51
|
Kiefer A, Prüfer M, Röder J, Pfeifer Serrahima J, Bodden M, Kühnel I, Oberoi P, Wels WS. Dual Targeting of Glioblastoma Cells with Bispecific Killer Cell Engagers Directed to EGFR and ErbB2 (HER2) Facilitates Effective Elimination by NKG2D-CAR-Engineered NK Cells. Cells 2024; 13:246. [PMID: 38334638 PMCID: PMC10854564 DOI: 10.3390/cells13030246] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/17/2024] [Accepted: 01/24/2024] [Indexed: 02/10/2024] Open
Abstract
NKG2D is an activating receptor of natural killer cells that recognizes stress-induced ligands (NKG2DL) expressed by many tumor cells. Nevertheless, NKG2DL downregulation or shedding can still allow cancer cells to evade immune surveillance. Here, we used lentiviral gene transfer to engineer clinically usable NK-92 cells with a chimeric antigen receptor (NKAR) which contains the extracellular domain of NKG2D for target recognition, or an NKAR, together with the IL-15 superagonist RD-IL15, and combined these effector cells with recombinant NKG2D-interacting bispecific engagers that simultaneously recognize the tumor-associated antigens epidermal growth factor receptor (EGFR) or ErbB2 (HER2). Applied individually, in in vitro cell-killing assays, these NKAB-EGFR and NKAB-ErbB2 antibodies specifically redirected NKAR-NK-92 and NKAR_RD-IL15-NK-92 cells to glioblastoma and other cancer cells with elevated EGFR or ErbB2 levels. However, in mixed glioblastoma cell cultures, used as a model for heterogeneous target antigen expression, NKAR-NK cells only lysed the EGFR- or ErbB2-expressing subpopulations in the presence of one of the NKAB molecules. This was circumvented by applying NKAB-EGFR and NKAB-ErbB2 together, resulting in effective antitumor activity similar to that against glioblastoma cells expressing both target antigens. Our results demonstrate that combining NK cells carrying an activating NKAR receptor with bispecific NKAB antibodies allows for flexible targeting, which can enhance tumor-antigen-specific cytotoxicity and prevent immune escape.
Collapse
Affiliation(s)
- Anne Kiefer
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, 60590 Frankfurt, Germany
| | - Maren Prüfer
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany
| | - Jasmin Röder
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, 60590 Frankfurt, Germany
| | - Jordi Pfeifer Serrahima
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, 60590 Frankfurt, Germany
| | - Malena Bodden
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany
| | - Ines Kühnel
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany
| | - Pranav Oberoi
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany
| | - Winfried S. Wels
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, 60590 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, a Partnership between DKFZ and University Hospital Frankfurt, 60590 Frankfurt, Germany
| |
Collapse
|
52
|
Wang W, Liu Y, He Z, Li L, Liu S, Jiang M, Zhao B, Deng M, Wang W, Mi X, Sun Z, Ge X. Breakthrough of solid tumor treatment: CAR-NK immunotherapy. Cell Death Discov 2024; 10:40. [PMID: 38245520 PMCID: PMC10799930 DOI: 10.1038/s41420-024-01815-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 01/22/2024] Open
Abstract
As the latest and most anticipated method of tumor immunotherapy, CAR-NK therapy has received increasing attention in recent years, and its safety and high efficiency have irreplaceable advantages over CAR-T. Current research focuses on the application of CAR-NK in hematological tumors, while there are fewer studies on solid tumor. This article reviews the process of constructing CAR-NK, the effects of hypoxia and metabolic factors, NK cell surface receptors, cytokines, and exosomes on the efficacy of CAR-NK in solid tumor, and the role of CAR-NK in various solid tumor. The mechanism of action and the research status of the potential of CAR-NK in the treatment of solid tumor in clinical practice, and put forward the advantages, limitations and future problems of CAR-NK in the treatment of solid tumor.
Collapse
Affiliation(s)
- Wenkang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Yang Liu
- Department of Radiotherapy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Zhen He
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Lifeng Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Senbo Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mingqiang Jiang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bing Zhao
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meng Deng
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wendong Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuefang Mi
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Xin Ge
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
53
|
Aloi N, Drago G, Ruggieri S, Cibella F, Colombo P, Longo V. Extracellular Vesicles and Immunity: At the Crossroads of Cell Communication. Int J Mol Sci 2024; 25:1205. [PMID: 38256278 PMCID: PMC10816988 DOI: 10.3390/ijms25021205] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 01/24/2024] Open
Abstract
Extracellular vesicles (EVs), comprising exosomes and microvesicles, are small membranous structures secreted by nearly all cell types. They have emerged as crucial mediators in intercellular communication, playing pivotal roles in diverse physiological and pathological processes, notably within the realm of immunity. These roles go beyond mere cellular interactions, as extracellular vesicles stand as versatile and dynamic components of immune regulation, impacting both innate and adaptive immunity. Their multifaceted involvement includes immune cell activation, antigen presentation, and immunomodulation, emphasising their significance in maintaining immune homeostasis and contributing to the pathogenesis of immune-related disorders. Extracellular vesicles participate in immunomodulation by delivering a wide array of bioactive molecules, including proteins, lipids, and nucleic acids, thereby influencing gene expression in target cells. This manuscript presents a comprehensive review that encompasses in vitro and in vivo studies aimed at elucidating the mechanisms through which EVs modulate human immunity. Understanding the intricate interplay between extracellular vesicles and immunity is imperative for unveiling novel therapeutic targets and diagnostic tools applicable to various immunological disorders, including autoimmune diseases, infectious diseases, and cancer. Furthermore, recognising the potential of EVs as versatile drug delivery vehicles holds significant promise for the future of immunotherapies.
Collapse
Affiliation(s)
| | | | | | | | - Paolo Colombo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Via Ugo La Malfa 153, 90146 Palermo, Italy; (N.A.); (G.D.); (S.R.); (F.C.); (V.L.)
| | | |
Collapse
|
54
|
Wu Q, Li X, Yang Y, Huang J, Yao M, Li J, Huang Y, Cai X, Geller DA, Yan Y. MICA+ Tumor Cell Upregulated Macrophage-Secreted MMP9 via PROS1-AXL Axis to Induce Tumor Immune Escape in Advanced Hepatocellular Carcinoma (HCC). Cancers (Basel) 2024; 16:269. [PMID: 38254761 PMCID: PMC10813556 DOI: 10.3390/cancers16020269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/21/2023] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND tumor-associated macrophages (TAMs) constitute a significant proportion of non-cancerous cells within the intricate tumor microenvironment (TME) of hepatocellular carcinoma (HCC). Understanding the communication between macrophages and tumor cells, as well as investigating potential signaling pathways, holds promise for enhancing therapeutic responses in HCC. METHODS single-cell RNA-sequencing data and bulk RNA-sequencing data were derived from open source databases Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA). Through this analysis, we elucidated the interactions between MICA+ tumor cells and MMP9+ macrophages, primarily mediated via the PROS1-AXL axis in advanced HCC. Subsequently, we employed a range of experimental techniques including lentivirus infection, recombinant protein stimulation, and AXL inhibition experiments to validate these interactions and unravel the underlying mechanisms. RESULTS we presented a single-cell atlas of advanced HCC, highlighting the expression patterns of MICA and MMP9 in tumor cells and macrophages, respectively. Activation of the interferon gamma (IFN-γ) signaling pathway was observed in MICA+ tumor cells and MMP9+ macrophages. We identified the existence of an interaction between MICA+ tumor cells and MMP9+ macrophages mediated via the PROS1-AXL axis. Additionally, we found MMP9+ macrophages had a positive correlation with M2-like macrophages. Subsequently, experiments validated that DNA damage not only induced MICA expression in tumor cells via IRF1, but also upregulated PROS1 levels in HCC cells, stimulating macrophages to secrete MMP9. Consequently, MMP9 led to the proteolysis of MICA. CONCLUSION MICA+ HCC cells secreted PROS1, which upregulated MMP9 expression in macrophages through AXL receptors. The increased MMP9 activity resulted in the proteolytic shedding of MICA, leading to the release of soluble MICA (sMICA) and the subsequent facilitation of tumor immune escape.
Collapse
Affiliation(s)
- Qiulin Wu
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China; (Q.W.); (X.L.); (Y.Y.); (J.H.); (M.Y.); (J.L.); (Y.H.); (X.C.)
| | - Xicai Li
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China; (Q.W.); (X.L.); (Y.Y.); (J.H.); (M.Y.); (J.L.); (Y.H.); (X.C.)
| | - Yan Yang
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China; (Q.W.); (X.L.); (Y.Y.); (J.H.); (M.Y.); (J.L.); (Y.H.); (X.C.)
| | - Jingquan Huang
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China; (Q.W.); (X.L.); (Y.Y.); (J.H.); (M.Y.); (J.L.); (Y.H.); (X.C.)
| | - Ming Yao
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China; (Q.W.); (X.L.); (Y.Y.); (J.H.); (M.Y.); (J.L.); (Y.H.); (X.C.)
| | - Jianjun Li
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China; (Q.W.); (X.L.); (Y.Y.); (J.H.); (M.Y.); (J.L.); (Y.H.); (X.C.)
| | - Yubin Huang
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China; (Q.W.); (X.L.); (Y.Y.); (J.H.); (M.Y.); (J.L.); (Y.H.); (X.C.)
| | - Xiaoyong Cai
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China; (Q.W.); (X.L.); (Y.Y.); (J.H.); (M.Y.); (J.L.); (Y.H.); (X.C.)
| | - David A. Geller
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15260, USA
| | - Yihe Yan
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China; (Q.W.); (X.L.); (Y.Y.); (J.H.); (M.Y.); (J.L.); (Y.H.); (X.C.)
| |
Collapse
|
55
|
Kim S, Chung H, Kwak JE, Kim YR, Park CH, Kim Y, Cheong JW, Wu J, Shin EC, Cho H, Kim JS. Clearing soluble MIC reverses the impaired function of natural killer cells from patients with multiple myeloma. J Immunother Cancer 2024; 12:e007886. [PMID: 38191242 PMCID: PMC10806558 DOI: 10.1136/jitc-2023-007886] [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] [Accepted: 11/28/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Major histocompatibility complex (MHC) class I chain-related protein (MIC) is a stress-induced ligand released from multiple myeloma (MM) cells during progression, and soluble MIC impairs natural killer group 2D (NKG2D) activating receptor-mediated recognition and function of natural killer (NK) cells. However, whether clearing soluble MIC with a monoclonal antibody (mAb) can restore NK cell activity of MM patients remains undetermined. METHODS We analyzed The Cancer Genome Atlas (TCGA) Multiple Myeloma Research Foundation (MMRF) CoMMpass data set to examine the prognostic significance of MIC expression in MM. We examined the level of soluble MIC in paired peripheral blood (PB) and bone marrow (BM) plasma of patients with MM at diagnosis by ELISA. We evaluated the correlation between the level of soluble MIC and immunophenotype of NK cells from MM patients by multicolor flow cytometry. We also generated MIC-overexpressing MM cell line and characterized the cytotoxic function of patient NK cells in the presence of soluble MIC, and examined the impact of clearing soluble MIC with a humanized mAb (huB10G5). RESULTS We characterize the importance of MICA in MM by revealing the significantly better overall survival of patients with high MICA expression from TCGA MMRF CoMMpass data set. The level of soluble MICA is more highly elevated in MM than in precursor stages, and the concentration of soluble MICA is higher in BM plasma than in PB. The concentration of soluble MICA in BM was correlated with myeloma burden, while it was negatively correlated with the frequency of NKG2D+ NK cells in diagnostic BM aspirates of MM patients. Soluble MICA downregulated NKG2D expression and decreased cytotoxicity of MM patient NK cells ex vivo, which were reversed by a humanized soluble MIC-clearing mAb (huB10G5) with enhanced degranulation of NK cells. CONCLUSIONS Our findings indicate targeting soluble MIC with huB10G5 might be a viable therapeutic approach to promote NKG2D-dependent cellular immunotherapy outcome in MM.
Collapse
Affiliation(s)
- Sojeong Kim
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Haerim Chung
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Jeong-Eun Kwak
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Yu Ri Kim
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Chung Hyun Park
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Yeonhee Kim
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - June-Won Cheong
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Jennifer Wu
- Department of Urology and Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea (the Republic of)
| | - Hyunsoo Cho
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| | - Jin Seok Kim
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea (the Republic of)
| |
Collapse
|
56
|
Lyu DW. Immunomodulatory effects of exercise in cancer prevention and adjuvant therapy: a narrative review. Front Physiol 2024; 14:1292580. [PMID: 38239881 PMCID: PMC10794543 DOI: 10.3389/fphys.2023.1292580] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024] Open
Abstract
Successful application of cancer immunotherapy has rekindled hope in cancer patients. However, a number of patients are unresponsive to immunotherapy and related treatments. This unresponsiveness in cancer patients toward different treatment regimens can be mainly attributed to severe immune dysfunction in such patients. Several reports indicate that physical exercise can significantly lead to improved cancer patient outcomes. Since exercise gets immense response from the immune system, it can be utilized to improve immune function. Leukocytes with enhanced functions are substantially mobilized into the circulation by a single bout of intense physical exercise. Chronic physical exercise results in greater muscle endurance and strength and improved cardiorespiratory function. This exercise regime is also useful in improving T-cell abundance and reducing dysfunctional T cells. The current available data strongly justify for future clinical trials to investigate physical exercise use as an adjuvant in cancer therapy; however, optimal parameters using exercise for a defined outcome are yet to be established. The components of the immune system associate with almost every tumorigenesis step. The inter-relationship between inflammation, cancer, and innate immunity has recently gained acceptance; however, the underlying cellular and molecular mechanisms behind this relationship are yet to be solved. Several studies suggest physical exercise-mediated induction of immune cells to elicit anti-tumorigenic effects. This indicates the potential of exercising in modulating the behavior of immune cells to inhibit tumor progression. However, further mechanistic details behind physical exercise-driven immunomodulation and anticancer effects have to be determined. This review aims to summarize and discuss the association between physical exercise and immune function modulation and the potential of exercise as an adjuvant therapy in cancer prevention and treatment.
Collapse
Affiliation(s)
- Da-wei Lyu
- Physical Education and Health School, East China Jiaotong University, Nanchang, Jiangxi, China
| |
Collapse
|
57
|
Aguilar OA, Qualls AE, Gonzalez-Hinojosa MDR, Obeidalla S, Kerchberger VE, Tsao T, Singer JP, Looney MR, Raymond W, Hays SR, Golden JA, Kukreja J, Shaver CM, Ware LB, Christie J, Diamond JM, Lanier LL, Greenland JR, Calabrese DR. MICB Genomic Variant Is Associated with NKG2D-mediated Acute Lung Injury and Death. Am J Respir Crit Care Med 2024; 209:70-82. [PMID: 37878820 PMCID: PMC10870895 DOI: 10.1164/rccm.202303-0472oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023] Open
Abstract
Rationale: Acute lung injury (ALI) carries a high risk of mortality but has no established pharmacologic therapy. We previously found that experimental ALI occurs through natural killer (NK) cell NKG2D receptor activation and that the cognate human ligand, MICB, was associated with ALI after transplantation. Objectives: To investigate the association of a common missense variant, MICBG406A, with ALI. Methods: We assessed MICBG406A genotypes within two multicenter observational study cohorts at risk for ALI: primary graft dysfunction (N = 619) and acute respiratory distress syndrome (N = 1,376). Variant protein functional effects were determined in cultured and ex vivo human samples. Measurements and Main Results: Recipients of MICBG406A-homozygous allografts had an 11.1% absolute risk reduction (95% confidence interval [CI], 3.2-19.4%) for severe primary graft dysfunction after lung transplantation and reduced risk for allograft failure (hazard ratio, 0.36; 95% CI, 0.13-0.98). In participants with sepsis, we observed 39% reduced odds of moderately or severely impaired oxygenation among MICBG406A-homozygous individuals (95% CI, 0.43-0.86). BAL NK cells were less frequent and less mature in participants with MICBG406A. Expression of missense variant protein MICBD136N in cultured cells resulted in reduced surface MICB and reduced NKG2D ligation relative to wild-type MICB. Coculture of variant MICBD136N cells with NK cells resulted in less NKG2D activation and less susceptibility to NK cell killing relative to the wild-type cells. Conclusions: These data support a role for MICB signaling through the NKG2D receptor in mediating ALI, suggesting a novel therapeutic approach.
Collapse
Affiliation(s)
- Oscar A. Aguilar
- Department Microbiology and Immunology
- Parker Institute for Cancer Immunotherapy
| | | | | | | | | | | | | | | | | | | | | | - Jasleen Kukreja
- Department of Surgery, University of California San Francisco, San Francisco, California
| | | | - Lorraine B. Ware
- Department Medicine and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jason Christie
- Department Medicine and
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | | | - Lewis L. Lanier
- Department Microbiology and Immunology
- Parker Institute for Cancer Immunotherapy
| | - John R. Greenland
- Department Medicine
- San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Daniel R. Calabrese
- Department Medicine
- San Francisco Veterans Affairs Medical Center, San Francisco, California
| |
Collapse
|
58
|
Verbist K, Nichols KE. Cytokine Storm Syndromes Associated with Epstein-Barr Virus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1448:227-248. [PMID: 39117818 DOI: 10.1007/978-3-031-59815-9_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Epstein-Barr virus (EBV) is a ubiquitous and predominantly B cell tropic virus. One of the most common viruses to infect humans, EBV, is best known as the causative agent of infectious mononucleosis (IM). Although most people experience asymptomatic infection, EBV is a potent immune stimulus and as such it elicits robust proliferation and activation of the B-lymphocytes it infects as well as the immune cells that respond to infection. In certain individuals, such as those with inherited or acquired defects affecting the immune system, failure to properly control EBV leads to the accumulation of EBV-infected B cells and EBV-reactive immune cells, which together contribute to the development of often life-threatening cytokine storm syndromes (CSS). Here, we review the normal immune response to EBV and discuss several CSS associated with EBV, such as chronic active EBV infection, hemophagocytic lymphohistiocytosis, and post-transplant lymphoproliferative disorder. Given the critical role for cytokines in driving inflammation and contributing to disease pathogenesis, we also discuss how targeting specific cytokines provides a rational and potentially less toxic treatment for EBV-driven CSS.
Collapse
Affiliation(s)
- Katherine Verbist
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kim E Nichols
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
59
|
Laureano RS, Vanmeerbeek I, Sprooten J, Govaerts J, Naulaerts S, Garg AD. The cell stress and immunity cycle in cancer: Toward next generation of cancer immunotherapy. Immunol Rev 2024; 321:71-93. [PMID: 37937803 DOI: 10.1111/imr.13287] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/05/2023] [Accepted: 10/20/2023] [Indexed: 11/09/2023]
Abstract
The cellular stress and immunity cycle is a cornerstone of organismal homeostasis. Stress activates intracellular and intercellular communications within a tissue or organ to initiate adaptive responses aiming to resolve the origin of this stress. If such local measures are unable to ameliorate this stress, then intercellular communications expand toward immune activation with the aim of recruiting immune cells to effectively resolve the situation while executing tissue repair to ameliorate any damage and facilitate homeostasis. This cellular stress-immunity cycle is severely dysregulated in diseased contexts like cancer. On one hand, cancer cells dysregulate the normal cellular stress responses to reorient them toward upholding growth at all costs, even at the expense of organismal integrity and homeostasis. On the other hand, the tumors severely dysregulate or inhibit various components of organismal immunity, for example, by facilitating immunosuppressive tumor landscape, lowering antigenicity, and increasing T-cell dysfunction. In this review we aim to comprehensively discuss the basis behind tumoral dysregulation of cellular stress-immunity cycle. We also offer insights into current understanding of the regulators and deregulators of this cycle and how they can be targeted for conceptualizing successful cancer immunotherapy regimen.
Collapse
Affiliation(s)
- Raquel S Laureano
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Isaure Vanmeerbeek
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jenny Sprooten
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jannes Govaerts
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Stefan Naulaerts
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Cell Stress & Immunity (CSI) Lab, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
60
|
He Y, Ma P, Luo Y, Gong X, Gao J, Sun Y, Chen P, Zhang S, Tian Y, Shi B, Zhang B. Novel Association of KLRC4-KLRK1 Gene Polymorphisms with Susceptibility and Progression of Antithyroid Drug-Induced Agranulocytosis. Exp Clin Endocrinol Diabetes 2024; 132:17-22. [PMID: 38237612 DOI: 10.1055/a-2206-2242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
OBJECTIVE Antithyroid drug (ATD)-induced agranulocytosis (TIA) is the most serious adverse effect during ATD treatment of Graves' disease (GD). Previously, the MICA gene was reported to be associated with TIA. MICA protein is an important ligand for the NKG2D protein, which is encoded by the KLRK1 gene and KLRC4-KLRK1 read-through transcription. This study further investigated the association between KLRC4-KLRK1 gene polymorphisms and susceptibility to TIA. METHODS Twenty-eight candidate single nucleotide polymorphisms (SNPs) on KLRC4-KLRK1 read-through transcription were evaluated by the iPLEX MassARRAY system in 209 GD control patients and 38 TIA cases. RESULTS A significant association of rs2734565 polymorphism with TIA was found (p=0.02, OR=1.80, 95% CI=1.09-2.96). The haplotype C-A-A-C-G, including rs2734565-C, was associated with a significantly higher risk of TIA (p=4.79E-09, OR=8.361, 95% CI=3.737-18.707). In addition, the interval time from hyperthyroidism to agranulocytosis onset was shorter in patients carrying the rs2734565-C allele than in non-carrying groups (45.00 (14.00-6570.00) d vs. 1080.00 (30.00-3600.00) d, p=0.046), and the interval from ATD treatment to agranulocytosis onset was also shorter in patients carrying rs2734565-C allele (29.00 (13.00-75.00) d vs. 57.50 (21.00-240.00) d, p=0.023). CONCLUSIONS The findings suggest that the KLRC4-KLRK1 gene polymorphism is associated with susceptibility and progression of ATD-induced agranulocytosis. Patients carrying the rs2734565-C allele had a higher susceptibility and faster onset time of TIA.
Collapse
Affiliation(s)
- Yayi He
- Department of Endocrinology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Pan Ma
- College of Medicine & Forensic, Health Science Center, Xi'an Jiaotong University, Xi'an, China
- Kunming Center for Medical Service Quality Assessment, Kunming, China
| | - Yuanlin Luo
- Department of Endocrinology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaojuan Gong
- College of Medicine & Forensic, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Jiayang Gao
- College of Medicine & Forensic, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Yuxin Sun
- College of Medicine & Forensic, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Pu Chen
- Department of Endocrinology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Suliang Zhang
- College of Medicine & Forensic, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Yuxin Tian
- College of Medicine & Forensic, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Bingyin Shi
- Department of Endocrinology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Bao Zhang
- College of Medicine & Forensic, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
61
|
Canichella M, Molica M, Mazzone C, de Fabritiis P. Chimeric Antigen Receptor T-Cell Therapy in Acute Myeloid Leukemia: State of the Art and Recent Advances. Cancers (Basel) 2023; 16:42. [PMID: 38201469 PMCID: PMC10777995 DOI: 10.3390/cancers16010042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/07/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Chimeric antigen receptors (CAR)-T-cell therapy represents the most important innovation in onco-hematology in recent years. The progress achieved in the management of complications and the latest generations of CAR-T-cells have made it possible to anticipate in second-line the indication of this type of treatment in large B-cell lymphoma. While some types of B-cell lymphomas and B-cell acute lymphoid leukemia have shown extremely promising results, the same cannot be said for myeloid leukemias-in particular, acute myeloid leukemia (AML), which would require innovative therapies more than any other blood disease. The heterogeneities of AML cells and the immunological complexity of the interactions between the bone marrow microenvironment and leukemia cells have been found to be major obstacles to the clinical development of CAR-T in AML. In this review, we report on the main results obtained in AML clinical trials, the preclinical studies testing potential CAR-T constructs, and future perspectives.
Collapse
Affiliation(s)
- Martina Canichella
- Hematology, St. Eugenio Hospital, ASL Roma2, 00144 Rome, Italy; (C.M.); (P.d.F.)
| | - Matteo Molica
- Department of Hematology-Oncology, Azienda Ospedaliera Pugliese-Ciaccio, 88100 Catanzaro, Italy;
| | - Carla Mazzone
- Hematology, St. Eugenio Hospital, ASL Roma2, 00144 Rome, Italy; (C.M.); (P.d.F.)
| | - Paolo de Fabritiis
- Hematology, St. Eugenio Hospital, ASL Roma2, 00144 Rome, Italy; (C.M.); (P.d.F.)
- Department of Biomedicina e Prevenzione, Tor Vergata University, 00133 Rome, Italy
| |
Collapse
|
62
|
Sadrolvaezin A, Pezhman A, Zare I, Nasab SZ, Chamani S, Naghizadeh A, Mostafavi E. Systemic allergic contact dermatitis to palladium, platinum, and titanium: mechanisms, clinical manifestations, prevalence, and therapeutic approaches. MedComm (Beijing) 2023; 4:e386. [PMID: 37873514 PMCID: PMC10590457 DOI: 10.1002/mco2.386] [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: 11/10/2022] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 10/25/2023] Open
Abstract
Contact dermatitis (CD) is an inflammatory skin disease of eczema that is elicited by chemicals or metal ions that have toxic effects without eliciting a T-cell response (contact elicitation) or by small reactive chemicals that modify proteins and induce innate and adaptive immune responses (contact allergens). The clinical condition is characterized by localized skin rash, pruritus, redness, swelling, and lesions, which are mainly detected by patch tests and lymphocyte stimulation. Heavy metals such as palladium (Pd), platinum (Pt), and titanium (Ti) are ubiquitous in our environment. These heavy metals have shown CD effects as allergic agents. Immunological responses result from the interaction of cytokines and T cells. Occupational metal CD accounts for most cases of work-related cutaneous disorders. In this systematic review, the allergic effects of heavy metals, including Pd, Pt, and Ti, and the mechanisms, clinical manifestations, prevalence, and therapeutic approaches are discussed in detail. Furthermore, the therapeutic approaches introduced to treat CD, including corticosteroids, topical calcineurin inhibitors, systemic immunosuppressive agents, phototherapy, and antihistamines, can be effective in the treatment of these diseases in the future. Ultimately, the insights identified could lead to improved therapeutic and diagnostic pathways.
Collapse
Affiliation(s)
- Ali Sadrolvaezin
- Medical Toxicology and Drug Abuse Research CenterBirjand University of Medical SciencesBirjandIran
| | - Arezou Pezhman
- School of MedicineZahedan Azad University of Medical SciencesZahedanIran
| | - Iman Zare
- Research and Development DepartmentSina Medical Biochemistry Technologies Co. Ltd.ShirazIran
| | - Shima Zahed Nasab
- Department of Life Science EngineeringFaculty of New Sciences and TechnologiesUniversity of TehranTehranIran
| | - Sajad Chamani
- Medical Toxicology and Drug Abuse Research CenterBirjand University of Medical SciencesBirjandIran
| | - Ali Naghizadeh
- Medical Toxicology and Drug Abuse Research CenterBirjand University of Medical SciencesBirjandIran
| | - Ebrahim Mostafavi
- Stanford Cardiovascular InstituteStanford University School of MedicineStanfordCaliforniaUSA
- Department of MedicineStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
63
|
Chen C, Jung A, Yang A, Monroy I, Zhang Z, Chaurasiya S, Deshpande S, Priceman S, Fong Y, Park AK, Woo Y. Chimeric Antigen Receptor-T Cell and Oncolytic Viral Therapies for Gastric Cancer and Peritoneal Carcinomatosis of Gastric Origin: Path to Improving Combination Strategies. Cancers (Basel) 2023; 15:5661. [PMID: 38067366 PMCID: PMC10705752 DOI: 10.3390/cancers15235661] [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: 10/26/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 02/12/2024] Open
Abstract
Precision immune oncology capitalizes on identifying and targeting tumor-specific antigens to enhance anti-tumor immunity and improve the treatment outcomes of solid tumors. Gastric cancer (GC) is a molecularly heterogeneous disease where monoclonal antibodies against human epidermal growth factor receptor 2 (HER2), vascular endothelial growth factor (VEGF), and programmed cell death 1 (PD-1) combined with systemic chemotherapy have improved survival in patients with unresectable or metastatic GC. However, intratumoral molecular heterogeneity, variable molecular target expression, and loss of target expression have limited antibody use and the durability of response. Often immunogenically "cold" and diffusely spread throughout the peritoneum, GC peritoneal carcinomatosis (PC) is a particularly challenging, treatment-refractory entity for current systemic strategies. More adaptable immunotherapeutic approaches, such as oncolytic viruses (OVs) and chimeric antigen receptor (CAR) T cells, have emerged as promising GC and GCPC treatments that circumvent these challenges. In this study, we provide an up-to-date review of the pre-clinical and clinical efficacy of CAR T cell therapy for key primary antigen targets and provide a translational overview of the types, modifications, and mechanisms for OVs used against GC and GCPC. Finally, we present a novel, summary-based discussion on the potential synergistic interplay between OVs and CAR T cells to treat GCPC.
Collapse
Affiliation(s)
- Courtney Chen
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Audrey Jung
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Annie Yang
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Isabel Monroy
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA; (I.M.); (S.P.)
| | - Zhifang Zhang
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Shyambabu Chaurasiya
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Supriya Deshpande
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Saul Priceman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA; (I.M.); (S.P.)
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Yuman Fong
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Anthony K. Park
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA; (I.M.); (S.P.)
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Yanghee Woo
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
64
|
Yi M, Li T, Niu M, Mei Q, Zhao B, Chu Q, Dai Z, Wu K. Exploiting innate immunity for cancer immunotherapy. Mol Cancer 2023; 22:187. [PMID: 38008741 PMCID: PMC10680233 DOI: 10.1186/s12943-023-01885-w] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/23/2023] [Indexed: 11/28/2023] Open
Abstract
Immunotherapies have revolutionized the treatment paradigms of various types of cancers. However, most of these immunomodulatory strategies focus on harnessing adaptive immunity, mainly by inhibiting immunosuppressive signaling with immune checkpoint blockade, or enhancing immunostimulatory signaling with bispecific T cell engager and chimeric antigen receptor (CAR)-T cell. Although these agents have already achieved great success, only a tiny percentage of patients could benefit from immunotherapies. Actually, immunotherapy efficacy is determined by multiple components in the tumor microenvironment beyond adaptive immunity. Cells from the innate arm of the immune system, such as macrophages, dendritic cells, myeloid-derived suppressor cells, neutrophils, natural killer cells, and unconventional T cells, also participate in cancer immune evasion and surveillance. Considering that the innate arm is the cornerstone of the antitumor immune response, utilizing innate immunity provides potential therapeutic options for cancer control. Up to now, strategies exploiting innate immunity, such as agonists of stimulator of interferon genes, CAR-macrophage or -natural killer cell therapies, metabolic regulators, and novel immune checkpoint blockade, have exhibited potent antitumor activities in preclinical and clinical studies. Here, we summarize the latest insights into the potential roles of innate cells in antitumor immunity and discuss the advances in innate arm-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ming Yi
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Qi Mei
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China
| | - Bin Zhao
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| | - Zhijun Dai
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China.
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
65
|
Huizing GJ, Deutschmann IM, Peyré G, Cantini L. Paired single-cell multi-omics data integration with Mowgli. Nat Commun 2023; 14:7711. [PMID: 38001063 PMCID: PMC10673889 DOI: 10.1038/s41467-023-43019-2] [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: 02/02/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
The profiling of multiple molecular layers from the same set of cells has recently become possible. There is thus a growing need for multi-view learning methods able to jointly analyze these data. We here present Multi-Omics Wasserstein inteGrative anaLysIs (Mowgli), a novel method for the integration of paired multi-omics data with any type and number of omics. Of note, Mowgli combines integrative Nonnegative Matrix Factorization and Optimal Transport, enhancing at the same time the clustering performance and interpretability of integrative Nonnegative Matrix Factorization. We apply Mowgli to multiple paired single-cell multi-omics data profiled with 10X Multiome, CITE-seq, and TEA-seq. Our in-depth benchmark demonstrates that Mowgli's performance is competitive with the state-of-the-art in cell clustering and superior to the state-of-the-art once considering biological interpretability. Mowgli is implemented as a Python package seamlessly integrated within the scverse ecosystem and it is available at http://github.com/cantinilab/mowgli .
Collapse
Affiliation(s)
- Geert-Jan Huizing
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Machine Learning for Integrative Genomics Group, F-75015, Paris, France.
- Institut de Biologie de l'Ecole Normale Supérieure, CNRS, INSERM, Ecole Normale Supérieure, Université PSL, 75005, Paris, France.
| | - Ina Maria Deutschmann
- Institut de Biologie de l'Ecole Normale Supérieure, CNRS, INSERM, Ecole Normale Supérieure, Université PSL, 75005, Paris, France
| | - Gabriel Peyré
- CNRS and DMA de l'Ecole Normale Supérieure, CNRS, Ecole Normale Supérieure, Université PSL, 75005, Paris, France
| | - Laura Cantini
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Machine Learning for Integrative Genomics Group, F-75015, Paris, France.
- Institut de Biologie de l'Ecole Normale Supérieure, CNRS, INSERM, Ecole Normale Supérieure, Université PSL, 75005, Paris, France.
| |
Collapse
|
66
|
Bernal-Alferes B, Gómez-Mosqueira R, Ortega-Tapia GT, Burgos-Vargas R, García-Latorre E, Domínguez-López ML, Romero-López JP. The role of γδ T cells in the immunopathogenesis of inflammatory diseases: from basic biology to therapeutic targeting. J Leukoc Biol 2023; 114:557-570. [PMID: 37040589 DOI: 10.1093/jleuko/qiad046] [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: 07/15/2022] [Revised: 04/01/2023] [Accepted: 04/06/2023] [Indexed: 04/13/2023] Open
Abstract
The γδ T cells are lymphocytes with an innate-like phenotype that can distribute to different tissues to reside and participate in homeostatic functions such as pathogen defense, tissue modeling, and response to stress. These cells originate during fetal development and migrate to the tissues in a TCR chain-dependent manner. Their unique manner to respond to danger signals facilitates the initiation of cytokine-mediated diseases such as spondyloarthritis and psoriasis, which are immune-mediated diseases with a very strong link with mucosal disturbances, either in the skin or the gut. In spondyloarthritis, γδ T cells are one of the main sources of IL-17 and, therefore, the main drivers of inflammation and probably new bone formation. Remarkably, this population can be the bridge between gut and joint inflammation.
Collapse
Affiliation(s)
- Brian Bernal-Alferes
- Laboratorio de Inmunoquímica 1, Posgrado en Ciencias Quimicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Lázaro Cárdenas, Prolongación de Carpio y Plan de Ayala s/n, Col. Santo Tomás C.P. 11340 Alcaldía Miguel Hidalgo, Ciudad de México, México
| | - Rafael Gómez-Mosqueira
- Laboratorio de Inmunoquímica 1, Posgrado en Ciencias Quimicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Lázaro Cárdenas, Prolongación de Carpio y Plan de Ayala s/n, Col. Santo Tomás C.P. 11340 Alcaldía Miguel Hidalgo, Ciudad de México, México
| | - Graciela Teresa Ortega-Tapia
- Laboratorio de Inmunoquímica 1, Posgrado en Ciencias Quimicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Lázaro Cárdenas, Prolongación de Carpio y Plan de Ayala s/n, Col. Santo Tomás C.P. 11340 Alcaldía Miguel Hidalgo, Ciudad de México, México
| | - Rubén Burgos-Vargas
- Departamento de Reumatología, Hospital General de México "Dr. Eduardo Liceaga", Dr. Balmis No. 148 Col. Doctores C.P. 06720, Alcaldía Cuauhtémoc Ciudad de México, México
| | - Ethel García-Latorre
- Laboratorio de Inmunoquímica 1, Posgrado en Ciencias Quimicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Lázaro Cárdenas, Prolongación de Carpio y Plan de Ayala s/n, Col. Santo Tomás C.P. 11340 Alcaldía Miguel Hidalgo, Ciudad de México, México
| | - María Lilia Domínguez-López
- Laboratorio de Inmunoquímica 1, Posgrado en Ciencias Quimicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Lázaro Cárdenas, Prolongación de Carpio y Plan de Ayala s/n, Col. Santo Tomás C.P. 11340 Alcaldía Miguel Hidalgo, Ciudad de México, México
| | - José Pablo Romero-López
- Laboratorio de Patogénesis Molecular, Edificio A4, Red MEDICI, Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios Número 1, Colonia Los Reyes Ixtacala, C.P. 54090, Tlalnepantla, Estado de México, México
| |
Collapse
|
67
|
Klussmeier A, Putke K, Klasberg S, Kohler M, Sauter J, Schefzyk D, Schöfl G, Massalski C, Schäfer G, Schmidt AH, Roers A, Lange V. High population frequencies of MICA copy number variations originate from independent recombination events. Front Immunol 2023; 14:1297589. [PMID: 38035108 PMCID: PMC10684724 DOI: 10.3389/fimmu.2023.1297589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 10/24/2023] [Indexed: 12/02/2023] Open
Abstract
MICA is a stress-induced ligand of the NKG2D receptor that stimulates NK and T cell responses and was identified as a key determinant of anti-tumor immunity. The MICA gene is located inside the MHC complex and is in strong linkage disequilibrium with HLA-B. While an HLA-B*48-linked MICA deletion-haplotype was previously described in Asian populations, little is known about other MICA copy number variations. Here, we report the genotyping of more than two million individuals revealing high frequencies of MICA duplications (1%) and MICA deletions (0.4%). Their prevalence differs between ethnic groups and can rise to 2.8% (Croatia) and 9.2% (Mexico), respectively. Targeted sequencing of more than 70 samples indicates that these copy number variations originate from independent nonallelic homologous recombination events between segmental duplications upstream of MICA and MICB. Overall, our data warrant further investigation of disease associations and consideration of MICA copy number data in oncological study protocols.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Axel Roers
- Institute for Immunology, Medical Faculty Carl Gustav Carus, University of Technology (TU) Dresden, Dresden, Germany
- Institute for Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | | |
Collapse
|
68
|
Wang J, Nakafuku KM, Ziff J, Gelin CF, Gholami H, Thompson AA, Karpowich NK, Limon L, Coate HR, Damm-Ganamet KL, Shih AY, Grant JC, Côte M, Mak PA, Pascual HA, Rives ML, Edwards JP, Venable JD, Venkatesan H, Shi Z, Allen SJ, Sharma S, Kung PP, Shireman BT. Development of small molecule inhibitors of natural killer group 2D receptor (NKG2D). Bioorg Med Chem Lett 2023; 96:129492. [PMID: 37778428 DOI: 10.1016/j.bmcl.2023.129492] [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: 07/24/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/03/2023]
Abstract
Natural killer group 2D (NKG2D) is a homodimeric activating immunoreceptor whose function is to detect and eliminate compromised cells upon binding to the NKG2D ligands (NKG2DL) major histocompatibility complex (MHC) molecules class I-related chain A (MICA) and B (MICB) and UL16 binding proteins (ULBP1-6). While typically present at low levels in healthy cells and tissue, NKG2DL expression can be induced by viral infection, cellular stress or transformation. Aberrant activity along the NKG2D/NKG2DL axis has been associated with autoimmune diseases due to the increased expression of NKG2D ligands in human disease tissue, making NKG2D inhibitors an attractive target for immunomodulation. Herein we describe the discovery and optimization of small molecule PPI (protein-protein interaction) inhibitors of NKG2D/NKG2DL. Rapid SAR was guided by structure-based drug design and accomplished by iterative singleton and parallel medicinal chemistry synthesis. These efforts resulted in the identification of several potent analogs (14, 21, 30, 45) with functional activity and improved LLE.
Collapse
Affiliation(s)
- Jocelyn Wang
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States.
| | - Kohki M Nakafuku
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States.
| | - Jeannie Ziff
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| | - Christine F Gelin
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| | - Hadi Gholami
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| | - Aaron A Thompson
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| | - Nathan K Karpowich
- Janssen Research & Development L.L.C., 1400 McKean Rd., Spring House, PA 19477, United States
| | - Luis Limon
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| | - Heather R Coate
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| | - Kelly L Damm-Ganamet
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| | - Amy Y Shih
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| | - Joanna C Grant
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| | - Marjorie Côte
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| | - Puiying A Mak
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| | - Heather A Pascual
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| | - Marie-Laure Rives
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| | - James P Edwards
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| | - Jennifer D Venable
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| | - Hariharan Venkatesan
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| | - Zhicai Shi
- Janssen Research & Development L.L.C., 1400 McKean Rd., Spring House, PA 19477, United States
| | - Samantha J Allen
- Janssen Research & Development L.L.C., 1400 McKean Rd., Spring House, PA 19477, United States
| | - Sujata Sharma
- Janssen Research & Development L.L.C., 1400 McKean Rd., Spring House, PA 19477, United States
| | - Pei-Pei Kung
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| | - Brock T Shireman
- Janssen Research & Development, L.L.C., 3210 Merryfield Row, San Diego, CA, 92121, United States
| |
Collapse
|
69
|
Abstract
The use of cancer vaccines is considered a promising therapeutic strategy in clinical oncology, which is achieved by stimulating antitumor immunity with tumor antigens delivered in the form of cells, peptides, viruses, and nucleic acids. The ideal cancer vaccine has many advantages, including low toxicity, specificity, and induction of persistent immune memory to overcome tumor heterogeneity and reverse the immunosuppressive microenvironment. Many therapeutic vaccines have entered clinical trials for a variety of cancers, including melanoma, breast cancer, lung cancer, and others. However, many challenges, including single antigen targeting, weak immunogenicity, off-target effects, and impaired immune response, have hindered their broad clinical translation. In this review, we introduce the principle of action, components (including antigens and adjuvants), and classification (according to applicable objects and preparation methods) of cancer vaccines, summarize the delivery methods of cancer vaccines, and review the clinical and theoretical research progress of cancer vaccines. We also present new insights into cancer vaccine technologies, platforms, and applications as well as an understanding of potential next-generation preventive and therapeutic vaccine technologies, providing a broader perspective for future vaccine design.
Collapse
Affiliation(s)
- Nian Liu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| | - Xiangyu Xiao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| | - Ziqiang Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| | - Jian Shen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210023, China
| |
Collapse
|
70
|
Olayinka JT, Nagarkar A, Ma DJ, Wong NB, Romasco A, Piedra-Mora C, Wrijil L, David CN, Gardner HL, Robinson NA, Hughes KL, Barton B, London CA, Almela RM, Richmond JM. Cathepsin W, T-cell receptor-associated transmembrane adapter 1, lymphotactin and killer cell lectin like receptor K1 are sensitive and specific RNA biomarkers of canine epitheliotropic lymphoma. Front Vet Sci 2023; 10:1225764. [PMID: 38026637 PMCID: PMC10654980 DOI: 10.3389/fvets.2023.1225764] [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: 05/19/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Cutaneous T-cell lymphoma (CTCL) is an uncommon type of lymphoma involving malignant skin-resident or skin-homing T cells. Canine epitheliotropic lymphoma (EL) is the most common form of CTCL in dogs, and it also spontaneously arises from T lymphocytes in the mucosa and skin. Clinically, it can be difficult to distinguish early-stage CTCLs apart from other forms of benign interface dermatitis (ID) in both dogs and people. Our objective was to identify novel biomarkers that can distinguish EL from other forms of ID, and perform comparative transcriptomics of human CTCL and canine EL. Here, we present a retrospective gene expression study that employed archival tissue from biorepositories. We analyzed a discovery cohort of 6 canines and a validation cohort of 8 canines with EL which occurred spontaneously in client-owned companion dogs. We performed comparative targeted transcriptomics studies using NanoString to assess 160 genes from lesional skin biopsies from the discovery cohort and 800 genes from the validation cohort to identify any significant differences that may reflect oncogenesis and immunopathogenesis. We further sought to determine if gene expression in EL and CTCL are conserved across humans and canines by comparing our data to previously published human datasets. Similar chemokine profiles were observed in dog EL and human CTCL, and analyses were performed to validate potential biomarkers and drivers of disease. In dogs, we found enrichment of T cell gene signatures, with upregulation of IFNG, TNF, PRF1, IL15, CD244, CXCL10, and CCL5 in EL in dogs compared to healthy controls. Importantly, CTSW, TRAT1 and KLRK1 distinguished EL from all other forms of interface dermatitis we studied, providing much-needed biomarkers for the veterinary field. XCL1/XCL2 were also highly specific of EL in our validation cohort. Future studies exploring the oncogenesis of spontaneous lymphomas in companion animals will expand our understanding of these disorders. Biomarkers may be useful for predicting disease prognosis and treatment responses. We plan to use our data to inform future development of targeted therapies, as well as for repurposing drugs for both veterinary and human medicine.
Collapse
Affiliation(s)
- Jadesola Temitope Olayinka
- Department of Dermatology, UMass Chan Medical School, Worcester, MA, United States
- SUNY Downstate School of Medicine, New York, NY, United States
| | - Akanksha Nagarkar
- Department of Dermatology, UMass Chan Medical School, Worcester, MA, United States
| | - Diana Junyue Ma
- Department of Dermatology, UMass Chan Medical School, Worcester, MA, United States
| | - Neil B. Wong
- Department of Dermatology, UMass Chan Medical School, Worcester, MA, United States
| | - Andrew Romasco
- Department of Dermatology, UMass Chan Medical School, Worcester, MA, United States
| | - Cesar Piedra-Mora
- Pathology Department, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, United States
| | - Linda Wrijil
- Pathology Department, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, United States
| | | | - Heather L. Gardner
- Department of Clinical Sciences, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, United States
| | - Nicholas A. Robinson
- Pathology Department, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, United States
| | - Kelly L. Hughes
- Department of Microbiology, Immunology and Pathology, Colorado State University Veterinary Diagnostic Laboratory, Fort Collins, CO, United States
| | - Bruce Barton
- Department of Population and Quantitative Health Sciences, UMass Chan Medical School, Worcester, MA, United States
| | - Cheryl A. London
- Department of Clinical Sciences, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, United States
| | - Ramón M. Almela
- Department of Clinical Sciences, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, United States
| | - Jillian M. Richmond
- Department of Dermatology, UMass Chan Medical School, Worcester, MA, United States
| |
Collapse
|
71
|
Ali ES, Ben-Sahra I. Regulation of nucleotide metabolism in cancers and immune disorders. Trends Cell Biol 2023; 33:950-966. [PMID: 36967301 PMCID: PMC10518033 DOI: 10.1016/j.tcb.2023.03.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/05/2023] [Accepted: 03/08/2023] [Indexed: 04/03/2023]
Abstract
Nucleotides are the foundational elements of life. Proliferative cells acquire nutrients for energy production and the synthesis of macromolecules, including proteins, lipids, and nucleic acids. Nucleotides are continuously replenished through the activation of the nucleotide synthesis pathways. Despite the importance of nucleotides in cell physiology, there is still much to learn about how the purine and pyrimidine synthesis pathways are regulated in response to intracellular and exogenous signals. Over the past decade, evidence has emerged that several signaling pathways [Akt, mechanistic target of rapamycin complex I (mTORC1), RAS, TP53, and Hippo-Yes-associated protein (YAP) signaling] alter nucleotide synthesis activity and influence cell function. Here, we examine the mechanisms by which these signaling networks affect de novo nucleotide synthesis in mammalian cells. We also discuss how these molecular links can be targeted in diseases such as cancers and immune disorders.
Collapse
Affiliation(s)
- Eunus S Ali
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Issam Ben-Sahra
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
72
|
Lutz S, Klausz K, Albici AM, Ebinger L, Sellmer L, Teipel H, Frenzel A, Langner A, Winterberg D, Krohn S, Hust M, Schirrmann T, Dübel S, Scherließ R, Humpe A, Gramatzki M, Kellner C, Peipp M. Novel NKG2D-directed bispecific antibodies enhance antibody-mediated killing of malignant B cells by NK cells and T cells. Front Immunol 2023; 14:1227572. [PMID: 37965326 PMCID: PMC10641740 DOI: 10.3389/fimmu.2023.1227572] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/05/2023] [Indexed: 11/16/2023] Open
Abstract
The activating receptor natural killer group 2, member D (NKG2D) represents an attractive target for immunotherapy as it exerts a crucial role in cancer immunosurveillance by regulating the activity of cytotoxic lymphocytes. In this study, a panel of novel NKG2D-specific single-chain fragments variable (scFv) were isolated from naïve human antibody gene libraries and fused to the fragment antigen binding (Fab) of rituximab to obtain [CD20×NKG2D] bibodies with the aim to recruit cytotoxic lymphocytes to lymphoma cells. All bispecific antibodies bound both antigens simultaneously. Two bibody constructs, [CD20×NKG2D#3] and [CD20×NKG2D#32], efficiently activated natural killer (NK) cells in co-cultures with CD20+ lymphoma cells. Both bibodies triggered NK cell-mediated lysis of lymphoma cells and especially enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) by CD38 or CD19 specific monoclonal antibodies suggesting a synergistic effect between NKG2D and FcγRIIIA signaling pathways in NK cell activation. The [CD20×NKG2D] bibodies were not effective in redirecting CD8+ T cells as single agents, but enhanced cytotoxicity when combined with a bispecific [CD19×CD3] T cell engager, indicating that NKG2D signaling also supports CD3-mediated T cell activation. In conclusion, engagement of NKG2D with bispecific antibodies is attractive to directly activate cytotoxic lymphocytes or to support their activation by monoclonal antibodies or bispecific T cell engagers. As a perspective, co-targeting of two tumor antigens may allow fine-tuning of antibody cancer therapies. Our proposed combinatorial approach is potentially applicable for many existing immunotherapies but further testing in different preclinical models is necessary to explore the full potential.
Collapse
Affiliation(s)
- Sebastian Lutz
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, Ludwig Maximilians University (LMU) Munich, Munich, Germany
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Katja Klausz
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Anca-Maria Albici
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Lea Ebinger
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Lea Sellmer
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Hannah Teipel
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | | | - Anna Langner
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Dorothee Winterberg
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Steffen Krohn
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Michael Hust
- YUMAB GmbH, Braunschweig, Germany
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | | | - Stefan Dübel
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Regina Scherließ
- Department of Pharmaceutics and Biopharmaceutics, Kiel University, Kiel, Germany
| | - Andreas Humpe
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, Ludwig Maximilians University (LMU) Munich, Munich, Germany
| | - Martin Gramatzki
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Christian Kellner
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, Ludwig Maximilians University (LMU) Munich, Munich, Germany
| | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| |
Collapse
|
73
|
Zhang J, Luo Q, Li X, Guo J, Zhu Q, Lu X, Wei L, Xiang Z, Peng M, Ou C, Zou Y. Novel role of immune-related non-coding RNAs as potential biomarkers regulating tumour immunoresponse via MICA/NKG2D pathway. Biomark Res 2023; 11:86. [PMID: 37784183 PMCID: PMC10546648 DOI: 10.1186/s40364-023-00530-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 09/24/2023] [Indexed: 10/04/2023] Open
Abstract
Major histocompatibility complex class I related chain A (MICA) is an important and stress-induced ligand of the natural killer group 2 member D receptor (NKG2D) that is expressed in various tumour cells. Given that the MICA/NKG2D signalling system is critically embedded in the innate and adaptive immune responses, it is particularly involved in the surveillance of cancer and viral infections. Emerging evidence has revealed the important roles of non-coding RNAs (ncRNAs) including microRNAs (miRNAs), long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs) in different cancer types. We searched for all relevant publications in the PubMed, Scopus and Web of Science database using the keywords ncRNA, MICA, NKG2D, cancer, and miRNAs. All relevant studies published from 2008 to the 2023 were retrieved and collated. Notably, we found that miRNAs can target to NKG2D mRNA and MICA mRNA 3'-untranslated regions (3'-UTR), leading to translation inhibition of NKG2D and MICA degradation. Several immune-related MICA/NKG2D pathways may be dysregulated in cancer with aberrant miRNA expressions. At the same time, the competitive endogenous RNA (ceRNA) hypothesis holds that circRNAs, lncRNAs, and mRNAs induce an abnormal MICA expression by directly targeting downstream miRNAs to mediate mRNA suppression in cancer. This review summarizes the novel mechanism of immune escape in the ncRNA-related MICA/NKG2D pathway mediated by NK cells and cancer cells. Moreover, we identified the miRNA-NKG2D, miRNA-MICA and circRNA/lncRNA/mRNA-miRNA-mRNA/MICA axis. Thus, we were particularly concerned with the regulation of mediated immune escape in the MICA/NKG2D pathway by ncRNAs as potential therapeutic targets and diagnostic biomarkers of immunity and cancer.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Immunology, School of Basic Medicine, Central South University, Changsha, 410000, Hunan, China
| | - Qizhi Luo
- Department of Immunology, School of Basic Medicine, Central South University, Changsha, 410000, Hunan, China
| | - Xin Li
- Department of Immunology, School of Basic Medicine, Central South University, Changsha, 410000, Hunan, China
| | - Junshuang Guo
- Department of Immunology, School of Basic Medicine, Central South University, Changsha, 410000, Hunan, China
| | - Quan Zhu
- Department of Immunology, School of Basic Medicine, Central South University, Changsha, 410000, Hunan, China
| | - Xiaofang Lu
- Department of Immunology, School of Basic Medicine, Central South University, Changsha, 410000, Hunan, China
| | - Leiyan Wei
- Department of Immunology, School of Basic Medicine, Central South University, Changsha, 410000, Hunan, China
| | - Zhiqing Xiang
- Department of Immunology, School of Basic Medicine, Central South University, Changsha, 410000, Hunan, China
| | - Manqing Peng
- Department of Immunology, School of Basic Medicine, Central South University, Changsha, 410000, Hunan, China
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China.
| | - Yizhou Zou
- Department of Immunology, School of Basic Medicine, Central South University, Changsha, 410000, Hunan, China.
| |
Collapse
|
74
|
Zarei M, Abdoli S, Farazmandfar T, Shahbazi M. Lenalidomide improves NKG2D-based CAR-T cell activity against colorectal cancer cells invitro. Heliyon 2023; 9:e20460. [PMID: 37790973 PMCID: PMC10543764 DOI: 10.1016/j.heliyon.2023.e20460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 09/13/2023] [Accepted: 09/26/2023] [Indexed: 10/05/2023] Open
Abstract
Introduction Although CAR-based immunotherapy is viewed as a promising treatment for tumors, particularly hematological malignancies, solid tumors can pose challenges. It has been suggested that the immunomodulatory medication Lenalidomide (LEN) may increase the effectiveness of CAR T cells in the treatment of solid tumors. The purpose of our study was to investigate the effect of NKG2D-based CAR T cell therapy on colorectal cancer cell lines, and then we assessed combinatorial therapy using NKG2D CAR T cells and lenalidomide in vitro. Methods and results To prepare NKG2D CAR T cells, a second-generation NKG2D-CAR construct was designed and transfected into the T cells using a lentiviral system. The NKG2D CAR T cells showed significantly higher cytotoxic activity against colorectal cancer cell lines, HCT116 and SW480, compared to untransduced T cells. In addition, our data demonstrated that the cytotoxicity and cytokine secretion of NKG2D CAR T cells significantly increased in the presence of higher doses of lenalidomide. Conclusions The study findings suggest that combinational therapy, utilizing NKG2D-based CAR T cells and lenalidomide, has a high potential for effectively eliminating tumor cells in vitro.
Collapse
Affiliation(s)
- Mahdi Zarei
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Shahriyar Abdoli
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Touraj Farazmandfar
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Shahbazi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- AryaTina Gene (ATG) Biopharmaceutical Company Gorgan, Iran
| |
Collapse
|
75
|
Jiang J, Liu Y, Zeng Y, Fang B, Chen Y. Annihilation of Non-small Cell Lung Cancer by NKG2D CAR-T Cells Produced from T Cells from Peripheral Blood of Healthy Donors. J Interferon Cytokine Res 2023; 43:445-454. [PMID: 37819621 DOI: 10.1089/jir.2023.0043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023] Open
Abstract
Some progress has been made in immunotherapy with chimeric antigen receptor (CAR)-T cells targeting NKG2D-NKG2DL with the purpose of eradicating solid tumors. Non-small cell lung cancer (NSCLC) has been shown to express NKG2DL. This study hence evaluated the therapeutic effect of NKG2D CAR-T cells on NSCLC. Accordingly, NKG2D CAR-T cells were obtained from diverse human autologous T cell sources. T cells from peripheral blood T lymphocytes of healthy volunteers (without NKG2D CAR insertion) were used as NT-T cells. Coculture of effector cells (CAR-T cells or NT-T cells) with target cells (NSCLC cells such as PC-9 or NCL-H460 cells) was performed at different ratios. The cytotoxicity of CAR-T cells was examined using lactate dehydrogenase assay kits. Murine xenograft assay was conducted to investigate the in vivo antitumor effect of CAR-T cells. Cytokines secreted from CAR-T cells were assessed by enzyme-linked immunosorbent assay. CAR-T cell infiltration into xenografts was observed through immunochemical assay. Based on the results, NKG2DL was highly expressed in NSCLC cells. Compared with NT-T cells, NKG2D CAR-T cells from different sources of T cells delivered stronger toxicity, and secreted more effector and memory function-related cytokines to NSCLC cells, and those from the peripheral blood of healthy donors (H-T cells) exhibited the strongest effect. Furthermore, compared with NT-T cells, H-T cells and NKG2D CAR-T cells from NSCLC patients' peripheral blood diminished tumor, improved survival, increased body weight and tumor-infiltrating capacity, and upregulated serum IFN-γ level in NOG mice. Collectively speaking, NKG2D CAR-T cells exhibit a robust effect on eradicating NSCLC in a NKG2DL-dependent manner, thus making themselves a promising therapeutic candidate for NSCLC patients.
Collapse
Affiliation(s)
- Jinhong Jiang
- The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui City, China
| | - Yonghua Liu
- The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui City, China
| | - Yuxiao Zeng
- The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui City, China
| | - Bingmu Fang
- The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui City, China
| | | |
Collapse
|
76
|
Lopez-Montaño M, Jimenez-Ortega L, Cruz-Hernandez TR, Hernandez-Chavez VG, Montiel-Cervantes LA, Reyes-Maldonado E, Vela-Ojeda J. Significant increase in MIC-A and MIC-B and soluble MIC-A and MIC-B in canine lymphomas. Vet Immunol Immunopathol 2023; 264:110647. [PMID: 37672843 DOI: 10.1016/j.vetimm.2023.110647] [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: 06/14/2023] [Revised: 08/10/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023]
Abstract
Non-Hodkin's lymphoma (NHL) is the most frequent hematologic malignancy in humans and dogs. NKG2D is one of the most critical receptors on NK cells, recognizing their natural ligands on malignant cells such as A and B major histocompatibility complex-related proteins (MIC-A and MIC-B). Soluble molecules (sMIC-A and sMIC-B) can interfere with immune synapsis between NK cells and tumor cells, impeding NK cytotoxicity. The main objectives of this study were to analyze, in dogs with diffuse large B cell lymphoma, NK cell lymphoma, and reactive lymphadenopathies, the role of NK cells, their activating receptors NKG2D and NKp46, and their ligands MIC-A and MIC-B, as well as soluble molecules sMIC-A and sMIC-B. Thirty-six dogs with a possible diagnosis of NHL and eight healthy dogs were studied. NHL was diagnosed in 28 (78 %) dogs; in the other 8 (22 %), reactive lymphadenopathies were present. Most of the lymphomas corresponded to B cell NHL (82 %). The most predominant subtype was diffuse large B cell lymphoma (21, 71.5 %), followed by five cases (18 %) that were Non-B Non-T lymphomas (presumably NK cell lymphomas) and other B cell lymphomas (3, 10.5%). There were no cases of T cell NHL. MIC-A was positive in 7 of 27 (26 %) cases of NHL, and MIC-B in 20 of 27 (74 %) NHL. In non-malignant lymphadenopathies, three (37.5 %) dogs were positive for MIC-A, and five (62.5 %) expressed MIC-B. Dogs with lymphoma had higher numbers of NK cells than eight healthy dogs. In 15 dogs (12 cases with NHL and three cases with reactive adenopathies) and eight controls, there were no differences in the number of NK cells expressing NKP46 and NKG2D. NHL dogs had higher values of sMIC-A and sMIC-B. B-cell and NK cell lymphomas correspond to 86 % and 14 % of all canine lymphomas. MIC-A, MIC-B, and sMIC-A and sMIC-B were increased in canine lymphomas.
Collapse
Affiliation(s)
- Maresa Lopez-Montaño
- Departamento de Morfología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Manuel Carpio y Plan de Ayala, Del. Miguel Hidalgo, 11340 Mexico City, Mexico
| | - Laura Jimenez-Ortega
- Escuela Superior de Medicina, Sección de Estudios de Posgrado e Investigación, Instituto Politécnico Nacional, Plan de San Luis y Salvador Díaz Mirón S/N, Col. Casco de Santo Tomás, CP 11340 Mexico City, Mexico
| | - Teresa Rocio Cruz-Hernandez
- Centro de diagnóstico veterinario especializado (cedivete), Área de histopatología Calle Iztapalapa 9, San Antonio, Iztapalapa, CP 09900 Mexico City, Mexico
| | - Victor Gabriel Hernandez-Chavez
- Departamento de Morfología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Manuel Carpio y Plan de Ayala, Del. Miguel Hidalgo, 11340 Mexico City, Mexico
| | - Laura Arcelia Montiel-Cervantes
- Departamento de Morfología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Manuel Carpio y Plan de Ayala, Del. Miguel Hidalgo, 11340 Mexico City, Mexico; Departamento de Hematología, Unidad Médica de Alta Especialidad, Centro Médico Nacional La Raza, Instituto Mexicano del Seguro Social, Seris y Zaachila S/N Colonia La Raza, Azcapotzalco, 02990 Mexico City, Mexico
| | - Elba Reyes-Maldonado
- Departamento de Morfología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Manuel Carpio y Plan de Ayala, Del. Miguel Hidalgo, 11340 Mexico City, Mexico
| | - Jorge Vela-Ojeda
- Departamento de Morfología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Manuel Carpio y Plan de Ayala, Del. Miguel Hidalgo, 11340 Mexico City, Mexico.
| |
Collapse
|
77
|
Paolini R, Molfetta R. Dysregulation of DNAM-1-Mediated NK Cell Anti-Cancer Responses in the Tumor Microenvironment. Cancers (Basel) 2023; 15:4616. [PMID: 37760586 PMCID: PMC10527063 DOI: 10.3390/cancers15184616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/10/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
NK cells play a pivotal role in anti-cancer immune responses, thanks to the expression of a wide array of inhibitory and activating receptors that regulate their cytotoxicity against transformed cells while preserving healthy cells from lysis. However, NK cells exhibit severe dysfunction in the tumor microenvironment, mainly due to the reduction of activating receptors and the induction or increased expression of inhibitory checkpoint receptors. An activating receptor that plays a central role in tumor recognition is the DNAM-1 receptor. It recognizes PVR and Nectin2 adhesion molecules, which are frequently overexpressed on the surface of cancerous cells. These ligands are also able to trigger inhibitory signals via immune checkpoint receptors that are upregulated in the tumor microenvironment and can counteract DNAM-1 activation. Among them, TIGIT has recently gained significant attention, since its targeting results in improved anti-tumor immune responses. This review aims to summarize how the recognition of PVR and Nectin2 by paired co-stimulatory/inhibitory receptors regulates NK cell-mediated clearance of transformed cells. Therapeutic approaches with the potential to reverse DNAM-1 dysfunction in the tumor microenvironment will be also discussed.
Collapse
Affiliation(s)
| | - Rosa Molfetta
- Department of Molecular Medicine, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy;
| |
Collapse
|
78
|
Kiaei SZF, Nouralishahi A, Ghasemirad M, Barkhordar M, Ghaffari S, Kheradjoo H, Saleh M, Mohammadzadehsaliani S, Molaeipour Z. Advances in natural killer cell therapies for breast cancer. Immunol Cell Biol 2023; 101:705-726. [PMID: 37282729 DOI: 10.1111/imcb.12658] [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: 03/21/2023] [Revised: 04/28/2023] [Accepted: 05/06/2023] [Indexed: 06/08/2023]
Abstract
Breast cancer (BC) is the most common cause of cancer death in women. According to the American Cancer Society's yearly cancer statistics, BC constituted almost 15% of all the newly diagnosed cancer cases in 2022 for both sexes. Metastatic disease occurs in 30% of patients with BC. The currently available treatments fail to cure metastatic BC, and the average survival time for patients with metastatic BC is approximately 2 years. Developing a treatment method that terminates cancer stem cells without harming healthy cells is the primary objective of novel therapeutics. Adoptive cell therapy is a branch of cancer immunotherapy that utilizes the immune cells to attack cancer cells. Natural killer (NK) cells are an essential component of innate immunity and are critical in destroying tumor cells without prior stimulation with antigens. With the advent of chimeric antigen receptors (CARs), the autologous or allogeneic use of NK/CAR-NK cell therapy has raised new hopes for treating patients with cancer. Here, we describe recent developments in NK and CAR-NK cell immunotherapy, including the biology and function of NK cells, clinical trials, different sources of NK cells and their future perspectives on BC.
Collapse
Affiliation(s)
- Seyedeh Zahra Fotook Kiaei
- Department of Pulmonary and Critical Care, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Ghasemirad
- Department of Periodontics, Faculty of Dentistry, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Maryam Barkhordar
- Hematology, Oncology and Stem Cell Transplantation Research Center (HORCSCT), Tehran University of Medical Sciences, Tehran, Iran
| | - Sasan Ghaffari
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | | | - Mahshid Saleh
- Wisconsin National Primate Research Center, University of Wisconsin Graduate School, Madison, WI, USA
| | | | - Zahra Molaeipour
- Hematology Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
79
|
Hu B, Xin Y, Hu G, Li K, Tan Y. Fluid shear stress enhances natural killer cell's cytotoxicity toward circulating tumor cells through NKG2D-mediated mechanosensing. APL Bioeng 2023; 7:036108. [PMID: 37575881 PMCID: PMC10423075 DOI: 10.1063/5.0156628] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/19/2023] [Indexed: 08/15/2023] Open
Abstract
Tumor cells metastasize to distant organs mainly via hematogenous dissemination, in which circulating tumor cells (CTCs) are relatively vulnerable, and eliminating these cells has great potential to prevent metastasis. In vasculature, natural killer (NK) cells are the major effector lymphocytes for efficient killing of CTCs under fluid shear stress (FSS), which is an important mechanical cue in tumor metastasis. However, the influence of FSS on the cytotoxicity of NK cells against CTCs remains elusive. We report that the death rate of CTCs under both NK cells and FSS is much higher than the combined death induced by either NK cells or FSS, suggesting that FSS may enhance NK cell's cytotoxicity. This death increment is elicited by shear-induced NK activation and granzyme B entry into target cells rather than the death ligand TRAIL or secreted cytokines TNF-α and IFN-γ. When NK cells form conjugates with CTCs or adhere to MICA-coated substrates, NK cell activating receptor NKG2D can directly sense FSS to induce NK activation and degranulation. These findings reveal the promotive effect of FSS on NK cell's cytotoxicity toward CTCs, thus providing new insight into immune surveillance of CTCs within circulation.
Collapse
Affiliation(s)
| | | | | | | | - Youhua Tan
- Author to whom correspondence should be addressed:
| |
Collapse
|
80
|
Yang D, Sun B, Li S, Wei W, Liu X, Cui X, Zhang X, Liu N, Yan L, Deng Y, Zhao X. NKG2D-CAR T cells eliminate senescent cells in aged mice and nonhuman primates. Sci Transl Med 2023; 15:eadd1951. [PMID: 37585504 DOI: 10.1126/scitranslmed.add1951] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/10/2023] [Indexed: 08/18/2023]
Abstract
Cellular senescence, characterized by stable cell cycle arrest, plays an important role in aging and age-associated pathologies. Eliminating senescent cells rejuvenates aged tissues and ameliorates age-associated diseases. Here, we identified that natural killer group 2 member D ligands (NKG2DLs) are up-regulated in senescent cells in vitro, regardless of stimuli that induced cellular senescence, and in various tissues of aged mice and nonhuman primates in vivo. Accordingly, we developed and demonstrated that chimeric antigen receptor (CAR) T cells targeting human NKG2DLs selectively and effectively diminish human cells undergoing senescence induced by oncogenic stress, replicative stress, DNA damage, or P16INK4a overexpression in vitro. Targeting senescent cells with mouse NKG2D-CAR T cells alleviated multiple aging-associated pathologies and improved physical performance in both irradiated and aged mice. Autologous T cells armed with the human NKG2D CAR effectively delete naturally occurring senescent cells in aged nonhuman primates without any observed adverse effects. Our findings establish that NKG2D-CAR T cells could serve as potent and selective senolytic agents for aging and age-associated diseases driven by senescence.
Collapse
Affiliation(s)
- Dong Yang
- Department of Targeting Therapy and Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Bin Sun
- Department of Targeting Therapy and Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shirong Li
- Department of Targeting Therapy and Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wenwen Wei
- Department of Targeting Therapy and Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiuyun Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming,, Yunnan 650223, China
| | - Xiaoyue Cui
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming,, Yunnan 650223, China
| | - Xianning Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming,, Yunnan 650223, China
| | - Nan Liu
- Department of Targeting Therapy and Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lanzhen Yan
- Department of Targeting Therapy and Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yibin Deng
- Department of Urology, Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Xudong Zhao
- Department of Targeting Therapy and Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming,, Yunnan 650223, China
| |
Collapse
|
81
|
Kaminski VDL, Kulmann-Leal B, Tyska-Nunes GL, Beltrame BP, Riesgo RDS, Schüler-Faccini L, Roman T, Schuch JB, Chies JAB. Association between NKG2/KLR gene variants and epilepsy in Autism Spectrum Disorder. J Neuroimmunol 2023; 381:578132. [PMID: 37352688 DOI: 10.1016/j.jneuroim.2023.578132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/31/2023] [Accepted: 06/15/2023] [Indexed: 06/25/2023]
Abstract
Autism Spectrum Disorder (ASD) is a set of neurodevelopmental disorders mainly characterized by repetitive, restrictive and stereotypical behaviors, and impaired communication skills. Several lines of evidence indicate that alterations of the immune system account for ASD development, including the presence of brain-reactive antibodies, abnormal T cell activation, altered cytokine levels in brain, cerebrospinal fluid and peripheral blood circulation, increased levels of circulating monocytes, and dysregulation in Natural Killer (NK) cells activity. Regarding NK cells, a lower cytotoxic activity, a higher level of activation and an increased number of these cells in individuals with ASD have been described. In 2019, a study showed that NK cells derived from patients with ASD show a characteristic pattern of NKG2C overexpression, highlighting the importance of the NK cell pathway in ASD. In fact, the study of genes related to NK cell activity has proven to be an excellent research target, both in terms of susceptibility as well as a marker for the different clinical manifestations observed in ASD individuals. Here, we evaluated the influence of KLRC2 gene deletion as well as KLRK1 rs1049174 and rs2255336 variants in a cohort of 185 children diagnosed with ASD and their respective biological parents in southern Brazil. Of note, this is the first study concerning genetic variants of the KLRC2 and KLRK1 genes in an ASD sample. The KLRC2 gene deletion (p = 0.001; pc = 0.009), KLRK1 rs1049174 (p = 0.005; pc = 0.045) and KLRK1 rs2255336 (p = 0.001; pc = 0.009) were associated with epilepsy in ASD patients. The results indicate that KLRC2 deletion, KLRK1 rs2255336, and KLRK1 rs1049174 could be involved in epilepsy manifestation in ASD patients, possibly impacting the NK dysregulation already described in ASD and epileptic patients.
Collapse
Affiliation(s)
- Valéria de Lima Kaminski
- Laboratory of Immunobiology and Immunogenetics, Graduate Program in Genetics and Molecular Biology - PPGBM, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Applied Immunology Laboratory, Graduate Program in Biotechnology, Institute of Science and Technology - ICT, Universidade Federal de São Paulo - UNIFESP, São José dos Campos - São Paulo, Brazil; Universidade Anhembi Morumbi, São José dos Campos - São Paulo, Brazil
| | - Bruna Kulmann-Leal
- Laboratory of Immunobiology and Immunogenetics, Graduate Program in Genetics and Molecular Biology - PPGBM, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Guilherme Luís Tyska-Nunes
- Laboratory of Immunobiology and Immunogenetics, Graduate Program in Genetics and Molecular Biology - PPGBM, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Brenda Pedron Beltrame
- Laboratory of Immunobiology and Immunogenetics, Graduate Program in Genetics and Molecular Biology - PPGBM, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Rudimar Dos Santos Riesgo
- Child Neurology Unit, Hospital de Clínicas de Porto Alegre (HCPA), Universidade Federal do Rio Grande do Sul - UFRGS, Rua Ramiro Barcelos, 2350, Porto Alegre 90035-903, Brazil
| | - Lavinia Schüler-Faccini
- Laboratory of Immunobiology and Immunogenetics, Graduate Program in Genetics and Molecular Biology - PPGBM, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil; National Institute of Population Medical Genetics (INAGEMP), Porto Alegre, Brazil; Brazilian Teratogen Information Service (SIAT), Medical Genetics Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Tatiana Roman
- Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Jaqueline Bohrer Schuch
- Graduate Program in Psychiatry and Behavioral Sciences, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - José Artur Bogo Chies
- Laboratory of Immunobiology and Immunogenetics, Graduate Program in Genetics and Molecular Biology - PPGBM, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil.
| |
Collapse
|
82
|
Wang Z, Cui Y, Zhang Y, Wang X, Li J, Li J, Jiang N. Twelve-week treadmill endurance training in mice is associated with upregulation of interleukin-15 and natural killer cell activation and increases apoptosis rate in Hepa1-6 cell-derived mouse hepatomas. Braz J Med Biol Res 2023; 56:e12296. [PMID: 37585912 PMCID: PMC10427160 DOI: 10.1590/1414-431x2023e12296] [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: 03/05/2023] [Accepted: 06/23/2023] [Indexed: 08/18/2023] Open
Abstract
Regular exercise reduces the risk of malignancy and decreases the recurrence of cancer. However, the mechanisms behind this protection remain to be elucidated. Natural killer (NK) cells are lymphocytes of the innate immune system, which play essential roles in immune defense and effectively prevent cancer metastasis. Physical exercise can increase the activity of NK cells. Interleukin-15 (IL-15) is the best-studied cytokine activator of NK cells, and it was shown to have many positive functional effects on NK cells to improve antitumor responses. The aim of this study was to clarify the possible important mechanisms behind endurance exercise-induced changes in NK cell function, which may be highly correlated with IL-15. An animal model was used to study IL-15 expression level, tumor volume, cancer cell apoptosis, and NK cell infiltration after treadmill exercise. Although IL-15 was highly expressed in skeletal muscle, treadmill exercise further elevated IL-15 levels in plasma and muscle (P<0.05). In addition, tumor weight and volume of tumor-bearing mice were decreased (P<0.05), and liver tumor cell apoptosis was increased after 12 weeks of treadmill exercise (P<0.05). NK cell infiltration was upregulated in tumors from treadmill exercise mice, and the level of interferon-gamma (IFN-γ) and IL-15 were higher than in sedentary mice (P<0.05). The study indicated that regular endurance training can reduce cancer risk, which was related to increased IL-15 expression, activation of the immune killing effect of NK cells, and promotion of tumor cell apoptosis, which can ultimately control tumor growth.
Collapse
Affiliation(s)
- Zhe Wang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise & Health, Tianjin University of Sport, Tianjin, China
- Department of Common Subject, College of Basic Sciences, Logistics College of Chinese People’s Armed Police Force, Tianjin, China
| | - Yunlong Cui
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Hospital, Tianjin, China
| | - Yong Zhang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise & Health, Tianjin University of Sport, Tianjin, China
| | - Xinghao Wang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise & Health, Tianjin University of Sport, Tianjin, China
| | - Jing Li
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise & Health, Tianjin University of Sport, Tianjin, China
| | - Jialin Li
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise & Health, Tianjin University of Sport, Tianjin, China
| | - Ning Jiang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise & Health, Tianjin University of Sport, Tianjin, China
| |
Collapse
|
83
|
Yun HD, Goel Y, Gupta K. Crosstalk of Mast Cells and Natural Killer Cells with Neurons in Chemotherapy-Induced Peripheral Neuropathy. Int J Mol Sci 2023; 24:12543. [PMID: 37628724 PMCID: PMC10454469 DOI: 10.3390/ijms241612543] [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: 07/11/2023] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a major comorbidity of cancer. Multiple clinical interventions have been studied to effectively treat CIPN, but the results have been disappointing, with no or little efficacy. Hence, understanding the pathophysiology of CIPN is critical to improving the quality of life and clinical outcomes of cancer patients. Although various mechanisms of CIPN have been described in neuropathic anti-cancer agents, the neuroinflammatory process involving cytotoxic/proinflammatory immune cells remains underexamined. While mast cells (MCs) and natural killer (NK) cells are the key innate immune compartments implicated in the pathogenesis of peripheral neuropathy, their role in CIPN has remained under-appreciated. Moreover, the biology of proinflammatory cytokines associated with MCs and NK cells in CIPN is particularly under-evaluated. In this review, we will focus on the interactions between MCs, NK cells, and neuronal structure and their communications via proinflammatory cytokines, including TNFα, IL-1β, and IL-6, in peripheral neuropathy in association with tumor immunology. This review will help lay the foundation to investigate MCs, NK cells, and cytokines to advance future therapeutic strategies for CIPN.
Collapse
Affiliation(s)
- Hyun Don Yun
- Hematology, Oncology, Veterans Affairs Long Beach Healthcare System, Long Beach, CA 90822, USA
- Division of Hematology, Oncology, Department of Medicine, School of Medicine, University of California, Irvine, CA 92617, USA; (Y.G.); (K.G.)
| | - Yugal Goel
- Division of Hematology, Oncology, Department of Medicine, School of Medicine, University of California, Irvine, CA 92617, USA; (Y.G.); (K.G.)
| | - Kalpna Gupta
- Division of Hematology, Oncology, Department of Medicine, School of Medicine, University of California, Irvine, CA 92617, USA; (Y.G.); (K.G.)
| |
Collapse
|
84
|
Wang T, Li P, Qi Q, Zhang S, Xie Y, Wang J, Liu S, Ma S, Li S, Gong T, Xu H, Xiong M, Li G, You C, Luo Z, Li J, Du L, Wang C. A multiplex blood-based assay targeting DNA methylation in PBMCs enables early detection of breast cancer. Nat Commun 2023; 14:4724. [PMID: 37550304 PMCID: PMC10406825 DOI: 10.1038/s41467-023-40389-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 07/21/2023] [Indexed: 08/09/2023] Open
Abstract
The immune system can monitor tumor development, and DNA methylation is involved in the body's immune response to tumors. In this work, we investigate whether DNA methylation alterations in peripheral blood mononuclear cells (PBMCs) could be used as markers for early detection of breast cancer (BC) from the perspective of tumor immune alterations. We identify four BC-specific methylation markers by combining Infinium 850 K BeadChips, pyrosequencing and targeted bisulfite sequencing. Based on the four methylation markers in PBMCs of BC, we develop an efficient and convenient multiplex methylation-specific quantitative PCR assay for the detection of BC and validate its diagnostic performance in a multicenter cohort. This assay was able to distinguish early-stage BC patients from normal controls, with an AUC of 0.940, sensitivity of 93.2%, and specificity of 90.4%. More importantly, this assay outperformed existing clinical diagnostic methods, especially in the detection of early-stage and minimal tumors.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China
| | - Peilong Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China
| | - Qiuchen Qi
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China
| | - Shujun Zhang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China
| | - Yan Xie
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China
| | - Jing Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China
| | - Shibiao Liu
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China
| | - Suhong Ma
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China
| | - Shijun Li
- Clinical Laboratory, The First Hospital of Dalian Medical University, Dalian, 116011, P. R. China
| | - Tingting Gong
- Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China
| | - Huiting Xu
- Departmemt of Clinical Laboratory Medicine, Affiliated Tumor Hospital of Nantong University, 226361, Jiangsu, China; Medical School of Nantong University, Nantong, 226001, P. R. China
| | - Mengqiu Xiong
- Clinical Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, P. R. China
| | - Guanghua Li
- Department of clinical laboratory, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou, 510000, P. R. China
| | - Chongge You
- Laboratory Medicine Center, Lanzhou University Second Hospital, the Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, P. R. China
| | - Zhaofan Luo
- Department of Clinical Laboratory, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, P. R. China
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China.
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China.
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Shandong Provincial Key Laboratory of Innovation Technology in Laboratory Medicine, Jinan, 250012, P. R. China.
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, 250033, Shandong, China.
- Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, 250033, China.
- Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, 250033, China.
| |
Collapse
|
85
|
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
|
86
|
Wang M, Gong K, Zhu X, Chen S, Zhou J, Zhang H, Han J, Ma L, Duan Y. Identification of circulating T-cell immunoglobulin and mucin domain 4 as a potential biomarker for coronary heart disease. MedComm (Beijing) 2023; 4:e320. [PMID: 37426678 PMCID: PMC10329472 DOI: 10.1002/mco2.320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 05/27/2023] [Accepted: 06/01/2023] [Indexed: 07/11/2023] Open
Abstract
Efferocytosis, the process of engulfing and removing apoptotic cells, is attenuated in vulnerable plaques of advanced atherosclerosis. T-cell immunoglobulin and mucin domain 4 (TIMD4) is a recognition receptor protein for efferocytosis that has been implicated in atherosclerosis mouse models. However, the role of serum-soluble TIMD4 (sTIMD4) in coronary heart disease (CHD) remains unknown. In this study, we analyzed serum samples collected from two groups: Group 1 (36 healthy controls and 70 CHD patients) and Group 2 (44 chronic coronary syndrome [CCS]) and 81 acute coronary syndrome [ACS] patients). We found that sTIMD4 levels in patients with CHD were significantly higher than those in healthy controls and were also higher in ACS than in CCS patients. The area under the receiver operating characteristic curve was 0.787. Furthermore, our in vitro results showed that low-density lipoprotein/lipopolysaccharide activated p38 mitogen-activated protein kinase, which in turn enhanced a disintegrin and metalloproteinase 17, resulting in increased secretion of sTIMD4. This impairment of macrophage efferocytosis promoted inflammation. Thus, this study is not only the first identification of a potential novel biomarker of CHD, sTIMD4, but also demonstrated its pathogenesis mechanism, providing a new direction for the diagnosis and treatment of CHD.
Collapse
Affiliation(s)
- Mengyao Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological EngineeringHefei University of TechnologyHefeiChina
| | - Ke Gong
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological EngineeringHefei University of TechnologyHefeiChina
| | - Xinran Zhu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological EngineeringHefei University of TechnologyHefeiChina
| | - Shasha Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological EngineeringHefei University of TechnologyHefeiChina
| | - Jie Zhou
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological EngineeringHefei University of TechnologyHefeiChina
| | - Hui Zhang
- Department of CardiologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Jihong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological EngineeringHefei University of TechnologyHefeiChina
- Key Laboratory of Bioactive Materials of Ministry of EducationCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyNankai UniversityTianjinChina
| | - Likun Ma
- Department of CardiologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Yajun Duan
- Department of CardiologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| |
Collapse
|
87
|
Sacramento LA, Farias Amorim C, Campos TM, Saldanha M, Arruda S, Carvalho LP, Beiting DP, Carvalho EM, Novais FO, Scott P. NKG2D promotes CD8 T cell-mediated cytotoxicity and is associated with treatment failure in human cutaneous leishmaniasis. PLoS Negl Trop Dis 2023; 17:e0011552. [PMID: 37603573 PMCID: PMC10470908 DOI: 10.1371/journal.pntd.0011552] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/31/2023] [Accepted: 07/25/2023] [Indexed: 08/23/2023] Open
Abstract
Cutaneous leishmaniasis exhibits a spectrum of clinical presentations dependent upon the parasites' persistence and host immunopathologic responses. Although cytolytic CD8 T cells cannot control the parasites, they significantly contribute to pathologic responses. In a murine model of cutaneous leishmaniasis, we previously found that NKG2D plays a role in the ability of cytolytic CD8 T cells to promote disease in leishmanial lesions. Here, we investigated whether NKG2D plays a role in human disease. We found that NKG2D and its ligands were expressed within lesions from L. braziliensis-infected patients and that IL-15 and IL-1β were factors driving NKG2D and NKG2D ligand expression, respectively. Blocking NKG2D reduced degranulation by CD8 T cells in a subset of patients. Additionally, our transcriptional analysis of patients' lesions found that patients who failed the first round of treatment exhibited higher expression of KLRK1, the gene coding for NKG2D, than those who responded to treatment. These findings suggest that NKG2D may be a promising therapeutic target for ameliorating disease severity in cutaneous leishmaniasis caused by L. braziliensis infection.
Collapse
Affiliation(s)
- Laís A. Sacramento
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Camila Farias Amorim
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Taís M. Campos
- Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Maíra Saldanha
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Sérgio Arruda
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Lucas P. Carvalho
- Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| | - Daniel P. Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Edgar M. Carvalho
- Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| | - Fernanda O. Novais
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus,Ohio, United States of America
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
88
|
Liu J, Ye L, Lin K, Zhong T, Luo J, Wang T, Suo L, Mo Q, Li S, Chen Q, Yu Y. miR-4299 inhibits tumor progression in pancreatic cancer through targeting ADAM17. Mol Cell Biochem 2023; 478:1727-1742. [PMID: 36565360 DOI: 10.1007/s11010-022-04617-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 11/18/2022] [Indexed: 12/25/2022]
Abstract
Pancreatic cancer (PC) is one of the most aggressive malignant tumors in human beings. Tumor capacity of evading immune-mediated lysis is a critical step in PC malignant progression. We aimed to evaluate the underlying regulatory mechanism of miR-4299 in the proliferation, metastasis, apoptosis, and immune escape in PC. miR-4299 and ADAM17 expressions in PC tissues and cell lines were detected using qRT-PCR. MTT assay and flow cytometry were used to detect cell viability and apoptosis, respectively. A luciferase reporter gene assay was conducted to confirm the targeted relationship between miR-4299 and ADAM17. Xenograft tumors in nude mice were used to detect tumorigenesis in vivo. PC cells were co-cultured with NK cells for determining the immune escape ability. NKG2D-positive rate of NK cells was detected using flow cytometry; NK cell-killing ability was detected using MTT assay. miR-4299 was downregulated in PC tissues and cell lines. miR-4299 inhibited PC cell proliferation and invasion, promoted cell apoptosis, and reduced PC tumor growth in vivo. ADAM17 3'UTR directly bound to miR-4299. ADAM17 overexpression could reverse miR-4299 effects on PC cell viability, invasion, apoptosis, and immune escape. miR-4299 exerted suppressive effects on PC cell proliferation, invasion, and immune escape via targeting ADAM17 expression. This study revealed a novel miR-4299/ADAM17 axis-modulating PC progression and proposed to concern the immune regulatory mechanism of miRNAs in PC development.
Collapse
Affiliation(s)
- Junhong Liu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541002, China
| | - Lin Ye
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541002, China
| | - Kangqiang Lin
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541002, China
| | - Tieshan Zhong
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541002, China
| | - Jiguang Luo
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541002, China
| | - Tao Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541002, China
| | - Liya Suo
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541002, China
| | - Qingrong Mo
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541002, China
| | - Shuqun Li
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541002, China
| | - Qian Chen
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541002, China
| | - Yaqun Yu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541002, China.
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, The Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China.
| |
Collapse
|
89
|
Ramanathan G, Chen JH, Mehrotra N, Trieu T, Huang A, Mas E, Monterrosa Mena JE, Bliss B, Herman DA, Kleinman MT, Fleischman AG. Cigarette smoke stimulates clonal expansion of Jak2 V617F and Tet2 -/- cells. Front Oncol 2023; 13:1210528. [PMID: 37546389 PMCID: PMC10401270 DOI: 10.3389/fonc.2023.1210528] [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: 04/22/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction Somatic mutations in myeloid growth factor pathway genes, such as JAK2, and genes involved in epigenetic regulation, such as TET2, in hematopoietic stem cells (HSCs) leads to clonal hematopoiesis of indeterminate potential (CHIP) which presents a risk factor for hematologic malignancy and cardiovascular disease. Smoking behavior has been repeatedly associated with the occurrence of CHIP but whether smoking is an environmental inflammatory stressor in promoting clonal expansion has not been investigated. Methods We performed in vivo smoke exposures in both wildtype (WT) mice and transplanted mice carrying Jak2V617F mutant and Tet2 knockout (Tet-/-) cells to determine the impact of cigarette smoke (CS) in the HSC compartment as well as favoring mutant cell expansion. Results WT mice exposed to smoke displayed increased oxidative stress in long-term HSCs and suppression of the hematopoietic stem and progenitor compartment but smoke exposure did not translate to impaired hematopoietic reconstitution in primary bone marrow transplants. Gene expression analysis of hematopoietic cells in the bone marrow identified an imbalance between Th17 and Treg immune cells suggesting a local inflammatory environment. We also observed enhanced survival of Jak2V617F cells exposed to CS in vivo and cigarette smoke extract (CSE) in vitro. WT bone marrow hematopoietic cells from WT/Jak2V617F chimeric mice exposed to CS demonstrated an increase in neutrophil abundance and distinct overexpression of bone marrow stromal antigen 2 (Bst2) and retinoic acid early transcript 1 (Raet1) targets. Bst2 and Raet1 are indicative of increased interferon signaling and cellular stress including oxidative stress and DNA damage, respectively. In chimeric mice containing both WT and Tet2-/- cells, we observed an increased percentage of circulating mutant cells in peripheral blood post-cigarette smoke exposure when compared to pre-exposure levels while this difference was absent in air-exposed controls. Conclusion Altogether, these findings demonstrate that CS results in an inflamed bone marrow environment that provides a selection pressure for existing CHIP mutations such as Jak2V617F and Tet2 loss-of-function.
Collapse
Affiliation(s)
- Gajalakshmi Ramanathan
- Department of Medicine, Division of Hematology/Oncology, University of California, Irvine, Irvine, CA, United States
| | - Jane H. Chen
- Department of Medicine, Division of Hematology/Oncology, University of California, Irvine, Irvine, CA, United States
| | - Nitya Mehrotra
- Department of Medicine, Division of Hematology/Oncology, University of California, Irvine, Irvine, CA, United States
| | - Tiffany Trieu
- Department of Medicine, Division of Hematology/Oncology, University of California, Irvine, Irvine, CA, United States
| | - Aaron Huang
- Department of Medicine, Division of Hematology/Oncology, University of California, Irvine, Irvine, CA, United States
| | - Eduard Mas
- Department of Medicine, Division of Hematology/Oncology, University of California, Irvine, Irvine, CA, United States
| | - Jessica E. Monterrosa Mena
- Department of Medicine, Division of Occupational and Environmental Medicine, University of California, Irvine, Irvine, CA, United States
| | - Bishop Bliss
- Department of Medicine, Division of Occupational and Environmental Medicine, University of California, Irvine, Irvine, CA, United States
| | - David A. Herman
- Department of Medicine, Division of Occupational and Environmental Medicine, University of California, Irvine, Irvine, CA, United States
| | - Michael T. Kleinman
- Department of Medicine, Division of Occupational and Environmental Medicine, University of California, Irvine, Irvine, CA, United States
| | - Angela G. Fleischman
- Department of Medicine, Division of Hematology/Oncology, University of California, Irvine, Irvine, CA, United States
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
90
|
Goulding J, Yeh WI, Hancock B, Blum R, Xu T, Yang BH, Chang CW, Groff B, Avramis E, Pribadi M, Pan Y, Chu HY, Sikaroodi S, Fong L, Brookhouser N, Dailey T, Meza M, Denholtz M, Diaz E, Martin J, Szabo P, Cooley S, Ferrari de Andrade L, Lee TT, Bjordahl R, Wucherpfennig KW, Valamehr B. A chimeric antigen receptor uniquely recognizing MICA/B stress proteins provides an effective approach to target solid tumors. MED 2023; 4:457-477.e8. [PMID: 37172578 PMCID: PMC10524375 DOI: 10.1016/j.medj.2023.04.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/16/2023] [Accepted: 04/13/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND The advent of chimeric antigen receptor (CAR) T cell therapies has transformed the treatment of hematological malignancies; however, broader therapeutic success of CAR T cells has been limited in solid tumors because of their frequently heterogeneous composition. Stress proteins in the MICA and MICB (MICA/B) family are broadly expressed by tumor cells following DNA damage but are rapidly shed to evade immune detection. METHODS We have developed a novel CAR targeting the conserved α3 domain of MICA/B (3MICA/B CAR) and incorporated it into a multiplexed-engineered induced pluripotent stem cell (iPSC)-derived natural killer (NK) cell (3MICA/B CAR iNK) that expressed a shedding-resistant form of the CD16 Fc receptor to enable tumor recognition through two major targeting receptors. FINDINGS We demonstrated that 3MICA/B CAR mitigates MICA/B shedding and inhibition via soluble MICA/B while simultaneously exhibiting antigen-specific anti-tumor reactivity across an expansive library of human cancer cell lines. Pre-clinical assessment of 3MICA/B CAR iNK cells demonstrated potent antigen-specific in vivo cytolytic activity against both solid and hematological xenograft models, which was further enhanced in combination with tumor-targeted therapeutic antibodies that activate the CD16 Fc receptor. CONCLUSIONS Our work demonstrated 3MICA/B CAR iNK cells to be a promising multi-antigen-targeting cancer immunotherapy approach intended for solid tumors. FUNDING Funded by Fate Therapeutics and NIH (R01CA238039).
Collapse
Affiliation(s)
| | - Wen-I Yeh
- Fate Therapeutics Inc., San Diego, CA 92131, USA
| | | | - Robert Blum
- Fate Therapeutics Inc., San Diego, CA 92131, USA
| | - Tianhao Xu
- Fate Therapeutics Inc., San Diego, CA 92131, USA
| | - Bi-Huei Yang
- Fate Therapeutics Inc., San Diego, CA 92131, USA
| | | | - Brian Groff
- Fate Therapeutics Inc., San Diego, CA 92131, USA
| | - Earl Avramis
- Fate Therapeutics Inc., San Diego, CA 92131, USA
| | | | - Yijia Pan
- Fate Therapeutics Inc., San Diego, CA 92131, USA
| | - Hui-Yi Chu
- Fate Therapeutics Inc., San Diego, CA 92131, USA
| | | | - Lauren Fong
- Fate Therapeutics Inc., San Diego, CA 92131, USA
| | | | | | - Miguel Meza
- Fate Therapeutics Inc., San Diego, CA 92131, USA
| | | | - Evelyn Diaz
- Fate Therapeutics Inc., San Diego, CA 92131, USA
| | - Judy Martin
- Fate Therapeutics Inc., San Diego, CA 92131, USA
| | - Peter Szabo
- Fate Therapeutics Inc., San Diego, CA 92131, USA
| | - Sarah Cooley
- Fate Therapeutics Inc., San Diego, CA 92131, USA
| | | | - Tom T Lee
- Fate Therapeutics Inc., San Diego, CA 92131, USA
| | | | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Neurology, Brigham & Women's Hospital, Boston, MA 02115, USA; Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
91
|
Imširović V, Lenartić M, Wensveen FM, Polić B, Jelenčić V. Largely preserved functionality after the combined loss of NKG2D, NCR1 and CD16 demonstrates the remarkable plasticity of NK cell responsiveness. Front Immunol 2023; 14:1191884. [PMID: 37520575 PMCID: PMC10374020 DOI: 10.3389/fimmu.2023.1191884] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Natural killer (NK) cells play an important role in the early defense against tumors and virally infected cells. Their function is thought to be controlled by the balance between activating and inhibitory receptors, which often compete for the same ligands. Several activating receptors expressed on virtually all NK cells lack an inhibitory partner, most notably CD16, NCR1 and NKG2D. We therefore hypothesized that a signal through at least one of these receptors is always required for full NK cell activation. We generated animals lacking all three receptors (TKO) and analyzed their NK cells. In vitro, TKO NK cells did not show reduced ability to kill tumor targets but displayed hyperresponsiveness to NK1.1 stimulation. In vivo, TKO animals had a minor reduction in their ability to control non-hematopoietic tumors and cytomegalovirus infection, which was the result of reduced NK cell activity. Together, our findings show that activating NK cell receptors without an inhibitory partner do not provide a 'master' signal but are integrated in the cumulative balance of activating and inhibitory signals. Their activity is controlled through regulation of the responsiveness and expression of other activating receptors. Our findings may be important for future development of NK cell-based cancer immunotherapy.
Collapse
|
92
|
Wen J, Chen Y, Yang J, Dai C, Yu S, Zhong W, Liu L, He C, Zhang W, Yang T, Liu L, Hu J. Valproic acid increases CAR T cell cytotoxicity against acute myeloid leukemia. J Immunother Cancer 2023; 11:e006857. [PMID: 37524506 PMCID: PMC10391797 DOI: 10.1136/jitc-2023-006857] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2023] [Indexed: 08/02/2023] Open
Abstract
The treatment of B cell malignancies has dramatically changed with the introduction of immunotherapy, especially chimeric antigen receptor T (CAR-T) cell therapy. However, only limited efficacy is observed in acute myeloid leukaemia (AML). In the study, We detected CD123 and CLL-1 expression on leukaemia cells from Relapsed/Refractory AML (R/R AML) patients. Then, we constructed anti-CD123 CAR and CLL-1 CAR with different co-stimulation domains (CD28 or 4-1BB) and detected their anti-AML effects. To increase the efficacy of CAR-T cell therapy, we tested different strategies, including application of combined checkpoint inhibitors and histone deacetylase inhibitors (HDACi) in vivo and in vitro We found CD123 and CLL-1 were highly expressed on AML cells. The proportions of T cell subsets and NK cells involved in anti-tumour or anti-inflammation processes in AML patients significantly decreased when compared with healthy donors. Both CD123 CAR and CLL-1 CAR displayed specific anti-AML effects in vitro To improve the lysis effects of CAR-T cells, we combined CAR-T cell therapy with different agents. PD-1/PD-L1 antibodies only slightly improved the potency of CAR-T cell therapy (CD123 CAR-T 60.92% ± 2.9087% vs. 65.43% ± 2.1893%, 60.92% ± 2.9087% vs. 67.43% ± 3.4973%; 37.37% ± 3.908% vs. 41.89% ± 5.1568%, 37.37% ± 3.908% vs. 42.84% ± 4.2635%). However, one HDACi (valproic acid [VPA]) significantly improved CAR-T cell potency against AML cells (CLL-1 CAR-T 34.97% ± 0.3051% vs. 88.167% ± 1.5327%, p < 0.0001; CD123 CAR-T 26.87% ± 2.7010% vs. 82.56% ± 3.086%, p < 0.0001 in MV411; CLL-1 CAR-T 78.77% ± 1.2061% vs. 93.743% ± 1.2333%, p < 0.0001; CD123 CAR-T 64.10% ± 1.5130% vs. 94.427% ± 0.142%, p = 0.0001 in THP-1). Combination therapy prolonged the overall survival of mice when compared with single CD123 CAR-T cell therapy (median survival: 180 days vs. unfollowed). A possible mechanism is that activated CD8+T cells upregulate natural-killer group 2 member D (NKG2D), and VPA upregulates NKG2D ligand expression in AML cells, contributing to NKG2D-mediated cytotoxicity of CAR-T cells against tumour cells. In conclusion, CD123 and CLL-1 are promising targets for AML CAR-T cell therapy. A combination of VPA pre-treatment and CAR-T against AML exhibits synergic effects.
Collapse
MESH Headings
- Animals
- Mice
- Valproic Acid/pharmacology
- Valproic Acid/therapeutic use
- Receptors, Chimeric Antigen/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- NK Cell Lectin-Like Receptor Subfamily K/metabolism
- Cell Line, Tumor
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- T-Lymphocytes
Collapse
Affiliation(s)
- Jingjing Wen
- Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou, China
- Department of Lymphoma, Fujian Cancer Hospital, Fuzhou, People's Republic of China
| | - Yanxin Chen
- Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou, China
| | - Jiajie Yang
- Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou, China
- Institute of Precision Medicine, Fujian Medical University, Fuzhou, People's Republic of China
| | - Chunye Dai
- School of Life Sciences, Fudan University, Shanghai, China
| | - Shenjie Yu
- Department of Internal medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Wenting Zhong
- Department of Research and Development, ST Phi Therapeutics Co., Ltd, Hangzhou, China
| | - Lilin Liu
- Department of Research and Development, ST Phi Therapeutics Co., Ltd, Hangzhou, China
| | - Chengguanng He
- Department of Research and Development, ST Phi Therapeutics Co., Ltd, Hangzhou, China
| | - Wenmin Zhang
- Institute of Precision Medicine, Fujian Medical University, Fuzhou, People's Republic of China
- Pathological Diagnosis Center & Oncology Institution, Fujian Medical University, Fuzhou, China
| | - Ting Yang
- Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou, China
| | - Lingfeng Liu
- School of Life Sciences, Fudan University, Shanghai, China
| | - Jianda Hu
- Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou, China
- Institute of Precision Medicine, Fujian Medical University, Fuzhou, People's Republic of China
| |
Collapse
|
93
|
Liu M, Hu Z, Wang C, Zhang Y. The TLR/MyD88 signalling cascade in inflammation and gastric cancer: the immune regulatory network of Helicobacter pylori. J Mol Med (Berl) 2023; 101:767-781. [PMID: 37195446 DOI: 10.1007/s00109-023-02332-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/18/2023]
Abstract
Helicobacter pylori-induced chronic gastritis represents a well-established risk factor for gastric cancer (GC). However, the mechanism by which chronic inflammation caused by H. pylori induces the development of GC is unclear. H. pylori can influence host cell signalling pathways to induce gastric disease development and mediate cancer promotion and progression. Toll-like receptors (TLRs), as pattern recognition receptors (PRRs), play a key role in the gastrointestinal innate immune response, and their signalling has been implicated in the pathogenesis of an increasing number of inflammation-associated cancers. The core adapter myeloid differentiation factor-88 (MyD88) is shared by most TLRs and functions primarily in H. pylori-triggered innate immune signalling. MyD88 is envisioned as a potential target for the regulation of immune responses and is involved in the regulation of tumourigenesis in a variety of cancer models. In recent years, the TLR/MyD88 signalling pathway has received increasing attention for its role in regulating innate and adaptive immune responses, inducing inflammatory activation and promoting tumour formation. In addition, TLR/MyD88 signalling can manipulate the expression of infiltrating immune cells and various cytokines in the tumour microenvironment (TME). In this review, we discuss the pathogenetic regulatory mechanisms of the TLR/MyD88 signalling cascade pathway and its downstream molecules in H. pylori infection-induced-associated GC. The focus is to elucidate the immunomolecular mechanisms of pathogen recognition and innate immune system activation of H. pylori in the TME of inflammation-associated GC. Ultimately, this study will provide insight into the mechanism of H. pylori-induced chronic inflammation-induced GC development and provide thoughts for GC prevention and treatment strategies.
Collapse
Affiliation(s)
- Meiqi Liu
- Medical School, Cancer Research Institute, University of South China, Chang Sheng Xi Avenue 28, Hengyang City, Hunan, 421001, China
| | - Zhizhong Hu
- Medical School, Cancer Research Institute, University of South China, Chang Sheng Xi Avenue 28, Hengyang City, Hunan, 421001, China
| | - Chengkun Wang
- Medical School, Cancer Research Institute, University of South China, Chang Sheng Xi Avenue 28, Hengyang City, Hunan, 421001, China.
| | - Yang Zhang
- Medical School, Cancer Research Institute, University of South China, Chang Sheng Xi Avenue 28, Hengyang City, Hunan, 421001, China.
| |
Collapse
|
94
|
Xiao H, Lin R, Chen C, Lian R, Wu Y, Diao L, Yin T, Huang C. γδ-T cell with high toxic potential was associated with recurrent miscarriage. Am J Reprod Immunol 2023; 90:e13717. [PMID: 37382173 DOI: 10.1111/aji.13717] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 06/30/2023] Open
Abstract
PROBLEM RM is a common clinical disease in reproduction, affecting approximately 1%-3% of women worldwide. Previous studies have shown the role of peripheral blood γδ-T cells during physiological pregnancy. However, the relationship between the immune status of peripheral blood γδ-T cells and RM is still not well defined. METHOD OF STUDY In this study, mid-luteal peripheral blood from 51 RM patients and 40 healthy women was collected to determine the immune status of γδ-T cells. The percentage of peripheral blood γδ-T cells, and the molecules mediating their toxic potential, including cytotoxic granules (perforin, granzyme B, and granulysin) and receptors (NKG2D, CD158a, and CD158b), were detected by flow cytometry. RESULTS Compared to healthy control, an increase in the proportion of total CD3+ T cells in lymphocytes and a decrease in the ratio of γδ-T cells to CD3+ T cells were observed in patients with RM. The percentages of granzyme B+ γδ-T cells and CD158a+ γδ-T cells in total γδ-T cells or lymphocytes were significantly increased in patients with RM, compared with healthy control. Conversely, CD158b+ γδ-T cells in total γδ-T cells or lymphocytes were significantly decreased in the RM group. CONCLUSION Increased peripheral blood γδ-T cell with high toxic potential was associated with RM.
Collapse
Affiliation(s)
- Huan Xiao
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Rong Lin
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, Guangdong, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China
| | - Cong Chen
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, Guangdong, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China
| | - Ruochun Lian
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, Guangdong, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China
| | - Yulian Wu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, Guangdong, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China
| | - Lianghui Diao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, Guangdong, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China
| | - Tailang Yin
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chunyu Huang
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, Guangdong, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China
| |
Collapse
|
95
|
Shen C, Li M, Duan Y, Jiang X, Hou X, Xue F, Zhang Y, Luo Y. HDAC inhibitors enhance the anti-tumor effect of immunotherapies in hepatocellular carcinoma. Front Immunol 2023; 14:1170207. [PMID: 37304265 PMCID: PMC10250615 DOI: 10.3389/fimmu.2023.1170207] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/18/2023] [Indexed: 06/13/2023] Open
Abstract
Hepatocellular carcinoma (HCC), the most common liver malignancy with a poor prognosis and increasing incidence, remains a serious health problem worldwide. Immunotherapy has been described as one of the ideal ways to treat HCC and is transforming patient management. However, the occurrence of immunotherapy resistance still prevents some patients from benefiting from current immunotherapies. Recent studies have shown that histone deacetylase inhibitors (HDACis) can enhance the efficacy of immunotherapy in a variety of tumors, including HCC. In this review, we present current knowledge and recent advances in immunotherapy-based and HDACi-based therapies for HCC. We highlight the fundamental dynamics of synergies between immunotherapies and HDACis, further detailing current efforts to translate this knowledge into clinical benefits. In addition, we explored the possibility of nano-based drug delivery system (NDDS) as a novel strategy to enhance HCC treatment.
Collapse
Affiliation(s)
- Chen Shen
- Department of Laboratory Medicine, Medical Equipment Innovation Research Center/Medical Device Regulatory Research and Evaluation Center, West China Hospital, Sichuan University, Chengdu, China
| | - Mei Li
- Department of Laboratory Medicine, Medical Equipment Innovation Research Center/Medical Device Regulatory Research and Evaluation Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yujuan Duan
- School of Chemical Science and Engineering, Tongji University, Shanghai, China
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Jiang
- Department of Laboratory Medicine, Medical Equipment Innovation Research Center/Medical Device Regulatory Research and Evaluation Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoming Hou
- Department of Laboratory Medicine, Medical Equipment Innovation Research Center/Medical Device Regulatory Research and Evaluation Center, West China Hospital, Sichuan University, Chengdu, China
| | - Fulai Xue
- Department of Laboratory Medicine, Medical Equipment Innovation Research Center/Medical Device Regulatory Research and Evaluation Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yinan Zhang
- School of Chemical Science and Engineering, Tongji University, Shanghai, China
| | - Yao Luo
- Department of Laboratory Medicine, Medical Equipment Innovation Research Center/Medical Device Regulatory Research and Evaluation Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
96
|
Molfetta R, Petillo S, Cippitelli M, Paolini R. SUMOylation and related post-translational modifications in natural killer cell anti-cancer responses. Front Cell Dev Biol 2023; 11:1213114. [PMID: 37313439 PMCID: PMC10258607 DOI: 10.3389/fcell.2023.1213114] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 05/17/2023] [Indexed: 06/15/2023] Open
Abstract
SUMOylation is a reversible modification that involves the covalent attachment of small ubiquitin-like modifier (SUMO) to target proteins, leading to changes in their localization, function, stability, and interactor profile. SUMOylation and additional related post-translational modifications have emerged as important modulators of various biological processes, including regulation of genomic stability and immune responses. Natural killer (NK) cells are innate immune cells that play a critical role in host defense against viral infections and tumors. NK cells can recognize and kill infected or transformed cells without prior sensitization, and their activity is tightly regulated by a balance of activating and inhibitory receptors. Expression of NK cell receptors as well as of their specific ligands on target cells is finely regulated during malignant transformation through the integration of different mechanisms including ubiquitin- and ubiquitin-like post-translational modifications. Our review summarizes the role of SUMOylation and other related pathways in the biology of NK cells with a special emphasis on the regulation of their response against cancer. The development of novel selective inhibitors as useful tools to potentiate NK-cell mediated killing of tumor cells is also briefly discussed.
Collapse
|
97
|
Wu C, Spector SA, Theodoropoulos G, Nguyen DJM, Kim EY, Garcia A, Savaraj N, Lim DC, Paul A, Feun LG, Bickerdike M, Wangpaichitr M. Dual inhibition of IDO1/TDO2 enhances anti-tumor immunity in platinum-resistant non-small cell lung cancer. Cancer Metab 2023; 11:7. [PMID: 37226257 DOI: 10.1186/s40170-023-00307-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 05/10/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND The impact of non-small cell lung cancer (NSCLC) metabolism on the immune microenvironment is not well understood within platinum resistance. We have identified crucial metabolic differences between cisplatin-resistant (CR) and cisplatin-sensitive (CS) NSCLC cells with elevated indoleamine 2,3-dioxygenase-1 (IDO1) activity in CR, recognized by increased kynurenine (KYN) production. METHODS Co-culture, syngeneic, and humanize mice models were utilized. C57BL/6 mice were inoculated with either Lewis lung carcinoma mouse cells (LLC) or their platinum-resistant counterpart (LLC-CR) cells. Humanized mice were inoculated with either A (human CS cells) or ALC (human CR cells). Mice were treated with either IDO1 inhibitor or TDO2 (tryptophan 2,3-dioxygenase-2) inhibitor at 200 mg/kg P.O. once a day for 15 days; or with a new-in-class, IDO1/TDO2 dual inhibitor AT-0174 at 170 mg/kg P.O. once a day for 15 days with and without anti-PD1 antibody (10 mg/kg, every 3 days). Immune profiles and KYN and tryptophan (TRP) production were evaluated. RESULTS CR tumors exhibited a more highly immunosuppressive environment that debilitated robust anti-tumor immune responses. IDO1-mediated KYN production from CR cells suppressed NKG2D on immune effector natural killer (NK) and CD8+ T cells and enhanced immunosuppressive populations of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). Importantly, while selective IDO1 inhibition attenuated CR tumor growth, it concomitantly upregulated the TDO2 enzyme. To overcome the compensatory induction of TDO2 activity, we employed the IDO1/TDO2 dual inhibitor, AT-0174. Dual inhibition of IDO1/TDO2 in CR mice suppressed tumor growth to a greater degree than IDO1 inhibition alone. Significant enhancement in NKG2D frequency on NK and CD8+ T cells and a reduction in Tregs and MDSCs were observed following AT-1074 treatment. PD-L1 (programmed death-ligand-1) expression was increased in CR cells; therefore, we assessed dual inhibition + PD1 (programmed cell death protein-1) blocking and report profound anti-tumor growth and improved immunity in CR tumors which in turn extended overall survival in mice. CONCLUSION Our study reports the presence of platinum-resistant lung tumors that utilize both IDO1/TDO2 enzymes for survival, and to escape immune surveillance as a consequence of KYN metabolites. We also report early in vivo data in support of the potential therapeutic efficacy of the dual IDO1/TDO2 inhibitor AT-0174 as a part of immuno-therapeutic treatment that disrupts tumor metabolism and enhances anti-tumor immunity.
Collapse
Affiliation(s)
- Chunjing Wu
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA
| | - Sydney A Spector
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA
| | | | - Dan J M Nguyen
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA
| | - Emily Y Kim
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA
| | - Ashley Garcia
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA
| | - Niramol Savaraj
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA
- Department of Medicine, University of Miami School of Medicine, Miami, FL, USA
| | - Diane C Lim
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA
- Department of Medicine, University of Miami School of Medicine, Miami, FL, USA
| | - Ankita Paul
- Department of Electrical and Computer Engineering, Drexel University, Philadelphia, PA, USA
| | - Lynn G Feun
- Department of Medicine, University of Miami School of Medicine, Miami, FL, USA
| | | | - Medhi Wangpaichitr
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL, USA.
- Department of Surgery, University of Miami School of Medicine, Miami, FL, USA.
| |
Collapse
|
98
|
Mazinani M, Rahbarizadeh F. New cell sources for CAR-based immunotherapy. Biomark Res 2023; 11:49. [PMID: 37147740 PMCID: PMC10163725 DOI: 10.1186/s40364-023-00482-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 04/04/2023] [Indexed: 05/07/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy, in which a patient's own T lymphocytes are engineered to recognize and kill cancer cells, has achieved striking success in some hematological malignancies in preclinical and clinical trials, resulting in six FDA-approved CAR-T products currently available in the market. Despite impressive clinical outcomes, concerns about treatment failure associated with low efficacy or high cytotoxicity of CAR-T cells remain. While the main focus has been on improving CAR-T cells, exploring alternative cellular sources for CAR generation has garnered growing interest. In the current review, we comprehensively evaluated other cell sources rather than conventional T cells for CAR generation.
Collapse
Affiliation(s)
- Marzieh Mazinani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14115-111, Tehran, Iran
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14115-111, Tehran, Iran.
- Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
99
|
Thompson AA, Harbut MB, Kung PP, Karpowich NK, Branson JD, Grant JC, Hagan D, Pascual HA, Bai G, Zavareh RB, Coate HR, Collins BC, Côte M, Gelin CF, Damm-Ganamet KL, Gholami H, Huff AR, Limon L, Lumb KJ, Mak PA, Nakafuku KM, Price EV, Shih AY, Tootoonchi M, Vellore NA, Wang J, Wei N, Ziff J, Berger SB, Edwards JP, Gardet A, Sun S, Towne JE, Venable JD, Shi Z, Venkatesan H, Rives ML, Sharma S, Shireman BT, Allen SJ. Identification of small-molecule protein-protein interaction inhibitors for NKG2D. Proc Natl Acad Sci U S A 2023; 120:e2216342120. [PMID: 37098070 PMCID: PMC10160951 DOI: 10.1073/pnas.2216342120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 03/10/2023] [Indexed: 04/26/2023] Open
Abstract
NKG2D (natural-killer group 2, member D) is a homodimeric transmembrane receptor that plays an important role in NK, γδ+, and CD8+ T cell-mediated immune responses to environmental stressors such as viral or bacterial infections and oxidative stress. However, aberrant NKG2D signaling has also been associated with chronic inflammatory and autoimmune diseases, and as such NKG2D is thought to be an attractive target for immune intervention. Here, we describe a comprehensive small-molecule hit identification strategy and two distinct series of protein-protein interaction inhibitors of NKG2D. Although the hits are chemically distinct, they share a unique allosteric mechanism of disrupting ligand binding by accessing a cryptic pocket and causing the two monomers of the NKG2D dimer to open apart and twist relative to one another. Leveraging a suite of biochemical and cell-based assays coupled with structure-based drug design, we established tractable structure-activity relationships with one of the chemical series and successfully improved both the potency and physicochemical properties. Together, we demonstrate that it is possible, albeit challenging, to disrupt the interaction between NKG2D and multiple protein ligands with a single molecule through allosteric modulation of the NKG2D receptor dimer/ligand interface.
Collapse
Affiliation(s)
- Aaron A. Thompson
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Michael B. Harbut
- Therapeutics Discovery, Janssen Research & Development, Lower Gwynedd Township, PA19002
| | - Pei-Pei Kung
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Nathan K. Karpowich
- Therapeutics Discovery, Janssen Research & Development, Lower Gwynedd Township, PA19002
| | - Jeffrey D. Branson
- Therapeutics Discovery, Janssen Research & Development, Lower Gwynedd Township, PA19002
| | - Joanna C. Grant
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Deborah Hagan
- Therapeutics Discovery, Janssen Research & Development, Lower Gwynedd Township, PA19002
| | - Heather A. Pascual
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Guoyun Bai
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | | | - Heather R. Coate
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Bernard C. Collins
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Marjorie Côte
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Christine F. Gelin
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | | | - Hadi Gholami
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Adam R. Huff
- Therapeutics Discovery, Janssen Research & Development, Lower Gwynedd Township, PA19002
| | - Luis Limon
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Kevin J. Lumb
- Therapeutics Discovery, Janssen Research & Development, Lower Gwynedd Township, PA19002
| | - Puiying A. Mak
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Kohki M. Nakafuku
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Edmund V. Price
- Therapeutics Discovery, Janssen Research & Development, Lower Gwynedd Township, PA19002
| | - Amy Y. Shih
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Mandana Tootoonchi
- Discovery Immunology, Janssen Research & Development, San Diego, CA92121
| | - Nadeem A. Vellore
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Jocelyn Wang
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Na Wei
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Jeannie Ziff
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Scott B. Berger
- Discovery Immunology, Janssen Research & Development, Lower Gwynedd Township, PA19002
| | - James P. Edwards
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Agnès Gardet
- Discovery Immunology, Janssen Research & Development, San Diego, CA92121
| | - Siquan Sun
- Discovery Immunology, Janssen Research & Development, San Diego, CA92121
| | - Jennifer E. Towne
- Discovery Immunology, Janssen Research & Development, San Diego, CA92121
| | | | - Zhicai Shi
- Therapeutics Discovery, Janssen Research & Development, Lower Gwynedd Township, PA19002
| | | | - Marie-Laure Rives
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Sujata Sharma
- Therapeutics Discovery, Janssen Research & Development, Lower Gwynedd Township, PA19002
| | - Brock T. Shireman
- Therapeutics Discovery, Janssen Research & Development, San Diego, CA92121
| | - Samantha J. Allen
- Therapeutics Discovery, Janssen Research & Development, Lower Gwynedd Township, PA19002
| |
Collapse
|
100
|
Abstract
T cells and natural killer (NK) cells have complementary roles in tumor immunity, and dual T cell and NK cell attack thus offers opportunities to deepen the impact of immunotherapy. Recent work has also shown that NK cells play an important role in recruiting dendritic cells to tumors and thus enhance induction of CD8 T cell responses, while IL-2 secreted by T cells activates NK cells. Targeting of immune evasion mechanisms from the activating NKG2D receptor and its MICA and MICB ligands on tumor cells offers opportunities for therapeutic intervention. Interestingly, T cells and NK cells share several important inhibitory and activating receptors that can be targeted to enhance T cell- and NK cell-mediated immunity. These inhibitory receptor-ligand systems include CD161-CLEC2D, TIGIT-CD155, and NKG2A/CD94-HLA-E. We also discuss emerging therapeutic strategies based on inhibitory and activating cytokines that profoundly impact the function of both lymphocyte populations within tumors.
Collapse
Affiliation(s)
- Oleksandr Kyrysyuk
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA;
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA;
- Department of Neurology, Brigham & Women's Hospital, Boston, Massachusetts, USA
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|