Next Article in Journal
Steroid-Sparing Effect of Tocilizumab and Methotrexate in Patients with Polymyalgia Rheumatica: A Retrospective Cohort Study
Next Article in Special Issue
Telehealth and Screening Strategies in the Diagnosis and Management of Glaucoma
Previous Article in Journal
Evaluation of Strategies to Fight COVID-19: The French Paradigm
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Psychopharmacological Treatment, Intraocular Pressure and the Risk of Glaucoma: A Review of Literature

1
Neuroscience Department, Discipline of Psychiatry, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
2
Department of Psychiatry, ‘Prof. Dr. Alexandru Obregia’ Clinical Hospital of Psychiatry, 041914 Bucharest, Romania
3
Department of Psychiatry and Psychology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
4
Service d’Ophtalmologie, Centre Hospitalier ‘Rene Dubos’, 95300 Pontoise, France
5
Department of Stress Research and Prophylaxis, ‘Prof. Dr. Alexandru Obregia’ Clinical Hospital of Psychiatry, 041914 Bucharest, Romania
6
Romanian Academy of Medical Sciences, 927180 Bucharest, Romania
7
Department of Medical Psychology, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
*
Authors to whom correspondence should be addressed.
J. Clin. Med. 2021, 10(13), 2947; https://doi.org/10.3390/jcm10132947
Submission received: 2 June 2021 / Revised: 25 June 2021 / Accepted: 29 June 2021 / Published: 30 June 2021
(This article belongs to the Special Issue Going for Gaps in Glaucoma)

Abstract

:
Through the years, the available psychopharmacological treatments have expanded with numerous new drugs. Besides weight gain, gastro-intestinal problems or Parkinson-like symptoms, ocular adverse effects of psychiatric drugs have been reported. These adverse effects are not common, but can be dangerous for the patient. This review summarises the current knowledge on the risk of raised intraocular pressure and glaucoma entailed by psychopharmacological treatment. Also, it provides updated data for clinicians involved in the treatment of patients with glaucoma or glaucoma risk factors. For this purpose, we performed an extensive literature search in the PubMed database using specific terms. Selective serotonin and noradrenaline reuptake inhibitors are the best evidenced as having no association with glaucoma. Antipsychotics, and especially first generation, seem to have no correlation with an increased intraocular pressure and therefore possibly with a risk of glaucoma, although a special attention should be paid when using ziprasidone. Tricyclic antidepressants, benzodiazepines and topiramate should be avoided in patients diagnosed with glaucoma or at risk. Clinicians should be aware of the possible psychotropic drug induced glaucoma and monitor at risk patients closely in order to prevent this condition. Irrespective of the psychopharmacological regimen taken into consideration, the glaucoma patient should be under the strict supervision of the ophthalmologist.

1. Introduction

Glaucoma represents a heterogeneous group of chronically progressive neurodegenerative bilateral diseases of the optic nerve, clinically characterized by optical neuropathy, resulting in retinal ganglion cell death, optic nerve head cupping, and associated specific loss of the visual field [1,2,3]. The aetiology of the disease is considered to be multifactorial [4], while the clinical picture can differ, with a substantial risk of associated blindness, especially in adults over the age of 60 [5].
Studies have shown that in Europe glaucoma occurs in 2.93% of people aged 40 to 80, reaching 10% at the age of over 90 [6]. Several types of glaucoma are described, which form a group of eye diseases and are the main cause of permanent blindness worldwide. [7,8]. Based on the mechanism by which aqueous outflow is impaired with respect to the anterior chamber configuration, the disease is typically divided into 2 basic subtypes: open angle and angle closure [9,10]. Clinical presentation of open-angle glaucoma (OAG) includes mainly chronic, slow and irreversible loss of peripheral vision, ultimately leading to blindness. Because of the gradual and insidious development, more than 50% of patients are unaware of their condition (known as ‘the sneak thief of sight’), especially due to the pattern of visual field loss that spares the central vision until advanced stages [9]; therefore, periodic ophthalmologic evaluation is important [11,12]. On the other hand, acute angle-closure glaucoma (AACG) represents a dramatic urgent event that is caused by the sudden increase in the intraocular pressure (IOP) due to occlusion of the trabecular meshwork by the peripheral iris (in predisposed eyes with a narrow iridocorneal angle), obstructing aqueous outflow, that can lead to a potential risk of rapid blinding. ACG patients usually present more acute symptoms such as hyperaemia, teary, and painful eyes, sudden blurring of vision, or halos around lights secondary to corneal oedema from a sudden rise in IOP. Increased IOP is responsible for additional symptoms such as headache, nausea, and vomiting [9]. It must also be kept in mind that a narrow angle can be asymptomatic in the absence of a predisposing factor for angle-closure (e.g., pupillary dilatation) or that there are cited cases of insidious angle-closure glaucoma, which tends to be more visually destructive subtypes [9].
Above all, we have to make a firm distinction between acute angle-closure and angle-closure glaucoma. The major difference between the two entities is the absence and presence of optic nerve damage and visual field defects, respectively, especially when a specific treatment is rapidly received by the patient. The notion of glaucoma is under discussion when an optic neuropathy with elements specific to glaucoma damage occurs.
The scientific literature identifies several local and systemic risk factors associated with the development and progression of glaucoma [13]. In the case of OAG the local risk factors are represented by IOP (the key modifiable factor), family history of primary OAG, intraocular anti-VEGF (vascular endothelial growth factor) therapy, decreased thickness of the central region of the cornea, pre-existing myopia (low–moderate and high), low intraocular vascular pressure, optic nerve pathology, visual field changes, disc haemorrhage, and pseudo-exfoliation. Systemic risk factors include cardiovascular diseases, diabetes mellitus, dyslipidaemia, cerebral stroke, steroid treatment, arterial hypertension, with old age and male gender acting as additional risks [8,10,14]. Concerning the rapid progression of glaucoma, the most important favouring factors are considered to be high IOP and cardiovascular diseases [15].
On the other hand, the risk factors for developing ACG mentioned in the literature are the following: age (62 years being the average age at presentation), gender (more commonly female), race (Asian descent), family history, hyperopic eyes, short eyes. Those at risk for ACG with packed angle configuration can develop an attack exacerbated by mydriasis either spontaneously (primary) or pharmacologically (secondary) [9,10].
Medication represents a distinct risk for glaucoma. Corticosteroids are the most common cause of open-angle glaucoma (OAG), but several non-steroidal anti-inflammatory agents can also lead to OAG [16]. Regarding ACG, the list of potentially risky drugs includes antidepressants, such as selective serotonin reuptake inhibitors (SSRIs), tricyclic antidepressants and monoamine oxidase inhibitors, antipsychotic drugs, antihistaminic medication, antiparkinsonian agents, anticonvulsants (e.g., topiramate), mydriatic agents, sympathomimetic drops, antispasmodic drugs, and botulinum toxin.
Among the two types of glaucoma, ACG represents a pathology of interest concerning psychiatric treatment. Medications with anticholinergic effects could induce a precipitation of iridocorneal angle closure in patients with predisposition via mydriasis which is the primary pathogenic mechanism in the appearance of glaucoma in psychiatric patients [17,18] This rapid onset of the angle obstruction produces an imbalance between the production and drainage of aqueous humour in the anterior chamber, thus an accumulation of liquid and the increase of IOP. Other presumed involved mechanisms in the development of glaucoma are the anterior dislocation of the iris and lens or the inflammation of the ciliary body [7].
The challenges presented by glaucoma in psychiatry are bidirectional: on the one hand, patients with risk factor for angle-closure may develop clinical symptoms (eye pain, visual changes, headache) as a direct side effect of the psychiatric treatment. In case of patients with non-diagnosed OAG the raise in IOP can lead to severe aggravation of the disease, which can pass unnoticed until advanced stages [19]. On the other hand, glaucoma patients may exhibit a wide array of psychiatric symptoms, as a consequence of progression of vision loss, such as depression [20], anxiety [21] and insomnia [22].
Therefore, a minimal and specific screening before initiating a psychiatric treatment should be kept in mind, at least for the patients who have risk factors to develop OAG (IOP measure, fundus examination, +/− optic coherence tomography—OCT of the optic nerve head) or ACG (IOP measure, gonioscopy, +/− OCT of the optic nerve head) depending on the mechanism of action of the class of drugs used. Nevertheless, a good multidisciplinary collaboration between the psychiatrist and the ophthalmologist is strongly recommended, especially in complex cases.
Given the importance of glaucoma in influencing the choice of psychiatric drugs, we propose to review the main psychiatric therapeutic agents and their potential effects on glaucoma occurrence. Secondly, our aim is to provide a useful review of current data for clinicians facing dilemmas regarding the pharmacology treatment of psychiatric disorders in patients with glaucoma or glaucoma risk factors.

2. Methodology

We conducted an extensive literature search in the PubMed database from 1977 until 2021. The search was performed during March-May 2021 using the following terms: ‘psychotropic medication’, ‘SSRI’, ‘citalopram’, ‘escitalopram’, ‘paroxetine’, ‘fluvoxamine’, ‘fluoxetine’, ‘sertraline’, ‘SNRI’, ‘duloxetine’, ‘venlafaxine’, ‘tricyclic antidepressants’, ‘clomipramine’, ‘amitriptyline’, ‘imipramine’, ‘doxepin’, ‘desipramine’, ‘nortriptyline’ ‘NDRI’, ‘bupropion’, ‘benzodiazepine’, ‘alprazolam’, ‘diazepam’ ‘antipsychotics’, ‘haloperidol’, ‘ziprasidone’, ’risperidone’, ‘olanzapine’, ‘quetiapine’, ‘clozapine’, ‘topiramate’ cross-referenced with ‘glaucoma’ and ‘intraocular pressure’. We selected only articles written in English and based on clinical reports. After review of title, keywords and abstract, we retrieved 128 articles. Following removal of duplicates, full text assessment and then screening of the remaining articles for relevant studies that could be included in our paper, we finally included 90 articles divided as it follows: SSRI-7, citalopram-2, escitalopram-2, paroxetine-5, fluvoxamine-1, sertraline-1, SNRI-2, duloxetine-2, venlafaxine-5, tricyclic antidepressants-1, clomipramine-1, amitriptyline-1, imipramine-1, bupropion-4, benzodiazepine-2, diazepam-1, antipsychotics-8, haloperidol-3, risperidone-1, topiramate-40.

3. Antidepressants

3.1. Selective Serotonin Reuptake Inhibitors (SSRIs) and Serotonin and Noradrenaline Reuptake Inhibitors (SNRIs)

SSRIs and SNRIs are currently the first line drugs for the treatment of depression according to international guidelines [23,24]. SSRIs and SNRIs are the most prescribed drugs for depression and have the best overall tolerability and safety among all antidepressants. Also, these drugs are indicated as first choice for the treatment of anxiety, post-traumatic and obsessive compulsive disorders [25,26,27].
Since the discovery in 1974 of the first member of SSRI class, namely fluoxetine, continuing with sertraline, paroxetine, fluvoxamine, citalopram and escitalopram, these drugs have revolutionised the pharmacological therapy of depression. SSRIs mechanism of action implies a selective blockage of the reuptake of serotonin in the synaptic gap, therefore increasing the availability of the neurotransmitter and normalising the function of synapses. Until now seven families of serotonin receptors (5HT1-5HT7) have been described as having a diffuse localisation, including eye structures. Experimental studies have determined that 5HT1A, 5HT2A/2C and 5HT7 are located in the iris-ciliary body complex. Stimulation of 5HT1A receptor reduces the IOP through the reduction of aqueous humour, while 5HT2A/2C receptors increase IOP by stimulation of the ciliary body blood flow, therefore they enhance the production of aqueous humour. 5HT7 receptors are responsible for mydriasis through the relaxation of the sphincter muscle and for rising IOP by increasing the production of aqueous humour [28,29]. In addition, mydriatic effects might appear due to their weak anticholinergic and noradrenergic actions [30]. These contrary possible effects of stimulating serotonin receptors have determined researchers to study the possible real relationship between glaucoma and SSRIs and SNRIs in order to shed some light on the field (Table 1, Figure 1).
The SNRI class has similar indications in psychiatry to SSRI, although they have been later on introduced, in the twentieth century. The SNRI class comprises venlafaxine (the first drug discovered from this class) and duloxetine. Their main mechanism is the inhibition of serotonin and norepinephrine reuptake, with weak dopamine transporter blockage. Noradrenaline is suggested to cause mydriasis and lid retraction through stimulation of α1 receptors; α2 inhibitory receptors from the ciliary epithelium can cause an increase in outflow facility of the aqueous humour while the blockage of these receptors by SNRI could reverse these effects leading to increased IOP [31]. The noradrenergic effect of SNRI is more dominant than the one of SSRI, suggesting a possible high risk of ACG. However, current data suggest that the systemic usage of SNRI could lead in long-term treatment to a decrease in the IOP. Another possible cause of mydriasis due to SNRI treatment would be the stimulation of serotoninergic receptors, mainly 5HT7, which in turn could lead to relaxation of the sphincter muscle [29,32].

3.1.1. OAG

Zheng et al. (2018) documented a potential negative association between SSRIs with primary open angle glaucoma (POAG). More precisely, Zheng et al. (2018) showed that POAG patients treated with systemic medication and under SSRI therapy have a significant lower risk of developing POAG that would require a procedure (patients undergoing treatment with SSRI were at an averagely 30% lower risk for the development of OAG than non-SSRIs users). An approximately 30% reduced risk was also associated with the SNRI class, although less significantly. Also, it has been suggested that there is a dose-response relationship with lower odds of POAG with greater days of treatment [41].
These findings are similar to another cross-sectional study in which three groups of patients with open angle eyes were compared (patients receiving SSRI for 1 week to 6 months, longer than 6 months, or patients under no treatment). IOP was lower in patients under SSRI treatment for less 6 month or more than 6 months in comparison with controls, but the pupil diameter was higher in the abovementioned groups [30].
In contrast, a rise in IOP was documented in the case of a patient with chronic OAG during the initiation of treatment with venlafaxine (a SNRI). After the patient complained of headaches, the starting dose of 225 mg was reduced to 75 mg. No symptoms were reported after the dosage was lowered. Although asymptomatic at 3 months, the IOP increased and the retinal nerve suffered damages [42].

3.1.2. ACG

Regarding ACG, in a large population-based study it was associated with a recent exposure to antidepressants in older adults [43], whereas long exposure to SSRIs did not influence the risk of ACG [44]. Another study conducted by Chen et al. (2017) concluded that individuals under SSRIs therapy had a greater risk of glaucoma (OAG, PACG, glaucoma state, glaucoma suspicion, other forms of glaucoma) incidence. Also, long-term use (>365 days) and/or high dosage were associated with a greater risk of developing glaucoma with an additive effect when both variables were included [37]. In a case-control study that included 1456 ACG patients, immediate SSRI users had 5.80 higher chances to develop ACG as compared to nonusers [45].
Conversely, a recent meta-analysis outlined that treatment with SSRIs was not associated with a higher risk of POAG or PACG and that IOP seemed to be lower in patients exposed to SSRIs. Also, Wang et al. (2018) concluded that pupillary diameter was higher in subjects under this type of antidepressant treatment [46].
Regarding the individual risk of each drug from the SSRIs class to cause glaucoma, current research is scarce and only a few case reports are available in the literature.
Citalopram-induced glaucoma (unilateral ACG) was reported in the literature in a case of drug overdose where the patient presented with blurred vision, pain and corneal oedema, in association with a high IOP. This patient was noted to have shallow anterior chambers in both eyes. After initial ophthalmologic treatment her IOP maintained normal without anti-glaucoma maintenance treatment [47]. Another case of citalopram induced bilateral symptomatic acute angle closure was reported in a patient with a history of 5 months of treatment with a normal dosage (the patient presented with blurred vision and headache) [48].
A case report of escitalopram-induced bilateral ACG described ciliochoroidal effusions after 4 weeks of treatment with a daily dose of 20 mg in a patient that presented with blurred vision. Ophthalmic examination revealed: high IOP, bilateral shallow anterior chamber, best corrected visual acuity was 20/40 bilaterally and a myopic shift of 4 dioptres over the current spectacle prescription. The condition was resistant to medical and surgical treatment but the patient recovered completely after escitalopram was discontinued [49]. Another article reported headache, blurred vision, vomiting, and photophobia (typical symptoms of AACG) in a patient that suddenly stopped escitalopram 1 month before the debut of symptoms. The risk factors identified in this case included hypertension (under control with beta-blockers) and escitalopram use for 1 year [50].
Patients under paroxetine therapy have reported ACG specific symptoms (i.e., loss of visual acuity or blurred vision) between 1 day and 4 months of treatment [51,52,53,54,55]. Interestingly, Sierra-Rodriguez et al. (2013) presented a case report of a unilateral visual loss due to chronic ACG under paroxetine treatment for 4 months. After discontinuation of paroxetine and laser iridotomy, the IOP normalised. Unfortunately, the patient resumed treatment on her own with consequent IOP rise despite patent iridotomies [53].
Regarding fluvoxamine, a patient with a previous history of narrow angle glaucoma (with no iridectomy) presented with daily headaches for 3 months and depressive symptoms (for anxiety the patient was taking lorazepam 2mg/day) and was prescribed fluvoxamine. After two months treatment the patient reported severe orbital pain and blurred vision (increased IOP and mydriasis). Despite specific therapy, IOP decreased only after the withdraw of the antidepressant [56].
Similar ACG symptoms were reported after three days of sertraline treatment, in a 64 year old Chinese woman with hypermetropia. It is worth to mention that Chinese ethnicity, old age, female gender and hypermetropia are risk factors for AACG [57].
Concerning SNRIs, the current literature describes only two and four ACG case reports involving duloxetine and venlafaxine, respectively.
The possible association of duloxetine with the appearance of ACG symptoms was reported in two female patients (46 and 81 years old, respectively). It is important to mention that the 81 years old patient was suffering from other comorbidities, hypermetropia and cataract [33,58].
Regarding venlafaxine, literature reports 4 possible cases of AACG after recent administration of this antidepressant, with the onset of symptoms ranging between 4 h and 10 days. In all reported cases, the patients were females of different age (between 35 and 70 years old) and had blurred vision as a common symptom [59,60,61,62].
Taking into account all the described current literature data, SSRIs and SNRIs have in general no association with either types of glaucoma or increased IOP. Moreover, it is worth emphasizing that long term treatment with SSRIs or SNRIs is associated with a decrease in the IOP, which suggests a possible protective effect of these drugs that needs further investigation. Of course, studies that assess the relationship between specific SSRIs or SNRIs and the risk of raised IOP and glaucoma are necessary in order to better characterise each drug regarding this possible side effect. The current case reports that describe a possible relationship between a specific SSRI or SNRI and ACG should warn the prescribers to closely monitor patients during treatment, especially the individuals with associated risk factors.

3.2. Bupropion

Bupropion is a noradrenaline and dopamine reuptake inhibitor (NDRI) and has been used since 1985 mainly as an antidepressant and more recently as adjuvant for smoking cessation. Bupropion is known to have anti-tumour necrosis factor (TNF) effects and a decreased activity on acetylcholine receptors that result in less anticholinergic side effects [63]. Studies hypothesised that IOP might be raised by TNF through increased caspase activity or mitochondrial dysfunction in the aqueous humour outflow channels. TNF synthesis is decreased by noradrenaline (β2 receptor) and dopamine (D1 receptor) activation [39,64]. All these effects led to the possible conclusion that bupropion could have some protective proprieties regarding IOP and glaucoma (Table 1).
A cross-sectional study that included patients over 40 years old investigated the relationship between self-reported glaucoma and self-reported bupropion use for at least 1 year. Masis et al. (2017) concluded that the usage of bupropion for longer than one year may be associated with a lower risk of self-reported glaucoma. Other covariates associated with high risk included Hispanic/Black ethnicity, increased age, cataract extraction, and diabetic neuropathy. One important limitation of this study is the lack of specificity in glaucoma type (ACG or OAG) [64].

3.2.1. OAG

A cohort-type study regarding the risk of OAG coupled with bupropion treatment, reported a reduced hazard of developing this type of glaucoma. More precisely, the percentage of bupropion users that developed OAG was 1.8% and the percentage of non-users who developed OAG was 2.4%. Moreover, usage of this drug for 24–48 months has been associated with a 21% reduced chance of OAG [39].

3.2.2. ACG

A case-control study conducted on patients under 50 years old reported bupropion treatment to be associated with an increased risk of ACG. No new prescriptions were issued afterwards, which could imply that predisposed eyes with narrow angles and pupillary dilation were not likely since iridotomy would have allowed continuation of treatment. Although the manufacturer’s information references the occurrence of ACG secondary to a pharmacological pupillary dilation, there is a possibility that choroidal effusion can occur [36].
Also, 2 weeks bupropion treatment (300 mg/day) was incriminated as a cause ACG in a 40 years old woman, with complains of blurred vision. Ultrasound bio microscopy revealed bilateral choroidal effusions that caused shallow angles [65].

3.3. Tricyclic Antidepressants (TCAs)

TCAs are the first generation of antidepressants and have been used in the psychopharmacological treatment of depression since around 1950. TCAs inhibit, through action on specific transporters, the reuptake of serotonin and noradrenaline in the synapse cleft. Also, TCAs block the postsynaptic histamine, acetylcholine and alpha-adrenergic receptors. Unfortunately, due to the cardiovascular and gastrointestinal serious adverse effects and their lethality in overdose quantities, TCAs have been replaced over time by SSRIs and SNRIs in the management of depression [66].
According current literature data, tricyclic antidepressants (clomipramine, amitriptyline, imipramine, doxepin, desipramine, nortriptyline etc.) are reported to be involved only in ACG and to precipitate AACG. The pupillary block via pupil dilatation that occurs during treatment with TCAs is attributed to the significant anticholinergic and serotonergic effects of these antidepressants [67]. The most frequent anticholinergic effects on the eye are mydriasis and cycloplegia, which in turn may cause the blockage of the trabecular meshwork. These effects result in blurred vision due to loss of accommodation and in precipitation of ACG [67,68,69,70] (Figure 1).
A relative large body of case reports that linked different TCAs treatment to the raise of IOP and glaucoma occurrence is present in the literature. Schlingemann et al. (1996) described the case of a 59-year-old woman with developed monocular vision loss, increased IOP and narrowed anterior chambers supposing due to treatment with clomipramine (75 mg/day) [71]. Lowe et al. (1966) also reported the cases of 4 patients on small dosage amitriptyline therapy that developed ACG [72]. In addition, Ritch et al. (1994) documented 4 cases of narrow angled patients who developed ACG related to imipramine treatment. Ritch et al. stated that uveal tract problems could be associated with TCA, mydriasis being often transient, without major consequences. Moreover, ACG can be promoted in susceptible patients (e.g., narrow angle individuals) [73].
All things considered, current data point out the risk of using drugs with potent anticholinergics proprieties, such as TCAs. The development of the new classes of antidepressants (i.e., SSRI and SNRI) provides important alternatives. In the case that TCAs must be used, the drugs with the less anticholinergic effects, such as desipramine and nortriptyline, should be taken into consideration [74]. No doubt, further cohort studies that assess the potential risk of angle closure glaucoma associated with TCAs are necessary in order to make conclusive recommendations.

4. Benzodiazepines (BZD)

Benzodiazepines are among the most commonly prescribed drug class in psychiatry and exhibit sedative, hypnotic, anxiolytic and muscle-relaxing properties by enhancing the effect of gamma aminobutyric acid (GABA). Due to this effect, benzodiazepines are incriminated to influence the sphincter pupillae and to determine the narrowing of iridocorneal angle [75]. Current literature documents only the relationship of BZD with ACG and AACG.
Until recently, few cases have been reported about the association of AACG with BZD treatment. The conclusions were rather ambiguous, given the fact that other psychotropic agents have been concomitantly used during the time AACG was reported to occur [75]. Park et al. (2019) tried to reveal the clear relationship between benzodiazepine usage and the risk of glaucoma. In this population-based case-control study on elderly patients, who are more susceptible to the adverse effects to BZD, the authors demonstrated a significant correlation between immediate new use of BZD (within 7 days of AACG diagnosis) and the occurrence of AACG. Oppositely, no significant change in AACG incidence in the non-immediate new users was reported. In addition, there was no significant difference between short half-life (<24 h) vs. long half-life (>24 h) benzodiazepine agents [76]. These findings are similar to the ones of Kim et al. (2020), who outlined an association between BZD therapy and AACG in a cohort of 6709 patients [75]. In the study group the most frequent prescribed BZD were Diazepam and Alprazolam. These drugs were also associated with the highest risk of AACG occurrence [75]. However, a different study concluded that diazepam reduced the IOP and would actually be safe in procedures where lowered IOP is desirable [77].
Therefore, we conclude that benzodiazepines could precipitate ACG in predisposed eyes and clinicians should be aware of these possible side effects.

5. Antipsychotics

Antipsychotic drugs are the cornerstone in the management of schizophrenia. Other indications for this category include schizoaffective disorder, bipolar disorder, delusional disorder, severe agitation, delirium, or psychotic features of major depressive disorder. They can be divided in two categories: typical or first generation (Haloperidol and Chlorpromazine as most known) and atypical or second generation (clozapine, asenapine, olanzapine, quetiapine, paliperidone, risperidone, sertindole, ziprasidone, zotepine, and aripiprazole). Typical antipsychotics manage the symptoms of psychosis through the blockage of the dopaminergic postsynaptic receptors, with additional histaminergic, α1 adrenergic and cholinergic blockade. Atypical antipsychotics are serotonin and dopamine antagonists with affinities for serotonin (5HT1A, 5HT2C, 5HT6, 5HT7), dopamine (D1, D3, D4) receptors, but also histamine (H1), muscarinic (M1, M2, M3, M4, M5) and adrenergic (α1, α2) receptors [78]. Most AAPs have in common a more potent 5HT2A action than dopamine (D2) antagonism. They are also partial agonists of 5HT1A. All these features contribute to the low extrapyramidal effects. Some other actions include the inhibition of muscarinic receptors (anticholinergic activity). A possible explanation for the association between antipsychotic drugs and glaucoma might be the cholinergic receptor blockade (muscarinic) [34,79] (Table 1, Figure 1).
Unfortunately, current research reports only the possible effects of antipsychotics on IOP and does not scrutinise the relationship between these drugs and OAG or ACG.
A cross-sectional study including 28 patients with schizophrenia showed that individuals under typical antipsychotics treatment had normal IOP [79]. This result is similar to the one reported by Reid et al. (1976). Moreover, Reid et al. (1976) study concluded that there was no IOP raise despite high dosage of typical antipsychotics [80]. Actually, not only haloperidol was found to lower IOP in glaucomatous eyes, but also it was proposed as a possible treatment for glaucoma [81,82,83].
Atypical antipsychotics (AAP) have cardiovascular side effects through the vasoconstriction determined by α1 adrenoreceptors blockage. Risperidone, clozapine, iloperidone and quetiapine may lead to hypotension via this mechanism [34]. Animal studies showed instilled APP into the eye determined a significant reduction of IOP observed after 1 h. Also, reduction of blood pressure occurred within 10 min after administration [84].
Reactive oxygen species (ROS) production is involved in the pathogenic mechanism of glaucoma. Risperidone was demonstrated to have the ability to decrease oxidative stress (OS) in schizophrenic patients by controlling the inflammatory response [85]. Other AAP that have been shown to decrease OS are clozapine and olanzapine [86]. However, the relationship between AAP, OS and glaucoma has been incompletely investigated. Therefore, future studies are necessary to elucidate this possible mechanism.
Several studies described that IOP elevation may lead to the inhibition of brain-derived-neurotrophic factor (BDNF) which could have in turn a contributing effect to the visual loss. Risperidone and clozapine have been found to increase levels of BDNF [87,88,89].
There is some evidence that AAPs could enhance glaucoma through anti-muscarinic action. For example, clozapine and olanzapine have high affinity for muscarinic receptors (inhibition) and anticholinergic activity, which could possibly exacerbate glaucoma [90]). Thus, the actions of AAP by downregulating OS and neurotrophins may be unbalanced because of their anti- muscarinic receptor action. This observation could explain that, in general, AAPs are not associated with a glaucoma risk [66].
A cross-sectional analysis of 28 patients with schizophrenia and under antipsychotic therapy (4 on typical antipsychotics, 16 on AAP, and 8 on both types) provided interesting conclusions. More precisely, a raise of IOP has been found only in patients under AAP therapy, particularly all on ziprasidone. Ziprasidone is known to exert a potent serotoninergic (5HT2A) and dopaminergic (D2) affinity [79].
In conclusion, clinicians should be aware that second generation antipsychotics could have some implication in the variations of IOP, therefore a special attention should be paid to patients at risk and also when prescribing ziprasidone. Increased IOP could have no clinical significance in certain situations (e.g., minor increase or lower basal IOP) or could lead to the development or progression of glaucoma (OAG or ACG) depending on the characteristics of the patient. Typical antipsychotics are suggested to be safer in relation to a possible rise of IOP.

6. Topiramate

Topiramate is a sulpha-derivative monosaccharide and it is commonly prescribed for treatment of epilepsy and migraine prevention. Off label indications include eating disorders, obesity and tobacco dependence. Regarding psychiatric recommendations, the current literature describes the benefits of topiramate in weight gain prevention and metabolic dysfunction in schizophrenic patients as a result of treatment with certain antipsychotics (i.e., olanzapine, clozapine) [91].
Topiramate’s mechanism of action involves inhibition of carbonic anhydrase, calcium channels, and glutamate receptors, as well as blockage of the sodium channels and stimulation of gamma-aminobutyric acid receptors. Topiramate is suggested to cause or worsen glaucoma due to an acute hypersensitivity reaction and alteration of osmotic status. The mechanism is suggested to involve ciliochoroidal effusion which leads to anterior rotation of the swelled ciliary body with anterior shifting of lens-iris diaphragm and consequently shallowing of the anterior chamber and narrowing of the angle [92,93]. All these changes may also occur in normal eyes. Given this fact, clinicians should be more vigilant and it is advisable to adopt a watchful waiting approach for all patients treated with topiramate.
In a retrospective study, Ho JD et al. (2013) reported a greater risk of developing ACG after topiramate therapy in the first month of treatment (hazard ratio 7.41 times higher than control subjects) [93]. Other data showed an increased risk of ACG, myopia, suprachoroidal effusion, and abnormal vision, all reversible with the discontinuation of treatment [94,95].
Numerous case reports found in the current literature described the association of topiramate treatment with the development of ACG. In 38 reported cases [96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130], patients under topiramate treatment developed ACG after a short time from the treatment initiation (the majority between 1 day and 14 days), most of them being adult women (27 women / 38 patients) with an age ranging from 23 to 59 years. Table 2 presents a summary of current topiramate induced acute angle closure or AACG case reports (Table 2).

7. Conclusions

Based on the presented data, clinicians should be aware of the glaucoma-related risk-benefit profile of psychotropic medication and tailor their recommendations. The selective serotonin and noradrenaline reuptake inhibitors class is the medication group with the most solid results regarding a minimal possible risk of glaucoma. More precisely, SSRI and SNRI treatment seems to have even a protective role regarding OAG and no effect in relationship with ACG. Therefore, practitioners could use these drugs safely since there is no risk of a glaucoma induced effect. On the other hand, tricyclic antidepressants should be avoided in patients at risk to develop angle closure glaucoma or in angle closure glaucoma diagnosed individuals due to their strong anticholinergic and antimuscarinic proprieties. Regarding all antipsychotics, there is an important gap in the knowledge of their relationship with the risk of glaucoma. Based on the herein reviewed data, first generation antipsychotics do not seem to affect the intraocular pressure, but for the second generation antipsychotics, and especially ziprasidone, further studies are needed in order to bring some light to the current data. Also, topiramate is another drug that we advise not be used in the treatment of patients with possible risk factors or diagnosed with angle closure glaucoma, since current data point to an increased risk of trabecular obstruction and consequently a raise in intraocular pressure. Benzodiazepines should be prescribed carefully, especially in older patients. Whether or not it is identified as a contraindication, physicians should be aware of the possibility of psychotropic drug-induced glaucoma, especially angle closure type, and if the suspicion of glaucoma arises, ophthalmological assessment is recommended. Early recognition of this possible side effect and discontinuation of the drug in question are measures that should be immediately employed by the psychiatrist concomitantly with referring the patient to an ophthalmologist for a thorough evaluation. Due to the vast psychotropic medication and possible mechanisms and their interactions, future studies are needed to fill the literature gaps and enrich current knowledge on this subject.

Author Contributions

Conceptualization, A.M.C. and C.N.; methodology, A.M.C., V.D. and O.P.-V.; software, C.N.; validation, A.M.C., O.P.-V. and S.R.; formal analysis, C.N.; investigation, A.M.C., V.D., C.N.; resources, V.D., C.N., O.M.B.; data curation, V.D., O.M.B.; writing—original draft preparation, A.M.C., V.D., C.N.; writing—review and editing, A.M.C., O.P.-V., S.R., O.M.B.; visualization, O.P.-V.; supervision, S.R.; project administration, A.M.C.; funding acquisition, V.D., C.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data relevant to this paper are included in the article.

Acknowledgments

We would like to thank Ioana Robu for the professional language assistance.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Balendra, S.I.; Shah, P.A.; Jain, M.; Grzybowski, A.; Cordeiro, M.F. Glaucoma: Hot Topics in Pharmacology. Curr. Pharm. Des. 2017, 23, 596–607. [Google Scholar]
  2. Flaxman, S.R.; Bourne, R.R.A.; Resnikoff, S.; Ackland, P.; Braithwaite, T.; Cicinelli, M.V.; Das, A.; Jonas, J.B.; Keeffe, J.; Kempen, J.H.; et al. Global Causes of Blindness and Distance Vision Impairment 1990–2020: A Systematic Review and Meta-Analysis. Lancet Glob. Health 2017, 5, e1221–e1234. [Google Scholar] [CrossRef] [Green Version]
  3. Bertaud, S.; Aragno, V.; Baudouin, C.; Labbé, A. Le glaucome primitif à angle ouvert. Rev. Med. Interne 2019, 40, 445–452. [Google Scholar] [CrossRef]
  4. Casson, R.J.; Chidlow, G.; Wood, J.P.M.; Crowston, J.G.; Goldberg, I. Definition of Glaucoma: Clinical and Experimental Concepts: Definition of Glaucoma. Clin. Exp. Ophthalmol. 2012, 40, 341–349. [Google Scholar] [CrossRef] [Green Version]
  5. Kingman, S. Glaucoma Is Second Leading Cause of Blindness Globally. Bull. World Health Organ. 2004, 82, 887–888. [Google Scholar]
  6. Schuster, A.K.; Erb, C.; Hoffmann, E.M.; Dietlein, T.; Pfeiffer, N. The Diagnosis and Treatment of Glaucoma. Dtsch. Arztebl. Int. 2020, 117, 225–234. [Google Scholar]
  7. Weinreb, R.N.; Aung, T.; Medeiros, F.A. The Pathophysiology and Treatment of Glaucoma: A Review. JAMA 2014, 311, 1901–1911. [Google Scholar] [CrossRef] [Green Version]
  8. McMonnies, C.W. Glaucoma History and Risk Factors. J. Optom. 2017, 10, 71–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Mantravadi, A.V.; Vadhar, N. Glaucoma. Prim. Care 2015, 42, 437–449. [Google Scholar] [CrossRef] [PubMed]
  10. Salmon, J. Kanski’s Clinical Ophthalmology: A Systematic Approach, 9th ed.; Elsevier Health Sciences: London, UK, 2019; pp. 349, 370–374. [Google Scholar]
  11. Quigley, H.A. Number of People with Glaucoma Worldwide. Br. J. Ophthalmol. 1996, 80, 389–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Friedman, D.S.; Wolfs, R.C.W.; O’Colmain, B.J.; Klein, B.E.; Taylor, H.R.; West, S.; Leske, M.C.; Mitchell, P.; Congdon, N.; Kempen, J.; et al. Prevalence of Open-Angle Glaucoma among Adults in the United States. Arch. Ophthalmol. 2004, 122, 532–538. [Google Scholar] [PubMed] [Green Version]
  13. Pantalon, A.D.; Feraru, C.; Chiseliţă, D. Risk Factors and Long Term Progression in Open Angle Glaucoma Patients. Rom. J. Ophthalmol. 2016, 60, 174–180. [Google Scholar] [PubMed]
  14. Kim, K.E.; Kim, M.J.; Park, K.H.; Jeoung, J.W.; Kim, S.H.; Kim, C.Y.; Kang, S.W.; Epidemiologic Survey Committee of the Korean Ophthalmological Society. Prevalence, Awareness, and Risk Factors of Primary Open-Angle Glaucoma: Korea National Health and Nutrition Examination Survey 2008–2011. Ophthalmology 2016, 123, 532–541. [Google Scholar] [CrossRef]
  15. Chan, T.C.W.; Bala, C.; Siu, A.; Wan, F.; White, A. Risk Factors for Rapid Glaucoma Disease Progression. Am. J. Ophthalmol. 2017, 180, 151–157. [Google Scholar] [CrossRef] [PubMed]
  16. Phulke, S.; Kaushik, S.; Kaur, S.; Pandav, S.S. Steroid-Induced Glaucoma: An Avoidable Irreversible Blindness. J. Curr. Glaucoma Pract. 2017, 11, 67–72. [Google Scholar] [PubMed]
  17. Flores-Sánchez, B.C.; Tatham, A.J. Acute Angle Closure Glaucoma. Br. J. Hosp. Med. 2019, 80, C174–C179. [Google Scholar] [CrossRef]
  18. Boonyaleephan, S. Drug-Induced Secondary Glaucoma. J. Med. Assoc. Thai 2010, 93, S118–S122. [Google Scholar]
  19. Distelhorst, J.S.; Hughes, G.M. Open-Angle Glaucoma. Am. Fam. Physician 2003, 67, 1937–1944. [Google Scholar]
  20. Pelčić, G.; Ljubičić, R.; Barać, J.; Biuk, D.; Rogoić, V. Glaucoma, Depression and Quality of Life: Multiple Comorbidities, Multiple Assessments and Multidisciplinary Plan Treatment. Psychiatr. Danub. 2017, 29, 351–359. [Google Scholar] [CrossRef]
  21. Zhang, D.; Fan, Z.; Gao, X.; Huang, W.; Yang, Q.; Li, Z.; Lin, M.; Xiao, H.; Ge, J. Illness Uncertainty, Anxiety and Depression in Chinese Patients with Glaucoma or Cataract. Sci. Rep. 2018, 8, 11671. [Google Scholar] [CrossRef] [Green Version]
  22. Akhilesh, J. Associating Factors of Insomnia and Depression in Glaucoma: A Descriptive Analysis. Int. Multispec. J. Health IMJH 2018, 4, 41–49. [Google Scholar]
  23. Kennedy, S.H.; Lam, R.W.; McIntyre, R.S.; Tourjman, S.V.; Bhat, V.; Blier, P.; Hasnain, M.; Jollant, F.; Levitt, A.J.; MacQueen, G.M.; et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) 2016 Clinical Guidelines for the Management of Adults with Major Depressive Disorder: Section 3. Pharmacological Treatments: Section 3. Pharmacological Treatments. Can. J. Psychiatry 2016, 61, 540–560. [Google Scholar] [CrossRef]
  24. Bauer, M.; Pfennig, A.; Severus, E.; Whybrow, P.C.; Angst, J.; Möller, H.-J.; World Federation of Societies of Biological Psychiatry. Task Force on Unipolar Depressive Disorders. 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, 334–385. [Google Scholar]
  25. Kellner, M. Drug Treatment of Obsessive-Compulsive Disorder. Dialogues Clin. Neurosci. 2010, 12, 187–197. [Google Scholar]
  26. Strawn, J.R.; Geracioti, L.; Rajdev, N.; Clemenza, K.; Levine, A. Pharmacotherapy for Generalized Anxiety Disorder in Adult and Pediatric Patients: An Evidence-Based Treatment Review. Expert Opin. Pharmacother. 2018, 19, 1057–1070. [Google Scholar] [CrossRef] [PubMed]
  27. Asnis, G.M.; Kohn, S.R.; Henderson, M.; Brown, N.L. SSRIs versus Non-SSRIs in Post-Traumatic Stress Disorder: An Update with Recommendations. Drugs 2004, 64, 383–404. [Google Scholar] [CrossRef] [PubMed]
  28. Costagliola, C.; Parmeggiani, F.; Sebastiani, A. SSRIs and Intraocular Pressure Modifications: Evidence, Therapeutic Implications and Possible Mechanisms. CNS Drugs 2004, 18, 475–484. [Google Scholar] [CrossRef]
  29. Wiciński, M.; Kaluzny, B.J.; Liberski, S.; Marczak, D.; Seredyka-Burduk, M.; Pawlak-Osińska, K. Association between Serotonin-Norepinephrine Reuptake Inhibitors and Acute Angle Closure: What Is Known? Surv. Ophthalmol. 2019, 64, 185–194. [Google Scholar] [CrossRef] [PubMed]
  30. Gündüz, G.U.; Parmak-Yener, N.; Kılınçel, O.; Gündüz, C. Effects of Selective Serotonin Reuptake Inhibitors on Intraocular Pressure and Anterior Segment Parameters in Open Angle Eyes. Cutan. Ocul. Toxicol. 2018, 37, 36–40. [Google Scholar] [CrossRef]
  31. Loma, P.; Guzman-Aranguez, A.; de Lara, M.J.P.; Pintor, J. Beta2 Adrenergic Receptor Silencing Change Intraocular Pressure in New Zealand Rabbits. J. Optom. 2018, 11, 69–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Uçan Gündüz, G.; Parmak Yener, N.; Kılınçel, O.; Gündüz, C. How Does Usage of Serotonin Noradrenaline Reuptake Inhibitors Affect Intraocular Pressure in Depression Patients? J. Ocul. Pharmacol. Ther. 2018, 34, 354–359. [Google Scholar] [CrossRef] [PubMed]
  33. Mahmut, A.; Tunc, V.; Demiryurek, E.; Gursoy, A. Bilateral Acute Angle-Closure Glaucoma Induced by Duloxetine. Ideggyogy. Sz. 2017, 70, 358–360. [Google Scholar] [CrossRef] [PubMed]
  34. Vallée, A.; Vallée, J.-N.; Lecarpentier, Y. Lithium and Atypical Antipsychotics: The Possible WNT/β Pathway Target in Glaucoma. Biomedicines 2021, 9, 473. [Google Scholar] [CrossRef]
  35. Stahl, S.M. Essential Psychopharmacology Series: Stahl’s Essential Psychopharmacology: Neuroscientific Basis and Practical Applications, 3rd ed.; Cambridge University Press: Cambridge, UK, 2008; pp. 284–369. [Google Scholar]
  36. Symes, R.J.; Etminan, M.; Mikelberg, F.S. Risk of Angle-Closure Glaucoma with Bupropion and Topiramate. JAMA Ophthalmol. 2015, 133, 1187–1189. [Google Scholar] [CrossRef] [Green Version]
  37. Chen, V.C.-H.; Ng, M.-H.; Chiu, W.-C.; McIntyre, R.S.; Lee, Y.; Lin, T.-Y.; Weng, J.-C.; Chen, P.-C.; Hsu, C.-Y. Effects of Selective Serotonin Reuptake Inhibitors on Glaucoma: A Nationwide Population-Based Study. PLoS ONE 2017, 12, e0173005. [Google Scholar]
  38. Richelson, E. Pharmacology of antidepressants. Mayo Clin. Proc. 2001, 76, 511–527. [Google Scholar] [CrossRef] [Green Version]
  39. Stein, J.D.; Talwar, N.; Kang, J.H.; Okereke, O.I.; Wiggs, J.L.; Pasquale, L.R. Bupropion Use and Risk of Open-Angle Glaucoma among Enrollees in a Large U.S. Managed Care Network. PLoS ONE 2015, 10, e0123682. [Google Scholar] [CrossRef] [PubMed]
  40. Tripathi, R.; Tripathi, B.; Haggerty, C. Drug-Induced Glaucomas. Drug Saf. 2003, 26, 749–767. [Google Scholar] [CrossRef] [PubMed]
  41. Zheng, W.; Dryja, T.P.; Wei, Z.; Song, D.; Tian, H.; Kahler, K.H.; Khawaja, A.P. Systemic Medication Associations with Presumed Advanced or Uncontrolled Primary Open-Angle Glaucoma. Ophthalmology 2018, 125, 984–993. [Google Scholar] [CrossRef] [Green Version]
  42. Botha, V.E.; Bhikoo, R.; Merriman, M. Venlafaxine-Induced Intraocular Pressure Rise in a Patient with Open Angle Glaucoma: Venlafaxine Induced Intraocular Pressure Rise. Clin. Exp. Ophthalmol. 2016, 44, 734–735. [Google Scholar] [CrossRef]
  43. Seitz, D.P.; Campbell, R.J.; Bell, C.M.; Gill, S.S.; Gruneir, A.; Herrmann, N.; Newman, A.M.; Anderson, G.; Rochon, P.A. Short-Term Exposure to Antidepressant Drugs and Risk of Acute Angle-Closure Glaucoma among Older Adults. J. Clin. Psychopharmacol. 2012, 32, 403–407. [Google Scholar] [CrossRef] [PubMed]
  44. Chen, H.-Y.; Lin, C.-L.; Kao, C.-H. Long-Term Use of Selective Serotonin Reuptake Inhibitors and Risk of Glaucoma in Depression Patients. Medicine 2015, 94, e2041. [Google Scholar] [CrossRef] [PubMed]
  45. Chen, H.-Y.; Lin, C.-L.; Lai, S.-W.; Kao, C.-H. Association of Selective Serotonin Reuptake Inhibitor Use and Acute Angle–Closure Glaucoma. J. Clin. Psychiatry 2016, 77, e692–e696. [Google Scholar] [CrossRef]
  46. Wang, H.-Y.; Tseng, P.-T.; Stubbs, B.; Carvalho, A.F.; Li, D.-J.; Chen, T.-Y.; Lin, P.-Y.; Hsueh, Y.-T.; Chen, Y.-Z.; Chen, Y.-W.; et al. The Risk of Glaucoma and Serotonergic Antidepressants: A Systematic Review and Meta-Analysis. J. Affect. Disord. 2018, 241, 63–70. [Google Scholar] [CrossRef]
  47. Croos, R.; Thirumalai, S.; Hassan, S.; Davis, J.D.R. Citalopram Associated with Acute Angle-Closure Glaucoma: Case Report. BMC Ophthalmol. 2005, 5, 23. [Google Scholar] [CrossRef] [Green Version]
  48. Massaoutis, P.; Goh, D.; Foster, P.J. Bilateral Symptomatic Angle Closure Associated with a Regular Dose of Citalopram, an SSRI Antidepressant. Br. J. Ophthalmol. 2007, 91, 1086–1087. [Google Scholar] [CrossRef] [Green Version]
  49. Zelefsky, J.R.; Fine, H.F.; Rubinstein, V.J.; Hsu, I.S.; Finger, P.T. Escitalopram-Induced Uveal Effusions and Bilateral Angle Closure Glaucoma. Am. J. Ophthalmol. 2006, 141, 1144–1147. [Google Scholar] [CrossRef] [PubMed]
  50. AlQuorain, S.; Alfaraj, S.; Alshahrani, M. Bilateral Acute Closed Angle Glaucoma Associated with the Discontinuation of Escitalopram: A Case Report. Open Access Emerg. Med. 2016, 8, 61–65. [Google Scholar] [PubMed] [Green Version]
  51. Eke, T.; Bates, A.K. Acute Angle Closure Glaucoma Associated with Paroxetine. BMJ 1997, 314, 1387. [Google Scholar] [CrossRef] [PubMed]
  52. Kirwan, J.F.; Subak-Sharpe, I.; Teimory, M. Bilateral Acute Angle Closure Glaucoma after Administration of Paroxetine. Br. J. Ophthalmol. 1997, 81, 252. [Google Scholar] [CrossRef] [Green Version]
  53. Sierra-Rodriguez, M.A.; Saenz-Frances, F.; Santos-Bueso, E.; Garcia-Feijoo, J.; Gonzelez-Romero, J.C. Chronic Angle-Closure Glaucoma Related to Paroxetine Treatment. Semin. Ophthalmol. 2013, 28, 244–246. [Google Scholar] [CrossRef] [PubMed]
  54. Browning, A.C.; Reck, A.C.; Chisholm, I.H.; Nischal, K.K. Acute Angle Closure Glaucoma Presenting in a Young Patient after Administration of Paroxetine. EYE 2000, 14, 406–408. [Google Scholar] [CrossRef] [PubMed]
  55. Levy, J.; Tessler, Z.; Klemperer, I.; Shneck, M.; Lifshitz, T. Late Bilateral Acute Angle-Closure Glaucoma after Administration of Paroxetine in a Patient with Plateau Iris Configuration. Can. J. Ophthalmol. 2004, 39, 780–781. [Google Scholar] [CrossRef]
  56. Jiménez-Jiménez, F.J.; Ortí-Pareja, M.; Zurdo, J.M. Aggravation of Glaucoma with Fluvoxamine. Ann. Pharmacother. 2001, 35, 1565–1566. [Google Scholar] [CrossRef]
  57. Ho, H.Y.; Kam, K.-W.A.; Young, A.L.; Chan, L.K.; Yu, E.C.-S. Acute Angle Closure Glaucoma after Sertraline. Gen. Hosp. Psychiatry 2013, 35, 575. [Google Scholar] [CrossRef] [PubMed]
  58. Shifera, A.S.; Leoncavallo, A.; Sherwood, M. Probable Association of an Attack of Bilateral Acute Angle-Closure Glaucoma with Duloxetine. Ann. Pharmacother. 2014, 48, 936–939. [Google Scholar] [CrossRef]
  59. de Guzman, M.H.P.; Thiagalingam, S.; Ong, P.Y.; Goldberg, I. Bilateral Acute Angle Closure Caused by Supraciliary Effusions Associated with Venlafaxine Intake. Med. J. Aust. 2005, 182, 121–123. [Google Scholar] [CrossRef]
  60. Ezra, D.G.; Storoni, M.; Whitefield, L.A. Simultaneous Bilateral Acute Angle Closure Glaucoma Following Venlafaxine Treatment. EYE 2006, 20, 128–129. [Google Scholar] [CrossRef] [Green Version]
  61. Ng, B.; Sanbrook, G.M.C.; Malouf, A.J.; Agarwal, S.A. Venlafaxine and Bilateral Acute Angle Closure Glaucoma. Med. J. Aust. 2002, 176, 241. [Google Scholar] [CrossRef] [PubMed]
  62. Zhou, N.; Zhao, J.-X.; Zhu, Y.-N.; Zhang, P.; Zuo, Y. Acute Angle-Closure Glaucoma Caused by Venlafaxine. Chin. Med. J. Engl. 2018, 131, 1502–1503. [Google Scholar] [CrossRef]
  63. Rudorfer, M.V.; Manji, H.K.; Potter, W.Z. Comparative Tolerability Profiles of the Newer versus Older Antidepressants. Drug Saf. 1994, 10, 18–46. [Google Scholar] [CrossRef]
  64. Masís, M.; Kakigi, C.; Singh, K.; Lin, S. Association between Self-Reported Bupropion Use and Glaucoma: A Population-Based Study. Br. J. Ophthalmol. 2017, 101, 525–529. [Google Scholar] [CrossRef]
  65. Takusagawa, H.L.; Hunter, R.S.; Jue, A.; Pasquale, L.R.; Chen, T.C. Bilateral Uveal Effusion and Angle-Closure Glaucoma Associated with Bupropion Use. Arch. Ophthalmol. 2012, 130, 120–122. [Google Scholar] [CrossRef] [Green Version]
  66. Richa, S.; Yazbek, J.-C. Ocular Adverse Effects of Common Psychotropic Agents: A Review: A Review. CNS Drugs 2010, 24, 501–526. [Google Scholar] [CrossRef] [PubMed]
  67. Ah-Kee, E.Y.; Egong, E.; Shafi, A.; Lim, L.T.; Yim, J.L. A Review of Drug-Induced Acute Angle Closure Glaucoma for Non-Ophthalmologists. Qatar Med. J. 2015, 2015, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Lieberman, E.; Stoudemire, A. Use of Tricyclic Antidepressants in Patients with Glaucoma. Assessment and Appropriate Precautions. Psychosomatics 1987, 28, 145–148. [Google Scholar] [CrossRef]
  69. Sönmez, İ.; Aykan, Ü. Psychotropic Drugs and Ocular Side Effects. Turk. J. Ophthalmol. 2014, 44, 144–150. [Google Scholar] [CrossRef]
  70. Razeghinejad, M.R.; Myers, J.S.; Katz, L.J. Iatrogenic Glaucoma Secondary to Medications. Am. J. Med. 2011, 124, 20–25. [Google Scholar] [CrossRef]
  71. Schlingemann, R.O.; Smit, A.A.; Lunel, H.F.; Hijdra, A. Amaurosis Fugax on Standing and Angle-Closure Glaucoma with Clomipramine. Lancet 1996, 347, 465. [Google Scholar] [CrossRef]
  72. Lowe, R.F. Amitriptyline and Glaucoma. Med. J. Aust. 1966, 2, 509–510. [Google Scholar] [CrossRef]
  73. Ritch, R.; Krupin, T.; Henry, C.; Kurata, F. Oral Imipramine and Acute Angle Closure Glaucoma. Arch. Ophthalmol. 1994, 112, 67–68. [Google Scholar] [CrossRef]
  74. Epstein, N.E.; Goldbloom, D.S. Oral Imipramine and Acute Angle-Closure Glaucoma. Arch. Ophthalmol. 1995, 113, 698. [Google Scholar] [CrossRef]
  75. Kim, W.J.; Li, J.; Oh, I.-S.; Song, I.; Lee, E.; Namkoong, K.; Shin, J.-Y. Benzodiazepine Use and Risk of Acute Angle-Closure Glaucoma: A Population-Based Case-Crossover Study. Drug Saf. 2020, 43, 539–547. [Google Scholar] [CrossRef]
  76. Park, M.Y.; Kim, W.J.; Lee, E.; Kim, C.; Son, S.J.; Yoon, J.S.; Kim, W.; Namkoong, K. Association between Use of Benzodiazepines and Occurrence of Acute Angle-Closure Glaucoma in the Elderly: A Population-Based Study. J. Psychosom. Res. 2019, 122, 1–5. [Google Scholar] [CrossRef] [PubMed]
  77. Fragen, R.J.; Hauch, T. The Effect of Midazolam Maleate and Diazepam on Intraocular Pressure in Adults. Arzneimittelforschung 1981, 31, 2273–2275. [Google Scholar]
  78. Nasrallah, H.A. Atypical Antipsychotic-Induced Metabolic Side Effects: Insights from Receptor-Binding Profiles. Mol. Psychiatry 2008, 13, 27–35. [Google Scholar] [CrossRef]
  79. Souza, V.B.N.E.; Moura Filho, F.J.R.; de Souza, F.G.d.M.E.; Pereira Filho, S.A.C.; Coelho, S.S.; Furtado, F.A.M.L.; Gonçalves, T.B.A.; Vasconcelos, K.F.X. Intraocular Pressure in Schizophrenic Patients Treated with Psychiatric Medications. Arq. Bras. Oftalmol. 2008, 71, 660–664. [Google Scholar] [CrossRef] [Green Version]
  80. Reid, W.H.; Blouin, P. Outpatient Psychiatric Medications and Glaucoma. Psychosomatics 1976, 17, 83–85. [Google Scholar] [CrossRef]
  81. Khosla, P.; Kothari, S.; Gupta, M.C.; Srivastava, R.K. Evaluation of Haloperidol, a Dopamine Antagonist, on Intraocular Pressure in Experimental Glaucoma. Indian J. Exp. Biol. 1996, 34, 580–581. [Google Scholar] [PubMed]
  82. Chiou, G.C. Ocular Hypotensive Actions of Haloperidol, a Dopaminergic Antagonist. Arch. Ophthalmol. 1984, 102, 143–145. [Google Scholar] [CrossRef] [PubMed]
  83. Sheppard, J.D.; Schaid, D.J. Oral Haloperidol Lowers Human Intraocular Pressure. J. Ocul. Pharmacol. 1986, 2, 215–224. [Google Scholar] [CrossRef]
  84. Joshi, S.V.; Patel, E.P.; Vyas, B.A.; Lodha, S.R.; Kalyankar, G.G. Repurposing of Iloperidone: Antihypertensive and Ocular Hypotensive Activity in Animals. Eur. J. Pharm. Sci. 2020, 143, 105173. [Google Scholar] [CrossRef]
  85. Al-Amin, M.M.; Nasir-Uddin, M.M.; Mahmud Reza, H. Effects of Antipsychotics on the Inflammatory Response System of Patients with Schizophrenia in Peripheral Blood Mononuclear Cell Cultures. Clin. Psychopharmacol. Neurosci. 2013, 11, 144–151. [Google Scholar] [CrossRef]
  86. Caruso, G.; Grasso, M.; Fidilio, A.; Tascedda, F.; Drago, F.; Caraci, F. Antioxidant Properties of Second-Generation Antipsychotics: Focus on Microglia. Pharmaceuticals 2020, 13, 457. [Google Scholar] [CrossRef] [PubMed]
  87. Aringhieri, S.; Carli, M.; Kolachalam, S.; Verdesca, V.; Cini, E.; Rossi, M.; McCormick, P.J.; Corsini, G.U.; Maggio, R.; Scarselli, M. Molecular Targets of Atypical Antipsychotics: From Mechanism of Action to Clinical Differences. Pharmacol. Ther. 2018, 192, 20–41. [Google Scholar] [CrossRef] [PubMed]
  88. Carli, M.; Aringhieri, S.; Kolachalam, S.; Longoni, B.; Grenno, G.; Rossi, M.; Gemignani, A.; Fornai, F.; Maggio, R.; Scarselli, M. Is Adult Hippocampal Neurogenesis Really Relevant for the Treatment of Psychiatric Disorders? Curr. Neuropharmacol. 2020, 18. [Google Scholar] [CrossRef]
  89. Quigley, H.A.; McKinnon, S.J.; Zack, D.J.; Pease, M.E.; Kerrigan-Baumrind, L.A.; Kerrigan, D.F.; Mitchell, R.S. Retrograde Axonal Transport of BDNF in Retinal Ganglion Cells Is Blocked by Acute IOP Elevation in Rats. Investig. Ophthalmol. Vis. Sci. 2000, 41, 3460–3466. [Google Scholar]
  90. Chew, M.L.; Mulsant, B.H.; Pollock, B.G.; Lehman, M.E.; Greenspan, A.; Kirshner, M.A.; Bies, R.R.; Kapur, S.; Gharabawi, G. A Model of Anticholinergic Activity of Atypical Antipsychotic Medications. Schizophr. Res. 2006, 88, 63–72. [Google Scholar] [CrossRef]
  91. Narula, P.K.; Rehan, H.S.; Unni, K.E.S.; Gupta, N. Topiramate for Prevention of Olanzapine Associated Weight Gain and Metabolic Dysfunction in Schizophrenia: A Double-Blind, Placebo-Controlled Trial. Schizophr. Res. 2010, 118, 218–223. [Google Scholar] [CrossRef]
  92. Rapoport, Y.; Benegas, N.; Kuchtey, R.W.; Joos, K.M. Acute Myopia and Angle Closure Glaucoma from Topiramate in a Seven-Year-Old: A Case Report and Review of the Literature. BMC Pediatr. 2014, 14, 96. [Google Scholar] [CrossRef] [Green Version]
  93. Ho, J.-D.; Keller, J.J.; Tsai, C.-Y.; Liou, S.-W.; Chang, C.-J.; Lin, H.-C. Topiramate Use and the Risk of Glaucoma Development: A Population-Based Follow-up Study. Am. J. Ophthalmol. 2013, 155, 336–341. [Google Scholar] [CrossRef]
  94. Fraunfelder, F.W.; Fraunfelder, F.T.; Keates, E.U. Topiramate-Associated Acute, Bilateral, Secondary Angle-Closure Glaucoma. Ophthalmology 2004, 111, 109–111. [Google Scholar] [CrossRef]
  95. Etminan, M.; Maberley, D.; Mikelberg, F.S. Use of Topiramate and Risk of Glaucoma: A Case-Control Study. Am. J. Ophthalmol. 2012, 153, 827–830. [Google Scholar] [CrossRef] [PubMed]
  96. Alzendi, N.; Badawi, A.; Alhazzaa, B.; Alshahrani, A.; Owaidhah, O. Topiramate-Induced Angle Closure Glaucoma: Two Unique Case Reports. Saudi J. Ophthalmol. 2020, 34, 202. [Google Scholar] [CrossRef] [PubMed]
  97. Agarwal, A. Ciliochoroidal Effusion in Topiramate-Induced Bilateral Acute Angle Closure Glaucoma. Indian J. Ophthalmol. 2019, 67, 1466–1467. [Google Scholar] [CrossRef]
  98. Mahendradas, P.; Parab, S.; Sasikumar, R.; Kawali, A.; Shetty, B.K. Topiramate-Induced Acute Angle Closure with Severe Panuveitis: A Challenging Case Report. Indian J. Ophthalmol. 2018, 66, 1342–1344. [Google Scholar] [CrossRef] [PubMed]
  99. Sierra-Rodríguez, M.A.; Rodríguez-Vicente, L.; Chavarri-García, J.J.; Del Río-Mayor, J.L. Acute narrow-angle glaucoma induced by topiramate with acute myopia and macular striae: A case report. Arch. Soc. Esp. Oftalmol. Engl. Ed. 2019, 94, 130–133. [Google Scholar] [CrossRef]
  100. Lan, Y.-W.; Hsieh, J.-W. Bilateral Acute Angle Closure Glaucoma and Myopic Shift by Topiramate-Induced Ciliochoroidal Effusion: Case Report and Literature Review. Int. Ophthalmol. 2018, 38, 2639–2648. [Google Scholar] [CrossRef]
  101. Czyz, C.N.; Clark, C.M.; Justice, J.D.; Pokabla, M.J.; Weber, P.A. Delayed Topiramate-Induced Bilateral Angle-Closure Glaucoma. J. Glaucoma 2014, 23, 577–578. [Google Scholar] [CrossRef] [PubMed]
  102. Pikkel, Y.Y. Acute Bilateral Glaucoma and Panuveitis as a Side Effect of Topiramate for Weight Loss Treatment. BMJ Case Rep. 2014, 2014, bcr2014203787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Katsimpris, J.M.; Katsimpris, A.; Theoulakis, P.E.; Lepidas, J.; Petropoulos, I.K. Bilateral Severe Anterior Uveitis and Acute Angle-Closure Glaucoma Following Topiramate Use for Migraine Crisis. Klin. Monbl. Augenheilkd. 2014, 231, 439–441. [Google Scholar] [CrossRef] [PubMed]
  104. Quagliato, L.B.; Barella, K.; Abreu Neto, J.M.; Quagliato, E.M.A.B. Topiramate-Associated Acute, Bilateral, Angle-Closure Glaucoma: Case Report. Arq. Bras. Oftalmol. 2013, 76, 48–49. [Google Scholar] [CrossRef]
  105. Caglar, C.; Yasar, T.; Ceyhan, D. Topiramate Induced Bilateral Angle-Closure Glaucoma: Low Dosage in a Short Time. J. Ocul. Pharmacol. Ther. 2012, 28, 205–207. [Google Scholar] [CrossRef]
  106. Cole, K.L.; Wang, E.E.; Aronwald, R.M. Bilateral Acute Angle-Closure Glaucoma in a Migraine Patient Receiving Topiramate: A Case Report. J. Emerg. Med. 2012, 43, e89–e91. [Google Scholar] [CrossRef]
  107. Van Issum, C.; Mavrakanas, N.; Schutz, J.S.; Shaarawy, T. Topiramate-Induced Acute Bilateral Angle Closure and Myopia: Pathophysiology and Treatment Controversies. Eur. J. Ophthalmol. 2011, 21, 404–409. [Google Scholar] [CrossRef] [PubMed]
  108. Willett, M.C.; Edward, D.P. Refractory Topiramate-Induced Angle-Closure Glaucoma in a Man: A Case Report. J. Med. Case Rep. 2011, 5, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Natesh, S.; Rajashekhara, S.K.; Rao, A.S.D.; Shetty, B. Topiramate-Induced Angle Closure with Acute Myopia, Macular Striae. Oman J. Ophthalmol. 2010, 3, 26–28. [Google Scholar] [CrossRef]
  110. Acharya, N.; Nithyanandam, S.; Kamat, S. Topiramate-Associated Bilateral Anterior Uveitis and Angle Closure Glaucoma. Indian J. Ophthalmol. 2010, 58, 557–559. [Google Scholar] [CrossRef]
  111. Spaccapelo, L.; Leschiutta, S.; Aurea, C.; Ferrari, A. Topiramate-Associated Acute Glaucoma in a Migraine Patient Receiving Concomitant Citalopram Therapy: A Case-Report. Cases J. 2009, 2, 87. [Google Scholar] [CrossRef] [Green Version]
  112. Sbeity, Z.; Gvozdyuk, N.; Amde, W.; Liebmann, J.M.; Ritch, R. Argon Laser Peripheral Iridoplasty for Topiramate-Induced Bilateral Acute Angle Closure. J. Glaucoma 2009, 18, 269–271. [Google Scholar] [CrossRef]
  113. Chalam, K.V.; Tillis, T.; Syed, F.; Agarwal, S.; Brar, V.S. Acute Bilateral Simultaneous Angle Closure Glaucoma after Topiramate Administration: A Case Report. J. Med. Case Rep. 2008, 2, 1. [Google Scholar] [CrossRef] [Green Version]
  114. Boonyaleephan, S. Bilateral Acute Onset Myopia and Angle Closure Glaucoma after Oral Topiramate: A Case Report. J. Med. Assoc. Thai. 2008, 91, 1904–1907. [Google Scholar] [PubMed]
  115. Aminlari, A.; East, M.; Wei, W.; Quillen, D. Topiramate Induced Acute Angle Closure Glaucoma. Open Ophthalmol. J. 2008, 2, 46–47. [Google Scholar] [CrossRef] [Green Version]
  116. Singh, S.K.; Thapa, S.S.; Badhu, B.P. Topiramate Induced Bilateral Angle-Closure Glaucoma. Kathmandu Univ. Med. J. KUMJ 2007, 5, 234–236. [Google Scholar] [PubMed]
  117. Parikh, R.; Parikh, S.; Das, S.; Thomas, R. Choroidal Drainage in the Management of Acute Angle Closure after Topiramate Toxicity. J. Glaucoma 2007, 16, 691–693. [Google Scholar] [CrossRef] [PubMed]
  118. Viet Tran, H.; Ravinet, E.; Schnyder, C.; Reichhart, M.; Guex-Crosier, Y. Blood-Brain Barrier Disruption Associated with Topiramate-Induced Angle-Closure Glaucoma of Acute Onset. Klin. Monbl. Augenheilkd. 2006, 223, 425–427. [Google Scholar] [CrossRef]
  119. Sachi, D.; Vijaya, L. Topiramate Induced Secondary Angle Closure Glaucoma. J. Postgrad. Med. 2006, 52, 72–73. [Google Scholar]
  120. Rhee, D.J.; Ramos-Esteban, J.C.; Nipper, K.S. Rapid Resolution of Topiramate-Induced Angle-Closure Glaucoma with Methylprednisolone and Mannitol. Am. J. Ophthalmol. 2006, 141, 1133–1134. [Google Scholar] [CrossRef]
  121. Levy, J.; Yagev, R.; Petrova, A.; Lifshitz, T. Topiramate-Induced Bilateral Angle-Closure Glaucoma. Can. J. Ophthalmol. 2006, 41, 221–225. [Google Scholar] [CrossRef] [Green Version]
  122. Desai, C.M.; Ramchandani, S.J.; Bhopale, S.G.; Ramchandani, S.S. Acute Myopia and Angle Closure Caused by Topiramate, a Drug Used for Prophylaxis of Migraine. Indian J. Ophthalmol. 2006, 54, 195–197. [Google Scholar]
  123. Mansoor, Q.; Jain, S. Bilateral Angle-Closure Glaucoma Following Oral Topiramate Therapy. Acta Ophthalmol. Scand. 2005, 83, 627–628. [Google Scholar] [CrossRef]
  124. Craig, J.E.; Ong, T.J.; Louis, D.L.; Wells, J.M. Mechanism of Topiramate-Induced Acute-Onset Myopia and Angle Closure Glaucoma. Am. J. Ophthalmol. 2004, 137, 193–195. [Google Scholar] [CrossRef]
  125. Boentert, M.; Aretz, H.; Ludemann, P. Acute Myopia and Angle-Closure Glaucoma Induced by Topiramate. Neurology 2003, 61, 1306. [Google Scholar] [CrossRef] [PubMed]
  126. Medeiros, F.A.; Zhang, X.Y.; Bernd, A.S.; Weinreb, R.N. Angle-Closure Glaucoma Associated with Ciliary Body Detachment in Patients Using Topiramate. Arch. Ophthalmol. 2003, 12, 282–285. [Google Scholar] [CrossRef] [Green Version]
  127. Chen, T.C.; Chao, C.W.; Sorkin, J.A. Topiramate Induced Myopic Shift and Angle Closure Glaucoma. Br. J. Ophthalmol. 2003, 87, 648–649. [Google Scholar] [CrossRef] [PubMed]
  128. Banta, J.T.; Hoffman, K.; Budenz, D.L.; Ceballos, E.; Greenfield, D.S. Presumed Topiramate-Induced Bilateral Acute Angle-Closure Glaucoma. Am. J. Ophthalmol. 2001, 132, 112–114. [Google Scholar] [CrossRef]
  129. Sankar, P.S.; Pasquale, L.R.; Grosskreutz, C.L. Uveal Effusion and Secondary Angle-Closure Glaucoma Associated with Topiramate Use. Arch. Ophthalmol. 2001, 119, 1210–1211. [Google Scholar]
  130. Rhee, D.J.; Goldberg, M.J.; Parrish, R.K. Bilateral Angle-Closure Glaucoma and Ciliary Body Swelling from Topiramate. Arch. Ophthalmol. 2001, 119, 1721–1723. [Google Scholar]
Figure 1. Schematic representation of the relationship between antidepressant and antipsychotic drugs and the various ocular side effects related to glaucoma pathogenic mechanisms. AAP, atypical antipsychotics; IOP, intraocular pressure; AH, aqueous humour; TCA, tricyclic antidepressants; ↑, increased; ↓, decreased.
Figure 1. Schematic representation of the relationship between antidepressant and antipsychotic drugs and the various ocular side effects related to glaucoma pathogenic mechanisms. AAP, atypical antipsychotics; IOP, intraocular pressure; AH, aqueous humour; TCA, tricyclic antidepressants; ↑, increased; ↓, decreased.
Jcm 10 02947 g001
Table 1. Mechanisms of ction of psychopharmacological drugs and their possible effect on intraocular pressure, pupil size and glaucoma risk.
Table 1. Mechanisms of ction of psychopharmacological drugs and their possible effect on intraocular pressure, pupil size and glaucoma risk.
EffectorReceptorsLocationDrugsPossible Induced EffectEffect on IOP
Adrenaline𝛼1Iris dilator muscleCitalopram
Escitalopram
Paroxetine
Fluvoxamine
Fluoxetine
Sertraline
Ziprasidone
Other atypical antipsychotics
Mydriasis
(Hypertension, lid retraction) [33,34,35]
-
𝛼2Ciliary epitheliumEscitalopram,
Paroxetine, Sertraline
Atypical antipsychotics
Increased outflow of aqueous humour↓IOP [33,35]
𝛽2Ciliary epitheliumParoxetineIncrease production of aqueous humour↑IOP [36]
Serotonin5HT7sphincter of the pupil, iris ciliary bodyParoxetine,
Ziprasidone
Mydriasis↑IOP [28,33,37]
5HT1AIris-ciliary bodyEscitalopram, Paroxetine, Fluvoxamine, Sertraline,
Ziprasidone
Other atypical antipsychotics
-↓IOP [28,33,34,35,37]
5HT2AIris-ciliary bodyEscitalopram
Paroxetine,
Fluoxetine
Sertraline,
Venlafaxine,
Ziprasidone
Other atypical antipsychotics
-↓IOP [28,33,37]
5HT2CIris-ciliary bodyCitalopram
Escitalopram,
Fluvoxamine
Fluoxetine, Paroxetine, Sertraline,
Venlafaxine
Ziprasidone, Other atypical antipsychotics
-↑IOP [28,33,35,37]
DopamineDA1The ciliary body, trabecular meshwork, and uveoscleral tissueParoxetineIncreased production of aqueous humour↑IOP
[33,35,38]
DA2Anterior segmentEscitalopram
Paroxetine, Sertraline, Ziprasidone
Typical and other atypical antipsychotics
Suppression of the production of aqueous humour↓IOP
[33,35,38]
Acetylcholine
(miotic effect)
Muscarinic
(Blockade)
Smooth muscle around the pupilCitalopram,
Paroxetine
Escitalopram,
Fluoxetine.
Sertraline
Tricyclic antidepressants
Typical and atypical antipsychotics
Mydriasis↑IOP [36]
TNFTNF-R1Aqueous humour outflow channelsBupropionIncreased caspase activity, mitochondrial dysfunction↑IOP [39]
Sulpha based drugs--TopiramateAllergic reaction (myopia, swelling of the ciliary body, forward displacement of the lens-iris diaphragm)↑IOP [40]
↑, increased; ↓, decreased; IOP, intraocular pressure; 5HT, 5-hydroxytryptamine (serotonin); DA, dopamine; TNF, tumour necrosis factor; R, receptor.
Table 2. A summary of currently reported topiramate-induced acute (primary) angle-closure or acute (primary) angle-closure glaucoma cases (in adult patients).
Table 2. A summary of currently reported topiramate-induced acute (primary) angle-closure or acute (primary) angle-closure glaucoma cases (in adult patients).
Case ReportPatient’s Gender, Age and Other ComorbiditiesOnset after Drug
Initiation
Alzendi et al. (2020) [96]Female, 24 yo, migraines13 days
Agarwal et al. (2019) [97]Female, 25 yo, morbid obesity, obstructive sleep apnoea11 days
Mahendradas et al. (2018) [98]Female, 36 yo, hypothyroidism5 days
Sierra-Rodríguez et al. (2018) [99]Female, 29 yo, epilepsy9 days
Lan et al. (2017) [100]Female, 43 yo, arrhythmia4 weeks
Czyz et al. (2014) [101]Female, 40 yo, arterial hypertension, degenerative disk disease, fibromyalgia, migraines, chronic obstructive pulmonary disease262 days
Pikkel et al. (2014) [102]Male, 54 yo7 days
Katsimpris et al. (2014) [103]Female, 36 yo, migraines14 days
Quagliato et al. (2013) [104]Female, 55 yo, migraines, spasmodic torticollis, essential tremor7days
Caglar et al. (2012) [105]Female, 36 yo, migraine1 day
Cole et al. (2012) [106]Female, 56 yo, depression treated with venlafaxine2 days
van Issum et al. (2011) [107]Male, 34 yo, epilepsy14 days
Willett et al. (2011) [108]Male, 39 yo, arterial hypertension, migraines7 days
Natesh et al. (2010) [109]Male, 23 yo5 days
Acharya et al. (2010) [110]Male, 49 yo14 days
Spaccapelo et al. (2009) [111]Male, 34 yo, anxious-depressive syndrome treated with citalopram7 days
Sbeity et al. (2009) [112]Female, 59 yo, keratomileusis surgery, myopia11 days
Chalam et al. (2008) [113]Female, 34 yo, arterial hypertension, hypothyroidism7 days
Boonyaleephan et al. (2008) [114]Female, 23 yo7 days
Aminlari et al. (2008) [115]Female, 48 yo, bipolar disorder, depression, hypothyroidism, chronic pain14 days
Aminlari et al. (2008)Male, 53 yo, cluster headaches, hyperlipidaemia6 weeks
Singh et al. (2007) [116]Female, 33 yo, headaches7 days
Parikh et al. (2007) [117]Male, 51 yo, epilepsy14 days
Viet et al. (2006) [118]Male, 57 yo, bipolar disorder7 days
Sachi et al. (2006) [119]Female, 33 yo, migraines3 weeks
Rhee et al. (2006) [120]Female, 35 yo, migraines2 months
Levy et al. (2006) [121]Female, 35 yo, depression7 days
Desai et al. (2006) [122]Female, 36 yo, migraines10 days
Mansoor et al. (2005) [123]Female, 51 yo, surgery for hypermetropia, migraines7 days
Craig et al. (2004) [124]Female, 25 yo, epilepsy, depression treated with Venlafaxine7 days
Boentert et al. (2003) [125]Female, 23 yo, congenital hydrocephalus, Arnold-Chiari formation I, partial atrophy of the right optic nerve, astigmatism, vertical strabismus.6 days
Medeiros et al. (2003) [126]Male, 44 yo5 days
Medeiros et al. (2003) [126]Female, 42 yo, myopia10 days
Chen et al. (2003) [127]Female, 42 yo, hypertension, seizures2.5 weeks
Banta et al. (2001) [128]Male, 51 yo, bipolar disorder14 days
Sankar et al. (2001) [129]Female, 34 yo, depression14 days
Sankar et al. (2001) [129]Female, 53 yo, depression treated with venlafaxine, high cholesterol.10 days
Rhee et al. (2001) [130]Female, 43 yo, depression treated with paroxetine1 day
yo, years old.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ciobanu, A.M.; Dionisie, V.; Neagu, C.; Bolog, O.M.; Riga, S.; Popa-Velea, O. Psychopharmacological Treatment, Intraocular Pressure and the Risk of Glaucoma: A Review of Literature. J. Clin. Med. 2021, 10, 2947. https://doi.org/10.3390/jcm10132947

AMA Style

Ciobanu AM, Dionisie V, Neagu C, Bolog OM, Riga S, Popa-Velea O. Psychopharmacological Treatment, Intraocular Pressure and the Risk of Glaucoma: A Review of Literature. Journal of Clinical Medicine. 2021; 10(13):2947. https://doi.org/10.3390/jcm10132947

Chicago/Turabian Style

Ciobanu, Adela Magdalena, Vlad Dionisie, Cristina Neagu, Otilia Maria Bolog, Sorin Riga, and Ovidiu Popa-Velea. 2021. "Psychopharmacological Treatment, Intraocular Pressure and the Risk of Glaucoma: A Review of Literature" Journal of Clinical Medicine 10, no. 13: 2947. https://doi.org/10.3390/jcm10132947

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop