Abstract
Serotonin syndrome (SS) is a potentially fatal adverse drug reaction characterized by an exaggerated increase in serotonergic activity in the central and peripheral nervous systems. It presents a constellation of signs and symptoms related to behavioral changes, neuromuscular excitability, and autonomic instability. These symptoms can occur in both mild and severe forms. SS can be triggered by the therapeutic use of a drug that increases serotonin (5-HT) availability in the synaptic cleft or by the co-administration of two or more drugs that provide this increase. With the escalating use of antidepressants by the world's population, this adverse reaction may be more recurrent. However, SS is often overlooked by patients or not diagnosed by doctors. This review aims to improve awareness about SS and provide a pharmacological perspective to explain its occurrence. Evidence shows that other neurotransmitters may also be involved with the pathology of SS. Furthermore, SS and neuroleptic malignant syndrome (NMS) seem to be part of the same pathological spectrum, especially in atypical NMS cases. The emergence of the syndrome's symptoms may be closely related to pharmacokinetic and/or pharmacodynamic polymorphisms that lead to an increase in the 5-HT available to or 5-HT signaling by specific receptors, thus constituting an important area for future investigations.
Keywords: serotonin syndrome diagnosis, selective serotonin reuptake inhibitor (ssri), antidepressants, diagnostic criteria, genetic polymorphisms, toxidromes, atypical neuroleptic malignant syndrome, neuroleptic malignant syndrome (nms), serotonin toxicity, serotonin syndrome (ss)
Introduction and background
Serotonin syndrome (SS), which also can be referred to as serotonin toxicity or serotonin toxidrome, is an adverse drug reaction characterized by an exaggerated increase in serotonergic activity in the central and peripheral nervous systems (CNS and PNS, respectively) [1,2]. It comprises a constellation of signs and symptoms related to behavioral changes, neuromuscular excitability, and autonomic instability. These symptoms can occur in mild, severe, and potentially fatal forms [1]. This syndrome can occur with the therapeutic use of a drug that increases the availability of serotonin (5-hydroxytryptamine; 5-HT) in the synaptic cleft, usually an antidepressant of the selective 5-HT reuptake inhibitor (SSRI) class, or by the interaction of two or more drugs that provide this 5-HT increase [1,3].
Due to the lack of medical awareness about SS, its true incidence is unknown and difficult to measure [4-6]. In 2017, the "Toxic Exposure Surveillance System" reported 57,254 cases of exposure to SSRIs in the US (of which 24,819 or 43.3% of cases were single exposures), with 5 or 0.009% of these cases culminating in death [7]. According to the World Health Organization [8], the global number of people with depression increased by 18.4% from 2005 to 2015. Since the effectiveness of antidepressant drugs is considered suboptimal, associated with the slow development of new drugs in this class, knowing as much as possible about all aspects of existing drugs becomes paramount [9].
Another current aggravating factor, which further increases the need for attention and consciousness about SS, is the worsening mental health of the population due to the COVID-19 pandemic. In a cohort study, which used information obtained from the US Veterans Health Administration database, three groups of patients were compared: (1) patients who had COVID-19; (2) patients who did not test positive for the disease during the same period; (3) control group with patients who had influenza in the pre-pandemic period. The results suggest that the first group had a greater risk of developing various mental health disorders such as anxiety, depression, opioid and other substance use, sleep, and stress disorders [10]. In Brazil, the cohort study "Brazilian Longitudinal Health Study" (ELSA-Brasil) investigated the psychopathological changes generated by COVID-19 in 2,011 patients (those who were from the beginning of the study in 2008 and who remained until the year 2020). The results suggested that the level of depressive disorder remained stable during the pandemic, but this level was already considered very high, affecting about 30% of the current population [11]. Indeed, there is evidence of increased prescription and consumption of antidepressants, especially SSRIs, in several countries in Europe and North and Latin America [12].
Given the lack of medical awareness about the syndrome associated with the increased use of antidepressants with several drug interactions that can trigger it, this review aimed to improve the current understanding of SS occurrence using a pharmacological mechanistic approach. Specifically, we analyzed the underlying pharmacological mechanisms that contribute to the development of this syndrome and the drug interactions that can lead to it. Through this approach, we hope to provide new insights to identify and prevent this life-threatening condition.
Review
History of serotonin syndrome
The discovery of the antidepressant properties of drugs like iproniazid and imipramine in the 1950s transformed the treatment of depression and anxiety. Iproniazid, initially developed for tuberculosis, was found to inhibit the enzyme monoamine oxidase (MAO), and this inhibition was believed to be responsible for its antidepressant effects [13]. Similarly, imipramine, which was originally being investigated as an antipsychotic drug, was found to have significant antidepressant properties [14].
Oates and Sjoerdsma, in 1960, were the first to relate specific clinical manifestations to increased 5-HT levels in the CNS. They observed a syndrome characterized by nervousness, diaphoresis, mental dysfunction, ataxia, and hyperreflexia in patients who were taking an MAO inhibitor (MAOI) while on antihypertensive treatment with L-tryptophan. This syndrome was later named the "Indolamine Syndrome" [15]. In 1975, researchers observed abnormal behavior in male rats treated with serotonergic agonists, which they called the "5-HT Behavioral Syndrome" [16]. The term "Serotonin Syndrome" or "5-HT Syndrome" was coined by Gerson and Baldessarini in 1980, in their review of the clinical features and etiology of the syndrome [17].
Serotonin system
5-HT is a monoamine present in most organic systems, but its best-known action is as a neurotransmitter molecule in the CNS [18]. It is responsible for regulating neurobehavioral processes such as attention, sleep-wake, appetite [19], affective and sexual behaviors [20], thermoregulation [21], aggressive behavior [22], nociception, emesis, and motor tone [18]. In the periphery, 5-HT is produced by enterochromaffin cells distributed throughout the gastrointestinal (GI) tract [23,24]. It is estimated that more than 90% of circulating 5-HT is produced by these cells [24]. Figure 1 illustrates the process of serotonin synthesis in detail.
Figure 1. 5-HT synthesis and degradation.
The cell bodies of serotonergic neurons are in the raphe nuclei in the brainstem, and their axons propagate and extend widely throughout the brain and spinal cord. 5-HT biosynthesis occurs in these cell bodies and begins with the passage of the essential amino acid L-tryptophan across the blood-brain barrier via a non-tryptophan-selective amino acid transporter [25]. The first step and the rate-limiting factor in 5-HT synthesis is the hydroxylation of L-tryptophan to 5-hydroxytryptophan (5-HTP) by the enzyme tryptophan hydroxylase (found only in serotonergic neurons). Then, decarboxylation occurs by the enzyme L-aromatic amino acid decarboxylase, generating the final compound 5-hydroxytryptamine [25]. After synthesis, 5-HT is rapidly transported to storage vesicles via a vesicular monoamine transporter (VMAT) isoform 2 [25]. There are two isoforms of VMAT: 1 and 2. VMAT1 is mostly in neuroendocrine cells, and VMAT2 is in monoaminergic neurons of the CNS and PNS [26]. Free 5-HT in the synaptic cleft is transported back to the presynaptic neuron via the 5-HT reuptake transporter (SERT; 5-HTT). The MAO enzyme degrades 5-HT. There are two isoforms of MAO (A and B); MAO-A more selectively degrades 5-HT, while MAO-B nonspecifically degrades catecholamines [27].
Currently, 14 5-HT receptors have already been cloned and are classified into seven families of receptors (5-HT1 to 5-HT7) with different subtypes [28]. Table 1 describes the distribution and effects of each 5-HT receptor.
