Next Article in Journal
Prognostic Relevance of Global Myocardial Work Index in Patients with Moderate Aortic Valve Stenosis
Previous Article in Journal
Striving for Perfection: How Stable Is Orthodontic Treatment When Excellent Outcomes Are Achieved? A 9-Year Post-Treatment Retrospective Study
Previous Article in Special Issue
Practical Considerations for Cardiac Electronic Devices Reimplantation Following Transvenous Lead Extraction Due to Related Endocarditis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Antimicrobials and New Therapy Strategies for Endocarditis: Weapons That Should Be Defended

by
Alessandra Oliva
*,
Francesco Cogliati Dezza
,
Francesca Cancelli
,
Ambrogio Curtolo
,
Antonio Falletta
,
Lorenzo Volpicelli
and
Mario Venditti
Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(24), 7693; https://doi.org/10.3390/jcm12247693
Submission received: 5 October 2023 / Revised: 22 November 2023 / Accepted: 30 November 2023 / Published: 14 December 2023
(This article belongs to the Special Issue Multidisciplinary Endocarditis Perspectives)

Abstract

:
The overall low-quality evidence concerning the clinical benefits of different antibiotic regimens for the treatment of infective endocarditis (IE), which has made it difficult to strongly support or reject any regimen of antibiotic therapy, has led to a discrepancy between the available guidelines and clinical practice. In this complex scenario, very recently published guidelines have attempted to fill this gap. Indeed, in recent years several antimicrobials have entered the market, including ceftobiprole, ceftaroline, and the long-acting lipoglycopeptides dalbavancin and oritavancin. Despite being approved for different indications, real-world data on their use for the treatment of IE, alone or in combination, has accumulated over time. Furthermore, an old antibiotic, fosfomycin, has gained renewed interest for the treatment of complicated infections such as IE. In this narrative review, we focused on new antimicrobials and therapeutic strategies that we believe may provide important contributions to the advancement of Gram-positive IE treatment, providing a summary of the current in vitro, in vivo, and clinical evidence supporting their use in clinical practice.

1. Introduction

Infective endocarditis (IE) is a potentially lethal disease that always poses new diagnostic and therapeutic challenges. The yearly incidence is about 3–10 cases per 100,000 people, with an overall mortality of about 30% [1]. In 2019, the estimated incidence of IE was 13.8 cases per 100,000 subjects per year, and IE accounted for over 66,000 deaths worldwide [2]. The aetiological agents of IE can be Gram-positive or Gram-negative bacteria or, less frequently, fungi. Among them, Gram-positive staphylococci, streptococci, and enterococci represent 80–90% of all IE causes [3].
Notably, 2023 has been an incredible and singular year for scientific advancements in IE management, witnessing the proposal of new revised Duke criteria to help diagnose endocarditis [4] and the recent publication of the new official European guidelines for IE that update the old version published eight years ago [5,6].
Between the publication of the 2015 guidelines and the new ones, new antibiotic molecules such as ceftaroline, ceftobiprole, dalbavancin, and oritavancin were approved by the Food and Drug Administration (FDA) and the European Medicine Agency (EMA) to meet the needs of tailored therapy and, accordingly, new antibiotic strategies were investigated. Indeed, despite being approved for indications other than IE, real-world data on their use, alone or in combination, for the treatment of IE has accumulated over time, providing clinical evidence on their possible therapeutic benefits over traditional regimens [7,8,9,10,11].
Furthermore, these molecules are characterised by high bactericidal activity towards the majority of microorganisms that commonly cause IE and, most importantly, exhibit a high safety profile in comparison with glycopeptides, which still represent the recommended option for methicillin-resistant Staphylococci. Fosfomycin, an old antibiotic discovered in 1969, has gained renewed interest in this setting thanks to (i) its broad activity against both Gram-positive and Gram-negative pathogens, including resistant ones, (ii) its high anti-biofilm activity, and (iii) its ability to synergise with several antimicrobials.
After the publication of the 2015 guidelines, the only relevant published randomised clinical trial (BACSARM) on IE treatment explored the combination of daptomycin and fosfomycin for the treatment of S. aureus IE [10]; however, only a few IE cases were included (approximately 8–10% for each arm).
Given that the complexity of endocarditis renders it difficult to set up a randomised controlled clinical trial to investigate the efficacy and safety of new drugs and antibiotic strategies, the evidence from the literature comes almost exclusively from observational retrospective studies [12]. Thus, the collection of clinical evidence concerning the efficacy and tolerability of new therapeutic strategies is highly needed to address the incertitude in the most recent guidelines and in current clinical practice [5].
Furthermore, the evolution of antibiotic therapy is moving more and more towards treatment individualization and shortening. In this context, the possibility of step-down oral treatments or replacement with long-acting antibiotics represent the new therapeutic frontiers in selected and eligible patients [13,14].
To build this narrative review, we focused on new antimicrobials and therapeutic strategies that we believe may provide important contributions to the advancement of Gram-positive IE treatment, providing a summary of the current in vitro, in vivo, and clinical evidence supporting their use in the clinical practice. Some of these strategies are also recommended in the new guidelines, such as the use of a combination of daptomycin and fosfomycin or ceftaroline for the treatment of staphylococci- or enterococci-induced IE [5].
Since several other antimicrobials retain fundamental roles in the treatment of IE caused, for instance, by streptococci or susceptible E. faecalis, our review does not aim to substitute these consolidated and effective regimens with the new drugs. Rather, we attempted to summarise the potential therapeutic weapons we currently possess for the treatment of IE, such as ceftaroline, ceftobiprole, fosfomycin, dalbavancin, and oritavancin, and their most relevant therapeutic associations.
We consciously decided not to include daptomycin alone in the new therapeutic strategies. Indeed, it has earned a place as an “established treatment” for IE in recent years, a role confirmed in recently published guidelines.

2. Materials and Methods

We discussed the main topics of the narrative review in several meetings. In the first round of discussion, the following topics were identified to be addressed in this review: (i) new antimicrobials and new strategies for the management of IE caused by the most common Gram-positive pathogens, which included: ceftobiprole, ceftaroline, dalbavancin, oritavancin in monotherapy, ceftobiprole or ceftaroline in combination with daptomycin, and fosfomycin in combination with ß-lactams or daptomycin; (ii) the in vitro activity and synergism of the new antimicrobials recognised; (iii) animal studies; (iv) clinical evidence concerning the efficacy of the selected antimicrobials, alone or in combination, in the treatment of IE due to Gram-positive pathogens.
Afterwards, we retrieved scientific evidence supporting the proposals of the review by means of a PubMed-MEDLINE literature search up to July 2023. The following search strategy and key terms were adopted: “endocarditis” or “infective endocarditis” or “bacteraemia” or “bloodstream infection” or “synergism” or “in vitro activity” or “experimental model” AND the name of each single antimicrobial were searched. The antimicrobials searched were “ceftobiprole” or “ceftaroline” or “fosfomycin” or “dalbavancin” or “oritavancin”.
We selected all available categories of articles, including randomised controlled trials (RCTs), multicentre or single-centre prospective observational studies, multicentre or single-centre retrospective observational studies, case series, case reports, and in vivo/in vitro preclinical studies.
During the subsequent shared discussions, we reviewed the articles’ relevance based on the authors’ opinions and the quality of evidence, established according to a hierarchical scale of study designs. Guidelines, systematic reviews, and meta-analyses were also consulted to address our proposals.
We excluded abstracts or articles not written in English. We did not consider any timeline limitations, but we mainly focused our research on studies published in the last 10 years.
In the final round of discussion, the last version of the manuscript was approved by all authors.
The review is structured as follows: Section 3 (Section 3.1, with corresponding Table 1; Section 3.2, with corresponding Table 2; Section 3.3, with corresponding Table 3; Section 3.4, with corresponding Table 4; Section 3.5, with corresponding Table 5); Section 4, with corresponding Table 6; Section 5, with corresponding Figure 1A,B; Section 6.
Legend of color. Green: evidence supported by in vitro, animal, and preliminary clinical studies; Green–yellow lines: evidence supported by in vitro activity, animal studies, and case report series; Yellow: evidence supported by in vitro activity and animal studies but lacking clinical evidence; Yellow–red lines: poor in vitro data, no in vivo data, no clinical data; Red: absence of in vitro, animal, and clinical data and/or no drug activity.
Abbreviation. MSSA: methicillin-susceptible S. aureus; MRSA: methicillin-resistant S. aureus; CoNS: coagulase-negative Staphylococci; VISA: vancomycin-intermediate S. aureus; hVISA: heterogeneus vancomycin-intermediate S. aureus; DNS: Damptomycin unsusceptible; VR: vancomycin-resistant. CPT: ceftaroline; DAP: daptomycin; BPR: ceftobiprole; DAL: dalbavancin; ORI: oritavancin; FOS: fosfomycin

3. New Antimicrobials

3.1. Ceftobiprole

3.1.1. Mechanism of Action and Indication

Ceftobiprole (BPR) is a fifth-generation, novel broad-spectrum cephalosporin with a mechanism of action that involves binding to penicillin-binding proteins (PBPs), inhibiting cell growth and leading to bacterial cell death. A peculiarity of BPR is its ability to bind PBP2a, PBP2x, and PBP4, with increased activity against methicillin-resistant Staphylococcus aureus (MRSA), penicillin-resistant Streptococcus pneumoniae (PRP), and Enterococcus faecalis, respectively, as well as Gram-negative microorganisms, including non-extended spectrum β-lactamase (ESBL), non-AmpC and non–carbapenemase-producing Enterobacterales, and Pseudomonas aeruginosa [15,16,17,18,19,20].
Studies investigating BPR in vitro synergisms and experimental models of IE are discussed in Supplementary Material Sections S1.1 and S1.2 [18,21,22,23,24,25,26,27,28,29,30,31,32,33,34].
BPR is currently approved by the European Medicines Agency (EMA) for the treatment of community-acquired pneumonia (CAP), non-ventilator-associated hospital-acquired pneumonia (HAP), and acute bacterial skin and skin structure infections (ABSSSIs), including diabetic foot infections.

3.1.2. Clinical Evidence in Infective Endocarditis

The evidence available in the literature concerning the use of BPR in IE consists of a double-blinded, randomised, controlled non-inferiority study and observational and retrospective studies, case series, and case reports [7,35,36,37,38,39,40] (Table 1).
Table 1. Clinical studies investigating the treatment of infective endocarditis with ceftobiprole.
Table 1. Clinical studies investigating the treatment of infective endocarditis with ceftobiprole.
AuthorsStudy DesignEndpointN° Patients/
IE Type
PathogensDosage and DurationCombinationOutcomesSafety
Holland, T.L. et al., 2022 * [39]Randomised double-blind trial (ERADICATE study)
BPR vs. DAP
±Aztreonam
Clinical success
Success required survival, symptom improvement, SAB clearance, no new SAB complications, and no use of other potentially effective antibiotics
390 SAB
192 BPR vs. 198 DAP
IE 33
BPR:
20, 15 right-sided, 5 left-sided
DAP:
13, 10 right-sided, 3 left-sided
MSSA 287
MRSA 94
500 mg/6 h
up to 42 d
±AztreonamOverall clinical success:
69.8% in BPR vs. 68.7% for DAP
There were no significant differences in mortality or microbiological eradication between treatment groups
≥1 AE:
63% BPR vs. 59% DAP
Gentile, I. et al., 2023 [7]Multicentre observational and ambispective study
Mono vs. combination therapy
Clinical success:
As a composite of the clinical cure, improvement or de-scalation feasibility in 30 d FU
195,
34% mono vs. 66% combination
(pneumonia 74%; BSI 19%;
SSTI 5%; bone infection 4%)
IE 7 (4%), all combination
Polymicrobial infection (25%)
MSSA (11%)
MRSA (38%)
In IE subgroup:
2/7 MRSA;
5/7 MRCoNS
No data reportedMER 31%
In IE subgroup:
DAP 6/7 and LNZ 1/7
Overall, clinical success 79%,
microbiological cure 87%, 8 infection recurrences
In IE subgroup:
Clinical success 29%
Microbiological cure 29% (presumed eradication)
7 AE (2 rash, 2 myoclonus, 1 allergic reaction, 1 seizure, 1 CDI)
4 AE (rash or myoclonus) were BRP + DAP
Mahmoud, E. et al., 2020 [36]Case seriesN/A6 BSI (2 osteomyelitis,1 IE, 1 CLABSI, 1 SSTI, 1 pneumonia)
IE 1 NVE
MRSANo data reported on the dosage
31 d
All VAN All demonstrated microbiological and clinical cure at 14 dNo data reported
Tascini, C. et al., 2020 [37]Case series
BPR + DAP or BPR
N/AIE 12
8 PVE, 3 NVE, 1 CIED-IE
5 surgeries for vegetation size (n.3) or severe valve
disfunction with heart failure (n. 2)
9/12 previous therapy
BPR + DAP 11
BPR 1
25% polymicrobial
33.3% MSSA 33.3% MRSA
No data reported on dosage
Up to 84 d
91.7% DAP Clinical success:
10/12 (83%)
Microbiological cure:
In 9/12 (75%) cases, patients were switched to BPR following failure of the previous antimicrobial regimen.
In 3/3 patients in which BPR was administered because of
persistently positive blood culture, bacteraemia clearance was rapidly achieved.
No data reported
Zhanel, G.G. et al., 2021 [38]Case series
Mono and combination therapy
N/A38 infections
42.1% IE
23.7% BJIs
15.8% HABP
5.3% SSTI
2.6% CNS
2.6% DRI
2.6% BSI
9 mono and 29 combination
MRSA500 mg/8 h
No data on duration
Combination therapy 76.3%:
- DAP 21/29
- VAN 7/29
- FLUORO 1/29
Overall, clinical success 84.8%, microbiological cure 97.0%
In IE subgroup:
- Microbiological cure: 14/16, 2/16 unknown
- Clinical success: 11/16, 4/16 unknown; 1/16 death
2.6% AE (gastrointestinal symptoms)
Giuliano, S. et al., 2023 [40]Case seriesN/A21 BSI
13 left-sided IE
8 PVE, 5 NVE, 1 PVE + NVE
E. faecalis
AMP S
15/21 500 mg/8 h
3/21 500 mg/12 h
3/21 350 mg/8 h
Among patients
with IE, the mean duration of the ABPR regimen was 27.8 ± 14.5 days. In patients with
E. faecalis bacteraemia, the mean duration of ABPR treatment was 20.4 ± 11.1 days.
All ampicillinOverall clinical success 81%,
microbiological cure 86%
In IE subgroup:
- Clinical success: 9 (6 PVE, 3 NVE)
- Microbiological cure: 10 (5 PVE, 5 NVE)
1 relapse in NVE (pt did not adhere to the partial oral treatment)
9% experienced ABPR-related side effects (seizure and skin rash)
Oltolini, C. et al.,
2016 [35]
Case reportN/A1 PVEMRSA250 mg/2 h then
500 mg/8 h according to GRF
11 weeks
DAP Clearance of bacteraemia
Complete disappearance of
the vegetation at
echocardiography
IE recurrence
(it was not attributable to antibiotic failure but to EVS with the implantation of a new prosthesis during an uncontrolled infection status and also the recurrence of PVE and the need for chronic antibiotic therapy)
No data reported
Abbreviations: ABPR: ampicillin plus ceftobiprole combination; BJI: bone and joint infection; BPR: ceftobiprole; BSI: bloodstream infection; CIED-IE: cardiovascular implantable electronic device endocarditis; CDI: clostridioides difficile infection; CLABSI: central line-associated bloodstream infection; CNS: central nervous system; DAP: daptomycin; DRI: device-related infection; IE: infective endocarditis; EVS: early valve surgery; FLUORO: fluoroquinolone; HABP: hospital-associated bacterial pneumonia; LNZ: linezolid; MRSA: methicillin-resistant S. aureus; MR CoNS: methicillin-resistant coagulase-negative Staphylococci; MSSA: methicillin-sensible S. aureus; NVE: native valve infection; PVE: prosthetic valve infection; SAB: S. aureus bacteraemia; SSTI: skin and soft tissue infection; VAN: vancomycin; N/A: not applicable: AE: adverse events. Definitions: Clinical success was defined as clinical improvement with resolution of all signs and symptoms of infection during BPR treatment or at the end of therapy. Microbiological cure was defined as negative follow-up blood cultures after the index-positive blood culture at some point during treatment and a negative valve culture in patients who underwent surgery. Notes: * all the ERADICATE study results were published at the end of September 2023 and were not included in the review. As for the results published in 2022, the study confirmed the non-inferiority of BPR compared to DAP.
The recent ERADICATE study, a randomised double-blind trial, compared the efficacy of BPR versus daptomycin ± aztreonam in the treatment of S. aureus bacteraemia (SAB) (n = 390), including ABSSSI, osteomyelitis, and native-valve IE (8.5%). Daptomycin (DAP) was administered at a dosage ranging from 6 mg/Kg to 10 mg/Kg q24h, while BPR was given at a dosage of 500 mg q6h from Day 1 to Day 8 and 500 mg q8h from Day 9 onwards, with dose adjustments according to renal function. The study showed the non-inferiority of BPR compared to DAP in terms of mortality rates, microbiological eradication, and the occurrence of new complications associated with bacteraemia (overall clinical success: 69.8% in BPR-regimen vs 68.7% in DAP-regimen) [39,41].
In a recent Italian multicentre observational study on the real-life use of BPR, seven cases of IE were described: two from MRSA and five from methicillin-resistant coagulase-negative staphylococci (MR-CoNS). BPR was always used in combination with DAP (n = 6) and linezolid (n = 1). In this study, only two out of seven patients with IE achieved clinical success, with a mortality rate of 28.6%, while overall microbiological and clinical success was obtained in 29% of patients [7].
Tascini et al. described the use of BPR in 12 patients with EI caused by Staphylococcus spp., including MRSA (n = 4). Three patients had polymicrobial IE. The majority of patients (83%) were switched to BPR due to the failure of previous antimicrobial regimens, mostly represented by DAP. BPR was administered in combination with DAP in 11/12 patients, while in one patient, BPR was administered as monotherapy. The cure rate was 83% (10/12 patients). Notably, the addition of BPR resulted in a rapid microbial clearance in all the three patients with persistently positive blood cultures under previous treatments [37].
Taking into account BPR’s pharmacokinetic–pharmacodynamic (PK–PD) profile, its microbial activity against E. faecalis by means of a high level of enterococcal PBP saturation, its synergism in combination with amoxicillin, and its enhanced activity against biofilms, Giuliano et al. investigated the use of BPR in combination with ampicillin (AMP) in a case series of 21 patients hospitalised for infections due to E. faecalis, including IE (n = 13). Clinical success was reached in 81% patients, with a microbiological cure obtained in 86% of patients. In the EI subgroup, clinical and microbiological success was reached in 69% and 77% of patients, respectively [40]. Experiences from case reports and case series in the literature also suggest the effectiveness of BPR as a monotherapy or as a combination regimen with DAP in achieving the microbiological eradication of MRSA EI [35,36,38].
Overall, we recorded 70 IE episodes caused mostly by Staphylococcus aureus (both methicillin-resistant and susceptible (MSSA)) and 13 cases of left-side IE due to AMP-S E. faecalis. The cases occurred in both native and prosthetic valves. Notably, the RCT ERADICATE included mostly right-sided IE. The outcomes were frequently favourable, with a good percentage of cases ending in microbiological and clinical cure.

3.2. Ceftaroline

3.2.1. Mechanism of Action and Indication

Ceftaroline (CPT) is an intravenous fifth-generation cephalosporin which inhibits the bacterial cell wall by irreversibly binding PBPs. As in the case of ceftobiprole, its molecular structure confers an increased binding affinity to PBP-2a, improving its activity against MRSA [42]. CPT also exhibits in vitro activity against CoNS, streptococci (including S. pneumoniae and S. pyogenes), Moraxella catarralis, Haemophilus influentiae, and Gram-negative bacteria including Klebsiella spp. and Escherichia coli. Notably, the in vitro activity includes vancomycin-intermediate S. aureus (VISA) and cephalosporine-resistant S. pneumoniae [43]. In contrast, CPT seems to have no activity against E. faecium and a variable activity against E. faecalis [44].
The data available in the literature investigating CPT in vitro synergisms and experimental models of IE are discussed in Supplementary Material, Sections S2.1 and S2.2 [45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62].
CPT is currently approved by the FDA and EMA for the treatment of ABSSSI and CAP caused by susceptible microorganisms including MRSA. It is also approved in case of ABSSSI and CAP with intercurrent bacteriemia due to susceptible microorganisms with caution in MRSA bacteriemia in course of CAP [63].

3.2.2. Clinical Evidence in Infective Endocarditis

Several studies investigating the treatment of bacteriemia due to MRSA consider CPT an option even in IE populations. However, the results in IE were often not reported or were discussed separately, although two multicentre observational retrospective studies and one case series reported results only for IE. Relevant clinical studies and case reports on the use of CPT in IE are summarised in Table 2.
Only one RCT enrolling patients with MRSA bloodstream infection (BSI) (n = 40) included IE (n = 7) and randomised patients in combination therapy with CPT + DAP (600 mg/8 h or adjusted for renal function) or DAP/VAN monotherapy. The IE patients were randomised as follows: three were in the combination group vs. four in the monotherapy group (3 VAN and 1 DAP). Overall, the study showed that combination therapy was associated with a significantly lower in-hospital mortality rate (0% vs. 26%; p = 0.029), which was also reflected in the IE subgroup; the excess mortality observed in the monotherapy arm during the interim analysis led the investigators to stop the study early [8]. The study was a pilot clinical trial which did not reach an appropriate sample size; consequently, the results did not provide any strong evidence and no definitive conclusions could be drawn.
Brandariz-Nunez and colleagues described 70 IE cases caused by different pathogens (MSSA, MRSA, MS and MR CoNS, AMP-S E. faecalis, Streptococcus spp.), all of which were CPT in vitro susceptible, with a 30% overall in-hospital mortality rate and a 38.6% treatment failure ate at 42 days. CPT was used in combination, mostly with DAP, at a dosage of 600 mg every 8 h or 12 h (or adjusted based on renal function) [64].
The CAPTURE study, a multicentre observational retrospective cohort, reported 55 IE cases due to different Gram-positive bacteria, mostly MRSA (80%), with an overall clinical success of more than 70% and a high success rate when CPT was administered as a first, second, or later line therapy. CPT was used in 32 patients as a combination therapy, mostly with DAP or vancomycin (VAN) [65].
Three multicentre retrospective studies including patients with various Staphylococcal infections and treated with CPT both in combination or monotherapy reported data on IE patients’ outcomes: clinical success was observed in 69.7% and 78% of cases in two studies [56,66], with mortality rates of 22.9%, 7%, and 11%, respectively [56,66,67].
Zasowski and colleagues observed in both MRSA BSI and IE populations that CPT monotherapy was not inferior to DAP in terms of composite failure, expressed in terms of 30 d mortality, persistent bacteraemia > 7 d, and 60 d BSI recurrence [68].
In a large multicentre retrospective study, there was no significant difference in terms of the mortality rate, hospital readmission, or BSI recurrence between combination therapy with DAP plus CPT (with no data reported on dosage) and the standard of care monotherapy (mostly VAN) in the treatment of 171 patients with MRSA BSI, of which 70 had IE [69].
Few single-centre observational studies reported positive clinical and/or microbiological outcomes in MRSA BSI populations, with or without specific data on the IE subgroups [70,71,72,73,74,75,76]. Additionally, several case series and complicated case reports showed microbiological cure and clinical success in IE patients treated with CPT as a monotherapy or in combination [56,72,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92].
While the majority of studies described the use of CPT in combination, mostly with DAP but also with VAN, some studies investigated CPT use in monotherapy versus combination therapy. In 2017, Zasowski [93] and colleagues showed no statistical differences in mortality, microbiological cure, and clinical success between CPT monotherapy [most common dose 600 mg (61.8%) and frequency every 8 h (58.4%)] and combination therapy in 126 patients with MRSA BSI included in the efficacy population group, with 31 cases of IE. Likewise, a recent study observed no statistically significant differences in the composite outcomes of inpatient infection-related mortality, 60 day readmission, and 60 day BSI recurrence in MRSA BSI patients treated only with combination therapy (DAP + CPT) versus de-escalation to monotherapy (DAP/CPT/VAN) after a start with DAP + CPT [94].
Overall, the safety profile of CPT seemed to be similar to that of other beta-lactams also used in prolonged treatment for IE. In a recent systematic review, authors found 9% (83 out of 933) of adverse events were related to the use of CPT, mostly gastrointestinal events, rashes, and neutropenia [95]. In our review, we also found several cases of C. difficile infections, eosinophilia, and thrombocytopenia and a few cases requiring CPT withdrawal (Table 2).
Overall, we recorded 677 IE cases caused mostly by MRSA and involving both native and prosthetic valves (right and left sides) as well as CIEDs. The outcomes, when reported, were frequently positive, with microbiological and clinical cure.
Table 2. Clinical studies investigating the treatment of infective endocarditis with ceftaroline.
Table 2. Clinical studies investigating the treatment of infective endocarditis with ceftaroline.
AuthorsStudy DesignEndpointN° Patients/
IE Type
PathogensDosage and DurationCombination OutcomesSafety
Geriak, M. et al., 2019 [8]Randomised clinical trial
DAP + CPT vs. VAN/DAP
Primary endpoints:
duration of bacteraemia and in-hospital mortality
Secondary endpoints:
60 d and 90 d mortality, hospital stay
40 BSI,
17 DAP + CPT vs. 23 VAN/DAP (VAN 21, DAP 2)
7 IE,
3 DAP + CPT vs. 4 VAN/DAP
(1 bilateral, 1 right-sided, 1 aortic PVE, 1 mitral NVE, 1 aortic NVE, 2 CIED)
MRSACPT 600 mg 8 h (or adjusted for GFR)
Mean 11 d
DAP 8 mg/kg/24 h Overall, 30 d, 90 d, and in-hospital mortality:
DAP + CPT 0 vs. VAN/DAP 6, 0 vs. 7, 0 vs. 6
Treatment failure *:
1 vs. 3
IE subgroup:
in-hospital mortality, 0 vs. 2
No AE reported
Casapao, A.M. et al., 2014 [66] Multicentre observational retrospective study
CPT in various infections
Clinical and microbiological success/failure, hospital length of stay, AEs, 30 d readmission, in-hospital mortality, and 30 d mortality.527 infections
148 (28.1%) BSI
35 IE
138 SAB with 92% MRSA
in IE group
6 hVISA
Overall, 85.6% 600 mg/12 h, 14.4% 600 mg/8 h
Median 9 (4–15) in BSI group
29.2% combination therapy, 42% of which was with metronidazoleIn IE subgroup:
Clinical failure 30.3%
Mortality 22.9%
In the BSI group: 12.8% AE
Arshad, S. et al., 2017 [76]Retrospective case-control study
CPT vs. VAN vs. DAP
Composite failure:
30 d mortality from infection onset, 42 d BSI recurrence, or 30 d readmission
after the end of treatment
132 BSI, monotherapy 30 CPT vs. 46 VAN vs. 56 DAP
39 IE
7 vs. 13 vs. 19
MRSANo data reportedNo data reportedOverall, 30 d mortality:
CPT group 13% vs. DAP group 24% and VAN group 11% (p = 0.188)
Overall and in the IE subgroup, no statistically significant difference in 30 d mortality, 42 d recurrence, and 30 d readmission
No data reported
Britt, R.S. et al., 2017 [67]Multicentre observational retrospective study
CPT in various infections
AEs within 30 d of therapy initiation
All-cause in-hospital mortality
764 infections
46 IE
No data reportedNo data reportedNo data reportedOverall, in hospital mortality 5%, 30 d readmission 33%
IE subgroup
mortality 11%,
30 d readmission 28%
AE < 1% (eosinophilia, leukopenia, fibromyalgia, myalgia and myositis, and polymyalgia)
Zasowski, E.J. et al., 2017 [93]Multicentre observational-retrospective study
CPT mono vs. combination therapy in BSI
Safety and efficacy outcomes211 BSI,
126 included in the efficacy population
31 IE
20 CPT mono vs. 11 combination therapy
MRSA
1% VAN resistant strain
In efficacy population, most common dose 600 mg (60.3%) and frequency every 8 h (52.4%)
In efficacy population, median 13 d (IQR 5–21)
DAP combination in 75.7%In efficacy population no statistical differences between monotherapy and combination.
Clinical success § 86/126 (68.3%) monotherapy 69.7% vs. combination 64.9%,
BSI clearance 115/126 §§ (91.3), 88.8% vs. 97.3%,
Mortality 28/126 (22.2%), 19.1% vs. 29.7%
Overall, 16 AE (6 CDI, 7 rash, 3 neutropenia)
Cortes-Penfield, N. et al., 2018 [71]Observational retrospective study
DAP + CPT vs. DAP in BSI
Duration of bacteraemia, mortality,
BSI recurrence
17 BSI,
5 IE
12 DAP + CPT and 5 DAP
MRSANo data on dosage
Mean 32.5 d
DAP median dose 7.6 mg/kg/24 h (5.7–13.8)Overall, shorter duration of bacteraemia in DAP + CPT group
IE subgroup
mortality 3/5
No data reported
Destache, C.J. et al., 2019 [65]Multicentre observational retrospective study

CPT mono or combination therapy in IE
Clinical outcomes55 IE,
26 right-sided, 25 left sided,
4 bilateral
MRSA 44/55
MSSA 4
CoNS 4
E. faecalis 1 Streptococcus 1
Mainly 600 mg/12 h
Mean (SD) 13.4 d (9.7)
32,
most common drugs (>5% of pt) DAP (n. 19), VAN (n. 9), RIF (n. 7). Other drugs: CFZ, LVX, LNZ, GEN, AMP.
Overall, clinical successes 39 (70.9%): monotherapy 19/23 (82.6%), combination 20/32 (62.5%)
High success rate with CPT as first or second line therapy
2 AE (AKI and rash) with CPT withdrawal
McCreary, E.K. et al., 2019 [69]Multicentre observational retrospective study
DAP + CPT vs. SoC
(case-control)
All-cause mortality,
duration of bacteraemia, and BSI recurrence
171 BSI,
58 DAP + CPT vs. 113 SoC (VAN or DAP),
70 EI,
23 vs. 47
MRSANo data reportedNo data reportedNo statistically significant difference in all-cause 30 d mortality and 90 d BSI recurrenceNo data reported
Ahmad, O. et al., 2020 [70]Retrospective case-control study
VAN or DAP vs. VAN/DAP +CPT
Treatment outcomes:
in-hospital mortality, BSI recurrence, 30 d readmission,
AKI, leukopenia
30 BSI,
15 VAN/DAP vs.
15 VAN/DAP + CPT
21 IE, all NVE
(14 vs. 7)
MRSA600 mg/8–12 h
Median 6 weeks
VAN 15–20 mg/kg/8–12 h
DAP 8–10 mg/kg/24 h
No difference in AKI, leukopenia, BSI recurrence, 30 d readmission, or mortalityNo AE reported
Morrisette, T. et al., 2020 [75]Observational retrospective study
DAP vs. DAP + CPT
Composite success:
30 d mortality, 60 d recurrence, worsening of respiratory status, change in therapy due to failure
29 BSI with septic pulmonary emboli,
14 DAP vs. 15 DAP + CPT
24 IE, all NVE (11 vs. 13)
MRSA600 mg/8 h
Median 11 d (9–12)
DAP median 9.9 mg/kg (8.8–9.8)
duration median 36 d (22–42)
No difference in the primary outcome of compositive success1 AE (thrombocytopenia) with CPT withdrawal
Johnson, T.M. et al., 2021 [73]Observational retrospective study
DAP + CPT vs. SoC
Clinical failure:
MRSA-related mortality and 60 d recurrent infection
60 BSI,
30 DAP + CPT vs. 30 SoC,
22 IE, 15 vs. 7
(14 left-sided, 6 right-sided, 2 bilateral)
MRSA1800 mg/24 h (or adjusted for GFR)
DAP + CPT median 7 d (3–11)
DAP 10 mg/kg/24 hOverall, clinical failure DAP + CPT 20% vs. SoC 43%,
60 d BSI recurrence 0% vs. 30%,
90 d mortality 27% vs. 23%,
DAP + CPT inversely associated with clinical failure 90 d (p = 0.03)
No statistically significant AE reported
Nichols, C.N. et al., 2021 [94]Observational retrospective study
DAP + CPT vs. de-escalation with DAP/CPT/or VAN
Composite endpoint:
inpatient
infection-related mortality, 60 d readmission, and 60 d BSI recurrence
140 BSI,
66 DAP + CPT vs. 74 de-escalation in monotherapy DAP/CPT/VAN
63 IE, 37 vs. 26
MRSANo data on dosage
Median 56 d in combination group
DAP No differences between combo and monotherapy for inpatient infection-related mortality, 60 d readmission, or 60 d BSI recurrenceIn the combination group, 2 AE (bone marrow suppression, oedema)
Zasowski, E.J. et al., 2022 [68]Multicentre observational retrospective study
CPT vs. DAP monotherapy
Composite treatment failure:
30 d mortality, BSI duration ≥ 7 d on study drug, and 60 d MRSA BSI recurrence.
270 BSI, 83 CPT and 187 DAP
82 IE
27 vs. 55
MRSAMost common dose 600 mg (68.7%) and frequency every 12 h (56.6%)
Median 10 d (IQR 5–18)
No
Monotherapy DAP median 8.5 mg/kg 24 h
In all populations and the IE subgroup,
CPT not inferior to DAP
No differences in any endpoints
Overall, 17 AE (9 rash, 4 CDI, 5 others)
No data on CPT discontinuation was reported
Brandariz-Nunez, D. et al., 2022 [64]Observational retrospective study
CPT combination in IE
Treatment failure:
presence of fever or positive BC at 7 d, positive BC recurrence, early
antibiotic withdrawal due to lack of clinical response, AE or death
70 IE,
30 NVE, 36 PVE, 10 ICED-IE
MRSA 6/26;
MR CoNS 15/26;
E. faecalis AMP-S 5; Streptococcus 5
600 mg/8–12 h (or adjusted on GFR)
Mean 21.26 d (DS 16.17)
70/70 combination
DAP (n.52), GEN (n.18), RIF (n.6)
Overall, 42 d in-hospital mortality 30%;
42 d treatment failure 38.6%
6 AE
with 4 CPT discontinuation
Kufel, W.D. et al., 2023 [74]Observational retrospective study
CPT + VAN in BSI
Effectiveness
and safety
Bacteraemia clearance post-CPT
initiation
30 BSI,
20 IE,
7 tricuspid, 7 mitral, 4 aortic and 2 multiple valves
MRSA 600 mg/8 h
Median 16 d (IQR 13.2)
All combination,
VAN median 1250 mg/24 h
Overall, microbiological cure 96.7%;
90 d readmission for MRSA BSI 6.7%,
all-cause 90 d mortality 26.7%,
MRSAB-related mortality+ 13.3%
2 AE (rash) with CPT discontinuation
Lin, J.C. et al., 2013 [92]Case seriesN/A10 infections
5 IE,
4 probable and 1 possible.
1 right-sided, 1 CIED, 1 NV+ CIED-IE, 2 no vegetation
MRSA600 mg/8 h (or adjusted por GFR)
Between 3 d to 7 weeks
No data reportedIE subgroup
Clinical cure 3/5
Microbiological cure 4/5
2 AE,
1 CDI,
1 fever + rash + eosinophilia with CPT discontinuation
Ho, T.T. et al., 2012 [91]Case series
CPT monotherapy
N/A6 BSI,
3 IE
Cases 1 and 2: middle-aged men with mitral NVE
Case 3: middle-age woman with mitral NVE
MRSA600 mg/8 h
Case 1: 42 d
Case 2–3: 3 weeks
No IE subgroup
Case 1–3: microbiological cure and clinical cure
No data reported
Polenakovik, H.M. and Pleiman, C.M.
2013 [78]
Case seriesN/A31 BSI,
10 IE,
3 left-sided, 6 right-sided, and 1 CIED-IE
MRSACPT 1200–1800 mg/24 h (1 case GFR dose-adjusted)
Overall median 30.4 d (IQR 7–60)
4 IE combinations with DAP, RIF, GEN, LNZOverall, microbiological cure 64.5% (IE 9 pt);
Clinical success 74.2%
(IE 9 pt);
Treatment failure ° 25.8%
(IE 1 pt)
Recurrence 9.7% (IE 1 pt);
Death 6.5%
Overall, 2 AE (eosinophilia) without CPT discontinuation (1 IE)
3 AE (eosinophilic pneumoniae, rash, diarrhoea) with CPT discontinuation
Fabre, V. et al., 2014 [72]Case seriesN/A29 BSI
18 IE
4 right-sided, 11 left-sided, 1 CIED, 2 LVAD
MRSA600 mg/8 h
(or adjusted on GFR)
No data on duration
24 combination therapies:
22 with TMP-SMZ 10–15 mg/kg/24 h
2 with DAP
Overall, microbiological success: 26/29 (90%);
Treatment success # with 6 months FU: 9 (31%);
Treatment failure ##: 4 (13%) (1 death, 3 recurrence)
1 AE (rash) with CPT discontinuation
Tattevin, P. et al., 2014 [79]Multicentre
case series
CPT in IE
N/A8 IE
3 aortic PVE, 1 aortic PV plus pulmonary valve, 1 CIED,
1 mitral and aortic NVE, 1 aortic NVE, 1 CIED plus aortic NVE
5 MRSA
3 MR CoNS
From 400 mg/12 h to 800 mg/8 h
Median 13 d (5–42)
3 combination DAP (n 2) RIF (n 1)Clinical success: 5/8
Clinical failure: 3/8
No AE reported
Gritsenko, D. et al., 2017 [90]Case series
CPT + VAN
N/A5 BSI,
2 IE,
Case 2: 42 y man with tricuspid NVE
Case 5: 50 y mitral NVE
MRSACase 2: 400 mg/12 h (adjusted for GFR)
6 weeks
Case 5: 600 mg/12 h (then adjusted for GFR)
7 d
Case 2 and 5: combo with VAN
Case 5: 7 d
IE subgroup
Case 2: microbiological cure and clinical success
Case 5: death
No data reported
Hornak, J.P. et al., 2019 [77]Case series
CPT + DAP in BSI
N/A10 BSI
6 IE,
1 mitral NVE, 3 aortic NVE, 1 CIED, 1 LVAD
MRSA4600 mg/12 h, 1600 mg/8 h, 1 400 mg/h 8.
Overall, median time 9 d (IQR 6–24)
All IE combination with DAPIE subgroup microbiological cure 6/6;
no recurrence;
30 d mortality and in-hospital mortality 1/6
3 AE (rash, eosinophilia, thrombocytopenia) without CPT discontinuation
1 eosinophilia in IE group
Rose, W.E. el al., 2012 [89]Case report
Failure with DAP
N/A1
right atrial vegetation
MRSA and DNS200 mg/12 h (haemodialysis dose-adjusted)
54 d
DAP
10 mg/kg/24 h
Microbiological cure and clinical success after failure with 11 d of monotherapy with DAP 6 mg/kg 48 hNo data reported
Jongsma, K. et al., 2013 [88]Case reportN/A1
tricuspid and aortic NVE
MRSA and DNS600 mg/12 h
44 d
NoNo resolution after 23 d of DAP and VAN,
debridement on 19 d,
microbiological cure at 7 d after CPT start,
clinical success
No data reported
Sakoulas, G. et al., 2013 [87]Case report
Failure with AMP-based regimens
N/A1
aortic NVE
HLGR E. faecalis600 mg/8 h
6 weeks
DAP
8 mg/kg/24 h
Microbiological cure and clinical success achieved after failure with CRO + AMP (6 weeks) and then DAP + AMP (7 d).
2 weeks after CPT + DAP start, aortic valve replacement was performed
No data reported
Baxi, S.M. et al., 2015 [86]Case-report
CPT + DAP
N/A1
mitral NVE
MRSA VISA and DNS400 mg/12 h
6 weeks of CPT + DAP
DAP 10 mg/kg after dialysisNegative BC from day 11 of DAP + CPT, remain negative at 28 d after discontinuationNo AE reported
Cunha, B.A. et al., 2015 [85]Case report
Persistent bacteraemia with DAP
N/A1
aortic PVE
MRSA 600 mg/12 h
6 weeks
DAP 10–12 mg/kg/24 hPersistent bacteraemia for 14 d under DAP 10 mg/kg 24 h
BC negative after 4 d of DAP+ CPT, no recurrence
No data reported
Sundaragiri, P.R. et al., 2015 [84]Case reportN/A1
tricuspid NVE
MRSANo data reportedNo data reported9 d valve replacement
Death
No data reported
Duss, F.R. et al., 2019 [83]Case report
Persistent bacteraemia with VAN
N/A1
left NVE
MRSA
(MIC: VAN 1.5 mg/L, DAP 2 mg/L)
600 mg/12 h
6 weeks
DAP 10 mg/kg/24 hBC positive under VAN 5 d; switch DAP + FOS; day 10 surgery and culture valve negative. After surgery CPT + DAP for 6 weeks.
Negative BC and persistent negative at 6 months FU
No data reported
Jilani, T.N. and Masood, S.O.
2018 [82]
Case report
Failure with DAP and VAN
N/A1
pulmonic NVE
MRSA600 mg/8 h
4 weeks after 2 weeks of VAN and DAP
No Microbiological cure after 2 d of CPT and clinical successNo data reported
Lin, S.Y. et al., 2021 [81]Case report
Failure with DAP and VAN
N/A1
mitral NVE
hVISA 600 mg/12 h
5 weeks
DAP
9 mg/kg/24 h
Microbiological cure and clinical success achieved after failure with monotherapy VAN (14 d) and then DAP (7 d)No data reported
Warren, E.F. et al., 2022 [80]Case report
CPT+ nafcillin
N/ACase 1,
tricuspid NVE
Case 2,
CIED-IE
MSSACase 1
600 mg/8 h
Case 2
600 mg/12 h (GFR dose-adjusted)
Case 1: 11 d
Case 2: 7 d
Case 1 and 2: nafcillin 12 g 24 hMicrobiological cure and clinical successNo data reported
Abbreviations: CPT: ceftaroline; AE: adverse event; MRSA: methicillin-resistant Staphylococcus aureus; IE: infectious endocarditis; hVISA: heterogeneus vancomycin-intermediate S. aureus; BSI: bloodstream infection; SAB: S. aureus bacteraemia; d: day; VAN: vancomycin; DAP: daptomycin; CDI: C. difficile infection; AKI: acute kidney injury; CoNS: coagulase-negative staphylococci; MSSA: methicillin-susceptible S. aureus; LNZ: linezolid; LVX: levofloxacin; CFZ: cefazolin; GEN: gentamicin; AMP: ampicillin; RIF: rifampicin; PVE: prosthetic valve endocarditis; NVE: native valve endocarditis; CIED-IE: cardiovascular implantable electronic device endocarditis; SoC: standard of care; GFR: glomerular filtration rate; BC: blood culture; MR CoNS: methicillin-resistant coagulase-negative staphylococci; MRSAB: methicillin-resistant S. aureus bacteraemia; N/A: not applicable; FU: follow up; VISA: vancomycin-intermediate S. aureus; MRSE: methicillin-resistant Staphylococcus epidermidis. Definitions: Clinical success/cure was defined as clinical improvement with resolution of all signs and symptoms of infection during CPT treatment or at the end of therapy, unless otherwise specified. Casapao AM et al. and Destache CJ et al. defined clinical success as above or as a clinical improvement with no further need for escalation while on CPT treatment or during hospitalization [65,66]. Clinical failure was defined as inadequate response or resistance to CPT therapy, worsening of the clinical conditions during the treatment, or new recurrent signs and symptoms at the end of CPT therapy [66]. Microbiological success/cure was defined as a documented negative blood culture result or BC clearance. Duration of bacteraemia was calculated as the number of days between the first positive blood culture and the first negative blood culture without subsequent positive cultures. Bacteraemia recurrence was defined as at least one positive blood culture for MRSA after an initial microbiological cure. Notes: § Clinical success was defined as BSI clearance and cessation of BSI signs and symptoms (i.e., fever and leukocytosis) by the end of therapy or discharge and living patients at hospital discharge; §§ Clearance of bloodstream infection was defined as a series of two consecutive negative blood cultures. * Patients with persistent bacteraemia for ≥5 days or deemed to be failing clinically on the regimen selected by the randomization process. +MRSAB-related mortality was defined as death prior to blood culture clearance or within 2 weeks following blood culture clearance using the date of the first positive blood culture as Day 1. ° Treatment failure was defined as any of the following: (i) persistent signs and symptoms of infection at the end of CPT therapy; (ii) persistent MRSAB defined as >7 days; (iii) recurrent MRSAB after the end of CPT therapy; (iv) death that could be attributed to ongoing infection (defined as MRSA-positive blood cultures at the time of death, death occurring before resolution of the signs and symptoms of MRSAB, or autopsy finding indicating MRSA infection as a cause of death); and (v) adverse drug reaction requiring cessation of CPT treatment. # Treatment success was defined as the absence of microbiologic or clinical recurrence at least 6 weeks after the end of therapy; ## treatment failure was defined as recurrence of MRSA infection after completion of CPT therapy or death related to MRSA infection.

3.3. Dalbavancin

3.3.1. Mechanism of Action and Indication

Dalbavancin (DAL) is a semisynthetic lipoglycopeptide derived from teicoplanin which is characterised by a unique PK profile with a prolonged half-life, lasting just over two weeks [96]. Similar to glycopeptides, DAL binds the C-terminal D-alanyl-D-alanine motif of peptidoglycan, inhibiting wall biosynthesis [97]. DAL exhibits excellent in vitro activity against the main Gram-positive pathogens, including vancomycin-susceptible enterococci, VanB E. faecalis, and VanB E. faecium, although it is inactive against VanA-phenotype enterococci [98]. This second-generation lipoglycopeptide exhibits potential penetration of and activity against the established biofilm produced by Gram-positive bacteria [99].
Studies investigating DAL in vitro synergisms and experimental models of IE are shown in Supplementary Material, Sections S3.1 and S3.2 [100,101,102,103].
Currently, DAL is approved for ABSSSI in adults by the FDA and the EMA. Recently, the approval was extended to pediatric ABSSSI [104,105]. In fact, the off-label application of this antibiotic in more deep-seated infections commonly caused by Gram-positive bacteria and requiring prolonged antimicrobial treatment is supported by an ever-growing body of evidence, and it can be used in conditions including osteomyelitis, prosthetic joint infections, endovascular device infections, BSI, and IE [96].

3.3.2. Clinical Evidence in Infective Endocarditis

The available evidence in the literature concerning the application of DAL in IE is still mainly represented by observational and retrospective studies, case series, and case reports. No prospective randomised trial is available yet. Moreover, many data are only available in aggregate form because IE cases were a subgroup of larger studied populations. DAL prescription has been reserved primarily for the consolidation or completion phase of treatment in patients with already cleared bacteraemia. Published relevant clinical studies and cases on the use of DAL in IE are summarised in Table 3.
In a two-year retrospective cohort study, 27 patients with Gram-positive IE received primary or sequential DAL. The majority (88.9%) were previously treated with another with another antimicrobial and gaining bacteremia clearance antimicrobial agent for bacteraemia clearance. DAL was administered as a twice-weekly regimen [1500 mg loading dose (LD), then 1000 mg] in 63.0% of cases, with a median duration of 6 weeks. Failure was described in one patient with incomplete surgical control of cardiac device-related MRSA IE who received 30 weekly DAL infusions. Importantly, all cases received at least one DAL dose in hospital, but 23 continued DAL as OPAT [14].
The Italian multicentric study DALBITA retrospectively enrolled 206 patients treated with DAL, of which six had IE. In the whole cohort, MRSA (32%), CoNS (29%), and methicillin-susceptible S. aureus (MSSA) (18%) were the most frequent isolates, and 77.8% of patients received prior therapy for a median of 15 days. Clinical success was recorded in 83.3% of the IE subgroup [106].
In a system-wide retrospective analysis of 56 people receiving long-acting lipoglycopeptides, five had IE. Forty received DAL, fourteen received oritavancin, and two received both, but the outcomes of the two agents were not distinguishable. The success rate was 100% among the three IE cases included in the success/failure analysis [107].
A national cohort included 19 IE cases (nine native valve and ten prosthetic) among 75 patients. In the whole cohort, the main isolates were S. aureus (51.4%) and CoNS (44.4%); prior therapy was received in 98.7% of cases. DAL dosing for IE was a 1500 mg single or double dose, with a cure rate of 72.2%. Here, DAL was largely used as a rescue treatment, justifying the high failure rate [108].
In a retrospective multicentre study on real-life DAL use, 25 out of 101 subjects had IE. All received other antimicrobials before DAL and 64% received concomitant antibiotics while on DAL. The success rate was 92% among IE patients [109].
DALBACEN is a multicentre retrospective Spanish cohort that included 124 elderly, predominantly male patients with major comorbidities who received DAL for IE (46.8% native valve, 43.6% prosthetic valve, and 9.6% pacemaker lead IE). CoNS (38.7%), MSSA (22.6%), E. faecalis (19.4%), and Streptococcus spp. (9.7%) were the most isolated pathogens. Almost all patients (98.4%) received prior antibiotic treatment for a median of 9.5 days, followed in 60.5% of cases by a second regimen for a median of 24.5 days. DAL usually represented a sequential or consolidation therapy in hospitalised patients, with a single 1500 mg dose being the most frequent regimen. Surgery was undergone in 45.9% of cases, usually before DAL. The main reason for prescription was to accelerate the rate of discharge (95.2%), resulting in a median fourteen-day reduction in hospital stay. Overall clinical success in patients who completed the one-year follow-up was 95.9% [9].
An observational study enrolled 22 patients treated with DAL after previous antimicrobials, of whom three had IE. Overall, S. aureus and CoNS were the most isolated pathogens, and the success rate was 95% [110].
A single-centre retrospective experience described 10 IE cases (three native valve, five prosthetic, and two CIED IE) mainly caused by staphylococci and enterococci. A median of 2.5 DAL doses were administered after at least 2 weeks of antimicrobials. Microbiological cure was obtained in 70% of cases, but long-term mortality was high (60%) and two patients relapsed [111].
Another retrospective analysis included 102 individuals, 14 (13.7%) of them with IE. All received antibiotics before DAL for a median of 18.5 days. S. aureus was isolated in 70.6% of cases. IE patients had a DAL LD of 1500 mg followed by a range of one to six 1500 mg doses. Overall, 93.7% reached clinical and microbiological success, and hospitalization was reduced by a median of 14 days (range 7–84) [112].
Several other studies investigated DAL in poorly compliant people with IE including homeless people, people who inject drugs (PWID), and people with alcohol disorders. In the majority of cases, patients were treated with previous intravenous antimicrobial regimens and were unsuitable for OPAT. Overall, the clinical success of DAL use was high, ranging from 66% to 100% [113,114,115,116,117,118,119,120]. However, the number of patients lost at follow-up was not negligible.
Finally, several cases and case series have described prolonged DAL treatment in patients with IE, with conflicting results [121,122,123,124,125,126,127]. Among the seven individuals with IE included in the study of real-life experience by Bouza et al., DAL was mainly used as a targeted therapy and only one failure was recorded [128].
Some authors reviewed the clinical efficacy of DAL for IE, with an overall success rate ranging from 68% to 95% [129,130], but acknowledged that most of the evidence came from retrospective studies and that there was a huge heterogeneity in the population included (PWID, cardiac device-related IE), the definition of outcomes, the quality of studies, the indications, and the dosing strategies. Notably, only three cases of DAL resistance were detected [96]. Our search confirmed this landscape.
Overall, we analyzed 313 cases of IE treated with DAL (the most-used regimen was a 1500 mg single or repeated dose), caused mostly by S. aureus (with a slight predominance of MSSA), followed by CoNS. Native valves of the right side were predominantly involved but cases involving the left side, prosthetic valves, and CIEDs were reported as well. Previous antibiotic treatment before DAL was almost universal. Clinical and microbiological outcomes were generally positive although there was an elevated rate of patients lost to follow-up and the data are difficult to interpret because of high heterogeneity.
Table 3. Clinical studies investigating the treatment of infective endocarditis with dalbavancin.
Table 3. Clinical studies investigating the treatment of infective endocarditis with dalbavancin.
AuthorsStudy DesignEndpointN° Patients/
IE Type
PathogensDosage and DurationCombination, DosageOutcomesSafety
Bouza, E. et al., 2018 [128]Multicentre retrospective studyEfficacy, tolerability, and cost reductions in people receiving DAL for various indications69, mainly prosthetic joint infections (29%) and ABSSSI (21.7%)
Previous therapy 97%
7 IE, type unspecified.
IE subgroup: CoNS (2), Enterococcus spp. (2), MRSA (1),
Streptococcus spp. (1), negative culture (1)
Most common regimen: 1000 mg Day 1, then weekly 500 mg Overall, 36.2%Overall clinical success 84.1% and significant cost reduction
IE subgroup
Clinical success: 85.7%. Failure in 1 IE patient attributed to inadequate source control
Overall, AE in 13%. Most common AE: rash and tachycardia.
Tobudic, S. et al., 2018 [14]Observational retrospective study
DAL in IE mainly administered as OPAT
Clinical cure
and safety
27 IE
Previous therapy 88.9%
16 NVE, 6 PVE and 5 CIED-IE
S. aureus (33.3%), CoNS (22%), and E. faecalis (14.8%) main pathogensAdministered as twice-weekly regimen in 63.0%
Median duration of 6 weeks (range, 1–30 weeks).
NoClinical and microbiological success: 92.6%.
Failure in 1 patient with MRSA CIED-IE and incomplete surgical control
2 AE: 1
nausea and vomiting after the second dose,
therapy continued.
1 creatinine increase, resolved with dose reduction.
Bryson-Cahn, C. et al., 2019 [115]Observational retrospective study on vulnerable patients
S. aureus serious infection
Clinical response:
any patient who had an FU visit within 1 year without evidence of ongoing/relapsed infection
32 infections (BSI 40.6%, osteoarticular 28%)
Previous therapy 100%.
9 IE
tricuspid NVE
2 IE MSSA
7 IE MRSA
22 received a single 1000 mg dose, 7 received 2 weekly dosesNoIE subgroup:
Clinical response 5/9
Lost to FU 4/9
No AE reported
Bork, J.T. et al., 2019 [116]Multicentre retrospective study on vulnerable patients
Invasive Gram-positive infections
Clinical cure 45 infections (osteomyelitis 45%, endovascular 25%)
Previous therapy 100%.
6 IE, type unspecified
MRSA (29%) and MSSA (21%) main pathogensMedian of 3 doses prescribed6 patients with concomitant oral fluoroquinolone.Overall, 30 day cure was achieved by 50% of patients with endovascular infection; >25% loss to FU.
IE subgroup unspecified.
AEs documented in 6.7% (2 acute kidney injuries and 1 rash)
Dinh, A. et al., 2019 [108]Multicentre retrospective study
French national cohort
Clinical cure75 infections (most frequent bone and joint 64%, endocarditis 25%). Previous therapy 98.7%
19 IE: 9 NVE and 10 PVE
S. aureus (51.4%) and CoNS (44.4%) main pathogensIn IE most frequent regimen was 1500 mg single or double doseOverall, 45.3%, mainly rifampicin, cotrimoxazole, quinolones and tetracyclinesOverall, clinical cure 79%.
IE subgroup
Clinical cure: 72.2%
Five AE in the cohort (6.7%) with no treatment discontinuation
Hidalgo-Tenorio, C. et al., 2023 [9]Multicentre retrospective study
DAL as consolidation treatment
Effectiveness of DAL as consolidation therapy124 IE (46.8% native valve, 43.6% prosthetic valve and 9.6% pacemaker lead IE).
Previous therapy 100%.
CoNS (38.7%), MSSA (22.6%) E. faecalis (19.4%) and Streptococcus species (9.7%) the most isolated pathogensSingle 1500 mg dose the most prescribed DAL regimen (33.3%)No data reportedClinical success in subjects that completed the 1 year follow-up: 95.9%
Mean reduction in hospital stay: 14 days.
AE in 3.2%
Morrisette, T. et al., 2019 [107]Multicentre retrospective study
DAL or ORI in various infections
Clinical success56 infections (ABSSSI 36%, osteomyelitis 27%), 40 DAL, 14 ORI and 2 both.
Previous therapy 91%
5 IE, type unspecified.
MSSA (25%), MRSA (19%) and E. faecalis (11%) main pathogensNo data reported30% of the whole cohort (drugs unspecified)IE subgroup
Clinical success: 100% among the 3 evaluable IE
Mild AE in 11%.
Wunsch, S. et al., 2019 [109]Multicentre retrospective study
DAL as sequential treatment
Clinical success101 infections (prosthetic joint 31%, osteomyelitis 30%, IE 25%)
Previous therapy 100%
25 IE: 15 NVE, 6 PVE, 4 CIED-IE
CoNS (33%), MSSA (16%), MRSA (9%) main pathogensIn IE, 9 single 1500 mg dose and 1000 mg dose followed by 500 mg 1 week apart.Overall, 64% of the cohort, mainly rifampicin (64%) and fluoroquinolones (15%)Overall, clinical success 89%.
IE subgroup
Clinical success: 92%
Three AE in the cohort (3%), requiring treatment discontinuation
Ajaka, L. et al., 2020 [117]Observational retrospective study in people with barriers to SoC Cure:
lack of clinical or microbiological persistent/recurrent infection within 90 days or negative BCs within 90 days after completion of DAL
28 infections (24 BSI and 4 IE)
Previous therapy 100%.
PWID 67%
4 IE, type unspecified
MRSA (39%) and MSSA (17%) main pathogensLD of 1500 mg followed by 1 maintenance doseNoOverall, 44% clinical cure, 33% failed treatment, and 22% lost to FU.No data reported
Bai, F. et al., 2020 [106]Multicentre retrospective study
DAL in various infections
Clinical cure 206 infections (124 ABSSSI, 82 other site infection)
Previous therapy 77.8%
6 IE, type unspecified.
MRSA (29%), CoNS (35%) and MSSA (17%) in the non-ABSSSI group.Overall, single 1500 mg dose in 60.2%In 37.2% of non-ABSSSI patients, mainly fluoroquinolones, rifampicin, and tetracyclineOverall clinical cure in non-ABSSSI 75%.
IE subgroup
Clinical cure: 83.3%
5.4% had an AE, mainly dermatologic. One serious AE (Stevens–Johnson).
Núñez-Núñez, M. et al., 2020 [110]Observational prospective study.
DAL as sequential treatment
Clinical success22 infections (osteoarticular 46%, BSI 23%).
Previous therapy 100%.
3 IE, type unspecified.
S. aureus (55%), CoNS (27%)63% of the whole cohort received 1000 mg followed by 500 mgNo data reportedOverall, clinical success 95%AE 1 (4.5%), infusion site reaction
Veve, M.P. et al., 2020 [119]Observational retrospective study
DAL vs. SOC
Incidence of infection-related readmission
within 90 d of hospital discharge or outpatient
DAL administration
215 infections (most common BSI, osteoarticular and IE)
70 DAL vs. 145 SoC
Previous therapy 100%.
IE 54: 9 DAL vs. 45 SOC
MRSA 82% Most frequent regimen 2: 1500 mg doses 1 week apartin 13% of DAL treated.Overall, DAL was associated with lower 90-day infection-related readmissions and shorter length of stay.AE 2.9% in the DAL group, 1 required discontinuation.
Durante-Mangoni, E. et al., 2021 [111]Observational single-centre retrospective study
DAL in IE
Clinical and microbiological cure 10 IE: 3 NVE, 5 PVE, 2 CIED-IE
At least 2 weeks previous therapy 100%
Mainly caused by staphylococci and enterococci.Median of 2.5 DAL doses per patientNo data reportedClinical and microbiological cure 70%1 AE (rash after the third dose) with treatment withdrawal
Arrieta-Loitegui, M. et al., 2022 [112]Observational retrospective study
DAL as sequential treatment
Clinical and
microbiological cure
102 infections (SSTI 30%, BSI 15.7%, IE 13.7%)
Previous therapy 100%.
14 IE, type unspecified
S. aureus in 70.6%IE patients, 1500 mg as LD followed by a range of 1–6: 1500 mg doses16.7%, mainly moxifloxacin and linezolidOverall, clinical and microbiological success: 93.7%.
Median reduction in hospitalization 14 days (range 7–84).
AE in 3.9%,
1 patient discontinued.
Taylor, K. et al., 2022 [114]Observational retrospective study
DAL as sequential treatment
Clinical success48 infections (osteomyelitis 54%, IE 23%, BSI 15%).
11 IE, type unspecified.
Previous therapy 100%
MRSA (42%) and MSSA (19%) main pathogensMost patients received 1500 mg doses
44% 1 dose, 52% 2 doses.
27%, mainly rifampin and quinolonesOverall clinical success 85%.
IE subgroup:
Clinical success at 90 days 82%.
No AE reported
Lueking, R. et al., 2023 [120]Observational retrospective study
Vulnerable people receiving DAL
Clinical failure
(not defined)
40 infections
(BSI 67.5%, ABSSSI 45%)
Previous therapy 100%.
4 IE, type unspecified
MRSA (57.5%) and MSSA (30%) main pathogensMost frequent regimen 1500 mg single doseIn 15% of the whole cohort.IE subgroup:
Clinical success in all patients
AE in 5%
Vazquez Deida, A.A. et al., 2020 [118] Case series
Vulnerable people receiving DAL
N/A27 infections (BSI 26%, IE 26%).
Previous therapy 100%
PWID 67%
9 right side IE
S. aureus 100% (48% MRSA).Single DAL dose 7–10 days before the planned end of therapyNoIE subgroup:
Clinical success in 6/9
Estimated cost avoidance of USD 9600 per patient in the whole cohort
AE in 7.4% (mild events)
Guleri, A. et al., 2021 [113]Case series
DAL in IE
N/A11 IE,
4 aortic NVE, 3 aortic PVE, 1 mitro-aortic NVE, 1 mitral NVE, 1 ICD-IE, 1 tricuspid NVE)
Previous therapy 100%.
MSSA and E. faecalis, main pathogens1 or 2: 1500 mg doses9, mostly oral amoxicillin.Clinical cure in all but one patient No AE reported
Hitzenbichler, F. et al., 2021 [127]Case series
DAL after clearance of bacteraemia
N/A4 IE
2 PVE
2 LVAD
MRSA
E. faecalis
E. faecium
Long-term suppressive DAL, various regimensNoClinical success with prolonged infection suppression in all IE casesNo AE reported
Steele, J.M. et al. 2018 [121]Case report
DNS strain
N/A1
Tricuspid NVE
DNS MRSA1000 mg LD, then 3 weekly 500 mg dosesNoClinical and microbiological failure,
bacteraemia relapse, isolation of a VISA and telavancin-non susceptible MRSA
No AE reported
Kussmann, M. et al., 2018 [125]Case reportN/A1
CIED-IE with incomplete PMK explantation
MRSAUnspecified dosing
30 weekly administrations
NoClinical and microbiological failure, bacteraemia relapse, isolation of a SCV strain teicoplanin-resistant and DAL non-susceptibleNo AE reported
Howard-Anderson, J. et al., 2019 [122] Case report
Suppressive therapy
N/A1
LVDA
MRSAWeekly 1500 mg for 10 weeks, then 1500 mg biweekly.
Total DAL exposure: 235 days
NoClinical success with prolonged infection suppressionNo AE reported
Spaziante, M. et al., 2019 [126]Case reportN/A1
Aortic PVE in a man with unacceptable perioperative risk
MRSE1500 mg whenever serum bactericidal activity titers detected ≤ 1:8NoClinical and radiological improvement with no recurrenceNo AE reported
Hakim, A. et al., 2020 [123]Case report
DAL as primary regimen
N/A1
Tricuspid NVE
MSSA1500 mg LD, followed by 5 weekly 500 mg dosesNoClinical successNo AE reported
Teigell-Muñoz, F.J. et al., 2023 [124]Case report
DAL as consolidation therapy
N/A1
Aortic NVE
E. faecalis1000 mg single dose, after 4 weeks of therapy and valve replacementNoClinical successNo AE reported
Abbreviations: ABSSSI: acute bacterial skin and skin structure infection; AE: adverse event; BC: blood cultures; BSI, bloodstream infection; CIED: cardiovascular implantable electronic device; CoNS: coagulase-negative staphylococci; DAL: dalbavancin; IE: infectious endocarditis; IM: intramuscular; LD: loading dose; LVAD: left ventricular assist device; MRSA: methicillin-resistant S. aureus; MSSA: methicillin-susceptible S. aureus; N/A: not applicable; NVE: native valve endocarditis; OD: once daily; OPAT: outpatient parenteral antibiotic therapy; ORI: oritavancin; PVE: prosthetic valve endocarditis; PWID: people who inject drugs; SCV: small colony variant; SOC: standard of care. Definitions: Clinical cure/success was defined, unless otherwise specified, as resolution of clinical signs of infection; as absence of clinical signs of infection [107]; as no further evidence of infection or microbiological evidence of infection control (clearance of cultures) [106]; as improvement in lesions and resolution of signs and symptoms at end of treatment [105]; as completed treatment course without change or addition of antibiotic therapy, and with no additional antibiotics commenced within 48 h of discontinuation of the targeted antimicrobial therapy [109]; as no clinical, laboratory, or microbiological evidence of persistent or recurring infection during a 90 day follow-up [108]; as resolution of signs and symptoms of IE with negative BCs after end of therapy [110]; and as no need for additional therapy, and no additional positive cultures at 90 days [113]. Microbiological cure was defined as a documented negative blood culture result or BC clearance, unless otherwise specified.

3.4. Oritavancin

3.4.1. Mechanism of Action and Indication

Oritavancin (ORI) is a second-generation semisynthetic lipoglycopeptide with an extensive tissue distribution, a high binding affinity for plasma proteins, and a long terminal half-life (393 h). With its concentration-dependent bactericidal action, it disrupts the membranes of Gram-positive bacteria causing depolarization and inhibits the production of cell wall peptidoglycan by binding either to D-Ala-D-Ala or to D-Ala-D-Lac residues [131]. This bactericidal action through multiple mechanisms is considered to confer a low probability of resistance development [130]. ORI acts against streptococci, as well as S. aureus and S. epidermidis, regardless of susceptibility to methicillin. Differently from DAL and telavancin, ORI retains activity against both VanA- and VanB-phenotype enterococci. In addition, it is active against VISA and vancomycin-resistant S. aureus (VRSA) [132].
ORI maintains activity inside the biofilms of MSSA, MRSA, and vancomycin-susceptible and resistant enterococci [133]. Notably, the activity of ORI is not limited to the extracellular environment but concentrates in lysosomes and effectively addresses pathogens persisting intracellularly, as occurs with the SCV phenotype [134].
The currently available evidence concerning ORI in vitro synergisms and experimental models of IE is discussed in Supplementary Material, Sections S4.1 and S4.2 [135,136,137,138,139].
In 2014 and 2015, ORI was approved by the FDA and EMA, respectively, for ABSSSI [140]. Similar to DAL, given its optimal spectrum, tissue penetration, prolonged half-life, and side effect profile, ORI was explored for multiple off-label indications in invasive Gram-positive infections [141].

3.4.2. Clinical Evidence in Infective Endocarditis

Presently, data on ORI off-label use are limited, as shown in Table 4 [142].
In the multicentre retrospective cohort studied by Morrisette et al., 40 patients were treated with DAL, 14 were treated with ORI, and two were treated with both. In the whole cohort, five people had IE; however, unfortunately, it is not possible to distinguish how many received ORI. The success rate was 100% among the three IE cases analyzed [107].
A multicentre retrospective analysis was conducted among four hospitals and several clinics. Out of 75 patients receiving ORI, four had IE. The most common pathogens were MSSA and MRSA, and 13.3% of the population were PWID. In the whole cohort, the main reasons for ORI use were IV-line placement avoidance (61.3%) and social/insurance barriers (46.7%). Three patients with IE achieved clinical cure, the fourth was readmitted due to chest pain during the second infusion, subsequently attributed to cocaine use [11].
A retrospective single-centre analysis was performed on a very complex population (100% PWID, 70% with psychiatric illness, 67% homeless) treated with ORI. Two out of 23 patients had tricuspid IE. The first patient had MSSA and received 30 days of prior therapy followed by a single 1200 mg ORI dose and obtained clinical cure. The second had MRSA IE and, after 47 days of inpatient treatment, received two 1200 mg doses of ORI one week apart, but was finally recorded as a clinical failure [143]. Two single cases of IE treated with ORI reported clinical and microbiological success obtained after valve replacement surgery [144,145]. In a case series, after inpatient antibiotic therapy, five PWID with IE (two due to MSSA, two due to MRSA, one due to group A/F Streptococcus) were selected for ORI due to active illicit drug use and risk for IV-line manipulation. Clinical success was achieved by three patients, while two were lost to follow-up [146]
Overall, we retrieved only 13 IE cases of various types that were treated with ORI 1200 mg single or repeated doses, which were caused by staphylococci for the most part and frequently affected people with reduced compliance. Results were commonly good.
Table 4. Clinical studies investigating the treatment of infective endocarditis with oritavancin.
Table 4. Clinical studies investigating the treatment of infective endocarditis with oritavancin.
AuthorsStudy DesignEndpointN° Patients/
IE Type
PathogensDosage and DurationCombination, Dosage OutcomesSafety
Stewart, C.L. et al., 2017 [145]Observational retrospective study
ORI as an off-label indication
Clinical cure10 infections (BSI 50%)
1 tricuspid NVE in a PWID with previous therapy: VAN (3 days), then CRO (4 days)
Streptococcus agalactiaeIE patient 1200 mg 1 dose and then dischargedNoClinical failure with need for valve replacement 3 months after ORI administrationNo AE reported
Ahiskali, A. et al., 2020 [143]Observational retrospective study on a vulnerable population of
PWID receiving ORI
Clinical cure23 infections (BSI 50%)
Previous therapy 100%.
2 IE, type unspecified
1 MSSA
1 MRSA
MSSA IE: single 1200 mg dose,
MRSA IE: two 1200 mg doses
NoIE subgroup:
Clinical cure 1 (MSSA),
Clinical failure 1 (MRSA)
AE in 8.7%, mild
Brownell, L.E. et al., 2020 [11]Multicentre observational retrospective study
ORI as primary treatment
Clinical cure 75 infections (ABSSSI 49%)
No previous treatment
4 IE, type unspecified
MSSA (31.5%) and MRSA (17.8%)All patients included received initial 1200 mg dose followed by 1200 or 800 mg weeklyNo data reportedIE subgroup:
Clinical cure 75%
Average hospital days avoided in IE: 18 d
AE in 12%, most commonly back pain with infusion. All resolved upon discontinuation
Salcedo, D.A.T. et al., 2018 [146]Case series of
Gram-positive IE in PWID
N/A5 IE
Previous therapy 100%.
MRSA (20%), MSSA (20%), Streptococcus (10%)2 received 4 ORI doses, 3 received only 1 doseNoClinical cure: 3/5
Lost to FU: 2/5
AE in 1 patient (allergic reaction treated with oral prednisone)
Johnson, J.A. et al., 2015 [144]Case report
Limited treatment options
N/A1
Aortic PVE
VR E. faecium.1200 mg every other day for 3 doses, then weekly for 6 weeks, then
1200 mg biweekly for 10 weeks after recurrence and valve exchange
GEN for the first 4 days, discontinued due to renal toxicityRecurrence after the first treatment course attributed to lack in source control.
Clinical cure after valve exchange and a second prolonged course of ORI
Mild increase in
transaminases
Abbreviations: ABSSSI: acute bacterial skin and skin structure infection; AE: adverse event; IE: infectious endocarditis; MRSA: methicillin-resistant S. aureus; MSSA: methicillin-susceptible S. aureus; N/A: not applicable; ORI: oritavancin; PVE: prosthetic valve endocarditis; NVE: native valve endocarditis; PWID: people who inject drugs; VR: vancomycin resistant; VAN: vancomycin; GEN: gentamycin; CRO: ceftriaxone, FU: follow-up. Definitions: Clinical cure was defined as the resolution of all clinical signs and symptoms of infection or without need for additional antimicrobial therapy following completion of ORI.

3.5. Old Antibiotics with a Renewed Interest: Fosfomycin

3.5.1. Mechanism of Action and Indication

Fosfomycin (FOS) is a broad-spectrum bactericidal agent, with activity against several Gram-negative and Gram-positive pathogens, that enters the bacterial cell through the L-alpha-glycerophosphate and the hexose-6-phosphate transporter systems and acts by interfering with the formation of the peptidoglycan precursor uridine diphosphate N-acetylmuramic acid (UDP-MurNAc) [147]. This feature makes cross-resistance with other antibiotics highly uncommon [148].
Although discovered more than four decades ago, its use has only recently been repurposed for the treatment of severe infections caused by Gram-negative MDR [147,149,150,151] or Gram-positive pathogens such as MSSA/MRSA and VRE, showing promising results in terms of clinical efficacy and safety [10,148,152].
Indeed, its unique mechanism of action, along with its high level of in vitro synergism and its extensive tissue distribution, even in difficult-to-reach areas, renders FOS a very promising combination partner for the treatment of several infections, including IE [147,148].
Studies investigating FOS in vitro synergisms and experimental models of IE are shown in Supplementary Material, Sections S5.1 and S5.2 [153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177].
Current drug indications for FOS, namely infections for which no other antibiotics may be recommended, include complicated urinary tract infections, IE, bone and joint infections, pneumonia, skin and soft tissue infections, intra-abdominal infections, and meningitis, with or without bacteraemia [178].

3.5.2. Clinical Evidence in Infective Endocarditis

Clinical experience concerning the possible role of FOS-containing combinations for the treatment of Gram-positive IE has accumulated over time. Translating from in vitro and in vivo experiments, the most studied combinations were DAP and FOS and imipenem and FOS (Table 5).
The first report concerning the combination of imipenem and FOS dates back to 1994 [179]. Subsequently, Del Rio et al. performed a clinical trial including adults receiving appropriate antibiotic therapy for MRSA bacteraemia or IE but who needed imipenem and FOS as rescue therapy because of persistent bacteraemia, unacceptable side effects of antibiotics, or relapse. Among the 16 patients included, 12 suffered from IE. Overall, the primary outcome (defined as negative blood cultures 72 h after the first dose) was reached in all the patients, with no breakthrough episodes of MRSA bacteraemia and an overall clinical success rate of 91.6% [180].
In 2018, Pericas et al. performed an RCT comparing patients receiving imipenem and FOS with VAN for the treatment of MRSA BSI, among whom eight had IE (four in each regimen). The primary endpoint was persistent bacteraemia at seven days while secondary endpoints were the clearance of blood cultures at 72 h after the initiation of study treatment, relapse of bacteraemia, and mortality. Persistent bacteraemia was absent and blood cultures at 72 h were negative in all patients receiving imipenem and FOS, while cure rates were similar between the two regimens (4/8 vs. 3/7 imipenem and FOS vs. VAN, respectively) [181].
Subsequently, Pujol and colleagues performed an RCT comparing DAP (10 mg/kg/24 h) and FOS (2 g every 6 h) with DAP alone (10 mg/kg/24 h) for the treatment of MRSA BSI. Of the 155 patients included, 112 underwent echocardiography and 18/112 (11.6%) had left-side IE. Combination therapy achieved treatment success in a higher number of patients, although it was not statistically significant (54.1% vs. 42%). Notably, microbiological failure was significantly lower in the combination arm than in the monotherapy arm (0% vs. 11.1%). After stratification for patients with or without IE, no differences were observed. On the other hand, side effects were higher in patients receiving DAP and FOS than those receiving DAP alone [10].
A post hoc analysis of the INSTINCT prospective cohort study, including 578 patients with S. aureus bacteraemia, among whome 129 had IE, evaluated combination therapy with either rifampin (n = 242) or FOS (n = 58) versus monotherapy. The authors found that combination therapy was associated with a better outcome than monotherapy, and this was also observed in the subgroup of patients with IE. No differences between the rifampin of FOS combinations were observed for 90 day mortality [182,183]. The DAP or VAN and FOS combination was also reported in the case reports and case series [184,185,186].
Overall, we analyzed 294 IE episodes, mostly caused by MRSA and treated mainly with FOS in combination with different ß-lactams or DAP/VAN. When the data were reported, the native or prosthetic valves of the left side were predominantly involved. Clinical and microbiological outcomes were generally positive, leading the DAP and FOS regimen to be included in the recent guidelines [5].
Table 5. Clinical studies investigating the treatment of infective endocarditis with fosfomycin.
Table 5. Clinical studies investigating the treatment of infective endocarditis with fosfomycin.
AuthorsStudy DesignEndpointN° Patients/
IE Type
PathogensDosage and DurationCombination, Dosage OutcomesSafety
Del Rio, A. et al., 2014
[180]
Multicentre prospective clinical trial
IMI + FOS as rescue therapy for MRSA BSI
Primary endpoints: negative BC at 72 h,
clinical success § rate assessed
at the test-of-cure visit in the ITT population
16 BSI
12 IE
MRSA2 g/6 h *
Median 28 d
(SD 4–75)
IMI 1 g/6 h *Overall, negative BC 72 h after the first dose in all the patients,
No MRSAB breakthrough episodes,
Clinical success: 91.6%,
Mortality: 5 (31%), only 1 related to the infection or to the antibiotic therapy
5/16 (31%)
1: leukopenia
1: fungal BSI
3: sodium overload
Rieg, S. et al., 2017
[183]
Post hoc analysis of the INSTINCT prospective multicentre
cohort study
Patients with SAB
All-cause 30 d and 90 d mortality, death, or SAB-related late complications within 180 days964 BSI (452 monotherapy and 512 combination)
FOS was used in 99/512 (19%)
121 (12.6%) IE
[20/512 (4.4%)
monotherapy,
101/452 (19.7%)
combination]
MRSA 108/964 (11.2%)
MSSA
856/964 (88.8%)
5 g/8 h
Median duration 14 d (IQR 7–26, range 1–66)
MSSA:
FLU, VAN, TEC, DAP
MRSA:
VAN, TEIC, DAP, LNZ
Overall, 30 d mortality: monotherapy 82/443 (18.5%), combination
93/509 (18.3%), (p = 1)
90 d mortality: monotherapy 140/436 (32.1%), combination 156/503 (31%), (p = 0.87)
SAB-related late complications within 180 d: monotherapy 25/428 (5.8%), combination 19/490 (3.9%), (p = 0.18)
No specific outcomes in patients receiving FOS
No data reported
Pericas, J.M. et al., 2018
[181]
Open-label randomised clinical trial
IMI + FOS vs. VAN for MRSA BSI
Primary endpoint: persistent bacteraemia at 7 d
Secondary endpoints: negative BC at 72 h after the initiation of study treatment,
relapse of BSI,
mortality
15 BSI
8 IE
FOS + IMI (n = 8)
(4 complicated BSI, 4 IE: 2 NVE, 2 PVE)
VAN (n = 7)
(3 complicated BSI, 1 NVE, 3 CIED-IE)
MRSA2 g/6 h
EI group,
VAN: mean 35.7 d (range 27–42), IMI + FOS: mean 18.2 d (range 4–51)
Complicated bacteraemia
VAN: mean 18.3 d (range 17–21), IMI + FOS:
mean 27.2 d (range 15–42)
IMI 1 g/6 h
VAN 30–45 mg/kg/24 h (divided into 2–3 doses, trough levels ≥ 15 mg/L)
Overall, all patients in the FOS + IMI arm had negative BC at 3 days
Cure rates: IMI + FOS 4 (50%) VAN 3 (43%)
In-hospital mortality: IMI + FOS 3 (37.5%),
VAN 1 (14.2%)
Persistent bacteriemia:
IMI + FOS 0, VAN 1 (14.2%)
Relapse: IMI + FOS 0,
VAN 1 (14.2%)
IMI + FOS: 1 salt overload
VAN: 1 renal toxicity
Rieg, S. et al., 2020
[182]
Post hoc analysis of the INSTINCT prospective multicentre cohort study
Patients with SAB
All-cause 90 d mortality, death, or SAB-related late complications within 180 days578 BSI
[313 combination with RIF (n = 242) or FOS (n = 58) and 265 monotherapy
129 IE,
23% NVE, 7,1% of CIED or vascular grafts or PVE
MSSA
250 (94%) monotherapy
264 (84%)
combination
MRSA
15 (6%) monotherapy
49 (16%) combination
5 g/8 h
Median 23 d (IQR 13–33)
MSSA:
FLU or DAP
MRSA:
VAN, TEIC, DAP, LNZ
Overall, all-cause 90 d mortality: 190/565 (34%),
Death or SAB-related late complications within 180 d:
45% [52% (132/255) monotherapy vs. 39% (115/297) combination],
Combination therapy was associated with a better outcome than monotherapy (HR 0.65, 95% CI 0.46–0.92), especially in implanted foreign devices.
IE subgroup:
90 d mortality: 16/32 (50%) monotherapy, 27/81 (33%) RIF, 4/11 (36%) FOS
No data reported
Pujol, M. et al., 2021
[10]
Randomised clinical trial
DAP + FOS vs. DAP for MRSA BSI
Treatment success 6 weeks after the end of therapy155 BSI
18 left-side IE
MRSA2 g/6 h
DAP + FOS: median 14 d (IQR 11–21)
DAP 10 mg/kg/24 h
DAP
Median 14 days (IQR 10–18.5)
Overall, treatment success °: DAP + FOS 40/74 (54.1%), DAP 34/81 (42.0%) (p = 0.135)
Microbiological failure °°: DAP + FOS 0, DAP 9/81 (11.1%)
(p = 0.003)
Persistent bacteraemia at 7 d: DAP + FOS 0, DAP 5/81 (6.2%)

Complicated bacteraemia: DAP + FOS 12/74 (16.2%), DAP
26/81 (32.1%) (p = 0.022)
No differences were observed in patients with or without IE
DAP + FOS 13/74 (17.6%)
DAP 4/81 (4.9%)
(p = 0.018)
Aoyagi, S. et al., 1994
[179]
Case reportN/A1
IE on ventricular patch graft
MRSA300 mg/6 h
(paediatric dosage)
24 d
IMI 125 mg/6 h
(paediatric dosage)
Clearance of bacteraemia: 24 h from FOS start
Symptom-free during 12 months of follow-up
No data reported
Chen, L.Y. et al., 2011 [184]Case reportN/A1
CIED-IE plus osteomyelitis
DNS
MRSA
6 g/6 h
56 d
DAP 9 mg/kg/24 h, followed by 12 mg/kg/24 hClearance of bacteraemia: 7 d
Symptom free during 12 months of follow-up
No AE reported
Mirò, J.M. et al., 2012
[185]
Case series
Failure with high-dose DAP or VAN
N/A3 IE (1 aortic PVE, 2 left-sided NVE)1 MSSA (PVE)
2 MRSA (NVE)
2 g/6 h
6 weeks
DAP 10 mg/kg/24 hClearance of bacteraemia
Alive at 6 months (n = 1) and 12 months (n = 2) FU
No need of surgery
No AE reported
Vergara-Lopez, S. et al., 2015
[186]
Case reportN/A1
Aortic NVE
MRSE +
carbapenem-resistant Klebsiella
oxytoca
4 g/6 h
28 d
VAN (1 g/12 h)
AMK (1 g/24 h)
Clearance of bacteraemia
Complete disappearance of the vegetation at echocardiography
Self-limited hypokalaemia
Abbreviations: CIED-EI: cardiovascular implantable electronic device endocarditis; IE: infective endocarditis; FOS: fosfomycin; DAP: daptomycin; MRSA: methicillin-resistant S. aureus; MRSE: methicillin-resistant S. epidermidis; VAN: vancomycin; AMK: amikacin; IMI: imipenem; BC: blood culture; ITT: intention-to-treat; BSI: bloodstream infection; INSTINCT: invasive stapyhlococcus aureus infection; CohorT; SAB: S. aureus bacteraemia; MSSA: methicillin-susceptible S. aureus; FLU: flucloxacillin; TEC: teicoplanin; LNZ: linezolid; PVE: prosthetic valve endocarditis; MRSAB: methicillin-resistant S. aureus bacteraemia. Definitions: Clinical success was defined as clinical improvement with resolution of all signs and symptoms of infection during treatment or at the end of therapy unless otherwise specified. Notes: §: Treatment was classified as clinically successful when the patient was alive, lacked signs or symptoms of infection, and had sterile blood cultures at the test-of-cure visit. Failure was defined as death, positive blood cultures, or discontinuation of FOS plus IMI because of persistent bacteraemia or AEs; *: Between 2001 and 2005, all patients received VAN as initial therapy; this was continued, and FOS and IMI were added. After 2006, FOS and IMI were administered instead of the initial antibiotic regimen, which included either DAP at 6–10 mg/kg or VAN; °: Treatment success was considered when patient was alive and had resolution of clinical manifestations of infection and negative blood cultures at test-of-cure after completion of therapy; °°: Microbiological failure was considered in the case of persistent bacteraemia, recurrent bacteraemia, and the emergence of resistance to study drugs during treatment.

4. Oral Strategies

There has been great interest in oral step-down strategies for the treatment of IE; however, most of the evidence comes from old trials or retrospective and observational studies, with controversial results [187,188,189,190,191].
It is only with the recent multicentre unblinded non-inferiority POET trial that the long-lasting paradigm of treating IE always (and only) with prolonged intravenous treatment has changed. Indeed, this trial was able to show that, in stable patients with Streptococcus spp., E. faecalis, S. aureus, or CoNS left-side IE, changing to oral antibiotics after an initial phase of at least 10 days of intravenous treatment was not inferior to continued intravenous antibiotic treatment [192]. However, it should be noted that only 22% of the enrolled patients had S. aureus IE, only a small percentage of patients with IV drug use was included, and, although it was not an exclusion criterion, no patients with MRSA-IE or other antibiotic-resistant phenotypes were enrolled, rendering the results not fully generalizable. Among the several proposed schemes, the most commonly used during the trial were dicloxacillin or amoxicillin and rifampicin for S. aureus, linezolid and rifampicin or fusidic acid for CoNS, amoxicillin and linezolid or moxifloxacin for E. faecalis, and amoxicillin and rifampicin or moxifloxacin for streptococci [192].
The five-year follow-up of the same trial demonstrated that the composite primary outcome (defined as death from any cause, unplanned cardiac surgery, embolic events, and relapse of a blood culture result positive for the primary pathogen) occurred in 32.8% and 45.2% of step-down and continued intravenous treatment groups, respectively. Interestingly, this difference was mainly driven by a lower incidence of death from any cause in the first group, while no differences were observed for the other parameters of the composite outcome [193].
Taken together, these findings appear somehow reassuring concerning the potential role of oral step-down therapy for the treatment of selected and stable patients with left-side IE.
A recent published multicentre retrospective cohort confirmed this potential role, with no significant difference between the IV-only and oral groups in terms of clinical success at 90 days. Moreover, the oral group patients had significantly fewer adverse events. In this cohort, the most commonly used therapy was 600 mg of oral linezolid twice a day with or without rifampin [13]. Focused on E. faecalis IE, a small case series proposed an interesting oral step-down combination therapy with amoxicillin/clavulanate and cefditoren [194]. In a study published in 2009, the authors proposed an early switch from intravenous VAN to oral linezolid for the treatment of MRSA IE only after an aggressive surgical approach. This oral step-down showed a reduction in recurrences, hospitalization, and economic costs [195].
Possible oral strategies for the sequential step-down therapy are shown in Table 6.
Additional results will be available after the completion of the RODEO trials, which will compare oral switch and intravenous antibiotic therapies in patients with staphylococcal and streptococcal/enterococcal left-sided IE (RODEO-1 and RODEO-2, respectively) [196].
Tedizolid phosphate (TDZ) is a second-generation form of oxazolidinone. Compared to linezolid, TDZ is administered once daily with less myelotoxicity and fewer drug–drug interactions. There is no clinical data on TDZ in human IE. Based on in vitro and in vivo activity, TDZ may be considered a possible agent for the treatment of IE only as a sequential therapy after IV treatment with other agents in patients not eligible for other regimens [197,198]. Due to the lack of clinical evidence, no recommendation on its use for IE may be given and it remains a potential candidate without sufficient clinical evidence.
Table 6. Possible oral strategies for sequential step-down therapy. The decision to use sequential step-down oral therapy must only be made if the patient is clinically stable, and the choice of drug regimen must always be based on the antimicrobial susceptibility of the bacteria isolated (adapted from [192]).
Table 6. Possible oral strategies for sequential step-down therapy. The decision to use sequential step-down oral therapy must only be made if the patient is clinically stable, and the choice of drug regimen must always be based on the antimicrobial susceptibility of the bacteria isolated (adapted from [192]).
BacteriaOral Antibiotic Strategies for Step-Down Treatment #
MSSA/
MS-CONS
Dicloxacillin
+
rifampicin/fusidic acid
Levofloxacin/moxifloxacin
+
rifampicin/fusidic acid
Linezolid monotherapy
or
linezolid + adjunctive therapy
TMP-SMX + adjunctive therapy
MRSALinezolid *°
MR CONSLinezolid
+
levofloxacin/moxifloxacin
Levofloxacin/moxifloxacin
+
rifampicin/fusidic acid/clindamycin
Linezolid monotherapy
or
linezolid + rifampicin
TMP-SMX + adjunctive therapy
Oral Streptococci/
Streptococcus spp.
Amoxicillin monotherapy
or
amoxicillin + rifampicin
Moxifloxacin
+
rifampicin/clindamycin/amoxicillin
Linezolid monotherapy
or
linezolid + rifampicin/clindamycin/amoxicillin
Moxifloxacin
+
linezolid
E. faecalisAmoxicillin/clavulanate + cefditoren °
or
amoxicillin + rifampicin
Moxifloxacin
+
Amoxicillin/rifampicin
Linezolid monotherapy
or
linezolid
+
amoxicillin/rifampicin
Moxifloxacin
+
linezolid
GISA
(hVISA, VISA, DNS)
NOT RECOMMENDED
(No data available)
E. faecium
VVR Enterococcus spp.
Legend: # Only used in stable patients and always based on the antimicrobial susceptibility; * after surgical intervention; ° need of future investigations; adjunctive therapy: rifampicin, clindamycin, or fusidic acid. MSSA: methicillin-susceptible S. aureus; MRSA: methicillin-resistant S. aureus; CoNS: coagulase-negative Staphylococci; VISA: vancomycin-intermediate S. aureus; hVISA: heterogeneus vancomycin-intermediate S. aureus; DNS: Damptomycin-unsusceptible, VR: vancomycin-resistant; MS: methicillin-susceptible; MR: methicillin-resistant.

5. New Therapeutic Strategies: Considerations for Their Optimal Use in IE

IE is a major public health challenge associated with high morbidity and mortality [2]. Recently released guidelines have introduced several updates regarding its prevention, diagnosis, and management [5]. From a therapeutic point of view, by introducing the possibility of a step-down oral strategy in selected stable patients, the new recommendations divided the antibiotic treatment of IE into two phases: the first one (critical phase), which can last up to 2 weeks, includes in-hospital intravenous therapy using combinations of rapidly bactericidal antibiotics to destroy planktonic bacteria; after this period, selected clinically stable patients can end the antibiotic treatment at home with intravenous (OPAT) or oral antibiotic regimens for up to 6 weeks (continuation phase) [5].
Compared to the previous 2015 guidelines, the choice of antibiotics in the first phase has been expanded with the introduction of new molecules and combinations, including, among others, the combination DAP and FOS or CPT for MSSA and MRSA. As for the consolidation phase, weekly DAL schemes as an alternative to oral or OPAT strategies have been considered [5,6].
In the present manuscript, we reviewed the currently available in vitro, in vivo, and clinical evidence on the use of new beta-lactams (CPT, BPR), long-acting agents (DAL and ORI), and the repurposed drug FOS for their possible use in the treatment of IE.
As shown in Figure 1A, the evidence supporting the use of CPT and BPR (alone or in combination with DAP), FOS, and long-acting DAL and ORI for staphylococcal IE has accumulated over time [7,9,10,11,14,39,65,68,182]. Despite exhibiting pre-clinical evidence, the new beta-lactams and their associations with DAP have garnered less clinical evidence for MSSA IE, which has been limited to case series/case reports (shown as yellow or yellow/green colour, Figure 1A); this could be possibly explained by the strong efficacy of the currently recommended agents (i.e., cefazolin) [39,65].
In contrast, the combination of DAP and FOS has gained clinical evidence supporting its use thanks to the RCT by Pujol et al. (shown as green colour, Figure 1A). Likewise, for MRSA the combinations of DAP and FOS and DAP and CPT gained pre-clinical and clinical evidence supported by the RCTs by Pujol et al. and by Geriak et al., respectively, as well as by observational studies [8,10]. Choosing one of these two regimens over the other should be based on several factors, including beta-lactam allergies, which favuor DAP and FOS, or the risk of exacerbating cardiac or renal failure with the sodium overload associated with FOS, a condition favouring DAP and CPT.
According to the promising results of the recent ERADICATE RCT, which included 20 patients with S. aureus IE, a green/yellow colour was attributed to BPR for S. aureus, similar to the evidence available for BPR and DAP (Figure 1A) [39]. However, we believe that the use of BPR for the treatment of staphylococcal IE (alone or in combination with DAP) will increase over time.
As for the long-acting agents, so far, the majority of clinical evidence is available for DAL, especially with regard to MSSA and MRSA (shown as green colour, Figure 1A). Nevertheless, the most effective administration schedule is still not clear, since high variability is present in the literature concerning the number of dosages, their interval, and the duration of therapy [96]. Consensus agreement in this setting is highly warranted. In contrast, ORI’s clinical evidence for MSSA and MRSA is limited only to case reports/case series (shown as green/yellow colour, Figure 1A), probably due to its only recent introduction in the market [142]. However, based on ORI in vitro activity towards these pathogens, it is likely that additional clinical evidence will accumulate in the coming years, positioning ORI as a potential additional therapeutic strategy in the treatment of IE.
Although supported by less clinical evidence than S. aureus, the same considerations mentioned above may be drawn for CoNS (Figure 1A).
Since strong and consolidated clinical evidence exists concerning the management of beta-lactam-susceptible E. faecalis and streptococcal IE, we only reviewed the available literature data on the potential use of new agents for IE.
As shown by Figure 1B, most of the evidence regarding CPT+/−DAP or the long-acting drugs for streptococcal IE comes from evidence supported by in vitro activity, animal studies, and case reports/series (shown as yellow/green colour, Figure 1B), while, for BPR or beta-lactams and FOS, evidence is supported by in vitro activity and animal studies in the absence of clinical evidence for their effectiveness against streptococcal IE (shown as yellow colour, Figure 1B). As for E. faecalis IE, beta-lactams and FOS or CPT+/−DAP present poor in vitro data and no in vivo and clinical evidence and therefore are shown as yellow/red colour (Figure 1B).
Likewise, the combinations FOS or BPR and DAP for streptococcal IE present an absence of in vitro, animal, and clinical data (shown as red colour, Figure 1B). BPR in combination with ampicillin was investigated in a small series of E. faecalis IE cases, showing promising results [40] (shown as yellow/green colour, Figure 1B).
Much less knowledge has been gained concerning E. faecium or VAN-R enterococcal IE, where the currently available evidence only comes from in vitro and animal studies, while clinical evidence is still lacking (yellow/red or red colour, Figure 1B). In this regard, a recent study showed that the combination of high-dose daptomycin with FOS improved the survival rate of patients with VRE-BSI compared to daptomycin alone. However, only one case of IE was included, which was treated with DAP alone [152]. Additional clinical evidence on the potential role of DAP and FOS in the setting of IE is therefore needed.
The only regimen whose evidence is supported also by clinical evidence is DAL for E. faecalis IE, which therefore may be considered as a possible strategy after the initial phase of in-hospital intravenous therapy when other options are not feasible and may be associated with cost-effectiveness and reductions in hospitalization lengths [9,110]. Although active in vitro, ORI suffers from a lack or paucity (only case reports/case series) of clinical evidence concerning E. faecium and E. faecalis IE. However, similar to what we have hypothesised concerning staphylococcal IE, we believe that, as evidence accumulates, ORI will be an important therapeutic step-down regimen for enterococcal IE.

6. Conclusions

In conclusion, while for streptococcal, MSSA, and E. faecalis IE the use of new drugs/strategies may be only limited to particular cases since the currently recommended regimens are highly effective and well tolerated, the treatment of staphylococcal IE cases, in particular those sustained by MRSA and methicillin-resistant CoNS, may benefit from new strategies including: (i) CPT/BPR, alone or in combination with DAP, (ii) FOS in association with DAP, or (iii) long-acting DAL and ORI as step-down treatments.
Overall, only poor evidence is currently available concerning the potential roles of these new strategies for the treatment of E. faecium IE (only limited to cases when current recommended regimens are not feasible or effective) and vancomycin-resistant enterococcal IE, which represents one of the most difficult to treat conditions. We strongly believe that additional studies aiming to fill this gap are warranted.
A multidisciplinary approach to IE is highly recommended in order to use, as best as possible, the new therapeutic weapons we have at our disposal, which should be defended in accordance with antimicrobial stewardship principles.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm12247693/s1, supplementary sections on studies investigating in vitro synergisms of new antimicrobials and experimental animal models of IE.

Author Contributions

Conceptualization, M.V. and A.O.; data curation, A.O., F.C.D., L.V., F.C., A.C. and A.F.; methodology, A.O.; supervision, M.V.; writing—original draft, A.O., F.C.D., L.V., F.C., A.C. and A.F.; writing—review and editing, M.V. and A.O. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this review are retrieved and summarised from the different published studies available.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cahill, T.J.; Baddour, L.M.; Habib, G.; Hoen, B.; Salaun, E.; Pettersson, G.B.; Schäfers, H.J.; Prendergast, B.D. Challenges in Infective Endocarditis. J. Am. Coll. Cardiol. 2017, 69, 325–344. [Google Scholar] [CrossRef] [PubMed]
  2. Chen, H.; Zhan, Y.; Zhang, K.; Gao, Y.; Chen, L.; Zhan, J.; Chen, Z.; Zeng, Z. The Global, Regional, and National Burden and Trends of Infective Endocarditis from 1990 to 2019: Results From the Global Burden of Disease Study 2019. Front. Med. 2022, 9, 774224. [Google Scholar] [CrossRef] [PubMed]
  3. Selton-Suty, C.; Célard, M.; Le Moing, V.; Doco-Lecompte, T.; Chirouze, C.; Iung, B.; Strady, C.; Revest, M.; Vandenesch, F.; Bouvet, A.; et al. Preeminence of Staphylococcus aureus in Infective Endocarditis: A 1-Year Population-Based Survey. Clin. Infect. Dis. 2012, 54, 1230–1239. [Google Scholar] [CrossRef] [PubMed]
  4. Fowler, V.G.; Durack, D.T.; Selton-Suty, C.; Athan, E.; Bayer, A.S.; Chamis, A.L.; Dahl, A.; DiBernardo, L.; Durante-Mangoni, E.; Duval, X.; et al. The 2023 Duke-International Society for Cardiovascular Infectious Diseases Criteria for Infective Endocarditis: Updating the Modified Duke Criteria. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2023, 77, 518–526. [Google Scholar] [CrossRef]
  5. Delgado, V.; Ajmone Marsan, N.; de Waha, S.; Bonaros, N.; Brida, M.; Burri, H.; Caselli, S.; Doenst, T.; Ederhy, S.; Erba, P.A.; et al. 2023 ESC Guidelines for the Management of Endocarditis: Developed by the Task Force on the Management of Endocarditis of the European Society of Cardiology (ESC) Endorsed by the European Association for Cardio-Thoracic Surgery (EACTS) and the European Association of Nuclear Medicine (EANM). Eur. Heart J. 2023, 44, 3948–4042. [Google Scholar] [CrossRef] [PubMed]
  6. Habib, G.; Lancellotti, P.; Antunes, M.J.; Bongiorni, M.G.; Casalta, J.-P.; Del Zotti, F.; Dulgheru, R.; El Khoury, G.; Erba, P.A.; Iung, B.; et al. 2015 ESC Guidelines for the Management of Infective Endocarditis: The Task Force for the Management of Infective Endocarditis of the European Society of Cardiology (ESC). Endorsed by: European Association for Cardio-Thoracic Surgery (EACTS), the European Association of Nuclear Medicine (EANM). Eur. Heart J. 2015, 36, 3075–3128. [Google Scholar] [CrossRef] [PubMed]
  7. Gentile, I.; Buonomo, A.R.; Corcione, S.; Paradiso, L.; Giacobbe, D.R.; Bavaro, D.F.; Tiseo, G.; Sordella, F.; Bartoletti, M.; Palmiero, G.; et al. CEFTO-CURE Study: CEFTObiprole Clinical Use in Real-lifE—A Multi-Centre Experience in Italy. Int. J. Antimicrob. Agents 2023, 62, 106817. [Google Scholar] [CrossRef]
  8. Geriak, M.; Haddad, F.; Rizvi, K.; Rose, W.; Kullar, R.; LaPlante, K.; Yu, M.; Vasina, L.; Ouellette, K.; Zervos, M.; et al. Clinical Data on Daptomycin plus Ceftaroline versus Standard of Care Monotherapy in the Treatment of Methicillin-Resistant Staphylococcus aureus Bacteremia. Antimicrob. Agents Chemother. 2019, 63, e02483-18. [Google Scholar] [CrossRef]
  9. Hidalgo-Tenorio, C.; Sadyrbaeva-Dolgova, S.; Enríquez-Gómez, A.; Muñoz, P.; Plata-Ciezar, A.; Miró, J.M.; Alarcón, A.; Martínez-Marcos, F.J.; Loeches, B.; Escrihuela-Vidal, F.; et al. EN-DALBACEN 2.0 Cohort: Real-Life Study of Dalbavancin as Sequential/Consolidation Therapy in Patients with Infective Endocarditis Due to Gram-Positive Cocci. Int. J. Antimicrob. Agents 2023, 62, 106918. [Google Scholar] [CrossRef]
  10. Pujol, M.; Miró, J.-M.; Shaw, E.; Aguado, J.-M.; San-Juan, R.; Puig-Asensio, M.; Pigrau, C.; Calbo, E.; Montejo, M.; Rodriguez-Álvarez, R.; et al. Daptomycin Plus Fosfomycin Versus Daptomycin Alone for Methicillin-Resistant Staphylococcus aureus Bacteremia and Endocarditis: A Randomized Clinical Trial. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2021, 72, 1517–1525. [Google Scholar] [CrossRef]
  11. Brownell, L.E.; Adamsick, M.L.; McCreary, E.K.; Vanderloo, J.P.; Ernst, E.J.; Jackson, E.R.; Schulz, L.T. Clinical Outcomes and Economic Impact of Oritavancin for Gram-Positive Infections: A Single Academic Medical Center Health System Experience. Drugs-Real World Outcomes 2020, 7, 13–19. [Google Scholar] [CrossRef] [PubMed]
  12. Martí-Carvajal, A.J.; Dayer, M.; Conterno, L.O.; Gonzalez Garay, A.G.; Martí-Amarista, C.E. A Comparison of Different Antibiotic Regimens for the Treatment of Infective Endocarditis. Cochrane Database Syst. Rev. 2020, 5, CD009880. [Google Scholar] [CrossRef] [PubMed]
  13. Freling, S.; Wald-Dickler, N.; Banerjee, J.; Canamar, C.P.; Tangpraphaphorn, S.; Bruce, D.; Davar, K.; Dominguez, F.; Norwitz, D.; Krishnamurthi, G.; et al. Real-World Application of Oral Therapy for Infective Endocarditis: A Multicenter Retrospective, Cohort Study. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2023, 77, 672–679. [Google Scholar] [CrossRef] [PubMed]
  14. Tobudic, S.; Forstner, C.; Burgmann, H.; Lagler, H.; Ramharter, M.; Steininger, C.; Vossen, M.G.; Winkler, S.; Thalhammer, F. Dalbavancin as Primary and Sequential Treatment for Gram-Positive Infective Endocarditis: 2-Year Experience at the General Hospital of Vienna. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2018, 67, 795–798. [Google Scholar] [CrossRef] [PubMed]
  15. Deshpande, L.; Rhomberg, P.R.; Fritsche, T.R.; Sader, H.S.; Jones, R.N. Bactericidal Activity of BAL9141, a Novel Parenteral Cephalosporin against Contemporary Gram-Positive and Gram-Negative Isolates. Diagn. Microbiol. Infect. Dis. 2004, 50, 73–75. [Google Scholar] [CrossRef]
  16. Bogdanovich, T.; Ednie, L.M.; Shapiro, S.; Appelbaum, P.C. Antistaphylococcal Activity of Ceftobiprole, a New Broad-Spectrum Cephalosporin. Antimicrob. Agents Chemother. 2005, 49, 4210–4219. [Google Scholar] [CrossRef] [PubMed]
  17. Chong, Y.P.; Park, S.-J.; Kim, H.S.; Kim, E.S.; Kim, M.-N.; Kim, S.-H.; Lee, S.-O.; Choi, S.-H.; Jeong, J.-Y.; Woo, J.H.; et al. In Vitro Activities of Ceftobiprole, Dalbavancin, Daptomycin, Linezolid, and Tigecycline against Methicillin-Resistant Staphylococcus aureus Blood Isolates: Stratified Analysis by Vancomycin MIC. Diagn. Microbiol. Infect. Dis. 2012, 73, 264–266. [Google Scholar] [CrossRef]
  18. Pfaller, M.A.; Flamm, R.K.; Mendes, R.E.; Streit, J.M.; Smart, J.I.; Hamed, K.A.; Duncan, L.R.; Sader, H.S. Ceftobiprole Activity against Gram-Positive and -Negative Pathogens Collected from the United States in 2006 and 2016. Antimicrob. Agents Chemother. 2019, 63, e01566-18. [Google Scholar] [CrossRef]
  19. Hope, R.; Livermore, D.M.; Brick, G.; Lillie, M.; Reynolds, R. BSAC Working Parties on Resistance Surveillance Non-Susceptibility Trends among Staphylococci from Bacteraemias in the UK and Ireland, 2001–2006. J. Antimicrob. Chemother. 2008, 62 (Suppl. 2), ii65–ii74. [Google Scholar] [CrossRef]
  20. Pfaller, M.A.; Flamm, R.K.; Duncan, L.R.; Shortridge, D.; Smart, J.I.; Hamed, K.A.; Mendes, R.E.; Sader, H.S. Ceftobiprole Activity When Tested against Contemporary Bacteria Causing Bloodstream Infections in the United States (2016–2017). Diagn. Microbiol. Infect. Dis. 2019, 94, 304–313. [Google Scholar] [CrossRef]
  21. López Díaz, M.C.; Ríos, E.; Rodríguez-Avial, I.; Simaluiza, R.J.; Picazo, J.J.; Culebras, E. In-Vitro Activity of Several Antimicrobial Agents against Methicillin-Resistant Staphylococcus aureus (MRSA) Isolates Expressing Aminoglycoside-Modifying Enzymes: Potency of Plazomicin Alone and in Combination with Other Agents. Int. J. Antimicrob. Agents 2017, 50, 191–196. [Google Scholar] [CrossRef]
  22. Deshpande, L.M.; Jones, R.N. Bactericidal Activity and Synergy Studies of BAL9141, a Novel Pyrrolidinone-3-Ylidenemethyl Cephem, Tested against Streptococci, Enterococci and Methicillin-Resistant Staphylococci. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2003, 9, 1120–1124. [Google Scholar] [CrossRef]
  23. Rouse, M.S.; Steckelberg, J.M.; Patel, R. In Vitro Activity of Ceftobiprole, Daptomycin, Linezolid, and Vancomycin against Methicillin-Resistant Staphylococci Associated with Endocarditis and Bone and Joint Infection. Diagn. Microbiol. Infect. Dis. 2007, 58, 363–365. [Google Scholar] [CrossRef]
  24. Leonard, S.N.; Cheung, C.M.; Rybak, M.J. Activities of Ceftobiprole, Linezolid, Vancomycin, and Daptomycin against Community-Associated and Hospital-Associated Methicillin-Resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2008, 52, 2974–2976. [Google Scholar] [CrossRef]
  25. Rodríguez-García, R.; Rodríguez-Esteban, M.Á.; García-Carús, E.; Telenti, M.; Fernández, J. In Vitro Activity of Ceftaroline and Ceftobiprole against Clinical Isolates of Gram-Positive Bacteria from Infective Endocarditis: Are These Drugs Potential Options for the Initial Management of This Disease? Diagn. Microbiol. Infect. Dis. 2020, 98, 115153. [Google Scholar] [CrossRef]
  26. Campanile, F.; Bongiorno, D.; Mongelli, G.; Zanghì, G.; Stefani, S. Bactericidal Activity of Ceftobiprole Combined with Different Antibiotics against Selected Gram-Positive Isolates. Diagn. Microbiol. Infect. Dis. 2019, 93, 77–81. [Google Scholar] [CrossRef] [PubMed]
  27. Bongiorno, D.; Mongelli, G.; Stefani, S.; Campanile, F. Genotypic Analysis of Italian MRSA Strains Exhibiting Low-Level Ceftaroline and Ceftobiprole Resistance. Diagn. Microbiol. Infect. Dis. 2019, 95, 114852. [Google Scholar] [CrossRef]
  28. Peiffer-Smadja, N.; Guillotel, E.; Luque-Paz, D.; Maataoui, N.; Lescure, F.-X.; Cattoir, V. In Vitro Bactericidal Activity of Amoxicillin Combined with Different Cephalosporins against Endocarditis-Associated Enterococcus Faecalis Clinical Isolates. J. Antimicrob. Chemother. 2019, 74, 3511–3514. [Google Scholar] [CrossRef] [PubMed]
  29. Entenza, J.M.; Veloso, T.R.; Vouillamoz, J.; Giddey, M.; Majcherczyk, P.; Moreillon, P. In Vivo Synergism of Ceftobiprole and Vancomycin against Experimental Endocarditis Due to Vancomycin-Intermediate Staphylococcus aureus. Antimicrob. Agents Chemother. 2011, 55, 3977–3984. [Google Scholar] [CrossRef] [PubMed]
  30. Tattevin, P.; Basuino, L.; Bauer, D.; Diep, B.A.; Chambers, H.F. Ceftobiprole Is Superior to Vancomycin, Daptomycin, and Linezolid for Treatment of Experimental Endocarditis in Rabbits Caused by Methicillin-Resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2010, 54, 610–613. [Google Scholar] [CrossRef]
  31. Chambers, H.F. Evaluation of Ceftobiprole in a Rabbit Model of Aortic Valve Endocarditis Due to Methicillin-Resistant and Vancomycin-Intermediate Staphylococcus aureus. Antimicrob. Agents Chemother. 2005, 49, 884–888. [Google Scholar] [CrossRef] [PubMed]
  32. Lin, G.; Ednie, L.M.; Appelbaum, P.C. Antistaphylococcal Activity of ACHN-490 Tested Alone and in Combination with Other Agents by Time-Kill Assay. Antimicrob. Agents Chemother. 2010, 54, 2258–2261. [Google Scholar] [CrossRef] [PubMed]
  33. Barber, K.E.; Werth, B.J.; Ireland, C.E.; Stone, N.E.; Nonejuie, P.; Sakoulas, G.; Pogliano, J.; Rybak, M.J. Potent Synergy of Ceftobiprole plus Daptomycin against Multiple Strains of Staphylococcus aureus with Various Resistance Phenotypes. J. Antimicrob. Chemother. 2014, 69, 3006–3010. [Google Scholar] [CrossRef] [PubMed]
  34. Arias, C.A.; Singh, K.V.; Panesso, D.; Murray, B.E. Time-Kill and Synergism Studies of Ceftobiprole against Enterococcus Faecalis, Including Beta-Lactamase-Producing and Vancomycin-Resistant Isolates. Antimicrob. Agents Chemother. 2007, 51, 2043–2047. [Google Scholar] [CrossRef]
  35. Oltolini, C.; Castiglioni, B.; Tassan Din, C.; Castiglioni, A.; Ossi, C.; La Canna, G.; Pajoro, U.; Scarpellini, P. Meticillin-Resistant Staphylococcus aureus Endocarditis: First Report of Daptomycin plus Ceftobiprole Combination as Salvage Therapy. Int. J. Antimicrob. Agents 2016, 47, 502–504. [Google Scholar] [CrossRef]
  36. Mahmoud, E.; Al Mansour, S.; Bosaeed, M.; Alharbi, A.; Alsaedy, A.; Aljohani, S.; Alalwan, B.; Alothman, A. Ceftobiprole for Treatment of MRSA Blood Stream Infection: A Case Series. Infect. Drug Resist. 2020, 13, 2667–2672. [Google Scholar] [CrossRef] [PubMed]
  37. Tascini, C.; Attanasio, V.; Ripa, M.; Carozza, A.; Pallotto, C.; Bernardo, M.; Francisci, D.; Oltolini, C.; Palmiero, G.; Scarpellini, P. Ceftobiprole for the Treatment of Infective Endocarditis: A Case Series. J. Glob. Antimicrob. Resist. 2020, 20, 56–59. [Google Scholar] [CrossRef]
  38. Zhanel, G.G.; Kosar, J.; Baxter, M.; Dhami, R.; Borgia, S.; Irfan, N.; MacDonald, K.S.; Dow, G.; Lagacé-Wiens, P.; Dube, M.; et al. Real-Life Experience with Ceftobiprole in Canada: Results from the CLEAR (CanadianLEadership onAntimicrobialReal-Life Usage) Registry. J. Glob. Antimicrob. Resist. 2021, 24, 335–339. [Google Scholar] [CrossRef]
  39. Holland, T.L.; Cosgrove, S.E.; Doernberg, S.B.; Pavlov, O.; Titov, I.; Atanasov, B.; Gehr, M.A.; Engelhardt, M.; Hamed, K.; Ionescu, D.; et al. LB2302. Ceftobiprole Compared to Daptomycin with or without Optional Aztreonam for the Treatment of Complicated Staphylococcus aureus (SAB): Results of a Phase 3, Randomized, Double-Blind Trial (ERADICATE). Open Forum Infect. Dis. 2022, 9, ofac492.1892. [Google Scholar] [CrossRef]
  40. Giuliano, S.; Angelini, J.; D’Elia, D.; Geminiani, M.; Barison, R.D.; Giacinta, A.; Sartor, A.; Campanile, F.; Curcio, F.; Cotta, M.O.; et al. Ampicillin and Ceftobiprole Combination for the Treatment of Enterococcus Faecalis Invasive Infections: “The Times They Are A-Changin”. Antibiotics 2023, 12, 879. [Google Scholar] [CrossRef]
  41. Holland, T.L.; Cosgrove, S.E.; Doernberg, S.B.; Jenkins, T.C.; Turner, N.A.; Boucher, H.W.; Pavlov, O.; Titov, I.; Kosulnykov, S.; Atanasov, B.; et al. Ceftobiprole for Treatment of Complicated Staphylococcus aureus Bacteremia. N. Engl. J. Med. 2023, 389, 1390–1401. [Google Scholar] [CrossRef] [PubMed]
  42. Jorgenson, M.R.; DePestel, D.D.; Carver, P.L. Ceftaroline Fosamil: A Novel Broad-Spectrum Cephalosporin with Activity against Methicillin-Resistant Staphylococcus aureus. Ann. Pharmacother. 2011, 45, 1384–1398. [Google Scholar] [CrossRef] [PubMed]
  43. Espedido, B.A.; Jensen, S.O.; van Hal, S.J. Ceftaroline Fosamil Salvage Therapy: An Option for Reduced-Vancomycin-Susceptible MRSA Bacteraemia. J. Antimicrob. Chemother. 2015, 70, 797–801. [Google Scholar] [CrossRef] [PubMed]
  44. Kaushik, D.; Rathi, S.; Jain, A. Ceftaroline: A Comprehensive Update. Int. J. Antimicrob. Agents 2011, 37, 389–395. [Google Scholar] [CrossRef] [PubMed]
  45. Werth, B.J.; Barber, K.E.; Ireland, C.E.; Rybak, M.J. Evaluation of Ceftaroline, Vancomycin, Daptomycin, or Ceftaroline plus Daptomycin against Daptomycin-Nonsusceptible Methicillin-Resistant Staphylococcus aureus in an in Vitro Pharmacokinetic/Pharmacodynamic Model of Simulated Endocardial Vegetations. Antimicrob. Agents Chemother. 2014, 58, 3177–3181. [Google Scholar] [CrossRef] [PubMed]
  46. Dhand, A.; Bayer, A.S.; Pogliano, J.; Yang, S.-J.; Bolaris, M.; Nizet, V.; Wang, G.; Sakoulas, G. Use of Antistaphylococcal β-Lactams to Increase Daptomycin Activity in Eradicating Persistent Bacteremia Due to Methicillin-Resistant Staphylococcus aureus: Role of Enhanced Daptomycin Binding. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2011, 53, 158–163. [Google Scholar] [CrossRef]
  47. Werth, B.J.; Vidaillac, C.; Murray, K.P.; Newton, K.L.; Sakoulas, G.; Nonejuie, P.; Pogliano, J.; Rybak, M.J. Novel Combinations of Vancomycin plus Ceftaroline or Oxacillin against Methicillin-Resistant Vancomycin-Intermediate Staphylococcus aureus (VISA) and Heterogeneous VISA. Antimicrob. Agents Chemother. 2013, 57, 2376–2379. [Google Scholar] [CrossRef]
  48. Tran, N.; Rybak, M.J. β-Lactam Combinations with Vancomycin Show Synergistic Activity against Vancomycin-Susceptible Staphylococcus aureus, Vancomycin-Intermediate S. Aureus (VISA), and Heterogeneous VISA. Antimicrob. Agents Chemother. 2018, 62, e00157-18. [Google Scholar] [CrossRef]
  49. Eliazar, J.; Johnson, T.; Chbib, C. Pre-Clinical Impact of the Synergistic Mechanism of Daptomycin and Ceftaroline on Patients with Methicillin-Resistant Staphylococcus aureus Bacteremia Infections. Curr. Rev. Clin. Exp. Pharmacol. 2021, 16, 296–299. [Google Scholar] [CrossRef]
  50. Hutton, M.A.; Sundaram, A.; Perri, M.B.; Zervos, M.J.; Herc, E.S. Assessment of Invitrosynergy of Daptomycin or Vancomycin plus Ceftaroline for Daptomycin Non-Susceptible Staphylococcus aureus. Diagn. Microbiol. Infect. Dis. 2020, 98, 115126. [Google Scholar] [CrossRef]
  51. Werth, B.J.; Shireman, L.M. Pharmacodynamics of Ceftaroline plus Ampicillin against Enterococcus Faecalis in an In Vitro Pharmacokinetic/Pharmacodynamic Model of Simulated Endocardial Vegetations. Antimicrob. Agents Chemother. 2017, 61, e02235-16. [Google Scholar] [CrossRef] [PubMed]
  52. Thieme, L.; Klinger-Strobel, M.; Hartung, A.; Stein, C.; Makarewicz, O.; Pletz, M.W. In Vitro Synergism and Anti-Biofilm Activity of Ampicillin, Gentamicin, Ceftaroline and Ceftriaxone against Enterococcus Faecalis. J. Antimicrob. Chemother. 2018, 73, 1553–1561. [Google Scholar] [CrossRef]
  53. Cusumano, J.A.; Daffinee, K.E.; Piehl, E.C.; García-Solache, M.; Desbonnet, C.; Rice, L.B.; LaPlante, K.L. Meropenem plus Ceftaroline Is Active against Enterococcus Faecalis in an In Vitro Pharmacodynamic Model Using Humanized Dosing Simulations. Antimicrob. Agents Chemother. 2022, 66, e0042622. [Google Scholar] [CrossRef]
  54. Smith, J.R.; Barber, K.E.; Raut, A.; Aboutaleb, M.; Sakoulas, G.; Rybak, M.J. β-Lactam Combinations with Daptomycin Provide Synergy against Vancomycin-Resistant Enterococcus Faecalis and Enterococcus Faecium. J. Antimicrob. Chemother. 2015, 70, 1738–1743. [Google Scholar] [CrossRef] [PubMed]
  55. Mainardi, J.L.; Gutmann, L.; Acar, J.F.; Goldstein, F.W. Synergistic Effect of Amoxicillin and Cefotaxime against Enterococcus Faecalis. Antimicrob. Agents Chemother. 1995, 39, 1984–1987. [Google Scholar] [CrossRef] [PubMed]
  56. Sakoulas, G.; Moise, P.A.; Casapao, A.M.; Nonejuie, P.; Olson, J.; Okumura, C.Y.M.; Rybak, M.J.; Kullar, R.; Dhand, A.; Rose, W.E.; et al. Antimicrobial Salvage Therapy for Persistent Staphylococcal Bacteremia Using Daptomycin plus Ceftaroline. Clin. Ther. 2014, 36, 1317–1333. [Google Scholar] [CrossRef] [PubMed]
  57. Jacqueline, C.; Amador, G.; Batard, E.; Le Mabecque, V.; Miègeville, A.-F.; Biek, D.; Caillon, J.; Potel, G. Comparison of Ceftaroline Fosamil, Daptomycin and Tigecycline in an Experimental Rabbit Endocarditis Model Caused by Methicillin-Susceptible, Methicillin-Resistant and Glycopeptide-Intermediate Staphylococcus aureus. J. Antimicrob. Chemother. 2011, 66, 863–866. [Google Scholar] [CrossRef] [PubMed]
  58. Jacqueline, C.; Caillon, J.; Batard, E.; Le Mabecque, V.; Amador, G.; Ge, Y.; Biek, D.; Potel, G. Evaluation of the in Vivo Efficacy of Intramuscularly Administered Ceftaroline Fosamil, a Novel Cephalosporin, against a Methicillin-Resistant Staphylococcus aureus Strain in a Rabbit Endocarditis Model. J. Antimicrob. Chemother. 2010, 65, 2264–2265. [Google Scholar] [CrossRef]
  59. Jacqueline, C.; Caillon, J.; Le Mabecque, V.; Miègeville, A.-F.; Ge, Y.; Biek, D.; Batard, E.; Potel, G. In Vivo Activity of a Novel Anti-Methicillin-Resistant Staphylococcus aureus Cephalosporin, Ceftaroline, against Vancomycin-Susceptible and -Resistant Enterococcus Faecalis Strains in a Rabbit Endocarditis Model: A Comparative Study with Linezolid and Vancomycin. Antimicrob. Agents Chemother. 2009, 53, 5300–5302. [Google Scholar] [CrossRef]
  60. Jacqueline, C.; Caillon, J.; Le Mabecque, V.; Miègeville, A.-F.; Hamel, A.; Bugnon, D.; Ge, J.Y.; Potel, G. In Vivo Efficacy of Ceftaroline (PPI-0903), a New Broad-Spectrum Cephalosporin, Compared with Linezolid and Vancomycin against Methicillin-Resistant and Vancomycin-Intermediate Staphylococcus aureus in a Rabbit Endocarditis Model. Antimicrob. Agents Chemother. 2007, 51, 3397–3400. [Google Scholar] [CrossRef]
  61. García, P.; Moscoso, M.; Fernández, M.C.; Fuentes-Valverde, V.; Pérez, A.; Bou, G. Comparison of the in Vivo Efficacy of Ceftaroline Fosamil, Vancomycin and Daptomycin in a Murine Model of Methicillin-Resistant Staphylococcus aureus Bacteraemia. Int. J. Antimicrob. Agents 2023, 62, 106836. [Google Scholar] [CrossRef] [PubMed]
  62. Luther, M.K.; Rice, L.B.; LaPlante, K.L. Ampicillin in Combination with Ceftaroline, Cefepime, or Ceftriaxone Demonstrates Equivalent Activities in a High-Inoculum Enterococcus Faecalis Infection Model. Antimicrob. Agents Chemother. 2016, 60, 3178–3182. [Google Scholar] [CrossRef] [PubMed]
  63. EMA Zinforo. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/zinforo (accessed on 11 September 2023).
  64. Brandariz-Núñez, D.; Suanzes, J.; Gutiérrez-Urbón, J.M.; Fernández-Oliveira, C.; Margusino, L.; Martín-Herranz, I. Incidence and Risk Factors for Mortality in Patients Treated with Combined Ceftaroline for Gram-Positive Infective Endocarditis. Eur. J. Clin. Microbiol. Infect. Dis. Off. Publ. Eur. Soc. Clin. Microbiol. 2022, 41, 827–834. [Google Scholar] [CrossRef] [PubMed]
  65. Destache, C.J.; Guervil, D.J.; Kaye, K.S. Ceftaroline Fosamil for the Treatment of Gram-Positive Endocarditis: CAPTURE Study Experience. Int. J. Antimicrob. Agents 2019, 53, 644–649. [Google Scholar] [CrossRef] [PubMed]
  66. Casapao, A.M.; Davis, S.L.; Barr, V.O.; Klinker, K.P.; Goff, D.A.; Barber, K.E.; Kaye, K.S.; Mynatt, R.P.; Molloy, L.M.; Pogue, J.M.; et al. Large Retrospective Evaluation of the Effectiveness and Safety of Ceftaroline Fosamil Therapy. Antimicrob. Agents Chemother. 2014, 58, 2541–2546. [Google Scholar] [CrossRef]
  67. Britt, R.S.; Evoy, K.E.; Lee, G.C.; Reveles, K.R.; Sorensen, K.M.; Jones, X.; Bollinger, M.; Frei, C.R. Early Use of Ceftaroline Fosamil in the United States Veterans Health Care System. Drugs 2017, 77, 1345–1351. [Google Scholar] [CrossRef]
  68. Zasowski, E.J.; Trinh, T.D.; Claeys, K.C.; Lagnf, A.M.; Bhatia, S.; Klinker, K.P.; Veve, M.P.; Estrada, S.J.; Johns, S.T.; Sawyer, A.J.; et al. Multicenter Cohort Study of Ceftaroline Versus Daptomycin for Treatment of Methicillin-Resistant Staphylococcus aureus Bloodstream Infection. Open Forum Infect. Dis. 2022, 9, ofab606. [Google Scholar] [CrossRef]
  69. McCreary, E.K.; Kullar, R.; Geriak, M.; Zasowski, E.J.; Rizvi, K.; Schulz, L.T.; Ouellette, K.; Vasina, L.; Haddad, F.; Rybak, M.J.; et al. Multicenter Cohort of Patients With Methicillin-Resistant Staphylococcus aureus Bacteremia Receiving Daptomycin Plus Ceftaroline Compared With Other MRSA Treatments. Open Forum Infect. Dis. 2020, 7, ofz538. [Google Scholar] [CrossRef]
  70. Ahmad, O.; Crawford, T.N.; Myint, T. Comparing the Outcomes of Ceftaroline Plus Vancomycin or Daptomycin Combination Therapy Versus Monotherapy in Adults with Complicated and Prolonged Methicillin-Resistant Staphylococcus aureus Bacteremia Initially Treated with Supplemental Ceftaroline. Infect. Dis. Ther. 2020, 9, 77–87. [Google Scholar] [CrossRef] [PubMed]
  71. Cortes-Penfield, N.; Oliver, N.T.; Hunter, A.; Rodriguez-Barradas, M. Daptomycin and Combination Daptomycin-Ceftaroline as Salvage Therapy for Persistent Methicillin-Resistant Staphylococcus aureus Bacteremia. Infect. Dis. Lond. Engl. 2018, 50, 643–647. [Google Scholar] [CrossRef] [PubMed]
  72. Fabre, V.; Ferrada, M.; Buckel, W.R.; Avdic, E.; Cosgrove, S.E. Ceftaroline in Combination With Trimethoprim-Sulfamethoxazole for Salvage Therapy of Methicillin-Resistant Staphylococcus aureus Bacteremia and Endocarditis. Open Forum Infect. Dis. 2014, 1, ofu046. [Google Scholar] [CrossRef]
  73. Johnson, T.M.; Molina, K.C.; Miller, M.A.; Kiser, T.H.; Huang, M.; Mueller, S.W. Combination Ceftaroline and Daptomycin Salvage Therapy for Complicated Methicillin-Resistant Staphylococcus aureus Bacteraemia Compared with Standard of Care. Int. J. Antimicrob. Agents 2021, 57, 106310. [Google Scholar] [CrossRef]
  74. Kufel, W.D.; Parsels, K.A.; Blaine, B.E.; Steele, J.M.; Mahapatra, R.; Paolino, K.M.; Thomas, S.J. Vancomycin plus Ceftaroline for Persistent Methicillin-Resistant Staphylococcus aureus Bacteremia. Pharmacotherapy 2023, 43, 15–23. [Google Scholar] [CrossRef]
  75. Morrisette, T.; Lagnf, A.M.; Alosaimy, S.; Rybak, M.J. A Comparison of Daptomycin Alone and in Combination with Ceftaroline Fosamil for Methicillin-Resistant Staphylococcus aureus Bacteremia Complicated by Septic Pulmonary Emboli. Eur. J. Clin. Microbiol. Infect. Dis. Off. Publ. Eur. Soc. Clin. Microbiol. 2020, 39, 2199–2203. [Google Scholar] [CrossRef] [PubMed]
  76. Arshad, S.; Huang, V.; Hartman, P.; Perri, M.B.; Moreno, D.; Zervos, M.J. Ceftaroline Fosamil Monotherapy for Methicillin-Resistant Staphylococcus aureus Bacteremia: A Comparative Clinical Outcomes Study. Int. J. Infect. Dis. IJID Off. Publ. Int. Soc. Infect. Dis. 2017, 57, 27–31. [Google Scholar] [CrossRef]
  77. Hornak, J.P.; Anjum, S.; Reynoso, D. Adjunctive Ceftaroline in Combination with Daptomycin or Vancomycin for Complicated Methicillin-Resistant Staphylococcus aureus Bacteremia after Monotherapy Failure. Ther. Adv. Infect. Dis. 2019, 6, 2049936119886504. [Google Scholar] [CrossRef] [PubMed]
  78. Polenakovik, H.M.; Pleiman, C.M. Ceftaroline for Meticillin-Resistant Staphylococcus aureus Bacteraemia: Case Series and Review of the Literature. Int. J. Antimicrob. Agents 2013, 42, 450–455. [Google Scholar] [CrossRef] [PubMed]
  79. Tattevin, P.; Boutoille, D.; Vitrat, V.; Van Grunderbeeck, N.; Revest, M.; Dupont, M.; Alfandari, S.; Stahl, J.-P. Salvage Treatment of Methicillin-Resistant Staphylococcal Endocarditis with Ceftaroline: A Multicentre Observational Study. J. Antimicrob. Chemother. 2014, 69, 2010–2013. [Google Scholar] [CrossRef] [PubMed]
  80. Warren, E.F.; Crocker, R.J.; Tabor, B.; Pizzuti, M.; Tsai, Y.V.; Antosz, K.; Battle, S.; Ahuja, D.; Bookstaver, P.B. Successful Use of Nafcillin and Ceftaroline Combination Therapy for Persistent MSSA Bacteraemia and Endocarditis: A Case Series. JAC-Antimicrob. Resist. 2022, 5, dlac129. [Google Scholar] [CrossRef] [PubMed]
  81. Lin, S.-Y.; Hung, W.-C.; Lo, S.-H.; Tseng, Y.-T.; Lu, P.-L. Successful Treatment with Daptomycin and Ceftaroline of Heterogeneous Vancomycin-Intermediate Staphylococcus aureus (hVISA) Endocarditis: A Case Report. J. Glob. Antimicrob. Resist. 2021, 27, 335–336. [Google Scholar] [CrossRef] [PubMed]
  82. Jilani, T.N.; Masood, S.O. Ceftaroline Fosamil as an Alternative for a Severe Methicillin-Resistant Staphylococcus aureus Infection: A Case Report. Cureus 2018, 10, e3776. [Google Scholar] [CrossRef] [PubMed]
  83. Duss, F.-R.; Garcia de la Mària, C.; Croxatto, A.; Giulieri, S.; Lamoth, F.; Manuel, O.; Miró, J.M. Successful Treatment with Daptomycin and Ceftaroline of MDR Staphylococcus aureus Native Valve Endocarditis: A Case Report. J. Antimicrob. Chemother. 2019, 74, 2626–2630. [Google Scholar] [CrossRef] [PubMed]
  84. Sundaragiri, P.R.; Vallabhajosyula, S.; Haddad, T.M.; Esterbrooks, D.J. Tricuspid and Mitral Endocarditis Due to Methicillin-Resistant Staphylococcus aureus Exhibiting Vancomycin-Creep Phenomenon. BMJ Case Rep. 2015, 2015, bcr2015211974. [Google Scholar] [CrossRef]
  85. Cunha, B.A.; Gran, A. Successful Treatment of Meticillin-Resistant Staphylococcus aureus (MRSA) Aortic Prosthetic Valve Endocarditis with Prolonged High-Dose Daptomycin plus Ceftaroline Therapy. Int. J. Antimicrob. Agents 2015, 46, 225–226. [Google Scholar] [CrossRef] [PubMed]
  86. Baxi, S.M.; Chan, D.; Jain, V. Daptomycin Non-Susceptible, Vancomycin-Intermediate Staphylococcus aureus Endocarditis Treated with Ceftaroline and Daptomycin: Case Report and Brief Review of the Literature. Infection 2015, 43, 751–754. [Google Scholar] [CrossRef] [PubMed]
  87. Sakoulas, G.; Nonejuie, P.; Nizet, V.; Pogliano, J.; Crum-Cianflone, N.; Haddad, F. Treatment of High-Level Gentamicin-Resistant Enterococcus Faecalis Endocarditis with Daptomycin plus Ceftaroline. Antimicrob. Agents Chemother. 2013, 57, 4042–4045. [Google Scholar] [CrossRef]
  88. Jongsma, K.; Joson, J.; Heidari, A. Ceftaroline in the Treatment of Concomitant Methicillin-Resistant and Daptomycin-Non-Susceptible Staphylococcus aureus Infective Endocarditis and Osteomyelitis: Case Report. J. Antimicrob. Chemother. 2013, 68, 1444–1445. [Google Scholar] [CrossRef]
  89. Rose, W.E.; Schulz, L.T.; Andes, D.; Striker, R.; Berti, A.D.; Hutson, P.R.; Shukla, S.K. Addition of Ceftaroline to Daptomycin after Emergence of Daptomycin-Nonsusceptible Staphylococcus aureus during Therapy Improves Antibacterial Activity. Antimicrob. Agents Chemother. 2012, 56, 5296–5302. [Google Scholar] [CrossRef]
  90. Gritsenko, D.; Fedorenko, M.; Ruhe, J.J.; Altshuler, J. Combination Therapy With Vancomycin and Ceftaroline for Refractory Methicillin-Resistant Staphylococcus aureus Bacteremia: A Case Series. Clin. Ther. 2017, 39, 212–218. [Google Scholar] [CrossRef]
  91. Ho, T.T.; Cadena, J.; Childs, L.M.; Gonzalez-Velez, M.; Lewis, J.S. Methicillin-Resistant Staphylococcus aureus Bacteraemia and Endocarditis Treated with Ceftaroline Salvage Therapy. J. Antimicrob. Chemother. 2012, 67, 1267–1270. [Google Scholar] [CrossRef]
  92. Lin, J.C.; Aung, G.; Thomas, A.; Jahng, M.; Johns, S.; Fierer, J. The Use of Ceftaroline Fosamil in Methicillin-Resistant Staphylococcus aureus Endocarditis and Deep-Seated MRSA Infections: A Retrospective Case Series of 10 Patients. J. Infect. Chemother. Off. J. Jpn. Soc. Chemother. 2013, 19, 42–49. [Google Scholar] [CrossRef]
  93. Zasowski, E.J.; Trinh, T.D.; Claeys, K.C.; Casapao, A.M.; Sabagha, N.; Lagnf, A.M.; Klinker, K.P.; Davis, S.L.; Rybak, M.J. Multicenter Observational Study of Ceftaroline Fosamil for Methicillin-Resistant Staphylococcus aureus Bloodstream Infections. Antimicrob. Agents Chemother. 2017, 61, e02015-16. [Google Scholar] [CrossRef] [PubMed]
  94. Nichols, C.N.; Wardlow, L.C.; Coe, K.E.; Sobhanie, M.M.E. Clinical Outcomes With Definitive Treatment of Methicillin-Resistant Staphylococcus aureus Bacteremia With Retained Daptomycin and Ceftaroline Combination Therapy vs De-Escalation to Monotherapy With Vancomycin, Daptomycin, or Ceftaroline. Open Forum Infect. Dis. 2021, 8, ofab327. [Google Scholar] [CrossRef] [PubMed]
  95. Pani, A.; Colombo, F.; Agnelli, F.; Frantellizzi, V.; Baratta, F.; Pastori, D.; Scaglione, F. Off-Label Use of Ceftaroline Fosamil: A Systematic Review. Int. J. Antimicrob. Agents 2019, 54, 562–571. [Google Scholar] [CrossRef]
  96. Gatti, M.; Andreoni, M.; Pea, F.; Viale, P. Real-World Use of Dalbavancin in the Era of Empowerment of Outpatient Antimicrobial Treatment: A Careful Appraisal Beyond Approved Indications Focusing on Unmet Clinical Needs. Drug Des. Devel. Ther. 2021, 15, 3349–3378. [Google Scholar] [CrossRef]
  97. Zeng, D.; Debabov, D.; Hartsell, T.L.; Cano, R.J.; Adams, S.; Schuyler, J.A.; McMillan, R.; Pace, J.L. Approved Glycopeptide Antibacterial Drugs: Mechanism of Action and Resistance. Cold Spring Harb. Perspect. Med. 2016, 6, a026989. [Google Scholar] [CrossRef] [PubMed]
  98. Pfaller, M.A.; Mendes, R.E.; Duncan, L.R.; Flamm, R.K.; Sader, H.S. Activity of Dalbavancin and Comparator Agents against Gram-Positive Cocci from Clinical Infections in the USA and Europe 2015-16. J. Antimicrob. Chemother. 2018, 73, 2748–2756. [Google Scholar] [CrossRef] [PubMed]
  99. Oliva, A.; Stefani, S.; Venditti, M.; Di Domenico, E.G. Biofilm-Related Infections in Gram-Positive Bacteria and the Potential Role of the Long-Acting Agent Dalbavancin. Front. Microbiol. 2021, 12, 749685. [Google Scholar] [CrossRef]
  100. Johnson, D.M.; Fritsche, T.R.; Sader, H.S.; Jones, R.N. Evaluation of Dalbavancin in Combination with Nine Antimicrobial Agents to Detect Enhanced or Antagonistic Interactions. Int. J. Antimicrob. Agents 2006, 27, 557–560. [Google Scholar] [CrossRef]
  101. Xhemali, X.; Smith, J.R.; Kebriaei, R.; Rice, S.A.; Stamper, K.C.; Compton, M.; Singh, N.B.; Jahanbakhsh, S.; Rybak, M.J. Evaluation of Dalbavancin Alone and in Combination with β-Lactam Antibiotics against Resistant Phenotypes of Staphylococcus aureus. J. Antimicrob. Chemother. 2019, 74, 82–86. [Google Scholar] [CrossRef]
  102. Candiani, G.; Abbondi, M.; Borgonovi, M.; Romanò, G.; Parenti, F. In-Vitro and in-Vivo Antibacterial Activity of BI 397, a New Semi-Synthetic Glycopeptide Antibiotic. J. Antimicrob. Chemother. 1999, 44, 179–192. [Google Scholar] [CrossRef]
  103. Lefort, A.; Pavie, J.; Garry, L.; Chau, F.; Fantin, B. Activities of Dalbavancin In Vitro and in a Rabbit Model of Experimental Endocarditis Due to Staphylococcus aureus with or without Reduced Susceptibility to Vancomycin and Teicoplanin. Antimicrob. Agents Chemother. 2004, 48, 1061–1064. [Google Scholar] [CrossRef]
  104. Volpicelli, L.; Venditti, M.; Oliva, A. Acute Bacterial Skin and Skin Structure Infections in Pediatric Patients: Potential Role of Dalbavancin. Expert Rev. Anti Infect. Ther. 2023, 21, 329–341. [Google Scholar] [CrossRef] [PubMed]
  105. EMA Xydalba. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/xydalba (accessed on 11 September 2023).
  106. Bai, F.; Aldieri, C.; Cattelan, A.; Raumer, F.; Di Meco, E.; Moioli, M.C.; Tordato, F.; Morelli, P.; Borghi, F.; Rizzi, M.; et al. Efficacy and Safety of Dalbavancin in the Treatment of Acute Bacterial Skin and Skin Structure Infections (ABSSSIs) and Other Infections in a Real-Life Setting: Data from an Italian Observational Multicentric Study (DALBITA Study). Expert Rev. Anti Infect. Ther. 2020, 18, 1271–1279. [Google Scholar] [CrossRef] [PubMed]
  107. Morrisette, T.; Miller, M.A.; Montague, B.T.; Barber, G.R.; McQueen, R.B.; Krsak, M. On- and off-Label Utilization of Dalbavancin and Oritavancin for Gram-Positive Infections. J. Antimicrob. Chemother. 2019, 74, 2405–2416. [Google Scholar] [CrossRef] [PubMed]
  108. Dinh, A.; Duran, C.; Pavese, P.; Khatchatourian, L.; Monnin, B.; Bleibtreu, A.; Denis, E.; Etienne, C.; Rouanes, N.; Mahieu, R.; et al. French National Cohort of First Use of Dalbavancin: A High Proportion of off-Label Use. Int. J. Antimicrob. Agents 2019, 54, 668–672. [Google Scholar] [CrossRef] [PubMed]
  109. Wunsch, S.; Krause, R.; Valentin, T.; Prattes, J.; Janata, O.; Lenger, A.; Bellmann-Weiler, R.; Weiss, G.; Zollner-Schwetz, I. Multicenter Clinical Experience of Real Life Dalbavancin Use in Gram-Positive Infections. Int. J. Infect. Dis. IJID Off. Publ. Int. Soc. Infect. Dis. 2019, 81, 210–214. [Google Scholar] [CrossRef] [PubMed]
  110. Núñez-Núñez, M.; Casas-Hidalgo, I.; García-Fumero, R.; Vallejo-Rodríguez, I.; Anguita-Santos, F.; Hernández-Quero, J.; Cabeza-Barrera, J.; Ruiz-Sancho, A. Dalbavancin Is a Novel Antimicrobial against Gram-Positive Pathogens: Clinical Experience beyond Labelled Indications. Eur. J. Hosp. Pharm. Sci. Pract. 2020, 27, 310–312. [Google Scholar] [CrossRef] [PubMed]
  111. Durante-Mangoni, E.; Boccia, F.; Ursi, M.P.; Karruli, A.; Andini, R.; Galdo, M.; Zampino, R. Dalbavancin for Infective Endocarditis: A Single Centre Experience. J. Chemother. Florence Italy 2021, 33, 256–262. [Google Scholar] [CrossRef]
  112. Arrieta-Loitegui, M.; Caro-Teller, J.M.; Ortiz-Pérez, S.; López-Medrano, F.; San Juan-Garrido, R.; Ferrari-Piquero, J.M. Effectiveness, Safety and Cost Analysis of Dalbavancin in Clinical Practice. Eur. J. Hosp. Pharm. Sci. Pract. 2022, 29, 55–58. [Google Scholar] [CrossRef]
  113. Guleri, A.; More, R.; Sharma, R.; Wong, M.; Abdelrahman, A. Use of Dalbavancin in Infective Endocarditis: A Case Series. JAC-Antimicrob. Resist. 2021, 3, dlab099. [Google Scholar] [CrossRef]
  114. Taylor, K.; Williamson, J.; Luther, V.; Stone, T.; Johnson, J.; Gruss, Z.; Russ-Friedman, C.; Ohl, C.; Beardsley, J. Evaluating the Use of Dalbavancin for Off-Label Indications. Infect. Dis. Rep. 2022, 14, 266–272. [Google Scholar] [CrossRef]
  115. Bryson-Cahn, C.; Beieler, A.M.; Chan, J.D.; Harrington, R.D.; Dhanireddy, S. Dalbavancin as Secondary Therapy for Serious Staphylococcus aureus Infections in a Vulnerable Patient Population. Open Forum Infect. Dis. 2019, 6, ofz028. [Google Scholar] [CrossRef] [PubMed]
  116. Bork, J.T.; Heil, E.L.; Berry, S.; Lopes, E.; Davé, R.; Gilliam, B.L.; Amoroso, A. Dalbavancin Use in Vulnerable Patients Receiving Outpatient Parenteral Antibiotic Therapy for Invasive Gram-Positive Infections. Infect. Dis. Ther. 2019, 8, 171–184. [Google Scholar] [CrossRef]
  117. Ajaka, L.; Heil, E.; Schmalzle, S. Dalbavancin in the Treatment of Bacteremia and Endocarditis in People with Barriers to Standard Care. Antibiotics 2020, 9, 700. [Google Scholar] [CrossRef]
  118. Vazquez Deida, A.A.; Shihadeh, K.C.; Preslaski, C.R.; Young, H.L.; Wyles, D.L.; Jenkins, T.C. Use of a Standardized Dalbavancin Approach to Facilitate Earlier Hospital Discharge for Vulnerable Patients Receiving Prolonged Inpatient Antibiotic Therapy. Open Forum Infect. Dis. 2020, 7, ofaa293. [Google Scholar] [CrossRef]
  119. Veve, M.P.; Patel, N.; Smith, Z.A.; Yeager, S.D.; Wright, L.R.; Shorman, M.A. Comparison of Dalbavancin to Standard-of-Care for Outpatient Treatment of Invasive Gram-Positive Infections. Int. J. Antimicrob. Agents 2020, 56, 106210. [Google Scholar] [CrossRef]
  120. Lueking, R.; Wei, W.; Mang, N.S.; Ortwine, J.K.; Meisner, J. Evaluation of Dalbavancin Use on Clinical Outcomes, Cost-Savings, and Adherence at a Large Safety Net Hospital. Microbiol. Spectr. 2023, 11, e0238522. [Google Scholar] [CrossRef]
  121. Steele, J.M.; Seabury, R.W.; Hale, C.M.; Mogle, B.T. Unsuccessful Treatment of Methicillin-Resistant Staphylococcus aureus Endocarditis with Dalbavancin. J. Clin. Pharm. Ther. 2018, 43, 101–103. [Google Scholar] [CrossRef]
  122. Howard-Anderson, J.; Pouch, S.M.; Sexton, M.E.; Mehta, A.K.; Smith, A.L.; Lyon, G.M.; Friedman-Moraco, R. Left Ventricular Assist Device Infections and the Potential Role for Dalbavancin: A Case Report. Open Forum Infect. Dis. 2019, 6, ofz235. [Google Scholar] [CrossRef]
  123. Hakim, A.; Braun, H.; Thornton, D.; Strymish, J. Successful Treatment of Methicillin-Sensitive Staphylococcus aureus Tricuspid-Valve Endocarditis with Dalbavancin as an Outpatient in a Person Who Injects Drugs: A Case Report. Int. J. Infect. Dis. IJID Off. Publ. Int. Soc. Infect. Dis. 2020, 91, 202–205. [Google Scholar] [CrossRef] [PubMed]
  124. Teigell-Muñoz, F.J.; Mateos-González, M.; Bernal Hertfelder, E.; Sánchez de Torre, A.; García-Ferrón, M.; de Cáceres Velasco, C.; Bueno Muiño, C. A Dalbavancin for Successful Treatment of Infective Endocarditis Caused by Enterococcus Faecalis. Eur. J. Case Rep. Intern. Med. 2023, 10, 003654. [Google Scholar] [CrossRef]
  125. Kussmann, M.; Karer, M.; Obermueller, M.; Schmidt, K.; Barousch, W.; Moser, D.; Nehr, M.; Ramharter, M.; Poeppl, W.; Makristathis, A.; et al. Emergence of a Dalbavancin Induced Glycopeptide/Lipoglycopeptide Non-Susceptible Staphylococcus aureus during Treatment of a Cardiac Device-Related Endocarditis. Emerg. Microbes Infect. 2018, 7, 202. [Google Scholar] [CrossRef]
  126. Spaziante, M.; Franchi, C.; Taliani, G.; D’Avolio, A.; Pietropaolo, V.; Biliotti, E.; Esvan, R.; Venditti, M. Serum Bactericidal Activity Levels Monitor to Guide Intravenous Dalbavancin Chronic Suppressive Therapy of Inoperable Staphylococcal Prosthetic Valve Endocarditis: A Case Report. Open Forum Infect. Dis. 2019, 6, ofz427. [Google Scholar] [CrossRef]
  127. Hitzenbichler, F.; Mohr, A.; Camboni, D.; Simon, M.; Salzberger, B.; Hanses, F. Dalbavancin as Long-Term Suppressive Therapy for Patients with Gram-Positive Bacteremia Due to an Intravascular Source-a Series of Four Cases. Infection 2021, 49, 181–186. [Google Scholar] [CrossRef]
  128. Bouza, E.; Valerio, M.; Soriano, A.; Morata, L.; Carus, E.G.; Rodríguez-González, C.; Hidalgo-Tenorio, M.C.; Plata, A.; Muñoz, P.; Vena, A.; et al. Dalbavancin in the Treatment of Different Gram-Positive Infections: A Real-Life Experience. Int. J. Antimicrob. Agents 2018, 51, 571–577. [Google Scholar] [CrossRef]
  129. Cooper, M.M.; Preslaski, C.R.; Shihadeh, K.C.; Hawkins, K.L.; Jenkins, T.C. Multiple-Dose Dalbavancin Regimens as the Predominant Treatment of Deep-Seated or Endovascular Infections: A Scoping Review. Open Forum Infect. Dis. 2021, 8, ofab486. [Google Scholar] [CrossRef]
  130. Thomas, G.; Henao-Martínez, A.F.; Franco-Paredes, C.; Chastain, D.B. Treatment of Osteoarticular, Cardiovascular, Intravascular-Catheter-Related and Other Complicated Infections with Dalbavancin and Oritavancin: A Systematic Review. Int. J. Antimicrob. Agents 2020, 56, 106069. [Google Scholar] [CrossRef]
  131. Bouza, E.; Burillo, A. Oritavancin: A Novel Lipoglycopeptide Active against Gram-Positive Pathogens Including Multiresistant Strains. Int. J. Antimicrob. Agents 2010, 36, 401–407. [Google Scholar] [CrossRef] [PubMed]
  132. Zhanel, G.G.; Calic, D.; Schweizer, F.; Zelenitsky, S.; Adam, H.; Lagacé-Wiens, P.R.S.; Rubinstein, E.; Gin, A.S.; Hoban, D.J.; Karlowsky, J.A. New Lipoglycopeptides: A Comparative Review of Dalbavancin, Oritavancin and Telavancin. Drugs 2010, 70, 859–886. [Google Scholar] [CrossRef]
  133. Yan, Q.; Karau, M.J.; Patel, R. In Vitro Activity of Oritavancin against Planktonic and Biofilm States of Vancomycin-Susceptible and Vancomycin-Resistant Enterococci. Diagn. Microbiol. Infect. Dis. 2018, 91, 348–350. [Google Scholar] [CrossRef] [PubMed]
  134. Garcia, L.G.; Lemaire, S.; Kahl, B.C.; Becker, K.; Proctor, R.A.; Denis, O.; Tulkens, P.M.; Van Bambeke, F. Pharmacodynamic Evaluation of the Activity of Antibiotics against Hemin- and Menadione-Dependent Small-Colony Variants of Staphylococcus aureus in Models of Extracellular (Broth) and Intracellular (THP-1 Monocytes) Infections. Antimicrob. Agents Chemother. 2012, 56, 3700–3711. [Google Scholar] [CrossRef]
  135. Wu, T.; Meyer, K.; Harrington, A.T.; Danziger, L.H.; Wenzler, E. In Vitro Activity of Oritavancin Alone or in Combination against Vancomycin-Susceptible and -Resistant Enterococci. J. Antimicrob. Chemother. 2019, 74, 1300–1305. [Google Scholar] [CrossRef] [PubMed]
  136. Lagatolla, C.; Mehat, J.W.; La Ragione, R.M.; Luzzati, R.; Di Bella, S. In Vitro and In Vivo Studies of Oritavancin and Fosfomycin Synergism against Vancomycin-Resistant Enterococcus Faecium. Antibiotics 2022, 11, 1334. [Google Scholar] [CrossRef]
  137. Kaatz, G.W.; Seo, S.M.; Aeschlimann, J.R.; Houlihan, H.H.; Mercier, R.C.; Rybak, M.J. Efficacy of LY333328 against Experimental Methicillin-Resistant Staphylococcus aureus Endocarditis. Antimicrob. Agents Chemother. 1998, 42, 981–983. [Google Scholar] [CrossRef]
  138. Saleh-Mghir, A.; Lefort, A.; Petegnief, Y.; Dautrey, S.; Vallois, J.M.; Le Guludec, D.; Carbon, C.; Fantin, B. Activity and Diffusion of LY333328 in Experimental Endocarditis Due to Vancomycin-Resistant Enterococcus Faecalis. Antimicrob. Agents Chemother. 1999, 43, 115–120. [Google Scholar] [CrossRef]
  139. Lefort, A.; Saleh-Mghir, A.; Garry, L.; Carbon, C.; Fantin, B. Activity of LY333328 Combined with Gentamicin in Vitro and in Rabbit Experimental Endocarditis Due to Vancomycin-Susceptible or -Resistant Enterococcus Faecalis. Antimicrob. Agents Chemother. 2000, 44, 3017–3021. [Google Scholar] [CrossRef]
  140. EMA Tenkasi (Previously Orbactiv). Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/tenkasi-previously-orbactiv (accessed on 11 September 2023).
  141. Lupia, T.; De Benedetto, I.; Bosio, R.; Shbaklo, N.; De Rosa, F.G.; Corcione, S. Role of Oritavancin in the Treatment of Infective Endocarditis, Catheter- or Device-Related Infections, Bloodstream Infections, and Bone and Prosthetic Joint Infections in Humans: Narrative Review and Possible Developments. Life 2023, 13, 959. [Google Scholar] [CrossRef]
  142. Bloem, A.; Bax, H.I.; Yusuf, E.; Verkaik, N.J. New-Generation Antibiotics for Treatment of Gram-Positive Infections: A Review with Focus on Endocarditis and Osteomyelitis. J. Clin. Med. 2021, 10, 1743. [Google Scholar] [CrossRef]
  143. Ahiskali, A.; Rhodes, H. Oritavancin for the Treatment of Complicated Gram-Positive Infection in Persons Who Inject Drugs. BMC Pharmacol. Toxicol. 2020, 21, 73. [Google Scholar] [CrossRef]
  144. Johnson, J.A.; Feeney, E.R.; Kubiak, D.W.; Corey, G.R. Prolonged Use of Oritavancin for Vancomycin-Resistant Enterococcus Faecium Prosthetic Valve Endocarditis. Open Forum Infect. Dis. 2015, 2, ofv156. [Google Scholar] [CrossRef] [PubMed]
  145. Stewart, C.L.; Turner, M.S.; Frens, J.J.; Snider, C.B.; Smith, J.R. Real-World Experience with Oritavancin Therapy in Invasive Gram-Positive Infections. Infect. Dis. Ther. 2017, 6, 277–289. [Google Scholar] [CrossRef]
  146. Salcedo, D.A.T.; El-Herte, R.; Granada, M. Oritavancin for the Treatment of Infective Endocarditis Due to Gram-Positive Organism. Ann. Case Rep. 2018. [Google Scholar] [CrossRef]
  147. Falagas, M.E.; Vouloumanou, E.K.; Samonis, G.; Vardakas, K.Z. Fosfomycin. Clin. Microbiol. Rev. 2016, 29, 321–347. [Google Scholar] [CrossRef] [PubMed]
  148. Veganzones, J.; Montero, A.; Maseda, E. New Evidence on the Use of Fosfomycin for Bacteremia and Infectious Endocarditis. Rev. Esp. Quimioter. 2019, 32 (Suppl. S1), 25–29. [Google Scholar] [PubMed]
  149. Sojo-Dorado, J.; López-Hernández, I.; Rosso-Fernandez, C.; Morales, I.M.; Palacios-Baena, Z.R.; Hernández-Torres, A.; Merino de Lucas, E.; Escolà-Vergé, L.; Bereciartua, E.; García-Vázquez, E.; et al. Effectiveness of Fosfomycin for the Treatment of Multidrug-Resistant Escherichia Coli Bacteremic Urinary Tract Infections: A Randomized Clinical Trial. JAMA Netw. Open 2022, 5, e2137277. [Google Scholar] [CrossRef] [PubMed]
  150. Oliva, A.; Volpicelli, L.; Di Bari, S.; Curtolo, A.; Borrazzo, C.; Cogliati Dezza, F.; Cona, A.; Agrenzano, S.; Mularoni, A.; Trancassini, M.; et al. Effect of Ceftazidime/Avibactam plus Fosfomycin Combination on 30 Day Mortality in Patients with Bloodstream Infections Caused by KPC-Producing Klebsiella Pneumoniae: Results from a Multicentre Retrospective Study. JAC-Antimicrob. Resist. 2022, 4, dlac121. [Google Scholar] [CrossRef] [PubMed]
  151. Oliva, A.; Al Ismail, D.; Arcari, G.; Miele, M.C.; Casali, E.; Sacco, F.; Volpicelli, L.; De Angelis, M.; Mascellino, M.T.; Cancelli, F.; et al. Ceftazidime/Avibactam-Resistant Meropenem-Susceptible KPC-Producing Klebsiella Pneumoniae: Analysis of Cases and Evaluation of in Vitro Activity of Fosfomycin-Containing Combinations. J. Glob. Antimicrob. Resist. 2023, 33, 321–327. [Google Scholar] [CrossRef]
  152. Tseng, T.-C.; Chuang, Y.-C.; Yang, J.-L.; Lin, C.-Y.; Huang, S.-H.; Wang, J.-T.; Chen, Y.-C.; Chang, S.-C. The Combination of Daptomycin with Fosfomycin Is More Effective than Daptomycin Alone in Reducing Mortality of Vancomycin-Resistant Enterococcal Bloodstream Infections: A Retrospective, Comparative Cohort Study. Infect. Dis. Ther. 2023, 12, 589–606. [Google Scholar] [CrossRef]
  153. Antonello, R.M.; Principe, L.; Maraolo, A.E.; Viaggi, V.; Pol, R.; Fabbiani, M.; Montagnani, F.; Lovecchio, A.; Luzzati, R.; Di Bella, S. Fosfomycin as Partner Drug for Systemic Infection Management. A Systematic Review of Its Synergistic Properties from In Vitro and In Vivo Studies. Antibiotics 2020, 9, 500. [Google Scholar] [CrossRef]
  154. Antonello, R.M.; Canetti, D.; Riccardi, N. Daptomycin Synergistic Properties from In Vitro and In Vivo Studies: A Systematic Review. J. Antimicrob. Chemother. 2022, 78, 52–77. [Google Scholar] [CrossRef]
  155. García-de-la-Mària, C.; Gasch, O.; García-Gonzalez, J.; Soy, D.; Shaw, E.; Ambrosioni, J.; Almela, M.; Pericàs, J.M.; Tellez, A.; Falces, C.; et al. The Combination of Daptomycin and Fosfomycin Has Synergistic, Potent, and Rapid Bactericidal Activity against Methicillin-Resistant Staphylococcus aureus in a Rabbit Model of Experimental Endocarditis. Antimicrob. Agents Chemother. 2018, 62, e02633-17. [Google Scholar] [CrossRef] [PubMed]
  156. García-de-la-Mària, C.; Gasch, O.; Castañeda, X.; García-González, J.; Soy, D.; Cañas, M.-A.; Ambrosioni, J.; Almela, M.; Pericàs, J.M.; Téllez, A.; et al. Cloxacillin or Fosfomycin plus Daptomycin Combinations Are More Active than Cloxacillin Monotherapy or Combined with Gentamicin against MSSA in a Rabbit Model of Experimental Endocarditis. J. Antimicrob. Chemother. 2020, 75, 3586–3592. [Google Scholar] [CrossRef] [PubMed]
  157. Debbia, E.; Pesce, A.; Schito, G.C. In Vitro Activity of LY146032 Alone and in Combination with Other Antibiotics against Gram-Positive Bacteria. Antimicrob. Agents Chemother. 1988, 32, 279–281. [Google Scholar] [CrossRef] [PubMed]
  158. Berti, A.D.; Sakoulas, G.; Nizet, V.; Tewhey, R.; Rose, W.E. β-Lactam Antibiotics Targeting PBP1 Selectively Enhance Daptomycin Activity against Methicillin-Resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2013, 57, 5005–5012. [Google Scholar] [CrossRef]
  159. Berti, A.D.; Theisen, E.; Sauer, J.-D.; Nonejuie, P.; Olson, J.; Pogliano, J.; Sakoulas, G.; Nizet, V.; Proctor, R.A.; Rose, W.E. Penicillin Binding Protein 1 Is Important in the Compensatory Response of Staphylococcus aureus to Daptomycin-Induced Membrane Damage and Is a Potential Target for β-Lactam-Daptomycin Synergy. Antimicrob. Agents Chemother. 2016, 60, 451–458. [Google Scholar] [CrossRef]
  160. Gasch, O.; Pillai, S.K.; Dakos, J.; Miyakis, S.; Moellering, R.C.; Eliopoulos, G.M. Daptomycin In Vitro Activity against Methicillin-Resistant Staphylococcus aureus Is Enhanced by d-Cycloserine in a Mechanism Associated with a Decrease in Cell Surface Charge. Antimicrob. Agents Chemother. 2013, 57, 4537–4539. [Google Scholar] [CrossRef]
  161. Mishra, N.N.; Lew, C.; Abdelhady, W.; Lapitan, C.K.; Proctor, R.A.; Rose, W.E.; Bayer, A.S. Synergy Mechanisms of Daptomycin-Fosfomycin Combinations in Daptomycin-Susceptible and -Resistant Methicillin-Resistant Staphylococcus aureus: In Vitro, Ex Vivo, and In Vivo Metrics. Antimicrob. Agents Chemother. 2022, 66, e0164921. [Google Scholar] [CrossRef]
  162. Courcol, R.J.; Martin, G.R. In-Vitro Activity of the Combination of Ceftriaxone and Fosfomycin against Staphylococci. J. Antimicrob. Chemother. 1987, 19, 276–278. [Google Scholar] [CrossRef]
  163. Duez, J.M.; Kohli, E.; Pechinot, A.; Tremeaux, J.C.; Kazmierczak, A. Combination between fosfomycin and oxacillin or cefotaxime against methicillin-resistant Staphylococci and Enterococci. Pathol. Biol. 1983, 31, 515–518. [Google Scholar]
  164. Utsui, Y.; Ohya, S.; Magaribuchi, T.; Tajima, M.; Yokota, T. Antibacterial Activity of Cefmetazole Alone and in Combination with Fosfomycin against Methicillin- and Cephem-Resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 1986, 30, 917–922. [Google Scholar] [CrossRef] [PubMed]
  165. Grif, K.; Dierich, M.P.; Pfaller, K.; Miglioli, P.A.; Allerberger, F. In Vitro Activity of Fosfomycin in Combination with Various Antistaphylococcal Substances. J. Antimicrob. Chemother. 2001, 48, 209–217. [Google Scholar] [CrossRef] [PubMed]
  166. Del Río, A.; García-de-la-Mària, C.; Entenza, J.M.; Gasch, O.; Armero, Y.; Soy, D.; Mestres, C.A.; Pericás, J.M.; Falces, C.; Ninot, S.; et al. Fosfomycin plus β-Lactams as Synergistic Bactericidal Combinations for Experimental Endocarditis Due to Methicillin-Resistant and Glycopeptide-Intermediate Staphylococcus aureus. Antimicrob. Agents Chemother. 2016, 60, 478–486. [Google Scholar] [CrossRef] [PubMed]
  167. Guggenbichler, J.P.; Berchtold, D.; Allerberger, F.; Bonatti, H.; Hager, J.; Pfaller, W.; Dierich, M.P. In Vitro and in Vivo Effect of Antibiotics on Catheters Colonized by Staphylococci. Eur. J. Clin. Microbiol. Infect. Dis. Off. Publ. Eur. Soc. Clin. Microbiol. 1992, 11, 408–415. [Google Scholar] [CrossRef] [PubMed]
  168. Tang, H.-J.; Chen, C.-C.; Zhang, C.-C.; Su, B.-A.; Li, C.-M.; Weng, T.-C.; Chiang, S.-R.; Ko, W.-C.; Chuang, Y.-C. In Vitro Efficacy of Fosfomycin-Based Combinations against Clinical Vancomycin-Resistant Enterococcus Isolates. Diagn. Microbiol. Infect. Dis. 2013, 77, 254–257. [Google Scholar] [CrossRef] [PubMed]
  169. Farina, C.; Russello, G.; Chinello, P.; Pasticci, M.B.; Raglio, A.; Ravasio, V.; Rizzi, M.; Scarparo, C.; Vailati, F.; Suter, F.; et al. In Vitro Activity Effects of Twelve Antibiotics Alone and in Association against Twenty-Seven Enterococcus Faecalis Strains Isolated from Italian Patients with Infective Endocarditis: High in Vitro Synergistic Effect of the Association Ceftriaxone-Fosfomycin. Chemotherapy 2011, 57, 426–433. [Google Scholar] [CrossRef] [PubMed]
  170. Gonzalez Moreno, M.; Trampuz, A.; Di Luca, M. Synergistic Antibiotic Activity against Planktonic and Biofilm-Embedded Streptococcus Agalactiae, Streptococcus Pyogenes and Streptococcus Oralis. J. Antimicrob. Chemother. 2017, 72, 3085–3092. [Google Scholar] [CrossRef]
  171. Vicente, M.V.; Olay, T.; Rodríguez, A. Experimental Endocarditis Caused by Streptococcus Sanguis: Single and Combined Antibiotic Therapy. Antimicrob. Agents Chemother. 1981, 20, 10–14. [Google Scholar] [CrossRef]
  172. Olay, T.; Rodríguez, A.; Oliver, L.E.; Vicente, M.V.; Quecedo, M.C. Interaction of Fosfomycin with Other Antimicrobial Agents: In Vitro and in Vivo Studies. J. Antimicrob. Chemother. 1978, 4, 569–576. [Google Scholar] [CrossRef]
  173. Rice, L.B.; Eliopoulos, C.T.; Yao, J.D.; Eliopoulos, G.M.; Moellering, R.C. In Vivo Activity of the Combination of Daptomycin and Fosfomycin Compared with Daptomycin Alone against a Strain of Enterococcus Faecalis with High-Level Gentamicin Resistance in the Rat Endocarditis Model. Diagn. Microbiol. Infect. Dis. 1992, 15, 173–176. [Google Scholar] [CrossRef]
  174. Garrigós, C.; Murillo, O.; Lora-Tamayo, J.; Verdaguer, R.; Tubau, F.; Cabellos, C.; Cabo, J.; Ariza, J. Fosfomycin-Daptomycin and Other Fosfomycin Combinations as Alternative Therapies in Experimental Foreign-Body Infection by Methicillin-Resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2013, 57, 606–610. [Google Scholar] [CrossRef] [PubMed]
  175. Mihailescu, R.; Furustrand Tafin, U.; Corvec, S.; Oliva, A.; Betrisey, B.; Borens, O.; Trampuz, A. High Activity of Fosfomycin and Rifampin against Methicillin-Resistant Staphylococcus aureus Biofilm in Vitro and in an Experimental Foreign-Body Infection Model. Antimicrob. Agents Chemother. 2014, 58, 2547–2553. [Google Scholar] [CrossRef] [PubMed]
  176. Oliva, A.; Furustrand Tafin, U.; Maiolo, E.M.; Jeddari, S.; Bétrisey, B.; Trampuz, A. Activities of Fosfomycin and Rifampin on Planktonic and Adherent Enterococcus Faecalis Strains in an Experimental Foreign-Body Infection Model. Antimicrob. Agents Chemother. 2014, 58, 1284–1293. [Google Scholar] [CrossRef] [PubMed]
  177. Descourouez, J.L.; Jorgenson, M.R.; Wergin, J.E.; Rose, W.E. Fosfomycin Synergy in Vitro with Amoxicillin, Daptomycin, and Linezolid against Vancomycin-Resistant Enterococcus Faecium from Renal Transplant Patients with Infected Urinary Stents. Antimicrob. Agents Chemother. 2013, 57, 1518–1520. [Google Scholar] [CrossRef] [PubMed]
  178. EMA Fosfomycin-Containing Medicinal Products. Available online: https://www.ema.europa.eu/en/medicines/human/referrals/fosfomycin-containing-medicinal-products (accessed on 28 September 2023).
  179. Aoyagi, S.; Kawara, T.; Mizoguchi, T.; Ando, F.; Yanai, T.; Yamamoto, E.; Suzuki, K. Methicillin-Resistant Staphylococcus aureus Endocarditis Following Patch Closure of a Ventricular Septal Defect: Report of a Case. Surg. Today 1994, 24, 644–647. [Google Scholar] [CrossRef] [PubMed]
  180. Del Río, A.; Gasch, O.; Moreno, A.; Peña, C.; Cuquet, J.; Soy, D.; Mestres, C.A.; Suárez, C.; Pare, J.C.; Tubau, F.; et al. Efficacy and Safety of Fosfomycin plus Imipenem as Rescue Therapy for Complicated Bacteremia and Endocarditis Due to Methicillin-Resistant Staphylococcus aureus: A Multicenter Clinical Trial. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2014, 59, 1105–1112. [Google Scholar] [CrossRef] [PubMed]
  181. Pericàs, J.M.; Moreno, A.; Almela, M.; García-de-la-Mària, C.; Marco, F.; Muñoz, P.; Peña, C.; de Alarcón, A.; Del Río, A.; Eworo, A.; et al. Efficacy and Safety of Fosfomycin plus Imipenem versus Vancomycin for Complicated Bacteraemia and Endocarditis Due to Methicillin-Resistant Staphylococcus aureus: A Randomized Clinical Trial. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2018, 24, 673–676. [Google Scholar] [CrossRef]
  182. Rieg, S.; Ernst, A.; Peyerl-Hoffmann, G.; Joost, I.; Camp, J.; Hellmich, M.; Kern, W.V.; Kaasch, A.J.; Seifert, H. Combination Therapy with Rifampicin or Fosfomycin in Patients with Staphylococcus aureus Bloodstream Infection at High Risk for Complications or Relapse: Results of a Large Prospective Observational Cohort. J. Antimicrob. Chemother. 2020, 75, 2282–2290. [Google Scholar] [CrossRef]
  183. Rieg, S.; Joost, I.; Weiß, V.; Peyerl-Hoffmann, G.; Schneider, C.; Hellmich, M.; Seifert, H.; Kern, W.V.; Kaasch, A. Combination Antimicrobial Therapy in Patients with Staphylococcus aureus Bacteraemia-a Post Hoc Analysis in 964 Prospectively Evaluated Patients. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2017, 23, 406.e1–406.e8. [Google Scholar] [CrossRef]
  184. Chen, L.-Y.; Huang, C.-H.; Kuo, S.-C.; Hsiao, C.-Y.; Lin, M.-L.; Wang, F.-D.; Fung, C.-P. High-Dose Daptomycin and Fosfomycin Treatment of a Patient with Endocarditis Caused by Daptomycin-Nonsusceptible Staphylococcus aureus: Case Report. BMC Infect. Dis. 2011, 11, 152. [Google Scholar] [CrossRef]
  185. Miró, J.M.; Entenza, J.M.; Del Río, A.; Velasco, M.; Castañeda, X.; Garcia de la Mària, C.; Giddey, M.; Armero, Y.; Pericàs, J.M.; Cervera, C.; et al. High-Dose Daptomycin plus Fosfomycin Is Safe and Effective in Treating Methicillin-Susceptible and Methicillin-Resistant Staphylococcus aureus Endocarditis. Antimicrob. Agents Chemother. 2012, 56, 4511–4515. [Google Scholar] [CrossRef]
  186. Vergara-López, S.; Domínguez, M.C.; Conejo, M.C.; Pascual, Á.; Rodríguez-Baño, J. Prolonged Treatment with Large Doses of Fosfomycin plus Vancomycin and Amikacin in a Case of Bacteraemia Due to Methicillin-Resistant Staphylococcus Epidermidis and IMP-8 Metallo-β-Lactamase-Producing Klebsiella Oxytoca. J. Antimicrob. Chemother. 2015, 70, 313–315. [Google Scholar] [CrossRef] [PubMed]
  187. Brown, E.; Gould, F.K. Oral Antibiotics for Infective Endocarditis: A Clinical Review. J. Antimicrob. Chemother. 2020, 75, 2021–2027. [Google Scholar] [CrossRef]
  188. Stamboulian, D.; Bonvehi, P.; Arevalo, C.; Bologna, R.; Cassetti, I.; Scilingo, V.; Efron, E. Antibiotic Management of Outpatients with Endocarditis Due to Penicillin-Susceptible Streptococci. Rev. Infect. Dis. 1991, 13 (Suppl. S2), S160–S163. [Google Scholar] [CrossRef] [PubMed]
  189. Heldman, A.W.; Hartert, T.V.; Ray, S.C.; Daoud, E.G.; Kowalski, T.E.; Pompili, V.J.; Sisson, S.D.; Tidmore, W.C.; vom Eigen, K.A.; Goodman, S.N.; et al. Oral Antibiotic Treatment of Right-Sided Staphylococcal Endocarditis in Injection Drug Users: Prospective Randomized Comparison with Parenteral Therapy. Am. J. Med. 1996, 101, 68–76. [Google Scholar] [CrossRef] [PubMed]
  190. Demonchy, E.; Dellamonica, P.; Roger, P.M.; Bernard, E.; Cua, E.; Pulcini, C. Audit of Antibiotic Therapy Used in 66 Cases of Endocarditis. Med. Mal. Infect. 2011, 41, 602–607. [Google Scholar] [CrossRef] [PubMed]
  191. Tissot-Dupont, H.; Gouriet, F.; Oliver, L.; Jamme, M.; Casalta, J.-P.; Jimeno, M.-T.; Arregle, F.; Lavoute, C.; Hubert, S.; Philip, M.; et al. High-Dose Trimethoprim-Sulfamethoxazole and Clindamycin for Staphylococcus aureus Endocarditis. Int. J. Antimicrob. Agents 2019, 54, 143–148. [Google Scholar] [CrossRef] [PubMed]
  192. Iversen, K.; Ihlemann, N.; Gill, S.U.; Madsen, T.; Elming, H.; Jensen, K.T.; Bruun, N.E.; Høfsten, D.E.; Fursted, K.; Christensen, J.J.; et al. Partial Oral versus Intravenous Antibiotic Treatment of Endocarditis. N. Engl. J. Med. 2019, 380, 415–424. [Google Scholar] [CrossRef]
  193. Pries-Heje, M.M.; Wiingaard, C.; Ihlemann, N.; Gill, S.U.; Bruun, N.E.; Elming, H.; Povlsen, J.A.; Madsen, T.; Jensen, K.T.; Fursted, K.; et al. Five-Year Outcomes of the Partial Oral Treatment of Endocarditis (POET) Trial. N. Engl. J. Med. 2022, 386, 601–602. [Google Scholar] [CrossRef]
  194. Attanasio, V.; Di Luca, M.; Carozza, A.; Severino, S.; Pallotto, C.; Capoluongo, N.; Palmiero, G.; Bernardo, M.; Tascini, C. Clinical Efficacy of Amoxicillin/Clavulanate plus Cefditoren as de-Escalation Combination Therapy for Endocarditis Due to Strongly Biofilm-Forming Enterococcus Faecalis. Infect. Dis. Lond. Engl. 2020, 52, 376–379. [Google Scholar] [CrossRef]
  195. Colli, A.; Campodonico, R.; Gherli, T. Early Switch from Vancomycin to Oral Linezolid for Treatment of Gram-Positive Heart Valve Endocarditis. Ann. Thorac. Surg. 2007, 84, 87–91. [Google Scholar] [CrossRef] [PubMed]
  196. Lemaignen, A.; Bernard, L.; Tattevin, P.; Bru, J.-P.; Duval, X.; Hoen, B.; Brunet-Houdard, S.; Mainardi, J.-L.; Caille, A. Oral Switch versus Standard Intravenous Antibiotic Therapy in Left-Sided Endocarditis Due to Susceptible Staphylococci, Streptococci or Enterococci (RODEO): A Protocol for Two Open-Label Randomised Controlled Trials. BMJ Open 2020, 10, e033540. [Google Scholar] [CrossRef] [PubMed]
  197. Chan, L.C.; Basuino, L.; Dip, E.C.; Chambers, H.F. Comparative Efficacies of Tedizolid Phosphate, Vancomycin, and Daptomycin in a Rabbit Model of Methicillin-Resistant Staphylococcus aureus Endocarditis. Antimicrob. Agents Chemother. 2015, 59, 3252–3256. [Google Scholar] [CrossRef] [PubMed]
  198. Singh, K.V.; Arias, C.A.; Murray, B.E. Tedizolid as Step-Down Therapy Following Daptomycin versus Continuation of Daptomycin against Enterococci and Methicillin- and Vancomycin-Resistant Staphylococcus aureus in a Rat Endocarditis Model. Antimicrob. Agents Chemother. 2020, 64, e02303-19. [Google Scholar] [CrossRef]
Figure 1. (A). Summary of the available in vitro, in vivo, and clinical evidence for a possible place in therapy for new antimicrobial strategies for Staphylococcus spp. infective endocarditis. *: Other regimens recommended for the treatment of Staphylococcus spp. IE due to strong and consolidated clinical evidence are not shown in this figure but are discussed in the text; **: clinical evidence derives from randomised clinical trials [10]. (B). Summary of available in vitro, in vivo, and clinical evidence for a possible place in therapy for new antimicrobial strategies for Streptococcus spp. and Enterococcus spp. infective endocarditis. *: Other regimens recommended for the treatment of Streptococcus and E. faecalis spp. IE due to strong and consolidated clinical evidence are not shown in this figure but are discussed in the text. ** As for E. faecalis, the suggested green/yellow colour refers only to clinical evidence for BPR in combination with ampicillin.
Figure 1. (A). Summary of the available in vitro, in vivo, and clinical evidence for a possible place in therapy for new antimicrobial strategies for Staphylococcus spp. infective endocarditis. *: Other regimens recommended for the treatment of Staphylococcus spp. IE due to strong and consolidated clinical evidence are not shown in this figure but are discussed in the text; **: clinical evidence derives from randomised clinical trials [10]. (B). Summary of available in vitro, in vivo, and clinical evidence for a possible place in therapy for new antimicrobial strategies for Streptococcus spp. and Enterococcus spp. infective endocarditis. *: Other regimens recommended for the treatment of Streptococcus and E. faecalis spp. IE due to strong and consolidated clinical evidence are not shown in this figure but are discussed in the text. ** As for E. faecalis, the suggested green/yellow colour refers only to clinical evidence for BPR in combination with ampicillin.
Jcm 12 07693 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Oliva, A.; Cogliati Dezza, F.; Cancelli, F.; Curtolo, A.; Falletta, A.; Volpicelli, L.; Venditti, M. New Antimicrobials and New Therapy Strategies for Endocarditis: Weapons That Should Be Defended. J. Clin. Med. 2023, 12, 7693. https://doi.org/10.3390/jcm12247693

AMA Style

Oliva A, Cogliati Dezza F, Cancelli F, Curtolo A, Falletta A, Volpicelli L, Venditti M. New Antimicrobials and New Therapy Strategies for Endocarditis: Weapons That Should Be Defended. Journal of Clinical Medicine. 2023; 12(24):7693. https://doi.org/10.3390/jcm12247693

Chicago/Turabian Style

Oliva, Alessandra, Francesco Cogliati Dezza, Francesca Cancelli, Ambrogio Curtolo, Antonio Falletta, Lorenzo Volpicelli, and Mario Venditti. 2023. "New Antimicrobials and New Therapy Strategies for Endocarditis: Weapons That Should Be Defended" Journal of Clinical Medicine 12, no. 24: 7693. https://doi.org/10.3390/jcm12247693

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop