Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 173
1.
J Clin Invest ; 134(9)2024 Mar 26.
Article En | MEDLINE | ID: mdl-38530366

Aberrant expression of the E26 transformation-specific (ETS) transcription factors characterizes numerous human malignancies. Many of these proteins, including EWS:FLI1 and EWS:ERG fusions in Ewing sarcoma (EwS) and TMPRSS2:ERG in prostate cancer (PCa), drive oncogenic programs via binding to GGAA repeats. We report here that both EWS:FLI1 and ERG bind and transcriptionally activate GGAA-rich pericentromeric heterochromatin. The respective pathogen-like HSAT2 and HSAT3 RNAs, together with LINE, SINE, ERV, and other repeat transcripts, are expressed in EwS and PCa tumors, secreted in extracellular vesicles (EVs), and are highly elevated in plasma of patients with EwS with metastatic disease. High human satellite 2 and 3 (HSAT2,3) levels in EWS:FLI1- or ERG-expressing cells and tumors were associated with induction of G2/M checkpoint, mitotic spindle, and DNA damage programs. These programs were also activated in EwS EV-treated fibroblasts, coincident with accumulation of HSAT2,3 RNAs, proinflammatory responses, mitotic defects, and senescence. Mechanistically, HSAT2,3-enriched cancer EVs induced cGAS-TBK1 innate immune signaling and formation of cytosolic granules positive for double-strand RNAs, RNA-DNA, and cGAS. Hence, aberrantly expressed ETS proteins derepress pericentromeric heterochromatin, yielding pathogenic RNAs that transmit genotoxic stress and inflammation to local and distant sites. Monitoring HSAT2,3 plasma levels and preventing their dissemination may thus improve therapeutic strategies and blood-based diagnostics.


DNA Damage , Extracellular Vesicles , Oncogene Proteins, Fusion , Proto-Oncogene Protein c-fli-1 , RNA-Binding Protein EWS , Transcriptional Regulator ERG , Humans , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Transcriptional Regulator ERG/genetics , Transcriptional Regulator ERG/metabolism , Male , RNA-Binding Protein EWS/genetics , RNA-Binding Protein EWS/metabolism , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/immunology , Cell Line, Tumor , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Mice , Animals , Heterochromatin/metabolism , Heterochromatin/genetics
2.
Cells ; 10(11)2021 11 08.
Article En | MEDLINE | ID: mdl-34831294

Ewing's sarcoma (EwS) is a pediatric solid tumor entity with low somatic mutational burden and a low rate of tumor-infiltrating T cells, indicating a low extent of immunogenicity. In EwS, immunogenicity may furthermore be significantly diminished by a predominantly M2 macrophage driven pro-tumorigenic tumor microenvironment. In the past, we demonstrated that CHM1319-specific TCR-transgenic T cells are able to control EwS growth in a preclinical mouse model as well as in a patient with metastatic disease. However, new adjuvant techniques to induce long lasting and curative CHM1319-specific TCR-transgenic T cell-mediated anti-tumor responses are needed. In this work, we sought to identify a technique to improve the cytotoxic effect of CHM1319-specific TCR-transgenic T cell by altering the immunogenic cell surface marker expression on EwS cell lines using different cytokines. We demonstrate that TNF, IL-6, IL-1ß and PGE2 cause pro-immunogenic CD83, MHC class I and II as well as ICAM-1 upregulation in EwS cell lines. This observation was associated with significantly improved recognition and killing of the tumor cells by EwS-specific CHM1319/HLA-A*02:01-restricted TCR-transgenic T cells. Conclusively, we demonstrate that the induction of an inflammatory signature renders EwS more susceptible to adoptive T cell therapy. TNF, which is upregulated during inflammatory processes, is of particular translational interest as its secretion may be induced in the patients e.g., by irradiation and hyperthermia in the clinical setting. In future clinical protocols, this finding may be important to identify appropriate conditioning regimens as well as point of time for adoptive T cell-based immunotherapy in EwS patients.


Histocompatibility Antigens Class I/metabolism , Intercellular Adhesion Molecule-1/metabolism , Monocytes/pathology , Sarcoma, Ewing/immunology , T-Lymphocytes, Cytotoxic/immunology , Up-Regulation , Antigens, CD , Cell Line, Tumor , Dendritic Cells/metabolism , Dendritic Cells/pathology , Gene Expression Regulation, Neoplastic , Humans , Immunoglobulins , Membrane Glycoproteins , Sarcoma, Ewing/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/genetics , CD83 Antigen
3.
Front Immunol ; 12: 791206, 2021.
Article En | MEDLINE | ID: mdl-34804076

Osteosarcoma, Ewing sarcoma (EWS), and rhabdomyosarcoma (RMS) are the most common pediatric sarcomas. Conventional therapy for these sarcomas comprises neoadjuvant and adjuvant chemotherapy, surgical resection of the primary tumor and/or radiation therapy. Patients with metastatic, relapsed, or refractory tumors have a dismal prognosis due to resistance to these conventional therapies. Therefore, innovative therapeutic interventions, such as immunotherapy, are urgently needed. Recently, cancer research has focused attention on natural killer (NK) cells due their innate ability to recognize and kill tumor cells. Osteosarcoma, EWS and RMS, are known to be sensitive to NK cell cytotoxicity in vitro. In the clinical setting however, NK cell cytotoxicity against sarcoma cells has been mainly studied in the context of allogeneic stem cell transplantation, where a rapid immune reconstitution of NK cells plays a key role in the control of the disease, known as graft-versus-tumor effect. In this review, we discuss the evidence for the current and future strategies to enhance the NK cell-versus-pediatric sarcoma effect, with a clinical focus. The different approaches encompass enhancing antibody-dependent NK cell cytotoxicity, counteracting the NK cell mechanisms of self-tolerance, and developing adoptive NK cell therapy including chimeric antigen receptor-expressing NK cells.


Bone Neoplasms/immunology , Immunotherapy, Adoptive/methods , Immunotherapy/trends , Killer Cells, Natural/immunology , Osteosarcoma/immunology , Rhabdomyosarcoma/immunology , Sarcoma, Ewing/immunology , Animals , Antibodies/metabolism , Bone Neoplasms/therapy , Child , Cytotoxicity, Immunologic , Graft vs Tumor Effect , Humans , Lymphocyte Activation , Osteosarcoma/therapy , Rhabdomyosarcoma/therapy , Sarcoma, Ewing/therapy
4.
Immunotherapy ; 13(17): 1439-1451, 2021 12.
Article En | MEDLINE | ID: mdl-34670399

Ewing sarcoma is an aggressive tumor type with an age peak in adolescence. Despite the use of dose-intensified chemotherapy as well as radiation and surgery for local control, patients with upfront metastatic disease or relapsed disease have a dismal prognosis, highlighting the need for additional therapeutic options. Different types of immunotherapies have been investigated with only very limited clinical success, which may be due to the presence of immunosuppressive factors in the tumor microenvironment. Here we provide an overview on different factors contributing to Ewing sarcoma immune escape. We demonstrate ways to target these factors in order to make current and future immunotherapies more effective and achieve deeper and more durable responses in patients with Ewing sarcoma.


Lay abstract Ewing sarcoma is an aggressive type of cancer of the bones or soft tissues that mainly affects teenagers. Patients are often treated with intensive treatments which may include chemotherapy in combination with radiation and/or surgery. For patients who present with cancer that has already spread to other sites or that returns after treatment, the cancer can be very hard to cure. This leads to the need for different therapies. Therapies that use the help of the immune system to combat the cancer, called immunotherapies, have had limited success, which is thought to be due to factors in the environment of the tumor that weaken the immune system and so dampen any potential use of it to destroy the cancer cells. We provide an overview of these factors in the tumor environment that allow the cancer cells to escape the immune system; we also discuss potential options to target these factors and, in this way, allow the immunotherapies to destroy the cancer cells more effectively.


Bone Neoplasms/therapy , Immunotherapy , Sarcoma, Ewing/therapy , Tumor Microenvironment/immunology , Adolescent , Bone Neoplasms/immunology , Humans , Sarcoma, Ewing/immunology
5.
Aging (Albany NY) ; 13(13): 17516-17535, 2021 07 07.
Article En | MEDLINE | ID: mdl-34233293

INTRODUCTION: Owing to the poor prognosis of Ewing's sarcoma, reliable prognostic biomarkers are highly warranted for clinical diagnosis of the disease. MATERIALS AND METHODS: A combination of the weighted correlation network analysis and differentially expression analysis was used for initial screening; glycolysis-related genes were extracted and subjected to univariate Cox, LASSO regression, and multivariate Cox analyses to construct prognostic models. The immune cell composition of each sample was analysed using CIBERSORT software. Immunohistochemical analysis was performed for assessing the differential expression of modelled genes in Ewing's sarcoma and paraneoplastic tissues. RESULTS: A logistic regression model constructed for the prognosis of Ewing's sarcoma exhibited that the patient survival rate in the high-risk group is much lower than in the low-risk group. CIBERSORT analysis exhibited a strong correlation of Ewing's sarcoma with naïve B cells, CD8+ T cells, activated NK cells, and M0 macrophages (P < 0.05). Immunohistochemical analysis confirmed the study findings. CONCLUSIONS: GLCE and TPI1 can be used as prognostic biomarkers to predict the prognosis of Ewing's sarcoma, and a close association of Ewing's sarcoma with naïve B cells, CD8+ T cells, activated NK cells, and M0 macrophages provides a novel approach to the disease immunotherapy.


Carbohydrate Epimerases/genetics , Carbohydrate Epimerases/immunology , Glycolysis/genetics , Sarcoma, Ewing/genetics , Sarcoma, Ewing/immunology , Triose-Phosphate Isomerase/genetics , Triose-Phosphate Isomerase/immunology , Biomarkers, Tumor/analysis , Gene Expression Regulation, Neoplastic/genetics , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Macrophages/immunology , Models, Biological , Paraneoplastic Syndromes/pathology , Prognosis , Risk Assessment , Survival Analysis
6.
Cancer Res ; 81(8): 2171-2183, 2021 04 15.
Article En | MEDLINE | ID: mdl-33558334

Ewing sarcoma is the second most common pediatric bone cancer, with a 5-year survival rate for metastatic disease of only 20%. Recent work indicates that survival is strongly correlated with high levels of tumor-infiltrating lymphocytes (TIL), whose abundance is associated with IFN-inducible chemokines CXCL10 and CCL5. However, the tumor-intrinsic factors that drive chemokine production and TIL recruitment have not been fully elucidated. We previously showed that ubiquitin-specific protease 6 (USP6) directly deubiquitinates and stabilizes Jak1, thereby inducing an IFN signature in Ewing sarcoma cells. Here, we show that this gene set comprises chemokines associated with immunostimulatory, antitumorigenic functions, including CXCL10 and CCL5. USP6 synergistically enhanced chemokine production in response to exogenous IFN by inducing surface upregulation of IFNAR1 and IFNGR1. USP6-expressing Ewing sarcoma cells stimulated migration of primary human monocytes and T lymphocytes and triggered activation of natural killer (NK) cells in vitro. USP6 inhibited Ewing sarcoma xenograft growth in nude but not NSG mice and was accompanied by increased intratumoral chemokine production and infiltration and activation of NK cells, dendritic cells, and macrophages, consistent with a requirement for innate immune cells in mediating the antitumorigenic effects of USP6. High USP6 expression in patients with Ewing sarcoma was associated with chemokine production, immune infiltration, and improved survival. This work reveals a previously unrecognized tumor-suppressive function for USP6, which engenders an immunostimulatory microenvironment through pleiotropic effects on multiple immune lineages. This further raises the possibility that USP6 activity may be harnessed to create a "hot" tumor microenvironment in immunotherapy. SIGNIFICANCE: This study reveals a novel tumor-suppressive function for USP6 by inducing an immunostimulatory microenvironment, suggesting that USP6 activity may be exploited to enhance immunotherapy regimens.


Bone Neoplasms/genetics , Lymphocytes, Tumor-Infiltrating , Sarcoma, Ewing/genetics , Tumor Suppressor Proteins/physiology , Ubiquitin Thiolesterase/physiology , Animals , Bone Neoplasms/immunology , Bone Neoplasms/metabolism , Bone Neoplasms/mortality , Cell Movement/drug effects , Chemokine CCL5/biosynthesis , Chemokine CXCL10/biosynthesis , Dendritic Cells/drug effects , Humans , Immunotherapy , Interferons/pharmacology , Janus Kinase 1/metabolism , Killer Cells, Natural/drug effects , Macrophages/drug effects , Mice , Mice, Nude , Neoplasm Transplantation , Receptor, Interferon alpha-beta/metabolism , Receptors, Interferon/metabolism , Sarcoma, Ewing/immunology , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/mortality , Tumor Microenvironment/immunology , Ubiquitin Thiolesterase/immunology , Ubiquitin Thiolesterase/metabolism , Up-Regulation/drug effects , Xenograft Model Antitumor Assays , Interferon gamma Receptor
7.
J Cancer Res Clin Oncol ; 147(1): 153-165, 2021 Jan.
Article En | MEDLINE | ID: mdl-32968877

PURPOSE: Ewing sarcoma (ES) is one of the most common malignant bone tumors in children and adolescents. The immune microenvironment plays an important role in the development of ES. Here, we developed an optimal signature for determining ES patient prognosis based on immune-related genes (IRGs). METHODS: We analyzed the ES gene expression profile dataset, GSE17679, from the GEO database and extracted differential expressed IRGs (DEIRGs). Then, we conducted functional correlation and protein-protein interaction (PPI) analyses of the DEIRGs and used the machine learning algorithm-iterative Lasso Cox regression analysis to build an optimal DEIRG signature. In addition, we applied ES samples from the ICGC database to test the optimal gene signature. We performed univariate and multivariate Cox regressions on clinicopathological characteristics and optimal gene signature to evaluate whether signature is an important prognostic factor. Finally, we calculated the infiltration of 24 immune cells in ES using the ssGSEA algorithm, and analyzed the correlation between the DEIRGs in the optimal gene signature and immune cells. RESULTS: A total of 249 DEIRGs were screened and an 11-gene signature with the strongest correlation with patient prognoses was analyzed using a machine learning algorithm. The 11-gene signature also had a high prognostic value in the ES external verification set. Univariate and multivariate Cox regression analyses showed that 11-gene signature is an independent prognostic factor. We found that macrophages and cytotoxic, CD8 T, NK, mast, B, NK CD56bright, TEM, TCM, and Th2 cells were significantly related to patient prognoses; the infiltration of cytotoxic and CD8 T cells in ES was significantly different. By correlating prognostic biomarkers with immune cell infiltration, we found that FABP4 and macrophages, and NDRG1 and Th2 cells had the strongest correlation. CONCLUSION: Overall, the IRG-related 11-gene signature can be used as a reliable ES prognostic biomarker and can provide guidance for personalized ES therapy.


Biomarkers, Tumor/genetics , Bone Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Machine Learning , Sarcoma, Ewing/pathology , Tumor Microenvironment/immunology , Adolescent , Bone Neoplasms/genetics , Bone Neoplasms/immunology , Female , Follow-Up Studies , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Prognosis , Sarcoma, Ewing/genetics , Sarcoma, Ewing/immunology , Survival Rate
8.
Bull Exp Biol Med ; 170(1): 64-68, 2020 Nov.
Article En | MEDLINE | ID: mdl-33231796

The levels of sPD-1 and sPD-L1 were analyzed in blood serum of 132 patients (age 14-70 years) with primary bone tumors: osteosarcoma (N=39), chondrosarcoma (N=42), Ewing sarcoma (N=9), chordoma (N=12), giant-cell bone tumor (GCBT) (N=16), benign neoplasms (N=14) and in and practically healthy subjects (age 19-58 years; N=27). sPD-L1 levels in all studied bone neoplasms were significantly higher than in the control. Serum sPD-1 level in GCBT patients was significantly higher than in the control, benign neoplasms, chondrosarcoma, and chordoma patients, but did not differ from osteosarcoma group. sPD-1 concentration in Ewing sarcoma was significantly higher than in chordoma and chondrosarcoma, but did not differ from the control. sPD-1 level in chondrosarcoma patients was also lower than in osteosarcoma, Ewing sarcoma, and in the control. Both sPD-1 and sPD-L1 concentrations were not significantly associated with the type of affected bone, process localization, disease stage, tumor histological grade, patients' age and sex. These results suggest the possibility of using these biological markers for preliminary assessment of the character of the process in the bone.


B7-H1 Antigen/genetics , Bone Neoplasms/genetics , Carcinoma, Giant Cell/genetics , Chondrosarcoma/genetics , Chordoma/genetics , Osteosarcoma/genetics , Programmed Cell Death 1 Receptor/genetics , Sarcoma, Ewing/genetics , Adolescent , Adult , Aged , B7-H1 Antigen/blood , Bone Neoplasms/blood , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Carcinoma, Giant Cell/blood , Carcinoma, Giant Cell/immunology , Carcinoma, Giant Cell/pathology , Case-Control Studies , Chondrosarcoma/blood , Chondrosarcoma/immunology , Chondrosarcoma/pathology , Chordoma/blood , Chordoma/immunology , Chordoma/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Neoplasms/blood , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Osteosarcoma/blood , Osteosarcoma/immunology , Osteosarcoma/pathology , Programmed Cell Death 1 Receptor/blood , Sarcoma, Ewing/blood , Sarcoma, Ewing/immunology , Sarcoma, Ewing/pathology
9.
Pediatr Blood Cancer ; 67(10): e28313, 2020 10.
Article En | MEDLINE | ID: mdl-32729251

BACKGROUND: T cells engineered to express chimeric antigen receptors (CARs) are a novel modality to treat refractory cancers. The development of CAR T cells against Ewing sarcoma (EwS) is limited by a lack of targetable surface antigens. We investigated vascular endothelial growth factor receptor 2 (VEGFR2) expressed on tumor-associated blood vessels as potential CAR target in this cancer. METHODS: Expression of VEGFR2 was studied by immunohistochemistry in human EwS biopsies and in murine xenografts and by flow cytometry in EwS cell lines. CARs with short, medium, and long hinge domains against either human or murine VEGFR2 were generated and expressed in human T cells by retroviral gene transfer. The capacity of the individual CARs to activate T cells in response to VEGFR2-expressing cells was compared in vitro. RESULTS: Tumor-associated endothelial cells in human EwS biopsies and in xenografts expressed VEGFR2. Tumor cells in the majority of EwS biopsies were also VEGFR2-positive. Following modification with anti-mouse or anti-human VEGFR2-specific CAR genes, T cells specifically lysed VEGFR2-expressing target cells of the respective species. CAR T cells with short-length or medium-length hinge domains were functionally superior over those with the long hinge region by in vitro parameters, including antigen-specific degranulation responses, lysis of tumor spheroids, tumor necrosis factor α secretion, sequential killing, and proliferation. CONCLUSIONS: VEGFR2 is consistently expressed on endothelial cells of the tumor stroma in EwS and thus is a candidate target for CAR T cells in this cancer. Among various VEGFR2-specific CARs, a construct with a short hinge domain was chosen to be further developed toward clinical translation.


Bone Neoplasms/therapy , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/immunology , Sarcoma, Ewing/therapy , Vascular Endothelial Growth Factor Receptor-2/immunology , Animals , Apoptosis , Bone Neoplasms/immunology , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Proliferation , Humans , Mice , Prognosis , Sarcoma, Ewing/immunology , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Tumor Cells, Cultured , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Xenograft Model Antitumor Assays
10.
Int J Mol Sci ; 20(15)2019 Jul 31.
Article En | MEDLINE | ID: mdl-31370265

Osteosarcoma and Ewing sarcoma are the most common malignant primary bone tumors mainly occurring in children, adolescents and young adults. Current standard therapy includes multidrug chemotherapy and/or radiation specifically for Ewing sarcoma, associated with tumor resection. However, patient survival has not evolved for the past decade and remains closely related to the response of tumor cells to chemotherapy, reaching around 75% at 5 years for patients with localized forms of osteosarcoma or Ewing sarcoma but less than 30% in metastatic diseases and patients resistant to initial chemotherapy. Despite Ewing sarcoma being characterized by specific EWSR1-ETS gene fusions resulting in oncogenic transcription factors, currently, no targeted therapy could be implemented. It seems even more difficult to develop a targeted therapeutic strategy in osteosarcoma which is characterized by high complexity and heterogeneity in genomic alterations. Nevertheless, the common point between these different bone tumors is their ability to deregulate bone homeostasis and remodeling and divert them to their benefit. Therefore, targeting different actors of the bone tumor microenvironment has been hypothesized to develop new therapeutic strategies. In this context, it is well known that the Wnt/ß-catenin signaling pathway plays a key role in cancer development, including osteosarcoma and Ewing sarcoma as well as in bone remodeling. Moreover, recent studies highlight the implication of the Wnt/ß-catenin pathway in angiogenesis and immuno-surveillance, two key mechanisms involved in metastatic dissemination. This review focuses on the role played by this signaling pathway in the development of primary bone tumors and the modulation of their specific microenvironment.


Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic , Osteosarcoma/drug therapy , Sarcoma, Ewing/drug therapy , Tumor Microenvironment/drug effects , Adolescent , Bone Neoplasms/genetics , Bone Neoplasms/immunology , Bone Neoplasms/mortality , Bone and Bones , Child , Humans , Lymphatic Metastasis , Molecular Targeted Therapy/methods , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/mortality , Neovascularization, Pathologic/prevention & control , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/immunology , Osteosarcoma/genetics , Osteosarcoma/immunology , Osteosarcoma/mortality , Proto-Oncogene Proteins c-ets/antagonists & inhibitors , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/immunology , RNA-Binding Protein EWS/antagonists & inhibitors , RNA-Binding Protein EWS/genetics , RNA-Binding Protein EWS/immunology , Sarcoma, Ewing/genetics , Sarcoma, Ewing/immunology , Sarcoma, Ewing/mortality , Survival Analysis , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Wnt Signaling Pathway/drug effects , Young Adult , beta Catenin/antagonists & inhibitors , beta Catenin/genetics , beta Catenin/immunology
11.
Front Immunol ; 10: 651, 2019.
Article En | MEDLINE | ID: mdl-31001265

CD99 (MIC2; single-chain type-1 glycoprotein) is a heavily O-glycosylated transmembrane protein (32 kDa) present on leukocytes and activated endothelium. Expression of CD99 on endothelium is important in lymphocyte diapedesis. CD99 is a diagnostic marker for Ewing's Sarcoma (EWS), as it is highly expressed by these tumors. It has been reported that CD99 can affect the migration, invasion and metastasis of tumor cells. Our results show that CD99 is also highly expressed in the tumor vasculature of most solid tumors. Furthermore, we found that in vitro CD99 expression in cultured endothelial cells is induced by starvation. Targeting of murine CD99 by a conjugate vaccine, which induced antibodies against CD99 in mice, resulted in inhibition of tumor growth in both a tumor model with high CD99 (Os-P0109 osteosarcoma) and low CD99 (CT26 colon carcinoma) expression. We demonstrated that vaccination against CD99 is safe, since no toxicity was observed in mice with high antibody titers against CD99 in their sera during a period of almost 11 months. Targeting of CD99 in humans is more complicated due to the fact that the human and mouse CD99 protein are not identical. We are the first to show that growth factor activated endothelial cells express a distinct human CD99 isoform. We conclude that our observations provide an opportunity for specific targeting of CD99 isoforms in human tumor vasculature.


12E7 Antigen/immunology , Cancer Vaccines/therapeutic use , Endothelium, Vascular/immunology , Sarcoma, Ewing/therapy , Animals , Cell Line, Tumor , Female , Human Umbilical Vein Endothelial Cells/immunology , Humans , Mice, Inbred BALB C , Mice, Inbred C3H , Protein Splicing , Sarcoma, Ewing/immunology , Sarcoma, Ewing/pathology , Tumor Burden
12.
Mol Ther ; 27(5): 933-946, 2019 05 08.
Article En | MEDLINE | ID: mdl-30879952

Chimeric antigen receptor (CAR) engineering of T cells allows one to specifically target tumor cells via cell surface antigens. A candidate target in Ewing sarcoma is the ganglioside GD2, but heterogeneic expression limits its value. Here we report that pharmacological inhibition of Enhancer of Zeste Homolog 2 (EZH2) at doses reducing H3K27 trimethylation, but not cell viability, selectively and reversibly induces GD2 surface expression in Ewing sarcoma cells. EZH2 in Ewing sarcoma cells directly binds to the promoter regions of genes encoding for two key enzymes of GD2 biosynthesis, and EZH2 inhibition enhances expression of these genes. GD2 surface expression in Ewing sarcoma cells is not associated with distinct in vitro proliferation, colony formation, chemosensitivity, or in vivo tumorigenicity. Moreover, disruption of GD2 synthesis by gene editing does not affect its in vitro behavior. EZH2 inhibitor treatment sensitizes Ewing sarcoma cells to effective cytolysis by GD2-specific CAR gene-modified T cells. In conclusion, we report a clinically applicable pharmacological approach for enhancing efficacy of adoptively transferred GD2-redirected T cells against Ewing sarcoma, by enabling recognition of tumor cells with low or negative target expression.


Enhancer of Zeste Homolog 2 Protein/genetics , Gangliosides/genetics , Receptors, Chimeric Antigen/genetics , Sarcoma, Ewing/drug therapy , Antigens, Surface/drug effects , Antigens, Surface/genetics , Benzamides/pharmacology , Biphenyl Compounds , Cell Line, Tumor , Cell Survival/drug effects , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Gangliosides/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunotherapy/methods , Immunotherapy, Adoptive/methods , Indoles/pharmacology , Morpholines , Promoter Regions, Genetic/genetics , Pyridones/pharmacology , Receptors, Chimeric Antigen/immunology , Sarcoma, Ewing/genetics , Sarcoma, Ewing/immunology , Sarcoma, Ewing/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
13.
J Natl Cancer Inst ; 111(9): 970-982, 2019 09 01.
Article En | MEDLINE | ID: mdl-30698726

BACKGROUND: Ewing sarcoma (EWS) manifests one of the lowest somatic mutation rates of any cancer, leading to a scarcity of druggable mutations and neoantigens. Immunotherapeutics targeting differentially expressed cell surface antigens could provide therapeutic benefit for such tumors. Pregnancy-associated plasma protein A (PAPP-A) is a cell membrane-associated proteinase produced by the placenta that promotes fetal growth by inducing insulinlike growth factor (IGF) signaling. METHODS: By comparing RNA expression of cell surface proteins in EWS (n = 120) versus normal tissues (n = 42), we comprehensively characterized the surfaceome of EWS to identify highly differentially expressed molecules. Using CRISPR/Cas-9 and anti-PAPP-A antibodies, we investigated biological roles for PAPP-A in EWS in vitro and in vivo in NSG xenograft models and performed RNA-sequencing on PAPPA knockout clones (n = 5) and controls (n = 3). All statistical tests were two-sided. RESULTS: EWS surfaceome analysis identified 11 highly differentially overexpressed genes, with PAPPA ranking second in differential expression. In EWS cell lines, genetic knockout of PAPPA and treatment with anti-PAPP-A antibodies revealed an essential survival role by regulating local IGF-1 bioavailability. MAb-mediated PAPPA inhibition diminished EWS growth in orthotopic xenografts (leg area mm2 at day 49 IgG2a control (CTRL) [n = 14], mean = 397.0, SD = 86.1 vs anti-PAPP-A [n = 14], mean = 311.7, SD = 155.0; P = .03; median OS anti-PAPP-A = 52.5 days, 95% CI = 46.0 to 63.0 days vs IgG2a = 45.0 days, 95% CI = 42.0 to 52.0 days; P = .02) and improved the efficacy of anti-IGF-1R treatment (leg area mm2 at day 49 anti-PAPP-A + anti-IGF-1R [n = 15], mean = 217.9, SD = 148.5 vs IgG2a-CTRL; P < .001; median OS anti-PAPP-A + anti-IGF1R = 63.0 days, 95% CI = 52.0 to 67.0 days vs IgG2a-CTRL; P < .001). Unexpectedly, PAPPA knockout in EWS cell lines induced interferon (IFN)-response genes, including proteins associated with antigen processing/presentation. Consistently, gene expression profiles in PAPPA-low EWS tumors were enriched for immune response pathways. CONCLUSION: This work provides a comprehensive characterization of the surfaceome of EWS, credentials PAPP-A as a highly differentially expressed therapeutic target, and discovers a novel link between IGF-1 signaling and immune evasion in cancer, thus implicating shared mechanisms of immune evasion between EWS and the placenta.


Pregnancy-Associated Plasma Protein-A/metabolism , Sarcoma, Ewing/immunology , Sarcoma, Ewing/metabolism , Tumor Escape , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Disease Models, Animal , Gene Expression , Gene Expression Profiling , Humans , Immunohistochemistry , Insulin-Like Growth Factor I/metabolism , Male , Mice , Sarcoma, Ewing/mortality , Sarcoma, Ewing/pathology , Signal Transduction , Transcriptome , Tumor Burden , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
14.
Cancer Res ; 79(3): 611-624, 2019 02 01.
Article En | MEDLINE | ID: mdl-30545920

Neuroblastoma is the most common childhood solid tumor, yet the prognosis for high-risk disease remains poor. We demonstrate here that arginase 2 (ARG2) drives neuroblastoma cell proliferation via regulation of arginine metabolism. Targeting arginine metabolism, either by blocking cationic amino acid transporter 1 (CAT-1)-dependent arginine uptake in vitro or therapeutic depletion of arginine by pegylated recombinant arginase BCT-100, significantly delayed tumor development and prolonged murine survival. Tumor cells polarized infiltrating monocytes to an M1-macrophage phenotype, which released IL1ß and TNFα in a RAC-alpha serine/threonine-protein kinase (AKT)-dependent manner. IL1ß and TNFα established a feedback loop to upregulate ARG2 expression via p38 and extracellular regulated kinases 1/2 (ERK1/2) signaling in neuroblastoma and neural crest-derived cells. Proteomic analysis revealed that enrichment of IL1ß and TNFα in stage IV human tumor microenvironments was associated with a worse prognosis. These data thus describe an immune-metabolic regulatory loop between tumor cells and infiltrating myeloid cells regulating ARG2, which can be clinically exploited. SIGNIFICANCE: These findings illustrate that cross-talk between myeloid cells and tumor cells creates a metabolic regulatory loop that promotes neuroblastoma progression.


Arginine/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Neuroblastoma/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Arginase/metabolism , Cell Line, Tumor , Humans , Interleukin-1beta/immunology , MAP Kinase Signaling System , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Transgenic , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neuroblastoma/immunology , Neuroblastoma/pathology , Sarcoma, Ewing/immunology , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Tumor Microenvironment , Tumor Necrosis Factor-alpha/immunology
15.
J Cutan Pathol ; 46(3): 238-241, 2019 Mar.
Article En | MEDLINE | ID: mdl-30582191

Primary cutaneous Ewing sarcoma is a rare clinical presentation of Ewing sarcoma, usually occurring as a small, localized tumor on the extremities of young adults and associated with favorable prognosis. We report a case of primary cutaneous Ewing sarcoma, which presented on the sole of the foot of a 27-year-old patient with relapsed acute myeloid leukemia and neutropenia. Diagnosis was determined through histological features and staining, as well as fluorescence in situ hybridization and molecular testing. The patient underwent wide-local excision with plan to begin targeted chemotherapy, but unfortunately died from adenovirus pneumonia while neutropenic before targeted chemotherapy was initiated.


Immunocompromised Host , Leukemia, Myeloid, Acute/complications , Neutropenia/complications , Sarcoma, Ewing/immunology , Skin Neoplasms/immunology , Adult , Female , Humans
16.
Virchows Arch ; 472(5): 815-824, 2018 May.
Article En | MEDLINE | ID: mdl-29445891

Ewing's sarcoma family of tumors (ESFT) are aggressive neoplasms with scant tumor-infiltrating lymphocytes. We analyzed the immunohistochemical (IHC) expression of PD-L1 and PD-1 and their prognostic significance in clinically localized neoplasms in a cohort of 370 ESFT. Slides prepared from tissue microarrays were stained for PD-L1, PD-1, and CD8. Membranous/cytoplasmic staining over 5% of tumor cells was regarded as positive for PD-L1 and PD-1. Prognostic analysis was done considering only clinically localized tumors (n = 217). PD-L1 expression was present in 19% of ESFT, while PD-1 was expressed in 26%. Forty-eight percent of tumors were negative and 12% were positive for both PD-L1 and PD-1. Metastatic tumors displayed higher expression of PD-L1 (p < 0.0001). Histological subtypes were not correlated with PD-L1 or PD-1 positivity. ESFT with elevated proliferation index (Ki-67) were associated with higher PD-L1 expression (p = 0.049). Regarding prognosis, no significant association was found between PD-L1 expression and progression-free survival (PFS) or overall survival (OS), whereas lack of PD-1 expression in tumor cells was correlated with both poor PFS (p = 0.02) and poor OS (p = 0.004). Tumor-infiltrating CD8(+) T lymphocytes were observed in 15.4% of ESFT with informative results (347 tumors). No correlation was found between tumor-infiltrating CD8(+) T lymphocytes and ESFT histological subtypes, tumor location, or PD-1 and PD-L1 expression, nor with PFS (p = 0.473) or OS (p = 0.087). PD-L1 expression was not significantly related to prognosis. PD-1 was expressed in 26% of ESFT tumor cells and may have prognostic and therapeutic implications. CD8 expression in tumor-infiltrating lymphocytes was not related to prognosis.


Biomarkers, Tumor/immunology , Bone Neoplasms/pathology , CD8-Positive T-Lymphocytes/pathology , Lymphocytes, Tumor-Infiltrating/pathology , Sarcoma, Ewing/pathology , Adolescent , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/biosynthesis , Biomarkers, Tumor/analysis , Bone Neoplasms/immunology , Bone Neoplasms/mortality , Child , Child, Preschool , Disease-Free Survival , Female , Humans , Immunohistochemistry , Infant , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Programmed Cell Death 1 Receptor/biosynthesis , Sarcoma, Ewing/immunology , Sarcoma, Ewing/mortality , Young Adult
17.
Cancer ; 123(19): 3807-3815, 2017 Oct 01.
Article En | MEDLINE | ID: mdl-28608950

BACKGROUND: Programmed death 1 (PD-1) signaling in the tumor microenvironment dampens immune responses to cancer, and blocking this axis induces antitumor effects in several malignancies. Clinical studies of PD-1 blockade are only now being initiated in pediatric patients, and little is known regarding programmed death-ligand 1 (PD-L1) expression in common childhood cancers. The authors characterized PD-L1 expression and tumor-associated immune cells (TAICs) (lymphocytes and macrophages) in common pediatric cancers. METHODS: Whole slide sections and tissue microarrays were evaluated by immunohistochemistry for PD-L1 expression and for the presence of TAICs. TAICs were also screened for PD-L1 expression. RESULTS: Thirty-nine of 451 evaluable tumors (9%) expressed PD-L1 in at least 1% of tumor cells. The highest frequency histotypes comprised Burkitt lymphoma (80%; 8 of 10 tumors), glioblastoma multiforme (36%; 5 of 14 tumors), and neuroblastoma (14%; 17 of 118 tumors). PD-L1 staining was associated with inferior survival among patients with neuroblastoma (P = .004). Seventy-four percent of tumors contained lymphocytes and/or macrophages. Macrophages were significantly more likely to be identified in PD-L1-positive versus PD-L1-negative tumors (P < .001). CONCLUSIONS: A subset of diagnostic pediatric cancers exhibit PD-L1 expression, whereas a much larger fraction demonstrates infiltration with tumor-associated lymphocytes. PD-L1 expression may be a biomarker for poor outcome in neuroblastoma. Further preclinical and clinical investigation will define the predictive nature of PD-L1 expression in childhood cancers both at diagnosis and after exposure to chemoradiotherapy. Cancer 2017;123:3807-3815. © 2017 American Cancer Society.


B7-H1 Antigen/analysis , Lymphocytes, Tumor-Infiltrating , Macrophages , Neoplasm Proteins/analysis , Neoplasms/chemistry , Bone Neoplasms/chemistry , Bone Neoplasms/immunology , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Burkitt Lymphoma/chemistry , Burkitt Lymphoma/immunology , Burkitt Lymphoma/pathology , Child , Glioblastoma/chemistry , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Immunohistochemistry , Neoplasms/immunology , Neoplasms/mortality , Neoplasms/pathology , Neuroblastoma/chemistry , Neuroblastoma/immunology , Neuroblastoma/mortality , Neuroblastoma/pathology , Osteosarcoma/chemistry , Osteosarcoma/immunology , Osteosarcoma/pathology , Rhabdomyosarcoma/chemistry , Rhabdomyosarcoma/immunology , Rhabdomyosarcoma/pathology , Sarcoma, Ewing/chemistry , Sarcoma, Ewing/immunology , Sarcoma, Ewing/pathology , Tissue Array Analysis
18.
Oncotarget ; 7(28): 43267-43280, 2016 Jul 12.
Article En | MEDLINE | ID: mdl-27281613

The endochondral bone protein Chondromodulin-I (CHM1) provides oncogene addiction in Ewing sarcoma (ES). We pre-clinically tested the targetability of CHM1 by TCR transgenic, allo-restricted, peptide specific T cells to treat ES. We previously generated allo-restricted wildtype CD8+ T cells directed against the ES specific antigen CHM1319 causing specific responses against ES. However, utilization of these cells in current therapy protocols is hampered due to high complexity in production, relatively low cell numbers, and rapid T cell exhaustion.In order to provide off-the-shelf products in the future, we successfully generated HLA-A*02:01-restricted T cell receptor (TCR) transgenic T cells directed against CHM1319 by retroviral transduction.After short-term expansion a 100% purified CHM1319-TCR-transgenic T cell population expressed a CD62L+/CD45RO and CD62L+/CD45RA+ phenotype. These cells displayed specific in vitro IFNg and granzyme B release in co-culture with HLA-A*02:01+ ES cell lines expressing CHM1. When co-injected with ES cells in Rag2-/-É£c-/- mice, CHM1-specific TCR-transgenic T cells significantly inhibited the formation of lung and liver metastases in contrast to control mice. Lungs and livers of representative mice displayed CD8+ T cell infiltration in the presence (control group treated with unspecific T cells) and in the absence (study group) of metastatic disease, respectively. Furthermore, mice receiving unspecific T cells showed signs of graft-versus-host-disease in contrast to all mice, receiving CHM1319-TCR-transgenic T cells.CHM1319 specific TCR-transgenic T cells were successfully generated causing anti-ES responses in vitro and in vivo. In the future, CHM1319-TCR-transgenic T cells may control minimal residual disease rendering donor lymphocyte infusions more efficacious and less toxic.


CD8-Positive T-Lymphocytes/transplantation , Hematopoietic Stem Cell Transplantation/methods , Immunotherapy, Adoptive/methods , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Sarcoma, Ewing/therapy , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Coculture Techniques , DNA-Binding Proteins/genetics , Graft vs Host Disease/immunology , Granzymes/metabolism , HLA-A Antigens/genetics , HLA-A Antigens/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunotherapy, Adoptive/adverse effects , Interferon-gamma/metabolism , Liver/pathology , Lung/pathology , Mice , Mice, Inbred BALB C , Oncogene Addiction , Receptors, Antigen, T-Cell/genetics , Retroviridae/genetics , Sarcoma, Ewing/immunology , Sarcoma, Ewing/pathology , Transduction, Genetic , Transplantation, Homologous/adverse effects , Transplantation, Homologous/methods
19.
Cancer Lett ; 374(2): 192-201, 2016 May 01.
Article En | MEDLINE | ID: mdl-26902422

The interaction between tumors cells, tumor-derived humoral factors and the bone marrow in the bone niches has been shown to be essential for bone tumor initiation and promotion. Among the tumor stromal cells, tumor-associated macrophages (TAMs) are usually the most abundant immune population. Previously, we reported that let-7a functions as a tumor suppressor in ES. Herein, we found that the suppressive effects are not only limited on the malignant phenotype of tumor cells but also on the regulation of macrophage infiltration. We observed that the let-7a expression is negatively related to macrophage infiltrations in ES. Moreover, overexpression of putative ts-miRNA let-7a significantly suppressed the recruitment of PBMCs in vitro and decreased the macrophage infiltrations in ES-xenografted tumors in vivo. Most importantly, a positive regulatory feedback loop consisting of let-7a, signal transducer and activator of transcription 3 (STAT3), and nuclear factor-kappa B (NF-κB) (let-7a/STAT3/NF-κB) was involved in let-7a-mediated suppressive effects. These data might provide evidence of a novel intracellular signaling network function in ES pathogenesis, and manipulating this novel feedback loop will have therapeutic potential for ES patients.


Macrophages/immunology , MicroRNAs/biosynthesis , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Sarcoma, Ewing/genetics , Sarcoma, Ewing/immunology , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Macrophages/pathology , Mice , Mice, Nude , MicroRNAs/genetics , NF-kappa B/genetics , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/biosynthesis , STAT3 Transcription Factor/genetics , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Transfection
20.
Clin Cancer Res ; 22(13): 3182-91, 2016 07 01.
Article En | MEDLINE | ID: mdl-26823601

PURPOSE: Patients with metastatic or relapsed pediatric sarcomas receive cytotoxic regimens that induce high remission rates associated with profound lymphocyte depletion, but ultimately few survive long term. We administered adjuvant immunotherapy to patients with metastatic and recurrent pediatric sarcomas in an effort to improve outcomes. EXPERIMENTAL DESIGN: Mononuclear cells were collected via apheresis, and tumor lysate was acquired via percutaneous biopsy at enrollment. Participants received standard antineoplastic therapy, followed by autologous lymphocytes, tumor lysate/keyhole limpet hemocyanin-pulsed dendritic cell vaccinations ± recombinant human IL7. Primary outcomes were toxicity and vaccine responses. Secondary outcomes were immune reconstitution, event-free survival, and overall survival (OS). RESULTS: Forty-three patients enrolled and 29 received immunotherapy. The regimen was well tolerated. Intent-to-treat analysis demonstrated 5-year OS of 51% with significant differences based upon histologic group (63% vs. 0% for Ewing/rhabdomyosarcoma vs. other sarcomas) and response to standard therapy (74% no residual disease vs. 0% residual disease). Five-year intent-to-treat OS of patients with newly diagnosed metastatic Ewing/rhabdomyosarcoma was 77%, higher than previously reported in this population and higher than observed in a similar group treated with an earlier adjuvant immunotherapy regimen (25% 5-year OS). T-cell responses to autologous tumor lysate were identified in 62% of immunotherapy recipients, and survival was higher in those patients (73% 5-year OS with vs. 37% without immune response, P = 0.017). Immune reconstitution, measured by CD4 count recovery, was significantly enhanced in subjects treated with recombinant human IL7. CONCLUSIONS: Adjuvant immunotherapy may improve survival in patients with metastatic pediatric sarcoma. Clin Cancer Res; 22(13); 3182-91. ©2016 AACR.


Dendritic Cells/transplantation , Immunologic Factors/therapeutic use , Immunotherapy, Adoptive/methods , Interleukin-7/therapeutic use , Rhabdomyosarcoma/therapy , Sarcoma, Ewing/therapy , T-Lymphocytes/transplantation , Adolescent , Adult , Chemotherapy, Adjuvant/methods , Child , Child, Preschool , Combined Modality Therapy , Dendritic Cells/immunology , Disease-Free Survival , Female , Humans , Leukapheresis , Male , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/prevention & control , Rhabdomyosarcoma/immunology , Rhabdomyosarcoma/pathology , Sarcoma, Ewing/immunology , Sarcoma, Ewing/pathology , T-Lymphocytes/immunology , Young Adult
...