1
|
Hammoud MK, Meena C, Dietze R, Hoffmann N, Szymanski W, Finkernagel F, Nist A, Stiewe T, Graumann J, von Strandmann EP, Müller R. Arachidonic acid impairs natural killer cell functions by disrupting signaling pathways driven by activating receptors and reactive oxygen species. Cell Commun Signal 2024; 22:555. [PMID: 39563446 PMCID: PMC11575453 DOI: 10.1186/s12964-024-01940-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND High levels of the polyunsaturated fatty acid arachidonic acid (AA) within the ovarian carcinoma (OC) microenvironment correlate with reduced relapse-free survival. Furthermore, OC progression is tied to compromised immunosurveillance, partially attributed to the impairment of natural killer (NK) cells. However, potential connections between AA and NK cell dysfunction in OC have not been studied. METHODS We employed a combination of phosphoproteomics, transcriptional profiling and biological assays to investigate AA's impact on NK cell functions. RESULTS AA (i) disrupts interleukin-2/15-mediated expression of pro-inflammatory genes by inhibiting STAT1-dependent signaling, (ii) hampers signaling by cytotoxicity receptors through disruption of their surface expression, (iii) diminishes phosphorylation of NKG2D-induced protein kinases, including ERK1/2, LYN, MSK1/2 and STAT1, and (iv) alters reactive oxygen species production by transcriptionally upregulating detoxification. These modifications lead to a cessation of NK cell proliferation and a reduction in cytotoxicity. CONCLUSION Our findings highlight significant AA-induced alterations in the signaling network that regulates NK cell activity. As low expression of several NK cell receptors correlates with shorter OC patient survival, these findings suggest a functional linkage between AA, NK cell dysfunction and OC progression.
Collapse
Affiliation(s)
- Mohamad K Hammoud
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
- Institute of Physiological Chemistry, Philipps University, Marburg, Germany
| | - Celina Meena
- Institute of Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Raimund Dietze
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Nathalie Hoffmann
- Institute of Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Witold Szymanski
- Institute of Translational Proteomics, Biochemical/Pharmacological Centre, Philipps University, Marburg, Germany
- Core Facility Translational Proteomics, Philipps University, Marburg, Germany
| | - Florian Finkernagel
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
- Institute of Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Johannes Graumann
- Institute of Translational Proteomics, Biochemical/Pharmacological Centre, Philipps University, Marburg, Germany
- Core Facility Translational Proteomics, Philipps University, Marburg, Germany
| | - Elke Pogge von Strandmann
- Institute of Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany.
- Center for Tumor Biology and Immunology (ZTI), Philipps University, Hans-Meerwein-Strasse 3, 35043, Marburg, Germany.
| | - Rolf Müller
- Department of Translational Oncology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany.
- Center for Tumor Biology and Immunology (ZTI), Philipps University, Hans-Meerwein-Strasse 3, 35043, Marburg, Germany.
| |
Collapse
|
2
|
Liu Z, Yang L, Liu C, Wang Z, Xu W, Lu J, Wang C, Xu X. Identification and validation of immune-related gene signature models for predicting prognosis and immunotherapy response in hepatocellular carcinoma. Front Immunol 2024; 15:1371829. [PMID: 38933262 PMCID: PMC11199539 DOI: 10.3389/fimmu.2024.1371829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Background This study seeks to enhance the accuracy and efficiency of clinical diagnosis and therapeutic decision-making in hepatocellular carcinoma (HCC), as well as to optimize the assessment of immunotherapy response. Methods A training set comprising 305 HCC cases was obtained from The Cancer Genome Atlas (TCGA) database. Initially, a screening process was undertaken to identify prognostically significant immune-related genes (IRGs), followed by the application of logistic regression and least absolute shrinkage and selection operator (LASSO) regression methods for gene modeling. Subsequently, the final model was constructed using support vector machines-recursive feature elimination (SVM-RFE). Following model evaluation, quantitative polymerase chain reaction (qPCR) was employed to examine the gene expression profiles in tissue samples obtained from our cohort of 54 patients with HCC and an independent cohort of 231 patients, and the prognostic relevance of the model was substantiated. Thereafter, the association of the model with the immune responses was examined, and its predictive value regarding the efficacy of immunotherapy was corroborated through studies involving three cohorts undergoing immunotherapy. Finally, the study uncovered the potential mechanism by which the model contributed to prognosticating HCC outcomes and assessing immunotherapy effectiveness. Results SVM-RFE modeling was applied to develop an OS prognostic model based on six IRGs (CMTM7, HDAC1, HRAS, PSMD1, RAET1E, and TXLNA). The performance of the model was assessed by AUC values on the ROC curves, resulting in values of 0.83, 0.73, and 0.75 for the predictions at 1, 3, and 5 years, respectively. A marked difference in OS outcomes was noted when comparing the high-risk group (HRG) with the low-risk group (LRG), as demonstrated in both the initial training set (P <0.0001) and the subsequent validation cohort (P <0.0001). Additionally, the SVMRS in the HRG demonstrated a notable positive correlation with key immune checkpoint genes (CTLA-4, PD-1, and PD-L1). The results obtained from the examination of three cohorts undergoing immunotherapy affirmed the potential capability of this model in predicting immunotherapy effectiveness. Conclusions The HCC predictive model developed in this study, comprising six genes, demonstrates a robust capability to predict the OS of patients with HCC and immunotherapy effectiveness in tumor management.
Collapse
Affiliation(s)
- Zhiqiang Liu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Lingge Yang
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chun Liu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zicheng Wang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Wendi Xu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jueliang Lu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Chunmeng Wang
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xundi Xu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of General Surgery, South China Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
3
|
Seller A, Tegeler CM, Mauermann J, Schreiber T, Hagelstein I, Liebel K, Koch A, Heitmann JS, Greiner SM, Hayn C, Dannehl D, Engler T, Hartkopf AD, Hahn M, Brucker SY, Salih HR, Märklin M. Soluble NKG2DLs Are Elevated in Breast Cancer Patients and Associate with Disease Outcome. Int J Mol Sci 2024; 25:4126. [PMID: 38612935 PMCID: PMC11012452 DOI: 10.3390/ijms25074126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/02/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024] Open
Abstract
Ligands of the natural killer group 2D (NKG2DL) family are expressed on malignant cells and are usually absent from healthy tissues. Recognition of NKG2DLs such as MICA/B and ULBP1-3 by the activating immunoreceptor NKG2D, expressed by NK and cytotoxic T cells, stimulates anti-tumor immunity in breast cancer. Upregulation of membrane-bound NKG2DLs in breast cancer has been demonstrated by immunohistochemistry. Tumor cells release NKG2DLs via proteolytic cleavage as soluble (s)NKG2DLs, which allows for effective immune escape and is associated with poor prognosis. In this study, we collected serum from 140 breast cancer (BC) and 20 ductal carcinoma in situ (DCIS) patients at the time of initial diagnosis and 20 healthy volunteers (HVs). Serum levels of sNKG2DLs were quantified through the use of ELISA and correlated with clinical data. The analyzed sNKG2DLs were low to absent in HVs and significantly higher in BC patients. For some of the ligands analyzed, higher sNKG2DLs serum levels were associated with the classification of malignant tumor (TNM) stage and grading. Low sMICA serum levels were associated with significantly longer progression-free (PFS) and overall survival (OS). In conclusion, we provide the first insights into sNKG2DLs in BC patients and suggest their potential role in tumor immune escape in breast cancer. Furthermore, our observations suggest that serum sMICA levels may serve as a prognostic parameter in the patients analyzed in this study.
Collapse
Affiliation(s)
- Anna Seller
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany; (A.S.)
- Department of Women’s Health, University Hospital Tübingen, Calwerstraße 7, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Röntgenweg 11, 72076 Tübingen, Germany
| | - Christian M. Tegeler
- Department of Women’s Health, University Hospital Tübingen, Calwerstraße 7, 72076 Tübingen, Germany
- Department of Peptide-Based Immunotherapy, Institute of Immunology, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - Jonas Mauermann
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany; (A.S.)
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Röntgenweg 11, 72076 Tübingen, Germany
| | - Tatjana Schreiber
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany; (A.S.)
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Röntgenweg 11, 72076 Tübingen, Germany
| | - Ilona Hagelstein
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany; (A.S.)
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Röntgenweg 11, 72076 Tübingen, Germany
| | - Kai Liebel
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany; (A.S.)
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Röntgenweg 11, 72076 Tübingen, Germany
| | - André Koch
- Department of Women’s Health, University Hospital Tübingen, Calwerstraße 7, 72076 Tübingen, Germany
| | - Jonas S. Heitmann
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany; (A.S.)
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Röntgenweg 11, 72076 Tübingen, Germany
- Department of Peptide-Based Immunotherapy, Institute of Immunology, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - Sarah M. Greiner
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany; (A.S.)
- Department of Women’s Health, University Hospital Tübingen, Calwerstraße 7, 72076 Tübingen, Germany
| | - Clara Hayn
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany; (A.S.)
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Röntgenweg 11, 72076 Tübingen, Germany
| | - Dominik Dannehl
- Department of Women’s Health, University Hospital Tübingen, Calwerstraße 7, 72076 Tübingen, Germany
| | - Tobias Engler
- Department of Women’s Health, University Hospital Tübingen, Calwerstraße 7, 72076 Tübingen, Germany
| | - Andreas D. Hartkopf
- Department of Women’s Health, University Hospital Tübingen, Calwerstraße 7, 72076 Tübingen, Germany
| | - Markus Hahn
- Department of Women’s Health, University Hospital Tübingen, Calwerstraße 7, 72076 Tübingen, Germany
| | - Sara Y. Brucker
- Department of Women’s Health, University Hospital Tübingen, Calwerstraße 7, 72076 Tübingen, Germany
| | - Helmut R. Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany; (A.S.)
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Röntgenweg 11, 72076 Tübingen, Germany
| | - Melanie Märklin
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany; (A.S.)
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Röntgenweg 11, 72076 Tübingen, Germany
| |
Collapse
|
4
|
Jaksik R, Szumała K, Dinh KN, Śmieja J. Multiomics-Based Feature Extraction and Selection for the Prediction of Lung Cancer Survival. Int J Mol Sci 2024; 25:3661. [PMID: 38612473 PMCID: PMC11011391 DOI: 10.3390/ijms25073661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Lung cancer is a global health challenge, hindered by delayed diagnosis and the disease's complex molecular landscape. Accurate patient survival prediction is critical, motivating the exploration of various -omics datasets using machine learning methods. Leveraging multi-omics data, this study seeks to enhance the accuracy of survival prediction by proposing new feature extraction techniques combined with unbiased feature selection. Two lung adenocarcinoma multi-omics datasets, originating from the TCGA and CPTAC-3 projects, were employed for this purpose, emphasizing gene expression, methylation, and mutations as the most relevant data sources that provide features for the survival prediction models. Additionally, gene set aggregation was shown to be the most effective feature extraction method for mutation and copy number variation data. Using the TCGA dataset, we identified 32 molecular features that allowed the construction of a 2-year survival prediction model with an AUC of 0.839. The selected features were additionally tested on an independent CPTAC-3 dataset, achieving an AUC of 0.815 in nested cross-validation, which confirmed the robustness of the identified features.
Collapse
Affiliation(s)
- Roman Jaksik
- Department of Systems Biology and Engineering, Silesian University of Technology, 44-100 Gliwice, Poland;
| | - Kamila Szumała
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, 44-100 Gliwice, Poland;
| | - Khanh Ngoc Dinh
- Irving Institute for Cancer Dynamics and Department of Statistics, Columbia University, New York, NY 10027, USA;
| | - Jarosław Śmieja
- Department of Systems Biology and Engineering, Silesian University of Technology, 44-100 Gliwice, Poland;
| |
Collapse
|
5
|
Nersesian S, Carter EB, Lee SN, Westhaver LP, Boudreau JE. Killer instincts: natural killer cells as multifactorial cancer immunotherapy. Front Immunol 2023; 14:1269614. [PMID: 38090565 PMCID: PMC10715270 DOI: 10.3389/fimmu.2023.1269614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023] Open
Abstract
Natural killer (NK) cells integrate heterogeneous signals for activation and inhibition using germline-encoded receptors. These receptors are stochastically co-expressed, and their concurrent engagement and signaling can adjust the sensitivity of individual cells to putative targets. Against cancers, which mutate and evolve under therapeutic and immunologic pressure, the diversity for recognition provided by NK cells may be key to comprehensive cancer control. NK cells are already being trialled as adoptive cell therapy and targets for immunotherapeutic agents. However, strategies to leverage their naturally occurring diversity and agility have not yet been developed. In this review, we discuss the receptors and signaling pathways through which signals for activation or inhibition are generated in NK cells, focusing on their roles in cancer and potential as targets for immunotherapies. Finally, we consider the impacts of receptor co-expression and the potential to engage multiple pathways of NK cell reactivity to maximize the scope and strength of antitumor activities.
Collapse
Affiliation(s)
- Sarah Nersesian
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Emily B. Carter
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Stacey N. Lee
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | | | - Jeanette E. Boudreau
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
6
|
Song C, Liu W, Jiang G, He Z, Wang R, Wang X, Chen R, Mao W, Zhu S. Identification and validation of a novel NK cells-related signature to predict prognosis and immune microenvironment in LUAD. Immunobiology 2023; 228:152751. [PMID: 37774597 DOI: 10.1016/j.imbio.2023.152751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/11/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023]
Abstract
BACKGROUND The prevalence and fatality rates of lung cancer are experiencing a rapid escalation. Natural Killer (NK) cells have been established to have a crucial role in both tumor initiation and progression. Nevertheless, uncertainties persist regarding their precise implications in the prognosis of LUAD. METHODS The data were obtained from reputable sources, such as the Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) database, and our internally generated sequencing data. Utilizing the TCGA data as a background, we selected intersecting genes, validated by cluster analysis, to establish a Cox model and validated it using the GEO datasets. Furthermore, we conducted extensive analyses to investigate the significance of potential biomarkers in relation to immune cell infiltration, single-cell data, differential gene expression, and drug sensitivity. RESULTS 67 immune-related genes associated with NK cells (NK-IRGs) were identified in the TCGA datasets, whose research potential was demonstrated by cluster analysis. A prognostic signature was identified utilizing the univariate and multivariate Cox model, resulting in the identification of five genes, which was validated using GEO datasets. Additionally, the nomogram's calibration curve demonstrated exceptional concordance between the projected and actual survival rates. Subsequent investigations uncovered that this prognostic signature demonstrated its independence as a risk factor. Notably, in the low-risk group, NK cells exhibited elevated levels of immune checkpoint molecules, indicating heightened sensitivity to immune therapy. These findings highlight the potential of utilizing this signature as a valuable tool in the selection of patients who could benefit from targeted immune interventions.
Collapse
Affiliation(s)
- Chenghu Song
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China
| | - Weici Liu
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China
| | - Guanyu Jiang
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China
| | - Zhao He
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China
| | - Ruixin Wang
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China
| | - Xiaokun Wang
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China
| | - Ruo Chen
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China.
| | - Wenjun Mao
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China.
| | - Shaojin Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China.
| |
Collapse
|
7
|
Tan G, Spillane KM, Maher J. The Role and Regulation of the NKG2D/NKG2D Ligand System in Cancer. BIOLOGY 2023; 12:1079. [PMID: 37626965 PMCID: PMC10452210 DOI: 10.3390/biology12081079] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/22/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023]
Abstract
The family of human NKG2D ligands (NKG2DL) consists of eight stress-induced molecules. Over 80% of human cancers express these ligands on the surface of tumour cells and/or associated stromal elements. In mice, NKG2D deficiency increases susceptibility to some types of cancer, implicating this system in immune surveillance for malignancy. However, NKG2DL can also be shed, released via exosomes and trapped intracellularly, leading to immunosuppressive effects. Moreover, NKG2D can enhance chronic inflammatory processes which themselves can increase cancer risk and progression. Indeed, tumours commonly deploy a range of countermeasures that can neutralise or even corrupt this surveillance system, tipping the balance away from immune control towards tumour progression. Consequently, the prognostic impact of NKG2DL expression in human cancer is variable. In this review, we consider the underlying biology and regulation of the NKG2D/NKG2DL system and its expression and role in a range of cancer types. We also consider the opportunities for pharmacological modulation of NKG2DL expression while cautioning that such interventions need to be carefully calibrated according to the biology of the specific cancer type.
Collapse
Affiliation(s)
- Ge Tan
- CAR Mechanics Group, Guy’s Cancer Centre, School of Cancer and Pharmaceutical Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK;
| | | | - John Maher
- CAR Mechanics Group, Guy’s Cancer Centre, School of Cancer and Pharmaceutical Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK;
- Department of Immunology, Eastbourne Hospital, Kings Drive, Eastbourne BN21 2UD, UK
- Leucid Bio Ltd., Guy’s Hospital, Great Maze Pond, London SE1 9RT, UK
| |
Collapse
|
8
|
Moran J, Mylod E, Kane LE, Marion C, Keenan E, Mekhaeil M, Lysaght J, Dev KK, O’Sullivan J, Conroy MJ. Investigating the Effects of Olaparib on the Susceptibility of Glioblastoma Multiforme Tumour Cells to Natural Killer Cell-Mediated Responses. Pharmaceutics 2023; 15:360. [PMID: 36839682 PMCID: PMC9959685 DOI: 10.3390/pharmaceutics15020360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/09/2023] [Accepted: 01/14/2023] [Indexed: 01/24/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common adult primary brain malignancy, with dismal survival rates of ~14.6 months. The current standard-of-care consists of surgical resection and chemoradiotherapy, however the treatment response is limited by factors such as tumour heterogeneity, treatment resistance, the blood-brain barrier, and immunosuppression. Several immunotherapies have undergone clinical development for GBM but demonstrated inadequate efficacy, yet future combinatorial approaches are likely to hold more promise. Olaparib is FDA-approved for BRCA-mutated advanced ovarian and breast cancer, and clinical studies have revealed its utility as a safe and efficacious radio- and chemo-sensitiser in GBM. The ability of Olaparib to enhance natural killer (NK) cell-mediated responses has been reported in prostate, breast, and lung cancer. This study examined its potential combination with NK cell therapies in GBM by firstly investigating the susceptibility of the GBM cell line T98G to NK cells and, secondly, examining whether Olaparib can sensitise T98G cells to NK cell-mediated responses. Here, we characterise the NK receptor ligand profile of T98G cells and demonstrate that Olaparib does not dampen T98G susceptibility to NK cells or elicit immunomodulatory effects on the function of NK cells. This study provides novel insights into the potential combination of Olaparib with NK cell therapies for GBM.
Collapse
Affiliation(s)
- Jennifer Moran
- Cancer Immunology Research Group, Department of Physiology, Trinity College Dublin, D02 R590 Dublin, Ireland
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Eimear Mylod
- Cancer Immunology Research Group, Department of Physiology, Trinity College Dublin, D02 R590 Dublin, Ireland
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Laura E. Kane
- Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Caroline Marion
- Cancer Immunology Research Group, Department of Physiology, Trinity College Dublin, D02 R590 Dublin, Ireland
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Emily Keenan
- Cancer Immunology Research Group, Department of Physiology, Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Marianna Mekhaeil
- Drug Development Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Joanne Lysaght
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Kumlesh K. Dev
- Drug Development Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Jacintha O’Sullivan
- Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James’s Cancer Institute, St. James’s Hospital, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Melissa J. Conroy
- Cancer Immunology Research Group, Department of Physiology, Trinity College Dublin, D02 R590 Dublin, Ireland
| |
Collapse
|
9
|
Fan J, Shi J, Zhang Y, Liu J, An C, Zhu H, Wu P, Hu W, Qin R, Yao D, Shou X, Xu Y, Tong Z, Wen X, Xu J, Zhang J, Fang W, Lou J, Yin W, Chen W. NKG2D discriminates diverse ligands through selectively mechano-regulated ligand conformational changes. EMBO J 2022; 41:e107739. [PMID: 34913508 PMCID: PMC8762575 DOI: 10.15252/embj.2021107739] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 11/10/2021] [Accepted: 11/18/2021] [Indexed: 11/12/2022] Open
Abstract
Stimulatory immune receptor NKG2D binds diverse ligands to elicit differential anti-tumor and anti-virus immune responses. Two conflicting degeneracy recognition models based on static crystal structures and in-solution binding affinities have been considered for almost two decades. Whether and how NKG2D recognizes and discriminates diverse ligands still remain unclear. Using live-cell-based single-molecule biomechanical assay, we characterized the in situ binding kinetics of NKG2D interacting with different ligands in the absence or presence of mechanical force. We found that mechanical force application selectively prolonged NKG2D interaction lifetimes with the ligands MICA and MICB, but not with ULBPs, and that force-strengthened binding is much more pronounced for MICA than for other ligands. We also integrated steered molecular dynamics simulations and mutagenesis to reveal force-induced rotational conformational changes of MICA, involving formation of additional hydrogen bonds on its binding interface with NKG2D, impeding MICA dissociation under force. We further provided a kinetic triggering model to reveal that force-dependent affinity determines NKG2D ligand discrimination and its downstream NK cell activation. Together, our results demonstrate that NKG2D has a discrimination power to recognize different ligands, which depends on selective mechanical force-induced ligand conformational changes.
Collapse
Affiliation(s)
- Juan Fan
- Department of Cell Biology and Department of Cardiology of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jiawei Shi
- Key Laboratory for Biomedical Engineering of the Ministry of EducationZhejiang UniversityHangzhouChina
| | - Yong Zhang
- Key Laboratory of RNA BiologyCAS Center for Excellence in BiomacromoleculesInstitute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Junwei Liu
- Key Laboratory for Biomedical Engineering of the Ministry of EducationZhejiang UniversityHangzhouChina
- Department of Hepatobiliary and Pancreatic SurgeryThe Center for Integrated Oncology and Precision MedicineAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouChina
| | - Chenyi An
- Department of Cell Biology and Department of Cardiology of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Huaying Zhu
- Department of Cell Biology and Department of Cardiology of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Peng Wu
- Department of Cell Biology and Department of Cardiology of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Wei Hu
- Department of Cell Biology and Department of Cardiology of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Rui Qin
- Department of Cell Biology and Department of Cardiology of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Danmei Yao
- Department of Cell Biology and Department of Cardiology of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Xin Shou
- Institute of Translational MedicineSchool of MedicineZhejiang UniversityHangzhouChina
| | - Yibing Xu
- Institute of Translational MedicineSchool of MedicineZhejiang UniversityHangzhouChina
| | - Zhou Tong
- Department of Medical OncologyFirst Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Xue Wen
- Department of PathologyThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Jianpo Xu
- Center for Stem Cell and Regenerative MedicineDepartment of Basic Medical SciencesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jin Zhang
- Center for Stem Cell and Regenerative MedicineDepartment of Basic Medical SciencesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Laboratory for Systems and Precision MedicineZhejiang University Medical CenterHangzhouChina
- Institute of HematologyZhejiang UniversityHangzhouChina
| | - Weijia Fang
- Department of Medical OncologyFirst Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Jizhong Lou
- Key Laboratory of RNA BiologyCAS Center for Excellence in BiomacromoleculesInstitute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Weiwei Yin
- Key Laboratory for Biomedical Engineering of the Ministry of EducationZhejiang UniversityHangzhouChina
- Department of Thoracic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Provincial Key Laboratory of Cardio‐Cerebral Vascular Detection Technology and Medicinal Effectiveness AppraisalCollege of Biomedical Engineering and Instrument of ScienceZhejiang UniversityHangzhouChina
| | - Wei Chen
- Department of Cell Biology and Department of Cardiology of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory for Biomedical Engineering of the Ministry of EducationZhejiang UniversityHangzhouChina
- Department of Hepatobiliary and Pancreatic SurgeryThe Center for Integrated Oncology and Precision MedicineAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Laboratory for Systems and Precision MedicineZhejiang University Medical CenterHangzhouChina
- The MOE Frontier Science Center for Brain Science & Brain‐machine IntegrationZhejiang UniversityHangzhouChina
| |
Collapse
|
10
|
Peipp M, Klausz K, Boje AS, Zeller T, Zielonka S, Kellner C. Immunotherapeutic targeting of activating natural killer cell receptors and their ligands in cancer. Clin Exp Immunol 2022; 209:22-32. [PMID: 35325068 PMCID: PMC9307233 DOI: 10.1093/cei/uxac028] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/15/2022] [Accepted: 03/22/2022] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cells exert an important role in cancer immune surveillance. Recognition of malignant cells and controlled activation of effector functions are facilitated by the expression of activating and inhibitory receptors, which is a complex interplay that allows NK cells to discriminate malignant cells from healthy tissues. Due to their unique profile of effector functions, the recruitment of NK cells is attractive in cancer treatment and a key function of NK cells in antibody therapy is widely appreciated. In recent years, besides the low-affinity fragment crystallizable receptor for immunoglobulin G (FcγRIIIA), the activating natural killer receptors p30 (NKp30) and p46 (NKp46), as well as natural killer group 2 member D (NKG2D), have gained increasing attention as potential targets for bispecific antibody-derivatives to redirect NK cell cytotoxicity against tumors. Beyond modulation of the receptor activity on NK cells, therapeutic targeting of the respective ligands represents an attractive approach. Here, novel therapeutic approaches to unleash NK cells by engagement of activating NK-cell receptors and alternative strategies targeting their tumor-expressed ligands in cancer therapy are summarized.
Collapse
Affiliation(s)
- Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Katja Klausz
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ammelie Svea Boje
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Tobias Zeller
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Stefan Zielonka
- Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Christian Kellner
- Correspondence: Christian Kellner, Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
11
|
Ruan GT, Xie HL, Zhu LC, Ge YZ, Yan L, Liao C, Gong YZ, Shi HP. Immune ULBP1 is Elevated in Colon Adenocarcinoma and Predicts Prognosis. Front Genet 2022; 13:762514. [PMID: 35211154 PMCID: PMC8862730 DOI: 10.3389/fgene.2022.762514] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 01/10/2022] [Indexed: 01/05/2023] Open
Abstract
Background: Colon adenocarcinoma (COAD) is still the main cause of cancer deaths worldwide. Although immunotherapy has made progress in recent years, there is still a need to improve diagnosis, prognosis, and treatment tools. UL-16 binding protein 1 (ULBP1) is a ligand that activates the receptor natural killer cell group 2 receptor D (NKG2D) and plays an important immunomodulatory role. We aimed to investigate the clinical significance of ULBP1 in COAD. Methods: We obtained the relevant data from The Cancer Genome Atlas (TCGA). A total of 438 patients with COAD were included in this study, with a mean age of 67.1 ± 13.03 years old, of which 234 (53.42%) were male. The diagnostic value of COAD tumor tissues and adjacent tissues was analyzed by ROC curve. Univariate and multivariate survival analysis investigated the prognostic value of ULBP1 gene, and Gene Set Enrichment Analysis (GSEA) curve was performed to analyze the biological process and enriched enrichment pathway of ULBP1 in COAD. Combination survival analysis investigated the combined prognostic effect of prognostic genes. Results:ULBP1 gene had a high diagnostic value in COAD [AUC (TCGA) = 0.959; AUC (Guangxi) = 0.898]. Up-regulated ULBP1 gene of patients with COAD predicted a worse prognosis compared to those patients with down-regulated ULBP1 gene (Adjusted HR = 1.544, 95% CI = 1.020–2.337, p = 0.040). The GSEA showed that ULBP1 was involved in the apoptotic pathway and biological process of T cell mediated cytotoxicity, regulation of natural killer cell activation, and T cell mediated immunity of COAD. The combination survival analysis showed that the combination of high expression of ULBP1, AARS1, and DDIT3 would increase the 2.2-fold death risk of COAD when compared with those of low expression genes. Conclusion: The immune-related ULBP1 gene had diagnostic and prognostic value in COAD. The combination of ULBP1, AARS1, and DDIT3 genes could improve the prognostic prediction performance in COAD.
Collapse
Affiliation(s)
- Guo-Tian Ruan
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Hai-Lun Xie
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Li-Chen Zhu
- Department of Immunology, School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Yi-Zhong Ge
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| | - Lin Yan
- Department of Thoracic Surgery, Affiliated Hospital of Guilin Medical College, Guilin, China
| | - Cun Liao
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yi-Zhen Gong
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Han-Ping Shi
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China
| |
Collapse
|
12
|
Wilczyński JR, Wilczyński M, Paradowska E. Cancer Stem Cells in Ovarian Cancer-A Source of Tumor Success and a Challenging Target for Novel Therapies. Int J Mol Sci 2022; 23:ijms23052496. [PMID: 35269636 PMCID: PMC8910575 DOI: 10.3390/ijms23052496] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 02/04/2023] Open
Abstract
Ovarian cancer is the most lethal neoplasm of the female genital organs. Despite indisputable progress in the treatment of ovarian cancer, the problems of chemo-resistance and recurrent disease are the main obstacles for successful therapy. One of the main reasons for this is the presence of a specific cell population of cancer stem cells. The aim of this review is to show the most contemporary knowledge concerning the biology of ovarian cancer stem cells (OCSCs) and their impact on chemo-resistance and prognosis in ovarian cancer patients, as well as to present the treatment options targeted exclusively on the OCSCs. The review presents data concerning the role of cancer stem cells in general and then concentrates on OCSCs. The surface and intracellular OCSCs markers and their meaning both for cancer biology and clinical prognosis, signaling pathways specifically activated in OCSCs, the genetic and epigenetic regulation of OCSCs function including the recent studies on the non-coding RNA regulation, cooperation between OCSCs and the tumor microenvironment (ovarian cancer niche) including very specific environment such as ascites fluid, the role of shear stress, autophagy and metabolic changes for the function of OCSCs, and finally mechanisms of OCSCs escape from immune surveillance, are described and discussed extensively. The possibilities of anti-OCSCs therapy both in experimental settings and in clinical trials are presented, including the recent II phase clinical trials and immunotherapy. OCSCs are a unique population of cancer cells showing a great plasticity, self-renewal potential and resistance against anti-cancer treatment. They are responsible for the progression and recurrence of the tumor. Several completed and ongoing clinical trials have tested different anti-OCSCs drugs which, however, have shown unsatisfactory efficacy in most cases. We propose a novel approach to ovarian cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Jacek R Wilczyński
- Department of Gynecological Surgery and Gynecological Oncology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
- Correspondence:
| | - Miłosz Wilczyński
- Department of Gynecological, Endoscopic and Oncological Surgery, Polish Mother’s Health Center—Research Institute, 281/289 Rzgowska Str., 93-338 Lodz, Poland;
- Department of Surgical and Endoscopic Gynecology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, 106 Lodowa Str., 93-232 Lodz, Poland;
| |
Collapse
|
13
|
Oliviero B, Varchetta S, Mele D, Pessino G, Maiello R, Falleni M, Tosi D, Donadon M, Soldani C, Franceschini B, Torzilli G, Piccolo G, Barabino M, Opocher E, Maestri M, Bernuzzi S, Wucherpfennig KW, Mondelli MU, Mantovani S. MICA/B-targeted antibody promotes NK cell-driven tumor immunity in patients with intrahepatic cholangiocarcinoma. Oncoimmunology 2022; 11:2035919. [PMID: 35223192 PMCID: PMC8865231 DOI: 10.1080/2162402x.2022.2035919] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The major histocompatibility complex-class I chain related proteins A and B (MICA/B) is upregulated because of cellular stress and MICA/B shedding by cancer cells causes escape from NKG2D recognition favoring the emergence of cancers. Cholangiocarcinoma (CCA) is a relatively rare, though increasingly prevalent, primary liver cancer characterized by a late clinical presentation and a dismal prognosis. We explored the NKG2D-MICA/B axis in NK cells from 41 patients with intrahepatic cholangiocarcinoma (iCCA). The MICA/B-specific 7C6 mAb was used for ex vivo antibody-dependent cytotoxicity (ADCC) experiments using circulating, non tumor liver- and tumor-infiltrating NK cells against the HuCCT-1 cell line and patient-derived primary iCCA cells as targets. MICA/B were more expressed in iCCA than in non-tumoral tissue, MICA transcription being higher in moderately-differentiated compared with poorly-differentiated cancer. Serum MICA was elevated in iCCA patients in line with higher expression of ADAM10 and ADAM17 that are responsible for proteolytic release of MICA/B from tumor. Addition of 7C6 significantly boosted peripheral, liver- and tumor-infiltrating-NK cell degranulation and IFNγ production toward MICA/B-expressing established cell lines and autologous iCCA patient target cells. Our data show that anti-MICA/B drives NK cell anti-tumor activity, and provide preclinical evidence in support of 7C6 as a potential immunotherapeutic tool for iCCA.
Collapse
Affiliation(s)
- Barbara Oliviero
- Division of Clinical Immunology - Infectious Diseases, Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Stefania Varchetta
- Division of Clinical Immunology - Infectious Diseases, Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Dalila Mele
- Division of Clinical Immunology - Infectious Diseases, Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Greta Pessino
- Division of Clinical Immunology - Infectious Diseases, Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Roberta Maiello
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Monica Falleni
- Department of Pathology, Department of Health Sciences, ASST Santi Paolo e Carlo, State University of Milan, Milan, Italy
| | - Delfina Tosi
- Department of Pathology, Department of Health Sciences, ASST Santi Paolo e Carlo, State University of Milan, Milan, Italy
| | - Matteo Donadon
- Department of Hepatobiliary and General Surgery, Humanitas Clinical and Research Center, Humanitas University, Rozzano, Italy
| | - Cristiana Soldani
- Department of Hepatobiliary and General Surgery, Humanitas Clinical and Research Center, Humanitas University, Rozzano, Italy
| | - Barbara Franceschini
- Department of Hepatobiliary and General Surgery, Humanitas Clinical and Research Center, Humanitas University, Rozzano, Italy
| | - Guido Torzilli
- Department of Hepatobiliary and General Surgery, Humanitas Clinical and Research Center, Humanitas University, Rozzano, Italy
| | - Gaetano Piccolo
- Division of Gastrointestinal Surgery, ASST Santi Paolo e Carlo, and State University of Milan, Milan, Italy
| | - Matteo Barabino
- Division of Gastrointestinal Surgery, ASST Santi Paolo e Carlo, and State University of Milan, Milan, Italy
| | - Enrico Opocher
- Division of Gastrointestinal Surgery, ASST Santi Paolo e Carlo, and State University of Milan, Milan, Italy
| | - Marcello Maestri
- Division of General Surgery 1, Department of Surgery, Fondazione Irccs Policlinico San Matteo, Pavia, Italy
| | - Stefano Bernuzzi
- Immunohematology and Transfusion Service, Department of Diagnostic Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Kai W. Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Mario U. Mondelli
- Division of Clinical Immunology - Infectious Diseases, Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy,Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy,CONTACT Mario U. Mondelli UOC Immunologia Clinica – Malattie Infettive, Fondazione IRCCS Policlinico San Matteo, Viale Golgi 19, Pavia27100, Italy
| | - Stefania Mantovani
- Division of Clinical Immunology - Infectious Diseases, Department of Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
14
|
Campos-Silva C, López-Borrego S, Felgueres MJ, Esteso G, Vales-Gomez M. NKG2D Ligands in Liquid Biopsy: The Importance of Soluble and Vesicle-Bound Proteins for Immune Modulation. Crit Rev Immunol 2022; 42:21-40. [PMID: 36374819 DOI: 10.1615/critrevimmunol.2022045263] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The identification of biomarkers allowing diagnostics, prognostics and patient classification is still a challenge in oncological research for patient management. Improvements in patient survival achieved with immunotherapies substantiate that biomarker studies rely not only on cellular pathways contributing to the pathology, but also on the immune competence of the patient. If these immune molecules can be studied in a non-invasive manner, the benefit for patients and clinicians is obvious. The immune receptor Natural Killer Group 2 Member D (NKG2D) represents one of the main systems involved in direct recognition of tumor cells by effector lymphocytes (T and Natural Killer cells), and in immune evasion. The biology of NKG2D and its ligands comprises a complex network of cellular pathways leading to the expression of these tumor-associated ligands on the cell surface or to their release either as soluble proteins, or in extracellular vesicles that potently inhibit NKG2D-mediated responses. Increased levels of NKG2D-ligands in patient serum correlate with tumor progression and poor prognosis; however, most studies did not test the biochemical form of these molecules. Here we review the biology of the NKG2D receptor and ligands, their role in cancer and in patient response to immunotherapies, as well as the changes provoked in this system by non-immune cancer therapies. Further, we discuss the use of NKG2D-L in liquid biopsy, including methods to analyse vesicle-associated proteins. We propose that the evaluation in cancer patients of the whole NKG2D system can provide crucial information about patient immune competence and risk of tumor progression.
Collapse
Affiliation(s)
- Carmen Campos-Silva
- Department of Immunology and Oncology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, Spain
| | - Silvia López-Borrego
- Department of Immunology and Oncology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, Spain
| | - María José Felgueres
- Department of Immunology and Oncology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, Spain
| | - Gloria Esteso
- Department of Immunology and Oncology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, Spain
| | - Mar Vales-Gomez
- Department of Immunology and Oncology, National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, Spain
| |
Collapse
|
15
|
Carmena Moratalla A, Carpentier Solorio Y, Lemaitre F, Farzam-Kia N, Levert A, Zandee SEJ, Lahav B, Guimond JV, Haddad E, Girard M, Duquette P, Larochelle C, Prat A, Arbour N. Stress Signal ULBP4, an NKG2D Ligand, Is Upregulated in Multiple Sclerosis and Shapes CD8 + T-Cell Behaviors. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 9:9/1/e1119. [PMID: 34873031 PMCID: PMC8656234 DOI: 10.1212/nxi.0000000000001119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/19/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND OBJECTIVES We posit the involvement of the natural killer group 2D (NKG2D) pathway in multiple sclerosis (MS) pathology via the presence of specific NKG2D ligands (NKG2DLs). We aim to evaluate the expression of NKG2DLs in the CNS and CSF of patients with MS and to identify cellular stressors inducing the expression of UL16-binding protein 4 (ULBP4), the only detectable NKG2DL. Finally, we evaluate the impact of ULBP4 on functions such as cytokine production and motility by CD8+ T lymphocytes, a subset largely expressing NKG2D, the cognate receptor. METHODS Human postmortem brain samples and CSF from patients with MS and controls were used to evaluate NKG2DL expression. In vitro assays using primary cultures of human astrocytes and neurons were performed to identify stressors inducing ULBP4 expression. Human CD8+ T lymphocytes from MS donors and age/sex-matched healthy controls were isolated to evaluate the functional impact of soluble ULBP4. RESULTS We detected mRNA coding for the 8 identified human NKG2DLs in brain samples from patients with MS and controls, but only ULBP4 protein expression was detectable by Western blot. ULBP4 levels were greater in patients with MS, particularly in active and chronic active lesions and normal-appearing white matter, compared with normal-appearing gray matter from MS donors and white and gray matter from controls. Soluble ULBP4 was also detected in CSF of patients with MS and controls, but a smaller shed/soluble form of 25 kDa was significantly elevated in CSF from female patients with MS compared with controls and male patients with MS. Our data indicate that soluble ULBP4 affects various functions of CD8+ T lymphocytes. First, it enhanced the production of the proinflammatory cytokines GM-CSF and interferon-γ (IFNγ). Second, it increased CD8+ T lymphocyte motility and favored a kinapse-like behavior when cultured in the presence of human astrocytes. CD8+ T lymphocytes from patients with MS were especially altered by the presence of soluble ULBP4 compared with healthy controls. DISCUSSION Our study provides new evidence for the involvement of NKG2D and its ligand ULBP4 in MS pathology. Our results point to ULBP4 as a viable target to specifically block 1 component of the NKG2D pathway without altering immune surveillance involving other NKG2DL.
Collapse
Affiliation(s)
- Ana Carmena Moratalla
- From the Department of Neurosciences (A.C.M., Y.C.S., F.L., N.F-k., A.L., S.E.J.Z., M.G., P.D., C.L., A.P., N.A.), Université de Montréal and Centre de Recherche du CHUM (CRCHUM) Montreal; MS-CHUM Clinic (B.L., M.G., P.D., C.L., A.P.); CLSC des Faubourgs (J.V.G.), CIUSSS du Centre-Sud-de-l'Ile-de-Montréal; and Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics (E.H.), Université de Montréal, Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Yves Carpentier Solorio
- From the Department of Neurosciences (A.C.M., Y.C.S., F.L., N.F-k., A.L., S.E.J.Z., M.G., P.D., C.L., A.P., N.A.), Université de Montréal and Centre de Recherche du CHUM (CRCHUM) Montreal; MS-CHUM Clinic (B.L., M.G., P.D., C.L., A.P.); CLSC des Faubourgs (J.V.G.), CIUSSS du Centre-Sud-de-l'Ile-de-Montréal; and Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics (E.H.), Université de Montréal, Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Florent Lemaitre
- From the Department of Neurosciences (A.C.M., Y.C.S., F.L., N.F-k., A.L., S.E.J.Z., M.G., P.D., C.L., A.P., N.A.), Université de Montréal and Centre de Recherche du CHUM (CRCHUM) Montreal; MS-CHUM Clinic (B.L., M.G., P.D., C.L., A.P.); CLSC des Faubourgs (J.V.G.), CIUSSS du Centre-Sud-de-l'Ile-de-Montréal; and Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics (E.H.), Université de Montréal, Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Negar Farzam-Kia
- From the Department of Neurosciences (A.C.M., Y.C.S., F.L., N.F-k., A.L., S.E.J.Z., M.G., P.D., C.L., A.P., N.A.), Université de Montréal and Centre de Recherche du CHUM (CRCHUM) Montreal; MS-CHUM Clinic (B.L., M.G., P.D., C.L., A.P.); CLSC des Faubourgs (J.V.G.), CIUSSS du Centre-Sud-de-l'Ile-de-Montréal; and Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics (E.H.), Université de Montréal, Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Annie Levert
- From the Department of Neurosciences (A.C.M., Y.C.S., F.L., N.F-k., A.L., S.E.J.Z., M.G., P.D., C.L., A.P., N.A.), Université de Montréal and Centre de Recherche du CHUM (CRCHUM) Montreal; MS-CHUM Clinic (B.L., M.G., P.D., C.L., A.P.); CLSC des Faubourgs (J.V.G.), CIUSSS du Centre-Sud-de-l'Ile-de-Montréal; and Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics (E.H.), Université de Montréal, Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Stephanie E J Zandee
- From the Department of Neurosciences (A.C.M., Y.C.S., F.L., N.F-k., A.L., S.E.J.Z., M.G., P.D., C.L., A.P., N.A.), Université de Montréal and Centre de Recherche du CHUM (CRCHUM) Montreal; MS-CHUM Clinic (B.L., M.G., P.D., C.L., A.P.); CLSC des Faubourgs (J.V.G.), CIUSSS du Centre-Sud-de-l'Ile-de-Montréal; and Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics (E.H.), Université de Montréal, Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Boaz Lahav
- From the Department of Neurosciences (A.C.M., Y.C.S., F.L., N.F-k., A.L., S.E.J.Z., M.G., P.D., C.L., A.P., N.A.), Université de Montréal and Centre de Recherche du CHUM (CRCHUM) Montreal; MS-CHUM Clinic (B.L., M.G., P.D., C.L., A.P.); CLSC des Faubourgs (J.V.G.), CIUSSS du Centre-Sud-de-l'Ile-de-Montréal; and Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics (E.H.), Université de Montréal, Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Jean Victor Guimond
- From the Department of Neurosciences (A.C.M., Y.C.S., F.L., N.F-k., A.L., S.E.J.Z., M.G., P.D., C.L., A.P., N.A.), Université de Montréal and Centre de Recherche du CHUM (CRCHUM) Montreal; MS-CHUM Clinic (B.L., M.G., P.D., C.L., A.P.); CLSC des Faubourgs (J.V.G.), CIUSSS du Centre-Sud-de-l'Ile-de-Montréal; and Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics (E.H.), Université de Montréal, Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Elie Haddad
- From the Department of Neurosciences (A.C.M., Y.C.S., F.L., N.F-k., A.L., S.E.J.Z., M.G., P.D., C.L., A.P., N.A.), Université de Montréal and Centre de Recherche du CHUM (CRCHUM) Montreal; MS-CHUM Clinic (B.L., M.G., P.D., C.L., A.P.); CLSC des Faubourgs (J.V.G.), CIUSSS du Centre-Sud-de-l'Ile-de-Montréal; and Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics (E.H.), Université de Montréal, Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Marc Girard
- From the Department of Neurosciences (A.C.M., Y.C.S., F.L., N.F-k., A.L., S.E.J.Z., M.G., P.D., C.L., A.P., N.A.), Université de Montréal and Centre de Recherche du CHUM (CRCHUM) Montreal; MS-CHUM Clinic (B.L., M.G., P.D., C.L., A.P.); CLSC des Faubourgs (J.V.G.), CIUSSS du Centre-Sud-de-l'Ile-de-Montréal; and Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics (E.H.), Université de Montréal, Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Pierre Duquette
- From the Department of Neurosciences (A.C.M., Y.C.S., F.L., N.F-k., A.L., S.E.J.Z., M.G., P.D., C.L., A.P., N.A.), Université de Montréal and Centre de Recherche du CHUM (CRCHUM) Montreal; MS-CHUM Clinic (B.L., M.G., P.D., C.L., A.P.); CLSC des Faubourgs (J.V.G.), CIUSSS du Centre-Sud-de-l'Ile-de-Montréal; and Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics (E.H.), Université de Montréal, Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Catherine Larochelle
- From the Department of Neurosciences (A.C.M., Y.C.S., F.L., N.F-k., A.L., S.E.J.Z., M.G., P.D., C.L., A.P., N.A.), Université de Montréal and Centre de Recherche du CHUM (CRCHUM) Montreal; MS-CHUM Clinic (B.L., M.G., P.D., C.L., A.P.); CLSC des Faubourgs (J.V.G.), CIUSSS du Centre-Sud-de-l'Ile-de-Montréal; and Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics (E.H.), Université de Montréal, Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Alexandre Prat
- From the Department of Neurosciences (A.C.M., Y.C.S., F.L., N.F-k., A.L., S.E.J.Z., M.G., P.D., C.L., A.P., N.A.), Université de Montréal and Centre de Recherche du CHUM (CRCHUM) Montreal; MS-CHUM Clinic (B.L., M.G., P.D., C.L., A.P.); CLSC des Faubourgs (J.V.G.), CIUSSS du Centre-Sud-de-l'Ile-de-Montréal; and Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics (E.H.), Université de Montréal, Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Nathalie Arbour
- From the Department of Neurosciences (A.C.M., Y.C.S., F.L., N.F-k., A.L., S.E.J.Z., M.G., P.D., C.L., A.P., N.A.), Université de Montréal and Centre de Recherche du CHUM (CRCHUM) Montreal; MS-CHUM Clinic (B.L., M.G., P.D., C.L., A.P.); CLSC des Faubourgs (J.V.G.), CIUSSS du Centre-Sud-de-l'Ile-de-Montréal; and Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics (E.H.), Université de Montréal, Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada.
| |
Collapse
|
16
|
Johnson RL, Cummings M, Thangavelu A, Theophilou G, de Jong D, Orsi NM. Barriers to Immunotherapy in Ovarian Cancer: Metabolic, Genomic, and Immune Perturbations in the Tumour Microenvironment. Cancers (Basel) 2021; 13:6231. [PMID: 34944851 PMCID: PMC8699358 DOI: 10.3390/cancers13246231] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 02/07/2023] Open
Abstract
A lack of explicit early clinical signs and effective screening measures mean that ovarian cancer (OC) often presents as advanced, incurable disease. While conventional treatment combines maximal cytoreductive surgery and platinum-based chemotherapy, patients frequently develop chemoresistance and disease recurrence. The clinical application of immune checkpoint blockade (ICB) aims to restore anti-cancer T-cell function in the tumour microenvironment (TME). Disappointingly, even though tumour infiltrating lymphocytes are associated with superior survival in OC, ICB has offered limited therapeutic benefits. Herein, we discuss specific TME features that prevent ICB from reaching its full potential, focussing in particular on the challenges created by immune, genomic and metabolic alterations. We explore both recent and current therapeutic strategies aiming to overcome these hurdles, including the synergistic effect of combination treatments with immune-based strategies and review the status quo of current clinical trials aiming to maximise the success of immunotherapy in OC.
Collapse
Affiliation(s)
- Racheal Louise Johnson
- Department Gynaecological Oncology, St. James’s University Hospital, Leeds LS9 7TF, UK; (A.T.); (G.T.); (D.d.J.)
| | - Michele Cummings
- Leeds Institute of Medical Research, St. James’s University Hospital, Leeds LS9 7TF, UK; (M.C.); (N.M.O.)
| | - Amudha Thangavelu
- Department Gynaecological Oncology, St. James’s University Hospital, Leeds LS9 7TF, UK; (A.T.); (G.T.); (D.d.J.)
| | - Georgios Theophilou
- Department Gynaecological Oncology, St. James’s University Hospital, Leeds LS9 7TF, UK; (A.T.); (G.T.); (D.d.J.)
| | - Diederick de Jong
- Department Gynaecological Oncology, St. James’s University Hospital, Leeds LS9 7TF, UK; (A.T.); (G.T.); (D.d.J.)
| | - Nicolas Michel Orsi
- Leeds Institute of Medical Research, St. James’s University Hospital, Leeds LS9 7TF, UK; (M.C.); (N.M.O.)
| |
Collapse
|
17
|
Fuertes MB, Domaica CI, Zwirner NW. Leveraging NKG2D Ligands in Immuno-Oncology. Front Immunol 2021; 12:713158. [PMID: 34394116 PMCID: PMC8358801 DOI: 10.3389/fimmu.2021.713158] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/02/2021] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint inhibitors (ICI) revolutionized the field of immuno-oncology and opened new avenues towards the development of novel assets to achieve durable immune control of cancer. Yet, the presence of tumor immune evasion mechanisms represents a challenge for the development of efficient treatment options. Therefore, combination therapies are taking the center of the stage in immuno-oncology. Such combination therapies should boost anti-tumor immune responses and/or target tumor immune escape mechanisms, especially those created by major players in the tumor microenvironment (TME) such as tumor-associated macrophages (TAM). Natural killer (NK) cells were recently positioned at the forefront of many immunotherapy strategies, and several new approaches are being designed to fully exploit NK cell antitumor potential. One of the most relevant NK cell-activating receptors is NKG2D, a receptor that recognizes 8 different NKG2D ligands (NKG2DL), including MICA and MICB. MICA and MICB are poorly expressed on normal cells but become upregulated on the surface of damaged, transformed or infected cells as a result of post-transcriptional or post-translational mechanisms and intracellular pathways. Their engagement of NKG2D triggers NK cell effector functions. Also, MICA/B are polymorphic and such polymorphism affects functional responses through regulation of their cell-surface expression, intracellular trafficking, shedding of soluble immunosuppressive isoforms, or the affinity of NKG2D interaction. Although immunotherapeutic approaches that target the NKG2D-NKG2DL axis are under investigation, several tumor immune escape mechanisms account for reduced cell surface expression of NKG2DL and contribute to tumor immune escape. Also, NKG2DL polymorphism determines functional NKG2D-dependent responses, thus representing an additional challenge for leveraging NKG2DL in immuno-oncology. In this review, we discuss strategies to boost MICA/B expression and/or inhibit their shedding and propose that combination strategies that target MICA/B with antibodies and strategies aimed at promoting their upregulation on tumor cells or at reprograming TAM into pro-inflammatory macrophages and remodeling of the TME, emerge as frontrunners in immuno-oncology because they may unleash the antitumor effector functions of NK cells and cytotoxic CD8 T cells (CTL). Pursuing several of these pipelines might lead to innovative modalities of immunotherapy for the treatment of a wide range of cancer patients.
Collapse
Affiliation(s)
- Mercedes Beatriz Fuertes
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Carolina Inés Domaica
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Norberto Walter Zwirner
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina.,Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
18
|
Cadoux M, Caruso S, Pham S, Gougelet A, Pophillat C, Riou R, Loesch R, Colnot S, Nguyen CT, Calderaro J, Celton-Morizur S, Guerra N, Zucman-Rossi J, Desdouets C, Couty JP. Expression of NKG2D ligands is downregulated by β-catenin signalling and associates with HCC aggressiveness. J Hepatol 2021; 74:1386-1397. [PMID: 33484773 DOI: 10.1016/j.jhep.2021.01.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 12/30/2020] [Accepted: 01/07/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS The NKG2D system is a potent immunosurveillance mechanism in cancer, wherein the activating NK cell receptor (NKG2D) on immune cells recognises its cognate ligands on tumour cells. Herein, we evaluated the expression of NKG2D ligands in hepatocellular carcinoma (HCC), in both humans and mice, taking the genomic features of HCC tumours into account. METHODS The expression of NKG2D ligands (MICA, MICB, ULBP1 and ULBP2) was analysed in large human HCC datasets by Fluidigm TaqMan and RNA-seq methods, and in 2 mouse models (mRNA and protein levels) reproducing the features of both major groups of human tumours. RESULTS We provide compelling evidence that expression of the MICA and MICB ligands in human HCC is associated with tumour aggressiveness and poor patient outcome. We also found that the expression of ULBP1 and ULBP2 was associated with poor patient outcome, and was downregulated in CTNNB1-mutated HCCs displaying low levels of inflammation and associated with a better prognosis. We also found an inverse correlation between ULBP1/2 expression levels and the expression of β-catenin target genes in patients with HCC, suggesting a role for β-catenin signalling in inhibiting expression. We showed in HCC mouse models that β-catenin signalling downregulated the expression of Rae-1 NKG2D ligands, orthologs of ULBPs, through TCF4 binding. CONCLUSIONS We demonstrate that the expression of NKG2D ligands is associated with aggressive liver tumorigenesis and that the downregulation of these ligands by β-catenin signalling may account for the less aggressive phenotype of CTNNB1-mutated HCC tumours. LAY SUMMARY The NKG2D system is a potent immunosurveillance mechanism in cancer. However, its role in hepatocellular carcinoma development has not been widely investigated. Herein, we should that the expression of NKG2D ligands by tumour cells is associated with a more aggressive tumour subtype.
Collapse
Affiliation(s)
- Mathilde Cadoux
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Team Proliferation Stress and Liver Physiopathology, F-75006 Paris, France
| | - Stefano Caruso
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Functional genomics of solid tumors Team, Labex Immuno-Oncology, Paris, France
| | - Sandrine Pham
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Team Proliferation Stress and Liver Physiopathology, F-75006 Paris, France
| | - Angélique Gougelet
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Oncogenic functions of β-catenin signalling in the liver team F-75006 Paris, France
| | - Céline Pophillat
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Team Proliferation Stress and Liver Physiopathology, F-75006 Paris, France
| | - Rozenn Riou
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Oncogenic functions of β-catenin signalling in the liver team F-75006 Paris, France
| | - Robin Loesch
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Oncogenic functions of β-catenin signalling in the liver team F-75006 Paris, France
| | - Sabine Colnot
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Oncogenic functions of β-catenin signalling in the liver team F-75006 Paris, France
| | - Công Trung Nguyen
- Institut Mondor de Recherche Biomédicale, INSERM U955, Créteil, France
| | - Julien Calderaro
- Institut Mondor de Recherche Biomédicale, INSERM U955, Créteil, France
| | - Séverine Celton-Morizur
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Team Proliferation Stress and Liver Physiopathology, F-75006 Paris, France
| | - Nadia Guerra
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Functional genomics of solid tumors Team, Labex Immuno-Oncology, Paris, France
| | - Chantal Desdouets
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Team Proliferation Stress and Liver Physiopathology, F-75006 Paris, France
| | - Jean-Pierre Couty
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Team Proliferation Stress and Liver Physiopathology, F-75006 Paris, France.
| |
Collapse
|
19
|
Ruan GT, Wang S, Zhu LC, Liao XW, Wang XK, Liao C, Yan L, Xie HL, Gong YZ, Gan JL, Gao F. Investigation and verification of the clinical significance and perspective of natural killer group 2 member D ligands in colon adenocarcinoma. Aging (Albany NY) 2021; 13:12565-12586. [PMID: 33909599 PMCID: PMC8148460 DOI: 10.18632/aging.202935] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/14/2021] [Indexed: 12/24/2022]
Abstract
This study investigated and verified the diagnostic and prognostic values of natural killer group 2 member D ligand (NKG2DL) genes in colon adenocarcinoma (COAD). We downloaded NKG2DLs expression data and corresponding clinical parameters from The Cancer Genome Atlas (TCGA) and used bioinformatics techniques to investigate the values of NKG2DLs in COAD. Then, we used the GSE40967 cohort to verify the prognostic value of NKG2DLs. Finally, we verified the ULBP2 expression level in tissues, and also investigated the diagnostic and prognostic values of ULBP2 in COAD. The diagnostic receiver operating characteristic curves showed that ULBP1, ULBP2, ULBP3, and RAET1L had high diagnostic values in COAD [Area Under Curve (AUC) > 0.9]. In TCGA cohort, the univariate and multivariate survival analyses suggested that ULBP2 was correlated with the prognosis of COAD recurrence-free survival (RFS) and overall survival (OS). In GSE40967 cohort, ULBP2 was associated with CC RFS and OS. Reverse transcription-quantitative polymerase chain reaction and immunohistochemistry results showed that ULBP2 was highly expressed in COAD tumor tissues (P < 0.05) and both had diagnostic values (AUC > 0.7). Validated survival analysis showed that the high expression of ULBP2 had a worse prognosis in COAD OS and RFS. Thus, ULBP2 might be an independent diagnostic and prognostic biomarker of COAD.
Collapse
Affiliation(s)
- Guo-Tian Ruan
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Shuai Wang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Li-Chen Zhu
- Department of Immunology, School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xi-Wen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xiang-Kun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Cun Liao
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Ling Yan
- Department of Thoracic Surgery, Affiliated Hospital of Guilin Medical College, Guilin, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Hai-Lun Xie
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Yi-Zhen Gong
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical College, Guilin, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Jia-Liang Gan
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Feng Gao
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| |
Collapse
|
20
|
Terkelsen T, Pernemalm M, Gromov P, Børresen-Dale AL, Krogh A, Haakensen VD, Lethiö J, Papaleo E, Gromova I. High-throughput proteomics of breast cancer interstitial fluid: identification of tumor subtype-specific serologically relevant biomarkers. Mol Oncol 2021; 15:429-461. [PMID: 33176066 PMCID: PMC7858121 DOI: 10.1002/1878-0261.12850] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 08/13/2020] [Accepted: 11/09/2020] [Indexed: 12/24/2022] Open
Abstract
Despite significant advancements in breast cancer (BC) research, clinicians lack robust serological protein markers for accurate diagnostics and tumor stratification. Tumor interstitial fluid (TIF) accumulates aberrantly externalized proteins within the local tumor space, which can potentially gain access to the circulatory system. As such, TIF may represent a valuable starting point for identifying relevant tumor-specific serological biomarkers. The aim of the study was to perform comprehensive proteomic profiling of TIF to identify proteins associated with BC tumor status and subtype. A liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis of 35 TIFs of three main subtypes: luminal (19), Her2 (4), and triple-negative (TNBC) (12) resulted in the identification of > 8800 proteins. Unsupervised hierarchical clustering segregated the TIF proteome into two major clusters, luminal and TNBC/Her2 subgroups. High-grade tumors enriched with tumor infiltrating lymphocytes (TILs) were also stratified from low-grade tumors. A consensus analysis approach, including differential abundance analysis, selection operator regression, and random forest returned a minimal set of 24 proteins associated with BC subtypes, receptor status, and TIL scoring. Among them, a panel of 10 proteins, AGR3, BCAM, CELSR1, MIEN1, NAT1, PIP4K2B, SEC23B, THTPA, TMEM51, and ULBP2, was found to stratify the tumor subtype-specific TIFs. In particular, upregulation of BCAM and CELSR1 differentiates luminal subtypes, while upregulation of MIEN1 differentiates Her2 subtypes. Immunohistochemistry analysis showed a direct correlation between protein abundance in TIFs and intratumor expression levels for all 10 proteins. Sensitivity and specificity were estimated for this protein panel by using an independent, comprehensive breast tumor proteome dataset. The results of this analysis strongly support our data, with eight of the proteins potentially representing biomarkers for stratification of BC subtypes. Five of the most representative proteomics databases currently available were also used to estimate the potential for these selected proteins to serve as putative serological markers.
Collapse
Affiliation(s)
- Thilde Terkelsen
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Maria Pernemalm
- Cancer Proteomics Mass Spectrometry, Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Pavel Gromov
- Breast Cancer Biology Group, Genome Integrity Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Anna-Lise Børresen-Dale
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Norway
| | - Anders Krogh
- Department of Computer Science, University of Copenhagen, Denmark.,Department of Biology, University of Copenhagen, Denmark
| | - Vilde D Haakensen
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Norway
| | - Janne Lethiö
- Cancer Proteomics Mass Spectrometry, Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Elena Papaleo
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark.,Translational Disease System Biology, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Denmark
| | - Irina Gromova
- Breast Cancer Biology Group, Genome Integrity Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
| |
Collapse
|
21
|
Bowden M, Nadal R, Zhou CW, Werner L, Barletta J, Juanpere N, Lloreta J, Hernandez-Llodrà S, Morote J, de Torres I, Orsola A, Cejas P, Long H, Bellmunt J. Transcriptomic analysis of micropapillary high grade T1 urothelial bladder cancer. Sci Rep 2020; 10:20135. [PMID: 33208770 PMCID: PMC7675970 DOI: 10.1038/s41598-020-76904-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 10/20/2020] [Indexed: 11/30/2022] Open
Abstract
No consensus currently exist on the optimal treatment of patients with high-risk nonmuscle invasive (HGT1) micropapillary variant of bladder cancer (MPBC). Transcripsome analysis may allow stratification of MPBC-HGT1 enabling prediction of recurrence and guide therapeutic management for individual patients. Whole transcriptome RNA-Sequencing of tumors from 23 patients with MPBC-HGT1 and 64 conventional urothelial carcinomas (cUC) (reference set) was performed. Differentially expressed genes between MPBC-HGT1 and cUC-HGT1 were explored. Cox proportional hazard models and Kapplan–Meier methods were used to assess the relation between time to progression (TTP) and individual gene expression adjusting for clinical covariates. Over 3000 genes were differentially expressed in MPBC-HGT1 as compared with cUC-HGT1 and a 26-gene signature is characteristic of MPBC within HGT1. A set of three genes; CD36, FAPB3 and RAETE1; were significantly associated with TTP. High expression of FABP3 and CD36 were associated with shorter TTP (p = 0.045 and p = 0.08) as was low expression of RAET1E (p = 0.01). Our study suggest that a 26-gene signature can define MPBC-HGT1 within conventional urothelial carcinomas. A prognostic risk index of three genes (FABP3, CD36 and RAET1E) was found to be associated with shorter TTP and may help classify a group of patients with MPBC-HGT1 with high-risk of early progression. These observations might have implications in terms of radical cystectomy recommendation in MPBC patients.
Collapse
Affiliation(s)
- Michaela Bowden
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, USA.
| | - Rosa Nadal
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institutes, National Institutes of Health, Bethesda, MD, USA
| | - Chensheng W Zhou
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, USA
| | - Lillian Werner
- Department of Biostatistics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Justine Barletta
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Nuria Juanpere
- Department of Pathology, PSMAR-IMIM Research Institute, Barcelona, Spain.,Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Josep Lloreta
- Department of Pathology, PSMAR-IMIM Research Institute, Barcelona, Spain.,Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | | | - Juan Morote
- Department of Urology, Hospital Vall D'Hebron, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Ines de Torres
- Department of Pathology, Hospital Vall D'Hebron, Barcelona, Spain
| | - Anna Orsola
- PSMAR-IMIM Research Institute, Barcelona, Spain
| | - Paloma Cejas
- Center for Functional Cancer Epigenetics, Dana Farber Cancer Institute, Boston, MA, USA
| | - Henry Long
- Center for Functional Cancer Epigenetics, Dana Farber Cancer Institute, Boston, MA, USA
| | - Joaquim Bellmunt
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, USA. .,PSMAR-IMIM Research Institute, Barcelona, Spain. .,Department of Medical Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Av, Boston, 02215, USA.
| |
Collapse
|
22
|
Aruleba RT, Adekiya TA, Molefe PF, Ikwegbue PC, Oyinloye BE, Kappo AP. Insights into functional amino acids of ULBP2 as potential immunogens against cancer. SCIENTIFIC AFRICAN 2020. [DOI: 10.1016/j.sciaf.2020.e00581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
23
|
Macpherson AM, Barry SC, Ricciardelli C, Oehler MK. Epithelial Ovarian Cancer and the Immune System: Biology, Interactions, Challenges and Potential Advances for Immunotherapy. J Clin Med 2020; 9:E2967. [PMID: 32937961 PMCID: PMC7564553 DOI: 10.3390/jcm9092967] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/28/2020] [Accepted: 09/03/2020] [Indexed: 12/11/2022] Open
Abstract
Recent advances in the understanding of immune function and the interactions with tumour cells have led to the development of various cancer immunotherapies and strategies for specific cancer types. However, despite some stunning successes with some malignancies such as melanomas and lung cancer, most patients receive little or no benefit from immunotherapy, which has been attributed to the tumour microenvironment and immune evasion. Although the US Food and Drug Administration have approved immunotherapies for some cancers, to date, only the anti-angiogenic antibody bevacizumab is approved for the treatment of epithelial ovarian cancer. Immunotherapeutic strategies for ovarian cancer are still under development and being tested in numerous clinical trials. A detailed understanding of the interactions between cancer and the immune system is vital for optimisation of immunotherapies either alone or when combined with chemotherapy and other therapies. This article, in two main parts, provides an overview of: (1) components of the normal immune system and current knowledge regarding tumour immunology, biology and their interactions; (2) strategies, and targets, together with challenges and potential innovative approaches for cancer immunotherapy, with attention given to epithelial ovarian cancer.
Collapse
Affiliation(s)
- Anne M. Macpherson
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5000, Australia; (A.M.M.); (C.R.)
| | - Simon C. Barry
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide 5005, Australia;
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5000, Australia; (A.M.M.); (C.R.)
| | - Martin K. Oehler
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5000, Australia; (A.M.M.); (C.R.)
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide 5000, Australia
| |
Collapse
|
24
|
Basher F, Dhar P, Wang X, Wainwright DA, Zhang B, Sosman J, Ji Z, Wu JD. Antibody targeting tumor-derived soluble NKG2D ligand sMIC reprograms NK cell homeostatic survival and function and enhances melanoma response to PDL1 blockade therapy. J Hematol Oncol 2020; 13:74. [PMID: 32517713 PMCID: PMC7285527 DOI: 10.1186/s13045-020-00896-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/08/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Melanoma patients who have detectable serum soluble NKG2D ligands either at the baseline or post-treatment of PD1/PDL1 blockade exhibit poor overall survival. Among families of soluble human NKG2D ligands, the soluble human MHC I chain-related molecule (sMIC) was found to be elevated in melanoma patients and mostly associated with poor response to PD1/PDL1 blockade therapy. METHODS In this study, we aim to investigate whether co-targeting tumor-released sMIC enhances the therapeutic outcome of PD1/PDL1 blockade therapy for melanoma. We implanted sMIC-expressing B16F10 melanoma tumors into syngeneic host and evaluated therapeutic efficacy of anti-sMIC antibody and anti-PDL1 antibody combination therapy in comparison with monotherapy. We analyzed associated effector mechanism. We also assessed sMIC/MIC prevalence in metastatic human melanoma tumors. RESULTS We found that the combination therapy of the anti-PDL1 antibody with an antibody targeting sMIC significantly improved animal survival as compared to monotherapies and that the effect of combination therapy depends significantly on NK cells. We show that combination therapy significantly increased IL-2Rα (CD25) on NK cells which sensitizes NK cells to low dose IL-2 for survival. We demonstrate that sMIC negatively reprograms gene expression related to NK cell homeostatic survival and proliferation and that antibody clearing sMIC reverses the effect of sMIC and reprograms NK cell for survival. We further show that sMIC/MIC is abundantly present in metastatic human melanoma tumors. CONCLUSIONS Our findings provide a pre-clinical proof-of-concept and a new mechanistic understanding to underscore the significance of antibody targeting sMIC to improve therapeutic efficacy of anti-PD1/PDL1 antibody for MIC/sMIC+ metastatic melanoma patients.
Collapse
Affiliation(s)
- Fahmin Basher
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA.,Current address: Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Payal Dhar
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.,Driskill Graduate Program in Life Science, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Xin Wang
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Derek A Wainwright
- Driskill Graduate Program in Life Science, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.,Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.,Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Bin Zhang
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.,Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jeffrey Sosman
- Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Zhe Ji
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.,Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, 60628, USA
| | - Jennifer D Wu
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA. .,Driskill Graduate Program in Life Science, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA. .,Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
25
|
Zhao P, Yang L, Li X, Lu W, Lu F, Wang S, Wang Y, Hua L, Cui C, Dong B, Yu Y, Wang L. Rae1 drives NKG2D binding-dependent tumor development in mice by activating mTOR and STAT3 pathways in tumor cells. Cancer Sci 2020; 111:2234-2247. [PMID: 32333709 PMCID: PMC7385386 DOI: 10.1111/cas.14434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/17/2022] Open
Abstract
Natural killer group 2 member D (NKG2D) ligands (NKG2DLs) on tumor cells engage NKG2D and mediate killing by NKG2D+ immune cells. However, tumor cells with high levels of NKG2DLs are still malignant and proliferate rapidly. We investigated the reason for NKG2DL-expressing cell progression. Tumor cells in mice were assessed for their NKG2DL expression, ability to attract immune cells, tumorigenicity, mTOR, and signal transducer and activator of transcription 3 (STAT3) signaling activation. Antibody blockade was used to determine the effect of NKG2DL-NKG2D interaction on signaling activation in vitro. Retinoic acid early inducible gene 1 (Rae1) was related to the expression of other NKG2DLs, the promotion of tumorigenicity, Mmp2 expression, mTOR and STAT3 phosphorylation in GL261 cells, and the recruitment of NKG2D+ cells in mice. Rae1 also induced NKG2DL expression, mTOR, and STAT3 phosphorylation in GL261 cells and LLC cells, but not in B16 and Pan02 cells, which did not express NKG2DLs, when cocultured with PBMCs; the induced phosphorylation was eliminated by Rae1-NKG2D blockade. Inhibition of mTOR and/or STAT3 decreased PBMC-induced migration and proliferation of GL261 cells in vitro. Rae1, a NKG2DL on tumor cells, plays a driving role in the expression of other NKG2DLs and in tumor development in mice by activating mTOR and STAT3 pathways, relying on its interaction with NKG2D on immune cells.
Collapse
Affiliation(s)
- Peiyan Zhao
- Department of Molecular Biology, College of Basic Medical Sciences and Institute of Pediatrics, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Lei Yang
- Department of Molecular Biology, College of Basic Medical Sciences and Institute of Pediatrics, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Xin Li
- Department of Molecular Biology, College of Basic Medical Sciences and Institute of Pediatrics, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Wenting Lu
- Department of Molecular Biology, College of Basic Medical Sciences and Institute of Pediatrics, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Fangjie Lu
- Department of Molecular Biology, College of Basic Medical Sciences and Institute of Pediatrics, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Shengnan Wang
- Department of Molecular Biology, College of Basic Medical Sciences and Institute of Pediatrics, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Ying Wang
- Department of Molecular Biology, College of Basic Medical Sciences and Institute of Pediatrics, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Li Hua
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Cuiyun Cui
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Boqi Dong
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yongli Yu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Liying Wang
- Department of Molecular Biology, College of Basic Medical Sciences and Institute of Pediatrics, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
26
|
Baci D, Bosi A, Gallazzi M, Rizzi M, Noonan DM, Poggi A, Bruno A, Mortara L. The Ovarian Cancer Tumor Immune Microenvironment (TIME) as Target for Therapy: A Focus on Innate Immunity Cells as Therapeutic Effectors. Int J Mol Sci 2020; 21:ijms21093125. [PMID: 32354198 PMCID: PMC7247443 DOI: 10.3390/ijms21093125] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/24/2020] [Accepted: 04/26/2020] [Indexed: 12/27/2022] Open
Abstract
Ovarian cancer (OvCA) accounts for one of the leading causes of death from gynecologic malignancy. Despite progress in therapy improvements in OvCA, most patients develop a recurrence after first-line treatments, dependent on the tumor and non-tumor complexity/heterogeneity of the neoplasm and its surrounding tumor microenvironment (TME). The TME has gained greater attention in the design of specific therapies within the new era of immunotherapy. It is now clear that the immune contexture in OvCA, here referred as tumor immune microenvironment (TIME), acts as a crucial orchestrator of OvCA progression, thus representing a necessary target for combined therapies. Currently, several advancements of antitumor immune responses in OvCA are based on the characterization of tumor-infiltrating lymphocytes, which have been shown to correlate with a significantly improved clinical outcome. Here, we reviewed the literature on selected TIME components of OvCA, such as macrophages, neutrophils, γδ T lymphocytes, and natural killer (NK) cells; these cells can have a role in either supporting or limiting OvCA, depending on the TIME stimuli. We also reviewed and discussed the major (immune)-therapeutic approaches currently employed to target and/or potentiate macrophages, neutrophils, γδ T lymphocytes, and NK cells in the OvCA context.
Collapse
Affiliation(s)
- Denisa Baci
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (D.B.); (M.G.); (M.R.); (D.M.N.)
| | - Annalisa Bosi
- Laboratory of Pharmacology, Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy;
| | - Matteo Gallazzi
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (D.B.); (M.G.); (M.R.); (D.M.N.)
| | - Manuela Rizzi
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (D.B.); (M.G.); (M.R.); (D.M.N.)
| | - Douglas M. Noonan
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (D.B.); (M.G.); (M.R.); (D.M.N.)
- IRCCS MultiMedica, 20138 Milan, Italy;
| | - Alessandro Poggi
- UOSD Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy;
| | | | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (D.B.); (M.G.); (M.R.); (D.M.N.)
- Correspondence:
| |
Collapse
|
27
|
Papillary Thyroid Carcinoma Variants are Characterized by Co-dysregulation of Immune and Cancer Associated Genes. Cancers (Basel) 2019; 11:cancers11081179. [PMID: 31443155 PMCID: PMC6721495 DOI: 10.3390/cancers11081179] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/08/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023] Open
Abstract
Papillary thyroid carcinoma (PTC) variants exhibit different prognosis, but critical characteristics of PTC variants that contribute to differences in pathogenesis are not well-known. This study aims to characterize dysregulated immune-associated and cancer-associated genes in three PTC subtypes to explore how the interplay between cancer and immune processes causes differential prognosis. RNA-sequencing data from The Cancer Genome Atlas (TCGA) were used to identify dysregulated genes in each variant. The dysregulation profiles of the subtypes were compared using functional pathways clustering and correlations to relevant clinical variables, genomic alterations, and microRNA regulation. We discovered that the dysregulation profiles of classical PTC (CPTC) and the tall cell variant (TCPTC) are similar and are distinct from that of the follicular variant (FVPTC). However, unique cancer or immune-associated genes are associated with clinical variables for each subtype. Cancer-related genes MUC1, FN1, and S100-family members were the most clinically relevant in CPTC, while APLN and IL16, both immune-related, were clinically relevant in FVPTC. RAET-family members, also immune-related, were clinically relevant in TCPTC. Collectively, our data suggest that dysregulation of both cancer and immune associated genes defines the gene expression landscapes of PTC variants, but different cancer or immune related genes may drive the phenotype of each variant.
Collapse
|
28
|
Campos-Silva C, Kramer MK, Valés-Gómez M. NKG2D-ligands: Putting everything under the same umbrella can be misleading. HLA 2019. [PMID: 29521021 DOI: 10.1111/tan.13246] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
NKG2D is a key receptor for the activation of immune effector cells, mainly Natural Killer cells and T lymphocytes, in infection, cancer and autoimmune diseases. Since the detection of ligands for NKG2D in sera of cancer patients is, in many human models, indicative of prognosis, a large number of studies have been undertaken to improve understanding of the biology regulating this receptor and its ligands, with the aim of translating this knowledge into clinical practice. Although it is becoming clear that the NKG2D system can be used as a tool for diagnosis and manipulated for therapy, some questions remain open due to the complexity associated with the existence of a large number of ligands, each one of them displaying distinct biological properties. In this review, we have highlighted some key aspects of this system that differ between humans and mice, including the properties of NKG2D, as well as the genetic and biochemical complexity of NKG2D-ligands. All of these features affect the characteristics of the immune response exerted by NKG2D-expressing cells and are likely to be important factors in the clearance of a tumour or the development of autoimmunity. Implementation of more global analyses, including information on genotype, transcription and protein properties (cellular vs released to the blood stream) of NKG2D-ligands expressed in patients will be necessary to fully understand the links between this system and disease progression.
Collapse
Affiliation(s)
- C Campos-Silva
- Department of Immunology and Oncology, National Centre for Biotechnology, CNB-CSIC, Madrid, Spain
| | - M K Kramer
- Department of Immunology and Oncology, National Centre for Biotechnology, CNB-CSIC, Madrid, Spain
| | - M Valés-Gómez
- Department of Immunology and Oncology, National Centre for Biotechnology, CNB-CSIC, Madrid, Spain
| |
Collapse
|
29
|
Sheppard S, Ferry A, Guedes J, Guerra N. The Paradoxical Role of NKG2D in Cancer Immunity. Front Immunol 2018; 9:1808. [PMID: 30150983 PMCID: PMC6099450 DOI: 10.3389/fimmu.2018.01808] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 07/23/2018] [Indexed: 12/12/2022] Open
Abstract
The activating receptor NKG2D and its ligands are recognized as a potent immune axis that controls tumor growth and microbial infections. With regards to cancer surveillance, various studies have demonstrated the antitumor function mediated by NKG2D on natural killer cells and on conventional and unconventional T cells. The use of NKG2D-deficient mice established the importance of NKG2D in delaying tumor development in transgenic mouse models of cancer. However, we recently demonstrated an unexpected, flip side to this coin, the ability for NKG2D to contribute to tumor growth in a model of inflammation-driven liver cancer. With a focus on the liver, here, we review current knowledge of NKG2D-mediated tumor surveillance and discuss evidence supporting a dual role for NKG2D in cancer immunity. We postulate that in certain advanced cancers, expression of ligands for NKG2D can drive cancer progression rather than rejection. We propose that the nature of the microenvironment within and surrounding tumors impacts the outcome of NKG2D activation. In a form of autoimmune attack, NKG2D promotes tissue damage, mostly in the inflamed tissue adjacent to the tumor, facilitating tumor progression while being ineffective at rejecting transformed cells in the tumor bed.
Collapse
Affiliation(s)
- Sam Sheppard
- Department of Life Sciences, Imperial College London, London, United Kingdom.,Memorial Sloan Kettering Cancer Center, Zuckerman Research Center, New York, NY, United States
| | - Amir Ferry
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Joana Guedes
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Nadia Guerra
- Department of Life Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
30
|
Decreased expression of the NKG2D ligand ULBP4 may be an indicator of poor prognosis in patients with nasopharyngeal carcinoma. Oncotarget 2018; 8:42007-42019. [PMID: 28159927 PMCID: PMC5522045 DOI: 10.18632/oncotarget.14917] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 12/27/2016] [Indexed: 11/25/2022] Open
Abstract
U16-binding protein 4 (ULBP4), a human ligand for natural killer group 2, member D (NKG2D) receptor on NK cells and subsets of T cells, is thought to activate anticancer immune responses. However, the expression pattern and prognostic effect of ULBP4 in nasopharyngeal carcinoma (NPC) has not been investigated. We first compared ULBP4 expression between archival 15 NPC tissues and 8 normal nasopharynx (NP) tissues using qPCR. Then ULBP4 expression among 111 NPC specimens was validated on immunohistochemical examination. In addition, the association of ULBP4 expression with clinical characteristics and survival outcomes was analyzed. Furthermore, the impact of ULBP4 expression in NPC cells on the cytotoxic activity of NK cells was investigated. Both mRNA and protein ULBP4 expressions of NPC tissues were significantly lower than those in normal NP tissues. However, no association of ULBP4 expression with clinical characteristics was observed. Patients with NPC having decreased expression of UBLP4 had significantly poorer overall survival (OS), progression-free survival (PFS), and distant metastasis-free survival (DMFS) than those with preserved levels of ULBP4. On multivariate analyses, low expression of ULBP4 was of borderline significance for OS, PFS, and DMFS (P = 0.060, 0.053, and 0.076, respectively). Further, LDH analysis demonstrated that the cytotoxic activitity of NK cells against C666-1 or 5-8F NPC cells with lenti-ULBP4 was considerably increased as compared to those with lenti-vector at various E/T ratios. Hence, restoration of ULBP4 expression may be a novel therapeutic strategy for treatment of NPC. However, further study is required to confirm these findings.
Collapse
|
31
|
Zöller T, Wittenbrink M, Hoffmeister M, Steinle A. Cutting an NKG2D Ligand Short: Cellular Processing of the Peculiar Human NKG2D Ligand ULBP4. Front Immunol 2018; 9:620. [PMID: 29651291 PMCID: PMC5884875 DOI: 10.3389/fimmu.2018.00620] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/12/2018] [Indexed: 12/25/2022] Open
Abstract
Stress-induced cell surface expression of MHC class I-related glycoproteins of the MIC and ULBP families allows for immune recognition of dangerous "self cells" by human cytotoxic lymphocytes via the NKG2D receptor. With two MIC molecules (MICA and MICB) and six ULBP molecules (ULBP1-6), there are a total of eight human NKG2D ligands (NKG2DL). Since the discovery of the NKG2D-NKG2DL system, the cause for both redundancy and diversity of NKG2DL has been a major and ongoing matter of debate. NKG2DL diversity has been attributed, among others, to the selective pressure by viral immunoevasins, to diverse regulation of expression, to differential tissue expression as well as to variations in receptor interactions. Here, we critically review the current state of knowledge on the poorly studied human NKG2DL ULBP4. Summarizing available facts and previous studies, we picture ULBP4 as a peculiar ULBP family member distinct from other ULBP family members by various aspects. In addition, we provide novel experimental evidence suggesting that cellular processing gives rise to mature ULBP4 glycoproteins different to previous reports. Finally, we report on the proteolytic release of soluble ULBP4 and discuss these results in the light of known mechanisms for generation of soluble NKG2DL.
Collapse
Affiliation(s)
- Tobias Zöller
- Institute for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Mareike Wittenbrink
- Institute for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Meike Hoffmeister
- Institute of Biochemistry II, Goethe University Frankfurt am Main, Frankfurt am Main, Germany.,Brandenburg Medical School (MHB) Theodor Fontane, Institute of Biochemistry, Neuruppin, Germany
| | - Alexander Steinle
- Institute for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
32
|
Ding H, Yang X, Wei Y. Fusion Proteins of NKG2D/NKG2DL in Cancer Immunotherapy. Int J Mol Sci 2018; 19:ijms19010177. [PMID: 29316666 PMCID: PMC5796126 DOI: 10.3390/ijms19010177] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/22/2017] [Accepted: 01/03/2018] [Indexed: 01/25/2023] Open
Abstract
NKG2D (natural killer group 2, member D) is an important activating receptor in natural killer (NK) cells and some T cells. NKG2D ligands (NKG2DLs) are specifically expressed on most tumor cells. The engagement of these ligands on tumor cells to NKG2D on NK cells will induce cell-mediated cytotoxicity and have target cells destroyed. This gives NKG2D/NKG2DLs great potential in cancer therapeutic application. The creation of NKG2D/NKG2DL-based multi-functional fusion proteins is becoming one of the most promising strategies in immunotherapy for cancer. Antibodies, cytokines, and death receptors have been fused with NKG2D or its ligands to produce many powerful fusion proteins, including NKG2D-based chimeric antigen receptors (CARs). In this article, we review the recent developments of the fusion proteins with NKG2D/NKG2DL ligands in cancer immunotherapy.
Collapse
Affiliation(s)
- Hui Ding
- Department of Biological Sciences, Clemson University, 190 Collings Street, Clemson, SC 29634, USA.
| | - Xi Yang
- Department of Biological Sciences, Clemson University, 190 Collings Street, Clemson, SC 29634, USA.
| | - Yanzhang Wei
- Department of Biological Sciences, Clemson University, 190 Collings Street, Clemson, SC 29634, USA.
| |
Collapse
|
33
|
Structural Analysis and Epitope Prediction of MHC Class-1-Chain Related Protein-A for Cancer Vaccine Development. Vaccines (Basel) 2017; 6:vaccines6010001. [PMID: 29295563 PMCID: PMC5874642 DOI: 10.3390/vaccines6010001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/16/2017] [Accepted: 11/21/2017] [Indexed: 11/19/2022] Open
Abstract
Major histocompatibility complex class 1 chain-related gene sequence A is a polymorphic gene found at about 46.6 kb centromeric to HLA-B. It encodes a transmembrane protein, which is a non-classical human leukocyte antigen whose expression is normally induced by stress conditions like cancer and viral infections. The expression of MIC-A leads to the activation of NKG2D receptors of natural killer and T cells, leading to the generation of innate immune response that can easily eliminate/cleanse tumour cells and other cells that express the protein. Several bioinformatics and immunoinformatics tools were used to analyse the sequence and structure of the MIC-A protein. These tools were used in building and evaluating modelled structure of MIC-A, and to predict several antigenic determinant sites on the protein. The MIC-A protein structure generated an average antigenic propensity of 1.0289. Additionally, the hydrophilic regions on the surface of the MIC-A protein where antibodies can be attached were revealed. A total of fourteen antigenic epitopes were predicted, with six found in the transmembrane protein topology, and are predicted to play a role in the development of vaccines that can reactivate the functionalities of the MIC-A protein on the surface of cancer cells in order to elicit a desired immune response.
Collapse
|
34
|
Janssen LME, Ramsay EE, Logsdon CD, Overwijk WW. The immune system in cancer metastasis: friend or foe? J Immunother Cancer 2017; 5:79. [PMID: 29037250 PMCID: PMC5644253 DOI: 10.1186/s40425-017-0283-9] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 09/05/2017] [Indexed: 12/15/2022] Open
Abstract
Metastatic disease is the leading cause of death among cancer patients and involves a complex and inefficient process. Every step of the metastatic process can be rate limiting and is influenced by non-malignant host cells interacting with the tumor cell. Over a century ago, experiments first indicated a link between the immune system and metastasis. This phenomenon, called concomitant immunity, indicates that the primary tumor induces an immune response, which may not be sufficient to destroy the primary tumor, but prevents the growth of a secondary tumor or metastases. Since that time, many different immune cells have been shown to play a role in both inhibiting and promoting metastatic disease. Here we review classic and new observations, describing the links between the immune system and metastasis that inform the development of cancer therapies.
Collapse
Affiliation(s)
- Louise M E Janssen
- Departments of Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Emma E Ramsay
- Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Craig D Logsdon
- Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Willem W Overwijk
- Departments of Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA. .,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
35
|
Vyas M, Reinartz S, Hoffmann N, Reiners KS, Lieber S, Jansen JM, Wagner U, Müller R, von Strandmann EP. Soluble NKG2D ligands in the ovarian cancer microenvironment are associated with an adverse clinical outcome and decreased memory effector T cells independent of NKG2D downregulation. Oncoimmunology 2017; 6:e1339854. [PMID: 28932639 PMCID: PMC5599084 DOI: 10.1080/2162402x.2017.1339854] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/01/2017] [Accepted: 06/02/2017] [Indexed: 12/12/2022] Open
Abstract
The immune receptor NKG2D is predominantly expressed on NK cells and T cell subsets and confers anti-tumor activity. According to the current paradigm, immune surveillance is counteracted by soluble ligands shed into the microenvironment, which down-regulate NKG2D receptor expression. Here, we analyzed the clinical significance of the soluble NKG2D ligands sMICA and sULBP2 in the malignancy-associated ascites of ovarian cancer. We show that high levels of sMICA and sULBP2 in ascites were associated with a poor prognosis. Ascites inhibited the activation of normal NK cells, which, in contrast to the prevailing notion, was not associated with decreased NKG2D expression. Of note, an inverse correlation of soluble NKG2D ligands with effector memory T cells and a direct correlation with pro-tumorigenic CD163+CD206+ macrophages was observed. Thus, the role of soluble NKG2D ligands within the ovarian cancer microenvironment is more complex than anticipated and does not exclusively function via NKG2D downregulation.
Collapse
Affiliation(s)
- Maulik Vyas
- Experimental Tumor Research, Center for Tumor Biology and Immunology, Clinic for Hematology, Oncology and Immunology, Philipps University, Marburg, Germany
| | - Silke Reinartz
- Clinic for Gynecology, Gynecologic Oncology and Endocrinology, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Nathalie Hoffmann
- Experimental Tumor Research, Center for Tumor Biology and Immunology, Clinic for Hematology, Oncology and Immunology, Philipps University, Marburg, Germany.,Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Katrin S Reiners
- Experimental Tumor Research, Center for Tumor Biology and Immunology, Clinic for Hematology, Oncology and Immunology, Philipps University, Marburg, Germany
| | - Sonja Lieber
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Julia M Jansen
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, University Hospital Giessen and Marburg (UKGM), Marburg, Germany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, University Hospital Giessen and Marburg (UKGM), Marburg, Germany
| | - Rolf Müller
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Elke Pogge von Strandmann
- Experimental Tumor Research, Center for Tumor Biology and Immunology, Clinic for Hematology, Oncology and Immunology, Philipps University, Marburg, Germany
| |
Collapse
|
36
|
Demoulin B, Cook WJ, Murad J, Graber DJ, Sentman ML, Lonez C, Gilham DE, Sentman CL, Agaugue S. Exploiting natural killer group 2D receptors for CAR T-cell therapy. Future Oncol 2017; 13:1593-1605. [PMID: 28613086 DOI: 10.2217/fon-2017-0102] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chimeric antigen receptors (CARs) are genetically engineered proteins that combine an extracellular antigen-specific recognition domain with one or several intracellular T-cell signaling domains. When expressed in T cells, these CARs specifically trigger T-cell activation upon antigen recognition. While the clinical proof of principle of CAR T-cell therapy has been established in hematological cancers, CAR T cells are only at the early stages of being explored to tackle solid cancers. This special report discusses the concept of exploiting natural killer cell receptors as an approach that could broaden the specificity of CAR T cells and potentially enhance the efficacy of this therapy against solid tumors. New data demonstrating feasibility of this approach in humans and supporting the ongoing clinical trial are also presented.
Collapse
Affiliation(s)
- Benjamin Demoulin
- Research & Development Department, Celyad SA, Mont-Saint-Guibert, Belgium
| | - W James Cook
- Center for Sy+nthetic Immunity, Department of Microbiology & Immunology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | | | - David J Graber
- Center for Sy+nthetic Immunity, Department of Microbiology & Immunology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Marie-Louise Sentman
- Center for Sy+nthetic Immunity, Department of Microbiology & Immunology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Caroline Lonez
- Research & Development Department, Celyad SA, Mont-Saint-Guibert, Belgium
| | - David E Gilham
- Research & Development Department, Celyad SA, Mont-Saint-Guibert, Belgium
| | - Charles L Sentman
- Center for Sy+nthetic Immunity, Department of Microbiology & Immunology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Sophie Agaugue
- Research & Development Department, Celyad SA, Mont-Saint-Guibert, Belgium
| |
Collapse
|
37
|
Garrido-Tapia M, Hernández CJ, Ascui G, Kramm K, Morales M, Ga Rate V, Zúñiga R, Bustamante M, Aguillón JC, Catala N D, Ribeiro CH, Molina MAC. STAT3 inhibition by STA21 increases cell surface expression of MICB and the release of soluble MICB by gastric adenocarcinoma cells. Immunobiology 2017; 222:1043-1051. [PMID: 28578917 DOI: 10.1016/j.imbio.2017.05.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/27/2017] [Accepted: 05/14/2017] [Indexed: 02/04/2023]
Abstract
NKG2D is an activating receptor expressed on NK cells that binds to a variety of ligands, including MICA and MICB. These cell surface glycoproteins are overexpressed under cellular transformation, thus playing an important role in cell-mediated immune response to tumors. STAT3 is a transcription factor that is constitutively active in cancer. It negatively regulates MICA expression on target cells, while its inhibition enhances NK cell cytotoxicity against tumors. In this work, we aimed to describe the effect of STAT3 signaling inhibition by STA21 on the regulation of MICB expression in gastric adenocarcinoma cells and its effect on the cytotoxic function of NK cells. Treatment of gastric adenocarcinoma cells with STA21 induced an increase in MICB expression and soluble MICB secretion, as well as a variable pattern on effector cell degranulation. Soluble MICB secretion by gastric adenocarcinoma cells was not affected by metalloprotease inhibition. We also observed that primary gastric adenocarcinoma tissue released soluble MICB into the extracellular milieu. Recombinant MICB induced a significant decrease in the levels of NKG2D receptor on effector NK and CD8+ T cells, which correlated with an impaired cytotoxic function. Altogether, our data provide evidence that STAT3 signaling pathway regulates MICB expression on gastric adenocarcinoma cells and that recombinant soluble MICB compromises the cytolytic activity of NK cells.
Collapse
Affiliation(s)
- Macarena Garrido-Tapia
- Laboratorio de Anticuerpos Recombinantes e Inmunoterapia anti tumoral. Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Chile.
| | - Carolina J Hernández
- Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Chile.
| | - Gabriel Ascui
- Laboratorio de Inmunoedición del Cáncer, Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Chile.
| | - Karina Kramm
- Laboratorio de Anticuerpos Recombinantes e Inmunoterapia anti tumoral. Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Chile.
| | - Marcela Morales
- Laboratorio de Anticuerpos Recombinantes e Inmunoterapia anti tumoral. Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Chile.
| | - Valentina Ga Rate
- Laboratorio de Anticuerpos Recombinantes e Inmunoterapia anti tumoral. Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Chile.
| | - Roberto Zúñiga
- Centro de Inmunobiotecnología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Chile; Programa de Doctorado en Química, Universidad de la República Oriental de Uruguay, Uruguay.
| | - Marco Bustamante
- Departamento de Cirugía Digestiva, Hospital del Salvador, Facultad de Medicina, Universidad de Chile, Chile.
| | - Juan Carlos Aguillón
- Laboratorio de Enfermedades Autoinmunes e Inflamatorias, Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Chile.
| | - Diego Catala N
- Laboratorio de Inmunoregulación, Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Chile.
| | - Carolina H Ribeiro
- Laboratorio de Inmunoedición del Cáncer, Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Chile.
| | - Mari A Carmen Molina
- Laboratorio de Anticuerpos Recombinantes e Inmunoterapia anti tumoral. Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Chile; Centro de Inmunobiotecnología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Chile.
| |
Collapse
|
38
|
Cai X, Caballero-Benitez A, Gewe MM, Jenkins IC, Drescher CW, Strong RK, Spies T, Groh V. Control of Tumor Initiation by NKG2D Naturally Expressed on Ovarian Cancer Cells. Neoplasia 2017; 19:471-482. [PMID: 28499126 PMCID: PMC5429243 DOI: 10.1016/j.neo.2017.03.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/20/2017] [Accepted: 03/24/2017] [Indexed: 01/06/2023] Open
Abstract
Cancer cells may co-opt the NKG2D lymphocyte receptor to complement the presence of its ligands for autonomous stimulation of oncogenic signaling. Previous studies raise the possibility that cancer cell NKG2D may induce high malignancy traits, but its full oncogenic impact is unknown. Using epithelial ovarian cancer as model setting, we show here that ex vivo NKG2D+ cancer cells have stem-like capacities, and provide formal in vivo evidence linking NKG2D stimulation with the development and maintenance of these functional states. NKG2D+ ovarian cancer cell populations harbor substantially greater capacities for self-renewing in vitro sphere formation and in vivo tumor initiation in immunodeficient (NOD scid gamma) mice than NKG2D− controls. Sphere formation and tumor initiation are impaired by NKG2D silencing or ligand blockade using antibodies or a newly designed pan ligand-masking NKG2D multimer. In further support of pathophysiological significance, a prospective study of 47 high-grade serous ovarian cancer cases revealed that the odds of disease recurrence were significantly greater and median progression-free survival rates higher among patients with above and below median NKG2D+ cancer cell frequencies, respectively. Collectively, our results define cancer cell NKG2D as an important regulator of tumor initiation in ovarian cancer and presumably other malignancies and thus challenge current efforts in immunotherapy aimed at enhancing NKG2D function.
Collapse
Affiliation(s)
- Xin Cai
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA, 98112, USA.
| | - Andrea Caballero-Benitez
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA, 98112, USA.
| | - Mesfin M Gewe
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA, 98112, USA.
| | - Isaac C Jenkins
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA, 98112, USA.
| | - Charles W Drescher
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA, 98112, USA.
| | - Roland K Strong
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA, 98112, USA.
| | - Thomas Spies
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA, 98112, USA.
| | - Veronika Groh
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA, 98112, USA.
| |
Collapse
|
39
|
Liu J, Wang L, Wang Y, Zhang W, Cao Y. Phenotypic characterization and anticancer capacity of CD8+ cytokine-induced killer cells after antigen-induced expansion. PLoS One 2017; 12:e0175704. [PMID: 28426690 PMCID: PMC5398541 DOI: 10.1371/journal.pone.0175704] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/30/2017] [Indexed: 01/12/2023] Open
Abstract
Cytokine-induced killer cells (CIK) have been used in clinic for adoptive immunotherapy in a variety of malignant tumors and have improved the prognosis of cancer patients. However, there are individual differences in the CIK cell preparations including the obvious differences in the ratio of effector CIK cells among different cancer patients. Infusion of such heterogeneous immune cell preparation is an important factor that would affect the therapeutic efficacy. We report here the enrichment and expansion of CD8+ cells from CIK cells cultured for one week using magnetic activated cell sorting (MACS). These enriched CD8+ CIK cells expressed T cell marker CD3 and antigen recognition receptor NKG2D. Phenotypic analysis showed that CD8+ CIK cells contained 32.4% of CD3+ CD56+ natural killer (NK)-like T cells, 23.6% of CD45RO+ CD28+, and 50.5% of CD45RA+ CD27+ memory T cells. In vitro cytotoxic activity assay demonstrated that the enriched CD8+ CIK cells had significant cytotoxic activity against K562 cells and five ovarian cancer cell lines. Intriguingly, CD8+ CIK cells had strong cytotoxic activity against OVCAR3 cells that has weak binding capability to NKG2D. Flow cytometry and quantitative RT-PCR analysis revealed that OVCAR3 cells expressed HLA-I and OCT4 and Sox2, suggesting that CD8+ CIK cells recognize surface antigen via specific T cell receptor and effectively kill the target cells. The results suggest that transplantation of such in vitro enriched and expanded OCT4-specific CD8+ CIK cells may improve the specific immune defense mechanism against cancer stem cells, providing a novel avenue of cancer stem cell targeted immunotherapy for clinical treatment of ovarian cancer.
Collapse
Affiliation(s)
- Jianhua Liu
- Department of Plastic Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lu Wang
- Department of Plastic Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yaoling Wang
- Department of Plastic Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wenjie Zhang
- Department of Plastic Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
- * E-mail: (YC); (WZ)
| | - Yilin Cao
- Department of Plastic Surgery, Shanghai Ninth People’s Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
- * E-mail: (YC); (WZ)
| |
Collapse
|
40
|
Implication of combined PD-L1/PD-1 blockade with cytokine-induced killer cells as a synergistic immunotherapy for gastrointestinal cancer. Oncotarget 2016; 7:10332-44. [PMID: 26871284 PMCID: PMC4891123 DOI: 10.18632/oncotarget.7243] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/24/2016] [Indexed: 12/14/2022] Open
Abstract
Cytokine-induced killer (CIK) cells represent a realistic approach in cancer immunotherapy with confirmed survival benefits in the context of metastatic solid tumors. However, therapeutic effects are limited to a fraction of patients. In this study, immune-resistance elements and ideal combination therapies were explored. Initially, phenotypic analysis was performed to document CD3, CD56, NKG2D, DNAM-1, PD-L1, PD-1, CTLA-4, TIM-3, 2B4, and LAG-3 on CIK cells. Upon engagement of CIK cells with the tumor cells, expression of PD-1 on CIK cells and PD-L1 on both cells were up-regulated. Over-expression of PD-L1 levels on tumor cells via lentiviral transduction inhibited tumoricidal activity of CIK cells, and neutralizing of PD-L1/PD-1 signaling axis could enhance their tumor-killing effect. Conversely, blockade of NKG2D, a major activating receptor of CIK cells, largely caused dysfunction of CIK cells. Functional study showed an increase of NKG2D levels along with PD-L1/PD-1 blockade in the presence of other immune effector molecule secretion. Additionally, combined therapy of CIK infusion and PD-L1/PD-1 blockade caused a delay of in vivo tumor growth and exhibited a survival advantage over untreated mice. These results provide a preclinical proof-of-concept for simultaneous PD-L1/PD-1 pathways blockade along with CIK infusion as a novel immunotherapy for unresectable cancers.
Collapse
|
41
|
Altered Expression of Natural Cytotoxicity Receptors and NKG2D on Peripheral Blood NK Cell Subsets in Breast Cancer Patients. Transl Oncol 2016; 9:384-391. [PMID: 27641642 PMCID: PMC5024335 DOI: 10.1016/j.tranon.2016.07.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/10/2016] [Accepted: 07/13/2016] [Indexed: 12/13/2022] Open
Abstract
Human natural killer (NK) cells are considered professional cytotoxic cells that are integrated into the effector branch of innate immunity during antiviral and antitumoral responses. The purpose of this study was to examine the peripheral distribution and expression of NK cell activation receptors from the fresh peripheral blood mononuclear cells of 30 breast cancer patients prior to any form of treatment (including surgery, chemotherapy, and radiotherapy), 10 benign breast pathology patients, and 24 control individuals. CD3−CD56dimCD16bright NK cells (CD56dim NK) and CD3−CD56brightCD16dim/− NK cells (CD56bright NK) were identified using flow cytometry. The circulating counts of CD56dim and CD56bright NK cells were not significantly different between the groups evaluated, nor were the counts of other leukocyte subsets between the breast cancer patients and benign breast pathology patients. However, in CD56dim NK cells, NKp44 expression was higher in breast cancer patients (P = .0302), whereas NKp30 (P = .0005), NKp46 (P = .0298), and NKG2D (P = .0005) expression was lower with respect to healthy donors. In CD56bright NK cells, NKp30 (P = .0007), NKp46 (P = .0012), and NKG2D (P = .0069) expression was lower in breast cancer patients compared with control group. Only NKG2D in CD56bright NK cells (P = .0208) and CD56dim NK cells (P = .0439) showed difference between benign breast pathology and breast cancer patients. Collectively, the current study showed phenotypic alterations in activation receptors on CD56dim and CD56bright NK cells, suggesting that breast cancer patients have decreased NK cell cytotoxicity.
Collapse
|
42
|
Chen J, Zhu XX, Xu H, Fang HZ, Zhao JQ. Expression and prognostic significance of unique ULBPs in pancreatic cancer. Onco Targets Ther 2016; 9:5271-9. [PMID: 27621649 PMCID: PMC5010155 DOI: 10.2147/ott.s107771] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background Pancreatic cancer is one of the most lethal cancers worldwide, due to the lack of efficient therapy and difficulty in early diagnosis. ULBPs have been shown to behave as important protectors with prognostic significance in various cancers. Materials and methods Immunohistochemistry and enzyme-linked immunosorbent assays were used to explore the expression of ULBPs in cancer tissue and in serum, while survival analysis was used to evaluate the subsequent clinical value of ULBPs. Results Statistics showed that high expression of membrane ULBP1 was a good biomarker of overall survival (18 months vs 13 months), and a high level of soluble ULBP2 was deemed an independent poor indicator for both overall survival (P<0.001) and disease-free survival (P<0.001). Conclusion ULBP1 provides additional information for early diagnosis, and soluble ULBP2 can be used as a novel tumor marker to evaluate the risk of pancreatic cancer patients.
Collapse
Affiliation(s)
- Jiong Chen
- Department of General Surgery, Anhui Provincial Hospital, Anhui Medical University; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, People's Republic of China
| | - Xing-Xing Zhu
- Department of General Surgery, Anhui Provincial Hospital, Anhui Medical University; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, People's Republic of China
| | - Hong Xu
- Department of General Surgery, Anhui Provincial Hospital, Anhui Medical University; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, People's Republic of China
| | - Heng-Zhong Fang
- Department of General Surgery, Anhui Provincial Hospital, Anhui Medical University; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, People's Republic of China
| | - Jin-Qian Zhao
- Department of General Surgery, Anhui Provincial Hospital, Anhui Medical University; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, People's Republic of China
| |
Collapse
|
43
|
Abstract
Human and mouse NKG2D ligands (NKG2DLs) are absent or only poorly expressed by most normal cells but are upregulated by cell stress, hence, alerting the immune system in case of malignancy or infection. Although these ligands are numerous and highly variable (at genetic, genomic, structural, and biochemical levels), they all belong to the major histocompatibility complex class I gene superfamily and bind to a single, invariant, receptor: NKG2D. NKG2D (CD314) is an activating receptor expressed on NK cells and subsets of T cells that have a key role in the recognition and lysis of infected and tumor cells. Here, we review the molecular diversity of NKG2DLs, discuss the increasing appreciation of their roles in a variety of medical conditions, and propose several explanations for the evolutionary force(s) that seem to drive the multiplicity and diversity of NKG2DLs while maintaining their interaction with a single invariant receptor.
Collapse
Affiliation(s)
- Raphael Carapito
- ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.,Laboratoire International Associé (LIA) INSERM, Strasbourg (France) - Nagano (Japan), Strasbourg, France.,Fédération Hospitalo-Universitaire (FHU) OMICARE, Strasbourg, France
| | - Seiamak Bahram
- ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.,Laboratoire International Associé (LIA) INSERM, Strasbourg (France) - Nagano (Japan), Strasbourg, France.,Fédération Hospitalo-Universitaire (FHU) OMICARE, Strasbourg, France.,Laboratoire Central d'Immunologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
44
|
Mittica G, Capellero S, Genta S, Cagnazzo C, Aglietta M, Sangiolo D, Valabrega G. Adoptive immunotherapy against ovarian cancer. J Ovarian Res 2016; 9:30. [PMID: 27188274 PMCID: PMC4869278 DOI: 10.1186/s13048-016-0236-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 04/21/2016] [Indexed: 01/16/2023] Open
Abstract
The standard front-line therapy for epithelial ovarian cancer (EOC) is combination of debulking surgery and platinum-based chemotherapy. Nevertheless, the majority of patients experience disease recurrence. Although extensive efforts to find new therapeutic options, cancer cells invariably develop drug resistance and disease progression. New therapeutic strategies are needed to improve prognosis of patients with advanced EOC. Recently, several preclinical and clinical studies investigated feasibility and activity of adoptive immunotherapy in EOC. Our aim is to highlight prospective of adoptive immunotherapy in EOC, focusing on HLA-restricted Tumor Infiltrating Lymphocytes (TILs), and MHC-independent immune effectors such as natural killer (NK), and cytokine-induced killer (CIK). Adoptive cell therapy (ACT) has shown activity in several pre-clinical models. Available preclinical and clinical data suggest that adoptive cell therapy may provide the best benefit in settings of low tumor burden, minimal residual disease, or maintenance therapy. Further studies are needed to better define the optimal clinical setting.
Collapse
Affiliation(s)
- Gloria Mittica
- Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | | | - Sofia Genta
- Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | | | - Massimo Aglietta
- Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Dario Sangiolo
- Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Giorgio Valabrega
- Candiolo Cancer Institute-FPO- IRCCS, Candiolo, Turin, Italy. .,Department of Oncology, University of Torino, Turin, Italy. .,Division of Medical Oncology-1, Candiolo Cancer Institute- FPO- IRCCS, Strada Provinciale 142 km 3.95, Candiolo, 10060, Turin, Italy.
| |
Collapse
|
45
|
Tsukagoshi M, Wada S, Yokobori T, Altan B, Ishii N, Watanabe A, Kubo N, Saito F, Araki K, Suzuki H, Hosouchi Y, Kuwano H. Overexpression of natural killer group 2 member D ligands predicts favorable prognosis in cholangiocarcinoma. Cancer Sci 2016; 107:116-22. [PMID: 26608587 PMCID: PMC4768394 DOI: 10.1111/cas.12853] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/17/2015] [Accepted: 11/20/2015] [Indexed: 12/17/2022] Open
Abstract
The natural killer group 2 member D (NKG2D) receptor and its ligands are important mediators of immune responses to tumors. NKG2D ligands are overexpressed in several malignant tumor types; however, the prognostic value of these ligands is unclear. Here, we aimed to elucidate the role of NKG2D ligands in extrahepatic cholangiocarcinoma (EHCC). We therefore investigated the expression of the NKG2D receptor and its ligands MHC class I chain-related proteins A and B (MICA/B), unique long 16 binding protein (ULBP) 1, and ULBP2/5/6 in resected specimens from 82 patients with EHCC. All NKG2D ligands were highly expressed in EHCC. High expression of MICA/B or ULBP2/5/6 correlated with overall and disease-free survival. In contrast, high expression of ULBP1 was significantly associated with improved overall survival, but not disease-free survival. Concurrent high expression of multiple NKG2D ligands revealed significantly better overall and disease-free survival than that observed with the overexpression of any one NKG2D ligand. Co-expression of multiple NKG2D ligands was an independent prognostic indicator of improved survival. Furthermore, co-overexpression of multiple NKG2D ligands was significantly correlated with high expression of the NKG2D receptor. Inhibiting interactions between multiple NKG2D ligands and the NKG2D receptor might be a promising approach for controlling cancer progression and improving patient prognosis in EHCC.
Collapse
Affiliation(s)
- Mariko Tsukagoshi
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Satoshi Wada
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
- Department of Cancer ImmunotherapyKanagawa Cancer CenterYokohamaJapan
| | - Takehiko Yokobori
- Department of Molecular Pharmacology and OncologyGunma University Graduate School of MedicineMaebashiJapan
| | - Bolag Altan
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Norihiro Ishii
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Akira Watanabe
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Norio Kubo
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Fumiyoshi Saito
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Kenichiro Araki
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Hideki Suzuki
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| | - Yasuo Hosouchi
- Department of Surgery and Laparoscopic SurgeryGunma Prefecture Saiseikai‐Maebashi HospitalMaebashiJapan
| | - Hiroyuki Kuwano
- Department of General Surgical ScienceGunma University Graduate School of MedicineMaebashiJapan
| |
Collapse
|
46
|
Ribeiro CH, Kramm K, Gálvez-Jirón F, Pola V, Bustamante M, Contreras HR, Sabag A, Garrido-Tapia M, Hernández CJ, Zúñiga R, Collazo N, Sotelo PH, Morales C, Mercado L, Catalán D, Aguillón JC, Molina MC. Clinical significance of tumor expression of major histocompatibility complex class I-related chains A and B (MICA/B) in gastric cancer patients. Oncol Rep 2015; 35:1309-17. [PMID: 26708143 PMCID: PMC4750752 DOI: 10.3892/or.2015.4510] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 10/31/2015] [Indexed: 12/26/2022] Open
Abstract
Gastric cancer (GC) is the third most common cause of cancer death worldwide. Natural killer cells play an important role in the immune defense against transformed cells. They express the activating receptor NKG2D, whose ligands belong to the MIC and ULBP/RAET family. Although it is well established that these ligands are generally expressed in tumors, the association between their expression in the tumor and gastric mucosa and clinical parameters and prognosis of GC remains to be addressed. In the present study, MICA and MICB expression was analyzed, by flow cytometry, in 23 and 20 pairs of gastric tumor and adjacent non-neoplasic gastric mucosa, respectively. Additionally, ligands expression in 13 tumors and 7 gastric mucosa samples from GC patients were evaluated by immunohistochemistry. The mRNA levels of MICA in 9 pairs of tumor and mucosa were determined by quantitative PCR. Data were associated with the clinicopathological characteristics and the patient outcome. MICA expression was observed in 57% of tumors (13/23) and 44% of mucosal samples (10/23), while MICB was detected in 50% of tumors (10/20) and 45% of mucosal tissues (9/20). At the protein level, ligand expression was significantly higher in the tumor than in the gastric mucosa. MICA mRNA levels were also increased in the tumor as compared to the mucosa. However, clinicopathological analysis indicated that, in patients with tumors >5 cm, the expression of MICA and MICB in the tumor did not differ from that of the mucosa, and tumors >5 cm showed significantly higher MICA and MICB expression than tumors ≤5 cm. Patients presenting tumors >5 cm that expressed MICA and MICB had substantially shorter survival than those with large tumors that did not express these ligands. Our results suggest that locally sustained expression of MICA and MICB in the tumor may contribute to the malignant progression of GC and that expression of these ligands predicts an unfavorable prognosis in GC patients presenting large tumors.
Collapse
Affiliation(s)
- Carolina Hager Ribeiro
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Karina Kramm
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Felipe Gálvez-Jirón
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Víctor Pola
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Marco Bustamante
- Departamento de Cirugía Digestiva, Hospital del Salvador, Universidad de Chile, Santiago, Chile
| | - Hector R Contreras
- Programa de Fisiología y Biofísica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Andrea Sabag
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Macarena Garrido-Tapia
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Carolina J Hernández
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Roberto Zúñiga
- Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Norberto Collazo
- Centro de InmunoBiotecnología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Pablo Hernán Sotelo
- Centro de InmunoBiotecnología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Camila Morales
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Luis Mercado
- Instituto de Biología, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Diego Catalán
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Juan Carlos Aguillón
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - María Carmen Molina
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
47
|
Chen J, Xu H, Zhu XX. Abnormal expression levels of sMICA and NKG2D are correlated with poor prognosis in pancreatic cancer. Ther Clin Risk Manag 2015; 12:11-8. [PMID: 26730197 PMCID: PMC4694694 DOI: 10.2147/tcrm.s96869] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Soluble major histocompatibility complex class I-related chain A molecules (sMICA) and natural-killer group 2 member D (NKG2D) not only correlate with tumorigenesis and progression, but also with tumor invasion and metastasis. In this study, we used immunohistochemistry to investigate the correlation and prognostic significance of the differential expression of sMICA and NKG2D in pancreatic carcinoma and paracarcinoma tissues from 70 patients with pancreatic carcinomas. The results showed that sMICA expression was significantly (P<0.05) higher in tumor tissues (67.1%) than that in adjacent nontumor tissues (31.4%), whereas NKG2D expression was significantly (P<0.001) lower in tumor tissues (32.9%) than that in adjacent nontumor tissues (60.0%). Spearman’s rank correlation test showed a negative correlation between the expression of sMICA and that of NKG2D (r=−0.676, P<0.001). Kaplan–Meier survival analysis showed that a high sMICA expression was significantly correlated with decreased disease-free survival (DFS) (P<0.001) and overall survival (OS) (P<0.001), while a high NKG2D expression was significantly associated with increased DFS (P=0.001) and OS (P=0.001) of the patients. Multivariate analysis showed that a high sMICA expression was an independent predictive factor for poor DFS (P<0.001) and OS (P=0.012); but low NKG2D expression was not an independent prognostic factor for poor DFS (P=0.238) and OS (P=0.574). In conclusion, our findings suggest that the expression levels of sMICA and NKG2D are abnormal and negatively correlated with one another in pancreatic carcinoma tissues; they may be considered as valuable biomarkers for the prognosis of pancreatic carcinoma.
Collapse
Affiliation(s)
- Jiong Chen
- Department of General Surgery, Affiliated Provincial Hospital, Anhui Medical University, Hefei, People's Republic of China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, People's Republic of China
| | - Hong Xu
- Department of General Surgery, Affiliated Provincial Hospital, Anhui Medical University, Hefei, People's Republic of China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, People's Republic of China
| | - Xing-Xing Zhu
- Department of General Surgery, Affiliated Provincial Hospital, Anhui Medical University, Hefei, People's Republic of China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, People's Republic of China
| |
Collapse
|
48
|
Cox ST, Pearson H, Laza-Briviesca R, Pesoa S, Vullo C, Madrigal JA, Saudemont A. Characterization of 5' promoter and exon 1-3 polymorphism of the RAET1E gene. Hum Immunol 2015; 77:96-103. [PMID: 26519211 DOI: 10.1016/j.humimm.2015.10.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 10/26/2015] [Accepted: 10/26/2015] [Indexed: 11/15/2022]
Abstract
NKG2D is an activating receptor utilized by natural killer (NK) cells that recognizes upregulated ligands on infected, tumorigenic and damaged cells, leading to their cytolysis. However, the NKG2D ligand (NKG2DL) system is very complex with eight known gene loci encoding slightly different molecules. Furthermore, most NKG2DL gene loci such as MICA and MICB are highly polymorphic with potential for functional differences. NKG2DL expression on tumors varies depending on the malignancy and tumors can also release soluble NKG2DL that exert anergic effects on NK cells when engagement with NKG2D occurs, allowing escape from NK cell immunosurveillance. We carried out RAET1E typing of IHW cell line DNA, including a 580 bp proximal promoter fragment and exons 1-3 identifying 13 of 15 known RAET1E alleles. We determined 7 polymorphisms within the promoter region, including 2 already known that contributed to 9 promoter types. RAET1E alleles with variability in the extracellular region also differed with respect to promoter type and one allele, RAET1E(∗)003, associated with 5 promoter types. We then identified putative transcription factor binding sites for RAET1E, and found 5 of the 7 promoter polymorphisms may disrupt these sites, abrogating binding of transcription factors and varying the potential level of expression.
Collapse
Affiliation(s)
- Steven T Cox
- Anthony Nolan Research Institute, Royal Free Hospital, Hampstead, London NW3 2QU, UK.
| | - Hayley Pearson
- Anthony Nolan Research Institute, Royal Free Hospital, Hampstead, London NW3 2QU, UK
| | - Raquel Laza-Briviesca
- Anthony Nolan Research Institute, Royal Free Hospital, Hampstead, London NW3 2QU, UK
| | - Susanna Pesoa
- HLA Laboratory, Hospital Nacional de Clinicas, Cordoba, Argentina
| | - Carlos Vullo
- HLA Laboratory, Hospital Nacional de Clinicas, Cordoba, Argentina
| | - J Alejandro Madrigal
- Anthony Nolan Research Institute, Royal Free Hospital, Hampstead, London NW3 2QU, UK; UCL Cancer Institute, Royal Free Campus, London NW3 2QG, UK
| | - Aurore Saudemont
- Anthony Nolan Research Institute, Royal Free Hospital, Hampstead, London NW3 2QU, UK; UCL Cancer Institute, Royal Free Campus, London NW3 2QG, UK
| |
Collapse
|
49
|
Zhou C, Ji J, Cai Q, Shi M, Chen X, Yu Y, Zhu Z, Zhang J. MTA2 enhances colony formation and tumor growth of gastric cancer cells through IL-11. BMC Cancer 2015; 15:343. [PMID: 25929737 PMCID: PMC4419442 DOI: 10.1186/s12885-015-1366-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 04/24/2015] [Indexed: 01/09/2023] Open
Abstract
Background We have preliminarily reported MTA2 expression in gastric cancer and its biological functions by using knockdown cell models, while the molecular mechanisms of MTA2 in regulating malignant behaviors are still unclear. Methods MTA2 overexpression models were established by transfection assay in gastric cancer cells BGC-823 and MKN28. Cell proliferation assay, colony formation in soft agar, wound-healing assay and transwell migration assay were performed with MTA2 overexpression and negative control (NC) cells. Subcutaneous xenografts and pulmonary metastasis models by BGC-823/MTA2 and BGC-823/NC cells were used to observe the capacity of growth and metastasis in vivo. Differential gene expression in MTA2 knockdown and overexpression cells was analyzed by microarrays. IL-11, which demonstrated as differential expression in microarray, was detected by real-time PCR, western blot, ELISA and immunohistochemistry staining. Recombinant human IL-11 (rhIL-11) was administrated in cell proliferation and colony formation as rescue assay. Results The numbers of colonies in soft agar were significantly more in BGC-823/MTA2 and MKN28/MTA2 cells, comparing with those in their NC cells. Capabilities of cell proliferation, wound-healing and cell migration were not significantly changed in MTA2 overexpression cells. The sizes of subcutaneous xenografts and pulmonary metastases of BGC-832/MTA2 cells were significantly larger than those in BGC-823/NC group. Differential expression of IL-11 was identified by genome expression microarray both in MTA2 knockdown and overexpression cells. IL-11 expression was elevated in BGC-823/MTA2 cells, whereas reduced in SGC-7901/shMTA2 cells. Administration of rhIL-11 recovered colony formation capacity of SGC-7901/shMTA2 cells. Conclusions MTA2 overexpression enhances colony formation and tumor growth of gastric cancer cells, but not plays important role in cancer cell migration and metastasis. IL-11 is one of the downstream effectors of MTA2 in regulating gastric cancer cells growth. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1366-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chenfei Zhou
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China.
| | - Jun Ji
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China.
| | - Qu Cai
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China.
| | - Min Shi
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China.
| | - Xuehua Chen
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China.
| | - Yingyan Yu
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China.
| | - Zhenggang Zhu
- Department of Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China. .,Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China.
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China.
| |
Collapse
|
50
|
Zhang J, Basher F, Wu JD. NKG2D Ligands in Tumor Immunity: Two Sides of a Coin. Front Immunol 2015; 6:97. [PMID: 25788898 PMCID: PMC4349182 DOI: 10.3389/fimmu.2015.00097] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/18/2015] [Indexed: 11/13/2022] Open
Abstract
The activating/co-stimulatory receptor NKG2D (natural-killer group 2, member D) is expressed on the surface of all human NK, NKT, CD8(+) T, and subsets of γδ(+) T cells. The significance of NKG2D function in tumor immunity has been well demonstrated in experimental animal models. However, the role of human NKG2D ligands in regulating tumor immunity and cancer prognosis had been controversial in the literature. In this review, we summarize the latest advancement, discuss the controversies, and present evidence that membrane-bound and soluble NKG2D ligands oppositely regulate tumor immunity. We also discuss new perspectives of targeting NKG2D ligands for cancer immunotherapy.
Collapse
Affiliation(s)
- Jinyu Zhang
- Department of Microbiology and Immunology, Medical University of South Carolina , Charleston, SC , USA
| | - Fahmin Basher
- Department of Microbiology and Immunology, Medical University of South Carolina , Charleston, SC , USA
| | - Jennifer D Wu
- Department of Microbiology and Immunology, Medical University of South Carolina , Charleston, SC , USA ; Cancer Immunology Program, Hollings Cancer Center , Charleston, SC , USA
| |
Collapse
|