Fact-checked by Grok 2 weeks ago

Glycopeptide antibiotic

Glycopeptide antibiotics are a class of glycosylated cyclic or polycyclic nonribosomal peptides that inhibit bacterial cell wall synthesis in Gram-positive bacteria by binding to the D-alanyl-D-alanine (D-Ala-D-Ala) terminus of peptidoglycan precursors, such as lipid II, thereby preventing cross-linking and leading to cell death. These antibiotics, first discovered in the mid-20th century, have been essential in treating serious infections caused by multidrug-resistant pathogens, with vancomycin, approved in 1958, serving as the prototype and remaining on the World Health Organization's List of Essential Medicines for intravenous use against conditions like methicillin-resistant Staphylococcus aureus (MRSA) bacteremia and endocarditis. Clinically approved glycopeptides include teicoplanin (approved outside the United States), telavancin, dalbavancin, and oritavancin (FDA-approved in the United States), which are lipoglycopeptide derivatives featuring hydrophobic moieties that enhance membrane anchoring and activity against resistant strains. These agents are primarily administered intravenously for skin and soft tissue infections, pneumonia, and bloodstream infections due to Gram-positive organisms, including vancomycin-resistant enterococci (VRE), though oral vancomycin is specifically used for Clostridium difficile-associated diarrhea by targeting the gut lumen without systemic absorption. Their rigid heptapeptide core structure, often with vancosamine sugar substituents, enables high-affinity binding, but bacterial resistance—mediated by genes like vanA that modify the peptidoglycan precursor to D-Ala-D-Lac, reducing affinity by up to 1,000-fold—poses a significant clinical challenge and drives ongoing research into semisynthetic modifications. Despite these hurdles, glycopeptides continue to play a critical role in antimicrobial stewardship, particularly as last-resort options for life-threatening infections where other therapies fail.

Introduction

Definition and characteristics

Glycopeptide antibiotics are a class of natural antibiotics characterized by a glycosylated core, consisting of a heptapeptide aglycone backbone modified with moieties such as vancosamine and glucose. These compounds are primarily produced by actinomycete bacteria, including species of the Amycolatopsis, through pathways that involve large multimodular enzyme complexes. This biosynthetic process assembles the peptide from and subsequently attaches sugars to enhance and . Key characteristics of glycopeptide antibiotics include their high molecular weight, typically ranging from 1,500 to 2,000 , which contributes to their and results in poor oral , with rates often less than 1% when administered orally. Due to this and large size, they are generally administered parenterally for systemic . They exhibit activity predominantly against by interfering with cell wall synthesis, acting as bactericidal agents against most susceptible organisms, though bacteriostatic effects can occur in certain cases like enterococcal . In distinction from other antibiotic classes, glycopeptide antibiotics do not mimic substrates to inhibit enzymes like beta-lactams, which target ; instead, they directly bind to precursors. Unlike aminoglycosides, which disrupt bacterial protein synthesis by binding to the ribosomal subunit, glycopeptides focus on assembly without ribosomal interference, providing a complementary role in treating Gram-positive infections.

Medical significance

Glycopeptide antibiotics play a critical role in clinical practice as drugs of last resort for treating severe infections caused by multidrug-resistant Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant enterococci (VRE), and Clostridium difficile. These agents are particularly vital for managing life-threatening conditions where other antibiotics fail, such as bacteremia, endocarditis, and skin and soft tissue infections in hospitalized patients. Their importance stems from the rising prevalence of antimicrobial resistance, which has made them indispensable in combating pathogens that contribute to high morbidity and mortality in healthcare settings. On a scale, glycopeptide antibiotics are essential for addressing hospital-acquired infections, which account for significant healthcare burdens worldwide. , the prototype glycopeptide, has been included on the World Health Organization's Model List of Essential Medicines since its early iterations, underscoring its ongoing relevance in resource-limited settings and critical care. By enabling effective treatment of resistant Gram-positive infections, these antibiotics help reduce associated mortality rates, with studies indicating improved outcomes in patients with MRSA bacteremia when glycopeptides are appropriately utilized. This epidemiological impact is amplified in intensive care units, where such infections are common and can prolong hospital stays and increase death risks. Despite their efficacy, glycopeptide antibiotics have a narrow spectrum of activity, primarily limited to Gram-positive organisms due to the impermeability of the Gram-negative bacterial outer membrane, which prevents adequate drug penetration. This limitation necessitates their reserved use under antibiotic stewardship guidelines to preserve effectiveness and minimize resistance development, positioning them as targeted therapies rather than broad-spectrum options.60003-9/fulltext) Economically, the global market for glycopeptide antibiotics reflects their clinical value, projected to reach approximately USD 3.78 billion in 2025, driven by a compound annual growth rate (CAGR) of 7.0% amid increasing resistance challenges.

Structure and classification

Chemical structure

Glycopeptide antibiotics are characterized by a rigid heptapeptide backbone composed of seven , including non-proteinogenic residues such as 3-hydroxytyrosine, 4-hydroxyphenylglycine, and β-hydroxytyrosine, which form the core aglycone . This backbone is extensively cross-linked through biaryl and diphenylether bonds—typically between rings A-B (biaryl), C-O-D (diphenylether), and D-O-E (diphenylether)—creating a cup-shaped, tricyclic or tetracyclic scaffold that provides structural rigidity essential for their function. At least three such cross-links are required to maintain the molecule's active conformation. Glycosylation occurs primarily at amino acid positions 4, 6, and sometimes 7, involving the attachment of amino sugars like L-vancosamine or its , often as part of disaccharides such as vancosamine-glucose or mannose-linked variants, which enhance aqueous and contribute to target specificity. These sugar moieties are typically added via enzymes during and can be removed under acidic conditions, yielding the aglycone core. Key functional groups include chlorine atoms substituted on aromatic rings—for instance, at positions 2 and 6 in representative structures—along with , , and hydroxyl groups that influence polarity and potential. The chain incorporates D-amino acids, such as D-leucine at the , which are introduced through epimerization during non-ribosomal . Structural variations exist across glycopeptide classes, ranging from fully cyclic forms with multiple cross-links to less rigid linear precursors, with molecular weights around 1,144 Da for typical examples like the aglycone, devoid of sugars. These differences arise from modifications in cross-linking patterns and substituents, classifying them into five main types based on ring systems and residues at positions 1 and 3.

Types and examples

Glycopeptide antibiotics are classified into natural and semi-synthetic categories based on their origin and structural modifications, with further subdivision into structural types I through V according to variations in residues and substituents on the core heptapeptide backbone. Natural glycopeptides are produced by actinomycetes bacteria and include type I examples like , isolated in 1953 from Amycolatopsis orientalis (formerly orientalis), which features at position 1 and at position 3. Type III natural glycopeptides, such as discovered in the late from Actinoplanes teichomyceticus, are characterized by conferred by a chain attached to an acylglucosamine at position 4, enabling longer half-lives compared to . Semi-synthetic glycopeptides, often termed lipoglycopeptides, are derived from natural scaffolds through chemical modifications to enhance potency against resistant strains, typically by adding lipophilic tails that promote bacterial disruption in addition to cell wall inhibition. These include derivatives of , such as telavancin, approved by the FDA in 2009, which incorporates a decylaminoethyl for dual membrane-depolarizing activity. Dalbavancin, a semi-synthetic analog of approved in 2014, features amide modifications that extend its plasma half-life to over two weeks, allowing infrequent dosing. , also approved in 2014 and derived from chlorobiphenyl , includes a chlorobiphenylmethyl group that enables multiple mechanisms, including inhibition of transglycosylation and perturbation. Other natural subtypes include type I glycopeptides like balhimycin, isolated from Amycolatopsis balhimycina, which possesses an additional sugar on the at position 4 but remains non-clinical due to limited development. Type II examples, such as from Amycolatopsis lurida, feature a (β-D-mannosyl-(1→2)-α-L-rhamnosyl) at position 6; however, it induces platelet aggregation, limiting its therapeutic use.
ExampleOrigin (Type)Base ScaffoldApproval Date (FDA)Primary Modifications
VancomycinNatural (Type I)A. orientalis1958None (core structure)
TeicoplaninNatural (Type III)A. teichomyceticus1988 (Europe; not FDA)Fatty acid chain on acylglucosamine
TelavancinSemi-synthetic (Type I derivative)VancomycinSeptember 2009Decylaminoethyl lipophilic tail
DalbavancinSemi-synthetic (Type III derivative)TeicoplaninMay 2014Amide groups for extended half-life
OritavancinSemi-synthetic (Type I derivative)VancomycinAugust 2014Chlorobiphenylmethyl group
BalhimycinNatural (Type I)A. balhimycinaNoneAdditional mannose on glucosamine
RistocetinNatural (Type II)A. luridaNoneDisaccharide (rhamnosyl-mannosyl) at position 6

Mechanism of action

Binding to bacterial targets

Glycopeptide antibiotics exert their antibacterial activity by specifically targeting Lipid II, the undecaprenyl-pyrophosphate-linked precursor of in bacterial cell walls, through binding at its D-alanyl-D-alanine (D-Ala-D-Ala) terminus. This interaction sequesters the precursor, preventing its incorporation into the growing layer. The binding is highly specific and occurs via a of hydrogen bonds formed between the amide carbonyl groups on the antibiotic's backbone and the carboxyl and amide groups of the D-Ala-D-Ala . For , the prototype glycopeptide, this association is characterized by an affinity constant of approximately $10^6 M^{-1}, reflecting strong but reversible binding under physiological conditions. The structural foundation of this lies in the rigid, cup-shaped aglycone moiety of the glycopeptide, which envelops the D-Ala-D-Ala terminus like a molecular clasp, positioning the hydrogen-bonding residues optimally for . This conformational rigidity, derived from the cross-linked heptapeptide , ensures precise and steric occlusion of the target, thereby inhibiting downstream enzymatic processing. Cooperative effects, such as dimerization of the on the bacterial surface, can further enhance to Lipid II clusters. In response to resistance mechanisms that modify the peptidoglycan terminus, second-generation glycopeptides have been engineered to accommodate altered substrates, such as D-Ala-D-Ser or D-Ala-D-Lac in vancomycin-resistant strains. These variants maintain hydrogen bonding but with adjusted geometries to restore affinity against modified targets. Additionally, derivatives like telavancin incorporate a lipophilic that enables hydrophobic interactions with the bacterial , augmenting the primary Lipid II binding and contributing to depolarization.

Inhibition of cell wall synthesis

Glycopeptide antibiotics exert their primary effect by disrupting biosynthesis through dual inhibition of key enzymatic steps. Following binding to the D-Ala-D-Ala terminus of the precursor Lipid II, these antibiotics sterically hinder the transglycosylase enzyme, which is responsible for polymerizing and N-acetylmuramic acid into linear chains, thereby preventing elongation of the backbone. Simultaneously, they block the transpeptidase (also known as transpeptidase or penicillin-binding protein), which catalyzes the cross-linking of stems between chains, resulting in an uncross-linked and fragile structure. This inhibition can be represented simply as: \text{Lipid II (with D-Ala-D-Ala)} + \text{Glycopeptide} \rightarrow \text{Inactive complex (no transglycosylation or transpeptidation)} The bactericidal consequences arise from the accumulation of unutilized peptidoglycan precursors, such as Lipid II, which weakens the cell wall in Gram-positive bacteria and triggers autolytic enzymes to degrade the existing peptidoglycan layer, leading to cell lysis. This process is particularly pronounced in staphylococci and other Gram-positives, where the thick peptidoglycan layer is essential for osmotic integrity, and the resulting imbalance promotes rapid cell death through concentration-dependent killing. Glycopeptide antibiotics are most effective against actively dividing bacterial cells, as peptidoglycan synthesis is upregulated during growth and division to support formation and cell expansion. For , the classic glycopeptide, killing is time-dependent, relying on prolonged exposure above the to sustain inhibition. In contrast, lipoglycopeptides such as telavancin and demonstrate concentration-dependent bactericidal activity, enhanced by additional membrane-disrupting effects that amplify compromise at higher doses.

Clinical use

Indications and spectrum

Glycopeptide antibiotics are primarily indicated for the treatment of serious infections caused by multidrug-resistant , particularly when are ineffective or contraindicated. Key indications include skin and soft tissue infections (SSTIs), such as acute bacterial skin and skin structure infections (ABSSSI), , and bacteremia due to methicillin-resistant Staphylococcus aureus (MRSA) or coagulase-negative staphylococci. For instance, is recommended as a first-line agent for MRSA-associated SSTIs, , and bacteremia in adults, per Infectious Diseases Society of America (IDSA) guidelines. Additionally, is preferred over for the initial episode of nonsevere Clostridioides difficile infection in adults, with oral as an acceptable alternative; both are options for recurrent episodes depending on prior therapy and severity. The spectrum of activity of glycopeptide antibiotics is restricted to Gram-positive organisms, with no clinically significant effect against due to their inability to penetrate the outer membrane. They exhibit potent activity against staphylococci (including MRSA and methicillin-susceptible S. aureus [MSSA]), streptococci, and enterococci (including vancomycin-susceptible strains). Some glycopeptides, such as , also demonstrate activity against certain anaerobes like difficile. Newer agents like dalbavancin and retain this Gram-positive focus but offer enhanced potency against resistant strains, such as vancomycin-intermediate S. aureus (VISA). Despite their utility, glycopeptide antibiotics have notable limitations that restrict their broader application. They exhibit poor tissue penetration into the (CSF), lungs, and abscesses, making them suboptimal for , , or deep-seated abscesses unless combined with other agents. IDSA guidelines emphasize that glycopeptides should not be used routinely for in non-severe or prophylaxis due to the risk of promoting and their narrower compared to beta-lactams. For example, while is used for MRSA , its efficacy is limited by inadequate lung penetration, often necessitating alternatives like in such cases. Dalbavancin, approved for ABSSSI, provides a convenient single-dose option but is similarly confined to Gram-positive SSTIs.

Therapeutic guidelines

Glycopeptide antibiotics, such as , are recommended to be reserved for the treatment of serious Gram-positive infections, particularly those involving methicillin-resistant Staphylococcus aureus (MRSA) or when fail due to resistance or patient allergy, as part of efforts to minimize resistance development and optimize outcomes. The 2011 IDSA guidelines for MRSA infections emphasize preferring beta-lactams for methicillin-susceptible strains unless contraindicated, positioning glycopeptides as second-line agents in such cases to reduce unnecessary broad-spectrum use. For serious infections like MRSA bacteremia or , the 2020 ASHP/IDSA/PIDS/SIDP consensus guidelines advocate (TDM) of to target trough concentrations of 15-20 mg/L, which correlates with an to () ratio of at least 400 to ensure efficacy while minimizing risk. Monitoring should commence within 48 hours of initiation, with adjustments based on serial levels to maintain these targets, particularly in critically ill patients where subtherapeutic exposure increases treatment failure rates. Combination therapy involving glycopeptides with s is indicated for synergistic bactericidal activity in specific scenarios. For enterococcal caused by -susceptible strains, if s are contraindicated, the guidelines recommend a double regimen such as plus ; in -allergic patients, plus an (if feasible) is used. This approach enhances clearance of persistent bacteremia compared to monotherapy, though it requires close monitoring for additive toxicities like renal impairment. In special populations, dosing adjustments are essential to achieve therapeutic targets safely. For obese adults, the 2020 vancomycin guidelines suggest empiric maintenance doses not exceeding 4,500 mg/day based on total body weight and renal function, often requiring Bayesian pharmacokinetic models for precise estimation to avoid under- or overdosing. Patients with renal impairment necessitate dose reductions proportional to creatinine clearance, with initial loading doses of 20-35 mg/kg followed by interval extensions to prevent accumulation and . In , vancomycin is dosed by actual body weight at 60 mg/kg/day divided every 6 hours for children over 1 year, with TDM targeting similar adult / goals, and higher rates (up to 70-80 mg/kg/day) considered for neonates or those with augmented clearance.

Pharmacology and administration

Routes of administration

Glycopeptide antibiotics are primarily administered intravenously for the treatment of systemic infections due to their large molecular size and poor oral bioavailability. Intravenous infusion is the standard route, as it ensures adequate systemic exposure; for instance, vancomycin is typically infused over 1 to 2 hours to minimize infusion-related reactions such as histamine release. Similarly, dalbavancin is given as a single intravenous dose infused over 30 minutes, leveraging its extended half-life for once-weekly or single-dose regimens in skin and soft tissue infections. Teicoplanin can also be administered intravenously, either as a bolus over 3 to 5 minutes or as a 30-minute infusion. Telavancin and oritavancin are administered solely intravenously, with telavancin infused over 60 minutes and oritavancin over 3 hours to avoid infusion reactions. Oral administration is limited to specific indications where systemic is not required, such as gastrointestinal infections. , for example, is used orally at a dose of 125 four times daily for Clostridioides difficile-associated , as it remains in the gut lumen due to negligible . This route is ineffective for systemic infections because of the drug's poor . Intramuscular administration is uncommon for most glycopeptides owing to local pain and tissue irritation. Teicoplanin is an exception, where it can be given intramuscularly as a single daily dose after an initial loading regimen, though injection-site pain may occur. Vancomycin intramuscular use is rare and discouraged due to risks of pain, tenderness, and necrosis. Topical applications remain experimental and are not clinically established for glycopeptide antibiotics. These agents are typically supplied as lyophilized powders that require reconstitution in sterile or saline prior to intravenous use, ensuring and proper .

Pharmacokinetics and dosing

Glycopeptide antibiotics are poorly absorbed from the and are primarily administered intravenously to achieve therapeutic concentrations. They exhibit limited , with the majority of the dose excreted unchanged via the kidneys, necessitating adjustments in patients with impaired renal . The volume of distribution for these agents typically ranges from 0.4 to 1 L/kg, reflecting distribution primarily into . For , the elimination in adults with normal renal function is approximately 4 to 11 hours, with clearance predominantly renal (over 80% of the dose excreted unchanged in ). Dalbavancin, a lipoglycopeptide , has a markedly prolonged of 200 to 400 hours, enabling once-weekly dosing regimens due to its high (93%) and slow renal elimination. similarly features a long elimination of 70 to 170 hours in patients with normal renal function, also supporting less frequent administration. Telavancin has an elimination of about 7 to 9 hours with ~99% protein binding and primarily renal excretion. exhibits a biphasic elimination with a of approximately 130 hours, high protein binding (>90%), and non-linear due to tissue distribution. Standard dosing for involves a of 25 to 30 mg/kg (actual body weight) followed by maintenance doses of 15 mg/kg every 12 hours, adjusted to achieve target area under the curve () values. For , typical regimens use 6 to 12 mg/kg daily after an initial loading phase. Dalbavancin dosing consists of 1,500 mg intravenously on day 1, with an optional 1,500 mg dose one week later for certain infections. Telavancin is dosed at 10 mg/kg intravenously once daily, with adjustments for renal impairment. is administered as a single 1,200 mg intravenous dose for skin and infections. Dosing adjustments for glycopeptides are primarily based on clearance (CrCl), with reductions recommended when CrCl falls below 50 mL/min; for example, maintenance intervals may extend to every 24 to 48 hours in moderate impairment. Therapeutic monitoring for often employs Bayesian pharmacokinetic models to estimate the 24-hour , calculated as AUC = dose / clearance, targeting 400 to 600 mg·h/L to optimize while minimizing .

Adverse effects

Common side effects

Glycopeptide antibiotics, particularly , are associated with infusion-related reactions primarily due to rapid intravenous administration. The most common is red man syndrome, characterized by release leading to flushing, pruritus, erythematous on the upper body, and occasionally . This non-immunologic reaction occurs in approximately 3.7% to 47% of patients receiving infusions, with higher rates linked to faster infusion speeds exceeding 1 g over 60 minutes. Slowing the infusion rate to over 2 hours or premedicating with antihistamines can mitigate these symptoms in most cases. Oral formulations of glycopeptides, such as used for infections, frequently cause gastrointestinal disturbances including , , and abdominal discomfort. Additionally, patients often report a bitter or unpleasant (), which is unique to and may contribute to poor . These effects are generally mild and self-limiting, resolving upon discontinuation. Mild reactions to glycopeptides manifest as maculopapular rashes, urticaria, pruritus, or low-grade fever, affecting a small percentage of patients. These are typically delayed and IgE-mediated in rare instances, but with is negligible due to structural differences, allowing safe use in penicillin-allergic individuals. Hematologic effects include reversible , reported in 1% to 7% of vancomycin-treated patients, often appearing after several days of therapy and resolving upon withdrawal. may also occur, particularly in association with reactions, though its isolated incidence remains low and is usually . Monitoring complete blood counts is recommended during prolonged use to detect these changes early.

Serious toxicities

Nephrotoxicity is a major serious adverse effect of glycopeptide antibiotics, particularly , manifesting as (AKI) through mechanisms involving , free production, and mitochondrial dysfunction in renal cells. The incidence of vancomycin-induced AKI ranges from 5% to 15% in patients receiving high doses, with rates escalating to 30-40% in those on aggressive dosing regimens, and meta-analyses indicate a of approximately 2.45 for AKI development compared to alternative therapies. Key risk factors include elevated trough concentrations (>15-20 mg/L), prolonged therapy (>7 days), concurrent use of nephrotoxic agents such as aminoglycosides or piperacillin-tazobactam, and patient factors like advanced age or admission, where AKI incidence can reach 10%. Ototoxicity represents another potentially life-threatening toxicity, primarily affecting the and , leading to irreversible or balance disturbances, though it occurs infrequently with modern dosing. The overall incidence is low, estimated at around 1% or less, but audiometric studies reveal subclinical high-frequency in up to 12% of older patients on prolonged vancomycin monotherapy. Risk factors encompass peak serum levels exceeding 40 mg/L (with some reports citing thresholds as low as 30 mg/L), treatment duration beyond 7-14 days, advanced age (>53 years), and concomitant ototoxic drugs, though long-term use does not consistently elevate risk in all populations. Additional serious toxicities include , which develops in approximately 2% to 8% of vancomycin-treated patients and may involve , as well as rare anaphylactic reactions characterized by severe manifesting as , , or urticaria. risks exist with , particularly for neutropenia and , with studies reporting adverse drug reactions in up to 35% of patients switching from vancomycin due to intolerance. Notably, teicoplanin carries a lower of compared to , with incidence rates often below 5% in similar patient cohorts.

Lipoglycopeptide derivatives

Lipoglycopeptide antibiotics such as telavancin, dalbavancin, and generally have adverse effect profiles similar to but with notable differences. Telavancin is associated with higher (up to 10-20% in some studies), prolongation, and increased mortality in patients with pre-existing renal impairment ( clearance ≤50 mL/min). Dalbavancin and exhibit lower rates of and red man syndrome but may cause mild gastrointestinal effects, headache, infusion-site reactions, and transient elevations in liver enzymes; serious and difficile-associated risks remain low. Management of these toxicities emphasizes (TDM), with current guidelines (as of 2020, endorsed through 2025) preferring area under the curve to minimum inhibitory concentration (AUC/MIC) ratio-guided monitoring targeting 400-600 mg·h/L for serious infections to minimize , over traditional trough levels (10-15 mg/L for non-severe, 15-20 mg/L for serious infections) which are associated with higher AKI risk. Peak levels >40-60 mg/L should be avoided to reduce . Adequate , avoidance of concomitant nephrotoxins, and prompt discontinuation upon suspicion of are essential. For , TDM targeting troughs of 20-40 mg/L for severe infections further minimizes risks.

Antimicrobial resistance

Mechanisms of resistance

Bacterial resistance to glycopeptide antibiotics primarily arises through modifications to the precursors in the , which prevent the drugs from binding effectively to their target site, the D-alanyl-D-alanine (D-Ala-D-Ala) . Glycopeptides such as exert their bactericidal action by forming s with this , inhibiting . The most prominent involves the VanA and VanB phenotypes, predominantly observed in enterococci. These phenotypes are mediated by acquired clusters that reprogram the of precursors. Specifically, enzymes encoded by vanA or vanB s catalyze the replacement of the normal D-Ala-D-Ala with D-alanyl-D-lactate (D-Ala-D-Lac). This reduces the binding affinity of by approximately 1,000-fold due to the loss of a critical and steric hindrance, rendering the ineffective at physiological concentrations. The VanA cluster confers high-level resistance to both and , while VanB provides resistance primarily to . Accompanying enzymes, such as VanH (a that produces D-lactate) and VanX (a D,D-dipeptidase that hydrolyzes residual D-Ala-D-Ala), ensure the selective incorporation of the modified precursor. Additional resistance strategies include structural alterations to the and . The , often clustered with vanA, contributes to low-level against teicoplanin-like lipoglycopeptides by reducing to the cell surface, possibly through modifications to anchoring or lipidation sites, though it has minimal on . In some strains, is enhanced by thickened cell walls, which sequester the in non-lethal outer layers, a phenomenon known as drug trapping; this traps excess glycopeptide molecules via free D-Ala-D-Ala ends without affecting the functional precursors. formation in staphylococci further promotes by creating a protective matrix that impedes glycopeptide penetration to bacterial cells. The genetic foundation of these mechanisms lies in mobile van operons, such as vanA and vanB, which are typically located on plasmids or transposons like Tn1546. These elements are often acquired from glycopeptide-producing actinomycetes in the environment, where they serve as self-protection mechanisms, and are disseminated horizontally among Gram-positive pathogens via conjugation or . In , resistance manifests differently through and heterogeneous VISA (hVISA) phenotypes, which do not involve van operons but rather mutations leading to alterations. VISA strains exhibit thickened s (up to twofold increase in layer) due to reduced autolysis, decreased cross-linking, and upregulated synthesis of wall teichoic acids, resulting in intermediate minimum inhibitory concentrations (4–8 μg/mL). hVISA represents a precursor state with subpopulations exhibiting variable resistance, often driven by mutations in regulatory genes like vraSR, walkR, or rpoB, which coordinately enhance turnover and trapping of in the outer layers.

Prevalence and management

Vancomycin-resistant Staphylococcus aureus (VRSA) remains exceedingly rare, with the first case identified in the United States in 2002 and 16 confirmed cases reported there as of 2022. In contrast, vancomycin-resistant enterococci (VRE) are far more prevalent in healthcare settings, with approximately 30% of healthcare-associated enterococcal infections resistant to vancomycin, and rates among hospital Enterococcus faecium isolates often ranging up to 40% or more in high-burden facilities. The prevalence of heterogeneous vancomycin-intermediate S. aureus (hVISA) has been rising gradually, increasing from about 4% before 2010 to 5.3% among S. aureus isolates between 2010 and 2019, with more recent studies from 2023–2025 reporting rates up to 22% in specific hospital settings and continued concerns. Key risk factors for acquiring glycopeptide resistance include prolonged exposure to and other antibiotics, inadequate control measures such as lapses in hand hygiene, and patient transfers between hospital units or facilities. These risks are amplified in intensive care units (ICUs), where rates of VRE and are notably higher due to the presence of immunocompromised patients, invasive devices like catheters, and frequent surgical interventions. Effective management of glycopeptide-resistant infections begins with routine antimicrobial susceptibility testing, using Clinical and Laboratory Standards Institute (CLSI) (MIC) breakpoints where isolates with MICs of ≤2 mg/L are considered susceptible for S. aureus. For VRE infections, alternatives such as , , or are recommended based on local susceptibility patterns and site, often guided by infectious disease consultation to optimize outcomes. control protocols, including contact precautions and environmental cleaning, are essential to limit transmission in hospital settings. Surveillance data from the Centers for Disease Control and Prevention (CDC) and the indicate stable but persistently concerning rates of methicillin-resistant S. aureus (MRSA) with reduced susceptibility, alongside an increasing trend in -resistant E. faecium bloodstream infections across the /Economic European Area from 2019 to 2023. Post-2023 reports highlight emerging community-associated VanA-type resistance in enterococci, exemplified by the spread of novel sequence type 1299 (ST1299) lineages in regions like , potentially signaling a shift from predominantly hospital-acquired patterns.

History

Discovery of vancomycin

Vancomycin, the prototype glycopeptide antibiotic, was isolated in 1953 by organic chemist Edmund C. Kornfeld and his team at from a soil sample collected in the interior jungles of . The producing was an actinomycete bacterium initially classified as Streptomyces orientalis and later reclassified as Amycolatopsis orientalis. The compound, originally labeled as 05865, was renamed , a term derived from "vanquish" to signify its ability to defeat bacterial infections, combined with the suffix "-mycin" common for antibiotics from actinomycetes. Early characterization revealed vancomycin's potent activity against , including penicillin-resistant staphylococci. In 1956, microbiologist M. H. McCormick and colleagues at reported its chemical and biological properties, demonstrating bactericidal effects in preclinical and animal models against staphylococcal infections. Due to the rising prevalence of staphylococcal resistance in the 1950s, the U.S. granted vancomycin fast-track approval in 1958 for intravenous administration in treating severe systemic infections. Initial clinical use was hampered by the drug's impure formulations, which contained up to 70% contaminants and imparted a brown, appearance—earning it the derogatory nickname "Mississippi ." These impurities contributed to high rates of toxicity, including and , limiting its early adoption despite efficacy. Structural elucidation began in the mid-1950s with partial studies but faced challenges due to the molecule's as a glycosylated heptapeptide; the complete structure was not fully determined until , when corrections to earlier models confirmed its tricyclic rigid scaffold.

Development of derivatives

Following the discovery of , efforts to develop glycopeptide derivatives began in the to address limitations such as tolerability and spectrum of activity. , the first major derivative, was isolated in 1978 from the actinomycete Actinoplanes teichomyceticus by researchers at the Lepetit Research Laboratories in . This lipoglycopeptide complex demonstrated improved pharmacokinetic properties, including longer and reduced compared to , making it suitable for once-daily dosing. received its first marketing approval in in 1987 and was subsequently approved across in 1988 for treating serious Gram-positive infections. In the , the emergence of vancomycin-resistant enterococci (VRE) prompted the development of semisynthetic glycopeptides with enhanced activity against resistant strains. These second- and third-generation agents incorporated modifications such as tails to improve anchoring and potency. Telavancin, developed by Theravance Biopharma, features a decylaminoethyl and was approved by the FDA in 2009 for complicated and structure infections and caused by Gram-positive pathogens, including MRSA. Dalbavancin and , both lipoglycopeptides, followed with FDA approvals in 2014 for acute bacterial and structure infections; dalbavancin allows for weekly dosing due to its long half-life, while provides single-dose treatment. These derivatives were designed to overcome VRE resistance mechanisms by altering binding affinity and adding bactericidal effects beyond inhibition. Key milestones in glycopeptide development included the total synthesis of vancomycin aglycon, first achieved in 1999 by independent groups led by , , and Dale L. Boger, enabling structure-activity relationship studies and analog design. The expiration of original patents on in the late facilitated the entry of generic formulations in the early , increasing accessibility amid rising demand. Regulatory approvals for these derivatives in the were spurred by the increasing prevalence of MRSA and VRE infections, with the FDA prioritizing agents active against multidrug-resistant under initiatives like the Qualified Infectious Disease Product designation. This progression marked a shift from natural products to engineered glycopeptides, enhancing options for resistant infections.

Research and future directions

New glycopeptide analogs

Since the approval of dalbavancin, , and telavancin around 2014, no major new glycopeptide antibiotics have received regulatory approval, though the pipeline includes candidates targeted at vancomycin-resistant enterococci (VRE). Promising experimental compounds include EVG7, a semisynthetic guanidino lipoglycopeptide developed in 2025 that demonstrates enhanced potency against while sparing beneficial family members in the , thereby preventing recurrent infections. In preclinical models, EVG7 exhibited superior efficacy compared to in treating infections, with minimal disruption to the . Another 2025 discovery is saarvienin A, a novel glycopeptide isolated from a rare Amycolatopsis strain in a mine, featuring a unique twisted scaffold that overcomes resistance mechanisms and shows potent activity against high-priority Gram-positive superbugs, including VRE and methicillin-resistant S. aureus. This compound binds differently from traditional glycopeptides, targeting resistant pathogens with minimum inhibitory concentrations (MICs) in the low microgram per milliliter range without cross-resistance to existing antibiotics. Kineomicins, also identified in 2025 from the rare actinomycete Actinokineospora auranticolor, represent a novel class of glycopeptides with an uncommon peptide scaffold derived from underexplored Pseudonocardiales genomes, offering potential against resistant through expanded biosynthetic diversity. Structural modifications, such as chlorination, have been explored to enhance binding affinity to the D-Ala-D-Lac terminus in resistant strains; for instance, trichlorinated derivatives alter target interactions and reduce resistance induction compared to parent compounds. In preclinical stages, strategies have generated numerous analogs with dual binding modes to D-Ala-D-Ala and D-Ala-D-Lac, achieving MICs below 1 mg/L against VRE and other resistant enterococci, thereby restoring activity lost in natural glycopeptides. These approaches, including peripheral lipidation and modifications, have yielded over 100 variants with synergistic mechanisms that disrupt multiple bacterial processes beyond inhibition.

Strategies to combat resistance

One key strategy to restore glycopeptide efficacy involves chemical modifications that enhance binding affinity or introduce additional mechanisms of action, countering mechanisms such as precursor remodeling. Chemical modifications often target the core to optimize hydrogen bonding interactions with altered precursors, such as D-Ala-D-Lac in VanA-type . For instance, replacing a in the backbone with a methylene unit eliminates electrostatic repulsion, resulting in a 40-fold increase in potency against VanA-resistant Enterococcus faecalis (MIC reduced to 31 μg/mL). Similarly, incorporating an amidine group into the binding pocket allows dual of D-Ala-D-Ala and D-Ala-D-Lac termini, achieving MICs as low as 0.5 μg/mL against resistant enterococci. These semisynthetic approaches, achieved through total or chemoenzymatic synthesis, prioritize structural tweaks that maintain the core scaffold while improving specificity. Another modification strategy incorporates quaternary ammonium groups at the to promote and increased permeability, complementing the primary inhibition of cell wall synthesis. This dual-action mechanism disrupts bacterial independently of targets, enhancing activity against Gram-positive pathogens with reduced susceptibility. Such modifications have been shown to induce cell wall permeability, providing synergistic effects that lower MICs against resistant strains. Combination therapies leverage synergistic interactions to overcome resistance without altering the glycopeptide itself. Pairing glycopeptides with β-lactam antibiotics, such as ceftaroline and , demonstrates synergy against methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-intermediate strains, often reducing bacterial burdens by 2–4 log10 CFU/mL in time-kill assays. This synergy arises from complementary inhibition of synthesis, where β-lactams bind altered in resistant isolates, restoring glycopeptide access. Clinical reports confirm improved outcomes in persistent MRSA bacteremia when using vancomycin-ceftaroline combinations compared to monotherapy. Efflux pump inhibitors represent an emerging adjunct to glycopeptide therapy, particularly against staphylococci where multidrug efflux contributes to reduced intracellular accumulation. Compounds like phenylalanine-arginine-β-naphthylamide (PAβN) potentiate vancomycin by blocking efflux pumps such as NorA in S. aureus, lowering MICs by 4- to 8-fold in resistant isolates. Although primarily studied in preclinical models, these combinations aim to mitigate low-level resistance mechanisms that exacerbate target-based alterations. Biosynthetic engineering expands glycopeptide diversity by manipulating producer strains to generate variants active against resistant targets. Genome mining of uncultured microbial communities via has uncovered novel synthetase (NRPS) gene clusters encoding glycopeptide-like scaffolds, revealing structural diversity in environmental Actinobacteria that evades common resistance pathways. Techniques like mutasynthesis feed modified precursors to engineered producers, incorporating fluorinated residues into the heptapeptide core to alter properties and enhance potency against Van-resistant enterococci. CRISPR-based editing of producer genomes enables precise reconfiguration of biosynthetic gene clusters for novel variants. In Amycolatopsis orientalis ( producer), CRISPR/Cas9 systems have been used to delete regulatory repressors or swap halogenase genes, yielding high-yield variants with modified chlorination patterns that improve activity against resistant . These approaches, combined with tools, facilitate rapid iteration of glycopeptide structures to address evolving resistance. Clinical trials are evaluating dual-action glycopeptides—those combining cell wall inhibition with membrane disruption—in phase II and III settings to treat complicated infections. A 2025 randomized trial (DOTS) of dalbavancin in patients with S. aureus bacteremia demonstrated noninferiority to standard therapy, with 70-day success rates of 73% versus 72% for comparator regimens, highlighting potential for single-dose administration to reduce recurrence. Post-2023 efforts focus on preventing recurrence in Clostridioides difficile infections (CDI), where oral or extended-release glycopeptide formulations are tested alongside probiotics. These trials emphasize outpatient regimens to minimize resistance selection.

References

  1. [1]
    Approved Glycopeptide Antibacterial Drugs: Mechanism of Action ...
    The glycopeptide antibiotics are a group of glycosylated cyclic or polycyclic nonribosomal peptides that inhibit Gram-positive bacterial cell-wall synthesis.
  2. [2]
    Glycopeptide and Lipoglycopeptide Antibiotics | Chemical Reviews
    This review will summarize what is known about the structure and function of the glycopeptide and lipoglycopeptide antibiotics, how glycopeptide resistance ...
  3. [3]
    Newest perspectives of glycopeptide antibiotics - PubMed Central
    Jan 3, 2023 · Glycopeptide antibiotics (GPAs) are a family of non-ribosomal peptide natural products with polypeptide skeleton characteristics, which are ...Introduction · Glycosylation Of Gpas · Vancomycin AnaloguesMissing: definition | Show results with:definition
  4. [4]
    Glycopeptide Antibiotic - an overview | ScienceDirect Topics
    Glycopeptide antibiotics are a class of antibiotics that are crucial for treating infections caused by Gram-positive bacteria, ...
  5. [5]
  6. [6]
    Glycopeptide Antibiotic - an overview | ScienceDirect Topics
    They have very poor oral absorption (less than 1% of a given dose) and must be given by the parenteral route. Their binding to plasma albumin is negligible ...
  7. [7]
    Glycopeptide antibiotics: Back to the future - Nature
    Aug 13, 2014 · Glycopeptide antibiotics have been a key weapon in the fight against bacterial infections for over half a century.
  8. [8]
    Developments in Glycopeptide Antibiotics - PMC - PubMed Central
    Glycopeptide antibiotics (GPAs) are a key weapon in the fight against drug resistant bacteria, with vancomycin still a mainstream therapy against serious Gram- ...Missing: definition | Show results with:definition
  9. [9]
    Vancomycin: A History | Clinical Infectious Diseases | Oxford Academic
    Vancomycin became available for clinical use >50 years ago but was soon discarded in favor of other antibiotics that were deemed to be more efficacious and ...
  10. [10]
    [PDF] WHO EML 23rd List (2023) - IRIS
    Medicines and dosage forms are listed in alphabetical order within each section and the order of listing does not imply preference for one form over another.
  11. [11]
    Limitations of presently available glycopeptides in the treatment of ...
    The glycopeptide antibacterial drugs vancomycin and teicoplanin are widely used in hospitals for therapy of severe or multiresistant Gram-positive infections.
  12. [12]
    Old and New Glycopeptide Antibiotics: Action and Resistance - PMC
    Glycopeptides are considered antibiotics of last resort for the treatment of life-threatening infections caused by relevant Gram-positive human pathogens.Missing: narrow | Show results with:narrow
  13. [13]
    Glycopeptide Antibiotics Market 2025 - Size, And Forecast
    It will grow from $3.54 billion in 2024 to $3.78 billion in 2025 at a compound annual growth rate (CAGR) of 7.0%. The growth in the historic period can be ...
  14. [14]
  15. [15]
    Vancomycin: The Tale of the Vanquisher and the Pyrrhic Victory - PMC
    Vancomycin was isolated in 1953 from a sample of dirt collected by a missionary in the interior jungle of Borneo. That sample contained the soil bacterium ...
  16. [16]
    Teicoplanin - an overview | ScienceDirect Topics
    Teicoplanin was first reported in 1978 by Italian researchers at Lepetit Research Center (Parenti et al., 1978) and then introduced to clinical use for treating ...
  17. [17]
    Drug Approval Package: Brand Name (Generic Name) NDA #
    Nov 24, 2009 · Vibativ (Telavancin) Injection Company: Theravance, Inc. Application No.: 022110. Approval Date: 9/11/2009 · Approval Letter(s) (PDF) · Summary ...
  18. [18]
    Drug Approval Package - accessdata.fda.gov
    Jun 24, 2014 · Dalvance (dalbavancin hydrochloride) Lyophilized Powder for Injection. Company: Durata Therapeutics. Application No.: 021883. Approval Date: 5/23/2014.
  19. [19]
    Dalbavancin - PMC - NIH
    It was approved by both the FDA in May 2014 and the EMA in February 2015 for the treatment of adult patients with acute bacterial skin and skin structure ...
  20. [20]
    [PDF] 206334Orig1s000 - accessdata.fda.gov
    Aug 6, 2014 · NDA 22,153 was submitted in February 2008 by Targanta Therapeutics Corp. seeking approval of oritavancin for the treatment of complicated skin ...<|separator|>
  21. [21]
    Balhimycin, a New Glycopeptide Antibiotic with an Unusual ...
    Highlighting the role of the chlorine substituents in the glycopeptide antibiotic balhimycin for chiral recognition in capillary electrophoresis. ... The ...
  22. [22]
    Abnormalities of factor VIII and platelet aggregation--use of ristocetin ...
    Mar 1, 1975 · Ristocetin was used to study platelet aggregation in platelet-rich plasma and to assay the von Willebrand factor activity of factor VIII ...
  23. [23]
    Therapeutic compounds targeting Lipid II for antibacterial purposes
    All glycopeptides antibiotics have in common that they inhibit the cell wall synthesis by binding to the carboxyl-terminal part of the pentapeptide of lipid II.
  24. [24]
    Molecular mechanisms of vancomycin resistance - Stogios - 2020
    Jan 3, 2020 · Vancomycin forms an intricate network of hydrogen bonds with the d-Ala-d-Ala region of Lipid II, interfering with the peptidoglycan layer maturation process.2 Vancomycin Resistance... · 2.1 D-Ala-D-Lac Based... · 2.2 D-Ala-D-Ser Based...
  25. [25]
    [PDF] Design and evaluation of analogues of the bacterial cell-wall ...
    Aug 28, 2007 · Keywords: Vancomycin; D-Ala-D-Ala derivatives; Microbiological assay ... measures of its affinity constant with vancomycin. (2.1 × 10 6 ...
  26. [26]
    Redesign of Glycopeptide Antibiotics: Back to the Future
    Feb 13, 2012 · Normal synthesis of lipid intermediate I and II, containing the d-Ala-d-Ala termini, continues but a late stage remodeling to d-Ala-d-Lac ensues ...
  27. [27]
    Telavancin Disrupts the Functional Integrity of the Bacterial ...
    The mechanism by which telavancin binds to and disrupts the function of the bacterial membrane has not yet been determined. Telavancin may bind to the membrane ...Missing: electrostatic | Show results with:electrostatic
  28. [28]
  29. [29]
  30. [30]
    Vancomycin - StatPearls - NCBI Bookshelf - NIH
    Vancomycin is a glycopeptide antibiotic that exerts its bactericidal effect by inhibiting the polymerization of peptidoglycans in the bacterial cell wall.[9] ...Missing: characteristics | Show results with:characteristics
  31. [31]
    Clinical Practice Guidelines by the Infectious Diseases Society of ...
    IV vancomycin 15–20 mg/kg/dose (actual body weight) every 8–12 h, not to exceed 2 g per dose, is recommended in patients with normal renal function (B-III). 61.
  32. [32]
    Clinical Practice Guidelines for the Management of Clostridioides ...
    Jun 14, 2021 · The previous iteration of the guidelines also recommends vancomycin (500 mg 4 times daily orally or by nasogastric tube) rather than fidaxomicin ...Background · Methodology · Full Recommendation on the... · Notes
  33. [33]
    Therapeutic monitoring of vancomycin for serious methicillin ... - IDSA
    Mar 19, 2020 · This document is an executive summary of the new vancomycin consensus guidelines for vancomycin dosing and monitoring.
  34. [34]
    Infective Endocarditis in Adults: Diagnosis, Antimicrobial Therapy ...
    For enterococci, bactericidal activity can be achieved by the combination of certain β-lactam antibiotics (eg, penicillin, ampicillin, and piperacillin) with an ...
  35. [35]
    [PDF] therapeutic-guidelines-monitoring-vancomycin-ASHP-IDSA-PIDS.pdf
    Mar 19, 2020 · ized clinical trials, vancomycin loading doses can be considered in the treatment of serious MRSA infections. Vancomycin should be ...
  36. [36]
    Red man syndrome - PMC - PubMed Central - NIH
    In summary, each intravenous dose of vancomycin should be administered over at least a 60 min interval to minimize the infusion-related adverse effects [20].
  37. [37]
    [PDF] Dalbavancin (Dalvance) February 2019 Non-formulary (OP) Criteria ...
    Feb 10, 2019 · Dalbavancin is administered as an IV infusion delivered over 30 minutes in order to prevent infusion-related reactions. If given too quickly, “ ...
  38. [38]
    [PDF] Targocid 400 mg lyophilised powder for injection with ... - Medsafe
    Teicoplanin can be administered either intravenously or intramuscularly. Intravenous dosing may be by rapid injection over 3-5 minutes, or more slowly over ...
  39. [39]
    Intravenous Vancomycin | Dosing Guidelines, Trough Levels, Side ...
    Oral vancomycin is given to treat Clostridiodes (Clostridium) difficile infections and infection-induced diarrhea. However, the drug has poor oral ...
  40. [40]
    Teicoplanin | Johns Hopkins ABX Guide
    Oct 18, 2022 · Pain at the injection site if given by IM injection. OCCASIONAL. OCCASIONAL. OCCASIONAL. Thrombophlebitis; Nephrotoxicity, especially with high ...
  41. [41]
    Glycopeptide antibiotic drug stability in aqueous solution - PMC
    Dec 12, 2022 · Vancomycin is a tricyclic glycopeptide comprising a heptapeptide chain that forms a tricyclic structure along with an attached disaccharide that ...Missing: bioavailability | Show results with:bioavailability
  42. [42]
    Pharmacokinetics of Vancomycin in Patients with Different Renal ...
    The estimated typical population clearance of vancomycin was lower in the patients with normal renal function (0.655 L/h) compared to the patients with impaired ...Missing: half- life<|control11|><|separator|>
  43. [43]
    Population pharmacokinetic model of Vancomycin based on ...
    In adults with normal renal function, elimination half-life (t1/2) is from 4 to 6 h, while in patients with severe renal impairment, it could be prolonged up ...
  44. [44]
    Vancomycin: Uses, Interactions, Mechanism of Action - DrugBank
    Half-life in normal renal patients is approximately 6 hours (range 4 to 11 hours). In anephric patients, the average half-life of elimination is 7.5 days.
  45. [45]
    Dalbavancin: Uses, Interactions, Mechanism of Action - DrugBank
    Mar 19, 2008 · Dalbavancin should be administered over 30 minutes by intravenous infusion Label,5.Identification · Pharmacology · Interactions · ProductsMissing: formulation | Show results with:formulation<|separator|>
  46. [46]
    Clinical pharmacokinetics of teicoplanin - PubMed
    The initial volume of distribution is 0.089 L/kg, the volume of distribution at steady-state is 0.86 L/kg, clearance is 0.0114 L/h/kg and renal clearance is 0. ...
  47. [47]
    [PDF] vancomycin dosing and monitoring guidelines - UC Davis Health
    ... kg / dose Maintenance Dose ~ 15mg/kg / dose. > 90. 1250. 1500. 76-90. 1000 ... Vancomycin Loading Dose: 1-2g (15-25mg/kg):. If > 1.5g consider two divided ...
  48. [48]
    Pharmacokinetics of teicoplanin in patients undergoing chronic ...
    It is proposed that teicoplanin should be administered at a dose of 10 mg/kg at 48–72 h intervals, in patients undergoing chronic haemodialysis for the therapy ...
  49. [49]
    Once-Weekly Dalbavancin versus Daily Conventional Therapy for ...
    Jun 5, 2014 · Dalbavancin has a terminal half-life of 2 weeks, related in part to 93% protein binding, and is more efficacious in animal models when ...
  50. [50]
    [PDF] Drug dosing consideration in patients with acute and chronic kidney ...
    Drug dosage adjustment for patients with acute or chronic kidney disease is an accepted standard of practice. The challenge is how to accurately estimate a ...
  51. [51]
    [PDF] Vancomycin Pharmacokinetic Calculations
    To be accurate, 2-4 half-lives should occur between the levels.1. • In pharmacokinetic calculations, the term e-kel(τ) represents the fraction of the serum.
  52. [52]
    Vancomycin (oral route) - Side effects & dosage - Mayo Clinic
    Vancomycin, when taken by mouth, is used to treat Clostridioides difficile-associated diarrhea (also called C diff).
  53. [53]
    Glycopeptide Hypersensitivity and Adverse Reactions - PMC - NIH
    Apr 21, 2020 · Common adverse effects associated with glycopeptide use include nephrotoxicity, ototoxicity, and Redman Syndrome (RMS).
  54. [54]
    Incidence and risk factors of Vancomycin-induced thrombocytopenia
    Jan 2, 2025 · Among the 1269 identified patients, the incidence of thrombocytopenia was 3.3% (42/1269) after a medium of 9 days (range, 2 to 22) of the ...
  55. [55]
    Vancomycin - LiverTox - NCBI Bookshelf - NIH
    Aug 5, 2025 · Vancomycin is largely well tolerated; common side effects include diarrhea, nausea, nephrotoxicity and neutropenia.
  56. [56]
    Vancomycin-induced nephrotoxicity in non-intensive care unit ...
    Oct 19, 2021 · Studies have suggested that vancomycin could induce nephrotoxicity by free radical production and subsequent oxidative stress and mitochondrial ...
  57. [57]
    Vancomycin-induced nephrotoxicity: mechanism, incidence, risk ...
    Vancomycin-induced renal toxicity was reported in 10-20 % and 30-40 % of patients following conventional and high doses of vancomycin therapy, respectively.Missing: meta- analysis
  58. [58]
    Incidence and risk factors of vancomycin-associated acute kidney ...
    A recent systematic review and meta-analysis suggested that use of vancomycin has a relative risk of 2.45 for development of AKI [7]. The exact mechanism of AKI ...
  59. [59]
    Vancomycin-associated nephrotoxicity: a critical appraisal of risk ...
    Available data suggest an association between vancomycin trough level and risk of nephrotoxicity as a function of intensity and duration of therapy (>7 days), ...
  60. [60]
    Systematic Review and Metaanalysis of Acute Kidney Injury ...
    The purpose of this systematic review and metaanalysis was to determine if concomitant vancomycin and PT is associated with a greater incidence of AKI compared ...
  61. [61]
    Vancomycin Ototoxicity: a Reevaluation in an Era of Increasing Doses
    We conclude that a significant rate of high-frequency hearing loss in older patients receiving vancomycin monotherapy was detected by audiometry.
  62. [62]
    Ototoxicity: Overview, Aminoglycosides, Other Antibiotics
    Apr 25, 2024 · Epidemiology. Incidence of ototoxicity is as high as 1% and is most commonly observed in elderly patients, even at low doses. Risk factors.
  63. [63]
    Ototoxicity and Nephrotoxicity With Elevated Serum Concentrations ...
    Previous reports suggested that possible risk factors are age >53 years, a high peak serum vancomycin concentration (>30–80 mg/L), prolonged exposure to a high ...<|separator|>
  64. [64]
    Can vancomycin cause ototoxicity? - Dr.Oracle
    Oct 2, 2025 · Risk Factors for Vancomycin-Associated Ototoxicity · Age: Patients older than 53 years appear to be at higher risk (19% incidence vs 0% in ...
  65. [65]
    Tolerability of teicoplanin in 117 hospitalized adults with previous ...
    Among patients with vancomycin-induced fever, rash, or neutropenia, there were no differences between patients with or without teicoplanin-induced fever, rash, ...
  66. [66]
    Anaphylactoid reactions to glycopeptide antibiotics - PubMed
    Antihistamine H1 blocking agents are also effective in preventing the reaction. Teicoplanin, a new glycopeptide antibiotic, does not appear to cause histamine ...<|separator|>
  67. [67]
    Cross-Reactivity Between Vancomycin and Teicoplanin
    Teicoplanin-induced agranulocytosis that followed vancomycin-induced agranulocytosis suggests a possible cross-reactivity between the 2 drugs. Both reactions ...
  68. [68]
    Incidence of teicoplanin adverse drug reactions among patients with ...
    Both vancomycin and teicoplanin are associated with relatively high rates of adverse drug reactions (ADRs), including hypersensitivity reactions. There is ...
  69. [69]
    [PDF] Therapeutic Monitoring of Vancomycin in Adult Patients - ASHP
    Vancomycin is a glycopeptide antibiotic that has been in clinical use for nearly 50 years as a penicillin alternative to treat penicillinase-producing ...
  70. [70]
    Therapeutic drug monitoring of glycopeptide antimicrobials
    Dec 22, 2023 · This review provides a comprehensive overview of LC-MS/MS methods available for TDM of glycopeptide antibiotics, including vancomycin, teicoplanin, dalbavancin ...
  71. [71]
    Clinical practice guidelines for therapeutic drug monitoring of ...
    Jan 12, 2022 · Teicoplanin, a glycopeptide antibiotic, has been extensively evaluated as a treatment for infections caused by Gram-positive bacteria including ...
  72. [72]
  73. [73]
  74. [74]
  75. [75]
  76. [76]
  77. [77]
    Vancomycin-resistant Staphylococcus aureus (VRSA) can overcome ...
    Aug 29, 2024 · Only 16 cases of vancomycin-resistant S. aureus (VRSA) have been documented in the United States. It is thought that the failure of VRSA to ...
  78. [78]
    Epidemiology and outcomes of vancomycin-resistant enterococcus ...
    Sep 18, 2023 · In this systematic review and meta-analysis, we showed a high disease burden of VRE, with infection rates ranging from 1% to 55%, depending on ...
  79. [79]
    Global prevalence and distribution of vancomycin resistant ... - Nature
    Jul 29, 2020 · The prolonged usage of vancomycin can lead to changes in cell wall patterning or reduced expression of penicillin-binding proteins. This may ...
  80. [80]
  81. [81]
    Risk Factors for the Spread of Antibiotic-Resistant Bacteria | Drugs
    Nov 27, 2012 · Risk factors for the spread of resistant bacteria in hospitals and the community can be summarised as overcrowding, lapses in hygiene or poor ...
  82. [82]
    Rationale for Revising the Clinical and Laboratory Standards ...
    The vancomycin MIC breakpoints for S. aureus were lowered (from ⩽4 μg/mL to ⩽2 μg/mL for “susceptible,” from 8–16 μg/mL to 4
  83. [83]
    Update on prevalence and mechanisms of resistance to linezolid ...
    In vitro, most VRE remain susceptible to last-resort antibiotics such as linezolid, tigecycline and daptomycin. However, neither tigecycline nor linezolid act ...
  84. [84]
    Antimicrobial resistance in the EU/EEA (EARS-Net) - ECDC
    Nov 18, 2024 · For the EU overall, there was no statistically significant trend detected between 2019 (baseline year) and 2023. The estimated total EU ...Missing: glycopeptide | Show results with:glycopeptide
  85. [85]
    Emergence of novel ST1299 vanA lineages as possible cause for ...
    Oct 31, 2023 · Emergence of novel ST1299 vanA lineages as possible cause for the striking rise of vancomycin resistance among invasive strains of Enterococcus ...
  86. [86]
    History of antimicrobial drug discovery: Major classes and health ...
    Jun 1, 2017 · The first representative of the class, vancomycin, was discovered in 1953 by Edmund Kornfeld and the team at Eli Lilly in the producer ...
  87. [87]
    Vancomycin Revisited – 60 Years Later - PMC - NIH
    Vancomycin was isolated in 1957 by Dr. E.C Kornfield, an organic chemist with Eli Lilly in the deep jungles in Borneo from a fungus named Streptomyces ...
  88. [88]
    [PDF] Teicoplanin - Art 5(3) - European Medicines Agency
    Mar 21, 2013 · Teicoplanin was first approved for marketing in Italy as Targocid, with an International Birth Date. (IBD) of 30 July 1987. Targocid ...
  89. [89]
    Teicoplanin—A New Use for an Old Drug in the COVID-19 Era? - NIH
    Nov 26, 2021 · Teicoplanin was approved in Europe in 1988, two years after its first application to treat bone and soft tissue infections, endocarditis, ...
  90. [90]
    Developments in Glycopeptide Antibiotics | ACS Infectious Diseases
    Glycopeptide antibiotics (GPAs) are a key weapon in the fight against drug resistant bacteria, with vancomycin still a mainstream therapy against serious ...
  91. [91]
    Drug Approval Package: Brand Name (Generic Name) NDA #
    Sep 5, 2014 · Oritavancin Diphosphate Lyophilized Powder for. Injection Company: The Medicines Company Application No.: 206334. Approval Date: 8/06/2014.
  92. [92]
    Total Synthesis of Vancomycin—Part 1: Design and Development of ...
    Sep 3, 1999 · A number of valuable new synthetic strategies, such as the triazene-driven biaryl ether synthesis, have been developed during the total synthesis of vancomycin.
  93. [93]
    Economic Barriers in the Treatment of Clostridium difficile Infection ...
    Despite this, the initial generic cost of vancomycin after patent expiration was high and it has remained so ever since. One potential influence on the ...<|control11|><|separator|>
  94. [94]
    Recent Advances in the Development of Semisynthetic ...
    Jul 27, 2022 · For decades, the treatment of serious Gram-positive infections relied upon the glycopeptide family of antibiotics, typified by vancomycin, as a ...Missing: aglycone | Show results with:aglycone
  95. [95]
    Experimental glycopeptide antibiotic EVG7 prevents recurrent ...
    Oct 10, 2025 · Recently, we reported the discovery and development of the new glycopeptide antibiotic EVG7 that holds promise as a treatment for challenging ...
  96. [96]
    Semisynthetic guanidino lipoglycopeptides with potent in vitro and in ...
    Aug 7, 2024 · The in vivo efficacy of guanidino lipoglycopeptide EVG7 was evaluated in a S. aureus murine thigh infection model and a 7-day sepsis survival ...<|separator|>
  97. [97]
    Saarvienin A—A Novel Glycopeptide with Potent Activity against ...
    Apr 18, 2025 · Saarvienin A exhibits potent activity against a range of Gram-positive bacteria, effectively overcoming resistance to several frontline antibiotics in clinical ...
  98. [98]
    New hope against superbugs: Promising antibiotic candidate ...
    May 14, 2025 · The novel glycopeptide saarvienin A is highly effective against resistant pathogens · Breaking the rules: A glycopeptide with a twist · A strong ...
  99. [99]
    A rare peptide scaffold in kineomicins, the glycopeptide antibiotics ...
    May 3, 2025 · Glycopeptide antibiotics (GPAs) constitute a diverse and still expanding group of naturally occurring non-ribosomally synthesized peptide ...
  100. [100]
    Trichlorination of a Teicoplanin-Type Glycopeptide Antibiotic by the ...
    Nov 26, 2018 · Chlorination of DS-A47934 had different effects on binding to the target d-Ala-d-Ala and induction of resistance (presumably by sensing by VanS ...
  101. [101]
    Peripheral modifications of [Ψ[CH2NH]Tpg4]vancomycin with added ...
    In a quest for antibiotics that may display durable clinical lifetimes, analogs of the glycopeptide antibiotics, including vancomycin, have been designed ...
  102. [102]
    Biological, chemical, and biochemical strategies for modifying ...
    The glycopeptide antibiotics (GPAs)—which include vancomycin (1) and teicoplanin (2)—are clinically utilized agents of last resort to treat resistant bacterial ...Missing: indications | Show results with:indications<|control11|><|separator|>
  103. [103]
  104. [104]
  105. [105]
    Novel Modifications of Glycopeptide Antibiotics via Total Synthesis
    Jan 26, 2018 · Total synthesis of derivatives has now led to a molecule with very significant activity against resistant strains.Abstract · Subjects · Figure 1Missing: MIC | Show results with:MIC
  106. [106]
    Peripheral modifications of [Ψ[CH2NH]Tpg4]vancomycin with ... - NIH
    A C-terminal peripheral modification, introducing a quaternary ammonium salt, is reported and was found to provide a binding pocket-modified vancomycin analog ...
  107. [107]
    Peripheral modifications of [Ψ[CH2NH]Tpg4]vancomycin with added ...
    A Nakayama, et al., Enzymatic glycosylation of vancomycin aglycon: Completion of a total synthesis of vancomycin and N- and C-terminus substituent effects ...<|control11|><|separator|>
  108. [108]
    β-Lactam Combinations with Vancomycin Show Synergistic Activity ...
    Several investigations have reported synergistic effects against Staphylococcus aureus when beta-lactams and glycopeptides including vancomycin and teicoplanin ...
  109. [109]
    Combinations of Vancomycin and β-Lactams Are Synergistic against ...
    Our data indicate that vancomycin given in combination with antistaphylococcal β-lactams demonstrates synergistic activity against a variety of staphylococcal ...
  110. [110]
    Combination Therapy With Vancomycin and Ceftaroline for ...
    In vitro studies have reported synergy between vancomycin and ceftaroline that may be greater than other β-lactam antibiotics.13, 14. Ceftaroline is an ...
  111. [111]
    Efflux Pump Mediated Antimicrobial Resistance by Staphylococci in ...
    Vancomycin, a glycopeptide antibiotic that inhibits cell wall biosynthesis, has been used as the drug of choice for the treatment of MRSA infections since the ...
  112. [112]
    Terpenes as bacterial efflux pump inhibitors: A systematic review
    Oct 12, 2022 · Terpenes are significant efflux pump inhibitors and, as such, can be used in drug development targeting the combat of antibacterial resistance.
  113. [113]
    Mining metagenomes reveals diverse antibiotic biosynthetic genes ...
    Mar 11, 2023 · This study focuses on capturing nonribosomal peptide synthase (NRPS) gene clusters implicated in the synthesis of many natural compounds of ...
  114. [114]
    Altering glycopeptide antibiotic biosynthesis through mutasynthesis ...
    Glycopeptide antibiotics (GPAs) are peptide natural products used as last resort treatments for antibiotic resistant bacterial infections. They are produced by ...
  115. [115]
    Strategy for Producing the High-Quality Glycopeptide Antibiotic ...
    Oct 10, 2021 · The CRISPR system for genome editing in A. orientalis was constructed based on the temperature-sensitive plasmid pKC1139. The CRISPR/Cas system ...
  116. [116]
    Expanding Glycopeptide Chemical Diversity | ACS Synthetic Biology
    We review these strategies here using the glycopeptides as an example and demonstrate how synthetic biology can expand antibiotic chemical diversity.
  117. [117]
    Dalbavancin for Treatment of Staphylococcus aureus Bacteremia
    Aug 30, 2025 · Here, we report the results of a randomized clinical trial assessing the efficacy and safety of dalbavancin among hospitalized participants with ...
  118. [118]
    Antibiotics in the clinical pipeline as of December 2022 - Nature
    Jun 8, 2023 · The mAb bezlotoxumab, which binds to toxin B produced by C. difficile [54, 55], was approved in 2016 to help prevent the recurrence of CDI after ...
  119. [119]
    Updates in Clinical Management of Recurrent Urinary Tract Infections
    Sep 4, 2025 · This clinical update underscores the need for individualized care plans that balance effective infection management while minimizing antibiotic- ...