Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 207(10): 2534-2544, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34625521

RESUMO

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.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos/imunologia , Infecções por Citomegalovirus/imunologia , Células Matadoras Naturais/imunologia , Subpopulações de Linfócitos/imunologia , Proteínas Repressoras/imunologia , Proteínas Supressoras de Tumor/imunologia , Animais , Complexo CD3/imunologia , Humanos , Camundongos , Camundongos Transgênicos , Transcriptoma
2.
J Infect Dis ; 223(11): 2001-2012, 2021 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-33031517

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos , Infecções por Citomegalovirus , Vacinas contra Citomegalovirus , Imunidade Celular , Linfócitos T CD8-Positivos/imunologia , Infecções por Citomegalovirus/prevenção & controle , Vacinas contra Citomegalovirus/imunologia , Humanos , Vacinação
3.
Clin Infect Dis ; 71(6): 1491-1498, 2020 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-31628849

RESUMO

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.


Assuntos
Infecções por Citomegalovirus , Globulinas , Complicações Infecciosas na Gravidez , Criança , Pré-Escolar , Citomegalovirus , Feminino , Humanos , Lactente , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Gravidez , Gestantes
4.
J Virol ; 93(23)2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31511385

RESUMO

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.


Assuntos
Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/prevenção & controle , Vacinas contra Citomegalovirus/imunologia , Citomegalovirus/imunologia , Imunidade Humoral/imunologia , Imunização , Adulto , Idoso , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Antígenos Virais/sangue , Linhagem Celular , Infecções por Citomegalovirus/congênito , Infecções por Citomegalovirus/virologia , Método Duplo-Cego , Feminino , Humanos , Imunidade Celular , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Masculino , Pessoa de Meia-Idade , Estados Unidos , Vacinação , Replicação Viral , Adulto Jovem
5.
J Infect Dis ; 220(3): 411-419, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31535143

RESUMO

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.


Assuntos
Infecções por Citomegalovirus/imunologia , Vacinas contra Citomegalovirus/imunologia , Citomegalovirus/imunologia , Replicação Viral/imunologia , Adjuvantes Imunológicos/administração & dosagem , Adolescente , Adulto , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Método Duplo-Cego , ELISPOT/métodos , Feminino , Humanos , Imunidade Celular/imunologia , Imunidade Humoral/imunologia , Imunização/métodos , Masculino , Pessoa de Meia-Idade , Linfócitos T/imunologia , Vacinação/métodos , Eliminação de Partículas Virais/imunologia , Adulto Jovem
6.
Med Microbiol Immunol ; 208(3-4): 323-328, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31062089

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Interações Hospedeiro-Patógeno , Memória Imunológica , Latência Viral , Replicação Viral , Idoso , Animais , Infecções por Citomegalovirus/epidemiologia , Infecções por Citomegalovirus/virologia , Modelos Animais de Doenças , Humanos , Camundongos , Recidiva , Medição de Risco
7.
Med Microbiol Immunol ; 208(1): 69-80, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30203132

RESUMO

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.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Citomegalovirus/imunologia , Imunoglobulinas Intravenosas/imunologia , Antígenos Virais/imunologia , Células Cultivadas , Feminino , Voluntários Saudáveis , Humanos , Testes de Neutralização , Gravidez , Ensaio de Placa Viral
8.
Int J Mol Sci ; 20(13)2019 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-31261659

RESUMO

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.


Assuntos
Anticorpos Neutralizantes/imunologia , Vacinas contra Citomegalovirus/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Células CHO , Cricetinae , Cricetulus , Humanos , Coelhos , Vacinas Sintéticas/imunologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
9.
J Virol ; 91(7)2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28077654

RESUMO

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.


Assuntos
Anticorpos Neutralizantes/química , Anticorpos Antivirais/química , Infecções por Citomegalovirus/prevenção & controle , Citomegalovirus/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Especificidade de Anticorpos , Linhagem Celular , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Mapeamento de Epitopos , Humanos , Macaca mulatta , Ligação Proteica , Coelhos , Vacinação , Vacinas Virais/administração & dosagem , Internalização do Vírus
10.
Virus Genes ; 53(4): 650-655, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28391502

RESUMO

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.


Assuntos
Infecções por Citomegalovirus/virologia , Vacinas contra Citomegalovirus/imunologia , Citomegalovirus/genética , Citomegalovirus/imunologia , Recombinação Genética , Anticorpos Antivirais/imunologia , Citomegalovirus/classificação , Citomegalovirus/isolamento & purificação , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/prevenção & controle , Vacinas contra Citomegalovirus/administração & dosagem , Vacinas contra Citomegalovirus/genética , Genoma Viral , Genômica , Humanos , Imunização
11.
J Infect Dis ; 214(9): 1341-1348, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27521362

RESUMO

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.


Assuntos
Quimera/imunologia , Infecções por Citomegalovirus/imunologia , Vacinas contra Citomegalovirus/imunologia , Citomegalovirus/imunologia , Vacinas Sintéticas/imunologia , Adulto , Anticorpos Antivirais/imunologia , Linfócitos T CD4-Positivos/imunologia , Humanos , Ativação Linfocitária/imunologia , Masculino , Pessoa de Meia-Idade , Vacinação/métodos , Vacinas Atenuadas/imunologia , Vacinas Virais/imunologia , Adulto Jovem
12.
Antimicrob Agents Chemother ; 59(3): 1558-68, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25534746

RESUMO

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.


Assuntos
Anticorpos Neutralizantes/imunologia , Afinidade de Anticorpos/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Anticorpos Antivirais/imunologia , Linfócitos B/imunologia , Linfócitos B/virologia , Linhagem Celular , Infecções por Citomegalovirus/virologia , Células Endoteliais/imunologia , Células Endoteliais/virologia , Células Epiteliais/imunologia , Células Epiteliais/virologia , Epitopos/imunologia , Feminino , Fibroblastos/imunologia , Fibroblastos/virologia , Humanos , Imunoglobulina G/imunologia , Miócitos de Músculo Liso/imunologia , Miócitos de Músculo Liso/virologia , Placenta/imunologia , Placenta/virologia , Gravidez , Proteínas do Envelope Viral/imunologia
13.
Clin Infect Dis ; 57 Suppl 4: S189-92, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24257425

RESUMO

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.


Assuntos
Infecções por Citomegalovirus/transmissão , Citomegalovirus/isolamento & purificação , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Complicações Infecciosas na Gravidez/prevenção & controle , Complicações Infecciosas na Gravidez/virologia , Infecções por Citomegalovirus/prevenção & controle , Feminino , Humanos , Imunização Passiva , Imunoglobulinas Intravenosas/administração & dosagem , Recém-Nascido , Controle de Infecções , Gravidez
14.
Clin Infect Dis ; 57 Suppl 4: S193-5, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24257426

RESUMO

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.


Assuntos
Infecções por Citomegalovirus/prevenção & controle , Citomegalovirus/imunologia , Imunoglobulinas/uso terapêutico , Complicações Infecciosas na Gravidez/prevenção & controle , Animais , Infecções por Citomegalovirus/tratamento farmacológico , Infecções por Citomegalovirus/imunologia , Feminino , Cobaias , Humanos , Gravidez , Complicações Infecciosas na Gravidez/tratamento farmacológico , Complicações Infecciosas na Gravidez/imunologia , Complicações Infecciosas na Gravidez/virologia
15.
Br Med Bull ; 107: 57-68, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23954939

RESUMO

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.


Assuntos
Infecções por Citomegalovirus/congênito , Infecções por Citomegalovirus/prevenção & controle , Vacinas contra Citomegalovirus/imunologia , Animais , Pré-Escolar , Infecções por Citomegalovirus/imunologia , Vacinas contra Citomegalovirus/química , Vacinas contra Citomegalovirus/uso terapêutico , Feminino , Cobaias , Humanos , Gravidez , Complicações Infecciosas na Gravidez/imunologia , Complicações Infecciosas na Gravidez/prevenção & controle
16.
J Infect Dis ; 205(2): 215-27, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22140265

RESUMO

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.


Assuntos
Infecções por Citomegalovirus/complicações , Infecções por Citomegalovirus/tratamento farmacológico , Citomegalovirus , Doenças Fetais/tratamento farmacológico , Imunização Passiva , Imunoglobulinas/uso terapêutico , Complicações Infecciosas na Gravidez/tratamento farmacológico , Adulto , Estudos de Casos e Controles , Pré-Escolar , Feminino , Doenças Fetais/virologia , Perda Auditiva/prevenção & controle , Perda Auditiva/virologia , Humanos , Lactente , Modelos Logísticos , Masculino , Gravidez , Complicações Infecciosas na Gravidez/virologia , Primeiro Trimestre da Gravidez , Segundo Trimestre da Gravidez , Transtornos Psicomotores/prevenção & controle , Transtornos Psicomotores/virologia , Adulto Jovem
17.
J Biomed Biotechnol ; 2012: 428498, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22187535

RESUMO

Bacterial artificial chromosome (BAC) clones have proven invaluable for genetic manipulation of herpesvirus genomes. BAC cloning can also be useful for capturing representative genomes that comprise a viral stock or mixture. The Towne live attenuated cytomegalovirus vaccine was developed in the 1970s by serial passage in cultured fibroblasts. Although its safety, immunogenicity, and efficacy have been evaluated in nearly a thousand human subjects, the vaccine itself has been little studied. Instead, genetic composition and in vitro growth properties have been inferred from studies of laboratory stocks that may not always accurately represent the viruses that comprise the vaccine. Here we describe the use of BAC cloning to define the genotypic and phenotypic properties of viruses from the Towne vaccine. Given the extensive safety history of the Towne vaccine, these BACs provide a logical starting point for the development of next-generation rationally engineered cytomegalovirus vaccines.


Assuntos
Cromossomos Artificiais Bacterianos , Citomegalovirus/genética , Vacinas Virais/genética , Linhagem Celular , Clonagem Molecular , Citomegalovirus/crescimento & desenvolvimento , Citomegalovirus/imunologia , Genoma Viral , Humanos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Ensaio de Placa Viral , Vacinas Virais/imunologia
18.
Am J Pathol ; 177(3): 1298-310, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20651234

RESUMO

Human cytomegalovirus (HCMV) is the major viral cause of birth defects worldwide. Affected infants can have temporary symptoms that resolve soon after birth, such as growth restriction, and permanent disabilities, including neurological impairment. Passive immunization of pregnant women with primary HCMV infection is a promising treatment to prevent congenital disease. To understand the effects of sustained viral replication on the placenta and passive transfer of protective antibodies, we performed immunohistological analysis of placental specimens from women with untreated congenital infection, HCMV-specific hyperimmune globulin treatment, and uninfected controls. In untreated infection, viral replication proteins were found in trophoblasts and endothelial cells of chorionic villi and uterine arteries. Associated damage included extensive fibrinoid deposits, fibrosis, avascular villi, and edema, which could impair placental functions. Vascular endothelial growth factor and its receptor fms-like tyrosine kinase 1 (Flt1) were up-regulated, and amniotic fluid contained elevated levels of soluble Flt1 (sFlt1), an antiangiogenic protein, relative to placental growth factor. With hyperimmune globulin treatment, placentas appeared uninfected, vascular endothelial growth factor and Flt1 expression was reduced, and sFlt1 levels in amniotic fluid were lower. An increase in the number of chorionic villi and blood vessels over that in controls suggested compensatory development for a hypoxia-like condition. Taken together the results indicate that antibody treatment can suppress HCMV replication and prevent placental dysfunction, thus improving fetal outcome.


Assuntos
Vilosidades Coriônicas/virologia , Infecções por Citomegalovirus/metabolismo , Placenta/virologia , Trofoblastos/virologia , Proteínas Virais/metabolismo , Análise de Variância , Vilosidades Coriônicas/metabolismo , Vilosidades Coriônicas/patologia , Citomegalovirus , Infecções por Citomegalovirus/patologia , Infecções por Citomegalovirus/virologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imuno-Histoquímica , Placenta/metabolismo , Placenta/patologia , Gravidez , Trofoblastos/metabolismo , Trofoblastos/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
19.
Curr Opin Obstet Gynecol ; 23(2): 123-8, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21157339

RESUMO

PURPOSE OF REVIEW: To review current prenatal diagnosis and management of congenital cytomegalovirus (CMV) infections with emphasis on maternal screening and available interventions. RECENT FINDINGS: Recent findings include an enhanced understanding of the epidemiology, pathogenesis, and treatment of CMV infections; a knowledge of high-risk women particularly those with chronic exposure to a young child in the home; the availability of accurate methods for the serologic diagnosis of a primary CMV infection using either single or serial blood samples; accurate methods for the diagnosis of fetal infection via amniotic fluid; sensitive fetal and placental indicators for neonatal outcomes, and the availability of potentially effective interventions such as hygienic intervention and CMV hyperimmune globulin. SUMMARY: These findings suggest that serologic testing for CMV during pregnancy may be appropriate either using one-time testing or serial serologic testing throughout the first two trimesters of pregnancy and that education of pregnant women about CMV is necessary so that they can asses their risk and make informed choices about serologic screening.


Assuntos
Infecções por Citomegalovirus/diagnóstico , Complicações Infecciosas na Gravidez/diagnóstico , Líquido Amniótico/virologia , Europa (Continente) , Feminino , Humanos , Programas de Rastreamento/métodos , Obstetrícia/métodos , Gravidez , Resultado da Gravidez , Diagnóstico Pré-Natal/métodos , Risco , Testes Sorológicos , Estados Unidos
20.
Infect Dis Obstet Gynecol ; 2011: 1-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21836812

RESUMO

The epidemiology and pathogenesis of CMV infections among pregnant women have been intensely studied over the last three decades. This paper highlights recent developments that make either universal or limited serologic screening for CMV during pregnancy potentially attractive. The developments include an understanding of the pathogenesis of CMV infections, a knowledge of high-risk women, the availability of accurate methods for the serologic diagnosis of a primary CMV infection using either single or serial blood samples, accurate methods for the diagnosis of fetal infection via amniotic fluid, sensitive fetal and placental indicators for neonatal outcomes, and the availability of potentially effective interventions.


Assuntos
Infecções por Citomegalovirus/diagnóstico , Citomegalovirus/isolamento & purificação , Complicações Infecciosas na Gravidez/diagnóstico , Complicações Infecciosas na Gravidez/virologia , Diagnóstico Pré-Natal/métodos , Feminino , Humanos , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA