Abstract
Depression is the fourth most serious disease in the world. Left untreated, it is a cause of suicide attempts, emergence or exacerbation worsening of serious diseases, bodily and mental disorders, as well as increased risk of cardiovascular diseases, stroke, diabetes, and obesity, as well as endocrine and neurological diseases. Frequent coexistence of depression and other diseases requires the simultaneous use of several drugs from different therapeutic groups, which very often interact and intensify comorbidities, sometimes unrelated mechanisms. Sufficient awareness of potential drug interactions is critical in clinical practice, as it allows both to avoid disruption of proper pharmacotherapy and achieve substantive results. Therefore, this review aims to analyze the interactions of antidepressants with other concomitant medications. Against the backdrop of experimental research and a thorough analysis of the up-to-date literature, the authors discuss in detail the mechanisms and effects of action of individual drug interactions and adaptogens, including the latest antidepressants.
Keywords: Depression, Antidepressants, Drug-drug interactions, Polypharmacotherapy
Introduction
According to the World Health Organization, depression is the fourth most serious disease in the world and one of the leading causes of suicide, appearing more and more often in all age groups. It is predicted that 2030 it will be the most common worldwide spread disorder [1]. Although the leading causes of depression remain vastly unexplored, it appears to be linked to various global circumstances, such as terrorism, pandemics, economic crises, and war [2]. In many cases, depression is seldom accompanied by other disorders, such as cardiovascular diseases [3], Parkinson's disease [4], epilepsy [5], and polycystic ovary syndrome (PCOS) [6]. Up-to-date studies indicate the coexistence of depression with somatic diseases. People suffering from depression are also exposed to cardiovascular diseases, stroke, diabetes, and obesity [7]. Such correlation is evident in older patients [8], while the depression of young people appears to be linked to thyroid diseases, type 2 diabetes, other endocrine diseases, as well as neurological diseases [9]. Untreated depression leads to suicide attempts [A1], the emergence or worsening of severe diseases, bodily and mental disorders, as well as numerous social and economic problems. Against this backdrop, it is agreed that major depression requires pharmacological treatment, which is a long-term process that should be continued for 6–12 months. In about 50–70% of cases, the first used antidepressant indicates positive results. However, the remaining patients require additional or alternative medication due to the ineffectiveness of the first attempt drug or experienced side effects [10]. Many patients also self-medicate, taking increasingly advertised and readily available adaptogens.
Simultaneous treatment of depression and other diseases leads to the use of several drugs from different therapeutic groups [8, 9]. At the same time, antidepressants are known for their everyday interaction with other medications and causing hurt comorbidities (drug-disease interactions), posing a threat to the health and life of patients. Therefore, one of the most severe problems of modern pharmacotherapy is the selection of appropriate therapy in patients treated for various diseases that require multidrug treatment [8, 9]. This applies to unfavorable interactions between concurrently used drugs and the resulting clinical effects. These may occur through unrelated mechanisms. Antidepressants can interact with other medications, primarily through pharmacokinetic processes affecting their absorption, distribution, metabolism, and excretion, which may alter their effectiveness or lead to toxicity. These interactions also involve pharmacodynamic mechanisms, such as synergism or antagonism, impacting drug efficacy and potential toxicity. There is a complex and not yet fully understood relationship between antidepressants and the drug efflux pump. This compound may significantly affect the therapeutic effectiveness of antidepressant treatment. P-glycoprotein plays an essential role in the functioning of the blood–brain barrier by selectively extruding drugs, thereby limiting the ability of its substrates to reach the brain [11]. On the one hand, this protein may limit the uptake of antidepressants into the brain, weakening antidepressant therapies. Drugs or other substances that are inhibitors or inducers of P-glycoprotein may also modulate the pharmacological activity of antidepressants. If other drugs simultaneously inhibit the transport protein, the penetration of the substrate drug increases, and its effect is enhanced. In case of excessive penetration of drugs through the blood–brain barrier, the side effects of antidepressants may be intensified, including serotonin syndrome, neurotoxicity, etc. However, controlled co-administration of transport protein inhibitors with antidepressants in antidepressant-resistant patients may represent a new therapeutic approach for the treatment of depression. In turn, drugs or other substances that are P-glycoprotein inducers may block the penetration of antidepressants into the central nervous system. They may weaken or even eliminate the antidepressant effect [12] P-glycoprotein in vitro, which may play a role in their therapeutic effect.
In clinical practice, a clear understanding of potential drug interactions is essential to avoid those that could interfere with the proper course of the pharmacotherapy process, as well as to use some interactions to benefit the patients. The currently available scientific literature does not provide a comprehensive summary of the interactions of all groups of antidepressants with other concomitantly taken medications or foods. The available literature lacks comprehensive reports on the interaction of antidepressants with other drugs. Therefore, the aim of this review is a thorough analysis of the interaction of antidepressants, taking into account the mechanisms of formation and side effects.
Interactions of tricyclic antidepressants (TCAs)
Tricyclic antidepressants (TCAs), introduced for the treatment of depression in the 1950s, are now rarely used due to numerous side effects [13]. The mechanism of action of these drugs is blocking the reuptake of neurotransmitters—norepinephrine and serotonin by nerve cells, which leads to an increase in the concentration of these transmitters in synaptic clefts. They also act as antagonists of muscarinic, histamine H1, and postsynaptic α1 and α2 adrenergic receptors [14]. TCAs interactions include primarily the risk of serotonin syndrome when taken together with monoamine oxidase inhibitors (MAOI) and selective serotonin reuptake inhibitor (SSRI) medications [15] (Table 1). There is also a risk of serotonin syndrome when tricyclic antidepressants are taken concomitantly with linezolid, tramadol, ondansetron, lithium compounds, fentanyl, buspirone, and preparations containing St. John's wort [16]. The serotonin syndrome is a phenomenon that directly threatens human life. It includes autonomic overactivity, a change in the patient's mental state, hypomania, normal neuromuscular tension, restlessness, confusion, myoclonus, and an increase in the patient's body temperature [17].
Table 1.
Interactions of TCAs with other medications drug classes and food
| Interactions of TCAs with other drugs classes | |||
|---|---|---|---|
| Drug | Interacting substance | Result of interaction | References |
| TCAs |
Ezyme CYP1A2 inhibitors: Cimetidine Tacrolimus Warfarin Antipyrine Verapamil Theophylline Diltiazem Ciprofloksacin Methoxsalen Norfloksacin Mexiletine Norethindrone Caffeine Nalidixic acid Citalopram Miodarone Clarithromycin Fluoxetine Erythromycin Melatonin Ethinylestriadol Fluvoxamine Isoniazid Troleandomycin Paroxetine Ticlopidine Tacrine Oral anticonceptives Grape juice Cumin |
Increased risk of side effects of TCAs | [15, 19] [16] |
|
Enzyme CYP 1A2 inductors: Carbamazepine Omeprazole Primidone Barbiturates Rifampicin |
Shorten the effect of the TCAs increase the effect of that batch of drugs that have undergone biotransformation in the body to their active metabolites | ||
| TCAs |
SSRI MAOI Linezolid Tramadol Ondasetron Lithium compounds Fentanyl Buspirone St. John’s wort |
Serotonin syndrome | [15–18, 21–26] |
|
Drugs that affect cardiac conduction Antiarrhythmic drugs diuretic Phenothiazine drugs Antipsychotic Proconvulsant |
Decrease the seizure threshold, loss of control over epileptic attacks | ||
|
Anticholinergic Sedative drugs |
Dxacerbate such side effects as confusion, constipation and bowel problems and visual disturbances | ||
|
Atropine Scopolamine |
Delirium | ||
|
Antimuscarinic substances peripheral Central anticholinergic |
Urinary retention, memory impairment, confusion, blurred vision, intestinal obstruction, dry mouth, glaucoma and constipation | ||
| CNS depressants | Deterioration of psychomotor performance and may also adversely affect cognitive and psychomotor functions | ||
| Epinephrine | Promoting an increase in blood pressure and cardiac arrhythmias | ||
| Cholestyramine | Significant decrease in the concentration of TLPD in the body of patients and the subsequent lack of pharmacological effect | ||
| Oral hormonal therapy Estrogens | Incidents of akathisia and a decrease in the potential and exacerbation of toxicity of TLPD | ||
| Enalapril | Incidents of accommodative dysfunction and blurred vision | ||
| Fluoroquinolones | Significant increase in TLPD and subsequent patient cardiac dysfunction | ||
| Halothane | Increased risk of hyperthermia | ||
| Antidiabetic drugs | Subsequent incidents of hypoglycemia and decrease in hypoglycemic potential | ||
| Levothyroxine | Incidents of tachycardia | ||
|
Melatonin Methadone Quetiapine |
Pathological increase in the patient's sedation | ||
| Pancuronium | The possibility of hyperthermia | ||
| Coffee or tea | Reducing effectiveness of drugs | ||
Cytochrome CYPA1A2 plays a significant role in the metabolism of drugs in this group [18]. Concurrent administration of TCAs and medicines or foods that are inhibitors or inducers of the CYPA1A2 isoenzyme may impair the metabolism of antidepressants. The use of isoenzyme inhibitors may increase the plasma concentration of tricyclic antidepressants and the intensification of their side effects. In contrast, enzyme inducers may weaken their impact and increase the concentration of their active metabolites [16]. TCA should be used with great caution in people who take drugs that affect cardiac conduction, antiarrhythmic drugs, diuretics, and phenothiazine drugs.
Moreover, individuals taking these drugs and antipsychotic and proconvulsant substances concurrently should also receive special monitoring and caution due to the ability of tricyclic antidepressants to decrease the seizure threshold [16]. In the case of maprotiline and clomipramine, the occurrence of this effect may lead to loss of control over epileptic seizures in people suffering from this condition [19]. Moreover, nortriptyline and imipramine increase the concentration of phenytoin in the body of epilepsy patients, which is related to the inhibitory effect of these antidepressants on the microsomal subunit of cytochromes CYP2C19 and CYP2D6. These processes lead, among others, to inhibit the reaction, resulting in p-hydroxylated phenytoin, indicating that both antidepressants act as competitive substrates for phenytoin [20].
As a result of the combination of TCAs with antimuscarinic substances, peripheral and central anticholinergic effects may be intensified, which may result in urinary retention, memory impairment, confusion, blurred vision, intestinal obstruction, dry mouth, glaucoma, and constipation {Citation} [16, 18]. The use of a combination of the tricyclic antidepressant amitriptyline and L-DOPA indicates a favorable interaction between the two drugs, which carries an increased antiparkinsonian effect compared to conventional therapy with L-DOPA alone [21].
Moreover, TCAs used in combination with central nervous system (CNS) depressants may lead to deterioration of psychomotor performance and may also adversely affect cognitive and psychomotor functions [22]. The combination of TCAs with antipsychotic drugs may lead to toxic concentrations of antidepressants in the body, which is related to the inhibition of the cytochrome subunit CYP2D6 [22]. TCAs block the reuptake of antihypertensive drugs by adrenergic neurons, which leads to the absence or weakening of the effects of drugs such as bretylium, guanethidine, betanidine, and debrisoquine. In addition, their interaction with α2 adrenergic receptors can attenuate the effect of drugs such as methyldopa and clonidine. On the other hand, drugs belonging to the calcium antagonist group, such as diltiazem and verapamil, enhance the pharmacological effect and overall bioavailability of imipramine. This effect results from these drugs competing as substrates with the cytochrome subunit CYP3A4 [22].
Studies have shown that the antifungal drug ketoconazole blocks the demethylation of imipramine while not affecting desipramine hydroxylation [22]. An increase in nortriptyline concentration was observed with simultaneous administration of fluconazole [22]. The concurrent use of warfarin and amitriptyline may cause hemorrhagic reactions [22]. Concomitant intake of yohimbine and TCAs can lead to hypertension. The discussed effect is even more potent in patients who already have hypertension [23]. Reducing the effectiveness of antidepressant therapy and possible intensification of adverse side effects may occur when consuming herbs such as Kava kava or the St. John's Wort [24]. In addition, laxative herbs such as Plantago spp. Plants, konjac, or guar gum can delay gastric emptying and diminish drug absorption [25]. Additionally, the cytochrome CYP1A2 subunit responsible for metabolizing procarcinogens contained in cigarette smoke can be induced by these substances, thereby altering the metabolism of tricyclic antidepressants [26]. Foods that affect the function of this enzyme include grilled meat, broccoli, tobacco, and brussels sprouts [24].
Interactions of SSRIs
Selective serotonin reuptake inhibitors (SSRIs) enhance the level of neurotransmitters such as serotonin, dopamine, and norepinephrine. SSRIs selectively affect the serotonergic system by inhibiting serotonin reuptake, which improves serotonin levels in the space between neurons and prolongs their action in the brain. An increase in serotonin levels occurs approximately two weeks after starting SSRI therapy [27]. The cytochrome P450 in the liver metabolizes SSRIs. This group of antidepressants can inhibit some CYP450 enzymes; thereby, drug-drug interactions can occur (Table 2).
Table 2.
Effects of selected SSRI drugs on isoenzyme inhibition
| Effects of selected SSRI drugs on isoenzyme inihibition | References | |||||
|---|---|---|---|---|---|---|
| CYP2D6 | CYP2C9 | CYP2C19 | CYP3A4 | CYP1A2 | ||
| Fluoxetine/norfluoxetine | Dominant | Limited | Moderate | Moderate | [3, 28–31] | |
| Paroxetine | Decided | |||||
| Sertraline | Dose-epended | |||||
| Fluvoxamine | Limited | Strong | Limited | Strong | ||
| Citalopram | Feeble | Irrelevant | Irrelevant | Irrelevant | ||
| Citalopram | Feeble | Irrelevant | Irrelevant | Irrelevant | ||
Based on available studies, severe depression symptoms can involve 45% of patients who have coronary artery disease (CAD), and circa 20% of them can have all the parameters of a full depressive episode. Woroń et al. analyzed 66 cases. Most antidepressants and cardiac drug interactions involved SSRIs. Bradycardia was the side effect that occurred the most often after the combination of SSRI and metoprolol or propranolol. In one case, the consequence of a combination of fluoxetine and propranolol was sudden cardiac arrest. Antidepressants inhibit the activity of CYP2D6—the enzyme that metabolizes metoprolol and propranolol the most [3]. Of the SSRIs, venlafaxine and sertraline have the least severe side effects in combination with metoprolol and propranolol. In case of use of other drugs of SSRI or serotonin and norepinephrine reuptake inhibitors (SNRI) group, metoprolol, and propranolol should be exchanged into nebivolol or carvedilol because they have a lower risk of interactions [3]. Except for the above study, there are available studies describing the case of a 63 years woman who had anterograde block after a combination of metoprolol and paroxetine [28] and the case of a 53 years man who had the same side effect after a combination of propranolol and fluoxetine [29]. A similar mechanism to that observed with SSRIs and β-blockers concerns propafenone and β-blockers. Propafenone is metabolized by CYP2D and 3A4 [3]. Case reports have documented instances of worsened propafenone tolerance (bradycardia, prolongation of QT) following its combination with citalopram [30]. Another group in which were observed side effects was a combination of SSRIs and calcium channel blockers. Side effects of calcium channel blockers increased, with cases of acute renal failure (after the combination of fluoxetine and amlodipine) and cases of myalgia, polyuria, elevated levels of transaminases (combination of SSRIs and lercanidipine), and one case of severe bradycardia (combination of fluvoxamine and diltiazem) [3]. The literature also describes the occurrence of edema, headaches, and weight gain after combining fluoxetine with verapamil [31]. These interactions between SSRIs and calcium channel blockers appear primarily pharmacokinetic, involving competition for the same CYP450 isoenzyme or inhibiting the isoenzyme responsible for metabolizing calcium channel blockers SSRIs [3].
SSRIs block the reuptake of serotonin from the platelets. As a result, the inhibitory of platelet aggregation occurs, and it can cause an increased risk of bleeding [32] in patients who take other drugs to decrease coagulation. This risk concerns patients who take antiplatelet drugs (acetylsalicylic acid, clopidogrel, ticagrelor), as well as patients taking anticoagulants, which have another mechanism of action (warfarin, dabigatran, rivaroxaban, acenocoumarol, apixaban, heparin). During the simultaneous treatment of SSRIs and warfarin, the additional hemorrhagic risk may be connected with inhibiting warfarin metabolism (CYP 450: 2C9, 2C19, 3A4) and its increased level in the blood. In most cases, this interaction may occur during the treatment of fluvoxamine, fluoxetine, and paroxetine, while minor cases may occur with sertraline. Moreover, fluvoxamine and fluoxetine block CYP2C19 and consequently reduce the antiplatelet effects of clopidogrel by inhibiting clopidogrel's transformation into an active metabolite. According to Woroń et al., the combination of anticoagulants, such as acenocoumarol, apixaban, acetylsalicylic acid, rivaroxaban, and SSRIs, such as citalopram, escitalopram, fluoxetine, fluvoxamine, sertraline, and venlafaxine may increase the risk of hemorrhagic complication syndrome, including upper or lower gastrointestinal bleeding [33]. An increased incidence of hemorrhagic complications is also observed with concomitant use of SSRIs with glucocorticosteroids and non-steroidal anti-inflammatory drugs [33]. Treatment of vitamin K antagonists (VKA) and SSRIs simultaneously may be associated with an increased risk of bleeding. There are three known mechanisms which may correspond to the above occurrence. Firstly, it is due to the inhibition of serotonin reuptake in platelets by SSRIs, which worsens their function and may cause an increased risk of bleeding. Secondly, SSRIs may increase the secretion of gastric acid, which may increase the risk of peptic ulcer disease and bleeding from the upper gastrointestinal tract. Third, inhibition of CYP2C9 may lead to a high international normalized ratio (INR) and an increased risk of bleeding [34]. SSRIs taken simultaneously with high doses of loop diuretics and thiazides may lead to hyponatremia, which is particularly dangerous in elderly patients [3]. Hyponatremia may occur as a result of thiazide diuretics with citalopram or escitalopram, as well as diuretics with sertraline fluoxetine, and fluvoxamine [33]. The mechanism of this side effect may be associated with the risk of summing hyponatremia, which stems from the action of the diuretic and the risk of induction of the syndrome of inappropriate antidiuretic hormone hypersecretion connected with blockade of serotonin reuptake [3]. Esomeprazol and omeprazole inhibit CYP 2C19, leading to the increased blood concentration of citalopram and escitalopram and subsequently amplifying their adverse effects. Moreover, esomeprasol inhibits CYP 3A4, potentially increasing sertraline concentration and its side effects. Fluoxetine inhibits isoenzymes CYP 2C9, 2C19, and 3A4, leading to increased lansoprazole concentration and the risk of side effects. A similar effect is observed with fluvoxamine, which inhibits 2C19 and 3A4 [33]. Concomitant use of itraconazole, fluconazole, ketoconazole, and miconazole with venlafaxine can increase its side effects by inhibiting CYP3A4, leading to elevated venlafaxine blood concentrations. Fluconazole’s inhibition of CYP2C19 and 3A4 may heighten adverse reactions to citalopram and escitalopram [33]. Citalopram and escitalopram can prolong the QT interval on electrocardiogram (ECG) by affecting potassium channels, potentially causing ventricular arrhythmia when co-administered with anti-arrhythmic drugs like amiodarone, propafenone, and sotalol. Statins, especially simvastatin, atorvastatin, and lovastatin, may interact with SSRIs by inhibiting CYP3A4, increasing the risk of statin-related side effects such as myopathy, myalgia, or rhabdomyolysis, particularly when combined with fluvoxamine [3, 35]. Fluoxetine strongly inhibits the CYP 3A4 isoenzyme and, as a result, may increase aripiprazole concentrations by up to 45%, thereby increasing the risk of adverse effects. Fluvoxamine inhibits CYP 2D6 and 3A4, resulting in an increase in aripiprazole concentrations and the need for a 50% dose reduction [33]. Detailed interactions of SSRIs are presented in Tables 3 and 4.
Table 3.
Interactions of SSRIs with other medications drug classes
| Interactions of SSRIs with other drug classes | |||
|---|---|---|---|
| Drug | Interacting substance | Result of interaction | References |
| Citalopram/escitalopram | Itopride | Risk of late diskinesis | [3, 32–34] |
| Clonidine | Cases of hyponatremia and sedation | ||
| Lamotrigine | Cases of myoclonus | ||
| Drugs that prolong QT interval | Increased risk of cardiac conduction and rhythm disturbances, including ventricular arrhythmias | ||
| Levomepromazine | Increased risk of side effects of citalopram or escitalopram | ||
| Melatonin |
exacerbation of its effects and increased risk of side effects |
||
| Rasagiline | Greater risk of disturbance of consciousness | ||
| Rifampicine | Decreased in efficacy of citalopram or escitalopram | ||
| Risperidon | Cases of ventricular arrhythmias | ||
| Activated carbon | Decreased absorption of drugs from the gastrointestinal tract | ||
| Fluoxetine | Amlodipine | Cases of acute renal failure and peripheral oedema | |
| Bupropion | Cases of unconsciousness | ||
| Buspirone | Reduced anti-anxiety effects, cases of akathisia, dystonia | ||
| Phenytoin | Increased side effects and toxicity and reduced efficacy of fluoxetine | ||
| Lamotrigine | Cases of delirium | ||
| Lercanidipine | Cases of hypotention | ||
| Lurasidone | Cases of pharyngeal dystonia | ||
| Metoprolol | Cases of bradycardia and conduction blocks | ||
|
Nifedipine Verapamil |
Cases of oedema, nausea, hot flashes | ||
|
Olanzapine Propafenone Haloperidol Iloperidol Alprazolam Atomoxetine Cyclosporine Diazepam Valproate |
Increased risk of side effects | ||
| Propranolol | Cases of bradycardia, conduction blocks and cardiac arrest | ||
| Selegiline | Increased risk of ataxia toxicity, increased blood pressure | ||
| Tamoxifen | Decreased efficacy | ||
| Tramadol | Decreased analgesic effect | ||
| Carbamazepine | Increased risk of side effects, hyponatremia and cases of blurred vision, tremors | ||
| Clarithromycin | Delirium | ||
| Zolpidem | Increased risk of hallucinations | ||
| Fluvoxamine |
Agomelatine Phenytoin Haloperidol Clozapine Tianisidine Alprazolam Asenapine Buspirone Cyclosporine Diazepam Diltiazem Fexofenadine Phenothiazine Caffeine Quetiapine |
Increased risk of side effects | |
| Celecoxib | Cases of agitation and delirium | ||
| Carbamazepine |
Increased side effects, decreased efficacy of fluvoxamine, risk of hyponatremia |
||
| Levomepromazine | Cases of convulsions | ||
| Lidocaine | Increased toxicity | ||
| Olanzapine | Increased risk of side effects, including convulsions, sedation, hypotonia, tachycardia, hepatotoxicity | ||
| Piracetam | Decreased its excretion, resulting in increased blood concentrations | ||
| Theophylline | Increased toxicity | ||
| Topiramate | Cases of tremors and myoclonus | ||
| Zolpidem | Increased risk of hallucinations and memory impairment | ||
| Sertraline | Amlodipine | cases of headache and oedema | |
| Cisapride | cardiac arrhythmias | ||
| Phenytoin |
Decreased sertraline concentration and efficacy, increased risk of phenytoin toxicity |
||
| Carbamazepine |
Decreased sertraline concentration and efficacy, increased carbamazepine concentrations and side effects |
||
| Lamotrigine | Cases of fatigue, sedation, cognitive impairment | ||
| Lercanidipine | Cases of myalgia and polyuria | ||
| Oxycodone | Cases of hallucinations and tremors | ||
| Venlafaxine |
Amiodarone Bupropion Celecoxib Diphenhydramine Duloxetine Erythromycin Phenothiazines Haloperidol Indinavir Clarithromycin Mexiletine Methadone Nilotinib Pindolol Risperidone Ritonavir Terbinafine Ticlopidine |
Increased concentration of vanlafaxine in blood and risk of side effects | |
| Amoxicillin with clavulanic acid | Cases of serotonin syndrome | ||
| Clozapine | Increased concentration of vanlafaxine in blood and risk of side effects, increased risk of tachycardia and blood pressure changes | ||
| Metoprolol | Decreased blood pressure lowering efficacy of metoprolol, increased venlafaxine concentrations and side effects | ||
| Piracetam | Decreased excretion of piracetam, resulting in an increase in blood concentrations of the drug | ||
| Propafenone | Cases of psychosis, bradycardia, dizziness, visual disturbances | ||
| Propranolol | Cases of increased urinary urgency and increased muscle tone | ||
Table 4.
Interactions of SSRI drugs with plant substances
| Interactions of selected SSRI drugs with plant substances | |||
|---|---|---|---|
| Drug | Interacting substance (herb) | Result of interaction | References |
| Citalopram, escitalopram, fluoxetine, fluvoxamine, venlafaxine, sertraline |
Ginkgo biloba Milk thistle Rhodiola rosea |
Increased risk of bleeding Coughing, ejaculatory disorders Myalgia, cardiac arrhytmias |
[40–43] |
| Citalopram or escitalopram, fluoxetine, fluvoxamine | Panax ginseng | Ejaculatory disorders, priapism and an increased risk of serotonergic syndrome | |
| Fluoxetine |
Bacopa monnieri Purple coneflower Rhiodola rosea and additionally duloxetine |
Increased risk of serotonergic syndrome Sudden hair loss and priapism Cases of diarrhoea, jaundice and hepatotoxicity |
|
| Sertraline |
Milk thistle and agomelatine Panax ginseng Starfruit |
Hepatotoxicity Chest pain, tachycardia, cortical arrhthmias Increased risk of serotonine syndrome, cases of bruxism, headaches |
|
| Venlafaxine |
Jujuba Panax ginseng |
Increased risk of serotoninergic syndrome Increased the risk of hyperhidrosis and itching of the skin |
|
Interactions of tetracyclic antidepressants
Maprotiline is a drug included in the group of quadricyclics used to treat depression, which very strongly inhibits the reuptake of norepinephrine, thereby increasing its concentration in the synaptic gap [36, 37]. Available literature also reports that it may prove effective in eradicating Francisella infection as an antiviral agent through its ability to decrease the virulence index of IgIC [38]. Moreover, due to its anabolic nature, it can be used by people who have anorexia, underweight, and those in recovery [39]. The cytochrome CYP2D6 subunit mainly metabolizes maprotiline, and concomitant administration of inhibitors or inductors carries the risk of drug adverse effects [40] (Table 5) [41–43].
Table 5.
Interactions of tetracyclic antidepressants with other medications. drug classes
| Interactions of tetracyclic antidepressants with with other drug classes | References | ||
|---|---|---|---|
| Drug | Interacting substance | Result of interaction | |
| Tetracyclic antidepressants (maprotiline) |
Ezyme CYP1A2 inhibitors: Diltiazem Celecoxib Moclobemide Bupropion Metoclopramide Encapone Yohimbine Methadone Codeine Dextropropoxyphene Esipramine Imipramine Clomipramine Lobeline Metoclopramide Tricyclic antidepressants Risperidone |
Elevation of the plasma level of tetracyclic antidepressants, increased cholinergic effects | [44–46] |
|
Enzyme CYP2D6 inductors: Griseofulvin Rifabutin Rifampicin Sulfadimidine Nafcillin Troglitazone Ethosuximide Carbamazepine Phenytoin Phenobarbital Nevirapine Nelfinavir Progesterone Refexib Phenylbutazone Dexamethasone |
Shorten the effect of the tetracyclic antidepressants, increase the effect of that batch of drugs that have undergone biotransformation in the body to their active metabolites | ||
| Tetracyclic antidepressants (maprotiline) | Dextromethorphan | Development of cardiomyopathy | |
|
Ritonavir Fluconazole |
Orthostatic hypotension | ||
| Phenytoin | Ventricular tachycardia | ||
| Betanidine | Increased lack of control over normal blood pressure | ||
| Reboxetine | Serotonin syndrome | ||
| Desmopressin |
Increase in water retention in the body, Pathological state of hyponatremia |
||
| Verapamil | Development of severe heart failure | ||
A case of cardiogenic shock associated with Takotsubo cardiomyopathy has also been reported during the concomitant administration of maprotiline and dextromethorphan. The development of cardiomyopathy was closely related to the presence of serotonergic interactions between the two drugs, which resulted later in the occurrence of serotonin syndrome. A woman aged 65 was treated with maprotiline due to the presence of depressive symptoms. The initial dose was 10 mg/24 h and was increased to 30 mg/24 h. Later, due to bronchitis, dextromethorphan and amoxicillin were included in her therapy. Three days after the list of medications was expanded, the woman developed symptoms such as hemoptysis, shock, severe dyspnoea, and acute agitation. An X-ray taken after admission showed lower lobe consolidation, ST-segment elevation, and pulmonary congestion. Other tests showed elevated levels of creatine kinase-MB and creatine kinase, basal hyperkinesis, apical akinesis, apical ballooning, and an elevated c-reactive protein (CRP) index. After intubating the patient and administering norepinephrine and dobutamine, she was diagnosed with Takotsubo cardiomyopathy. After taking the necessary measures, she was diagnosed with serotonin syndrome the next day. It was decided to exclude maprotiline from therapy and include cyproheptadine and midazolam. After this regimen, the patient’s temperature dropped, and hemodynamic indicators were regulated to safe levels [44].
Episodes of ventricular tachycardia have also been documented to affect the development of severe heart failure when maprotiline was combined with drugs used for epileptic seizures. The woman, aged 69, had been on therapy with glycoside and verapamil—antiarrhythmic drugs—for 13 years to normalize her atrial fibrillation and on long-term maprotiline therapy to treat her depression. The day before the onset of adverse and chewing-threatening symptoms, the woman used phenytoin to normalize the epileptic seizures she was experiencing. After the patient was admitted to the ward and the necessary examinations were performed, she was shown to have torsade de pointes tachycardia, reduced ejection fraction, coronary artery obstruction, left ventricular dilatation, and atherosclerotic plaques. After appropriate interventions, including defibrillation, the patient was administered xylocaine and magnesium, and maprotiline and antiarrhythmic medications were discontinued. After the implementation of these treatments, the characteristic tachycardia stopped, and the QT interval shortened. The occurrence of these symptoms results from possible drug interactions between antidepressants, arrhythmogenic drugs, and existing diseases. This case illustrates that the use of multidrug therapy with drugs of a narrow therapeutic range should be strictly controlled and subject to continuous observation [45]. Concurrent use of maprotiline and drugs that block adrenergic neurons can increase the risk of losing control over normal blood pressure. This interaction arises from competition between the two drug types for uptake into vesicles located in sympathetic neurons. In one study, patient subjects experienced a notable rise in blood pressure after receiving two 25 mg of maprotiline. Other clinical cases reported significant blood pressure dysregulation in patients receiving maprotiline doses ranging from 25 to 75 mg thrice daily. The introduction of maprotiline into the therapy of the time in four patients disrupted normal blood pressure in one patient [46].
Studies with animals also show evidence of interactions between maprotiline and drugs with central nervous system depressant effects [46]. The occurrence of life-threatening serotonin syndrome has also been described with the concomitant use of venlafaxine 300 mg/24 h, reboxetine 8 mg/24 h, and maprotiline 150 mg/24 h. A man undergoing such therapy developed serotonin syndrome. He experienced symptoms such as partial confusion, mutism, confusion, negativism, non-Parkinsonian rigidity, tremors in the upper extremities, episodes of psychomotor agitation, excessive sweating, hyper-responsiveness to bone-tendon reflexes, and hyperreflexia. The patient’s blood pressure was unstable and variable, 140–105 – 125–80 mmHg. To abolish the symptoms of serotonin syndrome, the drug treatment of the time was withdrawn, and diazepam therapy was introduced [47, 48].
The simultaneous use of maprotiline and desmopressin may result in a general increase in water retention in the body, which may, in turn, lead to the development of the pathological condition of hyponatremia. In severe cases, this condition can result in life-threatening seizures, coma, or death [49]. Clinical case reports report an increased risk of dangerous bronchospasm with concomitant use of maprotiline, as well as phenelzine, nortriptyline, and desipramine. A 36 year-old man was treated with maprotiline 200 mg/24 h due to the occurrence of depressive episodes. The therapy carried therapeutic success. The man had no clinical history of respiratory problems or allergies present, but he developed dangerous breathing problems—wheezing, shortness of breath, and intractable and increasing respiratory distress. After taking bronchodilators, the adverse symptoms subsided. After maprotiline was experimentally excluded from the therapy at the time, the respiratory problems disappeared as the patient's eosinophilia levels decreased. When attempting to re-include maprotiline at a lower dose of 150 mg/24 h, the dangerous and bothersome respiratory symptoms returned. The patient received phenelzine, which resulted in therapeutic success without shortness of breath or breathing problems [50]. A single study suggests the occurrence of Oglivi syndrome, an acute pseudo-obstruction of the colon caused by maprotiline used in co-therapy with rosuvastatin and clopidogrel [51].
Interactions of serotonin-norepinephrine reuptake inhibitors (SNRIs)
Selective serotonin and norepinephrine reuptake inhibitors SNRIs [52] mode of action involves a reuptake block of the neurotransmitters norepinephrine and serotonin – hence the reference to ‘dual-action’ drugs. The exact mechanism of action is based on the binding of norepinephrine transporter (NAT) and serotonin transporter SERT. They have no affinity for other receptors. Due to their nature, they cause an elevation of the overall level of the neurotransmitter dopamine in the prefrontal cortex [16]. These drug classes include venlafaxine, desvenlafaxine, milnacipran, duloxetine, and mirtazapine [53]. Duloxetine exhibits relatively unidirectional binding to both transporters, but the binding strength predominates toward serotonin. Venlafaxine shows significantly higher binding strength to the serotonin transporter than to the norepinephrine transporter, an effect that largely depends on the drug dose used. Milnacipran, on the other hand, shows the most equal affinity for both transporters compared to the drugs mentioned above [16]. Mirtazapine is characterized by its ability to raise neurotransmitters serotonin and norepinephrine levels. This effect occurs through an inhibitory effect on α-2 adrenergic heteroreceptors and autoreceptors. The drug also binds highly to histaminergic receptors [22].
The metabolism of duloxetine and mirtazapine mainly involves the cytochrome CYP2A1 subunit [26]. The drug may also act as an inhibitor of the cytochrome CYP2D6 subunit, which carries the risk of increasing the concentration in the body of drugs (Table 6). Also, medications from the SSRI group of selective serotonin reuptake inhibitors affect the concentration of duloxetine in the human organism. Paroxetine and fluvoxamine have been observed to raise the overall concentration of duloxetine in the body [16]. Studies show that mirtazapine is characterized by its ability to inhibit cytochrome CYP3A4, CYP2D6, and CYP1A2 subunits minimally.
Table 6.
Interactions of antidepressants with SNRIs
| Interactions of SNRIs with other drug classes | |||
|---|---|---|---|
| Drug | Interacting substance | Results of interaction | References |
| SNRI | Benzodiazepines | Risk of impaired motor skills | [16, 22, 33, 54–63, 65, 67, 68] |
|
Calcineurin inhibitors Lithium inhibitors |
Serotonin syndrome | ||
| Duloxetine | Anticoagulant | Change in INR | |
| Gastric juice antacids | Diarrhoea | ||
| Venlafaxine | Propafenone | Visual and auditory hallucinations | |
| Drugs used for weight reduction | Serotonin syndrome, tachycardia | ||
|
Itraconazole Fluconazole Ketoconazole Mycoconazole |
Increase in venlafaxine concentration | ||
|
Apixaban, COX-2 inhibitors Clopidogrel Aspirin Warfarin Acenocoumarol |
Increasing the risk of hemorrhagic complications and gastrointestinal bleeding | ||
|
Amoxicillin and clavulanic acid Dihydroergotamine Preparations of common starwort Oxycodone Thistle Palonosetron Pethidine Selegiline Tramadol Triprolidine Trazodone Pentazocine Promethazine |
Onset of serotonin syndrome | ||
| Ginseng | Hyperhidrosis, itching of the skin and also an increased risk of serotonin syndrome | ||
| Mirtazapine | Clonidine | Hypertension | |
| Phenothiazine | Increase of mirtazapine in the blood and subsequent side effects | ||
| Fluoxetine and Rhodiola Rosea | disturbance of consciousness | ||
| Levodopa | Occurrence of a psychotic state | ||
| Combination of sertraline and Echinacea | Serotonin syndrome | ||
|
Ketoconazole Cimetidine Fluvoxamine |
Increase of mirtazapine levels | ||
| Phenytoin Carbamazepine | Reduction of mirtazapine levels in the body | ||
Furthermore, there is a risk of impaired motor skills when this drug is used concomitantly with drugs from the benzodiazepine group [22]. In addition, it can exacerbate the sedative effects of medications with such properties as antipsychotics, alcohol, or benzodiazepines, which can lead to a significant increase in the risk of falls [16]. On the other hand, the CYP2D6 subunit and, to a slightly lower extent, CYP3A4 are involved in venlafaxine metabolism. Drugs in the group in question can also lead to serotonin syndrome when given concurrently with triptans or opiates. Also, venlafaxine used in simultaneous combination with compounds of calcineurin inhibitors, lithium inhibitors, tranylcypromine, or a combination of other SNRIs can lead to acute serotonin syndrome [54, 55].
Concomitant administration of duloxetine with anticoagulants also carries the risk of drug interactions [55]. A change in INR, the international normalized ratio, was observed as an effect of these interactions. There are many cases depicting this interaction. One of them is the case of a woman aged 63. The patient showed a decrease in her INR value when duloxetine 60 mg was added to her already existing acenocoumarol therapy. The value decreased from 2.54 to 1.49, and the index value at 1.49 persisted for three weeks since the duloxetine was discontinued. After this time, the INR value returned to normal [55]. Another case involves a woman aged 44 being treated with warfarin. After the inclusion of 30 mg of duloxetine in the current therapy, there was an increase in the INR value. The value rose even higher when warfarin withdrawal was applied. The index values decreased only when duloxetine exclusion from patient therapy was used [55]. Duloxetine may interact with gastric juice antacids, and the effect of this interaction may be the occurrence of diarrhea [56]. The literature also indicates the possibility of interaction between duloxetine and drugs metabolized by the cytochrome CYP2D6 subunit. Studies have shown that the simultaneous administration of desipramine and duloxetine leads to an increase in the Area under the curve (AUC) and maximum concentration (Cmax) values of desipramine. Because of the possibility of this phenomenon, particular caution, careful monitoring, and overall dose reduction of drugs such as tricyclic antidepressants or antiarrhythmic drugs (propafenone or flecainide) are recommended when used together with duloxetine [57]. Clinical cases also describe interactions between duloxetine and agomelatine administered concurrently. In a 62 year-old female patient treated with duloxetine hydrochloride, a significant increase in sweating was observed when agomelatine was added to her current therapy.
The occurrence of excessive drug-induced sweating was noted after treatment with agomelatine 25 mg and duloxetine hydrochloride 90 mg. The symptoms disappeared when agomelatine was discontinued, and trazodone was started [57]. Cases of drug-induced delirium have been reported with the combination of duloxetine and bupropion [58]. An 85 year-old man was treated with duloxetine at a dose of 60 mg/24 h. After seven days, bupropion was added to the therapy. It was observed that the patient was disoriented and could not recognize his place or the people with him. Additionally, there were visual and auditory delusions and persecution. These hallucinations occurred three days after bupropion was added to the therapy. It was decided to discontinue bupropion administration. After three days of discontinuing this drug from the patient’s treatment, the patient’s delusions and hallucinations gradually began to subside [58]. In addition, there is a risk of interaction when duloxetine is taken concomitantly with apixaban (increased risk of hemorrhagic complication syndrome, lower or upper gastrointestinal bleeding), brompheniramine (risk of dangerous serotonin syndrome), bupropion (cases of hallucinations and delirium), dihydroergotamine (risk of serotonin syndrome), encainide (a significant increase in the concentration of the drug in the body and subsequent side effects as a result of CYP 1A2 inhibition by duloxetine), ciprofloxacin (an enormously increased risk of increasing the concentration of duloxetine in the body—an absolute contraindication for combining the two drugs in pharmacotherapy), enoxacin (an increase in the concentration of duloxetine in the body based on CYP 1A2 inhibition) [33]. Furthermore, a marked increase in the risk syndrome of hemorrhagic complications and gastrointestinal bleeding was noted when duloxetine was co-administered with drugs such as corticosteroids, cyclooxygenase two inhibitors, aspirin, nonsteroidal anti-inflammatory drugs, rivaroxaban, ticlopidine, and warfarin [33].
The literature also indicates an increased possibility of stalling when venlafaxine is used concomitantly with drugs such as ibuprofen, aspirin, and naproxen [59, 60]. Cases of hallucinations and visual hallucinations can also occur when venlafaxine and propafenone are taken concurrently. Such symptoms were observed in a woman aged 85. The patient had been treated with propafenone 150 mg/12 h for three years. As a result of the occurrence of mood disorders, venlafaxine was included in the therapy at that time, initially at a dose of 75 mg/12 h. After one month, the dose of venlafaxine was raised to 150 mg/12 h. Ten days after increasing the dose of venlafaxine, the patient experienced significant psychomotor stimulation, hallucinations, and visual hallucinations. These symptoms worsened particularly at night. After four days of discontinuing venlafaxine, the unpleasant hallucinations ceased utterly [61]. In addition to the mentioned interactions, venlafaxine, when taken concurrently with cimetidine, can lead to the development of liver disease or hypertension. The literature also reports cases in which drugs such as clarithromycin, ketoconazole, or ritonavir that are co-administered with venlafaxine lead to an increase in venlafaxine concentrations in the body as a result of blocking its degradation. Also, the concomitant use of haloperidol with venlafaxine may lead to an interaction manifested as QT interval prolongation. It is also worth noting the risk of reducing the potency of metoprolol when taken together with venlafaxine [61]. There is also a risk of increased drowsiness and sedation when zolpidem or diphenhydramine is used concomitantly with venlafaxine [60]. The occurrence of interactions between venlafaxine and benzodiazepines is not entirely clear. Some sources mention the risk of increased sedation and tranquillity [60]. However, studies conducted with diazepam and venlafaxine show no significant pharmacodynamic or pharmacokinetic interactions [62]. J. Woroń et al. also report the possibility of interactions between venlafaxine and apixaban, COX-2 inhibitors, clopidogrel, aspirin, warfarin and acenocoumarol (increasing the risk of hemorrhagic complications and gastrointestinal bleeding), the combination of amoxicillin and clavulanic acid, dihydroergotamine, preparations of common starwort, oxycodone, thistle, palonosetron, pethidine, selegiline, tramadol, triprolidine, trazodone, pentazocine, and promethazine (onset of serotonin syndrome). In addition, combining therapy with ginseng can result in hyperhidrosis, itching of the skin, and also an increased risk of serotonin syndrome [63].
Clinical cases report the possible occurrence of significant hypertension with concomitant use of mirtazapine and clonidine. This effect occurred in a man in his 20 s who was treated with metoprolol, clonidine, and minoxidil. As a result of depression treatment, mirtazapine at a dose of 15 mg/24 h was included in his therapy. It has been noted that since taking mirtazapine treatment, the patient’s blood pressure began to rise rapidly to the point where it was no longer controlled as much as possible. After the patient was admitted to the intensive care unit and his blood pressure was regulated with nitroprusside, it was decided to discontinue mirtazapine treatment [64]. In addition, the occurrence of a possible interaction between benzodiazepines or alcohol and mirtazapine administered concurrently has been observed. Symptoms of such a combination include increased sedation and tranquilization [65]. No significant pharmacokinetic interactions were noted when mirtazapine was administered concurrently with lithium compounds, cimetidine, paroxetine, carbamazepine, amitriptyline, risperidone, and fluoxetine [65].
Additionally, there are single cases of the occurrence of interactions between mirtazapine and the combination of escitalopram and ginseng (the occurrence of serotonin syndrome), phenothiazine (increase of mirtazapine in the blood and subsequent side effects), combination with fluoxetine and Rhodiola Rosea (disturbance of consciousness), clonidine (mutual antagonization of the pharmacological profile of the drugs and subsequent risk of hypertensive breakthrough), combination therapy with clorazepate and ginseng (delirium along with excessive sedation), and levodopa (occurrence of a psychotic state) [63]. There are also cases of serotonin syndrome when mirtazapine therapy is combined with sertraline and Echinacea [63]. The study did not demonstrate a significant impact of food intake on metabolism and interactions with SNRIs [66]. A study suggests a possible increase in symptoms such as drowsiness, dizziness, motor coordination problems, impaired thinking, concentration problems, or confusion when taking duloxetine concurrently with valerian [67]. Another study suggests the risk of a pharmacokinetic interaction when venlafaxine is taken simultaneously with the Zuojin pill, which contains plant-derived substances such as Fructus Evodiae and Rhizoma Coptidis [68]. Other available scientific sources provide information on the possibility of hemorrhages and gastrointestinal bleeding during simultaneous intake of duloxetine with Ginkgo Bilobapreparations, incidents of anxiety and migraines, and headaches when taken with preparations containing thistle, incidents of intractable sore throat, jaundice, swallowing disorders, diarrhea, and hepatotoxicity when taken concurrently with Ginkgo preparations, as well as incidents of hyperhidrosis, pruritus, and risk of serotonin syndrome when combining duloxetine with ginseng [63].
Interactions of norepinephrine reuptake inhibitors (NRIs)
Reboxetine belongs to a group of norepinephrine reuptake inhibitors used to treat depression [69]. It increases the concentration of norepinephrine in the synaptic gap by binding the NAT ant, which also blocks the extracellular reuptake of norepinephrine relative to the terminals [69]. The study showed that the use of reboxetine in the treatment of depression led to a reduction in the effects of chronic social stress on the neurotrophic brain-derived factor BDFN, the typical expression of the peroxisome proliferator-activated receptor PPAR-α, and the phosphorylated protein responsible for binding the cAMP-mediated response pCREB. With this activity, reboxetine has been proven to exhibit an additional antidepressant mechanism by promoting PPAR alpha expression in hippocampal structures [70]. The drug is mainly metabolized by the cytochrome CYP3A4 subunit [71]. Drugs that are inhibitors or inducers of this isoenzyme affect the metabolism of the drug in question (Table 7) [71]. Inhibitors of the CYP3A4 subunit can be divided into compounds with reversible and irreversible mechanisms. This type of inhibition can occur through several mechanisms—acting on the active site and covalently attaching an amino acid-derived residue at that site or through a mechanism based on alkylation or arylation of a heme-derived prosthetic group. The group of inhibitors also includes substances of plant origin [72]. The group of inducers of this subunit is much smaller [72].
Table 7.
Interactions of NRIs with other medications drug classes and plant-derived agents
| Interactions of NRIs with other drug classes | |||
|---|---|---|---|
| Drug | Interacting substance | Results of interaction | References |
| Reboxetine |
Enzyme CYP3A4 inhibitors: Troleandomycin Roxithromycin Oleandomycin N-desmethylerythromycin Irinotecan Erythromycin Clarithromycin Azithromycin 14-OH- clarithromycin Amiodarone Amlodipine Atazanavir Buprenorphine Clozapine Desipramine Diltiazem Diclofenac Felodipine Indinavir Isoniazid Levonorgestrel Lidocaine Midazolam Nicardipine Nortriptyline Phenelzine Ritonavir Sertraline Selegiline Tadaliphil Thiotepa, and verapamil Rutacarpin and limonin from Evodie fruit Gomisin from Schisandra Chinesis Bergamottin and its derivatives from the juice of Grapefruit Propranolone Dipyridamole Methadone and phenothiazine Enzyme CYP3A4 inductors: Rifabutin Rifampicin Carbamazepine Primidone Phenytoin Phenobarbital Clotrimazole Statins Cyclophosphamide Paclitaxel Ifosfamide |
Increased plasma reboxetine concentrations, increased risk of bupropion side effects | [33, 71–75] |
| Decrease in plasma reboxetine concentrations, increased risk of bupropion side effects | |||
Studies confirm the inhibitory effect of ketoconazole on reboxetine metabolism. After five days of simultaneous intake of reboxetine 4 mg and ketoconazole 200 mg, studies showed an increase in the AUC values for both enantiomers of reboxetine and a decrease in the clearance values for both compounds. An increase in the half-life of ketoconazole was also observed [73]. There are also cases of side effects related to the potentiation of noradrenergic effects with the simultaneous use of reboxetine with the synthetic hormone triiodothyronine T3. Studies have been conducted to determine the impact of administering the synthetic hormone as a potential adjunctive therapy for people struggling with advanced stages of depression. Side effects of this drug combination were observed in patients with symptoms such as irritability, increased body sweating, adverse urinary symptoms, sleep disturbances, tremors, and anxiety [22, 74]. Concomitant use of NRIs with reboxetine can cause the occurrence of sleep problems and insomnia. This effect may occur due to blocking the structure transporter for norepinephrine [75]. There is also a possibility of drug interactions occurring when reboxetine is taken simultaneously with amiodarone, propranolone, dipyridamole, methadone, and phenothiazine (displacement of both drugs from the junctions and subsequent increase in the free fraction of the drug in the patient’s body). There are also reports of a significant increase in the risk of hypokalemia during simultaneous pharmacotherapy with reboxetine and loop and thiazide diuretics [33]. As a result of the in vivo study proved that rats receiving a mixture of reboxetine—curcumin-containing extract showed significantly lower immobility values for chronic fatigue and forced swimming tests. In addition, animals in the groups receiving the mixture had significantly elevated levels of norepinephrine in brain structures. This study demonstrated the synergistic effect of the reboxetine-turmeric extract combination [76].
Interactions of norepinephrine and dopamine reuptake inhibitors (NDRI)
Bupropion belongs to the group of selective norepinephrine and dopamine reuptake inhibitors [77]. The mechanism of action of this drug is based on blocking the reuptake of neurotransmitters such as dopamine and norepinephrine [22]. It blocks dopamine reuptake much more strongly compared to norepinephrine [78]. The drug also acts on nicotinic acetylcholine receptors, their non-competitive antagonist [79]. In addition, bupropion shows a weak affinity for receptors for serotonin [78]. It can reduce inflammatory cytokines like interferon-gamma or Tumour Necrosis Factor-alpha (TNF-alpha) [79].
The cytochrome CYP2B6 subunit mainly metabolizes the drug. It converts bupropion to hydroxybupropion with pharmacological activity [80]. Drugs that are inhibitors or inducers of this cytochrome subunit affect the metabolism of bupropion (Table 8) [80]. The inhibitor group of drugs causes a decrease in the enzymatic activity of the CYP2B6 subunit [80]. A reduction of 90% was reported with ticlopidine and 68% with clopidogrel [80].
Table 8.
Interactions of NDRI with other medications drug classes
| Interactions of NDRI with other drug classes | |||
|---|---|---|---|
| Drug | Interacting substance | Results of interaction | References |
| Bupropion |
Enzyme CYP2B6 inhibitors: Clopidogrel Ticlopidine Sertraline and thiotepa Orphenadrine Cyclophosphamide Paroxetine Clonazepam Fluvoxamine Norfluoxetine Nelfinavir, Diazepam and ritonavir Ketoconazole itraconazole |
Increased plasma bupropion concentrations, increased risk of bupropion side effects | [22, 33, 80–83] |
|
Enzyme CYP2B6 inductors: Rifampicin Phenobarbital Corticosteroids and dexamethasone Carbamazepine Hyperforin Ritonavir Clotrimazole Efavirenz |
Decreased plasma bupropion concentrations, impairment of the effects of bupropion | ||
| Flecainide | Elevation of flecainide concentration in the body entailing adverse side effects | ||
| Hormone replacement therapy | Significant elevation of bupropion concentration in the body and increased risk of seizures | ||
| Insulin | In case of hypoglycemia increased risk of convulsions | ||
| Linezolid | Risk of severe transient intraoperative pressure and dangerous risk of neurotoxicity | ||
| Moclobemide | Severe risk of neurotoxicity and bupropion intoxication | ||
| Metoprolol | Bradycardia, hypotension, severe sinus bradycardia, dizziness | ||
On the other hand, drugs that have inductors-properties can lead to increased metabolism and elevated levels of hydroxybupropion in the body [80]. Due to its ability to inhibit the cytochrome CYP2D6 subunit, bupropion is contraindicated for concomitant use with antipsychotics such as haloperidol, and other antidepressants such as sertraline, desipramine, group 1C antiarrhythmic drugs such as propafenone, flecainide and beta blockers like metoprolol [22]. The literature indicates an increased possibility of a seizure when bupropion is taken concomitantly with drugs that are characterized by their ability to lower the seizure threshold like tricyclic antidepressants and phenothiazines [22], tramadol, other drugs used to treat depression, drugs used for psychotic conditions, and theophylline [81]. Also, concomitant use with alcohol, lithium, glucocorticosteroids, olanzapine, and clozapine may lead to an increase in explosive effects on an additive basis by lowering the seizure threshold [33]. Bupropion should also be used with caution in combination with dopaminergic drugs due to its ability to enhance dopaminergic activity. Observations showed that the concomitant use of bupropion with levodopa used to treat Parkinson's disease increased patients' risk of nausea, hallucinations and delusions, confusion, agitation, and vomiting [22]. The simultaneous intake of levodopa-bupropion drug combinations can lead to a significant increase in the severity and frequency of levodopa side effects [82]. In addition, available scientific sources report that concomitant therapy with amantadine carries a high risk of patient neurotoxicity, and with memantine may exacerbate the risk of a seizure episode through a mechanism of cytochrome CYP 2B6 inhibition [33].
Monoamine oxidase inhibitors and bupropion used concurrently may pose a risk of interaction. This risk arises from the dual mechanism of action involving blocking monoamine reuptake and inhibiting their normal metabolism. Consequently, there's a potential for dangerous elevation of norepinephrine, serotonin, and dopamine levels in the body. To mitigate this risk, it is recommended to observe a minimum 14 day interval between discontinuing bupropion and initiating monoamine oxidase inhibitor therapy [81]. This break allows for restoring regular monoamine oxidase activity [81]. The case of a 57 year-old man presents the risk of combining bupropion with drugs that are mild reversible monoamine oxidase inhibitors with weak potency. As a result of a bypass graft infection, linezolid was included in the man's then-current therapy containing bupropion. After 24 h, the man developed severe hypertension with a blood pressure of 260/145 mmHg. Episodes of hypertension developed in the man due to the combination of bupropion and linezolid. In addition to its antimicrobial activity, linezolid is characterized by its ability to reversibly inhibit the enzyme low-potency monoxidase, which, combined with bupropion, manifested itself in episodes of hypertension [83]. For this reason, drugs belonging to the group of traditional monoamine oxidase inhibitors like tranylcypromine or phenelzine with bupropion have long been discontinued [83].
In addition to the cases described above, available scientific sources report the possibility of drug interactions when bupropion is taken concomitantly with efavirenz (the risk of lowering the concentration of bupropion in the body and lack of pharmacological effect preceding induction of CYP 2B6), flecainide (elevation of flecainide concentration in the body entailing adverse side effects, due to inhibition of cytochrome CYP 2D6), hormone replacement therapy (significant elevation of bupropion concentration in the body and increased risk of seizures), insulin (in case of hypoglycemia increased risk of convulsions), antifungal drugs such as ketconazole or itraconazole (induction of CYP 2B6 and increase of bupropion concentration in the body with risk of dangerous side effects), linezolid (concomitant use promotes risk of severe transient intraoperative pressure and dangerous risk of neurotoxicity), and moclobemide (severe risk of neurotoxicity and bupropion intoxication) [33]. Moreover, simultaneous intake with metoprolol can lead to cases of bradycardia, hypotension, severe sinus bradycardia, and also dizziness [33].
Also, substances of plant origin, such as baicalein, may affect the proper metabolism of bupropion. Through inhibition of the cytochrome CYP2D6 subunit, hydroxylation of bupropion was blocked. The study's results were analyzed by measuring the AUC field [84]. Also, plant extracts derived from ginkgo biloba may interfere with the normal metabolism of the drug in question. A study using recombinant liver-derived microsomal enzymes proved that extracts derived from this plant lead to blocking the hydroxylation of the antidepressant in question, which is related to the ability of Ginkgo-derived substances to inhibit CYP2D6 enzymes [85]. However, this effect needs to be clarified. There is another study conducted on men aged 19–25 who were given bupropion alone at a dose of 150 mg and in combination with a ginkgo preparation at a dose of 240 mg per day. The study was conducted for 14 days. Blood samples were collected from the subjects and evaluated for parameters of bupropion and its hydroxylated metabolite. A blood analysis showed that administration of the ginkgo preparation for 14 days did not significantly affect the pharmacokinetic parameters of hydroxybupropion or bupropion [86]. The literature also reports that substances from St. John’s wort herb can also interfere with the normal metabolism of bupropion [82].
MAOIs interactions
Monoamine oxidase inhibitors (MAOIs) are medicines used to treat various forms of depression [87]. MAOIs affect by inhibiting the breakdown of these neurotransmitters, thereby enhancing their levels and allowing them to continue to act on depressed cells [88]. There are two kinds of monoamine oxidase: A and B. The monoamine oxidase A (MAO-A) is mainly spread in the liver, gut, and placenta, while monoamine oxidase A (MAO-B) occurs in the brain, platelets, and liver. Noradrenaline and serotonin are substrates of MAO-A, and methylhistamine, tryptamine, and phenylethylamine are substrates of MAO-B. Tyramine and dopamine are metabolized by MAO-A and -B [89]. MAOIs include moclobemide, phenelzine, isocarboxazid, selegiline, and tranylcypromine [90]. Due to side effects, several dietary restrictions, and safety concerns, MAOIs are not the drug of first choice for the treatment of mental health disorders. MAOIs can potentially cause many interactions: drug-food interaction, drug-to-drug interactions, and overdoses [91]. MAOIs should not be used with other antidepressants, such as SSRIs. A combination of these drugs can cause serotonin syndrome, which can be potentially fatal. What is more, when changing MAOIs to different antidepressants, patients should wait 14 days before starting the new treatment to prevent any drug interaction. MAOIs avert the breakdown of tyramine in the body and some foods, drinks, and other medications. When taking MAOIs and eating tyramine-containing foods, patients show high serum tyramine levels, leading to a sudden rise in blood pressure, known as the tyramine pressure response [92, 93]. Although it is rare, it can cause a cerebral hemorrhage, resulting even in death. [94]. Meperidine, tramadol, methadone, and dextromethorphan are contraindicated in patients taking MAOIs because they are at high risk of serotonin syndrome [90]. Taking moclobemide with omeprazole may result in a doubling of omeprazole levels and an increased risk of side effects [33]. Overall, SSRIs, TCAs, SNRIs, Serotonin antagonists and reuptake inhibitors (SARI), mirtazapine, bupropion, and sympathomimetic amines, including stimulants and St. John's Wort, are contraindicated with MAOIs [90].
Interactions of drugs with receptor-based mechanisms of action
SARI works by inhibiting serotonin transporter and serotonin type 2 receptors. SARI blocks the alpha-1-adrenergic and histamine receptors and inhibits serotonin reuptake. SARI significantly induces a change in 5-HT presynaptic adrenoreceptors. However, the entire mechanism of action is not fully understood [94]. SARIs are inhibitors of the hepatic isoenzyme CYP3A4; therefore, when combined with drugs metabolized by this enzyme may increase the concentration of these drugs in the blood, increasing the risk of cardiotoxicity (Table 9) [18, 19, 23]. Administration of SARI with anxiolytic drugs, alprazolam, triazolam, and buspirone may improve their concentration in the blood [95]. The use of SARIs in combination with benzodiazepines is safe; however, increased soothing and sedative effects should be taken into account. Taking SARI drugs simultaneously with substances depressing the CNS, e.g., alcohol, can significantly intensify their effects [95].
Table 9.
Interaction of drugs with receptor-based mechanisms of action drugs with other medications drug classes and stimulants
| Interaction of drugs with receptor-based mechanisms of action with drugs | |||
|---|---|---|---|
| Drug | Interacting substance | Results of interaction | References |
| Mianserin | Alcohol | Increase in sedative effects and hepatopathy | [18, 19, 23, 33, 95–99] |
| Atomoxetine | Increase in seizure risk | ||
|
Phenothiazine Clozapine Quetiapine Olanzapine TCAs |
Summation of risk: sedation, myelotoxicity, hepatotoxicity | ||
| Carbamazepine | Increase in haematological and hepatological complications, attenuation of the effect of mianserin | ||
| NSAIDs | Possible decrease in analgesic effect due to central anticholinergic effect of mianserin | ||
| SARI |
Terfenadine Astemizole Pimozide |
Risk of cardiotoxicity | |
|
Alprazolam Triazolam Digoxin Diphenylhydantoin |
Increase concentration in the blood | ||
| Trazodone |
Buspirone Dextromethorphan Diphenhydramine Linezolid Pethidine Setrones Tramadol Tiprolidine Triptans |
Serotonin syndrome | |
| Omeprazole | Mobitz type 1 heart block | ||
What is more, taking other serotonergic medicines such as fentanyl, triptans, and TCA at the same time also increases serotonin levels [96]. Potential pharmacokinetic interaction of SARI drugs with digoxin and diphenylhydantoin has been identified, increasing serum concentrations of both drugs [97]. Nefazodone should not be used with TCAs because it increases their concentration in the blood [95]. Trazodone may also potentiate the effects of skeletal muscle relaxants and volatile anesthetics [98]. Trazodone also has a potential interaction with omeprazole. Trazodone may cause serotonin syndrome if taken together with buspirone, dextromethorphan, diphenhydramine, linezolid, pethidine, tramadol, triprolidine, and triptans [33].
Mianserin is referred to as an “atypical” antidepressant due to its unusual modes of action [99]. Mianserin acts as an antagonist of an α2-noradrenergic receptor. Moreover, it significantly exhibits antagonist properties at serotonin 5HT2A, 5HT2C, and 5HT3 receptors. It has also been noticed that mianserin increases dopamine and noradrenaline release in cortico-limbic areas of the brain [99]. The antidepressant effect of mianserin results from the blockade of presynaptic, hetero-, and auto-α2-adrenoceptors [100]. Interaction information is mainly derived from animal studies. It has been shown that caffeine and sildenafil increase the effect of MSR in the tail suspension test [101] and forced swim test [99]. This interaction is likely not pharmacokinetic. Studies on rats have shown that despite a moderate increase in blood pressure, mianserin does not affect the hypotensive effect of enalapril and propranolol and significantly increases the impact of prazosin [102].
There is also some indication that mianserin may intensify the antinociceptive effect of indomethacin and metamizole (but not morphine) in mice [103]. In addition, scientific sources report possible interactions of mianserin with alcohol (increase in sedative effects and hepatopathy), atomoxetine (increase in seizure risk), phenothiazine, clozapine, quetiapine, olanzapine, TCAs (summation of risk: sedation, myelotoxicity, hepatotoxicity), carbamazepine (increase in hematological and hepatological complications, attenuation of the effect of mianserin), NSAIDs (possible decrease in analgesic effect due to central anticholinergic effect of mianserin) [33].
Interactions of drugs with other mechanisms of action
Agomelatine (AGT) is an agonist of the MT1 and MT2 melatonin receptors and an antagonist of 5-HT2C receptors. The psychotropic effect of agomelatine is due to the combination of its 5-hydroxytryptaminergic and melatonergic impact [104]. Agomelatine can modulate circadian rhythms. This property may be therapeutically crucial for an antidepressant, as depression is associated with circadian rhythm disruption [105]. The antidepressant effect of agomelatine has been proven in studies on depressed rodents [106]. The biotransformation is mainly mediated by CYP1A2 and, to a minor extent, by CYP2C9/19 [107]. Drugs that interact with CYP1A2 can significantly change the plasma levels of AGT (Table 10) [108]. Fluvoxamine, a strong CYP1A2 and moderate CYP2C9 inhibitor, substantially inhibits the metabolism of AGT, resulting in a sixfold increase in agomelatine exposure. A combination of estrogens with AGT (temperate CYP1A2 inhibitors) causes a several-fold increased exposure to AGT, though in experiments, this did not cause clinically relevant effects [82]. It has been shown that smoking induces CYP1A2 and decreases the bioavailability of AGT [82]. The use of AGT together with alcohol adds up to potential hepatotoxicity. During the coadministration of AGT with duloxetine, a case of akathisia [22, 82] and hyperhidrosis [82] have been reported. Both were classified as pharmacodynamic DDI with noradrenergic hyperstimulation. In the mouse depression model, caffeine strengthened the effects of agomelatine without changing its plasma levels [101]. In other studies on mice, hesperidin has been affirmed to potentiate the antidepressant effect of agomelatine [109].
Table 10.
Interactions of drugs with other mechanisms of action with medications and plant-based agents
| Interactions of drugs with other mechanisms of action | References | ||
|---|---|---|---|
| Drug | Interacting substance | Results of interaction | |
| Agomelatine |
Enzyme CYP1A2 inhibitors: Fluvoxamine Amiodarone Ciprofloxacin Disulfiram Enoxacin Erythromycin Ketoconazole Clarithromycin Mexiletine Moclobemide Norfloxacin Ticlopidine |
Significant increase in agomelatine concentration, potentiate its side effects | |
|
Chlorpromazine Phenothiazines Carbamazepine Clozapine Milk thistle + sertraline Paracetamol Statins TCAs Alcohol |
Potential hepatotoxicity | ||
|
Phenobarbital Phenytoin Rifampicin Ritonavir Tipranavir |
Reduction in agomelatine blood concentration | ||
| Duloxetine | Akathisia and hyperhidrosis | ||
| Tianeptine | Salicylic acid | Increase in tianeptine concentration, potentiate its side effects | |
| Vilazodone | Ketoconazole | Increase in vilazodone concentration, potentiate its side effects | |
| Vortioxetine |
Enzyme CYP2D6 inhibitors: Bupropion Fluconazole Ketoconazole Terbinafine Warfarin Hydrochlorothiazide Buspirone Dextromethorphan Diphenhydramine MAO inhibitors Linezolid Moclobemide Pethidine Antiemetic setrones SNRIs SSRIs TLDs Tramadol Trazodone at doses > 150 mg Triprolidine Triptans Phenytoin carbamazepine Rifampicin Piracetam |
Increase vortioxetine concentrations and potentiate its side effects | |
| Hemorrhagic complications | |||
| Hyponatremia | |||
| Increased risk of serotonin syndrome | |||
| Decrease vortioxetine concentrations and reduce its activity | |||
| Decreased elimination of piracetam and its increased concentration in the blood | |||
Tianeptine (TNP) is an antidepressant drug with a unique neurochemical profile. It increases serotonin uptake in the brain and reduces stress-induced atrophy of neuronal dendrites [110]. TNP is an agonist of the δ-opioid receptor (DOR) and the μ-opioid receptor (MOR). It is believed that MOR-agonism (or combined DOR/MOR-agonism) underlies the preclinical, clinical, and in vitro effects of TNP [111]. TNP binds strongly to albumin, which could theoretically lead to interactions with other drugs that bind to proteins, such as nonsteroidal anti-inflammatory drugs and sulfonylureas. However, due to the wide therapeutic margin, increased TNP concentrations due to the concomitant administration of these drugs are unlikely to be dangerous. TNP varies from most antidepressant medications because it is not mainly metabolized by the hepatic cytochrome P450, which indicates a lower probability of drug-drug interactions, which is especially important in elderly patients [110]. Alcohol administration decreased the TNP absorption rate and reduced plasma levels by around 30%; however, it did not influence the pharmacokinetics of the C5 metabolite. In studies evaluating the effect of antipsychotics, benzodiazepines, and many other drugs on the binding of TNP to plasma proteins, it was found that only salicylic acid in high plasma concentrations can displace TNP from its binding sites, which may increase its effect [112]. TNP significantly decreased the "wet dog shakes" induced by 5-hyroxytrytophan, which could reduce the probability of serotonin syndrome if tianeptine is inadvertently used with drugs that can cause serotonin syndrome [112]. In a mouse model of epilepsy, TNP increased the anticonvulsant effects of carbamazepine and valproate, but not phenytoin, without changing the concentration of these drugs in the brain. However, TNP reduced phenobarbital concentration in the brain but enhanced its anticonvulsant activity [113]. In mice, the activity of TNP was enhanced, presumably by increasing its brain concentration [114].
Vilazodone exhibits a dual mechanism of action. It acts as a potent and selective inhibitor of serotonin 5-HT reuptake and selectively binds to 5-HT1A receptors with high affinity [115]. Functioning as both an SRI and a 5-HT1A partial agonist, vilazodone undergoes metabolism primarily via CYP3A4, with additional metabolism by CYP2C19 and CYP2D6, as well as possibly through non-CYP pathways such as carboxylesterases [116]. Simultaneous administration of vilazodone with a proton pump inhibitor or ethanol does not extend or affect the rate of vilazodone absorption. Consequently, when combined with potent CYP3A4 inhibitors, vilazodone dosage should be reduced to 20 mg. No dose adjustment is recommended for mild CYP3A4 inhibitors. CYP3A4 inducers are expected to minimize vilazodone concentration, which may decrease the effectiveness of the drug.
Minimal effects on vilazodone have been noted with other CYP isoenzyme inhibitors and inducers. Research in healthy volunteers revealed that vilazodone at a dosage of 20 mg/day administered for 8–10 days with flurbiprofen, nifedipine, caffeine, or debrisoquine, probes for CYP2C9, CYP3A4, CYP1A2, and CYP2D6, respectively—did not significantly affect their pharmacokinetic profiles. Additionally, in vitro experiments suggest that vilazodone may inhibit biotransformation of CYP2C8 substrates. Simultaneous administration of vilazodone with any drug that affects serotonergic neurotransmitter systems ought to be carefully applied, and it needs clinical monitoring because of the potential for serotonin syndrome [117]. Vortioxetine is a novel antidepressant for the treatment of major depression [118]. This drug is metabolized by many enzymes [119]. CYP2D6 is an enzyme that mainly metabolizes vortioxetine; CYP2C19 plays a minor role in this process. There are known interactions between vortioxetine and other drugs. Active metabolites of bupropion-threohydrobupropion and erythrohydrobupropion- are potent inhibitors of CYP2D6. Occurrences of side effects, such as nausea, vomiting, headache, and insomnia, were three times more likely after adding bupropion to vortioxetine monotherapy as when vortioxetine was added to bupropion therapy or when vortioxetine was used without other drugs [119]. Lots of CYP enzymes are induced by rifampicin, for example, CYP3A4/5, CYP2C19, CYP2C9, CYP2C8, and CYP2B6. Because of this, it is recommended to take into consideration increasing the dose of vortioxetine during treatment with rifampicin or other potent CYP inducer, for example, phenytoin or carbamazepine, when they are administered more than 14 days [119].
In the study of Woroń et al., there are described two cases of hemorrhagic complications—nasal and vaginal bleeding-after a combination of vortioxetine and warfarin. Another case concerns hyponatremia, which occurred as a result of a combination of vortioxetine and hydrochlorothiazide [3]. Available evidence indicates that vortioxetine does not significantly affect the pharmacokinetics of warfarin or increase its concentration [119]. The observed bleeding events may be linked to vortioxetine's mechanism of action, which involves blocking serotonin reuptake on platelets, similar to SSRIs and SNRIs [120]. It is presumed that the mechanism underlying hyponatremia may parallel that of SSRIs, involving the risk of hyponatremia due to diuretic action and the potential induction of syndrome of inappropriate antidiuretic hormone secretion associated with serotonin reuptake blockade [3].
New drug interactions – ifenprodil
Ifenprodil is a new drug from the anti-n-methyl-d-aspartate receptor (NMDAR) receptor antagonist group that has found use as an innovative antidepressant in recent years [121]. The mechanism of action of this drug is based on blocking G-protein-stimulated potassium channels [122]. Additionally, it blocks the previously mentioned n-methyl-d-aspartate receptor (NMDA) receptors through a conservative pathway by affecting GluN subunits [121]. The study, conducted with a mouse model of anxiety disorders, showed a change in the behavioral outcomes of the test animals in the track of simultaneous administration of ifenprodil together with dizocilpine. During the conducted experiment, the researchers noted a significant ability to suppress the deficit caused by the administration of dizocilpine, whereas, during the isolated administration of ifenprodil, the described effect did not occur. Both drugs were administered at a dose of 10 ml/kg (0.25 ml of the drug) via the intraperitoneal route. Mice evaluated during the maze plus test showed the effect of ifenprodil combined with dizocilpine—the addition of ifenprodil to the therapy induced a decrease in anti-anxiety effects and a decrease in the overall locomotor activity of dizocilpine. The experiment also noted the absence of degenerative effects of ifenprodil on working memory, with the simultaneous presence of such effects for dizocilpine [122, 123].
In another study involving mice, a synergistic antidepressant-like effect was observed with the combination of lithium and ifenprodil treatment. The test substances were administered to animals at a dose of 5 ml/kg (lithium) and 0.1/0.5/3 mg/kg (ifenprodil) of mouse body weight via the intraperitoneal route. The injections were performed 30 (lithium) and 45 (ifenprodil) minutes before the selected test. The combination of both drugs resulted in a significant enhancement of the antidepressant effect compared to the control group, as assessed by the forced swim test [124]. Single available publications also present the enhancing effect of ifenprodil when combined in pharmacotherapy together with paroxetine. In an experiment using a mouse model of depression, they showed a beneficial effect of the paroxetine-ifenprodil combination, characterized by an enhanced antidepressant effect [125].
Interactions of drugs with adaptogens
Recently, many patients suffering from depression and mood disorders often associated with the disease have been turning to so-called “adaptogens” Adaptogens are non-toxic substances of plant origin that increase “non-specific” resistance to adverse biological, chemical, and physical factors, normalizing body functions and enhancing stress resistance [126]. Because the principle of adaptogenic action has yet to be precisely explained, this term is currently not accepted in clinical and pharmacological terminology in the European Union; however, it is now widely used, sometimes even by doctors and pharmacists. However, there are scientific reports that taking adaptogens is associated with a better ability of the body to adapt to stress and normalize metabolic functions, as well as with better mental and physical performance [127–129]. Many reports suggest that adaptogens may improve sleep, alleviate fatigue, anxiety, memory impairment, and depressive symptoms, and reduce stress levels [130]. Therefore, in recent years, there has been a steady increase in interest in adaptogens among patients suffering from depressive disorders, who take them in addition to medications prescribed by doctors as a form of complementary treatment or to alleviate side effects occurring during psychopharmacotherapy [131]. As a rule, these substances are non-toxic and usually well tolerated, but they may cause adverse interactions with concurrently taken antidepressants.
Interestingly, research conducted by Woroń et al. showed that the frequency of interactions of “adaptogens” with antidepressants was twice as high as the occurrence of adverse events caused by interactions of antidepressants with over-the-counter drugs [132]. As the authors of the study note, in clinical practice, the incidence of these interactions may be much higher. It is worth emphasizing that dietary supplements referred to as adaptogens most often contain plant extracts consisting of many separate, pharmacologically active substances, which additionally significantly increases the risk of adverse events [63]. Woroń et al. performed a retrospective review assessing the incidence and clinical characteristics of adverse events associated with the concomitant use of antidepressants and adaptogens [131]. They evaluated 326 reports and found that 9% of adverse events due to antidepressant drug interactions were due to the concomitant use of adaptogens, especially Withania somnifera (Ashwagandha), Eleutherococcus senticosus, Schisandra chinensis, Tribulus terrestris, Coptis chinensis, Cimcifuga racemosa, Bacopa monnieri, Gynostema pentaphyllum, Cordyceps sinensis, Lepidium meyenii, and Scutellaria baicalensis. The undesirable effects of the interaction of adaptogens with antidepressants are presented in Table 11. The hypothesis put forward by the authors of the study may be confirmed by the fact that in all cases analyzed, discontinuation of adaptogen supplements led to remission of the side effects. The main mechanisms responsible for the occurrence of unfavorable phenomena when taking antidepressants and adaptogens simultaneously include interactions with cytochrome p-450 and P-glycoprotein. The main mechanisms responsible for the development of unfavorable phenomena when taking antidepressants and adaptogens simultaneously include interactions with cytochrome p-450 and P-glycoprotein, in particular, inhibition of isoenzymes involved in drug metabolism and inhibition of transport proteins. As a result of these interactions, many side effects may occur, in some cases life-threatening, such as bleeding from the upper gastrointestinal tract or myocardial infarction.
Table 11.
Interactions of drugs with adaptogens
| Interactions of drugs with adaptogens | References | ||
|---|---|---|---|
| Adaptogen | Interacting substance | Result of interaction | |
| Withania somnifera | Reboxetine | Testicle pain and ejaculatory dysfunctions | [63, 131, 132] |
| Sertraline | Severe diarrhea | ||
| Escitalopram | Myalgia, epigastric pain, nausea, vomiting, restless legs syndrome, and severe cough | ||
| Paroxetine | Generalized myalgia, ophthalmalgia, and ocular hypertension | ||
| Eleutherococcus senticosus | Duloxetine | Upper gastrointestinal bleeding | |
| Paroxetine | Epistaxis | ||
| Sertraline | Vaginal hemorrhage | ||
| Agomelatine | Irritability, agitation, headache, and dizziness | ||
| Schisandra chinensis | Bupropion | Arthralgia and thrombocytopenia | |
| Amitriptyline | Delirium | ||
| Fluoxetine | Dysuria | ||
| Tribulus terrestris | Citalopram | Generalized pruritus | |
| Escitalopram | Galactorrhea | ||
| Trazodone | Psoriasis relapse | ||
| Coptis chinensis | Mianserin | Arrhythmias | |
| Mirtazapine | Edema of lower limbs and myalgia | ||
| Fluoxetine | Gynecomastia | ||
| Cimicifuga racemosa | Mianserin | Restless legs syndrome | |
| Paroxetine | Gynecomastia and mastalgia | ||
| Venlafaxine | Hyponatremia | ||
| Bacopa monnieri | Agomelatine | Back pain and hyperhidrosis | |
| Moclobemide | Myocardial infarction | ||
| Gynostemma pentaphyllum | Duloxetine | Back pain | |
| Cordyceps sinensis | Sertraline | Upper gastrointestinal bleeding | |
| Lepidium meyenii | Mianserin | Restless legs syndrome | |
| Scutellaria baicalensis: | Bupropion | Seizures | |
| Japanese ginkgo biloba | SSRI or | Hemorrhagic complications | |
| SNRI | |||
| Panax ginseng | Haloperidol | Ventricular arrhythmias, pancreatitis | |
| Risperidone | Pancreatitis | ||
| Aripiprazole | Hepatotoxicity | ||
Interactions of drugs with CBD
Patients suffering from depression, often on their own, without consulting a doctor, use cannabidiol (CBD), for which scientific data on safety and effectiveness in the treatment of depression are limited and inconclusive. CBD is a natural compound found in cannabis, whose therapeutic effects are currently widely advertised. However, the FDA (Food and Drug Administration) has concluded that “The use of CBD raises various safety concerns, especially with long-term use” [133]. Studies have shown that CBD has potentially harmful effects and the possibility of severe negative drug interactions—the FDA-approved epilepsy drug (Epidiolex) has risks of liver damage, excessive sedation, suicidal thoughts, and hypersensitivity reactions [133]. Studies have also shown potential effects such as chronic psychosis and anxiety. Nowadays, the use of CBD, a phytocannabinoid derived from cannabis that does not have intoxicating properties, is becoming more and more popular. Unfortunately, this compound is not without risk of interaction with other substances, including antidepressants. Available studies show that CBD is metabolized by cytochrome CYP450 isoenzymes, including CYP2D6, CYP2C19, CyP3A4, CYP1A2, and CYP2C9, on which it has an inhibitory effect. This action may increase the concentration of SSRI and TCA drugs. In addition, CBD interacts with MAOIs by inhibiting their metabolism, which may increase side effects [134, 135].
Conclusions and future perspectives
Injudicious prescription of antidepressants to patients suffering from other diseases may cause unfavorable drug interactions, the most severe effects of which include gastrointestinal bleeding, cardiovascular disorders, bradycardia, serotonin syndrome, neuroleptic syndrome, hepatotoxicity, increased risk of seizures, and in some cases – even death. Therefore, pharmacotherapy of patients with depression and comorbidities should be balanced, preceded by a detailed analysis of the safety assessment, followed by the search for the safest alternative drug combinations. There is a need for monitoring and reporting of adverse events resulting from drug interactions, as well as in-depth education in this area. At the same time, medical practitioners should keep in mind the undesirable interactions are occurring in patients who, in the treatment process of other diseases, self-medicate depression with so-called adaptogens. Such cases unveil the significance of a thorough medical interview regarding prescription drugs and more and more commonly used dietary supplements.
Author Contributions
Conceptualization, methodology, project administration: MH, writing—original draft: A.D., E.B., G.B., M.H., writing—review & editing: I.P.Ch., M.H. All authors have read and agreed to the published version of the manuscript.
Funding
This research received no external funding.
Data availability
No datasets were generated or analyzed in the current study.
Declarations
Conflict of interest
The authors declare no conflict of interest.
Footnotes
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- 1.Takano T, Mizuguchi D, Omiya Y, Higuchi M, Nakamura M, Shinohara S, et al. Estimating depressive symptom class from voice. IJERPH. 2023;20:3965. 10.3390/ijerph20053965 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Mazza MG, Palladini M, Poletti S, Benedetti F. Post-COVID-19 depressive symptoms: epidemiology, pathophysiology, and pharmacological treatment. CNS Drugs. 2022;36:681–702. 10.1007/s40263-022-00931-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Woroń J, Siwek M, Gorostowicz A. Adverse effects of interactions between antidepressants and medications used in treatment of cardiovascular disorders. Psychiatr Pol. 2019;53:977–95. 10.12740/PP/OnlineFirst/96286 [DOI] [PubMed] [Google Scholar]
- 4.Rai NK, Goyal V, Kumar N, Shukla G, Srivastava AK, Singh S, et al. Neuropsychiatric co-morbidities in non-demented parkinson’s disease. Ann Indian Acad Neurol. 2015;18:33–8. 10.4103/0972-2327.144287 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Sundar U, Honrao ST, Shah N. Psychiatric co-morbidities in women with epilepsy. J Assoc Physicians India. 2017;65:30–2. [PubMed] [Google Scholar]
- 6.Dokras A, Sarwer DB, Allison KC, Milman L, Kris-Etherton PM, Kunselman AR, et al. Weight loss and lowering androgens predict improvements in health-related quality of life in women with PCOS. J Clin Endocrinol Metab. 2016;101:2966–74. 10.1210/jc.2016-1896 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Penninx BW, Milaneschi Y, Lamers F, Vogelzangs N. Understanding the somatic consequences of depression: biological mechanisms and the role of depression symptom profile. BMC Med. 2013;11:129. 10.1186/1741-7015-11-129 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Triolo F, Belvederi Murri M, Calderón-Larrañaga A, Vetrano DL, Sjöberg L, Fratiglioni L, Dekhtyar S. Bridging late-life depression and chronic somatic diseases: a network analysis. Transl Psychiatr. 2021;11:557. 10.1038/s41398-021-01686-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Leone M, Kuja-Halkola R, Leval A, D’Onofrio BM, Larsson H, Lichtenstein P, Bergen SE. Association of youth depression with subsequent somatic diseases and premature death. JAMA Psychiatr. 2021;78:1–9. 10.1001/jamapsychiatry.2020.3786 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ouazana-Vedrines C, Lesuffleur T, Cuerq A, Fagot-Campagna A, Rachas A, Gastaldi-Menager C, et al. Outcomes associated with antidepressant treatment according to the number of prescriptions and treatment changes: 5 year follow-up of a nation-wide chohort study. Front Psychiatr. 2022;13:923916. 10.3389/fpsyt.2022.923916 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.O’Brien FE, Dinan TG, Griffin BT, Cryan JF. Interactions between antidepressants and P-glycoprotein at the blood-brain barrier: clinical significance of in vitro and in vivo findings. Br J Pharmacol. 2012;165:289–312. 10.1111/j.1476-5381.2011.01557.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Löscher W, Potschka H. Drug resistance in brain diseases and the role of drug efflux transporters. Nat Rev Neurosci. 2005;6:591–602. 10.1038/nrn1728 [DOI] [PubMed] [Google Scholar]
- 13.Furukawa TA, McGuire H, Barbui C. Meta-analysis of effects and side effects of low dosage tricyclic antidepressants in depression: systematic review. BMJ. 2002. 10.1136/bmj.325.7371.991. 10.1136/bmj.325.7371.991 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Schneider J, Patterson M, Jimenez XF. Beyond depression: other uses for tricyclic antidepressants. CCJM. 2019;86:807–14. 10.3949/ccjm.86a.19005 [DOI] [PubMed] [Google Scholar]
- 15.Baldaçara L, Freitas R, Baldaçara R. Tricyclic antidepressants: an update. Res Soc Dev. 2024;13:4413445539. 10.33448/rsd-v13i4.45539 [DOI] [Google Scholar]
- 16.Bleakley S. Antidepressant drug interactions: evidence and clinical significance: antidepressant drug interactions. Prog Neurol Psychiatr. 2016;20:21–7. 10.1002/pnp.429 [DOI] [Google Scholar]
- 17.Buckley NA, Dawson AH, Isbister GK. Serotonin Syndrome. BMJ. 2014. 10.1136/bmj.g1626. 10.1136/bmj.g1626 [DOI] [PubMed] [Google Scholar]
- 18.Hicks JK, Sangkuhl K, Swen JJ, Ellingrod VL, Müller DJ, Shimoda K, et al. Clinical pharmacogenetics implementation consortium guideline (CPIC®) for CYP2D6 and CYP2C19 genotypes and dosing of tricyclic antidepressants: 2016 update. Clin Pharmacol Ther. 2017;102:37–44. 10.1002/cpt.597 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Saraghi M, Golden LR, Hersh EV. Anesthetic considerations for patients on antidepressant therapy—part I. Anesth Prog. 2017;64:253–61. 10.2344/anpr-64-04-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Shin JG, Park JY, Kim MJ, Shon JH, Yoon YR, Cha IJ. Inhibitory effects of tricyclic antidepressants (TCAs) on human cytochrome P450 enzymes in vitro: mechanism of drug interaction between TCAs and phenytoin. Drug Metab Dispos. 2002;30:1102–7. 10.1124/dmd.30.10.1102 [DOI] [PubMed] [Google Scholar]
- 21.Kamińska K, Lenda T, Konieczny J, Wardas J, Lorenc-Koci E. Interactions of the tricyclic antidepressant drug amitriptyline with L-DOPA in the striatum and substantia nigra of unilaterally 6-OHDA-lesioned rats. relevance to motor dysfunction in parkinson’s disease. Neurochem Inter. 2018;121:125–39. 10.1016/j.neuint.2018.10.004 [DOI] [PubMed] [Google Scholar]
- 22.Spina E, Scordo MG. Clinically significant drug interactions with antidepressants in the elderly. Drugs Aging. 2002;19:299–320. 10.2165/00002512-200219040-00004 [DOI] [PubMed] [Google Scholar]
- 23.Fugh-Berman A. Herb-drug interactions. Lancet. 2000;355:134–8. 10.1016/S0140-6736(99)06457-0 [DOI] [PubMed] [Google Scholar]
- 24.Kuhn MA. Herbal remedies: drug-herb interactions. Crit Care Nurse. 2002;22:22–8. 10.4037/ccn2002.22.2.22 [DOI] [PubMed] [Google Scholar]
- 25.Ismail MYM. Herb-drug interactions and patient counseling. Int J Pharm Sci. 2009;1:151–61. [Google Scholar]
- 26.Ayano G. Psychotropic medications metabolized by cytochromes P450 (CYP) 1A2 enzyme and relevant drug interactions: review of articles. Austin J Pharmacol Ther. 2016;4:1085. [Google Scholar]
- 27.Latendresse G, Elmore C, Deneris A. Selective serotonin reuptake inhibitors as first-line antidepressant therapy for perinatal depression. JMWH. 2017;62:317–28. [DOI] [PubMed] [Google Scholar]
- 28.Onalan O, Cumurcu BE, Bekar L. Complete atrioventricular block associated with concomitant use of metoprolol and paroxetine. Mayo Clin Proc. 2008;83:595–9. 10.1016/S0025-6196(11)60736-2 [DOI] [PubMed] [Google Scholar]
- 29.Drake WM, Gordon GD. Heart block in a patient on propranolol and fluoxetine. Lancet. 1994;343:425–6. 10.1016/S0140-6736(94)91265-3 [DOI] [PubMed] [Google Scholar]
- 30.Garcia A. Adverse effects of propafenone after long-term therapy with the addition of citalopram. Am J Geriatr Pharmacother. 2008;6:96–9. 10.1016/j.amjopharm.2008.05.001 [DOI] [PubMed] [Google Scholar]
- 31.Sternbach H. Fluoxetine-associated potentiation of calcium-channel blockers. J Clin Psychopharmacol. 1991;11:390–1. 10.1097/00004714-199112000-00019 [DOI] [PubMed] [Google Scholar]
- 32.RokachHirsh B, Spectre G, Shai E, Lotan A, Ritter A, Al-Aieshy F. Differential impact of selective serotonin reuptake inhibitors on platelet response to clopidogrel: a randomized, double-blind, crossover trial. Pharmacotherapy. 2015;35:140–7. 10.1002/phar.1542 [DOI] [PubMed] [Google Scholar]
- 33.Woroń J, Siwek M, Wasik A. Interakcje leków w psychiatrii. AsteriaMed. 2019. [Google Scholar]
- 34.Bakker S, Burggraaf JLI, Kruip MJHA, van der Meer FJM, Lijfering WM, van Rein N. Selective serotonin reuptake inhibitor use and risk of major bleeding during treatment with Vitamin K antagonists: results of a cohort study. Thromb Haemost. 2022;123:245–54. 10.1055/a-1957-6305 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Andrade C. Selective serotonin reuptake inhibitor drug interactions in patients receiving statins. J Clin Psychiatry. 2014;75:95–9. 10.4088/JCP.13f08941 [DOI] [PubMed] [Google Scholar]
- 36.Jan CR, Su JA, Teng CC, Sheu ML, Lin PY, Chi MC, et al. Mechanism of maprotiline-induced apoptosis: role of [Ca2+]i, ERK, JNK and caspase-3 signaling pathways. Toxicology. 2013;304:1–12. 10.1016/j.tox.2012.11.013 [DOI] [PubMed] [Google Scholar]
- 37.McNamara YM, Bright SA, Byrne AJ, Cloonan SM, McCabe T, Williams DC, et al. Synthesis and antiproliferative action of a novel series of maprotiline analogues. Eur J Med Chem. 2014;71:333–53. 10.1016/j.ejmech.2013.10.076 [DOI] [PubMed] [Google Scholar]
- 38.Dean SN, van Hoek ML. Screen of FDA-approved drug library identifies maprotiline, an antibiofilm and antivirulence compound with QseC sensor-kinase dependent activity in Francisellanovicida. Virulence. 2015;6:487–503. 10.1080/21505594.2015.1046029 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Lloyd AH. Practical considerations in the use of maprotiline (ludiomil) in general practice. J Int Med Res. 1977;5:122–38. [PubMed] [Google Scholar]
- 40.Dahl ML, Leander K, Vikström M, Frumerie C, Nordenmalm S, Möller J, et al. CYP2D6-inhibiting drugs and risk of fall injuries after newly initiated antidepressant and antipsychotic therapy in a Swedish register based case-crossover study. Sci Rep. 2021;11:5796. 10.1038/s41598-021-85022-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Rotzinger S, Bourin M, Akimoto Y, Coutts RT, Baker GB. Metabolism of some “second”- and “fourth”-generation antidepressants: iprindole, viloxazine, bupropion, mianserin, maprotiline, trazodone, nefazodone, and venlafaxine. Cell Mol Neurobiol. 1999;19:427–42. 10.1023/A:1006953923305 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Lithium overdose. React. Wkly. 2002;894:9.
- 43.Maprotiline interaction. React. Wkly. 2002;888:9–10.
- 44.Dextromethorphan/maprotiline interaction. React. Wkly. 2013;1440:15.
- 45.Lentini S, Rao M, Schröder R, Lüderitz B, Bauriedel G. QT-verlängerung und torsade de pointes-tachykardie bei therapie mit maprotilin. Dtsch med Wochenschr. 2001;126:1396–400. 10.1055/s-2001-18880 [DOI] [PubMed] [Google Scholar]
- 46.Wells BG, Gelenberg AJ. Chemistry, pharmacology, pharmacokinetics, adverse effects, and efficacy of the antidepressant maprotiline hydrochloride. Pharmacotherapy. 1981;1:121–38. 10.1002/j.1875-9114.1981.tb03559.x [DOI] [PubMed] [Google Scholar]
- 47.Bertolín-Guillén JM, Climent-Díaz B, Navarré-Gimeno A. Serotonin syndrome due to association of venlafaxine Maprotiline and Reboxetine. Eur Psychiatr. 2004;19:456–7. 10.1016/j.eurpsy.2004.06.012 [DOI] [PubMed] [Google Scholar]
- 48.Alotaibi MS, Yousuf AZ, Khan A, Alshammari NS. Late presentation of linezolid-induced serotonin syndrome after maprotiline and mirtazapine therapy: a case report. Clin Neuropharmacol. 2021;44:71. 10.1097/WNF.0000000000000431 [DOI] [PubMed] [Google Scholar]
- 49.Yan XY, Yin PW, Wu XM, Han JX. Prediction of the drug-drug interaction types with the unified embedding features from drug similarity networks. Front Pharmacol. 2021. 10.3389/fphar.2021.794205. 10.3389/fphar.2021.794205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Blackwell B, Simon JS. Antidepressant drugs. Side Eff Drugs Annu. 1989;13:6–16. 10.1016/S0378-6080(89)80007-8 [DOI] [Google Scholar]
- 51.Sreter K, Barisic B, Popovic-Grle S. Pharmacogenomics and tailored polypharmacy: an 80-year-old lady with rosuvastatin-associated rhabdomyolysis and maprotiline-related Ogilvie’s syndrome. Int J Clin Pharmacol Ther. 2017;55:442. 10.5414/CP202784 [DOI] [PubMed] [Google Scholar]
- 52.Dionisie V, Filip GA, Manea MC, Manea M, Riga S. The anti-inflammatory role of SSRI and SNRI in the treatment of depression: a review of human and rodent research studies. Inflammopharmacol. 2021;29:75–90. 10.1007/s10787-020-00777-5 [DOI] [PubMed] [Google Scholar]
- 53.R.-B. Lu, Ed., Effects of Antidepressants. InTech. 2012.
- 54.Francescangeli J, Karamchandani K, Powell M, Bonavia A. The serotonin syndrome: from molecular mechanisms to clinical practice. Int J Mol Sci. 2019;9(20):2288. 10.3390/ijms20092288 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Knadler MP, Lobo E, Chappell J, Bergstrom R. Duloxetine. Clin Pharmacokinet. 2011;50:281–94. 10.2165/11539240-000000000-00000 [DOI] [PubMed] [Google Scholar]
- 56.Wernicke JF, Gahimer J, Yalcin I, Wulster-Radcliffe M, Viktrup L. Safety and adverse event profile of duloxetine. Expert Opin Drug Saf. 2005;4:987–93. 10.1517/14740338.4.6.987 [DOI] [PubMed] [Google Scholar]
- 57.Smith TR. Duloxetine in diabetic neuropathy. Expert Opin Pharmacother. 2006;7(215):223. [DOI] [PubMed] [Google Scholar]
- 58.Štuhec M. Excessive sweating induced by interaction between agomelatine and duloxetine hydrochloride: case report and review of the literature. Wien Klin Wochenschr. 2015;127:703–6. 10.1007/s00508-014-0688-0 [DOI] [PubMed] [Google Scholar]
- 59.Ma SP, Tsai CJ, Chang CC, Hsu WY. Delirium associated with concomitant use of duloxetine and bupropion in an elderly patient: duloxetine & bupropion induced delirium. Psychogeriatr. 2017;17:130–2. 10.1111/psyg.12202 [DOI] [PubMed] [Google Scholar]
- 60.Singh D, Saadabadi A. Venlafaxine. StatPearls Publishing. 2023. [PubMed]
- 61.Gareri P, Fazio PD, Gallelli L, Fazio SD, Davoli A, Seminara G, et al. Venlafaxine-propafenone interaction resulting in hallucinations and psychomotor agitation. Ann Pharmacother. 2008;42:434–8. 10.1345/aph.1K405 [DOI] [PubMed] [Google Scholar]
- 62.Troy SM, Lucki I, Peirgies AA, Parker VD, Klockowski PM, Chiang ST. Pharmacokinetic and pharmacodynamic evaluation of the potential drug interaction between venlafaxine and diazepam. J Clin Pharmacol. 1995;35:410–9. 10.1002/j.1552-4604.1995.tb04082.x [DOI] [PubMed] [Google Scholar]
- 63.Woroń J, Siwek M. Unwanted effects of psychotropic drug interactions with medicinal products and diet supplements containing plant extracts. Psychiatr Pol. 2018;52:983–96. 10.12740/PP/OnlineFirst/80998 [DOI] [PubMed] [Google Scholar]
- 64.Abo-Zena RA, Bobek MB, Dweik RA. Hypertensive urgency induced by an interaction of mirtazapine and clonidine. Pharmacotherapy. 2000;20:476–8. 10.1592/phco.20.5.476.35061 [DOI] [PubMed] [Google Scholar]
- 65.Davis MP, Khawam E, Pozuelo L, Lagman R. Management of symptoms associated with advanced cancer: olanzapine and mirtazapine. A world health organization project. Expert Rev Anticancer Ther. 2002;2:365–76. 10.1586/14737140.2.4.365 [DOI] [PubMed] [Google Scholar]
- 66.Timmer CJ, Ad Sitsen JM, Delbressine LP. Clinical pharmacokinetics of mirtazapine. Clin Pharmacokinet. 2000;38:461–74. 10.2165/00003088-200038060-00001 [DOI] [PubMed] [Google Scholar]
- 67.Duloxetine Capsules and Valerian Root - Drugs Interactions https://doktika.com/duloxetine_capsules-valerian_root. Accessed 25 Dec 2022.
- 68.Yue L, Juan L, Dongmin Y, Qian W, Jingyi J, Bo T, Furong Q. Influence of zuojin pill on the metabolism of venlafaxine in vitro and in rats and associated herb-drug interaction. Drug Metab Dispos. 2020;48:1044–52. 10.1124/dmd.120.000048 [DOI] [PubMed] [Google Scholar]
- 69.Wong EHF, Sonders MS, Amara SG, Tinholt PM, Piercey MFP, Hoffmann WP, et al. Reboxetine: a pharmacologically potent, selective, and specific norepinephrine reuptake inhibitor. Biol Psychiatr. 2000;47:818–29. 10.1016/S0006-3223(99)00291-7 [DOI] [PubMed] [Google Scholar]
- 70.Gao S, Zhang X, Xu H, Miao D, Qian J, Wu Z, Shi W. Promoting the hippocampal PPARα expression participates in the antidepressant mechanism of reboxetine, a selective norepinephrine reuptake inhibitor. Behav Brain Res. 2022;416:113535. 10.1016/j.bbr.2021.113535 [DOI] [PubMed] [Google Scholar]
- 71.Sepede G, Corbo M, Fiori F, Martinotti G. Reboxetine in clinical practice: a review. Clin Ter. 2012;163:255–62. [PubMed] [Google Scholar]
- 72.Zhou SF. Drugs behave as substrates, inhibitors and inducers of human cytochrome P450 3A4. CDM. 2008;9:310–22. 10.2174/138920008784220664 [DOI] [PubMed] [Google Scholar]
- 73.Fleishaker JC. Clinical pharmacokinetics of reboxetine, a selective norepinephrine reuptake inhibitor for the treatment of patients with depression. Clin Pharmacokinet. 2000;39:413–27. 10.2165/00003088-200039060-00003 [DOI] [PubMed] [Google Scholar]
- 74.Krakowski MI, Czobor P. A prospective longitudinal study of cholesterol and aggression in patients randomized to clozapine, olanzapine, and haloperidol. J Clin Psychopharmacol. 2010;30:198–200. 10.1097/JCP.0b013e3181d47b5e [DOI] [PubMed] [Google Scholar]
- 75.Italiano D, Spina E, de Leon J. Pharmacokinetic and pharmacodynamic interactions between antiepileptics and antidepressants. Expert Opin Drug Metab Toxicol. 2014;10:1457–89. 10.1517/17425255.2014.956081 [DOI] [PubMed] [Google Scholar]
- 76.Gohil K. Effect of simultaneous administrations of Curcuma longa extracts and SNRI- reboxetin on 2- week, chronic, oral, once daily administrations in animals: a research study. World J Pharm Pharm Sci 2015;4:1405–29. [Google Scholar]
- 77.Stahl SM, Pradko J, Haight BR, Modell JG, Rockett CB, Learned-Coughlin S. A review of the neuropharmacology of bupropion, a dual norepinephrine and dopamine reuptake inhibitor. Prim. Care Companion CNS Disord. 2004. 10.4088/PCC.v06n0403. 10.4088/PCC.v06n0403 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Huecker MR, Smiley A, Saadabadi A. Bupropion. StatPearls Publishing. 2024. [PubMed]
- 79.Rashidian A, Dejban P, Karami Fard K, Abdollahi A, Chamanara M, Dehpour A, Hasanvand A. Bupropion ameliorates acetic acid-induced colitis in rat: the involvement of the TLR4/NF-kB signaling pathway. Inflammation. 2020;43:1999–2009. 10.1007/s10753-020-01273-2 [DOI] [PubMed] [Google Scholar]
- 80.Foley KF, DeSanty KP, Kast RE. Bupropion: pharmacology and therapeutic applications. Expert Rev Neurother. 2006;6:1249–65. 10.1586/14737175.6.9.1249 [DOI] [PubMed] [Google Scholar]
- 81.Jefferson JW, Pradko JF, Muir KT. Bupropion for major depressive disorder: pharmacokinetic and formulation considerations. Clin Ther. 2005;27:1685–95. 10.1016/j.clinthera.2005.11.011 [DOI] [PubMed] [Google Scholar]
- 82.Spina E, Trifirò G, Caraci F. Clinically significant drug interactions with newer antidepressants. CNS Drugs. 2012;26:39–67. 10.2165/11594710-000000000-00000 [DOI] [PubMed] [Google Scholar]
- 83.Marcucci C, Sandson NB, Dunlap JA. Linezolid-bupropion interaction as possible etiology of severe intermittent intraoperative hypertension? Anesthesiology. 2004;101:1487–8. 10.1097/00000542-200412000-00051 [DOI] [PubMed] [Google Scholar]
- 84.Liu MZ, Zhang YL, Zeng MZ, He FZ, Luo ZY, Luo JQ, et al. Pharmacogenomics and herb-drug interactions: merge of future and tradition. CAM. 2015;2015:321091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Hu M, Fan L, Zhou HH, Tomlinson B. Theranostics meets traditional chinese medicine: rational prediction of drug-herb interactions. Expert Rev Mol Diagn. 2012;12:815–30. 10.1586/erm.12.126 [DOI] [PubMed] [Google Scholar]
- 86.Lei HP, Ji W, Lin J, Chen H, Tan ZR, Hu DL, Liu LJ, Zhou HH. Effects of ginkgo biloba extract on the pharmacokinetics of bupropion in healthy volunteers. Br J Clin Pharmacol. 2009;68:201–6. 10.1111/j.1365-2125.2009.03442.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Thase ME. MAOIs and depression treatment guidelines. J Clin Psychiatr. 2012;73:24. 10.4088/JCP.11096tx4c [DOI] [PubMed] [Google Scholar]
- 88.Baker GB, Coutts RT, McKenna KF, Sherry-McKenna RL. Insights into the mechanisms of action of the MAO inhibitors phenelzine and tranylcypromine: a review. J Psychiatr Neurosci. 1992;17:206–14. [PMC free article] [PubMed] [Google Scholar]
- 89.Müller T, Riederer P, Grünblatt E. Determination of monoamine oxidase A and B activity in long-term treated patients with parkinson disease. Clin neuropharmacol. 2017;40:208–11. 10.1097/WNF.0000000000000233 [DOI] [PubMed] [Google Scholar]
- 90.Thomas SJ, Shin M, McInnis MG, Bostwick JR. Combination therapy with monoamine oxidase inhibitors and other antidepressants or stimulants: strategies for the management of treatment-resistant depression. Pharmacotherapy. 2015;35:433–49. 10.1002/phar.1576 [DOI] [PubMed] [Google Scholar]
- 91.Fiedorowicz JG, Swartz KL. The role of monoamine oxidase inhibitors in current psychiatric practice. J Psychiatr Pract. 2004;10:239–48. 10.1097/00131746-200407000-00005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Brown C, Taniguchi G, Yip K. The monoamine oxidase inhibitor-tyramine interaction. J Clin Pharmacol. 1989;29:529–32. 10.1002/j.1552-4604.1989.tb03376.x [DOI] [PubMed] [Google Scholar]
- 93.Sathyanarayana Rao TS, Yeragani VK. Hypertensive crisis and cheese. Indian J Psychiatr. 2009;51:65–6. 10.4103/0019-5545.44910 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Mandrioli R, Protti M, Mercolini L. New-generation, non-ssri antidepressants: therapeutic drug monitoring and pharmacological interactions. Part 1: SNRIs, SMSs. SARIs Curr Med Chem. 2018;25:772–92. 10.2174/0929867324666170712165042 [DOI] [PubMed] [Google Scholar]
- 95.Kent JM. SNaRIs, NaSSAs, and NaRIs: new agents for the treatment of depression. Lancet. 2000;355:911–8. 10.1016/S0140-6736(99)11381-3 [DOI] [PubMed] [Google Scholar]
- 96.Shin JJ, Saadabadi A. Trazodone. StatPearls Publishing; 2023. [PubMed] [Google Scholar]
- 97.Rauch PK, Jenike MA. Digoxin toxicity possibly precipitated by trazodone. Psychosomatics. 1984;25:334–5. 10.1016/S0033-3182(84)73050-7 [DOI] [PubMed] [Google Scholar]
- 98.Weisler RH, Johnston JA, Lineberry CG, Samara B, Branconnier RJ, Billow AA. Comparison of bupropion and trazodone for the treatment of major depression. J Clin Psychopharmacol. 1994;14:170–9. 10.1097/00004714-199406000-00004 [DOI] [PubMed] [Google Scholar]
- 99.Socała K, Nieoczym D, Wyska E, Poleszak E, Wlaź P. Sildenafil, a phosphodiesterase type 5 inhibitor, enhances the activity of two atypical antidepressant drugs, mianserin and tianeptine, in the forced swim test in mice. Prog Neuro psychopharmacol Biol Psychiatr. 2012;38:121–6. 10.1016/j.pnpbp.2012.02.013 [DOI] [PubMed] [Google Scholar]
- 100.Olianas MC, Dedoni S, Onali P. The atypical antidepressant mianserin exhibits agonist activity at κ-opioid receptors. Br J Pharmacol. 2012;167:1329–41. 10.1111/j.1476-5381.2012.02078.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Chow T, Hiroi T, Imaoka S, Chiba K, Funae Y. Isoform-selective metabolism of mianserin by cytochrome P-450 2D. Drug Metab Dispos. 1999;27:1200–4. [PubMed] [Google Scholar]
- 102.Górska D, Andrzejczak D. Influence of mianserin on the activity of some hypotensive drugs in spontaneously hypertensive rats. Pol J Pharmacol. 2003;55:409–17. [PubMed] [Google Scholar]
- 103.Protti M, Mandrioli R, Marasca C, Cavalli A, Serretti A, Mercolini L. New-generation, non-SSRI antidepressants: drug-drug interactions and therapeutic drug monitoring. Part 2: NaSSAs, NRIs, SNDRIs, MASSAs, NDRIs, and others. Med Res Rev. 2020;40:1794–832. 10.1002/med.21671 [DOI] [PubMed] [Google Scholar]
- 104.Guardiola-Lemaitre B, De Bodinat C, Delagrange P, Millan MJ, Munoz C, Mocaër E. Agomelatine: mechanism of action and pharmacological profile in relation to antidepressant properties. Br J Pharmacol. 2014;171:3604–19. 10.1111/bph.12720 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Kronfeld-Schor N, Einat H. Circadian rhythms and depression: human psychopathology and animal models. Neuropharmacology. 2012;62:101–14. 10.1016/j.neuropharm.2011.08.020 [DOI] [PubMed] [Google Scholar]
- 106.Bertaina-Anglade V, la Rochelle CD, Boyer P-A, Mocaër E. Antidepressant-like effects of agomelatine (S 20098) in the learned helplessness model. Behav Pharmacol. 2006;17:703–13. 10.1097/FBP.0b013e3280116e5c [DOI] [PubMed] [Google Scholar]
- 107.Dubocovich ML. Agomelatine targets a range of major depressive disorder symptoms. Curr Opin Investig Drugs. 2006;7:670–80. [PubMed] [Google Scholar]
- 108.Saiz-Rodríguez M, Ochoa D, Belmonte C, Román M, Vieira de Lara D, Zubiaur P, Koller D, Mejía G, Abad-Santos F. Polymorphisms in CYP1A2, CYP2C9 and ABCB1 affect agomelatine pharmacokinetics. J Psychopharmacol. 2019;33:522–31. 10.1177/0269881119827959 [DOI] [PubMed] [Google Scholar]
- 109.Nadar JS, Kale PP, Kadu PK, Prabhavalkar K, Dhanger R. Potentiation of antidepressant effects of agomelatine and bupropion by hesperidin in mice. Neurol Res Int. 2018;2018:9826639. 10.1155/2018/9828639 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Szafarz M, Wencel A, Pociecha K, Fedak FA, Wlaź P, Wyska E. Pharmacokinetic study of tianeptine and its active metabolite MC5 in rats following different routes of administration using a novel liquid chromatography tandem mass spectrometry analytical method. Naunyn Schmiedebergs Arch Pharmacol. 2018;391:185–96. 10.1007/s00210-017-1448-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Gassaway MM, Rives ML, Kruegel AC, Javitch JA, Sames D. The atypical antidepressant and neurorestorative agent tianeptine is a μ-opioid receptor agonist. Transl Psychiatr. 2014;4:411. 10.1038/tp.2014.30 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Preskorn SH. Tianeptine: a facilitator of the reuptake of serotonin and norepinephrine as an antidepressant? J Psychiatr Pract. 2004;10:323. 10.1097/00131746-200409000-00006 [DOI] [PubMed] [Google Scholar]
- 113.Borowicz KK, Banach M, Piskorska B, Czuczwar SJ. Effect of acute and chronic tianeptine on the action of classical antiepileptics in the mouse maximal electroshock model. Pharmacol Rep. 2013;65:379–88. 10.1016/S1734-1140(13)71013-2 [DOI] [PubMed] [Google Scholar]
- 114.Otani K, Sasa H, Kaneko S, Kondo T, Fukushima Y. Steady-state plasma concentrations of mianserin and its major active metabolite. Desmethylmianserin Ther Drug Monit. 1993;15:113–7. 10.1097/00007691-199304000-00008 [DOI] [PubMed] [Google Scholar]
- 115.Drug Approval Package: Brand Name (Generic Name) NDA https://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/022567Orig1s000TOC.cfm. Accessed 01 Mar 2011.
- 116.Bartoszyk GD, Hegenbart R, Ziegler H. EMD 68843, a serotonin reuptake inhibitor with selective presynaptic 5-HT1A receptor agonistic properties. Eur J Pharmacol. 1997;322:147–53. 10.1016/S0014-2999(96)00999-5 [DOI] [PubMed] [Google Scholar]
- 117.Wang SM, Han C, Lee SJ, Patkar AA, Masand PS, Pae CU. A review of current evidence for vilazodone in major depressive disorder. Int J Psychiatry Clin Pract. 2013;17:160–9. 10.3109/13651501.2013.794245 [DOI] [PubMed] [Google Scholar]
- 118.Sanchez C, Asin KE, Artigas F. Vortioxetine, a novel antidepressant with multimodal activity: review of preclinical and clinical data. Pharmacol Ther. 2015;145:43–57. 10.1016/j.pharmthera.2014.07.001 [DOI] [PubMed] [Google Scholar]
- 119.Chen G, Højer AM, Areberg J, Nomikos G. Vortioxetine: clinical pharmacokinetics and drug interactions. Clin Pharmacokinet. 2018;57:673–86. 10.1007/s40262-017-0612-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Siwek M. Zastosowanie Wortioksetyny w Leczeniu Zaburzeń Depresyjnych. Psychiatr. 2017;14:7–20. [Google Scholar]
- 121.Yao Y, Ju P, Liu H, Wu X, Niu Z, Zhu Y, Zhang C, Fang Y. Ifenprodil rapidly ameliorates depressive-like behaviors, activates mTOR signaling and modulates proinflammatory cytokines in the hippocampus of CUMS rats. Psychopharmacology. 2020;237:1421–33. 10.1007/s00213-020-05469-0 [DOI] [PubMed] [Google Scholar]
- 122.Kotajima-Murakami H, Takano A, Ogai Y, Tsukamoto S, Murakami M, Funada D, et al. study of effects of ifenprodil in patients with methamphetamine dependence: protocol for an exploratory, randomized, double-blind placebo-controlled trial. Neuropsychopharmacol Rep. 2019;39:90–9. 10.1002/npr2.12050 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Fraser CM, Cooke MJ, Fisher A, Thompson ID, Stone TW. Interactions between ifenprodil and dizocilpine on mouse behaviour in models of anxiety and working memory. ENP. 1996;6:311–6. [DOI] [PubMed] [Google Scholar]
- 124.Ghasemi M, Raza M, Dehpour A. NMDA receptor antagonists augment antidepressant-like effects of lithium in the mouse forced swimming test. J Psychopharmacol. 2010;24:585–94. 10.1177/0269881109104845 [DOI] [PubMed] [Google Scholar]
- 125.Ghasemi M, Montaser-Kouhsari L, Shafaroodi H, Nezami BG, Ebrahimi DAR. NMDA receptor/nitrergic system blockage augments antidepressant-like effects of paroxetine in the mouse forced swimming test. Psychopharmacology. 2009;206:325–33. 10.1007/s00213-009-1609-1 [DOI] [PubMed] [Google Scholar]
- 126.Adaptogenic Concept - Scientific Guideline. EMA https://www.ema.europa.eu/en/adaptogenic-concept-scientific-guideline. Accessed 08 May 2008.
- 127.Panossian A, Wikman G, Wagner H. Plant adaptogens. III. Earlier and more recent aspects and concepts on their mode of action. Phytomedicine. 1999;6:287–300. 10.1016/S0944-7113(99)80023-3 [DOI] [PubMed] [Google Scholar]
- 128.Wagner H, Nörr H, Winterhoff H. Plant adaptogens. Phytomedicine. 1994;1:63–76. 10.1016/S0944-7113(11)80025-5 [DOI] [PubMed] [Google Scholar]
- 129.Oliynyk S, Oh S. The pharmacology of actoprotectors: practical application for improvement of mental and physical performance. Biomol Ther. 2012;20:446–56. 10.4062/biomolther.2012.20.5.446 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Panossian AG, Efferth T, Shikov AN, Pozharitskaya ON, Kuchta K, Mukherjee PK, et al. Evolution of the adaptogenic concept from traditional use to medical systems: pharmacology of stress- and aging-related diseases. Med Res Rev. 2021;41:630–703. 10.1002/med.21743 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Siwek M, Woroń J, Wrzosek A, Gupało J, Chrobak AA. Harder, better, faster, stronger? retrospective chart review of adverse events of interactions between adaptogens and antidepressant drugs. Front Pharmacol. 2023;14:1271776. 10.3389/fphar.2023.1271776 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Woroń J, Chrobak AA, Ślęzak D, Siwek M. Unprescribed and unnoticed: retrospective chart review of adverse events of interactions between antidepressants and over-the-counter drugs. Front Pharmacol. 2022;13:965432. 10.3389/fphar.2022.965432 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Woodcock J. FDA concludes that existing regulatory frameworks for foods and supplements are not appropriate for cannabidiol, will work with Congress on a New Way Forward. 2023. https://www.fda.gov/news-events/press-announcements/fda-concludes-existing-regulatory-frameworks-foods-and-supplements-are-not-appropriate-cannabidiol
- 134.Bonaccorso S, Ricciardi A, Zangani C, Chiappini S, Schifano F. Cannabidiol (CBD) use in psychiatric disorders: a systematic review. NeuroToxicol. 2019;74:282–98. 10.1016/j.neuro.2019.08.002 [DOI] [PubMed] [Google Scholar]
- 135.Balachandran P, Elsohly M, Hill KP. Cannabidiol interactions with medications, illicit substances, and alcohol: a comprehensive review. J Gen Intern Med. 2021;36:2074–84. 10.1007/s11606-020-06504-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No datasets were generated or analyzed in the current study.