Table 1. 5-HT receptors and their distribution and effects.
| Receptor Family | Receptor Subtypes | Signaling Mechanisms | Distribution | Effects |
| 5-HT1 | 5-HT1A 5-HT1B 5-HT1D 5-HT1e 5-HT1F | G-protein coupled - Inhibit adenylate cyclase | Prefrontal cortex [28], raphe nuclei (autoreceptors), and the terminal regions as postsynaptic sites (mainly in cortico-limbic areas) [29] | 5-HT1A: anxious behavior [18] 5-HT1B/D: anti-migraine properties and an antiplatelet effect [18,30] |
| 5-HT2 | 5-HT2A 5-HT2B 5-HT2C | G-protein coupled - Activates phospholipase C | 5-HT2A: frontal cortex, nucleus accumbens, midbrain nuclei, hippocampus [31], smooth muscle, and platelets [18] | 5-HT2A: induces hallucinations (in high 5-HT concentration) in the prefrontal cortex [28]; the antagonism is an established treatment for schizophrenia [31,32] |
| 5-HT3 | 5-HT3A (single functional isoform) | Non-selective cation channel | Dorsal vagal complex, hippocampus, frontal cortex, cingulated cortex, ventral tegmental area, and immune cells [28] | 5-HT3A antagonism has an antiemetic effect [23] and may decrease the behavioral consequences of withdrawal syndromes caused by the treatment of drugs of abuse and for several psychiatric disorders (e.g., psychoses, anxiety, and cognitive dysfunction) [28,33] |
| 5-HT4 | 5-HT4A 5-HT4B 5-HT4C 5-HT4D | G-protein coupled - Activates adenylate cyclase | 5-HT4A-B-C are found in atrium, brain and intestine; 5-HT4A in bladder; 5-HT4B in the kidneys; and 5-HT4D in intestinal cells [34] | Modulate the motility and secretory response of the GI tract [23,28] |
| 5-HT5 | 5-HT5A 5-HT5B | G-protein coupled - Inhibit adenylate cyclase | The prefrontal cortex, hippocampus, amygdala, cerebellum, striatum, and substantia nigra [28] | 5-HT5A receptors have a distribution that could theoretically modulate emotion regulation, cognition, and nociception; however, due to the lack of selective substances for these receptors, none of these functions could be tested [28] |
| 5-HT6 | G-protein coupled - Activates adenylate cyclase | Hypothalamus, hippocampus, and cerebral cortex [28] | Some antidepressants and antipsychotics have an affinity for these receptors. Still, they also show selectivity for other targets, making it difficult to define the functions of this family of receptors [28] | |
| 5-HT7 | G-protein coupled - Activates adenylate cyclase | Hypothalamus, thalamus, and hippocampus. Smooth muscle cells, blood vessels, and gastrointestinal tract [28] | Seems to have a pro-depressive effect. Some preclinical studies in animal models using an unspecific antagonist suggest a significant role in depression [35] |
Pathophysiology of serotonin syndrome
Six possible mechanisms for the occurrence of SS are listed below and illustrated in Figure 2.
Figure 2. Possible mechanisms for the occurrence of serotonin syndrome.
1 - Increased levels of the 5-HT precursor, L-tryptophan; 2- Inhibition of monoamine oxidase; 3- Increased release of 5-HT by drugs such as amphetamine, cocaine, MDMA and levodopa; 4- Activation of the 5-HT1A receptor directly or indirectly; 5- 5-HT2A receptor antagonism; 6 – Inhibition of serotonin reuptake by SERT blockade. Adapted from [36], [37] and [38].
Increased Availability of the Essential Amino Acid L-Tryptophan
Theoretically, an increased supply can lead to an increment in endogenous 5-HT. The study that investigated this risk factor used patients who also used MAOIs, which are drugs that make up the second mechanism for the occurrence of SS [15]. Thus, an interaction between these factors leading to SS cannot be excluded.
Inhibition of MAO
By inhibiting MAO, 5-HT is once again stored in the vesicles of the presynaptic neuron and subsequently released into the synaptic cleft to perform its function, which allows for a longer duration of action [36].
Promotion of 5-HT Release
Some illicit substances, such as amphetamine, 3,4-methylenedioxymethamphetamine (MDMA), and cocaine can promote the release of 5-HT into the synaptic cleft and thus lead to syndrome onset [39]. It has also been described that levodopa can be decarboxylated into dopamine (DA) in serotonergic neurons. This newly synthesized DA competes with 5-HT for VMAT, after which it is released into the synaptic cleft along with 5-HT. Furthermore, this DA can hinder the entry or exit of 5-HT through the SERT [40,41].
Postsynaptic Receptors
The fourth and fifth mechanisms depend on the interaction of 5-HT and/or agonist or antagonist drugs with their postsynaptic receptors. They can also occur through increased availability of 5-HT in the cleft (also drug-mediated) [36,38].
SERT Blockade
Another way to prolong the action of 5-HT in the synaptic cleft is to inhibit its reuptake into the presynaptic neuron by blocking SERT [36].
5-HT receptors involved in serotonin syndrome
The symptoms that characterize SS can be mainly related to the agonism and/or antagonism of two subtypes of receptors: 5-HT1A and 5-HT2A [36,39]. There is no single receptor responsible. The 5-HT1A receptors seem to contribute to symptoms such as myoclonus, hyperreflexia, vasomotor tone alterations, anxiety, increased respiratory rate, and hyperactivity [1,39]. This receptor has been associated with several behaviors consistent with SS in rats and mice, such as Straub's tail, walking backward, stepping with forepaws, head shaking, tremors, and hindlimb abduction [42]. Accordingly, these behaviors are more apparent in mice overexpressing 5-HT1A receptors [43].
Another receptor that appears closely related to the syndrome is the 5-HT2A receptor. The appearance of the most severe SS symptoms may be related to the agonism of these receptors, as its stimulation can cause platelet aggregation, bronchoconstriction, vasoconstriction (and consequent hypertension), and the behavioral and cognitive changes characteristic of the syndrome [1,37,44]. This receptor has also been linked to SS-associated hyperthermia in animal models. Administration of the antipsychotic olanzapine (5-HT2A receptor antagonist) in an SS animal model (Wistar rats treated with fluoxetine 10 mg/kg + tranylcypromine 3.5 mg/kg) prevented the onset of hyperthermia, which supports the hypothesis of the involvement of these receptors on body temperature [45].
On the other hand, the antagonism of 5-HT2A receptors also seems to be associated with SS onset [46]. A 2018 study explored this topic with a bioinformatics approach, using the FDA Adverse Reactions Reporting System (FAERS) and pharmacokinetic data from the literature. They reported that when associated with SSRIs, second-generation antipsychotics exert an effect that may reflect 5-HT2A receptors antagonism with simultaneous activation of 5-HT1A receptors, which might be an SS onset mechanism for these two classes of drugs [38].
The two receptors, 5-HT1A and 5-HT2A, are extensively co-expressed in the prefrontal cortex (PFC) [28,29,47]. When SERT is inhibited, there is an increase in 5-HT in this brain area. Increased 5-HT will consequently elicit an excitatory effect through 5-HT2A receptor activation and an inhibitory effect caused by 5-HT1A receptor activation in pyramidal neurons, which project to other cortical areas and subcortical regions such as the limbic system [29,47]. By adding 5-HT2A receptor antagonists in this scenario, there will be an increase in neurotransmission mediated by postsynaptic 5-HT1A receptors, therefore modifying the control made by the PFC in the corticolimbic networks [29,47,48].
Moreover, the antagonism of 5-HT2A receptors is suggested to be implicated in the increase of serotonergic neurotransmission by inhibiting GABAergic interneurons in the dorsal raphe nucleus [49]. As shown in Figure 3, the GABAergic interneurons located in the dorsal nucleus of the raphe and periaqueductal gray matter are stimulated by 5-HT through 5-HT2A/2C receptors and inhibited by 5-HT1A receptors also present there. This activation promotes the release of GABA, which inhibits subsequent serotonergic neuron stimulation, blocking 5-HT release. When administered with a 5-HT2A/2C receptor antagonist, 5-HT will bind only to 5-HT1A receptors. This binding promotes the inhibition of GABAergic interneurons, which will not release GABA, and, consequently, will promote the release of 5-HT by serotonergic neurons [49].
Figure 3. Schematic representation of the negative feedback in the GABA and 5-HT circuit.
Adapted from [49]. Created with Biorender.com
A SS animal model, using substances with a strong affinity for 5-HT1A receptors like 8-OH-DPAT and gepirone, was developed utilizing male Sprague-Dawley rats (termed "behavioral syndrome" in rodents) [50]. After inducing the behavioral syndrome, these animals were treated with nonspecific 5-HT2/5-HT1C receptor antagonists (ritanserin, ICI 170,809, and ketanserin), and it was observed that the blockade caused by these agents increased the behavioral syndrome [50]. The authors suggest that 5-HT exerts an inhibitory effect on 5-HT1A receptor functions through 5-HT2 receptors or possibly through 5-HT1C receptors. Thus, blocking these receptors may release the 5-HT1A receptors from the inhibitory influence [50].
The activation of 5-HT3 receptors is related to SS symptoms such as diarrhea, nausea, and abdominal pain. Thus, 5-HT3 receptor antagonists have clinical relevance in relieving nausea and emesis, mainly those resulting from cancer treatment, as mentioned above. Some studies have reported SS cases associated with these antagonists, but the exact mechanism has yet to be elucidated [51].
