1
|
Nie Z, Chang J, Yang Z, Zeng K, Liu Y, Tu Q, Wang C, Yan Q, Shi H, Guo Y. PRMT5 highly expressed on CD16 + CD56- natural killer cells is correlated with NK cells exhaustion in colorectal cancer mesenchyme. Cancer Immunol Immunother 2025; 74:139. [PMID: 40056169 PMCID: PMC11890482 DOI: 10.1007/s00262-025-03981-w] [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: 12/25/2024] [Accepted: 02/11/2025] [Indexed: 03/10/2025]
Abstract
OBJECTIVE To investigate the relationships between changes in the phenotype of natural killer cells (NK cells) in the microenvironment of colorectal cancer (CRC) and the expression of important immune checkpoints. To assess the expression level of CD16 bright CD56 negative (CD16 + CD56-) NK cell-associated immune checkpoints, including protein arginine methyltransferase 5 (PRMT5) and T-cell immunoreceptor with Ig and ITIM domains (TIGIT), single-immunoglobulin interleukin-1-related receptor (SIGIRR), in CRC mesenchyme. METHODS A total of 194 patients who were diagnosed with CRC were screened. The percentage of NK cells and the expression levels of their surface receptors, including PRMT5, CD56, CD69, TIGIT, CD16, IFN-γ, and SIGIRR, in the tumor microenvironment (TME) of CRC were assessed. Immunohistochemical staining, multiplex immunohistochemistry, and single-cell sequencing were performed. RESULTS Compared with normal mesenchyme, NK cells were less in CRC mesenchyme. The percentage of CD16 + CD56- NK cells in tumor mesenchyme was significantly higher, the number of CD16 + NK cells was more, and the number of CD56 + NK cells was less in CRC mesenchyme. High expression of TIGIT and PRMT5 expression affected the progression of CRC. The expression of PRMT5 and SIGIRR expression was significantly increased in CD16 + CD56- NK cells, and both genes were identified as important morbidity factors. PRMT5 and SIGIRR may contribute to the phenotype changes of NK cells in CRC. CONCLUSION The microenvironment of CRC is in an immunosuppressive state characterized mainly by high expression of TIGIT, CD16, PRMT5, and SIGIRR; low expression of CD56, IFN-γ, and CD69; significantly decreased percentage of CD56 + NK cells; and significantly increased percentage of CD16 + CD56- NK cells with weakened killing ability. PRMT5 and TIGIT may be closely related to the formation of CD16 + CD56- NK cells with weakened killing ability.
Collapse
Affiliation(s)
- Zunzhen Nie
- Xi'an Daxing Hospital Affiliated to Yan'an University, Xi'an, People's Republic of China
| | - Juanjuan Chang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, People's Republic of China
| | - Zhiqin Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, People's Republic of China
| | - Kaixuan Zeng
- Precision Medical Research Institute, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yuangang Liu
- Xi'an Daxing Hospital Affiliated to Yan'an University, Xi'an, People's Republic of China
| | - Qian Tu
- Xi'an Daxing Hospital Affiliated to Yan'an University, Xi'an, People's Republic of China
| | - Chao Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, People's Republic of China
| | - Qingguo Yan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, People's Republic of China
| | - Hai Shi
- Department of Surgery, Xi'an Daxing Hospital, Xi'an, People's Republic of China
| | - Ying Guo
- Xi'an Daxing Hospital Affiliated to Yan'an University, Xi'an, People's Republic of China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, People's Republic of China.
| |
Collapse
|
2
|
Mantovani A, Marchesi F, Di Mitri D, Garlanda C. Macrophage diversity in cancer dissemination and metastasis. Cell Mol Immunol 2024; 21:1201-1214. [PMID: 39402303 PMCID: PMC11528009 DOI: 10.1038/s41423-024-01216-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/06/2024] [Indexed: 11/02/2024] Open
Abstract
Invasion and metastasis are hallmarks of cancer. In addition to the well-recognized hematogenous and lymphatic pathways of metastasis, cancer cell dissemination can occur via the transcoelomic and perineural routes, which are typical of ovarian and pancreatic cancer, respectively. Macrophages are a universal major component of the tumor microenvironment and, in established tumors, promote growth and dissemination to secondary sites. Here, we review the role of tumor-associated macrophages (TAMs) in cancer cell dissemination and metastasis, emphasizing the diversity of myeloid cells in different tissue contexts (lungs, liver, brain, bone, peritoneal cavity, nerves). The generally used models of lung metastasis fail to capture the diversity of pathways and tissue microenvironments. A better understanding of TAM diversity in different tissue contexts may pave the way for tailored diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Alberto Mantovani
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy.
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy.
- William Harvey Research Institute, Queen Mary University, London, UK.
| | - Federica Marchesi
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
- Department Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Diletta Di Mitri
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy
| | - Cecilia Garlanda
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy
| |
Collapse
|
3
|
Ma C, Hao Y, Shi B, Wu Z, Jin D, Yu X, Jin B. Unveiling mitochondrial and ribosomal gene deregulation and tumor microenvironment dynamics in acute myeloid leukemia. Cancer Gene Ther 2024; 31:1034-1048. [PMID: 38806621 DOI: 10.1038/s41417-024-00788-2] [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: 01/10/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
Acute myeloid leukemia (AML) is a malignant clonal hematopoietic disease with a poor prognosis. Understanding the interaction between leukemic cells and the tumor microenvironment (TME) can help predict the prognosis of leukemia and guide its treatment. Re-analyzing the scRNA-seq data from the CSC and G20 cohorts, using a Python-based pipeline including machine-learning-based scVI-tools, recapitulated the distinct hierarchical structure within the samples of AML patients. Weighted correlation network analysis (WGCNA) was conducted to construct a weighted gene co-expression network and to identify gene modules primarily focusing on hematopoietic stem cells (HSCs), multipotent progenitors (MPPs), and natural killer (NK) cells. The analysis revealed significant deregulation in gene modules associated with aerobic respiration and ribosomal/cytoplasmic translation. Cell-cell communications were elucidated by the CellChat package, revealing an imbalance of activating and inhibitory immune signaling pathways. Interception of genes upregulated in leukemic HSCs & MPPs as well as in NKG2A-high NK cells was used to construct prognostic models. Normal Cox and artificial neural network models based on 10 genes were developed. The study reveals the deregulation of mitochondrial and ribosomal genes in AML patients and suggests the co-occurrence of stimulatory and inhibitory factors in the AML TME.
Collapse
Affiliation(s)
- Chao Ma
- Institute of Cancer Stem Cell, Dalian Medical University, West Section Lvshun South Road, Dalian, 116044, Liaoning, China
| | - Yuchao Hao
- Department of Hematology, The Second Hospital of Dalian Medical University, West Section Lvshun South Road, Dalian, 116027, Liaoning, China
| | - Bo Shi
- Institute of Cancer Stem Cell, Dalian Medical University, West Section Lvshun South Road, Dalian, 116044, Liaoning, China
| | - Zheng Wu
- Institute of Cancer Stem Cell, Dalian Medical University, West Section Lvshun South Road, Dalian, 116044, Liaoning, China
| | - Di Jin
- Institute of Cancer Stem Cell, Dalian Medical University, West Section Lvshun South Road, Dalian, 116044, Liaoning, China
| | - Xiao Yu
- NHC Key Laboratory of Pneumoconiosis, The First Hospital of Shanxi Medical University, South Jiefang Road, Taiyuan, 030001, Shanxi, China.
| | - Bilian Jin
- Institute of Cancer Stem Cell, Dalian Medical University, West Section Lvshun South Road, Dalian, 116044, Liaoning, China.
| |
Collapse
|
4
|
Singhal S, Rao AS, Stadanlick J, Bruns K, Sullivan NT, Bermudez A, Honig-Frand A, Krouse R, Arambepola S, Guo E, Moon EK, Georgiou G, Valerius T, Albelda SM, Eruslanov EB. Human Tumor-Associated Macrophages and Neutrophils Regulate Antitumor Antibody Efficacy through Lethal and Sublethal Trogocytosis. Cancer Res 2024; 84:1029-1047. [PMID: 38270915 PMCID: PMC10982649 DOI: 10.1158/0008-5472.can-23-2135] [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: 07/17/2023] [Revised: 11/29/2023] [Accepted: 01/23/2024] [Indexed: 01/26/2024]
Abstract
The clinical benefits of tumor-targeting antibodies (tAb) are modest in solid human tumors. The efficacy of many tAbs is dependent on Fc receptor (FcR)-expressing leukocytes that bind Fc fragments of tAb. Tumor-associated macrophages (TAM) and neutrophils (TAN) represent the majority of FcR+ effectors in solid tumors. A better understanding of the mechanisms by which TAMs and TANs regulate tAb response could help improve the efficacy of cancer treatments. Here, we found that myeloid effectors interacting with tAb-opsonized lung cancer cells used antibody-dependent trogocytosis (ADT) but not antibody-dependent phagocytosis. During this process, myeloid cells "nibbled off" tumor cell fragments containing tAb/targeted antigen (tAg) complexes. ADT was only tumoricidal when the tumor cells expressed high levels of tAg and the effectors were present at high effector-to-tumor ratios. If either of these conditions were not met, which is typical for solid tumors, ADT was sublethal. Sublethal ADT, mainly mediated by CD32hiCD64hi TAM, led to two outcomes: (i) removal of surface tAg/tAb complexes from the tumor that facilitated tumor cell escape from the tumoricidal effects of tAb; and (ii) acquisition of bystander tAgs by TAM with subsequent cross-presentation and stimulation of tumor-specific T-cell responses. CD89hiCD32loCD64lo peripheral blood neutrophils (PBN) and TAN stimulated tumor cell growth in the presence of the IgG1 anti-EGFR Ab cetuximab; however, IgA anti-EGFR Abs triggered the tumoricidal activity of PBN and negated the stimulatory effect of TAN. Overall, this study provides insights into the mechanisms by which myeloid effectors mediate tumor cell killing or resistance during tAb therapy. SIGNIFICANCE The elucidation of the conditions and mechanisms by which human FcR+ myeloid effectors mediate cancer cell resistance and killing during antibody treatment could help develop improved strategies for treating solid tumors.
Collapse
Affiliation(s)
- Sunil Singhal
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Abhishek S. Rao
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jason Stadanlick
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kyle Bruns
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Neil T. Sullivan
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andres Bermudez
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Adam Honig-Frand
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ryan Krouse
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sachinthani Arambepola
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Emily Guo
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Edmund K. Moon
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - George Georgiou
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas
| | - Thomas Valerius
- Department of Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Steven M. Albelda
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Evgeniy B. Eruslanov
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
5
|
Di Ceglie I, Carnevale S, Rigatelli A, Grieco G, Molisso P, Jaillon S. Immune cell networking in solid tumors: focus on macrophages and neutrophils. Front Immunol 2024; 15:1341390. [PMID: 38426089 PMCID: PMC10903099 DOI: 10.3389/fimmu.2024.1341390] [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: 11/20/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
The tumor microenvironment is composed of tumor cells, stromal cells and leukocytes, including innate and adaptive immune cells, and represents an ecological niche that regulates tumor development and progression. In general, inflammatory cells are considered to contribute to tumor progression through various mechanisms, including the formation of an immunosuppressive microenvironment. Macrophages and neutrophils are important components of the tumor microenvironment and can act as a double-edged sword, promoting or inhibiting the development of the tumor. Targeting of the immune system is emerging as an important therapeutic strategy for cancer patients. However, the efficacy of the various immunotherapies available is still limited. Given the crucial importance of the crosstalk between macrophages and neutrophils and other immune cells in the formation of the anti-tumor immune response, targeting these interactions may represent a promising therapeutic approach against cancer. Here we will review the current knowledge of the role played by macrophages and neutrophils in cancer, focusing on their interaction with other immune cells.
Collapse
Affiliation(s)
| | | | | | - Giovanna Grieco
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Piera Molisso
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Sebastien Jaillon
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
6
|
Yang L, Zhuang L, Ye Z, Li L, Guan J, Gong W. Immunotherapy and biomarkers in patients with lung cancer with tuberculosis: Recent advances and future Directions. iScience 2023; 26:107881. [PMID: 37841590 PMCID: PMC10570004 DOI: 10.1016/j.isci.2023.107881] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023] Open
Abstract
Lung cancer (LC) and tuberculosis (TB) are two major global public health problems, and the incidence of LC-TB is currently on the rise. Therefore effective clinical interventions are crucial for LC-TB. The aim of this review is to provide up-to-date information on the immunological profile and therapeutic biomarkers in patients with LC-TB. We discuss the immune mechanisms involved, including the immune checkpoints that play an important role in the treatment of patients with LC-TB. In addition, we explore the susceptibility of patients with LC to TB and summarise the latest research on LC-TB. Finally, we discuss future prospects in this field, including the identification of potential targets for immune intervention. In conclusion, this review provides important insights into the complex relationship between LC and TB and highlights new advances in the detection and treatment of both diseases.
Collapse
Affiliation(s)
- Ling Yang
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, Eighth Medical Center of PLA General Hospital, Beijing 100091, China
- Hebei North University, Zhangjiakou, Hebei 075000, China
- Senior Department of Oncology, Fifth Medical Center of PLA General Hospital, Beijing 100071, China
| | - Li Zhuang
- Hebei North University, Zhangjiakou, Hebei 075000, China
| | - Zhaoyang Ye
- Hebei North University, Zhangjiakou, Hebei 075000, China
| | - Linsheng Li
- Hebei North University, Zhangjiakou, Hebei 075000, China
| | - Jingzhi Guan
- Senior Department of Oncology, Fifth Medical Center of PLA General Hospital, Beijing 100071, China
| | - Wenping Gong
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, Eighth Medical Center of PLA General Hospital, Beijing 100091, China
| |
Collapse
|
7
|
Lu J, Li J, Lin Z, Li H, Lou L, Ding W, Ouyang S, Yonghui W, Wen Y, Chen X, Yue P, Wang Y, Liu P, Lu J, Zhang J, Feng W, Zhang X. Reprogramming of TAMs via the STAT3/CD47-SIRPα axis promotes acquired resistance to EGFR-TKIs in lung cancer. Cancer Lett 2023; 564:216205. [PMID: 37146936 DOI: 10.1016/j.canlet.2023.216205] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/23/2023] [Accepted: 04/22/2023] [Indexed: 05/07/2023]
Abstract
Cross-talk between the tumor microenvironment (TME) and cancer cells plays an important role in acquired drug resistance to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs). The role of tumor-associated macrophages (TAMs), the major component of the TME, in acquired resistance remains unclear. In this study, M2-like reprogramming of TAMs and reduced phagocytosis by macrophages were observed in gefitinib-resistant lung cancer cells and tumor xenografts. CD47 was upregulated in TKI-resistant lung cancer cells, and M2 macrophage polarization and cancer cell escape from macrophage phagocytosis were enhanced. Culture medium from TKI-resistant cells led to metabolic reprogramming of TAMs. STAT3 was associated with CD47 expression in TKI-resistant lung cancer cells. Genetic and pharmacological inhibition of STAT3 enhanced the phagocytic activity of TAMs and alleviated the acquired resistance to EGFR-TKIs via inhibiting the CD47-SIRPα signaling axis and M2 polarization in the co-culture system. Moreover, STAT3 transcriptionally regulated CD47 expression by binding to consensus DNA response elements in the intron of the CD47 gene. Furthermore, the combination of gefitinib with a STAT3 inhibitor and an anti-CD47 monoclonal antibody alleviated the acquired resistance to gefitinib in vitro and in vivo. Collectively, our study reveals the role of TAM reprogramming and the CD47-SIRPα axis in acquired EGFR-TKI resistance and provides a novel therapeutic strategy to overcome the acquired resistance to EGFR-TKIs in lung cancer.
Collapse
Affiliation(s)
- Jiaye Lu
- National-Local Joint Engineering Laboratory of Draggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Jingwei Li
- National-Local Joint Engineering Laboratory of Draggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Ziyou Lin
- National-Local Joint Engineering Laboratory of Draggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Huaxuan Li
- National-Local Joint Engineering Laboratory of Draggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Linlin Lou
- National-Local Joint Engineering Laboratory of Draggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Wen Ding
- National-Local Joint Engineering Laboratory of Draggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Shumin Ouyang
- National-Local Joint Engineering Laboratory of Draggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Wu Yonghui
- Department of Thoracic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yuanzhen Wen
- Increasepharm (Hengqin) Innovative Medicine Institute Co. Ltd, Zhuhai, 519000, China
| | - Xiaobing Chen
- Increasepharm (Hengqin) Innovative Medicine Institute Co. Ltd, Zhuhai, 519000, China
| | - Peibin Yue
- Department of Medicine, Division of Hematology-Oncology, and Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Yuanxiang Wang
- National-Local Joint Engineering Laboratory of Draggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Peiqing Liu
- National-Local Joint Engineering Laboratory of Draggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Jinjian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, 519000, Macau.
| | - Jian Zhang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Weineng Feng
- Department of Head and Neck/Thoracic Medical Oncology, The First People's Hospital of Foshan, Foshan, 52800, China.
| | - Xiaolei Zhang
- National-Local Joint Engineering Laboratory of Draggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.
| |
Collapse
|
8
|
Wang L, Chen Z, Liu G, Pan Y. Functional crosstalk and regulation of natural killer cells in tumor microenvironment: Significance and potential therapeutic strategies. Genes Dis 2023; 10:990-1004. [PMID: 37396514 PMCID: PMC10308134 DOI: 10.1016/j.gendis.2022.07.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/11/2022] [Accepted: 07/15/2022] [Indexed: 11/19/2022] Open
Abstract
Natural killer (NK) cells eliminate a large variety of tumor cells and abnormal cells. However, NK cells in the tumor microenvironment (TME) are often functionally depleted. A few subsets of NK cells even promote tumor growth. This study reviewed the biological properties of NK cells, the dynamic phenotypic changes of NK cells in the TME, and the communication between NK cells and other immune and nonimmune cells.
Collapse
Affiliation(s)
- Liping Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China
| | - Zhe Chen
- Department of Otolaryngology Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China
| | - Guohong Liu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China
| | - Yunbao Pan
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China
| |
Collapse
|
9
|
Improved Antitumor Effect of NK Cells Activated by Neutrophils in a Bone Marrow Transplant Model. Mediators Inflamm 2023; 2023:6316581. [PMID: 36762286 PMCID: PMC9904906 DOI: 10.1155/2023/6316581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 12/23/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
The licensing process mediated by inhibitory receptors of the Ly49 C-type lectin superfamily that recognizes self-major histocompatibility complex (MHC) class I in mice is essential for the proper antitumor function of natural killer (NK) cells. Several models for NK cell licensing can be exploited for adoptive immunotherapy for cancer. However, the appropriate adoptive transfer setting to induce efficient graft versus tumor/leukemia effects remains elusive, especially after hematopoietic stem cell transplantation (HSCT). In our previous experiment, we showed that intraperitoneal neutrophil administration with their corresponding NK receptor ligand-activated NK cells using congenic mice without HSCT. In this experiment, we demonstrate enhanced antitumor effects of licensed NK cells induced by weekly intraperitoneal injections of irradiated neutrophil-enriched peripheral blood mononuclear cells (PBMNCs) in recipient mice bearing lymphoma. Bone marrow transplantation was performed using BALB/c mice (H-2d) as the recipient and B10 mice (H-2b) as the donor. The tumor was A20, a BALB/c-derived lymphoma cell line, which was injected subcutaneously into the recipient at the same time as the HSCT. Acute graft versus host disease was not exacerbated in this murine MHC class I mismatched HSCT setting. The intraperitoneal injection of PBMNCs activated a transient licensing of NK subsets expressed Ly49G2, its corresponding NK receptor ligand to H-2d, and reduced A20 tumor growth in the recipient after HSCT. Pathological examination revealed that increased donor-oriented NK1.1+NK cells migrated into the recipient tumors, depending on neutrophil counts in the administered PBMNCs. Collectively, our data reveal a pivotal role of neutrophils in promoting NK cell effector functions and adoptive immunotherapy for cancer.
Collapse
|
10
|
Azumi J, Takeda T, Shimada Y, Zhuang T, Tokuji Y, Sakamoto N, Aso H, Nakamura T. Organogermanium THGP Induces Differentiation into M1 Macrophages and Suppresses the Proliferation of Melanoma Cells via Phagocytosis. Int J Mol Sci 2023; 24:ijms24031885. [PMID: 36768216 PMCID: PMC9915250 DOI: 10.3390/ijms24031885] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 01/21/2023] Open
Abstract
M1 macrophages are an important cell type related to tumor immunology and are known to phagocytose cancer cells. In previous studies, the organogermanium compound poly-trans-[(2-carboxyethyl)germasesquioxane] (Ge-132) and its hydrolysate, 3-(trihydroxygermyl) propanoic acid (THGP), have been reported to exert antitumor effects by activating NK cells and macrophages through the induction of IFN-γ activity in vivo. However, the detailed molecular mechanism has not been clarified. In this study, we found that macrophages differentiate into the M1 phenotype via NF-κB activation under long-term culture in the presence of THGP in vitro and in vivo. Furthermore, long-term culture with THGP increases the ability of RAW 264.7 cells to suppress B16 4A5 melanoma cell proliferation. These mechanisms indicate that THGP promotes the M1 polarization of macrophages and suppresses the expression of signal-regulatory protein alpha (SIRP-α) in macrophages and CD47 in cancers. Based on these results, THGP may be considered a new regulatory reagent that suppresses tumor immunity.
Collapse
Affiliation(s)
- Junya Azumi
- Research Division, Asai Germanium Research Institute Co., Ltd., Suzuranoka 3-131, Hakodate 042-0958, Japan
| | - Tomoya Takeda
- Research Division, Asai Germanium Research Institute Co., Ltd., Suzuranoka 3-131, Hakodate 042-0958, Japan
| | - Yasuhiro Shimada
- Research Division, Asai Germanium Research Institute Co., Ltd., Suzuranoka 3-131, Hakodate 042-0958, Japan
| | - Tao Zhuang
- Laboratory of Animal Health Science, Graduate School of Agricultural Science, Tohoku University, 468-1 Aoba, Aramaki, Aoba-ku, Sendai 980-8572, Japan
| | - Yoshihiko Tokuji
- Department of Human Sciences, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2 Sen, Inada, Obihiro 080-8555, Japan
| | - Naoya Sakamoto
- Isotope Imaging Laboratory, Creative Research Institution, Hokkaido University, Kita 10 Jo-Nishi 5, Kita, Sapporo 060-0810, Japan
| | - Hisashi Aso
- Laboratory of Animal Health Science, Graduate School of Agricultural Science, Tohoku University, 468-1 Aoba, Aramaki, Aoba-ku, Sendai 980-8572, Japan
| | - Takashi Nakamura
- Research Division, Asai Germanium Research Institute Co., Ltd., Suzuranoka 3-131, Hakodate 042-0958, Japan
- Correspondence: ; Tel.: +81-138-32-0032; Fax: +81-138-31-0132
| |
Collapse
|
11
|
Fang Q, Stehr AM, Naschberger E, Knopf J, Herrmann M, Stürzl M. No NETs no TIME: Crosstalk between neutrophil extracellular traps and the tumor immune microenvironment. Front Immunol 2022; 13:1075260. [PMID: 36618417 PMCID: PMC9816414 DOI: 10.3389/fimmu.2022.1075260] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
The tumor immune microenvironment (TIME) controls tumorigenesis. Neutrophils are important components of TIME and control tumor progression and therapy resistance. Neutrophil extracellular traps (NETs) ejected by activated neutrophils are net-like structures composed of decondensed extracellular chromatin filaments decorated with a plethora of granules as well as cytoplasmic proteins. Many of these harbour post translational modifications. Cancer cells reportedly trigger NET formation, and conversely, NETs alter the TIME and promote tumor cell proliferation and migration. The specific interactions between NETs and TIME and the respective effects on tumor progression are still elusive. In certain tumors, a CD4+ T helper (Th) 2 cell-associated TIME induces NETs and exerts immunosuppressive functions via programmed death 1 (PD-1)/PD-L1, both associated with poorer prognosis. In other cases, NETs induce the proliferation of Th1 cells, associated with an improved prognosis in cancer. In addition, NETs can drive macrophage polarization and often rely on macrophages to promote cancer cell invasion and metastasis. In turn, macrophages can swiftly clear NETs in an immunologically silent manner. The aim of this review is to summarize the knowledge about the mutual interaction between NETs and TIME and its impact on tumor growth and therapy.
Collapse
Affiliation(s)
- Qi Fang
- Division of Molecular and Experimental Surgery, Translational Research Center, Department of Surgery, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Antonia Margarethe Stehr
- Division of Molecular and Experimental Surgery, Translational Research Center, Department of Surgery, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Elisabeth Naschberger
- Division of Molecular and Experimental Surgery, Translational Research Center, Department of Surgery, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jasmin Knopf
- Department of Internal Medicine 3, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Martin Herrmann
- Department of Internal Medicine 3, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Michael Stürzl
- Division of Molecular and Experimental Surgery, Translational Research Center, Department of Surgery, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
12
|
Exploring the Utility of NK Cells in COVID-19. Biomedicines 2022; 10:biomedicines10051002. [PMID: 35625739 PMCID: PMC9138257 DOI: 10.3390/biomedicines10051002] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 02/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) can manifest as acute respiratory distress syndrome and is associated with substantial morbidity and mortality. Extensive data now indicate that immune responses to SARS-CoV-2 infection determine the COVID-19 disease course. A wide range of immunomodulatory agents have been tested for the treatment of COVID-19. Natural killer (NK) cells play an important role in antiviral innate immunity, and anti-SARS-CoV-2 activity and antifibrotic activity are particularly critical for COVID-19 control. Notably, SARS-CoV-2 clearance rate, antibody response, and disease progression in COVID-19 correlate with NK cell status, and NK cell dysfunction is linked with increased SARS-CoV-2 susceptibility. Thus, NK cells function as the key element in the switch from effective to harmful immune responses in COVID-19. However, dysregulation of NK cells has been observed in COVID-19 patients, exhibiting depletion and dysfunction, which correlate with COVID-19 severity; this dysregulation perhaps contributes to disease progression. Given these findings, NK-cell-based therapies with anti-SARS-CoV-2 activity, antifibrotic activity, and strong safety profiles for cancers may encourage the rapid application of functional NK cells as a potential therapeutic strategy to eliminate SARS-CoV-2-infected cells at an early stage, facilitate immune–immune cell interactions, and favor inflammatory processes that prevent and/or reverse over-inflammation and inhibit fibrosis progression, thereby helping in the fight against COVID-19. However, our understanding of the role of NK cells in COVID-19 remains incomplete, and further research on the involvement of NK cells in the pathogenesis of COVID-19 is needed. The rationale of NK-cell-based therapies for COVID-19 has to be based on the timing of therapeutic interventions and disease severity, which may be determined by the balance between beneficial antiviral and potential detrimental pathologic actions. NK cells would be more effective early in SARS-CoV-2 infection and prevent the progression of COVID-19. Immunomodulation by NK cells towards regulatory functions could be useful as an adjunct therapy to prevent the progression of COVID-19.
Collapse
|
13
|
Najem H, Ott M, Kassab C, Rao A, Rao G, Marisetty A, Sonabend AM, Horbinski C, Verhaak R, Shankar A, Krishnan SN, Varn FS, Arrieta VA, Gupta P, Ferguson SD, Huse JT, Fuller GN, Long JP, Winkowski DE, Freiberg BA, James CD, Platanias LC, Lesniak MS, Burks JK, Heimberger AB. Central nervous system immune interactome is function of cancer lineage, tumor microenvironment and STAT3 expression. JCI Insight 2022; 7:157612. [PMID: 35316217 PMCID: PMC9090258 DOI: 10.1172/jci.insight.157612] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/18/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Immune cell profiling of primary and metastatic CNS tumors has been focused on the tumor, not the tumor microenvironment (TME), or has been analyzed via biopsies. METHODS En bloc resections of gliomas (n = 10) and lung metastases (n = 10) were analyzed via tissue segmentation and high-dimension Opal 7-color multiplex imaging. Single-cell RNA analyses were used to infer immune cell functionality. RESULTS Within gliomas, T cells were localized in the infiltrating edge and perivascular space of tumors, while residing mostly in the stroma of metastatic tumors. CD163+ macrophages were evident throughout the TME of metastatic tumors, whereas in gliomas, CD68+, CD11c+CD68+, and CD11c+CD68+CD163+ cell subtypes were commonly observed. In lung metastases, T cells interacted with CD163+ macrophages as dyads and clusters at the brain-tumor interface and within the tumor itself and as clusters within the necrotic core. In contrast, gliomas typically lacked dyad and cluster interactions, except for T cell CD68+ cell dyads within the tumor. Analysis of transcriptomic data in glioblastomas revealed that innate immune cells expressed both proinflammatory and immunosuppressive gene signatures. CONCLUSION Our results show that immunosuppressive macrophages are abundant within the TME and that the immune cell interactome between cancer lineages is distinct. Further, these data provide information for evaluating the role of different immune cell populations in brain tumor growth and therapeutic responses. FUNDING This study was supported by the NIH (NS120547), a Developmental research project award (P50CA221747), ReMission Alliance, institutional funding from Northwestern University and the Lurie Comprehensive Cancer Center, and gifts from the Mosky family and Perry McKay. Performed in the Flow Cytometry & Cellular Imaging Core Facility at MD Anderson Cancer Center, this study received support in part from the NIH (CA016672) and the National Cancer Institute (NCI) Research Specialist award 1 (R50 CA243707). Additional support was provided by CCSG Bioinformatics Shared Resource 5 (P30 CA046592), a gift from Agilent Technologies, a Research Scholar Grant from the American Cancer Society (RSG-16-005-01), a Precision Health Investigator Award from University of Michigan (U-M) Precision Health, the NCI (R37-CA214955), startup institutional research funds from U-M, and a Biomedical Informatics & Data Science Training Grant (T32GM141746).
Collapse
Affiliation(s)
- Hinda Najem
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Martina Ott
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Cynthia Kassab
- Department of General Surgery, University of Texas Galveston, Galveston, United States of America
| | - Arvind Rao
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, United States of America
| | - Ganesh Rao
- Department of Neurosurgery, Baylor College of Medicine, Houston, United States of America
| | - Anantha Marisetty
- Department of Neurosurgery, Baylor College of Medicine, Houston, United States of America
| | - Adam M Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine Northwestern University, Chicago, United States of America
| | - Craig Horbinski
- Department of Neurological Surgery, Feinberg School of Medicine Northwestern University, Chicago, United States of America
| | - Roel Verhaak
- The Jackson Laboratory, Farmington, United States of America
| | - Anand Shankar
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, United States of America
| | - Santhoshi N Krishnan
- Department of Electrical and Computer Engineering, Rice University, Houston, United States of America
| | | | - Víctor A Arrieta
- Department of Neurological Surgery, Feinberg School of Medicine Northwestern University, Chicago, United States of America
| | - Pravesh Gupta
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Sherise D Ferguson
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Jason T Huse
- Department of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Gregory N Fuller
- Department of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - James P Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | | | | | - C David James
- Department of Neurological Surgery, Feinberg School of Medicine Northwestern University, Chicago, United States of America
| | - Leonidas C Platanias
- Department of Neurological Surgery, Feinberg School of Medicine Northwestern University, Chicago, United States of America
| | - Maciej S Lesniak
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, United States of America
| | - Jared K Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Amy B Heimberger
- Department of Neurological Surgery, Feinberg School of Medicine Northwestern University, Chicago, United States of America
| |
Collapse
|
14
|
Neutrophil and Natural Killer Cell Interactions in Cancers: Dangerous Liaisons Instructing Immunosuppression and Angiogenesis. Vaccines (Basel) 2021; 9:vaccines9121488. [PMID: 34960234 PMCID: PMC8709224 DOI: 10.3390/vaccines9121488] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/03/2021] [Accepted: 12/11/2021] [Indexed: 01/21/2023] Open
Abstract
The tumor immune microenvironment (TIME) has largely been reported to cooperate on tumor onset and progression, as a consequence of the phenotype/functional plasticity and adaptation capabilities of tumor-infiltrating and tumor-associated immune cells. Immune cells within the tumor micro (tissue-local) and macro (peripheral blood) environment closely interact by cell-to-cell contact and/or via soluble factors, also generating a tumor-permissive soil. These dangerous liaisons have been investigated for pillars of tumor immunology, such as tumor associated macrophages and T cell subsets. Here, we reviewed and discussed the contribution of selected innate immunity effector cells, namely neutrophils and natural killer cells, as "soloists" or by their "dangerous liaisons", in favoring tumor progression by dissecting the cellular and molecular mechanisms involved.
Collapse
|
15
|
Zhou J, Zhang S, Guo C. Crosstalk between macrophages and natural killer cells in the tumor microenvironment. Int Immunopharmacol 2021; 101:108374. [PMID: 34824036 DOI: 10.1016/j.intimp.2021.108374] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/17/2022]
Abstract
The tumor microenvironment (TME) is jointly constructed by a variety of cell types, including tumor cells, immune cells, fibroblasts, and epithelial cells, among others. The cells within the TME interact with each other and with tumor cells to influence tumor development and progression. As the most abundant immune cells in the TME, macrophages regulate the immune network by not only secreting a large amount of versatile cytokines but also expressing a series of ligands or receptors on the surface to interact with other cells directly. Due to their strong plasticity, they exert both immunostimulatory and immunosuppressive effects in the complex TME. The major effector cells of the immune system that directly target cancer cells include but are not limited to natural killer cells (NKs), dendritic cells (DCs), macrophages, polymorphonuclear leukocytes, mast cells, and cytotoxic T lymphocytes (CTLs). Among them, NK cells are the predominant innate lymphocyte subsets that mediate antitumor and antiviral responses. The activation and inhibition of NK cells are regulated by cytokines and the balance between activating and inhibitory receptors. There is an inextricable regulatory relationship between macrophages and NK cells. Herein, we systematically elaborate on the regulatory network between macrophages and NK cells through soluble mediator crosstalk and cell-to-cell interactions. We believe that a better understanding of the crosstalk between macrophages and NKs in the TME will benefit the development of novel macrophage- or NK cell-focused therapeutic strategies with superior efficacies in cancer therapy.
Collapse
Affiliation(s)
- Jingping Zhou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, PR China
| | - Shaolong Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, PR China
| | - Changying Guo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, PR China.
| |
Collapse
|
16
|
De Lerma Barbaro A, Palano MT, Cucchiara M, Gallazzi M, Mortara L, Bruno A. Metabolic Rewiring in the Tumor Microenvironment to Support Immunotherapy: A Focus on Neutrophils, Polymorphonuclear Myeloid-Derived Suppressor Cells and Natural Killer Cells. Vaccines (Basel) 2021; 9:vaccines9101178. [PMID: 34696286 PMCID: PMC8539473 DOI: 10.3390/vaccines9101178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/09/2021] [Accepted: 10/11/2021] [Indexed: 12/13/2022] Open
Abstract
Leukocytes often undergo rapid changes in cell phenotype, for example, from a resting to an activated state, which places significant metabolic demands on the cell. These rapid changes in metabolic demand need to be tightly regulated to support immune cell effector functions during the initiation and downregulation of an immune response. Prospects for implementing cancer immunotherapy also rest on the idea of optimizing the metabolic profile of immune cell effectors. Here, we examine this issue by focusing on neutrophils and NK cells as cells of increasing interest in cancer immunology and tumor immunometabolism, because they can be targeted or, in the case of NK, used as effectors in immunotherapy. In addition, neutrophils and NK cells have been shown to functionally interact. In the case of neutrophils, we also extended our interest to polymorphonuclear MDSC (PMN-MDSCs), since the granulocytic subset of MDSCs share many phenotypes and are functionally similar to pro-tumor neutrophils. Finally, we reviewed relevant strategies to target tumor metabolism, focusing on neutrophils and NK cells.
Collapse
Affiliation(s)
- Andrea De Lerma Barbaro
- Laboratory of Comparative Physiopathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
- Correspondence: (A.D.L.B.); (A.B.)
| | - Maria Teresa Palano
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, IRCCS MultiMedica, 20100 Milan, Italy; (M.T.P.); (M.C.)
| | - Martina Cucchiara
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, IRCCS MultiMedica, 20100 Milan, Italy; (M.T.P.); (M.C.)
| | - Matteo Gallazzi
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (M.G.); (L.M.)
| | - Lorenzo Mortara
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (M.G.); (L.M.)
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, IRCCS MultiMedica, 20100 Milan, Italy; (M.T.P.); (M.C.)
- Correspondence: (A.D.L.B.); (A.B.)
| |
Collapse
|
17
|
Hussain K, Cragg MS, Beers SA. Remodeling the Tumor Myeloid Landscape to Enhance Antitumor Antibody Immunotherapies. Cancers (Basel) 2021; 13:4904. [PMID: 34638388 PMCID: PMC8507767 DOI: 10.3390/cancers13194904] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/16/2021] [Accepted: 09/26/2021] [Indexed: 12/30/2022] Open
Abstract
Among the diverse tumor resident immune cell types, tumor-associated macrophages (TAMs) are often the most abundant, possess an anti-inflammatory phenotype, orchestrate tumor immune evasion and are frequently associated with poor prognosis. However, TAMs can also be harnessed to destroy antibody-opsonized tumor cells through the process of antibody-dependent cellular phagocytosis (ADCP). Clinically important tumor-targeting monoclonal antibodies (mAb) such as Rituximab, Herceptin and Cetuximab, function, at least in part, by inducing macrophages to eliminate tumor cells via ADCP. For IgG mAb, this is mediated by antibody-binding activating Fc gamma receptors (FcγR), with resultant phagocytic activity impacted by the level of co-engagement with the single inhibitory FcγRIIb. Approaches to enhance ADCP in the tumor microenvironment include the repolarization of TAMs to proinflammatory phenotypes or the direct augmentation of ADCP by targeting so-called 'phagocytosis checkpoints'. Here we review the most promising new strategies targeting the cell surface molecules present on TAMs, which include the inhibition of 'don't eat me signals' or targeting immunostimulatory pathways with agonistic mAb and small molecules to augment tumor-targeting mAb immunotherapies and overcome therapeutic resistance.
Collapse
Affiliation(s)
| | | | - Stephen A. Beers
- Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Tremona Road, Southampton SO16 6YD, UK; (K.H.); (M.S.C.)
| |
Collapse
|
18
|
Abstract
Abstract
The innate immune system is mandatory for the activation of antiviral host defense and eradication of the infection. In this regard, dendritic cells, natural killer cells, macrophages, neutrophils representing the cellular component, and cytokines, interferons, complement or Toll-Like Receptors, representing the mediators of unspecific response act together for both activation of the adaptive immune response and viral clearance. Of great importance is the proper functioning of the innate immune response from the very beginning. For instance, in the early stages of viral infection, the defective interferon response leads to uncontrolled viral replication and pathogen evasion, while hypersecretion during the later stages of infection generates hyperinflammation. This cascade activation of systemic inflammation culminates with cytokine storm syndrome and hypercoagulability state, due to a close interconnection between them. Thus an unbalanced reaction, either under- or over- stimulation of the innate immune system will lead to an uncoordinated response and unfavorable disease outcomes. Since both cellular and humoral factors are involved in the time-course of the innate immune response, in this review we aimed to address their gradual involvement in the antiviral response with emphasis on key steps in SARS-CoV-2 infection.
Collapse
|
19
|
Zheng W, Zhang S, Jiang S, Huang Z, Chen X, Guo H, Li M, Zheng S. Evaluation of immune status in testis and macrophage polarization associated with testicular damage in patients with nonobstructive azoospermia. Am J Reprod Immunol 2021; 86:e13481. [PMID: 34192390 DOI: 10.1111/aji.13481] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 06/08/2021] [Accepted: 06/28/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Immune cells residing in the testicular interstitial space form the immunological microenvironment of the testis. They are assumed to play a role in maintaining testicular homeostasis and immune privilege. However, the immune status and related cell polarization in patients with nonobstructive azoospermia (NOA) remains poorly characterized. System evaluation of the testis immunological microenvironment in NOA patients may help to reveal the mechanisms of idiopathic azoospermia. STUDY DESIGN The gene expression patterns of immune cells in normal human testes were systematically analyzed by single-cell RNA sequencing (scRNA-seq) and preliminarily verification by the human protein atlas (HPA) online database. The immune cell infiltration profiles and immune status of patients with NOA was analyzed by single-sample gene set enrichment analysis (ssGSEA) and gene set variation analysis (GSVA) based on four independent public microarray datasets (GSE45885, GSE45887, GSE9210, and GSE145467), obtained from Gene Expression Omnibus (GEO) online database. The relationship between immune cells and spermatogenesis score was further analyzed by Spearman correlation analysis. Finally, immunohistochemistry (IHC) staining was performed to identify the main immune cell types and their polarization status in patients with NOA. RESULTS Both scRNA-seq and HPA analysis showed that testicular macrophages represent the largest pool of immune cells in the normal testis, and also exhibit an attenuated inflammatory response by expressing high levels of tolerance proteins (CD163, IL-10, TGF-β, and VEGF) and reduced expression of TLR signaling pathway-related genes. Correlation analysis revealed that the testicular immune score and macrophages including M1 and M2 macrophages were significantly negatively correlated with spermatogenesis score in patients with NOA (GSE45885 and GSE45887). In addition, the number of M1 and M2 macrophages was significantly higher in patients with NOA (GSE9210 and GSE145467) than in normal testis. GSVA analysis indicated that the immunological microenvironment in NOA tissues was manifested by activated immune system and pro-inflammatory status. IHC staining results showed that the number of M1 and M2 macrophages was significantly higher in NOA tissues than in normal testis and negatively correlated with the Johnson score. CONCLUSION Testicular macrophage polarization may play a vital role in NOA development and is a promising potential therapeutic target.
Collapse
Affiliation(s)
- Wenzhong Zheng
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shiqiang Zhang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Shaoqin Jiang
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhangcheng Huang
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaobao Chen
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Huan Guo
- Department of Urology, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, China
| | - Mengqiang Li
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Song Zheng
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
20
|
Crawford L, Wyatt M, Bryers J, Ratner B. Biocompatibility Evolves: Phenomenology to Toxicology to Regeneration. Adv Healthc Mater 2021; 10:e2002153. [PMID: 33829678 PMCID: PMC8221530 DOI: 10.1002/adhm.202002153] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/26/2021] [Indexed: 12/20/2022]
Abstract
The word "biocompatibility," is inconsistent with the observations of healing for so-called biocompatible biomaterials. The vast majority of the millions of medical implants in humans today, presumably "biocompatible," are walled off by a dense, avascular, crosslinked collagen capsule, hardly suggestive of life or compatibility. In contrast, one is now seeing examples of implant biomaterials that lead to a vascularized reconstruction of localized tissue, a biological reaction different from traditional biocompatible materials that generate a foreign body capsule. Both the encapsulated biomaterials and the reconstructive biomaterials qualify as "biocompatible" by present day measurements of biocompatibility. Yet, this new generation of materials would seem to heal "compatibly" with the living organism, where older biomaterials are isolated from the living organism by the dense capsule. This review/perspective article will explore this biocompatibility etymological conundrum by reviewing the history of the concepts around biocompatibility, today's standard methods for assessing biocompatibility, a contemporary view of the foreign body reaction and finally, a compendium of new biomaterials that heal without the foreign body capsule. A new definition of biocompatibility is offered here to address advances in biomaterials design leading to biomaterials that heal into the body in a facile manner.
Collapse
Affiliation(s)
- Lars Crawford
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Meghan Wyatt
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - James Bryers
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Buddy Ratner
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
21
|
Perera Molligoda Arachchige AS. Human NK cells: From development to effector functions. Innate Immun 2021; 27:212-229. [PMID: 33761782 PMCID: PMC8054151 DOI: 10.1177/17534259211001512] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 02/19/2021] [Indexed: 12/11/2022] Open
Abstract
NK cells are the major lymphocyte subset of the innate immune system that mediates antiviral and anti-tumor responses. It is well established that they develop mechanisms to distinguish self from non-self during the process of NK cell education. Unlike T and B cells, natural killer cells lack clonotypic receptors and are activated after recognizing their target via germline-encoded receptors through natural cytotoxicity, cytokine stimulation, and Ab-dependent cellular cytotoxicity. Subsequently, they utilize cytotoxic granules, death receptor ligands, and cytokines to perform their effector functions. In this review, we provide a general overview of human NK cells, as opposed to murine NK cells, discussing their ontogeny, maturation, receptor diversity, types of responses, and effector functions. Furthermore, we also describe recent advances in human NK cell biology, including tissue-resident NK cell populations, NK cell memory, and novel approaches used to target NK cells in cancer immunotherapy.
Collapse
|
22
|
Mantovani A, Marchesi F, Jaillon S, Garlanda C, Allavena P. Tumor-associated myeloid cells: diversity and therapeutic targeting. Cell Mol Immunol 2021; 18:566-578. [PMID: 33473192 PMCID: PMC8027665 DOI: 10.1038/s41423-020-00613-4] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 11/25/2020] [Indexed: 12/15/2022] Open
Abstract
Myeloid cells in tumor tissues constitute a dynamic immune population characterized by a non-uniform phenotype and diverse functional activities. Both tumor-associated macrophages (TAMs), which are more abundantly represented, and tumor-associated neutrophils (TANs) are known to sustain tumor cell growth and invasion, support neoangiogenesis and suppress anticancer adaptive immune responses. In recent decades, several therapeutic approaches have been implemented in preclinical cancer models to neutralize the tumor-promoting roles of both TAMs and TANs. Some of the most successful strategies have now reached the clinic and are being investigated in clinical trials. In this review, we provide an overview of the recent literature on the ever-growing complexity of the biology of TAMs and TANs and the development of the most promising approaches to target these populations therapeutically in cancer patients.
Collapse
Affiliation(s)
- Alberto Mantovani
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy.
- Department of Biomedical Science, Humanitas University, Rozzano, Italy.
- The William Harvey Research Institute, Queen Mary University of London, London, UK.
| | - Federica Marchesi
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
- Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Sebastien Jaillon
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
- Department of Biomedical Science, Humanitas University, Rozzano, Italy
| | - Cecilia Garlanda
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
- Department of Biomedical Science, Humanitas University, Rozzano, Italy
| | - Paola Allavena
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
| |
Collapse
|
23
|
Luu TT, Schmied L, Nguyen NA, Wiel C, Meinke S, Mohammad DK, Bergö M, Alici E, Kadri N, Ganesan S, Höglund P. Short-term IL-15 priming leaves a long-lasting signalling imprint in mouse NK cells independently of a metabolic switch. Life Sci Alliance 2021; 4:4/4/e202000723. [PMID: 33593878 PMCID: PMC7918643 DOI: 10.26508/lsa.202000723] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 11/25/2022] Open
Abstract
NK cell reactivity is dynamically regulated by IL-15, and NK cells do not need more than a few minutes of exposure to remember the cytokine for several hours. IL-15 priming of NK cells is a broadly accepted concept, but the dynamics and underlying molecular mechanisms remain poorly understood. We show that as little as 5 min of IL-15 treatment in vitro, followed by removal of excess cytokines, results in a long-lasting, but reversible, augmentation of NK cell responsiveness upon activating receptor cross-linking. In contrast to long-term stimulation, improved NK cell function after short-term IL-15 priming was not associated with enhanced metabolism but was based on the increased steady-state phosphorylation level of signalling molecules downstream of activating receptors. Inhibition of JAK3 eliminated this priming effect, suggesting a cross talk between the IL-15 receptor and ITAM-dependent activating receptors. Increased signalling molecule phosphorylation levels, calcium flux, and IFN-γ secretion lasted for up to 3 h after IL-15 stimulation before returning to baseline. We conclude that IL-15 rapidly and reversibly primes NK cell function by modulating activating receptor signalling. Our findings suggest a mechanism by which NK cell reactivity can potentially be maintained in vivo based on only brief encounters with IL-15 trans-presenting cells.
Collapse
Affiliation(s)
- Thuy T Luu
- Department of Medicine Huddinge, Centre for Haematology and Regenerative Medicine (HERM), Karolinska Institutet, Huddinge, Sweden
| | - Laurent Schmied
- Department of Medicine Huddinge, Centre for Haematology and Regenerative Medicine (HERM), Karolinska Institutet, Huddinge, Sweden
| | - Ngoc-Anh Nguyen
- Department of Medicine Huddinge, Centre for Haematology and Regenerative Medicine (HERM), Karolinska Institutet, Huddinge, Sweden
| | - Clotilde Wiel
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Stephan Meinke
- Department of Medicine Huddinge, Centre for Haematology and Regenerative Medicine (HERM), Karolinska Institutet, Huddinge, Sweden
| | - Dara K Mohammad
- Department of Medicine Huddinge, Centre for Haematology and Regenerative Medicine (HERM), Karolinska Institutet, Huddinge, Sweden.,Department of Food Technology, College of Agricultural Engineering Sciences, Salahaddin University-Erbil, KRG-Kurdistan Region, Iraq
| | - Martin Bergö
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Evren Alici
- Department of Medicine Huddinge, Centre for Haematology and Regenerative Medicine (HERM), Karolinska Institutet, Huddinge, Sweden.,Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Nadir Kadri
- Department of Medicine Huddinge, Centre for Haematology and Regenerative Medicine (HERM), Karolinska Institutet, Huddinge, Sweden
| | - Sridharan Ganesan
- Department of Medicine Huddinge, Centre for Haematology and Regenerative Medicine (HERM), Karolinska Institutet, Huddinge, Sweden
| | - Petter Höglund
- Department of Medicine Huddinge, Centre for Haematology and Regenerative Medicine (HERM), Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
24
|
Bay ML, Heywood S, Wedell-Neergaard AS, Schauer T, Lehrskov LL, Christensen RH, Legård GE, Jensen PØ, Krogh-Madsen R, Ellingsgaard H. Human immune cell mobilization during exercise: effect of IL-6 receptor blockade. Exp Physiol 2020; 105:2086-2098. [PMID: 33006190 DOI: 10.1113/ep088864] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/28/2020] [Indexed: 12/18/2022]
Abstract
NEW FINDINGS What is the central question of this study? Does blockade of the IL-6 receptor by tocilizumab inhibit immune cell mobilization to the blood stream in humans during an acute bout of exercise? What is the main finding and its importance? Blockade of IL-6 receptor signalling by tocilizumab attenuates mobilization of NK cells and dendritic cells to the blood stream during exercise. This implies an inhibitory effect of tocilizumab on the innate immune response to physical stress, which could be considered in clinical settings. ABSTRACT Immune cells are recruited from their storage organs and the endothelial walls to the blood stream in response to physiological stress. This is essential for the recognition and clearing of infected, transformed or damaged cells. One of the most potent stimuli to recruit immune cells to the circulation is exercise. Accordingly, exercise has proven beneficial in disease settings, such as cancer and diabetes. Interleukin-6 (IL-6) is released from contracting skeletal muscle in response to exercise, and rodent studies have established a link between exercise-induced IL-6 and recruitment of natural killer (NK) cells. Whether exercise-induced IL-6 is involved in regulating NK cell mobilization in humans is unclear. This study explored the effect of IL-6 receptor blockade on immune cell mobilization during an acute bout of exercise in humans. In a randomized, placebo-controlled clinical study, abdominally obese humans receiving placebo infusions or tocilizumab infusions performed an acute bout of exercise before and after the intervention. Immune cell recruitment was measured by flow cytometry. IL-6 receptor blockade attenuated the increase of NK cells by 53% (mean difference -0.49 (95% CI: -0.89 to -0.08) × 109 cells L-1 , P < 0.001) and dendritic cells by 66% (mean difference -0.14 (95% CI: -0.28 to 0.010) × 109 cells L-1 , P < 0.001) induced by an acute bout of exercises. No changes were observed for T cells, monocytes and neutrophils. Treatments which interact with the exercise-mediated immune surveillance provide relevant clinical information in pursuing the 'exercise as medicine' concept.
Collapse
Affiliation(s)
- Marie Lund Bay
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sarah Heywood
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anne-Sophie Wedell-Neergaard
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Tim Schauer
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Louise Lang Lehrskov
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Regitse Højgaard Christensen
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Grit Elster Legård
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | - Rikke Krogh-Madsen
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Helga Ellingsgaard
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
Höglund P. Scandinavian Society for Immunology turns 50: Snapshots of Scandinavian immunology today and the future of this learned society. Scand J Immunol 2020; 92:e12976. [DOI: 10.1111/sji.12976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
26
|
Abstract
Natural killer (NK) cells are cytotoxic lymphocytes of the innate immune system that are capable of killing virally infected and/or cancerous cells. Nearly 20 years ago, NK cell-mediated immunotherapy emerged as a safe and effective treatment approach for patients with advanced-stage leukaemia. Subsequently, the field of NK cell-based cancer therapy has grown exponentially and currently constitutes a major area of immunotherapy innovation. In general, the development of NK cell-directed therapies has two main focal points: optimizing the source of therapeutic NK cells for adoptive transfer and enhancing NK cell cytotoxicity and persistence in vivo. A wide variety of sources of therapeutic NK cells are currently being tested clinically, including haploidentical NK cells, umbilical cord blood NK cells, stem cell-derived NK cells, NK cell lines, adaptive NK cells, cytokine-induced memory-like NK cells and chimeric antigen receptor NK cells. A plethora of methods to augment the cytotoxicity and longevity of NK cells are also under clinical investigation, including cytokine-based agents, NK cell-engager molecules and immune-checkpoint inhibitors. In this Review, we highlight the variety of ways in which diverse NK cell products and their auxiliary therapeutics are being leveraged to target human cancers. We also identify future avenues for NK cell therapy research.
Collapse
|
27
|
Jaillon S, Ponzetta A, Di Mitri D, Santoni A, Bonecchi R, Mantovani A. Neutrophil diversity and plasticity in tumour progression and therapy. Nat Rev Cancer 2020; 20:485-503. [PMID: 32694624 DOI: 10.1038/s41568-020-0281-y] [Citation(s) in RCA: 710] [Impact Index Per Article: 142.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/04/2020] [Indexed: 12/11/2022]
Abstract
Neutrophils play a key role in defence against infection and in the activation and regulation of innate and adaptive immunity. In cancer, tumour-associated neutrophils (TANs) have emerged as an important component of the tumour microenvironment. Here, they can exert dual functions. TANs can be part of tumour-promoting inflammation by driving angiogenesis, extracellular matrix remodelling, metastasis and immunosuppression. Conversely, neutrophils can also mediate antitumour responses by direct killing of tumour cells and by participating in cellular networks that mediate antitumour resistance. Neutrophil diversity and plasticity underlie the dual potential of TANs in the tumour microenvironment. Myeloid checkpoints as well as the tumour and tissue contexture shape neutrophil function in response to conventional therapies and immunotherapy. We surmise that neutrophils can provide tools to tailor current immunotherapy strategies and pave the way to myeloid cell-centred therapeutic strategies, which would be complementary to current approaches.
Collapse
Affiliation(s)
- Sebastien Jaillon
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy.
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy.
| | - Andrea Ponzetta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy
| | - Diletta Di Mitri
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy
| | - Angela Santoni
- Dipartimento di Medicina Molecolare Istituto Pasteur-Fondazione Cenci Bolognetti, Università di Roma 'La Sapienza', Rome, Italy
- IRCCS Neuromed, Pozzilli (IS), Italy
| | - Raffaella Bonecchi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy
| | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy.
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy.
- The William Harvey Research Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
28
|
Masselli E, Vaccarezza M, Carubbi C, Pozzi G, Presta V, Mirandola P, Vitale M. NK cells: A double edge sword against SARS-CoV-2. Adv Biol Regul 2020; 77:100737. [PMID: 32773100 PMCID: PMC7292949 DOI: 10.1016/j.jbior.2020.100737] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 06/04/2020] [Indexed: 02/06/2023]
Abstract
Natural killer (NK) cells are pivotal effectors of the innate immunity protecting an individual from microbes. They are the first line of defense against invading viruses, given their substantial ability to directly target infected cells without the need for specific antigen presentation. By establishing cellular networks with a variety of cell types such as dendritic cells, NK cells can also amplify and modulate antiviral adaptive immune responses. In this review, we will examine the role of NK cells in SARS-COV2 infections causing the ongoing COVID19 pandemic, keeping in mind the controversial role of NK cells specifically in viral respiratory infections and in inflammatory-driven lung damage. We discuss lessons learnt from previous coronavirus outbreaks in humans (caused by SARS-CoV-1 and MERS-COV).
Collapse
Affiliation(s)
- Elena Masselli
- Department of Medicine and Surgery, Anatomy Unit, University of Parma, Via Gramsci 14, 43126, Parma, Italy; University Hospital of Parma, AOU-PR, Via Gramsci 14, 43126, Parma, Italy
| | - Mauro Vaccarezza
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Bentley, Perth, WA, 6102, Australia
| | - Cecilia Carubbi
- Department of Medicine and Surgery, Anatomy Unit, University of Parma, Via Gramsci 14, 43126, Parma, Italy
| | - Giulia Pozzi
- Department of Medicine and Surgery, Anatomy Unit, University of Parma, Via Gramsci 14, 43126, Parma, Italy
| | - Valentina Presta
- Department of Medicine and Surgery, Anatomy Unit, University of Parma, Via Gramsci 14, 43126, Parma, Italy
| | - Prisco Mirandola
- Department of Medicine and Surgery, Anatomy Unit, University of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Marco Vitale
- Department of Medicine and Surgery, Anatomy Unit, University of Parma, Via Gramsci 14, 43126, Parma, Italy; University Hospital of Parma, AOU-PR, Via Gramsci 14, 43126, Parma, Italy.
| |
Collapse
|
29
|
Martinsen JT, Gunst JD, Højen JF, Tolstrup M, Søgaard OS. The Use of Toll-Like Receptor Agonists in HIV-1 Cure Strategies. Front Immunol 2020; 11:1112. [PMID: 32595636 PMCID: PMC7300204 DOI: 10.3389/fimmu.2020.01112] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/07/2020] [Indexed: 12/16/2022] Open
Abstract
Toll-like receptors (TLRs) are a family of pattern recognition receptors and part of the first line of defense against invading microbes. In humans, we know of 10 different TLRs, which are expressed to varying degrees in immune cell subsets. Engaging TLRs through their specific ligands leads to activation of the innate immune system and secondarily priming of the adaptive immune system. Because of these unique properties, TLR agonists have been investigated as immunotherapy in cancer treatment for many years, but in recent years there has also been growing interest in the use of TLR agonists in the context of human immunodeficiency virus type 1 (HIV-1) cure research. The primary obstacle to curing HIV-1 is the presence of a latent viral reservoir in transcriptionally silent immune cells. Due to the very limited transcription of the integrated HIV-1 proviruses, latently infected cells cannot be targeted and cleared by immune effector mechanisms. TLR agonists are very interesting in this context because of their potential dual effects as latency reverting agents (LRAs) and immune modulatory compounds. Here, we review preclinical and clinical data on the impact of TLR stimulation on HIV-1 latency as well as antiviral and HIV-1-specific immunity. We also focus on the promising role of TLR agonists in combination strategies in HIV-1 cure research. Different combinations of TLR agonists and broadly neutralizing antibodies or TLRs agonists as adjuvants in HIV-1 vaccines have shown very encouraging results in non-human primate experiments and these concepts are now moving into clinical testing.
Collapse
Affiliation(s)
| | | | | | - Martin Tolstrup
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
30
|
Locati M, Curtale G, Mantovani A. Diversity, Mechanisms, and Significance of Macrophage Plasticity. ANNUAL REVIEW OF PATHOLOGY 2020; 15:123-147. [PMID: 31530089 PMCID: PMC7176483 DOI: 10.1146/annurev-pathmechdis-012418-012718] [Citation(s) in RCA: 1320] [Impact Index Per Article: 264.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Macrophages are a diverse set of cells present in all body compartments. This diversity is imprinted by their ontogenetic origin (embryonal versus adult bone marrow-derived cells); the organ context; by their activation or deactivation by various signals in the contexts of microbial invasion, tissue damage, and metabolic derangement; and by polarization of adaptive T cell responses. Classic adaptive responses of macrophages include tolerance, priming, and a wide spectrum of activation states, including M1, M2, or M2-like. Moreover, macrophages can retain long-term imprinting of microbial encounters (trained innate immunity). Single-cell analysis of mononuclear phagocytes in health and disease has added a new dimension to our understanding of the diversity of macrophage differentiation and activation. Epigenetic landscapes, transcription factors, and microRNA networks underlie the adaptability of macrophages to different environmental cues. Macrophage plasticity, an essential component of chronic inflammation, and its involvement in diverse human diseases, most notably cancer, is discussed here as a paradigm.
Collapse
Affiliation(s)
- Massimo Locati
- Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, 20089 Milan, Italy
- Humanitas Clinical and Research Center, 20089 Milan, Italy;
| | - Graziella Curtale
- Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, 20089 Milan, Italy
- Humanitas Clinical and Research Center, 20089 Milan, Italy;
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, 20089 Milan, Italy;
- Humanitas University, 20090 Milan, Italy
- The William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| |
Collapse
|
31
|
Aghdassi AA, John DS, Sendler M, Storck C, van den Brandt C, Krüger B, Weiss FU, Mayerle J, Lerch MM. Absence of the neutrophil serine protease cathepsin G decreases neutrophil granulocyte infiltration but does not change the severity of acute pancreatitis. Sci Rep 2019; 9:16774. [PMID: 31727956 PMCID: PMC6856518 DOI: 10.1038/s41598-019-53293-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/30/2019] [Indexed: 12/20/2022] Open
Abstract
Acute pancreatitis is characterized by an early intracellular protease activation and invasion of leukocytes into the pancreas. Cathepsins constitute a large group of lysosomal enzymes, that have been shown to modulate trypsinogen activation and neutrophil infiltration. Cathepsin G (CTSG) is a neutrophil serine protease of the chymotrypsin C family known to degrade extracellular matrix components and to have regulatory functions in inflammatory disorders. The aim of this study was to investigate the role of CTSG in pancreatitis. Isolated acinar cells were exposed to recombinant CTSG and supramaximal cholezystokinin stimulation. In CTSG-/- mice and corresponding controls acute experimental pancreatitis was induced by serial caerulein injections. Severity was assessed by histology, serum enzyme levels and zymogen activation. Neutrophil infiltration was quantified by chloro-acetate ersterase staining and myeloperoxidase measurement. CTSG was expessed in inflammatory cells but not in pancreatic acinar cells. CTSG had no effect on intra-acinar-cell trypsinogen activation. In CTSG-/- mice a transient decrease of neutrophil infiltration into the pancreas and lungs was found during acute pancreatitis while the disease severity remained largely unchanged. CTSG is involved in pancreatic neutrophil infiltration during pancreatitis, albeit to a lesser degree than the related neutrophil (PMN) elastase. Its absence therefore leaves pancreatitis severity essentially unaffected.
Collapse
Affiliation(s)
- Ali A Aghdassi
- Department of Medicine A, University Medicine Greifswald, 17475, Greifswald, Germany.
| | - Daniel S John
- Department of Medicine A, University Medicine Greifswald, 17475, Greifswald, Germany
| | - Matthias Sendler
- Department of Medicine A, University Medicine Greifswald, 17475, Greifswald, Germany
| | - Christian Storck
- Department of Medicine A, University Medicine Greifswald, 17475, Greifswald, Germany
| | - Cindy van den Brandt
- Department of Medicine A, University Medicine Greifswald, 17475, Greifswald, Germany
| | - Burkhard Krüger
- Division of Medical Biology, University of Rostock, 18051, Rostock, Germany
| | - Frank Ulrich Weiss
- Department of Medicine A, University Medicine Greifswald, 17475, Greifswald, Germany
| | - Julia Mayerle
- Department of Medicine II, Ludwigs-Maximilians University Munich, 80539, Munich, Germany
| | - Markus M Lerch
- Department of Medicine A, University Medicine Greifswald, 17475, Greifswald, Germany
| |
Collapse
|
32
|
Meinke S, Karlström C, Höglund P. Complement as an Immune Barrier in Platelet Transfusion Refractoriness. Transfus Med Rev 2019; 33:231-235. [PMID: 31679761 DOI: 10.1016/j.tmrv.2019.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 09/05/2019] [Indexed: 12/15/2022]
Abstract
Patients with hematological cancers often have low platelet counts because of progressing bone marrow failure or cytostatic therapy. A large fraction of those patients need platelet transfusions, which can be life-saving if bleedings occur and also allow diagnostic and therapeutic interventions. The outcomes of platelet transfusions are not always easy to predict in terms of bleeding control or increase in platelet count. Reasons could be disease-specific factors, fever, or infections leading to platelet consumption, but the immune system may also be involved, in particular, in patients previously immunized against foreign human leukocyte antigens (HLA). Mechanisms underlying immune-mediated platelet destruction in the presence of antibodies again HLA are not well understood in clinical situations. This review discusses the role of complement in platelet refractoriness, with a focus on HLA antibody-mediated platelet refractoriness. We summarize recent work in this area, discuss complement-platelet interactions in general terms, and a suggest a possible role of complement in platelet transfusion in general.
Collapse
Affiliation(s)
- Stephan Meinke
- Center for Hematology and Regenerative Medicine (HERM), Department of Medicine Huddinge, Karolinska Institutet, Sweden
| | - Cecilia Karlström
- Center for Hematology and Regenerative Medicine (HERM), Department of Medicine Huddinge, Karolinska Institutet, Sweden; Theme Hematology, Karolinska University Hospital, Sweden
| | - Petter Höglund
- Center for Hematology and Regenerative Medicine (HERM), Department of Medicine Huddinge, Karolinska Institutet, Sweden; Function area Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Sweden.
| |
Collapse
|
33
|
Mantovani A, Ponzetta A, Inforzato A, Jaillon S. Innate immunity, inflammation and tumour progression: double-edged swords. J Intern Med 2019; 285:524-532. [PMID: 30873708 PMCID: PMC7174018 DOI: 10.1111/joim.12886] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Components of the cellular and the humoral arm of the immune system are essential elements of the tumour microenvironment (TME). The TME includes tumour-associated macrophages which have served as a paradigm for the cancer-promoting inflammation. Cytokines, IL-1 in particular, and complement have emerged as important players in tumour promotion. On the other hand, myeloid cells, innate lymphoid cells and complement have the potential, if unleashed, to mediate anticancer resistance. Targeting checkpoints restraining innate immunity, macrophages and natural killer (NK) cells in particular holds promise as a therapeutic strategy.
Collapse
Affiliation(s)
- A Mantovani
- Humanitas Clinical and Research Center - IRCCS, via Manzoni 56, 20089, Rozzano, (Mi), Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele Milan, Italy.,The William Harvey Research Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - A Ponzetta
- Humanitas Clinical and Research Center - IRCCS, via Manzoni 56, 20089, Rozzano, (Mi), Italy
| | - A Inforzato
- Humanitas Clinical and Research Center - IRCCS, via Manzoni 56, 20089, Rozzano, (Mi), Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele Milan, Italy
| | - S Jaillon
- Humanitas Clinical and Research Center - IRCCS, via Manzoni 56, 20089, Rozzano, (Mi), Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele Milan, Italy
| |
Collapse
|
34
|
Bassani B, Baci D, Gallazzi M, Poggi A, Bruno A, Mortara L. Natural Killer Cells as Key Players of Tumor Progression and Angiogenesis: Old and Novel Tools to Divert Their Pro-Tumor Activities into Potent Anti-Tumor Effects. Cancers (Basel) 2019; 11:461. [PMID: 30939820 PMCID: PMC6521276 DOI: 10.3390/cancers11040461] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/21/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023] Open
Abstract
. Immune cells, as a consequence of their plasticity, can acquire altered phenotype/functions within the tumor microenvironment (TME). Some of these aberrant functions include attenuation of targeting and killing of tumor cells, tolerogenic/immunosuppressive behavior and acquisition of pro-angiogenic activities. Natural killer (NK) cells are effector lymphocytes involved in tumor immunosurveillance. In solid malignancies, tumor-associated NK cells (TANK cells) in peripheral blood and tumor-infiltrating NK (TINK) cells show altered phenotypes and are characterized by either anergy or reduced cytotoxicity. Here, we aim at discussing how NK cells can support tumor progression and how induction of angiogenesis, due to TME stimuli, can be a relevant part on the NK cell-associated tumor supporting activities. We will review and discuss the contribution of the TME in shaping NK cell response favoring cancer progression. We will focus on TME-derived set of factors such as TGF-β, soluble HLA-G, prostaglandin E₂, adenosine, extracellular vesicles, and miRNAs, which can exhibit a dual function. On one hand, these factors can suppress NK cell-mediated activities but, on the other hand, they can induce a pro-angiogenic polarization in NK cells. Also, we will analyze the impact on cancer progression of the interaction of NK cells with several TME-associated cells, including macrophages, neutrophils, mast cells, cancer-associated fibroblasts, and endothelial cells. Then, we will discuss the most relevant therapeutic approaches aimed at potentiating/restoring NK cell activities against tumors. Finally, supported by the literature revision and our new findings on NK cell pro-angiogenic activities, we uphold NK cells to a key host cellular paradigm in controlling tumor progression and angiogenesis; thus, we should bear in mind NK cells like a TME-associated target for anti-tumor therapeutic approaches.
Collapse
Affiliation(s)
- Barbara Bassani
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Via Monte Generoso, n. 71, 21100 Varese, Italy.
| | - Denisa Baci
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy.
| | - Matteo Gallazzi
- Vascular Biology and Angiogenesis Laboratory, Scientific and Technologic Park, IRCCS MultiMedica, 20138 Milan, Italy.
| | - Alessandro Poggi
- UOSD Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy.
| | - Antonino Bruno
- Vascular Biology and Angiogenesis Laboratory, Scientific and Technologic Park, IRCCS MultiMedica, 20138 Milan, Italy.
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Via Monte Generoso, n. 71, 21100 Varese, Italy.
| |
Collapse
|
35
|
Tsoutsou PG, Zaman K, Martin Lluesma S, Cagnon L, Kandalaft L, Vozenin MC. Emerging Opportunities of Radiotherapy Combined With Immunotherapy in the Era of Breast Cancer Heterogeneity. Front Oncol 2018; 8:609. [PMID: 30619749 PMCID: PMC6305124 DOI: 10.3389/fonc.2018.00609] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022] Open
Abstract
The association of radiotherapy and immunotherapy has recently emerged as an exciting combination that might improve outcomes in many solid tumor settings. In the context of breast cancer, this opportunity is promising and under investigation. Given the heterogeneity of breast cancer, it might be meaningful to study the association of radiotherapy and immunotherapy distinctly among the various breast cancer subtypes. The use of biomarkers, such as tumor infiltrating lymphocytes, which are also associated to breast cancer heterogeneity, might provide an opportunity for tailored studies. This review highlights current knowledge of the association of radiotherapy and immunotherapy in the setting of breast cancer and attempts to highlight the therapeutic opportunities among breast cancer heterogeneity.
Collapse
Affiliation(s)
- Pelagia G Tsoutsou
- Division of Oncology, Radio-oncology Department, Vaudois University Hospital Centre (CHUV), Lausanne, Switzerland.,Radio-Oncology Research Laboratory, Vaudois University Hospital Centre (CHUV), Epalinges, Switzerland.,Radiation Oncology Department, Hôpital Neuchâtelois, La Chaux-de-Fonds, Switzerland
| | - Khalil Zaman
- Department of Oncology, Breast Center, Vaudois University Hospital Centre (CHUV), Lausanne, Switzerland
| | - Silvia Martin Lluesma
- Department of Oncology, Center of Experimental Therapeutics, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Laurene Cagnon
- Department of Oncology, Center of Experimental Therapeutics, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Lana Kandalaft
- Department of Oncology, Center of Experimental Therapeutics, Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Marie-Catherine Vozenin
- Radio-Oncology Research Laboratory, Vaudois University Hospital Centre (CHUV), Epalinges, Switzerland
| |
Collapse
|
36
|
Höglund P. Dear readers of theScandinavian Journal of Immunologyand ECI 2018 attendees. Scand J Immunol 2018. [DOI: 10.1111/sji.12706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|