Skip to main content
Erschienen in: European Journal of Clinical Microbiology & Infectious Diseases 6/2021

Open Access 07.01.2021 | Original Article

Serious adverse events with novel beta-lactam/beta-lactamase inhibitor combinations: a large-scale pharmacovigilance analysis

verfasst von: Milo Gatti, Emanuel Raschi, Fabrizio De Ponti

Erschienen in: European Journal of Clinical Microbiology & Infectious Diseases | Ausgabe 6/2021

Abstract

The purpose of this study is to characterize adverse events (AEs) of clinical interest reported with ceftolozane-tazobactam and ceftazidime-avibactam, as an aid in monitoring patients affected by severe multidrug-resistant Gram-negative infections. We queried the worldwide FDA Adverse Event Reporting System (FAERS) and performed disproportionality analysis, selecting only designated medical events (DMEs) where ceftolozane-tazobactam and ceftazidime-avibactam were reported as suspect. Serious neurological AEs were further investigated. The reporting odds ratios were calculated, deemed significant by the lower limit of the 95% confidence interval (LL95% CI) > 1. All other drugs/events recorded in FAERS and cephalosporins showing clinical evidence of neurological AEs were respectively selected as comparator for analysis of DMEs and neurotoxicity. Qualitative analysis including case-by-case assessment and deduplication was also performed. Overall, 654 and 506 reports mentioning respectively ceftolozane-tazobactam and ceftazidime-avibactam were found, with DMEs accounting respectively for 13.1% and 10.9% of cases. Agranulocytosis (N = 12; LL95% CI = 12.40) and pancytopenia (14; 6.18) emerged as unexpected AEs with ceftolozane-tazobactam, while acute pancreatitis (7; 8.63) was an over-reported unexpected DME with ceftazidime-avibactam. After deduplication, four unequivocally different cases of agranulocytosis with ceftolozane-tazobactam were retained, occurring on average after 8.8 days. Causality was probable and possible respectively in three and one case. Among neurological AEs exhibiting significant disproportionality, encephalopathy with both antibiotics and mental status changes with ceftazidime-avibactam were retained in at least three cases after deduplication. Although rare, clinicians should monitor high-risk patients (i.e. individuals affected by haematological malignances, HIV infection, or treated with concomitant myelotoxic agents) for early unexpected occurrence of agranulocytosis with ceftolozane-tazobactam.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s10096-020-04149-3.

Publisher’s note

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

Introduction

Ceftolozane-tazobactam (C/T) and ceftazidime-avibactam (C/A) are novel beta-lactam/beta-lactamase inhibitors (BL/BLIs), respectively approved by the Food and Drug Administration (FDA) in 2014 and in 2015, and by the European Medicines Agency (EMA) in 2015 and in 2016, characterized by activity against multidrug-resistant (MDR) Gram-negative pathogens, including extensively drug-resistant Pseudomonas aeruginosa (XDR-PA) and carbapenemase producing Enterobacteriaceae (CPE) [1].
Although the risk-benefit ratio is generally favourable in severe MDR infections, the safety profile of these antibiotics still requires thorough investigation. Specifically, no differences were found in serious adverse events (AEs) between novel BL/BLIs and controlled treatments in pivotal trials [2], being tolerability predictable and comparable to those seen with other beta-lactams, while real-world evidence is limited to the description of AEs reported with C/A [3]. However, given their expanding use at higher dosage in challenging scenarios, including patients affected by severe renal impairment [4], characterization of potential unexpected AEs with novel BL/BLIs becomes necessary.
The FDA Adverse Event Reporting System (FAERS) has attracted considerable interest among clinicians for accurate and timely characterization of drug-related risks occurring in real-world patients, also including the assessment of antimicrobials in challenging settings [57]. These post-marketing studies are particularly suited to early detect rare, unexpected and delayed AEs, which cannot be fully appreciated in pivotal trials, and are recommended for real-time safety assessment of recently marketed drugs.
We queried the FAERS database to characterize AEs of clinical interest reported with novel BL/BLIs, as an aid in prioritizing monitoring in patients affected by severe MDR Gram-negative infections.

Methods

An observational, retrospective disproportionality analysis was performed to highlight and characterize AEs of clinical interest with higher-than-expected reporting. The FAERS database (public dashboard), the US repository of AEs and medication errors comprising more than 20 million reports gathered worldwide, was queried to retrieve C/T and C/A reports recorded between the first quarter (Q1) of 2015 and Q2 of 2020.
In order to assign a clinical priority to emerging safety issues, the public list developed by the EMA including 62 different reactions was used to select designated medical events (DMEs), namely rare, serious AEs with a recognized drug-attributable risk, which may constitute a safety issue under certain circumstances (e.g. plausible causality with exclusion of alternative causes) [8].
Furthermore, given the non-negligible risk of neurotoxicity reported with cephalosporins [9], we searched clinical signs/symptoms, known as preferred terms (PTs) according to the Medical Dictionary for Regulatory Activities, in the following High Level Group Terms: “Seizures” (comprising 77 PTs), “Deliria” and “Hallucinations” (12 PTs each). Specific PTs concerning neurotoxicity (namely “encephalopathy”, “tremor”, “agitation”, “anxiety”, “cognitive disorder”, “mental impairment”, “altered state of consciousness”, “mental disorder”, “mental status changes”, “myoclonus”, “neurotoxicity”) were also analysed.
The reporting odds ratio (ROR) with relevant 95% confidence interval (CI) was calculated as a measure of disproportionality. All other drugs/events recorded in FAERS and cephalosporins showing clinical evidence of neurological AEs (namely cefazolin, ceftriaxone, cefixime, cefotiam, ceftazidime and cefepime according to the review of Sutter et al. [10]) were respectively selected as comparator for analysis of DMEs and neurotoxicity. Specifically, a case‑non-case approach was applied: Cases were defined by reports of the event of interest for C/T or C/A in which the drug was suspiciously recorded, while non-cases were represented by AE reports recorded for comparators. The ROR is the odds of exposure to C/T or C/A among the cases divided by the odds of exposure to C/T or C/A among the non-cases. If the proportion of the event of interest is greater in patients exposed to C/T or C/A (cases) than in patients exposed to all other drugs reported in FAERS, or for the selected cephalosporins (non-cases), a disproportionality signal emerges. Cases counted as many-fold as the number of DMEs or “neurological” events recorded in a given report. Traditional criteria for signal detections were used, i.e. lower limit of the 95% CI of the ROR > 1 with at least three cases of interest reported [11].
DMEs or neurological AEs emerging from disproportionality analysis were further scrutinized to better describe relevant clinical features: demographic information, reported indication, proportion of death, proportion of septic shock and/or multi-organ dysfunction syndrome and concomitant risk factors and/or drugs implicated in the events of interest (i.e. proportion of renal impairment or underlying nervous abnormalities for neurological events).
DMEs with disproportionality signal were also classified into three broad categories, according to the predictability of the reaction: (1) expected AEs, anticipated from pre-marketing pivotal trials; (2) disease-related AEs, for which underlying sepsis/septic shock represents per se a risk factor; and (3) unexpected AEs, on the basis of pharmacodynamic properties.
Unexpected DMEs and neurological AEs were further scrutinized to detect potential duplicates, based on overlapping data in six key fields, as previously performed [6]: event date, age, sex, reporter’s country, concomitant reactions and concomitant drugs. Records with at least five out of six overlaps were considered duplicates and excluded by case assessment.
Finally, unexpected DMEs with at least 3 unduplicated cases, a widely accepted signalling criterion [11], were further characterized by accessing original narratives submitted by the reporter through a Freedom of Information Act requested to the FDA: Information on medical history, laboratory findings, dechallenge/rechallenge, medical management and latency were used to assess causality according to the WHO system [12].

Results

Overall, 654 and 506 reports mentioning respectively C/T and C/A as suspect were found, of which 65.4% and 87.2% were serious. Subjects aged > 50 years old were the most represented, with slight male preponderance for both C/T and C/A (Supplementary Table 1).
DMEs were respectively reported in 86 (13.1%) and 55 cases (10.9%) with C/T and C/A; 12 and 13 DMEs were reported at least once, respectively. Disproportionality analysis was performed for 4 and 6 DMEs respectively with C/T and C/A (eight and seven AEs were reported in less than three cases; Supplementary Table 2). Increased reporting was found for acute kidney injury (N = 24; ROR 5.50; 95% CI 3.66–8.27), agranulocytosis (12; 21.96; 12.40–38.87), pancytopenia (14; 10.50; 6.18–17.82) and renal failure (27; 7.88; 5.36–11.58) with C/T. Acute kidney injury (16; 4.71; 2.86–7.76), anaphylactic shock (3; 6.85; 2.20–21.33), haemolytic anaemia (3; 11.56; 3.72–35.98), hepatic failure (4; 5.74; 2.15–15.36), acute pancreatitis (7; 18.19; 8.63–38.36) and renal failure (13; 4.82; 2.78–8.37) emerged as over-reported DMEs with C/A (Table 1).
Table 1
Designated medical events (DMEs) and selected neurological adverse events (AEs) reported with ceftolozane-tazobactam and ceftazidime-avibactam showing statistically significant disproportionality. Main clinical features after deduplication are also showed
DMEs or neurological AEs
No. of cases
ROR (95% CI)
Predictability
No. of cases after deduplication
Proportion of potential confounders*
Ceftolozane-tazobactam – DMEs
  Acute kidney injury
24
5.50 (3.66–8.27)
Expected
Disease-related
10
5 (50.0%)
  Agranulocytosis
12
21.96 (12.40–38.87)
Unexpected
4
1 (25.0%)
  Pancytopenia
14
10.50 (6.18–17.82)
Unexpected
2
1 (50.0%)
  Renal failure
27
7.88 (5.36–11.58)
Expected
Disease-related
12
6 (50.0%)
Ceftazidime-avibactam – DMEs
  Acute kidney injury
16
4.71 (2.86–7.76)
Expected
Disease-related
9
4 (44.4%)
  Anaphylactic shock
3
6.85 (2.20–21.33)
Expected
2
0 (0.0%)
  Haemolytic anaemia
3
11.56 (3.72–35.98)
Expected
3
0 (0.0%)
  Hepatic failure
4
5.74 (2.15–15.36)
Disease-related
3
0 (0.0%)
  Pancreatitis acute
7
18.19 (8.63–38.36)
Unexpected
2
2 (100.0%)
  Renal failure
13
4.82 (2.78–8.37)
Expected
Disease-related
8
6 (75.0%)
Ceftolozane-tazobactam – selected neurological AEs
  Encephalopathy
19
2.63 (1.66–4.19)
Expected
3
1 (33.3%)
  Epilepsy
11
3.61 (1.96–6.65)
Expected
1
0 (0.0%)
  Generalized tonic-clonic seizure
10
6.60 (3.43–12.70)
Expected
2
0 (0.0%)
  Status epilepticus
10
1.93 (1.03–3.65)
Expected
1
0 (0.0%)
Ceftazidime-avibactam – selected neurological AEs
  Encephalopathy
18
3.25 (2.01–5.24)
Expected
10
3 (30.0%)
  Mental status changes
8
4.04 (1.98–8.26)
Expected
7
3 (42.9%)
  Tonic convulsion
4
14.42 (4.99–41.71)
Expected
1
0 (0.0%)
AEs adverse events; DMEs designated medical events; ROR reporting odds ratio; CI confidence interval
*Concomitant drugs or underlying conditions potentially implicated in the specific adverse event
Agranulocytosis and pancytopenia with C/T and pancreatitis acute with C/A emerged as unexpected AEs. Overall, concomitant drugs or underlying conditions potentially confounding were retrieved in 45.5% of cases (Supplementary Table 3).
After deduplication, only two cases of pancytopenia with C/T and two cases of pancreatitis acute with C/A were found as unequivocally different. Narratives were requested for the four confirmed cases of agranulocytosis with C/T (Table 2). Patients were ≥ 70 years old with female preponderance (75.0%). Agranulocytosis occurred after an average of 8.8 days (range 4–17 days) following the beginning of C/T treatment. In two cases, potential confounders related to underlying conditions (i.e. HIV infection, acute leukaemia, lymphoma) or concomitant therapies (i.e. rituximab, allopurinol, trimethoprim-sulfamethoxazole, metimazole) were recorded. In three cases, C/T was withdrawn, and in two patients, the administration of granulocyte colony-stimulating factor was required. All events were serious, leading to prolonged hospitalization. Recovery and increased in neutrophil count occurred in all cases after approximately 5–10 days. Causality was probable in three cases and possible in one case.
Table 2
Narrative review of cases of agranulocytosis reported with ceftolozane-tazobactam and submitted to FAERS
Case ID
Age/sex
Reporter country
Dose/route of administration
Time onset (days)*
Medical history
Concomitant therapies
Acute medical condition
Laboratory and imaging findings
Specific treatment
Dechallenge/rechallenge
Outcome
Causality assessment
13844290
87/M
Spain
1 g q8h IV
17
High blood pressure, congestive heart failure, atrial fibrillation, chronic obstructive pulmonary disease (COPD) with oxygen therapy, insomnia, loss of personal independence in daily activities, arthralgia
Amiodarone
200 mg/day
Furosemide
40 mg q12h
Trazodone
100 mg/day
Tramadol
50 mg q8h
COPD decompensation due to LRTI caused by PA XDR
Neutrophil count:
2.96 × 109/L before treatment with C/T
1.33 × 109/L at the moment of C/T withdrawn
5.45 × 109/L at recovering
C/T withdrawn 1 day after the occurrence of agranulocytosis
Progressive increase in leucocyte count after C/T withdrawn
Yes/No
Prolonged hospitalization
Recovered after 6 days
Probable
(reasonable time relationship with drug intake; unlikely role of other diseases/drugs; positive dechallenge)
14034547
79/F
France
LD 1 g IV
MD 250 mg q8h IV
Undergoing haemodialysis
8
Atrial fibrillation, pulmonary embolism
Amiodarone
Fluindione
cUTI with bacteraemia caused by PA MDR
Before treatment with C/T
Neutrophil count:
1.76 G/L
White cell count:
2.84 G/L
During C/T treatment
Neutrophil count:
0.13 G/L
White cell count:
2.61 G/L
Osteomedullar biopsy:
decrease of the granulocytic density at 10%, no impairment of other cell lines. Suspected toxic cause.
At recovering
Neutrophil count:
6.13 G/L
White cell count:
8.29 G/L
C/T withdrawn 1 day after occurrence of agranulocytosis
Switch to ceftazidime-avibactam and administration of filgrastim
Progressive increase in leucocytes and neutrophil count after C/T withdrawn
Yes/No
Prolonged hospitalization
Recovered
Probable
(osteomedullar biopsy suggestive for toxic cause; reasonable time relationship with drug intake; unlikely role of other diseases/drugs; positive dechallenge)
15505937
71/F
Portugal
3 g q8h IV
4
NHL with medullar invasion
Mature B cell type acute leukaemia undergoing induction chemotherapy
Controlled HIV infection
Fluconazole
200 mg/day
Tigecycline
100 mg q12h
Metamizole**
Rituximab**
TMP-SMX**
Allopurinol**
Pneumonia caused by XDR PA
Repeated myelogram: no presence of lymphoblast
C/T continuation
Administration of growth factors with no benefit
No/No
Prolonged hospitalization
Absolute neutropenia for 10 days with refractoriness to growth factors. Haemoglobin and platelet recovery was maintained.
Recovered
Possible
(concomitant drugs/diseases potentially explaining the event; dechallenge not performed)
15659483
70/F
Portugal
1.5 g q8h IV
6
HIV infection
Depressive symptom
Lymphoma
Vertigo
Abacavir/Lamivudine
600/300 mg/day
Tigecycline
100 mg/day
Nevirapine
400 mg/day
Allopurinol***
300 mg/day
TMP-SMX*** 800/160 mg q8h
Sepsis
NA
C/T withdrawn 5 days after the occurrence of agranulocytosis
Yes/No
Prolonged hospitalization
Life-threatening
Recovered/resolved
Probable
(reasonable time relationship with drug intake; unlikely role of other diseases/drugs: no worsening of HIV infection; no ongoing treatment for lymphoma; allopurinol and TMP-SMX prescribed 45 days before the event; positive dechallenge)
COPD chronic obstructive pulmonary disease; LRTI lower tract respiratory infection; PA Pseudomonas aeruginosa; XDR extensively drug-resistant; MDR multidrug-resistant; C/T ceftolozane-tazobactam; cUTI complicated urinary tract infection; NHL non-Hodgkin’s lymphoma; TMP-SMX cotrimoxazole; NA not available
*After ceftolozane-tazobactam administration
**The indicated drugs were previously administered to the patient before admission without experience of agranulocytosis
***Prescribed 45 days before occurrence of agranulocytosis
Overall, 78 (11.9%) and 71 (14.0%) neurological AEs were respectively found with C/T and C/A. Disproportionality analysis was performed for six and seven AEs respectively with C/T and C/A. Compared to selected cephalosporins, encephalopathy (19; 2.63; 1.66–4.19), epilepsy (10; 6.60; 3.43–12.70), generalized tonic-clonic seizure (11; 3.61; 1.96–6.65) and status epilepticus (10; 1.93; 1.03–3.65) reported with C/T exhibited significant ROR. Increased reporting was found for encephalopathy (18; 3.25; 2.01–5.24), mental status changes (8; 4.04; 1.98–8.26) and tonic convulsion (4; 14.42; 4.99–41.71) with C/A (Table 1; Supplementary Table 4).
After deduplication, only encephalopathy with both BL and BLIs and mental status changes with C/A were confirmed in at least three cases. Overall, concomitant renal impairment was found in 28.0% of cases, while no underlying nervous abnormalities were recognized (Supplementary Table 5).

Discussion

To the best of our knowledge, this is the first large-scale study reporting serious AEs with novel BL/BLIs. Haematological reactions emerged as unexpected life-threatening AEs for C/T, while C/A exhibited a predictable safety profile according to anticipated common AEs found in pivotal trials (i.e. anaphylactic shock, haemolytic anaemia) and expected clinical complications of severe infections caused by CPE (i.e. septic shock with hepatic/renal failure). Furthermore, an over-reporting of different serious neurological AEs (namely encephalopathy and mental status changes) compared to other cephalosporins [10] in patients receiving C/T or C/A was found.
Notably, signals of unexpected pancytopenia and agranulocytosis with C/T, detected by disproportionality, were refuted by our qualitative analysis, including concomitant use of different agents (namely linezolid, ganciclovir, valacyclovir and trimethoprim-sulfamethoxazole) known to cause myelotoxicity. Therefore, possible confounders responsible for synergic toxicity may be implicated in the occurrence of pancytopenia. Conversely, in the four retained cases of agranulocytosis with C/T, the limited presence of concomitant risk factors together with causality assessment suggests a true safety issue. Although currently no case reports or preclinical evidence of ceftolozane-induced agranulocytosis exist, the immune-mediated hypothesis may reasonably explain the occurrence of this unexpected AE with C/T, as reported for other agents, including beta-lactams [13, 14]. However, a dose-dependent direct toxic effect on granulocytopoiesis cannot be excluded [15]. Additionally, elderly and female gender are recognized risk factors for drug-induced agranulocytosis [13]. Notably, mean age of our cases was approximately 77 years with female preponderance (75.0%). Furthermore, the mean onset time of our cases (8.8 days) overlaps with agranulocytosis reported in 12 patients with piperacillin-tazobactam (17.6 days) and in 62 subjects treated with different beta-lactams (16 days) [14, 15], consistently with the immune-mediated hypothesis. Although in two of our cases potential confounders (underlying diseases and/or concomitant myelotoxic drugs) were found, all these agents were previously administered for several cycles without occurrence of agranulocytosis, nor was any previous event of neutropenia noted in patients affected by leukaemia or lymphoma. Notably, in three cases, a probable causal association was detected, and none was classified as unlikely. Given the non-negligible proportion of XDR-PA infections in haematological patients [16], C/T may assume a leading role in this scenario. Consequently, the over-reporting of agranulocytosis calls for clinical monitoring in patients treated with C/T. Particularly, assessment of host-dependent risk factors (i.e. elderly, haematological malignances, HIV infection, concomitant myelotoxic agents) coupled with intensive blood cell count monitoring (every 24 h during the entire treatment with C/T) should be implemented (Fig. 1).
The two unduplicated cases of acute pancreatitis with C/A deserve attention for the following reasons: In the literature, no case of definite or probable association was found for ceftazidime [17]; in our reports, concomitant use of tigecycline, known to cause pancreatitis [18], was noted. Although C/A could not be considered the only primary suspect, considering its wide use in combination with tigecycline in complicated intraabdominal infections caused by CPE [19], clinicians should evaluate the risk of pancreatitis on a case-by-case basis in critically ill patients treated with drug combination, possibly implementing additional laboratory (i.e. serum amylase/lipase) or imaging monitoring (Fig. 1).
Our analysis found an over-reporting of different serious neurological AEs with novel BL/BLIs compared to selected cephalosporins. Although neurotoxicity is a well-known safety concern in susceptible patients with risk factors (i.e. elderly, renal impairment, underlying nervous abnormalities) [9, 10], the higher reporting with novel BL/BLIs, rather than reflecting an intrinsically higher risk related to these agents per se, may be the result of a channelling bias favoured by their well-established use at standard/higher dosage to improve efficacy in severe MDR infections in critical settings [4]. To this regard, although C/A includes a maximum dose of ceftazidime (namely 6 g/day) for subject with normal renal function, novel BL/BLIs are commonly used in real-world scenarios at higher than recommended dosage also in patients with severe renal impairment or requiring continuous renal replacement therapy (CRRT), in order to overcome pharmacokinetic issues usually observed in critically ill subjects (e.g. wide increase in volume of distribution, high intensity CRRT, prompt recovery of renal function in the first 48–72 h), and achieve aggressive PK/PD targets directly associated with better clinical outcome and prevention of resistance development [4, 2022]. However, in this scenario, it could be possible that toxic serum concentrations are more likely achieved, thus leading to a greater risk of neurological AEs. Adaptive therapeutic drug monitoring of novel BL/BLIs could be useful, retaining steady-state concentrations below tenfold the minimum inhibitory concentration (Fig. 1) [23].
Notwithstanding the well-known limitations of our approach (e.g. potential reporting biases, including under-reporting phenomenon, lack of exposure data and clinical details, inability in establishing firm causality between drug exposure and occurrence of AEs, possible remaining duplicates), agranulocytosis emerged as an early and unexpected, albeit rare, AEs with C/T, thus calling for both stringent clinical monitoring in high-risk patients (namely those affected by haematological malignancies, HIV infection, acute or chronic kidney injury, or treated with concomitant myelotoxic or nephrotoxic agents) affected by severe MDR Gram-negative infections, and further observational studies to better characterize this safety aspect.

Compliance with ethical standards

Conflict of interest

The authors declare that they have no conflict of interest.

Ethics approval

Under current legislation, institutional review board approval is not required when performing analysis of the publicly available FAERS database, because it contains anonymized data that cannot allow patients’ identification.
Not applicable.
Not applicable.

Code availability

Not applicable.
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/​.

Publisher’s note

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Koulenti D, Song A, Ellingboe A, Abdul-Aziz MH, Harris P, Gavey E, Lipman J (2019) Infections by multidrug-resistant Gram-negative Bacteria: what’s new in our arsenal and what’s in the pipeline? Int J Antimicrob Agents. 53(3):211–224CrossRef Koulenti D, Song A, Ellingboe A, Abdul-Aziz MH, Harris P, Gavey E, Lipman J (2019) Infections by multidrug-resistant Gram-negative Bacteria: what’s new in our arsenal and what’s in the pipeline? Int J Antimicrob Agents. 53(3):211–224CrossRef
2.
Zurück zum Zitat Chen M, Zhang M, Huang P, Lin Q, Sun C, Zeng H, Deng Y (2018) Novel β-lactam/β-lactamase inhibitors versus alternative antibiotics for the treatment of complicated intra-abdominal infection and complicated urinary tract infection: a meta-analysis of randomized controlled trials. Expert Rev Anti Infect Ther. 16(2):111–120CrossRef Chen M, Zhang M, Huang P, Lin Q, Sun C, Zeng H, Deng Y (2018) Novel β-lactam/β-lactamase inhibitors versus alternative antibiotics for the treatment of complicated intra-abdominal infection and complicated urinary tract infection: a meta-analysis of randomized controlled trials. Expert Rev Anti Infect Ther. 16(2):111–120CrossRef
3.
Zurück zum Zitat Gatti M, Raschi E, De Ponti F (2019) Relationship between adverse drug reactions to antibacterial agents and the Klebsiella pneumoniae carbapenemase-producing (KPC) Klebsiella pneumoniae outbreak: insight from a pharmacovigilance study. BMC Pharmacol Toxicol. 20, 65(1) Gatti M, Raschi E, De Ponti F (2019) Relationship between adverse drug reactions to antibacterial agents and the Klebsiella pneumoniae carbapenemase-producing (KPC) Klebsiella pneumoniae outbreak: insight from a pharmacovigilance study. BMC Pharmacol Toxicol. 20, 65(1)
4.
Zurück zum Zitat Giannella M, Bartoletti M, Gatti M, Viale P (2020) Advances in the therapy of bacterial bloodstream infections. Clin Microbiol Infect. 26(2):158–167CrossRef Giannella M, Bartoletti M, Gatti M, Viale P (2020) Advances in the therapy of bacterial bloodstream infections. Clin Microbiol Infect. 26(2):158–167CrossRef
6.
Zurück zum Zitat Poluzzi E, Raschi E, Motola D, Moretti U, De Ponti F (2010) Antimicrobials and the risk of torsades de pointes: the contribution from data mining of the US FDA Adverse Event Reporting System. Drug Saf. 33(4):303–314CrossRef Poluzzi E, Raschi E, Motola D, Moretti U, De Ponti F (2010) Antimicrobials and the risk of torsades de pointes: the contribution from data mining of the US FDA Adverse Event Reporting System. Drug Saf. 33(4):303–314CrossRef
7.
Zurück zum Zitat Patek TM, Teng C, Kennedy KE, Alvarez CA, Frei CR (2020) Comparing acute kidney injury reports among antibiotics: a pharmacovigilance study of the FDA Adverse Event Reporting System (FAERS). Drug Saf. 43(1):17–22CrossRef Patek TM, Teng C, Kennedy KE, Alvarez CA, Frei CR (2020) Comparing acute kidney injury reports among antibiotics: a pharmacovigilance study of the FDA Adverse Event Reporting System (FAERS). Drug Saf. 43(1):17–22CrossRef
9.
Zurück zum Zitat Deshayes S, Coquerel A, Verdon R (2017) Neurological adverse effects attributable to β-lactam antibiotics: a literature review. Drug Saf. 40(12):1171–1198CrossRef Deshayes S, Coquerel A, Verdon R (2017) Neurological adverse effects attributable to β-lactam antibiotics: a literature review. Drug Saf. 40(12):1171–1198CrossRef
10.
Zurück zum Zitat Sutter R, Rüegg S, Tschudin-Sutter S (2015) Seizures as adverse events of antibiotic drugs: a systematic review. Neurology. 85(15):1332–1341CrossRef Sutter R, Rüegg S, Tschudin-Sutter S (2015) Seizures as adverse events of antibiotic drugs: a systematic review. Neurology. 85(15):1332–1341CrossRef
11.
Zurück zum Zitat Raschi E, Poluzzi E, Salvo F et al (2018) Pharmacovigilance of sodium-glucose co-transporter-2 inhibitors: what a clinician should know on disproportionality analysis of spontaneous reporting systems. Nutr Metab Cardiovasc Dis. 28(6):533–542CrossRef Raschi E, Poluzzi E, Salvo F et al (2018) Pharmacovigilance of sodium-glucose co-transporter-2 inhibitors: what a clinician should know on disproportionality analysis of spontaneous reporting systems. Nutr Metab Cardiovasc Dis. 28(6):533–542CrossRef
13.
Zurück zum Zitat Lorenzo-Villalba N, Alonso-Ortiz MB, Maouche Y, Zulfiqar AA, Andrès E (2020) Idiosyncratic drug-induced neutropenia and agranulocytosis in elderly patients. J Clin Med. 9(6):1808CrossRef Lorenzo-Villalba N, Alonso-Ortiz MB, Maouche Y, Zulfiqar AA, Andrès E (2020) Idiosyncratic drug-induced neutropenia and agranulocytosis in elderly patients. J Clin Med. 9(6):1808CrossRef
15.
Zurück zum Zitat Vial T, Bailly H, Perault-Pochat MC, Default A, Boulay C, Chouchana L, Kassai B (2019) French Network of Pharmacovigilance Centres. Beta-lactam-induced severe neutropaenia: a descriptive study. Fundam Clin Pharmacol. 33(2):225–231CrossRef Vial T, Bailly H, Perault-Pochat MC, Default A, Boulay C, Chouchana L, Kassai B (2019) French Network of Pharmacovigilance Centres. Beta-lactam-induced severe neutropaenia: a descriptive study. Fundam Clin Pharmacol. 33(2):225–231CrossRef
16.
Zurück zum Zitat Willmann M, Bezdan D, Zapata L, Susak H, Vogel W, Schröppel K, Liese J, Weidenmaier C, Autenrieth IB, Ossowski S, Peter S (2015) Analysis of a long-term outbreak of XDR Pseudomonas aeruginosa: a molecular epidemiological study. J Antimicrob Chemother. 70(5):1322–1330CrossRef Willmann M, Bezdan D, Zapata L, Susak H, Vogel W, Schröppel K, Liese J, Weidenmaier C, Autenrieth IB, Ossowski S, Peter S (2015) Analysis of a long-term outbreak of XDR Pseudomonas aeruginosa: a molecular epidemiological study. J Antimicrob Chemother. 70(5):1322–1330CrossRef
17.
Zurück zum Zitat Nitsche C, Maertin S, Scheiber J, Ritter CA, Lerch MM, Mayerle J (2012) Drug-induced pancreatitis. Curr Gastroenterol Rep. 14(2):131–138CrossRef Nitsche C, Maertin S, Scheiber J, Ritter CA, Lerch MM, Mayerle J (2012) Drug-induced pancreatitis. Curr Gastroenterol Rep. 14(2):131–138CrossRef
18.
Zurück zum Zitat McGovern PC, Wible M, Korth-Bradley JM, Quintana A (2014) Pancreatitis in tigecycline Phase 3 and 4 clinical studies. J Antimicrob Chemother. 69(3):773–778CrossRef McGovern PC, Wible M, Korth-Bradley JM, Quintana A (2014) Pancreatitis in tigecycline Phase 3 and 4 clinical studies. J Antimicrob Chemother. 69(3):773–778CrossRef
19.
Zurück zum Zitat Rodríguez-Baño J, Gutiérrez-Gutiérrez B, Machuca I, Pascual A (2018) Treatment of infections caused by extended-spectrum-beta-lactamase-, AmpC-, and carbapenemase-producing Enterobacteriaceae. Clin Microbiol Rev. 31(2):e00079–e00017CrossRef Rodríguez-Baño J, Gutiérrez-Gutiérrez B, Machuca I, Pascual A (2018) Treatment of infections caused by extended-spectrum-beta-lactamase-, AmpC-, and carbapenemase-producing Enterobacteriaceae. Clin Microbiol Rev. 31(2):e00079–e00017CrossRef
20.
Zurück zum Zitat Crass RL, Rodvold KA, Mueller BA, Pai MP (2019) Renal dosing of antibiotics: are we jumping the gun? Clin Infect Dis. 68(9):1596–1602CrossRef Crass RL, Rodvold KA, Mueller BA, Pai MP (2019) Renal dosing of antibiotics: are we jumping the gun? Clin Infect Dis. 68(9):1596–1602CrossRef
21.
Zurück zum Zitat Gatti M, Giannella M, Raschi E, Viale P, De Ponti F (2021) Ceftolozane/tazobactam exposure in critically ill patients undergoing continuous renal replacement therapy: a PK/PD approach to tailor dosing. J Antimicrob Chemother. 76(1):199–205CrossRef Gatti M, Giannella M, Raschi E, Viale P, De Ponti F (2021) Ceftolozane/tazobactam exposure in critically ill patients undergoing continuous renal replacement therapy: a PK/PD approach to tailor dosing. J Antimicrob Chemother. 76(1):199–205CrossRef
22.
Zurück zum Zitat Sumi CD, Heffernan AJ, Lipman J, Roberts JA, Sime FB (2019) What antibiotic exposures are required to suppress the emergence of resistance for Gram-negative bacteria? A Systematic Review. Clin Pharmacokinet. 58(11):1407–1443CrossRef Sumi CD, Heffernan AJ, Lipman J, Roberts JA, Sime FB (2019) What antibiotic exposures are required to suppress the emergence of resistance for Gram-negative bacteria? A Systematic Review. Clin Pharmacokinet. 58(11):1407–1443CrossRef
23.
Zurück zum Zitat Abdul-Aziz MH, Alffenaar JC, Bassetti M, Bracht H, Dimopoulos G, Marriott D, Neely MN, Paiva JA, Pea F, Sjovall F, Timsit JF, Udy AA, Wicha SG, Zeitlinger M, De Waele JJ, Roberts JA (2020) Infection Section of European Society of Intensive Care Medicine (ESICM); Pharmacokinetic/pharmacodynamic and Critically Ill Patient Study Groups of European Society of Clinical Microbiology and Infectious Diseases (ESCMID); Infectious Diseases Group of International Association of Therapeutic Drug Monitoring and Clinical Toxicology (IATDMCT); Infections in the ICU and Sepsis Working Group of International Society of Antimicrobial Chemotherapy (ISAC). Antimicrobial therapeutic drug monitoring in critically ill adult patients: a Position Paper. Intensive Care Med. 46(6):1127–1153CrossRef Abdul-Aziz MH, Alffenaar JC, Bassetti M, Bracht H, Dimopoulos G, Marriott D, Neely MN, Paiva JA, Pea F, Sjovall F, Timsit JF, Udy AA, Wicha SG, Zeitlinger M, De Waele JJ, Roberts JA (2020) Infection Section of European Society of Intensive Care Medicine (ESICM); Pharmacokinetic/pharmacodynamic and Critically Ill Patient Study Groups of European Society of Clinical Microbiology and Infectious Diseases (ESCMID); Infectious Diseases Group of International Association of Therapeutic Drug Monitoring and Clinical Toxicology (IATDMCT); Infections in the ICU and Sepsis Working Group of International Society of Antimicrobial Chemotherapy (ISAC). Antimicrobial therapeutic drug monitoring in critically ill adult patients: a Position Paper. Intensive Care Med. 46(6):1127–1153CrossRef
Metadaten
Titel
Serious adverse events with novel beta-lactam/beta-lactamase inhibitor combinations: a large-scale pharmacovigilance analysis
verfasst von
Milo Gatti
Emanuel Raschi
Fabrizio De Ponti
Publikationsdatum
07.01.2021
Verlag
Springer Berlin Heidelberg
Erschienen in
European Journal of Clinical Microbiology & Infectious Diseases / Ausgabe 6/2021
Print ISSN: 0934-9723
Elektronische ISSN: 1435-4373
DOI
https://doi.org/10.1007/s10096-020-04149-3

Weitere Artikel der Ausgabe 6/2021

European Journal of Clinical Microbiology & Infectious Diseases 6/2021 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

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