Next Article in Journal
Effect of the App-Based Video Guidance on Prenatal Pelvic Floor Muscle Training Combined with Global Postural Re-education for Stress Urinary Incontinence Prevention: A Protocol for a Multicenter, Randomized Controlled Trial
Next Article in Special Issue
Vestibular Disorders after Kidney Transplantation: Focus on the Pathophysiological Mechanisms Underlying the Vertical Nystagmus Associated with Tacrolimus-Related Hypomagnesamia
Previous Article in Journal
New Insights on the Mediating Role of Emotional Intelligence and Social Support on University Students’ Mental Health during COVID-19 Pandemic: Gender Matters
Previous Article in Special Issue
Videoconference-Based Adapted Physical Exercise Training Is a Good and Safe Option for Seniors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Drug–Drug Interactions in Vestibular Diseases, Clinical Problems, and Medico-Legal Implications

by
Giulio Di Mizio
1,†,
Gianmarco Marcianò
2,†,
Caterina Palleria
2,
Lucia Muraca
2,3,
Vincenzo Rania
2,
Roberta Roberti
2,
Giuseppe Spaziano
4,
Amalia Piscopo
1,
Valeria Ciconte
1,2,
Nunzio Di Nunno
5,
Massimiliano Esposito
6,
Pasquale Viola
7,
Davide Pisani
7,
Giovambattista De Sarro
2,8,
Milena Raffi
9,*,
Alessandro Piras
9,
Giuseppe Chiarella
7,‡ and
Luca Gallelli
2,8,10,‡
1
Department of Law, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
2
Department of Health Science, School of Medicine, University of Catanzaro, Clinical Pharmacology and Pharmacovigilance Unit, Mater Domini Hospital, 88100 Catanzaro, Italy
3
Department of Primary Care, ASP 7, 88100 Catanzaro, Italy
4
Department of Experimental Medicine L. Donatelli, Section of Pharmacology, School of Medicine, University of Campania Luigi Vanvitelli, 80123 Naples, Italy
5
Department of History, Society and Studies on Humanity, University of Salento, 83100 Lecce, Italy
6
Department of Medical, Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, University of Catania, 95121 Catania, Italy
7
Unit of Audiology, Department of Experimental and Clinical Medicine, Regional Centre of Cochlear Implants and ENT Diseases, Magna Graecia University, 88100 Catanzaro, Italy
8
Research Center FAS@UMG, Department of Health Science, University of Catanzaro, 88100 Catanzaro, Italy
9
Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
10
Medifarmagen SRL, University of Catanzaro, 88100 Catanzaro, Italy
*
Author to whom correspondence should be addressed.
Marcianò and Di Mizio share the authorship.
Gallelli and Chiarella share the authorship.
Int. J. Environ. Res. Public Health 2021, 18(24), 12936; https://doi.org/10.3390/ijerph182412936
Submission received: 31 October 2021 / Revised: 28 November 2021 / Accepted: 3 December 2021 / Published: 8 December 2021

Abstract

:
Peripheral vestibular disease can be treated with several approaches (e.g., maneuvers, surgery, or medical approach). Comorbidity is common in elderly patients, so polytherapy is used, but it can generate the development of drug–drug interactions (DDIs) that play a role in both adverse drug reactions and reduced adherence. For this reason, they need a complex kind of approach, considering all their individual characteristics. Physicians must be able to prescribe and deprescribe drugs based on a solid knowledge of pharmacokinetics, pharmacodynamics, and clinical indications. Moreover, full information is required to reach a real therapeutic alliance, to improve the safety of care and reduce possible malpractice claims related to drug–drug interactions. In this review, using PubMed, Embase, and Cochrane library, we searched articles published until 30 August 2021, and described both pharmacokinetic and pharmacodynamic DDIs in patients with vestibular disorders, focusing the interest on their clinical implications and on risk management strategies.

1. Introduction

The function of balance is regulated by the vestibular system. The main subdivision of this system, includes a peripheral portion (the posterior labyrinth, in the inner ear, and the vestibular portion of the eighth cranial nerve) and a central portion (vestibular nuclei, the oculomotor nuclei, the vestibulo–ocular reflex tracts, the cerebellum, the brainstem reticular formation, the area postrema, and other associated areas, including many refined connections with cognitive areas) [1]. Both, peripheral and central, acute and chronic impairment of the vestibular system determine the appearance of vertigo/dizziness with other symptoms (e.g., postural imbalance, nausea, vomiting, and in some particular clinical pictures, also sensorineural hearing loss and tinnitus) [2,3,4].
Vertigo is a highly disabling disorder that impairs the daily activities, especially in elderly patients, exposing this population to the risk of falling.
In elderly, a very significant complication is represented by both multiorgan impairment and alteration of pharmacokinetic parameters. Therefore, several factors—(i) polytherapy, (ii) changes in body composition, (iii) renal or liver impairment—play a role in the development of adverse drug reactions (ADRs) [5] and also induce an increased vulnerability of older patients [6,7].
In addition, polytherapy can increase the risk of drug–drug interactions (DDIs) and drug–disease interactions that are involved in falls, longer time of hospitalization, and even death [6,8].
Many pharmacological treatments modulate the activity of neurotransmitters, neuromodulators, and voltage-gated channels involved in the modulation of neuronal excitability.
Among all of these drugs, cinnarizine is the first-line therapy for the management of some kind of vertigo. It may act on the peripheral vestibular system with several mechanism [9,10,11,12] able to induce both an anti-vasoconstrictor activity and a decrease in blood viscosity of the inner ear’s circulatory system [11]. Usually in clinical studies, cinnarizine is used at different doses, from 15 mg tid to 150 mg od; however, the common dosage is 25 mg tidor 75 mg od, up to a maximum of 225 mg [13]. Today, cinnarizine is administered with dimenhydrinate, an histamine (H1) receptor antagonist and phosphodiesterase inhibitor in the vestibular nuclei and the vomiting center, with anti-vertigo and anti-emetic effects [14,15].
Different mechanisms of action, related to the drug use, reduce the strength of symptoms (e.g., vestibular suppressants) or modify the underlying disease processes that have led to the development of symptoms (e.g., calcium channel antagonists in the case of vestibular migraine).
Previously, we reported that DDIs represent a risk factor in poly-treated patients, because they can induce the development of ADRs [16,17,18,19]. In the present review we assessed the possibility of DDIs and ADRs in elderly people with vestibular disease.

2. Methods

The PubMed, Embase and Cochrane library databases were searched for articles published until 30 August 2021 in English language, in agreement with our recent papers [20,21].
Secondary search included articles cited in reference lists identified by the primary search. Records were first screened by title/abstract before full-text articles were retrieved for eligibility evaluation. Remaining articles were then subject to a citation search of all reference lists. Papers were deemed eligible if they included any form of words: “adverse drug reaction”, “drug interactions”, “polytherapy”, “vestibular disease/s”, “systemic diseases”. All citations were downloaded into Mendeley, and duplicates deleted. GM and PV screened all articles by title/abstract to determine their eligibility and RR reviewed a random sample of 20% to evaluate the reliability of the selection process. To avoid a bias of exclusion, the full text articles were retrieved following first round exclusions and were also subject to two independent eligibility reviews (CP 100%, LM 20%), this time with perfect agreement. The studies evaluated as eligible were enclosed in the present review. We excluded manuscript without full text and without indications of adverse drug reactions. Moreover, we also excluded manuscript not in English language. MR revised the manuscript elucidating the physiological mechanisms.

2.1. Drugs Used in the Management of Peripheral Vestibular Vertigo

2.1.1. Betahistine

Betahistine dihydrochloride is an oral drug, a strong antagonist of histamine H3 receptors and a weak agonist of H1 receptors [22]. Betahistine increases vasodilation and restores microcirculation [23] and blood flow, but also facilitates vestibular compensation and inhibits spike generation in lateral and medial vestibular nuclei [24]. Its main clinical indication is represented by MD (Meniere’s disease) [22], but it may be also used in BPPV (benign paroxysmal vertigo, in selected patients) [25], vestibular neuritis, and other cases of peripheral vestibular vertigo [24].
A systematic review by Murdin et al. [26] assessed betahistine’s efficacy in the treatment of vertigo despite low level of evidence. According to the BEMED trial performed by Adrion et al., low and high dose betahistine seem to be not superior to placebo on reducing the incidence of attacks caused by MD. Both placebo and betahistine lowered attack ratio and this result was probably related to natural history of MD [27]. Another systematic review showed no efficacy of betahistine in MD, sustaining the need of high quality studies [28].
Monoamine oxidases (MAOs) A/B are responsible of betahistine metabolism, therefore MAO inhibitors may increase its concentration [23,29], inducing the development of few ADRs [23,27]: (Table 1, Table 2, Table 3, Table 4, Table 5, Table 6, Table 7 and Table 8).

2.1.2. Diuretic Therapy

Diuretic drugs are commonly used for the treatment of hypertension and heart failure. Their action is mainly characterized by fluid excretion [217].
Endolymphatic hydrops plays a key role in the pathogenesis of MD. Therefore, fluids reduction is a rational strategy of treatment. Diuretics showed a certain efficacy in the control of hearing loss and vertigo, especially in patients with non-advanced disease. However, further analysis demonstrated that they didn’t prevent a long-term deterioration of hearing [41]. A systematic review by Crowson et al. [218] stated that diuretics are more effective in the prevention of vertigo in patient with MD, with a lower action on hearing outcomes.
Hydrochlorothiazide/triamterene, acetazolamide, spironolactone are the most important diuretics used in MD, whereas loop diuretics may be employed, but need a close monitoring of ADRs [22,41,219].
Hydrochlorothiazide and acetazolamide are carbonic anhydrase inhibitors, whereas triamterene and spironolactone have a potassium sparing mechanism (Table 1).

2.1.3. Acetazolamide

According to magnetic resonance imaging (MRI) study, Sepahdari et al. documented that acetazolamide reduces endolymphatic hydrops in subjects with MD. However, the action of acetazolamide could not be effective on hydrops in all patients and fluid could increase after therapy discontinuation [220].

2.1.4. Hydrochlorothiazide

Hydrochlorothiazide is a thiazide-type diuretic. It exerts its action inhibiting sodium chloride co-transporter in the distal convoluted tubule [31,32]. Its role in MD is well described [219,221], and it can be commonly administered with triamterene [22,41] (Table 1, Table 2, Table 3 and Table 4).

2.1.5. Triamterene

Triamterene, a potassium-sparing diuretic, inhibits the epithelium sodium channel (ENaC), avoiding sodium-potassium exchange [35,38]. Triamterene is often administered with hydrochlorothiazide since it enhances the effect of hydrochlorothiazide in lowering blood pressure and prevents hypokalemia. In MD the two drugs are often prescribed in a coformulation [22,35,41] (Table 1, Table 2, Table 3 and Table 4).

2.1.6. Spironolactone

Spironolactone is a potassium-sparing diuretic. It is a mineralocorticoid receptor antagonist which blocks the consequential action of aldosterone on ENaC (Epithelium Sodium Channel). Spironolactone determines the excretion of chlorine, sodium and water, whereas potassium is retained [33,34,35], ad is recommended in MD [22,41].
In an experimental study, Degerman et al. showed that spironolactone blocks the phosphodiesterase 4 (PDE4) pathway, involved in the generation of endolymphatic hydrops [222].
Other studies described a positive effect of aldosterone on endolymphatic hydrops that is related to salt-reduced diet (generating an increase of aldosterone levels), that is part of the management of MD [223] (Table 1, Table 2, Table 3 and Table 4).

2.2. Vestibular Suppressants

2.2.1. Benzodiazepines

Benzodiazepines (BDZ) are recommended in MD, VN and may be used for particular classes of patients in BPPV [22,25,41,42,56]. BDZ act on GABAA receptor as allosteric modulators, increasing receptor affinity for its substrate, with an increase in GABA inhibitory activity [39,106]. Despite the effectiveness of BDZ being demonstrated [22,224,225,226,227], it has been suggested that they might impair compensation for vestibular lesions [228]. In fact, GABAB activation seems to be involved in vestibular compensation after damage [229,230]. Diazepam, lorazepam, and clonazepam are more frequently used in the treatment of vestibular peripheral vertigo [22,228], due to short onset of action [41].
Lorazepam is metabolized by conjugation with glucuronic acid, diazepam is metabolized by CYP2C19 and CYP3A4 (part of its metabolites is conjugated) and clonazepam by CYP3A4 [43,45,46,81,82,84]. Inhibition/induction of these CYPs may generate increase/reduction of drugs concentrations (see Table 6).
BDZ main adverse events are neurologic symptoms (somnolence, motor difficulties, amnesia, difficulty concentrating, headache, vertigo, behavioral changes) and gastrointestinal symptoms. Incontinence, articular pain, hypersensitivity, hepatic damage, and hematologic alterations are rare. Abstinence, abuse, and tolerance are very common after BDZ administration. To avoid this kind of reaction physicians should use a gradual taper to discontinue BDZ, use a lower dosage. High dose and prolonged treatment are more frequently associated to these side effects [40,43,45,46,81,82,84].

2.2.2. Anticholinergics

Anticholinergics are used as vestibular suppressants in MD, BPPV, VN, and BV [22,25,41,42,56]. The most common anticholinergics are scopolamine, atropine and glycopyrrolate [22,41]. These drugs, acting on acetylcholine (ACh), inhibit the ACh muscarinic receptor (mAChR) [22,40]. Clinical trials showed a certain efficacy of scopolamine (transdermal route) [55] and glycopyrrolate (oral route) [50] in the control of vertigo. ACh pathways seem to have a crucial role in the activity of the mammalian vestibular system. In an experimental model, Schneider et al. described that atropine and scopolamine inhibited vestibular spike responses in mice [231]. Other authors sustain that muscarinic and nicotinic receptors are involved in both inhibitory and excitatory responses [232] (Table 1, Table 2, Table 3 and Table 4).
Glycopyrrolate does not pass the blood–brain barrier; therefore, it has less neurologic side effects. It also shows minor ocular and cardiovascular events [40,49].

2.2.3. Antihistamines

Antihistamines are commonly used as vestibular suppressants in the treatment of peripheral vestibular vertigo. Dimenhydrinate, meclizine and diphenhydramine (first-generation antihistamines) are the most common compounds. Diphenhydramine is a dimenhydrinate metabolite [22,25,40,41,56]. They exert their action blocking histamine receptor H1 [40].Cinnarizine, a calcium channel blocker, may be administered in combination with dimenhydrinate: The combination was more effective than monotherapy [14,233]. Phenothiazines are antipsychotic drugs that also act on histaminergic pathways and may be employed in the treatment of vertigo (e.g., promethazine) [41].An experimental model by Takatani et al. described an inhibitory action of diphenhydramine more selectively in medial vestibular nucleus (MVN) rather than lateral vestibular nucleus (LVN). Evidence about an important role of histaminergic pathways in vestibular system come from experimental models, showing a role of H1, H2, and H3 receptors. Counterintuitively, a high expression of H1 and H2 receptors is associated with a better vestibular compensation [62,234,235]. Not only the activity of histaminergic receptors seems to be effective in improving balance, but they also seem to modulate GABA pathways [234], reducing the concentration of the inhibitory neurotransmitter [235]. However, further studies are needed.
This positive role of histamine receptors seems to assess the need of a histaminergic agonist to treat vertigo, and betahistine (strong H3 antagonist and weak H1 agonist) was the main candidate [234]. However, trials described a superiority of betahistine compared with promethazine [236], but a minor effect than cinnarizine/dimenhydrinate [237]. Furthermore, a systematic review showed that evidence for betahistine in the treatment of peripheral vestibular vertigo has a low level of quality [26].Moreover, the clinical effectiveness of the other H1 antagonists was observed in other clinical trials [22,238,239,240].
Promethazine, meclizine, dimenhydrinate, and diphenhydramine are contraindicated in patients affected by lower respiratory tract infections, asthma [57,61,67,69].Physicians must pay attention also in cardiovascular diseases (promethazine) [69]. Dimenhydrinate should be used with caution in porphyria [57].Diphenhydramine is also contraindicated in pheochromocytoma, arrythmias (or risk of rhythm alterations) [61].
Diphenhydramine is a substrate and an inhibitor of CYP2D6 [92]. It is also metabolized with minor affinity by CYP1A2, CYP2C9 and CYP2C19 [92]. Meclizine reduces the expression of CYP2B10, 3A11, 1A2 [65], and it is metabolized by CYP2D6 [94].
Cinnarizine seems to be mainly metabolized by CYP2D family (CYP2D6) and CYP2B6, although other cytochromes are secondarily involved [88,89].
Meclizine is an agonist for murine constitutive androstane receptor (mCAR) and an inverse agonist for human constitutive androstane receptor (hCAR), involved in the regulation of hepatic enzymes [65].

2.2.4. Other Antiemetics

Other drugs may be used for the management of symptoms related to vertigo. Among them, 5HT3 antagonists play an important role. This category includes ondansetron (the most used), granisetron, tropisetron, dolasetron, and palonosetron (second generation antagonist) [40]. In an experimental model by Zhang et al., serotonin had an important role in vestibular compensation: the increase of its levels was documented after semi-circular canal occlusion [241]. Ondansetron showed a better effect in the control of nausea and vomiting, but a minor efficacy in the treatment of peripheral vertigo in comparison with promethazine [238]. Nevertheless, an experimental model by Dyhrfjeld-Johnsen etal. showed a positive post-insult effect of ondansetron in vestibular neuritis [242]. Ondansetron may be also used in the management of MD [41]. It is metabolized by CYP1A2, CYP2D6 and CYP3A4 [40,75,99], and It is also a P glycoprotein (P-gp) substrate [100].
Metoclopramide is a pro-kinetic drug used in the management of nausea and vomiting. It acts on 5HT4 (agonist), 5HT3 (antagonist) and dopamine D2 (antagonist) receptors [40,71,72]. Dopamine may have an important role in vestibular activity. Jansen et al. showed a reduction of D2/D3 receptor in patients with bilateral vestibular failure. They speculate that this may obstacle vestibular compensation. Dopamine pathways are complex and regulate vestibular activity involving also cortex and basal ganglia [243]. An experimental model by Toro et al. showed a dopamine modulation of sensory hair cells activity acting in a paracrine fashion [244]. Metoclopramide had a similar efficacy to dimenhydrinate in reducing vertigo, nausea and vomiting in an emergency setting [245]. Metoclopramide (non-specific compound) is commonly prescribed in the management of MD, BPPV, VN, and other forms of vertigo [73].Metoclopramide is metabolized to mono-de-ethyl-metoclopramide at least in part by CYP2D6 isoform, and possibly CYP1A2 and CYP3A [97].

2.2.5. Corticosteroids

Glucocorticoids have anti-inflammatory, anti-pyretic, and anti-allergic activity [133]. They modulate immune response through different mechanisms, for example, encoding pro-inflammatory cytokines IL-4, IL-10, IL-13, and TGFβ or inhibiting activation of nuclear factor (NF)-κB genes [246]. They act on the hypothalamic-pituitary-adrenal (HPA) axis [105,247]. Oral corticosteroids may be used in MD to reduce inflammatory response or to treat exacerbations. Evidence about their application is limited and their usage is based on expert opinion [248]. Even if dexamethasone, methylprednisolone and prednisone are the most common corticosteroids used [41], their role in VN is controversial. In fact, some authors suggest to start the treatment until 72h [56], whereas a systematic review failed to report a strong evidence [249]. Recent works described a role of corticosteroids in accelerating the recovery, without affecting prognosis [250]. Dexamethasone, methylprednisolone, and prednisone are metabolized by CYP3A4 [131,132,133,134]. Dexamethasone induces CYP3A4 and is both a substrate and inducer of P-gp [133,147]

2.2.6. Other Treatment Options

Calcium antagonists other than cinnarizine (flunarizine and verapamil) may be useful in MD, especially in patients with migraine as prophylactic medications [219]. Flunarizine showed good results (clinical improvement) in a small court of 32 patients [251].
However, flunarizine efficacy was inferior to betahistine in MD in clinical trial [252].
A prostaglandin F2α analogue, latanoprost, may have a certain efficacy, since the F2 receptor has been found in inner ear. Intratympanic administration was associated with MD symptoms improvement in a small court, but further studies are needed [219].
Compounds mimicking and inducing glutathione peroxidase may exert anti-oxidant and anti-inflammatory effect in MD. Ebselen is a drug with these characteristics: Its administration in a phase 2 trial led to improvement of hearing and tinnitus in MD [219].
These drugs are not mentioned in MD guidelines [22].
Intratympanic gentamicin and intratympanic corticosteroids are two therapeutic options used in surgical setting [219,253].

3. Discussion

DDIs are an important concern in clinical practice. Assessing the relevance of DDIs is not easy since this task needs pharmaceutical, chemical, and clinical skills. Risk: benefit ratio should be always considered in the evaluation of DDIs. Minor interactions may be accepted, if necessary, when patients obtain a tangible improvement of their conditions [254]. Scott et al. defined deprescribing as “the systematic process of identifying and discontinuing drugs in instances in which existing or potential harms outweigh existing or potential benefits within the context of an individual patient’s care goals, current level of functioning, life expectancy, values, and preferences” [254]. This kind of approach is usually needed with elderly people, since they consume multiple medications, creating space for interactions [255]. Potentially inappropriate medications (PIM) generate an important part of DDIs. Their role is crucial because the reduction of their prescription or discontinuation may consistently reduce DDIs. When evaluating a patient, physician must be aware of all the drugs taken, consider the risk related to a specific drug and, above all, the real presence of a clinical indication. In fact, PIM will not only lead to no benefit, but will probably determine adverse events [254,255]. Therefore, physician should prescribe therapies according to guidelines and specific characteristics of patient.
Ageing is associated with multiple pathophysiological changes. An increase of adipose tissue may be observed in elderly people, especially women: then, the volume of distribution (Vd) of lipophilic drugs may be increased. Conversely, the reduction of body water is associated to a reduction of Vd for hydrophilic drugs. Minor differences in CYP450 metabolism were observed, whereas significant changes may be described in hepatic first pass metabolism (with a possible increase of bioavailability), α1acid glycoprotein (often increased), renal elimination (worsening). More data about transporters activity are needed [5,127]. These factors should be held in account in the management of elderly patients.
Sex differences should be evaluated. Vd is generally higher in men due to their larger body. However, women have a major percentage of fat compared to men, but the difference decreases with older age. This factor may be important in the evaluation of lipophilic drug distribution: For example, diazepam has a larger Vd in women; therefore, it may show an increased duration of its effect. Renal processes are faster in men, whereas CYP450 activity may vary greatly based on the single isoform, or the specific substance metabolized. CYP1A, CYP2D6 metabolism and hepatic conjugation are generally faster in men, whereas CYP3A activity seems to be quicker in women. These observations were often made on young and healthy subjects [256]. Specific and generalizable data about pharmacokinetics sex differences in elderly people are lacking. However, elderly females are predicted to have the lowest elimination rates, since they live longer than men and according to age-related increase of comorbidities, clearance reduction, and decrease of body size [256]. Another important statement is that there are important differences in adaptive homeostasis between males and females. Nevertheless, these sex-related distinctions seem to be lost with age [257]. Furthermore, sex-aging related differences in genomic instability were found, including a major tendency of DNA mutations, telomere attrition and sometimes methylation in men [258]. This assumption may affect gene expression related drug development, one of the present and future direction of pharmaceutic industry [259,260]. In addition, physicians should remember that females live longer than males, but have a higher risk of frailty and morbidity. Despite being less affected by comorbidities, males are characterized by a higher level of physiological dysregulation. Authors called it “male–female dysregulation frailty paradox” [257,261]. This concept may influence a therapy including multiple drugs.
Interaction in elderly may be various and unpredictable because this category of patients consumes multiple medications.
Drugs with anticholinergic (anticholinergic and antihistamines) effect should be avoided in patients suffering for urinary retention [262]. The concomitant consumption of cholinergic drugs is another contraindication to the use of this kind of therapy [54].In contrast, a difficulty in clinician’s decisions can be described in dementia or other neurologic pathologies related to urinary incontinence [263]. In fact, anticholinergic drugs (or compounds with anticholinergic effect) may worsen cognitive impairment. Crossing the blood-brain barrier (BBB) anticholinergics block muscarinic receptors M1 and M4. Furthermore, these drugs may interact with cholinesterase inhibitors, used in Alzheimer’s disease [198,263].
Diabetes is a difficult setting. If patients develop peripheral vestibular vertigo, metformin-glipizide and liraglutide are associated with dizziness. Other compounds may create difficulties in balance, affecting vision [264]. Diabetic neuropathy may cause urinary incontinence [265], since using anticholinergics to treat vertigo may be a right choice.
Acarbose is associated with flatulence and diarrhea, some of the most frequent adverse events of metoclopramide and other vestibular suppressants [72,138].
Cardiovascular setting is difficult since several interactions may happen (Table 5, Table 6 and Table 7). Despite some action of CYP450 enzymes on ACE inhibitors been described [102], they seem not to be part of important interactions, whereas angiotensin receptor blockers (ARB) may be involved in this kind of mechanism [101]. Nevertheless, the role of CYP450 enzymes on ARB metabolism is not always clear [103] and then predicting interaction is difficult.
NSAIDs are also associated with dizziness and then they may obstacle the treatment of vertigo [18].
Obstructive sleep apnea syndrome (OSAS) seems to be related to vertigo and to vestibular system. A reciprocal influence and neuroanatomical route between vestibular nuclei and orexinergic neurons have been reported. Sleep also stimulates neuroplasticity that helps vestibular compensation. Therefore, OSAS and sleep deprivation impair vestibular control, but also vestibular pathologies induce sleep disturbances [266,267]. Vestibular-evoked myogenic potentials may be used to evaluate vestibular lesions at the early stage of OSAS [268]. Furthermore, continuous positive airway pressure therapy in refractory MD patients with concomitant OSAS demonstrated an improvement of dizziness and audiometric testing [269]. According to these observations, the treatment of dizziness may improve OSAS. In this clinical situation, it is important to avoid drugs inhibiting respiratory system (e.g., opioids, barbiturates) [270]. Although characterized by a lower potential of respiratory depression, benzodiazepines may be associated with acute respiratory failure in patients with sleep apnea [40,271]. Physicians must remember it in the management of peripheral vestibular vertigo with concomitant pneumological pathologies. Furthermore, anticholinergics and antihistamines seem to reduce respiratory secretions generating rare and mild respiratory symptoms [40,49,53,54,61,214]. Atropine may generate respiratory depression [47].
Gastrointestinal symptoms or illness should be evaluated carefully. The induction of diarrhea by metoclopramide [74] may be troublesome in the management of electrolytic disturbances, gastrointestinal infections, colon cancer, and diverticulosis.
Oncologic patients may experience ototoxicity when treated with platinum compounds, surgery, or radiotherapy. This may also worsen or ingenerate vertigo [272]. Physician must pay attention to vertigo in oncologic patients since it may be the sign of brain metastasis. A series of red flags may add value to the clinical hypothesis: altered mental status, ataxia, headache, visual symptoms, seizure, and focal signs and symptoms [273]. Some of the drugs used for the management of peripheral vestibular vertigo may generate headache (e.g., ondansetron, triamterene, benzodiazepines, betahistine) [23,36,45,75,76,80,82] and blurred vision (e.g., antihistamines, anticholinergics) [274], therefore clinician must distinguish these side effects from cancer spread.
The susceptibility to infection in the elderly is increased by immune senescence as well as altered skin and mucosal barrier function [275].Among antibiotics, macrolides, aminoglycosides, and glycopeptides may generate ototoxicity, worsening clinical status of patients with peripheral vestibular vertigo [276].
An association between osteoporosis and BPPV has been reported, mainly in women [277]. Use of corticosteroids in this clinical setting may increase the risk of bone fracture [278]. Bisphosphonates undergo negligible liver metabolism [40], so no important interactions are expected. In the past, episodic vertigo after bisphosphonates intravenous administration was described [279]. On the other side, vertigo may increase the risk of falls in elderly people with osteoporosis, raising the possibility of fracture [280].
Rheumatology may be another field of interactions. For example, triamterene, anticholinergics, and antihistamines are associated with dry mouth [36,40,53,57,61], that could be an important issue in patients with Sjogren’s syndrome (SS) [281]. Hydrochlorothiazide may cause skin manifestation (common in rheumatologic pathologies) and SLE exacerbations [31,282,283]. Hydrochlorothiazide and triamterene may generate hyperuricemia, that would be a trouble in the management of gout [36,80,284].
Liver and kidney function is often altered in elderly people. CYP450 function is reduced in elderly, liver volume and blood flow decrease with age, similarly to immune response. An increased susceptibly to autoimmunity and hepatic infections is another major concern. All these factors may reduce tolerance to treatment [285]. A declination of glomerular filtration rate and a decreased number of functional glomeruli (alongside other anatomical alterations) are typical of aging and senescence [286]. Therefore, physicians must pay attention to vertigo treatment effect on liver and kidney status (Table 2, Table 3 and Table 4), especially if administered with other nephrotoxic or hepatotoxic medications [276,287].
Furthermore, hepatic cirrhosis may degenerate in hepatic encephalopathy [288] and then the use of CNS depressants drugs (see the sections before) should be avoided.
Another important chapter is related to nutraceuticals for the treatment of peripheral vestibular vertigo (e.g., Ginkgo biloba, lemon balm, ginger, citicoline, sage). These substances are not properly recommended in guidelines [22,25,56], but are often used in clinical practice. Nutraceuticals are usually safe compounds, but adverse events and interactions (including CYP450 enzymes) were reported [289,290,291,292,293,294,295,296].
Pharmacogenomics could be used to evaluate the individual response for each treatment. Therefore, clinicians should be aware of this kind of genetic variability to perform specific analysis (genotype) and adequate dosage to the result. However, non-genetic factors may generate a phenomenon called phenoconversion [297], creating a mismatch between genetic polymorphisms and the real capacity to metabolize drugs (Table 9).

4. Medico Legal Aspects: A Multidisciplinary Approach

DDI is a rising phenomenon due to the ageing of the population and the consequent increasing number of patients with comorbidities requiring multiple drug use [323]. For example, multiple drug use (polypharmacy) is common in diabetes mellitus (DM) patients [324]. DDIs are potentially harmful to patients, so full information is required also to improve the safety of care and to avoid malpractice claims. An essential requirement before administering or prescribing any substance is to adequately inform the patient of the expected benefits and any risks of the proposed activity. The Italian Law No. 219—“Provisions for informed consent and advance treatment directives”—specifically defines the issue of information and consent. In observance of the principles referred to in Articles 2, 12, and 32 of the Constitution and Articles 1, 2, and 3 of the Charter of Fundamental Rights of the European Union, the Law No. 2019 of 22 December 2017 not only stipulates that “no medical treatment shall be initiated or continued unless the person concerned has given free and informed consent, except in cases expressly provided for by law”, but also states that “The communication time between the doctor and patient constitutes care time”. Although legislation differs between countries, the promotion of a close care relationship based on informed consent should be widely considered as a goal of treatment, which requires a communicative method appropriate to the patient’s condition and abilities, which can be critical in the elderly population [325]. Informed consent to pharmacological treatment should not be limited to provide a correct information about the risk:benefit ratio of the drug but should be extended on the side effects and their frequency. Clinicians should sensitize patients to alert them if any abnormal symptom appears, in order to modify current polytherapy cares. Extended knowledge of possible adverse drug reactions may improve patient adherence to therapeutic regimen even in case of interactions that may be accepted, if not unknown. By strengthening the relationship between doctor and patient, reaching a real therapeutic alliance, clinicians could guarantee the safety of treatments and reduce possible malpractice claims related to drug–drug interactions. It is necessary, then, to facilitate and promote a constant conversation with the patient [326], as well as careful clinical and laboratory monitoring in order to quickly discover any adverse event. DDIs, as previously discussed, can also determine liver or kidney overload and, therefore, require careful monitoring to quickly detect any drug interaction. The timely identification of pathological effects of drug interactions allows the right therapeutic changes, thus avoiding potential organ damage and subsequent harm to the patient. The benefits of this strategy will not only be economic, allowing to reduce the costs of compensation and civil disputes, but will strengthen the patient’s trust in the doctor, thus enhancing his diagnostic and therapeutic role and making the therapeutic alliance more effective.

5. Conclusions

The management of peripheral vestibular vertigo in elderly requires a deep knowledge of clinical pharmacology and of multidisciplinary issues in the treatment of these patients. Pharmacogenomics are a key point of the therapeutic algorithm. Individual differences in ageing seem to be strictly related to genotype. The evaluation of the different alleles linked to CYP450 isoforms and transporters may be useful to choose the best therapeutic option [102,115,125,159,327]. Clinicians must always remember the important changes related to ageing, involving Vd, renal clearance, hepatic function, and multiple organ impairment. However, individual differences in concomitant treatment, sex, comorbidities, physician decisions, and lifestyle may add great variability to this process [5,254,256]. A good number of therapeutic options is available for peripheral vestibular vertigo. None of the mentioned drugs has shown a superiority in clinical trials; therefore, physician’s challenge consists in the choice of the proper compound [14,22,25,41,236,237,238,245]. The knowledge of DDIs can be used to select treatments for patients, thus optimizing clinical response, minimizing toxicity, and improving the safety of care. DDIs are potentially harmful to patients, so full information is an essential requirement also to enhance a therapeutic alliance and to avoid malpractice claims.
However, we need to underline that most of DDIs described do not show a clinically significant effect because in clinical practice other variables play a role (e.g., inter-individual differences, enzyme availability, concentration of drug into the site of metabolism, affinity of drugs for the enzyme [19,328,329,330,331,332], and activity of genetic and epigenetic factors [333,334]).

Author Contributions

Conceptualization, G.D.M., G.M., P.V. and M.R.; methodology, C.P., L.M., V.R., M.E. and D.P.; formal analysis, R.R., G.S. and A.P. (Amalia Piscopo); investigation, V.C. and N.D.N.; resources, G.D.S.; writing—original draft preparation, G.D.M., G.M., M.R. and A.P. (Alessandro Piras); writing—review and editing, G.C. and L.G.; supervision, G.D.S.; project administration, G.D.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Yoo, H.; Mihaila, D.M. Neuroanatomy, Vestibular Pathways. StatPearls; 2020. Available online: https://www.ncbi.nlm.nih.gov/books/NBK557380/ (accessed on 25 August 2021).
  2. Honrubia, V.; House, M. Mechanism of posterior semicircular canal stimulation in patients with benign paroxysmal positional vertigo. Acta Otolaryngol. 2001, 121, 234–240. [Google Scholar] [CrossRef]
  3. Imai, T.; Takeda, N.; Ikezono, T.; Shigeno, K.; Asai, M.; Watanabe, Y.; Suzuki, M.; Committee for Standards in Diagnosis of Japan Society for Equilibrium Research. Classification, diagnostic criteria and management of benign paroxysmal positional vertigo. Auris Nasus Larynx 2017, 44, 1–6. [Google Scholar] [CrossRef]
  4. Plescia, F.; Salvago, P.; Dispenza, F.; Messina, G.; Cannizzaro, E.; Martines, F. Efficacy and Pharmacological Appropriateness of Cinnarizine and Dimenhydrinate in the Treatment of Vertigo and Related Symptoms. Int. J. Environ. Res. Public Health 2021, 18, 4787. [Google Scholar] [CrossRef]
  5. Klotz, U. Pharmacokinetics and drug metabolism in the elderly. Drug Metab. Rev. 2009, 41, 67–76. [Google Scholar] [CrossRef]
  6. Gutiérrez-Valencia, M.; Izquierdo, M.; Cesari, M.; Casas-Herrero, Á.; Inzitari, M.; Martínez-Velilla, N. The relationship between frailty and polypharmacy in older people: A systematic review. Br. J. Clin. Pharmacol. 2018, 84, 1432–1444. [Google Scholar] [CrossRef] [Green Version]
  7. Raffi, M.; Piras, A.; Persiani, M.; Perazzolo, M.; Squatrito, S. Angle of gaze and optic flow direction modulate body sway. J. Electromyogr. Kinesiol. 2017, 35, 61–68. [Google Scholar] [CrossRef]
  8. Hilmer, S.N.; Gnjidic, D. The effects of polypharmacy in older adults. Clin. Pharmacol. Ther. 2009, 85, 86–88. [Google Scholar] [CrossRef]
  9. Almanza, A.; Navarrete, F.; Vega, R.; Soto, E. Modulation of voltage-gated Ca2+ current in vestibular hair cells by nitric oxide. J. Neurophysiol. 2007, 97, 1188–1195. [Google Scholar] [CrossRef] [Green Version]
  10. Clark, K.C.; Sword, B.A.; Dupree, J.L. Oxidative Stress Induces Disruption of the Axon Initial Segment. ASN Neuro 2017, 9, 1–13. [Google Scholar] [CrossRef] [Green Version]
  11. Kirtane, M.V.; Bhandari, A.; Narang, P.; Santani, R. Cinnarizine: A Contemporary Review. Indian J. Otolaryngol. Head Neck Surg. 2019, 71, 1060–1068. [Google Scholar] [CrossRef]
  12. Haasler, T.; Homann, G.; Duong Dinh, T.A.; Jüngling, E.; Westhofen, M.; Lückhoff, A. Pharmacological modulation of transmitter release by inhibition of pressure-dependent potassium currents in vestibular hair cells. Naunyn. Schmiedebergs. Arch. Pharmacol. 2009, 380, 531–538. [Google Scholar] [CrossRef]
  13. Hausler, R.; Sabani, E.; Rohr, M. L’effet de la Cinnarizine sur divers types de vertiges. Résultatscliniques et électronystagmographiquesd’une étude en double aveugle. Acta Otorhinolaryngol. Belg. 1989, 43, 177–185. [Google Scholar]
  14. Hahn, A.; Novotný, M.; Shotekov, P.M.; Cirek, Z.; Bognar-Steinberg, I.; Baumann, W. Comparison of cinnarizine dimenhydrinate fixed combination with the respective monotherapies for vertigo of various origins: A randomized, double-blind, active-controlled, multicentre study. Clin. Drug Investig. 2011, 31, 371–383. [Google Scholar] [CrossRef]
  15. Scholtz, A.W.; Steindl, R.; Burchardi, N.; Bognar-Steinberg, I.; Baumann, W. Comparison of the therapeutic efficacy of a fixed low-dose combination of cinnarizine and dimenhydrinate with betahistine in vestibular neuritis: A randomized, double-blind, non-inferiority study. Clin. Drug Investig. 2012, 32, 387–399. [Google Scholar] [CrossRef] [PubMed]
  16. Gallelli, L.; Ferreri, G.; Colosimo, M.; Pirritano, D.; Guadagnino, L.; Pelaia, G.; Maselli, R.; De Sarro, G.B. Adversedrug reactions to antibioticsobserved in twopulmonologydivisions of Catanzaro, Italy: A six-yearretrospective study. Pharmacol. Res. 2002, 46, 395–400. [Google Scholar] [CrossRef] [PubMed]
  17. Gallelli, L.; Ferreri, G.; Colosimo, M.; Pirritano, D.; Flocco, M.A.; Pelaia, G.; Maselli, R.; De Sarro, G.B. Retrospectiveanalysis of adversedrug reactions to bronchodilatorsobserved in twopulmonarydivisions of Catanzaro, Italy. Pharmacol. Res. 2003, 47, 493–499. [Google Scholar] [CrossRef]
  18. Gallelli, L.; Colosimo, M.; Pirritano, D.; Ferraro, M.; De Fazio, S.; Marigliano, N.M.; De Sarro, G. Retrospectiveevaluation of adversedrug reactions induced by nonsteroidal anti-inflammatorydrugs. Clin. Drug Investig. 2007, 27, 115–122. [Google Scholar] [CrossRef]
  19. Gallelli, L.; Colosimo, M.; Tolotta, G.A.; Falcone, D.; Luberto, L.; Curto, L.S.; Rende, P.; Mazzei, F.; Marigliano, N.M.; De Sarro, G.; et al. Prospective randomized double-blind trial of racecadotril compared with loperamide in elderly people with gastroenteritis living in nursing homes. Eur. J. Clin. Pharmacol. 2010, 66, 137–144. [Google Scholar] [CrossRef] [Green Version]
  20. Palleria, C.; Di Paolo, A.; Giofrè, C.; Caglioti, C.; Leuzzi, G.; Siniscalchi, A.; De Sarro, G.; Gallelli, L. Pharmacokineticdrug-drug interaction and their implication in clinical management. J. Res. Med. Sci. 2013, 18, 601–610. [Google Scholar]
  21. Staltari, O.; Leporini, C.; Caroleo, B.; Russo, E.; Siniscalchi, A.; De Sarro, G.; Gallelli, L. Drug-drug interactions: Antiretroviraldrugs and recreationaldrugs. Recent Pat. CNS Drug Discov. 2014, 9, 153–163. [Google Scholar] [CrossRef]
  22. Basura, G.J.; Adams, M.E.; Monfared, A.; Schwartz, S.R.; Antonelli, P.J.; Burkard, R.; Bush, M.L.; Bykowski, J.; Colandrea, M.; Derebery, J.; et al. Clinical Practice Guideline: Ménière’s Disease. Otolaryngol.-Head Neck Surg. 2020, 162, S1–S55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Betaistina; Agenzia Italiana del Farmaco: Rome, Italy, 2021.
  24. Ramos Alcocer, R.; Ledezma Rodríguez, J.G.; Navas Romero, A.; Cardenas Nuñez, J.L.; Rodríguez Montoya, V.; Deschamps, J.J.; LiviacTicse, J.A. Use of betahistine in the treatment of peripheralvertigo. Acta Otolaryngol. 2015, 135, 1205–1211. [Google Scholar] [CrossRef] [PubMed]
  25. Bhattacharyya, N.; Gubbels, S.P.; Schwartz, S.R.; Edlow, J.A.; El-Kashlan, H.; Fife, T.; Holmberg, J.M.; Mahoney, K.; Hollingsworth, D.B.; Roberts, R.; et al. Clinical Practice Guideline: Benign Paroxysmal Positional Vertigo (Update). Otolaryngol.-Head Neck Surg. 2017, 156, S1–S47. [Google Scholar] [CrossRef]
  26. Murdin, L.; Hussain, K.; Schilder, A.G.M. Betahistine for symptoms of vertigo. Cochrane Database Syst. Rev. 2016, 2016, 1–55. [Google Scholar]
  27. Adrion, C.; Fischer, C.S.; Wagner, J.; Gürkov, R.; Mansmann, U.; Strupp, M.; BEMED Study Group. Efficacy and safety of betahistine treatment in patients with Meniere’s disease: Primary results of a long term, multicentre, double blind, randomised, placebo controlled, dose defining trial (BEMED trial). BMJ 2016, 352, h6816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Van Esch, B.; Van Der Zaag-Loonen, H.; Bruintjes, T.; Van Benthem, P.P. Betahistine in Ménière’s Disease or Syndrome: A Systematic Review. Audiol. Neurotol. 2021, 1–33. [Google Scholar] [CrossRef]
  29. Tighilet, B.; Léonard, J.; Watabe, I.; Bernard-Demanze, L.; Lacour, M. Betahistine treatment in a cat model of vestibular pathology: Pharmacokinetic and pharmacodynamic approaches. Front. Neurol. 2018, 9, 431. [Google Scholar] [CrossRef]
  30. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Diamox; Agenzia Italiana del Farmaco: Rome, Italy, 2016.
  31. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Idroclorotiazide; Agenzia Italiana del Farmaco: Rome, Italy, 2021.
  32. Vongpatanasin, W. Hydrochlorothiazide is not the most useful nor versatile thiazide diuretic. Curr. Opin. Cardiol. 2015, 30, 361–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. National Institutes of Health Spironolactone-Label. 2020. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=fb66327e-8261-46ae-b39a-4fa79d520844#section-11.2 (accessed on 17 August 2021).
  34. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Aldactone; Agenzia Italiana del Farmaco: Rome, Italy, 2019.
  35. Tu, W.; Decker, B.S.; He, Z.; Erdel, B.L.; Eckert, G.J.; Hellman, R.N.; Murray, M.D.; Oates, J.A.; Pratt, J.H. Triamterene Enhances the Blood Pressure Lowering Effect of Hydrochlorothiazide in Patients with Hypertension. J. Gen. Intern. Med. 2016, 31, 30–36. [Google Scholar] [CrossRef] [Green Version]
  36. National Institutes of Health Dyrenium-Label. 2020. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=ebb177c0-d45d-4443-a85a-e76bd9931a42 (accessed on 20 August 2021).
  37. FDA. Food and Drug Administration Dyazide-Label; Food and Drug Administration: Montgomery County and Prince George’s County, MD, USA, 2009.
  38. van Deelen, G.W.; Huizing, E.H. Use of a diuretic (Dyazide) in the treatment of Menière’s disease. A double-blind cross-over placebo-controlled study. ORL J. Otorhinolaryngol. Relat. Spec. 1986, 48, 287–292. [Google Scholar] [CrossRef]
  39. Nielsen, S. Benzodiazepines. Curr. Top. Behav. Neurosci. 2015, 34, 141–159. [Google Scholar]
  40. Laurence Brunton, R.H.D. Goodman & Gillman’s. The Pharmacological Basis of Therapy; McGraw-Hill Education: New York, NY, USA, 2018. [Google Scholar]
  41. Babu, S.; Schutt, C.A.; Bojrab, D.I. Diagnosis and Treatment of Vestibular Disorders; Springer: Berlin/Heidelberg, Germany, 2019. [Google Scholar] [CrossRef]
  42. Kanegaonkar, R.G.; Tysome, J.R. Dizziness and Vertigo: An Introduction and Pratical Guide; CRC Press: Boca Raton, FL, USA, 2014. [Google Scholar]
  43. AIFA. Agenzia Italiana del Farmaco Riassunto Delle Caratteristiche del Prodotto-Rivotril; Agenzia Italiana del Farmaco: Rome, Italy, 2021.
  44. Altamura, A.C.; Moliterno, D.; Paletta, S.; Maffini, M.; Mauri, M.C.; Bareggi, S. Understanding the pharmacokinetics of anxiolyticdrugs. Expert Opin. Drug Metab. Toxicol. 2013, 9, 423–440. [Google Scholar] [CrossRef]
  45. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Valium; Agenzia Italiana del Farmaco: Rome, Italy, 2021.
  46. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-LorazepamAlmus; Agenzia Italiana del Farmaco: Rome, Italy, 2021.
  47. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Atropina; Agenzia Italiana del Farmaco: Rome, Italy, 2018.
  48. National Institutes of Health Atropine-Label. 2016. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=85225d12-3d96-427a-8027-a4530b8840b0#La57aa34a-5fd5-4d6a-9619-fc7b55a524f1 (accessed on 25 August 2021).
  49. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Glicopirronio; Agenzia Italiana del Farmaco: Rome, Italy, 2020.
  50. Storper, I.S.; Spitzer, J.B.; Scanlan, M. Use of glycopyrrolate in the treatment of Meniere’s disease. Laryngoscope 1998, 108, 1442–1445. [Google Scholar] [CrossRef]
  51. National Institutes of Health Glycopyrrolate Tablet-Label. 2020. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=26cdd84d-ffa0-4e52-b359-4acef4bd8093 (accessed on 30 August 2021).
  52. National Institutes of Health Glycopyrrolate Injection, Solution-Label. 2021. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=88d3e865-8dca-4c89-b6f7-a34c481f75b9 (accessed on 30 August 2021).
  53. Chabicovsky, M.; Winkler, S.; Soeberdt, M.; Kilic, A.; Masur, C.; Abels, C. Pharmacology, toxicology and clinical safety of glycopyrrolate. Toxicol. Appl. Pharmacol. 2019, 370, 154–169. [Google Scholar] [CrossRef]
  54. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Scopolamina Bromidrato; Agenzia Italiana del Farmaco: Rome, Italy, 2016.
  55. Rahko, T.; Karma, P. Transdermal scopolamine for peripheral vertigo (A double-blind study). J. Laryngol. Otol. 1985, 99, 653–656. [Google Scholar] [CrossRef]
  56. Le, T.N.; Westerberg, B.D.; Lea, J. Vestibular neuritis: Recent advances in etiology, diagnostic evaluation, and treatment. Adv. Otorhinolaryngol. 2019, 82, 87–92. [Google Scholar]
  57. AIFA. Agenzia Italiana del Farmaco-Riassunto delle Caratteristiche del Prodotto-Dimenidrinato; Agenzia Italiana del Farmaco: Rome, Italy, 2021.
  58. National Institutes of Health Dimenhydrinate Injection, Solution-Label. 2020. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=bc71539e-1a33-4709-8a24-c2894e8dbc1c (accessed on 2 September 2021).
  59. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Cinnarizina; Agenzia Italiana del Farmaco: Rome, Italy, 2018.
  60. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Arlevertan; Agenzia Italiana del Farmaco: Rome, Italy, 2016.
  61. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Difenidramina; Agenzia Italiana del Farmaco: Rome, Italy, 2017.
  62. Takatani, T.; Ito, J.; Matsuoka, I.; Sasa, M.; Takaori, S. Effects of diphenhydramine iontophoretically applied onto neurons in the medial and lateral vestibular nuclei. Jpn. J. Pharmacol. 1983, 33, 557–561. [Google Scholar] [CrossRef]
  63. National Institutes of Health Diphenhydramine-Label. 2019. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=f3b177ef-55d5-4525-bf33-99205c414a5b (accessed on 3 September 2021).
  64. National Institutes of Health Nighttime Sleep Aid Maximum Strength-Diphenhydramine Hydrochloride 50 mg Capsule, Liquid Filled. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=df1c519f-eb50-4f9a-801a-4baa8a99c5bd (accessed on 3 September 2021).
  65. Huang, W.; Zhang, J.; Wei, P.; Schrader, W.T.; Moore, D.D. Meclizine is an agonist ligand for mouse constitutive androstane receptor (CAR) and an inverse agonist for human CAR. Mol. Endocrinol. 2004, 18, 2402–2408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Fujimoto, C.; Yagi, M.; Murofushi, T. Recent advances in idiopathic bilateral vestibulopathy: A literature review. Orphanet J. Rare Dis. 2019, 14, 202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. National Institutes of Health Meclizine-Label. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=b13cb390-20ed-43ce-aa74-5a754539f6e2 (accessed on 3 September 2021).
  68. National Institutes of Health Meclizine HCL 12.5 MG Tablet-Label. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=76efad7e-e765-430f-9232-63553d02f2c5 (accessed on 3 September 2021).
  69. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche Del Prodotto-Prometazina; Agenzia Italiana del Farmaco: Rome, Italy, 2021.
  70. National Institutes of Health Promethazine-Label. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=51e3baab-eba7-460c-9de1-bf6cfc378310 (accessed on 3 September 2021).
  71. Zabirowicz, E.S.; Gan, T.J. 34—Pharmacology of Postoperative Nausea and Vomiting. Pharmacology and Physiology for Anesthesia; Elsevier Inc.: Amsterdam, The Netherlands, 2019. [Google Scholar] [CrossRef]
  72. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Metoclopramide (Oral); Agenzia Italiana del Farmaco: Rome, Italy, 2021.
  73. Numata, K.; Shiga, T.; Omura, K.; Umibe, A.; Hiraoka, E.; Yamanaka, S.; Azuma, H.; Yamada, Y.; Kobayashi, D. Comparison of acute vertigo diagnosis and treatment practices between otolaryngologists and non-otolaryngologists: A multicenter scenario-based survey. PLoS ONE 2019, 14, e0213196. [Google Scholar] [CrossRef] [PubMed]
  74. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Metoclopramide (Intravenous); Agenzia Italiana del Farmaco: Rome, Italy, 2016.
  75. National Institutes of Health Ondansetron-Label. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=16ed3ca8-c85c-4d06-9c4f-d5bbf344b12e (accessed on 5 September 2021).
  76. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Zofran; Agenzia Italiana del Farmaco: Rome, Italy, 2021.
  77. Van Berkel, M.A.; Elefritz, J.L. Evaluating off-label uses of acetazolamide. Am. J. Health-Syst. Pharm. 2018, 75, 524–531. [Google Scholar] [CrossRef]
  78. Kondrack, R.; Mohiuddin, S. Valsartan/hydrochlorothiazide: Pharmacology and clinical efficacy. Expert Opin. Drug Metab. Toxicol. 2009, 5, 1125–1134. [Google Scholar] [CrossRef]
  79. Mutschler, E.; Gilfrich, H.J.; Knauf, H.; Möhrke, W.; Völger, K.D. Pharmacokinetics of triamterene. Clin. Exp. Hypertens. 1983, 5, 249–269. [Google Scholar] [CrossRef]
  80. AIFA. Agenzia Italiana del Farmaco-Riassunto delle Caratteristiche del Prodotto-Fluss; Agenzia Italiana del Farmaco: Rome, Italy, 2016.
  81. National Institutes of Health-Clonazepam. 2008. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=acbce0e8-5098-4785-943b-8bdb5ff17fab (accessed on 7 September 2021).
  82. National Institutes of Health Diazepam-Injection, Solution. 2020. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=1e6c2f52-73ff-429b-8f1b-9b5b61ce1ac5 (accessed on 7 September 2021).
  83. National Institutes of Health Lorazepam Injection. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=b5b17cde-a94c-4105-871c-54e7d2bd47e8 (accessed on 7 September 2021).
  84. National Institutes of Health Lorazepam Tablet-Label. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=ad2a0633-50fe-4180-b743-c1e49fc110c6 (accessed on 7 September 2021).
  85. Cornelissen, A.S.; Klaassen, S.D.; van Groningen, T.; Bohnert, S.; Joosen, M.J.A. Comparative physiology and efficacy of atropine and scopolamine in sarin nerve agent poisoning. Toxicol. Appl. Pharmacol. 2020, 396, 114994. [Google Scholar] [CrossRef]
  86. EMA. European Medicines Agency Summary of Product Characteristics-Glycopyrronium; European Medicines Agency: Amsterdam, The Netherlands, 2021. [Google Scholar]
  87. NIH. National Institutes of Health Scopolamine Transdermal System-Scolopamine Transdermal System Patch, Extended Release. 2019. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=cb93bfad-3e37-4a27-92f6-f21703ae7bdf (accessed on 5 September 2021).
  88. Kariya, S.; Isozaki, S.; Narimatsu, S.; Suzuki, T. Oxidative metabolism of cinnarizine in rat liver microsomes. Biochem. Pharmacol. 1992, 44, 1471–1474. [Google Scholar] [CrossRef]
  89. Kariya, S.; Isozaki, S.; Uchino, K.; Suzuki, T.; Narimatsu, S. Oxidative metabolism of flunarizine and cinnarizine by microsomes from B-lymphoblastoid cell lines expressing human cytochrome P450 enzymes. Biol. Pharm. Bull. 1996, 19, 1511–1514. [Google Scholar] [CrossRef] [Green Version]
  90. Nguyen, T.; Polyakova, B.; Cerenzio, J.; Ramilo, J.R. Diphenhydramine Use in End-Stage Kidney Disease. Am. J. Ther. 2021, 28, 232–237. [Google Scholar] [CrossRef]
  91. Pham, A.Q.; Scarlino, C. Diphenhydramine and Acute Kidney Injury. Pharm. Ther. 2013, 38, 453–461. [Google Scholar]
  92. Akutsu, T.; Kobayashi, K.; Sakurada, K.; Ikegaya, H.; Furihata, T. Identification of Human Cytochrome P450 Isozymes Involved in Diphenhydramine N -Demethylation. Curr. Drug Metab. 2007, 35, 72–78. [Google Scholar] [CrossRef] [Green Version]
  93. Food and Drug Administration Antivert-Label; Food and Drug Administration: Montgomery County/Prince George’s County, MD, USA, 2019.
  94. Wang, Z.; Lee, B.; Pearce, D.; Qian, S.; Wang, Y. Meclizine Metabolism and Pharmacokinetics: Formulation on Its Absorption. J. Clin. Pharmacol. 2012, 52, 1343–1349. [Google Scholar] [CrossRef]
  95. Taylor, G.; Houston, J.B.; Shaffer, J.; Maweri, G. Pharmacokinetics of promethazine and its sulphoxide metabolite after intravenous and oral administration to man. Br. J. Clin. Pharmacol. 1983, 15, 287–293. [Google Scholar] [CrossRef] [Green Version]
  96. National Institutes of Health Metoclopramide Tablet-Label. 2010. Available online: https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=55f1ee88-a455-44ba-b51a-2fcefe8eda65 (accessed on 8 September 2021).
  97. Shakhatreh, M.; Jehangir, A.; Malik, Z.; Parkman, H.P. Metoclopramide for the treatment of diabetic gastroparesis Metoclopramide for the treatment of diabetic gastroparesis. Expert Rev. Gastroenterol. Hepatol. 2019, 13, 711–721. [Google Scholar] [CrossRef]
  98. Bae, J.W.; Oh, K.Y.; Yoon, S.J.; Shin, H.B.; Jung, E.H.; Cho, C.K.; Lim, C.W.; Kang, P.; Choi, C.I.; Jang, C.G.; et al. Effects of CYP2D6 genetic polymorphism on the pharmacokinetics of metoclopramide. Arch. Pharm. Res. 2020, 43, 1207–1213. [Google Scholar] [CrossRef] [PubMed]
  99. Lemon, L.S.; Zhang, H.; Hebert, M.F.; Hankins, G.D.; Haas, D.M.; Caritis, S.N.; Venkataramanan, R. Ondansetron Exposure Changes in a Pregnant Woman. Pharmacotherapy 2016, 36, e139–e141. [Google Scholar] [CrossRef] [Green Version]
  100. Scott, J.A.; Wood, M.; Flood, P. The Pronociceptive Effect of Ondansetron in the Setting of P-Glycoprotein Inhibition. Anesth. Analg. 2006, 103, 742–746. [Google Scholar] [CrossRef]
  101. Scheen, A.J. Cytochrome P450-mediated cardiovascular drug interactions. Expert Opin. Drug Metab. Toxicol. 2011, 7, 1065–1082. [Google Scholar] [CrossRef] [PubMed]
  102. Siest, G.; Jeannesson, E.; Visvikis-Siest, S. Enzymes and pharmacogenetics of cardiovascular drugs. Clin. Chim. Acta 2007, 381, 26–31. [Google Scholar] [CrossRef]
  103. Akbulut, M.; Urun, Y. Onco-cardiology: Drug-drug interactions of antineoplastic and cardiovascular drugs. Crit. Rev. Oncol. Hematol. 2020, 145, 102822. [Google Scholar] [CrossRef] [PubMed]
  104. Klomp, F.; Wenzel, C.; Drozdzik, M.; Oswald, S. Drug–drug interactions involving intestinal and hepatic CYP1A enzymes. Pharmaceutics 2020, 12, 1201. [Google Scholar] [CrossRef] [PubMed]
  105. Marcianò, G.; Roberti, R.; Palleria, C.; Mirra, D.; Rania, V.; Casarella, A.; De Sarro, G.; Gallelli, L. SARS-CoV-2 Treatment: Current Therapeutic Options and the Pursuit of Tailored Therapy. Appl. Sci. 2021, 11, 7457. [Google Scholar] [CrossRef]
  106. Ahonen, J. Midazolam and Other Benzodiazepines. In Handbook of Experimental Pharmacology; Springer: Berlin/Heidelberg, Germany, 2008; Volume 52, pp. 335–360. [Google Scholar]
  107. El Rouby, N.; Lima, J.J.; Johnson, J.A. Proton pump inhibitors: From CYP2C19 pharmacogenetics to precision medicine. Expert Opin. Drug Metab. Toxicol. 2018, 14, 447–460. [Google Scholar] [CrossRef]
  108. Shirasaka, Y.; Sager, J.E.; Lutz, J.D.; Davis, C.; Isoherranen, N. Inhibition of CYP2C19 and CYP3A4 by Omeprazole Metabolites and Their Contribution to Drug-Drug Interactions s. Drug Metab. Dispos. 2013, 41, 1414–1424. [Google Scholar] [CrossRef] [Green Version]
  109. Food and Drug Administration-Tables. Available online: https://www.fda.gov/drugs/drug-interactions-labeling/drug-development-and-drug-interactions-table-substrates-inhibitors-and-inducers (accessed on 10 September 2021).
  110. Ortiz-Guerrero, G.; Amador-Muñoz, D.; Calderón-Ospina, C.A.; López-Fuentes, D.; Mesa, M.O.N. Proton pump inhibitors and dementia: Physiopathological mechanisms and clinical consequences. Neural Plast. 2018, 2018, 5257285. [Google Scholar] [CrossRef] [Green Version]
  111. Wen, X.; Wang, J.S.; Kivistö, K.T.; Neuvonen, P.J.; Backman, J.T. In Vitro evaluation of valproic acid as an inhibitor of human cytochrome P450 isoforms: Preferential inhibition of cytochrome P450 2C9 (CYP2C9). Br. J. Clin. Pharmacol. Clin. Pharmacol. 2001, 52, 547–553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Schmider, J.; Greenblatt, D.J.; Von Moltke, L.L.; Karsov, D.; Shader, R.I. Inhibition of CYP2C9 by selective serotonin reuptake inhibitors in vitro: Studies of phenytoin p-hydroxylation. Br. J. Clin. Pharmacol. 1997, 44, 495–498. [Google Scholar] [CrossRef] [Green Version]
  113. Krasniqi, V.; Dimovski, A.; Domjanović, I.K.; Bilić, I.; Božina, N. How polymorphisms of the cytochrome P450 genes affect ibuprofen and diclofenac metabolism and toxicity. Arh. Hig. Rada Toksikol. 2016, 67, 1–8. [Google Scholar] [CrossRef] [Green Version]
  114. Macías, Y.; Tabales, J.G.; García-Martín, E.; José, A. An update on the pharmacogenomics of NSAID metabolism and the risk of gastrointestinal bleeding. Expert Opin. Drug Metab. Toxicol. 2020, 16, 319–332. [Google Scholar] [CrossRef]
  115. Wyatt, J.E.; Pettit, W.L.; Harirforoosh, S. Pharmacogenetics of nonsteroidal anti-inflammatory drugs. Pharm. J. 2012, 12, 462–467. [Google Scholar] [CrossRef]
  116. McEvoy, L.; Carr, D.F.; Pirmohamed, M. Pharmacogenomics of NSAID-Induced Upper Gastrointestinal Toxicity. Front. Pharmacol. 2021, 12, 684162. [Google Scholar] [CrossRef]
  117. Zhou, S.-F. Drugs Behave as Substrates, Inhibitors and Inducers of Human Cytochrome P450 3A4. Curr. Drug Metab. 2008, 9, 310–322. [Google Scholar] [CrossRef]
  118. Bahar, M.A.; Setiawan, D.; Hak, E.; Wilffert, B. Pharmacogenetics of drug-drug interaction and drug-drug-gene interaction: A systematic review on CYP2C9, CYP2C19 and CYP2D6. Pharmacogenomics 2017, 18, 701–739. [Google Scholar] [CrossRef]
  119. Schrezenmeier, E.; Dörner, T. Mechanisms of action of hydroxychloroquine and chloroquine: Implications for rheumatology. Nat. Rev. Rheumatol. 2020, 16, 155–166. [Google Scholar] [CrossRef] [PubMed]
  120. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratterisiche del Prodotto-Ciqorin; Agenzia Italiana del Farmaco: Rome, Italy, 2016.
  121. EMA. European Medicines Agency Advagraf-Summary of Product Characteristics; European Medicines Agency: Amsterdam, The Netherlands, 2020. [Google Scholar]
  122. EMA. European Medicines Agency Rapamune-Summary of Product Characteristics; European Medicines Agency: Amsterdam, The Netherlands, 2021. [Google Scholar]
  123. EMA. European Medicines Agency Afinitor-Summary of Product Characteristics; European Medicines Agency: Amsterdam, The Netherlands, 2020. [Google Scholar]
  124. Vanhove, T.; Annaert, P.; Kuypers, D.R.J. Clinical determinants of calcineurin inhibitor disposition: A mechanistic review. Drug Metab. Rev. 2016, 48, 88–112. [Google Scholar] [CrossRef] [PubMed]
  125. Stocco, G.; Lucafò, M.; Decorti, G. Pharmacogenomics of antibiotics. Int. J. Mol. Sci. 2020, 21, 5975. [Google Scholar] [CrossRef]
  126. Zhou, S.; Yung Chan, S.; Cher Goh, B.; Chan, E.; Duan, W.; Huang, M.; McLeod, H.L. Mechanism-based inhibition of cytochrome P450 3A4 by therapeutic drugs. Clin. Pharmacokinet. 2005, 44, 279–304. [Google Scholar] [CrossRef]
  127. Pea, F. Pharmacokinetics and drug metabolism of antibiotics in the elderly. Expert Opin. Drug Metab. Toxicol. 2018, 14, 1087–1100. [Google Scholar] [CrossRef]
  128. Guo, X.; Li, W.; Li, Q.; Chen, Y.; Zhao, G.; Peng, Y.; Zheng, J. Tofacitinib Is a Mechanism-Based Inactivator of Cytochrome P450 3A4. Chem. Res. Toxicol. 2019, 32, 1791–1800. [Google Scholar] [CrossRef]
  129. Mohebbi, N.; Talebi, A.; Moghadamnia, M.; Taloki, Z.N.; Shakiba, A. Drug interactions of psychiatric and COVID-19 medications. Basic Clin. Neurosci. 2020, 11, 185–200. [Google Scholar] [CrossRef]
  130. Plasencia-García, B.O.; Rodríguez-Menéndez, G.; Rico-Rangel, M.I.; Rubio-García, A.; Torelló-Iserte, J.; Crespo-Facorro, B. Drug-drug interactions between COVID-19 treatments and antipsychotics drugs: Integrated evidence from 4 databases and a systematic review. Psychopharmacology 2021, 238, 329–340. [Google Scholar] [CrossRef] [PubMed]
  131. National Institutes of Health Prednisone-Label. Available online: https://dailymed.nlm.nih.gov/dailymed/fda/fdaDrugXsl.cfm?setid=d88845ae-5b7c-465c-982d-8210d37ae190&type=display (accessed on 10 September 2021).
  132. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Prednisone; Agenzia Italiana del Farmaco: Rome, Italy, 2018.
  133. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Soldesam; Agenzia Italiana del Farmaco: Rome, Italy, 2021.
  134. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Metilprednisolone; Agenzia Italiana del Farmaco: Rome, Italy, 2021.
  135. MacLeod, S.M.; Sellers, E.M.; Giles, H.G.; Billings, B.J.; Martin, P.R.; Greenblatt, D.J.; Marshman, J.A. Interaction of disulfiram with benzodiazepines. Clin. Pharmacol. Ther. 1978, 24, 583–589. [Google Scholar] [CrossRef]
  136. Callaghan, R.; Higgins, C.F. Interaction of tamoxifen with the multidrug resistance P-glycoprotein. Br. J. Cancer 1995, 71, 294–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Pauli-Magnus, C.; Rekersbrink, S.; Klotz, U.; Fromm, M.F. Interaction of omeprazole, lansoprazole and pantoprazole with P-glycoprotein. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2001, 364, 551–557. [Google Scholar] [CrossRef]
  138. Vanhove, T.; Remijsen, Q.; Kuypers, D.; Gillard, P. Drug-drug interactions between immunosuppressants and antidiabetic drugs in the treatment of post-transplant diabetes mellitus. Transplant. Rev. 2017, 31, 69–77. [Google Scholar] [CrossRef]
  139. Scheen, A.J. Pharmacokinetics of dipeptidylpeptidase-4 inhibitors. Diabetes Obes. Metab. 2010, 12, 648–658. [Google Scholar] [CrossRef]
  140. Scheen, A.J. Pharmacokinetic interactions with thiazolidinediones. Clin. Pharmacokinet. 2007, 46, 1–12. [Google Scholar] [CrossRef]
  141. EMA. European Medicines Agency Kaletra-Summary of Product Characteristics. 2021. Available online: https://www.ema.europa.eu/en/documents/product-information/kaletra-epar-product-information_en.pdf (accessed on 11 September 2021).
  142. Croxtall, J.D.; Perry, C.M. Lopinavir/Ritonavir: A review of its use in the management of HIV-1 infection. Drugs 2010, 70, 1885–1915. [Google Scholar] [CrossRef] [PubMed]
  143. NIH. Coronavirus Disease 2019 (COVID-19) Treatment Guidelines. NIH 2020, 2019, 130. Available online: https://covid19treatmentguidelines.nih.gov/ (accessed on 11 September 2021).
  144. Liu, X.; Pan, G. Drug Transporters in Drug Disposition, Effects and Toxicity. Advances in Experimental Medicine and Biology; Springer: Berlin/Heidelberg, Germany, 2019; Volume 1141. [Google Scholar]
  145. Kim, R.B. Drugs as P-glycoprotein substrates, inhibitors, and inducers. Drug Metab. Rev. 2002, 34, 47–54. [Google Scholar] [CrossRef] [PubMed]
  146. Saneja, A.; Khare, V.; Alam, N.; Dubey, R.D.; Gupta, P.N. Advances in P-glycoprotein-based approaches for delivering anticancer drugs: Pharmacokinetic perspective and clinical relevance. Expert Opin. Drug Deliv. 2014, 11, 121–138. [Google Scholar] [CrossRef]
  147. EMA. European Medicines Agency Neofordex-Summary of Product Characteristics. 2021. Available online: https://www.ema.europa.eu/en/documents/product-information/neofordex-epar-product-information_en.pdf (accessed on 11 September 2021).
  148. Reiss, W.G.; Oles, K.S. Acetazolamide in the treatment of seizures. Ann. Pharmacother. 1996, 30, 514–519. [Google Scholar] [CrossRef] [PubMed]
  149. Spina, E.; Pisani, F.; Perucca, E. Clinically significant pharmacokinetic drug interactions with carbamazepine. Clin. Pharmacokinet. 1996, 31, 198–214. [Google Scholar] [CrossRef]
  150. Kassamali, R.; Sica, D.A. Acetazolamide: A forgotten diuretic agent. Cardiol. Rev. 2011, 19, 276–278. [Google Scholar] [CrossRef]
  151. Mehta, B.R.; Robinson, B.H.B. Lithium toxicity induced by triamterene-hydrochlorothiazide. Postgrad. Med. J. 1980, 56, 783–784. [Google Scholar] [CrossRef] [PubMed]
  152. Finley, P.R. Drug Interactions with Lithium: An Update. Clin. Pharmacokinet. 2016, 55, 925–941. [Google Scholar] [CrossRef]
  153. Saavedra, I.N.; Aguilera, L.I.; Faure, E.; Galdames, D.G. Phenytoin/clonazepam interaction. Ther. Drug Monit. 1985, 7, 481–484. [Google Scholar] [CrossRef]
  154. Windorfer, A.W., Jr.; Sauer, W. Drug interactions during anticonvulsant therapy in childhood: Diphenylhydantoin, primidone, phenobarbitone, clonazepam, nitrazepam, carbamazepin and dipropylacetate. Neuropadiatrie 1977, 8, 29–41. [Google Scholar]
  155. Blake, K.; Lima, J. Pharmacogenomics of long-acting β2-agonists. Expert Opin. Drug Metab. Toxicol. 2015, 11, 1733–1751. [Google Scholar] [CrossRef]
  156. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Ultibro Breezhaler; Agenzia Italiana del Farmaco: Rome, Italy, 2018.
  157. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Revinty Ellipta; Agenzia Italiana del Farmaco: Rome, Italy, 2018.
  158. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Striverdi Respimat; Agenzia Italiana del Farmaco: Rome, Italy, 2020.
  159. Carr, D.F.; Turner, R.M.; Pirmohamed, M. Pharmacogenomics of anticancer drugs: Personalising the choice and dose to manage drug response. Br. J. Clin. Pharmacol. 2021, 87, 237–255. [Google Scholar] [CrossRef]
  160. Fujita, K. Cytochrome P450 and Anticancer Drugs. Curr. Drug Metab. 2005, 7, 23–37. [Google Scholar] [CrossRef] [PubMed]
  161. Bertholee, D.; Maring, J.G.; van Kuilenburg, A.B.P. Genotypes Affecting the Pharmacokinetics of Anticancer Drugs. Clin. Pharmacokinet. 2017, 56, 317–337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Xeljanz; Agenzia Italiana del Farmaco: Rome, Italy, 2018.
  163. AIFA. Agenzia Italiana del Farmaco Riassunto delle Caratteristiche del Prodotto-Rinvoq; Agenzia Italiana del Farmaco: Rome, Italy, 2019.
  164. Siwek, M.; Woroń, J.; Gorostowicz, A.; Wordliczek, J. Adverse effects of interactions between antipsychotics and medications used in the treatment of cardiovascular disorders. Pharmacol. Rep. 2020, 72, 350–359. [Google Scholar] [CrossRef] [Green Version]
  165. Wyska, E. Pharmacokinetic considerations for current state-of-the-art antidepressants. Expert Opin. Drug Metab. Toxicol. 2019, 15, 831–847. [Google Scholar] [CrossRef]
  166. Trescot, A.M.; Datta, S.; Lee, M.; Hansen, H. Opioid Pharmacology. Pain Physician 2008, 11, 133–153. [Google Scholar] [CrossRef]
  167. Young, W. Review of lithium effects on brain and blood. Cell Transplant. 2009, 18, 951–975. [Google Scholar] [CrossRef] [PubMed]
  168. Bahji, A.; Ermacora, D.; Stephenson, C.; Hawken, E.R.; Vazquez, G. Comparative efficacy and tolerability of pharmacological treatments for the treatment of acute bipolar depression: A systematic review and network meta-analysis. J. Affect. Disord. 2020, 269, 154–184. [Google Scholar] [CrossRef] [PubMed]
  169. Johnson, B.F.; Bustrack, J.A.; Urbach, D.R.; Heyward Hull, J.; Marwaha, R. Effect of metoclopramide on digoxin absorption from tablets and capsules. Clin. Pharmacol. Ther. 1984, 36, 724–730. [Google Scholar] [CrossRef] [PubMed]
  170. Wadhwa, N.K.; Schroeder, T.J.; O’Flaherty, E.; Pesce, A.J.; Myre, S.A.; First, M.R. The effect of oral metoclopramide on the absorption of cyclosporine. Transplantation 1987, 43, 211–213. [Google Scholar] [CrossRef] [PubMed]
  171. Holtzman, C.W.; Wiggins, B.S.; Spinler, S.A. Role of P-glycoprotein in Statin Drug Interactions. Pharmacotherapy 2006, 26, 1601–1607. [Google Scholar] [CrossRef]
  172. Chang, C.; Bahadduri, P.M.; Polli, J.E.; Swaan, P.W.; Ekins, S. Rapid Identification of P-glycoprotein Substrates and Inhibitors. Drug Metab. Dispos. 2006, 34, 1976–1984. [Google Scholar] [CrossRef] [Green Version]
  173. Hedman, A.; Angelin, B.; Arvidsson, A.; Dahlqvist, R. Digoxin-interactions in man: Spironolactone reduces renal but not biliary digoxin clearance. Eur. J. Clin. Pharmacol. 1992, 42, 481–485. [Google Scholar] [CrossRef]
  174. Fenster, P.E.; David Hager, W.; Goodman, M.M. Digoxin-quinidine-spironolactone interaction. Clin. Pharmacol. Ther. 1984, 36, 70–73. [Google Scholar] [CrossRef]
  175. Smellie, W.S.A.; Coleman, J.J. Pitfalls of testing and summary of guidance on safety monitoring with amiodarone and digoxin. Br. Med. J. 2007, 334, 312–315. [Google Scholar] [CrossRef] [Green Version]
  176. Lian, J.; Huang, X.F.; Pai, N.; Deng, C. Chronic betahistine co-treatment reverses olanzapine’s effects on dopamine D2 but not 5-HT2A/2C bindings in rat brains. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2015, 56, 75–80. [Google Scholar] [CrossRef] [Green Version]
  177. Webster, J. Interactions of NSAIDs with Diuretics and β-Blockers: Mechanisms and Clinical Implications. Drugs 1985, 30, 32–41. [Google Scholar] [CrossRef]
  178. Laurent, S. Antihypertensive drugs. Pharmacol. Res. 2017, 124, 116–125. [Google Scholar] [CrossRef] [PubMed]
  179. Trinkley, K.E.; Lee Page, R.; Lien, H.; Yamanouye, K.; Tisdale, J.E. QT interval prolongation and the risk of torsades de pointes: Essentials for clinicians. Curr. Med. Res. Opin. 2013, 29, 1719–1726. [Google Scholar] [CrossRef]
  180. Singh, B.N.; Hollenberg, N.K.; Poole-Wilson, P.A.; Robertson, J.I.S. Diuretic-induced potassium and magnesium deficiency: Relation to drug-induced QT prolongation, cardiac arrhythmias and sudden death. J. Hypertens. 1992, 10, 301–316. [Google Scholar] [CrossRef]
  181. Akylbekova, E.L.; Crow, R.S.; Johnson, W.D.; Buxbaum, S.G.; Njemanze, S.; Fox, E.; Sarpong, D.F.; Taylor, H.A.; Newton-Cheh, C. Clinical correlates and heritability of QT interval duration in blacks: The Jackson Heart Study. Circ. Arrhythmia Electrophysiol. 2009, 2, 427–432. [Google Scholar] [CrossRef] [Green Version]
  182. Scheen, A.J. Type 2 Diabetes and Thiazide Diuretics. Curr. Diab. Rep. 2018, 18, 6. [Google Scholar] [CrossRef]
  183. Humbert, X.; Dolladille, C.; Chrétien, B.; Sassier, M.; Fedrizzi, S.; Puddu, P.E.; Alexandre, J. Thiazides and nonmelanoma skin cancer: Is it a class effect? J. Am. Acad. Dermatol. 2020, 82, e25–e26. [Google Scholar] [CrossRef] [Green Version]
  184. Collins, L.; Quinn, A.; Stasko, T. Skin Cancer and Immunosuppression. Dermatol. Clin. 2019, 37, 83–94. [Google Scholar] [CrossRef]
  185. Scheel, P.J.; Whelton, A.; Rossiter, K.; Watson, A. Cholestyramine-Induced Hyperchloremic Metabolic Acidosis. J. Clin. Pharmacol. 1992, 32, 536–538. [Google Scholar] [CrossRef]
  186. Bostwick, J.R.; Guthrie, S.K.; Ellingrod, V.L. Antipsychotic-Induced Hyperprolactinemia. Pharmacotherapy 2009, 29, 64–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Dehelean, L.; Romosan, A.M.; Papava, I.; Bredicean, C.A.; Dumitrascu, V.; Ursoniu, S.; Romosan, R.S. Prolactin response to antipsychotics: An inpatient study. PLoS ONE 2020, 15, e0228648. [Google Scholar] [CrossRef]
  188. Shimon, I.; Benbassat, C. Male prolactinomas presenting with normal testosterone levels. Pituitary 2014, 17, 246–250. [Google Scholar] [CrossRef]
  189. Li, Y.; Cheng, K.C.; Niu, C.S.; Lo, S.H.; Cheng, J.T.; Niu, H.S. Investigation of triamterene as an inhibitor of the TGR5 receptor: Identification in cells and animals. Drug Des. Devel. Ther. 2017, 11, 1127–1134. [Google Scholar] [CrossRef] [Green Version]
  190. Cheng, K.C.; Chang, W.T.; Kuo, F.Y.; Chen, Z.C.; Li, Y.; Cheng, J.T. TGR5 activation ameliorates hyperglycemia-induced cardiac hypertrophy in H9c2 cells. Sci. Rep. 2019, 9, 3633. [Google Scholar] [CrossRef] [Green Version]
  191. Daly, J.W. Caffeine analogs: Biomedical impact. Cell. Mol. Life Sci. 2007, 64, 2153–2169. [Google Scholar] [CrossRef] [Green Version]
  192. Wein, A.J. Re: A Systematic Review of Amnestic and Non-Amnestic Mild Cognitive Impairment Induced by Anticholinergic, Antihistamine, GABAergic and Opioid Drugs. J. Urol. 2013, 190, 2168. [Google Scholar] [CrossRef]
  193. Pourcher, E.; Bonnet, A.M.; Kefalos, J.; Dubois, B.; Agid, Y. Effects of etybenzatropine and diazepam on levodopa-induced diphasic dyskinesias in Parkinson’s disease. Mov. Disord. 1989, 4, 195–201. [Google Scholar] [CrossRef]
  194. Schelp, S.A.; Brodnik, Z.D.; Rakowski, D.R.; Pultorak, K.J.; Sambells, A.T.; España, R.A.; Oleson, E.B. Diazepam concurrently increases the frequency and decreases the amplitude of transient dopamine release events in the nucleus accumbenss. J. Pharmacol. Exp. Ther. 2018, 364, 145–155. [Google Scholar] [CrossRef]
  195. Klimke, E.K.A. Sudden death after intravenous application of lorazepam in a patient treated with clozapine. Am. J. Psychiatry 1994, 151, 780. [Google Scholar]
  196. Gueta, I.; Markovits, N.; Halkin, H.; Loebstein, R. Concomitant oral potassium chloride and anticholinergic therapy is associated with upper gastrointestinal bleeding: A cohort study. Br. J. Clin. Pharmacol. 2021, 87, 2064–2069. [Google Scholar] [CrossRef]
  197. EMA. European Medicines Agency Sialanar-Summary of Product Characteristics; European Medicines Agency: Amsterdam, The Netherlands, 2016. [Google Scholar]
  198. Valladales-Restrepo, L.F.; Duran-Lengua, M.; Machado-Alba, J.E. Potentially inappropriate prescriptions of anticholinergics drugs in Alzheimer’s disease patients. Geriatr. Gerontol. Int. 2019, 19, 913–917. [Google Scholar] [CrossRef]
  199. Gillman, P.K. Tricyclic antidepressant pharmacology and therapeutic drug interactions updated. Br. J. Pharmacol. 2007, 151, 737–748. [Google Scholar] [CrossRef] [Green Version]
  200. Connolly, B.S.; Lang, A.E. Pharmacological treatment of Parkinson disease: A review. JAMA-J. Am. Med. Assoc. 2014, 311, 1670–1683. [Google Scholar] [CrossRef] [PubMed]
  201. Brocks, D.R. Anticholinergic drugs used in Parkinson’s disease: An overlooked class of drugs from a pharmacokinetic perspective. J. Pharm. Pharm. Sci. 1999, 2, 39–46. [Google Scholar]
  202. Els, C.; Jackson, T.D.; Kunyk, D.; Lappi, V.G.; Sonnenberg, B.; Hagtvedt, R.; Sharma, S.; Kolahdooz, F.; Straube, S. Adverse events associated with medium- and long-term use of opioids for chronic non-cancer pain: An overview of Cochrane Reviews. Cochrane Database Syst. Rev. 2017, 10, CD012509. [Google Scholar]
  203. Valladales-Restrepo, L.F.; Paredes-Mendoza, M.; MacHado-Alba, J.E. Potentially Inappropriate Prescriptions for Anticholinergic Medications for Patients with Constipation. Dig. Dis. 2020, 38, 500–506. [Google Scholar] [CrossRef]
  204. Thakur, A.C.; Aslam, A.K.; Aslam, A.F.; Vasavada, B.C.; Sacchi, T.J.; Khan, I.A. QT interval prolongation in diphenhydraminetoxicity. Int. J. Cardiol. 2005, 98, 341–343. [Google Scholar] [CrossRef]
  205. Isbister, G.K.; Page, C.B. Drug induced QT prolongation: The measurement and assessment of the QT interval in clinical practice. Br. J. Clin. Pharmacol. 2013, 76, 48–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Cheung, B.M.Y.; Wong, Y.L.; Lau, C.P. Queen Mary Utilization of Antihypertensive Drugs Study: Side-effects of antihypertensive drugs. J. Clin. Pharm. Ther. 2005, 30, 391–399. [Google Scholar] [CrossRef]
  207. Freedman, S.B.; Uleryk, E.; Rumantir, M.; Finkelstein, Y. Ondansetron and the risk of cardiac arrhythmias: A systematic review and postmarketing analysis. Ann. Emerg. Med. 2014, 64, 19–25. [Google Scholar] [CrossRef] [PubMed]
  208. Stevens, A.J.; Woodman, R.J.; Owen, H. The effect of ondansetron on the efficacy of postoperative tramadol: A systematic review and meta-Analysis of a drug interaction. Anaesthesia 2015, 70, 209–218. [Google Scholar] [CrossRef] [Green Version]
  209. Nguyen, T.T.; PetzelGimbar, R.M. Sustained Hypotension Following Intravenous Metoclopramide. Ann. Pharmacother. 2013, 47, 1577–1580. [Google Scholar] [CrossRef] [PubMed]
  210. Hwang, J.L.; Weiss, R.E. Steroid-induced diabetes: A clinical and molecular approach to understanding and treatment. Diabetes Metab. Res. Rev. 2014, 30, 96–102. [Google Scholar] [CrossRef] [PubMed]
  211. Narum, S.; Westergren, T.; Klemp, M. Corticosteroids and risk of gastrointestinal bleeding: A systematic review and meta-analysis. BMJ Open 2014, 4, e004587. [Google Scholar] [CrossRef] [Green Version]
  212. Wharton, S.; Raiber, L.; Serodio, K.J.; Lee, J.; Christensen, R.A.G. Medications that cause weight gain and alternatives in Canada: A narrative review. Diabetes Metab. Syndr. Obes. Targets Ther. 2018, 11, 427–438. [Google Scholar] [CrossRef] [Green Version]
  213. Kenna, H.A.; Poon, A.W.; De Los Angeles, C.P.; Koran, L.M. Psychiatric complications of treatment with corticosteroids: Review with case report. Psychiatry Clin. Neurosci. 2011, 65, 549–560. [Google Scholar] [CrossRef]
  214. Renner, U.D.; Oertel, R.; Kirch, W. Pharmacokinetics and pharmacodynamics in clinical use of scopolamine. Ther. Drug Monit. 2005, 27, 655–665. [Google Scholar] [CrossRef]
  215. Hripcsak, G.; Suchard, M.A.; Shea, S.; Chen, R.; You, S.C.; Pratt, N.; Madigan, D.; Krumholz, H.M.; Ryan, P.B.; Schuemie, M.J. Comparison of Cardiovascular and Safety Outcomes of Chlorthalidone vs. Hydrochlorothiazide to Treat Hypertension. JAMA Intern. Med. 2020, 180, 542–551. [Google Scholar] [CrossRef] [PubMed]
  216. Lainscak, M.; Pelliccia, F.; Rosano, G.; Vitale, C.; Schiariti, M.; Greco, C.; Speziale, G.; Gaudio, C. Safetyprofile of mineralocorticoid receptor antagonists: Spironolactone and eplerenone. Int. J. Cardiol. 2015, 200, 25–29. [Google Scholar] [CrossRef]
  217. Titko, T.; Perekhoda, L.; Drapak, I.; Tsapko, Y. Modern trends in diuretics development. Eur. J. Med. Chem. 2020, 208, 112855. [Google Scholar] [CrossRef]
  218. Crowson, M.G.; Patki, A.; Tucci, D.L. A Systematic Review of Diuretics in the Medical Management of Ménière’s Disease. Otolaryngol.-Head Neck Surg. 2016, 154, 824–834. [Google Scholar] [CrossRef]
  219. Espinosa-Sanchez, J.M.; Lopez-Escamez, J.A. The pharmacological management of vertigo in Meniere disease. Expert Opin. Pharmacother. 2020, 21, 1753–1763. [Google Scholar] [CrossRef]
  220. Sepahdari, A.; Vorasubin, N.; Ishiyama, G.; Ishiyama, A. Endolymphatic hydrops reversal following acetazolamide therapy: Demonstration with delayed intravenous contrast-enhanced 3D-FLAIR MRI. Am. J. Neuroradiol. 2016, 37, 151–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. Norell, I.; Stahle, J. Treatment of menière’s disease with hydrochloro-thiazide. Acta Otolaryngol. 1962, 54, 447–456. [Google Scholar] [CrossRef]
  222. Degerman, E.; In ’t Zandt, R.; Pålbrink, A.; Magnusson, M. Endolymphatic hydrops induced by different mechanisms responds differentially to spironolactone: A rationale for understanding the diversity of treatment responses in hydropic inner ear disease. Acta Otolaryngol. 2019, 139, 685–691. [Google Scholar] [CrossRef] [Green Version]
  223. Mori, N.; Miyashita, T.; Inamoto, R.; Matsubara, A.; Mori, T.; Akiyama, K.; Hoshikawa, H. Ion transport its regulation in the endolymphatic sac: Suggestions for clinical aspects of Meniere’s disease. Eur. Arch. Oto-Rhino-Laryngol. 2017, 274, 1813–1820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Amini, A.; Heidari, K.; Asadollahi, S.; Habibi, T.; Shahrami, A.; Mansouri, B.; Kariman, H. Intravenous promethazine versus lorazepam for the treatment of peripheral vertigo in the emergency department: A double blind, randomized clinical trial of efficacy and safety. J. Vestib. Res. 2014, 24, 39–47. [Google Scholar] [CrossRef]
  225. Marill, K.A.; Walsh, M.J.; Nelson, B.K. Intravenous Lorazepam versus Dimenhydrinate for Treatment of Vertigo in the Emergency Department: A Randomized Clinical Trial. Ann. Emerg. Med. 2000, 36, 310–319. [Google Scholar] [CrossRef]
  226. Shih, R.D.; Walsh, B.; Eskin, B.; Allegra, J.; Fiesseler, F.W.; Salo, D.; Silverman, M. Diazepam and Meclizine Are Equally Effective in the Treatment of Vertigo: An Emergency Department Randomized Double-Blind Placebo-Controlled Trial. J. Emerg. Med. 2017, 52, 23–27. [Google Scholar] [CrossRef]
  227. Brandt, T.; Zwergal, A.; Strupp, M. Medical treatment of vestibular disorders. Expert Opin. Pharmacother. 2009, 10, 1537–1548. [Google Scholar] [CrossRef] [PubMed]
  228. Hain, T.C.; Uddin, M. Pharmacological Treatment of Vertigo. CNS Drugs 2003, 17, 85–100. [Google Scholar] [CrossRef] [PubMed]
  229. Heskin-Sweezie, R.; Titley, H.K.; Baizer, J.S.; Broussard, D.M. GABA B receptors contribute to the restoration of balance during vestibular compensation in mice. Neuroscience 2010, 169, 302–314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Shao, M.; Reddaway, R.; Hirsch, J.C.; Peusner, K.D. Presynaptic GABA B Receptors Decrease Neurotransmitter Release in Vestibular Nuclei Neurons during Vestibular Compensation. Neuroscience 2012, 223, 333–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  231. Schneider, G.T.; Lee, C.; Sinha, A.K.; Jordan, P.M.; Holt, J.C. The mammalian efferent vestibular system utilizes cholinergic mechanisms to excite primary vestibular afferents. Sci. Rep. 2021, 11, 1231. [Google Scholar] [CrossRef]
  232. Lee, C.; Jones, T.A. Neuropharmacological targets for drug action in vestibular sensory pathways. J. Audiol. Otol. 2017, 21, 125–132. [Google Scholar] [CrossRef] [Green Version]
  233. Takatani, T.; Ito, J.; Matsuoka, I.; Sasa, M.; Takaori, S. Extrapyramidal symptoms after exposure to calcium channel blocker-flunarizine or cinnarizine. Eur. J. Clin. Pharmacol. 2017, 73, 911–916. [Google Scholar]
  234. Chen, Z.-P.; Zhang, X.Y.; Peng, S.Y.; Yang, Z.Q.; Wang, Y.B.; Zhang, Y.X.; Chen, X.; Wang, J.J.; Zhu, J.N. Histamine H1 receptor contributes to vestibular compensation. J. Neurosci. 2019, 39, 420–433. [Google Scholar] [CrossRef] [Green Version]
  235. Zhou, L.; Zhou, W.; Zhang, S.; Liu, B.; Leng, Y.; Zhou, R.; Kong, W. Changes in Histamine Receptors (H1, H2, and H3) Expression in Rat Medial Vestibular Nucleus and Flocculus after Unilateral Labyrinthectomy: Histamine Receptors in Vestibular Compensation. PLoS ONE 2013, 8, e66684. [Google Scholar]
  236. Motamed, H.; Moezzi, M.; Rooyfard, A.D.; Angali, K.A.; Izadi, Z. A Comparison of the Effects and Side Effects of Oral Betahistine with Injectable Promethazine in the Treatment of Acute Peripheral Vertigo in Emergency. J. Clin. Med. Res. 2017, 9, 994–997. [Google Scholar] [CrossRef] [Green Version]
  237. Scholtz, A.W.; Hahn, A.; Stefflova, B.; Medzhidieva, D.; Ryazantsev, S.V.; Paschinin, A.; Kunelskaya, N.; Schumacher, K.; Weisshaar, G. Efficacy and Safety of a Fixed Combination of Cinnarizine 20 mg and Dimenhydrinate 40 mg vs Betahistine Dihydrochloride 16 mg in Patients with Peripheral Vestibular Vertigo: A Prospective, Multinational, Multicenter, Double-Blind, Randomized, Non-inferior. Clin. Drug Investig. 2019, 39, 1045–1056. [Google Scholar] [CrossRef] [Green Version]
  238. Saberi, A.; Pourshafie, S.H.; Kazemnejad-Leili, E.; Nemati, S.; Sutohian, S.; Sayad-Fathi, S. Ondansetron or promethazine: Which one is better for the treatment of acute peripheral vertigo? Am. J. Otolaryngol.-Head Neck Med. Surg. 2019, 40, 10–15. [Google Scholar] [CrossRef] [PubMed]
  239. Kim, M.B.; Lee, H.S.; Ban, J.H. Vestibular suppressants after canalith repositioning in benign paroxysmal positional vertigo. Laryngoscope 2014, 124, 2400–2403. [Google Scholar] [CrossRef]
  240. Cohen, B.; Dejong, J.M.B.V. Meclizine and Placebo in Treating Vertigo of Vestibular Origin: Relative Efficacy in a Double-Blind Study. Arch. Neurol. 1972, 27, 129–135. [Google Scholar] [CrossRef] [PubMed]
  241. Zhang, K.; Li, Q.; Xu, J.; Liu, J.; Ke, J.; Kang, W.; Li, T.; Ma, F. Unilateral horizontal semicircular canal occlusion in ducesserotonin increase in medial vestibular nuclei: A study using microdialysis in vivo coupled. Analyst 2015, 140, 3846–3851. [Google Scholar] [CrossRef] [PubMed]
  242. Dyhrfjeld-Johnsen, J.; Gaboyard-Niay, S.; Broussy, A.; Saleur, A.; Brugeaud, A.; Chabbert, C. Ondansetron reduces lasting vestibular deficits in a model of severe peripheral excitotoxic injury. J. Vestib. Res. 2013, 23, 177–186. [Google Scholar] [CrossRef]
  243. Jansen, N.L.; Feuerecker, R.; Becker-Bense, S.; Zwergal, A.; Wulff, M.; Xiong, G.; Wängler, B.; Cumming, P.; Bartenstein, P.; Dieterich, M.; et al. Assessment of cerebral dopamine D 2/3 -receptors in patients with bilateral vestibular failure. J. Vestib. Res. 2014, 24, 403–413. [Google Scholar] [CrossRef] [Green Version]
  244. Toro, C.; Trapani, J.G.; Pacentine, I.; Maeda, R.; Sheets, L.; Mo, W.; Nicolson, T. Dopamine Modulates the Activity of Sensory Hair Cells. J. Neurosci. 2015, 35, 16494–16503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Ercin, D.; Erdur, B.; Turkcuer, I.; Seyit, M.; Ozen, M.; Yilmaz, A.; ZincirErcin, D.O. Comparison of efficacy dimenhydrinate and metoclopramide in the treatment of nausea due to vertigo; a randomized study. Am. J. Emerg. Med. 2020, 40, 77–82. [Google Scholar] [CrossRef]
  246. Pelaia, G.; Cuda, G.; Vatrella, A.; Gallelli, L.; Fratto, D.; Gioffrè, V.; D’Agostino, B.; Caputi, M.; Maselli, R.; Rossi, F.; et al. Effects of hydrogen peroxide on MAPK activation, IL-8 production and cell viability in primary cultures of human bronchial epithelial cells. J. Cell. Biochem. 2004, 93, 142–152. [Google Scholar] [CrossRef]
  247. Ramamoorthy, S.; Cidlowski, J.A. Corticosteroids—Mechanisms of action in health and disease. Rheum. Dis. Clin. N. Am. 2016, 42, 15–31. [Google Scholar] [CrossRef] [Green Version]
  248. Fisher, L.M.; Derebery, M.J.; Friedman, R.A. Oral Steroid Treatment for Hearing Improvement in Meniere’s Disease and Endolymphatic Hydrops. Otol. Neurotol. 2012, 33, 1685–1691. [Google Scholar] [CrossRef]
  249. Fishman, J.M.; Burgess, C.; Waddell, A. Corticosteroids for the treatment of idiopathic acute vestibular dysfunction (vestibular neuritis). Cochrane Database Syst. Rev. 2011, 1–30. [Google Scholar] [CrossRef] [PubMed]
  250. Ismail, E.I.; Morgan, A.E.; Rahman, A.M.A. Corticosteroids versus vestibular rehabilitation in long-term outcomes in vestibular neuritis. J. Vestib. Res. 2018, 28, 417–424. [Google Scholar] [CrossRef] [Green Version]
  251. Haid, T. Evaluation of flunarizine in patients with Menière’s disease. Subjective and vestibular findings. Acta Oto-Laryngol. Suppl. 1988, 460, 149–153. [Google Scholar] [CrossRef]
  252. Fraysse, B.; Bebear, J.P.; Dubreuil, C.; Berges, C.; Dauman, R. Betahistinedihydrochloride versus flunarizine. A double-blind study on recurrent vertigo with or without cochlear syndrome typical of Menière’s disease. Acta Oto-Laryngol. 1991, 490, 1–10. [Google Scholar] [CrossRef]
  253. Chiarella, G.; Marcianò, G.; Viola, P.; Palleria, C.; Pisani, D.; Rania, V.; Casarella, A.; Astorina, A.; Scarpa, A.; Esposito, M.; et al. Nutraceuticals for Peripheral Vestibular Pathology: Properties, Usefulness, Future Perspectives and Medico-Legal Aspects. Nutrients 2021, 13, 3646. [Google Scholar] [CrossRef] [PubMed]
  254. Scott, I.A.; Hilmer, S.N.; Reeve, E.; Potter, K.; Le Couteur, D.; Rigby, D.; Gnjidic, D.; Del Mar, C.B.; Roughead, E.E.; Page, A.; et al. Reducing inappropriate polypharmacy: The process of deprescribing. JAMA Intern. Med. 2015, 175, 827–834. [Google Scholar] [CrossRef] [Green Version]
  255. Mortazavi, S.S.; Shati, M.; Keshtkar, A.; Malakouti, S.K.; Bazargan, M.; Assari, S. Defining polypharmacy in the elderly: A systematic review protocol. BMJ Open 2016, 6, e010989. [Google Scholar] [CrossRef] [PubMed]
  256. Schwartz, J.B. The Influence of Sex on Pharmacokinetics. Clin. Pharmacokinet. 2003, 42, 107–121. [Google Scholar] [CrossRef]
  257. Le Couteur, D.G.; Anderson, R.M.; de Cabo, R. Sex and Aging. J. Gerontol.-Ser. A Biol. Sci. Med. Sci. 2018, 73, 139–140. [Google Scholar] [CrossRef]
  258. Fischer, K.E.; Riddle, N.C. Sex differences in aging: Genomic instability. J. Gerontol.-Ser. A Biol. Sci. Med. Sci. 2018, 73, 166–174. [Google Scholar] [CrossRef]
  259. Spreafico, R.; Soriaga, L.B.; Grosse, J.; Virgin, H.W.; Telenti, A. Advances in genomics for drug development. Genes 2020, 11, 942. [Google Scholar] [CrossRef]
  260. Sirota, M.; Dudley, J.T.; Kim, J.; Chiang, A.P.; Morgan, A.A.; Sweet-Cordero, A.; Sage, J.; Butte, A.J. Discovery and preclinical validation of drug indications using compendia of public gene expression data. Sci. Transl. Med. 2011, 3, 96ra77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  261. Gordon, E.H.; Peel, N.M.; Samanta, M.; Theou, O.; Howlett, S.E.; Hubbard, R.E. Sex differences in frailty: A systematic review and meta-analysis. Exp. Gerontol. 2017, 89, 30–40. [Google Scholar] [CrossRef] [PubMed]
  262. Verhamme, K.M.C.; Sturkenboom, M.C.J.M.; Stricker, B.H.C.; Bosch, R. Drug-induced urinary retention: Incidence, management and prevention. Drug Saf. 2008, 31, 373–388. [Google Scholar] [CrossRef]
  263. Sakakibara, R.; Uchiyama, T.; Yamanishi, T.; Kishi, M. Dementia and lower urinary dysfunction: With a reference to anticholinergic use in elderly population. Int. J. Urol. 2008, 15, 778–788. [Google Scholar] [CrossRef]
  264. DiSogra, R.M.; Meece, J. Auditory and Vestibular Side Effects of FDA-Approved Drugs for Diabetes. Semin. Hear. 2019, 40, 315–326. [Google Scholar] [CrossRef] [PubMed]
  265. Vinik, A.I.; Maser, R.E.; Mitchell, B.D.; Freeman, R. Diabetic Autonomic Neuropathy. Diabetes Care 2003, 26, 1553–1579. [Google Scholar] [CrossRef] [Green Version]
  266. Tsai, M.-S.; Lee, L.A.; Tsai, Y.T.; Yang, Y.H.; Liu, C.Y.; Lin, M.H.; Hsu, C.M.; Chen, C.K.; Li, H.Y. Sleep Apnea and Risk of Vertigo: A Nationwide Population-Based Cohort Study. Laryngoscope 2018, 128, 763–768. [Google Scholar] [CrossRef]
  267. Besnard, S.; Tighilet, B.; Chabbert, C.; Hitier, M.; Toulouse, J.; Le Gall, A.; Machado, M.L.; Smith, P.F. The Balance of Sleep: Role of the Vestibular Sensory System. Sleep Med. Rev. 2018, 42, 220–228. [Google Scholar] [CrossRef]
  268. Gao, T.; Zhang, Q.; Hou, J.; Zhu, K.; Sun, B.; Chen, J.; Xia, C.; Ren, X.; Cheng, Y.; Wang, Z. Vestibular-evoked myogenic potentials in patients with severe obstructive sleep apnea. J. Int. Med. Res. 2020, 48, 0300060520909717. [Google Scholar] [CrossRef] [PubMed]
  269. Nakayama, M.; Masuda, A.; Ando, K.B.; Arima, S.; Kabaya, K. A Pilot Study on the Efficacy of Continuous Positive Airway Pressure on the Manifestations of Ménière’s Disease in Patients with Concomitant Obstructive Sleep Apnea Syndrome. J. Clin. Sleep Med. 2015, 11, 1101–1107. [Google Scholar] [CrossRef] [Green Version]
  270. Guy Edwards, J. Adverse Effects of Antianxiety Drugs. Drugs 1981, 22, 495–514. [Google Scholar] [CrossRef]
  271. Wang, S.; Chen, W.; Tang, S.; Lin, H. Benzodiazepines Associated With Acute Respiratory Failure in Patients With Obstructive Sleep Apnea. Front. Pharmacol. 2019, 9, 1513. [Google Scholar] [CrossRef] [PubMed]
  272. Landier, W. Ototoxicity and cancer therapy. Cancer 2016, 122, 1647–1658. [Google Scholar] [CrossRef] [Green Version]
  273. Cacho-Díaz, B.; Salmerón-Moreno, K.; Mendoza-Olivas, L.G.; Reynoso-Noverón, N.; Gómez-Amador, J.L. Vertigo in patients with cancer: Red flag symptoms. J. Clin. Neurosci. 2019, 69, 175–178. [Google Scholar] [CrossRef]
  274. Collamati, A.; Martone, A.M.; Poscia, A.; Brandi, V.; Celi, M.; Marzetti, E.; Cherubini, A.; Landi, F. Anticholinergic drugs and negative outcomes in the olderp opulation: From biological plausibility to clinical evidence. Aging Clin. Exp. Res. 2016, 28, 25–35. [Google Scholar] [CrossRef] [PubMed]
  275. Heppner, H.J.; Cornel, S.; Peter, W.; Philipp, B.; Katrin, S. Infections in the Elderly. Crit. Care Clin. 2013, 29, 757–774. [Google Scholar] [CrossRef] [PubMed]
  276. Altissimi, G.; Colizza, A.; Cianfrone, G.; de Vincentiis, M.; Greco, A.; Taurone, S.; Musacchio, A.; Ciofalo, A.; Turchetta, R.; Angeletti, D.; et al. Drugs inducing hearing loss, tinnitus, dizziness and vertigo: An updated guide. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 7946–7952. [Google Scholar]
  277. Kim, S.Y.; Kim, H.J.; Min, C.; Choi, H.G. Association between benign paroxysmal positional vertigo and osteoporosis: Two nested case-control studies. Osteoporos. Int. 2020, 31, 2017–2024. [Google Scholar] [CrossRef]
  278. Lane, N.E. Glucocorticoid-Induced Osteoporosis: New Insights into the Pathophysiology and Treatments. Curr. Osteoporos. Rep. 2019, 17, 1–7. [Google Scholar] [CrossRef] [PubMed]
  279. Reid, I.R.; Mills, D.A.J.; Wattie, D.J. Ototoxicity associated with intravenous bisphosphonate administration. Calcif. Tissue Int. 1995, 56, 584–585. [Google Scholar] [CrossRef]
  280. Balatsouras, D.G.; Koukoutsis, G.; Fassolis, A.; Moukos, A.; Apris, A. Benign paroxysmal positional vertigo in the elderly: Current insights. Clin. Interv. Aging 2018, 13, 2251–2266. [Google Scholar] [CrossRef] [Green Version]
  281. Vivino, F.B. Sjogren’s syndrome: Clinical aspects. Clin. Immunol. 2017, 182, 48–54. [Google Scholar] [CrossRef] [PubMed]
  282. Kalus, A. Rheumatologic Skin Disease. Med. Clin. N. Am. 2015, 99, 1287–1303. [Google Scholar] [CrossRef] [PubMed]
  283. Rendon, A.; Schäkel, K. Psoriasis pathogenesis and treatment. Int. J. Mol. Sci. 2019, 20, 1475. [Google Scholar] [CrossRef] [Green Version]
  284. Bach, M.H.; Simkin, P.A. Uricosuric drugs: The once and future therapy for hyperuricemia? Curr. Opin. Rheumatol. 2014, 26, 169–175. [Google Scholar] [CrossRef] [PubMed]
  285. Tajiri, K.; Shimizu, Y. Liver physiology and liver diseases in the elderly. World J. Gastroenterol. 2013, 19, 8459–8467. [Google Scholar] [CrossRef] [PubMed]
  286. Denic, A.; Glassock, R.J.; Rule, A.D. Structural and Functional Changes with the Aging Kidney. Adv. Chronic Kidney Dis. 2016, 23, 19–28. [Google Scholar] [CrossRef] [Green Version]
  287. Björnsson, E.S. Hepatotoxicity by drugs: The most common implicated agents. Int. J. Mol. Sci. 2016, 17, 224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  288. Weissenborn, K. Hepatic Encephalopathy: Definition, Clinical Grading and Diagnostic Principles. Drugs 2019, 79, 5–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  289. Aronson, J.K. Defining ‘nutraceuticals’: Neither nutritious nor pharmaceutical. Br. J. Clin. Pharmacol. 2017, 83, 8–19. [Google Scholar] [CrossRef]
  290. Martines, F.; Salvago, P.; Dispenza, F.; Rizzo, S.; Letizia Mauro, G.; Puglisi, S. Treatment with a new nutraceutical compound on patients suffering from balance disorders: Dizziness handicap inventory scores. Acta Med. Mediterr. 2019, 35, 2029–2034. [Google Scholar]
  291. Russo, R.; Gallelli, L.; Cannataro, R.; Perri, M.; Calignano, A.; Citraro, R.; Russo, E.; Gareri, P.; Corsonello, A.; De Sarro, G. WhenNutraceuticalsReinforceDrugs Side Effects: A Case Report. Curr. Drug Saf. 2016, 11, 264–266. [Google Scholar] [CrossRef] [PubMed]
  292. Committee on Herbal Medicinal Products (HMPC). European Medicines Agency-Assessment report on Ginkgo biloba L., folium, EMA/HMPC/321095/2012. Agency Eur. Med. 2014, 44, 116. [Google Scholar]
  293. Shakeri, A.; Sahebkar, A.; Javadi, B. Melissa officinalis L.—A review of its traditional uses, phytochemistry and pharmacology. J. Ethnopharmacol. 2016, 188, 204–228. [Google Scholar] [CrossRef]
  294. Ghorbani, A.; Esmaeilizadeh, M. Pharmacological properties of Salvia officinalis and its components. J. Tradit. Complementary Med. 2017, 7, 433–440. [Google Scholar] [CrossRef]
  295. European Food Standards Agency-Scientific Opinion on the safety of “citicoline” as a Novel Food ingredient. EFSA J. 2013, 11, 1–22.
  296. Grøntved, A.; Hentzer, E. Vertigo-reducing effect of ginger root. A controlled clinical study. ORL J. Otorhinolaryngol. Relat. Spec. 1986, 48, 282–286. [Google Scholar] [CrossRef]
  297. Klomp, S.D.; Manson, M.L.; Guchelaar, H.J.; Swen, J.J. Phenoconversion of cytochrome p450 metabolism: A systematic review. J. Clin. Med. 2020, 9, 2890. [Google Scholar] [CrossRef]
  298. Korpi, E.R.; Sinkkonen, S.T. GABAA receptor subtypes as targets for neuropsychiatric drug development. Pharmacol. Ther. 2006, 109, 12–32. [Google Scholar] [CrossRef] [PubMed]
  299. Tóth, K.; Csukly, G.; Sirok, D.; Belic, A.; Kiss, Á.; Háfra, E.; Déri, M.; Menus, Á.; Bitter, I.; Monostory, K. Optimization of clonazepam therapy adjusted to patient’s CYP3A status and NAT2 genotype. Int. J. Neuropsychopharmacol. 2016, 19, pyw083. [Google Scholar] [CrossRef] [Green Version]
  300. Ho, T.T.; Gupta, S.V.; Sanchez-Valle, A. Prolonged clonazepam-induced withdrawal symptoms in an NAT2 ultraslow acetylator. Pharmacogenomics 2019, 20, 69–73. [Google Scholar] [CrossRef]
  301. Dean, L. Diazepam Therapy and CYP2C19 Genotype. In Medical Genetics Summaries; National Center for Biotechnology Information: Bethesda, MD, USA, 2012. [Google Scholar]
  302. Tiwari, A.K.; Souza, R.P.; Müller, D.J. Pharmacogenetics of anxiolytic drugs. J. Neural Transm. 2009, 116, 667–677. [Google Scholar] [CrossRef]
  303. Fukasawa, T.; Suzuki, A.; Otani, K. Effects of genetic polymorphism of cytochrome P450 enzymes on the pharmacokinetics of benzodiazepines. J. Clin. Pharm. Ther. 2007, 32, 333–341. [Google Scholar] [CrossRef] [PubMed]
  304. Guttman, Y.; Nudel, A.; Kerem, Z. Polymorphism in cytochrome P450 3A4 is ethnicity related. Front. Genet. 2019, 10, 224. [Google Scholar] [CrossRef]
  305. Saiz-Rodríguez, M.; Almenara, S.; Navares-Gómez, M.; Ochoa, D.; Román, M.; Zubiaur, P.; Koller, D.; Santos, M.; Mejía, G.; Borobia, A.M.; et al. Effect of the most relevant CYP3A4 and CYP3A5 polymorphisms on the pharmacokinetic parameters of 10 CYP3A substrates. Biomedicines 2020, 8, 94. [Google Scholar] [CrossRef] [PubMed]
  306. Li, J.; Chen, W.; Peng, C.; Zhu, W.; Liu, Z.; Zhang, W.; Su, J.; Li, J.; Chen, X. Human H1 receptor (HRH1) gene polymorphism is associated with the severity of side effects after desloratadine treatment in Chinese patients with chronic spontaneous uticaria. Pharm. J. 2020, 20, 87–93. [Google Scholar] [CrossRef]
  307. Chu, J.T. Histamine H1 receptor gene polymorphism acts as a biological indicator of the prediction of therapeutic efficacy in patients with allergic rhinitis in the Chinese Han population. J. Cell. Biochem. 2019, 120, 164–170. [Google Scholar] [CrossRef] [Green Version]
  308. Gu, J.; Mao, X.H.; Yang, X.Z.; Ao, H.F.; Zhang, Z.; Li, Y. Histamine H4 receptor gene polymorphisms: A potential predictor of oral H1 antihistamine efficacy for allergic rhinitis. Int. Forum Allergy Rhinol. 2017, 7, 268–275. [Google Scholar] [CrossRef] [PubMed]
  309. Yan, S.; Chen, W.; Wen, S.; Zhu, W.; Guo, A.; Chen, X.; Zhang, C.; Chen, M.; Zhang, J.; Su, J.; et al. Influence of component 5a receptor 1 (C5AR1) -1330T/G polymorphism on nonsedating H1-antihistamines therapy in Chinese patients with chronic spontaneous urticaria. J. Dermatol. Sci. 2014, 76, 240–245. [Google Scholar] [CrossRef]
  310. Zanger, U.M.; Raimundo, S.; Eichelbaum, M. Cytochrome P450 2D6: Overview and update on pharmacology, genetics, biochemistry. Naunyn. Schmiedebergs. Arch. Pharmacol. 2004, 369, 23–37. [Google Scholar] [CrossRef] [PubMed]
  311. Zhang, L.; Brown, S.J.; Shan, Y.; Lee, A.M.; Allen, J.D.; Eum, S.; de Leon, J.; Bishop, J.R. CYP2D6 Genetic Polymorphisms and Risperidone Pharmacokinetics: A Systematic Review and Meta-analysis. Pharmacotherapy 2020, 40, 632–647. [Google Scholar] [CrossRef]
  312. de Leon, J.; Nikoloff, D.M. Paradoxical excitation on diphenhydramine may be associated with being a CYP2D6 ultrarapid metabolizer: Three case reports. CNS Spectr. 2008, 13, 133–135. [Google Scholar] [CrossRef]
  313. Michel, M.C.; Teitsma, C.A. Polymorphisms in human muscarinic receptor subtype genes. Handb. Exp. Pharmacol. 2012, 208, 49–59. [Google Scholar]
  314. Koonrungsesomboon, N.; Khatsri, R.; Wongchompoo, P.; Teekachunhatean, S. The impact of genetic polymorphisms on CYP1A2 activity in humans: A systematic review and meta-analysis. Pharm. J. 2018, 18, 760–768. [Google Scholar] [CrossRef]
  315. Parkman, H.P.; Mishra, A.; Jacobs, M.; Pathikonda, M.; Sachdeva, P.; Gaughan, J.; Krynetskiy, E. Clinical response and side effects of metoclopramide: Associations with clinical, demographic, and pharmacogenetic parameters. J. Clin. Gastroenterol. 2012, 46, 494–503. [Google Scholar] [CrossRef]
  316. Bell, G.C.; Caudle, K.E.; Whirl-Carrillo, M.; Gordon, R.J.; Hikino, K.; Prows, C.A.; Gaedigk, A.; Agundez, J.; Sadhasivam, S.; Klein, T.E.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for CYP2D6 genotype and use of ondansetron and tropisetron. Clin. Pharmacol. Ther. 2017, 102, 213–218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  317. PharmVar. CYP1A2 Allele Nomenclature. Available online: https://www.pharmvar.org/gene/CYP1A2 (accessed on 21 November 2021).
  318. Tornio, A.; Backman, J.T. Cytochrome P450 in Pharmacogenetics: An Update. In Advances in Pharmacology; Elsevier Inc.: Amsterdam, The Netherlands, 2018; Volume 83. [Google Scholar]
  319. Ruan, C.J.; De Leon, J. Is there a future for CYP1A2 pharmacogenetics in the optimal dosing of clozapine? Pharmacogenomics 2020, 21, 369–373. [Google Scholar] [CrossRef]
  320. Tzvetkov, M.V.; Saadatmand, A.R.; Bokelmann, K.; Meineke, I.; Kaiser, R.; Brockmöller, J. Effects of OCT1 polymorphisms on the cellular uptake, plasma concentrations and efficacy of the 5-HT 3 antagonists tropisetron and ondansetron. Pharm. J. 2012, 12, 22–29. [Google Scholar]
  321. He, H.; Yin, J.Y.; Xu, Y.J.; Li, X.; Zhang, Y.; Liu, Z.G.; Zhou, F.; Zhai, M.; Li, Y.; Li, X.P.; et al. Association of ABCB1 polymorphisms with the efficacy of ondansetron in chemotherapy-induced nausea and vomiting. Clin. Ther. 2014, 36, 1242–1252. [Google Scholar] [CrossRef] [PubMed]
  322. Tremblay, P.B.; Kaiser, R.; Sezer, O.; Rosler, N.; Schelenz, C.; Possinger, K.; Roots, I.; Brockmoller, J. Variations in the 5-hydroxytryptamine type 3B receptor gene as predictors of the efficacy of antiemetic treatment in cancer patients. J. Clin. Oncol. 2003, 21, 2147–2155. [Google Scholar] [CrossRef]
  323. Rashid, K.; Khan, Y.; Ansar, F.; Waheed, A.; Aizaz, M. Potential Drug-Drug Interactions in Hospitalized Medical Patients: Data from Low Resource Settings. Cureus 2021, 13, e17336. [Google Scholar] [CrossRef] [PubMed]
  324. İnci, H. Evaluation of multiple drug use in patients with type 2 diabetes mellitus. Diabetol. Int. 2021, 12, 399–404. [Google Scholar] [CrossRef]
  325. Bolcato, M.; Sanavio, M.; Fassina, G.; Rodriguez, D.; Aprile, A. Healthcare professionals and patient information: A fresh look from the recent Italian law on consent. Clin. Ter. 2021, 172, 104–108. [Google Scholar]
  326. Truglio-Londrigan, M.; Slyer, J.T.; Singleton, J.K.; Worral, P.S. A qualitative systematic review of internal and external influences on shared decision-making in all health care settings. JBI Database Syst. Rev. Implement. Rep. 2012, 10, 4633–4646. [Google Scholar] [CrossRef]
  327. Melzer, D.; Pilling, L.C.; Ferrucci, L. The genetics of human ageing. Nat. Rev. Genet. 2020, 21, 88–101. [Google Scholar] [CrossRef] [PubMed]
  328. Bachmann, K.; Ghosh, R. The use of in vitro methods to predict in vivo pharmacokinetics and drug interactions. Curr. Drug Metab. 2001, 2, 299–314. [Google Scholar] [CrossRef] [PubMed]
  329. Venkatakrishnan, K.; von Moltke, L.L.; Obach, R.S.; Greenblatt, D.J. Drug metabolism and drug interactions: Application and clinical value of in vitro models. Curr. Drug Metab. 2003, 4, 423–459. [Google Scholar] [CrossRef]
  330. De Fazio, S.; Gallelli, L.; De Siena, A.; De Sarro, G.; Scordo, M.G. Role of CYP3A5 in abnormal clearance of methadone. Ann. Pharmacother. 2008, 42, 893–897. [Google Scholar] [CrossRef] [PubMed]
  331. Gallelli, L.; Galasso, O.; Urzino, A.; Saccà, S.; Falcone, D.; Palleria, C.; Longo, P.; Corigliano, A.; Terracciano, R.; Savino, R.; et al. Characteristics and clinical implications of the pharmacokinetic profile of ibuprofen in patients with knee osteoarthritis. Clin. Drug Investig. 2012, 32, 827–833. [Google Scholar] [CrossRef]
  332. Pelaia, G.; Vatrella, A.; Gallelli, L.; Cazzola, M.; Maselli, R.; Marsico, S.A. Potential genetic influences on the response to asthma treatment. Pulm. Pharmacol. Ther. 2004, 17, 253–261. [Google Scholar] [CrossRef]
  333. Gallelli, L.; Siniscalchi, A.; Carotenuto, M.; Caroleo, M.C.; Cione, E.; Guidetti, V. microRNAs-based Predictor Factor in Patients with Migraine-ischemic Stroke. MicroRNA 2017, 6, 17–21. [Google Scholar] [CrossRef]
  334. Gallelli, L.; Siniscalchi, A.; Palleria, C.; Mumoli, L.; Staltari, O.; Squillace, A.; Maida, F.; Russo, E.; Gratteri, S.; De Sarro, G.; et al. Adverse drug reactions related to drug administration in hospitalized patients. Curr. Drug Saf. 2017, 12, 171–177. [Google Scholar] [CrossRef]
Table 1. Rationale, current clinical indications, and dosages of peripheral vestibular vertigo drugs.
Table 1. Rationale, current clinical indications, and dosages of peripheral vestibular vertigo drugs.
Mechanism(s) of ActionCurrent Clinical IndicationsDosage SuggestedRoute of Administration
BetahistineStrong antagonist of histamine H3 receptors and a weak agonist of H1 receptors [22,23]MD and other causes of peripheral vestibular vertigo [22,24,25]24/48 mg daily [23]OS [23]
Diuretics
AcetazolamideInhibition of carbonic anhydrase [30]MD [22]250–375 mg daily [30]OS [30]
HydrochlorothiazideInhibition of sodium chloride co-transporter in the distal convoluted tubule [31,32]MD [22]12.5/25 mg daily (hypertension) [31]OS [31]
SpironolactoneMineralocorticoid receptor antagonist [33,34,35]MD [22]25–100 mg daily (hypertension) [33,34]OS, IV [33,34]
TriamtereneInhibition of ENaC [35]MD [22]50/100 mg daily [36], but may vary depending on formulation/coformulation [37,38]OS [36]
Benzodiazepines
ClonazepamAllosteric modulation of GABAA receptor [39,40]All forms of PVV [41,42]1.5–20 mg/die, generally 3–6 mg/die (OS) [43,44]OS [40]
DiazepamAllosteric modulation of GABAA receptor [39,40]All forms of PVV [41,42]4–60 mg/daily (OS)
10–60 mg/daily (IV, IM) [45]
OS, IV, IM, rectal [40]
LorazepamAllosteric modulation of GABAA receptor [39,40]All forms of PVV [41,42]2–10 mg/daily [44,46]OS, IM, IV [40]
Anticholinergics
AtropineNon-selective muscarinic blocker [47]All forms of PVV [22,25,41]0.3–4 mg (depending on clinical indication) [47,48]IV, IM, SC [47,48]
GlycopyrrolateNon-selective muscarinic blocker [49]All forms of PVV [22,25,41]2 mg in clinical trial [50], but may vary depending on clinical indication
1–8 mg (OS) [51]
Various (parenteral) [49,52]
IV, OS, IM [49,51,53]
ScopolamineNon-selective muscarinic blocker [54]All forms of PVV [22,25,41]0,25–1 mg daily (IM, IV) [54].
0.5 mg (TD) in clinical trial [55]
IM, IV, TD [54,55]
Antihistamines
Dimenhydrinate + cinnarizine D: antagonist of H1 receptor [4]
C:It blocks voltage-gated calcium channels, preventing calcium translocations across
the vestibular air cells and, thus, regulating hair cell afferent vestibular transmission, anti-vasoconstrictor activity, reduction in the blood viscosity of the inner ear’s circulatory system [4]
All forms of PVV [22,25,41,56]Dimenhydrinate:
25–200 mg (OS) [57]
Basis of 50 mg (IV-IM), but may vary [58]
Cinnarizine: 15–225 mg (OS) [13,59].
Recommended clinical dose (vestibular disorders): varies between 25 mg thrice-daily and 75 mg once-daily, up to a maximum of 225 mg [13]
Dimenhydrinate/
cinnarizine
(co-formulation): 20/40 mg thrice a day [60]
IV, IM, OS (d) [57,58]
OS (c) [59]
OS (co-formulation) [60]
DiphenhydramineAntagonist of H1 receptor [61,62]All forms of PPV [22,25,41,56]25 mg-50 mg [61,63,64]OS [61,63]
MeclizineAntagonist of H1 receptor [65]All forms of PVV [22,25,41,56,66]12.5–25 mg [67,68]OS [67]
PromethazineAntagonist of H1 receptor [69,70]All forms of PVV [22,25,41,56]25–100 mg (OS)
25–50 mg; max: 100 mg (IM, IV) [69]
May vary [71]
OS, IM, IV [69]
Other antiemetics
MetoclopramideIt acts on 5HT4 (agonist), 5HT3 (antagonist) and dopamine D2 (antagonist) receptors [40,71,72]All forms of PVV [41,73]10 mg–30 mg or max 0.5 mg/kg (IV-IM-OS) [72,74]OS, IM, IV, rectal [72,74]
Ondansetron5HT3 antagonist [75,76]All forms of PVV [22,25,41,56]4–8 mg capsules (OS), multiple administration also
8 mg (IV-IM)
16 mg (rectal) [76]
OS, IM IV, rectal [76]
ENaC, epithelial sodium channel; GABA, gamma aminobutyric acid; IM, intramuscular; IV, intravenous; MD, Meniere’s disease; OS, oral; PVV, peripheral vestibular vertigo; SC, subcutaneous; TD, transdermal.
Table 2. Pharmacokinetics of drugs used in peripheral vestibular vertigo.
Table 2. Pharmacokinetics of drugs used in peripheral vestibular vertigo.
Oral BioavailabilityTime to Peak ConcentrationSerum Half-Life (t1/2)Protein BindingTransporter ProteinsMetabolismMetabolites
BHSNA1 h3.5 h5%-Monoamine oxidases (MAO) A/B2-pyridylacetic acid (2-PAA)
Diuretics
AAZNA2 h8 h90–95%---
HCT70%4 h6–25 h40–70%-Mainly unmodified2-Amino-4-chlorobenzenesulfonamide
SPL~65%2–4 h * (including metabolites)1.5 h *90% *-First pass metabolismCanrenone, 6-ß-hydroxy-7-α-(thiomethyl) spironolactone, 7-α-(thiomethyl) spironolactone (TMS),
TRI52%3 h2–4 h67%-LiverHydroxytriamterene
Benzodiazepines
CLZ90%1.2 h23 ± 5 h82–86%-Liver (glucuronidation), CYP3A47-aminoclonazepam and 7-acetamido-clonazepam
DZP90–100%0.5–1.5 h24–48 h96–98%-Liver (glucuronidation), CYP3A4, CYP2C19Desmethyldiazepam, oxazepam, temazepam
LOR90%2–3 h12–16 h85–90%-Liver (glucuronidation)3-O-phenolic glucuronide
Anticholinergic drugs
ATP-10 min (IM)4 h--50% liver 50% unmodifiedNA
GLY3% (children) NA, but higher (adults)NA0.83 ± 0.27 h (IV) 75 min (IM) 2.5–4 h (OS, solution)--NANA
SCONA2 min (IM) 24 h (TD)8 h--HepaticNA
* Protein binding is about 98% for canrenone; canrenone half-life = 9–24 h; time to peak concentration of spironolactone alone = 2.6 h. AAZ; acetazolamide; ATP, atropine; BHS, betahistine; CLZ, clonazepam; CYP, cytochromes P450; DZP, diazepam; GLY, glycopyrrolate; HCT, hydro-chlorothiazide; IM, intramuscular; IV, intravenous; LOR, lorazepam; MAO, monoamine oxidase; NA, not available; OS, oral; P-gp, p-glycoprotein; SCO, scopolamine; SPL, spironolactone; TD, transdermal; TRI, triamterene.
Table 3. Pharmacokinetics of antihistamines and other antiemetics.
Table 3. Pharmacokinetics of antihistamines and other antiemetics.
Oral BioavailabilityTime to Peak ConcentrationSerum Half-Life (t1/2)Protein BindingTransporter ProteinsMetabolismMetabolites
Antihistamines
(H1 Antagonists)
DIM + CNZ43–72% (d)1–4 h (d)
2–4 h (c)
6–7 h (d)
4–5 h (c)
80–85% (d)-Hepatic (d, see the section below).