Occurrences of serotonin syndrome
It is known that SS can occur by an overdose of a drug that increases 5-HT availability or because of a drug interaction between two or more drugs at therapeutic doses (more common in clinical practice). Generally, in SS cases caused by a single drug, the symptoms are moderate, and there is a better prognosis. In contrast, when SS is triggered by drug interactions, mainly by MAOIs and SSRIs, the symptoms are more severe and serious and can lead to death [44,52].
Other neurotransmitters may also be involved in developing the severe form of SS [37]. For example, 5-HT has been shown to cause direct stimulation of neurotransmitter release (e.g., GABA, glutamate, and DA) by the dorsal raphe nuclei [25,37]. It is also suggested to influence noradrenaline (NA) release by the hypothalamus in animals [53]. Indeed, previous studies showed that SS causes a hyperadrenergic state that culminates in physiological stress [54,55]. In this sense, some case reports consider SS an inducer of Takotsubo syndrome [54-56], characterized by transient left ventricular dysfunction caused by physical and/or emotional stress, without the individual having previous heart disease [56]. Additionally, NA is known for its effect on blood pressure, in addition to causing tachycardia, tachypnea, tremor, and agitation [57].
Drugs associated with serotonin syndrome
According to the data found in the literature, the drugs capable of developing SS alone are all SSRIs and nefazodone [2,6,57]. On the other hand, 5-HT3 and 5-HT2A receptor antagonists and opioids can cause SS when used concomitantly with any antidepressant. Drugs, such as lithium and methylphenidate, do not have confirmed affinity or activity in any of the serotonergic receptors, yet several case reports associate their use with SS [58,59]. These drugs appear to increase 5-HT release and postsynaptic 5-HT1A receptor sensitivity [60,61]. Table 2 describes the drugs associated with SS according to their mechanisms.
Table 2. Drugs associated with SS according to their mechanisms.
*Lithium - as explained earlier in the text, it does not seem to have proven action on serotonergic receptors. Adapted from [2,37,38,57].
| Mechanism | Drugs associated with SS |
| Increased 5-HT synthesis | Food supplement: tryptophan |
| Inhibition of 5-HT metabolism | MAOIs: selegiline, rasagiline, tranylcypromine, linezolid (antibiotic), methylene blue |
| Increased 5-HT release | Amphetamine and derivatives: amphetamine, methylphenidate, cocaine, MDMA. Lithium* |
| 5-HT1 receptor agonism | Triptans: sumatriptan, naratriptan, rizatriptan, Opioids: fentanyl, meperidine, sufentanil. Antidepressants and mood stabilizers: mirtazapine, trazodone, lithium* |
| 5-HT2A receptor antagonism | Second-generation antipsychotics: quetiapine, risperidone, olanzapine, clozapine, aripiprazole, and ziprasidone Antidepressant: nefazodone |
| Inhibition of 5-HT reuptake (SERT) from the synaptic cleft | SSRI: citalopram, escitalopram, fluoxetine, fluvoxamine, paroxetine, sertraline 5-HT and NA reuptake inhibitors (SNRIs): venlafaxine, duloxetine, desvenlafaxine Tricyclic antidepressants (TCA): amitriptyline, clomipramine, desipramine, doxepin, imipramine, nortriptyline Multimodal antidepressant: vortioxetine. Opioids: meperidine, methadone, tramadol, codeine, oxycodone, morphine 5-HT3 receptor antagonists: ondansetron and granisetron. Antidepressants and other classes: trazodone, nefazodone, and cyclobenzaprine. |
Drugs associated with SS interact with several receptors which go beyond their desired initial interaction. Culbertson et al. (2018) demonstrated additional targets than those described in the technical information leaflets using a bioinformatics approach. They show how these interactions are diverse and with different levels of affinities. For example, naratriptan is a high affinity 5-HT1 receptor subfamily agonist, with a moderate affinity towards 5-HT2A, 5-HT2C, 5-HT3, and 5-HT7 receptors, with no evidence of activity on these receptors being as agonists or antagonists. In addition, this drug has also been shown to have a moderate affinity for SERT, managing to exert a critical inhibitory activity [57]. This study also showed that the additional antagonistic activity of some drugs on muscarinic acetylcholine receptors might result in more SS cases than with drugs inhibiting NET and SERT alone [57].
Clinical conditions associated with serotonin syndrome
The risk of developing SS appears to be associated with kidney diseases that may predispose the patient due to impaired drug clearance, which circulates longer in the individual's body [37]. Some studies have shown that there is an increase in circulating 5-HT in patients with cardiovascular diseases such as hypertension [62,63], thrombosis [44,64], myocardial infarction, and coronary atherosclerosis [64,65], and in patients who use tobacco [66].
Hepatic metabolism plays an essential role in developing several drug interactions. Cytochrome P450 (CYP450) is a family of oxidative isoenzymes found in the microsomes of various tissues (e.g., liver and intestines) responsible for metabolizing several drugs. The drugs involved in SS are mainly metabolized by the isoenzymes CYP2C19, CYP2D6, and CYP3A4 [67]. Some factors, such as age, sex, and some diseases, contribute to natural variations in the metabolism performed by these isoenzymes. Particular attention should be paid to elderly patients, who should have SS as a differential diagnosis for cases of mental status changes [67].
Interestingly, changes in the pharmacokinetic parameters of SSRIs leading to adverse reactions have been associated with polymorphisms in CYP2D6 and CYP2C19 [9,68,69], mainly in slow metabolizers and intermediate metabolizers, as shown by several case reports [70-73]. A study with 100 patients treated with only venlafaxine (orally), within the recommended doses for major depressive disorder, identified 25 with polymorphisms in CYP2D6 (at least one defective allele) responsible for most of the metabolism of venlafaxine. Four patients were classified as poor metabolizers, with significantly slower venlafaxine metabolization than normal or fast metabolizers. These individuals also had more adverse reactions than the others, showing that poor metabolizers have a greater risk of having adverse reactions to venlafaxine at normal doses [74].
In a recent systematic review and meta-analysis carried out with data from 8,379 individuals genotyped for CYP2D6 and CYP2C19, using any of the 14 drugs analyzed (antipsychotics and antidepressants), it was possible to observe that patients with polymorphisms in CYP2D6 had more adverse effects when administered aripiprazole, haloperidol, risperidone, nortriptyline, paroxetine, quetiapine, amitriptyline, mirtazapine, or fluvoxamine. Additionally, patients with CYP2C19 polymorphisms exhibited more effects from escitalopram, sertraline, clozapine, fluoxetine, and venlafaxine [9]. These polymorphisms could explain why many patients can take several serotonergic drugs without experiencing adverse reactions.
Another interesting variable is 5-HT2A receptor polymorphisms found in some individuals [75]. Genetic variations in this receptor can alter the membrane surface expression, signal transduction, and ligand affinity [76]. One example is the T102C polymorphism located in the first exon of the 5HTR2A gene. The thymine (T) for cytosine (C) substitution does not significantly alter the amino acid sequence (i.e., silent polymorphism); however, it is positioned close to the promoter region and could affect gene and/or protein expression [77]. Indeed, a study with human post-mortem tissue observed that the presence of the C allele reduced the production of 5-HT2A receptors by 20% in the temporal cortex [78]. Some authors suggest a correlation between homozygous individuals for the C102 allele of the 5-HTR2A gene and a greater risk of developing adverse reactions related to 5-HT [79,80].
However, in a study carried out with 95 patients who overdosed on various antidepressant classes, a correlation between polymorphisms in the C102 allele of the 5-HTR2A gene with a higher risk of developing SS was not observed [75]. It is important to emphasize that only Hunter's criteria were used to diagnose SS in those patients, limiting the diagnosis of the syndrome to only 14 participants. Using this stricter criterion (see the detailed description of diagnostic classifications below) may have masked the result obtained. For example, 61 participants had hyperreflexia without concomitant tremors, which considers these patients "without SS". Furthermore, the 13 participants classified as "without SS" had inducible clonus without simultaneous agitation, diaphoresis, or hyperthermia [75].
These studies pointing to correlations between SS and polymorphisms in CYPs, or 5-HT2A receptors, suggest that more than one polymorphism is related to a higher risk of developing SS symptoms [76].
Diagnosis
As shown in Figure 4, the symptoms can range from mild to severe, with potentially fatal forms. Mild symptoms are flu-like and easily ignored.
Figure 4. Signs and symptoms of SS within the severity spectrum.
Starting with mild symptoms (yellow), going through moderate symptoms (orange), and reaching severe symptoms (red). Adapted from [37] and [81].
As it is a purely clinical diagnosis syndrome, it is essential to have a diagnostic algorithm. Currently, three diagnostic classification systems are available: I) the Sternbach Criteria, II) the Radomski Criteria, and III). the Hunter Serotonin Toxicity Criteria. Each system tries to list the characteristic SS symptoms [5].
Sternbach proposed the first diagnostic criteria in 1991. These were based on 3 or more most described symptoms in 38 SS cases [82] presented in Table 3. However, these symptoms (e.g., confusion, agitation, and fever) can be observed in several other clinical conditions, which may indicate a false positive for SS [3,81].
Table 3. Sternbach criteria.
Adapted from [82].
| Sternbach Criteria | |
| Signs and Symptoms (3 or more) | Confusion or hypomania |
| Agitation | |
| Myoclonus | |
| Hyperreflexia | |
| Diaphoresis | |
| Tremor | |
| Shivering | |
| Fever | |
| Incoordination | |
| Diarrhea | |
| Inclusion Criteria | Coincidence with the addition or increased dose of a known serotonergic agent within an established therapeutic regimen |
| Exclusion of other etiology (e.g., infection, substance abuse, etc.) | |
| A neuroleptic drug was not started, or at an increased dose, before the onset of the signs and symptoms | |
The second classification system was developed by Radomski et al. (2000), through the review of 24 additional cases to those analyzed by Sternbach, from 1991 to 1995. In this classification, the cases were divided into I) a moderate state of problems related to 5-HT, II) SS, and III) a toxic state [83]. However, this classification tool, like Sternbach's, also generates false positive results because it excludes symptoms specific to SS.
The Hunter Serotonin Toxicity Criteria are based on data collected from a regional toxicology unit called the Hunter Area Toxicology Service (HATS), which serves the entire Hunter Division, Australia population. All patients admitted to HATS from January 1987 to November 2002 and diagnosed with an overdose of only one serotonergic drug were included in the study (n=473 cases) [3]. As exemplified in Figure 5, based on the statistically significant symptoms presented by these patients, a diagnostic algorithm was produced as a flowchart. This algorithm has improved sensitivity (69%) and specificity (97%) compared to the Sternbach criteria [3,5].
Figure 5: Hunter serotonin toxicity criteria
Adapted from [3], [37] and [39].
However, despite the apparent superiority of the Hunter criteria, the gold standard for SS diagnosis, there is no official consensus on which criterion should be used since the algorithm was developed from cases of overdose by a serotonergic agent, and this may make it less appropriate for diagnosing cases in which there is no overdose [5,84]. The work of Culbertson et al. (2018) suggests that the lower specificity of the Sternbach diagnostic criterion may be adequate for detecting mild to moderate SS cases and that the Hunter criterion is more appropriate for severe SS manifestations [57].
Differential diagnosis
Clinical diagnosis of SS can be challenging due to comparison with other syndromes (Table 4) and infections (such as meningitis and encephalitis [1]), that have very similar symptoms, which makes it difficult to distinguish between them. Moreover, changes in mental status can present symptoms that mirror those observed in alcohol and drug withdrawal [4,81].
Table 4. Comparison between SS and other syndromes.
| Syndrome | Causative Agent | Vital Signs | Mental Status | Other clinical symptoms |
| Serotonergic Syndrome | Serotonergic Drugs | Hyperthermia (>40°C), tachycardia, hypertension, and tachypnea | Delirium, agitation, and coma | Neuromuscular hyperactivity (tremor, myoclonus, hyperreflexia, clonus), diaphoresis, and hyperactive bowel sounds |
| Neuroleptic Malignant Syndrome | Dopamine antagonists and the absence of dopamine | Hyperthermia (>41.1°C), tachycardia, hypertension, and tachypnea | Delirium, agitation | Neuromuscular hypoactivity (rigidity and bradykinesia), hypoactive bowel sounds |
| Anticholinergic Syndrome | Anticholinergic agents | Hyperthermia (<38.8°C), tachycardia, hypertension (mild), and tachypnea | Hypervigilance, agitation, hallucination, delirium with grumbling, and coma | Normal muscle tone and reflexes, urinary retention, hypoactive bowel sounds, dry skin and mucous membranes |
| Malignant Hyperthermia | Inhalational anesthetics and depolarizing neuromuscular blockers (succinylcholine) | Hyperthermia (may be greater than 43°C), tachycardia, hypertension, and tachypnea | Agitation | Rigor-mortis-like rigidity, hyporeflexia, skin with signs of flushing and cyanosis, hypoactive bowel sounds |
Table 4, which compares the syndromes and clinical conditions that may generate more uncertainties at the time of diagnosis, demonstrates that hyperreflexia and clonus are unique SS symptoms, as advocated by Hunter's criterion, a possible differential. However, not all patients will present these symptoms, which may be related to 5-HT1A receptor agonism [5,37]. About 50% of patients have symptoms related to augmented neuromuscular activity, such as tremors, hyperreflexia, hypertonia, myoclonus, and ataxia [85]. Symptoms related to autonomic instability are present in 40% of patients and include tachycardia, tachypnea, diarrhea, diaphoresis, and increased body temperature [3]. Ignoring a possible case of SS because the symptoms do not meet the Hunter criteria can be a harmful and potentially less safe decision for the patient [5,84].
Serotonin syndrome versus neuroleptic malignant syndrome
Neuroleptic malignant syndrome (NMS) is a significant confounding factor for SS diagnosis [1,5,86]. This syndrome is caused by an adverse reaction to excessive DA D2 receptor blockade or genetically reduced function of this receptor [87]. According to the Diagnostic and Statistical Manual of Mental Disorders (DSM-5), NMS incidence in patients using antipsychotics varies between 0.07% and 1.4% [87]. Despite being a rare syndrome, NMS is better known and present in physicians' memory than SS, which could be because NMS is described in DSM-5, and SS, unfortunately, is not.
A vital factor in differentiating the syndromes is the class of drug that initiated the symptoms. While in SS, they would be serotonergic agents, in NMS, they are antipsychotics [86,88]. However, since second-generation antipsychotics (atypical) act by blocking DA D2 receptors and serotonergic 5-HT2A receptors (and some of them also can act as 5-HT1A agonists [89]), this differentiation factor may be outdated.
Symptoms are also used to guide the differential diagnosis of the two syndromes; for example: in NMS, patients may have stiffness, rhabdomyolysis (characterized by muscle fiber destruction), and increased serum creatine phosphokinase (CPK) levels, which should not occur in SS [90]. However, some SS case reports show that patients present rigidity, sometimes associated with hyperreflexia [91-94] and increased serum CPK levels [92,95,96]. In the meta-analysis carried out by Werneke et al. (2016), with all cases of SS published in the PubMed databases and Thomson Reuter's Web of Science from 2004 to 2014, they observed that in the 299 cases of SS included in the study, 45.4% (136 cases) had stiffness, and 14% had rhabdomyolysis (42 cases) [5].
Another critical point for differentiating the syndromes is the speed with which the symptoms appear after introducing the new drug to the therapy. Many authors support that SS symptoms appear in less than 24 hours and that in NMS, symptoms present between 24 and 72 hours [1,86]. However, some studies have demonstrated that this no longer seems accurate since many patients may present new SS-related symptoms after a week of the new therapy [5,91,95,97].
It is known that MAOIs have a very potent effect due to their mechanism of action that inhibits the main pathway of 5-HT metabolism. Thus, some authors argue that the interaction between an MAOI and an SSRI is more likely to cause more severe and even fatal cases of SS [2,97]. Fox et al. (2007) demonstrated that administering tranylcypromine and 5-HTP (or clorgyline and 5-HTP) was necessary for SS symptoms to appear in mice with normal SERT functioning. In cases where only MAOIs were administered, only SERT knockout mice presented SS symptoms [98]. Fox et al. (2007) demonstrated that administering tranylcypromine and 5-HTP (or clorgyline and 5-HTP) was necessary for SS symptoms to appear in mice with normal SERT functioning [98]. It is important to note that central SS studies, still used as a reference today, were carried out when MAOIs were more frequently prescribed [1,39,44,82].
Therefore, this delay in the appearance of SS symptoms may be due to the change in the pharmacological profile of drugs currently prescribed. This result may modify that well-established temporal parameter of the fastest initiation of SS symptoms. In addition, Werneke et al. (2016) demonstrated that the dose and speed of drug distribution might cause a more rapid or slower syndrome onset [5].
Given all these similarities and difficulties in differentiating these two syndromes, some authors consider them part of the same pathological spectrum [5,99]. Interestingly, some case reports reported a different presentation of NMS (atypical NMS), which does not necessarily present rigidity or hyperthermia as primary symptoms [100,101]. These cases always occur in the presence of atypical antipsychotics. One of the most widely accepted hypotheses about this "atypical" NMS pathophysiology is that, due to the action of these drugs on the serotonergic system, in addition to the lower degree of dopaminergic receptors antagonism, the motor symptoms could be attenuated. In addition, adrenergic and muscarinic receptor activity disruption can lead to autonomic dysfunction [100-102]. Therefore, SS and NMS may be part of the same pathological spectrum since the atypical NMS seems to occur due to an augmented serotonergic action. Indeed, as previously mentioned, some SS cases present NMS symptoms.
Treatment and prevention
The first and foremost treatment of the syndrome is removing the causative serotonergic agent. The symptoms of SS disappear spontaneously in 70% of the cases, with only the withdrawal of the drug(s) involved. The level of additional support will be given depending on the severity of the patient's symptoms; in general, 40% of patients end up needing intensive care, with 25% requiring drastic measures such as orotracheal intubation [44,84].
In addition to withdrawal from serotonergic agents, some mild cases also require benzodiazepines to improve akathisia and anxiety. Benzodiazepines can also be used in cases where the patient has myoclonus, muscle rigidity, and seizures. Beta-blockers (e.g., propranolol and pindolol) also block 5-HT1A receptors, which would be beneficial in moderate cases with hypertension [36,84]. However, animals treated with propranolol did not have an adequate response to SS [103].
In the most severe SS cases, often considered a medical emergency, some authors advocate administering cyproheptadine (a non-selective 5-HT1A and 5-HT2A receptor antagonist) if benzodiazepines fail to control symptoms [1,37,104]. This treatment neutralizes excess 5-HT in the synaptic cleft [105]. However, some researchers point out that there is no significant difference in the outcome of treatment with cyproheptadine compared to supportive therapy without this antagonist [36,38,95].
In patients with elevated body temperature caused by SS, cooling measures and reducing muscular activity is an effective treatment strategy [1,104]. This is because the temperature increase is not regulated by the hypothalamic thermostat, but rather by hyperexcitability and direct serotoninergic effects on the muscle [106]. As a result, antipyretics are not recommended for this condition [1,104,106]. However, cyproheptadine may be helpful in these cases as it targets the 5-HT2A receptors, which have been implicated in hyperthermia in animal models [103].
While most cases resolve spontaneously, there are commercially available extended-release formulations releasing the drug associated with SS slowly over time, which may change this estimate. In addition, it is always necessary to consider these drugs' half-life and whether their metabolites have pharmacological effects [37].
The main form of SS prevention is knowledge of its possible causative agents and symptoms by the entire care team, physicians, and patients [1,4,52]. In 1999, Mackay, Dunn, and Mann reported that 85% of physicians across England who prescribed nefazodone within one year (1996-1997) were unfamiliar with SS as a clinical diagnosis [6]. In 2009, a lack of medical awareness was also observed, where seven SS cases were initially misdiagnosed (due to the symptoms presented) in the emergency department of an Israeli hospital. Later, the correct diagnosis was made when the patients showed more classic and severe SS symptoms, including clonus and hyperreflexia [4]. This case demonstrates how unawareness can be harmful because if professionals are not attentive to SS and do not relate the symptoms with the syndrome, the patient will not be treated appropriately. Sometimes, mild and moderate cases will naturally evolve, increasing the syndrome's severity.
Conclusions
SS is commonly associated with patients receiving treatment for psychiatric disorders. However, since antidepressants are also used to treat other medical conditions, all physicians (not only psychiatrists and neurologists) who prescribe drugs that can induce SS should be educated about this syndrome. Moreover, providing education to patients who are starting antidepressant treatment about the potential adverse reactions and the risk of developing SS is crucial.
The appearance of the syndrome's symptoms in some patients, despite others with very similar conditions, leads us to consider that polymorphisms are the risk factors favoring the increase of 5-HT available in the synaptic cleft. Future studies investigating polymorphisms in critical molecules for the kinetics and/or targets (CYPs, 5-HT receptors, SERT, and MAO) of SS-inducing drugs will yield a greater understanding of the syndrome.
Acknowledgments
The Sao Paulo Research Foundation (FAPESP) grant # 17/06100-8 to SC
The authors have declared that no competing interests exist.
References
- 1.The serotonin syndrome. Boyer EW, Shannon M. N Engl J Med. 2005;352:1112–1120. doi: 10.1056/NEJMra041867. [DOI] [PubMed] [Google Scholar]
- 2.Serotonin syndrome. Buckley NA, Dawson AH, Isbister GK. BMJ. 2014;348:0. doi: 10.1136/bmj.g1626. [DOI] [PubMed] [Google Scholar]
- 3.The Hunter Serotonin Toxicity Criteria: simple and accurate diagnostic decision rules for serotonin toxicity. Dunkley EJ, Isbister GK, Sibbritt D, Dawson AH, Whyte IM. QJM. 2003;96:635–642. doi: 10.1093/qjmed/hcg109. [DOI] [PubMed] [Google Scholar]
- 4.The serotonin syndrome: initial misdiagnosis. Attar-Herzberg D, Apel A, Gang N, Dvir D, Mayan H. https://pubmed.ncbi.nlm.nih.gov/19697589/ Isr Med Assoc J. 2009;11:367–370. [PubMed] [Google Scholar]
- 5.Conundrums in neurology: diagnosing serotonin syndrome - a meta-analysis of cases. Werneke U, Jamshidi F, Taylor DM, Ott M. BMC Neurol. 2016;16:97. doi: 10.1186/s12883-016-0616-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Antidepressants and the serotonin syndrome in general practice. Mackay FJ, Dunn NR, Mann RD. https://pubmed.ncbi.nlm.nih.gov/10818650/ Br J Gen Pract. 1999;49:871–874. [PMC free article] [PubMed] [Google Scholar]
- 7.2017 Annual Report of the American Association of Poison Control Centers' National Poison Data System (NPDS): 35th Annual Report. Gummin DD, Mowry JB, Spyker DA, Brooks DE, Osterthaler KM, Banner W. Clin Toxicol (Phila) 2018;56:1213–1415. doi: 10.1080/15563650.2018.1533727. [DOI] [PubMed] [Google Scholar]
- 8.World Health Organization. World Heal. Organ. Vol. 24. 2017; 2017. Depression and Other Common Mental Disorders Global Health Estimates. [Google Scholar]
