1
|
Ge Z, Wu Q, Lv C, He Q. The Roles of T Cells in the Development of Metabolic Dysfunction-Associated Steatohepatitis. Immunology 2025. [PMID: 40414629 DOI: 10.1111/imm.13943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/28/2025] [Accepted: 04/28/2025] [Indexed: 05/27/2025] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH), the progressed period of metabolic dysfunction-associated steatotic liver disease (MASLD), is a multifaceted liver disease characterised by inflammation and fibrosis that develops from simple steatosis, even contributing to hepatocellular carcinoma and death. MASH involves several immune cell-mediated inflammation and fibrosis, where T cells play a crucial role through the release of pro-inflammatory cytokines and pro-fibrotic factors. This review discusses the complex role of various T cell subsets in the pathogenesis of MASH and highlights the progress of ongoing clinical trials involving T cell-targeted MASH therapies.
Collapse
Affiliation(s)
- Zhifa Ge
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qingwei Wu
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chengyu Lv
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qifeng He
- Department of General Surgery, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Stewart J, Damania B. Innate Immune Recognition of EBV. Curr Top Microbiol Immunol 2025. [PMID: 40399572 DOI: 10.1007/82_2025_297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Epstein-Barr virus (EBV) is a very successful human pathogen, with ~95% seroprevalence worldwide (Mentzer et al, Nat Commun 13:1818, 2022). If contracted in early childhood, EBV infection is typically asymptomatic; however, infections in adolescence and adulthood can manifest as infectious mononucleosis (IM). The innate immune response is the first line of defense, and its function is critical for controlling EBV infection. During EBV infection, components of the virus, known as pathogen-associated molecular patterns (PAMPs), are recognized by germline-encoded pattern recognition receptors (PRRs). PRRs are found on both non-immune and immune cells including antigen-presenting cells, such as macrophages, monocytes, dendritic cells, natural killer (NK), and mast cells. PRRs are also found on B cells and epithelial cells, the primary targets of EBV infection. Without immune surveillance, EBV can transform cells inducing various malignancies. Conversely, a prolonged innate immune response can lead to chronic inflammation which increases the likelihood of cancer. This review discusses innate immune recognition of EBV and its associated diseases.
Collapse
Affiliation(s)
- Jessica Stewart
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Blossom Damania
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
3
|
Tong J, Tan Y, Ouyang W, Chang H. Targeting immune checkpoints in hepatocellular carcinoma therapy: toward combination strategies with curative potential. Exp Hematol Oncol 2025; 14:65. [PMID: 40317077 PMCID: PMC12046748 DOI: 10.1186/s40164-025-00636-5] [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: 01/20/2025] [Accepted: 03/07/2025] [Indexed: 05/04/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a primary liver cancer characterized by poor immune cell infiltration and a strongly immunosuppressive microenvironment. Traditional treatments have often yielded unsatisfactory outcomes due to the insidious onset of the disease. Encouragingly, the introduction of immune checkpoint inhibitors (ICIs) has significantly transformed the approach to HCC treatment. Moreover, combining ICIs with other therapies or novel materials is considered the most promising opportunity in HCC, with some of these combinations already being evaluated in large-scale clinical trials. Unfortunately, most clinical trials fail to meet their endpoints, and the few successful ones also face challenges. This indicates that the potential of ICIs in HCC treatment remains underutilized, prompting a reevaluation of this promising therapy. Therefore, this article provides a review of the role of immune checkpoints in cancer treatment, the research progress of ICIs and their combination application in the treatment of HCC, aiming to open up avenues for the development of safer and more efficient immune checkpoint-related strategies for HCC treatment.
Collapse
Affiliation(s)
- Jing Tong
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
| | - Yongci Tan
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
| | - Wenwen Ouyang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
| | - Haocai Chang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China.
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
4
|
Fisher JG, Bartlett LG, Kashyap T, Walker CJ, Khakoo SI, Blunt MD. Modulation of anti-tumour immunity by XPO1 inhibitors. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002310. [PMID: 40291981 PMCID: PMC12022495 DOI: 10.37349/etat.2025.1002310] [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: 01/13/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Exportin-1 (XPO1) is a nuclear export protein that, when overexpressed, can facilitate cancer cell proliferation and survival and is frequently overexpressed or mutated in cancer patients. As such, selective inhibitors of XPO1 (XPO1i) function have been developed to inhibit cancer cell proliferation and induce apoptosis. This review outlines the evidence for the immunomodulatory properties of XPO1 inhibition and discusses the potential for combining and sequencing XPO1i with immunotherapy to improve the treatment of patients with cancer. Selinexor is a first-in-class XPO1i that is FDA-approved for the treatment of patients with relapsed and refractory (RR) multiple myeloma and RR diffuse large B cell lymphoma. In addition to the cancer cell intrinsic pro-apoptotic activity, increasing evidence suggests that XPO1 inhibition has immunomodulatory properties. In this review, we describe how XPO1i can lead to a skewing of macrophage polarisation, inhibition of neutrophil extracellular traps, modulation of immune checkpoint expression, blockade of myeloid-derived suppressor cells (MDSCs) and sensitisation of cancer cells to T cell and NK (natural killer) cell immunosurveillance. As such, there is an opportunity for selinexor to enhance immunotherapy efficacy and thus a need for clinical trials assessing selinexor in combination with immunotherapies such as immune checkpoint inhibitors, direct targeting monoclonal antibodies, chimeric antigen receptor (CAR)-T cells and cereblon E3 ligase modulators (CELMoDs).
Collapse
Affiliation(s)
- Jack G. Fisher
- Clinical and Experimental Sciences, University of Southampton, SO16 7YD Southampton, UK
| | - Laura G. Bartlett
- Clinical and Experimental Sciences, University of Southampton, SO16 7YD Southampton, UK
| | | | | | - Salim I. Khakoo
- Clinical and Experimental Sciences, University of Southampton, SO16 7YD Southampton, UK
| | - Matthew D. Blunt
- Clinical and Experimental Sciences, University of Southampton, SO16 7YD Southampton, UK
| |
Collapse
|
5
|
Vigano M, Wang L, As’sadiq A, Samarani S, Ahmad A, Costiniuk CT. Impact of cannabinoids on cancer outcomes in patients receiving immune checkpoint inhibitor immunotherapy. Front Immunol 2025; 16:1497829. [PMID: 40109334 PMCID: PMC11919899 DOI: 10.3389/fimmu.2025.1497829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025] Open
Abstract
Cannabinoids relieve pain, nausea, anorexia and anxiety, and improve quality of life in several cancer patients. The immunotherapy with checkpoint inhibitors (ICIs), although very successful in a subset of patients, is accompanied by moderate to severe immune-related adverse events (ir-AE) that often necessitate its discontinuation. Because of their role in symptomatic relief, cannabinoids have been used in combination with immune checkpoint inhibitor (ICI) immunotherapy. A few studies strongly suggest that the use of medicinal cannabis in cancer patients attenuates many of the ir-AE associated with the use of ICI immunotherapy and increase its tolerability. However, no significant beneficial effects on overall survival, progression free survival or cancer relapses were observed; rather, some of the studies noted adverse effects of concurrent administration of cannabinoids with ICI immunotherapy on the clinical benefits of the latter. Because of cannabinoids' well documented immunosuppressive effects mediated through the cannabinoid recptor-2 (CB2), we propose considering this receptor as an inhibitory immune checkpoint per se. A simultaneous neutralization of CB2, concurrent with cannabinoid treatment, may lead to better clinical outcomes in cancer patients receiving ICI immunotherapy. In this regard, cannabinoids such as cannabidiol (CBD) and cannabigerol (CBG), with little agonism for CB2, may be better therapeutic choices. Additional strategies e.g., the use of monoacylglycerol lipase (MAGL) inhibitors that degrade some endocannabinoids as well as lipogenesis and formation of lipid bilayers in cancer cells may also be explored. Future studies should take into consideration gut microbiota, CYP450 polymorphism and haplotypes, cannabinoid-drug interactions as well as genetic and somatic variations occurring in the cannabinoid receptors and their signaling pathways in cancer cells for personalized cannabis-based therapies in cancer patients receiving ICIs. This may lead to rational knowledge-based regimens tailored to individual cancer patients.
Collapse
Affiliation(s)
- MariaLuisa Vigano
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Lixing Wang
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Alia As’sadiq
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Suzanne Samarani
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Ali Ahmad
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Cecilia T. Costiniuk
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Infectious Diseases and Chronic Viral Illnesses Service, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
6
|
Lindenbergh PL, van der Stegen SJ. Adoptive Cell Therapy from the Dish: Potentiating Induced Pluripotent Stem Cells. Transfus Med Hemother 2025; 52:27-41. [PMID: 39944411 PMCID: PMC11813279 DOI: 10.1159/000540473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/19/2024] [Indexed: 02/16/2025] Open
Abstract
Background The clinical success of autologous adoptive cell therapy (ACT) is substantial but wide application is challenged by the quality and quantity of the patient's immune cells and the need for personalized manufacturing processes. Induced pluripotent stem cells (iPSCs) can be differentiated into immune effectors and thus provide an alternative, allogeneic cell source for ACT. Here, we compare iPSC-derived immune effectors to their PBMC-derived counterparts and review iPSC-derived ACT products currently under preclinical and clinical development. Summary iPSC-derived T cells, NK cells, macrophages, and neutrophils largely mimic their PBMC-derived counterparts in terms of cell-surface marker expression and cytotoxic effector functions. iPSC-derived immune effectors can be engineered with chimeric antigen receptors and other activating receptors to redirect their cytotoxic potential specifically to tumor-associated antigens (TAAs). However, several differences between iPSC- and PBMC-derived immune effectors remain and have inspired additional engineering strategies to enhance the antitumor capacity of iPSC-derived immune effectors. Key Messages iPSCs can be engineered to facilitate the generation of immune effectors with homogenous specificity for TAAs and enhanced effector functions. TAA-specific and functionally enhanced iPSC-derived T and NK cells are currently undergoing clinical evaluation in phase 1 trials. Engineered iPSC-derived macrophages and neutrophils are in preclinical development.
Collapse
Affiliation(s)
- Pieter L. Lindenbergh
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | | |
Collapse
|
7
|
Qu Y, Zeng A, Cheng Y, Li S. Natural killer cell memory: challenges and opportunities for cancer immunotherapy. Cancer Biol Ther 2024; 25:2376410. [PMID: 38987282 PMCID: PMC11238922 DOI: 10.1080/15384047.2024.2376410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Substantial advancements have been made in recent years in comprehending immune memory, which enhances the secondary response through prior infections. The ability of vertebrate T and B lymphocytes to exhibit classic recall responses has long been regarded as a distinguishing characteristic. However, natural killer (NK) cells have been found to acquire immunological memory in a manner akin to T and B cells. The fundamental principles derived from the investigation of NK cell memory offer novel insights into innate immunity and have the potential to pave the way for innovative strategies to enhance therapeutic interventions against multiple diseases including cancer. Here, we reviewed the fundamental characteristics, memory development and regulatory mechanism of NK cell memory. Moreover, we will conduct a comprehensive evaluation of the accomplishments, obstacles, and future direction pertaining to the utilization of NK cell memory in the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Yuhua Qu
- Department of Anorectal Surgery, Hospital of Chengdu University of Traditional Chinese Medicine and Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Anhui Zeng
- Department of Anorectal Surgery, Hospital of Chengdu University of Traditional Chinese Medicine and Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yulu Cheng
- Department of Disinfection Supply Center, Hospital of Chengdu University of Traditional Chinese Medicine and Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shengchun Li
- Department of Anorectal Surgery, Hospital of Chengdu University of Traditional Chinese Medicine and Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
8
|
Zhu A, Bai Y, Nan Y, Ju D. Natural killer cell engagers: From bi-specific to tri-specific and tetra-specific engagers for enhanced cancer immunotherapy. Clin Transl Med 2024; 14:e70046. [PMID: 39472273 PMCID: PMC11521791 DOI: 10.1002/ctm2.70046] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/25/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
Natural killer cell engagers (NKCEs) are a specialised subset of antibodies capable of simultaneously targeting endogenous NK cells and tumour cells, generating precise and effective cytolytic responses against cancer. This review systematically explores NK engagers as a rising star in NK-mediated immunotherapy, specifically focusing on multi-specific engagers. It examines the diverse configuration of NKCEs and how certain biologics could be employed to boost NK activity, including activating receptor engagement and cytokine incorporation. Some challenges and future perspectives of current NKCEs therapy are also discussed, including optimising pharmacokinetics, addressing the immunosuppressive tumour microenvironment and exploring potential combinatorial approaches. By offering an in-depth analysis of the current landscape and future trajectories of multi-specific NKCEs in cancer treatment, this review serves as a valuable resource for understanding this promising field of immunotherapy. HIGHLIGHTS Innovative NKCEs: NK cell engagers (NKCEs) represent a promising new class of immunotherapeutics targeting tumours by activating NK cells. Multi-specific formats: The transition from bi-specific to multi-specific NKCEs enhances their versatility and therapeutic efficacy. MECHANISMS OF ACTION NKCEs have the potential to improve NK cell activation by engaging activating receptors and incorporating cytokines. CLINICAL POTENTIAL Current clinical trials demonstrate the safety and efficacy of various NKCEs across different cancer types. Future research directions: Optimising NKCE designs and exploring combination therapies are essential for overcoming challenges in cancer treatment.
Collapse
Affiliation(s)
- An Zhu
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunotherapeuticsFudan University School of PharmacyShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
| | - Yu Bai
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunotherapeuticsFudan University School of PharmacyShanghaiChina
| | - Yanyang Nan
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunotherapeuticsFudan University School of PharmacyShanghaiChina
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunotherapeuticsFudan University School of PharmacyShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
| |
Collapse
|
9
|
Li Y, Li Z, Tang Y, Zhuang X, Feng W, Boor PPC, Buschow S, Sprengers D, Zhou G. Unlocking the therapeutic potential of the NKG2A-HLA-E immune checkpoint pathway in T cells and NK cells for cancer immunotherapy. J Immunother Cancer 2024; 12:e009934. [PMID: 39486805 PMCID: PMC11529472 DOI: 10.1136/jitc-2024-009934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/29/2024] [Indexed: 11/04/2024] Open
Abstract
Immune checkpoint blockade, which enhances the reactivity of T cells to eliminate cancer cells, has emerged as a potent strategy in cancer therapy. Besides T cells, natural killer (NK) cells also play an indispensable role in tumor surveillance and destruction. NK Group 2 family of receptor A (NKG2A), an emerging co-inhibitory immune checkpoint expressed on both NK cells and T cells, mediates inhibitory signal via interaction with its ligand human leukocyte antigen-E (HLA-E), thereby attenuating the effector and cytotoxic functions of NK cells and T cells. Developing antibodies to block NKG2A, holds promise in restoring the antitumor cytotoxicity of NK cells and T cells. In this review, we delve into the expression and functional significance of NKG2A and HLA-E, elucidating how the NKG2A-HLA-E axis contributes to tumor immune escape via signal transduction mechanisms. Furthermore, we provide an overview of clinical trials investigating NKG2A blockade, either as monotherapy or in combination with other therapeutic antibodies, highlighting the responses of the immune system and the clinical benefits for patients. We pay special attention to additional immune co-signaling molecules that serve as potential targets on both NK cells and T cells, aiming to evoke more robust immune responses against cancer. This review offers an in-depth exploration of the NKG2A-HLA-E pathway as a pivotal checkpoint in the anti-tumor responses, paving the way for new immunotherapeutic strategies to improve cancer patient outcomes.
Collapse
Affiliation(s)
- Yan Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Zhu Li
- Department of Dermatology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yisen Tang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiaomei Zhuang
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Wanhua Feng
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Patrick P C Boor
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Sonja Buschow
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Dave Sprengers
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Guoying Zhou
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|
10
|
Hurtado M, Khajavi L, Essabbar A, Kammer M, Xie T, Coullomb A, Pradines A, Casanova A, Kruczynski A, Gouin S, Clermont E, Boutillet L, Senosain MF, Zou Y, Zhao S, Burq P, Mahfoudi A, Besse J, Launay P, Passioukov A, Chetaille E, Favre G, Maldonado F, Cruzalegui F, Delfour O, Mazières J, Pancaldi V. Transcriptomics profiling of the non-small cell lung cancer microenvironment across disease stages reveals dual immune cell-type behaviors. Front Immunol 2024; 15:1394965. [PMID: 39606240 PMCID: PMC11600981 DOI: 10.3389/fimmu.2024.1394965] [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: 03/02/2024] [Accepted: 09/24/2024] [Indexed: 11/29/2024] Open
Abstract
Background Lung cancer is the leading cause of cancer death worldwide, with poor survival despite recent therapeutic advances. A better understanding of the complexity of the tumor microenvironment is needed to improve patients' outcome. Methods We applied a computational immunology approach (involving immune cell proportion estimation by deconvolution, transcription factor activity inference, pathways and immune scores estimations) in order to characterize bulk transcriptomics of 62 primary lung adenocarcinoma (LUAD) samples from patients across disease stages. Focusing specifically on early stage samples, we validated our findings using an independent LUAD cohort with 70 bulk RNAseq and 15 scRNAseq datasets and on TCGA datasets. Results Through our methodology and feature integration pipeline, we identified groups of immune cells related to disease stage as well as potential immune response or evasion and survival. More specifically, we reported a duality in the behavior of immune cells, notably natural killer (NK) cells, which was shown to be associated with survival and could be relevant for immune response or evasion. These distinct NK cell populations were further characterized using scRNAseq data, showing potential differences in their cytotoxic activity. Conclusion The dual profile of several immune cells, most notably T-cell populations, have been discussed in the context of diseases such as cancer. Here, we report the duality of NK cells which should be taken into account in conjunction with other immune cell populations and behaviors in predicting prognosis, immune response or evasion.
Collapse
Affiliation(s)
- Marcelo Hurtado
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
| | - Leila Khajavi
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
| | - Abdelmounim Essabbar
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
| | - Michael Kammer
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Ting Xie
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
| | - Alexis Coullomb
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
| | - Anne Pradines
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
- Laboratory Medicine, Oncopole Claudius Regaud, Toulouse, France
| | - Anne Casanova
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
- Laboratory Medicine, Oncopole Claudius Regaud, Toulouse, France
| | | | - Sandrine Gouin
- Pulmonology Department, Larrey Hospital, University Hospital of Toulouse, Toulouse, France
| | - Estelle Clermont
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
- Laboratory Medicine, Oncopole Claudius Regaud, Toulouse, France
| | - Léa Boutillet
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
| | - Maria Fernanda Senosain
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical. Center, Nashville, TN, United States
| | - Yong Zou
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical. Center, Nashville, TN, United States
| | - Shillin Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Prosper Burq
- Data Science, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | | | - Jerome Besse
- Institut de Recherche Pierre Fabre, Toulouse, France
| | - Pierre Launay
- Institut de Recherche Pierre Fabre, Toulouse, France
| | | | | | - Gilles Favre
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
| | - Fabien Maldonado
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | | | | | - Julien Mazières
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
- Pulmonology Department, Larrey Hospital, University Hospital of Toulouse, Toulouse, France
| | - Vera Pancaldi
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
- Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Spain
| |
Collapse
|
11
|
Salminen A. Inhibitory immune checkpoints suppress the surveillance of senescent cells promoting their accumulation with aging and in age-related diseases. Biogerontology 2024; 25:749-773. [PMID: 38954358 PMCID: PMC11374851 DOI: 10.1007/s10522-024-10114-w] [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: 04/19/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024]
Abstract
The accumulation of pro-inflammatory senescent cells within tissues is a common hallmark of the aging process and many age-related diseases. This modification has been called the senescence-associated secretory phenotype (SASP) and observed in cultured cells and in cells isolated from aged tissues. Currently, there is a debate whether the accumulation of senescent cells within tissues should be attributed to increased generation of senescent cells or to a defect in their elimination from aging tissues. Emerging studies have revealed that senescent cells display an increased expression of several inhibitory immune checkpoint ligands, especially those of the programmed cell death protein-1 (PD-1) ligand-1 (PD-L1) proteins. It is known that the PD-L1 ligands, especially those of cancer cells, target the PD-1 receptor of cytotoxic CD8+ T and natural killer (NK) cells disturbing their functions, e.g., evoking a decline in their cytotoxic activity and promoting their exhaustion and even apoptosis. An increase in the level of the PD-L1 protein in senescent cells was able to suppress their immune surveillance and inhibit their elimination by cytotoxic CD8+ T and NK cells. Senescent cells are known to express ligands for several inhibitory immune checkpoint receptors, i.e., PD-1, LILRB4, NKG2A, TIM-3, and SIRPα receptors. Here, I will briefly describe those pathways and examine whether these inhibitory checkpoints could be involved in the immune evasion of senescent cells with aging and age-related diseases. It seems plausible that an enhanced inhibitory checkpoint signaling can prevent the elimination of senescent cells from tissues and thus promote the aging process.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
12
|
Shimojima Y, Ichikawa T, Kishida D, Takamatsu R, Sekijima Y. HLA-E-expressing macrophage polarization and increased NKG2A/CD94 expression in adult-onset Still's disease. Immunol Res 2024; 72:1108-1119. [PMID: 38958876 DOI: 10.1007/s12026-024-09512-6] [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: 05/02/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024]
Abstract
We investigated the phenotypic characteristics of human leukocyte antigen (HLA)-E-expressing macrophages, NKG2A/CD94 expression in T and natural killer (NK) cells, and their interactions in patients with adult-onset Still's disease (AOSD). Peripheral blood mononuclear cells from 22 patients with AOSD and 22 healthy controls (HC) were used. Isolated monocytes were cultured first with macrophage colony-stimulating factor to differentiate into M0 macrophages and subsequently with lipopolysaccharide/interferon-γ or interleukin-4 to differentiate into M1 or M2 macrophages, respectively. HLA-E and NKG2A/CD94 expression levels were evaluated using quantitative RT-PCR and flow cytometry. HLA-E expression in M0 and M2 macrophages was significantly higher in patients with AOSD than in HC, and was positively correlated with serum C-reactive protein levels and erythrocyte sedimentation rate. NKG2A/CD94 expression in CD4 + and CD8 + T cells was significantly higher in patients with AOSD than in HC, but that in NK cells was not significantly different. In patients with AOSD, NKG2A expression in CD4 + T cells positively correlated with HLA-E expression in M0, M1, and M2 macrophages. CD94 expression in CD8 + T cells inversely correlated with HLA-E expression in M1 and M2 macrophages. NKG2A and CD94 expression in NK cells inversely correlated with HLA-E expression in M0, M1, and M2 macrophages. No significant correlation was observed between HLA-E and NKG2A/CD94 expression in HC. Increased expression of HLA-E in macrophages and NKG2A/CD94 in T cells can be observed in the inflammatory condition of AOSD. HLA-E-expressing macrophages may be associated with NKG2A/CD94 expression in T and NK cells with different correlations.
Collapse
Affiliation(s)
- Yasuhiro Shimojima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan.
| | - Takanori Ichikawa
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Dai Kishida
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Ryota Takamatsu
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Yoshiki Sekijima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| |
Collapse
|
13
|
Yang Y, Huang G, Lian J, Long C, Zhao B, Liu X, Zhang B, Ye W, Chen J, Du L, Jiang Z, Liu J, Zhang J, Hu C, Chen Q, Hong X. Circulating tumour cell clusters: isolation, biological significance and therapeutic implications. BMJ ONCOLOGY 2024; 3:e000437. [PMID: 39886139 PMCID: PMC11557725 DOI: 10.1136/bmjonc-2024-000437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/04/2024] [Indexed: 01/06/2025]
Abstract
Circulating tumour cells (CTCs) and CTC clusters are considered metastatic precursors due to their ability to seed distant metastasis. However, navigating the bloodstream presents a significant challenge for CTCs, as they must endure fluid shear forces and resist detachment-induced anoikis. Consequently, while a large number of cells from the primary tumour may enter the circulation, only a tiny fraction will result in metastasis. Nevertheless, the metastatic potency dramatically increases when CTCs travel in conjunction with other cell types to form CTC clusters, including neutrophils, myeloid-derived suppressor cells, macrophages, platelets, cancer-associated fibroblasts and red blood cells found in circulation. Such heterotypic CTC clustering events have been identified in a variety of cancer types and may serve as intriguing therapeutic targets and novel biomarkers for liquid biopsy. This review summarises recent advances in microfluidic technologies designed for the isolation of CTC clusters and explores the biological properties of distinct types of CTC clusters within the circulatory system. Investigation of the mechanisms of CTC cluster-blood microenvironment interactions may offer a promising avenue for gaining fresh insights into CTC cluster-mediated metastatic progression and reveal potential opportunities for devising personalised antimetastasis treatments.
Collapse
Affiliation(s)
- Yufan Yang
- Department of Biochemistry, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Guanyin Huang
- Department of Biochemistry, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jingru Lian
- Department of Biochemistry, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Chunhao Long
- Department of Biochemistry, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Boxi Zhao
- Department of Biochemistry, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xuefei Liu
- Department of Biochemistry, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Binyu Zhang
- Department of Biochemistry, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Weijian Ye
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Junhao Chen
- Department of Biochemistry, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Department of Dermatology, Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Longxiang Du
- Department of Biochemistry, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Zhuofeng Jiang
- Department of Biochemistry, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jialing Liu
- Department of Biochemistry, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jianglin Zhang
- Department of Dermatology, Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Chengzhi Hu
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Xin Hong
- Department of Biochemistry, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
14
|
Vietzen H, Simonitsch C, Friedel B, Berger SM, Kühner LM, Furlano PL, Florian DM, Görzer I, Koblischke M, Aberle JH, Puchhammer-Stöckl E. Torque teno viruses exhaust and imprint the human immune system via the HLA-E/NKG2A axis. Front Immunol 2024; 15:1447980. [PMID: 39295866 PMCID: PMC11408220 DOI: 10.3389/fimmu.2024.1447980] [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: 06/12/2024] [Accepted: 08/09/2024] [Indexed: 09/21/2024] Open
Abstract
The ubiquitous Torque teno virus (TTV) establishes a chronically persistent infection in the human host. TTV has not been associated with any apparent disease, but, as part of the human virome, it may confer a regulatory imprint on the human immune system with as yet unclear consequences. However, so far, only few studies have characterized the TTV-specific immune responses or the overall immunological imprints by TTV. Here, we reveal that TTV infection leads to a highly exhausted TTV-specific CD8+ T-cell response, hallmarked by decreased IFN-γ production and the expression of the inhibitory NKG2A-receptor. On a functional level, we identified a panel of highly polymorphic TTV-encoded peptides that lead to an expansion of regulatory NKG2A+ natural killer, NKG2A+CD4+, and NKG2A+CD8+ T cells via the stabilization of the non-classical HLA-E molecule. Our results thus demonstrate that TTV leads to a distinct imprint on the human immune system that may further regulate overall human immune responses in infectious, autoimmune, and malignant diseases.
Collapse
Affiliation(s)
- Hannes Vietzen
- Center for Virology, Medical University Vienna, Vienna, Austria
| | - Cara Simonitsch
- Center for Virology, Medical University Vienna, Vienna, Austria
| | | | - Sarah M Berger
- Center for Virology, Medical University Vienna, Vienna, Austria
| | - Laura M Kühner
- Center for Virology, Medical University Vienna, Vienna, Austria
| | | | - David M Florian
- Center for Virology, Medical University Vienna, Vienna, Austria
| | - Irene Görzer
- Center for Virology, Medical University Vienna, Vienna, Austria
| | | | - Judith H Aberle
- Center for Virology, Medical University Vienna, Vienna, Austria
| | | |
Collapse
|
15
|
Patin EC, Nenclares P, Chan Wah Hak C, Dillon MT, Patrikeev A, McLaughlin M, Grove L, Foo S, Soliman H, Barata JP, Marsden J, Baldock H, Gkantalis J, Roulstone V, Kyula J, Burley A, Hubbard L, Pedersen M, Smith SA, Clancy-Thompson E, Melcher AA, Ono M, Rullan A, Harrington KJ. Sculpting the tumour microenvironment by combining radiotherapy and ATR inhibition for curative-intent adjuvant immunotherapy. Nat Commun 2024; 15:6923. [PMID: 39134540 PMCID: PMC11319479 DOI: 10.1038/s41467-024-51236-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
The combination of radiotherapy/chemoradiotherapy and immune checkpoint blockade can result in poor outcomes in patients with locally advanced head and neck squamous cell carcinoma (HNSCC). Here, we show that combining ATR inhibition (ATRi) with radiotherapy (RT) increases the frequency of activated NKG2A+PD-1+ T cells in animal models of HNSCC. Compared with the ATRi/RT treatment regimen alone, the addition of simultaneous NKG2A and PD-L1 blockade to ATRi/RT, in the adjuvant, post-radiotherapy setting induces a robust antitumour response driven by higher infiltration and activation of cytotoxic T cells in the tumour microenvironment. The efficacy of this combination relies on CD40/CD40L costimulation and infiltration of activated, proliferating memory CD8+ and CD4+ T cells with persistent or new T cell receptor (TCR) signalling, respectively. We also observe increased richness in the TCR repertoire and emergence of numerous and large TCR clonotypes that cluster based on antigen specificity in response to NKG2A/PD-L1/ATRi/RT. Collectively, our data point towards potential combination approaches for the treatment of HNSCC.
Collapse
Affiliation(s)
- Emmanuel C Patin
- Targeted Therapy Team, The Institute of Cancer Research, London, UK.
| | - Pablo Nenclares
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Charleen Chan Wah Hak
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Magnus T Dillon
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Anton Patrikeev
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | - Lorna Grove
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Shane Foo
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | | | | | - Holly Baldock
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Jim Gkantalis
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | - Joan Kyula
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Amy Burley
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Lisa Hubbard
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Malin Pedersen
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | | | - Alan A Melcher
- Translational Immunotherapy Team, The Institute of Cancer Research, London, UK
| | - Masahiro Ono
- Department of Life Sciences, Imperial College London, London, UK
| | - Antonio Rullan
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Kevin J Harrington
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| |
Collapse
|
16
|
Mohammad Taheri M, Javan F, Poudineh M, Athari SS. Beyond CAR-T: The rise of CAR-NK cell therapy in asthma immunotherapy. J Transl Med 2024; 22:736. [PMID: 39103889 PMCID: PMC11302387 DOI: 10.1186/s12967-024-05534-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024] Open
Abstract
Asthma poses a major public health burden. While existing asthma drugs manage symptoms for many, some patients remain resistant. The lack of a cure, especially for severe asthma, compels exploration of novel therapies. Cancer immunotherapy successes with CAR-T cells suggest its potential for asthma treatment. Researchers are exploring various approaches for allergic diseases including membrane-bound IgE, IL-5, PD-L2, and CTLA-4 for asthma, and Dectin-1 for fungal asthma. NK cells offer several advantages over T cells for CAR-based immunotherapy. They offer key benefits: (1) HLA compatibility, meaning they can be used in a wider range of patients without the need for matching tissue types. (2) Minimal side effects (CRS and GVHD) due to their limited persistence and cytokine profile. (3) Scalability for "off-the-shelf" production from various sources. Several strategies have been introduced that highlight the superiority and challenges of CAR-NK cell therapy for asthma treatment including IL-10, IFN-γ, ADCC, perforin-granzyme, FASL, KIR, NCRs (NKP46), DAP, DNAM-1, TGF-β, TNF-α, CCL, NKG2A, TF, and EGFR. Furthermore, we advocate for incorporating AI for CAR design optimization and CRISPR-Cas9 gene editing technology for precise gene manipulation to generate highly effective CAR constructs. This review will delve into the evolution and production of CAR designs, explore pre-clinical and clinical studies of CAR-based therapies in asthma, analyze strategies to optimize CAR-NK cell function, conduct a comparative analysis of CAR-T and CAR-NK cell therapy with their respective challenges, and finally present established novel CAR designs with promising potential for asthma treatment.
Collapse
Affiliation(s)
| | - Fatemeh Javan
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohadeseh Poudineh
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Seyed Shamseddin Athari
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Immunology, Zanjan School of Medicine, Zanjan University of Medical Sciences, 12th Street, Shahrake Karmandan, Zanjan, 45139-561111, Iran.
| |
Collapse
|
17
|
Noviello M, De Lorenzo R, Chimienti R, Maugeri N, De Lalla C, Siracusano G, Lorè NI, Rancoita PMV, Cugnata F, Tassi E, Dispinseri S, Abbati D, Beretta V, Ruggiero E, Manfredi F, Merolla A, Cantarelli E, Tresoldi C, Pastori C, Caccia R, Sironi F, Marzinotto I, Saliu F, Ghezzi S, Lampasona V, Vicenzi E, Cinque P, Manfredi AA, Scarlatti G, Dellabona P, Lopalco L, Di Serio C, Malnati M, Ciceri F, Rovere-Querini P, Bonini C. The longitudinal characterization of immune responses in COVID-19 patients reveals novel prognostic signatures for disease severity, patients' survival and long COVID. Front Immunol 2024; 15:1381091. [PMID: 39136010 PMCID: PMC11317765 DOI: 10.3389/fimmu.2024.1381091] [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: 02/02/2024] [Accepted: 06/07/2024] [Indexed: 08/15/2024] Open
Abstract
Introduction SARS-CoV-2 pandemic still poses a significant burden on global health and economy, especially for symptoms persisting beyond the acute disease. COVID-19 manifests with various degrees of severity and the identification of early biomarkers capable of stratifying patient based on risk of progression could allow tailored treatments. Methods We longitudinally analyzed 67 patients, classified according to a WHO ordinal scale as having Mild, Moderate, or Severe COVID-19. Peripheral blood samples were prospectively collected at hospital admission and during a 6-month follow-up after discharge. Several subsets and markers of the innate and adaptive immunity were monitored as putative factors associated with COVID-19 symptoms. Results More than 50 immunological parameters were associated with disease severity. A decision tree including the main clinical, laboratory, and biological variables at admission identified low NK-cell precursors and CD14+CD91+ monocytes, and high CD8+ Effector Memory T cell frequencies as the most robust immunological correlates of COVID-19 severity and reduced survival. Moreover, low regulatory B-cell frequency at one month was associated with the susceptibility to develop long COVID at six months, likely due to their immunomodulatory ability. Discussion These results highlight the profound perturbation of the immune response during COVID-19. The evaluation of specific innate and adaptive immune-cell subsets allows to distinguish between different acute and persistent COVID-19 symptoms.
Collapse
Affiliation(s)
- Maddalena Noviello
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Cell Therapy Immunomonitoring Laboratory Monitoraggio Immunologico Terapie Cellulari (MITiCi), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Rebecca De Lorenzo
- Vita-Salute San Raffaele University, Milan, Italy
- Internal Medicine Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Raniero Chimienti
- Viral Evolution and Transmission Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Norma Maugeri
- Autoimmunity and Vascular Inflammation Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Claudia De Lalla
- Experimental Immunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Gabriel Siracusano
- Immunobiology of Human Immunodeficiency Virus (HIV) Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Nicola Ivan Lorè
- Emerging Bacterial Pathogens Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Paola Maria Vittoria Rancoita
- University Centre for Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy
| | - Federica Cugnata
- University Centre for Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy
| | - Elena Tassi
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Cell Therapy Immunomonitoring Laboratory Monitoraggio Immunologico Terapie Cellulari (MITiCi), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Stefania Dispinseri
- Viral Evolution and Transmission Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Danilo Abbati
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Valeria Beretta
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Cell Therapy Immunomonitoring Laboratory Monitoraggio Immunologico Terapie Cellulari (MITiCi), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Eliana Ruggiero
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Manfredi
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Aurora Merolla
- Vita-Salute San Raffaele University, Milan, Italy
- Internal Medicine Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Cantarelli
- Biological Resource Center Centro Risorse Biologiche-Ospedale San Raffaele (CRB-OSR), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Tresoldi
- Biological Resource Center Centro Risorse Biologiche-Ospedale San Raffaele (CRB-OSR), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Claudia Pastori
- Immunobiology of Human Immunodeficiency Virus (HIV) Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Caccia
- Neurovirology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Sironi
- Viral Evolution and Transmission Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Ilaria Marzinotto
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Fabio Saliu
- Emerging Bacterial Pathogens Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Ghezzi
- Viral Pathogenesis and Biosafety Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Vito Lampasona
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Elisa Vicenzi
- Viral Pathogenesis and Biosafety Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Paola Cinque
- Neurovirology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Angelo Andrea Manfredi
- Autoimmunity and Vascular Inflammation Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Dellabona
- Experimental Immunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Lopalco
- Immunobiology of Human Immunodeficiency Virus (HIV) Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Clelia Di Serio
- University Centre for Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy
| | - Mauro Malnati
- Viral Evolution and Transmission Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Ciceri
- Vita-Salute San Raffaele University, Milan, Italy
- Hematology and Bone Marrow Transplant Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Patrizia Rovere-Querini
- Vita-Salute San Raffaele University, Milan, Italy
- Internal Medicine Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Bonini
- Experimental Hematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Cell Therapy Immunomonitoring Laboratory Monitoraggio Immunologico Terapie Cellulari (MITiCi), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
18
|
Fert I, Douguet L, Vesin B, Moncoq F, Noirat A, Authié P, Ciret S, Le Chevalier F, Blanc C, Vitrenko Y, Charneau P, Majlessi L, Anna F. T-cell immunity induced and reshaped by an anti-HPV immuno-oncotherapeutic lentiviral vector. NPJ Vaccines 2024; 9:102. [PMID: 38858404 PMCID: PMC11164992 DOI: 10.1038/s41541-024-00894-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 05/23/2024] [Indexed: 06/12/2024] Open
Abstract
We recently developed an immuno-oncotherapy against human papillomavirus (HPV)-induced tumors based on a lentiviral vector encoding the Early E6 and E7 oncoproteins of HPV16 and HPV18 genotypes, namely "Lenti-HPV-07". The robust and long-lasting anti-tumor efficacy of Lenti-HPV-07 is dependent on CD8+ T-cell induction and remodeling of the tumor microenvironment. Here, we first established that anti-vector immunity induced by Lenti-HPV-07 prime has no impact on the efficacy of a homologous boost to amplify anti-HPV T-cell immunity. To longitudinally monitor the evolution of the T-cell repertoire generated after the prime, homologous or heterologous boost with Lenti-HPV-07, we tracked T-cell clonotypes by deep sequencing of T-Cell Receptor (TCR) variable β and α chain mRNA, applied to whole peripheral blood cells (PBL) and a T cell population specific of an immunodominant E7HPV16 epitope. We observed a hyper-expansion of clonotypes post prime, accompanied by increased frequencies of HPV-07-specific T cells. Additionally, there was a notable diversification of clonotypes post boost in whole PBL, but not in the E7HPV16-specific T cells. We then demonstrated that the effector functions of such Lenti-HPV-07-induced T cells synergize with anti-checkpoint inhibitory treatments by systemic administration of anti-TIM3 or anti-NKG2A monoclonal antibodies. While Lenti-HPV-07 is about to enter a Phase I/IIa clinical trial, these results will help better elucidate its mode of action in immunotherapy against established HPV-mediated malignancies.
Collapse
Affiliation(s)
- Ingrid Fert
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Laëtitia Douguet
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Benjamin Vesin
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Fanny Moncoq
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Amandine Noirat
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Pierre Authié
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Sylvain Ciret
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Fabien Le Chevalier
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Catherine Blanc
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Yakov Vitrenko
- Institut Pasteur, Université Paris Cité, Biomics Technology Platform, F-75015, Paris, France
| | - Pierre Charneau
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Laleh Majlessi
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France.
| | - François Anna
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France.
| |
Collapse
|
19
|
Riva E, Carboni S, di Berardino-Besson W, Moyat M, Belnoue E, Devy-Dimanche L, Rossi M. Bimodal Effect of NKG2A Blockade on Intratumoral and Systemic CD8 T Cell Response Induced by Cancer Vaccine. Cancers (Basel) 2024; 16:2036. [PMID: 38893156 PMCID: PMC11171001 DOI: 10.3390/cancers16112036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 05/22/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
Immune check-point blockade (ICB) has revitalized cancer immunotherapy, showing unprecedented efficacy despite only a narrow number of indications and with limited long-term protection. Cancer vaccines are promising combination partners for ICB to widen the patient population profiting from these treatments. Therapeutic heterologous prime-boost vaccination with KISIMATM protein vaccine and VSV-GP-TAg oncolytic virus was shown to inflame the tumor microenvironment, promoting significant infiltration of antigen-specific CD8 T cells resulting in robust antitumoral efficacy in mouse tumor models, and clinical trials are currently ongoing. Here, we report the impact of NKG2A blockade on antitumoral CD8 T cell immune response elicited by KISIMA-VSV-GP-TAg vaccination in tumor mouse models. Combination therapy significantly reduced the amount of vaccine-induced exhausted CD8 T cells infiltrating the tumor, resulting in short-term improved tumor growth control and prolonged mouse survival, while it also influenced the establishment of systemic effector memory CD8 T cell response. Taken together, these data show a compartment-dependent effect of NKG2A blockade on cancer vaccine-induced T cell immunity, increasing intratumoral T cell efficacy and attenuating the development of peripheral effector memory CD8 T cell response.
Collapse
Affiliation(s)
- Erika Riva
- Amal Therapeutics, Fondation Pour Recherches Médicales, Avenue de la Roseraie 64, 1205 Geneva, Switzerland
- Boehringer Ingelheim International GmbH, 55216 Ingelheim, Germany
| | - Susanna Carboni
- Amal Therapeutics, Fondation Pour Recherches Médicales, Avenue de la Roseraie 64, 1205 Geneva, Switzerland
- Boehringer Ingelheim International GmbH, 55216 Ingelheim, Germany
| | - Wilma di Berardino-Besson
- Amal Therapeutics, Fondation Pour Recherches Médicales, Avenue de la Roseraie 64, 1205 Geneva, Switzerland
- Boehringer Ingelheim International GmbH, 55216 Ingelheim, Germany
| | - Mati Moyat
- Amal Therapeutics, Fondation Pour Recherches Médicales, Avenue de la Roseraie 64, 1205 Geneva, Switzerland
- Boehringer Ingelheim International GmbH, 55216 Ingelheim, Germany
| | - Elodie Belnoue
- Amal Therapeutics, Fondation Pour Recherches Médicales, Avenue de la Roseraie 64, 1205 Geneva, Switzerland
- Boehringer Ingelheim International GmbH, 55216 Ingelheim, Germany
| | - Laetitia Devy-Dimanche
- Amal Therapeutics, Fondation Pour Recherches Médicales, Avenue de la Roseraie 64, 1205 Geneva, Switzerland
- Boehringer Ingelheim International GmbH, 55216 Ingelheim, Germany
| | - Matteo Rossi
- Amal Therapeutics, Fondation Pour Recherches Médicales, Avenue de la Roseraie 64, 1205 Geneva, Switzerland
- Boehringer Ingelheim International GmbH, 55216 Ingelheim, Germany
| |
Collapse
|
20
|
Stenger TD, Miller JS. Therapeutic approaches to enhance natural killer cell cytotoxicity. Front Immunol 2024; 15:1356666. [PMID: 38545115 PMCID: PMC10966407 DOI: 10.3389/fimmu.2024.1356666] [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: 12/16/2023] [Accepted: 02/05/2024] [Indexed: 04/14/2024] Open
Abstract
Enhancing the cytotoxicity of natural killer (NK) cells has emerged as a promising strategy in cancer immunotherapy, due to their pivotal role in immune surveillance and tumor clearance. This literature review provides a comprehensive overview of therapeutic approaches designed to augment NK cell cytotoxicity. We analyze a wide range of strategies, including cytokine-based treatment, monoclonal antibodies, and NK cell engagers, and discuss criteria that must be considered when selecting an NK cell product to combine with these strategies. Furthermore, we discuss the challenges and limitations associated with each therapeutic strategy, as well as the potential for combination therapies to maximize NK cell cytotoxicity while minimizing adverse effects. By exploring the wealth of research on this topic, this literature review aims to provide a comprehensive resource for researchers and clinicians seeking to develop and implement novel therapeutic strategies that harness the full potential of NK cells in the fight against cancer. Enhancing NK cell cytotoxicity holds great promise in the evolving landscape of immunotherapy, and this review serves as a roadmap for understanding the current state of the field and the future directions in NK cell-based therapies.
Collapse
Affiliation(s)
- Terran D. Stenger
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | | |
Collapse
|
21
|
Regmi M, Wang Y, Liu W, Dai Y, Liu S, Ma K, Lin G, Yang J, Liu H, Wu J, Yang C. From glioma gloom to immune bloom: unveiling novel immunotherapeutic paradigms-a review. J Exp Clin Cancer Res 2024; 43:47. [PMID: 38342925 PMCID: PMC10860318 DOI: 10.1186/s13046-024-02973-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/04/2024] [Indexed: 02/13/2024] Open
Abstract
In tumor therapeutics, the transition from conventional cytotoxic drugs to targeted molecular therapies, such as those targeting receptor tyrosine kinases, has been pivotal. Despite this progress, the clinical outcomes have remained modest, with glioblastoma patients' median survival stagnating at less than 15 months. This underscores the urgent need for more specialized treatment strategies. Our review delves into the progression toward immunomodulation in glioma treatment. We dissect critical discoveries in immunotherapy, such as spotlighting the instrumental role of tumor-associated macrophages, which account for approximately half of the immune cells in the glioma microenvironment, and myeloid-derived suppressor cells. The complex interplay between tumor cells and the immune microenvironment has been explored, revealing novel therapeutic targets. The uniqueness of our review is its exhaustive approach, synthesizing current research to elucidate the intricate roles of various molecules and receptors within the glioma microenvironment. This comprehensive synthesis not only maps the current landscape but also provides a blueprint for refining immunotherapy for glioma, signifying a paradigm shift toward leveraging immune mechanisms for improved patient prognosis.
Collapse
Affiliation(s)
- Moksada Regmi
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Peking University, Beijing, 100191, China
- Peking University Health Science Center, Beijing, 100191, China
- Henan Academy of Innovations in Medical Science (AIMS), Zhengzhou, 450003, China
| | - Yingjie Wang
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Peking University, Beijing, 100191, China
| | - Weihai Liu
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Peking University, Beijing, 100191, China
- Peking University Health Science Center, Beijing, 100191, China
| | - Yuwei Dai
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Peking University, Beijing, 100191, China
- Peking University Health Science Center, Beijing, 100191, China
| | - Shikun Liu
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Peking University, Beijing, 100191, China
- Peking University Health Science Center, Beijing, 100191, China
| | - Ke Ma
- Peking University Health Science Center, Beijing, 100191, China
| | - Guozhong Lin
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Peking University, Beijing, 100191, China
| | - Jun Yang
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Peking University, Beijing, 100191, China
| | - Hongyi Liu
- Henan Academy of Innovations in Medical Science (AIMS), Zhengzhou, 450003, China
- National Engineering Research Center for Ophthalmology, Beijing, 100730, China
- Engineering Research Center of Ophthalmic Equipment and Materials, Ministry of Education, Beijing, 100730, China
- Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, 100730, China
| | - Jian Wu
- Henan Academy of Innovations in Medical Science (AIMS), Zhengzhou, 450003, China.
- National Engineering Research Center for Ophthalmology, Beijing, 100730, China.
- Engineering Research Center of Ophthalmic Equipment and Materials, Ministry of Education, Beijing, 100730, China.
- Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, 100730, China.
| | - Chenlong Yang
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, 100191, China.
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Peking University, Beijing, 100191, China.
- Henan Academy of Innovations in Medical Science (AIMS), Zhengzhou, 450003, China.
| |
Collapse
|
22
|
Lin Z, Zou S, Wen K. The crosstalk of CD8+ T cells and ferroptosis in cancer. Front Immunol 2024; 14:1255443. [PMID: 38288118 PMCID: PMC10822999 DOI: 10.3389/fimmu.2023.1255443] [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: 07/08/2023] [Accepted: 12/28/2023] [Indexed: 01/31/2024] Open
Abstract
Ferroptosis is an iron-dependent, novel form of programmed cell death characterized by lipid peroxidation and glutathione depletion and is widespread in a variety of diseases. CD8+ T cells are the most important effector cells of cytotoxic T cells, capable of specifically recognizing and killing cancer cells. Traditionally, CD8+ T cells are thought to induce cancer cell death mainly through perforin and granzyme, and Fas-L/Fas binding. In recent years, CD8+ T cell-derived IFN-γ was found to promote cancer cell ferroptosis by multiple mechanisms, including upregulation of IRF1 and IRF8, and downregulation of the system XC-, while cancer cells ferroptosis was shown to enhance the anti-tumor effects of CD8+ T cell by heating the tumor immune microenvironment through the exposure and release of tumor-associated specific antigens, which results in a positive feedback pathway. Unfortunately, the intra-tumoral CD8+ T cells are more sensitive to ferroptosis than cancer cells, which limits the application of ferroptosis inducers in cancer. In addition, CD8+ T cells are susceptible to being regulated by other immune cell ferroptosis in the TME, such as tumor-associated macrophages, dendritic cells, Treg, and bone marrow-derived immunosuppressive cells. Together, these factors build a complex network of CD8+ T cells and ferroptosis in cancer. Therefore, we aim to integrate relevant studies to reveal the potential mechanisms of crosstalk between CD8+ T cells and ferroptosis, and to summarize preclinical models in cancer therapy to find new therapeutic strategies in this review.
Collapse
Affiliation(s)
- Zhengjun Lin
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- The First People's Hospital of Zunyi, The Third Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Songzhu Zou
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Kunming Wen
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
23
|
Patwekar M, Sehar N, Patwekar F, Medikeri A, Ali S, Aldossri RM, Rehman MU. Novel immune checkpoint targets: A promising therapy for cancer treatments. Int Immunopharmacol 2024; 126:111186. [PMID: 37979454 DOI: 10.1016/j.intimp.2023.111186] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/20/2023]
Abstract
The immune system frequently comprises immunological checkpoints. They serve as a barrier to keep the immune system from overreacting and damaging cells that are robust. Immune checkpoint inhibitors (ICIs) are utilized in immunotherapy to prevent the synergy of partner proteins of checkpoint proteins with auxiliary proteins. Moreover, the T cells may target malignant cells since the "off" signal cannot be conveyed. ICIs, which are mostly composed of monoclonal antibodies (mAbs) against cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and anti- programmed death-1/programmed ligand 1 (anti-PD-1/PD-L1), might transform the context of cancer therapy. Further, more patients continued to exhibit adaptive resistance, even though several ICIs demonstrated convincing therapeutic benefits in selective tumor types. Immune checkpoint therapy's overall effectiveness is still lacking at this time. A popular area of study involves investigating additional immune checkpoint molecules. Recent research has found a number of fresh immune checkpoint targets, including NKG2A ligands, TIGIT, B7-H6 ligands, Galectin 3, TIM3, and so on. These targets have been focus of the study, and recent investigational approaches have shown encouraging outcomes. In this review article, we covered the development and present level understanding of these recently identified immune checkpoint molecules, its effectiveness and limitations.
Collapse
Affiliation(s)
| | - Nouroz Sehar
- Centre for Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard University, New Delhi, 110062, India
| | - Faheem Patwekar
- Luqman College of Pharmacy, Gulbarga, 585102, Karnataka, India
| | | | - Shafat Ali
- Cytogenetics and Molecular Biology Laboratory, Centre of Research for Development, University of Kashmir, Srinagar, 190006, Jammu and Kashmir, India.
| | - Rana M Aldossri
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Muneeb U Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
24
|
Martínez-Domínguez SJ, García-Mateo S, Sainz-Arnal P, Martínez-García J, Gallego-Llera B, Lozano-Limones MJ, Hidalgo S, Gargallo-Puyuelo CJ, Latre-Santos M, Nocito-Colon MML, Martínez-Lostao L, Refaie E, Arroyo-Villarino MT, Del Rio-Nechaevsky M, Ramirez-Labrada A, Pardo J, Gomollón F, Baptista PM. Unravelling the cellular response to the SARS-COV-2 vaccine in inflammatory bowel disease patients on biologic drugs. Sci Rep 2023; 13:23061. [PMID: 38155275 PMCID: PMC10754931 DOI: 10.1038/s41598-023-50537-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 12/21/2023] [Indexed: 12/30/2023] Open
Abstract
Suboptimal vaccine response is a significant concern in patients with Inflammatory Bowel Disease (IBD) receiving biologic drugs. This single-center observational study involved 754 patients with IBD. In Phase I (October 2020-April 2021), 754 IBD participants who had not previously received the SARS-CoV-2 vaccine, underwent blood extraction to assess the seroprevalence of SARS-CoV-2 infection and IBD-related factors. Phase II (May 2021-October 2021) included a subgroup of 52 IBD participants with confirmed previous SARS-CoV-2 infection, who were studied for humoral and cellular response to the SARS-CoV-2 vaccine. In Phase I, treatment with anti-TNF was associated with lower rates of seroconversion (aOR 0.25 95% CI [0.10-0.61]). In Phase II, a significant increase in post-vaccination IgG levels was observed regardless of biologic treatment. However, patients treated with anti-TNF exhibited significantly lower IgG levels compared to those without IBD therapy (5.32 ± 2.47 vs. 7.99 ± 2.59 U/ml, p = 0.042). Following vaccination, a lymphocyte, monocyte, and NK cell activation pattern was observed, with no significant differences between patients receiving biologic drugs and those without IBD treatment. Despite lower seroprevalence and humoral response to the SARS-CoV-2 vaccine in patients treated with anti-TNF, the cellular response to the vaccine did not differ significantly from that patients without IBD therapy.
Collapse
Affiliation(s)
- Samuel J Martínez-Domínguez
- Digestive Diseases Department, University Hospital "Lozano Blesa", Av. San Juan Bosco, nº 15. PC: 50009, Zaragoza, Spain.
- Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain.
- University of Zaragoza, Zaragoza, Spain.
| | - Sandra García-Mateo
- Digestive Diseases Department, University Hospital "Lozano Blesa", Av. San Juan Bosco, nº 15. PC: 50009, Zaragoza, Spain
- Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain
- University of Zaragoza, Zaragoza, Spain
| | | | - Javier Martínez-García
- Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain
- University of Zaragoza, Zaragoza, Spain
| | | | | | - Sandra Hidalgo
- Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain
- University of Zaragoza, Zaragoza, Spain
| | - Carla J Gargallo-Puyuelo
- Digestive Diseases Department, University Hospital "Lozano Blesa", Av. San Juan Bosco, nº 15. PC: 50009, Zaragoza, Spain
- Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain
- University of Zaragoza, Zaragoza, Spain
| | - Marta Latre-Santos
- Digestive Diseases Department, University Hospital "Lozano Blesa", Av. San Juan Bosco, nº 15. PC: 50009, Zaragoza, Spain
| | | | - Luis Martínez-Lostao
- Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain
- University of Zaragoza, Zaragoza, Spain
- Immunology Department, University Hospital "Lozano Blesa", Zaragoza, Spain
- Institute of Nanoscience and Material of Aragón (INMA), Zaragoza, Spain
| | - Engy Refaie
- Scuola di Specializzazione in Chirurgia Generale, Università Degli Studi di Pavia Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Maria Teresa Arroyo-Villarino
- Digestive Diseases Department, University Hospital "Lozano Blesa", Av. San Juan Bosco, nº 15. PC: 50009, Zaragoza, Spain
- Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain
- University of Zaragoza, Zaragoza, Spain
| | - Marcela Del Rio-Nechaevsky
- CIBEREnfermedadesRaras (CIBERER), Madrid, Spain
- Biomedical Engineering Department, Universidad Carlos III de Madrid, Madrid, Spain
- IIS-Fundación Jiménez Díaz, Madrid, Spain
| | - Ariel Ramirez-Labrada
- Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain
- University of Zaragoza, Zaragoza, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Julián Pardo
- Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain
- University of Zaragoza, Zaragoza, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Fernando Gomollón
- Digestive Diseases Department, University Hospital "Lozano Blesa", Av. San Juan Bosco, nº 15. PC: 50009, Zaragoza, Spain
- Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain
- University of Zaragoza, Zaragoza, Spain
- CIBER Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Pedro M Baptista
- Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain
- Biomedical Engineering Department, Universidad Carlos III de Madrid, Madrid, Spain
- CIBER Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Fundación ARAID, Zaragoza, Spain
| |
Collapse
|
25
|
Vietzen H, Berger SM, Kühner LM, Furlano PL, Bsteh G, Berger T, Rommer P, Puchhammer-Stöckl E. Ineffective control of Epstein-Barr-virus-induced autoimmunity increases the risk for multiple sclerosis. Cell 2023; 186:5705-5718.e13. [PMID: 38091993 DOI: 10.1016/j.cell.2023.11.015] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/12/2023] [Accepted: 11/09/2023] [Indexed: 12/24/2023]
Abstract
Multiple sclerosis (MS) is a demyelinating disease of the CNS. Epstein-Barr virus (EBV) contributes to the MS pathogenesis because high levels of EBV EBNA386-405-specific antibodies cross react with the CNS-derived GlialCAM370-389. However, it is unclear why only some individuals with such high autoreactive antibody titers develop MS. Here, we show that autoreactive cells are eliminated by distinct immune responses, which are determined by genetic variations of the host, as well as of the infecting EBV and human cytomegalovirus (HCMV). We demonstrate that potent cytotoxic NKG2C+ and NKG2D+ natural killer (NK) cells and distinct EBV-specific T cell responses kill autoreactive GlialCAM370-389-specific cells. Furthermore, immune evasion of these autoreactive cells was induced by EBV-variant-specific upregulation of the immunomodulatory HLA-E. These defined virus and host genetic pre-dispositions are associated with an up to 260-fold increased risk of MS. Our findings thus allow the early identification of patients at risk for MS and suggest additional therapeutic options against MS.
Collapse
Affiliation(s)
- Hannes Vietzen
- Center for Virology, Medical University of Vienna, Vienna, Austria.
| | - Sarah M Berger
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Laura M Kühner
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | | | - Gabriel Bsteh
- Department of Neurology, Medical University of Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Paulus Rommer
- Department of Neurology, Medical University of Vienna, Vienna, Austria; Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
26
|
Good E, Åkerman L, Nyström S, Jonasson L, Ernerudh J, de Muinck E. Changes in natural killer and T lymphocyte phenotypes in response to cardiovascular risk management. Sci Rep 2023; 13:20810. [PMID: 38012327 PMCID: PMC10682417 DOI: 10.1038/s41598-023-48111-7] [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: 12/27/2022] [Accepted: 11/22/2023] [Indexed: 11/29/2023] Open
Abstract
The pro-inflammatory and regulatory roles of T lymphocytes in atherosclerosis are well established but less is known about natural killer (NK) cells and natural killer T (NKT)-like cells. The effects of cardiovascular risk management on the phenotypes of these cells are unknown. To assess changes in NK cell and lymphocyte phenotypes and circulating inflammatory proteins in response to cardiovascular risk management in patients with carotid atherosclerosis. Fifty patients were included in a prospective clinical study. Measurements were at baseline and after 12 months of cardiovascular risk management. Circulating NK, NKT-like and T lymphocyte subpopulations were phenotyped by multi-colour flow cytometry. Proximity extension assay was performed for 176 plasma proteins associated with inflammation and cardiovascular disease. At 12 months there were significant reductions in LDL (P = 0.001) and blood pressure (P = 0.028). NK cells responded with a reduction in pro-inflammatory (NKG2C+) cells (P = 0.0003), an increase in anti-inflammatory (NKG2A+) cells (P = 0.032), and a reduction in terminally differentiated (CD57+) NK cells. NKT-like cells showed a similar decrease in terminally differentiated subpopulations (P = 0.000002). Subpopulations of T helper cells exhibited a significant reduction in central memory (P = 1.09 × 10-8) and a significant increase in CD4+ naïve- (P = 0.0008) and effector memory T cells (P = 0.006). The protein analysis indicated that cardiovascular risk management affects proteins involved in the inflammatory NF-κB pathway. The consistent decrease in senescent phenotypes of NK, NKT-like and CD4+ cells with a concomitant increase in more naïve, phenotypes suggests a change towards a less pro-inflammatory lymphocyte profile in response to cardiovascular risk management.Trial registry name: CARotid MRI of Atherosclerosis (CARMA). ClinicalTrials.gov identifier NCT04835571 (08/04/2021). https://www.clinicaltrials.gov/study/NCT04835571 .
Collapse
Affiliation(s)
- Elin Good
- Department of Cardiology in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.
| | - Linda Åkerman
- Department of Clinical Immunology and Transfusion Medicine, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Sofia Nyström
- Department of Clinical Immunology and Transfusion Medicine, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Lena Jonasson
- Department of Cardiology in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Jan Ernerudh
- Department of Clinical Immunology and Transfusion Medicine, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ebo de Muinck
- Department of Cardiology in Linköping, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
27
|
Torki E, Gharezade A, Doroudchi M, Sheikhi S, Mansury D, Sullman MJM, Fouladseresht H. The kinetics of inhibitory immune checkpoints during and post-COVID-19: the knowns and unknowns. Clin Exp Med 2023; 23:3299-3319. [PMID: 37697158 DOI: 10.1007/s10238-023-01188-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/31/2023] [Indexed: 09/13/2023]
Abstract
The immune system is tightly regulated to prevent immune reactions to self-antigens and to avoid excessive immune responses during and after challenges from non-self-antigens. Inhibitory immune checkpoints (IICPs), as the major regulators of immune system responses, are extremely important for maintaining the homeostasis of cells and tissues. However, the high and sustained co-expression of IICPs in chronic infections, under persistent antigenic stimulations, results in reduced immune cell functioning and more severe and prolonged disease complications. Furthermore, IICPs-mediated interactions can be hijacked by pathogens in order to evade immune induction or effector mechanisms. Therefore, IICPs can be potential targets for the prognosis and treatment of chronic infectious diseases. This is especially the case with regards to the most challenging infectious disease of recent times, coronavirus disease-2019 (COVID-19), whose long-term complications can persist long after recovery. This article reviews the current knowledge about the kinetics and functioning of the IICPs during and post-COVID-19.
Collapse
Affiliation(s)
- Ensiye Torki
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arezou Gharezade
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehrnoosh Doroudchi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shima Sheikhi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Davood Mansury
- Department of Microbiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mark J M Sullman
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Social Sciences, University of Nicosia, Nicosia, Cyprus
| | - Hamed Fouladseresht
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
28
|
Bexte T, Reindl LM, Ullrich E. Nonviral technologies can pave the way for CAR-NK cell therapy. J Leukoc Biol 2023; 114:475-486. [PMID: 37403203 DOI: 10.1093/jleuko/qiad074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 05/25/2023] [Accepted: 06/22/2023] [Indexed: 07/06/2023] Open
Abstract
Natural killer cells are a promising platform for cancer immunotherapy. Natural killer cells have high intrinsic killing capability, and the insertion of a chimeric antigen receptor can further enhance their antitumor potential. In first-in-human trials, chimeric antigen receptor-natural killer cells demonstrated strong clinical activity without therapy-induced side effects. The applicability of natural killer cells as an "off-the-shelf" product makes them highly attractive for gene-engineered cell therapies. Traditionally, viral transduction has been used for gene editing; however, the use of viral vectors remains a safety concern and is associated with high costs and regulatory requirements. Here, we review the current landscape of nonviral approaches for chimeric antigen receptor-natural killer cell generation. This includes transfection of vector particles and electroporation of mRNA and DNA vectors, resulting in transient modification and chimeric antigen receptor expression. In addition, using nonviral transposon technologies, natural killer cells can be stably modified ensuring long-lasting chimeric antigen receptor expression. Finally, we discuss CRISPR/Cas9 tools to edit key genes for natural killer cell functionality.
Collapse
Affiliation(s)
- Tobias Bexte
- Goethe University Frankfurt, Department of Pediatrics, Experimental Immunology & Cell Therapy, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Paul-Ehrlich-Straße 42-44, 60596 Frankfurt am Main, Germany
- University Cancer Center (UCT), Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Mildred Scheel Career Center (MSNZ), Hospital of the Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Lisa Marie Reindl
- Goethe University Frankfurt, Department of Pediatrics, Experimental Immunology & Cell Therapy, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Paul-Ehrlich-Straße 42-44, 60596 Frankfurt am Main, Germany
| | - Evelyn Ullrich
- Goethe University Frankfurt, Department of Pediatrics, Experimental Immunology & Cell Therapy, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Paul-Ehrlich-Straße 42-44, 60596 Frankfurt am Main, Germany
- University Cancer Center (UCT), Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Mildred Scheel Career Center (MSNZ), Hospital of the Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Frankfurt/Mainz; Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| |
Collapse
|
29
|
Bjorgen JC, Dick JK, Cromarty R, Hart GT, Rhein J. NK cell subsets and dysfunction during viral infection: a new avenue for therapeutics? Front Immunol 2023; 14:1267774. [PMID: 37928543 PMCID: PMC10620977 DOI: 10.3389/fimmu.2023.1267774] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/25/2023] [Indexed: 11/07/2023] Open
Abstract
In the setting of viral challenge, natural killer (NK) cells play an important role as an early immune responder against infection. During this response, significant changes in the NK cell population occur, particularly in terms of their frequency, location, and subtype prevalence. In this review, changes in the NK cell repertoire associated with several pathogenic viral infections are summarized, with a particular focus placed on changes that contribute to NK cell dysregulation in these settings. This dysregulation, in turn, can contribute to host pathology either by causing NK cells to be hyperresponsive or hyporesponsive. Hyperresponsive NK cells mediate significant host cell death and contribute to generating a hyperinflammatory environment. Hyporesponsive NK cell populations shift toward exhaustion and often fail to limit viral pathogenesis, possibly enabling viral persistence. Several emerging therapeutic approaches aimed at addressing NK cell dysregulation have arisen in the last three decades in the setting of cancer and may prove to hold promise in treating viral diseases. However, the application of such therapeutics to treat viral infections remains critically underexplored. This review briefly explores several therapeutic approaches, including the administration of TGF-β inhibitors, immune checkpoint inhibitors, adoptive NK cell therapies, CAR NK cells, and NK cell engagers among other therapeutics.
Collapse
Affiliation(s)
- Jacob C. Bjorgen
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Jenna K. Dick
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Ross Cromarty
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Geoffrey T. Hart
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Joshua Rhein
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
30
|
Graydon EK, Conner TL, Dunham K, Olsen C, Goguet E, Coggins SA, Rekedal M, Samuels E, Jackson-Thompson B, Moser M, Lindrose A, Hollis-Perry M, Wang G, Maiolatesi S, Alcorta Y, Reyes A, Wong M, Ramsey K, Davies J, Parmelee E, Ortega O, Sanchez M, Moller S, Inglefield J, Tribble D, Burgess T, O’Connell R, Malloy AMW, Pollett S, Broder CC, Laing ED, Anderson SK, Mitre E. Natural killer cells and BNT162b2 mRNA vaccine reactogenicity and durability. Front Immunol 2023; 14:1225025. [PMID: 37711632 PMCID: PMC10497936 DOI: 10.3389/fimmu.2023.1225025] [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: 05/18/2023] [Accepted: 07/27/2023] [Indexed: 09/16/2023] Open
Abstract
Introduction Natural killer (NK) cells can both amplify and regulate immune responses to vaccination. Studies in humans and animals have observed NK cell activation within days after mRNA vaccination. In this study, we sought to determine if baseline NK cell frequencies, phenotype, or function correlate with antibody responses or inflammatory side effects induced by the Pfizer-BioNTech COVID-19 vaccine (BNT162b2). Methods We analyzed serum and peripheral blood mononuclear cells (PBMCs) from 188 participants in the Prospective Assessment of SARS-CoV-2 Seroconversion study, an observational study evaluating immune responses in healthcare workers. Baseline serum samples and PBMCs were collected from all participants prior to any SARS-CoV-2 infection or vaccination. Spike-specific IgG antibodies were quantified at one and six months post-vaccination by microsphere-based multiplex immunoassay. NK cell frequencies and phenotypes were assessed on pre-vaccination PBMCs from all participants by multi-color flow cytometry, and on a subset of participants at time points after the 1st and 2nd doses of BNT162b2. Inflammatory side effects were assessed by structured symptom questionnaires, and baseline NK cell functionality was quantified by an in vitro killing assay on participants that reported high or low post-vaccination symptom scores. Results Key observations include: 1) circulating NK cells exhibit evidence of activation in the week following vaccination, 2) individuals with high symptom scores after 1st vaccination had higher pre-vaccination NK cytotoxicity indices, 3) high pre-vaccination NK cell numbers were associated with lower spike-specific IgG levels six months after two BNT162b2 doses, and 4) expression of the inhibitory marker NKG2A on immature NK cells was associated with higher antibody responses 1 and 6 months post-vaccination. Discussion These results suggest that NK cell activation by BNT162b2 vaccination may contribute to vaccine-induced inflammatory symptoms and reduce durability of vaccine-induced antibody responses.
Collapse
Affiliation(s)
- Elizabeth K. Graydon
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Tonia L. Conner
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
| | - Kim Dunham
- Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Cara Olsen
- Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Emilie Goguet
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Si’Ana A. Coggins
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Marana Rekedal
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Emily Samuels
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Belinda Jackson-Thompson
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Matthew Moser
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Alyssa Lindrose
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Monique Hollis-Perry
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center (NMRC), Silver Spring, MD, United States
| | - Gregory Wang
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center (NMRC), Silver Spring, MD, United States
- General Dynamics Information Technology, Silver Spring, MD, United States
| | - Santina Maiolatesi
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center (NMRC), Silver Spring, MD, United States
| | - Yolanda Alcorta
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center (NMRC), Silver Spring, MD, United States
- General Dynamics Information Technology, Silver Spring, MD, United States
| | - Anatalio Reyes
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center (NMRC), Silver Spring, MD, United States
- General Dynamics Information Technology, Silver Spring, MD, United States
| | - Mimi Wong
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center (NMRC), Silver Spring, MD, United States
- General Dynamics Information Technology, Silver Spring, MD, United States
| | - Kathy Ramsey
- Clinical Trials Center, Infectious Diseases Directorate, Naval Medical Research Center (NMRC), Silver Spring, MD, United States
- General Dynamics Information Technology, Silver Spring, MD, United States
| | - Julian Davies
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Edward Parmelee
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Orlando Ortega
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Mimi Sanchez
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Sydney Moller
- Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Jon Inglefield
- Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - David Tribble
- Infectious Disease Clinical Research Program, Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Timothy Burgess
- Infectious Disease Clinical Research Program, Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Robert O’Connell
- Infectious Disease Clinical Research Program, Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Allison M. W. Malloy
- Department of Pediatrics, Uniformed Services University, Bethesda, MD, United States
| | - Simon Pollett
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Infectious Disease Clinical Research Program, Department of Preventive Medicine & Biostatistics, Uniformed Services University, Bethesda, MD, United States
| | - Christopher C. Broder
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
| | - Eric D. Laing
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
| | - Stephen K. Anderson
- Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Edward Mitre
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
| |
Collapse
|
31
|
Kent A, Crump LS, Davila E. Beyond αβ T cells: NK, iNKT, and γδT cell biology in leukemic patients and potential for off-the-shelf adoptive cell therapies for AML. Front Immunol 2023; 14:1202950. [PMID: 37654497 PMCID: PMC10465706 DOI: 10.3389/fimmu.2023.1202950] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/24/2023] [Indexed: 09/02/2023] Open
Abstract
Acute myeloid leukemia (AML) remains an elusive disease to treat, let alone cure, even after highly intensive therapies such as stem cell transplants. Adoptive cell therapeutic strategies based on conventional alpha beta (αβ)T cells are an active area of research in myeloid neoplasms given their remarkable success in other hematologic malignancies, particularly B-cell-derived acute lymphoid leukemia, myeloma, and lymphomas. Several limitations have hindered clinical application of adoptive cell therapies in AML including lack of leukemia-specific antigens, on-target-off-leukemic toxicity, immunosuppressive microenvironments, and leukemic stem cell populations elusive to immune recognition and destruction. While there are promising T cell-based therapies including chimeric antigen receptor (CAR)-T designs under development, other cytotoxic lymphocyte cell subsets have unique phenotypes and capabilities that might be of additional benefit in AML treatment. Of particular interest are the natural killer (NK) and unconventional T cells known as invariant natural killer T (iNKT) and gamma delta (γδ) T cells. NK, iNKT, and γδT cells exhibit intrinsic anti-malignant properties, potential for alloreactivity, and human leukocyte-antigen (HLA)-independent function. Here we review the biology of each of these unconventional cytotoxic lymphocyte cell types and compare and contrast their strengths and limitations as the basis for adoptive cell therapies for AML.
Collapse
Affiliation(s)
- Andrew Kent
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative, University of Colorado, Aurora, CO, United States
- Department of Medicine, University of Colorado Comprehensive Cancer Center, Aurora, CO, United States
| | | | - Eduardo Davila
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative, University of Colorado, Aurora, CO, United States
- Department of Medicine, University of Colorado Comprehensive Cancer Center, Aurora, CO, United States
- Department of Medicine, University of Colorado, Aurora, CO, United States
| |
Collapse
|
32
|
Hui J, Zhou M, An G, Zhang H, Lu Y, Wang X, Zhao X. Regulatory role of exosomes in colorectal cancer progression and potential as biomarkers. Cancer Biol Med 2023; 20:j.issn.2095-3941.2023.0119. [PMID: 37553810 PMCID: PMC10476469 DOI: 10.20892/j.issn.2095-3941.2023.0119] [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: 04/12/2023] [Accepted: 06/29/2023] [Indexed: 08/10/2023] Open
Abstract
Colorectal cancer (CRC) remains an enormous challenge to human health worldwide. Unfortunately, the mechanism underlying CRC progression is not well understood. Mounting evidence has confirmed that exosomes play a vital role in CRC progression, which has attracted extensive attention among researchers. In addition to acting as messengers between CRC cells, exosomes also participate in the CRC immunomodulatory process and reshape immune function. As stable message carriers and liquid biopsy option under development, exosomes are promising biomarkers in the diagnosis or treatment of CRC. In this review we have described and analyzed the biogenesis and release of exosomes and current research on the role of exosomes in immune regulation and metastasis of CRC. Moreover, we have discussed candidate exosomal molecules as potential biomarkers to diagnose CRC, predict CRC progression, or determine CRC chemoresistance, and described the significance of exosomes in the immunotherapy of CRC. This review provides insight to further understand the role of exosomes in CRC progression and identify valuable biomarkers that facilitate the clinical management of CRC patients.
Collapse
Affiliation(s)
- Juan Hui
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi’an 710038, China
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi’an 710032, China
| | - Mingzhen Zhou
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi’an 710032, China
| | - Guangzhou An
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi’an 710038, China
- Department of Radiation Protection Medicine, Ministry of Education Key Laboratory of Hazard Assessment and Control in Special Operational Environment, Faculty of Preventive Medicine, Air Force Medical University, Xi’an 710032, China
| | - Hui Zhang
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi’an 710038, China
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University, Xi’an 710038, China
| | - Yuanyuan Lu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi’an 710032, China
| | - Xin Wang
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi’an 710038, China
| | - Xiaodi Zhao
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi’an 710032, China
| |
Collapse
|
33
|
Pan S, Guan J, Xianyu B, Tan Y, Li T, Xu H. A Nanotherapeutic Strategy to Reverse NK Cell Exhaustion. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2211370. [PMID: 36917826 DOI: 10.1002/adma.202211370] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/10/2023] [Indexed: 06/09/2023]
Abstract
As a specialized immune effector cell, natural killer (NK) cells play a very important role in immunotherapy, but tumor immunosuppression caused by abnormal expression of cancer cells seriously weakens its therapeutic effect and leads to exhaustion. Here, self-assembled selenium-containing nanoparticles (NPs) composed of cetuximab, C5SeSeC5, and inhibitor LY345899 are developed to reverse NK cell exhaustion. The obtained NPs can target epidermal growth factor receptor on the surface of cancer cells and locate it in mitochondria. The released LY345899 can inhibit the activity of methylene tetrahydrofolate dehydrogenase 2 and produce excessive reactive oxygen species, leading to the formation of seleninic acid, further reducing the expression of human leukocyte antigen E , which is responsible for the NKG2A-related NK cell inhibition. As a result, the enhanced NK-cell-mediated immunotherapy in conjunction with the cetuximab-mediated antibody-dependent cell-mediated cytotoxicity effect can not only effectively inhibit the growth of xenograft tumors, but also significantly suppress the growth of untreated distant tumors via the abscopal effect. This work, the combination of seleninic acid, LY345899, and cetuximab, provides a new strategy for reversing NK cell exhaustion and has great potential for use in the treatment of metastatic tumors.
Collapse
Affiliation(s)
- Shuojiong Pan
- Key Lab of Organic Optoelectronics and Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Jun Guan
- Key Lab of Organic Optoelectronics and Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Banruo Xianyu
- Key Lab of Organic Optoelectronics and Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Yizheng Tan
- Key Lab of Organic Optoelectronics and Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Tianyu Li
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Huaping Xu
- Key Lab of Organic Optoelectronics and Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
34
|
Li L, Li J. Dimerization of Transmembrane Proteins in Cancer Immunotherapy. MEMBRANES 2023; 13:393. [PMID: 37103820 PMCID: PMC10143916 DOI: 10.3390/membranes13040393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/24/2023] [Accepted: 03/26/2023] [Indexed: 06/19/2023]
Abstract
Transmembrane proteins (TMEMs) are integrated membrane proteins that span the entire lipid bilayer and are permanently anchored to it. TMEMs participate in various cellular processes. Some TMEMs usually exist and perform their physiological functions as dimers rather than monomers. TMEM dimerization is associated with various physiological functions, such as the regulation of enzyme activity, signal transduction, and cancer immunotherapy. In this review, we focus on the dimerization of transmembrane proteins in cancer immunotherapy. This review is divided into three parts. First, the structures and functions of several TMEMs related to tumor immunity are introduced. Second, the characteristics and functions of several typical TMEM dimerization processes are analyzed. Finally, the application of the regulation of TMEM dimerization in cancer immunotherapy is introduced.
Collapse
Affiliation(s)
- Lei Li
- College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Jingying Li
- College of Chemistry, Fuzhou University, Fuzhou 350108, China
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| |
Collapse
|
35
|
Yang K, Zhao Y, Sun G, Zhang X, Cao J, Shao M, Liang X, Wang L. Clinical application and prospect of immune checkpoint inhibitors for CAR-NK cell in tumor immunotherapy. Front Immunol 2023; 13:1081546. [PMID: 36741400 PMCID: PMC9892943 DOI: 10.3389/fimmu.2022.1081546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/20/2022] [Indexed: 01/20/2023] Open
Abstract
Chimeric antigen receptor (CAR) engineering of natural killer (NK) cells is an attractive research field in tumor immunotherapy. While CAR is genetically engineered to express certain molecules, it retains the intrinsic ability to recognize tumor cells through its own receptors. Additionally, NK cells do not depend on T cell receptors for cytotoxic killing. CAR-NK cells exhibit some differences to CAR-T cells in terms of more precise killing, numerous cell sources, and increased effectiveness in solid tumors. However, some problems still exist with CAR-NK cell therapy, such as cytotoxicity, low transfection efficiency, and storage issues. Immune checkpoints inhibit immune cells from performing their normal killing function, and the clinical application of immune checkpoint inhibitors for cancer treatment has become a key therapeutic strategy. The application of CAR-T cells and immune checkpoint inhibitors is being evaluated in numerous ongoing basic research and clinical studies. Immune checkpoints may affect the function of CAR-NK cell therapy. In this review, we describe the combination of existing CAR-NK cell technology with immune checkpoint therapy and discuss the research of CAR-NK cell technology and future clinical treatments. We also summarize the progress of clinical trials of CAR-NK cells and immune checkpoint therapy.
Collapse
Affiliation(s)
- Kangdi Yang
- Department of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yuze Zhao
- School of Basic Medicine, Naval Medical University, Shanghai, China
| | - Guanqun Sun
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China
| | - Xu Zhang
- Department of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jinjin Cao
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China
| | - Mingcong Shao
- Department of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xijun Liang
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China,*Correspondence: Xijun Liang, ; Lina Wang,
| | - Lina Wang
- Department of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China,*Correspondence: Xijun Liang, ; Lina Wang,
| |
Collapse
|
36
|
Cianga VA, Rusu C, Pavel-Tanasa M, Dascalescu A, Danaila C, Harnau S, Aanei CM, Cianga P. Combined flow cytometry natural killer immunophenotyping and KIR/HLA-C genotyping reveal remarkable differences in acute myeloid leukemia patients, but suggest an overall impairment of the natural killer response. Front Med (Lausanne) 2023; 10:1148748. [PMID: 36960339 PMCID: PMC10028202 DOI: 10.3389/fmed.2023.1148748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 02/13/2023] [Indexed: 03/09/2023] Open
Abstract
Introduction Natural killer (NK) cells are key anti-tumor effectors of the innate immunity. Phenotypic differences allow us to discriminate in between three functional stages of maturation, named immature, mature and hypermature that are distinctive in terms of receptor expression, cytokine secretion, cytotoxic properties and organ trafficking. NKs display an impressive repertoire of highly polymorphic germline encoded receptors that can be either activating, triggering the effector's function, or inhibitory, limiting the immune response. In our study, we have investigated peripheral blood NK cells of acute myeloid leukemia (AML) patients. Methods The Killer Immunoglobulin-like receptors (KIRs) and the HLA-C genotypes were assessed, as HLA-C molecules are cognate antigens for inhibitory KIRs. Results The AA mainly inhibitory KIR haplotype was found in a higher proportion in AML, while a striking low frequency of the 2DS3 characterized the mainly activating Bx haplotype. Flow cytometry immunophenotyping evidenced a lower overall count of NK cells in AML versus healthy controls, with lower percentages of the immature and mature subpopulations, but with a markedly increase of the hypermature NKs. The analysis of the KIR2DL1, KIR2DL2, KIR2DL3, KIR3DL1, and NKG2A inhibitory receptors surface expression revealed a remarkable heterogeneity. However, an overall trend for a higher expression in AML patients could be noticed in all maturation subpopulations. Some of the AML patients with complex karyotypes or displaying a FLT3 gene mutation proved to be extreme outliers in terms of NK cells percentages or inhibitory receptors expression. Discussion We conclude that while the genetic background investigation in AML offers important pieces of information regarding susceptibility to disease or prognosis, it is flow cytometry that is able to offer details of finesse in terms of NK numbers and phenotypes, necessary for an adequate individual evaluation of these patients.
Collapse
Affiliation(s)
- Vlad Andrei Cianga
- Department of Hematology, University of Medicine and Pharmacy “Grigore T. Popa”, Iasi, Romania
- Department of Clinical Hematology, Regional Institute of Oncology, Iasi, Romania
| | - Cristina Rusu
- Department of Genetics, University of Medicine and Pharmacy “Grigore T. Popa”, Iasi, Romania
- *Correspondence: Cristina Rusu,
| | - Mariana Pavel-Tanasa
- Department of Immunology, University of Medicine and Pharmacy “Grigore T. Popa”, Iasi, Romania
| | - Angela Dascalescu
- Department of Hematology, University of Medicine and Pharmacy “Grigore T. Popa”, Iasi, Romania
- Department of Clinical Hematology, Regional Institute of Oncology, Iasi, Romania
| | - Catalin Danaila
- Department of Hematology, University of Medicine and Pharmacy “Grigore T. Popa”, Iasi, Romania
- Department of Clinical Hematology, Regional Institute of Oncology, Iasi, Romania
| | - Sebastian Harnau
- Department of Immunology, University of Medicine and Pharmacy “Grigore T. Popa”, Iasi, Romania
| | - Carmen-Mariana Aanei
- Laboratory of Hematology, Nord Hospital, CHU Saint Etienne, Cedex2, Saint-Étienne, France
- INSERM U1059-SAINBIOSE, Université de Lyon, Saint-Étienne, France
| | - Petru Cianga
- Department of Immunology, University of Medicine and Pharmacy “Grigore T. Popa”, Iasi, Romania
- Petru Cianga,
| |
Collapse
|