Skip to main content
Erschienen in: BMC Infectious Diseases 1/2024

Open Access 01.12.2024 | Research

Efficacy and safety of omadacycline for treating complicated skin and soft tissue infections: a meta-analysis of randomized controlled trials

verfasst von: Wenxin Liang, Hong Yin, Huiling Chen, Juan Xu, Yun Cai

Erschienen in: BMC Infectious Diseases | Ausgabe 1/2024

Abstract

Objective

In the present study, we aimed to compare the clinical efficacy and safety of omadacycline (OMC) with its comparators for the treatment of complicated skin and soft tissue infections (cSSTIs) in adult patients.

Methods

Randomized controlled trials (RCTs) evaluating OMC for cSSTIs were searched in databases of PubMed, Embase, Cochrane, Web of Science, and Clinical Trial, up to July 2022. The primary outcomes were clinical efficacy and microbiological response, with secondary outcome was safety.

Results

Four RCTs consisting of 1,757 patients were included, with linezolid (LZD) as a comparator drug. For clinical efficacy, OMC was not inferior to LZD in the modified intent-to-treat (MITT) (OR: 1.24, 95% Cl: [0.93, 1.66], P = 0.15) and clinically evaluable (CE) populations (OR: 1.92, 95% Cl: [0.94, 3.92], P = 0.07). For microbiological response, OMC was numerically higher than LZD in the microbiologically evaluable (ME) (OR: 1.74, 95% Cl: [0.81, 3.74], P = 0.16) and microbiological MITT (micro-MITT) populations (OR: 1.27, 95% Cl: [0.92, 1.76], P = 0.14). No significant difference was found in subpopulations of monomicrobial or polymicrobial mixed infection populations. The mortality and adverse event rates were similar between OMC and LZD.

Conclusions

OMC was as good as LZD in terms of clinical efficacy and microbiological response, and has similar safety issues in treating cSSTIs. OMC might be a promising option for treating cSSTIs in adult patients.
Hinweise
Wenxin Liang and Hong Yin contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Background

Skin and soft tissue infections (SSTIs) are caused by bacteria invading the skin and surrounding tissues, which is a common problem in hospitals. In the United States, there are more than 14 million outpatients with SSTIs and almost 900,000 hospitalized patients yearly [1].
According to the extent of infected skin, the Infections Diseases Society of America (IDSA) divides SSTIs into complicated SSTIs (cSSTIs) and uncomplicated SSTIs [2]. cSSTIs include deep soft tissue infections, such as necrotizing infections, infected ulcers, infected burns, and severe abscesses [3]. Uncomplicated SSTIs refer to superficial infections, including cellulitis, simple abscesses, impetigo, and furuncles [3]. Gram-positive bacteria are the primary pathogens of cSSTIs, among which Staphylococcus aureus is the most common one, and methicillin-resistant Staphylococcus aureus (MRSA) accounts for 46% of S. aureus isolates [4]. Other Gram-positive bacteria include Streptococcus pyogenes, Enterococcus faecalis, etc. In contrast, Gram-negative bacteria are less common in cSSTIs [4].
The most recent guideline of the Surgical Infection Society (SIS) on the management of cSSTIs suggests that vancomycin, linezolid (LZD), daptomycin, ceftaroline, and telavancin are first-line agents for cSSTIs caused by MRSA [5]. Vancomycin has always been the standard treatment for MRSA-caused cSSTIs, while LZD has been proven to be an effective substitute for vancomycin [6]. However, these drugs have limitations in their clinical application. Vancomycin has low tissue penetration and nephrotoxicity risk during treatment. Moreover, only intravenous dosage forms of vancomycin can be used. Long-term use of LZD may cause thrombocytopenia, as well as peripheral and central neuropathies [7, 8]. Since cSSTIs are characterized by deep tissue involvement and diverse pathogens, long-term antimicrobial therapy is usually required. Therefore, the optimal antibiotics for cSSTIs must have good tissue distribution, a broad antibacterial spectrum, and long-term medication safety and compliance.
Omadacycline (Nuzyra, PTK 0796, OMC) is a third-generation tetracycline derivative, but the first aminomethylcycline. It inhibits the synthesis of bacterial proteins by binding to 30 s ribosomal subunits and blocking the binding of aminoacyl tRNA [9, 10]. The structural modification of OMC makes it overcome the common tetracycline resistance mechanisms, such as the increasing number of efflux pumps and the production of ribosomal protective proteins [11, 12]. OMC has been proven to have good antibacterial activity against common clinical Gram-positive bacteria, Gram-negative bacteria, and anaerobes, showing low minimum inhibitory concentrations [13, 14]. OMC has high oral bioavailability, allowing intravenous injection and oral administration [15], which is convenient for outpatients and discharged patients with medicine. The dosage of once a day improves the patient's compliance with medication [16]. In addition, OMC has the characteristics of a large steady-state distribution volume and a high tissue penetration rate [10, 17, 18]. For patients with liver and kidney impairments, there is no need to adjust the dosage of OMC [19]. The FDA approved OMC to treat acute bacterial skin and skin structure infections and community-acquired bacterial pneumonia in October 2018 [20].
The present meta-analysis included all available randomized controlled trials (RCTs) to comprehensively evaluate the efficacy and safety of OMC in the treatment of cSSTIs. Collectively, our current findings provided valuable insights into the treatment for cSSTIs in clinical practice.

Methods

Study search and selection

This study was conducted in accordance with the Preferred Reporting Program for Systematic Review and Meta-Analysis (PRISMA) statement [21]. PubMed, Embase, Cochrane, Web of Science, and Clinical Trial databases were searched from the establishment of the database to July 2022, regardless of language, using the following search terms: “omadacycline” OR “nuzyra” OR “PTK 0796”. Duplicate records were eliminated using Endnote X9, and two reviewers (Liang and Yin) independently monitored the records to avoid bias according to the inclusion and exclusion criteria. If any disagreement arose during the review process, a third reviewer (Xu) would decide. Inclusion criteria were set as follows: ① RCTs of efficacy and safety, ② patients over 18 years old with SSTIs, and ③ the patients in the test group were treated with OMC, and the patients in the control group were treated with other drugs. Exclusion criteria were set as follows: ① republished literature, ② review, case report, etc., and ③ data from the same RCT.

Data extraction and quality assessment

In all included studies, the data were extracted primarily by two researchers independently, and if there was disagreement during extraction, it was examined and determined by a third investigator. The following information was extracted from included studies, such as the first author, publication year, research place, start and end time, intervention measures, sample size, outcome indicators, pathogenic microorganisms, infection types, size of lesion and drainage procedures. According to the items in the Cochrane Collaboration Risk of Bias Tool, the risk of bias in all included studies was rated as “low risk”, “unclear”, or “high risk” [22]. The quality of the included studies was evaluated using the Jadad scale [23]. A total score of 0 ~ 5 points, including 0 ~ 2 for randomization, 0 ~ 2 for blinding, and 0 ~ 1 for withdrawal.

Definitions

The modified intent-to-treat (MITT) population included all randomized patients who did not have a sole Gram-negative causative pathogen at baseline. The clinically evaluable (CE) population included patients in the MITT population who had a qualifying infection as per study-entry criteria, received the study drug, did not take other antibiotics that may confound with results, and had an assessment of outcome during the protocol-defined window.
The microbiologically evaluable (ME) population consisted of the CE population who had at least one Gram-positive pathogen at baseline. The microbiological MITT (micro-MITT) population was composed of MITT patients with at least one Gram-positive bacterial pathogen identified from blood culture or the sample obtained from the cSSTI site at baseline.

Outcome measurement

Clinical efficacy endpoints were defined as the infection was fully resolved, and no further antimicrobial treatment was required at the end of treatment and post-treatment evaluation (PTE) in the MITT and CE populations [24]. The microbiological response was determined in the ME and micro-MITT population as eradication (absence of original baseline pathogen) or presumed eradication (no source specimen to culture in a subject assessed with a clinical success) of baseline pathogens [24]. Adverse events (AEs) were defined as AEs that emerged during or after administration, increased in severity, or were associated with the study drug during the study period.

Data analysis

Review Manager 5.3 software was used for meta-analysis. Qualitative data were expressed by odds ratio (OR) and its 95% Cl, and quantitative data were described by mean difference (MD) and its 95% Cl. The heterogeneity test was evaluated by the Cochrane I2 statistics. When P < 0.10 or I2 > 50%, it was considered that there was statistical heterogeneity among the studies, and the random effects model was used for meta-analysis; otherwise, the fixed effects model was used. P < 0.05 was considered statistically significant.
This meta-analysis was registered in the International Prospective Register of Systematic Reviews (PROSPERO: CRD 42022362152).

Results

Study selection and characteristics

A total of 768 references were obtained (PubMed: 215, Embase: 261, Cochrane: 58, Web of Science: 223, Clinical Trial: 11), while 446 duplicates were excluded. By reading topics and abstracts, 302 references were excluded. Finally, 20 studies remained. By reading the full text, four RCTs [2528] consisting of 1,757 patients were included. Figure 1 illustrates the process of literature search and screening. Table 1 shows the essential characteristics, and all compared drugs included in this meta-analysis were LZD. Moreover, all included studies were high-quality RCTs, and Fig. 2 shows the risk chart of bias.
Table 1
Basic information of included studies
No
Author
Year
Sites
Start and End
Treatment Plan
Subjects
Pathogen Detected at Baseline
OMC
LZD
OMC
LZD
OMC
LZD
1
Gary J. Noel
2012
11 sites in the USA
2007.07–2008.01
100 mg (i.v.) q24h/200 mg (p.o.) q24h
a600 mg (i.v.) q12h/600 mg (p.o.) q12h
118
116
G+ pathogen:
S. aureus: 72
MRSA: 44
MSSA:\
G+ bacterium other than S. aureus: 3
G+ Pathogen:
S. aureus: 55
MRSA: 32
MSSA:\
G+ bacterium other than S. aureus: 7
2
NCT00865280
\
7 sites in the USA
2009.04–2010.04
100 mg (i.v.) q24h/300 mg (p.o.) q24h
b600 mg (i.v.) q12h/600 mg (p.o.) q12h
70
73
\
\
3
William O’Riordan (OASIS-1)
2019
55 sites in the USA, Peru, South Africa, and Europe
2015.06–2016.05
100 mg (i.v.) q12h for 1 day + 100 mg (i.v.) q24h for 2 days + 100 mg (i.v.) or 300 mg (p.o.) q24h
600 mg (i.v.) q12h for 3 days + 600 mg (i.v.) or 600 mg (p.o.) q12h
323
322
Mono-G+: 156
Poly-G+: 31
Mixed: 41
G+ Pathogen:
S. aureus: 156
MRSA: 69
MSSA: 88
S. pyogenes: 11
Streptococcus anginosis group: 47
E. faecalis: 10
Mono-G+: 171
Poly-G+: 27
Mixed: 29
G+ Pathogen:
S. aureus: 151
MRSA: 50
MSSA: 102
S. pyogenes: 18
Streptococcus anginosis group: 37
E. faecalis: 13
4
William O’Riordan (OASIS-2)
2019
33 sites in the USA
2016.08–2017.06
450 mg (p.o.) q24h for 2 days + 300 mg (p.o.) q24h
600 mg (p.o.) q12h for 2 days + 600 mg (p.o.) q12h
368
367
Mono-G+: 184
Poly-G+: 60
Mixed: 32
G+ Pathogen:
S. aureus: 220
MRSA: 104
MSSA: 120
S. pyogenes: 29
Streptococcus anginosis group: 57
E. faecalis: 7
Mono-G+: 212
Poly-G+: 37
Mixed: 38
G+ Pathogen:
S. aureus: 233
MRSA: 107
MSSA: 130
S. pyogenes: 16
Streptococcus anginosis group: 45
E. faecalis: 10
No
Author
Note
Quality score
Infection types
Size of lesion
Drainage Procedures
Cellulitis/Erysipelas
Major abscess
Wound infection
1
Gary J. Noel
cOMC 8 (7%) LZD 10 (9%)
cOMC 73 (66%) LZD 72 (67%)
cOMC 21 (19%) LZD 17 (16%)
gAbscesses: OMC 12.6 cm; LZD 11.3 cm Wound infection: 13.6 cm
Incision and drainage of the infected site in all abscess patients is permitted before or within 24 h after treatment
4
2
NCT00865280
dOMC 39 (57%) LZD 45 (63%)
\
dOMC 13 (19%) LZD 13 (18%)
\
\
3
3
William O’Riordan (OASIS-1)
eOMC 123 (39%) LZD 118 (38%)
eOMC 91 (29%) LZD 89 (29%)
eOMC 102 (32%) LZD 104 (33%)
hOMC 299.5 (77 ~ 4100) cm2; LZD 315.0 (88 ~ 6739) cm2
Patients with major abscesses had allowed drainage procedures before (or within 48 h following) the first dose
5
4
William O’Riordan (OASIS-2)
fOMC 86 (24%) LZD 84 (23%)
fOMC 64 (18%) LZD 62 (17%)
fOMC 210 (58%) LZD 214 (59%)
hOMC 322 (198 ~ 495) cm2; LZD 294 (190 ~ 462) cm2
Patients with major abscesses had allowed drainage procedures before (or within 48 h following) the first dose
5
S. aureus Staphylococcus aureus, MRSA meticillin-resistant S. aureus, MSSA meticillin-susceptible S. aureus, S. pyogenes, Streptococcus pyogenes, Streptococcus anginosis group: includes S. anginosus, Streptococcus intermedius, and Streptococcus constellatus, E.faecalis Enterococcus faecalis
Mono-G+: monomicrobial Gram-positive infection; Poly-G+: polymicrobial Gram-positive infection; Mixed: polymicrobial Gram-positive and Gram-negative infection
awith or without aztreonam
b-with moxifloxacin
cin intent-to-treat population, n = 111 and 108
din clinically evaluable population, n = 68 and 72
ein modified intent-to-treat population, n = 316 and 311
fin modified intent-to-treat population, n = 360 and 360
gmean maximum dimension
hmean maximal linear dimension

Clinical efficacy

The results of the meta-analysis showed that the efficacy of OMC was not inferior to LZD in both the MITT population (OR: 1.24, 95% Cl: [0.93, 1.66], P = 0.15) and CE population (OR: 1.92, 95% Cl: [0.94, 3.92], P = 0.07) (Fig. 3).

Microbiological response

Two studies reported microbiological responses in the micro-MITT and ME populations under the same baseline. Generally, the microbiological response of the OMC group was numerically higher compared with the LZD group (micro-MITT population: OR: 1.27, 95% Cl: [0.92, 1.76], P = 0.14; ME population: OR: 1.74, 95% Cl: [0.81, 3.74], P = 0.16) (Fig. 4).
The microbiological response of OMC in the micro-MITT population at PTE was not inferior to LZD among patients with monomicrobial Gram-positive infection (OR: 1.38, 95% Cl: [0.92, 2.06], P = 0.11), polymicrobial Gram-positive infection (OR: 1.26, 95% Cl: [0.59, 2.68], P = 0.55), and polymicrobial mixed Gram-positive and Gram-negative infection (OR: 1.03, 95% Cl: [0.45, 2.35], P = 0.94) (Fig. 5A). Next, we compared the ability of OMC and LZD to eradicate common Gram-positive bacteria that caused cSSTIs. The results suggested that OMC was not inferior to LZD in eradicating Gram-positive pathogens (S. aureus: OR: 1.12, 95% Cl: [0.77, 1.63], P = 0.55; S. pyogenes: OR: 1.06, 95% Cl: [0.38, 3.00], P = 0.91; S. anginosus group: OR:1.64, 95% Cl: [0.83, 3.27], P = 0.16; E. faecalis: OR: 2.47, 95% Cl: [0.36, 16.97], P = 0.36) (Fig. 5B).

Safety

There was no significant difference in treatment-emergent adverse events (TEAEs) (OR: 1.17, 95% Cl: [0.74, 1.86], P = 0.50), serious AEs (OR: 1.40, 95% Cl: [0.72, 2.70], P = 0.32), treatment discontinuation for AEs (OR: 1.24, 95% Cl: [0.59, 2.63], P = 0.57), and treatment related TEAEs (OR: 1.29, 95% Cl: [0.56, 3.00], P = 0.55) between OMC and LZD (Fig. 6). In addition, no significant difference was found in nausea (OR: 1.88, 95% Cl: [0.79, 4.44], P = 0.15), vomiting (OR: 1.53, 95% Cl: [0.46, 5.09], P = 0.49), diarrhea (OR: 0.35, 95% Cl: [0.09, 1.43], P = 0.14), and blood and lymphatic system disorders (OR: 0.63, 95% Cl: [0.24, 1.64], P = 0.34) (Fig. 7).
In terms of mortality, there was no significant difference between OMC and LZD (OR: 0.76, 95% Cl: [0.17, 3.40], P = 0.72) (Fig. 8).

Discussion

Our meta-analysis consisting of four RCTs showed that the clinical efficacy of OMC was as good as LZD. A previous meta-analysis has evaluated the efficacy and safety of OMC in the treatment of acute bacterial infection, which includes three studies on cSSTIs and one study on CABP. The results show that the clinical efficacy of OMC is numerically higher compared with the comparator antibiotics, and OMC shows similar clinical efficacy to LZD in treating cSSTIs [29]. However, the study did not appear to unify the criteria for the MITT and CE populations included in the study (two studies excluded patients with Gram-negative bacteria only; one study did not and added aztreonam against Gram-negative bacteria). After intravenous injection of OMC in rats, the skin-to-blood concentration ratio is increased by 6.6 times [30]. In healthy subjects, the steady-state distribution volumes of OMC are 190 ~ 204 L [17, 18], and the plasma protein binding rate is about 21% [10]. In contrast, the steady-state distribution volumes of LZD are 40 ~ 50 L [31], and the plasma protein binding rate is about 31% and has poor penetration into adipose tissue [32]. These findings may indicate that OMC has a high rate of tissue penetration than LZD.
The microbiological response of OMC was numerically higher than LZD, which might be related to its lower MIC. S. aureus is the most common pathogen causing cSSTIs, accounting for 81% of isolated pathogens [4]. Compared with LZD, OMC has a lower MIC against common Gram-positive bacteria in vitro. For example, for S. aureus (including MRSA and MSSA), S. pyogenes, and E. faecalis, the MICs are ≤ 0.015 ~ 2 mg/L for OMC and ≤ 0.12 ~ 8 for LZD [33]. The tissue distribution concentration of OMC and LZD is 378 mg/L and 15.5 mg/L, respectively [30, 34]. Free concentrations of OMC and LZD at the skin and soft tissue sites could cover common Gram-positive pathogen bacteria.
Although Gram-positive bacteria are the most common pathogens, Gram-negative bacteria or mixed infections still account for about 12 ~ 21% of severe skin infections [6, 35]. Among Gram-negative pathogens, Enterobacteriaceae is the most common isolated bacteria [36]. OMC shows activity against Gram-negative bacteria in vitro, such as Escherichia coli (E. coli), Klebsiella pneumoniae (K. pneumoniae), and Haemophilus influenzae, with MIC50/90 values of 0.5/2, 1/4, and 0.5/1 mg/L, respectively [33]. In recent years, multidrug-resistant (MDR)/extensively drug-resistant (XDR) bacteria have also been isolated from cSSTIs (such as MDR/XDR Enterobacteriaceae) [35]. A multicenter, observational study involving nine patients has found that oral OMC (450 mg loading dose plus 300 mg maintenance dose) is effective against MDR E. coli and K. pneumoniae-induced bone/joint or intra-abdominal infection [37]. Since LZD is not active against Gram-negative pathogens [38, 39], patients with sole Gram-negative pathogenic bacterial infections were excluded from RCTs. Therefore, we could not analyze how effective OMC was in treating cSSTIs caused by Gram-negative pathogens. In this analysis that integrated OASIS-1 and OASIS-2, the microbiological response of mixed infection with Gram-negative strains of OMC and LZD was similar in the micro-MITT population at PTE, and this finding might be attributed to the small number of cases. Considering that OMC is effective against Gram-negative bacteria or mixed infections, it might be preferred for treating cSSTIs.
Regarding safety, the incidence of adverse drug reactions in OMC was similar to that in LZD, and most of them were transient AEs. Unsurprisingly, since OMC is structurally similar to tetracycline antibiotics, the most common AEs are gastrointestinal events [40]. It was observed that the incidence of nausea and vomiting in the OMC group was slightly higher compared with the LZD group in this meta-analysis. Some studies have shown that the gastrointestinal AEs of OMC are dose-dependent in healthy and diseased subjects [41, 42]. A higher incidence of gastrointestinal AEs in the OASIS-2 study with a higher dosage applied is also observed. Studies have shown that oral OMC after meals can reduce gastrointestinal AEs. However, compared with fasting, taking meals 2 to 4 h before the administration will reduce the bioavailability of OMC [43]. Therefore, to obtain an excellent therapeutic effect, oral OMC should be taken on a fasting state and avoided in combination with dairy products. Some AEs associated with the long-term use of LZD include myelosuppression, peripheral and optic neuropathy, serotonin syndrome, and so on [44]. Only a trend of a higher number of AEs in the hematological system was observed in the present meta-analysis (1.60% in LZD vs. 1.01% in OMC). This finding might be related to the short duration of treatment (the treatment time was 7 ~ 14 days). Studies have shown that the hematological system response of LZD is increased with the prolongation of medication time (treatment duration ≤ 14 days reported 1.9%; 5.1% for 15 ~ 28 days; 7.4% for > 28 days) [45]. Although the recommended duration of cSSTIs treatment is 7 to 10 days, the treatment time should be extended if the infection is not improved during treatment [46]. Therefore, it is still necessary to pay attention to the hematological toxicity of LZD.
This study has several limitations. Firstly, the study failed to demonstrate that OMC was superior to LZD against Gram-positive infection. The ME population included a CE population with at least one Gram-positive pathogen at baseline, but there was no significant difference between OMC and LZD. Moreover, although no restrictions were set on the types of control drugs, only LZD was included in this meta-analysis. Comparisons between OMC and other antibiotics, such as vancomycin, for the treatment of cSSTIs could not be evaluated here. Lastly, because LZD was ineffective against Gram-negative bacteria, we could not assess the efficacy of OMC against infections caused by Gram-negative bacteria.

Conclusions

This meta-analysis showed that OMC was as good as LZD regarding clinical efficacy and microbiological response, and has a similar safety profile. Therefore, OMC might be a promising option for treating cSSTIs in adult patients. However, we need to further study the hepatorenal impact and low immune function populations before applying OMC in Gram-negative or mixed cSSTIs.

Acknowledgements

Not applicable.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
2.
Zurück zum Zitat Stevens DL, Bisno AL, Chambers HF, et al. Practice guidelines for the diagnosis and management of skin and soft tissue infections: 2014 update by the infectious diseases society of America. Clin Infect Dis. 2014;59(2):147–59.PubMedCrossRef Stevens DL, Bisno AL, Chambers HF, et al. Practice guidelines for the diagnosis and management of skin and soft tissue infections: 2014 update by the infectious diseases society of America. Clin Infect Dis. 2014;59(2):147–59.PubMedCrossRef
3.
Zurück zum Zitat Sartelli M, Guirao X, Hardcastle TC, et al. 2018 WSES/SIS-E consensus conference: recommendations for the management of skin and soft-tissue infections. World J Emerg Surg. 2018;13:58.PubMedPubMedCentralCrossRef Sartelli M, Guirao X, Hardcastle TC, et al. 2018 WSES/SIS-E consensus conference: recommendations for the management of skin and soft-tissue infections. World J Emerg Surg. 2018;13:58.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Sartelli M, Coccolini F, Kluger Y, et al. WSES/GAIS/WSIS/SIS-E/AAST global clinical pathways for patients with skin and soft tissue infections. World J Emerg Surg. 2022;17(1):3.PubMedPubMedCentralCrossRef Sartelli M, Coccolini F, Kluger Y, et al. WSES/GAIS/WSIS/SIS-E/AAST global clinical pathways for patients with skin and soft tissue infections. World J Emerg Surg. 2022;17(1):3.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Duane TM, Huston JM, Collom M, et al. Surgical Infection Society 2020 updated guidelines on the management of complicated skin and soft tissue infections. Surg Infect (Larchmt). 2021;22(4):383–99.PubMedCrossRef Duane TM, Huston JM, Collom M, et al. Surgical Infection Society 2020 updated guidelines on the management of complicated skin and soft tissue infections. Surg Infect (Larchmt). 2021;22(4):383–99.PubMedCrossRef
6.
Zurück zum Zitat Golan Y. Current treatment options for acute skin and skin-structure infections. Clin Infect Dis. 2019;68(Suppl 3):206-S212.CrossRef Golan Y. Current treatment options for acute skin and skin-structure infections. Clin Infect Dis. 2019;68(Suppl 3):206-S212.CrossRef
7.
Zurück zum Zitat Pollack CV Jr, Amin A, Ford WT Jr, et al. Acute bacterial skin and skin structure infections (ABSSSI): practice guidelines for management and care transitions in the emergency department and hospital. J Emerg Med. 2015;48(4):508–19.PubMedCrossRef Pollack CV Jr, Amin A, Ford WT Jr, et al. Acute bacterial skin and skin structure infections (ABSSSI): practice guidelines for management and care transitions in the emergency department and hospital. J Emerg Med. 2015;48(4):508–19.PubMedCrossRef
8.
Zurück zum Zitat Esposito S, Bassetti M, Concia E, et al. Diagnosis and management of skin and soft-tissue infections (SSTI). A literature review and consensus statement: an update. J Chemother. 2017;29(4):197–214.PubMedCrossRef Esposito S, Bassetti M, Concia E, et al. Diagnosis and management of skin and soft-tissue infections (SSTI). A literature review and consensus statement: an update. J Chemother. 2017;29(4):197–214.PubMedCrossRef
10.
Zurück zum Zitat Watkins RR, Deresinski S. Omadacycline: a novel tetracycline derivative with oral and intravenous formulations. Clin Infect Dis. 2019;69(5):890–6.PubMedCrossRef Watkins RR, Deresinski S. Omadacycline: a novel tetracycline derivative with oral and intravenous formulations. Clin Infect Dis. 2019;69(5):890–6.PubMedCrossRef
11.
Zurück zum Zitat Bidell MR, Lodise TP. Use of oral tetracyclines in the treatment of adult outpatients with skin and skin structure infections: focus on doxycycline, minocycline, and omadacycline. Pharmacotherapy. 2021;41(11):915–31.PubMedPubMedCentralCrossRef Bidell MR, Lodise TP. Use of oral tetracyclines in the treatment of adult outpatients with skin and skin structure infections: focus on doxycycline, minocycline, and omadacycline. Pharmacotherapy. 2021;41(11):915–31.PubMedPubMedCentralCrossRef
12.
13.
Zurück zum Zitat Huband MD, Pfaller MA, Shortridge D, et al. Surveillance of omadacycline activity tested against clinical isolates from the United States and Europe: results from the SENTRY Antimicrobial Surveillance Programme, 2017. J Glob Antimicrob Resist. 2019;19:56–63.PubMedCrossRef Huband MD, Pfaller MA, Shortridge D, et al. Surveillance of omadacycline activity tested against clinical isolates from the United States and Europe: results from the SENTRY Antimicrobial Surveillance Programme, 2017. J Glob Antimicrob Resist. 2019;19:56–63.PubMedCrossRef
14.
Zurück zum Zitat Karlowsky JA, Steenbergen J, Zhanel GG. Microbiology and preclinical review of omadacycline. Clin Infect Dis. 2019;69(Suppl 1):6-S15.CrossRef Karlowsky JA, Steenbergen J, Zhanel GG. Microbiology and preclinical review of omadacycline. Clin Infect Dis. 2019;69(Suppl 1):6-S15.CrossRef
15.
Zurück zum Zitat Honeyman L, Ismail M, Nelson ML, et al. Structure-activity relationship of the aminomethylcyclines and the discovery of omadacycline. Antimicrob Agents Chemother. 2015;59(11):7044–53.PubMedPubMedCentralCrossRef Honeyman L, Ismail M, Nelson ML, et al. Structure-activity relationship of the aminomethylcyclines and the discovery of omadacycline. Antimicrob Agents Chemother. 2015;59(11):7044–53.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Sun H, Ting L, Machineni S, et al. Randomized, open-label study of the pharmacokinetics and safety of oral and intravenous administration of omadacycline to healthy subjects. Antimicrob Agents Chemother. 2016;60(12):7431–5.PubMedPubMedCentralCrossRef Sun H, Ting L, Machineni S, et al. Randomized, open-label study of the pharmacokinetics and safety of oral and intravenous administration of omadacycline to healthy subjects. Antimicrob Agents Chemother. 2016;60(12):7431–5.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Gotfried MHHK, Garrity-Ryan L, et al. Comparison of omadacycline and tigecycline pharmacokinetics in the plasma, epithelial lining fluid, and alveolar cells of healthy adult subjects. Antimicrob Agents Chemother. 2017;61(9):e01135-01117.PubMedPubMedCentralCrossRef Gotfried MHHK, Garrity-Ryan L, et al. Comparison of omadacycline and tigecycline pharmacokinetics in the plasma, epithelial lining fluid, and alveolar cells of healthy adult subjects. Antimicrob Agents Chemother. 2017;61(9):e01135-01117.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Berg JK, Tzanis E, Garrity-Ryan L, et al. Pharmacokinetics and safety of omadacycline in subjects with impaired renal function. Antimicrob Agents Chemother. 2018;62:62 (2).CrossRef Berg JK, Tzanis E, Garrity-Ryan L, et al. Pharmacokinetics and safety of omadacycline in subjects with impaired renal function. Antimicrob Agents Chemother. 2018;62:62 (2).CrossRef
19.
Zurück zum Zitat Rodvold KA, Burgos RM, Tan X, et al. Omadacycline: a review of the clinical pharmacokinetics and Pharmacodynamics. Clin Pharmacokinet. 2020;59(4):409–25.PubMedCrossRef Rodvold KA, Burgos RM, Tan X, et al. Omadacycline: a review of the clinical pharmacokinetics and Pharmacodynamics. Clin Pharmacokinet. 2020;59(4):409–25.PubMedCrossRef
20.
Zurück zum Zitat Dougherty JA, Sucher AJ, Chahine EB, et al. Omadacycline: a new tetracycline antibiotic. Ann Pharmacother. 2019;53(5):486–500.PubMedCrossRef Dougherty JA, Sucher AJ, Chahine EB, et al. Omadacycline: a new tetracycline antibiotic. Ann Pharmacother. 2019;53(5):486–500.PubMedCrossRef
21.
Zurück zum Zitat Moher D, Shamseer L, Clarke M, et al. Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Syst Reviews. 2015;4(1):1.CrossRef Moher D, Shamseer L, Clarke M, et al. Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Syst Reviews. 2015;4(1):1.CrossRef
22.
Zurück zum Zitat Puljak L, Ramic I, Arriola Naharro C, et al. Cochrane risk of bias tool was used inadequately in the majority of non-cochrane systematic reviews. J Clin Epidemiol. 2020;123:114–9.PubMedCrossRef Puljak L, Ramic I, Arriola Naharro C, et al. Cochrane risk of bias tool was used inadequately in the majority of non-cochrane systematic reviews. J Clin Epidemiol. 2020;123:114–9.PubMedCrossRef
23.
Zurück zum Zitat Jadad AR, Moore RA, Carroll D, et al. Assessing the quality of reports of randomized clinical trials: is blinding necessary? Control Clin Trials. 1996;17(1):1–12.PubMedCrossRef Jadad AR, Moore RA, Carroll D, et al. Assessing the quality of reports of randomized clinical trials: is blinding necessary? Control Clin Trials. 1996;17(1):1–12.PubMedCrossRef
26.
Zurück zum Zitat Noel GJ, Draper MP, Hait H, et al. A randomized, evaluator-blind, phase 2 study comparing the safety and efficacy of omadacycline to those of linezolid for treatment of complicated skin and skin structure infections. Antimicrob Agents Chemother. 2012;56(11):5650–4.PubMedPubMedCentralCrossRef Noel GJ, Draper MP, Hait H, et al. A randomized, evaluator-blind, phase 2 study comparing the safety and efficacy of omadacycline to those of linezolid for treatment of complicated skin and skin structure infections. Antimicrob Agents Chemother. 2012;56(11):5650–4.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat O’Riordan W, Green S, Overcash JS, et al. Omadacycline for acute bacterial skin and skin-structure infections. N Engl J Med. 2019;380(6):528–38.PubMedCrossRef O’Riordan W, Green S, Overcash JS, et al. Omadacycline for acute bacterial skin and skin-structure infections. N Engl J Med. 2019;380(6):528–38.PubMedCrossRef
28.
Zurück zum Zitat O’Riordan W, Cardenas C, Shin E, et al. Once-daily oral omadacycline versus twice-daily oral linezolid for acute bacterial skin and skin structure infections (OASIS-2): a phase 3, double-blind, multicentre, randomised, controlled, non-inferiority trial. Lancet Infect Dis. 2019;19(10):1080–90.PubMedCrossRef O’Riordan W, Cardenas C, Shin E, et al. Once-daily oral omadacycline versus twice-daily oral linezolid for acute bacterial skin and skin structure infections (OASIS-2): a phase 3, double-blind, multicentre, randomised, controlled, non-inferiority trial. Lancet Infect Dis. 2019;19(10):1080–90.PubMedCrossRef
29.
Zurück zum Zitat Lan SH, Chang SP, Lai CC, et al. The efficacy and safety of omadacycline in treatment of acute bacterial infection: a systemic review and meta-analysis of randomized controlled trials. Medicine (Baltimore). 2019;98(51):e18426.PubMedCrossRef Lan SH, Chang SP, Lai CC, et al. The efficacy and safety of omadacycline in treatment of acute bacterial infection: a systemic review and meta-analysis of randomized controlled trials. Medicine (Baltimore). 2019;98(51):e18426.PubMedCrossRef
30.
Zurück zum Zitat Lin W, Flarakos J, Du Y, et al. Pharmacokinetics, distribution, metabolism, and excretion of omadacycline following a single intravenous or oral dose of 14 C-omadacycline in rats. Antimicrob Agents Chemother. 2017;61(1):e01784-16.PubMedCrossRef Lin W, Flarakos J, Du Y, et al. Pharmacokinetics, distribution, metabolism, and excretion of omadacycline following a single intravenous or oral dose of 14 C-omadacycline in rats. Antimicrob Agents Chemother. 2017;61(1):e01784-16.PubMedCrossRef
31.
Zurück zum Zitat Stein GE, Wells EM. The importance of tissue penetration in achieving successful antimicrobial treatment of nosocomial pneumonia and complicated skin and soft-tissue infections caused by methicillin-resistant Staphylococcus aureus: Vancomycin and linezolid. Curr Med Res Opin. 2010;26(3):571–88.PubMedCrossRef Stein GE, Wells EM. The importance of tissue penetration in achieving successful antimicrobial treatment of nosocomial pneumonia and complicated skin and soft-tissue infections caused by methicillin-resistant Staphylococcus aureus: Vancomycin and linezolid. Curr Med Res Opin. 2010;26(3):571–88.PubMedCrossRef
32.
Zurück zum Zitat Rao GG, Konicki R, Cattaneo D, et al. Therapeutic drug monitoring can improve Linezolid Dosing regimens in current clinical practice: a review of linezolid pharmacokinetics and pharmacodynamics. Ther Drug Monit. 2020;42(1):83–92.PubMedCrossRef Rao GG, Konicki R, Cattaneo D, et al. Therapeutic drug monitoring can improve Linezolid Dosing regimens in current clinical practice: a review of linezolid pharmacokinetics and pharmacodynamics. Ther Drug Monit. 2020;42(1):83–92.PubMedCrossRef
33.
Zurück zum Zitat Pfaller MA, Huband MD, Shortridge D, et al. Surveillance of omadacycline activity tested against clinical isolates from the USA: report from the SENTRY Antimicrobial Surveillance Program, 2019. J Glob Antimicrob Resist. 2021;27:337–51.PubMedCrossRef Pfaller MA, Huband MD, Shortridge D, et al. Surveillance of omadacycline activity tested against clinical isolates from the USA: report from the SENTRY Antimicrobial Surveillance Program, 2019. J Glob Antimicrob Resist. 2021;27:337–51.PubMedCrossRef
34.
Zurück zum Zitat Traunmuller F, Schintler MV, Spendel S, et al. Linezolid concentrations in infected soft tissue and bone following repetitive doses in diabetic patients with bacterial foot infections. Int J Antimicrob Agents. 2010;36(1):84–6.PubMedCrossRef Traunmuller F, Schintler MV, Spendel S, et al. Linezolid concentrations in infected soft tissue and bone following repetitive doses in diabetic patients with bacterial foot infections. Int J Antimicrob Agents. 2010;36(1):84–6.PubMedCrossRef
35.
Zurück zum Zitat Jabbour JF, Kanj SS. Gram-negative skin and soft tissue infections. Infect Dis Clin North Am. 2021;35(1):157–67.PubMedCrossRef Jabbour JF, Kanj SS. Gram-negative skin and soft tissue infections. Infect Dis Clin North Am. 2021;35(1):157–67.PubMedCrossRef
36.
Zurück zum Zitat Falcone M, Concia E, Giusti M, et al. Acute bacterial skin and skin structure infections in internal medicine wards: old and new drugs. Intern Emerg Med. 2016;11(5):637–48.PubMedCrossRef Falcone M, Concia E, Giusti M, et al. Acute bacterial skin and skin structure infections in internal medicine wards: old and new drugs. Intern Emerg Med. 2016;11(5):637–48.PubMedCrossRef
37.
Zurück zum Zitat Morrisette T, Alosaimy S, Lagnf AM, et al. Real-world, multicenter case series of patients treated with oral omadacycline for resistant gram-negative pathogens. Infect Dis Ther. 2022;11(4):1715–23.PubMedPubMedCentral Morrisette T, Alosaimy S, Lagnf AM, et al. Real-world, multicenter case series of patients treated with oral omadacycline for resistant gram-negative pathogens. Infect Dis Ther. 2022;11(4):1715–23.PubMedPubMedCentral
39.
Zurück zum Zitat Livermore DM. Linezolid in vitro: mechanism and antibacterial spectrum. J Antimicrob Chemother. 2003;51(Suppl 2):ii9-16.PubMed Livermore DM. Linezolid in vitro: mechanism and antibacterial spectrum. J Antimicrob Chemother. 2003;51(Suppl 2):ii9-16.PubMed
41.
Zurück zum Zitat Bundrant LATE, Garrity-Ryan L, et al. Safety and pharmacokinetics of the aminomethylcycline antibiotic omadacycline administered to healthy subjects in oral multiple-dose regimens. Antimicrob Agents Chemother. 2018;62(2):e01487-01417.PubMedPubMedCentralCrossRef Bundrant LATE, Garrity-Ryan L, et al. Safety and pharmacokinetics of the aminomethylcycline antibiotic omadacycline administered to healthy subjects in oral multiple-dose regimens. Antimicrob Agents Chemother. 2018;62(2):e01487-01417.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Overcash JS, Bhiwandi P, Garrity-Ryan L, et al. Pharmacokinetics, safety, and clinical outcomes of omadacycline in women with cystitis: results from a phase 1b study. Antimicrob Agents Chemother. 2019;63(5):e02083.PubMedPubMedCentralCrossRef Overcash JS, Bhiwandi P, Garrity-Ryan L, et al. Pharmacokinetics, safety, and clinical outcomes of omadacycline in women with cystitis: results from a phase 1b study. Antimicrob Agents Chemother. 2019;63(5):e02083.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Tzanis E, Manley A, Villano S, et al. Effect of food on the bioavailability of omadacycline in healthy participants. J Clin Pharmacol. 2017;57(3):321–7.PubMedCrossRef Tzanis E, Manley A, Villano S, et al. Effect of food on the bioavailability of omadacycline in healthy participants. J Clin Pharmacol. 2017;57(3):321–7.PubMedCrossRef
44.
Zurück zum Zitat Hashemian SMR, Farhadi T, Ganjparvar M. Linezolid: a review of its properties, function, and use in critical care. Drug Des Devel Ther. 2018;12:1759–67.PubMedPubMedCentralCrossRef Hashemian SMR, Farhadi T, Ganjparvar M. Linezolid: a review of its properties, function, and use in critical care. Drug Des Devel Ther. 2018;12:1759–67.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Shaw KJ, Barbachyn MR. The oxazolidinones: past, present, and future. Ann N Y Acad Sci. 2011;1241:48–70.ADSPubMedCrossRef Shaw KJ, Barbachyn MR. The oxazolidinones: past, present, and future. Ann N Y Acad Sci. 2011;1241:48–70.ADSPubMedCrossRef
46.
Zurück zum Zitat Stevens DL, Bisno AL, Chambers HF, et al. Practice guidelines for the diagnosis and management of skin and soft tissue infections: 2014 update by the Infectious Diseases Society of America. Clin Infect Dis. 2014;59(2):e10-52.PubMedCrossRef Stevens DL, Bisno AL, Chambers HF, et al. Practice guidelines for the diagnosis and management of skin and soft tissue infections: 2014 update by the Infectious Diseases Society of America. Clin Infect Dis. 2014;59(2):e10-52.PubMedCrossRef
Metadaten
Titel
Efficacy and safety of omadacycline for treating complicated skin and soft tissue infections: a meta-analysis of randomized controlled trials
verfasst von
Wenxin Liang
Hong Yin
Huiling Chen
Juan Xu
Yun Cai
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
BMC Infectious Diseases / Ausgabe 1/2024
Elektronische ISSN: 1471-2334
DOI
https://doi.org/10.1186/s12879-024-09097-3

Weitere Artikel der Ausgabe 1/2024

BMC Infectious Diseases 1/2024 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Neue S3-Leitlinie zur unkomplizierten Zystitis: Auf Antibiotika verzichten?

15.05.2024 Harnwegsinfektionen Nachrichten

Welche Antibiotika darf man bei unkomplizierter Zystitis verwenden und wovon sollte man die Finger lassen? Welche pflanzlichen Präparate können helfen? Was taugt der zugelassene Impfstoff? Antworten vom Koordinator der frisch überarbeiteten S3-Leitlinie, Prof. Florian Wagenlehner.

Schadet Ärger den Gefäßen?

14.05.2024 Arteriosklerose Nachrichten

In einer Studie aus New York wirkte sich Ärger kurzfristig deutlich negativ auf die Endothelfunktion gesunder Probanden aus. Möglicherweise hat dies Einfluss auf die kardiovaskuläre Gesundheit.

Intervallfasten zur Regeneration des Herzmuskels?

14.05.2024 Herzinfarkt Nachrichten

Die Nahrungsaufnahme auf wenige Stunden am Tag zu beschränken, hat möglicherweise einen günstigen Einfluss auf die Prognose nach akutem ST-Hebungsinfarkt. Darauf deutet eine Studie an der Uniklinik in Halle an der Saale hin.

Klimaschutz beginnt bei der Wahl des Inhalators

14.05.2024 Klimawandel Podcast

Auch kleine Entscheidungen im Alltag einer Praxis können einen großen Beitrag zum Klimaschutz leisten. Die neue Leitlinie zur "klimabewussten Verordnung von Inhalativa" geht mit gutem Beispiel voran, denn der Wechsel vom klimaschädlichen Dosieraerosol zum Pulverinhalator spart viele Tonnen CO2. Leitlinienautor PD Dr. Guido Schmiemann erklärt, warum nicht nur die Umwelt, sondern auch Patientinnen und Patienten davon profitieren.

Zeitschrift für Allgemeinmedizin, DEGAM

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.