Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 14(10): 1014-22, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24048123

RESUMEN

Most tumor cells express antigens that can mediate recognition by host CD8(+) T cells. Cancers that are detected clinically must have evaded antitumor immune responses to grow progressively. Recent work has suggested two broad categories of tumor escape based on cellular and molecular characteristics of the tumor microenvironment. One major subset shows a T cell-inflamed phenotype consisting of infiltrating T cells, a broad chemokine profile and a type I interferon signature indicative of innate immune activation. These tumors appear to resist immune attack through the dominant inhibitory effects of immune system-suppressive pathways. The other major phenotype lacks this T cell-inflamed phenotype and appears to resist immune attack through immune system exclusion or ignorance. These two major phenotypes of tumor microenvironment may require distinct immunotherapeutic interventions for maximal therapeutic effect.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Neoplasias/inmunología , Microambiente Tumoral/inmunología , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Activación de Linfocitos , Neoplasias/terapia , Células del Estroma/inmunología , Células del Estroma/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
2.
Immunity ; 44(6): 1444-54, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27332733

RESUMEN

Genetically modified T cells expressing chimeric antigen receptors (CARs) demonstrate robust responses against lineage restricted, non-essential targets in hematologic cancers. However, in solid tumors, the full potential of CAR T cell therapy is limited by the availability of cell surface antigens with sufficient cancer-specific expression. The majority of CAR targets have been normal self-antigens on dispensable hematopoietic tissues or overexpressed shared antigens. Here, we established that abnormal self-antigens can serve as targets for tumor rejection. We developed a CAR that recognized cancer-associated Tn glycoform of MUC1, a neoantigen expressed in a variety of cancers. Anti-Tn-MUC1 CAR T cells demonstrated target-specific cytotoxicity and successfully controlled tumor growth in xenograft models of T cell leukemia and pancreatic cancer. These findings demonstrate the therapeutic efficacy of CAR T cells directed against Tn-MUC1 and present aberrantly glycosylated antigens as a novel class of targets for tumor therapy with engineered T cells.


Asunto(s)
Adenocarcinoma/terapia , Epítopos de Linfocito T/inmunología , Inmunoterapia/métodos , Mucina-1/inmunología , Linfocitos T/fisiología , Adenocarcinoma/inmunología , Animales , Línea Celular Tumoral , Citotoxicidad Inmunológica , Ingeniería Genética , Glicosilación , Humanos , Células Jurkat , Ratones , Ratones Endogámicos , Mucina-1/química , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Nature ; 545(7652): 98-102, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28445461

RESUMEN

The relative contribution of the effector molecules produced by T cells to tumour rejection is unclear, but interferon-γ (IFNγ) is critical in most of the analysed models. Although IFNγ can impede tumour growth by acting directly on cancer cells, it must also act on the tumour stroma for effective rejection of large, established tumours. However, which stroma cells respond to IFNγ and by which mechanism IFNγ contributes to tumour rejection through stromal targeting have remained unknown. Here we use a model of IFNγ induction and an IFNγ-GFP fusion protein in large, vascularized tumours growing in mice that express the IFNγ receptor exclusively in defined cell types. Responsiveness to IFNγ by myeloid cells and other haematopoietic cells, including T cells or fibroblasts, was not sufficient for IFNγ-induced tumour regression, whereas responsiveness of endothelial cells to IFNγ was necessary and sufficient. Intravital microscopy revealed IFNγ-induced regression of the tumour vasculature, resulting in arrest of blood flow and subsequent collapse of tumours, similar to non-haemorrhagic necrosis in ischaemia and unlike haemorrhagic necrosis induced by tumour necrosis factor. The early events of IFNγ-induced tumour ischaemia resemble non-apoptotic blood vessel regression during development, wound healing or IFNγ-mediated, pregnancy-induced remodelling of uterine arteries. A better mechanistic understanding of how solid tumours are rejected may aid the design of more effective protocols for adoptive T-cell therapy.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Hipoxia de la Célula/inmunología , Interferón gamma/inmunología , Isquemia/inmunología , Neoplasias/irrigación sanguínea , Neoplasias/inmunología , Remodelación Vascular , Animales , Vasos Sanguíneos/inmunología , Vasos Sanguíneos/metabolismo , Línea Celular Tumoral , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Femenino , Interferón gamma/biosíntesis , Microscopía Intravital , Isquemia/metabolismo , Isquemia/patología , Masculino , Ratones , Necrosis , Neoplasias/metabolismo , Neoplasias/patología , Receptores de Interferón/metabolismo , Células del Estroma/inmunología , Células del Estroma/metabolismo , Especificidad por Sustrato , Cicatrización de Heridas , Receptor de Interferón gamma
4.
Proc Natl Acad Sci U S A ; 117(26): 15148-15159, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32541028

RESUMEN

The potency of adoptive T cell therapies targeting the cell surface antigen CD19 has been demonstrated in hematopoietic cancers. It has been difficult to identify appropriate targets in nonhematopoietic tumors, but one class of antigens that have shown promise is aberrant O-glycoprotein epitopes. It has long been known that dysregulated synthesis of O-linked (threonine or serine) sugars occurs in many cancers, and that this can lead to the expression of cell surface proteins containing O-glycans comprised of a single N-acetylgalactosamine (GalNAc, known as Tn antigen) rather than the normally extended carbohydrate. Previously, we used the scFv fragment of antibody 237 as a chimeric antigen receptor (CAR) to mediate recognition of mouse tumor cells that bear its cognate Tn-glycopeptide epitope in podoplanin, also called OTS8. Guided by the structure of the 237 Fab:Tn-OTS8-glycopeptide complex, here we conducted a deep mutational scan showing that residues flanking the Tn-glycan contributed significant binding energy to the interaction. Design of 237-scFv libraries in the yeast display system allowed us to isolate scFv variants with higher affinity for Tn-OTS8. Selection with a noncognate human antigen, Tn-MUC1, yielded scFv variants that were broadly reactive with multiple Tn-glycoproteins. When configured as CARs, engineered T cells expressing these scFv variants showed improved activity against mouse and human cancer cell lines defective in O-linked glycosylation. This strategy provides CARs with Tn-peptide specificities, all based on a single scFv scaffold, that allows the same CAR to be tested for toxicity in mice and efficacy against mouse and human tumors.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/fisiología , Secuencia de Aminoácidos , Animales , Anticuerpos , Línea Celular Tumoral , Evolución Molecular Dirigida , Epítopos/genética , Humanos , Ratones , Modelos Moleculares , Mutación , Conformación Proteica , Receptores Quiméricos de Antígenos/genética
5.
Exp Cell Res ; 393(2): 112014, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32439494

RESUMEN

Understanding interactions between immune cells and their targets is an important step on the path to fully characterizing the immune system, and in doing so, learning how it combats disease. Many studies of these interactions have a narrow focus, often looking only at a binary result of whether or not a specific treatment was successful or only focusing on the interactions between two individual cells. Therefore, in an effort to more comprehensively study multicellular interactions among immune cells and their targets, we used in vitro longitudinal time-lapse imaging and developed an automated cell cluster analysis tool, or macro, to investigate the formation of cell clusters. In particular, we investigated the behavior of cancer-specific CD8+ and CD4+ T cells on how they interact around their targets: cancer cells and antigen-presenting cells. The macro that we established allowed us to examine these large-scale clustering behaviors taking place between those four cell types. Thus, we were able to distinguish directed immune cell clustering from random cell movement. Furthermore, this macro can be generalized to be applicable to systems consisting of any number of differently labeled species and can be used to track clustering behaviors and compare them to randomized simulations.


Asunto(s)
Comunicación Celular/fisiología , Técnicas de Cultivo de Célula , Movimiento Celular/fisiología , Linfocitos T/citología , Animales , Análisis por Conglomerados , Ratones Endogámicos C57BL
6.
Carcinogenesis ; 41(11): 1605-1615, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-32221533

RESUMEN

Human papillomavirus (HPV) infection is necessary but insufficient for progression of epithelial cells from dysplasia to carcinoma-in situ (CIS) to invasive cancer. The combination of mutant cellular and viral oncogenes that regulate progression of cervical cancer (CC) remains unclear. Using combinations of HPV16 E6/E7 (E+), mutant Kras (mKras) (K+) and/or loss of Pten (P-/-), we generated autochthonous models of CC without exogenous estrogen, carcinogen or promoters. Furthermore, intravaginal instillation of adenoCre virus enabled focal activation of the oncogenes/inactivation of the tumor suppressor gene. In P+/+ mice, E6/E7 alone (P+/+E+K-) failed to cause premalignant changes, while mKras alone (P+/+E-K+) caused persistent mucosal abnormalities in about one-third of mice, but no cancers. To develop cancer, P+/+ mice needed both E6/E7 and mKras expression. Longitudinal endoscopies of P+/+E+K+ mice predicted carcinoma development by detection of mucosal lesions, found on an average of 23 weeks prior to death, unlike longitudinal quantitative PCRs of vaginal lavage samples from the same mice. Endoscopy revealed that individual mice differed widely in the time required for mucosal lesions to appear after adenoCre and in the time required for these lesions to progress to cancer. These cancers developed in the transition zone that extends, unlike in women, from the murine cervix to the distal vagina. The P-/-E+K+ genotype led to precipitous cancer development within a few weeks and E6/E7-independent cancer development occurred in the P-/-E-K+ genotype. In the P-/-E+K- genotype, mice only developed CIS. Thus, distinct combinations of viral and cellular oncogenes are involved in distinct steps in cervical carcinogenesis.


Asunto(s)
Carcinógenos/toxicidad , Endoscopía/métodos , Estrógenos/toxicidad , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Proteínas Represoras/metabolismo , Neoplasias del Cuello Uterino/patología , Neoplasias Vaginales/patología , Animales , Carcinogénesis , Femenino , Papillomavirus Humano 16/aislamiento & purificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Fosfohidrolasa PTEN/fisiología , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/virología , Proteínas Proto-Oncogénicas p21(ras)/genética , Neoplasias del Cuello Uterino/diagnóstico por imagen , Neoplasias del Cuello Uterino/etiología , Neoplasias del Cuello Uterino/metabolismo , Neoplasias Vaginales/diagnóstico por imagen , Neoplasias Vaginales/etiología , Neoplasias Vaginales/metabolismo
7.
Glycobiology ; 29(4): 307-319, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30726901

RESUMEN

Successful application of potent antibody-based T-cell engaging immunotherapeutic strategies is currently limited mainly to hematological cancers. One major reason is the lack of well-characterized antigens on solid tumors with sufficient cancer specific expression. Aberrantly O-glycosylated proteins contain promising cancer-specific O-glycopeptide epitopes suitable for immunotherapeutic applications, but currently only few examples of such antibody epitopes have been identified. We previously showed that chimeric antigen receptor T-cells directed towards aberrantly O-glycosylated MUC1 can control malignant growth in a mouse model. Here, we present a discovery platform for the generation of cancer-specific monoclonal antibodies targeting aberrant O-glycoproteins. The strategy is based on cancer cell lines engineered to homogeneously express the truncated Tn O-glycoform, the so-called SimpleCells. We used SimpleCells of different cancer origin to elicit monoclonal antibodies with selectivity for aberrant O-glycoproteins. For validation we selected and characterized one monoclonal antibody (6C5) directed to a Tn-glycopeptide in dysadherin (FXYD5), known to be upregulated in cancer and promote metastasis. While dysadherin is widely expressed also in normal cells, we demonstrated that the 6C5 epitope is specifically expressed in cancer.


Asunto(s)
Anticuerpos Monoclonales/análisis , Anticuerpos Monoclonales/biosíntesis , Glicoproteínas/metabolismo , Neoplasias/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Línea Celular Tumoral , Epítopos/inmunología , Epítopos/metabolismo , Glicoproteínas/inmunología , Humanos , Ratones , Neoplasias/inmunología , Neoplasias/patología
8.
Proc Natl Acad Sci U S A ; 113(27): 7551-6, 2016 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-27317748

RESUMEN

Fibroblasts are common cell types in cancer stroma and lay down collagen required for survival and growth of cancer cells. Although some cancer therapy strategies target tumor fibroblasts, their origin remains controversial. Multiple publications suggest circulating mesenchymal precursors as a source of tumor-associated fibroblasts. However, we show by three independent approaches that tumor fibroblasts derive primarily from local, sessile precursors. First, transplantable tumors developing in a mouse expressing green fluorescent reporter protein (EGFP) under control of the type I collagen (Col-I) promoter (COL-EGFP) had green stroma, whereas we could not find COL-EGFP(+) cells in tumors developing in the parabiotic partner lacking the fluorescent reporter. Lack of incorporation of COL-EGFP(+) cells from the circulation into tumors was confirmed in parabiotic pairs of COL-EGFP mice and transgenic mice developing autochthonous intestinal adenomas. Second, transplantable tumors developing in chimeric mice reconstituted with bone marrow cells from COL-EGFP mice very rarely showed stromal fibroblasts expressing EGFP. Finally, cancer cells injected under full-thickness COL-EGFP skin grafts transplanted in nonreporter mice developed into tumors containing green stromal cells. Using multicolor in vivo confocal microscopy, we found that Col-I-expressing fibroblasts constituted approximately one-third of the stromal mass and formed a continuous sheet wrapping the tumor vessels. In summary, tumors form their fibroblastic stroma predominantly from precursors present in the local tumor microenvironment, whereas the contribution of bone marrow-derived circulating precursors is rare.


Asunto(s)
Fibroblastos Asociados al Cáncer/fisiología , Neoplasias Experimentales/patología , Actinas/metabolismo , Animales , Fibroblastos Asociados al Cáncer/patología , Línea Celular Tumoral , Colágeno Tipo I/metabolismo , Proteínas Fluorescentes Verdes , Ratones Endogámicos C57BL , Ratones Noqueados
9.
J Immunol ; 192(3): 1286-93, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24367029

RESUMEN

Myeloid-derived CD11b(+)Gr1(+) suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) are considered a major obstacle for effective adoptive T cell therapy. Myeloid cells suppress naive T cell proliferation ex vivo and can prevent the generation of T cell responses in vivo. We find, however, that adoptively transferred immune T cells eradicate well-established tumors in the presence of MDSCs and TAMs, which are strongly immunosuppressive ex vivo. These MDSCs and TAMs were comparable in numbers and immunosuppressive capacity among different tumor models. Longitudinal microscopy of tumors in vivo revealed that after T cell transfer, tumor vasculature and cancer cells disappeared simultaneously. During T cell-mediated tumor destruction, the tumor stroma contained abundant myeloid cells (mainly TAMs) that retained their suppressive properties. Preimmunized but not naive mice resisted immune suppression caused by an unrelated tumor burden, supporting the idea that in vivo, myeloid immunosuppressive cells can suppress naive but not memory T cell responses.


Asunto(s)
Inmunoterapia Adoptiva , Macrófagos/inmunología , Células Mieloides/inmunología , Neoplasias Experimentales/terapia , Subgrupos de Linfocitos T/trasplante , Escape del Tumor/inmunología , Animales , Antígeno CD11b/análisis , Vacunas contra el Cáncer/inmunología , Proteínas de Unión al ADN/deficiencia , Proteínas de Homeodominio/genética , Inmunización , Memoria Inmunológica , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Modelos Biológicos , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neovascularización Patológica/inmunología , Neovascularización Patológica/terapia , Receptores de Quimiocina/análisis , Técnica de Ventana Cutánea , Subgrupos de Linfocitos T/inmunología , Carga Tumoral
10.
Proc Natl Acad Sci U S A ; 110(20): 8158-63, 2013 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-23637340

RESUMEN

A major challenge of cancer immunotherapy is the persistence and outgrowth of subpopulations that lose expression of the target antigen. IL-15 is a potent cytokine that can promote organ-specific autoimmunity when up-regulated on tissue cells. Here we report that T cells eradicated 2-wk-old solid tumors that expressed IL-15, eliminating antigen-negative cells. In contrast, control tumors that lacked IL-15 expression consistently relapsed. Interestingly, even tumors lacking expression of cognate antigen were rejected when expressing IL-15, indicating that rejection after adoptive T-cell transfer was independent of cognate antigen expression. Nevertheless, the T-cell receptor of the transferred T cells influenced the outcome, consistent with the notion that T-cell receptor activation and effector status determine whether IL-15 can confer lymphokine killer activity-like properties to T cells. The effect was limited to the microenvironment of tumors expressing IL-15; there were no noticeable effects on contralateral tumors lacking IL-15. Taken together, these results indicate that expression of IL-15 in the tumor microenvironment may prevent the escape of antigen loss variants and subsequent tumor recurrence by enabling T cells to eliminate cancer cells lacking cognate antigen expression in a locally restricted manner.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Interleucina-15/metabolismo , Neoplasias/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Microambiente Tumoral , Animales , Antígenos de Neoplasias/metabolismo , Autoinmunidad , Linfocitos T CD8-positivos/citología , Línea Celular Tumoral , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Interleucina-15/genética , Células Asesinas Naturales/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Perforina/metabolismo , Bazo/citología , Células del Estroma/citología
11.
Semin Cancer Biol ; 22(1): 41-9, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22212863

RESUMEN

All cancers depend on stroma for support of growth. Leukemias, solid tumors, cancer cells causing effusions, metastases as well as micro-disseminated cancer cells release factors that stimulate stromal cells, which in turn produce ligands that stimulate cancer cells. Therefore, elimination of stromal support by destroying the stromal cells or by inhibiting feedback stimulation of cancer growth is in the focus of many evolving therapies. A stringent evaluation of the efficacy of stromal targeting requires testing in animal models. Most current studies emphasize the successes of stromal targeting rather than deciphering its limitations. Here we show that many of the stromal targeting approaches, while often reducing tumor growth rates, are rarely curative. Therefore, we will also discuss conditions where stromal targeting can eradicate large established tumors. Finally, we will examine still unanswered questions of this promising and exciting area of cancer research.


Asunto(s)
Neoplasias/terapia , Comunicación Paracrina/fisiología , Células del Estroma/fisiología , Progresión de la Enfermedad , Humanos , Neoplasias/fisiopatología
12.
Sci Immunol ; 9(99): eadp6529, 2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39270007

RESUMEN

Cancers eventually kill hosts even when infiltrated by cancer-specific T cells. We examined whether cancer-specific T cell receptors of CD4+ T cells (CD4TCRs) from tumor-bearing hosts can be exploited for adoptive TCR therapy. We focused on CD4TCRs targeting an autochthonous mutant neoantigen that is only presented by stroma surrounding the MHC class II-negative cancer cells. The 11 most common tetramer-sorted CD4TCRs were tested using TCR-engineered CD4+ T cells. Three TCRs were characterized by convergent recombination for which multiple T cell clonotypes differed in their nucleotide sequences but encoded identical TCR α and ß chains. These preferentially selected TCRs destroyed tumors equally well and halted progression through reprogramming of the tumor stroma. TCRs represented by single T cell clonotypes were similarly effective only if they shared CDR elements with preferentially selected TCRs in both α and ß chains. Selecting candidate TCRs on the basis of these characteristics can help identify TCRs that are potentially therapeutically effective.


Asunto(s)
Linfocitos T CD4-Positivos , Inmunoterapia Adoptiva , Linfocitos T CD4-Positivos/inmunología , Animales , Inmunoterapia Adoptiva/métodos , Ratones , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/genética , Ratones Endogámicos C57BL , Humanos , Ratones Transgénicos , Femenino , Recombinación Genética/inmunología
13.
Clin Cancer Res ; 30(8): 1642-1654, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38190111

RESUMEN

PURPOSE: To achieve eradication of solid tumors, we examined how many neoantigens need to be targeted with how many T-cell receptors (TCR) by which type of T cells. EXPERIMENTAL DESIGN: Unmanipulated, naturally expressed (autochthonous) neoantigens were targeted with adoptively transferred TCR-engineered autologous T cells (TCR-therapy). TCR-therapy used CD8+ T-cell subsets engineered with TCRs isolated from CD8+ T cells (CD8+TCR-therapy), CD4+ T-cell subsets engineered with TCRs isolated from CD4+ T cells (CD4+TCR-therapy), or combinations of both. The targeted tumors were established for at least 3 weeks and derived from primary autochthonous cancer cell cultures, resembling natural solid tumors and their heterogeneity as found in humans. RESULTS: Relapse was common with CD8+TCR-therapy even when targeting multiple different autochthonous neoantigens on heterogeneous solid tumors. CD8+TCR-therapy was only effective against homogenous tumors artificially derived from a cancer cell clone. In contrast, a combination of CD8+TCR-therapy with CD4+TCR-therapy, each targeting one neoantigen, eradicated large and established solid tumors of natural heterogeneity. CD4+TCR-therapy targeted a mutant neoantigen on tumor stroma while direct cancer cell recognition by CD8+TCR-therapy was essential for cure. In vitro data were consistent with elimination of cancer cells requiring a four-cell cluster composed of TCR-engineered CD4+ and CD8+ T cells together with antigen-presenting cells and cancer cells. CONCLUSIONS: Two cancer-specific TCRs can be essential and sufficient to eradicate heterogeneous solid tumors expressing unmanipulated, autochthonous targets. We demonstrate that simplifications to adoptive TCR-therapy are possible without compromising efficacy.


Asunto(s)
Antígenos de Neoplasias , Neoplasias , Humanos , Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Neoplasias/inmunología , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/genética , Inmunoterapia Adoptiva/métodos
14.
Cancer Gene Ther ; 31(4): 574-585, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38267626

RESUMEN

Aberrant TGFß signaling is linked to metastasis and tumor immune escape of many cancers including metastatic triple negative breast cancer (mTNBC). Previously, we have found that oncolytic adenoviruses expressing a TGFß signaling inhibitory protein (sTGFßRIIFc) induced immune activation in a mouse TNBC (4T1) immunocompetent subcutaneous model with intratumoral injection. Systemic administration of adenoviruses can be a superior route to treat mTNBC but faces the challenges of increased toxicity and viral clearance. Thus, we created a liver-de-targeted sTGFßRIIFc- and LyP-1 peptide-expressing adenovirus (mHAdLyp.sT) with enhanced breast cancer cell tropism. Its safety and immune response features were profiled in the 4T1 model. Our data showed that the systemic administration of mHAdLyp.sT resulted in reduced hepatic and systemic toxicity. mHAdLyp.sT was also effective in increasing Th1 cytokines and anti-tumor cell populations by cytokine analysis, spleen/tumor qRT-PCR, and flow cytometry. We further tested the therapeutic effects of mHAdLyp.sT alone and in combination with immune checkpoint inhibitors (ICIs). mHAdLyp.sT alone and with all ICI combinations elicited significant inhibition of lung metastasis by histological analysis. When mHAdLyp.sT was combined with both anti-PD-1 and anti-CTLA-4 antibodies, primary 4T1 tumor growth was also significantly inhibited. We are confident in advancing this new treatment option for mTNBC.


Asunto(s)
Infecciones por Adenoviridae , Neoplasias Mamarias Animales , Neoplasias de la Mama Triple Negativas , Ratones , Animales , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Adenoviridae/metabolismo , Transducción de Señal , Citocinas/metabolismo , Hígado/patología , Neoplasias de la Mama Triple Negativas/terapia , Neoplasias de la Mama Triple Negativas/patología , Línea Celular Tumoral
15.
J Exp Med ; 204(1): 49-55, 2007 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-17210731

RESUMEN

Targeting cancer cells, as well as the nonmalignant stromal cells cross-presenting the tumor antigen (Ag), can lead to the complete destruction of well-established solid tumors by adoptively transferred Ag-specific cytotoxic T lymphocytes (CTLs). If, however, cancer cells express only low levels of the Ag, then stromal cells are not destroyed, and the tumor escapes as Ag loss variants. We show that treating well-established tumors expressing low levels of Ag with local irradiation or a chemotherapeutic drug causes sufficient release of Ag to sensitize stromal cells for destruction by CTLs. This was shown directly using high affinity T cell receptor tetramers for visualizing the transient appearance of tumor-specific peptide-MHC complexes on stromal cells. Maximum loading of tumor stroma with cancer Ag occurred 2 d after treatment and coincided with the optimal time for T cell transfer. Under these conditions, tumor rejection was complete. These findings may set the stage for developing rational clinical protocols for combining irradiation or chemotherapy with CTL therapy.


Asunto(s)
Neoplasias Experimentales/inmunología , Células del Estroma/inmunología , Linfocitos T Citotóxicos/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno , Células Presentadoras de Antígenos/inmunología , Antígenos de Neoplasias , Antineoplásicos/farmacología , Línea Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Inmunización , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/radioterapia , Receptores de Antígenos de Linfocitos T/metabolismo , Gemcitabina
16.
Cancer Immunol Immunother ; 62(2): 359-69, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22926060

RESUMEN

Clinical studies with immunotherapies for cancer, including adoptive cell transfers of T cells, have shown promising results. It is now widely believed that recruitment of CD4(+) helper T cells to the tumor would be favorable, as CD4(+) cells play a pivotal role in cytokine secretion as well as promoting the survival, proliferation, and effector functions of tumor-specific CD8(+) cytotoxic T lymphocytes. Genetically engineered high-affinity T-cell receptors (TCRs) can be introduced into CD4(+) helper T cells to redirect them to recognize MHC-class I-restricted antigens, but it is not clear what affinity of the TCR will be optimal in this approach. Here, we show that CD4(+) T cells expressing a high-affinity TCR (nanomolar K (d) value) against a class I tumor antigen mediated more effective tumor treatment than the wild-type affinity TCR (micromolar K (d) value). High-affinity TCRs in CD4(+) cells resulted in enhanced survival and long-term persistence of effector memory T cells in a melanoma tumor model. The results suggest that TCRs with nanomolar affinity could be advantageous for tumor targeting when expressed in CD4(+) T cells.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/inmunología , Genes MHC Clase I/inmunología , Melanoma Experimental/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Neoplasias Cutáneas/inmunología , Traslado Adoptivo , Animales , Antineoplásicos/uso terapéutico , Linfocitos T CD4-Positivos/química , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Supervivencia Celular/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Interferón gamma/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/biosíntesis , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/genética
17.
Mol Ther ; 20(3): 652-60, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22233579

RESUMEN

Most T cells have T cell receptors (TCR) of micromolar affinity for peptide-major histocompatibility complex (MHC) ligands, but genetic engineering can generate TCRs of nanomolar affinity. The affinity of the TCR used, m33, for its cognate non-self peptide-MHC-I complex (SIYRYYGL-K(b)) is 1,000-fold higher than of the wild-type TCR 2C. The affinity of m33 for the self-peptide dEV-8 on K(b) is only twofold higher. Mouse CD8(+) T cells transduced with an m33-encoding retrovirus showed binding of SIY-K(b) and potent function in vitro, but in vivo these T cells disappeared within hours after transfer into syngeneic hosts without causing graft-versus-host disease (GVHD). Accordingly, in cases where such CD8-dependent self-reactivity might occur in human adoptive T cell therapies, our results show that a peripheral T-cell deletion mechanism could operate to avoid reactions with the host. In contrast to CD8(+) T cells, we show that CD4(+) T cells expressing m33 survived for months in vivo. Furthermore, the m33-transduced CD4(+) T cells were able to mediate antigen-specific rejection of 6-day-old tumors. Together, we show that CD8(+) T cell expressing a MHC class I-restricted high-affinity TCR were rapidly deleted whereas CD4(+) T cells expressing the same TCR survived and provided function while being directed against a class I-restricted antigen.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Línea Celular , Supervivencia Celular/inmunología , Expresión Génica , Vectores Genéticos/genética , Inmunoterapia Adoptiva , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/terapia , Oligopéptidos/inmunología , Péptidos/química , Péptidos/inmunología , Retroviridae/genética , Transducción Genética
18.
Proc Natl Acad Sci U S A ; 107(22): 10056-61, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20479270

RESUMEN

Aberrant glycosylation and the overexpression of certain carbohydrate moieties is a consistent feature of cancers, and tumor-associated oligosaccharides are actively investigated as targets for immunotherapy. One of the most common aberrations in glycosylation patterns is the presentation of a single O-linked N-acetylgalactosamine on a threonine or serine residue known as the "Tn antigen." Whereas the ubiquitous nature of Tn antigens on cancers has made them a natural focus of vaccine research, such carbohydrate moieties are not always tumor-specific and have been observed on embryonic and nonmalignant adult tissue. Here we report the structural basis of binding of a complex of a monoclonal antibody (237mAb) with a truly tumor-specific glycopeptide containing the Tn antigen. In contrast to glycopeptide-specific antibodies in complex with simple peptides, 237mAb does not recognize a conformational epitope induced in the peptide by sugar substitution. Instead, 237mAb uses a pocket coded by germ-line genes to completely envelope the carbohydrate moiety itself while interacting with the peptide moiety in a shallow groove. Thus, 237mAb achieves its striking tumor specificity, with no observed physiological cross-reactivity to the unglycosylated peptide or the free glycan, by a combination of multiple weak but specific interactions to both the peptide and to the glycan portions of the antigen.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/química , Animales , Anticuerpos Monoclonales , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Complejo Antígeno-Anticuerpo/química , Cristalografía por Rayos X , Epítopos/química , Glicopéptidos/química , Glicopéptidos/inmunología , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Técnicas In Vitro , Ratones , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Electricidad Estática , Resonancia por Plasmón de Superficie
19.
Res Sq ; 2023 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-37790556

RESUMEN

Aberrant TGFß signaling is linked to metastasis and tumor immune escape of many cancers including metastatic triple negative breast cancer (mTNBC). Previously, we have found that oncolytic adenoviruses expressing a TGFß signaling inhibitory protein (sTGFßRIIFc) induced immune activation in a mouse TNBC (4T1) immunocompetent subcutaneous model with intratumoral injection. Systemic administration of adenoviruses can be a superior route to treat mTNBC but faces the challenges of increased toxicity and viral clearance. Thus, we created a liver-de-targeted sTGFßRIIFc- and LyP-1 peptide-expressing adenovirus (mHAdLyp.sT) with enhanced breast cancer cell tropism. Its safety and immune response features were profiled in the 4T1 model. Our data showed that the systemic administration of mHAdLyp.sT resulted in reduced hepatic and systemic toxicity. mHAdLyp.sT was also effective in increasing Th1 cytokines and anti-tumor cell populations by cytokine analysis, spleen/tumor qRT-PCR, and flow cytometry. We further tested the therapeutic effects of mHAdLyp.sT alone and in combination with immune checkpoint inhibitors (ICIs). mHAdLyp.sT alone and with all ICI combinations elicited significant inhibition of lung metastasis by histological analysis. When mHAdLyp.sT was combined with both anti-PD-1 and anti-CTLA-4 antibodies, primary 4T1 tumor growth was also significantly inhibited. We are confident in advancing this new treatment option for mTNBC.

20.
J Immunother Cancer ; 11(5)2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37258040

RESUMEN

BACKGROUND: Treatment of some blood cancers with T cells that express a chimeric antigen receptor (CAR) against CD19 have shown remarkable results. In contrast, CAR-T cell efficacy against solid tumors has been difficult to achieve. METHODS: To examine the potential of CAR-T cell treatments against ovarian cancers, we used the mouse ovarian cancer cell line ID8 in an intraperitoneal model that exhibits disseminated solid tumors in female C57BL/6J mice. The CAR contained a single-chain Fv from antibody 237 which recognizes a Tn-glycopeptide-antigen expressed by ID8 due to aberrant O-linked glycosylation in the absence of the transferase-dependent chaperone Cosmc. The efficacy of four Tn-dependent CARs with varying affinity to Tn antigen, and each containing CD28/CD3ζ cytoplasmic domains, were compared in vitro and in vivo in this study. RESULTS: In line with many observations about the impact of aberrant O-linked glycosylation, the ID8Cosmc knock-out (ID8Cosmc-KO) exhibited more rapid tumor progression compared with wild-type ID8. Despite the enhanced tumor growth in vivo, 237 CAR and a mutant with 30-fold higher affinity, but not CARs with lower affinity, controlled advanced ID8Cosmc-KO tumors. Tumor regression could be achieved with a single intravenous dose of the CARs, but intraperitoneal administration was even more effective. The CAR-T cells persisted over a period of months, allowing CAR-treated mice to delay tumor growth in a re-challenge setting. The most effective CARs exhibited the highest affinity for antigen. Antitumor effects observed in vivo were associated with increased numbers of T cells and macrophages, and higher levels of cleaved caspase-3, in the tumor microenvironment. Notably, the least therapeutically effective CAR mediated tonic signaling leading to antigen-independent cytokine expression and it had higher levels of the immunosuppressive cytokine interleukin10. CONCLUSION: The findings support the development of affinity-optimized CAR-T cells as a potential treatment for established ovarian cancer, with the most effective CARs mediating a distinct pattern of inflammatory cytokine release in vitro. Importantly, the most potent Tn-dependent CAR-T cells showed no evidence of toxicity in tumor-bearing mice in a syngeneic, immunocompetent system.


Asunto(s)
Neoplasias Ováricas , Receptores Quiméricos de Antígenos , Humanos , Femenino , Ratones , Animales , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T , Inmunoterapia Adoptiva/métodos , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Endogámicos C57BL , Citocinas/metabolismo , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA