- 1Psychiatry Department, Hospital Universitario Reina Sofía, Córdoba, Spain
- 2Family and Community Medicine, Centro de Salud Torreblanca, Seville, Spain
- 3Family and Community Medicine, Centro de Salud Alagón, Zaragoza, Spain
- 4Family and Community Medicine, Centro de Salud Huerta de los Frailes, Madrid, Spain
- 5Family and Community Medicine, Clínica Eliana, Torrent, Valencia, Spain
The management of patients with major depressive disorder (MDD) in primary care (PC) represents a significant challenge and a great opportunity for early and effective intervention. Primary care physicians, as first-line physicians, play a key role in the identification, diagnosis and initial treatment of depression, often being the first and sometimes the only point of contact for these patients with the healthcare system. In this context, the search for effective and well-tolerated therapeutic strategies is constant, and vortioxetine represents a good pharmacological option within the therapeutic armamentarium available to the PC physician. This article explores best practices in the management of MDD from the PC perspective, addressing the specific challenges faced by these professionals and examining the potential role of vortioxetine in the treatment of different patient profiles.
Introduction
Major depressive disorder (MDD) is characterized by a heterogeneous set of symptoms involving emotional, physical and cognitive impairment (1). The overall burden of MDD has increased in recent years, particularly since 2020, associated with the aftermath of the COVID-19 pandemic (2). According to data from the World Health Organization (WHO), it is estimated that 5% of all adults worldwide suffer depression (6% of women and 4% of men) and 5.7% of all adults over 60 years of age (3). With these data, primary care (PC) is fundamental in the detection, diagnosis, treatment and follow-up of major depressive disorder.
Complexity of the diagnosis of MDD in primary care
The diagnosis of depression in the PC setting presents significant challenges that require special consideration (Figure 1). The difficulty in establishing an early diagnosis is related at least in part to that fact that some patients with depressive disorder attending PC present cognitive or physical manifestations (pain), with no predominance of affective symptoms. This variability in the presentation of symptoms may lead to a delay in diagnosis and the start of the most adequate treatment, as well as to errors in diagnosis (4–6). In this regard, depression remains an underdiagnosed disorder in PC, with only approximately 40% of all patients receiving the necessary treatment (5). The fact that, according to different estimates, depression presents with complaints of psychological distress in 45% of the cases, with complaints of somatic discomfort (fatigue, joint pain or vague pain) in 36%, and organic comorbidities in 19%, means that emotional, behavioral, cognitive and somatic symptoms should be jointly assessed in diagnosing the disorder (7, 8).

Figure 1. Recommendations and issues to consider regarding the diagnosis of major depressive disorder (MDD) and joint management with a mental health specialist, from the primary care perspective. PC, Primary Care; HADS, Hospital Anxiety and Depression Scale; MADRS, Montgomery-Asberg Depression Rating Scale; PHQ-9, Patient Health Questionnaire-9.
To address this complexity, it is crucial to incorporate standardized assessment tools into clinical practice. Thus, there is a need to incorporate scales to objectify symptoms, suggesting the use of guiding questions in cases where, due to time or resource issues, the complete assessment tools cannot be used. Diagnostic scales such as the Hamilton Scale (9), the Montgomery-Asberg Rating Scale (MADRS) (10), the Patient Health Questionnaire (PHQ-9) (11), among others, may provide an objective assessment of the symptoms. However, the reality of the PC setting often makes it difficult to administer patient-reported outcome scales or questionnaires, which, although widely known by healthcare professionals, are not used due to time and resource constraints (12). However, some of the most widely used scales for the diagnosis of MDD require specialized training and are generally more specific for use in mental health settings. In primary care, simpler instruments are needed-tools that do not require specific training and can be self-administered by patients, such as the previously mentioned PHQ-9 and the Hospital Anxiety and Depression Scale (HADS) (13).
In addition to the assessment scales, continuous training of PC professionals in mental health is essential. In this regard, the general perception is that it should be greater in general practitioners in this area. Studies conducted in Spain have shown that 40.6% of the general practitioners considered their training in the management of patients with depression through the use of psychotropic drugs to be insufficient, and the vast majority (92.5%) considered their participation in training activities in this field to be necessary (14). These data are in line with similar analyses conducted at international level, evidencing that 25% of the general practitioners were not comfortable in being able to provide adequate treatment for patients with depression (15). This perception of insufficient training may negatively impact confidence and the management of depression in PC, even more so in a setting where a strong link must be established with the patient to ensure adherence to the care plan and to facilitate early and effective intervention (16). In addition, a substantial number of patients are reluctant to receive treatment for the management of depressive disorder, even after having accepted the diagnosis. Both the causes to which patients attribute the origin of the disorder, and negative beliefs about antidepressants, as well as fear of addiction or adverse effects, can account for such reluctance (17–20).
Management of MDD patients in PC through treatment with vortioxetine
Various categories of antidepressant drugs have been commonly used in the treatment of MDD, including selective serotonin reuptake inhibitors (SSRIs), serotonin norepinephrine reuptake inhibitors (SNRIs), and tricyclic antidepressants, among others (21). However, approximately half of all patients do not achieve remission of the depressive episode with first-line treatment, so there is considerable debate about its effectiveness (21). Thus, the search for new antidepressants that offer greater effectiveness and tolerability in the management of MDD is essential.
The multimodal antidepressant vortioxetine is a first-line treatment option that directly modulates different serotonin receptors and inhibits the serotonin transporter, affording efficacy in adults comparable to that of most other antidepressants (21–30). Positive results in controlled clinical trials and meta-analyses of these trials have been corroborated in real-life studies (31).
Vortioxetine has a unique multimodal mechanism of action. Its pharmacodynamic profile includes: 1) Inhibits serotonin transporters (SERT), increasing serotonin (5-HT) levels, though with lower transporter occupancy than most SSRIs; and 2) It acts directly on several serotonin (5-HT) receptor subtypes (32). Through its serotonergic actions, vortioxetine indirectly modulates other neurotransmitter systems, including norepinephrine, dopamine, acetylcholine, and histamine, particularly in brain regions relevant to mood and cognition (33). These pharmacodynamic properties are associated with antidepressant efficacy, improvement in cognitive symptoms, and a lower risk of certain side effects such as emotional blunting and sexual dysfunction compared to conventional SSRIs (32).
Key benefits and practical considerations in PC
Dosing flexibility
Treatment with vortioxetine allows dosing flexibility, since the dose can be increased to achieve complete functional recovery, without a proportional increase in adverse effects (Figure 2) (34, 35). Clinical trials and systematic reviews have shown a dose-response relationship with vortioxetine, in contrast to other antidepressants. In this regard, a dose of 20 mg/day is related to greater efficacy than a 10 mg/day dose, without increasing the adverse effects (34, 35). The assessment of tolerability at 15 days after treatment initiation and of response at 2–4 weeks is advised, with dose adjustments if needed (22). The half-life of vortioxetine facilitates treatment discontinuation with a lesser risk of withdrawal symptoms (6, 22, 32). Discontinuation symptoms (DS) with vortioxetine are rare, occurring in only 3% of patients who stopped treatment (36). DS is more likely when vortioxetine is stopped accidentally and without medical advice. The risk of DS is not related to age, sex, discontinuation method, dose, comorbidities, polytherapy, or treatment duration (36). This adaptation of treatment is a key point in the management of MDD with antidepressants, particularly from the perspective of PC, because it allows for avoiding frequent changes in drug. A high frequency of changes in medication may cause uncertainty and anxiety in patients, decrease confidence in the treatment and lead to lower long-term adherence, as well as create treatment resistance and modify the course of depression (20, 37, 38).

Figure 2. Recommendations and considerations regarding the management of patients with MDD in PC through treatment with vortioxetine. PC, Primary care; GAD, Generalized anxiety disorder; MDD, Major depressive disorder; VTX, Vortioxetine.
Recent analyses also suggest that higher doses of vortioxetine are associated with a more pronounced reduction in depressive symptoms, a faster onset of therapeutic effects, greater improvement in anxiety symptoms, increased effectiveness in patients with a history of trauma, and more substantial alleviation of specific symptoms such as anhedonia and anergy (39–43).
Benefit in the treatment of cognitive deficits
Vortioxetine has shown benefits in the treatment of cognitive deficits, which tend to be common in patients with depression, both in controlled clinical trials and in real-life studies (Figure 2) (31, 44–46). This is a relevant factor in the functional recovery of the patient, since some symptoms related to cognitive deficit (mainly attention, memory and processing speed) are residual symptoms that often persist in the remission phases of MDD (47). Specifically, this is an important aspect in the profile of young patients and those of working age, with high cognitive requirements.
Improvement of cognitive function is also a particularly relevant issue in the elderly population, with cognitive changes associated with age and/or with Alzheimer’s disease, where treatment with vortioxetine may have important therapeutic implications, given the need for effective therapies that address both the depressive symptoms and cognitive impairment (48, 49). In addition, different studies have addressed the treatment and improvement of cognitive function in specific patient groups, such as patients with MDD after stroke or with Parkinson’s disease (50, 51).
Benefit in terms of emotional blunting as an adverse effect of treatment with antidepressants
Emotional blunting is a common adverse effect reported by patients with MDD who are receiving antidepressant treatment, particularly SSRIs and SNRIs (52). This phenomenon is characterized by a reduced ability to experience emotions, both positive and negative; feelings of disengagement; and a decreased ability to respond emotionally. It is estimated that 40-60% of all patients treated with SSRIs or SNRIs experience some degree of emotional blunting (53). It has been identified as one of the most prominent reasons for drug discontinuation, and in response, clinicians often consider reducing the dose of antidepressant treatment or switching to another treatment in an attempt to control this effect (Figure 2) (53).
Based on real-life results, treatment with vortioxetine 5–20 mg effectively improves the emotional blunting associated with antidepressant treatment in patients with MDD and an inadequate response to SSRIs/SNRIs (54, 55). Such recovery is accompanied by improvement in the core of symptoms associated with depression, sleep duration and quality, motivation, and cognitive and global functioning of the patient (54, 55). Furthermore, the reduction in the severity of emotional blunting after vortioxetine initiation occurs rapidly and is sustained over time (54).
Tolerability and perception of treatment safety
Tolerability is one of the key aspects to ensure adherence to antidepressant treatment in MDD (20). Treatment with vortioxetine has a good tolerability profile, as clinical trials and real-life studies have shown that the incidence of adverse effects such as weight gain, insomnia and sexual dysfunction, and the cardiovascular safety profile, are comparable to placebo or other antidepressants (Figure 2) (24, 30, 31, 35). Broad clinical profile or vortioxetine and high level of safety are supported by evidence from randomized trials, post-marketing and case studies, including populations with epilepsy, elderly patients, people with dementia, and those with somatic comorbidities (39, 56–58). In fact, real-life data estimate a high acceptability and tolerability of treatment, with low discontinuation ratios, according to the conclusions of a recently published meta-analysis (31).
As discussed above, treatment with vortioxetine allows for the possibility of increasing the dose without a proportional increase in adverse effects (34, 35). A systematic review of clinical trials and real-life studies focused on vortioxetine indicated that the incidence of serious adverse effects was 2.9% for the 5–10 mg dose and 2.2% for the 15–20 mg dose (33). This is an important advantage, because the most commonly used second-generation antidepressants (citalopram, escitalopram, fluoxetine, paroxetine, sertraline, venlafaxine and mirtazapine) have an optimum balance of efficacy, tolerability and acceptability only in the lowest ranges of their doses approved in the Summary of Product Characteristics (59). Thus, it is estimated that the discontinuation rate of these drugs due to adverse effects increases sharply as the dosage is increased (59).
As an exception, it should be noted that treatment with vortioxetine 5–15 mg has been associated with a low risk of treatment-related sexual dysfunction, while the 20 mg dose was associated with a slight increase of this risk (22, 32).
Antidepressants have been reported to exert different effects on sleep architecture; in this regard, some treatments (e.g., SSRIs) can eventually lead to sleep disturbances, which may exacerbate the depressive symptoms (60, 61). This may help to create a vicious cycle, as it may lead to an increase in the dose of the antidepressant or the addition of drugs to improve sleep quality (62). In contrast, data on improvement of subjective sleep quality during treatment with vortioxetine, which does not usually cause insomnia or drowsiness, are found in the literature (63, 64).
Strategies in the case of lack of response to vortioxetine monotherapy
The response and remission rates in patients with MDD treated with vortioxetine in real life have been estimated to be 66.4% (95%CI = 51.2-81.5%) and 58% (95%CI = 48.9-67.1%), respectively. It is very important to assess patient adherence to therapy, because poor adherence has an impact on the recovery process and may increase the risk of relapse (Figure 2) (37, 38). In the event of lack of response to antidepressant treatment, the current guidelines on the management of MDD in PC recommend a number of strategies, including: i) increasing or optimizing the dose of the drug being used; ii) switching to another antidepressant; iii) combining antidepressants with different profiles; and iv) enhancing the antidepressant with other pharmacological agents (6). In addition, in depressive disorders refractory to treatment, referral to a mental health specialist should be considered (6).
There is growing evidence of the effectiveness of combining vortioxetine with other antidepressants or agents, mainly in patients in whom the therapeutic target has not been reached with ≥ 2 antidepressants. In general, the combination of vortioxetine and bupropion (a potent inhibitor of cytrochrome P4502D6) may be a useful and well-tolerated option, though it is underused in the PC setting. The association of vortioxetine with a slow metabolizer such as bupropion requires dose adjustment of both drugs, due to the potential incidence of certain side effects that do not appear when either drug is used alone (65, 66).
Mirtazapine is presented as a more attractive option for combined antidepressant therapy, being easy to use in the primary care setting (67). Its pharmacodynamic profile effectively complements most antidepressants, offering additional advantages (6). However, it is important to consider that mirtazapine involves an increased risk of weight gain and metabolic alterations (68). Therefore, careful and continuous assessment of its long-term tolerability is recommended in each patient in order to control potential cardiovascular risk factors.
Similarly, the addition of trazodone to vortioxetine therapy may be considered. Recent studies have demonstrated the strong clinical effectiveness of trazodone (69), and, when used at appropriate doses, this combination does not present a significant risk of serotonin syndrome. As regards vortioxetine augmentation therapy with other agents such as lithium salts or antipsychotics (practices more common in psychiatry than in PC), such treatments have been shown to be effective in two ways: on one hand, they improve resistant depressions, and on the other hand, they offer the advantage of including vortioxetine versus other antidepressants for the treatment of MDD with other comorbidities (66, 67).
Specific patient profiles
Patients with concomitant disorders or polymedicated individuals
Antidepressant therapies, particularly SSRIs and SNRIs, interact with other drugs, mainly through cytochrome P450 (CYP450) inhibition (70). This mechanism may lead to increased or decreased levels of other drugs in patients with concomitant diseases requiring drug treatment, causing toxicity. The above is particularly important in patients with MDD and cardiovascular diseases, renal and/or liver failure, diabetes or chronic pain (70–73). In these cases, vortioxetine would be recommended as the first option, since it offers advantages thanks to its pharmacological profile (6). Vortioxetine is metabolized in the body more simply than other antidepressants, mainly through CYP2D6, without affecting (inhibiting or inducing) the different cytochrome P450 isoenzymes involved, thus reducing potential interactions with other drugs (Figure 2) (32, 74). Clinical pharmacokinetic studies have shown that co-administering vortioxetine with strong CYP2D6 inhibitors such as bupropion, fluoxetine, or paroxetine leads to approximately a two-fold increase in vortioxetine exposure (AUC and Cmax) (75). Therefore, it is recommended to reduce the vortioxetine dose by half when used alongside strong CYP2D6 inhibitors to minimize the risk of concentration-dependent side effects. Specifically, in patients with cardiovascular comorbidity, vortioxetine has been shown to have no significant effect on heart rate or blood pressure (74). In patients with renal and/or liver failure, no dose adjustment of vortioxetine is required, unlike with other antidepressants (6, 76–78).
Patients over 65 years of age
As previously commented, vortioxetine shows significant improvement of outcomes in real-life and clinical trials in relation to the cognitive symptoms associated with MDD, which may be relevant in the elderly population. Depression in older patients has been linked to mild cognitive impairment and dementia, so treatment with a drug with procognitive effects may be beneficial (Figure 2) (79–81). Preliminary and specific studies on this patient profile demonstrated clinically significant improvement in the depressive symptoms, cognitive performance, functional recovery and quality of life of patients with MDD and cognitive impairment, including early-stage Alzheimer’s disease (49, 82).
Females of childbearing potential and in the perimenopausal phase
There are limited human data on the use of vortioxetine during pregnancy, but animal studies have shown reproductive toxicity. For this reason, the recommendation is to administer this medication during pregnancy only if the benefits outweigh the potential risks to the fetus (22). With regard to lactation, data from studies in animals show vortioxetine to be excreted in milk, and the same is expected to occur in humans. The Relative Infant Dose (RID) index for vortioxetine is low (less than 2%) so the risk of exposure of the child to significant doses of the drug received in milk is small (83). However, until more data are available, vortioxetine should be used with careful infant monitoring during breastfeeding. Menopause is characterized by a complex interaction of biological, psychological and social factors. The hormonal changes in menopause, particularly estrogen reduction, affect mood and mental health, and depression is an important concern due to its high prevalence in these patients (84, 85). In the case of patients with MDD and in transition to menopause, preliminary data have indicated that vortioxetine treatment not only significantly improves depression-related symptoms and indicators (with response and remission rates of 75% and 70.8%, respectively), but also improves menopause-related symptoms (Figure 2) (86).
Patients with chronic pain
Chronic pain is a state of stress in which depression becomes one of the most common psychiatric complications in the affected individual (87). Depression in patients with chronic pain is associated with a reduction in pain threshold, an increase in pain perception, and a poorer response to analgesics (87). In addition, the presence of pain is related to more severe depressive symptoms in patients, including sleep disturbances, increased anxiety, and cognitive deficit (88). Preliminary data obtained in routine clinical practice in patients treated for chronic pain and diagnosed with MDD who had not previously responded to other antidepressants show that vortioxetine significantly improves depressive and pain symptoms, with an adequate safety profile (Figure 2) (89). Recent studies have also demonstrated that vortioxetine can effectively treat neuropathic pain and has shown efficacy in the management of patients with burning mouth syndrome (90, 91).
Patients with MDD and generalized anxiety disorder
Comorbidity of MDD and generalized anxiety disorder (GAD) is common, and there is evidence that patients with depressive disorders are 11.7 times more at risk of developing GAD than patients without depressive symptoms (92–94). Furthermore, the comorbid presence of GAD increases the risk of relapse of the depressive disorder (Figure 2) (95). The multimodal mechanism of action of vortioxetine, at the level of the serotonergic pathways, could make vortioxetine a tool for the treatment of patients with MDD and generalized anxiety disorder (96, 97). In fact, the use of vortioxetine in real life in a subgroup of patients with MDD and comorbid GAD has shown significant improvement in global function, cognitive function, and depressive and anxiety-related symptoms (98). In addition, good tolerability is observed at the standard doses (5–20 mg) (98).
Primary care perspective of MDD management with vortioxetine in combination with non-pharmacological therapy
Psychotherapy enhances and complements drug therapy from the mildest stages of MDD (Figure 1) (6). However, the options for patient referral from PC to the psychotherapy services vary among the different regions (Autonomous Communities) of the country, and in general, in the context of the public healthcare system, there are no facilities for making such referrals. In the context of public health, few resources are available in terms of professionals specializing in psychology in PC, and their activity is limited (99). In fact, in Spain the rate is 5.71 clinical psychologists/100,000 inhabitants, with waiting times for the first visit of approximately 200 days (100). Demand from PC therefore would be aimed at securing greater coordination with the specialized services, since failure to do so may lead to a deficit in the detection of mental disorders, and particularly of MDD (101). Several proposals for the integration into PC teams of specialists in psychology have been developed in this regard - the results of which suggest that treatment is up to three times more effective than if patient management is only carried out by the general practitioner (102, 103). The main consequence of these integration programs is an increase in the percentage of patients who recover (102, 103).
Future research and clinical practice
Recent evidence highlights the efficacy and tolerability of vortioxetine in MDD and its potential benefits in cognitive symptoms. However, important gaps persist that require further research and focused enhancements.
First, most available data derive from randomized clinical trials conducted under conditions which may not fully represent real-world patient populations. Future studies should prioritize real-world research to better understand effectiveness and safety of vortioxetine across diverse clinical settings (31).
There is also a need to clarify the management of adverse events in special populations, such as those with complex psychiatric or neurological comorbidities (104). Finally, mechanistic studies exploring the drug’s multimodal effects could inform personalized treatment approaches and the development of combination therapies.
Addressing these gaps will help optimize the clinical use of vortioxetine and expand its therapeutic potential.
Conclusions
In conclusion, vortioxetine is a very valuable treatment option in the management of MDD in PC. Its multimodal mechanism of action offers significant advantages, including dosing flexibility, benefits in the treatment of cognitive deficits, and an improvement of the emotional blunting normally associated with other antidepressants. The good tolerability of vortioxetine, with a low incidence of adverse effects, favors treatment adherence. In addition, its effectiveness in the management of cognitive symptoms positions it as a particularly relevant option for patients of working age and older adults. Vortioxetine represents an important tool in the therapeutic armamentarium of the general practitioner, offering an alternative that can significantly improve the quality of life of patients with depression.
Data availability statement
The original contributions presented in the study are included in the article/supplementary material. Further inquiries can be directed to the corresponding author.
Author contributions
JÁA: Conceptualization, Writing – original draft, Writing – review & editing. VOD: Conceptualization, Writing – original draft, Writing – review & editing. GA: Conceptualization, Writing – original draft, Writing – review & editing. SL: Conceptualization, Writing – original draft, Writing – review & editing. VGM: Conceptualization, Writing – original draft, Writing – review & editing.
Funding
The author(s) declare that financial support was received for the research and/or publication of this article. This work was funded by Lundbeck. The funder was not involved in the study design, collection, analysis, interpretation of data, the writing of this article, or the decision to submit it for publication.
Acknowledgments
The authors thank Laura Prieto del Val and Alicia Subtil-Rodriguez of Evidenze Health España, S.L.U, for providing medical writing support (writing of the original draft, implementing changes in the manuscript after authors’ revisions, adapting the manuscript to journal requirements and submission to the journal), which was funded by Lundbeck Spain in accordance with Good Publication Practice (GPP3) guidelines.
Conflict of interest
VGM has received payment and honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events and support for attending meetings and/or travel from Esteve and Lundbeck. VOD has received grants from Schwabe and Esteve; consulting fees from Lundbeck; and payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events, and support for attending meetings and/or travel from Lundbeck, Neuraxpharm and Pfizer. JÁA has received honoraria for lectures and as advisory board member from Adamed, Esteve, Lundbeck and Johnson & Johnson.
The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Generative AI statement
The author(s) declare that no Generative AI was used in the creation of this manuscript.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
References
1. IsHak WW, James DM, Mirocha J, Youssef H, Tobia G, Pi S, et al. Patient-reported functioning in major depressive disorder. Ther Adv Chronic Dis. (2016) 7:160–9. doi: 10.1177/2040622316639769
2. COVID-19 Mental Disorders Collaborators. Global prevalence and burden of depressive and anxiety disorders in 204 countries and territories in 2020 due to the COVID-19 pandemic. Lancet. (2021) 398:1700–12. doi: 10.1016/S0140-6736(21)02143-7
3. WHO. Depressive disorder (depression). World Health Organization (2023). Available at: https://www.who.int/es/news-room/fact-sheets/detail/depression. (Accessed September 5, 2024).
4. Dumesnil H, Cortaredona S, Verdoux H, Sebbah R, Paraponaris A, and Verger P. General practitioners' choices and their determinants when starting treatment for major depression: a cross sectional, randomized case-vignette survey. PloS One. (2012) 7:e52429. doi: 10.1371/journal.pone.0052429
5. Martín-Agueda B, López-Muñoz F, Rubio G, Guerra JA, Silva A, and Alamo C. Management of depression in primary care: a survey of general practitioners in Spain. Gen Hosp Psychiatry. (2005) 27:305–12. doi: 10.1016/j.genhosppsych.2005.05.002
6. SEMERGEN. Guías Clínicas de Depresión (2023). Available online at: https://semergen.es/?seccion=biblioteca&subSeccion=detalleDocumento&idD=1212. (Accessed September 5, 2024).
7. APA. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition. Washington DC, USA: American Psychiatric Association Publishing. (2013).
8. López Chamón S. Depresión Mayor. SEMERGEN (2016), Spain: SEMERGEN. ISBN: 978-84-6087635-9978-84-6087635-92016.
9. Hamilton M. A rating scale for depression. J Neurol Neurosurg Psychiatry. (1960) 23:56–62. doi: 10.1136/jnnp.23.1.56
10. Montgomery SA and Asberg M. A new depression scale designed to be sensitive to change. Br J Psychiatry. (1979) 134:382–9. doi: 10.1192/bjp.134.4.382
11. Kroenke K, Spitzer RL, and Williams JB. The PHQ-9: validity of a brief depression severity measure. J Gen Intern Med. (2001) 16:606–13. doi: 10.1046/j.1525-1497.2001.016009606.x
12. Waheed A, Afridi AK, Rana M, Arif M, Barrera T, Patel F, et al. Knowledge and behavior of primary care physicians regarding utilization of standardized tools in screening and assessment of anxiety, depression, and mood disorders at a large integrated health system. J Prim Care Community Health. (2024) 15:21501319231224711. doi: 10.1177/21501319231224711
13. Zigmond AS and Snaith RP. The hospital anxiety and depression scale. Acta Psychiatr Scand. (1983) 67:361–70. doi: 10.1111/j.1600-0447.1983.tb09716.x
14. Latorre Postigo JM, López-Torres Hidalgo J, Montañés Rodríguez J, and Parra Delgado M. Percepción de la demanda y necesidades de formación en salud mental de los médicos de atención primaria. Atención Primaria. (2005) 36:85–92. doi: 10.1157/13076608
15. Grung I, Hjørleifsson S, Anderssen N, Bringedal B, Ruths S, and Hetlevik Ø. Norwegian general practitioners' perceptions of their depression care - a national survey. BMC Prim Care. (2024) 25:184. doi: 10.1186/s12875-024-02434-0
16. Torres Saldaña A, Ortiz Sánchez Y, Martínez Suárez H, Fernández Corrales YN, and Pacheco Ballester D. Factores asociados a la adherencia al tratamiento en pacientes con depresión. Multimed. (2019) 23:1–10.
17. Acosta F, Rodríguez L, and Cabrera B. Beliefs about depression and its treatments: associated variables and the influence of beliefs on adherence to treatment. Rev Psiquiatr Salud Ment. (2013) 6:86–92. doi: 10.1016/j.rpsm.2012.08.001
18. Acosta F, Rodríguez L, and Cabrera B. Creencias sobre la depresión y sus tratamientos: variables asociadas e influencia de las creencias en la adherencia. Rev Psiquiatría y Salud Mental. (2024) 53:86–92. doi: 10.1016/j.rpsm.2012.08.001
19. Hung CI. Factors predicting adherence to antidepressant treatment. Curr Opin Psychiatry. (2014) 27:344–9. doi: 10.1097/YCO.0000000000000086
20. Muhammad N, Ullah SR, Nagi TK, and Yousaf RA. Factors associated with non-adherence to anti-depressant medication in adults: A systematic review and meta-analysis. Cureus. (2023) 15:e37828. doi: 10.7759/cureus.37828
21. Cipriani A, Furukawa TA, Salanti G, Chaimani A, Atkinson LZ, Ogawa Y, et al. Comparative efficacy and acceptability of 21 antidepressant drugs for the acute treatment of adults with major depressive disorder: a systematic review and network meta-analysis. Lancet. (2018) 391:1357–66. doi: 10.1016/S0140-6736(17)32802-7
22. EMA. Summary of product characteristics brintellix (Vortioxetine). (2024). Available online at: https://www.ema.europa.eu/en/documents/product-information/brintellix-epar-product-information_en.pdf. (Accessed September 5, 2024).
23. Thase ME, Jacobsen PL, Hanson E, Xu R, Tolkoff M, and Murthy NV. Vortioxetine 5, 10, and 20 mg significantly reduces the risk of relapse compared with placebo in patients with remitted major depressive disorder: The RESET study. J Affect Disord. (2022) 303:123–30. doi: 10.1016/j.jad.2022.02.002
24. Zhang X, Cai Y, Hu X, Lu CY, Nie X, and Shi L. Systematic review and meta-analysis of vortioxetine for the treatment of major depressive disorder in adults. Front Psychiatry. (2022) 13:922648. doi: 10.3389/fpsyt.2022.922648
25. Zheng J, Wang Z, and Li E. The efficacy and safety of 10 mg/day vortioxetine compared to placebo for adult major depressive disorder: a meta-analysis. Afr Health Sci. (2019) 19:1716–26. doi: 10.4314/ahs.v19i1.48
26. Wagner G, Schultes MT, Titscher V, Teufer B, Klerings I, and Gartlehner G. Efficacy and safety of levomilnacipran, vilazodone and vortioxetine compared with other second-generation antidepressants for major depressive disorder in adults: A systematic review and network meta-analysis. J Affect Disord. (2018) 228:1–12. doi: 10.1016/j.jad.2017.11.056
27. Nishimura A, Aritomi Y, Sasai K, Kitagawa T, and Mahableshwarkar AR. Randomized, double-blind, placebo-controlled 8-week trial of the efficacy, safety, and tolerability of 5, 10, and 20 mg/day vortioxetine in adults with major depressive disorder. Psychiatry Clin Neurosci. (2018) 72:64–72. doi: 10.1111/pcn.2018.72.issue-2
28. Inoue T, Sasai K, Kitagawa T, Nishimura A, and Inada I. Randomized, double-blind, placebo-controlled study to assess the efficacy and safety of vortioxetine in Japanese patients with major depressive disorder. Psychiatry Clin Neurosci. (2020) 74:140–8. doi: 10.1111/pcn.12956
29. Inoue T, Nishimura A, Sasai K, and Kitagawa T. Randomized, 8-week, double-blind, placebo-controlled trial of vortioxetine in Japanese adults with major depressive disorder, followed by a 52-week open-label extension trial. Psychiatry Clin Neurosci. (2018) 72:103–15. doi: 10.1111/pcn.2018.72.issue-2
30. Thase ME, Mahableshwarkar AR, Dragheim M, Loft H, and Vieta E. A meta-analysis of randomized, placebo-controlled trials of vortioxetine for the treatment of major depressive disorder in adults. Eur Neuropsychopharmacol. (2016) 26:979–93. doi: 10.1016/j.euroneuro.2016.03.007
31. Li Z, Liu S, Wu Q, Li J, Yang Q, Wang X, et al. Effectiveness and safety of vortioxetine for the treatment of major depressive disorder in the real world: A systematic review and meta-analysis. Int J Neuropsychopharmacol. (2023) 26:373–84. doi: 10.1093/ijnp/pyad018
32. Krupa AJ, Wojtasik-Bakalarz K, and Siwek M. Vortioxetine - pharmacological properties and use in mood disorders. The current state of knowledge. Psychiatr Pol. (2023) 57:1109–26. doi: 10.12740/PP/OnlineFirst/151570
33. Salagre E, Grande I, Solé B, Sanchez-Moreno J, and Vieta E. Vortioxetine: A new alternative for the treatment of major depressive disorder. Rev Psiquiatr Salud Ment (Engl Ed). (2018) 11:48–59. doi: 10.1016/j.rpsm.2017.06.006
34. Wang P, Wang WW, Liu YQ, Li WQ, Hu JX, Su YA, et al. The dose-response relationship of vortioxetine on major depressive disorder: an umbrella review. Psychiatry Res. (2024) 340:116118. doi: 10.1016/j.psychres.2024.116118
35. Baldwin DS, Chrones L, Florea I, Nielsen R, Nomikos GG, Palo W, et al. The safety and tolerability of vortioxetine: Analysis of data from randomized placebo-controlled trials and open-label extension studies. J Psychopharmacol. (2016) 30:242–52. doi: 10.1177/0269881116628440
36. Siwek M, Chrobak AA, Gorostowicz A, Krupa AJ, and Dudek D. Withdrawal symptoms following discontinuation of vortioxetine-retrospective chart review. Pharm (Basel). (2021) 14(5):451. doi: 10.3390/ph14050451
37. Del Pino-Sedeño T, Infante-Ventura D, Hernández-González D, González-Hernández Y, González de León B, Rivero-Santana A, et al. Sociodemographic and clinical predictors of adherence to antidepressants in depressive disorders: a systematic review with a meta-analysis. Front Pharmacol. (2024) 15:1327155. doi: 10.3389/fphar.2024.1327155
38. Stahl ST, Kincman J, Karp JF, and Anne Gebara M. Psychosocial interventions to improve adherence in depressed and anxious older adults prescribed antidepressant pharmacotherapy: a scoping review. Ther Adv Psychopharmacol. (2023) 13:20451253231212322. doi: 10.1177/20451253231212322
39. Baldwin DS, Florea I, Jacobsen PL, Zhong W, and Nomikos GG. A meta-analysis of the efficacy of vortioxetine in patients with major depressive disorder (MDD) and high levels of anxiety symptoms. J Affect Disord. (2016) 206:140–50. doi: 10.1016/j.jad.2016.07.015
40. Cao B, Park C, Subramaniapillai M, Lee Y, Iacobucci M, Mansur RB, et al. The efficacy of vortioxetine on anhedonia in patients with major depressive disorder. Front Psychiatry. (2019) 10:17. doi: 10.3389/fpsyt.2019.00017
41. Christensen MC, Florea I, Loft H, and McIntyre RS. Efficacy of vortioxetine in patients with major depressive disorder reporting childhood or recent trauma. J Affect Disord. (2020) 263:258–66. doi: 10.1016/j.jad.2019.11.074
42. Christensen MC, McIntyre RS, Adair M, Florea I, Loft H, and Fagiolini A. Clinical benefits of vortioxetine 20 mg/day in patients with major depressive disorder. CNS Spectr. (2023) 28:693–701. doi: 10.1017/S1092852923002249
43. Watanabe K, Fujimoto S, Marumoto T, Kitagawa T, Ishida K, Nakajima T, et al. Therapeutic potential of vortioxetine for anhedonia-like symptoms in depression: A post hoc analysis of data from a clinical trial conducted in Japan. Neuropsychiatr Dis Treat. (2022) 18:363–73. doi: 10.2147/NDT.S340281
44. Cumbo E, Cumbo S, Torregrossa S, and Migliore D. Treatment effects of vortioxetine on cognitive functions in mild alzheimer's disease patients with depressive symptoms: A 12 month, open-label, observational study. J Prev Alzheimers Dis. (2019) 6:192–7. doi: 10.14283/jpad.2019.24
45. Bennabi D, Haffen E, and Van Waes V. Vortioxetine for cognitive enhancement in major depression: from animal models to clinical research. Front Psychiatry. (2019) 10:771. doi: 10.3389/fpsyt.2019.00771
46. Huang IC, Chang TS, Chen C, and Sung JY. Effect of vortioxetine on cognitive impairment in patients with major depressive disorder: A systematic review and meta-analysis of randomized controlled trials. Int J Neuropsychopharmacol. (2022) 25:969–78. doi: 10.1093/ijnp/pyac054
47. Bortolato B, Carvalho AF, and McIntyre RS. Cognitive dysfunction in major depressive disorder: a state-of-the-art clinical review. CNS Neurol Disord Drug Targets. (2014) 13:1804–18. doi: 10.2174/1871527313666141130203823
48. Burke AD, Goldfarb D, Bollam P, and Khokher S. Diagnosing and treating depression in patients with alzheimer's disease. Neurol Ther. (2019) 8:325–50. doi: 10.1007/s40120-019-00148-5
49. Bishop MM, Fixen DR, Linnebur SA, and Pearson SM. Cognitive effects of vortioxetine in older adults: a systematic review. Ther Adv Psychopharmacol. (2021) 11:20451253211026796. doi: 10.1177/20451253211026796
50. Gamberini G, Masuccio FG, Ferriero G, Cattaneo D, and Solaro C. Safety and efficacy of vortioxetine on depressive symptoms and cognition in post-stroke patients: a pilot study. J Affect Disord. (2021) 286:108–9. doi: 10.1016/j.jad.2021.02.075
51. Santos García D, Alonso Losada MG, Cimas Hernando I, Cabo López I, Yáñez Baña R, Alonso Redondo R, et al. Vortioxetine improves depressive symptoms and cognition in parkinson's disease patients with major depression: an open-label prospective study. Brain Sci. (2022) 12(11):1466. doi: 10.3390/brainsci12111466
52. Christensen MC, Ren H, and Fagiolini A. Emotional blunting in patients with depression. Part I: clinical characteristics. Ann Gen Psychiatry. (2022) 21:10. doi: 10.1186/s12991-022-00387-1
53. Ma H, Cai M, and Wang H. Emotional blunting in patients with major depressive disorder: A brief non-systematic review of current research. Front Psychiatry. (2021) 12:792960. doi: 10.3389/fpsyt.2021.792960
54. Christensen MC, Canellas F, Loft H, and Montejo ÁL. Effectiveness of vortioxetine for the treatment of emotional blunting in patients with major depressive disorder experiencing inadequate response to SSRI/SNRI monotherapy in Spain: results from the COMPLETE study. Neuropsychiatr Dis Treat. (2024) 20:1475–89. doi: 10.2147/NDT.S473056
55. Fagiolini A, Florea I, Loft H, and Christensen MC. Effectiveness of Vortioxetine on Emotional Blunting in Patients with Major Depressive Disorder with inadequate response to SSRI/SNRI treatment. J Affect Disord. (2021) 283:472–9. doi: 10.1016/j.jad.2020.11.106
56. Furutani N and Nagoshi Y. Vortioxetine as an alternative treatment for somatic symptom disorder: case report. Front Psychiatry. (2024) 15:1496072. doi: 10.3389/fpsyt.2024.1496072
57. Siwek M, Gorostowicz A, Bosak M, and Dudek D. Case report: vortioxetine in the treatment of depressive symptoms in patients with epilepsy-case series. Front Pharmacol. (2022) 13:852042. doi: 10.3389/fphar.2022.852042
58. Zhang Y, Sun S, and Ning Y. Post-marketing safety surveillance of vortioxetine hydrobromide: a pharmacovigilance study leveraging FAERS database. Front Psychiatry. (2025) 16. doi: 10.3389/fpsyt.2025.1532803
59. Furukawa TA, Cipriani A, Cowen PJ, Leucht S, Egger M, and Salanti G. Optimal dose of selective serotonin reuptake inhibitors, venlafaxine, and mirtazapine in major depression: a systematic review and dose-response meta-analysis. Lancet Psychiatry. (2019) 6:601–9. doi: 10.1016/S2215-0366(19)30217-2
60. Hutka P, Krivosova M, Muchova Z, Tonhajzerova I, Hamrakova A, Mlyncekova Z, et al. Association of sleep architecture and physiology with depressive disorder and antidepressants treatment. Int J Mol Sci. (2021) 22(3):1333. doi: 10.3390/ijms22031333
61. Steiger A and Pawlowski M. Depression and sleep. Int J Mol Sci. (2019) 20(3):607. doi: 10.3390/ijms20030607
62. Wichniak A, Wierzbicka A, Walęcka M, and Jernajczyk W. Effects of antidepressants on sleep. Curr Psychiatry Rep. (2017) 19:63. doi: 10.1007/s11920-017-0816-4
63. Cao B, Park C, Rosenblat JD, Chen Y, Iacobucci M, Subramaniapillai M, et al. Changes in sleep predict changes in depressive symptoms in depressed subjects receiving vortioxetine: An open-label clinical trial. J Psychopharmacol. (2019) 33:1388–94. doi: 10.1177/0269881119874485
64. Liguori C, Ferini-Strambi L, Izzi F, Mari L, Manfredi N, D'Elia A, et al. Preliminary evidence that vortioxetine may improve sleep quality in depressed patients with insomnia: a retrospective questionnaire analysis. Br J Clin Pharmacol. (2019) 85:240–4. doi: 10.1111/bcp.13772
65. Chen G, Højer AM, Areberg J, and Nomikos G. Vortioxetine: clinical pharmacokinetics and drug interactions. Clin Pharmacokinet. (2018) 57:673–86. doi: 10.1007/s40262-017-0612-7
66. Kotzalidis GD, Lombardozzi G, Matrone M, Amici E, Perrini F, Cuomo I, et al. Vortioxetine vs. Other antidepressants in patients with major depressive episode with or without substance use disorder. Curr Neuropharmacol. (2021) 19:2296–307. doi: 10.2174/1570159X19666210113150123
67. Aragonès E, Roca M, Mora F, Zarco J, Armenteros L, García-Portilla M, et al. Abordaje compartido de la depresión: Consenso multidisciplinar. Spain: EUROMEDICE Ediciones Médicas S.L. (2018).
68. Puzhko S, Schuster T, Barnett TA, Renoux C, Munro K, Barber D, et al. Difference in patterns of prescribing antidepressants known for their weight-modulating and cardiovascular side effects for patients with obesity compared to patients with normal weight. J Affect Disord. (2021) 295:1310–8. doi: 10.1016/j.jad.2021.08.018
69. Dudek D, Chrobak AA, Krupa AJ, Gorostowicz A, Gerlich A, Juryk A, et al. TED-trazodone effectiveness in depression: a naturalistic study of the effeciveness of trazodone in extended release formulation compared to SSRIs in patients with a major depressive disorder. Front Pharmacol. (2023) 14:1296639. doi: 10.3389/fphar.2023.1296639
70. Low Y, Setia S, and Lima G. Drug-drug interactions involving antidepressants: focus on desvenlafaxine. Neuropsychiatr Dis Treat. (2018) 14:567–80. doi: 10.2147/NDT.S157708
71. Ereshefsky L, Jhee S, and Grothe D. Antidepressant drug-drug interaction profile update. Drugs R D. (2005) 6:323–36. doi: 10.2165/00126839-200506060-00002
72. Castaldelli-Maia JM, Hofmann C, Chagas ACP, Liprandi AS, Alcocer A, Andrade LH, et al. Major cardiac-psychiatric drug-drug interactions: a systematic review of the consistency of drug databases. Cardiovasc Drugs Ther. (2021) 35:441–54. doi: 10.1007/s10557-020-06979-x
73. Chen Y and Ding L. Potential drug-drug interactions in outpatients with depression of a psychiatry department. Saudi Pharm J. (2023) 31:207–13. doi: 10.1016/j.jsps.2022.12.004
74. Bordet C, Rousseau V, Montastruc F, and Montastruc JL. QT prolongation and vortioxetine: a post-marketing study and comparison with other serotonin reuptake inhibitors. Psychopharmacol (Berl). (2020) 237:1245–7. doi: 10.1007/s00213-020-05461-8
75. Spina E and Santoro V. Drug interactions with vortioxetine, a new multimodal antidepressant. Riv Psichiatr. (2015) 50:210–5. doi: 10.1708/2040.22160
76. Menon V, Ransing R, and Praharaj SK. Management of psychiatric disorders in patients with hepatic and gastrointestinal diseases. Indian J Psychiatry. (2022) 64:S379–s93. doi: 10.4103/indianjpsychiatry.indianjpsychiatry_18_22
77. Mauri MC, Fiorentini A, Paletta S, and Altamura AC. Pharmacokinetics of antidepressants in patients with hepatic impairment. Clin Pharmacokinet. (2014) 53:1069–81. doi: 10.1007/s40262-014-0187-5
78. Zhu N, Lisinski A, Lagerberg T, Johnell K, Xu H, Carrero JJ, et al. Kidney function and prescribed dose in middle-aged and older patients starting selective serotonin reuptake inhibitors. Pharmacoepidemiol Drug Saf. (2022) 31:1091–101. doi: 10.1002/pds.v31.10
79. Alexopoulos GS. Mechanisms and treatment of late-life depression. Transl Psychiatry. (2019) 9:188. doi: 10.1038/s41398-019-0514-6
80. Aziz R and Steffens D. Overlay of late-life depression and cognitive impairment. Focus (Am Psychiatr Publ). (2017) 15:35–41. doi: 10.1176/appi.focus.20160036
81. Gastó C. ¿En qué consiste el efecto procognitivo de los antidepresivos? Psiquiatría Biológica. (2016) 23:34–9.
82. Christensen MC, Schmidt SN, and Grande I. Effectiveness of vortioxetine in patients with major depressive disorder and early-stage dementia: The MEMORY study. J Affect Disord. (2023) 338:423–31. doi: 10.1016/j.jad.2023.06.024
83. Hale TW and Krutsch K. Hale's Medications & Mothers' Milk 2023: A Manual of Lactational Pharmacology. New York, USA: Springer Publishing Company, Incorporated (2022).
84. Freeman EW, Sammel MD, Boorman DW, and Zhang R. Longitudinal pattern of depressive symptoms around natural menopause. JAMA Psychiatry. (2014) 71:36–43. doi: 10.1001/jamapsychiatry.2013.2819
85. de Kruif M, Spijker AT, and Molendijk ML. Depression during the perimenopause: A meta-analysis. J Affect Disord. (2016) 206:174–80. doi: 10.1016/j.jad.2016.07.040
86. Freeman MP, Cheng LJ, Moustafa D, Davies A, Sosinsky AZ, Wang B, et al. Vortioxetine for major depressive disorder, vasomotor, and cognitive symptoms associated with the menopausal transition. Ann Clin Psychiatry. (2017) 29:249–57. doi: 10.1177/1040123717029004
87. Gerrits M, van Oppen P, van Marwijk HWJ, Penninx B, and van der Horst HE. Pain and the onset of depressive and anxiety disorders. Pain. (2014) 155:53–9. doi: 10.1016/j.pain.2013.09.005
88. Bair MJ, Robinson RL, Katon W, and Kroenke K. Depression and pain comorbidity: a literature review. Arch Intern Med. (2003) 163:2433–45. doi: 10.1001/archinte.163.20.2433
89. Folch Ibáñez J, Vargas Domingo M, Coma Alemany J, Callao Sánchez R, and Guitart Vela J. Effectiveness of vortioxetine in patients with major depressive disorder associated with chronic pain: an observational study in a spanish population. Pain Ther. (2024) 13:621–35. doi: 10.1007/s40122-024-00597-3
90. Adamo D, Pecoraro G, Coppola N, Calabria E, Aria M, and Mignogna M. Vortioxetine versus other antidepressants in the treatment of burning mouth syndrome: An open-label randomized trial. Oral Dis. (2021) 27:1022–41. doi: 10.1111/odi.13602
91. Eliaçık S and Erdogan Kaya A. Vortioxetine treatment for neuropathic pain in major depressive disorder: a three-month prospective study. Front Neurol. (2024) 15:1398417. doi: 10.3389/fneur.2024.1398417
92. Zhou Y, Cao Z, Yang M, Xi X, Guo Y, Fang M, et al. Comorbid generalized anxiety disorder and its association with quality of life in patients with major depressive disorder. Sci Rep. (2017) 7:40511. doi: 10.1038/srep40511
93. Saha S, Lim CCW, Cannon DL, Burton L, Bremner M, Cosgrove P, et al. Co-morbidity between mood and anxiety disorders: A systematic review and meta-analysis. Depress Anxiety. (2021) 38:286–306. doi: 10.1002/da.23113
94. McGrath JJ, Lim CCW, Plana-Ripoll O, Holtz Y, Agerbo E, Momen NC, et al. Comorbidity within mental disorders: a comprehensive analysis based on 145–990 survey respondents from 27 countries. Epidemiol Psychiatr Sci. (2020) 29:e153. doi: 10.1017/S2045796020000633
95. Buckman JEJ, Underwood A, Clarke K, Saunders R, Hollon SD, Fearon P, et al. Risk factors for relapse and recurrence of depression in adults and how they operate: A four-phase systematic review and meta-synthesis. Clin Psychol Rev. (2018) 64:13–38. doi: 10.1016/j.cpr.2018.07.005
96. Sanchez C, Asin KE, and Artigas F. Vortioxetine, a novel antidepressant with multimodal activity: review of preclinical and clinical data. Pharmacol Ther. (2015) 145:43–57. doi: 10.1016/j.pharmthera.2014.07.001
97. Gonda X, Sharma SR, and Tarazi FI. Vortioxetine: a novel antidepressant for the treatment of major depressive disorder. Expert Opin Drug Discov. (2019) 14:81–9. doi: 10.1080/17460441.2019.1546691
98. Almeida SS, Christensen MC, Simonsen K, and Adair M. Effectiveness of vortioxetine in patients with major depressive disorder and co-morbid generalized anxiety disorder in routine clinical practice: A subgroup analysis of the RELIEVE study. J Psychopharmacol. (2023) 37:279–88. doi: 10.1177/02698811221132468
99. García-Haro J and Fernández-Briz N. Necesidad de criterios específicos para la derivación a psicoterapia: una propuesta. SEMERGEN - Medicina Familia. (2015) 41:214–20. doi: 10.1016/j.semerg.2014.01.011
100. Bueno Izquierdo M, Borràs Borràs M, Izquierdo Fuentes MT, and Fornós C. Discussion on the ability of primary care to meet the demand for healthcare due to the psychological effects derived from the COVID-19 pandemic. Aten Primaria. (2022) 54:102147. doi: 10.1016/j.aprim.2021.102147
101. Alonso Gómez R, Lorenzo Reina L, Flores Méndez I, Martín García J, and García Briñol L. The clinical psychologist in health centres. A joint work between primary care and mental health. Aten Primaria. (2019) 51:310–3. doi: 10.1016/j.aprim.2018.08.012
102. Cano-Vindel A, Ruiz-Rodríguez P, Moriana JA, Medrano LA, González-Blanch C, Aguirre E, et al. Improving access to psychological therapies in Spain: from IAPT to psicap. Psicothema. (2022) 34:18–24. doi: 10.7334/psicothema2021.113
103. Wakefield S, Kellett S, Simmonds-Buckley M, Stockton D, Bradbury A, and Delgadillo J. Improving Access to Psychological Therapies (IAPT) in the United Kingdom: A systematic review and meta-analysis of 10-years of practice-based evidence. Br J Clin Psychol. (2021) 60:1–37. doi: 10.1111/bjc.12259
Keywords: major depressive disorder, primary care, vortioxetine, antidepressants, cognitive deficits, emotional blunting
Citation: Alcalá JÁ, Olmo Dorado V, Arilla Herrera GdP, López Chamón S and Gasull Molinera V (2025) Optimizing the diagnosis and treatment of depression in primary care: the emerging role of vortioxetine treatment. Front. Psychiatry 16:1568777. doi: 10.3389/fpsyt.2025.1568777
Received: 04 February 2025; Accepted: 15 May 2025;
Published: 01 July 2025.
Edited by:
Matej Stuhec, University of Maribor, SloveniaReviewed by:
Marcin Siwek, Jagiellonian University, PolandMariacristina Mazzitelli, Texas Tech University Health Sciences Center, United States
Copyright © 2025 Alcalá, Olmo Dorado, Arilla Herrera, López Chamón and Gasull Molinera. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: José Ángel Alcalá, amFhbGNhbGFwYXJ0ZXJhQGNvbWNvcmRvYmEuY29t