Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Nat Commun ; 15(1): 2758, 2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38553439

RESUMEN

Hospital surfaces can harbour bacterial pathogens, which may disseminate and cause nosocomial infections, contributing towards mortality in low- and middle-income countries (LMICs). During the BARNARDS study, hospital surfaces from neonatal wards were sampled to assess the degree of environmental surface and patient care equipment colonisation by Gram-negative bacteria (GNB) carrying antibiotic resistance genes (ARGs). Here, we perform PCR screening for extended-spectrum ß-lactamases (blaCTX-M-15) and carbapenemases (blaNDM, blaOXA-48-like and blaKPC), MALDI-TOF MS identification of GNB carrying ARGs, and further analysis by whole genome sequencing of bacterial isolates. We determine presence of consistently dominant clones and their relatedness to strains causing neonatal sepsis. Higher prevalence of carbapenemases is observed in Pakistan, Bangladesh, and Ethiopia, compared to other countries, and are mostly found in surfaces near the sink drain. Klebsiella pneumoniae, Enterobacter hormaechei, Acinetobacter baumannii, Serratia marcescens and Leclercia adecarboxylata are dominant; ST15 K. pneumoniae is identified from the same ward on multiple occasions suggesting clonal persistence within the same environment, and is found to be identical to isolates causing neonatal sepsis in Pakistan over similar time periods. Our data suggests persistence of dominant clones across multiple time points, highlighting the need for assessment of Infection Prevention and Control guidelines.


Asunto(s)
Países en Desarrollo , Sepsis Neonatal , Recién Nacido , Humanos , beta-Lactamasas/genética , Proteínas Bacterianas/genética , Hospitales , Antibacterianos/farmacología , Klebsiella pneumoniae/genética , Bacterias Gramnegativas/genética , Pruebas de Sensibilidad Microbiana
2.
Lancet Infect Dis ; 23(11): 1280-1290, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37454672

RESUMEN

BACKGROUND: Bacteria of the order Enterobacterales are common pathogens causing bloodstream infections in sub-Saharan Africa and are frequently resistant to third-generation cephalosporin antibiotics. Although third-generation cephalosporin resistance is believed to lead to adverse outcomes, this relationship is difficult to quantify and has rarely been studied in this region. We aimed to measure the effects associated with resistance to third-generation cephalosporins in hospitalised patients with Enterobacterales bloodstream infection in Africa. METHODS: We conducted a prospective, matched, parallel cohort study at eight hospitals across sub-Saharan Africa. We recruited consecutive patients of all age groups with laboratory-confirmed Enterobacterales bloodstream infection and matched them to at least one patient without bloodstream infection on the basis of age group, hospital ward, and admission date. Date of infection onset (and enrolment) was defined as the day of blood sample collection for culturing. Patients infected with bacteria with a cefotaxime minimum inhibitory concentration of 1 mg/L or lower were included in the third-generation cephalosporin-susceptible (3GC-S) cohort, and the remainder were included in the third-generation cephalosporin-resistant (3GC-R) cohort. The primary outcomes were in-hospital death and death within 30 days of enrolment. We used adjusted multivariable regression models to first compare patients with bloodstream infection against matched patients within the 3GC-S and 3GC-R cohorts, then compared estimates between cohorts. FINDINGS: Between Nov 1, 2020, and Jan 31, 2022, we recruited 878 patients with Enterobacterales bloodstream infection (221 [25·2%] to the 3GC-S cohort and 657 [74·8%] to the 3GC-R cohort) and 1634 matched patients (420 [25·7%] and 1214 [74·3%], respectively). 502 (57·2%) bloodstream infections occurred in neonates and infants (age 0-364 days). Klebsiella pneumoniae (393 [44·8%] infections) and Escherichia coli (224 [25·5%] infections) were the most common Enterobacterales species identified. The proportion of patients who died in hospital was higher in patients with bloodstream infection than in matched controls in the 3GC-S cohort (62 [28·1%] of 221 vs 22 [5·2%] of 420; cause-specific hazard ratio 6·79 [95% CI 4·06-11·37] from Cox model) and the 3GC-R cohort (244 [37·1%] of 657 vs 115 [9·5%] of 1214; 5·01 [3·96-6·32]). The ratio of these cause-specific hazard ratios showed no significant difference in risk of in-hospital death in the 3GC-R cohort versus the 3GC-S cohort (0·74 [0·42-1·30]). The ratio of relative risk of death within 30 days (0·82 [95% CI 0·53-1·27]) also indicated no difference between the cohorts. INTERPRETATION: Patients with bloodstream infections with Enterobacterales bacteria either resistant or susceptible to third-generation cephalosporins had increased mortality compared with uninfected matched patients, with no differential effect related to third-generation cephalosporin-resistance status. However, this finding does not account for time to appropriate antibiotic treatment, which remains clinically important to optimise. Measures to prevent transmission of Enterobacterales could reduce bloodstream infection-associated mortality from both drug-resistant and drug-susceptible bacterial strains in Africa. FUNDING: Bill & Melinda Gates Foundation.


Asunto(s)
Cefalosporinas , Sepsis , Recién Nacido , Humanos , Cefalosporinas/farmacología , Cefalosporinas/uso terapéutico , Estudios Prospectivos , Resistencia a las Cefalosporinas , Estudios de Cohortes , Mortalidad Hospitalaria , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Escherichia coli , Sepsis/tratamiento farmacológico , Hospitales
3.
Gates Open Res ; 7: 21, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38532981

RESUMEN

Background: Bloodstream infections caused by Enterobacterales show high frequency of antimicrobial resistance (AMR) in many Low- and Middle-Income Countries. We aimed to describe the variation in circumstances for management of such resistant infections in a group of African public-sector hospitals participating in a major research study. Methods: We gathered data from eight hospitals across sub-Saharan Africa to describe hospital services, infection prevention and antibiotic stewardship activities, using two WHO-generated tools. We collected monthly cross-sectional data on availability of antibiotics in the hospital pharmacies for bloodstream infections caused by Enterobacterales. We compared the availability of these antibiotics to actual patient-level use of antibiotics in confirmed Enterobacterales bloodstream infections (BSI). Results: Hospital circumstances for institutional management of resistant BSI varied markedly. This included self-evaluated infection prevention level (WHO-IPCAF score: median 428, range 155 to 687.5) and antibiotic stewardship activities (WHO stewardship toolkit questions: median 14.5, range 2 to 23). These results did not correlate with national income levels. Across all sites, ceftriaxone and ciprofloxacin were the most consistently available antibiotic agents, followed by amoxicillin, co-amoxiclav, gentamicin and co-trimoxazole. There was substantial variation in the availability of some antibiotics, especially carbapenems, amikacin and piperacillin-tazobactam with degree of access linked to national income level. Investigators described out-of-pocket payments for access to additional antibiotics at 7/8 sites. The in-pharmacy availability of antibiotics correlated well with actual use of antibiotics for treating BSI patients. Conclusions: There was wide variation between these African hospitals for a range of important circumstances relating to treatment and control of severe bacterial infections, though these did not all correspond to national income level. For most antibiotics, patient-level use reflected in-hospital drug availability, suggesting external antibiotics supply was infrequent. Antimicrobial resistant bacterial infections could plausibly show different clinical impacts across sub-Saharan Africa due to this contextual variation.


Asunto(s)
Infecciones Bacterianas , Sepsis , Humanos , Estudios Transversales , Antibacterianos/uso terapéutico , Hospitales , Infecciones Bacterianas/tratamiento farmacológico , África del Sur del Sahara , Sepsis/tratamiento farmacológico
4.
Sci Rep ; 12(1): 14822, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-36050365

RESUMEN

Infections caused by multi-drug resistant Escherichia coli cause significant morbidity and mortality especially in developing countries. In this study, we describe the molecular characteristics of E. coli isolated from clinical specimens and the patients' outcomes. Phenotypic methods were used in the identification and antimicrobial susceptibility testing of E. coli from clinical specimens from a tertiary hospital in Abuja, Nigeria. Whole genome sequencing was used to describe the antimicrobial resistance genes, serotypes, sequence types/clonal complexes, and mobile genetic elements. The mean age of the patients was 20.3 years with 70.1% females and majority of isolates 75% from urine, 21% from blood cultures, and 3% each from cerebrospinal fluid and endo-cervical swabs. Of the 107 non-duplicate E. coli isolates, 101 (94.3%) were resistant to ampicillin, 95 (88.8%) to trimethoprim/sulfamethoxazole, 86 (80.4%) to ceftriaxone, 60 (56.1%) to gentamicin, and eight (7.5%) to meropenem. There were 102 (95.3%) isolates that were multi-drug resistant (MDR). Expression of Extended Spectrum Beta Lactamase (ESBL) phenotype was detected in 54 (50%) and blaCTX-M-15 genes detected in 75 (70.1%) isolates. The carbapenemase genes blaNDM-1 and blaNDM-5 were detected in six (5.6%), while the AmpC gene- blaCMY-2, was detected in seven (6.5%) isolates. Two (1.9%) isolates simultaneously harboured the blaOXA-1, blaCMY-2, blaCTX-M-15, and blaNDM-5 genes. In total, 35 sequence types (STs) were found with the majority being ST131 (n = 23; 21.5%). The most common serotype was O25:H4 associated with all 23 strains of ST131, followed by O1:H6/ST648 (n = 6). The ST410, ST671, and ST101 strains displayed phenotypic resistance to wide array of antibiotic classes and harbored high numbers of antibiotic resistance genes via in-silico analysis. The ST410 strain in particular harbored a higher number of antibiotic resistance genes and was phenotypically resistant to a wider array of antibiotics. Four pairs of isolates were closely related with three isolates (ST131, ST38, ST652) having a pairwise SNP difference of zero. 71/72 75/76 52/14. The MDR E. coli lineages circulating in this setting pose a clinical and public health threat as they can hinder effective prevention and management of infections. The genetic diversity and MDR E. coli with the emergence of ST410 and ST101 clones is concerning because of the potential for rapid dissemination in hospitals and communities- further increasing the problems of antibiotic resistance. Continuous routine surveillance of E. coli infections for AMR in hospitals becomes imperative, aimed at development of effective antimicrobial stewardship programs, facilitating prudent use of antimicrobial agents, and limiting dissemination of resistant strains.


Asunto(s)
Farmacorresistencia Bacteriana Múltiple , Infecciones por Escherichia coli , Escherichia coli , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Farmacorresistencia Bacteriana Múltiple/genética , Escherichia coli/genética , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/epidemiología , Proteínas de Escherichia coli/genética , Femenino , Humanos , Masculino , Pruebas de Sensibilidad Microbiana , Nigeria/epidemiología , Centros de Atención Terciaria , beta-Lactamasas/genética , beta-Lactamasas/metabolismo
5.
BMC Infect Dis ; 22(1): 593, 2022 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-35790903

RESUMEN

BACKGROUND: In low- and middle-income countries (LMIC) Staphylococcus aureus is regarded as one of the leading bacterial causes of neonatal sepsis, however there is limited knowledge on the species diversity and antimicrobial resistance caused by Gram-positive bacteria (GPB). METHODS: We characterised GPB isolates from neonatal blood cultures from LMICs in Africa (Ethiopia, Nigeria, Rwanda, and South Africa) and South-Asia (Bangladesh and Pakistan) between 2015-2017. We determined minimum inhibitory concentrations and performed whole genome sequencing (WGS) on Staphylococci isolates recovered and clinical data collected related to the onset of sepsis and the outcome of the neonate up to 60 days of age. RESULTS: From the isolates recovered from blood cultures, Staphylococci species were most frequently identified. Out of 100 S. aureus isolates sequenced, 18 different sequence types (ST) were found which unveiled two small epidemiological clusters caused by methicillin resistant S. aureus (MRSA) in Pakistan (ST8) and South Africa (ST5), both with high mortality (n = 6/17). One-third of S. aureus was MRSA, with methicillin resistance also detected in Staphylococcus epidermidis, Staphylococcus haemolyticus and Mammaliicoccus sciuri. Through additional WGS analysis we report a cluster of M. sciuri in Pakistan identified between July-November 2017. CONCLUSIONS: In total we identified 14 different GPB bacterial species, however Staphylococci was dominant. These findings highlight the need of a prospective genomic epidemiology study to comprehensively assess the true burden of GPB neonatal sepsis focusing specifically on mechanisms of resistance and virulence across species and in relation to neonatal outcome.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Sepsis Neonatal , Cultivo de Sangre , Países en Desarrollo , Etiopía , Humanos , Recién Nacido , Sepsis Neonatal/epidemiología , Estudios Prospectivos , Staphylococcus aureus/genética
7.
Lancet Glob Health ; 10(5): e661-e672, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35427523

RESUMEN

BACKGROUND: Neonatal sepsis is a primary cause of neonatal mortality and is an urgent global health concern, especially within low-income and middle-income countries (LMICs), where 99% of global neonatal mortality occurs. The aims of this study were to determine the incidence and associations with neonatal sepsis and all-cause mortality in facility-born neonates in LMICs. METHODS: The Burden of Antibiotic Resistance in Neonates from Developing Societies (BARNARDS) study recruited mothers and their neonates into a prospective observational cohort study across 12 clinical sites from Bangladesh, Ethiopia, India, Pakistan, Nigeria, Rwanda, and South Africa. Data for sepsis-associated factors in the four domains of health care, maternal, birth and neonatal, and living environment were collected for all mothers and neonates enrolled. Primary outcomes were clinically suspected sepsis, laboratory-confirmed sepsis, and all-cause mortality in neonates during the first 60 days of life. Incidence proportion of livebirths for clinically suspected sepsis and laboratory-confirmed sepsis and incidence rate per 1000 neonate-days for all-cause mortality were calculated. Modified Poisson regression was used to investigate factors associated with neonatal sepsis and parametric survival models for factors associated with all-cause mortality. FINDINGS: Between Nov 12, 2015 and Feb 1, 2018, 29 483 mothers and 30 557 neonates were enrolled. The incidence of clinically suspected sepsis was 166·0 (95% CI 97·69-234·24) per 1000 livebirths, laboratory-confirmed sepsis was 46·9 (19·04-74·79) per 1000 livebirths, and all-cause mortality was 0·83 (0·37-2·00) per 1000 neonate-days. Maternal hypertension, previous maternal hospitalisation within 12 months, average or higher monthly household income, ward size (>11 beds), ward type (neonatal), living in a rural environment, preterm birth, perinatal asphyxia, and multiple births were associated with an increased risk of clinically suspected sepsis, laboratory-confirmed sepsis, and all-cause mortality. The majority (881 [72·5%] of 1215) of laboratory-confirmed sepsis cases occurred within the first 3 days of life. INTERPRETATION: Findings from this study highlight the substantial proportion of neonates who develop neonatal sepsis, and the high mortality rates among neonates with sepsis in LMICs. More efficient and effective identification of neonatal sepsis is needed to target interventions to reduce its incidence and subsequent mortality in LMICs. FUNDING: Bill & Melinda Gates Foundation.


Asunto(s)
Sepsis Neonatal , Nacimiento Prematuro , Sepsis , Países en Desarrollo , Femenino , Humanos , Mortalidad Infantil , Recién Nacido , Sepsis Neonatal/epidemiología , Embarazo , Estudios Prospectivos , Sepsis/epidemiología
8.
Infect Drug Resist ; 15: 933-946, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35299860

RESUMEN

Neonatal sepsis is defined as a systemic infection within the first 28 days of life, with early-onset sepsis (EOS) occurring within the first 72h, although the definition of EOS varies in literature. Whilst the global incidence has dramatically reduced over the last decade, neonatal sepsis remains an important cause of neonatal mortality, highest in low- and middle-income countries (LMICs). Symptoms at the onset of neonatal sepsis can be subtle, and therefore EOS is often difficult to diagnose from clinical presentation and laboratory testing and blood cultures are not always conclusive or accessible, especially in resource limited countries. Although the World Health Organisation (WHO) currently advocates a ß-lactam, and gentamicin for first line treatment, availability and cost influence the empirical antibiotic therapy administered. Antibiotic treatment of neonatal sepsis in LMICs is highly variable, partially caused by factors such as cost of antibiotics (and who pays for them) and access to certain antibiotics. Antimicrobial resistance (AMR) has increased considerably over the past decade and this review discusses current microbiology data available in the context of the diagnosis, and treatment for EOS. Importantly, this review highlights a large variability in data availability, methodology, availability of diagnostics, and aetiology of sepsis pathogens.

9.
Lancet Infect Dis ; 22(5): e143-e152, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34914924

RESUMEN

Neonates and children in low-income and middle-income countries (LMICs) contribute to the highest number of sepsis-associated deaths globally. Interventions to prevent sepsis mortality are hampered by a lack of comprehensive epidemiological data and pathophysiological understanding of biological pathways. In this review, we discuss the challenges faced by LMICs in diagnosing sepsis in these age groups. We highlight a role for multi-omics and health care data to improve diagnostic accuracy of clinical algorithms, arguing that health-care systems urgently need precision medicine to avoid the pitfalls of missed diagnoses, misdiagnoses, and overdiagnoses, and associated antimicrobial resistance. We discuss ethical, regulatory, and systemic barriers related to the collection and use of big data in LMICs. Technologies such as cloud computing, artificial intelligence, and medical tricorders might help, but they require collaboration with local communities. Co-partnering (joint equal development of technology between producer and end-users) could facilitate integration of these technologies as part of future care-delivery systems, offering a chance to transform the global management and prevention of sepsis for neonates and children.


Asunto(s)
Ciencia de los Datos , Sepsis , Inteligencia Artificial , Niño , Países en Desarrollo , Salud Global , Humanos , Recién Nacido , Sepsis/diagnóstico
10.
Lancet Infect Dis ; 21(12): 1677-1688, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34384533

RESUMEN

BACKGROUND: Sepsis is a major contributor to neonatal mortality, particularly in low-income and middle-income countries (LMICs). WHO advocates ampicillin-gentamicin as first-line therapy for the management of neonatal sepsis. In the BARNARDS observational cohort study of neonatal sepsis and antimicrobial resistance in LMICs, common sepsis pathogens were characterised via whole genome sequencing (WGS) and antimicrobial resistance profiles. In this substudy of BARNARDS, we aimed to assess the use and efficacy of empirical antibiotic therapies commonly used in LMICs for neonatal sepsis. METHODS: In BARNARDS, consenting mother-neonates aged 0-60 days dyads were enrolled on delivery or neonatal presentation with suspected sepsis at 12 BARNARDS clinical sites in Bangladesh, Ethiopia, India, Pakistan, Nigeria, Rwanda, and South Africa. Stillborn babies were excluded from the study. Blood samples were collected from neonates presenting with clinical signs of sepsis, and WGS and minimum inhibitory concentrations for antibiotic treatment were determined for bacterial isolates from culture-confirmed sepsis. Neonatal outcome data were collected following enrolment until 60 days of life. Antibiotic usage and neonatal outcome data were assessed. Survival analyses were adjusted to take into account potential clinical confounding variables related to the birth and pathogen. Additionally, resistance profiles, pharmacokinetic-pharmacodynamic probability of target attainment, and frequency of resistance (ie, resistance defined by in-vitro growth of isolates when challenged by antibiotics) were assessed. Questionnaires on health structures and antibiotic costs evaluated accessibility and affordability. FINDINGS: Between Nov 12, 2015, and Feb 1, 2018, 36 285 neonates were enrolled into the main BARNARDS study, of whom 9874 had clinically diagnosed sepsis and 5749 had available antibiotic data. The four most commonly prescribed antibiotic combinations given to 4451 neonates (77·42%) of 5749 were ampicillin-gentamicin, ceftazidime-amikacin, piperacillin-tazobactam-amikacin, and amoxicillin clavulanate-amikacin. This dataset assessed 476 prescriptions for 442 neonates treated with one of these antibiotic combinations with WGS data (all BARNARDS countries were represented in this subset except India). Multiple pathogens were isolated, totalling 457 isolates. Reported mortality was lower for neonates treated with ceftazidime-amikacin than for neonates treated with ampicillin-gentamicin (hazard ratio [adjusted for clinical variables considered potential confounders to outcomes] 0·32, 95% CI 0·14-0·72; p=0·0060). Of 390 Gram-negative isolates, 379 (97·2%) were resistant to ampicillin and 274 (70·3%) were resistant to gentamicin. Susceptibility of Gram-negative isolates to at least one antibiotic in a treatment combination was noted in 111 (28·5%) to ampicillin-gentamicin; 286 (73·3%) to amoxicillin clavulanate-amikacin; 301 (77·2%) to ceftazidime-amikacin; and 312 (80·0%) to piperacillin-tazobactam-amikacin. A probability of target attainment of 80% or more was noted in 26 neonates (33·7% [SD 0·59]) of 78 with ampicillin-gentamicin; 15 (68·0% [3·84]) of 27 with amoxicillin clavulanate-amikacin; 93 (92·7% [0·24]) of 109 with ceftazidime-amikacin; and 70 (85·3% [0·47]) of 76 with piperacillin-tazobactam-amikacin. However, antibiotic and country effects could not be distinguished. Frequency of resistance was recorded most frequently with fosfomycin (in 78 isolates [68·4%] of 114), followed by colistin (55 isolates [57·3%] of 96), and gentamicin (62 isolates [53·0%] of 117). Sites in six of the seven countries (excluding South Africa) stated that the cost of antibiotics would influence treatment of neonatal sepsis. INTERPRETATION: Our data raise questions about the empirical use of combined ampicillin-gentamicin for neonatal sepsis in LMICs because of its high resistance and high rates of frequency of resistance and low probability of target attainment. Accessibility and affordability need to be considered when advocating antibiotic treatments with variance in economic health structures across LMICs. FUNDING: The Bill & Melinda Gates Foundation.


Asunto(s)
Antibacterianos/uso terapéutico , Farmacorresistencia Microbiana , Infecciones por Enterobacteriaceae/tratamiento farmacológico , Sepsis Neonatal/tratamiento farmacológico , Sepsis Neonatal/microbiología , Infecciones Estafilocócicas/tratamiento farmacológico , Antibacterianos/economía , Estudios de Cohortes , Quimioterapia Combinada , Enterobacteriaceae/patogenicidad , Humanos , Recién Nacido , Staphylococcus aureus/patogenicidad , Virulencia
11.
Nat Microbiol ; 6(4): 512-523, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33782558

RESUMEN

Antimicrobial resistance in neonatal sepsis is rising, yet mechanisms of resistance that often spread between species via mobile genetic elements, ultimately limiting treatments in low- and middle-income countries (LMICs), are poorly characterized. The Burden of Antibiotic Resistance in Neonates from Developing Societies (BARNARDS) network was initiated to characterize the cause and burden of antimicrobial resistance in neonatal sepsis for seven LMICs in Africa and South Asia. A total of 36,285 neonates were enrolled in the BARNARDS study between November 2015 and December 2017, of whom 2,483 were diagnosed with culture-confirmed sepsis. Klebsiella pneumoniae (n = 258) was the main cause of neonatal sepsis, with Serratia marcescens (n = 151), Klebsiella michiganensis (n = 117), Escherichia coli (n = 75) and Enterobacter cloacae complex (n = 57) also detected. We present whole-genome sequencing, antimicrobial susceptibility and clinical data for 916 out of 1,038 neonatal sepsis isolates (97 isolates were not recovered from initial isolation at local sites). Enterobacterales (K. pneumoniae, E. coli and E. cloacae) harboured multiple cephalosporin and carbapenem resistance genes. All isolated pathogens were resistant to multiple antibiotic classes, including those used to treat neonatal sepsis. Intraspecies diversity of K. pneumoniae and E. coli indicated that multiple antibiotic-resistant lineages cause neonatal sepsis. Our results will underpin research towards better treatments for neonatal sepsis in LMICs.


Asunto(s)
Farmacorresistencia Bacteriana Múltiple , Bacterias Gramnegativas/patogenicidad , Infecciones por Bacterias Gramnegativas/microbiología , Sepsis Neonatal/microbiología , África/epidemiología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Asia/epidemiología , Proteínas Bacterianas/genética , Países en Desarrollo , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/genética , Variación Genética , Genoma Bacteriano/genética , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/genética , Bacterias Gramnegativas/aislamiento & purificación , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Infecciones por Bacterias Gramnegativas/mortalidad , Humanos , Recién Nacido , Sepsis Neonatal/tratamiento farmacológico , Sepsis Neonatal/mortalidad , Filogenia , Plásmidos/genética , beta-Lactamasas/genética
12.
Niger J Clin Pract ; 24(2): 225-232, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33605913

RESUMEN

BACKGROUND: Infections with Staphylococcus aureus cause significant morbidity and mortality worldwide. Resistant strains of S. aureus to commonly used antibiotics are being increasingly encountered in clinical practice, necessitating the need to determine the resistance pattern in Nigeria. METHODS: Antibiotic susceptibility testing was performed on 360 S. aureus isolates from clinical specimen from seven hospitals across the six geo-political regions of Nigeria using Kirby Bauer disc diffusion technique, and E-test for vancomycin. Cefoxitin 30 µg disc was used to determine methicillin resistance, and D-test for inducible clindamycin resistance. RESULTS: Methicillin-resistant S. aureus was confirmed in 176 (48.9%) of the isolates, 346 (96%) for penicillin G and 311 (86.4%) for trimethoprim. 175 (99.4%) of the 176 resistant to methicillin were susceptible to vancomycin. Linezolid, tigecycline, chloramphenicol and clindamycin had susceptibilities of 341 (94.7%), 332 (92.2%), 298 (82.8%) and 290 (80.6%) respectively. Inducible clindamycin resistance was elucidated in 25 (29.1%) of the 86 isolates. Generally, MRSA isolates were more resistant than methicillin-sensitive S. aureus (MSSA) to all antibiotics tested. CONCLUSION: Staphylococcus aureus rates of resistance are high and call for urgent action such as antibiotic stewardship programmes and periodic surveillance to enhance clinical outcomes. While targeted therapy is preferred, options for empiric treatment include chloramphenicol, clindamycin, linezolid or vancomycin.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Staphylococcus aureus , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Hospitales , Humanos , Pruebas de Sensibilidad Microbiana , Nigeria
13.
PLoS One ; 16(2): e0246644, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33556108

RESUMEN

OBJECTIVES: To determine if the presence of microorganisms in follicular fluid of women undergoing in vitro fertilization-embryo transfer (IVF-ET) adversely affects the outcome of the treatment cycles. METHODS: Follicular fluid was collected from 86 women enrolled for IVF-ET at the National Hospital, Abuja from June 1, 2018 to December 1, 2018. Microscopy imaging and cultures were performed to identify bacteria and fungi in the follicular fluid and the vagina. Women with follicular fluid microorganisms were the test group while the controls were those without follicular fluid microorganisms. Fertilization and pregnancy rates were subsequently determined and their association with the presence of follicular fluid microorganisms was assessed using univariable and multivariable logistic regression modelling. RESULTS: The mean age and mean Body Mass Index (BMI) of the participants were 35(± 3.5) years and 28(± 4.9) kg/m2 respectively. Bacteria and fungi were isolated in 17% of the follicular fluid samples collected (n = 15/86). Most common isolates were Streptococcus spp. (n = 4/15), Staphylococcus aureus (n = 7/15), Enterococcus spp. (n = 4/15), Lactobacillus species (n = 2/15) and Candida albicans (n = 2/15). There was no statistically significant difference in the fertilization rates (adjusted odds ratio [AdjOR] 0.55, 95% confidence interval [CI] 0.04-7.34; P = 0.10) and pregnancy rates (AdjOR 4.02, 95% CI 0.56-28.92; P = 0.19) between the group of women with positive follicular fluid bacterial /fungal colonization as compared against those with negative follicular fluid colonization. CONCLUSION: Isolation of microorganisms from follicular fluid did not adversely affect fertilization and pregnancy rates following IVF-ET treatment cycles at the fertility centre of National Hospital, Abuja.


Asunto(s)
Transferencia de Embrión/métodos , Fertilización In Vitro/métodos , Líquido Folicular/microbiología , Vagina/microbiología , Adulto , Femenino , Humanos , Infertilidad Femenina/terapia , Nigeria/epidemiología , Oportunidad Relativa , Oocitos/citología , Oocitos/fisiología , Embarazo , Índice de Embarazo/tendencias
14.
Front Glob Womens Health ; 2: 788157, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35098214

RESUMEN

BACKGROUND: Stillbirths are a poignant representation of global inequality. Nigeria is documented to have the second highest rate; yet, the reporting system is inadequate in most Nigerian healthcare facilities. The aim was to identify the determinants of stillbirth among deliveries in the Murtala Muhammad Specialist Hospital (MMSH), Kano, Nigeria. METHODS: Two study designs were used: a case-control study (S1) and a prospective cohort study (S2). Both studies were carried out at the MMSH. For S1, stillbirths were retrospectively matched to a livebirth by time (target of 24 hours' time variation) to establish a case-control study with a 1:1 ratio. Eligibility into S2 included all mothers who were presented at the MMSH in labour regardless of birth outcome. Both were based on recruitment durations, not sample sizes (3 months and 2 months, respectively, 2017-2018). The demographic and clinical data were collected through paper-based questionnaires. Univariable logistic regression was used. Multivariable logistic regression was used to explore relationships between area type and other specific factors. FINDINGS: Stillbirth incidence in S2 was 180/1,000 births. Stillbirth was associated with the following factors; no maternal education, previous stillbirth(s), prematurity, living in both semi-rural and rural settings, and having extended time periods between rupture of membranes and delivery. Findings of the multivariable analysis (S1 and S2) indicated that the odds of stillbirth, for those living in a rural area, were further exacerbated in those mothers who had no education, lived in a shack, or had any maternal disease. INTERPRETATION: This research identifies the gravity of this situation in this area and highlights the need for action. Further understanding of some of the findings and exploration into associations are required to inform intervention development. FUNDING: This collaboration was partially supported by funding from Health and Care Research Wales.

16.
Int J Gynaecol Obstet ; 148(3): 338-343, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31833060

RESUMEN

OBJECTIVE: To determine whether intermittent preventive therapy in pregnancy (IPTp) eradicates peripheral and placental malaria and improves birth weight. METHOD: A cross-sectional study was conducted of 426 pregnant mothers on IPTp with sulphadoxine-pyrimethamine against malaria who presented in labor, at National Hospital Abuja, Nigeria between January and June 2017. The hospital is within the malaria-endemic zone of West Africa. Consenting pregnant women with uncomplicated singleton term pregnancy who had antenatal care in the hospital and lived in the study area for at least 6 months were consecutively recruited. Peripheral and placental blood were collected and examined for malaria parasite by microscopy. Babies were weighed at birth. RESULTS: The prevalence of peripheral malaria parasitemia and placental parasitization were 12.9% (95% confidence interval [CI] 10.0-16.6) and 9.4% (95% CI 7.0-12.7), respectively. Parasite density in both peripheral parasitemia and placental parasitization was low among the women that took IPTp, decreasing with increasing doses, with no parasitemia or parasitization in women that took up to three doses. Birth weight was lower in babies of mothers with plasmodium infestation than in those without infestation (P<0.001, P=0.024). CONCLUSION: IPTp reduces both peripheral parasitemia and placental parasitization, with the capacity to eliminate or prevent them. IPTp also reduces low birth weight.


Asunto(s)
Antimaláricos/uso terapéutico , Malaria/prevención & control , Parasitemia/prevención & control , Placenta/microbiología , Complicaciones Parasitarias del Embarazo/prevención & control , Pirimetamina/uso terapéutico , Sulfadoxina/uso terapéutico , Adolescente , Adulto , Peso al Nacer , Estudios Transversales , Combinación de Medicamentos , Femenino , Humanos , Recién Nacido , Nigeria/epidemiología , Parasitemia/epidemiología , Embarazo , Complicaciones Parasitarias del Embarazo/epidemiología , Prevalencia , Centros de Atención Terciaria
17.
Lancet Infect Dis ; 19(6): 601-610, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31047852

RESUMEN

BACKGROUND: Low-income and middle-income countries (LMICs) are under-represented in reports on the burden of antimicrobial resistance. We aimed to quantify the clinical effect of carbapenem resistance on mortality and length of hospital stay among inpatients in LMICs with a bloodstream infection due to Enterobacteriaceae. METHODS: The PANORAMA study was a multinational prospective cohort study at tertiary hospitals in Bangladesh, Colombia, Egypt, Ghana, India, Lebanon, Nepal, Nigeria, Pakistan, and Vietnam, recruiting consecutively diagnosed patients with carbapenem-susceptible Enterobacteriaceae (CSE) and carbapenem-resistant Entero-bacteriaceae (CRE) bloodstream infections. We excluded patients who had previously been enrolled in the study and those not treated with curative intent at the time of bloodstream infection onset. There were no age restrictions. Central laboratories in India and the UK did confirmatory testing and molecular characterisation, including strain typing. We applied proportional subdistribution hazard models with inverse probability weighting to estimate the effect of carbapenem resistance on probability of discharge alive and in-hospital death, and multistate modelling for excess length of stay in hospital. All patients were included in the analysis. FINDINGS: Between Aug 1, 2014, and June 30, 2015, we recruited 297 patients from 16 sites in ten countries: 174 with CSE bloodstream infection and 123 with CRE bloodstream infection. Median age was 46 years (IQR 15-61). Crude mortality was 20% (35 of 174 patients) for patients with CSE bloodstream infection and 35% (43 of 123 patients) for patients with CRE bloodstream infection. Carbapenem resistance was associated with an increased length of hospital stay (3·7 days, 95% CI 0·3-6·9), increased probability of in-hospital mortality (adjusted subdistribution hazard ratio 1·75, 95% CI 1·04-2·94), and decreased probability of discharge alive (0·61, 0·45-0·83). Multilocus sequence typing showed various clades, with marginal overlap between strains in the CRE and CSE clades. INTERPRETATION: Carbapenem resistance is associated with increased length of hospital stay and mortality in patients with bloodstream infections in LMICs. These data will inform global estimates of the burden of antimicrobial resistance and reinforce the need for better strategies to prevent, diagnose, and treat CRE infections in LMICs. FUNDING: bioMérieux.


Asunto(s)
Antibacterianos/uso terapéutico , Bacteriemia/tratamiento farmacológico , Enterobacteriaceae Resistentes a los Carbapenémicos/efectos de los fármacos , Carbapenémicos/uso terapéutico , Infecciones por Enterobacteriaceae/tratamiento farmacológico , Enfermedades Hematológicas/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Bacteriemia/epidemiología , Estudios de Cohortes , Países en Desarrollo , Infecciones por Enterobacteriaceae/epidemiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Estudios Retrospectivos
18.
PLoS One ; 13(7): e0200350, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30016358

RESUMEN

BACKGROUND: Group B Streptococcus (GBS) causes invasive infections in neonates and has been implicated as a cause of prelabour rupture of membranes, preterm delivery and stillbirths. The success of phase II trials of polyvalent polysaccharide GBS vaccines indicates that these infections are potentially preventable. Nigeria is the most populous country in Africa with one of the highest birth rates, one of the highest neonatal sepsis incidence rates and one of the highest mortality rates in the world. Therefore, before the possible introduction of preventive strategies such as intrapartum antibiotic prophylaxis or GBS vaccine into Nigeria, it is vital that there is accurate data on the aetiology of neonatal sepsis and on the incidence of GBS neonatal sepsis in particular. The objective of this study was to determine the incidence and aetiology of neonatal sepsis in Nigeria with a focus on GBS sepsis and also to assess the potential impact of a GBS vaccine. METHODS: A literature search was conducted on the databases of African journals online, PubMed and Google Scholar for works conducted between 1987 to 2017. Case reports, reviews, and studies not stating specific culture methods or specific bacteria isolated were excluded. Data extracted included; incidence of neonatal sepsis, method of blood culture, blood volume, sample size, bacterial agents isolated and history of antibiotic use. PRISMA guidelines were followed and modified Down's and Black criteria used to evaluate the quality of studies. RESULTS: A total of 5,114 studies were reviewed for neonatal sepsis out of which 24 consisting of a total of 2,280 cases were selected for final review. Nine studies met criteria for assessment of hospital based incidence of neonatal sepsis representing 31,305 hospital births. The incidence of neonatal sepsis was 18.2/1000 livebirths with range from 7-55/1000 livebirths while the GBS incidence was 0.06/1000 livebirths with range from 0-2/1000 live births. We discovered various limitations such as identification techniques that could result in underestimation of the true incidence of GBS sepsis. Pathogens such as Klebsiella pneumoniae and Staphylococcus aureus were more commonly isolated than GBS. IMPLICATIONS OF KEY FINDINGS: The hospital based incidence of neonatal sepsis was high at 18.2/1000 live births while that due to GBS was 0.06/1000 live births. The burden of neonatal sepsis, including that attributable to GBS is substantial and could be reduced by preventive strategies such as intrapartum antibiotic prophylaxis or GBS vaccine. There is however very sparse meaningful data currently. Well planned prospective studies with larger sample sizes, more advanced isolation and identification techniques and those following up invasive disease cases for possible short and long term sequelae are needed-not only prior to possible introduction of the vaccine to determine the baseline epidemiology, but also thereafter to monitor its impact on the population. Strategies need to be developed to also reduce the morbidity and mortality attributable to other bacteria that have an incidence even greater than that of GBS.


Asunto(s)
Sepsis Neonatal/epidemiología , Sepsis Neonatal/etiología , Infecciones Estreptocócicas/complicaciones , Infecciones Estreptocócicas/epidemiología , Humanos , Recién Nacido , Sepsis Neonatal/prevención & control , Nigeria/epidemiología , Infecciones Estreptocócicas/prevención & control
19.
BMC Cancer ; 17(1): 611, 2017 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-28865422

RESUMEN

BACKGROUND: Throughout most of sub-Saharan Africa (and, indeed, most resource-limited areas), lack of death registries prohibits linkage of cancer diagnoses and precludes the most expeditious approach to determining cancer survival. Instead, estimation of cancer survival often uses clinical records, which have some mortality data but are replete with patients who are lost to follow-up (LTFU), some of which may be caused by undocumented death. The end result is that accurate estimation of cancer survival is rarely performed. A prominent example of a common cancer in Africa for which survival data are needed but for which frequent LTFU has precluded accurate estimation is Kaposi sarcoma (KS). METHODS: Using electronic records, we identified all newly diagnosed KS among HIV-infected adults at 33 primary care clinics in Kenya, Uganda, Nigeria, and Malawi from 2009 to 2012. We determined those patients who were apparently LTFU, defined as absent from clinic for ≥90 days at database closure and unknown to be dead or transferred. Using standardized protocols which included manual chart review, telephone calls, and physical tracking in the community, we attempted to update vital status amongst patients who were LTFU. RESULTS: We identified 1222 patients with KS, of whom 440 were LTFU according to electronic records. Manual chart review revealed that 18 (4.1%) were classified as LFTU due to clerical error, leaving 422 as truly LTFU. Of these 422, we updated vital status in 78%; manual chart review was responsible for updating in 5.7%, telephone calls in 26%, and physical tracking in 46%. Among 378 patients who consented at clinic enrollment to be tracked if they became LTFU and who had sufficient geographic contact/locator information, we updated vital status in 88%. Duration of LTFU was not associated with success of tracking, but tracking success was better in Kenya than the other sites. CONCLUSION: It is feasible to update vital status in a large fraction of patients with HIV-associated KS in sub-Saharan Africa who have become LTFU from clinical care. This finding likely applies to other cancers as well. Updating vital status amongst lost patients paves the way towards accurate determination of cancer survival.


Asunto(s)
Infecciones Oportunistas Relacionadas con el SIDA/epidemiología , Epidemias , Infecciones por VIH/epidemiología , Sarcoma de Kaposi/epidemiología , Infecciones Oportunistas Relacionadas con el SIDA/mortalidad , Infecciones Oportunistas Relacionadas con el SIDA/fisiopatología , Infecciones Oportunistas Relacionadas con el SIDA/virología , Adulto , África del Sur del Sahara/epidemiología , Fármacos Anti-VIH/uso terapéutico , Femenino , Infecciones por VIH/complicaciones , Infecciones por VIH/fisiopatología , Infecciones por VIH/virología , Humanos , Perdida de Seguimiento , Masculino , Sarcoma de Kaposi/mortalidad , Sarcoma de Kaposi/fisiopatología , Sarcoma de Kaposi/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...