- 9.Association of CYP2C19 and CYP2D6 poor and intermediate metabolizer status with antidepressant and antipsychotic exposure: a systematic review and meta-analysis. Milosavljevic F, Bukvic N, Pavlovic Z, et al. JAMA Psychiatry. 2021;78:270–280. doi: 10.1001/jamapsychiatry.2020.3643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Risks of mental health outcomes in people with covid-19: cohort study. Xie Y, Xu E, Al-Aly Z. BMJ. 2022;376:0. doi: 10.1136/bmj-2021-068993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Examining the impact of the COVID-19 pandemic through the lens of the network approach to psychopathology: analysis of the Brazilian Longitudinal Study of Health (ELSA-Brasil) cohort over a 12-year timespan. Suen PJ, Bacchi PS, Razza L, et al. J Anxiety Disord. 2022;85:102512. doi: 10.1016/j.janxdis.2021.102512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Consumption and ocurrence of antidepressants (SSRIs) in pre- and post-COVID-19 pandemic, their environmental impact and innovative removal methods: a review. Diaz-Camal N, Cardoso-Vera JD, Islas-Flores H, Gómez-Oliván LM, Mejía-García A. Sci Total Environ. 2022;829:154656. doi: 10.1016/j.scitotenv.2022.154656. [DOI] [PubMed] [Google Scholar]
- 13.Action of isonicotinic acid hydrazide and related compounds on enzymes involved in the autonomic nervous system. Zeller EA, Barsky J, Berman ER, Fouts JR. https://scholar.google.com/scholar_lookup?title=Action%20of%20isonicotinic%20acid%20hydrazid%20and%20related%20compounds%20on%20enzymes%20involved%20in%20the%20autonomic%20nervous%20system&publication_year=1952&author=E.A.%20Zeller&author=J.%20Barsky&author=E.R.%20Berman&author=J.R.%20Fouts J Pharmacol Exp Ther. 1952;106:427–428. [Google Scholar]
- 14.Learning from the past and looking to the future: Emerging perspectives for improving the treatment of psychiatric disorders. Millan MJ, Goodwin GM, Meyer-Lindenberg A, Ove Ögren S. Eur Neuropsychopharmacol. 2015;25:599–656. doi: 10.1016/j.euroneuro.2015.01.016. [DOI] [PubMed] [Google Scholar]
- 15.Neurologic effects of tryptophan in patients receiving a monoamine oxidase inhibitor. Oates JA, Sjoerdsma A. Neurology. 1960;10:1076–1078. doi: 10.1212/wnl.10.12.1076. [DOI] [PubMed] [Google Scholar]
- 16.Brain stem and spinal cord mediation of a serotonergic behavioral syndrome. Jacobs BL, Klemfuss H. Brain Res. 1975;100:450–457. doi: 10.1016/0006-8993(75)90500-4. [DOI] [PubMed] [Google Scholar]
- 17.Motor effects of serotonin in the central nervous system. Gerson SC, Baldessarini RJ. Life Sci. 1980;27:1435–1451. doi: 10.1016/0024-3205(80)90368-9. [DOI] [PubMed] [Google Scholar]
- 18.The expanded biology of serotonin. Berger M, Gray JA, Roth BL. Annu Rev Med. 2009;60:355–366. doi: 10.1146/annurev.med.60.042307.110802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.A review of central 5-HT receptors and their function. Barnes NM, Sharp T. Neuropharmacology. 1999;38:1083–1152. doi: 10.1016/s0028-3908(99)00010-6. [DOI] [PubMed] [Google Scholar]
- 20.Long story short: the serotonin transporter in emotion regulation and social cognition. Canli T, Lesch KP. Nat Neurosci. 2007;10:1103–1109. doi: 10.1038/nn1964. [DOI] [PubMed] [Google Scholar]
- 21.Pharmacological analysis of the behavioural and thermoregulatory effects of the putative 5-HT1 receptor agonist, RU 24969, in the rat. Tricklebank MD, Middlemiss DN, Neill J. Neuropharmacology. 1986;25:877–886. doi: 10.1016/0028-3908(86)90014-6. [DOI] [PubMed] [Google Scholar]
- 22.Molecular basis of aggression. Nelson RJ, Chiavegatto S. Trends Neurosci. 2001;24:713–719. doi: 10.1016/s0166-2236(00)01996-2. [DOI] [PubMed] [Google Scholar]
- 23.The serotonin signaling system: from basic understanding to drug development for functional GI disorders. Gershon MD, Tack J. Gastroenterology. 2007;132:397–414. doi: 10.1053/j.gastro.2006.11.002. [DOI] [PubMed] [Google Scholar]
- 24.The ever-changing roles of serotonin. Jones LA, Sun EW, Martin AM, Keating DJ. Int J Biochem Cell Biol. 2020;125:105776. doi: 10.1016/j.biocel.2020.105776. [DOI] [PubMed] [Google Scholar]
- 25.Embracing diversity in the 5-HT neuronal system. Okaty BW, Commons KG, Dymecki SM. Nat Rev Neurosci. 2019;20:397–424. doi: 10.1038/s41583-019-0151-3. [DOI] [PubMed] [Google Scholar]
- 26.VMAT2: a dynamic regulator of brain monoaminergic neuronal function interacting with drugs of abuse. Eiden LE, Weihe E. Ann N Y Acad Sci. 2011;1216:86–98. doi: 10.1111/j.1749-6632.2010.05906.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.The therapeutic potential of monoamine oxidase inhibitors. Youdim MB, Edmondson D, Tipton KF. Nat Rev Neurosci. 2006;7:295–309. doi: 10.1038/nrn1883. [DOI] [PubMed] [Google Scholar]
- 28.International Union of Basic and Clinical Pharmacology. CX. Classification of receptors for 5-hydroxytryptamine; pharmacology and function. Barnes NM, Ahern GP, Becamel C, et al. Pharmacol Rev. 2021;73:310–520. doi: 10.1124/pr.118.015552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.The therapeutic role of 5-HT1A and 5-HT2A receptors in depression. Celada P, Puig MV, Amargós-Bosch M, Adell A, Artigas F. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC446220/pdf/20040700s00002p252.pdf. J Psychiatry Neurosci. 2004;29:252–265. [PMC free article] [PubMed] [Google Scholar]
- 30.Targeting the 5-HT(1B/1D) and 5-HT(1F) receptors for acute migraine treatment. Huang PC, Yang FC, Chang CM, Yang CP. Prog Brain Res. 2020;255:99–121. doi: 10.1016/bs.pbr.2020.05.010. [DOI] [PubMed] [Google Scholar]
- 31.Functional study of rat 5-HT2A receptors using antisense oligonucleotides. Van Oekelen D, Megens A, Meert T, Luyten WH, Leysen JE. J Neurochem. 2003;85:1087–1100. doi: 10.1046/j.1471-4159.2003.01738.x. [DOI] [PubMed] [Google Scholar]
- 32.The medical benefit of 5-HT research. Jones BJ, Blackburn TP. Pharmacol Biochem Behav. 2002;71:555–568. doi: 10.1016/s0091-3057(01)00745-6. [DOI] [PubMed] [Google Scholar]
- 33.The 5-HT3 receptor as a therapeutic target. Thompson AJ, Lummis SCR. Expert Opin Ther Targets. 2007;11:527–540. doi: 10.1517/14728222.11.4.527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Cloning, expression, and pharmacology of four human 5-hydroxytryptamine 4 receptor isoforms produced by alternative splicing in the carboxyl terminus. Blondel O, Gastineau M, Dahmoune Y, Langlois M, Fischmeister R. J Neurochem. 1998;70:2252–2261. doi: 10.1046/j.1471-4159.1998.70062252.x. [DOI] [PubMed] [Google Scholar]
- 35.Effects of lurasidone in behavioral models of depression. Role of the 5-HT₇ receptor subtype. Cates LN, Roberts AJ, Huitron-Resendiz S, Hedlund PB. Neuropharmacology. 2013;70:211–217. doi: 10.1016/j.neuropharm.2013.01.023. [DOI] [PubMed] [Google Scholar]
- 36.The anaesthetist, opioid analgesic drugs, and serotonin toxicity: a mechanistic and clinical review. Baldo BA, Rose MA. Br J Anaesth. 2020;124:44–62. doi: 10.1016/j.bja.2019.08.010. [DOI] [PubMed] [Google Scholar]
- 37.Serotonin syndrome: pathophysiology, clinical features, management, and potential future directions. Scotton WJ, Hill LJ, Williams AC, Barnes NM. Int J Tryptophan Res. 2019;12:1178646919873925. doi: 10.1177/1178646919873925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Association between serotonin syndrome and second-generation antipsychotics via pharmacological target-adverse event analysis. Racz R, Soldatos TG, Jackson D, Burkhart K. Clin Transl Sci. 2018;11:322–329. doi: 10.1111/cts.12543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.The pathophysiology of serotonin toxicity in animals and humans: implications for diagnosis and treatment. Isbister GK, Buckley NA. Clin Neuropharmacol. 2005;28:205–214. doi: 10.1097/01.wnf.0000177642.89888.85. [DOI] [PubMed] [Google Scholar]
- 40.L-DOPA elicits non-vesicular releases of serotonin and dopamine in hemiparkinsonian rats in vivo. Miguelez C, Navailles S, Delaville C, et al. Eur Neuropsychopharmacol. 2016;26:1297–1309. doi: 10.1016/j.euroneuro.2016.05.004. [DOI] [PubMed] [Google Scholar]
- 41.L-dopa induced "serotonin syndrome" in a parkinsonian patient on bromocriptine. Sandyk R. J Clin Psychopharmacol. 1986;6:194–195. doi: 10.1097/00004714-198606000-00022. [DOI] [PubMed] [Google Scholar]
- 42.The evidence for the involvement of the 5-HT1A receptor in 5-HT syndrome induced in mice by tryptamine. Yamada J, Sugimoto Y, Horisaka K. Jpn J Pharmacol. 1989;51:421–424. doi: 10.1254/jjp.51.421. [DOI] [PubMed] [Google Scholar]
- 43.Mice over-expressing the 5-HT(1A) receptor in cortex and dentate gyrus display exaggerated locomotor and hypothermic response to 8-OH-DPAT. Bert B, Fink H, Hörtnagl H, Veh RW, Davies B, Theuring F, Kusserow H. Behav Brain Res. 2006;167:328–341. doi: 10.1016/j.bbr.2005.09.020. [DOI] [PubMed] [Google Scholar]
- 44.Serotonin syndrome and other serotonergic disorders. Ener RA, Meglathery SB, Van Decker WA, Gallagher RM. Pain Med. 2003;4:63–74. doi: 10.1046/j.1526-4637.2003.03005.x. [DOI] [PubMed] [Google Scholar]
- 45.Effect of olanzapine on hyperthermia in serotonin syndrome model. Nikolov R, Bogdanov G. Int J Pharm Sci Res. 2020;11:4509–4512. [Google Scholar]
- 46.Serotonin syndrome in elderly patients treated for psychotic depression with atypical antipsychotics and antidepressants: two case reports. Kohen I, Gordon ML, Manu P. CNS Spectr. 2007;12:596–598. doi: 10.1017/s1092852900021386. [DOI] [PubMed] [Google Scholar]
- 47.Serotonin receptors involved in antidepressant effects. Artigas F. Pharmacol Ther. 2013;137:119–131. doi: 10.1016/j.pharmthera.2012.09.006. [DOI] [PubMed] [Google Scholar]
- 48.The selective 5-HT2A receptor antagonist M100907 enhances antidepressant-like behavioral effects of the SSRI fluoxetine. Marek GJ, Martin-Ruiz R, Abo A, Artigas F. Neuropsychopharmacology. 2005;30:2205–2215. doi: 10.1038/sj.npp.1300762. [DOI] [PubMed] [Google Scholar]
- 49.Serotonin 5-HT2 receptors activate local GABA inhibitory inputs to serotonergic neurons of the dorsal raphe nucleus. Liu R, Jolas T, Aghajanian G. Brain Res. 2000;873:34–45. doi: 10.1016/s0006-8993(00)02468-9. [DOI] [PubMed] [Google Scholar]
- 50.Behavioural evidence for a functional interaction between central 5-HT2 and 5-HT1A receptors. Backus LI, Sharp T, Grahame-Smith DG. Br J Pharmacol. 1990;100:793–799. doi: 10.1111/j.1476-5381.1990.tb14094.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Possible serotonin syndrome in association with 5-HT(3) antagonist agents. Turkel SB, Nadala JG, Wincor MZ. Psychosomatics. 2001;42:258–260. doi: 10.1176/appi.psy.42.3.258. [DOI] [PubMed] [Google Scholar]
- 52.Serotonin toxicity associated with the use of linezolid: a review of postmarketing data. Lawrence KR, Adra M, Gillman PK. Clin Infect Dis. 2006;42:1578–1583. doi: 10.1086/503839. [DOI] [PubMed] [Google Scholar]
- 53.Biochemical evidence for the regulation of central noradrenergic activity by 5-HT1A and 5-HT2 receptors: Microdialysis studies in the awake and anaesthetized rat. Done CJG, Sharp T. Neuropharmacology. 1994;33:411–421. doi: 10.1016/0028-3908(94)90071-x. [DOI] [PubMed] [Google Scholar]
- 54.Reverse Takotsubo cardiomyopathy after an episode of serotonin syndrome. Mehta NK, Aurigemma G, Rafeq Z, Starobin O. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3231548/ Tex Heart Inst J. 2011;38:568–572. [PMC free article] [PubMed] [Google Scholar]
- 55.Cardiogenic shock due to Takotsubo cardiomyopathy associated with serotonin syndrome. Sasaki H, Yumoto K, Nanao T, Nishizawa H, Funada S, Aoki H, Kato K. J Cardiol Cases. 2013;7:0–3. doi: 10.1016/j.jccase.2012.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Takotsubo cardiomyopathy associated with serotonin syndrome in a patient with stroke: a case report. Jang SH, Nam JH, Lee J, Chang MC. Medicine (Baltimore) 2019;98:0. doi: 10.1097/MD.0000000000015057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Implications of off-target serotoninergic drug activity: an analysis of serotonin syndrome reports using a aystematic bioinformatics approach. Culbertson VL, Rahman SE, Bosen GC, Caylor ML, Echevarria MM, Xu D. Pharmacotherapy. 2018;38:888–898. doi: 10.1002/phar.2163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Serotonin syndrome induced by fluvoxamine-lithium interaction. Ohman R, Spigset O. Pharmacopsychiatry. 1993;26:263–264. doi: 10.1055/s-2007-1014367. [DOI] [PubMed] [Google Scholar]
- 59.Manic switch and serotonin syndrome induced by augmentation of paroxetine with methylphenidate in a patient with major depression. Park YM, Jung YK. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34:719–720. doi: 10.1016/j.pnpbp.2010.03.016. [DOI] [PubMed] [Google Scholar]
- 60.Effects of methylphenidate on extracellular dopamine, serotonin, and norepinephrine: comparison with amphetamine. Kuczenski R, Segal DS. J Neurochem. 1997;68:2032–2037. doi: 10.1046/j.1471-4159.1997.68052032.x. [DOI] [PubMed] [Google Scholar]
- 61.The enhancement by lithium of the 5-HT1A mediated serotonin syndrome produced by 8-OH-DPAT in the rat: evidence for a post-synaptic mechanism. Goodwin GM, De Souza RJ, Wood AJ, Green AR. Psychopharmacology (Berl) 1986;90:488–493. doi: 10.1007/BF00174066. [DOI] [PubMed] [Google Scholar]
- 62.Antidepressant drugs effects on blood pressure. Calvi A, Fischetti I, Verzicco I, et al. Front Cardiovasc Med. 2021;8:704281. doi: 10.3389/fcvm.2021.704281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Serotonin uptake rates in platelets from angiotensin II-induced hypertensive mice. Singh P, Fletcher TW, Li Y, Rusch NJ, Kilic F. Health (Irvine Calif) 2013;5:31–39. doi: 10.4236/health.2013.54A005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Serotonin is associated with coronary artery disease and cardiac events. Vikenes K, Farstad M, Nordrehaug JE. Circulation. 1999;100:483–489. doi: 10.1161/01.cir.100.5.483. [DOI] [PubMed] [Google Scholar]
- 65.Frontiers of serotonin beyond the brain. Maroteaux L, Kilic F. Pharmacol Res. 2019;140:1–6. doi: 10.1016/j.phrs.2018.10.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Cigarette smoking-associated alterations in serotonin/adrenalin signaling pathways of platelets. Lowery CL 3rd, Elliott C, Cooper A, et al. J Am Heart Assoc. 2017;6:0. doi: 10.1161/JAHA.116.005465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Drug interactions due to cytochrome P450. Ogu CC, Maxa JL. Proc (Bayl Univ Med Cent) 2000;13:421–423. doi: 10.1080/08998280.2000.11927719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Pharmacogenetics of antipsychotics. Brandl EJ, Kennedy JL, Müller DJ. Can J Psychiatry. 2014;59:76–88. doi: 10.1177/070674371405900203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2D6 and CYP2C19 genotypes and dosing of selective serotonin reuptake inhibitors. Hicks JK, Bishop JR, Sangkuhl K, et al. Clin Pharmacol Ther. 2015;98:127–134. doi: 10.1002/cpt.147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Palpitations and asthenia associated with venlafaxine in a CYP2D6 poor metabolizer and CYP2C19 intermediate metabolizer. Garcia S, Schuh M, Cheema A, Atwal H, Atwal PS. Case Rep Genet. 2017;2017:6236714. doi: 10.1155/2017/6236714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Clinical relevance of pharmacogenetics in serotonin syndrome. Pandya D, Tran M, Verduzco-Gutierrez M. Case Rep Psychiatry. 2020;2020:8860434. doi: 10.1155/2020/8860434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Serotonin toxicity and cytochrome p450 poor metaboliser genotype patient case. Piatkov I, Mann G, Jones T, McLean M, Gunja N. J Investig Genomics. 2017;4:1–5. [Google Scholar]
- 73.Citalopram overdose and severe serotonin syndrome in an intermediate metabolizing patient. Schult RF, Morris AJ, Picard L, Wiegand TJ. Am J Emerg Med. 2019;37:1993–1996. doi: 10.1016/j.ajem.2019.06.038. [DOI] [PubMed] [Google Scholar]
- 74.CYP2D6 polymorphism and clinical effect of the antidepressant venlafaxine. Shams ME, Arneth B, Hiemke C, et al. J Clin Pharm Ther. 2006;31:493–502. doi: 10.1111/j.1365-2710.2006.00763.x. [DOI] [PubMed] [Google Scholar]
- 75.Serotonin toxicity from antidepressant overdose and its association with the T102C polymorphism of the 5-HT2A receptor. Cooper JM, Newby DA, Whyte IM, Carter G, Jones AL, Isbister GK. Pharmacogenomics J. 2014;14:390–394. doi: 10.1038/tpj.2013.47. [DOI] [PubMed] [Google Scholar]
- 76.The genetic foundations of serotonin syndrome, neuroleptic malignant syndrome, and malignant hyperthermia: is there a genetic association between these Disorders? Ortiz JF, Wirth M, Eskander N, Cozar JC, Fatade O, Rathod B. Cureus. 2020;12:0. doi: 10.7759/cureus.10635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.The role of serotonergic system at the interface of aggression and suicide. Bortolato M, Pivac N, Seler DM, Perkovic MN, Pessia M, Di Giovanni G. Neuroscience. 1990;236:160–185. doi: 10.1016/j.neuroscience.2013.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Differential expression of the "C" and "T" alleles of the 5-HT2A receptor gene in the temporal cortex of normal individuals and schizophrenics. Polesskaya OO, Sokolov BP. J Neurosci Res. 2002;67:812–822. doi: 10.1002/jnr.10173. [DOI] [PubMed] [Google Scholar]
- 79.Serotonin syndrome and the T102-->C polymorphism of the 5-HT2A receptor: a case report. Lattanzi L, Danesi R, Lastella M, et al. Bipolar Disord. 2008;10:655–656. doi: 10.1111/j.1399-5618.2008.00598.x. [DOI] [PubMed] [Google Scholar]
- 80.Pharmacogenetics of antidepressant medication intolerance. Murphy GM Jr, Kremer C, Rodrigues HE, Schatzberg AF. Am J Psychiatry. 2003;160:1830–1835. doi: 10.1176/appi.ajp.160.10.1830. [DOI] [PubMed] [Google Scholar]
- 81.The serotonin syndrome: from molecular mechanisms to clinical practice. Francescangeli J, Karamchandani K, Powell M, Bonavia A. Int J Mol Sci. 2019;20:2288. doi: 10.3390/ijms20092288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.The serotonin syndrome. Sternbach H. Am J Psychiatry. 1991;148:705–713. doi: 10.1176/ajp.148.6.705. [DOI] [PubMed] [Google Scholar]
- 83.An exploratory approach to the serotonin syndrome: an update of clinical phenomenology and revised diagnostic criteria. Radomski JW, Dursun SM, Reveley MA, Kutcher SP. Med Hypotheses. 2000;55:218–224. doi: 10.1054/mehy.2000.1047. [DOI] [PubMed] [Google Scholar]
- 84.Controversies in serotonin syndrome diagnosis and management: a review. Uddin MF, Alweis R, Shah SR, et al. J Clin Diagn Res. 2017;11:0–7. doi: 10.7860/JCDR/2017/29473.10696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Reineke EL. Small Anim Crit Care Med Second Ed. 414-8: Elsevier; Serotonin syndrome; pp. 414–418. [Google Scholar]
- 86.The serotonin syndrome and its treatment. Gillman PK. J Psychopharmacol. 1999;13:100–109. doi: 10.1177/026988119901300111. [DOI] [PubMed] [Google Scholar]
- 87.American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders: DSM-5. Vol. 5. Arlington, VA: American Psychiatric Publishing; 2013. Medication-Induced Movement Disorders and Other Adverse Effects of Medication; p. 991. [Google Scholar]
- 88.Neuroleptic malignant syndrome and serotonin syndrome. Tormoehlen LM, Rusyniak DE. Handb Clin Neurol. 2018;157:663–675. doi: 10.1016/B978-0-444-64074-1.00039-2. [DOI] [PubMed] [Google Scholar]
- 89.Treatment strategies for dosing the second generation antipsychotics. Schwartz TL, Stahl SM. CNS Neurosci Ther. 2011;17:110–117. doi: 10.1111/j.1755-5949.2011.00234.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Distinguishing between neuroleptic malignant syndrome and serotonin syndrome in polypharmacy: an overview with a case report. Debeljak T, Plesničar BK. https://www.psychiatria-danubina.com/UserDocsImages/pdf/dnb_vol33_noSuppl%204/dnb_vol33_noSuppl%204_1227.pdf. Psychiatr Danub. 2021;33:1227–1229. [PubMed] [Google Scholar]
- 91.Delayed onset serotonin syndrome in the setting of polypharmacy. Acharya R, Kafle S, Basnet S, Poudel D, Ghimire S. J Community Hosp Intern Med Perspect. 2021;11:76–78. doi: 10.1080/20009666.2020.1834929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Slow on the uptake, progression to heartbreak. Binder AK, Haydek JP, Parihar S, Modlin CE, Tannu M, Aldredge A, Sueblinvong V. J Investig Med High Impact Case Rep. 2020;8:2324709620936832. doi: 10.1177/2324709620936832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.An atypical presentation of serotonin syndrome. Landa E, Wagner S, Makkar A, Liu A, Jung D. Cureus. 2021;13:0. doi: 10.7759/cureus.13377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Linezolid‑induced serotonin syndrome. Shouan A, Kumar R, Lal V, Grover S. Ind Psychiatry J. 2021;29:345–348. doi: 10.4103/ipj.ipj_37_19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Refining the clinical features of serotonin syndrome: a prospective observational study of 45 patients. Prakash S, Rathore C, Rana KK, Dave A, Joshi H, Patel J. Ann Indian Acad Neurol. 2019;22:52–60. doi: 10.4103/aian.AIAN_344_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Serotonin syndrome associated with vilazodone overdose in a 22-month-old treated with dexmedetomidine. Schlichting E, Welter C, Schaeffer T, Strout TD. J Emerg Med. 2021;60:0–21. doi: 10.1016/j.jemermed.2020.10.012. [DOI] [PubMed] [Google Scholar]
- 97.Serotonin syndrome: analysis of cases registered in the French pharmacovigilance database. Abadie D, Rousseau V, Logerot S, Cottin J, Montastruc JL, Montastruc F. J Clin Psychopharmacol. 2015;35:382–388. doi: 10.1097/JCP.0000000000000344. [DOI] [PubMed] [Google Scholar]
- 98.Receptor mediation of exaggerated responses to serotonin-enhancing drugs in serotonin transporter (SERT)-deficient mice. Fox MA, Jensen CL, Gallagher PS, Murphy DL. Neuropharmacology. 2007;53:643–656. doi: 10.1016/j.neuropharm.2007.07.009. [DOI] [PubMed] [Google Scholar]
- 99.Neuroleptic malignant syndrome and serotonin syndrome. Nisijima K, Shioda K, Iwamura T. Prog Brain Res. 2007;162:81–104. doi: 10.1016/S0079-6123(06)62006-2. [DOI] [PubMed] [Google Scholar]
- 100.Atypical neuroleptic malignant syndrome: A systematic review of case reports. Singhai K, Kuppili PP, Nebhinani N. Gen Hosp Psychiatry. 2019;60:12–19. doi: 10.1016/j.genhosppsych.2019.06.009. [DOI] [PubMed] [Google Scholar]
- 101.Atypical neuroleptic malignant syndrome (NMS) Uvais NA. Asian J Psychiatr. 2017;30:120–121. doi: 10.1016/j.ajp.2017.08.020. [DOI] [PubMed] [Google Scholar]
- 102.Drug information update. Atypical antipsychotics and neuroleptic malignant syndrome: nuances and pragmatics of the association. Sarkar S, Gupta N. BJPsych Bull. 2017;41:211–216. doi: 10.1192/pb.bp.116.053736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Potent serotonin (5-HT)2A receptor antagonists completely prevent the development of hyperthermia in an animal model of the 5-HT syndrome. Nisijima K, Yoshino T, Yui K, Katoh S. Brain Res. 2001;890:23–31. doi: 10.1016/s0006-8993(00)03020-1. [DOI] [PubMed] [Google Scholar]
- 104.Gordon MF, Leder AN. Movement Disorder Emergencies. Vol. 10. Totowa, NJ: Humana Press; 2013. Serotonin Syndrome; p. 1007. [Google Scholar]
- 105.Cyproheptadine and the treatment of an unconscious patient with the serotonin syndrome. Baigel GD. Eur J Anaesthesiol. 2003;20:586–588. doi: 10.1017/s0265021503270927. [DOI] [PubMed] [Google Scholar]
- 106.Serotonin syndrome: a clinical review of current controversies. Werneke U, Truedson-Martiniussen P, Wikström H, Ott M. J Integr Neurosci. 2020;19:719–727. doi: 10.31083/j.jin.2020.04.314. [DOI] [PubMed] [Google Scholar]