CYP2D6 and CYP2B6, but other CYP may be involved (c)
D: Diphenhydramine, DMDP
C: Conjugated with glucuronic acid
DPH43–72%1–4 h3–9.3 h80–85%-Hepatic first-pass metabolism CYP2D6, and to a minor extent CYP1A2, CYP2C9 and CYP2C19DMDP
MECNA1.5–6 h5.21 ± 0.80 hNA-Hepatic CYP2D6Norchlorcyclizine (rats), 10 different metabolites in human urines. Human metabolites have not been identified, but meclizine undergoes aromatic hydroxylation or benzylic oxidation.
PMZ25%2–3 h4–6 h (OS) 9–16 (IV) 6–13 (IM)--Hepatic first-pass metabolismPromethazine sulfoxide (PMZSO), N-demethylpromethazine
Other antiemetics
MCP35–100%0.5–2 h (OS)
3 h (IM)
5–6 h13–40%-Hepatic: CYP2D6 isoform, and possibly CYP1A2 (minor role), CYP2C19and CYP3A; conjugation.Argikar et al. identified 10 metabolites of metoclopramide (M1-M10) in the urine after oral administration. Of those (M1, M2, M6, M7, and M8) were conjugated to either glucuronide or sulfate. Mono-de-ethyl-metoclopramide and N-4 sulphate conjugated are two important products.
OND56% (OS)
60% (rectal)
1.5 h (OS)
6 h (rectal)
3–6 h70–76%P-gp substrateHepatic first-pass metabolism, CYP1A2, CYP2D6, CYP3A4Hydroxylation on the indole ring followed by subsequent glucuronide or sulphate conjugation.
CNZ, cinnarizine; CYP, cytochromes P450; DIM, dimenhydrinate; DMDP, monodesmethyldi-phenhydramine; DPH, diphenhydramine; IM, intramuscular; IV, intravenous; MCP, metoclopramide; MEC, meclizine; NA, not available; OND, ondansetron; OS, oral; P-gp, p-glycoprotein; PMZ, promethazine; P-gp, p-glycoprotein; TD, transdermal.
Table 4. Dose changes, elimination, and inductor/inhibitor activity of peripheral vestibular vertigo drugs.
Table 4. Dose changes, elimination, and inductor/inhibitor activity of peripheral vestibular vertigo drugs.
Enzymes Inductor/InhibitorEliminationDose Changes in Hepatic DiseaseDose Changes in Renal DiseaseReferences
BHS-85% urine
Low levels in bile
No dosage adjustment seems to be neededNo dosage adjustment seems to be needed[23,29]
Diuretics
AAZ-100% urineContraindicated in hepatic impairmentDosage adjustment needed. Contraindicated in patients with IR but may be used in cardiopathic patients without severe renal impairment[30,77]
HCT-60–80% urine
24% feces
Caution needed in patients with severe hepatic impairmentThe compound is not effective in patients with eGFR <30 mL/min
Do not administer in patients with severe renal impairment
[31,78]
SPL-Mainly in urine, secondary in bileCaution needed in patients with severe hepatic impairmentAvoid in acute kidney injury, anuria or eGFR <30 mL/min. Pay attention in case of renal impairment[33,34,40]
TRI-The majority is expelled in urine, a minor quote in bileCaution needed in patients with impaired hepatic function. In patients with severe hepatic impairment, triamterene’s levels may increase. In NIH data bank it is contraindicates in severe hepatic impairmentCaution needed. Periodic BUN and serum potassium determinations should be made to check kidney function, especially in patients with suspected or confirmed renal insufficiency[36,37,40,79,80]
Benzodiazepines
CLZ-50–70% in urine
10–30% feces
Protein binding may be changed by cirrhosis, increasing the free fraction. Caution needed. Contraindicated in severe hepatic impairmentCaution needed[40,43,44,81]
DZP-100% urineContraindicated in severe hepatic impairment.
Caution needed in other mild and moderate hepatic impairment
Caution needed[40,44,45,82]
LOR-88 ± 4% urine
7 ± 2% feces.
Caution needed. Contraindicated in severe hepatic impairmentCaution needed in severe hepatic impairment[44,46,83,84]
Anticholinergic drugs
ATP-50% liver50% urine
Caution neededCaution needed[47,48,85]
GLY-Urine, only 5% bileFurther studies needed. Since kidney elimination has a major role, hepatic impairment seems not to be relevant, despite a certain negative effect of anticholinergic drugs on hepatic damageDose reduction by 30% in patients with mild to moderate renal impairment. Contraindicated in severe renal impairment[49,52,86]
SCO-UrineCaution needed for the risk of CNS reactionsCaution needed for the risk of CNS reactions[54,85,87]
AHs (H1-antagonists)
DIM + CNZInhibition of CYP2D6 (d)Mainly in urine (d)
40–60% feces and minor quote in urines (c)
Caution needed (d)
Coadministration contraindicated in patients with severe hepatic impairment
Caution needed (d)
Coadministration contraindicated in patients with eGFR < 25 mL/min
[57,60,61,71,88,89]
DPHIt inhibits CYP2D6Mainly in urineCaution neededCaution needed[61,71,90,91,92]
MECMeclizine seems to reduce the expression of CYP2B10, 3A11, 1A2 in experimental modelsUrine, fecesCaution needed (need further evaluation)Caution needed (need further evaluation)[65,67,93,94]
PMZ-UrineCaution needed (need further evaluation)Caution needed (need further evaluation)[69,70,71,95]
Other antiemetics
MCP-86% urine, minor quote in bileCaution neededCaution needed
In patients with last stage renal impairment (eGFR ≤ 15 mL/min): Dose reduction of 75%.
Severe/moderate renal impairment (eGFR 15–60 mL/min): Dose reduction of 50%
[71,72,74,96,97,98]
OND-Majority hepatic, 5% urineCaution needed, especially in severe hepatic impairmentCaution needed, although studies on moderate renal impairment did not show significant changes[40,75,76,99,100]
AAZ, acetazolamide; ACDs, anticholinergic drugs; AHs, antihistamines; ALT, Alanine Aminotransferase; ATP, atropine; BDZ, benzodiazepines; BHS, betahistine; BUN, Blood Urea Nitrogen; CLZ, clonazepam; CNS, central nervous system; CNZ, cinnarizine; CYP, cytochromes P450; DIM, dimenhydrinate; DPH, diphenhydramine; DZP, diazepam; eGFR, estimated glomerular filtration rate; GLY, glycopyrrolate; HCT, hydrochlorothiazide; LOR, lorazepam; MCP, metoclopramide; MEC, meclizine; NIH, National Institutes of Health; NA, not available; OND, ondansetron; P-gp, P-glycoprotein; SCO, scopolamine; PMZ, promethazine; SPL, spironolactone; TRI, triamterene.
Table 5. Peripheral vestibular vertigo drugs pharmacokinetic interactions.
Table 5. Peripheral vestibular vertigo drugs pharmacokinetic interactions.
Effect of the CombinationMechanism of InteractionSelection of Drugs AffectedClinical Comment
BetahistineSerum level ↑ by MAOIs [23]Inhibition of MAOIs-Decrease the dosage
CYP1A2 substratesSerum level may be slightly increased by amiodarone, ciprofloxacin, fluvoxamine, fluoxetine, antifungals, duloxetine, isoniazid [101,102,103,104]Inhibition of CYP1A2Diphenhydramine (minor extent) [92]Reduce the dosage
Serum level slightly decreased by tocilizumab, rifampicin, antiseizure drugs, smoke [104,105]Induction of CYP1A2
CYP2C19 substratesSerum level ↑
by inhibitors:
voriconazole, fluconazole, omeprazole, esomeprazole, fluvoxamine,
fluoxetine, cimetidine, modafinil, felbamate, armodafinil, ticlopidine [45,106,107,108,109]
Inhibition of CYP2C19Diazepam, diphenhydramine (minor extent) [45,92]Reduce the dosage (of 5 mg)
Omeprazole may increase diazepam levels, associated to dementia risk. PPI inhibitors are also related to dementia [110]
Serum level ↓ by
inducers: rifampicin, enzalutamide, efavirenz, ritonavir, tocilizumab [45,105,106,109]
Induction of CYP2C19 Evaluate dose increase
CYP2C9 substratesSerum level ↑ by amiodarone, some antifungals, fluoxetine, fluvoxamine, valproic acid, disulfiram, fluvastatin [101,109,111,112]CYP2C9 inhibitionDiphenhydramine (minor extent) [92]Low grade of interaction
Serum level ↓ by tocilizumab, antiseizure drugs, rifampicin, enzalutamide, ritonavir, aprepitant [105,109]CYP2C9 induction
CYP2D6 substratesSerum level ↑ by amiodarone, abiraterone, other anticancer drugs, bupropion, celecoxib, HCQ, labetalol, vemurafenib some antipsychotics, some antidepressants [101,105,109,113,114,115,116,117,118,119]CYP2D6 inhibitionDiphenhydramine, meclizine, metoclopramide [92,94,97]Dose decrease
Meclizine (from 25 mg to 12.5 mg)
CYP3A4 substratesSerum level ↑
by inhibitors: some antifungals
omeprazole, esomeprazole, fluvoxamine, cimetidine, isoniazid, cimetidine, idelasilib, antidepressants, calcium- antagonists, amiodarone, erythromycin, crizotinib, imatinib other anticancer drugs, calcineurin inhibitors, mTOR inhibitors, clarithromycin, erythromycin and troleandomycin, remdesivir, tofacitinib, some antidepressants [43,45,101,102,103,105,106,107,108,109,117,118,120,121,122,123,124,125,126,127,128,129,130]
Inhibition of CYP3A4
Diazepam, clonazepam [43,45],
corticosteroids [131,132,133,134]
Dose decrease (Diazepam of 5 mg, Clonazepam, and corticosteroids of one half)
Antidepressants don’t affect clonazepam levels [43]
Serum level ↓ by
inducers: modafinil, armodafinil, carbamazepine and other anti-seizure drugs, rifampicin, tocilizumab [45,105,106]
Induction of CYP3A4 Evaluate dose increase
DiazepamSerum level ↑ by disulfiram [45]Inhibition of oxidative metabolism [135]-Evaluate dose reduction
Serum level ↑ by ciprofloxacin [106]CYP1A2 inhibition (even if diazepam is not metabolized by this isoform) or unknown mechanism of reduced clearance-Evaluate dose reduction
Serum levels ↓ by food, antiacids, narcotics [45]Reduction of diazepam absorption-Dose increase related to patient condition. Food and antiacids reduce concentration peak, but not AUC
Serum levels ↑ by prokinetics [45] -Evaluate dose reduction
LorazepamSerum levels ↑ by valproate and probenecid Probenecid decreases lorazepam
clearance, reducing the production of lorazepam-glucuronide. Similar mechanism for valproate [46,106]
-Dose decrease
P-gp substratesSerum level ↑ by CQ, HCQ, LPV/r, canagliflozin, lansoprazole, tamoxifen [136], some antipsychotics, some calcium antagonists, calcineurin inhibitors, lovastatin
[101,103,105,109,117,119,120,121,122,123,124,129,130,133,137,138,139,140,141,142,143,144,145,146]
P-gp inhibitionOndansetron, dexamethasone [100,147]Ondansetron may have a pronociceptive effect with P-gp inhibitors [100], therefore evaluate a dose decrease, even if ondansetron is used for few days (maximum of 5) [76]
Dexamethasone or P-gp inhibitor reduction may be needed
Serum level ↓ by dexamethasone [147]P-gp inductionOndansetron, dexamethasone [100,147]Risk for reduced efficacy
AUC, area under the curve; CQ, chloroquine; CYP, cytochrome P450; HCQ, hydroxychloroquine; LPV/r, lopinavir/ritonavir; MAOIs, monoamineoxidaseinhibitors; mTOR, mechanistic target of rapamycin; P-gp, p-glycoprotein.
Table 6. Peripheral vestibular vertigo drugs pharmacokinetic interactions (perpetrators).
Table 6. Peripheral vestibular vertigo drugs pharmacokinetic interactions (perpetrators).
Effect of the CombinationMechanism of InteractionSelection of Drugs AffectedClinical Comment
AAZ↑ Phenytoin and carbamazepine concentration.
↓ Primidone levels
It acts on the cited anti-seizure drugs absorption or has an unknown mechanism [30,148,149]-Risk for increased or decreased efficacy
↑ Tricyclic antidepressants and amphetamine levels.
↓ Lithium and aspirin levels
It facilitates/lasts the elimination of these substances [30,150]-Risk for adverse events or decreased efficacy
↑ Lithium levelsIt reduces lithium urinary excretion [31,151,152]-Risk of increased lithium side effects (see the spironolactone section)
CLZVariation of phenytoin levels [43]Unknown. Probably clonazepam stimulate inhibition of enzymatic system in some patients, and induction in others [153]-Risk for adverse events or decreased efficacy
↑ Primidone levels [43,154]Unknown pharmacokinetic mechanism [154]-Risk for adverse events
CYP3A4 substratesDexamethasone ↓ substrates levels [147]Induction of CYP3A4Gliquidone, glyburide, repaglinide, pioglitazone, saxagliptin, warfarin, antiplatelet drugs, some antiarrhythmics, calcium antagonists, direct oral anticoagulants, some statins, some NSAIDs, some opioids, some β2-agonists, PPI (minor extent), anticancer drugs (e.g., cyclophosphamide, docetaxel, etoposide, gefitinib), calcineurin and mTOR inhibitors, macrolides (e.g., clarithromycin and telithromycin), remdesivir, HCQ, some JAK inhibitors, some antipsychotics, some antidepressants [101,102,103,105,107,108,113,114,115,116,117,119,120,121,122,123,124,125,126,127,128,138,139,140,155,156,157,158,159,160,161,162,163,164,165]CYP3A4 induction accelerates the production of antiplatelets active metabolites [101,102,103]

Pravastatin is not a relevant substrate of CYP450 and then may be a safer choice [101]
The interaction may accelerate the production of metabolites or delay opioids’ clearance [166]
CYP2D6 substratesSerum level ↑by diphenhydramine and dimenhydrinateCYP2D6 inhibitionBeta-blockers (e.g., metoprolol, bisoprolol, carvedilol), calcium antagonists,
antidepressants, antipsychotics, codeine, hydrocodone, antiarrhythmic drugs (often class I), formoterol, anticancer drugs(e.g., tamoxifen, gefitinib), remdesivir, HCQ, upadacitinib, some antipsychotics, some antidepressants [101,102,103,105,119,155,159,160,161,163,164,165,166]
Risk of adverse events

Interactions
may stop the production of metabolites or opioids’ clearance [166]
DPHBarbiturates, sulfacetamide, sodium salicylateIt may reduce the absorption of these drugs [61]-Dosage increase may be needed
HCT↑ lithium levels [31,151,152]Reduction of lithium clearance-Risk for ADRs. It is important to maintain lithium in the optimal range [167], therefore clinician should evaluate dose reduction for spironolactone or therapeutic switch. However, lithium itself may be switched to use other mood stabilizers [168]
MCPDigoxinMetoclopramide decrease digoxin absorption acting on gut motility [72,169]-Decrease of metoclopramide dosage, monitoring digoxin plasmatic range
CiclosporinIncrease of ciclosporin absorption. Metoclopramide hastens gastric emptying and facilitates small intestine absorption [170]-Dosage reduction of metoclopramide/
ciclosporin may be needed
Mivacurium, succinylcholine It may reduce their clearance blocking plasmatic cholinesterase [72]-Dosage reduction of metoclopramide may be needed
P-gp substratesDexamethasone ↓ substrates levels [147]P-gp induction by dexamethasoneDPP-4 inhibitors, SGLT-2 inhibitors, DOAC, verapamil, aliskiren, digoxin, amiodarone, atorvastatin and simvastatin, some β2-agonists, some anticancer drugs, calcineurin and mTOR inhibitors, Fluoroquinolones, daptomycin, linezolid, baricitinib, dexamethasone, remdesivir, upadacitinib (in vitro) [101,102,103,105,117,120,121,122,123,124,125,126,127,138,139,140,144,145,146,155,156,157,158,163,171,172]Increase dosage of P-gp substrates/decrease dexamethasone depending on clinical control of hypertension, diabetes, or other pathologies and on temporal duration of corticosteroid treatment
SPL↑ Lithium levelsReduction of lithium clearance-Risk for adverse events. It is important to maintain lithium in the optimal range [167], therefore clinician should evaluate dose reduction for spironolactone or therapeutic switch. However, lithium itself may be switched to use other mood stabilizers [168]
↑ Digoxin levelReduction of digoxin clearance [173,174]-Risk for adverse events. Evaluate dose reduction to maintain digoxin in its therapeutic range [175]
TRI↑ Lithium levels may increaseReduction of lithium clearance-Risk for increased adverse events (see the spironolactone section)
AAZ, acetazolamide; CLZ, clonazepam; CYP, cytochrome P450; DOAC, direct-acting oral anticoagulants; DPH, diphenhydramine; DPP-4, dipeptidyl peptidase-4; HCQ, hydroxychloroquine; JAK, Janus kinase; mTOR, mechanistic target of rapamycin; MCP, metoclopramide; NSAID, Nonsteroidal anti-inflammatory drugs; P-gp, p-glycoprotein; PPI, proton pump inhibitors; SGLT-2, sodium glucose co-transporter-2; SPL, spironolactone; TRI, triamterene.
Table 7. Pharmacodynamic drug interactions.
Table 7. Pharmacodynamic drug interactions.
Drug(s) InvolvedMechanism of InteractionClinical Comment
BHSAntihistaminesBetahistine acts as an histamine analogue, with possible interactions with antihistamines as a consequence [23]Risk for reduced efficacy
OlanzapineCoadministration with olanzapine showed a reduction of olanzapine binding to D2RThis may prevent adverse events (weight gain) related to olanzapine, but also “dopaminergic supersensitivity” due to high stimulation on D2R. Despite this evidence, betahistine may also affect drugs effect on D2R [176]
Diuretics
Class effectsNSAIDsVarious mechanisms including NSAID related sodium and water retention, suppression of plasma renin activity, alterations in
adrenoceptor sensitivity and impaired synthesis of vasodilator prostaglandins. Nephrotoxic effect and inhibition of natriuretic response to diuretic should be considered
[31,36,40,80,177]
Risk for reduced efficacy of diuretics and kidney injury
Other diureticsDiuretic therapy combined with other antihypertensive drugs may lower blood pressure [40,41,178]In case of multidrug therapy in a patient with MD and hypertension, the combination of two or more drugs acting on blood pressure should be evaluated carefully [34,36,150]. Clinicians may evaluate adding or replacing of one or more drugs used for hypertension
Drugs prolonging QT, proarrhythmic drugsHypocalcemia, hypokalemia, and hypomagnesemia may also prolong QT interval [179], so diuretic therapy (associated with electrolytic disturbances) may worsen this side effect [180,181]Caution needed. Switching to a drug which doesn’t alter QT, may be a good choice
HCTDrugs which lower potassium (e.g., amphotericin B, corticosteroids, diuretics, laxatives, digitalis) and sodium levels (e.g., desmopressin, antidepressants, diuretics, carbamazepine)The thiazide diuretic lowers potassium and sodium concentration [31]Risk of hypokalemia and hyponatremia
Antiarrhythmic drugs (e.g., amiodarone), antipsychotics (e.g., chlorpromazine) antimalarial drugs (e.g., chinidine)Hypokalemia may facilitate torsade de points [31]Risk of torsade de points
Antidiabetics or drugs raising glucose levelsIncrease of glucose levels [31,182]Risk of hyperglycemia or hypoglycemia
Calcineurin inhibitors, corticosteroids, mycophenolate, azathioprine and (lower risk) mTOR inhibitorsImmunosuppression and kidney transplant have a certain association with non-melanoma skin cancer: Thiazides, used in the management of MD, are associated with a certain skin cancer risk [183]mTOR inhibitors (sirolimus and everolimus) have a lower risk, partially reverting oncogenic effects [184]. Avoid the combination is the optimal choice. Sun protection has an important role
SPLDrugs generating hyperkaliemia (e.g., trimethoprim, ARB, ACE inhibitors, other potassium sparing diuretics, heparin, NSAIDs, ciclosporin, tacrolimus)Spironolactone may increase potassium levels [33,34]Risk for hyperkaliemia
CholestyramineCholestyramine increases the urinary excretion of bicarbonate and spironolactone favors acidosis with potassium reabsorption and reduction of urine acidity [185]Risk of hyperkalemic metabolic acidosis
Antipsychotics and metoclopramideHyperprolactinemia is a possible side effect of antipsychotics, related to their inhibition on D2R. This condition causes several clinical manifestations including menstrual disturbances, sexual dysfunction, galactorrhea, gynecomastia, infertility [186]. Spironolactone may cause gynecomastia or sexual disturbances [34].
Metoclopramide is also associated to hyperprolactinemia and, moreover, to depression [97]
Hyperprolactinemia related to antipsychotics seems to be more frequent in females and to be mainly connected with some specific compounds (risperidone, haloperidol, amisulpride) [187]. Spironolactone should be avoided in male patients in treatment with prolactin raising antipsychotics or prolactinoma [188]
TRISkeletal muscle relaxants (non-depolarizing) and preanesthetic/anesthetic agentsTriamterene enhances the effect of the cited drugs [36]Risk for adverse events
ACE inhibitors, angiotensin receptor blockers (ARB) or potassium containing medicationTriamterene causes the raise of potassium levels Risk of hyperkaliemia [36]
Antidiabetic drugs, or drugs increasing glucose levelsIt may also raise blood glucose levels [36,37,80].
The reason for this mechanism seems to rely in triamterene’s inhibition of G-protein-coupled bile acid receptor 1 (GPBAR1, also known as TGR5) [189,190]
Risk of hyperglycemia or hypoglycemia
BDZ
Class effectsXanthines contrast BDZ anxiolytic effect [45]Xanthines may give anxiety [191]Reduced clinical effect
CNS depressants (opioids, antipsychotics, antidepressants, hypnotics, antiseizure drugs, anesthetics, antihistamines)When associated with other central nervous system (CNS) depressants BDZ effect may be enhanced [43,45,46,192]Risk benefit-management is needed
Aminophylline and theophylline can reverse at least partially the sedation from
BDZ
This interaction appears to be due to
the blockade of adenosine receptors by aminophylline [106]
Risk for reduced efficacy
DZPDopamine levels down when administered with L-DOPA [45,193]Benzodiazepines influence dopamine release [194]Difficult management of Parkinson’s disease. Coadministration of antipsychotics in this clinical setting may worsen clinical status, increasing extrapyramidal side effects [193]
LORClozapine: concomitant use of lorazepam may produce marked sedation, excessive salivation, hypotension, ataxia, delirium, and respiratory arrest [84,195]UnknownEvaluate the real necessity of this combination, switching eventually to other compounds
ACDs
Class effectsKCLAnticholinergics reduce gastric transit and KCL has an irritating effect [47,196]Risk of gastric bleeding
Other drugs with anticholinergic activity (e.g., antipsychotics, amantadine, tricyclic antidepressants, nefopam, MAO inhibitors, anti-Parkinson drugs) may worsen side effectsConcomitant action on ACh pathway [47,49,54]Increased adverse events
AChE inhibitors, cholinergic drugsConcomitant administration of AChE inhibitors and anticholinergics must be avoided in patients with QT prolongation [47,49,54].
The concomitant consumption of cholinergic drugs is another contraindication to the use of anticholinergics [54]
Risk of arrythmias, pharmacodynamic antagonism
Antihypertensive/hypertensive drugsAnticholinergic drugs may increase (or lower) blood pressure and raise heart rate [54,197]Evaluate the real utility of coadministration and monitor patient
PPIChronic use of PPI may be associated with dementia, and this risk should be added to that generated using [110,198]Avoid chronic treatment with PPI and ACD
Tricyclic antidepressants These drugs act also on histaminergic and cholinergic receptors with antagonist effect [40,199] and for this reason they may enforce side effects related to antihistamines and anticholinergicsAvoid coadministration
AntipsychoticsSome anticholinergics drugs are used in the treatment of Parkinson’s disease, whereas other are associated with parkinsonism. Antipsychotics generate extrapyramidal effects [54,193,200,201]Avoid coadministration
Opioids Opioids and anticholinergics both lead to constipation [202,203]Avoid coadministration
Antihistamines
Class effectsAnticholinergicsIt has anticholinergic effects so it must be avoided in BPH, glaucoma, myasthenia gravis, bradycardia, gastrointestinal paralysis, or obstructions [57,61,67,69]Avoid coadministration
CNS suppressant drugs (e.g., MAOIs) [57,61,67,69].Sedation is a common adverse event of antihistamines [192]Avoid coadministration
Tricyclic antidepressantsThese drugs act also on histaminergic and cholinergic receptors with antagonist effect [40,199] and for this reason they may enforce side effects related to antihistamines and anticholinergicsAvoid coadministration
DIMOtotoxic drugsWhen administered with ototoxic drugs, dimenhydrinate may hide ototoxicity [57]Caution needed
Drugs prolonging QT, proarrhythmic drugsDiphenhydramine (and then dimenhydrinate) may also prolong QT, but rarely [204,205]Avoid coadministration
DPHAntihypertensive drugsIt may cause fatigue, that is a possible adverse event of antihypertensive drugs also [61,206]Prefer a short time of treatment
Drugs prolonging QT, proarrhythmic drugsDiphenhydramine (and then dimenhydrinate) may also prolong QT, but rarely [204,205]Avoid coadministration
PMZEpinephrinePromethazine may reverse epinephrine’s vasopressor effect [70]Epinephrine should not be used to treat hypotension associated with promethazine overdose
Other drugs
ONDDrugs prolonging QT interval (e.g., antiarrhythmics, antipsychotics, antidepressant, antihistamines, some antimicrobial, methadone, HCQ), cardio-toxic, pro-arrhythmic drugsOndansetron may prolong QT interval and should be administered carefully in cardiologic setting and in people with Long QT Syndrome (LQTS) [76]Avoid the coadministration. Consider patient risk factors is also important. Freedman et al. showed that most cases were related to intravenous ondansetron, especially if administered with other drugs prolonging QT. Therefore, they suggest ECG and electrolyte screening only in patients at high risk and receiving ondansetron intravenously [205,207]
Tramadol, SSRI, SNRI, serotoninergic drugsTramadol acts on serotonin reuptake, whereas ondansetron is a 5HT3 receptor antagonist. Other authors suggest also a pharmacokinetic mechanism: competition at CYP2D6 leading to the reduction of tramadol active metabolite [208]Ondansetron may reduce tramadol’s analgesic effect and increase the risk of serotoninergic syndrome [76,208]
MCPL-DOPA, dopamine agonistsMetoclopramide is an antagonist of dopamine D2 receptors [40,71,72]Avoid coadministration
Alcohol and CNS depressants Increase of sedative action [72,192]Avoid coadministration
Anticholinergics and morphine derivativesThese drugs may antagonize the gastrointestinal effect of metoclopramide (a pro-kinetic drug). They are also related to constipation. However, this last effect is useful to avoid diarrhea from MCP [202,203]Avoid coadministration of anticholinergics and MCP for the best clinical effect. Evaluate risk benefit or the use of another compound if patient consumes opioids for pain management
Psychiatric drugsAntipsychotics may increase the risk of extrapyramidal side effects and antidepressants or serotoninergic drugs (e.g., SSRI, SNRI) may increase the risk of serotoninergic syndrome, since metoclopramide acts on dopamine and serotonin receptor [72]Avoid coadministration
Antihypertensive, hypertensive drugsMetoclopramide leads to hypotension [209]Administer carefully. Metoclopramide is administered for a short interval of time (5 days) [72]. Therefore, clinician should adequate antihypertensive therapy, reducing metoclopramide and/or antihypertensive drug dosage
CCSAntidiabetics or drugs increasing glucose levelsGlucocorticoids are associated to the raise of glucose levels [210]Avoid the coadministration
NSAIDs, PPIRisk of bleeding related to CCS [211], creating antagonism with PPICCS may be dangerous, especially if patient is affected by gastrointestinal ulceration (bleeding risk). The situation may be even more complex in the case patient consumes NSAIDs for pain treatment [211]
Antipsychotics, antidepressantsPsychiatric drugs and CCS are often associated to weight gain [212]. CCS can induce psychiatric symptoms, worsening patients conditions [213]Evaluate risk/benefit. Low-dose corticosteroid is not expected to generate psychiatric symptoms, whereas wait gain is a not desirable adverse event, which may happen in coadministration
ACDs, anticholinergic drugs; ACE, angiotensin-converting enzyme; ACh, acetylcholine; AH, antihistamines; ARB, angiotensin II receptor blockers: BDZ, benzodiazepines; BHS, betahistine; BPH, Benign prostatic hyperplasia; CCS, corticosteroids; CNS, central nervous system; CYP, cytochrome P 450; DIM, dimenhydrinate; DPH, diphenhydramine; DZP, diazepam;D2R, dopamine receptor D2; ECG, electrocardiogram; HCT, hydrochlorothiazide; KCL, potassium chloride; L-DOPA, levodopa; LOR, lorazepam; MAO, monoaminoxidase; MAOI, monoaminoxidase inhibitors; MCP, metoclopramide; MD, Meniere’s disease; mTOR, mechanistic target of rapamycin; NSAIDs, nonsteroidal anti-inflammatory drugs; OND, ondansetron; P-gp, p-glycoprotein; PMZ, promethazine; PPI, proton pump inhibitors; SNRI, serotonin-norepinephrine reuptake inhibitor; SPL, spironolactone; SSRI, serotonin reuptake inhibitors; TRI, triamterene.
Table 8. Adverse drug reactions related to the most common drugs used in the management of peripheral vestibular vertigo.
Table 8. Adverse drug reactions related to the most common drugs used in the management of peripheral vestibular vertigo.
AnticholinergicsMost common adverse events include stypsis, dyspepsia, blurred vision, dry mouth, cognitive decline, urinary retention, flushing, tachycardia, hypertension (but also hypotension), and mydriasis [40,47,49,53,54,214]. Psychiatric, ocular, cutaneous, vascular, nervous symptoms, and dryness of the respiratory mucous membranes of the nasopharynx have also been reported [47,49,53,54,214]
AntihistaminesSedation, CNS effects (e.g., vertigo, tinnitus, headache, blurred vision, diplopia, insomnia, tremor, fatigue), gastrointestinal symptoms, anti-muscarinic adverse events (e.g., urinary retention, dry mouth), allergic dermatitis, respiratory symptoms (related to the reduction of respiratory secretions) are common adverse events. Hematologic alterations, fever, cardiovascular symptoms, extrapyramidal symptoms (promethazine), jaundice are also described. Cinnarizine (administered with dimenhydrinate) is associated with CNS effects (e.g., extrapyramidal symptoms), somnolence, gastrointestinal adverse events, hypersensitivity, hematologic alterations [40,57,61,67,69].
BetahistineGastrointestinal symptoms and headache are the most common; cutaneous reactions, hypersensitivity, palpitation, feeling hot, eye irritation were also reported [23,27].
Diuretics
AcetazolamideAcidosis, electrolytic alterations (lowering of potassium and sodium levels), paresthesia, anorexia, tinnitus, gastrointestinal symptoms, polyuria, somnolence, confusion, altered taste. Suicide, cutaneous reactions, alteration of glucose levels, hepatic damage, hematopoietic alterations, other neurologic symptoms, nephrolithiasis and other reactions related to kidney damage were also reported [30,77].
HydrochlorothiazideHypercalcemia, impaired glucose tolerance, increase of cholesterol and triglycerides, skin cancer, closed angle glaucoma, hypersensitivity, thrombocytopenia, hypomagnesemia, hypokalemia, hyponatremia, hypotension, gastrointestinal symptoms, cutaneous manifestations, impotence and others [31,182,183]. It may also cause exacerbations in patients with systemic lupus erythematosus (SLE) [31].
Despite this, hydrochlorothiazide showed a better safety than other thiazides [215].
SpironolactoneHyperkaliemia, hyponatremia, hypovolemia, metabolic acidosis, gastrointestinal symptoms, sexual or hormonal changes (e.g., gynecomastia, erectile disfunction, amenorrhea), vocal alterations, kidney damage and urologic adverse events, hepatitis, cutaneous reactions, neurologic symptoms (e.g., lethargy, vertigo, ataxia; mainly in patients with hepatic encephalopathy), and leukopenia/thrombocytopenia are possible adverse events [34]. Gynecomastia is the most common collateral effect. In fact, spironolactone lacks of specificity for mineralocorticoid receptors and acts also on androgen and progesterone receptors [33,216].
TriamtereneMost frequent adverse events are hyperkalemia and gastrointestinal symptoms. Cutaneous reactions (including photosensitivity), dry mouth, megaloblastic anemia, pancytopenia, renal impairment and urinary symptoms, headache, and hyperuricemia were described [36,80].
Other antiemetics
MetoclopramideMetoclopramide’s more frequent adverse events are extrapyramidal effects, parkinsonism, galactorrhea, asthenia, somnolence, diarrhea, depression, and hypotension (intravenous administration) [72].
OndansetronOndansetron’s main adverse events are: headache, fatigue, flushing, gastrointestinal symptoms, and cutaneous reactions in the site of injection [75,76].
CNS, central nervous system; SLE, systemic lupus erythematosus.
Table 9. Pharmacogenomics of vertigo drugs.
Table 9. Pharmacogenomics of vertigo drugs.
Drug NameProtein InvolvedPolymorphismsDose Adjustment
Benzodiazepines
Class related
Elements
GABAA
receptor
19 subunits of GABAA receptor are described in literature: α (1–6), β (1–3), γ (1–3), δ, ε, π, θ, ρ (1–3). Properties of the assembled receptor will depend on nature of subunits. For example, DZP acts on receptor having α 1/2/3/5 subunits, but not on those expressing α4/6.
For further details, Korpi and Sinkkonen wrote on this topic [298]
Unknown clinical implications. The process may lead to development of new benzodiazepines or selective use of available drugs
CLZCYP3A4CYP3A4 1*B has a doubtful effect (see DZP section) [299]Toth et al. showed a role of CYP3A4 different expression in dosage adjustment, but CYP expression was not different between people expressing different polymorphisms (no relation genotype-phenotype). However, the evaluation of low and normal expressers may be useful to dispose dose reduction (half- dose in low expressers) [299]
CYP3A4*22 resulted in decreased activity [299]See the section above.
See DZP section for other possible polymorphisms of CYP3A4
NAT2NAT2*4 is associated with rapid acetylation of 7-amino-clonazepam (main metabolite), whereas NAT2*5, *6 and *7 are related to slow acetylation [299]Slow acetylation pathway, associated with high CYP3A4-induced metabolism, may lead to the accumulation of 7-amino-clonazepam. This metabolite has a partial agonist effect on GABAA receptor and may modify clonazepam effects. Furthermore, it is associated to withdrawal symptoms. In patients with high CYP3A4 activity and low NAT2 acetylation a longer dose reduction period may be needed [299,300]
DZPCYP2C19CYP2C19*1 allele is wild type (normal activity) [301]Not required
CYP2C19*17 allele is related to increased activity. Heterozygote individuals (*1/*17) are considered rapid metabolizers, whereas homozygote (17*/17*) are ultrarapid metabolizers [301]Dose increase based on clinical effectiveness and holding in account other drugs taken
CYP2C19*2 and *3 are associated with the absence of activity. Individuals with one nonfunctional allele are intermediate metabolizers. Instead of, people with 2 nonfunctional alleles are poor metabolizers [301]Dose reduction based on clinical effectiveness and side effects.
Consider other drugs taken
CYP3A4Several polymorphisms have been identified (e.g., CYP3A4 1*B), but their frequency or their effect on benzodiazepines is not clear. In general isoform activity seem to be preserved [302,303]. Punctiform mutations may also affect CYP3A4 function and ethnicity is another important factor [304]. More than 856 CYP3A4 variants are documented [304]
CYP3A4*1 is related to normal function [305]-
CYP3A4*20 is related to loss of function. CYP3A4*22 is associated with reduced activity [305]Unknown impact on benzodiazepines, but possible dose reduction
CYP3A4 1*B has an uncertain effect on enzyme’s activity. Some studies described a decreased action, other an increased action (tacrolimus, ciclosporin, simvastatin) [305]Unknown impact of benzodiazepines
CYP3A4 1*G seem to be associated with higher substrate clearance, but further studies are needed and some controversial results are described [305]Unknown impact of benzodiazepines, but possible dose increase
Antihistamines
Class related
Elements
H1 receptorPolymorphisms in this gene (rs901865 allele) were associated to increased side
effects for desloratadine, and may have similar effects on other antihistamines [306]
The experience of Chu showed that H1 polymorphisms (CC genotype) were
associated to a better clinical response in allergic rhinitis, whereas other
polymorphisms (CT + TT) generated a worse effect [307]
Further studies needed in vestibular diseases
H4 receptorThe rs77041280 mutant genotype
(TA + TT) and the rs77485247
mutant genotype (TA + AA) may lower drug effects and enhance adverse events after using antihistamines for allergic
rhinitis [308]
Further studies needed in vestibular diseases
C5AR1Polymorphisms of this protein (-1330 T/G) seem to be a predictor of H1 antihistamines efficacy in urticaria [309]Further studies needed
DPHCYP2D6CYP2D6*1, *2, *33, *35 alleles generate a normal function CYP2D6. On the basis of alleles homo-heterozygosis patients may be classified in ultrarapid metabolizers, extensive (or normal) metabolizers, intermediate metabolizers, and poor metabolizers [310]

Zhang et al. described some example of genotype related to a specific phenotype [311]
No dose adjustment in homozygosis. Evaluate in case of heterozygosis
CYP2D6*3, *4, *5, *6, *7,*8, *11, *12, *13, *14,*15, *16, *18, *19, *20, *21, *38, *40, *42 are examples of nonfunctional alleles [310].Dose reduction may be needed, depending on homo-heterozygosis
*9, *10, *17, *36, *41 alleles have decreased function [310].Dose reduction may be needed, depending on homo-heterozygosis
CYP2D6 *1×N, *2×N, *35×N are associated with increased activity [310]Dose increase may be needed, depending on homo-heterozygosis. In ultrarapid metabolizers DPH generated paradoxical excitation [312]
MCZCYP2D6No dedicated literature on meclizine (see DPH section for CYP2D6 polymorphisms possible influence)
Anticholinergics
Class related
Elements
Muscarinic
receptors
There are documented polymorphisms of M1 and M2 in literature, whereas M3
variants are less known. No clear link between phenotype and genotype has been described. Implications on drug
responsiveness is not available [313]
Further studies needed
Other drugs
MCPCYP2D6CYPD6 *1 and *2 represent wild type variants. Genotype CYP2D6*wild type/wild type is associated with normal enzyme activity (for further information on single CYP2D6 alleles, see DPH section) [98]No dose adjustment
CYP2D6 *wild type/*10, *5/*10, and *10/*10 were associated with reduced activity and higher metoclopramide levels [98]Dose reduction
CYP3A4, CYP2C19No dedicated literature on MCP (see previous sections for CYP3A4 and CYP2C19 main polymorphisms)
CYP1A2It has a minor role on MCP metabolism. Koonrungsesomboonet al. performed a systematic review evaluating polymorphisms involved in CYP1A2 activity [314]
KCNH2This gene determines the production of a subunit of a potassium channel, whom mechanism is voltage dependent. KCNH2 seems to be a predictive factor for MCP response or side effects development. The drug may exert its action on
potassium channels [315]
Adequate dosage on the basis of different polymorphisms. Further studies needed
ADRAIDADRAID codifies α1D adrenergic receptor, that may be a target of MCP or matter of interaction with dopaminergic system. It seems to be related to MCP clinical efficacy [315].Adequate dosage on the basis of different polymorphisms. Further studies needed
HTR4HTR4 is responsible of 5HT4 receptor production. MCP is a partial agonist of this target. HTR4 polymorphisms are associated with MCP side effects [315]Adequate dosage on the basis of different polymorphisms. Further studies needed
ONDCYP2D6Genotypes *1/*1, *1/*2, *1/*4, *1/*5, *1/*9, *1/*41,*2/*2,*41/*41 are associated with normal activity (further information on single alleles in previous sections) [316]

In general, genetic deficits may be compensated by other CYP450 isoforms [76]
No dose adjustment
Genotypes *3/*4,*4/*4, *5/*5, *5/*6 are related to poor metabolism [316]Few studies available, possible dose reduction
Genotypes *4/*10,*4/*41, *5/*9 are associated with intermediate metabolism [316]Few studies available, possible dose reduction
Genotypes 1/*1xN, *1/*2×N, *2/*2×N are related to ultrarapid metabolism [316]Choose a drug not metabolized by CYP2D6 or dose increase [316]
CYP3A4No dedicated literature available (see previous sections for CYP3A4 polymorphisms)
CYP1A2CYP1A2*1A is the wild type allele in vitro and in vivo [317]No dose adjustment
CYP1A2*1F has been associated with increased inducibility (in vitro) [317,318]Possible dose increase, but further studies needed
CYP1A2*1K, *3, * 4, *6 and*7 (in vitro) are associated with abolished (*6) or decreased activity [317,319]
CYP1A2*8, *11, *15, *16 have the same effect in vivo [317]
Possible dose reduction but further studies are needed
OCT1Tzvetkov et al. showed that OCT1
polymorphisms resulting in a loss of function may moderately increase ondansetron concentration, efficacy, and side effects [320]
Evaluate OCT1 polymorphisms in patients with CYP2D6 ultrarapid metabolism may be useful to predict clinical response [320]
P-gpCG haplotype (C3435TandG2677T) of P-gp was associated to an increased risk of nausea and vomiting in cancer. 3435C > T polymorphism was related to efficacy in the early phase of chemotherapy. TT genotype was associated to a higher effectiveness. CTG(C3435T, C1236T, and G2677T) patients suffered for nausea and vomiting with a major frequency [321]Evaluate dose increase or shift to other drugs. Further studies needed
5HT3B receptorPolymorphisms of this protein are associated with different clinical effect.
Deletions-100-102delAAG of the
promotor region are characterized by worse clinical effect [322]
Evaluate dose increase or shift to other drugs. Further studies needed
CLZ, clonazepam; CYP450, cytochrome 450; C5AR, component 5a receptor; DPH, diphenhydramine; DZP, diazepam; GABA, gamma-aminobutyric acid; MCP, metoclopramide; MEC, meclizine; NAT, N-acetyltransferase; OCT, organic cation transporter; OND, ondansetron; P-gp, p-glycoprotein.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Di Mizio, G.; Marcianò, G.; Palleria, C.; Muraca, L.; Rania, V.; Roberti, R.; Spaziano, G.; Piscopo, A.; Ciconte, V.; Di Nunno, N.; et al. Drug–Drug Interactions in Vestibular Diseases, Clinical Problems, and Medico-Legal Implications. Int. J. Environ. Res. Public Health 2021, 18, 12936. https://doi.org/10.3390/ijerph182412936

AMA Style

Di Mizio G, Marcianò G, Palleria C, Muraca L, Rania V, Roberti R, Spaziano G, Piscopo A, Ciconte V, Di Nunno N, et al. Drug–Drug Interactions in Vestibular Diseases, Clinical Problems, and Medico-Legal Implications. International Journal of Environmental Research and Public Health. 2021; 18(24):12936. https://doi.org/10.3390/ijerph182412936

Chicago/Turabian Style

Di Mizio, Giulio, Gianmarco Marcianò, Caterina Palleria, Lucia Muraca, Vincenzo Rania, Roberta Roberti, Giuseppe Spaziano, Amalia Piscopo, Valeria Ciconte, Nunzio Di Nunno, and et al. 2021. "Drug–Drug Interactions in Vestibular Diseases, Clinical Problems, and Medico-Legal Implications" International Journal of Environmental Research and Public Health 18, no. 24: 12936. https://doi.org/10.3390/ijerph182412936

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

Article Metrics

Back to TopTop