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











Intervalo de año de publicación
1.
iScience ; 26(4): 106320, 2023 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-36968078

RESUMEN

HER2-targeted therapy has improved breast cancer survival, but treatment resistance and disease prevention remain major challenges. Genes that enable HER2/Neu oncogenesis are the next intervention targets. A bioinformatics discovery platform of HER2/Neu-expressing Diversity Outbred (DO) F1 Mice was established to identify cancer-enabling genes. Quantitative Trait Loci (QTL) associated with onset ages and growth rates of spontaneous mammary tumors were sought. Twenty-six genes in 3 QTL contain sequence variations unique to the genetic backgrounds that are linked to aggressive tumors and 21 genes are associated with human breast cancer survival. Concurrent identification of TSC22D3, a transcription factor, and its target gene LILRB4, a myeloid cell checkpoint receptor, suggests an immune axis for regulation, or intervention, of disease. We also investigated TIEG1 gene that impedes tumor immunity but suppresses tumor growth. Although not an actionable target, TIEG1 study revealed genetic regulation of tumor progression, forming the basis of the genetics-based discovery platform.

2.
J Immunol ; 205(6): 1554-1563, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32796024

RESUMEN

The genetic basis and mechanisms of disparate antitumor immune response was investigated in Diversity Outbred (DO) F1 mice that express human HER2. DO mouse stock samples nearly the entire genetic repertoire of the species. We crossed DO mice with syngeneic HER2 transgenic mice to study the genetics of an anti-self HER2 response in a healthy outbred population. Anti-HER2 IgG was induced by Ad/E2TM or naked pE2TM, both encoding HER2 extracellular and transmembrane domains. The response of DO F1 HER2 transgenic mice was remarkably variable. Still, immune sera inhibited HER2+ SKBR3 cell survival in a dose-dependent fashion. Using DO quantitative trait locus (QTL) analysis, we mapped the QTL that influences both total IgG and IgG2(a/b/c) Ab response to either Ad/E2TM or pE2TM. QTL from these four datasets identified a region in chromosome 17 that was responsible for regulating the response. A/J and NOD segments of genes in this region drove elevated HER2 Ig levels. This region is rich in MHC-IB genes, several of which interact with inhibitory receptors of NK cells. (B6xA/J)F1 and (B6xNOD)F1 HER2 transgenic mice received Ad/E2TM after NK cell depletion, and they produced less HER2 IgG, demonstrating positive regulatory function of NK cells. Depletion of regulatory T cells enhanced response. Using DO QTL analysis, we show that MHC-IB reactive NK cells exert positive influence on the immunity, countering negative regulation by regulatory T cells. This new, to our knowledge, DO F1 platform is a powerful tool for revealing novel immune regulatory mechanisms and for testing new interventional strategies.


Asunto(s)
Autoantígenos/metabolismo , Isoantígenos/metabolismo , Células Asesinas Naturales/fisiología , Sitios de Carácter Cuantitativo/genética , Receptor ErbB-2/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Animales no Consanguíneos , Autoantígenos/genética , Autoantígenos/inmunología , Femenino , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Inmunidad , Inmunoglobulina G/sangre , Isoantígenos/genética , Isoantígenos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Dominios Proteicos/genética , Receptor ErbB-2/genética , Receptor ErbB-2/inmunología
3.
Cancer Immunol Immunother ; 68(7): 1143-1155, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31177328

RESUMEN

Enhancement of endogenous immunity to tumor-associated self-antigens and neoantigens is the goal of preventive vaccination. Toward this goal, we compared the efficacy of the following HER2 DNA vaccine constructs: vaccines encoding wild-type HER2, hybrid HER2 vaccines consisting of human HER2 and rat Neu, HER2 vaccines with single residue substitutions and a novel human HER2 DNA vaccine, ph(es)E2TM. ph(es)E2TM was designed to contain five evolution-selected substitutions: M198V, Q398R, F425L, H473R and A622T that occur frequently in 12 primate HER2 sequences. These ph(es)E2TM substitutions score 0 to 1 in blocks substitutions matrix (BLOSUM), indicating minimal biochemical alterations. h(es)E2TM recombinant protein is recognized by a panel of anti-HER2 mAbs, demonstrating the preservation of HER2 protein structure. Compared to native human HER2, electrovaccination of HER2 transgenic mice with ph(es)E2TM induced a threefold increase in HER2-binding antibody (Ab) and elevated levels of IFNγ-producing T cells. ph(es)E2TM, but not pE2TM immune serum, recognized HER2 peptide p95 355LPESFDGDPASNTAP369, suggesting a broadening of epitope recognition induced by the minimally modified HER2 vaccine. ph(es)E2TM vaccination reduced tumor growth more effectively than wild-type HER2 or HER2 vaccines with more extensive modifications. The elevation of tumor immunity by ph(es)E2TM vaccination would create a favorable tumor microenvironment for neoantigen priming, further enhancing the protective immunity. The fundamental principle of exploiting evolution-selected amino acid substitutions is novel, effective and applicable to vaccine development in general.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Inmunoterapia/métodos , Neoplasias Mamarias Experimentales/terapia , Receptor ErbB-2/inmunología , Sustitución de Aminoácidos/genética , Sustitución de Aminoácidos/inmunología , Animales , Antígenos de Neoplasias/genética , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/uso terapéutico , Línea Celular Tumoral/trasplante , Células Dendríticas/inmunología , Evolución Molecular , Femenino , Inmunogenicidad Vacunal/genética , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Receptor ErbB-2/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Autotolerancia/genética , Microambiente Tumoral/inmunología , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Vacunas de ADN/uso terapéutico
4.
Cancer Res ; 78(19): 5706-5717, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30115693

RESUMEN

IFNγ is an attractive target for imaging active antitumor immunity due to its function in the T-cell signaling axis. Here, we test an IFNγ immuno-PET (immunoPET) probe for its capacity to identify adaptive immunotherapy response after HER2/neu vaccination in both spontaneous salivary and orthotopic neu+ mouse mammary tumors. IFNγ immunoPET detected elevated cytokine levels in situ after vaccination, which inversely correlated with tumor growth rate, an indicator of response to therapy. In a model of induced T-cell anergy where CD8 T cells infiltrate the tumor, but upregulate PD-1, IFNγ tracer uptake was equivalent to isotype control, illustrating a lack of antitumor T-cell activity. The IFNγ immunoPET tracer detected IFNγ protein sequestered on the surface of tumor cells, likely in complex with the IFNγ receptor, which may explain imaging localization of this soluble factor in vivo Collectively, we find that the activation status of cytotoxic T cells is annotated by IFNγ immunoPET, with reduced off-target binding to secondary lymphoid tissues compared with imaging total CD3+ tumor-infiltrating lymphocytes. Targeting of soluble cytokines such as IFNγ by PET imaging may provide valuable noninvasive insight into the function of immune cells in situ Significance: This study presents a novel approach to monitor therapeutic outcomes via IFNγ-targeted positron emission tomography. Cancer Res; 78(19); 5706-17. ©2018 AACR.


Asunto(s)
Inmunoterapia , Interferón gamma/metabolismo , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Tomografía de Emisión de Positrones , Animales , Unión Competitiva , Complejo CD3/metabolismo , Linfocitos T CD8-positivos/citología , Línea Celular Tumoral , Islas de CpG , Femenino , Heterocigoto , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Mamarias Animales/diagnóstico por imagen , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/terapia , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Receptor de Muerte Celular Programada 1/metabolismo , Radiofármacos , Saliva/metabolismo , Linfocitos T Citotóxicos/citología
5.
Nanoscale ; 10(19): 9311-9319, 2018 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-29737353

RESUMEN

Nanotechnology has demonstrated tremendous clinical utility, with potential applications in cancer immunotherapy. Although nanoparticles with intrinsic cytotoxicity are often considered unsuitable for clinical applications, such toxicity may be harnessed in the fight against cancer. Nanoparticle-associated toxicity can induce acute necrotic cell death, releasing tumor-associated antigens which may be captured by antigen-presenting cells to initiate or amplify tumor immunity. To test this hypothesis, cytotoxic cationic silica nanoparticles (CSiNPs) were directly administered into B16F10 melanoma implanted in C57BL/6 mice. CSiNPs caused plasma membrane rupture and oxidative stress of tumor cells, inducing local inflammation, tumor cell death and the release of tumor-associated antigens. The CSiNPs were further complexed with bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP), a molecular adjuvant which activates the stimulator of interferon genes (STING) in antigen-presenting cells. Compared with unformulated c-di-GMP, the delivery of c-di-GMP with CSiNPs markedly prolonged its local retention within the tumor microenvironment and activated tumor-infiltrating antigen-presenting cells. The combination of CSiNPs and a STING agonist showed dramatically increased expansion of antigen-specific CD8+ T cells, and potent tumor growth inhibition in murine melanoma. These results demonstrate that cationic nanoparticles can be used as an effective in situ vaccine platform which simultaneously causes tumor destruction and immune activation.


Asunto(s)
Muerte Celular , Melanoma Experimental/inmunología , Proteínas de la Membrana/metabolismo , Nanopartículas , Dióxido de Silicio , Microambiente Tumoral , Animales , Linfocitos T CD8-positivos , GMP Cíclico/administración & dosificación , GMP Cíclico/análogos & derivados , Femenino , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo
6.
Cancer Immunol Res ; 3(7): 777-86, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25711535

RESUMEN

Domestic cats share human living environments and genetic traits. They develop spontaneous feline mammary carcinoma (FMC) with similar histopathology to human breast cancer. HER2 and AKT phosphorylation was demonstrated in primary FMC by immunoblot analysis, indicating HER2 as a therapeutic target. FMC lines K12 and K248 expressing HER1, HER2, and HER3 were sensitive to receptor tyrosine kinase (RTK) inhibitors gefitinib and lapatinib. To test HER2 vaccine response in cats, purpose-bred, healthy cats were electrovaccinated with heterologous (xenogeneic) or point-mutated feline HER2 DNA. T-cell reactivity to feline self-HER2 was detected in 4 of 10 cats that received bear HER2, human-rat fusion HER2 (E2Neu) or mutant feline HER2 (feHER2-K), which contains a single amino acid substitution. The variable T-cell responses may resemble that in the genetically heterogeneous human population. All immune sera to heterologous HER2 recognized feline HER2 expressed in 3T3 cells (3T3/HER2), but not that in FMC K12 or K248. Immune sera to mutant pfeHER2-K bound 3T3/HER2 cells weakly, but they showed better recognition of K12 and K248 cells that also express HER1 and HER3, suggesting distinct HER2 epitopes displayed by FMC that may be simulated by feHER2-K. In summary, HER2 DNA electroporation overcomes T-cell immune tolerance in approximately 40% of healthy cats and induces antibodies with distinct specificity. Vaccination studies in domestic cats can expedite vaccine iteration to guide human vaccine design and better predict outcome, with the added benefit of helping feline mammary tumor patients.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Neoplasias Mamarias Animales/inmunología , Receptor ErbB-2/genética , Linfocitos T/inmunología , Vacunas de ADN/inmunología , Animales , Gatos , Línea Celular Tumoral , Proliferación Celular , Electroporación , Femenino , Humanos , Tolerancia Inmunológica , Ratones , Ratones Endogámicos BALB C , Ratas , Ursidae
7.
Cancer Res ; 74(19): 5409-20, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25092895

RESUMEN

Percutaneous cryoablation is a minimally invasive procedure for tumor destruction, which can potentially initiate or amplify antitumor immunity through the release of tumor-associated antigens. However, clinically efficacious immunity is lacking and regional recurrences are a limiting factor relative to surgical excision. To understand the mechanism of immune activation by cryoablation, comprehensive analyses of innate immunity and HER2/neu humoral and cellular immunity following cryoablation with or without peritumoral CpG injection were conducted using two HER2/neu(+) tumor systems in wild-type (WT), neu-tolerant, and SCID mice. Cryoablation of neu(+) TUBO tumor in BALB/c mice resulted in systemic immune priming, but not in neu-tolerant BALB NeuT mice. Cryoablation of human HER2(+) D2F2/E2 tumor enabled the functionality of tumor-induced immunity, but secondary tumors were refractory to antitumor immunity if rechallenge occurred during the resolution phase of the cryoablated tumor. A step-wise increase in local recurrence was observed in WT, neu-tolerant, and SCID mice, indicating a role of adaptive immunity in controlling residual tumor foci. Importantly, local recurrences were eliminated or greatly reduced in WT, neu tolerant, and SCID mice when CpG was incorporated in the cryoablation regimen, showing significant local control by innate immunity. For long-term protection, however, adaptive immunity was required because most SCID mice eventually succumbed to local tumor recurrence even with combined cryoablation and CpG treatment. This improved understanding of the mechanisms by which cryoablation affects innate and adaptive immunity will help guide appropriate combination of therapeutic interventions to improve treatment outcomes.


Asunto(s)
Islas de CpG , Crioterapia , Genes erbB-2 , Recurrencia Local de Neoplasia/prevención & control , Oligonucleótidos/farmacología , Animales , Terapia Combinada , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Recurrencia Local de Neoplasia/inmunología
8.
Cancer Res ; 70(1): 119-28, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20048073

RESUMEN

Immune tolerance to tumor-associated self-antigens poses a major challenge in the ability to mount an effective cancer vaccine response. To overcome immune tolerance to HER-2, we formulated DNA vaccines that express both human HER-2 and heterologous rat Neu sequences in separate plasmids or as single hybrid constructs that encode HER-2/Neu fusion proteins. Candidate vaccines were tested in Her-2 transgenic (Tg) mice of BALB/c (BALB), BALB/cxC57BL/6 F1 (F1), or C57BL/6 (B6) background, which exhibit decreasing immune responsiveness to HER-2. Analysis of various cocktails or hybrid vaccines defined a requirement for particular combination of HER/2/Neu sequences to effectively prime immune effector cells in HER-2 Tg mice. In B6 HER-2 Tg mice, rejection of HER-2-positive tumors protected mice from HER-2-negative tumors, providing evidence of epitope spreading. Our findings show that a strategy of combining heterologous antigen with self-antigens could produce a potent DNA vaccine that may be applicable to other tumor-associated antigens.


Asunto(s)
Vacunas contra el Cáncer/genética , Genes erbB-2/genética , Tolerancia Inmunológica/inmunología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Vacunas de ADN/inmunología , Animales , Femenino , Humanos , Neoplasias Mamarias Experimentales/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA