C58H73N7O17
166663-25-8
Bloodstream Infections (BSI), Candidemia, Candidiasis, Esophageal Candidiasis, Fungal peritonitis caused by Candida, Intraabdominal Abscess caused by Candida, Invasive Aspergillosis, Oropharyngeal Candidiasis
Anidulafungin is a second-generation, semi-synthetic echinocandin antifungal agent administered intravenously for the treatment of serious Candida infections. It is distinguished within its class by a unique, non-enzymatic degradation pathway, which confers a highly favorable drug interaction profile and obviates the need for dose adjustments in patients with hepatic or renal impairment.[1] This characteristic positions anidulafungin as a first-line therapeutic option, particularly in critically ill patients with complex comorbidities and polypharmacy.
Key attributes of anidulafungin include its potent and selective inhibition of fungal (1→3)-β-D-glucan synthase, an enzyme essential for fungal cell wall integrity but absent in mammals, which results in fungicidal activity against most Candida species.[1] Its pharmacokinetic profile is characterized by a long terminal half-life of 40–50 hours, low systemic clearance, and a unique metabolic profile dominated by slow chemical degradation at physiological temperature and pH, independent of hepatic cytochrome P450 (CYP450) and renal elimination pathways.[1]
Clinically, anidulafungin is indicated for the treatment of candidemia, intra-abdominal abscess, peritonitis, and esophageal candidiasis in adults.[6] Its approval extends to pediatric patients aged one month and older for invasive candidiasis.[6] The drug is generally well-tolerated, with the most frequently reported adverse effects being hypokalemia, gastrointestinal disturbances (nausea, diarrhea), and pyrexia.[7] While it carries important warnings regarding the potential for hepatotoxicity and infusion-related hypersensitivity reactions, it does not have a black box warning from the U.S. Food and Drug Administration (FDA), reflecting a manageable risk profile in clinical practice.[11] Its minimal potential for drug-drug interactions makes it a valuable agent in the management of invasive fungal infections in complex patient populations, solidifying its place as a first-line therapy for invasive candidiasis.[13]
Anidulafungin is the established generic name for this small molecule antifungal agent.[1] It is marketed globally under several brand names, most notably
Eraxis in the United States and Russia, and Ecalta in Europe.[17] During its development phase at Eli Lilly and Company and later Vicuron Pharmaceuticals, it was known by the developmental code LY303366.[1]
For precise identification in scientific and regulatory databases, anidulafungin is assigned several unique identifiers:
Anidulafungin is classified as an echinocandin antifungal, a structural subclass of lipopeptides.[2] Chemically, it is a semi-synthetic, cyclic hexapeptide distinguished by an N-linked acyl lipid side chain. The formal IUPAC name is N--11,20,21,25-tetrahydroxy-3,15-bis-26-methyl-2,5,8,14,17,23-hexaoxo-1,4,7,13,16,22-hexaazatricyclo[22.3.0.09,13]heptacosan-18-yl]- 4-{4-[4-(pentyloxy)phenyl]phenyl}benzamide.[1] A more common chemical name is 1--4-yl]carbonyl]-L-ornithine]echinocandin B.[2]
The drug is produced via a semi-synthetic process that begins with echinocandin B, a natural lipopeptide obtained from the fermentation of the fungus Aspergillus nidulans or the related species Aspergillus rugulosus.[1] This starting material undergoes enzymatic deacylation, where the native linoleoyl side chain is cleaved by a deacylase enzyme derived from the bacterium
Actinoplanes utahensis. The resulting peptide core is then subjected to a series of synthetic chemical steps, including a reacylation process, to attach the unique terphenyl side chain that defines the anidulafungin molecule.[1]
Anidulafungin is supplied commercially as a sterile, lyophilized (freeze-dried) powder for concentrate for solution for infusion.[2] It is available in single-use vials containing either 50 mg or 100 mg of the active drug.[7]
In addition to the active pharmaceutical ingredient, the formulation contains several inactive ingredients (excipients) that are critical for the drug's stability, solubility, and administration. These include fructose (50 mg in 50 mg vial, 119 mg in 100 mg vial), mannitol (250 mg), polysorbate 80 (125 mg), tartaric acid, and sodium hydroxide and/or hydrochloric acid for pH adjustment.[2]
The choice of these excipients introduces critical safety considerations that are entirely independent of anidulafungin's primary pharmacological activity. The presence of fructose in the formulation leads to a direct and absolute contraindication in patients with Hereditary Fructose Intolerance (HFI).[24] This rare genetic disorder prevents the proper metabolism of fructose, and intravenous administration to affected individuals can precipitate a life-threatening metabolic crisis, including severe hypoglycemia, hypophosphatemia, lactic acidosis, and acute hepatic failure.[7] This necessitates a specific and careful patient history regarding HFI symptoms (e.g., nausea, vomiting, abdominal pain after consuming sugar) before anidulafungin can be safely administered.[8] The risk is particularly acute in the pediatric population, especially in infants and young children under two years of age, who may have undiagnosed HFI, making this a crucial point of clinical vigilance.[8]
Furthermore, the inclusion of polysorbate 80 as a solubilizing agent raises concerns for use in neonates. While toxicity has not been directly reported with the anidulafungin formulation, high doses of polysorbate 80 administered parenterally to low-birth-weight infants have been associated with a severe toxicity syndrome characterized by thrombocytopenia, renal dysfunction, hepatomegaly, cholestasis, and metabolic acidosis.[25] This formulation component contributes to the cautious approach and lack of approval for anidulafungin use in neonates (<1 month of age).
Anidulafungin exerts its antifungal effect through the potent and selective inhibition of the enzyme (1→3)-β-D-glucan synthase.[1] This enzyme is essential for the synthesis of 1,3-β-D-glucan, a fundamental structural polysaccharide that constitutes 30% to 60% of the cell wall in many pathogenic fungi, including
Candida and Aspergillus species.[27] By non-competitively inhibiting this enzyme, anidulafungin disrupts the formation of this critical polymer, leading to a severely weakened and defective fungal cell wall.[4] The compromised cell wall is unable to withstand osmotic pressure, resulting in cellular lysis and, ultimately, fungal cell death.[4]
The clinical safety and high therapeutic index of anidulafungin are rooted in its selective toxicity. The target enzyme, (1→3)-β-D-glucan synthase, is a vital component of the fungal cell but is entirely absent in mammalian cells.[1] This specificity ensures that the drug's activity is directed exclusively against the fungal pathogen, minimizing off-target effects on the human host.
The pharmacodynamic profile of anidulafungin is characterized by concentration-dependent antifungal activity. It is generally fungicidal against most Candida species and fungistatic against Aspergillus species.[30]
A defining and highly unusual pharmacodynamic feature of anidulafungin is its primary mechanism of clearance. Rather than being metabolized by host enzymes, it undergoes slow, spontaneous chemical degradation at physiological body temperature (37∘C) and pH.[1] This non-enzymatic hydrolysis process opens the cyclic peptide ring structure, converting anidulafungin into a primary degradant that lacks any antifungal activity.[1] The
in vitro half-life of this degradation process under physiological conditions is approximately 24 hours.[2] This inherent chemical instability is a central element of its pharmacokinetic profile and is paradoxically responsible for its most significant clinical advantages.
The pharmacokinetic profile of anidulafungin is predictable, with low intersubject variability (<25% coefficient of variation) and dose-proportional systemic exposures.[2] Its most notable characteristic is its clearance pathway via chemical degradation, which makes its disposition independent of hepatic and renal function and results in a minimal potential for drug-drug interactions.
Anidulafungin is administered exclusively by intravenous infusion, which results in 100% bioavailability.[1] Following the administration of a loading dose (twice the maintenance dose), steady-state plasma concentrations are rapidly achieved on the first day of therapy, with a plasma accumulation factor of approximately 2.[2]
The distribution of anidulafungin is characterized by a short initial distribution half-life of 0.5 to 1 hour.[2] It has a volume of distribution (
Vd) of 30–50 L, which is similar in magnitude to the total body fluid volume, suggesting distribution into tissues.[2] Anidulafungin is extensively bound to human plasma proteins, with reported binding of 84% to over 99%.[1]
Preclinical studies in rats have demonstrated rapid and extensive distribution into tissues, particularly those organs most commonly affected by invasive mycoses. Peak drug concentrations were achieved within 30 minutes in most tissues, and the total drug exposure (AUC) in the liver, lung, kidney, and spleen was found to be 9- to 12-fold higher than the exposure in plasma.[32] While the drug appears to persist longer in these key tissues than in plasma, penetration into the central nervous system (CNS) is limited, with only low levels of radioactivity detected in the cerebrospinal fluid (CSF) and brain tissue.[32]
The metabolism of anidulafungin is unique among antifungals and is the cornerstone of its predictable clinical behavior. Hepatic metabolism has not been observed.[1] The drug is not a substrate, inducer, or inhibitor of any cytochrome P450 (CYP450) isoenzymes at clinically relevant concentrations.[2]
Instead of enzymatic biotransformation, its clearance is driven by slow, non-enzymatic chemical degradation at physiological temperature and pH. This process of chemical hydrolysis opens the cyclic peptide structure to form a ring-opened peptide that is devoid of antifungal activity.[2] This primary degradant is then further broken down into smaller peptidic fragments and eliminated.[2] This clearance mechanism, being independent of the body's major drug-metabolizing systems, is the direct reason for anidulafungin's lack of clinically significant drug interactions and the absence of a need for dose adjustments in patients with organ dysfunction.
Elimination of anidulafungin and its degradants occurs primarily via the biliary route into the feces. Following administration of a single radiolabeled dose to healthy subjects, approximately 30% of the radioactive dose was recovered in the feces over a period of nine days. Intact, unchanged anidulafungin accounted for less than 10% of the material recovered in feces, indicating that the majority is eliminated as degradation products.[1]
Renal excretion is a negligible pathway for elimination. Less than 1% of the administered dose is excreted in the urine.[1] Plasma concentrations of the drug typically fall below the lower limit of quantitation by six days after a dose.[2]
Anidulafungin exhibits a low systemic clearance of approximately 1 L/h.[2] Its elimination profile is multiphasic, with a predominant elimination half-life of approximately 24–27 hours and a longer terminal elimination half-life of 40–50 hours.[1] This long half-life profile supports convenient once-daily dosing.
Table 1: Summary of Steady-State Pharmacokinetic Parameters of Anidulafungin in Adults
Dose Regimen (LD/MD, mg) | Cmax,ss (mg/L) | AUCss (mg·h/L) | Clearance (L/h) | Terminal Half-Life (t1/2, h) |
---|---|---|---|---|
100 / 50 | 4.2 | 55.2 | 0.84 | 43.2 |
200 / 100 | 7.2 - 8.6 | 110.3 - 111.8 | 0.94 | 26.5 - 52 |
Data compiled from healthy subjects and patients in clinical studies. LD = Loading Dose; MD = Maintenance Dose; Cmax,ss = Steady-state peak concentration; AUCss = Steady-state area under the curve; ss = steady state. Sources:.[2]
Anidulafungin is approved by regulatory agencies for the treatment of several serious fungal infections caused by Candida species. Its primary indications are:
Therapy may be initiated empirically before culture results are available, but treatment should be adjusted accordingly once the causative organism and its susceptibilities are identified.[6]
The product labeling for anidulafungin specifies several important limitations of use based on the populations and conditions studied in clinical trials:
Anidulafungin possesses a targeted spectrum of activity, demonstrating potent efficacy against specific fungal pathogens while lacking activity against others.
The echinocandin class as a whole, including anidulafungin, is intrinsically not active against several important fungal pathogens. These include:
This inherent resistance pattern is critical for guiding appropriate empiric therapy; anidulafungin should not be used if these organisms are suspected pathogens.
A notable phenomenon in laboratory testing is the "paradoxical effect" or "Eagle effect" observed when echinocandins are tested against Aspergillus species in vitro. This effect is characterized by an apparent increase in fungal metabolic activity or growth at drug concentrations well above the inhibitory level.[34] This artifact can make standard Minimum Inhibitory Concentration (MIC) testing, which measures growth inhibition, difficult to interpret and potentially misleading. Consequently, the clinically relevant susceptibility endpoint for echinocandins against molds is the Minimum Effective Concentration (MEC), a value determined by microscopic examination of hyphal morphology.[34] This distinction is vital for clinicians and microbiologists, as a high MIC value for anidulafungin against an
Aspergillus isolate may represent this in vitro paradox rather than true clinical resistance. Therefore, therapeutic decisions for mold infections should be guided by clinical trial data, practice guidelines, and MEC testing where available, rather than relying solely on standard MIC results.
The administration of anidulafungin requires strict adherence to approved dosing regimens, preparation procedures, and infusion rates to ensure both efficacy and patient safety.
Dosing is indication- and population-specific and involves an initial loading dose to rapidly achieve therapeutic concentrations, followed by a once-daily maintenance dose.
Treatment Duration: For invasive candidiasis, antifungal therapy should generally continue for at least 14 days after the last positive blood culture and the resolution of clinical signs and symptoms.[6] For esophageal candidiasis, patients should be treated for a minimum of 14 days and for at least 7 days following the resolution of symptoms.[17]
Anidulafungin must be prepared in a two-step aseptic process of reconstitution followed by dilution.
The maximum infusion rate is a critical safety parameter, not merely a guideline. This restriction is directly linked to the prevention of potentially severe, histamine-mediated infusion-related reactions.[7] Rapid administration of anidulafungin can trigger mast cell degranulation and the release of histamine, leading to symptoms such as rash, urticaria, flushing, pruritus, dyspnea, bronchospasm, and hypotension.[12] Adherence to the prescribed infusion duration is therefore a primary strategy for mitigating this risk. This necessitates careful programming of infusion pumps and clear communication between pharmacy and nursing staff to ensure the drug is never administered as a rapid IV push or bolus, which could provoke a serious adverse event.
Storage: The reconstituted solution (in the vial) is chemically and physically stable for up to 24 hours at room temperature (25°C or 77°F) prior to dilution. The final diluted infusion solution is stable for up to 48 hours at room temperature and must not be frozen.[12]
Table 2: Recommended Dosing and Administration Regimens for Anidulafungin
Indication | Patient Population | Loading Dose (Day 1) | Maintenance Dose (Daily) | Minimum Infusion Duration |
---|---|---|---|---|
Candidemia / Invasive Candidiasis | Adults | 200 mg | 100 mg | 90 minutes (100 mg dose) 180 minutes (200 mg dose) |
Candidemia / Invasive Candidiasis | Pediatrics (≥1 month) | 3.0 mg/kg (max 200 mg) | 1.5 mg/kg (max 100 mg) | Dose-dependent; rate must not exceed 1.1 mg/min |
Esophageal Candidiasis | Adults | 100 mg | 50 mg | 45 minutes (50 mg dose) 90 minutes (100 mg dose) |
Infusion durations are calculated based on the maximum infusion rate of 1.1 mg/min (equivalent to 1.4 mL/min) for the final 0.77 mg/mL solution. Sources:.[7]
Anidulafungin is generally well-tolerated, with a safety profile consistent with the echinocandin class. Adverse events are typically mild to moderate in severity.
The most frequently observed adverse reactions in clinical trials for invasive candidiasis vary slightly by patient population and indication.
While infrequent, several serious adverse reactions warrant clinical attention:
Anidulafungin does not carry a black box warning from the U.S. FDA.[5] The absence of this highest-level warning, despite the presence of significant warnings for hepatotoxicity and anaphylaxis in the product labeling, is a notable regulatory assessment of the drug's overall risk-benefit profile. Black box warnings are reserved for adverse reactions that are severe, potentially fatal, or require specific risk mitigation strategies beyond standard clinical practice. The FDA's decision suggests that while the risks of liver injury and anaphylaxis are real and require clinical vigilance, their incidence and manageability (e.g., monitoring liver function tests, discontinuing the drug if a reaction occurs) do not meet the threshold for the most stringent warning. This provides important context for clinicians, framing the safety profile as generally favorable but with specific, identifiable risks that must be monitored, rather than as a drug with an inherently high and unavoidable level of danger.
Anidulafungin is contraindicated in patients with:
Experience with anidulafungin overdose is limited. In one clinical trial, a single 400 mg dose was inadvertently administered as a loading dose without any clinical adverse events being reported.[5] Preclinical studies in rats found the maximum non-lethal dose to be 50 mg/kg, a dose equivalent to 5 to 10 times the recommended daily human dose based on relative body surface area.[5] In the event of an overdose, general supportive measures should be employed as necessary.[23]
Table 3: Incidence of Common Adverse Reactions (≥5%) in Adults with Candidemia
Adverse Reaction | Anidulafungin 100 mg (%) | Fluconazole (%) |
---|---|---|
Hypokalemia | 10 | 13 |
Diarrhea | 10 | 8 |
Nausea | 8 | 7 |
Vomiting | 6 | 6 |
Pyrexia | 6 | 5 |
Increased ALT | 5 | 3 |
Increased AST | 4 | 3 |
Data from the pivotal comparative trial in patients with candidemia/invasive candidiasis. Source:.[7]
A key clinical advantage of anidulafungin is its exceptionally low potential for clinically significant drug-drug interactions (DDIs). This favorable profile is a direct consequence of its unique clearance mechanism—slow, non-enzymatic chemical degradation—which completely bypasses the hepatic cytochrome P450 (CYP450) enzyme system and renal excretion pathways that are responsible for the metabolism and elimination of most drugs.[2] Anidulafungin is not a substrate, inducer, or inhibitor of CYP450 isoenzymes, making it a predictable agent in patients receiving multiple medications.[2]
Formal drug interaction studies have been conducted with several commonly co-administered medications, confirming this lack of interaction.
While pharmacokinetic interactions are minimal, a few pharmacodynamic or other interactions have been noted:
Table 4: Summary of Anidulafungin Drug Interaction Studies and Dosing Recommendations
Co-administered Drug | Known Mechanism/Substrate | Effect on Anidulafungin PK | Effect on Co-administered Drug PK | Clinical Recommendation |
---|---|---|---|---|
Cyclosporine | CYP3A4 Substrate | AUC increased by 22% | No significant effect | No dose adjustment required for either drug |
Tacrolimus | CYP3A4 Substrate | No significant effect | No significant effect | No dose adjustment required for either drug |
Voriconazole | CYP Inhibitor/Substrate | No significant effect | No significant effect | No dose adjustment required for either drug |
Rifampicin | Potent CYP450 Inducer | No significant effect | Not studied | No dose adjustment required for anidulafungin |
Liposomal Amphotericin B | Antifungal | No significant effect | Not studied | No dose adjustment required for anidulafungin |
Saccharomyces boulardii | Probiotic Yeast | N/A | Efficacy may be decreased | Avoid combination |
Voclosporin | Calcineurin Inhibitor | N/A | Potential for increased toxicity | Monitor closely; modify therapy if needed |
PK = Pharmacokinetics; AUC = Area under the curve; N/A = Not applicable. Sources:.[2]
The clinical use of anidulafungin is well-defined across various patient populations, with its unique pharmacokinetic profile simplifying its application in many complex scenarios.
Anidulafungin's clearance pathway via non-enzymatic chemical degradation renders it effectively independent of both renal and hepatic function, a profound clinical advantage that makes it an "organ-proof" antifungal. Numerous studies and regulatory documents confirm that no dosage adjustments are required for patients with any degree of renal insufficiency, including those with end-stage renal disease requiring hemodialysis.[13] The drug is not dialyzable, so it can be administered without regard to the timing of dialysis sessions.[2]
Similarly, no dosage adjustments are necessary for patients with any degree of hepatic impairment, including mild, moderate, or severe (Child-Pugh Class A, B, or C) insufficiency.[23] This is a direct result of its non-hepatic clearance.[2] This characteristic dramatically simplifies prescribing for critically ill patients, who often present with fluctuating or compromised organ function. It eliminates the need for dose calculations based on creatinine clearance or Child-Pugh scores, reducing the cognitive burden on clinicians and the potential for dosing errors. This provides a clear therapeutic advantage over other antifungals, such as caspofungin (which requires dose reduction in moderate hepatic impairment) and many azoles, positioning anidulafungin as a preferred echinocandin for patients with established or evolving liver disease.[13]
Clinical studies have shown no significant differences in the pharmacokinetics or safety profile of anidulafungin in geriatric patients (≥65 years) compared to younger adults. Although clearance was observed to be slightly lower in the elderly, the difference was not considered clinically relevant. Therefore, no dosage adjustment is required based on age.[2]
While the three clinically available echinocandins—anidulafungin, caspofungin, and micafungin—share a common mechanism of action and a similar spectrum of activity, they are not therapeutically interchangeable. Subtle but significant differences in their pharmacokinetics, metabolism, dosing requirements, drug interaction profiles, and approved indications create distinct clinical niches, allowing for tailored therapy based on specific patient characteristics.
All three agents are available only as intravenous formulations, are highly effective against most Candida species, and have an excellent overall safety profile.[28] However, a comparative analysis reveals key differentiators.
The choice among these agents is therefore a strategic clinical decision. For a critically ill adult with liver failure and polypharmacy, anidulafungin's predictable, organ-independent profile is highly advantageous. For a neonate with suspected candidemia or a patient requiring prophylaxis during a stem cell transplant, micafungin is the indicated choice. Caspofungin remains a valuable option, particularly in settings where its long-term efficacy and safety data are valued, provided its interaction and hepatic dosing considerations are managed.
Table 5: Comparative Profile of Anidulafungin, Caspofungin, and Micafungin
Feature | Anidulafungin | Caspofungin | Micafungin |
---|---|---|---|
Loading Dose Required | Yes (200 mg or 100 mg) | Yes (70 mg) | No |
Primary Metabolism | Chemical Degradation (Non-enzymatic) | Hepatic (Hydrolysis, N-acetylation) | Hepatic (Enzymatic, minor CYP3A4) |
Dose Adj. - Hepatic Impairment | No | Yes (Moderate Impairment) | No (but not studied in severe) |
Dose Adj. - Renal Impairment | No | No | No |
Key DDI (with CYP Inducers) | No interaction | Yes (requires dose increase) | No significant interaction |
Approved for Neonates (<1 mo) | No | No | Yes |
Prophylaxis Indication | No | No | Yes (HSCT patients) |
Adj. = Adjustment; DDI = Drug-Drug Interaction; HSCT = Hematopoietic Stem Cell Transplant. Sources:.[14]
Anidulafungin was originally discovered in the laboratories of Eli Lilly and Company and was subsequently developed by Vicuron Pharmaceuticals under the designation LY303366.[1] In 2005, Pfizer Inc. acquired Vicuron Pharmaceuticals and, with it, the rights to anidulafungin.[1] Pfizer brought the drug through the final stages of regulatory approval and continues to manufacture and market it globally.[16]
Anidulafungin has received marketing authorization from major regulatory agencies worldwide.
In recognition of its efficacy and safety profile for treating serious fungal infections, anidulafungin is included on the World Health Organization's (WHO) Model List of Essential Medicines. It is listed under the "Antifungal medicines" section for the indication of systemic or invasive candidiasis.[16]
Published at: August 3, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.