Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Curr Opin Infect Dis ; 37(4): 232-237, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38748563

ABSTRACT

PURPOSE OF REVIEW: Congenital CMV (cCMV) infection is the most common infection of newborns and a leading cause of hearing loss and other neurologic disabilities in children. This review focuses on the diagnosis, presentation and management of cCMV infection. RECENT FINDINGS: Cytomegalovirus is one of the leading causes of sensorineural hearing loss in children. It also leads to neurodevelopmental disabilities and learning problems throughout childhood in both symptomatic and asymptomatic newborns. Urine and saliva PCR testing are the preferred methods of testing newborn infants for cCMV. In recent years, newborn-targeted and universal screening programs have been implemented in several states and major medical centers with the goal of identifying infected infants at risk for hearing loss. Treatment for infants diagnosed with cCMV infection should be limited to those who are moderately to severely symptomatic at birth with cCMV infection, though treatment may be beneficial for children who are asymptomatic with isolated sensorineural hearing loss. SUMMARY: As more children with cCMV are being identified through newborn screening, understanding the clinical presentation and sequelae is important for appropriate management of children with cCMV.


Subject(s)
Cytomegalovirus Infections , Hearing Loss, Sensorineural , Neonatal Screening , Humans , Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/congenital , Cytomegalovirus Infections/therapy , Cytomegalovirus Infections/drug therapy , Infant, Newborn , Neonatal Screening/methods , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/virology , Antiviral Agents/therapeutic use , Cytomegalovirus/isolation & purification , Disease Management
2.
Vaccine ; 41 Suppl 2: S53-S75, 2023 11 03.
Article in English | MEDLINE | ID: mdl-37806805

ABSTRACT

Cytomegalovirus (CMV) is the most common infectious cause of congenital malformation and a leading cause of developmental disabilities such as sensorineural hearing loss (SNHL), motor and cognitive deficits. The significant disease burden from congenital CMV infection (cCMV) led the US National Institute of Medicine to rank CMV vaccine development as the highest priority. An average of 6.7/1000 live births are affected by cCMV, but the prevalence varies across and within countries. In contrast to other congenital infections such as rubella and toxoplasmosis, the prevalence of cCMV increases with CMV seroprevalence rates in the population. The true global burden of cCMV disease is likely underestimated because most infected infants (85-90 %) have asymptomatic infection and are not identified. However, about 7-11 % of those with asymptomatic infection will develop SNHL throughout early childhood. Although no licensed CMV vaccine exists, several candidate vaccines are in development, including one currently in phase 3 trials. Licensure of one or more vaccine candidates is feasible within the next five years. Various models of CMV vaccine strategies employing different target populations have shown to provide substantial benefit in reducing cCMV. Although CMV can cause end-organ disease with significant morbidity and mortality in immunocompromised individuals, the focus of this vaccine value profile (VVP) is on preventing or reducing the cCMV disease burden. This CMV VVP provides a high-level, comprehensive assessment of the currently available data to inform the potential public health, economic, and societal value of CMV vaccines. The CMV VVP was developed by a working group of subject matter experts from academia, public health groups, policy organizations, and non-profit organizations. All contributors have extensive expertise on various elements of the CMV VVP and have described the state of knowledge and identified the current gaps. The VVP was developed using only existing and publicly available information.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus Vaccines , Hearing Loss, Sensorineural , Infant , Humans , Child, Preschool , Cytomegalovirus , Asymptomatic Infections , Seroepidemiologic Studies , Cytomegalovirus Infections/epidemiology , Cytomegalovirus Infections/prevention & control , Hearing Loss, Sensorineural/congenital , Hearing Loss, Sensorineural/epidemiology
3.
J Pediatric Infect Dis Soc ; 12(4): 230-233, 2023 Apr 28.
Article in English | MEDLINE | ID: mdl-36879401

ABSTRACT

Viral load in infant saliva and urine was assessed to predict sensorineural hearing loss (SNHL) in children with congenital cytomegalovirus infection. Viral load was higher in symptomatic infants. Viral load in asymptomatic children with and without SNHL did not differ. Congenital cytomegalovirus infection viral load in urine and saliva does not predict hearing loss.


Subject(s)
Cytomegalovirus Infections , Hearing Loss, Sensorineural , Infant , Humans , Child , Saliva , Cytomegalovirus , Viral Load
4.
J Am Chem Soc ; 145(9): 5553-5560, 2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36848549

ABSTRACT

Carbodiimide-fueled anhydride bond formation has been used to enhance the mechanical properties of permanently crosslinked polymer networks, giving materials that exhibit transitions from soft gels to covalently reinforced gels, eventually returning to the original soft gels. Temporary changes in mechanical properties result from a transient network of anhydride crosslinks, which eventually dissipate by hydrolysis. Over an order of magnitude increase in the storage modulus is possible through carbodiimide fueling. The time-dependent mechanical properties can be modulated by the concentration of carbodiimide, temperature, and primary chain architecture. Because the materials remain rheological solids, new material functions such as temporally controlled adhesion and rewritable spatial patterns of mechanical properties have been realized.

5.
Open Forum Infect Dis ; 10(1): ofad009, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36686629

ABSTRACT

Cardiac outcomes of 131 children with multisystem inflammatory syndrome (MIS-C) were examined. The majority of the cohort was male (66.4%) and half were Black (49.6%). Cardiac involvement was evident in 25% of the cohort at diagnosis. Favorable short- and mid-term outcomes were documented on follow-up, irrespective of the severe acute respiratory syndrome coronavirus 2 variants causing the infection.

6.
Antiviral Res ; 203: 105326, 2022 07.
Article in English | MEDLINE | ID: mdl-35504491

ABSTRACT

Congenital cytomegalovirus (cCMV) infection is the leading non-genetic cause of long-term neurological and sensory sequelae, the most common being sensorineural hearing loss (SNHL). Standard therapy for infants with symptomatic cCMV is valganciclovir for six months. However, little is known about the effects of antiviral therapy on CMV diversity while patients are on treatment. In this study, CMV variation was analyzed from urine specimens isolated from two patients with cCMV shortly after birth and at seven months. One was treated with valganciclovir for six weeks and the other for six months. In order to track these variants a novel bioinformatic approach was employed to analyze changes in low frequency variants over time. In the infant receiving antivirals for only six weeks, there was a fourfold increase in variation in UL97 from the seven month specimen. Furthermore, an eightfold increase in variation was seen in UL83 (pp65) with seven potential escape mutations occurring, and a twofold increase in UL73 (gN). In contrast variation did not increase or was reduced in these coding regions in the infant receiving valganciclovir for six months. However, there were increases in other CMV regions in samples isolated from both patients indicating further longitudinal studies are warranted to better understand the interplay between CMV diversity, antiviral therapy and patient outcome.


Subject(s)
Cytomegalovirus Infections , Hearing Loss, Sensorineural , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cytomegalovirus , Hearing Loss, Sensorineural/congenital , Humans , Infant , Infant, Newborn , Valganciclovir/pharmacology , Valganciclovir/therapeutic use
7.
J Pediatr Pharmacol Ther ; 26(7): 659-668, 2021.
Article in English | MEDLINE | ID: mdl-34588929

ABSTRACT

The goals of antimicrobial stewardship programs (ASPs) are to optimize antimicrobial prescribing habits in order to improve patient outcomes, reduce antimicrobial resistance, and reduce hospital costs. Multiple society-endorsed guidelines and government policies reinforce the importance of ASP implementation. Effective antimicrobial stewardship can impact unique patients, hospitals, and societal antibiotic-resistance burden. The role and subsequent success of these programs has largely been reported in the adult population. Pediatric and neonatal intensive care units present unique challenges for traditional antimicrobial stewardship approaches. The purpose of this review article is to explore the challenges of appropriate antibiotic use in the pediatric and neonatal intensive care units and to summarize strategies ASPs can use to overcome these challenges. These problems include non-specific disease presentations, limited evidence for definitive treatment durations in many pediatric infections, fewer pediatric-trained infectious disease physicians, and applicability of intensive laboratory obtainment, collection, and interpretation. Additionally, many ASP implementation studies evaluating the efficacy of ASPs exclude the PICU and NICU. Areas of focus for pediatric ASPs should likely include appropriate antibiotic initiation, appropriate antibiotic duration, and appropriate antibiotic de-escalation.

8.
Antiviral Res ; 191: 105083, 2021 07.
Article in English | MEDLINE | ID: mdl-33964331

ABSTRACT

Congenital cytomegalovirus (cCMV) infection is a leading cause of hearing loss and neurological disabilities in children worldwide. Although a minority of infants with cCMV will have symptoms at a birth, these children are at high risk of long-term sequelae. Most infants with cCMV have no clinical signs at birth (asymptomatic), but 10-15% will develop hearing loss. The diagnosis of cCMV relies on detection of the virus from urine or saliva within the first three weeks of life, with saliva PCR being the preferred method due to ease of collection and high sensitivity of the assay. Measures to prevent mother-to-child transmission of CMV are limited, and antiviral therapy with valganciclovir for 6 months is the standard of care for infants with symptomatic cCMV. As more infants with cCMV are being identified through newborn screening, studies are urgently needed to address antiviral treatment in asymptomatic infants and the implementation of prevention strategies to prevent fetal infection. This article is part of the symposium "New drugs and vaccines for DNA virus infections: a symposium in memory of Mark Prichard."


Subject(s)
Antiviral Agents/therapeutic use , Cytomegalovirus Infections/drug therapy , Cytomegalovirus/drug effects , Antiviral Agents/classification , Cytomegalovirus/genetics , Cytomegalovirus Infections/congenital , Cytomegalovirus Infections/transmission , Female , Humans , Infant , Infant, Newborn , Infectious Disease Transmission, Vertical/prevention & control , Neonatal Screening , Pregnancy , Pregnancy Complications, Infectious/drug therapy , Pregnancy Complications, Infectious/virology , Saliva/virology
9.
J Infect Dis ; 221(Suppl 1): S9-S14, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32134480

ABSTRACT

Congenital cytomegalovirus (cCMV) infection is a leading cause of hearing loss and neurological disabilities in children, with the disease burden and disabilities due to cCMV greater than many other well recognized childhood conditions. A minority of infants with cCMV will have symptoms at birth. Infants with symptomatic cCMV are at higher risk for sequelae than those born without symptoms. The majority of infants with cCMV are asymptomatic at birth, but 10%-15% will develop hearing loss. Although clinical symptoms can help predict which infants will have sensorineural hearing loss, among asymptomatic cCMV there are currently no predictors of adverse outcome. The identification of a biomarker to identify those at highest risk of sequelae is highly desirable to target interventions to those who could potentially benefit. Because there is increasing rationale for establishing both targeted and universal screening programs for cCMV in the United States and worldwide, this is an urgent priority.


Subject(s)
Cytomegalovirus Infections/epidemiology , Cytomegalovirus Infections/virology , Cytomegalovirus , Infectious Disease Transmission, Vertical , Pregnancy Complications, Infectious/epidemiology , Pregnancy Complications, Infectious/virology , Child , Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/transmission , Disabled Persons , Female , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/epidemiology , Hearing Loss, Sensorineural/etiology , Humans , Infant, Newborn , Infectious Disease Transmission, Vertical/statistics & numerical data , Neonatal Screening , Pregnancy , Pregnancy Complications, Infectious/diagnosis , Prognosis , Symptom Assessment
10.
BMC Infect Dis ; 20(1): 111, 2020 Feb 10.
Article in English | MEDLINE | ID: mdl-32039707

ABSTRACT

After publication of the original article [1], we were notified that Fig. 3 has "Fig. 1" posted on the top of it and Figs. 4 and 5 have "Genomic Position" in a different spot.

12.
J Infect Dis ; 221(5): 715-720, 2020 02 18.
Article in English | MEDLINE | ID: mdl-31593588

ABSTRACT

BACKGROUND: Infection with multiple cytomegalovirus (CMV) strains (mixed infection) was reported in a variety of hosts. As the virus genetic diversity in primary CMV infection and the changes over time remain incompletely defined, we examined CMV diversity and changes in diversity over time in healthy adolescent females who participated in a phase 2 CMV gB/MF59 vaccine trial. METHODS: CMV genetic diversity was determined by genotyping of 5 genes-gB (UL55), gH (UL75), gN (UL73), US28, and UL144-in urine, saliva, and plasma samples from 15 study subjects. RESULTS: At the time of primary infection, 5 of 12 (42%) urine samples had multiple virus strains, and 50% of vaccine recipients were infected with gB1 genotype (vaccine strain). Mixed infection was documented in all 15 subjects within 3 months after primary infection, and the majority had different CMV genotypes in different compartments. Changes in genotypes over time were observed in all subjects. CONCLUSIONS: Infection with multiple CMV genotypes was common during primary infection and further diversification occurred over time. Infection with gB1 genotype in vaccine recipients suggests a lack of strain-specific protection from the vaccine. As only 5 polymorphic genes were assessed, this study likely underestimated the true genetic diversity in primary CMV infection.


Subject(s)
Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/therapeutic use , Cytomegalovirus/genetics , Polymorphism, Genetic , Vaccination , Adolescent , Coinfection/diagnosis , Coinfection/virology , Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/virology , Double-Blind Method , Female , Genotype , Humans , Membrane Glycoproteins/blood , Membrane Glycoproteins/genetics , Membrane Glycoproteins/urine , Real-Time Polymerase Chain Reaction , Receptors, Chemokine/blood , Receptors, Chemokine/genetics , Saliva/virology , Viral Envelope Proteins/blood , Viral Envelope Proteins/genetics , Viral Envelope Proteins/urine , Viral Load , Viral Proteins/blood , Viral Proteins/genetics , Viral Proteins/urine
13.
BMC Infect Dis ; 19(1): 1046, 2019 Dec 10.
Article in English | MEDLINE | ID: mdl-31822287

ABSTRACT

BACKGROUND: Congenital cytomegalovirus (cCMV) infection is the most common congenital infection and a leading cause of long-term neurological and sensory sequelae, the most common being sensorineural hearing loss (SNHL). Despite extensive research, clinical or laboratory markers to identify CMV infected children with increased risk for disease have not been identified. This study utilizes viral whole-genome next generation-sequencing (NGS) of specimens from congenitally infected infants to explore viral diversity and specific viral variants that may be associated with symptomatic infection and SNHL. METHODS: CMV DNA from urine specimens of 30 infants (17 asymptomatic, 13 symptomatic) was target enriched and next generation sequenced resulting in 93% coverage of the CMV genome allowing analysis of viral diversity. RESULTS: Variant frequency distribution was compared between children with symptomatic and asymptomatic cCMV and those with (n = 13) and without (n = 17) hearing loss. The CMV genes UL48A, UL88, US19 and US22 were found to have an increase in nucleotide diversity in symptomatic children; while UL57, UL20, UL104, US14, UL115, and UL35 had an increase in diversity in children with hearing loss. An analysis of single variant differences between symptomatic and asymptomatic children found UL55 to have the highest number, while the most variants associated with SNHL were in the RL11 gene family. In asymptomatic infants with SNHL, mutations were observed more frequently in UL33 and UL20. CONCLUSION: CMV genomes from infected newborns can be mapped to 93% of the genome at a depth allowing accurate and reproducible analysis of polymorphisms for variant and gene discovery that may be linked to symptomatic and hearing loss outcomes.


Subject(s)
Cytomegalovirus Infections/diagnosis , Cytomegalovirus/genetics , DNA, Viral/genetics , Hearing Loss, Sensorineural/diagnosis , Child , Cytomegalovirus/classification , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/virology , DNA, Viral/metabolism , DNA, Viral/urine , Female , Hearing Loss, Sensorineural/complications , High-Throughput Nucleotide Sequencing , Humans , Infant , Infant, Newborn , Male , Mutation , Phylogeny , Principal Component Analysis
14.
J Pediatr ; 200: 196-201.e1, 2018 09.
Article in English | MEDLINE | ID: mdl-29784513

ABSTRACT

OBJECTIVE: To evaluate the impact of race and ethnicity upon the prevalence and clinical spectrum of congenital cytomegalovirus infection (cCMV). STUDY DESIGN: From 2007 to 2012, 100 332 infants from 7 medical centers were screened for cCMV while in the hospital. Ethnicity and race were collected and cCMV prevalence rates were calculated. RESULTS: The overall prevalence of cCMV in the cohort was 4.5 per 1000 live births (95% CI, 4.1-4.9). Black infants had the highest cCMV prevalence (9.5 per 1000 live births; 95% CI, 8.3-11.0), followed by multiracial infants (7.8 per 1000 live births; 95% CI, 4.7-12.0). Significantly lower prevalence rates were observed in non-Hispanic white infants (2.7 per 1000 live births; 95% CI, 2.2-3.3), Hispanic white infants (3.0 per 1000 live births; 95% CI, 2.4-3.6), and Asian infants (1.0 per 1000 live births; 95% CI, 0.3-2.5). After adjusting for socioeconomic status and maternal age, black infants were significantly more likely to have cCMV compared with non-Hispanic white infants (adjusted prevalence OR, 1.9; 95% CI, 1.4-2.5). Hispanic white infants had a slightly lower risk of having cCMV compared with non-Hispanic white infants (adjusted prevalence OR, 0.7; 95% CI, 0.5-1.0). However, no significant differences in symptomatic cCMV (9.6%) and sensorineural hearing loss (7.8%) were observed between the race/ethnic groups. CONCLUSIONS: Significant racial and ethnic differences exist in the prevalence of cCMV, even after adjusting for socioeconomic status and maternal age. Although once infected, the newborn disease and rates of hearing loss in infants are similar with respect to race and ethnicity.


Subject(s)
Cytomegalovirus Infections/ethnology , Ethnicity , Mass Screening/methods , Racial Groups , Adult , Cytomegalovirus Infections/congenital , Female , Follow-Up Studies , Humans , Infant, Newborn , Male , Prevalence , Retrospective Studies , United States/epidemiology
17.
J Infect Dis ; 217(10): 1612-1615, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29401303

ABSTRACT

Real-time polymerase chain reaction (PCR) of saliva is highly sensitive for newborn congenital cytomegalovirus (CMV) screening. This study uses nationally published CMV seroprevalence and breastfeeding rates to estimate the contribution of CMV DNA in breast milk to false-positive saliva PCR results. The false-positive rates adjusted for breastfeeding ranged from 0.03% in white Hispanic persons to 0.14% in white non-Hispanic persons. Saliva CMV PCR for newborn screening is highly sensitive, and the low false-positive rates in this study suggest that saliva PCR results are unlikely to be significantly influenced by breastfeeding or other perinatal exposures.


Subject(s)
Breast Feeding/adverse effects , Cytomegalovirus Infections/diagnosis , Cytomegalovirus/genetics , Saliva/virology , DNA, Viral/genetics , Female , Humans , Infant, Newborn , Male , Neonatal Screening/methods , Real-Time Polymerase Chain Reaction/methods
18.
J Infect Dis ; 216(suppl_10): S912-S918, 2017 12 16.
Article in English | MEDLINE | ID: mdl-29267912

ABSTRACT

A widespread epidemic of Zika virus (ZIKV) infection was reported in 2015 from South and Central America and the Caribbean. Although the full spectrum of ZIKV infection of the newborn has yet to be determined, other maternal viral infections resulting in transmission to the fetus provide instructive lessons that can be applied to the prospective evaluation of individuals with ZIKV infection. This review focuses on those other congenital infections, including rubella, congenital cytomegalovirus, human immunodeficiency virus, hepatitis B virus, and neonatal herpes simplex virus, from which lessons for the evaluation of ZIKV in the newborn can be applied.


Subject(s)
Infant, Newborn, Diseases/virology , Virus Diseases/virology , Zika Virus Infection/virology , Zika Virus/pathogenicity , Americas , Caribbean Region , Female , Hepatitis B/transmission , Hepatitis B/virology , Herpes Simplex/transmission , Herpes Simplex/virology , Humans , Infant, Newborn , Pregnancy Complications, Infectious/virology , Rubella/transmission , Rubella/virology , Virus Diseases/transmission , Zika Virus Infection/transmission
19.
Life Sci Space Res (Amst) ; 15: 1-10, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29198308

ABSTRACT

Human immune response is compromised and bacteria can become more antibiotic resistant in space microgravity (MG). We report that under low-shear modeled microgravity (LSMMG), stationary-phase uropathogenic Escherichia coli (UPEC) become more resistant to gentamicin (Gm), and that this increase is dependent on the presence of σs (a transcription regulator encoded by the rpoS gene). UPEC causes urinary tract infections (UTIs), reported to afflict astronauts; Gm is a standard treatment, so these findings could impact astronaut health. Because LSMMG findings can differ from MG, we report preparations to examine UPEC's Gm sensitivity during spaceflight using the E. coli Anti-Microbial Satellite (EcAMSat) as a free-flying "nanosatellite" in low Earth orbit. Within EcAMSat's payload, a 48-microwell fluidic card contains and supports study of bacterial cultures at constant temperature; optical absorbance changes in cell suspensions are made at three wavelengths for each microwell and a fluid-delivery system provides growth medium and predefined Gm concentrations. Performance characterization is reported here for spaceflight prototypes of this payload system. Using conventional microtiter plates, we show that Alamar Blue (AB) absorbance changes can assess the Gm effect on E. coli viability, permitting telemetric transfer of the spaceflight data to Earth. Laboratory results using payload prototypes are consistent with wellplate and flask findings of differential sensitivity of UPEC and its ∆rpoS strain to Gm. if σs plays the same role in space MG as in LSMMG and Earth gravity, countermeasures discovered in recent Earth studies (aimed at weakening the UPEC antioxidant defense) to control UPEC infections would prove useful also in space flights. Further, EcAMSat results should clarify inconsistencies from previous space experiments on bacterial antibiotic sensitivity and other issues.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Drug Resistance, Bacterial , Gentamicins/pharmacology , Sigma Factor/genetics , Uropathogenic Escherichia coli/growth & development , Weightlessness , Cell Survival/drug effects , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Humans , Microbial Viability , Mutation , Urinary Tract Infections/drug therapy , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/drug effects , Uropathogenic Escherichia coli/genetics
20.
J Infect Dis ; 216(9): 1176-1179, 2017 11 27.
Article in English | MEDLINE | ID: mdl-28968907

ABSTRACT

The role of human cytomegalovirus (HCMV)-specific T-cell responses in breast milk of HCMV-seropositive mothers is not well defined. In these studies, we demonstrate that the frequency of cytomegalovirus (CMV)-pp65-specific T-cell responses in peripheral blood mononuclear cells (PBMCs) and breast milk cells (BMCs) is increased for CD8+ T cells in both sample sources when compared with CD4+ T cells. The frequency of pp55-specific CD8 T cells producing interferon γ (IFN-γ) alone or dual IFN-γ/granzyme rB producers is increased in breast milk compared with PBMCs. Lastly, we observed a positive correlation between breast milk viral load and the CD8 pp65-specific response, suggesting that local virus replication drives antigen-specific CD8 T cells into the breast.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Cytomegalovirus/pathogenicity , Milk, Human/immunology , Milk, Human/virology , Adult , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Cytomegalovirus Infections/physiopathology , Female , Humans , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL