Next Article in Journal
Current Updates on the Role of Microbiome in Endometriosis: A Narrative Review
Previous Article in Journal
Rumen and Fecal Microbiota Characteristics of Qinchuan Cattle with Divergent Residual Feed Intake
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Molecular and Clinical Data of Antimicrobial Resistance in Microorganisms Producing Bacteremia in a Multicentric Cohort of Patients with Cancer in a Latin American Country

by
Sergio Andrés Cruz-Vargas
1,
Laura García-Muñoz
1,
Sonia Isabel Cuervo-Maldonado
1,2,3,
Carlos Arturo Álvarez-Moreno
1,4,
Carlos Humberto Saavedra-Trujillo
1,
José Camilo Álvarez-Rodríguez
2,3,5,
Angélica Arango-Gutiérrez
2,
Julio César Gómez-Rincón
2,
Katherine García-Guzman
2,
Aura Lucía Leal
6,
Javier Garzón-Herazo
7,
Samuel Martínez-Vernaza
7,8,
Fredy Orlando Guevara
9,
Leydy Paola Jiménez-Cetina
10,
Liliana Marcela Mora
10,
Sandra Yamile Saavedra
6 and
Jorge Alberto Cortés
1,11,*
1
Department of Internal Medicine, Universidad Nacional de Colombia, Sede Bogotá, Bogotá 111321, Colombia
2
Infectious Diseases Group, Instituto Nacional de Cancerología-ESE, Bogotá 111511, Colombia
3
Research Group in Cancer Infectious Diseases and Hematological Alterations (GREICAH), Bogotá 111321, Colombia
4
Clínica Universitaria Colombia, Bogota 111321, Colombia
5
Hospital Universitario Clínica San Rafael, Bogotá 110111, Colombia
6
Department of Microbiology, Universidad Nacional de Colombia, Bogotá 111321, Colombia
7
Infectious Diseases Unit, Hospital Universitario San Ignacio, Bogotá 110231, Colombia
8
Research Group in Infectious Diseases, Hospital Universitario San Ignacio, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
9
Infectious Diseases Department, Clínica Reina Sofía, Bogotá 110121, Colombia
10
Microbiology Laboratory, Instituto Nacional de Cancerología-ESE, Bogotá 111511, Colombia
11
Diseases Unit, Hospital Universitario Nacional, Bogotá 111321, Colombia
*
Author to whom correspondence should be addressed.
Microorganisms 2023, 11(2), 359; https://doi.org/10.3390/microorganisms11020359
Submission received: 15 December 2022 / Revised: 17 January 2023 / Accepted: 20 January 2023 / Published: 31 January 2023
(This article belongs to the Section Medical Microbiology)

Abstract

:
Patients with cancer have a higher risk of severe bacterial infections. This study aims to determine the frequency, susceptibility profiles, and resistance genes of bacterial species involved in bacteremia, as well as risk factors associated with mortality in cancer patients in Colombia. In this prospective multicenter cohort study of adult patients with cancer and bacteremia, susceptibility testing was performed and selected resistance genes were identified. A multivariate regression analysis was carried out for the identification of risk factors for mortality. In 195 patients, 206 microorganisms were isolated. Gram-negative bacteria were more frequently found, in 142 cases (68.9%): 67 Escherichia coli (32.5%), 36 Klebsiella pneumoniae (17.4%), and 21 Pseudomonas aeruginosa (10.1%), and 18 other Gram-negative isolates (8.7%). Staphylococcus aureus represented 12.4% (n = 25). Among the isolates, resistance to at least one antibiotic was identified in 63% of them. Genes coding for extended-spectrum beta-lactamases and carbapenemases, blaCTX-M and blaKPC, respectively, were commonly found. Mortality rate was 25.6% and it was lower in those with adequate empirical antibiotic treatment (22.0% vs. 45.2%, OR: 0.26, 95% CI: 0.1–0.63, in the multivariate model). In Colombia, in patients with cancer and bacteremia, bacteria have a high resistance profile to beta-lactams, with a high incidence of extended-spectrum beta-lactamases and carbapenemases. Adequate empirical treatment diminishes mortality, and empirical selection of treatment in this environment of high resistance is of key importance.

1. Introduction

The complications associated with cancer are multiple. Among them, bacterial infections stand out, which vary according to the type of neoplasia and the treatment used. One in four patients with post-chemotherapy severe febrile neutropenia develops bacteremia [1], with a negative effect on initiating a specific cancer treatment, prolonged hospital stays, morbidity, and mortality, particularly when inappropriate treatment is used [2]. Cancer patients represent a particularly complex group due to multiple risk factors for developing infectious complications. These include immunosuppression due to the underlying disease and its treatment [3], exposure to broad-spectrum antimicrobials, with the consequent selection of multidrug-resistant microorganisms, and morbidity and mortality due to septic shock [4].
Antimicrobial resistance is a public health problem. According to the WHO, it is one of the crucial issues of the twenty-first century, with a significant socioeconomic impact worldwide [5]. This problem is especially important among Enterobacterales, such as Escherichia coli, Klebsiella spp., Enterobacter spp., but also among isolates of Pseudomonas aeruginosa, in which the resistance to third-generation cephalosporins and carbapenems is specially problematic [6]. González et al. conducted a systematic review in 2014, which demonstrated an increase in antimicrobial resistance between 2006 and 2011 in Enterobacteriaceae to third-generation cephalosporin in third-level hospitals in Colombia [7]. The expected effect of the resistance is a lower probability of an appropriate empirical therapy, which affects the morbidity and mortality of these immunocompromised patients [2].
Currently, there is no information about the susceptibility profile and molecular mechanisms of resistance in Latin America, among cancer patients with bacterial infection. The aim of this work is to identify the resistance profiles and describe genes related to resistant microorganisms in cancer patients who present an episode of bacteremia in hospitals in Colombia.

2. Materials and Methods

2.1. Setting

Patients from six reference hospitals in the city of Bogotá, Colombia, were included: Instituto Nacional de Cancerología (INC), Clínica Universitaria Colombia (CUC), Hospital Universitario Nacional de Colombia (HUNC), Clínica Reina Sofía (CRS), Hospital Universitario Clínica San Rafael (HUSR), and Hospital Universitario San Ignacio (HUSI), during the period between 1 July 2018, and 31 July 2020. All these hospitals have oncology or hemato-oncology care units; one of them is public (INC) and all serve patients belonging to both public and private health care providers. They serve Colombian patients from Bogotá or other cities by referral.

2.2. Study Design

A prospective, multicenter, cohort study in adult patients was designed with those diagnosed with cancer of any tumor location and stage who developed a bacteremia confirmed by the laboratory of the participating institutions.
The inclusion criteria considered adult individuals of any race, with a confirmed cancer diagnosis, with or without treatment (chemotherapy, radiotherapy, surgical management). For microorganisms that are part of the microbiota of the skin, bacteremia was considered when there were at least two positive blood cultures for the same germ; similarly, at least one blood culture was considered for other Gram-positive microorganisms or Gram-negative ones. Patients with incomplete medical history information were excluded from the study.

2.3. Study Size and Sample

Sampling was non-probabilistic, sequential, and based on compliance with inclusion and exclusion criteria. The sample size was calculated considering an expected prevalence of 30% for Staphylococcus aureus (most frequent individual microorganism expected) and 60 to 70% Gram-negatives (as a group) in bacteremia among cancer patients, with a precision of 10%, a power of 90%, and an alpha error of 0.05 (two-tailed). A sample of 200 patients was estimated.

2.4. Definitions

Bacteremia is the infection of the bloodstream caused by bacteria. A systemic inflammatory response is understood as the presence of two or more of the following criteria: temperature greater than 38 °C or less than 36 °C; heart rate greater than 90 beats per minute; respiratory rate greater than 20 breaths per minute; or leukocyte count greater than 12,000 cells/mm3, less than 4000 cells/mm3, or more than 10% of bandemia [8]. Sepsis implies the presence of systemic inflammatory response, given that all patients had an identified infectious focus. Febrile neutropenia is an absolute count below 500 cells per milliliter or 1000 cells expected to fall below 500 cells per milliliter within 48 h. Additionally, it includes the presence of fever, defined as two consecutive temperature measurements of 38 °C for 2 h or an oral temperature greater than 38.3 °C [9]. Lymphopenia consists of a lymphocyte count of fewer than 1500 cells/mm3 and is severe with a count of less than 500 cells/mm3 [10].
Natural resistance profile is defined as the resistance pattern of each bacterial family, species, or group isolated in the antibiogram [11]. Acquired resistance profile, evidenced through the antibiogram, is characterized by not being typical of the bacterial family, species, or group; it is variable and obtained by a strain of a particular species [11]. Adequate antimicrobial treatment is antibiotic management with activity against the bacteria isolated in the blood culture, applied at the onset of symptoms or signs of infection, and maintained during the following 72 h. It is considered effective in vitro against the isolated microorganisms [12].

2.5. Microbiological Procedures

Isolates of the bacteria identified in the blood cultures of cancer patients who met the inclusion criteria and not the exclusion criteria were collected. Initial identification at the genus and species level and measurement of sensitivity to antimicrobials of the isolates were carried out in the clinical laboratory of the selected hospitals of Bogotá, with the automated systems of each institution: VITEK (INC, HUSI, CUC, CRS, Biomerieux, France) and Phoenix 100 (HUNC, HUSR, BD, USA), according to the manufacturers’ instructions. No molecular methods were used for identification. Antibiotics tested by every hospital were chosen by each center. Commonly tested antibiotics include ampicillin, cephalosporins, carbapenems, aminoglycosides (amikacin and gentamicin), ciprofloxacin, and trimethoprim/sulfametoxazole. Colistin was tested against P. aeruginosa isolates. Multidrug resistance (MDR) was defined as the presence of resistance to 3 or more antibiotic classes.
Two infectious disease specialists (JAC and SCM) reviewed, individually, the antibiograms and gave their conclusions according to the resistance phenotypic profiles of the microorganism evaluated. A third infectologist (JSB) evaluated the discordances that arose, and the final phenotypic profile was defined by agreement.
The isolates that showed some type of resistance as described in the study protocol were transferred in Cary–Blair medium to the Microbiology Laboratory of the Universidad Nacional de Colombia to be stored for subsequent molecular testing. Before the molecular study, the cryopreservation of strains was verified. The DNA of the isolates was obtained using cell lysis from fresh bacterial colonies of culture in non-selective agar. The sample was diluted at 1:10, taking 50 μL of the supernatant and placing it in a tube for DNA dilution (450 μL of sterile Type I water). This DNA sample was stored in a cold room when processed within three days; otherwise, in a −20 °C refrigerator until processing (the samples at −20 °C were thawed gradually by placing them in a cold room). Detection of resistance determinants was performed by conventional PCR. In Gram-negative bacilli, the presence of beta-lactamase-encoding genes was evaluated using specific primers for blaSHV, blaTEM, blaCTX-M [11], blaKPC, blaGES, blaVIM [12], blaNDM [13], blaIMP [14], and blaOXA-48 [15]. In Staphylococcus spp., resistance to methicillin was confirmed by detecting the mecA gene [16], and in Enterococcus spp., for vancomycin resistance, the presence of the vanA and vanB genes was evaluated [17]. DNA was added to tubes, vortexed, and 5 μL of each sample was placed in a thermocycler. A 1.2% agarose gel was prepared, and, finally, the electrophoresis was carried out.

2.6. Ethical Considerations

The study was conducted in accordance with good clinical practice (GCP) standards. The level of ethical risk at which the study is classified according to Article 11 of Resolution No. 008430 of 1993 issued by the Ministry of Health of Colombia is “Research with minimum risk”. To guarantee confidentiality, anonymous data were used. The protocol was submitted for review and approved by the Ethics Committee of each of the participating institutions.

2.7. Statistical Analysis

Data were entered into the REDCAP platform, and their statistical analysis was carried out using the R program (version 4.0.6). Demographic characteristics were recorded in relative and absolute frequencies, and measures of central tendency were calculated for continuous variables as mean (standard deviation) and median (interquartile range) according to their distribution. The exploratory analysis compared the proportions of qualitative variables using the X2 test, and a Yates correction was applied when there were fewer than 100 observations. The multivariate analysis used a logistic regression method to predict mortality, considering clinical variables and inappropriate antimicrobial therapy as predictors. Results with p < 0.05 were considered significant.

3. Results

3.1. Characteristics of the Population

In the 195 patients included, 206 microorganisms were identified: 120 (61.5%) from INC, 25 (12.8%) from CUC, 19 (9.7%) from HUSI, 16 (8.2%) from HUSR, 9 (4.6%) from HUNC, and 6 (3.1%) from CRS. There were 109 women (55.8%), and the average age was 52 years (SD 17.2 years). The majority (104, 53.3%) had hematological cancer. Of the total number of patients, 83 (42.5%) had active oncological pathology, 43 (22%) were in relapse, 41 (21%) had de novo cancer, 15 (7.7%) were in remission, and 13 (6.6%) were in palliative care for their pathology. As for cancer treatment, in the last month, 105 (53.8%) had received chemotherapy, 23 (11.8%) radiation therapy, and 49 (25.1%) some surgery. Only six (5.7%) of the patients with hematological cancer had a hematopoietic progenitor transplantation. Among the patients who received chemotherapy, 24 (22.8%) had an implantable catheter. Of the total patients included, 52 (26.6%) had had a previous infection, and 70 (35.8%) had received antibiotic treatment during the previous month. The most frequently used ones were non-carbapenem beta-lactams with 43 episodes (21.9%), followed by carbapenems with 18 episodes (9.2%), and vancomycin with 14 episodes (7.1%). At the time of presenting bacteremia, 131 (67.1%) were in hospital, 56 (28.7%) in the emergency room, and 8 (4.1%) in the intensive care unit; more than a half of them had a medical device (Table 1).
Severe lymphopenia was also identified in 111 patients (56.9%), hyperbilirubinemia in 43 (33.3%, tested in 129), and acidemia in 7 (8.8%, tested in 80), with significant metabolic acidosis/acidemia in 40% of these. Hypoxemia was observed in 37 cases (48.6%, measured in 76) and hyperlactatemia in 31 patients (37.3%, measured in 83), and acute kidney injury was found in 19 (10.4%).

3.2. Microbiological Results

In the 195 patients included, 206 microorganisms were identified; bacteremia was polymicrobial in 11 patients (5.6%). A total of 142 Gram-negative (68.9%) and 64 Gram-positive (31.1%) isolates were found (Table 2). The principal Gram-negative germ isolated was Escherichia coli, followed by Klebsiella pneumoniae and Pseudomonas aeruginosa. Among the Gram-positive bacteria, the main isolates correspond to coagulase-negative Staphylococci, followed by S. aureus, Enterococcus species, Streptococcus species, Bacillus, and Lysinibacillus. Most isolates, 130 (63.1%), had an acquired resistance pattern. In 192 patients (98.4%), initial antibiotic treatment was prescribed, which was adequate in 155 cases (79.5%).
Among Gram-negative bacteria, the phenotype observed in the resistance antibiograms of third- and fourth-generation cephalosporins was found in E. coli in 13 cases (19.7%), in 3 in K. pneumoniae (8.3%), and in 2 cases in P. aeruginosa (9.5%). No resistance to carbapenems was observed in E. coli, but it was found in 12 isolates of K. pneumoniae (34.2%) and 4 of P. aeruginosa (19%) (Table 3). Regarding the resistance of Gram-negative bacteria, in general, resistance to third- and fourth-generation cephalosporins was evidenced in 29 cases (22.4%) and to carbapenems in 15 (10.9%). MDR isolates were seen in E. coli (12, 17.4%), K. pneumoniae (16, 44.4%), P. aeruginosa (7, 33.3%), Serratia marcescens (1, 100%), and S. maltophilia (1, 100%). Among Gram-positives, MDR isolates were seen in S. aureus (7, 28%, all methicillin-resistant S. aureus isolates were classified as MDR), CNS (14, 53.8%), and E. faecalis (1, 25%)
Genes associated with resistance were found in 33 bacterial isolates studied: the principal gene was blaCTX-M in 14 cases, followed by blaTEM in 11, and blaSHV in 4 isolates of E. coli or K. pneumoniae. In two isolates, all three genes were found simultaneously. Among the carbapenem-resistant bacteria, the most common gene was blaKPC in 13 of 15 isolates (87%), followed by blaVIM in 2 (13%). No other causative genes, such as blaGES, blaOXA-48, blaNDM, or blaIMP (Table 4), were found; it is important to highlight that the analysis of one of the isolates of P. aeruginosa was not possible due to limited sample availability.
Among the Gram-positive microorganisms, six (24%) isolates of S. aureus resistant to oxacillin were observed, and none were resistant to vancomycin. In E. faecalis, no resistance to ampicillin or vancomycin was found. In E. faecium, four (100%) ampicillin-resistant and two (50%) vancomycin-resistant isolates were found. Of the isolates of S. aureus resistant to oxacillin, 83% had mecA. Among the vancomycin-resistant isolates of E. faecium, the vanA gene caused 100%.
A history of antibiotic use during the month before bacteremia was observed in 52 patients (40.9%) with microorganisms with an acquired profile (n = 127), and in 18 (26.4%) with microorganisms with a natural susceptibility profile (n = 68; OR: 1.91, 95% CI: 0.97–3.9). Among patients with Gram-negative bacilli isolates (n = 142), 22 (59.5%) had received a previous antibiotic from those identified as having a third- or fourth-generation cephalosporin-resistant or carbapenem-resistant microorganism (n = 37). This exposure occurred in 26 (24.8%) patients without this profile (n = 105; OR: 4.5, 95% CI: 2.0–9.8).

3.3. Mortality and Risk Factors

In the group of patients with resistant Gram-negative infections, four (22% of 18) with bacteria resistant to third- or fourth-generation cephalosporins and three (18.7% of 16) with bacteria resistant to carbapenems died at 30 days (p = 1.0). Two patients (33.3% of 6) in the group with MRSA bacteremia and two (100%) patients with vancomycin-resistant E. faecium died at 30 days (p = 0.42).
At day 30, from the time of identifying bacteremia, 134 (68.7%) patients had been discharged, 50 (25.6%) had died, and 11 (5.6%) were still hospitalized. Among the survivors, 56 (38.6%) patients had a natural profile, while 12 (24%) of those who died had this profile (OR: 0.50, 95% CI: 0.22–1.09, p = 0.09). Differences in mortality were observed according to the clinical stage of the patient: 0% mortality (0 of 15 patients) in patients in complete remission; 19.2% (16 of 83 patients) in those with active disease (p = 0.12); 23.8% (10 of 42) in those with de novo disease; 39.7% (17 of 43) in patients in relapse (p = 0.21); and 53.8% (7 of 13) for patients in palliative care (p < 0.01, compared to those in remission).
In the multivariate analysis, an inverse relationship was found between adequate treatment and mortality (OR: 0.26, 95% CI: 0.1–0.63), in addition to a relationship between mortality and palliative care (OR: 3.51, 95% CI: 1.05–12.04). Finally, a relationship was observed between survival and the number of platelets (per 10,000 additional platelets) (OR: 0.97, 95% CI: 0.94–0.99).

4. Discussion

Our study shows high antibiotic resistance among bacteremia-causing microorganisms in cancer patients in a developing country. Resistance rates were higher among Gram-negative bacteria and especially among K. pneumoniae isolates. In addition, it presents the characterization of the principal genes responsible for resistant bacteria, among which blaCTX-M and blaKPC stand out in the group of extended-spectrum beta-lactamases and carbapenems, respectively.
The microbiological findings showed a predominance of Gram-negative microorganisms in 68.6% of the isolates. These findings are in concordance with recent studies that evidence a predominance of this type of bacteria [18,19,20] and contrast with research carried out in the 2000s where the main bacteria causing infection in the bloodstream were Gram-positive bacteria [21]. It has been observed that the frequency of Gram-negative and Gram-positive microorganisms changes according to the geographical area [22,23], hence the importance of having regional information. Changes in epidemiology may be related to multiple causes, such as the utilization of antibiotic prophylaxis, campaigns to promote the reduction of device-associated bacteremia, the types of chemotherapy applied that have been more intensive during the last decade, or the lower use of chemotherapy catheters, among others [18,24].
Among the microorganisms identified, an acquired resistance profile was found in 63% of the isolates, in agreement with recent studies that have shown a gradual increase in antimicrobial resistance; among the proposed mechanisms, selective pressure due to inadequate use of antibiotics has been considered [22,25]. In South Korea, Nham et al. found a relationship between the development of BLEE-producing K. pneumoniae and the previous use of antimicrobials [20], without ruling out cross-bacterial transmission in different institutions, especially in those with low antimicrobial use, and the introduction of resistant bacteria from the community [26].
The most frequent resistance profiles varied according to the species identified. In E. coli, the most phenotypically and genotypically recognized resistance was due to BLEE; resistance to carbapenems was observed in K. pneumoniae, and multiresistance in P. aeruginosa. Upon genetic evaluation, 29 genes associated with bacteria resistant to third- and fourth-generation cephalosporins were found. The main gene involved was blaCTX-M, followed by blaTEM and finally blaSHV. In a group of neutropenic patients with hematological cancer and colonized by BLEE-producing E. coli¸ Arnan et al. (Spain) found blaCTX-M as the main gene associated with 81% of the isolates, followed by blaSHV (15%) and blaTEM (3.3%) [27]. In a group of patients with bacteremia and hematological cancer with BLEE-producing E. coli infection, Trecarichi et al. (Italy) described blaTEM as the main gene related to 36.1%, followed by blaCTX-M (33.3%) and blaSHV (30.6%) [28], which indicates that resistance patterns may be due to the geographical distribution of resistance genes. On the other hand, resistance to carbapenems was found in 34.2% and 19% of the K. pneumoniae and P. aeruginosa isolates, respectively. The main gene related to this resistance was blaKPC, followed by the blaVIM gene. Satlin et al. performed the characterization of carbapenem-resistant Enterobacteriaceae in patients with hematological cancer, neutropenia, and bacteremia in New York [29]. In this study, the main gene related to this resistance was blaKPC in 91% of the isolates. The high mortality rate of 53% at 30 days in the study by Satlin et al., compared to 18.7% in our study, associated with carbapenem-resistant bacteria, is noteworthy. Our data are consistent with what was observed in general among carbapenem-resistant isolates in Colombia, where the most frequently identified gene is blaKPC, regardless of the species [30]. We found a genetic variation according to the geographical area studied, which may be relevant when developing molecular diagnostic technology [31].
In patients with S. aureus, resistance to oxacillin was found in 24%; this is similar to what has been found in the international literature, which varies from 18–44% [32,33]. Regarding the result of 83% with evidence of the mecA gene as a mechanism of resistance, this is striking given it is the most commonly found gene, but other mechanisms not described to date in Colombia, such as mecC, can have the same resistance profile [34]. As for E. faecium, 50% resistance to vancomycin was found, 100% related to the vanA gene, with a mortality of 100% at 30 days. Observed mortality among patients with cancer infected with vancomycin-resistant E. faecium has been very high [35]. Vancomycin, piperacillin/tazobactam, meropenem, and metronidazole have been implicated in its appearance. It may cause bacteremia, but also complicated abdominal surgery, and skin-related infection. Long-term mortality has also been observed among patients with urological or colorectal malignancies [36]. Additionally, there is a considerable risk of outbreaks of these MDR microorganisms that might put a large number of patients in danger [37].
Regarding the outcomes, we did not find significant differences in relation to survival in patients with an acquired resistance profile compared to natural susceptibility. The information regarding this finding is contradictory. Tumbarello et al. in 2009 described a relationship between mortality and resistant bacteria [38]; among other factors related to mortality, age, comorbidities, and inappropriate use of antimicrobials have also been suggested [39].
A relationship was found between adequate initial antimicrobial treatment and survival, which is consistent with what has been described in multiple international studies that demonstrated a clear association between the use of inadequate antibiotic treatment and increased mortality [29,40,41,42]. Similarly, the group of patients in palliative care had a higher mortality rate, similar to what was previously described in the literature [18]. This finding, in relation to the prudent use of antimicrobials at the end of life, raises the need for interdisciplinary interventions to define the duration of antibiotics in this group of patients to reduce their impact on the microbiota [43]. Finally, thrombocytopenia was also associated with mortality in this group of patients. In 2012, Horino et al. described thrombocytopenia as a factor of poor prognosis in patients with P. aeruginosa bacteremia of which about 60% were cancer patients [44], which may represent poor control of the oncologic disease or an effect of septic shock.
Our study has some limitations, including the fact that despite being multicenter, most patients came from the INC because it is the largest public hospital in Colombia that treats cancer patients; the other participating institutions have oncology units in addition to treating other types of pathologies. When studying cancer patients, we might be analyzing a heterogeneous group due to the epidemiological and clinical differences of different types of oncologic pathologies.
On the other hand, this is the first Colombian study that reports phenotypic and molecular resistance patterns in patients with cancer and bacteremia. Considering that bacterial resistance is different in each geographic area, the results presented here are of great interest, given that they contribute to recommendations to strengthen programs aimed at the prudent use of antimicrobials and to encourage the creation or improved functioning of infection committees in institutions that treat cancer patients [45]. This information might also be helpful for the development and implementation of molecular diagnostic tests in the country to identify bacterial resistance genes as a useful tool in decision-making for sensible antimicrobial use [31].

5. Conclusions

In this cohort study in hospitals in a developing country, most of the bacteria identified were Gram-negative, with elevated resistance to beta-lactams with the production of ESBL-type beta-lactamases and carbapenemases, mediated by specific genes. The observed resistance profile is high and had an effect on the probability of administering an appropriate antibiotic therapy. Patients with bacteremia and palliative care had also a higher risk of dying by day 30.

Author Contributions

Conceptualization, L.G.-M., S.I.C.-M. and J.A.C.; methodology, A.L.L. and J.A.C.; formal analysis, S.A.C.-V., S.I.C.-M. and J.A.C.; investigation, S.A.C.-V., L.G.-M., S.I.C.-M., C.A.Á.-M., C.H.S.-T., J.C.Á.-R., A.A.-G., J.C.G.-R., K.G.-G., A.L.L., J.G.-H., S.M.-V., F.O.G., L.P.J.-C., L.M.M., S.Y.S. and J.A.C.; resources, S.I.C.-M.; data curation, S.A.C.-V., L.G.-M., S.I.C.-M., A.A.-G., K.G.-G., S.M.-V., L.P.J.-C., L.M.M. and J.A.C.; writing—original draft preparation, S.A.C.-V., L.G.-M., S.I.C.-M. and J.A.C.; writing—review and editing, S.I.C.-M., C.A.Á.-M., C.H.S.-T., J.C.Á.-R., A.A.-G., J.C.G.-R., K.G.-G., A.L.L., J.G.-H., S.M.-V., F.O.G., L.P.J.-C., L.M.M., S.Y.S. and J.A.C.; project administration, K.G.-G.; funding acquisition, S.I.C.-M. All authors have read and agreed to the published version of the manuscript.

Funding

This project was funded by Instituto Nacional de Cancerologia, ESE, nation investment resources (code C19010300-416, 2018-2021).

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Institutional Review Board of the following institutions: Instituto Nacional de Cancerologia (C19010300-416, reference INT-OFI-06665-2017, 5 July 2017); Hospital Universitario de Colombia (CEI-2019-03-01, 18 March 2019); Hospital Universitario San Ignacio (FM-CIE-0579-19, 12 September 2019); Hospital Universitario Clínica San Rafael (CEI-009-2019, 15 February 2019); Clínica Reina Sofía and Clínica Universitaria Colombia (Fundacion Universitaria Sanitas, CEIFUS 254-19. 9 April 2019).

Data Availability Statement

Not applicable.

Acknowledgments

Juan Sebastián Bravo (JSB) provided input for the phenotypic interpretation of the isolates.

Conflicts of Interest

J.A.C. and C.A.Á.-M. have a research grant sponsored by Pfizer and the International Society of Infectious Diseases. C.H.S.-T. has research grants sponsored by Merck and Pfizer. A.L.L. has research grants sponsored by Merck Sharp and Dohme and Pfizer. The other authors have no conflicts of interest to declare.

References

  1. Freifeld, A.G.; Bow, E.J.; Sepkowitz, K.A.; Boeckh, M.J.; Ito, J.I.; Mullen, C.A.; Raad, I.I.; Rolston, K.V.; Young, J.A.; Wingard, J.R.; et al. Clinical practice guideline for the use of antimicrobial agents in neutropenic patients with cancer: 2010 Update by the Infectious Diseases Society of America. Clin. Infect. Dis. 2011, 52, 427–431. [Google Scholar] [CrossRef] [Green Version]
  2. Cortes, J.A.; Cuervo, S.; Gomez, C.A.; Bermudez, D.; Martinez, T.; Arroyo, P. Febrile neutropenia in the tropics: A description of clinical and microbiological findings and their impact on inappropriate therapy currently used at an oncological reference center in Colombia. Biomedica 2013, 33, 70–77. [Google Scholar] [CrossRef] [PubMed]
  3. Viscoli, C.; Castagnola, E. Factors predisposing cancer patients to infection. Cancer Treat Res. 1995, 79, 1–30. [Google Scholar] [CrossRef] [PubMed]
  4. Gonzalez-Barca, E.; Fernandez-Sevilla, A.; Carratala, J.; Salar, A.; Peris, J.; Granena, A.; Gudiol, F. Prognostic factors influencing mortality in cancer patients with neutropenia and bacteremia. Eur. J. Clin. Microbiol. Infect. Dis. 1999, 18, 539–544. [Google Scholar] [CrossRef] [PubMed]
  5. World Health Organization. Global Action Plan on Antimicrobial Resistance; WHO: Geneva, Switzerland, 2015; pp. 1–28. [Google Scholar]
  6. Nouer, S.A.; Nucci, M.; Anaissie, E. Tackling antibiotic resistance in febrile neutropenia: Current challenges with and recommendations for managing infections with resistant Gram-negative organisms. Expert Rev. Hematol. 2015, 8, 647–658. [Google Scholar] [CrossRef] [PubMed]
  7. Gonzalez, L.; Cortes, J.A. Systematic review of antimicrobial resistance in Enterobacteriaceae isolates from Colombian hospitals. Biomedica 2014, 34, 180–197. [Google Scholar] [CrossRef]
  8. Huerta, L.E.; Rice, T.W. Pathologic Difference between Sepsis and Bloodstream Infections. J. Appl. Lab. Med. 2019, 3, 654–663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Klastersky, J.; de Naurois, J.; Rolston, K.; Rapoport, B.; Maschmeyer, G.; Aapro, M.; Herrstedt, J.; Committee, E.G. Management of febrile neutropaenia: ESMO Clinical Practice Guidelines. Ann. Oncol. 2016, 27, v111–v118. [Google Scholar] [CrossRef]
  10. Berezne, A.; Bono, W.; Guillevin, L.; Mouthon, L. Diagnosis of lymphocytopenia. Presse Med. 2006, 35, 895–902. [Google Scholar] [CrossRef] [PubMed]
  11. Monstein, H.J.; Ostholm-Balkhed, A.; Nilsson, M.V.; Nilsson, M.; Dornbusch, K.; Nilsson, L.E. Multiplex PCR amplification assay for the detection of blaSHV, blaTEM and blaCTX-M genes in Enterobacteriaceae. APMIS 2007, 115, 1400–1408. [Google Scholar] [CrossRef]
  12. Monteiro, J.; Widen, R.H.; Pignatari, A.C.; Kubasek, C.; Silbert, S. Rapid detection of carbapenemase genes by multiplex real-time PCR. J. Antimicrob. Chemother. 2012, 67, 906–909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Bonnin, R.A.; Naas, T.; Poirel, L.; Nordmann, P. Phenotypic, biochemical, and molecular techniques for detection of metallo-beta-lactamase NDM in Acinetobacter baumannii. J. Clin. Microbiol. 2012, 50, 1419–1421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Garza-Ramos, U.; Morfin-Otero, R.; Sader, H.S.; Jones, R.N.; Hernandez, E.; Rodriguez-Noriega, E.; Sanchez, A.; Carrillo, B.; Esparza-Ahumada, S.; Silva-Sanchez, J. Metallo-beta-lactamase gene bla(IMP-15) in a class 1 integron, In95, from Pseudomonas aeruginosa clinical isolates from a hospital in Mexico. Antimicrob. Agents Chemother. 2008, 52, 2943–2946. [Google Scholar] [CrossRef] [Green Version]
  15. Poirel, L.; Walsh, T.R.; Cuvillier, V.; Nordmann, P. Multiplex PCR for detection of acquired carbapenemase genes. Diagn. Microbiol. Infect. Dis. 2011, 70, 119–123. [Google Scholar] [CrossRef]
  16. Montazeri, E.A.; Khosravi, A.D.; Jolodar, A.; Ghaderpanah, M.; Azarpira, S. Identification of methicillin-resistant Staphylococcus aureus (MRSA) strains isolated from burn patients by multiplex PCR. Burns 2015, 41, 590–594. [Google Scholar] [CrossRef]
  17. Clark, N.C.; Cooksey, R.C.; Hill, B.C.; Swenson, J.M.; Tenover, F.C. Characterization of glycopeptide-resistant enterococci from U.S. hospitals. Antimicrob. Agents Chemother. 1993, 37, 2311–2317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Velasco, E.; Byington, R.; Martins, C.A.; Schirmer, M.; Dias, L.M.; Goncalves, V.M. Comparative study of clinical characteristics of neutropenic and non-neutropenic adult cancer patients with bloodstream infections. Eur. J. Clin. Microbiol. Infect. Dis. 2006, 25, 1–7. [Google Scholar] [CrossRef]
  19. Marin, M.; Gudiol, C.; Ardanuy, C.; Garcia-Vidal, C.; Calvo, M.; Arnan, M.; Carratala, J. Bloodstream infections in neutropenic patients with cancer: Differences between patients with haematological malignancies and solid tumours. J. Infect. 2014, 69, 417–423. [Google Scholar] [CrossRef]
  20. Trecarichi, E.M.; Pagano, L.; Candoni, A.; Pastore, D.; Cattaneo, C.; Fanci, R.; Nosari, A.; Caira, M.; Spadea, A.; Busca, A.; et al. 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, 337–343. [Google Scholar] [CrossRef] [Green Version]
  21. Wisplinghoff, H.; Seifert, H.; Wenzel, R.P.; Edmond, M.B. Current trends in the epidemiology of nosocomial bloodstream infections in patients with hematological malignancies and solid neoplasms in hospitals in the United States. Clin. Infect. Dis. 2003, 36, 1103–1110. [Google Scholar] [CrossRef]
  22. Montassier, E.; Batard, E.; Gastinne, T.; Potel, G.; de La Cochetiere, M.F. Recent changes in bacteremia in patients with cancer: A systematic review of epidemiology and antibiotic resistance. Eur. J. Clin. Microbiol. Infect. Dis. 2013, 32, 841–850. [Google Scholar] [CrossRef] [PubMed]
  23. Al Battashi, A.; Al Harrassi, B.; Al Maskari, N.; Al Hashami, H.; Al Awaidy, S. Alarming Antibiotic Resistance in Pediatric Oncology Patients: A Three-Year Prospective Cohort Study from Oman. Infect. Drug. Resist. 2022, 15, 3939–3947. [Google Scholar] [CrossRef] [PubMed]
  24. Belloni, S.; Caruso, R.; Cattani, D.; Mandelli, G.; Donizetti, D.; Mazzoleni, B.; Tedeschi, M. Occurrence rate and risk factors for long-term central line-associated bloodstream infections in patients with cancer: A systematic review. Worldviews Evid. Based Nurs. 2022, 19, 100–111. [Google Scholar] [CrossRef] [PubMed]
  25. De la Court, J.R.; Woudt, S.H.S.; Schoffelen, A.F.; Heijmans, J.; de Jonge, N.A.; van der Bruggen, T.; Bomers, M.K.; Lambregts, M.M.C.; Schade, R.P.; Sigaloff, K.C.E.; et al. Third-generation cephalosporin resistant gram-negative bacteraemia in patients with haematological malignancy; an 11-year multi-centre retrospective study. Ann. Clin. Microbiol. Antimicrob. 2022, 21, 54. [Google Scholar] [CrossRef] [PubMed]
  26. Montoya-Urrego, D.; Tellez-Carrasquilla, S.; Vanegas, J.M.; Quiceno, J.N.J. High frequency of colonization by extended-spectrum beta-lactamase-producing Gram-negative bacilli in hemodialysis patients and their household contacts in Colombia: Dissemination between the community and the hospital. Epidemiol. Health 2022, 44, e2022069. [Google Scholar] [CrossRef]
  27. Arnan, M.; Gudiol, C.; Calatayud, L.; Linares, J.; Dominguez, M.A.; Batlle, M.; Ribera, J.M.; Carratala, J.; Gudiol, F. Risk factors for, and clinical relevance of, faecal extended-spectrum beta-lactamase producing Escherichia coli (ESBL-EC) carriage in neutropenic patients with haematological malignancies. Eur. J. Clin. Microbiol. Infect. Dis. 2011, 30, 355–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Trecarichi, E.M.; Tumbarello, M.; Spanu, T.; Caira, M.; Fianchi, L.; Chiusolo, P.; Fadda, G.; Leone, G.; Cauda, R.; Pagano, L. Incidence and clinical impact of extended-spectrum-beta-lactamase (ESBL) production and fluoroquinolone resistance in bloodstream infections caused by Escherichia coli in patients with hematological malignancies. J. Infect. 2009, 58, 299–307. [Google Scholar] [CrossRef]
  29. Satlin, M.J.; Cohen, N.; Ma, K.C.; Gedrimaite, Z.; Soave, R.; Askin, G.; Chen, L.; Kreiswirth, B.N.; Walsh, T.J.; Seo, S.K. Bacteremia due to carbapenem-resistant Enterobacteriaceae in neutropenic patients with hematologic malignancies. J. Infect. 2016, 73, 336–345. [Google Scholar] [CrossRef] [Green Version]
  30. Remolina, G.S.; Conde, M.C.; Escobar, C.J.; Leal, C.A.; Bravo, O.J.; Saavedra, R.S.; Rosa, N.Z.; Sanchez, F.N.; Santana, G.A.; Cortes, C.S.; et al. Carbapenemases produced in Klebsiella spp., and Pseudomonas aeruginosa in six hospitals in Bogota—Colombia. Rev. Chilena Infectol. 2021, 38, 720–723. [Google Scholar] [CrossRef]
  31. Boolchandani, M.; D’Souza, A.W.; Dantas, G. Sequencing-based methods and resources to study antimicrobial resistance. Nat. Rev. Genet. 2019, 20, 356–370. [Google Scholar] [CrossRef] [PubMed]
  32. Kang, C.I.; Song, J.H.; Chung, D.R.; Peck, K.R.; Yeom, J.S.; Son, J.S.; Wi, Y.M.; Korean Network for Study on Infectious, D. Bloodstream infections in adult patients with cancer: Clinical features and pathogenic significance of Staphylococcus aureus bacteremia. Support Care Cancer 2012, 20, 2371–2378. [Google Scholar] [CrossRef] [PubMed]
  33. Li, Z.; Zhuang, H.; Wang, G.; Wang, H.; Dong, Y. Prevalence, predictors, and mortality of bloodstream infections due to methicillin-resistant Staphylococcus aureus in patients with malignancy: Systemic review and meta-analysis. BMC Infect. Dis. 2021, 21, 74. [Google Scholar] [CrossRef] [PubMed]
  34. Paterson, G.K.; Harrison, E.M.; Holmes, M.A. The emergence of mecC methicillin-resistant Staphylococcus aureus. Trends Microbiol. 2014, 22, 42–47. [Google Scholar] [CrossRef] [Green Version]
  35. Rafey, A.; Nizamuddin, S.; Qureshi, W.; Anjum, A.; Parveen, A. Trends of Vancomycin-Resistant Enterococcus Infections in Cancer Patients. Cureus 2022, 14, e31335. [Google Scholar] [CrossRef]
  36. Cabiltes, I.; Coghill, S.; Bowe, S.J.; Athan, E. Enterococcal bacteraemia ‘silent but deadly’: A population-based cohort study. Intern. Med. J. 2020, 50, 434–440. [Google Scholar] [CrossRef] [Green Version]
  37. Bender, J.K.; Hermes, J.; Zabel, L.T.; Haller, S.; Murter, N.; Blank, H.P.; Werner, G.; Huttner, I.; Eckmanns, T. Controlling an Unprecedented Outbreak with Vancomycin-Resistant Enterococcus faecium in Germany, October 2015 to November 2019. Microorganisms 2022, 10, 1603. [Google Scholar] [CrossRef]
  38. Tumbarello, M.; Spanu, T.; Caira, M.; Trecarichi, E.M.; Laurenti, L.; Montuori, E.; Fianchi, L.; Leone, F.; Fadda, G.; Cauda, R.; et al. Factors associated with mortality in bacteremic patients with hematologic malignancies. Diagn. Microbiol. Infect. Dis. 2009, 64, 320–326. [Google Scholar] [CrossRef] [PubMed]
  39. Cuervo, S.I.; Cortes, J.A.; Sanchez, R.; Rodriguez, J.Y.; Silva, E.; Tibavizco, D.; Arroyo, P. Risk factors for mortality caused by Staphylococcus aureus bacteremia in cancer patients. Enferm. Infecc. Microbiol. Clin. 2010, 28, 349–354. [Google Scholar] [CrossRef] [PubMed]
  40. Cattaneo, C.; Antoniazzi, F.; Casari, S.; Ravizzola, G.; Gelmi, M.; Pagani, C.; D’Adda, M.; Morello, E.; Re, A.; Borlenghi, E.; et al. P. aeruginosa bloodstream infections among hematological patients: An old or new question? Ann. Hematol. 2012, 91, 1299–1304. [Google Scholar] [CrossRef]
  41. Kleinhendler, E.; Cohen, M.J.; Moses, A.E.; Paltiel, O.; Strahilevitz, J.; Cahan, A. Empiric antibiotic protocols for cancer patients with neutropenia: A single-center study of treatment efficacy and mortality in patients with bacteremia. Int. J. Antimicrob. Agents 2018, 51, 71–76. [Google Scholar] [CrossRef]
  42. Martinez-Nadal, G.; Puerta-Alcalde, P.; Gudiol, C.; Cardozo, C.; Albasanz-Puig, A.; Marco, F.; Laporte-Amargos, J.; Moreno-Garcia, E.; Domingo-Domenech, E.; Chumbita, M.; et al. Inappropriate Empirical Antibiotic Treatment in High-risk Neutropenic Patients With Bacteremia in the Era of Multidrug Resistance. Clin. Infect. Dis. 2020, 70, 1068–1074. [Google Scholar] [CrossRef] [PubMed]
  43. Kwon, K.T. Implementation of Antimicrobial Stewardship Programs in End-of-Life Care. Infect. Chemother. 2019, 51, 89–97. [Google Scholar] [CrossRef] [PubMed]
  44. Horino, T.; Chiba, A.; Kawano, S.; Kato, T.; Sato, F.; Maruyama, Y.; Nakazawa, Y.; Yoshikawa, K.; Yoshida, M.; Hori, S. Clinical characteristics and risk factors for mortality in patients with bacteremia caused by Pseudomonas aeruginosa. Intern. Med. 2012, 51, 59–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Barlam, T.F.; Cosgrove, S.E.; Abbo, L.M.; MacDougall, C.; Schuetz, A.N.; Septimus, E.J.; Srinivasan, A.; Dellit, T.H.; Falck-Ytter, Y.T.; Fishman, N.O.; et al. Implementing an Antibiotic Stewardship Program: Guidelines by the Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America. Clin. Infect. Dis. 2016, 62, e51–e77. [Google Scholar] [CrossRef] [PubMed]
Table 1. Characteristics of the patients included.
Table 1. Characteristics of the patients included.
Initial Clinical Variables and Historyn = 195
n (%)
Age (years, average)52 (SD 17.2)
Female109 (55.8)
Type of tumor
Solid tumor91 (46.7)
Hematological tumor104 (53.3)
Tumor status
Active tumor83 (42.5)
Tumor in palliative care13 (6.6)
De novo tumor41 (21)
Relapsed tumor43 (22)
Tumor in remission15 (7.7)
Treatment received
Chemotherapy (%)105 (53.8)
Radiation therapy (%)23 (11.8)
Surgery (%)49 (25.1)
Hematopoietic progenitor transplantation (%)6 (5.7)
Previous bacterial infection (%)52 (26.6)
Antibiotic during the previous month (%)70 (35.8)
Non-carbapenem beta-lactams43 (22)
Carbapenems18 (9.2)
Glycopeptides14 (7.1)
Quinolones4 (2.0)
Aminoglycosides2 (1)
Other9 (4.6)
≥2 antibiotics30 (15.3)
Steroid use10 (5.1)
Patient service
Hospitalization131 (67.1)
Emergency room56 (28.7)
Intensive care unit8 (4.1)
Implantable catheter24 (12.2%)
Device usage112 (57.4)
Central venous catheter69 (35.3)
Bladder catheter18 (9.2)
Nasogastric tube14 (7.1)
Drain10 (5.1)
Hemodialysis catheter3 (1.5)
Other32 (16.4)
Comorbidities
Diabetes mellitus17 (8.7)
Diabetes complications1 (0.5)
Chronic kidney disease16 (8.2)
Charlson index (median, IQR)3 (2–3)
Systolic blood pressure mm Hg (mean, SD)110 (21)
Diastolic blood pressure mm Hg (mean, SD)65 (13)
Heart rate (mean, SD)102 (20)
Temperature °C (mean, SD)37.5 (1.2)
Respiratory rate (mean, SD)20 (2)
Neutropenia63 (32.3)
Febrile neutropenia59 (30.2)
Systemic inflammatory response and sepsis119 (61)
Fever78 (40)
Table 2. Microbiological results of 195 patients with cancer and bacteremia.
Table 2. Microbiological results of 195 patients with cancer and bacteremia.
Microbiological Resultsn = 206 (%)
Gram-negative142 (68.9)
Escherichia coli67 (32.5)
Klebsiella pneumoniae36 (17.4)
Pseudomonas aeruginosa21 (10.1)
Other Enterobacteriaceae15 (7.2)
Acinetobacter2 (1)
Stenotrophomonas maltophilia1 (0.5)
Gram-positive64 (31)
Staphylococcus aureus25 (12.4)
Coagulase-negative Staphylococci26 (12.6)
Enterococcus faecium4 (1.9)
Enterococcus faecalis4 (1.9)
Other Gram-positive5 (2.4)
Natural profile (%)76 (36.9)
Acquired profile (%)130 (63.1)
Table 3. Percentage of resistance of selected Gram-negative bacteria.
Table 3. Percentage of resistance of selected Gram-negative bacteria.
AntibioticE. coli
n = 67
K. pneumoniae
n = 36
P. aeruginosa
n = 21
Ampicillin R/n (%R)42/57 (73)
Ampicillin/sulbactam R/n (%R)11/65 (16.4)18/35 (51.4)
Cefazolin R/n (%R)8/52 (15.3)6/21 (28.5)
Piperacillin/tazobactam R/n (%R)2/65 (3.1)14/35 (40)1/17 (5,8)
Ceftazidime R/n (%R)13/66 (19.7)15/35 (42.8)5/17 (29.4)
Ceftriaxone R/n (%R)13/66 (19.7)15/35 (42.8)
Cefepime R/n (%R)13/66 (19.7)15/35 (42.8)5/21 (23.8)
Ertapenem R/n (%R)012/35 (34.2)
Meropenem R/n (%R)012/35 (34.2)4/21 (19)
Amikacin R/n (%R)01/36 (2.7)3/21 (14.2)
Gentamicin R/n (%R)10/64 (15.6)5/34 (14.7)3/21 (14.2)
Ciprofloxacin R/n (%R)24/67 (35.8)6/35 (17.1)2/21 (9.5)
Trimethoprim–ulfamethoxazole R/n (%R)38/59 (64.4)11/23 (47.8)
Colistin R/n (%R) 0
R/n: antibiotic-resistant bacteria/antibiotic-tested isolates.
Table 4. Resistance genes identified in Gram-negative bacilli isolates in cancer patients according to species.
Table 4. Resistance genes identified in Gram-negative bacilli isolates in cancer patients according to species.
BacteriumGene IdentifiedTotal
E. coliblaCTX-M4
blaCTX-M, blaTEM7
blaCTX-M, blaTEM, blaSHV1
K. pneumoniaeblaTEM1
blaSHV1
blaCTX-M, blaTEM1
blaCTX-M, blaTEM, blaSHV1
blaKPC11
blaVIM1
P. aeruginosablaSHV1
blaKPC2
blaVIM1
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cruz-Vargas, S.A.; García-Muñoz, L.; Cuervo-Maldonado, S.I.; Álvarez-Moreno, C.A.; Saavedra-Trujillo, C.H.; Álvarez-Rodríguez, J.C.; Arango-Gutiérrez, A.; Gómez-Rincón, J.C.; García-Guzman, K.; Leal, A.L.; et al. Molecular and Clinical Data of Antimicrobial Resistance in Microorganisms Producing Bacteremia in a Multicentric Cohort of Patients with Cancer in a Latin American Country. Microorganisms 2023, 11, 359. https://doi.org/10.3390/microorganisms11020359

AMA Style

Cruz-Vargas SA, García-Muñoz L, Cuervo-Maldonado SI, Álvarez-Moreno CA, Saavedra-Trujillo CH, Álvarez-Rodríguez JC, Arango-Gutiérrez A, Gómez-Rincón JC, García-Guzman K, Leal AL, et al. Molecular and Clinical Data of Antimicrobial Resistance in Microorganisms Producing Bacteremia in a Multicentric Cohort of Patients with Cancer in a Latin American Country. Microorganisms. 2023; 11(2):359. https://doi.org/10.3390/microorganisms11020359

Chicago/Turabian Style

Cruz-Vargas, Sergio Andrés, Laura García-Muñoz, Sonia Isabel Cuervo-Maldonado, Carlos Arturo Álvarez-Moreno, Carlos Humberto Saavedra-Trujillo, José Camilo Álvarez-Rodríguez, Angélica Arango-Gutiérrez, Julio César Gómez-Rincón, Katherine García-Guzman, Aura Lucía Leal, and et al. 2023. "Molecular and Clinical Data of Antimicrobial Resistance in Microorganisms Producing Bacteremia in a Multicentric Cohort of Patients with Cancer in a Latin American Country" Microorganisms 11, no. 2: 359. https://doi.org/10.3390/microorganisms11020359

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

Article Metrics

Back to TopTop