Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Mol Immunol ; 157: 91-100, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37002957

RESUMEN

Breast cancer is one of the leading causes of death that affects the female population worldwide. Despite advances in treatments and a greater understanding of the disease, there are still difficulties in successfully treating patients. Currently, the main challenge in the field of cancer vaccines is antigenic variability which can reduce antigen-specific T- cell response efficacy. The search for and validation of immunogenic antigen targets increased dramatically over the past few decades and, with the advent of modern sequencing techniques, permitting the fast and accurate identification of the neoantigen landscape of tumor cells, will undoubtedly continue to grow exponentially for years to come. We have previously implemented Variable Epitope Libraries (VEL) as an unconventional vaccine strategy in preclinical models and for identifying and selecting mutant epitope variants. Here, we used an alanine-based sequence to generate a 9-mer VEL-like combinatorial mimotope library G3d as a new class of vaccine immunogen. An in silico analysis of the 16,000 G3d-derived sequences revealed potential MHC-I binders and immunogenic mimotopes. We demonstrated the antitumor effect of treatment with G3d in the 4T1 murine model of breast cancer. Moreover, two different T cell proliferation screening assays against a panel of randomly selected G3d-derived mimotopes allowed the isolation of both stimulatory and inhibitory mimotopes showing differential therapeutic vaccine efficacy. Thus, the mimotope library is a promising vaccine immunogen and a reliable source for isolating molecular cancer vaccine components.


Asunto(s)
Neoplasias , Biblioteca de Péptidos , Femenino , Animales , Ratones , Epítopos , Modelos Animales de Enfermedad , Antígenos de Neoplasias
2.
Vaccine ; 40(33): 4796-4805, 2022 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-35788294

RESUMEN

The identification of novel targets for cancer immunotherapy and the development of new vaccine immunogens are subjects of permanent interest. MUC1 is an overexpressed antigen found in most tumors, and its overexpression correlates with poor prognosis. Many attempts to direct the immune response against MUC1 in tumor cells have failed, including several clinical trials. We have previously developed an innovative Variable Epitope Library (VEL) vaccine platform that carries massively substituted mutant variants of defined epitopes or epitope regions as an alternative to using wild-type peptide sequences-based immunogens. Here, two murine MUC1-derived epitopes equivalent to the previously tested in cancer immunotherapy human MUC1 regions were used to generate VELs. We observed that vaccination with the 23L VEL immunogens, encompassing the entire signal peptide region of MUC1, reduces the tumor area compared to the wild-type sequence treatment. Contrastingly, vaccination with the MUC1 signal peptide-derived predicted CD8++ T cell epitope-based VEL, 9MUC1spL, showed similar tumor area reduction as the wild-type treatment; however, a decrease in lung metastasis after 9MUC1spL treatment was observed. In addition, vaccination induced a large pool of CD8+ T cells which recognized most variant epitopes from 9MUC1spL. Also, we generated MUC1 variable number tandem repeat (VNTR)-based VELs that reduced the metastatic burden when dendritic cells and M13 recombinant bacteriophages were used as vaccine carriers. Collectively, our data demonstrate the immunogenic and antitumor properties of MUC1 signal peptide- and VNTR-derived VEL immunogens.


Asunto(s)
Neoplasias de la Mama , Vacunas contra el Cáncer , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Linfocitos T CD8-positivos , Modelos Animales de Enfermedad , Epítopos de Linfocito T/genética , Femenino , Humanos , Ratones , Mucina-1/genética , Señales de Clasificación de Proteína
3.
Mol Immunol ; 139: 65-75, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34454186

RESUMEN

After decades of cancer vaccine efforts, there is an imperious necessity for novel ideas that may result in better tumor control in patients. We have proposed the use of a novel Variable Epitope Library (VEL) vaccine strategy, which incorporates an unprecedented number of mutated epitopes to target antigenic variability and break tolerance against tumor-associated antigens. Here, we used an oncofetal antigen/immature laminin receptor protein-derived sequence to generate 9-mer and 43-mer VEL immunogens. 4T1 tumor-bearing mice developed epitope-specific CD8+IFN-γ+ and CD4+IFN-γ+ T cell responses after treatment. Tumor and lung analysis demonstrated that VELs could increase the number of tumor-infiltrating lymphocytes with diverse effector functions while reducing the number of immunosuppressive myeloid-derived suppressor and regulatory T cells. Most importantly, VEL immunogens inhibited tumor growth and metastasis after a single dose. The results presented here are consistent with our previous studies and provide evidence for VEL immunogens' feasibility as promising cancer immunotherapy.


Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias de la Mama , Vacunas contra el Cáncer/inmunología , Epítopos de Linfocito T/inmunología , Receptores de Laminina/inmunología , Animales , Vacunas contra el Cáncer/farmacología , Modelos Animales de Enfermedad , Mapeo Epitopo/métodos , Femenino , Ratones , Ratones Endogámicos BALB C
4.
Cell Immunol ; 358: 104235, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33137645

RESUMEN

Antibodies, T cell receptors and major histocompatibility complex molecules are members of the immunoglobulin superfamily and have pivotal roles in the immune system. The fine interrelation between them regulates several immune functions. Here, we describe lesser-known functions ascribed to these molecules in generating and maintaining immune response. Particularly, we outline the contribution of antibody- and T cell receptor-derived complementarity-determining region neoantigens, antigenized antibodies, as well as major histocompatibility complex class I molecules-derived epitopes to the induction of protective/therapeutic immune responses against pathogens and cancer. We discuss findings of our own and other studies describing protective mechanisms, based on immunogenic properties of immunoglobulin superfamily members, and evaluate the perspectives of application of this class of immunogens in molecular vaccines design.


Asunto(s)
Anticuerpos/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Secuencia de Aminoácidos/genética , Animales , Formación de Anticuerpos , Epítopos/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inmunidad/inmunología , Cadenas Pesadas de Inmunoglobulina , Región Variable de Inmunoglobulina , Inmunoglobulinas/inmunología , Inmunoglobulinas/metabolismo
5.
Immunology ; 161(2): 123-138, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32619293

RESUMEN

Immune tolerance is the main challenge in the field of cancer vaccines, so modified peptide sequences or naturally occurring mutated versions of cancer-related wild-type (WT) antigens represent a promising pathway. However, the low immunogenicity of mutation-induced neoantigens and, particularly, their incapacity to activate CD8+ T cells are generating doubts on the success of neoantigen-based cancer vaccines in clinical trials. We developed a novel vaccine approach based on a new class of vaccine immunogens, called variable epitope libraries (VELs). We used three regions of survivin (SVN), composed of 40, 49 and 51 amino acids, along with the complete SVN protein to generate the VELs as multiepitope vaccines. BALB/c mice, challenged with the aggressive and highly metastatic 4T1 cell line, were vaccinated in a therapeutic setting. We showed significant tumor growth inhibition and, most importantly, strong suppression of lung metastasis after a single immunization using VEL vaccines. We demonstrated vaccine-induced broad cellular immune responses concomitant with extensive tumor infiltration of T cells, the activation of CD107a+  IFN-γ+ T cells in the spleen and a significant increase in the number of CD3+  CD8+  Ly6C+ effector T cells. In addition, we observed the presence of interferon-γ-, granzyme B- and perforin-producing lymphocytes along with modifications in the amount of CD11b+  Ly6Cint/low  Ly6G+ granulocytic myeloid-derived suppressor cells and CD4+  CD25+  FoxP3+ regulatory T cells in the lungs and tumors of mice. In summary, we showed that the VELs represent a potent new class of cancer immunotherapy and propose the application of the VEL vaccine concept as a true alternative to currently available vaccine platforms.


Asunto(s)
Neoplasias de la Mama/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Epítopos de Linfocito T/inmunología , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Células Supresoras de Origen Mieloide/inmunología , Survivin/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Biblioteca de Péptidos , Survivin/genética , Survivin/inmunología , Vacunación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA