Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Cell ; 170(1): 142-157.e19, 2017 Jun 29.
Article in English | MEDLINE | ID: mdl-28648661

ABSTRACT

Immune-checkpoint-blockade (ICB)-mediated rejuvenation of exhausted T cells has emerged as a promising approach for treating various cancers and chronic infections. However, T cells that become fully exhausted during prolonged antigen exposure remain refractory to ICB-mediated rejuvenation. We report that blocking de novo DNA methylation in activated CD8 T cells allows them to retain their effector functions despite chronic stimulation during a persistent viral infection. Whole-genome bisulfite sequencing of antigen-specific murine CD8 T cells at the effector and exhaustion stages of an immune response identified progressively acquired heritable de novo methylation programs that restrict T cell expansion and clonal diversity during PD-1 blockade treatment. Moreover, these exhaustion-associated DNA-methylation programs were acquired in tumor-infiltrating PD-1hi CD8 T cells, and approaches to reverse these programs improved T cell responses and tumor control during ICB. These data establish de novo DNA-methylation programming as a regulator of T cell exhaustion and barrier of ICB-mediated T cell rejuvenation.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Epigenesis, Genetic , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Adenocarcinoma/drug therapy , Animals , CD8-Positive T-Lymphocytes/immunology , DNA Methylation , Female , Male , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Prostatic Neoplasms/drug therapy , Virus Diseases/drug therapy
2.
J Immunol ; 186(6): 3327-35, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21317394

ABSTRACT

Previous work from our laboratory showed that hydrocortisone (HC) combined with IL-15 induces expansion of activated human NK cells. We set up an experimental tumor model to evaluate the use of adoptively transferred, HC plus IL-15 (HC/IL-15)-activated and -expanded murine NK cells in the treatment of syngeneic mice carrying established lung metastases of the CT26 transplantable tumor. We also examined the effect of denileukin diftitox (Ontak) on the depletion of regulatory T cells to enhance the in vivo antitumor immunity induced by the adoptively transferred NK cells. Our results clearly demonstrate that murine DX5(+) NK cells are largely expanded in the presence of IL-15 plus HC while retaining intact their functional status. Moreover, when intravenously infused, they mediated significant antitumor responses against CT26 lung tumors in syngeneic BALB/c animals that were further enhanced upon pretreatment of the tumor-bearing animals with Ontak. Total splenocytes and isolated splenic T cells from NK-treated mice responded in vitro against CT26 tumor cells as evidenced by IFN-γ-based ELISPOT, proliferation, and cytotoxicity assays. Importantly, animals treated with Ontak plus adoptive transfer of HC/IL-15-expanded NK cells significantly retarded CT26 tumor growth after a rechallenge with the same tumor s.c. in their flanks. Taken altogether, our data suggest that NK cell adoptive transfer can trigger adaptive antitumor T cell responses, and regulatory T cell depletion by Ontak is mandatory for enabling HC/IL-15-activated NK cells to promote long-lasting adaptive antitumor immunity.


Subject(s)
Adoptive Transfer/methods , Antineoplastic Agents/therapeutic use , Diphtheria Toxin/therapeutic use , Interleukin-2/therapeutic use , Killer Cells, Natural/transplantation , Lung Neoplasms/immunology , Lymphocyte Depletion/methods , T-Lymphocytes, Regulatory/immunology , Adaptive Immunity , Animals , Antineoplastic Agents/administration & dosage , Cell Proliferation/drug effects , Cells, Cultured , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/secondary , Diphtheria Toxin/administration & dosage , Hydrocortisone/administration & dosage , Hydrocortisone/therapeutic use , Interleukin-15/administration & dosage , Interleukin-2/administration & dosage , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Random Allocation , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/pathology
3.
Sci Immunol ; 7(68): eabf6136, 2022 02 04.
Article in English | MEDLINE | ID: mdl-35119937

ABSTRACT

The immune system undergoes a progressive functional remodeling with age. Understanding how age bias shapes antitumor immunity is essential in designing effective immunotherapies, especially for pediatric patients. Here, we explore antitumor CD8+ T cell responses generated in young (prepubescent) and adult (presenescent) mice. Using an MHCI-deficient tumor model, we observed that tumor-reactive CD8+ T cells expanded in young tumor-bearing (TB) mice acquired a terminally differentiated phenotype characterized by overexpression of inhibitory receptors and the transcription factor Tox1. Furthermore, tumor-infiltrating CD8+ T cells from young tumors yielded a poor cytokine response compared with CD8+ T cells infiltrating adult tumors. Young migratory dendritic cells (migDCs) from the draining lymph nodes (dLNs), and mononuclear phagocytic cells (MPCs) infiltrating young tumors, were more competent in capturing and cross-presenting tumor antigen, leading to enhanced priming of CD8+ T cells in dLNs and their subsequent terminal differentiation in the tumors. Single-cell transcriptional profiling of tumor-infiltrating MPCs demonstrated that young MPCs are polarized toward an inflammatory, effector phenotype. Consistent with our observations in young versus adult TB mice, analysis of immune infiltrates from pediatric solid tumors showed a correlation between tumor-infiltrating CD8+ T cells with an exhaustion phenotype and the frequency of PD-L1-expressing monocytes/macrophages. Collectively, these data indicate that a young tissue microenvironment contributes to the generation of an immune response skewed toward a less pliable terminal effector state, thus narrowing the window for immunotherapeutic interventions.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Animals , Cell Differentiation/immunology , Cell Line, Tumor , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
4.
Cancer Immunol Immunother ; 60(12): 1683-95, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21706285

ABSTRACT

We have previously reported a synergistic effect between hydrocortisone (HC) and IL-15 on promoting natural killer (NK) cell expansion and function. In the present study, we extend our findings to methylprednisolone (MeP) and dexamethasone (Dex), thus ascribing to glucocorticoids (GCs) a general feature as positive regulators of IL-15-mediated effects on NK cells. We demonstrate that each GC when combined with IL-15 in cultures of peripheral blood (PB)-derived CD56(+) cells induces increased expansion of CD56(+)CD3(-) cells displaying high cytolytic activity, IFN-γ production potential and activating receptor expression, including NKp30, NKp44, NKp46, 2B4, NKG2D and DNAM-1. Furthermore, GCs protected NK cells from IL-15-induced cell death. The combination of IL-15 with GCs favored the expansion of a relatively more immature CD16(low/neg) NK cell population, with high expression of NKG2A and CD94, and significantly lower expression of KIR (CD158a and CD158b) and CD57, compared to IL-15 alone. IL-15-expanded NK cells, in the presence or absence of GCs, did not express CD62L, CXCR1 or CCR7. However, the presence of GCs significantly increased the density of CXCR3 and induced strong CXCR4 expression on the surface of NK cells. Our data indicate that IL-15/GC-expanded NK cells, apart from their increased proliferation rate, retain their functional integrity and exhibit a migratory potential rendering them useful for adoptive transfer in NK cell-based cancer immunotherapy.


Subject(s)
Cell Proliferation/drug effects , Glucocorticoids/pharmacology , Interleukin-15/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Adult , Cell Separation , Cells, Cultured , Dexamethasone/pharmacology , Flow Cytometry , Humans , Immunophenotyping , Killer Cells, Natural/cytology , Methylprednisolone/pharmacology , Middle Aged , Phenotype
5.
Clin Cancer Res ; 25(24): 7554-7564, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31455682

ABSTRACT

PURPOSE: Immunotherapy with IL2, GM-CSF, and an anti-disialoganglioside (GD2) antibody significantly increases event-free survival in children with high-risk neuroblastoma. However, therapy failure in one third of these patients and IL2-related toxicities pose a major challenge. We compared the immunoadjuvant effects of IL15 with those of IL2 for enhancing antibody-dependent cell-mediated cytotoxicity (ADCC) in neuroblastoma. EXPERIMENTAL DESIGN: We tested ADCC against neuroblastoma patient-derived xenografts (PDX) in vitro and in vivo and examined the functional and migratory properties of NK cells activated with IL2 and IL15. RESULTS: In cell culture, IL15-activated NK cells induced higher ADCC against two GD+ neuroblastoma PDXs than did IL2-activated NK cells (P < 0.001). This effect was dose-dependent (P < 0.001) and was maintained across several effector-to-tumor ratios. As compared with IL2, IL15 also improved chemotaxis of NK cells, leading to higher numbers of tumorsphere-infiltrating NK cells in vitro (P = 0.002). In an orthotopic PDX model, animals receiving chemoimmunotherapy with an anti-GD2 antibody, GM-CSF, and a soluble IL15/IL15Rα complex had greater tumor regression than did those receiving chemotherapy alone (P = 0.012) or combined with anti-GD2 antibody and GM-CSF with (P = 0.016) or without IL2 (P = 0.035). This was most likely due to lower numbers of immature tumor-infiltrating NK cells (DX5+CD27+) after IL15/IL15Rα administration (P = 0.029) and transcriptional upregulation of Gzmd. CONCLUSIONS: The substitution of IL15 for IL2 leads to significant tumor regression in vitro and in vivo and supports clinical testing of IL15 for immunotherapy in pediatric neuroblastoma.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Immunotherapy/methods , Interleukin-15/immunology , Killer Cells, Natural/immunology , Neuroblastoma/pathology , Animals , Antibodies, Monoclonal/administration & dosage , Child , Female , Gangliosides/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Interleukin-2/immunology , Neuroblastoma/immunology , Neuroblastoma/therapy , Xenograft Model Antitumor Assays
6.
Sci Transl Med ; 11(498)2019 06 26.
Article in English | MEDLINE | ID: mdl-31243155

ABSTRACT

Cancer arises from the accumulation of genetic alterations, which can lead to the production of mutant proteins not expressed by normal cells. These mutant proteins can be processed and presented on the cell surface by major histocompatibility complex molecules as neoepitopes, allowing CD8+ T cells to mount responses against them. For solid tumors, only an average 2% of neoepitopes predicted by algorithms have detectable endogenous antitumor T cell responses. This suggests that low mutation burden tumors, which include many pediatric tumors, are poorly immunogenic. Here, we report that pediatric patients with acute lymphoblastic leukemia (ALL) have tumor-associated neoepitope-specific CD8+ T cells, responding to 86% of tested neoantigens and recognizing 68% of the tested neoepitopes. These responses include a public neoantigen from the ETV6-RUNX1 fusion that is targeted in seven of nine tested patients. We characterized phenotypic and transcriptional profiles of CD8+ tumor-infiltrating lymphocytes (TILs) at the single-cell level and found a heterogeneous population that included highly functional effectors. Moreover, we observed immunodominance hierarchies among the CD8+ TILs restricted to one or two putative neoepitopes. Our results indicate that robust antitumor immune responses are induced in pediatric ALL despite their low mutation burdens and emphasize the importance of immunodominance in shaping cellular immune responses. Furthermore, these data suggest that pediatric cancers may be amenable to immunotherapies aimed at enhancing immune recognition of tumor-specific neoantigens.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Antigen Presentation/immunology , Child , Genetic Heterogeneity , Humans , Immunodominant Epitopes/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Reproducibility of Results , Transcription, Genetic
7.
J Exp Med ; 214(6): 1593-1606, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28490440

ABSTRACT

Antigen-independent homeostasis of memory CD8 T cells is vital for sustaining long-lived T cell-mediated immunity. In this study, we report that maintenance of human memory CD8 T cell effector potential during in vitro and in vivo homeostatic proliferation is coupled to preservation of acquired DNA methylation programs. Whole-genome bisulfite sequencing of primary human naive, short-lived effector memory (TEM), and longer-lived central memory (TCM) and stem cell memory (TSCM) CD8 T cells identified effector molecules with demethylated promoters and poised for expression. Effector-loci demethylation was heritably preserved during IL-7- and IL-15-mediated in vitro cell proliferation. Conversely, cytokine-driven proliferation of TCM and TSCM memory cells resulted in phenotypic conversion into TEM cells and was coupled to increased methylation of the CCR7 and Tcf7 loci. Furthermore, haploidentical donor memory CD8 T cells undergoing in vivo proliferation in lymphodepleted recipients also maintained their effector-associated demethylated status but acquired TEM-associated programs. These data demonstrate that effector-associated epigenetic programs are preserved during cytokine-driven subset interconversion of human memory CD8 T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epigenesis, Genetic , Homeostasis/genetics , Homeostasis/immunology , Immunologic Memory/genetics , Adoptive Transfer , CD8-Positive T-Lymphocytes/drug effects , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Proliferation/drug effects , Cellular Reprogramming/drug effects , Cellular Reprogramming/genetics , Cytokines/pharmacology , DNA Methylation/drug effects , DNA Methylation/genetics , Epigenesis, Genetic/drug effects , Genetic Loci , Genome, Human , Hematopoietic Stem Cell Transplantation , Hepatocyte Nuclear Factor 1-alpha/metabolism , Homeostasis/drug effects , Humans , Immunocompromised Host , Immunologic Memory/drug effects , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology , Phenotype , Receptors, CCR7/metabolism , Tissue Donors
8.
Stem Cells Dev ; 20(9): 1549-61, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21204633

ABSTRACT

Mesenchymal stem cells (MSCs) are a very important adult stem cell population with a multitude of potential applications in regenerative medicine. The thorough characterization of the bone marrow MSC (BM-MSC) population derived from the BALB/c species was essential, considering the significance of the murine model amongst animal models. In the present study, we examined the effect of gender, age, and in vitro culture on the basic properties (proliferation, differentiation, and immunosuppressive potential) of BM-MSCs. We found a decline in the progenitor frequencies from the BM of adult mice, lower MSC frequencies in all female donors, and an increase in the BM-MSC proliferation rate upon in vitro propagation. We also examined BM-MSCs for the expression of the 3 major embryonic stem cell transcription factors, Oct3/4, Sox-2, and Nanog, as well as 2 mRNA binding proteins, coding region determinant binding protein/insulin-like growth factor 2 mRNA binding protein 1 (Crd-bp/Imp1) and Deleted in azoospermia-like (Dazl), which are expressed in primitive stem cells, umbilical cord blood-hematopoietic stem cells and amniotic fluid stem cells, respectively. Further, it has been reported that these 2 genes are critical for embryonic development. In this study, therefore, we report, for the first time, the expression of Crd-bp/Imp1 and Dazl in BM-MSCs. Dazl, Oct3/4, and Sox2 were detected in relatively low levels in contrast to Crd-bp/Imp1, its major target c-Myc, as well as Nanog, which were expressed redundantly, irrespective of sex, donor age, or in vitro passaging. These findings could further support the extrinsic theory of aging of the MSC population and the potential implication of embryonic genes in adult stem cell physiology.


Subject(s)
Bone Marrow Cells/cytology , Cellular Senescence , Mesenchymal Stem Cells/physiology , Age Factors , Animals , Antigens, Differentiation/metabolism , Cell Differentiation , Cell Proliferation , Cell Size , Cells, Cultured , Female , Gene Expression Profiling , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phenotype , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
9.
J Natl Cancer Inst ; 100(20): 1464-76, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18840818

ABSTRACT

BACKGROUND: Tumor cells in malignant pleural effusions (MPEs) are an important source of monocyte chemoattractant protein (MCP)-1. However, the role of tumor-derived MCP-1 in the pathogenesis and progression of MPE has not been determined. METHODS: B16 mouse skin melanoma cells, which are deficient in MCP-1 expression, and mouse Lewis lung cancer (LLC) cells, which express high levels of MCP-1, were engineered to stably express MCP-1 and short hairpin RNAs (shRNAs) targeting the MCP-1 transcript, respectively. Cells were injected into the pleural cavities of syngeneic immunocompetent mice, and MPE volume and pleural tumors were quantified at necropsy (day 14). MCP-1 and other mediators were determined by cytometric bead array and enzyme-linked immunosorbent assay, and mononuclear and endothelial cells were identified by immunolabeling of F4/80 and factor VIII-related antigen respectively. Mouse survival was assessed using Kaplan-Meier analysis. Vascular permeability in mice with MPE was assessed using albumin-binding Evans blue. Statistical tests were two-sided. RESULTS: LLC cells expressing shRNA against MCP-1 elaborated less than 5% of the MCP-1 level in cells expressing nonspecific shRNA (control cells), and intrapleural delivery of these cells resulted in less MPE (mean MPE volume = 86 and 585 muL, respectively; difference = 499 muL; 95% confidence interval [CI] = 331 to 669 muL; P < .001), reduced MCP-1 levels in the pleural fluid, and lower mortality than when control cells were delivered. Overexpression of MCP-1 in intrapleurally injected B16 melanoma cells led to increased MPE and reduced survival. In mice with MPE, MCP-1 was a potent inducer of vascular permeability, mononuclear recruitment, and, in pleural tumors, of angiogenesis. CONCLUSION: MCP-1 produced by tumor cells is an important determinant of their capacity to induce the formation of MPE and may be a useful target for the treatment of malignant pleural disease.


Subject(s)
Chemokine CCL2/biosynthesis , Neoplasms, Experimental/immunology , Pleural Effusion, Malignant/immunology , Animals , Capillary Permeability , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Disease Models, Animal , Female , Macrophages/immunology , Macrophages/pathology , Male , Melanoma, Experimental/blood supply , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Plasmids/genetics , Pleural Effusion, Malignant/genetics , Pleural Effusion, Malignant/pathology , RNA, Small Interfering/genetics , Skin Neoplasms/blood supply , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL