You are on page 1of 6

SUPPLEMENT ARTICLE

Oritavancin: Mechanism of Action


George G. Zhanel,1,2,3,4 Frank Schweizer,1,2 and James A. Karlowsky1,2,3,4
Departments of 1Medical Microbiology, and 2Chemistry, University of Manitoba, and Departments of 3Clinical Microbiology, and 4Medicine, Health
Sciences Centre, Winnipeg, Canada

Downloaded from https://academic.oup.com/cid/article-abstract/54/suppl_3/S214/290034 by guest on 16 March 2019


Oritavancin is a semisynthetic lipoglycopeptide analogue of vancomycin that contains the heptapeptide
core common to all glycopeptides. It differs from vancomycin by the presence of a hydrophobic
N-4-(4-chlorophenyl)benzyl (also referred to as 4#-chlorobiphenylmethyl) substituent on the disaccharide
sugar, the addition of a 4-epi-vancosamine monosaccharide to the amino acid residue in ring 6, and the
replacement of the vancosamine moiety by 4-epi-vancosamine. One mechanism of action of oritavancin is
inhibition of transglycosylation (important in peptidoglycan synthesis) by binding to D-alanyl-D-alanine stem
termini in Gram-positive bacteria. The inhibition of peptidoglycan synthesis via inhibition of transglycosylation is
common to all glycopeptides (vancomycin) and lipoglycopeptides. Secondary binding of oritavancin to the
pentaglycyl (Asp/Asn) bridging segment in peptidoglycan also occurs, which distinguishes it from vancomycin
and contributes to oritavancin’s activity versus vancomycin-resistant organisms. The presence of the hydrophobic
4#-chlorobiphenylmethyl group allows for interaction and disruption of the cell membrane, resulting in
depolarization, permeabilization, and concentration-dependent, rapid cell death. This mechanism is shared with
telavancin but not vancomycin and results in activity against daptomycin-nonsusceptible organisms. In conclusion,
oritavancin’s mechanism of action involves at least 3 known mechanisms: inhibition of transglycosylation,
inhibition of transpeptidation, and cell membrane interaction/disruption. Oritavancin’s multiple mechanisms of
action confer activity against vancomycin-susceptible and -resistant organisms, as well as rapid, concentration-
dependent killing versus actively growing, stationary phase, and biofilm-producing Gram-positive bacteria.

Reports of multidrug-resistant, Gram-positive patho- problem is the emergence of vancomycin-intermediate


gens such as methicillin-resistant Staphylococcus aureus S. aureus (VISA), vancomycin-resistant S. aureus (VRSA),
(MRSA) appeared in the scientific literature as early as and vancomycin-resistant Enterococcus (VRE), which has
the 1960s [1, 2]. However, it was not until the 1980s that added a greater sense of urgency to the search for novel
MRSA became widespread [1]. The steep rise in the antimicrobials with activity against resistant Gram-
prevalence of MRSA in hospitals during that decade positive pathogens [1, 3].
resulted in a sharp increase in vancomycin use [1, 2]. For decades vancomycin was viewed by many as the
Today, healthcare-associated MRSA is a worldwide con- gold standard for the treatment of MRSA infections
cern, and the percentage of S. aureus isolates resistant [4–6]. Although it is active against the vast majority of
to methicillin exceeds 60% in some intensive care units MRSA, vancomycin is plagued by poor penetration into
[1, 2]. Many countries, including the United States and certain tissues (eg, lung) and slow bactericidal activity
Canada, are also experiencing a significant increase in the [4, 6]. In addition, vancomycin’s minimum inhibitory
number of community-associated MRSA infections in concentration (MIC) for Gram-positive pathogens such
otherwise healthy individuals [1, 2]. Compounding the as MRSA has been reported to be increasing, with ther-
apeutic failures also being reported [7–11]. Furthermore,
Correspondence: George G. Zhanel, PhD, MS 673 Microbiology, Health investigators are reporting the development of hetero-
Sciences Centre, 820 Sherbrook St, Winnipeg, Manitoba R3A 1R9, Canada
resistant vancomycin-intermediate S. aureus (hVISA) and
(ggzhanel@pcs.mb.ca).
Clinical Infectious Diseases 2012;54(S3):S214–9
VISA [1, 3]. The rampant spread of MRSA and VRE and
Ó The Author 2012. Published by Oxford University Press on behalf of the the inherent limitations of vancomycin therapy highlight
Infectious Diseases Society of America. All rights reserved. For Permissions,
please e-mail: journals.permissions@oup.com.
the need for novel, rapidly bactericidal agents to combat
DOI: 10.1093/cid/cir920 infections with these pathogens [12].

S214 d CID 2012:54 (Suppl 3) d Zhanel et al


Eli Lilly identified and developed vancomycin in the 1950s STRUCTURE OF ORITAVANCIN
as a response to the emergence of penicillin-resistant S. aureus
[6, 13, 14]. In the early 1980s, Eli Lilly renewed its efforts to All glycopeptides, including vancomycin, contain a common
identify other glycopeptides with enhanced properties relative to heptapeptide core and are glycosylated (Figure 1) [5, 6, 13].
vancomycin (ie, active vs vancomycin-resistant strains, better The heptapeptide core contains at least 5 amino acid residues
pharmacodynamics, etc). They identified chloroeremomycin, bearing aromatic side chains (these 5 rings are highlighted in
a vancomycin analogue with a different vancosamine sugar than Figure 1). Oritavancin, a vancomycin analogue, is a synthetic
vancomycin and an additional vancosamine epimer not present derivative of the naturally occurring glycopeptide chloroer-
in vancomycin’s structure (Figure 1) [13, 14]. Chloroeremomycin emomycin (Figure 1) [13, 14]. Chloroeremomycin belongs to
displayed modestly improved activity compared with vanco- the eremomycin class, which differs from vancomycin by the
mycin, against methicillin-susceptible S. aureus (MSSA), MRSA, presence of a 4-epi-vancosamine monosaccharide at the amino
and vancomycin-susceptible and -resistant strains of enterococci acid residue in ring 6 and the substitution of the vancosamine by

Downloaded from https://academic.oup.com/cid/article-abstract/54/suppl_3/S214/290034 by guest on 16 March 2019


[13, 14]. Eli Lilly medicinal chemists synthesized .500 analogues 4-epi-vancosamine (Figure 1) [13, 14]. These structural differ-
of chloroeremomycin and identified a chlorobiphenylmethyl ences, compared with vancomycin, confer on chloroeremomycin
side chain analogue, LY333328 (later known as oritavancin) an enhanced antimicrobial activity against both vancomycin-
[13, 14]. From 1994 to 2001 Eli Lilly, in a screen to gain sub- susceptible organisms (through increased affinity for the wild-type
stantial anti-VRE activity while maintaining anti-MRSA activity, target), as well as improved activity against vancomycin-resistant
developed oritavancin, but terminated its antibacterial discovery/ organisms [14]. Oritavancin differs from chloroeremomycin by
development program and sold oritavancin to InterMune, Inc, in the addition of a 4#-chlorobiphenylmethyl substituent on the
2002 [14]. InterMune conducted a variety of clinical trials with disaccharide sugar (Figure 1) [13, 14]. This structural alteration
oritavancin but subsequently sold it to Targanta Therapeutics imparts improved activity against vancomycin-susceptible En-
Corporation (now owned by The Medicines Company) in 2006 terococcus (VSE) and significantly improved activity against VRE,
[14]. The Medicines Company is continuing to develop orita- including VanA enterococci [13, 28, 29]. The lipophilic side chain
vancin, including performing additional clinical trials. in oritavancin anchors the compound to the cell membrane
Oritavancin, a vancomycin analogue, is a semisynthetic through hydrophobic interactions, and it stabilizes the oritavancin
lipoglycopeptide with activity against MRSA, VISA, VRSA, dimer (Figure 2) [15, 29–32]. The formation of dimers is achieved
daptomycin-nonsusceptible S. aureus, and VRE (both VanA by interactions involving the disaccharides attached to ring 4 and
and VanB phenotypes) [3, 5, 13–21]. Oritavancin, which has the 4-epi-vancosamine on ring 6, as well as the chlorine on ring 2
novel pharmacokinetics [22, 23], is rapidly bactericidal in [13, 32–35]. The dimers formed improve oritavancin’s ability to
a concentration-dependent manner, and the area under the bind to its target, including D-alanyl-D-lactate (D-Ala-D-Lac)
concentration–time curve to MIC ratio is the pharmacodynamic present in VanA enterococci [29].
parameter that best describes its activity [5, 15, 24, 25]. This agent
is currently in phase III development as a 1200-mg single dose for MECHANISM OF ACTION
the treatment of acute bacterial skin and skin structure infections,
including those due to MRSA [13, 14, 26, 27]. The purpose of Peptidoglycan Synthesis Inhibition
this report is to review the data available on the mechanisms of The mechanism of action common to all members of the gly-
action of oritavancin against Gram-positive pathogens. copeptide class, including vancomycin, is the inhibition of

Figure 1. Chemical structures of vancomycin, chloroeremomycin, and oritavancin.

Oritavancin Mechanism of Action d CID 2012:54 (Suppl 3) d S215


bacterial cell wall synthesis [5, 6]. The cell wall and its major Figure 2). Oritavancin’s hydrophobic side chain allows bacterial
component, peptidoglycan, are responsible for maintaining cell cell membrane anchoring, which results in stabilizing the inter-
shape and structural integrity of the bacterial cell. Peptidoglycan action with lipid II [32]. The self-association into dimers may
is a polymer composed of repeating disaccharide-pentapeptide promote cooperative interactions between dimers and adjacent
units, which are synthesized in the cytoplasm. Transport of the pentapeptides of the peptidoglycan, which results in increased
disaccharide-pentapeptide units across the cell membrane oc- binding avidity [13, 15]. Unlike vancomycin, oritavancin is able to
curs in the form of a complex with a lipid carrier (undecaprenyl bind to depsipeptides, including D-Ala-D-Lac, which facilitates its
pyrophosphate), known as lipid II. Translocation of lipid II inhibition of cell wall synthesis even in organisms that exhibit
across the cell membrane provides the substrate for trans- VanA-type resistance [28, 29]. Oritavancin forms homodimers
glycosylase enzymes to incorporate the disaccharide-pentapeptide prior to binding to D-Ala-D-Ala or D-Ala-D-Lac, which increases
monomer into the newly synthesized or nascent peptidoglycan. its binding affinity for the target site.
Vancomycin and other glycopeptides bind to the carboxyl- Unlike vancomycin, oritavancin has also been reported to

Downloaded from https://academic.oup.com/cid/article-abstract/54/suppl_3/S214/290034 by guest on 16 March 2019


terminal acyl-D-alanyl-D-alanine (acyl-D-Ala-D-Ala) residues of inhibit transpeptidation by binding to a secondary site (mecha-
the pentapeptide moiety of lipid II (mechanism 1 in Figure 2) nism 2 in Figure 2) [28, 33, 34]. Using solid-state nuclear
[13]. This specific binding, which is referred to as the primary magnetic resonance (NMR), the Schaefer group has described
binding site, sterically hinders the transglycosylase enzyme from the molecular interactions of oritavancin with the cell wall and
incorporating the disaccharide-pentapeptide monomer into na- changes in cell wall structure as a result of these interactions
scent peptidoglycan. This inhibition of peptidoglycan synthesis by [28, 33, 34]. These studies performed on either S. aureus or
vancomycin results in a relatively slow (compared with b-lactams) Enterococcus faecium, using metabolic labeling of cell wall
and concentration-independent bactericidal activity [31]. components with amino acids containing NMR-active nuclei,
Like vancomycin, oritavancin has also been reported to inhibit demonstrated that oritavancin is able to interact with newly
transglycosylation by binding to the primary site [28, 32, 33]. The made peptidoglycan components in a more extensive manner
addition of the hydrophobic side chain 4#-chlorobiphenylmethyl than does vancomycin. Specifically, oritavancin was reported
onto the disaccharide sugar confers on oritavancin an increased to bind the peptidic cross-linking portion of the cell wall of
binding affinity to the target molecule lipid II (mechanism 1 in S. aureus (pentaglycine bridge [33, 34]) and of E. faecium (Asx

Figure 2. Proposed model depicting vancomycin and oritavancin mechanisms of action. Comparison of the single mechanism of action of vancomycin
(inhibition of transglycosylation—mechanism 1) to the 3 different mechanisms of action of oritavancin (inhibition of transglycosylation—mechanism 1,
inhibition of transpeptidation—mechanism 2, and disruption of bacterial membrane integrity—mechanism 3, depicting oritavancin dimers).

S216 d CID 2012:54 (Suppl 3) d Zhanel et al


bridge [28]), in addition to binding the D-Ala-D-Ala termini oritavancin in clinical trials for acute bacterial skin and skin
of the stem peptide. Thus, oritavancin may retain microbio- structure infections, that is, single-dose (1200 mg) therapy [27].
logical activity against vancomycin-resistant organisms (such as Belley et al have recently reported that, unlike vancomycin, or-
VRE and VRSA, where the termini are modified to D-Ala-D-Lac) itavancin exhibited concentration-dependent bactericidal activity
by instead binding the peptide bridge [12, 28, 34]. The addition versus stationary phase inocula of MSSA, MRSA, and VRSA
of a hydrophobic side chain 4#-chlorobiphenylmethyl on the [29, 31]. As demonstrated for exponentially growing organisms,
disaccharide sugar thus confers on oritavancin increased binding oritavancin induced membrane depolorization, increased per-
affinity to lipid II, resulting in inhibition of transglycosylation and meability, and resulted in rapid cell death. These investigators
a more pronounced impact on transpeptidation (mechanism 2 reported a correlation between membrane depolarization and cell
in Figure 2), owing to bridge peptide binding. Thus, the si- death [15]. Also, unlike vancomycin, oritavancin sterilized bio-
multaneous accumulation of D-Ala-D-Ala stems and decrease in films of MSSA, MRSA, and VRSA at minimal biofilm eradication
the level of cross-linking is suggestive of an agent acting on both concentrations (MBECs) of between 0.5 and 8 lg/mL [31]. The

Downloaded from https://academic.oup.com/cid/article-abstract/54/suppl_3/S214/290034 by guest on 16 March 2019


transglycosylation and transpeptidation [28, 33]. bactericidal potency of oritavancin was evident as the MBECs
were within 1 dilution of the respective MIC performed with
Cell Membrane Alterations planktonic cells [31]. The authors hypothesized that the hydro-
The addition of a hydrophobic side chain 4#-chlorobiphenylmethyl phobic 4#-chlorobiphenylmethyl side chain of oritavancin dis-
on the disaccharide sugar confers on oritavancin not only in- rupts membrane potential and increases membrane permeability
creased binding affinity, which results in inhibition of trans- in Gram-positive pathogens. It is the concentration-dependent
glycosylation and transpeptidation, but also cell membrane oritavancin-induced cell membrane perturbation and resulting
anchoring and self-association into dimers, which result in permeability that results in cell death.
perturbation of cell membrane integrity in Gram-positive or-
ganisms [13, 29–32, 36]. Studies using the fluorescent probe Ultrastructural Changes
3,3#-dipropylthiadicarbocyanine iodide [DiSC3(5)] as an indi- The ultrastructural effects of oritavancin and vancomycin on
cator of the degree of depolarization of the plasma membrane MRSA and VRE were examined using transmission electron
following exposure to drug have shown that challenge with microcroscopy [29]. Both vancomycin-exposed (exposure at
oritavancin but not vancomycin results in loss of membrane 16 lg/mL for 3 hours) and oritavancin-exposed (exposure at
integrity [15, 31, 32]. The hydrophobic 4#-chlorobiphenylmethyl 1 lg/mL for 10 minutes) MRSA demonstrated cell wall thick-
side chain of oritavancin was implicated in the mechanism, ening and membrane inclusions, whereas only oritavancin-
resulting in concentration-dependent membrane depolarization exposed cells demonstrated deformed septa that were thickened
and increased permeability in various resistance phenotypes of and misshapen with loss of staining intensity of nascent septal
S. aureus and enterococci, which resulted in cell death (VISA, cross walls. In VRE, oritavancin exposure (0.12 lg/mL or
VRSA, VRE) [15]. Oritavancin interacts with the cell membrane, 1 lg/mL for 10 minutes) induced formation of large mem-
facilitating a prime antimicrobial position for peptidoglycan brane inclusions and septal distortions. The authors hypoth-
interactions, disrupting the bacterial membrane potential and esized that oritavancin-induced septal defects may be due to
increasing membrane permeability (mechanism 3 in Figure 2) membrane disruption because of oritavancin’s ability to di-
[15]. Figure 2 compares the single mechanism of action of merize and form cooperative interactions [29]. It is clear that in
vancomycin (inhibition of transglycosylation) to the 3 different both MRSA and VRE, oritavancin demonstrates pronounced
mechanisms of action of oritavancin (inhibition of transgly- effects on cell division that are not observed with vancomycin.
cosylation, inhibition of transpeptidation, and disruption of This is likely an indication that oritavancin is able to target
bacterial membrane integrity). critical yet vulnerable sites to achieve rapid concentration-
Kim et al have recently shown that the hydrophobic 4#- dependent bactericidal activity.
chlorobiphenylmethyl side chain of oritavancin results in in-
teraction with isolated membrane protoplasts and increases MECHANISM OF RESISTANCE
membrane permeability in artificial liposomes composed of bac-
terial membrane phospholipids [32]. It is important to note that, Even though oritavancin demonstrates activity against clinical
although oritavancin’s effects and interactions with the bacterial isolates of VanA, VanB, and VanC enterococci, it has been
cell membrane only occur above a threshold concentration, re- demonstrated in vitro using agar-based methods that moderate
searchers have concluded that its rapid, concentration-dependent decreases in susceptibility level resistance to oritavancin can
bactericidal effects against Gram-positive pathogens result from occur in enterococci isolates exhibiting the VanA or VanB
altering the integrity of the cell membrane [13, 15, 29, 31]. These phenotype [37]. However, resistance in isolates that harbor the
findings also argue in favor of the currently studied dosing of vanB operon was only observed when the operon was altered

Oritavancin Mechanism of Action d CID 2012:54 (Suppl 3) d S217


such that it became inducible by teicoplanin or constitutively aureus in Canada: results of the CANWARD 2007–2009 study. Diag
expressed [37]. Isolates that produce peptidoglycan precursors Microbiol Infect Dis 2011; 69:320–5.
2. Naimi TS, LeDell KH, Como-Sabetti K, et al. Comparison of community-
terminating in D-Lac may exhibit reduced susceptibility to or- and health care-associated methicillin-resistant Staphylococcus aureus in-
itavancin if all precursors terminating in D-Ala are eliminated fection. JAMA 2003; 290:2976–84.
[37]. This has been achieved in vitro by increasing resistance 3. Saravolatz LD, Pawlak J, Johnson JB. In vitro activity of oritavancin
against community-associated methicillin-resistant Staphylococcus aureus
gene expression or reducing D-Ala-D-Ala production [37]. Such (CA-MRSA), vancomycin-intermediate S. aureus (VISA), vancomycin-
changes resulted in a maximum 16-fold increase in oritavancin resistant S. aureus (VRSA) and daptomycin-non-susceptible S. aureus
MIC compared with the parent strain [37]. The expression of (DNSSA). Int J Antimicrob Agents 2010; 36:69–72.
4. Rybak M, Lomaestro B, Rotschafer JC, et al. Therapeutic monitoring of
the vanZ gene of the vanA gene cluster also resulted in a 4-fold
vancomycin in adult patients: a consensus review of the American Society
increase in MIC through an unknown mechanism [37]. of Health-System Pharmacists, the Infectious Diseases Society of America,
Limited data are available regarding the selection of and the Society of Infectious Diseases Pharmacists. Am J Health Syst
oritavancin-resistant mutants in the laboratory. This is likely Pharm 2009; 66:82–98.

Downloaded from https://academic.oup.com/cid/article-abstract/54/suppl_3/S214/290034 by guest on 16 March 2019


5. Zhanel GG, Calic D, Schweizer F, et al. Dalbavancin, oritavancin and
because, in agar assays, oritavancin diffuses slowly and binds to telavancin: a comparative review. Drugs 2010; 70:859–86.
agar, which makes interpretation of experiments to investigate 6. Matzke GR, Zhanel GG, Guay DR. Clinical pharmacokinetics of vanco-
single-step resistance selection difficult [38]. Bozdogan et al mycin. Clin Pharmacokinet 1986; 11:257–82.
7. Wang G, Hindler JF, Ward KW, Bruckner DA. Increased vancomycin
[38], studying a vancomycin-resistant S. aureus that was serially MICs for Staphylococcus aureus clinical isolates from a university
selected using vancomycin, showed a 32- to 64-fold increase in hospital during a 5-year period. J Clin Microbiol 2006; 44:3883–6.
vancomycin MIC, a 16- to 128-fold increase in teicoplanin 8. Sakoulas G, Moise-Broder PA, Schentag J, Forrest A, Moellering RC Jr,
Eliopoulos GM. Relationship of MIC and bactericidal activity to efficacy
MIC, and a 16- to 64-fold increase in dalbavancin MIC, but only
of vancomycin for treatment of methicillin-resistant Staphylococcus aureus
a 2- to 4-fold increase in oritavancin MIC. Thus far, it has not bacteremia. J Clin Microbiol 2004; 42:2398–402.
been possible to select for high-level oritavancin resistance in 9. Moise-Broder PA, Sakoulas G, Eliopoulos GM, Schentag JJ, Forrest A,
the laboratory, and there have been no instances of decreased Moellering RC Jr. Accessory gene regulator group II polymorphism in
methicillin-resistant Staphylococcus aureus is predictive of failure of
oritavancin susceptibility emerging during or after oritavancin vancomycin therapy. Clin Infect Dis 2004; 38:1700–5.
therapy in clinical studies to date [5, 13, 27]. Whether this is due 10. Hidayat LK, Hsu DI, Quist R, Shriner KA, Wong-Beringer A. High-dose
to the fact that oritavancin demonstrates multiple different vancomycin therapy for methicillin-resistant Staphylococcus aureus in-
fections: efficacy and toxicity. Arch Intern Med 2006; 166:2138–44.
mechanisms of action is unknown. 11. Moise PA, Sakoulas G, Forrest A, Schentag JJ. Vancomycin in vitro bac-
tericidal activity and its relationship to efficacy in clearance of methicillin-
CONCLUSIONS resistant Staphylococcus aureus bacteremia. Antimicrob Agents Chemother
2007; 51:2582–6.
12. Arias CA, Contreras GA, Murray BE. Management of multi-drug re-
Oritavancin’s mechanism of action involves at least 3 known
sistant enterococcal infections. Clin Microbiol Infect 2010; 16:555–62.
mechanisms: inhibition of transglycosylation, inhibition of 13. Arhin FF, Belley A, Rafai Far A, Lehoux D, Moeck G, Parr TR. Glyco-
transpeptidation, and cell membrane interaction/disruption. peptides and lipopeptides. Chapter 2011; In press.
Oritavancin’s multiple mechanisms of action confer activity 14. Allen NE. From vancomycin to oritavancin: the discovery and
development of a novel lipoglycopeptide antibiotic. Antinfect Agents
against vancomycin-susceptible and -resistant organisms as Med Chem 2010; 9:23–47.
well as rapid, concentration-dependent killing versus acti- 15. Belley A, McKay GA, Arhin FF, et al. Oritavancin disrupts membrane
vely growing, stationary phase, and biofilm-producing Gram- integrity of Staphylococcus aureus and vancomycin-resistant enterococci
to effect rapid bacterial killing. Antimicrob Agents Chemother 2010;
positive bacteria.
54:5369–71.
16. Arhin FF, Drahi DC, Pillar CM, et al. Comparative in vitro activity
profile of oritavancin against recent gram-positive clinical isolates.
Notes Antimicrob Agents Chemother 2009; 53:4762–71.
17. Arhin FF, Sarmiento I, Belley A, et al. Effect of polysorbate 80 on
Supplement sponsorship. This article was published as part of a sup-
plement entitled ‘‘Oritavancin for the Treatment of Serious Gram-Positive oritavancin binding to plastic surfaces: implications for susceptibility
Infections,’’ sponsored by The Medicines Company. testing. Antimicrob Agents Chemother 2008; 52:1597–603.
Potential conflicts of interest. G. Z. received funding from The 18. Arhin FF, Sarmiento I, Parr TR, Moeck G. Comparative in vitro activity
Medicines Company. All other authors report no potential conflicts. of oritavancin against Staphylococcus aureus strains that are resistant
All authors have submitted the ICMJE Form for Disclosure of Potential intermediate or heteroresistant to vancomycin. J Antimicrob Chemother
Conflicts of Interest. Conflicts that the editors consider relevant to the 2009; 64:868–70.
content of the manuscript have been disclosed in the Acknowledgements 19. Lemaire S, Kosowska-Shick K, Julian K, et al. Activities of anti-
section. staphylococcal antibiotics towards the extracellular and intraphagocytic
forms of Staphylococcus aureus isolates from a patient with persistent
bacteraemia and endocarditis. Clin Microbiol Infect 2008; 14:766–77.
References 20. Zelenitsky SA, Booker B, Laing N, Karlowsky JA, Hoban DJ, Zhanel GG.
Pharmacodynamics of an investigational glycopeptide LY333328 plus
1. Nichol KA, Adam HJ, Hussain Z, et al. Comparison of community- gentamicin against vancomycin sensitive and vancomycin resistant En-
associated and health care-associated methicillin-resistant Staphylococcus terococcus faecium as demonstrated by static time-kill and multiple dose

S218 d CID 2012:54 (Suppl 3) d Zhanel et al


in vitro pharmacodynamic model experiments. Antimicrob Agents 30. Allen NE, Nicas TI. Mechanism of action of oritavancin and related
Chemother 1999; 43:592–7. glycopeptide antibiotics. FEMS Microbiol Rev 2003; 26:511–32.
21. Jones RN, Barrett MS, Erwin ME. In vitro activity of LY333328, 31. Belley A, Neesham-Grenon E, McKay G, et al. Oritavancin kills stationary-
a novel glycopeptides derivative. Antimicrob Agents Chemother phase and biofilm Staphylococcus aureus cells in vitro. Antimicrob Agents
1997; 41:488–93. Chemother 2009; 53:918–25.
22. Rubino CM, Van Wart SA, Bhavnani SM, et al. Oritavancin population 32. Kim SJ, Singh M, Schaefer J. Oritavancin binds to isolated protoplast
pharmacokinetics in healthy subjects and patients with complicated membranes but not intact protoplasts of Staphylococcus aureus. J Mol
skin and skin structure infections or bacteremia. Antimicrob Agents Biol 2009; 391:414–25.
Chemother 2009; 53:4422–8. 33. Kim SJ, Cegelski L, Stueber D, et al. Oritavancin exhibits dual mode of
23. Fetterly GJ, Ong CM, Bhavnani SM, et al. Pharmacokinetics of action to inhibit cell-wall biosynthesis in Staphylococcus aureus. J Mol
oritavancin in plasma and skin blister fluid following administration of Biol 2008; 377:281–93.
a 200-milligram dose for 3 days or a single 800-milligram dose. Anti- 34. Kim SJ, Cegelski L, Preobrazhenskaya M, Schaefer J. Structures of
microb Agents Chemother 2005; 49:148–52. Staphylococcus aureus cell-wall complexes with vancomycin, eremo-
24. McKay GA, Beaulieu S, Arhin FF, et al. Time-kill kinetics of oritavancin mycin, and chloroeremomycin derivatives by 13C{19F} and 15N{19F}
and comparator agents against Staphylococcus aureus, Enterococcus faecalis rotational-echo double resonance. Biochem 2006; 45:5235–50.

Downloaded from https://academic.oup.com/cid/article-abstract/54/suppl_3/S214/290034 by guest on 16 March 2019


and Enterococcus faecium. J Antimicrob Chemother 2009; 63:1191–9. 35. Groves P, Searle MS, Mackay JP, Williams DH. The structure of an
25. Bhavnani SM, Passarell JA, Owen JS, et al. Pharmacokinetic- asymmetric dimer relevant to the mode of action of the glycopeptide
pharmacodynamic relationships describing the efficacy of oritavancin antibiotics. Structure 1994; 2:747–54.
in patients with Staphylococcus aureus bacteremia. Antimicrob Agents 36. Domenech O, Francius G, Tulkens PM, et al. Interactions of
Chemother 2006; 50:994–1000. oritavancin a new lipoglycopeptide derived from vancmomycin with
26. Dunbar LM, Milata J, Fitzpatrick M, et al. Efficacy of oritavancin at phospholipid bilayers: effect on membrane permeability and nanoscale
single or infrequent doses for the treatment of complicated skin and lipid membrane organization. Biochim Biophys Acta 2009; 1788:
skin structure infections. In: 19th European Congress of Clinical 1832–40.
Microbiology and Infectious Disease. Helsinki, Finland: ECCMID, 37. Arthur M, Depardieu F, Reynolds P, et al. Moderate-level resistance
2009. Poster 1849.75. to glycopeptide LY333328 mediated by genes of the vanA and
27. Greenblatt DJ, Zhao Y, Duan SX, et al. Effect of oritavancin on human vanB clusters in enterococci. Antimicrob Agents Chemother 1999; 43:
cytochromes P450 in vitro. In: 49th Interscience Conference on Anti- 1875–80.
microbial Agents and Chemotherapy, San Francisco, CA: ECCMID, 38. Blosser RS, Karlowsky JA, Loutit JS, et al. Evaluation of agar-based
2009. Poster A1–1284. susceptibility testing of oritavancin against gram-positive cocci
28. Patti GJ, Kim SJ, Yu TY, et al. Vancomycin and oritavancin have dif- [abstract C-070]. In: 103rd American Society for Microbiology
ferent modes of action in Enterococcus faecium. J Mol Biol 2009; 392: Meeting. Washington, DC: 2003.
1178–91. 39. Bozdogan B, Ednie L, Credito K, Kosowska K, Appelbaum PC.
29. Belley A, Harris R, Beveridge T, et al. Ultrastructural effects of oritavancin Derivatives of a vancomycin-resistant Staphylococcus aureus strain
on methicillin-resistant Staphylococcus aureus and vancomycin-resistant isolated at Hershey Medical Center. Antimicrob Agents Chemother
Enterococcus. Antimicrob Agents Chemother 2009; 53:800–4. 2004; 48:4762–5.

Oritavancin Mechanism of Action d CID 2012:54 (Suppl 3) d S219

You might also like