Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
1.
Nat Immunol ; 24(3): 414-422, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36732425

RESUMEN

Interferon-γ (IFNγ) is an important mediator of cellular immune responses, but high systemic levels of this cytokine are associated with immunopathology. IFNγ binds to its receptor (IFNγR) and to extracellular matrix (ECM) via four positively charged C-terminal amino acids (KRKR), the ECM-binding domain (EBD). Across evolution, IFNγ is not well conserved, but the EBD is highly conserved, suggesting a critical function. Here, we show that IFNγ lacking the EBD (IFNγΔKRKR) does not bind to ECM but still binds to the IFNγR and retains bioactivity. Overexpression of IFNγΔKRKR in tumors reduced local ECM binding, increased systemic levels and induced sickness behavior, weight loss and toxicity. To analyze the function of the EBD during infection, we generated IFNγΔKRKR mice lacking the EBD by using CRISPR-Cas9. Infection with lymphocytic choriomeningitis virus resulted in higher systemic IFNγΔKRKR levels, enhanced sickness behavior, weight loss and fatal toxicity. We conclude that local retention of IFNγ is a pivotal mechanism to protect the organism from systemic toxicity during prolonged immune stimulation.


Asunto(s)
Citocinas , Neoplasias , Ratones , Animales , Citocinas/metabolismo , Interferón gamma/metabolismo , Transducción de Señal , Matriz Extracelular/metabolismo
4.
Blood ; 143(14): 1365-1378, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38277625

RESUMEN

ABSTRACT: Acquired aplastic anemia is a bone marrow failure syndrome characterized by hypocellular bone marrow and peripheral blood pancytopenia. Frequent clinical responses to calcineurin inhibition and antithymocyte globulin strongly suggest critical roles for hematopoietic stem/progenitor cell-reactive T-cell clones in disease pathophysiology; however, their exact contribution and antigen specificities remain unclear. We determined differentiation states and targets of dominant T-cell clones along with their potential to eliminate hematopoietic progenitor cells in the bone marrow of 15 patients with acquired aplastic anemia. Single-cell sequencing and immunophenotyping revealed oligoclonal expansion and effector differentiation of CD8+ T-cell compartments. We reexpressed 28 dominant T-cell receptors (TCRs) of 9 patients in reporter cell lines to determine reactivity with (1) in vitro-expanded CD34+ bone marrow, (2) CD34- bone marrow, or (3) peptide pools covering immunodominant epitopes of highly prevalent viruses. Besides 5 cytomegalovirus-reactive TCRs, we identified 3 TCRs that recognized antigen presented on hematopoietic progenitor cells. T cells transduced with these TCRs eliminated hematopoietic progenitor cells of the respective patients in vitro. One progenitor cell-reactive TCR (11A5) also recognized an epitope of the Epstein-Barr virus-derived latent membrane protein 1 (LMP1) presented on HLA-A∗02:01. We identified 2 LMP1-related mimotopes within the human proteome as activating targets of TCR 11A5, providing proof of concept that molecular mimicry of viral and self-epitopes can drive T cell-mediated elimination of hematopoietic progenitor cells in aplastic anemia.


Asunto(s)
Anemia Aplásica , Infecciones por Virus de Epstein-Barr , Humanos , Imitación Molecular , Infecciones por Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4 , Células Madre Hematopoyéticas/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo
5.
Haematologica ; 109(4): 1121-1127, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-37767552

RESUMEN

The null allele HLA-C*04:09N differs from HLA-C*04:01 in a frameshift mutation within its cytoplasmic domain, resulting in translation of 32 additional amino acids that are assumed to prevent cell surface expression. However, we recently identified a multiple myeloma-reactive T-cell receptor (TCR) that appeared to recognize antigen presented on HLA-C*04:09N and encouraged us to ask whether HLA-C*04:09N, albeit not easily detectable at the cell surface, can present antigen sufficient for T-cell activation. We generated two HLA-class I-deficient cell lines, re-expressed HLAC* 04:09N, detected HLA expression by flow cytometry, and tested for T-cell activation using a cytomegalovirus peptide- specific HLA-C*04:01-restricted TCR. In both cell lines, HLA-C*04:09N expression was detectable at the cell surface and could be enhanced by IFN-γ exposure. Recombinant HLA-C*04:09N expression was sufficient for T-cell activation in vitro, which could be blocked by an HLA-class I-specific antibody, suggesting HLA-TCR interaction at the cell surface. Peripheral blood mononuclear cells isolated from an individual who physiologically expressed HLA-C*04:09N triggered peptide-specific T-cell activation, confirming our results with cells with natural HLA expression levels. In conclusion, we present peptide-specific HLA-C*04:09N-restricted T-cell activation and suggest consideration of this allele in the appropriate clinical context, such as allogeneic stem cell transplantation, or in the setting of cellular therapy.


Asunto(s)
Antígenos HLA-C , Leucocitos Mononucleares , Humanos , Antígenos HLA-C/genética , Péptidos , Linfocitos T Citotóxicos , Receptores de Antígenos de Linfocitos T
6.
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
7.
Cytotherapy ; 24(8): 818-826, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35525797

RESUMEN

BACKGROUND AND AIMS: Epstein-Barr virus (EBV) is associated with solid and hematopoietic malignancies. After allogeneic stem cell transplantation, EBV infection or reactivation represents a potentially life-threatening condition with no specific treatment available in clinical routine. In vitro expansion of naturally occurring EBV-specific T cells for adoptive transfer is time-consuming and influenced by the donor's T-cell receptor (TCR) repertoire and requires a specific memory compartment that is non-existent in seronegative individuals. The authors present highly efficient identification of EBV-specific TCRs that can be expressed on human T cells and recognize EBV-infected cells. METHODS AND RESULTS: Mononuclear cells from six stem cell grafts were expanded in vitro with three HLA-B*35:01- or four HLA-A*02:01-presented peptides derived from six EBV proteins expressed during latent and lytic infection. Epitope-specific T cells expanded on average 42-fold and were single-cell-sorted and TCRαß-sequenced. To confirm specificity, 11 HLA-B*35:01- and six HLA-A*02:01-restricted dominant TCRs were expressed on reporter cell lines, and 16 of 17 TCRs recognized their presumed target peptides. To confirm recognition of virus-infected cells and assess their value for adoptive therapy, three selected HLA-B*35:01- and four HLA-A*02:01-restricted TCRs were expressed on human peripheral blood lymphocytes. All TCR-transduced cells recognized EBV-infected lymphoblastoid cell lines. CONCLUSIONS: The authors' approach provides sets of EBV epitope-specific TCRs in two different HLA contexts. Resulting cellular products do not require EBV-seropositive donors, can be adjusted to cell subsets of choice with exactly defined proportions of target-specific T cells, can be tracked in vivo and will help to overcome unmet clinical needs in the treatment and prophylaxis of EBV reactivation and associated malignancies.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Herpesvirus Humano 4 , Epítopos , Infecciones por Virus de Epstein-Barr/terapia , Antígenos HLA-A , Humanos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Complemento 3d , Linfocitos T
8.
Gynecol Oncol ; 164(1): 68-75, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34794839

RESUMEN

BACKGROUND: The impact of adjuvant radiotherapy (RT) to the vulva with regard to prognosis and local recurrence in patients with vulvar squamous cell cancer (VSCC) is poorly described. PATIENTS AND METHODS: In the AGO-CaRE-1 study 1618 patients with primary VSCC FIGO stage ≥ IB, treated between 1998-2008, were documented. In this retrospective subanalysis, 360 patients were included based on the following criteria: nodal involvement (pN+), known RT treatment and known radiation fields. RESULTS: The majority had pT1b/pT2 tumors (n=299; 83.1%). In 76.7%, R0 resection was achieved. 57/360 (15.8%) N+ patients were treated with adjuvant RT to the groins/pelvis and 146/360 (40.5%) received adjuvant RT to the vulva and groins/pelvis. 157/360 (43.6%) patients did not receive any adjuvant RT. HPV status was available in 162/360 patients (45.0%), 75/162 tumors were HPV+(46.3%), 87/162 (53.7%) HPV-. During a median follow-up of 17.2 months, recurrence at the vulva only occurred in 25.5% of patients without adjuvant RT, in 22.8% of patients with adjuvant RT to groins/pelvis and in 15.8% of patients with adjuvant RT to the vulva and groins/pelvis respectively. The risk reducing effect of local RT was independent of the resection margin status. 50% disease free survival time (50% DFST) indicated a stronger impact of adjuvant RT to the vulva in HPV+ compared to HPV- patients (50% DFST 20.7 months vs. 17.8 months). CONCLUSION: Adjuvant RT to the vulva was associated with a lower risk for local recurrence in N+ VSCC independent of the resection margin status. This observation was more pronounced in patients with HPV+ tumors in comparison to HPV- tumors.


Asunto(s)
Carcinoma de Células Escamosas/radioterapia , Recurrencia Local de Neoplasia/radioterapia , Neoplasias de la Vulva/radioterapia , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/patología , Supervivencia sin Enfermedad , Femenino , Alemania , Humanos , Persona de Mediana Edad , Recurrencia Local de Neoplasia/mortalidad , Recurrencia Local de Neoplasia/patología , Radioterapia Adyuvante , Neoplasias de la Vulva/mortalidad , Neoplasias de la Vulva/patología , Adulto Joven
9.
Int J Cancer ; 148(12): 3097-3110, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33600609

RESUMEN

Adoptive T cell therapy (ATT) has revolutionized the treatment of cancer patients. A sufficient number of functional T cells are indispensable for ATT efficacy; however, several ATT dropouts have been reported due to T cell expansion failure or lack of T cell persistence in vivo. With the aim of providing ATT also to those patients experiencing insufficient T cell manufacturing via standard protocol, we evaluated if minimally manipulative prolongation of in vitro expansion (long-term [LT] >3 weeks with IL-7 and IL-15 cytokines) could result in enhanced T cell yield with preserved T cell functionality. The extended expansion resulted in a 39-fold increase of murine CD8+ T central memory cells (Tcm). LT expanded CD8+ and CD4+ Tcm cells retained a gene expression profile related to Tcm and T memory stem cells (Tscm). In vivo transfer of LT expanded Tcm revealed persistence and antitumor capacity. We confirmed our in vitro findings on human T cells, on healthy donors and diffuse large B cell lymphoma patients, undergoing salvage therapy. Our study demonstrates the feasibility of an extended T cell expansion as a practicable alternative for patients with insufficient numbers of T cells after the standard manufacturing process thereby increasing ATT accessibility.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Linfoma de Células B Grandes Difuso/terapia , Linfocitos T/citología , Linfocitos T/trasplante , Animales , Estudios de Casos y Controles , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Células Cultivadas , Humanos , Memoria Inmunológica , Inmunoterapia Adoptiva , Interleucina-15/farmacología , Interleucina-7/farmacología , Masculino , Ratones , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Blood ; 132(9): 924-934, 2018 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-30002144

RESUMEN

To date, little is known about the interaction between (pre-)malignant B cells and T cells. We generated transgenic mice that allow B cell-specific induction of the oncogene SV40 large T-antigen (TAg) to analyze the role of oncogene-specific T cells during sporadic B-cell lymphoma development. Constitutive TAg expression in CD19-Cre × LoxP-Tag mice resulted in TAg-tolerant CD8+ T cells and development of B-cell lymphomas. In contrast, CD19-CreERT2 × LoxP-Tag mice retained TAg-competent CD8+ T cells at time of oncogene induction and TAg expression in few B cells of adult mice resulted in exceptionally rare lymphoma formation late in life. Increased lymphoma incidence in the absence of TAg-specific T cells suggested T cell-mediated inhibition of lymphoma progression. However, TAg-initiated B cells were not eliminated by T cells and detected long term. Our results demonstrate a failure of the immune system to eradicate lymphoma-initiating B cells, retaining the risk of lymphoma development.


Asunto(s)
Antígenos Transformadores de Poliomavirus/inmunología , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunidad Celular , Linfoma de Células B/inmunología , Animales , Antígenos Transformadores de Poliomavirus/genética , Linfocitos B/patología , Linfocitos T CD8-positivos/patología , Linfoma de Células B/genética , Linfoma de Células B/patología , Ratones , Ratones Noqueados
11.
BMC Bioinformatics ; 20(1): 450, 2019 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-31477009

RESUMEN

BACKGROUND: Mutational signatures are specific patterns of somatic mutations introduced into the genome by oncogenic processes. Several mutational signatures have been identified and quantified from multiple cancer studies, and some of them have been linked to known oncogenic processes. Identification of the processes contributing to mutations observed in a sample is potentially informative to understand the cancer etiology. RESULTS: We present here SigsPack, a Bioconductor package to estimate a sample's exposure to mutational processes described by a set of mutational signatures. The package also provides functions to estimate stability of these exposures, using bootstrapping. The performance of exposure and exposure stability estimations have been validated using synthetic and real data. Finally, the package provides tools to normalize the mutation frequencies with respect to the tri-nucleotide contents of the regions probed in the experiment. The importance of this effect is illustrated in an example. CONCLUSION: SigsPack provides a complete set of tools for individual sample exposure estimation, and for mutation catalogue & mutational signatures normalization.


Asunto(s)
Carcinogénesis/genética , Genoma Humano , Mutación , Proteínas de Neoplasias/genética , Neoplasias/genética , Programas Informáticos , Carcinogénesis/patología , Análisis Mutacional de ADN , Humanos , Tasa de Mutación , Neoplasias/patología
12.
Int J Cancer ; 141(12): 2551-2561, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-28833076

RESUMEN

Targeted oncogene inactivation by small molecule inhibitors can be very effective but tumor recurrence is a frequent problem in the clinic. Therapy by inactivation of the cancer-driving oncogene in transplanted tumors was shown to be augmented in the presence of T cells. However, these experiments did not take into account the long-term, usually tolerogenic, interaction of de novo malignancies with the immune system. Here, we employed mice, in which SV40 large T (Tag) and firefly luciferase (Luc) as fusion protein (TagLuc) could be regulated with the Tet-on system and upon activation resulted in tumors after a long latency. TagLuc inactivation induced profound tumor regression, demonstrating sustained oncogene addiction. While tumor relapse after TagLuc inactivation was prevented in immunocompetent mice bearing transplanted tumors, autochthonous tumors relapsed or recurred after therapy discontinuation indicating that the immune system that coevolved with the malignancy over an extended period of time lost the potency to mount an efficient anti-tumor immune response. By contrast, adoptively transferred CD8+ T cells targeting the cancer-driving oncogene eradicated recurrent autochthonous tumors, highlighting a suitable therapy option in a clinically relevant model.


Asunto(s)
Linfocitos T CD8-positivos/trasplante , Doxiciclina/administración & dosificación , Silenciador del Gen , Sistema Inmunológico/metabolismo , Neoplasias Experimentales/terapia , Traslado Adoptivo , Animales , Antígenos Transformadores de Poliomavirus/genética , Antígenos Transformadores de Poliomavirus/metabolismo , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Doxiciclina/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trasplante de Neoplasias , Neoplasias Experimentales/inmunología , Proteínas Recombinantes de Fusión/genética
13.
Proc Natl Acad Sci U S A ; 110(6): 2276-81, 2013 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-23341634

RESUMEN

The contribution of molecules such as perforin, IFN-γ (IFNγ), and particularly Fas ligand (FasL) by transferred CD8(+) effector T (T(E)) cells to rejection of large, established tumors is incompletely understood. Efficient attack against large tumors carrying a surrogate tumor antigen (mimicking a "passenger" mutation) by T(E) cells requires action of IFNγ on tumor stroma cells to avoid selection of antigen-loss variants. Because "cancer-driving" antigens (CDAs) are rarely counterselected, IFNγ may be expected to be dispensable in elimination of cancers by targeting a CDA. Here, initial regression of large, established tumors required neither IFNγ, FasL, nor perforin by transferred CD8(+) T(E) cells targeting Simian Virus (SV) 40 large T as CDA. However, cytotoxic T(E) cells lacking IFNγ or FasL could not prevent relapse despite retention of the rejection antigen by the cancer cells. Complete tumor rejection required IFNγ-regulated Fas by the tumor stroma. Therefore, T(E) cells lacking IFNγ or FasL cannot prevent progression of antigenic cancer because the tumor stroma escapes destruction if its Fas expression is down-regulated.


Asunto(s)
Neoplasias/inmunología , Neoplasias/patología , Receptor fas/metabolismo , Animales , Línea Celular Tumoral , Proteína Ligando Fas/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inmunoterapia Adoptiva , Interferón gamma/deficiencia , Interferón gamma/genética , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Neoplasias/metabolismo , Neoplasias/terapia , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Células del Estroma/inmunología , Células del Estroma/patología , Linfocitos T Citotóxicos/inmunología , Receptor de Interferón gamma
14.
Int J Cancer ; 136(8): 1814-26, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25242680

RESUMEN

Tumor-induced immunosuppression remains a major challenge for immunotherapy of cancer patients. To further elucidate why an allogeneic gene-modified [interleukin-7 (IL-7)/CD80-cotransfected] renal cell cancer (RCC) vaccine failed to induce clinically relevant TH-1-polarized immune responses, peripheral blood mononuclear cells from enrolled study patients were analyzed by gene expression profiling (GEP) both prior and after vaccination. At baseline before vaccination, a profound downregulation of gene signatures associated with antigen presentation, immune response/T cells, cytokines/chemokines and signaling/transcription factors was observed in RCC patients as compared to healthy controls. Vaccination led to a partial reversion of preexisting immunosuppression, however, GEP indicated that an appropriate TH-1 polarization could not be achieved. Most interestingly, our results suggest that the nuclear factor-kappa B signaling pathway might be involved in the impairment of immunological responsiveness and the observed TH-2 deviation. In summary, our data suggest that GEP might be a powerful tool for the prediction of immunosuppression and the monitoring of immune responses within immunotherapy trials.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/inmunología , Neoplasias Renales/genética , Neoplasias Renales/inmunología , FN-kappa B/inmunología , Transcriptoma/genética , Adulto , Anciano , Citocinas/inmunología , Perfilación de la Expresión Génica/métodos , Humanos , Terapia de Inmunosupresión/métodos , Inmunoterapia/métodos , Leucocitos Mononucleares/inmunología , Masculino , Persona de Mediana Edad , FN-kappa B/genética , Transducción de Señal/inmunología , Células TH1/inmunología , Células Th2/inmunología , Transcriptoma/inmunología
15.
Cancer Metastasis Rev ; 33(4): 943-51, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25319202

RESUMEN

The family of metastasis-associated (MTA) genes is a small group of transcriptional co-regulators which are involved in various physiological functions, ranging from lymphopoietic cell differentiation to the development and maintenance of epithelial cell adhesions. By recruiting histone-modifying enzymes to specific promoter sequences, MTA proteins can function both as transcriptional repressors and activators of a number of cancer-relevant proteins, including Snail, E-cadherin, signal transducer and activator of transcriptions (STATs), and the estrogen receptor. Their involvement in the epithelial-mesenchymal transition process and regulatory interactions with estrogen receptor activity has made MTA proteins highly interesting research candidates, especially in the field of hormone-sensitive breast cancer and malignancies of the female reproductive tract. This review focuses on the current knowledge about the function and regulation of MTA1 and MTA3 proteins in gynecological cancer, including ovarian, endometrial, and cervical tumors.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Neoplasias de los Genitales Femeninos/genética , Histona Desacetilasas/genética , Proteínas de Neoplasias/genética , Proteínas Represoras/genética , Diferenciación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Neoplasias de los Genitales Femeninos/patología , Histona Desacetilasas/biosíntesis , Humanos , Proteínas de Neoplasias/biosíntesis , Receptores de Estrógenos/metabolismo , Proteínas Represoras/biosíntesis , Transactivadores
16.
Eur J Immunol ; 44(9): 2811-21, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24846220

RESUMEN

Immunity to tumor differentiation antigens, such as melanoma antigen recognized by T cells 1 (MART-1), has been comprehensively studied. Intriguingly, CD8(+) T cells specific for the MART-1(26(27)-35) epitope in the context of HLA-A0201 are about 100 times more abundant compared with T cells specific for other tumor-associated antigens. Moreover, MART-1-specific CD8(+) T cells show a highly biased usage of the Vα-region gene TRAV12-2. Here, we provide independent support for this notion, by showing that the combinatorial pairing of different TCRα- and TCRß- chains derived from HLA-A2-MART-1(26-35) -specific CD8(+) T-cell clones is unusually permissive in conferring MART-1 specificity, provided the CDR1α TRAV12-2 region is used. Whether TCR bias alone accounts for the unusual abundance of HLA-A2-MART-1(26-35) -specific CD8(+) T cells has remained conjectural. Here, we provide an alternative explanation: misinitiated transcription of the MART-1 gene resulting in truncated mRNA isoforms leads to lack of promiscuous transcription of the MART-1(26-35) epitope in human medullary thymic epithelial cells and, consequently, evasion of central self-tolerance toward this epitope. Thus, biased TCR usage and leaky central tolerance might act in an independent and additive manner to confer high frequency of MART-1(26-35) -specific CD8(+) T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Epiteliales/inmunología , Epítopos de Linfocito T/inmunología , Antígeno MART-1/inmunología , Timo/inmunología , Iniciación de la Transcripción Genética/inmunología , Linfocitos T CD8-positivos/citología , Línea Celular , Células Epiteliales/citología , Femenino , Antígeno HLA-A2/inmunología , Humanos , Masculino , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Timo/citología
17.
Blood ; 121(10): 1740-8, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23305737

RESUMEN

Tumors frequently induce immature myeloid cells (iMC), which suppress specific and unrelated cytotoxic T lymphocyte (CTL) responses and are termed myeloid-derived suppressor cells (MDSC). Mainly analyzed by in vitro assays in tumor transplantation models, little is known about their function in autochthonous tumor models in vivo. We analyzed iMC in 3 SV40 large T (Tag)-driven conditional autochthonous cancer models with different immune status: (1) Early Tag-specific CTL competence and rare stochastic Tag activation leading to sporadic cancer, which induces an aberrant immune response and CTL tolerance; (2) Cre/LoxP recombinase-mediated hepatocellular carcinoma (HCC) development in neonatal Tag-tolerant mice; and (3) Tag-activation through Cre recombinase-encoding viruses in the liver and HCC development with systemic anti-Tag CTL immunity. In the first but not two latter models, tumors induced CTL hyporesponsiveness to tumor-unrelated antigens. Regardless of the model, tumors produced interleukin-6 and vascular endothelial growth factor but not granulocyte macrophage­colony-stimulating factor (GM-CSF) and induced iMC (CD11b(+)Gr-1(int)) that suppressed CTL responses in vitro. None of the iMC from the different tumor models suppressed CTL responses in adoptive cell transfer experiments unless GM-CSF was provided in vivo. Together, iMC expand independent of the type of antitumor response and are not immunosuppressive in a cell-autonomous fashion.


Asunto(s)
Antígenos Transformadores de Poliomavirus/genética , Linfocitos T CD8-positivos/inmunología , Carcinoma Hepatocelular/etiología , Neoplasias Hepáticas Experimentales/etiología , Células Mieloides/inmunología , Linfocitos T Citotóxicos/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Citocinas/sangre , Citometría de Flujo , Humanos , Tolerancia Inmunológica , Inmunización , Técnicas In Vitro , Integrasas/metabolismo , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/metabolismo , Células Mieloides/patología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/patología
18.
Mol Med ; 18: 1499-508, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23269976

RESUMEN

Our previously reported phase I clinical trial with the allogeneic gene-modified tumor cell line RCC-26/CD80/IL-2 showed that vaccination was well tolerated and feasible in metastatic renal cell carcinoma (RCC) patients. Substantial disease stabilization was observed in most patients despite a high tumor burden at study entry. To investigate alterations in immune responses that might contribute to this effect, we performed an extended immune monitoring that included analysis of reactivity against multiple antigens, cytokine/chemokine changes in serum and determination of the frequencies of immune suppressor cell populations, including natural regulatory T cells (nTregs) and myeloid-derived suppressor cell subsets (MDSCs). An overall immune response capacity to virus-derived control peptides was present in 100% of patients before vaccination. Vaccine-induced immune responses to tumor-associated antigens occurred in 75% of patients, demonstrating the potent immune stimulatory capacity of this generic vaccine. Furthermore, some patients reacted to peptide epitopes of antigens not expressed by the vaccine, showing that epitope-spreading occurred in vivo. Frequencies of nTregs and MDSCs were comparable to healthy donors at the beginning of study. A significant decrease of nTregs was detected after vaccination (p = 0.012). High immune response rates, decreased frequencies of nTregs and a mixed T helper 1/T helper 2 (T(H)1/T(H)2)-like cytokine pattern support the applicability of this RCC generic vaccine for use in combination therapies.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/patología , Inmunidad/inmunología , Neoplasias Renales/inmunología , Linfocitos T Reguladores/inmunología , Vacunación , Carcinoma de Células Renales/sangre , Carcinoma de Células Renales/prevención & control , Citocinas/biosíntesis , Citocinas/sangre , Humanos , Neoplasias Renales/sangre , Neoplasias Renales/patología , Neoplasias Renales/prevención & control , Recuento de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Células Mieloides/inmunología , Células Mieloides/patología , Metástasis de la Neoplasia , Estadificación de Neoplasias , Péptidos/inmunología , Análisis de Supervivencia , Células TH1/inmunología , Células Th2/inmunología , Factores de Tiempo , Resultado del Tratamiento
19.
Blood ; 118(4): 946-54, 2011 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-21613253

RESUMEN

Adoptive therapy with T-cell receptor (TCR)-engineered T cells is a promising approach in cancer treatment. While usage of T cells specific for tumor-associated antigens (TAAs) can lead to serious side effects because of autoimmunity, targeting true tumor-specific mutations, such as the products of translocations in leukemias, should reduce such a risk. A potentially ideal target might be the chimeric protein TEL-AML1, which results from the chromosomal translocation 12;21 and represents the most common fusion gene in childhood B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Within the fusion region of TEL-AML1, a single epitope has been described by reverse immunology as immunogenic in HLA-A*0201 restriction settings. As a potential source of TCRs specific for this TEL-AML1 epitope, we have used mice expressing a human TCR-αß repertoire and human MHC class I. Surprisingly, we have found that, although a specific functional CD8(+) T-cell response against this peptide could be evoked, the described epitope was in fact not endogenously processed. Analyses done with a potent antigen-presenting cell line, as well as with purified human proteasomes, support the conclusion that this peptide cannot be proposed as a potential target in immunotherapy of ALL in HLA-A*0201-restricted fashion.


Asunto(s)
Presentación de Antígeno/inmunología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/inmunología , Epítopos de Linfocito T/inmunología , Proteínas de Fusión Oncogénica/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Separación Celular , Cromatografía Líquida de Alta Presión , Técnicas de Cocultivo , Citometría de Flujo , Humanos , Activación de Linfocitos/inmunología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Translocación Genética
20.
J Immunother Cancer ; 11(2)2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36822673

RESUMEN

BACKGROUND: Adoptive transfer of patient's T cells, engineered to express a T cell receptor (TCR) with defined novel antigen specificity, is a convenient form of cancer therapy. In most cases, major histocompatibility complex (MHC) I-restricted TCRs are expressed in CD8+ T cells and the development of CD4+ T cells engineered to express an MHC II-restricted TCR lacks behind. Critical is the choice of the target antigen, whether the epitope is efficiently processed and binds with high affinity to MHC molecules. A mutation in the transforming growth factor ß receptor 2 (TGFßR2(-1)) gene creates a frameshift peptide caused by the deletion of one adenine (-1) within a microsatellite sequence. This somatic mutation is recurrent in microsatellite instable colorectal and gastric cancers and, therefore, is a truly tumor-specific antigen detected in many patients. METHODS: ABabDR4 mice, which express a diverse human TCR repertoire restricted to human MHC II molecule HLA-DRA/DRB1*0401 (HLA-DR4), were immunized with the TGFßR2(-1) peptide and TGFßR2(-1)-specific TCRs were isolated from responding CD4+ T cells. The TGFßR2(-1)-specific TCRs were expressed in human CD4+ T cells and their potency and safety profile were assessed by co-cultures and other functional assays. RESULTS: We demonstrated that TGFßR2(-1) neoantigen is immunogenic and elicited CD4+ T cell responses in ABabDR4 mice. When expressed in human CD4+ T cells, the HLA-DR4 restricted TGFßR2(-1)-specific TCRs induced IFNy expression at low TGFßR2(-1) peptide amounts. The TGFßR2(-1)-specific TCRs recognized HLA-DR4+ lymphoblastoid cells, which endogenously processed and presented the neoantigen, and colorectal cancer cell lines SW48 and HCT116 naturally expressing the TGFßR2(-1) mutation. No MHC II alloreactivity or cross-reactivity to peptides with a similar TCR-recognition motif were observed, indicating the safety of the TCRs. CONCLUSIONS: The data suggest that HLA-DR4-restricted TCRs specific for the TGFßR2(-1) recurrent neoantigen can be valuable candidates for adoptive T cell therapy of a sizeable number of patients with cancer.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Humanos , Ratones , Animales , Antígeno HLA-DR4/metabolismo , Receptores de Antígenos de Linfocitos T , Antígenos de Neoplasias , Péptidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA