1
|
Looi CK, Loo EM, Lim HC, Chew YL, Chin KY, Cheah SC, Goh BH, Mai CW. Revolutionizing the treatment for nasopharyngeal cancer: the impact, challenges and strategies of stem cell and genetically engineered cell therapies. Front Immunol 2024; 15:1484535. [PMID: 39450176 PMCID: PMC11499120 DOI: 10.3389/fimmu.2024.1484535] [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: 08/22/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a distinct malignancy of the nasopharynx and is consistently associated with the Epstein-Barr virus (EBV) infection. Its unique anatomical location and complex aetiology often result in advanced-stage disease at first diagnosis. While radiotherapy (RT) and chemotherapy have been the mainstays of treatment, they often fail to prevent tumour recurrence and metastasis, leading to high rates of treatment failure and mortality. Recent advancement in cell-based therapies, such as chimeric antigen receptor (CAR)-T cell therapy, have shown great promise in hematological malignancies and are now being investigated for NPC. However, challenges such as targeting specific tumour antigens, limited T cell persistence and proliferation, and managing treatment-related toxicities must be addressed. Extensive research is needed to enhance the effectiveness and safety of these therapies, paving the way for their integration into standard clinical practice for better management of NPC and a better quality of life for human health.
Collapse
Affiliation(s)
- Chin-King Looi
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Ee-Mun Loo
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
- Advanced Genomics Laboratory, AGTC Genomics, Kuala Lumpur, Malaysia
| | - Heng-Chee Lim
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Yik-Ling Chew
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Shiau-Chuen Cheah
- Faculty of Medicine and Health Sciences, UCSI University, Port Dickson, Negeri Sembilan, Malaysia
| | - Bey Hing Goh
- Sunway Biofunctional Molecules Discovery Centre, School of Medical and Life Sciences, Sunway University Malaysia, Bandar Sunway, Selangor Darul Ehsan, Malaysia
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Zhejiang, China
| | - Chun-Wai Mai
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
| |
Collapse
|
2
|
Huang M, Liu Y, Yan Q, Peng M, Ge J, Mo Y, Wang Y, Wang F, Zeng Z, Li Y, Fan C, Xiong W. NK cells as powerful therapeutic tool in cancer immunotherapy. Cell Oncol (Dordr) 2024; 47:733-757. [PMID: 38170381 DOI: 10.1007/s13402-023-00909-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Natural killer (NK) cells have gained considerable attention and hold great potential for their application in tumor immunotherapy. This is mainly due to their MHC-unrestricted and pan-specific recognition capabilities, as well as their ability to rapidly respond to and eliminate target cells. To artificially generate therapeutic NK cells, various materials can be utilized, such as peripheral blood mononuclear cells (PBMCs), umbilical cord blood (UCB), induced pluripotent stem cells (iPSCs), and NK cell lines. Exploiting the therapeutic potential of NK cells to treat tumors through in vivo and in vitro therapeutic modalities has yielded positive therapeutic results. CONCLUSION This review provides a comprehensive description of NK cell therapeutic approaches for tumors and discusses the current problems associated with these therapeutic approaches and the prospects of NK cell therapy for tumors.
Collapse
Affiliation(s)
- Mao Huang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yixuan Liu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Qijia Yan
- Department of Pathology, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Miao Peng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Junshang Ge
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yongzhen Mo
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yumin Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, China
| | - Fuyan Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yong Li
- Department of Medicine, Comprehensive Cancer Center, Baylor College of Medicine, Alkek Building, RM N720, Houston, TX, USA
| | - Chunmei Fan
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, 410013, Changsha, Hunan Province, China.
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| |
Collapse
|
3
|
Zhang Q, Lin J, Yang M, Li Z, Zhang M, Bu B. Therapeutic potential of natural killer cells in neuroimmunological diseases. Biomed Pharmacother 2024; 173:116371. [PMID: 38430631 DOI: 10.1016/j.biopha.2024.116371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/05/2024] Open
Abstract
Natural killer (NK) cells, a major component of the innate immune system, have prominent immunoregulatory, antitumor proliferation, and antiviral activities. NK cells act as a double-edged sword with therapeutic potential in neurological autoimmunity. Emerging evidence has identified NK cells are involved in the development and progression of neuroimmunological diseases such as multiple sclerosis, neuromyelitis optica spectrum disorders, autoimmune encephalitis, Guillain-Barré Syndrome, chronic inflammatory demyelinating polyneuropathy, myasthenia gravis, and idiopathic inflammatory myopathy. However, the regulatory mechanisms and functional roles of NK cells are highly variable in different clinical states of neuroimmunological diseases and need to be further determined. In this review, we summarize the evidence for the heterogenic involvement of NK cells in the above conditions. Further, we describe cutting-edge NK-cell-based immunotherapy for neuroimmunological diseases in preclinical and clinical development and highlight challenges that must be overcome to fully realize the therapeutic potential of NK cells.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jing Lin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mengge Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhijun Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Min Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Bitao Bu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
4
|
Wang S, Chen K, Jiang Y, Zhao G, Wang C, Fang H, Tang Q, Sun C, Zhang L, Wu H, Zhang LF, Li N. Breaking boundaries: Current progress of anticancer NK cell-based drug development. Drug Discov Today 2023; 28:103436. [PMID: 36370993 DOI: 10.1016/j.drudis.2022.103436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 10/14/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022]
Abstract
Natural killer (NK) cell therapy is emerging as a cancer treatment. NK cells are innate cytotoxic lymphocytes that act as first-line responders to kill target cells without prior encounters. NK cells recognize cancer cells, virus-infected cells, and other types of stressed cell through a reservoir of germline-encoded receptors. NK cells are safe for allogeneic applications. Therefore, they are the ideal off-the-shelf cell, which overcome the low efficiency issue caused by the patient-by-patient nature of autologous cell therapy. Unlike T cells, NK cells cannot form a strong immune memory; therefore, they suffer from short in vivo persistence. However, different from T cells, NK cells have a reservoir of innate immune receptors targeting a variety of malignant cells. In addition, they can utilize antibody guidance in target recognition. With suitable engineering, NK cells can function as universal anticancer drugs that are not restricted to HLA and cancer types, which will benefit the large cohort of patients with rare cancer types and patients with no convenient drug targets for precision and personalized medicine. Here, we summarize and discuss the designs of current anticancer NK cell therapies.
Collapse
Affiliation(s)
- Shuhang Wang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Kun Chen
- Guizhou Provincial People's Hospital, Guiyang, China
| | - Yale Jiang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Guo Zhao
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Caie Wang
- Department of Pharmacy, the First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan, China
| | - Hong Fang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qiyu Tang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Chao Sun
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | | | - Haiyang Wu
- TCRCure Biological Technology Co Ltd, Guangdong, China
| | - Li-Feng Zhang
- TCRCure Biological Technology Co Ltd, Guangdong, China.
| | - Ning Li
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
5
|
Cord Blood-Derived Natural Killer Cell Exploitation in Immunotherapy Protocols: More Than a Promise? Cancers (Basel) 2022; 14:cancers14184439. [PMID: 36139598 PMCID: PMC9496735 DOI: 10.3390/cancers14184439] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/25/2022] [Accepted: 09/09/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary NK cell anti-tumor activity against hematological malignancies is well-established and many studies support their role in the control of solid tumor growth and metastasis generation. However, tumor microenvironment may affect NK cell function. Ongoing studies are aimed to design novel immunotherapeutic protocols to combine NK cell-based immunotherapy with other therapeutic strategies to improve the anti-tumor NK cell response. In this context, UCB is one of the main sources of both mature NK cells and of CD34+ HSPC that can generate NK cells, both in-vivo and in-vitro. UCB-derived NK cells represent a valuable tool to perform in-vitro and preclinical analyses and are already used in several clinical settings, particularly against hematological malignancies. The present review describes the characteristics of different types of UCB-derived NK cells and the in-vitro models to expand them, both for research and clinical purposes in the context of cancer immunotherapy. Abstract In the last 20 years, Natural Killer (NK) cell-based immunotherapy has become a promising approach to target various types of cancer. Indeed, NK cells play a pivotal role in the first-line defense against tumors through major histocompatibility complex-independent immunosurveillance. Their role in the control of leukemia relapse has been clearly established and, moreover, the presence of NK cells in the tumor microenvironment (TME) generally correlates with good prognosis. However, it has also been observed that, often, NK cells poorly infiltrate the tumor tissue, and, in TME, their functions may be compromised by immunosuppressive factors that contribute to the failure of anti-cancer immune response. Currently, studies are focused on the design of effective strategies to expand NK cells and enhance their cytotoxic activity, exploiting different cell sources, such as peripheral blood (PB), umbilical cord blood (UCB) and NK cell lines. Among them, UCB represents an important source of mature NK cells and CD34+ Hematopoietic Stem and Progenitor Cells (HSPCs), as precursors of NK cells. In this review, we summarize the UCB-derived NK cell activity in the tumor context, review the different in-vitro models to expand NK cells from UCB, and discuss the importance of their exploitation in anti-tumor immunotherapy protocols.
