Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 129(7): 866-878, 2017 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-28003273

RESUMEN

Human and mouse chronic lymphocytic leukemia (CLL) develops from CD5+ B cells that in mice and macaques are known to define the distinct B1a B-cell lineage. B1a cells are characterized by lack of germinal center (GC) development, and the B1a cell population is increased in mice with reduced GC formation. As a major mediator of follicular B-cell migration, the G protein-coupled receptor Epstein-Barr virus-induced gene 2 (EBI2 or GPR183) directs B-cell migration in the lymphoid follicles in response to its endogenous ligands, oxysterols. Thus, upregulation of EBI2 drives the B cells toward the extrafollicular area, whereas downregulation is essential for GC formation. We therefore speculated whether increased expression of EBI2 would lead to an expanded B1 cell subset and, ultimately, progression to CLL. Here, we demonstrate that B-cell-targeted expression of human EBI2 (hEBI2) in mice reduces GC-dependent immune responses, reduces total immunoglobulin M (IgM) and IgG levels, and leads to increased proliferation and upregulation of cellular oncogenes. Furthermore, hEBI2 overexpression leads to an abnormally expanded CD5+ B1a B-cell subset (present as early as 4 days after birth), late-onset lymphoid cancer development, and premature death. These findings are highly similar to those observed in CLL patients and identify EBI2 as a promoter of B-cell malignancies.


Asunto(s)
Linfocitos B/patología , Centro Germinal/patología , Leucemia Linfocítica Crónica de Células B/genética , Linfoma/genética , Receptores Acoplados a Proteínas G/genética , Regulación hacia Arriba , Animales , Linfocitos B/inmunología , Antígenos CD5/análisis , Antígenos CD5/inmunología , Regulación Neoplásica de la Expresión Génica , Centro Germinal/citología , Centro Germinal/inmunología , Leucemia Linfocítica Crónica de Células B/inmunología , Leucemia Linfocítica Crónica de Células B/patología , Linfoma/inmunología , Linfoma/patología , Ratones , Receptores Acoplados a Proteínas G/inmunología
2.
Int J Mol Sci ; 18(4)2017 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-28420073

RESUMEN

Vaccination is one of the most efficient tools for disease prevention, and a continuously growing field of research. However, despite progress, we still need more efficient and cost-effective vaccines that would improve access to those in need. In this review, we will describe the status of virus-vectored vaccine technology with a focus on adenoviral-based vaccines. Adenovirus (Ad) vaccines have proven to be efficient in military vaccinations against Ad4 and Ad7 and as highly efficient vectored vaccines against rabies. The question of how other adenovirus-based vaccines can become as efficient as the rabies vaccine is the underlying theme in this review. Here, we will first give an overview of the basic properties of vectored vaccines, followed by an introduction to the characteristics of adenoviral vectors and previously tested modifications of the vector backbone and expression cassettes, with a focus on how they can contribute to increased vaccine cost-effectiveness. Finally, we will highlight a few successful examples of research that have attempted to improve the use of adenoviral-based vaccines by improving the transgene immunogenicity.


Asunto(s)
Adenoviridae/genética , Análisis Costo-Beneficio , Vectores Genéticos/genética , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Animales , Biotecnología , Humanos , Inmunidad , Transgenes/genética , Transgenes/inmunología , Vacunas Sintéticas/administración & dosificación , Replicación Viral
3.
Front Immunol ; 15: 1338492, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38380318

RESUMEN

Modified vaccinia virus Ankara is a versatile vaccine vector, well suited for transgene delivery, with an excellent safety profile. However, certain transgenes render recombinant MVA (rMVA) genetically unstable, leading to the accumulation of mutated rMVA with impaired transgene expression. This represents a major challenge for upscaling and manufacturing of rMVA vaccines. To prevent transgene-mediated negative selection, the continuous avian cell line AGE1.CR pIX (CR pIX) was modified to suppress transgene expression during rMVA generation and amplification. This was achieved by constitutively expressing a tetracycline repressor (TetR) together with a rat-derived shRNA in engineered CR pIX PRO suppressor cells targeting an operator element (tetO) and 3' untranslated sequence motif on a chimeric poxviral promoter and the transgene mRNA, respectively. This cell line was instrumental in generating two rMVA (isolate CR19) expressing a Macaca fascicularis papillomavirus type 3 (MfPV3) E1E2E6E7 artificially-fused polyprotein following recombination-mediated integration of the coding sequences into the DelIII (CR19 M-DelIII) or TK locus (CR19 M-TK), respectively. Characterization of rMVA on parental CR pIX or engineered CR pIX PRO suppressor cells revealed enhanced replication kinetics, higher virus titers and a focus morphology equaling wild-type MVA, when transgene expression was suppressed. Serially passaging both rMVA ten times on parental CR pIX cells and tracking E1E2E6E7 expression by flow cytometry revealed a rapid loss of transgene product after only few passages. PCR analysis and next-generation sequencing demonstrated that rMVA accumulated mutations within the E1E2E6E7 open reading frame (CR19 M-TK) or deletions of the whole transgene cassette (CR19 M-DelIII). In contrast, CR pIX PRO suppressor cells preserved robust transgene expression for up to 10 passages, however, rMVAs were more stable when E1E2E6E7 was integrated into the TK as compared to the DelIII locus. In conclusion, sustained knock-down of transgene expression in CR pIX PRO suppressor cells facilitates the generation, propagation and large-scale manufacturing of rMVA with transgenes hampering viral replication.


Asunto(s)
Vacunas Sintéticas , Virus Vaccinia , Ratas , Animales , Virus Vaccinia/genética , Linfocitos T CD8-positivos , Transgenes
4.
Basic Clin Pharmacol Toxicol ; 133(4): 378-389, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37621135

RESUMEN

Adhesion G protein-coupled receptors (aGPCRs) constitute the second largest subclass of the GPCR superfamily. Although canonical GPCRs are explored pharmacologically as drug targets, no clinically approved drugs target the aGPCR family so far. The aGPCR GPR56/ADGRG1 stands out as an especially promising target, given its direct link to the monogenetic disease bilateral frontoparietal polymicrogyria and implications in cancers. Key to understanding GPCR pharmacology has been mapping out intracellular signalling activity. Detection of GPCR signalling in the Gαs /Gαi /Gαq G protein pathways is feasible with second messenger detection systems. However, in the case of Gα12/13 -coupled receptors, like GPR56, signalling detection is more challenging due to the lack of direct second messenger generation. To overcome this challenge, we engineered a Gαq chimera to translate Gα12/13 signalling. We show the ability of the chimeric GαΔ6q12myr and GαΔ6q13myr to translate basal Gα12/13 signalling of GPR56 to a Gαq readout in transcription factor luciferase reporter systems and show that the established peptide ligands (P7 and P19) function to enhance this signal. We further demonstrate the ability to directly influence the generation of second messengers in inositol-3-phosphate assays. In the future, these chimeric G proteins could facilitate basic functional studies, drug screenings and deorphanization of other aGPCRs.


Asunto(s)
Receptores Acoplados a Proteínas G , Transducción de Señal , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Unión al GTP/metabolismo , Péptidos
5.
J Biol Chem ; 286(33): 29292-29302, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21673108

RESUMEN

The Epstein-Barr virus-induced receptor 2 (EBI2) is a constitutively active seven-transmembrane receptor, which was recently shown to orchestrate the positioning of B cells in the follicle. To date, no ligands, endogenously or synthetic, have been identified that modulate EBI2 activity. Here we describe an inverse agonist, GSK682753A, which selectively inhibited the constitutive activity of EBI2 with high potency and efficacy. In cAMP-response element-binding protein-based reporter and guanosine 5'-3-O-(thio)triphosphate (GTPγS) binding assays, the potency of this compound was 2.6-53.6 nm, and its inhibitory efficacy was 75%. In addition, we show that EBI2 constitutively activated extracellular signal-regulated kinase (ERK) in a pertussis toxin-insensitive manner. Intriguingly, GSK682753A inhibited ERK phosphorylation, GTPγS binding, and cAMP-response element-binding protein activation with similar potency. Overexpression of EBI2 profoundly potentiated antibody-stimulated ex vivo proliferation of murine B cells compared with WT cells, whereas this was equivalently reduced for EBI2-deficient B cells. Inhibition of EBI2 constitutive activity suppressed the proliferation in all cases. Importantly, the suppression was of much higher potency (32-fold) in WT or EBI2-overexpressing B cells compared with EBI2-deficient counterparts. Finally, we screened GSK682753A against an EBI2 mutant library to determine putative molecular binding determinants in EBI2. We identified Phe(111) at position III:08/3.32 as being crucial for GSK682753A inverse agonism because Ala substitution resulted in a >500-fold decrease in IC(50). In conclusion, we present the first ligand targeting EBI2. In turn, this molecule provides a useful tool for further characterization of EBI2 as well as serving as a potent lead compound.


Asunto(s)
Linfocitos B/metabolismo , Proliferación Celular/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Oxazoles/farmacología , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Compuestos de Espiro/farmacología , Animales , Linfocitos B/citología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HEK293 , Compuestos Heterocíclicos de 4 o más Anillos/química , Humanos , Ratones , Ratones Mutantes , Oxazoles/química , Fosforilación/efectos de los fármacos , Fosforilación/genética , Receptores Acoplados a Proteínas G/genética , Elementos de Respuesta/fisiología , Compuestos de Espiro/química
6.
Eur J Immunol ; 39(10): 2725-36, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19637230

RESUMEN

Antigen-specific immunotherapy is an attractive strategy for cancer control. In the context of antiviral vaccines, adenoviral vectors have emerged as a favorable means for immunization. Therefore, we chose a strategy combining use of these vectors with another successful approach, namely linkage of the vaccine antigen to invariant chain (Ii). To evaluate this strategy we used a mouse model, in which an immunodominant epitope (GP33) of the LCMV glycoprotein (GP) represents the tumor-associated neoantigen. Prophylactic vaccination of C57BL/6 mice with a replication-deficient human adenovirus 5 vector encoding GP linked to Ii (Ad-Ii-GP) resulted in complete protection against GP33-expressing B16.F10 tumors. Therapeutic vaccination with Ad-Ii-GP delayed tumor growth by more than 2 wk compared with sham vaccination. Notably, therapeutic vaccination with the linked vaccine was significantly better than vaccination with adenovirus expressing GP alone (Ad-GP), or GP and Ii unlinked (Ad-GP+Ii). Ad-Ii-GP- induced tumor control depended on an improved generation of the tumor-associated neoantigen-specific CD8(+) T-cell response and was independent of CD4(+) T cells. IFN-gamma was shown to be a key player during the tumor degradation. Finally, Ad-Ii-GP but not Ad-GP vaccination can break the immunological non-reactivity in GP transgenic mice indicating that our vaccine strategy will prove efficient also against endogenous tumor antigens.


Asunto(s)
Adenoviridae/genética , Antígenos de Diferenciación de Linfocitos B/inmunología , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Vectores Genéticos/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Animales , Antígenos de Diferenciación de Linfocitos B/genética , Antígenos de Neoplasias/genética , Antígenos Virales/genética , Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/uso terapéutico , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citotoxicidad Inmunológica/inmunología , Genes MHC Clase II/genética , Glicoproteínas/genética , Glicoproteínas/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Interferón gamma/metabolismo , Interferón gamma/farmacología , Activación de Linfocitos/inmunología , Depleción Linfocítica , Coriomeningitis Linfocítica/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Melanoma Experimental/prevención & control , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Bazo/citología , Bazo/inmunología , Análisis de Supervivencia , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Vacunas de ADN/uso terapéutico , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología
7.
Vaccine ; 37(22): 2952-2959, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-31006497

RESUMEN

CD8+ T cells are known to control infections, but their role in preventing latent infection from establishing has not been thoroughly investigated. We hypothesized that a potent CD8+ T cell response patrolling the mucosal viral entry points could kill the first infected cells and thereby abrogate the infection before latency is established. To investigate this, replication deficient adenovirus serotype 5 vectors encoding murine γ-herpesvirus-68 CD8+ T cell epitopes linkedto the T cell adjuvant Invariant chain, were developed. We show that intranasal vaccination of mice reduces the risk of establishment of latent infection from multiple intranasal ID50 challenges with murine γ-herpesvirus-68 by 81% per exposure at 14 days post vaccination. Protection waned over time, but immune responses were extended by heterologous prime-boost vaccination applied simultaneously intramuscularly and intranasally, and animals vaccinated 66 days prior to challenge showed a strong trend of long-term protection. Our data provides evidence that CD8+ T cells are able to protect against establishment of latent infection. Although the protective efficacy is difficult to maintain over time, this proof-of-concept study suggests a role for a CD8+ T cell arm in future vaccine strategies against latent human viral infections caused by pathogens such as HIV and multiple herpes virus.


Asunto(s)
Adenoviridae/genética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/prevención & control , Herpesviridae/patogenicidad , Animales , Femenino , Citometría de Flujo , Herpesviridae/inmunología , Ratones , Ratones Endogámicos C57BL
8.
Vaccines (Basel) ; 6(1)2018 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-29439476

RESUMEN

Protection against chronic infections has necessitated the development of ever-more potent vaccination tools. HIV seems to be the most challenging foe, with a remarkable, poorly immunogenic and fragile surface glycoprotein and the ability to overpower the cell immune system. Virus-like-particle (VLP) vaccines have emerged as potent inducers of antibody and helper T cell responses, while replication-deficient viral vectors have yielded potent cytotoxic T cell responses. Here, we review the emerging concept of merging these two technologies into virus-like-vaccines (VLVs) for the targeting of HIV. Such vaccines are immunologically perceived as viruses, as they infect cells and produce VLPs in situ, but they only resemble viruses, as the replication defective vectors and VLPs cannot propagate an infection. The inherent safety of such a platform, despite robust particle production, is a distinct advantage over live-attenuated vaccines that must balance safety and immunogenicity. Previous studies have delivered VLVs encoded in modified Vaccinia Ankara vectors and we have developed the concept into a single-reading adenovirus-based technology capable of eliciting robust CD8⁺ and CD4⁺ T cells responses and trimer binding antibody responses. Such vaccines offer the potential to display the naturally produced immunogen directly and induce an integrated humoral and cellular immune response.

9.
Vaccine ; 36(41): 6212-6222, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30190120

RESUMEN

The human adenovirus type 19a/64 (hAd19a) is a rare serotype in the human population that transduces human dendritic cells (DCs) and human muscle cells more efficiently than the well-characterized human adenovirus type 5 (hAd5). To further characterize the potential of this vector as a vaccine we designed replication deficient hAd19a, hAd5 and MVA vectors expressing a papillomavirus (PV) antigen fused to the human MHC class II associated invariant chain T cell adjuvant (hIi) and investigated their immunogenicity in vivo in mice and cynomolgus macaques. We initially showed that the hIi encoded in the hAd5 enhanced PV specific CD8+ T cell responses in mice. The T cell responses induced after hAd19a vaccination was similar to those induced by hAd5 vaccination. The hAd19a induced responses were not reduced in presence of preexisting Ad5 immunity in mice. In macaques both vaccines were equally potent at inducing CD8+ T cells after MVA boost, while the level of CD4+ T cell responses were found to be broader in hAd19a primed animals. These data demonstrate the potential of hAd19a as an alternative vector to hAd5 to elicit potent T cell responses to PV.


Asunto(s)
Adenovirus Humanos/genética , Adenovirus Humanos/inmunología , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Vectores Genéticos , Humanos , Macaca fascicularis , Ratones , Vacunas contra Papillomavirus/genética , Serogrupo , Vacunación/efectos adversos , Vacunación/métodos
10.
Vaccine ; 35(8): 1140-1147, 2017 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-28131394

RESUMEN

The malaria parasite Plasmodium falciparum presents antigens on the infected erythrocyte surface that bind human receptors expressed on the vascular endothelium. The VAR2CSA mediated binding to a distinct chondroitin sulphate A (CSA) is a crucial step in the pathophysiology of placental malaria and the CSA binding region of VAR2CSA has been identified as a promising vaccine target against placental malaria. Here we designed adenovirus encoded virus-like particles (VLP) by co-encoding Simian Immunodeficiency Virus (SIV) gag and VAR2CSA. The VAR2CSA antigen was fused to the transmembrane (TM) and cytoplasmic tail (CT) domains of either the envelope protein of mouse mammary tumour virus (MMTV) or the hemagglutinin (HA) of influenza A. For a non-VLP incorporation control, a third design was made where VAR2CSA was expressed without TM-CT domains. In the primary immunogenicity study in Balb/c mice, VAR2CSA fused to HA TM-CT was significantly superior in inducing ID1-ID2a specific antibodies after the first immunization. A sequential study was performed to include a comparison to the soluble VAR2CSA protein vaccine, which has entered a phase I clinical trial (NCT02647489). The results revealed the induction of higher antibody responses and increased inhibition of parasite binding to CSA using either VAR2CSA HA TM-CT or VAR2CSA MMTV TM-CT as priming vaccines for protein double-boost immunizations, compared to protein prime-double boost regimen. Analysis of pooled serum samples on peptide arrays revealed a unique targeting of several epitopes in mice that had been primed with VAR2CSA HA TM-CT. Consequently, modification of VLP anchors is an important point of optimization in virus-encoded retroviral VLP-based vaccines, and adenovirus VLPs boosted by recombinant proteins offer hope of increasing the levels of protective VAR2CSA specific antibodies.


Asunto(s)
Anticuerpos Antiprotozoarios/biosíntesis , Antígenos de Protozoos/inmunología , Vacunas contra la Malaria/administración & dosificación , Malaria Falciparum/prevención & control , Plasmodium falciparum/inmunología , Vacunas de Partículas Similares a Virus/administración & dosificación , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Antígenos de Protozoos/química , Antígenos de Protozoos/genética , Sulfatos de Condroitina/química , Sulfatos de Condroitina/inmunología , Eritrocitos/inmunología , Eritrocitos/parasitología , Femenino , Productos del Gen gag/química , Productos del Gen gag/genética , Productos del Gen gag/inmunología , Vectores Genéticos/química , Vectores Genéticos/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Inmunización , Vacunas contra la Malaria/biosíntesis , Vacunas contra la Malaria/genética , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Virus del Tumor Mamario del Ratón , Ratones , Ratones Endogámicos BALB C , Placenta/química , Placenta/inmunología , Placenta/parasitología , Plasmodium falciparum/química , Plasmodium falciparum/efectos de los fármacos , Embarazo , Unión Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Virus de la Inmunodeficiencia de los Simios , Vacunas de Partículas Similares a Virus/biosíntesis , Vacunas de Partículas Similares a Virus/genética
11.
J Immunother ; 40(2): 51-61, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28166180

RESUMEN

Currently available prophylactic vaccines have no therapeutic efficacy for preexisting human papillomavirus (HPVs) infections, do not target all oncogenic HPVs and are insufficient to eliminate the burden of HPV induced cancer. We aim to develop an alternative HPV vaccine which is broadly effective and capable of clearing preexisting infection. In an initial attempt to develop a broadly reactive therapeutic vaccine, we designed a putative papillomavirus (PV) ancestor antigen (circulating sequence derived antigenic sequences E1E2-CDSE1E2) based on the conserved E1 and E2 protein sequences from existing oncogenic HPV strains. This antigen was found to be as related to circulating oncogenic Macaca fascicularis papillomaviruses (MfPVs) as to oncogenic HPVs. The CDSE1E2 antigen was fused to a T-cell adjuvant and encoded in chimpanzee 3 and 63 adenoviral vectors. We first showed that the combination of these 2 vaccines induced long-lasting potent CDSE1E2 specific T cell responses in outbred mice. This prime-boost regimen was then tested in female macaques naturally infected with MfPVs. All immunized animals (16/16) responded to the vaccine antigen but with reduced cross-reactivity against existing PVs. Preexisting MfPV infections did not prime vaccine inducible immune responses. Importantly, immunized oncogenic MfPV type 3 (MfPV3) infected animals that responded toward MfPV3 were able to diminish cervical MfPV3 DNA content. Although insufficient breadth was achieved, our results suggest that a relevant level of E1E2 specific T cell immunity is achievable and might be sufficient for the elimination of PV infection. Importantly, naturally infected macaques, offer a relevant model for testing vaccines aimed at eliminating mucosal PV infections.


Asunto(s)
Cuello del Útero/inmunología , Virus Oncogénicos/inmunología , Papillomaviridae/inmunología , Infecciones por Papillomavirus/inmunología , Vacunas contra Papillomavirus/inmunología , Linfocitos T/inmunología , Neoplasias del Cuello Uterino/prevención & control , Animales , Animales no Consanguíneos , Antígenos de Diferenciación de Linfocitos B/genética , Antígenos Virales/genética , Células Cultivadas , Cuello del Útero/virología , ADN Viral/análisis , Modelos Animales de Enfermedad , Femenino , Ingeniería Genética , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Inmunidad Celular , Inmunización Secundaria , Macaca fascicularis , Ratones , Virus Oncogénicos/genética , Pan troglodytes , Papillomaviridae/genética , Infecciones por Papillomavirus/complicaciones , Proteínas Recombinantes de Fusión/genética , Linfocitos T/virología , Neoplasias del Cuello Uterino/etiología , Vacunas de ADN , Proteínas Virales/genética
12.
Vaccine ; 34(18): 2147-56, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27004934

RESUMEN

Previous studies have shown promising results when using an agonistic anti-4-1BB antibody treatment against established tumors. While this is promising, this type of treatment can induce severe side effects. Therefore, we decided to incorporate the membrane form of 4-1BB ligand (4-1BBL) in a replicative deficient adenovirus vaccine expressing the invariant chain (Ii) adjuvant fused to a tumor associated antigen (TAA). The Ii adjuvant increases and prolongs TAA specific CD8+ T cells as previously shown and local expression of 4-1BBL was chosen to avoid the toxicity associated with systemic antibody administration. Furthermore, adenovirus encoded 4-1BBL expression has previously been successfully used to enhance responses toward Plasmodium falciparum and Influenza A antigens. We showed that the incorporation of 4-1BBL in the adenovirus vector led to surface expression of 4-1BBL on antigen presenting cells, but it did not enhance T cell responses in mice towards the Ii linked antigen. In tumor-bearing mice, our vaccine was found to decrease the frequency of TAA specific CD8+ T cells, but this difference did not alter the therapeutic efficacy. In order to reconcile our findings with the previous reports of increased anti-cancer efficacy using systemically delivered 4-1BB agonists, we incorporated a secreted version of 4-1BBL (Fc-4-1BBL) in our vaccine and co-expressed it with the Ii linked to TAA. In tumor bearing mice, this vaccine initially delayed tumor growth and slightly increased survival compared to the vaccine expressing the membrane form of 4-1BBL. Accordingly, secreted 4-1BBL co-encoded with the Ii linked antigen may offer a simplification compared to administration of drug and vaccine separately.


Asunto(s)
Ligando 4-1BB/inmunología , Adenoviridae , Adyuvantes Inmunológicos/administración & dosificación , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Melanoma Experimental/terapia , Animales , Células Presentadoras de Antígenos/inmunología , Antígeno B7-2/metabolismo , Linfocitos T CD8-positivos/inmunología , Células COS , Femenino , Fibroblastos/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
13.
Sci Rep ; 6: 38666, 2016 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-27929135

RESUMEN

Intracellular pathogens represent a serious threat during early life. Importantly, even though the immune system of newborns may be characterized as developmentally immature, with a propensity to develop Th2 immunity, significant CD8+ T-cell responses may still be elicited in the context of optimal priming. Replication deficient adenoviral vectors have been demonstrated to induce potent CD8+ T-cell response in mice, primates and humans. The aim of the present study was therefore to assess whether replication-deficient adenovectors could overcome the risk of overwhelming antigen stimulation during the first period of life and provide a pertinent alternative in infant vaccinology. To address this, infant mice were vaccinated with three different adenoviral vectors and the CD8+ T-cell response after early life vaccination was explored. We assessed the frequency, polyfunctionality and in vivo cytotoxicity of the elicited memory CD8+ T cells, as well as the potential of these cells to respond to secondary infections and confer protection. We further tested the impact of maternal immunity against our replication-deficient adenoviral vector during early life vaccination. Overall, our results indicate that memory CD8+ T cells induced by adenoviral vectors in infant mice are of good quality and match those elicited in the adult host.


Asunto(s)
Adenoviridae/inmunología , Linfocitos T CD8-positivos/inmunología , Vectores Genéticos/inmunología , Memoria Inmunológica , Vacunación , Vacunas/inmunología , Infecciones por Adenoviridae/inmunología , Infecciones por Adenoviridae/prevención & control , Factores de Edad , Animales , Biomarcadores , Linfocitos T CD8-positivos/metabolismo , Femenino , Vectores Genéticos/administración & dosificación , Inmunidad , Inmunofenotipificación , Activación de Linfocitos , Ratones , Fenotipo , Vacunación/métodos , Vacunas/administración & dosificación , Vacunas/genética
14.
Sci Rep ; 6: 20137, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26831578

RESUMEN

The threat from unpredictable influenza virus pandemics necessitates the development of a new type of influenza vaccine. Since the internal proteins are highly conserved, induction of T cells targeting these antigens may provide the solution. Indeed, adenoviral (Ad) vectors expressing flu nucleoprotein have previously been found to induce short-term protection in mice. In this study we confirm that systemic (subcutaneous (s.c.) immunization rapidly induced heterosubtypic protection predominantly mediated by CD8 T cells, but within three months clinical protection completely disappeared. Local (intranasal (i.n.)) immunization elicited delayed, but more lasting protection despite relatively inefficient immunization. However, by far, the most robust protection was induced by simultaneous, combined (i.n. + s.c.) vaccination, and, notably, in this case clinical protection lasted at least 8 months without showing any evidence of fading. Interestingly, the superior ability of the latter group to resist reinfection correlated with a higher number of antigen-specific CD8 T cells in the spleen. Thus, detailed analysis of the underlying CD8 T cell responses highlights the importance of T cells already positioned in the lungs prior to challenge, but at the same time underscores an important back-up role for circulating antigen-specific cells with the capacity to expand and infiltrate the infected lungs.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunidad , Inmunización , Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Adenoviridae/metabolismo , Animales , Antígenos Virales/inmunología , Vías de Administración de Medicamentos , Femenino , Memoria Inmunológica , Vacunas contra la Influenza/inmunología , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/prevención & control , Fenotipo , Especificidad de la Especie , Factores de Tiempo , Vacunación
15.
Vaccine ; 34(44): 5344-5351, 2016 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-27633665

RESUMEN

The low number of envelope (Env) spikes presented on native HIV-1 particles is a major impediment for HIV-1 prophylactic vaccine development. We designed virus-like particle encoding adenoviral vectors utilizing SIVmac239 Gag as an anchor for full length and truncated HIV-1 M consensus Env. Truncated Env overexpressed VRC01 and 17b binding antigen on the surface of transduced cells while the full length Env vaccine presented more and similar amounts of antigen binding to the trimer conformation sensitive antibodies PGT151 and PGT145, respectively. The adenoviral vectors were used to prime Balb/c mice followed by sequential boosting with chimpanzee type 63, and chimpanzee type 3 adenoviral vectors encoding SIVmac239 Gag and full length consensus Env. Both vaccine regimens induced increasing titers of binding antibody responses after each immunization, and significant differences in immune responses between the two groups were observed after the final immunization. Full length Env priming skewed antibody responses towards gp41, while truncated Env priming induced responses primarily targeting gp120 containing and derived antigens. Importantly, no differences in neutralizing antibody responses were found between the different priming regimens as both induced high titered tier 1 neutralizing antibodies, but no tier 2 antibodies, possibly reflecting the similar presentation of trimer specific antibody epitopes. The described vaccine regimens provide insight into the effects of the HIV-1 Env cytoplasmic tail on epitope presentation and subsequent immune responses, which is relevant for the interpretation of current clinical trials that are using truncated Env as an immunogen. The regimens described here provide similar neutralization titers, and thus are useful for investigating the importance of specificity in non-neutralizing antibody mediated protection against viral challenge.


Asunto(s)
Vacunas contra el SIDA/inmunología , Adenoviridae/genética , Anticuerpos Anti-VIH/sangre , Proteína gp120 de Envoltorio del VIH/inmunología , VIH-1/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/genética , Adenoviridae/inmunología , Adenovirus Humanos/genética , Adenovirus Humanos/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Epítopos/inmunología , Genes gag , Vectores Genéticos , Anticuerpos Anti-VIH/inmunología , Inmunidad Humoral , Inmunogenicidad Vacunal , Ratones Endogámicos BALB C , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/inmunología
16.
Mol Cancer Res ; 14(12): 1288-1299, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27655130

RESUMEN

Many tumors express proteoglycans modified with oncofetal chondroitin sulfate glycosaminoglycan chains (ofCS), which are normally restricted to the placenta. However, the role of ofCS in cancer is largely unknown. The function of ofCS in cancer was analyzed using the recombinant ofCS-binding VAR2CSA protein (rVAR2) derived from the malaria parasite, Plasmodium falciparum We demonstrate that ofCS plays a key role in tumor cell motility by affecting canonical integrin signaling pathways. Binding of rVAR2 to tumor cells inhibited the interaction of cells with extracellular matrix (ECM) components, which correlated with decreased phosphorylation of Src kinase. Moreover, rVAR2 binding decreased migration, invasion, and anchorage-independent growth of tumor cells in vitro Mass spectrometry of ofCS-modified proteoglycan complexes affinity purified from tumor cell lines on rVAR2 columns revealed an overrepresentation of proteins involved in cell motility and integrin signaling, such as integrin-ß1 (ITGB1) and integrin-α4 (ITGA4). Saturating concentrations of rVAR2 inhibited downstream integrin signaling, which was mimicked by knockdown of the core chondroitin sulfate synthesis enzymes ß-1,3-glucuronyltransferase 1 (B3GAT1) and chondroitin sulfate N-acetylgalactosaminyltransferase 1 (CSGALNACT1). The ofCS modification was highly expressed in both human and murine metastatic lesions in situ and preincubation or early intravenous treatment of tumor cells with rVAR2 inhibited seeding and spreading of tumor cells in mice. This was associated with a significant increase in survival of the animals. These data functionally link ofCS modifications with cancer cell motility and further highlights ofCS as a novel therapeutic cancer target. IMPLICATIONS: The cancer-specific expression of ofCS aids in metastatic phenotypes and is a candidate target for therapy. Mol Cancer Res; 14(12); 1288-99. ©2016 AACR.


Asunto(s)
Antígenos de Protozoos/genética , Sulfatos de Condroitina/metabolismo , Integrinas/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Animales , Antígenos de Protozoos/metabolismo , Carcinoma Pulmonar de Lewis/metabolismo , Línea Celular Tumoral , Sulfatos de Condroitina/genética , Humanos , Melanoma Experimental/metabolismo , Ratones , Metástasis de la Neoplasia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Transducción de Señal
17.
PLoS One ; 10(2): e0117242, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25679375

RESUMEN

In this study, we compared adenoviral vaccine vectors with the capacity to induce equally potent immune responses against non-dominant and immunodominant epitopes of murine lymphocytic choriomeningitis virus (LCMV). Our results demonstrate that vaccination targeting non-dominant epitopes facilitates potent virus-induced T-cell responses against immunodominant epitopes during subsequent challenge with highly invasive virus. In contrast, when an immunodominant epitope was included in the vaccine, the T-cell response associated with viral challenge remained focussed on that epitope. Early after challenge with live virus, the CD8+ T cells specific for vaccine-encoded epitopes, displayed a phenotype typically associated with prolonged/persistent antigenic stimulation marked by high levels of KLRG-1, as compared to T cells reacting to epitopes not included in the vaccine. Notably, this association was lost over time in T cells specific for the dominant T cell epitopes, and these cells were fully capable of expanding in response to a new viral challenge. Overall, our data suggests a potential for broadening of the antiviral CD8+ T-cell response by selecting non-dominant antigens to be targeted by vaccination. In addition, our findings suggest that prior adenoviral vaccination is not likely to negatively impact the long-term and protective immune response induced and maintained by a vaccine-attenuated chronic viral infection.


Asunto(s)
Antígenos Virales/inmunología , Epítopos de Linfocito T/inmunología , Linfocitos T/inmunología , Vacunas Virales/inmunología , Virosis/inmunología , Adenoviridae/genética , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Enfermedad Crónica , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Epítopos Inmunodominantes/inmunología , Memoria Inmunológica , Inmunofenotipificación , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Vacunación , Proteínas Virales/inmunología , Virosis/metabolismo , Virosis/prevención & control
18.
Cancer Cell ; 28(4): 500-514, 2015 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-26461094

RESUMEN

Plasmodium falciparum engineer infected erythrocytes to present the malarial protein, VAR2CSA, which binds a distinct type chondroitin sulfate (CS) exclusively expressed in the placenta. Here, we show that the same CS modification is present on a high proportion of malignant cells and that it can be specifically targeted by recombinant VAR2CSA (rVAR2). In tumors, placental-like CS chains are linked to a limited repertoire of cancer-associated proteoglycans including CD44 and CSPG4. The rVAR2 protein localizes to tumors in vivo and rVAR2 fused to diphtheria toxin or conjugated to hemiasterlin compounds strongly inhibits in vivo tumor cell growth and metastasis. Our data demonstrate how an evolutionarily refined parasite-derived protein can be exploited to target a common, but complex, malignancy-associated glycosaminoglycan modification.


Asunto(s)
Antígenos de Protozoos/genética , Sulfatos de Condroitina/metabolismo , Melanoma Experimental/terapia , Placenta/metabolismo , Proteínas Recombinantes/administración & dosificación , Neoplasias Cutáneas/terapia , Animales , Antígenos de Protozoos/metabolismo , Línea Celular Tumoral , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Femenino , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Receptores de Hialuranos/metabolismo , Melanoma Experimental/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Terapia Molecular Dirigida , Oligopéptidos/genética , Oligopéptidos/metabolismo , Especificidad de Órganos , Embarazo , Proteínas Recombinantes/farmacología , Neoplasias Cutáneas/metabolismo
19.
Microbes Infect ; 5(2): 179-87, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12650776

RESUMEN

Several viruses encode chemokine elements in their genome. This review focuses on the roles of such elements in the ongoing battle between the virus and the host. The biological and pharmacological characterizations of several of these chemokine elements have highlighted their importance in the mammalian immune system for antiviral responses and suggested future antiviral and anti-inflammatory therapeutic strategies.


Asunto(s)
Quimiocinas , Imitación Molecular , Virus/inmunología , Virus/patogenicidad , Animales , Quimiocinas/genética , Quimiocinas/inmunología , Humanos , Receptores de Quimiocina/metabolismo
20.
Contrib Microbiol ; 10: 232-52, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12530329

RESUMEN

Large DNA viruses such as pox- and in particular herpesviruses are notorious in their ability to evade the immune system and to be maintained in the general population. Based on the accumulated knowledge reviewed in this study it is evident that important mechanisms of these actions are the acquisition and modification of host-encoded chemokines and chemokine receptors. The described viral molecules leave nothing to chance and have thoroughly and efficiently corrupted the host immune system. Through this process viruses have identified key molecules in antiviral responses by their inhibition of these or potent ways to alter an efficient antiviral response to a weak Th2-driven response. Examples here are the chemokine scavenging by US28, attractance of Th2 cells and regulatory cells by vMIP1-3 and the selective engaging of CCR8 by MC148. Important insights into viral pathology and possible targets for antiviral therapies have been provided by UL33, UL78 and in particular ORF74 and the chances are that many more will follow. In HHV8 vMIP-2 and the chemokine-binding proteins potent anti-inflammatory agents have been provided. These have already had their potential demonstrated in animal models and may in their native or modified forms represent useful therapies in humans.


Asunto(s)
Quimiocinas/farmacología , Infecciones por Poxviridae/inmunología , Receptores de Quimiocina/metabolismo , Receptores Virales/metabolismo , Proteínas Virales/farmacología , Animales , Virus ADN/inmunología , Virus ADN/metabolismo , Herpesviridae/metabolismo , Herpesviridae/patogenicidad , Humanos , Mamíferos , Receptores Virales/clasificación , Receptores Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA