Fact-checked by Grok 2 weeks ago

Amphotericin B

Amphotericin B is a polyene derived from the bacterium nodosus, widely recognized for its broad-spectrum activity against serious and life-threatening fungal and certain protozoal infections in humans. It functions by binding to in the fungal , forming pores that disrupt membrane integrity, leading to leakage of cellular contents, oxidative damage, and ultimately fungal . This mechanism provides fungicidal effects against a range of pathogens, including spp., spp., , and dimorphic fungi like . First isolated in 1955 and introduced clinically in the late 1950s, it remains a cornerstone therapy for invasive mycoses, particularly in immunocompromised patients, despite the development of alternative antifungals. Amphotericin B is available in several formulations to balance efficacy and tolerability, with the conventional deoxycholate form (Fungizone®) approved by the FDA in 1965, followed by lipid-based versions like liposomal amphotericin B (AmBisome®) in 1997 and amphotericin B lipid complex (Abelcet®) in 1995. These lipid formulations encapsulate the drug in liposomes or lipid complexes, reducing nephrotoxicity—the most significant adverse effect of the conventional form, which affects up to 80% of patients—while maintaining antifungal potency. Indications include empirical therapy for febrile neutropenia, treatment of disseminated candidiasis, cryptococcal meningitis, and aspergillosis, often as first-line or salvage therapy when other agents fail. Resistance remains rare, primarily due to alterations in ergosterol biosynthesis, but emerging concerns with species like Candida auris highlight the need for judicious use. Despite its efficacy, amphotericin B is reserved for severe infections due to potential toxicities, including acute infusion-related reactions (fever, chills, rigors) occurring in approximately 80% of patients with initial doses and electrolyte imbalances like . is essential, with lipid formulations preferred in patients at risk for kidney injury. Over six decades of use, amphotericin B has saved countless lives in critical care settings, and ongoing research explores less toxic oral or delivery systems to expand its .

Medical Uses

Fungal Infections

Amphotericin B is a first-line or salvage therapy for severe systemic fungal infections, particularly those caused by dimorphic fungi and molds in immunocompromised patients. It is indicated for when is not feasible, , , disseminated , severe , , , and severe . In invasive aspergillosis, lipid formulations of amphotericin B serve as an alternative initial for patients unable to tolerate , with evidence supporting its use in salvage settings for refractory cases. For invasive , including candidemia and deep-seated infections, amphotericin B deoxycholate or lipid formulations are recommended in cases of intolerance or resistance, particularly for krusei or glabrata. Cryptococcosis treatment, especially , involves liposomal amphotericin B at 3-4 mg/kg daily combined with for induction in high-income settings. For endemic mycoses, amphotericin B is preferred for severe or disseminated , with lipid formulations at 3-5 mg/kg daily for initial therapy in immunocompromised patients. In , it is used for life-threatening pulmonary or disseminated disease, followed by step-down to . For severe disseminated , lipid formulation amphotericin B is recommended initially; for , azoles are first-line, with amphotericin B (IV or intrathecal) as alternative for severe cases or azole failure. Mucormycosis guidelines strongly recommend high-dose liposomal amphotericin B (5-7.5 mg/kg daily) as first-line therapy due to its broad activity against Mucorales species. Severe , such as disseminated or osteoarticular forms, requires lipid formulations of amphotericin B at 3-5 mg/kg daily until clinical improvement, followed by step-down to . Standard dosing for systemic fungal infections typically involves amphotericin B deoxycholate at 0.5-1.5 mg/kg/day intravenously, while lipid formulations range from 3-5 mg/kg/day, adjusted higher (up to 10 mg/kg/day) for involvement or . Therapy duration varies by site and response, often 2-6 weeks initially before transitioning to oral azoles. Amphotericin B, particularly the liposomal formulation at 3 mg/kg/day, plays a key role in empirical antifungal therapy for high-risk patients with persistent fever despite broad-spectrum antibiotics, as endorsed by IDSA guidelines for presumed invasive .

Protozoal Infections

Amphotericin B is indicated as a first-line treatment for (also known as kala-azar) in regions with significant resistance to pentavalent antimonials, such as parts of the where treatment failure rates were historically up to 60% but are currently around 30-45% in areas like (as of 2024). For , it is reserved for select cases, including complicated, severe, or antimony-resistant infections, particularly those caused by species like Leishmania braziliensis or L. panamensis in the . The liposomal formulation of amphotericin B is preferred for due to its improved safety profile and efficacy, typically administered intravenously at 3 mg/kg/day for 5–7 days, achieving total doses of 15–21 mg/kg with cure rates exceeding 95% in endemic areas like . In , liposomal amphotericin B dosing ranges from 2–3 mg/kg/day for a total of 20–40 mg/kg over 10–20 doses, offering effective resolution in complex cases with minimal . While conventional deoxycholate amphotericin B remains an option at 0.75–1 mg/kg every other day for 15–20 doses, its use is limited by higher , making liposomal forms the standard in resource-limited settings. Combination therapies incorporating amphotericin B enhance outcomes for by improving efficacy and shortening treatment duration; for example, a single 5 mg/kg dose of liposomal amphotericin B combined with (100 mg/day for 7–8 days) or (15 mg/kg/day for 10–11 days) yields cure rates of 98–99% in antimony-resistant regions of . Such regimens reduce the risk of relapse and are particularly beneficial in high-burden areas. The endorses amphotericin B, especially the liposomal form, as a key therapy for antimony-unresponsive cases, recommending it in elimination programs for the and other endemic foci to address resistance and achieve high cure rates. Recent WHO guidelines (2022) for VL/HIV co-infection emphasize liposomal amphotericin B-based regimens. In , intensified elimination efforts have reduced VL incidence dramatically, with fewer than 1,000 cases reported annually as of 2023, though vigilance for resistance continues. These guidelines emphasize its role in combination strategies to optimize resource use and patient outcomes in global control efforts.

Spectrum of Activity

Amphotericin B demonstrates a broad spectrum of activity, primarily targeting pathogenic fungi by binding to in their cell membranes, which disrupts membrane integrity and leads to cell death. It is highly effective against a range of yeasts, including most species (such as C. albicans, C. glabrata, and C. parapsilosis) and , with minimum inhibitory concentrations (MICs) typically in the susceptible range. The drug also shows strong activity against molds, notably species (e.g., A. fumigatus and A. flavus) and members of the Mucorales order (e.g., and spp.), where it exhibits fungicidal effects at low concentrations. Additionally, amphotericin B is active against dimorphic fungi such as and , which can cause systemic infections in immunocompromised hosts. Beyond fungi, it possesses notable activity against the protozoan parasite species, making it a key therapeutic option for . Susceptibility testing for amphotericin B is guided by established breakpoints from organizations like the European Committee on Antimicrobial Susceptibility Testing (EUCAST). For most Candida species, isolates are considered susceptible at MICs ≤1 μg/mL and resistant at >1 μg/mL; similar breakpoints apply to Aspergillus species (susceptible ≤1 μg/mL, resistant >1 μg/mL). The Clinical and Laboratory Standards Institute (CLSI) does not provide formal clinical breakpoints but uses epidemiological cutoff values (ECVs) to distinguish wild-type from non-wild-type populations, typically around 1–2 μg/mL for Candida and 2–4 μg/mL for Aspergillus species. These thresholds help predict clinical efficacy, though in vivo outcomes can vary due to host factors and infection site. Despite its broad activity, amphotericin B has limitations against certain pathogens. It shows poor clinical utility and relatively weak in vitro activity against dermatophytes (e.g., ), with MICs comparable to but inferior to terbinafine. The drug lacks activity against bacteria such as species, which are treated with sulfonamides or other antibacterials rather than antifungals. Emerging resistance is a concern with , where many isolates exhibit elevated MICs (often >1 μg/mL) and reduced killing despite appearing susceptible by breakpoint criteria. Susceptibility to amphotericin B is primarily influenced by the content in the target organism's cell membranes, as the drug's binding affinity is higher for than for mammalian , conferring selectivity. Organisms with altered or reduced membrane levels may show decreased susceptibility. Typically, there is no cross-resistance with azoles, which inhibit synthesis upstream, allowing amphotericin B to remain effective against azole-resistant strains in most cases.

Formulations and Administration

Deoxycholate Amphotericin B

Deoxycholate amphotericin B, also known as conventional or Fungizone amphotericin B, is the original intravenous formulation of the antifungal agent, introduced in as a groundbreaking for systemic fungal infections. It consists of amphotericin B complexed with sodium deoxycholate to enhance solubility, as the parent compound is poorly soluble at physiological . Each 50 mg typically contains 50 mg of amphotericin B, 41 mg of sodium deoxycholate, and 20.2 mg of sodium phosphates (mono- and dibasic) as a , with the vial headspace filled with to prevent oxidation. For preparation, the lyophilized powder is reconstituted with 10 mL of sterile to yield a 5 mg/mL colloidal suspension, which is then further diluted in 5% dextrose injection (not saline, to maintain stability) to a final concentration of 0.1 mg/mL for administration; bacteriostatic agents or electrolytes must be avoided to prevent . Administration requires a slow intravenous over 2 to 6 hours to minimize infusion-related reactions, with an initial test dose of 1 mg infused in 20 mL of 5% dextrose over 20 to 30 minutes while monitoring . Dosing typically begins at 0.25 mg/kg/day and is gradually increased to 0.5 to 1 mg/kg/day based on tolerance, with a maximum of 1.5 mg/kg/day; for equivalence to lipid formulations, 1 mg/kg of deoxycholate amphotericin B is approximately comparable to 3 to 5 mg/kg of liposomal amphotericin B in terms of therapeutic effect, though direct substitution requires clinical judgment. Premedication with acetaminophen, diphenhydramine, or 30 to 60 minutes prior is often recommended to attenuate acute reactions. As the historical standard for over five decades, deoxycholate amphotericin B offered broad-spectrum efficacy against life-threatening mycoses at a low cost, making it accessible in resource-limited settings. However, its significant toxicity profile, including a high risk of affecting up to 80% of patients, has led to its gradual replacement by safer lipid-based alternatives in many developed healthcare systems, though it remains a viable option where cost is a barrier.

Liposomal Amphotericin B

Liposomal amphotericin B is a lipid-encapsulated formulation designed to improve the safety profile of amphotericin B by reducing its toxicity, particularly to the kidneys. The most widely used product, AmBisome, consists of amphotericin B intercalated into small unilamellar liposomes formed by a phospholipid bilayer comprising hydrogenated soy phosphatidylcholine, cholesterol, distearoylphosphatidylglycerol, and alpha-tocopherol. This structure limits the drug's distribution to renal tissues, thereby minimizing direct tubular exposure and nephrotoxic effects. Administration of liposomal amphotericin B is exclusively intravenous, with the lyophilized powder reconstituted in sterile water and diluted in 5% dextrose for over approximately 60 to 120 minutes using an in-line with a pore diameter of at least 1.0 micron. Unlike the conventional deoxycholate formulation, no or test dose is required prior to , facilitating easier clinical use. Dosing regimens vary by indication but generally range from 3 to 5 mg/kg body weight per day for empirical or systemic fungal infections such as invasive or . For , a total cumulative dose of up to 21 mg/kg is administered, typically as 3 mg/kg daily on days 1 through 5, 14, and 21 in immunocompetent patients, or higher and more frequent dosing in immunocompromised individuals. Adjustments may be made based on renal function, though the formulation's tolerability allows continuation in patients with mild impairment. The primary clinical advantage of liposomal amphotericin B is its substantially reduced , with studies reporting 50% to 60% lower rates of renal adverse events compared to amphotericin B deoxycholate, including fewer instances of doubling or discontinuation due to . This makes it the preferred option for high-risk patients, such as those with preexisting renal or requiring prolonged , where it demonstrates better overall tolerability and allows for higher cumulative doses without escalating toxicity. Infusion-related reactions, such as fever or chills, can occur but are generally milder than with other formulations.

Lipid Complex Amphotericin B

Lipid complex formulations of amphotericin B, such as amphotericin B lipid complex (ABLC) and amphotericin B colloidal dispersion (ABCD), consist of the antifungal agent complexed with lipids to form large, ribbon-like or disk-shaped structures that enhance tolerability while maintaining efficacy against systemic fungal infections. ABLC, marketed as Abelcet, incorporates amphotericin B in a 1:1 molar ratio with a phospholipid mixture of dimyristoyl phosphatidylcholine (DMPC) and dimyristoyl phosphatidylglycerol (DMPG) at a 7:3 ratio, resulting in multilamellar ribbon-like complexes approximately 1.6–11 μm in size. In contrast, ABCD, previously available as Amphotec, features amphotericin B complexed with cholesteryl sulfate in a 1:1 molar ratio, forming uniform disk-shaped particles around 0.12 μm in diameter upon reconstitution. These colloidal dispersions differ from liposomal formulations by their aggregate, non-vesicular architecture, which influences tissue distribution and reduces accumulation in renal tissues. Administration of lipid complex amphotericin B occurs via intravenous , typically over 1–2 hours to minimize infusion-related reactions, with precautions similar to those for the deoxycholate formulation, including with antipyretics or antihistamines and close monitoring for . For ABLC, the infusion rate is 2.5 mg/kg per hour, while is infused at 1 mg/kg per hour initially, potentially shortening to 2 hours for subsequent doses if tolerated. The standard dosing for systemic fungal infections is 5 mg/kg per day for ABLC and 3–4 mg/kg per day for in adults and children, allowing higher cumulative doses than conventional amphotericin B without proportional increases. These regimens are particularly suited for patients intolerant to or failing deoxycholate , with adjustments based on renal function and response. The primary advantages of lipid complex formulations include a reduced of compared to conventional amphotericin B deoxycholate, with incidence rates approximately 30–50% lower, enabling safer use in patients with renal impairment or those requiring prolonged therapy. This improvement stems from decreased renal uptake and , as the lipid complexes preferentially target the over the kidneys. Additionally, these formulations serve as a cost-effective to liposomal amphotericin B, offering similar against invasive mycoses at a lower while exhibiting an intermediate toxicity profile. Note that ABCD has been discontinued in many markets since 2011 due to higher rates of infusion-related adverse events, leaving ABLC as the predominant lipid complex option.

Oral Amphotericin B

Oral amphotericin B has historically been limited by its negligible systemic absorption following , with approaching zero, which confines its effects to the . This poor absorption has restricted its use primarily to topical applications or as part of selective digestive decontamination () regimens in critically ill patients, where it is administered orally in combination with other nonabsorbable antimicrobials like polymyxin E and tobramycin to prevent endogenous infections by targeting fungal overgrowth in the gut. In SDD protocols, oral amphotericin B is typically given as a multiple times daily, providing local coverage without significant systemic exposure. Recent advancements have focused on investigational oral formulations designed to enhance while minimizing toxicity. One prominent example is MAT2203, an oral lipid nanocrystal (LNC) formulation of amphotericin B developed by Matinas BioPharma, which encapsulates the drug in cochleate structures to improve gastrointestinal absorption and targeted delivery to fungal cells. This formulation is administered as oral capsules and has been evaluated in clinical trials for systemic fungal infections where is challenging. In a Phase 2 trial for cryptococcal (NCT04031833), MAT2203 combined with demonstrated noninferiority to standard intravenous amphotericin B plus in terms of early fungicidal activity, with similar survival rates and reduced , based on data from 2023 involving 141 HIV-positive patients. As of 2025, oral amphotericin B formulations like MAT2203 remain primarily investigational and are not approved by the FDA for systemic indications, though they hold promise for enabling outpatient therapy and improving access in resource-limited settings where intravenous administration is logistically difficult. The potential to shift treatment from hospital-based intravenous regimens to oral options could reduce healthcare burdens, particularly for conditions like in low-income regions, pending successful Phase 3 outcomes and regulatory approval.

Adverse Effects

Nephrotoxicity

Amphotericin B induces primarily through renal and direct damage to renal tubular cells, resulting from its binding to in mammalian cell membranes and subsequent disruption of cellular integrity. This leads to reduced renal blood flow and impaired tubular function, often manifesting as (AKI). The incidence of nephrotoxicity is notably high with conventional deoxycholate amphotericin B, affecting over 80% of patients, and is dose- and duration-dependent, with risks escalating at cumulative doses exceeding 1 mg/kg. In contrast, lipid-based formulations, such as liposomal amphotericin B, significantly lower this rate to around 15%, due to reduced exposure of renal tissues to the active . For example, in patients with hematologic malignancies, conventional deoxycholate was associated with AKI in approximately 36% of cases, often emerging within days of initiation. Key risk factors include pre-existing renal impairment, concurrent administration of other nephrotoxic agents such as cyclosporine or aminoglycosides, and , which can amplify vasoconstrictive effects and tubular toxicity. Concurrent nephrotoxic drugs like cyclosporine have been shown to synergistically increase the risk of severe AKI. Monitoring involves regular assessment of serum and electrolytes, including and magnesium, with daily checks initially and then weekly, to detect early elevations in creatinine (e.g., ≥0.3 mg/dL or 50% from baseline). Preventive measures include pre-infusion with 1 L of normal saline to mitigate , alongside potential dose adjustments if renal function declines. Infusion-related reactions (IRRs) to amphotericin B occur acutely during or shortly after intravenous administration and are primarily attributed to the release of pro-inflammatory cytokines triggered by the drug's interaction with host cells. These reactions are mediated by amphotericin B deoxycholate stimulating cytokine genes in vitro and in vivo, leading to symptoms such as fever, chills, rigors, nausea, vomiting, back pain, and arthralgias. Fever and chills are common on the first day of therapy with conventional amphotericin B deoxycholate, occurring in over 50% of patients. The incidence of IRRs is notably higher with amphotericin B deoxycholate, ranging from 30% to 72% across patient populations, compared to reduced rates with formulations. Premedication with agents such as acetaminophen, diphenhydramine, and administered 30 minutes prior to can significantly mitigate these reactions by suppressing cytokine-mediated . Additionally, infusing the drug over 2 to 6 hours, rather than more rapidly, helps minimize symptom severity by allowing gradual exposure. Tolerance to IRRs often develops with repeated administration, typically within the first few doses as the patient's adapts during therapy. Management protocols emphasize and controlled rates, with symptoms generally becoming less frequent after initial exposures. Among formulations, conventional amphotericin B deoxycholate exhibits the highest IRR rates, while liposomal amphotericin B shows the lowest, with significantly reduced incidence of fever and chills compared to conventional forms. Lipid complex formulations fall intermediately, offering a balance between efficacy and tolerability. Recent studies as of 2023 on single high-dose liposomal amphotericin B indicate further reductions in infusion-related adverse events.

Other Adverse Effects

Amphotericin B therapy is frequently associated with disturbances, particularly and hypomagnesemia. occurs in 30% to 50% of patients receiving liposomal amphotericin B, often necessitating supplementation and close monitoring to prevent complications such as cardiac arrhythmias. Hypomagnesemia, resulting from renal tubular loss, is also common and typically accompanies , with incidences reported up to 50% in treated patients; it can exacerbate and requires magnesium replacement. Hematologic adverse effects of amphotericin B include normocytic, normochromic anemia, which develops in up to 25% of patients during prolonged therapy due to suppression of erythropoietin production and is generally reversible upon discontinuation. Thrombocytopenia is a rarer manifestation, observed in fewer than 10% of cases and often linked to underlying hematologic conditions or concurrent myelotoxic therapies rather than the drug alone. Hepatic effects are usually mild and transient, with elevations in alanine aminotransferase (ALT) and aspartate aminotransferase (AST) occurring in up to 20% of patients, typically resolving after therapy cessation. Cholestasis is uncommon but has been reported in isolated cases, presenting as hyperbilirubinemia with minimal transaminase elevation, particularly in critically ill individuals. Neurological adverse effects are rare and generally limited to prolonged use, including isolated reports of seizures in patients with predisposing factors such as AIDS. Peripheral neuropathy has been documented infrequently (0.1% to 1% of cases), sometimes manifesting as demyelinating encephalopathy in vulnerable populations like bone marrow transplant recipients.

Drug Interactions

Nephrotoxic Drug Interactions

Amphotericin B exhibits significant interactions with other nephrotoxic agents, leading to enhanced renal impairment when used concurrently. These interactions primarily involve additive or synergistic effects on renal tubular cells and glomerular function, resulting in elevated serum creatinine levels and reduced glomerular filtration rate. Common culprits include aminoglycosides such as gentamicin, vancomycin, cyclosporine, and tacrolimus, which are frequently co-administered in patients with severe infections or undergoing immunosuppression. This risk is lower with lipid formulations of amphotericin B. The interaction with aminoglycosides, exemplified by gentamicin, arises from shared mechanisms of proximal tubular toxicity, where both agents disrupt lysosomal integrity and induce in renal epithelial cells, leading to cellular . Similarly, vancomycin potentiates amphotericin B's through additive damage to the renal tubules, with studies reporting an of 5.21 for when combined. Cyclosporine and , calcineurin inhibitors used in transplant settings, exacerbate amphotericin B-induced of renal arterioles and direct tubular injury, further impairing renal blood flow and solute reabsorption. Concurrent administration of these agents can lead to greater elevations in serum , reflecting accelerated tubular dysfunction and potential for acute renal failure. Such combinations significantly increase the incidence of , particularly when multiple nephrotoxins are involved, as evidenced in patients receiving empirical antifungal therapy alongside antibiotics. This heightened risk underscores the need for vigilant assessment in high-risk populations, such as those with hematologic malignancies or post-transplant. To mitigate these interactions, renal function—including serum , electrolytes, and estimated —should be monitored at least twice weekly during therapy, with more frequent checks if baseline exists. Dose reductions of amphotericin B (e.g., to 0.3–0.5 mg/kg/day) or the interacting agent may be necessary if creatinine rises by 0.5 mg/dL or more, alongside protocols to maintain output above 2 L/day. In severe cases, switching to lipid-formulated amphotericin B or alternative antifungals like is recommended to minimize cumulative renal insult.

Other Pharmacological Interactions

Amphotericin B exhibits additive or synergistic effects when combined with in the treatment of cryptococcal , leading to improved survival rates compared to amphotericin B monotherapy. This combination enhances fungal clearance and is recommended as first-line induction therapy for at least two weeks in severe cases. In contrast, concurrent use with azoles such as often results in antagonistic interactions against and species, as azoles reduce availability in fungal membranes, thereby diminishing amphotericin B's binding and fungicidal activity. Corticosteroids, while potentially exacerbating underlying fungal infections by suppressing immune responses, can mitigate amphotericin B-associated infusion-related reactions when used as , such as , to attenuate release. However, their concurrent administration requires careful monitoring due to the risk of compounded imbalances. Hypokalemia induced by amphotericin B can potentiate by increasing cardiac sensitivity to the , potentially leading to arrhythmias; supplementation and electrocardiographic monitoring are essential in patients receiving both agents. of amphotericin B plasma levels may be beneficial in select cases, such as pediatric or transplant patients, to optimize and minimize non-renal toxicity, though it is not routinely required. As amphotericin B is a substrate of (P-gp), co-administration with strong P-gp inducers (e.g., rifampin) or inhibitors (e.g., verapamil) should be avoided or closely monitored to prevent altered and .

Pharmacology

Mechanism of Action

Amphotericin B exerts its and effects primarily by interacting with components in the cell membranes of target organisms. The drug, a polyene , binds selectively to , the predominant in fungal and protozoal membranes, through hydrophobic and hydrogen-bonding interactions. This binding facilitates the aggregation of amphotericin B molecules into transmembrane channels or pores, typically composed of 7–10 drug molecules forming a barrel-like structure with embedded molecules. These pores span the , creating aqueous conduits that disrupt membrane integrity. The formation of these ion channels leads to uncontrolled leakage of essential ions, including (K⁺), sodium (Na⁺), and protons, from the cell interior. This ion efflux causes rapid membrane depolarization, osmotic imbalance, and subsequent cell lysis, rendering amphotericin B fungicidal and leishmanicidal at therapeutic concentrations. Additionally, the pores promote the generation of (ROS), which induce oxidative damage to cellular components, further contributing to pathogen death. The drug's activity is independent of the fungal , allowing it to target both proliferating and quiescent cells effectively. Amphotericin B's selectivity for fungal and protozoal cells over mammalian ones stems from its approximately 10- to 50-fold higher affinity for compared to , the primary in membranes. Ergosterol's rigid and higher membrane concentration in pathogens enhance pore stability and ion conductance, amplifying the drug's potency against these organisms. Although amphotericin B can bind cholesterol and form less stable pores in host cells, this differential affinity underlies its therapeutic window. to amphotericin B is rare and typically arises from mutations in ergosterol biosynthesis genes (e.g., ERG3 or ERG11), altering composition, rather than common mechanisms like overexpression seen with azoles.

Pharmacokinetics

Amphotericin B exhibits negligible oral , estimated at less than 5%, due to its poor aqueous and instability in gastrointestinal conditions, necessitating parenteral administration for . Intravenous of the conventional deoxycholate at typical doses of 0.5–1 mg/kg achieves peak plasma concentrations of approximately 1–2 μg/mL. Lipid-based formulations, such as liposomal amphotericin B, yield substantially higher peak levels, up to 58 μg/mL at a 5 mg/kg dose, owing to their larger particle size and prolonged circulation. The drug demonstrates a large , approximately 4 L/kg, reflecting extensive tissue penetration and binding, with over 90% accumulation in organs such as the liver, , and . exceeds 90%, further contributing to its wide distribution. Penetration into the is limited, reaching only 2–4% of simultaneous concentrations, which often requires for infections. Lipid formulations exhibit formulation-specific distribution patterns, with liposomal variants showing a smaller apparent (around 0.22–0.42 L/kg) due to uptake by mononuclear phagocytic cells in the liver, , and lungs, while achieving lower kidney concentrations (about 15% of those in hepatic tissues). Metabolism of amphotericin B is minimal and does not involve hepatic enzymes, with the parent drug remaining largely unchanged in circulation. Elimination occurs slowly, primarily through renal excretion of unchanged drug (2–5% over several days for conventional formulations), with minor biliary contributions; lipid formulations show even lower urinary output (<10% over 7 days) and favor fecal elimination. The initial is 24–48 hours for the deoxycholate form, extending to a terminal phase of about 15 days due to gradual release from tissue depots, leading to accumulation in patients with renal impairment. In contrast, lipid formulations prolong the terminal significantly, up to 7 days for liposomal amphotericin B, enabling less frequent dosing and reduced peak exposures.

Toxicity

Mechanism of Toxicity

Amphotericin B exerts its toxicity primarily through binding to in mammalian cell membranes, forming barrel-shaped pores that disrupt membrane integrity and lead to uncontrolled ion leakage, particularly and magnesium, causing cellular dysfunction and death. This interaction is especially pronounced in renal tubular cells, where the high content facilitates pore formation and subsequent ion imbalance, contributing to direct in the proximal tubules. Additionally, amphotericin B induces afferent vasoconstriction in the via stimulation of production, reducing renal blood flow and exacerbating ischemic damage to tubular epithelium. Systemically, amphotericin B triggers the release of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-1 (IL-1) from monocytes and macrophages, leading to infusion-related reactions including fever and chills. In erythrocytes, the drug promotes by generating , which damage lipid membranes and induce , resulting in . Lipid-based formulations of amphotericin B, such as liposomal amphotericin B, mitigate by encapsulating the drug in structures that limit the availability of amphotericin B to bind host , thereby reducing formation and associated cellular damage while preserving efficacy. This approach preferentially targets fungal membranes due to lower drug concentrations in .

Toxicity Management

Management of amphotericin B toxicity focuses on preventive measures to minimize and disturbances, alongside prompt when adverse effects occur. Aggressive hydration is a cornerstone of prevention, with administration of 1 liter of normal saline containing 20 mEq over 2 hours before each infusion recommended to reduce the risk of renal impairment. Lipid formulations, such as liposomal amphotericin B, are preferred over conventional deoxycholate due to their significantly lower nephrotoxic potential, allowing for safer use in patients at high risk for kidney injury. Alternate-day dosing may be considered after initial therapy to further mitigate cumulative toxicity once renal function stabilizes. For treatment of established toxicity, discontinuation is advised in cases of severe renal impairment, such as when serum rises to ≥2 times baseline and persists despite interventions, or in overdose scenarios where supportive care is provided but offers limited removal of the drug. Supportive measures include replacement, with intravenous (up to 40 mEq) and oral supplements for (serum K+ <3.3 mmol/L), alongside magnesium supplementation (e.g., 250 mg tablets twice daily) to address common deficiencies. If is severe, switching to antifungals like echinocandins is recommended to avoid further deterioration. Monitoring protocols are essential for early detection, involving baseline and 2–3 times weekly assessments of serum creatinine, , magnesium, and , particularly during the second week of therapy when risks peak. Daily of fluid intake, output, and weight is also advised, especially in hospitalized patients. In special populations, elderly patients require heightened vigilance due to age-related declines in renal function, potentially necessitating dose reductions or more frequent to prevent of . For those with hepatic disease, no routine dose adjustment is needed as amphotericin B elimination is primarily renal, but close observation for any emerging complications is warranted.

Biosynthesis

Microbial Production

Amphotericin B is primarily produced by the actinomycete bacterium nodosus, a soil-dwelling first identified as the key producer of this polyene . The biosynthesis occurs through a complex pathway involving modular (PKS) enzymes that assemble a 37-carbon polyene chain from , propionate, and butyrate-derived units, forming the macrolactone . Subsequent post-PKS modifications, including with the mycosamine at the C19 position, confer the molecule's amphoteric properties and enhance its solubility and activity. This pathway is encoded by a large spanning approximately 135 kb, which includes PKS modules and accessory genes for tailoring reactions such as hydroxylations and epoxidations. Industrial production of amphotericin B relies on submerged of S. nodosus in nutrient-rich media, typically composed of glucose as the primary carbon source and soybean flour or soy-based hydrolysates providing nitrogen and additional nutrients. The process involves seed culture preparation followed by inoculation into large-scale fermenters, where controlled and maintain dissolved oxygen levels above 20-30% to support polyketide chain elongation and oxidation steps. Yields have been optimized through , such as gene cluster overexpression and precursor supplementation, achieving titers up to 5 g/L in fed-batch cultures, a significant improvement over wild-type production of around 1-2 g/L. Biosynthesis is tightly regulated by environmental factors, including carbon catabolite repression mediated by glucose, which suppresses PKS when glucose levels are high, delaying production until later growth phases. Oxygen availability plays a critical role in formation, as it is required for P450-mediated hydroxylations and the incorporation of molecular oxygen into the polyene structure; suboptimal reduces precursor flux and overall yield. These regulatory mechanisms ensure efficient resource allocation, with industrial processes often employing fed-batch strategies to mitigate repression and maintain aerobic conditions for maximal output.

Chemical Synthesis

Amphotericin B presents formidable challenges in chemical synthesis owing to its intricate as a heptaene polyene , featuring 20 stereogenic centers, a 38-membered , and a sensitive conjugated polyene moiety prone to degradation by light and oxygen. These factors demand rigorous control over , efficient fragment assembly, and protection strategies to prevent or oxidation during multi-step processes. Semi-synthetic routes typically start from product isolated via microbial and involve targeted modifications to generate analogs with altered properties. A classic example is the preparation of N-iodoacetyl amphotericin B, formed by of the mycosamine amino group, which facilitated crystallographic analysis and served as a precursor for further derivatization. Contemporary semi-syntheses focus on toxicity mitigation; for instance, the analog AM-2-19 is produced through a two-step process from amphotericin B, entailing C2' epimerization to install a methyl configuration that disrupts binding, followed by C16 amidation with a serinol to enhance selectivity. This modification yields a compound with retained broad-spectrum activity but significantly reduced , as demonstrated and in murine models. Efforts toward have been pursued to enable structural diversification, with the landmark achievement by Nicolaou and coworkers in 1987 marking the first enantioselective route to amphotericin B. This multi-step endeavor, exceeding 70 linear steps, assembled key fragments including the polyol chain and aglycone via stereocontrolled aldol reactions and macrolactonization, culminating in glycosidation with a mycosamine derivative; notably, Suzuki-Miyaura cross-coupling was employed to construct the sensitive polyene segment of the aglycone. The overall yield remained below 1%, underscoring the inefficiency of such routes for large-scale production. These chemical and semi-chemical syntheses primarily support into less toxic amphotericin B derivatives, such as those targeting ergosterol-specific channels, but are not commercially viable compared to fermentation-based due to high costs, low yields, and handling complexities.

History

Discovery and Early Development

Amphotericin B was isolated in from a soil sample collected in the Orinoco River region of by researchers at the Squibb Institute for Medical Research. The compound was produced through fermentation of the actinomycete Streptomyces nodosus, which had been identified in the sample. It was named amphotericin B in due to its amphoteric chemical properties, capable of reacting as both an acid and a base. The initial isolation efforts were led by a team including R. Donovick, W. Gold, J.F. Pagano, and H.A. Stout, who reported its broad-spectrum antifungal activity against various pathogenic fungi . Early studies in the mid-1950s confirmed amphotericin B's potent effects, particularly against yeasts and dimorphic fungi, positioning it as a promising agent for systemic previously lacking effective treatments. The first reported human use occurred in 1957, when it was administered to a patient with disseminated , marking a critical step in its transition from laboratory to clinical application. Subsequent pre-clinical evaluations highlighted its in animal models of fungal , though challenges with and were immediately evident, requiring formulation with sodium deoxycholate for intravenous administration. The U.S. approved amphotericin B in 1965 under the trade name Fungizone, formulated as a deoxycholate complex, based on emerging clinical evidence of its life-saving potential. Pivotal early trials demonstrated its efficacy in treating severe cases of , with significant improvements in patient survival rates compared to prior therapies. These trials, conducted in the late , underscored the drug's role in managing disseminated fungal diseases, while also noting early concerns over renal toxicity and infusion-related reactions that would influence future management strategies.

Formulation Advancements

The development of lipid-based formulations of amphotericin B in the marked a significant advancement in mitigating the drug's while preserving its . These formulations, including amphotericin B (ABLC; Abelcet), approved by the FDA in November 1995 for treating invasive fungal infections in patients to or intolerant of conventional amphotericin B, encapsulated the drug in structures to reduce renal exposure. Similarly, amphotericin B cholesteryl sulfate (ABCD; Amphotec) received FDA approval in November 1996 for in patients intolerant to conventional therapy, and liposomal amphotericin B (L-AmB; AmBisome) was approved in August 1997 for empirical therapy in and cryptococcal in HIV-infected patients. These innovations shifted amphotericin B from a last-resort option to a more tolerable standard for severe systemic mycoses. The push for these lipid formulations stemmed from clinical trials in the demonstrating substantial reductions in , with early studies showing up to 50% lower incidence of renal impairment compared to the deoxycholate form, alongside cost-benefit analyses highlighting improved outcomes and reduced hospitalization costs despite higher upfront expenses. For instance, lipid complexes were found to allow higher cumulative doses with fewer discontinuations due to , balancing against the economic burden of renal support therapies. These advancements were driven by the need to address the conventional formulation's 50-80% rate in prolonged use. Recent investigational progress has focused on oral and less toxic analogs to further enhance and . The oral lipid nanocrystal formulation MAT2203 completed 2 trials in 2023, demonstrating noninferior early fungicidal activity to intravenous amphotericin B for cryptococcal , with significantly lower toxicity, including reduced and renal effects, in a of HIV-associated cases in . Preclinically, the amphotericin B analog AM-2-19, engineered in 2023 to selectively target fungal over human , has shown potent antifungal activity against and species in mouse models with markedly reduced kidney damage, positioning it as a promising candidate for clinical translation as of 2025. As a , amphotericin B, particularly its liposomal form, remains on the WHO Model List of Essential Medicines for treating and other severe infections; however, access remains limited in low-income countries. For example, as of , only 17.5% of clinics in reported availability of liposomal amphotericin B, exacerbated by high costs, with no-profit pricing increasing from $16.25 to $23 per vial in 2024, cold-chain requirements, and supply shortages for control in regions like and .

References

  1. [1]
    Amphotericin B - StatPearls - NCBI Bookshelf - NIH
    Feb 28, 2024 · Amphotericin B is primarily indicated for treating progressive and potentially life-threatening fungal infections. Amphotericin B should not be ...
  2. [2]
  3. [3]
    Amphotericin B Injection: MedlinePlus Drug Information
    ### Summary of Amphotericin B Injection
  4. [4]
    Aspergillosis - IDSA
    Jun 29, 2016 · Amphotericin B (AmB) deoxycholate and its lipid derivatives are appropriate options for initial and salvage therapy of Aspergillus infections ...Missing: cryptococcosis sporotrichosis
  5. [5]
    Clinical Practice Guideline for the Management of Candidiasis - IDSA
    Dec 16, 2015 · Lipid formulation amphotericin B (AmB) (3–5 mg/kg daily) is a reasonable alternative if there is intolerance, limited availability, or ...Antifungal Agents · Amphotericin B · Full Recommendations
  6. [6]
    ECMM/ISHAM/ASM Global Guideline for the Diagnosis and ... - IDSA
    Feb 14, 2024 · Liposomal amphotericin B 3–4 mg/kg daily and flucytosine 25 mg/kg four times a day is the most optimal induction therapy option for cryptococcal ...
  7. [7]
    Histoplasmosis - IDSA
    Mar 12, 2025 · These recommendations span treatment of asymptomatic Histoplasma pulmonary nodules (histoplasmomas) and mild and moderate acute pulmonary histoplasmosis.
  8. [8]
    Clinical Practice Guidelines for the Management of Blastomycosis
    Jun 1, 2008 · Evidence-based guidelines for the management of patients with blastomycosis were prepared by an Expert Panel of the Infectious Diseases Society of America.
  9. [9]
    IDSA 2016 Clinical Practice Guideline for the Treatment of ...
    Jul 27, 2016 · In this practice guideline, we have organized our recommendations to address actionable questions concerning the entire spectrum of clinical syndromes.
  10. [10]
    Global guideline for the diagnosis and management of mucormycosis
    First-line treatment with high-dose liposomal amphotericin B is strongly recommended, while intravenous isavuconazole and intravenous or delayed release tablet ...
  11. [11]
    Practice Guidelines for the Management of Patients with ...
    Amphotericin B is indicated for patients with life-threatening or extensive pulmonary sporotrichosis [21] (BIII). The most effective therapy appears to be a ...
  12. [12]
    Clinical Practice Guideline for the Use of Antimicrobial Agents in ...
    Nonetheless, the Panel recommends that every patient with fever and neutropenia receive empirical antibiotic therapy urgently (ie, within 2 h) after ...
  13. [13]
    [PDF] Control of the leishmaniases - The Global Health Network
    Control of the leishmaniasis: report of a meeting of the WHO Expert Committee on the Control of. Leishmaniases, Geneva, 22-26 March 2010. (WHO technical ...
  14. [14]
  15. [15]
    Clinical Care of Leishmaniasis - CDC
    Mar 13, 2024 · The FDA-approved regimen for immunosuppressed patients consists of 4 mg per kg daily on days 1–5, 10, 17, 24, 31, and 38 (total dose of 40 mg/kg) ...
  16. [16]
    Sixty years of Amphotericin B: An Overview of the Main Antifungal ...
    Amphotericin B is a life-saving drug in the treatment of serious systemic fungal infections and is still the most widely used antifungal in and intensive care, ...
  17. [17]
    Label: AMPHOTERICIN B injection, powder, lyophilized, for solution
    Prior to reconstitution Amphotericin B for Injection USP should be stored under refrigeration, 2˚ to 8˚C (36˚to 46˚F), protected against exposure to light.
  18. [18]
    Liposomal Amphotericin B for Empirical Therapy in Patients with ...
    Mar 11, 1999 · The mean daily doses throughout the study were 3.0±0.9 mg per kilogram for liposomal amphotericin B and 0.6±0.2 mg per kilogram for conventional ...
  19. [19]
    Amphotericin B: Time for a New “Gold Standard” - Oxford Academic
    When introduced in 1959, amphotericin B deoxycholate (AmBD) was clearly a life-saving drug. Randomized studies demonstrating its efficacy were not thought to be ...
  20. [20]
    Amphotericin B deoxycholate: time to retire our old standard?
    Jan 10, 2014 · It was the first orally available systemic antifungal agent. However, the oral formulation had problematic inter-patient differences in ...
  21. [21]
    [PDF] AmBisome® (amphotericin B) liposome for injection
    AmBisome consists of these unilamellar bilayer liposomes with amphotericin B intercalated within the membrane.
  22. [22]
    Liposomal Amphotericin B (AmBisome®): A review of the ...
    Amphotericin B is a polyene antifungal agent with a broad range of activity against yeasts and molds, as well as the protozoan parasite Leishmania spp. LAmB ...<|control11|><|separator|>
  23. [23]
    Optimizing efficacy of amphotericin B through nanomodification - PMC
    All the lipid formulations of AmB demonstrate improved efficacy, primarily because of the higher administered dose, and reduced kidney toxicity, compared to ...Role Of Lipid Formulations · Ambisome · Other Lipid Based...
  24. [24]
    [PDF] ABELCET® (Amphotericin B Lipid Complex Injection)
    Zidovudine: Increased myelotoxicity and nephrotoxicity were observed in dogs when either. ABELCET® (at doses 0.16 or 0.5 times the recommended human dose) or ...Missing: advantages | Show results with:advantages
  25. [25]
    [PDF] PRODUCT MONOGRAPH AMPHOTEC® (Amphotericin B ...
    Jan 12, 2006 · Despite generally less nephrotoxicity of AMPHOTEC® at the recommended doses compared with amphotericin B deoxycholate at the dose range of 0.8 ...
  26. [26]
    Amphotericin B formulations: a comparative review of efficacy and ...
    For the treatment of most invasive fungal infections, an amphotericin B lipid formulation provides a safer alternative than conventional amphotericin B, with at ...
  27. [27]
    Decontamination of the Digestive Tract and Oropharynx in ICU ...
    Jan 1, 2009 · SDD consisted of 4 days of intravenous cefotaxime and topical application of tobramycin, colistin, and amphotericin B in the oropharynx and ...
  28. [28]
    Selective decontamination of the digestive tract: an update of ... - NIH
    Selective decontamination of the digestive tract has been proven to prevent severe infections and to reduce mortality in critically ill patients.
  29. [29]
    MAT2203 :: Matinas BioPharma Holdings, Inc. (MTNB)
    An oral and non-toxic encochleated form of amphotericin B (CAmB). This antifungal drug is the primary broad-spectrum treatment for immunocompromised patients.Missing: prophylaxis | Show results with:prophylaxis
  30. [30]
    Oral Lipid Nanocrystal Amphotericin B for Cryptococcal Meningitis
    Aug 22, 2023 · We conducted a randomized clinical trial to evaluate oral LNC amphotericin (MAT2203; Matinas BioPharma, Bedminster, NJ) combined with ...
  31. [31]
    NCT03187691 | Safety and PK of Oral Encochleated Amphotericin B ...
    This is an open label phase II clinical study to evaluate the safety and pharmacokinetics of oral encochleated Amphotericin B (CAMB/MAT2203) for prevention of ...Missing: dosing | Show results with:dosing
  32. [32]
    Matinas BioPharma Provides Update to MAT2203 Regulatory and ...
    Dec 21, 2023 · Written Preliminary Comments on the Phase 3 Registration Trial for MAT2203 Move Company Closer to Full Alignment with FDA.
  33. [33]
    Matinas BioPharma Successfully Reaches Agreement with FDA for ...
    Feb 20, 2024 · Matinas BioPharma Successfully Reaches Agreement with FDA for a Single Phase 3 Registration Trial to Support an NDA for MAT2203 for the ...
  34. [34]
    Effects of tenofovir and amphotericin B deoxycholate co ... - NIH
    Amphotericin-related nephrotoxicity occurs from direct damage to renal tubules and constriction of the renal vasculature. Acute kidney injury has been estimated ...
  35. [35]
    Nephrotoxicity of Different Formulations of Amphotericin B
    The nephrotoxicity of amphotericin B (AmB) has long been known, but the risk of this side effect can be reduced if the drug is formulated as either an ...
  36. [36]
    Conventional Amphotericin B Associated Nephrotoxicity in Patients ...
    Jul 17, 2021 · AmB-d is associated with a significant risk of nephrotoxicity. In most hematological patients, antifungal treatment is initiated empirically, and patients ...
  37. [37]
    Risk factors for Amphotericin B-associated nephrotoxicity - PubMed
    Risk factors for amphotericin B-associated nephrotoxicity include higher average daily doses (approximately a doubling for each 0.10 mg/kg/day increment), ...Missing: cyclosporine | Show results with:cyclosporine
  38. [38]
    Influence of cyclosporine on the occurrence of nephrotoxicity after ...
    Some studies, which show an association between cyclosporine and nephrotoxicity, have found synergism with other nephrotoxic drugs such as amphotericin B and ...Missing: dehydration | Show results with:dehydration
  39. [39]
    The dose of the normal saline pre-infusion and other risk factors for ...
    Dec 28, 2023 · The dose of the normal saline pre-infusion and other risk factors for amphotericin B deoxycholate-associated acute kidney injury · Mathurot ...Missing: monitoring | Show results with:monitoring
  40. [40]
    Amphotericin-Associated Infusion-Related Reactions - PubMed
    Oct 23, 2021 · Findings: In vitro and in vivo studies have reported that amphotericin B deoxycholate stimulates pro-inflammatory cytokine genes causing IRAEs.
  41. [41]
    Amphotericin-Associated Infusion-Related Reactions: A Narrative ...
    Oct 22, 2021 · Amphotericin B has been reported to cause infusion-related adverse effects (IRAEs). To prevent IRAEs, pre-medications may be administered prior to the ...
  42. [42]
    Amphotericin Toxicity | PSNet - Patient Safety Network - AHRQ
    (1,2) Infusion-related adverse events, including fever, chills, and rigors, occur in 30%–72% of patients treated with amphotericin B deoxycholate (Fungizone).Missing: tolerance | Show results with:tolerance
  43. [43]
    Amphotericin B and its new formulations: pharmacologic ...
    Jan 5, 2002 · Tolerance to infusion-related reactions usually develops during therapy. Pretreatment with acetaminophen, diphenhydramine, chlorpheniramine ...Amphotericin B · Renal Toxicity · Amphotericin Lipid Complex<|control11|><|separator|>
  44. [44]
    Evaluation of hypokalemia and potassium supplementation during ...
    The incidence of hypokalemia in patients treated with L-AMB was reported as 36% by Ringden in 1994 (10) and as 51.3% by Sunakawa in 2012 (11). In the present ...
  45. [45]
    Hematological toxicities associated with amphotericin B formulations
    Anemia is a well-described adverse event related to the use of amphotericin B (AmB) [1]. Up to one-quarter of patients receiving this drug develop anemia, ...
  46. [46]
    Hematological toxicities associated with amphotericin B formulations
    In this study, significant hematological toxicity was associated with amphotericin B treatment, along with previous hematological disease and use of myelotoxic ...
  47. [47]
    Amphotericin B - LiverTox - NCBI Bookshelf - NIH
    Apr 8, 2016 · Amphotericin B is an antifungal agent with a broad spectrum of activity against many fungal species. Amphotericin B commonly causes mild to ...
  48. [48]
    Amphotericin B–Induced Seizures in a Patient with AIDS
    Amphotericin B seems to be the probable cause of the seizures. To date, only three cases of seizures associated with amphotericin B have been reported in the ...
  49. [49]
    Combination Antifungal Therapy for Cryptococcal Meningitis
    Apr 4, 2013 · Amphotericin B plus flucytosine, as compared with amphotericin B alone, is associated with improved survival among patients with cryptococcal meningitis.
  50. [50]
    Flucytosine | Johns Hopkins ABX Guide
    May 1, 2017 · Recommended in combination with amphotericin or liposomal amphotericin for treatment of cryptococcal meningitis for at least 2 weeks.
  51. [51]
    Dynamic Interaction between Fluconazole and Amphotericin B ...
    When these antifungals were tested in combination, fluconazole impaired the amphotericin B activity. The ergosterol content decreased with the increase of ...
  52. [52]
    Antagonistic interactions between azoles and amphotericin B with ...
    These four azoles antagonized the fungistatic activity of AmB at sub-MICs if both substances acted simultaneously.
  53. [53]
    Mechanism of Action | AmBisome (amphotericin B) liposome for ...
    However, the following drugs are known to interact with amphotericin B and may interact with AmBisome: antineoplastic agents, corticosteroids and ...
  54. [54]
    [PDF] Amphotericin B for Injection USP
    Amphotericin B for Injection should be administered by slow intravenous infusion. Intravenous infusion should be given over a period of approximately 2 to 6 ...Missing: Amphotec | Show results with:Amphotec
  55. [55]
    Therapeutic Drug Monitoring of Amphotericin-B in Plasma and ... - NIH
    May 11, 2022 · Our results showed that peritoneal amphotericin-B levels were significantly lower than plasma and that the correlation coefficient was 0.72 (p = ...
  56. [56]
    Sensitive LC-MS/MS Methods for Amphotericin B Analysis ... - Frontiers
    Nov 28, 2021 · Based on experimental studies in mice, AmB is probably a substrate of P-glycoprotein (P-gp) (Wu et al., 2015) and breast cancer resistance ...
  57. [57]
    Pharmacokinetics of antifungal drugs: practical implications for ...
    Jul 12, 2017 · In a preclinical study, enhanced cerebral amphotericin B uptake was achieved by exposure to P-glycoprotein (P-gp) inhibitors verapamil and ...
  58. [58]
    Antifungal Macrocycle Antibiotic Amphotericin B—Its Present and ...
    The mechanism of action of amphotericin B and its analogues is based on their interaction with cellular and lipid membranes, followed by formation of ion ...
  59. [59]
    Amphotericin B and Other Polyenes—Discovery, Clinical Use, Mode ...
    The active efflux of drugs, a common mechanism of resistance for azole drugs, does not appear to play a role for polyene resistance. Although one report ...
  60. [60]
    High-throughput and sensitive assay for amphotericin B interaction ...
    ... amphotericin B (AmB) ... AmB showed an approximately 18-fold higher affinity for ergosterol-containing membrane than for cholesterol-containing membrane.
  61. [61]
    Biodegradable Nanoparticles Improve Oral Bioavailability of ...
    Feb 13, 2009 · Amphotericin B (AMB), an effective antifungal and antileishmanial agent associated with low oral bioavailability (0.3%) and severe ...
  62. [62]
    Clinical Pharmacokinetics, Pharmacodynamics, Safety and Efficacy ...
    May 2, 2019 · In this article, we review the clinical pharmacokinetics, pharmacodynamics, safety, and efficacy of LAmB in a wide variety of patient populations and in ...Abstract · CLINICAL... · SAFETY AND TOLERABILITY · EFFICACY AGAINST...
  63. [63]
    Pharmacology of amphotericin B - UpToDate
    Aug 18, 2025 · INTRODUCTION. Amphotericin B is a polyene antifungal agent with activity in vitro against a wide variety of fungal pathogens [1].Missing: adjustment | Show results with:adjustment
  64. [64]
    Clinical Aspects of Amphotericin B Nephrotoxicity
    This article reviews the clinical aspects, histopathological changes, pathophysiology and prevention of AmB nephrotoxicity with emphasis on the renal tolerance.
  65. [65]
    The effect of thromboxane A2 receptor antagonism on amphotericin ...
    We investigated the role of thromboxane as a mediator for this nephrotoxicity. Vehicle or amphotericin (0.60 mg/kg) was infused into the left renal artery in ...
  66. [66]
    Amphotericin B Activation of Human Genes Encoding for Cytokines
    Amphotericin B results in a febrile response in humans, which peaks at ∼90 min after infusion and continues for up to 3 h. Chills and hypotension are also ...
  67. [67]
    Involvement of oxidative damage in erythrocyte lysis ... - PubMed - NIH
    Consistent with a role for oxidative damage in lysis, the mixture of erythrocytes and amphotericin B showed an increase in malonyldialdehyde, the product of ...Missing: anemia | Show results with:anemia
  68. [68]
    Liposomal and lipid-based formulations of amphotericin B - PubMed
    Encapsulating amphotericin B (AmB) into liposomes or binding of AmB to other lipid carriers results in a significant reduction of toxicity of AmB.
  69. [69]
    Amphotericin B lipid preparations: what are the differences?
    The markedly reduced nephrotoxicity of AmBi as compared to other AMB lipid formulations has led to its being tested at doses ≥10 mg/kg in several animal models.
  70. [70]
  71. [71]
    Amphotericin B deoxycholate versus liposomal amphotericin B
    Liposomal amphotericin B was found to be significantly safer than conventional amphotericin B in terms of serum creatinine increase.
  72. [72]
    RECOMMENDATIONS ON INDUCTION, CONSOLIDATION AND ...
    The 2018 WHO guidelines recommended seven days of amphotericin B deoxycholate and flucytosine-based therapy (followed by seven days of fluconazole 1200 mg) as ...
  73. [73]
    Amphotericin B biosynthesis in Streptomyces nodosus
    Jan 31, 2020 · The main AmB-producing microorganism is Streptomyces nodosus. Another fungus Penicillium nalgiovense Laxa was reported to synthesize AmB ...
  74. [74]
    Research Paper Amphotericin biosynthesis in Streptomyces nodosus
    The structure of amphotericin B [22] suggests that the polyketide precursor is synthesised from the following sequence of starter and extender units: AP2A8PA7, ...
  75. [75]
    Article Redesign of Polyene Macrolide Glycosylation: Engineered ...
    Feb 26, 2010 · Most polyene macrolide antibiotics are glycosylated with mycosamine (3,6-dideoxy-3-aminomannose). In the amphotericin B producer, ...
  76. [76]
    Enhancing the production of amphotericin B by Strepyomyces ...
    The fermentation media used for S. nodosus in 50-tons bioreactor consisted of 47 g/L soybean flour, 59 g/L glucose, 3.5 g/L cottonseed meal, 9 g ...
  77. [77]
    Enhanced AmB Production in Streptomyces nodosus by ... - Frontiers
    A novel combined feeding strategy was ultimately developed to improve the production of AmB by 85.9%, reaching 18.39 g/L that is the highest titer of AmB ever ...
  78. [78]
    Amphotericin B biosynthesis in Streptomyces nodosus
    Jan 31, 2020 · The AmB yield of modified strain S. nodosus VMR4A was 6.58 g/L, which was increased significantly in comparison with that of strain S. nodosus ...Missing: soy | Show results with:soy
  79. [79]
    Molecular regulation of antibiotic biosynthesis in streptomyces.
    Carbon Catabolite Repression of Antibiotic Production. It has long been known that extracellular glucose represses the production of many antibiotics, including ...<|control11|><|separator|>
  80. [80]
    Amphotericin B: 50 Years of Chemistry and Biochemistry
    The last century has seen the isolation and synthesis of a multitude of molecules with remarkable biological activity. ... Total Synthesis of Amphotericin ...
  81. [81]
    Total synthesis of amphotericin B - ACS Publications
    Solubility, aggregation and stability of Amphotericin B drug in pure organic solvents: Thermodynamic analysis and solid form characterization.
  82. [82]
    Polyene macrolide antibiotic amphotericin B. Crystal structure of the ...
    Spectroscopic evidence for self-organization of N-iodoacetylamphotericin B in crystalline and amorphous phases.
  83. [83]
    Tuning sterol extraction kinetics yields a renal-sparing polyene ...
    Nov 8, 2023 · We paired the C2′ epimerization with the most potency-promoting serinol side chain to form the hybrid derivative AM-2-19 (Fig. 3h). AM-2-19 ...Missing: analog | Show results with:analog
  84. [84]
    Synthesis and biological evaluation of amphotericin B derivatives
    Jun 16, 2010 · Amphotericin B ( AmB ) analogs prepared by semi-synthetic modifications as well as those produced via genetic modification of the AmB polyketide ...
  85. [85]
    Amphotericins A and B, antifungal antibiotics produced by ... - PubMed
    Amphotericins A and B, antifungal antibiotics ... R DONOVICK, W GOLD, J F PAGANO, H A STOUT. PMID: 13355330. No abstract available. MeSH terms. Amphotericin B* ...Missing: discovery | Show results with:discovery
  86. [86]
    Amphotericin B - American Chemical Society
    Aug 12, 2019 · Amphotericin B is an antifungal antibiotic produced by the bacterium Streptomyces nodosus. In 1955, it was isolated from soil collected near the Orinoco River ...
  87. [87]
    Systemic antifungal agents | Indian Journal of Pediatrics
    Since the discovery in 1955, amphotericin B has been the cornerstone of anti-fungal treatment. ... Gold W, Stout NA, Pagano JF, Donovick R. Amphotericin A and B, ...
  88. [88]
    Pharmacokinetics and Pharmacodynamics of Amphotericin B ...
    Amphotericin B (AmB) is a polyene derived from Streptomyces nodosus. This compound was discovered in the mid-1950s and remains a first-line agent for the ...
  89. [89]
    Coccidioidomycosis and its treatment with amphotericin B - PubMed
    Coccidioidomycosis and its treatment with amphotericin B. Am J Med. 1958 Apr;24(4):568-92. doi: 10.1016/0002-9343(58)90297-3. Authors. M L LITTMAN, P L ...Missing: pivotal trials
  90. [90]
    Amphotericin B - ScienceDirect
    It was originally isolated from a soil culture from the Orinoco River region, Venezuela. Used topically as a cream, or parenterally as a Na-desoxycholate ...