Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Blood ; 136(10): 1155-1160, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32573723

RESUMEN

Hematological and solid cancers catabolize the semiessential amino acid arginine to drive cell proliferation. However, the resulting low arginine microenvironment also impairs chimeric antigen receptor T cells (CAR-T) cell proliferation, limiting their efficacy in clinical trials against hematological and solid malignancies. T cells are susceptible to the low arginine microenvironment because of the low expression of the arginine resynthesis enzymes argininosuccinate synthase (ASS) and ornithine transcarbamylase (OTC). We demonstrate that T cells can be reengineered to express functional ASS or OTC enzymes, in concert with different chimeric antigen receptors. Enzyme modifications increase CAR-T cell proliferation, with no loss of CAR cytotoxicity or increased exhaustion. In vivo, enzyme-modified CAR-T cells lead to enhanced clearance of leukemia or solid tumor burden, providing the first metabolic modification to enhance CAR-T cell therapies.


Asunto(s)
Arginina/metabolismo , Argininosuccinato Sintasa/metabolismo , Inmunoterapia Adoptiva/métodos , Leucemia Mieloide Aguda/terapia , Neuroblastoma/terapia , Ornitina Carbamoiltransferasa/metabolismo , Linfocitos T/trasplante , Animales , Apoptosis , Argininosuccinato Sintasa/genética , Proliferación Celular , Humanos , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ingeniería Metabólica/métodos , Ratones , Ratones Desnudos , Neuroblastoma/inmunología , Neuroblastoma/metabolismo , Neuroblastoma/patología , Ornitina Carbamoiltransferasa/genética , Receptores Quiméricos de Antígenos/química , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
PLoS Biol ; 16(9): e2005046, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30180168

RESUMEN

The microenvironment of lymphoid organs can aid healthy immune function through provision of both structural and molecular support. In mice, fibroblastic reticular cells (FRCs) create an essential T-cell support structure within lymph nodes, while human FRCs are largely unstudied. Here, we show that FRCs create a regulatory checkpoint in human peripheral T-cell activation through 4 mechanisms simultaneously utilised. Human tonsil and lymph node-derived FRCs constrained the proliferation of both naïve and pre-activated T cells, skewing their differentiation away from a central memory T-cell phenotype. FRCs acted unilaterally without requiring T-cell feedback, imposing suppression via indoleamine-2,3-dioxygenase, adenosine 2A Receptor, prostaglandin E2, and transforming growth factor beta receptor (TGFßR). Each mechanistic pathway was druggable, and a cocktail of inhibitors, targeting all 4 mechanisms, entirely reversed the suppressive effect of FRCs. T cells were not permanently anergised by FRCs, and studies using chimeric antigen receptor (CAR) T cells showed that immunotherapeutic T cells retained effector functions in the presence of FRCs. Since mice were not suitable as a proof-of-concept model, we instead developed a novel human tissue-based in situ assay. Human T cells stimulated using standard methods within fresh tonsil slices did not proliferate except in the presence of inhibitors described above. Collectively, we define a 4-part molecular mechanism by which FRCs regulate the T-cell response to strongly activating events in secondary lymphoid organs while permitting activated and CAR T cells to utilise effector functions. Our results define 4 feasible strategies, used alone or in combinations, to boost primary T-cell responses to infection or cancer by pharmacologically targeting FRCs.


Asunto(s)
Diferenciación Celular/inmunología , Microambiente Celular , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Linfocitos T/citología , Adulto , Proliferación Celular , Niño , Fibroblastos/citología , Humanos , Memoria Inmunológica , Fenotipo
3.
Haematologica ; 104(9): 1892-1905, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30573509

RESUMEN

Ca2+ entry via Orai1 store-operated Ca2+ channels in the plasma membrane is critical to cell function, and Orai1 loss causes severe immunodeficiency and developmental defects. The tetraspanins are a superfamily of transmembrane proteins that interact with specific 'partner proteins' and regulate their trafficking and clustering. The aim of this study was to functionally characterize tetraspanin Tspan18. We show that Tspan18 is expressed by endothelial cells at several-fold higher levels than most other cell types analyzed. Tspan18-knockdown primary human umbilical vein endothelial cells have 55-70% decreased Ca2+ mobilization upon stimulation with the inflammatory mediators thrombin or histamine, similar to Orai1-knockdown. Tspan18 interacts with Orai1, and Orai1 cell surface localization is reduced by 70% in Tspan18-knockdown endothelial cells. Tspan18 overexpression in lymphocyte model cell lines induces 20-fold activation of Ca2+ -responsive nuclear factor of activated T cell (NFAT) signaling, in an Orai1-dependent manner. Tspan18-knockout mice are viable. They lose on average 6-fold more blood in a tail-bleed assay. This is due to Tspan18 deficiency in non-hematopoietic cells, as assessed using chimeric mice. Tspan18-knockout mice have 60% reduced thrombus size in a deep vein thrombosis model, and 50% reduced platelet deposition in the microcirculation following myocardial ischemia-reperfusion injury. Histamine- or thrombin-induced von Willebrand factor release from endothelial cells is reduced by 90% following Tspan18-knockdown, and histamine-induced increase of plasma von Willebrand factor is reduced by 45% in Tspan18-knockout mice. These findings identify Tspan18 as a novel regulator of endothelial cell Orai1/Ca2+ signaling and von Willebrand factor release in response to inflammatory stimuli.


Asunto(s)
Calcio/metabolismo , Daño por Reperfusión Miocárdica/genética , Proteína ORAI1/genética , Tetraspaninas/genética , Trombosis de la Vena/genética , Factor de von Willebrand/genética , Animales , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Pollos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Histamina/farmacología , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Transporte Iónico/efectos de los fármacos , Células Jurkat , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Proteína ORAI1/metabolismo , Transducción de Señal , Tetraspaninas/metabolismo , Trombina/farmacología , Trombosis de la Vena/metabolismo , Trombosis de la Vena/patología , Factor de von Willebrand/metabolismo
4.
J Immunother Cancer ; 12(2)2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38413223

RESUMEN

BACKGROUND: Targeting of solid cancers with chimeric antigen receptor (CAR)-T cells is limited by the lack of suitable tumor-specific antigens and the immunosuppressive, desmoplastic tumor microenvironment that impedes CAR-T cell infiltration, activity and persistence. We hypothesized that targeting the endosialin (CD248) receptor, strongly expressed by tumor-associated pericytes and perivascular cancer-associated fibroblasts, would circumvent these challenges and offer an exciting antigen for CAR-T cell therapy due to the close proximity of target cells to the tumor vasculature, the limited endosialin expression in normal tissues and the lack of phenotype observed in endosialin knockout mice. METHODS: We generated endosialin-directed E3K CAR-T cells from three immunocompetent mouse strains, BALB/c, FVB/N and C57BL/6. E3K CAR-T cell composition (CD4+/CD8+ ratio), activity in vitro against endosialin+ and endosialin- cells, and expansion and activity in vivo in syngeneic tumor models as well as in tumor-naive healthy and wounded mice and tumor-bearing endosialin knockout mice was assessed. RESULTS: E3K CAR-T cells were active in vitro against both mouse and human endosialin+, but not endosialin-, cells. Adoptively transferred E3K CAR-T cells exhibited no activity in endosialin knockout mice, tumor-naive endosialin wildtype mice or in wound healing models, demonstrating an absence of off-target and on-target/off-tumor activity. By contrast, adoptive transfer of E3K CAR-T cells into BALB/c, FVB/N or C57BL/6 mice bearing syngeneic breast or lung cancer lines depleted target cells in the tumor stroma resulting in increased tumor necrosis, reduced tumor growth and a substantial impairment in metastatic outgrowth. CONCLUSIONS: Together these data highlight endosialin as a viable antigen for CAR-T cell therapy and that targeting stromal cells closely associated with the tumor vasculature avoids CAR-T cells having to navigate the harsh immunosuppressive tumor microenvironment. Further, the ability of E3K CAR-T cells to recognize and target both mouse and human endosialin+ cells makes a humanized and optimized E3K CAR a promising candidate for clinical development applicable to a broad range of solid tumor types.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Ratones , Animales , Pericitos/metabolismo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Ratones Endogámicos C57BL , Neoplasias/metabolismo , Linfocitos T/metabolismo , Ratones Noqueados , Microambiente Tumoral , Antígenos de Neoplasias/metabolismo , Antígenos CD/metabolismo
5.
Am J Pathol ; 180(3): 1215-1222, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22226739

RESUMEN

The substantial T lymphocyte infiltrate found in cases of nasopharyngeal carcinoma (NPC) has been implicated in the promotion of both tumor growth and immune escape. Conversely, because malignant NPC cells harbor the Epstein-Barr virus, this tumor is a candidate for virus-specific T cell-based therapies. Preventing the accumulation of tumor-promoting T cells or enhancing the recruitment of tumor-specific cytotoxic T cells offers therapeutic potential. However, the mechanisms involved in T cell recruitment to this tumor are poorly understood. Comparing memory T cell subsets that have naturally infiltrated NPC tissue with their counterparts from matched blood revealed enrichment of CD8(+), CD4(+), and regulatory T cells expressing the chemokine receptor CXCR6 in tumor tissue. CD8(+) and (nonregulatory) CD4(+) T cells also were more frequently CCR5(+) in tumor than in blood. Ex vivo studies demonstrated that both receptors were functional. CXCL16 and CCL4, unique chemokine ligands for CXCR6 and CCR5, respectively, were expressed by the malignant cells in tumor tissue from the majority of NPC cases, as was another CCR5 ligand, CCL5. The strongest expression of CXCL16 was found on tumor-infiltrating cells. CCL4 was detected on the tumor vasculature in a majority of cases. These findings suggest that CXCR6 and CCR5 play important roles in T cell recruitment and/or retention in NPC and have implications for the pathogenesis and treatment of this tumor.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias Nasofaríngeas/inmunología , Receptores CCR5/metabolismo , Receptores de Quimiocina/metabolismo , Receptores Virales/metabolismo , Subgrupos de Linfocitos T/inmunología , Carcinoma , Línea Celular Tumoral , Humanos , Memoria Inmunológica/fisiología , Linfocitos Infiltrantes de Tumor/inmunología , Carcinoma Nasofaríngeo , Receptores CCR5/fisiología , Receptores CXCR6 , Receptores de Quimiocina/fisiología , Receptores Virales/fisiología
6.
Cancer Immunol Immunother ; 61(5): 615-28, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22021067

RESUMEN

4-1BB ligation co-stimulates T cell activation, and agonistic antibodies have entered clinical trials. Natural killer (NK) cells also express 4-1BB following activation and are implicated in the anti-tumour efficacy of 4-1BB stimulation in mice; however, the response of human NK cells to 4-1BB stimulation is not clearly defined. Stimulation of non-adherent PBMC with OVCAR-3 cells expressing 4-1BB ligand (4-1BBL) or IL-12 resulted in preferential expansion of the NK cell population, while the combination 4-1BBL + IL-12 was superior for the activation and proliferation of functional NK cells from healthy donors and patients with renal cell or ovarian carcinoma, supporting long-term (21 day) NK cell proliferation. The expanded NK cells are predominantly CD56(bright), and we show that isolated CD56(dim)CD16(+) NK cells can switch to a CD56(bright)CD16(-) phenotype and proliferate in response to 4-1BBL + IL-12. Whereas 4-1BB upregulation on NK cells in response to 4-1BBL required 'help' from other PBMC, it could be induced on isolated NK cells by IL-12, but only in the presence of target (OVCAR-3) cells. Following primary stimulation with OVCAR-3 cells expressing 4-1BBL + IL-12 and subsequent resting until day 21, NK cells remained predominantly CD56(bright) and retained both high cytotoxic capability against K562 targets and enhanced ability to produce IFNγ relative to NK cells in PBMC. These data support the concept that NK cells could contribute to anti-tumour activity of 4-1BB agonists in humans and suggest that combining 4-1BB-stimulation with IL-12 could be beneficial for ex vivo or in vivo expansion and activation of NK cells for cancer immunotherapy.


Asunto(s)
Ligando 4-1BB/inmunología , Interleucina-12/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Neoplasias/inmunología , Neoplasias/patología , Ligando 4-1BB/biosíntesis , Ligando 4-1BB/genética , Antígeno CD56/inmunología , Procesos de Crecimiento Celular/genética , Procesos de Crecimiento Celular/inmunología , Línea Celular Tumoral , Femenino , Proteínas Ligadas a GPI/inmunología , Humanos , Interferón gamma/inmunología , Interleucina-12/biosíntesis , Interleucina-12/genética , Leucocitos Mononucleares/inmunología , Activación de Linfocitos , Neoplasias/genética , Fenotipo , Receptores de IgG/inmunología , Regulación hacia Arriba
7.
Blood ; 116(19): 3695-704, 2010 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-20671118

RESUMEN

Therapeutic targeting of virus-encoded proteins using cellular immunotherapy has proved successful for Epstein-Barr virus (EBV)-associated posttransplant lymphoproliferative disease. However, the more limited repertoire and immunogenicity of EBV-encoded proteins in other malignancies such as Hodgkin lymphoma and extranodal natural killer (NK)/T lymphoma has been more challenging to target. The immunosubdominant latent membrane protein 2 (LMP2) is considered the optimal target in such Latency II tumors, although data relating to its expression in T/NK malignancies are limited. In addressing the validity of LMP2 as an immunotherapeutic target we found that LMP2-specific effector CD8(+) T cells recognized and killed EBV-positive NK- and T-cell tumor lines, despite an apparent absence of LMP2A protein and barely detectable levels of LMP2 transcripts from the conventional LMP2A and LMP2B promoters. We resolved this paradox by identifying in these lines a novel LMP2 mRNA, initiated from within the EBV terminal repeats and containing downstream, epitope-encoding exons. This same mRNA was also highly expressed in primary (extra-nodal) NK/T lymphoma tissue, with virtually undetectable levels of conventional LMP2A/B transcripts. Expression of this novel transcript in T/NK-cell lymphoproliferative diseases validates LMP2 as an attractive target for cellular immunotherapy and implicates this truncated LMP2 protein in NK- and T-cell lymphomagenesis. This study is registered at clinicaltrials.gov as NCT00062868.


Asunto(s)
Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/inmunología , Leucemia Linfocítica Granular Grande/inmunología , Leucemia Linfocítica Granular Grande/virología , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/inmunología , Secuencia de Bases , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Cartilla de ADN/genética , Infecciones por Virus de Epstein-Barr/terapia , Expresión Génica , Genes Virales , Herpesvirus Humano 4/patogenicidad , Humanos , Inmunoterapia Adoptiva , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/virología , Leucemia Linfocítica Granular Grande/terapia , Linfoma Extranodal de Células NK-T/inmunología , Linfoma Extranodal de Células NK-T/terapia , Linfoma Extranodal de Células NK-T/virología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Linfocitos T/inmunología , Linfocitos T/virología
8.
Mol Pharmacol ; 79(3): 411-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21149638

RESUMEN

Verapamil is a prototypical phenylalkylamine (PAA), and it was the first calcium channel blocker to be used clinically. It tonically blocks L-type channels in the inner pore with micromolar affinity, and its affinity increases at depolarized membrane potentials. In T-type calcium channels, verapamil blocks with micromolar affinity and has modestly increased affinity at depolarized potentials. We found that a related PAA, 4-desmethoxyverapamil (D888), is comparable with verapamil both in affinity and in state-dependence. Permanently charged verapamil was more effective intracellularly than neutral verapamil. Charged PAAs were able to access their binding site from both inside and outside the cell. Furthermore, membrane-impermeant [2-(trimethylammonium)ethyl]methanethiosulfonate was able to access the inner pore from outside of the cell. We examined a homology model of the T-type calcium channel to look for possible routes of drug entry. Mutation of L1825W produced a channel that was blocked significantly more slowly by charged verapamil from the outside, with an increase in apparent affinity when the drug was applied from the inside. Data suggest that T-type channels have a back pathway through which charged drugs can access the inner pore of the channel without passing through the plasma membrane.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo T/efectos de los fármacos , Verapamilo/farmacología , Canales de Calcio Tipo T/química , Relación Dosis-Respuesta a Droga , Espacio Extracelular/efectos de los fármacos , Células HEK293 , Humanos , Potenciales de la Membrana/efectos de los fármacos , Mutagénesis Sitio-Dirigida , Estructura Terciaria de Proteína , Verapamilo/análogos & derivados
9.
J Immunol ; 182(4): 1919-28, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19201845

RESUMEN

CD8(+) T cells specific for EBV latent cycle epitopes can be reactivated in vitro by stimulating with the autologous EBV-transformed B lymphoblastoid cell line (LCL). The resultant CD8(+) clones kill epitope peptide-loaded targets, but frequently do not kill or show only low levels of lysis of the unmanipulated LCL in 5-h cytotoxicity assays. However, they reproducibly show clear LCL recognition in cytokine (IFN-gamma) release assays and inhibit LCL outgrowth in long-term coculture assays. We show that this growth inhibition is not mediated by cytokines, but by slow killing detectable in extended cytotoxicity assays. The paradoxical earlier findings reflect the fact that cytokine assays are more sensitive indicators of Ag-specific recognition in situations in which the target population is heterogeneous at the single-cell level in terms of epitope display. Such heterogeneity exists within LCLs with, at any one time, subpopulations showing large differences in sensitivity to T cell detection. These differences are not cell cycle related, but correlate with differing levels of EBV latent membrane protein (LMP)1 expression at the single-cell level. In this study, LMP1 is not itself a CD8(+) T cell target, but its expression enhances Ag-processing capacity and HLA class I expression. We propose that LMP1 levels fluctuate cyclically in individual cells and, over time, all cells within a LCL pass through a LMP1(high) T cell-detectable phase.


Asunto(s)
Linfocitos B/virología , Linfocitos T CD8-positivos/inmunología , Activación de Linfocitos/inmunología , Proteínas de la Matriz Viral/inmunología , Presentación de Antígeno/inmunología , Linfocitos B/inmunología , Western Blotting , Linfocitos T CD8-positivos/virología , Línea Celular Transformada , Transformación Celular Viral/inmunología , Citocinas/biosíntesis , Citotoxicidad Inmunológica/inmunología , Ensayo de Inmunoadsorción Enzimática , Epítopos de Linfocito T/inmunología , Herpesvirus Humano 4/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Proteínas de la Matriz Viral/biosíntesis
10.
CBE Life Sci Educ ; 20(1): es2, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33635126

RESUMEN

Maintaining your research team's productivity during the COVID-19 era can be a challenge. Developing new strategies to mentor your research trainees in remote work environments will not only support research productivity and progress toward degree, but also help to keep your mentees' academic and research careers on track. We describe a three-step process grounded in reflective practice that research mentors and mentees can use together to reassess, realign, and reimagine their mentoring relationships to enhance their effectiveness, both in the current circumstances and for the future. Drawing on evidence-based approaches, a series of questions for mentees around documented mentoring competencies provide structure for remote mentoring plans. Special consideration is given to how these plans must address the psychosocial needs and diverse backgrounds of mentors and mentees in the unique conditions that require remote interactions.


Asunto(s)
COVID-19 , Tutoría , Humanos , Mentores , SARS-CoV-2 , Estudiantes
11.
J Exp Med ; 199(10): 1409-20, 2004 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-15148339

RESUMEN

The Epstein-Barr virus (EBV) nuclear antigen (EBNA)1 contains a glycine-alanine repeat (GAr) domain that appears to protect the antigen from proteasomal breakdown and, as measured in cytotoxicity assays, from major histocompatibility complex (MHC) class I-restricted presentation to CD8+ T cells. This led to the concept of EBNA1 as an immunologically silent protein that although unique in being expressed in all EBV malignancies, could not be exploited as a CD8 target. Here, using CD8+ T cell clones to native EBNA1 epitopes upstream and downstream of the GAr domain and assaying recognition by interferon gamma release, we show that the EBNA1 naturally expressed in EBV-transformed lymphoblastoid cell lines (LCLs) is in fact presented to CD8+ T cells via a proteasome/peptide transporter-dependent pathway. Furthermore, LCL recognition by such CD8+ T cells, although slightly lower than seen with paired lines expressing a GAr-deleted EBNA1 protein, leads to strong and specific inhibition of LCL outgrowth in vitro. Endogenously expressed EBNA1 is therefore accessible to the MHC class I pathway despite GAr-mediated stabilization of the mature protein. We infer that EBNA1-specific CD8+ T cells do play a role in control of EBV infection in vivo and might be exploitable in the control of EBV+ malignancies.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Antígenos Nucleares del Virus de Epstein-Barr/inmunología , Alanina , Secuencia de Aminoácidos , Linfoma de Burkitt , Línea Celular Tumoral , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/inmunología , Antígenos Nucleares del Virus de Epstein-Barr/genética , Glicina , Herpesvirus Humano 4/inmunología , Humanos , Complejo Mayor de Histocompatibilidad , Fragmentos de Péptidos/inmunología , Secuencias Repetitivas de Aminoácido
12.
Drug News Perspect ; 23(4): 221-8, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20520851

RESUMEN

Epstein-Barr virus (EBV) is present in the malignant cells of several human cancers including post-transplant lymphoproliferative disease (PTLD), Hodgkin's lymphoma, nasopharyngeal carcinoma, natural killer/T lymphomas and Burkitt's lymphoma. Yet in > 90% of the world's adult population, who carry EBV as a lifelong asymptomatic infection, the oncogenic potential of this virus is controlled by a strong virus-specific T-cell response. Accordingly, EBV-associated malignancies represent good candidates for a T-cell-based therapy and provide an important model for developing such therapies for other human cancers. This review summarizes the impressive results seen with T-cell therapy for PTLD and discusses, in the light of recent technological advances, the prospects for developing more effective approaches for other EBV-associated cancers.


Asunto(s)
Infecciones por Virus de Epstein-Barr/complicaciones , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Inmunoterapia , Trastornos Linfoproliferativos/terapia , Neoplasias/terapia , Trasplante de Órganos/efectos adversos , Linfocitos T Citotóxicos/inmunología , Herpesvirus Humano 4/fisiología , Humanos , Trastornos Linfoproliferativos/virología , Neoplasias/virología , Receptores de Antígenos de Linfocitos T/fisiología , Latencia del Virus
13.
J Pathol Clin Res ; 6(4): 308-319, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32696621

RESUMEN

Earlier studies identified the transmembrane cell surface C-type lectin CLEC14A as a putative tumour endothelial marker. For CLEC14A to progress as a vascular target in solid tumours an in-depth analysis of CLEC14A expression in human healthy and tumour tissue is needed. It is here shown that an analysis of 5332 RNA expression profiles in the public domain confirmed high expression of CLEC14A in tumour compared to healthy human tissue. It is further shown by immunohistochemistry that CLEC14A protein is absent, or expressed at a very low level, in healthy human and primate tissue. In contrast, CLEC14A is expressed on the vasculature of a range of human solid tumours, with particularly high expression in more than half of renal cell carcinomas. Elevated levels of CLEC14A transcripts were identified in some non-cancer pathologies; such comorbidities may need to be excluded from trials of therapies targeting this marker. It is further shown that, as CLEC14A expression can be induced by the absence of shear stress, it is imperative that freshly collected as opposed to aged or post-mortem tissue be analysed. We conclude that CLEC14A is a promising target to enable development of novel anti-cancer therapies for solid tumours.


Asunto(s)
Biomarcadores de Tumor/análisis , Moléculas de Adhesión Celular/análisis , Células Endoteliales/química , Lectinas Tipo C/análisis , Neoplasias/irrigación sanguínea , Neovascularización Patológica , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Biomarcadores de Tumor/genética , Moléculas de Adhesión Celular/genética , Bases de Datos Genéticas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Humanos , Inmunohistoquímica , Lectinas Tipo C/genética , Macaca fascicularis , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Regulación hacia Arriba
14.
JCI Insight ; 5(19)2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-33004686

RESUMEN

Engineering T cells to express chimeric antigen receptors (CARs) specific for antigens on hematological cancers has yielded remarkable clinical responses, but with solid tumors, benefit has been more limited. This may reflect lack of suitable target antigens, immune evasion mechanisms in malignant cells, and/or lack of T cell infiltration into tumors. An alternative approach, to circumvent these problems, is targeting the tumor vasculature rather than the malignant cells directly. CLEC14A is a glycoprotein selectively overexpressed on the vasculature of many solid human cancers and is, therefore, of considerable interest as a target antigen. Here, we generated CARs from 2 CLEC14A-specific antibodies and expressed them in T cells. In vitro studies demonstrated that, when exposed to their target antigen, these engineered T cells proliferate, release IFN-γ, and mediate cytotoxicity. Infusing CAR engineered T cells into healthy mice showed no signs of toxicity, yet these T cells targeted tumor tissue and significantly inhibited tumor growth in 3 mouse models of cancer (Rip-Tag2, mPDAC, and Lewis lung carcinoma). Reduced tumor burden also correlated with significant loss of CLEC14A expression and reduced vascular density within malignant tissues. These data suggest the tumor vasculature can be safely and effectively targeted with CLEC14A-specific CAR T cells, offering a potent and widely applicable therapy for cancer.


Asunto(s)
Carcinoma Pulmonar de Lewis/prevención & control , Carcinoma Ductal Pancreático/prevención & control , Moléculas de Adhesión Celular/metabolismo , Inmunoterapia Adoptiva/métodos , Lectinas Tipo C/metabolismo , Neovascularización Patológica/prevención & control , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Animales , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patología , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Moléculas de Adhesión Celular/genética , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lectinas Tipo C/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/inmunología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/prevención & control
15.
Cancer Immunol Immunother ; 58(1): 85-94, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18488221

RESUMEN

Circulating T lymphocytes enter a tissue if they express appropriate chemokine receptors and adhesion molecules to engage ligands presented at this site. To aid rational development of T cell-based therapies for Hodgkin's lymphoma (HL), we have assessed the expression and function of homing receptors on tumour-infiltrating T cells in HL and compared them with T cells from unaffected lymph nodes and colorectal cancer tissue. Chemokine receptors CXCR3, CXCR4 and CCR7 were expressed on a large proportion of T cells within HL tissue and mediated chemotaxis to purified chemokine. The corresponding ligands (CXCL10, CXCL12, CCL21) were expressed on the malignant cells and/or vascular endothelium. Adhesion molecules including CD62L were widely expressed on HL-derived T cells and their corresponding ligands were detected on vessels within the tumour. This homing phenotype was distinct from T cells isolated from colorectal cancer, but matched closely the phenotype of T cells from unaffected lymph nodes. Thus, T cell recruitment to HL resembles entry of naïve/central memory T cells into normal lymph nodes. This has important implications for current approaches to treat HL using T cells activated and expanded in vitro that lack CCR7 and CD62L expression.


Asunto(s)
Enfermedad de Hodgkin/inmunología , Enfermedad de Hodgkin/metabolismo , Receptores Mensajeros de Linfocitos/metabolismo , Adolescente , Adulto , Anciano , Niño , Femenino , Citometría de Flujo , Enfermedad de Hodgkin/fisiopatología , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Receptores CXCR/inmunología , Receptores CXCR/metabolismo , Receptores Mensajeros de Linfocitos/inmunología , Adulto Joven
16.
PLoS One ; 14(10): e0224015, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31622431

RESUMEN

Engineering T-cells to express receptors specific for antigens present on tumour tissue is proving a highly effective treatment for some leukaemias. However, extending this to solid tumours requires antigens that can be safely and effectively targeted. TEM8, a marker overexpressed on the vasculature of some solid tumours, has been proposed as one such target. A recent report stated that T-cells engineered to express a TEM8-specific chimeric antigen receptor (CAR), when injected into mouse models of triple negative breast cancer, are both safe and effective in controlling tumour growth. Here we report contrasting data with a panel of TEM8-specific CAR-T-cells including one generated from the same antibody used in the other study. We found that the CAR-T-cells demonstrated clear TEM8-specific cytotoxic and cytokine release responses in vitro, but when injected into healthy C57BL6 and NSG mice they rapidly and selectively disappeared from the circulation and in most cases caused rapid toxicity. Infusing CAR-T-cells into a TEM8-knockout mouse indicated that selective loss of cells from the circulation was due to targeting of TEM8 in healthy tissues. Histological analysis of mice treated with a TEM8-specific CAR revealed evidence of inflammation in the lung and spleen with large collections of infiltrating neutrophils. Therefore our data raise concerns over potential on-target off-tumour toxicity with CARs targeting TEM8 and these should be considered carefully before embarking upon clinical trials with such agents.


Asunto(s)
Inmunoterapia Adoptiva , Proteínas de Microfilamentos/metabolismo , Receptores de Superficie Celular/metabolismo , Anticuerpos de Cadena Única/metabolismo , Linfocitos T/metabolismo , Neoplasias de la Mama Triple Negativas/terapia , Animales , Línea Celular Tumoral , Femenino , Humanos , Interferón gamma/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Infiltración Neutrófila , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Anticuerpos de Cadena Única/genética , Bazo/patología , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Clin Cancer Res ; 25(17): 5315-5328, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31182433

RESUMEN

PURPOSE: Intratumoral hypoxia and immunity have been correlated with patient outcome in various tumor settings. However, these factors are not currently considered for treatment selection in head and neck cancer (HNC) due to lack of validated biomarkers. Here we sought to develop a hypoxia-immune classifier with potential application in patient prognostication and prediction of response to targeted therapy. EXPERIMENTAL DESIGN: A 54-gene hypoxia-immune signature was constructed on the basis of literature review. Gene expression was analyzed in silico using the The Cancer Genome Atlas (TCGA) HNC dataset (n = 275) and validated using two independent cohorts (n = 130 and 123). IHC was used to investigate the utility of a simplified protein signature. The spatial distribution of hypoxia and immune markers was examined using multiplex immunofluorescence staining. RESULTS: Unsupervised hierarchical clustering of TCGA dataset (development cohort) identified three patient subgroups with distinct hypoxia-immune phenotypes and survival profiles: hypoxialow/immunehigh, hypoxiahigh/immunelow, and mixed, with 5-year overall survival (OS) rates of 71%, 51%, and 49%, respectively (P = 0.0015). The prognostic relevance of the hypoxia-immune gene signature was replicated in two independent validation cohorts. Only PD-L1 and intratumoral CD3 protein expression were associated with improved OS on multivariate analysis. Hypoxialow/immunehigh and hypoxiahigh/immunelow tumors were overrepresented in "inflamed" and "immune-desert" microenvironmental profiles, respectively. Multiplex staining demonstrated an inverse correlation between CA-IX expression and prevalence of intratumoral CD3+ T cells (r = -0.5464; P = 0.0377), further corroborating the transcription-based classification. CONCLUSIONS: We developed and validated a hypoxia-immune prognostic transcriptional classifier, which may have clinical application to guide the use of hypoxia modification and targeted immunotherapies for the treatment of HNC.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/metabolismo , Hipoxia/inmunología , Hipoxia/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Estudios de Cohortes , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Hipoxia/genética , Hipoxia/patología , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia , Adulto Joven
18.
J Immunother Cancer ; 6(1): 70, 2018 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-30001747

RESUMEN

Mutation-derived neoantigens represent an important class of tumour-specific, tumour rejection antigens, and are attractive targets for TCR gene therapy of cancer. The majority of such mutations are patient-specific and targeting these requires a fully personalized approach. However, some mutations are found recurrently among cancer patients, and represent potential targets for neoantigen-specific TCR gene therapy that is more widely applicable. Therefore, we have investigated if some cancer mutations found recurrently in hematological malignancies encode immunogenic neoantigens presented by common European Caucasoid HLA class I alleles and can form targets for TCR gene therapy. We initially focused on identifying HLA class I neoepitopes derived from calreticulin (CALR) exon 9 mutations, found in ~ 80% of JAK2wt myeloproliferative neoplasms (MPN). Based on MHC class I peptide predictions, a number of peptides derived from mutant CALR (mCALR) were predicted to bind to HLA-A*03:01 and HLA-B*07:02. However, using mass spectrometry and ex vivo pMHC multimer staining of PBMC from MPN patients with CALR exon 9 mutations, we found no evidence that these peptides were naturally processed and presented on the surface of mCALR-expressing target cells. We next developed a protocol utilizing pMHC multimers to isolate CD8+ T cells from healthy human donor PBMC that are specific for mCALR and additional putative neoepitopes found recurrently in hematological malignancies. Using this approach, CD8+ T cells specific for HLA-A*03:01- and HLA-B*07:02-presented mCALR peptides and an HLA-A*11:01-presented mutant FBXW7 (mFBXW7) peptide were successfully isolated. TCRs isolated from mCALR-specific CD8+ T cell populations were not able to recognize target cells engineered to express mCALR. In contrast, mFBXW7-specific CD8+ T cells were able to recognize target cells engineered to express mFBXW7. In conclusion, while we found no evidence for mCALR derived neoepitope presentation in the context of the HLA class I alleles studied, our data suggests that the recurrent pR465H mutation in FBXW7 may encode an HLA-A*11:01 presented neoepitope, and warrants further investigation as a target for T cell based immunotherapy of cancer.


Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Epítopos de Linfocito T/inmunología , Antígenos HLA/genética , Antígenos HLA/inmunología , Neoplasias Hematológicas/patología , Humanos , Activación de Linfocitos/inmunología , Mutación , Péptidos/genética , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/genética , Recurrencia , Especificidad del Receptor de Antígeno de Linfocitos T
19.
Cancer Immunol Res ; 3(10): 1138-47, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25711537

RESUMEN

Infusing virus-specific T cells is effective treatment for rare Epstein-Barr virus (EBV)-associated posttransplant lymphomas, and more limited success has been reported using this approach to treat a far more common EBV-associated malignancy, nasopharyngeal carcinoma (NPC). However, current approaches using EBV-transformed lymphoblastoid cell lines to reactivate EBV-specific T cells for infusion take 2 to 3 months of in vitro culture and favor outgrowth of T cells targeting viral antigens expressed within EBV(+) lymphomas, but not in NPC. Here, we explore T-cell receptor (TCR) gene transfer to rapidly and reliably generate T cells specific for the NPC-associated viral protein LMP2. We cloned a human leukocyte antigen (HLA) A*1101-restricted TCR, which would be widely applicable because 40% of NPC patients carry this HLA allele. Studying both the wild-type and modified forms, we have optimized expression of the TCR and demonstrated high-avidity antigen-specific function (proliferation, cytotoxicity, and cytokine release) in both CD8(+) and CD4(+) T cells. The engineered T cells also inhibited LMP2(+) epithelial tumor growth in a mouse model. Furthermore, transduced T cells from patients with advanced NPC lysed LMP2-expressing NPC cell lines. Using this approach, within a few days large numbers of high-avidity LMP2-specific T cells can be generated reliably to treat NPC, thus providing an ideal clinical setting to test TCR gene transfer without the risk of autoimmunity through targeting self-antigens.


Asunto(s)
Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/inmunología , Antígenos HLA-A/inmunología , Herpesvirus Humano 4/inmunología , Neoplasias Nasofaríngeas/etiología , Receptores de Antígenos de Linfocitos T/genética , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Carcinoma , Línea Celular Tumoral , Citocinas/biosíntesis , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología , Femenino , Expresión Génica , Técnicas de Transferencia de Gen , Humanos , Inmunoterapia , Interferón gamma/biosíntesis , Ratones , Datos de Secuencia Molecular , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/terapia , Transducción Genética , Carga Tumoral , Proteínas de la Matriz Viral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Nat Med ; 20(3): 255-64, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24531378

RESUMEN

The elderly often suffer from progressive muscle weakness and regenerative failure. We demonstrate that muscle regeneration is impaired with aging owing in part to a cell-autonomous functional decline in skeletal muscle stem cells (MuSCs). Two-thirds of MuSCs from aged mice are intrinsically defective relative to MuSCs from young mice, with reduced capacity to repair myofibers and repopulate the stem cell reservoir in vivo following transplantation. This deficiency is correlated with a higher incidence of cells that express senescence markers and is due to elevated activity of the p38α and p38ß mitogen-activated kinase pathway. We show that these limitations cannot be overcome by transplantation into the microenvironment of young recipient muscles. In contrast, subjecting the MuSC population from aged mice to transient inhibition of p38α and p38ß in conjunction with culture on soft hydrogel substrates rapidly expands the residual functional MuSC population from aged mice, rejuvenating its potential for regeneration and serial transplantation as well as strengthening of damaged muscles of aged mice. These findings reveal a synergy between biophysical and biochemical cues that provides a paradigm for a localized autologous muscle stem cell therapy for the elderly.


Asunto(s)
Envejecimiento , Fuerza Muscular , Músculos/citología , Regeneración , Rejuvenecimiento , Células Madre/citología , Animales , Proliferación Celular , Trasplante de Células , Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Hidrogeles/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Proteína Quinasa 11 Activada por Mitógenos/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Fenotipo , Fosfoproteínas/metabolismo , Trasplante de Células Madre , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA