Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
PLoS Pathog ; 18(6): e1010403, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35737741

RESUMEN

There is now convincing evidence that the successful development of an effective CMV vaccine will require improved formulation and adjuvant selection that is capable of inducing both humoral and cellular immune responses. Here, we have designed a novel bivalent subunit vaccine formulation based on CMV-encoded oligomeric glycoprotein B (gB) and polyepitope protein in combination with human compatible TLR9 agonist CpG1018. The polyepitope protein includes multiple minimal HLA class I-restricted CD8+ T cell epitopes from different antigens of CMV. This subunit vaccine generated durable anti-viral antibodies, CMV-specific CD4+ and CD8+ T cell responses in multiple HLA expressing mice. Antibody responses included broad TH1 isotypes (IgG2a, IgG2b and IgG3) and potently neutralized CMV infection in fibroblasts and epithelial cells. Furthermore, polyfunctional antigen-specific T cell immunity and antiviral antibody responses showed long-term memory maintenance. These observations argue that this novel vaccine strategy, if applied to humans, could facilitate the generation of robust humoral and cellular immune responses which may be more effective in preventing CMV-associated complications in various clinical settings.


Asunto(s)
Infecciones por Citomegalovirus , Inmunidad Humoral , Adyuvantes Inmunológicos , Animales , Anticuerpos Antivirales , Linfocitos T CD8-positivos , Epítopos de Linfocito T/metabolismo , Humanos , Inmunidad Celular , Ratones , Oligodesoxirribonucleótidos , Receptor Toll-Like 9/metabolismo , Vacunas Combinadas , Vacunas de Subunidad
2.
Immunol Cell Biol ; 94(10): 964-974, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27297581

RESUMEN

Cross-presentation of exogenous protein antigens by B cells through the major histocompatibility complex (MHC) class I pathway in lymphoid malignancies, and transplant setting has been recognised as an important mediator of immune pathogenesis and T cell-mediated immune regulation. However, the precise mechanism of cross-presentation of exogenous antigens in B cells has remained unresolved. Here we have delineated a novel pathway for cross-presentation in B cells, which involves synergistic cooperation of the proteasome and autophagy. After endocytosis, protein antigen is processed through an autophagy- and proteasome-dependent pathway and CD8+ T-cell epitopes are loaded onto MHC class I molecules within the autophagolysomal compartment rather than the conventional secretory pathway, which requires transporters associated with antigen processing-dependent transport. Interestingly, this cross-presentation was critically dependent on valosin-containing protein (VCP)/p97 ATPase through its participation in autophagy. Loss of VCP/p97 ATPase was coincident with accumulation of LC3-II and marked reduction in antigen presentation. These observations provide unique insight on how the autophagy and proteasomal degradation systems interconnect to coordinate MHC class I-restricted cross-presentation in B cells.


Asunto(s)
Autofagia/inmunología , Linfocitos B/citología , Linfocitos B/inmunología , Reactividad Cruzada/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Complejo de la Endopetidasa Proteasomal/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Adenosina Trifosfato/farmacología , Autofagia/efectos de los fármacos , Proteínas Relacionadas con la Autofagia/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Reactividad Cruzada/efectos de los fármacos , Citomegalovirus/metabolismo , Epítopos de Linfocito T/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Proteína que Contiene Valosina/metabolismo , Proteínas Virales/metabolismo
3.
J Virol ; 89(17): 9137-41, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26109734

RESUMEN

Here we present evidence for previously unappreciated B-cell immune dysregulation during acute Epstein-Barr virus (EBV)-associated infectious mononucleosis (IM). Longitudinal analyses revealed that patients with acute IM have undetectable EBV-specific neutralizing antibodies and gp350-specific B-cell responses, which were associated with a significant reduction in memory B cells and no evidence of circulating antibody-secreting cells. These observations correlate with dysregulation of tumor necrosis factor family members BAFF and APRIL and increased expression of FAS on circulating B cells.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Linfocitos B/inmunología , Herpesvirus Humano 4/inmunología , Mononucleosis Infecciosa/inmunología , Factor Activador de Células B/inmunología , Linfocitos B/virología , Linfocitos T CD8-positivos/inmunología , Proteínas de la Cápside/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/virología , Humanos , Memoria Inmunológica/inmunología , Mononucleosis Infecciosa/virología , Activación de Linfocitos/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Proteínas de la Matriz Viral/inmunología , Receptor fas/metabolismo
4.
J Infect Dis ; 212(12): 1957-61, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26080368

RESUMEN

Acute infectious mononucleosis (IM) is associated with altered expression of inflammatory cytokines and disturbed T-cell homeostasis, however, the precise mechanism of this immune dysregulation remains unresolved. In the current study we demonstrated a significant loss of circulating myeloid and plasmacytoid dendritic cells (DCs) during acute IM, a loss correlated with the severity of clinical symptoms. In vitro exposure of blood DCs to acute IM plasma resulted in loss of plasmacytoid DCs, and further studies with individual cytokines showed that exposure to interleukin 10 could replicate this effect. Our data provide important mechanistic insight into dysregulated immune homeostasis during acute IM.


Asunto(s)
Células Sanguíneas/efectos de los fármacos , Células Dendríticas/inmunología , Tolerancia Inmunológica , Mononucleosis Infecciosa/patología , Interleucina-10/sangre , Adolescente , Adulto , Supervivencia Celular/efectos de los fármacos , Humanos , Mononucleosis Infecciosa/inmunología , Interleucina-10/metabolismo , Adulto Joven
5.
Blood ; 120(16): 3237-45, 2012 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-22942184

RESUMEN

T-box transcription factors T-bet (Tbx21) and Eomesodermin (Eomes) are critical players in CD8(+) cytotoxic T lymphocyte effector function and differentiation, but how the expression of these transcription factors is regulated remains poorly defined. Here we show that dominant T cells directed toward human CMV, expressing significantly higher levels of T-bet with graded loss of Eomes expression (T-bet(hi)Eomes(hi/lo)), are more efficient in recognizing endogenously processed peptide-major histocompatibility complexes (pMHC) compared with subdominant virus-specific T cells expressing lower levels of T-bet and high levels of Eomes (T-bet(int)Eomes(hi)). Paradoxically, the T-bet(hi)Eomes(hi/lo) dominant populations that efficiently recognized endogenous antigen demonstrated lower intrinsic avidity for pMHC, whereas T-bet(int)Eomes(hi) subdominant populations were characterized by higher pMHC avidity and less efficient recognition of virus-infected cells. Importantly, differential endogenous viral antigen recognition by CMV-specific CD8(+) T cells also correlated with the differentiation status and expression of perforin, granzyme B and K. Furthermore, we demonstrate that the expression of T-bet correlates with clonal expansion, differentiation status, and expression of perforin, granzyme B and K in antigen-specific T cells. These findings illustrate how endogenous viral antigen presentation during persistent viral infection may influence the transcriptional program of virus-specific T cells and their functional profile in the peripheral blood of humans.


Asunto(s)
Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/inmunología , Proteínas de Dominio T Box/metabolismo , Linfocitos T Citotóxicos/inmunología , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Células Cultivadas , Citocinas/metabolismo , Citomegalovirus/inmunología , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/microbiología , Citometría de Flujo , Granzimas/metabolismo , Humanos , Interferón gamma/metabolismo , Activación de Linfocitos , Complejo Mayor de Histocompatibilidad/inmunología , Perforina/metabolismo , Proteínas de Dominio T Box/inmunología
6.
Nat Commun ; 14(1): 4371, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37553346

RESUMEN

The recent emergence of a causal link between Epstein-Barr virus (EBV) and multiple sclerosis has generated considerable interest in the development of an effective vaccine against EBV. Here we describe a vaccine formulation based on a lymph node targeting Amphiphile vaccine adjuvant, Amphiphile-CpG, admixed with EBV gp350 glycoprotein and an engineered EBV polyepitope protein that includes 20 CD8+ T cell epitopes from EBV latent and lytic antigens. Potent gp350-specific IgG responses are induced in mice with titers >100,000 in Amphiphile-CpG vaccinated mice. Immunization including Amphiphile-CpG also induces high frequencies of polyfunctional gp350-specific CD4+ T cells and EBV-specific CD8+ T cells that are 2-fold greater than soluble CpG and are maintained for >7 months post immunization. This combination of broad humoral and cellular immunity against multiple viral determinants is likely to provide better protection against primary infection and control of latently infected B cells leading to protection against the development of EBV-associated diseases.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Herpesvirus Humano 4 , Ratones , Animales , Infecciones por Virus de Epstein-Barr/prevención & control , Linfocitos T CD8-positivos , Epítopos de Linfocito T , Ganglios Linfáticos , Vacunas de Subunidad
7.
J Gen Virol ; 92(Pt 5): 1021-1031, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21307228

RESUMEN

Natural human cytomegalovirus (CMV) infection is characterized by a strain-specific neutralizing antibody response. This is particularly relevant in clinical settings such as transplantation and pregnancy where reinfection with heterologous strains occurs and the immune system does not mount an effective response against the infecting strain due to underlying immunosuppression. There is an emerging argument that a CMV vaccine that induces high titres of cross-neutralizing antibodies will be more effective in protecting individuals from infection with antigenically different CMV strains. In addition, induction of cell-mediated immunity offers the additional advantage of targeting virus-infected cells. This study presents a novel formulation of a CMV vaccine that, by combining recombinant soluble gB protein with a Toll-like receptor 9 agonist (CpG ODN1826) and immune-stimulating complexes (AbISCO 100), was able to elicit strong polyfunctional CMV-specific cellular and cross-neutralizing humoral immune responses. These data demonstrated that prime-boost immunization of human leukocyte antigen (HLA)-A2 mice with gB protein in combination with CpG ODN1826 and AbISCO 100 induced long-lasting CMV-specific CD4(+) and CD8(+) T-cell and humoral responses. Furthermore, these responses neutralized infection with multiple strains of CMV expressing different gB genotypes and afforded protection against challenge with recombinant vaccinia virus encoding the gB protein. These observations argue that this novel vaccine strategy, if applied to humans, should facilitate the generation of a robust, pluripotent immune response, which may be more effective in preventing infection with multiple strains of CMV.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacunas contra Citomegalovirus/inmunología , Oligodesoxirribonucleótidos/administración & dosificación , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Complejo Antígeno-Anticuerpo/administración & dosificación , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra Citomegalovirus/administración & dosificación , Femenino , Humanos , Inmunización Secundaria/métodos , Ratones , Ratones Transgénicos , Receptor Toll-Like 9/agonistas , Vacunación/métodos , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/administración & dosificación
8.
Expert Rev Vaccines ; 18(5): 457-474, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30987475

RESUMEN

INTRODUCTION: Epstein-Barr virus (EBV) infects more than 95% of the world's population and is associated with infectious mononucleosis as well as a number of cancers in various geographical locations. Despite its significant health burden, no licenced prophylactic or therapeutic vaccines are available. Areas covered: Over the last two decades, our understanding of the role of EBV infection in the pathogenesis and immune regulation of EBV-associated diseases has provided new lines of research to conceptualize various novel prophylactic and therapeutic approaches to control EBV-associated disease. In this review, we evaluate the prophylactic and therapeutic vaccine approaches against EBV and various immunotherapeutic strategies against a number of EBV-associated malignancies. This review also describes the existing and future prospects of improved EBV-targeted therapeutic strategies. Expert opinion: It is anticipated that these emerging strategies will provide answers for the major challenges in EBV vaccine development and help improve the efficacy of novel therapeutic strategies.


Asunto(s)
Infecciones por Virus de Epstein-Barr/prevención & control , Infecciones por Virus de Epstein-Barr/terapia , Herpesvirus Humano 4/inmunología , Inmunoterapia/métodos , Vacunas Virales/inmunología , Vacunas Virales/aislamiento & purificación , Infecciones por Virus de Epstein-Barr/complicaciones , Trastornos Linfoproliferativos/prevención & control , Carcinoma Nasofaríngeo/prevención & control , Neoplasias Nasofaríngeas/prevención & control
9.
Blood Adv ; 2(6): 656-668, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29563122

RESUMEN

Immune reconstitution following hematopoietic stem cell transplantation (HSCT) is critical in preventing harmful sequelae in recipients with cytomegalovirus (CMV) infection. To understand the molecular mechanisms underlying immune reconstitution kinetics, we profiled the transcriptome-chromatin accessibility landscape of CMV-specific CD8+ T cells from HCST recipients with different immune reconstitution efficiencies. CMV-specific T cells from HSCT recipients with stable antiviral immunity expressed higher levels of interferon/defense response and cell cycle genes in an interconnected network involving PI3KCG, STAT5B, NFAT, RBPJ, and lower HDAC6, increasing chromatin accessibility at the enhancer regions of immune and T-cell receptor signaling pathway genes. By contrast, the transcriptional and epigenomic signatures of CMV-specific T cells from HSCT recipients with unstable immune reconstitution showed commonalities with T-cell responses in other nonresolving chronic infections. These signatures included higher levels of EGR and KLF factors that, along with lower JARID2 expression, maintained higher accessibility at promoter and CpG-rich regions of genes associated with apoptosis. Furthermore, epigenetic targeting via inhibition of HDAC6 or JARID2 enhanced the transcription of genes associated with differential responses, suggesting that drugs targeting epigenomic modifiers may have therapeutic potential for enhancing immune reconstitution in HSCT recipients. Taken together, these analyses demonstrate that transcription factors and chromatin modulators create different chromatin accessibility landscapes in T cells of HSCT recipients that not only affect immediate gene expression but also differentially prime cells for responses to additional signals. Epigenetic therapy may be a promising strategy to promote immune reconstitution in HSCT recipients.


Asunto(s)
Reprogramación Celular/genética , Reprogramación Celular/inmunología , Epigénesis Genética , Reconstitución Inmune , Linfocitos T/inmunología , Linfocitos T/metabolismo , Ensamble y Desensamble de Cromatina , Citomegalovirus/inmunología , Infecciones por Citomegalovirus/etiología , Regulación de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Especificidad del Receptor de Antígeno de Linfocitos T , Factores de Transcripción/metabolismo , Transcriptoma , Receptores de Trasplantes , Replicación Viral
10.
Expert Rev Vaccines ; 16(4): 377-390, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28276306

RESUMEN

INTRODUCTION: Epstein-Barr virus (EBV) is a ubiquitous herpesvirus associated with a number of clinical manifestations. Primary EBV infection in young adolescents often manifests as acute infectious mononucleosis and latent infection is associated with multiple lymphoid and epithelial cancers and autoimmune disorders, particularly multiple sclerosis. Areas covered: Over the last decade, our understanding of pathogenesis and immune regulation of EBV-associated diseases has provided an important platform for the development of novel vaccine formulations. In this review, we discuss developmental strategies for prophylactic and therapeutic EBV vaccines which have been assessed in preclinical and clinical settings. Expert commentary: Major roadblocks in EBV vaccine development include no precise understanding of the clinical correlates of protection, uncertainty about adjuvant selection and the unavailability of appropriate animal models. Recent development of new EBV vaccine formulations provides exciting opportunities for the formal clinical assessment of novel formulations.


Asunto(s)
Infecciones por Virus de Epstein-Barr/prevención & control , Herpesvirus Humano 4/inmunología , Vacunas Virales/inmunología , Animales , Carcinoma/prevención & control , Carcinoma/virología , Modelos Animales de Enfermedad , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/virología , Humanos , Linfoma/prevención & control , Linfoma/virología , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/prevención & control , Neoplasias Nasofaríngeas/virología , Ensayos Clínicos Controlados Aleatorios como Asunto , Vacunas Virales/administración & dosificación , Vacunas Virales/química
11.
Mol Ther Methods Clin Dev ; 3: 16058, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27606351

RESUMEN

Viral infections including cytomegalovirus, Epstein-Barr virus, adenovirus, and BK virus are a common and predictable problem in transplant recipients. While cellular immune therapies have been successfully used to tackle infectious complications in transplant recipients, manufacturing immunotherapies to address the multitude of possible pathogens can be technically challenging and labor-intensive. Here we describe a novel adenoviral antigen presentation platform (Ad-MvP) as a tool for rapid generation of multivirus-specific T-cells in a single step. Ad-MvP encodes 32 CD8+ T-cell epitopes from cytomegalovirus, Epstein-Barr virus, adenovirus, and BK virus as a contiguous polyepitope. We demonstrate that Ad-MvP vector can be successfully used for rapid in vitro expansion of multivirus-specific T-cells from transplant recipients and in vivo priming of antiviral T-cell immunity. Most importantly, using an in vivo murine model of Epstein-Barr virus-induced lymphoma, we also show that adoptive immunotherapy with Ad-MvP expanded autologous and allogeneic multivirus-specific T-cells is highly effective in controlling Epstein-Barr virus tumor outgrowth and improving overall survival. We propose that Ad-MvP has wide ranging therapeutic applications in greatly facilitating in vivo priming of antiviral T-cells, the generation of third-party T-cell banks as "off-the-shelf" therapeutics as well as autologous T-cell therapies for transplant patients.

12.
Hum Vaccin Immunother ; 10(4): 1064-77, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24463331

RESUMEN

Recent studies have suggested that a successful subunit human cytomegalovirus (CMV) vaccine requires improved formulation to generate broad-based anti-viral immunity following immunization. Here we report the development of a non-live protein-based vaccine strategy for CMV based on a polyepitope protein and CMV glycoprotein B (gB) adjuvanted with TLR4 and/or TLR9 agonists. The polyepitope protein includes contiguous multiple MHC class I-restricted epitopes with an aim to induce CD8(+) T cell immunity, while gB is an important target for CD4(+) T cell immunity and neutralizing antibodies. Optimal immunogenicity of this bivalent non-live protein vaccine formulation was dependent upon the co-administration of both the TLR4 and TLR9 agonist, which was associated with the activation of innate immune signatures and the influx of different DC subsets including plasmacytoid DCs and migratory CD8-DEC205+CD103-CD326- langerin-negative dermal DCs into the draining lymph nodes. Furthermore these professional antigen presenting cells also expressed IL-6, IL-12p70, TNFα, and IFNα which play a crucial role in the activation of adaptive immunity. In summary, this study provides a novel platform technology in which broad-based anti-CMV immune responses upon vaccination can be maximised by co-delivery of viral antigens and TLR4 and 9 agonists which induce activation of innate immune signatures and promote potent antigen acquisition and cross-presentation by multiple DC subsets.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacunas contra Citomegalovirus/inmunología , Epítopos/inmunología , Inmunización/métodos , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 9/agonistas , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Linfocitos T CD8-positivos/inmunología , Vacunas contra Citomegalovirus/administración & dosificación , Vacunas contra Citomegalovirus/genética , Células Dendríticas/inmunología , Epítopos/genética , Ganglios Linfáticos/inmunología , Ratones Transgénicos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética
13.
Cancer Res ; 74(13): 3466-76, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24795429

RESUMEN

Glioblastoma multiforme (GBM) is one of the most aggressive human brain malignancies. Even with optimal treatment, median survival is less than 6 months for patients with recurrent GBM. Immune-based therapies have the potential to improve patient outcome by supplementing standard treatment. Expression of human cytomegalovirus (CMV) antigens in GBM tissues provides the unique opportunity to target viral antigens for GBM therapy. Here, we report findings of a formal clinical assessment of safety and potential clinical efficacy of autologous CMV-specific T-cell therapy as a consolidative treatment for recurrent GBM. From a total of 19 patients with recurrent GBM, CMV-specific T cells were successfully expanded from 13 patients (68.4%), 11 of whom received up to four T-cell infusions. Combination therapy based on T-cell infusion and chemotherapy was well tolerated, and we detected only minor adverse events. The overall survival of these patients since first recurrence ranged from 133 to 2,428 days, with a median overall survival of 403 days. Most importantly, 4 of 10 patients that completed the treatment remained progression free during the study period. Furthermore, molecular profiling of CMV-specific T-cell therapy from these patients revealed distinct gene expression signatures, which correlated with their clinical response. Our study suggests that a combination therapy with autologous CMV-specific T cells and chemotherapy is a safe novel treatment option and may offer clinical benefit for patients with recurrent GBM.


Asunto(s)
Neoplasias Encefálicas/terapia , Citomegalovirus/inmunología , Glioblastoma/terapia , Inmunoterapia Adoptiva/efectos adversos , Linfocitos T/trasplante , Adulto , Anciano , Antígenos Virales/inmunología , Antígenos Virales/uso terapéutico , Encéfalo/patología , Neoplasias Encefálicas/mortalidad , Tratamiento Basado en Trasplante de Células y Tejidos , Supervivencia sin Enfermedad , Femenino , Expresión Génica , Glioblastoma/mortalidad , Humanos , Inmunoterapia Adoptiva/métodos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/inmunología , Trasplante Autólogo/efectos adversos , Trasplante Autólogo/métodos , Adulto Joven
14.
Expert Rev Vaccines ; 12(6): 661-76, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23750795

RESUMEN

It is now well over a decade since the US Institute of Medicine of the National Academy of Sciences assigned the highest priority for a vaccine to prevent congenital human CMV infection, which was subsequently endorsed by the US National Vaccine Program Office. In spite of extensive efforts over many years, successful licensure of a CMV vaccine formulation remains elusive. While the understanding of immune regulation of CMV infection in healthy virus carriers and diseased patients has dramatically improved, traditional vaccine development programs have failed to exploit this knowledge. Until recently, most efforts have concentrated on designing vaccine formulations that block CMV infection through neutralizing antibodies. However, studies carried out in various disease settings, especially in transplant patients, have clearly emphasized the importance of cellular immunity and it is indeed encouraging to see that recent CMV vaccine development programs have started to incorporate this arm of the immune system. A number of new vaccine candidates have been found to be effective in preclinical studies, and are able to induce CMV-specific immune responses in clinical studies, although firm evidence for long-term efficacy is not yet available. For successful implementation of these vaccines in clinical settings, it will be important to demonstrate that the vaccine can induce effective levels of immunity for prevention of transmission of viral infection from mother to unborn baby and thus reduce CMV-related pathogenesis. For transplant recipients, vaccine strategies should be aimed at the induction of immunity that restricts viral reactivation and limits development of disease.


Asunto(s)
Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/prevención & control , Vacunas contra Citomegalovirus/inmunología , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Ensayos Clínicos como Asunto , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/transmisión , Vacunas contra Citomegalovirus/administración & dosificación , Descubrimiento de Drogas/tendencias , Humanos , Estados Unidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA