1
|
Balkhi S, Zuccolotto G, Di Spirito A, Rosato A, Mortara L. CAR-NK cell therapy: promise and challenges in solid tumors. Front Immunol 2025; 16:1574742. [PMID: 40260240 PMCID: PMC12009813 DOI: 10.3389/fimmu.2025.1574742] [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: 02/11/2025] [Accepted: 03/17/2025] [Indexed: 04/23/2025] Open
Abstract
Over the past few years, cellular immunotherapy has emerged as a promising treatment for certain hematologic cancers, with various CAR-T therapies now widely used in clinical settings. However, challenges related to the production of autologous cell products and the management of CAR-T cell toxicity highlight the need for new cell therapy options that are universal, safe, and effective. Natural killer (NK) cells, which are part of the innate immune system, offer unique advantages, including the potential for off-the-shelf therapy. A recent first-in-human trial of CD19-CAR-NK infusion in patients with relapsed/refractory lymphoid malignancies demonstrated safety and promising clinical activity. Building on these positive clinical outcomes, current research focuses on enhancing CAR-NK cell potency by increasing their in vivo persistence and addressing functional exhaustion. There is also growing interest in applying the successes seen in hematologic malignancies to solid tumors. This review discusses current trends and emerging concepts in the engineering of next-generation CAR- NK therapies. It will cover the process of constructing CAR-NK cells, potential targets for their manufacturing, and their role in various solid tumors. Additionally, it will examine the mechanisms of action and the research status of CAR-NK therapies in the treatment of solid tumors, along with their advantages, limitations, and future challenges. The insights provided may guide future investigations aimed at optimizing CAR-NK therapy for a broader range of malignancies.
Collapse
Affiliation(s)
- Sahar Balkhi
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Gaia Zuccolotto
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Anna Di Spirito
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| |
Collapse
|
2
|
Nabekura T. Immunological memory in natural killer cells. Int Immunol 2025:dxaf016. [PMID: 40388217 DOI: 10.1093/intimm/dxaf016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/18/2025] [Indexed: 05/20/2025] Open
Abstract
Immune cells are classified into adaptive and innate immune cells. Adaptive immune cells-i.e. T cells and B cells-respond to pathogens in an antigen-specific manner and then provide immunological memory, contributing to long-term host defense against reinfection. In contrast, innate immune cells promptly respond to pathogens, but they are short-lived and have been thought not to contribute to immunological memory. Natural killer (NK) cells are lymphocytes essential for controlling viral infections and cancer. NK cells-which have traditionally been classified as innate immune cells-have recently been revealed as being capable of differentiating into memory NK cells, thus participating in immunological memory, formerly considered to be restricted to adaptive immune cells. Like memory T and B cells, memory NK cells (i) can be long-lived; (ii) display distinct phenotypes from naïve and activated NK cells; (iii) show augmented cellular functions, as compared with naïve NK cells; (iv) have secondary proliferation capacity; and (v) confer an improved host defense when transferred to naïve recipients. Therefore, at least in a broad sense, they fulfill the definition of immunological memory. In this article, I provide an overview of NK cell memory and recent research trends regarding this phenomenon.
Collapse
Affiliation(s)
- Tsukasa Nabekura
- Division of Immune Response, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi 464-8681, Japan
- Division of Cellular Oncology, Department of Cancer Diagnostics and Therapeutics, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya, Aichi 466-8550, Japan
| |
Collapse
|
3
|
Li S, Wang K, Wu J, Zhu Y. The immunosenescence clock: A new method for evaluating biological age and predicting mortality risk. Ageing Res Rev 2025; 104:102653. [PMID: 39746402 DOI: 10.1016/j.arr.2024.102653] [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: 09/25/2024] [Revised: 12/12/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Precisely assessing an individual's immune age is critical for developing targeted aging interventions. Although traditional methods for evaluating biological age, such as the use of cellular senescence markers and physiological indicators, have been widely applied, these methods inherently struggle to capture the full complexity of biological aging. We propose the concept of an 'immunosenescence clock' that evaluates immune system changes on the basis of changes in immune cell abundance and omics data (including transcriptome and proteome data), providing a complementary indicator for understanding age-related physiological transformations. Rather than claiming to definitively measure biological age, this approach can be divided into a biological age prediction clock and a mortality prediction clock. The main function of the biological age prediction clock is to reflect the physiological state through the transcriptome data of peripheral blood mononuclear cells (PBMCs), whereas the mortality prediction clock emphasizes the ability to identify people at high risk of mortality and disease. We hereby present nearly all of the immunosenescence clocks developed to date, as well as their functional differences. Critically, we explicitly acknowledge that no single diagnostic test can exhaustively capture the intricate changes associated with biological aging. Furthermore, as these biological functions are based on the acceleration or delay of immunosenescence, we also summarize the factors that accelerate immunosenescence and the methods for delaying it. A deep understanding of the regulatory mechanisms of immunosenescence can help establish more accurate immune-age models, providing support for personalized longevity interventions and improving quality of life in old age.
Collapse
Affiliation(s)
- Shuyu Li
- Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ke Wang
- Department of Breast Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jingni Wu
- Department of International Healthcare Center and General Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yongliang Zhu
- Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
Meggyes M, Nagy DU, Mezosi L, Polgar B, Szereday L. CD8+ and CD8- NK Cells and Immune Checkpoint Networks in Peripheral Blood During Healthy Pregnancy. Int J Mol Sci 2025; 26:428. [PMID: 39796279 PMCID: PMC11720283 DOI: 10.3390/ijms26010428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Pregnancy involves significant immunological changes to support fetal development while protecting the mother from infections. A growing body of evidence supports the importance of immune checkpoint pathways, especially at the maternal-fetal interface, although limited information is available about the peripheral expression of these molecules by CD8+ and CD8- NK cell subsets during the trimesters of pregnancy. Understanding the dynamics of these immune cells and their checkpoint pathways is crucial for elucidating their roles in pregnancy maintenance and potential complications. This study aims to investigate the peripheral expression and functional characteristics of CD8+ and CD8- NK cell subsets throughout pregnancy, providing insights into their contributions to maternal and fetal health. A total of 34 healthy women were enrolled from the first, 30 from the second and 40 from the third trimester of pregnancy. At the same time, 35 healthy age-matched non-pregnant women formed the control group. From peripheral blood, mononuclear cells were separated and stored at -80 °C. CD8+ and CD8- NK cell subsets were analyzed from freshly thawed samples, and surface and intracellular staining was performed using flow cytometric analyses. The proportions of CD56+ NK cells in peripheral blood were similar across groups. While CD8- NKdim cells increased significantly in all trimesters compared to non-pregnant controls, CD8+ NKdim cells showed no significant changes. CD8- NKbright cells had higher frequencies throughout pregnancy, whereas CD8+ NKbright cells significantly increased only in the first and second trimesters. The expression levels of immune checkpoint molecules, such as PD-1 and PD-L1, and cytotoxic-activity-related molecules were stable, with notable perforin and granzyme B increases in CD8- NKbright cells throughout pregnancy. Our study shows that peripheral NK cell populations, especially CD8- subsets, are predominant during pregnancy. This shift suggests a crucial role for CD8- NK cells in balancing maternal immune tolerance and surveillance. The stable expression of immune checkpoint molecules indicates that other regulatory mechanisms may be at work. These findings enhance our understanding of peripheral immune dynamics in pregnancy and suggest that targeting CD8- NKbright cell functions could help manage pregnancy-related immune complications. This research elucidates the stable distribution and functional characteristics of peripheral NK cells during pregnancy, with CD8- subsets being more prevalent. The increased activity of CD8- NKbright cells suggests their critical role in maintaining immune surveillance. Our findings provide a basis for future studies to uncover the mechanisms regulating NK cell function in pregnancy, potentially leading to new treatments for immune-related pregnancy complications.
Collapse
Affiliation(s)
- Matyas Meggyes
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 12 Szigeti Street, 7624 Pecs, Hungary
- Janos Szentagothai Research Centre, 20 Ifjusag Street, 7624 Pecs, Hungary
| | - David U. Nagy
- Institute of Geobotany/Plant Ecology, Martin-Luther-University, Große Steinstraße 79/80, D-06108 Halle, Germany
| | - Livia Mezosi
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 12 Szigeti Street, 7624 Pecs, Hungary
| | - Beata Polgar
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 12 Szigeti Street, 7624 Pecs, Hungary
| | - Laszlo Szereday
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 12 Szigeti Street, 7624 Pecs, Hungary
- Janos Szentagothai Research Centre, 20 Ifjusag Street, 7624 Pecs, Hungary
| |
Collapse
|
5
|
Rados M, Landegger A, Schmutzler L, Rabidou K, Taschner-Mandl S, Fetahu IS. Natural killer cells in neuroblastoma: immunological insights and therapeutic perspectives. Cancer Metastasis Rev 2024; 43:1401-1417. [PMID: 39294470 PMCID: PMC11554946 DOI: 10.1007/s10555-024-10212-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 09/10/2024] [Indexed: 09/20/2024]
Abstract
Natural killer (NK) cells have multifaceted roles within the complex tumor milieu. They are pivotal components of innate immunity and shape the dynamic landscape of tumor-immune cell interactions, and thus can be leveraged for use in therapeutic interventions. NK-based immunotherapies have had remarkable success in hematological malignancies, but these therapies are met with many challenges in solid tumors, including neuroblastoma (NB), a childhood tumor arising from the sympathetic nervous system. With a focus on NB, this review outlines the mechanisms employed by NK cells to recognize and eliminate malignant cells, delving into the dynamic relationship between ligand-receptor interactions, cytokines, and other molecules that facilitate the cross talk between NK and NB cells. We discuss the immunomodulatory functions of NK cells and the mechanisms that contribute to loss of this immunosurveillance in NB, with a focus on how this dynamic has been utilized in recent immunotherapy advancements for NB.
Collapse
Affiliation(s)
- Magdalena Rados
- St. Anna Children's Cancer Research Institute, Vienna, Austria
| | | | - Lukas Schmutzler
- Department of Otorhinolaryngology - Head and Neck Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Kimberlie Rabidou
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, USA
| | | | - Irfete S Fetahu
- Department of Neurology, Division of Neuropathology and Neurochemistry, Medical University of Vienna, Vienna, Austria.
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
6
|
Shi J, Shen L, Xiao Y, Wan C, Wang B, Zhou P, Zhang J, Han W, Hu R, Yu F, Wang H. Identification and validation of diagnostic biomarkers and immune cell abundance characteristics in Staphylococcus aureus bloodstream infection by integrative bioinformatics analysis. Front Immunol 2024; 15:1450782. [PMID: 39654884 PMCID: PMC11626409 DOI: 10.3389/fimmu.2024.1450782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/01/2024] [Indexed: 12/12/2024] Open
Abstract
Staphylococcus aureus (S. aureus) is an opportunistic pathogen that could cause life-threatening bloodstream infections. The objective of this study was to identify potential diagnostic biomarkers of S. aureus bloodstream infection. Gene expression dataset GSE33341 was optimized as the discovery dataset, which contained samples from human and mice. GSE65088 dataset was utilized as a validation dataset. First, after overlapping the differentially expressed genes (DEGs) in S. aureus infection samples from GSE33341-human and GSE33341-mice samples, we detected 63 overlapping genes. Subsequently, the hub genes including DRAM1, PSTPIP2, and UPP1 were identified via three machine-learning algorithms: random forest, support vector machine-recursive feature elimination, and least absolute shrinkage and selection operator. Additionally, the receiver operating characteristic curve was leveraged to verify the efficacy of the hub genes. DRAM1 (AUC=1), PSTPIP2 (AUC=1), and UPP1 (AUC=1) were investigated and demonstrated significant expression differences (all P < 0.05) and diagnostic efficacy in the training and validation datasets. Furthermore, the relationship between the diagnostic markers and the abundance of immune cells was assessed using cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT). These three diagnostic indicators also correlated with multiple immune cells to varying degrees. The expression of DRAM1 was significantly positively correlated with B cell naive and mast cell activation, and negatively correlated with NK cells and CD4/CD8+ T cells. The expression of PSTPIP2 was significantly positively correlated with macrophage M0, macrophage M1, B cell naive, and dendritic cell activation, while the expression of PSTPIP2 was negatively correlated with NK cells and CD4/CD8+ T cells. Significant negative correlations between UPP1 expression and T cell CD4 memory rest and neutrophils were also observed. Finally, we established a mouse model of S. aureus bloodstream infection and collected the blood samples for RNA-Seq analysis and RT-qPCR experiments. The analysis results in RNA-Seq and RT-qPCR experiments further confirmed the significant expression differences (all P < 0.05) of these three genes. Overall, three candidate hub genes (DRAM1, PSTPIP2, and UPP1) were identified initially for S. aureus bloodstream infection diagnosis. Our study could provide potential diagnostic biomarkers for S. aureus bloodstream infection patients.
Collapse
Affiliation(s)
- Junhong Shi
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Li Shen
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanghua Xiao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Cailing Wan
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bingjie Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Peiyao Zhou
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiao Zhang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weihua Han
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Rongrong Hu
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Fangyou Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hongxiu Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
7
|
Newman MJ. Invention and characterization of a systemically administered, attenuated and killed bacteria-based multiple immune receptor agonist for anti-tumor immunotherapy. Front Immunol 2024; 15:1462221. [PMID: 39606250 PMCID: PMC11599860 DOI: 10.3389/fimmu.2024.1462221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
Activation of immune receptors, such as Toll-like (TLR), NOD-like (NLR) and Stimulator of Interferon Genes (STING) is critical for efficient innate and adaptive immunity. Gram-negative bacteria (G-NB) contain multiple TLR, NOD and STING agonists. Potential utility of G-NB for cancer immunotherapy is supported by observations of tumor regression in the setting of infection and Coley's Toxins. Coley reported that intravenous (i.v.) administration was likely most effective but produced uncontrollable toxicity. The discovery of TLRs and their agonists, particularly the potent TLR4 agonist lipopolysaccharide (LPS)-endotoxin, comprising ~75% of the outer membrane of G-NB, suggests that LPS may be both a critical active ingredient and responsible for dose-limiting i.v. toxicity of G-NB. This communication reports the production of killed, stabilized, intact bacteria products from non-pathogenic G-NB with ~96% reduction of LPS-endotoxin activity. One resulting product candidate, Decoy10, was resistant to standard methods of cell disruption and contained TLR2,4,8,9, NOD2 and STING agonist activity. Decoy10 also exhibited reduced i.v. toxicity in mice and rabbits, and a largely uncompromised ability to induce cytokine and chemokine secretion by human immune cells in vitro, all relative to unprocessed, parental bacterial cells. Decoy10 and a closely related product, Decoy20, produced single agent anti-tumor activity or combination-mediated durable regression of established subcutaneous, metastatic or orthotopic colorectal, hepatocellular (HCC), pancreatic, and non-Hodgkin's lymphoma (NHL) tumors in mice, with induction of both innate and adaptive immunological memory (syngeneic and human tumor xenograft models). Decoy bacteria combination-mediated regressions were observed with a low-dose, oral non-steroidal anti-inflammatory drug (NSAID), anti-PD-1 checkpoint therapy, low-dose cyclophosphamide (LDC), and/or a targeted antibody (rituximab). Efficient tumor eradication was associated with plasma expression of 15-23 cytokines and chemokines, broad induction of cytokine, chemokine, innate and adaptive immune pathway genes in tumors, cold to hot tumor inflammation signature transition, and required NK, CD4+ and CD8+ T cells, collectively demonstrating a role for both innate and adaptive immune activation in the anti-tumor immune response.
Collapse
|
8
|
Boum Y, Matchim L, Guimsop DK, Buri BD, Bebell LM, Jaudel YSF, Njuwa FKG, Danirla DB, Youm E, Ntone R, Tchame CR, Tchiasso D, Essaka R, Eyong JB, Ngosso A, Nanda H, Fondze NR, Ndifon MN, Eteki L, Ghislain YFC, Messi BYE, Moustapha H, Hamdja M, Essomba RG, Mandeng N, Modeste TAK, Bisseck ACZK, Eyangoh SI, Njouom R, Okomo MC, Esso L, Emilienne E, Mballa GAE. High immunity and low mortality after Omicron and mass event in Cameroon despite low vaccination. J Public Health Afr 2024; 15:649. [PMID: 39649435 PMCID: PMC11622603 DOI: 10.4102/jphia.v15i1.649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 09/07/2024] [Indexed: 12/10/2024] Open
Abstract
Background Little is known about the evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunity in African communities. Aim We evaluated changes in anti-SARS-CoV-2 antibodies, mortality and vaccination status in Cameroon between August 2021 and September 2022 to begin describing the evolution of the pandemic in Africa. Setting The study was conducted across Cameroon's 10 regional capitals, between 2021 and 2022 as the country hosted a mass gathering. Methods We conducted a cross-sectional population-based survey in 2022, including SARS-CoV-2 seroprevalence testing and retrospective mortality estimation using two-stage cluster sampling. We estimated and compared seroprevalence and crude mortality rates (CMR) to a survey conducted in 2021 using the same methodology. Results We performed serologic testing on 8400 individuals and collected mortality data from 22 314 individuals. Approximately 5% in each survey reported SARS-CoV-2-vaccination. Rapid diagnostic test-based seroprevalence increased from 11.2% (95% confidence interval [CI]: 10-12.5) to 59.8% (95% CI: 58.3-61.2) between 2021 and 2022, despite no increase in the proportion vaccinated. The CMR decreased from 0.17 to 0.06 deaths per 10 000 persons per day between 2021 and 2022. In 2022, no deaths were reportedly attributable to COVID-19 as compared to 17 deaths in 2021. Conclusion Over a 12-month period encompassing two waves of omicron variant SARS-CoV-2 and a mass gathering, SARS-CoV-2 seropositivity in Cameroon approached 60%, and deaths declined despite low vaccination coverage. Contribution This study challenges the assumption that high immunisation coverage is the sole determinant of epidemic control in the African context and encourages policymakers to increasingly rely on local research when designing response strategies for more effective outbreak management.
Collapse
Affiliation(s)
- Yap Boum
- Public Health Emergency Operation Center, Ministry of Public Health, Yaoundé, Cameroon
- Epicentre, Yaoundé, Cameroon
- Faculty of Biomedical Medicine and Science, University of Yaoundé I, Yaoundé, Cameroon
| | | | - Dominique K Guimsop
- Department for the Control of Disease, Epidemics and Pandemics, Ministry of Public Health, Yaoundé, Cameroon
- Western Africa Regional Coordination Center, Africa Centers for Disease Control, Abuja, Nigeria
| | | | - Lisa M Bebell
- Department of Medicine, Massachusetts General Hospital, and Harvard Medical School, Boston, Massachusetts, United States of America
| | | | | | | | | | | | - Claudric Roosevelt Tchame
- Department for the Control of Disease, Epidemics and Pandemics, Ministry of Public Health, Yaoundé, Cameroon
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Nadia Mandeng
- Public Health Emergency Operation Center, Ministry of Public Health, Yaoundé, Cameroon
| | | | | | | | | | | | - Linda Esso
- Public Health Emergency Operation Center, Ministry of Public Health, Yaoundé, Cameroon
- Department for the Control of Disease, Epidemics and Pandemics, Yaoundé, Cameroon
| | - Epee Emilienne
- Public Health Emergency Operation Center, Ministry of Public Health, Yaoundé, Cameroon
- Department for the Control of Disease, Epidemics and Pandemics, Ministry of Public Health, Yaoundé, Cameroon
| | - Georges-Alain Etoundi Mballa
- Public Health Emergency Operation Center, Ministry of Public Health, Yaoundé, Cameroon
- Department for the Control of Disease, Epidemics and Pandemics, Ministry of Public Health, Yaoundé, Cameroon
| |
Collapse
|
9
|
Tahri S, Piccinelli S, Su NK, Lampe L, Dong H, Vergara Cadavid J, Boiarsky R, Papazian N, Lightbody ED, Cao A, Alberge JB, Ferrari de Andrade L, Rahmat M, Shen Y, Blanco Fernández L, Zabaleta A, Günther A, Getz G, Sonneveld P, Cupedo T, Wucherpfennig KW, Ghobrial IM, Romee R. Inhibition of MICA and MICB shedding enhances memory-like NK-cell-mediated cytotoxicity against multiple myeloma. Blood Adv 2024; 8:5365-5370. [PMID: 39106432 PMCID: PMC11568787 DOI: 10.1182/bloodadvances.2023010869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/14/2024] [Accepted: 07/02/2024] [Indexed: 08/09/2024] Open
Affiliation(s)
- Sabrin Tahri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medical Oncology, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA
- Center for Prevention of Progression of Blood Cancers, Dana-Farber Cancer Institute, Boston, MA
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sara Piccinelli
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medical Oncology, Harvard Medical School, Boston, MA
| | - Nang Kham Su
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medical Oncology, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA
- Center for Prevention of Progression of Blood Cancers, Dana-Farber Cancer Institute, Boston, MA
| | - Luisa Lampe
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medical Oncology, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA
- Center for Prevention of Progression of Blood Cancers, Dana-Farber Cancer Institute, Boston, MA
- Medical Department, Christian-Albrechts University, Kiel, Germany
| | - Han Dong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Rebecca Boiarsky
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA
| | - Natalie Papazian
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Elizabeth D. Lightbody
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medical Oncology, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA
- Center for Prevention of Progression of Blood Cancers, Dana-Farber Cancer Institute, Boston, MA
| | - Amanda Cao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medical Oncology, Harvard Medical School, Boston, MA
| | - Jean-Baptiste Alberge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medical Oncology, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA
- Center for Prevention of Progression of Blood Cancers, Dana-Farber Cancer Institute, Boston, MA
| | - Lucas Ferrari de Andrade
- Precision Immunology Institute, Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai Hospital, New York, NY
| | - Mahshid Rahmat
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medical Oncology, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA
- Center for Prevention of Progression of Blood Cancers, Dana-Farber Cancer Institute, Boston, MA
| | - Yujia Shen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Cancer Science Institute of Singapore, National University of Singapore Cancer Science, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Laura Blanco Fernández
- Clínica Universidad de Navarra, Center for Applied Medical Research, CIBERONC, IDISNA, Pamplona, Spain
| | - Aintzane Zabaleta
- Clínica Universidad de Navarra, Center for Applied Medical Research, CIBERONC, IDISNA, Pamplona, Spain
| | - Andreas Günther
- Medical Department, Christian-Albrechts University, Kiel, Germany
| | - Gad Getz
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA
- Krantz Family Center for Cancer Research and Deptartment of Pathology, Massachusetts General Hospital, Boston, MA
- Department of Pathology, Harvard Medical School, Boston, MA
| | - Pieter Sonneveld
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Tom Cupedo
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Kai W. Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Irene M. Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medical Oncology, Harvard Medical School, Boston, MA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA
- Center for Prevention of Progression of Blood Cancers, Dana-Farber Cancer Institute, Boston, MA
| | - Rizwan Romee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medical Oncology, Harvard Medical School, Boston, MA
| |
Collapse
|
10
|
Naidoo KK, Altfeld M. The Role of Natural Killer Cells and Their Metabolism in HIV-1 Infection. Viruses 2024; 16:1584. [PMID: 39459918 PMCID: PMC11512232 DOI: 10.3390/v16101584] [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: 08/02/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
Natural killer (NK) cells are multifaceted innate effector cells that critically influence antiviral immunity, and several protective NK cell features that modulate HIV-1 acquisition and viral control have been described. Chronic HIV-1 infection leads to NK cell impairment that has been associated with metabolic dysregulations. Therapeutic approaches targeting cellular immune metabolism represent potential novel interventions to reverse defective NK cell function in people living with HIV.
Collapse
Affiliation(s)
- Kewreshini K. Naidoo
- Department of Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany
| | - Marcus Altfeld
- Department of Virus Immunology, Leibniz Institute of Virology, 20251 Hamburg, Germany
- German Center for Infection Disease (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, 20251 Hamburg, Germany
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
11
|
Hoffmann MH, Kirchner H, Krönke G, Riemekasten G, Bonelli M. Inflammatory tissue priming: novel insights and therapeutic opportunities for inflammatory rheumatic diseases. Ann Rheum Dis 2024; 83:1233-1253. [PMID: 38702177 DOI: 10.1136/ard-2023-224092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024]
Abstract
Due to optimised treatment strategies and the availability of new therapies during the last decades, formerly devastating chronic inflammatory diseases such as rheumatoid arthritis or systemic sclerosis (SSc) have become less menacing. However, in many patients, even state-of-the-art treatment cannot induce remission. Moreover, the risk for flares strongly increases once anti-inflammatory therapy is tapered or withdrawn, suggesting that underlying pathological processes remain active even in the absence of overt inflammation. It has become evident that tissues have the ability to remember past encounters with pathogens, wounds and other irritants, and to react more strongly and/or persistently to the next occurrence. This priming of the tissue bears a paramount role in defence from microbes, but on the other hand drives inflammatory pathologies (the Dr Jekyll and Mr Hyde aspect of tissue adaptation). Emerging evidence suggests that long-lived tissue-resident cells, such as fibroblasts, macrophages, long-lived plasma cells and tissue-resident memory T cells, determine inflammatory tissue priming in an interplay with infiltrating immune cells of lymphoid and myeloid origin, and with systemically acting factors such as cytokines, extracellular vesicles and antibodies. Here, we review the current state of science on inflammatory tissue priming, focusing on tissue-resident and tissue-occupying cells in arthritis and SSc, and reflect on the most promising treatment options targeting the maladapted tissue response during these diseases.
Collapse
Affiliation(s)
| | - Henriette Kirchner
- Institute for Human Genetics, Epigenetics and Metabolism Lab, University of Lübeck, Lübeck, Germany
| | - Gerhard Krönke
- Department of Rheumatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Michael Bonelli
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| |
Collapse
|
12
|
Bao K, Gu X, Song Y, Zhou Y, Chen Y, Yu X, Yuan W, Shi L, Zheng J, Hong M. TCF-1 and TOX regulate the memory formation of intestinal group 2 innate lymphoid cells in asthma. Nat Commun 2024; 15:7850. [PMID: 39245681 PMCID: PMC11381517 DOI: 10.1038/s41467-024-52252-2] [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: 12/18/2023] [Accepted: 08/31/2024] [Indexed: 09/10/2024] Open
Abstract
Immune memory has been expanded to group 2 innate lymphoid cells (ILC2s), but the cellular and molecular bases remain incompletely understood. Based on house dust mite (HDM)-induced mice asthma models and human samples, we applied flow cytometry, parabiosis, in vivo imaging and adoptive transplantation to confirm the persistence, migration and function of CD45+lineage-CD90.2+NK1.1-NKp46-ST2-KLRG1+IL-17RB+ memory-like ILC2s (ml-ILC2s). Regulated by CCR9/CCL25 and S1P signaling, ml-ILC2s reside in the lamina propria of small intestines (siLP) in asthma remission, and subsequently move to airway upon re-encountering antigens or alarmins. Furthermore, ml-ILC2s possess properties of longevity, potential of rapid proliferation and producing IL-13, and display transcriptional characteristics with up-regulation of Tox and Tcf-7. ml-ILC2s transplantation restore the asthmatic changes abrogated by Tox and Tcf7 knockdown. Our data identify siLP ml-ILC2s as a memory-like subset, which promotes asthma relapse. Targeting TCF-1 and TOX might be promising for preventing asthma recurrence.
Collapse
Affiliation(s)
- Kaifan Bao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Department of Immunology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xiaoqun Gu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yajun Song
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yijing Zhou
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yanyan Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xi Yu
- Nanjing Haikerui Pharmaceutical Technology Co., LTD, Nanjing, 210023, China
| | - Weiyuan Yuan
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Liyun Shi
- Department of Immunology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jie Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Min Hong
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
13
|
Wang D, Dou L, Sui L, Xue Y, Xu S. Natural killer cells in cancer immunotherapy. MedComm (Beijing) 2024; 5:e626. [PMID: 38882209 PMCID: PMC11179524 DOI: 10.1002/mco2.626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 06/18/2024] Open
Abstract
Natural killer (NK) cells, as innate lymphocytes, possess cytotoxic capabilities and engage target cells through a repertoire of activating and inhibitory receptors. Particularly, natural killer group 2, member D (NKG2D) receptor on NK cells recognizes stress-induced ligands-the MHC class I chain-related molecules A and B (MICA/B) presented on tumor cells and is key to trigger the cytolytic response of NK cells. However, tumors have developed sophisticated strategies to evade NK cell surveillance, which lead to failure of tumor immunotherapy. In this paper, we summarized these immune escaping strategies, including the downregulation of ligands for activating receptors, upregulation of ligands for inhibitory receptors, secretion of immunosuppressive compounds, and the development of apoptosis resistance. Then, we focus on recent advancements in NK cell immune therapies, which include engaging activating NK cell receptors, upregulating NKG2D ligand MICA/B expression, blocking inhibitory NK cell receptors, adoptive NK cell therapy, chimeric antigen receptor (CAR)-engineered NK cells (CAR-NK), and NKG2D CAR-T cells, especially several vaccines targeting MICA/B. This review will inspire the research in NK cell biology in tumor and provide significant hope for improving cancer treatment outcomes by harnessing the potent cytotoxic activity of NK cells.
Collapse
Affiliation(s)
- DanRu Wang
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - LingYun Dou
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - LiHao Sui
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - Yiquan Xue
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - Sheng Xu
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
- Shanghai Institute of Stem Cell Research and Clinical Translation Dongfang Hospital Shanghai China
| |
Collapse
|
14
|
Lam PY, Souza-Fonseca-Guimaraes F. Highlight of 2023: Unlocking the therapeutic potential of natural killer cells-advances in adaptive functions, cellular engineering and immunotherapy. Immunol Cell Biol 2024; 102:444-447. [PMID: 38693888 DOI: 10.1111/imcb.12769] [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] [Indexed: 05/03/2024]
Abstract
Natural killer (NK) cells possess potent cytotoxicity against infected and cancerous cells and hold promise in the development of new immunotherapies. This article for the Highlights of 2023 Series focuses on current advances in NK cell biology in cancerous and infectious settings and highlights opportunities for therapeutic interventions, including engineered NK cell therapies and advancements in feeder cell technologies.
Collapse
Affiliation(s)
- Pui Yeng Lam
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, Australia
| | | |
Collapse
|
15
|
Ding Y, Lavaert M, Grassmann S, Band VI, Chi L, Das A, Das S, Harly C, Shissler SC, Malin J, Peng D, Zhao Y, Zhu J, Belkaid Y, Sun JC, Bhandoola A. Distinct developmental pathways generate functionally distinct populations of natural killer cells. Nat Immunol 2024; 25:1183-1192. [PMID: 38872000 DOI: 10.1038/s41590-024-01865-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 05/08/2024] [Indexed: 06/15/2024]
Abstract
Natural killer (NK) cells function by eliminating virus-infected or tumor cells. Here we identified an NK-lineage-biased progenitor population, referred to as early NK progenitors (ENKPs), which developed into NK cells independently of common precursors for innate lymphoid cells (ILCPs). ENKP-derived NK cells (ENKP_NK cells) and ILCP-derived NK cells (ILCP_NK cells) were transcriptionally different. We devised combinations of surface markers that identified highly enriched ENKP_NK and ILCP_NK cell populations in wild-type mice. Furthermore, Ly49H+ NK cells that responded to mouse cytomegalovirus infection primarily developed from ENKPs, whereas ILCP_NK cells were better IFNγ producers after infection with Salmonella and herpes simplex virus. Human CD56dim and CD56bright NK cells were transcriptionally similar to ENKP_NK cells and ILCP_NK cells, respectively. Our findings establish the existence of two pathways of NK cell development that generate functionally distinct NK cell subsets in mice and further suggest these pathways may be conserved in humans.
Collapse
Affiliation(s)
- Yi Ding
- T Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marieke Lavaert
- T Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Simon Grassmann
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Victor I Band
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Liang Chi
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Arundhoti Das
- T Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sumit Das
- T Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christelle Harly
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, Nantes, France
- LabEx IGO "Immunotherapy, Graft Oncology", Nantes, France
| | - Susannah C Shissler
- T Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Justin Malin
- T Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dingkang Peng
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yongge Zhao
- T Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Joseph C Sun
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Avinash Bhandoola
- T Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
16
|
Gui Z, Al Moussawy M, Sanders SM, Abou-Daya KI. Innate Allorecognition in Transplantation: Ancient Mechanisms With Modern Impact. Transplantation 2024; 108:1524-1531. [PMID: 38049941 PMCID: PMC11188633 DOI: 10.1097/tp.0000000000004847] [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: 08/14/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 12/06/2023]
Abstract
Through the effective targeting of the adaptive immune system, solid organ transplantation became a life-saving therapy for organ failure. However, beyond 1 y of transplantation, there is little improvement in transplant outcomes. The adaptive immune response requires the activation of the innate immune system. There are no modalities for the specific targeting of the innate immune system involvement in transplant rejection. However, the recent discovery of innate allorecognition and innate immune memory presents novel targets in transplantation that will increase our understanding of organ rejection and might aid in improving transplant outcomes. In this review, we look at the latest developments in the study of innate allorecognition and innate immune memory in transplantation.
Collapse
Affiliation(s)
- Zeping Gui
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA
| | - Mouhamad Al Moussawy
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA
| | - Steven M. Sanders
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA
| | - Khodor I. Abou-Daya
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
17
|
Qi Y, Li Y, Wang H, Wang A, Liu X, Liang Z, Gao Y, Wei L. Natural killer cell-related anti-tumour adoptive cell immunotherapy. J Cell Mol Med 2024; 28:e18362. [PMID: 38837666 PMCID: PMC11151221 DOI: 10.1111/jcmm.18362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/04/2024] [Accepted: 04/16/2024] [Indexed: 06/07/2024] Open
Abstract
Chimeric antigen receptor- (CAR-)modified T cells have been successfully used to treat blood cancer. With the improved research on anti-tumour adoptive cell therapy, researchers have focused on immune cells other than T lymphocytes. Natural killer (NK) cells have received widespread attention as barriers to natural immunity. Compared to T lymphocyte-related adoptive cell therapy, the use of NK cells to treat tumours does not cause graft-versus-host disease, significantly improving immunity. Moreover, NK cells have more sources than T cells, and the related modified cells are less expensive. NK cells function through several pathways in anti-tumour mechanisms. Currently, many anti-tumour clinical trials have used NK cell-related adoptive cell therapies. In this review, we have summarized the recent progress in NK cell-related adoptive cellular immunotherapy for tumour treatment and propose the current challenges faced by CAR-NK cell therapy.
Collapse
Affiliation(s)
- Yuwen Qi
- Department of Gynecological OncologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of Tumor Biological BehaviorsWuhanChina
- Hubei Cancer Clinical Study CenterWuhanChina
| | - Ying Li
- Physical Examination CenterRenmin Hospital of Wuhan UniversityWuhanChina
| | - Hua Wang
- Department of Gynecological OncologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of Tumor Biological BehaviorsWuhanChina
- Hubei Cancer Clinical Study CenterWuhanChina
| | - Anjin Wang
- Department of Gynecological OncologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of Tumor Biological BehaviorsWuhanChina
- Hubei Cancer Clinical Study CenterWuhanChina
| | - Xuelian Liu
- Department of Gynecological OncologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of Tumor Biological BehaviorsWuhanChina
- Hubei Cancer Clinical Study CenterWuhanChina
| | - Ziyan Liang
- Department of Gynecological OncologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of Tumor Biological BehaviorsWuhanChina
- Hubei Cancer Clinical Study CenterWuhanChina
| | - Yang Gao
- Department of Gynecological OncologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of Tumor Biological BehaviorsWuhanChina
- Hubei Cancer Clinical Study CenterWuhanChina
| | - Liqing Wei
- Wuhan Wuchang HospitalWuhan University of Science and TechnologyWuhanChina
| |
Collapse
|
18
|
Tepale-Segura A, Gajón JA, Muñoz-Cruz S, Castro-Escamilla O, Bonifaz LC. The cholera toxin B subunit induces trained immunity in dendritic cells and promotes CD8 T cell antitumor immunity. Front Immunol 2024; 15:1362289. [PMID: 38812523 PMCID: PMC11133619 DOI: 10.3389/fimmu.2024.1362289] [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: 12/28/2023] [Accepted: 04/30/2024] [Indexed: 05/31/2024] Open
Abstract
Introduction Innate immune training is a metabolic, functional, and epigenetic long-term reprogramming of innate cells triggered by different stimuli. This imprinting also reaches hematopoietic precursors in the bone marrow to sustain a memory-like phenotype. Dendritic cells (DCs) can exhibit memory-like responses, enhanced upon subsequent exposure to a pathogen; however, whether this imprinting is lineage and stimulus-restricted is still being determined. Nevertheless, the functional consequences of DCs training on the adaptive and protective immune response against non-infectious diseases remain unresolved. Methods We evaluated the effect of the nontoxic cholera B subunit (CTB), LPS and LTA in the induction of trained immunity in murine DCs revealed by TNFa and LDH expression, through confocal microscopy. Additionally, we obtained bone marrow DCs (BMDCs) from mice treated with CTB, LPS, and LTA and evaluated training features in DCs and their antigen-presenting cell capability using multiparametric cytometry. Finally, we design an experimental melanoma mouse model to demonstrate protection induced by CTB-trained DCs in vivo. Results CTB-trained DCs exhibit increased expression of TNFa, and metabolic reprogramming indicated by LDH expression. Moreover, CTB training has an imprint on DC precursors, increasing the number and antigen-presenting function in BMDCs. We found that training by CTB stimulates the recruitment of DC precursors and DCs infiltration at the skin and lymph nodes. Interestingly, training-induced by CTB promotes a highly co-stimulatory phenotype in tumor-infiltrating DCs (CD86+) and a heightened functionality of exhausted CD8 T cells (Ki67+, GZMB+), which were associated with a protective response against melanoma challenge in vivo. Conclusion Our work indicates that CTB can induce innate immune training on DCs, which turns into an efficient adaptive immune response in the melanoma model and might be a potential immunotherapeutic approach for tumor growth control.
Collapse
Affiliation(s)
- Araceli Tepale-Segura
- Unidad de Investigación Médica en Inmunoquímica, Unidad Médica de Alta Especialidad (UMAE) Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Departamento de Inmunología, Mexico City, Mexico
| | - Julián A. Gajón
- Unidad de Investigación Médica en Inmunoquímica, Unidad Médica de Alta Especialidad (UMAE) Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
- Posgrado en Ciencias Bioquímicas, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Samira Muñoz-Cruz
- Unidad de Investigación Médica en Inmunoquímica, Unidad Médica de Alta Especialidad (UMAE) Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Octavio Castro-Escamilla
- División de Investigación Clínica, Coordinación de Investigación en Salud, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Laura C. Bonifaz
- Unidad de Investigación Médica en Inmunoquímica, Unidad Médica de Alta Especialidad (UMAE) Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
- Coordinación de Investigación en Salud, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| |
Collapse
|
19
|
Hermans L, O’Sullivan TE. No time to die: Epigenetic regulation of natural killer cell survival. Immunol Rev 2024; 323:61-79. [PMID: 38426615 PMCID: PMC11102341 DOI: 10.1111/imr.13314] [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] [Indexed: 03/02/2024]
Abstract
NK cells are short-lived innate lymphocytes that can mediate antigen-independent responses to infection and cancer. However, studies from the past two decades have shown that NK cells can acquire transcriptional and epigenetic modifications during inflammation that result in increased survival and lifespan. These findings blur the lines between the innate and adaptive arms of the immune system, and suggest that the homeostatic mechanisms that govern the persistence of innate immune cells are malleable. Indeed, recent studies have shown that NK cells undergo continuous and strictly regulated adaptations controlling their survival during development, tissue residency, and following inflammation. In this review, we summarize our current understanding of the critical factors regulating NK cell survival throughout their lifespan, with a specific emphasis on the epigenetic modifications that regulate the survival of NK cells in various contexts. A precise understanding of the molecular mechanisms that govern NK cell survival will be important to enhance therapies for cancer and infectious diseases.
Collapse
Affiliation(s)
- Leen Hermans
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
| | - Timothy E. O’Sullivan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
20
|
Dulfer EA, Joosten LAB, Netea MG. Enduring echoes: Post-infectious long-term changes in innate immunity. Eur J Intern Med 2024; 123:15-22. [PMID: 38135583 DOI: 10.1016/j.ejim.2023.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/16/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023]
Abstract
Upon encountering pathogens, the immune system typically responds by initiating an acute and self-limiting reaction, with symptoms subsiding after the pathogen has been cleared. However, long-term post-infectious clinical symptoms can manifest months or even years after the initial infection. 'Trained immunity', the functional reprogramming of innate immune cells through epigenetic and metabolic rewiring, has been proposed as a key concept for understanding these long-term effects. Although trained immunity can result in enhanced protection against reinfection with heterologous pathogens, it can also contribute to detrimental outcomes. Persisting and excessive inflammation can cause tissue damage and aggravate immune-mediated conditions and cardiovascular complications. On the other hand, suppression of immune cell effector functions by long-lasting epigenetic changes can result in post-infectious immune paralysis. Distinct stimuli can evoke different trained immunity programs, potentially resulting in different consequences for the host. In this review, we provide an overview of both the adaptive and maladaptive consequences of infectious diseases. We discuss how long-term immune dysregulation in patients can be addressed by tailoring host-directed interventions and identify areas of scientific and therapeutic potential to advance further.
Collapse
Affiliation(s)
- Elisabeth A Dulfer
- Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud university medical center, Nijmegen, the Netherlands.
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud university medical center, Nijmegen, the Netherlands; Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G Netea
- Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud university medical center, Nijmegen, the Netherlands; Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Germany
| |
Collapse
|
21
|
Zhang J, Li AM, Kansler ER, Li MO. Cancer immunity by tissue-resident type 1 innate lymphoid cells and killer innate-like T cells. Immunol Rev 2024; 323:150-163. [PMID: 38506480 PMCID: PMC11102320 DOI: 10.1111/imr.13319] [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] [Indexed: 03/21/2024]
Abstract
Cancer progression can be restrained by tumor-infiltrating lymphocytes in a process termed cancer immunosurveillance. Based on how lymphocytes are activated and recruited to the tumor tissue, cancer immunity is either pre-wired, in which innate lymphocytes and innate-like T cells are directly recruited to and activated in tumors following their differentiation in primary lymphoid organs; or priming-dependent, in which conventional adaptive T cells are first primed by cognate antigens in secondary lymphoid organs before homing to and reactivated in tumors. While priming-dependent cancer immunity has been a focus of cancer immunology research for decades, in part due to historical preconception of cancer theory and tumor model choice as well as clinical success of conventional adaptive T cell-directed therapeutic programs, recent studies have revealed that pre-wired cancer immunity mediated by tissue-resident type 1 innate lymphoid cells (ILC1s) and killer innate-like T cells (ILTCKs) is an integral component of the cancer immunosurveillance process. Herein we review the distinct ontogenies and cancer-sensing mechanisms of ILC1s and ILTCKs in murine genetic cancer models as well as the conspicuously conserved responses in human malignancies. How ILC1s and ILTCKs may be targeted to broaden the scope of cancer immunotherapy beyond conventional adaptive T cells is also discussed.
Collapse
Affiliation(s)
- Jing Zhang
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Albert M. Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Emily R. Kansler
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ming O. Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Graduate School of Biomedical Sciences, Cornell University, New York, NY, USA
| |
Collapse
|
22
|
Joshi VR, Altfeld M. Harnessing natural killer cells to target HIV-1 persistence. Curr Opin HIV AIDS 2024; 19:141-149. [PMID: 38457230 DOI: 10.1097/coh.0000000000000848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
PURPOSE OF REVIEW The purpose of this article is to review recent advances in the role of natural killer (NK) cells in approaches aimed at reducing the latent HIV-1 reservoir. RECENT FINDINGS Multiple approaches to eliminate cells harboring latent HIV-1 are being explored, but have been met with limited success so far. Recent studies have highlighted the role of NK cells and their potential in HIV-1 cure efforts. Anti-HIV-1 NK cell function can be optimized by enhancing NK cell activation, antibody dependent cellular cytotoxicity, reversing inhibition of NK cells as well as by employing immunotherapeutic complexes to enable HIV-1 specificity of NK cells. While NK cells alone do not eliminate the HIV-1 reservoir, boosting NK cell function might complement other strategies involving T cell and B cell immunity towards an HIV-1 functional cure. SUMMARY Numerous studies focusing on targeting latently HIV-1-infected cells have emphasized a potential role of NK cells in these strategies. Our review highlights recent advances in harnessing NK cells in conjunction with latency reversal agents and other immunomodulatory therapeutics to target HIV-1 persistence.
Collapse
Affiliation(s)
- Vinita R Joshi
- Department of Virus Immunology, Leibniz Institute of Virology
| | - Marcus Altfeld
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
23
|
Dang Q, Li B, Jin B, Ye Z, Lou X, Wang T, Wang Y, Pan X, Hu Q, Li Z, Ji S, Zhou C, Yu X, Qin Y, Xu X. Cancer immunometabolism: advent, challenges, and perspective. Mol Cancer 2024; 23:72. [PMID: 38581001 PMCID: PMC10996263 DOI: 10.1186/s12943-024-01981-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 03/06/2024] [Indexed: 04/07/2024] Open
Abstract
For decades, great strides have been made in the field of immunometabolism. A plethora of evidence ranging from basic mechanisms to clinical transformation has gradually embarked on immunometabolism to the center stage of innate and adaptive immunomodulation. Given this, we focus on changes in immunometabolism, a converging series of biochemical events that alters immune cell function, propose the immune roles played by diversified metabolic derivatives and enzymes, emphasize the key metabolism-related checkpoints in distinct immune cell types, and discuss the ongoing and upcoming realities of clinical treatment. It is expected that future research will reduce the current limitations of immunotherapy and provide a positive hand in immune responses to exert a broader therapeutic role.
Collapse
Affiliation(s)
- Qin Dang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Borui Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Bing Jin
- School of Clinical Medicine, Zhengzhou University, Zhengzhou, China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xin Lou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Ting Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Yan Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xuan Pan
- Department of Hepatobiliary Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Qiangsheng Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Zheng Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Chenjie Zhou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| |
Collapse
|
24
|
Safaei S, Fadaee M, Farzam OR, Yari A, Poursaei E, Aslan C, Samemaleki S, Shanehbandi D, Baradaran B, Kazemi T. Exploring the dynamic interplay between exosomes and the immune tumor microenvironment: implications for breast cancer progression and therapeutic strategies. Breast Cancer Res 2024; 26:57. [PMID: 38553754 PMCID: PMC10981336 DOI: 10.1186/s13058-024-01810-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/18/2024] [Indexed: 04/01/2024] Open
Abstract
Breast cancer continues to pose a substantial worldwide health concern, demanding a thorough comprehension of the complex interaction between cancerous cells and the immune system. Recent studies have shown the significant function of exosomes in facilitating intercellular communication and their participation in the advancement of cancer. Tumor-derived exosomes have been identified as significant regulators in the context of breast cancer, playing a crucial role in modulating immune cell activity and contributing to the advancement of the illness. This study aims to investigate the many effects of tumor-derived exosomes on immune cells in the setting of breast cancer. Specifically, we will examine their role in influencing immune cell polarization, facilitating immunological evasion, and modifying the tumor microenvironment. Furthermore, we explore the nascent domain of exosomes produced from immune cells and their prospective involvement in the prevention of breast cancer. This paper focuses on new research that emphasizes the immunomodulatory characteristics of exosomes produced from immune cells. It also explores the possibility of these exosomes as therapeutic agents or biomarkers for the early identification and prevention of breast cancer. The exploration of the reciprocal connections between exosomes formed from tumors and immune cells, together with the rising significance of exosomes derived from immune cells, presents a potential avenue for the advancement of novel approaches in the field of breast cancer therapy and prevention.
Collapse
Affiliation(s)
- Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Manouchehr Fadaee
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Omid Rahbar Farzam
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Amirhossein Yari
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
- Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Elham Poursaei
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Cynthia Aslan
- Research Center for Integrative Medicine in Aging, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Samemaleki
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran.
| |
Collapse
|
25
|
Li R, Galindo CC, Davidson D, Guo H, Zhong MC, Qian J, Li B, Ruzsics Z, Lau CM, O'Sullivan TE, Vidal SM, Sun JC, Veillette A. Suppression of adaptive NK cell expansion by macrophage-mediated phagocytosis inhibited by 2B4-CD48. Cell Rep 2024; 43:113800. [PMID: 38386559 DOI: 10.1016/j.celrep.2024.113800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 12/21/2023] [Accepted: 01/31/2024] [Indexed: 02/24/2024] Open
Abstract
Infection of mice by mouse cytomegalovirus (MCMV) triggers activation and expansion of Ly49H+ natural killer (NK) cells, which are virus specific and considered to be "adaptive" or "memory" NK cells. Here, we find that signaling lymphocytic activation molecule family receptors (SFRs), a group of hematopoietic cell-restricted receptors, are essential for the expansion of Ly49H+ NK cells after MCMV infection. This activity is largely mediated by CD48, an SFR broadly expressed on NK cells and displaying augmented expression after MCMV infection. It is also dependent on the CD48 counter-receptor, 2B4, expressed on host macrophages. The 2B4-CD48 axis promotes expansion of Ly49H+ NK cells by repressing their phagocytosis by virus-activated macrophages through inhibition of the pro-phagocytic integrin lymphocyte function-associated antigen-1 (LFA-1) on macrophages. These data identify key roles of macrophages and the 2B4-CD48 pathway in controlling the expansion of adaptive NK cells following MCMV infection. Stimulation of the 2B4-CD48 axis may be helpful in enhancing adaptive NK cell responses for therapeutic purposes.
Collapse
Affiliation(s)
- Rui Li
- Laboratory of Molecular Oncology, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada; Department of Medicine, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Cristian Camilo Galindo
- Laboratory of Molecular Oncology, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada; Department of Medicine, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Dominique Davidson
- Laboratory of Molecular Oncology, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada
| | - Huaijian Guo
- Laboratory of Molecular Oncology, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada; Department of Medicine, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Ming-Chao Zhong
- Laboratory of Molecular Oncology, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada
| | - Jin Qian
- Laboratory of Molecular Oncology, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada
| | - Bin Li
- Laboratory of Molecular Oncology, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada; Molecular Biology Program, University of Montréal, Montréal, QC H3T 1J4, Canada
| | - Zsolt Ruzsics
- Institute of Virology, University Medical Center, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Colleen M Lau
- Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Silvia M Vidal
- Department of Human Genetics, McGill University, Montréal, QC H3A 0C7, Canada; Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, QC H3A 0G1, Canada
| | - Joseph C Sun
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY 10065, USA
| | - André Veillette
- Laboratory of Molecular Oncology, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada; Department of Medicine, McGill University, Montréal, QC H3G 1Y6, Canada; Molecular Biology Program, University of Montréal, Montréal, QC H3T 1J4, Canada.
| |
Collapse
|
26
|
Shang J, Hu S, Wang X. Targeting natural killer cells: from basic biology to clinical application in hematologic malignancies. Exp Hematol Oncol 2024; 13:21. [PMID: 38396050 PMCID: PMC10885621 DOI: 10.1186/s40164-024-00481-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 01/18/2024] [Indexed: 02/25/2024] Open
Abstract
Natural killer (NK) cell belongs to innate lymphoid cell family that contributes to host immunosurveillance and defense without pre-immunization. Emerging studies have sought to understand the underlying mechanism behind NK cell dysfunction in tumor environments, and provide numerous novel therapeutic targets for tumor treatment. Strategies to enhance functional activities of NK cell have exhibited promising efficacy and favorable tolerance in clinical treatment of tumor patients, such as immune checkpoint blockade (ICB), chimeric antigen receptor NK (CAR-NK) cell, and bi/trispecific killer cell engager (BiKE/TriKE). Immunotherapy targeting NK cell provides remarkable advantages compared to T cell therapy, including a decreased rate of graft versus-host disease (GvHD) and neurotoxicity. Nevertheless, advanced details on how to support the maintenance and function of NK cell to obtain better response rate and longer duration still remain to be elucidated. This review systematically summarizes the profound role of NK cells in tumor development, highlights up-to-date advances and current challenges of therapy targeting NK cell in the clinical treatment of hematologic malignancies.
Collapse
Affiliation(s)
- Juanjuan Shang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, 250021, Shandong, China
| | - Shunfeng Hu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, 250021, Shandong, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Taishan Scholars Program of Shandong Province, Jinan, 250021, Shandong, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| |
Collapse
|
27
|
He S, Su L, Hu H, Liu H, Xiong J, Gong X, Chi H, Wu Q, Yang G. Immunoregulatory functions and therapeutic potential of natural killer cell-derived extracellular vesicles in chronic diseases. Front Immunol 2024; 14:1328094. [PMID: 38239346 PMCID: PMC10795180 DOI: 10.3389/fimmu.2023.1328094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024] Open
Abstract
Extracellular vesicles (EVs) have been proven to play a significant immunoregulatory role in many chronic diseases, such as cancer and immune disorders. Among them, EVs derived from NK cells are an essential component of the immune cell functions. These EVs have been demonstrated to carry a variety of toxic proteins and nucleic acids derived from NK cells and play a therapeutic role in diseases like malignancies, liver fibrosis, and lung injury. However, natural NK-derived EVs (NKEVs) have certain limitations in disease treatment, such as low yield and poor targeting. Concurrently, NK cells exhibit characteristics of memory-like NK cells, which have stronger proliferative capacity, increased IFN-γ production, and enhanced cytotoxicity, making them more advantageous for disease treatment. Recent research has shifted its focus towards engineered extracellular vesicles and their potential to improve the efficiency, specificity, and safety of disease treatments. In this review, we will discuss the characteristics of NK-derived EVs and the latest advancements in disease therapy. Specifically, we will compare different cellular sources of NKEVs and explore the current status and prospects of memory-like NK cell-derived EVs and engineered NKEVs.
Collapse
Affiliation(s)
- Shuang He
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Lanqian Su
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Haiyang Hu
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Haiqi Liu
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Jingwen Xiong
- Department of Sports Rehabilitation, Southwest Medical University, Luzhou, China
| | - Xiangjin Gong
- Department of Sports Rehabilitation, Southwest Medical University, Luzhou, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Qibiao Wu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| |
Collapse
|
28
|
Choi S, Valente D, Virone‐Oddos A, Mauriac C. Developing a mechanistic translational PK/PD model for a trifunctional NK cell engager to predict the first-in-human dose for acute myeloid leukemia. Clin Transl Sci 2024; 17:e13689. [PMID: 37990450 PMCID: PMC10772472 DOI: 10.1111/cts.13689] [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: 10/03/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/23/2023] Open
Abstract
Natural killer cell engagers (NKCEs), a treatment that stimulates innate immunity, have lately gained attention owing to their favorable safety profile, and their efficacy. Natural killer (NK) cell activation is driven by immune synapse formation between drugs, NK cells, and tumor cells. However, no clear translational modeling approach has been reported for first-in-human (FIH) dose estimation of humanized NKCEs. We developed the first translational mechanistic synapse-driven pharmacokinetic/pharmacodynamic (PK/PD) model for a trifunctional NKp46/CD16a-CD123 (CD123-NKCE) by integrating (i) in vitro target cell cytotoxicity in MOLM-13 tumor cell lines at varying effector-to-tumor cell ratios and incubation intervals; (ii) nonhuman primate PK and profiles of CD123+ cells and NKP46+ NK cells; and (iii) healthy human or patients with acute myeloid leukemia system-specific parameters. To depict direct tumor cell killing by the innate immunity, no transit compartment was included in PK/PD model structures. Model predictions suggested an intrapatient dose escalation of 10/30/100 μg/kg twice weekly to be selected as the starting dose in the FIH trial. However, sensitivity analyses revealed that CD123+ cell growth rate constant and maximal tumor killing rate constant were the key uncertainties to the recommended active dose. This novel translational model structure can be used as the basis to predict clinical PK/PD data for CD123-NKCE, and the translational strategy may serve as a foundation for future advancements of NKCEs.
Collapse
|
29
|
Perales SG, Rajasingh S, Zhou Z, Rajasingh J. Therapy of infectious diseases using epigenetic approaches. EPIGENETICS IN HUMAN DISEASE 2024:853-882. [DOI: 10.1016/b978-0-443-21863-7.00007-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
30
|
Bakhtiyaridovvombaygi M, Yazdanparast S, Mikanik F, Izadpanah A, Parkhideh S, Shahbaz Ghasabeh A, Roshandel E, Hajifathali A, Gharehbaghian A. Cytokine-Induced Memory-Like NK Cells: Emerging strategy for AML immunotherapy. Biomed Pharmacother 2023; 168:115718. [PMID: 37857247 DOI: 10.1016/j.biopha.2023.115718] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease developed from the malignant expansion of myeloid precursor cells in the bone marrow and peripheral blood. The implementation of intensive chemotherapy and hematopoietic stem cell transplantation (HSCT) has improved outcomes associated with AML, but relapse, along with suboptimal outcomes, is still a common scenario. In the past few years, exploring new therapeutic strategies to optimize treatment outcomes has occurred rapidly. In this regard, natural killer (NK) cell-based immunotherapy has attracted clinical interest due to its critical role in immunosurveillance and their capabilities to target AML blasts. NK cells are cytotoxic innate lymphoid cells that mediate anti-viral and anti-tumor responses by producing pro-inflammatory cytokines and directly inducing cytotoxicity. Although NK cells are well known as short-lived innate immune cells with non-specific responses that have limited their clinical applications, the discovery of cytokine-induced memory-like (CIML) NK cells could overcome these challenges. NK cells pre-activated with the cytokine combination IL-12/15/18 achieved a long-term life span with adaptive immunity characteristics, termed CIML-NK cells. Previous studies documented that using CIML-NK cells in cancer treatment is safe and results in promising outcomes. This review highlights the current application, challenges, and opportunities of CIML-NK cell-based therapy in AML.
Collapse
Affiliation(s)
- Mehdi Bakhtiyaridovvombaygi
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Yazdanparast
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mikanik
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhossein Izadpanah
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Parkhideh
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amin Shahbaz Ghasabeh
- Department of Hematology and Blood Bank, School of Allied Medical Science, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Elham Roshandel
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ahmad Gharehbaghian
- Department of Hematology and Blood Bank, School of Allied Medical Science, Shahid Beheshti University of Medical Science, Tehran, Iran; Pediatric Congenital Hematologic Disorders Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Huntington ND. Ironman training for NK cells. Nat Immunol 2023; 24:1599-1601. [PMID: 37697098 DOI: 10.1038/s41590-023-01626-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Affiliation(s)
- Nicholas D Huntington
- Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia.
- oNKo-Innate, Moonee Ponds, Victoria, Australia.
| |
Collapse
|
32
|
Lu L, Yang C, Zhou X, Wu L, Hong X, Li W, Wang X, Yang Y, Cao D, Zhang A, Di W, Deng L. STING signaling promotes NK cell antitumor immunity and maintains a reservoir of TCF-1 + NK cells. Cell Rep 2023; 42:113108. [PMID: 37708030 DOI: 10.1016/j.celrep.2023.113108] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 08/03/2023] [Accepted: 08/24/2023] [Indexed: 09/16/2023] Open
Abstract
Natural killer (NK) cells are cytotoxic innate lymphocytes that eradicate tumor cells. Inducing durable antitumor immune responses by NK cells represents a major priority of cancer immunotherapy. While cytosolic DNA sensing plays an essential role in initiating antitumor immunity, the role of NK cell-intrinsic STING signaling remains unclear. Here, we find that NK cell-intrinsic STING promotes antitumor responses and maintains a reservoir of TCF-1+ NK cells. In contrast, tumor cell-intrinsic cGAS and mtDNA are required for NK cell antitumor activity, indicating that tumor mtDNA recognition by cGAS partially triggers NK cell-intrinsic STING activation. Moreover, addition of cGAMP enables STING activation and type I interferon production in NK cells, thereby supporting the activation of NK cells in vitro. In humans, STING agonism promotes the expansion of TCF-1+ NK cells. This study provides insight into understanding how STING signaling drives NK cell antitumor immunity and the development of NK cell-based cancer immunotherapy.
Collapse
Affiliation(s)
- Lu Lu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chao Yang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xingyue Zhou
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lingling Wu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaochuan Hong
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wenwen Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xinran Wang
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yuanqin Yang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Dongqing Cao
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ao Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wen Di
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Liufu Deng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
33
|
Chang Y, Jin G, Luo W, Luo Q, Jung J, Hummel SN, Torregrosa-Allen S, Elzey BD, Low PS, Lian XL, Bao X. Engineered human pluripotent stem cell-derived natural killer cells with PD-L1 responsive immunological memory for enhanced immunotherapeutic efficacy. Bioact Mater 2023; 27:168-180. [PMID: 37091063 PMCID: PMC10113709 DOI: 10.1016/j.bioactmat.2023.03.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/14/2023] [Accepted: 03/22/2023] [Indexed: 04/08/2023] Open
Abstract
Adoptive chimeric antigen receptor (CAR)-engineered natural killer (NK) cells have shown promise in treating various cancers. However, limited immunological memory and access to sufficient numbers of allogenic donor cells have hindered their broader preclinical and clinical applications. Here, we first assess eight different CAR constructs that use an anti-PD-L1 nanobody and/or universal anti-fluorescein (FITC) single-chain variable fragment (scFv) to enhance antigen-specific proliferation and anti-tumor cytotoxicity of NK-92 cells against heterogenous solid tumors. We next genetically engineer human pluripotent stem cells (hPSCs) with optimized CARs and differentiate them into functional dual CAR-NK cells. The tumor microenvironment responsive anti-PD-L1 CAR effectively promoted hPSC-NK cell proliferation and cytotoxicity through antigen-dependent activation of phosphorylated STAT3 (pSTAT3) and pSTAT5 signaling pathways via an intracellular truncated IL-2 receptor β-chain (ΔIL-2Rβ) and STAT3-binding tyrosine-X-X-glutamine (YXXQ) motif. Anti-tumor activities of PD-L1-induced memory-like hPSC-NK cells were further boosted by administering a FITC-folate bi-specific adapter that bridges between a programmable anti-FITC CAR and folate receptor alpha-expressing breast tumor cells. Collectively, our hPSC CAR-NK engineering platform is modular and could constitute a realistic strategy to manufacture off-the-shelf CAR-NK cells with immunological memory-like phenotype for targeted immunotherapy.
Collapse
|
34
|
Hatami Z, Hashemi ZS, Eftekhary M, Amiri A, Karpisheh V, Nasrollahi K, Jafari R. Natural killer cell-derived exosomes for cancer immunotherapy: innovative therapeutics art. Cancer Cell Int 2023; 23:157. [PMID: 37543612 PMCID: PMC10403883 DOI: 10.1186/s12935-023-02996-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 07/19/2023] [Indexed: 08/07/2023] Open
Abstract
Chimeric antigen receptor natural killer cells (CAR-NK) promote off-the-shelf cellular therapy for solid tumors and malignancy.However,, the development of CAR-NK is due to their immune surveillance uncertainty and cytotoxicity challenge was restricted. Natural killer cell-derived exosome (NK-Exo) combine crucial targeted cellular therapies of NK cell therapies with unique non-toxic Exo as a self-origin shuttle against cancer immunotherapy. This review study covers cytokines, adoptive (autologous and allogenic) NK immunotherapy, stimulatory and regulatory functions, and cell-free derivatives from NK cells. The future path of NK-Exo cytotoxicity and anti-tumor activity with considering non-caspase-independent/dependent apoptosis and Fas/FasL pathway in cancer immunotherapy. Finally, the significance and implication of NK-Exo therapeutics through combination therapy and the development of emerging approaches for the purification and delivery NK-Exo to severe immune and tumor cells and tissues were discussed in detail.
Collapse
Affiliation(s)
- Zahra Hatami
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Sadat Hashemi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| | - Mohamad Eftekhary
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ala Amiri
- Department of Biotechnology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Vahid Karpisheh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kaveh Nasrollahi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reza Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
35
|
Camarasa TMN, Torné J, Chevalier C, Rasid O, Hamon MA. Streptococcus pneumoniae drives specific and lasting Natural Killer cell memory. PLoS Pathog 2023; 19:e1011159. [PMID: 37486946 PMCID: PMC10399893 DOI: 10.1371/journal.ppat.1011159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 08/03/2023] [Accepted: 06/27/2023] [Indexed: 07/26/2023] Open
Abstract
NK cells are important mediators of innate immunity and play an essential role for host protection against infection, although their responses to bacteria are poorly understood. Recently NK cells were shown to display memory properties, as characterized by an epigenetic signature leading to a stronger secondary response. Although NK cell memory could be a promising mechanism to fight against infection, it has not been described upon bacterial infection. Using a mouse model, we reveal that NK cells develop specific and long-term memory following sub-lethal infection with the extracellular pathogen Streptococcus pneumoniae. Memory NK cells display intrinsic sensing and response to bacteria in vitro, in a manner that is enhanced post-bacterial infection. In addition, their transfer into naïve mice confers protection from lethal infection for at least 12 weeks. Interestingly, NK cells display enhanced cytotoxic molecule production upon secondary stimulation and their protective role is dependent on Perforin and independent of IFNγ. Thus, our study identifies a new role for NK cells during bacterial infection, opening the possibility to harness innate immune memory for therapeutic purposes.
Collapse
Affiliation(s)
- Tiphaine M. N. Camarasa
- Chromatin and Infection Unit, Institut Pasteur, Paris, France
- Université Paris Cité, 562 Bio Sorbonne Paris Cité, Paris, France
| | - Júlia Torné
- Chromatin and Infection Unit, Institut Pasteur, Paris, France
| | | | - Orhan Rasid
- Chromatin and Infection Unit, Institut Pasteur, Paris, France
| | | |
Collapse
|
36
|
Carannante V, Wiklund M, Önfelt B. In vitro models to study natural killer cell dynamics in the tumor microenvironment. Front Immunol 2023; 14:1135148. [PMID: 37457703 PMCID: PMC10338882 DOI: 10.3389/fimmu.2023.1135148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Immunotherapy is revolutionizing cancer therapy. The rapid development of new immunotherapeutic strategies to treat solid tumors is posing new challenges for preclinical research, demanding novel in vitro methods to test treatments. Such methods should meet specific requirements, such as enabling the evaluation of immune cell responses like cytotoxicity or cytokine release, and infiltration into the tumor microenvironment using cancer models representative of the original disease. They should allow high-throughput and high-content analysis, to evaluate the efficacy of treatments and understand immune-evasion processes to facilitate development of new therapeutic targets. Ideally, they should be suitable for personalized immunotherapy testing, providing information for patient stratification. Consequently, the application of in vitro 3-dimensional (3D) cell culture models, such as tumor spheroids and organoids, is rapidly expanding in the immunotherapeutic field, coupled with the development of novel imaging-based techniques and -omic analysis. In this paper, we review the recent advances in the development of in vitro 3D platforms applied to natural killer (NK) cell-based cancer immunotherapy studies, highlighting the benefits and limitations of the current methods, and discuss new concepts and future directions of the field.
Collapse
Affiliation(s)
- Valentina Carannante
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Martin Wiklund
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Björn Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
- Center for Infectious Medicine, Department of Medicine Huddinge, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
37
|
Riffelmacher T, Paynich Murray M, Wientjens C, Chandra S, Cedillo-Castelán V, Chou TF, McArdle S, Dillingham C, Devereaux J, Nilsen A, Brunel S, Lewinsohn DM, Hasty J, Seumois G, Benedict CA, Vijayanand P, Kronenberg M. Divergent metabolic programmes control two populations of MAIT cells that protect the lung. Nat Cell Biol 2023; 25:877-891. [PMID: 37231163 PMCID: PMC10264248 DOI: 10.1038/s41556-023-01152-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 04/18/2023] [Indexed: 05/27/2023]
Abstract
Although mucosal-associated invariant T (MAIT) cells provide rapid, innate-like responses, they are not pre-set, and memory-like responses have been described for MAIT cells following infections. The importance of metabolism for controlling these responses, however, is unknown. Here, following pulmonary immunization with a Salmonella vaccine strain, mouse MAIT cells expanded as separate CD127-Klrg1+ and CD127+Klrg1- antigen-adapted populations that differed in terms of their transcriptome, function and localization in lung tissue. These populations remained altered from steady state for months as stable, separate MAIT cell lineages with enhanced effector programmes and divergent metabolism. CD127+ MAIT cells engaged in an energetic, mitochondrial metabolic programme, which was critical for their maintenance and IL-17A synthesis. This programme was supported by high fatty acid uptake and mitochondrial oxidation and relied on highly polarized mitochondria and autophagy. After vaccination, CD127+ MAIT cells protected mice against Streptococcus pneumoniae infection. In contrast, Klrg1+ MAIT cells had dormant but ready-to-respond mitochondria and depended instead on Hif1a-driven glycolysis to survive and produce IFN-γ. They responded antigen independently and participated in protection from influenza virus. These metabolic dependencies may enable tuning of memory-like MAIT cell responses for vaccination and immunotherapies.
Collapse
Affiliation(s)
- Thomas Riffelmacher
- La Jolla Institute for Immunology, La Jolla, CA, USA.
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
| | | | | | | | | | | | - Sara McArdle
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | | | - Aaron Nilsen
- Oregon Health and Science University, Portland, OR, USA
| | - Simon Brunel
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Jeff Hasty
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | | | | | | | - Mitchell Kronenberg
- La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
38
|
Cimpean M, Cooper MA. Metabolic regulation of NK cell antiviral functions during cytomegalovirus infection. J Leukoc Biol 2023; 113:525-534. [PMID: 36843434 PMCID: PMC11262056 DOI: 10.1093/jleuko/qiad018] [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: 11/18/2022] [Revised: 01/23/2023] [Accepted: 02/09/2023] [Indexed: 02/28/2023] Open
Abstract
Natural killer (NK) cells quickly mount cytotoxic responses, produce cytokines, and proliferate in response to infected or transformed cells. Moreover, they can develop memory, with enhanced effector responses following activation, in some cases with antigen specificity. To optimally execute these functions, NK cells undergo metabolic reprogramming. Here, we discuss the interplay between metabolism and NK cell function in the context of viral infections. We review findings supporting metabolic regulation of NK cell effector functions, with a focus on NK cell antiviral infection in the context of cytomegalovirus in the mouse (MCMV) and human (HCMV).
Collapse
Affiliation(s)
- Maria Cimpean
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University in St. Louis, St. Louis, USA
| | - Megan A. Cooper
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University in St. Louis, St. Louis, USA
| |
Collapse
|
39
|
Lizana-Vasquez GD, Torres-Lugo M, Dixon R, Powderly JD, Warin RF. The application of autologous cancer immunotherapies in the age of memory-NK cells. Front Immunol 2023; 14:1167666. [PMID: 37205105 PMCID: PMC10185894 DOI: 10.3389/fimmu.2023.1167666] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/17/2023] [Indexed: 05/21/2023] Open
Abstract
Cellular immunotherapy has revolutionized the oncology field, yielding improved results against hematological and solid malignancies. NK cells have become an attractive alternative due to their capacity to activate upon recognition of "stress" or "danger" signals independently of Major Histocompatibility Complex (MHC) engagement, thus making tumor cells a perfect target for NK cell-mediated cancer immunotherapy even as an allogeneic solution. While this allogeneic use is currently favored, the existence of a characterized memory function for NK cells ("memory-like" NK cells) advocates for an autologous approach, that would benefit from the allogeneic setting discoveries, but with added persistence and specificity. Still, both approaches struggle to exert a sustained and high anticancer effect in-vivo due to the immunosuppressive tumor micro-environment and the logistical challenges of cGMP production or clinical deployment. Novel approaches focused on the quality enhancement and the consistent large-scale production of highly activated therapeutic memory-like NK cells have yielded encouraging but still unconclusive results. This review provides an overview of NK biology as it relates to cancer immunotherapy and the challenge presented by solid tumors for therapeutic NKs. After contrasting the autologous and allogeneic NK approaches for solid cancer immunotherapy, this work will present the current scientific focus for the production of highly persistent and cytotoxic memory-like NK cells as well as the current issues with production methods as they apply to stress-sensitive immune cells. In conclusion, autologous NK cells for cancer immunotherapy appears to be a prime alternative for front line therapeutics but to be successful, it will be critical to establish comprehensives infrastructures allowing the production of extremely potent NK cells while constraining costs of production.
Collapse
Affiliation(s)
- Gaby D. Lizana-Vasquez
- Department of Chemical Engineering, University of Puerto Rico-Mayagüez, Mayagüez, Puerto Rico
- Cancer Research Clinic, Carolina BioOncology Institute (CBOI), Huntersville, NC, United States
| | - Madeline Torres-Lugo
- Department of Chemical Engineering, University of Puerto Rico-Mayagüez, Mayagüez, Puerto Rico
| | - R. Brent Dixon
- Cancer Research Clinic, Carolina BioOncology Institute (CBOI), Huntersville, NC, United States
- Human Applications Lab (HAL) - BioCytics, Huntersville, NC, United States
| | - John D. Powderly
- Cancer Research Clinic, Carolina BioOncology Institute (CBOI), Huntersville, NC, United States
- Human Applications Lab (HAL) - BioCytics, Huntersville, NC, United States
| | - Renaud F. Warin
- Cancer Research Clinic, Carolina BioOncology Institute (CBOI), Huntersville, NC, United States
- Human Applications Lab (HAL) - BioCytics, Huntersville, NC, United States
| |
Collapse
|
40
|
Hojjatipour T, Maali A, Azad M. Natural killer cell epigenetic reprogramming in tumors and potential for cancer immunotherapy. Epigenomics 2023; 15:249-266. [PMID: 37125432 DOI: 10.2217/epi-2022-0454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Natural killer (NK) cells are critical members of the innate lymphoid cell population and have a pivotal role in cancer eradication. NK cell maturation, development and function are tightly regulated by epigenetic modifications, which can also be recruited for cancer propagation and immune escape. NK cells have the potential to be activated against tumors through several epigenetic regulators. Given that epigenetic changes are inducible and reversible, focusing on aberrant epigenetic regulations recruited by tumor cells provides a tremendous opportunity for cancer treatment. This review presents a comprehensive picture of NK cell normal epigenetic regulation and cancer-driven epigenetic modifications. From our perspective, a better understanding of epigenetic regulators that can edit and revise NK cells' activity is a promising avenue for NK cell-based therapy in cancer management.
Collapse
Affiliation(s)
- Tahereh Hojjatipour
- Department of Hematology & Blood Transfusion, Students Research Center, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Department of Medical Biotechnology, School of Paramedicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mehdi Azad
- Department of Medical Laboratory Sciences, School of Paramedicine, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
41
|
Ma K, Zheng ZR, Meng Y. Natural Killer Cells, as the Rising Point in Tissues, Are Forgotten in the Kidney. Biomolecules 2023; 13:biom13050748. [PMID: 37238618 DOI: 10.3390/biom13050748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/13/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Natural killer (NK) cells are members of a rapidly expanding family of innate lymphoid cells (ILCs). NK cells play roles in the spleen, periphery, and in many tissues, such as the liver, uterine, lung, adipose, and so on. While the immunological functions of NK cells are well established in these organs, comparatively little is known about NK cells in the kidney. Our understanding of NK cells is rapidly rising, with more and more studies highlighting the functional significance of NK cells in different types of kidney diseases. Recent progress has been made in translating these findings to clinical diseases that occur in the kidney, with indications of subset-specific roles of NK cells in the kidney. For the development of targeted therapeutics to delay kidney disease progression, a better understanding of the NK cell with respect to the mechanisms of kidney diseases is necessary. In order to promote the targeted treatment ability of NK cells in clinical diseases, in this paper we demonstrate the roles that NK cells play in different organs, especially the functions of NK cells in the kidney.
Collapse
Affiliation(s)
- Ke Ma
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou 510000, China
| | - Zi-Run Zheng
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou 510000, China
| | - Yu Meng
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou 510000, China
- Department of Nephrology, The Fifth Affiliated Hospital of Jinan University, Heyuan 570000, China
| |
Collapse
|
42
|
Berrien-Elliott MM, Jacobs MT, Fehniger TA. Allogeneic natural killer cell therapy. Blood 2023; 141:856-868. [PMID: 36416736 PMCID: PMC10023727 DOI: 10.1182/blood.2022016200] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/17/2022] [Accepted: 11/20/2022] [Indexed: 11/24/2022] Open
Abstract
Interest in adoptive cell therapy for treating cancer is exploding owing to early clinical successes of autologous chimeric antigen receptor (CAR) T lymphocyte therapy. However, limitations using T cells and autologous cell products are apparent as they (1) take weeks to generate, (2) utilize a 1:1 donor-to-patient model, (3) are expensive, and (4) are prone to heterogeneity and manufacturing failures. CAR T cells are also associated with significant toxicities, including cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and prolonged cytopenias. To overcome these issues, natural killer (NK) cells are being explored as an alternative cell source for allogeneic cell therapies. NK cells have an inherent ability to recognize cancers, mediate immune functions of killing and communication, and do not induce graft-versus-host disease, cytokine release syndrome, or immune effector cell-associated neurotoxicity syndrome. NK cells can be obtained from blood or cord blood or be derived from hematopoietic stem and progenitor cells or induced pluripotent stem cells, and can be expanded and cryopreserved for off-the-shelf availability. The first wave of point-of-care NK cell therapies led to the current allogeneic NK cell products being investigated in clinical trials with promising preliminary results. Basic advances in NK cell biology and cellular engineering have led to new translational strategies to block inhibition, enhance and broaden target cell recognition, optimize functional persistence, and provide stealth from patients' immunity. This review details NK cell biology, as well as NK cell product manufacturing, engineering, and combination therapies explored in the clinic leading to the next generation of potent, off-the-shelf cellular therapies for blood cancers.
Collapse
Affiliation(s)
| | - Miriam T. Jacobs
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Todd A. Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
43
|
Harnessing Innate Immunity to Treat Mycobacterium tuberculosis Infections: Heat-Killed Caulobacter crescentus as a Novel Biotherapeutic. Cells 2023; 12:cells12040560. [PMID: 36831226 PMCID: PMC9954702 DOI: 10.3390/cells12040560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/12/2023] Open
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis (Mtb), is a serious and devastating infectious disease worldwide. Approximately a quarter of the world population harbors latent Mtb infection without pathological consequences. Exposure of immunocompetent healthy individuals with Mtb does not result in active disease in more than 90% individuals, suggesting a defining role of host immunity to prevent and/or clear early infection. However, innate immune stimulation strategies have been relatively underexplored for the treatment of tuberculosis. In this study, we used cell culture and mouse models to examine the role of a heat-killed form of a non-pathogenic microbe, Caulobacter crescentus (HKCC), in inducing innate immunity and limiting Mtb infection. We also examined the added benefits of a distinct chemo-immunotherapeutic strategy that incorporates concurrent treatments with low doses of a first-line drug isoniazid and HKCC. This therapeutic approach resulted in highly significant reductions in disseminated Mtb in the lungs, liver, and spleen of mice compared to either agent alone. Our studies demonstrate the potential of a novel innate immunotherapeutic strategy with or without antimycobacterial drugs in controlling Mtb infection in mice and open new avenues for the treatment of tuberculosis in humans.
Collapse
|
44
|
Roles of natural killer cells in immunity to cancer, and applications to immunotherapy. Nat Rev Immunol 2023; 23:90-105. [PMID: 35637393 DOI: 10.1038/s41577-022-00732-1] [Citation(s) in RCA: 255] [Impact Index Per Article: 127.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 02/04/2023]
Abstract
Great strides have been made in recent years towards understanding the roles of natural killer (NK) cells in immunity to tumours and viruses. NK cells are cytotoxic innate lymphoid cells that produce inflammatory cytokines and chemokines. By lysing transformed or infected cells, they limit tumour growth and viral infections. Whereas T cells recognize peptides presented by MHC molecules, NK cells display receptors that recognize stress-induced autologous proteins on cancer cells. At the same time, their functional activity is inhibited by MHC molecules displayed on such cells. The enormous potential of NK cells for immunotherapy for cancer is illustrated by their broad recognition of stressed cells regardless of neoantigen presentation, and enhanced activity against tumours that have lost expression of MHC class I owing to acquired resistance mechanisms. As a result, many efforts are under way to mobilize endogenous NK cells with therapeutics, or to provide populations of ex vivo-expanded NK cells as a cellular therapy, in some cases by equipping the NK cells with chimeric antigen receptors. Here we consider the key features that underlie why NK cells are emerging as important new additions to the cancer therapeutic arsenal.
Collapse
|
45
|
Metabolic regulation of NK cell function: implications for immunotherapy. IMMUNOMETABOLISM (COBHAM (SURREY, ENGLAND)) 2023; 5:e00020. [PMID: 36710923 PMCID: PMC9869966 DOI: 10.1097/in9.0000000000000020] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/21/2022] [Indexed: 01/31/2023]
Abstract
Natural killer (NK) cells are innate immune lymphocytes capable of rapidly responding to tumors and infection without prior sensitization. There is increasing interest and success in harnessing NK cell function for the treatment of disease, in particular cancers. NK cell activation is dependent on integration of signals through cytokine and germline-encoded activating and inhibitory receptors. The availability of metabolic fuels and pathways is required for NK effector functions including proliferation, killing, and production of interferon gamma (IFN-γ). An understanding of NK cell immunometabolism is thus essential for developing immunotherapy approaches that will allow for optimal effector functions in patients. Studies in mice and humans have demonstrated stimulation-dependent metabolic changes that are required for NK cell function. Here we review the most recent findings in NK cell immunometabolism relevant to disease models and translation to therapy of patients.
Collapse
|
46
|
Oh BLZ, Chan LWY, Chai LYA. Manipulating NK cellular therapy from cancer to invasive fungal infection: promises and challenges. Front Immunol 2023; 13:1044946. [PMID: 36969979 PMCID: PMC10034767 DOI: 10.3389/fimmu.2022.1044946] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/09/2022] [Indexed: 01/13/2023] Open
Abstract
The ideal strategy to fight an infection involves both (i) weakening the invading pathogen through conventional antimicrobial therapy, and (ii) strengthening defense through the augmentation of host immunity. This is even more pertinent in the context of invasive fungal infections whereby the majority of patients have altered immunity and are unable to mount an appropriate host response against the pathogen. Natural killer (NK) cells fit the requirement of an efficient, innate executioner of both tumour cells and pathogens – their unique, targeted cell killing mechanism, combined with other arms of the immune system, make them potent effectors. These characteristics, together with their ready availability (given the various sources of extrinsic NK cells available for harvesting), make NK cells an attractive choice as adoptive cellular therapy against fungi in invasive infections. Improved techniques in ex vivo NK cell activation with expansion, and more importantly, recent advances in genetic engineering including state-of-the-art chimeric antigen receptor platform development, have presented an opportune moment to harness this novel therapeutic as a key component of a multipronged strategy against invasive fungal infections.
Collapse
Affiliation(s)
- Bernice Ling Zhi Oh
- VIVA-University Children’s Cancer Centre, Khoo-Teck Puat-National University Children’s Medical Institute, National University Hospital, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| | - Louis Wei Yong Chan
- Clinician Scientist Academy, National University Health System, Singapore, Singapore
| | - Louis Yi Ann Chai
- Division of Infectious Diseases, Department of Medicine, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- *Correspondence: Louis Yi Ann Chai,
| |
Collapse
|
47
|
Ghaedrahmati F, Esmaeil N, Abbaspour M. Targeting immune checkpoints: how to use natural killer cells for fighting against solid tumors. Cancer Commun (Lond) 2022; 43:177-213. [PMID: 36585761 PMCID: PMC9926962 DOI: 10.1002/cac2.12394] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 10/08/2022] [Accepted: 11/15/2022] [Indexed: 01/01/2023] Open
Abstract
Natural killer (NK) cells are unique innate immune cells that mediate anti-viral and anti-tumor responses. Thus, they might hold great potential for cancer immunotherapy. NK cell adoptive immunotherapy in humans has shown modest efficacy. In particular, it has failed to demonstrate therapeutic efficiency in the treatment of solid tumors, possibly due in part to the immunosuppressive tumor microenvironment (TME), which reduces NK cell immunotherapy's efficiencies. It is known that immune checkpoints play a prominent role in creating an immunosuppressive TME, leading to NK cell exhaustion and tumor immune escape. Therefore, NK cells must be reversed from their dysfunctional status and increased in their effector roles in order to improve the efficiency of cancer immunotherapy. Blockade of immune checkpoints can not only rescue NK cells from exhaustion but also augment their robust anti-tumor activity. In this review, we discussed immune checkpoint blockade strategies with a focus on chimeric antigen receptor (CAR)-NK cells to redirect NK cells to cancer cells in the treatment of solid tumors.
Collapse
Affiliation(s)
- Farhoodeh Ghaedrahmati
- Department of ImmunologySchool of MedicineIsfahan University of Medical SciencesIsfahanIran
| | - Nafiseh Esmaeil
- Department of ImmunologySchool of MedicineIsfahan University of Medical SciencesIsfahanIran,Research Institute for Primordial Prevention of Non‐Communicable DiseaseIsfahan University of Medical SciencesIsfahanIran
| | - Maryam Abbaspour
- Department of Pharmaceutical BiotechnologyFaculty of PharmacyIsfahan University of Medical SciencesIsfahanIran
| |
Collapse
|
48
|
Abstract
Osteoclasts are the only cells that can efficiently resorb bone. They do so by sealing themselves on to bone and removing the mineral and organic components. Osteoclasts are essential for bone homeostasis and are involved in the development of diseases associated with decreased bone mass, like osteoporosis, or abnormal bone turnover, like Paget's disease of bone. In addition, compromise of their development or resorbing machinery is pathogenic in multiple types of osteopetrosis. However, osteoclasts also have functions other than bone resorption. Like cells of the innate immune system, they are derived from myeloid precursors and retain multiple immune cell properties. In addition, there is now strong evidence that osteoclasts regulate osteoblasts through a process known as coupling, which coordinates rates of bone resorption and bone formation during bone remodeling. In this article we review the non-resorbing functions of osteoclasts and highlight their importance in health and disease.
Collapse
Affiliation(s)
- Kyung-Hyun Park-Min
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA.
| | - Joseph Lorenzo
- The Departments of Medicine and Orthopaedics, UConn Health, Farmington, CT 06030, USA.
| |
Collapse
|
49
|
Hasan MZ, Höltermann C, Petersen B, Schrod A, Mätz-Rensing K, Kaul A, Salinas G, Dressel R, Walter L. Detailed phenotypic and functional characterization of CMV-associated adaptive NK cells in rhesus macaques. Front Immunol 2022; 13:1028788. [PMID: 36518759 PMCID: PMC9742600 DOI: 10.3389/fimmu.2022.1028788] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/27/2022] [Indexed: 11/29/2022] Open
Abstract
Previous research on adaptive NK cells in rhesus macaques suffered from the lack of specific antibodies to differentiate between inhibitory CD94/NKG2A and stimulatory CD94/NKG2C heterodimeric receptors. Recently we reported an expansion of NKG2C receptor-encoding genes in rhesus macaques, but their expression and functional role on primary NK cells remained unknown due to this deficit. Thus, we established monoclonal antibodies 4A8 and 7B1 which show identical specificities and bind to both NKG2C-1 and NKG2C-2 but neither react with NKG2C-3 nor NKG2A on transfected cells. Using a combination of 4A8 and Z199 antibodies in multicolor flow cytometry we detected broad expression (4-73%) of NKG2C-1 and/or NKG2C-2 (NKG2C-1/2) on primary NK cells in rhesus macaques from our breeding colony. Stratifying our data to CMV-positive and CMV-negative animals, we noticed a higher proportion (23-73%) of primary NK cells expressing NKG2C-1/2 in CMV+ as compared to CMV- macaques (4-5%). These NKG2C-1/2-positive NK cells in CMV+ macaques are characterized by lower expression of IL12RB2, ZBTB16, SH2D1B, but not FCER1G, as well as high expression of IFNG, indicating that antibody 4A8 detects CMV-associated adaptive NK cells. Single cell RNA seq data of 4A8-positive NK cells from a rhCMV-positive macaque demonstrated that a high proportion of these adaptive NK cells transcribe in addition to NKG2C-1 and NKG2C-2 also NKG2C-3, but interestingly NKG2A as well. Remarkably, in comparison to NKG2A, NKG2C-1 and in particular NKG2C-2 bind Mamu-E with higher avidity. Primary NK cells exposed to Mamu-E-expressing target cells displayed strong degranulation as well as IFN-gamma expression of 4A8+ adaptive NK cells from rhCMV+ animals. Thus, despite co-expression of inhibitory and stimulatory CD94/NKG2 receptors the higher number of different stimulatory NKG2C receptors and their higher binding avidity to Mamu-E outreach inhibitory signaling via NKG2A. These data demonstrate the evolutionary conservation of the CMV-driven development of NKG2C-positive adaptive NK cells with particular molecular signatures in primates and with changes in gene copy numbers and ligand-binding strength of NKG2C isotypes. Thus, rhesus macaques represent a suitable and valuable nonhuman primate animal model to study the CMV-NKG2C liaison in vivo.
Collapse
Affiliation(s)
- Mohammad Zahidul Hasan
- Primate Genetics Laboratory, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
- PhD program Molecular Biology of Cells, GGNB, Georg August University, Göttingen, Germany
| | - Charlotte Höltermann
- Primate Genetics Laboratory, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
- PhD program Molecular Biology of Cells, GGNB, Georg August University, Göttingen, Germany
| | - Beatrix Petersen
- Primate Genetics Laboratory, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Annette Schrod
- Animal Husbandry, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Kerstin Mätz-Rensing
- Pathology Unit, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Artur Kaul
- Infection Biology Unit, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Gabriela Salinas
- NGS Core Unit for Integrative Genomics, Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Ralf Dressel
- Institute for Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Lutz Walter
- Primate Genetics Laboratory, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| |
Collapse
|
50
|
Della Camera G, Liu T, Yang W, Li Y, Puntes VF, Gioria S, Italiani P, Boraschi D. Induction of Innate Memory in Human Monocytes Exposed to Mixtures of Bacterial Agents and Nanoparticles. Int J Mol Sci 2022; 23:ijms232314655. [PMID: 36498992 PMCID: PMC9738562 DOI: 10.3390/ijms232314655] [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: 09/30/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022] Open
Abstract
We assessed whether concomitant exposure of human monocytes to bacterial agents and different engineered nanoparticles can affect the induction of protective innate memory, an immune mechanism that affords better resistance to diverse threatening challenges. Monocytes were exposed in vitro to nanoparticles of different chemical nature, shape and size either alone or admixed with LPS, and cell activation was assessed in terms of production of inflammatory (TNFα, IL-6) and anti-inflammatory cytokines (IL-10, IL-1Ra). After return to baseline conditions, cells were re-challenged with LPS and their secondary "memory" response measured. Results show that nanoparticles alone are essentially unable to generate memory, while LPS induced a tolerance memory response (less inflammatory cytokines, equal or increased anti-inflammatory cytokines). LPS-induced tolerance was not significantly affected by the presence of nanoparticles during the memory generation phase, although with substantial donor-to-donor variability. This suggests that, despite the overall lack of significant effects on LPS-induced innate memory, nanoparticles may have donor-specific effects. Thus, future nanosafety assessment and nanotherapeutic strategies will need a personalized approach in order to ensure both the safety and efficacy of nano medical compounds for individual patients.
Collapse
Affiliation(s)
- Giacomo Della Camera
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), 80131 Napoli, Italy
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy
| | - Tinghao Liu
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518055, China
- China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation (CNR, SIAT, SZN), SIAT, CAS, Shenzhen 518055, China
| | - Wenjie Yang
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518055, China
- China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation (CNR, SIAT, SZN), SIAT, CAS, Shenzhen 518055, China
| | - Yang Li
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518055, China
- China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation (CNR, SIAT, SZN), SIAT, CAS, Shenzhen 518055, China
| | - Victor F. Puntes
- Institut Català de Nanociència i Nanotecnologia (ICN2), Consejo Superior de Investigaciones Científicas (CSIC) and The Barcelona Institute of Science and Technology (BIST), 08036 Barcelona, Spain
- Vall d’Hebron Research Institute (VHIR), 08035 Barcelona, Spain
- Institució Catalana de Recerca I Estudis Avançats (ICREA), 08193 Barcelona, Spain
| | - Sabrina Gioria
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy
| | - Paola Italiani
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), 80131 Napoli, Italy
- Stazione Zoologica Anton Dohrn (SZN), 80121 Napoli, Italy
- China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation (CNR, SIAT, SZN), IBBC, CNR, 80131 Napoli, Italy
| | - Diana Boraschi
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), 80131 Napoli, Italy
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen 518055, China
- China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation (CNR, SIAT, SZN), SIAT, CAS, Shenzhen 518055, China
- Stazione Zoologica Anton Dohrn (SZN), 80121 Napoli, Italy
- China-Italy Joint Laboratory of Pharmacobiotechnology for Medical Immunomodulation (CNR, SIAT, SZN), IBBC, CNR, 80131 Napoli, Italy
- Correspondence:
| |
Collapse
|