Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Immunother Cancer ; 12(2)2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38413223

RESUMO

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.


Assuntos
Neoplasias , Receptores de Antígenos Quiméricos , Humanos , Camundongos , Animais , Pericitos/metabolismo , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Camundongos Endogâmicos C57BL , Neoplasias/metabolismo , Linfócitos T/metabolismo , Camundongos Knockout , Microambiente Tumoral , Antígenos de Neoplasias/metabolismo , Antígenos CD/metabolismo
2.
JCI Insight ; 5(19)2020 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-33004686

RESUMO

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.


Assuntos
Carcinoma Pulmonar de Lewis/prevenção & controle , Carcinoma Ductal Pancreático/prevenção & controle , Moléculas de Adesão Celular/metabolismo , Imunoterapia Adotiva/métodos , Lectinas Tipo C/metabolismo , Neovascularização Patológica/prevenção & controle , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Animais , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Moléculas de Adesão Celular/genética , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Lectinas Tipo C/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/imunologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/prevenção & controle
3.
J Pathol Clin Res ; 6(4): 308-319, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32696621

RESUMO

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.


Assuntos
Biomarcadores Tumorais/análise , Moléculas de Adesão Celular/análise , Células Endoteliais/química , Lectinas Tipo C/análise , Neoplasias/irrigação sanguínea , Neovascularização Patológica , Inibidores da Angiogênese/uso terapêutico , Animais , Biomarcadores Tumorais/genética , Moléculas de Adesão Celular/genética , Bases de Dados Genéticas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Humanos , Imuno-Histoquímica , Lectinas Tipo C/genética , Macaca fascicularis , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Regulação para Cima
4.
Blood ; 136(10): 1155-1160, 2020 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-32573723

RESUMO

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.


Assuntos
Arginina/metabolismo , Argininossuccinato Sintase/metabolismo , Imunoterapia Adotiva/métodos , Leucemia Mieloide Aguda/terapia , Neuroblastoma/terapia , Ornitina Carbamoiltransferase/metabolismo , Linfócitos T/transplante , Animais , Apoptose , Argininossuccinato Sintase/genética , Proliferação de Células , Humanos , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Engenharia Metabólica/métodos , Camundongos , Camundongos Nus , Neuroblastoma/imunologia , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Ornitina Carbamoiltransferase/genética , Receptores de Antígenos Quiméricos/química , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
PLoS One ; 14(10): e0224015, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31622431

RESUMO

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.


Assuntos
Imunoterapia Adotiva , Proteínas dos Microfilamentos/metabolismo , Receptores de Superfície Celular/metabolismo , Anticorpos de Cadeia Única/metabolismo , Linfócitos T/metabolismo , Neoplasias de Mama Triplo Negativas/terapia , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Interferon gama/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Infiltração de Neutrófilos , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Anticorpos de Cadeia Única/genética , Baço/patologia , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Clin Cancer Res ; 25(17): 5315-5328, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31182433

RESUMO

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.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias de Cabeça e Pescoço/imunologia , Neoplasias de Cabeça e Pescoço/metabolismo , Hipóxia/imunologia , Hipóxia/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígeno B7-H1/imunologia , Antígeno B7-H1/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/imunologia , Estudos de Coortes , Feminino , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Hipóxia/genética , Hipóxia/patologia , Linfócitos do Interstício Tumoral/imunologia , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Retrospectivos , Taxa de Sobrevida , Adulto Jovem
7.
Haematologica ; 104(9): 1892-1905, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30573509

RESUMO

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.


Assuntos
Cálcio/metabolismo , Traumatismo por Reperfusão Miocárdica/genética , Proteína ORAI1/genética , Tetraspaninas/genética , Trombose Venosa/genética , Fator de von Willebrand/genética , Animais , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Galinhas , Modelos Animais de Doenças , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Histamina/farmacologia , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Transporte de Íons/efeitos dos fármacos , Células Jurkat , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Proteína ORAI1/metabolismo , Transdução de Sinais , Tetraspaninas/metabolismo , Trombina/farmacologia , Trombose Venosa/metabolismo , Trombose Venosa/patologia , Fator de von Willebrand/metabolismo
8.
PLoS Biol ; 16(9): e2005046, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30180168

RESUMO

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.


Assuntos
Diferenciação Celular/imunologia , Microambiente Celular , Linfonodos/imunologia , Ativação Linfocitária/imunologia , Linfócitos T/citologia , Adulto , Proliferação de Células , Criança , Fibroblastos/citologia , Humanos , Memória Imunológica , Fenótipo
9.
J Immunother Cancer ; 6(1): 70, 2018 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-30001747

RESUMO

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.


Assuntos
Antígenos de Neoplasias/imunologia , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Epitopos de Linfócito T/imunologia , Antígenos HLA/genética , Antígenos HLA/imunologia , Neoplasias Hematológicas/patologia , Humanos , Ativação Linfocitária/imunologia , Mutação , Peptídeos/genética , Peptídeos/imunologia , Receptores de Antígenos de Linfócitos T/genética , Recidiva , Especificidade do Receptor de Antígeno de Linfócitos T
10.
Cancer Immunol Res ; 3(10): 1138-47, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25711537

RESUMO

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.


Assuntos
Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/imunologia , Antígenos HLA-A/imunologia , Herpesvirus Humano 4/imunologia , Neoplasias Nasofaríngeas/etiologia , Receptores de Antígenos de Linfócitos T/genética , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Carcinoma , Linhagem Celular Tumoral , Citocinas/biossíntese , Citotoxicidade Imunológica , Modelos Animais de Doenças , Epitopos de Linfócito T/imunologia , Feminino , Expressão Gênica , Técnicas de Transferência de Genes , Humanos , Imunoterapia , Interferon gama/biossíntese , Camundongos , Dados de Sequência Molecular , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/terapia , Transdução Genética , Carga Tumoral , Proteínas da Matriz Viral/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Nat Med ; 20(3): 255-64, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24531378

RESUMO

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.


Assuntos
Envelhecimento , Força Muscular , Músculos/citologia , Regeneração , Rejuvenescimento , Células-Tronco/citologia , Animais , Proliferação de Células , Transplante de Células , Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Feminino , Proteínas de Fluorescência Verde/metabolismo , Hidrogéis/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Proteína Quinase 11 Ativada por Mitógeno/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Fenótipo , Fosfoproteínas/metabolismo , Transplante de Células-Tronco , Fatores de Tempo
12.
Am J Pathol ; 180(3): 1215-1222, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22226739

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Neoplasias Nasofaríngeas/imunologia , Receptores CCR5/metabolismo , Receptores de Quimiocinas/metabolismo , Receptores Virais/metabolismo , Subpopulações de Linfócitos T/imunologia , Carcinoma , Linhagem Celular Tumoral , Humanos , Memória Imunológica/fisiologia , Linfócitos do Interstício Tumoral/imunologia , Carcinoma Nasofaríngeo , Receptores CCR5/fisiologia , Receptores CXCR6 , Receptores de Quimiocinas/fisiologia , Receptores Virais/fisiologia
13.
Eur Urol ; 61(2): 385-94, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22079021

RESUMO

BACKGROUND: Evidence suggests that some patients with renal cell carcinoma (RCC) respond to immunomodulatory therapies that activate T lymphocytes. A prerequisite for effective T cell therapy is efficient targeting of effector T cells to the tumour site, yet the molecular basis of T cell recruitment to RCC is unknown. Furthermore, some T cells that naturally infiltrate this cancer are regulatory T cells (Tregs) that may suppress antitumour immune responses. OBJECTIVE: Determine the mechanisms of effector and regulatory T cell recruitment to RCC to allow targeted therapy that promotes local anti-tumour immunity. DESIGN, SETTING, AND PARTICIPANTS: Tumour-infiltrating and peripheral blood T cells were collected from 70 patients undergoing nephrectomy for RCC. MEASUREMENTS: T cells were analysed by multicolour flow cytometry for expression of 19 chemokine receptors and 7 adhesion molecules. Receptors that were expressed at higher levels on tumour-infiltrating lymphocytes (TILs) compared with matched peripheral blood lymphocytes (PBLs) were analysed further for their ability to mediate migration responses in TILs and for expression of corresponding ligands in tumour tissue. RESULTS AND LIMITATIONS: Three chemokine receptors-CCR5, CXCR3, and CXCR6-were significantly overexpressed on TILs compared with matched PBLs (n=16 cases) and were capable of promoting migration in vitro. Their corresponding ligands CCL4-5, CXCL9-11, and CXCL16 were all detected in RCC tissue. However, since they were present in all cases studied, it was not possible to correlate ligand expression with levels of T cell infiltration. Foxp3(+) Tregs were enriched within TILs compared with matched PBLs and expressed high levels of CCR5, CXCR3, and CXCR6, as well as CCR6, the ligand for which (CCL20) was detectable in RCC tissue. CONCLUSIONS: Our data support a role for CCR5, CXCR3, and CXCR6 in the selective recruitment of T cells into RCC tissue and, together with CCR6, in the recruitment of Tregs.


Assuntos
Carcinoma de Células Renais/imunologia , Neoplasias Renais/imunologia , Ativação Linfocitária/imunologia , Linfócitos do Interstício Tumoral/imunologia , Receptores CCR5/imunologia , Receptores CCR6/imunologia , Receptores CXCR3/imunologia , Receptores de Quimiocinas/imunologia , Receptores Virais/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Movimento Celular/imunologia , Feminino , Citometria de Fluxo/métodos , Fatores de Transcrição Forkhead/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Receptores CXCR6 , Linfócitos T/imunologia
14.
Cancer Immunol Immunother ; 61(5): 615-28, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22021067

RESUMO

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.


Assuntos
Ligante 4-1BB/imunologia , Interleucina-12/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Neoplasias/imunologia , Neoplasias/patologia , Ligante 4-1BB/biossíntese , Ligante 4-1BB/genética , Antígeno CD56/imunologia , Processos de Crescimento Celular/genética , Processos de Crescimento Celular/imunologia , Linhagem Celular Tumoral , Feminino , Proteínas Ligadas por GPI/imunologia , Humanos , Interferon gama/imunologia , Interleucina-12/biossíntese , Interleucina-12/genética , Leucócitos Mononucleares/imunologia , Ativação Linfocitária , Neoplasias/genética , Fenótipo , Receptores de IgG/imunologia , Regulação para Cima
15.
Blood ; 116(19): 3695-704, 2010 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-20671118

RESUMO

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.


Assuntos
Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/virologia , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/imunologia , Leucemia Linfocítica Granular Grande/imunologia , Leucemia Linfocítica Granular Grande/virologia , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/imunologia , Sequência de Bases , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Primers do DNA/genética , Infecções por Vírus Epstein-Barr/terapia , Expressão Gênica , Genes Virais , Herpesvirus Humano 4/patogenicidade , Humanos , Imunoterapia Adotiva , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/virologia , Leucemia Linfocítica Granular Grande/terapia , Linfoma Extranodal de Células T-NK/imunologia , Linfoma Extranodal de Células T-NK/terapia , Linfoma Extranodal de Células T-NK/virologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Linfócitos T/imunologia , Linfócitos T/virologia
16.
Drug News Perspect ; 23(4): 221-8, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20520851

RESUMO

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.


Assuntos
Infecções por Vírus Epstein-Barr/complicações , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Imunoterapia , Transtornos Linfoproliferativos/terapia , Neoplasias/terapia , Transplante de Órgãos/efeitos adversos , Linfócitos T Citotóxicos/imunologia , Herpesvirus Humano 4/fisiologia , Humanos , Transtornos Linfoproliferativos/virologia , Neoplasias/virologia , Receptores de Antígenos de Linfócitos T/fisiologia , Latência Viral
17.
J Immunol ; 182(4): 1919-28, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19201845

RESUMO

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.


Assuntos
Linfócitos B/virologia , Linfócitos T CD8-Positivos/imunologia , Ativação Linfocitária/imunologia , Proteínas da Matriz Viral/imunologia , Apresentação de Antígeno/imunologia , Linfócitos B/imunologia , Western Blotting , Linfócitos T CD8-Positivos/virologia , Linhagem Celular Transformada , Transformação Celular Viral/imunologia , Citocinas/biossíntese , Citotoxicidade Imunológica/imunologia , Ensaio de Imunoadsorção Enzimática , Epitopos de Linfócito T/imunologia , Herpesvirus Humano 4/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Proteínas da Matriz Viral/biossíntese
18.
Cancer Immunol Immunother ; 58(1): 85-94, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18488221

RESUMO

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.


Assuntos
Doença de Hodgkin/imunologia , Doença de Hodgkin/metabolismo , Receptores de Retorno de Linfócitos/metabolismo , Adolescente , Adulto , Idoso , Criança , Feminino , Citometria de Fluxo , Doença de Hodgkin/fisiopatologia , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Receptores CXCR/imunologia , Receptores CXCR/metabolismo , Receptores de Retorno de Linfócitos/imunologia , Adulto Jovem
19.
J Gen Virol ; 88(Pt 9): 2417-2425, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17698650

RESUMO

Adenovirus is a significant pathogen in immunocompromised patients and is widely utilized as a gene delivery vector, so a detailed understanding of the human immune response to adenovirus infection is critical. This study characterized the adenovirus-specific CD4(+) T-cell response of healthy donors by incubation with whole virus or with individual hexon and fiber proteins. Adenovirus-specific CD4(+) T cells averaged 0.26 % of the CD4(+) T-cell pool and were detectable in all donors. T cells recognizing the highly conserved hexon protein accounted for 0.09 %, whereas no response was observed against the fiber protein. A panel of hexon-specific CD4(+) T-cell clones was generated and shown to lyse targets infected with adenovirus from different serotypes and species. Three CD4 T-cell epitopes are described, which map to highly conserved regions of the hexon protein.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Proteínas do Capsídeo/imunologia , Linfócitos T/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteínas do Capsídeo/química , Linhagem Celular , Sequência Conservada , Antígenos HLA/imunologia , Humanos , Depleção Linfocítica
20.
Int Immunol ; 18(4): 591-601, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16507598

RESUMO

EBV is associated with a broad range of malignancies. Adoptive immunotherapy of these tumors with EBV-specific CTL proved useful. We generated a panel of primary human T cells specific to various EBV antigens (i.e. Epstein-Barr nuclear antigen 3A, 3B and BamHI-M leftward reading frame) via transfer of modified TCR genes that are either coupled to CD3zeta or Fc(epsilon)RIgamma. TCR-transduced T cells from 20-60% of donors (total number of 25) demonstrated specific lysis of EBV peptide-loaded target cells, whereas lymphoblastoid cell lines expressing native EBV antigens were not killed by any of the EBV-specific T cell populations. This non-responsiveness, confirmed at the level of nuclear factor of activated T cells activation, is not due to receptor configuration since identical receptor formats specific for melanoma antigens successfully re-targeted T cells to native melanoma cells. In an effort to generate a more potent receptor, we introduced a CD28 domain into one of the EBV-specific TCR. This TCR did not affect the cytotoxic response of re-targeted T cells, but dramatically enhanced antigen-specific IFNgamma production. We therefore conclude that these novel CD28-containing EBV-specific TCRs provide a basis for further development of TCR gene transfer to treat EBV-induced diseases.


Assuntos
Antígenos Nucleares do Vírus Epstein-Barr/imunologia , Técnicas de Transferência de Genes , Genes Codificadores dos Receptores de Linfócitos T , Interferon gama/biossíntese , Linfócitos T/imunologia , Antígenos CD28/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Terapia Genética/métodos , Humanos , Células Jurkat , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA