MedPath

Amphotericin B Advanced Drug Monograph

Published:Aug 2, 2025

Generic Name

Amphotericin B

Brand Names

Abelcet, Ambisome, Amphotec, Fungizone

Drug Type

Small Molecule

Chemical Formula

C47H73NO17

CAS Number

1397-89-3

Associated Conditions

Coccidioidomycosis, Fungal Infections, Histoplasmosis, Invasive Aspergillosis, Invasive Fungal Infections, Leishmaniasis, Meningitis, Cryptococcal, Meningitis, Fungal, Mucocutaneous Leishmaniasis, Mycotic endophthalmitis, Penicillium marneffei infection, Visceral Leishmaniasis, Candidal cystitis, Disseminated Cryptococcosis, Fungal osteoarticular infections, Ocular aspergillosis, Refractory aspergillosis, Severe Coccidioidomycosis, Severe Cryptococcosis, Severe Fungal infection caused by Basidiobolus spp., Severe Fungal infection caused by Conidiobolus spp., Severe Fungal infection caused by sporotrichosis spp., Severe Histoplasmosis, Severe Mucocutaneous leishmaniasis, Severe North American blastomycosis, Severe Systemic candidiasis

Amphotericin B: A Comprehensive Pharmacological and Clinical Review

Introduction and Historical Context

Discovery and Enduring Legacy

Amphotericin B (AmB) represents one of the most significant milestones in the history of antimicrobial chemotherapy and stands as a foundational agent in the treatment of severe fungal diseases.[1] Its discovery in the 1950s marked the dawn of effective systemic antifungal therapy. The journey began with a broad screening program of actinomycete cultures, leading to the isolation of a potent antifungal compound from a strain of

Streptomyces nodosus.[1] This particular microorganism, originally identified as M-4575, was cultured from a soil sample collected in the Orinoco River region of Venezuela, a testament to the power of natural product screening in drug discovery.[1]

As a therapeutic agent, AmB was licensed in 1959 based on the available clinical data and became commercially accessible in 1960 under the trade name Fungizone® (Bristol-Myers-Squibb).[1] This initial formulation was a colloidal suspension of AmB complexed with the bile salt sodium deoxycholate, a necessary innovation to overcome the drug's inherent insolubility in water.[2] For decades following its introduction, AmB was the only reliable treatment for life-threatening systemic mycoses and quickly established itself as the "gold standard" against which all subsequent antifungals would be measured.[2] Even after more than 60 years and the development of newer antifungal classes like the azoles and echinocandins, AmB remains a first-line or indispensable second-line therapy for many of the most aggressive fungal infections, including cryptococcal meningitis, invasive mucormycosis, and severe forms of aspergillosis and candidiasis.[2] Its enduring relevance is rooted in its exceptionally broad spectrum of activity and a remarkably low incidence of acquired microbial resistance, a feature that sets it apart from nearly all other antimicrobial agents.[1]

The Clinical Imperative for Innovation: The Rise of Lipid Formulations

The profound efficacy of Amphotericin B has always been shadowed by a significant and often dose-limiting toxicity profile.[1] From its earliest clinical use, the drug became notorious for causing severe adverse effects, earning it the memorable and dreaded nickname "amphoterrible".[5] The most prominent of these toxicities are acute, severe infusion-related reactions and chronic, cumulative nephrotoxicity.[2] These adverse events created a persistent clinical dilemma: the need to administer a life-saving medication that could simultaneously inflict substantial harm, particularly on the kidneys.[8]

This central paradox—a drug of immense power but poor tolerability—became the primary impetus for pharmaceutical innovation. The challenge was not to discover a new antifungal molecule but to re-engineer the delivery of the existing one to improve its therapeutic index. This led to a landmark advance in the late 1980s and 1990s: the development of lipid-based formulations of AmB.[1] These advanced drug delivery systems were designed to encapsulate or complex the AmB molecule within a lipid carrier, thereby altering its pharmacokinetic properties to minimize exposure of mammalian cells to the free drug while preserving its ability to reach and kill the target fungus. Three major lipid formulations were introduced: Amphotericin B Lipid Complex (ABLC; Abelcet®), Liposomal Amphotericin B (L-AmB; AmBisome®), and a third formulation, Amphotericin B Colloidal Dispersion (ABCD; Amphotec®), which was later discontinued due to a high rate of infusion-related events.[1] The history of Amphotericin B is therefore a quintessential case study in the evolution of pharmacotherapy. It demonstrates how a potent but challenging natural product can be systematically refined through the application of advanced pharmaceutical technology—in this case, lipid-based drug delivery—to overcome its inherent limitations and maintain its status as a vital clinical tool for over half a century.

Physicochemical Properties and Molecular Structure

Chemical Identification and Nomenclature

Amphotericin B is a well-characterized small molecule with a unique set of identifiers that precisely define its chemical nature. These identifiers are crucial for regulatory, research, and clinical purposes, ensuring accuracy and consistency in scientific communication.

  • DrugBank ID: DB00681 [5]
  • CAS Number: 1397-89-3 [11]
  • Molecular Formula: C47​H73​NO17​ [11]
  • Molecular Weight: Approximately 924.1 g/mol [10]
  • IUPAC Name: (1R,3S,5R,6R,9R,11R,15S,16R,17R,18S,19E,21E,23E,25E,27E,29E,31E,33R,35S,36R,37S)−33−oxy−1,3,5,6,9,11,17,37−octahydroxy−15,16,18−trimethyl−13−oxo−14,39−dioxabicyclo[33.3.1]nonatriaconta−19,21,23,25,27,29,31−heptaene−36−carboxylicacid [11]

The Amphipathic Architecture

Amphotericin B is classified as a polyene macrolide antibiotic, a structural definition that encapsulates its key features.[11] The molecule's architecture is dominated by a large, 38-membered macrocyclic lactone ring, which confers a distinct amphipathic character—possessing both hydrophilic (water-loving) and lipophilic (fat-loving) regions.[4] This structural duality is the master key to understanding its entire pharmacological profile, from its mechanism of action and toxicity to the pharmaceutical challenges of its formulation.

The macrocycle is functionally divided into two opposing faces:

  1. A rigid, lipophilic polyene region composed of seven conjugated double bonds in an all-trans configuration. This heptaene system is responsible for the molecule's chromophore properties and, more importantly, its affinity for sterols in lipid membranes.[4]
  2. A flexible, hydrophilic polyhydroxyl region containing a chain of seven or eight hydroxyl groups. This face of the molecule imparts water solubility and allows for interaction with the aqueous environment and the polar head groups of membrane phospholipids.[4]

Attached to this macrocyclic ring via a glycosidic bond is a critical sugar moiety, mycosamine (an aminohexose).[4] The presence of this sugar, particularly its amino group, has been shown to be indispensable for the high-affinity binding to its primary fungal target, ergosterol.[18] Furthermore, the molecule is amphoteric, containing both a primary amino group on the mycosamine sugar and a carboxylic acid group on the macrocycle, allowing it to form salts in either acidic or basic conditions.[17]

This unique amphipathic structure dictates a direct causal chain of pharmacological properties. The molecule's ability to interact with and insert into lipid bilayers is the foundation of its mechanism of action. Its stronger binding affinity for fungal ergosterol compared to mammalian cholesterol provides a degree of selectivity, which is the basis of its therapeutic effect.[19] However, its significant affinity for cholesterol is sufficient to cause damage to host cell membranes, particularly in the kidney, which explains its primary toxicity.[14] Finally, the molecule's large size and conflicting solubility preferences make it practically insoluble in water at physiological pH, a fundamental pharmaceutical challenge that necessitated the development of all its clinical formulations, from the original deoxycholate complex to the modern lipid carriers.[5]

Physical and Chemical Characteristics

In its pure form, Amphotericin B is an orange-yellow, odorless, crystalline solid or powder.[11] Its solubility is highly pH-dependent, a direct consequence of its amphoteric nature. It is practically insoluble in water at a neutral pH of 6 to 7 but becomes soluble in highly acidic (pH 2) or highly alkaline (pH 11) aqueous media.[17] This poor aqueous solubility at physiological pH is the central reason why it cannot be administered as a simple solution and requires complex formulations for intravenous use.[5] It displays good solubility in certain organic solvents, most notably dimethyl sulfoxide (DMSO) at 30-40 mg/mL and, to a lesser extent, dimethylformamide (DMF) at 2-4 mg/mL.[12] The molecule is also known to be sensitive to light and moisture and will degrade upon exposure; it also decomposes at temperatures exceeding 170°C.[15]

Mechanism of Action: From Dogma to a Dual-Action Model

The mechanism by which Amphotericin B exerts its potent fungicidal effect has been a subject of intense study for over 60 years. While the classical model of pore formation has long been accepted dogma, recent discoveries have refined this understanding, revealing a more complex, multi-faceted mode of action centered on a primary mechanism of sterol sequestration.[2]

The Classical Model: Ergosterol Binding and Pore Formation

The traditional and widely cited mechanism of action posits that AmB functions by disrupting the integrity of the fungal cell membrane.[2] The process begins with the high-affinity binding of the AmB molecule to ergosterol, the principal sterol component of fungal cell membranes.[6] This interaction is selective; while AmB can bind to cholesterol in mammalian membranes, its affinity for ergosterol is substantially higher, providing the basis for its antifungal activity.[19]

Following this initial binding, it is believed that multiple AmB molecules, along with ergosterol, self-assemble into aggregates that span the lipid bilayer, forming a stable transmembrane channel or pore.[2] The hydrophilic polyhydroxyl faces of the AmB molecules line the interior of this pore, creating an aqueous channel through the otherwise impermeable membrane. This structural disruption compromises the membrane's essential function as a selective barrier, leading to the rapid and uncontrolled leakage of vital intracellular components, particularly monovalent cations like potassium (

K+) and sodium (Na+), as well as protons (H+) and chloride ions (Cl−).[5] The resulting loss of the electrochemical gradient and intracellular contents leads to metabolic arrest and, ultimately, fungal cell death. This action can be either fungistatic (inhibiting growth) or fungicidal (killing the fungus), depending on the drug concentration achieved and the intrinsic susceptibility of the target organism.[10]

A New Paradigm: The "Sterol Sponge" Hypothesis

More recent and sophisticated biophysical studies have challenged the primacy of the pore-formation model, leading to a paradigm shift in understanding AmB's mechanism.[18] This new model proposes that the primary fungicidal action of AmB is not the formation of pores but rather the direct sequestration of ergosterol from the fungal membrane.

According to this "sterol sponge" hypothesis, AmB molecules first form large, extramembranous aggregates in the aqueous environment near the fungal cell surface.[24] These aggregates then act as powerful "sponges," actively extracting ergosterol molecules directly from the lipid bilayer.[19] Research using functional group-deficient derivatives of AmB has compellingly demonstrated that this process of ergosterol sequestration is, by itself, sufficient to kill yeast cells.[18] The formation of transmembrane channels is now viewed as a potential secondary or complementary effect that may contribute to the rate and potency of killing but is not an absolute requirement for the drug's lethal activity.[18]

This updated model provides a more robust explanation for AmB's enduring efficacy and, critically, its remarkably low rate of clinical resistance. The classical pore-formation model, involving a complex structural assembly, presents several theoretical avenues for a fungus to evolve resistance, such as by altering membrane fluidity or charge. In contrast, the sterol sponge model proposes a simpler and more direct assault: physically removing a lipid component that is absolutely essential for fungal membrane structure, fluidity, and the function of numerous membrane-bound proteins.[18] A fungus cannot easily survive without ergosterol or with a drastically altered version of it, meaning that the drug targets a fundamental vulnerability with very limited evolutionary escape routes. This inherent link between the drug's target and the pathogen's viability helps explain why, after more than six decades of clinical use, significant resistance to AmB remains a rare event.[5] This reframes AmB not just as a pore-former but as a potent "lipid sequestrant," a concept that holds valuable lessons for the design of future antimicrobial agents that may be inherently refractory to resistance.

Ancillary and Host-Interactive Mechanisms

Beyond its direct interaction with ergosterol, Amphotericin B's biological activity is augmented by at least two other mechanisms.

  • Oxidative Damage: There is evidence that AmB can undergo auto-oxidation, particularly its polyene chain, leading to the generation of reactive oxygen species (ROS).[2] These free radicals can induce oxidative damage to fungal membrane lipids and proteins, contributing to the overall cellular injury and death.
  • Immunomodulation: AmB is not a passive antimicrobial agent; it actively stimulates the host's innate immune system. It has been shown to be a potent activator of macrophages and monocytes, likely through interaction with Toll-like receptors, triggering the release of proinflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-1-beta (IL-1$\beta$).[2] This immunomodulatory effect is a double-edged sword. On one hand, it can enhance the host's ability to clear the fungal infection by stimulating phagocytic cells.[6] On the other hand, this same cytokine release is believed to be a primary driver of the acute, systemic infusion-related reactions—the "shake and bake" syndrome—that constitute one of the drug's most prominent toxicities.[5]

Pharmacokinetics and Pharmacodynamics (PK/PD)

The pharmacokinetic and pharmacodynamic profile of Amphotericin B is complex and, most notably, highly dependent on its formulation. Understanding these properties is essential for its safe and effective clinical use, as the different formulations are not therapeutically interchangeable.

Core Pharmacokinetic Profile (ADME)

The fundamental pharmacokinetic properties of the Amphotericin B molecule itself dictate its clinical behavior and the necessity for specialized formulations.

  • Absorption: AmB exhibits very poor absorption from the gastrointestinal tract, rendering oral administration ineffective for treating systemic infections.[6] Therefore, it must be administered parenterally, typically via intravenous infusion, through which its bioavailability is 100%.[10]
  • Distribution: Once in the bloodstream, AmB is highly bound (>90%) to plasma proteins, primarily lipoproteins.[6] It has a large volume of distribution, indicating extensive partitioning from the plasma into tissues. High concentrations are achieved in the organs of the reticuloendothelial system (RES), such as the liver, spleen, and bone marrow, as well as in the lungs and kidneys.[6] However, its penetration into the cerebrospinal fluid (CSF) is poor, with concentrations reaching only about 5% of those in the serum. For this reason, direct intrathecal or intraventricular administration is sometimes employed for fungal infections of the central nervous system, though this route is associated with a high risk of neurotoxicity.[6]
  • Metabolism: Current evidence suggests that Amphotericin B is not significantly metabolized by the liver or other tissues.[6]
  • Elimination: The elimination of AmB is exceedingly slow and follows a complex, multi-compartment kinetic model. After an initial plasma half-life of approximately 24 hours, the drug exhibits a very long terminal elimination half-life of around 15 days.[10] This prolonged terminal phase reflects the slow redistribution and release of the drug from deep tissue compartments where it is sequestered. The primary route of elimination is believed to be via the kidneys, but the process is inefficient; only a small fraction (2-5%) of the administered dose is excreted in the urine as active drug over a prolonged period.[6]

Formulation-Dependent Pharmacokinetics: A Tale of Two Deliveries

The development of lipid formulations dramatically altered the pharmacokinetic profile of AmB, creating distinct drug delivery platforms that behave very differently in the body. Unlike conventional AmB, the lipid formulations exhibit non-linear, dose-dependent pharmacokinetics.[6]

  • Conventional Amphotericin B Deoxycholate (C-AmB): This formulation consists of small micellar aggregates (less than 0.04 µm) of AmB and deoxycholate.[27] In the bloodstream, these micelles readily dissociate, releasing free AmB that can distribute widely and interact non-selectively with both fungal (ergosterol-containing) and mammalian (cholesterol-containing) cell membranes. This widespread, non-targeted distribution is responsible for its high toxicity.[27]
  • Liposomal Amphotericin B (L-AmB; AmBisome®): L-AmB is a true unilamellar liposome, a small (approximately 80 nm) and stable spherical vesicle into which the AmB molecule is intercalated.[2] These liposomes remain largely intact in the circulation for extended periods. This sequestration of AmB within the lipid carrier leads to a significantly smaller volume of distribution, a longer residence time in the plasma, and much higher peak plasma concentrations (Cmax) and area under the concentration-time curve (AUC) values compared to C-AmB at equivalent doses.[26] This altered biodistribution, which limits the interaction of free AmB with host tissues like the kidney, is the primary reason for its substantially improved safety profile.[23]
  • Amphotericin B Lipid Complex (ABLC; Abelcet®): ABLC is composed of large (1,600 to 11,000 nm), ribbon-like lipid complexes of AmB and two phospholipids.[2] Because of their large size, these particles are rapidly cleared from the circulation via uptake by phagocytic cells of the RES. This results in relatively low plasma concentrations but a very large volume of distribution, with rapid and extensive accumulation in RES-rich tissues, particularly the liver, spleen, and lungs.[3]

The distinct pharmacokinetic profiles of L-AmB and ABLC suggest they are not interchangeable and may be suited for different clinical scenarios. For a bloodstream infection like candidemia, the high and sustained plasma concentrations achieved with L-AmB may be advantageous. Conversely, for a deep-seated tissue infection like invasive pulmonary aspergillosis (IPA), the rapid and extensive lung tissue accumulation of ABLC could, in theory, lead to faster fungal clearance at the site of infection.[3] This highlights that the choice of a lipid formulation is a sophisticated clinical decision that goes beyond simply reducing toxicity and involves optimizing drug delivery based on the specific site of infection.

Pharmacodynamic Principles

The antifungal activity of Amphotericin B is governed by key pharmacodynamic principles that link drug exposure to its therapeutic effect.

  • AmB exhibits concentration-dependent fungicidal activity, meaning that higher drug concentrations lead to a more rapid and extensive killing of the target fungus.[6]
  • The primary PK/PD index that correlates with efficacy is the ratio of the peak plasma concentration to the minimum inhibitory concentration (Cmax/MIC).[28] Achieving a high Cmax relative to the pathogen's MIC is therefore a key therapeutic goal.
  • The drug also demonstrates a prolonged post-antifungal effect (PAFE), where it continues to suppress fungal growth for a period even after drug concentrations in the plasma have fallen below the MIC.[6] This effect, combined with its long half-life, provides a rationale for intermittent or less frequent dosing regimens in some situations.

Clinical Applications and Spectrum of Activity

Therapeutic Indications

Amphotericin B is a potent medication reserved for the treatment of progressive and potentially life-threatening fungal and certain protozoal infections.[5] Its use is explicitly contraindicated for the treatment of non-invasive fungal diseases, such as oral thrush (oropharyngeal candidiasis) or vaginal candidiasis, in patients with a normal immune system, due to its significant toxicity profile.[6] The specific FDA-approved indications can vary depending on the formulation.

Key indications include:

  • Empirical Therapy: For presumed fungal infections in high-risk patients, particularly those with febrile neutropenia who are unresponsive to broad-spectrum antibacterial therapy.[31]
  • Invasive Fungal Infections: As a first-line or alternative therapy for severe systemic infections caused by a wide range of pathogens, including:
  • Aspergillus species (Invasive Aspergillosis) [5]
  • Candida species (Candidemia, deep-seated Candidiasis) [5]
  • Cryptococcus neoformans (Cryptococcosis), especially for the induction phase of treatment for cryptococcal meningitis in HIV-infected patients.[5]
  • Endemic mycoses such as histoplasmosis, blastomycosis, and coccidioidomycosis.[5]
  • Mucormycosis (caused by fungi in the order Mucorales), for which AmB is considered a first-line therapy.[5]
  • Protozoal Infections: For the treatment of visceral leishmaniasis and the rare but devastating primary amoebic meningoencephalitis (PAM) caused by Naegleria fowleri.[5]
  • Salvage Therapy: For patients with infections refractory to, or in whom unacceptable toxicity precludes the use of, conventional amphotericin B deoxycholate.[31]

Research continues to explore new applications for AmB. For instance, a Phase 3 clinical trial is currently recruiting to evaluate the efficacy and safety of high-dose liposomal amphotericin B for the treatment of disseminated histoplasmosis in patients with AIDS.[35]

Spectrum of Activity and In Vitro Susceptibility

Amphotericin B possesses one of the broadest spectra of activity among all available antifungal agents, covering most medically important yeasts and molds.[2] It has no clinically relevant activity against bacteria, rickettsiae, or viruses.[10] The in vitro susceptibility of various fungi to AmB is a critical factor in guiding therapy, with some species being highly susceptible while others exhibit intrinsic resistance.

Table 1 provides a summary of the typical in vitro susceptibility for key fungal pathogens.

Table 1: In Vitro Susceptibility of Medically Important Fungi to Amphotericin B

Fungal SpeciesTypical MIC Breakpoint (mg/L)Source(s)
Aspergillus fumigatus15
Aspergillus terreusResistant5
Candida albicans15
Candida glabrata15
Candida krusei15
Candida lusitaniaeIntrinsically Resistant5
Cryptococcus neoformans25
Histoplasma capsulatum≤1.010
Blastomyces dermatitidis≤1.010
Coccidioides immitis≤1.010
Fusarium spp.Often Resistant10
Pseudallescheria boydiiOften Resistant10

Use in Special Populations

The use of Amphotericin B in specific patient populations requires careful consideration of the potential risks and benefits.

  • Pregnancy: While it appears to be relatively safe compared to other systemic antifungals, AmB should be used during pregnancy only if the potential benefit to the mother justifies the potential risk to the fetus. There are no adequate and well-controlled studies in pregnant women.[5]
  • Breastfeeding: It is not known whether AmB is excreted in human milk. Because of the potential for serious adverse reactions in nursing infants, a decision should be made whether to discontinue nursing or to discontinue the drug, taking into account the importance of the drug to the mother.[36]
  • Pediatrics: AmB is used in children and is not expected to cause different side effects or problems than it does in adults. However, some data suggest that children may clear the drug from plasma more rapidly than adults, which may have implications for dosing.[6]
  • Geriatrics: Although specific comparative studies are lacking, AmB is not expected to work differently or cause different side effects in older people compared to younger adults. However, elderly patients may have a higher prevalence of baseline renal impairment, which increases their risk of nephrotoxicity.[36]

Formulations: A Comparative Analysis

The clinical utility of Amphotericin B has been defined by the evolution of its formulations. The transition from the original conventional preparation to advanced lipid-based systems represents a major triumph of pharmaceutical science, aimed at mitigating the drug's formidable toxicity.

Conventional Amphotericin B Deoxycholate (C-AmB; Fungizone®)

This is the original formulation of Amphotericin B, introduced in 1960.[1] Due to AmB's poor water solubility, it is co-formulated with the bile salt detergent sodium deoxycholate. When reconstituted in a dextrose solution, this mixture forms a colloidal dispersion of micellar aggregates.[2] This formulation allows for intravenous administration but is associated with the highest rates of both acute infusion-related reactions and chronic nephrotoxicity, as the micelles readily release free AmB into the circulation to interact with host cells.[2]

Liposomal Amphotericin B (L-AmB; AmBisome®)

L-AmB is a true liposomal formulation, consisting of small, unilamellar (single-bilayer) vesicles with a diameter of approximately 60-80 nm.[2] The AmB molecule is stably intercalated within the lipid bilayer of the liposome, which is composed of hydrogenated soy phosphatidylcholine, cholesterol, and distearoyl phosphatidylglycerol.[14] This sophisticated structure effectively sequesters the AmB molecule, dramatically reducing its interaction with cholesterol-containing mammalian cell membranes while in circulation. This sequestration is the key to its significantly reduced toxicity, particularly nephrotoxicity, compared to both C-AmB and ABLC.[20] The intact liposomes are thought to preferentially accumulate at sites of infection and inflammation, where they can be taken up by phagocytic cells or interact directly with fungal cells to release their payload.

Amphotericin B Lipid Complex (ABLC; Abelcet®)

ABLC is structurally distinct from L-AmB. It is a complex of AmB with two phospholipids (dimyristoylphosphatidylcholine and dimyristoylphosphatidylglycerol) that form large, ribbon-like or disc-shaped particles with a size ranging from 1,600 to 11,000 nm.[2] Due to their large size, these lipid complexes are rapidly cleared from the bloodstream by phagocytic cells of the reticuloendothelial system (RES) in the liver, spleen, and lungs.[29] This results in a unique pharmacokinetic profile characterized by low plasma concentrations and rapid, high-level accumulation in these tissues.

Comparative Efficacy and Safety

The choice between AmB formulations is a complex clinical decision involving a trade-off between toxicity, efficacy, pharmacokinetics, and cost.

  • Toxicity: There is a clear and well-established hierarchy of safety. C-AmB is the most toxic formulation. All lipid formulations are significantly less nephrotoxic and cause fewer infusion-related reactions than C-AmB.[5] Among the lipid formulations, clinical data consistently demonstrate that L-AmB has a superior safety profile, with significantly lower rates of both nephrotoxicity and infusion-related reactions compared to ABLC.[38]
  • Efficacy: The comparative efficacy is less straightforward. While the lipid formulations offer a major advantage in tolerability, allowing for higher doses to be administered, they are not necessarily more efficacious than C-AmB on a milligram-per-milligram basis.[5] In head-to-head clinical trials comparing L-AmB and ABLC, overall therapeutic success rates have often been found to be similar, despite the clear differences in their safety profiles.[39] This suggests that factors other than just the formulation type, such as host immunity and the specific pathogen, play a crucial role in clinical outcomes.
  • Cost: A major real-world determinant of use is the substantial cost difference between the formulations. C-AmB is by far the least expensive option. The lipid formulations are significantly more costly, with L-AmB typically being the most expensive and ABLC often positioned as an intermediate-cost alternative.[38] This cost differential frequently influences institutional formularies and treatment guidelines, particularly in resource-limited settings.

The nuanced differences between the formulations underscore that the clinical choice is not simply about which drug is "best" but involves a complex risk-benefit calculation. L-AmB's superior safety profile is undisputed. However, ABLC's unique pharmacokinetic property of rapid and extensive uptake into tissues like the lung provides a theoretical advantage for certain deep-seated infections, though this has not been definitively proven in clinical outcomes. This makes the selection of a lipid formulation a multi-variable equation involving the patient's underlying condition (especially renal function), the site and severity of infection, and critical economic considerations.

Table 2 provides a comparative summary of the key characteristics of the three main AmB formulations to aid in clinical decision-making.

Table 2: Comparative Profile of Amphotericin B Formulations

ParameterC-AmB (Fungizone®)L-AmB (AmBisome®)ABLC (Abelcet®)
StructureMicellar dispersionSmall unilamellar liposomeLarge ribbon-like lipid complex
Particle Size (nm)< 40~801,600 - 11,000
Typical Dose (mg/kg/day)0.5 - 1.53 - 65
Peak Plasma Conc. (Cmax)LowHighLow
Volume of Distribution (Vd)HighLow (central compartment)Very High (tissue-sequestered)
Tissue DistributionBroad, non-specificPrimarily plasma, slow tissue distributionRapid RES uptake (liver, spleen, lung)
NephrotoxicityHighLowModerate (higher than L-AmB)
Infusion ReactionsHighLowModerate (higher than L-AmB)
Relative CostLowHighIntermediate

Adverse Effects and Toxicity Management

The clinical use of Amphotericin B is fundamentally limited by its propensity to cause a range of significant adverse effects. Proactive monitoring and management of these toxicities are paramount to ensuring patient safety and enabling the completion of a full therapeutic course. The adverse effects are not random but are direct, predictable consequences of AmB's core mechanism of action and its interactions with mammalian physiology.

Infusion-Related Reactions (IRRs)

Nearly all patients receiving intravenous AmB, particularly the conventional deoxycholate formulation, experience some form of acute infusion-related reaction.[42] These reactions are often severe and have led to the drug's "shake and bake" moniker.[5] The syndrome typically begins within 1 to 3 hours of starting the infusion and is characterized by a constellation of symptoms including high fever, violent shaking chills (rigors), hypotension or hypertension, headache, anorexia, nausea, and vomiting.[5] Dyspnea and tachypnea may also occur.[5]

The underlying mechanism of these reactions is believed to be an acute inflammatory response triggered by the AmB molecule itself. As a microbial product, AmB is recognized by the host's innate immune system, stimulating macrophages and monocytes to release a cascade of proinflammatory cytokines, including TNF-α and IL-1$\beta$, as well as prostaglandins.[5] The deoxycholate formulation may also directly stimulate the release of histamine from mast cells and basophils.[5]

Management of IRRs is primarily prophylactic. Premedication, administered 30 to 60 minutes prior to the AmB infusion, is standard practice. This typically includes an antipyretic (e.g., acetaminophen), an antihistamine (e.g., diphenhydramine), and sometimes a corticosteroid (e.g., hydrocortisone) to blunt the inflammatory response.[6] For severe rigors that occur despite premedication, a small dose of intravenous meperidine can be effective in reducing their duration and severity.[42] Additionally, slowing the rate of infusion can sometimes ameliorate the reaction's intensity.[44] The lipid formulations, especially L-AmB, are associated with a significantly lower incidence and severity of IRRs compared to C-AmB.[8]

Nephrotoxicity

Nephrotoxicity, or kidney damage, is the most significant and frequent dose-limiting chronic toxicity of Amphotericin B.[5] Some degree of renal impairment occurs in a majority of patients receiving C-AmB. The damage is a direct consequence of AmB's primary mechanism of action being applied to host cells. The mechanism is twofold:

  1. Renal Vasoconstriction: AmB induces constriction of the afferent arterioles in the glomerulus, which reduces renal blood flow (RBF) and the glomerular filtration rate (GFR).[8]
  2. Direct Tubular Toxicity: AmB binds to cholesterol present in the membranes of renal tubular epithelial cells, forming pores and causing direct cellular damage. This leads to impaired tubular function and the wasting of electrolytes.[5]

The clinical manifestations of AmB nephrotoxicity are predictable. Patients develop azotemia, with a progressive rise in blood urea nitrogen (BUN) and serum creatinine levels.[42] The tubular damage leads to Type 1 (distal) renal tubular acidosis, an inability to concentrate urine (nephrogenic diabetes insipidus), and profound electrolyte wasting, particularly of potassium and magnesium.[5] While often reversible upon discontinuation of the drug, some permanent renal impairment can occur, especially in patients who receive high cumulative doses (greater than 5 grams) of C-AmB.[42]

The primary strategy for mitigating nephrotoxicity is the use of lipid formulations, which are designed to limit the exposure of the kidneys to free AmB.[38] When C-AmB must be used, the most effective preventative measure is aggressive hydration with intravenous normal saline, often referred to as "sodium loading." Administering 500 mL to 1 L of 0.9% sodium chloride solution before the AmB infusion has been shown to significantly attenuate the decline in renal function.[6] Concomitant use of other nephrotoxic drugs should be avoided whenever possible, and renal function and electrolytes must be monitored meticulously throughout the course of therapy.

Electrolyte and Hematologic Disturbances

  • Hypokalemia and Hypomagnesemia: These are extremely common adverse effects, occurring as a direct result of renal tubular damage and wasting.[8] Low potassium (hypokalemia) and low magnesium (hypomagnesemia) can lead to severe muscle weakness, cramps, and potentially life-threatening cardiac arrhythmias.[8] Diligent monitoring of serum electrolyte levels and aggressive intravenous or oral supplementation are essential components of care for any patient receiving AmB.[43]
  • Anemia: A reversible, normochromic, normocytic anemia develops in up to 75% of patients undergoing prolonged therapy.[8] This is not due to hemolysis but is thought to result from a direct suppressive effect of AmB on erythropoietin production by the kidney and/or direct toxicity to bone marrow erythroid precursors.[8] The hematocrit typically stabilizes at 25-30% and recovers after therapy is completed.[8]

Other Systemic Toxicities

While less common than IRRs and nephrotoxicity, AmB can cause damage to other organ systems.

  • Hepatotoxicity: Elevations in liver function tests can occur, and in rare instances, acute hepatic failure has been reported.[8]
  • Cardiotoxicity: In addition to arrhythmias secondary to electrolyte disturbances, AmB has been associated with hypotension, shock, and rare cases of direct cardiotoxicity, including myocarditis and dilated cardiomyopathy.[5]
  • Neurotoxicity: Neurological side effects are less common but can be serious, including headache, peripheral neuropathy, dizziness, convulsions, and, rarely, encephalopathy.[8]

Drug and Disease Interactions

The interaction profile of Amphotericin B is a direct reflection of its own toxicity profile. It does not significantly alter the metabolism of other drugs via the cytochrome P450 system; instead, it creates a physiological state of renal impairment and electrolyte imbalance that renders the body more vulnerable to the effects of other medications.[26]

Drug-Drug Interactions

The most clinically significant interactions are pharmacodynamic in nature, involving additive toxicities.

  • Additive Nephrotoxicity: The risk of severe and potentially irreversible kidney damage is substantially increased when AmB is co-administered with other nephrotoxic agents. This includes aminoglycoside antibiotics (e.g., gentamicin, tobramycin), the antiviral foscarnet, the immunosuppressants cyclosporine and tacrolimus, and the chemotherapeutic agent cisplatin.[5] Intensive monitoring of renal function is mandatory if such combinations are unavoidable.
  • Potentiation of Hypokalemia: The renal potassium wasting induced by AmB is exacerbated by the concurrent use of other drugs that also lower potassium levels, such as loop and thiazide diuretics, and high-dose corticosteroids.[5] This can lead to profound hypokalemia.
  • Interactions Secondary to Hypokalemia: The hypokalemia caused by AmB can potentiate the toxicity of cardiac glycosides like digoxin, increasing the risk of arrhythmias. It can also enhance the effects of neuromuscular-blocking agents (paralytics), potentially leading to prolonged respiratory depression.[5] Close monitoring of serum potassium is crucial when these drugs are used together.
  • Interaction with Flucytosine: AmB and flucytosine exhibit synergistic antifungal activity. AmB is thought to increase the permeability of the fungal cell membrane, which facilitates the entry of flucytosine into the cell. However, this combination also has synergistic toxicity. AmB-induced nephrotoxicity can impair the renal excretion of flucytosine, leading to elevated levels of the drug and an increased risk of its primary side effects, myelosuppression and hepatotoxicity.[5]
  • Potential Antagonism with Azoles: There is a theoretical concern, supported by some in vitro data, that azole antifungals (e.g., fluconazole, itraconazole) could antagonize the activity of AmB. Azoles work by inhibiting the synthesis of ergosterol. By depleting the fungal membrane of ergosterol, they could reduce the number of available binding sites for AmB, thereby diminishing its effect. The clinical significance of this interaction remains uncertain, but caution is advised, especially in immunocompromised patients.[5]
  • Leukocyte Transfusions: Acute pulmonary toxicity, manifesting as respiratory distress, has been reported in patients receiving AmB infusions concurrently with leukocyte (white blood cell) transfusions. It is recommended to separate the administration times of these two therapies and to monitor pulmonary function closely.[5]

Disease State Interactions

The presence of certain pre-existing medical conditions significantly increases the risks associated with Amphotericin B therapy.

  • Renal Dysfunction: This is the most critical contraindication. Patients with pre-existing kidney disease are at an extremely high risk of developing severe, progressive, and potentially irreversible nephrotoxicity when treated with AmB, particularly the conventional formulation.[30] In such patients, a lipid formulation is strongly preferred.
  • Electrolyte Disturbances and Anemia: Patients with baseline hypokalemia, hypomagnesemia, or anemia are more susceptible to the worsening of these conditions during AmB therapy.[47] These conditions should be corrected as much as possible before initiating treatment.

Mechanisms of Fungal Resistance

One of the most remarkable features of Amphotericin B is the exceptionally low rate of clinically significant, acquired resistance, despite over 60 years of widespread use.[5] This phenomenon is not accidental but is rooted in the drug's unique mechanism of action and the profound biological cost that resistance imposes on the fungus.

Primary Mechanism: Alterations in Membrane Ergosterol

The principal and best-characterized mechanism of acquired resistance to AmB involves a reduction in the drug's primary target, ergosterol, within the fungal cell membrane.[48] This is typically achieved through mutations in the genes encoding enzymes of the ergosterol biosynthesis pathway (the

ERG genes).

In various fungal species, including Candida and Cryptococcus, mutations leading to the loss of function of enzymes such as C-5 sterol desaturase (Erg3), C-8 sterol isomerase (Erg2), C-24 sterol methyltransferase (Erg6), or lanosterol 14$\alpha$-demethylase (Erg11) have been identified in resistant isolates.[48] These genetic alterations disrupt the normal synthesis pathway, leading to a depletion of ergosterol in the cell membrane and its replacement by various precursor sterols (e.g., lanosterol, fecosterol). These precursor sterols have a much lower binding affinity for AmB, effectively removing the drug's target and rendering the fungus resistant.[48] In some cases, prior exposure to azole antifungals, which directly inhibit Erg11, can select for mutations that lead to ergosterol depletion, resulting in secondary cross-resistance to AmB.[49]

The "Fitness Cost" Trade-Off: The Key to Low Resistance

While the mutations described above can confer resistance to the drug, they come at a tremendous biological price for the pathogen. This concept of a "fitness cost" is the most critical insight into why AmB resistance remains rare in the clinical setting.[51] Ergosterol is not merely a passive target for a drug; it is a vital structural component of the fungal cell membrane, essential for maintaining proper fluidity, integrity, permeability, and the function of numerous membrane-bound enzymes.[18]

When a fungus develops resistance by altering its ergosterol content, it creates a "brittle" or compromised cell that is poorly adapted to survive within a mammalian host. Studies have shown that AmB-resistant mutants are often markedly less virulent than their drug-susceptible parent strains.[51] They exhibit a range of defects, including:

  • Hypersensitivity to environmental stresses, such as the febrile temperatures and oxidative stress encountered during infection.[49]
  • Increased susceptibility to killing by host immune cells, such as neutrophils.[51]
  • Defects in key virulence traits, such as the ability to form hyphae and invade host tissues.[51]

In essence, for the fungus to survive the drug, it must adopt a state in which it can no longer effectively survive the host. This creates a powerful evolutionary trade-off that selects against the emergence and propagation of resistant strains in a clinical environment. This makes Amphotericin B a powerful model for the development of "evolution-proof" antimicrobials, where the target is so fundamental to pathogen biology that resistance mutations inherently cripple the pathogen's ability to cause disease.

Other and Ancillary Resistance Mechanisms

While ergosterol alteration is the primary mechanism, other factors may contribute to reduced AmB susceptibility in some organisms.

  • Changes in Membrane Properties: Some resistant isolates have been shown to have increased membrane fluidity or decreased membrane permeability, which could theoretically hinder the drug's ability to bind, insert, and disrupt the membrane.[25]
  • Enhanced Oxidative Stress Response: In the parasite Leishmania, resistance has been linked to the upregulation of the thiol metabolic pathway (the trypanothione cascade). This enhanced antioxidant capacity allows the parasite to more effectively neutralize the reactive oxygen species generated by AmB, contributing to its survival.[25]
  • Drug Efflux: While a major mechanism of resistance for azole antifungals, the role of drug efflux pumps (such as those from the ATP-binding cassette transporter family) in AmB resistance appears to be less prominent, though it may play a contributing role in some cases.[25]

Conclusion and Future Directions

Summary of Clinical Standing

For more than six decades, Amphotericin B has held a unique and indispensable position in the antifungal armamentarium. It remains a cornerstone of therapy for severe, life-threatening mycoses, a status attributable to its potent, concentration-dependent fungicidal activity, its exceptionally broad spectrum of action, and a low rate of clinical resistance that is nearly unparalleled among antimicrobial agents. The clinical history of Amphotericin B is a narrative of balance—a continuous effort to harness its profound efficacy while mitigating its formidable toxicity. The development of sophisticated lipid formulations has been the key to tipping this balance in favor of the patient, transforming a drug once considered a "therapy of last resort" into a more manageable, albeit still complex, first-line option for many critical infections. The rational selection of a formulation, based on a nuanced understanding of its distinct pharmacokinetic profile, safety, and cost, is central to its modern clinical use.

Future Research and Development

Despite its long history, research into optimizing and expanding the utility of Amphotericin B continues, driven by the persistent threat of invasive fungal diseases and the emergence of multidrug-resistant pathogens.

  • Novel Formulations: The development of a safe, effective, and affordable oral formulation of Amphotericin B remains a "holy grail" of antifungal research. An oral agent would revolutionize the treatment of many endemic mycoses and could provide a pathway for long-term suppressive therapy or prophylaxis without the need for intravenous access.[8] Research into other novel drug delivery systems, such as nanodisks, cochleates, and microspheres, is also ongoing, with the goal of further improving the drug's therapeutic index.[8]
  • Optimizing Dosing Strategies: There is a need for further clinical research to refine dosing strategies for the existing lipid formulations. This includes exploring higher doses, intermittent or single-dose regimens (leveraging the drug's long half-life and post-antifungal effect), and tailoring doses based on the specific site of infection, pathogen susceptibility, and patient population, guided by robust pharmacokinetic and pharmacodynamic data.[26]
  • New Routes of Administration: The investigation of alternative delivery routes, such as inhaled Amphotericin B for the treatment or prevention of pulmonary fungal infections like allergic bronchopulmonary aspergillosis (ABPA), represents a promising area for reducing systemic toxicity while delivering high drug concentrations directly to the target organ.[52]
  • Confronting Emerging Threats: The rise of multidrug-resistant fungi, most notably Candida auris, underscores the continued importance of Amphotericin B. As resistance to azoles and echinocandins becomes more prevalent, AmB often remains one of the few viable therapeutic options. A deeper understanding of its activity and the subtle mechanisms of resistance in these emerging pathogens will be critical for preserving its efficacy in the years to come.[24]

Works cited

  1. Sixty years of Amphotericin B: An Overview of the Main Antifungal ..., accessed August 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7954977/
  2. It only takes one to do many jobs: Amphotericin B as antifungal and immunomodulatory drug, accessed August 2, 2025, https://www.frontiersin.org/journals/microbiology/articles/10.3389/fmicb.2012.00286/full
  3. Pharmacokinetics and Pharmacodynamics of Amphotericin B ..., accessed August 2, 2025, https://journals.asm.org/doi/10.1128/aac.01586-09
  4. (PDF) Sixty years of Amphotericin B: An Overview of the Main Antifungal Agent Used to Treat Invasive Fungal Infections - ResearchGate, accessed August 2, 2025, https://www.researchgate.net/publication/348942736_Sixty_years_of_Amphotericin_B_An_Overview_of_the_Main_Antifungal_Agent_Used_to_Treat_Invasive_Fungal_Infections
  5. Amphotericin B - Wikipedia, accessed August 2, 2025, https://en.wikipedia.org/wiki/Amphotericin_B
  6. Amphotericin B - StatPearls - NCBI Bookshelf, accessed August 2, 2025, https://www.ncbi.nlm.nih.gov/books/NBK482327/
  7. Managing side effects of amphotericin B infusions - Life Worldwide, accessed August 2, 2025, https://en.fungaleducation.org/amphotericin-b-infusions/
  8. Amphotericin B: side effects and toxicity | Revista Iberoamericana de ..., accessed August 2, 2025, https://www.elsevier.es/es-revista-revista-iberoamericana-micologia-290-articulo-amphotericin-b-side-effects-toxicity-S1130140609000291
  9. Comparative Efficacies of Four Amphotericin B Formulations ..., accessed August 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC353056/
  10. Amphotericin B: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed August 2, 2025, https://go.drugbank.com/drugs/DB00681
  11. Amphotericin B | C47H73NO17 | CID 5280965 - PubChem, accessed August 2, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Amphotericin-B
  12. Amphotericin B (AmB), Antimycotic polyene (CAS 1397-89-3) (ab141199) | Abcam, accessed August 2, 2025, https://www.abcam.com/en-us/products/biochemicals/amphotericin-b-amb-antimycotic-polyene-ab141199
  13. Amphotericin B (LNS-AmB, NSC 527017, CAS Number: 1397-89-3) | Cayman Chemical, accessed August 2, 2025, https://www.caymanchem.com/product/11636/amphotericin-b
  14. AmBisome® (amphotericin B) liposome for injection DESCRIPTION - Gilead Sciences, accessed August 2, 2025, https://www.gilead.com/-/media/files/pdfs/medicines/other/ambisome/ambisome_pi.pdf
  15. Amphotericin B deionized water 250ug/mL, 0.1 um filtered, BioReagent, cell culture mammalian 1397-89-3 - Sigma-Aldrich, accessed August 2, 2025, https://www.sigmaaldrich.com/US/en/product/sigma/a2942
  16. Amphotericin B | C47H73NO17 - ChemSpider, accessed August 2, 2025, https://www.chemspider.com/Chemical-Structure.10237579.html
  17. Amphotericin B | 1397-89-3 - ChemicalBook, accessed August 2, 2025, https://www.chemicalbook.com/ChemicalProductProperty_EN_CB3425912.htm
  18. Amphotericin primarily kills yeast by simply binding ergosterol - PNAS, accessed August 2, 2025, https://www.pnas.org/doi/10.1073/pnas.1117280109
  19. The Antifungal Mechanism of Amphotericin B Elucidated in Ergosterol and Cholesterol-Containing Membranes Using Neutron Reflectometry, accessed August 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7762259/
  20. Liposomal and Deoxycholate Amphotericin B Formulations: Effectiveness against Biofilm Infections of Candida spp. - MDPI, accessed August 2, 2025, https://www.mdpi.com/2076-0817/6/4/62
  21. Amphotericin B (E3789) - Datasheet - Sigma-Aldrich, accessed August 2, 2025, https://www.sigmaaldrich.com/deepweb/assets/sigmaaldrich/product/documents/290/384/e3789dat.pdf
  22. Amphotericin B - LKT Labs, accessed August 2, 2025, https://lktlabs.com/product/amphotericin-b/
  23. Liposomal amphotericin B—the past | Journal of Antimicrobial Chemotherapy, accessed August 2, 2025, https://academic.oup.com/jac/article/77/Supplement_2/ii3/6845632
  24. Antifungal Drug Resistance: Molecular Mechanisms in Candida albicans and Beyond - PMC, accessed August 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8519031/
  25. Mechanism of Amphotericin B Resistance in Clinical Isolates of Leishmania donovani - PMC, accessed August 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3264217/
  26. Clinical Pharmacokinetics, Pharmacodynamics, Safety and Efficacy of Liposomal Amphotericin B - Oxford Academic, accessed August 2, 2025, https://academic.oup.com/cid/article/68/Supplement_4/S260/5482449
  27. Liposomal Amphotericin B (AmBisome®): A review of the pharmacokinetics, pharmacodynamics, clinical experience and future directions - PMC - PubMed Central, accessed August 2, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4856207/
  28. Pharmacology of Liposomal Amphotericin B: An Introduction to Preclinical and Clinical Advances for Treatment of Life-threatening Invasive Fungal Infections - Oxford Academic, accessed August 2, 2025, https://academic.oup.com/cid/article/68/Supplement_4/S241/5482448
  29. Comparative Analysis of Amphotericin B Lipid Complex and Liposomal Amphotericin B Kinetics of Lung Accumulation and Fungal Clearance in a Murine Model of Acute Invasive Pulmonary Aspergillosis, accessed August 2, 2025, https://journals.asm.org/doi/10.1128/aac.01449-06
  30. Amphotericin B Injection: MedlinePlus Drug Information, accessed August 2, 2025, https://medlineplus.gov/druginfo/meds/a682643.html
  31. AmBisome® (amphotericin B) liposome for ... - accessdata.fda.gov, accessed August 2, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2008/050740s016lbl.pdf
  32. Fujisawa - accessdata.fda.gov, accessed August 2, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2000/50740S2LBL.pdf
  33. cancidas - This label may not be the latest approved by FDA. For current labeling information, please visit https://www.fda.gov/drugsatfda, accessed August 2, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/021227s028lbl.pdf
  34. medlineplus.gov, accessed August 2, 2025, https://medlineplus.gov/druginfo/meds/a616018.html#:~:text=Why%20is%20this%20medication%20prescribed%3F&text=Amphotericin%20B%20liposomal%20injection%20is,bone%20marrow)%20in%20certain%20people.%2520in%2520certain%2520people.&sa=D&source=editors&ust=1754123935877910&usg=AOvVaw3kIabCYZQ1scNoFp1FGHte)
  35. Fungal Infection Recruiting Phase 3 Trials for Amphotericin B (DB00681) | DrugBank Online, accessed August 2, 2025, https://go.drugbank.com/indications/DBCOND0031276/clinical_trials/DB00681?phase=3&status=recruiting
  36. Amphotericin b (intravenous route, injection route) - Side effects & dosage - Mayo Clinic, accessed August 2, 2025, https://www.mayoclinic.org/drugs-supplements/amphotericin-b-intravenous-route-injection-route/description/drg-20061771
  37. Amphotericin b liposome (intravenous route) - Side effects & dosage - Mayo Clinic, accessed August 2, 2025, https://www.mayoclinic.org/drugs-supplements/amphotericin-b-liposome-intravenous-route/description/drg-20061777
  38. Nephrotoxicity of three formulations of amphotericin B: trial sequential analysis, accessed August 2, 2025, https://www.archivesofmedicalscience.com/Nephrotoxicity-of-three-formulations-of-amphotericin-B-trial-sequential-analysis,102467,0,2.html
  39. Amphotericin B lipid complex versus liposomal amphotericin B monotherapy for invasive aspergillosis in patients with hematologic malignancy - PubMed, accessed August 2, 2025, https://pubmed.ncbi.nlm.nih.gov/18224662/
  40. A randomized, double-blind comparative trial evaluating the safety of liposomal amphotericin B versus amphotericin B lipid complex in the empirical treatment of febrile neutropenia. L Amph/ABLC Collaborative Study Group - PubMed, accessed August 2, 2025, https://pubmed.ncbi.nlm.nih.gov/11073745/
  41. COMPARATIVE EFFECTIVENESS AND SAFETY BETWEEN ..., accessed August 2, 2025, https://www.cambridge.org/core/journals/international-journal-of-technology-assessment-in-health-care/article/comparative-effectiveness-and-safety-between-amphotericin-b-lipidformulations-a-systematic-review/F35BA0380FA6F00682652676843D41D2
  42. Amphotericin B Side Effects: Common, Severe, Long Term - Drugs.com, accessed August 2, 2025, https://www.drugs.com/sfx/amphotericin-b-side-effects.html
  43. What are the side effects of Amphotericin B? - Patsnap Synapse, accessed August 2, 2025, https://synapse.patsnap.com/article/what-are-the-side-effects-of-amphotericin-b
  44. Mechanism of Action | AmBisome (amphotericin B) liposome for injection, accessed August 2, 2025, https://www.ambisome.com/ambisome-mechanism-of-action
  45. Systemic antifungal agents. Drug interactions of clinical significance - PubMed, accessed August 2, 2025, https://pubmed.ncbi.nlm.nih.gov/9512916/
  46. AmBisome (amphotericin B liposomal) dosing, indications ..., accessed August 2, 2025, https://reference.medscape.com/drug/ambisome-amphotericin-b-liposomal-999576
  47. Amphotericin b Interactions - Drugs.com, accessed August 2, 2025, https://www.drugs.com/drug-interactions/amphotericin-b.html
  48. Amphotericin B and Other Polyenes—Discovery, Clinical Use, Mode of Action and Drug Resistance - MDPI, accessed August 2, 2025, https://www.mdpi.com/2309-608X/6/4/321
  49. Insight into Virulence and Mechanisms of Amphotericin B Resistance in the Candida haemulonii Complex - MDPI, accessed August 2, 2025, https://www.mdpi.com/2309-608X/10/9/615
  50. Aspergillus terreus: Novel lessons learned on amphotericin B resistance - Oxford Academic, accessed August 2, 2025, https://academic.oup.com/mmy/article/56/suppl_1/S73/4925972
  51. Fitness Trade-offs Restrict the Evolution of Resistance to ..., accessed August 2, 2025, https://journals.plos.org/plosbiology/article?id=10.1371/journal.pbio.1001692
  52. Regulatory Perspective for Inhaled Antifungal Products - FDA, accessed August 2, 2025, https://www.fda.gov/media/142637/download

Published at: August 2, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.