Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncoimmunology ; 10(1): 1912473, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33907631

RESUMEN

High-risk human papillomavirus (HPV) encoding E6/E7-HPV oncogenes are responsible for a subgroup of head and neck squamous-cell carcinoma (HNSCC) and thus therapeutic E7-vaccines may be used to control HPV+HNSCC tumors. Herein we investigated the effects of an optimized nanoparticle-conjugated E7 long-peptide vaccine adjuvanted with CpG (NP-E7LP) in an orthotopic immunocompetent mouse model of HPV+HNSCC which is based on injection of HPV16 E6/E7-expressing mEERL95-cells into the submental space. In absence of surgery, vaccination performed before or after tumor-cell injection decreased tumor growth or prolonged mice survival only marginally, despite the high numbers of vaccine-induced circulating E7-specific IFN-γ-secreting CD8+ T-cells. This contrasts with the high-efficacy of NP-E7LP-vaccination reported in the genital and subcutaneous HPV16-E6/E7-expressing TC-1 models. Our data show that in a direct comparison, NP-E7LP-vaccination fully controlled TC-1, but not mEERL95, tumors subcutaneously growing in the flanks. Immune-cell infiltration was 10-fold higher in TC-1-tumors, than in mEERL95-tumors, suggesting that vaccine-induced CD8+ T-cells can only poorly infiltrate mEERL95-tumors. Indeed, immunofluorescence staining of orthotopic mEERL95-tumors showed that CD3+ T-cells are preferentially located peritumorally. However, when NP-E7LP-vaccination was performed after mEERL95-cell injection, but before resection of primary tumors, no postsurgical recurrence was observed and 100% of the mice survived until the experimental endpoint (day 70) in the NP-E7LP-vaccinated group. In contrast, we observed a 60% recurrence rate and only 35% survival in PBS-vaccinated mice. This suggests that removal of the primary tumor modified the tumor microenvironment, allowing a therapeutic effect of the vaccine-induced anti-tumor response. E7-vaccination combined with surgery may thus benefit patients with HPV+HNSCC.


Asunto(s)
Alphapapillomavirus , Vacunas contra el Cáncer , Neoplasias de Cabeza y Cuello , Nanopartículas , Vacunas contra Papillomavirus , Animales , Linfocitos T CD8-positivos , Humanos , Ratones , Ratones Endogámicos C57BL , Recurrencia Local de Neoplasia/prevención & control , Papillomaviridae , Microambiente Tumoral , Vacunación
2.
Cancer Immunol Res ; 8(1): 131-145, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31771984

RESUMEN

Cancers induced by human papillomaviruses (HPV) should be responsive to immunotherapy by virtue of expressing the immunogenic oncoproteins E6/E7. However, advanced forms of cervical cancer, driven by HPV, are poorly responsive to immune response-enhancing treatments involving therapeutic vaccination against these viral neoantigens. Leveraging a transgenic mouse model of HPV-derived cancers, K14HPV16/H2b, we demonstrated that a potent nanoparticle-based E7 vaccine, but not a conventional "liquid" vaccine, induced E7 tumor antigen-specific CD8+ T cells in cervical tumor-bearing mice. Vaccination alone or in combination with anti-PD-1/anti-CTLA4 did not elicit tumor regression nor increase CD8+ T cells in the tumor microenvironment (TME), suggesting the presence of immune-suppressive barriers. Patients with cervical cancer have poor dendritic cell functions, have weak cytotoxic lymphocyte responses, and demonstrate an accumulation of myeloid cells in the periphery. Here, we illustrated that myeloid cells in K14HPV16/H2b mice possess potent immunosuppressive activity toward antigen-presenting cells and CD8+ T cells, dampening antitumor immunity. These immune-inhibitory effects inhibited synergistic effects of combining our oncoprotein vaccine with immune checkpoint-blocking antibodies. Our data highlighted a link between HPV-induced cancers, systemic amplification of myeloid cells, and the detrimental effects of myeloid cells on CD8+ T-cell activation and recruitment into the TME. These results established immunosuppressive myeloid cells in lymphoid organs as an HPV+ cancer-induced means of circumventing tumor immunity that will require targeted abrogation to enable the induction of efficacious antitumor immune responses.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Linfocitos T CD8-positivos/inmunología , Células Mieloides/inmunología , Papillomaviridae/inmunología , Infecciones por Papillomavirus/inmunología , Vacunas contra Papillomavirus/inmunología , Neoplasias del Cuello Uterino/inmunología , Animales , Antígeno CTLA-4/antagonistas & inhibidores , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Humanos , Terapia de Inmunosupresión , Inmunoterapia/métodos , Ratones , Células Mieloides/efectos de los fármacos , Nanopartículas/administración & dosificación , Proteínas E7 de Papillomavirus/inmunología , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/tratamiento farmacológico , Infecciones por Papillomavirus/virología , Vacunas contra Papillomavirus/administración & dosificación , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Microambiente Tumoral , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/virología
3.
J Immunother Cancer ; 7(1): 122, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-31060612

RESUMEN

High-risk human papillomavirus (HPV) are responsible for genital and oral cancers associated with the expression of the E6/E7 HPV oncogenes. Therapeutic vaccines targeting those oncogenes can only partially control tumor progression, highlighting the necessity to investigate different treatment strategies. Using the genital orthotopic HPV16 TC-1 model, herein we sequentially investigated in progressively more stringent settings the effects of systemic administration of carboplatin/paclitaxel (C + P) chemotherapy combined with HPV16-E7 synthetic long peptide (E7LP) vaccination, followed by intravaginal immunostimulation with the synthetic toll-like-receptor-9 agonist CpG. Our data show that systemic delivery of C + P prior to E7LP vaccination significantly increased mice survival. This survival benefit was associated with both reduced genital tumor growth at the time of vaccination, and a decreased infiltration of Ly6G myeloid cells and tumor-associated macrophages. Adding intravaginal CpG, which results in increased E7-specific CD8 T cells locally, to E7LP vaccination and the chemotherapy formed a tri-therapy, which significantly increased mice survival as compared to any of the dual treatments. When the tri-therapy was further refined by using a recently optimized nanoparticle-conjugated E7LP vaccine, even larger end-stage genital-TC-1 tumors responded, with 90% of mice showing a survival benefit as compared to 30% of mice with the tri-therapy involving the traditional E7LP 'liquid' vaccine. C + P is commonly used to treat cervical cancer patients and its combination with E7/E6 vaccination is currently being tested in a phase I/II trial (NCT02128126). Our data suggests that new vaccine formulations combined with local immunostimulation and standard-of-care chemotherapy have promise to further benefit patients with HPV-associated cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Neoplasias de los Genitales Femeninos/terapia , Inmunoterapia/métodos , Oligodesoxirribonucleótidos/administración & dosificación , Infecciones por Papillomavirus/terapia , Administración Intravaginal , Animales , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Femenino , Neoplasias de los Genitales Femeninos/inmunología , Neoplasias de los Genitales Femeninos/patología , Neoplasias de los Genitales Femeninos/virología , Papillomavirus Humano 16/inmunología , Papillomavirus Humano 16/aislamiento & purificación , Papillomavirus Humano 16/patogenicidad , Humanos , Inyecciones Intraperitoneales , Inyecciones Subcutáneas , Ratones , Paclitaxel/administración & dosificación , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/inmunología , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/virología , Receptor Toll-Like 9/agonistas , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vagina/inmunología , Vagina/patología , Vagina/virología
4.
Cancer Immunol Res ; 6(11): 1301-1313, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30131378

RESUMEN

Treatment of patients bearing human papillomavirus (HPV)-related cancers with synthetic long-peptide (SLP) therapeutic vaccines has shown promising results in clinical trials against premalignant lesions, whereas responses against later stage carcinomas have remained elusive. We show that conjugation of a well-documented HPV-E7 SLP to ultra-small polymeric nanoparticles (NP) enhances the antitumor efficacy of therapeutic vaccination in different mouse models of HPV+ cancers. Immunization of TC-1 tumor-bearing mice with a single dose of NP-conjugated E7LP (NP-E7LP) generated a larger pool of E7-specific CD8+ T cells with increased effector functions than unconjugated free E7LP. At the tumor site, NP-E7LP prompted a robust infiltration of CD8+ T cells that was not accompanied by concomitant accumulation of regulatory T cells (Tregs), resulting in a higher CD8+ T-cell to Treg ratio. Consequently, the amplified immune response elicited by the NP-E7LP formulation led to increased regression of large, well-established tumors, resulting in a significant percentage of complete responses that were not achievable by immunizing with the non-NP-conjugated long-peptide. The partial responses were characterized by distinct phases of regression, stable disease, and relapse to progressive growth, establishing a platform to investigate adaptive resistance mechanisms. The efficacy of NP-E7LP could be further improved by therapeutic activation of the costimulatory receptor 4-1BB. This NP-E7LP formulation illustrates a "solid-phase" antigen delivery strategy that is more effective than a conventional free-peptide ("liquid") vaccine, further highlighting the potential of using such formulations for therapeutic vaccination against solid tumors. Cancer Immunol Res; 6(11); 1301-13. ©2018 AACR.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacología , Nanopartículas/química , Proteínas E7 de Papillomavirus/química , Animales , Anticuerpos/farmacología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/química , Femenino , Neoplasias Pulmonares/secundario , Ratones Endogámicos C57BL , Ratones Transgénicos , Recurrencia Local de Neoplasia , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/mortalidad , Neoplasias Experimentales/terapia , Proteínas E7 de Papillomavirus/inmunología , Proteínas E7 de Papillomavirus/farmacología , Linfocitos T Reguladores/inmunología , Resultado del Tratamiento , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Neoplasias Vaginales/inmunología , Neoplasias Vaginales/patología , Neoplasias Vaginales/prevención & control
5.
Sci Transl Med ; 9(415)2017 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-29118259

RESUMEN

Immune checkpoint blockade exhibits considerable antitumor activity, but previous studies have reported instances of severe treatment-related adverse events. We sought to explore local immune checkpoint blockade, with an antibody (Ab) form that would be retained intra- or peritumorally, limiting systemic exposure. To accomplish this, we conjugated the checkpoint blockade Abs to an extracellular matrix (ECM)-super-affinity peptide derived from placenta growth factor-2 (PlGF-2123-144). We show enhanced tissue retention and lower Ab concentrations in blood plasma after PlGF-2123-144 conjugation, reducing systemic side effects such as the risk of autoimmune diabetes. Peritumoral injections of PlGF-2123-144-anti-CTLA4 (cytotoxic T lymphocyte antigen 4) and PlGF-2123-144-anti-PD-L1 (programmed death ligand 1) Abs delayed tumor growth and prolonged survival compared to the unmodified Abs in genetically engineered murine tumor models of melanoma and breast cancer. The PlGF-2123-144-Abs increased tumor-infiltrating activated CD8+ and CD4+ T cells, resulting in a delay of distant tumor growth as well. This simple and translatable approach of engineered ECM-binding Abs may present a viable and safer approach in checkpoint blockade.


Asunto(s)
Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Matriz Extracelular/metabolismo , Inmunoterapia , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Animales , Anticuerpos/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4 , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/metabolismo , Fibrinolisina/metabolismo , Ingeniería Genética , Heparina/metabolismo , Inmunidad , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Péptidos/farmacología , Péptidos/uso terapéutico , Factor de Crecimiento Placentario/química , Resultado del Tratamiento
7.
Sci Rep ; 7: 45961, 2017 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-28401888

RESUMEN

A robust and reproducible method for culturing monolayers of adherent and well-spread primary islet cells on glass coverslips is required for detailed imaging studies by super-resolution and live-cell microscopy. Guided by an observation that dispersed islet cells spread and adhere well on glass surfaces in neuronal co-culture and form a monolayer of connected cells, we demonstrate that in the absence of neurons, well-defined surface coatings combined with components of neuronal culture media collectively support robust attachment and growth of primary human or rat islet cells as monolayers on glass surfaces. The islet cell monolayer cultures on glass stably maintain distinct mono-hormonal insulin+, glucagon+, somatostatin+ and PP+ cells and glucose-responsive synchronized calcium signaling as well as expression of the transcription factors Pdx-1 and NKX-6.1 in beta cells. This technical advance enabled detailed observation of sub-cellular processes in primary human and rat beta cells by super-resolution microscopy. The protocol is envisaged to have broad applicability to sophisticated analyses of pancreatic islet cells that reveal new biological insights, as demonstrated by the identification of an in vitro protocol that markedly increases proliferation of primary beta cells and is associated with a reduction in ciliated, ostensibly proliferation-suppressed beta cells.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Cilios/metabolismo , Vidrio/química , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Microscopía/métodos , Organogénesis , Actinas/metabolismo , Adolescente , Adulto , Animales , Calcio/metabolismo , Adhesión Celular , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Femenino , Glucosa/metabolismo , Hipocampo/citología , Humanos , Insulina/metabolismo , Masculino , Microtúbulos/metabolismo , Persona de Mediana Edad , Neuronas/citología , Neuronas/metabolismo , Fenotipo , Ratas Sprague-Dawley , Adulto Joven
8.
Cell Rep ; 5(2): 323-30, 2013 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-24120862

RESUMEN

Bisphosphonates are a class of drugs that are widely used to inhibit loss of bone mass in patients. We show here that the administration of clinically relevant doses of bisphosphonates in mice increases antibody responses to live and inactive viruses, proteins, haptens, and existing commercial vaccine formulations. Bisphosphonates exert this adjuvant-like activity in the absence of CD4(+) and γδ T cells, neutrophils, or dendritic cells, and their effect does not rely on local macrophage depletion, Toll-like receptor signaling, or the inflammasome. Rather, bisphosphonates target directly B cells and enhance B cell expansion and antibody production upon antigen encounter. These data establish bisphosphonates as an additional class of adjuvants that boost humoral immune responses.


Asunto(s)
Linfocitos B/metabolismo , Difosfonatos/farmacología , Inmunidad Humoral/efectos de los fármacos , Adyuvantes Inmunológicos , Animales , Formación de Anticuerpos/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Humanos , Inmunoglobulina G/metabolismo , Inflamasomas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Transducción de Señal , Receptores Toll-Like/metabolismo , Vesiculovirus/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...