1
|
Li J, Chen L, Chen M, Lin M, Xie Z, Wu H, Zhou Z, Lin W. Dap10 co-stimulation enhances the anti-HCC efficacy of NKp30 chimeric antigen receptor T cells. Transl Oncol 2025; 57:102425. [PMID: 40393250 DOI: 10.1016/j.tranon.2025.102425] [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: 08/23/2024] [Revised: 03/30/2025] [Accepted: 05/16/2025] [Indexed: 05/22/2025] Open
Abstract
Chimeric antigen receptor (CAR) T-cell immunotherapy has made significant breakthroughs in the treatment of relapsed or refractory hematologic malignancies, but its efficacy in solid tumors remains limited. In this study, we developed a chimeric NKp30 (chNKp30) receptor whose ligand, B7H6, is often up-regulated in various tumor cells and sparsely expressed in healthy cells. Introduction of the cytoplasmic structural domain of dnax-activating protein 10 (DAP10) into CAR resulted in chNKp30-Dap10 CAR-T cells that showed superior cell proliferation, activation, and apoptosis inhibition after antigenic stimulation compared with conventional chNKp30-CD28 and chNKp30-Wt CAR-T cells lacking any structural domains, along with inducing a central memory T cell phenotype, whereas chNKp30-CD28 and chNKp30-Wt triggered an effector memory phenotype. In addition, chNKp30-Dap10 T cells secreted higher levels of pro-inflammatory cytokines such as IL-2, IFN-γ, and TNF-α, while chNKp30-CD28 T cells secreted more of the anti-inflammatory cytokine IL-10. In the killing assay, chNKp30-Dap10 T cells demonstrated stronger anti-tumor effects. Similarly, better tumor regression was observed in the hepatocellular carcinoma transplantation tumor model. These findings suggest that B7H6 is an attractive therapeutic target and DAP10 signaling is involved in the functional regulation of CAR-T cells in hepatocellular carcinoma, which may induce preferential cytokine profiling and differentiation for cancer therapy, and that NKp30-Dap10 CAR-T cell therapy offers a potential option for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- JieYu Li
- Laboratory of Immuno-Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, PR China; Fuzhou University College of Chemistry, Fuzhou 350002, PR China; The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, Fujian Province, PR China
| | - LiMei Chen
- Department of Clinical Laboratory, Fuzhou 350014, PR China
| | - MingShui Chen
- Laboratory of Immuno-Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, PR China; Fuzhou University College of Chemistry, Fuzhou 350002, PR China; The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China
| | - Miao Lin
- Fuzhou University College of Chemistry, Fuzhou 350002, PR China
| | - Zineng Xie
- Fuzhou University College of Chemistry, Fuzhou 350002, PR China
| | - HuiLing Wu
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China
| | - ZhiFeng Zhou
- Laboratory of Immuno-Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, PR China; Fuzhou University College of Chemistry, Fuzhou 350002, PR China; The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, Fujian Province, PR China.
| | - WanSong Lin
- Laboratory of Immuno-Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, PR China; Fuzhou University College of Chemistry, Fuzhou 350002, PR China; The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, Fujian Province, PR China.
| |
Collapse
|
2
|
Lei W, Zhou K, Lei Y, Li Q, Zhu H. Pathogenesis and Systemic Treatment of Hepatocellular Carcinoma: Current Status and Prospects. Mol Cancer Ther 2025; 24:692-708. [PMID: 39417575 DOI: 10.1158/1535-7163.mct-24-0403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/14/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024]
Abstract
Hepatocellular carcinoma (HCC) remains one of the major threats to human health worldwide. The emergence of systemic therapeutic options has greatly improved the prognosis of patients with HCC, particularly those with advanced stages of the disease. In this review, we discussed the pathogenesis of HCC, genetic alterations associated with the development of HCC, and alterations in the tumor immune microenvironment. Then, important indicators and emerging technologies related to the diagnosis of HCC are summarized. Also, we reviewed the major advances in treatments for HCC, offering insights into future prospects for next-generation managements.
Collapse
Affiliation(s)
- Wanting Lei
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kexun Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ye Lei
- College of Liberal Arts, Neijiang Normal University, Neijiang, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Whalen KA, Henry CC, Mehta NK, Rakhra K, Yalcin S, Meetze K, Gibson NW, Baeuerle PA, Michaelson JS. CLN-619, a MICA/B monoclonal antibody that promotes innate immune cell-mediated antitumor activity. J Immunother Cancer 2025; 13:e008987. [PMID: 40274283 PMCID: PMC12020772 DOI: 10.1136/jitc-2024-008987] [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: 02/01/2024] [Accepted: 04/02/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Major histocompatibility complex class I-related protein A and B (MICA/B) are ligands for the natural killer group 2 member D (NKG2D) receptor and are broadly expressed on tumor cells but minimally on normal tissues. When cytotoxic NKG2D-expressing immune cells engage MICA/B, the ligand-expressing cells are targeted for lysis. Cancer cells can evade NKG2D-mediated destruction by shedding MICA/B from their cell surface via proteases present in the tumor microenvironment. CLN-619 is a humanized IgG1 monoclonal antibody (mAb) which binds MICA/B and inhibits shedding resulting in accumulation of MICA/B on the tumor cell surface. CLN-619 may thereby have therapeutic effects in a broad range of malignancies by re-establishing the MICA/B-NKG2D axis to enable NKG2D-mediated, as well as Fc-gamma receptor-mediated, tumor cell lysis. METHODS CLN-619 was characterized for binding epitope and affinity, effects on surface and soluble levels of MICA/B, and in vitro tumor cell killing. In mouse models, the mAb was tested for tumor growth inhibition. The contribution of the Fc-gamma (Fcγ) 1 domain to CLN-619 activity was also assessed. RESULTS CLN-619 bound with high affinity to the alpha-3 domain of MICA/B without encumbering the interaction with NKG2D on natural killer cells. CLN-619 increased the level of cell surface expression of MICA/B and concomitantly decreased the levels of soluble MICA/B in cell culture assays. Treatment of cancer cell lines with CLN-619 induced antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis. CLN-619 resulted in potent inhibition of tumor growth in multiple xenograft models and increased survival of mice in a disseminated cancer model. CONCLUSIONS CLN-619 inhibited the shedding of MICA/B to effectively restore cytotoxic signaling pathways in immune cells. Potent antitumor activity of CLN-619 as a monotherapy was observed in several preclinical models. Activity of CLN-619 required a functional Fcγ1 domain, suggesting the requirement of simultaneous engagement of NKG2D and cluster of differentiation 16A (CD16A) on immune cells for optimal cytotoxicity. The preclinical data reported here support the assessment of CLN-619 in patients with cancer.
Collapse
Affiliation(s)
- Kerry A Whalen
- Cullinan Therapeutics Inc, Cambridge, Massachusetts, USA
| | | | - Naveen K Mehta
- Cullinan Therapeutics Inc, Cambridge, Massachusetts, USA
| | - Kavya Rakhra
- Cullinan Therapeutics Inc, Cambridge, Massachusetts, USA
| | | | - Kristan Meetze
- Cullinan Therapeutics Inc, Cambridge, Massachusetts, USA
| | | | | | | |
Collapse
|
4
|
Biały S, Bogunia-Kubik K. Uncovering the mysteries of human gamma delta T cells: from origins to novel therapeutics. Front Immunol 2025; 16:1543454. [PMID: 40276509 PMCID: PMC12018481 DOI: 10.3389/fimmu.2025.1543454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Gamma delta (γδ) T cells represent a unique and distinct population of lymphocytes that bridge the innate and adaptive immune responses. This functional duality positions them as one of the pivotal elements in the evolution and development of the human body's defense mechanisms. This review aims to provide a comprehensive and in-depth overview of γδ T cells, covering their origins, development, classification, and functional roles in immunology. Special attention is given to their involvement in the pathogenesis of autoimmune and cancer-related diseases-areas that remain subjects of intensive research with many unanswered questions. Additionally, this article explores the therapeutic potential of γδ T cells, which hold promise as a novel approach to treating various difficult-to-manage diseases. The review also presents an analysis of the latest clinical studies utilizing γδ T cells, emphasizing their emerging role in modern medicine. The ultimate goal of this work is to offer a holistic perspective on the current state of research on γδ T cells and their prospective applications in immunotherapy and cancer treatment, highlighting their potential to become a groundbreaking tool in future medical interventions.
Collapse
Affiliation(s)
- Sylwia Biały
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of
Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | | |
Collapse
|
5
|
Liu J, Quan ZR, Zhu TH, Zhong YP, Jiang RH, Yang BN, Zhang YM, Song JM, Zou HY, Deng ZH. Allele and Haplotype Frequencies of 17 HLA-Related Loci in Shenzhen Chinese Population by Next-Generation Sequencing. HLA 2025; 105:e70148. [PMID: 40193066 DOI: 10.1111/tan.70148] [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: 09/03/2024] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 04/20/2025]
Abstract
Although the allele and haplotype frequencies of 11 HLA loci (HLA-A, B, C, DRB1, DRB3/4/5, DQA1, DQB1, DPA1 and DPB1) have been reported in different populations, rare studies have simultaneously assessed the allele distributions of non-classical HLA class I genes (HLA-E/F/G/H) and MICA/MICB together with the 11 classical HLA loci, or further analysed the haplotype frequencies covering the 17 loci. The present study aims to investigate the allele diversity and haplotype frequencies of 17 HLA-related loci including HLA genes and MICA/MICB simultaneously using a hybrid capture (HC)-based NGS method. A total of 358 HLA alleles including 177 class I and 137 class II alleles, as well as 29 MICA and 15 MICB alleles were identified in this project. The most frequent alleles at each locus were A*11:01 (29.10%), B*40:01 (14.46%), C*01:02 (19.90%), DRB1*09:01 (15.61%), DQB1*03:01 (18.48%), DPB1*05:01 (40.13%), DQA1*01:02 (22.58%), DPA1*02:02 (55.27%), DRB3*02:02 (65.95%), DRB4*01:03 (95.20%), DRB5*01:01 (75.97%), E*01:03 (62.63%), F*01:01 (97.07%), G*01:01 (70.74%), H*01:01 (35.87%), MICA*010:01 (19.90%) and MICB*005:02 (57.53%), respectively. The haplotype frequencies for different combinations of HLA loci were estimated and linkage disequilibrium (LD) between alleles for all pairs of neighbouring loci were calculated. The most frequent haplotype covering 17 loci was F*01:01-G*01:01-H*01:01-A*02:07-E*01:03-C*01:02-B*46:01-MICA*010:01-MICB*005:02-DRB4*01:03-DRB1*09:01-DQA1*03:02-DQB1*03:03-DPA1*02:02-DPB1*05:01 with a frequency of 3.18%. This is the first study on allelic polymorphism, haplotype inference and LD covering 17 HLA-related loci simultaneously in the Shenzhen Chinese population. These results will extend our knowledge of the allelic diversity of the HLA complex and provide population genetics data for transplantation and HLA-associated disease studies.
Collapse
Affiliation(s)
- Jie Liu
- Institute of Blood Transfusion Medicine, Shenzhen Blood Center, Shenzhen, Guangdong, China
- Immunogenetics Laboratory, Shenzhen Blood Center, Shenzhen, Guangdong, China
| | - Zhan-Rou Quan
- Institute of Blood Transfusion Medicine, Shenzhen Blood Center, Shenzhen, Guangdong, China
- Immunogenetics Laboratory, Shenzhen Blood Center, Shenzhen, Guangdong, China
| | - Tian-Hui Zhu
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, Guangdong, China
| | - Yan-Ping Zhong
- Shenzhen Pingle Orthopaedic Hospital (Shenzhen Pingshan District Hospital of Traditional Chinese Medicine), Shenzhen, Guangdong, China
| | - Ren-Hui Jiang
- Institute of Blood Transfusion Medicine, Shenzhen Blood Center, Shenzhen, Guangdong, China
- Immunogenetics Laboratory, Shenzhen Blood Center, Shenzhen, Guangdong, China
| | - Bing-Na Yang
- Institute of Blood Transfusion Medicine, Shenzhen Blood Center, Shenzhen, Guangdong, China
- Immunogenetics Laboratory, Shenzhen Blood Center, Shenzhen, Guangdong, China
| | - Yin-Ming Zhang
- Institute of Blood Transfusion Medicine, Shenzhen Blood Center, Shenzhen, Guangdong, China
- Immunogenetics Laboratory, Shenzhen Blood Center, Shenzhen, Guangdong, China
| | - Jia-Min Song
- Institute of Blood Transfusion Medicine, Shenzhen Blood Center, Shenzhen, Guangdong, China
- Immunogenetics Laboratory, Shenzhen Blood Center, Shenzhen, Guangdong, China
| | - Hong-Yan Zou
- Institute of Blood Transfusion Medicine, Shenzhen Blood Center, Shenzhen, Guangdong, China
- Immunogenetics Laboratory, Shenzhen Blood Center, Shenzhen, Guangdong, China
| | - Zhi-Hui Deng
- Institute of Blood Transfusion Medicine, Shenzhen Blood Center, Shenzhen, Guangdong, China
- Immunogenetics Laboratory, Shenzhen Blood Center, Shenzhen, Guangdong, China
| |
Collapse
|
6
|
Cieslak SG, Shahbazi R. Gamma delta T cells and their immunotherapeutic potential in cancer. Biomark Res 2025; 13:51. [PMID: 40148988 PMCID: PMC11951843 DOI: 10.1186/s40364-025-00762-6] [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: 12/18/2024] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Gamma-delta (γδ) T cells are a unique subset of T lymphocytes that play diverse roles in immune responses, bridging innate and adaptive immunity. With growing interest in their potential for cancer immunotherapy, a comprehensive and inclusive exploration of γδ T cell families, their development, activation mechanisms, functions, therapeutic implications, and current treatments is essential. This review aims to provide an inclusive and thorough discussion of these topics. Through our discussion, we seek to uncover insights that may harbinger innovative immunotherapeutic strategies. Beginning with an overview of γδ T cell families including Vδ1, Vδ2, and Vδ3, this review highlights their distinct functional properties and contributions to anti-tumor immunity. Despite γδ T cells exhibiting both anti-tumor and pro-tumor activities, our review elucidates strategies to harness the anti-tumor potential of γδ T cells for therapeutic benefit. Moreover, our paper discusses the structural intricacies of the γδ T cell receptor and its significance in tumor recognition. Additionally, this review examines conventional and emerging γδ T cell therapies, encompassing both non-engineered and engineered approaches, with a focus on their efficacy and safety profiles in clinical trials. From multifunctional capabilities to diverse tissue distribution, γδ T cells play a pivotal role in immune regulation and surveillance. By analyzing current research findings, this paper offers insights into the dynamic landscape of γδ T cell-based immunotherapies, underscoring their promise as a potent armamentarium against cancer. Furthermore, by dissecting the complex biology of γδ T cells, we learn valuable information about the anti-cancer contributions of γδ T cells, as well as potential targets for immunotherapeutic interventions.
Collapse
Affiliation(s)
- Stephen G Cieslak
- Division of Hematology/Oncology, Department of Medicine, Indiana University, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, USA
| | - Reza Shahbazi
- Division of Hematology/Oncology, Department of Medicine, Indiana University, Indianapolis, IN, USA.
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, USA.
- Tumor Microenvironment & Metastasis, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA.
- Brown Center for Immunotherapy, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
7
|
Puig-Gámez M, Van Attekum M, Theis T, Dick A, Park JE. Transcriptional signature of rapidly responding NK cells reveals S1P5 and CXCR4 as anti-tumor response disruptors. Sci Rep 2025; 15:10769. [PMID: 40155684 PMCID: PMC11953373 DOI: 10.1038/s41598-025-95211-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/19/2025] [Indexed: 04/01/2025] Open
Abstract
Natural killer (NK) cells are prototypic cytotoxic innate lymphocytes that can kill target cells, such as tumor cells, in the absence of antigen-restriction. Peripheral NK cells exhibit a high degree of heterogeneity. Here, we set out to broadly assess intrinsic modulators of NK cell degranulation in an unbiased manner. We stimulated human primary blood-borne NK cells pre-treated with different cytokine regimens with the HCT116 human colon cancer cell line and used detection of lysosome-associated membrane glycoprotein 1 (LAMP1) as an identifier of rapid NK cell degranulation. RNA sequencing of FACS-sorted LAMP1hi NK cells showed CXCR4 and S1PR5 were top down-regulated genes. Using compounds that modulate activity of CXCR4 and S1P receptor family members S1P1 and S1P5, we confirmed they play an important immunosuppressive role in NK cell cytotoxicity. Mechanistically, engagement of CXCR4 and S1P1/5 receptors triggered phosphorylation of p42 and Ca2+ influx. CXCR4 activation promoted S1P5 upregulation and vice versa, and joint activation of both receptors amplified the defect NK cell degranulation. Intriguingly, in tumor samples the expression of both receptors and the synthesis of their ligands themselves appear to be coordinately regulated. Together, these data suggest that specifically and simultaneously targeting CXCR4 and S1P5 activity in the tumor microenvironment (TME) could be a beneficial strategy to unleash full cytotoxic potential of cytotoxic NK effector cells in the tumor.
Collapse
Affiliation(s)
- Marta Puig-Gámez
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, 88387, Biberach an der Riss, Germany
| | - Martijn Van Attekum
- Department of Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88387, Biberach an der Riss, Germany
| | - Theodor Theis
- Department of Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, 88387, Biberach an der Riss, Germany
| | - Alec Dick
- Department of Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88387, Biberach an der Riss, Germany
| | - John E Park
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, 88387, Biberach an der Riss, Germany.
| |
Collapse
|
8
|
Yamane K, Yamaguchi K, Teruya Y, Miyake N, Nakayama Y, Nonaka T, Chikumi H, Yamasaki A. ULBP2 Promotes Tumor Progression by Suppressing NKG2D-Mediated Anti-Tumor Immunity. Int J Mol Sci 2025; 26:2950. [PMID: 40243581 PMCID: PMC11988498 DOI: 10.3390/ijms26072950] [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: 01/22/2025] [Revised: 03/01/2025] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
UL-16 binding protein 2 (ULBP2), a human NKG2D ligand, has been identified as a poor prognostic factor in several cancers based on recent comprehensive analyses of immune-related genes using the Cancer Genome Atlas datasets. Despite its clinical significance, the functional role of ULBP2 in vivo remains largely unknown. In this study, we investigated the role of ULBP2 in modulating anti-tumor immunity using murine melanoma cell lines engineered to stably express surface-expressed or soluble ULBP2. Subcutaneous transplantation of ULBP2-expressing melanoma cells into syngeneic mice resulted in accelerated tumor growth, mediated by surface-expressed ULBP2, through the suppression of NKG2D-dependent immune responses. In vitro experiments revealed that sustained exposure to tumor-expressed ULBP2 reduced NKG2D expression and cytotoxic activity of splenocytes. In contrast, soluble ULBP2 did not significantly affect tumor growth or immune responses. These findings suggest that surface-expressed ULBP2 plays a pivotal role in tumor immune evasion and highlight its potential as a therapeutic target to enhance anti-tumor immunity.
Collapse
Affiliation(s)
- Kohei Yamane
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago 683-8504, Japan; (K.Y.); (Y.T.); (N.M.); (T.N.); (A.Y.)
| | - Kosuke Yamaguchi
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago 683-8504, Japan; (K.Y.); (Y.T.); (N.M.); (T.N.); (A.Y.)
| | - Yasuhiko Teruya
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago 683-8504, Japan; (K.Y.); (Y.T.); (N.M.); (T.N.); (A.Y.)
| | - Naomi Miyake
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago 683-8504, Japan; (K.Y.); (Y.T.); (N.M.); (T.N.); (A.Y.)
| | - Yuji Nakayama
- Division of Radioisotope Science, Research Initiative Center, Organization for Research Initiative and Promotion, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan;
| | - Takafumi Nonaka
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago 683-8504, Japan; (K.Y.); (Y.T.); (N.M.); (T.N.); (A.Y.)
| | - Hiroki Chikumi
- Division of Infectious Diseases, School of Medicine, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago 683-8504, Japan;
| | - Akira Yamasaki
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago 683-8504, Japan; (K.Y.); (Y.T.); (N.M.); (T.N.); (A.Y.)
| |
Collapse
|
9
|
Poluektov YM, Lopina OD, Strelkova MA, Kuleshova ID, Makarov AA, Petrushanko IY. Mechanisms mediating effects of cardiotonic steroids in mammalian blood cells. Front Pharmacol 2025; 16:1520927. [PMID: 40196366 PMCID: PMC11973394 DOI: 10.3389/fphar.2025.1520927] [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: 10/31/2024] [Accepted: 02/21/2025] [Indexed: 04/09/2025] Open
Abstract
Cardiotonic steroids (CTSs) were known as steroidal plant compounds that exert cellular effects by the binding to Na,K-ATPase. Earlier, plant (exogenous) CTSs were used to treat chronic heart failure. By now, endogenous CTS have been identified in mammals, and their concentrations in the blood, normally in a subnanomolar range, are altered in numerous pathologies. This indicates their role as endogenous regulators of physiological processes. CTS transport occurs primarily in the blood, yet the CTS effects on blood cells remain poorly understood. This review summarizes the CTS effects on blood cells of animals and humans under normal and pathological conditions, and analyzes their action based on known mechanisms of action in mammalian cells. At high concentrations (greater than 10-9 M), CTS binding to Na,K-ATPase inhibits the enzyme, whereas lower concentrations of CTSs induce signaling cascades or activate the enzyme. All these mechanisms are shown to be present in blood cells. The particular CTS effect is determined by the CTS type, its concentration, the isoform composition of the catalytic α-subunit of Na,K-ATPase in the cell, and other cell features. It has been demonstrated that all blood cell types (erythrocytes, leukocytes, and platelets) expressed both ubiquitously distributed α1-isoform and tissue-specific α3-subunit, which exhibits a different ion and CTS affinity compared to α1. This results in a wide spectrum of blood cell responses to fluctuations in CTS levels in the blood. In particular, an increase in the level of endogenous CTSs by a more twofold is sufficient to induce a decline in the activity of erythrocyte Na,K-ATPase. The administration of exogenous CTSs is able to modulate the proinflammatory activity of leukocytes, which is attributed to the activation of signaling cascades, and to exert an influence on platelet activation. Hence, alterations of CTS levels in bloodstream significantly affect the functionality of blood cells, contributing to the organism's adaptive response. On top of this, a comparison of the effects of CTSs on human leukocytes and rodent leukocytes carrying the CTS-resistant α1-isoform often reveals opposite effects, thus indicating that rodents are an unsuitable model for studying CTS effects on these cells.
Collapse
Affiliation(s)
- Yuri M. Poluektov
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Olga D. Lopina
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Maria A. Strelkova
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Iuliia D. Kuleshova
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Alexander A. Makarov
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Irina Yu. Petrushanko
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
10
|
Subhi-Issa N, Tovar Manzano D, Pereiro Rodriguez A, Sanchez Ramon S, Perez Segura P, Ocaña A. γδ T Cells: Game Changers in Immune Cell Therapy for Cancer. Cancers (Basel) 2025; 17:1063. [PMID: 40227601 PMCID: PMC11987767 DOI: 10.3390/cancers17071063] [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: 12/18/2024] [Revised: 02/04/2025] [Accepted: 02/13/2025] [Indexed: 04/15/2025] Open
Abstract
Gamma delta (γδ) T cells are a unique subset of T lymphocytes with distinctive features that make them highly promising candidates for cancer therapy. Their MHC-independent recognition of tumor antigens, ability to mediate direct cytotoxicity, and role in modulating the tumor microenvironment position them as versatile agents in cancer immunotherapy. This review integrates and synthesizes the existing data on γδ T cells, with an emphasis on the development and optimization of in vitro expansion protocols. Critical aspects are detailed such as activation strategies, co-culture systems, cytokine use, and other parameters to ensure robust cell proliferation and functionality, which may be valuable for those developing or optimizing clinical practices. Finally, we discuss current advancements in γδ T cell research, clinical experience, and highlight areas needing further exploration. Considering these data, we hypothesize and propose potential new applications such as engineering γδ T cells for enhanced resistance to immune checkpoint pathways or for localized cytokine delivery within the tumor microenvironment, which could broaden their therapeutic impact in the treatment of cancer and beyond.
Collapse
Affiliation(s)
- Nabil Subhi-Issa
- Department of Immunology, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain
| | - Daniel Tovar Manzano
- Department of Immunology, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain
| | | | - Silvia Sanchez Ramon
- Department of Immunology, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain
- Department of Immunology, Ophthalmology, and ORL, School of Medicine, Complutense University, 28040 Madrid, Spain
| | - Pedro Perez Segura
- Department of Oncology, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain (A.O.)
| | - Alberto Ocaña
- Department of Oncology, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain (A.O.)
| |
Collapse
|
11
|
Al-Sharabass EA, El-Houseini ME, Effat H, Ibrahim SA, Abdellateif MS. The clinical potential of PDL-1 pathway and some related micro-RNAs as promising diagnostic markers for breast cancer. Mol Med 2025; 31:106. [PMID: 40108523 PMCID: PMC11921724 DOI: 10.1186/s10020-025-01137-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Immune checkpoint pathways play important roles in breast cancer (BC) pathogenesis and therapy. METHODS Expression levels of programmed cell death protein 1 (PD-1), cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed death-ligand 1 (PD-L1), Forkhead box P3 (FOXP3), miR-155, and miR-195 were assessed in the peripheral blood of 90 BC patients compared to 30 healthy controls using quantitative real-time PCR (qRt-PCR). The plasma level of soluble MHC class I chain related-protein B (MIC-B) protein was assessed using the enzyme linked immunosorbent assay (ELISA) technique. The data were correlated to the clinico-pathological characteristics of the patients. RESULTS There was a significant increase in the expression levels of PDL-1 [17.59 (3.24-123), p < 0.001], CTLA-4 [23.34 (1.3-1267), p = 0.006], PD-1 [10.25 (1-280), p < 0.001], FOXP3 [11.5 (1-234.8), p = 0.001], miR-155 [87.3 (1.5-910), p < 0.001] in BC patients compared to normal controls. The miR-195 was significantly downregulated in BC patients [0.23 (0-0.98, p < 0.001]. The plasma level of MIC-B was significantly increased in the BC patients [0.941 (0.204-6.38) ng/ml], compared to the control group [0.351 (0.211-0.884) ng/mL, p < 0.00]. PDL-1, CTLA-4, PD-1, and FOXP3 achieved a specificity of 100% for distinguishing BC patients, at a sensitivity of 93.3%, 82.2%, 62.2%, and 71.1% respectively. The combined expression of PDL-1 and CTLA-4 scored a 100% sensitivity and 100% specificity for diagnosing BC (p < 0.001). The sensitivity, specificity, and AUC of miR-155 were 88.9%, 96.7%, and 0.934; respectively (p < 0.001). While those of miR-195 were 73.3%, 60%, and 0.716; respectively (p = 0.001). MIC-B expression showed a 77.8% sensitivity, 80% specificity, and 0.811 AUC at a cutoff of 1.17 ng/ml (p < 0.001). Combined expression of miR-155 and miR-195 achieved a sensitivity of 91.1%, a specificity of 96.7%, and AUC of 0.926 (p < 0.001). Multivariate analysis showed that PDL-1 (OR:13.825, p = 0.004), CTLA-4 (OR: 20.958, p = 0.010), PD-1(OR:10.550, p = 0.044), MIC-B (OR: 17.89, p = 0.003), miR-155 (OR: 211.356, P < 0.001), and miR-195(OR:0.006, P < 0.001) were considered as independent risk factors for BC. CONCLUSIONS The PB levels of PDL-1, CTLA-4, PD-1, FOXP3, MIC-B, miR-155, and miR-195 could be used as promising diagnostic markers for BC patients.
Collapse
Affiliation(s)
| | - Motawa E El-Houseini
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Heba Effat
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | | | - Mona S Abdellateif
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt.
| |
Collapse
|
12
|
Pascoe RD, Kim Y, Rhodes A, Ong J, Tumpach C, Gubser C, Chang JJ, McMahon JH, Lewin SR, Rasmussen TA. Targeting Ikaros and Aiolos with pomalidomide fails to reactivate or induce apoptosis of the latent HIV reservoir. J Virol 2025; 99:e0167624. [PMID: 39902962 PMCID: PMC11915836 DOI: 10.1128/jvi.01676-24] [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: 09/23/2024] [Accepted: 01/12/2025] [Indexed: 02/06/2025] Open
Abstract
HIV persists in people living with HIV (PLHIV) on antiretroviral therapy (ART) in long-lived and proliferating latently infected CD4+ T cells that selectively express pro-survival proteins, including the zinc finger proteins, Ikaros and Aiolos. In this study, we investigated whether pomalidomide, an immunomodulatory agent that induces degradation of Ikaros and Aiolos, could increase the death of HIV-infected cells and/or reverse HIV latency. Using an in vitro model of CD4+ T cells infected with a green fluorescent protein (GFP) reporter virus, pomalidomide increased the expression of the pro-survival protein B cell lymphoma (Bcl)-2 and did not increase apoptosis of GFP+ HIV productively infected CD4+ T cells. Pomalidomide also increased the expression of CD155 and UL16-binding protein (ULBP) stress proteins on GFP+ HIV productively infected CD4+ T cells, but this did not translate to enhanced clearance following co-culture with a natural killer (NK) cell line. Using CD4+ T cells from PLHIV on ART, pomalidomide ex vivo activated memory CD4+ T cells resulting in elevated HLA-DR expression and induced CD4+ T cell proliferation but only in the presence of T cell receptor stimulation with anti-CD3 and anti-CD28. There was no effect on cell-associated HIV RNA or the frequency of intact HIV DNA. In conclusion, despite an increase in stress protein expression, promoting Ikaros and Aiolos degradation in CD4+ T cells using pomalidomide did not directly induce apoptosis of HIV-infected cells or induce HIV latency reversal.IMPORTANCEPeople living with HIV (PLHIV) require lifelong antiretroviral therapy (ART) due to the persistence of latently infected cells. The zinc finger proteins, Ikaros and Aiolos, have recently been implicated in promoting the persistence of latently infected cells. In this study, we investigated the effects of pomalidomide, an immunomodulatory imide drug that induces the degradation of Ikaros and Aiolos, on HIV latency reversal and death of infected cells. Using CD4+ T cells from people living with HIV on suppressive antiretroviral therapy, as well as an in vitro model of productive HIV infection, we found that pomalidomide induced T cell activation and expression of stress proteins but no evidence of latency reversal or selective death of infected cells.
Collapse
Affiliation(s)
- Rachel D. Pascoe
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Youry Kim
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Ajantha Rhodes
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jesslyn Ong
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Carolin Tumpach
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Celine Gubser
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - J. Judy Chang
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - James H. McMahon
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Sharon R. Lewin
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Victoria, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Thomas A. Rasmussen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
13
|
Henden C, Fjerdingstad HB, Bjørnsen EG, Thiruchelvam-Kyle L, Daws MR, Inngjerdingen M, Glover JC, Dissen E. NK-cell cytotoxicity toward pluripotent stem cells and their neural progeny: impacts of activating and inhibitory receptors and KIR/HLA mismatch. Stem Cells 2025; 43:sxae083. [PMID: 39708357 PMCID: PMC11929945 DOI: 10.1093/stmcls/sxae083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/14/2024] [Indexed: 12/23/2024]
Abstract
Pluripotent stem cells provide opportunities for treating injuries and previously incurable diseases. A major concern is the immunogenicity of stem cells and their progeny. Here, we have dissected the molecular mechanisms that allow natural killer (NK) cells to respond to human pluripotent stem cells, investigating a wide selection of activating and inhibitory NK-cell receptors and their ligands. Reporter cells expressing the activating receptor NKG2D responded strongly to embryonic stem (ES) cell lines and induced pluripotent stem (iPS) cell lines, whereas reporter cells expressing the activating receptors NKp30, NKp46, KIR2DS1, KIR2DS2, and KIR2DS4 did not respond. Human ES and iPS cells invariably expressed several ligands for NKG2D. Expression of HLA-C and HLA-E was lacking or low, insufficient to trigger reporter cells expressing the inhibitory receptors KIR2DL1, -2DL2, or -2DL3. Similar results were obtained for the pluripotent embryonic carcinoma cell lines NTERA-2 and 2102Ep, and also iPS-cell-derived neural progenitor cells. Importantly, neural progenitor cells and iPS-cell-derived motoneurons also expressed B7H6, the ligand for the activating receptor NKp30. In line with these observations, IL-2-stimulated NK cells showed robust cytotoxic responses to ES and iPS cells as well as to iPS-cell-derived motoneurons. No significant differences in cytotoxicity levels were observed between KIR/HLA matched and mismatched combinations of NK cells and pluripotent targets. Together, these data indicate that pluripotent stem cells and their neural progeny are targets for NK-cell killing both by failing to sufficiently express ligands for inhibitory receptors and by expression of ligands for activating receptors.
Collapse
Affiliation(s)
- Camilla Henden
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
| | - Hege B Fjerdingstad
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
- Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital, N-0317 Oslo, Norway
| | - Elisabeth G Bjørnsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
| | - Lavanya Thiruchelvam-Kyle
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
| | - Michael R Daws
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
| | - Marit Inngjerdingen
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, N-0317 Oslo, Norway
| | - Joel C Glover
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
- Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital, N-0317 Oslo, Norway
| | - Erik Dissen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
| |
Collapse
|
14
|
Kolossváry M, Schnittman SR, Zanni MV, Fitch KV, Fichtenbaum CJ, Aberg JA, Bloomfield GS, Malvestutto CD, Currier J, Diggs MR, deFilippi C, Eckard AR, Curran A, Centinbas M, Sadreyev R, Foldyna B, Mayrhofer T, Karady J, Taron J, McCallum S, Lu MT, Ribaudo HJ, Douglas PS, Grinspoon SK. Pitavastatin, Procollagen Pathways, and Plaque Stabilization in Patients With HIV: A Secondary Analysis of the REPRIEVE Randomized Clinical Trial. JAMA Cardiol 2025; 10:254-264. [PMID: 39661372 PMCID: PMC11771813 DOI: 10.1001/jamacardio.2024.4115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/29/2024] [Indexed: 12/12/2024]
Abstract
Importance In a mechanistic substudy of the Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE) randomized clinical trial, pitavastatin reduced noncalcified plaque (NCP) volume, but specific protein and gene pathways contributing to changes in coronary plaque remain unknown. Objective To use targeted discovery proteomics and transcriptomics approaches to interrogate biological pathways beyond low-density lipoprotein cholesterol (LDL-C), relating statin outcomes to reduce NCP volume and promote plaque stabilization among people with HIV (PWH). Design, Setting, and Participants This was a post hoc analysis of the double-blind, placebo-controlled, REPRIEVE randomized clinical trial. Participants underwent coronary computed tomography angiography (CTA), plasma protein analysis, and transcriptomic analysis at baseline and 2-year follow-up. The trial enrolled PWH from April 2015 to February 2018 at 31 US research sites. PWH without known cardiovascular diseases taking antiretroviral therapy and with low to moderate 10-year cardiovascular risk were eligible. Data analyses were conducted from October 2023 to February 2024. Intervention Oral pitavastatin calcium, 4 mg per day. Main Outcomes and Measures Relative change in plasma proteomics, transcriptomics, and noncalcified plaque volume among those receiving treatment vs placebo. Results Among 558 individuals (mean [SD] age, 51 [6] years; 455 male [82%]) included in the proteomics assessment, 272 (48.7%) received pitavastatin and 286 (51.3%) received placebo. After adjusting for false discovery rates, pitavastatin increased abundance of procollagen C-endopeptidase enhancer 1 (PCOLCE), neuropilin 1 (NRP-1), major histocompatibility complex class I polypeptide-related sequence A (MIC-A) and B (MIC-B), and decreased abundance of tissue factor pathway inhibitor (TFPI), tumor necrosis factor ligand superfamily member 10 (TRAIL), angiopoietin-related protein 3 (ANGPTL3), and mannose-binding protein C (MBL2). Among these proteins, the association of pitavastatin with PCOLCE (a rate-limiting enzyme of collagen deposition) was greatest, with an effect size of 24.3% (95% CI, 18.0%-30.8%; P < .001). In a transcriptomic analysis, individual collagen genes and collagen gene sets showed increased expression. Among the 195 individuals with plaque at baseline (88 [45.1%] taking pitavastatin, 107 [54.9%] taking placebo), changes in NCP volume were most strongly associated with changes in PCOLCE (%change NCP volume/log2-fold change = -31.9%; 95% CI, -42.9% to -18.7%; P < .001), independent of changes in LDL-C level. Increases in PCOLCE related most strongly to change in the fibro-fatty (<130 Hounsfield units) component of NCP (%change fibro-fatty volume/log2-fold change = -38.5%; 95% CI, -58.1% to -9.7%; P = .01) with a directionally opposite, although nonsignificant, increase in calcified plaque (%change calcified volume/log2-fold change = 34.4%; 95% CI, -7.9% to 96.2%; P = .12). Conclusions and Relevance Results of this secondary analysis of the REPRIEVE randomized clinical trial suggest that PCOLCE may be associated with the atherosclerotic plaque stabilization effects of statins by promoting collagen deposition in the extracellular matrix transforming vulnerable plaque phenotypes to more stable coronary lesions. Trial Registration ClinicalTrials.gov Identifier: NCT02344290.
Collapse
Affiliation(s)
- Márton Kolossváry
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Samuel R. Schnittman
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
- Division of Infectious Diseases, Massachusetts General Hospital, Boston
| | - Markella V. Zanni
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Kathleen V. Fitch
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Carl J. Fichtenbaum
- Division of Infectious Diseases, Department of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Judith A. Aberg
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Gerald S. Bloomfield
- Department of Medicine, Duke Global Health Institute, Duke Clinical Research Institute, Duke University, Durham, North Carolina
| | | | - Judith Currier
- Division of Infectious Diseases, University of California, Los Angeles, Los Angeles, California
| | - Marissa R. Diggs
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | | | - Allison Ross Eckard
- Departments of Pediatrics and Medicine, Divisions of Infectious Diseases, Medical University of South Carolina, Charleston
| | - Adrian Curran
- Infectious Diseases Department, Vall d’Hebron Research Institute, Hospital Universitari Vall d’Hebron, Barcelona, Spain
| | - Murat Centinbas
- Department of Molecular Biology, Massachusetts General Hospital, Boston
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Ruslan Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Borek Foldyna
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Thomas Mayrhofer
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
- School of Business Studies, Stralsund University of Applied Sciences, Stralsund, Germany
| | - Julia Karady
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
- Cardiovascular Imaging Research Group, Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Jana Taron
- Department of Molecular Biology, Massachusetts General Hospital, Boston
- Department of Radiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Sara McCallum
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Michael T. Lu
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Heather J. Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Pamela S. Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| | - Steven K. Grinspoon
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
15
|
Rojas-Domínguez A, Martínez-Vargas IU, Alvarado-Mentado M. Modeling and simulation of genotypic Tumor Mutational Burden and Phenotypic Immunogenicity biomarkers in cancer immunoediting with Ising-Hamiltonian characterization. Comput Biol Med 2025; 187:109717. [PMID: 39894008 DOI: 10.1016/j.compbiomed.2025.109717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND AND OBJECTIVE In the Tumor Micro-Environment, cancer progression and its relationship with the Immune System (IS) are described in terms of cancer immunoediting (CI) phases, each of which is characterized by different types and levels of interaction between the tumor cells and elements of the IS, such as CD8+T cells. Said interactions are governed by genotypical (Tumor Mutational Burden, TMB) and phenotypical aspects pertaining to the tumor, as well as by the strength of the IS. In this work, a computational model of CI is presented that incorporates the TMB and the biomarker Tumor Immunogenic Phenotype (TIP) as its control parameters, and which employs the Ising-model Hamiltonian to characterize the system with respect to the CI phases. METHODS Our model is a probabilistic multi-agent system with agents for tumor cells and for the IS. The computer implementation includes the parametrization of the TMB and the TIP, which is useful for identifying whether a tumor is hot or cold based on tumor immunogenicity and inflammation. For modeling the interactions between tumor and immune cells, the relevant elements are integrated under a Michaelis-Menten equation that regulates the recruitment rate of CD8+T cells and other IS elements. This novel quantification of immune cell recruitment encompasses the growth of neoantigen production, which in turn triggers the growth of CD8+T cells. RESULTS Our model reliably captures the Elimination, Equilibrium, and Escape phases of tumor-immune cell interactions, modulating the observed behaviors through the introduced parametrization of TMB and TIP biomarkers. Notably, these results align well with the combination of genotypical and phenotypical biomarkers analyzed in recent literature. A remarkable instance is the appreciable inhibition of the tumor activity during the Escape phase, observed for phenotypically hot tumors with relatively high TMB, and pointing towards improved efficacy of the IS against such tumors. The Ising-Hamiltonian provides precise quantification of diverse tumor-immune interactions across different TMB and TIP value combinations. CONCLUSIONS The presented model, formed by relatively simple agents, generates emergent behaviors through which the phases of CI are identified. The flexible choice of control parameters is robust enough and provides a plausible explanation for the mechanisms through which tumors with high TMB and high immunogenicity (i.e., hot tumors) exhibit a higher probability of responding to immunotherapy treatment. Characterization via the Ising-model Hamiltonian supports this explanation by summarizing the system's dynamics, which, in turn, facilitates its analysis and methodical improvements. The complex interplay of TMB, TIP, and individual physiology is finely captured.
Collapse
Affiliation(s)
- Alfonso Rojas-Domínguez
- Centro de Investigación en Computación-IPN, Av. Juan de Dios Bátiz, esq. Miguel Othón de Mendizábal, Col.Nueva Industrial Vallejo, CDMX, 0700, Mexico
| | - Irving Ulises Martínez-Vargas
- Depto. de Computación CINVESTAV, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco., CDMX, 07360, Mexico; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place., New York, 10029, NY, USA
| | - Matías Alvarado-Mentado
- Depto. de Computación CINVESTAV, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco., CDMX, 07360, Mexico.
| |
Collapse
|
16
|
Ozer EA, Keskin A, Berrak YH, Cankara F, Can F, Gursoy-Ozdemir Y, Keskin O, Gursoy A, Yapici-Eser H. Shared interactions of six neurotropic viruses with 38 human proteins: a computational and literature-based exploration of viral interactions and hijacking of human proteins in neuropsychiatric disorders. DISCOVER MENTAL HEALTH 2025; 5:18. [PMID: 39987419 PMCID: PMC11846830 DOI: 10.1007/s44192-025-00128-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/09/2025] [Indexed: 02/24/2025]
Abstract
INTRODUCTION Viral infections may disrupt the structural and functional integrity of the nervous system, leading to acute conditions such as encephalitis, and neuropsychiatric conditions as mood disorders, schizophrenia, and neurodegenerative diseases. Investigating viral interactions of human proteins may reveal mechanisms underlying these effects and offer insights for therapeutic interventions. This study explores molecular interactions of virus and human proteins that may be related to neuropsychiatric disorders. METHODS Herpes Simplex Virus-1 (HSV-1), Cytomegalovirus (CMV), Epstein-Barr Virus (EBV), Influenza A virus (IAV) (H1N1, H5N1), and Human Immunodeficiency Virus (HIV1&2) were selected as key viruses. Protein structures for each virus were accessed from the Protein Data Bank and analyzed using the HMI-Pred web server to detect interface mimicry between viral and human proteins. The PANTHER classification system was used to categorize viral-human protein interactions based on function and cellular localization. RESULTS Energetically favorable viral-human protein interactions were identified for HSV-1 (467), CMV (514), EBV (495), H1N1 (3331), H5N1 (3533), and HIV 1&2 (62425). Besides immune and apoptosis-related pathways, key neurodegenerative pathways, including those associated with Parkinson's and Huntington's diseases, were frequently interacted. A total of 38 human proteins, including calmodulin 2, Ras-related botulinum toxin substrate 1 (Rac1), PDGF-β, and vimentin, were found to interact with all six viruses. CONCLUSION The study indicates a substantial number of energetically favorable interactions between human proteins and selected viral proteins, underscoring the complexity and breadth of viral strategies to hijack host cellular mechanisms. Further in vivo and in vitro validation is required to understand the implications of these interactions.
Collapse
Affiliation(s)
| | - Aleyna Keskin
- School of Medicine, Koç University, Istanbul, Turkey
| | | | - Fatma Cankara
- Graduate School of Sciences and Engineering, Computational Sciences and Engineering, Koç University, Istanbul, Turkey
| | - Fusun Can
- Department of Microbiology, School of Medicine, Koç University, Istanbul, Turkey
| | - Yasemin Gursoy-Ozdemir
- Department of Neurology, School of Medicine, Koç University, Istanbul, Turkey
- Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Ozlem Keskin
- Department of Chemical and Biological Engineering, College of Engineering, Koç University, Istanbul, Turkey
| | - Attila Gursoy
- Department of Computer Science and Engineering, College of Engineering, Koç University, Istanbul, Turkey.
| | - Hale Yapici-Eser
- Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey.
- Department of Psychiatry, School of Medicine, Koç University, Istanbul, Turkey.
| |
Collapse
|
17
|
Chang TD, Chen YJ, Luo JL, Zhang C, Chen SY, Lin ZQ, Zhang PD, Shen YX, Tang TX, Li H, Dong LM, Tang ZH, Chen D, Wang YM. Adaptation of Natural Killer Cells to Hypoxia: A Review of the Transcriptional, Translational, and Metabolic Processes. Immunotargets Ther 2025; 14:99-121. [PMID: 39990274 PMCID: PMC11846490 DOI: 10.2147/itt.s492334] [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: 08/21/2024] [Accepted: 02/08/2025] [Indexed: 02/25/2025] Open
Abstract
As important innate immune cells, natural killer (NK) cells play an essential role in resisting pathogen invasion and eliminating transformed cells. However, the hypoxic microenvironment caused by disease conditions is an important physicochemical factor that impairs NK cell function. With the increasing prominence of NK cells in immunotherapy, there has been a surge of interest in developing biological means through which NK cells may overcome the inhibition caused by hypoxia in disease conditions. Although the effects of hypoxic conditions in shaping the functions of NK cells have been increasingly recognized and investigated, reviews have been scantly. A comprehensive understanding of how NK cells adapt to hypoxia can provide valuable insights into how the functional capacity of NK cells may be restored. This review focuses on the functional alterations of NK cells in response to hypoxia. It delineates the mechanisms by which NK cells adapt to hypoxia at the transcriptional, metabolic, translational levels. Furthermore, given the complexity of the hypoxic microenvironment, we also elucidated the effects of key hypoxic metabolites on NK cells. Finally, this review discusses the current clinical therapies derived from targeting hypoxic NK cells. The study of NK cell adaptation to hypoxia has yielded new insights into immunotherapy. These insights may lead to development of novel strategies to improve the treatment of infectious diseases and cancer.
Collapse
Affiliation(s)
- Te-Ding Chang
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yu-Jie Chen
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jia-Liu Luo
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Cong Zhang
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Shun-Yao Chen
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhi-Qiang Lin
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Pei-Dong Zhang
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - You-Xie Shen
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Ting-Xuan Tang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Hui Li
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Li-Ming Dong
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhao-Hui Tang
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Deng Chen
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yu-Man Wang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
18
|
Guo P, Zhong L, Wang T, Luo W, Zhou A, Cao D. NK cell-based immunotherapy for hepatocellular carcinoma: Challenges and opportunities. Scand J Immunol 2025; 101:e13433. [PMID: 39934640 DOI: 10.1111/sji.13433] [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: 09/07/2024] [Revised: 12/22/2024] [Accepted: 01/01/2025] [Indexed: 02/13/2025]
Abstract
Hepatocellular carcinoma (HCC) remains one of the most challenging malignancies globally, characterized by significant heterogeneity, late-stage diagnosis, and resistance to treatment. In recent years, the advent of immune-checkpoint blockades (ICBs) and targeted immune cell therapies has marked a substantial advancement in HCC treatment. However, the clinical efficacy of these existing therapies is still limited, highlighting the urgent need for new breakthroughs. Natural killer (NK) cells, a subset of the innate lymphoid cell family, have shown unique advantages in the anti-tumour response. With increasing evidence suggesting the crucial role of dysfunctional NK cells in the pathogenesis and progression of HCC, considerable efforts have been directed toward exploring NK cells as a potential therapeutic target for HCC. In this review, we will provide an overview of the role of NK cells in normal liver immunity and in HCC, followed by a detailed discussion of various NK cell-based immunotherapies and their potential applications in HCC treatment.
Collapse
Affiliation(s)
- Pei Guo
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Liyuan Zhong
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Tao Wang
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weijia Luo
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Aiqiang Zhou
- Guangzhou Hospital of Integrated Chinese and Western Medicine, Guangzhou, Guangdong, P.R China
| | - Deliang Cao
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
19
|
Wang R, Hu M, Lozzi I, Jin CZJ, Ma D, Splith K, Mengwasser J, Wolf V, Feldbrügge L, Tang P, Timmermann L, Hillebrandt KH, Kirchner M, Mertins P, Hilfenhaus G, Neumann CCM, Kammertoens T, Pratschke J, Malinka T, Sauer IM, Noessner E, Guo ZS, Felsenstein M. Cytokine-armed vaccinia virus promotes cytotoxicity toward pancreatic carcinoma cells via activation of human intermediary CD56 dimCD16 dim natural killer cells. Int J Cancer 2025; 156:638-651. [PMID: 39400317 PMCID: PMC11621990 DOI: 10.1002/ijc.35209] [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: 01/09/2024] [Revised: 08/20/2024] [Accepted: 09/18/2024] [Indexed: 10/15/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a particularly aggressive disease with few effective treatments. The PDAC tumor immune microenvironment (TIME) is known to be immune suppressive. Oncolytic viruses can increase tumor immunogenicity via immunogenic cell death (ICD). We focused on tumor-selective (vvDD) and cytokine-armed Western-reserve vaccinia viruses (vvDD-IL2 and vvDD-IL15) and infected carcinoma cell lines as well as patient-derived primary PDAC cells. In co-culture experiments, we investigated the cytotoxic response and the activation of human natural killer (NK). Infection and virus replication were assessed by measuring virus encoded YFP. We then analyzed intracellular signaling processes and oncolysis via in-depth proteomic analysis, immunoblotting and TUNEL assay. Following the co-culture of mock or virus infected carcinoma cell lines with allogenic PBMCs or NK cell lines, CD56+ NK cells were analyzed with respect to their activation, cytotoxicity and effector function. Both, dose- and time-dependent release of danger signals following infection were measured. Viruses effectively entered PDAC cells, emitted YFP signals and resulted in concomitant oncolysis. The proteome showed reprogramming of normally active core signaling pathways in PDAC (e.g., MAPK-ERK signaling). Danger-associated molecular patterns were released upon infection and stimulated co-cultured NK cells for enhanced effector cytotoxicity. NK cell subtyping revealed enhanced numbers and activation of a rare CD56dimCD16dim population. Tumor cell killing was primarily triggered via Fas ligands rather than granule release, resulting in marked apoptosis. Overall, the cytokine-armed vaccinia viruses induced NK cell activation and enhanced cytotoxicity toward human PDAC cells in vitro. We could show that cytokine-armed virus targets the carcinoma cells and thus has great potential to modulate the TIME in PDAC.
Collapse
Affiliation(s)
- Ruonan Wang
- Department of Surgery, CCMCVK, Experimental SurgeryCharité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Mengwen Hu
- Department of Surgery, CCMCVK, Experimental SurgeryCharité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Isis Lozzi
- Department of Surgery, CCMCVK, Experimental SurgeryCharité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Cao Zhong Jing Jin
- Department of Surgery, CCMCVK, Experimental SurgeryCharité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Dou Ma
- Department of Surgery, CCMCVK, Experimental SurgeryCharité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Katrin Splith
- Department of General, Visceral and Transplant SurgeryMedizinische Hochschule HannoverHannoverGermany
| | - Jörg Mengwasser
- Department of General, Visceral and Transplant SurgeryMedizinische Hochschule HannoverHannoverGermany
| | - Vincent Wolf
- Department of General, Visceral and Transplant SurgeryMedizinische Hochschule HannoverHannoverGermany
| | - Linda Feldbrügge
- Department of General, Visceral and Transplant SurgeryMedizinische Hochschule HannoverHannoverGermany
| | - Peter Tang
- Department of Surgery, CCMCVK, Experimental SurgeryCharité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Lea Timmermann
- Department of Surgery, CCMCVK, Experimental SurgeryCharité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Berlin Institute of Health at Charité—Universitätsmedizin BerlinBIH Biomedical Innovation Academy, BIH Charité Clinician Scientist ProgramBerlinGermany
| | - Karl Herbert Hillebrandt
- Department of Surgery, CCMCVK, Experimental SurgeryCharité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Berlin Institute of Health at Charité—Universitätsmedizin BerlinBIH Biomedical Innovation Academy, BIH Charité Clinician Scientist ProgramBerlinGermany
| | - Marieluise Kirchner
- Core Unit ProteomicsBerlin Institute of Health at Charité—Universitätsmedizin Berlin and Max‐Delbrück‐Center for Molecular MedicineBerlinGermany
| | - Philipp Mertins
- Core Unit ProteomicsBerlin Institute of Health at Charité—Universitätsmedizin Berlin and Max‐Delbrück‐Center for Molecular MedicineBerlinGermany
| | - Georg Hilfenhaus
- Berlin Institute of Health at Charité—Universitätsmedizin BerlinBIH Biomedical Innovation Academy, BIH Charité Clinician Scientist ProgramBerlinGermany
- Medical Department, Division of Hematology, Oncology and Tumor ImmunologyCCM, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Christopher Claudius Maximilian Neumann
- Berlin Institute of Health at Charité—Universitätsmedizin BerlinBIH Biomedical Innovation Academy, BIH Charité Clinician Scientist ProgramBerlinGermany
- Medical Department, Division of Hematology, Oncology and Tumor ImmunologyCCM, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Thomas Kammertoens
- Institute of Immunology, Charité Unversitätsmedizin, Campus BuchBerlinGermany
| | - Johann Pratschke
- Department of Surgery, CCMCVK, Experimental SurgeryCharité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Thomas Malinka
- Department of Surgery, CCMCVK, Experimental SurgeryCharité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Igor Maximillian Sauer
- Department of Surgery, CCMCVK, Experimental SurgeryCharité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Berlin Institute of Health at Charité—Universitätsmedizin BerlinBIH Biomedical Innovation Academy, BIH Charité Clinician Scientist ProgramBerlinGermany
| | | | - Zong Sheng Guo
- Department of ImmunologyRoswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | - Matthäus Felsenstein
- Department of Surgery, CCMCVK, Experimental SurgeryCharité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
- Berlin Institute of Health at Charité—Universitätsmedizin BerlinBIH Biomedical Innovation Academy, BIH Charité Clinician Scientist ProgramBerlinGermany
| |
Collapse
|
20
|
Dallmann-Sauer M, Fava VM, Malherbe ST, MacDonald CE, Orlova M, Kroon EE, Cobat A, Boisson-Dupuis S, Hoal EG, Abel L, Möller M, Casanova JL, Walzl G, Du Plessis N, Schurr E. Mycobacterium tuberculosis resisters despite HIV exhibit activated T cells and macrophages in their pulmonary alveoli. J Clin Invest 2025; 135:e188016. [PMID: 39836471 PMCID: PMC11957701 DOI: 10.1172/jci188016] [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: 10/11/2024] [Accepted: 01/16/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUNDNatural resistance to Mycobacterium tuberculosis (Mtb) infection in some people with HIV (PWH) is unexplained.METHODSWe performed single cell RNA-sequencing of bronchoalveolar lavage cells, unstimulated or ex vivo stimulated with Mtb, for 7 PWH who were tuberculin skin test (TST) and IFN-γ release assay (IGRA) positive (called LTBI) and 6 who were persistently TST and IGRA negative (called resisters).RESULTSAlveolar macrophages (AM) from resisters displayed a baseline M1 macrophage phenotype while AM from LTBI did not. Resisters displayed alveolar lymphocytosis, with enrichment of all T cell subpopulations including IFNG-expressing cells. In both groups, mycobactericidal granulysin was expressed almost exclusively by a T cell subtype that coexpressed granzyme B, perforin and NK cell receptors. These poly-cytotoxic T lymphocytes (poly-CTL) overexpressed activating NK cell receptors and were increased in resister BAL. Following challenge with Mtb, only intraepithelial lymphocyte-like cells from LTBI participants responded with increased transcription of IFNG. AM from resisters responded with a stronger TNF signature at 6 hours after infection while at 24 hours after infection, AM from LTBI displayed a stronger IFN-γ signature. Conversely, at 24 hours after infection, only AM from resisters displayed an upregulation of MHC class I polypeptide-related sequence A (MICA) transcripts, which encode an activating ligand for poly-CTL.CONCLUSIONThese results suggest that poly-CTL and M1-like pre-activated AM mediate the resister phenotype in PWH.FUNDINGNational Institutes of Health. Canadian Institutes of Health Research. Digital Research Alliance of Canada. French National Research Agency. French National Agency for Research on AIDS and Viral Hepatitis. St. Giles Foundation. General Atlantic Foundation. South African Medical Research Council Centre for Tuberculosis Research.
Collapse
Affiliation(s)
- Monica Dallmann-Sauer
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, Montreal, Canada
- McGill International TB Centre, and
- Departments of Human Genetics and Medicine, Faculty of Medicine and Health Science, McGill University, Montreal, Canada
| | - Vinicius M. Fava
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, Montreal, Canada
- McGill International TB Centre, and
| | - Stephanus T. Malherbe
- South African Medical Research Council Centre for Tuberculosis Research; Biomedical Research Institute, Division of Immunology, Department of Biomedical Sciences and
| | - Candice E. MacDonald
- South African Medical Research Council Centre for Tuberculosis Research; Biomedical Research Institute, Division of Immunology, Department of Biomedical Sciences and
| | - Marianna Orlova
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, Montreal, Canada
- McGill International TB Centre, and
- Departments of Human Genetics and Medicine, Faculty of Medicine and Health Science, McGill University, Montreal, Canada
| | - Elouise E. Kroon
- South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Aurélie Cobat
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Université Paris Cité, Imagine Institute, Paris, France
| | - Stéphanie Boisson-Dupuis
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Université Paris Cité, Imagine Institute, Paris, France
| | - Eileen G. Hoal
- South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Université Paris Cité, Imagine Institute, Paris, France
| | - Marlo Möller
- South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Université Paris Cité, Imagine Institute, Paris, France
- Howard Hughes Medical Institute, New York, New York, USA
- Pediatric Hematology and Immunology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Gerhard Walzl
- South African Medical Research Council Centre for Tuberculosis Research; Biomedical Research Institute, Division of Immunology, Department of Biomedical Sciences and
| | - Nelita Du Plessis
- South African Medical Research Council Centre for Tuberculosis Research; Biomedical Research Institute, Division of Immunology, Department of Biomedical Sciences and
| | - Erwin Schurr
- Program in Infectious Diseases and Immunity in Global Health, The Research Institute of the McGill University Health Centre, Montreal, Canada
- McGill International TB Centre, and
- Departments of Human Genetics and Medicine, Faculty of Medicine and Health Science, McGill University, Montreal, Canada
| |
Collapse
|
21
|
Pogoda-Wesołowska A, Sługocka N, Synowiec A, Brodaczewska K, Mejer-Zahorowski M, Ziękiewicz M, Szypowski W, Szymański P, Stępień A. The current state of knowledge on the role of NKG2D ligands in multiple sclerosis and other autoimmune diseases. Front Mol Neurosci 2025; 17:1493308. [PMID: 39866909 PMCID: PMC11758245 DOI: 10.3389/fnmol.2024.1493308] [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: 09/08/2024] [Accepted: 12/18/2024] [Indexed: 01/28/2025] Open
Abstract
Multiple sclerosis (MS) is a chronic central nervous system (CNS) disease with demyelinating inflammatory characteristics. It is the most common nontraumatic and disabling disease affecting young adults. The incidence and prevalence of MS have been increasing. However, its exact cause remains unclear. The main tests used to support the diagnosis are magnetic resonance imaging (MRI) examination and cerebrospinal fluid (CSF) analysis. Nonetheless, to date, no sensitive or specific marker has been identified for the detection of the disease at its initial stage. In recent years, researchers have focused on the fact that the number of natural killer cell group 2 member D (NKG2D) family of C-type lectin-like receptor + (NKG2D+) T cells in the peripheral blood, CSF, and brain tissue has been shown to be higher in patients with MS than in controls. The activating receptor belonging to the NKG2D is stimulated by specific ligands: in humans these are major histocompatibility complex (MHC) class I polypeptide-related sequence A (MICA) and MHC class I polypeptide-related sequence B (MICB) proteins and UL16 binding 1-6 proteins (ULBP1-6). Under physiological conditions, the aforementioned ligands are expressed at low or undetectable levels but can be induced in response to stress factors. NKG2D ligands (NKG2DLs) are involved in epigenetic regulation of their expression. To date, studies in cell cultures, animal models, and brain tissues have revealed elevated expression of MICA/B, ULPB4, and its mouse homolog murine UL16 binding protein-like transcript (MULT1), in oligodendrocytes and astrocytes from patients with MS. Furthermore, soluble forms of NKG2DLs were elevated in the plasma and CSF of patients with MS compared to controls. In this review, we aim to describe the role of NKG2D and NKG2DLs, and their interactions in the pathogenesis of MS, as well as in other autoimmune diseases such as rheumatoid arthritis (RA), inflammatory bowel disease (IBD), systemic lupus erythematosus (SLE), and celiac disease (CeD). We also assess the potential of these proteins as diagnostic markers and consider future perspectives for targeting NKG2D ligands and their pathways as therapeutic targets in MS.
Collapse
Affiliation(s)
| | - Nina Sługocka
- Faculty of Medicine, University of Warsaw, Warsaw, Poland
| | - Agnieszka Synowiec
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine–National Research Institute, Warsaw, Poland
| | - Klaudia Brodaczewska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine–National Research Institute, Warsaw, Poland
| | - Marcin Mejer-Zahorowski
- Neurology Clinic, Military Institute of Medicine- National Research Institute, Warsaw, Poland
| | - Maciej Ziękiewicz
- Neurology Clinic, Military Institute of Medicine- National Research Institute, Warsaw, Poland
| | - Wojciech Szypowski
- Neurology Clinic, Military Institute of Medicine- National Research Institute, Warsaw, Poland
| | - Piotr Szymański
- Neurology Clinic, Military Institute of Medicine- National Research Institute, Warsaw, Poland
| | - Adam Stępień
- Neurology Clinic, Military Institute of Medicine- National Research Institute, Warsaw, Poland
| |
Collapse
|
22
|
Lee MJ, Litchford ML, Vendrame E, Vergara R, Ranganath T, Fish CS, Chebet D, Langat A, Mburu C, Neary J, Benki S, Wamalwa D, John-Stewart G, Lehman DA, Blish CA. Distinct immune profiles in children living with HIV based on timing and duration of suppressive antiretroviral treatment. Virology 2025; 602:110318. [PMID: 39612623 PMCID: PMC11645197 DOI: 10.1016/j.virol.2024.110318] [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: 09/17/2024] [Revised: 11/05/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024]
Abstract
Timely initiation of antiretroviral therapy (ART) remains a major challenge in the effort to treat children living with HIV ("CLH") and little is known regarding the dynamics of immune normalization following ART in CLH with varying times to and durations of ART. Here, we leveraged two cohorts of virally-suppressed CLH from Nairobi, Kenya to examine differences in the peripheral immune systems between two cohorts of age-matched children (to control for immune changes with age): one group which initiated ART during early HIV infection and had been on ART for 5-6 years at evaluation (early, long-term treated; "ELT" cohort), and one group which initiated ART later and had been on ART for approximately 9 months at evaluation (delayed, short-term treated; "DST" cohort). We profiled PBMC and purified NK cells from these two cohorts by mass cytometry time-of-flight (CyTOF). Although both groups of CLH had undetectable viral RNA load at evaluation, there were marked differences in both immune composition and immune phenotype between the ELT cohort and the DST cohort. DST donors had reduced CD4 T cell percentages, decreased naive to effector memory T cell ratios, and markedly higher expression of stress-induced markers. Conversely, ELT donors had higher naive to effector memory T cell ratios, low expression of stress-induced markers, and increased expression of markers associated with an effective antiviral response and resolution of inflammation. Collectively, our results demonstrate key differences in the immune systems of virally-suppressed CLH with different ages at ART initiation and durations of treatment and provide further rationale for emphasizing early onset of ART.
Collapse
Affiliation(s)
- Madeline J Lee
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Stanford Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Morgan L Litchford
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Elena Vendrame
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rosemary Vergara
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Thanmayi Ranganath
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Carolyn S Fish
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Daisy Chebet
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Agnes Langat
- Division of Global HIV & TB., Center for Global Health, U.S Centers for Disease Control and Prevention, USA
| | - Caren Mburu
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Jillian Neary
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Sarah Benki
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Dalton Wamalwa
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | | | - Dara A Lehman
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA
| | - Catherine A Blish
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
23
|
Girardi FM, Schuch LF, Martins MD. Oral cancer immunology: state of the art and future perspectives. Braz Oral Res 2024; 38:e129. [PMID: 39775418 DOI: 10.1590/1807-3107bor-2024.vol38.0129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 12/12/2022] [Indexed: 01/11/2025] Open
Abstract
Oral cancer is a multifactorial disease involving genetic, epigenetic, and environmental factors. The literature indicates that inflammatory cells at the advancing front of the tumor induce a host immune response, preventing the spread of the tumor. However, cancer cells adopt various continued strategies to circumvent this immune surveillance. The complexity of immune mechanisms suggests that there must be virtually individual patterns of anti-tumor immune responses. Due to this important interaction of cancer with the immune system, the objective of the present study was to provide an up-to-date overview of immuno-oncology focused on oral cancer, summarizing the basic immunology, the classic risk factors, immunotherapy, and future treatment and prognostic perspectives.
Collapse
Affiliation(s)
| | - Lauren Frenzel Schuch
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Oral Diagnosis, Piracicaba, SP, Brazil
| | - Manoela Domingues Martins
- Universidade Federal do Rio Grande do Sul - UFRS, School of Dentistry, Department of Oral Pathology, Porto Alegre, RS, Brazil
| |
Collapse
|
24
|
Luo X, Lv Y, Yang J, Long R, Qiu J, Deng Y, Tang G, Zhang C, Li J, Zuo J. Gamma delta T cells in cancer therapy: from tumor recognition to novel treatments. Front Med (Lausanne) 2024; 11:1480191. [PMID: 39748921 PMCID: PMC11693687 DOI: 10.3389/fmed.2024.1480191] [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: 08/13/2024] [Accepted: 12/09/2024] [Indexed: 01/04/2025] Open
Abstract
Traditional immunotherapies mainly focus on αβ T cell-based strategies, which depend on MHC-mediated antigen recognition. However, this approach poses significant challenges in treating recurrent tumors, as immune escape mechanisms are widespread. γδ T cells, with their ability for MHC-independent antigen presentation, offer a promising alternative that could potentially overcome limitations observed in traditional immunotherapies. These cells play a role in tumor immune surveillance through a unique mechanism of antigen recognition and synergistic interactions with other immune effector cells. In this review, we will discuss the biological properties of the Vδ1 and Vδ2 T subsets of γδ T cells, their immunomodulatory role within the tumor microenvironment, and the most recent clinical advances in γδ T cell-based related immunotherapies, including cell engaging strategies and adoptive cell therapy.
Collapse
Affiliation(s)
- Xinyu Luo
- The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yufan Lv
- The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jinsai Yang
- Computer Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Rou Long
- Transformation Research Lab, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jieya Qiu
- Transformation Research Lab, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuqi Deng
- Transformation Research Lab, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Guiyang Tang
- Transformation Research Lab, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Chaohui Zhang
- The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jiale Li
- Computer Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jianhong Zuo
- The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Computer Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Transformation Research Lab, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Third Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
25
|
Xu N, Wu Z, Pan J, Xu X, Wei Q. CAR-T cell therapy: Advances in digestive system malignant tumors. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200872. [PMID: 39377038 PMCID: PMC11456800 DOI: 10.1016/j.omton.2024.200872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Malignant tumors of the digestive system have had a notoriously dismal prognosis throughout history. Immunotherapy, radiotherapy, surgery, and chemotherapy are the primary therapeutic approaches for digestive system cancers. The rate of recurrence and metastasis, nevertheless, remains elevated. As one of the immunotherapies, chimeric antigen receptor T cell (CAR-T) therapy has demonstrated a promising antitumor effect in hematologic cancer. Despite undergoing numerous clinical trials, the ineffective antitumor effect and adverse effects of CAR-T cell therapy in the treatment of digestive system cancers continue to impede its clinical translation. It is necessary to surmount the restricted options for targeting proteins, the obstacles that impede CAR-T cell infiltration into solid tumors, and the limited survival time in vivo. We examined and summarized the developments, obstacles, and countermeasures associated with CAR-T therapy in digestive system cancers. Emphasis was placed on the regulatory functions of potential antigen targets, the tumor microenvironment, and immune evasion in CAR-T therapy. Thus, our analysis has furnished an all-encompassing comprehension of CAR-T cell therapy in digestive system cancers, which will generate tremendous enthusiasm for subsequent in-depth research into CAR-T-based therapies in digestive system cancers.
Collapse
Affiliation(s)
- Nan Xu
- Zhejiang University School of Medicine, Hangzhou 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Zhonglin Wu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Jun Pan
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiao Xu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310053, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Qiang Wei
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310053, China
| |
Collapse
|
26
|
Park S, Maus MV, Choi BD. CAR-T cell therapy for the treatment of adult high-grade gliomas. NPJ Precis Oncol 2024; 8:279. [PMID: 39702579 DOI: 10.1038/s41698-024-00753-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 10/30/2024] [Indexed: 12/21/2024] Open
Abstract
Treatment for malignant primary brain tumors, including glioblastoma, remains a significant challenge despite advances in therapy. CAR-T cell immunotherapy represents a promising alternative to conventional treatments. This review discusses the landscape of clinical trials for CAR-T cell therapy targeting brain tumors, highlighting key advancements like novel target antigens and combinatorial strategies designed to address tumor heterogeneity and immunosuppression, with the goal of improving outcomes for patients with these aggressive cancers.
Collapse
Affiliation(s)
- Sangwoo Park
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Bryan D Choi
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
27
|
Gibbs RJ, Chambers AC, Hill DJ. The emerging role of Fusobacteria in carcinogenesis. Eur J Clin Invest 2024; 54 Suppl 2:e14353. [PMID: 39674881 DOI: 10.1111/eci.14353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/04/2024] [Indexed: 12/17/2024]
Abstract
The Fusobacterium genus comprises Gram-negative, obligate anaerobic bacteria that typically reside in the periodontium of the oral cavity, gastrointestinal tract, and female genital tract. The association of Fusobacterial spp. with colorectal tumours is widely accepted, with further evidence that this pathogen may also be implicated in the development of other malignancies. Fusobacterial spp. influence malignant cell behaviours and the tumour microenvironment in various ways, which can be related to the multiple surface adhesins expressed. These adhesins include Fap2 (fibroblast-activated protein 2), CpbF (CEACAM binding protein of Fusobacteria), FadA (Fusobacterium adhesin A) and FomA (Fusobacterial outer membrane protein A). This review outlines the influence of Fusobacteria in promoting cancer initiation and progression, impacts of therapeutic outcomes and discusses potential therapeutic interventions where appropriate.
Collapse
|
28
|
Lim W, Iyer N. A GD (Gamma-Delta) type of cancel culture. IMMUNO-ONCOLOGY TECHNOLOGY 2024; 24:100740. [PMID: 39717204 PMCID: PMC11664092 DOI: 10.1016/j.iotech.2024.100740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
γδ T cells represent an 'unconventional' class of CD3+ lymphocytes with unique phenotypical and functional attributes that distinguishes them from their αβ T-cell receptor-expressing counterparts. Studies investigating the roles of γδ T cells in cancer have shown that these cells are indispensable for effective tumor control and their presence within the tumor may be of prognostic significance. Currently, there is significant interest in harnessing γδ T cells for cancer treatment, and research efforts have focused on the development of γδ T-cell-based strategies that are efficacious against cancer. Several therapeutic approaches using γδ T cells have been described, premised on the expansion of γδ T cells or γδ chimeric antigen receptor T therapy. The potential for broad, unbiased and 'off-the-shelf' applicability in cancer treatment, drives ongoing and future research and methodologies by which γδ T cells can be exploited for therapeutic use. In this review, we will briefly outline the characteristics of γδ T cells and describe how these work within and promote proper functioning of the cancer-immunity cycle. Additionally, we will introduce strategies that are less commonly described and may potentially be more efficacious than other types of therapy. Our discussion will expand upon presently known applications and even highlight the versatility of this immune subset as cancer therapeutics. γδ T-cell-based treatment is an emerging strategy and should be considered for cancelling cancer.
Collapse
Affiliation(s)
- W.K. Lim
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore
| | - N.G. Iyer
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
- Cancer Therapeutics Research Laboratory, National Cancer Centre, Singapore
- Department of Head and Neck Surgery, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
29
|
Lee S, Chae SJ, Jang IH, Oh SC, Kim SM, Lee SY, Kim JH, Ko J, Kim HJ, Song IC, Kim JK, Kim TD. B7H6 is the predominant activating ligand driving natural killer cell-mediated killing in patients with liquid tumours: evidence from clinical, in silico, in vitro, and in vivo studies. EBioMedicine 2024; 110:105459. [PMID: 39579618 PMCID: PMC11621501 DOI: 10.1016/j.ebiom.2024.105459] [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: 06/06/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Natural killer (NK) cells are a subset of innate lymphoid cells that are inherently capable of recognizing and killing infected or tumour cells. This has positioned NK cells as a promising live drug for tumour immunotherapy, but limited success suggests incomplete knowledge of their killing mechanism. NK cell-mediated killing involves a complex decision-making process based on integrating activating and inhibitory signals from various ligand-receptor repertoires. However, the relative importance of the different activating ligand-receptor interactions in triggering NK killing remains unclear. METHODS We employed a systematic approach combining clinical, in silico, in vitro, and in vivo data analysis to quantify the impact of various activating ligands. Clinical data analysis was conducted using massive pan-cancer data (n = 10,595), where patients with high NK cell levels were stratified using CIBERSORT. Subsequently, multivariate Cox regression and Kaplan-Meier (KM) survival analysis were performed based on activating ligand expression. To examine the impact of ligand expression on NK killing at the cellular level, we assessed surface expression of five major activating ligands (B7H6, MICA/B, ULBP1, ULBP2/5/6, and ULBP3) of human tumour cell lines of diverse origins (n = 33) via flow cytometry (FACs) and their NK cell-mediated cytotoxicity on by calcein-AM assay using human primary NK cells and NK-92 cell lines. Based on this data, we quantified the contribution of each activating ligand to the NK killing activity using mathematical models and Bayesian statistics. To further validate the results, we performed calcein-AM assays upon ligand knockdown and overexpression, conjugation assays, and co-culture assays in activating ligand-downregulated/overexpressed in liquid tumour (LT) cell lines. Moreover, we established LT-xenograft mouse models to assess the efficacy of NK cell targeting toward tumours with dominant ligands. FINDINGS Through the clinical analysis, we discovered that among nearly all 18 activating ligands, only patients with LT who were NK cell-rich and specifically had higher B7H6 level exhibited a favorable survival outcome (p = 0.0069). This unexpected dominant role of B7H6 was further confirmed by the analysis of datasets encompassing multiple ligands and a variety of tumours, which showed that B7H6 exhibited the highest contribution to NK killing among five representative ligands. Furthermore, LT cell lines (acute myeloid leukemia (AML), B cell lymphoma, and T-acute lymphocytic leukemia (ALL)) with lowered B7H6 demonstrated decreased susceptibility to NK cell-mediated cytotoxicity compared to those with higher levels. Even within the same cell line, NK cells selectively targeted cells with higher B7H6 levels. Finally, LT-xenograft mouse models (n = 24) confirmed that higher B7H6 results in less tumour burden and longer survival in NK cell-treated LT mice (p = 0.0022). INTERPRETATION While NK cells have gained attention for their potent anti-tumour effects without causing graft-versus-host disease (GvHD), thus making them a promising off-the-shelf therapy, our limited understanding of NK killing mechanisms has hindered their clinical application. This study illuminates the crucial role of the activating ligand B7H6 in driving NK cell killing, particularly in the context of LT. Therefore, the expression level of B7H6 could serve as a prognostic marker for patients with LT. Moreover, for the development of NK cell-based immunotherapy, focusing on increasing the level of B7H6 on its cognate receptor, NKp30, could be the most effective strategy. FUNDING This work was supported by the National Research Council of Science & Technology (NST) grant (CAP-18-02-KRIBB, GTL24021-000), a National Research Foundation grant (2710012258, 2710004815), and an Institute for Basic Science grant (IBS-R029-C3).
Collapse
Affiliation(s)
- Sunyoung Lee
- Center for Cell and Gene Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; Division of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Seok Joo Chae
- Department of Mathematical Sciences, KAIST, Daejeon, 34141, Republic of Korea; Biomedical Mathematics Group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon, 34126, Republic of Korea; Department of Bioengineering, Rice University, Houston, TX, 77005, USA
| | - In-Hwan Jang
- Center for Cell and Gene Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Se-Chan Oh
- Center for Cell and Gene Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Seok-Min Kim
- Center for Cell and Gene Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Soo Yun Lee
- Center for Cell and Gene Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Ji Hyun Kim
- Center for Cell and Gene Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Jesang Ko
- Division of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Hang J Kim
- Division of Statistics and Data Science, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Ik-Chan Song
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, 35015, South Korea
| | - Jae Kyoung Kim
- Department of Mathematical Sciences, KAIST, Daejeon, 34141, Republic of Korea; Biomedical Mathematics Group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon, 34126, Republic of Korea; Department of Medicine, College of Medicine, Korea University, Seoul, 02481, Republic of Korea.
| | - Tae-Don Kim
- Center for Cell and Gene Therapy, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; Biomedical Mathematics Group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon, 34126, Republic of Korea; KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea; Department of Biopharmaceutical Convergence, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
30
|
Greppi M, De Franco F, Obino V, Rebaudi F, Goda R, Frumento D, Vita G, Baronti C, Melaiu O, Bozzo M, Candiani S, Vellone VG, Papaccio F, Pesce S, Marcenaro E. NK cell receptors in anti-tumor and healthy tissue protection: Mechanisms and therapeutic advances. Immunol Lett 2024; 270:106932. [PMID: 39303993 DOI: 10.1016/j.imlet.2024.106932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/10/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Natural Killer (NK) cells are integral to the innate immune system, renowned for their ability to target and eliminate cancer cells without the need for antigen presentation, sparing normal tissues. These cells are crucial in cancer immunosurveillance due to their diverse array of activating and inhibitory receptors that modulate their cytotoxic activity. However, the tumor microenvironment can suppress NK cell function through various mechanisms. Over recent decades, research has focused on overcoming these tumor escape mechanisms. Initially, efforts concentrated on enhancing T cell activity, leading to impressive results with immunotherapeutic approaches aimed at boosting T cell responses. Nevertheless, a substantial number of patients do not benefit from these treatments and continue to seek effective alternatives. In this context, NK cells present a promising avenue for developing new treatments, given their potent cytotoxic capabilities, safety profile, and activity against T cell-resistant tumors, such as those lacking HLA-I expression. Recent advancements in immunotherapy include strategies to restore and amplify NK cell activity through immune checkpoint inhibitors, cytokines, adoptive NK cell therapy, and CAR-NK cell technology. This review provides a comprehensive overview of NK cell receptors, the tumor escape mechanisms that hinder NK cell function, and the evolving field of NK cell-based cancer immunotherapy, highlighting ongoing efforts to develop more effective and targeted cancer treatment strategies.
Collapse
Affiliation(s)
- Marco Greppi
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Fabiana De Franco
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Valentina Obino
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Federico Rebaudi
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Rayan Goda
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Davide Frumento
- Department of Education Sciences, University of Rome Tre, Rome, Italy
| | - Giorgio Vita
- Department of Internal Medicine (DIMI), University of Genoa, Genoa, Italy
| | - Camilla Baronti
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Ombretta Melaiu
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Matteo Bozzo
- Department of Earth, Environmental and Life Sciences (DISTAV), University of Genoa, Genoa, Italy
| | - Simona Candiani
- Department of Earth, Environmental and Life Sciences (DISTAV), University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Valerio G Vellone
- Department of Integrated Surgical and Diagnostic Sciences (DISC), University of Genoa, Genoa, Italy; Pathology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Federica Papaccio
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy.
| | - Silvia Pesce
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Emanuela Marcenaro
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| |
Collapse
|
31
|
Qiao W, Dong P, Chen H, Zhang J. Advances in Induced Pluripotent Stem Cell-Derived Natural Killer Cell Therapy. Cells 2024; 13:1976. [PMID: 39682724 PMCID: PMC11640743 DOI: 10.3390/cells13231976] [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: 10/11/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Natural killer (NK) cells are cytotoxic lymphocytes of the innate immune system capable of killing virus-infected cells and/or cancer cells. The commonly used NK cells for therapeutic applications include primary NK cells and immortalized NK cell lines. However, primary NK cell therapy faces limitations due to its restricted proliferation capacity and challenges in stable storage. Meanwhile, the immortalized NK-92 cell line requires irradiation prior to infusion, which reduces its cytotoxic activity, providing a ready-made alternative and overcoming these bottlenecks. Recent improvements in differentiation protocols for iPSC-derived NK cells have facilitated the clinical production of iPSC-NK cells. Moreover, iPSC-NK cells can be genetically modified to enhance tumor targeting and improve the expansion and persistence of iPSC-NK cells, thereby achieving more robust antitumor efficacy. This paper focuses on the differentiation-protocols efforts of iPSC-derived NK cells and the latest progress in iPSC-NK cell therapy. Additionally, we discuss the current challenges faced by iPSC-NK cells and provide an outlook on future applications and developments.
Collapse
Affiliation(s)
- Wenhua Qiao
- CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China;
| | - Peng Dong
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China;
| | - Hui Chen
- CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China;
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China;
| | - Jianmin Zhang
- CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China;
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China;
| |
Collapse
|
32
|
Noh KM, Jangid AK, Park J, Kim S, Kim K. Membrane-immobilized gemcitabine for cancer-targetable NK cell surface engineering. J Mater Chem B 2024; 12:12087-12102. [PMID: 39465499 DOI: 10.1039/d4tb01639d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Although natural killer (NK) cell-based adoptive cell transfer (ACT) has shown promise in cancer immunotherapy, its efficacy against solid tumors is limited in the immunosuppressive tumor microenvironment (TME). Combinatorial therapies involving chemotherapeutic drugs such as gemcitabine (Gem) and NK cells have been developed to modulate the TME; however, their clinical application is constrained by low drug delivery efficiency and significant off-target toxicity. In this study, we developed cell membrane-immobilized Gem conjugates (i.e., lipid-Gem conjugates), designed to anchor seamlessly onto NK cell surfaces. Our modular-designed ex vivo cell surface engineeringmaterials comprise a lipid anchor for membrane immobilization, poly(ethylene glycol) to inhibit endocytosis, a disulfide bond as cleavable linker by glutathione (GSH) released during cancer cell lysis, and Gem for targeted sensitization. We demonstrated that the intrinsic properties of NK cells, such as proliferation and surface ligand availability, were preserved despite coating with lipid-Gem conjugates. Moreover, delivery of Gem prodrugs by lipid-Gem coated NK (GCNK) cells was shown to enhance antitumor efficacy against pancreatic cancer cells (PANC-1) through the following mechanisms: (1) NK cells recognized and attacked cancer cells, (2) intracellular GSH was leaked out from the lysed cancer cells, enabling cleavage of disulfide bond, (3) released Gem from the GCNK cells delivered to the target cells, (4) Gem upregulated MHC class I-related chain A and B on cancer cells, and (5) thereby activating NK cells led to enhance antitumor efficacy. The simultaneous co-delivery of membrane-immobilized Gem with NK cells could potentially facilitate both immune synapse-mediated cancer recognition and chemotherapeutic effects, offering a promising approach to enhance the anticancer efficacy of conventional ACTs.
Collapse
Affiliation(s)
- Kyung Mu Noh
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea.
| | - Ashok Kumar Jangid
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea.
| | - Jaewon Park
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea.
| | - Sungjun Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea.
| | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea.
| |
Collapse
|
33
|
Zhou Y, Song L, Li H. Full-resolution HLA and KIR gene annotations for human genome assemblies. Genome Res 2024; 34:1931-1941. [PMID: 38839374 PMCID: PMC11610593 DOI: 10.1101/gr.278985.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024]
Abstract
The human leukocyte antigen (HLA) genes and the killer cell immunoglobulin-like receptor (KIR) genes are critical to immune responses and are associated with many immune-related diseases. Located in highly polymorphic regions, it is difficult to study them with traditional short-read alignment-based methods. Although modern long-read assemblers can often assemble these genes, using existing tools to annotate HLA and KIR genes in these assemblies remains a nontrivial task. Here, we describe Immuannot, a new computation tool to annotate the gene structures of HLA and KIR genes and to type the allele of each gene. Applying Immuannot to 56 regional and 212 whole-genome assemblies from previous studies, we annotate 9931 HLA and KIR genes and found that almost half of these genes, 4068, have novel sequences compared with the current Immuno Polymorphism Database (IPD). These novel gene sequences are represented by 2664 distinct alleles, some of which contained nonsynonymous variations, resulting in 92 novel protein sequences. We demonstrate the complex haplotype structures at the two loci and report the linkage between HLA/KIR haplotypes and gene alleles. We anticipate that Immuannot will speed up the discovery of new HLA/KIR alleles and enable the association of HLA/KIR haplotype structures with clinical outcomes in the future.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | - Li Song
- Department of Biomedical Data Science, Dartmouth College, Hanover, New Hampshire 03755, USA
| | - Heng Li
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA;
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
34
|
Demaria O, Habif G, Vetizou M, Gauthier L, Remark R, Chiossone L, Vagne C, Rebuffet L, Courtois R, Denis C, Le Floch F, Muller M, Girard-Madoux M, Augier S, Lopez J, Carrette B, Maguer A, Vallier JB, Grondin G, Baron W, Galluso J, Yessaad N, Giordano M, Simon L, Chanuc F, Alvarez AB, Perrot I, Bonnafous C, Represa A, Rossi B, Morel A, Morel Y, Paturel C, Vivier E. A tetraspecific engager armed with a non-alpha IL-2 variant harnesses natural killer cells against B cell non-Hodgkin lymphoma. Sci Immunol 2024; 9:eadp3720. [PMID: 39546590 DOI: 10.1126/sciimmunol.adp3720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/07/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024]
Abstract
NK cells offer a promising alternative to T cell therapies in cancer. We evaluated IPH6501, a clinical-stage, tetraspecific NK cell engager (NKCE) armed with a non-alpha IL-2 variant (IL-2v), which targets CD20 and was developed for treating B cell non-Hodgkin lymphoma (B-NHL). CD20-NKCE-IL2v boosts NK cell proliferation and cytotoxicity, showing activity against a range of B-NHL cell lines, including those with low CD20 density. Whereas it presented reduced toxicities compared with those commonly associated with T cell therapies, CD20-NKCE-IL2v showed greater killing efficacy over a T cell engager targeting CD20 in in vitro preclinical models. CD20-NKCE-IL2v also increased the cell surface expression of NK cell-activating receptors, leading to activity against CD20-negative tumor cells. In vivo studies in nonhuman primates and tumor mouse models further validated its efficacy and revealed that CD20-NKCE-IL2v induces peripheral NK cell homing at the tumor site. CD20-NKCE-IL2v emerges as a potential alternative in the treatment landscape of B-NHL.
Collapse
Affiliation(s)
- Olivier Demaria
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Guillaume Habif
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Marie Vetizou
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Laurent Gauthier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Romain Remark
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Laura Chiossone
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Constance Vagne
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Lucas Rebuffet
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Rachel Courtois
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Caroline Denis
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - François Le Floch
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Marianna Muller
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | | | - Séverine Augier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Julie Lopez
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Barbara Carrette
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Aurélie Maguer
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | | | | | - William Baron
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Justine Galluso
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Nadia Yessaad
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Marilyn Giordano
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Léa Simon
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Fabien Chanuc
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | | | - Ivan Perrot
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Cécile Bonnafous
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Agnès Represa
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Benjamin Rossi
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Ariane Morel
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Yannis Morel
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Carine Paturel
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- APHM, Hôpital de la Timone, Marseille-Immunopôle Profiling Platform, Marseille, France
- Paris-Saclay Cancer Cluster, Le Kremlin-Bicêtre, France
- Université Paris-Saclay, Gustave Roussy, INSERM, Prédicteurs moléculaires et nouvelles cibles en oncologie, 94800, Villejuif, France
| |
Collapse
|
35
|
Chen S, Zhu H, Jounaidi Y. Comprehensive snapshots of natural killer cells functions, signaling, molecular mechanisms and clinical utilization. Signal Transduct Target Ther 2024; 9:302. [PMID: 39511139 PMCID: PMC11544004 DOI: 10.1038/s41392-024-02005-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/25/2024] [Accepted: 09/17/2024] [Indexed: 11/15/2024] Open
Abstract
Natural killer (NK) cells, initially identified for their rapid virus-infected and leukemia cell killing and tumor destruction, are pivotal in immunity. They exhibit multifaceted roles in cancer, viral infections, autoimmunity, pregnancy, wound healing, and more. Derived from a common lymphoid progenitor, they lack CD3, B-cell, or T-cell receptors but wield high cytotoxicity via perforin and granzymes. NK cells orchestrate immune responses, secreting inflammatory IFNγ or immunosuppressive TGFβ and IL-10. CD56dim and CD56bright NK cells execute cytotoxicity, while CD56bright cells also regulate immunity. However, beyond the CD56 dichotomy, detailed phenotypic diversity reveals many functional subsets that may not be optimal for cancer immunotherapy. In this review, we provide comprehensive and detailed snapshots of NK cells' functions and states of activation and inhibitions in cancer, autoimmunity, angiogenesis, wound healing, pregnancy and fertility, aging, and senescence mediated by complex signaling and ligand-receptor interactions, including the impact of the environment. As the use of engineered NK cells for cancer immunotherapy accelerates, often in the footsteps of T-cell-derived engineering, we examine the interactions of NK cells with other immune effectors and relevant signaling and the limitations in the tumor microenvironment, intending to understand how to enhance their cytolytic activities specifically for cancer immunotherapy.
Collapse
Affiliation(s)
- Sumei Chen
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China.
| | - Haitao Zhu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Youssef Jounaidi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
36
|
Chaoul N, Lauricella E, Giglio A, D'Angelo G, Ganini C, Cives M, Porta C. The future of cellular therapy for the treatment of renal cell carcinoma. Expert Opin Biol Ther 2024; 24:1245-1259. [PMID: 39485013 DOI: 10.1080/14712598.2024.2418321] [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: 06/13/2024] [Revised: 08/26/2024] [Accepted: 10/15/2024] [Indexed: 11/03/2024]
Abstract
INTRODUCTION Systemic treatment options for renal cell carcinoma (RCC) have expanded considerably in recent years, and both tyrosine kinase inhibitors and immune checkpoint inhibitors, alone or in combination, have entered the clinical arena. Adoptive cell immunotherapies have recently revolutionized the treatment of cancer and hold the promise to further advance the treatment of RCC. AREAS COVERED In this review, we summarize the latest preclinical and clinical development in the field of adoptive cell immunotherapy for the treatment of RCC, focusing on lymphokine-activated killer (LAK) cells, cytokine-induced killer (CIK) cells, tumor-infiltrating T cells (TILs), TCR-engineered T cells, chimeric antigen receptor (CAR) T cells, and dendritic cell vaccination strategies. Perspectives on emerging cellular products including CAR NK cells, CAR macrophages, as well as γδ T cells are also included. EXPERT OPINION So far, areas of greater therapeutic success of adoptive cell therapies include the adjuvant administration of CIK cells and the transfer of anti-CD70 CAR T cells in patients with metastatic RCC. Bench to bedside and back research will be needed to overcome current limitations of adoptive cell therapies in RCC, primarily aiming at improving the safety of immune cell products, optimizing their antitumor activity and generating off-the-shelf products ready for clinical use.
Collapse
Affiliation(s)
- Nada Chaoul
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Eleonora Lauricella
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Andrea Giglio
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Gabriella D'Angelo
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Carlo Ganini
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy
| | - Mauro Cives
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy
| | - Camillo Porta
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy
| |
Collapse
|
37
|
Almutairi A, Alshehri NA, Al Subayyil A, Bahattab E, Alshabibi M, Abomaray F, Basmaeil YS, Khatlani T. Human decidua basalis mesenchymal stem/stromal cells enhance anticancer properties of human natural killer cells, in vitro. Front Cell Dev Biol 2024; 12:1435484. [PMID: 39539962 PMCID: PMC11557523 DOI: 10.3389/fcell.2024.1435484] [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: 05/20/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Mesenchymal stem cells/stromal cells from the Decidua Basalis of the human placenta (DBMSCs) express wide range of effector molecules that modulate the functions of their target cells. These properties make them potential candidate for use in cellular therapy. In this study, we have investigated the consequences of interaction between DBMSCs and natural killer (NK) cells for both cell types. Methods DBMSCs were cultured with IL-2-activated and resting non-activated NK cells isolated from healthy human peripheral blood and various functional assays were performed including, NK cell proliferation and cytolytic activities. Flow cytometry and microscopic studies were performed to examine the expression of NK cell receptors that mediate these cytolytic activities against DBMSCs. Moreover, the mechanism underlying these effects was also investigated. Results Our findings revealed that, co-culture of DBMSCs and NK cells resulted in inhibition of proliferation of resting NK cells, while proliferation of IL-2 activated NK cells was increased. Contrarily, treatment of DBMSC's with comparatively high numbers of IL-2 activated NK cells, resulted in their lysis, whereas treatment with low numbers resulted in reduction in their proliferation. Cytolytic activity of NK cells against DBMSCs was mediated by several activating NK cell receptors. In spite of the expression of HLA class I molecules by DBMSCs, they were still lysed by NK cells, excluding their involvement in cytolytic activity. In addition, preconditioning NK cells by DBMSCs, enhanced their ability to suppress tumor cell proliferation and in severe cases resulted in their partial lysis. Lysis and decrease of tumor cell proliferation is associated with increased expression of important molecules involved in anticancer activities. Discussion We conclude that DBMSCs exhibit dualfunctions on NK cells that enhance their anticancer therapeutic potential.
Collapse
Affiliation(s)
- Abdulaziz Almutairi
- College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdul Aziz University for Medical Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh, Saudi Arabia
| | - Najlaa A. Alshehri
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdul Aziz University for Medical Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh, Saudi Arabia
- School of Education, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Abdullah Al Subayyil
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdul Aziz University for Medical Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh, Saudi Arabia
| | - Eman Bahattab
- National Center for Stem Cell Technology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Manal Alshabibi
- National Center for Stem Cell Technology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Fawaz Abomaray
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet, Stockholm, Sweden
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yasser S. Basmaeil
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdul Aziz University for Medical Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh, Saudi Arabia
| | - Tanvir Khatlani
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdul Aziz University for Medical Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Ministry of National Guard Health Affairs (MNGHA), Riyadh, Saudi Arabia
| |
Collapse
|
38
|
Ariolli A, Agolini E, Mazza T, Petrizzelli F, Petrini S, D’Oria V, Cudini A, Nardella C, Pesce V, Comparcola D, Cappa M, Fierabracci A. The Putative Role of TIM-3 Variants in Polyendocrine Autoimmunity: Insights from a WES Investigation. Int J Mol Sci 2024; 25:10994. [PMID: 39456777 PMCID: PMC11506967 DOI: 10.3390/ijms252010994] [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: 08/28/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Autoimmune polyglandular syndrome (APS) comprises a complex association of autoimmune pathological conditions. APS Type 1 originates from loss-of-function mutations in the autoimmune regulator (AIRE) gene. APS2, APS3 and APS4 are linked to specific HLA alleles within the major histocompatibility complex, with single-nucleotide polymorphisms (SNPs) in non-HLA genes also contributing to disease. In general, variability in the AIRE locus and the presence of heterozygous loss-of-function mutations can impact self-antigen presentation in the thymus. In this study, whole-exome sequencing (WES) was performed on a sixteen-year-old female APS3A/B patient to investigate the genetic basis of her complex phenotype. The analysis identified two variants (p.Arg111Trp and p.Thr101Ile) of the hepatitis A virus cell receptor 2 gene (HAVCR2) encoding for the TIM-3 (T cell immunoglobulin and mucin domain 3) protein. These variants were predicted, through in silico analysis, to impact protein structure and stability, potentially influencing the patient's autoimmune phenotype. While confocal microscopy analysis revealed no alteration in TIM-3 fluorescence intensity between the PBMCs isolated from the patient and those of a healthy donor, RT-qPCR showed reduced TIM-3 expression in the patient's unfractionated PBMCs. A screening conducted on a cohort of thirty APS patients indicated that the p.Thr101Ile and p.Arg111Trp mutations were unique to the proband. This study opens the pathway for the search of TIM-3 variants possibly linked to complex autoimmune phenotypes, highlighting the potential of novel variant discovery in contributing to APS classification and diagnosis.
Collapse
Affiliation(s)
- Andrea Ariolli
- Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00146 Rome, Italy; (A.A.); (T.M.); (A.C.); (C.N.); (V.P.); (D.C.)
| | - Emanuele Agolini
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00146 Rome, Italy;
| | - Tommaso Mazza
- Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00146 Rome, Italy; (A.A.); (T.M.); (A.C.); (C.N.); (V.P.); (D.C.)
- Laboratory of Bioinformatics, Casa Sollievo della Sofferenza, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 70013 San Giovanni Rotondo, Foggia, Italy;
| | - Francesco Petrizzelli
- Laboratory of Bioinformatics, Casa Sollievo della Sofferenza, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 70013 San Giovanni Rotondo, Foggia, Italy;
| | - Stefania Petrini
- Confocal Microscopy Core Facility, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00146 Rome, Italy; (S.P.); (V.D.)
| | - Valentina D’Oria
- Confocal Microscopy Core Facility, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00146 Rome, Italy; (S.P.); (V.D.)
| | - Annamaria Cudini
- Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00146 Rome, Italy; (A.A.); (T.M.); (A.C.); (C.N.); (V.P.); (D.C.)
| | - Caterina Nardella
- Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00146 Rome, Italy; (A.A.); (T.M.); (A.C.); (C.N.); (V.P.); (D.C.)
| | - Vanessa Pesce
- Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00146 Rome, Italy; (A.A.); (T.M.); (A.C.); (C.N.); (V.P.); (D.C.)
| | - Donatella Comparcola
- Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00146 Rome, Italy; (A.A.); (T.M.); (A.C.); (C.N.); (V.P.); (D.C.)
| | - Marco Cappa
- Research Unit Innovative Therapies for Endocrinopathies, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00146 Rome, Italy;
| | - Alessandra Fierabracci
- Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00146 Rome, Italy; (A.A.); (T.M.); (A.C.); (C.N.); (V.P.); (D.C.)
| |
Collapse
|
39
|
Toyoda H, Kuramasu A, Hosonuma M, Murayama M, Narikawa Y, Isobe J, Baba Y, Tajima K, Funayama E, Shida M, Maruyama Y, Sasaki A, Hirasawa Y, Tsurui T, Ariizumi H, Ishiguro T, Suzuki R, Kobayashi S, Horiike A, Hida N, Sambe T, Nobe K, Wada S, Kobayashi H, Tsuji M, Kobayashi S, Tsunoda T, Kudo Y, Kiuchi Y, Yoshimura K. MHC class I polypeptide-related sequence B shedding modulates pancreatic tumor immunity via the activation of NKG2D Low T cells. Sci Rep 2024; 14:23401. [PMID: 39379424 PMCID: PMC11461622 DOI: 10.1038/s41598-024-73712-1] [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: 02/16/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024] Open
Abstract
Natural killer group 2 member D ligands (NKG2DLs) are expressed as stress response proteins in cancer cells. NKG2DLs induce immune cell activation or tumor escape responses, depending on their expression. Human pancreatic cancer cells, PANC-1, express membrane MHC class I polypeptide-related sequence A/B (mMICA/B), whereas soluble MICB (sMICB) is detected in the culture supernatant. We hypothesized that sMICB saturates NKG2D in NKG2DLow T cells and inhibits the activation signal from mMICB to NKG2D. Knockdown of MICB by siRNA reduced sMICB level, downregulated mMICB expression, maintained NKG2DLow T cell activation, and inhibited NKG2DHigh T cell activation. To maintain mMICB expression and downregulate sMICB expression, we inhibited a disintegrin and metalloproteinase (ADAM), a metalloproteinase that sheds MICB. Subsequently, the shedding of MICB was prevented using ADAM17 inhibitors, and the activation of NKG2DLow T cells was maintained. In vivo xenograft model revealed that NKG2DHigh T cells have superior anti-tumor activity. These results elucidate the mechanism of immune escape via sMICB and show potential for the activation of NKG2DLow T cells within the tumor microenvironment.
Collapse
Affiliation(s)
- Hitoshi Toyoda
- Department of Clinical Immuno Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya-ku, Tokyo, 157-8577, Japan
- Department of Pharmacology, Showa University School of Medicine, Tokyo, 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
- Department of Orthopedic Surgery, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Atsuo Kuramasu
- Department of Clinical Immuno Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya-ku, Tokyo, 157-8577, Japan
| | - Masahiro Hosonuma
- Department of Clinical Immuno Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya-ku, Tokyo, 157-8577, Japan
- Department of Pharmacology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Masakazu Murayama
- Department of Clinical Immuno Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya-ku, Tokyo, 157-8577, Japan
- Department of Pharmacology, Showa University School of Medicine, Tokyo, 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
- Department of Otorhinolaryngology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Yoichiro Narikawa
- Department of Clinical Immuno Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya-ku, Tokyo, 157-8577, Japan
- Department of Pharmacology, Showa University School of Medicine, Tokyo, 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
- Department of Otorhinolaryngology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Junya Isobe
- Department of Hospital Pharmaceutics, Showa University School of Pharmacy, Tokyo, 142-8555, Japan
| | - Yuta Baba
- Department of Clinical Immuno Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya-ku, Tokyo, 157-8577, Japan
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Kohei Tajima
- Department of Clinical Immuno Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya-ku, Tokyo, 157-8577, Japan
- Department of Gastroenterological Surgery, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Eiji Funayama
- Department of Clinical Immuno Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya-ku, Tokyo, 157-8577, Japan
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
- Division of Pharmacology, Department of Pharmacology, Toxicology and Therapeutics, School of Pharmacy, Showa University, Tokyo, 142-8555, Japan
| | - Midori Shida
- Department of Clinical Immuno Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya-ku, Tokyo, 157-8577, Japan
| | - Yuki Maruyama
- Department of Clinical Immuno Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya-ku, Tokyo, 157-8577, Japan
- Department of Pharmacology, Showa University School of Medicine, Tokyo, 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Aya Sasaki
- Department of Clinical Immuno Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya-ku, Tokyo, 157-8577, Japan
- Department of Pharmacology, Showa University School of Medicine, Tokyo, 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Yuya Hirasawa
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Toshiaki Tsurui
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Hirotsugu Ariizumi
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Tomoyuki Ishiguro
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Risako Suzuki
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Sei Kobayashi
- Department of Otorhinolaryngology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Atsushi Horiike
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Noriko Hida
- Division of Clinical Pharmacology, Department of Pharmacology, Showa University School of Medicine, Tokyo, 142-8555, Japan
- Division of Clinical Research and Development, Department of Clinical Pharmacy, School of Pharmacy, Showa University, Tokyo, 142-8555, Japan
| | - Takehiko Sambe
- Division of Clinical Research and Development, Department of Clinical Pharmacy, School of Pharmacy, Showa University, Tokyo, 142-8555, Japan
| | - Koji Nobe
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
- Division of Pharmacology, Department of Pharmacology, Toxicology and Therapeutics, School of Pharmacy, Showa University, Tokyo, 142-8555, Japan
| | - Satoshi Wada
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, 157-8577, Japan
| | - Hitome Kobayashi
- Department of Otorhinolaryngology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Mayumi Tsuji
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Shinichi Kobayashi
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, 157-8577, Japan
| | - Takuya Tsunoda
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Yoshifumi Kudo
- Department of Orthopedic Surgery, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Yuji Kiuchi
- Department of Pharmacology, Showa University School of Medicine, Tokyo, 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Kiyoshi Yoshimura
- Department of Clinical Immuno Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kita-karasuyama, Setagaya-ku, Tokyo, 157-8577, Japan.
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, 142-8555, Japan.
| |
Collapse
|
40
|
Gunasena M, Alles M, Wijewantha Y, Mulhern W, Bowman E, Gabriel J, Kettelhut A, Kumar A, Weragalaarachchi K, Kasturiratna D, Horowitz JC, Scrape S, Pannu SR, Liu SL, Vilgelm A, Wijeratne S, Bednash JS, Demberg T, Funderburg NT, Liyanage NP. Synergy Between NK Cells and Monocytes in Potentiating Cardiovascular Disease Risk in Severe COVID-19. Arterioscler Thromb Vasc Biol 2024; 44:e243-e261. [PMID: 38989579 PMCID: PMC11448863 DOI: 10.1161/atvbaha.124.321085] [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: 04/15/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Evidence suggests that COVID-19 predisposes to cardiovascular diseases (CVDs). While monocytes/macrophages play a central role in the immunopathogenesis of atherosclerosis, less is known about their immunopathogenic mechanisms that lead to CVDs during COVID-19. Natural killer (NK) cells, which play an intermediary role during pathologies like atherosclerosis, are dysregulated during COVID-19. Here, we sought to investigate altered immune cells and their associations with CVD risk during severe COVID-19. METHODS We measured plasma biomarkers of CVDs and determined phenotypes of circulating immune subsets using spectral flow cytometry. We compared these between patients with severe COVID-19 (severe, n=31), those who recovered from severe COVID-19 (recovered, n=29), and SARS-CoV-2-uninfected controls (controls, n=17). In vivo observations were supported using in vitro assays to highlight possible mechanistic links between dysregulated immune subsets and biomarkers during and after COVID-19. We performed multidimensional analyses of published single-cell transcriptome data of monocytes and NK cells during severe COVID-19 to substantiate in vivo findings. RESULTS During severe COVID-19, we observed alterations in cardiometabolic biomarkers including oxidized-low-density lipoprotein, which showed decreased levels in severe and recovered groups. Severe patients exhibited dysregulated monocyte subsets, including increased frequencies of proinflammatory intermediate monocytes (also observed in the recovered) and decreased nonclassical monocytes. All identified NK-cell subsets in the severe COVID-19 group displayed increased expression of activation and tissue-resident markers, such as CD69 (cluster of differentiation 69). We observed significant correlations between altered immune subsets and plasma oxidized-low-density lipoprotein levels. In vitro assays revealed increased uptake of oxidized-low-density lipoprotein into monocyte-derived macrophages in the presence of NK cells activated by plasma of patients with severe COVID-19. Transcriptome analyses confirmed enriched proinflammatory responses and lipid dysregulation associated with epigenetic modifications in monocytes and NK cells during severe COVID-19. CONCLUSIONS Our study provides new insights into the involvement of monocytes and NK cells in the increased CVD risk observed during and after COVID-19.
Collapse
Affiliation(s)
- Manuja Gunasena
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
- Department of Veterinary Bioscience, College of Veterinary Medicine, The Ohio State University
| | - Mario Alles
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
| | - Yasasvi Wijewantha
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
| | - Will Mulhern
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
| | - Emily Bowman
- School of Health and Rehabilitation Sciences, College of Medicine, the Ohio State University
| | - Janelle Gabriel
- School of Health and Rehabilitation Sciences, College of Medicine, the Ohio State University
| | - Aaren Kettelhut
- School of Health and Rehabilitation Sciences, College of Medicine, the Ohio State University
| | - Amrendra Kumar
- Department of pathology, College of Medicine, The Ohio State University
| | | | - Dhanuja Kasturiratna
- Department of Mathematics and Statistics, Northern Kentucky University, KY, Highland Heights, KY, USA
| | - Jeffrey C Horowitz
- Department of Internal Medicine, College of Medicine, The Ohio State University
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University
| | - Scott Scrape
- Department of pathology, College of Medicine, The Ohio State University
| | - Sonal R Pannu
- Department of Internal Medicine, College of Medicine, The Ohio State University
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University
| | - Shan-Lu Liu
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
- Department of Veterinary Bioscience, College of Veterinary Medicine, The Ohio State University
| | - Anna Vilgelm
- Department of pathology, College of Medicine, The Ohio State University
| | - Saranga Wijeratne
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Joseph S Bednash
- Department of Internal Medicine, College of Medicine, The Ohio State University
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University
| | - Thorsten Demberg
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Nicholas T Funderburg
- Department of Veterinary Bioscience, College of Veterinary Medicine, The Ohio State University
| | - Namal P.M. Liyanage
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University
- Department of Veterinary Bioscience, College of Veterinary Medicine, The Ohio State University
| |
Collapse
|
41
|
Dong B, Obermajer N, Tsuji T, Matsuzaki J, Bonura CM, Sander C, Withers H, Long MD, Chavel C, Olejniczak SH, Minderman H, Kirkwood JM, Edwards RP, Storkus WJ, Romero P, Kalinski P. NK Receptor Signaling Lowers TCR Activation Threshold, Enhancing Selective Recognition of Cancer Cells by TAA-Specific CTLs. Cancer Immunol Res 2024; 12:1421-1437. [PMID: 38949179 PMCID: PMC11706306 DOI: 10.1158/2326-6066.cir-24-0061] [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: 01/16/2024] [Revised: 05/10/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024]
Abstract
Cytotoxic CD8+ T lymphocyte (CTL) recognition of non-mutated tumor-associated antigens (TAA), present on cancer cells and also in healthy tissues, is an important element of cancer immunity, but the mechanism of its selectivity for cancer cells and opportunities for its enhancement remain elusive. In this study, we found that CTL expression of the NK receptors (NKR) DNAM1 and NKG2D was associated with the effector status of CD8+ tumor-infiltrating lymphocytes and long-term survival of patients with melanoma. Using MART1 and NY-ESO-1 as model TAAs, we demonstrated that DNAM1 and NKG2D regulate T-cell receptor (TCR) functional avidity and set the threshold for TCR activation of human TAA-specific CTLs. Superior co-stimulatory effects of DNAM1 over CD28 involved enhanced TCR signaling, CTL killer function, and polyfunctionality. Double transduction of human CTLs with TAA-specific TCR and NKRs resulted in strongly enhanced antigen sensitivity, without a reduction in antigen specificity and selectivity of killer function. In addition, the elevation of NKR ligand expression on cancer cells due to chemotherapy also increased CTL recognition of cancer cells expressing low levels of TAAs. Our data help explain the ability of self-antigens to mediate tumor rejection in the absence of autoimmunity and support the development of dual-targeting adoptive T-cell therapies that use NKRs to enhance the potency and selectivity of recognition of TAA-expressing cancer cells.
Collapse
MESH Headings
- Humans
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Signal Transduction
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Melanoma/immunology
- Melanoma/metabolism
- NK Cell Lectin-Like Receptor Subfamily K/metabolism
- Lymphocyte Activation/immunology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Antigens, Differentiation, T-Lymphocyte/immunology
- Cell Line, Tumor
- MART-1 Antigen/immunology
- MART-1 Antigen/metabolism
- Cytotoxicity, Immunologic
- T Lineage-Specific Activation Antigen 1
Collapse
Affiliation(s)
- Bowen Dong
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
- Department of Medicine, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Nataša Obermajer
- Department of Surgery, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| | - Takemasa Tsuji
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Junko Matsuzaki
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Cindy M. Bonura
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Cindy Sander
- Department of Medicine, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| | - Henry Withers
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Mark D. Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Colin Chavel
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Scott H. Olejniczak
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Hans Minderman
- Department of Flow and Immune Analysis Shared Resource, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - John M. Kirkwood
- Department of Medicine, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| | - Robert P. Edwards
- Department of OB-GYN, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| | - Walter J. Storkus
- Department of Dermatology, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| | - Pedro Romero
- University of Lausanne and Ludwig Institute for Cancer Research; Lausanne, Switzerland
| | - Pawel Kalinski
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
- Department of Medicine, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
- Department of Surgery, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| |
Collapse
|
42
|
Xu J, Gao H, Azhar MS, Xu H, Chen S, Li M, Ni X, Yan T, Zhou H, Long Q, Yi W. Interleukin signaling in the regulation of natural killer cells biology in breast cancer. Front Immunol 2024; 15:1449441. [PMID: 39380989 PMCID: PMC11459090 DOI: 10.3389/fimmu.2024.1449441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
In the field of breast cancer treatment, the immunotherapy involving natural killer (NK) cells is increasingly highlighting its distinct potential and significance. Members of the interleukin (IL) family play pivotal regulatory roles in the growth, differentiation, survival, and apoptosis of NK cells, and are central to their anti-tumor activity. These cytokines enhance the ability of NK cells to recognize and eliminate tumor cells by binding to specific receptors and activating downstream signaling pathways. Furthermore, interleukins do not function in isolation; the synergistic or antagonistic interactions between different interleukins can drive NK cells toward various functional pathways, ultimately leading to diverse outcomes for breast cancer patients. This paper reviews the intricate relationship between NK cells and interleukins, particularly within the breast cancer tumor microenvironment. Additionally, we summarize the latest clinical studies and advancements in NK cell therapy for breast cancer, along with the potential applications of interleukin signaling in these therapies. In conclusion, this article underscores the critical role of NK cells and interleukin signaling in breast cancer treatment, providing valuable insights and a significant reference for future research and clinical practice.
Collapse
Affiliation(s)
- Jiachi Xu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Hongyu Gao
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Muhammad Salman Azhar
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haifan Xu
- Breast and Thyroid Surgery, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Siyuan Chen
- Breast and Thyroid Surgery, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Mingcan Li
- Breast and Thyroid Surgery, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xinxi Ni
- Breast and Thyroid Surgery, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Ting Yan
- Breast and Thyroid Surgery, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Hui Zhou
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Long
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Wenjun Yi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| |
Collapse
|
43
|
Ochayon DE, DeVore SB, Chang WC, Krishnamurthy D, Seelamneni H, Grashel B, Spagna D, Andorf S, Martin LJ, Biagini JM, Waggoner SN, Khurana Hershey GK. Progressive accumulation of hyperinflammatory NKG2D low NK cells in early childhood severe atopic dermatitis. Sci Immunol 2024; 9:eadd3085. [PMID: 38335270 PMCID: PMC11107477 DOI: 10.1126/sciimmunol.add3085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 12/21/2023] [Indexed: 02/12/2024]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease that often precedes the development of food allergy, asthma, and allergic rhinitis. The prevailing paradigm holds that a reduced frequency and function of natural killer (NK) cell contributes to AD pathogenesis, yet the underlying mechanisms and contributions of NK cells to allergic comorbidities remain ill-defined. Here, analysis of circulating NK cells in a longitudinal early life cohort of children with AD revealed a progressive accumulation of NK cells with low expression of the activating receptor NKG2D, which was linked to more severe AD and sensitivity to allergens. This was most notable in children co-sensitized to food and aeroallergens, a risk factor for development of asthma. Individual-level longitudinal analysis in a subset of children revealed coincident reduction of NKG2D on NK cells with acquired or persistent sensitization, and this was associated with impaired skin barrier function assessed by transepidermal water loss. Low expression of NKG2D on NK cells was paradoxically associated with depressed cytolytic function but exaggerated release of the proinflammatory cytokine tumor necrosis factor-α. These observations provide important insights into a potential mechanism underlying the development of allergic comorbidity in early life in children with AD, which involves altered NK cell functional responses, and define an endotype of severe AD.
Collapse
Affiliation(s)
- David E. Ochayon
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center
| | - Stanley B. DeVore
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center
- Medical Scientist Training Program, University of Cincinnati College of Medicine
- Cancer and Cell Biology Program, University of Cincinnati College of Medicine
| | - Wan-Chi Chang
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center
| | - Durga Krishnamurthy
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center
| | - Harsha Seelamneni
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center
| | - Brittany Grashel
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center
| | - Daniel Spagna
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center
| | - Sandra Andorf
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center
- Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - Lisa J. Martin
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - Jocelyn M. Biagini
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - Stephen N. Waggoner
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center
- Medical Scientist Training Program, University of Cincinnati College of Medicine
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - Gurjit K. Khurana Hershey
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center
- Medical Scientist Training Program, University of Cincinnati College of Medicine
- Cancer and Cell Biology Program, University of Cincinnati College of Medicine
- Department of Pediatrics, University of Cincinnati College of Medicine
| |
Collapse
|
44
|
Panicker AJ, Prokop LJ, Hacke K, Jaramillo A, Griffiths LG. Outcome-based Risk Assessment of Non-HLA Antibodies in Heart Transplantation: A Systematic Review. J Heart Lung Transplant 2024; 43:1450-1467. [PMID: 38796046 DOI: 10.1016/j.healun.2024.05.012] [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: 11/06/2023] [Revised: 05/15/2024] [Accepted: 05/19/2024] [Indexed: 05/28/2024] Open
Abstract
BACKGROUND Current monitoring after heart transplantation (HT) employs repeated invasive endomyocardial biopsies (EMB). Although positive EMB confirms rejection, EMB fails to predict impending, subclinical, or EMB-negative rejection events. While non-human leukocyte antigen (non-HLA) antibodies have emerged as important risk factors for antibody-mediated rejection after HT, their use in clinical risk stratification has been limited. A systematic review of the role of non-HLA antibodies in rejection pathologies has the potential to guide efforts to overcome deficiencies of EMB in rejection monitoring. METHODS Databases were searched to include studies on non-HLA antibodies in HT recipients. Data collected included the number of patients, type of rejection, non-HLA antigen studied, association of non-HLA antibodies with rejection, and evidence for synergistic interaction between non-HLA antibodies and donor-specific anti-human leukocyte antigen antibody (HLA-DSA) responses. RESULTS A total of 56 studies met the inclusion criteria. Strength of evidence for each non-HLA antibody was evaluated based on the number of articles and patients in support versus against their role in mediating rejection. Importantly, despite previous intense focus on the role of anti-major histocompatibility complex class I chain-related gene A (MICA) and anti-angiotensin II type I receptor antibodies (AT1R) in HT rejection, evidence for their involvement was equivocal. Conversely, the strength of evidence for other non-HLA antibodies supports that differing rejection pathologies are driven by differing non-HLA antibodies. CONCLUSIONS This systematic review underscores the importance of identifying peri-HT non-HLA antibodies. Current evidence supports the role of non-HLA antibodies in all forms of HT rejection. Further investigations are required to define the mechanisms of action of non-HLA antibodies in HT rejection.
Collapse
Affiliation(s)
- Anjali J Panicker
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota; Department of Immunology, Mayo Clinic, Rochester, Minnesota; Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Larry J Prokop
- Mayo Clinic Libraries, Mayo Clinic, Rochester, Minnesota
| | - Katrin Hacke
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, Arizona
| | - Andrés Jaramillo
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, Arizona
| | - Leigh G Griffiths
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota; Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota; Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
45
|
Petersdorf EW. HLA structure and function in hematopoietic-cell transplantation. Best Pract Res Clin Haematol 2024; 37:101564. [PMID: 39396254 DOI: 10.1016/j.beha.2024.101564] [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: 12/15/2023] [Revised: 07/11/2024] [Accepted: 07/23/2024] [Indexed: 10/15/2024]
Abstract
The degree of HLA compatibility between a patient and donor has formed the basis of donor selection since the development of allogeneic hematopoietic cell transplantation over 50 years ago and has advanced understanding of the basic immunobiology of HLA. New evidence supports a role for germline variation in the patient and the donor that do not require HLA matching for their effects to have clinical consequences. The discovery of novel non-coding polymorphisms, structural features of HLA molecules, and expression provide new models for donor selection and inspire the development of tools for clinical translation. Pairwise effects of HLA ligand/donor NK receptors may play an important role in transplant outcomes and showcase the value of understanding the role played by each constituent of the NK pathway in modulating donor responses to target antigens.
Collapse
Affiliation(s)
- Effie W Petersdorf
- Division of Translational Science and Therapeutics, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, USA.
| |
Collapse
|
46
|
Jiang P, Jing S, Sheng G, Jia F. The basic biology of NK cells and its application in tumor immunotherapy. Front Immunol 2024; 15:1420205. [PMID: 39221244 PMCID: PMC11361984 DOI: 10.3389/fimmu.2024.1420205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Natural Killer (NK) cells play a crucial role as effector cells within the tumor immune microenvironment, capable of identifying and eliminating tumor cells through the expression of diverse activating and inhibitory receptors that recognize tumor-related ligands. Therefore, harnessing NK cells for therapeutic purposes represents a significant adjunct to T cell-based tumor immunotherapy strategies. Presently, NK cell-based tumor immunotherapy strategies encompass various approaches, including adoptive NK cell therapy, cytokine therapy, antibody-based NK cell therapy (enhancing ADCC mediated by NK cells, NK cell engagers, immune checkpoint blockade therapy) and the utilization of nanoparticles and small molecules to modulate NK cell anti-tumor functionality. This article presents a comprehensive overview of the latest advances in NK cell-based anti-tumor immunotherapy, with the aim of offering insights and methodologies for the clinical treatment of cancer patients.
Collapse
Affiliation(s)
- Pan Jiang
- Department of General Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Department of Infectious Diseases, Jingzhou First People’s Hospital, Jingzhou, China
| | - Shaoze Jing
- Department of Orthopedics, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Gaohong Sheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fajing Jia
- Department of General Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
47
|
Banerjee A, Farci P. Fibrosis and Hepatocarcinogenesis: Role of Gene-Environment Interactions in Liver Disease Progression. Int J Mol Sci 2024; 25:8641. [PMID: 39201329 PMCID: PMC11354981 DOI: 10.3390/ijms25168641] [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: 06/26/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
The liver is a complex organ that performs vital functions in the body. Despite its extraordinary regenerative capacity compared to other organs, exposure to chemical, infectious, metabolic and immunologic insults and toxins renders the liver vulnerable to inflammation, degeneration and fibrosis. Abnormal wound healing response mediated by aberrant signaling pathways causes chronic activation of hepatic stellate cells (HSCs) and excessive accumulation of extracellular matrix (ECM), leading to hepatic fibrosis and cirrhosis. Fibrosis plays a key role in liver carcinogenesis. Once thought to be irreversible, recent clinical studies show that hepatic fibrosis can be reversed, even in the advanced stage. Experimental evidence shows that removal of the insult or injury can inactivate HSCs and reduce the inflammatory response, eventually leading to activation of fibrolysis and degradation of ECM. Thus, it is critical to understand the role of gene-environment interactions in the context of liver fibrosis progression and regression in order to identify specific therapeutic targets for optimized treatment to induce fibrosis regression, prevent HCC development and, ultimately, improve the clinical outcome.
Collapse
Affiliation(s)
- Anindita Banerjee
- Department of Transfusion Transmitted Diseases, ICMR-National Institute of Immunohaematology, Mumbai 400012, Maharashtra, India;
| | - Patrizia Farci
- Hepatic Pathogenesis Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
48
|
Herault A, Mak J, de la Cruz-Chuh J, Dillon MA, Ellerman D, Go M, Cosino E, Clark R, Carson E, Yeung S, Pichery M, Gador M, Chiang EY, Wu J, Liang Y, Modrusan Z, Gampa G, Sudhamsu J, Kemball CC, Cheung V, Nguyen TTT, Seshasayee D, Piskol R, Totpal K, Yu SF, Lee G, Kozak KR, Spiess C, Walsh KB. NKG2D-bispecific enhances NK and CD8+ T cell antitumor immunity. Cancer Immunol Immunother 2024; 73:209. [PMID: 39112670 PMCID: PMC11306676 DOI: 10.1007/s00262-024-03795-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 07/30/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Cancer immunotherapy approaches that elicit immune cell responses, including T and NK cells, have revolutionized the field of oncology. However, immunosuppressive mechanisms restrain immune cell activation within solid tumors so additional strategies to augment activity are required. METHODS We identified the co-stimulatory receptor NKG2D as a target based on its expression on a large proportion of CD8+ tumor infiltrating lymphocytes (TILs) from breast cancer patient samples. Human and murine surrogate NKG2D co-stimulatory receptor-bispecifics (CRB) that bind NKG2D on NK and CD8+ T cells as well as HER2 on breast cancer cells (HER2-CRB) were developed as a proof of concept for targeting this signaling axis in vitro and in vivo. RESULTS HER2-CRB enhanced NK cell activation and cytokine production when co-cultured with HER2 expressing breast cancer cell lines. HER2-CRB when combined with a T cell-dependent-bispecific (TDB) antibody that synthetically activates T cells by crosslinking CD3 to HER2 (HER2-TDB), enhanced T cell cytotoxicity, cytokine production and in vivo antitumor activity. A mouse surrogate HER2-CRB (mHER2-CRB) improved in vivo efficacy of HER2-TDB and augmented NK as well as T cell activation, cytokine production and effector CD8+ T cell differentiation. CONCLUSION We demonstrate that targeting NKG2D with bispecific antibodies (BsAbs) is an effective approach to augment NK and CD8+ T cell antitumor immune responses. Given the large number of ongoing clinical trials leveraging NK and T cells for cancer immunotherapy, NKG2D-bispecifics have broad combinatorial potential.
Collapse
Affiliation(s)
- Aurelie Herault
- Department of Molecular Oncology, Genentech, South San Francisco, CA, USA
| | - Judy Mak
- Department of Molecular Oncology, Genentech, South San Francisco, CA, USA
| | - Josefa de la Cruz-Chuh
- Department of Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, USA
| | - Michael A Dillon
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - Diego Ellerman
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - MaryAnn Go
- Department of In Vivo Pharmacology, Genentech, South San Francisco, CA, USA
| | - Ely Cosino
- Department of In Vivo Pharmacology, Genentech, South San Francisco, CA, USA
| | - Robyn Clark
- Department of In Vivo Pharmacology, Genentech, South San Francisco, CA, USA
| | - Emily Carson
- Department of In Vivo Pharmacology, Genentech, South San Francisco, CA, USA
| | - Stacey Yeung
- Department of Molecular Oncology, Genentech, South San Francisco, CA, USA
| | - Melanie Pichery
- Immuno-Oncology-In Vitro Biology Department, Evotec, Toulouse, France
| | - Mylène Gador
- Immuno-Oncology-In Vitro Biology Department, Evotec, Toulouse, France
| | - Eugene Y Chiang
- Department of Cancer Immunology, Genentech, South San Francisco, CA, USA
| | - Jia Wu
- Department of Antibody Discovery, Genentech, South San Francisco, CA, USA
| | - Yuxin Liang
- Department of Next-GenSequencing, South San Francisco, CA, USA
| | - Zora Modrusan
- Department of Next-GenSequencing, South San Francisco, CA, USA
| | - Gautham Gampa
- Department of Development Sciences PTPK, Genentech, South San Francisco, CA, USA
| | - Jawahar Sudhamsu
- Department of Structural Biology, Genentech, South San Francisco, CA, USA
| | - Christopher C Kemball
- Department of Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, USA
| | - Victoria Cheung
- Department of Molecular Oncology, Genentech, South San Francisco, CA, USA
| | | | - Dhaya Seshasayee
- Department of Antibody Discovery, Genentech, South San Francisco, CA, USA
| | - Robert Piskol
- Department of Bioinformatics, Genentech, South San Francisco, CA, USA
| | - Klara Totpal
- Department of In Vivo Pharmacology, Genentech, South San Francisco, CA, USA
| | - Shang-Fan Yu
- Department of In Vivo Pharmacology, Genentech, South San Francisco, CA, USA
| | - Genee Lee
- Department of Molecular Oncology, Genentech, South San Francisco, CA, USA
| | - Katherine R Kozak
- Department of Biochemical and Cellular Pharmacology, Genentech, South San Francisco, CA, USA
| | - Christoph Spiess
- Department of Antibody Engineering, Genentech, South San Francisco, CA, USA
| | - Kevin B Walsh
- Department of Molecular Oncology, Genentech, South San Francisco, CA, USA.
| |
Collapse
|
49
|
Lanier LL. Five decades of natural killer cell discovery. J Exp Med 2024; 221:e20231222. [PMID: 38842526 PMCID: PMC11157086 DOI: 10.1084/jem.20231222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/12/2024] [Accepted: 04/17/2024] [Indexed: 06/07/2024] Open
Abstract
The first descriptions of "non-specific" killing of tumor cells by lymphocytes were reported in 1973, and subsequently, the mediators of the activity were named "natural killer" (NK) cells by Rolf Kiessling and colleagues at the Karolinska Institute in 1975. The activity was detected in mice, rats, and humans that had no prior exposure to the tumors, major histocompatibility complex (MHC) antigen matching of the effectors and tumor cells was not required, and the cells responsible were distinct from MHC-restricted, antigen-specific T cells. In the ensuing five decades, research by many labs has extended knowledge of NK cells beyond an in vitro curiosity to demonstrate their in vivo relevance in host defense against tumors and microbial pathogens and their role in regulation of the immune system. This brief Perspective highlights a timeline of a few selected advancements in NK cell biology from a personal perspective of being involved in this quest.
Collapse
Affiliation(s)
- Lewis L. Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
50
|
Fiala GJ, Lücke J, Huber S. Pro- and antitumorigenic functions of γδ T cells. Eur J Immunol 2024; 54:e2451070. [PMID: 38803018 DOI: 10.1002/eji.202451070] [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: 04/03/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024]
Abstract
γδ T cells are a subset of T cells that are characterized by the expression of a TCR-γδ instead of a TCR-αβ. Despite being outnumbered by their αβ T cell counterpart in many tissues, studies from the last 20 years underline their important and non-redundant roles in tumor and metastasis development. However, whether a γδ T cell exerts pro- or antitumorigenic effects seems to depend on a variety of factors, many of them still incompletely understood today. In this review, we summarize mechanisms by which γδ T cells exert these seemingly contradictory effector functions in mice and humans. Furthermore, we discuss the current view on inducing and inhibiting factors of γδ T cells during cancer development.
Collapse
Affiliation(s)
- Gina J Fiala
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Biological Signalling Studies BIOSS, University of Freiburg, Freiburg, Germany
| | - Jöran Lücke
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Huber
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|