Next Article in Journal
Gastrointestinal Cancer: Outcomes and Therapeutic Management
Previous Article in Journal
Anxiety and Depression in Adolescents with Prelingual Hearing Loss: Prevalence and Risk Factors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pharmacological Management of Oral and Esophageal Candidiasis: A Clinical Pharmacotherapy Perspective

1
Department of Pharmacy, Institute of Science Tokyo Hospital, 1-5-45 Yushima, Bunkyo-ku 113-8519, Tokyo, Japan
2
Department of Oral and Maxillofacial Surgery, Nishinomiya Watanabe Hospital, 10-22 Murokawa-cho, Nishinomiya 662-0863, Hyogo, Japan
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2025, 14(21), 7537; https://doi.org/10.3390/jcm14217537 (registering DOI)
Submission received: 12 September 2025 / Revised: 9 October 2025 / Accepted: 20 October 2025 / Published: 24 October 2025
(This article belongs to the Section Dentistry, Oral Surgery and Oral Medicine)

Abstract

Candida spp. are common components of normal microbiota in the oral cavity. However, Candida albicans can be a primary cause of superficial infections in the oral cavity and esophagus, especially in immunocompromised individuals. While these infections are rarely life-threatening, they can significantly impair quality of life and, in severe cases, progress to hematogenous dissemination. Oral candidiasis often exhibits as pseudomembranous, erythematous (atrophic), chronic hyperplastic, denture stomatitis, or angular cheilitis. Esophageal candidiasis is typically diagnosed by upper endoscopy, which involves histological examination and brushing. Clinical guidelines recommend topical antifungal agents for mild oral candidiasis, and systemic agents for moderate-to-severe disease or when topical therapy fails. However, azole antifungals pose a substantial risk of drug–drug interactions, primarily due to the inhibition of cytochrome P450 enzymes and drug transporters, which dramatically alters the pharmacokinetics of co-administered drugs. Additionally, amphotericin B, a polyene macrolide antibiotic, may cause nephrotoxicity and electrolyte disturbances (e.g., hypokalemia and hypomagnesemia). Moreover, the co-administration of nephrotoxic drugs may augment the toxicity associated with amphotericin B. Therefore, this review aimed to provide a comprehensive overview of the management of oral and esophageal candidiasis from the viewpoint of clinical pharmacology, with a particular focus on drug–drug interactions and adverse effect profiles.

Graphical Abstract

1. Introduction

Candida is a yeast that resides on the surface epithelium of the oral cavity in healthy individuals [1]. There are over one hundred Candida spp., including C. albicans, C. glabrata, C. parapsilosis, and C. tropicalis [2], which are causal pathogens of human mucosal and invasive infections [3]. C. albicans is a major pathogen responsible for oral and esophageal candidiasis [4,5]. C. albicans is the most common cause of oropharyngeal disease (approximately 70–80% of isolated cases); other causes include non-albicans species or mixed infections [3]. This review focuses on C. albicans and highlights the contributions of non-albicans species to disease patterns relevant to dental practice. C. albicans has a prevalence of approximately 30–50% in the oral cavity of healthy individuals, 50–65% in denture wearers, 65–90% in long-term hospitalized patients, and up to 90–95% in immunocompromised hosts [1], highlighting its role as an opportunistic pathogen. When immune function is compromised, Candida can colonize and proliferate in the oral and esophageal mucosa, leading to fungal invasion and the development of characteristic white plaques with underlying erythema [6]. Oral candidiasis commonly presents as pseudomembranous, erythematous (atrophic), chronic hyperplastic, denture stomatitis, or angular cheilitis [7]. Clinical diagnosis may be supported by KOH microscopy and culture (e.g., chromogenic media), with biopsy for hyperplastic lesions to exclude leukoplakia/dysplasia [8,9].
Oral and esophageal candidiasis typically occurs in immunocompromised individuals, such as those with malignancies, solid organ transplants, HIV infection, or those receiving cytotoxic chemotherapy and immunosuppressive agents (e.g., corticosteroids) [10,11,12,13,14]. Thus, individuals with poor oral condition, including those with dental prostheses or hyposalivation caused by radiation therapy or Sjögren’s syndrome, are at high risk of oral and esophageal candidiasis [15,16,17,18]. Pooled estimates suggest that the prevalence of oropharyngeal candidiasis is approximately 50% in high-risk cohorts, such as people with HIV, with C. albicans being the most frequently isolated species [19]. Upper endoscopy with histology or brush cytology is used to confirm esophageal candidiasis [5]. C. albicans is also the leading etiology for esophageal disease; upper endoscopy with histology and brushing is the diagnostic standard and helps distinguish Candida esophagitis from cytomegalovirus and herpes simplex virus infection, pill esophagitis, reflux esophagitis, and eosinophilic esophagitis [5]. Although the prognosis of oral and esophageal candidiasis is generally favorable, severe cases may result in systemic dissemination, especially in immunocompromised hosts [20,21]. Additionally, candidemia is associated with high mortality and morbidity due to complications such as endocarditis and ocular candidiasis [22]. A previous study reported that approximately 20% of patients experienced recurrence, often involving different Candida species from those isolated during the initial episode [23]. Therefore, optimal pharmacotherapy is essential for effective treatment.
However, considerable interindividual variability exists in the response to antifungal therapy, stemming from genetic and environmental factors (e.g., organ dysfunction, gene polymorphisms, and drug–drug interactions) [24,25,26]. Consequently, drug–drug interactions can complicate treatment, leading to therapeutic failure or unforeseen toxicity. Although previous reviews have focused on drug selection, only a few have comprehensively summarized pharmacokinetics, pharmacodynamics, and drug–drug interactions.
Therefore, this review aims to provide a comprehensive overview of drug–drug interactions and toxicity in oral and esophageal candidiasis, with a focus on clinical pharmacotherapy.

2. Oral Health Care

Effective management of oral candidiasis requires a holistic strategy that extends beyond routine oral hygiene. Oral health care refers to the comprehensive management of the oral cavity as a functional system including daily hygiene, maintenance of oral function, and targeted reduction in local risk factors. Rather than serving merely as an adjunct to antifungal pharmacotherapy, oral health care forms the foundation of treatment. It reduces the Candida burden and disrupts biofilms, which are intrinsically drug-tolerant, thereby enhancing the clinical response [27,28]. These measures are applicable across clinical forms of oral candidiasis and denture-related disease, in line with guidelines [29].
The central pillar of oral health care is the mechanical disruption and removal of biofilms and dental plaques. Dentate patients should brush their teeth twice daily, followed by interdental cleaning with floss or interdental brushes once daily. Routine cleansing of mucosal surfaces prone to Candida colonization, particularly the tongue dorsum—which serves as a major reservoir for Candida—is also recommended. Edentulous patients or those with compromised mucosal integrity such as those undergoing chemotherapy, require gentler cleaning techniques [30]. This may involve the use of soft-bristled toothbrushes, sponge swabs, or sterile gauze to clean the mucosa, palate, and tongue without causing trauma; however, the efficacy of plaque removal is relatively low [31].
Denture hygiene is another critical aspect of oral health care, as poorly maintained or long-term prostheses are established risk factors for denture stomatitis, which is a common manifestation of oral candidiasis [15]. An evidence-based regimen suggests combining daily mechanical cleaning with a non-abrasive cleanser and brief chemical disinfection (e.g., hypochlorite immersion). For acrylic appliances, short immersion in approximately 0.5% dilute sodium hypochlorite for no more than 10 min is recommended, while hypochlorite should be avoided for metal-containing dentures due to corrosion risk [32,33]. Additionally, nocturnal wear increases oral colonization and is associated with adverse outcomes in older adults [15]. Therefore, patients should be instructed to remove dentures overnight, which facilitates mucosal recovery and reduces exposure to fungal reservoirs.
Xerostomia (dry mouth) is a common and clinically significant cause of oral candidiasis. Reduced salivary flow compromises self-cleansing and diminishes key antimicrobial constituents, including mucins, histatins, lysozymes, lactoferrin, and secretory IgA, thereby favoring Candida adherence and persistence [34,35,36,37]. Moreover, acidic salivary conditions and a relatively closed, low-saliva environment at the denture–mucosa interface promote Candida growth and virulence; low salivary pH has been repeatedly linked to higher Candida carriage in denture wearers [38,39]. Overnight wearing of dentures exacerbates this condition, promoting early Candida adherence and increasing the risk of denture-related candidiasis [40].
Recommended management strategies include maintaining adequate fluid intake, stimulating saliva production with sugar-free gum or candy, and using saliva substitutes [29,41]. The management of xerostomia requires a coordinated multidisciplinary approach because it often arises from multiple causes, including systemic diseases, medications, oral hypofunction, and chemotherapy. Consequently, collaboration among physicians, dentists, and pharmacists is essential for effective intervention [42,43,44].

3. Clinical Guidelines for the Treatment of Oral and Esophageal Candidiasis

Therapeutic options for oral and esophageal candidiasis are summarized in Table 1 and Table 2, and treatment is determined by disease severity and site of infection [29]. In pregnant women, intravenous amphotericin B is an alternative, as systemic azole antifungals are contraindicated in the first trimester owing to teratogenicity. In such cases, local administration is preferred.

3.1. Oral Candidiasis

First-line treatment for mild oral candidiasis involves the topical administration of clotrimazole troches, miconazole mucoadhesive buccal tablets, nystatin suspensions, or pastilles [45,46,47,48]. Oral fluconazole is recommended in cases of moderate-to-severe disease or unresponsiveness to first-line treatment [49,50,51]. When fluconazole treatment fails, increased dose of fluconazole or other azole antifungals, including itraconazole, posaconazole, and voriconazole are administered [52,53,54]. Recommended treatment duration for patients without serious comorbidities is 7–14 days, with further extension for 14–28 days in patients with refractory disease [29].

3.2. Esophageal Candidiasis

Systemic fluconazole is recommended as the initial treatment of esophageal candidiasis, regardless of the severity [29,55]. For patients who cannot tolerate fluconazole or in whom it is contraindicated, intravenous echinocandins or amphotericin B are recommended as alternative initial therapies. In refractory cases, options include dose escalation of fluconazole or switching to another antifungal agent (itraconazole, posaconazole, voriconazole, intravenous echinocandins, or liposomal amphotericin B). The recommended duration of therapy is 14–21 days.

4. Pharmacological Mechanism

The pharmacological actions of the individual antifungal agents are shown in Figure 1. Notably, azole antifungals and echinocandins exhibit selective toxicity toward Candida because their target molecules are specific to fungi and are not found in host cells.
Amphotericin B, a polyene macrolide, specifically binds to ergosterol in the fungal cell membrane and forms pores that allow the passage of protons and monovalent cations such as potassium through increased membrane permeability [56,57,58]. This leads to membrane depolarization, affecting fungal and host cells. A liposomal formulation of amphotericin B has been developed and is widely used to reduce toxicity while maintaining antifungal efficacy [62]. Topical nystatin, which is poorly absorbed systemically, has minimal adverse effects and is particularly useful for the reducing colonization of Candida on the tongue dorsum, a major reservoir of this organism [63].
In contrast, azole antifungals, including triazoles and imidazoles, inhibit a key enzyme 14-α-demethylase (CYP51A1), which catalyzes the conversion of lanosterol to ergosterol, an essential component of fungal membranes [59,60]. Inhibition of this enzyme results in the accumulation of toxic sterol intermediates. Consequently, azole antifungals disrupt membrane integrity and function, thereby hindering fungal growth.
The fungal cell wall consists of several polysaccharides, including 1,3-β-D-glucan, 1,4-β-D-glucan, 1,6-β-D-glucan, and diverse glycoproteins [64]. Accordingly, echinocandins, a class of cyclic hexapeptides, noncompetitively block the Fks1p subunit of the enzyme responsible for synthesizing 1,3-β-D-glucan, which is a specific component of the fungal cell wall [61]. This action weakens the fungal cell wall, leading to osmotic instability and cell lysis.

5. Clinical Pharmacokinetics

Antifungals used to treat candidiasis exhibit diverse pharmacokinetic profiles [65,66,67], and determining their ability to reach infection sites is a primary clinical concern. Amphotericin B has poor oral bioavailability (approximately 0%) and remains confined to the gastrointestinal lumen when administered orally [68]. In contrast to conventional amphotericin B, liposomal amphotericin B exhibits lower tissue permeability [69,70]. Amphotericin B is not metabolized by cytochrome P450 (CYP) enzymes and is primarily eliminated via renal excretion [70]. In contrast, liposomal amphotericin B exhibits nonlinear pharmacokinetics [58] because lipid carriers are relatively large particles that restrict the diffusion of drugs into non-pathological tissues, while preferentially accumulating in infected tissues due to increased vascular permeability.
Azole antifungals, except fluconazole, are substrates of CYP enzymes and drug transporters [71]. For instance, CYP3A4 mediates the hepatic metabolism of itraconazole to its active metabolite, 14-hydroxyitraconazole, whereas plasma esterases convert isavuconazonium to its active moiety, isavuconazole [72,73]. Voriconazole is primarily metabolized by both CYP2C19 and CYP3A4. Its nonlinear pharmacokinetics results from the saturation of CYP2C19, whose activity varies substantially due to genetic polymorphisms [74,75]. In contrast, posaconazole predominantly undergoes metabolism via uridine diphosphate glucuronosyltransferase and is carried via P-glycoprotein (P-gp) to efflux from the cell into the lumen [76]. Fluconazole is largely eliminated via glomerular filtration following tubular reabsorption [74].
Caspofungin, an echinocandin, is degraded by hydrolysis and N-acetylation, and its metabolites are mainly excreted in bile and feces [77]. Micafungin is metabolized by sulfatase into the catechol form, which is further metabolized by catechol O-methyltransferase into the methoxy form and finally undergoes non-enzymatic ring-opening to form a methoxy derivative [78].

6. Toxicity

Systemic administration of amphotericin B causes infusion-related reactions, typically manifesting as fever and chills [68] (Table 3). These reactions are believed to result from the increased prostaglandin E2 synthesis [79]. Premedication is recommended to prevent infusion-related reactions [80]. For instance, premedication with corticosteroids can mitigate these symptoms, unlike non-steroidal anti-inflammatory drugs and acetaminophen [81].
Nephrotoxicity, a dose-dependent and generally reversible adverse effect of amphotericin B, often limits its systemic administration [68,82,83]. The drug binds to cholesterol in host cell membranes, increasing membrane permeability in the renal vasculature. Additionally, amphotericin B increases the permeability of the macula densa cells, which sense sodium chloride delivery. This leads to the activation of the tubuloglomerular feedback mechanism, afferent arteriolar vasoconstriction, and a subsequent reduction in the glomerular filtration rate. Altered membrane permeability subsequently results in electrolyte disturbances, including the loss of potassium and magnesium [68]. Indeed, sodium enters distal tubular cells via amphotericin B–induced pores, followed by potassium efflux [84,85].
Long-term use of azole antifungals can result in hepatotoxicity and hormone-related adverse effects (e.g., gynecomastia and adrenal insufficiency) [86]. Hepatotoxicity associated with azole antifungals manifests as hepatocellular and cholestatic patterns [86]. However, the mechanisms underlying this hepatotoxicity are unclear. Nevertheless, the blood concentration of voriconazole is useful for identifying populations at high risk of hepatotoxicity, especially among Asian populations [87,88].
Azole antifungals block lanosterol 14-α-demethylase, which is involved in the production of ergosterol [89]. Azoles can also inhibit mammalian CYP enzymes, which are involved in steroidogenesis, leading to endocrine-related adverse effects such as gynecomastia, adrenal insufficiency, and pseudo-hyperaldosteronism [90]. They are also known to prolong QT interval; however, the exact mechanism is not fully understood. Several azoles also inhibit the human Ether-à-go-go–Related Gene (hERG) potassium channel, which is involved in cardiac repolarization [91]. Therefore, drug interactions are a notable concern, as azoles can inhibit multiple CYP enzymes and transporters, resulting in elevated plasma concentrations of co-administered drugs with a potency of QT prolongation [92]. However, a previous study reported that isavuconazole shortened QT intervals in a dose-dependent fashion [93]. Moreover, voriconazole is associated with visual disturbances (e.g., photopsia) and concentration-dependent central nervous system side effects, while itraconazole may cause gastrointestinal symptoms, partly due to its additive agent with β-cyclodextrin [94,95].
Echinocandins exhibit better tolerability than amphotericin B and azoles [96]. The most common adverse effects of echinocandins include infusion-related reactions such as rashes and flushing [97]. Although fever is particularly common with caspofungin, it may be alleviated by reducing the infusion rate [98]. Mild elevations in hepatic aminotransferases and alkaline phosphatase can also occur due to caspofungin but are typically asymptomatic and reversible [99,100].

7. Drug–Drug Interaction

Drug–drug interactions can occur through different mechanisms, resulting from either pharmacokinetics or pharmacodynamics. Antifungal agents interact with a variety of drugs. Some common antifungal drug–drug interactions are discussed below.

7.1. Pharmacokinetic Interaction

Pharmacokinetic interactions arise when one drug alters the pharmacokinetic processes of another drug, including absorption, distribution, metabolism, and excretion. Most clinically relevant interactions involve drug metabolizing enzymes and transporters such as CYP and P-gp. The mechanism of drug–drug interactions is illustrated in Figure 2. CYP3A4 is the most abundant drug-metabolizing enzyme in the gut and liver [101,102,103], with inter-individual variability largely explained by genetic polymorphisms [102]. In contrast, P-gp expressed on the apical membrane exports substrates out of the cell [104].
Additionally, drug–drug–gene interactions should be considered when planning a treatment strategy [105,106]. Genetic variations exist in drug-metabolizing enzymes and transporters. Thus, the extent of drug interactions varies according to genetic polymorphisms, even when patients receive the same victim–perpetrator combination. For example, a common interaction between tacrolimus and voriconazole results from the inhibition of CYP3A4 metabolism [106,107,108,109]. This effect is more pronounced in carriers of CYP2C19 loss-of-function alleles, since CYP2C19 polymorphisms strongly influence voriconazole clearance [110,111].
Azole antifungals are particularly problematic in clinical practice because they inhibit multiple CYP isoenzymes and P-gp [74,112,113]. The addition of azole antifungals leads to a sustained increase in the co-administered victim drug concentrations. Unlike other antifungal classes, since azoles are often administered for weeks, even moderate inhibition can accumulate over time. Consequently, azoles are recognized as “high-risk perpetrators” of drug–drug interactions and require therapeutic drug monitoring or careful dose adjustment as needed.
Although azole antifungals inhibit CYP enzymes and transporters, their targets vary among the individual agents (Table 4). Itraconazole is a strong inhibitor of CYP3A4 and P-gp [114,115]. Additionally, a metabolite of itraconazole (14-hydroxyitraconazole) is an equally potent CYP3A4 inhibitor [115]. Meanwhile, voriconazole is recognized as a substrate and inhibitor of CYP2C9, CYP2C19, and, to a lesser extent, CYP3A4 [67]. The extent of this interaction depends on the plasma concentration of voriconazole. The extent of interaction depends on plasma voriconazole concentration; tacrolimus exposure increases markedly when voriconazole levels exceed 2.0 µg/mL [116]. Fluconazole is a moderate inhibitor of CYP2C9, CYP2C19, and CYP3A4 [117].

7.1.1. Opioids

The pharmacokinetics of opioids, such as oxycodone and fentanyl, involve CYP3A4 metabolism [118]. Consequently, azole antifungals interfere with oxycodone metabolism [119]. The co-administration of voriconazole increases the risk of oxycodone-induced somnolence [120]. In contrast, the inhibition of CYP3A4 slightly affects fentanyl when administered intravenously [121]. This is because fentanyl clearance depends primarily on hepatic blood flow rather than CYP activity. Thus, the administration route produces variations in drug–drug interactions between opioids and azole antifungals.

7.1.2. Antithrombotic Drugs

Warfarin is a racemic mixture of R and S isomers [122]. S-warfarin is more than five times as potent as R-warfarin [123,124]. S-warfarin mainly undergoes the metabolism of CYP2C9 [125,126]. Thus, the CYP2C9 inhibitor fluconazole inhibits warfarin metabolism and increases bleeding risk [127,128]. Similarly, a clear elevation in the prothrombin time international normalized ratio (PT-INR) is observed when voriconazole is used with warfarin [129]. Miconazole, a topical formulation, has been reported to interact with the CYP2C9 metabolism of warfarin, and thereby elevates PT-INR [130,131,132]. This is attributed to the absorption of miconazole by the gut. Dose optimization is a valuable approach for managing bleeding risk when warfarin is administered.
All direct oral anticoagulants are substrates of P-gp in the gut and proximal tubules [133]. Accordingly, several azole antifungals that inhibit P-gp are associated with increased blood concentrations of direct oral anticoagulants [134]. A study of the drug interactions between rivaroxaban and fluconazole suggested a notable impact on the pharmacokinetics of rivaroxaban. Moreover, this interaction is harmful when rivaroxaban is co-administered with fluconazole in addition to cyclosporine (inhibitors of CYP3A4 and P-gp) [135]. The contribution of CYP3A4 varies among DOACs; it is particularly important for apixaban and rivaroxaban. Co-administration with strong CYP3A4 inhibitors or inducers should be avoided in these cases [136,137].
Clopidogrel undergoes a 2-step conversion with an intermediate active metabolite via mainly CYP2C19 [138]. Prasugrel is activated by CYP3A4 and 2B6 [139]. Ketoconazole decreases the active metabolite formation of clopidogrel and attenuates the pharmacodynamic effect on platelet function but does not affect the pharmacokinetics of prasugrel [140]. However, ticagrelor, a pure CYP3A4 substrate, is not recommended when a strong CYP3A4 inhibitor (itraconazole) is administered [141].

7.1.3. Cardiovascular Drugs

The primary elimination route of digoxin involves glomerular filtration and secretion via P-gp [142]. In addition to amiodarone, the inhibition of P-gp by itraconazole and posaconazole alters the bioavailability and renal clearance, increasing the risk of digoxin intoxication [143,144,145,146].
CYP3A4 is essential for the metabolism of several mineralocorticoid receptor antagonists, including eplerenone, esaxerenone, and finerenone [147,148,149]. Close monitoring should be performed for the hyperkalemic risk of mineralocorticoid receptor antagonists co-administered with CYP3A4 inhibitors, including azole antifungals [112]. Older age increases the risk of hyperkalemia when mineralocorticoid receptor antagonists and CYP3A4 inhibitor simultaneously [150].
The pharmacokinetic profiles of statins differ in terms of drug transporters and metabolism. Hepatic metabolism occurs primarily through CYP3A4 for simvastatin, lovastatin, and atorvastatin, whereas fluvastatin is metabolized mainly through CYP2C9 and pitavastatin and rosuvastatin are converted through to lesser extent, CYP2C9 [151,152]. The CYP3A4 inhibitor dramatically increases statins metabolized by CYP3A4 (e.g., simvastatin), whereas fluconazole interacts with fluvastatin via the CYP2C9 interaction [153]. Hepatic uptake of statins is mediated by organic anion-transporting polypeptides 1B1 and 1B3, but not P-gp [152]. Thus, the switch from simvastatin to pravastatin can be ignored when co-administrating with itraconazole [154].

7.1.4. Immunosuppressant Drugs

An immunosuppressive regimen of calcineurin inhibitors (e.g., tacrolimus and cyclosporine) and glucocorticoids is often used to treat autoimmune diseases and transplantation.
There is large variability in the pharmacokinetics of calcineurin inhibitors, suggesting their utility in therapeutic drug monitoring [155,156,157]. Cyclosporine is a substrate of CYP3A4 and P-gp [89]. A randomized study identified a slow increase in cyclosporine concentration during the 2 weeks following fluconazole administration [158]. In particular, close monitoring of drug concentration is recommended for fluconazole doses above 200 mg/day [159]. Posaconazole has a greater impact on the pharmacokinetics of cyclosporine than fluconazole [160]. Tacrolimus is primarily metabolized by CYP3A5, and to a lesser extent by CYP3A4 [161]. Numerous differences exist in the effects of azole antifungals on tacrolimus pharmacokinetics [162]. The interaction between tacrolimus and azole antifungals was blunted by CYP3A5 expressor [163]. Moreover, drug–drug interactions between tacrolimus and azole antifungals differ depending on the administration route [164]. This is because intravenous administration of tacrolimus is not relevant to the metabolism of CYP3A4 in the intestine.
Co-administration of azole antifungals blocks CYP3A4, which potentially increases the exposure to both methylprednisolone and prednisolone. However, itraconazole causes no change in the pharmacokinetic profile of prednisolone despite notable adrenal suppression by methylprednisolone [165]. Isavuconazole did not affect the exposure to prednisolone [166]. Thus, the combination of glucocorticoids and azole antifungals affects the magnitude of drug–drug interactions.

7.1.5. Chemotherapeutic Drugs

Several studies have suggested that the interactions between drugs and azole antifungals drive the development of peripheral neuropathy caused by vinca alkaloids. Azole antifungals inhibit CYP3A4 metabolism of vincristine and increase the risk of toxicity, including peripheral neuropathy and secondary constipation, due to neuropathy in hematologic malignancies [167]. In addition, itraconazole inhibits CYP3A4 and P-gp, further contributing to the elevation of intracellular concentrations in nerve cells [168]. In contrast, fluconazole has less potential to increase the risk of neurotoxicity than itraconazole and voriconazole [169].
Most kinase inhibitors are metabolized by CYP3A4 [170]. Co-administration of azole antifungals predisposes patients to drug–drug interactions with tyrosine kinase inhibitors [171]. Therefore, individual doses should be personalized based on in-depth discussions among the medical staff to avoid undesirable adverse events.

7.2. Pharmacodynamic Interaction

Pharmacodynamic interactions occur when the pharmacological effects of a drug are altered by another co-administered drug. This interaction is generally classified as synergistic, additive, or antagonistic, according to the combination therapy. In clinical practice, the additive effects pose significant risks, particularly when they lead to unintended toxicity.

Nephrotoxicity and Electrolyte Disorder

Drug-induced nephrotoxicity involves multiple mechanisms [172]. For instance, nephrotoxicity with amphotericin B is exacerbated when co-administered with other nephrotoxic drugs, such as calcineurin inhibitors and antihypertensive drugs (angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers) [173,174,175].
Likewise, additive effects on electrolyte depletion are considered when amphotericin B is combined with drugs that promote the urinary excretion of electrolytes. Loop and thiazide diuretics increase the urinary excretion of potassium and magnesium by inhibiting sodium-potassium-chloride and sodium-chloride symporters, respectively [176]. Glycyrrhizin blocks 11β-hydroxysteroid dehydrogenase 2 that converts cortisol (active form) to cortisone (inactive form), thereby elevating cortisol level [177]. Consequently, cortisol acts on mineralocorticoid receptors and enhances renal potassium excretion. In addition, older age is a risk factor for Kampo medicine-induced hypokalemia [178]. Fludrocortisone also binds to mineralocorticoid receptors and thereby increases the risk of hypokalemia [179].
Epidermal growth factor receptor (EGFR) plays a key role in renal tubular signaling [180]. The anti-epidermal growth factor receptor (EGFR) monoclonal antibodies cetuximab and panitumumab, which have relatively long half-lives, target the EGFR cascade and cause hypomagnesemia [181]. This mechanism impairs the expression of channel transient receptor potential M6 that reabsorbs magnesium in the distal nephron [182]. Therefore, the combination of amphotericin B and anti-EGFR monoclonal antibodies may increase the risk of hypomagnesemia. Furthermore, magnesium deficiency often leads to refractory hypokalemia because of the release of magnesium-mediated inhibition of ROMK, which promotes potassium secretion into urinary lumen [183,184].
Additional pharmacodynamic concerns include additive QT prolongation and hormone-related effects of azole antifungals. However, these largely overlap with the intrinsic toxic profiles described earlier.

8. Discussion

The management of oral and esophageal candidiasis extends beyond symptom relief to influence long-term prognosis, particularly in immunocompromised patients. Although prognosis is generally favorable with appropriate treatment, recurrence occurs in approximately 20% of patients, and systemic dissemination can be life-threatening. Therefore, the pharmacological issues described in this review, such as drug–drug interactions, adverse effect profiles, and interindividual variability in drug metabolism, are critical determinants of treatment outcomes.
For example, drug–drug interactions with antifungals may lead to supratherapeutic drug concentrations or toxicity by immunosuppressants, anticoagulants, or chemotherapeutic agents, and increase the risk of treatment failure and Candida recurrence. Similarly, the impact of CYP2C19 polymorphisms on voriconazole metabolism underscores how genetic variability can affect both efficacy and safety. These findings suggest that antifungal therapy should be individualized, taking into account comorbidities, concomitant medications, and genetic background, to improve prognosis and prevent refractory or disseminated infections.
Antifungal resistance is a significant factor affecting prognosis. Repeated or prolonged azole exposure, particularly in immunocompromised hosts, has contributed to the increasing prevalence of fluconazole-resistant species, such as C. glabrata, and intrinsically resistant species, such as C. krusei [185]. Resistance not only compromises the efficacy of first-line agents but also necessitates the use of broader spectrum or more toxic alternatives, which may increase the risk of drug interaction and adverse events. Thus, antifungal susceptibility patterns and timely adjustments to therapy are essential for preventing treatment failure and improving long-term outcomes.
The effective management of oral candidiasis requires a multidisciplinary collaborative approach. Physicians assess and manage systemic risk factors, such as immune deficiency and diabetes. They also guide decisions regarding systemic antifungal therapy. Dental professionals confirm the diagnosis through intraoral examination and address local predisposing factors such as ill-fitting dentures and xerostomia. Mechanical disruption of Candida biofilms via professional denture cleaning and oral care is essential to enhance antifungal efficacy [186]. Pharmacists play a key role in ensuring the safe and effective use of antifungal agents, especially azole antifungals, which can cause complex drug interactions [187]. Prescription verification, drug–drug interaction monitoring, and patient counseling help optimize therapeutic outcomes.
In addition to the pharmacological considerations discussed above, emerging antifungal resistance and the increasing complexity of polypharmacy highlight the need for more individualized treatment strategies. Despite the availability of guidelines, limited attention has been paid to pharmacogenetic variability and real-world drug–drug interactions, particularly in elderly and immunocompromised populations. Future research should integrate pharmacokinetic–pharmacodynamic modeling, therapeutic drug monitoring, and pharmacogenomic testing to optimize antifungal exposure and minimize toxicity. Moreover, interprofessional collaboration among physicians, dentists, and pharmacists is critical not only for preventing recurrent or refractory infections but also for developing personalized antifungal stewardship frameworks. Such efforts are expected to bridge the current gap between pharmacological knowledge and clinical implementation.
Effective management of oral candidiasis requires both appropriate antifungal selection and seamless interdisciplinary collaboration among physicians, dentists, and pharmacists.

9. Conclusions

The optimal management of oral and esophageal candidiasis requires effective pharmacological therapy. Appropriate drug selection, careful monitoring of drug–drug interactions, and attention to toxicity are essential for therapeutic efficacy maximization and toxicity minimization. In combination with interindividual variability, including genetic polymorphisms that affect drug metabolism and transport, this underscores the need for personalized treatment strategies.
Although oral health care and multidisciplinary collaboration are necessary to support the treatment of oral and esophageal candidiasis, the central focus should be on the safe and effective use of antifungal agents. Future research should clarify how pharmacogenomic testing and therapeutic drug monitoring can be integrated into routine care to further improve prognosis in patients with oral and esophageal candidiasis.

Author Contributions

Conceptualization, T.H. and M.N.; writing—original draft preparation, T.H. and M.N.; writing—review and editing, T.H. and M.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Ethical review and approval were waived for this study due to Review Article.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable.

Acknowledgments

The figures were created using Servier Medical Art accessed on 1 September 2025. (https://smart.servier.com/).

Conflicts of Interest

The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; in the decision to publish the results.

Abbreviations

14-α demethylase: CYP51A1; cytochrome P450: CYP; P-glycoprotein: P-gp; Ether-à-go-go–Related Gene: hERG; prothrombin time international normalized ratio: PT-INR; Epidermal growth factor receptor: EGFR.

References

  1. Jørgensen, M.R. Pathophysiological microenvironments in oral candidiasis. APMIS 2024, 132, 956–973. [Google Scholar] [CrossRef] [PubMed]
  2. Silva, S.; Negri, M.; Henriques, M.; Oliveira, R.; Williams, D.W.; Azeredo, J. Candida glabrata, Candida parapsilosis and Candida tropicalis: Biology, epidemiology, pathogenicity and antifungal resistance. FEMS Microbiol. Rev. 2012, 36, 288–305. [Google Scholar] [CrossRef] [PubMed]
  3. Pappas, P.G.; Lionakis, M.S.; Arendrup, M.C.; Ostrosky-Zeichner, L.; Kullberg, B.J. Invasive candidiasis. Nat. Rev. Dis. Primers 2018, 4, 18026. [Google Scholar] [CrossRef] [PubMed]
  4. Singh, A.; Verma, R.; Murari, A.; Agrawal, A. Oral candidiasis: An overview. J. Oral Maxillofac. Pathol. 2014, 18, S81–S85. [Google Scholar] [CrossRef]
  5. Mohamed, A.A.; Lu, X.L.; Mounmin, F.A. Diagnosis and Treatment of Esophageal Candidiasis: Current Updates. Can. J. Gastroenterol. Hepatol. 2019, 2019, 3585136. [Google Scholar] [CrossRef]
  6. Talapko, J.; Juzbašić, M.; Matijević, T.; Pustijanac, E.; Bekić, S.; Kotris, I.; Škrlec, I. Candida albicans-The Virulence Factors and Clinical Manifestations of Infection. J. Fungi 2021, 7, 79. [Google Scholar] [CrossRef]
  7. Patil, S.; Rao, R.S.; Majumdar, B.; Anil, S. Clinical Appearance of Oral Candida Infection and Therapeutic Strategies. Front. Microbiol. 2015, 6, 1391. [Google Scholar] [CrossRef]
  8. Coronado-Castellote, L.; Jiménez-Soriano, Y. Clinical and microbiological diagnosis of oral candidiasis. J. Clin. Exp. Dent. 2013, 5, e279–e286. [Google Scholar] [CrossRef]
  9. Rich, A.M.; Hussaini, H.M.; Nizar, M.A.M.; Gavidi, R.O.; Tauati-Williams, E.; Yakin, M.; Seo, B. Diagnosis of oral potentially malignant disorders: Overview and experience in Oceania. Front. Oral Health 2023, 4, 1122497. [Google Scholar] [CrossRef]
  10. Sims, C.R.; Ostrosky-Zeichner, L.; Rex, J.H. Invasive candidiasis in immunocompromised hospitalized patients. Arch. Med. Res. 2005, 36, 660–671. [Google Scholar] [CrossRef]
  11. Dropulic, L.K.; Lederman, H.M. Overview of Infections in the Immunocompromised Host. Microbiol. Spectrum. 2016, 4. [Google Scholar] [CrossRef]
  12. Fidel, P.L., Jr. Candida-host interactions in HIV disease: Implications for oropharyngeal candidiasis. Adv. Dent. Res. 2011, 23, 45–49. [Google Scholar] [CrossRef]
  13. Teoh, F.; Pavelka, N. How Chemotherapy Increases the Risk of Systemic Candidiasis in Cancer Patients: Current Paradigm and Future Directions. Pathogens 2016, 5, 6. [Google Scholar] [CrossRef]
  14. Pappas, P.G.; Silveira, F.P. Candida in solid organ transplant recipients. Am. J. Transplant. 2009, 9 (Suppl. 4), S173–S179. [Google Scholar] [CrossRef]
  15. Perić, M.; Miličić, B.; Kuzmanović Pfićer, J.; Živković, R.; Arsić Arsenijević, V. A Systematic Review of Denture Stomatitis: Predisposing Factors, Clinical Features, Etiology, and Global Candida spp. Distribution. J. Fungi 2024, 10, 328. [Google Scholar] [CrossRef] [PubMed]
  16. Tarapan, S.; Matangkasombut, O.; Trachootham, D.; Sattabanasuk, V.; Talungchit, S.; Paemuang, W.; Phonyiam, T.; Chokchaitam, O.; Mungkung, O.O.; Lam-Ubol, A. Oral Candida colonization in xerostomic postradiotherapy head and neck cancer patients. Oral Dis. 2019, 25, 1798–1808. [Google Scholar] [CrossRef] [PubMed]
  17. Goh, E.Z.; Beech, N.; Johnson, N.R.; Batstone, M. The dental management of patients irradiated for head and neck cancer. Br. Dent. J. 2023, 234, 800–804. [Google Scholar] [CrossRef] [PubMed]
  18. Serrano, J.; Lopez-Pintor, R.M.; Gonzalez-Serrano, J.; Fernandez-Castro, M.; Casanas, E.; Hernandez, G. Oral lesions in Sjogren’s syndrome: A systematic review. Med. Oral Patol. Oral Cir. Bucal 2018, 23, e391–e400. [Google Scholar] [CrossRef]
  19. Musinguzi, B.; Obuku, E.A.; Mwesigwa, A.; Migisha, R.; Kinengyere, A.A.; Ndagire, R.; Baguma, A.; Okek, E.J.; Olum, R.; Itabangi, H.; et al. Distribution of Candida species isolated from people living with human immunodeficiency virus with oropharyngeal and oral candidiasis in Africa in the era of universal test and treat policy: A systematic review and meta-analysis. Trop. Med. Health 2024, 52, 88. [Google Scholar] [CrossRef]
  20. Bassetti, M.; Vena, A.; Meroi, M.; Cardozo, C.; Cuervo, G.; Giacobbe, D.R.; Salavert, M.; Merino, P.; Gioia, F.; Fernández-Ruiz, M.; et al. Factors associated with the development of septic shock in patients with candidemia: A post hoc analysis from two prospective cohorts. Crit. Care 2020, 24, 117. [Google Scholar] [CrossRef]
  21. Mazi, P.B.; Olsen, M.A.; Stwalley, D.; Rauseo, A.M.; Ayres, C.; Powderly, W.G.; Spec, A. Attributable Mortality of Candida Bloodstream Infections in the Modern Era: A Propensity Score Analysis. Clin. Infect. Dis. 2022, 75, 1031–1036. [Google Scholar] [CrossRef]
  22. Antinori, S.; Milazzo, L.; Sollima, S.; Galli, M.; Corbellino, M. Candidemia and invasive candidiasis in adults: A narrative review. Eur. J. Intern. Med. 2016, 34, 21–28. [Google Scholar] [CrossRef]
  23. Darwazeh, A.M.G.; Darwazeh, T.A. What Makes Oral Candidiasis Recurrent Infection? A Clinical View. J. Mycol. 2014, 2014, 758394. [Google Scholar] [CrossRef]
  24. Jackson, K.D.; Achour, B.; Lee, J.; Geffert, R.M.; Beers, J.L.; Latham, B.D. Novel Approaches to Characterize Individual Drug Metabolism and Advance Precision Medicine. Drug Metab. Dispos. 2023, 51, 1238–1253. [Google Scholar] [CrossRef]
  25. Rawson, T.M.; Wilson, R.C.; O’Hare, D.; Herrero, P.; Kambugu, A.; Lamorde, M.; Ellington, M.; Georgiou, P.; Cass, A.; Hope, W.W.; et al. Optimizing antimicrobial use: Challenges, advances and opportunities. Nat. Rev. Microbiol. 2021, 19, 747–758. [Google Scholar] [CrossRef] [PubMed]
  26. Carmo, A.; Rocha, M.; Pereirinha, P.; Tomé, R.; Costa, E. Antifungals: From Pharmacokinetics to Clinical Practice. Antibiotics 2023, 12, 884. [Google Scholar] [CrossRef]
  27. Silva, S.; Rodrigues, C.F.; Araújo, D.; Rodrigues, M.E.; Henriques, M. Candida Species Biofilms’ Antifungal Resistance. J. Fungi 2017, 3, 8. [Google Scholar] [CrossRef] [PubMed]
  28. Kaur, J.; Nobile, C.J. Antifungal drug-resistance mechanisms in Candida biofilms. Curr. Opin. Microbiol. 2023, 71, 102237. [Google Scholar] [CrossRef] [PubMed]
  29. Pappas, P.G.; Kauffman, C.A.; Andes, D.R.; Clancy, C.J.; Marr, K.A.; Ostrosky-Zeichner, L.; Annette, C.R.; Mindy, G.S.; Jose, A.V.; Thomas, J.W.; et al. Clinical Practice Guideline for the Management of Candidiasis: 2016 Update by the Infectious Diseases Society of America. Clin. Infect. Dis. 2016, 62, e1–e50. [Google Scholar] [CrossRef]
  30. Elad, S.; Cheng, K.K.F.; Lalla, R.V.; Yarom, N.; Hong, C.; Logan, R.M.; Bowen, J.; Gibson, R.; Saunders, D.P.; Zadik, Y.; et al. MASCC/ISOO clinical practice guidelines for the management of mucositis secondary to cancer therapy. Cancer 2020, 126, 4423–4431. [Google Scholar] [CrossRef]
  31. Patel, M. Oral Cavity and Candida albicans: Colonisation to the Development of Infection. Pathogens 2022, 11, 335. [Google Scholar] [CrossRef] [PubMed]
  32. Felton, D.; Cooper, L.; Duqum, I.; Minsley, G.; Guckes, A.; Haug, S.; Meredith, P.; Solie, C.; Avery, D.; Deal Chandler, N.; et al. Evidence-based guidelines for the care and maintenance of complete dentures: A publication of the American College of Prosthodontists. J. Prosthodont. 2011, 20 (Suppl. 1), S1–S12. [Google Scholar] [CrossRef]
  33. Mylonas, P.; Milward, P.; McAndrew, R. Denture cleanliness and hygiene: An overview. Br. Dent. J. 2022, 233, 20–26. [Google Scholar] [CrossRef]
  34. Epstein, J.B.; Emerton, S.; Le, N.D.; Stevenson-Moore, P. A double-blind crossover trial of Oral Balance gel and Biotene toothpaste versus placebo in patients with xerostomia following radiation therapy. Oral Oncol. 1999, 35, 132–137. [Google Scholar] [CrossRef]
  35. Sugimoto, J.; Kanehira, T.; Mizugai, H.; Chiba, I.; Morita, M. Relationship between salivary histatin 5 levels and Candida CFU counts in healthy elderly. Gerodontology 2006, 23, 164–169. [Google Scholar] [CrossRef]
  36. Edgerton, M.; Koshlukova, S.E. Salivary histatin 5 and its similarities to the other antimicrobial proteins in human saliva. Adv. Dent. Res. 2000, 14, 16–21. [Google Scholar] [CrossRef] [PubMed]
  37. Mathews, S.A.; Kurien, B.T.; Scofield, R.H. Oral manifestations of Sjögren’s syndrome. J. Dent. Res. 2008, 87, 308–318. [Google Scholar] [CrossRef]
  38. Lu, S.Y. Oral Candidosis: Pathophysiology and Best Practice for Diagnosis, Classification, and Successful Management. J. Fungi 2021, 7, 555. [Google Scholar] [CrossRef]
  39. Martori, E.; Ayuso-Montero, R.; Willaert, E.; Viñas, M.; Peraire, M.; Martinez-Gomis, J. Status of Removable Dentures and Relationship with Oral Candida-Associated Factors in a Geriatric Population in Catalonia. J. Prosthodont. 2017, 26, 370–375. [Google Scholar] [CrossRef] [PubMed]
  40. Compagnoni, M.A.; Souza, R.F.; Marra, J.; Pero, A.C.; Barbosa, D.B. Relationship between Candida and nocturnal denture wear: Quantitative study. J. Oral Rehabil. 2007, 34, 600–605. [Google Scholar] [CrossRef]
  41. Stoopler, E.T.; Villa, A.; Bindakhil, M.; Díaz, D.L.O.; Sollecito, T.P. Common Oral Conditions: A Review. Jama 2024, 331, 1045–1054. [Google Scholar] [CrossRef]
  42. Wolff, A.; Joshi, R.K.; Ekström, J.; Aframian, D.; Pedersen, A.M.; Proctor, G.; Narayana, N.; Villa, A.; Sia, Y.W.; Aliko, A.; et al. A Guide to Medications Inducing Salivary Gland Dysfunction, Xerostomia, and Subjective Sialorrhea: A Systematic Review Sponsored by the World Workshop on Oral Medicine VI. Drugs R D 2017, 17, 1–28. [Google Scholar] [CrossRef]
  43. Mercadante, V.; Jensen, S.B.; Smith, D.K.; Bohlke, K.; Bauman, J.; Brennan, M.T.; Coppes, R.P.; Jessen, N.; Malhotra, N.K.; Murphy, B.; et al. Salivary Gland Hypofunction and/or Xerostomia Induced by Nonsurgical Cancer Therapies: ISOO/MASCC/ASCO Guideline. J. Clin. Oncol. 2021, 39, 2825–2843. [Google Scholar] [CrossRef] [PubMed]
  44. Minakuchi, S.; Tsuga, K.; Ikebe, K.; Ueda, T.; Tamura, F.; Nagao, K.; Furuya, J.; Matsuo, K.; Yamamoto, K.; Kanazawa, M.; et al. Oral hypofunction in the older population: Position paper of the Japanese Society of Gerodontology in 2016. Gerodontology 2018, 35, 317–324. [Google Scholar] [CrossRef]
  45. Yap, B.S.; Bodey, G.P. Oropharyngeal candidiasis treated with a troche form of clotrimazole. Arch. Intern. Med. 1979, 139, 656–657. [Google Scholar] [CrossRef]
  46. Van Roey, J.; Haxaire, M.; Kamya, M.; Lwanga, I.; Katabira, E. Comparative efficacy of topical therapy with a slow-release mucoadhesive buccal tablet containing miconazole nitrate versus systemic therapy with ketoconazole in HIV-positive patients with oropharyngeal candidiasis. J. Acquir. Immune Defic. Syndr. 2004, 35, 144–150. [Google Scholar] [CrossRef]
  47. Vazquez, J.A.; Patton, L.L.; Epstein, J.B.; Ramlachan, P.; Mitha, I.; Noveljic, Z.; Fourie, J.; Conway, B.; Lalla, R.V.; Barasch, A.; et al. Randomized, comparative, double-blind, double-dummy, multicenter trial of miconazole buccal tablet and clotrimazole troches for the treatment of oropharyngeal candidiasis: Study of miconazole Lauriad® efficacy and safety (SMiLES). HIV Clin. Trials 2010, 11, 186–196. [Google Scholar] [CrossRef] [PubMed]
  48. Patton, L.L.; Bonito, A.J.; Shugars, D.A. A systematic review of the effectiveness of antifungal drugs for the prevention and treatment of oropharyngeal candidiasis in HIV-positive patients. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 2001, 92, 170–179. [Google Scholar] [CrossRef] [PubMed]
  49. Finlay, P.M.; Richardson, M.D.; Robertson, A.G. A comparative study of the efficacy of fluconazole and amphotericin B in the treatment of oropharyngeal candidosis in patients undergoing radiotherapy for head and neck tumours. Br. J. Oral Maxillofac. Surg. 1996, 34, 23–25. [Google Scholar] [CrossRef]
  50. Pons, V.; Greenspan, D.; Lozada-Nur, F.; McPhail, L.; Gallant, J.E.; Tunkel, A.; Johnson, C.C.; McCarty, J.; Panzer, H.; Levenstein, M.; et al. Oropharyngeal candidiasis in patients with AIDS: Randomized comparison of fluconazole versus nystatin oral suspensions. Clin. Infect. Dis. 1997, 24, 1204–1207. [Google Scholar] [CrossRef]
  51. Koletar, S.L.; Russell, J.A.; Fass, R.J.; Plouffe, J.F. Comparison of oral fluconazole and clotrimazole troches as treatment for oral candidiasis in patients infected with human immunodeficiency virus. Antimicrob. Agents Chemother. 1990, 34, 2267–2268. [Google Scholar] [CrossRef] [PubMed]
  52. Saag, M.S.; Fessel, W.J.; Kaufman, C.A.; Merrill, K.W.; Ward, D.J.; Moskovitz, B.L.; Thomas, C.; Oleka, N.; Guarnieri, J.A.; Lee, J.; et al. Treatment of fluconazole-refractory oropharyngeal candidiasis with itraconazole oral solution in HIV-positive patients. AIDS Res. Hum. Retroviruses 1999, 15, 1413–1417. [Google Scholar] [CrossRef]
  53. Skiest, D.J.; Vazquez, J.A.; Anstead, G.M.; Graybill, J.R.; Reynes, J.; Ward, D.; Hare, R.; Boparai, N.; Isaacs, R. Posaconazole for the treatment of azole-refractory oropharyngeal and esophageal candidiasis in subjects with HIV infection. Clin. Infect. Dis. 2007, 44, 607–614. [Google Scholar] [CrossRef]
  54. Hegener, P.; Troke, P.F.; Fätkenheuer, G.; Diehl, V.; Ruhnke, M. Treatment of fluconazole-resistant candidiasis with voriconazole in patients with AIDS. Aids 1998, 12, 2227–2228. [Google Scholar]
  55. Lake, D.E.; Kunzweiler, J.; Beer, M.; Buell, D.N.; Islam, M.Z. Fluconazole versus amphotericin B in the treatment of esophageal candidiasis in cancer patients. Chemotherapy 1996, 42, 308–314. [Google Scholar] [CrossRef]
  56. Kristanc, L.; Božič, B.; Jokhadar, Š.Z.; Dolenc, M.S.; Gomišček, G. The pore-forming action of polyenes: From model membranes to living organisms. Biochim. Biophys. Acta Biomembr. 2019, 1861, 418–430. [Google Scholar] [CrossRef]
  57. Ramos, H.; Valdivieso, E.; Gamargo, M.; Dagger, F.; Cohen, B.E. Amphotericin B kills unicellular leishmanias by forming aqueous pores permeable to small cations and anions. J. Membr. Biol. 1996, 152, 65–75. [Google Scholar] [CrossRef]
  58. Brüggemann, R.J.; Jensen, G.M.; Lass-Flörl, C. Liposomal amphotericin B—The past. J. Antimicrob. Chemother. 2022, 77 (Suppl. 2), ii3–ii10. [Google Scholar] [CrossRef]
  59. Ghannoum, M.A.; Rice, L.B. Antifungal agents: Mode of action, mechanisms of resistance, and correlation of these mechanisms with bacterial resistance. Clin. Microbiol. Rev. 1999, 12, 501–517. [Google Scholar] [CrossRef] [PubMed]
  60. Sheehan, D.J.; Hitchcock, C.A.; Sibley, C.M. Current and emerging azole antifungal agents. Clin. Microbiol. Rev. 1999, 12, 40–79. [Google Scholar] [CrossRef] [PubMed]
  61. Szymański, M.; Chmielewska, S.; Czyżewska, U.; Malinowska, M.; Tylicki, A. Echinocandins—Structure, mechanism of action and use in antifungal therapy. J. Enzyme Inhib. Med. Chem. 2022, 37, 876–894. [Google Scholar] [CrossRef]
  62. Subirà, M.; Martino, R.; Gómez, L.; Martí, J.M.; Estany, C.; Sierra, J. Low-dose amphotericin B lipid complex vs. conventional amphotericin B for empirical antifungal therapy of neutropenic fever in patients with hematologic malignancies—A randomized, controlled trial. Eur. J. Haematol. 2004, 72, 342–347. [Google Scholar] [CrossRef]
  63. Haro-Reyes, T.; Díaz-Peralta, L.; Galván-Hernández, A.; Rodríguez-López, A.; Rodríguez-Fragoso, L.; Ortega-Blake, I. Polyene Antibiotics Physical Chemistry and Their Effect on Lipid Membranes; Impacting Biological Processes and Medical Applications. Membranes 2022, 12, 681. [Google Scholar] [CrossRef]
  64. Mroczyńska, M.; Brillowska-Dąbrowska, A. Review on Current Status of Echinocandins Use. Antibiotics 2020, 9, 227. [Google Scholar] [CrossRef]
  65. Bellmann, R.; Smuszkiewicz, P. Pharmacokinetics of antifungal drugs: Practical implications for optimized treatment of patients. Infection 2017, 45, 737–779. [Google Scholar] [CrossRef] [PubMed]
  66. Hope, W.W.; Drusano, G.L. Antifungal pharmacokinetics and pharmacodynamics: Bridging from the bench to bedside. Clin. Microbiol. Infect. 2009, 15, 602–612. [Google Scholar] [CrossRef] [PubMed]
  67. Gómez-López, A. Antifungal therapeutic drug monitoring: Focus on drugs without a clear recommendation. Clin. Microbiol. Infect. 2020, 26, 1481–1487. [Google Scholar] [CrossRef] [PubMed]
  68. Laniado-Laborín, R.; Cabrales-Vargas, M.N. Amphotericin B: Side effects and toxicity. Rev. Iberoam. Micol. 2009, 26, 223–227. [Google Scholar] [CrossRef]
  69. Stone, N.R.; Bicanic, T.; Salim, R.; Hope, W. Liposomal Amphotericin B (AmBisome(®)): A Review of the Pharmacokinetics, Pharmacodynamics, Clinical Experience and Future Directions. Drugs 2016, 76, 485–500. [Google Scholar] [CrossRef]
  70. Bekersky, I.; Fielding, R.M.; Dressler, D.E.; Lee, J.W.; Buell, D.N.; Walsh, T.J. Pharmacokinetics, excretion, and mass balance of liposomal amphotericin B (AmBisome) and amphotericin B deoxycholate in humans. Antimicrob. Agents Chemother. 2002, 46, 828–833. [Google Scholar] [CrossRef]
  71. Falci, D.R.; Pasqualotto, A.C. Profile of isavuconazole and its potential in the treatment of severe invasive fungal infections. Infect. Drug Resist. 2013, 6, 163–174. [Google Scholar] [CrossRef]
  72. Heykants, J.; Van Peer, A.; Van de Velde, V.; Van Rooy, P.; Meuldermans, W.; Lavrijsen, K.; Woestenborghs, R.; Van Cutsem, J.; Cauwenbergh, G. The clinical pharmacokinetics of itraconazole: An overview. Mycoses 1989, 32 (Suppl. 1), 67–87. [Google Scholar] [CrossRef]
  73. Lewis, J.S., II; Wiederhold, N.P.; Hakki, M.; Thompson, G.R., III. New Perspectives on Antimicrobial Agents: Isavuconazole. Antimicrob. Agents Chemother. 2022, 66, e0017722. [Google Scholar] [CrossRef]
  74. Brüggemann, R.J.; Alffenaar, J.W.; Blijlevens, N.M.; Billaud, E.M.; Kosterink, J.G.; Verweij, P.E.; Burger, D.M. Clinical relevance of the pharmacokinetic interactions of azole antifungal drugs with other coadministered agents. Clin. Infect. Dis. 2009, 48, 1441–1458. [Google Scholar] [CrossRef]
  75. Lee, S.; Kim, B.H.; Nam, W.S.; Yoon, S.H.; Cho, J.Y.; Shin, S.G.; Jang, I.J.; Yu, K.S. Effect of CYP2C19 polymorphism on the pharmacokinetics of voriconazole after single and multiple doses in healthy volunteers. J. Clin. Pharmacol. 2012, 52, 195–203. [Google Scholar] [CrossRef]
  76. Lipp, H.P. Clinical pharmacodynamics and pharmacokinetics of the antifungal extended-spectrum triazole posaconazole: An overview. Br. J. Clin. Pharmacol. 2010, 70, 471–480. [Google Scholar] [CrossRef]
  77. Perlin, D.S. Current perspectives on echinocandin class drugs. Future Microbiol. 2011, 6, 441–457. [Google Scholar] [CrossRef]
  78. Wiederhold, N.P.; Lewis, J.S., II. The echinocandin micafungin: A review of the pharmacology, spectrum of activity, clinical efficacy and safety. Expert Opin. Pharmacother. 2007, 8, 1155–1166. [Google Scholar] [CrossRef]
  79. Gigliotti, F.; Shenep, J.L.; Lott, L.; Thornton, D. Induction of prostaglandin synthesis as the mechanism responsible for the chills and fever produced by infusing amphotericin B. J. Infect. Dis. 1987, 156, 784–789. [Google Scholar] [CrossRef]
  80. Goodwin, S.D.; Cleary, J.D.; Walawander, C.A.; Taylor, J.W.; Grasela, T.H., Jr. Pretreatment regimens for adverse events related to infusion of amphotericin B. Clin. Infect. Dis. 1995, 20, 755–761. [Google Scholar] [CrossRef]
  81. Paterson, D.L.; David, K.; Mrsic, M.; Cetkovsky, P.; Weng, X.H.; Sterba, J.; Krivan, G.; Boskovic, D.; Lu, M.; Zhu, L.P.; et al. Pre-medication practices and incidence of infusion-related reactions in patients receiving AMPHOTEC: Data from the Patient Registry of Amphotericin B Cholesteryl Sulfate Complex for Injection Clinical Tolerability (PRoACT) registry. J. Antimicrob. Chemother. 2008, 62, 1392–1400. [Google Scholar] [CrossRef]
  82. Sabra, R.; Branch, R.A. Amphotericin B nephrotoxicity. Drug Saf. 1990, 5, 94–108. [Google Scholar] [CrossRef]
  83. Adler-Moore, J.; Lewis, R.E.; Brüggemann, R.J.M.; Rijnders, B.J.A.; Groll, A.H.; Walsh, T.J. Preclinical Safety, Tolerability, Pharmacokinetics, Pharmacodynamics, and Antifungal Activity of Liposomal Amphotericin, B. Clin. Infect. Dis. 2019, 68, S244–S259. [Google Scholar] [CrossRef]
  84. Rosner, M.H. Polyuria in a Patient with Aspergillus Infection. Clin. J. Am. Soc. Nephrol. 2017, 12, 1343–1346. [Google Scholar] [CrossRef]
  85. Vallés, P.G.; Batlle, D. Hypokalemic Distal Renal Tubular Acidosis. Adv. Chronic Kidney Dis. 2018, 25, 303–320. [Google Scholar] [CrossRef] [PubMed]
  86. Benitez, L.L.; Carver, P.L. Adverse Effects Associated with Long-Term Administration of Azole Antifungal Agents. Drugs 2019, 79, 833–853. [Google Scholar] [CrossRef]
  87. Tan, K.; Brayshaw, N.; Tomaszewski, K.; Troke, P.; Wood, N. Investigation of the potential relationships between plasma voriconazole concentrations and visual adverse events or liver function test abnormalities. J. Clin. Pharmacol. 2006, 46, 235–243. [Google Scholar] [CrossRef]
  88. Jin, H.; Wang, T.; Falcione, B.A.; Olsen, K.M.; Chen, K.; Tang, H.; Hui, J.; Zhai, S. Trough concentration of voriconazole and its relationship with efficacy and safety: A systematic review and meta-analysis. J. Antimicrob. Chemother. 2016, 71, 1772–1785. [Google Scholar] [CrossRef]
  89. Saad, A.H.; DePestel, D.D.; Carver, P.L. Factors influencing the magnitude and clinical significance of drug interactions between azole antifungals and select immunosuppressants. Pharmacotherapy 2006, 26, 1730–1744. [Google Scholar] [CrossRef]
  90. Balcerek, M.I.; Stewart, A.G.; Chapman, P.; Lazarus, S. Reducing the off-target endocrinologic adverse effects of azole antifungals-can it be done? Int. J. Antimicrob. Agents 2022, 59, 106587. [Google Scholar] [CrossRef]
  91. Sung, D.J.; Kim, J.G.; Won, K.J.; Kim, B.; Shin, H.C.; Park, J.Y.; Bae, Y.M. Blockade of K+ and Ca2+ channels by azole antifungal agents in neonatal rat ventricular myocytes. Biol. Pharm. Bull. 2012, 35, 1469–1475. [Google Scholar] [CrossRef]
  92. Owens, R.C., Jr.; Nolin, T.D. Antimicrobial-associated QT interval prolongation: Pointes of interest. Clin. Infect. Dis. 2006, 43, 1603–1611. [Google Scholar] [CrossRef]
  93. Keirns, J.; Desai, A.; Kowalski, D.; Lademacher, C.; Mujais, S.; Parker, B.; Schneidkraut, M.J.; Townsend, R.; Wojtkowski, T.; Yamazaki, T.; et al. QT Interval Shortening with Isavuconazole: In Vitro and In Vivo Effects on Cardiac Repolarization. Clin. Pharmacol. Ther. 2017, 101, 782–790. [Google Scholar] [CrossRef]
  94. Lewis, R.E. Current concepts in antifungal pharmacology. Mayo Clin. Proc. 2011, 86, 805–817. [Google Scholar] [CrossRef]
  95. Yang, L.; Wang, C.; Zhang, Y.; Wang, Q.; Qiu, Y.; Li, S.; Yang, B.; Du, Q.; Chen, J.; Teng, M.; et al. Central Nervous System Toxicity of Voriconazole: Risk Factors and Threshold—A Retrospective Cohort Study. Infect. Drug Resist. 2022, 15, 7475–7484. [Google Scholar] [CrossRef]
  96. Aguilar-Zapata, D.; Petraitiene, R.; Petraitis, V. Echinocandins: The Expanding Antifungal Armamentarium. Clin. Infect. Dis. 2015, 61 (Suppl. 6), S604–S611. [Google Scholar] [CrossRef] [PubMed]
  97. Grover, N.D. Echinocandins: A ray of hope in antifungal drug therapy. Indian J. Pharmacol. 2010, 42, 9–11. [Google Scholar] [CrossRef]
  98. Denning, D.W. Echinocandin antifungal drugs. Lancet 2003, 362, 1142–1151. [Google Scholar] [CrossRef]
  99. Kyriakidis, I.; Tragiannidis, A.; Munchen, S.; Groll, A.H. Clinical hepatotoxicity associated with antifungal agents. Expert Opin. Drug Saf. 2017, 16, 149–165. [Google Scholar] [CrossRef] [PubMed]
  100. Song, J.C.; Deresinski, S. Hepatotoxicity of antifungal agents. Curr. Opin. Investig. Drugs 2005, 6, 170–177. [Google Scholar] [PubMed]
  101. Song, Y.; Li, C.; Liu, G.; Liu, R.; Chen, Y.; Li, W.; Cao, Z.; Zhao, B.; Lu, C.; Liu, Y. Drug-Metabolizing Cytochrome P450 Enzymes Have Multifarious Influences on Treatment Outcomes. Clin. Pharmacokinet. 2021, 60, 585–601. [Google Scholar] [CrossRef]
  102. Zhang, H.F.; Wang, H.H.; Gao, N.; Wei, J.Y.; Tian, X.; Zhao, Y.; Fang, Y.; Zhou, J.; Wen, Q.; Gao, J.; et al. Physiological Content and Intrinsic Activities of 10 Cytochrome P450 Isoforms in Human Normal Liver Microsomes. J. Pharmacol. Exp. Ther. 2016, 358, 83–93. [Google Scholar] [CrossRef]
  103. Paine, M.F.; Hart, H.L.; Ludington, S.S.; Haining, R.L.; Rettie, A.E.; Zeldin, D.C. The human intestinal cytochrome P450 “pie”. Drug Metab. Dispos. 2006, 34, 880–886. [Google Scholar] [CrossRef]
  104. König, J.; Müller, F.; Fromm, M.F. Transporters and drug-drug interactions: Important determinants of drug disposition and effects. Pharmacol. Rev. 2013, 65, 944–966. [Google Scholar] [CrossRef]
  105. Bruckmueller, H.; Cascorbi, I. Drug-Drug-Gene Interactions: A Call for Clinical Consideration. Clin. Pharmacol. Ther. 2021, 110, 549–551. [Google Scholar] [CrossRef] [PubMed]
  106. Malki, M.A.; Pearson, E.R. Drug–drug–gene interactions and adverse drug reactions. Pharmacogenomics J. 2020, 20, 355–366. [Google Scholar] [CrossRef]
  107. Brunet, M.; van Gelder, T.; Åsberg, A.; Haufroid, V.; Hesselink, D.A.; Langman, L.; Lemaitre, F.; Marquet, P.; Seger, C.; Shipkova, M.; et al. Therapeutic Drug Monitoring of Tacrolimus-Personalized Therapy: Second Consensus Report. Ther. Drug Monit. 2019, 41, 261–307. [Google Scholar] [CrossRef] [PubMed]
  108. Huppertz, A.; Ott, C.; Bruckner, T.; Foerster, K.I.; Burhenne, J.; Weiss, J.; Zorn, M.; Haefeli, W.E.; Czock, D. Prolonged-Release Tacrolimus Is Less Susceptible to Interaction with the Strong CYP3A Inhibitor Voriconazole in Healthy Volunteers. Clin. Pharmacol Ther. 2019, 106, 1290–1298. [Google Scholar] [CrossRef]
  109. Venkataramanan, R.; Zang, S.; Gayowski, T.; Singh, N. Voriconazole inhibition of the metabolism of tacrolimus in a liver transplant recipient and in human liver microsomes. Antimicrob. Agents Chemother. 2002, 46, 3091–3093. [Google Scholar] [CrossRef]
  110. Huang, X.; Zhou, Y.; Zhang, J.; Xiang, H.; Mei, H.; Liu, L.; Tong, L.; Zeng, F.; Huang, Y.; Zhou, H.; et al. The importance of CYP2C19 genotype in tacrolimus dose optimization when concomitant with voriconazole in heart transplant recipients. Br. J. Clin. Pharmacol. 2022, 88, 4515–4525. [Google Scholar] [CrossRef] [PubMed]
  111. Imamura, C.K.; Furihata, K.; Okamoto, S.; Tanigawara, Y. Impact of cytochrome P450 2C19 polymorphisms on the pharmacokinetics of tacrolimus when coadministered with voriconazole. J. Clin. Pharmacol. 2016, 56, 408–413. [Google Scholar] [CrossRef]
  112. Lewis, R.; Niazi-Ali, S.; McIvor, A.; Kanj, S.S.; Maertens, J.; Bassetti, M.; Levine, D.; Groll, A.H.; Denning, D.W. Triazole antifungal drug interactions-practical considerations for excellent prescribing. J. Antimicrob. Chemother. 2024, 79, 1203–1217. [Google Scholar] [CrossRef]
  113. Dodds-Ashley, E. Management of drug and food interactions with azole antifungal agents in transplant recipients. Pharmacotherapy 2010, 30, 842–854. [Google Scholar] [CrossRef] [PubMed]
  114. Isoherranen, N.; Kunze, K.L.; Allen, K.E.; Nelson, W.L.; Thummel, K.E. Role of itraconazole metabolites in CYP3A4 inhibition. Drug Metab. Dispos. 2004, 32, 1121–1131. [Google Scholar] [CrossRef]
  115. Liu, L.; Bello, A.; Dresser, M.J.; Heald, D.; Komjathy, S.F.; O’Mara, E.; Rogge, M.; Stoch, S.A.; Robertson, S.M. Best practices for the use of itraconazole as a replacement for ketoconazole in drug-drug interaction studies. J. Clin. Pharmacol. 2016, 56, 143–151. [Google Scholar] [CrossRef] [PubMed]
  116. Hirai, T.; Ino, K.; Ikejiri, M.; Tawara, I.; Iwamoto, T. Dual Inhibition of CYP3A4 by Voriconazole and Clarithromycin Influences Tacrolimus Pharmacokinetics: Case Series Study. Eur. J. Drug Metab. Pharmacokinet. 2022, 47, 889–893. [Google Scholar] [CrossRef]
  117. Nivoix, Y.; Levêque, D.; Herbrecht, R.; Koffel, J.C.; Beretz, L.; Ubeaud-Sequier, G. The enzymatic basis of drug-drug interactions with systemic triazole antifungals. Clin. Pharmacokinet. 2008, 47, 779–792. [Google Scholar] [CrossRef]
  118. Smith, H.S. Opioid metabolism. Mayo Clin. Proc. 2009, 84, 613–624. [Google Scholar] [CrossRef]
  119. Kinnunen, M.; Piirainen, P.; Kokki, H.; Lammi, P.; Kokki, M. Updated Clinical Pharmacokinetics and Pharmacodynamics of Oxycodone. Clin. Pharmacokinet. 2019, 58, 705–725. [Google Scholar] [CrossRef]
  120. Watanabe, M.; Homma, M.; Momo, K.; Okoshi, Y.; Wada, T.; Hara, A.; Chiba, S.; Kohda, Y. Effects of voriconazole co-administration on oxycodone-induced adverse events: A case in the retrospective survey. Eur. J. Clin. Pharmacol. 2011, 67, 859–861. [Google Scholar] [CrossRef] [PubMed]
  121. Saari, T.I.; Laine, K.; Neuvonen, M.; Neuvonen, P.J.; Olkkola, K.T. Effect of voriconazole and fluconazole on the pharmacokinetics of intravenous fentanyl. Eur. J. Clin. Pharmacol. 2008, 64, 25–30. [Google Scholar] [CrossRef] [PubMed]
  122. Wittkowsky, A.K. Warfarin and other coumarin derivatives: Pharmacokinetics, pharmacodynamics, and drug interactions. Semin. Vasc. Med. 2003, 3, 221–230. [Google Scholar] [CrossRef] [PubMed]
  123. Breckenridge, A.; Orme, M.; Wesseling, H.; Lewis, R.J.; Gibbons, R. Pharmacokinetics and pharmacodynamics of the enantiomers of warfarin in man. Clin. Pharmacol. Ther. 1974, 15, 424–430. [Google Scholar] [CrossRef] [PubMed]
  124. O’Reilly, R.A. Studies on the optical enantiomorphs of warfarin in man. Clin. Pharmacol. Ther. 1974, 16, 348–354. [Google Scholar] [CrossRef]
  125. Kaminsky, L.S.; Zhang, Z.Y. Human P450 metabolism of warfarin. Pharmacol. Ther. 1997, 73, 67–74. [Google Scholar] [CrossRef]
  126. Yamazaki, H.; Shimada, T. Human liver cytochrome P450 enzymes involved in the 7-hydroxylation of R- and S-warfarin enantiomers. Biochem. Pharmacol. 1997, 54, 1195–1203. [Google Scholar] [CrossRef]
  127. Schelleman, H.; Bilker, W.B.; Brensinger, C.M.; Han, X.; Kimmel, S.E.; Hennessy, S. Warfarin with fluoroquinolones, sulfonamides, or azole antifungals: Interactions and the risk of hospitalization for gastrointestinal bleeding. Clin. Pharmacol. Ther. 2008, 84, 581–588. [Google Scholar] [CrossRef]
  128. Black, D.J.; Kunze, K.L.; Wienkers, L.C.; Gidal, B.E.; Seaton, T.L.; McDonnell, N.D.; Evans, J.S.; Bauwens, J.E.; Trager, W.F. Warfarin-fluconazole. II. A metabolically based drug interaction: In vivo studies. Drug Metab. Dispos. 1996, 24, 422–428. [Google Scholar] [CrossRef]
  129. Purkins, L.; Wood, N.; Kleinermans, D.; Nichols, D. Voriconazole potentiates warfarin-induced prothrombin time prolongation. Br. J. Clin. Pharmacol. 2003, 56 (Suppl. 1), 24–29. [Google Scholar] [CrossRef]
  130. Pemberton, M.N.; Oliver, R.J.; Theaker, E.D. Miconazole oral gel and drug interactions. Br. Dent. J. 2004, 196, 529–531. [Google Scholar] [CrossRef]
  131. Naberhaus, T.; Jones, M.J.; Burns, A.; Raney, E.C. Topical Miconazole Cream and Warfarin Interaction: A Case Report. J. Pharm. Technol. 2022, 38, 127–129. [Google Scholar] [CrossRef] [PubMed]
  132. Ezsiás, A.; Wojnarowska, F.; Juniper, R. Topical use of miconazole antifungal oral gel on warfarinized patients: A word of caution. Dent. Update 1997, 24, 421–422. [Google Scholar]
  133. Hindley, B.; Lip, G.Y.H.; McCloskey, A.P.; Penson, P.E. Pharmacokinetics and pharmacodynamics of direct oral anticoagulants. Expert Opin. Drug Metab. Toxicol. 2023, 19, 911–923. [Google Scholar] [CrossRef]
  134. Foerster, K.I.; Hermann, S.; Mikus, G.; Haefeli, W.E. Drug-Drug Interactions with Direct Oral Anticoagulants. Clin. Pharmacokinet. 2020, 59, 967–980. [Google Scholar] [CrossRef]
  135. Brings, A.; Lehmann, M.L.; Foerster, K.I.; Burhenne, J.; Weiss, J.; Haefeli, W.E.; Czock, D. Perpetrator effects of ciclosporin (P-glycoprotein inhibitor) and its combination with fluconazole (CYP3A inhibitor) on the pharmacokinetics of rivaroxaban in healthy volunteers. Br. J. Clin. Pharmacol. 2019, 85, 1528–1537. [Google Scholar] [CrossRef] [PubMed]
  136. Terrier, J.; Gaspar, F.; Fontana, P.; Youssef, D.; Reny, J.L.; Csajka, C.; Samer, C.F. Drug-Drug Interactions with Direct Oral Anticoagulants: Practical Recommendations for Clinicians. Am. J. Med. 2021, 134, 939–942. [Google Scholar] [CrossRef]
  137. Mar, P.L.; Gopinathannair, R.; Gengler, B.E.; Chung, M.K.; Perez, A.; Dukes, J.; Ezekowitz, M.D.; Lakkireddy, D.; Lip, G.Y.H.; Miletello, M.; et al. Drug Interactions Affecting Oral Anticoagulant Use. Circ. Arrhythm. Electrophysiol. 2022, 15, e007956. [Google Scholar] [CrossRef]
  138. Mullangi, R.; Srinivas, N.R. Clopidogrel: Review of bioanalytical methods, pharmacokinetics/pharmacodynamics, and update on recent trends in drug-drug interaction studies. Biomed. Chromatogr. 2009, 23, 26–41. [Google Scholar] [CrossRef] [PubMed]
  139. Rehmel, J.L.; Eckstein, J.A.; Farid, N.A.; Heim, J.B.; Kasper, S.C.; Kurihara, A.; Wrighton, S.A.; Ring, B.J. Interactions of two major metabolites of prasugrel, a thienopyridine antiplatelet agent, with the cytochromes P450. Drug Metab. Dispos. 2006, 34, 600–607. [Google Scholar] [CrossRef]
  140. Farid, N.A.; Payne, C.D.; Small, D.S.; Winters, K.J.; Ernest, C.S., II; Brandt, J.T.; Darstein, C.; Jakubowski, J.A.; Salazar, D.E. Cytochrome P450 3A inhibition by ketoconazole affects prasugrel and clopidogrel pharmacokinetics and pharmacodynamics differently. Clin. Pharmacol. Ther. 2007, 81, 735–741. [Google Scholar] [CrossRef]
  141. Teng, R.; Butler, K. Effect of the CYP3A inhibitors, diltiazem and ketoconazole, on ticagrelor pharmacokinetics in healthy volunteers. J. Drug Assess. 2013, 2, 30–39. [Google Scholar] [CrossRef]
  142. Iisalo, E. Clinical pharmacokinetics of digoxin. Clin. Pharmacokinet. 1977, 2, 1–16. [Google Scholar] [CrossRef]
  143. Jalava, K.M.; Partanen, J.; Neuvonen, P.J. Itraconazole decreases renal clearance of digoxin. Ther. Drug Monit. 1997, 19, 609–613. [Google Scholar] [CrossRef]
  144. Mathis, A.S.; Friedman, G.S. Coadministration of digoxin with itraconazole in renal transplant recipients. Am. J. Kidney Dis. 2001, 37, E18. [Google Scholar] [CrossRef]
  145. Shumaker, A.C.; Bullard, H.M.; Churpek, J.; Knoebel, R.W. Posaconazole-digoxin drug-drug interaction mediated by inhibition of P-glycoprotein. J. Oncol. Pharm. Pract. 2019, 25, 1758–1761. [Google Scholar] [CrossRef]
  146. Hirai, T.; Kasai, H.; Shiga, T. Population pharmacokinetic analysis of the interaction of digoxin with N-desethylamiodarone in patients with atrial fibrillation and heart failure. Br. J. Clin. Pharmacol. 2025, 91, 2827–2835. [Google Scholar] [CrossRef] [PubMed]
  147. McGraw, J.; Cherney, M.; Bichler, K.; Gerhardt, A.; Nauman, M. The relative role of CYP3A4 and CYP3A5 in eplerenone metabolism. Toxicol. Lett. 2019, 315, 9–13. [Google Scholar] [CrossRef]
  148. Yamada, M.; Mendell, J.; Takakusa, H.; Shimizu, T.; Ando, O. Pharmacokinetics, Metabolism, and Excretion of [(14)C] Esaxerenone, a Novel Mineralocorticoid Receptor Blocker in Humans. Drug Metab. Dispos. 2019, 47, 340–349. [Google Scholar] [CrossRef] [PubMed]
  149. Gerisch, M.; Heinig, R.; Engelen, A.; Lang, D.; Kolkhof, P.; Radtke, M.; Platzek, J.; Lovis, K.; Rohde, G.; Schwarz, T. Biotransformation of Finerenone, a Novel Nonsteroidal Mineralocorticoid Receptor Antagonist, in Dogs, Rats, and Humans, In Vivo and In Vitro. Drug Metab. Dispos. 2018, 46, 1546–1555. [Google Scholar] [CrossRef]
  150. Hirai, T.; Ueda, S.; Ogura, T.; Katayama, K.; Dohi, K.; Hosohata, K.; Aoyama, T.; Matsumoto, Y.; Iwamoto, T. Hyperkalemia by eplerenone or esaxerenone in the presence or absence of clarithromycin in hypertensive patients: A retrospective observational cohort study. J. Hypertens. 2023, 41, 580–586. [Google Scholar] [CrossRef] [PubMed]
  151. Schachter, M. Chemical, pharmacokinetic and pharmacodynamic properties of statins: An update. Fundam. Clin. Pharmacol. 2005, 19, 117–125. [Google Scholar] [CrossRef]
  152. Zheng, E.; Madura, P.; Grandos, J.; Broncel, M.; Pawlos, A.; Woźniak, E.; Gorzelak-Pabiś, P. When the same treatment has different response: The role of pharmacogenomics in statin therapy. Biomed. Pharmacother. 2024, 170, 115966. [Google Scholar] [CrossRef] [PubMed]
  153. Neuvonen, P.J.; Niemi, M.; Backman, J.T. Drug interactions with lipid-lowering drugs: Mechanisms and clinical relevance. Clin. Pharmacol. Ther. 2006, 80, 565–581. [Google Scholar] [CrossRef] [PubMed]
  154. Neuvonen, P.J.; Kantola, T.; Kivistö, K.T. Simvastatin but not pravastatin is very susceptible to interaction with the CYP3A4 inhibitor itraconazole. Clin. Pharmacol. Ther. 1998, 63, 332–341. [Google Scholar] [CrossRef]
  155. Hesselink, D.A.; van Gelder, T.; van Schaik, R.H. The pharmacogenetics of calcineurin inhibitors: One step closer toward individualized immunosuppression? Pharmacogenomics 2005, 6, 323–337. [Google Scholar] [CrossRef]
  156. Knops, N.; Levtchenko, E.; van den Heuvel, B.; Kuypers, D. From gut to kidney: Transporting and metabolizing calcineurin-inhibitors in solid organ transplantation. Int. J. Pharm. 2013, 452, 14–35. [Google Scholar] [CrossRef]
  157. Udomkarnjananun, S.; Francke, M.I.; De Winter, B.C.M.; Mulder, M.B.; Baan, C.C.; Metselaar, H.J.; den Hoed, C.M.; Hesselink, D.A. Therapeutic drug monitoring of immunosuppressive drugs in hepatology and gastroenterology. Best Pract. Res. Clin. Gastroenterol. 2021, 54-55, 101756. [Google Scholar] [CrossRef]
  158. Canafax, D.M.; Graves, N.M.; Hilligoss, D.M.; Carleton, B.C.; Gardner, M.J.; Matas, A.J. Interaction between cyclosporine and fluconazole in renal allograft recipients. Transplantation 1991, 51, 1014–1018. [Google Scholar] [CrossRef]
  159. López-Gil, J.A. Fluconazole-cyclosporine interaction: A dose-dependent effect? Ann. Pharmacother. 1993, 27, 427–430. [Google Scholar] [CrossRef]
  160. Tian, Y.; Song, Y.; Qiao, Y.; Song, L.; Zhao, Q.; Yin, D.; Wang, S.; Hou, R. Effects of Triazole Antifungal Agents on the Plasma Concentration and Dosage of Cyclosporin in Patients with Aplastic Anaemia. J. Clin. Pharm. Ther. 2024, 2024, 6850289. [Google Scholar] [CrossRef]
  161. Kamdem, L.K.; Streit, F.; Zanger, U.M.; Brockmöller, J.; Oellerich, M.; Armstrong, V.W.; Wojnowski, L. Contribution of CYP3A5 to the in vitro hepatic clearance of tacrolimus. Clin. Chem. 2005, 51, 1374–1381. [Google Scholar] [CrossRef] [PubMed]
  162. Gu, T.M.; Lewis, J.S., II; Le, H.; Bubalo, J.S. Comparative effects of fluconazole, posaconazole, and isavuconazole upon tacrolimus and cyclosporine serum concentrations. J. Oncol. Pharm. Pract. 2022, 28, 1357–1362. [Google Scholar] [CrossRef]
  163. Vanhove, T.; Bouwsma, H.; Hilbrands, L.; Swen, J.J.; Spriet, I.; Annaert, P.; Vanaudenaerde, B.; Verleden, G.; Vos, R.; Kuypers, D.R.J. Determinants of the Magnitude of Interaction Between Tacrolimus and Voriconazole/Posaconazole in Solid Organ Recipients. Am. J. Transpl. 2017, 17, 2372–2380. [Google Scholar] [CrossRef]
  164. Suetsugu, K.; Ikesue, H.; Miyamoto, T.; Shiratsuchi, M.; Yamamoto-Taguchi, N.; Tsuchiya, Y.; Matsukawa, K.; Uchida, M.; Watanabe, H.; Akashi, K.; et al. Analysis of the variable factors influencing tacrolimus blood concentration during the switch from continuous intravenous infusion to oral administration after allogeneic hematopoietic stem cell transplantation. Int. J. Hematol. 2017, 105, 361–368. [Google Scholar] [CrossRef] [PubMed]
  165. Lebrun-Vignes, B.; Archer, V.C.; Diquet, B.; Levron, J.C.; Chosidow, O.; Puech, A.J.; Warot, D. Effect of itraconazole on the pharmacokinetics of prednisolone and methylprednisolone and cortisol secretion in healthy subjects. Br. J. Clin. Pharmacol. 2001, 51, 443–450. [Google Scholar] [CrossRef]
  166. Groll, A.H.; Desai, A.; Han, D.; Howieson, C.; Kato, K.; Akhtar, S.; Kowalski, D.; Lademacher, C.; Lewis, W.; Pearlman, H.; et al. Pharmacokinetic Assessment of Drug-Drug Interactions of Isavuconazole with the Immunosuppressants Cyclosporine, Mycophenolic Acid, Prednisolone, Sirolimus, and Tacrolimus in Healthy Adults. Clin. Pharmacol. Drug Dev. 2017, 6, 76–85. [Google Scholar] [CrossRef]
  167. Madsen, M.L.; Due, H.; Ejskjær, N.; Jensen, P.; Madsen, J.; Dybkær, K. Aspects of vincristine-induced neuropathy in hematologic malignancies: A systematic review. Cancer Chemother. Pharmacol. 2019, 84, 471–485. [Google Scholar] [CrossRef] [PubMed]
  168. Wang, E.J.; Lew, K.; Casciano, C.N.; Clement, R.P.; Johnson, W.W. Interaction of common azole antifungals with P glycoprotein. Antimicrob. Agents Chemother. 2002, 46, 160–165. [Google Scholar] [CrossRef]
  169. Venkatakrishnan, K.; von Moltke, L.L.; Greenblatt, D.J. Effects of the antifungal agents on oxidative drug metabolism: Clinical relevance. Clin. Pharmacokinet. 2000, 38, 111–180. [Google Scholar] [CrossRef]
  170. Scripture, C.D.; Figg, W.D. Drug interactions in cancer therapy. Nat. Rev. Cancer 2006, 6, 546–558. [Google Scholar] [CrossRef]
  171. van Leeuwen, R.W.; van Gelder, T.; Mathijssen, R.H.; Jansman, F.G. Drug-drug interactions with tyrosine-kinase inhibitors: A clinical perspective. Lancet Oncol. 2014, 15, e315-26. [Google Scholar] [CrossRef] [PubMed]
  172. Sharma, V.; Singh, T.G. Drug induced nephrotoxicity- A mechanistic approach. Mol. Biol. Rep. 2023, 50, 6975–6986. [Google Scholar] [CrossRef] [PubMed]
  173. Luber, A.D.; Maa, L.; Lam, M.; Guglielmo, B.J. Risk factors for amphotericin B-induced nephrotoxicity. J. Antimicrob. Chemother. 1999, 43, 267–271. [Google Scholar] [CrossRef]
  174. Takazono, T.; Tashiro, M.; Ota, Y.; Obata, Y.; Wakamura, T.; Miyazaki, T.; Nishino, T.; Izumikawa, K. Factor analysis of acute kidney injury in patients administered liposomal amphotericin B in a real-world clinical setting in Japan. Sci. Rep. 2020, 10, 15033. [Google Scholar] [CrossRef]
  175. Stanzani, M.; Vianelli, N.; Cavo, M.; Maritati, A.; Morotti, M.; Lewis, R.E. Retrospective Cohort Analysis of Liposomal Amphotericin B Nephrotoxicity in Patients with Hematological Malignancies. Antimicrob. Agents Chemother. 2017, 61, e02651-16. [Google Scholar] [CrossRef]
  176. Novak, J.E.; Ellison, D.H. Diuretics in States of Volume Overload: Core Curriculum 2022. Am. J. Kidney Dis. 2022, 80, 264–276. [Google Scholar] [CrossRef] [PubMed]
  177. Desai, N.B.; Hoenig, M.P. Potassium Homeostasis. Adv. Kidney Dis. Health 2024, 31, 504–513. [Google Scholar] [CrossRef]
  178. Hirai, T.; Yamaga, R.; Kei, M.; Hosohata, K.; Itoh, T. Geriatric Patients Are at a High Risk of Hypokalemia Associated with Yokukansan Preparation: A Retrospective Cohort Study. Biol. Pharm. Bull. 2020, 43, 1742–1748. [Google Scholar] [CrossRef]
  179. Veltri, K.T.; Mason, C. Medication-induced hypokalemia. PT 2015, 40, 185–190. [Google Scholar]
  180. Orofiamma, L.A.; Vural, D.; Antonescu, C.N. Control of cell metabolism by the epidermal growth factor receptor. Biochim. Biophys. Acta Mol. Cell Res. 2022, 1869, 119359. [Google Scholar] [CrossRef]
  181. Hsieh, M.C.; Wu, C.F.; Chen, C.W.; Shi, C.S.; Huang, W.S.; Kuan, F.C. Hypomagnesemia and clinical benefits of anti-EGFR monoclonal antibodies in wild-type KRAS metastatic colorectal cancer: A systematic review and meta-analysis. Sci. Rep. 2018, 8, 2047. [Google Scholar] [CrossRef] [PubMed]
  182. Workeneh, B.T.; Uppal, N.N.; Jhaveri, K.D.; Rondon-Berrios, H. Hypomagnesemia in the Cancer Patient. Kidney360 2021, 2, 154–166. [Google Scholar] [CrossRef] [PubMed]
  183. Huang, C.L.; Kuo, E. Mechanism of hypokalemia in magnesium deficiency. J. Am. Soc. Nephrol. 2007, 18, 2649–2652. [Google Scholar] [CrossRef]
  184. Rosner, M.H.; Ha, N.; Palmer, B.F.; Perazella, M.A. Acquired Disorders of Hypomagnesemia. Mayo Clin. Proc. 2023, 98, 581–596. [Google Scholar] [CrossRef] [PubMed]
  185. Arendrup, M.C.; Patterson, T.F. Multidrug-Resistant Candida: Epidemiology, Molecular Mechanisms, and Treatment. J. Infect. Dis. 2017, 216, S445–S451. [Google Scholar] [CrossRef]
  186. Rautemaa, R.; Ramage, G. Oral candidosis—clinical challenges of a biofilm disease. Crit. Rev. Microbiol. 2011, 37, 328–336. [Google Scholar] [CrossRef]
  187. Akbar, Z.; Aamir, M.; Saleem, Z. Optimizing antifungal therapy: A systematic review of pharmacist interventions, stewardship approaches, and outcomes. Front. Med. 2024, 11, 1489109. [Google Scholar] [CrossRef]
Figure 1. Mechanism of action of individual antifungal agents. Prepared using previously published data [56,57,58,59,60,61].
Figure 1. Mechanism of action of individual antifungal agents. Prepared using previously published data [56,57,58,59,60,61].
Jcm 14 07537 g001
Figure 2. Summary of drug–drug interaction regarding CYP isoforms and P-gp. Abbreviations: CYP: cytochrome P450 and P-gp: P-glycoprotein. Each circle represents an abundance of CYP enzymes. Prepared using previously published data [74,101,102,103,104].
Figure 2. Summary of drug–drug interaction regarding CYP isoforms and P-gp. Abbreviations: CYP: cytochrome P450 and P-gp: P-glycoprotein. Each circle represents an abundance of CYP enzymes. Prepared using previously published data [74,101,102,103,104].
Jcm 14 07537 g002
Table 1. Therapeutic options for oral candidiasis.
Table 1. Therapeutic options for oral candidiasis.
DrugFormulationRouteDoseFrequency
First line (mild)
ClotrimazoleTrochesTopical (oral cavity)10 mg5 times/day
NystatinSuspension
Pastilles
Topical (oral cavity)400,000–600,000 U (suspension)
1 pastille (pastilles)
4 times/day
MiconazoleMucoadhesive buccal tabletTopical (buccal)50 mgOnce daily
First line (Moderate to Severe)
FluconazoleTablet/capsuleOral100–200 mg (Day 1: 200 mg)Once daily
Alternative option
ItraconazoleOral solutionOral200 mgOnce daily
PosaconazoleOral suspensionOral400 mgTwice daily for 3 days, then once daily
VoriconazoleTabletOral200 mgTwice daily
The maximum recommended daily dose of fluconazole is 400 mg.
Table 2. Therapeutic options for esophageal candidiasis.
Table 2. Therapeutic options for esophageal candidiasis.
DrugFormulationRouteDoseFrequency
First line
FluconazoleTablet/capsule, IV solutionOral/IV200–400 mg (Day 1: 400 mg)Once daily
Second line
ItraconazoleOral solutionOral200 mgOnce daily
PosaconazoleOral suspension
DR tablet
Oral400 mg (suspension)
300 mg (tablet)
Once or twice daily
VoriconazoleTablet, InjectionOral/IV200 mgTwice daily
MicafunginInjectionIV150 mgOnce daily
Amphotericin B deoxycholateInjectionIV3 mg/kgOnce daily
Liposomal amphotericin BInjectionIV3 mg/kgOnce daily
Abbreviations: IV: intravenous. The maximum recommended daily dose of fluconazole is 800 mg.
Table 3. Major adverse effects of systemic antifungal agents.
Table 3. Major adverse effects of systemic antifungal agents.
Drug ClassAdverse EventsComment
Polyene macrolides
(Amphotericin B)
Infusion reaction
Nephrotoxicity, hypokalemia, hypomagnesemia
Liposomal formulations reduce toxicity
AzolesHepatotoxicity
Gynecomastia, adrenal suppression
QT prolongation (except isavuconazole)
Voriconazole; CNS toxicity and visual disturbance
EchinocandinsInfusion reaction
Hepatotoxicity
Generally tolerated
Abbreviations: CNS: central nervous system.
Table 4. Inhibition of azole antifungals with CYP enzymes and drug transporters.
Table 4. Inhibition of azole antifungals with CYP enzymes and drug transporters.
FluconazoleMiconazoleItraconazoleVoriconazolePosaconazoleIsavuconazole
Substrate
 CYP2C9 Y
 CYP2C19 Y
 CYP3A4 YY Y
 P-gp Y
Inhibitor
 CYP2C9++++ +
 CYP2C19+++ ++
 CYP3A4++ +++++++++++
 P-gp+ + +
Abbreviations: CYP: cytochrome P450, P-gp: P-glycoprotein. +: Mild, ++: Moderate, +++: Severe, Y: yes.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hirai, T.; Nashi, M. Pharmacological Management of Oral and Esophageal Candidiasis: A Clinical Pharmacotherapy Perspective. J. Clin. Med. 2025, 14, 7537. https://doi.org/10.3390/jcm14217537

AMA Style

Hirai T, Nashi M. Pharmacological Management of Oral and Esophageal Candidiasis: A Clinical Pharmacotherapy Perspective. Journal of Clinical Medicine. 2025; 14(21):7537. https://doi.org/10.3390/jcm14217537

Chicago/Turabian Style

Hirai, Toshinori, and Masanori Nashi. 2025. "Pharmacological Management of Oral and Esophageal Candidiasis: A Clinical Pharmacotherapy Perspective" Journal of Clinical Medicine 14, no. 21: 7537. https://doi.org/10.3390/jcm14217537

APA Style

Hirai, T., & Nashi, M. (2025). Pharmacological Management of Oral and Esophageal Candidiasis: A Clinical Pharmacotherapy Perspective. Journal of Clinical Medicine, 14(21), 7537. https://doi.org/10.3390/jcm14217537

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