Collapse
|
6
|
Valeri A, García-Ortiz A, Castellano E, Córdoba L, Maroto-Martín E, Encinas J, Leivas A, Río P, Martínez-López J. Overcoming tumor resistance mechanisms in CAR-NK cell therapy. Front Immunol 2022; 13:953849. [PMID: 35990652 PMCID: PMC9381932 DOI: 10.3389/fimmu.2022.953849] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Despite the impressive results of autologous CAR-T cell therapy in refractory B lymphoproliferative diseases, CAR-NK immunotherapy emerges as a safer, faster, and cost-effective approach with no signs of severe toxicities as described for CAR-T cells. Permanently scrutinized for its efficacy, recent promising data in CAR-NK clinical trials point out the achievement of deep, high-quality responses, thus confirming its potential clinical use. Although CAR-NK cell therapy is not significantly affected by the loss or downregulation of its CAR tumor target, as in the case of CAR-T cell, a plethora of common additional tumor intrinsic or extrinsic mechanisms that could also disable NK cell function have been described. Therefore, considering lessons learned from CAR-T cell therapy, the emergence of CAR-NK cell therapy resistance can also be envisioned. In this review we highlight the processes that could be involved in its development, focusing on cytokine addiction and potential fratricide during manufacturing, poor tumor trafficking, exhaustion within the tumor microenvironment (TME), and NK cell short in vivo persistence on account of the limited expansion, replicative senescence, and rejection by patient’s immune system after lymphodepletion recovery. Finally, we outline new actively explored alternatives to overcome these resistance mechanisms, with a special emphasis on CRISPR/Cas9 mediated genetic engineering approaches, a promising platform to optimize CAR-NK cell function to eradicate refractory cancers.
Collapse
Affiliation(s)
- Antonio Valeri
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Almudena García-Ortiz
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Eva Castellano
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Laura Córdoba
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Elena Maroto-Martín
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Jessica Encinas
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Alejandra Leivas
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Paula Río
- Division of Hematopoietic Innovative Therapies, Biomedical Innovation Unit, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) and Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
| | - Joaquín Martínez-López
- Hospital Universitario 12 de Octubre-Centro Nacional de Investigaciones Oncológicas (H12O-CNIO) Haematological Malignancies Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
- Department of Hematology, Hospital Universitario 12 de Octubre-Universidad Complutense, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- *Correspondence: Joaquín Martínez-López,
| |
Collapse
|
7
|
Chaudhry K, Dowlati E, Long MD, Geiger A, Lang H, Gomez EC, Muniraj N, Sanchez CE, Singh PK, Liu S, Bollard CM, Cruz CRY. Comparable transforming growth factor beta-mediated immune suppression in ex vivo-expanded natural killer cells from cord blood and peripheral blood: implications for adoptive immunotherapy. Cytotherapy 2022; 24:802-817. [PMID: 35589475 PMCID: PMC10258734 DOI: 10.1016/j.jcyt.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 04/01/2022] [Accepted: 04/03/2022] [Indexed: 11/27/2022]
Abstract
T cell-based therapies like genetically modified immune cells expressing chimeric antigen receptors have shown robust anti-cancer activity in vivo, especially in patients with blood cancers. However, extending this approach to an "off-the-shelf" setting can be challenging, as allogeneic T cells carry a significant risk of graft-versus-host disease (GVHD). By contrast, allogeneic natural killer (NK) cells recognize malignant cells without the need for prior antigen exposure and have been used safely in multiple cancer settings without the risk of GVHD. However, similar to T cells, NK cell function is negatively impacted by tumor-induced transforming growth factor beta (TGF-β) secretion, which is a ubiquitous and potent immunosuppressive mechanism employed by most malignancies. Allogeneic NK cells for adoptive immunotherapy can be sourced from peripheral blood (PB) or cord blood (CB), and the authors' group and others have previously shown that ex vivo expansion and gene engineering can overcome CB-derived NK cells' functional immaturity and poor cytolytic activity, including in the presence of exogenous TGF-β. However, a direct comparison of the effects of TGF-β-mediated immune suppression on ex vivo-expanded CB- versus PB-derived NK cell therapy products has not previously been performed. Here the authors show that PB- and CB-derived NK cells have distinctive gene signatures that can be overcome by ex vivo expansion. Additionally, exposure to exogenous TGF-β results in an upregulation of inhibitory receptors on NK cells, a novel immunosuppressive mechanism not previously described. Finally, the authors provide functional and genetic evidence that both PB- and CB-derived NK cells are equivalently susceptible to TGF-β-mediated immune suppression. The authors believe these results provide important mechanistic insights to consider when using ex vivo-expanded, TGF-β-resistant PB- or CB-derived NK cells as novel immunotherapy agents for cancer.
Collapse
Affiliation(s)
- Kajal Chaudhry
- Center for Cancer and Immunology Research, Children's National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA
| | - Ehsan Dowlati
- Department of Neurosurgery, Georgetown University Medical Center, Washington, DC, USA
| | - Mark D Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Ashley Geiger
- Center for Cancer and Immunology Research, Children's National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA
| | - Haili Lang
- Center for Cancer and Immunology Research, Children's National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA
| | - Eduardo C Gomez
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Nethaji Muniraj
- Center for Cancer and Immunology Research, Children's National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA
| | - Carlos E Sanchez
- Center for Cancer and Immunology Research, Children's National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA
| | - Prashant K Singh
- Genomics Shared Resource, Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; George Washington University Cancer Center, George Washington University, Washington, DC, USA.
| | - Conrad Russell Y Cruz
- Center for Cancer and Immunology Research, Children's National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; George Washington University Cancer Center, George Washington University, Washington, DC, USA.
| |
Collapse
|
8
|
Tang J, Zhu Q, Li Z, Yang J, Lai Y. Natural killer cell-targeted immunotherapy for cancer. Curr Stem Cell Res Ther 2022; 17:513-526. [PMID: 34994316 DOI: 10.2174/1574888x17666220107101722] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/12/2021] [Accepted: 11/15/2021] [Indexed: 11/22/2022]
Abstract
Natural killer (NK) cells were initially described in the early 1970s as major histocompatibility complex unrestricted killers due to their ability to spontaneously kill certain tumor cells. In the past decade, the field of NK cell-based treatment has been accelerating exponentially, holding a dominant position in cancer immunotherapy innovation. Generally, research on NK cell-mediated antitumor therapies can be categorized into three areas: choosing the optimal source of allogenic NK cells to yield massively amplified "off-the-shelf" products, improving NK cell cytotoxicity and longevity, and engineering NK cells with the ability of tumor-specific recognition. In this review, we focused on NK cell manufacturing techniques, some auxiliary methods to enhance the therapeutic efficacy of NK cells, chimeric antigen receptor NK cells, and monoclonal antibodies targeting inhibitory receptors, which can significantly augment the antitumor activity of NK cells. Notably, emerging evidence suggests that NK cells are a promising constituent of multipronged therapeutic strategies, strengthening immune responses to cancer.
Collapse
Affiliation(s)
- Jingyi Tang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qi Zhu
- Sichuan Fine Arts Institute, Chongqing, China
| | - Zhaoyang Li
- Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Jiahui Yang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yu Lai
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Nguyen OTP, Misun PM, Lohasz C, Lee J, Wang W, Schroeder T, Hierlemann A. An Immunocompetent Microphysiological System to Simultaneously Investigate Effects of Anti-Tumor Natural Killer Cells on Tumor and Cardiac Microtissues. Front Immunol 2021; 12:781337. [PMID: 34925361 PMCID: PMC8675866 DOI: 10.3389/fimmu.2021.781337] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/16/2021] [Indexed: 12/26/2022] Open
Abstract
Existing first-line cancer therapies often fail to cope with the heterogeneity and complexity of cancers, so that new therapeutic approaches are urgently needed. Among novel alternative therapies, adoptive cell therapy (ACT) has emerged as a promising cancer treatment in recent years. The limited clinical applications of ACT, despite its advantages over standard-of-care therapies, can be attributed to (i) time-consuming and cost-intensive procedures to screen for potent anti-tumor immune cells and the corresponding targets, (ii) difficulties to translate in-vitro and animal-derived in-vivo efficacies to clinical efficacy in humans, and (iii) the lack of systemic methods for the safety assessment of ACT. Suitable experimental models and testing platforms have the potential to accelerate the development of ACT. Immunocompetent microphysiological systems (iMPS) are microfluidic platforms that enable complex interactions of advanced tissue models with different immune cell types, bridging the gap between in-vitro and in-vivo studies. Here, we present a proof-of-concept iMPS that supports a triple culture of three-dimensional (3D) colorectal tumor microtissues, 3D cardiac microtissues, and human-derived natural killer (NK) cells in the same microfluidic network. Different aspects of tumor-NK cell interactions were characterized using this iMPS including: (i) direct interaction and NK cell-mediated tumor killing, (ii) the development of an inflammatory milieu through enrichment of soluble pro-inflammatory chemokines and cytokines, and (iii) secondary effects on healthy cardiac microtissues. We found a specific NK cell-mediated tumor-killing activity and elevated levels of tumor- and NK cell-derived chemokines and cytokines, indicating crosstalk and development of an inflammatory milieu. While viability and morphological integrity of cardiac microtissues remained mostly unaffected, we were able to detect alterations in their beating behavior, which shows the potential of iMPS for both, efficacy and early safety testing of new candidate ACTs.
Collapse
Affiliation(s)
- Oanh T. P. Nguyen
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Patrick M. Misun
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Christian Lohasz
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Jihyun Lee
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Weijia Wang
- Cell Systems Dynamics Group, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Timm Schroeder
- Cell Systems Dynamics Group, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Andreas Hierlemann
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| |
Collapse
|
10
|
Reevaluation of NOD/SCID Mice as NK Cell-Deficient Models. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8851986. [PMID: 34805408 PMCID: PMC8598338 DOI: 10.1155/2021/8851986] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 07/21/2021] [Accepted: 10/21/2021] [Indexed: 12/02/2022]
Abstract
Objective Natural killer (NK) cell-deficient mice are useful models in biomedical research. NOD/SCID mice have been used as a model of this type in research. However, the actual status of NK cells in NOD/SCID mice and CB17/SCID mice in comparison with that in BALB/c mice has not been sufficiently evaluated. Methods Splenocytes from naïve or poly(I:C)-treated mice were isolated for phenotyping and analysis of cytotoxicity-related molecules and inhibitory receptors; for cytotoxicity assay, purified NK cells were also used. Results The proportion of splenic NK cells did not differ significantly between NOD/SCID and CB17/SCID mice. The perforin levels in NK cells were similar between the poly(I:C)-treated CB17/SCID and NOD/SCID mice, while the granzyme B and NKG2A/C/E levels in NK cells from NOD/SCID mice were significantly lower than those from CB17/SCID mice. Moreover, the NKG2D and Ly49A levels in NK cells from NOD/SCID mice were higher than those from CB17/SCID. The splenocytes from CB17/SCID mice showed higher cytotoxicity than those from NOD/SCID mice, while the cytotoxicity of purified NK cells basically did not differ between the two strains. After in vitro stimulation with cytokines, the splenocytes from CB17/SCID mice showed higher IFN-γ production than those from NOD/SCID mice; however, NK cells did not. Conclusion There was no significant difference in the proportion of splenic NK cells between CB17/SCID and NOD/SCID mice, and the function of NK cells was only partially compromised in NOD/SCID mice. Caution should be taken when considering the use of NOD/SCID mice as an NK-deficient model.
Collapse
|
11
|
Castleman MJ, Dillon SM, Thompson TA, Santiago ML, McCarter MD, Barker E, Wilson CC. Gut Bacteria Induce Granzyme B Expression in Human Colonic ILC3s In Vitro in an IL-15-Dependent Manner. THE JOURNAL OF IMMUNOLOGY 2021; 206:3043-3052. [PMID: 34117105 DOI: 10.4049/jimmunol.2000239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/13/2021] [Indexed: 12/13/2022]
Abstract
Group 3 innate lymphoid cells (ILC3s) in the gut mucosa have long been thought to be noncytotoxic lymphocytes that are critical for homeostasis of intestinal epithelial cells through secretion of IL-22. Recent work using human tonsillar cells demonstrated that ILC3s exposed to exogenous inflammatory cytokines for a long period of time acquired expression of granzyme B, suggesting that under pathological conditions ILC3s may become cytotoxic. We hypothesized that inflammation associated with bacterial exposure might trigger granzyme B expression in gut ILC3s. To test this, we exposed human colon lamina propria mononuclear cells to a panel of enteric bacteria. We found that the Gram-negative commensal and pathogenic bacteria induced granzyme B expression in a subset of ILC3s that were distinct from IL-22-producing ILC3s. A fraction of granzyme B+ ILC3s coexpressed the cytolytic protein perforin. Granzyme B expression was mediated, in part, by IL-15 produced upon exposure to bacteria. ILC3s coexpressing all three IL-15R subunits (IL15Rα/β/γ) increased following bacterial stimulation, potentially allowing for cis presentation of IL-15 during bacterial exposure. Additionally, a large frequency of colonic myeloid dendritic cells expressed IL-15Rα, implicating myeloid dendritic cells in trans presentation of IL-15 to ILC3s. Tonsillar ILC3s minimally expressed granzyme B when exposed to the same bacteria or to rIL-15. Overall, these data establish the novel, to our knowledge, finding that human colonic ILC3s can express granzyme B in response to a subset of enteric bacteria through a process mediated by IL-15. These observations raise new questions about the multifunctional role of human gut ILC3s.
Collapse
Affiliation(s)
- Moriah J Castleman
- Division of Infectious Disease, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Stephanie M Dillon
- Division of Infectious Disease, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Tezha A Thompson
- Division of Infectious Disease, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Mario L Santiago
- Division of Infectious Disease, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Martin D McCarter
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO; and
| | - Edward Barker
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL
| | - Cara C Wilson
- Division of Infectious Disease, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO;
| |
Collapse
|
12
|
Chaudhry K, Dowlati E, Bollard CM. Chimeric antigen receptor-engineered natural killer cells: a promising cancer immunotherapy. Expert Rev Clin Immunol 2021; 17:643-659. [PMID: 33821731 DOI: 10.1080/1744666x.2021.1911648] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Introduction:Widespread success of CD19 chimeric antigen receptor (CAR) T cells for the treatment of hematological malignancies have shifted the focus from conventional cancer treatments toward adoptive immunotherapy. There are major efforts to improve CAR constructs and to identify new target antigens. Even though the Food and Drug Administration has approved commercialization of some CD19 CART cell therapies, there are still some limitations that restrict their widespread clinical use. The manufacture of autologous products for individual patients is logistically cumbersome and expensive and allogeneic T cell products may pose an appreciable risk of graft-versus-host disease (GVHD).Areas covered:Natural killer (NK) cells are an attractive alternative for CART-based immunotherapies. They have the innate ability to detect and eliminate malignant cells and are safer in the 'off-the-shelf' setting. This review discusses the current progress within the CAR NK cell field, including the challenges, and future prospects. Gene engineered NK cells was used as the search term in PubMed and Google Scholar through to December 2020.Expert opinion:CAR NK cell therapies hold promise as an 'off-the-shelf' cell therapy for cancer. It is hoped that an enhanced understanding of their immunobiology and molecular mechanisms of action will improve their in vivo potency.
Collapse
Affiliation(s)
- Kajal Chaudhry
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, WA, USA
| | - Ehsan Dowlati
- Department of Neurosurgery, Georgetown University Medical Center, Washington, WA, USA
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, WA, USA.,GW Cancer Center, George Washington University, Washington, DC, WA, USA.,Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, WA, USA
| |
Collapse
|
13
|
NK Cell Adoptive Immunotherapy of Cancer: Evaluating Recognition Strategies and Overcoming Limitations. Transplant Cell Ther 2020; 27:21-35. [PMID: 33007496 DOI: 10.1016/j.bbmt.2020.09.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/14/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023]
Abstract
Natural killer (NK) cells, the primary effector cells of the innate immune system, utilize multiple strategies to recognize tumor cells by (1) detecting the presence of activating receptor ligands, which are often upregulated in cancer; (2) targeting cells that have a loss of major histocompatibility complex (MHC); and (3) binding to antibodies that bind to tumor-specific antigens on the tumor cell surface. All these strategies have been successfully harnessed in adoptive NK cell immunotherapies targeting cancer. In this review, we review the applications of NK cell therapies across different tumor types. Similar to other forms of immunotherapy, tumor-induced immune escape and immune suppression can limit NK cell therapies' efficacy. Therefore, we also discuss how these limitations can be overcome by conferring NK cells with the ability to redirect their tumor-targeting capabilities and survive the immune-suppressive tumor microenvironment. Finally, we also discuss how future iterations can benefit from combination therapies with other immunotherapeutic agents.
Collapse
|
14
|
UV Light-inactivated HSV-1 Stimulates Natural Killer Cell-induced Killing of Prostate Cancer Cells. J Immunother 2020; 42:162-174. [PMID: 30933043 DOI: 10.1097/cji.0000000000000261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Herein we demonstrate that ultraviolet light-inactivated Herpes Simplex Virus-1 (UV-HSV-1) stimulates peripheral blood mononuclear cells (PBMCs) to lyse both androgen-sensitive and androgen-independent prostate cancer (PrCA) cell lines, but not the benign prostatic hyperplastic epithelial cell line, BPH-1, and is 1000-10,000-fold more potent at stimulating this killing than ultraviolet light-inactivated Vesicular Stomatitis Virus, adenovirus, reovirus or cytomegalovirus. Among PBMCs, natural killer (NK) cells appear to be a major cell type involved in this killing and UV-HSV-1 appears to directly and potently stimulate NK cell expression of CD69, degranulation, cytokine production, and migration to IL-8 in PC3 conditioned medium. We also found that UV-HSV-1 stimulates glycolysis in PBMCs and NK cells, and that 2-deoxyglucose and the protein kinase C inhibitor, Go6976, and the NFκB inhibitor, Bay 11-7082, all abrogate UV-HSV-1 activated killing of PC3 cells by PBMCs and NK cells. Using neutralizing anti-Toll-like receptor 2 (TLR2) we found that UV-HSV-1, like HSV-1, activates NK cells via TLR2. Taken together, these results are consistent with Toll-like receptor 2 ligands on UV-HSV-1 stimulating TLR2 on NK cells to activate protein kinase C, leading to enhanced glycolysis and NFκB activation, both of which play a critical role in this anti-PrCA innate immune response. Importantly, UV-HSV-1 synergizes with IL-15 to increase the cytolytic activity of PBMCs against PC3 cells and there was considerable donor-to-donor variation in killing ability. These results support the preclinical development of UV-HSV-1 as an adjuvant, in combination with IL-15, for cell infusions of healthy, preselected NK cells to treat PrCA.
Collapse
|
15
|
Mu YX, Zhao YX, Li BY, Bao HJ, Jiang H, Qi XL, Bai LY, Wang YH, Ma ZJ, Wu XY. A simple method for in vitro preparation of natural killer cells from cord blood. BMC Biotechnol 2019; 19:80. [PMID: 31752805 PMCID: PMC6869212 DOI: 10.1186/s12896-019-0564-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 09/27/2019] [Indexed: 12/12/2022] Open
Abstract
Background Cord Blood (CB) has been considered a promising source of natural killer (NK) cells for cellular immunotherapy. However, it is difficult to expand the large numbers of highly pure NK cells from CB without cell sorting and feeder cells/multiple cytokines. In this study, we try to develop a simple, safe and economical method for ex vivo expansion and purification of NK cells from CB without cell sorting and feeder cells/multiple cytokines. Results The large numbers (mean: 1.59 × 1010) of highly pure (≥90%) NK cells from CB could be obtained through interleukin-2, group A streptococcus and zoledronate stimulation of mononuclear cells using the 21-day culture approach. When compared to resting NK cells, expanded NK cells were a higher expression of activating receptors CD16, NKG2D, NKp30, NKp44, NKp46 and activating markers CD62L and CD69, while the inhibitory receptors, CD158a and CD158b remained largely unchanged. In addition, these cells showed a higher concentration of IFN-γ, TNF-α and GM-CSF secretion and cytotoxicity to K562 cells and acute myeloid leukemia targets than resting NK cells. Conclusion We develop a simple, safe and economical method to obtain high yield, purity, and functionality NK cells from CB without cell sorting and feeder cells/multiple cytokines.
Collapse
Affiliation(s)
- Yong Xu Mu
- Interventional Department, the First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Yu Xia Zhao
- Department of Blood, the People's Hospital of Xing'an League, Xing'an League, Inner Mongolia, China
| | - Bing Yao Li
- Department of Medicine, Chifeng Cancer Hospital, Chifeng, Inner Mongolia, China
| | - Hong Jing Bao
- Department of Blood, the People's Hospital of Xing'an League, Xing'an League, Inner Mongolia, China
| | - Hui Jiang
- Department of Blood, the People's Hospital of Xing'an League, Xing'an League, Inner Mongolia, China
| | - Xiao Lei Qi
- Department of Blood, the People's Hospital of Xing'an League, Xing'an League, Inner Mongolia, China
| | - Li Yun Bai
- Department of Blood, the People's Hospital of Xing'an League, Xing'an League, Inner Mongolia, China
| | - Yun Hong Wang
- Department of Technology, Stem Cell Medicine Engineering & Technology Research Center of Inner Mongolia, Huhhot, Inner Mongolia, China.,Department of Research and Development, Beijing Jingmeng Stem Cell Technology CO., LTD, Beijing, China
| | - Zhi Jie Ma
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Xiao Yun Wu
- Department of Technology, Stem Cell Medicine Engineering & Technology Research Center of Inner Mongolia, Huhhot, Inner Mongolia, China. .,Department of Research and Development, Beijing Jingmeng Stem Cell Technology CO., LTD, Beijing, China.
| |
Collapse
|
16
|
Ghaemdoust F, Keshavarz-Fathi M, Rezaei N. Natural killer cells and cancer therapy, what we know and where we are going. Immunotherapy 2019; 11:1231-1251. [DOI: 10.2217/imt-2019-0040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cells are among the significant components of innate immune system and they have come to the first line of defense against tumor cells developing inside the body. CD56lo/CD16+ NK cells are highly cytotoxic and CD56hi NK cells can produce cytokines and perform a regulatory function. Specific features of NK cells have made them a unique choice for cancer immunotherapy. Simple interventions like cytokine-injection to boost the internal NK cells were the first trials to target these cells. Nowadays, many other types of intervention are under investigation, such as adoptive NK cell immunotherapy. In this paper, we will discuss the biology and function of NK cells in cancer immunosurveillance and therapeutic approaches against cancer via using NK cells.
Collapse
Affiliation(s)
- Faezeh Ghaemdoust
- School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, 1419733151, Iran
| | - Mahsa Keshavarz-Fathi
- School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, 1419733151, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, 1419733151, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, 1419733151, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| |
Collapse
|
17
|
Ex Vivo-expanded Natural Killer Cells Derived From Long-term Cryopreserved Cord Blood are Cytotoxic Against Primary Breast Cancer Cells. J Immunother 2019; 41:64-72. [PMID: 29189387 DOI: 10.1097/cji.0000000000000192] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
With over 600,000 units of umbilical cord blood (CB) stored on a global scale, it is important to elucidate the therapeutic abilities of this cryopreserved reservoir. In the advancing field of natural killer (NK) cell cancer immunotherapy, CB has proven to be a promising and noninvasive source of therapeutic NK cells. Although studies have proven the clinical efficacy of using long-term cryopreserved CB in the context of hematopoietic stem cell transplantations, little is known about its use for the ex vivo expansion of effector immune cells. Therefore, our group sought to derive ex vivo-expanded NK cells from long-term cryopreserved CB, using an artificial antigen presenting cell-mediated expansion technique. We compared the expansion potential and antitumor effector function of CB-derived NK (CB-NK) cells expanded from fresh (n=4), short-term cryopreserved (<1-year old, n=5), and long-term cryopreserved (1-10-year old, n=5) CB. Here, we demonstrated it is possible to obtain an exponential amount of expanded CB-NK cells from long-term cryopreserved CB. Ex vivo-expanded CB-NK cells had an increased surface expression of activating markers and showed potent antitumor function by producing robust levels of proinflammatory cytokines, interferon-γ, and tumor necrosis factor-α. Moreover, expanded CB-NK cells (n=3-5) demonstrated cytotoxicity towards primary breast cancer cells (n=2) derived from a triple-negative breast cancer and an estrogen receptor-positive/progesterone receptor-positive breast cancer patient. Long-term cryopreservation had no effect on the expansion potential or effector function of expanded CB-NK cells. Therefore, we propose that long-term cryopreserved CB remains clinically useful for the ex vivo expansion of therapeutic NK cells.
Collapse
|
18
|
Natural Killer Cells as Allogeneic Effectors in Adoptive Cancer Immunotherapy. Cancers (Basel) 2019; 11:cancers11060769. [PMID: 31163679 PMCID: PMC6628161 DOI: 10.3390/cancers11060769] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 05/25/2019] [Accepted: 05/30/2019] [Indexed: 02/07/2023] Open
Abstract
Natural killer (NK) cells are attractive within adoptive transfer settings in cancer immunotherapy due to their potential for allogeneic use; their alloreactivity is enhanced under conditions of killer immunoglobulin-like receptor (KIR) mismatch with human leukocyte antigen (HLA) ligands on cancer cells. In addition to this, NK cells are platforms for genetic modification, and proliferate in vivo for a shorter time relative to T cells, limiting off-target activation. Current clinical studies have demonstrated the safety and efficacy of allogeneic NK cell adoptive transfer therapies as a means for treatment of hematologic malignancies and, to a lesser extent, solid tumors. However, challenges associated with sourcing allogeneic NK cells have given rise to controversy over the contribution of NK cells to graft-versus-host disease (GvHD). Specifically, blood-derived NK cell infusions contain contaminating T cells, whose activation with NK-stimulating cytokines has been known to lead to heightened release of proinflammatory cytokines and trigger the onset of GvHD in vivo. NK cells sourced from cell lines and stem cells lack contaminating T cells, but can also lack many phenotypic characteristics of mature NK cells. Here, we discuss the available published evidence for the varying roles of NK cells in GvHD and, more broadly, their use in allogeneic adoptive transfer settings to treat various cancers.
Collapse
|
19
|
Rechavi E, Somech R. Maturation of the immune system in the fetus and the implications for congenital CMV. Best Pract Res Clin Obstet Gynaecol 2019; 60:35-41. [PMID: 30981539 DOI: 10.1016/j.bpobgyn.2019.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 03/01/2019] [Indexed: 12/28/2022]
Abstract
Congenital cytomegalovirus (CMV) infection is the most prevalent and consequential congenital infection, among others, that affects approximately 0.6% of all live births worldwide. Timing of maternal infection and maternal immune status largely determine the likelihood of a symptomatic infection. However, recent studies suggest that the fetal immune system, long perceived as naïve and immature, may also play a role in deciding the outcome of congenital CMV infection. Here, we review the development of four immune cells most pertinent to CMV control in the human fetus. αβT cells, B cells, natural killer (NK) cells, and γδT cells are all present, mature and partially functional in utero, and are capable of mounting some form of response to congenital CMV infection. Whether this response is negligible, effective, or harmful remains an open question. Expanding our knowledge of normal and abnormal immune development could provide clinicians with more accurate tools for the detection, monitoring, and treatment of congenital CMV infection in fetuses.
Collapse
Affiliation(s)
- Erez Rechavi
- Pediatric Department A and Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Affiliated with Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Raz Somech
- Pediatric Department A and Immunology Service, Jeffrey Modell Foundation Center, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Affiliated with Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
20
|
Zoledronate increases enrichment, activation and expansion of natural killer cells from umbilical cord blood. Hum Cell 2018; 31:310-312. [PMID: 29948879 DOI: 10.1007/s13577-018-0213-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 05/30/2018] [Indexed: 12/12/2022]
|
21
|
Manzini C, Venè R, Cossu I, Gualco M, Zupo S, Dono M, Spagnolo F, Queirolo P, Moretta L, Mingari MC, Pietra G. Cytokines can counteract the inhibitory effect of MEK-i on NK-cell function. Oncotarget 2018; 7:60858-60871. [PMID: 27563819 PMCID: PMC5308621 DOI: 10.18632/oncotarget.11504] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/03/2016] [Indexed: 01/23/2023] Open
Abstract
Oncogene-targeted therapies based on mutated BRAF- and/or MEK-specific inhibitors have been developed for melanoma treatment. Although these drugs induce tumor regression in a high percentage of patients, clinical responses are frequently limited in time and tumors often recur. Recent studies suggested that the combination of BRAF/MEK inhibition with immunotherapy could represent a promising strategy for the cure of melanoma. NK cells are suitable effectors for tumor immunotherapy. Here we show that PLX4032 (a mutant BRAFV600 inhibitor) had no effect on the functional properties of NK cells cultured in the presence of IL-2 or IL-15. In contrast, PD0325901 (a MEK inhibitor) induced the down-regulation of the main activating NK receptors and inhibited NK cell function. Importantly, PD0325901 did not affect the anti-tumor activity of NK cells that had been exposed to a combination of IL-15 and IL-18. In addition, both PLX4032 and PD0325901 did not exert any inhibitory effect on in vitro IL-2 or IL-15 pre-activated NK cells. Our data may provide a rationale for future clinical protocols that combine IL-15/IL-18 cytokine administration with MEK inhibitors. In addition, they suggest that oncogene-targeting drugs are compatible with NK-based adoptive therapy.
Collapse
Affiliation(s)
| | - Roberta Venè
- Oncologia Molecolare e Angiogenesi, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Irene Cossu
- IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Marina Gualco
- Anatomia Patologica, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Simonetta Zupo
- Diagnostica Molecolare, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Mariella Dono
- Diagnostica Molecolare, IRCCS AOU San Martino-IST, Genoa, Italy
| | | | - Paola Queirolo
- Oncologia Medica 2, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Lorenzo Moretta
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Maria Cristina Mingari
- Immunologia, IRCCS AOU San Martino-IST, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Gabriella Pietra
- Immunologia, IRCCS AOU San Martino-IST, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| |
Collapse
|
22
|
Oberschmidt O, Kloess S, Koehl U. Redirected Primary Human Chimeric Antigen Receptor Natural Killer Cells As an "Off-the-Shelf Immunotherapy" for Improvement in Cancer Treatment. Front Immunol 2017. [PMID: 28649246 PMCID: PMC5465249 DOI: 10.3389/fimmu.2017.00654] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Primary human natural killer (NK) cells recognize and subsequently eliminate virus infected cells, tumor cells, or other aberrant cells. However, cancer cells are able to develop tumor immune escape mechanisms to undermine this immune control. To overcome this obstacle, NK cells can be genetically modified to express chimeric antigen receptors (CARs) in order to improve specific recognition of cancer surface markers (e.g., CD19, CD20, and ErbB2). After target recognition, intracellular CAR domain signaling (CD3ζ, CD28, 4-1BB, and 2B4) leads to activation of PI3K or DNAX proteins (DAP10, DAP12) and finally to enhanced cytotoxicity, proliferation, and/or interferon γ release. This mini-review summarizes both the first preclinical trials with CAR-engineered primary human NK cells and the translational implications for “off-the-shelf immunotherapy” in cancer treatment. Signal transduction in NK cells as well as optimization of CAR signaling will be described, becoming more and more a focal point of interest in addition to redirected T cells. Finally, strategies to overcome off-target effects will be discussed in order to improve future clinical trials and to avoid attacking healthy tissues.
Collapse
Affiliation(s)
- Olaf Oberschmidt
- Institute of Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| | - Stephan Kloess
- Institute of Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| | - Ulrike Koehl
- Institute of Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| |
Collapse
|
23
|
Dickinson AM, Norden J, Li S, Hromadnikova I, Schmid C, Schmetzer H, Jochem-Kolb H. Graft-versus-Leukemia Effect Following Hematopoietic Stem Cell Transplantation for Leukemia. Front Immunol 2017. [PMID: 28638379 PMCID: PMC5461268 DOI: 10.3389/fimmu.2017.00496] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The success of hematopoietic stem cell transplantation (HSCT) lies with the ability of the engrafting immune system to remove residual leukemia cells via a graft-versus-leukemia effect (GvL), caused either spontaneously post-HSCT or via donor lymphocyte infusion. GvL effects can also be initiated by allogenic mismatched natural killer cells, antigen-specific T cells, and activated dendritic cells of leukemic origin. The history and further application of this GvL effect and the main mechanisms will be discussed and reviewed in this chapter.
Collapse
Affiliation(s)
- Anne M Dickinson
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Jean Norden
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Shuang Li
- Third Faculty of Medicine, Department of Molecular Biology and Cell Pathology, Charles University, Prague, Czechia
| | - Ilona Hromadnikova
- Third Faculty of Medicine, Department of Molecular Biology and Cell Pathology, Charles University, Prague, Czechia
| | - Christoph Schmid
- Department for Hematopoietic Cell Transplantation, University Hospital Augsburg, Munich, Germany
| | - Helga Schmetzer
- Department for Hematopoietic Cell Transplantation, Internal Medicine III, Hospital of the University of Munich, Munich, Germany
| | - Hans Jochem-Kolb
- Department of Hematology-Oncology Immunology Infectious Diseases, Klinikum München-Schwabing, Munich, Germany
| |
Collapse
|
24
|
Domogala A, Blundell M, Thrasher A, Lowdell MW, Madrigal JA, Saudemont A. Natural killer cells differentiated in vitro from cord blood CD34 + cells are more advantageous for use as an immunotherapy than peripheral blood and cord blood natural killer cells. Cytotherapy 2017; 19:710-720. [PMID: 28428057 DOI: 10.1016/j.jcyt.2017.03.068] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/06/2017] [Accepted: 03/13/2017] [Indexed: 12/24/2022]
Abstract
BACKGROUND AIMS Natural killer (NK) cells have the potential to become a successful immunotherapy as they can target malignant cells without being direct effectors of graft-versus-host disease. Our group has previously shown that large numbers of functional NK cells can be differentiated in vitro from umbilical cord blood (CB) CD34+ cells. To produce a clinically relevant and effective immunotherapy, we hypothesized that it is essential that the NK cells are able to proliferate and persist in vivo while maintaining an optimal activation status and killing capacity. METHODS We evaluated the proliferation capacity, telomere length and terminal differentiation markers expressed by NK cells differentiated in vitro. We also determined how their cytotoxicity compared with peripheral blood (PB) NK cells and CBNK cells when targeting patient acute myeloid leukemia (AML) blasts and solid tumor cell lines. RESULTS We found that the differentiated NK cells could respond to interleukin-2 and proliferate in vitro. Telomere length was significantly increased, whereas CD57 expression was significantly reduced compared with PBNK cells. The cytotoxicity of the differentiated NK cells was equivalent to that of the PBNK and CBNK cell controls, and priming consistently led to higher levels of killing of patient leukemic blasts and solid tumor cell lines in vitro. Interestingly, this activation step was not required to observe killing of patient AML blasts in vivo. CONCLUSION We are able to generate NK cells from CBCD34+ cells in high numbers, allowing for multiple infusions of highly cytotoxic NK cells that have potential to further proliferate in vivo, making them a desirable product for application as an immunotherapy in the clinic.
Collapse
Affiliation(s)
- Anna Domogala
- Anthony Nolan Research Institute, London, UK; University College London, London, UK
| | | | | | | | - J Alejandro Madrigal
- Anthony Nolan Research Institute, London, UK; University College London, London, UK
| | - Aurore Saudemont
- Anthony Nolan Research Institute, London, UK; University College London, London, UK.
| |
Collapse
|
25
|
Sarvaria A, Jawdat D, Madrigal JA, Saudemont A. Umbilical Cord Blood Natural Killer Cells, Their Characteristics, and Potential Clinical Applications. Front Immunol 2017; 8:329. [PMID: 28386260 PMCID: PMC5362597 DOI: 10.3389/fimmu.2017.00329] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/07/2017] [Indexed: 12/13/2022] Open
Abstract
Natural killer (NK) cells are lymphocytes of the innate immune system able to kill different targets such as cancer cells and virally infected cells without prior activation making then attractive candidates for cancer immunotherapy. Umbilical cord blood (UCB) has become a source of hematopoietic stem cells for transplantation but as we gain a better understanding of the characteristics of each immune cell that UCB contains, we will also be able to develop new cell therapies for cancer. In this review, we present what is currently known of the phenotype and functions of UCB NK cells and how these cells could be used in the future for cancer immunotherapy.
Collapse
Affiliation(s)
- Anushruti Sarvaria
- Anthony Nolan Research Institute, London, UK; Cancer Institute, University College London, London, UK
| | - Dunia Jawdat
- King Abdullah International Medical Research Center , Riyadh , Saudi Arabia
| | - J Alejandro Madrigal
- Anthony Nolan Research Institute, London, UK; Cancer Institute, University College London, London, UK
| | - Aurore Saudemont
- Anthony Nolan Research Institute, London, UK; Cancer Institute, University College London, London, UK
| |
Collapse
|
26
|
da Silva MB, da Cunha FF, Terra FF, Camara NOS. Old game, new players: Linking classical theories to new trends in transplant immunology. World J Transplant 2017; 7:1-25. [PMID: 28280691 PMCID: PMC5324024 DOI: 10.5500/wjt.v7.i1.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/16/2016] [Accepted: 12/07/2016] [Indexed: 02/05/2023] Open
Abstract
The evolutionary emergence of an efficient immune system has a fundamental role in our survival against pathogenic attacks. Nevertheless, this same protective mechanism may also establish a negative consequence in the setting of disorders such as autoimmunity and transplant rejection. In light of the latter, although research has long uncovered main concepts of allogeneic recognition, immune rejection is still the main obstacle to long-term graft survival. Therefore, in order to define effective therapies that prolong graft viability, it is essential that we understand the underlying mediators and mechanisms that participate in transplant rejection. This multifaceted process is characterized by diverse cellular and humoral participants with innate and adaptive functions that can determine the type of rejection or promote graft acceptance. Although a number of mediators of graft recognition have been described in traditional immunology, recent studies indicate that defining rigid roles for certain immune cells and factors may be more complicated than originally conceived. Current research has also targeted specific cells and drugs that regulate immune activation and induce tolerance. This review will give a broad view of the most recent understanding of the allogeneic inflammatory/tolerogenic response and current insights into cellular and drug therapies that modulate immune activation that may prove to be useful in the induction of tolerance in the clinical setting.
Collapse
|
27
|
Saudemont A, Madrigal JA. Immunotherapy after hematopoietic stem cell transplantation using umbilical cord blood-derived products. Cancer Immunol Immunother 2017; 66:215-221. [PMID: 27271550 PMCID: PMC11028513 DOI: 10.1007/s00262-016-1852-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 05/25/2016] [Indexed: 02/02/2023]
Abstract
Umbilical cord blood (UCB) is being increasingly used as a source of hematopoietic stem cells (HSC) for transplantation. UCB transplantation (UCBT) has some advantages such as less stringent HLA-matching requirements, fast availability of the graft and reduced incidence and severity of graft-versus-host disease. However, UCBT is also associated with a higher incidence of infection, graft failure, slow engraftment and slow immune reconstitution. UCB is mainly used as a source of HSC; however, it is also rich in immune cells that could be used to treat some of the main complications post-UCBT as well as other diseases, thus implicating the use of UCB for immunotherapy. Here, we aim to describe some of the therapies currently developed that use UCB as a cell source, focusing in particular on regulatory T cells and natural killer cells.
Collapse
Affiliation(s)
- Aurore Saudemont
- University College London, London, UK
- Anthony Nolan Research Institute, Fleet Road, London, NW3 2QG, UK
| | - J Alejandro Madrigal
- University College London, London, UK.
- Anthony Nolan Research Institute, Fleet Road, London, NW3 2QG, UK.
| |
Collapse
|
28
|
Strauss-Albee DM, Liang EC, Ranganath T, Aziz N, Blish CA. The newborn human NK cell repertoire is phenotypically formed but functionally reduced. CYTOMETRY PART B-CLINICAL CYTOMETRY 2016; 92:33-41. [PMID: 27718327 DOI: 10.1002/cyto.b.21485] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 09/15/2016] [Accepted: 10/05/2016] [Indexed: 11/11/2022]
Abstract
BACKGROUND Infection is a leading cause of death worldwide in babies under 1 month of age. Better vaccines and therapeutics are desperately needed for this vulnerable population. METHODS Because newborns rely heavily on the innate immune system, we evaluated cell phenotype and function of some of the earliest cellular responders during infection, natural killer (NK) cells. We used mass cytometry to provide a comprehensive comparison of NK cells from umbilical cord blood and adult peripheral blood. RESULTS In unsupervised analyses, including viSNE and principal component analysis, the structure of the cord blood and adult NK cell repertoires are highly similar, distinguishable mainly by maturity-related markers expressed on rare subpopulations of cells. However, in functional analyses, cord blood NK cells show reduced degranulation and cytokine production following target recognition, as well as antibody-dependent cell-mediated cytotoxicity and apoptosis induction in targets. CONCLUSIONS These findings show that the structure of the NK cell repertoire is intact at birth, suggesting great potential for vaccine and therapeutic strategies targeting this cell population. © 2016 International Clinical Cytometry Society.
Collapse
Affiliation(s)
- Dara M Strauss-Albee
- Immunology Program, Stanford University School of Medicine, Stanford, California, 94305.,Department of Medicine, Stanford University School of Medicine, Stanford, California, 94305
| | - Emily C Liang
- Department of Medicine, Stanford University School of Medicine, Stanford, California, 94305
| | - Thanmayi Ranganath
- Department of Medicine, Stanford University School of Medicine, Stanford, California, 94305
| | - Natali Aziz
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California, 94305
| | - Catherine A Blish
- Immunology Program, Stanford University School of Medicine, Stanford, California, 94305.,Department of Medicine, Stanford University School of Medicine, Stanford, California, 94305
| |
Collapse
|
29
|
Laza-Briviesca R, Saudemont A, Madrigal JA. [Treatment of umbilical cord blood units with interleukin-15 improves hematopoietic stem cell engraftment]. CIR CIR 2016; 84:267-8. [PMID: 27324929 DOI: 10.1016/j.circir.2016.05.001] [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]
Affiliation(s)
| | - A Saudemont
- Anthony Nolan Research Institute, London, Reino Unido; University College London, London, Reino Unido
| | - J A Madrigal
- Anthony Nolan Research Institute, London, Reino Unido; University College London, London, Reino Unido.
| |
Collapse
|
30
|
Samudio I, Rezvani K, Shaim H, Hofs E, Ngom M, Bu L, Liu G, Lee JTC, Imren S, Lam V, Poon GFT, Ghaedi M, Takei F, Humphries K, Jia W, Krystal G. UV-inactivated HSV-1 potently activates NK cell killing of leukemic cells. Blood 2016; 127:2575-86. [PMID: 26941401 PMCID: PMC4892253 DOI: 10.1182/blood-2015-04-639088] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 02/26/2016] [Indexed: 11/20/2022] Open
Abstract
Herein we demonstrate that oncolytic herpes simplex virus-1 (HSV-1) potently activates human peripheral blood mononuclear cells (PBMCs) to lyse leukemic cell lines and primary acute myeloid leukemia samples, but not healthy allogeneic lymphocytes. Intriguingly, we found that UV light-inactivated HSV-1 (UV-HSV-1) is equally effective in promoting PBMC cytolysis of leukemic cells and is 1000- to 10 000-fold more potent at stimulating innate antileukemic responses than UV-inactivated cytomegalovirus, vesicular stomatitis virus, reovirus, or adenovirus. Mechanistically, UV-HSV-1 stimulates PBMC cytolysis of leukemic cells, partly via Toll-like receptor-2/protein kinase C/nuclear factor-κB signaling, and potently stimulates expression of CD69, degranulation, migration, and cytokine production in natural killer (NK) cells, suggesting that surface components of UV-HSV-1 directly activate NK cells. Importantly, UV-HSV-1 synergizes with interleukin-15 (IL-15) and IL-2 in inducing activation and cytolytic activity of NK cells. Additionally, UV-HSV-1 stimulates glycolysis and fatty acid oxidation-dependent oxygen consumption in NK cells, but only glycolysis is required for their enhanced antileukemic activity. Last, we demonstrate that T cell-depleted human PBMCs exposed to UV-HSV-1 provide a survival benefit in a murine xenograft model of human acute myeloid leukemia (AML). Taken together, our results support the preclinical development of UV-HSV-1 as an adjuvant, alone or in combination with IL-15, for allogeneic donor mononuclear cell infusions to treat AML.
Collapse
Affiliation(s)
- Ismael Samudio
- Programa de Investigacion e Innovacion en Leucemia Aguda y Cronica, Bogotá, Colombia; Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX; and
| | - Hila Shaim
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX; and
| | - Elyse Hofs
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Mor Ngom
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Luke Bu
- Brain Research Centre, University of British Columbia, Vancouver, Canada
| | - Guoyu Liu
- Brain Research Centre, University of British Columbia, Vancouver, Canada
| | - Jason T C Lee
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Suzan Imren
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Vivian Lam
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Grace F T Poon
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Maryam Ghaedi
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Fumio Takei
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Keith Humphries
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - William Jia
- Brain Research Centre, University of British Columbia, Vancouver, Canada
| | - Gerald Krystal
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Natural killer (NK) cells are innate lymphoid cells specialized to eliminate malignant cells via direct cytotoxicity and immunoregulatory cytokine production. As such, NK cells are ideal as cellular therapy for cancer patients, and several studies have provided proof of principle that adoptively transferred NK cells can induce remissions in patients with leukemia. A clear understanding of the mechanisms underlying NK cell antitumor responses, including target cell recognition, activation status, and negative regulatory signals will improve NK cellular therapy for cancer patients. RECENT FINDINGS Clinical studies have demonstrated the safety and preliminary efficacy of NK cell adoptive transfer, especially in hematologic malignancies. Various NK cell sources, isolation techniques, activation approaches, and ex-vivo expansion strategies are under investigation. New approaches have been developed and are being tested to optimize NK cell therapy, including ways to better target NK cells to malignant cells, increase their functional competence, facilitate expansion in patients, and limit inhibitory signals or cells. SUMMARY NK cells represent a promising cellular immunotherapy for the treatment of cancer. In addition to adoptive cellular therapy, adjunct treatments that optimize NK cell targeting and function will enhance their potency and broaden their potential use to many cancer types.
Collapse
|
32
|
Rezvani K, Rouce RH. The Application of Natural Killer Cell Immunotherapy for the Treatment of Cancer. Front Immunol 2015; 6:578. [PMID: 26635792 PMCID: PMC4648067 DOI: 10.3389/fimmu.2015.00578] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 10/29/2015] [Indexed: 12/17/2022] Open
Abstract
Natural killer (NK) cells are essential components of the innate immune system and play a critical role in host immunity against cancer. Recent progress in our understanding of NK cell immunobiology has paved the way for novel NK cell-based therapeutic strategies for the treatment of cancer. In this review, we will focus on recent advances in the field of NK cell immunotherapy, including augmentation of antibody-dependent cellular cytotoxicity, manipulation of receptor-mediated activation, and adoptive immunotherapy with ex vivo-expanded, chimeric antigen receptor (CAR)-engineered, or engager-modified NK cells. In contrast to T lymphocytes, donor NK cells do not attack non-hematopoietic tissues, suggesting that an NK-mediated antitumor effect can be achieved in the absence of graft-vs.-host disease. Despite reports of clinical efficacy, a number of factors limit the application of NK cell immunotherapy for the treatment of cancer, such as the failure of infused NK cells to expand and persist in vivo. Therefore, efforts to enhance the therapeutic benefit of NK cell-based immunotherapy by developing strategies to manipulate the NK cell product, host factors, and tumor targets are the subject of intense research. In the preclinical setting, genetic engineering of NK cells to express CARs to redirect their antitumor specificity has shown significant promise. Given the short lifespan and potent cytolytic function of mature NK cells, they are attractive candidate effector cells to express CARs for adoptive immunotherapies. Another innovative approach to redirect NK cytotoxicity towards tumor cells is to create either bispecific or trispecific antibodies, thus augmenting cytotoxicity against tumor-associated antigens. These are exciting times for the study of NK cells; with recent advances in the field of NK cell biology and translational research, it is likely that NK cell immunotherapy will move to the forefront of cancer immunotherapy over the next few years.
Collapse
Affiliation(s)
- Katayoun Rezvani
- Department of Stem Cell Transplantation, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Rayne H Rouce
- Department of Pediatrics, Texas Children's Cancer and Hematology Centers, Baylor College of Medicine , Houston, TX , USA ; Center for Cell and Gene Therapy, Baylor College of Medicine Houston Methodist Hospital and Texas Children's Hospital , Houston, TX , USA
| |
Collapse
|
33
|
Escobedo-Cousin M, Jackson N, Laza-Briviesca R, Ariza-McNaughton L, Luevano M, Derniame S, Querol S, Blundell M, Thrasher A, Soria B, Cooper N, Bonnet D, Madrigal A, Saudemont A. Natural Killer Cells Improve Hematopoietic Stem Cell Engraftment by Increasing Stem Cell Clonogenicity In Vitro and in a Humanized Mouse Model. PLoS One 2015; 10:e0138623. [PMID: 26465138 PMCID: PMC4605799 DOI: 10.1371/journal.pone.0138623] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 08/31/2015] [Indexed: 11/30/2022] Open
Abstract
Cord blood (CB) is increasingly used as a source of hematopoietic stem cells (HSC) for transplantation. Low incidence and severity of graft-versus-host disease (GvHD) and a robust graft-versus-leukemia (GvL) effect are observed following CB transplantation (CBT). However, its main disadvantages are a limited number of HSC per unit, delayed immune reconstitution and a higher incidence of infection. Unmanipulated grafts contain accessory cells that may facilitate HSC engraftment. Therefore, the effects of accessory cells, particularly natural killer (NK) cells, on human CB HSC (CBSC) functions were assessed in vitro and in vivo. CBSC cultured with autologous CB NK cells showed higher levels of CXCR4 expression, a higher migration index and a higher number of colony forming units (CFU) after short-term and long-term cultures. We found that CBSC secreted CXCL9 following interaction with CB NK cells. In addition, recombinant CXCL9 increased CBSC clonogenicity, recapitulating the effect observed of CB NK cells on CBSC. Moreover, the co-infusion of CBSC with CB NK cells led to a higher level of CBSC engraftment in NSG mouse model. The results presented in this work offer the basis for an alternative approach to enhance HSC engraftment that could improve the outcome of CBT.
Collapse
Affiliation(s)
- Michelle Escobedo-Cousin
- University College London, Cancer Institute, London, United Kingdom
- Anthony Nolan Research Institute, Royal Free Campus, London, United Kingdom
| | - Nicola Jackson
- Anthony Nolan Research Institute, Royal Free Campus, London, United Kingdom
| | | | | | - Martha Luevano
- University College London, Cancer Institute, London, United Kingdom
- Anthony Nolan Research Institute, Royal Free Campus, London, United Kingdom
| | - Sophie Derniame
- University College London, Cancer Institute, London, United Kingdom
- Anthony Nolan Research Institute, Royal Free Campus, London, United Kingdom
| | - Sergio Querol
- Programa Concordia Banc de Sang i Teixits, Barcelona, Spain
| | - Michael Blundell
- University College London, Institute of Child Health, London, United Kingdom
| | - Adrian Thrasher
- University College London, Institute of Child Health, London, United Kingdom
| | - Bernat Soria
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Fundación Progreso y Salud, Seville, Spain
- Biomedical Research Network on Diabetes and Related Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Nichola Cooper
- Haematology Department, Hammersmith Hospital, London, United Kingdom
| | - Dominique Bonnet
- Cancer Research UK, London Research Institute, London, United Kingdom
| | - Alejandro Madrigal
- University College London, Cancer Institute, London, United Kingdom
- Anthony Nolan Research Institute, Royal Free Campus, London, United Kingdom
| | - Aurore Saudemont
- University College London, Cancer Institute, London, United Kingdom
- Anthony Nolan Research Institute, Royal Free Campus, London, United Kingdom
| |
Collapse
|
34
|
Cord blood: a promising source of allogeneic natural killer cells for immunotherapy. Cytotherapy 2015; 17:1-2. [PMID: 25527863 DOI: 10.1016/j.jcyt.2014.12.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 12/02/2014] [Indexed: 11/23/2022]
|
35
|
Pittari G, Filippini P, Gentilcore G, Grivel JC, Rutella S. Revving up Natural Killer Cells and Cytokine-Induced Killer Cells Against Hematological Malignancies. Front Immunol 2015; 6:230. [PMID: 26029215 PMCID: PMC4429635 DOI: 10.3389/fimmu.2015.00230] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 04/29/2015] [Indexed: 01/29/2023] Open
Abstract
Natural killer (NK) cells belong to innate immunity and exhibit cytolytic activity against infectious pathogens and tumor cells. NK-cell function is finely tuned by receptors that transduce inhibitory or activating signals, such as killer immunoglobulin-like receptors, NK Group 2 member D (NKG2D), NKG2A/CD94, NKp46, and others, and recognize both foreign and self-antigens expressed by NK-susceptible targets. Recent insights into NK-cell developmental intermediates have translated into a more accurate definition of culture conditions for the in vitro generation and propagation of human NK cells. In this respect, interleukin (IL)-15 and IL-21 are instrumental in driving NK-cell differentiation and maturation, and hold great promise for the design of optimal NK-cell culture protocols. Cytokine-induced killer (CIK) cells possess phenotypic and functional hallmarks of both T cells and NK cells. Similar to T cells, they express CD3 and are expandable in culture, while not requiring functional priming for in vivo activity, like NK cells. CIK cells may offer some advantages over other cell therapy products, including ease of in vitro propagation and no need for exogenous administration of IL-2 for in vivo priming. NK cells and CIK cells can be expanded using a variety of clinical-grade approaches, before their infusion into patients with cancer. Herein, we discuss GMP-compliant strategies to isolate and expand human NK and CIK cells for immunotherapy purposes, focusing on clinical trials of adoptive transfer to patients with hematological malignancies.
Collapse
Affiliation(s)
- Gianfranco Pittari
- Department of Medical Oncology, National Center for Cancer Care and Research, Hamad Medical Corporation , Doha , Qatar
| | - Perla Filippini
- Deep Immunophenotyping Core, Division of Translational Medicine, Sidra Medical and Research Center , Doha , Qatar
| | - Giusy Gentilcore
- Deep Immunophenotyping Core, Division of Translational Medicine, Sidra Medical and Research Center , Doha , Qatar
| | - Jean-Charles Grivel
- Deep Immunophenotyping Core, Division of Translational Medicine, Sidra Medical and Research Center , Doha , Qatar
| | - Sergio Rutella
- Clinical Research Center, Division of Translational Medicine, Sidra Medical and Research Center , Doha , Qatar
| |
Collapse
|