Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Acta Neuropathol ; 134(2): 297-316, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28332095

RESUMEN

Glioblastoma (GBM) is a highly aggressive brain tumor and still remains incurable. Among others, an immature subpopulation of self-renewing and therapy-resistant tumor cells-often referred to as glioblastoma stem-like cells (GSCs)-has been shown to contribute to disease recurrence. To target these cells personalized immunotherapy has gained a lot of interest, e.g. by reactivating pre-existing anti-tumor immune responses against GSC antigens. To identify T cell targets commonly presented by GSCs and their differentiated counterpart, we used a proteomics-based separation of GSC proteins in combination with a T cell activation assay. Altogether, 713 proteins were identified by LC-ESI-MS/MS mass spectrometry. After a thorough filtering process, 32 proteins were chosen for further analyses. Immunogenicity of corresponding peptides was tested ex vivo. A considerable number of these antigens induced T cell responses in GBM patients but not in healthy donors. Moreover, most of them were overexpressed in primary GBM and also highly expressed in recurrent GBM tissues. Interestingly, expression of the most frequent T cell target antigens could also be confirmed in quiescent, slow-cycling GSCs isolated in high purity by the DEPArray technology. Finally, for a subset of these T cell target antigens, an association between expression levels and higher T cell infiltration as well as an increased expression of positive immune modulators was observed. In summary, we identified novel immunogenic proteins, which frequently induce tumor-specific T cell responses in GBM patients and were also detected in vitro in therapy-resistant quiescent, slow-cycling GSCs. Stable expression of these T cell targets in primary and recurrent GBM support their suitability for future clinical use.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Neoplasias Encefálicas/patología , Glioblastoma/patología , Proteómica , Subgrupos de Linfocitos T/patología , Animales , Anexina A1/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Pruebas de Carcinogenicidad , Proteínas Portadoras/metabolismo , Células Cultivadas , Chaperonina 60/metabolismo , Cistatina A/metabolismo , Modelos Animales de Enfermedad , Mapeo Epitopo , Femenino , Humanos , Interferón gamma/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Ratones , Proteínas de Microfilamentos/metabolismo , Proteínas Mitocondriales/metabolismo , Células Madre Neoplásicas/patología
2.
J Immunol ; 189(6): 3249-59, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22875801

RESUMEN

Prostate cancer is the most common noncutaneous malignancy in men. The prostate stem cell Ag (PSCA) is a promising target for immunotherapy of advanced disease. Based on a novel mAb directed to PSCA, we established and compared a series of murine and humanized anti-CD3-anti-PSCA single-chain bispecific Abs. Their capability to redirect T cells for killing of tumor cells was analyzed. During these studies, we identified a novel bispecific humanized Ab that efficiently retargets T cells to tumor cells in a strictly Ag-dependent manner and at femtomolar concentrations. T cell activation, cytokine release, and lysis of target cells depend on a cross-linkage of redirected T cells with tumor cells, whereas binding of the anti-CD3 domain alone does not lead to an activation or cytokine release. Interestingly, both CD8+ and CD4+ T cells are activated in parallel and can efficiently mediate the lysis of tumor cells. However, the onset of killing via CD4+ T cells is delayed. Furthermore, redirecting T cells via the novel humanized bispecific Abs results in a delay of tumor growth in xenografted nude mice.


Asunto(s)
Anticuerpos Biespecíficos/fisiología , Anticuerpos Monoclonales Humanizados/fisiología , Antígenos de Neoplasias/biosíntesis , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proteínas de Neoplasias/biosíntesis , Neoplasias de la Próstata/inmunología , Células Madre/inmunología , Ensayo de Tumor de Célula Madre , Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Muerte Celular/inmunología , Epítopos de Linfocito T/inmunología , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/biosíntesis , Proteínas Ligadas a GPI/inmunología , Humanos , Masculino , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/inmunología , Neoplasias de la Próstata/patología , Células Madre/patología , Ensayo de Tumor de Célula Madre/métodos
3.
J Immunol ; 188(3): 1551-8, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22184723

RESUMEN

Bispecific Abs hold great potential for immunotherapy of malignant diseases. Because the first components of this new drug class are now entering clinical trials, all aspects of their mode of action should be well understood. Several studies proved that CD8(+) and CD4(+) effector T cells can be successfully redirected and activated against tumor cells by bispecific Abs both in vitro and in vivo. To our knowledge, this study provides the first evidence that bispecific Abs can also redirect and activate regulatory T cells against a surface Ag, independently of their TCR specificity. After cross-linking, via a bispecific Ab, redirected regulatory T cells upregulate the activation markers CD69 and CD25, as well as regulatory T cell-associated markers, like CTLA-4 and FOXP3. The activated regulatory T cells secrete the immunosuppressive cytokine IL-10, but, in contrast to CD8(+) and CD4(+) effector T cells, almost no inflammatory cytokines. In addition, the redirected regulatory T cells are able to suppress effector functions of activated autologous CD4(+) T cells both in vitro and in vivo. Therefore, the potential risk for activation of regulatory T cells should be taken into consideration when bispecific Abs are applied for the treatment of malignant diseases. In contrast, an Ag/tissue-specific redirection of regulatory T cells with bispecific Abs holds great potential for the treatment of autoimmune diseases and graft rejection.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Terapia Molecular Dirigida/métodos , Linfocitos T Reguladores/efectos de los fármacos , Anticuerpos Biespecíficos/farmacología , Antígenos de Superficie/efectos de los fármacos , Línea Celular , Humanos , Interleucina-10 , Activación de Linfocitos
4.
Cancer Cell ; 42(6): 938-941, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38861930

RESUMEN

Cancer immunotherapy is a pillar of clinical oncology but only achieves long-term remissions in a minority of cases. In this issue, van Elsas et al. show that effective immunotherapy requires a series of processes orchestrated by CD8+ T cells that result in the recruitment and local activation of M1-like macrophages.


Asunto(s)
Linfocitos T CD8-positivos , Inmunoterapia , Macrófagos , Linfocitos T CD8-positivos/inmunología , Humanos , Inmunoterapia/métodos , Macrófagos/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Animales , Microambiente Tumoral/inmunología
5.
J Autoimmun ; 42: 105-16, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23352111

RESUMEN

The nuclear autoantigen La can be detected on the surface of dying cells. Here we present an assay which enables us to show that La protein is not limited to the surface of dying cells but will be released upon stress-induced cell death. As released La protein tightly binds to the surface of neighboring intact cells we asked the question whether or not La protein could serve as a stress-inducible target e.g. for redirecting of regulatory T cells (Tregs) into damaged tissues to downregulate an immune response. In order to provide first proof of concept we developed a novel fully humanized single-chain bispecific antibody (bsAb) which on the one hand is directed to the La antigen and on the other hand to the CD3 complex of T cells. A cross-linkage of Tregs with La-decorated target cells mediated by this bsAb resulted indeed in the activation of the Tregs in a target-dependent manner. Moreover, such bsAb activated Tregs displayed a potent suppressive capacity and negatively influenced proliferation, expansion and cytokine production of autologous CD4(+) and CD8(+) Teff cells.


Asunto(s)
Autoantígenos/inmunología , Complejo CD3/inmunología , Terapia de Inmunosupresión , Proteínas de la Membrana/inmunología , Ribonucleoproteínas/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Células 3T3 , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Autoantígenos/genética , Complejo CD3/genética , Muerte Celular/efectos de los fármacos , Muerte Celular/inmunología , Células HEK293 , Células HeLa , Humanos , Activación de Linfocitos/efectos de los fármacos , Proteínas de la Membrana/genética , Ratones , Receptor Cross-Talk/inmunología , Ribonucleoproteínas/genética , Anticuerpos de Cadena Única/inmunología , Estrés Fisiológico/inmunología , Linfocitos T Reguladores/inmunología , Antígeno SS-B
6.
Acta Neuropathol Commun ; 11(1): 75, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-37158962

RESUMEN

Glioblastoma (GB) IDH-wildtype is the most malignant primary brain tumor. It is particularly resistant to current immunotherapies. Translocator protein 18 kDa (TSPO) is upregulated in GB and correlates with malignancy and poor prognosis, but also with increased immune infiltration. Here, we studied the role of TSPO in the regulation of immune resistance of human GB cells. The role of TSPO in tumor immune resistance was experimentally determined in primary brain tumor initiating cells (BTICs) and cell lines through genetic manipulation of TSPO expression and subsequent cocultures with antigen specific cytotoxic T cells and autologous tumor-infiltrating T cells. Death inducing intrinsic and extrinsic apoptotic pathways affected by TSPO were investigated. TSPO-regulated genes mediating apoptosis resistance in BTICs were identified through gene expression analysis and subsequent functional analyses. TSPO transcription in primary GB cells correlated with CD8+ T cell infiltration, cytotoxic activity of T cell infiltrate, expression of TNFR and IFNGR and with the activity of their downstream signalling pathways, as well as with the expression of TRAIL receptors. Coculture of BTICs with tumor reactive cytotoxic T cells or with T cell-derived factors induced TSPO up-regulation through T cell derived TNFα and IFNγ. Silencing of TSPO sensitized BTICs against T cell-mediated cytotoxicity. TSPO selectively protected BTICs against TRAIL-induced apoptosis by regulating apoptosis pathways. TSPO also regulated the expression of multiple genes associated with resistance against apoptosis. We conclude that TSPO expression in GB is induced through T cell-derived cytokines TNFα and IFNγ and that TSPO expression protects GB cells against cytotoxic T cell attack through TRAIL. Our data thereby provide an indication that therapeutic targeting of TSPO may be a suitable approach to sensitize GB to immune cell-mediated cytotoxicity by circumventing tumor intrinsic TRAIL resistance.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/genética , Factor de Necrosis Tumoral alfa , Encéfalo , Linfocitos T CD8-positivos , Neoplasias Encefálicas/genética , Receptores de GABA/genética
7.
Anal Biochem ; 423(2): 261-8, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22274538

RESUMEN

There is growing interest in the development of novel single-chain bispecific antibodies for retargeting of immune effector T cells to tumor cells. Until today, functional fusion constructs consisting of a single-chain bispecific antibody and a fluorescent protein were not reported. Such molecules could be useful for an in vivo visualization of this retargeting process. Recently, we established two novel single-chain bispecific antibodies. One is capable of retargeting T cells to CD33, and the other is capable of retargeting T cells to the prostate stem cell antigen (PSCA). CD33 is an attractive immunotarget on the surface of tumor cells from patients with acute myeloid leukemia (AML). The PSCA is a potential target on prostate cancer cells. Flanking the reading frame encoding the green fluorescent protein (GFP) with a recently described novel helical linker element allowed us to establish novel single-chain bispecific fusion antibodies. These fluorescent fusion antibodies were useful to efficiently retarget T cells to the respective tumor cells and visualize the formation of immune synapses between effector and target cells.


Asunto(s)
Anticuerpos Biespecíficos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Sinapsis Inmunológicas/patología , Microscopía Confocal , Linfocitos T/metabolismo , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/inmunología , Complejo CD3/inmunología , Línea Celular Tumoral , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Humanos , Sinapsis Inmunológicas/metabolismo , Antígeno Prostático Específico/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Lectina 3 Similar a Ig de Unión al Ácido Siálico , Linfocitos T/inmunología
8.
J Immunother Cancer ; 10(5)2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35606086

RESUMEN

BACKGROUND: Cancer immunotherapeutic strategies showed unprecedented results in the clinic. However, many patients do not respond to immuno-oncological treatments due to the occurrence of a plethora of immunological obstacles, including tumor intrinsic mechanisms of resistance to cytotoxic T-cell (TC) attack. Thus, a deeper understanding of these mechanisms is needed to develop successful immunotherapies. METHODS: To identify novel genes that protect tumor cells from effective TC-mediated cytotoxicity, we performed a genetic screening in pancreatic cancer cells challenged with tumor-infiltrating lymphocytes and antigen-specific TCs. RESULTS: The screening revealed 108 potential genes that protected tumor cells from TC attack. Among them, salt-inducible kinase 3 (SIK3) was one of the strongest hits identified in the screening. Both genetic and pharmacological inhibitions of SIK3 in tumor cells dramatically increased TC-mediated cytotoxicity in several in vitro coculture models, using different sources of tumor and TCs. Consistently, adoptive TC transfer of TILs led to tumor growth inhibition of SIK3-depleted cancer cells in vivo. Mechanistic analysis revealed that SIK3 rendered tumor cells susceptible to tumor necrosis factor (TNF) secreted by tumor-activated TCs. SIK3 promoted nuclear factor kappa B (NF-κB) nuclear translocation and inhibited caspase-8 and caspase-9 after TNF stimulation. Chromatin accessibility and transcriptome analyses showed that SIK3 knockdown profoundly impaired the expression of prosurvival genes under the TNF-NF-κB axis. TNF stimulation led to SIK3-dependent phosphorylation of the NF-κB upstream regulators inhibitory-κB kinase and NF-kappa-B inhibitor alpha on the one side, and to inhibition of histone deacetylase 4 on the other side, thus sustaining NF-κB activation and nuclear stabilization. A SIK3-dependent gene signature of TNF-mediated NF-κB activation was found in a majority of pancreatic cancers where it correlated with increased cytotoxic TC activity and poor prognosis. CONCLUSION: Our data reveal an abundant molecular mechanism that protects tumor cells from cytotoxic TC attack and demonstrate that pharmacological inhibition of this pathway is feasible.


Asunto(s)
FN-kappa B , Factor de Necrosis Tumoral alfa , Apoptosis , Humanos , FN-kappa B/metabolismo , Fosforilación , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
9.
Oncoimmunology ; 11(1): 2008110, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35141051

RESUMEN

Carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6), a cell surface receptor, is expressed on normal epithelial tissue and highly expressed in cancers of high unmet medical need, such as non-small cell lung, pancreatic, and colorectal cancer. CEACAM receptors undergo homo- and heterophilic interactions thereby regulating normal tissue homeostasis and angiogenesis, and in cancer, tumor invasion and metastasis. CEACAM6 expression on malignant plasma cells inhibits antitumor activity of T cells, and we hypothesize a similar function on epithelial cancer cells. The interactions between CEACAM6 and its suggested partner CEACAM1 on T cells were studied. A humanized CEACAM6-blocking antibody, BAY 1834942, was developed and characterized for its immunomodulating effects in co-culture experiments with T cells and solid cancer cells and in comparison to antibodies targeting the immune checkpoints programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), and T cell immunoglobulin mucin-3 (TIM-3). The immunosuppressive activity of CEACAM6 was mediated by binding to CEACAM1 expressed by activated tumor-specific T cells. BAY 1834942 increased cytokine secretion by T cells and T cell-mediated killing of cancer cells. The in vitro efficacy of BAY 1834942 correlated with the degree of CEACAM6 expression on cancer cells, suggesting potential in guiding patient selection. BAY 1834942 was equally or more efficacious compared to blockade of PD-L1, and at least an additive efficacy was observed in combination with anti-PD-1 or anti-TIM-3 antibodies, suggesting an efficacy independent of the PD-1/PD-L1 axis. In summary, CEACAM6 blockade by BAY 1834942 reactivates the antitumor response of T cells. This warrants clinical evaluation.


Asunto(s)
Antígenos CD , Neoplasias , Receptor de Muerte Celular Programada 1 , Antígenos CD/inmunología , Antígeno B7-H1/inmunología , Moléculas de Adhesión Celular/inmunología , Proteínas Ligadas a GPI/inmunología , Humanos , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T
10.
Prostate ; 71(9): 998-1011, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21541976

RESUMEN

BACKGROUND: Prostate cancer (PCa) is the most common malignant disease in men. Novel treatment options are needed for patients after development of metastatic, hormone-refractory disease or for those who have failed a local treatment. The prostate stem cell antigen (PSCA) is expressed in >80% of primary PCa samples and bone metastases. Its expression is increased both in androgen-dependent and independent prostate tumors, particularly in carcinomas of high stages and Gleason scores. Therefore, PSCA is an attractive target for immunotherapy of PCa by retargeting of T cells to tumor cells. METHODS: A series of different bispecific antibody formats for retargeting of T cells to tumor cells were described but, only very limited data obtained by side by side comparison of the different antibody formats are available. We established two novel bispecific antibodies in different formats. The functionality of both constructs was analyzed by FACS and chromium release assays. In parallel, the release of pro-inflammatory cytokines was determined by ELISA. RESULTS AND CONCLUSIONS: Irrespective of the underlying antibody format, both novel bispecific antibodies cause an efficient killing of PSCA-positive tumor cells by pre- and non-pre-activated T cells. Killing and release of pro-inflammatory cytokines requires an antigen specific cross-linkage of the T cells with the target cells.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Antígenos de Neoplasias/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias de la Próstata/inmunología , Linfocitos T/inmunología , Anticuerpos Biespecíficos/inmunología , Línea Celular Tumoral , Citocinas/análisis , Pruebas Inmunológicas de Citotoxicidad , Citometría de Flujo , Proteínas Ligadas a GPI/inmunología , Humanos , Inmunización Pasiva/métodos , Masculino , Neoplasias Hormono-Dependientes/inmunología , Neoplasias Hormono-Dependientes/terapia , Neoplasias de la Próstata/terapia , ARN/química , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
J Immunol Methods ; 496: 113086, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34146580

RESUMEN

Rare subpopulations of tumor antigen-reactive memory T cells, which actively secrete type-1 effector cytokines, particularly TNF-α in situ, possess anti-tumor activity and prognostic relevance. These cells are relevant for cancer immunotherapy; however, their low frequencies make them difficult to study and novel protocols for their culture and expansion ex vivo are needed. Here, we studied the presence of T cells secreting type-1 cytokines (Cy+T cells) in the blood and tumors of 24 patients with oral squamous cell carcinomas (OSCC) and explored possibilities for their isolation and expansion. More than 90% of OSCC patients contained enriched numbers Cy+T cells in the blood and tumors compared to healthy donors in which these were hardly detectable. The majority of TNF-α+T cells were CD4+ T helper cells while IFN-γ+TIL were predominantly CD8+. Cy+T helper cells in the blood were early-differentiated memory T cells while Cy+TIL and Cy+CD8+T cells showed advanced-differentiated memory T cell phenotypes. We explored different conditions for their in vitro culture and found that Cy+T cells can be efficiently expanded in vitro to similar levels as Cy-T cells and after expansion maintained their TNF-α secreting capacity. However, for optimal expansion they required specific culture conditions to support the maintenance of stem-like and central memory T cell phenotype. In conclusion, we show that Cy+T cells are enriched in OSCC patients and report a novel cell culture protocol optimized to specifically expand and functionally maintain these cells for further functional characterization or for their exploitation in immunotherapy of OSCC.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Interferón gamma/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias de la Boca/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Adulto , Anciano , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/metabolismo , Separación Celular , Células Cultivadas , Femenino , Humanos , Memoria Inmunológica , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/sangre , Neoplasias de la Boca/patología , Fenotipo , Carcinoma de Células Escamosas de Cabeza y Cuello/sangre , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Linfocitos T Colaboradores-Inductores/metabolismo
12.
J Biomed Biotechnol ; 2010: 956304, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20467460

RESUMEN

CD4+ and CD8+ T lymphocytes are powerful components of adaptive immunity, which essentially contribute to the elimination of tumors. Due to their cytotoxic capacity, T cells emerged as attractive candidates for specific immunotherapy of cancer. A promising approach is the genetic modification of T cells with chimeric antigen receptors (CARs). First generation CARs consist of a binding moiety specifically recognizing a tumor cell surface antigen and a lymphocyte activating signaling chain. The CAR-mediated recognition induces cytokine production and tumor-directed cytotoxicity of T cells. Second and third generation CARs include signal sequences from various costimulatory molecules resulting in enhanced T-cell persistence and sustained antitumor reaction. Clinical trials revealed that the adoptive transfer of T cells engineered with first generation CARs represents a feasible concept for the induction of clinical responses in some tumor patients. However, further improvement is required, which may be achieved by second or third generation CAR-engrafted T cells.


Asunto(s)
Antígenos de Neoplasias , Inmunoterapia/métodos , Neoplasias/terapia , Ingeniería de Proteínas/métodos , Receptores de Antígenos de Linfocitos T , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Humanos , Ratones , Modelos Inmunológicos , Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/inmunología
13.
Oncoimmunology ; 8(12): e1671762, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31741774

RESUMEN

The efficacy of cancer immunotherapy may be improved by increasing the number of circulating tumor-reactive T cells. The bone marrow is a priming site and reservoir for such T cells. The characteristics of bone marrow-derived tumor-reactive T cells are poorly understood in patients with non-small-cell lung cancer (NSCLC). To compare the responsiveness of tumor antigen-reactive T cells from the bone marrow with matched peripheral blood samples in patients with resectable NSCLC, we used flow cytometry, cytokine capture assays and enzyme-linked immunospot assays to examine the responsiveness of T cells to 14 tumor antigens in matched bone marrow and peripheral blood samples from patients with resectable NSCLC or benign tumors and tumor-free patients. T cells with reactivity to tumor antigens were detected in the bone marrow of 20 of 39 (51%) NSCLC patients. The panel of tumor antigens recognized by bone marrow-derived T cells was distinct from that recognized by peripheral blood-derived T cells in NSCLC patients. Unlike for peripheral blood T cells, the presence of tumor-reactive T cells in the bone marrow did not correlate with recurrence-free survival after curative intent resection of NSCLC. T cells with reactivity to tumor antigens are common in the bone marrow of patients with NSCLC. Tumor-reactive T cells of the bone marrow have the potential to significantly broaden the total repertoire of tumor-reactive T cells in the body. To clarify the role of tumor-reactive T cells of the bone marrow in T cell-based immunotherapy approaches, clinical studies are needed (ClinicalTrials.gov: NCT02515760).

14.
Clin Cancer Res ; 24(12): 2951-2962, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29563135

RESUMEN

Purpose: Successful immunotherapies for IDHmut gliomas require better knowledge of T-cell target antigens. Here, we elucidated their antigen repertoire recognized by spontaneous T-cell responses using an unbiased proteomic approach.Experimental Design: Protein fractionations of tissue lysates from IDHmut gliomas (n = 4) were performed. Fractions were tested by IFNγ ELISpot assay for recognition through patients' T cells. Proteins of immunogenic fractions were identified by mass spectrometry and validated by in silico-predicted synthetic long peptides in patients of origin, additional IDHmut glioma patients (n = 16), and healthy donors (n = 13). mRNA and protein expression of immunogenic antigens was analyzed in tumor tissues and IDHmut glioma stem-like cells (GSC). HLA-A*02-restricted T-cell epitopes were functionally determined by short peptides and numbers of antigen-specific T cells by HLA-peptide tetramer analysis.Results: A total of 2,897 proteins were identified in immunogenic tumor fractions. Based on a thorough filter process, 79 proteins were selected as potential T-cell antigens. Twenty-six of these were recognized by the patients' T cells, and five of them (CRKII, CFL1, CNTN1, NME2, and TKT) in up to 56% unrelated IDHmut glioma patients. Most immunogenic tumor-associated antigens (TAA) were expressed in IDHmut gliomas and GSCs, while being almost absent in normal brain tissues. Finally, we identified HLA-A*02-restricted epitopes for CRKII, NME2, and TKT that were recognized by up to 2.82% of antigen-specific peripheral cytotoxic T cells in IDHmut glioma patients.Conclusions: By analyzing the repertoire of T-cell target antigens in IDHmut glioma patients, we identified five novel immunogenic TAAs and confirmed their expression on IDHmut tumors and GSCs. Clin Cancer Res; 24(12); 2951-62. ©2018 AACR.


Asunto(s)
Biomarcadores de Tumor , Glioma/genética , Glioma/metabolismo , Isocitrato Deshidrogenasa/genética , Mutación , Linfocitos T/metabolismo , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Cromatografía Liquida , Cofilina 1/genética , Cofilina 1/metabolismo , Contactina 1/genética , Contactina 1/metabolismo , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Mapeo Epitopo , Glioma/inmunología , Humanos , Inmunohistoquímica , Isocitrato Deshidrogenasa/metabolismo , Nucleósido Difosfato Quinasas NM23/genética , Nucleósido Difosfato Quinasas NM23/metabolismo , Proteoma , Proteómica/métodos , Proteínas Proto-Oncogénicas c-crk/genética , Proteínas Proto-Oncogénicas c-crk/metabolismo , Linfocitos T/inmunología , Espectrometría de Masas en Tándem
15.
Oncoimmunology ; 6(11): e1360458, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29147626

RESUMEN

In this prospective study, we examined postoperative follow-up and preoperative IFN-γ T cell responses against 14 non-small cell lung cancer (NSCLC)-associated antigens in the blood of 51 patients with NSCLC, 7 patients with benign pulmonary tumors, and 10 tumor-free patients by enzyme-linked immunospot assay. The phenotype and function of T cells specific for tumor-associated antigens (TAAs) in the blood or tumor tissue of 9 NSCLC patients were characterized in detail using TNF-α, IL-2, and IFN-γ cytokine capture assays. We found that circulating TAA-specific T cells were significantly enriched in NSCLC compared with tumor-free patients. The most frequently recognized TAAs were Aurora kinase A, HER2/neu, NY-ESO-1, and p53. TNF-α was the most abundant cytokine secreted by TAA-specific T cells in the blood as well as by in situ-activated tumor-infiltrating lymphocytes, most of which were effector memory cells. The absence of TAA-reactive T cells identified patients at higher risk of tumor recurrence, irrespective of tumor stage (OR = 8.76, 95% CI: 1.57-34.79, p = 0.008). We conclude that pre-existing TAA-reactive circulating T cells are a strong independent prognostic factor for recurrence-free survival. These data may help discriminating high-risk from low-risk patients, improving prognostication, and redirecting adjuvant therapy. Our findings suggest the therapeutic relevance of Aurora kinase A, HER2/neu, NY-ESO-1, and p53 as targets for immunotherapy. This study is registered on Clinicaltrials.gov with trial identification number: NCT02515760.

16.
J Clin Invest ; 125(2): 739-51, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25562322

RESUMEN

The composition of tumor-targeted T cell infiltrates is a major prognostic factor in colorectal cancer (CRC) outcome; however, the functional role of these populations in prolonging patient survival remains unclear. Here, we evaluated 190 patients with CRC for the presence of functionally active tumor-infiltrating lymphocytes (TILs), the tumor specificity of these TILs, and the correlation between patient TILs and long-term survival. Using intracytoplasmic cytokine staining in conjunction with HLA multimers loaded with tumor peptide and antigen-specific cytokine secretion assays, we determined that TNF-α expression delineates a population of tumor antigen-specific (TA-specific) cytotoxic T lymphocytes (CTLs) present within tumors from patients with CRC. Upregulation of TNF-α expression in TILs strongly correlated with an increase in the total amount of intratumoral TNF-α, which is indicative of tumor-specific CTL activity. Moreover, a retrospective multivariate analysis of 102 patients with CRC, which had multiple immune parameters evaluated, revealed that increased TNF-α concentration was an independent prognostic factor. Together, these results indicate that the prognostic impact of T cell infiltrates for CRC maybe largely based on subpopulations of active TA-specific T cells within the tumor, suggesting causal implication for these cells in patient survival. Additionally, these results support the use of intratumoral TNF-α, which is indicative of T cell function, as a prognostic parameter for CRC.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/mortalidad , Linfocitos T CD8-positivos/patología , Neoplasias Colorrectales/patología , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Masculino , Proteínas de Neoplasias/inmunología , Estudios Prospectivos , Estudios Retrospectivos , Tasa de Supervivencia , Factor de Necrosis Tumoral alfa/inmunología
17.
Oncoimmunology ; 4(4): e1001217, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26137397

RESUMEN

VEGFR-2 is expressed on tumor vasculature and a target for anti-angiogenic intervention. VXM01 is a first in kind orally applied tumor vaccine based on live, attenuated Salmonella bacteria carrying an expression plasmid, encoding VEGFR-2. We here studied the safety, tolerability, T effector (Teff), T regulatory (Treg) and humoral responses to VEGFR2 and anti-angiogenic effects in advanced pancreatic cancer patients in a randomized, dose escalation phase I clinical trial. Results of the first 3 mo observation period are reported. Locally advanced or metastatic, pancreatic cancer patients were enrolled. In five escalating dose groups, 30 patients received VXM01 and 15 placebo on days 1, 3, 5, and 7. Treatment was well tolerated at all dose levels. No dose-limiting toxicities were observed. Salmonella excretion and salmonella-specific humoral immune responses occurred in the two highest dose groups. VEGFR2 specific Teff, but not Treg responses were overall increased in vaccinated patients. We furthermore observed a significant reduction of tumor perfusion after 38 d in vaccinated patients together with increased levels of serum biomarkers indicative of anti-angiogenic activity, VEGF-A, and collagen IV. Vaccine specific Teff responses significantly correlated with reductions of tumor perfusion and high levels of preexisting VEGFR2-specific Teff while those showing no antiangiogenic activity had low levels of preexisting VEGFR2 specific Teff, showed a transient early increase of VEGFR2-specific Treg and reduced levels of VEGFR2-specific Teff at later time points - pointing to the possibility that early anti-angiogenic activity might be based at least in part on specific reactivation of preexisting memory T cells.

18.
PLoS One ; 9(4): e93745, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24699869

RESUMEN

Genetically engineered T lymphocytes are a promising option for cancer therapy. Prior to adoptive transfer they have to be expanded in vitro to reach therapeutically sufficient numbers. So far, no universal method exists for selective in vitro expansion of engineered T lymphocytes. In order to overcome this problem and for proof of concept we incorporated a novel unique peptide sequence of ten amino acids as epitope (E-Tag) into the binding domains of two novel chimeric antigen receptors (ECARs) directed against either prostate stem cell antigen (PSCA) for the treatment of prostate cancer (PCa) or CD33 for the treatment of acute myeloide leukemia (AML). The epitope tag then was utilized for expanding ECAR engrafted T cells by triggering the modified T cells via a monoclonal antibody directed against the E-Tag (Emab). Moreover, the E-Tag served as an efficient selection epitope for immunomagnetic isolation of modified T cells to high purity. ECAR engrafted T cells were fully functional and mediated profound anti-tumor effects in the respective models of PCa or AML both in vitro and in vivo. The method can be integrated straightforward into clinical protocols to improve therapeutic efficiency of tumor treatment with CAR modified T lymphocytes.


Asunto(s)
Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Traslado Adoptivo , Epítopos , Humanos , Receptores de Antígenos/genética
19.
Oncoimmunology ; 2(12): e26770, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24498554

RESUMEN

We have recently described a novel modular targeting platform for T cell recruitment that not only efficiently replaces but also is superior to conventional T cell-engaging bispecific antibodies as it allows for the flexible targeting of several antigens and the delivery of co-stimulatory ligands to malignant lesions, thereby enhancing the antitumor potential of redirected T cells.

20.
Methods Mol Biol ; 869: 433-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22585507

RESUMEN

For some instances, protein gels need to be dried after SDS-PAGE, for example, if autoradiography should be performed from radioactive-labeled proteins after their separation on SDS-polyacrylamide gels. Another reason may be to simply store the gel in the laboratory book. Aside from expensive commercial solutions, especially for storage of the dried gel in the lab book, the simple and cheap drying protocol here presented may be sufficient.


Asunto(s)
Desecación/métodos , Electroforesis en Gel de Poliacrilamida/métodos , Tampones (Química) , Celofán/química , Desecación/instrumentación , Proteínas/química , Proteínas/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA