Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
1.
APMIS ; 132(7): 479-491, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38757248

RESUMEN

The Melan-A (melanocyte antigen) protein, also termed 'melanoma antigen recognized by T cells 1' (MART-1) is a protein with unknown function whose expression is specific for the melanocyte lineage. Antibodies against Melan-A are thus used for identifying melanocytic tumors, but some Melan-A antibodies show an additional - diagnostically useful - cross-reactivity against an unspecified protein involved in corticosteroid hormone synthesis. To comprehensively compare the staining patterns of a specific and a cross-reactive Melan-A antibody in normal and neoplastic tissues, tissue microarrays containing 15,840 samples from 133 different tumor types and subtypes as well as 608 samples of 76 different normal tissue types were analyzed by immunohistochemistry. For the Melan-A-specific antibody 'Melan-A specific' (MSVA-900M), Melan-A positivity was seen in 96.0% of 25 benign nevi, 93.0% of 40 primary and 86.7% of 75 metastatic melanomas, 82.4% of 85 renal angiomyolipomas as well as 96.4% of 84 neurofibromas, 2.2% of 46 granular cell tumors, 1.0% of 104 schwannomas, and 1.1% of 87 leiomyosarcomas. The cross-reactive antibody 'Melan-A+' (MSVA-901M+) stained 98.1% of the tumors stained by 'Melan-A specific'. In addition, high positivity rates were seen in sex-cord-stroma tumors of the ovary (35.3%-100%) and the testis (86.7%) as well as for adrenocortical neoplasms (76.3%-83.0%). Only nine further tumor groups showed Melan-A+ staining, including five different categories of urothelial carcinomas. Our data provide a comprehensive overview on the staining patterns of specific and cross-reactive Melan-A antibodies. The data demonstrate that both antibodies are highly useful for their specific purpose. It is important for pathologists to distinguish these two Melan-A antibody subtypes for their daily work.


Asunto(s)
Reacciones Cruzadas , Inmunohistoquímica , Antígeno MART-1 , Neoplasias , Humanos , Reacciones Cruzadas/inmunología , Antígeno MART-1/inmunología , Antígeno MART-1/análisis , Inmunohistoquímica/métodos , Neoplasias/inmunología , Neoplasias/diagnóstico , Neoplasias/patología , Melanoma/inmunología , Melanoma/diagnóstico , Melanoma/patología , Biomarcadores de Tumor/inmunología , Biomarcadores de Tumor/análisis , Análisis de Matrices Tisulares , Femenino
2.
J Immunol ; 208(3): 562-570, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35031578

RESUMEN

Aging is associated with functional deficits in the naive T cell compartment, which compromise the generation of de novo immune responses against previously unencountered Ags. The mechanisms that underlie this phenomenon have nonetheless remained unclear. We found that naive CD8+ T cells in elderly humans were prone to apoptosis and proliferated suboptimally in response to stimulation via the TCR. These abnormalities were associated with dysregulated lipid metabolism under homeostatic conditions and enhanced levels of basal activation. Importantly, reversal of the bioenergetic anomalies with lipid-altering drugs, such as rosiglitazone, almost completely restored the Ag responsiveness of naive CD8+ T cells. Interventions that favor lipid catabolism may therefore find utility as adjunctive therapies in the elderly to promote vaccine-induced immunity against targetable cancers and emerging pathogens, such as seasonal influenza viruses and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).


Asunto(s)
Envejecimiento/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunocompetencia/efectos de los fármacos , Metabolismo de los Lípidos , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis , Linfocitos T CD8-positivos/metabolismo , COVID-19/inmunología , Vacunas contra el Cáncer/inmunología , División Celular , Femenino , Fenofibrato/farmacología , Glucosa/metabolismo , Antígeno HLA-A2/inmunología , Humanos , Hipolipemiantes/farmacología , Hipolipemiantes/uso terapéutico , Gripe Humana/inmunología , Metabolismo de los Lípidos/efectos de los fármacos , Activación de Linfocitos , Antígeno MART-1/química , Antígeno MART-1/inmunología , Masculino , Persona de Mediana Edad , Neoplasias/inmunología , Fragmentos de Péptidos/inmunología , Rosiglitazona/farmacología , Método Simple Ciego , Vacunación , Vacunas Virales/inmunología , Adulto Joven
3.
J Immunother Cancer ; 9(12)2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34933966

RESUMEN

BACKGROUND: Anti-programmed cell death protein 1 (PD-1) antibodies are now routinely administered for metastatic melanoma and for increasing numbers of other cancers, but still only a fraction of patients respond. Better understanding of the modes of action and predictive biomarkers for clinical outcome is urgently required. Cancer rejection is mostly T cell-mediated. We previously showed that the presence of NY-ESO-1-reactive and/or Melan-A-reactive T cells in the blood correlated with prolonged overall survival (OS) of patients with melanoma with a heterogeneous treatment background. Here, we investigated whether such reactive T cells can also be informative for clinical outcomes in metastatic melanoma under PD-1 immune-checkpoint blockade (ICB). METHODS: Peripheral blood T cell stimulation by NY-ESO-1 and Melan-A overlapping peptide libraries was assessed before and during ICB in two independent cohorts of a total of 111 patients with stage IV melanoma. In certain cases, tumor-infiltrating lymphocytes could also be assessed for such responses. These were characterized using intracellular cytokine staining for interferon gamma (IFN-γ), tumor negrosis factor (TNF) and CD107a. Digital pathology analysis was performed to quantify NY-ESO-1 and Melan-A expression by tumors. Endpoints were OS and progression-free survival (PFS). RESULTS: The initial presence in the circulation of NY-ESO-1- or Melan-A-reactive T cells which became no longer detectable during ICB correlated with validated, prolonged PFS (HR:0.1; p>0.0001) and OS (HR:0.2; p=0.021). An evaluation of melanoma tissue from selected cases suggested a correlation between tumor-resident NY-ESO-1- and Melan-A-reactive T cells and disease control, supporting the notion of a therapy-associated sequestration of cells from the periphery to the tumor predominantly in those patients benefitting from ICB. CONCLUSIONS: Our findings suggest a PD-1 blockade-dependent infiltration of melanoma-reactive T cells from the periphery into the tumor and imply that this seminally contributes to effective treatment.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Biomarcadores de Tumor/metabolismo , Linfocitos T CD8-positivos/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Antígeno MART-1/metabolismo , Melanoma/mortalidad , Proteínas de la Membrana/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Anciano , Anciano de 80 o más Años , Antígenos de Neoplasias/inmunología , Biomarcadores de Tumor/inmunología , Femenino , Estudios de Seguimiento , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Linfocitos Infiltrantes de Tumor/inmunología , Antígeno MART-1/inmunología , Masculino , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Melanoma/patología , Proteínas de la Membrana/inmunología , Persona de Mediana Edad , Pronóstico , Tasa de Supervivencia
4.
Front Immunol ; 12: 674276, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34566953

RESUMEN

Adoptive immunotherapy based on the transfer of anti-tumor cytotoxic T lymphocytes (CTLs) is a promising strategy to cure cancers. However, rapid expansion of numerous highly functional CTLs with long-lived features remains a challenge. Here, we constructed NIH/3T3 mouse fibroblast-based artificial antigen presenting cells (AAPCs) and precisely evaluated their ability to circumvent this difficulty. These AAPCs stably express the essential molecules involved in CTL activation in the HLA-A*0201 context and an immunogenic HLA-A*0201 restricted analogue peptide derived from MART-1, an auto-antigen overexpressed in melanoma. Using these AAPCs and pentamer-based magnetic bead-sorting, we defined, in a preclinical setting, the optimal conditions to expand pure MART-1-specific CTLs. Numerous highly purified MART-1-specific CTLs were rapidly obtained from healthy donors and melanoma patients. Both TCR repertoire and CDR3 sequence analyses revealed that MART-1-specific CTL responses were similar to those reported in the literature and obtained with autologous or allogeneic presenting cells. These MART-1-specific CTLs were highly cytotoxic against HLA-A*0201+ MART-1+ tumor cells. Moreover, they harbored a suitable phenotype for immunotherapy, with effector memory, central memory and, most importantly, stem cell-like memory T cell features. Notably, the cells harboring stem cell-like memory phenotype features were capable of self-renewal and of differentiation into potent effector anti-tumor T cells. These "off-the-shelf" AAPCs represent a unique tool to rapidly and easily expand large numbers of long-lived highly functional pure specific CTLs with stem cell-like memory T cell properties, for the development of efficient adoptive immunotherapy strategies against cancers.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Técnicas de Cultivo de Célula/métodos , Inmunoterapia Adoptiva/métodos , Melanoma , Linfocitos T Citotóxicos/inmunología , Animales , Humanos , Memoria Inmunológica/inmunología , Activación de Linfocitos/inmunología , Antígeno MART-1/inmunología , Ratones , Células 3T3 NIH
5.
Int Immunopharmacol ; 98: 107848, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34126342

RESUMEN

The efficacy of adoptive cell therapy (ACT) relies on the abilities of T cells in self-expansion, survival and the secretion of effector molecules. Here, we presented an optimized method to generate T cells with improved functions by supplementing the culture medium with p38 inhibitor and the combination of IL-7 and IL-15 or IL-2 alone. The addition of p38 inhibitor, Doramapimod or SB202190, to IL-7 and IL-15 culture largely increased the capacity of T cells in the proliferation with enrichment of the naïve-like subsets and expression of CD62L. Importantly, we found this regimen has generated complete T cell resistance to TGF-ß-induced functional suppression, with sustained levels of the IFN-γ and Granzyme-B productions. Such findings were also validated in the melanoma-associated antigen recognized by T cells (MART-1) specific T cell receptor (TCR) engineered T cells, which were expanded in Doramapimod and IL-7 + IL-15 added media. In conclusion, we have established and optimized a protocol with the combination of p38 inhibitor, IL-7 and IL-15, rather than IL-2, for the generation of functionally enhanced T cells applicable for ACT.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Naftalenos/farmacología , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Línea Celular , Ingeniería Genética , Granzimas/metabolismo , Humanos , Tolerancia Inmunológica , Interferón gamma/metabolismo , Activación de Linfocitos , Antígeno MART-1/inmunología , Linfocitos T/trasplante , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
6.
J Immunother Cancer ; 9(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34021033

RESUMEN

BACKGROUND: The adoptive transfer of tumor-infiltrating lymphocytes (TIL) has demonstrated robust efficacy in metastatic melanoma patients. Tumor antigen-loaded dendritic cells (DCs) are believed to optimally activate antigen-specific T lymphocytes. We hypothesized that the combined transfer of TIL, containing a melanoma antigen recognized by T cells 1 (MART-1) specific population, with MART-1-pulsed DC will result in enhanced proliferation and prolonged survival of transferred MART-1 specific T cells in vivo ultimately leading to improved clinical responses. DESIGN: We tested the combination of TIL and DC in a phase II clinical trial of patients with advanced stage IV melanoma. HLA-A0201 patients whose early TIL cultures demonstrated reactivity to MART-1 peptide were randomly assigned to receive TIL alone or TIL +DC pulsed with MART-1 peptide. The primary endpoint was to evaluate the persistence of MART-1 TIL in the two arms. Secondary endpoints were to evaluate clinical response and survival. RESULTS: Ten patients were given TIL alone while eight patients received TIL+DC vaccine. Infused MART-1 reactive CD8+ TIL were tracked in the blood over time by flow cytometry and results show good persistence in both arms, with no difference in the persistence of MART-1 between the two arms. The objective response rate was 30% (3/10) in the TIL arm and 50% (4/8) in the TIL+DC arm. All treatments were well tolerated. CONCLUSIONS: The combination of TIL +DC showed no difference in the persistence of MART-1 TIL compared with TIL therapy alone. Although more patients showed a clinical response to TIL+DC therapy, this study was not powered to resolve differences between groups. TRIAL REGISTRATION NUMBER: NCT00338377.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/trasplante , Inmunoterapia Adoptiva , Depleción Linfocítica , Linfocitos Infiltrantes de Tumor/trasplante , Melanoma/terapia , Neoplasias Cutáneas/terapia , Linfocitos T/trasplante , Adolescente , Adulto , Vacunas contra el Cáncer/efectos adversos , Terapia Combinada , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Humanos , Inmunoterapia Adoptiva/efectos adversos , Depleción Linfocítica/efectos adversos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Antígeno MART-1/inmunología , Antígeno MART-1/metabolismo , Masculino , Melanoma/inmunología , Melanoma/metabolismo , Melanoma/secundario , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
7.
Theranostics ; 11(10): 4957-4974, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33754038

RESUMEN

Rationale: TCR-T cell therapy plays a critical role in the treatment of malignant cancers. However, it is unclear how TCR-T cells are affected by PD-L1 molecule in the tumor environment. We performed an in-depth evaluation on how differential expressions of tumor PD-L1 can affect the functionality of T cells. Methods: We used MART-1-specific TCR-T cells (TCR-TMART-1), stimulated with MART-127-35 peptide-loaded MEL-526 tumor cells, expressing different proportions of PD-L1, to perform cellular assays and high-throughput single-cell RNA sequencing. Results: Different clusters of activated or cytotoxic TCR-TMART-1 responded divergently when stimulated with tumor cells expressing different percentages of PD-L1 expression. Compared to control T cells, TCR-TMART-1 were more sensitive to exhaustion, and secreted not only pro-inflammatory cytokines but also anti-inflammatory cytokines with increasing proportions of PD-L1+ tumor cells. The gene profiles of chemokines were modified by increased expression of tumor PD-L1, which concurrently downregulated pro-inflammatory and anti-inflammatory transcription factors. Furthermore, increased expression of tumor PD-L1 showed distinct effects on different inhibitory checkpoint molecules (ICMs). In addition, there was a limited correlation between the enrichment of cell death signaling in tumor cells and T cells and increased tumor PD-L1 expression. Conclusion: Overall, though the effector functionality of TCR-T cells was suppressed by increased expression percentages of tumor PD-L1 in vitro, scRNA-seq profiles revealed that both the anti-inflammatory and pro-inflammatory responses were triggered by a higher expression of tumor PD-L1. This suggests that the sole blockade of tumor PD-L1 might inhibit not only the anti-inflammatory response but also the pro-inflammatory response in the complicated tumor microenvironment. Thus, the outcome of PD-L1 intervention may depend on the final balance among the highly dynamic and heterogeneous immune regulatory circuits.


Asunto(s)
Antígeno B7-H1/inmunología , Linfocitos T/inmunología , Microambiente Tumoral/inmunología , Antígeno B7-H1/genética , Línea Celular Tumoral , Quimiocinas/genética , Quimiocinas/inmunología , Citocinas/genética , Citocinas/inmunología , Pruebas Inmunológicas de Citotoxicidad , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Inmunoterapia Adoptiva , Inflamación/genética , Inflamación/inmunología , Antígeno MART-1/inmunología , Melanoma/inmunología , RNA-Seq , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Análisis de la Célula Individual , Neoplasias Cutáneas/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T/metabolismo , Microambiente Tumoral/genética
8.
J Immunol Methods ; 484-485: 112831, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32758496

RESUMEN

Neoantigen-based cancer immunotherapies hold the promise of being a truly personalized, effective treatment for diverse cancer types. ELISPOT assays, as a powerful experimental technique, can verify the existence of antigen specific T cells to support basic clinical research and monitor clinical trials. However, despite the high sensitivity of ELISPOT assays, detecting immune responses of neoantigen specific T cells in a patient or healthy donor's PBMCs is still extremely difficult, since the frequency of these T cells can be very low. We developed a novel experimental method, by co-stimulation of T cells with anti-CD28 and IL-2 at the beginning of ELISPOT, to further increase the sensitivity of ELISPOT and mitigate the challenge introduced by low frequency T cells. Under the optimal concentration of 1 µg/ml for anti-CD28 and 1 U/ml for IL-2, an 11.7-fold increase of T cell response against CMV peptide was observed by using our method, and it outperforms other cytokine stimulation alternatives (5-10 folds). We also showed that this method can be effectively applied to detect neoantigen-specific T cells in healthy donors' and a melanoma patient's PBMCs. To the best of our knowledge, this is the first report that the co-stimulation of anti-CD28 and IL-2 is able to significantly improve the sensitivity of ELISPOT assays, indicating that anti-CD28 and IL-2 signaling can act in synergy to lower the T cell activation threshold and trigger more neoantigen-specific T cells.


Asunto(s)
Anticuerpos/farmacología , Antígenos de Neoplasias/inmunología , Antígenos CD28/inmunología , Ensayo de Immunospot Ligado a Enzimas , Ensayos de Liberación de Interferón gamma , Interleucina-2/farmacología , Activación de Linfocitos/efectos de los fármacos , Neoplasias/inmunología , Linfocitos T/efectos de los fármacos , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Células Cultivadas , Sinergismo Farmacológico , Humanos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Antígeno MART-1/inmunología , Mutación , Neoplasias/genética , Neoplasias/metabolismo , Fragmentos de Péptidos/inmunología , Reproducibilidad de los Resultados , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Proteínas de la Matriz Viral/inmunología
9.
Eur J Immunol ; 50(7): 949-958, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32112565

RESUMEN

Long-lasting CD8+ T cell responses are critical in combatting infections and tumors. The pro-inflammatory cytokine IFN-γ is a key effector molecule herein. We recently showed that in murine T cells the production of IFN-γ is tightly regulated through adenylate uridylate-rich elements (AREs) that are located in the 3' untranslated region (UTR) of the Ifng mRNA molecule. Loss of AREs resulted in prolonged cytokine production in activated T cells and boosted anti-tumoral T cell responses. Here, we investigated whether these findings can be translated to primary human T cells. Utilizing CRISPR-Cas9 technology, we deleted the ARE region from the IFNG 3' UTR in peripheral blood-derived human T cells. Loss of AREs stabilized the IFNG mRNA in T cells and supported a higher proportion of IFN-γ protein-producing T cells. Importantly, combining MART-1 T cell receptor engineering with ARE-Del gene editing showed that this was also true for antigen-specific activation of T cells. MART-1-specific ARE-Del T cells showed higher percentages of IFN-γ producing T cells in response to MART-1 expressing tumor cells. Combined, our study reveals that ARE-mediated posttranscriptional regulation is conserved between murine and human T cells. Furthermore, generating antigen-specific ARE-Del T cells is feasible, a feature that could potentially be used for therapeutical purposes.


Asunto(s)
Elementos Ricos en Adenilato y Uridilato , Linfocitos T CD8-positivos/inmunología , Interferón gamma/inmunología , Sistemas CRISPR-Cas , Línea Celular Tumoral , Femenino , Humanos , Interferón gamma/genética , Antígeno MART-1/genética , Antígeno MART-1/inmunología , Masculino , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología
10.
ChemMedChem ; 15(9): 799-807, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32162475

RESUMEN

A click-chemistry-based approach was implemented to prepare peptidomimetics designed in silico and made from aromatic azides and a propargylated GIGI-mimicking platform derived from the altered Melan-A/MART-126(27L)-35 antigenic peptide ELAGIGILTV. The CuI -catalyzed Huisgen cycloaddition was carried out on solid support to generate rapidly a first series of peptidomimetics, which were evaluated for their capacity to dock at the interface between the major histocompatibility complex class-I (MHC-I) human leucocyte antigen (HLA)-A2 and T-cell receptors (TCRs). Despite being a weak HLA-A2 ligand, one of these 11 first synthetic compounds bearing a p-nitrobenzyl-triazole side chain was recognized by the receptor proteins of Melan-A/MART-1-specific T-cells. After modification of the N and C termini of this agonist, which was intended to enhance HLA-A2 binding, one of the resulting seven additional compounds triggered significant T-cell responses. Thus, these results highlight the capacity of naturally circulating human TCRs that are specific for the native Melan-A/MART-126-35 peptide to cross-react with peptidomimetics bearing organic motifs structurally different from the native central amino acids.


Asunto(s)
Haptenos/química , Antígeno MART-1/química , Oligopéptidos/síntesis química , Química Clic , Antígeno HLA-A2/inmunología , Haptenos/inmunología , Humanos , Antígeno MART-1/inmunología , Estructura Molecular , Oligopéptidos/química , Oligopéptidos/inmunología , Peptidomiméticos , Receptores de Antígenos de Linfocitos T/inmunología
11.
Appl Immunohistochem Mol Morphol ; 28(9): e76-e81, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-30499815

RESUMEN

BACKGROUND: Metastatic melanoma in sentinel lymph nodes is often elusive to detect with morphology alone. Per American Joint Committee on Cancer staging guidelines, a single atypical melanocyte in lymph node qualifies as metastasis, whether identified by morphology or immunohistochemistry, but single cell staining must be convincing. We propose that the use of a second immunohistochemical run performed on a single slide will allow for more confident diagnosis of micrometastases. MATERIALS AND METHODS: We designed a technical study to determine whether a second antibody application on previously stained slides can successfully detect the same population of cells. Melanocytic neoplasms were stained with SOX-10 using Ventana Benchmark Ultra stainers, coverslipped, and examined, followed by coverslip removal and application of MART-1 (Ventana A103). The order of antibody application and chromagen detection kit (AP-RED vs. DAB) was reversed to establish reliability and robustness of the protocol. RESULTS: All melanocytes marked with SOX-10 and MART-1, and produced a range of staining quality that varied based on order of stain application and chromagen kit were used. The optimal combination was red MART-1 applied first followed by brown SOX-10 applied second. CONCLUSIONS: Consecutive staining of melanocytes with SOX-10 and MART-1 may improve diagnostic confidence of melanocyte identification, particularly in detection of single cell, micrometastases in sentinel lymph nodes or in situations where dual immunohistochemical stains may be unavailable.


Asunto(s)
Inmunohistoquímica/métodos , Metástasis Linfática/diagnóstico , Melanocitos/metabolismo , Melanoma/diagnóstico , Ganglio Linfático Centinela/metabolismo , Neoplasias Cutáneas/diagnóstico , Biomarcadores de Tumor , Diagnóstico Diferencial , Humanos , Metástasis Linfática/patología , Antígeno MART-1/inmunología , Antígeno MART-1/metabolismo , Melanocitos/patología , Melanoma/patología , Estadificación de Neoplasias , Guías de Práctica Clínica como Asunto , Factores de Transcripción SOXE/inmunología , Factores de Transcripción SOXE/metabolismo , Ganglio Linfático Centinela/patología , Análisis de la Célula Individual , Neoplasias Cutáneas/patología , Coloración y Etiquetado
12.
Xenotransplantation ; 27(1): e12558, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31565822

RESUMEN

BACKGROUND: Tolerance-inducing approaches to xenotransplantation would be optimal and may be necessary for long-term survival of transplanted pig organs in human patients. The ideal approach would generate donor-specific unresponsiveness to the pig organ without suppressing the patient's normal immune function. Porcine thymus transplantation has shown efficacy in promoting xenotolerance in humanized mice and large animal models. However, murine studies demonstrate that T cells selected in a swine thymus are positively selected only by swine thymic epithelial cells, and therefore, cells expressing human HLA-restricted TCRs may not be selected efficiently in a transplanted pig thymus. This may lead to suboptimal patient immune function. METHODS: To assess human thymocyte selection in a pig thymus, we used a TCR transgenic humanized mouse model to study positive selection of cells expressing the MART1 TCR, a well-characterized human HLA-A2-restricted TCR, in a grafted pig thymus. RESULTS: Positive selection of T cells expressing the MART1 TCR was inefficient in both a non-selecting human HLA-A2- or swine thymus compared with an HLA-A2+ thymus. Additionally, CD8 MART1 TCRbright T cells were detected in the spleens of mice transplanted with HLA-A2+ thymi but were significantly reduced in the spleens of mice transplanted with swine or HLA-A2- thymi. [Correction added on October 15, 2019, after first online publication: The missing superscript values +, -, and bright have been included in the Results section.] CONCLUSIONS: Positive selection of cells expressing a human-restricted TCR in a transplanted pig thymus is inefficient, suggesting that modifications to improve positive selection of cells expressing human-restricted TCRs in a pig thymus may be necessary to support development of a protective human T-cell pool in future patients.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo , Timo/fisiología , Animales , Células Cultivadas , Selección Clonal Mediada por Antígenos , Antígeno HLA-A2/metabolismo , Humanos , Tolerancia Inmunológica , Antígeno MART-1/inmunología , Ratones , Ratones SCID , Ratones Transgénicos , Trasplante de Órganos , Porcinos , Trasplante Heterólogo
13.
Front Immunol ; 10: 2115, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31555299

RESUMEN

The efficacy of T cells depends on their functional avidity, i. e., the strength of T cell interaction with cells presenting cognate antigen. The overall T cell response is composed of multiple T cell clonotypes, involving different T cell receptors and variable levels of functional avidity. Recently, it has been proposed that the presence of low avidity tumor antigen-specific CD8 T cells hinder their high avidity counterparts to protect from tumor growth. Here we analyzed human cytotoxic CD8 T cells specific for the melanoma antigen Melan-A/MART-1. We found that the presence of low avidity T cells did not result in reduced cytotoxicity of tumor cells, nor reduced cytokine production, by high avidity T cells. In vivo in NSG-HLA-A2 mice, the anti-tumor effect of high avidity T cells was similar in presence or absence of low avidity T cells. These data indicate that low avidity T cells are not hindering anti-tumor T cell responses, a finding that is reassuring because low avidity T cells are an integrated part of natural T cell responses.


Asunto(s)
Afinidad de Anticuerpos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/uso terapéutico , Melanoma/inmunología , Animales , Citotoxicidad Inmunológica/inmunología , Xenoinjertos , Humanos , Antígeno MART-1/inmunología , Ratones , Células Tumorales Cultivadas
14.
Otol Neurotol ; 40(7): e674-e678, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31295198

RESUMEN

OBJECTIVE: To illustrate a case of sensorineural hearing loss (SNHL) after immunotherapy based on T cell receptor (TCR) gene therapy using modified T cells recognizing melanoma antigen recognized by T cells 1 for disseminated melanoma. PATIENT: We present a 59-year-old woman with profound subacute bilateral SNHL including unilateral deafness after immunotherapy based on TCR gene therapy using modified T cells recognizing melanoma antigen recognized by T cells 1 for disseminated melanoma. Ten days after treatment, the patient developed hearing loss of 57 dB hearing loss air conduction at pure-tone average 0.5-1-2-4 kHz in the right ear, and >100 dB hearing loss air conduction at pure-tone average 0.5-1-2-4 in the left ear. The right ear recovered partially, while the left ear remained deaf, despite oral prednisolone (1.0 mg/kg) and salvage treatment with three transtympanic injections of 0.5 ml dexamethasone (4.0 mg/ml). CONCLUSION: Based on our presented case and a vast amount of literature there is circumstantial evidence that TCR gene therapy for melanoma targets the perivascular macrophage-like melanocytes in the stria vascularis, resulting in SNHL. We suggest that SNHL after TCR gene therapy may be caused by a disruption of the blood-labyrinth-barrier and the endolymphatic potential and/or a sterile inflammation of the stria vascularis. In severe cases like our subject, we posit that endolymphatic hydrops or hair cell loss may cause irreversible and asymmetrical deafness. Steroid prophylaxis via transtympanic application is debatable.


Asunto(s)
Pérdida Auditiva Sensorineural/etiología , Inmunoterapia Adoptiva/efectos adversos , Melanoma/terapia , Neoplasias Cutáneas/terapia , Femenino , Humanos , Antígeno MART-1/inmunología , Persona de Mediana Edad , Melanoma Cutáneo Maligno
15.
Cancer Immunol Immunother ; 68(5): 709-720, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30783693

RESUMEN

We used four-color ImmunoSpot® assays, in conjunction with peptide pools that cover the sequence of tyrosinase (Tyr), melanoma-associated antigen A3 (MAGE-A3), melanocyte antigen/melanoma antigen recognized by T cells 1 (Melan-A/MART-1), glycoprotein 100 (gp100), and New York esophageal squamous cell carcinoma-1 (NY-ESO-1) to characterize the melanoma antigen (MA)-specific CD8 + cell repertoire in PBMC of 40 healthy human donors (HD). Tyr triggered interferon gamma (IFN-γ)-secreting CD8 + T cells in 25% of HD within 24 h of antigen stimulation ex vivo. MAGE-A3, Melan-A/MART-1, and gp100 also induced recall responses in 10%, 7.5%, and 2.5% of HD, respectively. At this time point, these CD8 + T cells did not yet produce GzB (granzyme B). However, they engaged in GzB production after 72 h of antigen stimulation. By this 72-h time point, 57.5% of the HD responded to at least one, and typically several, of the MA. A closer characterization of the Tyr-specific CD8 + T cell repertoire indicated that it was low-affinity, and to primarily entail a stem cell-like subpopulation. Collectively, our data reveal pre-existing endogenous T cell immunity against melanoma antigens in healthy donors, and analogous to natural autoantibodies, we have termed this "natural T cell autoreactivity".


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Ensayo de Immunospot Ligado a Enzimas/métodos , Monofenol Monooxigenasa/inmunología , Antígenos de Neoplasias/inmunología , Autoinmunidad , Proliferación Celular , Células Clonales , Voluntarios Sanos , Humanos , Memoria Inmunológica , Interferón gamma/metabolismo , Activación de Linfocitos , Antígeno MART-1/inmunología , Proteínas de Neoplasias/inmunología , Antígeno gp100 del Melanoma/inmunología
16.
Front Immunol ; 10: 63, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30778347

RESUMEN

Thymic selection constitutes the first checkpoint in T-cell development to purge autoreactive T cells. Most of our understanding of this process comes from animal models because of the challenges of studying thymopoiesis and how T cell receptor (TCR) specificity impacts thymocyte phenotype in humans. We developed a humanized mouse model involving the introduction of autoreactive TCRs and cognate autoantigens that enables the analysis of selection of human T cells in human thymic tissue in vivo. Here, we describe the thymic development of MART1-specific autoreactive CD8+ T cells that normally escape deletion and how their phenotype and survival are affected by introduction of the missing epitope in the hematopoietic lineage. Expression of the epitope in a fraction of hematopoietic cells, including all major types of antigen-presenting cells (APCs), led to profound yet incomplete deletion of these T cells. Upregulation of PD-1 upon antigen encounter occurred through the different stages of thymocyte development. PD-1 and CCR7 expression were mutually exclusive in both transgenic and non-transgenic thymocytes, challenging the view that CCR7 is necessary for negative selection in humans. In the presence of antigen, MART1-reactive T cells down-regulated TCR, CD3, CD8, and CD4 in the thymus and periphery. Moreover, expression of secondary TCRs influences MHC class I-restricted T cells to develop as CD4+, particularly regulatory T cells. This new model constitutes a valuable tool to better understand the development of autoreactive T cells identified in different human autoimmune diseases and the role of different APC subsets in their selection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Timo/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Femenino , Humanos , Antígeno MART-1/inmunología , Masculino , Ratones , Ratones Transgénicos , Timectomía
17.
Biomater Sci ; 7(3): 773-788, 2019 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-30601510

RESUMEN

The clinical success of dendritic cell (DC)-based genetic immunization remains critically dependent on the availability of effective and safe nano-carriers for targeting antigen-encoded DNA vaccines to DCs, the most potent antigen-presenting cells in the human body in vivo. Recent studies revealed the efficacies of mannose receptor-mediated in vivo DC-targeted genetic immunization by liposomal DNA vaccine carriers containing both mannose-mimicking shikimoyl and transfection enhancing guanidinyl functionalities. However, to date, the efficacies of this approach have not been examined for metal-based nanoparticle DNA vaccine carriers. Herein, we report for the first time, the design, synthesis, physico-chemical characterization and bioactivities of gold nanoparticles covalently functionalized with a thiol ligand containing both shikimoyl and guanidinyl functionalities (Au-SGSH). We show that Au-SGSH nanoparticles can deliver DNA vaccines to mouse DCs under in vivo conditions. Subcutaneous administration of near infrared (NIR) dye-labeled Au-SGSH showed significant accumulation of the NIR dye in the DCs of the nearby lymph nodes compared to that for the non-targeting NIR-labeled Au-GSH nanoconjugate containing only a covalently tethered guanidinyl group, not the shikimoyl-functionality. Under prophylactic settings, in vivo immunization (s.c.) with the Au-SGSH-pCMV-MART1 nanoplex induced a long-lasting (180 days) immune response against murine melanoma. Notably, mannose receptor-mediated in vivo DC-targeted immunization (s.c.) with the Au-SGSH-MART1 nanoplex significantly inhibited established melanoma growth and increased the overall survivability of melanoma-bearing mice under therapeutic settings. The Au-SGSH nanoparticles reported herein have potential use for in vivo DC-targeted genetic immunization against cancer and infectious diseases.


Asunto(s)
Células Dendríticas/metabolismo , Oro/química , Nanopartículas del Metal/química , Nanoconjugados/química , Vacunas de ADN/inmunología , Animales , Células de la Médula Ósea/citología , Línea Celular Tumoral , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/citología , Células Dendríticas/inmunología , Femenino , Colorantes Fluorescentes/química , Inmunidad Activa , Antígeno MART-1/química , Antígeno MART-1/inmunología , Masculino , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/prevención & control , Nanopartículas del Metal/toxicidad , Ratones , Ratones Endogámicos C57BL , Plásmidos/química , Plásmidos/metabolismo , Compuestos de Sulfhidrilo/química , Vacunas de ADN/química
18.
Mol Ther ; 27(2): 300-313, 2019 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30617019

RESUMEN

T cell receptors (TCRs) have emerged as a new class of immunological therapeutics. However, though antigen specificity is a hallmark of adaptive immunity, TCRs themselves do not possess the high specificity of monoclonal antibodies. Although a necessary function of T cell biology, the resulting cross-reactivity presents a significant challenge for TCR-based therapeutic development, as it creates the potential for off-target recognition and immune toxicity. Efforts to enhance TCR specificity by mimicking the antibody maturation process and enhancing affinity can inadvertently exacerbate TCR cross-reactivity. Here we demonstrate this concern by showing that even peptide-targeted mutations in the TCR can introduce new reactivities against peptides that bear similarity to the original target. To counteract this, we explored a novel structure-guided approach for enhancing TCR specificity independent of affinity. Tested with the MART-1-specific TCR DMF5, our approach had a small but discernible impact on cross-reactivity toward MART-1 homologs yet was able to eliminate DMF5 cross-recognition of more divergent, unrelated epitopes. Our study provides a proof of principle for the use of advanced structure-guided design techniques for improving TCR specificity, and it suggests new ways forward for enhancing TCRs for therapeutic use.


Asunto(s)
Receptores de Antígenos de Linfocitos T/metabolismo , Inmunidad Adaptativa/fisiología , Anticuerpos Monoclonales/inmunología , Humanos , Antígeno MART-1/inmunología , Estructura Secundaria de Proteína , Resonancia por Plasmón de Superficie , Especificidad del Receptor de Antígeno de Linfocitos T
19.
Immunol Cell Biol ; 97(4): 427-434, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30633397

RESUMEN

The differentiation of human hematopoietic stem cells into CD8 T cells can be achieved in vitro with the OP9-DL4 system. We considered that in the absence of medullary thymic epithelial cells, which serve to restrict the breath of the T-cell receptor (TCR) repertoire by expressing tissue-restricted antigens, a distinct repertoire would be generated in vitro. To test this notion, we compared the TCR-Vα/Vß (TRAV/TRBV) gene usage of major histocompatibility complex-restricted antigen (MART-1)-specific T cells generated in vitro to that from ex vivo naïve T cells and tumor-infiltrating lymphocytes (TILs) using high-throughput DNA sequencing. In contrast to naïve T cells and TILs, which showed the expected narrow TRAV repertoire, in vitro-generated MART-1-specific T cells used almost all TRAV gene families and displayed unique CDR3 lengths. Our work demonstrates that the OP9-DL4 system supports the creation of a broad antigen-specific TCR repertoire, suggesting that T cells generated in vitro may undergo a different set of selection events that otherwise constrains the TCR repertoire of thymus-derived T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Antígeno MART-1/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Línea Celular , Regiones Determinantes de Complementariedad/genética , Células Madre Hematopoyéticas/metabolismo , Humanos
20.
Melanoma Res ; 29(4): 420-427, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30520800

RESUMEN

A vaccine that could expand melanoma-specific T cells might reduce the risk of recurrence of resected melanoma and could provide an alternative or adjunct to standard immunotherapy options. We tested the safety and immunogenicity of a vaccine coupling a melanoma-associated peptide with a xenogenic peptide (to promote epitope spreading) and/or resiquimod (to activate antigen-presenting cells). HLA-A2-positive patients with resected stage II, III, and IV melanoma were assigned to treatment on one of three schedules. All patients received three subcutaneous doses of the peptide MART-1a mixed with Montanide. In addition, patients on schedule 1 received the xenoantigen peptide Gag267-274, patients on schedule 2 received topical resiquimod, and patients on schedule 3 received both Gag267-274 and resiquimod. Blood samples were tested for the frequency of antigen-specific T cells by tetramer assay, as well as immune cell subtypes and plasma cytokine levels. Patients enrolled from October 2012 to December 2014, with 10 patients enrolling to each schedule. The most common adverse events were injection site reaction (26 patients) and fatigue (15 patients). Tetramer analysis revealed antigen-specific responses (defined as doubling of MART-1a-specific T cells from pretreatment to post-treatment) in 20, 60, and 40% of patients treated on schedules 1, 2, and 3, respectively. Vaccine treatment consisting of MART-1a peptide, Gag267-274, Montanide, and topical resiquimod was well-tolerated. The addition of the Gag267-274 xenoantigen was not associated with an increase in the response to MART-1a, whereas use of topical resiquimod was associated with a higher frequency of MART-1a-specific T-cell responses that did not meet statistical significance.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Imidazoles/uso terapéutico , Antígeno MART-1/inmunología , Melanoma/terapia , Neoplasias Cutáneas/terapia , Administración Tópica , Anciano , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Terapia Combinada , Citocinas/inmunología , Femenino , Productos del Gen gag/inmunología , Antígeno HLA-A2/inmunología , Humanos , Masculino , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Persona de Mediana Edad , Fragmentos de Péptidos/inmunología , Proyectos Piloto , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...