Abstract

Introduction

Daptomycin is a bactericidal lipopeptide antibiotic approved for the treatment of systemic infections (i.e. skin and soft tissue infections, bloodstream infections, infective endocarditis) caused by Gram-positive cocci. It is often prescribed in association with a partner drug to increase its bactericidal effect and to prevent the emergence of resistant strains during treatment; however, its synergistic properties are still under evaluation.

Methods

We performed a systematic review to offer clinicians an updated overview of daptomycin synergistic properties from in vitro and in vivo studies. Moreover, we reported all in vitro and in vivo data evaluating daptomycin in combination with other antibiotic agents, subdivided by antibiotic classes, and a summary graph presenting the most favourable combinations at a glance.

Results

A total of 92 studies and 1087 isolates (723 Staphylococcus aureus, 68 Staphylococcus epidermidis, 179 Enterococcus faecium, 105 Enterococcus faecalis, 12 Enterococcus durans) were included. Synergism accounted for 30.9% of total interactions, while indifferent effect was the most frequently observed interaction (41.9%). Antagonistic effect accounted for 0.7% of total interactions. The highest synergistic rates against S. aureus were observed with daptomycin in combination with fosfomycin (55.6%). For S. epidermidis and Enterococcus spp., the most effective combinations were daptomycin plus ceftobiprole (50%) and daptomycin plus fosfomycin (63.6%) or rifampicin (62.8%), respectively.

Future perspectives

We believe this systematic review could be useful for the future updates of guidelines on systemic infections where daptomycin plays a key role.

Introduction

Daptomycin is a semisynthetic cyclic lipopeptide first marketed in the USA in 2003 and in Europe in 2006.1,2 Daptomycin acts with a unique mechanism of action causing depolarization of the bacterial cell wall and membrane reorganization in lipid and protein sites, determining a rapid, concentration-dependent bactericidal effect.3–5 Due to its activity against clinically relevant Gram-positive cocci, including MRSA and VRE,2 daptomycin is a milestone in the treatment of systemic infections, including severe skin and soft tissue infections (SSTIs), bloodstream infections (BSIs), catheter-related BSIs (CRBSIs) and infective endocarditis sustained by Gram-positive cocci.6,7 In fact, besides prescription based on antimicrobial susceptibility testing, it is often prescribed pending microbiological results, if the patient has known rectal or nasal colonization by MDR Gram-positive bacteria.8,9 Daptomycin is generally reported to have a good safety profile and its pharmacokinetics (PK) and pharmacodynamics (PD) allow once-a-day administration, making daptomycin a valuable option for outpatient treatments also.10

Ultimately, there are some methodological and ethical concerns that cannot be ignored in conducting clinical trials, in particular comparative trials between monotherapies and combination regimens, in life-threatening infections such as endocarditis. With this in mind, translational studies may be the most reasonable starting point.

We performed a systematic review to provide an updated overview of daptomycin synergistic properties from in vitro and in vivo studies.

Materials and methods

The preferred reporting items for systematic reviews and meta-analyses (PRISMA) were considered for conception and design of the present systematic review (Table 1).11

Table 1.

PRISMA 2020 checklist11

Title
TitleIdentify the report as a systematic review.YES
Abstract
AbstractSee the PRISMA 2020 for Abstracts checklist.YES
Introduction
RationaleDescribe the rationale for the review in the context of existing knowledge.YES
ObjectivesProvide an explicit statement of the objective(s) or question(s) the review addresses.YES
Materials and methods
Eligibility criteriaSpecify the inclusion and exclusion criteria for the review and how studies were grouped for the syntheses.YES
Information sourcesSpecify all databases, registers, websites, organizations, reference lists and other sources searched or consulted to identify studies. Specify the date when each source was last searched or consulted.YES
Search strategyPresent the full search strategies for all databases, registers and websites, including any filters and limits used.YES
Selection processSpecify the methods used to decide whether a study met the inclusion criteria of the review, including how many reviewers screened each record and each report retrieved, whether they worked independently, and if applicable, details of automation tools used in the process.YES
Data collection processSpecify the methods used to collect data from reports, including how many reviewers collected data from each report, whether they worked independently, any processes for obtaining or confirming data from study investigators, and if applicable, details of automation tools used in the process.YES
Data itemsList and define all outcomes for which data were sought. Specify whether all results that were compatible with each outcome domain in each study were sought (e.g. for all measures, timepoints, analyses), and if not, the methods used to decide which results to collect. List and define all other variables for which data were sought (e.g. participant and intervention characteristics, funding sources). Describe any assumptions made about any missing or unclear information.YES
Study risk of bias assessmentSpecify the methods used to assess risk of bias in the included studies, including details of the tool(s) used, how many reviewers assessed each study and whether they worked independently, and if applicable, details of automation tools used in the process.NA
Effect measuresSpecify for each outcome the effect measure(s) (e.g. risk ratio, mean difference) used in the synthesis or presentation of results.NA
Synthesis methodsDescribe the processes used to decide which studies were eligible for each synthesis (e.g. tabulating the study intervention characteristics and comparing against the planned groups for each synthesis). Describe any methods required to prepare the data for presentation or synthesis, such as handling of missing summary statistics, or data conversions. If meta-analysis was performed, describe the model(s), method(s) to identify the presence and extent of statistical heterogeneity, and software package(s) used. Describe any methods used to explore possible causes of heterogeneity among study results (e.g. subgroup analysis, meta-regression). Describe any sensitivity analyses conducted to assess robustness of the synthesized results.NA
Describe any methods used to tabulate or visually display results of individual studies and syntheses. Describe any methods used to synthesize results and provide a rationale for the choice(s).YES
Reporting bias assessmentDescribe any sensitivity analyses conducted to assess robustness of the synthesized results.NA
Certainty assessmentDescribe any methods used to assess certainty (or confidence) in the body of evidence for an outcome.NA
Results
Study selectionDescribe the results of the search and selection process, from the number of records identified in the search to the number of studies included in the review, ideally using a flow diagram. Cite studies that might appear to meet the inclusion criteria, but which were excluded, and explain why they were excluded.YES
Study characteristicsCite each included study and present its characteristics.YES
Risk of bias in studiesPresent assessments of risk of bias for each included study.YES
Results of individual studiesFor all outcomes, present, for each study: (a) summary statistics for each group (where appropriate) and (b) an effect estimate and its precision (e.g. confidence/credible interval), ideally using structured tables or plots.NA
Results of synthesesFor each synthesis, briefly summarize the characteristics and risk of bias among contributing studies.YES
Present results of all statistical syntheses conducted. If meta-analysis was done, present for each the summary estimate and its precision (e.g. confidence/credible interval) and measures of statistical heterogeneity. If comparing groups, describe the direction of the effect. Present results of all investigations of possible causes of heterogeneity among study results. Present results of all investigations of possible causes of heterogeneity among study results.NA
Reporting biasesPresent assessments of risk of bias due to missing results (arising from reporting biases) for each synthesis assessed.NA
Certainty of evidencePresent assessments of certainty (or confidence) in the body of evidence for each outcome assessed.NA
Discussion
DiscussionProvide a general interpretation of the results in the context of other evidence. Discuss any limitations of the evidence included in the review. Discuss any limitations of the review processes used. Discuss implications of the results for practice, policy, and future research.YES
Other information
Registration and protocolProvide registration information for the review, including register name and registration number, or state that the review was not registered. Indicate where the review protocol can be accessed, or state that a protocol was not prepared. Describe and explain any amendments to information provided at registration or in the protocol.YES
SupportDescribe sources of financial or non-financial support for the review, and the role of the funders or sponsors in the review.YES
Competing interestsDeclare any competing interests of review authors.YES
Availability of data, code and other materialsReport which of the following are publicly available and where they can be found: template data collection forms; data extracted from included studies; data used for all analyses; analytic code; any other materials used in the review.YES
Title
TitleIdentify the report as a systematic review.YES
Abstract
AbstractSee the PRISMA 2020 for Abstracts checklist.YES
Introduction
RationaleDescribe the rationale for the review in the context of existing knowledge.YES
ObjectivesProvide an explicit statement of the objective(s) or question(s) the review addresses.YES
Materials and methods
Eligibility criteriaSpecify the inclusion and exclusion criteria for the review and how studies were grouped for the syntheses.YES
Information sourcesSpecify all databases, registers, websites, organizations, reference lists and other sources searched or consulted to identify studies. Specify the date when each source was last searched or consulted.YES
Search strategyPresent the full search strategies for all databases, registers and websites, including any filters and limits used.YES
Selection processSpecify the methods used to decide whether a study met the inclusion criteria of the review, including how many reviewers screened each record and each report retrieved, whether they worked independently, and if applicable, details of automation tools used in the process.YES
Data collection processSpecify the methods used to collect data from reports, including how many reviewers collected data from each report, whether they worked independently, any processes for obtaining or confirming data from study investigators, and if applicable, details of automation tools used in the process.YES
Data itemsList and define all outcomes for which data were sought. Specify whether all results that were compatible with each outcome domain in each study were sought (e.g. for all measures, timepoints, analyses), and if not, the methods used to decide which results to collect. List and define all other variables for which data were sought (e.g. participant and intervention characteristics, funding sources). Describe any assumptions made about any missing or unclear information.YES
Study risk of bias assessmentSpecify the methods used to assess risk of bias in the included studies, including details of the tool(s) used, how many reviewers assessed each study and whether they worked independently, and if applicable, details of automation tools used in the process.NA
Effect measuresSpecify for each outcome the effect measure(s) (e.g. risk ratio, mean difference) used in the synthesis or presentation of results.NA
Synthesis methodsDescribe the processes used to decide which studies were eligible for each synthesis (e.g. tabulating the study intervention characteristics and comparing against the planned groups for each synthesis). Describe any methods required to prepare the data for presentation or synthesis, such as handling of missing summary statistics, or data conversions. If meta-analysis was performed, describe the model(s), method(s) to identify the presence and extent of statistical heterogeneity, and software package(s) used. Describe any methods used to explore possible causes of heterogeneity among study results (e.g. subgroup analysis, meta-regression). Describe any sensitivity analyses conducted to assess robustness of the synthesized results.NA
Describe any methods used to tabulate or visually display results of individual studies and syntheses. Describe any methods used to synthesize results and provide a rationale for the choice(s).YES
Reporting bias assessmentDescribe any sensitivity analyses conducted to assess robustness of the synthesized results.NA
Certainty assessmentDescribe any methods used to assess certainty (or confidence) in the body of evidence for an outcome.NA
Results
Study selectionDescribe the results of the search and selection process, from the number of records identified in the search to the number of studies included in the review, ideally using a flow diagram. Cite studies that might appear to meet the inclusion criteria, but which were excluded, and explain why they were excluded.YES
Study characteristicsCite each included study and present its characteristics.YES
Risk of bias in studiesPresent assessments of risk of bias for each included study.YES
Results of individual studiesFor all outcomes, present, for each study: (a) summary statistics for each group (where appropriate) and (b) an effect estimate and its precision (e.g. confidence/credible interval), ideally using structured tables or plots.NA
Results of synthesesFor each synthesis, briefly summarize the characteristics and risk of bias among contributing studies.YES
Present results of all statistical syntheses conducted. If meta-analysis was done, present for each the summary estimate and its precision (e.g. confidence/credible interval) and measures of statistical heterogeneity. If comparing groups, describe the direction of the effect. Present results of all investigations of possible causes of heterogeneity among study results. Present results of all investigations of possible causes of heterogeneity among study results.NA
Reporting biasesPresent assessments of risk of bias due to missing results (arising from reporting biases) for each synthesis assessed.NA
Certainty of evidencePresent assessments of certainty (or confidence) in the body of evidence for each outcome assessed.NA
Discussion
DiscussionProvide a general interpretation of the results in the context of other evidence. Discuss any limitations of the evidence included in the review. Discuss any limitations of the review processes used. Discuss implications of the results for practice, policy, and future research.YES
Other information
Registration and protocolProvide registration information for the review, including register name and registration number, or state that the review was not registered. Indicate where the review protocol can be accessed, or state that a protocol was not prepared. Describe and explain any amendments to information provided at registration or in the protocol.YES
SupportDescribe sources of financial or non-financial support for the review, and the role of the funders or sponsors in the review.YES
Competing interestsDeclare any competing interests of review authors.YES
Availability of data, code and other materialsReport which of the following are publicly available and where they can be found: template data collection forms; data extracted from included studies; data used for all analyses; analytic code; any other materials used in the review.YES

NA, not available (not pertinent for the present systematic review).

Table 1.

PRISMA 2020 checklist11

Title
TitleIdentify the report as a systematic review.YES
Abstract
AbstractSee the PRISMA 2020 for Abstracts checklist.YES
Introduction
RationaleDescribe the rationale for the review in the context of existing knowledge.YES
ObjectivesProvide an explicit statement of the objective(s) or question(s) the review addresses.YES
Materials and methods
Eligibility criteriaSpecify the inclusion and exclusion criteria for the review and how studies were grouped for the syntheses.YES
Information sourcesSpecify all databases, registers, websites, organizations, reference lists and other sources searched or consulted to identify studies. Specify the date when each source was last searched or consulted.YES
Search strategyPresent the full search strategies for all databases, registers and websites, including any filters and limits used.YES
Selection processSpecify the methods used to decide whether a study met the inclusion criteria of the review, including how many reviewers screened each record and each report retrieved, whether they worked independently, and if applicable, details of automation tools used in the process.YES
Data collection processSpecify the methods used to collect data from reports, including how many reviewers collected data from each report, whether they worked independently, any processes for obtaining or confirming data from study investigators, and if applicable, details of automation tools used in the process.YES
Data itemsList and define all outcomes for which data were sought. Specify whether all results that were compatible with each outcome domain in each study were sought (e.g. for all measures, timepoints, analyses), and if not, the methods used to decide which results to collect. List and define all other variables for which data were sought (e.g. participant and intervention characteristics, funding sources). Describe any assumptions made about any missing or unclear information.YES
Study risk of bias assessmentSpecify the methods used to assess risk of bias in the included studies, including details of the tool(s) used, how many reviewers assessed each study and whether they worked independently, and if applicable, details of automation tools used in the process.NA
Effect measuresSpecify for each outcome the effect measure(s) (e.g. risk ratio, mean difference) used in the synthesis or presentation of results.NA
Synthesis methodsDescribe the processes used to decide which studies were eligible for each synthesis (e.g. tabulating the study intervention characteristics and comparing against the planned groups for each synthesis). Describe any methods required to prepare the data for presentation or synthesis, such as handling of missing summary statistics, or data conversions. If meta-analysis was performed, describe the model(s), method(s) to identify the presence and extent of statistical heterogeneity, and software package(s) used. Describe any methods used to explore possible causes of heterogeneity among study results (e.g. subgroup analysis, meta-regression). Describe any sensitivity analyses conducted to assess robustness of the synthesized results.NA
Describe any methods used to tabulate or visually display results of individual studies and syntheses. Describe any methods used to synthesize results and provide a rationale for the choice(s).YES
Reporting bias assessmentDescribe any sensitivity analyses conducted to assess robustness of the synthesized results.NA
Certainty assessmentDescribe any methods used to assess certainty (or confidence) in the body of evidence for an outcome.NA
Results
Study selectionDescribe the results of the search and selection process, from the number of records identified in the search to the number of studies included in the review, ideally using a flow diagram. Cite studies that might appear to meet the inclusion criteria, but which were excluded, and explain why they were excluded.YES
Study characteristicsCite each included study and present its characteristics.YES
Risk of bias in studiesPresent assessments of risk of bias for each included study.YES
Results of individual studiesFor all outcomes, present, for each study: (a) summary statistics for each group (where appropriate) and (b) an effect estimate and its precision (e.g. confidence/credible interval), ideally using structured tables or plots.NA
Results of synthesesFor each synthesis, briefly summarize the characteristics and risk of bias among contributing studies.YES
Present results of all statistical syntheses conducted. If meta-analysis was done, present for each the summary estimate and its precision (e.g. confidence/credible interval) and measures of statistical heterogeneity. If comparing groups, describe the direction of the effect. Present results of all investigations of possible causes of heterogeneity among study results. Present results of all investigations of possible causes of heterogeneity among study results.NA
Reporting biasesPresent assessments of risk of bias due to missing results (arising from reporting biases) for each synthesis assessed.NA
Certainty of evidencePresent assessments of certainty (or confidence) in the body of evidence for each outcome assessed.NA
Discussion
DiscussionProvide a general interpretation of the results in the context of other evidence. Discuss any limitations of the evidence included in the review. Discuss any limitations of the review processes used. Discuss implications of the results for practice, policy, and future research.YES
Other information
Registration and protocolProvide registration information for the review, including register name and registration number, or state that the review was not registered. Indicate where the review protocol can be accessed, or state that a protocol was not prepared. Describe and explain any amendments to information provided at registration or in the protocol.YES
SupportDescribe sources of financial or non-financial support for the review, and the role of the funders or sponsors in the review.YES
Competing interestsDeclare any competing interests of review authors.YES
Availability of data, code and other materialsReport which of the following are publicly available and where they can be found: template data collection forms; data extracted from included studies; data used for all analyses; analytic code; any other materials used in the review.YES
Title
TitleIdentify the report as a systematic review.YES
Abstract
AbstractSee the PRISMA 2020 for Abstracts checklist.YES
Introduction
RationaleDescribe the rationale for the review in the context of existing knowledge.YES
ObjectivesProvide an explicit statement of the objective(s) or question(s) the review addresses.YES
Materials and methods
Eligibility criteriaSpecify the inclusion and exclusion criteria for the review and how studies were grouped for the syntheses.YES
Information sourcesSpecify all databases, registers, websites, organizations, reference lists and other sources searched or consulted to identify studies. Specify the date when each source was last searched or consulted.YES
Search strategyPresent the full search strategies for all databases, registers and websites, including any filters and limits used.YES
Selection processSpecify the methods used to decide whether a study met the inclusion criteria of the review, including how many reviewers screened each record and each report retrieved, whether they worked independently, and if applicable, details of automation tools used in the process.YES
Data collection processSpecify the methods used to collect data from reports, including how many reviewers collected data from each report, whether they worked independently, any processes for obtaining or confirming data from study investigators, and if applicable, details of automation tools used in the process.YES
Data itemsList and define all outcomes for which data were sought. Specify whether all results that were compatible with each outcome domain in each study were sought (e.g. for all measures, timepoints, analyses), and if not, the methods used to decide which results to collect. List and define all other variables for which data were sought (e.g. participant and intervention characteristics, funding sources). Describe any assumptions made about any missing or unclear information.YES
Study risk of bias assessmentSpecify the methods used to assess risk of bias in the included studies, including details of the tool(s) used, how many reviewers assessed each study and whether they worked independently, and if applicable, details of automation tools used in the process.NA
Effect measuresSpecify for each outcome the effect measure(s) (e.g. risk ratio, mean difference) used in the synthesis or presentation of results.NA
Synthesis methodsDescribe the processes used to decide which studies were eligible for each synthesis (e.g. tabulating the study intervention characteristics and comparing against the planned groups for each synthesis). Describe any methods required to prepare the data for presentation or synthesis, such as handling of missing summary statistics, or data conversions. If meta-analysis was performed, describe the model(s), method(s) to identify the presence and extent of statistical heterogeneity, and software package(s) used. Describe any methods used to explore possible causes of heterogeneity among study results (e.g. subgroup analysis, meta-regression). Describe any sensitivity analyses conducted to assess robustness of the synthesized results.NA
Describe any methods used to tabulate or visually display results of individual studies and syntheses. Describe any methods used to synthesize results and provide a rationale for the choice(s).YES
Reporting bias assessmentDescribe any sensitivity analyses conducted to assess robustness of the synthesized results.NA
Certainty assessmentDescribe any methods used to assess certainty (or confidence) in the body of evidence for an outcome.NA
Results
Study selectionDescribe the results of the search and selection process, from the number of records identified in the search to the number of studies included in the review, ideally using a flow diagram. Cite studies that might appear to meet the inclusion criteria, but which were excluded, and explain why they were excluded.YES
Study characteristicsCite each included study and present its characteristics.YES
Risk of bias in studiesPresent assessments of risk of bias for each included study.YES
Results of individual studiesFor all outcomes, present, for each study: (a) summary statistics for each group (where appropriate) and (b) an effect estimate and its precision (e.g. confidence/credible interval), ideally using structured tables or plots.NA
Results of synthesesFor each synthesis, briefly summarize the characteristics and risk of bias among contributing studies.YES
Present results of all statistical syntheses conducted. If meta-analysis was done, present for each the summary estimate and its precision (e.g. confidence/credible interval) and measures of statistical heterogeneity. If comparing groups, describe the direction of the effect. Present results of all investigations of possible causes of heterogeneity among study results. Present results of all investigations of possible causes of heterogeneity among study results.NA
Reporting biasesPresent assessments of risk of bias due to missing results (arising from reporting biases) for each synthesis assessed.NA
Certainty of evidencePresent assessments of certainty (or confidence) in the body of evidence for each outcome assessed.NA
Discussion
DiscussionProvide a general interpretation of the results in the context of other evidence. Discuss any limitations of the evidence included in the review. Discuss any limitations of the review processes used. Discuss implications of the results for practice, policy, and future research.YES
Other information
Registration and protocolProvide registration information for the review, including register name and registration number, or state that the review was not registered. Indicate where the review protocol can be accessed, or state that a protocol was not prepared. Describe and explain any amendments to information provided at registration or in the protocol.YES
SupportDescribe sources of financial or non-financial support for the review, and the role of the funders or sponsors in the review.YES
Competing interestsDeclare any competing interests of review authors.YES
Availability of data, code and other materialsReport which of the following are publicly available and where they can be found: template data collection forms; data extracted from included studies; data used for all analyses; analytic code; any other materials used in the review.YES

NA, not available (not pertinent for the present systematic review).

On 1 June 2022 we performed a PubMed/MEDLINE search including papers published starting from 1 January 2003 (year of first authorization of daptomycin for clinical purposes). The PubMed/MEDLINE search engine was chosen as it was a free and accessible option, with optimal update frequency, including online early articles, and the authors were more familiar with it than other search engines as they used it for previous systematic reviews. The complete search string was ‘((Daptomycin[Title/Abstract]) AND (((((((((((((((synergistic[Title/Abstract]) OR (synergic[Title/Abstract])) OR (synergism[Title/Abstract])) OR (antagonism[Title/Abstract])) OR (antagonistic[Title/Abstract])) OR (combination[Title/Abstract])) OR (combined[Title/Abstract])) OR (plus[Title/Abstract])) OR (together[Title/Abstract])) OR (time kill[Title/Abstract])) OR (time-kill[Title/Abstract])) OR (time killing[Title/Abstract])) OR (time-killing[Title/Abstract])) OR (checkerboard[Title/Abstract])) OR (chequerboard[Title/Abstract]))) AND ((‘2003/01/01’[Date—Publication] : ‘2022/05/31’[Date—Publication]))’.

Inclusion criteria were papers written in English, papers evaluating daptomycin in combination with another antibiotic against Gram-positive cocci in vitro or in vivo in animal models (no limitations on the site of infection) and full text available online. Exclusion criteria were papers evaluating daptomycin alone or in combination with molecules that are not antibiotics (i.e. antimicrobial peptides, phytotherapeutics), combinations tested against bacteria other than Gram-positive cocci, papers reporting clinical data (i.e. case reports, case series, clinical trials) or expert opinions, papers written in languages other than English and full text not available online.

A total of 796 published papers were identified and screened (Figure 1). Of these, 19 were excluded by language screening (written in languages other than English) and 481 by title/abstract screening. The remaining 296 papers and their pertinent references were jointly reviewed and discussed by authors. Ninety-two studies were finally included and considered for further analysis. Fifteen papers, though pertinent for the aim of the present review, had no extractable data, i.e. only general statements about synergistic interactions but no details on how the synergism was established. Studies evaluating daptomycin synergistic properties in bacteria other than Gram-positive cocci were not included in the data analysis but are considered in the Discussion section. The full database of articles screened and included in the present systematic review is available from the corresponding author.

Stepwise procedure for study selection.
Figure 1.

Stepwise procedure for study selection.

To ensure homogeneous criteria of paper selection, analysis and data extraction, the following common definitions were established and adopted by all authors (R.M.A., D.C., N.R.), who reviewed the papers independently.

Synergistic effect

Chequerboard assay or Etest: FIC index (FICI) ≤0.5. FICI is defined as follows: A/MICA + B/MICB = FICA + FICB = FICI. Time–kill assay: ratio of effective concentrations concordant with FICI or ≥2 log kill.

Additive effect

Chequerboard assay or Etest: 0.5 < FICI ≤ 1. Time–kill assay: ratio of effective concentrations concordant with FICI or 1 < log kill <2.

Indifferent effect

Chequerboard assay or Etest: 1 < FICI < 4. Time–kill assay: ratio of effective concentrations concordant with FICI or 1 log kill.

Antagonistic effect

Chequerboard assay or Etest: FICI 4. Time–kill assay: ratio of effective concentrations concordant with FICI or <1 log kill.

For in vitro studies using a method different from chequerboard, Etest or time–kill assay (i.e. PK/PD models), synergism was evaluated according to the ratio of effective concentrations in combination or the log kill.

For studies performed in vivo, synergism was established with the same ratio of effective concentrations considered for chequerboard assays or with the same log kill considered for time–kill assays.

Studies in which there was no distinction between additive and indifferent effect were included and the interaction was considered indifferent (we remarked about this point in the note section for these studies).

For further analysis, i.e. descriptive statistics calculating percentages of synergism for daptomycin in combination with each antibiotic class, if a study evaluated daptomycin in combination with more than one antimicrobial agent belonging to the same antibiotic class, the most unfavourable association was considered.

The present systematic review was not registered and no specific protocol was written.

Results

A total of 92 studies and 1087 isolates (723 Staphylococcus aureus, 68 Staphylococcus epidermidis, 179 Enterococcus faecium, 105 Enterococcus faecalis, 12 Enterococcus durans) were included in the present systematic review. For better comprehension, a table with the included studies and a brief summary of the main findings is provided for each antibiotic class.

Penicillins and penicillins/β-lactamase inhibitors (BLIs)

Twenty-two studies evaluated daptomycin in combination with penicillins or penicillins/BLIs (Table 2).12–33

Table 2.

Studies evaluating daptomycin in combination with penicillins and penicillins + BLIs

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2004, USA18MRSA (100)in vitro (ET, TK)OxacillinET: 6 (33.3); TK: 11 (61)ET: 12 (66.6); TK: 7 (39)12
E. faecium2004, USA19VRE (100)in vitro (ET)Ampicillin13 (68.4)1 (5.3)5 (26.3)13
SA2005, USA40MRSA (50)in vitro (ET) (TK on 1 MSSA)AmpicillinET: 4 (10)ET: 35 (87.5); TK: 1 (100)ET: 1 (2.5)No distinction between additive and indifferent effect. TK was performed on the isolate showing antagonistic effect with ET.14
in vitro (ET, 37 isolates tested)OxacillinET: 4 (11)ET: 33 (89)
E. faecalis, E. faecium40VRE (50)in vitro (ET) (TK on 1 VSE)AmpicillinET: 3 (7.5)ET: 37 (92.5); TK: 1 (100)
SA2006, USA2GISA (100)in vitro (ET, TK)Ampicillin/sulbactamET: 2 (100); TK: 2 (100)15
E. faecium2006, Turkey42VRE (100)in vitro (ET)Ampicillin27 (64.2)15 (35.8)No distinction between additive and indifferent effect.16
SA30MRSA (100)Ampicillin/sulbactam28 (93.3)2 (6.7)
Piperacillin/tazobactam22 (73.3)8 (26.7)
Ticarcillin/clavulanate24 (80)6 (20)
SE36MRSE (100)Ampicillin/sulbactam14 (38.8)22 (61.2)
Piperacillin/tazobactam18 (50)18 (50)
Ticarcillin/clavulanate12 (33.3)24 (66.7)
SA2008, Taiwan1VISA (100)in vitro (TK)Oxacillin1 (100)17
SA2010, USA2MRSA (100)in vitro (TK); in vivo (rabbit, catheter-induced IE with kidney and spleen dissemination)OxacillinTK: 2 (100)in vivo: 2 (100)DNS isolates. In the in vivo study, equivalent human dosage of DAP 12 mg/kg was administered.18
SA2010, USA2MRSA (100)in vitro (PK/PD model)Nafcillin2 (100)DNS isolates. High inoculum tested.19
E. faecium2012, USA1VRE (100)in vitro (TK, PD model)Ampicillin1 (100)High inoculum tested.20
SA2012, Spain1MRSA (100)in vitro (TK), in vivo (rat, tissue cage infection model)CloxacillinTK: 1 (100); in vivo: 1 (100)Standard and high inoculum tested.
In the in vivo study, equivalent human dosage of DAP 6 and 10 mg/kg was administered (synergism observed with both dosages).
21
SA2012, USA2MRSA (100)in vitro (TK)Oxacillin, Amoxicillin/clavulanate2 (100)In the in vivo study, equivalent human dosage of DAP 10 mg/kg was administered.22
in vivo (G. mellonella infection model)Nafcillin2 (100)
SA2013, USA20VISA (100)in vitro (TK, PK/PD model)Nafcillin11 (55)9 (45)In the PK/PD model, the superiority of the combination compared with monotherapy was not as marked as in TK analysis.23
SA2014, Spain1MRSA (100)in vitro (TK); in vivo (rat, foreign body infection model)Cloxacillinin vitro: 1 (100); in vivo: 1 (100)Resistance developed with monotherapies, but not with combinations. Standard and high inoculum tested. In the in vivo study, equivalent human dosage of DAP 8–10 mg/kg was administered.24
E. faecalis2015, USA2VRE (100)in vitro (CB, TK)Ampicillin2 (100)25
E. faecium22 (100)
E. faecium2015, USA4VRE (100)in vitro (TK)Ampicillin2 (50)2 (50)DNS isolates.26
E. faecalis22 (100)
E. faecalis2015, USA1VRE (100)in vitro (PK/PD model)Ampicillin1 (100)27
E. faecium22 (100)
SA2016, USA8MRSA (100)in vitro (CB, TK)Piperacillin/tazobactam7 (87.5)1 (12.5)4 DNS isolates.28
Ampicillin/sulbactam7 (87.5)1 (12.5)
E. faecalis2017, Spain8in vitro (TK); in vivo (rabbit, IE, 2 strains)AmpicillinTK: 8 (100); in vivo: 2 (100)When testing high inoculum, 2 isolates showed synergism and 6 isolates showed additive effect. In the in vivo study, equivalent human dosage of DAP 10 mg/kg was administered.31
SA2018, Spain5MRSA (100)in vitro (TK); in vivo (rabbit, IE, 1 isolate)CloxacillinTK: 5 (100)in vivo: 1 (100)Synergism was confirmed also when high inoculum was tested. In the in vivo study, equivalent human dosage of DAP 6 and 10 mg/kg was administered (synergism observed with DAP 6  mg/kg, indifferent effect with DAP 10 mg/kg).29
SA2019, Taiwan100MRSA (100)in vitro (CB)Oxacillin11 (11)2 (2)87 (87)No distinction between indifferent and antagonistic effect.30
SA2020, Spain5in vitro (TK), in vivo (rabbit, IE, 1 isolate)CloxacillinTK: 5 (100); in vivo: 1 (100)Standard and high inoculum tested. In the in vivo study, equivalent human dosage of DAP 6 mg/kg was administered.32
E. faecalis2021, USA2VRE (100)in vitro (TK)Ampicillin2 (100)33
E. faecium21 (50)1 (50)
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2004, USA18MRSA (100)in vitro (ET, TK)OxacillinET: 6 (33.3); TK: 11 (61)ET: 12 (66.6); TK: 7 (39)12
E. faecium2004, USA19VRE (100)in vitro (ET)Ampicillin13 (68.4)1 (5.3)5 (26.3)13
SA2005, USA40MRSA (50)in vitro (ET) (TK on 1 MSSA)AmpicillinET: 4 (10)ET: 35 (87.5); TK: 1 (100)ET: 1 (2.5)No distinction between additive and indifferent effect. TK was performed on the isolate showing antagonistic effect with ET.14
in vitro (ET, 37 isolates tested)OxacillinET: 4 (11)ET: 33 (89)
E. faecalis, E. faecium40VRE (50)in vitro (ET) (TK on 1 VSE)AmpicillinET: 3 (7.5)ET: 37 (92.5); TK: 1 (100)
SA2006, USA2GISA (100)in vitro (ET, TK)Ampicillin/sulbactamET: 2 (100); TK: 2 (100)15
E. faecium2006, Turkey42VRE (100)in vitro (ET)Ampicillin27 (64.2)15 (35.8)No distinction between additive and indifferent effect.16
SA30MRSA (100)Ampicillin/sulbactam28 (93.3)2 (6.7)
Piperacillin/tazobactam22 (73.3)8 (26.7)
Ticarcillin/clavulanate24 (80)6 (20)
SE36MRSE (100)Ampicillin/sulbactam14 (38.8)22 (61.2)
Piperacillin/tazobactam18 (50)18 (50)
Ticarcillin/clavulanate12 (33.3)24 (66.7)
SA2008, Taiwan1VISA (100)in vitro (TK)Oxacillin1 (100)17
SA2010, USA2MRSA (100)in vitro (TK); in vivo (rabbit, catheter-induced IE with kidney and spleen dissemination)OxacillinTK: 2 (100)in vivo: 2 (100)DNS isolates. In the in vivo study, equivalent human dosage of DAP 12 mg/kg was administered.18
SA2010, USA2MRSA (100)in vitro (PK/PD model)Nafcillin2 (100)DNS isolates. High inoculum tested.19
E. faecium2012, USA1VRE (100)in vitro (TK, PD model)Ampicillin1 (100)High inoculum tested.20
SA2012, Spain1MRSA (100)in vitro (TK), in vivo (rat, tissue cage infection model)CloxacillinTK: 1 (100); in vivo: 1 (100)Standard and high inoculum tested.
In the in vivo study, equivalent human dosage of DAP 6 and 10 mg/kg was administered (synergism observed with both dosages).
21
SA2012, USA2MRSA (100)in vitro (TK)Oxacillin, Amoxicillin/clavulanate2 (100)In the in vivo study, equivalent human dosage of DAP 10 mg/kg was administered.22
in vivo (G. mellonella infection model)Nafcillin2 (100)
SA2013, USA20VISA (100)in vitro (TK, PK/PD model)Nafcillin11 (55)9 (45)In the PK/PD model, the superiority of the combination compared with monotherapy was not as marked as in TK analysis.23
SA2014, Spain1MRSA (100)in vitro (TK); in vivo (rat, foreign body infection model)Cloxacillinin vitro: 1 (100); in vivo: 1 (100)Resistance developed with monotherapies, but not with combinations. Standard and high inoculum tested. In the in vivo study, equivalent human dosage of DAP 8–10 mg/kg was administered.24
E. faecalis2015, USA2VRE (100)in vitro (CB, TK)Ampicillin2 (100)25
E. faecium22 (100)
E. faecium2015, USA4VRE (100)in vitro (TK)Ampicillin2 (50)2 (50)DNS isolates.26
E. faecalis22 (100)
E. faecalis2015, USA1VRE (100)in vitro (PK/PD model)Ampicillin1 (100)27
E. faecium22 (100)
SA2016, USA8MRSA (100)in vitro (CB, TK)Piperacillin/tazobactam7 (87.5)1 (12.5)4 DNS isolates.28
Ampicillin/sulbactam7 (87.5)1 (12.5)
E. faecalis2017, Spain8in vitro (TK); in vivo (rabbit, IE, 2 strains)AmpicillinTK: 8 (100); in vivo: 2 (100)When testing high inoculum, 2 isolates showed synergism and 6 isolates showed additive effect. In the in vivo study, equivalent human dosage of DAP 10 mg/kg was administered.31
SA2018, Spain5MRSA (100)in vitro (TK); in vivo (rabbit, IE, 1 isolate)CloxacillinTK: 5 (100)in vivo: 1 (100)Synergism was confirmed also when high inoculum was tested. In the in vivo study, equivalent human dosage of DAP 6 and 10 mg/kg was administered (synergism observed with DAP 6  mg/kg, indifferent effect with DAP 10 mg/kg).29
SA2019, Taiwan100MRSA (100)in vitro (CB)Oxacillin11 (11)2 (2)87 (87)No distinction between indifferent and antagonistic effect.30
SA2020, Spain5in vitro (TK), in vivo (rabbit, IE, 1 isolate)CloxacillinTK: 5 (100); in vivo: 1 (100)Standard and high inoculum tested. In the in vivo study, equivalent human dosage of DAP 6 mg/kg was administered.32
E. faecalis2021, USA2VRE (100)in vitro (TK)Ampicillin2 (100)33
E. faecium21 (50)1 (50)

CB, chequerboard; ET, Etest; IE, infective endocarditis; DAP, daptomycin; DNS, daptomycin non-susceptible; SA, S. aureus; SE, S. epidermidis; TK, time–kill assay.

Table 2.

Studies evaluating daptomycin in combination with penicillins and penicillins + BLIs

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2004, USA18MRSA (100)in vitro (ET, TK)OxacillinET: 6 (33.3); TK: 11 (61)ET: 12 (66.6); TK: 7 (39)12
E. faecium2004, USA19VRE (100)in vitro (ET)Ampicillin13 (68.4)1 (5.3)5 (26.3)13
SA2005, USA40MRSA (50)in vitro (ET) (TK on 1 MSSA)AmpicillinET: 4 (10)ET: 35 (87.5); TK: 1 (100)ET: 1 (2.5)No distinction between additive and indifferent effect. TK was performed on the isolate showing antagonistic effect with ET.14
in vitro (ET, 37 isolates tested)OxacillinET: 4 (11)ET: 33 (89)
E. faecalis, E. faecium40VRE (50)in vitro (ET) (TK on 1 VSE)AmpicillinET: 3 (7.5)ET: 37 (92.5); TK: 1 (100)
SA2006, USA2GISA (100)in vitro (ET, TK)Ampicillin/sulbactamET: 2 (100); TK: 2 (100)15
E. faecium2006, Turkey42VRE (100)in vitro (ET)Ampicillin27 (64.2)15 (35.8)No distinction between additive and indifferent effect.16
SA30MRSA (100)Ampicillin/sulbactam28 (93.3)2 (6.7)
Piperacillin/tazobactam22 (73.3)8 (26.7)
Ticarcillin/clavulanate24 (80)6 (20)
SE36MRSE (100)Ampicillin/sulbactam14 (38.8)22 (61.2)
Piperacillin/tazobactam18 (50)18 (50)
Ticarcillin/clavulanate12 (33.3)24 (66.7)
SA2008, Taiwan1VISA (100)in vitro (TK)Oxacillin1 (100)17
SA2010, USA2MRSA (100)in vitro (TK); in vivo (rabbit, catheter-induced IE with kidney and spleen dissemination)OxacillinTK: 2 (100)in vivo: 2 (100)DNS isolates. In the in vivo study, equivalent human dosage of DAP 12 mg/kg was administered.18
SA2010, USA2MRSA (100)in vitro (PK/PD model)Nafcillin2 (100)DNS isolates. High inoculum tested.19
E. faecium2012, USA1VRE (100)in vitro (TK, PD model)Ampicillin1 (100)High inoculum tested.20
SA2012, Spain1MRSA (100)in vitro (TK), in vivo (rat, tissue cage infection model)CloxacillinTK: 1 (100); in vivo: 1 (100)Standard and high inoculum tested.
In the in vivo study, equivalent human dosage of DAP 6 and 10 mg/kg was administered (synergism observed with both dosages).
21
SA2012, USA2MRSA (100)in vitro (TK)Oxacillin, Amoxicillin/clavulanate2 (100)In the in vivo study, equivalent human dosage of DAP 10 mg/kg was administered.22
in vivo (G. mellonella infection model)Nafcillin2 (100)
SA2013, USA20VISA (100)in vitro (TK, PK/PD model)Nafcillin11 (55)9 (45)In the PK/PD model, the superiority of the combination compared with monotherapy was not as marked as in TK analysis.23
SA2014, Spain1MRSA (100)in vitro (TK); in vivo (rat, foreign body infection model)Cloxacillinin vitro: 1 (100); in vivo: 1 (100)Resistance developed with monotherapies, but not with combinations. Standard and high inoculum tested. In the in vivo study, equivalent human dosage of DAP 8–10 mg/kg was administered.24
E. faecalis2015, USA2VRE (100)in vitro (CB, TK)Ampicillin2 (100)25
E. faecium22 (100)
E. faecium2015, USA4VRE (100)in vitro (TK)Ampicillin2 (50)2 (50)DNS isolates.26
E. faecalis22 (100)
E. faecalis2015, USA1VRE (100)in vitro (PK/PD model)Ampicillin1 (100)27
E. faecium22 (100)
SA2016, USA8MRSA (100)in vitro (CB, TK)Piperacillin/tazobactam7 (87.5)1 (12.5)4 DNS isolates.28
Ampicillin/sulbactam7 (87.5)1 (12.5)
E. faecalis2017, Spain8in vitro (TK); in vivo (rabbit, IE, 2 strains)AmpicillinTK: 8 (100); in vivo: 2 (100)When testing high inoculum, 2 isolates showed synergism and 6 isolates showed additive effect. In the in vivo study, equivalent human dosage of DAP 10 mg/kg was administered.31
SA2018, Spain5MRSA (100)in vitro (TK); in vivo (rabbit, IE, 1 isolate)CloxacillinTK: 5 (100)in vivo: 1 (100)Synergism was confirmed also when high inoculum was tested. In the in vivo study, equivalent human dosage of DAP 6 and 10 mg/kg was administered (synergism observed with DAP 6  mg/kg, indifferent effect with DAP 10 mg/kg).29
SA2019, Taiwan100MRSA (100)in vitro (CB)Oxacillin11 (11)2 (2)87 (87)No distinction between indifferent and antagonistic effect.30
SA2020, Spain5in vitro (TK), in vivo (rabbit, IE, 1 isolate)CloxacillinTK: 5 (100); in vivo: 1 (100)Standard and high inoculum tested. In the in vivo study, equivalent human dosage of DAP 6 mg/kg was administered.32
E. faecalis2021, USA2VRE (100)in vitro (TK)Ampicillin2 (100)33
E. faecium21 (50)1 (50)
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2004, USA18MRSA (100)in vitro (ET, TK)OxacillinET: 6 (33.3); TK: 11 (61)ET: 12 (66.6); TK: 7 (39)12
E. faecium2004, USA19VRE (100)in vitro (ET)Ampicillin13 (68.4)1 (5.3)5 (26.3)13
SA2005, USA40MRSA (50)in vitro (ET) (TK on 1 MSSA)AmpicillinET: 4 (10)ET: 35 (87.5); TK: 1 (100)ET: 1 (2.5)No distinction between additive and indifferent effect. TK was performed on the isolate showing antagonistic effect with ET.14
in vitro (ET, 37 isolates tested)OxacillinET: 4 (11)ET: 33 (89)
E. faecalis, E. faecium40VRE (50)in vitro (ET) (TK on 1 VSE)AmpicillinET: 3 (7.5)ET: 37 (92.5); TK: 1 (100)
SA2006, USA2GISA (100)in vitro (ET, TK)Ampicillin/sulbactamET: 2 (100); TK: 2 (100)15
E. faecium2006, Turkey42VRE (100)in vitro (ET)Ampicillin27 (64.2)15 (35.8)No distinction between additive and indifferent effect.16
SA30MRSA (100)Ampicillin/sulbactam28 (93.3)2 (6.7)
Piperacillin/tazobactam22 (73.3)8 (26.7)
Ticarcillin/clavulanate24 (80)6 (20)
SE36MRSE (100)Ampicillin/sulbactam14 (38.8)22 (61.2)
Piperacillin/tazobactam18 (50)18 (50)
Ticarcillin/clavulanate12 (33.3)24 (66.7)
SA2008, Taiwan1VISA (100)in vitro (TK)Oxacillin1 (100)17
SA2010, USA2MRSA (100)in vitro (TK); in vivo (rabbit, catheter-induced IE with kidney and spleen dissemination)OxacillinTK: 2 (100)in vivo: 2 (100)DNS isolates. In the in vivo study, equivalent human dosage of DAP 12 mg/kg was administered.18
SA2010, USA2MRSA (100)in vitro (PK/PD model)Nafcillin2 (100)DNS isolates. High inoculum tested.19
E. faecium2012, USA1VRE (100)in vitro (TK, PD model)Ampicillin1 (100)High inoculum tested.20
SA2012, Spain1MRSA (100)in vitro (TK), in vivo (rat, tissue cage infection model)CloxacillinTK: 1 (100); in vivo: 1 (100)Standard and high inoculum tested.
In the in vivo study, equivalent human dosage of DAP 6 and 10 mg/kg was administered (synergism observed with both dosages).
21
SA2012, USA2MRSA (100)in vitro (TK)Oxacillin, Amoxicillin/clavulanate2 (100)In the in vivo study, equivalent human dosage of DAP 10 mg/kg was administered.22
in vivo (G. mellonella infection model)Nafcillin2 (100)
SA2013, USA20VISA (100)in vitro (TK, PK/PD model)Nafcillin11 (55)9 (45)In the PK/PD model, the superiority of the combination compared with monotherapy was not as marked as in TK analysis.23
SA2014, Spain1MRSA (100)in vitro (TK); in vivo (rat, foreign body infection model)Cloxacillinin vitro: 1 (100); in vivo: 1 (100)Resistance developed with monotherapies, but not with combinations. Standard and high inoculum tested. In the in vivo study, equivalent human dosage of DAP 8–10 mg/kg was administered.24
E. faecalis2015, USA2VRE (100)in vitro (CB, TK)Ampicillin2 (100)25
E. faecium22 (100)
E. faecium2015, USA4VRE (100)in vitro (TK)Ampicillin2 (50)2 (50)DNS isolates.26
E. faecalis22 (100)
E. faecalis2015, USA1VRE (100)in vitro (PK/PD model)Ampicillin1 (100)27
E. faecium22 (100)
SA2016, USA8MRSA (100)in vitro (CB, TK)Piperacillin/tazobactam7 (87.5)1 (12.5)4 DNS isolates.28
Ampicillin/sulbactam7 (87.5)1 (12.5)
E. faecalis2017, Spain8in vitro (TK); in vivo (rabbit, IE, 2 strains)AmpicillinTK: 8 (100); in vivo: 2 (100)When testing high inoculum, 2 isolates showed synergism and 6 isolates showed additive effect. In the in vivo study, equivalent human dosage of DAP 10 mg/kg was administered.31
SA2018, Spain5MRSA (100)in vitro (TK); in vivo (rabbit, IE, 1 isolate)CloxacillinTK: 5 (100)in vivo: 1 (100)Synergism was confirmed also when high inoculum was tested. In the in vivo study, equivalent human dosage of DAP 6 and 10 mg/kg was administered (synergism observed with DAP 6  mg/kg, indifferent effect with DAP 10 mg/kg).29
SA2019, Taiwan100MRSA (100)in vitro (CB)Oxacillin11 (11)2 (2)87 (87)No distinction between indifferent and antagonistic effect.30
SA2020, Spain5in vitro (TK), in vivo (rabbit, IE, 1 isolate)CloxacillinTK: 5 (100); in vivo: 1 (100)Standard and high inoculum tested. In the in vivo study, equivalent human dosage of DAP 6 mg/kg was administered.32
E. faecalis2021, USA2VRE (100)in vitro (TK)Ampicillin2 (100)33
E. faecium21 (50)1 (50)

CB, chequerboard; ET, Etest; IE, infective endocarditis; DAP, daptomycin; DNS, daptomycin non-susceptible; SA, S. aureus; SE, S. epidermidis; TK, time–kill assay.

S. aureus [MSSA, MRSA, VISA, glycopeptide-intermediate S. aureus (GISA)] and methicillin-resistant S. epidermidis (MRSE)

Two hundred and thirty-seven isolates of S. aureus (25 MSSA, 189 MRSA, 21 VISA, 2 GISA) were tested in vitro. Indifferent effect was observed against most isolates (145; 61.2%). The combination was synergistic against 77 (32.5%) isolates. Antagonistic effect was found only against one isolate of S. aureus when the combination was tested with Etest, but the result was not confirmed by time–kill assay (indifferent effect).14 Eight isolates of S. aureus were tested in vivo; synergistic effect was observed against five (62.5%) isolates, additive effect against two isolates (25%) and indifferent effect against one isolate (12.5%).

Only one study evaluated daptomycin in combination with penicillins/BLIs (ampicillin/sulbactam, piperacillin/tazobactam, ticarcillin/clavulanate) in vitro against S. epidermidis; the most effective combination was daptomycin + piperacillin/tazobactam, showing synergistic effect against 18 (50%) isolates and indifferent effect against 18 (50%) isolates.16

Enterococcus spp. [vancomycin-susceptible Enterococcus (VSE), VRE]

A total of 127 Enterococcus spp. isolates (28 VSE, 99 VRE) were tested: the most frequent interaction was synergism (65 isolates; 51.2%), followed by additive effect (31.5%) and indifferent effect (17.3%). No antagonistic effect was described. Only one study evaluated the combination in vivo against two isolates of E. faecalis in a rabbit infective endocarditis model; the combination was highly effective and acted synergistically against both isolates.31

Cephalosporins and cephalosporins/BLIs

Daptomycin in combination with cephalosporins or cephalosporins/BLIs was evaluated in 26 studies (Table 3).14,19,25–27,33–53

Table 3.

Studies evaluating daptomycin in combination with cephalosporins and cephalosporins + BLIs

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2005, USA2MRSA (100)in vitro (PK/PD model)Cefepime2 (100)34
SA2005, USA40MRSA (50)in vitro (ET) (TK on 1 MRSA)CefepimeET: 2 (5); TK: 1 (100)ET: 38 (95)No distinction between additive and indifferent effect.14
in vitro (ET)CeftriaxoneET: 1 (2.5)ET: 39 (97.5)
E. faecalis, E. faecium40VRE (50)in vitro (ET) (TK on 1 VSE)CefepimeET: 9 (22.5)ET: 31 (77.5); TK: 1 (100)
in vitro (ET) (TK on 2 VSE and 1 VRE)CeftriaxoneET: 18 (45)ET: 22 (55); TK: 3 (100)
SA2010, USA2MRSA (100)in vitro (PD model)Cefepime1 (50)1 (50)DNS isolates. High inoculum tested.19
SA2013, USA2hVISA (50)in vitro (PK/PD model)Ceftaroline2 (100)Two isogenic isolates, one of which mutated into a hVISA DNS strain. High inoculum tested.35
SA2014, USA3MRSA (66.7), hVISA (33.3)in vitro (biofilm model)Ceftaroline, cefazolin3 (100)Synergy was observed against all isolated for both combinations.36
SA2014, USA3VISA (100)in vitro (PK/PD model)Ceftaroline2 (66.7)1 (33.3)DNS isolates. High inoculum tested.37
E. faecium2014, USA2VRE (100)in vitro (PK/PD model)Ceftriaxone1 (50)1 (50)High inoculum tested.38
E. faecalis1Ceftriaxone1 (100)
SA2014, USA2MRSA (50)in vitro (CB, TK)Ceftaroline2 (100)39
SA2014, USA5MRSA (60); VISA (40)in vitro (TK)Ceftobiprole5 (100)40
SA2015, USA1MRSA (100)in vitro (PK/PD model)Ceftaroline1 (100)41
E. faecalis2015, USA2VRE (100)in vitro (CB, TK)Ceftaroline2 (100)25
2Cefepime1 (50)1 (50)
2Ceftriaxone1 (50)1 (50)
2Cefotaxime1 (50)1 (50)
2Cefazolin2 (100)
E. faecium2Ceftaroline2 (100)
2Cefepime1 (50)1 (50)
2Ceftriaxone1 (50)1 (50)
2Cefotaxime1 (50)1 (50)
2Cefazolin1 (50)1 (50)
E. faecium2015, USA4VRE (100)in vitro (TK)Ceftobiprole2 (50)2 (50)DNS isolates.26
E. faecalis2Ceftobiprole2 (100)
E. faecalis2015, USA1VRE (100)in vitro (PK/PD model)Ceftaroline1 (100)27
E. faecium2Ceftaroline2 (100)
Enterococcus spp.2015, USA9VRE (88.9) (2 E. faecalis, 7 E. faecium)in vitro (TK)Ceftriaxone4 (44.4)5 (55.6)DNS isolates.42
9Cefazolin2 (22.2)7 (77.8)
9Ceftaroline5 (55.6)4 (44.4)
SA2015, USA3MRSA (100)in vitro (PK/PD biofilm model)Ceftaroline3 (100)43
SA2016, USA2MRSA (100)in vitro (Hollow-fibre model)Cefazolin2 (100)Isogenic pair of DAP-susceptible and DNS isolates. High inoculum tested.44
2Ceftolozane/tazobactam1 (50)1 (50)
SA2016, USA1MRSA (100)in vivo (rabbit, IE, spleen and kidney dissemination)Ceftriaxone1 (100)Synergistic effect on spleen. In the in vivo study, equivalent human dosage of DAP 12 mg/kg was administered.45
SA2016, Spain71MRSA (100)in vitro (ET)Ceftaroline10 (14.1)41 (57.7)20 (28.2)18 isolates were also resistant to linezolid.46
SA2017, Turkey30MRSA (100)in vitro (CB)Ceftriaxone23 (76.7)7 (23.3)No distinction between additive and indifferent effect.47
SA2017, USA2MRSA (100)in vitro (TK)Ceftaroline2 (100)Isogenic pair with one DNS strain.48
SA2019, Italy17MRSA (35.3), hVISA (29.4)in vitro (CB,TK)Ceftobiprole4 (23.5)6 (35.3)7 (41.2)49
SE16MRSE (37.5)Ceftobiprole8 (50)3 (18.8)5 (31.2)
E. faecium6VRE (83.3)Ceftobiprole1 (16.7)5 (83.3)
E. faecalis7VRE (71.4)Ceftobiprole2 (28.6)5 (71.4)
SA2019, Taiwan15VISA (40), hVISA (60)in vitro (CB, TK)Cefazolin15 (100)DNS isolates.52
15Cefmetazole13 (86.7)2 (13.3)
15Cefotaxime15 (100)
15Cefepime12 (80)3 (20)
SA2020, USA4VISA (50)in vitro (TK, PK/PD model)Cefazolin4 (100)DNS isolates.50
SA2020, USA13MRSA (69.2)in vitro (TK)Ceftaroline8 (61.5)5 (38.5)8 DNS isolates.51
E. faecalis2021, USA2VRE (100)in vitro (TK)Ceftriaxone1 (50)1 (50)33
E. faecium2Ceftriaxone2 (100)
SA2022, Taiwan10MRSA (100)in vitro (CB, TK)Ceftaroline3 (30)7 (70)53
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2005, USA2MRSA (100)in vitro (PK/PD model)Cefepime2 (100)34
SA2005, USA40MRSA (50)in vitro (ET) (TK on 1 MRSA)CefepimeET: 2 (5); TK: 1 (100)ET: 38 (95)No distinction between additive and indifferent effect.14
in vitro (ET)CeftriaxoneET: 1 (2.5)ET: 39 (97.5)
E. faecalis, E. faecium40VRE (50)in vitro (ET) (TK on 1 VSE)CefepimeET: 9 (22.5)ET: 31 (77.5); TK: 1 (100)
in vitro (ET) (TK on 2 VSE and 1 VRE)CeftriaxoneET: 18 (45)ET: 22 (55); TK: 3 (100)
SA2010, USA2MRSA (100)in vitro (PD model)Cefepime1 (50)1 (50)DNS isolates. High inoculum tested.19
SA2013, USA2hVISA (50)in vitro (PK/PD model)Ceftaroline2 (100)Two isogenic isolates, one of which mutated into a hVISA DNS strain. High inoculum tested.35
SA2014, USA3MRSA (66.7), hVISA (33.3)in vitro (biofilm model)Ceftaroline, cefazolin3 (100)Synergy was observed against all isolated for both combinations.36
SA2014, USA3VISA (100)in vitro (PK/PD model)Ceftaroline2 (66.7)1 (33.3)DNS isolates. High inoculum tested.37
E. faecium2014, USA2VRE (100)in vitro (PK/PD model)Ceftriaxone1 (50)1 (50)High inoculum tested.38
E. faecalis1Ceftriaxone1 (100)
SA2014, USA2MRSA (50)in vitro (CB, TK)Ceftaroline2 (100)39
SA2014, USA5MRSA (60); VISA (40)in vitro (TK)Ceftobiprole5 (100)40
SA2015, USA1MRSA (100)in vitro (PK/PD model)Ceftaroline1 (100)41
E. faecalis2015, USA2VRE (100)in vitro (CB, TK)Ceftaroline2 (100)25
2Cefepime1 (50)1 (50)
2Ceftriaxone1 (50)1 (50)
2Cefotaxime1 (50)1 (50)
2Cefazolin2 (100)
E. faecium2Ceftaroline2 (100)
2Cefepime1 (50)1 (50)
2Ceftriaxone1 (50)1 (50)
2Cefotaxime1 (50)1 (50)
2Cefazolin1 (50)1 (50)
E. faecium2015, USA4VRE (100)in vitro (TK)Ceftobiprole2 (50)2 (50)DNS isolates.26
E. faecalis2Ceftobiprole2 (100)
E. faecalis2015, USA1VRE (100)in vitro (PK/PD model)Ceftaroline1 (100)27
E. faecium2Ceftaroline2 (100)
Enterococcus spp.2015, USA9VRE (88.9) (2 E. faecalis, 7 E. faecium)in vitro (TK)Ceftriaxone4 (44.4)5 (55.6)DNS isolates.42
9Cefazolin2 (22.2)7 (77.8)
9Ceftaroline5 (55.6)4 (44.4)
SA2015, USA3MRSA (100)in vitro (PK/PD biofilm model)Ceftaroline3 (100)43
SA2016, USA2MRSA (100)in vitro (Hollow-fibre model)Cefazolin2 (100)Isogenic pair of DAP-susceptible and DNS isolates. High inoculum tested.44
2Ceftolozane/tazobactam1 (50)1 (50)
SA2016, USA1MRSA (100)in vivo (rabbit, IE, spleen and kidney dissemination)Ceftriaxone1 (100)Synergistic effect on spleen. In the in vivo study, equivalent human dosage of DAP 12 mg/kg was administered.45
SA2016, Spain71MRSA (100)in vitro (ET)Ceftaroline10 (14.1)41 (57.7)20 (28.2)18 isolates were also resistant to linezolid.46
SA2017, Turkey30MRSA (100)in vitro (CB)Ceftriaxone23 (76.7)7 (23.3)No distinction between additive and indifferent effect.47
SA2017, USA2MRSA (100)in vitro (TK)Ceftaroline2 (100)Isogenic pair with one DNS strain.48
SA2019, Italy17MRSA (35.3), hVISA (29.4)in vitro (CB,TK)Ceftobiprole4 (23.5)6 (35.3)7 (41.2)49
SE16MRSE (37.5)Ceftobiprole8 (50)3 (18.8)5 (31.2)
E. faecium6VRE (83.3)Ceftobiprole1 (16.7)5 (83.3)
E. faecalis7VRE (71.4)Ceftobiprole2 (28.6)5 (71.4)
SA2019, Taiwan15VISA (40), hVISA (60)in vitro (CB, TK)Cefazolin15 (100)DNS isolates.52
15Cefmetazole13 (86.7)2 (13.3)
15Cefotaxime15 (100)
15Cefepime12 (80)3 (20)
SA2020, USA4VISA (50)in vitro (TK, PK/PD model)Cefazolin4 (100)DNS isolates.50
SA2020, USA13MRSA (69.2)in vitro (TK)Ceftaroline8 (61.5)5 (38.5)8 DNS isolates.51
E. faecalis2021, USA2VRE (100)in vitro (TK)Ceftriaxone1 (50)1 (50)33
E. faecium2Ceftriaxone2 (100)
SA2022, Taiwan10MRSA (100)in vitro (CB, TK)Ceftaroline3 (30)7 (70)53

CB, chequerboard; ET, Etest; IE, infective endocarditis; DAP, daptomycin; DNS, daptomycin non-susceptible; SA, S. aureus; SE, S. epidermidis; TK, time–kill assay.

Table 3.

Studies evaluating daptomycin in combination with cephalosporins and cephalosporins + BLIs

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2005, USA2MRSA (100)in vitro (PK/PD model)Cefepime2 (100)34
SA2005, USA40MRSA (50)in vitro (ET) (TK on 1 MRSA)CefepimeET: 2 (5); TK: 1 (100)ET: 38 (95)No distinction between additive and indifferent effect.14
in vitro (ET)CeftriaxoneET: 1 (2.5)ET: 39 (97.5)
E. faecalis, E. faecium40VRE (50)in vitro (ET) (TK on 1 VSE)CefepimeET: 9 (22.5)ET: 31 (77.5); TK: 1 (100)
in vitro (ET) (TK on 2 VSE and 1 VRE)CeftriaxoneET: 18 (45)ET: 22 (55); TK: 3 (100)
SA2010, USA2MRSA (100)in vitro (PD model)Cefepime1 (50)1 (50)DNS isolates. High inoculum tested.19
SA2013, USA2hVISA (50)in vitro (PK/PD model)Ceftaroline2 (100)Two isogenic isolates, one of which mutated into a hVISA DNS strain. High inoculum tested.35
SA2014, USA3MRSA (66.7), hVISA (33.3)in vitro (biofilm model)Ceftaroline, cefazolin3 (100)Synergy was observed against all isolated for both combinations.36
SA2014, USA3VISA (100)in vitro (PK/PD model)Ceftaroline2 (66.7)1 (33.3)DNS isolates. High inoculum tested.37
E. faecium2014, USA2VRE (100)in vitro (PK/PD model)Ceftriaxone1 (50)1 (50)High inoculum tested.38
E. faecalis1Ceftriaxone1 (100)
SA2014, USA2MRSA (50)in vitro (CB, TK)Ceftaroline2 (100)39
SA2014, USA5MRSA (60); VISA (40)in vitro (TK)Ceftobiprole5 (100)40
SA2015, USA1MRSA (100)in vitro (PK/PD model)Ceftaroline1 (100)41
E. faecalis2015, USA2VRE (100)in vitro (CB, TK)Ceftaroline2 (100)25
2Cefepime1 (50)1 (50)
2Ceftriaxone1 (50)1 (50)
2Cefotaxime1 (50)1 (50)
2Cefazolin2 (100)
E. faecium2Ceftaroline2 (100)
2Cefepime1 (50)1 (50)
2Ceftriaxone1 (50)1 (50)
2Cefotaxime1 (50)1 (50)
2Cefazolin1 (50)1 (50)
E. faecium2015, USA4VRE (100)in vitro (TK)Ceftobiprole2 (50)2 (50)DNS isolates.26
E. faecalis2Ceftobiprole2 (100)
E. faecalis2015, USA1VRE (100)in vitro (PK/PD model)Ceftaroline1 (100)27
E. faecium2Ceftaroline2 (100)
Enterococcus spp.2015, USA9VRE (88.9) (2 E. faecalis, 7 E. faecium)in vitro (TK)Ceftriaxone4 (44.4)5 (55.6)DNS isolates.42
9Cefazolin2 (22.2)7 (77.8)
9Ceftaroline5 (55.6)4 (44.4)
SA2015, USA3MRSA (100)in vitro (PK/PD biofilm model)Ceftaroline3 (100)43
SA2016, USA2MRSA (100)in vitro (Hollow-fibre model)Cefazolin2 (100)Isogenic pair of DAP-susceptible and DNS isolates. High inoculum tested.44
2Ceftolozane/tazobactam1 (50)1 (50)
SA2016, USA1MRSA (100)in vivo (rabbit, IE, spleen and kidney dissemination)Ceftriaxone1 (100)Synergistic effect on spleen. In the in vivo study, equivalent human dosage of DAP 12 mg/kg was administered.45
SA2016, Spain71MRSA (100)in vitro (ET)Ceftaroline10 (14.1)41 (57.7)20 (28.2)18 isolates were also resistant to linezolid.46
SA2017, Turkey30MRSA (100)in vitro (CB)Ceftriaxone23 (76.7)7 (23.3)No distinction between additive and indifferent effect.47
SA2017, USA2MRSA (100)in vitro (TK)Ceftaroline2 (100)Isogenic pair with one DNS strain.48
SA2019, Italy17MRSA (35.3), hVISA (29.4)in vitro (CB,TK)Ceftobiprole4 (23.5)6 (35.3)7 (41.2)49
SE16MRSE (37.5)Ceftobiprole8 (50)3 (18.8)5 (31.2)
E. faecium6VRE (83.3)Ceftobiprole1 (16.7)5 (83.3)
E. faecalis7VRE (71.4)Ceftobiprole2 (28.6)5 (71.4)
SA2019, Taiwan15VISA (40), hVISA (60)in vitro (CB, TK)Cefazolin15 (100)DNS isolates.52
15Cefmetazole13 (86.7)2 (13.3)
15Cefotaxime15 (100)
15Cefepime12 (80)3 (20)
SA2020, USA4VISA (50)in vitro (TK, PK/PD model)Cefazolin4 (100)DNS isolates.50
SA2020, USA13MRSA (69.2)in vitro (TK)Ceftaroline8 (61.5)5 (38.5)8 DNS isolates.51
E. faecalis2021, USA2VRE (100)in vitro (TK)Ceftriaxone1 (50)1 (50)33
E. faecium2Ceftriaxone2 (100)
SA2022, Taiwan10MRSA (100)in vitro (CB, TK)Ceftaroline3 (30)7 (70)53
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2005, USA2MRSA (100)in vitro (PK/PD model)Cefepime2 (100)34
SA2005, USA40MRSA (50)in vitro (ET) (TK on 1 MRSA)CefepimeET: 2 (5); TK: 1 (100)ET: 38 (95)No distinction between additive and indifferent effect.14
in vitro (ET)CeftriaxoneET: 1 (2.5)ET: 39 (97.5)
E. faecalis, E. faecium40VRE (50)in vitro (ET) (TK on 1 VSE)CefepimeET: 9 (22.5)ET: 31 (77.5); TK: 1 (100)
in vitro (ET) (TK on 2 VSE and 1 VRE)CeftriaxoneET: 18 (45)ET: 22 (55); TK: 3 (100)
SA2010, USA2MRSA (100)in vitro (PD model)Cefepime1 (50)1 (50)DNS isolates. High inoculum tested.19
SA2013, USA2hVISA (50)in vitro (PK/PD model)Ceftaroline2 (100)Two isogenic isolates, one of which mutated into a hVISA DNS strain. High inoculum tested.35
SA2014, USA3MRSA (66.7), hVISA (33.3)in vitro (biofilm model)Ceftaroline, cefazolin3 (100)Synergy was observed against all isolated for both combinations.36
SA2014, USA3VISA (100)in vitro (PK/PD model)Ceftaroline2 (66.7)1 (33.3)DNS isolates. High inoculum tested.37
E. faecium2014, USA2VRE (100)in vitro (PK/PD model)Ceftriaxone1 (50)1 (50)High inoculum tested.38
E. faecalis1Ceftriaxone1 (100)
SA2014, USA2MRSA (50)in vitro (CB, TK)Ceftaroline2 (100)39
SA2014, USA5MRSA (60); VISA (40)in vitro (TK)Ceftobiprole5 (100)40
SA2015, USA1MRSA (100)in vitro (PK/PD model)Ceftaroline1 (100)41
E. faecalis2015, USA2VRE (100)in vitro (CB, TK)Ceftaroline2 (100)25
2Cefepime1 (50)1 (50)
2Ceftriaxone1 (50)1 (50)
2Cefotaxime1 (50)1 (50)
2Cefazolin2 (100)
E. faecium2Ceftaroline2 (100)
2Cefepime1 (50)1 (50)
2Ceftriaxone1 (50)1 (50)
2Cefotaxime1 (50)1 (50)
2Cefazolin1 (50)1 (50)
E. faecium2015, USA4VRE (100)in vitro (TK)Ceftobiprole2 (50)2 (50)DNS isolates.26
E. faecalis2Ceftobiprole2 (100)
E. faecalis2015, USA1VRE (100)in vitro (PK/PD model)Ceftaroline1 (100)27
E. faecium2Ceftaroline2 (100)
Enterococcus spp.2015, USA9VRE (88.9) (2 E. faecalis, 7 E. faecium)in vitro (TK)Ceftriaxone4 (44.4)5 (55.6)DNS isolates.42
9Cefazolin2 (22.2)7 (77.8)
9Ceftaroline5 (55.6)4 (44.4)
SA2015, USA3MRSA (100)in vitro (PK/PD biofilm model)Ceftaroline3 (100)43
SA2016, USA2MRSA (100)in vitro (Hollow-fibre model)Cefazolin2 (100)Isogenic pair of DAP-susceptible and DNS isolates. High inoculum tested.44
2Ceftolozane/tazobactam1 (50)1 (50)
SA2016, USA1MRSA (100)in vivo (rabbit, IE, spleen and kidney dissemination)Ceftriaxone1 (100)Synergistic effect on spleen. In the in vivo study, equivalent human dosage of DAP 12 mg/kg was administered.45
SA2016, Spain71MRSA (100)in vitro (ET)Ceftaroline10 (14.1)41 (57.7)20 (28.2)18 isolates were also resistant to linezolid.46
SA2017, Turkey30MRSA (100)in vitro (CB)Ceftriaxone23 (76.7)7 (23.3)No distinction between additive and indifferent effect.47
SA2017, USA2MRSA (100)in vitro (TK)Ceftaroline2 (100)Isogenic pair with one DNS strain.48
SA2019, Italy17MRSA (35.3), hVISA (29.4)in vitro (CB,TK)Ceftobiprole4 (23.5)6 (35.3)7 (41.2)49
SE16MRSE (37.5)Ceftobiprole8 (50)3 (18.8)5 (31.2)
E. faecium6VRE (83.3)Ceftobiprole1 (16.7)5 (83.3)
E. faecalis7VRE (71.4)Ceftobiprole2 (28.6)5 (71.4)
SA2019, Taiwan15VISA (40), hVISA (60)in vitro (CB, TK)Cefazolin15 (100)DNS isolates.52
15Cefmetazole13 (86.7)2 (13.3)
15Cefotaxime15 (100)
15Cefepime12 (80)3 (20)
SA2020, USA4VISA (50)in vitro (TK, PK/PD model)Cefazolin4 (100)DNS isolates.50
SA2020, USA13MRSA (69.2)in vitro (TK)Ceftaroline8 (61.5)5 (38.5)8 DNS isolates.51
E. faecalis2021, USA2VRE (100)in vitro (TK)Ceftriaxone1 (50)1 (50)33
E. faecium2Ceftriaxone2 (100)
SA2022, Taiwan10MRSA (100)in vitro (CB, TK)Ceftaroline3 (30)7 (70)53

CB, chequerboard; ET, Etest; IE, infective endocarditis; DAP, daptomycin; DNS, daptomycin non-susceptible; SA, S. aureus; SE, S. epidermidis; TK, time–kill assay.

S. aureus (MSSA, MRSA, hVISA, VISA) and S. epidermidis [methicillin-susceptible S. epidermidis (MSSE), MRSE]

A total of 228 S. aureus isolates (34 MSSA, 165 MRSA, 16 hVISA, 13 VISA) were tested. In in vitro studies (227 isolates tested), the combination had an indifferent effect against 82 isolates (36.1%). On the other hand, synergism was observed when the combination was tested against 75 (33%) isolates. One study evaluated one strain in an in vivo model of infective endocarditis with spleen and kidney dissemination, reporting an indifferent effect of the combination daptomycin + ceftriaxone on valve vegetations, but a synergistic effect on spleen septic emboli.45

Against 16 isolates of S. epidermidis (5 MSSE, 6 MRSE, 5 linezolid-resistant S. epidermidis), the most frequent effect of the combination was synergistic (8 isolates; 50%), followed by indifferent effect (31.2%) and additive effect (18.8%).

Enterococcus spp. (VSE, VRE)

A total of 82 isolates of Enterococcus spp. (24 VSE, 58 VRE) were tested in vitro. Indifferent effect was observed in most cases (51 isolates; 62.2%), while synergism was observed against 23 isolates (28%). No antagonism was observed against any of the tested strains.

Carbapenems

Seven studies evaluated daptomycin + carbapenems in vitro (Table 4).14,17,22,25,27,42,45

Table 4.

Studies evaluating daptomycin in combination with carbapenems

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2005, USA40MRSA (50)in vitro (ET)ImipenemET: 1 (2.5)ET: 39 (97.5)No distinction between additive and indifferent effect.14
E. faecalis, E. faecium40VRE (50)in vitro (ET) (TK on 1 VSE and 1 VRE)ImipenemET: 9 (22.5)ET: 31 (77.5); TK: 2 (100)
SA2008, Taiwan1VISA (100)in vitro (TK)Imipenem1 (100)17
SA2012, USA2MRSA (100)in vitro (TK)Imipenem2 (100)22
E. faecalis2015, USA2VRE (100)in vitro (CB, TK)Ertapenem1 (50)1 (50)25
E. faecium2Ertapenem2 (100)
E. faecalis2015, USA1VRE (100)in vitro (PK/PD model)Ertapenem1 (100)27
E. faecium2Ertapenem2 (100)
Enterococcus spp.2015, USA9VRE (88.9) (2 E. faecalis and 7 E. faecium)in vitro (TK)Ertapenem4 (44.4)5 (55.6)DNS isolates.42
SA2016, USA1MRSA (100)in vivo (rabbit, IE, spleen and kidney dissemination)Ertapenem1 (100)Synergistic effect on kidneys. In the in vivo study, equivalent human dosage of DAP 12 mg/kg was administered.45
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2005, USA40MRSA (50)in vitro (ET)ImipenemET: 1 (2.5)ET: 39 (97.5)No distinction between additive and indifferent effect.14
E. faecalis, E. faecium40VRE (50)in vitro (ET) (TK on 1 VSE and 1 VRE)ImipenemET: 9 (22.5)ET: 31 (77.5); TK: 2 (100)
SA2008, Taiwan1VISA (100)in vitro (TK)Imipenem1 (100)17
SA2012, USA2MRSA (100)in vitro (TK)Imipenem2 (100)22
E. faecalis2015, USA2VRE (100)in vitro (CB, TK)Ertapenem1 (50)1 (50)25
E. faecium2Ertapenem2 (100)
E. faecalis2015, USA1VRE (100)in vitro (PK/PD model)Ertapenem1 (100)27
E. faecium2Ertapenem2 (100)
Enterococcus spp.2015, USA9VRE (88.9) (2 E. faecalis and 7 E. faecium)in vitro (TK)Ertapenem4 (44.4)5 (55.6)DNS isolates.42
SA2016, USA1MRSA (100)in vivo (rabbit, IE, spleen and kidney dissemination)Ertapenem1 (100)Synergistic effect on kidneys. In the in vivo study, equivalent human dosage of DAP 12 mg/kg was administered.45

CB, chequerboard; ET, Etest; DAP, daptomycin; DNS, daptomycin non-susceptible; SA, S. aureus; TK, time–kill assay.

Table 4.

Studies evaluating daptomycin in combination with carbapenems

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2005, USA40MRSA (50)in vitro (ET)ImipenemET: 1 (2.5)ET: 39 (97.5)No distinction between additive and indifferent effect.14
E. faecalis, E. faecium40VRE (50)in vitro (ET) (TK on 1 VSE and 1 VRE)ImipenemET: 9 (22.5)ET: 31 (77.5); TK: 2 (100)
SA2008, Taiwan1VISA (100)in vitro (TK)Imipenem1 (100)17
SA2012, USA2MRSA (100)in vitro (TK)Imipenem2 (100)22
E. faecalis2015, USA2VRE (100)in vitro (CB, TK)Ertapenem1 (50)1 (50)25
E. faecium2Ertapenem2 (100)
E. faecalis2015, USA1VRE (100)in vitro (PK/PD model)Ertapenem1 (100)27
E. faecium2Ertapenem2 (100)
Enterococcus spp.2015, USA9VRE (88.9) (2 E. faecalis and 7 E. faecium)in vitro (TK)Ertapenem4 (44.4)5 (55.6)DNS isolates.42
SA2016, USA1MRSA (100)in vivo (rabbit, IE, spleen and kidney dissemination)Ertapenem1 (100)Synergistic effect on kidneys. In the in vivo study, equivalent human dosage of DAP 12 mg/kg was administered.45
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2005, USA40MRSA (50)in vitro (ET)ImipenemET: 1 (2.5)ET: 39 (97.5)No distinction between additive and indifferent effect.14
E. faecalis, E. faecium40VRE (50)in vitro (ET) (TK on 1 VSE and 1 VRE)ImipenemET: 9 (22.5)ET: 31 (77.5); TK: 2 (100)
SA2008, Taiwan1VISA (100)in vitro (TK)Imipenem1 (100)17
SA2012, USA2MRSA (100)in vitro (TK)Imipenem2 (100)22
E. faecalis2015, USA2VRE (100)in vitro (CB, TK)Ertapenem1 (50)1 (50)25
E. faecium2Ertapenem2 (100)
E. faecalis2015, USA1VRE (100)in vitro (PK/PD model)Ertapenem1 (100)27
E. faecium2Ertapenem2 (100)
Enterococcus spp.2015, USA9VRE (88.9) (2 E. faecalis and 7 E. faecium)in vitro (TK)Ertapenem4 (44.4)5 (55.6)DNS isolates.42
SA2016, USA1MRSA (100)in vivo (rabbit, IE, spleen and kidney dissemination)Ertapenem1 (100)Synergistic effect on kidneys. In the in vivo study, equivalent human dosage of DAP 12 mg/kg was administered.45

CB, chequerboard; ET, Etest; DAP, daptomycin; DNS, daptomycin non-susceptible; SA, S. aureus; TK, time–kill assay.

S. aureus (MSSA, MRSA, VISA)

Forty-four isolates of S. aureus (20 MSSA, 23 MRSA, 1 VISA) were tested in three in vitro studies, and the combination had an indifferent effect in most cases (90.7%). One study evaluated one strain in an in vivo model of infective endocarditis with spleen and kidney dissemination, reporting an indifferent effect of the combination daptomycin + ertapenem on valve vegetations, but a synergistic effect on kidneys.45

Enterococcus spp. (VSE, VRE)

When daptomycin + carbapenems was tested against Enterococcus spp. (56 isolates; 21 VSE, 35 VRE), synergism was observed against 19 isolates (33.9%), additive effect against 1 isolate (1.8%) and indifferent effect against 36 isolates (64.3%). No antagonistic effect was reported.

Macrolides

S. aureus (MSSA, MRSA, hVISA) and S. epidermidis (MSSE, MRSE)

Four in vitro and one in vivo studies evaluated daptomycin + macrolides against 5 S. aureus isolates (1 MSSA, 3 MRSA, 1 hVISA) and 15 S. epidermidis isolates (10 MSSE, 5 MRSE) (Table 5).54–58 Overall, the combination exerted indifferent effect against most S. aureus and S. epidermidis isolates. In fact, against 4 S. aureus isolates (80%), including 2 isolates tested in vivo, and against 14 S. epidermidis isolates (93.3%), the combination showed no significant enhanced activity compared with monotherapy. Synergism was reported sporadically (two isolates). One study reporting synergistic activity of the combination against one S. aureus isolate described enhanced antibacterial activity against both planktonic and biofilm bacteria.55 No antagonistic effect was reported.

Table 5.

Studies evaluating daptomycin in combination with macrolides

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SE2009, Austria10in vitro (CB)Azithromycin10 (100)54
SA2010, USA1in vitro (PK/PD model)Clarithromycin1 (100)Synergism was observed both on planktonic and biofilm bacteria.55
SA2014, USA2MRSA (50), hVISA (50)in vitro (PK/PD biofilm model)Clarithromycin2 (100)Clarithromycin-resistant isolates.56
SE1MRSE (100)Clarithromycin1 (100)
SE2015, Spain4MRSE (100)in vitro (PK/PD catheter model)Clarithromycin1 (25)3 (75)57
SA2016, Spain2MRSA (50)in vivo (rat, foreign body infection)Clarithromycin2 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.58
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SE2009, Austria10in vitro (CB)Azithromycin10 (100)54
SA2010, USA1in vitro (PK/PD model)Clarithromycin1 (100)Synergism was observed both on planktonic and biofilm bacteria.55
SA2014, USA2MRSA (50), hVISA (50)in vitro (PK/PD biofilm model)Clarithromycin2 (100)Clarithromycin-resistant isolates.56
SE1MRSE (100)Clarithromycin1 (100)
SE2015, Spain4MRSE (100)in vitro (PK/PD catheter model)Clarithromycin1 (25)3 (75)57
SA2016, Spain2MRSA (50)in vivo (rat, foreign body infection)Clarithromycin2 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.58

CB, chequerboard; DAP, daptomycin; SA, S. aureus; SE, S. epidermidis.

Table 5.

Studies evaluating daptomycin in combination with macrolides

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SE2009, Austria10in vitro (CB)Azithromycin10 (100)54
SA2010, USA1in vitro (PK/PD model)Clarithromycin1 (100)Synergism was observed both on planktonic and biofilm bacteria.55
SA2014, USA2MRSA (50), hVISA (50)in vitro (PK/PD biofilm model)Clarithromycin2 (100)Clarithromycin-resistant isolates.56
SE1MRSE (100)Clarithromycin1 (100)
SE2015, Spain4MRSE (100)in vitro (PK/PD catheter model)Clarithromycin1 (25)3 (75)57
SA2016, Spain2MRSA (50)in vivo (rat, foreign body infection)Clarithromycin2 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.58
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SE2009, Austria10in vitro (CB)Azithromycin10 (100)54
SA2010, USA1in vitro (PK/PD model)Clarithromycin1 (100)Synergism was observed both on planktonic and biofilm bacteria.55
SA2014, USA2MRSA (50), hVISA (50)in vitro (PK/PD biofilm model)Clarithromycin2 (100)Clarithromycin-resistant isolates.56
SE1MRSE (100)Clarithromycin1 (100)
SE2015, Spain4MRSE (100)in vitro (PK/PD catheter model)Clarithromycin1 (25)3 (75)57
SA2016, Spain2MRSA (50)in vivo (rat, foreign body infection)Clarithromycin2 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.58

CB, chequerboard; DAP, daptomycin; SA, S. aureus; SE, S. epidermidis.

Aminoglycosides

The combination daptomycin + gentamicin was evaluated in 16 studies, and daptomycin + tobramycin in one study (Table 6).14–16,30,42,57,59–69

Table 6.

Studies evaluating daptomycin in combination with aminoglycosides

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2004, USA2MRSA (50)in vitro (PK/PD model)Gentamicin2 (100)An additive effect was described also when a high inoculum was tested.59
SA2005, USA2MRSA (50)in vitro (PK/PD model)Gentamicin2 (100)High inoculum tested.60
SA2005, USA40MRSA (50)in vitro (ET) (TK on 1 MSSA and 1 MRSA)GentamicinET: 1 (2.5); TK 1 (50)ET: 39 (97.5); TK 1 (50)14
E. faecalis, E. faecium40VRE (50)in vitro (ET)GentamicinET: 3 (7.5)ET: 37 (92.5)
SA2006, USA2GISA (100)in vitro (ET, TK)GentamicinET: 2 (100); TK: 1 (50)TK: 1 (50)15
E. faecium2006, Turkey42VRE (100)in vitro (ET)Gentamicin9 (21.4)33 (78.6)No distinction between additive and indifferent effect.16
SA2007, USA49VISA (12), VRSA (6), MRSA (64)in vitro (TK)Gentamicin34 (68)16 (32)61
SA2008, USA2in vitro (TK)Gentamicin2 (100)62
SA2009, USA2MRSA (100)in vitro (PK/PD model)Gentamicin2 (100)High inoculum tested. Both isolates exhibited biofilm formation. DAP monotherapy demonstrated significantly better activity than DAP in combination.63
SA2009, Spain3MRSA (100)in vitro (TK); in vivo (rabbit, IE)GentamicinTK: 3 (100)in vivo: 1 (100)In the in vivo study, equivalent human dosage of DAP 6 mg/kg was administered.64
E. faecalis2011, Switzerland1in vitro (TK), in vivo (Guinea pig, foreign body infection model)GentamicinTK: 1 (100); in vivo: 1 (100)In the in vivo study, equivalent human dosage of DAP 10 mg/kg was administered.65
E. faecalis2014, USA1in vitro (PK/PD model), in vivo (G. mellonella infection model)Gentamicinin vitro: 1 (100)in vivo: 1 (100)High inoculum tested. In the in vivo study, equivalent human dosage of DAP 6 and 10 mg/kg was administered (indifferent effect observed with both dosages).66
E. faecium1VRE (100)Gentamicinin vitro: 1 (100); in vivo: 1 (100)
Enterococcus spp.2015, USA4VRE (100)in vitro (TK)Gentamicin3 (75)1 (25)42
SE2015, Spain4MRSE (100)in vitro (PK/PD catheter model)Gentamicin3 (75)1 (25)57
SA2015, Australia8MRSA (50), VISA (25)in vitro (TK)Gentamicin4 (50)3 (37.5)1 (12.5)4 DNS isolates.67
SA2017, Turkey25MRSA (100)in vitro (CB)Gentamicin17 (68)7 (28)1 (4)68
SA2019, Taiwan100MRSA (100)in vitro (CB)Gentamicin5 (5)38 (38)57 (57)No distinction between indifferent and antagonistic effect.30
SA2022, India3in vitro (CB, TK)Tobramycin3 (100)69
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2004, USA2MRSA (50)in vitro (PK/PD model)Gentamicin2 (100)An additive effect was described also when a high inoculum was tested.59
SA2005, USA2MRSA (50)in vitro (PK/PD model)Gentamicin2 (100)High inoculum tested.60
SA2005, USA40MRSA (50)in vitro (ET) (TK on 1 MSSA and 1 MRSA)GentamicinET: 1 (2.5); TK 1 (50)ET: 39 (97.5); TK 1 (50)14
E. faecalis, E. faecium40VRE (50)in vitro (ET)GentamicinET: 3 (7.5)ET: 37 (92.5)
SA2006, USA2GISA (100)in vitro (ET, TK)GentamicinET: 2 (100); TK: 1 (50)TK: 1 (50)15
E. faecium2006, Turkey42VRE (100)in vitro (ET)Gentamicin9 (21.4)33 (78.6)No distinction between additive and indifferent effect.16
SA2007, USA49VISA (12), VRSA (6), MRSA (64)in vitro (TK)Gentamicin34 (68)16 (32)61
SA2008, USA2in vitro (TK)Gentamicin2 (100)62
SA2009, USA2MRSA (100)in vitro (PK/PD model)Gentamicin2 (100)High inoculum tested. Both isolates exhibited biofilm formation. DAP monotherapy demonstrated significantly better activity than DAP in combination.63
SA2009, Spain3MRSA (100)in vitro (TK); in vivo (rabbit, IE)GentamicinTK: 3 (100)in vivo: 1 (100)In the in vivo study, equivalent human dosage of DAP 6 mg/kg was administered.64
E. faecalis2011, Switzerland1in vitro (TK), in vivo (Guinea pig, foreign body infection model)GentamicinTK: 1 (100); in vivo: 1 (100)In the in vivo study, equivalent human dosage of DAP 10 mg/kg was administered.65
E. faecalis2014, USA1in vitro (PK/PD model), in vivo (G. mellonella infection model)Gentamicinin vitro: 1 (100)in vivo: 1 (100)High inoculum tested. In the in vivo study, equivalent human dosage of DAP 6 and 10 mg/kg was administered (indifferent effect observed with both dosages).66
E. faecium1VRE (100)Gentamicinin vitro: 1 (100); in vivo: 1 (100)
Enterococcus spp.2015, USA4VRE (100)in vitro (TK)Gentamicin3 (75)1 (25)42
SE2015, Spain4MRSE (100)in vitro (PK/PD catheter model)Gentamicin3 (75)1 (25)57
SA2015, Australia8MRSA (50), VISA (25)in vitro (TK)Gentamicin4 (50)3 (37.5)1 (12.5)4 DNS isolates.67
SA2017, Turkey25MRSA (100)in vitro (CB)Gentamicin17 (68)7 (28)1 (4)68
SA2019, Taiwan100MRSA (100)in vitro (CB)Gentamicin5 (5)38 (38)57 (57)No distinction between indifferent and antagonistic effect.30
SA2022, India3in vitro (CB, TK)Tobramycin3 (100)69

CB, chequerboard; ET, Etest; IE, infective endocarditis; DAP, daptomycin; DNS, daptomycin non-susceptible; SA, S. aureus; SE, S. epidermidis; TK, time–kill assay.

Table 6.

Studies evaluating daptomycin in combination with aminoglycosides

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2004, USA2MRSA (50)in vitro (PK/PD model)Gentamicin2 (100)An additive effect was described also when a high inoculum was tested.59
SA2005, USA2MRSA (50)in vitro (PK/PD model)Gentamicin2 (100)High inoculum tested.60
SA2005, USA40MRSA (50)in vitro (ET) (TK on 1 MSSA and 1 MRSA)GentamicinET: 1 (2.5); TK 1 (50)ET: 39 (97.5); TK 1 (50)14
E. faecalis, E. faecium40VRE (50)in vitro (ET)GentamicinET: 3 (7.5)ET: 37 (92.5)
SA2006, USA2GISA (100)in vitro (ET, TK)GentamicinET: 2 (100); TK: 1 (50)TK: 1 (50)15
E. faecium2006, Turkey42VRE (100)in vitro (ET)Gentamicin9 (21.4)33 (78.6)No distinction between additive and indifferent effect.16
SA2007, USA49VISA (12), VRSA (6), MRSA (64)in vitro (TK)Gentamicin34 (68)16 (32)61
SA2008, USA2in vitro (TK)Gentamicin2 (100)62
SA2009, USA2MRSA (100)in vitro (PK/PD model)Gentamicin2 (100)High inoculum tested. Both isolates exhibited biofilm formation. DAP monotherapy demonstrated significantly better activity than DAP in combination.63
SA2009, Spain3MRSA (100)in vitro (TK); in vivo (rabbit, IE)GentamicinTK: 3 (100)in vivo: 1 (100)In the in vivo study, equivalent human dosage of DAP 6 mg/kg was administered.64
E. faecalis2011, Switzerland1in vitro (TK), in vivo (Guinea pig, foreign body infection model)GentamicinTK: 1 (100); in vivo: 1 (100)In the in vivo study, equivalent human dosage of DAP 10 mg/kg was administered.65
E. faecalis2014, USA1in vitro (PK/PD model), in vivo (G. mellonella infection model)Gentamicinin vitro: 1 (100)in vivo: 1 (100)High inoculum tested. In the in vivo study, equivalent human dosage of DAP 6 and 10 mg/kg was administered (indifferent effect observed with both dosages).66
E. faecium1VRE (100)Gentamicinin vitro: 1 (100); in vivo: 1 (100)
Enterococcus spp.2015, USA4VRE (100)in vitro (TK)Gentamicin3 (75)1 (25)42
SE2015, Spain4MRSE (100)in vitro (PK/PD catheter model)Gentamicin3 (75)1 (25)57
SA2015, Australia8MRSA (50), VISA (25)in vitro (TK)Gentamicin4 (50)3 (37.5)1 (12.5)4 DNS isolates.67
SA2017, Turkey25MRSA (100)in vitro (CB)Gentamicin17 (68)7 (28)1 (4)68
SA2019, Taiwan100MRSA (100)in vitro (CB)Gentamicin5 (5)38 (38)57 (57)No distinction between indifferent and antagonistic effect.30
SA2022, India3in vitro (CB, TK)Tobramycin3 (100)69
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2004, USA2MRSA (50)in vitro (PK/PD model)Gentamicin2 (100)An additive effect was described also when a high inoculum was tested.59
SA2005, USA2MRSA (50)in vitro (PK/PD model)Gentamicin2 (100)High inoculum tested.60
SA2005, USA40MRSA (50)in vitro (ET) (TK on 1 MSSA and 1 MRSA)GentamicinET: 1 (2.5); TK 1 (50)ET: 39 (97.5); TK 1 (50)14
E. faecalis, E. faecium40VRE (50)in vitro (ET)GentamicinET: 3 (7.5)ET: 37 (92.5)
SA2006, USA2GISA (100)in vitro (ET, TK)GentamicinET: 2 (100); TK: 1 (50)TK: 1 (50)15
E. faecium2006, Turkey42VRE (100)in vitro (ET)Gentamicin9 (21.4)33 (78.6)No distinction between additive and indifferent effect.16
SA2007, USA49VISA (12), VRSA (6), MRSA (64)in vitro (TK)Gentamicin34 (68)16 (32)61
SA2008, USA2in vitro (TK)Gentamicin2 (100)62
SA2009, USA2MRSA (100)in vitro (PK/PD model)Gentamicin2 (100)High inoculum tested. Both isolates exhibited biofilm formation. DAP monotherapy demonstrated significantly better activity than DAP in combination.63
SA2009, Spain3MRSA (100)in vitro (TK); in vivo (rabbit, IE)GentamicinTK: 3 (100)in vivo: 1 (100)In the in vivo study, equivalent human dosage of DAP 6 mg/kg was administered.64
E. faecalis2011, Switzerland1in vitro (TK), in vivo (Guinea pig, foreign body infection model)GentamicinTK: 1 (100); in vivo: 1 (100)In the in vivo study, equivalent human dosage of DAP 10 mg/kg was administered.65
E. faecalis2014, USA1in vitro (PK/PD model), in vivo (G. mellonella infection model)Gentamicinin vitro: 1 (100)in vivo: 1 (100)High inoculum tested. In the in vivo study, equivalent human dosage of DAP 6 and 10 mg/kg was administered (indifferent effect observed with both dosages).66
E. faecium1VRE (100)Gentamicinin vitro: 1 (100); in vivo: 1 (100)
Enterococcus spp.2015, USA4VRE (100)in vitro (TK)Gentamicin3 (75)1 (25)42
SE2015, Spain4MRSE (100)in vitro (PK/PD catheter model)Gentamicin3 (75)1 (25)57
SA2015, Australia8MRSA (50), VISA (25)in vitro (TK)Gentamicin4 (50)3 (37.5)1 (12.5)4 DNS isolates.67
SA2017, Turkey25MRSA (100)in vitro (CB)Gentamicin17 (68)7 (28)1 (4)68
SA2019, Taiwan100MRSA (100)in vitro (CB)Gentamicin5 (5)38 (38)57 (57)No distinction between indifferent and antagonistic effect.30
SA2022, India3in vitro (CB, TK)Tobramycin3 (100)69

CB, chequerboard; ET, Etest; IE, infective endocarditis; DAP, daptomycin; DNS, daptomycin non-susceptible; SA, S. aureus; SE, S. epidermidis; TK, time–kill assay.

S. aureus (MSSA, MRSA, GISA, VISA, VRSA) and S. epidermidis (MRSE)

The combination was tested against 238 isolates of S. aureus [37 MSSA, 188 MRSA, 2 GISA, 8 VISA, 3 vancomycin-resistant S. aureus (VRSA)] in vitro. The most frequent interaction was indifferent effect (99 isolates; 41.4%). One study evaluating daptomycin + gentamicin in a PK/PD model against two S. aureus isolates reported an antagonistic effect against both isolates.63 Only one isolate of MRSA was tested in vivo (indifferent effect).64

Four isolates of S. epidermidis were tested in vitro, resulting in indifferent effect in most cases (75%). The combination showed an antagonistic effect against one isolate.57

Enterococcus spp. (VSE, VRE)

Against 89 isolates of Enterococcus spp. (22 VSE, 67 VRE), the combination had a synergistic effect against 16 isolates (18%), additive effect against 1 isolate only, and indifferent effect against 72 (80.9%) isolates. When tested in vivo (three isolates; two Galleria mellonella infection models and one Guinea pig foreign body infection model), additive effect was observed against one isolate (foreign body infection model) and indifferent effect against the other two isolates.65,66

Glycopeptides

Five in vitro and one in vivo studies evaluated daptomycin in combination with glycopeptides (four studies included vancomycin, one study oritavancin, and one study dalbavancin) (Table 7).15,70–74

Table 7.

Studies evaluating daptomycin in combination with glycopeptides

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2006, USA2GISA (100)in vitro (ET, TK)VancomycinET: 1 (50)ET: 1 (500); TK: 2 (100)15
SA2016, Denmark1in vivo (mouse, implant-associated osteomyelitis)Vancomycin1 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.70
SA2017, Turkey30MRSA (100)in vitro (CB)Dalbavancin20 (67)10 (33)No distinction between additive and indifferent effect.71
E. faecium2019, USA4VRE (100)in vitro (TK)Oritavancin2 (50)2 (50)72
E. faecalis1Oritavancin1 (100)Synergistic, but non-bactericidal effect.
E. faecium2019, Turkey4VRE (100)in vitro (CB, TK)Vancomycin4 (100)73
SA2021, Turkey25MRSA (100)in vitro (ET)Vancomycin23 (92)2 (8)74
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2006, USA2GISA (100)in vitro (ET, TK)VancomycinET: 1 (50)ET: 1 (500); TK: 2 (100)15
SA2016, Denmark1in vivo (mouse, implant-associated osteomyelitis)Vancomycin1 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.70
SA2017, Turkey30MRSA (100)in vitro (CB)Dalbavancin20 (67)10 (33)No distinction between additive and indifferent effect.71
E. faecium2019, USA4VRE (100)in vitro (TK)Oritavancin2 (50)2 (50)72
E. faecalis1Oritavancin1 (100)Synergistic, but non-bactericidal effect.
E. faecium2019, Turkey4VRE (100)in vitro (CB, TK)Vancomycin4 (100)73
SA2021, Turkey25MRSA (100)in vitro (ET)Vancomycin23 (92)2 (8)74

CB, chequerboard; DAP, daptomycin; ET, Etest; SA, S. aureus; TK, time–kill assay.

Table 7.

Studies evaluating daptomycin in combination with glycopeptides

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2006, USA2GISA (100)in vitro (ET, TK)VancomycinET: 1 (50)ET: 1 (500); TK: 2 (100)15
SA2016, Denmark1in vivo (mouse, implant-associated osteomyelitis)Vancomycin1 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.70
SA2017, Turkey30MRSA (100)in vitro (CB)Dalbavancin20 (67)10 (33)No distinction between additive and indifferent effect.71
E. faecium2019, USA4VRE (100)in vitro (TK)Oritavancin2 (50)2 (50)72
E. faecalis1Oritavancin1 (100)Synergistic, but non-bactericidal effect.
E. faecium2019, Turkey4VRE (100)in vitro (CB, TK)Vancomycin4 (100)73
SA2021, Turkey25MRSA (100)in vitro (ET)Vancomycin23 (92)2 (8)74
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2006, USA2GISA (100)in vitro (ET, TK)VancomycinET: 1 (50)ET: 1 (500); TK: 2 (100)15
SA2016, Denmark1in vivo (mouse, implant-associated osteomyelitis)Vancomycin1 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.70
SA2017, Turkey30MRSA (100)in vitro (CB)Dalbavancin20 (67)10 (33)No distinction between additive and indifferent effect.71
E. faecium2019, USA4VRE (100)in vitro (TK)Oritavancin2 (50)2 (50)72
E. faecalis1Oritavancin1 (100)Synergistic, but non-bactericidal effect.
E. faecium2019, Turkey4VRE (100)in vitro (CB, TK)Vancomycin4 (100)73
SA2021, Turkey25MRSA (100)in vitro (ET)Vancomycin23 (92)2 (8)74

CB, chequerboard; DAP, daptomycin; ET, Etest; SA, S. aureus; TK, time–kill assay.

S. aureus (MSSA, MRSA, GISA)

Fifty-eight isolates of S. aureus (1 MSSA, 55 MRSA, 2 GISA) were tested. In vitro, the most frequent interaction was additive effect (23 isolates; 82.1%). Four S. aureus isolates showed indifferent effect. One isolate of MSSA was evaluated in vivo in a mouse implant-associated osteomyelitis and the combination had an indifferent effect compared with monotherapy.70 Daptomycin was tested in combination with dalbavancin, a novel glycopeptide, in one in vitro study, and synergistic effect was observed against 67% (20/30) of tested strains.71

Enterococcus spp. (VSE, VRE)

With regard to Enterococcus spp., nine isolates (eight vancomycin-resistant E. faecium, one vancomycin-susceptible E. faecalis) were evaluated in vitro. Daptomycin + vancomycin had a synergistic effect against four vancomycin-resistant E. faecium, and daptomycin + oritavancin was synergistic against one vancomycin-susceptible E. faecalis. Daptomycin + oritavancin showed antagonistic effect against two isolates of vancomycin-resistant E. faecium.

Oxazolidinones

Eight in vitro and one in vivo studies evaluating daptomycin in combination with oxazolidinones were included in the present systematic review (Table 8).15,19,30,70,71,75–78 Linezolid was evaluated in eight studies and tedizolid in one study only.

Table 8.

Studies evaluating daptomycin in combination with oxazolidinones

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2006, USA2GISA (100)in vitro (ET, TK)LinezolidET: 2 (100); TK: 2 (100)15
SA2010, USA2MRSA (100)in vitro (PK/PD model)Linezolid1 (50)1 (50)DNS isolates. High inoculum tested.19
SA2012, Spain3MRSA (100)in vitro (PK/PD model of biofilm)Linezolid2 (66.7)1 (33.3)Neither DAP nor linezolid alone were active against biofilm-embedded bacteria, while the combination was active against both planktonic and biofilm-embedded bacteria.75
E. faecalis2014, Switzerland3VRE (100)in vitro (CB, TK)Linezolid3 (100)76
E. faecium4VRE (100)Linezolid1 (33.3)1 (33.3)2 (33.3)
SA2015, USA2MRSA (100)in vitro (PK/PD biofilm model)Linezolid2 (100)77
SA2016, Denmark1in vivo (mouse, implant-associated osteomyelitis)Linezolid1 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.70
SA2017, Turkey30MRSA (100)in vitro (CB)Linezolid20 (67)10 (33)No distinction between additive and indifferent effect.71
SA2018, USA2MRSA (100)in vitro (PK/PD model)Tedizolid2 (100)78
SA2019, Taiwan100MRSA (100)in vitro (CB)Linezolid3 (3)74 (74)23 (23)No distinction between indifferent and antagonistic effect.30
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2006, USA2GISA (100)in vitro (ET, TK)LinezolidET: 2 (100); TK: 2 (100)15
SA2010, USA2MRSA (100)in vitro (PK/PD model)Linezolid1 (50)1 (50)DNS isolates. High inoculum tested.19
SA2012, Spain3MRSA (100)in vitro (PK/PD model of biofilm)Linezolid2 (66.7)1 (33.3)Neither DAP nor linezolid alone were active against biofilm-embedded bacteria, while the combination was active against both planktonic and biofilm-embedded bacteria.75
E. faecalis2014, Switzerland3VRE (100)in vitro (CB, TK)Linezolid3 (100)76
E. faecium4VRE (100)Linezolid1 (33.3)1 (33.3)2 (33.3)
SA2015, USA2MRSA (100)in vitro (PK/PD biofilm model)Linezolid2 (100)77
SA2016, Denmark1in vivo (mouse, implant-associated osteomyelitis)Linezolid1 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.70
SA2017, Turkey30MRSA (100)in vitro (CB)Linezolid20 (67)10 (33)No distinction between additive and indifferent effect.71
SA2018, USA2MRSA (100)in vitro (PK/PD model)Tedizolid2 (100)78
SA2019, Taiwan100MRSA (100)in vitro (CB)Linezolid3 (3)74 (74)23 (23)No distinction between indifferent and antagonistic effect.30

CB, chequerboard; ET, Etest; DAP, daptomycin; DNS, daptomycin non-susceptible; SA, S. aureus; TK, time–kill assay.

Table 8.

Studies evaluating daptomycin in combination with oxazolidinones

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2006, USA2GISA (100)in vitro (ET, TK)LinezolidET: 2 (100); TK: 2 (100)15
SA2010, USA2MRSA (100)in vitro (PK/PD model)Linezolid1 (50)1 (50)DNS isolates. High inoculum tested.19
SA2012, Spain3MRSA (100)in vitro (PK/PD model of biofilm)Linezolid2 (66.7)1 (33.3)Neither DAP nor linezolid alone were active against biofilm-embedded bacteria, while the combination was active against both planktonic and biofilm-embedded bacteria.75
E. faecalis2014, Switzerland3VRE (100)in vitro (CB, TK)Linezolid3 (100)76
E. faecium4VRE (100)Linezolid1 (33.3)1 (33.3)2 (33.3)
SA2015, USA2MRSA (100)in vitro (PK/PD biofilm model)Linezolid2 (100)77
SA2016, Denmark1in vivo (mouse, implant-associated osteomyelitis)Linezolid1 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.70
SA2017, Turkey30MRSA (100)in vitro (CB)Linezolid20 (67)10 (33)No distinction between additive and indifferent effect.71
SA2018, USA2MRSA (100)in vitro (PK/PD model)Tedizolid2 (100)78
SA2019, Taiwan100MRSA (100)in vitro (CB)Linezolid3 (3)74 (74)23 (23)No distinction between indifferent and antagonistic effect.30
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2006, USA2GISA (100)in vitro (ET, TK)LinezolidET: 2 (100); TK: 2 (100)15
SA2010, USA2MRSA (100)in vitro (PK/PD model)Linezolid1 (50)1 (50)DNS isolates. High inoculum tested.19
SA2012, Spain3MRSA (100)in vitro (PK/PD model of biofilm)Linezolid2 (66.7)1 (33.3)Neither DAP nor linezolid alone were active against biofilm-embedded bacteria, while the combination was active against both planktonic and biofilm-embedded bacteria.75
E. faecalis2014, Switzerland3VRE (100)in vitro (CB, TK)Linezolid3 (100)76
E. faecium4VRE (100)Linezolid1 (33.3)1 (33.3)2 (33.3)
SA2015, USA2MRSA (100)in vitro (PK/PD biofilm model)Linezolid2 (100)77
SA2016, Denmark1in vivo (mouse, implant-associated osteomyelitis)Linezolid1 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.70
SA2017, Turkey30MRSA (100)in vitro (CB)Linezolid20 (67)10 (33)No distinction between additive and indifferent effect.71
SA2018, USA2MRSA (100)in vitro (PK/PD model)Tedizolid2 (100)78
SA2019, Taiwan100MRSA (100)in vitro (CB)Linezolid3 (3)74 (74)23 (23)No distinction between indifferent and antagonistic effect.30

CB, chequerboard; ET, Etest; DAP, daptomycin; DNS, daptomycin non-susceptible; SA, S. aureus; TK, time–kill assay.

S. aureus (MSSA, MRSA, GISA)

One hundred and forty-two S. aureus (1 MSSA, 139 MRSA, 2 GISA) isolates were tested. With regard to S. aureus, in vitro the combination showed an additive effect against most isolates (75; 53.2%). Synergism occurred against 26 isolates (18.4%). Indifferent effect was reported against 36 isolates (25.6%) and antagonism against 4 isolates (2.8%). One study evaluated daptomycin + linezolid against one isolate of S. aureus in a mouse implant-associated osteomyelitis, exhibiting indifferent effect.70

Enterococcus spp. (VRE)

When the combination was tested against Enterococcus spp. (seven isolates, all VRE), synergism was observed against four isolates (57.1%), while additive and indifferent effect was described against one (14.3%) and two (28.6%) isolates, respectively.

Rifampicin

We included 31 studies evaluating daptomycin in combination with rifampicin (Table 9).13,15–17,30,33,42,55,56,58,61,63,64,66,68,70,79–93

Table 9.

Studies evaluating daptomycin in combination with rifampicin

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Synergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2003, USA1MRSA (100)in vivo (rat, aortic valve IE)1 (100)Equivalent human dosage of DAP 4–6 mg/kg was administered.79
E. faecium2004, USA19VRE (100)in vitro (ET)13 (68.4)6 (31.6)13
E. faecium2005, USA24VRE (100)in vitro (ET, TK)ET: 21 (87.5); TK: 18 (75)ET: 2 (8.3); TK: 2 (8.3)ET: 1 (4.2); TK: 4 (16.7)Linezolid-resistant isolates.80
SA2006, USA2GISA (100)in vitro (ET, TK)ET: 2 (100); TK: 2 (100)15
E. faecium2006, Turkey42VRE (100)in vitro (ET)24 (57.1)18 (42.9)No distinction between additive and indifferent effect.16
SA2007, USA50VISA (12), VRSA (6), MRSA (64)in vitro (TK)1 (2)49 (98)61
SA2008, Taiwan1VISA (100)in vitro (TK)1 (100)Additive effect was observed at 8 h, while antagonism at 24 h.17
SA2008, USA6MRSA (100)in vitro (TK)5 (83.3)1 (16.7)All isolates were rifampicin-resistant.81
SA2009, Switzerland1MRSA (100)in vivo (Guinea pig foreign body infection model)1 (100)The synergistic effect was observed in both planktonic and adherent bacteria. Equivalent human dosage of DAP 6 mg/kg was administered.82
SA2009, USA2MRSA (100)in vitro (PK/PD model)1 (50)1 (50)High inoculum tested. Both MRSA isolates exhibited biofilm formation.63
SA2009, Spain3MRSA (100)in vitro (TK); in vivo (rabbit, IE)TK: 2 (66.6)TK: 1 (33.3); in vivo: 1 (100)64
SA2010, USA1MRSA (100)in vitro (PK/PD model)1 (100)55
SA2010, France1MRSA (100)in vivo (rabbit, acute osteomyelitis)1 (100)Equivalent human dosage of DAP 6 mg/kg was administered.83
SA2010, Italy6in vitro (CB); in vivo (rat, vascular graft infection)CB: 6 (100); in vivo: 1 (100)In the in vivo study, the combination was tested with DAP administered intraperitoneally, in a rat implanted with a rifampicin-soaked graft. In the in vivo study, equivalent human dosage of DAP 6 mg/kg was administered.84
SA2010, Spain1MRSA (100)in vitro (TK), in vivo (rat, foreign body infection model)TK: 1 (100); in vivo: 1 (100)In the in vivo study, equivalent human dosages of DAP 6 and 10 mg/kg were administered.85
E. faecium2013, USA24VRE (100)in vitro (ET)21 (88)3 (12)No distinction between additive and indifferent effect.86
SA2013, China8MRSA (100)in vitro (biofilm model)2 (25)6 (75)87
SA2014, Switzerland1MRSA (100)in vivo (Guinea pig, foreign body infection model)1 (100)Equivalent human dosage of DAP 10 mg/kg was administered.88
E. faecium2014, Sweden3in vitro (TK)3 (100)Activity tested against biofilms formed on beads of bone cement.89
E. faecalis2014, USA1in vitro (PK/PD model), in vivo (G. mellonella infection model)in vitro: 1 (100)in vivo: 1 (100)High inoculum tested. In the in vivo study, equivalent human dosages of DAP 6 and 10 mg/kg were administered.66
E. faecium1VRE (100)in vitro: 1 (100); in vivo: 1 (100)
SA2014, USA2MRSA (50), hVISA (50)in vitro (PK/PD biofilm model)2 (100)56
SE1MRSE (100)1 (100)
Enterococcus spp.2015, USA7VRE (100)in vitro (TK)7 (100)42
SA2015, Spain1in vitro (TK); in vivo (rat, tissue-cage infection model)in vivo: 1 (100)in vitro: 1 (100)In the in vivo study, equivalent human dosage of DAP 8–10 mg/kg was administered.90
SA2016, Denmark1in vivo (mouse, implant-associated osteomyelitis)1 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.70
SA2016, Germany58MRSA (84.5)in vitro (CB)1 (1.7)57 (98.3)6 DNS isolates.91
SA2016, Spain2MRSA (50)in vivo (rat, foreign body infection)2 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.58
SA2017, Turkey25MRSA (100)in vitro (CB)3 (12)7 (28)15 (60)68
E. faecalis2019, China4in vitro (TK)4 (100)92
SA2019, Taiwan100MRSA (100)in vitro (CB)1 (1)51 (51)48 (48)No distinction between indifferent and antagonistic effect.30
SE2020, France1MRSE (100)in vivo (rat, osteitis)1 (100)Equivalent human dosage of DAP 10 mg/kg was administered.93
E. faecalis2021, USA2VRE (100)in vitro (TK)1 (50)1 (50)33
E. faecium21 (50)1 (50)
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Synergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2003, USA1MRSA (100)in vivo (rat, aortic valve IE)1 (100)Equivalent human dosage of DAP 4–6 mg/kg was administered.79
E. faecium2004, USA19VRE (100)in vitro (ET)13 (68.4)6 (31.6)13
E. faecium2005, USA24VRE (100)in vitro (ET, TK)ET: 21 (87.5); TK: 18 (75)ET: 2 (8.3); TK: 2 (8.3)ET: 1 (4.2); TK: 4 (16.7)Linezolid-resistant isolates.80
SA2006, USA2GISA (100)in vitro (ET, TK)ET: 2 (100); TK: 2 (100)15
E. faecium2006, Turkey42VRE (100)in vitro (ET)24 (57.1)18 (42.9)No distinction between additive and indifferent effect.16
SA2007, USA50VISA (12), VRSA (6), MRSA (64)in vitro (TK)1 (2)49 (98)61
SA2008, Taiwan1VISA (100)in vitro (TK)1 (100)Additive effect was observed at 8 h, while antagonism at 24 h.17
SA2008, USA6MRSA (100)in vitro (TK)5 (83.3)1 (16.7)All isolates were rifampicin-resistant.81
SA2009, Switzerland1MRSA (100)in vivo (Guinea pig foreign body infection model)1 (100)The synergistic effect was observed in both planktonic and adherent bacteria. Equivalent human dosage of DAP 6 mg/kg was administered.82
SA2009, USA2MRSA (100)in vitro (PK/PD model)1 (50)1 (50)High inoculum tested. Both MRSA isolates exhibited biofilm formation.63
SA2009, Spain3MRSA (100)in vitro (TK); in vivo (rabbit, IE)TK: 2 (66.6)TK: 1 (33.3); in vivo: 1 (100)64
SA2010, USA1MRSA (100)in vitro (PK/PD model)1 (100)55
SA2010, France1MRSA (100)in vivo (rabbit, acute osteomyelitis)1 (100)Equivalent human dosage of DAP 6 mg/kg was administered.83
SA2010, Italy6in vitro (CB); in vivo (rat, vascular graft infection)CB: 6 (100); in vivo: 1 (100)In the in vivo study, the combination was tested with DAP administered intraperitoneally, in a rat implanted with a rifampicin-soaked graft. In the in vivo study, equivalent human dosage of DAP 6 mg/kg was administered.84
SA2010, Spain1MRSA (100)in vitro (TK), in vivo (rat, foreign body infection model)TK: 1 (100); in vivo: 1 (100)In the in vivo study, equivalent human dosages of DAP 6 and 10 mg/kg were administered.85
E. faecium2013, USA24VRE (100)in vitro (ET)21 (88)3 (12)No distinction between additive and indifferent effect.86
SA2013, China8MRSA (100)in vitro (biofilm model)2 (25)6 (75)87
SA2014, Switzerland1MRSA (100)in vivo (Guinea pig, foreign body infection model)1 (100)Equivalent human dosage of DAP 10 mg/kg was administered.88
E. faecium2014, Sweden3in vitro (TK)3 (100)Activity tested against biofilms formed on beads of bone cement.89
E. faecalis2014, USA1in vitro (PK/PD model), in vivo (G. mellonella infection model)in vitro: 1 (100)in vivo: 1 (100)High inoculum tested. In the in vivo study, equivalent human dosages of DAP 6 and 10 mg/kg were administered.66
E. faecium1VRE (100)in vitro: 1 (100); in vivo: 1 (100)
SA2014, USA2MRSA (50), hVISA (50)in vitro (PK/PD biofilm model)2 (100)56
SE1MRSE (100)1 (100)
Enterococcus spp.2015, USA7VRE (100)in vitro (TK)7 (100)42
SA2015, Spain1in vitro (TK); in vivo (rat, tissue-cage infection model)in vivo: 1 (100)in vitro: 1 (100)In the in vivo study, equivalent human dosage of DAP 8–10 mg/kg was administered.90
SA2016, Denmark1in vivo (mouse, implant-associated osteomyelitis)1 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.70
SA2016, Germany58MRSA (84.5)in vitro (CB)1 (1.7)57 (98.3)6 DNS isolates.91
SA2016, Spain2MRSA (50)in vivo (rat, foreign body infection)2 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.58
SA2017, Turkey25MRSA (100)in vitro (CB)3 (12)7 (28)15 (60)68
E. faecalis2019, China4in vitro (TK)4 (100)92
SA2019, Taiwan100MRSA (100)in vitro (CB)1 (1)51 (51)48 (48)No distinction between indifferent and antagonistic effect.30
SE2020, France1MRSE (100)in vivo (rat, osteitis)1 (100)Equivalent human dosage of DAP 10 mg/kg was administered.93
E. faecalis2021, USA2VRE (100)in vitro (TK)1 (50)1 (50)33
E. faecium21 (50)1 (50)

CB, chequerboard; DAP, daptomycin; ET, Etest; IE, infective endocarditis; DNS, daptomycin non-susceptible; SA, S. aureus; SE, S. epidermidis; TK, time–kill assay.

Table 9.

Studies evaluating daptomycin in combination with rifampicin

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Synergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2003, USA1MRSA (100)in vivo (rat, aortic valve IE)1 (100)Equivalent human dosage of DAP 4–6 mg/kg was administered.79
E. faecium2004, USA19VRE (100)in vitro (ET)13 (68.4)6 (31.6)13
E. faecium2005, USA24VRE (100)in vitro (ET, TK)ET: 21 (87.5); TK: 18 (75)ET: 2 (8.3); TK: 2 (8.3)ET: 1 (4.2); TK: 4 (16.7)Linezolid-resistant isolates.80
SA2006, USA2GISA (100)in vitro (ET, TK)ET: 2 (100); TK: 2 (100)15
E. faecium2006, Turkey42VRE (100)in vitro (ET)24 (57.1)18 (42.9)No distinction between additive and indifferent effect.16
SA2007, USA50VISA (12), VRSA (6), MRSA (64)in vitro (TK)1 (2)49 (98)61
SA2008, Taiwan1VISA (100)in vitro (TK)1 (100)Additive effect was observed at 8 h, while antagonism at 24 h.17
SA2008, USA6MRSA (100)in vitro (TK)5 (83.3)1 (16.7)All isolates were rifampicin-resistant.81
SA2009, Switzerland1MRSA (100)in vivo (Guinea pig foreign body infection model)1 (100)The synergistic effect was observed in both planktonic and adherent bacteria. Equivalent human dosage of DAP 6 mg/kg was administered.82
SA2009, USA2MRSA (100)in vitro (PK/PD model)1 (50)1 (50)High inoculum tested. Both MRSA isolates exhibited biofilm formation.63
SA2009, Spain3MRSA (100)in vitro (TK); in vivo (rabbit, IE)TK: 2 (66.6)TK: 1 (33.3); in vivo: 1 (100)64
SA2010, USA1MRSA (100)in vitro (PK/PD model)1 (100)55
SA2010, France1MRSA (100)in vivo (rabbit, acute osteomyelitis)1 (100)Equivalent human dosage of DAP 6 mg/kg was administered.83
SA2010, Italy6in vitro (CB); in vivo (rat, vascular graft infection)CB: 6 (100); in vivo: 1 (100)In the in vivo study, the combination was tested with DAP administered intraperitoneally, in a rat implanted with a rifampicin-soaked graft. In the in vivo study, equivalent human dosage of DAP 6 mg/kg was administered.84
SA2010, Spain1MRSA (100)in vitro (TK), in vivo (rat, foreign body infection model)TK: 1 (100); in vivo: 1 (100)In the in vivo study, equivalent human dosages of DAP 6 and 10 mg/kg were administered.85
E. faecium2013, USA24VRE (100)in vitro (ET)21 (88)3 (12)No distinction between additive and indifferent effect.86
SA2013, China8MRSA (100)in vitro (biofilm model)2 (25)6 (75)87
SA2014, Switzerland1MRSA (100)in vivo (Guinea pig, foreign body infection model)1 (100)Equivalent human dosage of DAP 10 mg/kg was administered.88
E. faecium2014, Sweden3in vitro (TK)3 (100)Activity tested against biofilms formed on beads of bone cement.89
E. faecalis2014, USA1in vitro (PK/PD model), in vivo (G. mellonella infection model)in vitro: 1 (100)in vivo: 1 (100)High inoculum tested. In the in vivo study, equivalent human dosages of DAP 6 and 10 mg/kg were administered.66
E. faecium1VRE (100)in vitro: 1 (100); in vivo: 1 (100)
SA2014, USA2MRSA (50), hVISA (50)in vitro (PK/PD biofilm model)2 (100)56
SE1MRSE (100)1 (100)
Enterococcus spp.2015, USA7VRE (100)in vitro (TK)7 (100)42
SA2015, Spain1in vitro (TK); in vivo (rat, tissue-cage infection model)in vivo: 1 (100)in vitro: 1 (100)In the in vivo study, equivalent human dosage of DAP 8–10 mg/kg was administered.90
SA2016, Denmark1in vivo (mouse, implant-associated osteomyelitis)1 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.70
SA2016, Germany58MRSA (84.5)in vitro (CB)1 (1.7)57 (98.3)6 DNS isolates.91
SA2016, Spain2MRSA (50)in vivo (rat, foreign body infection)2 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.58
SA2017, Turkey25MRSA (100)in vitro (CB)3 (12)7 (28)15 (60)68
E. faecalis2019, China4in vitro (TK)4 (100)92
SA2019, Taiwan100MRSA (100)in vitro (CB)1 (1)51 (51)48 (48)No distinction between indifferent and antagonistic effect.30
SE2020, France1MRSE (100)in vivo (rat, osteitis)1 (100)Equivalent human dosage of DAP 10 mg/kg was administered.93
E. faecalis2021, USA2VRE (100)in vitro (TK)1 (50)1 (50)33
E. faecium21 (50)1 (50)
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Synergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2003, USA1MRSA (100)in vivo (rat, aortic valve IE)1 (100)Equivalent human dosage of DAP 4–6 mg/kg was administered.79
E. faecium2004, USA19VRE (100)in vitro (ET)13 (68.4)6 (31.6)13
E. faecium2005, USA24VRE (100)in vitro (ET, TK)ET: 21 (87.5); TK: 18 (75)ET: 2 (8.3); TK: 2 (8.3)ET: 1 (4.2); TK: 4 (16.7)Linezolid-resistant isolates.80
SA2006, USA2GISA (100)in vitro (ET, TK)ET: 2 (100); TK: 2 (100)15
E. faecium2006, Turkey42VRE (100)in vitro (ET)24 (57.1)18 (42.9)No distinction between additive and indifferent effect.16
SA2007, USA50VISA (12), VRSA (6), MRSA (64)in vitro (TK)1 (2)49 (98)61
SA2008, Taiwan1VISA (100)in vitro (TK)1 (100)Additive effect was observed at 8 h, while antagonism at 24 h.17
SA2008, USA6MRSA (100)in vitro (TK)5 (83.3)1 (16.7)All isolates were rifampicin-resistant.81
SA2009, Switzerland1MRSA (100)in vivo (Guinea pig foreign body infection model)1 (100)The synergistic effect was observed in both planktonic and adherent bacteria. Equivalent human dosage of DAP 6 mg/kg was administered.82
SA2009, USA2MRSA (100)in vitro (PK/PD model)1 (50)1 (50)High inoculum tested. Both MRSA isolates exhibited biofilm formation.63
SA2009, Spain3MRSA (100)in vitro (TK); in vivo (rabbit, IE)TK: 2 (66.6)TK: 1 (33.3); in vivo: 1 (100)64
SA2010, USA1MRSA (100)in vitro (PK/PD model)1 (100)55
SA2010, France1MRSA (100)in vivo (rabbit, acute osteomyelitis)1 (100)Equivalent human dosage of DAP 6 mg/kg was administered.83
SA2010, Italy6in vitro (CB); in vivo (rat, vascular graft infection)CB: 6 (100); in vivo: 1 (100)In the in vivo study, the combination was tested with DAP administered intraperitoneally, in a rat implanted with a rifampicin-soaked graft. In the in vivo study, equivalent human dosage of DAP 6 mg/kg was administered.84
SA2010, Spain1MRSA (100)in vitro (TK), in vivo (rat, foreign body infection model)TK: 1 (100); in vivo: 1 (100)In the in vivo study, equivalent human dosages of DAP 6 and 10 mg/kg were administered.85
E. faecium2013, USA24VRE (100)in vitro (ET)21 (88)3 (12)No distinction between additive and indifferent effect.86
SA2013, China8MRSA (100)in vitro (biofilm model)2 (25)6 (75)87
SA2014, Switzerland1MRSA (100)in vivo (Guinea pig, foreign body infection model)1 (100)Equivalent human dosage of DAP 10 mg/kg was administered.88
E. faecium2014, Sweden3in vitro (TK)3 (100)Activity tested against biofilms formed on beads of bone cement.89
E. faecalis2014, USA1in vitro (PK/PD model), in vivo (G. mellonella infection model)in vitro: 1 (100)in vivo: 1 (100)High inoculum tested. In the in vivo study, equivalent human dosages of DAP 6 and 10 mg/kg were administered.66
E. faecium1VRE (100)in vitro: 1 (100); in vivo: 1 (100)
SA2014, USA2MRSA (50), hVISA (50)in vitro (PK/PD biofilm model)2 (100)56
SE1MRSE (100)1 (100)
Enterococcus spp.2015, USA7VRE (100)in vitro (TK)7 (100)42
SA2015, Spain1in vitro (TK); in vivo (rat, tissue-cage infection model)in vivo: 1 (100)in vitro: 1 (100)In the in vivo study, equivalent human dosage of DAP 8–10 mg/kg was administered.90
SA2016, Denmark1in vivo (mouse, implant-associated osteomyelitis)1 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.70
SA2016, Germany58MRSA (84.5)in vitro (CB)1 (1.7)57 (98.3)6 DNS isolates.91
SA2016, Spain2MRSA (50)in vivo (rat, foreign body infection)2 (100)Equivalent human dosage of DAP 8–10 mg/kg was administered.58
SA2017, Turkey25MRSA (100)in vitro (CB)3 (12)7 (28)15 (60)68
E. faecalis2019, China4in vitro (TK)4 (100)92
SA2019, Taiwan100MRSA (100)in vitro (CB)1 (1)51 (51)48 (48)No distinction between indifferent and antagonistic effect.30
SE2020, France1MRSE (100)in vivo (rat, osteitis)1 (100)Equivalent human dosage of DAP 10 mg/kg was administered.93
E. faecalis2021, USA2VRE (100)in vitro (TK)1 (50)1 (50)33
E. faecium21 (50)1 (50)

CB, chequerboard; DAP, daptomycin; ET, Etest; IE, infective endocarditis; DNS, daptomycin non-susceptible; SA, S. aureus; SE, S. epidermidis; TK, time–kill assay.

S. aureus (MSSA, MRSA, GISA, hVISA, VRSA) and S. epidermidis (MRSE)

Twenty-one studies (11 in vitro, 4 in vitro and in vivo, 6 in vivo) evaluated the combination against S. aureus (273 isolates; 27 MSSA, 233 MRSA, 2 GISA, 1 hVISA, 7 VISA, 3 VRSA). Two hundred and sixty-six isolates were tested in vitro. The combination exerted a synergistic effect against only 13 (4.9%) isolates, while the most frequent interaction was indifferent effect, which was observed against 138 (51.9%) isolates. Three studies (two time–kill assay, one PK/PD model) reported an antagonistic effect of the combination against the tested strains.63,64,81 Daptomycin + rifampicin was tested in vivo against 11 isolates, and synergistic effect was observed against 6 isolates (54.5%), thus suggesting a higher synergism rate than observed in vitro. One study reported an antagonistic effect of the combination against one isolate of S. aureus in a rabbit infective endocarditis model.64

With regard to S. epidermidis, one isolate was tested in vitro (PK/PD biofilm model, synergistic effect), and one isolate was tested in vivo (osteitis model in rats, additive effect).56,93

Enterococcus spp. (VSE, VRE)

A total of 129 isolates of Enterococcus spp. (8 VSE, 121 VRE) were tested in eight in vitro studies and one in vitro and in vivo study. Synergism was observed in most cases (81 isolates; 62.8%), while additive and indifferent effects occurred for 10.9% and 25.6% of tested strains, respectively. Antagonism was observed in vitro against one isolate of E. faecium evaluated in a PK/PD model.66 Only two isolates (one E. faecalis, one E. faecium; G. mellonella infection model) were tested in vivo and the combination exerted an antagonistic effect against both isolates.66

Fosfomycin

We included 14 studies evaluating daptomycin in combination with fosfomycin in the present systematic review (Table 10).29,30,32,33,68,85,88,92,94–99

Table 10.

Studies evaluating daptomycin in combination with fosfomycin

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Synergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2011, Austria1MRSA (100)in vivo (rat, acute osteomyelitis)1 (100)High inoculum tested. Equivalent human dosage of DAP 6 mg/kg was tested.94
SA2012, Spain14MRSA (35.7); GISA (14.3)in vitro (TK)11 (79)3 (21)95
SA2013, Spain1MRSA (100)in vitro (TK); in vivo (rat, foreign body infection)in vivo: 1 (100)TK: 1 (100)In the in vivo study, equivalent human dosages of DAP 6 and 10 mg/kg were administered.85
E. faecium2013, USA4VRE (100)in vitro (TK)4 (100)96
SA2014, Switzerland1MRSA (100)in vivo (Guinea pig, foreign body infection model)1 (100)Equivalent human dosage of DAP 10 mg/kg was administered.88
SA2015, Austria1MRSA (100)in vivo (rat, implant-associated osteomyelitis)1 (100)High inoculum tested. Equivalent human dosage of DAP 6 mg/kg was administered.97
E. faecalis2016, USA2VRE (100)in vitro (PK/PD model)2 (100)High inoculum tested.98
E. faecium21 (50)1 (50)High inoculum tested. Isogenic pair of DAP-susceptible and DNS isolates.
SA2017, Turkey25MRSA (100)in vitro (CB)25 (100)68
SA2018, Spain5MRSA (100)in vitro (TK); in vivo (rabbit, IE, 1 isolate)TK: 5 (100)in vivo: 1 (100)Synergism was confirmed also when high inoculum was tested. In the in vivo study, equivalent human dosages of DAP 6 and 10 mg/kg were administered.29
E. faecalis2019, China4in vitro (TK)4 (100)92
SA2019, Taiwan100MRSA (100)in vitro (CB)37 (37)44 (44)19 (19)No distinction between indifferent and antagonistic effect.30
SA2020, Spain5in vitro (TK), in vivo (rabbit, IE, 1 isolate)TK: 5 (100); in vivo: 1 (100)Standard and high inoculum tested. In the in vivo study, equivalent human dosage of DAP 6 mg/kg was administered.32
E. faecalis2020, China6in vitro (TK)2 (33.3)1 (16.7)3 (50)5 linezolid-resistant isolates.99
E. faecalis2021, USA2VRE (100)in vitro (TK)1 (50)1 (50)33
E. faecium21 (50)1 (50)
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Synergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2011, Austria1MRSA (100)in vivo (rat, acute osteomyelitis)1 (100)High inoculum tested. Equivalent human dosage of DAP 6 mg/kg was tested.94
SA2012, Spain14MRSA (35.7); GISA (14.3)in vitro (TK)11 (79)3 (21)95
SA2013, Spain1MRSA (100)in vitro (TK); in vivo (rat, foreign body infection)in vivo: 1 (100)TK: 1 (100)In the in vivo study, equivalent human dosages of DAP 6 and 10 mg/kg were administered.85
E. faecium2013, USA4VRE (100)in vitro (TK)4 (100)96
SA2014, Switzerland1MRSA (100)in vivo (Guinea pig, foreign body infection model)1 (100)Equivalent human dosage of DAP 10 mg/kg was administered.88
SA2015, Austria1MRSA (100)in vivo (rat, implant-associated osteomyelitis)1 (100)High inoculum tested. Equivalent human dosage of DAP 6 mg/kg was administered.97
E. faecalis2016, USA2VRE (100)in vitro (PK/PD model)2 (100)High inoculum tested.98
E. faecium21 (50)1 (50)High inoculum tested. Isogenic pair of DAP-susceptible and DNS isolates.
SA2017, Turkey25MRSA (100)in vitro (CB)25 (100)68
SA2018, Spain5MRSA (100)in vitro (TK); in vivo (rabbit, IE, 1 isolate)TK: 5 (100)in vivo: 1 (100)Synergism was confirmed also when high inoculum was tested. In the in vivo study, equivalent human dosages of DAP 6 and 10 mg/kg were administered.29
E. faecalis2019, China4in vitro (TK)4 (100)92
SA2019, Taiwan100MRSA (100)in vitro (CB)37 (37)44 (44)19 (19)No distinction between indifferent and antagonistic effect.30
SA2020, Spain5in vitro (TK), in vivo (rabbit, IE, 1 isolate)TK: 5 (100); in vivo: 1 (100)Standard and high inoculum tested. In the in vivo study, equivalent human dosage of DAP 6 mg/kg was administered.32
E. faecalis2020, China6in vitro (TK)2 (33.3)1 (16.7)3 (50)5 linezolid-resistant isolates.99
E. faecalis2021, USA2VRE (100)in vitro (TK)1 (50)1 (50)33
E. faecium21 (50)1 (50)

CB, chequerboard; IE, infective endocarditis; DAP, daptomycin; DNS, daptomycin non-susceptible; SA, S. aureus; TK, time–kill assay.

Table 10.

Studies evaluating daptomycin in combination with fosfomycin

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Synergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2011, Austria1MRSA (100)in vivo (rat, acute osteomyelitis)1 (100)High inoculum tested. Equivalent human dosage of DAP 6 mg/kg was tested.94
SA2012, Spain14MRSA (35.7); GISA (14.3)in vitro (TK)11 (79)3 (21)95
SA2013, Spain1MRSA (100)in vitro (TK); in vivo (rat, foreign body infection)in vivo: 1 (100)TK: 1 (100)In the in vivo study, equivalent human dosages of DAP 6 and 10 mg/kg were administered.85
E. faecium2013, USA4VRE (100)in vitro (TK)4 (100)96
SA2014, Switzerland1MRSA (100)in vivo (Guinea pig, foreign body infection model)1 (100)Equivalent human dosage of DAP 10 mg/kg was administered.88
SA2015, Austria1MRSA (100)in vivo (rat, implant-associated osteomyelitis)1 (100)High inoculum tested. Equivalent human dosage of DAP 6 mg/kg was administered.97
E. faecalis2016, USA2VRE (100)in vitro (PK/PD model)2 (100)High inoculum tested.98
E. faecium21 (50)1 (50)High inoculum tested. Isogenic pair of DAP-susceptible and DNS isolates.
SA2017, Turkey25MRSA (100)in vitro (CB)25 (100)68
SA2018, Spain5MRSA (100)in vitro (TK); in vivo (rabbit, IE, 1 isolate)TK: 5 (100)in vivo: 1 (100)Synergism was confirmed also when high inoculum was tested. In the in vivo study, equivalent human dosages of DAP 6 and 10 mg/kg were administered.29
E. faecalis2019, China4in vitro (TK)4 (100)92
SA2019, Taiwan100MRSA (100)in vitro (CB)37 (37)44 (44)19 (19)No distinction between indifferent and antagonistic effect.30
SA2020, Spain5in vitro (TK), in vivo (rabbit, IE, 1 isolate)TK: 5 (100); in vivo: 1 (100)Standard and high inoculum tested. In the in vivo study, equivalent human dosage of DAP 6 mg/kg was administered.32
E. faecalis2020, China6in vitro (TK)2 (33.3)1 (16.7)3 (50)5 linezolid-resistant isolates.99
E. faecalis2021, USA2VRE (100)in vitro (TK)1 (50)1 (50)33
E. faecium21 (50)1 (50)
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Synergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2011, Austria1MRSA (100)in vivo (rat, acute osteomyelitis)1 (100)High inoculum tested. Equivalent human dosage of DAP 6 mg/kg was tested.94
SA2012, Spain14MRSA (35.7); GISA (14.3)in vitro (TK)11 (79)3 (21)95
SA2013, Spain1MRSA (100)in vitro (TK); in vivo (rat, foreign body infection)in vivo: 1 (100)TK: 1 (100)In the in vivo study, equivalent human dosages of DAP 6 and 10 mg/kg were administered.85
E. faecium2013, USA4VRE (100)in vitro (TK)4 (100)96
SA2014, Switzerland1MRSA (100)in vivo (Guinea pig, foreign body infection model)1 (100)Equivalent human dosage of DAP 10 mg/kg was administered.88
SA2015, Austria1MRSA (100)in vivo (rat, implant-associated osteomyelitis)1 (100)High inoculum tested. Equivalent human dosage of DAP 6 mg/kg was administered.97
E. faecalis2016, USA2VRE (100)in vitro (PK/PD model)2 (100)High inoculum tested.98
E. faecium21 (50)1 (50)High inoculum tested. Isogenic pair of DAP-susceptible and DNS isolates.
SA2017, Turkey25MRSA (100)in vitro (CB)25 (100)68
SA2018, Spain5MRSA (100)in vitro (TK); in vivo (rabbit, IE, 1 isolate)TK: 5 (100)in vivo: 1 (100)Synergism was confirmed also when high inoculum was tested. In the in vivo study, equivalent human dosages of DAP 6 and 10 mg/kg were administered.29
E. faecalis2019, China4in vitro (TK)4 (100)92
SA2019, Taiwan100MRSA (100)in vitro (CB)37 (37)44 (44)19 (19)No distinction between indifferent and antagonistic effect.30
SA2020, Spain5in vitro (TK), in vivo (rabbit, IE, 1 isolate)TK: 5 (100); in vivo: 1 (100)Standard and high inoculum tested. In the in vivo study, equivalent human dosage of DAP 6 mg/kg was administered.32
E. faecalis2020, China6in vitro (TK)2 (33.3)1 (16.7)3 (50)5 linezolid-resistant isolates.99
E. faecalis2021, USA2VRE (100)in vitro (TK)1 (50)1 (50)33
E. faecium21 (50)1 (50)

CB, chequerboard; IE, infective endocarditis; DAP, daptomycin; DNS, daptomycin non-susceptible; SA, S. aureus; TK, time–kill assay.

S. aureus (MSSA, MRSA, GISA)

A total of 153 isolates of S. aureus (12 MSSA, 139 MRSA, 2 GISA) were tested (147 in vitro, 3 in vitro and in vivo, 3 in vivo). In vitro, the combination was synergistic against most isolates (55.4%). Additive effect accounted for 31.3% of interactions, while indifferent effect accounted for 13.3% of interactions. In vivo (two infective endocarditis, two osteomyelitis, two foreign body infection model), the combination acted synergistically against four isolates (66.7%), while indifferent effect was observed against two isolates (33.3%; one infective endocarditis and one osteomyelitis model).

Enterococcus spp. (VSE, VRE)

Five in vitro studies evaluated 22 isolates of Enterococcus spp. (14 E. faecalis, 8 E. faecium). The most frequent interaction was synergism (14 isolates; 63.6%). The combination had an additive effect against four isolates (18.2%) and an indifferent effect against four isolates (18.2%). No antagonistic effect was observed, either in vitro or in vivo.

Miscellanea

Table 11 summarizes the studies evaluating daptomycin in combination with other antimicrobial agents not belonging to the previously discussed antibiotic classes. The following daptomycin combinations were evaluated in three or fewer studies: aztreonam (one study),14 quinolones (three studies),69,90,100 trimethoprim/sulfamethoxazole (three studies),19,101,102 tigecycline (two studies),42,103 fusidic acid (one study)68 and quinupristin/dalfopristin (one study).15 Due to the limited number of studies and tested strains, it appears inappropriate to reach any conclusion.

Table 11.

Studies evaluating daptomycin in combination with other antimicrobial agents (miscellanea)

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2005, USA40MRSA (50)in vitro (ET) (TK on 1 MRSA)AztreonamET: 2 (5); TK: 1 (100)ET: 38 (95)14
E. faecalis, E. faecium40 (21 E. faecalis, 9 E. faecium)VRE (50)in vitro (ET)ET: 40 (100)
SA2017, Turkey25MRSA (100)in vitro (CB)Fusidic acid4 (16)7 (28)14 (56)68
SA2006, USA2GISA (100)in vitro (ET, TK)Quinupristin/dalfopristinET: 2 (100); TK: 2 (100)15
SA2010, USA2MRSA (100)in vitro (PD model)TMP/SMX2 (100)DNS isolates. High inoculum tested.19
SA2012, Spain4MRSA (50), VISA (25), hVISA (25)in vitro (PK/PD model)TMP/SMX4 (100)High inoculum tested.101
SA2015, USA17MRSA (100)in vitro (TK)TMP/SMX17 (100)6 DNS isolates.102
SA2012, Italy13MRSA (92.3, 1 MSSA for in vivo study)in vitro (CB, TK), in vivo (mouse, wound infection model)Tigecyclinein vitro: 9 (75); in vivo: 1 (100)in vitro: 3 (25)No distinction between additive and indifferent effect. In the in vivo study, equivalent human dosage of DAP 7 mg/kg was administered.103
Enterococcus spp.12 E. faecalis, 12 E. faecium, 12 E. durans, 1 E. faecalis ATCC for in vivo studyin vitro: 12 (100) E. faecalis, 7 (58) E. faecium, 5 (42) E. durans; in vivo: 1 (100)in vitro: 5 (42) E. faecium, 7 (58) E. durans
Enterococcus spp.2015, USA7VRE (100)in vitro (TK)Tigecycline2 (28.6)3 (42.8)2 (28.6)42
E. faecalis2011, Italy27VRE (100)in vitro (ET)Moxifloxacin23 (85.2)4 (14.8)No distinction between additive and indifferent effect100
SA2015, Spain1in vitro (TK); in vivo (rat, tissue-cage infection model)Levofloxacinin vitro: 1 (100)in vivo: 1 (100)In the in vivo study, equivalent human dosage of DAP 8–10 mg/kg was administered.90
SA2022, India3in vitro (CB, TK)Ciprofloxacin2 (66.7)1 (33.3)69
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2005, USA40MRSA (50)in vitro (ET) (TK on 1 MRSA)AztreonamET: 2 (5); TK: 1 (100)ET: 38 (95)14
E. faecalis, E. faecium40 (21 E. faecalis, 9 E. faecium)VRE (50)in vitro (ET)ET: 40 (100)
SA2017, Turkey25MRSA (100)in vitro (CB)Fusidic acid4 (16)7 (28)14 (56)68
SA2006, USA2GISA (100)in vitro (ET, TK)Quinupristin/dalfopristinET: 2 (100); TK: 2 (100)15
SA2010, USA2MRSA (100)in vitro (PD model)TMP/SMX2 (100)DNS isolates. High inoculum tested.19
SA2012, Spain4MRSA (50), VISA (25), hVISA (25)in vitro (PK/PD model)TMP/SMX4 (100)High inoculum tested.101
SA2015, USA17MRSA (100)in vitro (TK)TMP/SMX17 (100)6 DNS isolates.102
SA2012, Italy13MRSA (92.3, 1 MSSA for in vivo study)in vitro (CB, TK), in vivo (mouse, wound infection model)Tigecyclinein vitro: 9 (75); in vivo: 1 (100)in vitro: 3 (25)No distinction between additive and indifferent effect. In the in vivo study, equivalent human dosage of DAP 7 mg/kg was administered.103
Enterococcus spp.12 E. faecalis, 12 E. faecium, 12 E. durans, 1 E. faecalis ATCC for in vivo studyin vitro: 12 (100) E. faecalis, 7 (58) E. faecium, 5 (42) E. durans; in vivo: 1 (100)in vitro: 5 (42) E. faecium, 7 (58) E. durans
Enterococcus spp.2015, USA7VRE (100)in vitro (TK)Tigecycline2 (28.6)3 (42.8)2 (28.6)42
E. faecalis2011, Italy27VRE (100)in vitro (ET)Moxifloxacin23 (85.2)4 (14.8)No distinction between additive and indifferent effect100
SA2015, Spain1in vitro (TK); in vivo (rat, tissue-cage infection model)Levofloxacinin vitro: 1 (100)in vivo: 1 (100)In the in vivo study, equivalent human dosage of DAP 8–10 mg/kg was administered.90
SA2022, India3in vitro (CB, TK)Ciprofloxacin2 (66.7)1 (33.3)69

CB, chequerboard; ET, Etest; DAP, daptomycin; DNS, daptomycin non-susceptible; SA, S. aureus; TK, time–kill assay; TMP/SMX, trimethoprim/sulfamethoxazole.

Table 11.

Studies evaluating daptomycin in combination with other antimicrobial agents (miscellanea)

StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2005, USA40MRSA (50)in vitro (ET) (TK on 1 MRSA)AztreonamET: 2 (5); TK: 1 (100)ET: 38 (95)14
E. faecalis, E. faecium40 (21 E. faecalis, 9 E. faecium)VRE (50)in vitro (ET)ET: 40 (100)
SA2017, Turkey25MRSA (100)in vitro (CB)Fusidic acid4 (16)7 (28)14 (56)68
SA2006, USA2GISA (100)in vitro (ET, TK)Quinupristin/dalfopristinET: 2 (100); TK: 2 (100)15
SA2010, USA2MRSA (100)in vitro (PD model)TMP/SMX2 (100)DNS isolates. High inoculum tested.19
SA2012, Spain4MRSA (50), VISA (25), hVISA (25)in vitro (PK/PD model)TMP/SMX4 (100)High inoculum tested.101
SA2015, USA17MRSA (100)in vitro (TK)TMP/SMX17 (100)6 DNS isolates.102
SA2012, Italy13MRSA (92.3, 1 MSSA for in vivo study)in vitro (CB, TK), in vivo (mouse, wound infection model)Tigecyclinein vitro: 9 (75); in vivo: 1 (100)in vitro: 3 (25)No distinction between additive and indifferent effect. In the in vivo study, equivalent human dosage of DAP 7 mg/kg was administered.103
Enterococcus spp.12 E. faecalis, 12 E. faecium, 12 E. durans, 1 E. faecalis ATCC for in vivo studyin vitro: 12 (100) E. faecalis, 7 (58) E. faecium, 5 (42) E. durans; in vivo: 1 (100)in vitro: 5 (42) E. faecium, 7 (58) E. durans
Enterococcus spp.2015, USA7VRE (100)in vitro (TK)Tigecycline2 (28.6)3 (42.8)2 (28.6)42
E. faecalis2011, Italy27VRE (100)in vitro (ET)Moxifloxacin23 (85.2)4 (14.8)No distinction between additive and indifferent effect100
SA2015, Spain1in vitro (TK); in vivo (rat, tissue-cage infection model)Levofloxacinin vitro: 1 (100)in vivo: 1 (100)In the in vivo study, equivalent human dosage of DAP 8–10 mg/kg was administered.90
SA2022, India3in vitro (CB, TK)Ciprofloxacin2 (66.7)1 (33.3)69
StrainYear and countryNumber of isolatesResistance (%)In vitro (methods)/in vivo (animal and site of infection)Partner drugSynergistic effect (%)Additive effect (%)Indifferent effect (%)Antagonistic effect (%)CommentsReference
SA2005, USA40MRSA (50)in vitro (ET) (TK on 1 MRSA)AztreonamET: 2 (5); TK: 1 (100)ET: 38 (95)14
E. faecalis, E. faecium40 (21 E. faecalis, 9 E. faecium)VRE (50)in vitro (ET)ET: 40 (100)
SA2017, Turkey25MRSA (100)in vitro (CB)Fusidic acid4 (16)7 (28)14 (56)68
SA2006, USA2GISA (100)in vitro (ET, TK)Quinupristin/dalfopristinET: 2 (100); TK: 2 (100)15
SA2010, USA2MRSA (100)in vitro (PD model)TMP/SMX2 (100)DNS isolates. High inoculum tested.19
SA2012, Spain4MRSA (50), VISA (25), hVISA (25)in vitro (PK/PD model)TMP/SMX4 (100)High inoculum tested.101
SA2015, USA17MRSA (100)in vitro (TK)TMP/SMX17 (100)6 DNS isolates.102
SA2012, Italy13MRSA (92.3, 1 MSSA for in vivo study)in vitro (CB, TK), in vivo (mouse, wound infection model)Tigecyclinein vitro: 9 (75); in vivo: 1 (100)in vitro: 3 (25)No distinction between additive and indifferent effect. In the in vivo study, equivalent human dosage of DAP 7 mg/kg was administered.103
Enterococcus spp.12 E. faecalis, 12 E. faecium, 12 E. durans, 1 E. faecalis ATCC for in vivo studyin vitro: 12 (100) E. faecalis, 7 (58) E. faecium, 5 (42) E. durans; in vivo: 1 (100)in vitro: 5 (42) E. faecium, 7 (58) E. durans
Enterococcus spp.2015, USA7VRE (100)in vitro (TK)Tigecycline2 (28.6)3 (42.8)2 (28.6)42
E. faecalis2011, Italy27VRE (100)in vitro (ET)Moxifloxacin23 (85.2)4 (14.8)No distinction between additive and indifferent effect100
SA2015, Spain1in vitro (TK); in vivo (rat, tissue-cage infection model)Levofloxacinin vitro: 1 (100)in vivo: 1 (100)In the in vivo study, equivalent human dosage of DAP 8–10 mg/kg was administered.90
SA2022, India3in vitro (CB, TK)Ciprofloxacin2 (66.7)1 (33.3)69

CB, chequerboard; ET, Etest; DAP, daptomycin; DNS, daptomycin non-susceptible; SA, S. aureus; TK, time–kill assay; TMP/SMX, trimethoprim/sulfamethoxazole.

Aztreonam

Forty S. aureus (20 MSSA, 20 MRSA) and 40 Enterococcus spp. (20 VSE, 20 VRE) isolates were tested, resulting in indifferent effect in 78 out of 80 cases (97.5%). Synergistic effect was observed against two isolates of S. aureus.

Fusidic acid

Only one study evaluated daptomycin in combination with fusidic acid against 25 MRSA isolates, resulting in synergistic effect against 4 (16%) isolates, additive effect against 7 (28%) and indifferent effect against 14 (56%) isolates.

Quinupristin/dalfopristin

One study evaluated daptomycin + quinupristin/dalfopristin against two isolates of GISA, and indifferent effect was observed against both isolates.

Trimethoprim/sulfamethoxazole

A total of 23 isolates of S. aureus were tested with daptomycin + trimethoprim/sulfamethoxazole; synergistic effect was observed against 19 (82.6%) isolates, while additive effect was observed against 4 (17.4%) isolates.

Tigecycline

The daptomycin + tigecycline combination was tested against 13 isolates of S. aureus and 44 isolates of Enterococcus spp. Synergistic effect was observed in most cases in vitro (75% isolates of S. aureus and 60.5% isolates of Enterococcus spp.). Antagonistic effect was observed against two isolates of Enterococcus spp. (4.5%) in vitro. Daptomycin + tigecycline was evaluated in vivo (wound infection model in mice) against one MSSA and one vancomycin-susceptible E. faecalis and resulted in synergistic effect against both isolates.

Quinolones

Daptomycin in combination with moxifloxacin, levofloxacin and ciprofloxacin was evaluated in three studies (two in vitro studies, one in vitro and in vivo study). Four isolates of S. aureus were tested and synergistic effect accounted for 75% of interactions. Antagonistic effect was observed against one isolate of S. aureus in vitro. In vivo, daptomycin + levofloxacin exerted an additive effect against one isolate of S. aureus.90

Overview

Overall, synergism accounted for 30.9% of total interactions, while indifferent effect was the most frequently observed interaction (41.9%). Antagonism accounted for 0.7% of total interactions. Against S. aureus isolates, the highest synergistic rates were observed when daptomycin was tested in combination with fosfomycin (55.6%). For S. epidermidis and Enterococcus spp., the most effective combinations were daptomycin plus cephalosporins (50%) and daptomycin plus fosfomycin (63.6%) or rifampicin (62.8%), respectively. Against S. epidermidis, daptomycin plus rifampicin could also be a valuable therapeutic option, but the extremely limited number of tested isolates (only two isolates) does not allow us to make generalistic considerations. Figure 2 provides an overview of the activity of daptomycin in combination with different antibiotic classes against isolates of S. aureus, S. epidermidis and Enterococcus spp.

Effect of daptomycin in combination with different antibiotic classes: a summary from the in vitro and in vivo studies included in the present systematic review. Numbers in square brackets report the total number of studies evaluating daptomycin in combination with different antibiotic classes. Numbers in round brackets refer to the number of the tested isolates. This figure appears in colour in the online version of JAC and in black and white in the print version of JAC.
Figure 2.

Effect of daptomycin in combination with different antibiotic classes: a summary from the in vitro and in vivo studies included in the present systematic review. Numbers in square brackets report the total number of studies evaluating daptomycin in combination with different antibiotic classes. Numbers in round brackets refer to the number of the tested isolates. This figure appears in colour in the online version of JAC and in black and white in the print version of JAC.

Discussion

To the best of our knowledge, this is the first systematic review on daptomycin synergistic properties including in vitro and in vivo studies, including all antibiotic classes previously tested. Compared with a narrative review on daptomycin synergistic combination, where no antagonism was reported, our study reported antagonistic effect for some daptomycin combinations, like daptomycin + glycopeptides against Enterococcus spp. and daptomycin + aminoglycosides against S. epidermidis (Figure 2).104 Our systematic review suggests that a tailored approach is strongly recommended, taking into account not only the causative agent and clinical aspects that might limit prescription (i.e. drug allergies, comorbidities, drug–drug interactions), but also the site of infection. In fact, the same can act differently in different in vivo models of infection (the number of in vivo studies for each combination is too limited to make general statements about the superiority of a combination regimen compared with others in a specific model of infection).45,64,82,83,96

Although some meta-analyses have been conducted on daptomycin in combination with β-lactams in clinical settings, none was performed investigating the efficacy of daptomycin in combination with molecules belonging to other antibiotic classes. Three meta-analyses on daptomycin in combination with β-lactams for the treatment of MRSA BSI reported superiority of the combination compared with monotherapy, though with an alert on the combination therapy for the increased risk of Clostridioides difficile infection.105–107 In particular, one of these studies underlined that the combination showed no significant improvement of survival, but was associated with a shorter duration of bacteraemia, a lower risk of persistent bacteraemia and of recurrence within 60–90 days, suggesting careful evaluation of both harms and benefits of daptomycin plus β-lactam combinations.107

Daptomycin is a milestone for systemic infections caused by MDR Gram-positive cocci and its administration is on the rise due to increased antimicrobial resistance worldwide and its manageable profile. Firstly, it has a good safety and tolerability profile (the most frequent side effect is rhabdomyolysis, mainly occurring in patients with acute kidney injury, end-stage renal disease, or who are taking statins) and can also be administered in children and neonates.108–110 Secondly, it exerts its bactericidal activity rapidly (in vitro, up to 90% of tested bacteria killed after 1 h) and it is also active against biofilm-embedded bacteria, therefore being a valuable therapeutic option in infections involving prosthetic materials.111,112 Thirdly, as it is active against MDR Gram-positive cocci and its PK/PD properties allow its administration once a day, its administration has also grown in recent years in outpatient settings.113 This consideration strengthens the necessity of avoiding the emergence of daptomycin non-susceptible strains.

The clinical superiority of daptomycin-containing regimens compared with monotherapy is still under debate. It is reasonable to hypothesize that the benefits of prescribing daptomycin in combination mainly depend on the partner drug, the infection site, the microbial burden and the type of microorganism involved, as emerged in the ‘Penicillins and penicillin/BLIs’ to ‘Miscellanea’ sections. Some case reports documented clinical failure when daptomycin was administered as monotherapy for BSI or endocarditis, while the infection was cleared by a daptomycin-containing regimen, thus supporting the superiority of the combination compared with monotherapy in the prevention of resistance emergence, enhanced bactericidal effect and subsequent better clinical outcomes.108,114–117 In a pilot study, the combination of daptomycin plus ceftaroline was associated with such a reduction in in-hospital mortality (0% versus 26%) compared with daptomycin monotherapy that it caused the study to be halted.118 In two cohorts of 597 and 229 patients with MRSA BSI, combination therapy (daptomycin + β-lactams) was associated with better clinical outcomes (lower 60 day mortality, 60 day recurrence and persistent bacteraemia rates).119,120 In agreement with the high synergism between daptomycin and fosfomycin that emerged in the present review, in a randomized clinical trial evaluating daptomycin plus fosfomycin and daptomycin monotherapy for MRSA BSI and endocarditis, higher treatment success rates (although not statistically significant; 54.1% versus 42.0%) were reported for combination treatment, although a higher occurrence of adverse events (17.6% versus 4.9%) was observed.121 In addition, the combination of daptomycin plus fosfomycin successfully treated a total femoral replacement infection sustained by S. epidermidis.122 A systematic review on fosfomycin synergistic properties reported consistent results.123 On the other hand, a randomized clinical trial reported daptomycin monotherapy as non-inferior to vancomycin plus gentamicin for the treatment of MRSA BSI and right-sided endocarditis.124 In another randomized clinical trial involving MRSA bacteraemic patients, daptomycin in combination with an antistaphylococcal β-lactam (IV flucloxacillin, cloxacillin or cefazolin) was non-superior to daptomycin monotherapy.125 Non superiority of the daptomycin + β-lactam (ceftaroline) combination compared with daptomycin monotherapy was also reported in a cohort of patients admitted with MRSA BSI.126 For MSSA BSI, a randomized, double-blind, placebo-controlled trial reported no significant advantages of daptomycin + cefazolin or cloxacillin compared with β-lactam monotherapy in terms of both duration of the bacteraemia (primary outcome of the study) and 90 day mortality.127

With regard to enterococcal systemic infections, although high-dose daptomycin (10–12 mg/kg/day) is generally considered effective based on available PK/PD data, EUCAST warned about the insufficient clinical evidence of the role of daptomycin in enterococcal endocarditis and BSI.128

Our systematic review suggests great heterogeneity in daptomycin synergistic properties according to the antibiotic class tested in combination. As stated in the Materials and methods section, when no distinction was possible between additive and indifferent effect, the combination was considered to exert an indifferent effect compared with monotherapy. This should be stressed, as slightly higher percentages of additive effect against some strains could be possible (see Tables 2–10 for studies that did not make this distinction). Of note, in some PD models, the little benefit observed from the combination of daptomycin with other antimicrobials is mainly due to the extremely effective and rapid bactericidal effect that daptomycin exerted alone.34,59

Conflicting results are reported about the avoidance of emergence of daptomycin resistance when daptomycin is co-administered with another active agent; one in vitro study reported that co-administration of gentamicin or rifampicin was not able to prevent daptomycin resistance, while amoxicillin/clavulanic acid or ampicillin greatly delayed the emergence of daptomycin resistance in S. aureus and Enterococcus spp. isolates.129 Similar results, and in particular the prevention of daptomycin resistance in the presence of β-lactams, clarithromycin or rifampicin, was also confirmed in other studies.130–133

Daptomycin also has antibiofilm properties, which is particularly relevant when prosthetic materials are present in the site of infection (e.g. CRBSI, prosthetic valve or prosthetic joint infection).112,134,135 When biofilm-related infections are diagnosed or strongly suspected, association with another antimicrobial agent with antibiofilm properties could be an effective treatment. In in vitro biofilm models of staphylococcal infection, daptomycin in combination with rifampicin or tigecycline was more effective than either agent tested alone.82,112,136,137

Treatment failure is often due to the development of daptomycin resistance, especially in MRSA isolates. When daptomycin was tested in vitro with β-lactams, the ‘seesaw effect’ was observed; the development of daptomycin resistance was accompanied by a concomitant restoration of β-lactam susceptibility.18,138 Taking this into account, co-administration of daptomycin and a β-lactam could offer the advantage of preventing daptomycin resistance, and in case it develops, somehow taking advantage of the seesaw effect and counting on an eventually restored susceptibility to β-lactams.

Limitations and strengths

Neither clinical studies nor abstracts presented in congresses and preprint papers were included in the current review, nor databases other than PubMed/MEDLINE were consulted. Indeed, because they did not meet inclusion criteria, studies evaluating in vitro daptomycin in combination with another active agent (e.g. aztreonam, ceftazidime, colistin) against Gram-negative bacteria, including MDR Acinetobacter baumannii isolates, even with promising results, were not included in the present review.139–142

On the other hand, this is the first evaluation of daptomycin synergistic properties carried out with rigorous methods and including all antibiotic classes. It could help clinicians define the best treatment strategy, which should however always be guided by the bacterial species isolated, antimicrobial susceptibility testing, patient allergies and comorbidities and drug–drug interactions. This systematic review also highlighted the combinations and the pathogens less tested, suggesting the fields where further research is particularly needed (e.g. in vivo studies comparing different combination regimens, daptomycin combination regimens against S. epidermidis, daptomycin plus oxazolidinones or glycopeptides against Enterococcus spp.). The updated overview of PD interactions between daptomycin and molecules of other antibiotic classes could also be useful for the future updates of guidelines on systemic infections where daptomycin plays a key role.

Conclusions

Daptomycin is a valuable therapeutic option for systemic infections caused by Gram-positive cocci. Current guidelines for the treatment of systemic infections such as infective endocarditis, SSTI, BSI and CRBSI suggest evaluating co-prescription of daptomycin with another active drug.6,7 Despite this, clinical evidence is limited and mainly based on case series rather than on randomized clinical trials regarding the superiority of combination regimens compared with monotherapy. Our systematic review presented an updated overview of most synergistic combinations, suggesting that a tailored approach is needed evaluating both clinical characteristics of the patient and the causative agent. In particular, against S. aureus, daptomycin in combination with fosfomycin, glycopeptides or oxazolidinones seems to be particularly beneficial compared with monotherapy. When treating infections by S. epidermidis, daptomycin plus penicillins, penicillins/BLIs, cephalosporins or cephalosporins/BLIs could be valuable options. Finally, daptomycin plus fosfomycin or rifampicin resulted as the most beneficial combinations compared with monotherapy against Enterococcus spp. isolates.

Funding

We carried out the study as part of our routine work without any internal or external financial support.

Transparency declarations

None to declare.

References

1

Gonzalez-Ruiz
A
,
Beiras-Fernandez
A
,
Lehmkuhl
H
et al.
Clinical experience with daptomycin in Europe: the first 2.5 years
.
J Antimicrob Chemother
2011
;
66
:
912
9
. https://doi.org/10.1093/jac/dkq528

2

Rybak
MJ
.
The efficacy and safety of daptomycin: first in a new class of antibiotics for Gram-positive bacteria
.
Clin Microb Infect
2006
;
12
:
24
32
. https://doi.org/10.1111/j.1469-0691.2006.01342.x

3

Pokorny
A
,
Almeida
PF
.
The antibiotic peptide daptomycin functions by reorganizing the membrane
.
J Membr Biol
2021
;
254
:
97
108
. https://doi.org/10.1007/s00232-021-00175-0

4

Lee
M-T
,
Yang
P-Y
,
Charron
NE
et al.
Comparison of the effects of daptomycin on bacterial and model membranes
.
Biochemistry
2018
;
57
:
5629
39
. https://doi.org/10.1021/acs.biochem.8b00818

5

Taylor
SD
,
Palmer
M
.
The action mechanism of daptomycin
.
Bioorg Med Chem
2016
;
24
:
6253
68
. https://doi.org/10.1016/j.bmc.2016.05.052

6

Habib
G
,
Lancellotti
P
,
Antunes
MJ
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
128
. https://doi.org/10.1093/eurheartj/ehv319

7

Stevens
DL
,
Bisno
AL
,
Chambers
HF
et al.
Practice guidelines for the diagnosis and management of skin and soft tissue infections: 2014 update by the Infectious Diseases Society of America
.
Clin Infect Dis
2014
;
59
:
e10
52
. https://doi.org/10.1093/cid/ciu296

8

Webb
BJ
,
Majers
J
,
Healy
R
et al.
Antimicrobial stewardship in a hematological malignancy unit: carbapenem reduction and decreased vancomycin-resistant Enterococcus infection
.
Clin Infect Dis
2020
;
71
:
960
7
. https://doi.org/10.1093/cid/ciz900

9

Moses
VK
,
Kandi
V
,
Rao
SKD
.
Minimum inhibitory concentrations of vancomycin and daptomycin against methicillin-resistant Staphylococcus aureus isolated from various clinical specimens: a study from South India
.
Cureus
2020
;
12
:
e6749
. https://doi.org/10.7759/cureus.6749

10

Beardsley
J
,
Patel
S
,
Cook
C
et al.
Inpatient antibiotic costs associated with switching from vancomycin to daptomycin for outpatient parenteral antibiotic therapy
.
Hosp Pharm
2022
;
57
:
17
9
. https://doi.org/10.1177/0018578720970466

11

Page
MJ
,
McKenzie
JE
,
Bossuyt
PM
et al.
The PRISMA 2020 statement: an updated guideline for reporting systematic reviews
.
Rev Esp Cardiol
2021
;
74
:
790
9
. https://doi.org/10.1016/j.recesp.2021.06.016

12

Rand
KH
,
Houck
HJ
.
Synergy of daptomycin with oxacillin and other beta-lactams against methicillin-resistant Staphylococcus aureus
.
Antimicrob Agents Chemother
2004
;
48
:
2871
5
. https://doi.org/10.1128/AAC.48.8.2871-2875.2004

13

Rand
KH
,
Houck
H
.
Daptomycin synergy with rifampicin and ampicillin against vancomycin-resistant enterococci
.
J Antimicrob Chemother
2004
;
53
:
530
2
. https://doi.org/10.1093/jac/dkh104

14

Snydman
DR
,
McDermott
LA
,
Jacobus
NV
.
Evaluation of in vitro interaction of daptomycin with gentamicin or beta-lactam antibiotics against Staphylococcus aureus and Enterococci by FIC index and timed-kill curves
.
J Chemother
2005
;
17
:
614
21
. https://doi.org/10.1179/joc.2005.17.6.614

15

Tsuji
BT
,
Rybak
MJ
.
Etest synergy testing of clinical isolates of Staphylococcus aureus demonstrating heterogeneous resistance to vancomycin
.
Diagn Microbiol Infect Dis
2006
;
54
:
73
7
. https://doi.org/10.1016/j.diagmicrobio.2005.08.014

16

Cilli
F
,
Aydemir
S
,
Tunger
A
.
In vitro activity of daptomycin alone and in combination with various antimicrobials against Gram-positive cocci
.
J Chemother
2006
;
18
:
27
32
. https://doi.org/10.1179/joc.2008.18.1.27

17

Huang
Y-T
,
Liao
C-H
,
Teng
L-J
et al.
Comparative bactericidal activities of daptomycin, glycopeptides, linezolid and tigecycline against blood isolates of Gram-positive bacteria in Taiwan
.
Clin Microbiol Infect
2008
;
14
:
124
9
. https://doi.org/10.1111/j.1469-0691.2007.01888.x

18

Yang
S-J
,
Xiong
YQ
,
Boyle-Vavra
S
et al.
Daptomycin-oxacillin combinations in treatment of experimental endocarditis caused by daptomycin-nonsusceptible strains of methicillin-resistant Staphylococcus aureus with evolving oxacillin susceptibility (the ‘seesaw effect’)
.
Antimicrob Agents Chemother
2010
;
54
:
3161
9
. https://doi.org/10.1128/AAC.00487-10

19

Steed
ME
,
Vidaillac
C
,
Rybak
MJ
.
Novel daptomycin combinations against daptomycin-nonsusceptible methicillin-resistant Staphylococcus aureus in an in vitro model of simulated endocardial vegetations
.
Antimicrob Agents Chemother
2010
;
54
:
5187
92
. https://doi.org/10.1128/AAC.00536-10

20

Sakoulas
G
,
Bayer
AS
,
Pogliano
J
et al.
Ampicillin enhances daptomycin- and cationic host defense peptide-mediated killing of ampicillin- and vancomycin-resistant Enterococcus faecium
.
Antimicrob Agents Chemother
2012
;
56
:
838
44
. https://doi.org/10.1128/AAC.05551-11

21

Garrigós
C
,
Murillo
O
,
Lora-Tamayo
J
et al.
Efficacy of daptomycin-cloxacillin combination in experimental foreign-body infection due to methicillin-resistant Staphylococcus aureus
.
Antimicrob Agents Chemother
2012
;
56
:
3806
11
. https://doi.org/10.1128/AAC.00127-12

22

Mehta
S
,
Singh
C
,
Plata
KB
et al.
β-Lactams increase the antibacterial activity of daptomycin against clinical methicillin-resistant Staphylococcus aureus strains and prevent selection of daptomycin-resistant derivatives
.
Antimicrob Agents Chemother
2012
;
56
:
6192
200
. https://doi.org/10.1128/AAC.01525-12

23

Leonard
SN
,
Rolek
KM
.
Evaluation of the combination of daptomycin and nafcillin against vancomycin-intermediate Staphylococcus aureus
.
J Antimicrob Chemother
2013
;
68
:
644
7
. https://doi.org/10.1093/jac/dks453

24

El Haj
C
,
Murillo
O
,
Ribera
A
et al.
Comparative efficacies of cloxacillin-daptomycin and the standard cloxacillin-rifampin therapies against an experimental foreign-body infection by methicillin-susceptible Staphylococcus aureus
.
Antimicrob Agents Chemother
2014
;
58
:
5576
80
. https://doi.org/10.1128/AAC.02681-14

25

Smith
JR
,
Barber
KE
,
Raut
A
et al.
β-Lactam combinations with daptomycin provide synergy against vancomycin-resistant Enterococcus faecalis and Enterococcus faecium
.
J Antimicrob Chemother
2015
;
70
:
1738
43
. https://doi.org/10.1093/jac/dkv007

26

Werth
BJ
,
Barber
KE
,
Tran
K-NT
et al.
Ceftobiprole and ampicillin increase daptomycin susceptibility of daptomycin-susceptible and -resistant VRE
.
J Antimicrob Chemother
2015
;
70
:
489
93
. https://doi.org/10.1093/jac/dku386

27

Smith
JR
,
Barber
KE
,
Raut
A
et al.
β-Lactams enhance daptomycin activity against vancomycin-resistant Enterococcus faecalis and Enterococcus faecium in in vitro pharmacokinetic/pharmacodynamic models
.
Antimicrob Agents Chemother
2015
;
59
:
2842
8
. https://doi.org/10.1128/AAC.00053-15

28

Henson
KER
,
Yim
J
,
Smith
JR
et al.
β-Lactamase inhibitors enhance the synergy between β-lactam antibiotics and daptomycin against methicillin-resistant Staphylococcus aureus
.
Antimicrob Agents Chemother
2017
;
61
:
e01564-16
. https://doi.org/10.1128/AAC.01564-16

29

García-de-la-Mària
C
,
Gasch
O
,
García-Gonzalez
J
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
. https://doi.org/10.1128/AAC.02633-17

30

Lee
Y-C
,
Chen
P-Y
,
Wang
J-T
et al.
A study on combination of daptomycin with selected antimicrobial agents: in vitro synergistic effect of MIC value of 1 mg/L against MRSA strains
.
BMC Pharmacol Toxicol
2019
;
20
:
25
. https://doi.org/10.1186/s40360-019-0305-y

31

Pericàs
JM
,
García-de-la-Mària
C
,
Brunet
M
et al.
Early in vitro development of daptomycin non-susceptibility in high-level aminoglycoside-resistant Enterococcus faecalis predicts the efficacy of the combination of high-dose daptomycin plus ampicillin in an in vivo model of experimental endocarditis
.
J Antimicrob Chemother
2017
;
72
:
1714
22
. https://doi.org/10.1093/jac/dkx016

32

García-de-la-Mària
C
,
Gasch
O
,
Castañeda
X
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
92
. https://doi.org/10.1093/jac/dkaa354

33

Barber
KE
,
Shammout
Z
,
Smith
JR
et al.
Biofilm time-kill curves to assess the bactericidal activity of daptomycin combinations against biofilm-producing vancomycin-resistant Enterococcus faecium and faecalis
.
Antibiotics
2021
;
10
:
897
. https://doi.org/10.3390/antibiotics10080897

34

Huang
V
,
Rybak
MJ
.
Pharmacodynamics of cefepime alone and in combination with various antimicrobials against methicillin-resistant Staphylococcus aureus in an in vitro pharmacodynamic infection model
.
Antimicrob Agents Chemother
2005
;
49
:
302
8
. https://doi.org/10.1128/AAC.49.1.302-308.2005

35

Werth
BJ
,
Sakoulas
G
,
Rose
WE
et al.
Ceftaroline increases membrane binding and enhances the activity of daptomycin against daptomycin-nonsusceptible vancomycin-intermediate Staphylococcus aureus in a pharmacokinetic/pharmacodynamic model
.
Antimicrob Agents Chemother
2013
;
57
:
66
73
. https://doi.org/10.1128/AAC.01586-12

36

Barber
KE
,
Werth
BJ
,
McRoberts
JP
et al.
A novel approach utilizing biofilm time-kill curves to assess the bactericidal activity of ceftaroline combinations against biofilm-producing methicillin-resistant Staphylococcus aureus
.
Antimicrob Agents Chemother
2014
;
58
:
2989
92
. https://doi.org/10.1128/AAC.02764-13

37

Werth
BJ
,
Barber
KE
,
Ireland
CE
et al.
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
81
. https://doi.org/10.1128/AAC.00088-14

38

Snyder
ADH
,
Werth
BJ
,
Barber
KE
et al.
Evaluation of the novel combination of daptomycin plus ceftriaxone against vancomycin-resistant enterococci in an in vitro pharmacokinetic/pharmacodynamic simulated endocardial vegetation model
.
J Antimicrob Chemother
2014
;
69
:
2148
54
. https://doi.org/10.1093/jac/dku113

39

Sakoulas
G
,
Moise
PA
,
Casapao
AM
et al.
Antimicrobial salvage therapy for persistent staphylococcal bacteremia using daptomycin plus ceftaroline
.
Clin Ther
2014
;
36
:
1317
33
. https://doi.org/10.1016/j.clinthera.2014.05.061

40

Barber
KE
,
Werth
BJ
,
Ireland
CE
et al.
Potent synergy of ceftobiprole plus daptomycin against multiple strains of Staphylococcus aureus with various resistance phenotypes
.
J Antimicrob Chemother
2014
;
69
:
3006
10
. https://doi.org/10.1093/jac/dku236

41

Barber
KE
,
Rybak
MJ
,
Sakoulas
G
.
Vancomycin plus ceftaroline shows potent in vitro synergy and was successfully utilized to clear persistent daptomycin-non-susceptible MRSA bacteraemia
.
J Antimicrob Chemother
2015
;
70
:
311
3
. https://doi.org/10.1093/jac/dku322

42

Hindler
JA
,
Wong-Beringer
A
,
Charlton
CL
et al.
In vitro activity of daptomycin in combination with β-lactams, gentamicin, rifampin, and tigecycline against daptomycin-nonsusceptible enterococci
.
Antimicrob Agents Chemother
2015
;
59
:
4279
88
. https://doi.org/10.1128/AAC.05077-14

43

Barber
KE
,
Smith
JR
,
Ireland
CE
et al.
Evaluation of ceftaroline alone and in combination against biofilm-producing methicillin-resistant Staphylococcus aureus with reduced susceptibility to daptomycin and vancomycin in an in vitro pharmacokinetic/pharmacodynamic model
.
Antimicrob Agents Chemother
2015
;
59
:
4497
503
. https://doi.org/10.1128/AAC.00386-15

44

Smith
JR
,
Arya
A
,
Yim
J
et al.
Daptomycin in combination with ceftolozane-tazobactam or cefazolin against daptomycin-susceptible and -nonsusceptible Staphylococcus aureus in an in vitro, hollow-fiber model
.
Antimicrob Agents Chemother
2016
;
60
:
3970
5
. https://doi.org/10.1128/AAC.01666-15

45

Chambers
HF
,
Basuino
L
,
Hamilton
SM
et al.
Daptomycin-β-lactam combinations in a rabbit model of daptomycin-nonsusceptible methicillin-resistant Staphylococcus aureus endocarditis
.
Antimicrob Agents Chemother
2016
;
60
:
3976
9
. https://doi.org/10.1128/AAC.00589-16

46

García
AB
,
Candel
FJ
,
López
L
et al.
In vitro ceftaroline combinations against meticillin-resistant Staphylococcus aureus
.
J Med Microbiol
2016
;
65
:
1119
22
. https://doi.org/10.1099/jmm.0.000341

47

Aktas
G
.
In-vitro activity of ceftriaxone combined with newer agents against MRSA
.
J Chemother
2017
;
29
:
383
5
. https://doi.org/10.1080/1120009X.2016.1246633

48

Shafiq
I
,
Bulman
ZP
,
Spitznogle
SL
et al.
A combination of ceftaroline and daptomycin has synergistic and bactericidal activity in vitro against daptomycin nonsusceptible methicillin-resistant Staphylococcus aureus (MRSA)
.
Infect Dis
2017
;
49
:
410
6
. https://doi.org/10.1080/23744235.2016.1277587

49

Campanile
F
,
Bongiorno
D
,
Mongelli
G
et al.
Bactericidal activity of ceftobiprole combined with different antibiotics against selected Gram-positive isolates
.
Diagn Microbiol Infect Dis
2019
;
93
:
77
81
. https://doi.org/10.1016/j.diagmicrobio.2018.07.015

50

Abdul-Mutakabbir
JC
,
Kebriaei
R
,
Stamper
KC
et al.
Dalbavancin, vancomycin and daptomycin alone and in combination with cefazolin against resistant phenotypes of Staphylococcus aureus in a pharmacokinetic/pharmacodynamic model
.
Antibiotics
2020
;
9
:
696
. https://doi.org/10.3390/antibiotics9100696

51

Hutton
MA
,
Sundaram
A
,
Perri
MB
et al.
Assessment of in vitro synergy of daptomycin or vancomycin plus ceftaroline for daptomycin non-susceptible Staphylococcus aureus
.
Diagn Microbiol Infect Dis
2020
;
98
:
115126
. https://doi.org/10.1016/j.diagmicrobio.2020.115126

52

Lai
C-C
,
Chen
C-C
,
Lu
Y-C
et al.
The potential role of sulbactam and cephalosporins plus daptomycin against daptomycin-nonsusceptible VISA and H-VISA isolates: an in vitro study
.
Antibiotics (Basel)
2019
;
8
:
184
. https://doi.org/10.3390/antibiotics8040184

53

Tsai
C-E
,
Yang
C-J
,
Chuang
Y-C
et al.
Evaluation of the synergistic effect of ceftaroline against methicillin-resistant Staphylococcus aureus
.
Int J Infect Dis
2022
;
122
:
230
6
. https://doi.org/10.1016/j.ijid.2022.05.057

54

Presterl
E
,
Hajdu
S
,
Lassnigg
AM
et al.
Effects of azithromycin in combination with vancomycin, daptomycin, fosfomycin, tigecycline, and ceftriaxone on Staphylococcus epidermidis biofilms
.
Antimicrob Agents Chemother
2009
;
53
:
3205
10
. https://doi.org/10.1128/AAC.01628-08

55

Parra-Ruiz
J
,
Vidaillac
C
,
Rose
WE
et al.
Activities of high-dose daptomycin, vancomycin, and moxifloxacin alone or in combination with clarithromycin or rifampin in a novel in vitro model of Staphylococcus aureus biofilm
.
Antimicrob Agents Chemother
2010
;
54
:
4329
34
. https://doi.org/10.1128/AAC.00455-10

56

Snyder
ADH
,
Vidaillac
C
,
Rose
W
et al.
Evaluation of high-dose daptomycin versus vancomycin alone or combined with clarithromycin or rifampin against Staphylococcus aureus and S. epidermidis in a novel in vitro PK/PD model of bacterial biofilm
.
Infect Dis Ther
2015
;
4
:
51
65
. https://doi.org/10.1007/s40121-014-0055-5

57

Parra
D
,
Peña-Monje
A
,
Coronado-Álvarez
NM
et al.
In vitro efficacy of daptomycin and teicoplanin combined with ethanol, clarithromycin or gentamicin as catheter lock solutions
.
BMC Microbiol
2015
;
15
:
245
. https://doi.org/10.1186/s12866-015-0585-3

58

El Haj
C
,
Murillo
O
,
Ribera
A
et al.
The anti-biofilm effect of macrolides in a rat model of S. aureus foreign-body infection: might it be of clinical relevance?
Med Microbiol Immunol
2017
;
206
:
31
9
. https://doi.org/10.1007/s00430-016-0479-5

59

LaPlante
KL
,
Rybak
MJ
.
Impact of high-inoculum Staphylococcus aureus on the activities of nafcillin, vancomycin, linezolid, and daptomycin, alone and in combination with gentamicin, in an in vitro pharmacodynamic model
.
Antimicrob Agents Chemother
2004
;
48
:
4665
72
. https://doi.org/10.1128/AAC.48.12.4665-4672.2004

60

Tsuji
BT
,
Rybak
MJ
.
Short-course gentamicin in combination with daptomycin or vancomycin against Staphylococcus aureus in an in vitro pharmacodynamic model with simulated endocardial vegetations
.
Antimicrob Agents Chemother
2005
;
49
:
2735
45
. https://doi.org/10.1128/AAC.49.7.2735-2745.2005

61

Credito
K
,
Lin
G
,
Appelbaum
PC
.
Activity of daptomycin alone and in combination with rifampin and gentamicin against Staphylococcus aureus assessed by time-kill methodology
.
Antimicrob Agents Chemother
2007
;
51
:
1504
7
. https://doi.org/10.1128/AAC.01455-06

62

Baltch
AL
,
Ritz
WJ
,
Bopp
LH
et al.
Activities of daptomycin and comparative antimicrobials, singly and in combination, against extracellular and intracellular Staphylococcus aureus and its stable small-colony variant in human monocyte-derived macrophages and in broth
.
Antimicrob Agents Chemother
2008
;
52
:
1829
33
. https://doi.org/10.1128/AAC.01480-07

63

LaPlante
KL
,
Woodmansee
S
.
Activities of daptomycin and vancomycin alone and in combination with rifampin and gentamicin against biofilm-forming methicillin-resistant Staphylococcus aureus isolates in an experimental model of endocarditis
.
Antimicrob Agents Chemother
2009
;
53
:
3880
6
. https://doi.org/10.1128/AAC.00134-09

64

Miró
JM
,
García-de-la-Mària
C
,
Armero
Y
et al.
Addition of gentamicin or rifampin does not enhance the effectiveness of daptomycin in treatment of experimental endocarditis due to methicillin-resistant Staphylococcus aureus
.
Antimicrob Agents Chemother
2009
;
53
:
4172
7
. https://doi.org/10.1128/AAC.00051-09

65

Tafin
UF
,
Majic
I
,
Belkhodja
CZ
et al.
Gentamicin improves the activities of daptomycin and vancomycin against Enterococcus faecalis in vitro and in an experimental foreign-body infection model
.
Antimicrob Agents Chemother
2011
;
55
:
4821
7
. https://doi.org/10.1128/AAC.00141-11

66

Luther
MK
,
Arvanitis
M
,
Mylonakis
E
et al.
Activity of daptomycin or linezolid in combination with rifampin or gentamicin against biofilm-forming Enterococcus faecalis or E. faecium in an in vitro pharmacodynamic model using simulated endocardial vegetations and an in vivo survival assay using Galleria mellonella larvae
.
Antimicrob Agents Chemother
2014
;
58
:
4612
20
. https://doi.org/10.1128/AAC.02790-13

67

Jiang
J-H
,
Peleg
AY
.
Daptomycin-nonsusceptible Staphylococcus aureus: the role of combination therapy with daptomycin and gentamicin
.
Genes (Basel)
2015
;
6
:
1256
67
. https://doi.org/10.3390/genes6041256

68

Aktas
G
,
Derbentli
S
.
In vitro activity of daptomycin combinations with rifampicin, gentamicin, fosfomycin and fusidic acid against MRSA strains
.
J Glob Antimicrob Resist
2017
;
10
:
223
7
. https://doi.org/10.1016/j.jgar.2017.05.022

69

Kamble
E
,
Sanghvi
P
,
Pardesi
K
.
Synergistic effect of antibiotic combinations on Staphylococcus aureus biofilms and their persister cell populations
.
Biofilms
2022
;
4
:
100068
. https://doi.org/10.1016/j.bioflm.2022.100068

70

Jørgensen
NP
,
Skovdal
SM
,
Meyer
RL
et al.
Rifampicin-containing combinations are superior to combinations of vancomycin, linezolid and daptomycin against Staphylococcus aureus biofilm infection in vivo and in vitro
.
Pathog Dis
2016
;
74
:
ftw019
. https://doi.org/10.1093/femspd/ftw019

71

Aktas
G
,
Derbentli
S
.
In vitro activity of daptomycin combined with dalbavancin and linezolid, and dalbavancin with linezolid against MRSA strains
.
J Antimicrob Chemother
2017
;
72
:
441
3
. https://doi.org/10.1093/jac/dkw416

72

Wu
T
,
Meyer
K
,
Harrington
AT
et al.
In vitro activity of oritavancin alone or in combination against vancomycin-susceptible and -resistant enterococci
.
J Antimicrob Chemother
2019
;
74
:
1300
5
. https://doi.org/10.1093/jac/dkz010

73

Aktas
G
.
In vitro synergistic effect of vancomycin combined with daptomycin against vancomycin-resistant enterococci
.
Microb Drug Resist
2019
;
25
:
1484
9
. https://doi.org/10.1089/mdr.2019.0033

74

Aktas
G
.
Efficacy of vancomycin in combination with various antimicrobial agents against clinical methicillin resistant Staphylococcus aureus strains
.
Pak J Med Sci Q
2021
;
37
:
151
6
. https://doi.org/10.12669/pjms.37.1.2887

75

Parra-Ruiz
J
,
Bravo-Molina
A
,
Peña-Monje
A
et al.
Activity of linezolid and high-dose daptomycin, alone or in combination, in an in vitro model of Staphylococcus aureus biofilm
.
J Antimicrob Chemother
2012
;
67
:
2682
5
. https://doi.org/10.1093/jac/dks272

76

Entenza
JM
,
Giddey
M
,
Vouillamoz
J
et al.
Assessment of the in vitro synergy of daptomycin plus linezolid against multidrug-resistant enterococci
.
J Glob Antimicrob Resist
2014
;
2
:
306
8
. https://doi.org/10.1016/j.jgar.2014.09.004

77

Luther
MK
,
LaPlante
KL
.
Observed antagonistic effect of linezolid on daptomycin or vancomycin activity against biofilm-forming methicillin-resistant Staphylococcus aureus in an in vitro pharmacodynamic model
.
Antimicrob Agents Chemother
2015
;
59
:
7790
4
. https://doi.org/10.1128/AAC.01604-15

78

Smith
JR
,
Yim
J
,
Rice
S
et al.
Combination of tedizolid and daptomycin against methicillin-resistant Staphylococcus aureus in an in vitro model of simulated endocardial vegetations
.
Antimicrob Agents Chemother
2018
;
62
:
e00101-18
. https://doi.org/10.1128/AAC.00101-18

79

Sakoulas
G
,
Eliopoulos
GM
,
Alder
J
et al.
Efficacy of daptomycin in experimental endocarditis due to methicillin-resistant Staphylococcus aureus
.
Antimicrob Agents Chemother
2003
;
47
:
1714
8
. https://doi.org/10.1128/AAC.47.5.1714-1718.2003

80

Pankey
G
,
Ashcraft
D
,
Patel
N
.
In vitro synergy of daptomycin plus rifampin against Enterococcus faecium resistant to both linezolid and vancomycin
.
Antimicrob Agents Chemother
2005
;
49
:
5166
8
. https://doi.org/10.1128/AAC.49.12.5166-5168.2005

81

Khasawneh
FA
,
Ashcraft
DS
,
Pankey
GA
.
In vitro testing of daptomycin plus rifampin against methicillin-resistant Staphylococcus aureus resistant to rifampin
.
Saudi Med J
2008
;
29
:
1726
9
.

82

John
A-K
,
Baldoni
D
,
Haschke
M
et al.
Efficacy of daptomycin in implant-associated infection due to methicillin-resistant Staphylococcus aureus: importance of combination with rifampin
.
Antimicrob Agents Chemother
2009
;
53
:
2719
24
. https://doi.org/10.1128/AAC.00047-09

83

Lefebvre
M
,
Jacqueline
C
,
Amador
G
et al.
Efficacy of daptomycin combined with rifampicin for the treatment of experimental meticillin-resistant Staphylococcus aureus (MRSA) acute osteomyelitis
.
Int J Antimicrob Agents
2010
;
36
:
542
4
. https://doi.org/10.1016/j.ijantimicag.2010.07.008

84

Cirioni
O
,
Mocchegiani
F
,
Ghiselli
R
et al.
Daptomycin and rifampin alone and in combination prevent vascular graft biofilm formation and emergence of antibiotic resistance in a subcutaneous rat pouch model of staphylococcal infection
.
Eur J Vasc Endovasc Surg
2010
;
40
:
817
22
. https://doi.org/10.1016/j.ejvs.2010.08.009

85

Garrigós
C
,
Murillo
O
,
Euba
G
et al.
Efficacy of usual and high doses of daptomycin in combination with rifampin versus alternative therapies in experimental foreign-body infection by methicillin-resistant Staphylococcus aureus
.
Antimicrob Agents Chemother
2010
;
54
:
5251
6
. https://doi.org/10.1128/AAC.00226-10

86

Pankey
GA
,
Ashcraft
DS
.
In vitro synergy of telavancin and rifampin against Enterococcus faecium resistant to both linezolid and vancomycin
.
Ochsner J
2013
;
13
:
61
5
.

87

Tang
H-J
,
Chen
C-C
,
Cheng
K-C
et al.
In vitro efficacies and resistance profiles of rifampin-based combination regimens for biofilm-embedded methicillin-resistant Staphylococcus aureus
.
Antimicrob Agents Chemother
2013
;
57
:
5717
20
. https://doi.org/10.1128/AAC.01236-13

88

Mihailescu
R
,
Furustrand Tafin
U
,
Corvec
S
et al.
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
53
. https://doi.org/10.1128/AAC.02420-12

89

Holmberg
A
,
Rasmussen
M
.
Antibiotic regimens with rifampicin for treatment of Enterococcus faecium in biofilms
.
Int J Antimicrob Agents
2014
;
44
:
78
80
. https://doi.org/10.1016/j.ijantimicag.2014.03.008

90

El Haj
C
,
Murillo
O
,
Ribera
A
et al.
Daptomycin combinations as alternative therapies in experimental foreign-body infection caused by meticillin-susceptible Staphylococcus aureus
.
Int J Antimicrob Agents
2015
;
46
:
189
95
. https://doi.org/10.1016/j.ijantimicag.2015.04.004

91

Stein
C
,
Makarewicz
O
,
Forstner
C
et al.
Should daptomycin-rifampin combinations for MSSA/MRSA isolates be avoided because of antagonism?
Infection
2016
;
44
:
499
504
. https://doi.org/10.1007/s15010-016-0874-2

92

Zheng
J-X
,
Sun
X
,
Lin
Z-W
et al.
In vitro activities of daptomycin combined with fosfomycin or rifampin on planktonic and adherent linezolid-resistant isolates of Enterococcus faecalis
.
J Med Microbiol
2019
;
68
:
493
502
. https://doi.org/10.1099/jmm.0.000945

93

Albac
S
,
Labrousse
D
,
Hayez
D
et al.
Activity of different antistaphylococcal therapies, alone or combined, in a rat model of methicillin-resistant Staphylococcus epidermidis osteitis without implant
.
Antimicrob Agents Chemother
2020
;
64
:
e01865-19
. https://doi.org/10.1128/AAC.01865-19

94

Poeppl
W
,
Tobudic
S
,
Lingscheid
T
et al.
Daptomycin, fosfomycin, or both for treatment of methicillin-resistant Staphylococcus aureus osteomyelitis in an experimental rat model
.
Antimicrob Agents Chemother
2011
;
55
:
4999
5003
. https://doi.org/10.1128/AAC.00584-11

95

Miró
JM
,
Entenza
JM
,
del Río
A
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
5
. https://doi.org/10.1128/AAC.06449-11

96

Descourouez
JL
,
Jorgenson
MR
,
Wergin
JE
et al.
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
20
. https://doi.org/10.1128/AAC.02099-12

97

Lingscheid
T
,
Poeppl
W
,
Bernitzky
D
et al.
Daptomycin plus fosfomycin, a synergistic combination in experimental implant-associated osteomyelitis due to methicillin-resistant Staphylococcus aureus in rats
.
Antimicrob Agents Chemother
2015
;
59
:
859
63
. https://doi.org/10.1128/AAC.04246-14

98

Snyder
ADH
,
Werth
BJ
,
Nonejuie
P
et al.
Fosfomycin enhances the activity of daptomycin against vancomycin-resistant enterococci in an in vitro pharmacokinetic-pharmacodynamic model
.
Antimicrob Agents Chemother
2016
;
60
:
5716
23
. https://doi.org/10.1128/AAC.00687-16

99

Yu
W
,
Zhang
J
,
Tong
J
et al.
In vitro antimicrobial activity of fosfomycin, vancomycin and daptomycin alone, and in combination, against linezolid-resistant Enterococcus faecalis
.
Infect Dis Ther
2020
;
9
:
927
34
. https://doi.org/10.1007/s40121-020-00342-1

100

Farina
C
,
Russello
G
,
Chinello
P
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
33
. https://doi.org/10.1159/000330458

101

Steed
ME
,
Werth
BJ
,
Ireland
CE
et al.
Evaluation of the novel combination of high-dose daptomycin plus trimethoprim-sulfamethoxazole against daptomycin-nonsusceptible methicillin-resistant Staphylococcus aureus using an in vitro pharmacokinetic/pharmacodynamic model of simulated endocardial vegetations
.
Antimicrob Agents Chemother
2012
;
56
:
5709
14
. https://doi.org/10.1128/AAC.01185-12

102

Claeys
KC
,
Smith
JR
,
Casapao
AM
et al.
Impact of the combination of daptomycin and trimethoprim-sulfamethoxazole on clinical outcomes in methicillin-resistant Staphylococcus aureus infections
.
Antimicrob Agents Chemother
2015
;
59
:
1969
76
. https://doi.org/10.1128/AAC.04141-14

103

Silvestri
C
,
Cirioni
O
,
Arzeni
D
et al.
In vitro activity and in vivo efficacy of tigecycline alone and in combination with daptomycin and rifampin against Gram-positive cocci isolated from surgical wound infection
.
Eur J Clin Microbiol Infect Dis
2012
;
31
:
1759
64
. https://doi.org/10.1007/s10096-011-1498-1

104

Steenbergen
JN
,
Mohr
JF
,
Thorne
GM
.
Effects of daptomycin in combination with other antimicrobial agents: a review of in vitro and animal model studies
.
J Antimicrob Chemother
2009
;
64
:
1130
8
. https://doi.org/10.1093/jac/dkp346

105

Kale-Pradhan
PB
,
Giuliano
C
,
Jongekrijg
A
et al.
Combination of vancomycin or daptomycin and beta-lactam antibiotics: a meta-analysis
.
Pharmacotherapy
2020
;
40
:
648
58
. https://doi.org/10.1002/phar.2437

106

Wang
C
,
Ye
C
,
Liao
L
et al.
Adjuvant β-lactam therapy combined with vancomycin or daptomycin for methicillin-resistant Staphylococcus aureus bacteremia: a systematic review and meta-analysis
.
Antimicrob Agents Chemother
2020
;
64
:
e01377-20
. https://doi.org/10.1128/AAC.01377-20

107

Yi
Y-H
,
Wang
J-L
,
Yin
W-J
et al.
Vancomycin or daptomycin plus a β-lactam versus vancomycin or daptomycin alone for methicillin-resistant Staphylococcus aureus bloodstream infections: a systematic review and meta-analysis
.
Microb Drug Resist
2021
;
27
:
1044
56
. https://doi.org/10.1089/mdr.2020.0350

108

Beneri
CA
,
Nicolau
DP
,
Seiden
HS
et al.
Successful treatment of a neonate with persistent vancomycin-resistant enterococcal bacteremia with a daptomycin-containing regimen
.
Infect Drug Resist
2008
;
1
:
9
11
. https://doi.org/10.2147/IDR.S3649

109

Vlashyn
OO
,
Lorenz
AM
,
Sobhanie
MM
et al.
Safety outcomes with high-dose daptomycin in patients with acute kidney injury and/or end-stage renal disease
.
J Clin Pharm Ther
2021
;
46
:
363
8
. https://doi.org/10.1111/jcpt.13289

110

Samura
M
,
Takada
K
,
Hirose
N
et al.
Incidence of elevated creatine phosphokinase between daptomycin alone and concomitant daptomycin and statins: a systematic review and meta-analysis
.
Br J Clin Pharmacol
2022
;
88
:
1985
98
. https://doi.org/10.1111/bcp.15172

111

Pankuch
GA
,
Jacobs
MR
,
Appelbaum
PC
.
Bactericidal activity of daptomycin against Streptococcus pneumoniae compared with eight other antimicrobials
.
J Antimicrob Chemother
2003
;
51
:
443
6
. https://doi.org/10.1093/jac/dkg091

112

Raad
I
,
Hanna
H
,
Jiang
Y
et al.
Comparative activities of daptomycin, linezolid, and tigecycline against catheter-related methicillin-resistant Staphylococcus bacteremic isolates embedded in biofilm
.
Antimicrob Agents Chemother
2007
;
51
:
1656
60
. https://doi.org/10.1128/AAC.00350-06

113

Wu
KH
,
Sakoulas
G
,
Geriak
M
.
Vancomycin or daptomycin for outpatient parenteral antibiotic therapy: does it make a difference in patient satisfaction?
Open Forum Infect Dis
2021
;
8
:
ofab418
. https://doi.org/10.1093/ofid/ofab418

114

Ahmad
NM
,
Rojtman
AD
.
Successful treatment of daptomycin-nonsusceptible methicillin-resistant Staphylococcus aureus bacteremia with the addition of rifampin to daptomycin
.
Ann Pharmacother
2010
;
44
:
918
21
. https://doi.org/10.1345/aph.1M665

115

Hagiya
H
,
Terasaka
T
,
Kimura
K
et al.
Successful treatment of persistent MRSA bacteremia using high-dose daptomycin combined with rifampicin
.
Intern Med
2014
;
53
:
2159
63
. https://doi.org/10.2169/internalmedicine.53.2711

116

Hall
AM
,
McTigue
SM
.
Ceftaroline plus daptomycin for refractory methicillin-resistant Staphylococcus aureus bacteremia in a child
.
J Pediatr Pharmacol Ther
2018
;
23
:
490
3
. https://doi.org/10.5863/1551-6776-23.6.490

117

Arias
CA
,
Torres
HA
,
Singh
KV
et al.
Failure of daptomycin monotherapy for endocarditis caused by an Enterococcus faecium strain with vancomycin-resistant and vancomycin-susceptible subpopulations and evidence of in vivo loss of the vanA gene cluster
.
Clin Infect Dis
2007
;
45
:
1343
6
. https://doi.org/10.1086/522656

118

Geriak
M
,
Haddad
F
,
Rizvi
K
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
. https://doi.org/10.1128/AAC.02483-18

119

Alosaimy
S
,
Sabagha
NL
,
Lagnf
AM
et al.
Monotherapy with vancomycin or daptomycin versus combination therapy with β-lactams in the treatment of methicillin-resistant Staphylococcus aureus bloodstream infections: a retrospective cohort analysis
.
Infect Dis Ther
2020
;
9
:
325
39
. https://doi.org/10.1007/s40121-020-00292-8

120

Jorgensen
SCJ
,
Zasowski
EJ
,
Trinh
TD
et al.
Daptomycin plus β-lactam combination therapy for methicillin-resistant Staphylococcus aureus bloodstream infections: a retrospective, comparative cohort study
.
Clin Infect Dis
2020
;
71
:
1
10
. https://doi.org/10.1093/cid/ciz746

121

Pujol
M
,
Miró
J-M
,
Shaw
E
et al.
Daptomycin plus fosfomycin versus daptomycin alone for methicillin-resistant Staphylococcus aureus bacteremia and endocarditis: a randomized clinical trial
.
Clin Infect Dis
2021
;
72
:
1517
25
. https://doi.org/10.1093/cid/ciaa1081

122

Luengo
G
,
Lora-Tamayo
J
,
Paredes
D
et al.
Daptomycin plus fosfomycin as salvage therapy in a difficult-to-treat total femoral replacement infection
.
J Bone Jt Infect
2018
;
3
:
207
11
. https://doi.org/10.7150/jbji.27811

123

Antonello
RM
,
Principe
L
,
Maraolo
AE
et al.
Fosfomycin as partner drug for systemic infection management. A systematic review of its synergistic properties from in vitro and in vivo studies
.
Antibiotics (Basel)
2020
;
9
:
500
. https://doi.org/10.3390/antibiotics9080500

124

Rehm
SJ
,
Boucher
H
,
Levine
D
et al.
Daptomycin versus vancomycin plus gentamicin for treatment of bacteraemia and endocarditis due to Staphylococcus aureus: subset analysis of patients infected with methicillin-resistant isolates
.
J Antimicrob Chemother
2008
;
62
:
1413
21
. https://doi.org/10.1093/jac/dkn372

125

Tong
SYC
,
Lye
DC
,
Yahav
D
et al.
Effect of vancomycin or daptomycin with vs without an antistaphylococcal β-lactam on mortality, bacteremia, relapse, or treatment failure in patients with MRSA bacteremia: a randomized clinical trial
.
JAMA
2020
;
323
:
527
37
. https://doi.org/10.1001/jama.2020.0103

126

Ahmad
O
,
Crawford
TN
,
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
. https://doi.org/10.1007/s40121-019-00277-2

127

Cheng
MP
,
Lawandi
A
,
Butler-Laporte
G
et al.
Adjunctive daptomycin in the treatment of methicillin-susceptible Staphylococcus aureus bacteremia: a randomized, controlled trial
.
Clin Infect Dis
2021
;
72
:
e196
203
. https://doi.org/10.1093/cid/ciaa1000

128

Turnidge
J
,
Kahlmeter
G
,
Cantón
R
et al.
Daptomycin in the treatment of enterococcal bloodstream infections and endocarditis: a EUCAST position paper
.
Clin Microbiol Infect
2020
;
26
:
1039
43
. https://doi.org/10.1016/j.cmi.2020.04.027

129

Entenza
JM
,
Giddey
M
,
Vouillamoz
J
et al.
In vitro prevention of the emergence of daptomycin resistance in Staphylococcus aureus and enterococci following combination with amoxicillin/clavulanic acid or ampicillin
.
Int J Antimicrob Agents
2010
;
35
:
451
6
. https://doi.org/10.1016/j.ijantimicag.2009.12.022

130

Berti
AD
,
Wergin
JE
,
Girdaukas
GG
et al.
Altering the proclivity towards daptomycin resistance in methicillin-resistant Staphylococcus aureus using combinations with other antibiotics
.
Antimicrob Agents Chemother
2012
;
56
:
5046
53
. https://doi.org/10.1128/AAC.00502-12

131

Rose
WE
,
Schulz
LT
,
Andes
D
et al.
Addition of ceftaroline to daptomycin after emergence of daptomycin-nonsusceptible Staphylococcus aureus during therapy improves antibacterial activity
.
Antimicrob Agents Chemother
2012
;
56
:
5296
302
. https://doi.org/10.1128/AAC.00797-12

132

García-de-la-Mària
C
,
Pericas
JM
,
Del Río
A
et al.
Early in vitro and in vivo development of high-level daptomycin resistance is common in mitis group streptococci after exposure to daptomycin
.
Antimicrob Agents Chemother
2013
;
57
:
2319
25
. https://doi.org/10.1128/AAC.01921-12

133

Golikova
MV
,
Strukova
EN
,
Portnoy
YA
et al.
A novel parameter to predict the effects of antibiotic combinations on the development of Staphylococcus aureus resistance: in vitro model studies at subtherapeutic daptomycin and rifampicin exposures
.
J Chemother
2019
;
31
:
1
9
. https://doi.org/10.1080/1120009X.2018.1533266

134

LaPlante
KL
,
Mermel
LA
.
In vitro activity of daptomycin and vancomycin lock solutions on staphylococcal biofilms in a central venous catheter model
.
Nephrol Dial Transplant
2007
;
22
:
2239
46
. https://doi.org/10.1093/ndt/gfm141

135

Smith
K
,
Perez
A
,
Ramage
G
et al.
Comparison of biofilm-associated cell survival following in vitro exposure of meticillin-resistant Staphylococcus aureus biofilms to the antibiotics clindamycin, daptomycin, linezolid, tigecycline and vancomycin
.
Int J Antimicrob Agents
2009
;
33
:
374
8
. https://doi.org/10.1016/j.ijantimicag.2008.08.029

136

Schutt
AC
,
Bohm
NM
.
Multidrug-resistant Enterococcus faecium endocarditis treated with combination tigecycline and high-dose daptomycin
.
Ann Pharmacother
2009
;
43
:
2108
12
. https://doi.org/10.1345/aph.1M324

137

Polidori
M
,
Nuccorini
A
,
Tascini
C
et al.
Vancomycin-resistant Enterococcus faecium (VRE) bacteremia in infective endocarditis successfully treated with combination daptomycin and tigecycline
.
J Chemother
2011
;
23
:
240
1
. https://doi.org/10.1179/joc.2011.23.4.240

138

Ortwine
JK
,
Werth
BJ
,
Sakoulas
G
et al.
Reduced glycopeptide and lipopeptide susceptibility in Staphylococcus aureus and the ‘seesaw effect’: taking advantage of the back door left open?
Drug Resist Updat
2013
;
16
:
73
9
. https://doi.org/10.1016/j.drup.2013.10.002

139

LaPlante
KL
,
Sakoulas
G
.
Evaluating aztreonam and ceftazidime pharmacodynamics with Escherichia coli in combination with daptomycin, linezolid, or vancomycin in an in vitro pharmacodynamic model
.
Antimicrob Agents Chemother
2009
;
53
:
4549
55
. https://doi.org/10.1128/AAC.00180-09

140

Phee
L
,
Hornsey
M
,
Wareham
DW
.
In vitro activity of daptomycin in combination with low-dose colistin against a diverse collection of Gram-negative bacterial pathogens
.
Eur J Clin Microbiol Infect Dis
2013
;
32
:
1291
4
. https://doi.org/10.1007/s10096-013-1875-z

141

Galani
I
,
Orlandou
K
,
Moraitou
H
et al.
Colistin/daptomycin: an unconventional antimicrobial combination synergistic in vitro against multidrug-resistant Acinetobacter baumannii
.
Int J Antimicrob Agents
2014
;
43
:
370
4
. https://doi.org/10.1016/j.ijantimicag.2013.12.010

142

Yang
H
,
Chen
G
,
Hu
L
et al.
In vivo activity of daptomycin/colistin combination therapy in a Galleria mellonella model of Acinetobacter baumannii infection
.
Int J Antimicrob Agents
2015
;
45
:
188
91
. https://doi.org/10.1016/j.ijantimicag.2014.10.012

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)