You are on page 1of 14

STAT E O F T H E A RT R E V I E W

Advances in the medical management of


skin and soft tissue infections
Sarah L McClain,1 Jefferson G Bohan,1 Dennis L Stevens1 2
1
Boise Veterans Affairs Medical
Center, 500 W Fort St, Boise, ID A B S T RAC T
83702, USA
2
Skin and soft tissue infections are some of the most common infectious disease
University of Washington School of
Medicine, Seattle, WA 98195, USA diagnoses in both inpatient and outpatient settings. With bacterial resistance
Correspondence to: S L McClain
sarah.mcclain@va.gov
to antimicrobials growing, decision making on empiric antibiotics is becoming
Cite this as: BMJ 2016;355:i6004 increasingly difficult. Additionally, the most recent guidance from a professional
doi: 10.1136/bmj.i6004
society on the treatment of skin and soft tissue infections was published in 2014
by the Infectious Diseases Society of America and is now two years old. New
antimicrobial agents have been developed and approved for the treatment of skin
and soft tissue infections since then, and more are in the pipeline. This review
summarizes the evidence on treatments that are new or in development and the
potential repurposing of old antimicrobials. The clinical utility of these treatments is
also discussed.

Introduction This review discusses the evidence surrounding new


The management of skin and soft tissue infections (SSTIs) treatments, potential repurposing of old antimicrobials,
in ambulatory and inpatient settings is challenging. The and treatments that may become available in the future. It
incidence is twice as high as that of pneumonia and primarily focuses on the treatment of SSTIs in adults, but
urinary tract infections combined.1 Also, antimicrobial data about new antimicrobials in children are presented
resistance to available treatments remains a problem, where available.
particularly with resistant staphylococci.
In 2010 the US Food and Drug Administration (FDA) Incidence and prevalence
published guidance changing the terminology for more The incidence of SSTIs in ambulatory and inpatient
severe SSTIs from complicated skin and skin structure settings in the United States has been relatively stable
infections (cSSSIs) to acute bacterial skin and skin struc- from 2005 to 2010, at 47.9 to 48.5 cases per 1000 per-
ture infections (ABSSSIs). The 2010 draft guidance was son years.1 Of those SSTI cases, 95% were treated in the
finalized in 2013 and served to update the FDA’s 1998 ambulatory care setting and 60% were diagnosed as
document that provided clinical trial guidance for the abscesses or cellulitis. Streptococci and staphylococci are
drug industry.2 cSSSIs were defined as infections that the primary SSTI pathogens.5 Community associated met-
involved deeper soft tissue, required significant surgi- icillin resistant Staphylococcus aureus (MRSA, USA300
cal intervention, or occurred in patients with significant strain) is of particular importance in the US, given the
underlying disease complicating the response to treat- high rates of purulent SSTIs treated in the community.
ment.2 ABSSSI is defined as cellulitis/erysipelas, wound SSTIs accounted for 62.9% of infections with MRSA strain
infection, and major cutaneous abscess, with a minimum USA300 from 2000 to 2013, increasing the need for addi-
lesion size of 75 cm2.3 Milder SSTIs such as impetigo and tional treatment options.6 The prevalence of MRSA dif-
minor cutaneous abscesses are excluded from the ABSSSI fers between the US and the rest of the world. In areas of
designation. Alternatively, the 2014 guideline on SSTI lower MRSA prevalence, such as the United Kingdom and
from the Infectious Diseases Society of America (IDSA) northern Europe, treatment guidelines may vary consid-
differentiates SSTIs by the presence of purulence and erably from the IDSA guidance, with more emphasis on
severity of illness rather than using ABSSSI designation.4 β lactams effective against meticillin susceptible Staphy-
Antimicrobial resistance makes selecting empiric anti- lococcus aureus (MSSA).7
microbial treatment difficult; choosing definitive therapy
is nearly impossible, especially if a microbiological diag- Diagnostic considerations
nosis is absent. Recently, several antimicrobials have Epidemiology plays an important role when diagnos-
been approved to treat SSTIs, with more in the pipeline. ing SSTIs with potentially diverse etiologies.4 Obtaining
A limitation of using these new drugs is the lack of guide- careful, complete, and accurate medical histories may
line consensus about their clinical utility. Because of these provide insight into clinical and microbiological diag-
concerns, evidence based decision making about empiric nosis and treatment. Specifically, patients’ immune sta-
and definitive antimicrobial therapy is im­perative. tus, geographical locale, travel history, recent trauma or

For personal use only 1 of 14


STAT E O F T H E A RT R E V I E W

Terms used in search strategy therapy can be considered for nasal decolonization in
patients with recurrent SSTIs or for treatment of mild,
Drug related terms
uncomplicated SSTIs, such as bullous or non-bullous
Free text terms—“tedizolid”, “brilacidin”, “oral sodium
impetigo. 4 Despite incision and drainage or topical
fusidate”, “sodium fusidate”, “delafloxacin”
therapy, some SSTIs will need systemic antimicrobial
MESH terms—“anti-infective agents”, “fosfomycin”,
“fusidic acid”
tr­eatment.
The IDSA treatment algorithm (figure) incorporates
MESH supplementary concepts—“dalbavancin”,
“oritavancin”, “iclaprim”, “ABT 492”, “omadacycline”, the presence or absence of purulence, along with clini-
“torezolid phosphate” cal status, when providing treatment recommenda-
Indication related terms tions. According to this algorithm, moderate or severe
Free text terms—“SSTI”, “skin and soft tissue infection”, purulent infections require empiric oral or intravenous
“acute bacterial skin and skin structure infection”, antistaphylococcal antimicrobial therapy, respectively,
“ABSSSI”, “major cutaneous abscess” along with incision and drainage plus culture and sensi-
MESH terms—“soft tissue infections”, “skin diseases, tivity.2 Non-purulent infections may be treated with oral
infectious,” “cellulitis,” “paronychia”, “skin diseases, or intravenous antibiotics targeted against streptococci
bacterial”, “wound infection”, “impetigo”, “cervicofacial for mild and moderate infections, respectively. Severe
actinomycosis”, “bacillary angiomatosis”, “ecthyma”, non-purulent infections including necrotizing fasciitis
“erysipelas”, “erythema chronicum migrans”, may require broad spectrum antibiotics, toxin inhibition,
“erythrasma”, “granuloma inguinale”, “pinta”,
and surgical intervention.2
“rhinoscleroma”, “staphylococcal scalded skin
syndrome”, “furunculosis”, “carbuncle”, “cutaneous
syphilis”, “erythema induratum”, “lupus vulgaris”, “yaws” New antimicrobial agents
Dalbavancin
surgery, previous antimicrobial therapy, and exposure Properties and activity
to animals are crucial to limit the differential diagnosis.4 Dalbavancin, a semi-synthetic lipoglycopeptide, binds
Recognition of clinical findings and understanding ana- the terminal D-alanyl-D-alanine residues, thus inhibit-
tomical relations of skin and soft tissue are also essential ing the transpeptidation and transglycosylation steps of
for establishing the correct diagnosis. In patients who are peptidoglycan synthesis, in the bacterial cell wall.8 In
immunocompromised or have evidence of systemic signs addition, dalbavancin’s structure features a lipophilic
of infection such as tachycardia, hypotension, or organ side chain allowing it to anchor to the bacterial cell
dysfunction, more invasive techniques including tissue membrane, thereby increasing potency and improving
biopsy or aspiration need to be used.4 Radiographic evi- half life.
dence may help to determine the level of infection and Dalbavancin has in vitro activity against Gram positive
presence of gas or abscess. Lastly, surgical debridement organisms, including Staphylococcus aureus (MSSA and
or exploration may be diagnostically and therapeutically MRSA), coagulase negative staphylococci (CoNS), Strepto-
important, especially in immunocompromised patients or coccus spp, and Enterococcus spp.9 Dalbavancin also has
those with necrotizing infections.4 More definitive diag- in vitro activity against rarer Gram positive pathogens,
nostic information can be found in the full IDSA SSTI including Listeria monocytogenes, Micrococcus spp, and
guideline. Corynebacterium spp.10 Dalbavancin has shown eightfold
and 16-fold greater in vitro activity against S aureus than
Methods daptomycin and vancomycin, respectively.11 Vancomycin
We did a structured PubMed search to identify primary lit- susceptibility can be used as a surrogate for dalbavancin
erature published from 1 January 2006 through 1 March susceptibility with 97.7-100% accuracy.12 13 Furthermore,
2016 using indication related and drug related free text dalbavancin retains in vitro activity against pathogens
terms, MESH terms, and MESH supplementary concepts with reduced susceptibility to vancomycin, such as van-
(box). We used filters to select clinical trials, prospective comycin intermediate S aureus (VISA), although most
and retrospective cohort studies, guidelines, and review minimum inhibitory concentrations (MICs) fall above
articles. We screened 2400 articles and applied exclusion the FDA’s breakpoint of 0.12 μg/mL.14
criteria based on title, then abstract, then full text. We Staphylococcal resistance to dalbavancin has been
also screened reference lists of pertinent articles. Topics reported in less than 1% of isolates and occurs similarly
of interest included pharmacology, pharmacokinetics, to vancomycin resistance despite higher potency, through
pharmacodynamics, dosing, adverse effects, FDA/Euro- cell wall thickening and increased D-alanine-D-alanine
pean Medicines Agency (EMA) approved indications, binding sites.11 14 Lastly, dalbavancin has in vitro activity
and outcomes related to drugs approved since 2014 or against enterococci carrying VanB but not VanA, making
in phase II or III clinical development. We also searched it ineffective against most vancomycin resistant Entero-
clinical trials databases. coccus spp (VRE) in the US.14
Dalbavancin was approved for ABSSSI by the FDA in
Treatment overview 2014 and EMA in 2015. The dosing was initially 1000
SSTI treatment is generally divided into two different mg followed by an additional 500 mg seven days later.
groups: purulent infections (for example, furuncles, In 2016 a single dose regimen of 1500 mg was approved
carbuncles, abscesses) and non-purulent infections (for on the basis of results of a randomized trial showing non-
example, cellulitis, erysipelas). Topical antimicrobial inferiority to the two dose regimen.9 15

For personal use only 2 of 14


STAT E O F T H E A RT R E V I E W

0$1$*(0(172)667,V

1213858/(17 3858/(17
1HFURWL]LQJLQIHFWLRQFHOOXOLWLVHU\VLSHODV )XUXQFOHFDUEXQFOHDEVFHVV

6(9(5( 02'(5$7( 0,/' 6(9(5( 02'(5$7( 0,/'

(0(5*(17685*,&$/ ,175$9(12867+(5$3< 25$/7+(5$3< , ' , ' , '


,163(&7,21'(%5,'(0(17 3HQLFLOOLQRU 3HQLFLOOLQ9.RU & 6 & 6 & 6
5XOHRXWQHFURWL]LQJSURFHVV &HIWULD[RQHRU &HSKDORVSRULQRU
(03,5,&7+(5$3< &HID]ROLQRU 'LFOR[DFLOOLQRU
9DQFRP\FLQ3/86 &OLQGDP\FLQ &OLQGDP\FLQ (03,5,&7+(5$3< 
(03,5,&7+(5$3<
SLSHUDFLOOLQWD]REDFWDP 9DQFRP\FLQRU
'DSWRP\FLQRU 70360;RU
/LQH]ROLGRU 'R[\F\FOLQH
7HOHYDQFLQRU
& 6 &HIWDUROLQH

'(),1('7+(5$3< QHFURWL]LQJLQIHFWLRQV
0RQRPLFURELDO '(),1('7+(5$3< '(),1('7+(5$3<
6WUHSWRFRFFXVS\RJHQHVSHQLFLOOLQ3/86FOLQGDP\FLQ 056$ 056$
&ORVWULGLDOVSSHQLFLOOLQ3/86FOLQGDP\FLQ 6HH(PSLULF 70360;
$VGDSWRP\FLQDQGWHOHYDQFLQDUHQRW
9LEULRYXOQLƟFXVGR[\F\FOLQH3/86FHIWD]LGLPH 066$ 066$
DSSURYHGIRUXVHLQFKLOGUHQYDQFRP\FLQLV
$HURPRQDVK\GURSKLOLDGR[\F\FOLQH3/86FLSURƠR[DFLQ UHFRPPHQGHGFOLQGDP\FLQPD\EHXVHGLI 1DIFLOOLQRU 'LFOR[DFLOOLQRU
3RO\PLFURELDO FOLQGDP\FLQUHVLVWDQFHLVDWWKH &HID]ROLQRU &HSKDOH[LQ
LQVWLWXWLRQ &OLQGDP\FLQ
9DQFRP\FLQ3/86SLSHUDFLOOLQWD]REDFWDP

Purulent skin and soft tissue infections (SSTIs)—Mild infection: for purulent SSTI, incision and drainage is indicated. Moderate infection: patients with purulent
infection with systemic signs of infection. Severe infection: patients who have not responded to incision and drainage plus oral antibiotics, those with systemic
signs of infection such as temperature >38°C, tachycardia (heart rate >90 beats per minute), tachypnea (respiratory rate >24 breaths per minute), or abnormal white
blood cell count (>12 000 or <4000 cells/µL), or immunocompromised patients. Non-purulent SSTIs—Mild infection: typical cellulitis/erysipelas with no focus of
purulence. Moderate infection: typical cellulitis/erysipelas with systemic signs of infection. Severe infection: patients who have not responded to oral antibiotic
treatment, those with systemic signs of infection (as defined above under purulent infection), those who are immunocompromised, or those with clinical signs
of deeper infection such as bullae, skin sloughing, hypotension, or evidence of organ dysfunction. C&S=culture and sensitivity; I&D=incision and drainage;
MRSA=meticillin resistant Staphylococcus aureus; MSSA=meticillin susceptible Staphylococcus aureus; TMP/SMX=trimethoprim-sulfamethoxazole. First
appeared in “Practice guidelines for the diagnosis and management of skin and soft tissue infections: 2014 update” by the Infectious Diseases Society of America;
adapted with permission from Oxford University Press

Pharmacokinetics and dosing by intermittent hemodialysis and with low permeability


Dalbavancin has linear, dose dependent pharmacokinet- continuous hemodialyzers; however, dalbavancin clear-
ics. It widely distributes into tissues, with extensive and ance matched or exceeded renal clearance in patients
reversible protein binding of approximately 93%, and with normal creatinine clearance using high permeability
achieves therapeutic concentrations in blister fluid for continuous dialyzers.17 No dosage adjustment is needed
at least seven days.9 Dalbavancin does not interact with in patients with hepatic impairment owing to little effect
the cytochrome P450 system and has no important drug on dalbavancin’s metabolism and clearance.16 Dalba-
interactions. Approximately one third of dalbavancin is vancin does not prolong the QTc interval and was shown
excreted unchanged in urine, with another 20% being to have no effect on heart rate or PR or QRS intervals.18
excreted in feces up to 70 days after the initial dose. The
terminal half life of dalbavancin is 346 hours, allowing Evidence summary
for one or two doses to provide an entire course of ther- The first phase III, randomized, double blind study of
apy. No dosage adjustments are needed for patients with dalbavancin for SSTIs was published in 2005 and com-
normal renal function (creatinine clearance ≥30 mL/min) pared two doses of dalbavancin against 14 days of lin-
or those receiving hemodialysis.9 ezolid treatment. Dalbavancin showed non-inferiority to
Interestingly, patients with creatinine clearance below linezolid with a lower frequency of adverse effects.19 This
30 mL/min but not receiving hemodialysis have a higher study was followed by two identical randomized, double
area under of the curve for dalbavancin and reduced blind, double dummy, multicenter phase III trials, DIS-
clearance, so dosage adjustment is needed to 1125 mg COVER 1 and 2 (table 1). Two doses of dalbavancin were
for a single dose or 750 mg followed by 375 mg for the compared with at least three days of intravenous vanco-
two dose series.16 Dalbavancin is not significantly cleared mycin, followed by an optional switch to oral linezolid

For personal use only 3 of 14


STAT E O F T H E A RT R E V I E W

Table 1 | Phase III clinical evidence summary for dalbavancin, oritavancin, and tedizolid in acute bacterial skin and skin structure infections
Patient Secondary Statistical
Trial Design Intervention population Demographics Primary endpoint* endpoints* analyses Results
Dalbavancin
Randomized, Randomized, Dalbavancin 1000 mg Adults with ITT (n=854): mean Clinical success (no Microbiologic Non-inferiority, Primary endpoint:
double blind double blind, double IV on day 1, 500 mg cSSSIs† and at age 47; males further antibiotics response and margin 12.5% 88.9% response to
comparison of once dummy, multicenter; on day 8 v linezolid least one sign 62%; abscess 32%; needed) at TOC overall response at dalbavancin v 91.2%
weekly dalbavancin conducted in 2003- 600 mg every 12 h for of systemic cellulitis 28%. mITT visit 14±2 days EOT and TOC response to linezolid,
versus twice daily 04 14 days infection (64%, n=550): S after completion lower limit of
linezolid therapy for aureus 89%; MRSA CI=−7.28%; met non-
cSSSIs19 51%; GAS 5.6% inferiority criteria
DISCOVER-1 and Randomized, Dalbavancin 1000 mg Adults with ITT (n=1312): mean Early clinical Clinical status Non-inferiority, Pooled analysis of
DISCOVER-220 double blind, double IV on day 1, 500 mg on ABSSSIs‡ and at age 49.5; males response at 48-72 at EOT; clinical margin 10% primary endpoints for
dummy, multicenter; day 8 v IV vancomycin least one sign of 58%; white 89.3%; hours (cessation response at EOT both trials: 79.7% for
conducted in 2011- for ≥3 days followed systemic illness abscess 25.5%; of spread of dalbavancin v 79.8%
12 by optional switch to cellulitis 53.5%; erythema, no for vancomycin-
oral linezolid for 10-14 pathogen isolated increase in size, linezolid, 95% CI
days at baseline 51%: S temperature −4.5 to 4.2); met
aureus 77%; MRSA ≤37.6°C) non-inferiority criteria
24%; GAS 11%
Oritavancin
SOLO-121 Randomized, Oritavancin 1200 Adults with mITT (n=954): mean Composite of Investigator Non-inferiority, Primary efficacy
double blind, double mg IV on day 1 or ABSSSIs and age 45; males 63%; cessation of assessed clinical margin 10% endpoint at early
dummy multicenter; vancomycin 1000 mg signs of systemic white 58%; abscess spreading or cure at PTE; clinical evaluation:
conducted in 2011- or 15 mg/kg IV every inflammation 29%;cellulitis 50%; reduction in lesion decrease in lesion 82.3% for oritavancin
12 12 h for 7-10 days positive infection size, absence size by ≥20% v 78.9% for
site culture 61%: S of fever, and no vancomycin, 95% CI
aureus 45% use of rescue −1.6 to 8.4; met non-
antibiotics at ECE inferiority criteria
SOLO-222 Randomized, Oritavancin 1200 Adults with mITT (n=1005): Composite of Investigator Non-inferiority, Primary efficacy
double blind, double mg IV on day 1 or ABSSSIs and mean age 45; males cessation of assessed clinical margin 10% outcome at ECE
dummy, multicenter; vancomycin 1000 mg signs of systemic 68%; white 71%; spreading or cure at PTE; (mITT): 80.1% for
conducted in 2011- or 15 mg/kg IV every inflammation abscess 32.5%; reduction in lesion decrease in lesion oritavancin v 82.9%
13 12 h for 7-10 days cellulitis 30.9%; size, absence of size by ≥20% for vancomycin, 95%
positive infection fever and no use of CI −7.5 to 2.0; met
site culture 70%: S rescue antibiotics non-inferiority criteria
aureus 74%; MRSA at ECE
29%
Tedizolid
ESTABLISH-1 and Randomized, Tedizolid 200 mg daily ESTABLISH-1: ITT (n=1333): Early clinical Investigator Non-inferiority, Pooled analysis for
ESTABLISH-223 double blind, double for 6 days v linezolid adults with median age 44; response at 48-72 assessed response margin 10% primary efficacy
dummy, multicenter; 600 mg twice daily for ABSSSIs; males 63%; abscess hours*§ at day 7, EOT, and endpoint in both
ESTABLISH-1 10 days; ESTABLISH-1 ESTABLISH-2: 25%; cellulitis 46%; PTE trials§: 81.6% for
conducted in 2010- patients received oral patients ≥12 baseline cultures: S tedizolid v 79.4%
11; ESTABLISH-2 drugs; ESTABLISH-2 years with aureus 50%; MRSA for linezolid, 95% CI
conducted in 2011- patients received ≥2 ABSSSIs 21.5%; GAS 4% −2.0 to 6.5; met non-
13 IV doses inferiority criteria
GAS=group A streptococci; MRSA=meticillin resistant Staphylococcus aureus; TOC=test of cure; EOT=end of treatment; ITT=intention to treat; ECE=early clinical evaluation; PTE=post-therapy evaluation.
*US FDA issued draft guidance for industry for ABSSSI drug development in 2010 and final guidance in 2013. In final document, primary efficacy endpoint became lesion response at 48-72 hours, defined as ≥20%
size reduction from baseline in patients who did not receive rescue therapy and are alive. Resolution of ABSSSI 7-14 days after completion of therapy is suggested as secondary endpoint.3
†Complicated skin and soft tissue infections (cSSSIs) defined as infection that involved deeper soft tissue or required significant surgical intervention (major abscesses, burns, traumatic or surgical wound infection,
deep SSTIs such as extensive/ulcerating cellulitis).16
‡Acute bacterial skin and skin structure infection required the presence of cellulitis, major abscess, or wound infection, each with ≥75 cm2 of erythema.
§ESTABLISH-1 definition for primary endpoint differed slightly owing to draft industry guidance available from FDA at time study was designed. Pooled data published in 2015 updated endpoint to reflect 2013 final
guidance document.

for a total of 10-14 days, in 1312 patients with ABSSSIs.20 ard adult dosing (1000 mg) for children weighing at least
Similarly, dalbavancin showed non-inferiority for the pri- 60 kg and a dose of 15 mg/kg for those weighing under 60
mary endpoint of early clinical response, defined as ces- kg.24 Dalbavancin was well tolerated and pharmacokinet-
sation of the spread of erythema and absence of fever at ics were similar among children receiving each dosing
48-72 hours. Adverse events occurred less frequently in strategy, although the values were slightly lower than
the dalbavancin arm (overall P=0.5), with nausea, head- those measured in adults.
ache, and diarrhea being most common. Median dura-
tion of adverse effects was three days for dalbavancin Clinical application
compared with four days for the comparator.9 20 Notably, Dalbavancin has shown safety and efficacy for treating
patients in this study had more severe disease than did ABSSSIs, and its place in therapy is similar to that of other
those included in previous studies of dalbavancin, as glycopeptides. The biggest advantage of dalbavancin is
DISCOVER enrollment limited patients with abscess to no convenience; however, economic considerations must be
more than 30% and approximately 50% of patients had weighed carefully against other treatment options (table
two or more systemic inflammatory response syndrome 2). The single dose, short infusion time, lack of need for
criteria at enrollment.20 routine monitoring, and absence of drug-drug or drug-
Limited data in children are available for dalbavancin. laboratory interactions are desirable characteristics.
A phase I study in patients aged 12-17 years used stand- Dalbavancin may help in avoiding hospital admissions

For personal use only 4 of 14


STAT E O F T H E A RT R E V I E W

Table 2 | Economic impact of antimicrobial agents used to treat skin and soft tissue infections ceptibility to vancomycin, MIC values often exceed the
Cost per day Cost (AWP*) based on standard laboratory breakpoints, warranting cautious use
(AWP*) based on standard dosing regimen in clinical settings.26
Medication Dosing regimen dosing regimen and duration indicated
Newer agents Pharmacokinetics and dosing
Ceftaroline 600 mg IV twice daily $366.24 7 days: $2563.68 Oritavancin was approved for ABSSSIs by the FDA in
Dalbavacin† 1500 mg IV once daily $383.14 One dose: $5363.96
2014 and the EMA in 2015. Oritavancin is administered
Daptomycin‡ 500 mg IV once daily $534.59 7 days: $3742.13
intravenously as a single 1200 mg dose. Oritavancin has
Oritavancin† 1200 mg IV once daily $248.57 One dose: $3479.98
linear pharmacokinetics, and it extensively distributes
Tedizolid 200 mg IV once daily $296.10 6 days:$1776.60
into tissues.32 Plasma protein binding of oritavancin is
200 mg PO once daily $371.70 6 days: $2230.10
Telavancin‡ 750 mg IV once daily $428.92 7 days: $3002.44
approximately 85%.32
Older agents Oritavancin clearance primarily occurs through the
Cefazolin 2 g IV every 8 h $198.00 7 days: $1386.00 reticuloendothelial system, and excretion from this sys-
Clindamycin 900 mg IV every 8 h $62.64 7 days: $438.48 tem occurs slowly, with less than 1% recovered in the feces
300 mg PO every 8 h $7.14 7 days: $49.98 and 5% in urine after two weeks of collection.26 32 The half
Doxycycline 100 mg PO twice daily $12.30 7 days: $86.10 life of oritavancin is approximately 245 hours, allowing a
Linezolid 600 mg IV twice daily $192.00 10 days: $1920.00 single dose to provide an entire course of therapy.33 No dos-
600 mg PO twice daily $368.00 10 days: $3680.00 age adjustments are needed for mild to moderate renal or
Trimethoprim/sulfamethoxazole 160 mg/800 mg PO twice daily $2.30 7 days: $16.10 hepatic impairment. Oritavancin has not been adequately
Vancomycin 1000 mg IV every 12 h $30.96 7 days: $216.72 evaluated in severe hepatic or renal impairment, but it is
IV=intravenously; PO=orally.
not significantly cleared by low or high flux hemodialysis
*Average wholesale price as of 6 March 2016, US dollars ($1=£0.79; €0.94).
†One dose was considered to provide two weeks of therapy for purpose of cost per day calculations. or continuous renal replacement therapy.32 34 Oritavancin
‡Dosing rounded to nearest vial size. has not been associated with QTc prolongation.35
Oritavancin is a non-specific, weak inhibitor of CYP2CP
solely for administration of intravenous antibiotic. Dalba- and CYP2C19 and an inducer of CYP3A4 and CYP2D6,
vancin may also have a niche in treating patients who find leading to drug interactions, most notably with warfarin
adherence difficult, those who live remotely and would (31% increase in mean area under the curve).32 Owing
have difficultly being monitored on outpatient parenteral to its ability to bind the phospholipid reagent, orita-
antimicrobial therapy, and those with a history of inject- vancin also has some important drug-laboratory interac-
ing drug misuse to avoid central line placement. tions with commonly used coagulation tests, including
activated partial thromboplastin time (aPTT), activated
Oritavancin clotting time, and prothrombin time/international nor-
Properties and activity malized ratio.32 As a result of these interactions, the use
Oritavancin is a semisynthetic lipoglycopeptide with at of unfractionated heparin is contraindicated for 120
least three mechanisms of action (MOAs). Similarly to hours after administration of oritavancin owing to the
other glycopeptides, oritavancin inhibits transglycosyla- potential for false elevation of the aPTT. For patients who
tion, a step of cell wall synthesis, by binding the terminal need aPTT monitoring within 120 hours of oritavancin
D-alanyl-D-alanine.25 It also binds peptide bridging seg- administration, a non-phospholipid dependent coagula-
ments to inhibit transpeptidation, which also contributes tion test, such as the factor Xa assay, is recommended.32
to its activity against vancomycin resistant organisms.
Oritavancin is thought to inhibit transpeptidation more Evidence summary
than transglycosylation.26 Lastly, oritavancin interacts The major clinical trials for oritavancin, SOLO I and II,
with the cell membrane, causing depolarization and were both randomized, double blind studies of a single
permeabilization, ultimately leading to rapid, concen- dose of intravenous oritavancin 1200 mg versus vanco-
tration dependent bactericidal activity.25 Activity on mycin 1000 mg or 15 mg/kg intravenously twice daily for
the cell membrane has been attributed to oritavancin’s seven to 10 days in nearly 2000 adults with ABSSSI.21 22
activity against stationary phase bacteria, such as those The primary endpoint was a composite of cessation of
in biofilms.27 spreading infection or reduction in lesion size, absence
Oritavancin has in vitro activity against many Gram of fever, and no use of a rescue antibiotic 48-72 hours
positive organisms, including S aureus (MSSA, MRSA, after oritavancin administration. Secondary endpoints
VISA, vancomycin resistant S aureus (VRSA)), CoNS, were investigator determined clinical cure seven to 14
streptococci, and enterococci, including VRE.28 Addi- days after end of treatment and lesion size reduction of
tionally, oritavancin has shown in vitro activity against L at least 20% 48-72 hours post-oritavancin. Oritavancin
monocytogenes, Micrococcus spp, and Corynebacterium showed non-inferiority to vancomycin in both studies for
spp.29 Vancomycin sensitivities can be used as a surro- all efficacy endpoints. The most common adverse reac-
gate for oritavancin sensitivities, with 99.4% cross sus- tions occurring in more than 3% of patients were nausea,
ceptibility; however, vancomycin non-susceptibility to headache, vomiting, diarrhea, and limb and subcutane-
enterococci is not an accurate predictor of oritavancin ous abscesses. Oritavancin was well tolerated, with sim-
non-susceptibility.30 31 Oritavancin non-susceptibility is ilar rates of adverse reactions to vancomycin. In SOLO
thought to be exceedingly rare.31 Although oritavancin I, nausea was (non-significantly) more common in the
has in vitro activity against S aureus with reduced sus- oritavancin group.21

For personal use only 5 of 14


STAT E O F T H E A RT R E V I E W

A pediatric phase I study is recruiting patients under Tedizolid has been approved by the EMA and FDA for
18 with suspected or confirmed bacterial infections to the treatment of ABSSSIs including cellulitis and skin
assess the pharmacokinetics of oritavancin. The starting abscesses.45 The dose of tedizolid is 200 mg, adminis-
dose of oritavancin for this study seems to be 15 mg/kg.36 tered orally or intravenously, once daily, with no dose
adjustment needed for patients with renal or hepatic
Clinical application impairment.48 49
Oritavancin’s place in therapy for ABSSSIs is similar to
that of other glycopeptides (table 1). Oritavancin’s con- Evidence summary
venience is a shared feature with dalbavancin, which ESTABLISH-1 and 2 were phase III clinical trials that eval-
must be carefully weighed with economic and patient uated tedizolid compared with linezolid. ESTABLISH-1
specific factors when considering all treatment options randomized 667 patients with cellulitis, major cutaneous
for SSTIs (table 2). The use of oritavancin could improve abscesses, or wound infection to either tedizolid 200 mg
and simplify patient care scenarios in which older treat- by mouth daily for six days or linezolid 600 mg by mouth
ment options are less desirable owing to toxicity or mon- twice daily for 10 days. The primary outcome was early
itoring requirements. Oritavancin also provides more clinical response at 48-72 hours. With a 10% margin, tedi-
robust activity against resistant Gram positive organisms. zolid was non-inferior to linezolid. Early and sustained
The three hour infusion time and large volume of dextrose clinical response rates were similar between groups.50
it must be prepared in have been criticized; however, a ESTABLISH-2 randomized 666 patients with similar
new formulation with a shorter infusion time is being infections to either tedizolid 200 mg intravenously daily
investigated.37 Drug-drug and drug-laboratory interac- for six days or linezolid 600 mg intravenously twice daily
tions with anticoagulants are also problematic in some for 10 days, with the option for oral step-down therapy
patients, but a study of oritavancin in patients on chronic after at least two intravenous doses in both groups. The
warfarin treatment is under way.38 primary outcome studied was early clinical response
48-72 hours after starting treatment. With a 10% margin,
Tedizolid tedizolid was non-inferior to linezolid. Clinical response
Properties and activity rates were similar at all time points.51 Notably, clinically
Tedizolid, an oxazolidinone prodrug, is metabolized by significant adverse effects were significantly decreased
endogenous phosphatases from tedizolid phosphate to with tedizolid compared with linezolid, including nausea
tedizolid, the active metabolite. The MOA of oxazolidi- (P=0.02) and thrombocytopenia (P<0.001).23 One pre-
none involves binding the 23S ribosomal RNA of the 50S clinical study suggests that prolonged therapy beyond
subunit, thus preventing formation of the 70S initiation six days of tedizolid may also have advantages of less
complex and causing inhibition of protein synthesis.39 myelosuppression and neuropathy than linezolid.52
Tedizolid’s spectrum of activity is similar to that of lin-
ezolid, with bacteriostatic in vitro killing and suggested Clinical application
bactericidal in vivo killing due to granulocyte augmen- Tedizolid’s place in therapy is similar to that of linezolid
tation in some animal models of Gram positive organ- owing to its microbiologic spectrum and MOA; however,
isms including MRSA and VRE.40 41 Tedizolid has greater tedizolid provides some advantages over linezolid, such
potency than linezolid, with lower MIC50 and MIC90 values as a more convenient dosing regimen and fewer side
against organisms including staphylococci, streptococci, effects and drug interactions, specifically with serotonin
and enterococci, which may be due to greater ribosomal modulating agents.53 Owing to the MOA, tedizolid may
binding affinity.39 Despite tedizolid’s increased potency, also be beneficial in treating necrotizing fasciitis because
linezolid resistance does predict tedizolid resistance.42 of its ability to suppress toxin production. These charac-
However, tedizolid has maintained activity against some teristics could be appealing for clinicians when treating
staphylococci and enterococci expressing the chloram- patients with multiple comorbidities, but use would need
phenicol resistance gene (cfr), suggesting that tedizolid to be balanced against the pharmacoeconomic advan-
may be used to treat infections caused by linezolid resist- tages of linezolid (table 2). Future studies are planned
ant organisms in addition to vancomycin and daptomycin in diabetic patients with wound infections and pediatric
resistant organisms.43 44 patients with ABSSSI.54 55

Pharmacokinetics and dosing Retapamulin


Tedizolid has greater than 90% oral bioavailability and Properties and activity
linear, two compartment pharmacokinetics indicating Retapamulin, a topical pleuromutilin antibiotic, exerts
central and peripheral compartment absorption.45 46 Tedi- action by binding to an allosteric site of the 50S ribosomal
zolid is a reversible inhibitor of monoamine oxidase in subunit, thus altering the conformational shape of the
vitro; however, in contrast to linezolid, preclinical studies peptidyl transferase center and inhibiting protein syn-
suggest a lack of clinically significant drug interactions thesis. The barrier to resistance is thought to be higher
with serotonin modulating agents.39 The terminal half life owing to the allosteric binding.56 Retapamulin has activ-
is longer and the volume of distribution larger than lin- ity against both staphylococci and streptococci, including
ezolid’s, and excretion of tedizolid is primarily fecal with MRSA strains. Additional data show activity against Pro-
small fractions excreted renally.46 Similar pharmacokinet- pionibacterium, Fusobacterium, and other Gram positive
ics have been observed in adolescents.47 cocci.57 58

For personal use only 6 of 14


STAT E O F T H E A RT R E V I E W

One study showed that retapamulin remained active minimal systemic exposure of the topical formulation.
against S aureus isolates resistant to meticillin, fusidic It may also be useful in treating wound infections but
acid, or mupirocin with 99.9% inhibition.59 However, needs to be studied further for this indication. Retapa-
in 2014 retapamulin resistance increased to 9.5% for mulin is being studied for use in reducing MRSA nasal
S aureus, of which more than 50% of the isolates were carriage.67
also meticillin resistant and less than 1% were linezolid
resistant, suggesting that retapamulin resistance may Phase III agents
be increasing slowly in both MSSA and MRSA.60 Resist- Ozenoxacin
ance to retapamulin primarily occurs as a result of muta- Pharmacology
tions in the rplC gene and needs three different stepwise Ozenoxacin represents a new generation of non-fluori-
mutations to reduce susceptibility significantly.61 Slower nated, quinolone antibiotics and exerts its MOA by inhib-
growth rates have been noted in resistant strains, which iting DNA gyrase and topoisomerase IV, affecting DNA
led to the emergence of more susceptible, faster growing replication, transcription, repair, and recombination.68
strains.62 Ozenoxacin has greater bactericidal activity against
Gram positive organisms including MRSA, CoNS, and
Pharmacokinetics and dosing streptococcal species than do other fluoroquinolone anti-
Retapamulin is a topical antibiotic with minimal absorp- biotics.69 Resistance to ozenoxacin occurs as a result of
tion. In studies, measurable blood concentrations of reta- mutations in the grlA and gyrA genes coding topoisomer-
pamulin were found in only 3-11% of patients using the ase IV and DNA gyrase, respectively. Interestingly, muta-
recommended dose.63 tions to grlA did not confer resistance whereas grlA-gyrA
Retapamulin is approved by the FDA for treatment of double mutants and grlA-gyrA triple mutants conferred
impetigo and by the EMA for treatment of impetigo and a lower level of resistance to S aureus than with other
staphylococcal skin infections.63 It is dosed by applying a fluoroquinolones.68
thin layer topically to the affected area twice daily for five In a phase I open label study involving 46 patients
days. Owing to limited systemic exposure, retapamulin (adult and pediatric), systemic bioavailability was unde-
does not need dose adjustment in patients with renal or tectable in all but four samples, indicating minimal sys-
hepatic impairment. temic exposure to topical ozenoxacin.70 Another phase I
trial studied the skin tissue exposure of topical ozenoxa-
Evidence summary cin 2% dosed either once or twice daily. Results indicated
A randomized, double blind, phase III study conducted that the drug does not penetrate well into the dermis,
in 17 international centers randomized 139 patients to steady state conditions are reached at day three, and
receive retapamulin and 71 patients to receive placebo skin concentrations correlate linearly with total amount
topically twice daily for five days to treat impetigo. The of drug administered.71
primary endpoint was end of therapy response rate
measured at day seven. Success rates at day 7 differed Evidence summary
significantly at 85.6% for retapamulin and 52.1% for Japan has approved a 2% ozenoxacin topical preparation
placebo (difference 33.5%, 95% confidence interval for use in superficial skin infections, and other countries
20.5% to 46.5%). The most common adverse effect including the US and Europe are still evaluating a 1%
related to retapamulin compared with placebo was appli- preparation.72 A phase III clinical trial randomized 465
cation site pruritis (6.5% v 1.4%).64 patients aged at least 2 years with impetigo to ozenoxa-
A phase III non-inferiority study randomized 519 cin 1% cream, placebo, or retapamulin ointment applied
patients with impetigo to retapamulin or fusidic acid. twice daily for five days. The primary endpoint was clini-
With a 10% margin, retapamulin was non-inferior to cal success or failure at the end of therapy. In the inten-
fusidic acid for the primary endpoint of clinical success tion to treat population, ozenoxacin was associated with
at end of therapy in both the intention to treat and per significantly higher clinical success rates than placebo at
protocol populations.65 Both clinical trials enrolled adult 34.8% compared with 19.2% (P=0.003); retapamulin’s
and pediatric (age ≥9 months) patients. clinical success was similar to ozenoxacin’s (37.7%).73
In a murine wound infection model, topical retapa- Ozenoxacin has been tolerated well in studies, with mini-
mulin was compared with oral linezolid with a primary mal side effects related to the drug.
endpoint of bacterial load reduction. Retapamulin
reduced bacterial loads more significantly than did oral Clinical application
linezolid.66 Ozenoxacin’s place in therapy is likely to be similar
to that of other topical agents including retapamulin.
Clinical application An advantage of ozenoxacin is the new generation of
Retapamulin should primarily be used to treat mild quinolone antibiotics it represents, to which bacteria
to moderate impetigo with minimal systemic signs or likely have not yet developed resistance. Also, its MOA
symptoms of infection. Retapamulin may be particularly is different from that of other newer topical antimicro-
useful in patients known to be carriers of MRSA or who bials, increasing the diversity of topical antimicrobials
have a history of previous MRSA infections. Retapamulin and maintaining active agents in the face of increasing
may be beneficial in patients who have had systemic side resistance. All clinical trials registered to date have been
effects (such as diarrhea) from antibiotic use, owing to completed.

For personal use only 7 of 14


STAT E O F T H E A RT R E V I E W

Iclaprim Evidence summary and clinical application


Pharmacology Phase II studies of brilacidin have been completed in
Iclaprim, a diaminopyrimidine derivative, is a bacterial more than 300 patients with ABSSIs.78 In the phase IIb
dihydrofolate reductase inhibitor. Inhibition of dihydro- study, two single dose brilacidin strategies (0.6 mg/kg
folate reductase leads to a shortage of nucleosides, inter- intravenously × 1 and 0.8 mg/kg intravenously × 1) and
fering with DNA synthesis and leading to cell death.74 one three dose brilacidin strategy (0.6 mg/kg × 1 followed
Iclaprim’s microbiologic spectrum is broad against Gram by 0.3 mg/kg × 2) were compared against daptomycin
positive organisms, including MRSA, VISA, VRSA, CoNS, 4 mg/kg intravenously daily for seven days. All three
streptococci, and E faecalis. It also has activity against brilacidin dosing strategies performed similarly to dap-
atypical organisms and some Enterobacteriaceae.74 tomycin for the FDA endpoint of early clinical response at
Iclaprim’s half life is about three hours, and it widely 48-72 hours and the EMA endpoint of sustained clinical
distributes throughout tissues. Oral and intravenous response (days 7-8 and 10-14). Brilacidin was generally
preparations are being developed. well tolerated.
All of the brilacidin dosing strategies used in the phase
Evidence summary and clinical application IIb study were lower doses than those used in the phase
Two phase III clinical trials, ASSIST-1 and ASSIST-2, IIa studies and seemed to be better tolerated, with less
have evaluated the use of iclaprim in cSSSIs. In each paresthesia and hypoesthesia observed. These adverse
study, iclaprim 0.8 mg/kg intravenously every 12 hours events have not been associated with neurotoxicity and
was compared with linezolid 600 mg intravenously are reported to have acute onset and rapid resolution.
every 12 hours for 10-14 days in adults.74 Iclaprim Hypertension also seems to be an adverse effect with
showed non-inferiority to linezolid with regard to clini- brilacidin, in a dose dependent and transient fashion.
cal cure rates at end of treatment. Iclaprim was well tol- Phase III studies in ABSSSI are planned.78
erated overall, and the frequency of adverse events was Brilacidin’s novel MOA, high barrier to resistance, and
similar in the two groups, with the rate of any probably favorable efficacy and safety profile make it a welcome
related adverse effects being 6.0% for iclaprim and 9.4% addition to the SSTI treatment armamentarium. Brilacidin
for linezolid.74 is also being developed in topical form for mucositis and
Transient QTc interval prolongation of 4-6 ms has been for otic and ocular administration.79
reported.74 Although no cardiac adverse events were
attributed to iclaprim induced QTc prolongation, cau- Delafloxacin
tion may be warranted in patients with prolonged QTc at Pharmacology
baseline or receiving concomitant drugs that also prolong Delafloxacin, a novel, anionic (non-zwitterionic) fluoro-
the QTc interval.74 In 2008 and 2009 the FDA and EMA quinolone, works by equally inhibiting DNA gyrase and
respectively rejected the approval of iclaprim and called topoisomerase IV.80 It is potently active compared with
for additional studies to demonstrate efficacy. other fluoroquinolones against Gram positive organisms,
Two new phase III studies of iclaprim in ABSSSI, including MSSA and MRSA, especially in inherently acidic
REVIVE-1 and REVIVE-2, are active and recruiting.75 76 environments.81 82 Delafloxacin has a similar Gram nega-
Of note, iclaprim is being studied at a fixed dose of tive spectrum of activity to other fluoroquinolones and
80 mg intravenously every 12 hours in these trials, is also active against anaerobic organisms.83 Because of
in c­ontrast to the previously studied 0.8 mg/kg dose. the MOA, mutations in gyrA and glrA would be needed to
Iclaprim would be an attractive treatment option for confer resistance. Additionally, delafloxacin has not been
SSTIs given its oral and intravenous preparations, shown to readily select mutants in vitro, possibly owing to
as well as its activity against trimethoprim resistant R groups at the C-7 and C-8 structural positions.81
org­anisms.74 Three phase I studies were conducted to determine
safety, tolerability, and pharmacokinetics of intrave-
Brilacidin nously administered delafloxacin in healthy volun-
Pharmacology teers.84 Two additional studies examined delafloxacin
Brilacidin represents the first member of a new class of administered orally.85 Delafloxacin’s terminal half life is
antibiotics, defensin mimetics, and is a cationic, hydro- about 12 hours when administered intravenously and
phobic molecule designed to work similarly to innate host six to eight hours when administered orally; however, in
defense proteins. Brilacidin causes dose dependent depo- the latter studies, the half life lengthened after multiple
larization of the bacterial membrane in S aureus, similarly oral doses. Steady state is expected to be achieved by
to daptomycin, and has broad spectrum of action, includ- day three. Food had no significant effect on total expo-
ing MRSA and VRE.77 sure. Delafloxacin excretion is approximately two thirds
Brilacidin has concentration dependent, bactericidal in urine and one third in feces, primarily as unchanged
activity with linear pharmacokinetics.78 It has a long half drug.86
life of 15-23 hours and post-antibiotic effect, allowing The studies also found that delafloxacin was well toler-
single dose administration. Brilacidin is also thought to ated, with the most common adverse effects being gas-
have anti-inflammatory properties and seems to be active trointestinal and occurring more frequently at doses of
in the stationary phase of bacterial growth, making it 800 mg or above. Notably, no QTc interval prolongation
useful for infections with biofilms. The MOA means that occurred, which differentiates delafloxacin from other
development of resistance may be unlikely. fluoroquinolones.87

For personal use only 8 of 14


STAT E O F T H E A RT R E V I E W

Evidence summary Evidence summary


Two phase II studies have been completed comparing A phase II randomized, double blind, multicenter, non-
delafloxacin with tigecycline, vancomycin, or linezolid inferiority study randomized 161 patients who had ABSS-
in patients with SSTIs.88 89 The first study randomized SIs to either avarofloxacin or linezolid.95 The primary
150 patients with cSSTIs, including wound infection, endpoint evaluated cure rates with a non-inferiority mar-
abscess, and cellulitis, and measured cure rates at the gin of 15% in the intention to treat population. In the pri-
test-of-cure visit 14-21 days after the last dose adminis- mary analysis, avarofloxacin did not meet non-inferiority
tered. They found no significant difference in cure rates criteria; however, the post hoc analyses using 2010 FDA
between delafloxacin 300 mg, delafloxacin 450 mg, and guidance standards and endpoint criteria (lack of lesion
tigecyline standard dosing at 94.3%, 92.5%, and 91.2%, spread and afebrile) indicated non-inferiority compared
respectively; any treatment related adverse effect rates with linezolid (61.4% v 57.7%; P=0.024).96
were lower with delafloxacin 300 mg (44.9%, 62.7%, and
72.0%, respectively). Clinical application
The second study randomized 256 patients with With the available phase II data, avarofloxacin’s place
ABSSSI, including cellulitis, wound infection, abscess, in therapy will be difficult to determine until outcomes
or burn infection, and measured cure rates at the follow- of phase III clinical trials are available. Despite current
up visit. The study found that delafloxacin significantly results, this drug does have potential to compete with
improved cure rates compared with vancomycin (70.4% delafloxacin, as both are potent, broad spectrum, anti-
v 54.1%; P=0.031), with no difference in cure rates MRSA fluoroquinolones.
between delafloxacin and linezolid (70.4% v 64.9%;
P=0.496). Adverse effects related to delafloxacin were Omadacycline
primarily mild and gastrointestinal in nature, similar to Pharmacology
previous phase I studies. Rates of at least one adverse Omadacycline, a semi-synthetic derivative of mino-
effect for delafloxacin, linezolid, and vancomycin were cycline, is the first member of the new 9-aminomethyl
74.4%, 72.0%, and 64.6%, respectively. class of tetracycline drugs in clinical development. It
inhibits protein synthesis similarly to other tetracyclines,
Clinical application although binding the bacterial ribosome.97 Omadacycline
Delafloxacin’s place in therapy will likely reside in has broad activity against Gram positive, Gram nega-
the area of polymicrobial, difficult to treat infections, tive, atypical, and anaerobic pathogens.98 It also retains
owing to its broad spectrum and activity against mul- activity against multidrug resistant organisms, including
tidrug resistant S aureus. Other benefits include oral MRSA and VRE.97 98 Similarly to tigecycline, omadacycline
and in­travenous formulations and negligible effect on has shown activity in the presence of both efflux and ribo-
the QTc interval. Further studies will need to confirm somal protection tetracycline resistance genes.
its safety profile, especially measuring the outcome of Omadacycline has good bioavailability and will likely
Clostridium difficile infection owing to the broad spec- be available in both intravenous and oral preparations.98
trum of a­ctivity. Phase III clinical trials for delafloxacin In a phase I study, no metabolites were found and it
in SSTIs have been completed and are awaiting analysis achieved high concentrations in the urine while being
and publication.90 91 eliminated by renal and gastrointestinal routes.98

Avarofloxacin Evidence summary and clinical application


Pharmacology In a phase II study, omadacycline showed comparable
Avarofloxacin, a broad spectrum fluoroquinolone antibi- efficacy to linezolid for treatment of cSSTI and was well
otic, targets DNA gyrase and topoisomerase IV, much like tolerated, with the most common side effects being gas-
other fluoroquinolones. However, this agent has potent trointestinal (18.9% with omadacycline; 18.5% with
bactericidal activity against MRSA, including fluoroqui- linezolid).99 A phase II clinical trial comparing omada-
nolone resistant strains, and is being developed to treat cycline to linezolid for ABSSSI is active.100 In the future,
ABSSSIs.92 During a phase II clinical trial, avarofloxacin omadacycline may be beneficial in treating SSTIs caused
was effective against S aureus isolates with MIC values by resistant organisms owing to its broad activity and oral
of 0.25 μg/mL or below.93 As avarofloxacin has balanced formulation.
activity against DNA gyrase and topoisomerase IV, muta-
tions at both sites will likely be needed before resistance Fosfomycin
develops. Pharmacology
Evaluated by phase I, placebo controlled, double Fosfomycin, a phosphonic antibiotic, inhibits phospho-
blind, and open label trials in healthy volunteers, enolpyruvate transferase, an enzyme involved in early
av­arofloxacin’s half life is between 12.9 hours and phases of bacterial cell wall peptidoglycan synthesis.101
16.7 hours, and oral bioavailability is around 66%.94 Fosfomycin has broad activity, including MRSA, VRE,
In­travenous doses of up to 150 mg twice daily have been resistant Enterobacteriaceae, and Pseudomonas aer-
tolerated, with gastrointestinal adverse effects, head- uginosa. In the US, fosfomycin trometamol is an oral
ache, and contact dermatitis being the most prominent. formulation approved for the treatment of urinary tract
These adverse effects were similar with oral administra- infections. Outside of the urinary tract, intravenous fos-
tion.94 fomycin, which has approval in some countries outside

For personal use only 9 of 14


STAT E O F T H E A RT R E V I E W

of the US, is generally used in combination with other was cure rate seven to 14 days after the completion of
antimicrobials owing to the potential for development therapy, and ceftobiprole was found to be non-inferior
of resistance. Several resistance mechanisms have been to vancomycin. The second study was a multicenter,
described, including target site modification, decreased randomized, double blind trial in adults with cSSSIs
cellular uptake, and inactivation by fosfomycin resistance comparing a different dosing strategy of ceftobiprole,
kinases.101 102 500 mg intravenously every eight hours infused over two
Intravenous fosfomycin is widely distributed into tis- hours, with vancomycin 1000 mg intravenously every
sues and does not undergo significant metabolism.117 12 hours plus ceftazidime 1000 mg intravenously every
The half life of fosfomycin is approximately two hours eight hours.108 The primary outcome was rate of clinical
and may be prolonged to 3.6-3.8 hours in people with cure seven to 14 days after the completion of therapy,
advanced age or critical illness.103 As fosfomycin’s and ceftobiprole was again found to be non-inferior to
elimination is primary renal, the half life also increases the comparator regimen. In both studies, adverse events
proportionally with the degree of renal insufficiency.103 were not significantly different from comparators and
Standard dosing varies by indication and ranges from most commonly included gastrointestinal symptoms
12-24 g in two to four divided doses; dosage adjust- and headache.108 109
ments are needed in patients with impaired renal
fu­nction.103 Clinical application
Ceftobiprole has yet to receive FDA approval after site
Evidence summary and clinical application inspections and audits found unverifiable data; however,
Fosfomycin has several attributes that could make the ceftobiprole has since been granted qualified infectious
intravenous preparation desirable for future research disease product status by the FDA and clinical phase
in the treatment of SSTIs. In addition to a broad antimi- III development is being pursued again in the US.107 110
crobial spectrum, intravenous fosfomycin achieves high Ceftobiprole is approved for use in Canada for cSSSIs,
concentrations in bone and peripheral soft tissues when dosed at 500 mg intravenously every 12 hours, with
administered intravenously in the setting of diabetic foot dose adjustments needed in patients with decreased renal
infections and also shows synergy with linezolid against function.111 Ceftobiprole also has approval in 13 Euro-
MRSA.104 105 It also has several immunomodulatory pean countries and Australia for community acquired
effects, including decreased production of pro-inflam- and hospital acquired pneumonia. Ceftobiprole’s role
matory cytokines, in vitro inhibition of B cell and T cell would likely be in the treatment of severe SSTIs owing to
activation, and improved susceptibility of some bacteria its broad spectrum of activity, and it would be the first β
to phagocytosis.106 Lastly, fosfomycin is thought to help lactam to provide coverage against both MRSA and sus-
to mitigate the nephrotoxicity of glycopeptides, which are ceptible Pseudomonas aeruginosa.
commonly used in SSTI treatment.106
Phase II agents
Ceftobiprole Lefamulin
Pharmacology Pharmacology
Ceftobiprole, a broad spectrum cephalosporin, inhib- Lefamulin (formerly BC-3781), a semi-synthetic pleuro-
its cell wall synthesis by binding with strong affinity to mutilin antibiotic, works by inhibiting bacterial protein
penicillin binding proteins, including PBP2a, responsible synthesis through binding the peptidyl transferase center
for resistance in S aureus with the mecA gene.107 Cefto- of the ribosome.112 Before the development of intrave-
biprole’s spectrum of action includes Gram positives, nous and oral formulations of lefamulin, the only other
including MRSA, penicillin resistant Streptococcus pneu- pleuromutilin developed for use in humans was topical
moniae, Enterococcus faecalis, Gram negatives, includ- retapamulin. Lefamulin’s spectrum of activity includes
ing susceptible Pseudomonas spp, and some anaerobes. Gram positive organisms such as staphylococci (including
It has shown bactericidal activity and low likelihood of MRSA), streptococci, and enterococci (including VRE),
resistance development. as well as Haemophilus influenzae, Moraxella catarrhalis,
The intravenous formulation is ceftobiprole medocaril and atypical organisms.112
sodium, a pro-drug, which is rapidly converted to the
ceftobiprole drug by plasma esterases.107 No clinically Evidence summary and clinical application
significant drug interactions are expected with ceftobi- A phase II, double blind, multicenter study compared
prole owing to its minimal metabolism and low protein lefamulin 100 mg or 150 mg intravenously every 12
binding (16%).106 Elimination of ceftobiprole occurs pri- hours against vancomycin 1000 mg intravenously every
marily through renal excretion as unchanged drug, and 12 hours for five to 14 days in 207 adults admitted to hos-
the half life is approximately three hours.107 pital with ABSSSI.113 The primary outcome was clinical
success at the test of cure visit seven to 14 days after treat-
Evidence summary ment. Success rates were comparable across all treatment
Two phase III trials have been completed in patients groups, and lefamulin was well tolerated overall, with
with cSSSIs. The first was a multicenter, randomized, patients most commonly experiencing nausea, headache,
double blind trial of ceftobiprole 500 mg intravenously or diarrhea. Lefamulin would be the first systemic pleu-
every 12 hours versus vancomycin 1000 mg intrave- romutilin compound and would offer advantages of both
nously every 12 hours in adults.108 The primary outcome oral and intravenous administration.

For personal use only 10 of 14


STAT E O F T H E A RT R E V I E W

GLOSSARY OF ABBREVIATIONS is exciting to see our SSTI armamentarium expanding,


ABSSSIs—acute bacterial skin and skin structure infections we must be conscientious antimicrobial stewards and
aPTT—activated partial thromboplastin time use these agents responsibly to ensure their future utility.
CoNS—coagulase negative staphylococci Contributors: JGB did the literature search. SLM, JGB, and DLS wrote the
draft article and revised the manuscript. SLM is the guarantor.
cSSSI—complicated skin and skin structure infection
Competing interests: We have read and understood the BMJ policy on
EMA—European Medicines Agency
declaration of interests and declare the following interests: none.
FDA—Food and Drug Administration
Provenance and peer review: Commissioned; externally peer reviewed.
IDSA—Infectious Diseases Society of America
Patient involvement: Owing to the technical nature of this review, patient
MIC—minimum inhibitory concentration involvement was not requested by BMJ.
MOA—mechanism of action 1 Miller LG, Eisenberg DF, Liu H, et al. Incidence of skin and soft tissue
MRSA—meticillin resistant Staphylococcus aureus infections in ambulatory and inpatient settings, 2005-2010. BMC Infect Dis
2015;15:362-9. doi:10.1186/s12879-015-1071-0 pmid:26293161.
MSSA—meticillin susceptible Staphylococcus aureus 2 Corey GR, Stryjewski ME. New rules for clinical trials of patients with
SSTI—skin and soft tissue infection acute bacterial skin and skin-structure infections: do not let the perfect
be the enemy of the good. Clin Infect Dis 2011;52(Suppl 7):S469-76.
VISA—vancomycin intermediate Staphylococcus aureus doi:10.1093/cid/cir162 pmid:21546623.
VRE—vancomycin resistant enterococci 3 US Department of Health and Human Services, Food and Drug
Administration Center for Drug Evaluation Research. Guidance for industry.
VRSA—vancomycin resistant Staphylococcus aureus Acute bacterial skin and skin structure infections: developing drugs for
treatment. 2013. http://www.fda.gov/downloads/Drugs/.../Guidances/
ucm071185.pdf.
Auriclosene 4 Stevens DL, Bisno AL, Chambers HF, et al. Infectious Diseases Society
of America. Practice guidelines for the diagnosis and management of
Pharmacology skin and soft tissue infections: 2014 update by the Infectious Diseases
Auriclosene (formerly NVC-422), an aganocide com- Society of America. Clin Infect Dis 2014;59:e10-52. doi:10.1093/cid/
ciu296 pmid:24973422.
pound and the first in the synthetic stable chlorourine 5 Eriksson B, Jorup-Rönström C, Karkkonen K, Sjöblom AC, Holm SE.
class, preferentially inactivates sulfur groups in amino Erysipelas: clinical and bacteriologic spectrum and serological
aspects. Clin Infect Dis 1996;23:1091-8. doi:10.1093/
acid residues of surface proteins and has broad spectrum clinids/23.5.1091 pmid:8922808.
antibacterial activity, including multidrug resistant MRSA 6 Carrel M, Perencevich EN, David MZ. USA300 Methicillin-Resistant
Staphylococcus aureus, United States, 2000-2013. Emerg Infect Dis
and VRE.114 115 Because auriclosene has activity against 2015;21:1973-80. doi:10.3201/eid2111.150452 pmid:26484389.
multiple surface proteins, development of resistance will 7 Bassetti M, Baguneid M, Bouza E, Dryden M, Nathwani D, Wilcox M.
likely be difficult. European perspective and update on the management of complicated
skin and soft tissue infections due to methicillin-resistant Staphylococcus
aureus after more than 10 years of experience with linezolid. Clin
Evidence summary and clinical application Microbiol Infect 2014;20(Suppl 4):3-18. doi:10.1111/1469-
0691.12463 pmid:24580738.
A phase II study of auriclosene in 129 children aged 8 Zhanel GG, Calic D, Schweizer F, et al. New lipoglycopeptides: a comparative
2-12 years with non-bullous impetigo compared three review of dalbavancin, oritavancin and telavancin. Drugs 2010;70:859-86.
doi:10.2165/11534440-000000000-00000 pmid:20426497.
different concentrations of auriclosene (0.1%, 0.5%, 9 Dalvance® (dalbavancin) [package insert]. Durata Therapeutics, 2016.
and 1.5%) administered topically three times daily for 10 Jones RN, Stilwell MG. Comprehensive update of dalbavancin activity
when tested against uncommonly isolated streptococci, Corynebacterium
seven days.114 Clinical response was defined as clinical spp., Listeria monocytogenes, and Micrococcus spp. (1357 strains).
success or improvement at end of treatment (day 8) and/ Diagn Microbiol Infect Dis 2013;76:239-40. doi:10.1016/j.
or follow-up on day 15 and was attained in more than diagmicrobio.2013.01.002 pmid:23414746.
11 Jones RN, Sader HS, Flamm RK. Update of dalbavancin spectrum and
85% of patients across all concentrations tested. Adverse potency in the USA: report from the SENTRY Antimicrobial Surveillance
events were infrequent and included itching and rash. Program (2011). Diagn Microbiol Infect Dis 2013;75:304-7.
doi:10.1016/j.diagmicrobio.2012.11.024 pmid:23357293.
Auriclosene is a novel treatment agent that would provide 12 Dunne MW, Sahm D, Puttagunta S. Use of vancomycin as a surrogate for
another option for topical treatment of mild SSTIs. dalbavancin in vitro susceptibility testing: results from the DISCOVER
studies. Ann Clin Microbiol Antimicrob 2015;14:19-21. doi:10.1186/
s12941-015-0081-5 pmid:25885674.
Conclusion 13 Jones RN, Farrell DJ, Flamm RK, Sader HS, Dunne MW, Mendes RE.
Surrogate analysis of vancomycin to predict susceptible categorization of
SSTIs place a substantial burden on patients and health- dalbavancin. Diagn Microbiol Infect Dis 2015;82:73-7. doi:10.1016/j.
care systems each year. Increasing resistance among diagmicrobio.2015.01.017 pmid:25724854.
Staphylococcus aureus, in particular, presents major 14 Smith JR, Roberts KD, Rybak MJ. Dalbavancin: a novel lipoglycopeptide
antibiotic with extended activity against Gram-positive infections.
challenges to healthcare providers. The antimicrobi- Infect Dis Ther 2015;4:245-58. doi:10.1007/s40121-015-0077-
als included in this review do not yet have established 7 pmid:26341488.
15 Dunne MW, Puttagunta S, Giordano P, Krievins D, Zelasky M, Baldassarre J.
places in therapy, as they were approved or in clinical A randomized clinical trial of single-dose versus weekly dalbavancin for
development when the 2014 ISDA SSTI guidelines were treatment of acute bacterial skin and skin structure infection. Clin Infect Dis
2016;62:545-51. doi:10.1093/cid/civ982 pmid:26611777.
published. Many new treatment options offer improved 16 Marbury T, Dowell JA, Seltzer E, Buckwalter M. Pharmacokinetics of
convenience and safety. Several agents also have novel dalbavancin in patients with renal or hepatic impairment. J Clin Pharmacol
2009;49:465-76. doi:10.1177/0091270008330162 pmid:19318696.
MOAs or offer advantages in treatment of resistant organ- 17 Vilay AM, Shah KH, Churchwell MD, Patel JH, DePestel DD, Mueller BA.
isms. Alternatively, many SSTIs can be treated with more Modeled dalbavancin transmembrane clearance during intermittent and
continuous renal replacement therapies. Blood Purif 2010;30:37-43.
cost effective oral agents. Several recent studies, along doi:10.1159/000316685 pmid:20588011.
with years of clinical experience, continue to support 18 Dunne MW, Zhou M, Darpo B. A thorough QT study with dalbavancin: a
the use of oral agents such as clindamycin and trimetho- novel lipoglycopeptide antibiotic for the treatment of acute bacterial skin
and skin-structure infections. Int J Antimicrob Agents 2015;45:393-8.
prim/sulfamethoxazole for patients with mild to mod- doi:10.1016/j.ijantimicag.2014.12.021 pmid:25681068.
erate SSTIs.116 117 Practitioners must carefully consider 19 Jauregui LE, Babazadeh S, Seltzer E, et al. Randomized, double-blind
comparison of once-weekly dalbavancin versus twice-daily linezolid
several patient specific factors to appropriately select therapy for the treatment of complicated skin and skin structure infections.
and in­dividualize SSTI treatment regimens. Although it Clin Infect Dis 2005;41:1407-15. doi:10.1086/497271 pmid:16231250.

For personal use only 11 of 14


STAT E O F T H E A RT R E V I E W

20 Boucher HW, Wilcox M, Talbot GH, Puttagunta S, Das AF, Dunne MW. 43 Rodríguez-Avial I, Culebras E, Betriu C, Morales G, Pena I, Picazo JJ.
Once-weekly dalbavancin versus daily conventional therapy for In vitro activity of tedizolid (TR-700) against linezolid-resistant
skin infection. N Engl J Med 2014;370:2169-79. doi:10.1056/ staphylococci. J Antimicrob Chemother 2012;67:167-9. doi:10.1093/jac/
NEJMoa1310480 pmid:24897082. dkr403 pmid:21954458.
21 Corey GR, Kabler H, Mehra P, et al. SOLO I Investigators. Single-dose 44 Barber KE, Smith JR, Raut A, Rybak MJ. Evaluation of tedizolid against
oritavancin in the treatment of acute bacterial skin infections. N Engl J Med Staphylococcus aureus and enterococci with reduced susceptibility
2014;370:2180-90. doi:10.1056/NEJMoa1310422 pmid:24897083. to vancomycin, daptomycin or linezolid. J Antimicrob Chemother
22 Corey GR, Good S, Jiang H, et al. SOLO II Investigators. Single-dose 2016;71:152-5. doi:10.1093/jac/dkv302 pmid:26476277.
oritavancin versus 7-10 days of vancomycin in the treatment of gram- 45 Sivextro® (tedizolid) [package insert]. Merck & Co, 2015.
positive acute bacterial skin and skin structure infections: the SOLO II 46 Ong V, Flanagan S, Fang E, et al. Absorption, distribution, metabolism,
noninferiority study. Clin Infect Dis 2015;60:254-62. doi:10.1093/cid/ and excretion of the novel antibacterial prodrug tedizolid
ciu778 pmid:25294250. phosphate. Drug Metab Dispos 2014;42:1275-84. doi:10.1124/
23 Shorr AF, Lodise TP, Corey GR, et al. Analysis of the phase 3 ESTABLISH dmd.113.056697 pmid:24875463.
trials of tedizolid versus linezolid in acute bacterial skin and skin 47 Bradley JS, Flanagan SD, Arrieta AC, Jacobs R, Capparelli E, Prokocimer P.
structure infections. Antimicrob Agents Chemother 2015;59:864-71. Pharmacokinetics, safety and tolerability of single oral or
doi:10.1128/AAC.03688-14 pmid:25421472. intravenous administration of 200 mg tedizolid phosphate in
24 Bradley JS, Puttagunta S, Rubino CM, Blumer JL, Dunne M, Sullivan JE. adolescents. Pediatr Infect Dis J 2016;35:628-33. doi:10.1097/
Pharmacokinetics, safety and tolerability of single dose dalbavancin INF.0000000000001096 pmid:26910588.
in children 12-17 years of age. Pediatr Infect Dis J 2015;34:748-52. 48 Prokocimer P, Bien P, Surber J, et al. Phase 2, randomized, double-blind,
doi:10.1097/INF.0000000000000646 pmid:25551831. dose-ranging study evaluating the safety, tolerability, population
25 Zhanel GG, Schweizer F, Karlowsky JA. Oritavancin: mechanism of pharmacokinetics, and efficacy of oral torezolid phosphate in patients with
action. Clin Infect Dis 2012;54(Suppl 3):S214-9. doi:10.1093/cid/ complicated skin and skin structure infections. Antimicrob Agents Chemother
cir920 pmid:22431851. 2011;55:583-92. doi:10.1128/AAC.00076-10 pmid:21115795.
26 Brade KD, Rybak JM, Rybak MJ. Oritavancin: a new lipoglycopeptide 49 Flanagan S, Minassian SL, Morris D, et al. Pharmacokinetics of tedizolid in
antibiotic in the treatment of Gram-positive infections. Infect Dis Ther subjects with renal or hepatic impairment. Antimicrob Agents Chemother
2016;5:1-15. doi:10.1007/s40121-016-0103-4 pmid:26831328. 2014;58:6471-6. doi:10.1128/AAC.03431-14 pmid:25136024.
27 Belley A, Neesham-Grenon E, McKay G, et al. Oritavancin kills stationary- 50 Prokocimer P, De Anda C, Fang E, Mehra P, Das A. Tedizolid phosphate vs
phase and biofilm Staphylococcus aureus cells in vitro. Antimicrob linezolid for treatment of acute bacterial skin and skin structure infections:
Agents Chemother 2009;53:918-25. doi:10.1128/AAC.00766- the ESTABLISH-1 randomized trial. JAMA 2013;309:559-69. doi:10.1001/
08 pmid:19104027. jama.2013.241 pmid:23403680.
28 Mendes RE, Farrell DJ, Sader HS, Flamm RK, Jones RN. Activity of 51 Moran GJ, Fang E, Corey GR, Das AF, De Anda C, Prokocimer P. Tedizolid for 6
oritavancin against Gram-positive clinical isolates responsible for days versus linezolid for 10 days for acute bacterial skin and skin-structure
documented skin and soft-tissue infections in European and US hospitals infections (ESTABLISH-2): a randomised, double-blind, phase 3, non-
(2010-13). J Antimicrob Chemother 2015;70:498-504. doi:10.1093/ inferiority trial. Lancet Infect Dis 2014;14:696-705. doi:10.1016/S1473-
jac/dku421 pmid:25362568. 3099(14)70737-6 pmid:24909499.
29 Mendes RE, Sader HS, Flamm RK, Jones RN. Activity of oritavancin tested 52 Flanagan S, McKee EE, Das D, et al. Nonclinical and pharmacokinetic
against uncommonly isolated Gram-positive pathogens responsible for assessments to evaluate the potential of tedizolid and linezolid to affect
documented infections in hospitals worldwide. J Antimicrob Chemother mitochondrial function. Antimicrob Agents Chemother 2015;59:178-85.
2014;69:1579-81. doi:10.1093/jac/dku016 pmid:24505091. doi:10.1128/AAC.03684-14 pmid:25331703.
30 Jones RN, Turnidge JD, Moeck G, Arhin FF, Mendes RE. Use of in vitro 53 Flanagan S, Bartizal K, Minassian SL, Fang E, Prokocimer P. In vitro, in vivo,
vancomycin testing results to predict susceptibility to oritavancin, and clinical studies of tedizolid to assess the potential for peripheral or
a new long-acting lipoglycopeptide. Antimicrob Agents Chemother central monoamine oxidase interactions. Antimicrob Agents Chemother
2015;59:2405-9. doi:10.1128/AAC.05098-14 pmid:25666152. 2013;57:3060-6. doi:10.1128/AAC.00431-13 pmid:23612197.
31 Jones RN, Moeck G, Arhin FF, Dudley MN, Rhomberg PR, Mendes RE. 54 ClinicalTrials.gov. Tedizolid tissue penetration in diabetic patients with
Results from oritavancin resistance surveillance programs (2011 wound infections and healthy volunteers via in vivo microdialysis. 2015.
to 2014): clarification for using vancomycin as a surrogate to infer https://clinicaltrials.gov/ct2/show/NCT02620787.
oritavancin susceptibility. Antimicrob Agents Chemother 2016;60:3174- 55 ClinicalTrials.gov. Study of tedizolid phosphate in adolescents with
7. doi:10.1128/AAC.03029-15 pmid:26926647. complicated skin and soft tissue infections (cSSTI) (MK-1986-012). 2016.
32 Orbactiv® (oritavancin) [package insert]. The Medicines Company, 2016. https://clinicaltrials.gov/ct2/show/NCT02276482.
33 Rubino CM, Bhavnani SM, Moeck G, Bellibas SE, Ambrose PG. Population 56 Scangarella-Oman NE, Shawar RM, Bouchillon S, Hoban D. Microbiological
pharmacokinetic analysis for a single 1,200-milligram dose of profile of a new topical antibacterial: retapamulin ointment 1%. Expert Rev
oritavancin using data from two pivotal phase 3 clinical trials. Antimicrob Anti Infect Ther 2009;7:269-79. doi:10.1586/eri.09.7 pmid:19344241.
Agents Chemother 2015;59:3365-72. doi:10.1128/AAC.00176- 57 Rittenhouse S, Biswas S, Broskey J, et al. Selection of retapamulin,
15 pmid:25824211. a novel pleuromutilin for topical use. Antimicrob Agents Chemother
34 Kumar A, Mann HJ, Keshtgarpour M, et al. In vitro characterization of 2006;50:3882-5. doi:10.1128/AAC.00178-06 pmid:17065625.
oritavancin clearance from human blood by low-flux, high-flux, and 58 Jones RN, Fritsche TR, Sader HS, Ross JE. Activity of retapamulin (SB-
continuous renal replacement therapy dialyzers. Int J Artif Organs 275833), a novel pleuromutilin, against selected resistant gram-positive
2011;34:1067-74. doi:10.5301/ijao.5000008 pmid:22183520. cocci. Antimicrob Agents Chemother 2006;50:2583-6. doi:10.1128/
35 Darpo B, Lee SK, Moon TE, Sills N, Mason JW. Oritavancin, AAC.01432-05 pmid:16801451.
a new lipoglycopeptide antibiotic: results from a 59 Woodford N, Afzal-Shah M, Warner M, Livermore DM. In vitro activity of
thorough QT study. J Clin Pharmacol 2010;50:895-903. retapamulin against Staphylococcus aureus isolates resistant to fusidic acid
doi:10.1177/0091270009355449 pmid:20484618. and mupirocin. J Antimicrob Chemother 2008;62:766-8. doi:10.1093/jac/
36 ClinicalTrials.gov. Open-label, dose-finding, pharmacokinetics, safety and dkn266 pmid:18567573.
tolerability study of oritavancin in pediatric patients with suspected or 60 McNeil JC, Hulten KG, Kaplan SL, Mason EO. Decreased susceptibilities
confirmed bacterial infections. 2015. https://clinicaltrials.gov/ct2/show/ to Retapamulin, Mupirocin, and Chlorhexidine among Staphylococcus
NCT02134301. aureus isolates causing skin and soft tissue infections in otherwise healthy
37 ClinicalTrials.gov. Study to evaluate the safety, tolerability and children. Antimicrob Agents Chemother 2014;58:2878-83. doi:10.1128/
pharmacokinetics of a new formulation of oritavancin in healthy AAC.02707-13 pmid:24614375.
volunteers. 2016. https://clinicaltrials.gov/ct2/show/NCT02471690. 61 Kosowska-Shick K, Clark C, Credito K, et al. Single- and multistep resistance
38 ClinicalTrials.gov. A study to evaluate the safety of a single IV dose of selection studies on the activity of retapamulin compared to other agents
Orbactiv (oritavancin) in subjects on chronic warfarin therapy being against Staphylococcus aureus and Streptococcus pyogenes. Antimicrob
treated for acute bacterial skin and skin structure infection (ABSSSI). Agents Chemother 2006;50:765-9. doi:10.1128/AAC.50.2.765-
2016. https://clinicaltrials.gov/ct2/show/NCT02452918. 769.2006 pmid:16436741.
39 Burdette SD, Trotman R. Tedizolid: the first once-daily oxazolidinone 62 Gentry DR, Rittenhouse SF, McCloskey L, Holmes DJ. Stepwise exposure
class antibiotic. Clin Infect Dis 2015;61:1315-21. doi:10.1093/cid/ of Staphylococcus aureus to pleuromutilins is associated with stepwise
civ501 pmid:26105167. acquisition of mutations in rplC and minimally affects susceptibility
40 Rybak JM, Marx K, Martin CA. Early experience with tedizolid: clinical to retapamulin. Antimicrob Agents Chemother 2007;51:2048-52.
efficacy, pharmacodynamics, and resistance. Pharmacotherapy doi:10.1128/AAC.01066-06 pmid:17404009.
2014;34:1198-208. doi:10.1002/phar.1491 pmid:25266820. 63 Altabax® (retapamulin) [package insert]. GlaxoSmithKline, 2007.
41 Drusano GL, Liu W, Kulawy R, Louie A. Impact of granulocytes on the 64 Koning S, van der Wouden JC, Chosidow O, et al. Efficacy and safety of
antimicrobial effect of tedizolid in a mouse thigh infection model. retapamulin ointment as treatment of impetigo: randomized double-blind
Antimicrob Agents Chemother 2011;55:5300-5. doi:10.1128/ multicentre placebo-controlled trial. Br J Dermatol 2008;158:1077-82.
AAC.00502-11 pmid:21911576. doi:10.1111/j.1365-2133.2008.08485.x pmid:18341664.
42 Zurenko G, Bien P, Bensaci M, Patel HN, Thorne G. Use of linezolid 65 Oranje AP, Chosidow O, Sacchidanand S, et al. TOC100224
susceptibility test results as a surrogate for the susceptibility of Study Team. Topical retapamulin ointment, 1%, versus sodium
Gram-positive pathogens to tedizolid, a novel oxazolidinone. Ann Clin fusidate ointment, 2%, for impetigo: a randomized, observer-
Microbiol Antimicrob 2014;13:46-52. doi:10.1186/s12941-014-0046- blinded, noninferiority study. Dermatology 2007;215:331-40.
0 pmid:25238753. doi:10.1159/000107776 pmid:17911992.

For personal use only 12 of 14


STAT E O F T H E A RT R E V I E W

66 Vingsbo Lundberg C, Frimodt-Møller N. Efficacy of topical and 88 O’Riordan W, Mehra P, Manos P, Kingsley J, Lawrence L, Cammarata S.
systemic antibiotic treatment of meticillin-resistant Staphylococcus A randomized phase 2 study comparing two doses of delafloxacin
aureus in a murine superficial skin wound infection model. with tigecycline in adults with complicated skin and skin-structure
Int J Antimicrob Agents 2013;42:272-5. doi:10.1016/j. infections. Int J Infect Dis 2015;30:67-73. doi:10.1016/j.
ijantimicag.2013.05.008 pmid:23837927. ijid.2014.10.009 pmid:25448332.
67 ClinicalTrials.gov. Retapamulin for reducing MRSA nasal carriage. 2016. 89 Kingsley J, Mehra P, Lawrence LE, et al. A randomized, double-blind,
https://clinicaltrials.gov/ct2/show/NCT01461668. Phase 2 study to evaluate subjective and objective outcomes in
68 Yamakawa T, Mitsuyama J, Hayashi K. In vitro and in vivo antibacterial patients with acute bacterial skin and skin structure infections treated
activity of T-3912, a novel non-fluorinated topical quinolone. with delafloxacin, linezolid or vancomycin. J Antimicrob Chemother
J Antimicrob Chemother 2002;49:455-65. doi:10.1093/ 2016;71:821-9. doi:10.1093/jac/dkv411 pmid:26679243.
jac/49.3.455 pmid:11864945. 90 ClinicalTrials.gov. Delafloxacin versus vancomycin and aztreonam for
69 López Y, Tato M, Espinal P, et al. In vitro activity of Ozenoxacin against the treatment of acute bacterial skin and skin structure infections. 2015
quinolone-susceptible and quinolone-resistant gram-positive bacteria. https://clinicaltrials.gov/ct2/show/NCT01811732.
Antimicrob Agents Chemother 2013;57:6389-92. doi:10.1128/ 91 ClinicalTrials.gov. Delafloxacin vs vancomycin and aztreonam for the
AAC.01509-13 pmid:24080666. treatment of acute bacterial skin and skin structure infections. 2016.
70 Gropper S, Cepero AL, Santos B, Kruger D. Systemic bioavailability and https://clinicaltrials.gov/ct2/show/NCT01984684.
safety of twice-daily topical ozenoxacin 1% cream in adults and children 92 Farrell DJ, Liverman LC, Biedenbach DJ, Jones R. JNJ-Q2: a new
with impetigo. Future Microbiol 2014;9(Suppl):S33-40. doi:10.2217/ fluoroquinolone with potent in vitro activity against staphylococcus
fmb.14.85 pmid:25209523. aureus, including methicillin- and fluoroquinolone-resistant strains
71 Gropper S, Albareda N, Santos B, Febbraro S. Skin tissue exposure [abstract]. IDSA, Vancouver, Canada, Oct 21-24 2010.
of once- versus twice-daily topical ozenoxacin 2% cream: a Phase I 93 Farrell DJ, Turner LL, Castanheira M, Jones RN. Activity of JNJ-Q2 against
study in healthy volunteers. Future Microbiol 2014;9(Suppl):S17-22. Staphylococcus aureus isolated from patients with acute bacterial
doi:10.2217/fmb.14.83 pmid:25209520. skin and skin-structure infection obtained during a Phase 2 clinical
72 Kanayama S, Ikeda F, Okamoto K, et al. In vitro antimicrobial activity trial. Diagn Microbiol Infect Dis 2012;74:73-4. doi:10.1016/j.
of ozenoxacin against methicillin-susceptible Staphylococcus aureus, diagmicrobio.2012.05.031 pmid:22819242.
methicillin-resistant S. aureus and Streptococcus pyogenes isolated from 94 Davenport JM, Covington P, Gotfried M, et al. Summary of
clinical cutaneous specimens in Japan. J Infect Chemother 2016;22:720- Pharmacokinetics and Tissue Distribution of a Broad-Spectrum
3. doi:10.1016/j.jiac.2016.03.006 pmid:27091753. Fluoroquinolone, JNJ-Q2. Clin Pharmacol Drug Dev 2012;1:121-30.
73 Gropper S, Albareda N, Chelius K, et al. Ozenoxacin in Impetigo Trial doi:10.1177/2160763X12454714 pmid:27121454.
Investigators Group. Ozenoxacin 1% cream in the treatment of impetigo: 95 Covington P, Davenport JM, Andrae D, et al. Randomized, double-blind,
a multicenter, randomized, placebo- and retapamulin-controlled phase II, multicenter study evaluating the safety/tolerability and
clinical trial. Future Microbiol 2014;9:1013-23. doi:10.2217/ efficacy of JNJ-Q2, a novel fluoroquinolone, compared with linezolid for
fmb.14.78 pmid:25340832. treatment of acute bacterial skin and skin structure infection. Antimicrob
74 Morgan A, Cofer C, Stevens DL. Iclaprim: a novel dihydrofolate Agents Chemother 2011;55:5790-7. doi:10.1128/AAC.05044-
reductase inhibitor for skin and soft tissue infections. Future Microbiol 11 pmid:21947389.
2009;4:131-44. doi:10.2217/17460913.4.2.131 pmid:19257839. 96 Corey GR, Stryjewski ME. New rules for clinical trials of patients with
75 ClinicalTrials.gov. Phase 3 study to evaluate safety and efficacy of acute bacterial skin and skin-structure infections: do not let the perfect
iclaprim versus vancomycin for ABSSSI: REVIVE-1 (REVIVE-1). 2016. be the enemy of the good. Clin Infect Dis 2011;52(Suppl 7):S469-76.
https://clinicaltrials.gov/ct2/show/NCT02600611. doi:10.1093/cid/cir162 pmid:21546623.
76 ClinicalTrials.gov. Phase 3 study to evaluate safety and efficacy of 97 Draper MP, Weir S, Macone A, et al. Mechanism of action of the
iclaprim versus vancomycin for ABSSSI: REVIVE-2 (REVIVE-2). 2016. novel aminomethylcycline antibiotic omadacycline. Antimicrob
https://clinicaltrials.gov/ct2/show/NCT02607618. Agents Chemother 2014;58:1279-83. doi:10.1128/AAC.01066-
77 Mensa B, Howell GL, Scott R, DeGrado WF. Comparative mechanistic 13 pmid:24041885.
studies of brilacidin, daptomycin, and the antimicrobial peptide LL16. 98 Macone AB, Caruso BK, Leahy RG, et al. In vitro and in vivo antibacterial
Antimicrob Agents Chemother 2014;58:5136-45. doi:10.1128/ activities of omadacycline, a novel aminomethylcycline. Antimicrob
AAC.02955-14 pmid:24936592. Agents Chemother 2014;58:1127-35. doi:10.1128/AAC.01242-
78 Jorgensen D, Scott R, O’Riordan W, Tack K. A randomized, double-blind 13 pmid:24295985.
study comparing single-dose and short-course brilacidin to daptomycin 99 Noel GJ, Draper MP, Hait H, Tanaka SK, Arbeit RD. A randomized,
in the treatment of acute bacterial skin & skin structure infections evaluator-blind, phase 2 study comparing the safety and efficacy of
(ABSSSI) [abstract 2969]. ECCMID, Copenhagen, Denmark, Apr 25-28 omadacycline to those of linezolid for treatment of complicated skin and
2015. skin structure infections. Antimicrob Agents Chemother 2012;56:5650-
79 Cellceutix. Product pipeline. 2016. http://cellceutix.com/ 4. doi:10.1128/AAC.00948-12 pmid:22908151.
pipeline/#sthash.PRtOfp0B.dpbs. 100 ClinicalTrials.gov. Omadacycline versus linezolid for the treatment of
80 Nilius AM, Shen LL, Hensey-Rudloff D, et al. In vitro antibacterial ABSSSI (EudraCT #2013-003644-23). 2016. https://clinicaltrials.gov/
potency and spectrum of ABT-492, a new fluoroquinolone. Antimicrob ct2/show/NCT02378480.
Agents Chemother 2003;47:3260-9. doi:10.1128/AAC.47.10.3260- 101 Reffert JL, Smith WJ. Insights from the Society of Infectious Diseases
3269.2003 pmid:14506039. Pharmacists. Fosfomycin for the treatment of resistant gram-negative
81 Remy JM, Tow-Keogh CA, McConnell TS, Dalton JM, Devito JA. Activity bacterial infections. Pharmacotherapy 2014;34:845-57. doi:10.1002/
of delafloxacin against methicillin-resistant Staphylococcus aureus: phar.1434 pmid:24782335.
resistance selection and characterization. J Antimicrob Chemother 102 Thompson MK, Keithly ME, Goodman MC, et al. Structure and function
2012;67:2814-20. doi:10.1093/jac/dks307 pmid:22875850. of the genomically encoded fosfomycin resistance enzyme, FosB, from
82 Lemaire S, Tulkens PM, Van Bambeke F. Contrasting effects of acidic pH Staphylococcus aureus. Biochemistry 2014;53:755-65. doi:10.1021/
on the extracellular and intracellular activities of the anti-gram-positive bi4015852 pmid:24447055.
fluoroquinolones moxifloxacin and delafloxacin against Staphylococcus 103 Fomicyt [summary of product characteristics]. Nordic Pharma UK Ltd,
aureus. Antimicrob Agents Chemother 2011;55:649-58. doi:10.1128/ 2014.
AAC.01201-10 pmid:21135179. 104 Xu-hong Y, Falagas ME, Dong W, Karageorgopoulos DE, De-feng L,
83 Van Bambeke F. Delafloxacin, a non-zwitterionic fluoroquinolone Rui W. In vitro activity of fosfomycin in combination with linezolid
in Phase III of clinical development: evaluation of its against clinical isolates of methicillin-resistant Staphylococcus
pharmacology, pharmacokinetics, pharmacodynamics and clinical aureus. J Antibiot (Tokyo) 2014;67:369-71. doi:10.1038/
efficacy. Future Microbiol 2015;10:1111-23. doi:10.2217/ ja.2014.5 pmid:24517925.
fmb.15.39 pmid:26119479. 105 Schintler MV, Traunmüller F, Metzler J, et al. High fosfomycin
84 Hoover R, Hunt T, Benedict M, et al. Safety, Tolerability, and concentrations in bone and peripheral soft tissue in diabetic patients
Pharmacokinetic Properties of Intravenous Delafloxacin After Single presenting with bacterial foot infection. J Antimicrob Chemother
and Multiple Doses in Healthy Volunteers. Clin Ther 2016;38:53-65. 2009;64:574-8. doi:10.1093/jac/dkp230 pmid:19578081.
doi:10.1016/j.clinthera.2015.11.019 pmid:26718605. 106 Roussos N, Karageorgopoulos DE, Samonis G, Falagas ME. Clinical
85 Hoover R, Hunt T, Benedict M, et al. Single and Multiple significance of the pharmacokinetic and pharmacodynamic
Ascending-dose Studies of Oral Delafloxacin: Effects of Food, characteristics of fosfomycin for the treatment of patients with systemic
Sex, and Age. Clin Ther 2016;38:39-52. doi:10.1016/j. infections. Int J Antimicrob Agents 2009;34:506-15. doi:10.1016/j.
clinthera.2015.10.016 pmid:26581327. ijantimicag.2009.08.013 pmid:19828298.
86 McEwen A, Lawrence L, Hoover R, et al. Disposition, metabolism 107 Dauner DG, Nelson RE, Taketa DC. Ceftobiprole: A novel, broad-spectrum
and mass balance of delafloxacin in healthy human volunteers cephalosporin with activity against methicillin-resistant Staphylococcus
following intravenous administration. Xenobiotica 2015;45:1054-62. aureus. Am J Health Syst Pharm 2010;67:983-93. doi:10.2146/
doi:10.3109/00498254.2015.1042946 pmid:25986539. ajhp090285 pmid:20516468.
87 Litwin JS, Benedict MS, Thorn MD, Lawrence LE, Cammarata SK, Sun E. 108 Noel GJ, Bush K, Bagchi P, Ianus J, Strauss RS. A randomized, double-
A thorough QT study to evaluate the effects of therapeutic and blind trial comparing ceftobiprole medocaril with vancomycin plus
supratherapeutic doses of delafloxacin on cardiac repolarization. ceftazidime for the treatment of patients with complicated skin
Antimicrob Agents Chemother 2015;59:3469-73. doi:10.1128/ and skin-structure infections. Clin Infect Dis 2008;46:647-55.
AAC.04813-14 pmid:25845864. doi:10.1086/526527 pmid:18225981.

For personal use only 13 of 14


STAT E O F T H E A RT R E V I E W

109 Basilea Pharmaceutica. Ceftobiprole. 2016. http://www.basilea.com/ 113 Prince WT, Ivezic-Schoenfeld Z, Lell C, et al. Phase II clinical study of
Portfolio/Ceftobiprole/. BC-3781, a pleuromutilin antibiotic, in treatment of patients with acute
110 Noel GJ, Strauss RS, Amsler K, Heep M, Pypstra R, Solomkin JS. Results bacterial skin and skin structure infections. Antimicrob Agents Chemother
of a double-blind, randomized trial of ceftobiprole treatment of 2013;57:2087-94. doi:10.1128/AAC.02106-12 pmid:23422913.
complicated skin and skin structure infections caused by gram-positive 114 Iovino SM, Krantz KD, Blanco DM, et al. NVC-422 topical gel for the treatment
bacteria. Antimicrob Agents Chemother 2008;52:37-44. doi:10.1128/ of impetigo. Int J Clin Exp Pathol 2011;4:587-95.pmid:21904634.
AAC.00551-07 pmid:17954698. 115 NovaBay Pharmaceuticals. Aganocides. 2016. http://novabay.com/
111 Health Canada. Regulatory decision summary: Zevtera. 2015. http:// technology/aganocides.
www.hc-sc.gc.ca/dhp-mps/prodpharma/rds-sdr/drug-med/rds_sdr_ 116 Talan DA, Lovecchio F, Abrahamian FM, et al. A randomized trial of
zevtera_178459-eng.php. clindamycin versus trimethoprim-sulfamethoxazole for uncomplicated
112 Paukner S, Sader HS, Ivezic-Schoenfeld Z, Jones RN. Antimicrobial activity wound infection. Clin Infect Dis 2016;62:1505-13. doi:10.1093/cid/
of the pleuromutilin antibiotic BC-3781 against bacterial pathogens ciw177 pmid:27025829.
isolated in the SENTRY antimicrobial surveillance program in 2010. 117 Talan DA, Mower WR, Krishnadasan A, et al. Trimethoprim-sulfamethoxazole
Antimicrob Agents Chemother 2013;57:4489-95. doi:10.1128/ versus placebo for uncomplicated skin abscess. N Engl J Med 2016;374:823-
AAC.00358-13 pmid:23836172. 32. doi:10.1056/NEJMoa1507476 pmid:26962903.

For personal use only 14 of 14

You might also like