Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 207(10): 2534-2544, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34625521

RESUMEN

Human CMV (HCMV) is a ubiquitous pathogen that indelibly shapes the NK cell repertoire. Using transcriptomic, epigenomic, and proteomic approaches to evaluate peripheral blood NK cells from healthy human volunteers, we find that prior HCMV infection promotes NK cells with a T cell-like gene profile, including the canonical markers CD3ε, CD5, and CD8ß, as well as the T cell lineage-commitment transcription factor Bcl11b. Although Bcl11b expression is upregulated during NK maturation from CD56bright to CD56dim, we find a Bcl11b-mediated signature at the protein level for FcεRIγ, PLZF, IL-2Rß, CD3γ, CD3δ, and CD3ε in later-stage, HCMV-induced NK cells. BCL11B is targeted by Notch signaling in T cell development, and culture of NK cells with Notch ligand increases cytoplasmic CD3ε expression. The Bcl11b-mediated gain of CD3ε, physically associated with CD16 signaling molecules Lck and CD247 in NK cells is correlated with increased Ab-dependent effector function, including against HCMV-infected cells, identifying a potential mechanism for their prevalence in HCMV-infected individuals and their prospective clinical use in Ab-based therapies.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Infecciones por Citomegalovirus/inmunología , Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/inmunología , Proteínas Represoras/inmunología , Proteínas Supresoras de Tumor/inmunología , Animales , Complejo CD3/inmunología , Humanos , Ratones , Ratones Transgénicos , Transcriptoma
2.
Circulation ; 143(18): 1754-1762, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33820423

RESUMEN

BACKGROUND: Left atrial appendage (LAA) occlusion provides an alternative to oral anticoagulation for thromboembolic risk reduction in patients with nonvalvular atrial fibrillation. Since regulatory approval in 2015, the WATCHMAN device has been the only LAA closure device available for clinical use in the United States. The PINNACLE FLX study (Protection Against Embolism for Nonvalvular AF Patients: Investigational Device Evaluation of the Watchman FLX LAA Closure Technology) evaluated the safety and effectiveness of the next-generation WATCHMAN FLX LAA closure device in patients with nonvalvular atrial fibrillation in whom oral anticoagulation is indicated, but who have an appropriate rationale to seek a nonpharmaceutical alternative. METHODS: This was a prospective, nonrandomized, multicenter US Food and Drug Administration study. The primary safety end point was the occurrence of one of the following events within 7 days after the procedure or by hospital discharge, whichever was later: death, ischemic stroke, systemic embolism, or device- or procedure-related events requiring cardiac surgery. The primary effectiveness end point was the incidence of effective LAA closure (peri-device flow ≤5 mm), as assessed by the echocardiography core laboratory at 12-month follow-up. RESULTS: A total of 400 patients were enrolled. The mean age was 73.8±8.6 years and the mean CHA2DS2-VASc score was 4.2±1.5. The incidence of the primary safety end point was 0.5% with a 1-sided 95% upper CI of 1.6%, meeting the performance goal of 4.2% (P<0.0001). The incidence of the primary effectiveness end point was 100%, with a 1-sided 95% lower CI of 99.1%, again meeting the performance goal of 97.0% (P<0.0001). Device-related thrombus was reported in 7 patients, no patients experienced pericardial effusion requiring open cardiac surgery, and there were no device embolizations. CONCLUSIONS: LAA closure with this next-generation LAA closure device was associated with a low incidence of adverse events and a high incidence of anatomic closure. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT02702271.


Asunto(s)
Apéndice Atrial/fisiopatología , Anciano , Humanos , Estudios Prospectivos , Resultado del Tratamiento
3.
J Infect Dis ; 223(11): 2001-2012, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-33031517

RESUMEN

BACKGROUND: Cytomegalovirus (CMV) can cause congenital infection and is the leading cause of nongenetic newborn disabilities. V160, a conditionally replication-defective virus, is an investigational vaccine under evaluation for prevention of congenital CMV. The vaccine was well tolerated and induced both humoral and cellular immunity in CMV-seronegative trial participants. T-cell-mediated immunity is important for immune control of CMV. Here we describe efforts to understand the quality attributes of the T-cell responses induced by vaccination. METHODS: Using multicolor flow cytometry, we analyzed vaccine-induced T cells for memory phenotype, antigen specificity, cytokine profiles, and cytolytic potential. Moreover, antigen-specific T cells were sorted from 4 participants, and next-generation sequencing was used to trace clonal lineage development during the course of vaccination using T-cell receptor ß-chain sequences as identifiers. RESULTS: The results demonstrated that vaccination elicited polyfunctional CD4 and CD8 T cells to 2 dominant antigens, pp65 and IE1, with a predominantly effector phenotype. Analysis of T-cell receptor repertoires showed polyclonal expansion of pp65- and IE1-specific T cells after vaccination. CONCLUSION: V160 induced a genetically diverse and polyfunctional T-cell response and the data support further clinical development of V160 for prevention of CMV infection and congenital transmission. CLINICAL TRIALS REGISTRATION: NCT01986010.


Asunto(s)
Linfocitos T CD8-positivos , Infecciones por Citomegalovirus , Vacunas contra Citomegalovirus , Inmunidad Celular , Linfocitos T CD8-positivos/inmunología , Infecciones por Citomegalovirus/prevención & control , Vacunas contra Citomegalovirus/inmunología , Humanos , Vacunación
4.
Clin Infect Dis ; 71(6): 1491-1498, 2020 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-31628849

RESUMEN

BACKGROUND: After primary maternal cytomegalovirus (CMV) infection during pregnancy, infants are at risk for disease. METHODS: Factors predictive of infant outcome were analyzed in a database of 304 pregnant women with primary infection. These women were enrolled between 2010 and 2017 and delivered 281 infants, of whom 108 were CMV infected. Long term follow-up occurred for 173 uninfected and 106 infected infants at age 4 years (range, 1-8 years). One hundred fifty-seven women were treated with an average of 2 doses (range, 1-6 doses) of high-dose hyperimmune globulin (HIG: 200 mg/kg/infusion). We used a regression model to define predictors of fetal infection, symptoms at birth, and long-term sequelae; 31 covariates were tested. RESULTS: Four factors predicted fetal infection: a 1.8-fold increase (30% vs 56%) in the rate of congenital infection without HIG (adjusted odds ratio [AOR], 5.2; P < .0001), a 1.8-fold increase (32% vs 56%) associated with maternal viral DNAemia prior to HIG administration (AOR, 3.0; P = .002), abnormal ultrasounds (AOR, 59; P = .0002), and diagnosis of maternal infection by seroconversion rather than avidity (AOR, 3.3; P = .007). Lack of HIG and abnormal ultrasounds also predicted symptoms (P = .001). Long-term sequelae were predicted by not receiving HIG (AOR, 13.2; P = .001), maternal infection in early gestation (odds ratio [OR], 0.9; P = .017), and abnormal ultrasounds (OR, 7.6; P < .003). Prevalence and copy/number of DNAemia declined after HIG. CONCLUSIONS: Maternal viremia predicts fetal infection and neonatal outcome. This may help patient counseling. High-dose HIG may prevent fetal infection and disease and is associated with the resolution of DNAemia.


Asunto(s)
Infecciones por Citomegalovirus , Globulinas , Complicaciones Infecciosas del Embarazo , Niño , Preescolar , Citomegalovirus , Femenino , Humanos , Lactante , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Embarazo , Mujeres Embarazadas
5.
J Virol ; 93(23)2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31511385

RESUMEN

Human cytomegalovirus (HCMV) can cause congenital infections, which are a leading cause of childhood disabilities. Since the rate of maternal-fetal transmission is much lower in naturally infected (HCMV-seropositive) women, we hypothesize that a vaccine candidate capable of eliciting immune responses analogous to those of HCMV-seropositive subjects may confer protection against congenital HCMV. We have previously described a replication-defective virus vaccine based on strain AD169 (D. Wang, D. C. Freed, X. He, F. Li, et al., Sci Transl Med 8:362ra145, 2016, https://doi.org/10.1126/scitranslmed.aaf9387). The vaccine, named V160, has been shown to be safe and immunogenic in HCMV-seronegative human subjects, eliciting both humoral and cellular immune responses (S. P. Adler, S. E. Starr, S. A. Plotkin, S. H. Hempfling, et al., J Infect Dis 220:411-419, 2019, https://doi.org/10.1093/infdis/171.1.26). Here, we further showed that sera from V160-immunized HCMV-seronegative subjects have attributes similar in quality to those from seropositive subjects, including high-avidity antibodies to viral antigens, coverage against a panel of genetically distinct clinical isolates, and protection against viral infection in diverse types of human cells in culture. More importantly, vaccination appeared efficient in priming the human immune system, inducing memory B cells in six V160 recipients at frequencies comparable to those of three HCMV-seropositive subjects. Our results demonstrate the ability of V160 to induce robust and durable humoral memory responses to HCMV, justifying further clinical evaluation of the vaccine against congenital HCMV.IMPORTANCEIn utero HCMV infection can lead to miscarriage or childhood disabilities, and an effective vaccine is urgently needed. Since children born to women who are seropositive prior to pregnancy are less likely to be affected by congenital HCMV infection, it has been hypothesized that a vaccine capable of inducing an immune response resembling the responses in HCMV-seropositive women may be effective. We previously described a replication-defective virus vaccine that has been demonstrated safe and immunogenic in HCMV-seronegative subjects. Here, we conducted additional analyses to show that the vaccine can induce antibodies with functional attributes similar to those from HCMV-seropositive subjects. Importantly, vaccination can induce long-lived memory B cells at frequencies comparable to those seen in HCMV-seropositive subjects. We conclude that this vaccine is a promising candidate that warrants further clinical evaluation for prevention of congenital HCMV.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/prevención & control , Vacunas contra Citomegalovirus/inmunología , Citomegalovirus/inmunología , Inmunidad Humoral/inmunología , Inmunización , Adulto , Anciano , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Antígenos Virales/sangre , Línea Celular , Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/virología , Método Doble Ciego , Femenino , Humanos , Inmunidad Celular , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Masculino , Persona de Mediana Edad , Estados Unidos , Vacunación , Replicación Viral , Adulto Joven
6.
J Infect Dis ; 220(3): 411-419, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31535143

RESUMEN

BACKGROUND: A conditionally replication-defective human cytomegalovirus (CMV) vaccine (V160) derived from AD169 and genetically engineered to express CMV pentameric complex (gH/gL/pUL128/pUL130/pUL131) was developed and evaluated for phase 1 vaccine safety and immunogenicity in CMV-seronegative and CMV-seropositive adults. METHODS: Subjects received 3 doses of V160 or placebo on day 1, month 1, and month 6. Four vaccine dose levels, formulated with or without aluminum phosphate adjuvant, were evaluated. Injection-site and systemic adverse events (AEs) and vaccine viral shedding were monitored. CMV-specific cellular and humoral responses were measured by interferon-gamma ELISPOT and virus neutralization assay up to 12 months after last dose. RESULTS: V160 was generally well-tolerated, with no serious AEs observed. Transient, mild-to-moderate injection-site and systemic AEs were reported more frequently in vaccinated subjects than placebo. Vaccine viral shedding was not detected in any subject, confirming the nonreplicating feature of V160. Robust neutralizing antibody titers were elicited and maintained through 12 months postvaccination. Cellular responses to structural and nonstructural viral proteins were observed, indicating de novo expression of viral genes postvaccination. CONCLUSIONS: V160 displayed an acceptable safety profile. Levels of neutralizing antibodies and T-cell responses in CMV-seronegative subjects were within ranges observed following natural CMV infection. CLINICAL TRIAL REGISTRATION: . NCT01986010.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Vacunas contra Citomegalovirus/inmunología , Citomegalovirus/inmunología , Replicación Viral/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adolescente , Adulto , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Método Doble Ciego , Ensayo de Immunospot Ligado a Enzimas/métodos , Femenino , Humanos , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Inmunización/métodos , Masculino , Persona de Mediana Edad , Linfocitos T/inmunología , Vacunación/métodos , Esparcimiento de Virus/inmunología , Adulto Joven
7.
Med Microbiol Immunol ; 208(3-4): 323-328, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31062089

RESUMEN

The establishment of a lifelong latent infection after resolution of primary infection is a hallmark of cytomegalovirus (CMV) biology. Primary infection with human CMV is possible any time in life, but most frequently, virus transmission occurs already perinatally or in early childhood. Many years or even decades later, severe clinical problems can result from recurrence of infectious virus by reactivation from latency in individuals who undergo immunocompromising medical treatment, for instance, transplant recipients, but also in septic patients without canonical immunosuppression, and in elderly people with a weakened immune system. The diversity of disease manifestations, such as retinitis, pneumonia, hepatitis, gastrointestinal disease, and others, has remained an enigma. In clinical routine, seropositivity for IgG antibodies against human CMV is taken to indicate latent infection and thus to define a qualitative risk of recurrence, but it is insufficient as a predictor for the quantitative risk of recurrence. Early experimental studies in the mouse model, comparing primary infection of neonatal and adult mice, led to the hypothesis that high load of latent viral genomes is a better predictor for the quantitative risk. A prolonged period of virus multiplication in the immunologically immature neonatally infected host increased the risk of virus recurrence by an enhanced copy number of latent virus genomes from which reactivation can initiate. In extension of this hypothesis, one would predict today that a higher incidence of reactivation events will also fuel the expansion of virus-specific T cells observed in the elderly, a phenomenon known as "memory inflation". Notably, the mouse model also indicated a stochastic nature of reactivation, thus offering an explanation for the diversity and organ selectivity of disease manifestations observed in patients. As the infection history is mostly undefined in humans, such predictions from the mouse model are difficult to verify by clinical investigation, and moreover, such questions were actually rarely addressed. Here, we have surveyed the existing literature for reports that may help to retrospectively relate the individual infection history to the risk of virus recurrence and recrudescent organ disease.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Interacciones Huésped-Patógeno , Memoria Inmunológica , Latencia del Virus , Replicación Viral , Anciano , Animales , Infecciones por Citomegalovirus/epidemiología , Infecciones por Citomegalovirus/virología , Modelos Animales de Enfermedad , Humanos , Ratones , Recurrencia , Medición de Riesgo
8.
Med Microbiol Immunol ; 208(1): 69-80, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30203132

RESUMEN

BACKGROUND: HCMV hyperimmunoglobulin-preparations (HIG) contain high concentrations of HCMV-specific IgG. The reduced maternofetal-HCMV-transmission rate of IgG may be due to HCMV-specific neutralizing antibodies against the HCMV pentameric complex (PC). In contrast to HIG, standard intravenous immunoglobulin (IVIG) may have more neutralization (NT) capacity than HIG due to higher IgG subclass 3 levels (Planitzer et al., 2011). METHODS: We investigated the HCMV-specific NT-capacity of HIG Cytotect®, using a recombinant pentameric complex (gHgLUL128-131A) for specific antibody-depletion. We used a modified UL130-peptide (TANQNPSPPWSKLTYSKPH) based on original-sequence of Saccoccio et al. (Vaccine 29(15):2705-2711, 2011) (SWSTLTANQNPSPPWSKLTY) as neutralization target. Both UL130-peptides and the PC were bound via sixfold HisTag and anti-HisTag mAbs to magnetic beads to deplete HCMV-specific IgGs from HIG (Cytotect®). Modifying this depletion strategy, we analyzed the role of IgG subclass 3 in both HIG and IVIG. RESULTS: After CMV IgG-normalization of HIG and IVIG, we found a significant trend towards a decrease (16%) of neutralization-capacity for the UL130 TAN-peptide, but not for the original UL130 SWS-peptide. However, highly significant loss of NT-capacity could be only observed by PC depletion (42%). The IgG subclass 3 depletion revealed no significant reduction of NT-capacity in both HIG and IVIG. CONCLUSION: Via specific antibody depletion, we could demonstrate that pentameric complex-specific antibodies are present in HIG and bind to the recombinant PC resulting in a highly significant reduction of NT-capacity compared to the UL130 TAN-and SWS-peptides. We could not confirm the functional role of IgG subclass 3 neutralizing antibodies in IgG-preparations.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Citomegalovirus/inmunología , Inmunoglobulinas Intravenosas/inmunología , Antígenos Virales/inmunología , Células Cultivadas , Femenino , Voluntarios Sanos , Humanos , Pruebas de Neutralización , Embarazo , Ensayo de Placa Viral
9.
Int J Mol Sci ; 20(13)2019 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-31261659

RESUMEN

Congenital human cytomegalovirus (HCMV) infection and HCMV infection of immunosuppressed patients cause significant morbidity and mortality, and vaccine development against HCMV is a major public health priority. HCMV envelope glycoproteins gB, gH, and gL, which constitute the core fusion machinery, play critical roles in HCMV fusion and entry into host cells. HCMV gB and gH/gL have been reported to elicit potent neutralizing antibodies. Recently, the gB/gH/gL complex was identified in the envelope of HCMV virions, and 16-50% of the total gH/gL bound to gB, forming the gB/gH/gL complex. These findings make the gB/gH/gL a unique HCMV vaccine candidate. We previously reported the production of HCMV trimeric gB and gH/gL heterodimers, and immunization with a combination of trimeric gB and gH/gL heterodimers elicited strong synergistic HCMV-neutralizing activity. To further improve the immunogenicity of gH/gL, we produced trimeric gH/gL. Rabbits immunized with HCMV trimeric gH/gL induced up to 38-fold higher serum titers of gH/gL-specific IgG relative to HCMV monomeric gH/gL, and elicited ~10-fold higher titers of complement-dependent and complement-independent HCMV-neutralizing activity for both epithelial cells and fibroblasts. HCMV trimeric gH/gL in combination with HCMV trimeric gB would be a novel promising HCMV vaccine candidate that could induce highly potent neutralizing activities.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Vacunas contra Citomegalovirus/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Células CHO , Cricetinae , Cricetulus , Humanos , Conejos , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética
10.
J Virol ; 91(7)2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28077654

RESUMEN

Human cytomegalovirus (HCMV) is the leading cause of congenital viral infection, and developing a prophylactic vaccine is of high priority to public health. We recently reported a replication-defective human cytomegalovirus with restored pentameric complex glycoprotein H (gH)/gL/pUL128-131 for prevention of congenital HCMV infection. While the quantity of vaccine-induced antibody responses can be measured in a viral neutralization assay, assessing the quality of such responses, including the ability of vaccine-induced antibodies to cross-neutralize the field strains of HCMV, remains a challenge. In this study, with a panel of neutralizing antibodies from three healthy human donors with natural HCMV infection or a vaccinated animal, we mapped eight sites on the dominant virus-neutralizing antigen-the pentameric complex of glycoprotein H (gH), gL, and pUL128, pUL130, and pUL131. By evaluating the site-specific antibodies in vaccine immune sera, we demonstrated that vaccination elicited functional antiviral antibodies to multiple neutralizing sites in rhesus macaques, with quality attributes comparable to those of CMV hyperimmune globulin. Furthermore, these immune sera showed antiviral activities against a panel of genetically distinct HCMV clinical isolates. These results highlighted the importance of understanding the quality of vaccine-induced antibody responses, which includes not only the neutralizing potency in key cell types but also the ability to protect against the genetically diverse field strains.IMPORTANCE HCMV is the leading cause of congenital viral infection, and development of a preventive vaccine is a high public health priority. To understand the strain coverage of vaccine-induced immune responses in comparison with natural immunity, we used a panel of broadly neutralizing antibodies to identify the immunogenic sites of a dominant viral antigen-the pentameric complex. We further demonstrated that following vaccination of a replication-defective virus with the restored pentameric complex, rhesus macaques can develop broadly neutralizing antibodies targeting multiple immunogenic sites of the pentameric complex. Such analyses of site-specific antibody responses are imperative to our assessment of the quality of vaccine-induced immunity in clinical studies.


Asunto(s)
Anticuerpos Neutralizantes/química , Anticuerpos Antivirales/química , Infecciones por Citomegalovirus/prevención & control , Citomegalovirus/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Especificidad de Anticuerpos , Línea Celular , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Mapeo Epitopo , Humanos , Macaca mulatta , Unión Proteica , Conejos , Vacunación , Vacunas Virales/administración & dosificación , Internalización del Virus
11.
Virus Genes ; 53(4): 650-655, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28391502

RESUMEN

Human cytomegalovirus (HCMV) is an important opportunistic pathogen in immunocompromised patients and a major cause of congenital birth defects when acquired in utero. In the 1990s, four chimeric viruses were constructed by replacing genome segments of the high passage Towne strain with segments of the low passage Toledo strain, with the goal of obtaining live attenuated vaccine candidates that remained safe but were more immunogenic than the overly attenuated Towne vaccine. The chimeras were found to be safe when administered to HCMV-seronegative human volunteers, but to differ significantly in their ability to induce seroconversion. This suggests that chimera-specific genetic differences impacted the ability to replicate or persist in vivo and the consequent ability to induce an antibody response. To identify specific genomic breakpoints between Towne and Toledo sequences and establish whether spontaneous mutations or rearrangements had occurred during construction of the chimeras, complete genome sequences were determined. No major deletions or rearrangements were observed, although a number of unanticipated mutations were identified. However, no clear association emerged between the genetic content of the chimeras and the reported levels of vaccine-induced HCMV-specific humoral or cellular immune responses, suggesting that multiple genetic determinants are likely to impact immunogenicity. In addition to revealing the genome organization of the four vaccine candidates, this study provided an opportunity to probe the genetics of HCMV attenuation in humans. The results may be valuable in the future design of safe live or replication-defective vaccines that optimize immunogenicity and efficacy.


Asunto(s)
Infecciones por Citomegalovirus/virología , Vacunas contra Citomegalovirus/inmunología , Citomegalovirus/genética , Citomegalovirus/inmunología , Recombinación Genética , Anticuerpos Antivirales/inmunología , Citomegalovirus/clasificación , Citomegalovirus/aislamiento & purificación , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/prevención & control , Vacunas contra Citomegalovirus/administración & dosificación , Vacunas contra Citomegalovirus/genética , Genoma Viral , Genómica , Humanos , Inmunización
12.
Pacing Clin Electrophysiol ; 40(11): 1291-1297, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28940232

RESUMEN

BACKGROUND: A cardiac lead with a side helix for active fixation to the coronary vein wall (Attain Stability® , Model 20066, Medtronic, Minneapolis, MN, USA) recently received CE Mark. The lead is designed to improve left ventricular (LV) placement and reduce dislodgement rates. The extractability of this active fixation LV lead has not been studied extensively. METHODS: Seventeen sheep were implanted with either an LV lead with a side helix (Model 20066, Model 20096, Medtronic) or a unipolar LV lead (Model 4193, Medtronic) as a control. Leads were extracted at approximately 26, 52, or 118 weeks. Standard extraction methodology was employed with quantitative traction up to 907 g (2 lbs.) using a locking stylet. Gross pathology and histology of the heart with particular attention to the lead tracts were performed. RESULTS: All leads were successfully removed in their entirety and required significantly less than 1 kg of traction force. The side helix disengaged from the vein as designed and resulted in no complications. No cardiac tissue was observed on any extracted lead. Gross pathology and histology were devoid of any helix-induced lesions in the vascular structures. The epicardium over the side helix was normal and the fibrotic reaction around the helix was not significantly different from that around the nonhelix portions of the study leads or the control lead. CONCLUSION: Extraction of the side helix, active fixation LV lead from the coronary veins in the sheep model is safe, without procedural complexity, and free of complications after long-term LV lead implant duration.


Asunto(s)
Dispositivos de Terapia de Resincronización Cardíaca , Vasos Coronarios , Ventrículos Cardíacos , Animales , Medios de Contraste/administración & dosificación , Angiografía Coronaria , Remoción de Dispositivos , Diseño de Equipo , Modelos Animales , Oveja Doméstica
13.
J Infect Dis ; 214(9): 1341-1348, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27521362

RESUMEN

BACKGROUND: Human cytomegalovirus (HCMV) infection causes disease in newborns and transplant recipients. A HCMV vaccine (Towne) protects transplant recipients. METHODS: The genomes of Towne and the nonattenuated Toledo strain were recombined, yielding 4 Towne/Toledo chimera vaccines. Each of 36 HCMV-seronegative men received 1 subcutaneous dose of 10, 100, or 1000 plaque-forming units (PFU) in cohorts of 3. Safety and immunogenicity were evaluated over 12 weeks after immunization and for 52 weeks for those who seroconverted. RESULTS: There were no serious local or systemic reactions. No subject had HCMV in urine or saliva. For chimera 3, none of 9 subjects seroconverted. For chimera 1, 1 of 9 seroconverted (the seroconverter received 100 PFU). For chimera 2, 3 subjects seroconverted (1 received 100 PFU, and 2 received 1000 PFU). For chimera 4, 7 subjects seroconverted (1 received 10 PFU, 3 received 100 PFU, and 3 received 1000 PFU). All 11 seroconverters developed low but detectable levels of neutralizing activity. CD4+ T-cell responses were detectable in 1 subject (who received 100 PFU of chimera 4). Seven subjects receiving chimera 2 or 4 had detectable CD8+ T-cell responses to IE1; 3 responded to 1-2 additional antigens. CONCLUSIONS: The Towne/Toledo chimera vaccine candidates were well tolerated and were not excreted. Additional human trials of chimeras 2 and 4 are appropriate. CLINICAL TRIALS REGISTRATION: NCT01195571.


Asunto(s)
Quimera/inmunología , Infecciones por Citomegalovirus/inmunología , Vacunas contra Citomegalovirus/inmunología , Citomegalovirus/inmunología , Vacunas Sintéticas/inmunología , Adulto , Anticuerpos Antivirales/inmunología , Linfocitos T CD4-Positivos/inmunología , Humanos , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Vacunación/métodos , Vacunas Atenuadas/inmunología , Vacunas Virales/inmunología , Adulto Joven
14.
Antimicrob Agents Chemother ; 59(3): 1558-68, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25534746

RESUMEN

Human cytomegalovirus (HCMV) is the most common infection causing poor outcomes among transplant recipients. Maternal infection and transplacental transmission are major causes of permanent birth defects. Although no active vaccines to prevent HCMV infection have been approved, passive immunization with HCMV-specific immunoglobulin has shown promise in the treatment of both transplant and congenital indications. Antibodies targeting the viral glycoprotein B (gB) surface protein are known to neutralize HCMV infectivity, with high-affinity binding being a desirable trait, both to compete with low-affinity antibodies that promote the transmission of virus across the placenta and to displace nonneutralizing antibodies binding nearby epitopes. Using a miniaturized screening technology to characterize secreted IgG from single human B lymphocytes, 30 antibodies directed against gB were previously cloned. The most potent clone, TRL345, is described here. Its measured affinity was 1 pM for the highly conserved site I of the AD-2 epitope of gB. Strain-independent neutralization was confirmed for 15 primary HCMV clinical isolates. TRL345 prevented HCMV infection of placental fibroblasts, smooth muscle cells, endothelial cells, and epithelial cells, and it inhibited postinfection HCMV spread in epithelial cells. The potential utility for preventing congenital transmission is supported by the blockage of HCMV infection of placental cell types central to virus transmission to the fetus, including differentiating cytotrophoblasts, trophoblast progenitor cells, and placental fibroblasts. Further, TRL345 was effective at controlling an ex vivo infection of human placental anchoring villi. TRL345 has been utilized on a commercial scale and is a candidate for clinical evaluation.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Afinidad de Anticuerpos/inmunología , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Anticuerpos Antivirales/inmunología , Linfocitos B/inmunología , Linfocitos B/virología , Línea Celular , Infecciones por Citomegalovirus/virología , Células Endoteliales/inmunología , Células Endoteliales/virología , Células Epiteliales/inmunología , Células Epiteliales/virología , Epítopos/inmunología , Femenino , Fibroblastos/inmunología , Fibroblastos/virología , Humanos , Inmunoglobulina G/inmunología , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/virología , Placenta/inmunología , Placenta/virología , Embarazo , Proteínas del Envoltorio Viral/inmunología
15.
Clin Infect Dis ; 57 Suppl 4: S189-92, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24257425

RESUMEN

Reported maternal-to-fetal rates of primary cytomegalovirus (CMV) infection during pregnancy have been between 30% and 50%. The highest rate of symptomatic congenital infection and sequelae occurs in about 25% of infected infants born of mothers with a primary infection during pregnancy. Symptomatic infants demonstrate a constellation of clinical features that reflect placental dysfunction and probable viral infection of the central nervous system of the fetus. In the United States, we estimate that about 8000 affected infants are born each year. Two options may be available to prevent or treat maternal CMV infection during pregnancy, especially for women with exposure to young children in the home. The first is hygienic intervention. Two studies support the simplicity, harmlessness, and effectiveness of hygienic intervention to prevent child-to-mother transmission of CMV among high-risk pregnant women who know they are susceptible. The second is CMV immunoglobulin. A meta-analysis of 2 clinical trials showed an efficacy of 50% if immunoglobulin is given to pregnant women who have acquired a primary CMV infection during pregnancy. These results mean that seronegative pregnant women have options to prevent fetal infection.


Asunto(s)
Infecciones por Citomegalovirus/transmisión , Citomegalovirus/aislamiento & purificación , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Complicaciones Infecciosas del Embarazo/prevención & control , Complicaciones Infecciosas del Embarazo/virología , Infecciones por Citomegalovirus/prevención & control , Femenino , Humanos , Inmunización Pasiva , Inmunoglobulinas Intravenosas/administración & dosificación , Recién Nacido , Control de Infecciones , Embarazo
16.
Clin Infect Dis ; 57 Suppl 4: S193-5, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24257426

RESUMEN

Primary cytomegalovirus (CMV) infection during the first half of pregnancy is responsible for the majority of symptomatic congenital infections. Between one-third and one-half of fetuses become infected, and up to one-half of infected fetuses will have neurologic or sensorineural sequelae at birth or later in life. Following favorable results obtained in animal experiments, observational studies have shown beneficial effects after administration of high-titer CMV hyperimmunoglobulin to pregnant women with fetal infection or disease subsequent to primary CMV infection. The mechanisms of action of hyperimmunoglobulin are multiple and not yet fully understood. However, they could reside in 2 major properties: (1) antiviral activities due to high-avidity neutralizing antibodies and (2) immunomodulating activities mostly including downregulation of cytokine-mediated cellular immune responses. A decreased viral pathogenicity occurs as an immediate consequence, whereas reduced placental inflammation and restored function are the long-term effects.


Asunto(s)
Infecciones por Citomegalovirus/prevención & control , Citomegalovirus/inmunología , Inmunoglobulinas/uso terapéutico , Complicaciones Infecciosas del Embarazo/prevención & control , Animales , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/inmunología , Femenino , Cobayas , Humanos , Embarazo , Complicaciones Infecciosas del Embarazo/tratamiento farmacológico , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/virología
17.
Br Med Bull ; 107: 57-68, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23954939

RESUMEN

INTRODUCTION: A primary maternal cytomegalovirus (CMV) during pregnancy causes newborn disease that includes hearing deficit and/or mental retardation. SOURCES OF DATA: Relevant published literature. AREAS OF AGREEMENT: There are no biologic obstacles to immunization against fetal/placental infection with CMV. AREAS OF UNCERTAINTY: CMV vaccine trials may be difficult due to a lack of public awareness of CMV. Vaccine trials that use fetal infection as an endpoint will be prolonged, since vaccination will need to occur preconception. AREAS TIMELY FOR DEVELOPING RESEARCH: Vaccines in preclinical development include antigens of the CMV gB glycoprotein and the gH/gL UL128, 130 and 131 pentameric complex. These antigens induce antibodies that block viral entry into fibroblasts and endothelial/epithelial cells. Vaccines immunogenic in animals include an inactivated virus with a wild-type UL131 gene, a DNA vaccine using a wild-type UL130 gene and peptide vaccines using peptides from UL130 and 131. CONCLUSIONS: In spite of these potential obstacles, successful evaluation of CMV vaccines is possible.


Asunto(s)
Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/prevención & control , Vacunas contra Citomegalovirus/inmunología , Animales , Preescolar , Infecciones por Citomegalovirus/inmunología , Vacunas contra Citomegalovirus/química , Vacunas contra Citomegalovirus/uso terapéutico , Femenino , Cobayas , Humanos , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/prevención & control
18.
J Infect Dis ; 205(2): 215-27, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22140265

RESUMEN

BACKGROUND: Primary cytomegalovirus (CMV) infection early in gestation causes severe disease. METHODS: Case patients were 32 congenitally infected children aged 1-5 years who had either hearing deficit and/or psychomotor retardation and whose mothers had a confirmed or probable primary CMV infection at ≤ 20 weeks' gestation. Control subjects were 32 congenitally infected normal children whose mothers had a confirmed primary infection at ≤ 20 weeks' gestation. Case patients and control subjects were matched by the weeks of maternal gestation (± 1 week) at the mother's infection and by the child's age (± 1 year) at evaluation. RESULTS: For the case patients and control subjects, the mean age was 3.0 years. The mean number of weeks of gestation at maternal infection was 11 weeks. The only risk factor for an affected child was the mother not receiving immunoglobulin (P = .001). Of the 32 case patients, only 4 mothers received CMV immunoglobulin, compared with 27 of the 32 mothers of control infants (adjusted odds ratio, 14 [95% confidence interval, 1.7-110]). The rate of both psychomotor retardation and hearing deficit decreased with immunoglobulin. CONCLUSIONS: These results support the efficacy of immunoglobulins for decreasing the severity of disabilities caused by fetal CMV infection after a primary maternal infection during pregnancy.


Asunto(s)
Infecciones por Citomegalovirus/complicaciones , Infecciones por Citomegalovirus/tratamiento farmacológico , Citomegalovirus , Enfermedades Fetales/tratamiento farmacológico , Inmunización Pasiva , Inmunoglobulinas/uso terapéutico , Complicaciones Infecciosas del Embarazo/tratamiento farmacológico , Adulto , Estudios de Casos y Controles , Preescolar , Femenino , Enfermedades Fetales/virología , Pérdida Auditiva/prevención & control , Pérdida Auditiva/virología , Humanos , Lactante , Modelos Logísticos , Masculino , Embarazo , Complicaciones Infecciosas del Embarazo/virología , Primer Trimestre del Embarazo , Segundo Trimestre del Embarazo , Trastornos Psicomotores/prevención & control , Trastornos Psicomotores/virología , Adulto Joven
19.
J Am Heart Assoc ; 12(4): e026295, 2023 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-36789852

RESUMEN

Background The PINNACLE FLX (Protection Against Embolism for Non-valvular AF [Atrial Fibrillation] Patients: Investigational Device Evaluation of the Watchman FLX LAA [Left Atrial Appendage] Closure Technology) trial evaluated the safety and efficacy of a next-generation left atrial appendage closure device (WATCHMAN FLX; Boston Scientific, Marlborough, MA). At 1 year, the study met the primary end points of safety and anatomical efficacy/appendage closure. This final report of the PINNACLE FLX trial includes the prespecified secondary end point of ischemic stroke or systemic embolism at 2 years, also making it the first report of 2-year outcomes with this next-generation left atrial appendage closure device. Methods and Results Patients with nonvalvular atrial fibrillation with CHA2DS2-VASc score ≥2 (men) or ≥3 (women), with an appropriate rationale for left atrial appendage closure, were enrolled to receive the left atrial appendage closure device at 29 US centers. Adverse events were assessed by an independent clinical events committee, and imaging was assessed by independent core laboratories. Among 395 implanted patients (36% women; mean age, 74 years; CHA2DS2-VASc, 4.2±1.5), the secondary efficacy end point of 2-year ischemic stroke or systemic embolism was met, with an absolute rate of 3.4% (annualized rate, 1.7%) and an upper 1-sided 95% confidence bound of 5.3%, which was superior to the 8.7% performance goal. Two-year rates of adverse events were as follows: 9.3% all-cause mortality, 5.5% cardiovascular death, 3.4% all stroke, and 10.1% major bleeding (Bleeding Academic Research Consortium 3 or 5). There were no additional systemic embolisms, device embolizations, pericardial effusions, or symptomatic device-related thrombi after 1 year. Conclusions The secondary end point of 2-year stroke or systemic embolism was met at 3.4%. In these final results of the PINNACLE FLX trial, the next-generation WATCHMAN FLX device demonstrated favorable safety and efficacy outcomes.


Asunto(s)
Apéndice Atrial , Fibrilación Atrial , Embolia , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Masculino , Humanos , Femenino , Anciano , Fibrilación Atrial/complicaciones , Fibrilación Atrial/cirugía , Apéndice Atrial/diagnóstico por imagen , Apéndice Atrial/cirugía , Resultado del Tratamiento , Accidente Cerebrovascular/prevención & control , Accidente Cerebrovascular/complicaciones , Embolia/prevención & control , Embolia/complicaciones , Hemorragia/etiología , Accidente Cerebrovascular Isquémico/etiología , Cateterismo Cardíaco/efectos adversos
20.
J Card Fail ; 18(2): 153-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22300784

RESUMEN

BACKGROUND: Several clinical trials have confirmed that cardiac resynchronization therapy (CRT) improves outcomes in well defined patient populations. It is uncertain, however, whether outcomes are similar in real-world clinical settings. This study compared outcomes after CRT with defibrillator (CRT-D) in a large real-world private-practice cardiology setting with those in the COMPANION multicenter trial. METHODS AND RESULTS: A total of 429 consecutive patients who received CRT-D for standard indications (group 1) were retrospectively compared with the 595 patients (group 3) in the COMPANION CRT-D cohort regarding survival and survival free of cardiovascular (CV) hospitalization. A subgroup of the group 1 patients who met the COMPANION entrance criteria (group 2) was also compared with the COMPANION cohort (group 3) both with and without propensity-matching statistical analysis. Survival and survival free of CV hospitalization was better in group 1 than in group 3. Survival in group 2 with and without propensity matching was similar to group 3. However, survival free of CV hospitalization was better in the real-world patients (group 2) even after adjustment for differences in baseline characteristics. CONCLUSIONS: Survival and CV hospitalization outcomes in a real-world clinical setting are as good as, or better than, those demonstrated in the COMPANION research trial.


Asunto(s)
Estimulación Cardíaca Artificial , Terapia de Resincronización Cardíaca , Insuficiencia Cardíaca/mortalidad , Insuficiencia Cardíaca/terapia , Anciano , Estudios de Cohortes , Bases de Datos Factuales , Ecocardiografía , Femenino , Insuficiencia Cardíaca/diagnóstico por imagen , Hospitalización , Humanos , Masculino , Minnesota , Ensayos Clínicos Controlados Aleatorios como Asunto , Estudios Retrospectivos , Análisis de Supervivencia , Resultado del Tratamiento , Wisconsin
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA