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










Database
Language
Publication year range
1.
Cancer Res ; 80(12): 2575-2585, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32107211

ABSTRACT

Because of the high coverage of international vaccination programs, most people worldwide have been vaccinated against common pathogens, leading to acquired pathogen-specific immunity with a robust memory T-cell repertoire. Although CD8+ antitumor cytotoxic T lymphocytes (CTL) are the preferred effectors of cancer immunotherapy, CD4+ T-cell help is also required for an optimal antitumor immune response to occur. Hence, we investigated whether the pathogen-related CD4+ T-cell memory populations could be reengaged to support the CTLs, converting a weak primary antitumor immune response into a stronger secondary one. To this end, we used our PeptiCRAd technology that consists of an oncolytic adenovirus coated with MHC-I-restricted tumor-specific peptides and developed it further by introducing pathogen-specific MHC-II-restricted peptides. Mice preimmunized with tetanus vaccine were challenged with B16.OVA tumors and treated with the newly developed hybrid TT-OVA-PeptiCRAd containing both tetanus toxoid- and tumor-specific peptides. Treatment with the hybrid PeptiCRAd significantly enhanced antitumor efficacy and induced TT-specific, CD40 ligand-expressing CD4+ T helper cells and maturation of antigen-presenting cells. Importantly, this approach could be extended to naturally occurring tumor peptides (both tumor-associated antigens and neoantigens), as well as to other pathogens beyond tetanus, highlighting the usefulness of this technique to take full advantage of CD4+ memory T-cell repertoires when designing immunotherapeutic treatment regimens. Finally, the antitumor effect was even more prominent when combined with the immune checkpoint inhibitor anti-PD-1, strengthening the rationale behind combination therapy with oncolytic viruses. SIGNIFICANCE: These findings establish a novel technology that enhances oncolytic cancer immunotherapy by capitalizing on pre-acquired immunity to pathogens to convert a weak antitumor immune response into a much stronger one.


Subject(s)
Cancer Vaccines/administration & dosage , Diphtheria-Tetanus-Pertussis Vaccine/administration & dosage , Immunologic Memory , Immunotherapy/methods , Melanoma, Experimental/therapy , Poliovirus Vaccine, Inactivated/administration & dosage , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/administration & dosage , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cell Line, Tumor/transplantation , Diphtheria-Tetanus-Pertussis Vaccine/immunology , Female , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Poliovirus Vaccine, Inactivated/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Combined/administration & dosage , Vaccines, Combined/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...