluvoxamine strongly inhibits melatonin metabolism in a patient with low-amplitude melatonin profile. Day Care & Early Education. 2000;20
  62. 62. Friesland M, Mingorance L, Chung J, Chisari FV, Gastaminza P. Sigma-1 receptor regulates early steps of viral RNA replication at the onset of hepatitis C virus infection. Journal of Virology. 2013;87(11):6377-6390
  63. 63. Bagchi P. Endoplasmic reticulum in viral infection. International Review of Cell and Molecular Biology. 2020;350:265-284
  64. 64. Spack NP, Shumer DE, NJN. A SARS-CoV-2 protein interaction map reveals targets for drug repurposing. Physiology & Behavior. 2017;176(12):139-148
  65. 65. Gordon DE, Hiatt J, Bouhaddou M, Rezelj VV, Ulferts S, Braberg H, et al. Comparative host-coronavirus protein interaction networks reveal pan-viral disease mechanisms. Science (80- ). 2020;370:6521
  66. 66. Hashimoto K. Activation of sigma-1 receptor chaperone in the treatment of neuropsychiatric diseases and its clinical implication. Journal of Pharmacological Sciences. 2015;127(1):6-9
  67. 67. Vela JM. Repurposing sigma-1 receptor ligands for COVID-19 therapy? Frontiers in Pharmacology. 2020;11:1-23
  68. 68. Irons J. Fluvoxamine in the treatment of anxiety disorders. Neuropsychiatric Disease and Treatment. 2005;1(4):289-299
  69. 69. Hindmarch I, Hashimoto K. Cognition and depression: The effects of fluvoxamine, a sigma-1 receptor agonist, reconsidered. Human Psychopharmacology: Clinical and Experimental. 2010;25(2):193-200
  70. 70. Iyo M, Shirayama Y, Watanabe H, Fujisaki M, Miyatake R, Fukami G, et al. Fluvoxamine as a sigma-1 receptor agonist improved cognitive impairments in a patient with schizophrenia. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2008;32(4):1072-1073
  71. 71. Ishima T, Fujita Y, Hashimoto K. Interaction of new antidepressants with sigma-1 receptor chaperones and their potentiation of neurite outgrowth in PC12 cells. European Journal of Pharmacology. 2014;15(727):167-173
  72. 72. Alonso G, Phan VL, Guillemain I, Saunier M, Legrand A, Anoal M, et al. Immunocytochemical localization of the sigma1 receptor in the adult rat central nervous system. Neuroscience. 2000;97(1):155-170
  73. 73. Alshammari MA, Alshammari TK, Nenov MN, Scala F, Laezza F. Fibroblast growth factor 14 modulates the neurogenesis of granule neurons in the adult dentate gyrus. PMol Neurobiology. 2016;53(10):7254-7270
  74. 74. Sahay A, Hen R. Adult hippocampal neurogenesis in depression. Nature Neuroscience. 2007;10(9):1110-1115
  75. 75. Moriguchi S, Shinoda Y, Yamamoto Y, Sasaki Y, Miyajima K, Tagashira H, et al. Stimulation of the sigma-1 receptor by DHEA enhances synaptic efficacy and neurogenesis in the hippocampal dentate gyrus of olfactory bulbectomized mice. PLoS One. 2013;8(4):e60863
  76. 76. Ahn H, Weaver M, Lyon D, Choi E, Roger B, Fillingim P. Pridopidine stabilizes mushroom spines in mouse models of Alzheimer’s disease by acting on the sigma-1 receptor. Physiology & Behavior. 2017;176(1):139-148
  77. 77. Duman CH, Duman RS. Spine synapse remodeling in the pathophysiology and treatment of depression. Neuroscience Letters. 2015;5(601):20-29
  78. 78. Castany S, Gris G, Vela JM, Verdú E, Boadas-Vaello P. Critical role of sigma-1 receptors in central neuropathic pain-related behaviours after mild spinal cord injury in mice. Scientific Reports. 2018;8(1):1-13
  79. 79. Mao J, Hu Y, Ruan L, Ji Y, Lou Z. Role of endoplasmic reticulum stress in depression (Review). Molecular Medicine Reports. 2019;20(6):4774-4780
  80. 80. Voronin MV, Vakhitova YV, Seredenin SB. Chaperone Sigma1R and antidepressant effect. International Journal of Molecular Sciences. 2020;21(7088):1-33
  81. 81. Chen YW, Lin PY, Tu KY, Cheng YS, Wu CK, Tseng PT. Significantly lower nerve growth factor levels in patients with major depressive disorder than in healthy subjects: A meta-analysis and systematic review. Neuropsychiatric Disease and Treatment. 2015;11:925-933
  82. 82. De Azevedo CT, Mondin TC, Wiener CD, Marques MB, Fucolo BDÁ, Pinheiro RT, et al. Neurotrophic factors, clinical features and gender differences in depression. Neurochemical Research. 2014;39(8):1571-1578
  83. 83. Angelucci F, Aloe L, Jiménez-Vasquez P, Mathé AA. Lithium treatment alters brain concentrations of nerve growth factor, brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor in a rat model of depression. The International Journal of Neuropsychopharmacology. 2003;6(3):225-231
  84. 84. Song C, Xiang YZ, Manku M. Increased phospholipase A2 activity and inflammatory response but decreased nerve growth factor expression in the olfactory bulbectomized rat model of depression: Effects of chronic ethyl-eicosapentaenoate treatment. The Journal of Neuroscience. 2009;29(1):14-22
  85. 85. Angelucci F, Aloe L, Jiménez-Vasquez P, Mathé AA. Electroconvulsive stimuli alter nerve growth factor but not brain-derived neurotrophic factor concentrations in brains of a rat model of depression. Neuropeptides. 2003;37(1):51-56
  86. 86. Valdes R, Millner LM. Modulation of the Sigma-1 Receptor-IRE1 pathway is beneficial in preclinical models of inflammation and sepsis. Physiology & Behavior. 2019;176(3):139-148
  87. 87. Allahtavakoli M, Jarrott B. Sigma-1 receptor ligand PRE-084 reduced infarct volume, neurological deficits, pro-inflammatory cytokines and enhanced anti-inflammatory cytokines after embolic stroke in rats. Brain Research Bulletin. 2011;85(3-4):219-224
  88. 88. Shen YC, Wang YH, Chou YC, Liou KT, Yen JC, Wang WY, et al. Dimemorfan protects rats against ischemic stroke through activation of sigma-1 receptor-mediated mechanisms by decreasing glutamate accumulation. Journal of Neurochemistry. 2008;104(2):558-572
  89. 89. Hosszu A, Antal Z, Lenart L, Hodrea J, Koszegi S, Balogh DB, et al. Σ1-receptor agonism protects against renal ischemia-reperfusion injury. Journal of American Society of Nephrology. 2017;28(1):152-165
  90. 90. Furuse T, Hashimoto K. Sigma-1 receptor agonist fluvoxamine for delirium in intensive care units: Report of five cases. Annals of General Psychiatry. 2010;9:2-5
  91. 91. Tynan RJ, Weidenhofer J, Hinwood M, Cairns MJ, Day TA, Walker FR. A comparative examination of the anti-inflammatory effects of SSRI and SNRI antidepressants on LPS stimulated microglia. Brain, Behavior, and Immunity. 2012;26(3):469-479
  92. 92. Sacre S, Medghalchi M, Gregory B, Brennan F, Williams R. Fluoxetine and citalopram exhibit potent antiinflammatory activity in human and murine models of rheumatoid arthritis and inhibit toll-like receptors. Arthritis and Rheumatism. 2010;62(3):683-693
  93. 93. Abdo SA, Wadie W, Abdelsalam RM, Khattab MM. Potential anti-inflammatory effect of escitalopram in iodoacetamide-induced colitis in depressed ovariectomized rats: Role of α7-nAChR. Inflammation. 2019;42(6):2056-2064
  94. 94. Obuchowicz E, Bielecka-Wajdman A, Zieliński M, MacHnik G, Gołyszny M, Ludyga T. Imipramine and venlafaxine differentially affect primary glial cultures of prenatally stressed rats. Frontiers in Pharmacology. 2020;10(January):1-17
  95. 95. El Awdan SA. Anti-inflammatory, anti-nociceptive and anti-arthritic effects of antidepressants in rats. Journal of Pharma Care Health System. 2015;4(2):86

Written By

Jawza F. Alsabhan and Tahani K. Alshammari

Submitted: 27 February 2022 Reviewed: 21 April 2022 Published: 04 July 2022