Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cancer Lett ; 561: 216139, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37001752

ABSTRACT

Despite the success of immune checkpoint blockade for cancer therapy, many patients do not respond adequately. We aimed to improve this therapy by optimizing both the antibodies and their delivery route, using small monodomain antibodies (nanobodies) delivered locally with a self-amplifying RNA (saRNA) vector based on Semliki Forest virus (SFV). We generated nanobodies against PD-1 and PD-L1 able to inhibit both human and mouse interactions. Incorporation of a dimerization domain reduced PD-1/PD-L1 IC50 by 8- and 40-fold for anti-PD-L1 and anti-PD-1 nanobodies, respectively. SFV viral particles expressing dimeric nanobodies showed a potent antitumor response in the MC38 model, resulting in >50% complete regressions, and showed better therapeutic efficacy compared to vectors expressing conventional antibodies. These effects were also observed in the B16 melanoma model. Although a short-term expression of nanobodies was observed due to the cytopathic nature of the saRNA vector, it was enough to generate a strong proinflammatory response in tumors, increasing infiltration of NK and CD8+ T cells. Delivery of the SFV vector expressing dimeric nanobodies by local plasmid electroporation, which could be more easily translated to the clinic, also showed a potent antitumor effect.


Subject(s)
Neoplasms , Single-Domain Antibodies , Animals , Humans , Mice , B7-H1 Antigen/genetics , CD8-Positive T-Lymphocytes , Semliki forest virus/genetics , Single-Domain Antibodies/genetics , Programmed Cell Death 1 Receptor/metabolism
2.
J Immunother Cancer ; 10(8)2022 08.
Article in English | MEDLINE | ID: mdl-35918123

ABSTRACT

BACKGROUND: One of the main difficulties of adoptive cell therapies with chimeric antigen receptor (CAR)-T cells in solid tumors is the identification of specific target antigens. The tumor microenvironment can present suitable antigens for CAR design, even though they are not expressed by the tumor cells. We have generated a CAR specific for the splice variant extra domain A (EDA) of fibronectin, which is highly expressed in the tumor stroma of many types of tumors but not in healthy tissues. METHODS: EDA expression was explored in RNA-seq data from different human tumor types and by immunohistochemistry in paraffin-embedded tumor biopsies. Murine and human anti-EDA CAR-T cells were prepared using recombinant retro/lentiviruses, respectively. The functionality of EDA CAR-T cells was measured in vitro in response to antigen stimulation. The antitumor activity of EDA CAR-T cells was measured in vivo in C57BL/6 mice challenged with PM299L-EDA hepatocarcinoma cell line, in 129Sv mice-bearing F9 teratocarcinoma and in NSG mice injected with the human hepatocarcinoma cell line PLC. RESULTS: EDA CAR-T cells recognized and killed EDA-expressing tumor cell lines in vitro and rejected EDA-expressing tumors in immunocompetent mice. Notably, EDA CAR-T cells showed an antitumor effect in mice injected with EDA-negative tumor cells lines when the tumor stroma or the basement membrane of tumor endothelial cells express EDA. Thus, EDA CAR-T administration delayed tumor growth in immunocompetent 129Sv mice challenged with teratocarcinoma cell line F9. EDA CAR-T treatment exerted an antiangiogenic effect and significantly reduced gene signatures associated with epithelial-mesenchymal transition, collagen synthesis, extracellular matrix organization as well as IL-6-STAT5 and KRAS pathways. Importantly, the human version of EDA CAR, that includes the human 41BB and CD3ζ endodomains, exerted strong antitumor activity in NSG mice challenged with the human hepatocarcinoma cell line PLC, which expresses EDA in the tumor stroma and the endothelial vasculature. EDA CAR-T cells exhibited a tropism for EDA-expressing tumor tissue and no toxicity was observed in tumor bearing or in healthy mice. CONCLUSIONS: These results suggest that targeting the tumor-specific fibronectin splice variant EDA with CAR-T cells is feasible and offers a therapeutic option that is applicable to different types of cancer.


Subject(s)
Receptors, Chimeric Antigen , Teratocarcinoma , Animals , Endothelial Cells , Fibronectins , Humans , Mice , Mice, Inbred C57BL , T-Lymphocytes , Teratocarcinoma/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
3.
Oncoimmunology ; 11(1): 2070337, 2022.
Article in English | MEDLINE | ID: mdl-35529677

ABSTRACT

The high metabolic activity and insufficient perfusion of tumors leads to the acidification of the tumor microenvironment (TME) that may inhibit the antitumor T cell activity. We found that pharmacological inhibition of the acid loader chloride/bicarbonate anion exchanger 2 (Ae2), with 4,4'-diisothiocyanatostilbene-2,2'-disulfonicacid (DIDS) enhancedCD4+ andCD8+ T cell function upon TCR activation in vitro, especially under low pH conditions. In vivo, DIDS administration delayed B16OVA tumor growth in immunocompetent mice as monotherapy or when combined with adoptive T cell transfer of OVA-specificT cells. Notably, genetic Ae2 silencing in OVA-specificT cells improvedCD4+/CD8+ T cell function in vitro as well as their antitumor activity in vivo. Similarly, genetic modification of OVA-specificT cells to overexpress Hvcn1, a selectiveH+ outward current mediator that prevents cell acidification, significantly improved T cell function in vitro, even at low pH conditions. The adoptive transfer of OVA-specificT cells overexpressing Hvcn1 exerted a better antitumor activity in B16OVA tumor-bearingmice. Hvcn1 overexpression also improved the antitumor activity of CAR T cells specific for Glypican 3 (GPC3) in mice bearing PM299L-GPC3tumors. Our results suggest that preventing intracellular acidification by regulating the expression of acidifier ion channels such as Ae2 or alkalinizer channels like Hvcn1 in tumor-specificlymphocytes enhances their antitumor response by making them more resistant to the acidic TME.


Subject(s)
CD8-Positive T-Lymphocytes , Immunotherapy, Adoptive , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Hydrogen-Ion Concentration , Immunotherapy, Adoptive/methods , Mice
4.
Cancer Lett ; 528: 45-58, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34973390

ABSTRACT

Adoptive cell transfer therapy using CD8+ T lymphocytes showed promising results eradicating metastatic malignancies. However, several regulatory mechanisms limit its efficacy. We studied the role of the expression of the transcription factor FOXP3 on CD8+ T cell function and anti-tumor immunity. Here we show that suboptimal T cell receptor stimulation of CD8+ T cells upregulates FOXP3 in vitro. Similarly, CD8 T cells transferred into tumor-bearing mice upregulate FOXP3 in vivo. Cell-intrinsic loss of FOXP3 by CD8+ T cells resulted in improved functionality after TCR stimulation and better antitumor responses in vivo. Inhibition of the FOXP3/NFAT interaction likewise improved CD8+ T cell functionality. Transcriptomic analysis of cells after TCR stimulation revealed an enrichment of genes implicated in the response to IFN-γ, IFN-α, inflammatory response, IL-6/JAK/STAT, G2M checkpoint and IL-2/STAT signaling in FOXP3-deficient CD8+ T cells with respect to FOXP3-wt CD8+ T cells. Our results suggest that transient expression of FOXP3 by CD8+ T cells in the tumor microenvironment restrains their anti-tumor activity, with clear implications for improving T cell responses during immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/metabolism , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/immunology , Animals , Humans , Mice
5.
Biomedicines ; 9(2)2021 Feb 17.
Article in English | MEDLINE | ID: mdl-33671179

ABSTRACT

(1) Background: The ability of cancer cells to evade the immune system is due in part to their capacity to induce and recruit T regulatory cells (Tregs) to the tumor microenvironment. Strategies proposed to improve antitumor immunity by depleting Tregs generally lack specificity and raise the possibility of autoimmunity. Therefore, we propose to control Tregs by their functional inactivation rather than depletion. Tregs are characterized by the expression of the Forkhead box protein 3 (FOXP3) transcription factor, which is considered their "master regulator". Its interaction with DNA is assisted primarily by its interaction with other proteins in the so-called "Foxp3 interactome", which elicits much of the characteristic Treg cell transcriptional signature. We speculated that the disruption of such a protein complex by using synthetic peptides able to bind Foxp3 might have an impact on the functionality of Treg cells and thus have a therapeutic potential in cancer treatment. (2) Methods: By using a phage-displayed peptide library, or short synthetic peptides encompassing Foxp3 fragments, or by studying the crystal structure of the Foxp3:NFAT complex, we have identified a series of peptides that are able to bind Foxp3 and inhibit Treg activity. (3) Results: We identified some peptides encompassing fragments of the leuzin zipper or the C terminal domain of Foxp3 with the capacity to inhibit Treg activity in vitro. The acetylation/amidation of linear peptides, head-to-tail cyclization, the incorporation of non-natural aminoacids, or the incorporation of cell-penetrating peptide motifs increased in some cases the Foxp3 binding capacity and Treg inhibitory activity of the identified peptides. Some of them have shown antitumoral activity in vivo. (4) Conclusions: Synthetic peptides constitute an alternative to inhibit Foxp3 protein-protein interactions intracellularly and impair Treg immunosuppressive activity. These peptides might be considered as potential hit compounds on the design of new immunotherapeutic approaches against cancer.

6.
Biomed Res Int ; 2021: 8852233, 2021.
Article in English | MEDLINE | ID: mdl-33575350

ABSTRACT

BACKGROUND/AIM: Irreversible electroporation (IRE) showed promising results for small-size tumors and very early cancers. However, further development is needed to evolve this procedure into a more efficient ablation technique for long-term control of tumor growth. In this work, we show that it is possible to increase the antitumor efficiency of IRE by simmultaneously injecting c-di-GMP, a STING agonist, intratumorally. MATERIALS AND METHODS: Intratumoral administration of c-di-GMP simultaneously to IRE was evaluated in murine models of melanona (B16.OVA) and hepatocellular carcinoma (PM299L). RESULTS: The combined therapy increased the number of tumor-infiltrating IFN-γ/TNF-α-producing CD4 and CD8 T cells and delayed tumor growth, as compared to the effect observed in groups treated with c-di-GMP or IRE alone. CONCLUSION: These results can lead to the development of a new therapeutic strategy for the treatment of cancer patients refractory to other therapies.


Subject(s)
Ablation Techniques/methods , Carcinoma, Hepatocellular/therapy , Cyclic GMP/analogs & derivatives , Electroporation/methods , Liver Neoplasms/therapy , Membrane Proteins/agonists , Animals , Cell Line , Combined Modality Therapy/methods , Cyclic GMP/administration & dosage , Female , Liver Neoplasms, Experimental/therapy , Mice, Inbred C57BL
7.
Mol Ther ; 27(11): 1892-1905, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31563534

ABSTRACT

Immune checkpoint blockade has shown anti-cancer efficacy, but requires systemic administration of monoclonal antibodies (mAbs), often leading to adverse effects. To avoid toxicity, mAbs could be expressed locally in tumors. We developed adeno-associated virus (AAV) and Semliki Forest virus (SFV) vectors expressing anti-programmed death ligand 1 (aPDL1) mAb. When injected intratumorally in MC38 tumors, both viral vectors led to similar local mAb expression at 24 h, diminishing quickly in SFV-aPDL1-treated tumors. However, SFV-aPDL1 induced >40% complete regressions and was superior to AAV-aPDL1, as well as to aPDL1 mAb given systemically or locally. SFV-aPDL1 induced abscopal effects and was also efficacious against B16-ovalbumin (OVA). The higher SFV-aPDL1 antitumor activity could be related to local upregulation of interferon-stimulated genes because of SFV RNA replication. This was confirmed by combining local SFV-LacZ administration and systemic aPDL1 mAb, which provided higher antitumor effects than each separated agent. SFV-aPDL1 promoted tumor-specific CD8 T cells infiltration in both tumor models. In MC38, SFV-aPDL1 upregulated co-stimulatory markers (CD137/OX40) in tumor CD8 T cells, and its combination with anti-CD137 mAb showed more pronounced antitumor effects than each single agent. These results indicate that local transient expression of immunomodulatory mAbs using non-propagative RNA vectors inducing type I interferon (IFN-I) responses represents a potent and safe approach for cancer treatment.


Subject(s)
Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Gene Expression , Genetic Vectors/genetics , Neoplasms/genetics , Neoplasms/immunology , RNA Viruses/genetics , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cell Line , Dependovirus/genetics , Disease Models, Animal , Female , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Humans , Immunomodulation/drug effects , Immunophenotyping , Injections, Intralesional , Mice , Neoplasms/pathology , Neoplasms/therapy , Recombinant Fusion Proteins/genetics , Semliki forest virus/genetics , Survival Rate , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Burden
8.
J Vasc Interv Radiol ; 30(7): 1098-1105, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31101416

ABSTRACT

PURPOSE: To evaluate the therapeutic efficacy of irreversible electroporation (IRE) combined with the intratumoral injection of the immunogenic adjuvant poly-ICLC (polyinosinic-polycytidylic acid and poly-L-lysine, a dsRNA analog mimicking viral RNA) inmediately before IRE. MATERIALS AND METHODS: Mice and rabbits bearing hepatocellular carcinoma tumors (Hepa.129 and VX2 tumor models, respectively) were treated with IRE (2 pulses of 2500V), with poly-ICLC, or with IRE + poly-ICLC combination therapy. Tumor growth in mice was monitored using a digital caliper and by computed tomography in rabbits. RESULTS: Intratumoral administration of poly-ICLC immediately before IRE elicited shrinkage of Hepa.129 cell-derived tumors in 70% of mice, compared to 30% and 26% by poly-ICLC or IRE alone, respectively (P = .0004). This combined therapy induced the shrinkage of VX-2-based hepatocellular carcinoma tumors in 40% of rabbits, whereas no response was achieved by either individual treatment (P = .045). The combined therapy activated a systemic antitumor response able to inhibit the growth of other untreated tumors. CONCLUSIONS: IRE treatment, immediately preceded by the intratumoral administration of an immunogenic adjuvant such as poly-ICLC, might enhance the antitumor effect of the IRE procedure. This combination might facilitate the induction of a long-term systemic response to prevent tumor relapses and the appearance of metastases.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Carboxymethylcellulose Sodium/analogs & derivatives , Carcinoma, Hepatocellular/therapy , Electroporation/methods , Liver Neoplasms, Experimental/therapy , Poly I-C/administration & dosage , Polylysine/analogs & derivatives , Animals , Carboxymethylcellulose Sodium/administration & dosage , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Injections, Intralesional , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/pathology , Mice, Inbred C3H , Polylysine/administration & dosage , Rabbits , Tumor Burden
9.
Front Immunol ; 10: 2990, 2019.
Article in English | MEDLINE | ID: mdl-31921216

ABSTRACT

Adoptive immunotherapy with ex vivo-expanded tumor-infiltrating lymphocytes (TILs) has achieved objective clinical responses in a significant number of patients with cancer. The failure of many patients to develop long-term tumor control may be, in part, due to exhaustion of transferred T cells in the presence of a hostile tumor microenvironment. In several tumor types, growth and survival of carcinoma cells appear to be sustained by a network of receptors/ligands of the ErbB family. We speculated that if transferred T cells could benefit from EGFR ligands produced by the tumor, they might proliferate better and exert their anti-tumor activities more efficiently. We found that CD8+ T cells transduced with a retrovirus to express EGFR responded to EGFR ligands activating the EGFR signaling pathway. These EGFR-expressing effector T cells proliferated better and produced more IFN-γ and TNF-α in the presence of EGFR ligands produced by tumor cells in vitro. EGFR-expressing CD8 T cells from OT-1 mice were more efficient killing B16-OVA cells than control OT-1 CD8 T cells. Importantly, EGFR-expressing OT-1 T cells injected into B16-OVA tumor bearing mice were recruited into the tumor, expressed lower levels of the exhaustion markers PD1, TIGIT, and LAG3, and were more efficient in delaying tumor growth. Our results suggest that genetic modification of CD8+ T cells to express EGFR might be considered in immunotherapeutic strategies based on adoptive transfer of anti-tumor T cells against cancers expressing EGFR ligands.


Subject(s)
Adoptive Transfer , CD8-Positive T-Lymphocytes , ErbB Receptors , Genetic Vectors , Neoplasms , Retroviridae , Transduction, Genetic , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/transplantation , Cell Line, Tumor , ErbB Receptors/genetics , ErbB Receptors/immunology , Female , Mice , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy
10.
Front Immunol ; 9: 68, 2018.
Article in English | MEDLINE | ID: mdl-29422905

ABSTRACT

A complex network of interactions exists between the immune, the olfactory, and the central nervous system (CNS). Inhalation of different fragrances can affect immunological reactions in response to an antigen but also may have effects on the CNS and cognitive activity. We performed an exploratory study of the immunomodulatory ability of a series of compounds representing each of the 10 odor categories or clusters described previously. We evaluated the impact of each particular odor on the immune response after immunization with the model antigen ovalbumin in combination with the TLR3 agonist poly I:C. We found that some odors behave as immunostimulatory agents, whereas others might be considered as potential immunosuppressant odors. Interestingly, the immunomodulatory capacity was, in some cases, strain-specific. In particular, one of the fragrances, carvone, was found to be immunostimulatory in BALB/c mice and immunosuppressive in C57BL/6J mice, facilitating or impairing viral clearance, respectively, in a model of a viral infection with a recombinant adenovirus. Importantly, inhalation of the odor improved the memory capacity in BALB/c mice in a fear-conditioning test, while it impaired this same capacity in C57BL/6J mice. The improvement in memory capacity in BALB/c was associated with higher CD3+ T cell infiltration into the hippocampus and increased local expression of mRNA coding for IL-1ß, TNF-α, and IL-6 cytokines. In contrast, the memory impairment in C57BL/6 was associated with a reduction in CD3 numbers and an increase in IFN-γ. These data suggest an association between the immunomodulatory capacity of smells and their impact on the cognitive functions of the animals. These results highlight the potential of studying odors as therapeutic agents for CNS-related diseases.


Subject(s)
Fear/psychology , Immunologic Factors/pharmacology , Immunomodulation/drug effects , Memory/drug effects , Monoterpenes/pharmacology , Administration, Inhalation , Animals , Cognition , Conditioning, Psychological , Cyclohexane Monoterpenes , Disease Models, Animal , Disease Susceptibility , Female , Gene Expression Profiling , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Immunologic Factors/administration & dosage , Immunomodulation/genetics , Leukocytes/drug effects , Leukocytes/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monoterpenes/administration & dosage , Odorants , Virus Diseases/etiology
11.
Oncotarget ; 8(42): 71709-71724, 2017 Sep 22.
Article in English | MEDLINE | ID: mdl-29069740

ABSTRACT

Although T regulatory cells (Treg) are essential for the prevention of autoimmune diseases, their immunoregulatory function restrains the induction of immune responses against cancer. Thus, development of inhibitors of FOXP3, a key transcription factor for the immunosuppressive activity of Treg, might give new therapeutic opportunities. In a previous work we identified a peptide (named P60) able to enter into the cells, bind to FOXP3, and impair Treg activity in vitro and in vivo. Here we show that P60 binds to the intermediate region of FOXP3 and inhibits its homodimerization as well as its interaction with the transcription factor AML1. Alanine-scanning of P60 revealed the relevance of each position on FOXP3 binding, homodimerization, association with AML1 and inhibition of Treg activity. Introduction of alanine at positions 2, 5 and 11 improved the activity of the original P60, whereas alanine mutations at positions 1, 7, 8, 9, 10 and 12 were detrimental. Multiple mutation experiments allowed us to identify peptides with higher FOXP3 binding affinity and stronger biological activity than the original P60. Head to tail macrocyclization of peptide P60-D2A-S5A improved Treg inhibition and enhanced anti-tumor activity of anti-PD1 antibodies in a model of hepatocellular carcinoma. Introduction of a D-aminoacid at position 2 augmented significantly microsomal stability while maintained FOXP3 binding capacity and Treg inhibition in vitro. In vivo, when combined with the cytotoxic T-cell epitope AH1, it induced protection against CT26 tumor implantation. This study provides important structure-function relationships essential for further drug design to inhibit Treg cells in cancer.

12.
J Immunol ; 195(7): 3180-9, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26324768

ABSTRACT

Regulatory T cell (Treg) activity is modulated by a cooperative complex between the transcription factor NFAT and FOXP3, a lineage specification factor for Tregs. FOXP3/NFAT interaction is required to repress expression of IL-2, upregulate expression of the Treg markers CTLA4 and CD25, and confer suppressor function to Tregs. However, FOXP3 is expressed transiently in conventional CD4(+) T cells upon TCR stimulation and may lead to T cell hyporesponsiveness. We found that a short synthetic peptide able to inhibit FOXP3/NFAT interaction impaired suppressor activity of conventional Tregs in vitro. Specific inhibition of FOXP3/NFAT interaction with this inhibitory peptide revealed that FOXP3 downregulates NFAT-driven promoter activity of CD40L and IL-17. Inhibition of FOXP3/NFAT interaction upregulated CD40L expression on effector T cells and enhanced T cell proliferation and IL-2, IFN-γ, IL-6, or IL-17 production in response to TCR stimulation. The inhibitory peptide impaired effector T cell conversion into induced Tregs in the presence of TGF-ß. Moreover, in vivo peptide administration showed antitumor efficacy in mice bearing Hepa129 or TC1 tumor cells when combined with sorafenib or with an antitumor vaccine, respectively. Our results suggest that inhibition of NFAT/FOXP3 interaction might improve antitumor immunotherapies.


Subject(s)
CD40 Ligand/biosynthesis , Forkhead Transcription Factors/metabolism , Interleukin-17/biosynthesis , NFATC Transcription Factors/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antineoplastic Agents/pharmacology , CD40 Ligand/genetics , CTLA-4 Antigen/biosynthesis , Cell Proliferation/genetics , Female , Forkhead Transcription Factors/antagonists & inhibitors , Humans , Immunotherapy , Interferon-gamma/biosynthesis , Interleukin-17/genetics , Interleukin-2/biosynthesis , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-6/biosynthesis , Jurkat Cells , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , NFATC Transcription Factors/antagonists & inhibitors , Neoplasms/therapy , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Ovalbumin/immunology , Peptide Fragments/pharmacology , Phenylurea Compounds/pharmacology , Promoter Regions, Genetic/genetics , Sorafenib , Transforming Growth Factor beta/metabolism
13.
Biomed Res Int ; 2013: 864720, 2013.
Article in English | MEDLINE | ID: mdl-24093105

ABSTRACT

The development of tools for efficient targeting of antigens to antigen presenting cells is of great importance for vaccine development. We have previously shown that fusion proteins containing antigens fused to the extra domain A from fibronectin (EDA), an endogenous TLR4 ligand, which targets antigens to TLR4-expressing dendritic cells (DC), are highly immunogenic. To facilitate the procedure of joining EDA to any antigen of choice, we have prepared the fusion protein EDAvidin by linking EDA to the N terminus of streptavidin, allowing its conjugation with biotinylated antigens. We found that EDAvidin, as streptavidin, forms tetramers and binds biotin or biotinylated proteins with a Kd ~ 2.6 × 10(-14) mol/L. EDAvidin favours the uptake of biotinylated green fluorescent protein by DC. Moreover, EDAvidin retains the proinflammatory properties of EDA, inducing NF- κß by TLR4-expressing cells, as well as the production of TNF- α by the human monocyte cell line THP1 and IL-12 by DC. More importantly, immunization of mice with EDAvidin conjugated with the biotinylated nonstructural NS3 protein from hepatitis C virus induces a strong anti-NS3 T cell immune response. These results open a new way to use the EDA-based delivery tool to target any antigen of choice to DC for vaccination against infectious diseases and cancer.


Subject(s)
Antigen-Presenting Cells/immunology , Fibronectins/immunology , Recombinant Fusion Proteins/immunology , Toll-Like Receptor 4/immunology , Animals , Antigen-Presenting Cells/metabolism , Cell Line , Dendritic Cells/immunology , Dendritic Cells/metabolism , Fibronectins/chemistry , Fibronectins/genetics , Hepacivirus/immunology , Humans , Immunotherapy, Active , Ligands , Mice , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Streptavidin/chemistry , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Toll-Like Receptor 4/metabolism
14.
J Hepatol ; 51(3): 520-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19596480

ABSTRACT

BACKGROUND/AIMS: Vaccination strategies able to induce strong T-cell responses might contribute to eradicate hepatitis C virus (HCV) infection. We previously demonstrated that fusion of an antigen to the extra domain A from fibronectin (EDA) targets the antigen to TLR4-expressing dendritic cells (DC) and improves its immunogenicity. Here, we studied if fusion of EDA with the non-structural HCV protein NS3 might constitute an effective immunogen against HCV. METHODS: Recombinant NS3 and the fusion protein EDA-NS3 were produced and purified from E. coli, and tested in vitro for their capacity to activate maturation of DC and to favour antigen presentation. HHD transgenic mice expressing the human HLA-A2 molecule were immunized with recombinant proteins in the absence or presence of poly(I:C) and anti-CD40 agonistic antibodies and responses elicited by vaccination were tested in vitro, and in vivo, by their capacity to downregulate intrahepatic expression of HCV-NS3 RNA. RESULTS: EDA-NS3, but not NS3 alone, upregulated the expression of maturation markers, as well as Delta-like 1 and Delta-like 4 Notch ligands in DC and induced the production of IL-12. Mice immunized with EDA-NS3 had strong and long lasting NS3-specific CD4+ and CD8+ T-cell responses and, in combination with poly(I:C) and anti-CD40, downregulated intrahepatic expression of HCV-NS3 RNA. CONCLUSIONS: Recombinant EDA-NS3 may be considered for the development of vaccines against HCV infection.


Subject(s)
Fibronectins/therapeutic use , Hepacivirus/immunology , Hepatitis C/prevention & control , Recombinant Proteins/therapeutic use , Viral Fusion Proteins/therapeutic use , Viral Nonstructural Proteins/therapeutic use , Viral Vaccines/therapeutic use , Animals , Antiviral Agents/therapeutic use , Cell Line , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Disease Models, Animal , Drug Therapy, Combination , Escherichia coli/metabolism , Female , Fibronectins/biosynthesis , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatitis C/immunology , Hepatitis C/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Poly I-C/therapeutic use , Protein Structure, Tertiary , RNA, Viral/metabolism , Recombinant Proteins/biosynthesis , Viral Fusion Proteins/biosynthesis , Viral Nonstructural Proteins/biosynthesis , Viral Nonstructural Proteins/genetics , Viral Vaccines/biosynthesis
15.
J Immunol ; 178(2): 748-56, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17202335

ABSTRACT

Vaccination strategies based on the in vivo targeting of Ags to dendritic cells (DCs) are needed to improve the induction of specific T cell immunity against tumors and infectious agents. In this study, we have used a recombinant protein encompassing the extra domain A from fibronectin (EDA), an endogenous ligand for TLR4, to deliver Ags to TLR4-expressing DC. The purified EDA protein was shown to bind to TLR4-expressing HEK293 cells and to activate the TLR4 signaling pathway. EDA also stimulated the production by DC of proinflammatory cytokines such as IL-12 or TNF-alpha and induced their maturation in vitro and in vivo. A fusion protein between EDA and a cytotoxic T cell epitope from OVA efficiently presented this epitope to specific T cells and induced the in vivo activation of a strong and specific CTL response. Moreover, a fusion protein containing EDA and the full OVA also improved OVA presentation by DC and induced CTL responses in vivo. These EDA recombinant proteins protected mice from a challenge with tumor cells expressing OVA. These results strongly suggest that the fibronectin extra domain A may serve as a suitable Ag carrier for the development of antiviral or antitumoral vaccines.


Subject(s)
Fibronectins/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Toll-Like Receptor 4/metabolism , Amino Acid Motifs , Animals , Bone Marrow/metabolism , Cell Differentiation , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Female , Fibronectins/chemistry , Fibronectins/genetics , Humans , Interleukin-12/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Ovalbumin/immunology , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/biosynthesis
16.
Vaccine ; 23(27): 3493-9, 2005 May 20.
Article in English | MEDLINE | ID: mdl-15855007

ABSTRACT

The induction of protective or therapeutic cellular immunity against hepatitis C virus (HCV) is a difficult goal. In a previous work we showed that immunization with a recombinant adenovirus encoding HCV-NS3 (RAdNS3) could partially protect mice from challenge with a vaccinia virus encoding HCV antigens. We sought to investigate whether systemic administration of an immunostimulatory monoclonal antibody directed against the lymphocyte surface molecule CD137 could enhance the immunity elicited by RAdNS3. It was found that treatment with anti-CD137 mAb after the administration of a suboptimal dose of RAdNS3 enhanced cytotoxic and T helper cell responses against HCV NS3. Importantly, the ability of RAdNS3 to induce protective immunity against challenge with a recombinant vaccinia virus expressing HCV proteins was markedly augmented. Thus, combination of immunostimulatory anti-CD137 mAb with recombinant adenoviruses expressing HCV proteins might be useful in strategies of immunization against HCV.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Hepacivirus/immunology , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/immunology , Viral Hepatitis Vaccines/immunology , Adenoviridae/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antigens, CD , Cells, Cultured , Female , Hepacivirus/drug effects , Hepatitis C/immunology , Hepatitis C/prevention & control , Hepatitis C/virology , Humans , Mice , Mice, Inbred BALB C , Receptors, Nerve Growth Factor/administration & dosage , Receptors, Tumor Necrosis Factor/administration & dosage , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Viral Hepatitis Vaccines/administration & dosage , Viral Load/methods
17.
Hepatology ; 37(2): 461-70, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12540797

ABSTRACT

Hepatitis C virus (HCV) infection has a strong tendency to evolve to chronicity despite up-regulation of proapoptotic cytokines in the inflamed liver. The mechanisms responsible for persistent viral replication in this inflammatory environment are obscure. It is conceivable that viral replication would be facilitated if the infected hepatocytes are rendered resistant to cytokine-induced cytotoxicity. In this study, we investigated if an adenovirus encoding HCV core and E1 (RAdCE1) could reduce liver cell injury in different in vivo models of cytokine-mediated hepatotoxicity in mice. We show that RAdCE1 markedly attenuates hepatocellular apoptosis and the increase in serum transaminase levels after concanavalin A (con A) challenge. This protective effect is accompanied by an inhibition of nuclear translocation of nuclear factor kappaB (NF-kappaB); reduced expression of inducible nitric oxide synthase (iNOS); decreased hepatic messenger RNA levels of chemokines macrophage inflammatory protein 2 (MIP-2), monocyte chemoattractant protein 1 (MCP-1), and interferon-inducible protein 10 (IP-10); and abrogation of liver leukocyte infiltration. RAdCE1 also causes a reduction in serum transaminase levels and inhibits hepatocellular apoptosis in mice given tumor necrosis factor (TNF)-alpha plus D-galactosamine. In conclusion, HCV structural antigens can protect liver cells against the proapoptotic effects of proinflammatory cytokines. The antiapoptotic status of infected liver cells may represent a mechanism favoring viral persistence. Our findings also suggest that, in chronic hepatitis C, the burden of hepatocellular damage mainly affects noninfected liver cells.


Subject(s)
Adenoviridae/genetics , Cytokines/physiology , Gene Expression/physiology , Hepacivirus/genetics , Liver Diseases/etiology , Liver Diseases/prevention & control , Viral Core Proteins/genetics , Viral Envelope Proteins/genetics , Acute Disease , Animals , Cell Nucleus/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Concanavalin A , Cytokines/antagonists & inhibitors , Drug Combinations , Female , Galactosamine , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Recombination, Genetic , Transduction, Genetic , Tumor Necrosis Factor-alpha
18.
Vaccine ; 21(3-4): 202-10, 2002 Dec 13.
Article in English | MEDLINE | ID: mdl-12450695

ABSTRACT

Cellular immune response plays an important role in the clearance of hepatitis C virus (HCV). Thus, development of efficient ways to induce anti-viral cellular immune responses is an important step toward prevention and/or treatment of HCV infection. With this aim, we have constructed a replication-deficient recombinant adenovirus expressing HCV NS3 protein (RAdNS3). The efficacy of RAdNS3 was tested in vivo by measuring the protection against infection with a recombinant vaccinia virus expressing HCV-polyprotein (vHCV1-3011). Immunisation with 10(9)pfu of RAdNS3 induced anti-NS3 humoral, T helper and T cytotoxic responses. We identified eight epitopes recognised by IFN-gamma producing cells, five of them exhibiting lytic activity. Moreover, we show that RAdNS3 immunised mice were protected against challenge with vHCV1-3011 and that this protection was mediated by CD8(+) cells. In conclusion, our results suggest that adenoviral vectors encoding NS3 might be useful for the induction of prophylactic and/or therapeutic anti-HCV immunity.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/immunology , Hepacivirus/immunology , Hepatitis C Antibodies/biosynthesis , Hepatitis C/prevention & control , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic/immunology , Viral Hepatitis Vaccines/immunology , Viral Nonstructural Proteins/administration & dosage , Adenoviridae/immunology , Animals , Cell Line , Enzyme-Linked Immunosorbent Assay , Mice , Mice, Inbred BALB C , Vaccination , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...