Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Biol Sci ; 17(11): 2944-2956, 2021.
Article in English | MEDLINE | ID: mdl-34345218

ABSTRACT

The generation of successful anticancer vaccines relies on the ability to induce efficient and long-lasting immune responses to tumor antigens. In this scenario, dendritic cells (DCs) are essential cellular components in the generation of antitumor immune responses. Thus, delivery of tumor antigens to specific DC populations represents a promising approach to enhance the efficiency of antitumor immunotherapies. In the present study, we employed antibody-antigen conjugates targeting a specific DC C-type lectin receptor. For that purpose, we genetically fused the anti-DEC205 monoclonal antibody to the type 16 human papillomavirus (HPV-16) E7 oncoprotein to create a therapeutic vaccine to treat HPV-associated tumors in syngeneic mouse tumor models. The therapeutic efficacy of the αDEC205-E7 mAb was investigated in three distinct anatomical tumor models (subcutaneous, lingual and intravaginal). The immunization regimen comprised two doses of the αDEC205-E7 mAb coadministered with a DC maturation stimulus (Polyinosinic:polycytidylic acid, poly (I:C)) as an adjuvant. The combined immunotherapy produced robust antitumor effects on both the subcutaneous and orthotopic tumor models, stimulating rapid tumor regression and long-term survival. These outcomes were related to the activation of tumor antigen-specific CD8+ T cells in both systemic compartments and lymphoid tissues. The αDEC205-E7 antibody plus poly (I:C) administration induced long-lasting immunity and controlled tumor relapses. Our results highlight that the delivery of HPV tumor antigens to DCs, particularly via the DEC205 surface receptor, is a promising therapeutic approach, providing new opportunities for the development of alternative immunotherapies for patients with HPV-associated tumors at different anatomical sites.


Subject(s)
Antigens, CD/immunology , Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Lectins, C-Type/immunology , Minor Histocompatibility Antigens/immunology , Neoplasms, Experimental/prevention & control , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/prevention & control , Receptors, Cell Surface/immunology , Adjuvants, Immunologic , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Female , Humans , Immunologic Memory , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/virology , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Poly I-C/administration & dosage
2.
Cancer Immunol Immunother ; 68(5): 753-763, 2019 May.
Article in English | MEDLINE | ID: mdl-30806747

ABSTRACT

The presence of IL-10, produced either by tumor cells or immunosuppressive cells, is frequently associated with a poor prognosis for cancer progression. It may also negatively impact anticancer treatments, such as immunotherapies, that otherwise would promote the activation of cytotoxic T cells capable of detecting and destroying malignant cells. In the present study, we evaluated a new adjuvant approach for anticancer immunotherapy using a plasmid vector encoding a soluble form of the IL-10 receptor (pIL-10R). pIL-10R was coadministered to mice with a DNA vaccine encoding the type 16 human papillomavirus (HPV-16) E7 oncoprotein genetically fused with glycoprotein D of herpes simplex virus (HSV) (pgDE7h). Immunization regimens based on the coadministration of pIL-10R and pgDE7h enhanced the antitumor immunity elicited in mice injected with TC-1 cells, which express HPV-16 oncoproteins. The administration of the DNA vaccines by in vivo electroporation further enhanced the anticancer effects of the vaccines, leading to the activation of tumor-infiltrating polyfunctional E7-specific cytotoxic CD8+ T cells and control of the expansion of immunosuppressive cells. In addition, the combination of immunotherapy and pIL-10R allowed the control of tumors in more advanced growth stages that otherwise would not be treatable by the pgDE7h vaccine. In conclusion, the proposed treatment involving the expression of IL-10R enhanced the antitumor protective immunity induced by pgDE7h administration and may contribute to the development of more efficient clinical interventions against HPV-induced tumors.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Epithelial Cells/physiology , Human papillomavirus 16/physiology , Immunotherapy/methods , Neoplasms, Experimental/immunology , Papillomavirus Infections/immunology , Papillomavirus Vaccines/immunology , Receptors, Interleukin-10/immunology , Animals , Immune Tolerance , Interleukin-10/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Papillomavirus E7 Proteins/genetics , Papillomavirus Vaccines/genetics , Receptors, Interleukin-10/genetics , Vaccines, DNA , Viral Envelope Proteins/genetics
3.
Vaccine ; 35(52): 7240-7249, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29174677

ABSTRACT

In vivo electroporation (EP) has reignited the clinical interest on DNA vaccines as immunotherapeutic approaches to control different types of cancer. EP has been associated with increased immune response potency, but its capacity in influencing immunomodulation remains unclear. Here we evaluated the impact of in vivo EP on the induction of cellular immune responses and therapeutic effects of a DNA vaccine targeting human papillomavirus-induced tumors. Our results demonstrate that association of EP with the conventional intramuscular administration route promoted a more efficient activation of multifunctional and effector memory CD8+ T cells with enhanced cytotoxic activity. Furthermore, EP increased tumor infiltration of CD8+ T cells and avoided tumor recurrences. Finally, our results demonstrated that EP promotes local migration of antigen presenting cells that enhances with vaccine co-delivery. Altogether the present evidences shed further light on the in vivo electroporation action and its impact on the immunogenicity of DNA vaccines.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Electroporation/methods , Immunologic Memory , Neoplasms/therapy , Papillomavirus Vaccines/administration & dosage , Vaccines, DNA/immunology , Animals , Antigen-Presenting Cells/immunology , Cell Movement , Cytotoxicity, Immunologic , Humans , Immunity, Cellular , Immunogenicity, Vaccine , Injections, Intramuscular , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Neoplasms/immunology , Neoplasms/virology , Papillomaviridae/immunology , Papillomaviridae/isolation & purification , Papillomaviridae/physiology , Papillomavirus Infections/complications , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/therapeutic use , Recurrence , Vaccination/methods , Vaccines, DNA/administration & dosage
4.
Mol Cancer Ther ; 15(8): 1920-30, 2016 08.
Article in English | MEDLINE | ID: mdl-27222537

ABSTRACT

Active anticancer immunotherapeutic approaches have been shown to induce cellular or humoral immune responses in patients, but, thus far, the observed outcomes did not ensure their recommendation for clinical use. The induction of tumor-specific CD8(+) T cells, although required for the clearance of most solid tumors, was shown to be insufficient for the development of a successful immunotherapeutic approach. The suppressive immune environment triggered by tumors, including the expansion of myeloid-derived suppressor cells (MDSC), is detrimental to the development of antitumor immune responses and precludes the generation of more promising clinical outcomes. In this work, we characterized the CD8(+) T-cell population specifically involved in the control of tumor growth and the role of MDSCs after administration of an antitumor therapeutic DNA vaccine targeting human papillomavirus type 16 (HPV-16)-associated tumors. Activation of cytotoxic high-avidity CD8(+) T cells with an effector memory phenotype was found in mice grafted with tumor cells expressing the HPV-16 oncoproteins. In addition, MDSC antibody depletion further enhanced the immunotherapeutic effects of the vaccine, resulting in the complete eradication of tumor cells. Collectively, the current results indicate that the simultaneous control of MDSCs and activation of high-avidity tumor-specific effector memory CD8(+) T cells are key features for tumor protection by immunotherapeutic approaches and deserve further testing under clinical conditions. Mol Cancer Ther; 15(8); 1920-30. ©2016 AACR.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Human papillomavirus 16/immunology , Immunologic Memory , Lymphocyte Activation/immunology , Myeloid-Derived Suppressor Cells/immunology , Papillomavirus Infections/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/immunology , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immunization , Interferon-gamma/biosynthesis , Mice , Myeloid-Derived Suppressor Cells/metabolism , Neoplasms/etiology , Neoplasms/mortality , Neoplasms/pathology , Neoplasms/therapy , Papillomavirus Infections/complications , Papillomavirus Infections/virology , T-Cell Antigen Receptor Specificity/immunology , Tumor Burden/immunology , Vaccines, DNA/immunology , Xenograft Model Antitumor Assays
5.
Vaccine ; 33(20): 2328-34, 2015 May 11.
Article in English | MEDLINE | ID: mdl-25819710

ABSTRACT

Recently, Bacillus subtilis spores were shown to be endowed with strong adjuvant capacity when co-administered with purified antigenic proteins. In the present study we assessed whether spores possess adjuvant properties when combined with DNA vaccines. We showed that B. subtilis spores promoted the activation of dendritic cells in vitro and induced migration of pro-inflammatory cells after parenteral administration to mice. Likewise, co-administration of spores with a DNA vaccine encoding the human papillomavirus type 16 (HPV-16) E7 protein enhanced the activation of antigen-specific CD8(+) T cell responses in vivo. Mice immunized with the DNA vaccine admixed with spores presented a protective immunity increase to previously implanted tumor cells, capable of expressing HPV-16 oncoproteins. Finally, we observed that the adjuvant effect can vary accordingly to the number of co-administered spores which may be ascribed with the ability to induce. Collectively, the present results demonstrate for the first time that B. subtilis spores can also confer adjuvant effects to DNA vaccines.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bacillus subtilis/immunology , Spores, Bacterial/immunology , Vaccines, DNA/immunology , Animals , Bacillus subtilis/physiology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cytokines/immunology , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Interferon-gamma/immunology , Male , Mice, Inbred C57BL , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/immunology , Vaccines, DNA/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...