Skip to main content
Erschienen in: BMC Infectious Diseases 1/2022

Open Access 01.12.2022 | Research article

Multi-body-site colonization screening cultures for predicting multi-drug resistant Gram-negative and Gram-positive bacteremia in hematological patients

verfasst von: Ignacio Torres, Dixie Huntley, Mar Tormo, Marisa Calabuig, Juan Carlos Hernández-Boluda, María José Terol, Carlos Carretero, Paula de Michelena, Ariadna Pérez, José Luis Piñana, Javier Colomina, Carlos Solano, David Navarro

Erschienen in: BMC Infectious Diseases | Ausgabe 1/2022

Abstract

Background

To investigate the multi-drug resistant bacteria (MDRB) colonization rate in hematological patients hospitalized for any cause using a multi-body-site surveillance approach, and determine the extent to which this screening strategy helped anticipate MDRB bloodstream infections (BSI).

Methods

Single-center retrospective observational study including 361 admissions documented in 250 adult patients. Surveillance cultures of nasal, pharyngeal, axillary and rectal specimens (the latter two combined) were performed at admission and subsequently on a weekly basis. Blood culture samples were incubated in an automated continuous monitoring blood culturing instrument (BACTEC FX).

Results

In total, 3463 surveillance cultures were performed (pharyngeal, n = 1201; axillary-rectal, n = 1200; nasal, n = 1062). MDRB colonization was documented in 122 out of 361 (33.7%) admissions corresponding to 86 patients (34.4%). A total of 149 MDRB were isolated from one or more body sites, of which most were Gram-negative bacteria, most frequently non-fermenting (n = 83) followed by Enterobacterales (n = 51). BSI were documented in 102 admissions (28%) involving 87 patients. Overall, the rate of BSI caused by MDRB was significantly higher (p = 0.04) in the presence of colonizing MDRB (16 out of 47 admissions in 14 patients) than in its absence (9 out of 55 admissions in 9 patients). Colonization by any MDRB was independently associated with increased risk of MDRB-BSI (HR, 3.70; 95% CI, 1.38–9.90; p = 0.009).

Conclusion

MDRB colonization is a frequent event in hematological patients hospitalized for any reason and is associated with an increased risk of MDRB BSI. The data lend support to the use of MDRB colonization surveillance cultures for predicting the occurrence of MDRB BSI in this cohort.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AMR
Antimicrobial resistance
BHI
Brain–heart infusion broth
BSI
Bloodstream infections
ESBL
Extended-spectrum beta-lactamase
EUCAST
European Committee on Antimicrobial Susceptibility Testing
HR
Hazard ratio
ICU
Intensive care unit
MALDI-ToF–MS
Matrix-assisted laser desorption/ionization time-of-flight mass spectroscopy
MDRB
Multi-drug resistant bacteria
MIC
Minimum inhibitory concentration
MRSA
Methicillin-resistant Staphylococcus aureus
OR
Odss ratio
VIM
Verona integron-encoded metallo-beta-lactamase
VRE
Vancomycin-resistant enterococci

Background

Bloodstream infections (BSI) caused by multidrug-resistant bacteria (MDRB), particularly those involving extended-spectrum beta-lactamase (ESBL)- or carbapenemase-producing Enterobacterales, MDR Pseudomonas aeruginosa, and vancomycin-resistant enterococci (VRE), pose a major threat for patients with hematological malignancies undergoing chemotherapy or transplantation due to their inherent associated morbidity and mortality [18]. Hematological patients are at increased risk of MRDB BSI as a result of extensive broad-spectrum antibiotics use, prolonged hospitalizations, and disruption of mucosal surfaces and neutropenia due to cytotoxic chemotherapy [13]. Identifying MDRB-colonized patients through active surveillance could facilitate appropriate or early adjustment of empirical therapy for BSI, yet data supporting this assumption have been inconsistent [917].
Nose and rectum are the preferred sites for surveillance of methicillin-resistant Staphylococcus aureus (MRSA) and VRE/MDR Gram-negative bacteria, respectively [1823]; nonetheless, there is no consensus regarding anatomical sites to be sampled for MDR Gram-negative bacteria screening, microbiological methods for the purpose, or the benefit of performing screening cultures targeting certain MDR bacteria (i.e. MDR-Stenotrophomonas maltophilia) [18, 24]. This was highlighted in a recent study revealing striking differences across Spanish centers in sampling protocols, body sites chosen for screening and microbiological testing procedures [25]. The purpose of the current study was twofold. First, we investigated the MDRB colonization rate in a series of hematological patients hospitalized for any cause using a multi-body-site surveillance approach and identified risk factors for this event. Second, we ascertained the incidence of MDRB BSI in patients with or without previous detection of MDRB colonization and determined whether the above screening strategy permitted anticipation of its occurrence.

Methods

Study population

In this single-center retrospective observational study we included a total of 361 admissions to the hematology wards from January 2015 to December 2019, involving 250 adult (> 18 years) hematological patients. No exclusion criteria were established. Twenty-nine patients were eventually admitted to the Intensive Care Unit (ICU). The cohort comprised patients with the following underlying diseases: lymphoma (n = 93), acute myeloid leukemia (n = 69), multiple myeloma (n = 45), acute lymphoblastic leukemia (n = 16), myelodysplastic syndrome (n = 11), chronic lymphocytic leukemia (n = 2), chronic myeloid leukemia (n = 1) and others (n = 13). Reasons for hospital admission were receipt of allogeneic or autologous stem cell transplantation (n = 163), treatment of underlying disease (n = 136), neutropenic fever (n = 35), performance of diagnostic procedures (n = 9) acute or chronic graft versus host disease (n = 7), and other causes (n = 11). For patients with two or more admissions (n = 59), these took place at least 3 months apart. Colonization by the same one or more MDRB in two or more consecutive admissions for a given patient was considered as a single episode. Median hospital stay was 25 days (range, 4–96). Baseline characteristics, clinical data, recent antibiotic treatment and clinical outcomes were registered.

Microbiological analyses

During patient hospitalization, surveillance cultures of nasal, pharyngeal, axillary and rectal specimens were scheduled to be performed systematically within 48 h of admission (baseline) and on a weekly basis afterwards, as a part of routine patient care and according to predefined protocols, as detailed below. All patients colonized with MDRB underwent contact isolation. Surveillance specimens were delivered in AMIES transport medium (cliniswabLTS, Aptaca Spa., Canelli, Italy) to the Microbiology Service and immediately processed in accordance with the Procedures in Clinical Microbiology guidelines issued by the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC) [26]. Swabs were placed in brain–heart infusion broth tubes (BHI; Oxoid Limited, Hampshire, UK) containing a disc of either cefotaxime (30 µg) or imipenem (10 µg), for MDR-Gram negative enrichment and incubated at 37 °C in a CO2 incubator (Heracell 240i CO2 incubator, Thermo Fisher Scientific, Langenselbold, Germany) for 24 h. Pharyngeal and nasal swabs were processed individually, while axillary and rectal swabs were combined, as previous data from our group indicated that this strategy yielded comparable results to those obtained by processing both of these specimens individually (not shown) and resulted in lower cost burden. Specimens were subcultured on MacConkey Agar (Becton Dickinson, New Jersey, USA) in which imipenem (10 µg), cefotaxime (30 µg) and ceftazidime (30 µg) discs (Oxoid Limited, Hampshire, UK) were placed and Columbia Blood Agar with 5% Sheep Blood (Becton Dickinson) in which oxacillin (1 μg) and 30 μg vancomycin discs (Oxoid Limited) were placed. Isolated colonies growing near the discs were identified using Matrix-Assisted Laser Desorption/Ionization time-of-flight Mass spectroscopy -MALDI-TOF MS- (Bruker Daltonics, MA, USA). Conventional antimicrobial susceptibility testing from isolated bacteria was performed by broth microdilution using the MicroScan NM44 panel for Gram-negative bacteria and PM33 for Gram-positive bacteria, both from Beckman Coulter (Brea, CA, USA), and interpreted according to contemporary European Committee on Antimicrobial Susceptibility Testing (EUCAST) guidelines https://​www.​eucast.​org/​fileadmin/​src/​media/​PDFs/​EUCAST_​files/​Breakpoint_​tables/​v_​10.​0_​breakpoint_​Tables.​pdf.). The Antimicrobial Resistance (AMR) Direct Flow Chip (Máster Diagnóstica, Granada, Spain), DNA microarray-based assay was used for antimicrobial resistance gene characterization from bacterial isolated colonies, as described [28] and was performed either contemporarily or retrospectively.
Blood culture samples (BACTEC Plus Aerobic/F and Plus Anaerobic/F medium bottles, Becton Dickinson—BD—and Company, New Jersey, USA) were collected from patients with suspected bacteremia and incubated in an automated continuous monitoring blood culturing instrument (BACTEC FX; BD). Aliquots from each positive BC bottle were subjected to routine Gram stain microscopy, subcultured on chocolate blood medium (BD) and incubated for 24–48 h. From 2018 onwards, direct bacterial identification from BCs was performed by MALDI-TOF MS testing using intact bacteria, as previously described [27].

Definitions

MDRB was defined as resistance to one or more agents in three or more antimicrobial categories, as previously recommended [29]. The MDRB targeted in screening cultures were ESBL-carbapenemase- and plasmidic AmpC-producing Enterobacterales, MDR-P. aeruginosa, MDR-S. maltophilia, MDR-Acinetobacter spp. MRSA and VRE. A febrile neutropenia episode was defined as a single oral temperature of ≥ 38.3 °C or a temperature of ≥ 38.0 °C (100.4°F) sustained over 1 hour with an absolute neutrophil count < 500 cells/mm3 [1]. Administration of any systemic antibiotic within one month prior to admission was considered prior antibiotic therapy [1, 12]. MDRB colonization was defined as the detection of the respective organism in at least one surveillance culture (from any site). Empirical antibiotic therapy (antibiotic administration without prior identification of the causative bacteria) was initiated at physician discretion according to local guidelines, which take into consideration several factors including baseline risk for severe infection (i.e. neutropenia), site of infection and previous records of MDRB colonization. MDR Gram-negative bacteria were covered by administering beta-lactam antibiotics with antipseudomonal activity (usually, piperacillin-tazobactam or carbapenems) either in monotherapy or in combination with aminoglycosides. When appropriate, MDR Gram positive bacteria were covered by adding to the aforementioned regimens either vancomycin, linezolid or daptomycin. Similarity between susceptibility antimicrobial profile and genotypic resistant marker pattern among isolates recovered from blood and surveillance cultures was deemed to indicate bacterial identity.

Statistical analysis

Frequency comparisons for categorical variables were carried out using the Fisher exact test or the Chi-square test when appropriate. A P-value < 0.05 was considered statistically significant. Odds ratios (OR) and hazard ratios (HR) were determined by Cox and logistic regression analyses, respectively. For multivariate analyses, only variables with parameter estimates showing a P value ≤ 0.10 in univariate analyses were included; two-sided P-values < 0.05 were deemed to be significant. Analyses were performed using SPSS version 25.0 (SPSS, Chicago, IL, USA).

Results

Colonization by MRDB in hematological patients

A total of 3463 surveillance cultures (pharyngeal, n = 1201; axillary-rectal, n = 1200; nasal, n = 1062) were performed during the study period in hematological patients included in the study. MDRB colonization was documented in 122 out of 361 (33.7%) admissions corresponding to 86 out of 250 patients (34.4%), of whom 75 were hospitalized at the hematology ward (111 out of 332 admissions; 33.4%) and 11 at ICU (11 out of 29 admissions; 37.9%).
A total of 149 MDRB were isolated from one or more body sites (Table 1), of which most were Gram-negative bacteria, most frequently non-fermenting (n = 83) followed by Enterobacterales (n = 51). MDR Gram-positive bacteria (MRSA or VRE-Enterococcus faecium with vancomycin and teicoplanin MICs > 16 mg/L) were isolated in 15 admissions. In most cases, MDRB were cultured from a single body site (82/122; 67.2%), irrespective of the hospitalization ward (68.5% in hematology and 54.5% in ICU).
Table 1
Multi-drug resistant bacteria (MDRB) isolated from surveillance colonization cultures
Any MDRB
149 (100)
Gram-negative bacteria
134 (89.9)
 Enterobacterales
51 (34.2)
  ESBL-Escherichia coli
36 (24.1)
  Plasmidic AmpC Escherichia coli
5 (3.4)
  ESBL-Klebsiella pneumoniae
6 (4)
  Plasmidic AmpC Klebsiella pneumoniae
2 (1.3)
  ESBL-Enterobacter cloacae
1 (0.7)
  Class B carbapenemase (VIM type) Klebsiella pneumoniae
1 (0.7)
Non-fermenting Gram-negative bacteria
83 (55.7)
 MDR-Pseudomonas aeruginosa
37 (24.8)
 Class B carbapenemase (VIM type) Pseudomonas aeruginosa
16 (10.8)
 MDR-Acinetobacter spp.
3 (2)
 MDR-Stenotrophomonas maltophilia
27 (18.1)
Gram-positive bacteria
15 (10.1)
 MRSA
6 (4)
 VRE
9 (6.1)
ESBL extended spectrum β-lactamase, MDR multidrug-resistant, MRSA methicillin-resistant Staphylococcus aureus, VRE vancomycin-resistant enterococci
When considering all hospital admissions, one or more colonizing MDRB were present in 47 out of 329 available baseline specimens (14.2%) corresponding to 36 patients: ESBL-producing Enterobacterales (n = 22), MDR-P. aeruginosa (n = 10), MDR-Stenotrophomonas maltophilia (n = 8), MRSA (n = 4) and others (n = 8).
Interestingly, among patients with more than one hospital admission (n = 59), 14 tested negative during the first hospital stay and became colonized during the second one. All these patients received broad-spectrum antimicrobial therapy during the first hospital admission period. Nevertheless, the median length of hospital stay in first admissions was not significantly different (P = 0.27) between those who became colonized in subsequent admission periods (31 days; range, 6–64) and those who did not (27 days; range, 18–92).
Overall, recovery of any MDRB was consistently more likely from axillary-rectal specimens than from pharyngeal or nasal specimens, at both the hematology ward and ICU, as shown in Table 2. ESBL-producing Enterobacterales (specially E. coli) and MDR-P. aeruginosa represented the commonest MDRB recovered from both axillary-rectal specimens and pharyngeal specimens. MDR-S. maltophilia was cultured more frequently from pharyngeal than from axillary-rectal specimens. VRE were recovered at the same rate from axillary-rectal and pharyngeal specimens.
Table 2
Multi-drug resistant bacteria (MDRB) isolated from surveillance colonization cultures
MDRB
Specimen from which MDRB were isolated in patients admitted to the hematology ward/Intensive care unit
 
Pharyngeal, ward, no. (%) / ICU, no. (%)
Nasal, ward, no. (%) / ICU, no. (%)
Axillary-rectal, ward, no. (%) / ICU, no. (%)
Any MDRB
59 (17.7) / 6 (20.6)
13 (3.9) / 4 (13.7)
97 (29.2) / 9 (31.0)
Gram-negative bacteria
54 (16.2) / 6 (20.6)
7 (2.1) / 4 (13.7)
89 (26.8) / 9 (31.0)
 Enterobacterales
11 (3.3) / 1 (3.4)
0 (0) / 2 (6.8)
42 (12.6) / 6 (20.6)
  ESBL-Escherichia coli
7 (2.1) / 1 (3.4)
0 (0) / 1 (3.4)
30 (9.0) / 5 (26.2)
  Plasmidic AmpC Escherichia coli
0 (0) / 0 (0)
0 (0)
4 (1.2) / 1 (11.1)
  ESBL-Klebsiella pneumoniae
3 (0.9) / 0 (0)
0 (0)
6 (1.8) / 0 (0)
  Plasmidic AmpC Klebsiella pneumoniae
0 (0) / 0 (0)
0 (0)
2 (0.6) / 0 (0)
  ESBL-Enterobacter cloacae
1 (0.3) / 0 (0)
0 (0)
0 (0) / 0 (0)
  Class B carbapenemase (VIM type) Klebsiella pneumoniae
0 (0) / 0 (0)
0 (0) / 1 (3.4)
0 (0) / 0 (0)
Non-fermenting Gram-negative bacteria
43 (12.9) / 5 (17.2)
7 (2.1) / 2 (6.8)
47 (14.1) / 3 (10.3)
 MDR-Pseudomonas aeruginosa
17 (5.1) / 3 (10.3)
3 (0.9) / 0 (0)
24 (7.1) / 2 (6.8)
 Class B carbapenemase (VIM type) Pseudomonas aeruginosa
6 (1.8) / 0 (0)
0 (0) / 0 (0)
15 (4.5) / 0 (0)
 MDR-Acinetobacter spp
1 (0.3) / 0 (0)
1 (0.3) / 0 (0)
1 (0.3) / 0 (0)
 MDR-Stenotrophomonas maltophilia
19 (5.7) / 2 (6.8)
3 (0.9) / 2 (6.8)
7 (2.1) / 1 (3.4)
Gram-positive bacteria
5 (1.5) / 0 (0)
6 (1.8) / 0 (0)
8 (2.4) / 0 (0)
 MRSA
0 (0) / 0 (0)
6 (1.8) / 0 (0)
0 (0) / 0 (0)
 VRE
5 (1.5) / 0 (0)
0 (0) / 0 (0)
8 (2.4) / 0 (0)
ESBL extended spectrum β-lactamase, MDR multidrug-resistant, MRSA methicillin-resistant Staphylococcus aureus, VRE vancomycin-resistant enterococci
Surveillance of pharyngeal and nasal sites enabled documentation of colonization by one or more MDRB (n = 41) that were absent from axillary-rectal cultures in 31 out of the 122 (25.4%) admissions (Table 3). In most cases, these were MDR-non-fermenting Gram-negative bacteria, in particular MDR-P. aeruginosa and MDR-S. maltophilia recovered from nasal and/or pharyngeal specimens. In contrast, screening of pharyngeal and nasal sites seldom increased the detection rate of MDR-Enterobacterales or VRE colonization provided by axillary-rectal specimens. As expected, nasal site screening allowed recovery of MRSA which could not be cultured from the other specimen types.
Table 3
Multi-drug resistant bacteria (MDRB) isolated from pharyngeal, nasal specimens or both and missed by axillary-rectal surveillance cultures
MDRB
Specimen from which MDRB were isolated
 
Pharyngeal, no
Nasal, no
Any MDRB
33
13
Gram-negative bacteria
32
7
 Enterobacterales
  
  ESBL-Escherichia coli
2
0
  ESBL-Enterobacter cloacae
1
0
  Class B carbapenemase (VIM type) Klebsiella pneumoniae
0
0
Non-fermenting
  
 MDR-Pseudomonas aeruginosa
11
1
 Class B carbapenemase (VIM type) Pseudomonas aeruginosa
1
0
 MDR-Acinetobacter spp.
1
1
 MDR-Stenotrophomonas maltophilia
16
4
Gram-positive bacteria
1
6
 MRSA
0
6
 VRE
1
0
ESBL extended spectrum β-lactamase, MDR multidrug-resistant, MRSA methicillin-resistant Staphylococcus aureus, VRE vancomycin-resistant enterococci

BSI in patients colonized with MDRB

As depicted in Fig. 1, BSI were documented in 102 admissions (28%) from 87 patients, of which 98 were monomicrobial and 4 polymicrobial. Therefore, a total of 107 isolates were recovered from BC (Table 4). There were 47 BSI episodes (39 patients) among 122 hospitalizations in which colonizing MDRB were identified in the last surveillance cultures processed prior to BSI detection (within 2–7 days), and 55 (52 patients) among 239 admissions in which colonizing MDRB were not documented (p = 0.002). Overall, the rate of BSI caused by MDRB was significantly higher in the presence of colonizing MDRB (16 out of 47, in 14 patients) than in its absence (9 out of 55, in 9 patients) (p = 0.04). Out of the 16 BSI occurring in MDRB-colonized patients, 13 (in 11 patients) were deemed to be due to the colonizing isolate (ESBL-producing E. coli, n = 5; plasmidic AmpC-producing E. coli, n = 2; MDR P. aeruginosa, n = 2; class B carbapenemase (VIM type)-producing P. aeruginosa, n = 2; ESBL-producing K. pneumoniae, n = 1; VRE, n = 1). Colonization by any MDRB was associated with an increased risk of MDRB BSI (HR, 3.70; 95% CI, 1.38–9.90; p = 0.009) in multivariate models adjusted for age, sex, underlying hematological disease, receipt of transplant and previous antibiotic treatments. Colonization by MDRB had a positive and negative predictive value of 68.5% and 64% for the occurrence of MDRB-BSI, respectively. The 13 colonizing MDRB causing BSI could be recovered from one or more body sites (Table 5). Interestingly, all but one of these isolates (MDR-P. aeruginosa) were cultured from axillary-rectal screening cultures.
Table 4
Bacteria isolated from blood cultures
All isolates (%)
107
Gram-negative bacteria
46 (43)
 Enterobacterales
36 (33.7)
  Citrobacter freundii
1 (0.9)
  Escherichia coli
20 (18.8)
  Klebsiella oxytoca
1 (0.9)
  Serratia marcescens
1 (0.9)
  ESBL-E. coli
9 (8.5)
  ESBL-K. pneumoniae
1 (0.9)
  Plasmidic AmpC E. coli
3 (2.8)
 Non-fermenting
10 (9.3)
  Non MDR-Pseudomonas aeruginosa
3 (2.8)
  MDR-Pseudomonas aeruginosa
3 (2.8)
  Class B carbapenemase (VIM type) Pseudomonas aeruginosa
3 (2.8)
  MDR-Stenotrophomonas maltophilia
1 (0.9)
Gram-positive bacteria
58 (54.2)
 CNS
22 (20.5)
 MR-CNS
10 (9.3)
 MRSA
4 (3.7)
 Enterococcus spp.
19 (17.9)
 VRE
1 (0.9)
 Streptococcus spp.
2 (1.9)
Other microorganisms
3 (2.8)
 Candida spp.
2 (1.9)
 C. perfringens
1 (0.9)
ESBL extended spectrum β-lactamase, MDR multidrug-resistant, CNS coagulase-negative Staphylococcus spp, MR-CNS methicillin-resistant coagulase-negative Staphylococcus spp, MRSA methicillin-resistant Staphylococcus aureus, VRE vancomycin-resistant enterococci
Table 5
Multi-drug resistant bacteria (MDRB) isolated from surveillance colonization cultures causing bloodstream infection
MDRB
Specimen from which MDRB were isolated
 
Pharyngeal, no. (%)
Nasal, no. (%)
Axillary-rectal, no. (%)
Any MDRB
5 (100)
1 (100)
12 (100)
Gram-negative bacteria
5 (100)
1 (100)
11 (91.7)
 Enterobacterales
2 (40)
1 (100)
8 (66.7)
  ESBL-Escherichia coli
1 (20)
1 (100)
5 (41.7)
  Plasmidic AmpC Escherichia
0 (0)
0 (0)
2 (16.7)
  ESBL-Klebsiella pneumoniae
1 (20)
0 (0)
1 (8.3)
Non-fermenting Gram-negative bacteria
3 (60)
0 (0)
3 (25)
 MDR-Pseudomonas aeruginosa
1 (20)
0 (0)
1 (8.3)
 Class B carbapenemase (VIM type) Pseudomonas aeruginosa
2 (40)
0 (0)
2 (16.7)
Gram-positive bacteria
0 (0)
0 (0)
1 (8.3)
 VRE
0 (0)
0 (0)
1 (8.3)
ESBL extended spectrum β-lactamase, MDR multidrug-resistant, VRE vancomycin-resistant enterococci

Risk factors for MDRB colonization in hematological patients

We next investigated whether MDRB colonization was associated with any demographic or clinical characteristic of patients during hospital admission (Table 6). Frequency comparison analyses revealed statistically significant differences between colonized and non-colonized cases when comparing sex (p = 0.005), underlying hematological malignancy (p = 0.002), with increased MDRB colonization rate among patients with acute myeloid leukemia, and whether the patient had undergone hematopoietic stem cell transplantation (p = 0.025). A trend towards a higher MDRB colonization rate was seen in older patients (p = 0.06) and those who had been treated with antibiotics within the month prior to admission (p = 0.07). None of these factors except for sex and acute myeloid leukemia as the underlying disease were independently associated with MDRB colonization (Table 7).
Table 6
Risk factors for multi-drug bacteria (MDRB) colonization during hospital admissions
Demographic parameters
No MDRB colonization no. (%)
MDRB colonization n (%)
P value
Agea
   
 ≤ 58
140 (58.6)
59 (48.4)
0.06
 > 58
99 (41.4)
63 (51.6)
 
Sex
   
 Male
130 (54.4)
85 (69.7)
0.005
 Female
109 (45.6)
37 (30.3)
 
Diagnosis
  
0.002
 AA
1 (0.4)
0 (0)
 
 Lymphoma
82 (34.3)
20 (16.4)
 
 ALL
21 (8.8)
8 (6.6)
 
 CLL/LPD
2 (0.8)
0 (0)
 
 AML
81 (33.9)
68 (55.7)
 
 CML
0
1 (0.8)
 
 MM
33 (13.8)
13 (10.7)
 
 MDS
9 (3.8)
5 (4.1)
 
 Other
10 (4.2)
7 (5.7)
 
Urinary catheter
  
0.64
 No
218 (91.2)
113 (92.6)
 
 Yes
21 (8.8)
9 (7.4)
 
Chemotherapy
  
0.15
 No
40 (16.7)
28 (23)
 
 Yes
199 (83.3)
94 (77)
 
Peripheral or central catheter
  
0.98
 No
2 (0.8)
1 (0.8)
 
 Yes
237 (99.2)
121 (99.2)
 
Previous antibiotic treatment
  
0.07
 No
164 (68.6)
72 (59)
 
 Yes
75 (31.4)
50 (41)
 
Allogeneic or autologous hematopoietic stem cell transplantation
  
0.02
 No
90 (37.7)
61 (50)
 
 Yes
149 (62.3)
61 (50)
 
AA aplastic anemia, ALL acute lymphoblastic leukemia, CLL/LPD chronic lymphocytic leukemia/lymphoproliferative disorders, AML acute myeloid leukemia, CML chronic myeloid leukemia, MM multiple myeloma, MDS myelodysplastic syndrome
aMedian age of patients at admission
Table 7
Risk factors for multi-drug resistant bacteria (MDRB) colonization during hospital admission: logistic regression analysis
Parameter
Univariate
Multivariate
 
OR
CI 95%
P-value
OR
CI 95%
P-value
Age (> 58 vs. ≤ 58)
1.51
0.97–2.341
0.06
1.45
0.91–2.29
0.11
Sex (male vs. female)
1.92
1.21–3.05
0.005
1.69
1.04–2.74
0.03
Acute myeloid leukemia (yes vs. no)
2.45
1.57–3.83
 < 0.001
2.3
1.32–4.00
0.03
Previous antibiotic treatment (yes vs. no)
1.51
0.96–2.38
0.07
1.35
0.84–2.19
0.21
Transplant (yes vs. no)
1.65
1.06–2.57
0.02
0.92
0.52–1.63
0.79
OR odds ratio

Discussion

Several factors mainly including broad-spectrum antibiotics use and prolonged hospital stays pose hematological patients at increased risk of MRDB colonization [13]. In turn, disruption of mucosal surfaces and neutropenia due to cytotoxic chemotherapy or transplantation favor the occurrence of MDRB BSI, which are associated with high morbidity and mortality [18]. In this study, we used a multi-body site screening strategy to investigate the rate of MDRB colonization in hematological patients admitted to the hospital for any cause (most frequently receipt of allogeneic or autologous stem cell transplantation, treatment of underlying disease or neutropenic fever) and to what extent this approach allowed us to predict MDRB BSI. Detection of genotypic determinants of antimicrobial resistance in MDRB was performed using a DNA-based microarray, which displays results that fully agree with genome sequencing data [26]. In line with previous studies [30, 31], to increase the rate of detection of MDRB we performed a combination of upper respiratory tract, gastrointestinal tract and axillar sampling. We documented colonization by one or more MDRB, mostly ESBL-producing Enterobacterales and MDR P. aeruginosa (including VIM-type carbapenemase producers) and S. maltophilia, in 33.7% of admissions, corresponding to 34.4% of patients in this series. Although MDRB colonization was more frequently documented during hospital stay, at either hematology ward or ICU, a non-negligible percentage of patients (14% of admissions) were identified as being colonized at baseline. Of note, colonization by carbapenemase-producing Enterobacterales and VRE was anecdotal, as expected considering the very low prevalence of these MDRB in our setting (not shown). Direct comparison of the current study with others addressing this same issue [917] is simply unfeasible for a number of reasons, including differences across studies in patients characteristics, local epidemiology particularities and notably targeted MDRBs, screening strategy and methodology used for MDRB detection, all of which may impact dramatically on results. Regarding the latter, for example, we sampled the upper respiratory tract (pharyngeal and nasal specimens), axillary and rectal sites and combined rectal and axillary specimens, while others used only rectal specimens for surveillance. Although MDRB recovery was more likely from axillary-rectal specimens than from upper respiratory tract samples (in this context, nasal swabs were the most unrewarding specimen for detecting colonization with MDR Gram-negative bacteria) specially MDR Enterobacterales, surveillance of pharyngeal and nasal sites enabled us to document MDRB colonization, mostly by non-fermenting Gram-negative rods, that went undetected in axillary-rectal cultures in 25.4% of admissions.
Sex (male), underlying hematological malignancy (acute myeloid leukemia) and transplantation were associated with increased risk of MDRB colonization in univariate analysis, while a trend towards significance was observed in classically associated factors such as age and antibiotics treatment within the month prior to admission [13]. The impact of sex and acute myeloid leukemia on increasing the risk of colonization by MDRB has been previously reported in particular for carbapenemase-producing Enterobacterales [10, 14, 3235].
In our view, the key observation of the current study was that overall, the MDRB-based BSI rate was significantly higher in MDRB-colonized patients than those who were not (p = 0.04). Moreover, among the 16 MDRB BSI episodes developing in MDRB-colonized patients, 13 were deemed to be caused by the colonizing isolate, principally ESBL-producing Enterobacterales and MDR P. aeruginosa. Indeed, MDRB colonization was independently associated with increased risk of MDRB BSI. Of these 13 isolates, 12 could be recovered from axillary-rectal cultures, while one (MDR-P. aeruginosa) was cultured from a pharyngeal specimen, but missed in axillary-rectal screening cultures. MDRB surveillance cultures, notably those from axillary-rectal sites, therefore offered valuable information to predict the antimicrobial susceptibility profile of BSI-causing MDRB in 81% of episodes. Our data concur to some extent with findings of several studies. Cattaneo and colleagues [12] conducted a multicenter prospective observational study involving 18 hematologic centers during a 6-month period; overall, 37 MDRB-colonized patients (25.7%) developed at least one BSI, of which 23 (16% of the whole series) developed BSI from the same MDRB pathogen with a rate of 15.6% for ESBL-producing Enterobacterales and 14.1% for carbapenemase-producing Gram-negative bacteria. The study concluded that empiric antibiotic treatment selection should be guided by known colonization in hematologic patients. Likewise, data from a study by Jaiswal et al. [10] demonstrated that hematologic patients colonized with carbapenemase-producing Enterobacterales have the highest risk of MDRB-BSI and mortality, particularly those with acute myeloid leukemia. In turn, Ballo et al. [11] highlighted the importance of exhaustive MDRB screening in acute myeloid patients with febrile neutropenia, as they require special surveillance due to their high risk of invasive infection and mortality. Liss et al. [13] found that colonization with ESBL-producing Enterobacterales was associated with increased risk ratios towards BSI development due to ESBL-producing Enterobacterales. Finally, Sadowska-Klasa et al. [17] retrospectively analyzed the data of 120 patients who underwent HCT for hematologic disorders and found that prior colonization was significantly correlated with MDRB infections (p < 0.001), especially bacteremia (p = 0.038).
The main limitation of the study is the relatively low number of registered MDRB BSI episodes. Nonetheless, the multi-body-site surveillance approach and MDRB pre-enrichment using selective broths, both of which may increase sensitivity for MDRB detection, can be considered strengths.

Conclusion

In summary, our data indicated that MDRB colonization in a highly heterogeneous cohort of hematological patients is a frequent event associated with increased risk of MDRB BSI. Thus, MDRB colonization screening cultures may be useful for predicting MDRB BSI and tailoring empirical antibiotic treatments on an individual basis. The data also suggested that upper respiratory tract sampling for surveillance cultures adds little to axillary-rectal swabbing only in terms of predicting MDR Gram-negative BSI development.

Acknowledgements

Ignacio Torres holds a Río Hortega Contract (CM20/00090) from the Carlos III Health Institute.

Declarations

The current study was approved by the Institutional review Board (IRB) Research Ethics Committee of Hospital Clínico Universitario INCLIVA (September, 2019). A waiver of documentation of informed consent was issued by the IRB.
Not applicable.

Competing interests

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

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Gudiol C, Bodro M, Simonetti A, et al. Changing aetiology, clinical features, antimicrobial resistance, and outcomes of bloodstream infection in neutropenic cancer patients. Clin Microbiol Infect. 2013;19(5):474–9.CrossRefPubMed Gudiol C, Bodro M, Simonetti A, et al. Changing aetiology, clinical features, antimicrobial resistance, and outcomes of bloodstream infection in neutropenic cancer patients. Clin Microbiol Infect. 2013;19(5):474–9.CrossRefPubMed
2.
Zurück zum Zitat Satlin MJ, Jenkins SG, Walsh TJ. The global challenge of carbapenem-resistant Enterobacteriaceae in transplant recipients and patients with hematologic malignancies. Clin Infect Dis. 2014;58(9):1274–83.CrossRefPubMedPubMedCentral Satlin MJ, Jenkins SG, Walsh TJ. The global challenge of carbapenem-resistant Enterobacteriaceae in transplant recipients and patients with hematologic malignancies. Clin Infect Dis. 2014;58(9):1274–83.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Gudiol C, Tubau F, Calatayud L, Garcia-Vidal C, Cisnal M, Sanchez-Ortega I, et al. Bacteraemia due to multidrug-resistant Gram-negative bacilli in cancer patients: risk factors, antibiotic therapy and outcomes. J Antimicrob Chemother. 2011;66(3):657–63.CrossRefPubMed Gudiol C, Tubau F, Calatayud L, Garcia-Vidal C, Cisnal M, Sanchez-Ortega I, et al. Bacteraemia due to multidrug-resistant Gram-negative bacilli in cancer patients: risk factors, antibiotic therapy and outcomes. J Antimicrob Chemother. 2011;66(3):657–63.CrossRefPubMed
4.
Zurück zum Zitat Tumbarello M, Viale P, Viscoli C, Trecarichi EM, Tumietto F, Marchese A, Spanu T, Ambretti S, Ginocchio F, Cristini F, Losito AR, Tedeschi S, Cauda R, Bassetti M. Predictors of mortality in bloodstream infections caused by Klebsiella pneumoniae carbapenemase-producing K. pneumoniae: importance of combination therapy. Clin Infect Dis. 2012;55(7):943–50.CrossRefPubMed Tumbarello M, Viale P, Viscoli C, Trecarichi EM, Tumietto F, Marchese A, Spanu T, Ambretti S, Ginocchio F, Cristini F, Losito AR, Tedeschi S, Cauda R, Bassetti M. Predictors of mortality in bloodstream infections caused by Klebsiella pneumoniae carbapenemase-producing K. pneumoniae: importance of combination therapy. Clin Infect Dis. 2012;55(7):943–50.CrossRefPubMed
5.
Zurück zum Zitat Trecarichi EM, Tumbarello M. Antimicrobial-resistant Gram-negative bacteria in febrile neutropenic patients with cancer: current epidemiology and clinical impact. Curr Opin Infect Dis. 2014;27(2):200–10.CrossRefPubMed Trecarichi EM, Tumbarello M. Antimicrobial-resistant Gram-negative bacteria in febrile neutropenic patients with cancer: current epidemiology and clinical impact. Curr Opin Infect Dis. 2014;27(2):200–10.CrossRefPubMed
6.
Zurück zum Zitat Girmenia C, Rossolini GM, Piciocchi A, et al. Infections by carbapenem-resistant Klebsiella pneumoniae in SCT recipients: a nationwide retrospective survey from Italy. Bone Marrow Transplant. 2015;50(2):282–8.CrossRefPubMed Girmenia C, Rossolini GM, Piciocchi A, et al. Infections by carbapenem-resistant Klebsiella pneumoniae in SCT recipients: a nationwide retrospective survey from Italy. Bone Marrow Transplant. 2015;50(2):282–8.CrossRefPubMed
7.
Zurück zum Zitat Lalaoui R, Javelle E, Bakour S, Ubeda C, Rolain JM. Infections due to carbapenem-resistant bacteria in patients with hematologic malignancies. Front Microbiol. 2020;17(11):1422.CrossRef Lalaoui R, Javelle E, Bakour S, Ubeda C, Rolain JM. Infections due to carbapenem-resistant bacteria in patients with hematologic malignancies. Front Microbiol. 2020;17(11):1422.CrossRef
8.
Zurück zum Zitat Weinstock DM, Conlon M, Iovino C, Aubrey T, Gudiol C, Riedel E, Young JW, Kiehn TE, Zuccotti G. Colonization, bloodstream infection, and mortality caused by vancomycin-resistant enterococcus early after allogeneic hematopoietic stem cell transplant. Biol Blood Marrow Transplant. 2007;13(5):615–21.CrossRefPubMed Weinstock DM, Conlon M, Iovino C, Aubrey T, Gudiol C, Riedel E, Young JW, Kiehn TE, Zuccotti G. Colonization, bloodstream infection, and mortality caused by vancomycin-resistant enterococcus early after allogeneic hematopoietic stem cell transplant. Biol Blood Marrow Transplant. 2007;13(5):615–21.CrossRefPubMed
9.
Zurück zum Zitat Pagano L, Caira M, Trecarichi EM, Spanu T, Di Blasi R, Sica S, Sanguinetti M, Tumbarello M. Carbapenemase-producing Klebsiella pneumoniae and hematologic malignancies. Emerg Infect Dis. 2014;20(7):1235–6.CrossRefPubMedPubMedCentral Pagano L, Caira M, Trecarichi EM, Spanu T, Di Blasi R, Sica S, Sanguinetti M, Tumbarello M. Carbapenemase-producing Klebsiella pneumoniae and hematologic malignancies. Emerg Infect Dis. 2014;20(7):1235–6.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Jaiswal SR, Gupta S, Kumar RS, Sherawat A, Rajoreya A, Dash SK, et al. Gut colonization with carbapenem-resistant Enterobacteriaceae adversely impacts the outcome in patients with hematological malignancies: results of a prospective surveillance study. Mediterr J Hematol Infect Dis. 2018;10(1): e2018025.CrossRefPubMedPubMedCentral Jaiswal SR, Gupta S, Kumar RS, Sherawat A, Rajoreya A, Dash SK, et al. Gut colonization with carbapenem-resistant Enterobacteriaceae adversely impacts the outcome in patients with hematological malignancies: results of a prospective surveillance study. Mediterr J Hematol Infect Dis. 2018;10(1): e2018025.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Ballo O, Tarazzit I, Stratmann J, Reinheimer C, Hogardt M, Wichelhaus TA, Kempf V, Serve H, Finkelmeier F, Brandts C. Colonization with multidrug resistant organisms determines the clinical course of patients with acute myeloid leukemia undergoing intensive induction chemotherapy. PLoS ONE. 2019;14(1): e0210991.CrossRefPubMedPubMedCentral Ballo O, Tarazzit I, Stratmann J, Reinheimer C, Hogardt M, Wichelhaus TA, Kempf V, Serve H, Finkelmeier F, Brandts C. Colonization with multidrug resistant organisms determines the clinical course of patients with acute myeloid leukemia undergoing intensive induction chemotherapy. PLoS ONE. 2019;14(1): e0210991.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Cattaneo C, Di Blasi R, Skert C, Candoni A, Martino B, Di Renzo N, Delia M, Ballanti S, Marchesi F, Mancini V, Orciuolo E, Cesaro S, Prezioso L, Fanci R, Nadali G, Chierichini A, Facchini L, Picardi M, Malagola M, Orlando V, Trecarichi EM, Tumbarello M, Aversa F, Rossi G, Pagano L. Bloodstream infections in haematological cancer patients colonized by multidrug-resistant bacteria. Ann Hematol. 2018;97(9):1717–26.CrossRefPubMed Cattaneo C, Di Blasi R, Skert C, Candoni A, Martino B, Di Renzo N, Delia M, Ballanti S, Marchesi F, Mancini V, Orciuolo E, Cesaro S, Prezioso L, Fanci R, Nadali G, Chierichini A, Facchini L, Picardi M, Malagola M, Orlando V, Trecarichi EM, Tumbarello M, Aversa F, Rossi G, Pagano L. Bloodstream infections in haematological cancer patients colonized by multidrug-resistant bacteria. Ann Hematol. 2018;97(9):1717–26.CrossRefPubMed
13.
Zurück zum Zitat Liss BJ, Vehreschild JJ, Cornely OA, Hallek M, Fatkenheuer G, Wisplinghoff H, et al. Intestinal colonisation and blood stream infections due to vancomycin-resistant enterococci (VRE) and extended-spectrum beta-lactamase-producing Enterobacteriaceae (ESBLE) in patients with haematological and oncological malignancies. Infection. 2012;40(6):613–9 (Epub 2012/06/06).CrossRefPubMed Liss BJ, Vehreschild JJ, Cornely OA, Hallek M, Fatkenheuer G, Wisplinghoff H, et al. Intestinal colonisation and blood stream infections due to vancomycin-resistant enterococci (VRE) and extended-spectrum beta-lactamase-producing Enterobacteriaceae (ESBLE) in patients with haematological and oncological malignancies. Infection. 2012;40(6):613–9 (Epub 2012/06/06).CrossRefPubMed
14.
Zurück zum Zitat Kömürcü B, Tükenmez Tigen E, Toptaş T, Fıratlı Tuğlular T, Koten V. Korten V Rectal colonization with multidrug-resistant gram-negative bacteria in patients with hematological malignancies: a prospective study. Expert Rev Hematol. 2020;13(8):923–7.CrossRefPubMed Kömürcü B, Tükenmez Tigen E, Toptaş T, Fıratlı Tuğlular T, Koten V. Korten V Rectal colonization with multidrug-resistant gram-negative bacteria in patients with hematological malignancies: a prospective study. Expert Rev Hematol. 2020;13(8):923–7.CrossRefPubMed
15.
Zurück zum Zitat Vehreschild MJ, Hamprecht A, Peterson L, Schubert S, Häntschel M, Peter S, Schafhausen P, Rohde H, Lilienfeld-Toal MV, Bekeredjian-Ding I, Libam J, Hellmich M, Vehreschild JJ, Cornely OA, Seifert H. A multicentre cohort study on colonization and infection with ESBL-producing Enterobacteriaceae in high-risk patients with haematological malignancies. J Antimicrob Chemother. 2014;69(12):3387–92.CrossRefPubMed Vehreschild MJ, Hamprecht A, Peterson L, Schubert S, Häntschel M, Peter S, Schafhausen P, Rohde H, Lilienfeld-Toal MV, Bekeredjian-Ding I, Libam J, Hellmich M, Vehreschild JJ, Cornely OA, Seifert H. A multicentre cohort study on colonization and infection with ESBL-producing Enterobacteriaceae in high-risk patients with haematological malignancies. J Antimicrob Chemother. 2014;69(12):3387–92.CrossRefPubMed
17.
Zurück zum Zitat Sadowska-Klasa A, Piekarska A, Prejzner W, Bieniaszewska M, Hellmann A. Colonization with multidrug-resistant bacteria increases the risk of complications and a fatal outcome after allogeneic hematopoietic cell transplantation. Ann Hematol. 2018;97(3):509–17.CrossRefPubMed Sadowska-Klasa A, Piekarska A, Prejzner W, Bieniaszewska M, Hellmann A. Colonization with multidrug-resistant bacteria increases the risk of complications and a fatal outcome after allogeneic hematopoietic cell transplantation. Ann Hematol. 2018;97(3):509–17.CrossRefPubMed
18.
Zurück zum Zitat Tacconelli E, Cataldo MA, Dancer SJ, De Angelis G, Falcone M, Frank U, Kahlmeter G, Pan A, Petrosillo N, Rodríguez-Baño J, Singh N, Venditti M, Yokoe DS, Cookson B. ESCMID guidelines for the management of the infection control measures to reduce transmission of multidrug-resistant Gramnegative bacteria in hospitalized patients. Clin Microbiol Infect. 2014;20(Suppl. 1):1–55.CrossRefPubMed Tacconelli E, Cataldo MA, Dancer SJ, De Angelis G, Falcone M, Frank U, Kahlmeter G, Pan A, Petrosillo N, Rodríguez-Baño J, Singh N, Venditti M, Yokoe DS, Cookson B. ESCMID guidelines for the management of the infection control measures to reduce transmission of multidrug-resistant Gramnegative bacteria in hospitalized patients. Clin Microbiol Infect. 2014;20(Suppl. 1):1–55.CrossRefPubMed
19.
Zurück zum Zitat ECDC. European centre for disease prevention and control. Systematic review of the effectiveness of infection control measures to prevent the transmission of carbapenemase-producing Enterobacteriaceae through cross-border transfer of patients. 2014. ECDC. European centre for disease prevention and control. Systematic review of the effectiveness of infection control measures to prevent the transmission of carbapenemase-producing Enterobacteriaceae through cross-border transfer of patients. 2014.
21.
Zurück zum Zitat Royal College of Physicians of Ireland. Guidelines for the prevention and control of multi-drug resistant organisms (MDRO) excluding mrsa in the healthcare setting. 2012. Royal College of Physicians of Ireland. Guidelines for the prevention and control of multi-drug resistant organisms (MDRO) excluding mrsa in the healthcare setting. 2012.
22.
Zurück zum Zitat Public Health England (2013) Acute trust toolkit for the early detection, management and control of carbapenemase-producing Enterobacteriaceae. Public Health England (2013) Acute trust toolkit for the early detection, management and control of carbapenemase-producing Enterobacteriaceae.
23.
Zurück zum Zitat Health Protection Scotland. Carbapenemase producing Enterobacteriaceae (CPE) prevention and management toolkit for inpatient areas. 2013. Health Protection Scotland. Carbapenemase producing Enterobacteriaceae (CPE) prevention and management toolkit for inpatient areas. 2013.
24.
Zurück zum Zitat Magiorakos AP, Burns K, Rodríguez Baño J, Borg M, Daikos G, Dumpis U, Lucet JC, Moro ML, Tacconelli E, Simonsen GS, Szilágyi E, Voss A, Weber JT. Infection prevention and control measures and tools for the prevention of entry of carbapenem-resistant Enterobacteriaceae into healthcare settings: guidance from the European Centre for Disease Prevention and Control. Antimicrob Resist Infect Control. 2017;6:113.CrossRefPubMedPubMedCentral Magiorakos AP, Burns K, Rodríguez Baño J, Borg M, Daikos G, Dumpis U, Lucet JC, Moro ML, Tacconelli E, Simonsen GS, Szilágyi E, Voss A, Weber JT. Infection prevention and control measures and tools for the prevention of entry of carbapenem-resistant Enterobacteriaceae into healthcare settings: guidance from the European Centre for Disease Prevention and Control. Antimicrob Resist Infect Control. 2017;6:113.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Otter JA, Mutters NT, Tacconelli E, Gikas A, Holmes AH. Controversies in guidelines for the control of multidrug-resistant Gram-negative bacteria in EU countries. Clin Microbiol Infect. 2015;21:1057–66.CrossRefPubMed Otter JA, Mutters NT, Tacconelli E, Gikas A, Holmes AH. Controversies in guidelines for the control of multidrug-resistant Gram-negative bacteria in EU countries. Clin Microbiol Infect. 2015;21:1057–66.CrossRefPubMed
26.
Zurück zum Zitat Tormo N, Albert E, Borrajo E, Bosque M, Camarena JJ, Domínguez V, Fuentes E, Gascón I, Gomila B, Gonzalo N, Jiménez M, Martínez O, Nogueira JM, Orta N, Prat J, Rodríguez JC, Gimeno C, Navarro D. A survey on practices for active surveillance of carriage of multidrug-resistant bacteria in hospitals in the Autonomous Community of Valencia, Spain. Eur J Clin Microbiol Infect Dis. 2018;37(11):2069–74.CrossRefPubMed Tormo N, Albert E, Borrajo E, Bosque M, Camarena JJ, Domínguez V, Fuentes E, Gascón I, Gomila B, Gonzalo N, Jiménez M, Martínez O, Nogueira JM, Orta N, Prat J, Rodríguez JC, Gimeno C, Navarro D. A survey on practices for active surveillance of carriage of multidrug-resistant bacteria in hospitals in the Autonomous Community of Valencia, Spain. Eur J Clin Microbiol Infect Dis. 2018;37(11):2069–74.CrossRefPubMed
27.
Zurück zum Zitat Sánchez Carrillo C, Guerrero Gómez C, Cercenado E, Cantón R. Recogida, transporte y procesamiento general de muestras de laboratorio de Microbiología. Procedimientos de microbiología clínica. Recomendaciones de la Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica (SEIMC). 2003. Sánchez Carrillo C, Guerrero Gómez C, Cercenado E, Cantón R. Recogida, transporte y procesamiento general de muestras de laboratorio de Microbiología. Procedimientos de microbiología clínica. Recomendaciones de la Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica (SEIMC). 2003.
28.
Zurück zum Zitat Torres I, Gimenez E, Pascual T, Bueno F, Huntley D, Martínez M, Navarro D. Short-term incubation of positive blood cultures in brain-heart infusion broth accelerates identification of bacteria by matrix-assisted laser desorption/ionization time-of-flight mass-spectrometry. J Med Microbiol. 2017;66(12):1752–8.CrossRefPubMed Torres I, Gimenez E, Pascual T, Bueno F, Huntley D, Martínez M, Navarro D. Short-term incubation of positive blood cultures in brain-heart infusion broth accelerates identification of bacteria by matrix-assisted laser desorption/ionization time-of-flight mass-spectrometry. J Med Microbiol. 2017;66(12):1752–8.CrossRefPubMed
29.
Zurück zum Zitat Torres I, Palop N, Salvador RB, Gómez JB, Gimeno C, Navarro D. Evaluation of the DNA microarray “AMR Direct Flow Chip Kit” for detection of antimicrobial resistance genes from Gram-positive and Gram-negative bacterial isolated colonies. Enferm Infecc Microbiol Clin. 2019;37(7):454–7.CrossRef Torres I, Palop N, Salvador RB, Gómez JB, Gimeno C, Navarro D. Evaluation of the DNA microarray “AMR Direct Flow Chip Kit” for detection of antimicrobial resistance genes from Gram-positive and Gram-negative bacterial isolated colonies. Enferm Infecc Microbiol Clin. 2019;37(7):454–7.CrossRef
30.
Zurück zum Zitat Magiorakos AP, Srinivasan A, Carey RB, Carmeli Y, Falagas ME, Giske CG, Harbarth S, Hindler JF, Kahlmeter G, Olsson-Liljequist B, Paterson DL, Rice LB, Stelling J, Struelens MJ, Vatopoulos A, Weber JT, Monnet DL. 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(3):268–81. https://doi.org/10.1111/j.1469-0691.2011.03570.x.).CrossRefPubMed Magiorakos AP, Srinivasan A, Carey RB, Carmeli Y, Falagas ME, Giske CG, Harbarth S, Hindler JF, Kahlmeter G, Olsson-Liljequist B, Paterson DL, Rice LB, Stelling J, Struelens MJ, Vatopoulos A, Weber JT, Monnet DL. 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(3):268–81. https://​doi.​org/​10.​1111/​j.​1469-0691.​2011.​03570.​x.​).CrossRefPubMed
31.
Zurück zum Zitat Papadomichelakis E, Kontopidou F, Antoniadou A, Poulakou G, Koratzanis E, Kopterides P, Mavrou I, Armaganidis A, Giamarellou H. Screening for resistant gram-negative microorganisms to guide empiric therapy of subsequent infection. Intensive Care Med. 2008;34(12):2169–75.CrossRefPubMed Papadomichelakis E, Kontopidou F, Antoniadou A, Poulakou G, Koratzanis E, Kopterides P, Mavrou I, Armaganidis A, Giamarellou H. Screening for resistant gram-negative microorganisms to guide empiric therapy of subsequent infection. Intensive Care Med. 2008;34(12):2169–75.CrossRefPubMed
32.
Zurück zum Zitat Ayats J, Corbella X, Ardanuy C, Domínguez MA, Ricart A, Ariza J, Martin R, Liñares J. Epidemiological significance of cutaneous, pharyngeal, and digestive tract colonization by multiresistant Acinetobacter baumannii in ICU patients. J Hosp Infect. 1997;37(4):287–9.CrossRefPubMed Ayats J, Corbella X, Ardanuy C, Domínguez MA, Ricart A, Ariza J, Martin R, Liñares J. Epidemiological significance of cutaneous, pharyngeal, and digestive tract colonization by multiresistant Acinetobacter baumannii in ICU patients. J Hosp Infect. 1997;37(4):287–9.CrossRefPubMed
33.
Zurück zum Zitat Micozzi A, Gentile G, Minotti C, Cartoni C, Capria S, Ballarò D, Santilli S, Pacetti E, Grammatico S, Bucaneve G, Foà R. Carbapenem-resistant Klebsiella pneumoniae in high-risk haematological patients: factors favouring spread, risk factors and outcome of carbapenem-resistant Klebsiella pneumoniae bacteremias. BMC Infect Dis. 2017;17(1):203.CrossRefPubMedPubMedCentral Micozzi A, Gentile G, Minotti C, Cartoni C, Capria S, Ballarò D, Santilli S, Pacetti E, Grammatico S, Bucaneve G, Foà R. Carbapenem-resistant Klebsiella pneumoniae in high-risk haematological patients: factors favouring spread, risk factors and outcome of carbapenem-resistant Klebsiella pneumoniae bacteremias. BMC Infect Dis. 2017;17(1):203.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Trecarichi EM, Pagano L, Candoni A, Pastore D, Cattaneo C, Fanci R, Nosari A, Caira M, Spadea A, Busca A, Vianelli N, Tumbarello M. Current epidemiology and antimicrobial resistance data for bacterial bloodstream infections in patients with hematologic malignancies: an Italian multicentre prospective survey. Clin Microbiol Infect. 2015;21(4):337–43.CrossRefPubMed Trecarichi EM, Pagano L, Candoni A, Pastore D, Cattaneo C, Fanci R, Nosari A, Caira M, Spadea A, Busca A, Vianelli N, Tumbarello M. Current epidemiology and antimicrobial resistance data for bacterial bloodstream infections in patients with hematologic malignancies: an Italian multicentre prospective survey. Clin Microbiol Infect. 2015;21(4):337–43.CrossRefPubMed
35.
Zurück zum Zitat Trecarichi EM, Pagano L, Martino B, Candoni A, Di Blasi R, Nadali G, Fianchi L, Delia M, Sica S, Perriello V, Busca A, Aversa F, Fanci R, Melillo L, Lessi F, Del Principe MI, Cattaneo C, Tumbarello M. Bloodstream infections caused by Klebsiella pneumoniae in onco-hematological patients: clinical impact of carbapenem resistance in a multicentre prospective survey. Am J Hematol. 2016;91(11):1076–81.CrossRefPubMed Trecarichi EM, Pagano L, Martino B, Candoni A, Di Blasi R, Nadali G, Fianchi L, Delia M, Sica S, Perriello V, Busca A, Aversa F, Fanci R, Melillo L, Lessi F, Del Principe MI, Cattaneo C, Tumbarello M. Bloodstream infections caused by Klebsiella pneumoniae in onco-hematological patients: clinical impact of carbapenem resistance in a multicentre prospective survey. Am J Hematol. 2016;91(11):1076–81.CrossRefPubMed
Metadaten
Titel
Multi-body-site colonization screening cultures for predicting multi-drug resistant Gram-negative and Gram-positive bacteremia in hematological patients
verfasst von
Ignacio Torres
Dixie Huntley
Mar Tormo
Marisa Calabuig
Juan Carlos Hernández-Boluda
María José Terol
Carlos Carretero
Paula de Michelena
Ariadna Pérez
José Luis Piñana
Javier Colomina
Carlos Solano
David Navarro
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
BMC Infectious Diseases / Ausgabe 1/2022
Elektronische ISSN: 1471-2334
DOI
https://doi.org/10.1186/s12879-022-07154-3

Weitere Artikel der Ausgabe 1/2022

BMC Infectious Diseases 1/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Perioperative Checkpointhemmer-Therapie verbessert NSCLC-Prognose

28.05.2024 NSCLC Nachrichten

Eine perioperative Therapie mit Nivolumab reduziert das Risiko für Rezidive und Todesfälle bei operablem NSCLC im Vergleich zu einer alleinigen neoadjuvanten Chemotherapie um über 40%. Darauf deuten die Resultate der Phase-3-Studie CheckMate 77T.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Positiver FIT: Die Ursache liegt nicht immer im Dickdarm

27.05.2024 Blut im Stuhl Nachrichten

Immunchemischer Stuhltest positiv, Koloskopie negativ – in solchen Fällen kann die Blutungsquelle auch weiter proximal sitzen. Ein Forschungsteam hat nachgesehen, wie häufig und in welchen Lokalisationen das der Fall ist.

GLP-1-Agonisten können Fortschreiten diabetischer Retinopathie begünstigen

24.05.2024 Diabetische Retinopathie Nachrichten

Möglicherweise hängt es von der Art der Diabetesmedikamente ab, wie hoch das Risiko der Betroffenen ist, dass sich sehkraftgefährdende Komplikationen verschlimmern.

Update Innere Medizin

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