Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 May 1.
Published in final edited form as: Expert Opin Pharmacother. 2015 Mar 24;16(7):1091–1098. doi: 10.1517/14656566.2015.1026256

Oritavancin for acute bacterial skin and skin structure infection

Julia A Messina 1,2, Vance G Fowler Jr 1,2, G Ralph Corey 1,2
PMCID: PMC4580226  NIHMSID: NIHMS723681  PMID: 25803197

Abstract

Introduction

Inpatient treatment of acute bacterial skin and skin structure infections (ABSSSI) exerts a significant economic burden on the healthcare system. Oritavancin is a concentration-dependent, rapidly bactericidal agent approved for the treatment of ABSSSI. Its prolonged half-life with one-time intravenous (IV) dosing offers a potential solution to this burden. In addition, oritavancin represents an alternative therapy for Streptococci and multidrug resistant gram-positive bacteria including methicillin-resistant Staphylococcus aureus and vancomycin-resistant Enterococcus. Animal models have also shown promising results with oritavancin for other disease states including those that require long courses of IV therapy.

Areas covered

This review covers oritavancin’s basic chemistry, spectrum of activity, pharmacodynamics/ pharmacokinetics, efficacy in clinical trials, and provides expert opinion on future directions. To compose this review, a search of PubMed was performed, and articles written in the English language were selected based on full text availability.

Expert Opinion

If oritavancin is proven to be a cost-effective strategy for outpatient treatment and prevents complications of prolonged IV therapy, it will be sought as an alternative antibiotic therapy for ABSSSI. In addition, further clinical data demonstrating efficacy in gram-positive infections requiring prolonged therapy such as endocarditis and osteomyelitis could support oritavancin’s success in the current market.

Keywords: acute bacterial skin and skin structure infection, lipoglycopeptide, oritavancin, Staphylococcus aureus

1.0 Introduction

Acute bacterial skin and skin structure infections (ABSSSI) including cellulitis, wound infections, and major cutaneous abscesses, the majority of which are due to gram-positive organisms, exert significant economic pressure on the healthcare system13. An estimated 750,000 patients per year in the United States are admitted to the hospital for ABSSSI incurring an estimated cost of >6 billion dollars4.

Oritavancin is a long acting, concentration-dependent, rapidly bactericidal agent newly approved for use in ABSSSI5. It offers a potential solution to alleviate this economic burden with effective, one-time intravenous dosing over 3 hours, eliminating the need for prolonged intravenous (IV) catheters and therapeutic drug monitoring. In addition, oritavancin dosing does not have to be adjusted based on age, weight, or mild to moderate hepatic or renal dysfunction. This review focuses on the chemistry, spectrum of activity, pharmacodynamics/ pharmacokinetics (PD/PK), safety and efficacy in clinical trials, and future directions in the use of oritavancin.

2.0 Overview of the market

As demonstrated in clinical trials of ABSSSI, the majority of skin and soft tissue infections are due to S. aureus, and empiric therapy is often directed against methicillin-resistant S. aureus (MRSA). Historically the first choice for MRSA infection, vancomycin requires dose-adjustment for renal insufficiency and close therapeutic monitoring to achieve a narrow window of efficacy without inducing toxicity with supratherapeutic serum levels. With the evolution of S. aureus exhibiting reduced6 and complete7 resistance to vancomycin, alternative therapies are increasingly required. Unfortunately, most alternatives to vancomycin also have important limitations. For example, treatment-emergent resistance to daptomycin has occurred in approximately 5% of patients receiving the drug in the registration trial8 and has been described to occur in up to 60% of specific high-risk populations9.

Newer antibiotics for the treatment of ABSSSI with anti-MRSA activity include ceftaroline, tedizolid, telavancin, dalbavancin, and oritavancin. In clinical trials, ceftaroline has demonstrated non-inferiority to vancomycin in treatment of ABSSSI, but its dosing regimen is 2 to 3 times per day10. Tedizolid has superior pharmacokinetics, potentially reduced toxicity compared to linezolid, and activity against S. aureus isolates with the cfr-element (which confers linezolid resistance)11, 12. Compared to linezolid’s twice daily dosing for 7–10 days, tedizolid is taken once daily for 6 days potentially enhancing compliance. Telavancin is a lipoglycopeptide that allows daily dosing but requires monitoring of serum creatinine due to associated nephrotoxicity1315. Dalbavancin is another semi-synthetic lipoglycopeptide that is given as a single 1000mg IV dose initially followed by a single 500mg dose 1 week later, has excellent coverage of MRSA and MSSA, but is inactive against vanA strains of vancomycin-resistant Enterococcus (VRE)16.

3.0 Introduction to the compound

3.1 Chemistry

Oritavancin, a semi-synthetic lipoglycopeptide, was first described in 1996 as a derivative from the natural product, chloroeremomycin, with addition of a 4′-chlorobiphenylmethyl group17. Oritavancin has three mechanisms of action: 1) inhibition of the transglycosylation step of bacterial cell wall biosynthesis by binding the stem peptide of peptidoglycan precursors, 2) inhibition of the transpeptidation, or cross-linking step of cell wall biosynthesis by binding peptide bridging segments of the cell wall, and 3) disruption of bacterial membrane integrity leading to depolarization, increased membrane permeability, and cell death18.

Oritavancin demonstrates excellent in vitro activity (MIC) against S. aureus, notably methicillin-resistant S. aureus, Vancomycin-intermediate S. aureus (VISA), and Vancomycin-resistant S. aureus (VRSA) strains19, 20. Lin, et al. (2014) tested Oritavancin’s in vitro activity against 203 MRSA isolates of community and healthcare-acquired origin. The investigators report Oritavancin MIC50 and MIC90 of 0.12 and 0.25, respectively, versus 169 isolates of community- and hospital-associated MRSA. Oritavancin MIC ranges were 0.12–0.5 µg/mL versus 5 Heterogeneous Vancomycin-intermediate S. aureus (hVISA) strains, 0.5–2 µg/mL versus 5 VISA strains, and 0.12–1 µg/mL versus 5 VRSA strains. Of note, among VISA and VRSA isolates, only three possessed an oritavancin MIC >1 µg/mL.

Oritavancin has also demonstrated activity against coagulase-negative Staphylococci (CoNS), Streptococci, and other gram-positive organisms including vancomycin-resistant Enterococci21, 22 (Table 1).

Table 1.

Oritavancin’s spectrum of activity

Organism Oritavancin MIC50/MIC90 (µg/mL)
Methicillin-resistant Staphylococcus aureus43,# 0.06/0.06
Methicillin-sensitive Staphylococcus aureus43,# 0.03/0.06
Coagulase negative Staphylococci43,# 0.015/0.06
Streptococcus anginosus group43,# ≤0.008/0.015
Streptococcus pyogenes43,# 0.03/0.12
Streptococcus agalactiae43,# 0.03/0.12
Enterococcus faecalis43,# 0.015/0.06
Enterococcus faecium, Vancomycin-Susceptible43,# ≤0.008/≤0.008
Enterococcus faecium, Vancomycin-Resistant43,# 0.06/0.12
Listeria monocytogenes21 ≤0.008/0.015
Corynebacterium species21 ≤0.008/0.06
Micrococcus species21 ≤0.008/≤0.008
#

for United States isolates

3.2 Pharmacodynamics

Oritavancin’s pharmacodynamic (PD) profile was studied in vitro and in vivo models. In animal models, its antimicrobial activity correlated with the ratio of the area under the concentration-time curve to MIC (AUC/MIC)23. Oritavancin’s concentration-dependent killing was determined from in vitro time-kill studies in which increasing concentrations of oritavancin led to progressively greater rate and extent of killing24.

Belley et al. (2013) compared the in vitro PD based on population PD/PK of simulated dosing regimens of a single infusion of oritavancin 1200mg IV to daptomycin and vancomycin and noted that the single dose of oritavancin exerted rapidly bactericidal activity within the first 2–3 hours of infusion against all 3 MRSA isolates tested25. The mean decreases in bacterial viability after oritavancin administration were significantly greater at 2 hours compared to vancomycin for all 3 MRSA strains including an hVISA isolate (oritavancin vs. vancomycin [log CFU/mL ± standard deviation (SD)]: −4.3 ± 0.1 vs. −0.6 ± 0.3 for MRSA NRS123; −3.7 ± 0.4 vs. −0.2 ± 0.1 for MRSA ATCC 33591; −4.0 ± 0.1 vs. −0.1 ± 0.1 for MRSA-hVISA 1561603). Oritavancin administration resulted in significantly greater mean decreases in bacterial viability at 24 hours compared to daptomycin and vancomycin respectively (−3.3 ± 0.7 vs. −4.2 ± 0.0 vs. −3.2 ± 0.3 for MRSA NRS123, −4.1 ± 0.0 vs. −2.2 ± 0.5 vs. −3.2 ± 0.8 for MRSA ATCC 33591, and −4.1 ± 0.1 vs. −3.1 ± 0.6 vs. −2.7 ± 0.2 for MRSA-hVISA 1561603).

3.3 Pharmacokinetics and metabolism

The pharmacokinetics from Phase II and Phase III studies fit into a dose-linear, three-compartment model with two distributional half-life’s (T1/2’s), alpha and beta, of 2.29 and 13.4 hours respectively and a terminal elimination T1/2 (gamma) of 245 hours, which is >10 days2527. The terminal half-life is prolonged due to oritavancin’s increased protein binding, up to 85–90%, its lipophilic side chain, and its volume of distribution approaching 110 L with slow release from tissue absorption sites. Due to the prolonged terminal T1/2, recent clinical trials have utilized a one-time dosing strategy of 1200mg IV28, 29. To support this strategy, in vitro PD/PK modeling demonstrated that oritavancin single 1200mg IV infusion is as effective over 72 hours as daptomycin 6 mg/kg IV daily and vancomycin 1g IV twice daily25.

Oritavancin is not metabolized and is excreted slowly and unchanged in urine (<5%) and feces (<1%) up to 7 days after dosing30. Although no dose adjustment is recommended for patients with renal failure, the pharmacokinetics of oritavancin in severe renal impairment, defined by a creatinine clearance less than 30, or on hemodialysis has not been studied. Clearly further clinical studies are needed before initiating oritavancin therapy in this group of patients.

4.0 Clinical efficacy

4.1 Phase I studies

A Phase I open-label, non-controlled, dose escalation study of oritavancin 1200 mg IV in 11 healthy human subjects was performed to evaluate safety and pharmacokinetics of the drug30. Oritavancin was well tolerated without serious safety events. Of note, 5 subjects did experience transient, asymptomatic elevation in liver transaminases without associated elevation in bilirubin. In 3 subjects, liver transaminase elevation peaked at <1.5 times the upper limit of normal (ULN). In a fourth subject, liver transaminases increased <3 times ULN but subsequently declined. In the fifth subject, liver transaminases peaked at 70 days after study initiation with AST of 444 U/L and ALT of 334 U/L but subsequently declined. Of note, this subject did admit to significant alcohol consumption.

4.2 Phase II studies

SIMPLIFI was a Phase II multi-center, randomized, double blind, parallel active-comparator study testing non-inferiority of single (1200 mg IV) or infrequent doses to daily doses of oritavancin for adults with ABSSSI31. For infrequent dosing, patients received 800mg IV on day 1 with the option of additional 400mg IV on day 5. Primary efficacy was defined as clinical response at days 21 to 29. With a 15% non-inferiority margin, single dosing was non-inferior to infrequent dosing in efficacy and safety in treating ABSSSI.

Oritavancin has also been studied in an international, multicenter clinical trial evaluating the relationship between microbiologic and clinical response and oritavancin exposure in the treatment of S. aureus bacteremia32. Adult patients were randomized to receive either 10–14 days of oritavancin at four different weight-based doses or control (vancomycin or beta-lactam). A total of 86 patients received oritavancin and underwent plasma PK measurements at varying points on day 1 through day 42 ± 7 days. Of these 86 patients, 55 patients were evaluable for microbiologic and clinical responses. Forty-seven patients treated with oritavancin achieved microbiologic success, and 45 patients achieved clinical success. The investigators concluded that there was a positive exposure-response relationship in the treatment of S. aureus bacteremia with oritavancin based on PK-PD modeling. However, since this study was conducted, it was determined oritavancin in vitro potency was being underestimated by approximately 16- to 32-fold in the absence of polysorbate-80 in the MIC assay as discussed in the “Regulatory affairs” section below. Therefore, time above the MIC (T>MIC) is not a PK/PD parameter that is presently associated with oritavancin efficacy.

4.3 Phase III studies

SOLO I and SOLO II were large Phase III international, multi-center, randomized, double-blind clinical trials comparing a single dose of oritavancin 1200mg IV to twice daily vancomycin for ABSSSI28, 29. The primary composite endpoint was cessation of spreading or reduction of lesion size measured at 48–72 hours, the absence of fever, and no need for rescue antibiotics. The secondary endpoints were investigator-assessed clinical cure within 7–14 days after treatment cessation and ≥20% reduction in lesion size 48–72 hours after initiation of antibiotic therapy. Patients were followed for 60 days to evaluate safety and tolerability of oritavancin’s extended terminal T1/2. Both trials demonstrated that single dosing of oritavancin was non-inferior (margin of 10%) to vancomycin in a modified-intent-to treat analysis of efficacy.

5.0 Safety and tolerability

To date, over one thousand patients have been enrolled into clinical trials testing safety and efficacy of oritavancin for the treatment of patients with ABSSSI with minimal serious safety reports or trial withdrawal due to intolerability of the drug. In SOLO I, at least one treatment-emergent adverse event (TEAE) occurred in 108 (22.8%) patients in the oritavancin arm compared to 151 (31.4%) in the vancomycin arm, and at least one TEAE leading to discontinuation of study drug in 18 (3.8%) patients in the oritavancin arm versus 28 (5.8%) in the vancomycin arm. Serious TEAE leading to discontinuation of study drug occurred in 3 (0.6%) patients in the oritavancin arm and 3 (0.6%) in the vancomycin arm. In SOLO II, study drug-related TEAE occurred in 109 (21.7%) of patients in the oritavancin arm vs. 128 (25.5%) in the vancomycin arm. TEAE leading to study drug discontinuation occurred in 18 (3.6%) patients in the oritavancin group and 13 (2.6%) in the vancomycin group. Importantly, there was not a notable difference between time of onset and duration of adverse events in patients treated with oritavancin compared to vancomycin in either of the Phase III trials28, 29.

The most commonly reported side effects observed in Phase III SOLO trials were headache, nausea, vomiting, limb and subcutaneous abscess, and diarrhea33. Transient ALT elevations were also observed, but no cases that met Hy’s law criteria (>3-fold elevation of AST or ALT above the ULN and total bilirubin >2-fold above the ULN) were identified. Infusion reactions occurred and included pruritus, urticaria, and flushing but resolved with slowing rate of infusion and were less frequent than with the comparator, vancomycin. Hypersensitivity reactions occurred with median onset of 1.2 days and median duration of reaction of 2.4 days. Rare episodes of leukocytoclastic vasculitis, erythema multiforme, and angioedema were also reported at rates ≤1.5%. Oritavancin carries a pregnancy category C with safety only demonstrated in animal models. Similarly, safety of oritavancin in nursing mothers has not yet been established.

Although oritavancin does not have direct effect on the coagulation cascade, it has weak activity on the cytochrome P450 system. Thus, oritavancin co-administration with warfarin may result in higher exposure to warfarin and increased risk of bleeding. In addition, oritavancin artificially prolongs PT and PTT tests for up to 48 hours by binding to and preventing action of the phospholipid reagents which activate coagulation in commonly used laboratory coagulation tests and INR tests for up to 24 hours" after "PT" since oritavancin's in vitro effect on PT and INR is not longer than 24 hours. Therefore, use of unfractionated heparin is contra-indicated within 48 hours of oritavancin infusion.

6.0 Regulatory affairs

Oritavancin’s development was slow to progress due to succession of ownership among pharmaceutical companies and an initial overestimation of S. aureus and enterococci MICs. Eli Lilly synthesized the drug in 1994 with the designation LY333328, but the compound was acquired by Intermune Inc. in 2002, followed by Targanta Therapeutics Corporation in 2006, and finally by the Medicines Companies in 200917, 23. Initially, oritavancin’s MICs to methicillin-susceptible S. aureus (MSSA), MRSA, and Enterococci were substantially over-estimated as investigators did not account for oritavancin’s non-specific binding losses through sticking to the walls of the test tubes during MIC assays. However, in 2008, work by Arhin et al. confirmed a 16 to 32-fold overestimation of MIC against staphylococci and enterococci34.

The first FDA drug application for oritavancin, submitted in 2008, was unsuccessful leading to the request for another Phase III trial and further evaluation of the effects of the drug on macrophage function35. Concerns about the potential of oritavancin decreasing effectiveness of macrophage function arose from the observation of its marked accumulation in lysosomes of macrophages. Similar to excessive intracellular cholesterol and phospholipids, oritavancin accumulation in macrophages resulted in deposition of concentric lamellar structures, finely granular material, or less-defined osmiophilic material36. The appearance mimicked morphologic abnormalities noted in mixed-lipid storage disorders, invoking concern that these cellular alterations within macrophages reduced phagocytic function, potentially increasing risk for infection particularly due to fungi and mycobacteria. A study by Baquir et al. (2012) addressed these concerns by measuring killing function by 2 lines of macrophages against S. aureus, Candida albicans, and Acinetobacter baumannii when exposed to therapeutic dosing of oritavancin37. The investigators found that both lines of macrophages still killed pathogens after exposure to oritavancin but with the following differences in effect: S. aureus killing was enhanced by oritavancin, C. albicans killing was unchanged, and A. baumannii killing was unchanged among one line of macrophages and moderately reduced among the other line. Based on these results, the investigators concluded that oritavancin accumulation within macrophages does not significantly alter macrophage killing of key pathogens such as S. aureus.

In August of 2014 after completion of SOLO I and SOLOII, the FDA approved oritavancin for the treatment of ABSSSI due to gram-positive organisms in adults ≥18 years of age.

7.0 Conclusion

Oritavancin is a safe and effective drug for the treatment ABSSSI. It also has demonstrated bactericidal activity against multidrug resistant gram-positive bacteria such as VRE, MRSA, VISA, hVISA, and VRSA. The one-time intravenous dosing is an appealing solution to the economic burden that hospitalization and outpatient IV therapy for ABSSSI exerts upon the healthcare system. However, further assessment of cost and providers’ willingness to prescribe oritavancin over other antibiotics currently available is needed.

8.0 Expert opinion

Oritavancin offers advantages over current IV antibiotic therapy for the treatment of ABSSSI caused by Staphylococci and Streptococci. Its prolonged half-life allows treatment with a single infusion and does not require therapeutic drug monitoring. In comparison to current antibiotics approved for the treatment of ABSSSI, it is the only agent with a one-time dosing regimen. This dosing strategy is attractive, potentially avoiding hospitalization for IV antibiotics and the need for home health or skilled nursing facility care for outpatient IV antibiotic administration. One-time IV dosing may also improve therapeutic compliance compared to oral step-down therapy. Dalbavancin is in the beginning of a Phase 4 trial comparing one time versus the current two time dosing strategy for the treatment of ABSSSI. If the one time dosing strategy of dalbavancin is successful, the drug will compete strongly against oritavancin.

In addition to treatment of ABSSSI, oritavancin holds promise in treating other infectious diseases such as bacteremia, endocarditis, and C. difficile colitis based upon efficacy in a Phase II clinical trial in treatment of S. aureus bacteremia and animal models of infective endocarditis23, 32. In a rabbit model of MRSA endocarditis, oritavancin is as effective as vancomycin in clearance of bacteremia and reduction in bacterial counts in vegetations and tissues38. In a rat model of MSSA endocarditis, oritavancin, vancomycin, oxacillin, and daptomycin therapy significantly decreased S. aureus densities in vegetations by bioluminescent assay39. Oritavancin has also performed well in in animal models of endocarditis due to E. faecalis, maintaining activity against vancomycin-susceptible and vanA and vanB phenotypes40. Similarly, oritavancin in combination with gentamicin proved synergistic against all three strains of E. faecalis in the treatment of left-sided endocarditis without the emergence of mutants resistant to either component of therapy41. Further studies are needed to study oritavancin’s effectiveness in the treatment of E. faecalis and E. faecium infection in humans.

Drug-resistant S. aureus infections will continue to pose significant treatment challenges in the years to come42. Current data on efficacy in animal models and initial clinical trials should inspire further study of oritavancin in treatment of conditions that require prolonged IV therapy such as bacteremia, endocarditis, and meningitis. Further investigation into osteomyelitis and septic arthritis as potential future therapeutic targets for oritavancin is also warranted.

Whether or not physicians would be willing to prescribe oritavancin depends on whether one-time infusion will prevent hospitalization for ABSSSI and complications from prolonged IV therapy such as catheter-associated bloodstream infections, venous thrombosis, and thrombophlebitis. Ideally, patients could receive their first dose of oritavancin in the outpatient setting such as an emergency department or outpatient clinic without necessitating hospital admission, PICC line placement, and arrangements for home infusion services. Conversely, patients with more complicated ABSSSI that require admission for sepsis or surgical drainage could benefit from oritavancin’s dosing strategy with shorter hospital stays and potentially no need for home infusion services or oral step-down therapy.

In addition, further data are needed to determine whether oritavancin’s one time IV dosing strategy is cost effective. Costs of antibiotics for ABSSSI vary significantly (Table 2). However, the costs do not take into account costs of home IV infusion services nor can they account for potential non-compliance with oral therapy, relapse of infection, and hospital readmission. In 5 years’ time, if use of oritavancin is proven to be cost-effective in shortening duration or preventing hospitalization and preventing complications of IV therapy, it will be sought as an effective alternative to current antibiotics for the treatment of ABSSSI.

Table 2.

Prices of Commonly Used Anti-Staphylococcal Drugs for ABSSSI1

Antibiotic Price per dose Price per Treatment Course
Ceftaroline $151.62a $2122.68b
Dalbavancin $1788c $5364d
Daptomycin $455.04c $3185.28e
Linezolid N/A $3644.29f
Oritavancin $1160g $3480h
Tedizolid N/A $2124.00i
Telavancin $389.93j $2729.51e
Vancomycin $7.46k $104.44b
1

. Average wholesale price in U.S. Data obtained from Lexicomp Online®, UpToDate®, Hudson, Ohio: Lexi-Comp, Inc.; February 20, 2015.

a

. Same price for 400mg and 600mg reconstituted solution

b

. 7 day treatment course (twice daily)

c

. 500mg reconstituted solution

d

. One time dosing 1000mg followed by 500mg one week later

e

. 7 day treatment course (once daily)

f

. 10 day treatment course (twenty 600mg oral tablets)

g

. 400mg reconstituted solution

h

. 1200mg dose once

i

. 6 day treatment course (six 200mg oral tablets)

j

. 750mg reconstituted solution

k

. 1000mg reconstituted solution

Drug summary box.

Drug Name Oritavancin
Phase Completed Phase III trials
Indication Acute bacterial skin and skin structure infections
Pharmacology description/ mechanism of action Semi-synthetic lipoglycolipopeptide, bactericidal, disrupts cell membrane and inhibits the transglycosylation and transpeptidation steps of cell wall synthesis in a dose-dependent manner
Route of administration Intravenous
Chemical Structure graphic file with name nihms723681t1.jpg
Pivotal Trial(s) SIMPLIFI31
SOLO I29
SOLO II28

Acknowledgments

Dr. Messina, Dr. Fowler and Dr. Corey received funding support from the National Institute Of Allergy And Infectious Diseases of the National Institutes of Health under Award Number UM1AI104681. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. Dr. Fowler was also supported by K24-AI093969. Vance G. Fowler, Jr has received grant/ research Support from Advanced Liquid Logic, Cubist, Cerexa, MedImmune, Merck, NIH, Novartis, Pfizer, Theravance. Has acted as a paid consultant for Affinium, Baxter, Cerexa, Cubist, Debiopharm, Durata, Merck, Novartis, NovaDigm, The Medicines Company, MedImmune, Pfizer, Theravance, Trius. Has received honoraria from Arpida, Astellas, Cubist, Inhibitex, Merck, Pfizer, Targanta, Theravance, Wyeth, Ortho-McNeil, Novartis, Vertex Pharmaceuticals and has Membership at Merck Co-Chair V710 Vaccine. G. Ralph Corey reports acting on scientific advisory board for Nabriva, Metronic (and study design group), Cubist, TMC, Theravance, Cempra, Cerexa/Forest/Actavis, Trius/Merk, Achaogen, GlaxoSmithKline. He also reports involvement with Dr Reddy’s lab on adjudication committee, Merk Mortality board, he has also acted as consultant and scientific advisory board for Melinta and Motif, and the Pfizer Mortality Board.

Abbreviations and units

ABSSSI

Acute bacterial skin and skin structure infection

ALT

Alanine aminotransferase

AUC

Area under concentration-time curve

CFU

Colony-forming units

CoNS

Coagulase negative Staphylococci

g

Gram

T1/2

Half-life

hVISA

Heterogeneous vancomycin-intermediate Staphylococcus aureus

IV

Intravenous

L

Liters

MRSA

Methicillin-resistant Staphylococcus aureus

µg/mL

Microgram per milliliter

mg/kg

Milligram per kilogram

mg/L

Milligram per liter

MIC

Minimum inhibitory concentration

PD

Pharmacodynamic

SD

Standard deviation

ULN

Upper limit of normal

TEAE

Treatment-emergent adverse event

T>MIC

Time above MIC

VISA

Vancomycin-intermediate Staphylococcus aureus

VRSA

Vancomycin-resistant Staphylococcus aureus

VRE

Vancomycin-resistant Enterococcus

Footnotes

Financial and competing interests dislcosure

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed

References

  • 1.Suaya JA, Mera RM, Cassidy A, et al. Incidence and cost of hospitalizations associated with Staphylococcus aureus skin and soft tissue infections in the United States from 2001 through 2009. BMC infectious diseases. 2014;14:296. doi: 10.1186/1471-2334-14-296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Lee BY, Singh A, David MZ, et al. The economic burden of community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) Clinical microbiology and infection : the official publication of the European Society of Clinical Microbiology and Infectious Diseases. 2013 Jun;19(6):528–536. doi: 10.1111/j.1469-0691.2012.03914.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Garau J, Ostermann H, Medina J, et al. Current management of patients hospitalized with complicated skin and soft tissue infections across Europe (2010–2011): assessment of clinical practice patterns and real-life effectiveness of antibiotics from the REACH study. Clinical microbiology and infection : the official publication of the European Society of Clinical Microbiology and Infectious Diseases. 2013 Sep;19(9):E377–E385. doi: 10.1111/1469-0691.12235. [DOI] [PubMed] [Google Scholar]
  • 4.LaPensee KT, Fan W, Wang Y. PIN22 Economic Burden of Hospitalization With Antibiotic Treatment for Absssi in the United States: An Analysis Ofthe Premier Hospital Database. Value in Health. 2012;15(4):A240–A241. [Google Scholar]
  • 5.Tice A. Oritavancin: a new opportunity for outpatient therapy of serious infections. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2012 Apr;54(Suppl 3):S239–S243. doi: 10.1093/cid/cir1047. [DOI] [PubMed] [Google Scholar]
  • 6.Fridkin SK, Hageman J, McDougal LK, et al. Epidemiological and microbiological characterization of infections caused by Staphylococcus aureus with reduced susceptibility to vancomycin, United States, 1997–2001. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2003 Feb 15;36(4):429–439. doi: 10.1086/346207. [DOI] [PubMed] [Google Scholar]
  • 7.Hanaki H, Kuwahara-Arai K, Boyle-Vavra S, et al. Activated cell-wall synthesis is associated with vancomycin resistance in methicillin-resistant Staphylococcus aureus clinical strains Mu3 and Mu50. The Journal of antimicrobial chemotherapy. 1998 Aug;42(2):199–209. doi: 10.1093/jac/42.2.199. [DOI] [PubMed] [Google Scholar]
  • 8.Fowler VG, Jr, Boucher HW, Corey GR, et al. Daptomycin versus standard therapy for bacteremia and endocarditis caused by Staphylococcus aureus. The New England journal of medicine. 2006 Aug 17;355(7):653–665. doi: 10.1056/NEJMoa053783. [DOI] [PubMed] [Google Scholar]
  • 9.Sharma M, Riederer K, Chase P, Khatib R. High rate of decreasing daptomycin susceptibility during the treatment of persistent Staphylococcus aureus bacteremia. European journal of clinical microbiology & infectious diseases : official publication of the European Society of Clinical Microbiology. 2008 Jun;27(6):433–437. doi: 10.1007/s10096-007-0455-5. [DOI] [PubMed] [Google Scholar]
  • 10.File TM, Jr, Wilcox MH, Stein GE. Summary of ceftaroline fosamil clinical trial studies and clinical safety. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2012 Sep;55(Suppl 3):S173–S180. doi: 10.1093/cid/cis559. [DOI] [PubMed] [Google Scholar]
  • 11.Chambers HF. Pharmacology and the treatment of complicated skin and skin-structure infections. The New England journal of medicine. 2014 Jun 5;370(23):2238–2239. doi: 10.1056/NEJMe1405078. [DOI] [PubMed] [Google Scholar]
  • 12.Shorr AF, Lodise TP, Corey GR, et al. Analysis of the Phase 3 ESTABLISH Trials: Tedizolid Versus Linezolid in Acute Bacterial Skin and Skin Structure Infection. Antimicrobial agents and chemotherapy. 2014 Nov 24; doi: 10.1128/AAC.03688-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Stryjewski ME, Lentnek A, O'Riordan W, et al. A randomized Phase 2 trial of telavancin versus standard therapy in patients with uncomplicated Staphylococcus aureus bacteremia: the ASSURE study. BMC infectious diseases. 2014;14:289. doi: 10.1186/1471-2334-14-289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Rubinstein E, Lalani T, Corey GR, et al. Telavancin versus vancomycin for hospital-acquired pneumonia due to gram-positive pathogens. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2011 Jan 1;52(1):31–40. doi: 10.1093/cid/ciq031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Stryjewski ME, Graham DR, Wilson SE, et al. Telavancin versus vancomycin for the treatment of complicated skin and skin-structure infections caused by gram-positive organisms. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2008 Jun 1;46(11):1683–1693. doi: 10.1086/587896. [DOI] [PubMed] [Google Scholar]
  • 16.Boucher HW, Wilcox M, Talbot GH, et al. Once-weekly dalbavancin versus daily conventional therapy for skin infection. The New England journal of medicine. 2014 Jun 5;370(23):2169–2179. doi: 10.1056/NEJMoa1310480. [DOI] [PubMed] [Google Scholar]
  • 17.Schwalbe RS, McIntosh AC, Qaiyumi S, et al. In vitro activity of LY333328, an investigational glycopeptide antibiotic, against enterococci and staphylococci. Antimicrobial agents and chemotherapy. 1996 Oct;40(10):2416–2419. doi: 10.1128/aac.40.10.2416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Zhanel GG, Schweizer F, Karlowsky JA. Oritavancin: mechanism of action. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2012 Apr;54(Suppl 3):S214–S219. doi: 10.1093/cid/cir920. [DOI] [PubMed] [Google Scholar]
  • 19.Lin G, Pankuch G, Appelbaum PC, Kosowska-Shick K. Antistaphylococcal activity of oritavancin and its synergistic effect in combination with other antimicrobial agents. Antimicrobial agents and chemotherapy. 2014 Oct;58(10):6251–6254. doi: 10.1128/AAC.02932-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Saravolatz LD, Pawlak J, Johnson LB. In vitro activity of oritavancin against community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA), vancomycin-intermediate S. aureus (VISA), vancomycin-resistant S. aureus (VRSA) and daptomycin-non-susceptible S. aureus (DNSSA) International journal of antimicrobial agents. 2010 Jul;36(1):69–72. doi: 10.1016/j.ijantimicag.2010.02.023. [DOI] [PubMed] [Google Scholar]
  • 21.Mendes RE, Sader HS, Flamm RK, Jones RN. Activity of oritavancin tested against uncommonly isolated Gram-positive pathogens responsible for documented infections in hospitals worldwide. The Journal of antimicrobial chemotherapy. 2014 Jun;69(6):1579–1581. doi: 10.1093/jac/dku016. [DOI] [PubMed] [Google Scholar]
  • 22.Morrissey I, Seifert H, Canton R, et al. Activity of oritavancin against methicillin-resistant staphylococci, vancomycin-resistant enterococci and beta-haemolytic streptococci collected from western European countries in 2011. The Journal of antimicrobial chemotherapy. 2013 Jan;68(1):164–167. doi: 10.1093/jac/dks344. [DOI] [PubMed] [Google Scholar]
  • 23.Ambrose PG, Drusano GL, Craig WA. In vivo activity of oritavancin in animal infection models and rationale for a new dosing regimen in humans. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2012 Apr;54(Suppl 3):S220–S228. doi: 10.1093/cid/cis001. [DOI] [PubMed] [Google Scholar]
  • 24.McKay GA, Beaulieu S, Arhin FF, et al. Time-kill kinetics of oritavancin and comparator agents against Staphylococcus aureus, Enterococcus faecalis and Enterococcus faecium. The Journal of antimicrobial chemotherapy. 2009 Jun;63(6):1191–1199. doi: 10.1093/jac/dkp126. [DOI] [PubMed] [Google Scholar]
  • 25. Belley A, Arhin FF, Sarmiento I, et al. Pharmacodynamics of a simulated single 1,200-milligram dose of oritavancin in an in vitro pharmacokinetic/pharmacodynamic model of methicillin-resistant staphylococcus aureus infection. Antimicrobial agents and chemotherapy. 2013 Jan;57(1):205–211. doi: 10.1128/AAC.01428-12. *This study defined important PK/PD parameters through modeling that justified the once weekly dosing strategy for the clinical trails.
  • 26. Rubino CM, Van Wart SA, Bhavnani SM, et al. Oritavancin population pharmacokinetics in healthy subjects and patients with complicated skin and skin structure infections or bacteremia. Antimicrobial agents and chemotherapy. 2009 Oct;53(10):4422–4428. doi: 10.1128/AAC.00231-09. **The investigators created population pharmacokinetic model from data from phase 1, 2, and 3 clinical trial participants.
  • 27.Rubino CM, Bhavnani SM, Moeck G, et al. Population Pharmacokinetic (PPK) Analysis for Oritavancin (ORI) Based on Data from Patients with Acute Bacterial Skin and Skin Structure Infections (ABSSSI) 2014 [abstract]. [Google Scholar]
  • 28. Corey GR, Good S, Jiang H, et al. Single-dose Oritavancin Compared to 7–10 days of Vancomycin in the Treatment of Gram-Positive Acute Bacterial Skin and Skin Structure Infections; the SOLO II Non-inferiority Study. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2015;60(2):254–262. doi: 10.1093/cid/ciu778. ***This is a Phase 3 pivital clinical trial confirming results of SOLO I and contributing further pateint safety data that led to oritavancin's FDA approval for the treatment of ABSSSI.
  • 29. Corey GR, Kabler H, Mehra P, et al. Single-dose oritavancin in the treatment of acute bacterial skin infections. The New England journal of medicine. 2014 Jun 5;370(23):2180–2190. doi: 10.1056/NEJMoa1310422. ***This is a pivital Phase 3 clinical trial demonstrating that oritavancin is non-inferior to vancomycin in the treatment of ABSSSI.
  • 30.Bhavnani SM, Owen JS, Loutit JS, et al. Pharmacokinetics, safety, and tolerability of ascending single intravenous doses of oritavancin administered to healthy human subjects. Diagnostic microbiology and infectious disease. 2004 Oct;50(2):95–102. doi: 10.1016/j.diagmicrobio.2004.06.007. [DOI] [PubMed] [Google Scholar]
  • 31.Dunbar LM, Milata J, McClure T, et al. Comparison of the efficacy and safety of oritavancin front-loaded dosing regimens to daily dosing: an analysis of the SIMPLIFI trial. Antimicrobial agents and chemotherapy. 2011 Jul;55(7):3476–3484. doi: 10.1128/AAC.00029-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Bhavnani SM, Passarell JA, Owen JS, et al. Pharmacokinetic-pharmacodynamic relationships describing the efficacy of oritavancin in patients with Staphylococcus aureus bacteremia. Antimicrobial agents and chemotherapy. 2006 Mar;50(3):994–1000. doi: 10.1128/AAC.50.3.994-1000.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.ORBACTIV™ (Oritavancin) [prescribing information] Parsippany, NJ: 2014. The Medicines Company. [Google Scholar]
  • 34.Arhin FF, Sarmiento I, Belley A, et al. Effect of polysorbate 80 on oritavancin binding to plastic surfaces: implications for susceptibility testing. Antimicrobial agents and chemotherapy. 2008 May;52(5):1597–1603. doi: 10.1128/AAC.01513-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Van Bambeke F. Renaissance of antibiotics against difficult infections: Focus on oritavancin and new ketolides and quinolones. Annals of medicine. 2014 Jul 24;:1–18. doi: 10.3109/07853890.2014.935470. [DOI] [PubMed] [Google Scholar]
  • 36.Van Bambeke F, Saffran J, Mingeot-Leclercq MP, Tulkens PM. Mixed-lipid storage disorder induced in macrophages and fibroblasts by oritavancin (LY333328), a new glycopeptide antibiotic with exceptional cellular accumulation. Antimicrobial agents and chemotherapy. 2005 May;49(5):1695–1700. doi: 10.1128/AAC.49.5.1695-1700.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Baquir B, Lemaire S, Van Bambeke F, et al. Macrophage killing of bacterial and fungal pathogens is not inhibited by intense intracellular accumulation of the lipoglycopeptide antibiotic oritavancin. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2012 Apr;54(Suppl 3):S229–S232. doi: 10.1093/cid/cir921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kaatz GW, Seo SM, Aeschlimann JR, et al. Efficacy of LY333328 against experimental methicillin-resistant Staphylococcus aureus endocarditis. Antimicrobial agents and chemotherapy. 1998 Apr;42(4):981–983. doi: 10.1128/aac.42.4.981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Xiong YQ, Yin L, Hady WA, et al. Efficacy of oritavancin (ORI), a lipoglycopeptide antibiotic, in a rat Staphylococcal aureus endocarditis (IE) model: microbiological and bioluminescent assessments; 48th Interscience Conference on Antimicrobial Agents and Chemotherapy/Infectious Diseases Society of America 46th Annual Meeting; Washington, DC. 2008. [Google Scholar]
  • 40.Saleh-Mghir A, Lefort A, Petegnief Y, et al. Activity and diffusion of LY333328 in experimental endocarditis due to vancomycin-resistant Enterococcus faecalis. Antimicrobial agents and chemotherapy. 1999 Jan;43(1):115–120. doi: 10.1128/aac.43.1.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Lefort A, Saleh-Mghir A, Garry L, et al. Activity of LY333328 combined with gentamicin in vitro and in rabbit experimental endocarditis due to vancomycin-susceptible or -resistant Enterococcus faecalis. Antimicrobial agents and chemotherapy. 2000 Nov;44(11):3017–3021. doi: 10.1128/aac.44.11.3017-3021.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Rodvold KA, McConeghy KW. Methicillin-resistant Staphylococcus aureus therapy: past, present, and future. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2014 Jan;58(Suppl 1):S20–S27. doi: 10.1093/cid/cit614. [DOI] [PubMed] [Google Scholar]
  • 43.Mendes RE, et al. Activity of oritavancin against Gram-positive clinical isolates responsible for documented skin and soft-tissue infections in European and US hospitals (2010–13) Journal of Antimicrobial Chemotherapy 70.2. 2015:498–504. doi: 10.1093/jac/dku421. [DOI] [PubMed] [Google Scholar]

RESOURCES