+ All Categories
Home > Documents > Glycopeptide Antibiotics · 2017. 8. 28. · 914 Van Bambeke et al. so far, this paper reviews the...

Glycopeptide Antibiotics · 2017. 8. 28. · 914 Van Bambeke et al. so far, this paper reviews the...

Date post: 28-Jan-2021
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
24
Drugs 2004; 64 (9): 913-936 LEADING ARTICLE 0012-6667/04/0009-0913/$34.00/0 © 2004 Adis Data Information BV. All rights reserved. Glycopeptide Antibiotics from Conventional Molecules to New Derivatives Fran¸ coise Van Bambeke, 1 Yves Van Laethem, 2 Patrice Courvalin 3 and Paul M. Tulkens 1 1 Unit´ e de Pharmacologie Cellulaire et Mol´ eculaire, Universit´ e Catholique de Louvain, Brussels, Belgium 2 Service des Maladies Infectieuses, H ˆ opital Saint Pierre, Brussels, Belgium 3 Unit´ e des Agents Antibact´ eriens, Institut Pasteur, Paris, France Vancomycin and teicoplanin are still the only glycopeptide antibiotics avail- Abstract able for use in humans. Emergence of resistance in enterococci and staphylococci has led to restriction of their use to severe infections caused by Gram-positive bacteria for which no other alternative is acceptable (because of resistance or allergy). In parallel, considerable efforts have been made to produce semisynthet- ic glycopeptides with improved pharmacokinetic and pharmacodynamic proper- ties, and with activity towards resistant strains. Several molecules have now been obtained, helping to better delineate structure-activity relationships. Two are being currently evaluated for skin and soft tissue infections and are in phases II/ III. The first, oritavancin (LY333328), is the 4-chlorobiphenylmethyl derivative of chloroeremomycin, an analogue to vancomycin. It is characterised by: i) a spectrum covering vancomycin-resistant enterococci (VRE), methicillin-resistant Staphylococcus aureus (MRSA) and to some extent glycopeptide-intermediate S. aureus (GISA); ii) rapid bactericidal activity including against the intracellular forms of enterococci and staphylococci; and iii) a prolonged half-life, allowing for daily administration. The second molecule is dalbavancin (BI397), a derivative of the teicoplanin analogue A40926. Dalbavancin has a spectrum of activity similar to that of oritavancin against vancomycin-sensitive strains, but is not active against VRE. It can be administered once a week, based on its prolonged retention in the organism. Despite these remarkable properties, the use of these potent agents should be restricted to severe infections, as should the older glycopeptides, with an extension towards resistant or poorly sensitive bacteria, to limit the risk of potential selection of resistance. Glycopeptide antibiotics were introduced in clin- for livestock. The emergence and spreading of resis- ical practice 50 years ago, with vancomycin as the tance in enterococci and staphylococci towards van- only agent for almost 30 years. Teicoplanin was comycin has stimulated active research for new gly- launched in Europe in the mid 1980s and these two copeptides over the last 10 years, leading to the molecules remain the only members in this class production of a series of semisynthetic derivatives. available for human use; however, related deriva- Starting from vancomycin and moving through a tives have been used widely as growth promotants short description of the various compounds obtained
Transcript
  • Drugs 2004; 64 (9): 913-936LEADING ARTICLE 0012-6667/04/0009-0913/$34.00/0© 2004 Adis Data Information BV. All rights reserved.

    Glycopeptide Antibioticsfrom Conventional Molecules to New Derivatives

    Françoise Van Bambeke,1 Yves Van Laethem,2 Patrice Courvalin3 andPaul M. Tulkens1

    1 Unité de Pharmacologie Cellulaire et Moléculaire, Université Catholique de Louvain,Brussels, Belgium

    2 Service des Maladies Infectieuses, Hôpital Saint Pierre, Brussels, Belgium3 Unité des Agents Antibactériens, Institut Pasteur, Paris, France

    Vancomycin and teicoplanin are still the only glycopeptide antibiotics avail-Abstractable for use in humans. Emergence of resistance in enterococci and staphylococcihas led to restriction of their use to severe infections caused by Gram-positivebacteria for which no other alternative is acceptable (because of resistance orallergy). In parallel, considerable efforts have been made to produce semisynthet-ic glycopeptides with improved pharmacokinetic and pharmacodynamic proper-ties, and with activity towards resistant strains. Several molecules have now beenobtained, helping to better delineate structure-activity relationships. Two arebeing currently evaluated for skin and soft tissue infections and are in phases II/III. The first, oritavancin (LY333328), is the 4′-chlorobiphenylmethyl derivativeof chloroeremomycin, an analogue to vancomycin. It is characterised by: i) aspectrum covering vancomycin-resistant enterococci (VRE), methicillin-resistantStaphylococcus aureus (MRSA) and to some extent glycopeptide-intermediate S.aureus (GISA); ii) rapid bactericidal activity including against the intracellularforms of enterococci and staphylococci; and iii) a prolonged half-life, allowing fordaily administration. The second molecule is dalbavancin (BI397), a derivative ofthe teicoplanin analogue A40926. Dalbavancin has a spectrum of activity similarto that of oritavancin against vancomycin-sensitive strains, but is not activeagainst VRE. It can be administered once a week, based on its prolonged retentionin the organism. Despite these remarkable properties, the use of these potentagents should be restricted to severe infections, as should the older glycopeptides,with an extension towards resistant or poorly sensitive bacteria, to limit the risk ofpotential selection of resistance.

    Glycopeptide antibiotics were introduced in clin- for livestock. The emergence and spreading of resis-ical practice 50 years ago, with vancomycin as the tance in enterococci and staphylococci towards van-only agent for almost 30 years. Teicoplanin was comycin has stimulated active research for new gly-launched in Europe in the mid 1980s and these two copeptides over the last 10 years, leading to themolecules remain the only members in this class production of a series of semisynthetic derivatives.available for human use; however, related deriva- Starting from vancomycin and moving through atives have been used widely as growth promotants short description of the various compounds obtained

  • 914 Van Bambeke et al.

    so far, this paper reviews the basis of the selection of may allow for a cooperative binding to theoritavancin and dalbavancin as new glycopeptides ligand.[6,9-12] These dimers are formed by hydro-for clinical development (the chemical structure of gen bonds between the glycopeptide aglycone,these products is presented in figure 1). We also which are maintained in the rigid conformationaldiscuss their advantages and potential role in the state favourable for ligand binding by the sugartherapeutic armament. residues on the glycopeptide molecule.[13]

    • Anchoring of the antibiotic in the membrane, as1. Vancomycin and Teicoplanin proposed for glycopeptides carrying a lipophilic

    tail, such as teicoplanin derivatives.[11] Lipidmoieties are actually a common determinant in1.1 Mechanism of Actionthe structure of several antibiotics, such as ramo-

    Biochemical studies indicate that glycopeptides planin, moenomycin and tunicamycin, which in-inhibit the late stages of peptidoglycan synthesis.[1] hibit steps of peptidoglycan synthesis occurringThe biosynthetic pathway of this polymer involves close to the membrane. This suggests that suchthree steps: (i) the synthesis of cytosolic precursors moieties help to maintain the drug close to theirmade of pentapeptides fixed on a disaccharide; (ii) target. The importance of hydrophobic determi-the coupling of these precursors with a lipid carrier nants has been further emphasised by the obser-and the transfer of the resulting amphiphilic mole- vation that vancomycin derivatives may inhibitcule to the outer surface of the membrane; and (iii) transglycosylation without binding to D-Ala-D-the reticulation between individual precursors by Ala.[14,15]transpeptidation and transglycosylation reactions,accompanied by the release of the lipid carrier and 1.2 Mechanisms and Importanceits recycling to the inner face of the membrane. of ResistanceBacteria incubated with vancomycin accumulate

    The American National Committee for Clinicalcytosolic precursors,[1] suggesting that glycopep-Laboratory Standards (NCCLS) has set up break-tides interfere with the assembly of peptidoglycanpoints for susceptibility and resistance, respectively,and, in particular, with transglycosylation reactionsof ≤4 and ≥16 μg/mL for vancomycin and ≤8 and(figure 2). At the molecular level, the primary target≥32 μg/mL for teicoplanin (see table I for the mainof vancomycin was shown to be the D-Ala-D-Alacharacteristics of the resistant strains). Over theterminus of the precursors. Molecular modelling andyears, two organisms of medical interest, namely theexperimental studies[1-4] indicate that vancomycinenterococci and Staphylococcus aureus, have devel-forms a stoechiometric complex with the D-Ala-oped a different, ingenuous resistance mechanism.D-Ala dipeptide via the formation of five hydrogen

    bonds with the peptidic backbone of the glycopep- Resistance of enterococci to vancomycin wastide. The formation of this complex prevents the first reported in 1988.[27,28] The situation has evolvedtranspeptidation reactions by steric hindrance. The quite rapidly in the US, where resistance in bloodtightness of the interaction between the glycopep- isolates reached 13% in 1995 and 26% in 2000.[29]

    tide and the D-Ala-D-Ala motif can be enhanced by Colonisation by resistant strains is common in criti-two mechanisms. cally ill and immunosuppressed patients as well as

    • Formation of homodimers between glycopeptide in patients hospitalised in wards with high anti-molecules, which has been demonstrated for gly- microbial use. Hospitalisation is probably the maincopeptides of the vancomycin group, such as route for spread of these strains but, currently, out-vancomycin itself,[6] eremomycin[7] or chloroer- patients are also often colonised and may, therefore,emomycin.[8] Organisation of molecules in also represent another important source of contami-dimers confers a structural rigidity that locks the nation.[30] This alarming observation has stimulatedbinding pocket into the correct conformation and drastic measures in terms of hygiene, restriction in

    © 2004 Adis Data Information BV. All rights reserved. Drugs 2004; 64 (9)

  • New Glycopeptides 915

    HO OHOH

    HN

    HOOC

    NH

    O

    OHN

    O

    NH

    O

    CONH2O

    HN

    NH

    ONHCH3

    HOCI

    O

    O

    O

    CI

    OO

    HOHO

    OHO

    H2N

    HOH3C

    CH3

    HO OOH

    HN

    HOOC

    ONH

    OHN

    O

    NH

    OHN

    O

    NH

    O

    HO

    OCI

    O O

    O CI

    NH2

    OH

    NHAc

    OHO

    HOOH

    O

    HOHO

    HO

    HN CO

    OHOH

    O

    OH

    OH

    O

    HO O

    HN

    HOOC

    ONH

    OHN

    O

    NH

    OHN

    O

    NH

    O

    NHCH3

    CI

    HO

    OH

    HOCI

    O O

    O

    OH

    O

    OH

    OH

    OH

    OH

    OHOOC

    HOHO

    HNCO

    O

    HO OHOH

    HOOC

    HN

    OHN

    O

    NH

    OCI

    O

    NH

    OHN

    CONH2O

    NH

    ONHCH3

    O

    O

    O

    CI

    OH3CCH3

    H2NHO

    OO OH

    HOHO

    OCH3H2N

    OHH3C

    HO OHOH

    HN

    HOOC

    NH

    OHN

    O

    NH

    OHN

    OCONH2

    NH

    ONHCH3

    O

    OCI

    O O

    ClO

    OHOH3C

    CH3

    H2N

    OO

    OH

    HOHO

    OHN

    H3C

    CI

    OH

    CH3

    HO OOH

    HN

    CO

    NH

    O

    OHN

    O

    NH

    HN

    NH

    NH

    N

    HOCI

    O O

    O O

    OH

    CI

    HO

    NHCH3

    O

    O

    O

    OHOH

    OHOH

    OHOOC

    HOHO

    HNCO

    O

    Vancomycin

    Teicoplanin

    LY264826 A40926

    Oritavancin Dalbavancin

    Fig. 1. Chemical structure of glycopeptides. Upper panel: vancomycin and teicoplanin, which are the two glycopeptides currently availablein clinical settings; lower panel: oritavancin and dalbavancin, two semisynthetic glycopeptides currently under clinical development. Middlepanel: natural derivatives of vancomycin and teicoplanin used for the synthesis of oritavancin and dalbavancin, respectively. The part of themolecule that is common to all glycopeptides appears in bold. The arrows point to the differences between molecules.

    © 2004 Adis Data Information BV. All rights reserved. Drugs 2004; 64 (9)

  • 916 Van Bambeke et al.

    Susceptible strain

    ddl

    UDP

    Transpeptidase

    Transglycosylase

    UDP UMP UDP UDP

    VanSVanS

    VanR -PVanR

    -P

    ddl VanX VanT

    VanHpyr

    VanA-E

    UDP UMP UDP UDP

    L-ser

    UDPvan operon

    VanY

    Transglycoslase

    Transpeptidase

    Resistant Enterococcus (VRE)

    Intermediate resistant Staphylococcus aureus (VISA)

    Transpeptidase

    Transglycoslase

    Glycopeptide

    N-acetylmuramic acid

    N-acetylglucosamine

    Tripeptide

    Interpeptide bridge

    Lipid carrier

    D-Ala

    D-Lac

    D-Ser

    Depsipeptide

    Fig. 2. Peptidoglycan synthesis in glycopeptide-susceptible bacteria (upper panel), in glycopeptide-resistant enterococci (middle panel) andin glycopeptide-intermediate staphylococci (lower panel).[2,5] In susceptible strains, peptidoglycan precursors are ending by D-Ala-D-Alatermini synthesised by D-Ala:D-Ala ligases (ddl). Glycopeptides interact with D-Ala-D-Ala termini of pentapeptides (through five hydrogenbonds) at the cell surface, preventing transglycosylation reactions and therefore peptidoglycan reticulation. The figure also illustrates howdimerisation of glycopeptide molecules or anchoring in the membrane by a hydrophobic tail may increase the tightness of this interaction.Hydrophobic derivatives may also inhibit the transglycosylation step without binding to peptidoglycan precursors. In glycopeptide-resistantenterococci, the presence of glycopeptides activates the signal-transducing system VanS (sensor) – VanR (regulator), which allowstranscription of the resistance genes. These include enzymes involved in the hydrolysis of D-Ala-D-Ala termini (VanX and VanY) or in thesynthesis of precursors ending by either D-Ala-D-Lac (VanH and VanA, VanB or Van D) or D-Ala-D-Ser (VanT and VanC, VanE or VanG),characterised by a lower affinity for glycopeptides. In glycopeptide-intermediate staphylococci (so-called VISA [vancomycin-intermediateStaphylococcus aureus] or GISA [glycopeptide-intermediate S. aureus] strains), more murein monomers are supplied and incorporated inthe peptidoglycan, in which more free D-Ala-D-Ala residues remain present because of a decreased level of reticulation. More glycopeptidemolecules are therefore trapped in the multiple layers of the thicker cell wall but fewer reach their target at the cell membrane. UDP = uridinediphosphate; UMP = uridine monophosphate; VRE = vancomycin-resistant enterococci.

    © 2004 Adis Data Information BV. All rights reserved. Drugs 2004; 64 (9)

  • New Glycopeptides 917

    Table I. Main characteristics of glycopeptide-resistant bacteria

    Resistance Bacterial species Expression Mechanism of resistance MIC range of Referencestype glycopeptides (mg/L)

    vancomycin teicoplanin

    VanA Enterococcus faecium Inducible (by vancomycin Modified target 64–1000 16–512 16

    E. faecalis and teicoplanin) (D-Ala-D-Lac)

    E. avium

    E. durans

    E. hirae

    E. mundii

    E. raffinosus

    E. gallinarum

    E. casseliflavus

    Staphylococcus aureus Inducible (by vancomycin Modified target 17-20and teicoplanin) (D-Ala-D-Lac)

    VanB E. faecium Inducible (by vancomycin) Modified target 4–1000 0.5–1 16

    E. faecalis (D-Ala-D-Lac)

    Streptococcus bovis 21

    VanC E. gallinarum Constitutive/inducible Modified target 2–32 0.5–1 16

    E. casseliflavus (D-Ala-D-Ser)

    E. flavescens

    VanD E. faecium Constitutive Modified target 64–128 4–64 16,22(D-Ala-D-Lac)

    VanE E. faecalis Inducible (by vancomycin) Modified target 16 0.5 16,23(D-Ala-D-Ser)

    VanG E. faecalis Inducible (by vancomycin) Modified target 16 0.5 24(D-Ala-D-Ser)

    VISA S. aureus Thickened cell wall with 8 8–32 5,25increased proportion ofD-Ala-D-Ala termini

    GRSA S. aureus Stop codon in the gene of 26PBP4

    GRSA = glycopeptide-resistant Staphylococcus aureus; MIC = minimum inhibitory concentration; VISA = vancomycin-intermediateStaphylococcus aureus.

    antimicrobial use and surveillance to try to control The mechanism of resistance in enterococci re-or even reverse the situation.[29] In sharp contrast, lies on synthesis of peptidoglycan by an alternativeresistance of enterococci in Europe is minimal (3%) pathway, which produces precursors ending inand observed only in nosocomial infections;[31] out- D-Ala-D-Lac or D-Ala-D-Ser instead of D-Ala-D-breaks have been reported but remain sporadic. Ala and concomitantly eliminates precursors endingCarriage of resistant enterococci in animals, how- in D-Ala-D-Ala. The replacement of D-Ala by D-ever, is frequent in Europe and is thought to be due Lac suppresses one of the five hydrogen bondsto the massive use of avoparcin, a glycopeptide between the glycopeptides and their target, whichantibiotic used as growth promoter in animal leads to a 1000-fold decrease in the binding affini-feed,[32,33] but which has the same mode of action as ty.[2,37] The substitution in D-Ser causes a conforma-vancomycin and, therefore, shares cross-resistance. tional change, which also reduces vancomycin affin-The ban on avoparcin use instituted by EU countries ity, although not as markedly as with D-Lac. To behas caused, in most countries, a fall in the preva- phenotypically detectable and significant, resistancelence of resistance in both animals and requires the coordinated action of several enzymeshumans.[29,34-36] (see figure 2). Thus, the bacteria need to synthesise

    © 2004 Adis Data Information BV. All rights reserved. Drugs 2004; 64 (9)

  • 918 Van Bambeke et al.

    Table II. In vitro activity of oritavancin and dalbavancin compared with those of vancomycin and teicoplanina

    Organism MIC range (mg/L) References

    vancomycin oritavancin teicoplanin dalbavancin

    Staphylococcus aureus

    methicillin-susceptible 0.13–1 0.13–1 0.25–8 ≤0.03–0.5 490.25–2 0.06–4 0.125–4 0.06–0.5 50-55

    methicillin-resistant 0.5–4 0.13–4 0.13–8 0.06–1 49

    0.25–4 0.03–8 0.06–8 0.06–1 50-55

    vancomycin-intermediate 8 1–8 8–32 2 56-58

    Staphylococcus epidermidis

    methicillin-susceptible 0.13–1 0.25–1 0.25–16 ≤0.03–0.25 490.25–2 ≤0.03–8 ≤0.03–16 50,54

    methicillin-resistant 1–4 0.25–4 1–16 ≤0.03–1 490.5–4 0.25–16 0.125–16 50,54

    Staphylococcus haemolyticus

    methicillin-susceptible 1–4 0.06–1 1–32 ≤0.03–0.25 490.25–4 0.12–8 ≤0.12–8 50,54

    methicillin-resistant 0.5–8 0.13–1 2–128 ≤0.03–4 491–4 1–4 0.5–>16 50,54

    Other coagulase-negative staphylococci

    methicillin-susceptible 0.5–2 0.06–0.5 0.13–4 ≤0.03–0.13 490.25–4 ≤0.01–2 ≤0.12–32 ≤0.03–0.25 51-53

    methicillin-resistant 0.5–4 ≤0.03–0.5 0.06–32 ≤0.03–0.13 490.25–8 0.015–4 0.06–64 0.06–1 51-53

    Streptococcus pyogenes 0.5–0.5 0.016–0.13 0.008–0.06 ≤0.002–0.06 490.25–0.5 0.06–0.5 ≤0.03–0.12 55

    Streptococcus pneumoniae

    penicillin-susceptible 0.13–0.5 ≤0.002–0.06 0.008–0.06 0.016–0.13 490.06–0.5 ≤0.002–0.12 ≤0.01–0.25 51,52,54,55

    penicillin-resistant 0.25–2 ≤0.002–0.06 0.016–0.13 0.008–0.13 490.06–1 0.002–0.25 ≤0.01–0.25 51,52,54,55

    Viridans streptococci

    0.25–2 ≤0.12–2 ≤0.03–0.06 53penicillin-susceptible 0.12–1 ≤0.01 ≤0.01–0.5 52penicillin-resistant 0.25–1 0.01–0.06 ≤0.01–0.5 52β-Haemolytic streptococci 0.25–1 ≤0.12–0.25 ≤0.03–0.12 53

    Enterococcus spp.

    vancomycin-susceptible 0.25–4 0.06–0.25 0.13–0.5 0.06–0.13 49

    ≤0.01–4 ≤0.03–2 ≤0.012–8 ≤0.03–1 50-54,59VanA >128 0.06–1 64–>128 0.5–>128 49

    64–>256 0.06–1 32–>256 50

    VanB 8–128 ≤0.03–0.13 0.13–8 0.02–2 498–128 0.25–1 0.125–0.5 60

    VanC 4–16 ≤0.03–1 0.125–4 50Bacillus spp. ≤0.12–1 ≤0.015–0.5 ≤0.12–4 ≤0.03–2 53,54Corynebacterium spp. 0.25–0.5 ≤0.12–1 ≤0.03–0.12 53

    Continued next page

    © 2004 Adis Data Information BV. All rights reserved. Drugs 2004; 64 (9)

  • New Glycopeptides 919

    Table II. Contd

    Organism MIC range (mg/L) References

    vancomycin oritavancin teicoplanin dalbavancin

    Listeria spp. 0.25–2 ≤0.03–0.125 0.06–0.25 50Clostridium difficile 0.5–4 0.016–2 0.064–0.5 0.125–0.5 61,62

    Clostridium perfringens 0.025–4 0.016–2 0.064–4 0.03–0.125 61,62

    Haemophilus influenzae ≥16 16–32 8–64 53,54a When two rows of data appear for a single species, the first one refers to studies where the drugs are compared against the same

    strains; the second one represents the range of MICs reported in other publications.

    MIC = minimum inhibitory concentration.

    D-Lac (VanH) or D-Ser (VanT) and D-Ala-D-Lac an abundant extracellular material of still ill-charac-or D-Ala-D-Ser (VanA, B, D or C, E, G), and to terised nature and an impaired ability to divide.[43]

    degrade D-Ala-D-Ala (VanX) or to remove D-Ala These phenotypic changes can be explained by thefrom growing precursors (VanY) or both (VanXY). production of an altered peptidoglycan with an in-Moreover, a two-component regulatory system creased proportion of free D-Ala-D-Ala termini(VanS-VanR) resulting in induction by either van- (less reticulation), which can trap vancomycin mole-comycin (VanB, C, E, G phenotype) or vancomycin cules and prevent their access to the target at theand teicoplanin (VanA phenotype) plays a critical cytosolic membrane (figure 2). The thickened cellrole.[2,16,22,23] The resistance proteins are encoded by wall of a VISA strain may contain up to two to fourgenes physically grouped in operons that are located times more D-Ala-D-Ala residues than that of aon plasmids or in the chromosome, and can be easily susceptible strain and is able to bind up to three totransferred, even between different species.[2,38,39] six times more vancomycin molecules before pep-Six phenotypes of resistance have been described tidoglycan synthesis becomes impaired.[5] This(see table I), which differ by the genetic support, the mechanism also implies a reduced cross-linkage ofregulation of expression and the level of resistance peptidoglycan. The latter has been suggested to re-conferred. An intriguing phenotype of glycopeptide sult from decreased activity of penicillin-bindingdependence has also been described, in which syn- proteins (PBPs)[26] or from an alteration of mureinthesis of the D-Ala-D-Ala-ending peptidoglycan precursors.[44] Yet multiple, additive, but not clearlyprecursors is impaired through a mutation in the host identified mutations, are probably necessary to ob-ligase. Induction of production of the resistance tain resistance. VISA and GISA also need to importproteins by glycopeptides restores peptidoglycan a larger amount of precursors than normal strains,synthesis by allowing production of precursors end- which compromises their fitness in an antibiotic-ing in D-Ala-D-Lac.[40,41] free environment. This explains why VISA and

    GISA tend to lose their resistance when relievedResistance in S. aureus first emerged in the formfrom vancomycin pressure, giving rise to the so-of strains with elevated minimum inhibitory concen-called hetero-VISA phenotype.[45] Moreover, atration (MIC) values towards vancomycin (or bothglycopeptide-tolerant phenotype has been observedvancomycin and teicoplanin), which have beenin clinical isolates of MRSA, in which MICs ofnamed vancomycin-intermediate S. aureus (VISA)glycopeptides are not affected but minimum bacteri-or glycopeptide-intermediate S. aureus (GISA).cidal concentrations (MBCs) are considerably in-Originally described in Japan in 1996 in methicillin-creased.[46] In a still more frightening fashion, tworesistant S. aureus (MRSA),[25] VISA and GISAMRSA strains with high levels of resistance to van-strains have now been isolated in numerous coun-comycin and teicoplanin have now been reported intries, particularly from patients having received pro-two different hospital institutions in the US.[17-20]longed vancomycin therapy.[5,42] These bacteria are

    characterised by a thickened cell wall, production of Both of these strains harbour the vanA gene cluster,

    © 2004 Adis Data Information BV. All rights reserved. Drugs 2004; 64 (9)

  • 920 Van Bambeke et al.

    indicating that they had acquired the corresponding some extent, bone and cartilage, but not in theset of genes from enterococci. Should such strains CNS.[63]

    spread in hospitals, treatment options would rapidly In vitro pharmacodynamic models show that van-become very limited.[47] comycin exhibits time-dependent bactericidal ac-

    tivity against most Gram-positive organisms. How-ever, vancomycin is essentially bacteriostatic

    1.3 Pharmacological Propertiesagainst enterococci[71] but may become bactericidalwhen combined with an aminoglycoside.[72,73] Van-

    The spectrum of activity of glycopeptides covers comycin activity is adversely affected by a largeessentially the Gram-positive organisms and a few inoculum but not by acidic pH. The drug also dem-anaerobes, and their activity against Gram-negative onstrates faster killing rates against actively grow-organisms is most often marginal (table II). Vanco- ing organisms, which can be easily understood onmycin and teicoplanin have a similar intrinsic ac- the basis of its mechanism of action.[74] Finally,tivity, except against streptococci, which are more glycopeptides produce persistent effects (postantibi-susceptible to teicoplanin.[48] otic effect lasting 1–6 hours[71,75]). While the dosage

    The key pharmacokinetic properties of glycopep- recommendations for glycopeptides have insisted ontides are summarised in table III. The most striking low dosages because of the fear of toxicity, recentdifference between vancomycin and teicoplanin is developments in our understanding of pharmaco-their capacity to bind serum proteins, which is much dynamics have allowed a more rational basis forhigher for teicoplanin than for vancomycin. This recommendations. The first models suggested thatexplains the prolonged half-life of teicoplanin in the the free serum concentration of glycopeptides needsorganism. However, high protein binding also needs to remain above the MIC for the infecting organismto be taken into account for activity predictions for a prolonged period and, therefore, concluded that(table IV), since only the free serum fraction is activity was primarily driven by the so-called ‘timedirectly active. Teicoplanin, perhaps because of its above MIC’ parameter.[76,77] However, more recentlipophilic character, is characterised by a higher and thorough studies have indicated that the para-volume of distribution than vancomycin, allowing it meter which best predicts efficacy is the area underto reach therapeutic concentrations in fat, muscles the plasma concentration-time curve (AUC)/MIC(including pericardium and myocardium) and, to ratio,[56] as usually found for most antimicrobials

    Table III. Pharmacokinetic properties of glycopeptides as observed after administration of conventional clinical dosesa

    Parameter Vancomycin Oritavancin Teicoplanin Dalbavancin

    Peak concentration (mg/L) 20–50 31b 43 300

    Trough concentration (mg/L) 5–12 (24h) 1.7 (24h)b

  • New Glycopeptides 921

    Table IV. Pharmacodynamic parameters predictive of glycopeptide efficiency calculated for conventional dosages (see table III)

    PK/PD parameter Maximum reachable MIC based on PK/PD parameters

    vancomycin 15 mg/kg bid teicoplanin 6 mg/kg od oritavancin 3 mg/kg od dalbavancin 15 mg/kg od

    AUC/MIC = 125a

    total 4 4 1 185

    free 2 0.4 0.1 4

    Cmax/MIC = 10a

    total 10 4 3 30

    free 5 0.4 0.3 0.6

    a Target values based on general PD considerations for antibiotics in the absence of actual data for all glycopeptides (lower Cmax/MIC ratio may be sufficient on the basis of available data for vancomycin and teicoplanin).

    AUC = area under the concentration-time curve; bid = twice daily; Cmax = peak concentration; MIC = minimum inhibitory concentration; od= once daily; PD = pharmacodynamic; PK = pharmacokinetic.

    exhibiting time-dependent killing and significant 1.4 Clinical Use: Pros and Conspostantibiotic effect.[78] However, in non-neutropen-

    Originally introduced into clinical practice asic animals, the ratio of the free serum peak concen-an agent active against β-lactamase-producing S.tration to the MIC (Cmax-free/MIC) also plays aaureus, vancomycin remained largely unused be-determinant role in efficacy.[79] This Cmax-free/MICcause of development of less toxic alternatives,

    ratio needs to reach at least 5–6 for vancomycin and namely the β-lactamase-resistant penicillins (methi-2–3 for teicoplanin (note that the determination of cillin, isoxazolyl penicillins) and cephalosporins, asthe peak and trough levels for these drugs, which are well as the introduction of β-lactamase inhibitors

    such as clavulanic acid (mainly in Europe) or sul-determined in routine in hospital laboratories, can bebactam, which have become highly popular whenused for calculation of both Cmax/MIC and AUC/combined with ampicillin or amoxicillin (mostlyMIC ratios). As a consequence, glycopeptides areagainst Gram-negative bacteria, however). Yet oral-

    probably best given by discrete administrations withly administered vancomycin became popular for

    a total daily dose sufficiently large to match with the treatment of Clostridium difficile-associated diar-MIC of the infecting organism. A recent study com- rhoea and colitis.[86] The pandemic of nosocomialparing vancomycin 30 mg/kg once daily versus two MRSA infections which started in the mid 1970s,

    and the fact that these strains were resistant not onlyadministrations for the treatment of patients withto all β-lactam agents but often also to aminoglyco-bacteraemia or arthritis did not show a significantsides, macrolides, lincosamides and fluoroquino-difference between the two groups in terms of clin-lones,[87] heralded the return of vancomycin for sys-

    ical response (>92% in both cases;[80] however, notemic use. Alternatives such as fusidic acid, rifampi-

    patients with endocarditis were included in this cin and cotrimoxazole have indeed been poorlystudy). On a pharmacodynamic basis, there is there- studied and did not compete in the face of thefore little reason to administer vancomycin by con- extensive data and the everyday experience avail-

    able with glycopeptides.[88] Moreover, easy selec-tinuous infusion[81,82] even though other parameterstion of resistant mutants (especially for rifampicinsuch as more sustained concentrations in given tis-and fusidic acid when used alone) or toxicity prob-sues, or cost containment and ease of administration,lems has further limited their use. As we have seen,

    have been advocated.[83,84] (Note that the stability however, resistance developed in enterococci almostand compatibility of vancomycin with other drugs in in parallel with the increase in vancomycin use inrelation to its use by continuous infusion are current- the US.[89-91] This led health authorities and con-ly under investigation.[85]) cerned clinicians to try to restrict glycopeptide usage

    © 2004 Adis Data Information BV. All rights reserved. Drugs 2004; 64 (9)

  • 922 Van Bambeke et al.

    in order to maintain the activity of these usefulagents for situations where alternatives were notusable.[90,92-95] The introduction of quinupristin/dalfopristin and linezolid in the late 1990s has notreally changed this situation, since both drugs areexpensive, have rare but potentially worrying ad-verse effects (haematological toxicity for long-course linezolid treatments, thrombophlebitis, ar-thralgias and/or myalgias, and drug interactions forquinupristin/dalfopristin), and are already facingemergence of resistance.[96-101] In particular, linezo-lid-resistant strains are now increasingly reported,not only in enterococci but also in S. aureus, whichcould become a clinically significant problem in thefuture.

    The recommendations presented in table V wereissued in 1995 by the Hospital Infection ControlPractices Advisory Committee in the US(HICPAC[102]) and have been adopted, with adapta-tions, in most countries (see Gordts et al.[103] forBelgium). These recommendations have been fur-ther extended to special populations (see Nourse etal.[104] for children). In summary, and outside thefield of bacteriologically documented MRSA infec-tions, empirical therapy with glycopeptides shouldbe limited to severe infections in immuno-compromised patients (e.g. burn patients, clinicalsepsis in the intensive care unit) when local epide-miological data show a high percentage of MRSA,and/or to life-threatening infections associated withthe presence of foreign bodies (often infected withmethicillin-resistant coagulase-negative staphylo-cocci; e.g. indwelling percutaneous catheters, pros-thetic cardiac valves). Indeed, animal models and

    Table V. Appropriate clinical use of glycopeptides, based on therecommendations from Hospital Infection Control Practice AdvisoryCommittee (HICPAC)[102] for vancomycin and on pharmacodynamicconsiderations (table IV and Gruneberg et al.[105])

    Situations in which the use of glycopeptides is appropriateor acceptable

    Treatment

    Serious infections caused by β-lactam-resistant Gram-positivemicroorganisms

    Infections caused by Gram-positive microorganisms in patientswho have serious allergies to β-lactam agentsAntibiotic-associated colitis failing to respond to metronidazoletherapy or potentially life-threatening

    Prophylaxis

    Endocarditis following certain procedures in patients at high riskfor endocarditis

    Major surgical procedures involving implantation of prostheticmaterials or devices at institutions that have a high rate ofinfections caused by methicillin-resistant Staphylococcus aureus(MRSA) or methicillin-resistant S. epidermidis

    Situations in which the use of glycopeptides should bediscouraged

    Routine surgical prophylaxis other than in a patient who has alife-threatening allergy to β-lactam agentsEmpirical antimicrobial therapy for a febrile neutropenic patient,unless initial evidence indicates that the patient has an infectioncaused by Gram-positive microorganisms and the prevalence ofinfections caused by MRSA in the hospital is substantial

    Treatment in response to a single blood culture positive forcoagulase-negative staphylococci, if other blood cultures takenduring the same time frame are negative

    Continued empirical use for presumed infections in patientswhose cultures are negative for β-lactam-resistant Gram-positivemicroorganisms

    Appropriate dosagesa,b

    Vancomycin 15 mg/kg twice daily

    Teicoplanin 6–12 mg/kg once daily (after three loading doses of6 mg/kg every 12 hours)

    a Higher intravenous doses are needed for treating peritonitis inpatients undergoing peritoneal dialysis.[106,107]

    b Dosage reduction required in patients with renalinsufficiency.[63,64]

    clinical studies have shown that vancomycin is lessbactericidal than isoxazolyl penicillins against with the exception of true β-lactam allergy. Of thesemethicillin-sensitive staphylococci (causing pro- severe infections by multiresistant staphylococci,longed periods of fever and persistence of positive the main indications for vancomycin are, therefore,blood cultures). This should preclude its use (and, restricted to: (i) serious diphtheroid infectionsdespite fewer data, the use of teicoplanin) when such (when the strain is penicillin resistant or the patientstrains are involved, especially in life-threatening has IgE-mediated allergy to β-lactam agents); (ii)infections (e.g. endocarditis). Therefore, their indis- penicillin-resistant Streptococcus pneumoniae in-criminate use in non-documented infections (or in- fections of the CNS in combination with cefotaximefections caused by Gram-positive strains susceptible or ceftriaxone; and (iii) antibiotic-associated colitisto other common agents) should be discouraged that is life-threatening or fails to respond to metroni-

    © 2004 Adis Data Information BV. All rights reserved. Drugs 2004; 64 (9)

  • New Glycopeptides 923

    dazole (only oral vancomycin; low faecal concentra- lycosides or other drugs toxic for these organs.[112]

    Infrequent toxic manifestations include neutropenia,tions by intravenous route and few data with teico-thrombocytopenia, fever, bullous dermatosis, necro-planin). Routine prophylaxis with glycopeptidestising cutaneous vasculitis and toxic epidermalshould also be prohibited[102,103] and restricted tonecrolysis.[113] Teicoplanin has been claimed to beprevention of bacterial endocarditis in penicillin/less toxic than vancomycin.[114] However, a difficul-ampicillin-allergic patients at risk, undergoing gas-ty relates to the fact that the recommended doses oftrointestinal or genitourinary procedures (plusteicoplanin have been increasing over time and thatgentamicin), or dental procedures (vancomycinmost comparative studies with vancomycin werealone). In selected surgical wards with a high inci-done with teicoplanin doses (typically ≤6 mg/kg)dence of methicillin-resistant staphylococci infec-that are now considered insufficient (table V).tions, a single dose before surgery may be used inTherefore, further studies are needed to evaluateprosthetic surgery, but one should always try totoxicity of teicoplanin in the current conditions ofsolve the underlying fundamental hygiene problem.use.Teicoplanin could be used in most of the same

    indications, with the exception of the CNS infection,2. New Developments in thebecause of the poor penetration of the drug in theGlycopeptide Areainfected compartment. Its main advantages are the

    possibility of administering it by the intramuscularGiven the potential of glycopeptides in severe

    route with a once-daily schedule (because of itsinfections, considerable effort has been made to

    longer half-life) and a lower incidence of adverseobtain new derivatives with improved pharmacolog-

    effects. However, several indications are less docu-ical properties and activity against resistant strains.

    mented than for vancomycin, and there are no wellBecause of a highly complex structure, which makes

    conducted, large, comparative studies between the total synthesis of glycopeptides very challeng-two drugs. In severe infections (as in S. aureus ing,[115] most of the new molecules obtained so farendocarditis), teicoplanin needs to be given at a dose are semisynthetic derivatives of existing, naturalof at least 12 mg/kg/day once daily, after a mini- glycopeptides. Structure-activity relationshipsmum of three to four loading doses (each given (SARs) are presented in figure 3, based on ourevery 12 hours). current understanding of the molecular mode of

    interaction between glycopeptides and their pharma-Safety issues need also to be taken into accountcological target. Modifications can be subdividedand may limit glycopeptide use in given popula-into three main categories, each of them concerningtions. Earlier, vancomycin was notorious for toxici-distinct domains of the molecule and affecting thety related to impurities and to histamine releasegeneral mode of action of vancomycin in various(causing the so-called ‘red man syndrome’). Theseways. Other recent strategies have explored the for-adverse effects have been markedly reduced throughmation of hybrid or dimerised molecules, or thebetter purification procedures and by giving the drugsynthesis of inhibitors of the resistance mechanismsas a slow infusion over at least 1 hour.[108-110] Withto be administered in combination with unmodifiedthese precautions, vancomycin can be safely used,glycopeptides.causing only relatively mild and self-limiting gener-

    al toxicity.[111,112] However, the dose should be2.1 Semisynthetic Modification ofcorrected in the case of renal insufficiency. TheNatural Glycopeptidesmain adverse effects are phlebitis at the site of

    injection as well as nephrotoxicity and ototoxicity,2.1.1 Modification of the Binding Pocketthe latter remaining the most problematic because it

    can be irreversible.[64] Both nephro- and ototoxicity Binding studies have shown that five sites on theare aggravated by the concomitant use of aminog- peptidic backbone of glycopeptides are involved in

    © 2004 Adis Data Information BV. All rights reserved. Drugs 2004; 64 (9)

  • 924V

    an Bam

    beke et al.

    © 2004 A

    dis D

    ata

    Info

    rma

    tion

    BV. A

    ll righ

    ts rese

    rved

    .D

    rug

    s 2004; 64 (9)

    HO OHOH

    HN

    HOOC

    NH

    HN

    O

    ONH

    O

    CONH2O

    HN

    HN

    ONHCH3

    OCI

    O O

    CIO

    HO

    O OH

    HOHO

    OO

    H3C

    HO

    CH3H2N

    Acylation:↑ activity

    Alkylation:↑↑ activity

    Amino sugar:↑ dimer formation

    Carboxamide: ↑ activity

    (Staphylococcusepidermidis)

    CI:↑ dimer formation

    Vancomycin

    HO OHOH

    Membrane anchoringInhibition of transglycosylase

    Dimerisation

    HN

    NH

    HOOC

    O

    O

    O

    Cl

    O

    HN

    O

    NH

    OHN

    NH

    OCONH2 O

    NHCH 3

    HO

    O

    O OHHO

    HO

    O

    HOH3C

    CH3H2N

    CI

    Binding to D-Ala-D-Ala

    6 5 4

    3

    2

    1

    HO OOH

    HN

    NH

    O

    HOOC

    HN

    O

    NH

    OHN

    O

    NH

    O

    NH2

    HO

    OH

    OCI

    O O

    O CI

    O

    NHAc

    OHO

    HO

    OH

    OHO

    HO

    HN CO

    HO

    O

    OHOH

    OH

    OH

    O

    Teicoplanin

    Sugar: ↑ activity

    staphylococci↓ activity VanA

    COOH: ↑ activity Gram-negative

    ↓ activity VanA

    Sugar:↑ activity

    Alkyl:↑ half-life↑ activity↓ VanB induction

    CI:↑ activity

    Basic amide:↑ activity

    ↑ activity Gram-negative

    Acylation:↑ activitystaphylococci

    Fig. 3. Structure-activity relationship (SAR) of glycopeptides derived from vancomycin and teicoplanin. Upper panel: current view of the parts of the molecules involved in thevarious modes of action proposed for glycopeptides. Lower panel: most salient SARs that can be drawn for vancomycin (left) or teicoplanin (right) derivatives. ↑ indicates increased;↓ indicates decreased.

  • New Glycopeptides 925

    the interaction with the D-Ala-D-Ala termini (figure to cross their outer membrane.[118] Esterification3). Efforts have been made to change these amino with hydrophobic alcohols also leads to marginalacid residues, with the aim to generate compounds activity on Gram-negative bacteria. Similarly, basicwith increased affinity for both dipeptide and dep- amides of the A40,926 derivative of teicoplaninsipeptide termini. These have included trimming or (which lacks the N-acetylglucosamine sugar; figureenlarging the heptapeptide backbone, epimerisation 1) show increased activity against staphylococciof the C3 chiral centre (in teicoplanin only), or (particularly coagulase-negative) and streptococci.substitution of amino acids 1 and 3.[116] Unfortunate- The most active compounds also show moderately, most of these modifications have resulted in a activity against VanA enterococci.[119,120] At the N-reduction of antibacterial activity. Nevertheless, the terminus, acylation or alkylation causes either apossibility of obtaining active compounds by this slight increase or no modification in activity. An-type of approach remains open, based on the obser- other important modification in the teicoplaninvation that substitution of the Asn at position 3 by a backbone is the introduction of chlorine atoms,hydrophobic amino acid enhances the affinity of which increase both target affinity and antibacterialvancomycin for D-Ala-D-Lac precursors.[117] activity.[116]

    2.1.3 Alteration of Sugars2.1.2 Modification or Addition ofFunctional Groups Despite their location apparently far from the

    binding pocket, sugars play a determinant role inA few functional groups can be modified out ofglycopeptide activity, since aglycones of both van-the binding pocket, essentially causing changes incomycin and teicoplanin are systematically less ac-physicochemical properties. These studies have gen-tive than their parent compounds.[116,121] Substitut-erated useful knowledge in SAR and have alloweding these sugars can actually have a considerablechanges in the natural compounds to be proposed.impact on glycopeptide pharmacological properties.

    Modification of VancomycinModification of VancomycinAs stated earlier, the affinity of vancomycin forBesides the beneficial addition of a 4-epi-its target is increased by its capacity to dimerise.

    vancosamine on the benzylic function of residueThis process is facilitated by the addition of an6,[122] substitution of the vancosamine sugar by hy-amino sugar to residue 6 and by the presence of adrophobic substituents proved mostly useful, prob-chlorine atom in meta on the aromatic substituent ofably by somehow mimicking the lipophilic sideresidue 2.[10,13,116] Other modifications at the extre-chain of teicoplanin derivatives. N-alkyl derivativesmities of the peptidic backbone have also broughtwere found to be more active than N-acyl products,significant pieces of information. Thus, replacementparticularly against enterococci, streptococci andof the free carboxylate terminus by a carboxamidestaphylococci.[116] When combined with the addi-increases the activity on Staphylococcus epider-tional 4-epi-vancosamine (as in LY264826; see fig-midis, whereas D-amino acids at the N-terminusure 1), an alkyl side chain confers to the molecule anappear more active than their corresponding L-iso-unexpectedly rapid, concentration-dependent bacte-mers.[116]ricidal activity, including against VanA- and VanB-

    Modification of Teicoplanin type resistant enterococci.[123-125] The activity ofAs for vancomycin, structure-activity studies of these hydrophobic derivatives on resistant strains

    teicoplanin have examined the influence of substi- has been proposed to result from the combination oftuting the extremities of the peptidic backbone. At a facilitation of the binding to the peptidoglycanthe C-terminus, substitution by a basic, positively precursors due to their anchoring in the mem-charged amide considerably increases activity on brane[9,11] and of a direct inhibition of transglycosy-staphylococci and also confers moderate activity on lases.[15,126] Recent studies also suggest that some ofGram-negative bacteria, because of improved ability these derivatives may act as inhibitors not only of

    © 2004 Adis Data Information BV. All rights reserved. Drugs 2004; 64 (9)

  • 926 Van Bambeke et al.

    cell wall synthesis but also of phospholipid synthe- linkers and various linkage positions.[130,131] Im-sis.[127] Acidic and hydrophilic substituents have proved activity has been observed against Gram-also been added on the resorcinol in alpha of the free positive organisms, including GISA and VREcarboxylate of lipidated analogues of vancomycin, strains, opening promising lines of investigation inwith favourable consequences on distribution and this area. Secondly, taking into account the possibil-safety profiles, without negative effect on activity ity of a dual target for glycopeptides, namely theagainst MRSA or vancomycin-resistant enterococci dipeptide termini of peptidoglycan precursors, and(VRE).[128] the enzymes involved in the transglycosylation step,

    vancomycin derivatives have been prepared whereModification of Teicoplanin

    the aglycone is separated from the sugars by linkers,The impact of sugars on the activity is em-

    with the aim to correctly position each part of thephasised by the fact that the removal of N-acyl-

    molecule.[132] This approach could now be extendedglucosamine reduces activity, particularly against

    to other glycopeptides in which both the aglyconestreptococci and enterococci, whereas removal of N-

    and the carbohydrate moieties would be individuallyacetylglucosamine decreases the activity against

    optimised for interaction with their own target, so asstaphylococci but may confer activity against Gram-

    to generate very active compounds.negative as well as VanA-type enterococci. This isespecially true for derivatives that also have a basic

    2.3 Inhibitors of Resistance Mechanismsamide substitution of the C-terminus.[116]

    Substituents on sugars also play important roles.In the same way as clavulanic acid or sulbactam

    As mentioned in the previous section, lipophilichave been designed to restore the activity of β-

    substituents are of major interest for vancomycinlactam agents against β-lactamase-producing organ-

    activity. Teicoplanin contains naturally a lipophilicisms, inhibitors of glycopeptide resistance mechan-

    side chain that is responsible for its prolonged half-isms could be considered to specifically circumvent

    life. This side chain is also important for activity,the mechanisms of resistance developed by entero-

    since deacylated derivatives are only weak antibio-cocci (which, as described in section 1.2, may also

    tics.[116] The acyl chain has been proposed not onlyapply to the recently described highly resistant S.

    to favour anchoring in the membrane, and thereforeaureus strains). This goal has been achieved by

    the interaction with the target,[11] but also to circum-designing small molecules such as ε-aminopenta-

    vent VanB induction,[129] explaining teicoplanin ac-noylated prolinol,[133] which are able to selectively

    tivity against resistant strains of the VanB-type. Oncleave the ester linkage of the D-Ala-D-Lac dep-

    the other hand, the replacement of the N-acetyl-sipeptide of resistant bacteria. Concomitant admin-

    glucosamine by an N-acylglucuronic acid (as inistration of this compound and vancomycin in-

    A40926, see figure 1) has a detrimental effect oncreases the activity of the glycopeptide in vitro

    activity against VanA enterococci but improvesagainst VanA-type enterococci, without affecting its

    anti-Gram-negative activity.[116]activity against susceptible strains. However, ac-tivity remains globally low, probably because VanX2.2 Hybrid and Multivalent Glycopeptideshydrolyses the D-Ala-D-Ala dipeptide, so that there

    On the basis of current knowledge on the mode of is no pentapeptide left for binding glycopeptides.action of glycopeptides, more innovative strategies These inhibitors will, therefore, need to be com-have been explored to optimise activity against both bined with inhibitors of the VanX D-D-pepti-susceptible and resistant strains. First, considering dase.[134,135] They have been obtained with inhibito-the potential cooperative binding occurring in multi- ry potencies in the micromolar range, but microbio-valent drugs, as well as the enhanced activity of logical data are still lacking to demonstrate theglycopeptides capable of self-association, dimers of interest of combining these inhibitors with vanco-vancomycin have been prepared using different mycin.

    © 2004 Adis Data Information BV. All rights reserved. Drugs 2004; 64 (9)

  • New Glycopeptides 927

    3. Properties of the Ideal Glycopeptide cosaminyl moiety of vancomycin by a lipophilicside chain resulted in enhanced activity and in some

    Based on an examination of the limitations of the cases restored activity towards vancomycin-resis-current molecules and also on the promising proper- tant enterococci.[121,137]ties of some of the semisynthetic derivatives de-scribed in section 2, we delineate in table VI the 4.1.1 Microbiological andmain desirable characteristics expected from new Pharmacological Propertiesglycopeptides for future clinical development. Twomolecules, oritavancin and dalbavancin, fulfil these Spectrum of Activity and Resistanceconditions in various ways and are currently in The activity of oritavancin (table II) is similar tophase II/III of clinical development. that of vancomycin against staphylococci, with

    MICs ranging between 0.03 and 8 mg/L; no4. Glycopeptides Undergoing differences are seen between methicillin-susceptibleClinical Evaluation and -resistant stains. Against enterococci, orita-

    vancin is more active than vancomycin or teico-planin, with MICs consistently lower than 1 mg/L.4.1 OritavancinRemarkably, it is as active against glycopeptide-resistant strains as against glycopeptide-susceptibleOritavancin (LY333328) was discovered at Eliisolates. Oritavancin is also very potent againstLilly (Indianapolis, IN, USA) but is now developedpneumococci (both penicillin-susceptible and -resis-by InterMune (Brisbane, CA, USA)[136] [see figure 1tant). It shows low MICs against other Gram-posi-for structure]. This molecule was obtained by

    reductive alkylation with 4′-chloro-biphenyl-car- tive species including other streptococci, Listeriaboxaldehyde of A82846B (chloroeremomycin; spp., Clostridium spp. and corynebacteria,[50] but isLY264826), a natural glycopeptide that differs from not active against Gram-negative bacteria, includingvancomycin by the addition of a 4-epi-vancosamine Haemophilus influenzae. Resistance of S. aureus tosugar and the replacement of the vancosamine of the oritavancin has so far not been described in clinicaldisaccharide moiety by an epi-vancosamine.[122] The isolates,[51,52,138] including VISA strains. In contrast,design of this drug was based on previous observa- enterococci with reduced susceptibility to orita-tions that chloroeremomycin was more potent than vancin (MIC 8–16 mg/L) have been obtained invancomycin because of its stronger tendency to self- vitro, using strains harbouring the vanA or the vanBassociate,[3] and that substitution of the van- gene clusters.[139] Three mechanisms seem to

    operate:[139] (i) the complete elimination of D-Ala-ending precursors by overexpression of the vanAgene cluster or by reduced expression of the host D-Ala:D-Ala ligase; (ii) mutations in the VanSB sensorof the vanB cluster (which can occur in clinicalisolates[140]); or (iii) the expression of VanZ, a pro-tein encoded by a gene present in the resistancetransposon but the precise function of which is stillunknown. A clinical strain of oritavancin-dependentEnterococcus faecalis has also been isolated but theunderlying mechanism should be different from thatconferring dependence to vancomycin and teico-planin, since the growth of this strain is not restoredin the presence of D-Ala-D-Ala precursors.[141]

    Table VI. Properties of an ideal glycopeptide

    Microbiological properties

    High intrinsic activity against Gram-positive organisms, includingmethicillin-resistant Staphylococcus aureus, vancomycin-resistantenterococci and glycopeptide-resistant S. aureus

    Pharmacodynamic properties

    Rapid and concentration-dependent bactericidal activity

    Pharmacokinetic properties

    Area under the concentration/time curve and peak plasmaconcentration for free fraction adequate to cover minimuminhibitory concentration of target pathogens

    Prolonged half-life (once daily administration)

    High diffusibility in tissues, including in the CNS

    Safety profile

    Lower incidence of adverse effects than current molecules

    © 2004 Adis Data Information BV. All rights reserved. Drugs 2004; 64 (9)

  • 928 Van Bambeke et al.

    Pharmacodynamic Properties in Relation to the the other glycopeptides. Like teicoplanin, orita-Mode of Action vancin is characterised by high protein binding toOritavancin is bactericidal in vitro, with MBCs albumin, which explains its prolonged retention in

    only 1- to 8-fold higher than the corresponding mammals, but also reduces the rate and extent of itsMICs against most of the covered microorga- bactericidal activity and shortens the postantibioticnisms.[50] Killing curve experiments have revealed a effect.[146,148] This could be important when serumstriking difference between vancomycin and orita- levels become close to the MIC for the infectingvancin concerning the rate of killing and its concen- organism. Further studies should address pharmaco-tration-dependent character. Oritavancin indeed kinetic-pharmacodynamic relationships for theseshows very rapid and highly concentration-depen- drugs, but we may anticipate, on the basis of itsdent bactericidal activity (3-log reduction in bacter- concentration-dependent bactericidal activity inial counts after 1–8 hours) in conditions where van- vitro, that high serum levels or AUC relative to MICcomycin requires at least 8–24 hours to reach the will correlate with optimal efficacy. Studies on cul-same effect.[50,142] However, oritavancin acts more tured cells also show that oritavancin accumulates inslowly against VRE.[143] These properties suggest eukaryotic cells to exceptional levels (the apparentthat oritavancin could have another mode of action cellular concentration reaching values as high as 400than conventional glycopeptides. The current view times the extracellular ones), which may be an ad-is that oritavancin combines the advantages of a vantage for the eradication of intracellular infec-high capacity to dimerise and to interact with the tions.[149]membrane, and therefore may cooperatively bind topeptidoglycan residues of both susceptible and re- Models of Infectionsistant strains. It may also directly perturb the mem- The promising in vitro activity of oritavancin hasbrane properties and inhibit transglycosylation reac- prompted evaluation of its activity in models oftions.[125] difficult-to-treat infections. As anticipated, orita-

    Oritavancin also displays a concentration-depen- vancin given once daily is as effective as vancomy-dent postantibiotic effect, increasing from ~2 hours cin given every 8 hours against MRSA rabbit endo-at 1 × MIC to 4 hours against VRE and 8 hours carditis[150] and is bactericidal against S. pneu-against MRSA.[143] As with vancomycin, the combi- moniae rabbit meningitis,[151,152] even though thenation of oritavancin with gentamicin is synergis- drug penetration in the CSF reaches only 5% of thetic.[143-145] The combination with ampicillin en- serum concentration.[152] Oritavancin activity hashances the bactericidal activity of oritavancin, with- also been studied in models of infections byout being truly synergistic. It also prolongs its glycopeptide-resistant enterococci. While orita-postantibiotic effect against VRE from 18 to 23 vancin was effective in a model of rat central venoushours at 10 × MIC.[146] catheter infection,[153] its activity was more limited

    Oritavancin activity is negatively affected by in a rabbit endocarditis model, causing a reductionlarge inocula,[143] but not by acid pH or by the in the number of bacteria in the vegetations butgrowth phase of the bacteria.[147] However, as point- failing to sterilise them.[154] Increasing serum con-ed out by Mercier et al.,[147] activity might be slight- centrations to values as high as 80 mg/L did notly reduced against VRE in stationary growth phase, provide significant improvement and did not pre-as observed in animal model infective endocarditis vent the selection of resistant mutants, probablyor in acidic foci of infection. because of the heterogeneous distribution of the

    drug in the vegetations. Yet combination with genta-micin proved synergistic and bactericidal, and pre-Pharmacokinetic Profile and Relationship with

    Pharmacodynamic Properties vented the emergence of resistant mutants.[155] Intra-The pharmacokinetic properties of oritavancin cellular bactericidal activity of oritavancin has been

    are presented in table III in comparison with those of demonstrated in in vitro models of polymorphonu-

    © 2004 Adis Data Information BV. All rights reserved. Drugs 2004; 64 (9)

  • New Glycopeptides 929

    clear leucocytes infected by vancomycin-resistant terminal methylamino group.[164] In dalbavancin, aenterococci[156] or by MRSA[157] as well as in macro- 3,3-dimethylaminopropylamide replaces the peptidephages infected by S. aureus.[158] Taken together, carboxy group of A40926.[165] SAR studies of teico-these data suggest a definite clinical interest for this planin indicated an improvement in activity bydrug, provided it can reach the infected compart- derivatisation of its carboxy group, and improvedment at sufficiently high concentrations. Efficacy activity against VanA-type enterococci is expectedagainst infections occurring in less accessible com- to occur when removing the acetyl-partments as well as against intracellular organisms glucosamine.[48] The most active compound in theneeds, therefore, to be examined in the light of the series was actually the 6β-decarboxy-6β-hydrox-tissue distribution of the drug in vivo. ymethyl amide of A40926,[165] but development of

    this compound was halted because of its poor tolera-4.1.2 Clinical Studies bility in animals (causing adverse effects typical ofOritavancin is currently in phase III develop- histamine release, which have been ascribed to its

    ment, with two studies completed in the treatment of basic character[48]). Unfortunately, most data onpatients with complicated skin and skin structure dalbavancin have been published only as abstractsinfection caused by Gram-positive pathogens in- or as short quotations in review papers. This makescluding MRSA.[159] The intent-to-treat analysis of an in-depth comparison of dalbavancin withone of these, a double-blind, randomised study, has oritavancin difficult.been presented as an abstract[160] and showed equi-valent clinical success with oritavancin at 1.5 or 4.2.1 Microbiological and3 mg/kg for 3–7 days versus vancomycin (15 mg/kg Pharmacological Propertiestwice daily for 3–7 days followed by oral cephalexinfor up to 10–14 days in the vancomycin group). Spectrum of Activity and Resistance

    In general (table II), dalbavancin is more potent4.1.3 Safety Profilethan vancomycin, teicoplanin and, to some extent,The only published data available so far are thoseoritavancin against staphylococci (with MICsof the phase I studies, in which oritavancin was wellranging from

  • 930 Van Bambeke et al.

    trough concentrations of dalbavancin are above the with clindamycin, ceftriaxone, vancomycin orcurrent MIC of the targeted microorganisms. cefazolin) for the treatment of deep skin and soft

    tissue infections caused by methicillin-susceptiblePharmacodynamic Properties in Relation to the S. aureus or MRSA.Mode of ActionLike oritavancin, dalbavancin is bactericidal, 4.2.3 Safety Profile

    with MBC/MIC ratios close to 1, even in the pres- Preclinical studies in rats and dogs show thatence of 30% serum.[66] It is synergic with ampicillin, dalbavancin is well tolerated after intravenous bolusincluding against VanA-type enterococci. administration at doses several times higher than

    those expected to be used in humans.[48] Phase I orPharmacokinetic Profile and Relationship with

    phase II clinical trials have not reported major ad-Pharmacodynamic Propertiesverse effects in the range of concentrations whereThe pharmacokinetic profile of dalbavancin isdalbavancin is effective.[66,169] No dosage adjust-compared with that of the other glycopeptides inments are necessary in case of mild renal insuffi-table III. Phase I studies have used higher dosages,ciency.[170]providing higher AUC values. However, when

    corrected for the free fraction, the AUC/MIC and 5. New Glycopeptides in the Clinics: forCmax/MIC ratios of dalbavancin are in the same Which Indications?order of magnitude as those of teicoplanin. Plasmadalbavancin concentrations still exceed the MBC The improved pharmacological properties of newvalues for staphylococci 1 week after administration glycopeptides make them very potent agents, withof a single dose of 1000mg but free serum concen- potentially large clinical indications. However, theirtrations are close to the MIC.[66,67] use should be as restricted as that of conventional

    glycopeptides if one wishes to avoid the rapid emer-Models of Infection

    gence of resistance.The activity of dalbavancin has been assessed in

    The advantages of oritavancin that should provemodels of infections by glycopeptide-susceptibleto be useful in clinical practice include: (i) its fastGram-positive organisms and by GISA. Preliminaryrate of bacterial killing, its spectrum of activity,reports indicate a reduction in bacterial load after awhich covers VRE, methicillin-resistant staphylo-single-dose administration in models of penicillin-cocci and to some extent the GISA strains; (ii) itsresistant S. pneumoniae pneumonia or of MRSA ratactivity against intracellular forms of enterococcipouch infection.[48,167] Its activity is superior to thatand staphylococci; and (iii) its prolonged half-life.of conventional glycopeptides in models of endocar-For dalbavancin, the main improvement seems to beditis due to MRSA or GISA, and of acute septicae-its pharmacokinetic profile, which should allowmia caused by staphylococci, streptococci or entero-once-weekly administration. Microbiological prop-cocci.[66,168]erties of dalbavancin are less favourable than those

    4.2.2 Clinical Studies of oritavancin, since VRE are not covered.No clinical data have been published so far but On the basis of these properties, and also in view

    phase II/III clinical trials are ongoing to evaluate the of the results from animal models, oritavancin mayefficacy and safety of dalbavancin for the treatment become a drug of interest for infections caused byof complicated skin and soft tissue infections caused VRE, while both oritavancin and dalbavancin couldby Gram-positive organisms or the use of dalba- be considered for severe infections by multiresistantvancin in a once-a-week mode of administration for organisms, such as methicillin-resistant staphylo-other Gram-positive hospital infections.[66] In a first cocci or S. pneumoniae. In the latter case, however,report of a phase II study,[169] administration of two other alternatives such as fluoroquinolones (moxi-doses (1000mg on day 1 and 500mg on day 8) was floxacin or gatifloxacin) should be examined first.found to be as effective as comparators (7–21 days In some countries resistance to fluoroquinolones has

    © 2004 Adis Data Information BV. All rights reserved. Drugs 2004; 64 (9)

  • New Glycopeptides 931

    already reached alarming levels (15% in Southeast 6. ConclusionAsia [Hong Kong, South Korea[171]]). There are also

    Oritavancin, and to a lesser extent dalbavancin,situations such as CNS infections in which fluoro- offer considerable possibilities for improvementquinolones are poorly efficient.[172,173] Oritavancin over vancomycin and teicoplanin, essentially inand dalbavancin could, therefore, prove very useful terms of pharmacokinetic and pharmacodynamicin these ‘niche’ but important indications. Finally, properties, as well as of activity against resistanttheir activity against GISA strains appears limited strains (for oritavancin). Whether these properties

    can translate into improved clinical efficacy awaits(table II), which could be a deterrent to recom-completion of clinical studies. Should these mole-mending them in these situations. However, synergycules soon become approved for clinical use, it ishas been demonstrated for dalbavancin when com-vital that they are used rationally and prudently tobined with β-lactam agents.[57] Encouraging resultsprotect against the rapid emergence of resistancehave also been obtained in animal models,[168] whichand extend their useful life.

    should stimulate further testing of this moleculeagainst GISA infections. Acknowledgements

    Therefore, reasonable indications could include F. Van Bambeke is Chercheur Qualifié of the Belgianthe following: (i) nosocomial septicaemia or infec- Fonds National de la Recherche Scientifique. The authors

    have provided no information on sources of funding or ontions of deep organs caused by MRSA or VREconflicts of interest directly relevant to the content of this

    (oritavancin only); (ii) severe and recurrent skin and review.soft tissue infections caused by MRSA (in relationwith activity against both extra- and intracellular References

    1. Reynolds PE. Structure, biochemistry and mechanism of actionbacteria [demonstrated so far only for oritavancin]);of glycopeptide antibiotics. Eur J Clin Microbiol Infect Dis

    and (iii) endocarditis due to resistant organisms (but 1989 Nov; 8 (11): 943-502. Arthur M, Reynolds P, Courvalin P. Glycopeptide resistance inprobably in combination with an aminoglycoside for

    enterococci. Trends Microbiol 1996 Oct; 4 (10): 401-7oritavancin against VRE, based on disappointing 3. Williams DH, Waltho JP. Molecular basis of the activity of

    antibiotics of the vancomycin group. Biochem Pharmacolanimal data[155]). The usefulness of oritavancin in1988 Jan 1; 37 (1): 133-41

    nosocomial CNS infections caused by MRSA, resis- 4. Loll PJ, Axelsen PH. The structural biology of molecular recog-nition by vancomycin. Annu Rev Biophys Biomol Struct 2000;tant S. pneumoniae or VRE may be suggested on the29: 265-89

    basis of its activity in animal models. No corre- 5. Hiramatsu K. Vancomycin-resistant Staphylococcus aureus: anew model of antibiotic resistance. Lancet Infect Dis 2001 Oct;sponding data exist for dalbavancin, for which pene-1 (3): 147-55

    tration in CSF is low. The potential use of orita- 6. Schafer M, Schneider TR, Sheldrick GM. Crystal structure ofvancomycin. Structure 1996 Dec 15; 4 (12): 1509-15vancin in humans in this indication needs to be

    7. Groves P, Searle MS, Mackay JP, et al. The structure of anassessed, as well as the influence of corticosteroids asymmetric dimer relevant to the mode of action of theglycopeptide antibiotics. Structure 1994 Aug 15; 2 (8): 747-54on the CNS penetration of these new glycopeptides

    8. Shiozawa H, Chia BC, Davies NL, et al. Cooperative binding(influence of corticosteroids was found to be minor interactions of glycopeptide antibiotics. J Am Chem Soc 2002Apr 17; 124 (15): 3914-9for vancomycin).

    9. Mackay JP, Gerhard U, Beauregard DA, et al. GlycopeptideGlobally speaking, clinical studies or additional antibiotic activity and the possible role of dimerization: a

    model for biological signalling. J Am Chem Soc 1994; 116:animal data are needed to confirm the interest of4581-90

    new glycopeptides in these indications, since the 10. Mackay JP, Gerhard U, Beauregard DA, et al. Dissection of thecontributions towards dimerization of glycopeptide antibiotics.proposals made above rely almost exclusively on anJ Am Chem Soc 1994; 116: 4573-80

    analysis of the pharmacological properties of these 11. Beauregard DA, Williams DH, Gwynn MN, et al. Dimerizationand membrane anchors in extracellular targeting of vancomy-drugs and on the results obtained in animal modelscin group antibiotics. Antimicrob Agents Chemother 1995

    of infection. Mar; 39 (3): 781-5

    © 2004 Adis Data Information BV. All rights reserved. Drugs 2004; 64 (9)

  • 932 Van Bambeke et al.

    12. Williams DH, Maguire AJ, Tsuzuki W, et al. An analysis of the 30. Kenner J, O’Connor T, Piantanida N, et al. Rates of carriage oforigins of a cooperative binding energy of dimerization. methicillin-resistant and methicillin-susceptible Staphylococ-Science 1998 May 1; 280 (5364): 711-4 cus aureus in an outpatient population. Infect Control Hosp

    Epidemiol 2003 Jun; 24 (6): 439-4413. Kaplan J, Korty BD, Axelsen PH, et al. The role of sugar31. Schouten MA, Hoogkamp-Korstanje JA, Meis JF, et al. Preva-residues in molecular recognition by vancomycin. J Med

    lence of vancomycin-resistant enterococci in Europe. Eur JChem 2001 May 24; 44 (11): 1837-40Clin Microbiol Infect Dis 2000 Nov; 19 (11): 816-2214. Ge M, Chen Z, Onishi HR, et al. Vancomycin derivatives that

    32. van den Bogaard AE, Mertens P, London NH, et al. Highinhibit peptidoglycan biosynthesis without binding D-Ala-D-prevalence of colonization with vancomycin- and pristi-Ala. Science 1999 Apr 16; 284 (5413): 507-11namycin-resistant enterococci in healthy humans and pigs in15. Printsevskaya SS, Pavlov AY, Olsufyeva EN, et al. Role of theThe Netherlands: is the addition of antibiotics to animal feedsglycopeptide framework in the antibacterial activity of hydro-to blame? J Antimicrob Chemother 1997 Sep; 40 (3): 454-6phobic derivatives of glycopeptide antibiotics. J Med Chem

    33. van den Braak N, van Belkum A, van Keulen M, et al. Molecu-2003 Mar 27; 46 (7): 1204-9lar characterization of vancomycin-resistant enterococci from16. Gholizadeh Y, Courvalin P. Acquired and intrinsic glycopeptidehospitalized patients and poultry products in The Netherlands.resistance in enterococci. Int J Antimicrob Agents 2000 Nov;J Clin Microbiol 1998 Jul; 36 (7): 1927-3216 Suppl. 1: S11-7

    34. Klare I, Badstubner D, Konstabel C, et al. Decreased incidence17. Centers for Disease Control and Prevention. Staphylococcusof VanA-type vancomycin-resistant enterococci isolated fromaureus resistant to vancomycin: United States, 2002. MMWRpoultry meat and from fecal samples of humans in the com-Morb Mortal Wkly Rep 2002; 51: 565-7munity after discontinuation of avoparcin usage in animal

    18. Chang S, Sievert DM, Hageman JC, et al. Infection with husbandry. Microb Drug Resist 1999; 5 (1): 45-52vancomycin-resistant Staphylococcus aureus containing the

    35. Aarestrup FM, Seyfarth AM, Emborg HD, et al. Effect ofvanA resistance gene. N Engl J Med 2003 Apr 3; 348 (14):abolishment of the use of antimicrobial agents for growth1342-7promotion on occurrence of antimicrobial resistance in fecal

    19. Bozdogan B, Esel D, Whitener C, et al. Antibacterial suscepti- enterococci from food animals in Denmark. Antimicrobbility of a vancomycin-resistant Staphylococcus aureus strain Agents Chemother 2001 Jul; 45 (7): 2054-9isolated at the Hershey Medical Center. J Antimicrob

    36. van den Bogaard AE, Stobberingh EE. Epidemiology of resis-Chemother 2003 Nov; 52 (5): 864-8tance to antibiotics: links between animals and humans. Int J

    20. Tenover FC, Weigel LM, Appelbaum PC, et al. Vancomycin- Antimicrob Agents 2000 May; 14 (4): 327-35resistant Staphylococcus aureus isolate from a patient in Penn-

    37. Bugg TD, Wright GD, Dutka-Malen S, et al. Molecular basissylvania. Antimicrob Agents Chemother 2004 Jan; 48 (1):

    for vancomycin resistance in Enterococcus faecium BM4147:275-80

    biosynthesis of a depsipeptide peptidoglycan precursor by21. Poyart C, Pierre C, Quesne G, et al. Emergence of vancomycin vancomycin resistance proteins VanH and VanA. Biochemis-

    resistance in the genus Streptococcus: characterization of a try 1991 Oct 29; 30 (43): 10408-15vanB transferable determinant in Streptococcus bovis. Antimi- 38. Arthur M, Molinas C, Depardieu F, et al. Characterization ofcrob Agents Chemother 1997 Jan; 41 (1): 24-9 Tn1546, a Tn3-related transposon conferring glycopeptide re-

    22. Perichon B, Reynolds P, Courvalin P. VanD-type glycopeptide- sistance by synthesis of depsipeptide peptidoglycan precursorsresistant Enterococcus faecium BM4339. Antimicrob Agents in Enterococcus faecium BM4147. J Bacteriol 1993 Jan; 175Chemother 1997 Sep; 41 (9): 2016-8 (1): 117-27

    23. Fines M, Perichon B, Reynolds P, et al. VanE, a new type of 39. Quintiliani Jr R, Courvalin P. Conjugal transfer of the vancomy-acquired glycopeptide resistance in Enterococcus faecalis cin resistance determinant vanB between enterococci involvesBM4405. Antimicrob Agents Chemother 1999 Sep; 43 (9): the movement of large genetic elements from chromosome to2161-4 chromosome. FEMS Microbiol Lett 1994 Jun 15; 119 (3): 359-

    24. McKessar SJ, Berry AM, Bell JM, et al. Genetic characteriza- 63tion of vanG, a novel vancomycin resistance locus of Entero- 40. Baptista M, Depardieu F, Reynolds P, et al. Mutations leadingcoccus faecalis. Antimicrob Agents Chemother 2000 Nov; 44 to increased levels of resistance to glycopeptide antibiotics in(11): 3224-8 VanB-type enterococci. Mol Microbiol 1997 Jul; 25 (1): 93-

    25. Hiramatsu K, Hanaki H, Ino T, et al. Methicillin-resistant 105Staphylococcus aureus clinical strain with reduced vancomy- 41. Van Bambeke F, Chauvel M, Reynolds PE, et al. Vancomycin-cin susceptibility. J Antimicrob Chemother 1997 Jul; 40 (1): dependent Enterococcus faecalis clinical isolates and revertant135-6 mutants. Antimicrob Agents Chemother 1999 Jan; 43 (1): 41-7

    26. Sieradzki K, Tomasz A. Gradual alterations in cell wall struc- 42. Hamilton-Miller JM. Vancomycin-resistant Staphylococcus au-ture and metabolism in vancomycin-resistant mutants of reus: a real and present danger? Infection 2002 Jun; 30 (3):Staphylococcus aureus. J Bacteriol 1999 Dec; 181 (24): 7566- 118-2470 43. Hamilton-Miller JM. Glycopeptide-resistant staphylococci. Int J

    27. Leclercq R, Derlot E, Duval J, et al. Plasmid-mediated resis- Antimicrob Agents 1999 Sep; 13 (1): 63-5tance to vancomycin and teicoplanin in Enterococcus faecium. 44. Cui L, Murakami H, Kuwahara-Arai K, et al. Contribution of aN Engl J Med 1988 Jul 21; 319 (3): 157-61 thickened cell wall and its glutamine nonamidated component

    28. Uttley AH, Collins CH, Naidoo J, et al. Vancomycin-resistant to the vancomycin resistance expressed by Staphylococcusenterococci. Lancet 1988 Jan 2; I (8575-6): 57-8 aureus Mu50. Antimicrob Agents Chemother 2000 Sep; 44

    (9): 2276-8529. Bonten MJ, Willems R, Weinstein RA. Vancomycin-resistantenterococci: why are they here, and where do they come from? 45. Hiramatsu K, Okuma K, Ma XX, et al. New trends in Staphylo-Lancet Infect Dis 2001 Dec; 1 (5): 314-25 coccus aureus infections: glycopeptide resistance in hospital

    © 2004 Adis Data Information BV. All rights reserved. Drugs 2004; 64 (9)

  • New Glycopeptides 933

    and methicillin resistance in the community. Curr Opin Infect gram-positive species and corynebacteria. Antimicrob AgentsDis 2002 Aug; 15 (4): 407-13 Chemother 2003 Jun; 47 (6): 1968-71

    46. May J, Shannon K, King A, et al. Glycopeptide tolerance in 63. Wilson AP. Clinical pharmacokinetics of teicoplanin. ClinStaphylococcus aureus. J Antimicrob Chemother 1998 Aug; Pharmacokinet 2000 Sep; 39 (3): 167-8342 (2): 189-97 64. Feketi R. Vancomycin, teicoplanin, and the streptogramins:

    47. Johnson AP, Woodford N. Glycopeptide-resistant Staphylococ- quinupristin and dalfopristin. In: Mandell GE, Bennett JE,cus aureus. J Antimicrob Chemother 2002 Nov; 50 (5): 621-3 Dolin R, editors. Principles and practice of infectious diseases.

    Philadelphia (PA): Churchill Livingstone, 2000: 382-9248. Malabarba A, Ciabatti R. Glycopeptide derivatives. Curr MedChem 2001 Dec; 8 (14): 1759-73 65. Rowe PA, Brown TJ. Protein binding of 14C-oritavancin [ab-

    stract no. A-2193]. 41st Interscience Conference on Anti-49. Candiani GP, Abbondi M, Borgonovi M, et al. In-vitro and in-microbial Agents and Chemotherapy; 2001 Dec 16-18; Chica-vivo antibacterial activity of BI 397, a new semi-syntheticgo (IL)glycopeptide antibiotic. J Antimicrob Chemother 1999 Aug;

    44 (2): 179-92 66. Steiert M, Schmitz FJ. Dalbavancin (Biosearch Italia/Versicor).Curr Opin Investig Drugs 2002 Feb; 3 (2): 229-3350. Biavasco F, Vignaroli C, Lupidi R, et al. In vitro antibacterial

    activity of LY333328, a new semisynthetic glycopeptide. An- 67. Dowell JA, Gottlieb AB, Van Sanders C, et al. The pharmaco-timicrob Agents Chemother 1997 Oct; 41 (10): 2165-72 kinetics and renal excretion of dalbavancin in healthy subjects

    [abstract no. A-1386]. 42nd Interscience Conference on Anti-51. Zeckel ML, Preston DA, Allen BS. In vitro activities ofmicrobial Agents and Chemotherapy; 2002 Sep 27-30; SanLY333328 and comparative agents against nosocomial gram-Diego (CA)positive pathogens collected in a 1997 global surveillance

    study. Antimicrob Agents Chemother 2000 May; 44 (5): 1370- 68. Cavaleri M, Cooper A, Nutley MA, et al. Protein binding of4 dalbavancin using isothermal titration microcalorimetry [ab-

    stract no. A-1385]. 42nd Interscience Conference on Anti-52. Garcia-Garrote F, Cercenado E, Alcala L, et al. In vitro activitymicrobial Agents and Chemotherapy; 2002 Sep 27-30; Sanof the new glycopeptide LY333328 against multiply resistantDiego (CA)gram-positive clinical isolates. Antimicrob Agents Chemother

    1998 Sep; 42 (9): 2452-5 69. Owen J. Population pharmacokinetic model. Brisbane (CA):InterMune, Inc., 2003. (Data on file)53. Jones RN, Biedenbach DJ, Johnson DM, et al. In vitro evalua-

    tion of BI 397, a novel glycopeptide antimicrobial agent. J 70. Thomasson HR. Study ARKK. Brisbane (CA): InterMune, Inc.,Chemother 2001 Jun; 13 (3): 244-54 1997. (Data on file)

    54. Jones RN, Barrett MS, Erwin ME. In vitro activity and spectrum 71. Lowdin E, Odenholt I, Cars O. In vitro studies of pharmacody-of LY333328, a novel glycopeptide derivative. Antimicrob namic properties of vancomycin against Staphylococcus aure-Agents Chemother 1997 Feb; 41 (2): 488-93 us and Staphylococcus epidermidis. Antimicrob Agents Che-

    mother 1998 Oct; 42 (10): 2739-4455. Noviello S, Ianniello F, Esposito S. In vitro activity ofLY333328 (oritavancin) against gram-positive aerobic cocci 72. Houlihan HH, Stokes DP, Rybak MJ. Pharmacodynamics ofand synergy with ciprofloxacin against enterococci. J An- vancomycin and ampicillin alone and in combination withtimicrob Chemother 2001 Aug; 48 (2): 283-6 gentamicin once daily or thrice daily against Enterococcus

    faecalis in an in vitro infection model. J Antimicrob56. Aeschlimann JR, Allen GP, Hershberger E, et al. Activities ofChemother 2000 Jul; 46 (1): 79-86LY333328 and vancomycin administered alone or in combina-

    tion with gentamicin against three strains of vancomycin- 73. Fantin B, Carbon C. Importance of the aminoglycoside dosingintermediate Staphylococcus aureus in an in vitro pharmaco- regimen in the penicillin- netilmicin combination for treatmentdynamic infection model. Antimicrob Agents Chemother 2000 of Enterococcus faecalis-induced experimental endocarditis.Nov; 44 (11): 2991-8 Antimicrob Agents Chemother 1990 Dec; 34 (12): 2387-91

    57. Hackbarth CJ, Lopez S, Trias J, et al. In vitro activity of the 74. Lamp KC, Rybak MJ, Bailey EM, et al. In vitro pharmacody-glycopeptide BI 397 against Staphylococcus aureus and namic effects of concentration, pH, and growth phase on serumStaphylococcus epidermidis [abstract no. 1283]. 39th Inter- bactericidal activities of daptomycin and vancomycin. An-science Conference on Antimicrobial Agents and Chemother- timicrob Agents Chemother 1992 Dec; 36 (12): 2709-14apy; 1999 Sep 26-28; San Francisco (CA) 75. Drabu YJ, Blakemore PH. The post-antibiotic effect of teico-

    58. Tenover FC, Lancaster MV, Hill BC, et al. Characterization of planin: monotherapy and combination studies. J Antimicrobstaphylococci with reduced susceptibilities to vancomycin and Chemother 1991 Apr; 27 Suppl. B: 1-7other glycopeptides. J Clin Microbiol 1998 Apr; 36 (4): 1020-7 76. Chambers HF, Kennedy S. Effects of dosage, peak and trough

    59. Schwalbe RS, McIntosh AC, Qaiyumi S, et al. In vitro activity concentrations in serum, protein binding, and bactericidal rateof LY333328, an investigational glycopeptide antibiotic, on efficacy of teicoplanin in a rabbit model of endocarditis.against enterococci and staphylococci. Antimicrob Agents Antimicrob Agents Chemother 1990 Apr; 34 (4): 510-4Chemother 1996 Oct; 40 (10): 2416-9 77. Peetermans WE, Hoogeterp JJ, Hazekamp-van Dokkum AM, et

    60. Harland S, Tebbs SE, Elliott TS. Evaluation of the in-vitro al. Antistaphylococcal activities of teicoplanin and vancomy-activity of the glycopeptide antibiotic LY333328 in compari- cin in vitro and in an experimental infection. Antimicrobson with vancomycin and teicoplanin. J Antimicrob Chemoth- Agents Chemother 1990 Oct; 34 (10): 1869-74er 1998 Feb; 41 (2): 273-6 78. Craig WA. Does the dose matter? Clin Infect Dis 2001 Sep 15;

    61. Sillerstrom E, Wahlund E, Nord CE. In vitro activity of LY 33 Suppl. 3: S233-7333328 against anaerobic gram-positive bacteria. J Chemother 79. Knudsen JD, Fuursted K, Raber S, et al. Pharmacodynamics of1999 Apr; 11 (2): 90-2 glycopeptides in the mouse peritonitis model of Streptococcus

    62. Goldstein EJ, Citron DM, Merriam CV, et al. In vitro activities pneumoniae or Staphylococcus aureus infection. Antimicrobof Dalbavancin and nine comparator agents against anaerobic Agents Chemother 2000 May; 44 (5): 1247-54

    © 2004 Adis Data Information BV. All rights reserved. Drugs 2004; 64 (9)

  • 934 Van Bambeke et al.

    80. Cohen E, Dadashev A, Drucker M, et al. Once-daily versus 97. Eliopoulos GM. Quinupristin-dalfopristin and linezolid: evi-twice-daily intravenous administration of vancomycin for in- dence and opinion. Clin Infect Dis 2003 Feb 15; 36 (4): 473-81fections in hospitalized patients. J Antimicrob Chemother 98. Olsen KM, Rebuck JA, Rupp ME. Arthralgias and myalgias2002 Jan; 49 (1): 155-60 related to quinupristin-dalfopristin administration. Clin Infect

    81. Klepser ME, Patel KB, Nicolau DP, et al. Comparison of Dis 2001 Feb 15; 32 (4): e83-6bactericidal activities of intermittent and continuous infusion 99. Rubinstein E, Prokocimer P, Talbot GH. Safety and tolerabilitydosing of vancomycin against methicillin-resistant Staphylo- of quinupristin/dalfopristin: administration guidelines. J An-coccus aureus and Enterococcus faecalis. Pharmacotherapy timicrob Chemother 1999 Sep; 44 Suppl. A: 37-461998 Sep; 18 (5): 1069-74 100. Gerson SL, Kaplan SL, Bruss JB, et al. Hematologic effects of

    82. James JK, Palmer SM, Levine DP, et al. Comparison of linezolid: summary of clinical experience. Antimicrob Agentsconventional dosing versus continuous-infusion vancomycin Chemother 2002 Aug; 46 (8): 2723-6therapy for patients with suspected or documented gram-posi-

    101. Nilius AM. Have the oxazolidinones lived up to their billing?:tive infections. Antimicrob Agents Chemother 1996 Mar; 40future perspectives for this antibacterial class. Curr Opin In-(3): 696-700vestig Drugs 2003 Feb; 4 (2): 149-55

    83. Byl B, Jacobs F, Wallemacq P, et al. Vancomycin penetration102. Centers for Disease Control and Prevention. Recommendationsof uninfected pleural fluid exudate after continuous or inter-

    for preventing the spread of vancomycin resistance: Hospitalmittent infusion. Antimicrob Agents Chemother 2003 Jun; 47Infection Control Practices Advisory Committee (HICPAC).(6): 2015-7Infect Control Hosp Epidemiol 1995 Feb; 16 (2): 105-13

    84. Wysocki M, Delatour F, Faurisson F, et al. Continuous versus103. Gordts B, Firre E, Jordens P, et al. National guidelines for theintermittent infusion of vancomycin in severe staphylococcal

    judicious use of glycopeptides in Belgium. Clin Microbiolinfections: prospective multicenter randomized study. An-Infect 2000 Nov; 6 (11): 585-92timicrob Agents Chemother 2001 Sep; 45 (9): 2460-7

    104. Nourse C, Byrne C, Leonard L, et al. Glycopeptide prescribing85. Basma V, Van Bambeke F, Mingeot-Leclercq MP, et al. Stabili-in a tertiary referral paediatric hospital and applicability ofty and compatibility of vancomycin for administration byhospital infection control practices advisory committeecontinuous infusion in intensive care patients [abstract]. 14th(HICPAC) guidelines to children. Eur J Pediatr 2000 Mar; 159European Congress of Clinical Microbiology and Infectious(3): 193-7Diseases; 2004 May 1-4; Prague. In press

    105. Gruneberg RN, Antunes F, Chambers HF, et al. The role of86. Cheung RP, DiPiro JT. Vancomycin: an update. Pharmacother-glycopeptide antibiotics in the treatment of infective endocar-apy 1986 Jul; 6 (4): 153-69ditis. Int J Antimicrob Agents 1999 Aug; 12 (3): 191-8

    87. Livermore DM. Antibiotic resistance in staphylococci. Int J106. Manley HJ, Bailie GR, Frye RF, et al. Intravenous vancomycinAntimicrob Agents 2000 Nov; 16 Suppl. 1: S3-10

    pharmacokinetics in automated peritoneal dialysis patients.88. Michel M, Gutmann L. Methicillin-resistant Staphylococcus

    Perit Dial Int 2001 Jul; 21 (4): 378-85aureus and vancomycin-resistant enterococci: therapeutic real-

    107. Stamatiadis D, Papaioannou MG, Giamarellos-Bourboulis EJ,ities and possibilities. Lancet 1997 Jun 28; 349 (9069): 1901-6et al. Pharmacokinetics of teicoplanin in patients undergoing89. Kirst HA, Thompson DG, Nicas TI. Historical yearly usage ofcontinuous ambulatory peritoneal dialysis. Perit Dial Int 2003vancomycin. Antimicrob Agents Chemother 1998 May; 42Mar; 23 (2): 127-31(5): 1303-4

    108. Farber BF, Moellering Jr RC. Retrospective study of the toxicity90. Centers for Disease Control and Prevention. Nosocomial entero-of preparations of vancomycin from 1974 to 1981. Antimicrobcocci resistant to vancomycin: United States, 1989-1993.Agents Chemother 1983 Jan; 23 (1): 138-41MMWR Morb Mortal Wkly Rep 1993 Aug 6; 42 (30): 597-9

    109. Wang LS, Liu CY, Wang FD, et al. Chromatographically91. Fridkin SK, Edwards JR, Courval JM, et al. The effect ofpurified vancomycin: therapy of serious infections caused byvancomycin and third-generation cephalosporins on preva-Staphylococcus aureus and other gram-positive bacteria. Clinlence of vancomycin-resistant enterococci in 126 US adultTher 1988; 10 (5): 574-84intensive care units. Ann Intern Med 2001 Aug 7; 135 (3): 175-

    110. Sivagnanam S, Deleu D. Red man syndrome. Crit Care 200383Apr; 7 (2): 119-2092. Kumana CR, Ching TY, Kong Y, et al. Curtailing unnecessary

    111. Sorrell TC, Collignon PJ. A prospective study of adverse reac-vancomycin usage in a hospital with high rates of methicillintions associated with vancomycin therapy. J Antimicrobresistant Staphylococcus aureus infections. Br J Clin Pharma-Chemother 1985 Aug; 16 (2): 235-41col 2001 Oct; 52 (4): 427-32

    112. Elting LS, Rubenstein EB, Kurtin D, et al. Mississippi mud in93. Shojania KG, Yokoe D, Platt R, et al. Reducing vancomycinthe 1990s: risks and outcomes of vancomycin-associated tox-use utilizing a computer guideline: results of a randomizedicity in general oncology practice. Cancer 1998 Dec 15; 83controlled trial. J Am Med Inform Assoc 1998 Nov; 5 (6): 554-(12): 2597-60762

    113. Rocha JL, Kondo W, Baptista MI, et al. Uncommon vancomy-94. Goeckner BJ, Hendershot E, Scott K, et al. A vancomycincin-induced side effects. Braz J Infect Dis 2002 Aug; 6 (4):monitoring program at a community hospital. Jt Comm J Qual196-200Improv 1998 Jul; 24 (7): 379-85

    114. Wood MJ. The comparative efficacy and safety of teicoplanin95. Hamilton CD, Drew R, Janning SW, et al. Excessive use ofand vancomycin. J Antimicrob Chemother 1996 Feb; 37 (2):vancomycin: a successful intervention strategy at an academic209-22medical center. Infect Control Hosp Epidemiol 2000 Jan; 21

    (1): 42-5 115. Boger DL. Vancomycin, teicoplanin, and ramopl


Recommended