Skip to main content

Role of trazodone in treatment of major depressive disorder: an update

Abstract

Major depressive disorder (MDD) is the most common mood disorder and a leading cause of disability worldwide. Trazodone, a triazolopyridine serotonin receptor antagonist and reuptake inhibitor (SARI) antidepressant approved for major depressive disorder (MDD) in adults, has established efficacy that is comparable to other available antidepressants, and is effective for a range of depression symptoms, including insomnia, which is one of the most common and bothersome symptoms of depression. Also, trazodone’s pharmacodynamic properties allow it to avoid the side effects of insomnia, anxiety and sexual dysfunction often associated with selective serotonin reuptake inhibitor antidepressants. In this narrative review, we have summarized recent clinical trials and real-world data on trazodone, including the recently introduced once-daily formulation, which has single dose pharmacokinetic properties that maintain effective blood trazodone levels for 24 h, while avoiding concentration peaks associated with side effects. This, combined with a low incidence of weight gain, and sexual dysfunction, may improve adherence to treatment. The most common adverse effects of trazodone are somnolence, headache, dizziness and xerostomia. It has minimal anticholinergic activity but may be associated infrequently with orthostatic hypotension (especially in patients with cardiovascular disease or older adults), QT interval prolongation, cardiac arrhythmias, and rare episodes of priapism. The low liability for activating side effects, the efficacy on symptoms such as insomnia and psychomotor agitation and the rapid onset of action make it useful for many depressed patients, both in monotherapy at nominal dosages of 150–300 mg/day, and in combination with other antidepressants at lower dosages.

Introduction

Major depressive disorder (MDD) is the most common mood disorder and a leading cause of disability worldwide [1], affecting multiple functional domains that impact education, work productivity, and quality of life (QoL). Treatment can include psychotherapy, psychopharmacotherapy, or combinations of these. Antidepressant therapy, with the goal of achieving remission of symptoms and recovery of function is the most common treatment for severe depression [2, 3]; however, these goals are achieved in only about half of patients, and many experience relapses before recovery is achieved. This low success rate may be due to a lack of adequate treatment [4], or low treatment adherence [5]. Patients who require a longer treatment duration to achieve remission are at higher risk of early relapse [6]. Patients who fail to achieve remission after adequate trials with two or more different antidepressants have been described as treatment-resistant [7]. Difficult-to-treat depression (DTD) is a related concept describing “depression that continues to cause significant burden despite usual treatment efforts [8,9,10].

This narrative review provides an update on recent clinical data for trazodone in MDD and its place in current treatment strategies for MDD, with a focus on the once-daily trazodone Contramid® formulation, and symptoms that do not respond well to other treatments. In preparing it, we have conducted a literature search in PubMed using the keywords “trazodone” and “major depressive disorder” and focused on updating the 2012 review published by Fagiolini et al. [11].

Trazodone

Trazodone is a triazolopyridine derivative that inhibits the reuptake of serotonin and blocks histamine and alpha-1-adrenergic receptors. It is marketed in a variety of forms, including a once-daily formulation. Clinical studies have shown that the anti-depressive efficacy of trazodone, when administered at ≥ 150 mg/day, is comparable to that of tricyclic antidepressants [12,13,14], selective serotonin reuptake inhibitors [15,16,17,18], and serotonin-norepinephrine receptor inhibitors [19], (reviewed in [11]). Trazodone is generally well tolerated and may have additional advantages for patients who have both major depression and prevalent sleep disturbances [11].

Pharmacokinetic properties of trazodone formulations

The immediate-release formulation may be used as augmentation with another treatment, especially in patients who have major depressive disorder and accompanying symptoms such as insomnia, anxiety, irritability, or mild psychomotor agitation; however, its use requires administration in three separate doses, unless the medication is prescribed at relatively low doses, for example to target the symptom of insomnia. The immediate-release formulation is associated with fluctuations in plasma drug concentrations that increase the likelihood of adverse effects [11]. The Contramid® formulation is administered once daily at doses of up 300 mg at night, and provides controlled release of trazodone over 24 h [20]. Administration of trazodone Contramid® once daily for 7 days results in plasma AUC that is similar to that of the immediate-release formulation administered three times daily, but with peak maximum concentration (Cmax) that are 43% lower [21] (Fig. 1).

Fig. 1
figure 1

Pharmacokinetic properties of trazodone IR (immediate-release formulation, purple) dosed at 100 mg three times per day, and trazodone XR (extended-release, red) dosed at 300 mg once daily at night, showing plasma drug levels with respect to the minimum antidepressant concentration of 0.65 Mg/L over a 24 h period. (From Stahl [22])

This improves tolerability while increasing the probability of maintaining drug concentrations at levels adequate for antidepressant effectiveness.

Pharmacodynamic properties of trazodone and relationship to clinical efficacy

Dose-dependence of activity

Trazodone is a dose-dependent multifunctional drug that acts only on its highest affinity binding sites at low doses but has additional pharmacological actions at higher doses (Fig. 2).

Fig. 2
figure 2

Trazodone affinities at neurotransmitter receptors and transporters. The most potent action is antagonism at 5-HT2A receptors. 5-HT2A and 5-HT2C, subtypes of 5-hydroxytryptamine receptors; α1 and α2, subtypes of alpha adrenergic receptors; H1, histamine 1 receptor; SERT, serotonin reuptake transporter; D2, dopamine 2 receptor; DAT, dopamine transporter; M1, muscarinic 1 cholinergic receptor; NET, norepinephrine transporter. (From Stahl [22]

Doses of 150–600 mg are required to saturate serotonin transporters and achieve antidepressant activity (Box 1). At this dose, trazodone is a multifunctional serotonergic agent that inhibits the serotonin transporter (SERT) and antagonizes 5-HT2A and 5-HT2C receptors [22, 23], making it a multifunctional SARI.

Box 1 Summary of pharmacodynamic properties at 150–600 mg/day

Trazodone doses between 25 and 150 mg do not achieve antidepressant activity but have hypnotic activity through antagonism of 5-HT2a, H1, and α1 receptors. Estimates of brain receptor occupancy based on a pharmacokinetic model support the multimodal activity of trazodone, which may contribute to the rapid onset of its antidepressant action and its effectiveness for a variety of symptoms in depressed patients [24].

Rapid onset of action

Generally, the effectiveness of antidepressants remains suboptimal, and part of this may be due to their delayed onset of action. However, trazodone appears to have a rapid onset as seen in results from controlled clinical trials [25, 26], and the real-world setting [27]. This rapid onset of action may result from the efficacy on depression symptoms such as insomnia, anxiety, irritability, and psychomotor agitation [11, 28], as well as from the combined effects of partial 5-HT1A receptor agonism and serotonin transporter inhibition [29].

Safety and tolerability

Trazodone is generally well tolerated in patients with MDD and, compared to SSRIs and SNRIs it has a low risk for anxiety, insomnia, and sexual dysfunction, due to its simultaneous inhibition of 5-HT2A and 5-HT2C receptors, and serotonin transporters, along with its anti-alpha adrenergic and anti-histaminergic properties [11, 30]. The most common side effects include somnolence, headache, dizziness, and xerostomia.

Efforts should be made to minimize the impact of these side effects. Bedtime administration can reduce the impact of somnolence. Xerostomia, depending on severity, can cause difficulty in swallowing and increase the risk of oral infections and tooth decay, and may negatively affect quality of life with consequences for treatment adherence. Patients should be instructed to report side effects, including xerostomia, to the clinician, who may suggest strategies for managing them.

Other side effects

  • Arrhythmias—An ECG trial identified a modest, dose‐dependent effect of trazodone on cardiac repolarization, suggesting that caution is warranted in patients with concomitant medications known to prolong the QT interval [31].

  • Orthostatic hypotension, secondary to α1-adrenergic antagonism, may occur in patients receiving trazodone and could be more severe in patients also receiving antihypertensive therapy, especially in older adults or patients with heart disease [11].

  • Priapism is a rare AE with trazodone that is likely due to α-adrenergic antagonism. To reduce the risk, trazodone should be used with caution in patients with anatomical penis deformation or conditions including multiple myeloma or sickle cell anemia.

Tolerability of trazodone is further improved with the extended-release Contramid® formulation, which avoids concentration peaks associated with orthostatic hypotension and sedation, while allowing treatment to be initiated at a dose that is effective for depression [28]. In the RCT by Sheehan et al. side effects compared to placebo were mostly mild and their incidence decreased over time [25].

Pregnancy and lactation.

Trazodone is classified in the US FDA pregnancy category C; however, preliminary data suggest that maternal exposure to trazodone in early pregnancy is not associated with adverse outcomes [32]. Limited information indicates that trazodone levels in milk are low and would not be expected to cause any adverse effects in breastfed infants, especially if the infant is older than 2 months or when doses of 100 mg or less are used at bedtime for insomnia. Based on a safety scoring system for the use of psychotropic drugs during lactation, use of trazodone is considered “possible with caution” when necessary [33].

Update on clinical trials of trazodone in MDD

Since the last major review [11], three prospective trials have investigated the efficacy and safety of trazodone in patients with MDD [26, 34, 35].

The efficacy and safety of once-daily trazodone (Contramid® formulation) was compared to that of extended-release venlafaxine XR in an 8-week active controlled study that randomized 324 adults with MDD to receive trazodone 300 mg/day (n = 166) or venlafaxine 75–225 mg/day (n = 158) [26]. The primary efficacy endpoint was non-inferiority of the mean change from baseline on HAM-D-17 at the final study visit on day-56 in the ITT and per protocol populations. Secondary endpoints included the mean change from baseline in the Montgomery-Asberg Depression Rating Scale (MADRS) score at the final visit; (ii) Clinical Global Impression Severity (CGI-S) and Improvement (CGI-I) results at day-56, and the rates of response (≥ 50% reduction from baseline in HAM-D-17 total score) and remission (HAM-D-17 score ≤ 7) at the final visit. Patients were monitored at weeks 1, 3, 5, and 8, with additional safety visits after dose escalations.

The mean (SD) treatment doses administered were 311.4 ± 48.7 mg/day for trazodone and 84.1 ± 29.9 mg/day for venlafaxine. Both treatments effectively reduced HAM-D-17 scores at week 8 compared to baseline in the ITT population (trazodone − 12.9 ± 6.82, venlafaxine − 14.7 ± 6.56), and the per-protocol population (trazodone [n = 122] − 15.4 ± 5.32, venlafaxine [n = 127] − 16.4 ± 5.39). Efficacy was significantly higher in the venlafaxine group, while the reduction in HAM-D-17 score was significantly greater at day 7 in the trazodone group, compared to the venlafaxine group (p < 0.05). At treatment day 56, a response was achieved by 65.4% of patient in the trazodone group and 76.3% in the venlafaxine group (p < 0.05), while clinical remission was ongoing in 37.7% of patients in the trazodone group and 52.0% in the venlafaxine group.

Most reported AEs were mild or moderate in severity and a similar percentage of patients had treatment-related adverse events. The most frequent AEs in the trazodone group were dizziness and somnolence, while in the venlafaxine group they were nausea and headache [26].

Treatment-resistant depression [7] and difficult-to-treat depression [9] are common and burdensome. Effective treatment should address symptoms known to compromise response, functionality, and/or quality of life [8]. Pharmacological augmentation may be an effective component of a comprehensive, measurement-based strategy. Fang et al. assessed the effectiveness and tolerability of 5 different augmentation partners to pair with paroxetine 20 mg/day in an 8-week randomized study of 225 adults with treatment-resistant depression (TRD) [34]. Trazodone 100 mg/day was among the treatments assessed, along with buspirone 30 mg/day, risperidone 2 mg/day, thyroid hormone 80 mg/day, and valproic acid 600 mg/day. There were no statistically significant differences among groups for the primary outcome of remission defined as HAM-D-17 score of ≤ 7 at study end. Tolerability was also similar among groups, and no serious adverse events were reported during the study period. In the paroxetine plus trazodone group (n = 47), 20 patients (43%) achieved remission and 29 patients (62%) responded with a ≥ 50% reduction in HAM-D-17 total score at study end. There was no placebo group for comparison.

Zhang et al. assessed the efficacy, safety, and clinical benefit of prolonged-release trazodone (Trittico®; also referred to as slow-, delayed-, or controlled-release), which was administered twice daily in a 6-week randomized placebo-controlled flexible-dose study that randomly assigned 363 adults with MDD to receive prolonged release trazodone (150–450 mg/day) or placebo [35]. The prolonged-release trazodone group had received a mean maximum trazodone dosage of 273 mg/day, and had a significantly greater improvement from baseline in mean HAM-D-17 total score at study end (primary outcome) compared to placebo (− 11.07 vs. − 8.29, p < 0.001).

The rate of response defined as a ≥ 50% improvement in HAM-D-17 total score, was 59.6% (109/183) with prolonged-release trazodone vs. 37.2% (67/180) with placebo (p < 0.001), and the remission rate defined as a HAM-D-17 total score ≤ 7, was significantly higher with prolonged-release trazodone (35.5% [65/183] vs. 22.2% [40/180]; p = 0.005). Consistent with previous studies [25], sleep quality, assessed using the Pittsburgh Sleep Quality Index, was significantly improved from baseline with prolonged-release trazodone compared to placebo (p < 0.001).

Trazodone was well tolerated. AEs (n = 241) were mostly mild or moderate, and were reported by 156 of the 366 patients, including 100 patients who received prolonged-released trazodone (54.1%) and 56 patients who received placebo (30.9%). Dizziness, xerostomia, and somnolence were the most common; dizziness and somnolence were significantly more common with trazodone.

Recent real-world data

Several recent real-world studies have investigated the effectiveness and tolerability of trazodone [27, 36, 37]. Miljevic et al. conducted an eight-week multi-center open-label observational study to evaluate the effectiveness and tolerability of prolonged-release trazodone, in this case administered as a single daily dose in the evening to 242 adults with MDD experiencing a depressive episode without psychotic features [36]. Trazodone monotherapy was initiated at 50 mg and titrated to 150 mg over 2 weeks, with the possibility of increasing the dose up to 300 mg/day based on response (HAM-D-17) at week 2 compared to baseline. The 300 mg dose was administered to 54 patients. Outcomes were assessed at weeks 2, 4, and 8, using the HAM-D-17, HAM-A-14, and the CGI-S, and response was defined as ≥ 50% improvement from baseline on the HAM-D-17 or HAM-A-14. Significant improvements in depression were apparent at the first follow-up visit, with the HAM-D-17 baseline value of 23.74 (95% CI 23.15 to 24.33) reaching 17.32 (95% CI 16.59 to 18.06) after 2 weeks of treatment (p < 0.001) and continuing to improve to 8.67 (95% CI 8.02 to 9.32) at week 8 (p < 0.001). Similar improvements were observed on the HAM-A-14 scale. Responses were achieved by 12% of patients at week 2 and by more than 80% had responded at study end.

Adverse events (n = 73) were experienced by 52 patients, were mostly mild (62%), and were reported mainly at the first follow-up visit, when 42 patients reported events. The most frequent side effects were headache, sedation, and nausea. Severe side effects (vertigo or nausea) were reported by 11 patients. Overall, the results were similar to those from clinical trials [11], and supported the effectiveness and tolerability of trazodone for treating depression in routine clinical practice.

Some antidepressants have an initial delay before achieving a therapeutic response [38]. Intravenous administration has been suggested to accelerate attainment of steady-state therapeutic blood levels and treatment effects [39, 40]. A real-world study compared intravenous administration followed by oral dosing of trazodone vs clomipramine in 42 outpatient adults with MDD who were experiencing a major depressive episode [37]. Treatment was selected based on clinical judgement, with 26 patients receiving trazodone and 16 receiving clomipramine. Intravenous trazodone (25–100 mg/day, mean 44.2 ± 11.0 mg) or clomipramine (25–75 mg/day, mean 29.7 ± 9.30 mg) was administered for 1 week, with dosages based on the severity of depressive symptoms. Patients then switched to oral extended-release (Contramid®) trazodone (150–300 mg/day, mean 161.5 ± 67.4 mg) or clomipramine (50–225 mg/day, mean 98.4 ± 59.9 mg) for 4 weeks.

Patients were assessed at baseline and again after 1, 2, and 6 weeks by blinded raters using the HAM-D-17 and other psychometric scales. Response was defined as a ≥ 50% reduction in the HAM-D-17 total score and remission as achieving a HAM-D-17 total score < 8. The response rate at study end was 46.1% (12/26) in the trazodone group and 18.8% (3/16) in the clomipramine group. Remission was achieved by 34.6% (9/26) of patients receiving trazodone and 12.5% (2/16) of patients receiving clomipramine.

Side effects were reported by 4/26 patients receiving trazodone (2 sedation, 1 rash, and 1 dizziness), and 9/16 patients who received clomipramine (4 xerostomia, 1 sedation, 1 headache, and 1 dizziness). None of the patients discontinued treatment because of adverse events.

Češková et al. conducted a prospective real-world study to evaluate the efficacy, tolerability, and safety of once-daily trazodone Contramid® in 85 adults with moderate to severe MDD in routine clinical practice [27]. Trazodone Contramid® was effective in previously treated and treatment naïve patients, including those with depressive episodes not responding to previous antidepressant treatment. Mean reductions from baseline in overall MADRS score were significant after 1 week of titration (27.4 to 21.2; p < 0.001) decreased further after 4 weeks of full dosage treatment (7.9; p < 0.001), when scores for all MADRS items were significantly lower than baseline values. Depression severity also decreased, with 71/80 patients (94%) achieving a CGI-S of ≤ 3 after 5 weeks. Follow-up visits revealed that 79% of patients were still receiving trazodone at week 9 and 71% at week 21, and investigator-assessed depression status compared to the previous visit was either unchanged or improved in all patients. Investigator-reported treatment tolerability was good or excellent for all patients; the most frequent drug-related AEs were somnolence and fatigue.

Siwek et al. conducted a 12-week open-label study to compare the effectiveness of trazodone Contramid® vs. SSRIs for adults with MDD (n = 76) [41]. Based on clinical presentation, clinicians assigned 42 patients to trazodone (mean dose 209.4 mg/day), and 34 patients to an SSRI (25 sertraline and 9 escitalopram; mean dose 21.7 fluoxetine equivalent mg/day).

Assessments at baseline and weeks 2, 4, 8, and 12 included the change in depression severity (MADRS, Quick Inventory of Depressive Symptomatology [QIDS]), with response corresponding to a ≥ 50% reduction on the MADRS or QIDS, or improvement on the CGI-I. Response rates achieved after 12 weeks were similar in both groups, but trazodone was more effective than SSRIs at reducing insomnia severity on the Athens Insomnia Scale (− 12.5, 95% CI − 15.4 to − 9.5 vs. − 3.7, 95% CI − 5.7 to − 1.8; p < 0.001). Both treatments were well-tolerated; one patient discontinued due to adverse effects in each group (somnolence with trazodone, and hypomania with sertraline).

Candidate situations and populations for trazodone

MDD is heterogeneous [42, 43], and involves a variety of symptoms that may respond variably to a given treatment. For examples of clinical applications see Additional file 1. Because of its unique mechanism of action and low risk of adverse anticholinergic effects and sexual side effects, trazodone has several advantages when addressing comorbid anxiety, agitation, or insomnia [11], and has been used off label, especially for treating insomnia and anxiety [44]

Several studies have compared the effectiveness of trazodone in treating depression to other classes of antidepressants, including selective serotonin reuptake inhibitors (SSRIs) such as fluoxetine, paroxetine, sertraline, citalopram, and escitalopram, serotonin-norepinephrine reuptake inhibitors (SNRIs) like venlafaxine and mirtazapine, inhibitors of norepinephrine and dopamine reuptake (bupropion), and tricyclic antidepressants (specifically amitriptyline and imipramine) [11]. The consistent findings of these studies indicate that trazodone's efficacy is comparable to that of these other antidepressants. However, trazodone's mechanism of action makes it well-suited for addressing certain aspects of depression, including insomnia, agitation, anxiety, and irritability. It sets itself apart from benzodiazepines due to its lower risk of abuse, dependence, and tolerance. In contrast to tricyclic antidepressants, it typically avoids causing anticholinergic side effects. Unlike mirtazapine, it generally does not lead to increased appetite or body weight. Moreover, unlike certain selective serotonin reuptake inhibitors, it does not impact the levels of other drugs in the bloodstream through interactions with cytochrome P450 enzymes. Nonetheless, trazodone is not without potential drawbacks, and special caution should be exercised in patients with an elevated risk of arrhythmias or falls due to excessive sedation or orthostatic hypotension. The utility of the available trazodone formulations in different clinical settings are summarized in Box 2.

Box 2 Different forms of trazodone present options to tailor treatment based on individual patient symptoms and requirements

Residual insomnia in treated MDD

Insomnia is a common symptom in MDD [48] and is associated with substantial clinical and economic burden [49, 50]. Insomnia may be exacerbated by the activating effects of some antidepressants [51]. Trazodone has hypnotic action at doses lower than those used for treating depression (i.e., < 150 mg/day), which is primarily due to its ability to block 5-HT2A, H1, and alpha-1 adrenergic receptors [22]. The results of a systematic review suggest that trazodone is effective for insomnia, although its use for this indication is off label [52].

Results from RCTs have shown beneficial effects on sleep disorders with trazodone in the immediate-release [19, 53] and prolonged-release formulations [17, 18]. A post-hoc analysis [54] of the placebo-controlled study on once-daily extended-release trazodone (Contramid®) by Sheehan et al. [25], revealed that patients with MDD who had been randomized to receive trazodone had significant improvements in insomnia-related symptoms, and had the greatest improvement among the HAM-D-17 and MADRS items. This analysis also confirmed that the antidepressant effect of trazodone is independent of its effects on insomnia. In a double-blind RCT comparing trazodone Contramid® with the active control venlafaxine XR in patients with MDD, both treatments significantly reduced HAM-D-17 total scores at day 56, while trazodone was significantly more effective at reducing the HAM-D-17 items related to insomnia at all time points [26].

Major depressive episodes with mixed features

Concomitant manic symptoms not meeting bipolar disorder criteria occur during major depressive episodes in approximately one in five patients with major depressive disorder [55]. Mixed features correlate with more severe disease, longer time to recovery, and poor response to treatment [56]. This condition is recognized in the 5th edition of the American Psychiatric Association diagnostic and statistical manual of mental disorders, where it is described as major depressive episodes with mixed features (MDE-MF), and defined as a major depressive episode accompanied three or more (hypo)manic symptoms that occur on most days [57]. Anxiety and agitation are excluded from the DSM5 definition, but these so-called overlapping symptoms are often associated with mixed features in clinical practice and are included in the research‐based diagnostic criteria used in the Improving Diagnosis, Guidance and Education (BRIDGE)-II-MIX study [58, 59]. A cluster analysis of the BRIDGE Study suggested that these overlapping symptoms increase sensitivity for detecting MDE-MF [60].

Fagiolini et al. [61] proposed that patients with MDD who experience an MDE-MF, with or without psychomotor agitation (i.e., irritability, restlessness, lack of impulse control, high suicide risk, racing thoughts, increased energy, severe insomnia, or reduced need for sleep), should receive an antimanic agent during the acute episode, and be considered for treatment with an antidepressant that does not increase dopamine or norepinephrine levels, such as citalopram or trazodone. Some antidepressant treatments exacerbate agitation and worsen mania symptoms during mixed episodes, without resolving depressive symptoms. Trazodone has a low propensity for causing agitation or psychomotor activation, which allows trazodone to be used in MDD patients with isolated (hypo)manic symptoms.

Patients with MDD and alcohol use disorder

Individually, MDD and AUD pose significant challenges to public health due to their widespread occurrences and the serious impact they have on clinical and functional well-being [1, 62]. Their co-occurrence is common, yet effective treatment strategies for addressing both conditions together remain limited [63, 64]. Symptoms of anxiety and insomnia are often observed in individuals with AUD [65, 66], and in patients with MDD [48, 67].

Trazodone improved polysomnographic sleep efficiency after detoxification in patients with AUD in a small placebo controlled study (n = 16), with an immediate effect that was sustained at 4 weeks [68]; however, a placebo-controlled trial in subjects with self-reported sleep disturbances after alcohol detoxification revealed an improvement in sleep quality while receiving low dose trazodone (50 to 150 mg/day), but an increase in alcohol intake after treatment discontinuation at week 12 [69]. Di Nicola et al. retrospectively evaluated the effect of extended-release trazodone, flexibly dosed at 150–300 mg/day, in outpatients with MDD and comorbid alcohol use disorder (n = 100) [70]. After 6 months of treatment, there had been a significant improvement in depressive symptoms (primary outcome), with 55% of patients achieving remission. Similar improvements were observed in measures of anxiety, sleep quality, functioning, quality of life, clinical global severity, as well as alcohol craving.

Patients with history of non-adherence to antidepressants

Lack of adherence to therapy is a major contributor to poor outcomes in MDD, and simpler treatment regimens may overcome this [71]. Prescribing poorly tolerated antidepressants or agents with complex regimens can compromise patient adherence to antidepressant treatment [72]. The low incidence of side effects with trazodone and the simplified administration of once daily oral dosing at bedtime may improve tolerability and potentially lead to improved adherence.

Conclusions

Trazodone is a well-established antidepressant with good tolerability and a comparable efficacy profile to other antidepressants, as demonstrated in both clinical trials and clinical practice. Trazodone may be particularly useful in patients with major depressive disorder and accompanying symptoms such as insomnia, anxiety, irritability, or psychomotor agitation.

Appropriate doses for achieving antidepressant effect in adult patients are usually 150–300 mg/day and are often higher than the doses used when trazodone is prescribed to enhance the antidepressant effect of another drug (e.g., to treat insomnia). Trazodone is usually well tolerated and carries a low risk of anticholinergic side effects (e.g., constipation, urinary retention, xerostomia), weight gain, and sexual side effects.

The Contramid® ER formulation of trazodone provides sustained release over a 24-h period, avoiding peaks and troughs in drug concentration and the associated increase in occurrence or severity of certain side effects (e.g., sedation or orthostatic hypotension). This allows once daily dosing, which increases convenience and patient adherence to therapy.

Availability of data and materials

Data sharing is not applicable to this article as no datasets were generated or analysed during the current study.

Abbreviations

AEs:

Adverse Events

CGI-I:

Clinical Global Impression Improvement

CGI-S:

Clinical Global Impression Severity

DTD:

Difficult-to-Treat Depression

HAM-D-17:

Hamilton Rating Scale for Depression

ITT:

Intention to treat

MADRS:

Montgomery-Asberg Depression Rating Scale

MDD:

Major Depressive Disorder

MDE-MF:

Major Depressive Episodes with Mixed Features

QIDS:

Quick Inventory of Depressive Symptomatology

QoL:

Quality of Life

RCT:

Randomized Clinical Trial

SARI:

Serotonin receptor Antagonist and Reuptake Inhibitor

SERT:

Serotonin Transporter

SNRIs:

Serotonin and Norepinephrine Reuptake Inhibitors

SSRIs:

Selective Serotonin Reuptake Inhibitors

TRD:

Treatment-Resistant Depression

References

  1. GBD 2019 Diseases and Injuries Collaborators. Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet. 2020; 396: 1204–22.

  2. American Psychiatric Association. Clinical practice guideline for the treatment of depression across three age cohorts. 2019. https://www.apa.org/depression-guideline.

  3. Bauer M, Pfennig A, Severus E, Whybrow PC, Angst J, Möller H-J, et al. World federation of societies of biological psychiatry (WFSBP) guidelines for biological treatment of unipolar depressive disorders, part 1: update 2013 on the acute and continuation treatment of unipolar depressive disorders. World J Biol Psychiatry. 2013;14:334–85.

    PubMed  Google Scholar 

  4. Thase ME. Evaluating antidepressant therapies: remission as the optimal outcome. J Clin Psychiatry. 2003;64(Suppl 13):18–25.

    CAS  PubMed  Google Scholar 

  5. Sansone RA, Sansone LA. Antidepressant adherence: are patients taking their medications? Innov Clin Neurosci. 2012;9:41–6.

    PubMed  PubMed Central  Google Scholar 

  6. Kubo K, Sakurai H, Tani H, Watanabe K, Mimura M, Uchida H. Predicting relapse from the time to remission during the acute treatment of depression: a re-analysis of the STAR*D data. J Affect Disord. 2023;320:710–5.

    PubMed  Google Scholar 

  7. Sforzini L, Worrell C, Kose M, Anderson IM, Aouizerate B, Arolt V, et al. A Delphi-method-based consensus guideline for definition of treatment-resistant depression for clinical trials. Mol Psychiatry. 2022;27:1286–99.

    CAS  PubMed  Google Scholar 

  8. McAllister-Williams RH, Arango C, Blier P, Demyttenaere K, Falkai P, Gorwood P, et al. The identification, assessment and management of difficult-to-treat depression: An international consensus statement. J Affect Disord. 2020;267:264–82.

    CAS  PubMed  Google Scholar 

  9. Rush AJ, Aaronson ST, Demyttenaere K. Difficult-to-treat depression: A clinical and research roadmap for when remission is elusive. Aust N Z J Psychiatry. 2019;53:109–18.

    PubMed  Google Scholar 

  10. McAllister-Williams RH. When depression is difficult to treat. Eur Neuropsychopharmacol. 2022;56:89–91.

    CAS  PubMed  Google Scholar 

  11. Fagiolini A, Comandini A, Catena Dell’Osso M, Kasper S. Rediscovering trazodone for the treatment of major depressive disorder. CNS Drugs. 2012;26:1033–49.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Gerner R, Estabrook W, Steuer J, Jarvik L. Treatment of geriatric depression with trazodone, imipramine, and placebo: a double-blind study. J Clin Psychiatry. 1980;41:216–20.

    CAS  PubMed  Google Scholar 

  13. Ather SA, Ankier SI, Middleton RS. A double-blind evaluation of trazodone in the treatment of depression in the elderly. Br J Clin Pract. 1985;39:192–9.

    CAS  PubMed  Google Scholar 

  14. Altamura AC, Mauri MC, Rudas N, Carpiniello B, Montanini R, Perini M, et al. Clinical activity and tolerability of trazodone, mianserin, and amitriptyline in elderly subjects with major depression: a controlled multicenter trial. Clin Neuropharmacol. 1989;12(1):25–33.

    Google Scholar 

  15. Falk WE, Rosenbaum JF, Otto MW, Zusky PM, Weilburg JB, Nixon RA. Fluoxetine versus trazodone in depressed geriatric patients. J Geriatr Psychiatry Neurol. 1989;2:208–14.

    CAS  PubMed  Google Scholar 

  16. Beasley CM, Dornseif BE, Pultz JA, Bosomworth JC, Sayler ME. Fluoxetine versus trazodone: efficacy and activating-sedating effects. J Clin Psychiatry. 1991;52:294–9.

    PubMed  Google Scholar 

  17. Kasper S, Olivieri L, Di Loreto G, Dionisio P. A comparative, randomised, double-blind study of trazodone prolonged-release and paroxetine in the treatment of patients with major depressive disorder. Curr Med Res Opin. 2005;21:1139–46.

    CAS  PubMed  Google Scholar 

  18. Munizza C, Olivieri L, Di Loreto G, Dionisio P. A comparative, randomized, double-blind study of trazodone prolonged-release and sertraline in the treatment of major depressive disorder. Curr Med Res Opin. 2006;22:1703–13.

    CAS  PubMed  Google Scholar 

  19. Cunningham LA, Borison RL, Carman JS, Chouinard G, Crowder JE, Diamond BI, et al. A comparison of venlafaxine, trazodone, and placebo in major depression. J Clin Psychopharmacol. 1994;14:99–106.

    CAS  PubMed  Google Scholar 

  20. US Food and Drug Administration. Oleptro prescribing information. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/022411s008lbl.pdf

  21. Karhu D, Gossen ER, Mostert A, Cronjé T, Fradette C. Safety, tolerability, and pharmacokinetics of once-daily trazodone extended-release caplets in healthy subjects. Int J Clin Pharmacol Ther. 2011;49:730–43.

    CAS  PubMed  Google Scholar 

  22. Stahl SM. Mechanism of action of trazodone: a multifunctional drug. CNS Spectr. 2009;14:536–46.

    PubMed  Google Scholar 

  23. OleptroTM (trazodone hydrochloride) extended-release tablets. P T. 2011; 36: 2–18.

  24. Oggianu L, Di Dato G, Mangano G, Rosignoli MT, McFeely S, Ke AB, et al. Estimation of brain receptor occupancy for trazodone immediate release and once a day formulations. Clin Transl Sci. 2022;15:1417–29.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Sheehan DV, Croft HA, Gossen ER, Levitt RJ, Brullé C, Bouchard S, et al. Extended-release Trazodone in major depressive disorder: a randomized, double-blind, placebo-controlled study. Psychiatry (Edgmont). 2009;6:20–33.

    PubMed  Google Scholar 

  26. Fagiolini A, Albert U, Ferrando L, Herman E, Muntean C, Pálová E, et al. A randomized, double-blind study comparing the efficacy and safety of trazodone once-a-day and venlafaxine extended-release for the treatment of patients with major depressive disorder. Int Clin Psychopharmacol. 2020;35:137–46.

    PubMed  PubMed Central  Google Scholar 

  27. Češková E, Šedová M, Kellnerová R, Starobová O. Once-a-day Trazodone in the treatment of depression in routine clinical practice. Pharmacology. 2018;102:206–12.

    PubMed  Google Scholar 

  28. Goracci A, Forgione RN, De Giorgi R, Coluccia A, Cuomo A, Fagiolini A. Practical guidance for prescribing trazodone extended-release in major depression. Expert Opin Pharmacother. 2016;17:433–41.

    CAS  PubMed  Google Scholar 

  29. Albert U, Lamba P, Stahl SM. Early response to trazodone once-a-day in major depressive disorder: review of the clinical data and putative mechanism for faster onset of action. CNS Spectr. 2021;26:232–42.

    PubMed  Google Scholar 

  30. Cuomo A, Ballerini A, Bruni AC, Decina P, Di Sciascio G, Fiorentini A, et al. Clinical guidance for the use of trazodone in major depressive disorder and concomitant conditions: pharmacology and clinical practice. Riv Psichiatr. 2019;54:137–49.

    PubMed  Google Scholar 

  31. Tellone V, Rosignoli MT, Picollo R, Dragone P, Del Vecchio A, Comandini A, et al. Effect of 3 single doses of Trazodone on QTc Interval in Healthy Subjects. J Clin Pharmacol. 2020;60:1483–95.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Dao K, Shechtman S, Diav-Citrin O, George N, Richardson JL, Rollason V, et al. Reproductive safety of Trazodone after maternal exposure in early pregnancy: a comparative ENTIS cohort study. J Clin Psychopharmacol. 2023;43:12–9.

    CAS  PubMed  Google Scholar 

  33. Uguz F. A new safety scoring system for the use of psychotropic drugs during lactation. Am J Ther. 2021;28:e118–26.

    PubMed  Google Scholar 

  34. Fang Y, Yuan C, Xu Y, Chen J, Wu Z, Cao L, et al. A pilot study of the efficacy and safety of paroxetine augmented with risperidone, valproate, buspirone, trazodone, or thyroid hormone in adult Chinese patients with treatment-resistant major depression. J Clin Psychopharmacol. 2011;31:638–42.

    CAS  PubMed  Google Scholar 

  35. Zhang L, Xie W-W, Li L-H, Zhang H-G, Wang G, Chen D-C, et al. Efficacy and safety of prolonged-release trazodone in major depressive disorder: a multicenter, randomized, double-blind, flexible-dose trial. Pharmacology. 2014;94:199–206.

    CAS  PubMed  Google Scholar 

  36. Miljevic CD, Lečić-Toševski D. Trazodone Study Group Serbia null. Efficacy and tolerability of trazodone retard monotherapy: results of the Serbian non-interventional study. Int J Psychiatry Clin Pract. 2016;20:133–40.

    CAS  PubMed  Google Scholar 

  37. Buoli M, Rovera C, Pozzoli SM, Fiorentini A, Cremaschi L, Caldiroli A, et al. Is trazodone more effective than clomipramine in major depressed outpatients? A single-blind study with intravenous and oral administration. CNS Spectr. 2019;24:258–64.

    PubMed  Google Scholar 

  38. Witkin JM, Knutson DE, Rodriguez GJ, Shi S. Rapid-Acting Antidepressants. Curr Pharm Des. 2018;24:2556–63.

    CAS  PubMed  Google Scholar 

  39. Vieta E, Florea I, Schmidt SN, Areberg J, Ettrup A. Intravenous vortioxetine to accelerate onset of effect in major depressive disorder: a 2-week, randomized, double-blind, placebo-controlled study. Int Clin Psychopharmacol. 2019;34:153–60.

    PubMed  PubMed Central  Google Scholar 

  40. Rancans E, Zambori J, Dalsgaard M, Baayen C, Areberg J, Ettrup A, et al. Intravenous vortioxetine to accelerate onset of effect in major depressive disorder: a 7-day randomized, double-blind, placebo-controlled exploratory study. Int Clin Psychopharmacol. 2020;35:305–12.

    PubMed  PubMed Central  Google Scholar 

  41. Siwek M, Gorostowicz A, Chrobak AA, Gerlich A, Krupa AJ, Juryk A, et al. TED-trazodone efficacy in depression: a naturalistic study on the efficacy of trazodone in an extended-release formulation compared to SSRIS in patients with a depressive episode-preliminary report. Brain Sci. 2023;13:86.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Monroe SM, Anderson SF. Depression: the shroud of heterogeneity. Curr Dir Psychol Sci. 2015;24:227–31. https://doi.org/10.1177/0963721414568342.

    Article  Google Scholar 

  43. Fried EI, Nesse RM. Depression is not a consistent syndrome: An investigation of unique symptom patterns in the STAR*D study. J Affect Disord. 2015;172:96–102.

    PubMed  Google Scholar 

  44. Bossini L, Coluccia A, Casolaro I, Benbow J, Amodeo G, De Giorgi R, et al. Off-label trazodone prescription: evidence. Benefits and Risks Curr Pharm Des. 2015;21:3343–51.

    CAS  PubMed  Google Scholar 

  45. US Food and Drug Administration. Desyrel prescribing information. 2017. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/018207s032lbl.pdf

  46. Monteleone P, Delrio G. Pharmacokinetic and pharmacodynamic characteristics of a controlled-release formulation of trazodone versus the conventional formulation in healthy volunteers. Ital J Neuro Sci. 1993;14:443–9.

    CAS  Google Scholar 

  47. Ballerio M, Politi P, Crapanzano C, Emanuele E, Cuomo A, Goracci A, et al. Clinical effectiveness of parenteral trazodone for the management of psychomotor activation in patients with bipolar disorder. Neuro Endocrinol Lett. 2018;39:205–8.

    PubMed  Google Scholar 

  48. Nutt D, Wilson S, Paterson L. Sleep disorders as core symptoms of depression. Dialogues Clin Neurosci. 2008;10:329–36.

    PubMed  PubMed Central  Google Scholar 

  49. Tian H, Abouzaid S, Gabriel S, Kahler KH, Kim E. Resource utilization and costs associated with insomnia treatment in patients with major depressive disorder. Prim Care Companion CNS Disord. 2012;14:PCC.12m01374.

    PubMed  PubMed Central  Google Scholar 

  50. Joshi K, Cambron-Mellott MJ, Costantino H, Pfau A, Jha MK. The real-world burden of adults with major depressive disorder with moderate or severe insomnia symptoms in the United States. J Affect Disord. 2023;323:698–706.

    PubMed  Google Scholar 

  51. Wichniak A, Wierzbicka A, Walęcka M, Jernajczyk W. Effects of antidepressants on sleep. Curr Psychiatry Rep. 2017;19:63.

    PubMed  PubMed Central  Google Scholar 

  52. Jaffer KY, Chang T, Vanle B, Dang J, Steiner AJ, Loera N, et al. Trazodone for insomnia: a systematic review. Innov Clin Neurosci. 2017;14:24–34.

    PubMed  PubMed Central  Google Scholar 

  53. Weisler RH, Johnston JA, Lineberry CG, Samara B, Branconnier RJ, Billow AA. Comparison of bupropion and trazodone for the treatment of major depression. J Clin Psychopharmacol. 1994;14:170–9.

    CAS  PubMed  Google Scholar 

  54. Sheehan DV, Rozova A, Gossen ER, Gibertini M. The efficacy and tolerability of once-daily controlled-release trazodone for depressed mood, anxiety, insomnia, and suicidality in major depressive disorder. Psychopharmacol Bull. 2009;42:5–22.

    PubMed  Google Scholar 

  55. McIntyre RS, Soczynska JK, Cha DS, Woldeyohannes HO, Dale RS, Alsuwaidan MT, et al. The prevalence and illness characteristics of DSM-5-defined “mixed feature specifier” in adults with major depressive disorder and bipolar disorder: results from the International Mood Disorders Collaborative Project. J Affect Disord. 2015;172:259–64.

    PubMed  Google Scholar 

  56. Shim IH, Bahk W-M, Woo YS, Yoon B-H. Pharmacological treatment of major depressive episodes with mixed features: a systematic review. Clin Psychopharmacol Neurosci. 2018;16:376–82.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. DSM-5-TR. American Psychiatric Association Publishing; 2022. Doi: https://doi.org/10.1176/appi.books.9780890425787.Accessed 12 Mar 2023

  58. Angst J, Gamma A, Bowden CL, Azorin JM, Perugi G, Vieta E, et al. Evidence-based definitions of bipolar-I and bipolar-II disorders among 5,635 patients with major depressive episodes in the Bridge Study: validity and comorbidity. Eur Arch Psychiatry Clin Neurosci. 2013;263:663–73.

    CAS  PubMed  Google Scholar 

  59. Perugi G, Angst J, Azorin J-M, Bowden CL, Mosolov S, Reis J, et al. Mixed features in patients with a major depressive episode: the BRIDGE-II-MIX study. J Clin Psychiatry. 2015;76:e351-358.

    PubMed  Google Scholar 

  60. Brancati GE, Vieta E, Azorin J-M, Angst J, Bowden CL, Mosolov S, et al. The role of overlapping excitatory symptoms in major depression: are they relevant for the diagnosis of mixed state? J Psychiatr Res. 2019;115:151–7.

    PubMed  Google Scholar 

  61. Fagiolini A, Cuomo A. Treating major depressive disorder with mixed features. Eur Neuropsychopharmacol. 2023;69:58–9.

    CAS  PubMed  Google Scholar 

  62. Lim SS, Vos T, Flaxman AD, Danaei G, Shibuya K, Adair-Rohani H, et al. A comparative risk assessment of burden of disease and injury attributable to 67 risk factors and risk factor clusters in 21 regions, 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380:2224–60.

    PubMed  PubMed Central  Google Scholar 

  63. Agabio R, Trogu E, Pani PP. Antidepressants for the treatment of people with co-occurring depression and alcohol dependence. Cochrane Database Syst Rev. 2018;4:CD008581.

  64. Geoffroy PA, Lejoyeux M, Rolland B. Management of insomnia in alcohol use disorder. Expert Opin Pharmacother. 2020;21:297–306.

    PubMed  Google Scholar 

  65. Brower KJ, Aldrich MS, Robinson EA, Zucker RA, Greden JF. Insomnia, self-medication, and relapse to alcoholism. Am J Psychiatry. 2001;158:399–404.

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Anker JJ, Kushner MG. Co-occurring alcohol use disorder and anxiety: bridging psychiatric, psychological, and neurobiological perspectives. Alcohol Res. 2019. https://doi.org/10.35946/arcr.v40.1.03.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Hasin DS, Sarvet AL, Meyers JL, Saha TD, Ruan WJ, Stohl M, et al. Epidemiology of Adult DSM-5 major depressive disorder and its specifiers in the United States. JAMA Psychiat. 2018;75:336–46.

    Google Scholar 

  68. Le Bon O, Murphy JR, Staner L, Hoffmann G, Kormoss N, Kentos M, et al. Double-blind, placebo-controlled study of the efficacy of trazodone in alcohol post-withdrawal syndrome: polysomnographic and clinical evaluations. J Clin Psychopharmacol. 2003;23:377–83.

    PubMed  Google Scholar 

  69. Friedmann PD, Rose JS, Swift R, Stout RL, Millman RP, Stein MD. Trazodone for sleep disturbance after alcohol detoxification: a double-blind, placebo-controlled trial. Alcohol Clin Exp Res. 2008;32:1652–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Di Nicola M, Pepe M, Panaccione I, Moccia L, Janiri L, Sani G. Update on Pharmacological Treatment for Comorbid Major Depressive and Alcohol Use Disorders: The Role of Extended-release Trazodone. Curr Neuropharmacol. 2023. https://doi.org/10.2174/1570159X21666230403080624.

    Article  PubMed  PubMed Central  Google Scholar 

  71. De las Cuevas C, Peñate W, Sanz EJ. Risk factors for non-adherence to antidepressant treatment in patients with mood disorders. Eur J Clin Pharmacol. 2014;70:89–98.

    CAS  PubMed  Google Scholar 

  72. Solmi M, Miola A, Croatto G, Pigato G, Favaro A, Fornaro M, et al. How can we improve antidepressant adherence in the management of depression? A targeted review and 10 clinical recommendations. Braz J Psychiatry. 2021;43:189–202.

    PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Angelini sponsored the expert meeting from which this manuscript arose. Angelini also supported financially Ethos srl which supported the authors in the preparation of the manuscript. This has not impacted the content of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

AF planned and designed the study, interpreted the data, drafted, and revised the manuscript. AGP planned and designed the study, interpreted the data, drafted, and revised the manuscript. KWM planned and designed the study, interpreted the data, drafted, and revised the manuscript. PM planned and designed the study, interpreted the data, drafted, and revised the manuscript. AHY planned and designed the study, interpreted the data, drafted, and revised the manuscript. EV planned and designed the study, interpreted the data, drafted, and revised the manuscript. All authors approved the final submitted version.

Corresponding author

Correspondence to Andrea Fagiolini.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

AF is /has been a consultant and/or a speaker and/or has received research grants from Angelini, Apsen, Biogen, Boheringer Ingelheim, Glaxo Smith Kline, Lundbeck, Janssen, Mylan, Neuraxpharm, Otsuka, Pfizer, Recordati, Rovi, Sanofi Aventis, Viatris, Vifor. AGP has received grants from or served as a consultant, advisor, or CME speaker for the following entities: Almirall, AstraZeneca, Bristol-Myers Squibb, Cephalon, Eli Lilly, Glaxo-Smith-Kline, Janssen-Cilag, Ferrer, Johnson & Johnson, Lundbeck, Merck, Otsuka, Pfizer, Sanofi-Aventis, Servier, Shering-Plough, Solvay, the Spanish Ministries of Science and Innovation (through CIBERSAM) and of Science (through the Carlos III Institute of Health), the Basque Government, the Stanley Medical Research Institute, and Wyeth. KWM has received consultancy fees from Lundbeck, Allergan and Janssen in the past three years. PM has received in the past 3 years grants, CME-related honoraria, or consulting fees from Angelini, AstraZeneca, Bial, Biogen, DGS-Portugal, FCT, FLAD, Janssen-Cilag, Gulbenkian Foundation, Lundbeck, Springer Healthcare, Tecnimede, Viatris and 2CA-Braga. AHY Employed by King’s College London; Honorary Consultant South London and Maudsley NHS Foundation Trust (NHS UK). Editor of Journal of Psychopharmacology and Deputy Editor, BJPsych Open. Paid lectures and advisory boards for the following companies with drugs used in affective and related disorders: Astrazenaca, Boehringer Ingelheim, Eli Lilly, LivaNova, Lundbeck, Sunovion, Servier, Livanova, Janssen, Allegan, Bionomics, Sumitomo Dainippon Pharma, COMPASS, Sage, Novartis, Neurocentrx. Principal Investigator in the Restore-Life VNS registry study funded by LivaNova. Principal Investigator on ESKETINTRD3004: “An Open-label, Long-term, Safety and Efficacy Study of Intranasal Esketamine in Treatment-resistant Depression.” Principal Investigator on “The Effects of Psilocybin on Cognitive Function in Healthy Participants”. Principal Investigator on “The Safety and Efficacy of Psilocybin in Participants with Treatment-Resistant Depression (P-TRD)”. UK Chief Investigator for Compass; COMP006 & COMP007 studies. UK Chief Investigator for Novartis MDD study MIJ821A12201. Grant funding (past and present): NIMH (USA); CIHR (Canada); NARSAD (USA); Stanley Medical Research Institute (USA); MRC (UK); Wellcome Trust (UK); Royal College of Physicians (Edin); BMA (UK); UBC-VGH Foundation (Canada); WEDC (Canada); CCS Depression Research Fund (Canada); MSFHR (Canada); NIHR (UK). Janssen (UK) EU Horizon 2020. No shareholdings in pharmaceutical companies. EV has received grants and served as consultant, advisor or CME speaker for the following entities: AB-Biotics, AbbVie, Adamed, Angelini, Biogen, Biohaven, Boehringer-Ingelheim, Celon Pharma, Compass, Dainippon Sumitomo Pharma, Ethypharm, Ferrer, Gedeon Richter, GH Research, Glaxo-Smith Kline, HMNC, Idorsia, Janssen, Lundbeck, Medincell, Merck, Novartis, Orion Corporation, Organon, Otsuka, Roche, Rovi, Sage, Sanofi-Aventis, Sunovion, Takeda, and Viatris, outside the submitted work.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1:

Virtual patient cases.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fagiolini, A., González-Pinto, A., Miskowiak, K.W. et al. Role of trazodone in treatment of major depressive disorder: an update. Ann Gen Psychiatry 22, 32 (2023). https://doi.org/10.1186/s12991-023-00465-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12991-023-00465-y

Keywords