Skip to main content

Adverse events during intravenous fosfomycin therapy in a real-life scenario. Risk factors and the potential role of therapeutic drug monitoring

Abstract

Background

Intravenous fosfomycin (IVFOF) is gaining interest in severe infections. Its use may be limited by adverse events (AEs). Little experience exists on IVFOF therapeutic drug monitoring (TDM) in real-life setting.

Patients and methods

Retrospective study of patients receiving IVFOF for > 48 h at Policlinico Hospital (Milan, Italy) from 01/01/2019 to 01/01/2023. AEs associated to IVFOF graded CTCAE ≥ II were considered. Demographic and clinical risk factors for IVFOF-related AEs were analysed with simple and multivariable regression models. The determination of IVFOF TDM was made by a rapid ultraperformance liquid chromatography mass spectrometry method (LC-MS/MS) on plasma samples. The performance of TDM (trough levels (Cmin) in intermittent infusion, steady state levels (Css) in continuous infusion) in predicting AEs ≤ 5 days after its assessment was evaluated.

Results

Two hundred and twenty-four patients were included. At IVFOF initiation, 81/224 (36.2%) patients were in ICU and 35/224 (15.7%) had septic shock. The most frequent infection site was the low respiratory tract (124/224, 55.4%). Ninety-five patients (42.4%) experienced ≥ 1AEs, with median time of 4.0 (2.0–7.0) days from IVFOF initiation. Hypernatremia was the most frequent AE (53/224, 23.7%). Therapy discontinuation due to AEs occurred in 38/224 (17.0%). ICU setting, low respiratory tract infections and septic shock resulted associated with AEs (RRadjusted 1.59 (95%CI:1.09–2.31), 1.46 (95%CI:1.03–2.07) and 1.73 (95%CI:1.27–2.37), respectively), while IVFOF daily dose did not. Of the 68 patients undergone IVFOF TDM, TDM values predicted overall AEs and hypernatremia with AUROC of 0.65 (95%CI:0.44–0.86) and 0.91 (95%CI:0.79-1.0) respectively for Cmin, 0.67 (95%CI:0.39–0.95) and 0.76 (95%CI:0.52-1.0) respectively for Css.

Conclusions

We provided real world data on the use of IVFOF-based regimens and associated AEs. IVFOF TDM deserves further research as it may represent a valid tool to predict AEs.

Key points

Real world data on intravenous fosfomycin for severe bacterial infections. AEs occurred in over 40% (therapy discontinuation in 17%) and were related to baseline clinical severity but not to fosfomycin dose. TDM showed promising results in predicting AEs.

Peer Review reports

Background

Intravenous fosfomycin (IVFOF) has recently gained interest for the treatment of severe infections, particularly when caused by multidrug-resistant organisms (MDROs) [1]. Its unique mechanism of action provides activity against both gram-positive and gram-negative bacteria and prevents cross-resistance to other class of antibiotics [2].

IVFOF is generally prescribed in combination therapy, to avoid the occurrence of resistance and take advantage of its synergism with several other antimicrobials. [3, 4] Recently, it has also showed good results as monotherapy for the treatment of complicated urinary tract infections caused by Escherichia coli and other Enterobacterales. [5,6,7]

The use of IVFOF may however be limited by significant adverse events (AEs), specifically electrolyte disorders (hypernatremia and hypokalaemia), gastrointestinal intolerance or cardiac failure due to sodium overload [8, 9]. Furthermore, there is a great variability in IVFOF dosage (12 to 24 g per day, administered 2 to 4 times daily or as a continuous infusion), depending on renal function and source and severity of the infection, with potential risk of under-/over-dosing especially in critically ill patients. [10,11,12]

Therapeutic drug monitoring (TDM) is proved to be useful for antibiotic dose adjustments for maintaining effective and safe drug concentrations as well as lowering the risk of resistance development. [13] TDM of glycopeptides and aminoglycosides is routinely performed, while TDM of β-lactams is recommended in critically ill patients with altered pharmacokinetic/pharmacodynamic (PK/PD) parameters. [14,15,16] To date, IVFOF TDM is not routinely performed in most centers due to the lack of commercial kits for drug quantification and the absence of established cut-offs for drug safety and/or efficacy. However, observations from single case reports or small cases series have provided preliminary evidence on the role of TDM in optimizing IVFOF administration. [17,18,19,20,21]

Here we report our real-life experience of IVFOF-based antibiotic therapies in a large cohort of patients with severe bacterial infections, with a focus on drug-related AEs and factors associated with their development.

Patients and methods

Study design and setting

Real-life, single centre retrospective cohort study of patients treated with IVFOF at Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico (Milan, Italy) from January 1, 2019 to January 1, 2023. InfectoFos® (InfectoPharm s.r.l., Milan, Italy) preparation for intravenous use is employed in our Institution; IVFOF dosage was chosen based on source of infection, microbial isolates and renal function according to technical data sheet. Each gram contains 14 mEq (320 mg) of sodium. [22] As per clinical practice in our Hospital, IVFOF TDM was not standardised but requested on a case-by-case basis by the infectious diseases (ID) consultant in charge of the patient management. Samples are usually collected into EDTA plasma sampling tubes ≥ 48 h from IVFOF start, centrifuged and sent to the laboratory of the Unit of Clinical Pharmacology ASST Fatebenefratelli Sacco (Milan, Italy) on ice and frozen at -20 °C until analysis. The laboratory processes IVFOF TDM samples twice weekly using a rapid ultraperformance liquid chromatography mass spectrometry method as previously described. [23] In the absence of established cut off and clear IVFOF PK/PD parameters of microbiological eradication and clinical efficacy, TDM values were managed by the ID consultant in collaboration with the Clinical Pharmacologist. We considered IVFOF with a time-dependent killing activity (PK/PD index: T> MIC) based on previous studies [12, 24,25,26] and the microbiological characteristics of our cohort, with the majority of isolates represented by Pseudomonas aeruginosaandStaphylococcus aureus(Table S1). The value of 100% T> MIC (or ECOFF, if MIC not available) of the isolated pathogen was conservatively and arbitrarily chosen as the time-dependent killing activity index.

Study participants and data collection

All consecutive patients treated with IVFOF were considered for inclusion. Exclusion criteria was length of IVFOF treatment ≤ 48 h (discontinuation not due to AEs) and lack of data on the primary outcome (no information on AEs).

Demographic, clinical, laboratory and outcome data were collected from clinical records. Microbiological and therapeutic data were collected from dedicated hospital-databases.

Bacterial isolates were defined as MDROs when non-susceptible to at least one agent in three or more antimicrobial categories or when harbouring specific antibiotic resistance mechanisms (e.g., methicillin-resistant Staphylococcus aureus; vancomycin-resistant Enterococcus faecium; extended spectrum beta-lactamase (ESBL)- or carbapenemase-producing Enterobacterales). [27]

AEs were registered if: (i) reported by IVFOF technical data sheet [28], (ii) had severity grade II or higher according to Common Terminology Criteria for Adverse Events (CTCAE) Version 5.0 [29] and (iii) occurred from the first day of IVFOF administration until 10 days after its discontinuation. Specifically, the following AEs were registered: diarrhoea, if ≥4 stools/day; nausea, when resulting in reduced oral intake; hypernatremia, if evidence of sodium ≥150 mmol/L; hypokalemia, if evidence of potassium < 3 mmol/L with symptoms associated; hypertransaminasemia if evidence of alanine transaminase (ALT) > 3.0 x upper normal limit or > 3.0 x baseline if baseline was abnormal; any registered cardiac event (e.g., development of arrhythmias, QT prolongation, cardiac arrest).

TDM values were categorized as trough concentration (minimum concentration, Cmin) when IVFOF was administered as intermittent infusion (II) and the sample was collected 30 min before the next dose administration, or as steady-state concentration (Css) in case of continuous infusion (CI). For patients treated with multiple courses of IVFOF or managed with repeated TDM, only first IVFOF employment and first TDM assessment were considered.

The primary outcome was to investigate epidemiological and clinical factors associated with the development of moderate or severe IVFOF-related AEs. Secondary outcome was to evaluate whether IVFOF TDM could predict the development of AEs within 5 days from its assessment (AEs≤ 5days). The 5-day interval was set to include events occurred before treatment changes based on TDM.

Anonymized data were abstracted on standardized data collection forms in the web platform REDCap (Reaserch Electronic Data Capture). [30, 31]

The study was registered by the Milan Area 2 Ethical Committee (#664_2022bis) and was conducted in accordance with standards of the Helsinki Declaration. Informed consent for pseudonymized data processing for future research purposes was provided by all patients at the time hospital admission, as routine procedure. Specific written informed consent was waived because of the retrospective nature of the analysis.

Statistical analysis

Continuous variables were presented as medians and first and third quartiles (Q1-Q3), categorical variables were reported as frequencies and proportions. Group comparisons were conducted using appropriate statistical tests depending on the variable distribution, including the Mann-Whitney test for continuous variables and the Chi-squared test or Fisher’s exact test for categorical variables.

For the analysis of factors associated with the occurrence of IVFOF-related AEs, log-binomial regression models were used, or Poisson regression models with robust error variance when the log-binomial model failed to converge. Risk ratios (RRs) were estimated, along with their corresponding 95% CIs. [32] In all the multivariable regression models, factors were entered into the adjusted model on the basis of their univariate relation to outcome (p < 0.20) along with possible confounders. All factors were biologically plausible with a sound scientific rationale. However, if the Pearson or Spearman correlation coefficient (according to variables distribution), was > 0.20, the variable with the lower p-value was retained in the model (for example, when septic shock with vasopressors was considered, IVFOF treatment started in ICU was excluded). Confounders included in multivariable models are reported in footnotes.

To evaluate the performance of TDM values in predicting at least one AEs≤ 5days or hypernatremia≤ 5days, receiver operating characteristic (ROC) curves were constructed and their corresponding areas under the curve (AUC) were evaluated in both groups of IVFOF CI and II.

Statistical analyses were performed using SAS 9.4 software (Cary, NC, USA). For all tests, a two-tailed significance level was considered.

Results

Study population

Three-hundreds and twenty-one patients were considered. Of them, 70 were excluded from the analysis because treated for ≤ 48 h (surgical prophylaxis, early interruption due to death or other causes unrelated to treatment), 5 because IVFOF was started as empiric therapy and discontinued once microbial results were available and 22 patients for missing data related to the primary outcome (Fig. 1).

Demographic and clinical characteristics of the study cohort are reported in Table 1, overall and for patients who did and did not develop IVFOF-related AEs.

Table 1 Demographic and clinical characteristics of the overall study population. Comparison between patients who developed adverse events (AEs) or not (no-AEs) related to the use of IVFOF

Overall, median age was 63.0 (50.0-71.5) years, 86/224 (38.4%) were female and 4/224 (1.8%) were under 18 years of age. One hundred and forty-six patients (65.5%) had at least one chronic illness, with diabetes mellitus being the most frequent (67/224, 29.9%). The majority of patients (203/224, 90.6%) had a microbiologically defined infection, with MDROs in almost half of them (99/203, 48.3%). The most frequent infection site was represented by the lower respiratory tract (124/224, 55.4%). Bloodstream infections (BSI) were 72/224 (32.1%), with 53 cases secondary to another infectious site and 19/224 (8.5%) primary BSI. At IVFOF initiation, 81/224 (36.2%) patients were admitted in the ICU and 35/224 (15.7%) had septic shock. Median length of hospitalization was 43 (26-75.5) days and overall in-hospital mortality was 60/224 (26.8%), confirming the high clinical severity of the population in analysis. IVFOF was administered at a median daily dose of 16.6 (12.0–24.0) grams, started within 3 (1–7) days from pathogen identification. The majority of patients (190/224, 84.8%) received IVFOF as II, 34/224 (15.2%) as CI. Median length of treatment was 11.0 (7.0-16.5) days. IVFOF was employed as part of combination therapy in all but 4 cases (2 infections sustained by Enterobacterales and 2 empirical therapies based on previous Enterobacterales infection/colonization). Details on microbial isolates are reported in TableS1, details on therapeutic regimens in Table S2 and Table S3.

Over a third of the study population (95/224, 42.4%) experienced ≥ 1 IVFOF-related AEs, with therapy discontinuation in 38/224 (17.0%) (Table 1). AEs occurred at a median time of 4.0 (2.0–7.0) days from IVFOF initiation. Hypernatremia was the most frequent AE (53/224, 23.7%), followed by hypokalaemia (22/224 9.8%), diarrhoea (20/224, 8.9%), hypertransaminasemia (12/224, 5.4%), nausea (12/224, 5.4%) and cardiac events (5/224, 2.2%). Patients who developed AEs were older and had a more severe clinical condition compared to no-AEs group, with ICU setting and septic shock at IVFOF initiation in 47/95 (49.5%) vs. 34/129 (26.4%) (p < 0.001) and 24/95 (25.5%) vs. 11/129 (8.5%) (p < 0.001), respectively. IVFOF was started for lower respiratory tract infections more frequently in AEs group than no-AEs group (62/95 (65.3%) vs. 62/129 (48.1%), p 0.011). No significant differences were observed in IVFOF daily dose.

TDM subgroup

Among the study population, 68/224 patients (30.4%) underwent IVFOF TDM, with 33/68 (48.5%) having more than one TDM assessment. Patients assigned to TDM had a more severe baseline condition compared to no-TDM patients, with septic shock at IVFOF initiation in 16/68 (23.9%) vs. 19/156 (12.2%) (p 0.028). IVFOF mode of administration differed significantly between groups, with CI in 28/68 (41.2%) of TDM group compared to 6/156 (3.9%) of no-TDM group (p < 0.001). No significant differences were observed in IVFOF daily dose, length of treatment nor the occurrence of AEs related to IVFOF. On the contrary, IVFOF dose adjustment not related to AEs occurred more frequently in TDM compared to no-TDM group (17/68 (25.0%) vs. 10/156 (6.4%), p < 0.001) (Table S4). IVFOF TDM samples were obtained after a median of 3.5 (2.5-6.0) days from therapy initiation. Among the 40 patients treated with IVFOF as II, median Cmin value was 171.5 (68.5-244.5) mg/L. Among the 28 patients treated with IVFOF as CI, median Css value was 188.8 (138.0-329.0) mg/L.

Factors associated to the development of AEs during treatment

In multivariate analysis, ICU setting (adjusted RR 1.59 (95%CI 1.09–2.31), p 0.016), lower respiratory tract infection (adjusted RR 1.46 (95%CI 1.03–2.07), p 0.031) and septic shock at IVFOF initiation (adjusted RR 1.73 (95%CI 1.27–2.37), p < 0.001) resulted associated with the development of AEs. Age, baseline alteration of renal function or the presence of any chronic illness did not increase the risk of AEs. Likewise, IVFOF daily dose nor mode of administration (CI compared to II) were related to AEs during treatment (Table 2).

Table 2 Univariate and multivariate analysis of the association of selected demographic, clinical and therapeutic factors to the development of AEs during IVFOF treatment

Within IVFOF-based combination regimens, no specific antibiotic was found associated to the development of AEs (Table S5).

Among the 68 patients who underwent TDM, 37 (54.4%) developed ≥ 1 AEs during IVFOF treatment, with events occurring within 5 days after TDM assessment in 18 patients (12 with IVFOF II and 6 with IVFOF CI) (Supplementary Fig. S1). Patients who developed AEs≤ 5days showed higher TDM levels compared to those who did not, albeit not reaching statistical significance. When comparing patients who did and did not develop AEs≤ 5days, Cmin median values were 211.3 (113.0-302.5) mg/L vs. 140.6 (64.9-227.2) mg/L (p 0.167), Css median values were 241.0 (164.5–369.0) mg/L vs. 146.0 (111.0-314.0) mg/L (p 0.269), respectively. Receiver operating characteristic (ROC) curves showed AUC of 0.65 (95%CI 0.44–0.86) for Cmin and AUC of 0.67 (0.39–0.95) for Css. When comparing patients who developed hypernatremia≤ 5days (the most frequent AE registered) to those who did not develop AEs≤ 5days, Cmin median values were 419.5 (266.0-655.0) mg/L vs. 140.6 (64.9-227.2) mg/L (p 0.012), Css median values were 294.0 (188.0-369.0) mg/L vs. 146.0 (111.0-314.0) mg/L (p 0.103), respectively. ROC curve showed good discriminatory ability for Cmin with AUC of 0.91 (95%CI 0.79-1.0), and weak discriminatory ability for Css with AUC of 0.76 (0.52-1.0). (Supplementary Figures S1-S3).

Discussion

Our study provides real world data on the use of IVFOF-based regimens in clinical practice, particularly in treating severe infections caused by MDROs. AEs occurred in more than 40% of the study population and were mainly associated to critically ill conditions at IVFOF initiation, although treatment interruption was needed in only a minority of cases. We also highlighted a possible role of IVFOF TDM in predicting the development of AEs during treatment.

With over 220 patients evaluated, this study finds its place among the largest clinical studies on IVFOF so far. Over the last 20 years, only 4 randomized clinical trials (RCT) were published, with the remaining being prospective or retrospective observational studies. Great heterogeneity exists in published literature on patients’ severity and infection type. ICU population ranged from null to over 90% of examined patients. [4, 8, 33, 34] The majority of studies focused on gram-negative MDROs, but a few evaluated specifically infections by gram-positive bacteria. [35,36,37,38] Several types of infections have been reported, with pneumonia [39] (including pulmonary exacerbations in patient with cystic fibrosis [40]), urinary tract infections [5,6,7] and osteoarticular [36, 37, 41] infections being the most frequent. Consistently with the real-life study performed in 2012 in France by Dinh et al., [42] in our cohort 40% of patients started IVFOF in ICU and 15% had septic shock requiring vasopressors at treatment initiation, with gram-positive bacteria in slightly less than a third of cases.

The rate of AEs during treatment observed in our patients was above  40%, higher than what reported in most observational studies but comparable to findings of RCTs. [5, 6, 38, 43] This is likely due to the application of the same classification criteria for AEs, [29] but is also related to the setting of IVFOF use with 40% of treatment started in the ICU. Of note, in our cohort the factors independently associated with the development of AEs were ICU setting, the presence of deep-seated infections (lower respiratory tract infections) and septic shock at IVFOF initiation.

With 23.7% of the total study population experiencing moderate to severe hypernatremia during treatment and only 3.6% already hypernatremic at IVFOF initiation, our study confirmed the high sodium intake related to the drug, which could constitute an issue especially in patients with pre-existing heart conditions or renal failure. The finding, however, may have limited reliability as it is influenced by possible confounders (concomitant therapies, renal failure, critically-ill conditions) particularly in the ICU population, where hypernatremia is reported in 6–26% of patients. [44]

Overall, our findings support recent reviews corroborating the good safety profile of IVFOF, [9, 45] as AEs were generally non-serious and led to discontinuation of treatment in less than 20% of cases.

TDM-guided therapy of glycopeptides, aminoglycosides and, more recently, betalactams has shown that dose modification based on validated PK/PD targets is associated with better clinical outcomes, in terms of both efficacy and the reduction of AEs and resistance during treatment. [13, 15, 46]

Clinical data on IVFOF TDM are currently limited to animal models, case reports and case series [17,18,19,20,21, 47, 48] but no focus has been placed on its correlation to drug toxicity so far. Cojutti et al. described clinical efficacy of the combination therapy with CI IVFOF plus meropenem in bacteriemic VAP caused by MDR Klebsiella pneumoniae, with real-time TDM-based program allowing to achieve optimal PK/PD indexes [18]. Gatti et al. reported the effective treatment of post-neurosurgical ventriculitis caused by carbapenem-resistant-Pseudomonas aeruginosa with IVFOF plus ceftazidime/avibactam using TDM-guided approach [19]. The same authors reported positive outcomes in 6 patients with BSI and/or pneumonia caused by DTR (difficult-to-treat resistance)-Pseudomonas aeruginosa treated with CI IVFOF plus extended-infusion cefiderocol or continuous-infusion ceftazidime-avibactam, demonstrating that microbiological eradication was associated with the obtainment of pre-specified PK/PD targets. Median IVFOF Css was 504.9 (363.2–647.2) mg/L, higher than what observed in our study [21]. However, no treatment-related AEs was reported in any of the studies above. By contrast, in a German observational study on 17 patients with ventriculitis treated with 24 g/day CI IVFOF, median Css was 200 (159–289) mg/L, comparable with our findings. Cases of hypernatremia were reported, particularly in patients with high sodium levels prior to IVFOF start, but correlation with IVFOF TDM was not studied. [49]

In our cohort, nor IVFOF daily dose nor specific antibiotic combination regimen was associated with the development of AEs. On the other hand, albeit based on very few observations which limited the statistical significance of our results, we found a potential role of TDM in predicting IVFOF-related AEs that occur close to TDM assessment.

Our study has limitations. Firstly, the retrospective design, the lack of a control group and the presence of possible confounders (e.g., high proportion of ICU patients and organ dysfunction, use of combination therapy) warrant some caution in associating AEs to IVFOF alone. Yet, this is the most common real-life scenario where the drug is employed, and all the AEs in analysis are known to be associated with IVFOF. [8, 9, 50] As stated before, the frequency and types of AEs in our cohort were consistent with RCTs and prospective, multicenter studies. [4,5,6, 38, 43]. Secondly, the relatively small sample size, particularly in the TDM group and in the II and CI subgroups, have likely limited the statistical power of the association between IVFOF TDM and the development of AEs. Moreover, the choice to perform TDM was made on a case-by-case basis by the ID consultant, depending on patients’ severity and risk of microbiological failure. Further studies are needed to confirm TDM role in predicting drug toxicity during IVFOF treatment and to establish and validate cut-off values. Lastly, since MIC values were not available for all the bacterial isolates, we could not calculate PK/PD indexes associated to drug efficacy. This should be further investigated, but was beyond the scope of our current work.

Conclusions

Our real-life data confirms IVFOF-based combination regimens as promising and feasible options for the treatment of severe infections. In this setting, drug-related AEs are expected in a significant proportion of patients, especially in those with a baseline critical condition, but treatment discontinuation is needed in only a minority of cases. IVFOF TDM worth future research, since it may represent a valid tool not only to achieve effective PK/PD targets but also to reduce drug-related AEs.

Fig. 1
figure 1

Flowchart of the study population

Data availability

Anonymized data were abstracted on standardized data collection forms in the web platform REDCap (Reaserch Electronic Data Capture).

Abbreviations

AE:

Adverse Events

AUROC:

Area Under the Receiver Operating Characteristic

AUC:

Area Under the Curve

CI:

Continuous Infusion

Cmin:

Minimum Concentration

CRRT:

Continuous Renal Replacement Therapy

Css:

Steady-State Concentration

CTCAE:

Common Terminology Criteria for Adverse Events

DTR:

Difficult-to-Treat Resistance

ECOFF:

Epidemiologic Cutoff

eGFR:

Estimated Glomerular Filtration Rate

ESBL:

Extended Spectrum Beta-Lactamases

ICU:

Intensive Care Unit

II:

Intermittent Infusion

IVFOF:

Intravenous Fosfomycin

KDIGO:

Kidney Disease Improving Global Outcomes

MDROs:

Multidrug-Resistant Organisms

MIC:

Minimal Inhibitory Concentration

PK/PD:

Pharmacokinetic/Pharmacodinamic

RCT:

Randomized Clinical Trials

TDM:

Therapeutic Drug Monitoring

VAP:

Ventilator Associated Pneumonia

References

  1. Vardakas KZ, Legakis NJ, Triarides N, Falagas ME. Susceptibility of contemporary isolates to fosfomycin: a systematic review of the literature. Int J Antimicrob Agents. 2016;47:269–85.

    Article  CAS  PubMed  Google Scholar 

  2. Falagas ME, Vouloumanou EK, Samonis G, Vardakasa KZ. Fosfomycin Clin Microbiol Rev. 2016;29:321–47.

    Article  CAS  PubMed  Google Scholar 

  3. Falagas ME, Kastoris AC, Kapaskelis AM, Karageorgopoulos DE. Fosfomycin for the treatment of multidrug-resistant, including extended-spectrum β-lactamase producing, Enterobacteriaceae infections: a systematic review. Lancet Infect Dis. 2010;10:43–50.

    Article  CAS  PubMed  Google Scholar 

  4. Putensen C, Ellger B, Sakka SG, Weyland A, Schmidt K, Zoller M, et al. Current clinical use of intravenous fosfomycin in ICU patients in two European countries. Infection. 2019;47:827–36.

    Article  CAS  PubMed  Google Scholar 

  5. Kaye KS, Rice LB, Dane AL, Stus V, Sagan O, Fedosiuk E, et al. Fosfomycin for Injection (ZTI-01) Versus Piperacillin-Tazobactam for the treatment of complicated urinary tract infection including Acute Pyelonephritis: ZEUS, a phase 2/3 Randomized Trial. Clin Infect Dis. 2019;69:2045–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Sojo-Dorado J, López-Hernández I, Rosso-Fernandez C, Morales IM, Palacios-Baena ZR, Hernández-Torres A, et al. Effectiveness of Fosfomycin for the treatment of Multidrug-Resistant Escherichia coli Bacteremic urinary tract infections: a Randomized Clinical Trial. JAMA Netw Open. 2022;5:e2137277–2137277.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Merino-Bohórquez V, Docobo-Pérez F, Sojo J, Morales I, Lupión C, Martín D, et al. Population pharmacokinetics and pharmacodynamics of fosfomycin in non–critically ill patients with bacteremic urinary infection caused by multidrug-resistant Escherichia coli. Clin Microbiol Infect. 2018;24:1177–83.

    Article  PubMed  Google Scholar 

  8. Florent A, Chichmanian RM, Cua E, Pulcini C. Adverse events associated with intravenous fosfomycin. Int J Antimicrob Agents. 2011;37:82–3.

    Article  CAS  PubMed  Google Scholar 

  9. Iarikov D, Wassel R, Farley J, Nambiar S. Adverse events Associated with Fosfomycin Use: review of the literature and analyses of the FDA adverse event reporting System Database. Infect Dis Ther. 2015;4:433.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Roussos N, Karageorgopoulos DE, Samonis G, Falagas ME. Clinical significance of the pharmacokinetic and pharmacodynamic characteristics of fosfomycin for the treatment of patients with systemic infections. Int J Antimicrob Agents. 2009;34:506–15.

    Article  CAS  PubMed  Google Scholar 

  11. Parker S, Lipman J, Koulenti D, Dimopoulos G, Roberts JA. What is the relevance of fosfomycin pharmacokinetics in the treatment of serious infections in critically ill patients? A systematic review. Int J Antimicrob Agents. 2013;42:289–93.

    Article  CAS  PubMed  Google Scholar 

  12. Antonello RM, Di Bella S, Maraolo AE, Luzzati R. Fosfomycin in continuous or prolonged infusion for systemic bacterial infections: a systematic review of its dosing regimen proposal from in vitro, in vivo and clinical studies. Eur J Clin Microbiol Infect Dis. 2021;40:1117–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Mabilat C, Gros MF, Nicolau D, Mouton JW, Textoris J, Roberts JA, et al. Diagnostic and medical needs for therapeutic drug monitoring of antibiotics. Eur J Clin Microbiol Infect Dis. 2020;39:791–7.

    Article  PubMed  Google Scholar 

  14. Hong LT, Downes KJ, Fakhriravari A, Abdul-Mutakabbir JC, Kuti JL, Jorgensen S et al. International consensus recommendations for the use of prolonged-infusion beta-lactam antibiotics: Endorsed by the American College of Clinical Pharmacy, British Society for Antimicrobial Chemotherapy, Cystic Fibrosis Foundation, European Society of Clinical Microbiology and Infectious Diseases, Infectious Diseases Society of America, Society of Critical Care Medicine, and Society of Infectious Diseases Pharmacists. Pharmacotherapy: The Journal of Human Pharmacology and Drug Therapy. 2023;43:740–77.

  15. Abdul-Aziz MH, Alffenaar JWC, Bassetti M, Bracht H, Dimopoulos G, Marriott D, et al. Antimicrobial therapeutic drug monitoring in critically ill adult patients: a position paper. Intensive Care Med. 2020;46:1127–53.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Pea F, Viale P. Bench-to-bedside review: appropriate antibiotic therapy in severe sepsis and septic shock–does the dose matter? Crit Care. 2009;13:214.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Rinaldi M, Cojutti PG, Zamparini E, Tedeschi S, Rossi N, Conti M et al. Population Pharmacokinetics and Monte Carlo Simulation for Dosage Optimization of Fosfomycin in the Treatment of Osteoarticular Infections in patients without renal dysfunction. 2021.

  18. Cojutti PG, Fornaro G, Gatti M, Rinaldi M, Gaibani P, Giannella M, et al. Successful treatment of Bacteremia and Ventilator-Associated Pneumonia caused by KPC/OXA-48-like Klebsiella pneumoniae Co-producer with a continuous infusion of high-dose Meropenem Plus Fosfomycin guided by real-time therapeutic drug monitoring. Infect Dis Rep. 2022;14:88–92.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Gatti M, Virgili G, Cojutti PG, Gaibani P, Conti M, Sturiale C et al. Real-time optimization of pharmacodynamic target attainment at infection site during treatment of Post-neurosurgical Ventriculitis caused by Carbapenem-Resistant Gram Negatives with Ceftazidime-Avibactam-based regimens: a report of two cases. Microorganisms. 2022;10.

  20. Narayanasamy S, Nation RL, Mahony AA, Grayson ML, Kwong JC, Sherry NL et al. Cure of limb-threatening XDR Pseudomonas aeruginosa infection: combining genome sequencing, Therapeutic Drug Level Monitoring, and Surgical Debridement. Open Forum Infect Dis. 2020;8.

  21. Gatti M, Giannella M, Rinaldi M, Gaibani P, Viale P, Pea F. Pharmacokinetic/Pharmacodynamic Analysis of Continuous-Infusion Fosfomycin in Combination with Extended-Infusion Cefiderocol or Continuous-Infusion Ceftazidime-Avibactam in a Case Series of Difficult-to-Treat Resistant Pseudomonas aeruginosa Bloodstream Infections and/or Hospital-Acquired Pneumonia. Antibiotics 2022, Vol 11, Page 1739. 2022;11:1739.

  22. AIFA Agenzia Italiana del Farmaco. InfectoFos Riassunto delle caratteristiche del prodotto. 2019. https://farmaci.agenziafarmaco.gov.it/aifa/servlet/PdfDownloadServlet?pdfFileName=footer_003703_043646_RCP.pdf&sys=m0b1l3. Accessed 21 Mar 2024.

  23. Baldelli S, Cerea M, Mangioni D, Alagna L, Muscatello A, Bandera A, et al. Fosfomycin therapeutic drug monitoring in real-life: development and validation of a LC-MS/MS method on plasma samples. J Chemother. 2022;34:25–34.

    Article  CAS  PubMed  Google Scholar 

  24. Rodríguez-Gascón A, Canut-Blasco A. Deciphering pharmacokinetics and pharmacodynamics of fosfomycin. Revista Española De Quimioterapia. 2019;32(Suppl 1):19.

    PubMed  PubMed Central  Google Scholar 

  25. Edwina AE, Koch BCP, Muller AE, al Jalali V, Matzneller P, Zeitlinger M, et al. Population plasma and urine pharmacokinetics and the probability of target attainment of fosfomycin in healthy male volunteers. Eur J Clin Pharmacol. 2023;79:775–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Al Jalali V, Matzneller P, Wulkersdorfer B, Chou S, Bahmany S, Koch BCP et al. Clinical pharmacokinetics of Fosfomycin after continuous infusion compared with intermittent infusion: a randomized crossover study in healthy volunteers. Antimicrob Agents Chemother. 2021;65.

  27. Magiorakos AP, Srinivasan A, Carey RB, Carmeli Y, Falagas ME, Giske CG, et al. Multidrug-resistant, extensively drug-resistant and pandrug-resistant bacteria: an international expert proposal for interim standard definitions for acquired resistance. Clin Microbiol Infect. 2012;18:268–81.

    Article  CAS  PubMed  Google Scholar 

  28. European Medicines Agency (EMA). Annex III Amendments to relevant sections of the Product Information. https://www.ema.europa.eu/en/documents/referral/fosfomycin-article-31-referral-annex-iii_en.pdf. Accessed 28 Aug 2001.

  29. Cancer Institute N. Common Terminology Criteria for Adverse Events (CTCAE) Common Terminology Criteria for Adverse Events (CTCAE) v5.0. 2017.

  30. Harris PA, Taylor R, Minor BL, Elliott V, Fernandez M, O’Neal L, et al. The REDCap consortium: building an international community of software platform partners. J Biomed Inf. 2019;95:103208.

    Article  Google Scholar 

  31. Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. Research electronic data capture (REDCap)—A metadata-driven methodology and workflow process for providing translational research informatics support. J Biomed Inf. 2009;42:377–81.

    Article  Google Scholar 

  32. Spiegelman D, Hertzmark E. Easy SAS calculations for risk or prevalence ratios and differences. Am J Epidemiol. 2005;162:199–200.

    Article  PubMed  Google Scholar 

  33. Pontikis K, Karaiskos I, Bastani S, Dimopoulos G, Kalogirou M, Katsiari M, et al. Outcomes of critically ill intensive care unit patients treated with fosfomycin for infections due to pandrug-resistant and extensively drug-resistant carbapenemase-producing Gram-negative bacteria. Int J Antimicrob Agents. 2014;43:52–9.

    Article  CAS  PubMed  Google Scholar 

  34. Michalopoulos A, Virtzili S, Rafailidis P, Chalevelakis G, Damala M, Falagas ME. Intravenous fosfomycin for the treatment of nosocomial infections caused by carbapenem-resistant Klebsiella pneumoniae in critically ill patients: a prospective evaluation. Clin Microbiol Infect. 2010;16:184–6.

    Article  CAS  PubMed  Google Scholar 

  35. Del Río A, Gasch O, Moreno A, Peña C, Cuquet J, Soy D, et al. Efficacy and safety of fosfomycin plus imipenem as rescue therapy for complicated bacteremia and endocarditis due to methicillin-resistant Staphylococcus aureus: a multicenter clinical trial. Clin Infect Dis. 2014;59:1105–12.

    Article  PubMed  Google Scholar 

  36. Corti N, Sennhauser FH, Stauffer UG, Nadal D. Fosfomycin for the initial treatment of acute haematogenous osteomyelitis. Arch Dis Child. 2003;88:512–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Fitoussi F, Litzelmann E, Ilharreborde B, Morel E, Mazda K, Penneçot GF. Hematogenous osteomyelitis of the wrist in children. J Pediatr Orthop. 2007;27:810–3.

    Article  PubMed  Google Scholar 

  38. Pujol M, Miró JM, Shaw E, Aguado JM, Puig-Asensio M, Pigrau C, 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.

    Article  CAS  PubMed  Google Scholar 

  39. Apisarnthanarak A, Mundy LM. Carbapenem-resistant Pseudomonas aeruginosa pneumonia with intermediate minimum inhibitory concentrations to doripenem: combination therapy with high-dose, 4-h infusion of doripenem plus fosfomycin versus intravenous colistin plus fosfomycin. Int J Antimicrob Agents. 2012;39:271–2.

    Article  CAS  PubMed  Google Scholar 

  40. Mirakhur A, Gallagher MJ, Ledson MJ, Hart CA, Walshaw MJ. Fosfomycin therapy for multiresistant Pseudomonas aeruginosa in cystic fibrosis. J Cyst Fibros. 2003;2:19–24.

    Article  CAS  PubMed  Google Scholar 

  41. Stengel D, Görzer E, Schintler M, Legat FJ, Amann W, Pieber T, et al. Second-line treatment of limb-threatening diabetic foot infections with intravenous fosfomycin. J Chemother. 2005;17:527–35.

    Article  CAS  PubMed  Google Scholar 

  42. Dinh A, Salomon J, Bru JP, Bernard L. Fosfomycin: efficacy against infections caused by multidrug-resistant bacteria. Scand J Infect Dis. 2012;44:182–9.

    Article  CAS  PubMed  Google Scholar 

  43. Sirijatuphat R, Thamlikitkul V. Preliminary study of Colistin versus Colistin plus Fosfomycin for treatment of Carbapenem-Resistant Acinetobacter baumannii infections. Antimicrob Agents Chemother. 2014;58:5598–601.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Chand R, Chand R, Goldfarb DS. Hypernatremia in the intensive care unit. Curr Opin Nephrol Hypertens. 2022;31:199–204.

    Article  PubMed  Google Scholar 

  45. Grabein B, Graninger W, Rodríguez Baño J, Dinh A, Liesenfeld DB. Intravenous fosfomycin-back to the future. Systematic review and meta-analysis of the clinical literature. Clin Microbiol Infect. 2017;23:363–72.

    Article  CAS  PubMed  Google Scholar 

  46. Gatti M, Cojutti PG, Bartoletti M, Tonetti T, Bianchini A, Ramirez S, et al. Expert clinical pharmacological advice may make an antimicrobial TDM program for emerging candidates more clinically useful in tailoring therapy of critically ill patients. Crit Care. 2022;26:1–14.

    Article  Google Scholar 

  47. Lepak AJ, Zhao M, Vanscoy B, Taylor DS, Ellis-Grosse E, Ambrose PG et al. In vivo pharmacokinetics and pharmacodynamics of ZTI-01 (Fosfomycin for Injection) in the neutropenic murine thigh infection model against Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa. Antimicrob Agents Chemother. 2017;61.

  48. Parker SL, Frantzeskaki F, Wallis SC, Diakaki C, Giamarellou H, Koulenti D, et al. Population pharmacokinetics of fosfomycin in critically ill patients. Antimicrob Agents Chemother. 2015;59:6471–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. König C, Martens-Lobenhoffer J, Czorlich P, Westphal M, Bode-Böger SM, Kluge S, et al. Cerebrospinal fluid penetration of fosfomycin in patients with ventriculitis: an observational study. Ann Clin Microbiol Antimicrob. 2023;22:29.

    Article  PubMed  PubMed Central  Google Scholar 

  50. European Medicines Agency (EMA). Fosfomycin-containing medicinal products - referral | European Medicines Agency. https://www.ema.europa.eu/en/medicines/human/referrals/fosfomycin-containing-medicinal-products. Accessed 28 Aug 2001.

Download references

Acknowledgements

Preliminary results were presented at the European Congress of Clinical Microbiology and Infectious Diseases, 2023, Copenhagen, Denmark, 15-18 April 2023 (abstract no. 02745). We are grateful to Dr. Liliane Chatenoud for her supervision and support in statistical analysis.

Funding

This study was partially funded by the Italian Ministry of Health – Current Research IRCCS.

Author information

Authors and Affiliations

Authors

Contributions

DM, DC, AB were involved in conceptualization; CB ran statistical analysis; SBi, LM, LA, SBa, FB, CC, MC, FC, AG, GG, BM, AM, DC contributed to investigation; DM, SBi, CB, LM were involved in writing – original draft; LA, SBa, FC, MC, CC, FC, AG, GG, BM, AM, DC, AB contributed to writing – review & editing. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Davide Mangioni.

Ethics declarations

Ethics approval and consent to participate

The study was registered by the Milan Area 2 Ethical Committee (#664_2022bis) and was conducted in accordance with standards of the Helsinki Declaration. Informed consent for pseudonymized data processing for future research purposes was provided by all patients at the time hospital admission, as routine procedure. Specific written informed consent was waived because of the retrospective nature of the analysis.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Biscarini, S., Mangioni, D., Bobbio, C. et al. Adverse events during intravenous fosfomycin therapy in a real-life scenario. Risk factors and the potential role of therapeutic drug monitoring. BMC Infect Dis 24, 650 (2024). https://doi.org/10.1186/s12879-024-09541-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12879-024-09541-4

Keywords